09.12.2012 Views

Helicobacter pylori - Portal Neonatal

Helicobacter pylori - Portal Neonatal

Helicobacter pylori - Portal Neonatal

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

Textbook of<br />

Pediatric Gastroenterology<br />

and Nutrition


Edited by<br />

Textbook of<br />

Pediatric Gastroenterology<br />

and Nutrition<br />

Stefano Guandalini MD<br />

Professor of Pediatrics<br />

Chief, Section of Gastroenterology<br />

University of Chicago Children’s Hospital<br />

Director, University of Chicago Celiac Disease Program<br />

Chicago, IL<br />

USA


© 2004 Taylor & Francis, an imprint of the Taylor & Francis Group<br />

First published in the United Kingdom in 2004<br />

by Taylor & Francis,<br />

an imprint of the Taylor & Francis Group,<br />

11 New Fetter Lane,<br />

London EC4P 4EE<br />

This edition published in the Taylor & Francis e-Library, 2005.<br />

“To purchase your own copy of this or any of Taylor & Francis or Routledge’s<br />

collection of thousands of eBooks please go to www.eBookstore.tandf.co.uk.”<br />

Tel.: +44 (0) 20 7583 9855<br />

Fax.: +44 (0) 20 7842 2298<br />

Website: www.tandf.co.uk<br />

All rights reserved. No part of this publication may be reproduced, stored in a<br />

retrieval system, or transmitted, in any form or by any means, electronic, mechanical,<br />

photocopying, recording, or otherwise, without the prior permission of the<br />

publisher or in accordance with the provisions of the Copyright, Designs and<br />

Patents Act 1988 or under the terms of any licence permitting limited copying<br />

issued by the Copyright Licensing Agency, 90 Tottenham Court Road, London W1P<br />

0LP.<br />

Although every effort has been made to ensure that all owners of copyright material<br />

have been acknowledged in this publication, we would be glad to acknowledge<br />

in subsequent reprints or editions any omissions brought to our attention.<br />

British Library Cataloguing in Publication Data<br />

Data available on application<br />

Library of Congress Cataloging-in-Publication Data<br />

Data available on application<br />

ISBN 0-203-64045-4 Master e-book ISBN<br />

ISBN 0-203-67686-6 (Adobe eReader Format)<br />

ISBN 1-84184-315-6 (Print Edition)<br />

Distributed in North and South America by<br />

Taylor & Francis<br />

2000 NW Corporate Blvd<br />

Boca Raton, FL 33431, USA<br />

Within Continental USA<br />

Tel.: 800 272 7737; Fax.: 800 374 3401<br />

Outside Continental USA<br />

Tel.: 561 994 0555; Fax.: 561 361 6018<br />

E-mail: orders@crcpress.com<br />

Distributed in the rest of the world by<br />

Thomson Publishing Services<br />

Cheriton House<br />

North Way<br />

Andover, Hampshire SP10 5BE, UK<br />

Tel.: +44 (0) 1264 332424<br />

E-mail: salesorder.tandf@thomsonpublishingservices.co.uk


Contents<br />

Contributors vii<br />

Preface xi<br />

I Congenital Disorders<br />

1 Microvillus inclusion disease and 1<br />

epithelial dysplasia<br />

Olivier Goulet, Nelly Youssef and<br />

Frank M Ruemmele<br />

2 Congenital problems of the 13<br />

gastrointestinal tract<br />

Nigel Hall and Agostino Pierro<br />

II Diseases of the Esophagus and Stomach<br />

3 Infectious esophagitis 29<br />

Salvatore Cucchiara and<br />

Osvaldo Borrelli<br />

4 Gastroesophageal reflux disease 39<br />

Yvan Vandenplas, Silvia Salvatore<br />

and Bruno Hauser<br />

5 Achalasia 61<br />

Carl-Christian A Jackson and<br />

Donald C Liu<br />

6 <strong>Helicobacter</strong> <strong>pylori</strong> gastritis and 73<br />

peptic ulcer disease<br />

Costantino De Giacomo<br />

7 Other gastritides 95<br />

Salvatore Cucchiara and<br />

Osvaldo Borrelli<br />

III Infectious Diseases of the Gastrointestinal<br />

Tract<br />

8 HIV and the intestine 113<br />

Nigel C Rollins<br />

9 Viral diarrhea 127<br />

Alfredo Guarino and Fabio Albano<br />

10 Bacterial infections 145<br />

Alessio Fasano<br />

11 Intestinal parasites 161<br />

David Brewster<br />

12 Post-infectious persistent 193<br />

diarrhea in developing countries<br />

Zulfiqar A Bhutta<br />

13 Small-bowel bacterial overgrowth 201<br />

Mauro Batista de Morais and<br />

Ulysses Fagundes-Neto<br />

IV Functional Disorders and<br />

Neurogastroenterology<br />

14 Functional abdominal pain and 213<br />

other functional bowel disorders<br />

Miguel Saps and Carlo Di Lorenzo<br />

15 Disorders of sucking and swallowing 233<br />

Erasmo Miele and Annamaria Staiano<br />

16 Constipation and encopresis in 247<br />

childhood<br />

Jan Taminiau and Marc Benninga<br />

17 Hirschsprung’s disease and 259<br />

intestinal neuronal dysplasias<br />

Annamaria Staiano, Lucia Quaglietta<br />

and Renata Auricchio<br />

18 Chronic intestinal pseudo- 269<br />

obstruction in childhood<br />

Peter J Milla<br />

19 Gastrointestinal and nutritional 283<br />

problems in the neurologically<br />

handicapped child<br />

Jan Taminiau and Marc Benninga<br />

20 Cyclic vomiting syndrome 289<br />

Bhanu Sunku and B UK Li<br />

V Inflammatory and Immune-mediated<br />

Disorders of the Gastrointestinal Tract<br />

21 Acute and chronic pancreatitis 303<br />

Michelle M Pietzak<br />

22 Food allergies 319<br />

Simon Murch<br />

23 Crohn’s disease 347<br />

Qian Yuan and Harland S Winter


vi<br />

Contents<br />

24 Indeterminate colitis 379<br />

Barbara S Kirschner<br />

25 Ulcerative colitis 385<br />

Leslie M Higuchi and Athos Bousvaros<br />

26 Vasculitides 419<br />

Salvatore Cucchiara and<br />

Osvaldo Borrelli<br />

27 Celiac disease 435<br />

Stefano Guandalini<br />

28 Protein-losing enteropathy 451<br />

Jorge Amil Dias and Eunice Trindade<br />

29 Short-bowel syndrome 461<br />

Olivier Goulet<br />

30 Lymphonodular hyperplasia 479<br />

Jorma Kokkonen<br />

VI Nutritional Problems<br />

31 Malnutrition 489<br />

Michael H N Golden<br />

32 Biotherapeutic and 525<br />

nutraceutical agents<br />

Kirsi Laiho and Erika Isolauri<br />

33 Enteral nutrition 539<br />

Olivier Goulet and Virginie Colomb<br />

34 Parenteral nutrition in infants 555<br />

and children<br />

Olivier Goulet and Virginie Colomb<br />

VII Gastrointestinal Disorders and Nutrition<br />

in the <strong>Neonatal</strong> Age<br />

35 Gastrointestinal problems of 579<br />

the newborn<br />

Moti M Chowdhury and<br />

Agostino Pierro<br />

36 Enteral nutrition in 599<br />

preterm infants<br />

Mario De Curtis and Jacques Rigo<br />

37 Parenteral nutrition in 619<br />

premature infants<br />

Jacques Rigo and Mario De Curtis<br />

VIII Approach to Selected Gastrointestinal<br />

Disorders<br />

38 Approach to gastrointestinal 639<br />

bleeding<br />

Samy Cadranel and<br />

Michèle Scaillon<br />

39 Approach to the child with 655<br />

acute diarrhea<br />

Hania Szajewska and<br />

Jacek Z Mrukowicz<br />

40 Approach to the child with 677<br />

acute abdomen<br />

Luigi Dall’Oglio, Paola De Angelis and<br />

Giovanni Federici di Abriola<br />

41 Management of ingested 691<br />

foreign bodies<br />

Yvan Vandenplas, Said Hachimi-Idrissi<br />

and Bruno Hauser<br />

42 Medical aspects of intestinal 701<br />

transplantation<br />

Olivier Goulet<br />

IX ‘Surgical’ and Neoplastic Disorders of the<br />

Gastrointestinal Tract<br />

43 Intussusception 719<br />

Adolfo Bautista Casasnovas<br />

44 Meckel’s diverticulum 729<br />

Richard G Azizkhan<br />

45 Acute appendicitis 739<br />

Adolfo Bautista Casasnovas<br />

46 Vascular lesions of the 751<br />

gastrointestinal tract in childhood<br />

Steven R Allen and Richard G Azizkhan<br />

47 The role of minimally invasive 761<br />

surgery in pediatric<br />

gastrointestinal disease<br />

Stig Somme and Donald C Liu<br />

48 Polyps and other tumors of the 771<br />

gastrointestinal tract<br />

Warren Hyer and John Fell<br />

Index 787


Contributors<br />

Fabio Albano<br />

Department of Pediatrics<br />

University of Naples ‘Federico II’<br />

Via S. Pansini 5<br />

80131 Naples, Italy<br />

Steven R Allen<br />

University of Cincinnati College of Medicine<br />

Cincinnati, OH 45229, USA<br />

Jorge Amil Dias<br />

Pediatric Gastroenterology Unit<br />

Hospital S. João<br />

4200 Porto, Portugal<br />

Renata Auricchio<br />

Department of Pediatrics<br />

University of Naples ‘Federico II’<br />

Via S. Pansini 5<br />

80131 Naples, Italy<br />

Richard G Azizkhan<br />

Cincinnati Children’s Hospital Medical Center<br />

Division of Pediatric Surgery<br />

3333 Burnet Avenue<br />

Cincinnati, OH 45229, USA<br />

Mauro Batista de Morais<br />

Division of Pediatric Gastroenterology<br />

Escola Paulista de Medicina<br />

Universidade Federal de São Paulo<br />

São Paulo, Brazil<br />

Adolfo Bautista Casasnovas<br />

Section of Pediatric Surgery<br />

Clinical University Hospital of Santiago de<br />

Compostela<br />

Choupana s/n<br />

15706 Santiago, Spain<br />

Marc Benninga<br />

Emma Kinderziekenhuis AMC<br />

Department of Pediatrics<br />

Postbus 22660<br />

NL-1100 DD<br />

Amsterdam, The Netherlands<br />

Zulfiqar A Bhutta<br />

Department of Paediatrics<br />

The Aga Khan University<br />

Stadium Road<br />

Karachi 74800, Pakistan<br />

Osvaldo Borrelli<br />

Department of Pediatrics<br />

Gastroenterology Service<br />

University of Rome ‘La Sapienza’<br />

Viale Regina Elena 324<br />

00161 Rome, Italy<br />

Athos Bousvaros<br />

Division of Gastroenterology and Nutrition<br />

Children’s Hospital<br />

Boston, MA 02115, USA<br />

David Brewster<br />

Northern Territory Clinical School<br />

Flinders University<br />

Casuarina NT0811, Australia<br />

Samy Cadranel<br />

Queen Fabiola Children’s Hospital<br />

Department of Gastroenterology and Nutrition<br />

Avenue J J Crocq 15<br />

B-1020 Brussels, Belgium


viii<br />

Contributors<br />

Moti M Chowdhury<br />

Department of Paediatric Surgery<br />

Surgery Unit<br />

Institute of Child Health<br />

30 Guilford Street<br />

London WC1N 1EH, UK<br />

Virginie Colomb<br />

Service de Gastroentérologie et Nutrition<br />

Pédiatriques<br />

Hôpital Necker – Enfants Malades<br />

149 Rue de Sèvres<br />

75743 Paris, France<br />

Salvatore Cucchiara<br />

Department of Pediatrics<br />

Gastroenterology Service<br />

University of Rome ‘La Sapienza’<br />

Viale Regina Elena 324<br />

00160 Rome, Italy<br />

Luigi Dall’Oglio<br />

Digestive Surgical and Endoscopic Unit<br />

Ospedale Pediatrico Bambino Gesù-IRCCS<br />

Viale S. Onofrio<br />

00199 Rome, Italy<br />

Paola De Angelis<br />

Digestive Surgical and Endoscopic Unit<br />

Ospedale Pediatrico Bambino Gesù-IRCCS<br />

Viale S. Onofrio<br />

00199 Rome, Italy<br />

Mario De Curtis<br />

Dipartimento Materno-Infantile<br />

University of Palermo<br />

90142 Palermo, Italy<br />

Costantino De Giacomo<br />

Unità Operativa di Pediatria<br />

Ospedale Niguarda Cà Granda<br />

Piazza Ospedale Maggiore 3<br />

20162 Milano, Italy<br />

Carlo Di Lorenzo<br />

Division of Pediatric Gastroenterology<br />

Children’s Hospital of Pittsburgh<br />

University of Pittsburgh Medical Center<br />

3705 5th Avenue<br />

Pittsburgh, PA 15213, USA<br />

Ulysses Fagundes-Neto<br />

Division of Pediatric Gastroenterology<br />

Escola Paulista de Medicina<br />

Universidade Federal de São Paulo<br />

São Paulo, Brazil<br />

Alessio Fasano<br />

Mucosal Biology Research Center<br />

University of Maryland School of Medicine<br />

22 S. Greene Street<br />

Baltimore, MD 21201, USA<br />

Giovanni Federici di Abriola<br />

Digestive Surgical and Endoscopic Unit<br />

Ospedale Pediatrico Bambino Gesù-IRCCS<br />

Viale S. Onofrio<br />

00199 Rome, Italy<br />

John Fell<br />

Chelsea and Westminster Hospital<br />

London, UK<br />

Michael H N Golden<br />

Department of Medicine and Therapeutics<br />

University of Aberdeen<br />

Scotland, UK<br />

Olivier Goulet<br />

Service de Gastroentérologie et Nutrition<br />

Pédiatriques<br />

Hôpital Necker – Enfants Malades<br />

149 Rue de Sèvres<br />

75743 Paris, France<br />

Stefano Guandalini<br />

Section of Gastroenterology, Hepatology and<br />

Nutrition<br />

University of Chicago<br />

5839 S. Maryland Avenue<br />

Chicago, IL 60637, USA<br />

Alfredo Guarino<br />

Department of Pediatrics<br />

University of Naples ‘Federico II’<br />

Via S. Pansini 5<br />

80131 Naples, Italy<br />

Said Hachimi-Idrissi<br />

Academische Ziekenhuis Kinderen<br />

Vrije Universiteit Brussel<br />

Laarbeeklaan 101<br />

B-1090 Brussels, Belgium<br />

Nigel Hall<br />

Department of Paediatric Surgery<br />

Institute of Child Health<br />

30 Guilford Street<br />

London WC1N 1EH, UK


Bruno Hauser<br />

Academische Ziekenhuis Kinderen<br />

Vrije Universiteit Brussel<br />

Laarbeeklaan 101<br />

B-1090 Brussels, Belgium<br />

Leslie M Higuchi<br />

Division of Gastroenterology and Nutrition<br />

Children’s Hospital<br />

Boston, MA 02115, USA<br />

Warren Hyer<br />

Northwick Park and St Mark’s Hospital<br />

Middlesex, UK<br />

Erika Isolauri<br />

Department of Pediatrics<br />

University of Turku<br />

Kiinamyllynk. 4-8<br />

FIN-20520 Turku, Finland<br />

Carl-Christian A Jackson<br />

University of Chicago Pritzker School of Medicine<br />

5841 S. Maryland Avenue<br />

Chicago, IL 60637, USA<br />

Barbara S Kirschner<br />

University of Chicago Pritzker School of Medicine<br />

5841 S. Maryland Avenue<br />

Chicago, IL 60637, USA<br />

Jorma Kokkonen<br />

Department of Pediatrics<br />

University Hospital of Oulu<br />

Oulu<br />

FIN-90029, Finland<br />

Kirsi Laiho<br />

Department of Pediatrics<br />

University of Turku<br />

Kiinamyllynk. 4-8<br />

FIN-20520 Turku, Finland<br />

B UK Li<br />

Department of Pediatrics<br />

Northwestern University McGraw School of<br />

Medicine<br />

Children’s Memorial Hospital<br />

Chicago, IL 60614, USA<br />

Donald C Liu<br />

Pediatric Surgery<br />

University of Chicago Comer Children’s Hospital<br />

5841 S. Maryland Avenue<br />

Chicago, IL 60637, USA<br />

Erasmo Miele<br />

Department of Pediatrics<br />

University of Naples ‘Federico II’<br />

Via S. Pansini 5<br />

80131 Naples, Italy<br />

Peter J Milla<br />

Gastroenterology Unit<br />

Institute of Child Health<br />

30 Guilford Street<br />

London, WC1N 1EH, UK<br />

Contributors ix<br />

Jacek Z Mrukowicz<br />

Polish Institute for Evidence Based Medicine<br />

Cracow, Poland<br />

Simon Murch<br />

Centre for Paediatric Gastroenterology<br />

Royal Free Hospital and University College<br />

Medical School<br />

Royal Free Campus<br />

Rowland Hill Street<br />

London, NW3 2PF, UK<br />

Agostino Pierro<br />

Department of Paediatric Surgery<br />

Institute of Child Health<br />

30 Guilford Street<br />

London WC1N 1EH, UK<br />

Michelle M Pietzak<br />

Division of Gastroenterology and Nutrition<br />

Children’s Hospital of Los Angeles<br />

4650 Sunset Boulevard<br />

Los Angeles, CA 90027, USA<br />

Lucia Quaglietta<br />

Department of Pediatrics<br />

University of Naples ‘Federico II’<br />

Via S. Pansini 5<br />

80131 Naples, Italy<br />

Jacques Rigo<br />

Department of Pediatrics<br />

Division of Neonatology<br />

University of Liège<br />

CHR Citadelle<br />

4000 Liège, Belgium<br />

Nigel C Rollins<br />

Department of Paediatrics and Child Health<br />

Nelson R Mandela School of Medicine<br />

719 Umbilo Road<br />

Congella 4013<br />

Durban, South Africa


x<br />

Contributors<br />

Frank M Ruemmele<br />

Service de Gastroentérologie et Nutrition<br />

Pédiatriques<br />

Hôpital Necker – Enfants Malades<br />

149 Rue de Sèvres<br />

75743 Paris, France<br />

Silvia Salvatore<br />

Academische Ziekenhuis Kinderen<br />

Vrije Universiteit Brussel<br />

Laarbeeklaan 101<br />

B-1090 Brussels, Belgium<br />

Miguel Saps<br />

Division of Pediatric Gastroenterology<br />

Children’s Hospital of Pittsburgh<br />

University of Pittsburgh Medical Center<br />

3705 5th Avenue<br />

Pittsburgh, PA 15213, USA<br />

Michèle Scaillon<br />

Queen Fabiola Children’s Hospital<br />

Department of Gastroenterology and Nutrition<br />

Avenue J J Crocq 15<br />

B-1020 Brussels, Belgium<br />

Stig Somme<br />

Louisiana State University School of Medicine<br />

1542 Tulane Avenue<br />

New Orleans, LA 70112, USA<br />

Annamaria Staiano<br />

Department of Pediatrics<br />

University of Naples ‘Federico II’<br />

Via S. Pansini 5<br />

80131 Naples, Italy<br />

Bhanu Sunku<br />

Department of Pediatrics<br />

Northwestern University McGraw School of<br />

Medicine<br />

Children’s Memorial Hospital<br />

Chicago, IL 60614, USA<br />

Hania Szajewska<br />

Department of Pediatric Gastroenterology and<br />

Nutrition<br />

The Medical University of Warsaw<br />

Dzialdowska 1<br />

01-184 Warsaw, Poland<br />

Jan Taminiau<br />

Emma Kinderziekenhuis AMC<br />

Department of Pediatrics<br />

Postbus 22660<br />

NL-1100 DD<br />

Amsterdam, The Netherlands<br />

Eunice Trindade<br />

Pediatric Gastroenterology Unit<br />

Hospital S. João<br />

4200 Porto, Portugal<br />

Yvan Vandenplas<br />

Academische Ziekenhuis Kinderen<br />

Vrije Universiteit Brussel<br />

Laarbeeklaan 101<br />

B-1090 Brussels, Belgium<br />

Harland S Winter<br />

Pediatric IBD Center<br />

Department of Pediatrics<br />

Massachusetts General Hospital for Children<br />

55 Fruit Street<br />

Boston, MA 02114, USA<br />

Nelly Youssef<br />

Service de Gastroentérologie et Nutrition<br />

Pédiatriques<br />

Hôpital Necker – Enfants Malades<br />

149 Rue de Sèvres Paris<br />

75743 Paris, France<br />

Qian Yuan<br />

Pediatric IBD Center<br />

Department of Pediatrics<br />

Massachusetts General Hospital for Children<br />

55 Fruit Street<br />

Boston, MA 02114, USA


Preface<br />

Our discipline is young. When I began focusing on<br />

pediatric gastroenterology, in the early 1970s,<br />

there were no textbooks available. My training was<br />

based on the teaching of outstanding mentors<br />

(such as Salvatore Auricchio and Armido Rubino,<br />

to whom I will always be indebted), on eager clinical<br />

work and on tireless research; and reading of<br />

adult gastrointestinal textbooks that were regarded<br />

as the necessary theoretical fundaments of knowledge.<br />

Then the Silvermann, Roy and Cozzetto<br />

book (Pediatric Clinical Gastroenterology. St Louis,<br />

MO: Mosby, 1971) came along, somewhat officially<br />

identifying our field as a free-standing subspecialty<br />

and heralding a new exciting era of<br />

educational tools directed at our discipline.<br />

The knowledge then seems to have almost<br />

exploded, with technology rapidly introducing<br />

new investigative, diagnostic and therapeutic<br />

modalities; new acquisitions making old approaches<br />

quickly obsolete; and the sheer body of<br />

new information forcing us to become sub-subspecialists.<br />

Hepatologists (now rightly even transplant<br />

hepatologists), endoscopists, neurogastroenterologists<br />

and more ‘ologists’ have emerged and<br />

have gained or are gaining their cultural and<br />

operative independence.<br />

In this scenario of ever-changing evolution, and in<br />

the Internet era, why another pediatric gastroenterology<br />

and nutrition book? I believe the<br />

answer lies in the progressive globalization that<br />

the Internet has certainly catalyzed and that we<br />

see occurring before our eyes. We are living more<br />

and more in a true global village, where educational<br />

resources need not only to be scientifically<br />

correct and updated, but also able to offer, in<br />

a comprehensive, cohesive package, information<br />

that is also reflective of the different realities that<br />

gastroenterological and nutritional problems take<br />

on in different parts of the world. Intentionally,<br />

this book does not cover any liver-related disorder.<br />

Hepatology has become a discipline in its own<br />

right, and authoritative, specific books addressing<br />

only pediatric hepatology have already appeared<br />

and are available. We felt that focusing on the two<br />

intertwined areas of gastroenterology and nutrition<br />

would serve better the needs of the pediatric<br />

gastroenterologist. However, the book has a rather<br />

ambitious goal: that of having a global ‘flavor’. It is<br />

in fact born out of a new vision of worldwide cooperation<br />

among pediatric gastroenterologists,<br />

which is embodied by the recently created<br />

Federation of the International Societies of Pediatric<br />

Gastroenterology, Hepatology and Nutrition<br />

(FISPGHAN). I can only hope that its goal will be<br />

reached; certainly all the authors and I tried very<br />

hard. Happy Reading!<br />

Stefano Guandalini, MD<br />

Professor of Pediatrics<br />

President of FISPGHAN


1<br />

Introduction<br />

Microvillus inclusion disease<br />

and epithelial dysplasia<br />

Olivier Goulet, Nelly Youssef and Frank M Ruemmele<br />

The definition, presentation and outcome of<br />

intractable diarrhea of infancy (IDI) have changed<br />

considerably over the past three decades, owing to<br />

major improvements in nutritional management,<br />

and a better understanding of the pathology of the<br />

small bowel mucosa.<br />

Definition of protracted and<br />

intractable diarrhea of infancy<br />

Originally, the syndrome of IDI was described by<br />

Avery et al in 1968, based on the following<br />

features: diarrhea occurring in a newborn younger<br />

than 3 months of age, lasting more than 2 weeks,<br />

with three or more negative stool cultures for<br />

bacterial pathogens. 1 Most cases were managed in<br />

hospital, using intravenous fluids while the diarrhea<br />

was persistent and intractable, with a high<br />

mortality rate from infection or malnutrition. 2<br />

Recently, the term ‘severe diarrhea requiring<br />

parenteral nutrition’ was proposed. 3,4 Within this<br />

group of pathologies, two major subtypes can be<br />

differentiated. The first group is made up of<br />

patients with ‘protracted diarrhea of infancy’ (PDI),<br />

which subsides despite its initial severity. PDI can<br />

result from a specific immune deficiency, a sensitization<br />

to a common food protein (e.g. cow’s milk<br />

or gluten), or it can be secondary to a severe<br />

infection of the digestive tract (post-enteritis<br />

syndrome). The second group is characterized by<br />

an ‘intractable diarrhea of infancy’, with onset<br />

within the first 2 years of life. In this second group,<br />

diarrhea persists sometimes for years, despite<br />

prolonged bowel resting and various therapeutic<br />

trials. In most cases, such as constitutive enterocyte<br />

disorders 5 or autoimmune enteropathy, 6 the<br />

situation becomes rapidly life threatening, and<br />

these patients depend on long-term parenteral<br />

nutrition (PN). Some of them are candidates for<br />

intestinal transplantation. Table 1.1 shows the<br />

diagnostic heterogeneity of 65 cases with severe<br />

diarrhea requiring PN for more than 1 month, as<br />

recently analyzed by a French multicenter study. 7<br />

The so-called ‘intractable ulcerating enterocolitis’<br />

of early onset is difficult to classify. 8 It is very<br />

important to distinguish between IDI and PDI,<br />

since children with PDI always recover, sometimes<br />

after only several weeks or months of parenteral<br />

and/or enteral nutrition. In contrast, patients with<br />

IDI never recover, and are dependent on life-long<br />

parenteral nutrition or – in the case of autoimmune<br />

enteropathy – life-long massive immunosuppressive<br />

medication.<br />

Current classification<br />

An attempt to classify intractable diarrhea according<br />

to villus atrophy was proposed, on the basis of<br />

immunohistological criteria emphasizing the role<br />

of activated T cells in the intestinal mucosa. 9 A<br />

multicenter survey from the European Society for<br />

Paediatric Gastroenterology, Hepatology and<br />

Nutrition (ESPGHAN) collected different cases of<br />

IDI and villus atrophy with precisely defined light<br />

microscopic characteristics, categorizing several<br />

types of IDI. 10 On histological analysis, two clearly<br />

different groups of IDI can be separated. The first<br />

one is characterized by a mononuclear cell infiltration<br />

of the lamina propria, and is considered to<br />

be associated with activated T cells. Within this<br />

group, two different clinical presentations can be<br />

distinguished. Patients presenting with additional<br />

extradigestive autoimmune symptoms (such as<br />

diabetes, arthritis, thyroiditis, dermatitis and<br />

nephrotic syndrome) tended to have a later onset<br />

1


2<br />

Microvillus inclusion disease and epithelial dysplasia<br />

Table 1.1 Causes and outcomes of protracted and intractable<br />

diarrhea of infancy<br />

Cause Patients (n) Deceased (n)<br />

Protracted diarrhea 39 1<br />

Multiple food intolerance 15 0<br />

Infectious enteritis 14 0<br />

Colitis (including two with CMV) 6 1<br />

CDG syndrome 1 0<br />

Ganglioneuroblastoma 1 0<br />

Unknown 2 0<br />

Intractable diarrhea<br />

Abnormalities of the enterocyte<br />

21 4<br />

intestinal epithelial dysplasia 6 1<br />

microvillus atrophy 3 0<br />

Autoimmune enteropathy 5 3<br />

Phenotypic diarrhea 3 0<br />

Undefined 4 0<br />

CMV, cytomegalovirus; CDG, congenital disorders of glycosylation<br />

of diarrhea, which was more abundant and more<br />

severe than in patients with isolated gastrointestinal<br />

symptoms and gut autoantibodies. The second<br />

histological pattern includes the early onset of<br />

severe intractable diarrhea with villus atrophy,<br />

without mononuclear cell infiltration of the<br />

lamina propria but specific histological abnormalities<br />

involving the epithelium. To date, several<br />

types of primary epithelial abnormality inducing<br />

IDI have been identified. The first described was<br />

microvillus atrophy or microvillus inclusion<br />

disease (MVID) and, more recently, tufting<br />

enteropathy or epithelial dysplasia. 11 Some<br />

patients are small for gestational age and present<br />

with phenotypic abnormalities corresponding to<br />

the previously described syndromatic diarrhea. 12<br />

Microvillus inclusion disease<br />

In 1978, Davidson et al reported five infants with<br />

severe, persistent diarrhea beginning in the<br />

newborn period, in whom light microscopy<br />

revealed crypt hypoplastic villus atrophy. 13<br />

Electron microscopic examination of small intestinal<br />

biopsies from three of the patients showed<br />

severe brush-border abnormalities and increased<br />

liposome-like bodies and, in one, intracytoplasmic<br />

cysts made up of brush border. Further children<br />

were reported with these characteristic cytoplasmic<br />

inclusions of the brush-border membrane. 5,14<br />

From these first clinical and histological descriptions,<br />

MVID has been established as a distinct<br />

disease within the syndrome of IDI, based on the<br />

characteristic morphological features. MVID in its<br />

typical form is a congenital disorder of intestinal<br />

epithelial cells, presenting as intractable secretory<br />

diarrhea within the first days of life. 15<br />

Clinical expression<br />

In general, infants develop severe secretory diarrhea<br />

within the first days after birth. Stool volumes<br />

reach 250–300ml/kg body weight per day, with


electrolyte concentrations similar to those seen in<br />

small-intestinal fluid. This disorder is life threatening,<br />

since massive diarrhea leads to rapid dehydration<br />

and electrolyte imbalance, with subsequent<br />

metabolic decompensation within a few<br />

hours. 13–15 Severe watery diarrhea persists despite<br />

bowel rest. The differential diagnosis may include<br />

congenital chloride diarrhea or sodium malabsorption<br />

diarrhea, which can be easily distinguished<br />

from MVID by blood and stool electrolyte assessment.<br />

16,17 In contrast, clinical presentation of<br />

MVID may sometimes be atypical, in the form of a<br />

predominantly pseudo-occlusive syndrome, with<br />

full and distended small bowel and colon. Some<br />

newborns have been thought to present intestinal<br />

pseudo-obstruction syndrome, and unfortunately<br />

in some of them an ileostomy was created. The<br />

most common time of onset of diarrhea in MVID is<br />

within the first few days (early-onset or congenital<br />

form). However, in a few patients the onset of diarrhea<br />

is delayed (first month) and less severe (lateonset<br />

form).<br />

Histological examination<br />

Histological analysis of small bowel biopsies<br />

shows a variable degree of villus atrophy without<br />

any inflammatory infiltrate. Highly characteristic<br />

for this disorder is the accumulation of periodic<br />

acid Schiff (PAS)-positive secretory granules<br />

within the apical cytoplasm of enterocytes. 18–20 On<br />

(a) (b)<br />

Microvillus inclusion disease 3<br />

the ultrastructural level, rare or absent microvilli<br />

on intestinal epithelial cells along with inclusions<br />

of microvilli in the cytoplasm of enterocytes are<br />

seen, which define this entity. 21<br />

Diagnosis may be easily performed from light<br />

microscopic examination of a duodenal or jejunal<br />

biopsy specimen. On hematoxylin–eosin staining,<br />

the mucosa appears flattened with hypoplastic<br />

villus atrophy. PAS staining reveals an abnormal<br />

brush-border pattern with positive-staining material<br />

within the apical cytoplasm of enterocytes<br />

(Figure 1.1). A valuable new tool for the light<br />

microscopic diagnosis of MVID was recently<br />

proposed. 22,23 CD-10 is a membrane-associated<br />

neutral peptidase, shown to have a linear brushborder<br />

staining pattern in normal small intestine.<br />

In contrast to this surface staining in different<br />

controls (normal intestine, celiac disease, autoimmune<br />

enteropathy, allergy), all MVID cases<br />

revealed prominent intracytoplasmic CD-10<br />

immunoreactivity in surface enterocytes 22,23<br />

(Figure 1.2). Similar results were obtained with<br />

PAS, polyclonal carcinoembryonic antigen and<br />

alkaline phosphatase, three stains known to show<br />

cytoplasmic staining of surface enterocytes in<br />

MVID. 23 On electron microscopy, surface epithelial<br />

cells show absent or grossly abnormal<br />

microvilli, as well as numerous vesicular bodies of<br />

various sizes, and the characteristic microvillus<br />

inclusions (Figure 1.3). Crypt cells are morphologically<br />

almost normal, but do not contain increased<br />

Figure 1.1 Microvillus inclusion disease: periodic acid-Schiff (PAS) staining. (a) Normal mucosa, normal PAS, brushborder<br />

staining; (b) abnormal accumulation of PAS, positive material in the apical cytoplasm of epithelial cells in microvillus<br />

inclusion disease.


4<br />

Microvillus inclusion disease and epithelial dysplasia<br />

(a) (b)<br />

Figure 1.2 Microvillus inclusion disease: CD-10 immunostaining. (a) Normal mucosa, normal CD-10, brush-border<br />

immunostaining; (b) abnormal accumulation of positive material in the apical cytoplasm of epithelial cells in microvillus<br />

inclusion disease.<br />

Figure 1.3 Microvillus inclusion disease. Electron<br />

microscopy of jejunal biopsy specimen. The brush border<br />

is almost absent. The cytoplasm contains a microvillus<br />

inclusion.<br />

Figure 1.4 Microvillus inclusion disease. Electron<br />

microscopy of colonic biopsy specimen.<br />

numbers of apical vesicles and vesicular bodies.<br />

Microvillus inclusions, as well as increased secretory<br />

granules, are also present in the large bowel,<br />

more easily accessible for biopsy, especially in<br />

early infancy (Figure 1.4).<br />

Pathophysiology and mode of transmission<br />

A defect in the membrane trafficking of immature<br />

and/or differentiating enterocytes has been<br />

discussed as an etiopathogenic mechanism in<br />

MVID. 20,21 This membrane defect results, as a


direct functional consequence, in complete intestinal<br />

failure. It has been speculated that the disease<br />

is associated with a disorder of the enterocyte<br />

cytoskeleton, which produces an abnormal assembly<br />

of microvilli. Intestinal microvillus dystrophy<br />

was reported as being a hypothetic variant of<br />

MVID. 24 The underlying pathogenesis of MVID is<br />

still unclear, although a cytoskeletal myosin deficiency<br />

has been found. 25 When analyzing the<br />

turnover of sucrase-isomaltase, as a representative<br />

brush-border protein, there is clear evidence that<br />

the direct and indirect constitutive pathways are<br />

intact in MVID. 21 Therefore, a defect in endocytosis<br />

is rather unlikely. More recently, by investigating<br />

the glycobiological nature of the epithelial<br />

accumulation of PAS, Phillips et al 26 suggested<br />

that MVID involves a defect in exocytosis of the<br />

glycocalyx. 26 The absence of glycocalyx might<br />

impair normal cell functions.<br />

Considering the number of cases with affected<br />

siblings, and the frequency of consanguinity<br />

among patients in families of affected infants, this<br />

disease appears to be transmitted as an autosomal<br />

recessive trait. 13,15,27 No candidate gene has been<br />

identified to date. MVID has been reported in a girl<br />

with autosomal dominant hypochondroplasia. 28<br />

The gene defect of this disease was recently localized<br />

on the chromosome region 4p16.3, which<br />

might help in elucidating the genetic basis of MVID.<br />

Long-term outcome<br />

MVID is a congenital constitutive intestinal epithelial<br />

cell disorder leading, in its typical early-onset<br />

form, to permanent intestinal failure. The largest<br />

multicenter survey, of 23 MVID patients, 15<br />

revealed an extremely reduced life expectancy<br />

with a 1-year survival rate of less than 25%. Most<br />

children died of septic complications, liver failure,<br />

or metabolic decompensation. Few cases of MVID,<br />

especially with the late-onset form, may survive<br />

with limited stool output and may require only<br />

partial PN. 29 Treatment with corticosteroids,<br />

colostrum or epidermal growth factor has not been<br />

successful, but octreotide has been used with<br />

partial success in one patient. 15 In contrast to the<br />

initial outcomes before the 1980s, PN now allows<br />

most infants and children to survive. However,<br />

complications related to inadequate PN do limit<br />

long-term survival. These include recurrent<br />

Microvillus inclusion disease 5<br />

catheter-related sepsis, extensive thrombosis, fat<br />

overload syndrome and cholestasis. In addition,<br />

without evidence of an associated renal disease,<br />

some of these infants and children present chronic<br />

hydro-electrolytic imbalance and acidosis, with<br />

subsequent impaired length growth. Others,<br />

because of repeated dehydration episodes associated<br />

with unadapted phosphocalcic intakes,<br />

present with nephrocalcinosis. Finally, even with<br />

adequate long-term PN and normal growth, most<br />

children remain with high and uncomfortable<br />

stool output. This requires daily fluid replacement<br />

with the high risk of severe dehydration. Intestinal<br />

transplantation therefore became the only definitive<br />

treatment of this rare intestinal disease. 30–33<br />

Definitive treatment<br />

Since the introduction of tacrolimus (originally<br />

FK506) as an immunosuppressive drug after organ<br />

transplantation in the early 1990s, the outcome of<br />

intestinal transplantation markedly improved.<br />

Several cases of successful transplantation for<br />

MVID have been reported. 30–35 Transplantation<br />

involved isolated intestine, 30–33 or intestine<br />

combined with the liver. 31,32 Following the report<br />

of these cases, there has been an ongoing discussion<br />

on whether or not the colon should be transplanted<br />

together with the small bowel.<br />

We recently evaluated the possibility and outcome<br />

of intestinal transplantation in ten consecutive<br />

patients with early-onset congenital MVID at<br />

Necker-Enfants Malades Hospital (Paris) (Ruemmele<br />

et al, submitted for publication). Two<br />

patients died before they could be put on a waiting<br />

list for small-bowel transplantation; one patient is<br />

still waiting. We performed cadaveric intestinal<br />

transplantation in seven patients aged between 3<br />

and 11 years by using tacrolimus, steroids and<br />

interleukin (IL-2) blockers. Three transplantations<br />

were performed with isolated intestine, and four<br />

with intestine associated with the liver. Right<br />

colon transplantation was performed in five cases<br />

(two with isolated intestine). One patient died<br />

during transplantation surgery from acute liver<br />

failure and hemodynamic shock, probably due to<br />

re-perfusion shock. The six others (86%) survived,<br />

with a median follow-up of 3 years (range 1–8<br />

years). Graft rejections occurred in two patients<br />

(one with isolated intestinal transplantation, and


6<br />

Microvillus inclusion disease and epithelial dysplasia<br />

one with intestine and liver), who responded<br />

favorably to methylprednisolone pulses. Complete<br />

weaning from PN was achieved in all patients: the<br />

five patients with an additional colon graft were<br />

off PN after a median of 36 days after surgery, as<br />

opposed to those without colonic transplant who<br />

obtained full intestinal autonomy several months<br />

after transplantation. Thus, intestinal transplantation<br />

alone or in combination with liver, offered<br />

MVID children for the first time a long-term<br />

perspective. In this series at Necker-Enfants<br />

Malades Hospital, additional colon grafting<br />

markedly improved outcome and quality of life<br />

after transplantation in MVID. These preliminary<br />

results of intestinal transplantation in this rare<br />

disease are very encouraging, and demonstrate<br />

that the prognosis of MVID has changed dramatically<br />

during the past decade.<br />

Finally, in a child suspected of having MVID, a<br />

precise classification into one of three different<br />

subtypes (congenital–early-onset, late-onset or<br />

atypical) should be obtained as early as possible<br />

and confirmed by an experienced pathologist,<br />

according to clearly defined morphological criteria.<br />

Recently, Croft et al 34 reported a 5-year-old girl<br />

with a late-onset form of MVID who was weaned<br />

from total PN and is thriving on a normal unrestricted<br />

diet. This rare form of MVID probably has<br />

a better prognosis than the most frequent form of<br />

early-onset MVID, with a high mortality rate. 15<br />

Given the high success rate of intestinal transplantation<br />

in MVID, we recommend transplantation,<br />

once the definitive diagnosis of early-onset MVID<br />

is made. In addition, the potential candidate for<br />

transplantation has to be in a eutrophic phase<br />

under total PN. This policy should allow avoidance<br />

of liver impairment. Conversely, patients<br />

with a late-onset or atypical form of MVID should<br />

not be automatically scheduled for small-bowel<br />

transplantation. The individual course of the<br />

disease will help in deciding whether or not a<br />

child with MVID is a candidate for intestinal transplantation.<br />

Intestinal epithelial dysplasia (or<br />

tufting enteropathy)<br />

In 1994, three cases of neonatal severe diarrhea<br />

with abnormal epithelial pictures were reported by<br />

Reifen et al under the name of ‘tufting enteropa-<br />

thy’. 36 In our own series at Necker-Enfants<br />

Malades, we identified nine cases of severe neonatal<br />

diarrhea that were clearly different from<br />

MVID. 12 Further studies in these patients<br />

confirmed that intestinal epithelial dysplasia (IED)<br />

is a constitutive epithelial disorder involving both<br />

small intestine and colon. 37 In our experience, IED<br />

seems to be frequent in patients of Arabic origin,<br />

from the Middle-East or North Africa. Main characteristics<br />

of this disease are its clinical and histological<br />

heterogeneity and its association with<br />

malformations or other epithelial diseases.<br />

Clinical expression<br />

Typically, the patients present during the first<br />

weeks of life with severe diarrhea. Most have<br />

affected consanguineous parents and/or siblings,<br />

some of whom died during the first months of life<br />

with severe diarrhea of unknown origin. Most of<br />

the time, diarrhea persists despite bowel rest, but<br />

at a lower level when compared to MVID.<br />

Therefore, attempts at continuous enteral feeding<br />

with protein hydrolysates or amino acid-based<br />

formulas were performed in some patients.<br />

Unfortunately, most often the continuous enteral<br />

feeding exacerbated the diarrhea, and particularly<br />

the newborns with IED did not grow adequately.<br />

They rapidly developed failure to thrive, with<br />

severe protein energy malnutrition. Because of the<br />

early onset of diarrhea, MVID is often suspected in<br />

these children. However, morphological analysis<br />

of small- and large-bowel biopsies easily distinguishes<br />

these entities (see above). A major problem<br />

in the diagnosis of IED is its clinical and histological<br />

heterogeneity.<br />

Histological features<br />

Villus atrophy of variable severity is present. In the<br />

typical form, abnormalities are localized mainly in<br />

the epithelium and include a disorganization of<br />

surface enterocytes with focal crowding, resembling<br />

tufts (Figure 1.5). These characteristic ‘tufts’<br />

of extruding epithelium first described by Reifen et<br />

al 36 are seen towards the villus tip, and may affect<br />

up to 70% of villi. The tufting process is not<br />

limited to the small intestine but also involves the<br />

colonic mucosa. 11 This picture can also be<br />

observed in crypt epithelium and, in addition,


Figure 1.5 Epithelial dysplasia. Disorganization of<br />

surface epithelium showing tufts with apical rounding<br />

epithelial cells.<br />

crypts often have an abnormal aspect with dilatations<br />

such as pseudocysts and abnormal regeneration<br />

with branching 11 (Figure 1.6). The study of<br />

basement membrane components demonstrated<br />

an abnormal laminin and heparan sulfate proteoglycan<br />

deposition at that level, compared to biopsy<br />

specimens from patients with celiac disease or<br />

autoimmune enteropathy. 11 Relative to the<br />

controls, there was faint and irregular laminin<br />

deposition at the epithelial–lamina propria interface,<br />

while heparan sulfate proteoglycan depositions<br />

were large and lamellar, suggestive of abnormal<br />

development of basement membrane at the<br />

origin of the epithelial abnormalities. On the other<br />

hand, we observed an increased immunohistochemical<br />

expression of desmoglein in IED, and<br />

ultrastructural changes of desmosomes, which<br />

were increased in length and number (Figure<br />

1.7). 37<br />

Diagnosis<br />

Diagnosis of IED is sometimes difficult for several<br />

reasons. The early onset of severe and permanent<br />

diarrhea and the lack of features diagnostic of<br />

microvillus inclusion disease are important diagnostic<br />

elements. However, the characteristic tufts<br />

of extruding epithelium may not be obvious, especially<br />

early on. At the onset of the clinical course,<br />

IED is most often suspected after elimination of<br />

MVID, and the final diagnosis is made rather late,<br />

by performing repeated intestinal biopsies which<br />

Intestinal epithelial disease 7<br />

Figure 1.6 Epithelial dysplasia. Partial villus atrophy with<br />

crypt hyperplasia and/or pseudocystic crypt appearance,<br />

branching and disorganization of surface epithelium.<br />

Figure 1.7 Desmoglein staining. Increased expression of<br />

desmoglein in the tight junctions.<br />

change from normal in early life (only non-specific<br />

villus atrophy with or without mononuclear cell<br />

infiltration of the lamina propria) to the characteristic<br />

tufts. In addition, it is difficult to show the<br />

rare, specific abnormalities of basement<br />

membrane components, integrins or desmosomes<br />

in part of the mucosa, in the absence of tufts.<br />

Another difficulty is related to the infiltration of<br />

the lamina propria by T cells, a finding that would<br />

support the hypothesis of an immune-related<br />

enteropathy, especially when tufts are missing.<br />

One might speculate that defective cell adhesion<br />

increases intestinal permeability, with a subse-


8<br />

Microvillus inclusion disease and epithelial dysplasia<br />

quent inflammatory reaction. In a mouse model of<br />

dysfunctional E-cadherin, this primary disorder of<br />

epithelial integrity was responsible for secondary<br />

T-cell-mediated mucosal damage. 38 Murch et al 39<br />

described this type of lesion in infants with epithelial<br />

dysplasia. They showed that inhibition of<br />

secondary T-cell activation significantly improved<br />

enteral absorption and decreased crypt cell proliferation,<br />

without, however, permanent resolution<br />

of the severe diarrhea.<br />

Associated disorders<br />

Several cases of IED have been reported as being<br />

associated with phenotypic abnormalities (e.g.<br />

Dubowitz syndrome or malformation syndromes).<br />

40,41 An association between congenital<br />

IDI and choanal atresia was recently reported in<br />

four children. 40 In our experience, we have<br />

observed malformations including rectal atresia,<br />

choanal atresia or esophageal atresia. We recently<br />

reported the association with a non-specific punctiform<br />

keratitis in more than 50% of patients with<br />

clinically and histologically recognized IED. 42<br />

Therefore, the diagnostic work-up should include<br />

a systematic ophthalmological examination by an<br />

experienced specialist. The latter association is<br />

intriguing, since punctiform keratitis is also an<br />

epithelial disease; thus, studying this condition<br />

might help to elucidate the molecular mechanisms<br />

of the intestinal epithelial disease. The fact that<br />

some children have no ophthalmological symptoms<br />

confirms the heterogeneity of the disease.<br />

Interestingly, Lachaux et al have reported a case of<br />

a newborn presenting with <strong>pylori</strong>c atresia and<br />

intractable diarrhea. 43 Light microscopic examination<br />

showed extensive desquamation from fundus<br />

to rectum, with only a few epithelial cells remaining<br />

at the base of the crypts. Electron microscopy<br />

of the gut revealed normal desmosomes, but a<br />

cleavage located between the lamina propria and<br />

the basal pole of the enterocytes. This firstdescribed<br />

disease is supposed to be related to<br />

congenital deficiency of α6β4 integrin. This integrin<br />

is known to be defective in epidermolysis<br />

bullosa, in which gross epidermal shedding<br />

occurs, although the cutaneous expression of α6β4<br />

integrin appeared normal in this case of IDI. This<br />

is consistent with a mutation within an intestinal<br />

isoform of the α6β4 integrin, or a deficiency of a<br />

related and immunohistochemically cross-reactive<br />

intestinal integrin. 44,45 Rather like epidermolysis<br />

bullosa, which shares several similarities with<br />

deficiency of α6β4 integrin at the ultrastructural<br />

and possibly molecular level, there are likely to be<br />

several distinct mutations that can cause this<br />

phenotype.<br />

Pathophysiology and mode of transmission<br />

In infants with epithelial dysplasia with characteristic<br />

tufts, we have reported abnormal laminin<br />

and heparan sulfate proteoglycan deposition on<br />

the basement membrane, compared to biopsy<br />

specimens from patients with celiac disease or<br />

autoimmune enteropathy. 11 Basement membrane<br />

molecules are involved in epithelial–mesenchymal<br />

cell interactions, which are instrumental in<br />

intestinal development and differentiation. 46–49<br />

Alterations suggestive of abnormal cell–cell and<br />

cell–matrix interactions were seen in patients<br />

with epithelial dysplasia without any evidence of<br />

abnormalities in epithelial cell polarization and<br />

proliferation. 37 Alterations included abnormal<br />

distribution of adhesion molecules α2β1 integrin<br />

along the crypt–villus axis. The α2β1 integrin is<br />

involved in the interaction of epithelial cells to<br />

various basement membrane components, such as<br />

laminin and collagen. To date, the pathophysiological<br />

mechanisms resulting in an increased<br />

immunohistochemical expression of desmoglein,<br />

and the ultrastructural changes of desmosomes<br />

remain unclear (Figure 1.7). 37 Tufts correspond to<br />

non-apoptotic epithelial cells at the villous tips<br />

that are no longer in contact with the basement<br />

membrane. It can be speculated that a defect of<br />

normal enterocyte apoptosis at the end of their<br />

lifespan, or an altered cell–cell contact, is responsible<br />

for this effect. The primary or secondary<br />

nature of the formation of tufts remains, however,<br />

to be determined.<br />

To date, the genetic origin of this disorder is<br />

suspected from the clear association of parental<br />

consanguinity and/or affected siblings. These<br />

features suggest an autosomal recessive transmission.<br />

The gene involved in this congenital inherited<br />

autosomal recessive disease is not yet<br />

identified. This enteropathy appears more<br />

common than MVID, especially within the<br />

Middle-Eastern population.


Outcome<br />

This neonatal diarrhea, which resists all treatments,<br />

requires permanent PN. However, it seems<br />

that some infants have a rather milder phenotype<br />

than others. 50 Thus, because of partial intestinal<br />

function and limited amount of stool output, some<br />

patients only need partial long-term PN, with three<br />

to four weekly infusions. Careful monitoring<br />

should be performed, in order to avoid progressive<br />

growth retardation. In most patients, the severity<br />

of intestinal malabsorption and diarrhea make<br />

them totally dependent on daily long-term PN<br />

with subsequent risk of complications. This is<br />

therefore another indication for intestinal transplantation.<br />

51–53<br />

Other diseases of the intestinal<br />

epithelium<br />

The classification of IDI is probably incomplete,<br />

since other forms with abnormal small-bowel<br />

mucosa have been described. These include mitochondrial<br />

DNA rearrangements; congenital enterocyte<br />

heparan sulfate deficiency, phosphomannose<br />

isomerase deficiency; and a<br />

carbohydrate-deficient glycoprotein syndrome<br />

with hepatic-intestinal presentation. 54–59 Rare<br />

diseases involving the immune system and smallbowel<br />

mucosa, 60 or severe intractable enterocolitis<br />

of infancy, 6 seem clearly different from the<br />

above-described diseases. The so-called ‘phenotypic<br />

diarrhea’ that is an IDI syndrome associated<br />

with phenotypic abnormalities and immune deficiency<br />

is one of these rare diseases recently<br />

reported. The patients present with diarrhea starting<br />

within the first 6 months of life (


10<br />

Microvillus inclusion disease and epithelial dysplasia<br />

3. Guarino A, Spagnulo MI, Russo S et al. Etiology and<br />

risk factors of severe and protracted diarrhea. J Pediatr<br />

Gastroenterol Nutr 1995; 20: 173–178.<br />

4. Catassi C, Fabiani E, Spagnuolo MI et al. Severe and<br />

protracted diarrhea: results of the 3-year SIGEP multicenter<br />

survey. J Pediatr Gastroenterol Nutr 1999; 29:<br />

63–68.<br />

5. Phillips AD, Jenkins P, Raafat F, Walker-Smith JA.<br />

Congenital microvillus atrophy: specific diagnostic<br />

features. Arch Dis Child 1985; 60: 135–140.<br />

6. Unsworth DJ, Walker-Smith JA. Auto-immunity in diarrheal<br />

disease. J Pediatr Gastroenterol Nutr 1985; 4:<br />

375–380.<br />

7. Goulet O, Besnard M, Girardet JP, Lachaux A, Sarles J<br />

and the French Speaking Group of Hepatology,<br />

Gastroenterology and Nutrition. Clin Nutr 1998; 17: 9<br />

(A).<br />

8. Sanderson IR, Risdon RA, Walker-Smith JA. Intractable<br />

ulcerating enterocolitis of infancy. Arch Dis Child 1991;<br />

65: 295–299.<br />

9. Cuenod B, Brousse N, Goulet O et al. Classification of<br />

intractable diarrhea in infancy using clinical and<br />

immunohistological criteria. Gastroenterology 1990; 99:<br />

1037–1043.<br />

10. Goulet O, Brousse N, Canioni D et al. Syndrome of<br />

intractable diarrhoea with persistent villus atrophy in<br />

early childhood: a clinicopathological survey of 47<br />

cases. J Pediatr Gastroenterol Nutr 1998; 26: 151–161.<br />

11. Goulet O, Kedinger M, Brousse N et al. Intractable diarrhea<br />

of infancy: a new entity with epithelial and basement<br />

membrane abnormalities. J Pediatr 1995; 127:<br />

212–219.<br />

12. Girault D, Goulet O, Ledeist F et al. Intractable diarrhea<br />

syndrome associated with phenotypic abnormalities<br />

and immune deficiency. J Pediatr 1994; 125: 36–42.<br />

13. Davidson GP, Cuiz E, Hamilton JR, Gall DG. Familial<br />

enteropathy: a syndrome of protracted diarrhea from<br />

birth, failure to thrive, and hypoplastic villus atrophy.<br />

Gastroenterology 1978; 75: 783–790<br />

14. Schmitz J, Ginies JL, Arnaud-Battandier F et al.<br />

Congenital microvillus atrophy, a rare cause of neonatal<br />

intractable diarrhea. Pediatr Res 1982; 16: 1041a.<br />

15. Phillips AD, Schmitz J. Familial microvillus atrophy: a<br />

clinicopathological survey of 23 cases. J Pediatr<br />

Gastroenterol Nutr 1992; 14: 380–396.<br />

16. Homberg C, Perheentupa J. Congenital chloride diarrhoea.<br />

Ergeb Inn Med Kinderheilkd 1982; 49: 138–172.<br />

17. Booth IW, Stange G, Murer H et al. Defective jejunal<br />

brush-border Na+/H+ exchange: a cause of congenital<br />

secretory diarrhoea. Lancet 1985; 1: 1066-1069.<br />

18. Phillips AD, Jenkins P, Raafat F, Walker-Smith JA.<br />

Congenital microvillus atrophy: specific diagnostic<br />

features. Arch Dis Child 1985; 60: 135–140.<br />

19. Bell SW, Kerner JA Jr, Sibley RK. Microvillus inclusion<br />

disease. The importance of electron microscopy for<br />

diagnosis. Am J Surg Pathol 1991; 15: 1157–1164.<br />

20. Phillips AD, Szfranski M, Man L-Y, Wall W. Periodic<br />

acid Schiff staining abnormality in microvillus atrophy:<br />

photometric and ultrastructural studies. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 34–42.<br />

21. Phillips A, Fransen J, Hauri HP, Sterchi E. The constitutive<br />

exocytotic pathway in microvillus atrophy. J Pediatr<br />

Gastroenterol Nutr 1993; 17: 239–246<br />

22. Groisman GM, Amar M, Livne E. CD10: a valuable tool<br />

for the light microscopic diagnosis of microvillus inclusion<br />

disease (familial microvillus atrophy). Am J Surg<br />

Pathol 2002; 26: 902–907.<br />

23. Youssef N, Canioni D, Ruemmele F. CD-10 expression in<br />

microvillous inclusion disease. J Pediatr Gastroenterol<br />

Nutr 2003; 36: 563(A).<br />

24. Raafat F, Green NJ, Nathavitharana KA, Booth IW.<br />

Intestinal microvillus dystrophy: a variant of microvillus<br />

inclusion disease or a new entity? Hum Pathol 1994;<br />

25: 1243–1248.<br />

25. Carruthers L, Dourmaskhin R, Phillips A. Disorders of<br />

the cytoskeleton of the enterocyte. Clin Gastroenterol<br />

1986; 15 : 105–120.<br />

26. Phillips A, Brown A, Murch S, Walker-Smith JA.<br />

Histochemical studies of microvillus atrophy: acetylated<br />

sialic acid residues accumulate in the epithelium. J<br />

Pediatr Gastroenterol Nutr 1999; 28: 565 (abstract).<br />

27. Nathavitharana KA, Green NJ, Raafat F, Booth IW.<br />

Siblings with microvillus inclusion disease. Arch Dis<br />

Child 1994; 71: 71–73.<br />

28. Heinz-Erian P, Schmidt H, Le Merrer M et al. Congenital<br />

microvillus atrophy in a girl with autosomal dominant<br />

hypochondroplasia. J Pediatr Gastroenterol Nutr 1999;<br />

28: 203–205.<br />

29. Michail S, Collins JF, Xu H et al. Abnormal expression<br />

of brush-border membrane transporters in the duodenal<br />

mucosa of two patients with microvillus inclusion<br />

disease. J Pediatr Gastroenterol Nutr 1998; 27: 536–542.<br />

30. Oliva MM, Perman JA, Saavedra JM et al. Successful<br />

intestinal transplantation for microvillus inclusion<br />

disease. Gastroenterology 1994; 106: 771–774.<br />

31. Herzog D, Atkinson P, Grant D et al. Combined<br />

bowel–liver transplantation in an infant with microvillus<br />

inclusion disease. J Pediatr Gastroenterol Nutr 1996;<br />

22: 405–408.<br />

32. Randak C, Langnas AN, Kaufman SS et al. Pretransplant<br />

management and small bowel–liver transplantation in<br />

an infant with microvillus inclusion disease. J Pediatr<br />

Gastroenterol Nutr 1998; 27: 333–337.<br />

33. Bunn SK, Beath SV, McKeirnan PJ et al. Treatment of<br />

microvillus inclusion disease by intestinal transplantation.<br />

J Pediat Gastroenterol Nutr 2000; 31: 176–180.<br />

34. Croft NM, Howatson AG, Ling SC et al. Microvillus<br />

inclusion disease: an evolving condition. J Pediatr<br />

Gastroenterol Nutr 2000; 31: 185–189.<br />

35. Goulet O, Michel JL, Jobert A et al. Small bowel transplantation<br />

alone or with the liver in children: changes<br />

by using FK506. Transplant Proc 1998; 30: 1569–1570.<br />

36. Reifen RM, Cutz E, Griffiths AM et al. Tufting enteropathy:<br />

a newly recognized clinicopathological entity associated<br />

with refractory diarrhea in infants. J Pediatr<br />

Gastroenterol Nutr 1994; 18: 379–385.<br />

37. Patey N, Scoazec JY, Cuenod-Jabri B et al. Distribution<br />

of cell adhesion molecules in infants with intestinal<br />

epithelial dysplasia (tufting enteropathy).<br />

Gastroenterology 1997; 113: 833–843.<br />

38. Hermiston ML, Gordon JI. Inflammatory bowel disease<br />

and adenomas in mice expressing a dominant negative<br />

N-cadherin. Science 1995; 270: 1203–1207.<br />

39. Murch S, Graham A, Vermault A et al. Functionally<br />

significant secondary inflammation occurs in a primary<br />

epithelial enteropathy. J Pediatr Gastroenterol Nutr 1997;<br />

24: 467.<br />

40. Krantz M, Jansson U, Rectors S et al. Hereditary<br />

intractable diarrhea with choanal atresia. A new familial<br />

syndrome. J Pediatr Gastroenterol Nutr 1997; 24: 470.<br />

41. Abely M, Hankard GF, Hugot JP et al. Intractable infant<br />

diarrhea with epithelial dysplasia associated with polymalformative<br />

syndrome. J Pediatr Gastroenterol Nutr<br />

1998; 27: 348–352.<br />

42. Djeddi D, Verkarre V, Talbotec C et al. Tufting enteropathy<br />

and associated disorders. J Pediatr Gastroenterol<br />

Nutr 2002; 34: 446(A).<br />

43. Lachaux A, Bouvier R, Loras I et al. a6b4 integrin deficiency.<br />

A new aetiology for protracted diarrhoea in<br />

infancy. J Pediatr Gastroenterol Nutr 1997; 24: 470.


44. Beaulieu JF. Differential expression of the VLA family of<br />

integrins along the crypt–villus axis in the human small<br />

intestine. J Cell Sci 1992; 102: 427–436.<br />

45. Simon-Assmann P, Duclos B, Orian-Rousseau V et al.<br />

Differential expression of laminin isoforms and a6-b4<br />

integrin subunits in the developing human and mouse<br />

intestine. Dev Dynamics 1994; 201: 71–85.<br />

46. Simon-Assmann P, Bouziges F, Vigny M, Kedinger M.<br />

Origin and deposition of basement membrane. Heparan<br />

sulfate proteoglycan in the developing intestine. J Cell<br />

Biol 1989; 109: 1837–1848.<br />

47. Simo P, Simon-Assmann P, Bouziges F et al. Changes in<br />

the expression of laminin during intestinal development.<br />

Development 1991; 112: 477–487.<br />

48. Simo P, Bouziges F, Lissitzky JC et al. Dual and asynchronous<br />

deposition of laminin chains at the epithelial<br />

mesenchymal interface in the gut. Gastroenterology<br />

1992; 102: 1835–1845.<br />

49. Simon-Assmann P, Kedinger M. Heterotypic cellular<br />

cooperation in gut morphogenesis and differentiation.<br />

Cell Biol 1993; 4: 221–230.<br />

50. Cameron DJS, Barnes GL. Successful pregnancy<br />

outcome in tufting enteropathy. J Pediatr Gastroenterol<br />

Nutr 2003; 36: 158.<br />

51. Lacaille F, Cuenod B, Colomb V et al. Successful<br />

combined liver and small bowel transplantation in a<br />

child with epithelial dysplasia. J Pediatr Gastroenterol<br />

Nutr 1998; 2: 230–233.<br />

52. Goulet O. Intestinal transplantation. Curr Opin Clin<br />

Nutr Metab Care 1999; 2: 315–321.<br />

53. Paramesh AS, Fishbein T, Tschernia A et al. Isolated<br />

small bowel transplantation for tufting enteropathy. J<br />

Pediatr Gastroenterol Nutr 2003; 36: 138–140.<br />

54. Verloes A, Lombet J, Lambert Y et al. Tricho-hepatoenteric<br />

syndrome: further delineation of a distinct<br />

syndrome with neonatal hemochromatosis phenotype,<br />

intractable diarrhea, and hair anomalies. Am J Med Gen<br />

1997; 68: 391–395.<br />

References 11<br />

55. De Vries E, Visser DM, Van Dongen JJ et al. Oligoclonal<br />

gammopathy in ‘phenotypic diarrhea’. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 349–350<br />

56. Cormier-Daire V, Bonnefont JP, Rustin P et al.<br />

Mitochondrial DNA rearrangements with onset as<br />

chronic diarrhea with villus atrophy. J Pediatr 1994;<br />

124: 63–70.<br />

57. Murch S, Winyard PJD, Koletzko S et al. Congenital<br />

enterocyte heparan sulphate deficiency with massive<br />

albumin loss, secretory diarrhoea and malnutrition.<br />

Lancet 1996; 347: 1299–1301.<br />

58. Jaeken J, Matthijs G, Saudubray JM et al.<br />

Phosphomannose isomerase deficiency: a carbohydratedeficient<br />

glycoprotein syndrome with hepatic–intestinal<br />

presentation. Am J Hum Genet 1998; 62: 1535–1539.<br />

59. Oren A, Houwen RH. Phosphomannose isomerase deficiency<br />

as the cause of protein-losing enteropathy and<br />

congenital liver fibrosis. J Pediatr Gastroenterol Nutr<br />

1999; 29: 231–232.<br />

60. Smith LJ, Szymanski W, Foulston C et al. Familial<br />

enteropathy with villus edema and immunoglobulin G2<br />

subclass deficiency. J Pediatr 1994; 125: 541–548.<br />

61. Itin PH, Pittelkow MR. Trichothiodystrophy: review of<br />

sulfur-deficient brittle hair syndromes and association<br />

with the ectodermal dysplasia. J Am Acad Dermatol<br />

1990; 22: 705–717.<br />

62. Happle R, Traupe H, Gröbe H, Bonsmann G. The Tay<br />

syndrome (congenital ichthyosis with trichothiodystrophy).<br />

Eur J Pediatr 1984; 141: 147–152.<br />

63. Stefanini M, Vermeulen W, Weeda G et al. A new<br />

nucleotide-excision-repair gene associated with the<br />

disorder trichothiodystrophy. Am J Hum Genet 1993; 53:<br />

817–821.<br />

64. Mariani E, Facchini A, Honorati MC et al. Immune<br />

defects in families and patients with xeroderma pigmentosum<br />

and trichothiodystrophy. Clin Exp Immunol 1992;<br />

88: 376–382.


2<br />

Introduction<br />

Congenital problems of the<br />

gastrointestinal tract<br />

Nigel Hall and Agostino Pierro<br />

Congenital abnormalities of the gastrointestinal<br />

(GI) tract are relatively common. Owing to their<br />

nature, they frequently require surgical correction.<br />

On occasion this must be undertaken as a matter of<br />

emergency in order to avoid catastrophic intestinal<br />

ischemia and necrosis, resulting in loss of bowel or<br />

damage to a secondary organ system. It is not<br />

possible in the realms of one chapter to describe in<br />

detail all of the variants of congenital GI tract<br />

abnormalities that may be encountered and the<br />

precise nature of the treatment options available.<br />

Therefore, what follows is an overview of the most<br />

common conditions encountered and those which<br />

require intervention as a matter of urgency. For<br />

ease of understanding we commence with the<br />

upper GI tract and continue in a caudal direction.<br />

Conditions affecting the upper<br />

gastrointestinal tract<br />

Disorders of the mouth and oral cavity<br />

Cleft lip and palate is one of the more common<br />

congenital anomalies with an incidence of about 1<br />

in 600 live births. 1,2 The etiology is probably<br />

multifactorial: a genetic component is implicated<br />

as are a wide variety of exogenous factors and up<br />

to 50% of infants may have other associated anomalies.<br />

3 The presentation may be during routine<br />

antenatal scans, although some cases do not<br />

become apparent until birth. Whilst the esthetic<br />

appearance may be the most obvious, respiratory<br />

and feeding difficulties associated with clefts carry<br />

greater medical significance and in some cases<br />

may require early intervention. The long-term<br />

management of these disorders is best in the<br />

setting of a specialist multidisciplinary team in<br />

order to achieve a satisfactory functional and<br />

cosmetic repair.<br />

Esophageal atresia and tracheoesophageal<br />

fistula<br />

This complex group of anomalies, with an incidence<br />

of 1 in 2440–4500 live births, 4,5 results from<br />

failure of correct division of the tracheal<br />

primordium from the esophagus during early<br />

embryonic development. The precise etiology is<br />

unknown with a number of embryological theories<br />

proposed to explain the different variants of this<br />

anomaly. There is a high incidence of co-existing<br />

abnormalities including the VACTERL syndrome,<br />

CHARGE association and isolated cardiovascular<br />

anomalies. 6<br />

Classification<br />

A number of classification systems have been<br />

proposed over the years. The abbreviated list in<br />

Table 2.1 describes the most commonly encountered<br />

anatomical variants (Figure 2.1). The<br />

Table 2.1 Classification of esophageal<br />

atresia (EA)/tracheoesophageal fistula (TEF)<br />

anomalies and frequency 6<br />

Type of lesion Frequency (%)<br />

EA and distal TEF 88.8<br />

Isolated EA 7.3<br />

H-type fistula 4.2<br />

Distal and proximal TEF 2.8<br />

Proximal TEF 1.1<br />

13


14<br />

Congenital problems of the gastrointestinal tract<br />

Figure 2.1 Common anatomical variants of esophageal<br />

atresia (EA)/tracheoesophageal fistula (TEF) anomalies. (a)<br />

EA with distal TEF; (b) isolated EA with no TEF; (c) H-type<br />

TEF; (d) proximal and distal TEF; (e) EA with proximal TEF.<br />

Spitz classification 6 (Table 2.2), incorporating birth<br />

weight and presence of major congenital heart<br />

disease status, may be useful in predicting survival.<br />

Clinical features<br />

Esophageal atresia is commonly associated with<br />

maternal polyhydramnios. The diagnosis may be<br />

made in the antenatal period, particularly if there<br />

is no tacheoesophageal fistula. In the postnatal<br />

period, symptoms include excessive salivation,<br />

feeding difficulties, respiratory distress, and<br />

cyanotic episodes. Cases of esophageal atresia<br />

(with the exception of the rare esophageal atresia<br />

with double fistula) can be confirmed by failure of<br />

Table 2.2 Spitz classification of esophageal<br />

atresia (EA)/tracheoesophageal fistula (TEF)<br />

anomalies and outcome 6<br />

Group Clinical features Survival (%)<br />

I BW ≥1500g with no major CHD 97<br />

II BW


Obstructive lesions of the duodenum, jejunum<br />

and ileum<br />

The most common congenital conditions affecting<br />

the duodenum, jejunum and ileum all result in<br />

partial or complete gastrointestinal obstruction.<br />

The presenting features and investigations recommended<br />

to diagnose the underlying abnormality<br />

are similar for all conditions. The clinical features<br />

and investigations of these conditions are therefore<br />

presented first, followed by a description of each<br />

type of abnormality and the recommended treatment<br />

options.<br />

Clinical features<br />

Obstructive lesions of the small intestine from the<br />

pylorus down to the ileocecal valve may give rise<br />

to polyhydramnios in the antenatal period which<br />

is detectable by antenatal ultrasonography. As a<br />

general rule, the more proximal the lesion the<br />

more severe the degree of polyhydramnios; distal<br />

ileal lesions may be present in the absence of polyhydramnios.<br />

7 The list of differential diagnoses<br />

giving rise to polyhydramnios is extensive, and it<br />

is rare for an obstructive lesion of the GI tract to be<br />

confirmed before birth.<br />

Following birth, the most common and important<br />

clinical manifestation of obstructive lesions of the<br />

GI tract is bile-stained vomiting. Vomiting with<br />

truly bilious staining is abnormal in the neonatal<br />

period and always requires investigation. Lesions<br />

in the duodenum and jejunum usually result in<br />

bilious vomiting within hours. In addition, the<br />

abdomen may appear empty or even scaphoid and<br />

visible gastric peristalsis may be observed. Lesions<br />

lower in the ileum result in a distended abdomen<br />

if the obstruction is complete, and there may be<br />

failure to pass meconium. Obstructive lesions may<br />

also give rise to intestinal perforation in the neonatal<br />

period and occasionally antenatally. In all<br />

cases of neonatal intestinal obstruction, infants<br />

become progressively hypovolemic and are prone<br />

to circulatory and respiratory collapse. They<br />

require fluid resuscitation and may require ventilatory<br />

support. GI tract obstruction should therefore<br />

be considered in any infant who is dehydrated,<br />

especially if there is a history of vomiting.<br />

Stenotic lesions of the small bowel in which the<br />

obstruction is incomplete may give rise to<br />

Conditions affecting the upper gastrointestinal tract 15<br />

increased diagnostic difficulty. Affected infants<br />

often present with intermittent vomiting and<br />

episodes of partial obstruction. They eventually<br />

fail to thrive or develop complete obstruction, at<br />

which stage they are fully investigated and the<br />

diagnosis becomes apparent.<br />

Intestinal malrotation is considered separately, as<br />

it may present with a spectrum of clinical scenarios<br />

depending on the degree of intestinal obstruction<br />

or midgut volvulus or both. The clinical<br />

pictures of all types of abnormal rotation are those<br />

of acute or chronic intestinal obstruction and/or<br />

acute or chronic abdominal pain suggestive of<br />

intestinal ischemia. True malrotation typically<br />

presents in the first year of life with symptoms of<br />

upper gastrointestinal tract obstruction including<br />

vomiting, which is usually bile-stained. A coexisting<br />

volvulus may be suspected by abdominal<br />

pain, peritonitis and hypovolemic shock associated<br />

with intestinal ischemia. However, these<br />

signs may be relatively non-specific in the young<br />

infant. Malrotation may also present later in life<br />

with similar symptoms.<br />

Investigations<br />

The aim of investigating cases of suspected<br />

obstruction of the small intestine is two-fold: first,<br />

to identify the nature and anatomical location of<br />

the lesion, to allow for planning of correct treatment;<br />

and second, to identify patients with<br />

malrotation in whom there is a risk of midgut<br />

volvulus and intestinal ischemia. These cases<br />

require urgent surgical intervention to reduce the<br />

risk of potentially catastrophic intestinal necrosis.<br />

The history and examination may give clues as to<br />

the location of the lesion as described above. An<br />

abdominal X-ray may simply confirm the presence<br />

of dilated intestinal loops but may also give further<br />

clues in some cases. A double-bubble appearance<br />

on abdominal X-ray with a lack of air in the distal<br />

intestine (Figure 2.2) is characteristic of duodenal<br />

obstruction. Multiple air-filled loops of proximal<br />

bowel often with air–fluid levels along with a<br />

paucity or complete absence of gas in the distal<br />

bowel is highly suggestive of obstruction of the<br />

ileum. Intestinal perforation if present will usually<br />

be apparent on abdominal X-ray, and in the rare<br />

cases of antenatal perforation there may be widespread<br />

or localized flecks of calcification representing<br />

calcified meconium within the peritoneum.


16<br />

Congenital problems of the gastrointestinal tract<br />

In cases in which the diagnosis is not clear on<br />

abdominal X-ray or in which midgut malrotation<br />

or volvulus is suspected, a limited upper gastrointestinal<br />

contrast study is indicated. The classical<br />

finding in cases of malrotation is that the duodenojejunal<br />

flexure lies to the right side of the<br />

spine instead of in its normal left-sided position<br />

(Figure 2.3). This finding should prompt urgent<br />

surgical treatment, because of the risk of co-existing<br />

midgut volvulus. The contrast study may also<br />

identify the presence of a stenotic segment or<br />

complete obstruction.<br />

Cases of lower ileal stenosis or atresia are often<br />

more difficult to diagnose and a contrast enema is<br />

invaluable in distinguishing between ileal and<br />

colonic obstruction and could be therapeutic in<br />

cases of meconium ileus (see below).<br />

Conditions affecting the duodenum<br />

Duodenal atresia, duodenal stenosis and annular<br />

pancreas are the most common congenital condi-<br />

Figure 2.2 Abdominal X-ray of an infant with duodenal<br />

atresia showing the ‘double bubble’ appearance<br />

characteristic of duodenal obstruction.<br />

tions to affect the duodenum. All are capable of<br />

giving rise to duodenal obstruction. The incidence<br />

is reported to be between 1 in 5000 and 1 in 10 000<br />

live births. 8<br />

Explanations of the etiology of duodenal atresias<br />

are not universally accepted. Unlike atresias of the<br />

ileum, they are not thought to be due to vascular<br />

accidents and the most widely accepted explanation<br />

is that of failure of recanalization of the<br />

intestinal lumen during early embryonic development.<br />

Classification<br />

There are four basic types of duodenal obstruction<br />

(Figure 2.4). In type 1 there is a stenosis of the<br />

duodenum resulting from a diaphragm or web<br />

partially or totally occluding the lumen. Owing to<br />

the incomplete nature of the obstruction, cases<br />

Figure 2.3 Upper gastrointestinal contrast study of a<br />

case of malrotation. The contrast is seen within the<br />

duodenum (D) and flowing into the upper jejunum (J),<br />

both of which lie completely to the right of the midline.


may present in childhood rather than in the<br />

neonatal period. In type 2 duodenal atresia, the<br />

proximal and distal segments end blindly but<br />

remain connected by a fibrous cord. There is<br />

complete separation of the bowel segments in type<br />

3, and type 4 comprises extrinsic obstruction due<br />

to an annular pancreas although there may be an<br />

associated atretic segment. Multiple atresias are<br />

said to occur in approximately 15% of cases. 9<br />

Treatment<br />

The principles of treatment are to restore intestinal<br />

continuity whilst avoiding interference with the<br />

ductal system draining the pancreas and biliary<br />

tree. This is best achieved using a duodenoduo-<br />

Figure 2.4 Variants of duodenal atresia. (a) Type 1<br />

atresia due to an internal diaphragm; (b) type 2 atresia<br />

with blind-ending loops remaining connected by a fibrous<br />

cord; (c) type 3 atresia with blind ends completely separated;<br />

(d) type 4 – duodenal obstruction due to an annular<br />

pancreas.<br />

Conditions affecting the upper gastrointestinal tract 17<br />

denostomy in which the obstructed segment is<br />

bypassed by the proximal segment being joined<br />

directly to the distal segment. Following surgery<br />

the long-term gastrointestinal results are good. 10<br />

Conditions affecting the ileum and jejunum<br />

The main congenital problems directly affecting<br />

the small intestine from the duodenojejunal flexure<br />

down to the cecum are atresia and stenosis.<br />

Jejunoileal atresia occurs more commonly than its<br />

duodenal counterpart, with an incidence varying<br />

from 1 in 330 to 1 in 3000 livebirths. 11 Such lesions<br />

are one of the most common causes of neonatal<br />

intestinal obstruction. The major difference<br />

between atresias of the ileojejunum and those of<br />

the duodenum is in their etiology. It is postulated<br />

that atresia or stenosis of the jejunum and ileum is<br />

the result of a localized intravascular accident<br />

during intrauterine life. Subsequent ischemic<br />

necrosis and reabsorption of the affected segment<br />

or segments results in a contracted scarred bowel<br />

wall leading to stenosis at one end of the spectrum<br />

to a complete intestinal and mesenteric defect at<br />

the other. Fetal animal experiments have confirmed<br />

this hypothesis, at least in part, 12 and the absence<br />

of other congenital abnormalities found in association<br />

with jejunoileal stenoses and atresias supports<br />

the localized vascular accident theory.<br />

Classification<br />

Morphological classification of these lesions<br />

allows different surgeons and centers to compare<br />

outcomes and is also of therapeutic and prognostic<br />

value. The most commonly accepted system is that<br />

proposed by Louw13 and modified by Grosfeld et<br />

al. 14 Whether the lesion is classified as ileal or<br />

jejunal is determined by the most proximal<br />

affected segment (Figure 2.5).<br />

Stenosis is a localized narrowing of the lumen<br />

without any break in the continuity or mesenteric<br />

defect. The intestinal wall may be thickened and<br />

rigid at the stenotic site and there is a small, often<br />

minute, lumen. The overall intestinal length is not<br />

shortened. Type 1 atresia is the result of a membranous<br />

web occluding the lumen with no mesenteric<br />

defect and no intestinal shortening. The lumen is<br />

usually completely occluded and the proximal<br />

bowel is therefore dilated but remaining in continuity<br />

with the collapsed distal segment. Type 2


18<br />

Congenital problems of the gastrointestinal tract<br />

Figure 2.5 Variants of ileal atresia. (a) Type 1 due to an internal web (not shown) with no mesenteric defect; (b) type 2<br />

atresia with blind ends joined by a fibrous cord; (c) type 3(a) – blind ends separated with a mesenteric defect; (d) type 3(b),<br />

in which the ileum is coiled like an ‘apple peel’ around a single vessel and completely separated from the proximal dilated<br />

duodenum; (e) type 4 or multiple atresias.<br />

atresia arises from a complete obliteration of the<br />

intestinal segment into a fibrous cord, which joins<br />

two blind ends and runs in the free edge of the<br />

mesentery. There is no mesenteric defect and once<br />

again the total bowel length is usually normal. In<br />

both type 3(a) and type 3(b) atresia the intestine is<br />

likely to be shortened and this may have significant<br />

clinical consequences. Type 3(a) atresia<br />

consists of blind-ending proximal and distal bowel<br />

with no connection and an often large mesenteric<br />

defect. The blind ends are often physiologically<br />

abnormal with decreased or absent peristaltic<br />

activity, which may give rise to torsion, distension<br />

or perforation. Type 3(b) atresia, also known as<br />

apple peel atresia because of its gross morphology,<br />

may involve massive intestinal loss. It consists of<br />

intestinal atresia near the ligament of Treitz, oblit-<br />

eration of the superior mesenteric artery (SMA)<br />

beyond the origin of the middle colic branch and<br />

absence of the dorsal mesentery. The remaining<br />

intestine is coiled helically (like an apple peel)<br />

around a single perfusing vessel, often has<br />

impaired vascularity and is almost inevitably<br />

short. Furthermore, there may be additional<br />

segments of type 1 or 2 atresia within the apple<br />

peel segment. Such a configuration probably arises<br />

from occlusion of the SMA due to thrombus,<br />

embolus or strangulation as part of a midgut volvulus.<br />

In type 4 atresia there are multiple atretic<br />

segments and the intestine may resemble a string<br />

of sausages. Overall bowel length is usually shortened<br />

and the intestine grossly dilated. It has been<br />

proposed that the etiology of type 4 atresia may be<br />

due to failure of recanalization of solid epithelial-


ization throughout the length of the intestine<br />

rather than from multiple single vascular events.<br />

Whilst it is generally accepted that stenosis and<br />

atresia of types 1, 2 and 3(b) are the result of<br />

intrauterine vascular accidents, a genetic component<br />

has been suggested in types 3(b) and 4.<br />

Treatment<br />

The mainstay of surgical treatment for this type of<br />

lesion is resection of the atretic or stenotic segment<br />

and primary anastomosis, with closure of the<br />

mesenteric defect. The proximal intestinal<br />

segment is usually dilated and functionally abnormal<br />

with absent or ineffective peristalsis. This<br />

dilated proximal segment is excised along with a<br />

short segment distal to the stenosis or atresia. It is<br />

essential to establish patency of the distal bowel<br />

by irrigation or wash-out of the intestine and<br />

subsequently a primary anastomosis is performed.<br />

There is a balance to be struck between the length<br />

of the dilated proximal segment resected and the<br />

risk of leaving the infant with a short length of<br />

small bowel. It is almost inevitable that the caliber<br />

of the proximal bowel will be greater than that of<br />

the distal intestine, and a number of techniques<br />

including fishtailing and tapering exist to assist<br />

the anastomosis in such circumstances.<br />

Outcome following intestinal atresia is dependent<br />

primarily on the length of remaining intestine and<br />

the presence of the ileocecal valve. Short bowel<br />

syndrome is defined as the presence of less than<br />

75cm of the small intestine or 30% of the<br />

predicted intestinal length in a premature<br />

infant. 15,16 Outcomes following short bowel<br />

syndrome vary and there is a high level of dependence<br />

on parenteral nutrition. However, intestinal<br />

adaptation can occur such that more than 80% of<br />

babies with short bowel syndrome do eventually<br />

become entirely enterally nourished. 17<br />

Intestinal malrotation<br />

The incidence of intestinal malrotation is difficult<br />

to establish as not all affected patients develop<br />

symptoms, but autopsy studies estimate the incidence<br />

at approximately 1 in 500.<br />

The traditional embryological basis for disorders<br />

of intestinal rotation is that of abnormal position-<br />

Conditions affecting the upper gastrointestinal tract 19<br />

ing of the intestinal loops in relation to one<br />

another as they return to the abdominal cavity<br />

from the yolk sac. During normal development the<br />

midgut rotates through 270º so that the duodenum<br />

lies posterior to the colon and the duodenojejunal<br />

flexure is to the left of the midline. A consistent<br />

finding in cases of malrotation is abnormal positioning<br />

of the duodenum. In 1995, an alternative<br />

hypothesis was proposed based on animal<br />

studies. 18 Kluth et al proposed that malrotation is<br />

the result of failure of localized growth of the<br />

duodenal loop rather than a disorder of rotation.<br />

The term ‘malrotation’ covers a spectrum of<br />

anatomical abnormalities. In non-rotation the<br />

duodenojejunal flexure lies to the right of the spine<br />

along with most of the small intestine. The cecum<br />

and colon are typically on the left side of the<br />

abdominal cavity. Adhesions formed between<br />

loops of bowel or the intestine and abdominal wall<br />

are usually responsible for obstructive symptoms<br />

at presentation. In malrotation the distribution of<br />

intestinal contents within the abdominal cavity is<br />

such that the duodenum again lies to the right of<br />

the spine with the cecum anterior to it. Adhesions<br />

between these two structures (Ladd’s bands) are<br />

often present and may result in partial or total<br />

occlusion of the second part of the duodenum. In<br />

addition the mesenteric attachment to the posterior<br />

aspect of the abdominal cavity is typically<br />

very short and there is a risk of volvulus with<br />

ensuing intestinal ischemia. Other forms of abnormal<br />

intestinal rotation (inverse rotation, malrotation<br />

with mesocolic hernia and malposition of the<br />

cecum) are all rare.<br />

Treatment<br />

There are two aspects to this disorder which<br />

require surgical intervention. The first and most<br />

important aspect is that of midgut volvulus. Any<br />

infant in whom malrotation is suspected based on<br />

clinical findings and radiological investigations<br />

should undergo laparotomy as a matter of urgency<br />

in order to minimize the risk of intestinal ischemia<br />

due to volvulus. At laparotomy blood-stained peritoneal<br />

fluid may indicate the presence of ischemic<br />

intestine. Any volvulus should be derotated<br />

(usually in the clockwise direction) and the intestine<br />

examined for viability. Non-viable bowel is<br />

resected and a primary anastomosis performed. If<br />

there is doubt about the viability of the remaining


20<br />

Congenital problems of the gastrointestinal tract<br />

intestine a second-look laparotomy can be<br />

performed after 24 h.<br />

In cases of malrotation not complicated by volvulus,<br />

the procedure of choice for most surgeons is<br />

the Ladd’s procedure. This involves division of all<br />

adhesions or adhesive bands between the cecum,<br />

duodenum and parietal peritoneum, broadening of<br />

the mesenteric base around the superior mesenteric<br />

artery and repositioning of the intestine<br />

within the abdominal cavity so that the duodenum<br />

is on the right and the cecum lies in the left upper<br />

quadrant. It has become customary to perform an<br />

appendectomy, owing to the difficulties of diagnosis,<br />

should appendicitis develop later in life.<br />

Meconium ileus<br />

Meconium ileus is a common cause of neonatal<br />

intestinal obstruction and the most common cause<br />

of antenatal intestinal perforation. 19 It should be<br />

included in the differential diagnosis of infants<br />

presenting with GI tract obstruction. In approximately<br />

80% of cases it is associated with cystic<br />

fibrosis. 20–22 The underlying defect in cystic fibrosis,<br />

an abnormality in a transmembrane chloride<br />

channel, results in the production of abnormally<br />

viscid and sticky meconium. This meconium<br />

sticks to the intestinal mucosa causing intestinal<br />

obstruction usually occurring late in gestation.<br />

Why some infants with cystic fibrosis do not<br />

develop meconium ileus is unclear. Meconium<br />

ileus can be classified as: ‘uncomplicated’, when it<br />

is limited to intraluminal obstruction caused by<br />

the abnormal meconium; or ‘complicated’, when it<br />

is associated with intestinal atresia, volvulus or<br />

meconium peritonitis.<br />

Clinical features<br />

In cases of uncomplicated meconium ileus, the<br />

infant usually presents shortly after birth with<br />

symptoms of lower gastrointestinal obstruction<br />

including abdominal distension and vomiting<br />

which may or may not be bile stained. The rectum<br />

may be empty and narrow and the infant does not<br />

pass meconium. If meconium ileus is complicated<br />

by volvulus, intestinal ischemia or perforation, the<br />

infant can be systemically unwell with acidosis,<br />

undergo hypovolemic shock and may require<br />

ventilatory support. Abdominal X-ray showing<br />

dilated intestinal loops and occasionally abundance<br />

of meconium in the right lower quadrant are<br />

supportive of the diagnosis, as is a gastrograffin<br />

contrast enema revealing a small collapsed colon<br />

(microcolon) and often inspissated pellets of<br />

meconium in the right lower quadrant.<br />

Treatment<br />

In some cases, the gastrograffin enema mentioned<br />

above may relieve the obstruction sufficiently to<br />

be curative. However, a number of uncomplicated<br />

cases and all complicated cases require surgery.<br />

The procedure performed depends on the findings<br />

during laparotomy. Atretic or grossly dilated<br />

segments of bowel may be resected, the inspissated<br />

meconium removed from the intestinal<br />

lumen and the distal bowel flushed through.<br />

Occasionally a stoma is formed to allow intestinal<br />

decompression. Outcome of surgical treatment is<br />

generally good and gastrointestinal complications<br />

are of lesser significance than the pulmonary<br />

disease caused by the underlying cystic fibrosis.<br />

Meckel’s diverticulum<br />

Meckel’s diverticulum is the most common<br />

omphalomesenteric remnant with a reported incidence<br />

of approximately 2%. Of these only a small<br />

proportion become clinically significant. The<br />

diverticulum originates from incomplete obliteration<br />

of the omphalomesenteric duct and exists as a<br />

free-lying diverticulum on the antimesenteric<br />

border of the ileum.<br />

Clinical features<br />

There are a variety of disease entities attributed to<br />

Meckel’s diverticulum including gastrointestinal<br />

hemorrhage, intussusception, diverticulitis and<br />

perforation. The most common presenting<br />

symptom is that of gastrointestinal bleeding due to<br />

excessive acid and pepsin production from an<br />

ectopic gastric mucosa which may be present<br />

within the diverticulum. Bloody diarrhea in the<br />

absence of abdominal pain is the classical presenting<br />

picture. Other complications of Meckel’s diverticulum<br />

are intussusception in which the diverticulum<br />

acts as a lead point, diverticulitis with<br />

symptoms similar to those of appendicitis and<br />

perforation.


Treatment<br />

Management of all clinically significant cases of<br />

Meckel’s diverticulum is resection of the diverticulum<br />

after adequate preoperative resuscitation. At<br />

operation the diverticulum and a wedge of ileum<br />

are resected. The ileal wedge is included, as<br />

ectopic tissue may not be entirely confined to the<br />

diverticulum. Following surgical excision the<br />

outcome is good.<br />

Congenital hepatic, pancreatic and biliary<br />

abnormalities<br />

Abnormalities of the hepaticopancreaticobiliary<br />

system are all extremely rare. They are included<br />

here as knowledge of their existence is important,<br />

as they form part of the differential diagnosis for<br />

infants with jaundice, malabsorption and hypoglycemia.<br />

The most common lesions of the biliary tree are<br />

biliary atresia and congenital biliary dilatation. In<br />

biliary atresia the biliary tree is obliterated either<br />

completely or partially. Congenital biliary dilatation<br />

includes a variety of abnormalities of the<br />

biliary tree in which the dilated segment may be<br />

either intrahepatic or extrahepatic and either<br />

fusiform or cystic in nature. Dilatations of the<br />

extrahepatic biliary ducts are commonly known as<br />

choledochal cysts. The dilated bile duct is both<br />

anatomically and functionally abnormal, resulting<br />

in cholestasis. Infants with biliary atresia and<br />

severe cholestasis associated with biliary dilatation<br />

present in the neonatal period with prolonged<br />

jaundice due to accumulation of conjugated bilirubin.<br />

When the degree of obstruction to the biliary<br />

tree is not so severe, congenital biliary dilatation<br />

may present later in life with malabsorption, intermittent<br />

jaundice, abdominal pain or even pancreatitis.<br />

In addition, a choledochal cyst may present<br />

as an upper abdominal mass. Treatment of these<br />

lesions is centered around allowing drainage of the<br />

biliary tree into the intestine, and the surgery<br />

involved is often complex. The operation of choice<br />

for biliary atresia is the Kasai portoenterostomy. 23<br />

The atretic remnants of the extrahepatic biliary<br />

ducts are removed and the porta hepatis is anastomosed<br />

to a defunctioned loop of jejunum. The<br />

timing of surgery is of paramount importance to<br />

avoid hepatocellular damage, but even with<br />

prompt diagnosis and early surgical intervention<br />

Conditions affecting the lower gastrointestinal tract 21<br />

infants with biliary atresia often have residual<br />

hepatic impairment due to intrauterine cholestasis.<br />

In cases of choledochal cysts the dilated<br />

portion of the extrahepatic ducts are removed<br />

together with the gallbladder, and the common<br />

hepatic duct is anastomosed to the duodenum or a<br />

defunctioned loop of jejunum.<br />

There are a number of congenital hepatic anomalies<br />

that give rise to structural and/or functional<br />

abnormalities of the liver parenchyma or the intrahepatic<br />

biliary tree. These include infantile and<br />

adult-type polycystic disease, congenital hepatic<br />

fibrosis, biliary hypoplasia and congenital tumors<br />

of the liver such as hamartomas and hemangiomas.<br />

Presentation is usually with one of<br />

hepatomegaly, portal hypertension or cholangitis.<br />

Treatment is that of resection of suitable lesions<br />

and prevention or treatment of hepatic disease.<br />

Congenital lesions involving the pancreas are rare,<br />

the most common being annular pancreas (see the<br />

section on the duodenum p. 00). Other anatomical<br />

anomalies are seen including pancreatic ductal<br />

anomalies, pancreatic cysts and very rarely pancreatic<br />

agenesis. There are a group of infants who<br />

present in the neonatal period with hypoglycemia<br />

who are found to have inappropriately high levels<br />

of circulating insulin. The condition hyperinsulinemic<br />

hypoglycemia (previously commonly<br />

referred to as ‘nesidioblastosis’) is characterized by<br />

inappropriate endogenous insulin secretion in the<br />

presence of low blood glucose. It may result from<br />

an insulin-secreting tumor in the pancreas (a socalled<br />

‘insulinoma’) but more commonly no tumor<br />

is identified and the disease is a result of a genetic<br />

defect in a membrane channel controlling insulin<br />

secretion. Infants require high glucose intake to<br />

maintain normoglycemia whilst they are investigated<br />

for the presence of an isolated secretory<br />

tumor. Treatment is by surgical excision of the<br />

tumor if present, otherwise a 90–95% subtotal<br />

pancreatectomy is performed.<br />

Conditions affecting the lower<br />

gastrointestinal tract<br />

Hirschsprung’s disease<br />

Hirschsprung’s disease is the most common<br />

congenital malformation of the enteric nervous


22<br />

Congenital problems of the gastrointestinal tract<br />

system with an incidence of approximately 1 in<br />

5000 live births. 24–26 Whilst most cases are<br />

sporadic a positive familial occurrence exists in<br />

3.6–7.8% of cases 27 and the presence of co-existing<br />

abnormalities including trisomy 21 suggests a<br />

genetic involvement (see also Chapter 17).<br />

This condition is characterized by the absence of<br />

enteric neurons and hypertrophy of nerve trunks<br />

in the distal bowel always involving the rectum<br />

and for a variable distance proximally. There is an<br />

absence of peristaltic activity in this aganglionic<br />

segment resulting in symptoms of intractable<br />

constipation. The bowel proximal to the aganglionic<br />

segment contains ganglionated cells, and<br />

peristaltic activity is normal. However, in the socalled<br />

transitional zone, immediately proximal to<br />

the aganglionic segment, neuronal cells may exist<br />

but they are commonly of abnormal architecture<br />

and the intestinal peristalsis is abnormal. This<br />

zone is of fundamental importance when considering<br />

surgical treatment of this disorder.<br />

Clinical features<br />

In the neonatal period the disease should be<br />

considered in any infant who fails to pass<br />

meconium in the first 48h of life. The usual<br />

presentation in the neonatal period is with constipation,<br />

abdominal distension and eventually<br />

vomiting during the first few days of life. More<br />

severe symptoms may be present in the neonatal<br />

period including those of gastrointestinal obstruction,<br />

enterocolitis (see below) and rarely perforation.<br />

Later in infancy symptoms of intractable<br />

constipation may signify the presence of<br />

Hirschsprung’s disease.<br />

Diagnosis<br />

One of the most important factors in the management<br />

of Hirschsprung’s disease is early diagnosis<br />

and appropriate treatment to avoid complications<br />

of the disease. Following clinical suspicion a<br />

number of investigations may be of use in making<br />

a diagnosis which must always be confirmed by<br />

histological examination of intestinal tissue. Plain<br />

abdominal X-ray will often show dilated proximal<br />

intestinal loops (Figure 2.6) prompting a lower GI<br />

contrast study to be performed to exclude intestinal<br />

atresia/stenosis or meconium ileus. This may<br />

show a prompt transition from narrow distal<br />

aganglionic bowel to dilated proximal bowel. A<br />

contrast enema is not necessary for diagnosis in<br />

many cases of Hirschsprung’s disease. In addition,<br />

this investigation may be misleading in its indication<br />

of the length of intestinal aganglionosis.<br />

The gold standard for diagnosis is the suction<br />

rectal biopsy. Characteristic histological findings<br />

are absence of ganglion cells and increase in<br />

acetylcholinesterase (AChE) staining in the<br />

parasympathetic nerve fibers. In cases where<br />

suction rectal biopsy fails to provide adequate<br />

information, a full-thickness rectal biopsy should<br />

be considered.<br />

Treatment<br />

Treatment in the first instance is aimed at decompression<br />

of the distal GI tract by regular rectal<br />

washouts. Subsequently a number of surgical<br />

options exist, all of which aim to remove the aganglionic<br />

segment and restore intestinal continuity by<br />

means of an anastomosis of ganglionated bowel to<br />

the rectal stump (so-called ‘pull-through’). This may<br />

be performed either as a primary procedure or as a<br />

delayed procedure after initial colostomy formation<br />

Figure 2.6 Plain abdominal X-ray of a child with<br />

rectosigmoid Hirschsprung’s disease showing extensively<br />

dilated loops of bowel.


to allow distal intestinal decompression. 28 There are<br />

a number of surgical techniques, of which three are<br />

the most commonly used – the Swenson rectosigmoidectomy,<br />

the Duhammel retrorectal transanal<br />

pull-through and the endorectal pull-through of<br />

Soave. These techniques may also be carried out<br />

laparoscopically in suitable infants.<br />

Of paramount importance in all surgical approaches<br />

is removal of the full length of aganglionic intestine.<br />

Failure to do so may result in recurrence of symptoms<br />

and complications of Hirschsprung’s disease.<br />

To ensure normoganglionosis of the segment,<br />

pulled-through intraoperative biopsies are taken<br />

and rapidly examined histologically. Biopsies may<br />

be taken laparoscopically in infants undergoing a<br />

minimally invasive procedure. A number of authors<br />

have advocated laparoscopic colonic mapping prior<br />

to or during definitive surgery for exact characterization<br />

of the extent of the disease. 29,30 Recently<br />

there has been interest in infants who have retained<br />

a portion of the transition zone and have recurrent<br />

symptoms. 31,32 Attention has been drawn to the fact<br />

that the transition zone may not be linear around<br />

the circumference of the intestine, and it has been<br />

recommended that multiple circumferential biopsies<br />

be taken. 31,33<br />

Hirschsprung’s enterocolitis<br />

Despite appropriate treatment this serious complication<br />

of Hirschsprung’s disease can develop at<br />

any stage and may be the presenting condition.<br />

Profuse, often bloody diarrhea with abdominal<br />

distension is the main presenting symptom.<br />

Vomiting and fever may be present and the child<br />

may become rapidly unwell and dehydrated.<br />

Treatment is initially with fluid resuscitation and<br />

intestinal decompression by means of a nasogastric<br />

tube. Rectal examination should be performed<br />

as it may produce an explosion of foul-smelling<br />

gas and stools which aids diagnosis and intestinal<br />

decompression. The bowel is rested, during which<br />

time parenteral nutrition may be considered. Our<br />

practice is to give enteral vancomycin, targeting<br />

Clostridium species which are often implicated.<br />

Outcome<br />

The results following surgery for Hirschsprung’s<br />

disease are generally good. Long-term outcome<br />

may be difficult to assess fully, owing to the short<br />

Conditions affecting the lower gastrointestinal tract 23<br />

follow-up periods reported in the literature. The<br />

incidence of incontinence is reported as ranging<br />

from zero to 82% and that of constipation from<br />

zero to 56%, these two complications probably<br />

having the greatest influence on quality of life. 34–36<br />

Whilst some patients may be relieved of their symptoms<br />

by surgery altogether, it should be remembered<br />

that, for some, lifelong symptoms persist particularly<br />

if the entire colon is involved by the disease. 34,36<br />

Anorectal anomalies<br />

Congenital abnormalities of the anorectal region<br />

occur with an incidence of 1 in 4000 to 1 in 5000<br />

live births. 37–39 A very small minority may be familial<br />

with the majority being isolated findings or part<br />

of a congenital syndrome such as the VACTERL<br />

syndrome. There are a number of different types of<br />

anorectal anomalies resulting from the complex<br />

embryological development of the anorectal region<br />

involving differentiation of the cloaca. The<br />

Wingspread classification 40 divides them into high,<br />

intermediate or low based on the relationship of<br />

the terminal bowel or any fistula arising from the<br />

bowel to the pelvic diaphragm. Precise definition of<br />

the abnormal anatomy is of paramount importance<br />

when planning corrective surgical treatment.<br />

Clinical features<br />

Abnormalities of the anorectal region are usually<br />

diagnosed on inspection during the newborn period,<br />

but surprisingly this is not always the case. In many<br />

cases the anus will be absent and meconium may be<br />

seen to originate from an abnormal site including a<br />

mucocutaneous fistula, the urethra in males or the<br />

vagina in females. Anomalies in which the anus is<br />

present but abnormally sited or stenosed may be<br />

more difficult to diagnose in the absence of adequate<br />

experience. The most complex abnormality in<br />

females is shown by the cloaca, represented by a<br />

single opening in the perineum with rectum, vagina<br />

and urethra joining a single channel.<br />

Treatment<br />

Treatment is aimed at preventing complications<br />

associated with the anomaly, including urinary<br />

tract infection and lower GI obstruction and subsequently<br />

restoring the anatomy to as near to normal


24<br />

Congenital problems of the gastrointestinal tract<br />

a functional and cosmetic state as possible. In the<br />

majority of cases the initial surgery involves<br />

forming a colostomy in the descending or sigmoid<br />

colon to allow intestinal drainage and avoid dilatation<br />

of the lower bowel. 41 Following assessment,<br />

planning of surgery and growth of the infant reconstructive<br />

surgery is undertaken most commonly by<br />

the posterior sagittal approach. 42 Some anomalies<br />

also require a laparotomy to divide a high<br />

rectovesical fistula. Surgery of these cases and<br />

particularly of the cloaca is complex and should be<br />

performed by an experienced surgeon.<br />

Outcome<br />

In similarity to patients with Hirschsprung’s<br />

disease, incontinence and constipation are the<br />

most significant long-term complications of<br />

surgically treated anorectal anomalies and often<br />

have a significant impact on quality of life. In one<br />

large series, soiling occurred in 57% of 387 cases.<br />

The incidence of fecal incontinence was 25% and<br />

constipation 43.1%. 42 Ongoing medical and, on<br />

occasion, surgical treatment is necessary to minimize<br />

disruption to a normal lifestyle.<br />

Conditions which may occur at any<br />

point in the gastrointestinal tract<br />

Gastrointestinal duplications<br />

Duplication cysts of the GI tract are rare congenital<br />

abnormalities. They can occur at any point in<br />

the GI tract from mouth to anus, although they are<br />

most commonly found around the ileocecal region.<br />

Duplication cysts are defined according to strict<br />

criteria, as devised by Ladd and Gross; they are<br />

closely attached to some part of the GI tract, have<br />

a smooth muscle coat and have an epithelial lining<br />

that resembles some part of the alimentary canal. 43<br />

Duplications may be spherical or tubular in macroscopic<br />

appearance, those that are tubular accounting<br />

for 10–20% and often having a communication<br />

with the bowel.<br />

Clinical features<br />

Between 25 and 30% present in the neonatal<br />

period and most have presented by the age of 10<br />

years. Clinical features at presentation depend on<br />

anatomical site, size and secondary effects and<br />

include an oropharyngeal, abdominal or rectal<br />

mass, respiratory distress, GI bleeding, obstruction<br />

and intussusception. Duplication cysts may also<br />

be found as incidental findings at laparotomy and<br />

some lesions have been detected on antenatal<br />

ultrasound examination. 44,45<br />

Treatment<br />

The recommended management of duplication<br />

cysts is complete surgical excision wherever possible,<br />

in order to prevent recurrence and complications<br />

secondary to ectopic gastric mucosa. When<br />

complete excision is not possible it is essential to<br />

remove the mucosal lining.<br />

Conditions affecting the walls of the<br />

abdominal cavity<br />

Whilst not truly conditions of the GI tract, there<br />

are a number of conditions that cause the abdominal<br />

contents to develop outside the abdominal<br />

cavity. These conditions are included as they have<br />

secondary effects which may significantly affect<br />

the GI tract and be a cause of GI dysfunction.<br />

Congenital diaphragmatic hernia<br />

The incidence of congenital diaphragmatic hernia<br />

varies from 1 in 3500 to 1 in 5000 live births. 46 Its<br />

etiology is unknown, although it is probably multifactorial.<br />

The essential anatomical defect is a<br />

breach in the continuity of the diaphragm which<br />

allows herniation of the abdominal viscera into the<br />

thoracic cavity. This has a secondary effect of<br />

impeding development of the lungs during<br />

intrauterine life. The resulting hypoplastic lungs<br />

are a cause of significant morbidity and mortality<br />

in this condition. Compression by misplaced<br />

abdominal contents does not explain the severity<br />

of lung disease seen and it is well recognized that<br />

lung development is markedly abnormal in infants<br />

with congenital diaphragmatic hernia.<br />

Classification<br />

A number of different defects can occur, owing to<br />

the complex development of the diaphragm. The


majority of cases are of the Bochdalek type, in<br />

which the defect is posterolateral and most<br />

commonly on the left side. The defect can range in<br />

size from a small slit to involve almost the entire<br />

hemidiaphragm. Defects in the central tendon of<br />

the diaphragm result in Morgagni hernias, which<br />

are retrosternal in nature and most commonly on<br />

the right side. Finally, agenesis of the diaphragm<br />

may occur which is usually left sided and<br />

extremely rare.<br />

Clinical features<br />

In the current era, diaphragmatic hernia is often<br />

diagnosed during prenatal ultrasound scanning.<br />

The advantage of prenatal diagnosis is that delivery<br />

can be planned to take place in a unit with<br />

appropriate pediatric surgical and intensive care<br />

facilities. For those infants who avoid prenatal<br />

diagnosis, for whatever reason, the clinical<br />

features depend on the volume of abdominal<br />

contents within the thoracic cavity and the degree<br />

of lung hypoplasia. In the most severe cases, there<br />

will be severe respiratory distress and cyanosis<br />

from shortly after birth. At the other end of the<br />

spectrum are infants who have minimal if any<br />

respiratory symptoms or signs and in whom<br />

intestinal loops are noted to be in the abdomen on<br />

chest X-ray (Figure 2.7).<br />

Treatment<br />

Whilst the definitive treatment of diaphragmatic<br />

hernia is surgical closure, the timing of this is not<br />

of paramount importance and should be undertaken<br />

once the infant is stable from a cardiovascular<br />

and respiratory point of view. The respiratory<br />

management of these infants can be problematic<br />

owing to the severe lung hypoplasia and associated<br />

pulmonary hypertension. Most require<br />

conventional ventilatory support as a minimum<br />

and many require high-frequency oscillatory<br />

ventilation to ensure adequate oxygenation. Other<br />

measures to reduce pulmonary hypertension,<br />

including inhaled nitric oxide, adenosine or sildenafil,<br />

may be effective. Resistant cases may be<br />

candidates for extracorporeal membrane oxygenation,<br />

during which the infant is placed on a<br />

life-support system in the hope that the lungs and<br />

in particular the pulmonary vasculature will<br />

mature. Various criteria for extracorporeal<br />

membrane oxygenation exist47 with the aim of<br />

Conditions affecting the walls of the abdominal cavity 25<br />

Figure 2.7 Chest radiograph of an infant with congenital<br />

diaphragmatic hernia. Loops of intestine are clearly seen<br />

within the left hemithorax and there is mediastinal shift to<br />

the right.<br />

reserving it for those who have the most severe<br />

respiratory failure and those who are most likely to<br />

benefit. Unfortunately, there remain a number of<br />

infants with congenital diaphragmatic hernia<br />

whose lung disease presents too great a challenge<br />

and these do not survive.<br />

Surgery<br />

The principles of surgical repair are to return the<br />

abdominal contents to the abdominal cavity and<br />

repair the diaphragmatic defect. It may be possible<br />

to repair the defect by simply suturing the edges<br />

together. However, if the defect is large a patch<br />

repair may be undertaken using prosthetic material.<br />

The long-term outcome of congenital diaphragmatic<br />

hernia is dependent primarily on the degree<br />

of pulmonary hypoplasia. The main GI consequence<br />

appears to be gastroesophageal reflux, seen<br />

in up to 62% of cases. 48<br />

Anterior abdominal wall defects<br />

Although separate clinical entities, the two conditions<br />

exomphalos and gastroschisis, which comprise<br />

anterior abdominal wall defects are grouped<br />

together, owing to similarity in their clinical appearance<br />

and the recommended course of management.<br />

In both conditions some portion of the viscera lies<br />

outside the abdominal cavity extruding through a<br />

defect in the anterior abdominal wall.


26<br />

Congenital problems of the gastrointestinal tract<br />

Exomphalos<br />

The defect on the anterior abdominal wall in cases<br />

of exomphalos lies in the midline. Viscera herniate<br />

through this defect but remain contained within<br />

an avascular hernial sac comprising peritoneum<br />

and amniotic membrane (Figure 2.8). The size of<br />

the defect and hence the size of the sac may vary<br />

from a small swelling at the base of the umbilical<br />

cord (exomphalos minor) to a much larger sac<br />

containing liver and a large proportion of the small<br />

intestine (exomphalos major). The embryological<br />

origins of exomphalos are believed to be failure of<br />

complete closure of the anterior abdominal wall<br />

around a persistent body stalk. Visceral contents<br />

continue to develop within this body stalk and<br />

thus remain outside the abdominal cavity. Whilst<br />

the precise etiology of exomphalos is not clear, it is<br />

well recognized that exomphalos often co-exists<br />

with a number of other congenital abnormalities<br />

and this may suggest at least in part a genetic<br />

component. Associated abnormalities include<br />

Beckwith–Wiedemann syndrome, the trisomies 13,<br />

18 and 21 and the upper and lower midline<br />

associations.<br />

Gastroschisis<br />

The anterior abdominal wall defect in cases of<br />

gastroschisis is of full thickness and typically to<br />

the right of the umbilical cord. Unlike exomphalos<br />

there is no sac covering the eviscerated intestine,<br />

which is usually dilated and inflamed (Figure 2.9).<br />

Figure 2.8 Clinical appearance of an infant with exomphalos.<br />

The abdominal contents are enclosed within an<br />

avascular hernial sac.<br />

The liver is not herniated. The precise embryological<br />

basis of gastroschisis is unclear and a number<br />

of hypotheses have been proposed. The fact that<br />

gastroschisis is rarely associated with any other<br />

congenital abnormalities, with the exception of<br />

intestinal atresias and malrotation, suggests that it<br />

is most likely to have a separate embryological<br />

basis from the events resulting in exomphalos.<br />

Treatment<br />

It is now common for these two abnormalities to be<br />

detected in the antenatal period; delivery in a<br />

specialist center with pediatric surgical facilities is<br />

recommended. There is no consensus concerning<br />

the timing or mode of delivery of these babies and<br />

there is no convincing evidence to suggest that<br />

preterm or Cesarean section delivery confer any<br />

distinct advantage. 49–51 In gastroschisis, however,<br />

delivery is commonly induced at 37 weeks’ gestation<br />

to avoid late-gestation fetal death. What is of<br />

paramount importance is protection of the intestine<br />

and prevention of fluid loss in cases of gastroschisis<br />

from the moment of delivery. In exomphalos the<br />

hernial sac confers a degree of protection to the<br />

intestine. In cases of gastroschisis the eviscerated<br />

intestine should be wrapped in clingfilm and<br />

adequate support provided to prevent fluid loss and<br />

ischemic damage to the bowel. Cases of exomphalos<br />

in which the hernial sac ruptures during delivery<br />

should subsequently be treated as for gastroschisis.<br />

Surgery and attempted closure should take place as<br />

Figure 2.9 Clinical appearance of an infant with<br />

gastroschisis. There is no sac enclosing the herniated<br />

intestine, which is thickened and inflamed.


soon as possible following stabilization of the infant<br />

to prevent dehydration. Recent evidence suggests<br />

that a staged repair may result in favorable outcome<br />

when compared with primary closure. 52–54<br />

Intestinal dysmotility is always present in neonates<br />

with gastroschisis requiring parenteral nutrition for<br />

a period of usually 2–3 weeks. Parenteral nutrition<br />

has significantly improved the survival of neonates<br />

with gastroschisis.<br />

Surgical closure<br />

The aim of surgery in both conditions is the return<br />

of abdominal contents to the abdominal cavity and<br />

closure of the overlying skin. In some cases this<br />

REFERENCES<br />

1. Jensen BL, Kreiborg S, Dahl E et al, Cleft lip and palate<br />

in Denmark, 1976–1981: epidemiology, variability, and<br />

early somatic development. Cleft Palate J 1988; 25:<br />

258–269.<br />

2. Womersley J, Stone DH. Epidemiology of facial clefts.<br />

Arch Dis Child 1987 62: 717–720.<br />

3. Shprintzen RJ, Siegel-Sadewitz VL, Amato J et al.<br />

Anomalies associated with cleft lip, cleft palate, or both.<br />

Am J Med Genet 1985; 20: 585–595.<br />

4. Kyyronen P, Himminki K. Gastro-intestinal atresia in<br />

Finland in 1970–79, indicating time–place clustering. J<br />

Epidemiol Community Health 1988; 42: 257–265.<br />

5. Myers NA. Esophageal atresia: the epitome of modern<br />

surgery. Ann R Coll Surg Engl 1974; 54: 277–287.<br />

6. Spitz L, Kiely EM, Morecroft JA et al. Esophageal<br />

atresia: at-risk groups for the 1990s. J Pediatr Surg 1994;<br />

29: 723–725.<br />

7. Pierro A, Cozzi F, Colarossi G et al. Does fetal gut<br />

obstruction cause hydramnios and growth retardation? J<br />

Pediatr Surg 1987; 22: 454–457.<br />

8. Sweed Y. Duodenal obstruction. In Puri P, ed. Newborn<br />

Surgery, 2nd edn. New York: Arnold, 2003: 423–433.<br />

9. Menardi G. Duodenal atresia, stenosis and annular<br />

pancreas. In Freeman NV, Burge DM, Griffiths DM,<br />

References 27<br />

can be achieved in one surgical procedure<br />

(primary closure) but in many instances the<br />

abdominal cavity is not of sufficient volume to<br />

accommodate the eviscerated organs. In these<br />

cases a staged closure is performed in which a<br />

‘silo’ is attached under the fascia around the base<br />

of the defect and completely encloses the eviscerated<br />

abdominal contents. This gives the intestine<br />

protection from dehydration and contains the<br />

bowel within a manageable sac, reducing the risk<br />

of intestinal damage. The silo is gradually reduced<br />

in size as the abdominal cavity allows and the skin<br />

is closed in a final operation. The prognosis for<br />

infants with both these conditions in the absence<br />

of co-existing abnormalities is good.<br />

Malone PSJ, eds. Surgery of the Newborn, 1st edn.<br />

Edinburgh: Churchill Livingstone, 1994: 107–115.<br />

10. Stauffer UG, Irving I. Duodenal atresia and stenosis –<br />

long-term results. Prog Pediatr Surg 1977; 10: 49–60.<br />

11. Rode H, Millar AJW. Jejuno-ileal atresia and stenosis. In<br />

Puri P, ed. Newborn Surgery, 2nd edn. New York:<br />

Arnold, 2003: 445–456.<br />

12. Louw JH, Barnard CN. Congenital intestinal atresia:<br />

observations on its origin. Lancet 1955; 2:1065.<br />

13. Louw JH. Congenital intestinal atresia and stenosis in<br />

the newborn. Observations on its pathogenesis and<br />

treatment. Ann R Coll Surg Engl 1959; 25: 209.<br />

14. Grosfeld JL, Ballantine TV, Shoemaker R. Operative<br />

mangement of intestinal atresia and stenosis based on<br />

pathologic findings J Pediatr Surg 1979; 14: 368–375.<br />

15. Rickham PP. Massive small intestinal resection in<br />

newborn infants. Hunterian Lecture delivered at the<br />

Royal College of Surgeons of England on 13th April<br />

1967. Ann R Coll Surg Engl 1967; 41: 480–492.<br />

16. Touloukian RJ, Smith GJ. Normal intestinal length in<br />

preterm infants. J Pediatr Surg 1983; 18: 720–723.<br />

17. Hollwarth ME. Short bowel syndrome and surgical techniques<br />

for the baby with short intestines. In Puri P, ed. Newborn<br />

Surgery, 2nd edn. New York: Arnold, 2003; 569–576.


28<br />

Congenital problems of the gastrointestinal tract<br />

18. Kluth D, Kaestner M, Tibboel D et al. Rotation of the<br />

gut: fact or fantasy? J Pediatr Surg 1995; 30: 448–453.<br />

19. Farber SJ. The relation of pancreatic achylia to meconium<br />

ileus. J Pediatr 1944; 24: 387–392.<br />

20. Del Pin CA, Czyrko C, Ziegler MM et al. Management<br />

and survival of meconium ileus. A 30-year review. Ann<br />

Surg 1992; 215: 179–185.<br />

21. Fakhoury K, Durie PR, Levison H et al. Meconium ileus<br />

in the absence of cystic fibrosis. Arch Dis Child 1992;<br />

67: 1204–1206.<br />

22. Murshed R, Spitz L, Kiely E et al. Meconium ileus: a<br />

ten-year review of thirty-six patients. Eur J Pediatr Surg<br />

1997; 7: 275–277.<br />

23. Kasai M. Treatment of biliary atresia with special reference<br />

to hepatic porto-enterostomy and its modifications.<br />

Prog Pediatr Surg 1974; 6: 5–52.<br />

24. Passarge E. The genetics of Hirschsprung’s disease.<br />

Evidence for heterogeneous etiologogy and a study of<br />

sixty-three families. N Engl J Med 1967; 276: 138–143.<br />

25. Orr JD, Scobie WG. Presentation and incidence of<br />

Hirschsprung’s disease. Br Med J (Clin Res Ed) 1983:<br />

287: 1671.<br />

26. Spouge D, Baird PA. Hirschsprung’s disease in large<br />

birth cohort. Teratology 1985; 32: 171–177.<br />

27. Puri P. Hirschsprung’s disease. In Oldham TO,<br />

Colombani PM, Foglia RP, eds. Surgery of Infants and<br />

Children: Scientific Principles and Practice. New York:<br />

Lippincott-Raven, 1997: 1277–1299.<br />

28. Pierro A, Fasoli L, Kiely EM et al. Staged pull-through<br />

for rectosigmoid Hirschsprung’s disease is not safer<br />

than primary pull-through. J Pediatr Surg 1997; 32:<br />

505–509.<br />

29. Yamataka A, Yoshida R, Kobayashi H et al. Laparoscopyassisted<br />

suction colonic biopsy and intraoperative rapid<br />

acetylcholinesterase staining during transanal pullthrough<br />

for Hirschsprung’s disease. J Pediatr Surg 2002;<br />

37: 1661–1663.<br />

30. Carvalho JL, Campos M, Soares-Oliveira M et al.<br />

Laparoscopic colonic mapping of dysganglionosis.<br />

Pediatr Surg Int 2001; 17: 493–495.<br />

31. Ghose SI, Squire BR, Stringer MD et al. Hirschsprung’s<br />

disease: problems with transition-zone pull-through. J<br />

Pediatr Surg 2000; 35: 1805–1809.<br />

32. Proctor ML, Traubici J, Langer JC et al. Correlation<br />

between radiographic transition zone and level of aganglionosis<br />

in Hirschsprung’s disease: implications for<br />

surgical approach. J Pediatr Surg 2003; 38: 775–778.<br />

33. Farrugia M, Alexander N, Nicholls E et al. Does transitional<br />

zone pull-through in Hirschsprung’s disease<br />

imply a poor prognosis? Presented at the Pacific<br />

Association of Pediatric Surgeons 36th Annual Meeting,<br />

Sydney, Australia 2003.<br />

34. Ludman L, Spitz L, Tsuji H et al. Hirschsprung’s<br />

disease: functional and psychological follow up comparing<br />

total colonic and rectosigmoid aganglionosis. Arch<br />

Dis Child 2002; 86: 348–351.<br />

35. Teitelbaum DH, Coran AG. Long-term results and<br />

quality of life after treatment of Hirschsprung’s disease<br />

and allied disorders. In Holschneider AM, Puri P, eds.<br />

Hirschsprung’s Disease and Allied Disorders, 2nd edn.<br />

Amsterdam: Harwood Academic, 2000: 457–465.<br />

36. Tsuji H, Spitz L, Kiely EM et al. Management and longterm<br />

follow-up of infants with total colonic aganglionosis.<br />

J Pediatr Surg 1999; 34: 158–161.<br />

37. Brenner EC. Congenital defects of the anus and rectum.<br />

Surg Gynecol Obstet 1915; 20: 579–588.<br />

38. Santulli TV. Treatment of imperforate anus and associated<br />

fistulas. Surg Gynecol Obstet 1952; 95: 601–614.<br />

39. Trusler GA, Wilkinson RH. Imperforate anus: a review<br />

of 147 cases. Can J Surg 1962; 5: 169–177.<br />

40. Stephens FD, Smith ED. Classification, identification<br />

and assessment of surgical treatment of anorectal anomalies.<br />

Pediatr Surg Int 1986: 1:200–205.<br />

41. Patwardhan N, Kiely EM, Drake DP et al. Colostomy for<br />

anorectal anomalies: high incidence of complications. J<br />

Pediatr Surg 2001; 36: 795–798.<br />

42. Pena A. Anorectal anomalies. In Puri P, ed. Newborn<br />

Surgery, 2nd edn. New York: Arnold, 2003: 535–552.<br />

43. Ladd WE, Gross RE. Surgical treatment of duplication of<br />

the alimentary tract; enterogenous cysts, enteric cysts,<br />

or ileum duplex. Surg Gynecol Obstet 1940; 70: 295–307.<br />

44. Duncan BW, Adzick NS, Eraklis A. Retroperitoneal<br />

alimentary tract duplications detected in utero. J Pediatr<br />

Surg 1992; 27: 1231–1233.<br />

45. Goyert GL, Blitz D, Gibson P et al. Prenatal diagnosis of<br />

duplication cyst of the pylorus. Prenat Diagn 1991; 11:<br />

483–486.<br />

46. Robert E, Kallen B, Harris J. The epidemiology of<br />

diaphragmatic hernia. Eur J Epidemiol 1997; 13:<br />

665–673.<br />

47. UK Collaborative ECMO Trial Group. UK collaborative<br />

randomised trial of neonatal extracorporeal membrane<br />

oxygenation. Lancet 1996; 348: 75–82.<br />

48. Kieffer J, Sapin E, Berg A et al. Gastroesophageal reflux<br />

after repair of congenital diaphragmatic hernia. J Pediatr<br />

Surg 1995; 30: 1330–1333.<br />

49. Quirk JG Jr, Fortney J, Collins HB et al. Outcomes of<br />

newborns with gastroschisis: the effects of mode of<br />

delivery, site of delivery, and interval from birth to<br />

surgery. Am J Obstet Gynecol 1996; 174: 1134–1138.<br />

50. Dunn JC, Fonkalsrud EW, Atkinson JB. The influence of<br />

gestational age and mode of delivery on infants with<br />

gastroschisis. J Pediatr Surg 1999; 34: 1393–1395.<br />

51. Sheth NP. Preterm and particularly, pre-labour cesarean<br />

section to avoid complications of gastroschisis. Pediatr<br />

Surg Int 2000; 16: 229.<br />

52. Jona JZ. The ‘gentle touch’ technique in the treatment of<br />

gastroschisis. J Pediatr Surg 2003; 38: 1036–1038.<br />

53. Kidd JN Jr, Jackson RJ, Smith SD et al. Evolution of<br />

staged versus primary closure of gastroschisis. Ann Surg<br />

2003; 237: 759–764.<br />

54. Schlatter M, Norris K, Uitvlugt N et al. Improved<br />

outcomes in the treatment of gastroschisis using a<br />

preformed silo and delayed repair approach. J Pediatr<br />

Surg 2003; 38: 459–464.


3<br />

Infectious esophagitis<br />

Salvatore Cucchiara and Osvaldo Borrelli<br />

Epidemiology and predisposing<br />

conditions<br />

In the recent years the spectrum of infectious<br />

esophagitis in childhood has expanded, owing to<br />

the emergence of new conditions, such as the<br />

acquired immunodeficiency syndrome (AIDS), the<br />

advancement in therapy and survival of patients<br />

transplanted and treated with immunosuppressive<br />

drugs, and the improvement in endoscopic and<br />

microbiological techniques. The most common<br />

infectious causes of esophagitis are fungal, viral,<br />

bacterial and, more rarely, protozoal. Primary<br />

esophageal infection is quite rare in otherwise<br />

normal subjects without permissive factors and<br />

most cases of infectious esophagitis are described<br />

in immunocompromised patients.<br />

The immunocompetent subjects developing<br />

esophageal infections will have predisposing<br />

conditions that weaken defence mechanisms of<br />

the esophageal mucosa. Esophageal disorders that<br />

cause slowing of peristalsis and stasis of intraluminal<br />

content such as achalasia, systemic sclerosis,<br />

myopathies, neuropathies and esophageal<br />

strictures predispose to infections of the esophageal<br />

mucosa, usually candidiasis. Infections in<br />

the surrounding organs and structures may also<br />

involve the esophagus. Other conditions predisposing<br />

to esophageal infections are malnutrition<br />

and diabetes mellitus. The latter may derange<br />

esophageal peristalsis and emptying of intraluminal<br />

content, and may impair granulocyte function<br />

through hyperglycemia. Finally, antimicrobial<br />

drugs can alter the normal oropharyngeal flora<br />

leading to overgrowth of Candida organisms.<br />

Conditions affecting both humoral and cellular<br />

immunological variables also lead to esophageal<br />

infections. This occurs in children with either<br />

genetic or acquired immunodeficiency as well as<br />

in the course of diseases promoting development<br />

of opportunistic infections. Transplant recipients<br />

are candidates for infectious esophagitis through<br />

different mechanisms: drug-induced immunosuppression,<br />

chemotherapy and neutropenia. Furthermore,<br />

whereas bacterial and fungal infections<br />

predominate in the early phases following transplantation,<br />

when granulocyte number and function<br />

are more compromised, the cytomegalovirus<br />

(CMV) infection occurs a few months after<br />

transplantation when T-cell function is more<br />

impaired.<br />

In HIV-infected patients opportunistic infections<br />

develop and increase in frequency as immunodeficiency<br />

worsens. Esophageal and other opportunistic<br />

infections do not occur until immunodeficiency<br />

is severe, usually when the CD4<br />

lymphocyte count is below 100–200/mm 3 , according<br />

to references 1–3. More recently, however, the<br />

widespread availability of highly active antiretroviral<br />

therapy has been associated with an apparent<br />

decline in opportunistic infections in HIV-infected<br />

patients. The causes of esophageal disease in<br />

patients with HIV infection and AIDS are reported<br />

in Table 3.1. In contrast to the findings in other<br />

immunocompromised hosts, herpes simplex virus<br />

(HSV) esophagitis is uncommon, whereas by far<br />

the most common cause is Candida, accounting<br />

for about 50% of esophageal infections. 4,5<br />

Esophageal candidiasis can also co-exist with<br />

other esophageal infections. 6<br />

Fungal infections<br />

Candida species are the most common agents<br />

of infectious esophagitis. Candida albicans is<br />

the most common pathogen, but C. tropicalis,<br />

C. parapsilosis and C. glabrata have occasionally<br />

been reported. These organisms are usually<br />

29


30<br />

Infectious esophagitis<br />

Table 3.1 Causes of esophageal disease in HIV infection and AIDS<br />

Common Uncommon Rare<br />

Candida Herpes simplex virus Neoplasm<br />

Cytomegalovirus Gastroesophageal reflux disease Mycobacteria<br />

Idiopathic Protozoa<br />

present in the normal oral flora, where their<br />

growth is controlled by commensal organisms.<br />

Conditions predisposing to esophageal candidiasis<br />

in immunocompetent subjects are inhaled or<br />

ingested corticosteroids, prolonged antibiotic<br />

administration, acid suppressive therapy, disorders<br />

of esophageal motility, malnutrition, diabetes<br />

mellitus and neck or head radiotherapy because of<br />

malignancy. All abnormalities in cellular immunity<br />

lead to esophageal candidiasis, whereas<br />

improved management of immunosuppressive<br />

therapies and antifungal prophylaxis have<br />

reduced the occurrence of esophageal candidiasis<br />

in solid-organ transplant recipients. 7<br />

Esophageal candidiasis has the classical appearance<br />

of white or yellow plaques coating the<br />

esophageal mucosa (Figure 3.1). These plaques can<br />

extend up to the proximal esophagus, are usually<br />

thick and, characteristically, cannot be washed or<br />

brushed off, unlike food or milk residues overlying<br />

esophageal mucosa. They include desquamated<br />

esophageal epithelial cells intermingled with<br />

inflammatory cells, bacteria and mycelia and<br />

spores typical for Candida. 8 The underlying squamous<br />

esophageal epithelium usually appears<br />

uninvolved, and ulcerations occur rarely.<br />

Typical symptoms of esophageal candidiasis are<br />

painful swallowing (odynophagia) or dysphagia<br />

(difficulty in swallowing, described as food ‘sticking’)<br />

(Table 3.2). 3,9 Any patient with risk factors for<br />

esophageal infection and complaining of dysphagia<br />

should be suspected for esophageal<br />

candidiasis. The latter, however, can be detected<br />

by chance in asymptomatic subjects. When evaluating<br />

patients with esophageal complaints, an<br />

important part of the physical examination is a<br />

close inspection of the oropharynx. However, oral<br />

candidiasis is not predictive of esophageal involvement<br />

and the latter can occur in the absence of oral<br />

candidiasis even in the immunocompromised<br />

Figure 3.1 Esophageal candidiasis. White plaques<br />

coating the lower third of the esophageal mucosa.<br />

subject. Complications from esophageal candidiasis<br />

occur rarely. Hematemesis suggests underlying<br />

ulcerative esophagitis that occurs if the disease is<br />

severe and there is an associated coagulopathy.<br />

Given that esophageal candidiasis is the most<br />

common of opportunistic infections of the esophagus<br />

in subjects with a predisposing condition (e.g.<br />

HIV infection, transplantation, immunosuppressive<br />

therapy), an empirical antifungal therapy has been<br />

proposed, with further diagnostic evaluation based<br />

on the clinical response (Figure 3.2). Interestingly, a<br />

prospective study comparing empirical fluconazole<br />

to endoscopy in HIV-infected adults has shown<br />

empirical fluconazole to be the best initial management<br />

strategy. 3 Candida esophagitis usually<br />

responds rapidly to fluconazole. 10<br />

Before the advent of upper endoscopy, a barium<br />

esophagram was used as the initial diagnostic tool.<br />

However, a number of studies have shown the rela-


Table 3.2 Symptoms and clinical signs in patients with esophageal infection<br />

Fungal infections 31<br />

Candida Viruses Bacteria Parasites<br />

Dysphagia ++++ + + +<br />

Odynophagia ++ ++++ +++ ++<br />

Heartburn + + + +<br />

Chest pain + +++ ++ +<br />

Fever - + +++ -<br />

Hematemesis + ++ + +<br />

(-), Not occurring; (+), rare; (++), occasional; (+++), common; (++++), always present<br />

Immunocompetent subject Immunocompromised subject<br />

Upper gastrointestinal<br />

endoscopy<br />

Biopsies<br />

Pathogen identified and<br />

specific treatment given<br />

Dysphagia or<br />

odynophagia, chest pain<br />

Upper gastrointestinal<br />

endoscopy and biopsy<br />

Pathogen identified and<br />

specific treatment given<br />

Upper gastrointestinal<br />

endoscopy and biopsy<br />

Figure 3.2 Algorithm for management of infectious esophagitis.<br />

tive insensitivity and non-specificity of barium<br />

studies for evaluating esophageal mucosa infection.<br />

Commonly reported X-ray features in patients with<br />

esophageal candidiasis are plaque-like lesions in a<br />

linear configuration that, when severe, become<br />

systemic<br />

symptoms<br />

Refine diagnostic work-up<br />

and physical examination<br />

no symptom<br />

improvement<br />

no systemic<br />

symptoms<br />

+/- oral<br />

thrust<br />

Consider empiric treatment<br />

(systemic fluconazole for<br />

10–14 days)<br />

improvement<br />

of symptoms<br />

Follow-up program: recurrent<br />

problems after therapy;<br />

therapy to correct<br />

immunosuppression<br />

confluent; a ‘shaggy’ aspect of the esophagus<br />

ensues. 11,12 However, a normal barium esophagram<br />

does not exclude esophageal candidiasis. It should be<br />

remarked that a well-circumscribed ulceration should<br />

not indicate Candida infection of the esophagus.


32<br />

Infectious esophagitis<br />

Non-invasive methods to diagnose esophageal<br />

infections without endoscopy are cytology brush<br />

and balloon devices. The latter consists of a tube<br />

with a ridged cytology balloon on the distal end:<br />

when inflated, it can be withdrawn from the<br />

esophagus and the brushings removed and submitted<br />

for cytological evaluation as well as viral<br />

culture. Whereas these methods are useful to identify<br />

Candida, they are less sensitive for viral<br />

disease and have no real advantage over empirical<br />

antifungal therapy. 13<br />

Endoscopy with biopsy is the gold standard for<br />

diagnosing esophageal candidiasis because<br />

mucosal biopsies can be performed. Despite the<br />

fact that gross endoscopic features can suggest a<br />

diagnosis of esophageal candidiasis, this is<br />

confirmed by the presence of hyphae in biopsies.<br />

Cytological specimens, which can be obtained at<br />

the time of endoscopy, are also a sensitive tool for<br />

the diagnosis, especially when organisms are<br />

washed off the tissue surface in mild superficial<br />

candidiasis after processing of bioptic specimens.<br />

When endoscopy is performed in patients with<br />

esophageal candidiasis, removal of plaque material<br />

by scraping with the endoscope is important to<br />

evaluate the underlying mucosa for ulcers. 6 In<br />

general, Candida should not be considered a cause<br />

of esophageal ulcers in HIV-infected patients.<br />

Both oral and intravenous drugs are available for<br />

treating esophageal candidiasis. Oral medications<br />

are generally administered first, whereas intravenous<br />

therapy is reserved for refractory cases or<br />

when oral administration is contraindicated. Nonsystemic<br />

locally acting agents such as nystatin or<br />

clotrimazole are not very effective and must be<br />

reserved for the treatment of oropharyngeal<br />

disease. In patients with mild disease, with<br />

minimal or reversible immunodeficiency, a short<br />

course of therapy with a systemically absorbed<br />

agent can be given, whereas in patients with HIV<br />

infection or with transplantation immunodeficiency,<br />

longer courses of azoles are best given.<br />

Patients with granulocytopenia, at risk for<br />

systemic Candida infection, require the use of<br />

systemically acting intravenous agents such as<br />

azoles or amphotericin B. All available oral agents<br />

(ketoconazole, fluconazole and itraconazole) are<br />

efficacious for the treatment of esophageal<br />

candidiasis (Table 3.3). Ketoconazole and itraconazole<br />

should be avoided in patients requiring anti-<br />

acid therapy, as an alkaline pH limits their<br />

bioavailability. Fluconazole appears to be more<br />

efficacious than itraconazole; a poor response to<br />

fluconazole suggests non-compliance, drug resistance<br />

or other causes for esophageal symptoms. 14<br />

Randomized trials suggest that fluconazole is<br />

significantly more effective for the treatment of<br />

esophageal candidiasis in HIV-infected patients<br />

than ketoconazole and itraconazole. 14 Fluconazole<br />

is available in oral and intravenous preparations,<br />

is minimally metabolized, is highly water soluble<br />

and is slightly protein bound. The new oral<br />

suspension formulations of fluconazole and itraconazole<br />

may have superior efficacy over pills due<br />

to an additional local antifungal effect and<br />

improved absorption. 15 The adverse effects of ketoconazole,<br />

fluconazole and itraconazole are dose<br />

dependent and include nausea, hepatotoxicity,<br />

inhibition of steroid production and cyclosporin<br />

metabolism. 16 The latter effect is more pronounced<br />

with ketoconazole. 17 Minor increases in aminotransferases<br />

are commonly found in patients<br />

treated with oral azoles and do not require drug<br />

discontinuation.<br />

Amphotericin B represents the other family of<br />

antifungal agents (polyene antibiotics), which bind<br />

irreversibly to sterol in fungal cell membranes,<br />

causing cell death. This drug has a limited use for<br />

the treatment of esophageal candidiasis, owing to<br />

its severe side-effects (nephro- and hepatotoxicity).<br />

This drug is now available in an oral<br />

formulation. Patients with esophageal candidiasis<br />

that is resistant to treatment with fluconazole or<br />

other azoles can be treated effectively with lower<br />

doses of intravenous amphotericin B.<br />

The prophylaxis of esophageal candidiasis in<br />

malignancy and transplant patients has yielded<br />

controversial results. 18<br />

Viral infections<br />

HSV and CMV are the most common viral agents<br />

involved in infectious esophagitis, although some<br />

cases have been ascribed to HIV infection of the<br />

esophagus. HSV (HSV I or, rarely, HSV II)<br />

esophagitis causes a self-limited disease in normal<br />

subjects, but it can be severe and prolonged in the<br />

compromised patients.


Table 3.3 Drugs for esophageal candidiasis<br />

Drug/efficacy Duration Dosage Remarks<br />

HSV esophagitis occurs as a primary infection or<br />

as a reactivation of previously latent HSV, especially<br />

in the compromised patient. It is particularly<br />

common in patients receiving immunosuppression<br />

for solid organ or bone marrow transplantation,<br />

19 whereas it is infrequently reported in HIVinfected<br />

patients. 4 HSV generally infects the<br />

squamous epithelium, where the earliest lesion is<br />

a vesicle. As vesicles enlarge and ulcerate, they<br />

Viral infections 33<br />

Ketoconazole 7–14 days 5–10 mg/kg/per day Liver disorders can occur during therapy.<br />

(80%) infections; 6–12mg/kg/per day reactions are most common adverse events.<br />

for systemic infections May cause clinical hepatitis, cholestasis<br />

and fulminant hepatic failure with<br />

underlying clinical conditions (e.g. AIDS,<br />

malignancy)<br />

Itraconazole 7–14 days 3–5 mg/kg/per day Caution in liver insufficiencies; nausea,<br />

(approximately vomiting, diarrhea and abdominal<br />

80%) discomfort may occur; high doses may<br />

produce hypertension, hypokalemia or<br />

edema<br />

Amphotericin B 7 days 0.3–1.5 mg/kg/per day i.v. Infusion-related toxicity includes acute<br />

(>95%) infused in 5% dextrose over reactions occurring about 30–45 min after<br />

2–4 h starting infusion; typically chills, fever and<br />

tachypnea may present; patients receiving<br />

any parenteral form of the drug must be<br />

monitored for renal and liver functions;<br />

normochromic normocytic anemia may<br />

develop, usually after 7–10 days of therapy<br />

Amphotericin B, 7 days ABLC (amphotericin B lipid Novel lipid formulations of amphotericin B<br />

lipid formulations complex): 5 mg/kg/per day i.v. delivering higher concentrations of the drug<br />

(>95%) for 1–2 h; ABCD (amphotericin with a theoretical increase in the<br />

B colloidal dispersion): therapeutic potential and decreased<br />

3–6 mg/kg/per day i.v. for nephrotoxicity<br />

1–2 h; L-AMB (liposomal<br />

amphotericin B): 1–7 mg/kg<br />

per day i.v. for 1–2 h<br />

tend to form larger lesions with a characteristic<br />

central ulceration and raised edges.<br />

HSV esophagitis usually presents with a sudden<br />

onset of severe odynophagia, with inability to<br />

swallow liquids or solids. Herpes labialis and<br />

herpetic oropharyngeal ulcers can occur during<br />

the esophageal infection, whereas skin lesions are<br />

rarely present. 20


34<br />

Infectious esophagitis<br />

A definite diagnosis of HSV esophagitis requires<br />

endoscopy and histology of the esophageal<br />

mucosa, even though contrast radiographic<br />

appearance of the esophagus may be suggestive by<br />

showing multiple vesicles and ulcers as stellate or<br />

volcano structures. Endoscopy usually reveals<br />

small, well-circumscribed ulcers (Figure 3.3),<br />

rarely vesicles, whereas deep ulcers, as seen with<br />

CMV, are rare. 21,22 Brushing or biopsies should be<br />

taken from the edge or periphery of ulcerations,<br />

given that HSV infects squamous epithelial cells. 23<br />

Histology reveals intranuclear Cowdry type A<br />

inclusion bodies (eosinophilic material), ballooning<br />

degeneration cells, multinucleated giant cells,<br />

ground-glass-like nuclei and margination of chromatin.<br />

24 Confirmation may require immunoperoxidase<br />

staining or positive viral culture.<br />

Although HSV esophagitis has a spontaneous resolution<br />

in a normal host, antiviral therapy is<br />

commonly used both in immunocompetent and<br />

immunocompromised subjects. Several uncontrolled<br />

trials and clinical experience indicate the<br />

efficacy of acyclovir, a nucleoside analog, for the<br />

treatment of HSV esophagitis. 25 Parenteral<br />

acyclovir should be initiated until the patient can<br />

be converted to oral therapy (when dysphagia or<br />

odynophagia is resolved). Generally, patients are<br />

treated with acyclovir for 7–10 days. Because resistance<br />

to acyclovir has been reported, therapy with<br />

foscarnet should be considered in case of clinical<br />

failure with acyclovir. 25 Acyclovir is efficacious in<br />

prophylaxis for patients undergoing transplantation<br />

and those who are HSV-antibody-positive.<br />

Table 3.4 lists the available antiviral agents for<br />

treating viral esophagitis.<br />

CMV is the most frequent infectious complication<br />

of organ transplantation, occurring in 60–70% of<br />

these patients. Esophagitis is one of the most<br />

common manifestations in this setting. CMV is by<br />

far the most frequent cause of esophageal ulcer in<br />

patients with advanced HIV infection and a CD4<br />

count lower than 100mm 3 (see reference 26). In<br />

contrast to HSV esophagitis, CMV esophagitis has<br />

rarely been detected in an immunocompetent<br />

host; 27 however, CMV and HSV are equally<br />

common organisms in transplant patients who do<br />

not receive antiviral prophylaxis. 28 Odynophagia<br />

is a constant feature and is typically severe; chest<br />

pain of esophageal origin, mainly occurring upon<br />

deglutition, is also described. Prior or co-existent<br />

Figure 3.3 Herpes simplex virus esophagitis. Small, wellcircumscribed<br />

ulcers (short arrows) and two small vesicles<br />

(long arrows) in the lower third of the esophageal mucosa.<br />

CMV infection in other regions (e.g. retinitis,<br />

colitis) is not uncommonly found. 29<br />

As with HSV, a definite diagnosis of CMV<br />

esophagitis requires endoscopy and biopsy.<br />

Contrast X-ray examination of the esophagus<br />

reveals either focal or diffuse images of mucosal<br />

ulcerations. Ulcers may be vertical or linear with<br />

central umbilication, or may be diffuse and superficial;<br />

30 they are usually deep and large in patients<br />

with advanced HIV infection. Endoscopy remains<br />

the definitive diagnostic tool for CMV esophagitis.<br />

The endoscopic appearance is variable and may<br />

include multiple shallow ulcers, solitary ulcers or<br />

diffuse superficial esophagitis (Figure 3.4). In<br />

contrast to Candida and HSV, brushing cytology<br />

has a poor sensitivity and multiple biopsies should<br />

be taken. Since the cytopathic effect of CMV<br />

occurs at the level of endothelial and mesenchymal<br />

cells in the granulation tissue, endoscopic<br />

biopsies must be taken from the base of the ulcer. 31<br />

Histology typically shows large cells with intranuclear<br />

and intracytoplasmic inclusions having an<br />

eosinophilic appearance. Immunohistochemical<br />

stains can reveal more infected cells than routinely<br />

appreciated with hematoxylin and eosin staining.<br />

Mucosal biopsy is also more specific than viral<br />

culture as with other esophageal infections. 32


The drugs available for treating CMV disease<br />

include ganciclovir, an acyclovir derivative,<br />

foscarnet and cidofovir. CMV esophagitis responds<br />

clinically and endoscopically to ganciclovir in<br />

approximately 75–80% of patients (Table 3.4). 29<br />

Duration of treatment should be based on clinical<br />

and endoscopic variables, but there is wide agreement<br />

that a 2–4-week period is usually effective.<br />

Oral ganciclovir is not effective for treating active<br />

infection, owing to its low bioavailability<br />

(≤ 10%). 33 If an acute CMV infection occurs in<br />

transplant patients, ganciclovir should be given for<br />

about 1–2 months, until immunosuppressive<br />

drugs are reduced or discontinued. In patients<br />

receiving a protracted course of therapy, clinical<br />

and virological resistance to ganciclovir may<br />

occur. 34 In this condition foscarnet, a pyrophosphate<br />

analog inhibiting DNA polymerase and<br />

reverse transcriptase, is usually effective. 35 The<br />

relapse rate of CMV esophagitis in HIV-infected<br />

patients is approximately 50%, 29 but long-term<br />

ganciclovir maintenance therapy is not routinely<br />

recommended unless there is a co-existent retinitis.<br />

33<br />

Ganciclovir and high-dose acyclovir have been<br />

used with moderate success for the prophylaxis of<br />

CMV infection in transplant patients. However, in<br />

Figure 3.4 Cytomegalovirus esophagitis. A round,<br />

solitary deep ulcer is evident in the distal esophageal<br />

mucosa (arrow).<br />

Other viral pathogens 35<br />

this setting ganciclovir prophylaxis is limited to<br />

high-risk patients, i.e. CMV-seropositive patients,<br />

CMV-seronegative patients receiving CMVseropositive<br />

organ or blood products, and patients<br />

treated with immunosuppressive drugs because of<br />

episodes of rejection. 36<br />

Other viral pathogens<br />

Varicella zoster virus (VZV) is a relatively uncommon<br />

agent of infectious esophagitis in immunologically<br />

normal subjects, but it causes a severe<br />

esophagitis in immunocompromised patients,<br />

usually accompanied by other signs of systemic<br />

dissemination (e.g. pneumonitis, hepatitis,<br />

encephalitis). 37 The endoscopic appearance ranges<br />

from vesicles to necrotic ulcerations. Definite diagnosis<br />

requires biopsies both for routine histology<br />

(ballooning degeneration, multinucleated giant cells,<br />

intranuclear eosinophilic inclusion bodies) and for<br />

culture and immunohistochemical staining. The<br />

infection is treated with acyclovir or famciclovir;<br />

foscarnet is used for resistant viruses.<br />

In a subject with clinical features consistent with<br />

infectious mononucleosis due to Epstein–Barr virus<br />

(EBV), the occurrence of nausea, dysphagia or<br />

hematemesis should raise the possibility of an infectious<br />

esophagitis. This however, develops only in a<br />

small minority of immunocompetent individuals,<br />

whereas in immunocompromised subjects, EBVrelated<br />

infectious esophagitis has been reported. 38<br />

Oral acyclovir may be a reasonable therapy, even if<br />

benefits of antiviral therapy in EBV-related esophagitis<br />

are unproven.<br />

The human papillovirus, like HSV, usually infects<br />

squamous epithelial cells. The diagnosis requires<br />

histology 39 (multinucleated giant cells, koilocytosis,<br />

cellular atypia) and immunohistochemical staining.<br />

Endoscopy reveals non-specific lesions such as<br />

yellow plaques, small nodules or patches with small<br />

villous projections. The condition is generally<br />

asymptomatic, and specific treatment may be<br />

unnecessary<br />

In specific HIV-related esophagitis, HIV is a wellknown<br />

risk factor for many other infections.<br />

However, it is agreed that HIV itself can lead to<br />

esophageal ulcerations, 40 usually presenting as<br />

multiple, small and shallow lesions. This esophageal


36<br />

Infectious esophagitis<br />

Table 3.4 Drugs for viral esophagitis<br />

Drug Duration Dosage Remarks<br />

Acyclovir 7–10 days 20mg/kg per dose orally four A nucleoside analog available for both oral and<br />

times daily (80 mg/kg per day). intravenous administration. It is well tolerated;<br />

Severe infections refractory to occasionally, rash, reversible renal failure or<br />

normal oral dosages or in gastrointestinal symptoms occur. Dose<br />

which the patient is unable adjustment should be made with renal<br />

to swallow are treated with impairment. Risk of renal insufficiency is<br />

i.v. acyclovir, 15–30 mg/kg per reduced with adequate prehydration<br />

day in 3 divided doses<br />

Ganciclovir 14–28 days 5 mg/kg given intravenously twice Drugs of first choice for the treatment of serious<br />

per day systemic CMV infections. The need for<br />

maintenance therapy of gastrointestinal CMV<br />

infection remains unclear. Dosage reduction is<br />

recommended in patients with renal<br />

impairment. Main side-effects are: neutropenia,<br />

thrombocytopenia, central nervous system<br />

symptoms, abnormal liver function tests, fever<br />

and rash. Simultaneous administration of G-<br />

CSF or GM-CSF can prevent ganciclovirassociated<br />

neutropenia<br />

Foscarnet 14–28 days HSV infection: 40 mg/kg of body Second-line parenteral antiviral, reserved for<br />

weight given either every 8 or treatment of HSV or CMV infections resistant to<br />

every 12 h. This dose is injected conventional therapy. Main side-effects are:<br />

slowly into a vein by an infusion nephrotoxicity, anemia, gastrointestinal<br />

pump over at least 1 h. CMV toxicity, hyper- and hypocalcemia,<br />

infection: 60 mg/kg, as above hypomagnesemia, hyper- and<br />

hypophosphatemia, hypokalemia, seizures. It is<br />

mainly used in the treatment of acyclovirresistant<br />

HSV infections and ganciclovirresistant<br />

CMV infections<br />

Famciclovir 7 days In adults: 250 mg, three times Consider with acyclovir resistance, but limited<br />

daily orally experience in infectious esophagitis. Use with<br />

caution in subjects with renal insufficiency.<br />

Main side-effects similar to those for acyclovir<br />

Cidofovir 14 days 5 mg/kg i.v. once per week A novel monophosphate nucleotide analog<br />

effective against CMV, HVS-1, HVS-2 and EBV.<br />

Parenteral antiviral recently approved to treat<br />

CMV retinitis. No reports on the treatment of<br />

CMV esophagitis. Generally reserved for<br />

patients with serious CMV disease for whom<br />

ganciclovir and/or foscarnet therapy has failed.<br />

Main side-effects are: nephrotoxicity,<br />

proteinuria, glycosuria, neutropenia and<br />

metabolic acidosis<br />

CMV, cytomegalovirus; G, granulocyte; CSF, colony-stimulating factor; GM, granulocyte/macrophage; HSV, herpes<br />

simplex virus; EBV, Epstein–Barr virus


involvement may occur in the setting of a mononucleosis-like<br />

illness that develops around the time of<br />

the primary HIV infection. Biopsy specimens examined<br />

at electronic microscopy show enveloped viruslike<br />

particles with morphology compatible with that<br />

of retroviruses. In the later stages of HIV infection,<br />

when the CD4 lymphocyte count is below 100/mm 3 ,<br />

large esophageal ulcerations can be observed. These<br />

ulcers are commonly defined as idiopathic<br />

esophageal ulcers (IEU) and appear as uniformly<br />

well-circumscribed lesions, without histological<br />

features of a viral cytopathic effect. It has been<br />

observed that HIV-associated IEU are diagnosed<br />

when biopsy, cytology and cultures are negative.<br />

Electron micrographs from the margins of some<br />

superficial ulcers have revealed viral particles<br />

morphologically similar to retroviruses. The polymerase<br />

chain reaction (PCR) can identify the HIV<br />

genome in biopsy specimens, but it remains unclear<br />

whether HIV is pathogenic in esophageal ulcerations.<br />

Reports have revealed HIV histopathologically<br />

in esophageal biopsies from patients with Candida,<br />

HSV and CMV. It has been hypothesized that most<br />

esophageal disease in patients with advanced HIV<br />

infection is associated with specific pathogenic<br />

processes. However, HIV-induced ulcerations can<br />

occur and may require treatment with corticosteroids.<br />

41 IEU present clinically in a fashion similar<br />

to CMV esophagitis; whereas bleeding is reported,<br />

strictures are rare. These ulcers usually respond well<br />

to oral corticosteroids or thalidomide. Despite the<br />

fact that these drugs may entail some risk for<br />

patients already immunosuppressed, this treatment<br />

is usually well tolerated.<br />

REFERENCES<br />

1. Bacellar H, Munoz A, Hoover DR et al. Incidence of<br />

clinical AIDS conditions in a cohort of homosexual men<br />

with CD4+ cell counts


38<br />

Infectious esophagitis<br />

5. Connolly GM, Hawkins D, Harcourt-Webster JN et al.<br />

Oesophageal symptoms, their causes, treatment, and<br />

prognosis in patients with the acquired immunodeficiency<br />

syndrome. Gut 1989; 30: 1033-1039.<br />

6. Wilcox CM. A technique to examine the underlying<br />

mucosa in patients with AIDS and severe Candida<br />

esophagitis. Gastrointest Endosc 1995; 42: 360–363.<br />

7. Slavin MA, Osborne B, Adams R et al. Efficacy and<br />

safety of fluconazole prophylaxis for fungal infections<br />

after marrow transplantation – a prospective, randomized,<br />

double-blind study. J Infect Dis 1995; 171:<br />

1545–1552.<br />

8. Wilcox CM, Schwartz DA. Endoscopic–pathologic correlates<br />

of Candida esophagitis in acquired immunodeficiency<br />

syndrome. Dig Dis Sci 1996; 41: 1337–1345.<br />

9. Lopez-Dupla M, Mora Sanz P, Pintado Garcia V et al.<br />

Clinical, endoscopic, immunologic, and therapeutic<br />

aspects of oropharyngeal and esophageal candidiasis in<br />

HIV-infected patients: a survey of 114 cases. Am J<br />

Gastroenterol 1992; 87: 1771–1776.<br />

10. Wilcox CM. Time course of clinical response to fluconazole<br />

for Candida oesophagitis in AIDS. Alimentar<br />

Pharmacol Ther 1994; 8: 347–350.<br />

11. Levine MS, Macones AJ Jr, Laufer I. Candida esophagitis:<br />

accuracy of radiographic diagnosis. Radiology 1985;<br />

154: 581–587.<br />

12. Roberts L Jr, Gibbons R, Gibbons G et al. Adult<br />

esophageal candidiasis: a radiographic spectrum.<br />

Radiographics 1987; 7: 289–307.<br />

13. Brandt LJ, Coman E, Schwartz E et al. Use of a new<br />

cytology balloon for diagnosis of symptomatic<br />

esophageal disease in acquired immunodeficiency<br />

syndrome. Gastrointest Endosc 1993; 39: 559–561.<br />

14. Barbaro G, Barbarini G, Calderon W et al. Fluconazole<br />

versus itraconazole for Candida esophagitis in acquired<br />

immunodeficiency syndrome. Gastroenterology 1996;<br />

111: 1169–1177.<br />

15. Laine L, Rabeneck L. Prospective study of fluconazole<br />

suspension for the treatment of oesophageal candidiasis<br />

in patients with AIDS. Aliment Pharmacol Ther 1995; 9:<br />

553–556.<br />

16. Como JA, Dismukes WE. Oral azole drugs as systemic<br />

antifungal therapy. N Engl J Med 1994; 330: 263–272.<br />

17. Lewis JH, Zimmerman HJ, Benson GD et al. Hepatic<br />

injury associated with ketoconazole therapy. Analysis of<br />

33 cases. Gastroenterology 1984; 86: 503–513.<br />

18. Wilcox CM, Darouiche RO, Laine L et al. A randomized,<br />

double-blind comparison of itraconazole oral solution<br />

and fluconazole tablets in the treatment of esophageal<br />

candidiasis. J Infect Dis 1997; 176: 227–232.<br />

19. Mosimann F, Cuenoud PF, Steinhauslin F et al. Herpes<br />

simplex esophagitis after renal transplantation.<br />

Transplant Int 1994; 7: 79–82.<br />

20. Baehr PH, McDonald GB. Esophageal infections: risk<br />

factors, presentation, diagnosis, and treatment.<br />

Gastroenterology 1994; 106: 509–532.<br />

21. McBane RD, Gross JB Jr. Herpes esophagitis: clinical<br />

syndrome, endoscopic appearance, and diagnosis in 23<br />

patients. Gastrointest Endosc 1991; 37: 600–603.<br />

22. Agha FP, Lee HH, Nostrant TT. Herpetic esophagitis: a<br />

diagnostic challenge in immunocompromised patients.<br />

Am J Gastroenterol 1986; 81: 246–253.<br />

23. Ramanathan J, Rammouni M, Baran J Jr et al. Herpes<br />

simplex virus esophagitis in immunocompetent host: an<br />

overview. Am J Gastroenterol 2000; 95: 2171–2176.<br />

24. Feiden W, Borchard F, Burrig KF et al. Herpes<br />

oesophagitis. I. Light microscopical and immunohistochemical<br />

investigations. Virchows Arch Pathol Anat<br />

Histopathol 1984; 404: 167–176.<br />

25. Genereau T, Lortholary O, Bouchaud O et al. Herpes<br />

simplex esophagitis in patients with AIDS: report of 34<br />

cases. The Cooperative Study Group on Herpetic<br />

Esophagitis in HIV Infection. Clin Infect Dis 1996; 22:<br />

926–931.<br />

26. Wilcox CM, Schwartz DA, Clark WS. Esophageal ulceration<br />

in human immunodeficiency virus infection.<br />

Causes, response to therapy, and long-term outcome.<br />

Ann Intern Med 1995; 123: 143–149.<br />

27. Altman C, Bedossa P, Dussaix E et al. Cytomegalovirus<br />

infection of esophagus in immunocompetent adults. Dig<br />

Dis Sci 1995; 40: 606.<br />

28. Alexander JA, Brouillette DE, Chien MC et al. Infectious<br />

esophagitis following renal and liver transplantation.<br />

Dig Dis Sci 1988; 33: 1121.<br />

29. Wilcox CM, Straub RF, Schwartz DA. Cytomegalovirus<br />

esophagitis in AIDS: a prospective study of clinical<br />

response to ganciclovir therapy, relapse rate, and longterm<br />

outcome. Am J Med 1995; 98: 169.<br />

30. Balthazar EJ, Megibow AJ, Hulnick D et al.<br />

Cytomegalovirus esophagitis in AIDS: radiographic<br />

features in 16 patients. Am J Roentgenol 1987; 149: 919.<br />

31. Wilcox CM, Straub RF, Schwartz DA. A prospective<br />

evaluatuion of biopsy number for the diagnosis of viral<br />

esophagitis in patients with HIV infection and<br />

esophageal ulcer. Gastrointest Endosc 1996; 44: 587.<br />

32. Goodgame RW, Genta RM, Estrada R et al. Frequency of<br />

positive test for cytomegalovirus in AIDS patients:<br />

endoscopic lesions compared with normal mucosa. Am<br />

J Gastroenterol 1993; 88: 338.<br />

33. Crumpacker CS. Ganciclovir. N Engl J Med 1996; 335: 721.<br />

34. Erice A, Chou S, Biron KK et al. Progressive disease due<br />

to ganciclovir-resistant cytomegalovirus in immunocompromised<br />

patients. N Engl J Med 1989; 167: 1184.<br />

35. Blanshard C, Benhamou Y, Dohin E et al. Treatment of<br />

AIDS-associated gastrointestinal cytomegalovirus infection<br />

with foscarnet and gancyclovir: a randomized<br />

comparison. J Infect Dis 1995; 172: 622.<br />

36. Winston DJ, Wirin D, Shaked A et al. Randomised<br />

comparison of ganciclovir and high-dose acyclovir for<br />

long-term cytomegalovirus prophylaxis in liver-transplant<br />

recipients. Lancet 1995; 346: 69.<br />

37. Gill RA, Gebhard RL, Dozeman RL et al. Shingles<br />

esophagitis: endoscopic diagnosis in two patients.<br />

Gastrointest Endosc 1984; 30: 26.<br />

38. Kitchen VS, Helbert M, Francis ND et al. Epstein–Barr<br />

virus associated oesophageal ulcers in AIDS. Gut 1990;<br />

31: 1223.<br />

39. Schechter M, Pannain VNL, de Oliveira AV.<br />

Papovavirus-associated esophageal ulceration in a<br />

patient with AIDS. AIDS 1991; 5: 238.<br />

40. Rabeneck L, Popovic M, Gartner S et al. Acute HIV<br />

infection presenting with painful swallowing and<br />

esophageal ulcers. JAMA 1990; 263: 2318–2322.<br />

41. Wilcox CM, Schwartz DA. A pilot study of oral corticosteroid<br />

therapy for idiopatic esophageal ulceration associated<br />

with human immunodeficiancy virus infection.<br />

Am J Med 1992; 93: 131–134.<br />

42. Ezzell JH, Bremer J, Adamec TA. Bacterial esophagitis:<br />

an often forgotten cause of odynophagia. Am J<br />

Gastroenterol 1990; 85: 296.


4<br />

Introduction<br />

Gastroesophageal reflux<br />

disease<br />

Yvan Vandenplas, Silvia Salvatore and Bruno Hauser<br />

Gastroesophageal reflux (GER) is defined as the<br />

involuntary passage of gastric contents into the<br />

esophagus. GER is a physiological phenomenon,<br />

occurring in every individual. Most episodes of<br />

reflux are limited to the distal esophagus, and are<br />

brief and asymptomatic. The difference between<br />

physiological reflux and reflux disease is to a<br />

lesser extent defined by the frequency, duration<br />

and severity of the reflux episodes, than whether<br />

the reflux episodes result in the occurrence of<br />

symptoms, signs severe enough to impair the<br />

quality of life, or complications. GER disease<br />

(GERD) is reflux associated with mucosal damage<br />

or symptoms severe enough to impair quality of<br />

life. 1,2 When this occurs, and the forces overcome<br />

the defense, the esophagus may become damaged,<br />

with significant consequences for the affected<br />

individual.<br />

Definitions<br />

Regurgitation is defined as passage of refluxed<br />

gastric contents into the oral pharynx and the<br />

mouth and is accompanied by gastric content<br />

drooling out of the mouth. Spitting-up is synonymous<br />

with regurgitation. Vomiting is defined as<br />

expulsion of the refluxed gastric contents from the<br />

mouth. 3,4 Only a minority of reflux episodes is<br />

accompanied by regurgitation or vomiting.<br />

Rumination is characterized by the voluntary,<br />

habitual regurgitation of recently ingested food<br />

that is subsequently spitted up or re-swallowed.<br />

Sometimes GER is a normal esophageal function,<br />

serving a protective role, e.g. during meals, or in<br />

the immediate postprandial period; if the stomach<br />

is overdistended, GER serves to decompress it.<br />

Regurgitation may be physiological in healthy,<br />

thriving, happy infants. Primary GER results from<br />

a primary disorder of function of the upper<br />

gastrointestinal tract. In secondary GER, reflux<br />

results from dysmotility occurring in systemic<br />

disorders such as neurological impairment or<br />

systemic sclerosis. It may also result from mechanical<br />

factors at play in chronic lung disease or upper<br />

airway obstruction, as in chronic tonsillitis. Other<br />

causes include systemic or local infections<br />

(urinary tract infection, gastroenteritis), food<br />

allergy, metabolic disorders, intracranial hypertension<br />

and medications such as chemotherapy. In<br />

some cases, secondary reflux results from stimulation<br />

of the vomiting center by afferent impulses<br />

from circulating bacterial toxins, or stimulation<br />

from sites such as the eye, olfactory epithelium,<br />

labyrinths, pharynx, gastrointestinal and urinary<br />

tracts, and testes. 3,4 These stimuli usually cause<br />

vomiting. The symptoms and signs of primary and<br />

secondary reflux are similar, but a distinction is<br />

conceptually helpful in determining a therapeutic<br />

approach. Secondary GER is not discussed further.<br />

Epidemiology<br />

Determination of the exact prevalence of GER and<br />

GERD at any age is virtually impossible because<br />

most reflux episodes are asymptomatic, show the<br />

absence of specific symptoms, undergo selftreatment<br />

and lack medical referral. In normal 3–4month-old<br />

infants, three or four episodes of GER<br />

are detectable during 5 min of intermittent fluoroscopic<br />

evaluation, 5 and up to 31±21 acid reflux<br />

episodes are recorded within a 24-h period with<br />

pH monitoring. 6<br />

The frequency of regurgitation varies according to<br />

age. Daily regurgitation is present in 50% of infants<br />

39


40<br />

Gastroesophageal reflux disease<br />

younger than 3 months and in more than 66% at 4<br />

months, but only in 5% at 1 year of age. 7–9<br />

Complete resolution of regurgitation is frequent<br />

and expected, by 10 months in 55%, by 18 months<br />

in 60–80% and by 2 years of age in 98%. 10 A<br />

prospective follow-up of 63 regurgitating infants<br />

reported in all subjects, before 12 months, a<br />

complete disappearance of symptoms, although<br />

accompanied by a significant increase in feeding<br />

refusal, duration of meals, parental feeding-related<br />

distress and impaired quality of life, even after the<br />

disappearance of symptoms. 11<br />

About 5–9% of infants have troublesome GERD. 6,12<br />

According to parents, heartburn is present in 1.8%<br />

of 3–9-year-old healthy children and 3.5% of 10–<br />

17-year-old adolescents; regurgitation is said to<br />

occur in 2.3% and 1.4%, respectively, and 0.5%<br />

and 1.9% need anti-acid medication. In selfreports,<br />

adolescents complain about heartburn in<br />

5.2% and regurgitation in up to 8.2%, while antiacids<br />

are taken by 2.3% and histamine receptor<br />

antagonists (H2RA) by 1.3%, suggesting that symptoms<br />

of GER are not rare during childhood and are<br />

underreported by the parents or overestimated by<br />

the adolescents. 13 In a Western population, GERD<br />

affected 4–30% of adults 14,15 and heartburn and<br />

regurgitation resolved within 3–10 years only in<br />

12–33%, regardless of the presence of esophagitis<br />

at diagnosis. 1,16,17 In the absence of H2RA or proton<br />

pump inhibitors (PPI), symptoms improved after<br />

17–22 years in 60% but disappeared in only 12%. 18<br />

Despite anti-secretory treatment, a 10-year followup<br />

of esophagitis showed that over 70% had<br />

persisting symptoms and 2% had strictures. 19<br />

Reflux esophagitis is reported in 2–5% of the<br />

general population. 12 Children with GER symptoms<br />

present esophagitis in 15 up to 62% of cases,<br />

Barrett’s esophagus in 0.1–3% and refractory<br />

GERD requiring surgery in 6–13%. 20–24 In adults<br />

undergoing endoscopy, esophagitis is diagnosed in<br />

15 up to 80% of cases. 1,17,25,26 The huge differences<br />

in incidence are determined by patient recruitment<br />

and availability of self-treatment.<br />

Genetic and environmental factors<br />

It has been demonstrated that physiological reflux,<br />

heartburn, acid regurgitation and severe GERD are<br />

more frequent in men than in women. Barrett’s<br />

esophagus is in part genetically determined. 24<br />

There is much information (in adults) demonstrating<br />

the aggravating effects of alcohol, smoking,<br />

drugs, dietary components, etc. on the incidence<br />

of GER. A detailed discussion on these environmental<br />

factors is beyond the scope of this chapter.<br />

Changes in lifestyle in men and women may result<br />

in the fact that the differences in incidence in<br />

GERD between both sexes may be disappearing.<br />

With pH monitoring, we could not demonstrate a<br />

male predominance in children. All forms of<br />

GERD affect Caucasians more often than African-<br />

Americans or Native Americans (adult data). 14<br />

However, the same prevalence of troublesome<br />

infant regurgitation was found in Caucasian and<br />

Indonesian infants. 27 The importance of the<br />

genetic background was hypothesized by demonstrating<br />

that esophagitis and hiatus hernia were<br />

more common in a population with the same<br />

genetic background with dyspeptic symptoms in<br />

England than in Singapore. 28 Race, sex, body mass<br />

index and age were independently associated with<br />

hiatus hernia and esophagitis, race being the most<br />

important risk factor. 28 However, over-the-counter<br />

use of low-dose aspirin and non-steroidal antiinflammatory<br />

drugs has a greater impact on severe<br />

GERD, such as esophageal stricture than age. 29<br />

Carre et al described the autosomal dominant<br />

inheritance of hiatal hernia by discovering familial<br />

hiatal hernia in a large five-generation family, but<br />

without demonstrating the link to GERD. 30 The<br />

genetic influence on GERD is supported by<br />

increased GER symptoms in relatives of GERD<br />

patients. 31 Moreover, the concordance for GER is<br />

higher in monozygotic than dizygotic twins. 32 A<br />

locus on chromosome 13q, between microsatellites<br />

D13S171 and D13S263, has been linked to<br />

severe GERD in five multiply affected families, 33<br />

but not confirmed in another five families, possibly<br />

owing to the genetic heterogeneity of GERD<br />

and the different clinical presentations of<br />

patients. 34<br />

Pathophysiology<br />

The pathophysiology of GER is complex and<br />

diverse, as it is influenced by factors that are<br />

genetic, environmental (e.g. diet and smoking),<br />

anatomic, hormonal and neurogenic (Figure 4.1<br />

and Table 4.1). We have recently reviewed the


Gastric distension<br />

(gastric mechanoreceptors near cardia)<br />

Vagally mediated abnormal neural control of LES by CNS<br />

Defective LES motility<br />

Increased TLESRs<br />

Low basal LES tone<br />

Increase in GER<br />

pathophysiology of GER in infants and children. 35<br />

The most relevant factors are gastric (gastric acid<br />

and non-acid content, gastric emptying, fundus<br />

tone, triggering of transient lower esophageal<br />

sphincter (LES) relaxations (TLESRs)), the antireflux<br />

barrier (LES basal pressure, TLESRs, hiatal<br />

hernia), the composition of the refluxate<br />

(air–liquid, acid, non-acid, bile), the esophageal<br />

tone and the esophageal clearance (volume and<br />

chemical clearance). 35,36 GER occurs during<br />

episodes of transient relaxation of the LES or inadequate<br />

adaptation of the sphincter tone to changes<br />

in abdominal pressure.<br />

GERD was classically considered to be an acid<br />

peptic disease, although as a group, the majority of<br />

patients with reflux disease do not have a significant<br />

increase in gastric acid secretion. Three major<br />

Gastric acid<br />

Hiatal hernia, obtuse angle of His<br />

Delayed (acid) clearance<br />

Delayed volume clearance (motility)<br />

Impaired pH neutralization (saliva, esophageal secretion)<br />

Poor mucosal resistance<br />

Increased abdominal pressure<br />

Genetic factors<br />

Posture, handling, physical activity, sleep state, feeding, drugs<br />

Environmental factors<br />

Pathophysiology 41<br />

Figure 4.1 Illustration of multifactorial influences on gastroesophageal reflux (GER). LES, lower esophageal sphincter;<br />

CNS, central nervous system; TLESRs, transient lower esophageal sphincter relaxations.<br />

tiers of defense serve to limit the degree of GER,<br />

and to minimize the risk of reflux-induced injury<br />

to the esophagus. The first line of defense is the<br />

‘anti-reflux barrier’, consisting of the LES, and the<br />

diaphragmatic pinchcock and angle of His; this<br />

barrier serves to limit the frequency and volume of<br />

refluxed gastric contents. When this line of<br />

defense fails, the second, esophageal clearance,<br />

assumes greater importance, to limit the duration<br />

of contact between luminal contents and<br />

esophageal epithelium. Gravity and esophageal<br />

peristalsis serve to remove volume from the<br />

esophageal lumen, while salivary and esophageal<br />

secretions (the latter from esophageal submucosal<br />

glands), serve to neutralize acid. The third line of<br />

defense, ‘tissue or esophageal mucosal resistance’<br />

comes into play when acid contact time is<br />

prolonged, such as when esophageal clearance is


42<br />

Gastroesophageal reflux disease<br />

Table 4.1 Parameters influencing the incidence<br />

of gastroesophageal reflux<br />

Mastication, saliva secretion<br />

Swallowing<br />

Esophageal clearance<br />

Esophageal innervation and receptors<br />

Mucosal resistance<br />

Lower esophageal sphincter pressure<br />

Sphincter relaxation<br />

Abdominal esophagus<br />

Sphincter position<br />

Angle of His<br />

Gastric volume, gastric accommodation<br />

Gastric emptying<br />

Gastric acid output<br />

Gastric acid feed buffering<br />

Feeding regimen: type, frequency, volume<br />

Pepsin/trypsin/bile salts<br />

<strong>Helicobacter</strong> <strong>pylori</strong><br />

Intra-abdominal pressure<br />

Genetic factors<br />

Environmental factors<br />

Posture<br />

Physical activity<br />

Sleep state<br />

Respiratory disease<br />

Medication (e.g. xanthines)<br />

defective or not operative (motility disorders,<br />

sleep). 35<br />

Epidemiological data suggest that <strong>Helicobacter</strong><br />

<strong>pylori</strong> plays a protective role in GERD, presumably<br />

by decreasing acid secretion. In adults,<br />

<strong>Helicobacter</strong> <strong>pylori</strong> and especially the cagA positive<br />

strains, which cause more severe gastric<br />

inflammation, are less prevalent in patients with<br />

esophagitis or Barrett’s esophagus than in those<br />

with endoscopically negative reflux disease or in<br />

control patients. 37–41<br />

Recent detailed analysis of postprandial acidity in<br />

the gastroesophageal junction area suggests that<br />

local acid distribution rather than total gastric<br />

secretion might be more relevant to the pathogenesis<br />

of GERD. 42 Differences may exist in the degree<br />

of mixing of fundal contents leading to different<br />

distributions of acid in the stomach. Studies using<br />

pH monitoring and esophageal scintigraphy and<br />

gastric magnetic resonance imaging suggest that<br />

gastric mixing can be incomplete and different<br />

layers of viscosity within the stomach might therefore<br />

influence the distribution of the gastric<br />

contents. A collection of acid in the gastric part of<br />

the esophageal junction was shown in adults in a<br />

supine position, even in the postprandial period<br />

when the stomach content was neutralized by the<br />

meal. 43,44 This newly described mechanism is<br />

attractive as an explanation for postprandial<br />

distress in infants lying in the supine position.<br />

TLESRs are the major mechanism of GER episodes,<br />

in premature infants and in adults. 35,45,46 TLESRs<br />

are a neural reflex, triggered mainly by the distension<br />

of the proximal stomach and organized in the<br />

brain stem, with efferent and afferent pathways<br />

traveling in the vagus nerve, activating an intramural<br />

inhibitory neuron which releases nitric<br />

oxide to relax the LES. 47,48 When investigated in<br />

the supine position, the incidence of TLESRs in<br />

healthy adults and those with GERD did not differ.<br />

In healthy adults, only 30% of the TLSERs were<br />

accompanied by acid reflux, but in patients with<br />

GERD the reflux occurred in 65% of the TLESRs.<br />

Thus adults, controls and GERD patients have the<br />

same incidence of TLESRs, but in patients with<br />

GERD these TLESRs are more than twice as<br />

frequently accompanied by acid GER. 36,49,50 The<br />

initial studies performed in the recumbent position<br />

found a higher frequency of TLSERs in<br />

patients with GERD than in normals. 51 These older<br />

studies in the recumbent position may be more<br />

relevant for infants.<br />

Delayed gastric emptying may increase postprandial<br />

reflux possibly by increasing the rate of<br />

TLESRs and the likelihood of reflux during the<br />

TLESRs. A disturbed gastric accommodation to a<br />

meal and prolonged postprandial fundic relaxation<br />

where described in patients with GERD. 52 Both<br />

phenomena can influence postprandial fundic<br />

volume and pressure, which in turn may affect the<br />

rate of distension-induced triggerings of TLESRs


and the volume of the refluxate. A recent study has<br />

shown that esophageal acid exposure in patients<br />

with GERD is directly correlated with the emptying<br />

time of the proximal stomach. 53 These new<br />

findings are especially of interest in infants with<br />

postprandial distress or regurgitation who are not<br />

responding to dietary treatment with thickened<br />

casein-predominant formula. This has a delayed<br />

gastric emptying time, and thus is related to an<br />

increased incidence of TLESRs.<br />

Hiatal hernia increases the number of reflux<br />

episodes and delays esophageal clearance by<br />

promoting retrograde flow across the esophagogastric<br />

junction when the LES relaxes after a<br />

swallow. This mechanism underlies the so-called<br />

re-reflux phenomenon (acid reflux when the pH is<br />

still below 4).<br />

The refluxate might be highly acid, moderately<br />

acid, or non-acid. Reflux may be a mixture of gas<br />

and liquid or pure liquid, and may or may not<br />

contain bile. More than half of the acid and nonacid<br />

reflux episodes are associated with reflux of<br />

gas. 36 Non-acid reflux also occurs predominantly<br />

during TLESRs. With liquid meals, patients with<br />

reflux disease had a similar total rate of reflux<br />

episodes but a higher proportion of acid reflux<br />

events than controls. 54 Non-acid reflux may be<br />

responsible for the remaining symptoms in<br />

patients under anti-secretory treatment. 55<br />

Acid reflux in patients with GERD is associated<br />

with an inhibition of tone in the esophageal body,<br />

whereas normals have an increased contractile<br />

activity. In order to have an effective volume clearance,<br />

motility of the esophageal body needs to be<br />

preserved. Acid is emptied from the esophagus<br />

with one or two sequences of primary peristalsis,<br />

then the residual acidity is neutralized by swallowed<br />

saliva. 35 Secondary peristalsis is the<br />

response to esophageal distension with air or<br />

water, and is more important during sleep, when<br />

peristalsis is reduced. Patients may have normal<br />

primary peristalsis but abnormal secondary peristalsis.<br />

Thus, non-acid reflux, as occurs in the<br />

postprandial period, may be inefficiently cleared<br />

and cause prolonged esophageal distension. The<br />

esophageal mucosal defense can be divided into<br />

pre-epithelial (protective factors in swallowed<br />

saliva and esophageal secretions containing bicarbonate,<br />

mucin, prostaglandin E2, epidermal<br />

Pathophysiology 43<br />

growth factor, transforming growth factor), epithelial<br />

(tight junctions, intercellular glycoprotein<br />

material) and post-epithelial factors. 35 There is a<br />

very important interindividual variation of reflux<br />

perception suggesting different esophagussensitive<br />

thresholds. The esophageal mucosa<br />

contains acid, and temperature- and volumesensitive<br />

receptors. A widening of the intercellular<br />

spaces has been found in patients with esophagitis<br />

and in patients with endoscopy-negative disease.<br />

When esophagitis heals, esophageal sensitivity to<br />

acid decreases. The presence of fat in the duodenum<br />

increases the sensitivity to reflux. Hyposensitivity,<br />

as occurs in patients with Barrett’s<br />

esophagus, is a secondary phenomenon. 56<br />

Clinical signs/symptoms<br />

The most typical, although non-specific, symptoms<br />

of esophageal dysfunction are GER, regurgitation<br />

and vomiting. While reflux does occur physiologically<br />

at all ages, there is a continuum<br />

between physiological GER and GERD leading to<br />

significant symptoms and complications. GERD is<br />

a spectrum of diseases that can best be defined as<br />

the symptoms and/or signs of esophageal or adjacent<br />

organ injury secondary to the reflux of gastric<br />

contents into the esophagus or, beyond, into the<br />

oral cavity or airways.<br />

Presentation may be with decreased food intake<br />

and aversive behavior around feeds. There is often<br />

clearly abnormal sucking and swallowing. Not<br />

surprisingly, the mother–child interaction is<br />

affected, making the situation less easily treated. 57<br />

There may be poor weight gain. These infants have<br />

no apparent malformations, and may be diagnosed<br />

as ‘non-organic failure to thrive’ (NOFTT), 58 a<br />

‘disorder’ that is sometimes attributed to<br />

social/sensory deprivation, socioeconomic or<br />

primary maternal–child problems. Primary GERD<br />

is but one root cause of ‘feeding problems’ in<br />

infancy, others being structural abnormalities of<br />

the mouth/pharynx/upper gastrointestinal tract,<br />

neurological conditions, primary behavior disorders,<br />

cardiorespiratory problems, or metabolic<br />

dysfunction. However, no matter what the cause,<br />

so-called ‘feeding problems’ are bio-behavioral<br />

conditions, i.e. disorders in which biological and<br />

behavioral causes interact. 59


44<br />

Gastroesophageal reflux disease<br />

A wide spectrum of clinical presentations of GERD<br />

exists, with relevant differences within ages (Table<br />

4.2). Regurgitation is the most common presentation<br />

of infantile GER, with occasional projectile<br />

vomiting. 7,60 In infants and young children, verbal<br />

expression of symptoms is often vague or impossible,<br />

and persistent crying, irritability, feeding and<br />

sleeping difficulties have been proposed as equivalents<br />

for heartburn in adults. Nevertheless, the<br />

descriptions for infants are non-specific and even<br />

functional. Therefore, poor weight gain, feeding<br />

refusal, back-arching, irritability and sleep disturbances<br />

have also been reported to be unrelated to<br />

GERD. 61,62 Esophageal pain and behaviors<br />

perceived by the caregiver (usually the mother) to<br />

represent pain (e.g. crying and retching) potentially<br />

affect the response of the infant to visceral<br />

stimuli and the ability to cope with these sensations,<br />

either painful or non-painful. 63 In addition,<br />

cow’s milk allergy (CMA) may overlap with many<br />

symptoms of GER, and may coexist or complicate<br />

GERD in up to 40% of infants. 64–66<br />

Table 4.2 Spectrum of gastroesophageal reflux disease manifestations according to different ages<br />

Symptoms and signs Infants Children Adults<br />

Vomiting ++ ++ +<br />

Regurgitation +++ ++ ++<br />

Heartburn/pyrosis ? ++ +++<br />

Epigastric pain ? + ++<br />

Chest pain ? + ++<br />

Dysphagia ? + ++<br />

Excessive crying/irritability +++ + -<br />

Anemia/melena/hematemesis + + +<br />

Food refusal/feeding disturbancies/anorexia ++ + +<br />

Failure to thrive/poor growth ++ + -<br />

Abnormal posturing/Sandifer’s syndrome ++ + -<br />

Persisting hiccups ++ + +<br />

Dental erosions/halitosis/water brush - + +<br />

Hoarseness/globus pharyngeus - + +<br />

Persistent cough/aspiration pneumonia/wheezing + + +<br />

Laryngitis/ear problems + + +<br />

Laryngomalacia/stridor/croup + + -<br />

Laryngostenosis/resistant asthma/chronic sinusitis - + +<br />

Vocal nodule problems - - +<br />

ALTE/SIDS/apnea/desaturation + - -<br />

Bradycardia + ? ?<br />

Sleeping disturbancies + + +<br />

Impaired quality of life ? ? +++<br />

Esophagitis + + ++<br />

Stenosis - (+) +<br />

Barrett’s esophagus/esophageal adenocarcinoma - (+) +<br />

ALTE, apparent life-threatening events; SIDS, sudden infant death syndrome<br />

+++ very common; ++ common; + possible; (+) rare; - absent; ? unknown


Compared to adults, children report more regurgitation<br />

and emesis and less heartburn, dysphagia<br />

and chest pain. 13,23,67 The younger the children<br />

are, the more difficult it is to describe and perceive<br />

these ‘unpleasant sensations’. In 69 children with<br />

GERD, regurgitation and vomiting occurred in<br />

72%, symptoms attributed to the esophagus<br />

(epigastric/abdominal pain, feeding difficulties,<br />

irritability and Sandifer–Sutcliffe syndrome) in<br />

68%, failure to thrive in 28%, chronic respiratory<br />

symptoms in 13% and recurrent apnea in 12%,<br />

with more feeding difficulties in toddlers and more<br />

irritability in infants. 21 Clinical distinction is,<br />

however, not simple, as GERD may be occult or<br />

masquerade as respiratory or other manifestations<br />

co-existing at different ages.<br />

GERD in adolescents is more adult-like. Heartburn<br />

is the predominant GER symptom, occurring<br />

weekly in 15–20% 15,68 and daily in 5–10% of<br />

subjects. 16 Atypical symptoms such as epigastric<br />

pain, nausea, flatulence, hiccups, chronic cough,<br />

asthma, chest pain, hoarseness and earache,<br />

account for 30–60% of presentations of GERD. 1,16<br />

GERD is diagnosed in 50% of the adult patients<br />

with chest pain and in 80% presenting with<br />

chronic hoarseness and asthma. 69 The incidence<br />

of GERD in this group with atypical symptoms is<br />

determined by the selection (bias) of the patients.<br />

Recent evidence has shown that GERD affects the<br />

quality of life significantly in adults, and probably<br />

also in children (and their parents), although<br />

quality of life is more difficult to evaluate in<br />

infants and young children. The developing<br />

nervous system of infants exposed to acid seems<br />

susceptible to pain hypersensitivity despite the<br />

absence of tissue damage. 70 The role of hypersensitivity<br />

to dietary allergens, both in exclusively<br />

breast-fed and formula-fed infants, is likely to be<br />

underestimated at present. The ‘hygiene hypothesis’<br />

suggests that the Th2-predominant immune<br />

response at birth in the industrialized world is<br />

insufficiently skewed towards a well-balanced<br />

Th1/Th2 response. 71 Lack of controlled chronic or<br />

repetitive inflammation of the mucosa during the<br />

first months of life may account for the dramatic<br />

increase in atopic disease during infancy and<br />

childhood (which is a Th2 response) and the<br />

increase in autoimmune diseases such as diabetes<br />

and Crohn’s disease in adolescents and adults<br />

Pathophysiology 45<br />

(which are a Th1 response). 72 This hypothesis fits<br />

well with the observations that atopy is inversely<br />

related to family income and early attendance at a<br />

day-care center, and positively related to number<br />

of siblings and living on a farm. Repeated contacts<br />

with certain infectious organisms and endotoxins<br />

decrease the incidence of atopic disease. Infant<br />

distress and colic are now recognized as manifestations<br />

of food hypersensitivity during early childhood.<br />

Histology of the duodenal, gastric and<br />

esophageal mucosa reveals eosinophilic infiltration,<br />

characteristic of a Th2 type and thus allergic<br />

response, in many although not all infants.<br />

Infants with GERD learn to associate eating with<br />

discomfort and thus subsequently tend to avoid<br />

eating, although behavioral feeding difficulties are<br />

common even in control toddlers. 11 In adults,<br />

impaired quality of life, notably regarding pain,<br />

mental health and social function, has been<br />

demonstrated in patients with GERD, regardless of<br />

the presence of esophagitis. 17 In an unselected<br />

population, 28% of the adults reported heartburn,<br />

almost half of them weekly, with a significant<br />

impact on the quality of life in 76%, especially if<br />

the symptoms were frequent and long-lasting.<br />

Despite that, only half of heartburn complainers<br />

sought medical help, although 60% were taking<br />

medications. 73 Thus, some adults ‘learn to live<br />

with their symptoms’, and acquire tolerance to<br />

long-lasting symptoms, while others accept living<br />

with an impaired quality of life.<br />

The reason for the differences in presentation of<br />

GERD according to age remains unclear. The<br />

persistence of symptoms and progression to<br />

complications are unpredictable for a group of<br />

patients and for an individual patient. Alarm<br />

symptoms are similar in adults and children:<br />

weight loss, dysphagia, bleeding, anemia, chest<br />

pain and choking. 16,69 Additional alarm symptoms<br />

in children are failure to thrive, irritability/crying<br />

and feeding or sleeping difficulties. 74<br />

Gastroesophageal reflux disease and<br />

respiratory disease<br />

Reflux may cause respiratory symptoms through<br />

different pathways, such as (micro-) aspiration or<br />

vagally mediated mechanisms. Many patients with


46<br />

Gastroesophageal reflux disease<br />

chronic cough have gastrohypopharyngeal<br />

reflux. 35 A consequence of pulmonary aspiration<br />

of refluxed material may be the presence of an<br />

increased number of lipid-laden macrophages.<br />

Although simply observing the presence of lipidladen<br />

alveolar macrophages is likely to be nonspecific,<br />

it has been suggested that quantification<br />

would be a useful marker of silent aspiration. 75<br />

Data are lacking and are therefore needed on the<br />

diagnostic accuracy, sensitivity and specificity of<br />

the detection and quantification of other<br />

substances in tracheal aspirates, such as lactose,<br />

pepsin and intrinsic factor.<br />

In some patients it may not be GER that is causing<br />

respiratory disease, but the reverse. Respiratory<br />

difficulties cause greater respiratory breathing<br />

efforts and thus more pronounced negative<br />

intrathoracic pressure, and thus respiratory symptoms<br />

may provoke GER. However, the incidence of<br />

a direct temporal relation between reflux and<br />

cough episodes is relatively low. 76<br />

The relation between respiratory disease and GER<br />

may also be neurogenic, in this case designated as<br />

‘gastric asthma’. 77 The tracheobronchial tree and<br />

the esophagus have common embryonic foregut<br />

origins and share autonomic innervation through<br />

the vagus nerve. 78 In other words, it can be speculated<br />

that GER increases the irritability of the vagal<br />

nerve endings in the esophagus, and that as a<br />

result these nerve endings hyper-react together<br />

with the nerve endings in the airways because<br />

they have the same embryonic origin.<br />

Complications<br />

GERD is associated with severe complications<br />

such as esophagitis, Barrett’s esophagus, strictures<br />

and esophageal adenocarcinoma. The severity of<br />

the complications is not clearly related to the duration<br />

or severity of symptoms, as severe histological<br />

changes are detectable at the first investigation.<br />

Differences in esophageal mucosal resistance and<br />

genetic factors may partially explain the diversity<br />

of lesions and symptoms. 16 Esophageal ulcers may<br />

be diagnosed in adults with dysphagia, odynophagia<br />

or esophageal bleeding, but are rarely seen in<br />

children. 20,74<br />

The incidence of congenital esophageal stenosis is<br />

approximately 1 in 25000 to 1 in 50000 live births,<br />

with associated esophageal atresia in one-third of<br />

cases. 79 More than 40 years ago, in the absence of<br />

reflux treatment, esophageal strictures were<br />

reported in about 5% of children with reflux symptoms.<br />

80 Nowadays, except in Barrett’s esophagus,<br />

esophageal stenosis and ulceration in children<br />

have become rare. 20 Esophageal stenosis may be<br />

related to the initial severity of the esophagitis and<br />

the persistence of symptoms, even during treatment.<br />

16 Occasionally, esophageal stenosis is<br />

reported to have developed after an intervention<br />

for achalasia. 81<br />

Reflux esophagitis is reported in 2–5% of the<br />

general population. 82 Children with GER symptoms<br />

present esophagitis in 15–62%, Barrett’s esophagus<br />

in 1.5–3% and refractory GERD requiring surgery in<br />

6–13%. 20–24 In adults undergoing endoscopy,<br />

esophagitis is diagnosed in 15–80%. 1,17,25,26 The<br />

differences in incidence are determined by patient<br />

recruitment and availability of acid-blocking drugs<br />

over the counter (self-treatment). A 10-year followup<br />

of esophagitis showed that over 70% had<br />

persisting symptoms, and 2% had strictures. 19<br />

Thus, esophagitis is not a necessary prerequisite<br />

for diagnosing GERD or starting therapy, either in<br />

children or in adults.<br />

Abundant infiltration of the esophageal mucosa<br />

with eosinophils, as occurs in eosinophilic<br />

gastroenteritis and eosinophilic esophagitis, is<br />

increasing in prevalence and necessitates proper<br />

treatment (hypoallergenic feeding, corticoids, etc.).<br />

Patients with allergic esophagitis seem to have a<br />

younger age and common atopic features (allergic<br />

symptoms or positive allergy tests), but no specific<br />

symptoms. In children, eosinophilic esophagitis<br />

accounts for almost 1.0% of esophagitis in some<br />

selected series. 83 Atopic features are reported in<br />

more than 90% and peripheral eosinophilia in<br />

50% of patients. At endoscopy, a pale, granular,<br />

furrowed and occasionally ringed esophageal<br />

mucosa may appear. 84 In reflux esophagitis, the<br />

distal and lower eosinophilic infiltrate is limited to<br />

less than 5 per high-power field (HPF) with 85%<br />

positive response to GER treatment, compared to<br />

primary eosinophilic esophagitis with >20<br />

eosinophils per HPF. 83–85 In adults, allergic<br />

esophagitis and eosinophilic esophagitis are rarely<br />

found, suggesting an age-related regression or a<br />

possible underinvestigation. Patients with primary<br />

eosinophilic esophagitis may respond to dietary


elimination, cromolyn sodium or steroids. 84<br />

Recently, montelukast and fluticasone have also<br />

been reported to be of benefit. 86,87<br />

Barrett’s esophagus is a premalignant condition<br />

with metaplastic columnar epithelium with goblet<br />

cells in the esophagus, 88 detectable in up to 5–10%<br />

of the endoscopies performed in adults, 16 but<br />

rarely seen in children. Children with severe<br />

reflux, as in those with neurological impairment,<br />

chronic lung disease (especially cystic fibrosis)<br />

and esophageal atresia, are at a higher risk of<br />

developing Barrett’s esophagus. 24 In addition to<br />

severe reflux, a genetic component is also<br />

suggested. 20,24,88–91 In a series including 402 children<br />

with GERD without neurological or congenital<br />

anomalies, no case of Barrett’s esophagus was<br />

detected. 20 In another series including 103 children<br />

with long-lasting GERD, and not previously<br />

treated with H2-receptor antagonists or a proton<br />

pump inhibitor, Barrett’s esophagus was detected<br />

in 13%. An esophageal stricture was present in<br />

five of the 13 patients with Barrett’s esophagus<br />

(38%). 90 Reflux symptoms during childhood were<br />

not different in the adults without, than in the<br />

adults with, Barrett’s esophagus. 88<br />

Peptic ulcer, esophageal and gastric neoplastic<br />

changes in children are extremely rare. In adults,<br />

over the past 30 years, a decreased prevalence of<br />

gastric cancer and peptic ulcer with an opposite<br />

increase of esophageal adenocarcinoma and GERD<br />

have been noted. 92 This has been attributed to<br />

independent factors amongst which are changes in<br />

dietary habits such as a higher fat intake, an<br />

increased incidence of obesity and a decreased<br />

incidence of <strong>Helicobacter</strong> <strong>pylori</strong> infection. 17,92<br />

Nevertheless, the frequency, severity and duration<br />

of reflux symptoms are related to the risk of developing<br />

esophageal cancer. Among adults with longstanding<br />

and severe reflux the odds ratios are 43.5<br />

for esophageal adenocarcinoma and 4.4 for adenocarcinoma<br />

at the cardia. 93 It is unknown whether<br />

mild esophagitis or GER symptoms persisting from<br />

childhood are related to an increased risk for<br />

severe complications in adults.<br />

Diagnosis with differential<br />

GERD is a primary motility disorder, mainly<br />

caused by reflux of gastric content during TLESRs<br />

inducing symptoms. None of the symptoms asso-<br />

Pathophysiology 47<br />

ciated with GER and GERD are specific. Reflux<br />

disease can be a primary condition, or it can be the<br />

consequence of other abnormalities (such as<br />

neurological impairment, cystic fibrosis, <strong>pylori</strong>c<br />

hypertrophy) favoring GER. The long list of differential<br />

diagnoses is discussed under the other headings<br />

of this chapter, and depends on the age of the<br />

patient and the presenting symptom.<br />

Because of lack of space, diagnostic procedures are<br />

not discussed in full detail. Detailed information<br />

regarding the techniques, indications and pitfalls<br />

of radiologic contrast studies, reflux scintiscanning,<br />

ultrasound, pH metry, endoscopy and<br />

manometry can be found in other textbooks or<br />

review papers. Interest will be focused on recent<br />

developments such as impedancometry. The<br />

development of a validated ‘infant GERD questionnaire’<br />

is likely to be the development in diagnostic<br />

accuracy with the greatest impact.<br />

Radiological contrast studies, scintiscanning and<br />

ultrasound are techniques evaluating postprandial<br />

reflux, and provide some information on gastric<br />

emptying. Normal ranges are not well established<br />

for any of the three procedures. Regarding gastric<br />

emptying, 13-C breath tests are more standardized<br />

(but the role of delayed gastric emptying in GER(D)<br />

is controversial). Scintiscanning may show<br />

pulmonary aspiration, although the sensitivity of<br />

the technique is extremely low. Contrast radiology<br />

is of importance to rule out anatomic abnormalities<br />

such as malrotation, duodenal web, stenosis<br />

and achalasia.<br />

There are no age or weight limitations in performing<br />

endoscopy. Endoscopy shows anatomic<br />

malformations and esophagitis, not reflux.<br />

Endoscopy-negative reflux disease is common.<br />

Biopsies of duodenal, gastric and esophageal<br />

mucosa are mandatory to exclude eosinophilic<br />

infiltration.<br />

Ambulatory 24-h esophageal pH monitoring<br />

measures the incidence and duration of acid<br />

reflux, and should be considered the ‘silver standard’.<br />

It is the best method to measure the presence<br />

of acid in the esophagus, but not all reflux<br />

causing symptoms is acid. However, it is likely<br />

that the majority of GERD patients suffer acid<br />

reflux. Esophageal pH metry is useful in evaluating<br />

the effect of any therapeutic intervention on<br />

reducing esophageal acid exposure. Since medical


48<br />

Gastroesophageal reflux disease<br />

treatment currently focuses on reducing gastric<br />

acid secretion, the technique offers the possibility<br />

of measuring intragastric and esophageal recording<br />

of pH simultaneously. Manometry does not<br />

demonstrate reflux, but is of interest in showing<br />

the pathophysiological mechanisms causing the<br />

reflux (by measuring the frequency and duration<br />

of TLESRs), and is indicated in specific situations<br />

such as achalasia. Ambulatory 24-h esophageal<br />

manometry, in combination with pH metry, is now<br />

technically feasible. Long-lasting investigations<br />

offer the opportunity of measuring events in<br />

upright and recumbent positions, awake and<br />

asleep. Results of pH monitoring depend on the<br />

hardware and the software used. 94 The correlation<br />

between results obtained with different types of<br />

electrodes, glass and antimony, is extremely<br />

poor. 95<br />

Impedancometry is a technique gaining more and<br />

more interest, although it has existed for many<br />

years. The technique measures electrical potential<br />

differences, and is therefore not pH-dependent.<br />

The technique offers the possibility of distinguishing<br />

between acid and non-acid reflux (in combination<br />

with pH metry), and between liquid and gas<br />

reflux. Interpretation of the recording is still quite<br />

laborious and necessitates sufficient experience.<br />

(This is of course true for any of the investigation<br />

techniques.) Impedancometry seems especially of<br />

interest in patients with endoscopic and pH<br />

metric-negative symptoms suggesting GERD.<br />

Because reflux in infants is common, because<br />

there is no ‘gold standard’ investigation, and<br />

because investigations are invasive and expensive,<br />

interest has focused on the development of an<br />

‘infant GER-questionnaire’. 7 Recently, an improved<br />

questionnaire was developed. 96 The questionnaire<br />

offers the advantage of an objective, validated and<br />

repeatable quantification of symptoms suggesting<br />

GERD, and thus offers the possibility of measuring<br />

the impact of therapeutic intervention. However,<br />

although the correlation between the questionnaire<br />

and symptoms seems fair, the correlation<br />

between the questionnaire and results of investigations<br />

for reflux is poor.<br />

Investigation methods for GER all test different<br />

aspects involved in the mechanisms and characteristics<br />

of reflux. Therefore, it is not unexpected<br />

that the correlation between different techniques<br />

is extremely poor; non-acid reflux can cause<br />

esophagitis, severe heartburn can exist without<br />

esophagitis, etc. Also, the correlation between<br />

questionnaires for GER symptoms and results of<br />

pH metry and endoscopy is quite poor.<br />

Treatment options<br />

Because symptoms suggesting GERD are frequent<br />

and non-specific, especially during infancy, and<br />

because there is no ‘gold-standard’ diagnostic technique,<br />

many infants are exposed to anti-reflux<br />

treatment. Therefore, attention in the paragraphs<br />

on therapeutic possibilities is focused on safety<br />

aspects. Therapeutic intervention should always<br />

be a balance between intended improvement of<br />

symptoms and risk for side-effects.<br />

Complications of non-intervention<br />

It is difficult to know the true natural history of<br />

GER in infants and children because most patients<br />

obtain treatment. Knowledge on the natural<br />

history in untreated patients from the initial<br />

studies, when effective treatment was unavailable,<br />

is extremely limited, because of the limited<br />

description and identification of the patients. The<br />

paucity of long-term reports, the presence of multiple<br />

pathogenic factors and the absence of pathognomonic<br />

symptoms for complications make it<br />

currently impossible to predict, on an individual<br />

basis, which child will continue to have GERD into<br />

and during adult life. However, we know that<br />

untreated GERD may be associated with severe<br />

complications such as esophagitis, failure to thrive<br />

in children, esophageal stricture and Barrett’s<br />

esophagus.<br />

Recent observations suggest a decreased quality of<br />

life in regurgitating infants and their parents, even<br />

if the regurgitation has disappeared. A 10-year<br />

follow-up of esophagitis showed that over 70%<br />

had persisting symptoms, and 2% had strictures. 19<br />

Untreated or uncontrolled GERD is associated with<br />

severe complications such as esophagitis, Barrett’s<br />

mucosa, stricture formation and esophageal<br />

adenocarcinoma. The frequency, severity and<br />

duration of reflux symptoms are related to the risk<br />

of esophageal cancer. It is not known whether mild


esophagitis or GERD symptoms persisting from<br />

childhood into adulthood carry an increased risk<br />

for severe complications in adult life. Spontaneous<br />

improvement and healing of non-ulcerated<br />

esophagitis may exist. Nevertheless, complications<br />

and side-effects of medication have to be considered<br />

in relation to the natural evolution of<br />

untreated GERD.<br />

Non-pharmacological and non-surgical<br />

therapies for gastroesophageal reflux<br />

Non-pharmacological (and non-surgical) therapies<br />

for reflux do not have any proven efficacy on<br />

reflux, although some may decrease the incidence<br />

of regurgitation. Lifestyle changes (in adults) are<br />

rarely beneficial. 96 No significant difference was<br />

shown between the flat and head-elevated prone<br />

position. Despite gravity, the upright seated position<br />

leads to significantly more and larger reflux<br />

episodes than the simple prone and 30° elevated<br />

prone position, when the infant is awake or<br />

asleep. 97 This is likely to be due to increased<br />

abdominal or intragastric pressure. The supine<br />

(lying on the back) and lateral positions (lying on<br />

the left or right side) usually result in intermediate<br />

pH-metric GER values and do not appear to influence<br />

GER. 97,98 However, there is now ample<br />

evidence that the prone sleeping position is a risk<br />

factor in sudden infant death, independent of<br />

overheating, adult’s smoking or way of feeding.<br />

The impact of pacifier (‘dummy’) use on reflux<br />

frequency was equivocal and dependent on infant<br />

position. The protein content of formula was not<br />

found to affect reflux. Another study suggested<br />

that the lower the osmolality, the less acid reflux.<br />

Larger food volume and higher osmolality increase<br />

the rate of transient LES relaxations and drifts in<br />

LES pressure; a reduction of the food volume<br />

results in a decrease in the number of regurgitations<br />

but no change in acid reflux. 99<br />

The data of ten randomized controlled trials of<br />

non-pharmacological and non-surgical GERD in<br />

healthy infants were recently reviewed. 100<br />

Although no study demonstrated a significant<br />

reflux-reducing benefit of thickened formula<br />

compared to placebo, one study detected a significant<br />

benefit of formula thickened with carob bean<br />

gum compared with rice flour. Milk-thickening<br />

agents include bean gum preparations prepared<br />

Treatment options 49<br />

from St John’s bread, a galactomannan, carboxymethylcellulose,<br />

a combination of pectine and<br />

cellulose, cereals and starch from rice, potato, corn<br />

(maize), etc. There are as many different compositions<br />

of anti-regurgitation formulas as there are<br />

companies: some are casein-predominant, and<br />

others contain protein hydrolysates. Milk thickeners<br />

have been reported to reduce regurgitation in<br />

infants. 97 However, their effects on esophageal<br />

acid exposure are inconsistent. Increased coughing<br />

has also been demonstrated in infants receiving<br />

milk thickeners. 97 According to in vitro models<br />

testing the effect on one meal, bean gum may be<br />

associated with a malabsorption of minerals and<br />

micronutrients. 101 Studies of various thickening<br />

agents, including guar gum, carob bean gum and<br />

soybean polysaccharides, indicate the potential for<br />

decreased intestinal absorption of carbohydrates,<br />

fats, calcium, iron, zinc and copper. 102 Abdominal<br />

pain, colic and diarrhea may ensue from fermentation<br />

of bean gum derivatives in the colon. Carob<br />

bean gum induces frequent, loose, gelatinous<br />

stools. In some, but not all, animal studies, adding<br />

carob bean gum to the diet decreased growth. 102<br />

However, growth and nutritional parameters in<br />

infants receiving a casein-predominant formula<br />

thickened with bean gum were normal. 103<br />

Although rare, serious complications such as acute<br />

intestinal obstruction in newborns have been<br />

reported. 97 Milk thickeners are often wrongly<br />

considered as ‘inexpensive’. Allergic reactions to<br />

carob bean gum have been reported in adults<br />

exposed to it at their workplaces and in infants<br />

after exposure to formula thickened with carob<br />

bean gum. 102 Nevertheless, in view of their safety<br />

and efficacy in decreasing regurgitation, milk<br />

thickeners remain a valuable first-line measure in<br />

relieving regurgitation in many infants. In<br />

contrast, their efficacy in GERD is questionable.<br />

Moreover, they are not devoid of side-effects.<br />

Prokinetics<br />

Prokinetic agents, e.g. metoclopramide, domperidone,<br />

erythromycin and cisapride, act on regurgitation<br />

through their effects on LES pressure,<br />

esophageal peristalsis or clearance and/or gastric<br />

emptying. Metoclopramide and domperidone also<br />

have anti-emetic properties, owing to their<br />

dopamine-receptor blocking action, while


50<br />

Gastroesophageal reflux disease<br />

cisapride is a prokinetic mainly acting via indirect<br />

release of acetylcholine from the myenteric plexus.<br />

Metoclopramide<br />

Data supporting the efficacy of metoclopramide<br />

are contradictory, and positive results are limited<br />

to observations with intravenous administration.<br />

104 Application in infants is limited because of<br />

severe adverse events that occur quite frequently<br />

(in more than 20% of patients) including central<br />

nervous system effects and interactions with the<br />

endocrine system. 104 The adverse effects regarding<br />

the central nervous system are mainly related to its<br />

dopamine-receptor blocking properties in the<br />

substantia nigra, and include extrapyramidal<br />

effects (dystonic reactions, irritability) and drowsiness,<br />

but also asthenia and sleepiness. Isolated<br />

cases of metoclopramide-induced methemoglobinemia<br />

and sulfhemoglobinemia have been<br />

reported. 104,105 Neuroendocrine side-effects such<br />

as galactorrhea do occur. 106 Also, metoclopramide<br />

has been reported to induce torsade de pointes. 107<br />

Domperidone<br />

The studies supporting efficacy of domperidone in<br />

improving GER in infants are limited. 104 The<br />

ability of oral domperidone to increase the pressure<br />

of the LES or to promote healing of reflux<br />

esophagitis has not been demonstrated in placebocontrolled<br />

trials. Most studies have been<br />

performed in older children, or investigate the<br />

effects of domperidone co-administered with other<br />

anti-reflux agents. 104 Comparing domperidone to<br />

metoclopramide, elicited adverse effects on the<br />

central nervous system were more severe and more<br />

common with metoclopramide. 108 Because very<br />

little domperidone crosses the blood–brain barrier,<br />

reports of central nervous system adverse effects,<br />

such as dystonic reactions, are rare. 109<br />

Domperidone is better tolerated than metoclopramide,<br />

since dystonic reactions (tremors) and<br />

anxiety are infrequent. Prolactin plasma levels<br />

may increase, owing to pituary gland stimulation.<br />

110 Somnolence was acknowledged by 49% of<br />

patients after 4 weeks of metoclopramide treatment<br />

compared with 29% of patients after 4 weeks<br />

of domperidone. 108 A reduction in mental acuity<br />

was acknowledged by 33% of patients compared to<br />

20% in the domperidone group. Akathisia, asthenia,<br />

anxiety and depression were also acknowl-<br />

edged less often, and at a lower severity after 4<br />

weeks of domperidone, although these differences<br />

were not significant. Domperidone possesses<br />

cardiac electrophysiological effects similar to<br />

those of cisapride and class III antiarrhythmic<br />

drugs. 104 Intravenously administered domperidone<br />

clearly causes QT prolongation and ventricular<br />

fibrillation. 111,112<br />

Erythromycin<br />

Erythromycin has a prokinetic activity if it is<br />

administered intravenously. Systemic administration<br />

of erythromycin in young infants increases<br />

the risk of the infants developing hypertrophic<br />

<strong>pylori</strong>c stenosis. 113 Similarly, a possible association<br />

exists with maternal macrolide therapy in late<br />

pregnancy. 113 Intravenous erythromycin is<br />

reported to cause QT prolongation and ventricular<br />

fibrillation. 97,114 The use of erythromycin at doses<br />

far below the concentrations necessary for an<br />

inhibitory effect on susceptible bacteria provides<br />

close to ideal conditions for the induction of bacterial<br />

mutation and selection. 115 Emergence of bacteria<br />

increasingly resistant to macrolide antibiotics<br />

has been reported. 116 New erythromycin-like<br />

molecules without the antibiotic properties are in<br />

development.<br />

Cisapride<br />

Critical evaluations of published reports on the<br />

efficacy of different prokinetics (cisapride,<br />

domperidone and metoclopramide) concluded<br />

that cisapride was the preferred agent. 117,118<br />

According to these assessments, the vast majority<br />

of clinical trials on the efficacy of cisapride<br />

demonstrated that at least one of the end-points<br />

changed favorably as a result of the intervention.<br />

117 Cisapride is more effective than metoclopramide.<br />

118 A Cochrane review on cisapride in<br />

children analyzed data from seven trials, including<br />

236 patients; they compared the effect of cisapride<br />

to that of placebo on symptom presence and<br />

improvement. 119 It was concluded that there was a<br />

statistical difference in the parameter symptoms<br />

‘present/absent’ but that there was no statistically<br />

significant difference for ‘symptom change’<br />

between placebo and cisapride. The Cochrane<br />

review also concluded that cisapride compared to<br />

placebo significantly reduced the number and<br />

duration of acid reflux episodes, since there was a


significant decrease in reflux index, which is the<br />

percentage of time that esophageal pH is below<br />

4.0. 119 In general, cisapride is well tolerated. The<br />

most common adverse events at therapeutic doses<br />

are transient diarrhea and colic (in about 2%). The<br />

effect of cisapride on relevant cardiac events such<br />

as QT prolongation and arrhythmia is related to<br />

dose and risk factors. More serious adverse events<br />

such as extrapyramidal reactions, seizures in<br />

epileptic patients, and cholestasis in very premature<br />

infants have been the objects of isolated<br />

reports.<br />

The relation between cisapride, the P450<br />

cytochrome and cardiac effects was considered in<br />

1996. 120 The cytochrome P450 system and especially<br />

CYP3A4 metabolizes cisapride in the<br />

liver. 121 There is little doubt that cisapride has a<br />

QT-prolonging effect, 117 as do many other drugs or<br />

clinical situations. 122 Cisapride possesses class III<br />

antiarrhythmic properties and prolongs the action<br />

potential duration, delaying cardiac repolarization,<br />

123 although many studies do not report an<br />

increase in duration of QTc, in neonates or in older<br />

children. Torsades de pointes have been reported<br />

with cisapride use, especially in those receiving<br />

high doses or macrolides. 124 Co-treatment of<br />

cisapride and macrolides such as clarithromycin<br />

and erythromycin clearly prolongs the QT duration.<br />

125 Underlying cardiac disease, drug interactions<br />

and electrolyte imbalance are clearly interfering<br />

factors. 126 Cisapride causes prolongation of<br />

ventricular repolarization without causing<br />

increased heterogeneity of repolarization (QT<br />

dispersion), but all patients in the study remained<br />

asymptomatic without dysrhythmia. 127 The QTprolonging<br />

effect of cisapride may be related to<br />

age. 128 The cytochrome P4503A4, which is<br />

involved in the metabolism of cisapride, is immature<br />

at birth and reaches adult activity by the age<br />

of 3 months. Cisapride accumulation occurs in<br />

newborns because of enzymatic immaturity. A<br />

significant QTc prolongation occurs, especially in<br />

infants younger than 3 months, but not in older<br />

infants. 129,130 This effect was related to higher<br />

plasma levels. A more frequent administration of<br />

lower doses (resulting in a recommended daily<br />

dose of 0.8mg/kg per day) in premature infants<br />

results in lower peak levels. 131 Consumption of<br />

grapefruit juice also alters cisapride metabolism.<br />

132 According to recent data, gene mutation<br />

may be the fundamental culprit. 133 Molecular<br />

Treatment options 51<br />

screening may allow identification among family<br />

members of gene carriers potentially at risk if<br />

treated with I(Kr) blockers. 134<br />

Other molecules<br />

From the pathophysiological point of view, prokinetics<br />

seems a logical therapeutic approach.<br />

However, efficacy data for the whole group of<br />

prokinetic drugs are disappointing. Cisapride was<br />

shown to have some efficacy in esophageal acidexposure<br />

duration, 135 but it is now banned<br />

because of cardiac side-effects. Prucalopride, a 5-<br />

HT4 agonist, as been suggested has a possible<br />

option, but although the drug seems effective in<br />

adult constipation, 136 its use was prohibited for<br />

children because of the extrapyramidal sideeffects.<br />

Ondansetron is a 5-HT3 receptor antagonist<br />

that accelerates gastric emptying, inhibits<br />

chemotherapy-induced emesis, but prolongs<br />

colonic transit time. 137 The most frequently<br />

reported adverse events of ondansetron were mild<br />

to moderate headache, constipation and diarrhea<br />

in patients receiving chemotherapy. Tegaserod is a<br />

partial 5-HT4 agonist that has been mostly studied<br />

in constipation-predominant irritable bowel<br />

syndrome in adults. 138 Tegaserod was shown to<br />

accelerate small intestinal transit time, and to<br />

increase proximal colonic emptying. Tegaserod<br />

also improves gastric emptying and decreases<br />

GER, 139 and may be a promising drug. However, to<br />

date there are no efficacy data published on the<br />

treatment of pediatric GER. Baclofen, 4-amino-3-(chlorophenyl)-butanoic<br />

acid, is a γ-aminobutyric<br />

acid (GABA)-B receptor agonist, which is used in<br />

children with extreme spasticity. Given orally, it<br />

has been shown to decrease GER in healthy<br />

adults. 140 Administration in eight pediatric<br />

patients was reported to be safe. 141 Alosetron is a<br />

5-HT3 antagonist that increases colonic compliance<br />

to distension, delays (ascending) colonic<br />

transit time and increases basal fluid absorption.<br />

Alosetron induces severe constipation and causes<br />

ischemic colitis.<br />

(Alginate-) antacids<br />

Experience with antacids is limited in infants.<br />

Their efficacy in buffering gastric acidity in infants<br />

is strongly influenced by the time of administration,<br />

and requires multiple administrations.


52<br />

Gastroesophageal reflux disease<br />

Alginate-antacids form a viscous fluid with<br />

surface-active properties, floating as a raft on the<br />

surface of the gastric contents, and hence forming<br />

an artificial protective barrier against reflux. 142<br />

Their efficacy as monotherapy or in combination<br />

with prokinetics for reflux is not convincing.<br />

Important drug interactions with antacids include<br />

the prevention of the absorption of antibacterials<br />

such as tetracycline, azithromycin and quinolones.<br />

143 Antacid ingestion decreased the bioavailability<br />

of famotidione, ranitidine and cimetidine<br />

by 20–25%, and the bioavailability of nizatidine by<br />

12%. 144 Gaviscon ® contains a considerable<br />

amount of sodium carbonate, so that its administration<br />

may increase the sodium content of the<br />

feeds to an undesirable degree (1g of Gaviscon<br />

contains 46mg of sodium and the suspension<br />

contains twice this amount). Algicon ® , having a<br />

better taste than Gaviscon, has a lower sodium<br />

load, but a higher aluminium content. 142<br />

Occasional formation of large bezoar-like masses<br />

of agglutinated intragastric material has been<br />

reported in association with Gaviscon. Side-effects<br />

include diarrhea with magnesium-rich preparations,<br />

and excessive absorption of aluminium in<br />

infants. 142 Dimethicone is used in some regions for<br />

regurgitation, although there are no reliable<br />

studies demonstrating its efficacy in the treatment<br />

of GER in infants. Although often classified as an<br />

antacid, it acts more as a feed thickener, as it<br />

contains more than 50% of bean gum and has<br />

hardly any acid-neutralizing properties.<br />

H2-receptor antagonists<br />

Acid-suppressant therapy is recommended in<br />

severe esophagitis, but this does not rectify<br />

primary disordered motility, a major pathophysiological<br />

mechanism underlying GERD in children.<br />

Historically, cimetidine was the first H2RA that<br />

became available. Ranitidine and nizatidine are<br />

the most popular and best studied (although quite<br />

poorly) H2RAs in children. 145,146 Experience with<br />

other H2RAs such as roxatidine or ebrotidine is<br />

very limited or non-existent in children. In<br />

general, H2RAs are considered to be quite safe.<br />

Adverse events reported in clinical trials with ranitidine<br />

include headache, tiredness and mild<br />

gastrointestinal disturbances, but the incidence is<br />

not higher than that for placebo. 146 A high dose of<br />

cimetidine can cause reversible impotence and<br />

gynecomastia. 146 The endocrinological side-effects<br />

associated with long-term administration of cimetidine<br />

in adults essentially preclude its long-term<br />

use in children. 147 Fatigue, dizziness, headache,<br />

dyspepsia, nausea, abdominal pain, flatulence,<br />

constipation and diarrhea occur in 1–6% of<br />

patients. 148 H2RAs have been reported to provoke<br />

central nervous system dysfunction, but these are<br />

poorly documented in children. Ranitidine<br />

enhances ischemic neuronal damage. 148 Ranitidine<br />

has occasionally been associated with acute<br />

interstitial nephritis in native and transplanted<br />

kidneys. 149 While ranitidine exhibits no clinically<br />

significant drug–drug interactions, cimetidine<br />

interacts with many drugs metabolized by<br />

cytochrome P450. Theophylline plasma levels<br />

were 25–32% higher if cimetidine was administered,<br />

compared to ranitidine. Neither ranitidine<br />

nor nizatidine increased theophylline levels. 144,150<br />

H2RAs, PPIs and prokinetic agents undergo metabolism<br />

by the cytochrome P450 system present in<br />

the liver and gastrointestinal tract. 143 Cimetidine is<br />

an inhibitor of CYP3A and it may cause significant<br />

interactions with drugs of narrow therapeutic<br />

range and low bioavailability that are metabolized<br />

by those enzymes. 143<br />

Whether ranitidine may exceptionally cause QT<br />

prolongation or not, is still debated. 151 If ranitidine<br />

is administered intravenously after autonomic<br />

blockade, the sinus cycle length is prolonged, and<br />

the systolic and diastolic blood pressures are<br />

decreased. 152 Thus, ranitidine has to be administered<br />

by a slow intravenous infusion in patients<br />

with sinus node dysfunction. 152 The altered<br />

cardiac sympathovagal balance after oral administration<br />

of the H2RA ranitidine indicates a shift<br />

towards sympathetic predominance in the heart<br />

rate control. 153 Ranitidine modulates highfrequency<br />

power of heart rate, and this may be the<br />

underlying mechanism of cardiovascular sideeffects.<br />

154 Since H2 receptors are present in the<br />

stomach and the heart, they have a trend towards<br />

decreasing the heart rate and cardiac contractility.<br />

Nizatidine and ranitidine are susceptible to metabolism<br />

by colonic bacteria, but famotidine and<br />

cimetidine are not (see also the section on PPIs,<br />

below). 155 Ranitidine alters the gastrointestinal<br />

flora 156 and causes significantly more pneumonias<br />

in patients in intensive care units. 157 In the majority<br />

of patients on H2RAs, there is a relatively


important nocturnal breakthrough of acid secretion,<br />

sometimes limiting therapeutic efficacy, but<br />

on the other hand minimizing side-effects related<br />

to long-term blockade of acid secretion as with<br />

PPIs. There is a rapid development of tachyphylaxis<br />

or tolerance to H2RAs, limiting their longterm<br />

clinical use. 158<br />

The combination of ranitidine and pirenzipine, a<br />

muscarinic receptor antagonist, does not aid the<br />

healing of reflux esophagitis, but does improve<br />

symptom relief after 4 weeks. 159 However, sideeffects<br />

were reported in nine of 75 patients in the<br />

ranitidine group and 19 of 76 patients in the ranitidine<br />

and pirenpizine group. 159<br />

Proton pump inhibitors<br />

The suppression of gastric acid secretion achieved<br />

with H2RAs has, however, proved to be suboptimal.<br />

158 In this regard, the advent of the PPIs has<br />

been a major breakthrough. Drugs of this category<br />

have in fact been shown to be more effective than<br />

H2RAs. 96 Esomeprazole seems to be the most effective<br />

PPI commercialized today. 160 Step-down treatment<br />

is recommended in adults. 161 Failure to<br />

control symptoms with high-dose PPI treatment<br />

raises the likelihood of non-acid-related causes for<br />

the symptoms.<br />

The pharmacokinetics and tolerance of pantoprazole<br />

were similar in patients with moderate and<br />

severe hepatic impairment. 162 These were also<br />

evaluated for famotidine in 150 children. 163 Recent<br />

studies with lansoprazole showed its efficacy in<br />

reducing symptoms and healing esophagitis in<br />

children. 164 The availability of a syrup facilitates<br />

the use of this drug in children. 164 Zimmermann<br />

and colleagues reviewed the literature on the use<br />

and administration of omeprazole in children. 165<br />

In uncontrolled trials and case reports, omeprazole<br />

was used at a dosage of 0.2–3.5mg/kg per day for<br />

periods ranging from 14 days to 36 months, and<br />

found to be effective and well tolerated for the<br />

acute and chronic treatment of esophageal and<br />

peptic ulcer disease in children aged 2 months to<br />

18 years. 165<br />

The following side-effects of PPI have been<br />

reported: headache (about 3%); neurological and<br />

psychiatric side-effects, especially fatigue, dizziness<br />

and confusion in patients with hepatic<br />

Treatment options 53<br />

diseases and/or advanced age; cutaneous reactions,<br />

generally rash and urticaria; hemolytic<br />

anemia, leukopenia and agranulocytosis; gynecomastia;<br />

subacute myopathy; gastrointestinal sideeffects<br />

such as flatulence, constipation, diarrhea<br />

(about 4%), dyspepsia and nausea (about 2%),<br />

vomiting and abdominal pain; hepatic disorders,<br />

especially moderate elevation of aminotransferases;<br />

and excessive urinary sodium<br />

loss. 97,142,166,167 The high cost of PPIs can also be<br />

considered an important albeit not medical ‘sideeffect’.<br />

Omeprazole, esomeprazole, lansoprazole, rabeprazole<br />

and pantaprazole rarely exhibit clinically<br />

important interactions with other hepatically<br />

metabolized medications or pH-dependent<br />

drugs. 168 The absorption of drugs that are pH<br />

sensitive, as is the case with digoxin and ketoconazole,<br />

will be influenced by PPIs. 169<br />

The gastroparietal PPIs lansoprazole, omeprazole<br />

and pantoprazole are all primarily metabolized by<br />

a genetically polymorphic enzyme, CYP2C19, that<br />

is absent in approximately 3% of Caucasians and<br />

20% of Asians. 143 These drugs may also interact<br />

with CYP3A, but to a lesser extent. Esomeprazole<br />

has the potential to interact with CYP2C19. The<br />

slightly altered metabolism of cisapride was also<br />

suggested to be the result of inhibition of a minor<br />

metabolic pathway for cisapride mediated by<br />

CYP2C19. Esomeprazole did not interact with the<br />

CYP3A4 substrates clarithromycin and quinidine.<br />

169 Overall, the potential for drug–drug interactions<br />

with esomeprazole is low, and similar to<br />

that reported for omeprazole. 169 Salivary secretion<br />

is decreased with omeprazole. Prolonged use of<br />

PPIs can result in vitamin B12 deficiency as a<br />

consequence of impaired release of vitamin B12<br />

from food in a non-acid environment. However,<br />

cystic fibrosis patients treated for at least 2 years<br />

with a PPIs and cystic fibrosis patients without PPI<br />

had higher vitamin B12 levels than healthy<br />

controls. 170<br />

The safety of long-term administration of acidblocking<br />

medication needs to be considered in<br />

relation to potential consequences of prolonged<br />

acid suppression, including the risk of proliferation<br />

of gastric flora and the risk of developing enterochromaffin-like<br />

cell hyperplasia, which could,<br />

in turn, theoretically, lead to gastric malignancy.<br />

Hypergastrinemia occurs in nearly all patients


54<br />

Gastroesophageal reflux disease<br />

treated with omeprazole, causing hyperplasia and<br />

pseudohypertrophy of the parietal cells, as recently<br />

shown in 93% of adult patients on long-term<br />

omeprazole. Patients on omeprazole therapy for<br />

5–8 years remained without evidence of significant<br />

enterochromaffin cell hyperplasia, gastric<br />

atrophy, intestinal metaplasia, dysplasia or<br />

neoplastic changes. 171 Because of the excellent<br />

efficacy profile, these drugs tend to be<br />

overused. 172 Since PPIs could delay the diagnosis<br />

of gastric cancer, the long-term uncontrolled and<br />

unnecessary use of these drugs should be avoided.<br />

Bacterial proliferation in the gastric content may<br />

not only change colonic flora, but may also be a<br />

risk factor for the patient to develop nosocomial<br />

pneumonia.<br />

Other drugs<br />

Sucralfate causes bezoars, especially when given<br />

to patients in intensive care units, and diarrhea.<br />

Patients with renal failure treated with sucralfate<br />

are exposed to aluminium toxicity. 173–175 Other<br />

anti-emetic drugs such as batanopride also have a<br />

QTc-prolonging effect. 176 Octeotride is a longacting<br />

somatostatin, that induces a phase 3-like<br />

migrating motor complex response, which is not<br />

inhibited by meals (differing from normal), and<br />

has an inhibitory effect on the gastric antrum (and<br />

is therefore indicated in dumping syndrome).<br />

Motilin agonists have been studied with inconsistent<br />

results in adults, and are not yet available for<br />

pediatric use. 177<br />

Therapeutic endoscopic procedures<br />

During recent years, new endoscopic techniques<br />

intending to improve the function of the antireflux<br />

barrier have been developed. The first<br />

results of endoscopic gastroplasty (Endocinch ®<br />

system), radiofrequency delivery at the cardia<br />

(Stretta ® system) and injection therapy (Enteryx ®<br />

procedure) in adults have been reported. 178–181<br />

The first series in adolescents have been<br />

performed. Although experience is too limited to<br />

recommend broad use, the theoretical concept of<br />

these procedures is interesting. Further improvements<br />

to the techniques are still being introduced.<br />

Surgery<br />

While anti-reflux surgery in certain groups of children<br />

may be of considerable benefit, it also has a<br />

mortality and a failure rate. 184–187 Ninety per cent<br />

of patients remained free from significant reflux<br />

symptoms after a laparoscopic Nissen operation,<br />

although side-effects occurred in up to 22%. 186<br />

After a median follow-up of 16 years, the<br />

Nissen–Rosetti procedure in 24 consecutive children<br />

without congenital or acquired anomalies of<br />

the esophagus except GERD showed a result that<br />

was considered excellent in only 75%, good in 21%<br />

and poor in 4%. 187 Failure rates of 5–20% have been<br />

found after objective postoperative follow-up. 187 A<br />

protective anti-reflux surgical procedure in neurologically<br />

impaired children needing a gastrostomy<br />

increased the morbidity and mortality rate of the<br />

gastrostomy procedure itself. 188<br />

Conclusion<br />

GER and GERD are frequent conditions in infants,<br />

children and adolescents. Symptomatology differs<br />

with age, although the main pathophysiological<br />

mechanism, transient relaxations of the LES associated<br />

with reflux, is identical at all ages. Although<br />

infant regurgitation is likely to disappear with age,<br />

little is known about reflux. The majority of symptomatic<br />

reflux episodes are acid, but non-acid and gas<br />

reflux can also cause symptoms. Complications of<br />

reflux disease may be severe and even life threatening,<br />

such as esophageal stenosis and Barrett’s esophagus.<br />

There is no gold standard for a diagnostic technique.<br />

A simple questionnaire may be among the<br />

best diagnostic aids in infants; non-acid reflux is<br />

best investigated with impedancometry. Primary<br />

GERD is mainly a motility disorder. Guidelines for<br />

treatment struggle with the fact that there is no<br />

prokinetic drug with a convincing efficacy profile.<br />

As a consequence, treatment of GERD focuses on<br />

anti-acid drugs, and particularly on PPIs. There is<br />

little or no information on how to organize follow-up.


REFERENCES<br />

1. de Caestecker J. Oesophagus: heartburn. BMJ 2001; 323:<br />

736–739.<br />

2. Dent J, Brun J, Fendrick AM. An evidence-based<br />

appraisal of the reflux disease management – The<br />

Genval Workshop Report. Gut 1999; 44: S1–S16.<br />

3. Rudolph CD, Mazur LJ, Liptak GS et al. Guidelines for<br />

evaluation and treatment of gastroesophageal reflux in<br />

infants and children. Recommendations of the North<br />

American Society for Pediatric Gastroenterology and<br />

Nutrition. J Pediatr Gastroenterol Nutr 2001; 32(Suppl<br />

2): S1–S31.<br />

4. Vandenplas Y. Oesophageal pH Monitoring for Gastrooesophageal<br />

Reflux in Infants and Children. Chichester,<br />

UK: John Wiley and Sons, 1992: 27–36.<br />

5. Cleveland RH, Kushner DC, Schwartz AN.<br />

Gastroesophageal reflux in children: results of a standardized<br />

fluoroscopic approach. Am J Roentgenol 1983;<br />

141: 53–56.<br />

6. Vandenplas Y, Goyvaerts H, Helven R.<br />

Gastroesophageal reflux, as measured by 24-hour pHmonitoring,<br />

in 509 healthy infants screened for risk of<br />

sudden infant death syndrome. Pediatrics 1991; 88:<br />

834–840.<br />

7. Orenstein SR, Shalaby TM, Cohn J. Reflux symptoms in<br />

100 normal infants: diagnostic validity of the Infant<br />

Gastroesophageal Reflux Questionnaire. Clin Pediatr<br />

1996; 35: 607–614.<br />

8. Chouhou D, Rossignol C, Bernard F, Dupont C. Le reflux<br />

gastro-oesophagien dans les centres de bilan de santé de<br />

l’enfant de moins de 4 ans. Arch Franc Ped 1992; 49:<br />

843–845.<br />

9. Nelson SP, Chen EH, Syniar GM. Prevalence of symptoms<br />

of gastroesophageal reflux in infancy. Arch Pediatr<br />

Adolesc Med 1997; 151: 569–572.<br />

10. Shepherd R, Wren J, Evans S. Gastroesophageal reflux<br />

in children. Clinical profile, course and outcome with<br />

active therapy in 126 cases. Clin Pediatr 1987; 26:<br />

55–60.<br />

11. Nelson SP, Chen EH, Syniar GM. One year follow-up of<br />

symptoms of gastroesophageal reflux during infancy.<br />

Pediatrics 1998; 102: e67.<br />

12. Wienbeck M, Barnert J. Epidemiology of reflux disease<br />

and reflux esophagitis. Scand J Gastroenterol 1989; 24:<br />

7–13.<br />

13. Nelson SP, Chen EH, Syniar GM, Christoffel KK.<br />

Prevalence of symptoms of gastroesophageal reflux<br />

during childhood. Arch Pediatr Adolesc Med 2000; 154:<br />

150–154.<br />

14. Sonnenberg A, El-Serag HB. Clinical epidemiology and<br />

natural history of gastroesophageal reflux disease. Yale J<br />

Biol Med 1999; 72: 81–92.<br />

15. Isolauri J, Laippala P. Prevalence of symptoms suggestive<br />

of gastro-oesophageal reflux disease in an adult<br />

population. Ann Med 1995; 27: 67–70.<br />

16. The Jury of the Consensus Conference. French–Belgian<br />

Consensus Conference on adult gastro-oesophageal<br />

reflux disease ‘Diagnosis and Treatment’: report of a<br />

meeting held in Paris, France, on 21–22 January 1999.<br />

Eur J Gastroenterol Hepatol 2000; 12: 129–137.<br />

17. Nandurkar S, Talley NJ. Epidemiology and natural<br />

history of reflux disease. Baillière’s Clin Gastroenterol<br />

2000; 14: 743–757.<br />

18. Isolauri J, Luostarinen M, Isolauri E. Natural course of<br />

gastroesophageal reflux disease: 17–22 year follow-up of<br />

60 patients. Am J Gastroenterol 1997; 92: 37–41.<br />

19. McDougall NJ, Johnston BT, Kee F. Natural history of<br />

reflux oesophagitis: a 10 year follow up of its effect on<br />

References 55<br />

patient symptomatology and quality of life. Gut 1996;<br />

38: 481–486.<br />

20. El-Serag HB, Bailey NR, Gilger M, Rabeneck L.<br />

Endoscopic manifestations of gastroesophageal reflux<br />

disease in patients between 18 months and 25 years<br />

without neurological deficits. Am J Gastroenterol 2002;<br />

97: 1635–1639.<br />

21. Lee WS, Beattie RM, Meadows N, Walker-Smith JA.<br />

Gastro-oesophageal reflux: clinical profiles and<br />

outcome. J Paediatr Child Health 1999; 35: 568–571.<br />

22. Tolaymat N, Chapman DM. Gastroesophageal reflux<br />

disease in children older than two years of age. West<br />

Virginia Med J 1998; 94: 22–25.<br />

23. Treem W, Davis P, Hyams J. Gastroesophageal reflux in<br />

the older child: presentation, response to treatment, and<br />

long-term follow-up. Clin Pediatr 1991; 30: 435–440.<br />

24. Hassall E. Co-morbidities in childhood Barrett’s esophagus.<br />

J Pediatr Gastroenterol Nutr 1997; 25: 255–260.<br />

25. Voutilainen M, Sipponen P, Macklin JP.<br />

Gastroesophageal reflux disease: prevalence, clinical,<br />

endoscopic and histopathological findings in 1,128<br />

consecutive patients referred for endoscopy due to<br />

dyspeptic and reflux symptoms. Digestion 2000; 61:<br />

6–13.<br />

26. Loof L, Gotell P, Elfberg B. The incidence of reflux<br />

oesophagitis. Scand J Gastroenterol 1993; 28: 113–118.<br />

27. Hegar B, Boediarso A, Firmansyah A, Vandenplas Y.<br />

Incidence of regurgitation and symptoms of gastroesophageal<br />

reflux in Indonesian infants. Acta<br />

Gastroenterol Belg 2003; Suppl:69 (abstr).<br />

28. Kang JY, Ho KY. Different prevalences of reflux<br />

oesophagitis and hiatus hernia among dyspeptic<br />

patients in England and Singapore. Eur J Gastroenterol<br />

Hepatol 1999; 11: 845–850.<br />

29. Kim SL, Hunter JG, Wo JM et al. NSAIDs, aspirin, and<br />

esophageal strictures: are over-the-counter medications<br />

harmful to the esophagus? J Clin Gastroenterol 1999; 29:<br />

32–34.<br />

30. Carre IJ, Johnston BT, Thomas PS, Morrisson PJ.<br />

Familial hiatal hernia in a large five generation family<br />

confirming true autosomal dominant inheritance. Gut<br />

1999; 45: 649–652.<br />

31. Trudgill NJ, Kapur KC, Riley SA. Familial clustering of<br />

reflux symptoms. Am J Gastroenterol 1999; 94:<br />

1172–1178.<br />

32. Cameron AJ, Lagergren J, Henriksson C.<br />

Gastroesophageal reflux disease in monozygotic and<br />

dizygotic twins. Gastroenterology 2002; 122: 55–59.<br />

33. Hu FZ, Preston RA, Post JC. Mapping of a gene for<br />

severe pediatric gastroesophageal reflux to chromosome<br />

13q14. JAMA 2000; 284: 325–334.<br />

34. Orenstein SR, Shalaby TM, Barmada MM, Whitcomb<br />

DC. Genetics of gastroesophageal reflux disease: a<br />

review. J Pediatr Gastroenterol Nutr 2002; 34: 506–510.<br />

35. Vandenplas Y, Hassall E. Mechanisms of gastroesophageal<br />

reflux and gastroesophageal reflux disease. J<br />

Pediatr Gastroenterol 2002; 35: 119–136.<br />

36. Sifrim D, Holloway R, Silny J et al. Acid, non-acid and<br />

gas reflux in patients with gastroesophageal reflux<br />

disease during 24 hr ambulatory pH-impedance recordings.<br />

Gastroenterology 2001; 120: 1588–1598.<br />

37. McNamara D, O’Morain C. Gastro-oesophageal reflux<br />

disease and <strong>Helicobacter</strong> <strong>pylori</strong>: an intricate relation.<br />

Gut 1999; 45(Suppl 1) :S13–S17.<br />

38. Graham DY, Yamaoka Y. Disease-specific <strong>Helicobacter</strong><br />

<strong>pylori</strong> virulence factors: the unfulfilled promise.<br />

<strong>Helicobacter</strong> 2000; 5(Suppl 1): S3–S9.


56<br />

Gastroesophageal reflux disease<br />

39. Wu JC, Sung JJ, Chan FK et al. <strong>Helicobacter</strong> <strong>pylori</strong> infection<br />

is associated with milder gastro-oesophageal reflux<br />

disease. Aliment Pharmacol Ther 2000; 14: 427–432.<br />

40. De Koster E. Adverse events of HP eradication: long<br />

term negative consequences of HP eradication. Acta<br />

Gastroenterol Belg 1998; 61: 350–351.<br />

41. Fallone CA, Barkin AN, Friedman G et al. Is <strong>Helicobacter</strong><br />

<strong>pylori</strong> eradication associated with gastroesophageal<br />

reflux disease? Am J Gastroenterol 2000; 95: 914–920.<br />

42. Van Zanten SJ, Dixon MF, Lee A. The gastric transitional<br />

zones: neglected links between gastroduodenal pathology<br />

and helicobacter ecology. Gastroenterology 1999;<br />

116: 1217–1229.<br />

43. Kuiken S, Van Den Elzen B, Tytgat G et al. Evidence for<br />

pooling of gastric secretions in the proximal stomach in<br />

humans using single photon computed tomography.<br />

Gastroenterology 2002; 123: 2157-2158.<br />

44. Fletcher J, Wirz A, Young J et al. Unbuffered highly<br />

acidic gastric juice exists at the gastroesophageal junction<br />

after a meal. Gastroenterology 2001; 121: 775–783.<br />

45. Dodds WJ, Dent J, Hogan WJ et al. Mechanisms of<br />

gastroesophageal reflux in patients with reflux<br />

esophagitis. N Engl J Med 1982; 307: 1547–1552.<br />

46. Omari TI, Benninga MA, Barnett CP et al.<br />

Characterization of esophageal body and lower<br />

esophageal sphincter motor function in the very premature<br />

neonate. J Pediatr 1999; 135: 517–521.<br />

47. Boulant J, Fioramonti J, Dapoigny M et al.<br />

Cholecystokinin and nitric oxide in transient lower<br />

esophageal sphincter relaxation to gastric distention in<br />

dogs. Gastroenterology 1994; 107: 1059–1066.<br />

48. Hirsch DP, Hollowy RH, Tytgat GNJ, Boeckxstaens GE.<br />

Involvement of nitric oxide in human transient lower<br />

esophageal sphincter relaxations and esophageal<br />

primary peristalsis. Gastroenterology 1998; 115:<br />

1374–1380.<br />

49. Sifrim D, Tack J, Lerut T, Janssens J. Transient lower<br />

esophageal sphincter relaxations and esophageal body<br />

muscular contractile response in reflux esophagitis. Dig<br />

Dis Sci 2000; 45: 1293–1300.<br />

50. Mittal RK, McCallum RW. Characteristics and frequency<br />

of transient relaxations of the lower esophageal sphincter<br />

in patients with reflux esophagitis. Gastroenterology<br />

1988; 95: 593–599.<br />

51. Holloway RH, Kocyan P, Dent J. Provocation of transient<br />

lower esophageal sphincter relaxations by meals in<br />

patients with symptomatic gastroesophageal reflux. Dig<br />

Dis Sci 1991; 36: 1034–1039.<br />

52. Penagini R, Hebbard G, Horowitz M et al. Motor function<br />

of the proximal stomach and visceral perception in<br />

gastro-oesophageal reflux disease. Gut 1998; 42:<br />

251–257.<br />

53. Stacher G, Lenglinger J, Bergmann H. Gastric emptying:<br />

a contributory factor in gastro-oesophageal reflux activity?<br />

Gut 2000; 47: 661–666.<br />

54. Sifrim D, Holloway R. Transient lower esophageal<br />

sphincter relaxations: how many or how harmful? Am J<br />

Gastroenterol 2001; 96: 2529–2532.<br />

55. Vela MF, Camacho-Lobato L, Srinivasan R et al.<br />

Simultaneous intraesopahgeal impedance and pH<br />

measurement of acid and non-acid gastroesophageal<br />

reflux: effect of omeprazole. Gastroenterology 2001; 120:<br />

1599–1606.<br />

56. Niemantsverdriet EC, Timmer R, Breumelhof R, Smout<br />

AJ. The role of excessive gastro-oesophageal reflux,<br />

disordered oesophageal motility and decreased mucosal<br />

sensitivity in the pathogenesis of Barrett’s oesophagus.<br />

Eur J Gastroenterol Hepatol 1997; 9: 515–519.<br />

57. Mathisen B, Worall L, Masel J et al. Feeding problems in<br />

infants with gastro-oesophageal reflux disease: a<br />

controlled study. J Pediatr Child Health 1999; 35:<br />

163–169.<br />

58. Black MM, Dubowitz H, Huctheson J. A randomized<br />

clinical trial of home intervention for children with<br />

failure to thrive. Pediatrics 1995; 95: 807–814.<br />

59. Burklow KA, Phelps AN, Schultz JR et al. Classifying<br />

complex pediatric feeding disorders. J Pediatr<br />

Gastroenterol Nutr 1998; 27: 143–147.<br />

60. Orenstein SR, Cohn JF, Shalaby T. Reliability and validity<br />

of an infant gastroesophageal questionnaire. Clin<br />

Pediatrics 1993; 32: 472–484.<br />

61. Heine RG, Jaquiery A, Lubitz L et al. Role of gastrooesophageal<br />

reflux in infant irritability. Arch Dis Child<br />

1995; 73: 121–125.<br />

62. Snel A, Barnett CP, Cresp TL. Behavior and gastroesophageal<br />

reflux in the premature neonate. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 18–21.<br />

63. Hamilton AB, Zeltzer LK. Visceral pain in infants. J<br />

Pediatr 1994; 125(Suppl): S95–S102.<br />

64. Iacono G, Carroccio A, Cavataio F et al.<br />

Gastroesophageal reflux and cow’s milk allergy in<br />

infants: a prospective study. J All Clin Immunol 1996;<br />

97: 822–827.<br />

65. Salvatore S, Vandenplas Y. Gastroesophageal reflux and<br />

cow’s milk allergy: is there a link? Pediatrics 2002; 110:<br />

972–984.<br />

66. Hill DJ, Heine RG, Cameron DJ et al. Role of food<br />

protein intolerance in infants with persistent distress<br />

attributed to reflux esophagitis. J Pediatr 2000; 136:<br />

641–647.<br />

67. Orenstein SR, Izadnia F, Khan S. Gastroesophageal<br />

reflux disease in children. Gastroenterol Clin North Am<br />

1999; 28: 947–969.<br />

68. Locke GR, Talley NJ, Fett SL. Prevalence and clinical<br />

spectrum of gastroesophageal reflux: a population based<br />

study in Olmsted Country, Minnesota. Gastroenterology<br />

1997; 112: 1448–1456.<br />

69. De Vault KR, Castell DO. Updated guidelines for the<br />

diagnosis and treatment of gastroesophageal reflux<br />

disease. Am J Gastroenterol 1999; 94: 1434–1442.<br />

70. Hyman PE. Gastroesophageal reflux: one reason why<br />

baby won’t eat. J Pediatr 1994; 125: S103–S109.<br />

71. Holt PG. Parasites, atopy, and the hygiene hypothesis:<br />

resolution of a paradox? Lancet 2000; 356: 1699–1701.<br />

72. Van den Biggelaar AHJ, van Ree R, Rodrigues LC et al.<br />

Decreased atopy in children infected with Schistosoma<br />

haematobium: a role for parasite-induced interleukins-<br />

10. Lancet 2000; 356: 1723–1727.<br />

73. Louis E, DeLooze D, Deprez P. Heartburn in Belgium:<br />

prevalence, impact on daily life, and utilization of<br />

medical resources. Eur J Gastroenterol Hepatol 2002; 14:<br />

279–284.<br />

74. Vandenplas Y. Reflux esophagitis in infants and children.<br />

A report from the Working Group of the European<br />

Society of Pediatric Gastroenterology and Nutrition on<br />

Gastro-oesophageal Reflux Disease. J Pediatr<br />

Gastroenterol Nutr 1994; 18: 413–422.<br />

75. Ahrens P, Noll C, Kitz R et al. Lipid-laden alveolar<br />

macrophages: a useful marker of silent aspiration in<br />

children. Pediatr Pulmonol 1999; 28: 83–88.<br />

76. Paterson WG, Murat BW. Combined ambulatory<br />

esophageal manometry and dual-probe pH metry in the<br />

evaluation of patients with chronic unexplained cough.<br />

Dig Dis Sci 1994; 39: 1117–1125.<br />

77. Bruno G, Graf U, Andreozzi P. Gastric asthma: an unrecognized<br />

disease with an unsuspected frequency. J<br />

Asthma 1999; 36: 315–325.<br />

78. Cunningham ET Jr, Ravich WJ, Jones B, Donner MW.<br />

Vagal reflexes referred from the upper aerodigestive<br />

tract: an infrequently recognized cause of common


cardiorespiratory responses. Ann Intern Med 1992; 116:<br />

575–582.<br />

79. Murphy SG, Yazbeck S, Russo P. Isolated congenital<br />

esophageal stenosis. J Pediatr Surg 1995; 30: 1238–1241.<br />

80. Carre I. The natural history of the partial thoracic<br />

stomach (“hiatal hernia”) in children. Arch Dis Child<br />

1959; 34: 344–353.<br />

81. Karnak I, Senocak ME, Tanyel FC, Buyukpamukcu N.<br />

Achalasia in childhood: surgical treatment and<br />

outcome. Eur J Pediatr Surg 2001; 11: 223–229.<br />

82. Wienbeck M, Barnert J. Epidemiology of reflux disease and<br />

reflux esophagitis. Scand J Gastroenterol 1989; 156: 7–13.<br />

83. Liacouras CA, Wenner WJ, Brown K, Ruchelli E.<br />

Primary eosinophilic esophagitis in children: successful<br />

treatment with oral corticosteroids. J Pediatr<br />

Gastroenterol Nutr 1998; 26: 380–385.<br />

84. Orenstein SR, Shalaby TM, Di Lorenzo C. The spectrum<br />

of pediatric eosinophilic esophagitis beyond infancy: a<br />

clinical series of 30 children. Am J Gastroenterol 2000;<br />

95: 1422–1430.<br />

85. Ruchelli E, Wenner W, Voytek T et al. Severity of<br />

esophageal eosinophilia predicts response to conventional<br />

gastroesophageal reflux therapy. Pediatr Dev<br />

Pathol 1999; 2: 15–18.<br />

86. Attwood SE, Lewis CJ, Bronder CS et al. Eosinophilic<br />

oesophagitis: a novel treatment using Montelukast. Gut<br />

2003; 52: 181–185.<br />

87. Teitelbaum JE, Fox VL, Twarog FJ et al. Eosinophilic<br />

esophagitis in children: immunopathological analysis<br />

and response to fluticasone propionate. Gastroenterology<br />

2002; 122: 1216–1225.<br />

88. Hassall E. Barrett’s esophagus: new definitions and<br />

approaches in children. J Pediatr Gastroenterol Nutr<br />

1993; 16: 345–364.<br />

89. Romero Y, Cameron AJ, Locke GR. Familial aggregation<br />

of gastroesophageal reflux in patients with Barrett’s<br />

esophagus and esophageal carcinoma. Gastroenterology<br />

1997; 113: 1449–1456.<br />

90. Krug E, Bergmeijer JH, Dees J. Gastroesophageal reflux<br />

and Barrett’s esophagus in adults born with esophageal<br />

atresia. Am J Gastroenterol 1999; 94: 2825–2828.<br />

91. Dahms BB, Rothstein FC. Barrett’s esophagus in children:<br />

a consequence of chronic gastroesophageal reflux.<br />

Gastroenterology 1984; 86: 318–323.<br />

92. Sonnenberg A, El-Serag HB. Clinical epidemiology and<br />

natural history of gastroesophageal reflux disease. Yale J<br />

Biol Med 1999; 72: 81–92.<br />

93. Lagergren J, Bergstrom R, Lindgren A, Nyrén O.<br />

Symptomatic gastroesophageal reflux as a risk factor for<br />

esophageal adenocarcinoma. N Engl J Med 1999; 340:<br />

825–831.<br />

94. Vandenplas Y, de Pont S, Vandemaele C et al.<br />

Dependability of esophageal pH monitoring data on<br />

software. J Pediatr Gastroenterol Nutr 1996; 23: 203–204.<br />

95. Vandenplas Y, Helven R, Goyvaerts H. Comparative<br />

study of glass and antimony electrodes for continuous<br />

oesophageal pH monitoring. Gut 1991; 32: 708–712.<br />

96. Kaynard A, Flora K. Gastroesophageal reflux disease.<br />

Control of symptoms, prevention of complications.<br />

Postgrad Med 2001; 110: 42–44.<br />

97. Vandenplas Y, Belli D, Benhamou P et al. A critical<br />

appraisal of current management practicies for infant<br />

regurgitation – recommendations of a working party. Eur<br />

J Pediatr 1997; 156: 343–357.<br />

98. Ewer AK, James ME, Tobin JM. Prone and left lateral<br />

positioning reduce gastro-oesophageal reflux in preterm<br />

infants. Arch Dis Child Fetal <strong>Neonatal</strong> Ed 1999; 81:<br />

F201–F205.<br />

99. Khoshoo V, Ross G, Brown S, Edell D. Smaller volume,<br />

thickened formulas in the management of gastroe-<br />

References 57<br />

sophageal reflux in thriving infants. J Pediatr<br />

Gastroenterol Nutr 2000; 31: 554–556.<br />

100. Carroll AE, Garrison MM, Christakis DA. A systematic<br />

review of nonpharmacological and nonsurgical therapies<br />

for gastroesophageal reflux in infants. Arch Pediatr<br />

Adolesc Med 2002; 156: 109–113.<br />

101. Bosscher D, van Caillie-Bertrand M, van Dyck K.<br />

Thickening of infant formula with digestible and indigestible<br />

carbohydrate availability of calcium, iron and<br />

zinc in vitro. J Pediatr Gastroenterol Nutr 2000; 30: 373-<br />

378.<br />

102. Aggett PJ, Agostoni C, Goulet O et al. Antireflux or antiregurgitation<br />

milk products for infants and young children:<br />

a commentary by the ESPGHAN Committee on<br />

Nutrition. J Pediatr Gastroenterol Nutr 2002; 34:<br />

496–498.<br />

103. Levtchenko E, Hauser B, Vandenplas Y. Nutritional<br />

value of an ‘anti-regurgitation’ formula. Acta<br />

Gastroenterol Belg 1998; 61: 285–287.<br />

104. Vandenplas Y. Clinical use of cisapride and its<br />

risk–benefit in paediatric patients. Eur J Gastroenterol<br />

Hepatol 1998; 10: 871–881.<br />

105. Aravindhan N, Chisholm DG. Sulfhemoglobinemia<br />

presenting as pulse oximetry desaturations.<br />

Anesthesiology 2000; 93: 883–884.<br />

106. Cinquetti M, Bonetti P, Bertamini P. Current role of antidopaminergic<br />

drugs in pediatrics. Pediatr Med Chir<br />

2000; 22: 1–7.<br />

107. Chou CC, Wu D. Torsade de pointes induced by metoclopramide<br />

in an elderly woman with preexisting<br />

complete left bundle branch block. Chang Gubng Med J<br />

2001; 4: 805–809.<br />

108. Patterson D, Abell T, Rothstein R et al. A double-blind<br />

multicenter comparison of domperidone and metoclopramide<br />

in the treatment of diabetic patients with<br />

symptoms of gastroparesis. Am J Gastroenterol 1999; 94:<br />

1230–1234.<br />

109. Barone JA. Domperidone: a peripherally acting<br />

dopamine 2-receptor antagonist. Ann Pharmacother<br />

1999; 33: 429–440.<br />

110. Lu F, Wu H, Zhang K, Li D. Domperidone and hyperprolactinemia.<br />

Human Yi Ke Da Xue Xue Bao 1998; 23:<br />

100–102.<br />

111. Drolet B, Rousseau G, Daleau P et al. Domperidone<br />

should not be considered a no-risk alternative to<br />

cisapride in the treatment of gastrointestinal motility<br />

disorders. Circulation 2000; 102: 1883–1885.<br />

112. Cameron HA, Reyntjes AJ, Lake-Bakaar G. Cardiac arrest<br />

after treatment with intravenous domperidone. BMJ<br />

1985; 290: 160.<br />

113. Mahon BE, Rosenman MB, Kleiman MB. Maternal and<br />

infant use of erythromycin and other macrolide antibiotics<br />

as risk factors for infantile hypertrophic <strong>pylori</strong><br />

stenosis. J Pediatr 2001; 139: 380-384.<br />

114. Mishra A, Friedman HS, Sinha AK. The effects of<br />

erythromycin on the electrocardiogram. Chest 1999;<br />

115: 983-986.<br />

115. Guerin JM, Leibinger F. Why not use erythromycin in GI<br />

motility? Chest 2002; 121: 301.<br />

116. Gay K, Baughan W, Miller J. The emergence of<br />

Streptococcus pneumoniae resistant to macrolide<br />

antimicrobial agents: a 6-year population-based assessment.<br />

J Infect Dis 2000; 182: 1417–1424.<br />

117. Vandenplas Y, and the ESPGHAN Cisapride Panel.<br />

Current pediatric indications for cisapride. J Pediatr<br />

Gastroenterol 2000; 31: 480–489.<br />

118. Rode H, Millar AJW, Melis J, Cywes S. Esophageal<br />

assessment of gastroesophageal reflux in 18 patients<br />

and the effect of two prokinetic agents: cisapride and<br />

metoclopramide. J Pediatr Surg 1987; 22: 931–934.


58<br />

Gastroesophageal reflux disease<br />

119. Augood C, MacLennan S, Gilbert R, Logan S. Cisapride<br />

treatment for gastro-oesophageal reflux in children.<br />

Cochrane Database Syst Rev 2000; 3: CD002300.<br />

120. Shulman RJ. Report from the NASPGN therapeutics<br />

subcommittee. Cisapride and the attack of the P-450s. J<br />

Pediatr Gastroenterol Nutr 1996; 23: 395–397.<br />

121. Desta Z, Zoukhova N, Mahal SK, Flockhart DA.<br />

Interaction of cisapride with the human cytochrome<br />

P450 system: metabolism and inhibition studies. Drug<br />

Metab Dispos 2000; 28: 789–800.<br />

122. Viskin S. Long QT syndromes and torsades de pointes.<br />

Lancet 1999; 354: 1625–1633.<br />

123. Tonini M, De Ponti F, Di Nucci A, Crema F. Review<br />

article: cardiac adverse effects of gastrointestinal prokinetics.<br />

Aliment Pharmacol Ther 1999; 13: 1585–1591.<br />

124. Hill SL, Evangelista JK, Pizzi AM et al. Proarrhythmia<br />

associated with cisapride in children. Pediatrics 1998;<br />

101: 1053–1056.<br />

125. Ward RM, Lemons JA, Molteni RA. Cisapride: a survey<br />

of the frequency of use and adverse events in premature<br />

newborns. Pediatrics 1999; 103: 469–472.<br />

126. Levy J, Hayes C, Kern J et al. Does cisapride influence<br />

cardiac rhythm? Results of a US multicenter, doubleblind,<br />

placebo controlled pediatric study. J Pediatr<br />

Gastroenterol Nutr 2001; 32: 458–463.<br />

127. Tutar HE, Kansu A, Kalayci AG et al. Effects of cisapride<br />

on ventricular repolarization in children. Acta Paediatr<br />

2000; 89: 820–823.<br />

128. Semama DS, Bernardini S, Louf S et al. Effects of<br />

cisapride on QTc interval in term neonates. Arch Dis<br />

Child Fetal Neonat Ed 2001; 84: F44-F46.<br />

129. Benatar A, Feenstra A, De Craene T, Vandenplas Y. QTc<br />

interval in infants and serum concentrations. J Pediatr<br />

Gastroenterol Nutr 2001; 33: 41–46.<br />

130. Benatar A, Feenstra A, Decraene T, Vandenplas Y.<br />

Effects of cisapride on corrected QT interval, heart rate,<br />

and rhythm in infants undergoing polysomnography.<br />

Pediatrics 2000; 106: E85.<br />

131. Cools F, Benatar A, Bruneel E et al. A comparison of the<br />

pharmacokinetics of two dosing regimens of cisapride<br />

and their effects on corrected QT interval in premature<br />

infants. Eur J Clin Pharmacol. 2003; 59: 17–22.<br />

132. Kivisto KT, Lilja JJ, Backman JT, Neuvonen PJ. Repeated<br />

consumption of grapefruit juice considerably increases<br />

plasma concentrations of cisapride. Clin Pharmacol<br />

Ther 1999; 66: 448–453.<br />

133. Napolitano C, Schwartz PJ, Brown AM et al. Evidence<br />

for a cardiac ion channel mutation underlying druginduced<br />

QT prolongation and life-threatening arrhythmias.<br />

J Cardiovasc Electrophysiol 2000; 11: 691–696.<br />

134. Walker AM, Szneke P, Weatherby LB et al. The risk of<br />

serious cardiac arrhythmias among cisapride users in<br />

the United Kingdom and Canada. Am J Med 1999; 107:<br />

356–362.<br />

135. Augood C, Gilbert R, Logan S, MacLennan S. Cisapride<br />

treatment for gastro-oesophageal reflux in children.<br />

Cochrane Database Syst Rev 2002; 3: CD002300.<br />

136. Coremans G, Kerstens R, De Pauw M, Stevens M.<br />

Prucalopride is effective in patients with severe chronic<br />

constipation in whom laxatives fail to provide adequate<br />

relief. Results of a double-blind, placebo-controlled clinical<br />

trial. Digestion 2003; 67: 82–89.<br />

137. Culy CR, Bhana N, Plosker GL. Ondansetron: a review<br />

of its use as an antiemetic in children. Paediatr Drugs<br />

2001; 3: 441–79.<br />

138. Wagstaff A, Frampton J, Croom K. Tegaserod: a review of<br />

its use in the management of irritable bowel syndrome<br />

with constipation in women. Drugs 2003; 63:<br />

1101–1120.<br />

139. Kahrilas PJ, Quigley EM, Castell DO, Spechler SJ. The<br />

effects of tegaserod (HTF 919) on oesophageal acid<br />

exposure in gastro-oesophageal reflux disease. Aliment<br />

Pharmacol Ther 2000; 14: 1503-1509.<br />

140. Ciccaglione AF, Marzio L. Effect of acute and chronic<br />

administration of the GABA(B) agonist baclofen on 24<br />

hour pH metry and symptoms in control subjects and in<br />

patients with gastro-oesophageal reflux disease. Gut<br />

2003; 52: 464–470.<br />

141. Wiersma HE. Pharmacokinetics of a single oral dose of<br />

baclofen in pediatric patients with GERD. Ther Drug<br />

Monitor 2003; 25: 93–98.<br />

142. Vandenplas Y, Belli DC, Benatar A et al. The role of<br />

cisapride in the treatment of pediatric gastroesophageal<br />

reflux. The European Society of Pediatric<br />

Gastroenterology, Hepatology and Nutrition. J Pediatr<br />

Gastroenterol Nutr 1999; 28: 518–528.<br />

143. Flockhart DA, Desta Z, Mahal SK. Selection of drugs to<br />

treat gastro-oesophageal reflux disease: the role of drug<br />

interactions. Clin Pharmacokinet 2000; 39: 295–309.<br />

144. Bachmann KA, Sullivan TJ, Jauregui L et al. Drug interactions<br />

of H2-receptor antagonists. Scand J Gastroenterol<br />

1994; 206 (Suppl): 14–19.<br />

145. Paul K, Redman CM, Chen M. Effectiveness and safety<br />

of nizatidine, 75 mg, for the relief of episodic heartburn.<br />

Aliment Pharmacol Ther 2001; 15: 1571–1577.<br />

146. Sabesin SM. Safety issues relating to long-term treatment<br />

with histamine H2-receptor antagonists. Aliment<br />

Pharmacol 1993; 7(Suppl 2): S35–S40.<br />

147. Kelly DA. Do H2 receptor antagonists have a therapeutic<br />

role in childhood? J Pediatr Gastroenterol 1994; 19:<br />

270–276.<br />

148. Adachi N, Seyfried FJ, Arai T. Blockade of central histaminergic<br />

H2-receptors aggravates ischemic neuronal<br />

damage in gerbil hippocampus. Crit Care Med 2001; 29:<br />

1189–1194.<br />

149. Emovon OE, King JA, Holt CO, Browne BJ. Ranitidine<br />

induced acute interstitial nephritis in a cadaveric renal<br />

allograft. Am J Kidney Dis 2001; 38: 169–172.<br />

150. Boehning W. Effect of cimetidine and ranitidine on<br />

plasma theophylline in patients with chronic obstructive<br />

airways disease treated with theophylline and corticosteroids.<br />

Eur J Clin Pharmacol 1990; 38: 43–45.<br />

151. Alliet P, Devos E. Ranitidine-induced bradycardia in a<br />

neonate – secondary to a congenital long QT interval<br />

syndrome. Eur J Pediatr 1993; 152: 933–934.<br />

152. Hu WH, Wang KY, Hwang DS et al. Histamine 2 receptor<br />

blocker – ranitidine and sinus mode dysfunction.<br />

Zhonghua Yi Xue Za Zhi 1997; 60: 1–5.<br />

153. Nault MA, Milne B, Parlow JL. Effects of the selective<br />

H1 and H2 histamine receptor antagonists loretadine<br />

and ranitidine on autonomic control of the heart.<br />

Anesthesiology 2002; 96: 336–341.<br />

154. Ooie T, Saaikawa T, Hara M et al. H2-blocker modulates<br />

heart rate variability. Heart Vessels 1999; 14: 137–142.<br />

155. Basit AW, Newaton JM, Lacey LF. Susceptibility of the<br />

H2-receptor antagonists cimetidine, famotidine and<br />

nizatidine, to metabolism by the gastrointestinal<br />

microflora. Int J Pharm 2002; 237: 23–33.<br />

156. Cothran DS, Borowitz SM, Sutphen JL et al. Alteration<br />

of normal gastric flora in neonates receiving ranitidine. J<br />

Perinatol 1997; 17: 383–388.<br />

157. Messori A, Trippoli S, Vaiani M et al. Bleeding and<br />

pneumonia in intensive care patients given ranitidine<br />

and sucralfate for prevention of stress ulcer: meta-analysis<br />

of randomised controlled trials. BMJ 2000; 321:<br />

1103–1106.<br />

158. Huang JQ, Hunt RH. Pharmacological and pharmacodynamic<br />

essentials of H(2)-receptor antagonists and proton


pump inhibitors for the practising physician. Best Pract<br />

Res Clin Gastroenterol 2001; 15: 355–370.<br />

159. Londong W, Phillips J, Johnson NJ, Wood JR. The effect<br />

of combined therapy with ranitidine and pirenzipine in<br />

the treatment of reflux esophagitis. Aliment Pharmacol<br />

Ther 1992; 6: 609–618.<br />

160. Vakil NB, Shaker R, Johnson DA et al. The new proton<br />

pump inhibitor esomeprazole is effective as a maintenance<br />

therapy in GERD patients with healed erosive<br />

esophagitis: a 6-month, randomized, double-blind,<br />

placebo-controlled study of efficacy and safety. Aliment<br />

Pharmacol Ther 2001; 15: 927–935.<br />

161. Jones R, Bytzer P. Acid suppression in the management<br />

of gastro-oesophageal reflux disease – an appraisal of<br />

treatment options in primary care. Aliment Pharmacol<br />

Ther 2001; 16: 765–771.<br />

162. Ferron GM, Preston RA, Noveck RJ et al.<br />

Pharmacokinetics of pantoprazole in patients with<br />

moderate and severe hepatic dysfunction. Clin Ther<br />

2001; 23: 1180–1192.<br />

163. James LP, Kearns GL. Pharmacokinetics and pharmacodynamics<br />

of famotidine in paediatric patients. Clin<br />

Pharmacokinetic 1996; 31: 103–110.<br />

164. Scott LJ. Lansoprazole: in the management of gastroesophageal<br />

reflux disease in children. Paediatr Drugs<br />

2003; 5: 57–61.<br />

165. Zimmermann AE, Walters JK, Katona BG et al. A review<br />

of omeprazole use in the treatment of acid-related disorders<br />

in children. Clin Ther 2001; 23: 660-679.<br />

166. Leufkans H, Claessens A, Heerdink E et al. A prospective<br />

follow-up study of 5669 users of lansoprazole in<br />

daily practice. Aliment Pharmacol Ther 1997; 11:<br />

887–897.<br />

167. Castot A, Bidault I, Dahan R, Efthymiou ML. Evaluation<br />

of unexpected and toxic effects of omeprazole A90 –<br />

reported to the regional centers of pharmacovigilance<br />

during the first 22 postmarketing months. Therapie<br />

1993; 48: 469–474.<br />

168. Berardi RR. A critical evaluation of proton pump<br />

inhibitors in the treatment of gastroesophageal reflux<br />

disease. Am J Manag Care 2000; 6(9 Suppl): S491–505.<br />

169. Andersson T, Hanssan-alin M, Hasselgren G, Rohss K.<br />

Drug interaction studies with esomeprazole, the (S)isomer<br />

of omeprazole. Clin Pharmacokinet 2001; 40:<br />

523–537.<br />

170. ter Heide H, Hendriks HJ, Heijmans H et al. Are children<br />

with cystic fibrosis who are treated with a protonpump<br />

inhibitor at risk for vitamin B12 deficiency? J<br />

Pediatr Gastroenterol Nutr 2001; 33: 342–345.<br />

171. Singh P, Indaram A, Greenberg R et al. Long term<br />

omeprazole therapy for reflux esophagitis: follow-up in<br />

serum gastrin levels, EC cell hyperplasia and neoplasia.<br />

World J Gastroenterol 2000; 6: 789–792.<br />

172. Nuanton M, Peterson GM, Bleasel MD. Overuse of<br />

proton pump inhibitors. J Clin Pharm Ther 2000; 25:<br />

333–338.<br />

References 59<br />

173. Amaro R, Montelongo PC, Barkin JS. Sucralfate-induced<br />

diarrhea in an enterally fed patient. Am J Gastroenterol<br />

1999; 94: 2328–2329.<br />

174. Guy C, Ollagnier M. Sucralfate and bezoars: data from<br />

the system of pharmacologic vigilance and review of the<br />

literature. Therapie 1999; 54: 55–58.<br />

175. Hemstreet BA. Use of sucralfate in renal failure. Ann<br />

Pharmacother 2001; 35: 360–364.<br />

176. Fleming GF, Vokes EE, McEvilly JM et al. Double-blind,<br />

randomized crossover study of metoclopramide and<br />

batanopride for prevention of cisplatin-induced emesis.<br />

Cancer Chemother Pharmacol 1991; 28: 226–227.<br />

177. Talley NJ, Verlinden M, Snape W et al. Failure of a<br />

motilin receptor agonist (ABT-229) to relieve the symptoms<br />

of functional dyspepsia in patients with and<br />

without delayed gastric emptying: a randomized<br />

double-blind placebo-controlled trial. Aliment<br />

Pharmacol Ther 2000; 14: 1653–1661.<br />

178. Mahmood Z, McMahon BP, Arfin Q et al. Endocinch<br />

therapy for gastro-oesophageal reflux disease: a one year<br />

prospective follow up. Gut 2003; 52: 34–39.<br />

179. Wolfsen HC, Richards WO. The Stretta procedure for<br />

the treatment of GERD: a registry of 558 patients. J<br />

Laparoendosc Adv Surg Tech A 2002; 12: 395–402.<br />

180. Johnson DA, Ganz R, Aisenberg J et al. Endoscopic,<br />

deep mural implantation of enteryx for the treatment of<br />

GERD: 6-month follow-up of a multicenter trial. Am J<br />

Gastroenterol 2003; 98: 250–258.<br />

181. Galmiche JP, Bruley des Varannes S. Endoluminal therapies<br />

for gastro-oesophageal reflux disease. Lancet 2003;<br />

361: 1119–1121.<br />

182. Fonkalsrud EX, Bustorff-Silva J, Perez CA et al.<br />

Antireflux surgery in children under three months of<br />

age. J Pediatr Surg 1999; 34: 527–531.<br />

183. Pearl RH, Robie DK, Ein SH et al. Complications of<br />

gastroesophageal reflux surgery in neurologically<br />

impaired versus neurologically normal children. J<br />

Pediatr Surg 1990; 25: 1169–1173.<br />

184. Alexander F, Wyllie R, Jirousek K et al. Delayed gastric<br />

emptying affects outcome of Nissen fundoplication in<br />

neurologically impaired children. Surgery 1997; 122:<br />

690–697.<br />

185. Spitz L, Roth K, Kiely EM et al. Operation for gastrooesophageal<br />

reflux associated with severe mental retardation.<br />

Arch Dis Child 1993; 68: 347–351.<br />

186. Booth MI, Jones L, Stratford J, Dehn TC. Results of<br />

laparoscopic Nissen fundoplication 8 years after<br />

surgery. Br J Surg 2002; 89: 476–481.<br />

187. Bergmeijer JH, Harbers JS, Molenaar JC. Function of<br />

pediatric Nissen–Rosetti fundoplication followed up<br />

into adolescence and adulthood. J Am Coll Surg 1997;<br />

184: 259-261.<br />

188. Burd RS, Price MR, Whalen TV. The role of protective<br />

antireflux procedures in neurologically impaired children:<br />

a decision analysis. J Pediatr Surg 2002; 37: 500-<br />

506.


5<br />

Introduction<br />

Achalasia<br />

Carl-Christian A Jackson and Donald C Liu<br />

Diseases of the esophagus can generally be categorized<br />

into four types of abnormality: those of inadequate<br />

lower esophageal relaxation; those of<br />

uncoordinated contraction; those of hypercontraction;<br />

and those of hypocontraction. 1 Classic achalasia<br />

falls into this first category. Directly translated,<br />

achalasia means ‘failure to relax’, and<br />

describes a rare disease in which the lower<br />

esophageal sphincter (LES) remains constricted<br />

during swallowing and there is an absence of<br />

esophageal peristalsis. The first case of achalasia<br />

was reported in 1674 by Thomas Willis. 2 His<br />

patient was otherwise healthy, but suffered from<br />

frequent vomiting and regurgitation of food.<br />

Successful palliation was achieved through postprandial<br />

passage of a whalebone with a sponge<br />

attached to the tip, which accomplished both<br />

tamping of the food through the LES, as well as<br />

some degree of forceful dilatation. However, it<br />

was Sir Cooper Perry in 1913 who first used the<br />

term ‘achalasia’ to describe this disease. 3–5<br />

The exact etiology of achalasia is unknown, but<br />

certain factors remain consistent, namely inflammation<br />

of the myenteric plexuses with varying<br />

degrees of ganglion cell loss. When symptomatology<br />

suggests achalasia, the diagnosis is confirmed<br />

through radiographic and manometric studies. In<br />

treating children with achalasia, numerous<br />

modalities have been employed with varying<br />

degrees of success, including drug treatment,<br />

forceful dilatation and surgical esophagomyotomy.<br />

The purpose of this chapter is to provide<br />

an overview of this disease process, the tools with<br />

which to diagnose it and the available treatment<br />

options.<br />

Epidemiology<br />

Epidemiological studies of achalasia have been<br />

relatively few. A summary of the available investigations,<br />

which include both adults and children,<br />

shows an incidence of 0.4–1.2/10 5 persons/year in<br />

Europe and North America. 6–11 The incidence in<br />

children is far lower. Approximately 4–5% of all<br />

cases of achalasia are diagnosed before the age of<br />

15. More so than the adult form, childhood achalasia<br />

can occur with associated abnormalities,<br />

such as familial glucocorticoid deficiency,<br />

alacrima, autonomic system disorder, short<br />

stature, microcephaly and nerve deafness. 12–16<br />

Contrary to the generally accepted view of a female<br />

predominance, achalasia actually appears to affect<br />

men and women equally. 17,18 Studies that exhibit<br />

higher rates in females, when adjusted for their<br />

higher population of women, show that the incidence<br />

of achalasia approaches that expected in the<br />

general population. 6,9<br />

Etiology<br />

The etiology of achalasia remains uncertain.<br />

Proposed mechanisms for the disease include<br />

genetics, infectious agents, autoimmune destruction,<br />

and primary neural degeneration. Various<br />

case reports have identified instances of both horizontal<br />

and vertical ‘transmission’. 19–24 The majority<br />

of these familial cases, however, occur in the<br />

setting of consanguinity. While this raises the<br />

possibility of a rare, recessive gene being<br />

expressed, a large-scale study of more than 1000<br />

first-degree relatives of patients with achalasia<br />

showed no affected relatives – contrary to expected<br />

Mendelian genetics. 25<br />

61


62<br />

Achalasia<br />

Five per cent of patients with chronic Chagas’<br />

disease develop poor LES relaxation and megaesophagus<br />

that mimics classic achalasia, 26 which<br />

suggests that an infectious etiology of achalasia is<br />

possible. The protozoan Trypanosoma cruzi which<br />

causes Chagas’ disease acts through direct invasion<br />

of cells, toxin production or antigenic factors<br />

to cause degeneration of esophageal neurons. The<br />

mechanism of this process is also uncertain, and<br />

may be from direct injury to neurons of the esophagus,<br />

an inflammatory reaction or the generation<br />

of autoantibodies against these nerves. 27 Potential<br />

infectious candidates for idiopathic achalasia<br />

include measles and varicella viruses, but as yet<br />

no firm evidence exists to confirm this association.<br />

25,28–31<br />

Pathology<br />

While the etiology of achalasia remains unclear,<br />

the histopathological changes associated with it<br />

have been well described. Surgical specimens<br />

involving strips of esophageal muscle from<br />

esophagomyotomy or entire resected esophagi<br />

have formed the basis of study. 32–35 Gross examination<br />

of esophagectomy specimens from patients<br />

with severe/recurrent achalasia reveals the classic<br />

morphology of a dilated proximal esophagus, with<br />

distal tapering. The muscularis propria is<br />

frequently hypertrophied, and the mucosa exhibits<br />

exaggerated longitudinal folds. 32<br />

Upon microscopic examination, there is a range of<br />

associated changes in the esophagus. The most<br />

classic feature is an absence of myenteric ganglion<br />

cells, which is seen in specimens from patients<br />

with both early and late achalasia. 32,34–38 While<br />

there is no apparent association between the<br />

number of remaining ganglion cells and the degree<br />

of symptoms, there does seem to be a temporal<br />

relationship, i.e. patients with longstanding<br />

disease tend to exhibit fewer ganglion cells. 34,38<br />

The most consistent histological feature, seen in<br />

all esophageal specimens, is inflammation. 32,35<br />

The inflammation occurs in and around the myenteric<br />

nerves, predominantly involves lymphocytes<br />

and results in neural fibrosis. Goldblum et al made<br />

an interesting observation in their study of<br />

patients with early achalasia. 35 A woman with<br />

rapid onset of dysphagia and weight loss underwent<br />

esophagomyotomy only 2.5 months after the<br />

onset of symptoms. Histological examination was<br />

significant for mild myenteric inflammation,<br />

normal numbers of ganglion cells and an absence<br />

of fibrosis. These findings suggest that the earliest<br />

pathological feature of achalasia is myenteric<br />

inflammation.<br />

Additional histological changes result from<br />

esophageal obstruction and the associated stasis.<br />

Esophageal obstruction leads to varying degrees of<br />

muscular hypertrophy of the muscularis propria,<br />

most notably in the inner circular layer. 32,36,39<br />

Further findings include small leiomyomas of the<br />

inner circular layer, degenerative changes of the<br />

muscularis propria as well as focal fibrosis of the<br />

muscularis propria (again, predominantly in the<br />

inner circular layer). 32 Stasis of luminal contents<br />

results in both proximal and distal squamous<br />

hyperplasia, as well as changes similar to those<br />

seen in reflux esophagitis, namely papillomatosis<br />

and basal cell hyperplasia. 32 However, owing to<br />

the increased LES pressure limiting passage of<br />

gastric contents, these findings are unlikely to be<br />

due to true gastroesophageal reflux.<br />

Extra-esophageal pathology accompanies achalasia,<br />

predominantly related to esophageal innervation.<br />

Both the vagus nerve and the vagal dorsal<br />

motor nucleus are affected. Electron microscopy of<br />

periesophageal vagal nerve biopsies reveals<br />

changes resembling Wallerian degeneration (i.e.<br />

secondary to nerve transaction), changes that are<br />

not seen on light microscopy. 33,38 Whether the<br />

affected nerves are motor, sensory or vagalassociated<br />

sympathetic nerves is unclear.<br />

Evaluation of the dorsal motor nucleus of the<br />

vagus, performed upon autopsy, also reveals<br />

degenerative changes, neuron loss and Lewy<br />

bodies. 40<br />

Pathophysiology<br />

The resting state of the LES is one of contraction.<br />

Cholinergic neurons provide resting tone, whereas<br />

inhibitory neurons provide vagal-mediated relaxation<br />

34,41,42 Vagal efferents contain both excitatory<br />

and inhibitory signals; however, the predominant<br />

effect of vagal stimulation is LES relaxation, as<br />

mediated by non-adrenergic, non-cholinergic<br />

(NANC) nerves. 43 In normal patients, stimuli from<br />

gastric, esophageal and pharyngeal sensory nerves


are relayed to, and processed by, the dorsal motor<br />

nucleus of the vagus, which results in modulation<br />

of both excitatory and inhibitory impulses to the<br />

LES. 44<br />

Two strong candidates for mediation of LES relaxation<br />

are vasoactive intestinal polypeptide (VIP)<br />

and nitric oxide (NO). After it was realized that<br />

VIP was found in high concentrations in gut<br />

sphincters, subsequent investigations showed it to<br />

be a potent inhibitor of LES contraction in<br />

vitro. 45,46 Further association of VIP with sphincteric<br />

relaxation and achalasia is suggested by<br />

immunohistochemical studies that have shown<br />

decreased or absent VIP-containing neurons in LES<br />

specimens from patients with achalasia. 47–49<br />

Human manometric studies have shown that the<br />

LES in patients with achalasia, as compared to<br />

normal controls, is hypersensitive to exogenous<br />

VIP, again supporting a role for VIP in LES relaxation.<br />

44<br />

NO also figures prominently in gastrointestinal<br />

tract smooth muscle and sphincteric<br />

mechanisms. 50–57 The role of NO appears to be that<br />

of sphincter-muscle hyperpolarization, and therefore<br />

relaxation, and the loss of NO has been associated<br />

with conditions of persistent contraction,<br />

such as achalasia. 58–62 Preiksaitis et al elegantly<br />

demonstrated that blocking NO synthase prevents<br />

relaxation of LES muscles by NANC nerves, and<br />

that exogenous NO from sodium nitroprusside can<br />

overcome this inhibition. 57 Inhibitory regulation of<br />

gastrointestinal muscles by NANC transmitters<br />

was generally believed to be direct, until an intermediary,<br />

interstitial cell was proposed by Cajal (a<br />

cell that bears his name). 63 Recent work in both<br />

animal and human models confirms the relationship<br />

of interstitial cells of Cajal (ICC) to nitric<br />

Table 5.1 Symptoms of achalasia (from reference 67)<br />

Symptoms Number of patients Mean (%) Mean range (%)<br />

Dysphagia 1930 97 82–100<br />

Regurgitation 1892 75 56–97<br />

Weight loss 1675 58 30–91<br />

Chest pain 1894 43 17–95<br />

Heartburn 127 36 27–42<br />

Cough 732 30 11–46<br />

Signs and symptoms 63<br />

oxide synthase (NOS)-positive neurons. 53,64 It is<br />

accepted that NO release results in sphincteric<br />

relaxation; however, the exact mechanism has not<br />

been fully elucidated. While evaluation of the LES<br />

in adults shows that ICC are altered, this may be a<br />

result of disruption of the interaction between<br />

NOS-positive neurons and ICC, because examination<br />

of the pediatric esophagus in achalasia (and<br />

therefore a shorter duration of disease) shows<br />

morphologically normal ICC, but disrupted<br />

contact with NOS-positive neurons. 53,54<br />

The precise cause of achalasia remains to be elucidated<br />

but the loss of NO- and/or VIP-associated<br />

neurons appears to be integral to the disease. By<br />

their destruction, these neurons permit unopposed<br />

contraction of the LES.<br />

Signs and symptoms<br />

Because the incidence of achalasia in children is<br />

so low, the majority of information regarding the<br />

diagnosis comes from adults. The earliest, and<br />

most common, symptom of achalasia is dysphagia.<br />

This dysphagia initially is for solids, but<br />

frequently progresses to dysphagia for liquids by<br />

the time treatment is sought. 65–67 The next most<br />

common symptom in adults is regurgitation,<br />

which is non-acidic and non-bilious, owing to the<br />

contracted LES, and most often occurs right after<br />

eating or during sleep. Patients often find ways to<br />

accommodate these dysfunctions, and will ameliorate<br />

symptoms by such methods as drinking large<br />

volumes of water with meals, holding their arms<br />

above the head or performing a Valsalva maneuver.<br />

67–69 A summary of the most common presenting<br />

symptoms is provided in Table 5.1.


64<br />

Achalasia<br />

The presenting symptoms in children tend to<br />

correlate with patient age and can mimic those<br />

seen in adults, or they can be more vague, thus<br />

requiring a high degree of suspicion for diagnosis.<br />

Symptoms in older children (more than 7 years)<br />

tend to parallel those seen in adults, so dysphagia<br />

and regurgitation predominate, but with substernal<br />

chest pain and burning also appearing in about<br />

half the patients. 70 Children aged less than 6 years,<br />

particularly infants, more commonly present with<br />

respiratory symptoms, complaints, similar to those<br />

of gastroesophageal reflux disease (GERD), occasional<br />

emesis and failure to thrive. 12,71<br />

Regurgitation and dysphagia are the most common<br />

symptoms, present in 83% and 71–80% of<br />

patients, respectively, followed by failure to thrive<br />

in 54–70%. 70,72 Especially in non-verbal children,<br />

a diagnosis of achalasia should be entertained<br />

when presented with a patient experiencing significant<br />

chronic respiratory symptoms, such as<br />

choking, recurrent pneumonia, severe asthma,<br />

chronic bronchitis or chronic cough, because these<br />

are seen in 25–100% of patients. 12,70 The physical<br />

examination tends to be unremarkable, but can<br />

reveal findings secondary to dysphagia (and a<br />

resultant decrease in oral intake) and food stasis,<br />

such as weight loss, malnutrition and halitosis. 67<br />

Laboratory and instrumental<br />

investigations<br />

Radiographic studies<br />

For patients in whom a diagnosis is uncertain, a<br />

plain chest radiograph may be the first available<br />

study. The classically described finding on upright<br />

chest X-ray is an absent gastric air-bubble. This<br />

finding, present in nearly all normal individuals, is<br />

absent in approximately half of patients with achalasia.<br />

73,74 Additional findings include a widened<br />

mediastinum from esophageal dilatation, a posterior<br />

mediastinal air–fluid level from retained<br />

food/secretions and lung parenchymal abnormalities<br />

from chronic aspiration. 67,73,75<br />

Barium swallow, however, is the definitive radiographic<br />

study, and, in a review of the European<br />

experience with achalasia, it is the most<br />

commonly performed test. 76 On barium swallow,<br />

classic achalasia typically shows esophageal<br />

dilatation with distal narrowing at the gastroesophageal<br />

(GE) junction, the ‘bird’s beak’ deformity<br />

(Figure 5.1). Also, retained food/secretions,<br />

absent peristalsis, tortuosity of the esophagus and,<br />

occasionally, an epiphrenic diverticulum can be<br />

seen. 67,73,75 Because a barium swallow is<br />

performed under fluoroscopy it is a dynamic<br />

study, and may also reveal back-and-forth sloshing<br />

of the barium boluses when the patient is in the<br />

supine position, owing to the esophageal dilatation<br />

and the ineffective peristalsis. 67,75,77 However, the<br />

diagnostic accuracy of barium swallow is approximately<br />

85%, which can be due to a very early stage<br />

Figure 5.1 ‘Bird’s beak’ appearance characteristic of<br />

esophageal achalasia on esophogram (courtesy of Hans<br />

Björknäs, Gastrolabs, Finland).


of achalasia not showing the classic signs, due to a<br />

tumor of the GE junction mimicking achalasia or<br />

due to a peptic stricture. 67,73,76<br />

Computed tomography (CT) has no real role in the<br />

diagnosis of achalasia. 67 The findings are consistent<br />

with those already seen on plain X-ray or<br />

barium swallow, such as a dilated esophagus,<br />

esophageal air–fluid levels, and possibly displacement<br />

of mediastinal structures by a dilated esophagus.<br />

Furthermore, masses that may cause<br />

pseudoachalasia are infrequently identified on a<br />

CT scan. 78,79<br />

With the desire to limit radiation exposure, particularly<br />

in children, alternative means of diagnosis<br />

are sought. Radionuclide bolus transport has been<br />

evaluated as one such alternative, with achalasia<br />

diagnosed when esophageal emptying time is<br />

prolonged. When evaluating patients with manometrically<br />

diagnosed achalasia using radionuclide<br />

transport, Stacher et al found that sensitivity for<br />

this evaluation was 68%, with a specificity of<br />

95%. 80 Their criteria were an esophageal emptying<br />

time of ≥ 20 s (the time for 95% of the bolus to<br />

enter the stomach), compared to a normal median<br />

time of 7.3 s (range 5.5–12.0 s). Because radionuclide<br />

bolus transport is a functional study with<br />

relatively low sensitivity (other esophageal motility<br />

disorders also show prolonged transit time),<br />

this study may be best suited for follow-up of<br />

patients after treatment, or in the few cases in<br />

which achalasia is suspected, despite normal LES<br />

pressure on manometry. 67,73,80<br />

Manometry<br />

The gold standard for accurate diagnosis of achalasia<br />

remains esophageal manometry, using either<br />

a perfused-catheter or a solid-state system. The<br />

minimum information collected from manometry<br />

will include resting LES pressure, relaxed LES<br />

pressure and peristaltic function of the esophageal<br />

body. Depending on local protocol, upper<br />

esophageal sphincter (UES) pressure may also be<br />

assessed. Fluoroscopy may be used to assist in<br />

positioning of the sensors. Manometry is usually<br />

performed with a catheter containing multiple,<br />

evenly spaced sensors, which is passed by the oral<br />

or nasal route into the stomach. Resting gastric<br />

pressures are measured to provide a baseline. The<br />

Laboratory and instrumental investigations 65<br />

catheter is slowly withdrawn until the sensor is<br />

measuring LES pressure. Under quiet respirations,<br />

resting LES pressure is measured at midrespiration.<br />

Swallow-induced LES relaxation is<br />

measured with the patient taking several wet swallows<br />

using small volumes of liquid. The catheter is<br />

further withdrawn until the distal sensor is a few<br />

centimeters above the LES and the proximal<br />

sensors are spaced throughout the esophageal<br />

body. Esophageal peristalsis and wave progression<br />

are assessed during several more wet swallows.<br />

Because achalasia affects the smooth muscles of<br />

the esophagus, the manometric findings involve<br />

the mid- and distal esophagus, sparing the proximal<br />

regions of predominantly striated muscle. The<br />

two manometric abnormalities found in all patients<br />

with achalasia are aperistalsis of the esophageal<br />

body and abnormal LES relaxation. 1,67–69<br />

Simultaneous, low-amplitude contractions<br />

(< 40 mmHg) throughout the esophageal body are<br />

characteristic of the aperistalsis seen after wet<br />

swallows. Abnormal LES relaxation is characterized<br />

by either absent/incomplete relaxation<br />

(70–80% of patients) or normal relaxation of short,<br />

and therefore ineffective, duration – typically less<br />

than 6 s each (20–30% of patients). 69, 81 This latter<br />

finding commonly accompanies early stage achalasia.<br />

68 Manometric findings associated with achalasia,<br />

but not required for diagnosis, include<br />

elevated resting LES pressure and resting<br />

esophageal body pressure exceeding baseline<br />

gastric pressure. 1 Table 5.2 summarizes normal<br />

and abnormal manometric findings.<br />

Endoscopy<br />

Despite radiographic and manometric evidence<br />

typical of achalasia, pseudoachalasia secondary to<br />

a tumor at the GE junction must be ruled out. 67,73<br />

Findings consistent with achalasia may include a<br />

dilated, atonic esophageal body; mucosal thickening<br />

and/or erythema; and a puckered LES which<br />

fails to open with insufflation, but which is easily<br />

passed with the endoscope. If there is visual<br />

evidence of extrinsic compression or the endoscope<br />

fails to pass through the GE junction with<br />

gentle pressure, then a tumor must be excluded.<br />

Biopsies should be taken from suspicious areas<br />

seen above the GE junction or seen on retroflexion<br />

from within the stomach.


66<br />

Achalasia<br />

Table 5.2 Normal and abnormal manometric findings (from references 1, 67 and 68)<br />

Treatment<br />

Until such time as the primary cause of achalasia<br />

can be reversed, namely the loss of esophageal<br />

innervation by inhibitory neurons, treatment of<br />

achalasia must address the relief of symptoms.<br />

Since the primary disorders are aperistalsis and<br />

abnormal LES relaxation, the goal of non-invasive<br />

and invasive treatments is relief of the obstruction<br />

and its associated dysphagia. Medical treatment<br />

focuses on promoting LES relaxation, whereas<br />

endoscopic and surgical treatments address<br />

disruption of the LES muscle itself.<br />

Medical options<br />

Swallow-induced Peristaltic wave Distal wave<br />

Basal LES pressure LES relaxation progression amplitude<br />

Normal 10–45 mmHg complete (less than 2–8 cm/s from 30–180 mmHg<br />

8 mmHg above gastric UES to LES<br />

baseline, or 95% of<br />

LES baseline)<br />

Achalasia >45 mmHg (up to 40% incomplete (more simultaneous


Surgical options<br />

The gold standard in treatment of achalasia is<br />

surgical esophagomyotomy – the modified Heller<br />

procedure. It is against this procedure that other<br />

surgical, and medical, treatments are compared.<br />

Surgical treatments include endoscopy with botulinum<br />

toxin injection, forceful dilatation, open<br />

Heller myotomy and laparoscopic Heller myotomy.<br />

Endoscopic botulinum toxin injection<br />

With endoscopy playing an important role in the<br />

work-up of achalasia, it seems attractive to initiate<br />

therapy concurrently. Botulinum toxin (BoTox) is a<br />

potent inhibitor of presynaptic acetylcholine<br />

release, and can be injected into the LES through<br />

an endoscope. After a pilot study had demonstrated<br />

efficacy, Pasricha et al undertook a randomized,<br />

blinded and controlled study of BoTox. 89<br />

This study showed significant decreases in<br />

symptom scores, resting LES pressure and<br />

esophageal food retention. Approximately half of<br />

the injected patients, however, had no response or<br />

relapsed within 2 months of initial treatment and<br />

required either repeat injection or pneumatic<br />

dilatation. Subsequent studies in adults have<br />

shown that BoTox generally provides good initial<br />

results, but these improvements typically are not<br />

long-lasting. 90–92 Follow-up showed that 10–35%<br />

of patients had no initial response, 30–40%<br />

relapsed within 4 months, and 38–67% had<br />

‘lasting’ effects to an average of 1.3–2.5 years. 90–92<br />

An initial experience with BoTox in an 11-year-old<br />

child showed encouraging initial results, with<br />

repeat injection required 1 year after initial<br />

therapy. 93 Two recent studies have evaluated the<br />

role of BoTox in a series of children. 94,95 In both,<br />

the mean duration of symptomatic relief was<br />

short-lived (3–7 months) and required repeat injections<br />

or eventual Heller myotomy. Among the<br />

limited patients who did not progress to surgical<br />

treatment, a few showed lasting benefit over<br />

several years, and some opted for frequent<br />

repeated injections rather than undergoing<br />

surgery. While BoTox is generally considered safe,<br />

complications such as gastroesophageal reflux,<br />

esophageal inflammation and ulceration with<br />

hemorrhage have been noted. 96 The consensus at<br />

this time relegates BoTox injection to patients who<br />

are unwilling or medically unsuitable for more<br />

Treatment 67<br />

invasive correction, or as a subsequent adjunct to<br />

patients who are symptomatic after myotomy or<br />

dilatation. 67, 90,91,94,95<br />

Esophageal dilatation<br />

In contrast to the above-described treatments,<br />

esophageal dilatation addresses achalasia through<br />

forceful disruption of the LES muscle fibers. The<br />

largest experience with dilatation has been in<br />

adults, and current therapy involves the use of<br />

balloon dilators. The balloons are placed across<br />

the LES, rapidly inflated and then deflated after a<br />

period of 30 s to several minutes, depending on<br />

patient tolerance. As the procedure is performed<br />

under fluoroscopy, the dilatations are repeated<br />

until the waist of the LES is obliterated by the<br />

balloon. ‘Excellent’ to ‘good’ results, based on<br />

patient survey, are obtained in 65–93% of patients<br />

(this higher number was based on pooling results<br />

from multiple, graded dilatations). 82,97–99 With<br />

long-term follow-up, trends were seen in patients<br />

who either failed initial treatment or quickly<br />

relapsed, with the most significant in younger age<br />

(under 45 years). 73,100 While dilatation is relatively<br />

safe, early and late complications do occur. The<br />

most significant early complication is perforation<br />

(0–12%), which if recognized early can be treated<br />

conservatively with good effect. 73,82,101<br />

One of the major benefits of esophageal dilatations<br />

in adults is avoidance of general anesthesia, which<br />

is generally lost when treating children, making it<br />

a less attractive option. Despite showing some<br />

degree of success in a small series, 102 the typical<br />

experience is poor long-term response requiring<br />

frequent re-dilatation, and eventually surgery,<br />

particularly in younger children. 71,95,99,103 This<br />

requirement for multiple dilatations in children, as<br />

well as the need for monitored general anesthesia,<br />

also decrease the proposed benefit of lower hospital<br />

costs. 104 Given these results, balloon dilatation<br />

plays a minimal role in the treatment of children<br />

with achalasia. Balloon dilatation should be<br />

reserved for patients unwilling to undergo surgery,<br />

or as a treatment adjunct in patients with residual<br />

symptoms after surgery.<br />

Esophagomyotomy<br />

Surgical esophagomyotomy for achalasia was first<br />

performed in Germany by Ernest Heller, and


68<br />

Achalasia<br />

involved a laparotomy with both an anterior and a<br />

posterior esophagomyotomy. 105 The procedure<br />

was modified by Zaaijer in 1923 to utilize only an<br />

anterior esophagomyotomy; 106 all surgery for achalasia<br />

now employs a variant of this procedure. 107<br />

An example of the affected esophageal segment<br />

pre- and post-myotomy is shown in Figure 5.2. A<br />

consensus on the optimal surgical technique<br />

remains to be resolved. Traditional approaches<br />

have been through a standard midline laparotomy<br />

or a left thoracotomy, with equally good<br />

results. 108–111 A review of the literature by<br />

Ferguson showed that, regardless of operative<br />

approach, an open Heller procedure resulted in<br />

(a) (b)<br />

(c) (d)<br />

symptomatic improvement in 89% of patients, a<br />

mortality rate of 0.3%, a reoperative rate of 2.9%<br />

and a postoperative GERD incidence of 10%. 73<br />

The benefits of minimally invasive surgery,<br />

decreased pain, shorter hospital stay, and<br />

improved cosmesis have prompted surgeons to<br />

apply this approach to esophagomyotomy. As with<br />

open surgery, both thoracoscopic and laparoscopic<br />

approaches are in current use, and by nature of<br />

similar results to the open surgery, minimally invasive<br />

procedures are becoming the standard. 112–118<br />

In addition to controversy over whether an abdominal<br />

or thoracic approach is better, there is no<br />

Figure 5.2 (a) Transition between dilated (normal) and narrowed (abnormal) segment of esophageal achalasia prior to<br />

myotomy. (b) Performing myotomy with an endoscopic spreader. (c) Myotomy completed to the gastroesophageal junction.<br />

(d) Completed myotomy including dissection onto the lesser curvature of the stomach (circularis muscle seen distally).


consensus on the benefit of performing a simultaneous<br />

fundoplication. For example, at our institution<br />

we do not routinely perform concomitant<br />

fundoplication. Proponents of a fundoplication<br />

cite increased risk of late GERD, whereas opponents<br />

state that limiting the gastric portion of the<br />

esophagomyotomy to less than 1 cm limits the<br />

incidence of GERD and that a fundoplication may<br />

actually result in pseudoachalasia from making an<br />

over-tight wrap. 73,119–124<br />

The complications of Heller myotomy include<br />

intraoperative perforation (treated by over-sewing<br />

the tear, and buttressing with gastric fundus),<br />

recurrence of symptoms (typically the result of<br />

incomplete myotomy or addition of a fundoplication<br />

that is too tight) and esophageal<br />

leak. 67,70,109,125 Concern exists about performing a<br />

Heller myotomy after previous non-surgical therapies,<br />

as this may incur a higher complication rate.<br />

While previous treatment by balloon dilatations<br />

or BoTox injection seems to result in scar forma-<br />

REFERENCES<br />

1. Spechler SJ, Castell DO. Classification of oesophageal<br />

motility abnormalities. Gut 2001; 49: 145–151.<br />

2. Willis T. Pharmaceutice Rationalis sive Diatriba do<br />

Medicamentorum Operationibus in Humano Corpore.<br />

London: Hagae Comitis, 1674.<br />

3. Hertz AF. The bismuth meal. BMJ 1913; 1: 13.<br />

4. Hertz AF. Achalasia of the cardia. Q J Med 1916; 8:<br />

300–308.<br />

5. Mayberry JF, Probert CSJ, Sher KS et al. Some epidemiological<br />

and aetiological aspects of achalasia. Dig Dis<br />

1991; 9: 1–8.<br />

6. Erlam RJ, Ellis FH, Nobrega FT. Achalasia of the esophagus<br />

in a small urban community. Mayo Clin Proc 1960;<br />

44: 478–483.<br />

7. Galen EA, Switz M, Zfass AM. Achalasia: incidence and<br />

treatment in Virginia. Va Med 1982; 109: 183–186.<br />

8. Mayberry JF, Rhodes J. Achalasia in the city of Cardiff<br />

from 1926 to 1977. Digestion 1980; 20: 248–252.<br />

9. Mayberry JF, Atkinson M. Studies of incidence and<br />

prevalence of achalasia in the Nottingham area. Q J Med<br />

1985; 56: 451–456.<br />

10. Mayberry JF, Atkinson M. Variations in the prevalence<br />

of achalasia over a decade in Great Britain and Ireland:<br />

an epidemiological study based on hospital admissions.<br />

Q J Med 1987; 62: 67–74.<br />

11. Mayberry JF, Mayall M. The epidemiology of achalsia in<br />

children. Gut 1988; 29: 90–93.<br />

Conclusion 69<br />

tion, 126 it does not seem to affect subsequent<br />

esophagomyotomy. 127,128<br />

Conclusion<br />

Achalasia presents a diagnostic challenge in children,<br />

given the varied presentations. High clinical<br />

suspicion is necessary, particularly when treating<br />

infants and young children. The mainstays of<br />

diagnosis are barium swallow and manometry,<br />

each with findings highly suggestive of achalasia.<br />

Once the diagnosis is made, the most efficacious<br />

treatment is surgical esophagomyotomy. As the<br />

operative experience in children progresses,<br />

laparoscopic Heller myotomy emerges as the<br />

surgical treatment of choice, with or without an<br />

added fundoplication. Balloon dilatation or BoTox<br />

injection should be reserved for addressing postoperative<br />

recurrence of dysphagia. Medical treatment<br />

with calcium channel blockers currently has<br />

only very limited application in children.<br />

12. Nihoul-Fekete C, Bawab F, Lortat-Jacob S et al.<br />

Achalasia of the esophagus in childhood. Surgical treatment<br />

in 35 cases, with special reference to familial<br />

cases and glucocorticoid deficiency association. Hepatogastroenterol<br />

1991; 38: 510–513.<br />

13. Allgrove J, Clayden GS, Grant DB et al. Familial glucocorticoid<br />

deficiency with achalasia of the cardia and<br />

deficient tear production, Lancet 1978 26: 1284–1286.<br />

14. Ehrich E, Aranoff G, Johnson WG. Familial achalasia<br />

associated with adrenocortical insufficiency, alacrima<br />

and neurologic abnormalities. Am J Med Genet 1987; 26:<br />

637–644.<br />

15. Stuckey BG, Mastaglia FL, Reed WD et al.<br />

Glucocorticoid insuffiency, achalasia, alacrima with<br />

autonomic and motor neuropathy, Ann Intern Med 1987;<br />

106: 62–64.<br />

16. El-Rayyes K, Hegab S, Besisso M. A syndrome of<br />

alacrima, achalasia and neurologic anomalies without<br />

adrenocortical insufficiency. J Pediatr Ophthalmol<br />

Strabismus 1991; 28: 35–37.<br />

17. Arber N, Grossman A, Lurie B et al. Epidemiology of<br />

achalasia in central Israel. Rarity of esophageal cancer.<br />

Dig Dis Sci 1993; 38: 1920–1925.<br />

18. Sonnenberg A, Massey BT, McCarty DJ et al.<br />

Epidemiology of hospitalization for achalasia in the<br />

United States. Dig Dis Sci 1993; 38: 233–244.


70<br />

Achalasia<br />

19. Zimmerman FH, Rosenweig NS. Achalasia in a father<br />

and son. Am J Gastroenterol 1984; 79: 506–508.<br />

20. Bosher LP, Shaw A. Achalasia in siblings. Clinical and<br />

genetic aspects. Am J Dis Child 1981; 135: 709–710.<br />

21. Murphy MS, Gardner-Medwin D, Eastham EJ. Achalasia<br />

of the cardia associated with hereditary cerebellar<br />

ataxia. Am J Gastroenterol 1989; 84: 1329–1330.<br />

22. Chawla K, Chawla SK, Alexander LL. Familial achalasia<br />

of the esophagus in mother and son: a possible pathogenetic<br />

relationship. J Am Geriatr Soc 1979; 27:<br />

519–521.<br />

23. Kilpatrick ZM, Milles S. Achalasia in mother and<br />

daughter. Gastroenterology 1972; 62: 1042–1046.<br />

24. Mackler D, Schneider R. Achalasia in father and son.<br />

Dig Dis Sci 1978; 23: 1042–1045.<br />

25. Mayberry JF, Atkinson M. A study of swallowing difficulties<br />

in first degree relatives of patients with achalasia.<br />

Thorax 1985; 40: 391–393.<br />

26. Pinotti HW, Felix VN, Zilberstein B et al. Surgical<br />

complications of Chagas’ disease: megaesophagus, achalasia<br />

of the pylorus and cholelithiasis. World J Surg<br />

1991; 15: 198–204.<br />

27. Kirchoff LV. Trypanosoma species (American trypanosomiasis,<br />

Chagas’ disease): biology of trypanosomes. In<br />

Mandell GL, Bennett JE, Dolin R eds. In Principles and<br />

Practice of Infectious Diseases, 5th edn. Philadelphia:<br />

Churchill Livingstone, 2000: 2845-2851.<br />

28. Robertson CS, Martin BA, Atkinson M. Varicella-zoster<br />

virus DNA in the oesophageal myenteric plexus in achalasia.<br />

Gut 1993; 34: 299–302.<br />

29. Jones DB, Mayberry JF, Rhodes J et al. Preliminary<br />

report of an association between measles virus and<br />

achalasia. J Clin Pathol 1983; 36: 655–657.<br />

30. Castex F, Guillemot F, Talbodec N et al. Association of<br />

an attack of varicella and achalasia. Am J Gastroenterol<br />

1995; 90: 1188–1189.<br />

31. Niwatomto H, Okamoto E, Fujimoto J et al. Are human<br />

herpes viruses or measles virus associated with<br />

esophageal achalasia? Dig Dis Sci 1995; 40: 859–864.<br />

32. Goldblum JR, Whyte RI, Orringer MB et al. Achalasia: a<br />

morphologic study of 42 resected specimens. Am J Surg<br />

Pathol 1994; 18: 327–337.<br />

33. Cassella RR, Ellis FH, Brown AL. Fine-structure changes<br />

in achalasia of the esophagus. I: Vagus nerves. Am J<br />

Pathol 1965; 46: 279–283.<br />

34. Csendes A, Smok G, Braghetto I et al. Gastroesophageal<br />

sphincter pressure and histological changes in distal<br />

esophagus in patients with achalasia of the esophagus.<br />

Dig Dis Sci 1985; 30: 941–945.<br />

35. Goldblum JR, Rice TW, Richter JE. Histopathologic<br />

features in esophagomyotomy specimens from patients<br />

with achalasia. Gastroenterology 1996; 111: 648–654.<br />

36. Rake GW. A case of annular muscular hypertrophy of<br />

the esophagus. Guy’s Hosp Rep 1926; 76: 145–158.<br />

37. Cross FS. Pathologic changes in megaesophagus<br />

(esophageal dystonia). Surgery 1952; 31: 647–653.<br />

38. Cassella RR, Brown AL, Sayre GP et al. Achalasia of the<br />

esophagus: pathologic and etiologic considerations. Ann<br />

Surg 1964; 160: 474–487.<br />

39 Cassella RR, Ellis FH, Brown AL. Fine-structure changes<br />

in achalasia of the esophagus. II: Esophageal smooth<br />

muscle. Am J Pathol 1965; 46: 467–475.<br />

40. Qualman SJ, Haupt HM, Yang P et al. Esophageal Lewy<br />

bodies associated with ganglion cell loss in achalasia.<br />

Similarity to Parkinson’s disease. Gastroenterology 1984;<br />

87: 848–856.<br />

41. Holloway RH, Dodds WJ, Helm JF et al. Integrity of<br />

cholinergic innervation to the lower esophageal sphincter<br />

in achalasia. Gastroenterology 1986; 90: 924–929.<br />

42. Goyal RK, Rattan S. Nature of the vagal inhibitory<br />

innervation to the lower esophageal sphincter. J Clin<br />

Invest 1975; 55: 1119–1126.<br />

43. Yuan C, Costa M, Brookes SJH. Neuronal pathways and<br />

transmission to the lower esophageal sphincter of the<br />

guinea pig. Gastroenterology 1998; 118: 661–668.<br />

44. Hornby PJ, Abrahams TP. Central control of lower<br />

esophageal sphincter relaxation. Am J Med 2000; 108:<br />

90S–98S.<br />

45. Alumets J, Schaffalitzky De Muckadell O, Fahrenkrug J<br />

et al. A rich VIP nerve supply is characteristic of<br />

sphincters. Nature 1979; 280: 155–156.<br />

46. Goyal RK, Rattan S, Said SI. VIP as a possible neurotransmitter<br />

of non-cholinergic non-adrenergic inhibitory<br />

neurons. Nature 1980; 288: 378–380.<br />

47. Aggestrup S, Uddman R, Sundler F et al. Lack of<br />

vasoactive intestinal peptide nerves in esophageal achalasia.<br />

Gastroenterology 1983; 84: 924–927.<br />

48. Guelrud M, Rossiter A, Sourney PF et al. The effect of<br />

vasoactive intestinal polypeptide on the lower<br />

esophageal sphincter in achalasia. Gastroenterology<br />

1992; 103: 377–382.<br />

49. Rattan S, Said SI, Goyal RK. Effect of vasoactive intestinal<br />

polypeptide on lower esophageal sphincter pressure.<br />

Proc Soc Exp Bio Med 1977; 155: 40–43.<br />

50. Aimi Y, Kimura H, Kinoshita T et al. Histochemical<br />

localization of nitric oxide synthase in rat enteric<br />

nervous system, Neuroscience 1993; 53: 553–560.<br />

51. Furness JB, Li ZS, Young HM et al. Nitric oxide synthase<br />

in the enteric nervous system of the guinea pig: a quantitative<br />

description. Cell Tissue Res 1994; 277: 139–149.<br />

52. Ward SM, Xue C, Sanders KM. Localization of nitric<br />

oxide synthase immunoreactivity in ileocolonic and<br />

<strong>pylori</strong>c sphincters. Cell Tissue Res 1994; 275: 513–527.<br />

53. Singaram C, Sengupta A, Sweet MA et al. Nitrinergic<br />

and peptidergic innervation of the human oesophagus.<br />

Gut 1994; 35: 1690–1696.<br />

54. Christensen J, Fang S, Rick GA. NADPH-diaphorasepositive<br />

nerve fibers in smooth muscle layers of<br />

opossum esophagus: gradients in density. J Auton Nerv<br />

Sys 1995; 52: 99–105.<br />

55. Lynn RB, Sankey SL, Chakder S et al. Colocalization of<br />

NADPH-diaphorase staining and VIP immunoreactivitiy<br />

in neurons in opossum internal anal sphincter. Dig Dis<br />

Sci 1995; 40: 781–791.<br />

56. Wang XY, Wong WC, Ling EA. Localization of nicotinamide<br />

adenine dinucleotide phosphate diaphorase<br />

(NADPH-d) activity in the gastrointestinal sphincters in<br />

the guinea pig. J Auton Nerv Sys 1996; 58: 51–55.<br />

57. Preiksaitis HG, Tremblay L, Diamant NE. Nitric oxide<br />

mediates inhibitory nerve effects in human esophagus<br />

and lower esophageal sphincter. Dig Dis Sci 1994; 39:<br />

770–775.<br />

58. Conklin JL, Du C, Murray JA et al. Characterization and<br />

mediation of inhibitory junction potentials from<br />

opossum lower esophageal sphincter. Gastroenterology<br />

1993; 104: 1439–1444.<br />

59. Bayguinov O, Sanders KM. Role of nitric oxide as an<br />

inhibitory neurotransmitter in the canine <strong>pylori</strong>c<br />

sphincter. Am J Physiol 1993; 264: G975–G983.<br />

60. Tottrup A, Ny L, Alm P et al. The role of the larginine/nitric<br />

oxide pathway for relaxation of the<br />

human lower oesophageal sphincter. Acta Physiol Scand<br />

1993; 149: 451–459.<br />

61. Huang PL, Dawson TM, Bredt DS et al. Targetted<br />

disruption of the neuronal nitric oxide synthase gene.<br />

Cell 1993; 75: 1273–1286.<br />

62. Mearin F, Mourelle M, Guarner F et al. Patients with<br />

achalasia lack nitric oxide synthase in the gastro-


oesophageal junction. Eur J Clin Invest 1993; 23:<br />

724–728.<br />

63. Ward SM, Morris G, Reese L et al. Interstitial cells of<br />

Cajal mediate enteric inhibitory neurotransmission in<br />

the lower esophageal and <strong>pylori</strong>c sphincters.<br />

Gastroenterology 1998; 115: 314–329.<br />

64. Watanabe Y, Ando H, Seo T et al. Attenuated nitrergic<br />

inhibitory neurotransmission to interstitial cells of Cajal<br />

in the lower esophageal sphincter with esophageal<br />

achalasia in children. Pediatr Int 2002; 44: 145–148.<br />

65. Howard PJ, Maher L, Pryde A et al. Five-year prospective<br />

study of the incidence, clinical features, and diagnosis<br />

of achalasia in Edinburgh. Gut 1992; 33:<br />

1011–1015.<br />

66. Reynolds JC, Parkman HP. Achalasia. Gastroenterol Clin<br />

North Am 1989; 18: 223–255.<br />

67. Birgisson S, Richter JE. Achalasia: what’s new in diagnosis<br />

and treatment? Dig Dis 1997; 15(Suppl 1): 1–27.<br />

68. Couturier D, Samama J. Clinical aspects and monometric<br />

criteria in achalasia. Hepato-gastroenterol 1991; 38:<br />

481–487.<br />

69. Richter JE. Oesophageal motility disorders. Lancet 2001;<br />

358: 823–828.<br />

70. Vane DW, Cosby K, West K et al. Late results following<br />

esophagomyotomy in children with achalasia. J Pediatr<br />

Surg 1988; 23: 515–519.<br />

71. Azizkhan RG, Tapper D, Eraklis A. Achalasia in childhood:<br />

a 20-year experience. J Pediatr Surg 1980; 15:<br />

452–456.<br />

72. Myers NA, Jolley SG, Taylor R. Achalasia of the cardia<br />

in children: a worldwide survey. J Pediatr Surg 1994; 29:<br />

1375–1379.<br />

73. Ferguson MK. Achalasia: current evaluation and<br />

therapy. Ann Thorac Surg 1991; 52: 336–342.<br />

74. Orlando RC, Call DL, Bream CA. Achalasia and absent<br />

gastric air bubble. Ann Intern Med 1978; 88: 60–61.<br />

75. Stewart ET. Radiographic evaluations of the esophagus<br />

and its motor disorders. Med Clin North Am 1981; 65:<br />

1173–1190.<br />

76. Moreno Gonzalez E, Garcia Alvarez A, Landa Garcia I et<br />

al. Results of surgical treatment of esophageal achalasia.<br />

Multicenter retrospective study of 1856 cases. Int Surg<br />

1988; 73: 69–77.<br />

77. Hewson EG, Ott DJ, Dalton CB et al. Manometry and<br />

radiology. Complementary studies in the assessment of<br />

esophageal motility disorders. Gastroenterology 1990;<br />

98: 626–632.<br />

78. Tracey JP, Traube M. Difficulties in the diagnosis of<br />

pseudoachalasia. Am J Gastroenterol 1994; 89:<br />

2014–2018.<br />

79. Vilgrain V, Mompoint D, Palazzio L. Staging of<br />

esophageal carcinoma: comparison of results with endoscopic<br />

sonography and CT. Am J Roentgenol 1990; 155:<br />

277–281.<br />

80. Stacher G, Schima W, Bergmann H et al. Sensitivity of<br />

radionuclide bolus transport and videofluoroscopic<br />

studies compared with manometry in the detection of<br />

achalasia. Am J Gastroenterol 1994; 89: 1484–1488.<br />

81. Cohen S, Lipshutz W. Lower esophageal sphincter<br />

dysfunction in achalasia. Gastroenterology 1971; 61:<br />

814–820.<br />

82. Tack J, Janssens J, Vantrappen G. Non-surgical treatment<br />

of achalasia. Hepato-gastroenterol 1991; 38: 493–497.<br />

83. Lobis JB, Fischer RS. Anticholinergic therapy for achalasia.<br />

A controlled study (abstract). Gastroenterology 1976;<br />

70: 976.<br />

84. Christensen J. Effects of drugs on esophageal motility.<br />

Arch Intern Med 1976; 136: 532–537.<br />

References 71<br />

85. Triadafilopoulos G, Aaronson M, Sackel S et al. Medical<br />

treatment of esophageal achalasia: double-blind<br />

crossover study with oral nifedipine, verapamil and<br />

placebo. Dig Dis Sci 1991; 36: 260–267.<br />

86. Traube M, Dubovik S, Lange RC et al. The role of<br />

nifedipine therapy in achalasia: results of a randomized,<br />

double-blind, placebo-controlled study. Am J<br />

Gastroenterol 1989; 84: 1259–1262.<br />

87. Gelfond M, Rozen P, Keren S et al. Effect of nitrates on<br />

LEOS pressure in achalasia: a potential therapeutic aid.<br />

Gut 1981; 22: 312–318.<br />

88. Gelfond M, Rozen P, Gilat T. Isosorbide dinitrate and<br />

nifedipine treatement of achalasia: a clinical, manometric<br />

and radionuclide evaluation. Gastroenterology 1982;<br />

83: 963–969.<br />

89. Pasricha PJ, Ravich WJ, Hendrix TR et al.<br />

Intrasphincteric botulinum toxin for the treatment of<br />

achalasia. N Engl J Med 1995; 332: 774–778.<br />

90. Pasricha PJ, Rudra R, Ravich WJ et al. Botulinum toxin<br />

for achalasia: long-term outcome and predictors of<br />

response. Gastroenterology 1996; 110: 1410–1415.<br />

91. Neubrand M, Scheurlen C, Schepke M et al. Long-term<br />

results and prognostic factors in the treatment of achalasia<br />

with botulinum toxin. Endoscopy 2002; 34:<br />

519–523.<br />

92. Bansal R, Nostrant TT, Scheiman JM et al.<br />

Intrasphincteric botulinum toxin versus pneumatic<br />

balloon dilation for treatment of primary achalasia. J<br />

Clin Gastroenterol 2003; 36: 209–214.<br />

93. Walton JM, Tougas G. Botulinum toxin use in pediatric<br />

esophageal achalasia: a case report. J Pediatr Surg 1997;<br />

32: 916–917.<br />

94. Hurwitz M, Bahar RJ, Ament ME et al. Evaluation of the<br />

use of botulinum toxin in children with achalasia. J<br />

Pediatr Gastroenterol Nutr 2000; 30: 509–514.<br />

95. Hussain SZ, Thomas R, Tolia V. A review of achalasia in<br />

33 children. Dig Dis Sci 2002; 47: 2538–2543.<br />

96. Eaker EY, Gordon JM, Vogel SB. Untoward effects of<br />

esophageal botulinum toxin injection in the treatment<br />

of achalasia. Dig Dis Sci 1997; 42: 724–727.<br />

97. Barnett JL, Eisenman R, Nostrant TT et al. Witzel pneumatic<br />

dilation for achalasia: safety and long-term efficacy.<br />

Gastrointest Endosc 1990; 36: 482–485.<br />

98. Kadakia SC, Wong RKH. Graded pneumatic dilation<br />

using Rigiflex achalasia dilators in patients with<br />

primary esophageal achalasia. Am J Gastroenterol 1993;<br />

88: 34–38.<br />

99. Csendes A, Braghetto I, Henríquez A et al. Late results<br />

of a prospective randomized study comparing forceful<br />

dilatation and oesophagomyotomy in patients with<br />

achalasia. Gut 1989; 30: 299–304.<br />

100. Eckardt VF, Aignherr C, Bernhard G. Predictors of<br />

outcome in patients with achalasia treated by pneumatic<br />

dilation, Gastroenterology 1992; 103: 1732–1738.<br />

101. Nair, LA, Reynolds JC, Parkman HP et al. Complications<br />

during pneumatic dilation for achalasia or diffuse<br />

esophageal spasm: analysis of risk factors, early clinical<br />

characteristics, and outcome. Dig Dis Sci 1993; 38:<br />

1893–1903.<br />

102. Babu R, Grier D, Cusick E et al. Pneumatic dilatation for<br />

childhood achalasia. Pediatr Surg Int 2001; 17: 505–507.<br />

103. Wilkinson AG, Raine PAM, Fyfe AHB. Pneumatic dilatation<br />

in childhood cardio-achalasia. Pediatr Radiol 1997;<br />

27: 60–62.<br />

104. Parkman HP, Reynolds JC, Ouyang A et al. Pneumatic<br />

dilatation or esophagomyotomy treatment for idiopathic<br />

achalasia: clinical outcomes and cost analysis. Dig Dis<br />

Sci 1993; 38: 75–85.


72<br />

Achalasia<br />

105. Heller E. Extramuköse Kardioplastik beim chronischen<br />

Kardiospasmus mit Dilatation des Oesophagus. Mitt<br />

Grenzgeb Med Chir 1914; 27: 141–149.<br />

106. Zaaijer JN. Cardiospasm in the aged. Ann Surg 1923; 77:<br />

615–617.<br />

107. Crookes PF, DeMeester TR. Esophageal anatomy and<br />

physiology, and esophageal reflux. In Greenfield LJ,<br />

Mulholland MW, Oldham KT et al. eds. Surgery:<br />

Scientific Principles and Practice, 2nd Edn. Philadelphia:<br />

Lippincott-Raven, 1997: 653–694.<br />

108. Ellis FH, Crozier RE, Watkins E. Operation for<br />

esophageal achalasia: results of esophagomyotomy<br />

without an antireflux operation. J Thorac Cardiovasc<br />

Surg 1984; 88: 344–351.<br />

109. Csendes A. Results of surgical treatment of achalasia of<br />

the esophagus. Hepato-gastroenterol 1991; 38: 474–480.<br />

110. Bonavina L, Nosadini A, Bardini R et al. Primary treatment<br />

of esophageal achalasia: long-term results of<br />

myotomy and Dor fundoplication. Arch Surg 1992; 127:<br />

222–227.<br />

111. Mattioli S, DiSimone MP, Bassi F et al. Surgery for<br />

esophageal achalasia. Long-term results with three<br />

different techniques. Hepato-gastroenterol 1996; 43:<br />

492–500.<br />

112. Wang PC, Sharp KW, Holzman MD et al. The outcome<br />

of laparoscopic Heller myotomy without antireflux<br />

procedure in patients with achalasia. Am Surg 1998; 64:<br />

515–521.<br />

113. Pellegrini CA, Leichter R, Patti M et al. Thoracoscopic<br />

esophageal myotomy in the treatment of achalasia. Ann<br />

Thorac Surg 1993; 56: 680–682.<br />

114. Maher JW. Thoracoscopic esophagomyotomy for achalasia:<br />

maximum gain, minimal pain. Surgery 1997; 122:<br />

836–841.<br />

115. Vogt D, Curet M, Pitcher D et al. Successful treatment of<br />

esophageal achalasia with laparoscopic Heller myotomy<br />

and Toupet fundoplication. Am J Surg 1997; 174:<br />

709–714.<br />

116. Hunter JG, Trus TL, Branum GD et al. Laparoscopic<br />

Heller myotomy and fundoplication for achalasia. Ann<br />

Surg 1997; 225: 655–665.<br />

117. Ancona E, Anselmino M, Zaninotto G et al. Esophageal<br />

achalasia: laparoscopic versus conventional open<br />

Heller–Dor operation. Am J Surg 1995; 170: 265–270.<br />

118. Esposito C, Cucchiara S, Borrelli O et al. Laparoscopic<br />

esophagomyotomy for the treatment of achalasia in children.<br />

Surg Endosc 2000; 14: 110–113.<br />

119. Andreollo NA, Earlam RJ. Heller’s myotomy for achalasia:<br />

is an added anti-reflux procedure necessary? Br J<br />

Surg 1987; 74: 765–769.<br />

120. Csendes A, Velasco N, Braghetto I et al. A prospective<br />

randomized study comparing forceful dilatation and<br />

esophagomyotomy in patients with achalasia of the<br />

esophagus. Surgery 1988; 104: 469–475.<br />

121. Anselmino M, Zaninotto G, Costantini M et al. One-year<br />

follow-up after Heller–Dor operation for esophageal<br />

achalasia. Surg Endosc 1997; 11: 3–7.<br />

122. Jamieson GG. Gastro-esophageal reflux following<br />

myotomy for achalasia. Hepato-gastroenterol 1991; 38:<br />

506–509.<br />

123. Chen LW, Chughtai T, Sideris L. Long-term effects of<br />

myotomy and partial fundoplication for esophageal<br />

achalasia. Dis Esophagus 2002; 15: 171–179.<br />

124. Fernández AF, Martínez MA, Ruiz J et al. Six years of<br />

experience in laparoscopic surgery of esophageal achalasia.<br />

Surg Endosc 2003; 17: 153–156.<br />

125. Zaninotto G, Costantini M, <strong>Portal</strong>e G et al. Etiology,<br />

diagnosis and treatment of failures after laparoscopic<br />

Heller myotomy for achalasia. Ann Surg 2002; 235:<br />

186–192.<br />

126. Richardson WS, Willis GW, Smith JW. Evaluation of scar<br />

formation after botulinum toxin injection or forced<br />

balloon dilation to the lower esophageal sphincter. Surg<br />

Endosc 2003; 17: 696–698.<br />

127. Ferguson MK, Reeder LB, Olak J. Results of myotomy<br />

and partial fundoplication after pneumatic dilation for<br />

achalasia. Ann Thorac Surg 1996; 62: 327–330.<br />

128. Cosentini E, Berlakovich G, Zacherl J. Achalasia: results<br />

of myotomy and antireflux operation after failed dilatations.<br />

Arch Surg 1997; 132: 143–147.


6<br />

Introduction<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis<br />

and peptic ulcer disease<br />

Costantino De Giacomo<br />

In adults, peptic disease (PD) is an important<br />

public health problem because of its clinical and<br />

economic implications. In children it is a wellknown<br />

problem, but its prevalence and relative<br />

complications are less well defined.<br />

PD includes some gastric and duodenal diseases<br />

characterized by the presence of the digestive<br />

symptoms of dyspepsia, the endoscopic appearance<br />

of mucosal lesions, and the histological<br />

features of chronic inflammation. However, these<br />

three characteristics might be expressed in different<br />

ways, producing a mix of clinical pictures,<br />

which range from the asymptomatic patient with<br />

normal endoscopic picture and mild gastritis, to<br />

the patient with a bleeding peptic ulcer.<br />

Gastritis, duodenitis, gastric and duodenal ulcer<br />

are the definitive diagnoses of a patient who has<br />

gone through the appropriate endoscopic and<br />

histological investigation of the upper gastrointestinal<br />

tract. This means that most of the findings<br />

belonging to the pre-endoscopic era (in children,<br />

before 1980) are not reliable and not<br />

comparable with more recent data.<br />

The strict link between the above-mentioned<br />

pictures is based on two major pieces of evidence.<br />

First, is the presence of mucosal inflammation and<br />

ulcer in the same patient. Even if gastroduodenitis<br />

and ulcer could be seen as a continuum in the<br />

natural history of PD, the evolution toward the<br />

crater (ulcer) is not mandatory, and the majority of<br />

patients with PD show only gastritis. Second, is<br />

the existence of a common etiological factor of<br />

both gastritis and ulcer in the majority of patients:<br />

the <strong>Helicobacter</strong> <strong>pylori</strong> (H. <strong>pylori</strong>) infection.<br />

The history of the H. <strong>pylori</strong> infection begins in<br />

1982, and it is one of the most exciting examples of<br />

how a fortuitous observation has produced one of<br />

the greatest impacts on our knowledge and treatment<br />

of an old disease. The oversight of cultures of<br />

antral biopsies taken from patients with PD during<br />

some days of vacation allowed two Australian<br />

investigators, Barry Marshall and Robert Warren,<br />

to observe and describe the growth of colonies of<br />

previously unknown bacteria. 1 As these Gramnegative,<br />

spiral bacteria had been isolated on<br />

Campylobacter-specific media, they were initially<br />

identified as Campylobacter-like organisms, but it<br />

soon became clear that H. <strong>pylori</strong> belonged to a new<br />

genus, taxonomically identified as <strong>Helicobacter</strong>,<br />

which, to date, consists of 19 zoonotic species with<br />

another ten potentially novel species. The official<br />

species differ in site of infection, which is the<br />

stomach in eight and the gut in 11, and in the host,<br />

but they show great homology in the analysis of<br />

165 rRNA sequences (Table 6.1). 2<br />

During the past two decades, an impressive<br />

number of papers, consensus conferences, position<br />

documents and statements of the<br />

Gastroenterological Associations and Public<br />

Health Organizations (NIH) confirmed the existence<br />

of a strong association between H. <strong>pylori</strong> and<br />

PD. In particular, in 1994 a Consensus Conference<br />

of the NIH established the role of H. <strong>pylori</strong> in the<br />

pathogenesis and recurrence of peptic ulcer,<br />

emphasizing that the treatment of this condition<br />

should be considered the eradication of the H.<br />

<strong>pylori</strong> infection and not only the healing of the<br />

ulcer. 3 In the same year, the International Agency<br />

for Research on Cancer included H. <strong>pylori</strong> in the<br />

list of the carcinogenic factors of class I, as the<br />

main developing factor in gastric carcinoma. 4<br />

73


74<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

Table 6.1 Natural hosts and usual site of isolation of <strong>Helicobacter</strong><br />

species<br />

<strong>Helicobacter</strong> species Main host Origin<br />

H. <strong>pylori</strong> human gastric<br />

H. mustelae ferret gastric<br />

H. nemestrinae macaque monkey gastric<br />

H. felis cat, dog gastric<br />

H. acinonychis cheetah gastric<br />

H. bizzozeronii dog gastric<br />

H. salomonis dog gastric<br />

H. heilmannii human, cat, dog, pig gastric<br />

H. cinaedi human, hamster intestinal<br />

H. fennelliae human, hamster intestinal<br />

H. muridarum rat, mouse intestinal<br />

H. canis dog intestinal<br />

H. pullorum poultry intestinal<br />

H. pametensis tern intestinal<br />

H. hepaticus mouse intestinal<br />

H. bilis mouse intestinal<br />

H. cholecystus hamster intestinal<br />

H. trogontum rat intestinal<br />

H. rodentium mouse intestinal<br />

Epidemiology<br />

Epidemiological data about peptic ulcer disease<br />

(PUD) are scanty and often related to series investigated<br />

by X-rays. Endoscopic data appeared after<br />

1980 and showed a three-fold increase of cases of<br />

PUD over earlier findings. 5 A review of more than<br />

1000 cases stated that the expected number of new<br />

PUD cases in a large children’s hospital was evaluated<br />

as 3.5 per year. 6 Others found that the incidence<br />

of PUD was 4.4/10000 children with a<br />

frequency of 3–6% in children who had undergone<br />

gastroscopy. 7<br />

However, there is now evidence that the incidence<br />

of PUD is decreasing in adults as well as in children.<br />

The progressive decrease of PUD is probably<br />

related to modifications in the epidemiology of H.<br />

<strong>pylori</strong> infection. The slow but continuous improve-<br />

ment of socioeconomic conditions, the diffuse use<br />

of antibiotics, and the decrease of the so-called<br />

ulcerogenic strains of H. <strong>pylori</strong> might reasonably<br />

explain this phenomenon.<br />

H. <strong>pylori</strong> infection is one of the most diffuse infections<br />

in the world. There are areas at high risk of<br />

infection (in developing countries, such as those in<br />

the African or South American continents) and<br />

areas at low risk (such as Europe and North<br />

America). 8 In high-risk countries, the rate of infection<br />

is very high in the first 2 years of life, and<br />

older children and adults are almost totally<br />

infected. In a study on a cohort of 248 Gambian<br />

children aged 3–45 months, the prevalence of positive<br />

breath tests (a specific and non-invasive test to<br />

detect H. <strong>pylori</strong> infection, see below) rose from<br />

19% at 3 months of age to 84% by the age of 30<br />

months. Reversion to a negative breath test, in


association with declining specific antibody<br />

levels, occurred in 20% of children, suggesting<br />

that in the first years of life children may acquire<br />

and clear the infection before ultimately being<br />

infected with a persistent strain. 9<br />

In developed countries, transverse serological<br />

studies have demonstrated that the rate of infection<br />

increases slowly and regularly by 10% with<br />

each decade of life. 8 In Italy, a recent epidemiological<br />

study showed that 11% of subjects from 6 to<br />

18 years were infected, 10 while the prevalence rate<br />

in people of 50–60 years was 50–60%.<br />

Longitudinal studies demonstrated that this<br />

increase of infected subjects with aging was<br />

related to an effective decrease of infection in the<br />

youngest cohorts, rather than to an increase of new<br />

cases with aging. 11 This phenomenon is explained<br />

by the fact that each generation or cohort has a<br />

distinct and possibly unique environmental risk of<br />

exposure to the infection (cohort effect), linked to<br />

specific risk factors. These risk factors are well<br />

known and are socioeconomic, so that the cohort<br />

effect is considered to be the consequence of the<br />

improvement of socioeconomic conditions in the<br />

most recent decades. In Japan, a country that has<br />

had an impressive and fast economic development<br />

after the Second World War, epidemiological<br />

studies demonstrated a high prevalence rate of H.<br />

<strong>pylori</strong> infection in subjects born before the war,<br />

and a dramatic decrease in those born after the war<br />

and the industrial revolution. 12<br />

When the improvement of socioeconomic conditions<br />

was slow, but constant, as in the<br />

Netherlands, the decrease of the infection rate in<br />

childhood was continuous. 13<br />

The relationship between socioeconomic status<br />

and H. <strong>pylori</strong> infection has been emphasized by<br />

studies in subjects belonging to the same city or<br />

community, but with different socioeconomic<br />

conditions. Using family income as a measure of<br />

the socioeconomic class, the rate of acquisition of<br />

H. <strong>pylori</strong> in those children with family income less<br />

than $5000/year was twice that of those with<br />

incomes greater than $75 000/year in a study from<br />

Arkansas. 14 Absence of a fixed hot-water supply<br />

and domestic crowding in childhood were powerful<br />

independent risk factors for current infection<br />

with H. <strong>pylori</strong>. Among current living conditions,<br />

only the number of children living in the house-<br />

Epidemiology 75<br />

hold was independently associated with H. <strong>pylori</strong><br />

infection. 15 Different studies showed that the age<br />

of infection is in the first years of life. 16–18<br />

In a cohort of 224 children followed up retrospectively<br />

for 21 years by stored serum samples, the<br />

crude incidence rate per year was 1.4% for the<br />

whole cohort, ranging from 2.1% at 4–5 years and<br />

1.5% at age 7–9 years to 0.3% at 21–23 years. The<br />

median age for seroconversion was 7.5 years. Nine<br />

of the 58 seropositive children cleared the infection<br />

during follow-up. The rate of seroreversion<br />

per year was 1.1%; it was highest among children<br />

at age 4–5 years (2.2% vs 0.2% at ages 18–19). 16<br />

The finding that children infected before 5 years of<br />

age could eradicate the infection spontaneously, as<br />

well as be re-infected more easily than older children,<br />

has been confirmed by others. 14,17 A study of<br />

follow-up of 52 children successfully treated for H.<br />

<strong>pylori</strong> demonstrated that 11.5% of children who<br />

had eradicated the bacterium suffered reinfection,<br />

with the majority under 5 years of age. Only 4.3%<br />

of children older than 5 years were re-infected,<br />

even those living with H. <strong>pylori</strong>-positive parent in<br />

81% of cases. The age of < 5 years was the major<br />

risk factor for re-infection. 17 In adults, the infection<br />

rate is approximately 0.3–0.5% per year and<br />

infection is rarely eradicated spontaneously, as<br />

demonstrated by longitudinal studies on stored<br />

samples. 19,20<br />

In addition to socioeconomic factors, genetic<br />

factors could play a role in the acquisition of the<br />

infection. A sibling study showed that the seroprevalence<br />

rate was similar in genetically identical<br />

twins living under different socioeconomic<br />

circumstances. 21 It should be acknowledged,<br />

however, that ethnic, genetic and socioeconomic<br />

factors are often intertwined and their various<br />

contributions are difficult to assess.<br />

An important issue in the study of an infectious<br />

disease is its route of transmission. The absence of<br />

an animal reservoir, the demonstration of intrafamilial<br />

clustering, 22 as well as clustering of the<br />

infection among institutionalized children 23<br />

suggests that person-to-person transmission is the<br />

most probable route. Approximately 90% of<br />

parents and 70% of siblings of H. <strong>pylori</strong>-positive<br />

children showed seropositivity for H. <strong>pylori</strong>. 22 In<br />

institutionalized children, including those living<br />

in a monastery, an inverse relationship between


76<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

seroprevalence and age was demonstrated, with<br />

the increased rate of infection correlating with the<br />

age of entry more than the length of stay in the<br />

community. 23 Both vertical transmission (from<br />

mother to child as well as from father to child),<br />

and horizontal transmission from sibling to<br />

sibling, have been demonstrated. 24<br />

H. <strong>pylori</strong> has been isolated from saliva and dental<br />

plaque, 25 as well as from vomitus, 26 suggesting<br />

that the most diffuse routes of transmission<br />

between siblings or children in communities could<br />

be the oro-oral or gastro-oral. Epidemics of vomiting<br />

could easily diffuse the infection.<br />

In developing countries, H. <strong>pylori</strong> has been identified<br />

in water 27 and in feces 28 by studies using the<br />

polymerase chain reaction (PCR). Clusters of infection<br />

independent of income, but strictly related to<br />

water source, have been demonstrated; the infection<br />

rate was 12-fold higher in children from highincome<br />

families using community wells rather<br />

than municipal water in a study in Peru. 27 The<br />

fecal-oral route of transmission typical of waterborn<br />

diseases is quite possible, particularly in<br />

conditions where there is a lack of a municipal<br />

water supply.<br />

Etiology<br />

PUD is the progressive loss of the mucosal<br />

integrity up to the formation of a crater (ulcer).<br />

This is the final process of different etiological<br />

factors, which produce lesions by different mechanisms.<br />

These factors and mechanisms are not<br />

mutually exclusive and may co-operate to bring<br />

about their damage. The classical pathogenic<br />

theory was that an ulcer develops when the equilibrium<br />

is lost between defensive factors and<br />

aggressive factors. Defensive factors are the mucus<br />

layer, constituted by glycoproteins, bicarbonate<br />

and prostaglandins; and the gastric mucosa barrier,<br />

constituted by the epithelial layer, submucosal<br />

tissue and microcirculation. The best known<br />

aggressive factors are gastric acid, pepsins,<br />

gastrolesional substances, such as alcohol, drugs,<br />

non-steroidal anti-inflammatory drugs (NSAIDs)<br />

and, last to be identified, H. <strong>pylori</strong> infection). In<br />

primary ulcers (deep erosive lesions not secondary<br />

to other known causes) the most important factor<br />

was considered to be the acid output of the<br />

stomach. Both research and therapeutic interventional<br />

studies were directed towards evaluating<br />

and suppressing acid secretion (according to the<br />

theory ‘no acid, no ulcer’) until the discovery of H.<br />

<strong>pylori</strong>. The demonstration that duodenal ulcer is<br />

almost invariably associated with H. <strong>pylori</strong> infection,<br />

and recurrence depends on its persistence,<br />

established the importance of H. <strong>pylori</strong> as the main<br />

etiological factor in PUD. 3,29 Indeed, H. <strong>pylori</strong><br />

infection has been demonstrated in 92%<br />

(33–100%) of children with duodenal ulcer in 14<br />

studies, and in 25% (11–75%) of children with<br />

gastric ulcer in four studies. 30 Recurrence of ulcer<br />

after healing has been reported in 47% of children<br />

where <strong>Helicobacter</strong> was not eradicated, but it is<br />

rare, if not exceptional, in patients in whom eradication<br />

was achieved. 31 H. <strong>pylori</strong> is the main known<br />

etiological factor in chronic gastritis in adults as<br />

well as in children. 30<br />

H. <strong>pylori</strong> is a Gram-negative, unipolar flagellated<br />

bacterium with a spiral appearance (Figure 6.1). It<br />

was the first recognized bacterium of a new genus<br />

whose different species colonize the gastrointestinal<br />

tract of the human (H. <strong>pylori</strong> and H. heilmannii)<br />

and animals (all remaining species) (Table<br />

6.1). 2<br />

Figure 6.1 Electron micrograph of <strong>Helicobacter</strong> <strong>pylori</strong><br />

attached to the cell membrane of gastric epithelial cells<br />

with evident cytoplasmic vacuolization.


H. <strong>pylori</strong> colonizes only the gastric mucosa, wherever<br />

it may be present: in the stomach or in heterotopic<br />

areas, as in Barrett’s esophagus or in gastric<br />

metaplasia of the duodenum. 2 H. heilmannii is a<br />

rare (0.5% prevalence in humans) cause of dyspeptic<br />

symptoms and inflammation in adults, and its<br />

pathogenic role in children seems limited. 32<br />

The H. <strong>pylori</strong> genome (1.65 million bp) codes for<br />

about 1500 proteins and shows an extraordinary<br />

heterogeneity, 33 which could in part explain the<br />

wide spectrum of clinical pictures. Multiple<br />

strains may be present in the same patient, particularly<br />

in developing countries. Recombination and<br />

mutation between strains produces continuous<br />

changes of the genome during long-term gastric<br />

colonization in the same host. 34<br />

Pathogenesis<br />

Role of acid and pepsinogen secretion<br />

The bulk of our knowledge on acid secretion is<br />

derived from studies in adults. Gastric acid secretion<br />

begins in the newborn from the first day of<br />

life; it is not sensitive to gastrin, but rather to<br />

pentagastrin stimulation, which is the best way to<br />

evaluate it. Serum gastrin is usually elevated in the<br />

newborn period. 35,36<br />

Maximal acid output after pentagastrin stimulation<br />

increases from 0.031 mEq/kg per h at 1 month<br />

to 0.122 mEq/kg per h at 3 months, and up to<br />

0.218 mEq/kg per h at 20 months, reflecting the<br />

increase of the mass of parietal cells. 37 In another<br />

Table 6.2 Causes of gastric acid hypersecretion<br />

Zollinger–Ellison syndrome<br />

Antral G-cell hyperplasia or hyperfunction<br />

Mastocytosis<br />

Chronic renal failure<br />

Hypertrophic gastropathy<br />

Small intestinal resection – short gut<br />

Hyperparathyroidism<br />

Pathogenesis 77<br />

study, maximal acid output values were demonstrated<br />

not to change from 3 months to 12 years of<br />

life. 38<br />

Basal acid output has been found to be normal in<br />

children with PUD in all 39–41 but one study. 42<br />

Although children with PUD have been shown to<br />

have a higher average maximal acid output<br />

compared to age-matched controls, the occasional<br />

child with peptic ulcer may well have a<br />

completely normal maximal acid output.<br />

Similarly, control children may occasionally show<br />

increased maximal acid output. 39–42 This important<br />

overlap of values between children with or<br />

without ulcer is similar to that reported in adults.<br />

Interestingly, children with more severe PUD<br />

(requiring surgery, or presenting severe digestive<br />

bleeding) have been reported to have values of<br />

maximal acid output significantly higher than<br />

those with milder forms of disease or in remission.<br />

40,42 Basal 41 or meal-stimulated serum<br />

gastrin 43 can be normal or slightly increased in<br />

children with ulcer.<br />

Other disorders associated with severe gastric and<br />

duodenal ulcers secondary to increased gastric<br />

acid secretion are listed in Table 6.2. These situations<br />

are usually the consequence of an increased<br />

basal (Zollinger–Ellison syndrome, ‘ZES’) or stimulated<br />

(pseudo-ZES or antral G-cell hyperplasia/<br />

hyperfunction syndrome) 44 gastrin or histamine<br />

secretion.<br />

Pepsins are proteolytic enzymes generated by the<br />

action of hydrochloric acid on their precursor<br />

pepsinogens. Pepsinogens are secreted by the<br />

principal cells of the gastric fundus (PGI) and by


78<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

the mucous cells of the remaining stomach and<br />

part of the duodenum (PGII). Seven molecular<br />

forms (Pg1–Pg5 for PGI and Pg6–Pg7 for PGII) can<br />

be identified electrophoretically. Only traces of<br />

pepsins have been demonstrated in neonatal<br />

gastric juice, and their increase with age is slow. 37<br />

In children with PUD, as well as in members of<br />

families with a high incidence of PUD, an increase<br />

of serum pepsinogen I has been demonstrated, and<br />

high serum pepsinogen I has been considered a<br />

marker of ulcerogenicity. 45<br />

<strong>Helicobacter</strong> <strong>pylori</strong>-related mechanisms<br />

H. <strong>pylori</strong> may induce inflammation by two different<br />

pathways: direct toxicity mediated by secretion<br />

of specific toxins and other aggressive factors<br />

(Table 6.3); and immune-mediated toxicity secondary<br />

to stimulation of both innate and adaptive<br />

immune responses in the host. 46<br />

The gastric mucosa is a niche relatively resistant to<br />

bacterial infection. The main protective mechanism<br />

is the bactericidal activity of the gastric acid.<br />

However, H. <strong>pylori</strong> has developed some mechanisms<br />

to evade protection and to allow the colonization<br />

of the mucosa: urease production and<br />

motility. Urease activity is mediated by a unique<br />

pH-dependent urea channel, UreI, which works at<br />

low pH, and allows the influx of urea followed by<br />

hydrolysis into CO2 and ammonia, which buffers<br />

acid conditions close to the bacterium. 47,48 By<br />

means of flagellar motility, H. <strong>pylori</strong> swims into the<br />

mucous layer, and attaches to epithelial cells by<br />

multiple bacterial-surface adhesins. The best characterized<br />

adhesin is Baba, a 78-kDa outer<br />

membrane protein, which binds to the fucosylated<br />

Lewis B blood-group antigen. 49<br />

Table 6.3 Virulence factors demonstrated in <strong>Helicobacter</strong> <strong>pylori</strong> strains<br />

Colonization factors Toxicity factors<br />

Approximately half of the genomic variants of H.<br />

<strong>pylori</strong> are able to produce the thermolabile 95-kDa<br />

cytotoxin, called Vac-A, with a cytopathic effect<br />

induced by direct vacuolating action on epithelial<br />

cells (Figure 6.1). 50 The toxin inserts itself into the<br />

epithelial cell membrane, forming a channel<br />

through which bicarbonate and organic anions can<br />

reach the bacterium. Other mechanisms involved<br />

in the Vac-A-mediated epithelial damage are the<br />

release of cytochrome C from mitochondria, and<br />

apoptosis. 51 Even if the pathogenic role of Vac A is<br />

still under debate, it is relevant that Vac-A positive<br />

strains are isolated in 60% of patients with PUD,<br />

and in only 30% of patients with gastritis. 52<br />

Sixty-five to eighty per cent of strains of H. <strong>pylori</strong><br />

have a chromosomal region called cag-PAI (37-kb<br />

genomic fragment containing 29 genes) which<br />

codifies a type IV secretory apparatus translocating<br />

the 120–128-kDa Cag-A protein inside the host<br />

cell. 53,54 At the cellular level, a direct proliferative<br />

response and cytokine production are induced.<br />

Cag-A-positive strains are usually isolated from<br />

80–100% of patients with more severe peptic<br />

lesions and in 60–75% of patients with milder<br />

forms of disease.<br />

Even if the majority of strains isolated from<br />

patients with PUD are positive for both Cag-A and<br />

Vac-A, it is not possible to distinguish ulcerogenic<br />

from non-ulcerogenic strains with certainty.<br />

The second pathway involved in gastric injury is<br />

secondary to activation of an immune response.<br />

H. <strong>pylori</strong> produces gastric inflammation in virtually<br />

all infected persons. 55 This inflammatory<br />

response is triggered by bacterial attachment to<br />

epithelial cells, followed by recruitment of<br />

neutrophils, macrophages, T and B lymphocytes,<br />

Urease (acidity inhibition) vacuolizing cytotoxin (Vac-A)<br />

Flagellins (motility) cag pathogenicity island (cag-PAI)<br />

Adhesins (adhesivity)<br />

Catalase, superoxide dismutase (phagocytosis resistance)<br />

ammonium, lipopolysaccharide, phospholipase


and plasma cells. In the inflamed gastric epithelium,<br />

a high concentration of proinflammatory<br />

cytokines has been found. Increased production<br />

of interleukin (IL)-8, a potent neutrophil-activating<br />

cytokine, is induced by activation of NFKβ<br />

from Cag-A-positive, more than Cag-A-negative,<br />

strains. 56 IL-8 and other chemokines amplify the<br />

immune as well as the inflammatory response,<br />

producing severe epithelial damage. The multiplicity<br />

of possible pathogenic mechanisms is<br />

shown in Figure 6.2.<br />

A systemic and mucosal immune response is<br />

elicited by H. <strong>pylori</strong> infection. 57 This does not lead<br />

to eradication, but it may contribute to further<br />

damage. Immunological studies have demon-<br />

Pathogenesis 79<br />

strated that an IL-18-driven Th1 immune<br />

response, instead of the expected Th2, combined<br />

with Fas-mediated apoptosis of H. <strong>pylori</strong>-specific<br />

T-cell clones, may contribute to the persistence of<br />

the infection. Some patients produce autoantibodies<br />

against the H + /K + ATPase of gastric parietal<br />

cells that play a role in the induction of gastric<br />

atrophy. 58<br />

In addition to the direct and inflammatory mechanisms<br />

of mucosal injury, H. <strong>pylori</strong> may play an<br />

ulcerogenic role by interfering with acid and<br />

pepsin secretion. In children, H. <strong>pylori</strong> infection<br />

seems to increase serum PGI 59 and in some, but<br />

not all, children with H. <strong>pylori</strong>, a meal-induced<br />

hypergastrinemia secondary to G-cell hyperplasia<br />

Figure 6.2 <strong>Helicobacter</strong> <strong>pylori</strong>-host interactions in the pathogenesis of mucosal lesions from reference 46, with<br />

permission.


80<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

has been reported. Eradication of H. <strong>pylori</strong> usually<br />

normalizes pepsinogen and gastrin secretion 59 as<br />

well as G- and D-cell hyperplasia. 60 H. <strong>pylori</strong><br />

infection may interfere in the gastrin–HCl axis<br />

increasing gastric acid secretion, which in turn<br />

results in duodenal ulceration: in adults, an<br />

increased maximal acid output has been demonstrated<br />

to differentiate ulcer from non-ulcer H.<br />

<strong>pylori</strong>-positive patients. 61<br />

Clinical aspects<br />

Peptic ulcers are classified as either primary,<br />

when they occur in the absence of an underlying<br />

systemic disease, or secondary, when they are<br />

caused by medications or other diseases. Acute<br />

secondary (stress ulcers), which represent the<br />

majority of PUD during infancy and early childhood,<br />

occur in association with shock, burns,<br />

surgery, sepsis, or intracranial hypertension<br />

(Cushing’s ulcers). 62 Chronic PUD secondary to<br />

diseases which produce an increase of gastric acid<br />

secretion (Table 6.2) are rare at any age.<br />

Since its discovery, many clinical pictures have<br />

been attributed to H. <strong>pylori</strong> infection. Table 6.4<br />

shows the main conditions associated with H.<br />

<strong>pylori</strong> according to evidence-based criteria. By<br />

comparison of the results from different studies, it<br />

appears that there are two main populations of<br />

children with PUD: the first, predominantly<br />

constituted by females younger than 8 years of<br />

age, usually has a gastric localization of the<br />

ulcer, 6,7 which is not associated with H. <strong>pylori</strong> and<br />

rarely proceeds to relapse; the second, more<br />

similar to the adult PUD, is mainly found in males<br />

older than 8–10 years and shows an H. <strong>pylori</strong>associated,<br />

highly relapsing, bulbar ulcer.<br />

Symptoms depend strictly on the age of the<br />

subjects. 6,62,63 In infancy and in early childhood,<br />

PUD is characterized by vomiting and/or digestive<br />

bleeding. Children with ulcer may be referred for<br />

abdominal pain and/or vomiting. Epigastric localization,<br />

nocturnal pain, and meal or antacid relief<br />

of pain, are typical of so-called ‘ulcer-like’ dyspepsia,<br />

and might frequently be reported by older<br />

subjects. Hematemesis, weight loss and other<br />

alarm signs should alert the physician and<br />

strongly suggest further evaluation. 62<br />

Table 6.4 Human digestive diseases<br />

associated with <strong>Helicobacter</strong> <strong>pylori</strong> infection<br />

Acute gastritis<br />

Chronic gastritis<br />

Duodenal ulcer<br />

Gastric ulcer<br />

Gastric carcinoma<br />

B-Lymphoma (MALToma)<br />

Children with H. <strong>pylori</strong>-associated gastritis<br />

without ulcer are asymptomatic in the majority of<br />

cases 10 or, rarely, they may suffer from the same<br />

dyspeptic symptoms as patients with ulcer. No<br />

clinical picture has been found to be specific for H.<br />

<strong>pylori</strong>-associated gastritis.<br />

The clinical picture of recurrent abdominal pain<br />

(RAP) was first described over four decades ago by<br />

Apley and Naish. 64 They found RAP, defined as at<br />

least three bouts of abdominal pain, severe enough<br />

to affect the child’s activity, over a period of not<br />

less than 3 months, in 10.8% of 1000 school-age<br />

children. 64 The etiology, based on patient history<br />

and clinical grounds, was attributed to social and<br />

familial environment stress rather than to an<br />

organic disease. This generic definition served<br />

pediatricians for many years, but the development<br />

of new techniques (ultrasound, endoscopy, motility<br />

probes) and new acquisitions (identification of<br />

H. <strong>pylori</strong>, the role of motility disorders, etc.) ultimately<br />

proved that RAP is a description and not a<br />

single, homogeneous diagnosis 65 (see also Chapter<br />

7).<br />

Is H. <strong>pylori</strong> gastritis a cause of RAP? In a very large<br />

pediatric series, abdominal pain occurred in 90%<br />

of 110 cases of duodenal ulceration, and it was the<br />

main presenting feature in 88% of them. 63 Since<br />

1986, when H. <strong>pylori</strong> infection was first described<br />

in pediatric patients, 66 many endoscopic series<br />

supported the association between the infection<br />

and gastroduodenal pathology in children. 67–69<br />

However, abdominal pain was not more frequent<br />

in H. <strong>pylori</strong>-positive than in H. <strong>pylori</strong>-negative children<br />

submitted to endoscopy. 70–72 In addition,<br />

eradication did not resolve symptoms in all cases,


eing very effective only in those rare cases with<br />

ulcer. 73<br />

Moreover, population-based studies failed to show<br />

any association between RAP and H. <strong>pylori</strong> infection<br />

in both school-aged and pre-school-aged<br />

populations. 10,74,75 Nevertheless, it is evident that<br />

discordant results may also be dependent on the<br />

heterogeneity of the clinical populations studied<br />

(endoscopic series vs. school populations), and the<br />

differences in the inclusion criteria (age, duration<br />

of the pain, etc.) 30<br />

A recent report from the Committee on Childhood<br />

Functional Gastrointestinal Disorders 76 states that<br />

‘it seemed more appropriate to apply the most<br />

specific diagnostic category to a symptomatic<br />

child’, defining clinical criteria of functional<br />

dyspepsia as in adults. Gastroenterologists<br />

working on adults with upper gastrointestinal<br />

symptoms prefer to aggregate more symptoms in<br />

the complex picture of ulcer-like and dysmotilitylike<br />

forms of dyspepsia. Recent studies have<br />

shown that a relationship between dyspeptic<br />

symptoms and some pathogenic factors (mainly H.<br />

<strong>pylori</strong> infection and motility disturbances) is<br />

apparent. 77 However, a large intervention study on<br />

adults failed to demonstrate that symptoms are<br />

dependent on H. <strong>pylori</strong> infection and, by extrapolation,<br />

on gastritis. 78 In the eradication trials, the<br />

summary odds ratio for improvement in dyspeptic<br />

symptoms in patients with non-ulcer dyspepsia in<br />

whom H. <strong>pylori</strong> was eradicated was 1.9 (1.3–2.6).<br />

The presence of severe epigastric pain, associated<br />

with nocturnal pain, fasting pain and relief of pain<br />

after meal intake, characterizing the picture of<br />

ulcer-like dyspepsia in subjects aged over 10<br />

years, 10 could be the clinical picture more suggestive<br />

of gastroduodenitis, suggesting the need for<br />

further evaluation.<br />

Complications<br />

The most frequent complication of PUD is digestive<br />

bleeding, with hematemesis and melena<br />

reported in more than half the cases. 6<br />

Gastrointestinal bleeding may be present even in<br />

the absence of ulcer, if diffuse varioliform gastritis<br />

with erosions is present at endoscopy. Another<br />

complication of PUD is perforation (10%), which<br />

Complications 81<br />

may result in peritonitis, if anterior, or in pancreatic<br />

penetration, if posterior. Pyloric stenosis as a<br />

consequence of a para<strong>pylori</strong>c ulcer is quite rare; a<br />

malignant ulcer is exceptional in children.<br />

A vicious cycle between chronic diarrhea, malnutrition<br />

and H. <strong>pylori</strong> infection is evident, especially<br />

for children in developing countries. 79 Some other<br />

gastrointestinal disorders based on the presence of<br />

heterotopic gastric mucosa, such as Barrett’s<br />

ulcer 80 and Meckel’s diverticulum bleeding, 81 have<br />

been anecdotally associated with H. <strong>pylori</strong> infection,<br />

but further studies suggested that H. <strong>pylori</strong><br />

did not play a causal role in their determination.<br />

Extraintestinal manifestations have been controversially<br />

reported in patients with H. <strong>pylori</strong> infection;<br />

iron deficiency and sideropenic refractory<br />

anemia, short stature and growth failure, and<br />

sudden infant death are the most conflicting areas<br />

in childhood. 82<br />

Iron deficiency anemia may be a consequence of<br />

H. <strong>pylori</strong> infection in children. Case reports and<br />

series of children with sideropenic refractory<br />

anemia have been reported. 83 Iron deficiency<br />

without anemia has also been demonstrated in<br />

adults. 84 The efficacy of eradication in raising the<br />

hemoglobin level and in restoring ferritin values<br />

has been demonstrated in adults as well as in<br />

teenagers. 85 Potential mechanisms involved in<br />

producing iron deficiency are fecal occult blood<br />

loss, reduction in duodenal absorption and the<br />

iron-scavenging capability of the bacterium.<br />

Some studies have reported the existence of an<br />

effect of the infection on the final height of the<br />

patient. H. <strong>pylori</strong>-positive children have been<br />

demonstrated to be shorter than those who are H.<br />

<strong>pylori</strong>-negative. 27 Growth velocity has been<br />

demonstrated to be decreased by 1.1 cm in affected<br />

females between 7 and 11 years of age. 86 However,<br />

this association has not been confirmed by other<br />

studies, which showed, instead, that reduced<br />

growth is related to genetic determinants, such as<br />

parental height, and to mixed genetic and environmental<br />

factors, such as birth weight. Low socioeconomic<br />

status was clearly relevant. 87<br />

Concerning the role of <strong>Helicobacter</strong> infection in<br />

causing infant deaths by sudden infant death<br />

syndrome (SIDS), one study detected H. <strong>pylori</strong> in<br />

lungs from 25 out of 32 SIDS cases. 88 However, a


82<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

further study utilizing both histology and<br />

immunohistochemistry on 25 cases of infants<br />

with SIDS failed to confirm this hypothesis. 89<br />

Diagnosis<br />

Diagnosis of ulcer is based on the endoscopic<br />

examination of the stomach and the duodenal<br />

Figure 6.3 Endoscopic appearance of a <strong>Helicobacter</strong><br />

<strong>pylori</strong>-associated peptic ulcer of the posterior wall of the<br />

duodenal bulb in an 11-year-old girl.<br />

Figure 6.4 Endoscopic appearance of <strong>Helicobacter</strong><br />

<strong>pylori</strong>-associated multiple peptic ulcers of the gastric body<br />

and hyperplastic regeneration of the surrounding mucosa<br />

in a 12-year-old girl.<br />

bulb. Primary ulcers are often single and localized<br />

at the duodenal bulb (Figure 6.3) or in the<br />

distal part of the stomach (Figure 6.4) (antrum,<br />

lesser curvature); while secondary ulcers<br />

(NSAIDs, stress ulcer) can be located in all parts<br />

of the stomach and can be multiple. Endoscopy<br />

may show changes of the gastroduodenal mucosa<br />

typical of gastropathy. Dohil et al 90 have<br />

suggested an easy and quite useful endoscopic<br />

classification of gastropathies in erosive and<br />

non-erosive forms (Table 6.5). Even if some<br />

disorders may show both erosive and nonerosive<br />

lesions, each is classified by its most<br />

common clinical manifestation. The endoscopic<br />

picture of H. <strong>pylori</strong>-associated gastritis is characterized<br />

by the presence of micronodularity of the<br />

antral mucosa in more than 50% of cases (Figure<br />

6.5). 90<br />

Some endoscopic pictures might be suggestive of<br />

inflammation and/or be specific for a given etiology,<br />

but confirmation of the initial impression<br />

and definitive diagnosis is dependent on histological<br />

examination. For this reason, each endoscopic<br />

examination must be completed by biopsy<br />

sampling of both endoscopically abnormal and<br />

normal mucosa. At least two biopsies for H.<br />

<strong>pylori</strong> detection must be taken at the antral site.<br />

Further sampling from the fundus may be useful,<br />

especially after treatment, because of the<br />

tendency of the bacterial colonization to migrate<br />

proximally. Biopsies for other procedures<br />

(culture, urease test) should be considered after<br />

sampling for routine histology.<br />

H. <strong>pylori</strong>-associated gastritis is the most frequent<br />

microscopic finding. It is predominantly an<br />

antral gastritis, but in some patients, and particularly<br />

in adults, inflammation may involve the<br />

entire stomach (pangastritis). In children, its<br />

severity is usually less, and features of activity<br />

(presence of polymorphonuclear leukocytes) are<br />

reported in 40% of cases. 69,90 The presence of<br />

lymphoid follicle hyperplasia (follicular gastritis)<br />

(Figure 6.6), suspected of being the histological<br />

counterpart of the nodular appearance of the<br />

gastric mucosa at endoscopy, has been reported<br />

in 20% of patients. 69,90 In adults, pangastritis<br />

may show gastric atrophy and/or focal intestinal<br />

metaplasia, particularly in association with<br />

gastric ulcers. In children, these findings are<br />

rarely reported (Figure 6.7).


Table 6.5 Classification of gastritis and gastropathy in children* 90<br />

Erosive and/or hemorrhagic gastritis or gastropathy<br />

‘Stress’ gastropathy<br />

<strong>Neonatal</strong> gastropathies<br />

Traumatic gastropathy<br />

Aspirin and other non-steroidal anti-inflammatory drugs<br />

Other drugs<br />

<strong>Portal</strong> hypertensive gastropathy<br />

Uremic gastropathy<br />

Chronic varioliform gastritis<br />

Bile gastropathy<br />

Henoch–Schönlein gastropathy<br />

Corrosive gastropathy<br />

Exercise-induced gastropathy or gastritis<br />

Radiation gastropathy<br />

Non-erosive gastritis or gastropathy<br />

‘Non-specific’ gastritis<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis<br />

Crohn’s gastritis<br />

Allergic gastritis<br />

Proton pump inhibitor gastropathy<br />

Celiac gastritis<br />

Gastritis of chronic granulomatous disease<br />

Cytomegalovirus gastritis<br />

Eosinophilic gastritis<br />

Collagenous gastritis<br />

Graft-versus-host disease<br />

Ménétrier’s disease<br />

Pernicious anemia<br />

Gastritis with autoimmune disease<br />

Other granulomatous gastritides<br />

Phlegmonous and emphysematous gastritis<br />

Other infectious gastritides<br />

*Although some disorders can present as either erosive or non-erosive, each is<br />

classified by its most common manifestation<br />

In recent years, the importance of the histological<br />

diagnosis of gastritis, on the basis of routinely<br />

obtained antral and body biopsies, has increased<br />

enormously, particularly because of the discovery<br />

Diagnosis 83<br />

of H. <strong>pylori</strong>. The introduction of the Sydney<br />

system made it possible, for the first time, to grade<br />

histological parameters, to identify topographic<br />

distribution and, finally, to make a statement about


84<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

(a) (b)<br />

Figure 6.5 Endoscopic appearance of antral nodularity associated with <strong>Helicobacter</strong> <strong>pylori</strong> infection (a). After biopsy,<br />

mucosal bleeding enhances the evidence of the picture (b).<br />

Figure 6.6 Immunohistochemical staining for B lymphocytes<br />

of a mucosa-associated lymphoid follicle in a child<br />

with <strong>Helicobacter</strong> <strong>pylori</strong>-associated antral nodularity.<br />

the etiopathogenesis of the gastritis in H. <strong>pylori</strong>- or<br />

non-H. <strong>pylori</strong>-associated gastritis. 91<br />

Multiple samplings from the antrum, the body<br />

and the fundus of the stomach allow further<br />

subdivision of the group of H. <strong>pylori</strong>-associated<br />

gastritis into forms of gastritis whose morphological<br />

distribution patterns usually identify them as<br />

sequelae of H. <strong>pylori</strong> infection. Moreover, the<br />

group of gastritis not associated with H. <strong>pylori</strong> can<br />

be differentiated into autoimmune, chemically<br />

induced reactive gastritis, ex-H. <strong>pylori</strong> gastritis, H.<br />

heilmannii gastritis, Crohn’s gastritis and a<br />

Figure 6.7 Giemsa-stained antral specimen showing, on<br />

the left side, typical intestinal metaplasia and, on the right<br />

side, <strong>Helicobacter</strong> <strong>pylori</strong>-associated chronic gastritis.<br />

number of special forms of gastritis (see Table<br />

6.5).<br />

Diagnosis of H. <strong>pylori</strong> infection is based on the<br />

demonstration of H. <strong>pylori</strong> by either direct (invasive)<br />

or indirect (non-invasive) methods. H. <strong>pylori</strong><br />

can be demonstrated on the gastric mucosa specimen<br />

by staining with Warthin–Starry, Giemsa<br />

(Figure 6.7) or orange acridine stain, or it can grow<br />

in culture of gastric biopsies on specific media and<br />

in microaerophilic conditions. 2 Culture may be<br />

particularly useful for specific antibiotic sensitivity<br />

testing. An easy, diffuse and rapid test is the


urease test, which is based on the color reaction<br />

induced by the presence of urease in the gastric<br />

specimen within 1 h. 92<br />

Indirect tests are based on the demonstration of an<br />

elevated titer of IgG or IgA antibodies against H.<br />

<strong>pylori</strong> in serum 22 or in saliva, 93 or the presence of<br />

a positive immunoassay for H. <strong>pylori</strong> in the<br />

stools. 94 For research purposes, H. <strong>pylori</strong> may be<br />

detected by PCR in some human samples, such as<br />

dental plaque or feces. 25,28 Results obtained using<br />

these diagnostic methods are variable, but among<br />

invasive tests, histology and culture are superior. If<br />

indirect tests are considered, the 13 C-urea breath<br />

test (UBT) and fecal antigen tests show the best<br />

accuracy. In a recent study comparing more tests<br />

on 53 children, 1 all the diagnostic tests except<br />

serology were excellent methods of diagnosing H.<br />

<strong>pylori</strong> infection. The diagnostic accuracy was<br />

96.2% for the stool antigen test, 96.2% for the<br />

biopsy urease test, 98.1% for histology, 94.3% for<br />

PCR, 98.1% for culture, 100% for the 13 C-UBT and<br />

84.9% for serology. 95<br />

The North American Society of Pediatric<br />

Gastroenterology, Hepatology and Nutrition<br />

(NASPGHAN) clinical practice guidelines (Table<br />

6.6), 96 as well as the European (EPTFHP) (Table<br />

6.6) 97 and Canadian 98 pediatric consensus conferences<br />

on H. <strong>pylori</strong> infection, recommend making a<br />

definitive diagnosis of H. <strong>pylori</strong> infection through<br />

Treatment 85<br />

endoscopy with multiple biopsies of the stomach.<br />

The role of non-invasive methods has to be<br />

reserved to the demonstration of eradication and<br />

for follow-up. For this purpose, the 13 C-UBT is the<br />

best validated method, 99 even though some problems<br />

may exist in patients younger than 6 years of<br />

age.<br />

Treatment<br />

One of the most debated problems is who should<br />

be treated for H. <strong>pylori</strong> infection. In adults, the<br />

most recent indications for H. <strong>pylori</strong> treatment are<br />

listed in the Table 6.7, derived from the Maastricht<br />

2-2000 Consensus Report. 100<br />

Each of the three Pediatric Consensus Reports<br />

(North American, European and Canadian)<br />

outlined recommendations for the detection of<br />

H. <strong>pylori</strong> infection. 96–98 Regarding the H. <strong>pylori</strong>positive<br />

subjects to be treated, the NASPGHAN<br />

supports the treatment only in case of PUD and<br />

MALToma. 96 The European Pediatric Task Force on<br />

<strong>Helicobacter</strong> <strong>pylori</strong> (EPTFHP) consensus statement<br />

suggests providing treatment for the infection if H.<br />

<strong>pylori</strong> is identified in a child at endoscopy (Table<br />

6.6). 97<br />

Children with H. <strong>pylori</strong>-positive PUD must be<br />

submitted to eradicating treatment, because<br />

Table 6.6 Guidelines for the management of <strong>Helicobacter</strong> <strong>pylori</strong> infection in children<br />

Diagnosis Evidence Treatment Evidence<br />

North American Society of Pediatric Gastroenterology, Hepatology and Nutrition<br />

Endoscopy and histology II peptic ulcer disease I<br />

13C-urea breath test II MALToma<br />

atrophic gastritis<br />

III<br />

I. metaplasia<br />

triple therapy for 1–2 weeks<br />

III<br />

European Pediatric Task Force on <strong>Helicobacter</strong> <strong>pylori</strong><br />

Endoscopy and histology peptic ulcer disease<br />

13C-urea breath test for follow-up all <strong>Helicobacter</strong> <strong>pylori</strong>-positive children after<br />

endoscopy (information of the possibility<br />

of symptoms’ persistence)


86<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

Table 6.7 The Maastricht 2-2000 guidelines for the treatment of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection in adults<br />

Strongly recommended indications<br />

Peptic ulcer disease<br />

MALToma<br />

Atrophic gastritis<br />

Post-gastric cancer resection<br />

First-degree relatives of gastric cancer patients<br />

Patient’s wishes (after physician consultation)<br />

Advisable indications<br />

Functional dyspepsia (subset of patients)<br />

Gastroesophageal reflux disease (in patients requiring long-term proton pump<br />

inhibitor therapy)<br />

Users of non-steroidal anti-inflammatory drugs<br />

relapse after effective ulcer healing has been<br />

demonstrated in 47–80% of children with persistent<br />

H. <strong>pylori</strong> infection, and exceptionally in those<br />

in whom H. <strong>pylori</strong> has been eradicated. 31,37,97<br />

Symptomatic relief of dyspeptic symptoms is<br />

usually achieved. 73<br />

H. <strong>pylori</strong> eradication in children with chronic<br />

active gastritis is associated with evident<br />

histological improvement, 69 but remission of<br />

symptoms does not necessarily follow. 73,97 However,<br />

if the infection is not treated, worsening of<br />

gastritis and of the clinical picture, particularly in<br />

males and in children infected with Cag-A-positive<br />

strains, has been documented in a close follow-up<br />

study. 101 In adults as well as in teenagers with<br />

iron-deficient anemia, H. <strong>pylori</strong> eradication was<br />

associated with a rise in hemoglobin levels and in<br />

ferritin values, suggesting that treatment should be<br />

considered. 82–85<br />

In adults with peptic esophagitis, the treatment of<br />

associated H. <strong>pylori</strong> infection is a question of<br />

debate. 102 Some authors have suggested that<br />

esophagitis and gastroesophageal reflux disease<br />

(GERD) may be exacerbated by eliminating the<br />

protecting buffering effect of H. <strong>pylori</strong> infection. 103<br />

Nevertheless, a causal relationship between these<br />

phenomena remains unproven, and is mainly<br />

based on retrospective analyses or epidemiological<br />

hypothesis. The small number of prospective trials<br />

in adults has not consistently demonstrated an<br />

increased risk of GERD after H. <strong>pylori</strong> eradication.<br />

104<br />

In a study planned to answer the question of<br />

whether eradication of H. <strong>pylori</strong> infection in<br />

children with RAP should result in clinical<br />

improvement, no correlation was seen between<br />

eradication and disappearance of RAP, after 3 or<br />

after 6 months of observation. 105<br />

To date, extrapolation of data from randomized<br />

controlled trials in adults has been the basic<br />

approach to treating children with H. <strong>pylori</strong> infection.<br />

A systematic review of the small published<br />

open trials in 870 children from 1987 to 2000 has<br />

recently been published. 106 A total of 79 children<br />

were treated with one drug (monotherapy); 345<br />

were treated with dual therapy, including two<br />

antibiotics or one antibiotic plus bismuth<br />

compounds or H2-receptor antagonists (H2RA) or<br />

proton pump inhibitors (PPIs); and 446 with a<br />

bismuth- or PPI-based triple therapy plus two<br />

antibiotics. In pediatrics, the most frequently<br />

utilized antibiotics were amoxicillin, clar-


ithromycin, and the nitroimidazoles, metronidazole<br />

or tinidazole. The reasons for the development<br />

of combined therapies were:<br />

(1) Different strains with different antibiotic<br />

sensitivity may colonize the same patient.<br />

Sensitivity to amoxicillin, clarithromycin and<br />

nitroimidazoles is one of the key factors in<br />

the eradication rates. Unpublished data<br />

collected by the EPTFHP showed metronidazole,<br />

clarithromycin or double resistance<br />

before the first therapy in 24.5%, 20% and<br />

7%, respectively, of 400 H. <strong>pylori</strong>-infected<br />

children aged under 15 years.<br />

(2) Stability and efficacy of antibiotics are often<br />

impaired in the acid environment. This<br />

consideration prompted the combination of<br />

antibiotics with drugs able to increase the<br />

gastric pH (H2RA or PPI).<br />

In general, monotherapy with one antibiotic, or<br />

bismuth compounds, or H2RA or PPI showed a<br />

very low eradication rate and it is not recommended.<br />

Combination of one antibiotic with PPI<br />

was not satisfactory (mean eradication rate of<br />

39%), while combination of amoxicillin with<br />

nitroimidazole or one of the two antibiotics with<br />

bismuth salts was tried in 248 children, with a<br />

mean eradication rate of 73–76%. 106<br />

Table 6.8 Protocols of <strong>Helicobacter</strong> <strong>pylori</strong> treatment in children<br />

First-line therapy<br />

PPI (1 mg/kg per day up to 20 mg bid) plus<br />

Amoxicillin (50 mg/kg per day bid up to 1 g bid) plus<br />

Clarithromycin (15 mg/kg per day up to 500 mg bid) or<br />

Metro(ti)nidazole (20 mg/kg per day up to 500 mg bid) for 1 week<br />

In case of failure to eradicate the bacterium:<br />

Second-line therapy<br />

Treatment 87<br />

Triple therapy schedules did not work much<br />

better. A recent randomized clinical trial<br />

confirmed that 1 week of omeprazole–amoxicillin–<br />

clarithromycin resulted in successful eradication<br />

of H. <strong>pylori</strong> in 75% of 73 children


88<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

the most recommended. 97 Recent data showed that<br />

fecal H. <strong>pylori</strong> antigen assays could be an effective<br />

alternative. 109 Re-infection after eradication is<br />

uncommon in children, and treatment of the<br />

affected family member at present is not<br />

supported. 17<br />

In the case of relapse, biopsy culture for antibiotic<br />

sensitivity testing should be proposed for a better<br />

chance of eradication. If culture is not available or<br />

if H. <strong>pylori</strong> does not grow, substitution of clarithromycin<br />

with a nitroimidazole (and vice versa)<br />

is the first step, because of the high probability of<br />

development of a resistance to either drug. A 2week<br />

regimen should be offered to relapsing<br />

patients (Table 6.8).<br />

Short- and long-term prognosis<br />

In patients with H. <strong>pylori</strong>-associated PUD, successful<br />

eradication is followed by ulcer healing and<br />

usually evident clinical improvement. PUD does<br />

not relapse and reacquisition of the infection is<br />

rare after 5 years of age. 31,42,97 Children with<br />

abdominal complaints associated with chronic<br />

gastritis have a more unpredictable clinical outcome,<br />

even if H. <strong>pylori</strong> is eradicated. 73,97<br />

Among long-term complications of H. <strong>pylori</strong>associated<br />

chronic gastritis, the most important is<br />

the multistep disease progression towards gastric<br />

atrophy, intestinal metaplasia, dysplasia and<br />

gastric cancer. 110 In general, this is a rare evolution,<br />

affecting no more than 1% of H. <strong>pylori</strong>positive<br />

patients. This multifactorial process<br />

includes host, bacterial and dietary factors.<br />

Family studies have shown that gastric cancer is<br />

significantly higher in first-degree relatives of<br />

gastric cancer patients. 111 Epidemiological<br />

case–control and cohort studies have demonstrated<br />

that H. <strong>pylori</strong> is strongly associated with<br />

non-cardia gastric carcinoma of both the intestinal<br />

and the diffuse type. 112,113 An extensive review on<br />

this topic showed the best estimate of the relative<br />

risk of non-cardia gastric cancer associated with H.<br />

<strong>pylori</strong> infection to be 5.9. 113 The odds ratio<br />

decreased significantly with aging, from 9.29 in<br />

patients with carcinoma at age 20–29 years, to 1 in<br />

those older than 70 years of age. 112 For some<br />

authors, the basis of the genetic predisposition lies<br />

either in genetic susceptibility to the infection 20 or<br />

in mechanisms of DNA repair and carcinogenesis.<br />

114 Mutation in p53 is one of the most common<br />

genetic alterations found in human cancer, including<br />

gastric cancer. 115 Inhibition of apoptosis<br />

secondary to up-regulation of cyclo-oxygenase 2<br />

by H. <strong>pylori</strong>-induced inflammation could play a<br />

role in inducing intestinal metaplasia. In a<br />

subgroup of gastric cancer, as well as in intestinal<br />

metaplasia, microsatellite instability, secondary to<br />

germline mutation of the DNA mismatch repair<br />

genes, has also been demonstrated. The mechanisms<br />

inducing intestinal metaplasia and carcinogenesis<br />

have recently been reviewed. 114<br />

Acid output has long been investigated as a key<br />

factor of ulcer development and gastric cancer.<br />

Indeed, patients with higher acid output are likely<br />

to have antral-predominant gastritis and eventually<br />

duodenal ulcer, but they do not show any<br />

predisposition towards gastric cancer. 116 Patients<br />

with lower acid output, on the other hand, are<br />

more likely to have a body-predominant gastritis,<br />

which predisposes to gastric ulcer and to the<br />

multistep progression of disease that, in rare cases,<br />

leads to gastric carcinoma. Polymorphisms of the<br />

IL-1β promoter have been linked to altered gastric<br />

acid secretion and pre-malignant histological<br />

features. Severity of the host response to H. <strong>pylori</strong><br />

infection was related to individual ability to<br />

produce IL-1, which has been demonstrated to be<br />

a potent inhibitor of gastric acid secretion, as well<br />

as a proinflammatory cytokine. 117 The histological<br />

pathways of H. <strong>pylori</strong> infection and mechanisms<br />

involved in carcinogenesis are shown in Figure<br />

6.8. Genetic as well as acquired (H. <strong>pylori</strong>dependent)<br />

determinants co-operate, increasing<br />

the risk of gastric cancer up to 90-fold in patients<br />

with intestinal metaplasia.<br />

Despite the strong causal link between H. <strong>pylori</strong><br />

and gastric carcinoma, evidence that treatment for<br />

H. <strong>pylori</strong> infection may actually prevent gastric<br />

cancer is lacking. The long development time of<br />

gastric cancer is the main problem in performing<br />

such studies. It has been estimated that a trial with<br />

sufficient power to answer the question would<br />

have to recruit 100 000 infected subjects with a 20year<br />

follow-up. 118 In addition, studies planned to<br />

investigate the regression of pre-malignant lesions<br />

after eradication gave conflicting results. The<br />

progression rate of intestinal metaplasia does not


OR Cancer H. <strong>pylori</strong>+<br />

duodenal ulcer<br />

0<br />

gastric ulcer<br />

3.4<br />

NAG<br />

2–4.7<br />

Atrophic gastritis (MAG)<br />

p53 mutation<br />

MSL hypermethylation<br />

TGF-alpha EGFR<br />

intestinal metaplasia<br />

6.4 - > 90<br />

Metaplasia Dysplasia<br />

Cancer<br />

(intestinal type)<br />

appear to be arrested by eradication; however, eradicating<br />

H. <strong>pylori</strong> leads to resolution of inflammation,<br />

elimination of DNA damage and reduction of<br />

proliferation, all changes possibly preventing<br />

gastric cancer. 114<br />

Another malignancy strongly associated with H.<br />

<strong>pylori</strong> infection is gastric B-cell lymphoma, known<br />

as MALToma. Even if the stomach is the most<br />

common site of the extranodal lymphomas,<br />

MALTomas are rare in adults (0.71 cases/100 000<br />

per year in the USA) and definitely exceptional in<br />

children. The association of H. <strong>pylori</strong> infection and<br />

MALTomas is based on these considerations: H.<br />

<strong>pylori</strong> is found in more than 90% of cases; 119 H.<br />

<strong>pylori</strong> infection precedes the development of the<br />

malignancy; and eradicating the infection results in<br />

the regression of MALToma in approximately 70%<br />

of cases of low-grade disease (stage IE). 120 Thus, H.<br />

<strong>pylori</strong> infection in a patient with MALToma is a<br />

strong indication for eradication treatment. 100<br />

Regression of MALToma after H. <strong>pylori</strong> eradication<br />

was described in a 14-year-old female. 121<br />

Prophylactic and therapeutic immunization 89<br />

Antral gastritis<br />

(± duodenal ulcer)<br />

+<br />

Acid output<br />

–/=<br />

Corpus gastritis or<br />

pangastritis<br />

(± gastric ulcer)<br />

(NAG) non-atrophic<br />

gastritis<br />

Cancer<br />

(diffuse type)<br />

MALToma<br />

Figure 6.8 The natural history of <strong>Helicobacter</strong> <strong>pylori</strong> infection and associated diseases depends on aging, as well as<br />

genetic and bacterial factors (see text). Determinants of diseases are the acid output for the development of peptic ulcer,<br />

and the existence of carcinogenetic mechanisms which trigger the multistep gastric carcinogenesis pathway. On the left,<br />

are the odds ratios for gastric cancer associated with different clinical pictures. EGFR, epidermal growth factor receptor;<br />

MSL, microsatellite instability; TGF, transforming growth factor.<br />

A<br />

G<br />

I<br />

N<br />

G<br />

Prophylactic and therapeutic<br />

immunization<br />

As we have seen, effective therapies against<br />

<strong>Helicobacter</strong> require the child to ingest multiple<br />

drugs several times a day for at least 1 week. 106<br />

The common occurrence of adverse effects<br />

(nausea, diarrhea, abdominal pain and, more<br />

rarely, pseudomembranous colitis), the frequently<br />

observed low compliance and the high costs<br />

suggest the need to evaluate different approaches.<br />

Despite the fact that patients infected with H.<br />

<strong>pylori</strong> have a systemic and local immune<br />

response, 57 this response does not resolve the<br />

infection. Studies on H. <strong>pylori</strong>-specific cellular<br />

immunity suggested that H. <strong>pylori</strong> induces a proinflammatory<br />

cascade and a Th1 response that<br />

contributes to the chronicity (Figure 6.2). Since<br />

1993, several groups have developed animal<br />

models of <strong>Helicobacter</strong> infection. 122 Using the<br />

mouse model, oral immunization with H. felis


90<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

lysates with cholera toxin (CT) as adjuvant<br />

resulted in 76–96% of protection. Others challenged<br />

the same models using H. <strong>pylori</strong> antigens<br />

such as Vac-A, Cag-A, catalase or urease B subunit.<br />

On the basis of the results of clinical therapeutic<br />

vaccination trials, it is likely that several antigens<br />

are needed for a subunit vaccine in humans.<br />

Several groups administered the vaccine to<br />

infected animals for therapeutic and not for<br />

prophylactic use. Oral vaccination of H. felisinfected<br />

mice with either bacterial sonicate or H.<br />

<strong>pylori</strong> urease B subunit plus CT cured approximately<br />

half of the mice. Several groups have now<br />

developed murine models of H. <strong>pylori</strong> infection.<br />

Eradication in 70–92% was achieved by using<br />

recombinant Cag-A or Vac-A as oral antigens,<br />

respectively.<br />

Studying the H. mustelae ferret model, which<br />

represents a natural host–pathogen disease, therapeutic<br />

immunization with H. <strong>pylori</strong> urease and CT<br />

achieved eradication in one-third of ferrets.<br />

Recently, a clinical trial tested an oral therapeutic<br />

vaccine consisting of recombinant H. <strong>pylori</strong> urease<br />

apoenzyme coupled with Escherichia coli LT in<br />

REFERENCES<br />

1. Marshall BJ, Warren JR. Unidentified curved bacilli in<br />

the stomach of patients with gastritis and peptic ulcerations.<br />

Lancet 1984; 1: 1311–1315.<br />

2. Windsor HM, O’Rourke J. Bacteriology and taxonomy of<br />

<strong>Helicobacter</strong> <strong>pylori</strong>. Gastroenterol Clin North Am 2000;<br />

29: 633–648.<br />

3. NIH Consensus Development Panel on <strong>Helicobacter</strong><br />

<strong>pylori</strong> in peptic ulcer disease. JAMA 1994; 272: 65–69.<br />

4. International Agency for Research on Cancer.<br />

Monograph on the Evaluation of Carcinogenics Risks to<br />

Humans. Schistosomes, Liver Flukes and <strong>Helicobacter</strong><br />

<strong>pylori</strong>. Lyon, France: International Agency for Research<br />

on Cancer, 1994.<br />

5. Drumm B, Rhoads JM, Sringer DA et al. Peptic ulcer<br />

disease in children: etiology, clinical findings and clinical<br />

course. Pediatrics 1988; 82: 410–414.<br />

6. Eastham EJ. Peptic ulcer. In Walker WA, Durie P,<br />

Hamilton JR et al, eds. Pediatric Gastrointestinal<br />

humans; a significant reduction in gastric H. <strong>pylori</strong><br />

density was achieved along with an increase of<br />

specific IgA-producing cells. 123<br />

A different approach was based on the development<br />

of attenuated recombinant enteric pathogens<br />

expressing <strong>Helicobacter</strong> antigens. Cloning of<br />

urease A and B subunits in an attenuated strain of<br />

Salmonella typhimurium achieved protection in<br />

mice. Recently, the safety and immunogenicity of<br />

live recombinant Salmonella typhi vaccine<br />

expressing urease A and B from H. <strong>pylori</strong> in human<br />

volunteers has been tested with encouraging<br />

results. 124<br />

In summary, these immunization studies are very<br />

promising, but at the same time indicate that many<br />

problems are still unsolved: what constitutes the<br />

best antigen, the safest adjuvant and the best route<br />

of administration? In spite of these uncertainties, it<br />

is evident that childhood vaccination could, in<br />

theory, prevent one of the most diffuse chronic<br />

infections in the world, avoiding consistent<br />

morbidity in adult life, and reducing the incidence<br />

of gastric cancer later in life.<br />

Disease: Pathophysiology, Diagnosis, Management, vol 1.<br />

Philadelphia: BC Decker, 1991: 438–451.<br />

7. Nord KS, Rossi TM, Lebenthal E. Peptic ulcer in children:<br />

the predominance of gastric ulcers. Am J<br />

Gastroenterol 1981; 75: 153–157.<br />

8. Megraud F, Brassens-Rabbè MP, Denis F et al.<br />

Seroepidemiology of Campylobacter <strong>pylori</strong> infection in<br />

various populations. J Clin Microbiol 1989; 27:<br />

1870–1873.<br />

9. Thomas JE, Dale A, Harding M et al. <strong>Helicobacter</strong> <strong>pylori</strong><br />

colonization in early life. Pediatr Res 1999; 45: 218–223.<br />

10. De Giacomo C, Valdambrini V, Lizzoli F et al. A population-based<br />

survey on gastrointestinal tract symptoms<br />

and <strong>Helicobacter</strong> <strong>pylori</strong> infection in children and adolescents.<br />

<strong>Helicobacter</strong> 2002; 7: 356–363.<br />

11. Banatvala M, Mayo K, Megraud F et al. The cohort<br />

effect and <strong>Helicobacter</strong> <strong>pylori</strong>. J Infect Dis 1993; 168:<br />

219–221.


12. Fujisawa T, Kumagai T, Akamatsu T et al. Changes in<br />

sero-epidemiological pattern of <strong>Helicobacter</strong> <strong>pylori</strong> and<br />

hepatitis A virus over the last 20 years in Japan. Am J<br />

Gastroenterol 1999; 94: 2094–2099.<br />

13. Roosendaal R, Kuipers EJ, Buitenwerf J et al.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> and the birth cohort effect: evidence<br />

of a continuous decrease of infection rates in childhood.<br />

Am J Gastroenterol 1997; 92: 1480–1482.<br />

14. Fiodorek SC, Malaty HM, Evans DL et al. Factors influencing<br />

the epidemiology of <strong>Helicobacter</strong> <strong>pylori</strong> infection<br />

in children. Pediatrics 1991; 88: 578–582.<br />

15. Mendall MA, Goggin PM, Molineaux N et al. Childhood<br />

living conditions and <strong>Helicobacter</strong> <strong>pylori</strong> seropositivity<br />

in adult life. Lancet 1992; 339: 896–897.<br />

16. Malaty HM, El-Kasabany A, Graham DY et al. Age at<br />

acquisition of <strong>Helicobacter</strong> <strong>pylori</strong> infection: a follow-up<br />

study from infancy to adulthood. Lancet 2002; 359:<br />

931–935.<br />

17. Rowland M, Kumar D, Daly L et al. Low rates of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> reinfection in children.<br />

Gastroenterology 1999; 117: 336–341.<br />

18. Klein PD, Gilman RH, Leon-Barua R et al. The epidemiology<br />

of <strong>Helicobacter</strong> <strong>pylori</strong> in Peruvian children<br />

between 6 and 30 months of age. Am J Gastroenterol<br />

1994; 89: 2196–2200.<br />

19. Parsonnet J, Blaser MJ, Perez-Perez GI et al. Symptoms<br />

and risk factors of <strong>Helicobacter</strong> <strong>pylori</strong> infection in a<br />

cohort of epidemiologists. Gastroenterology 1992; 102:<br />

41–46.<br />

20. Kuipers EJ, Pena AS, van Kamp G et al. Seroconversion<br />

for <strong>Helicobacter</strong> <strong>pylori</strong>. Lancet 1993; 342: 328–331.<br />

21. Malaty HM, Engstrand L, Pederson NL, Graham DY.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection: genetic and environmental<br />

influences: a study of twins. Ann Intern Med 1994; 120:<br />

982–986.<br />

22. De Giacomo C, Lisato L, Negrini R et al. Serum immune<br />

response to <strong>Helicobacter</strong> <strong>pylori</strong> in children: epidemiologic<br />

end clinical applications. J Pediatr 1991; 119:<br />

205–210.<br />

23. Buckley M, Mitchell HM, Bolin TD et al. Impact of<br />

institutionalization on prevalence of <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection. Gastroenterology 2001; 120:<br />

A735A735(abstract).<br />

24. Drumm B, Rowland M. The epidemiology of<br />

<strong>Helicobacter</strong> <strong>pylori</strong>: where to from here? J Pediatr<br />

Gastroenterol Nutr 2003; 36: 7–8.<br />

25. Nguyen AMH, Engstrand L, Genta RM et al. Detection<br />

of <strong>Helicobacter</strong> <strong>pylori</strong> in dental plaque by reverse transciption-polymerase<br />

chain reaction. J Clin Microbiol<br />

1993; 31: 783–787.<br />

26. Parsonnet J, Shmuely H, Haggerty T. Fecal and oral<br />

shedding of <strong>Helicobacter</strong> <strong>pylori</strong> from healthy infected<br />

adults. JAMA 1999; 282: 2240–2245.<br />

27. Klein P, Graham DY, Gaillour A et al. Water source as<br />

risk factor for <strong>Helicobacter</strong> <strong>pylori</strong> in Peruvian children.<br />

Lancet 1991; 337: 1503–1506.<br />

28. Thomas JE, Gibson GR, Darboe MK et al. Isolation of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> from human feces. Lancet 1992; 340:<br />

1194–1195.<br />

29. Marshall BJ, Goodwin CS, Warren JR et al. Prospective<br />

double-blind trial of duodenal ulcer relapse after eradication<br />

of Campylobacter <strong>pylori</strong>. Lancet 1988; 2:<br />

1437–1442.<br />

30. Macarthur C, Saunders N, Feldman W. <strong>Helicobacter</strong><br />

<strong>pylori</strong>, gastroduodenal disease, and recurrent abdominal<br />

pain in children. JAMA 1995; 273: 729–734.<br />

31. Oderda G, Forni M, Dell’Olio D, Ansaldi N. Cure of<br />

peptic ulcer associated with eradication of <strong>Helicobacter</strong><br />

<strong>pylori</strong>. Lancet 1990; 335: 1599.<br />

References 91<br />

32. Mention K, Michaud L, Guimber D et al. Characteristics<br />

and prevalence of <strong>Helicobacter</strong> heilmannii infection in<br />

children undergoing upper gastrointestinal endoscopy. J<br />

Pediatr Gastroenterol Nutr 1999; 29: 533–539.<br />

33. Tomb JF, White O, Kerlavage AR et al. The complete<br />

genome sequence of the gastric pathogen <strong>Helicobacter</strong><br />

<strong>pylori</strong>. Nature 1997; 388: 539-547. [Erratum, Nature<br />

1997; 389: 412]<br />

34. Falush D, Kraft C, Taylor NS et al. Recombination and<br />

mutation during long-term gastric colonization by<br />

<strong>Helicobacter</strong> <strong>pylori</strong>: estimates of clock rates, recombination<br />

size, and minimal age. Proc Natl Acad Sci USA<br />

2001; 98: 15056–15061.<br />

35. Euler AR, Byrne WJ, Cousins LM et al. Increased serum<br />

gastrin concentrations and gastric acid hyposecretion in<br />

the immediate newborn period. Gastroenterology 1977;<br />

72: 1271–1273.<br />

36. Euler AR, Byrne WJ, Meis PJ et al. Basal and pentagastrin-stimulated<br />

acid secretion in newborn human<br />

infants. Pediat Res 1979; 13: 36–37.<br />

37. Agunod M, Yamaguchi N, Lopez R et al. Correlative<br />

study of hydrochloric acid, pepsin, and intrinsic factor<br />

secretion in newborns and infants. Am J Dig Dis 1969;<br />

14: 400–414.<br />

38. Kopel FB, Barbero GJ. Gastric acid secretion in infancy<br />

and childhood. Gastroenterology 1967; 52: 1101.<br />

39. Christie DL, Ament ME. Gastric acid hypersecretion in<br />

children with duodenal ulcer. Gastroenterology 1976;<br />

71: 242–244.<br />

40. Mohammed R, Hearns JB, Cream GP. Gastric acid secretion<br />

in children with duodenal ulceration. Scand J<br />

Gastroenterol 1982; 17: 289–292.<br />

41. Liebman WM. Gastric acid secretion and serum gastrin<br />

levels in children with recurrent abdominal pain,<br />

gastric and duodenal ulcers. J Clin Gastroenterol 1980;<br />

2: 243–246.<br />

42. Tam PKH, Saing H. Gastric acid secretion and emptying<br />

rates in chidren with duodenal ulcer. J Pediatr Surg<br />

1986; 21: 129–131.<br />

43. Christie DL, Ament ME. Gastrin response to a protein<br />

meal in children with duodenal ulcer. Pediatr Res 1976;<br />

10: 353.<br />

44. De Giacomo C, Fiocca R, Villani L. Omeprazole treatment<br />

of severe peptic disease associated with antral G<br />

cell hyperfunction and hyperpepsinogenemia I in an<br />

infant. J Pediatr 1990; 117: 898–993.<br />

45. Habibullah CM, Alis MM, Ishaq M. Study of duodenal<br />

ulcer disease in 100 families using total serum pepsinogen<br />

as a genetic marker. Gut 1984; 25: 1380–1383.<br />

46. Suerbaum S, Michetti P. <strong>Helicobacter</strong> <strong>pylori</strong> infection. N<br />

Engl J Med 2002; 347: 1175–1186.<br />

47. Mobley HLT. <strong>Helicobacter</strong> <strong>pylori</strong> urease. In Achtman M,<br />

Suerbaum S, eds. <strong>Helicobacter</strong> <strong>pylori</strong>: Molecular and<br />

Cellular Biology. Wymondham, UK: Horizon Scientific<br />

Press, 2001: 155–170.<br />

48. Weeks DL, Eskandari S, Scott DR, Sachs G. A H + -gated<br />

urea channel: the link between <strong>Helicobacter</strong> <strong>pylori</strong><br />

urease and gastric colonization. Science 2000; 287:<br />

482–485.<br />

49. Ilver D, Arnqvist A, Ogren J et al. <strong>Helicobacter</strong> <strong>pylori</strong><br />

adhesin binding fucosylated histo-blood group antigens<br />

revealed by retagging. Science 1998; 279: 373–377.<br />

50. Atherton JC, Cao P, Peek RM et al. Mosaicism in vacuolating<br />

citotoxin alleles of <strong>Helicobacter</strong> <strong>pylori</strong>. J Biol<br />

Chem 1995; 270: 17771–17777.<br />

51. Montecucco C, Papini E, de Bernard M et al.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> VacA vacuolating cytotoxin and H.<br />

<strong>pylori</strong>-Nap neutrophil activating protein. In Achtman M,<br />

Suerbaum S, eds. <strong>Helicobacter</strong> <strong>pylori</strong>: Molecular and


92<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastritis and peptic ulcer disease<br />

Cellular Biology. Wymondham, UK: Horizon Scientific<br />

Press, 2001: 245–263.<br />

52. Atherton JC, Peek RM Jr, Tham KT et al. Clinical and<br />

pathological importance of heterogeneity in vacA, the<br />

vacuolating cytotoxin gene of <strong>Helicobacter</strong> <strong>pylori</strong>.<br />

Gastroenterology 1997; 112: 92–99.<br />

53. Covacci A, Censini S, Bugnoli M et al. Molecular characterization<br />

of the 128 kDa immunodominant antigen of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> associated with cytotoxicity and<br />

duodenal ulcer. Proc Natl Acad Sci USA 1993; 90:<br />

5791–5795.<br />

54. Odenbreit S, Puls J, Sedlmaier B et al. Translocation of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> CagA into gastric epithelial cells by<br />

type IV secretion. Science 2000; 287: 1497–1500.<br />

55. Crabtree JE. Mucosal immune responses to <strong>Helicobacter</strong><br />

<strong>pylori</strong>. Eur J Gastroenterol Hepatol 1992; 4(Suppl 2):<br />

S30–S32.<br />

56. Naumann M, Wessler S, Bartsch C et al. Activation of<br />

activator protein 1 and stress response kinases in<br />

epithelial cells colonized by <strong>Helicobacter</strong> <strong>pylori</strong> encoding<br />

the cag pathogenicity island. J Biol Chem 1999; 274:<br />

31655–31662.<br />

57. Perez-Perez GI, Dworkin BM, Chodos JE, Blaser MJ.<br />

Campylobacter <strong>pylori</strong> antibodies in humans. Ann Intern<br />

Med 1988; 109: 11–17.<br />

58. Negrini R, Savio A, Appelmelk BJ. Autoantibodies to<br />

gastric mucosa in <strong>Helicobacter</strong> <strong>pylori</strong> infection.<br />

<strong>Helicobacter</strong> 1997; 2(Suppl 1): S13–S16.<br />

59. Oderda G, Vaira D, Holton J et al. Amoxycillin plus<br />

tinidazole for Campylobacter <strong>pylori</strong> gastritis in children:<br />

assessment by serum IgG antibody, pepsinogen I, and<br />

gastrin levels. Lancet 1989; 1: 690–692.<br />

60. Queiroz DMM, Moura SB, Mendes EN et al. Effects of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> eradication on G-cell and D-cell<br />

density in children. Lancet 1994; 343: 1191–1193.<br />

61. El-Omar EM, Penman IA, Ardill JES et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> infection and abnormalities of acid secretion in<br />

patients with duodenal ulcer disease. Gastroenterology<br />

1995; 109: 681–691.<br />

62. Nord KS. Peptic ulcer. In Lebenthal E, ed. Textbook of<br />

Pediatric Gastroenterology and Nutrition, 2nd edn. New<br />

York: Raven Press, 1989: 815–827.<br />

63. Murphy MS, Eastham EJ, Jimenez M et al. Duodenal<br />

ulceration: review of 110 cases. Arch Dis Child 1987; 62:<br />

554–558.<br />

64. Apley J, Naish N. Recurrent abdominal pains: a field<br />

survey of 1000 school children. Arch Dis Child 1958; 33:<br />

165–170.<br />

65. Hyams JS, Hyman PE. Recurrent abdominal pain and<br />

the biopsychosocial model of medical practice. J Pediatr<br />

1998; 133: 473–478.<br />

66. Czinn SJ, Dahms BB, Jacobs GH et al. Campylobacterlike<br />

organisms in association with symptomatic gastritis<br />

in children. J Pediatr 1986; 109: 80–83.<br />

67. Drumm B, Sherman P, Cutz E, Karmali M. Association<br />

of Campylobacter <strong>pylori</strong> on the gastric mucosa with<br />

antral gastritis in children. N Engl J Med 1987; 316:<br />

1557–1561.<br />

68. Oderda G, Holton J, Altare F et al. Amoxycillin plus<br />

tinidazole for Campylobacter <strong>pylori</strong> gastritis in children:<br />

assessment by serum IgG antibody, pepsinogen I, and<br />

gastrin levels. Lancet 1989; 1: 690–693.<br />

69. De Giacomo C, Fiocca R, Villani L et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> infection and chronic gastritis: clinical, serological,<br />

and histologic correlations in children treated with<br />

amoxicillin and colloidal bismuth subcitrate. J Pediatr<br />

Gastroenterol Nutr 1990; 11: 310–306.<br />

70. Glassman MS, Schwarz SM, Medow MS et al.<br />

Campylobacter <strong>pylori</strong>-related gastrointestinal disease in<br />

children. Incidence and clinical findings. Dig Dis Sci<br />

1989; 34: 1501–1504.<br />

71. Blecker U, Hauser B, Lanciers S et al. Symptomatology<br />

of <strong>Helicobacter</strong> <strong>pylori</strong> infection in children. Acta<br />

Paediatr 1996; 85: 1156–1158.<br />

72. Mahony MJ, Wyatt JI, Littlewood JM. Management and<br />

response to treatment of <strong>Helicobacter</strong> <strong>pylori</strong> gastritis.<br />

Arch Dis Child 1992; 67: 940–943.<br />

73. Gormally SM, Prakash N, Durnin MT et al. Association<br />

of symptoms with <strong>Helicobacter</strong> <strong>pylori</strong> infection in children.<br />

J Pediatr 1995; 126: 753–756.<br />

74. O’Donohoe JM, Sullivan PB, Scott R et al. Recurrent<br />

abdominal pain and <strong>Helicobacter</strong> <strong>pylori</strong> in a community-based<br />

sample of London children. Acta Paediatr<br />

1996; 85: 961–964.<br />

75. Bode G, Rothenbacher D, Brenner H, Adler G.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> and abdominal symptoms: a population-based<br />

study among pre-school children in southern<br />

Germany. Pediatrics 1998; 101: 634–637.<br />

76. Rasquin-Weber A, Hyman PE, Cucchiara S et al.<br />

Childhood functional gastrointestinal disorders. Gut<br />

1999; 45 (Suppl 2); II60–II68.<br />

77. Tucci A, Corinaldesi R, Stanghellini V et al.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection and gastric function in<br />

patients with chronic idiopathic dyspepsia.<br />

Gastroenterology 1992; 103: 768–774.<br />

78. Talley NJ. Dyspepsia management in the millennium:<br />

the death of test and treat? Gastroenterology 2002; 122:<br />

1521–1525.<br />

79. Sullivan PB, Thomas JE, Wight DG et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> in Gambian children with chronic diarrhoea and<br />

malnutrition. Arch Dis Child 1990; 65: 189–191.<br />

80. De Giacomo C, Fiocca R, Villani L et al. Barrett’s ulcer<br />

and Campylobacter-like organisms infection in a child. J<br />

Pediatr Gastroenterol Nutr 1988; 7: 766–768.<br />

81. Hill P, Rode J. <strong>Helicobacter</strong> <strong>pylori</strong> in ectopic gastric<br />

mucosa in Meckel’s diverticulum. Pathology 1998; 30:<br />

7–9.<br />

82. Sherman P, Macarthur C. Current controversies associated<br />

with <strong>Helicobacter</strong> <strong>pylori</strong> infection in the pediatric<br />

population. Frontiers Biosci 2001; 6: e187–192.<br />

83. Barabino A, Dufour C, Marino CE. Unexplained refractory<br />

iron-deficiency anemia associated with<br />

<strong>Helicobacter</strong> <strong>pylori</strong> gastric infection in children: further<br />

clinical evidence. J Pediatr Gastroenterol Nutr 1999; 28:<br />

116–119.<br />

84. Milman N, Rosenstock S, Andersen L et al. Serum<br />

ferritin, hemoglobin, and <strong>Helicobacter</strong> <strong>pylori</strong> infection: a<br />

seroepidemiologic survey comprising 2794 Danish<br />

adults. Gastroenterology 1998; 115: 274–278.<br />

85. Choe YH, Kwon YS, Jung MK et al. <strong>Helicobacter</strong> <strong>pylori</strong>associated<br />

iron-deficiency anemia in adolescent female<br />

athletes. J Pediatr 2001; 139: 100–104.<br />

86. Patel P, Mendall MA, Khulusi S et al. <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection in childhood: risk factors and effect on<br />

growth. BMJ 1994; 309: 1119–1123.<br />

87. Oderda G, Palli D, Saieva C et al. Short stature and<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection in italian children:<br />

prospective multicentre hospital based case–control<br />

study. The Italian Study Group on Short Stature and H.<br />

<strong>pylori</strong>. BMJ 1998; 317: 514–515.<br />

88. Kerr JR, Al-Khattaf A, Barson AJ, Burnie JP. An association<br />

between sudden infant death syndrome (SIDS) and<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection. Arch Dis Child 2000; 83:<br />

429–434.<br />

89. Elitsur Y. <strong>Helicobacter</strong> <strong>pylori</strong> and SIDS: the jury is in at<br />

last! Am J Gastroenterol 2002; 97: 1576–1577.


90. Dohil R, Hassal E, Jevon G, Dimmick J. Gastritis and<br />

gastropathy of childhood. J Pediatr Gastroenterol Nutr<br />

1999; 29: 378–394.<br />

91. Dixon MF, Genta RM, Yardley JH, Correa P.<br />

Classification and grading of gastritis. The updated<br />

Sydney system. Am J Surg Pathol 1996; 20: 161–81.<br />

92. Marshall BJ, Warren JR, Francis GJ et al. Rapid urease<br />

test in the management of Campylobacter <strong>pylori</strong>-associated<br />

gastritis. Am J Gastroenterol 1987; 82: 200–210.<br />

93. Luzza F, Oderda G, Maletta M et al. Salivary<br />

immunoglobulin G assay to diagnose <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection in children. J Clin Microbiol 1997; 35:<br />

3358–3360.<br />

94. Oderda G, Rapa A, Ronchi B et al. Detection of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> in stool specimens by non-invasive<br />

antigen enzyme immunoassay in children: multicentre<br />

Italian study. BMJ 2000; 320: 347–348.<br />

95. Ni Y-H, Lin J-T, Huang S-F et al. Accurate diagnosis of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection by stool antigen test and 6<br />

other currently available tests in children. J Pediatr<br />

2000; 136: 823–827.<br />

96. Gold BD, Colletti RB, Abbott M et al. <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection in children: recommendations for diagnosis<br />

and treatment. [Medical Position Papers: The North<br />

American Society for Pediatric Gastroenterology and<br />

Nutrition]. J Pediatr Gastroenterol Nutr 2000; 31:<br />

490–497.<br />

97. Drumm B, Koletzko S, Oderda S, on behalf of the<br />

European Pediatric Task force on <strong>Helicobacter</strong> <strong>pylori</strong>.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection in children: a consensus<br />

statement. J Pediatr Gastroenterol Nutr 2000; 30:<br />

207–213.<br />

98. Sherman P, Hassall E, Hunt RH et al. Canadian<br />

<strong>Helicobacter</strong> Study Group Consensus Conference on the<br />

Approach to <strong>Helicobacter</strong> <strong>pylori</strong> infection in Children<br />

and Adolescents. Can J Gastroenterol 1999; 13: 553–559.<br />

99. Bazzoli F, Cecchini L, Corvaglia L et al. Validation of the<br />

13C-urea breath test for the diagnosis of <strong>Helicobacter</strong><br />

<strong>pylori</strong> infection in children: a multicenter study. Am J<br />

Gastroenterol 2000; 95: 646–650.<br />

100. Malfertheiner P, Megraud F, O’Morain C et al. Current<br />

concepts in the management of <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection – the Maastricht 2-2000 Consensus Report.<br />

Aliment Pharmacol Ther 2002; 16: 167–180.<br />

101. Ganga-Zandzou PS, Michaud L, Vincent P et al. Natural<br />

outcome of <strong>Helicobacter</strong> <strong>pylori</strong> infection in asymptomatic<br />

children: a two-year follow-up study. Pediatrics<br />

1999; 104: 216–222.<br />

102. Malfertheiner P, O’ Connor HJ, Genta MR et al.<br />

Symposium: <strong>Helicobacter</strong> <strong>pylori</strong> and clinical risks –<br />

focus on gastro-esophageal reflux disease. Aliment<br />

Pharmacol Ther 2002; 16 (Suppl. 3): 1–10.<br />

103. Labenz J, Blum AL, Bayerdorffer E et al. Curing<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection in patients with duodenal<br />

ulcer may provoke reflux esophagitis. Gastroenterology<br />

1997; 112: 1442–1447.<br />

104. Moayyedi P, Bardhan C, Young L et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> eradication does not exacerbate reflux symptoms<br />

in gastroesophageal reflux disease. Gastroenterology<br />

2001; 121: 1120–1126.<br />

105. Wewer V, Andersen LP, Paerregaard A et al. Treatment of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> in children with recurrent abdominal<br />

pain. <strong>Helicobacter</strong> 2001; 6: 244–248.<br />

106. Oderda G, Rapa A, Bona G. A systematic review of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> eradication treatment schedules in<br />

References 93<br />

children. Aliment Pharmacol Ther 2000; 14 (Suppl 3):<br />

59–66.<br />

107. Gottrand F, Kalach N, Spyckerelle C et al. Omeprazole<br />

combined with amoxicillin and clarithromycin in the<br />

eradication of <strong>Helicobacter</strong> <strong>pylori</strong> in children with<br />

gastritis: a prospective randomized double-blind trial. J<br />

Pediatr 2001; 139: 664–668.<br />

108. Walsh D, Goggin N, Rowland M et al. One week treatment<br />

for <strong>Helicobacter</strong> <strong>pylori</strong> infection. Arch Dis Child<br />

1997; 76: 352–355.<br />

109. Roggero P, Bonfiglio A, Luzzani S et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> stool antigen test: a method to confirm eradication<br />

in children. J Pediatr 2002; 140: 775–777.<br />

110. Correa P. Human gastric carcinogenesis: a multistep and<br />

multifactorial process. First American Cancer Society<br />

Award Lecture on Cancer Epidemiology and Prevention.<br />

Cancer Res 1992; 52: 6735–6740.<br />

111. Woolf CM. A further study on the familial aspects of<br />

carcinoma of the stomach. Am J Hum Genet 1956; 8:<br />

102–109.<br />

112. Huang J-Q, Sridhar S, Chen Y et al. Meta-analysis of the<br />

relationship between <strong>Helicobacter</strong> <strong>pylori</strong> seropositivity<br />

and gastric cancer. Gastroenterology 1998; 114:<br />

1169–1179.<br />

113. <strong>Helicobacter</strong> and Cancer Collaborative Group. Gastric<br />

cancer and <strong>Helicobacter</strong> <strong>pylori</strong>: a combined analysis of<br />

12 case control studies nested within prospective<br />

cohorts. Gut 2001; 49: 347–353.<br />

114. Leung WK, Sung JJY. Intestinal metaplasia and gastric<br />

carcinogenesis. Aliment Pharm Ther 2002; 16:<br />

1209–1216.<br />

115. Shiao YH, Rugge M, Correa P et al. p53 alteration in<br />

gastric precancerous lesions. Am J Pathol 1994; 144:<br />

511–517.<br />

116. Hansson LE, Nyren O, Hsing AW et al. The risk of<br />

stomach cancer in patients with gastric or duodenal<br />

ulcer disease. N Engl J Med 1996; 335: 242–249.<br />

117. El-Omar EM, Carrington M, Chow WH et al.<br />

Interleukin-1 polymorphisms associated with increased<br />

risk of gastric cancer. Nature 2000; 404: 398–402.<br />

118. Forman D. Should we go further and screen and treat?<br />

Eur J Gastroenterol Hepatol 1999; 11(Suppl 2): S69–S71.<br />

119. Parsonnet J, Hansen S, Rodriguez L et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> infection and gastric lymphoma. N Engl J Med<br />

1994; 330: 1267–1271.<br />

120. Bayerdorffer E, Neubauer A, Rudolph B et al. Regression<br />

of primary gastric lymphoma of mucosa-associated<br />

lymphoid tissue type after cure of <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection. Lancet 1995; 345: 1591–1594.<br />

121. Blecker U, McKeithan TW, Hart J, Kirschner BS.<br />

Resolution of <strong>Helicobacter</strong> <strong>pylori</strong>-associated gastric<br />

lymphoproliferative disease in a child. Gastroenterology<br />

1995; 109: 973–977.<br />

122. Blanchard TG, Czinn SJ. Immunology of <strong>Helicobacter</strong><br />

<strong>pylori</strong> and prospects for vaccine, Gastroenterol Clin<br />

North Am 2000; 29: 671–685.<br />

123. Michetti P, Kreiss C, Kotloff KL et al. Oral immunization<br />

with urease and Escherichia coli heat-labile enterotoxin<br />

is safe and immunogenic in <strong>Helicobacter</strong> <strong>pylori</strong>-infected<br />

adults, Gastroenterology 1999; 116: 804–812.<br />

124. Bumann D, Metzger WG, Mansouri E et al. Safety and<br />

immunogenicity of live recombinant Salmonella enterica<br />

serovar Typhi Ty21a expressing a urease A and B<br />

from <strong>Helicobacter</strong> <strong>pylori</strong> in human volunteers Vaccine<br />

2001; 20: 845–852.


7<br />

Introduction<br />

Other gastritides<br />

Salvatore Cucchiara and Osvaldo Borrelli<br />

Epithelial damage, mucosal inflammation and<br />

epithelial cell regeneration represent the histological<br />

response of the stomach to injury. The term<br />

‘gastritis’ implies the microscopic evidence of<br />

inflammation, in which all processes of mucosal<br />

response to injury are present, whereas the term<br />

‘gastropathy’ is used for conditions in which<br />

epithelial injury is associated with cellular regeneration<br />

and the inflammation is not the prominent<br />

feature. 1 The definite diagnosis of gastritis is thus<br />

exclusively based on histological assessment of<br />

biopsy specimens, whereas gastropathies are occasionally<br />

diagnosed on endoscopic appearance. For<br />

this reason, great confusion has been generated by<br />

the inappropriate use of the term gastritis.<br />

Gastritis and gastropathy were previously classified<br />

either as primary (idiopathic) or secondary on<br />

the basis of the underlying etiology. 2 It is now clear<br />

that most cases of unexplained gastritis are caused<br />

by previously unrecognized <strong>Helicobacter</strong> <strong>pylori</strong> (H.<br />

<strong>pylori</strong>) infection. Recently, an endoscopic classification<br />

of gastritis and gastropathy, has been<br />

proposed, that differentiates between erosive<br />

and/or hemorrhagic gastritis or gastropathy and<br />

non-erosive gastritis or gastropathy. 3 The<br />

confounding aspect lies in the fact that some disorders<br />

can be either erosive or non-erosive. The most<br />

common causes of gastritis are summarized in<br />

Table 7.1.<br />

Infectious gastritides<br />

Bacterial gastritides<br />

<strong>Helicobacter</strong> heilmannii<br />

Although H. <strong>pylori</strong> is by far the most common<br />

agent in bacterial gastritides (discussed in detail in<br />

Chapter 6), a spirochete-like organism, <strong>Helicobacter</strong><br />

heilmannii (formerly Gastrospirillum<br />

hominis), has been found to infect the human<br />

stomach. H. heilmannii is closely related to<br />

<strong>Helicobacter</strong> types found in dogs and cats, such as<br />

<strong>Helicobacter</strong> felis. In a pediatric population, Oliva<br />

et al found a prevalence of infection of 0.3%,<br />

whereas in domestic cats and dogs the frequency<br />

of infection was up to 100%. 4,5 H. heilmannii<br />

infection is thought to be a zoonosis, and a<br />

fecal–oral route of transmission is suggested.<br />

Recently, van Loon et al found an identical H. heilmannii<br />

strain in a 5-year-old boy and his cats. 6<br />

Histology shows a typical chronic inflammation,<br />

milder than H. <strong>pylori</strong> gastritis, and the presence of<br />

a spiral organism with four to six coils per cell and<br />

up to 12 flagella at each pole. The bacterium is<br />

usually recognized focally and in small groups in<br />

the foveola, maintaining a large distance from the<br />

surface epithelium. Endoscopic features such as<br />

erosions, peptic ulcerations and antral nodularity<br />

have rarely been reported. Eradication treatment,<br />

as with H. <strong>pylori</strong> infection, should be considered in<br />

both humans and animals.<br />

Tuberculosis gastritis<br />

Tuberculosis infection of the stomach is very rare<br />

and usually occurs in patients with immune<br />

deficiency or after transplantation. 7 In adults,<br />

gastric tuberculosis typically involves the antrum<br />

and extends to the lesser curvature, although in<br />

patients with AIDS it may also involve the esophagogastric<br />

junction. Endoscopic features include<br />

hypertrophic nodular lesions and multiple ulcers,<br />

whereas outlet obstruction, caused by pre<strong>pylori</strong>c<br />

narrowing or mass effect, has rarely been<br />

described. Histology can show both caseous and<br />

non-caseous granulomas. Culture of biopsy specimens<br />

is essential to establish the diagnosis,<br />

95


96<br />

Other gastritides<br />

Table 7.1 Common causes of gastritis and gastropathy<br />

Infectious gastritis<br />

Bacteria <strong>Helicobacter</strong> <strong>pylori</strong><br />

<strong>Helicobacter</strong> heilmannii<br />

Mycobacterium tuberculosis,<br />

Mycobacterium avium-intracellulare<br />

Viruses cytomegalovirus<br />

herpes simplex virus<br />

varicella zoster virus<br />

Fungi – Protozoa Candida albicans<br />

Histoplasma, Mucormycosis<br />

Anisakis simplex<br />

Giardia lamblia<br />

Cryptosporidium<br />

Drugs and other agents<br />

Aspirin and other non-steroidal anti-inflammatory drugs<br />

Corticosteroids<br />

Potassium chloride, calcium<br />

Chemotherapeutic agents, antibiotics, prostaglandin E1<br />

Ethanol, cocaine, methamphetamines<br />

Gastrostomy tubes<br />

Endoscopic procedures<br />

Foreign-body ingestion<br />

Stress<br />

Ménétrier’s disease<br />

Bile reflux gastropathy<br />

<strong>Portal</strong> hypertensive gastropathy<br />

Zollinger–Ellison and pseudo-Zollinger–Ellison syndrome<br />

Lymphocytic gastritis<br />

Chronic varioliform gastritis<br />

Celiac gastritis<br />

<strong>Helicobacter</strong> <strong>pylori</strong> lymphocytic gastritis<br />

Crohn’s disease<br />

Chronic granulomatous disease<br />

Eosinophilic gastritis<br />

Collagenous gastritis<br />

Proton pump inhibitor gastritis<br />

Pernicious anemia<br />

Gastritis associated with other autoimmune diseases<br />

Graft-versus-host disease


ecause the organism may not be seen microscopically.<br />

Organisms of the Mycobacterium avium–intracellular<br />

complex are common pathogens in immunosuppressed<br />

patients, and mainly occur in the small<br />

and large intestines. However, the stomach may<br />

occasionally be involved, with refractory gastric<br />

ulcerations. 8<br />

Viral gastritides<br />

Viral infections of the stomach typically affect<br />

immunocompromised or severely debilitated children.<br />

Their frequency substantially increases in<br />

such children, whereas they are quite rare in<br />

healthy subjects. Rare cases of gastric involvement<br />

by herpes simplex virus (HSV) or disseminated<br />

varicella zoster virus (VZV) infections have been<br />

reported. 9,10 Bleeding from hemorrhagic gastropathy<br />

has been described in children with influenza<br />

A infection. 11 Cytomegalovirus (CMV) is the main<br />

recognizable viral agent, occurring in both healthy<br />

and immunodeficient children, and has been<br />

involved as an etiologic agent in Ménétrier’s<br />

disease.<br />

Fungal and protozoan gastritides<br />

Fungal infections, such as Candida albicans, histoplasmosis<br />

and mucormycosis, rarely affect the<br />

stomach, although bleeding, gastric ulcers and<br />

perforation during disseminated infections have<br />

been described in preterm or sick neonates, as well<br />

as in malnourished or immunodeficient children.<br />

In adults, acute gastric anisakiasis frequently<br />

occurs in countries where the consumption of raw<br />

or undercooked fish is high. It is caused by ingesting<br />

the organism’s larvae. Because humans represent<br />

an aberrant host, it is a dead-end infection<br />

characterized by an intense acute eosinophilic<br />

inflammation. Acute gastrointestinal symptoms,<br />

such as upper abdominal pain, nausea and vomiting<br />

occur within a few hours, and acute anaphylactic<br />

reactions have been described in patients<br />

previously exposed to the organism. 12 Endoscopic<br />

findings include multiple erosions, edema and<br />

single or multiple worms, while histology shows<br />

an acute eosinophilic inflammation. The removal<br />

Gastropathies due to drugs 97<br />

of worms with endoscopic forceps induces rapidly<br />

symptomatic relief.<br />

Gastric colonization of Giardia lamblia has been<br />

reported in 0.3% of adults undergoing upper<br />

gastrointestinal endoscopy. 13 Because giardiasis is<br />

associated with intestinal metaplasia of the<br />

stomach, some authors suggest that this organism<br />

is not a gastric pathogen.<br />

Gastric outlet obstruction due to enteric cryptosporidiosis<br />

has been reported in immunocompromised<br />

patients. 14<br />

Gastropathies due to drugs, toxins<br />

and other agents<br />

Acetylsalicylic acid and other non-steroidal<br />

anti-inflammatory drugs<br />

It is well known that acetylsalicylic acid and other<br />

non-steroidal anti-inflammatory drugs (NSAIDs)<br />

can cause gastric injury including gastritis, gastric<br />

ulcers and gastrointestinal hemorrhage.<br />

Gastroduodenal damage can be seen at endoscopy<br />

in 20–40% of adults taking NSAIDs, and the<br />

overall risk for peptic complications (hemorrhage<br />

and perforation) in these patients is about three<br />

times greater than in controls. 15 There are no data<br />

on the frequency of upper gastrointestinal injury<br />

in children related to NSAID use. Because of the<br />

risk of Reye syndrome and the availability of<br />

different types of antipyretic drugs, the use of<br />

acetylsalicylic acid and NSAIDs in children has<br />

significantly diminished.<br />

The routine use of these drugs in different clinical<br />

conditions is associated with severe complications<br />

such as mucosal erosions and ulceration, and<br />

gastrointestinal bleeding. Endoscopic features<br />

include hemorrhagic gastric erosions (most often<br />

in the corpus) and gastric ulcers (mainly in the<br />

antrum). Histology shows foveolar hyperplasia,<br />

edema, vascular dilatation and congestion,<br />

increased smooth muscle fibers in the lamina<br />

propria and, characteristically, a relative paucity of<br />

inflammation. The term ‘chemical gastritis’ is<br />

usually employed for this condition.<br />

Several mechanisms are thought to play a role in<br />

the pathogenesis of NSAID-induced injury,


98<br />

Other gastritides<br />

including inhibition of prostaglandin (PG) synthesis,<br />

increased platelet-activating factor, platelet<br />

dysfunction, increased oxygen free radicals,<br />

increased neutrophil adherence, decreased<br />

mucosal blood flow and topical irritant effects. 16<br />

Endogenous PGs are well-known mucosal defense<br />

factors, protecting the gastric mucosa against<br />

injury caused by a variety of toxic stimuli. PGs are<br />

synthesized through cyclo-oxygenase (COX),<br />

which is the target of NSAIDs. Two isoforms of<br />

COX molecules (COX-1 and COX-2) have been<br />

recognized. COX-1 is a constitutively expressed<br />

enzyme in many tissues, including the gastrointestinal<br />

tract, and is usually active, while COX-2<br />

is an inducible enzyme predominantly expressed<br />

at the site of inflammation. It has been shown that<br />

the side-effects of NSAIDs are related to COX-1<br />

affinity, while their therapeutic effects are dependent<br />

on activity against COX-2. 17<br />

Corticosteroids<br />

The association between peptic ulcer disease and<br />

corticosteroids has never been accepted unequivocally.<br />

Ng et al described two cases of gastric perforation<br />

in preterm babies treated with dexamethasone<br />

for bronchopulmonary dysplasia. 18 A<br />

meta-analysis of 71 controlled studies performed<br />

by Messer et al demonstrated an increased risk for<br />

gastrointestinal ulceration and hemorrhage among<br />

adult patients who had received corticosteroids,<br />

and the risk was correlated with the dosage<br />

employed. 19 Although to date there are no similar<br />

studies in childhood it is commonly agreed that,<br />

when steroids are administered to children for<br />

long periods, prophylactic therapy for gastric<br />

mucosal damage should be started concomitantly<br />

(see p.109).<br />

Other agents<br />

Gastropathies can be caused by a variety of drugs<br />

including potassium chloride, calcium, valproic<br />

acid, chemotherapeutic agents and antibiotics,<br />

such as penicillin, chloramphenicol, sulphonamides,<br />

tetracyclines and cephalosporins. In<br />

neonates the administration of PGE1 can induce<br />

gastric outlet obstruction due to antral hyperplasia.<br />

20<br />

In experimental animals, exposure to a high<br />

concentration of ethanol rapidly produces grossly<br />

evident focal hemorrhagic erosions. Chronic ingestion<br />

of alcohol may also result in human gastric<br />

pathology. Endoscopic findings include subepithelial<br />

hemorrhages (‘blood under plastic wrap’),<br />

usually in the fundus and corpus, and small whitebased<br />

erosions. 21 Histology reveals areas of subepithelial<br />

hemorrhage, associated with superficial<br />

and deep epithelial damage, and marked edema of<br />

the surrounding mucosa. Inflammatory cell infiltrate<br />

is usually minimal. Experimental studies in<br />

animals have shown that alteration of the mucosal<br />

microcirculation might be an early event in the<br />

evolution of ethanol-induced gastric injury, as<br />

suggested by the occurrence of focal damage of<br />

mucosal capillaries and increased permeability<br />

within 1 or 2 min after intragastric instillation of a<br />

high concentration of ethanol. 22<br />

Gastric ulcerations have been reported in association<br />

with cocaine and methamphetamine abuse. In<br />

children requiring enteral nutrition, ulcers and<br />

small multiple erosions, usually localized in the<br />

proximal stomach, have been described after<br />

placement of gastrostomy feeding tubes.<br />

Subepithelial hemorrhages and focal erosions are<br />

commonly detected after ingestion of a foreign<br />

body or endoscopic procedures.<br />

Stress gastropathy<br />

Although the overall prevalence is unknown, most<br />

critically ill infants and children are prone to<br />

develop stress-related gastropathy. In infants,<br />

stress gastropathy is usually related to traumatic<br />

delivery, respiratory or cardiac failure, sepsis,<br />

hypoglycemia or dehydration, while in older children<br />

it is related to severe life-threatening illness<br />

(e.g. respiratory or cardiac failure), intracranial<br />

lesion, trauma, burns, coagulopathy, or vasculitis.<br />

23 Endoscopic features include isolated gastroduodenal<br />

erosions, usually in the fundus and<br />

proximal body, and, in severe cases, mucosal<br />

ulcerations occurring at multiple sites within the<br />

stomach and duodenum (Figure 7.1). The majority<br />

of patients develop erosions and ulcers within<br />

minutes to hours after the initial insult, and the<br />

most common presenting symptoms are<br />

hematemesis and melena rather than abdominal


Figure 7.1 Congested gastric mucosa in an infant with<br />

stress hemorrhagic vomiting. The mucosa shows nonconfluent<br />

cherry red spots on a finely granular background.<br />

pain. Recently, Chaibou et al showed that more<br />

than 10% of children admitted to an intensive care<br />

unit experienced upper gastrointestinal bleeding,<br />

but the bleeding was clinically significant in only<br />

1.6% of them. 24<br />

Alterations of the mucosal microcirculation and<br />

mucosal ischemia have been implicated in the<br />

pathogenesis of stress-induced gastropathy, even<br />

though other mechanisms have been suggested,<br />

such as increased acid output, decreased production<br />

of mucus and impairment of local<br />

prostaglandin synthesis.<br />

Ménétrier’s disease<br />

Ménétrier’s disease, a rare disorder of unknown<br />

etiology, is characterized by enlarged gastric folds<br />

due to mucosal thickening in the gastric body. To<br />

date, fewer than 100 pediatric cases have been<br />

described. Significant differences between adult<br />

and pediatric disease in terms of onset, presentation,<br />

course and prognosis have been<br />

observed. 25,26 In children, the disease often begins<br />

abruptly and is usually self-limited, with resolution<br />

of clinical features within weeks or months. In<br />

contrast, in adults, Ménétrier’s disease is a persis-<br />

Ménétrier’s disease 99<br />

tent and more severe condition often associated<br />

with gastrointestinal bleeding, in most cases<br />

requiring surgical resection.<br />

Although this disease has also been reported in the<br />

neonatal period, the mean age of onset is 5 years<br />

(ranging from 2 days to 17 years). Common<br />

features include vomiting, abdominal pain,<br />

anorexia, hypoproteinemia and, in some cases,<br />

eosinophilia and raised IgE levels. Several studies<br />

have confirmed that hypoproteinemia is due to a<br />

non-selective increased loss of proteins through<br />

the gastric mucosa, occurring in a paracellular<br />

fashion via widened tight junctions. 27 In childhood,<br />

hematemesis and melena occur rarely,<br />

whereas in adults the incidence of gastrointestinal<br />

bleeding ranges from 20 to 40%.<br />

The etiology of Ménétrier’s disease is unknown,<br />

although its acute presentation and self-limited<br />

clinical course in childhood suggest an infectious<br />

cause. Nearly 30% of affected children show<br />

evidence of CMV infection, either through the<br />

presence of characteristic inclusion bodies and<br />

CMV antigen in the gastric tissue or by serology. 25<br />

It has also been proposed that an increase in both<br />

serum IgE levels and the number of peripheral<br />

blood eosinophils might be an idiosyncratic antibody-mediated<br />

response to the penetration of the<br />

gastric mucosa by allergens after the cytopathic<br />

effect of CMV on the gastric mucosa.<br />

H. <strong>pylori</strong> infection has also been reported in children<br />

with enlarged gastric folds and protein-losing<br />

enteropathy, and its eradication has resulted in<br />

disease resolution. 28 However, the classic clinical<br />

and histological features of Ménétrier’s disease<br />

were absent in all reported cases.<br />

Insights into the disease pathogenesis have been<br />

gained through the investigation of the mediators<br />

of gastric mucosal growth and function, such as<br />

transforming growth factor-α (TGFα). The latter<br />

stimulates gastric epithelial cell proliferation and<br />

mucin secretion, and inhibits parietal cell acid<br />

production, in transgenic mice reproducing many<br />

altered mucosal features of Ménétrier’s disease. As<br />

in adult patients, elevated levels of TGFα have<br />

been found in gastric mucosal biopsies taken from<br />

children with Ménétrier’s disease, suggesting a<br />

role for this polypeptide in the development and<br />

progression of the disesase. 29


100<br />

Other gastritides<br />

Upper gastrointestinal X-ray series identifies the<br />

characteristic gastric rugae hypertrophy, predominantly<br />

in the fundus and in the corpus of the<br />

stomach. Thickened gastric mucosal folds have<br />

also been demonstrated by additional diagnostic<br />

imaging modalities, such as ultrasound and<br />

computed axial tomography. Endoscopy and<br />

histology should be performed in all patients to<br />

confirm the diagnosis, to search for the pathogen<br />

and to exclude different diagnoses, such as<br />

eosinophilic gastritis, primary gastric lymphoma,<br />

gastric carcinoma, Crohn’s disease of the stomach<br />

and other conditions in which hypertrophic<br />

gastric rugae occur. Endoscopy shows swollen<br />

gastric folds predominantly in the greater curvature<br />

of the stomach or within the entire body<br />

region of the stomach, large quantities of gelatinous<br />

mucus and, less commonly, multiple areas of<br />

focal erosions in the fundus. Histology usually<br />

shows considerable elongation and complexity of<br />

the superficial epithelium, so-called foveolar<br />

hyperplasia (gastric pit), with reduction of chief<br />

and parietal cell glands and often with cystic<br />

dilatation, which may extend into the submucosa.<br />

An inflammatory infiltrate of eosinophils,<br />

neutrophils, lymphocytes and, occasionally,<br />

plasma cells is found in the edematous lamina<br />

propria.<br />

Bile reflux gastropathy<br />

This is also known as alkaline gastritis and<br />

describes an ongoing, chronic condition in which<br />

bile-containing intestinal contents reflux into the<br />

stomach. In animal studies it has been shown that<br />

bile salts and other intestinal contents, such as<br />

lysolecitin, break down the gastric mucosal<br />

barrier, leading to back-diffusion of H + ions and<br />

histological injury. Bile acid salts are also potent<br />

releasers of histamine and other mediators from<br />

mast cells and induce non-specific cytotoxicity. 30<br />

Duodenogastric bile reflux is thought to be a physiological<br />

event during the second phase of the<br />

interdigestive motor complex and during the postprandial<br />

phase, but it is rapidly flushed out during<br />

the third phase of the interdigestive period. It may<br />

be considered abnormal when it is frequent, when<br />

it is abundant or when it is associated with<br />

delayed gastric emptying.<br />

Typical endoscopic findings are ‘beefy’ redness or<br />

erythema and, occasionally, erosions, but the presence<br />

of bile in the stomach during endoscopy does<br />

not have clinical significance. The distinctive<br />

histological features, also defined as ‘chemical<br />

gastropathy’, include foveolar elongation, complexity<br />

and hypercellularity, together with edema,<br />

vasodilatation and a paucity of acute and chronic<br />

inflammatory cells in the lamina propria. 31 These<br />

findings do not correlate with gastric bile concentration.<br />

The diagnosis of bile reflux gastropathy should be<br />

considered in adults with abdominal pain and<br />

bile-stained vomiting, who had previously undergone<br />

partial gastric resection or drainage procedures,<br />

even though reflux of duodenal contents<br />

has been linked to the development of a number of<br />

pathological gastric conditions, such as nonspecific<br />

gastritis, gastric ulcer, gastric carcinoma<br />

and non-ulcer dyspepsia. In adults, an association<br />

between bile reflux and intestinal metaplasia in<br />

the gastric and cardiac mucosa has been reported,<br />

suggesting that the latter represents a defense<br />

response against a sustained adverse environment,<br />

in the same way that gastric metaplasia develops<br />

in the duodenum when subjected to a high acid<br />

load. 32<br />

<strong>Portal</strong> hypertensive gastropathy<br />

Involvement of the gastric mucosa is common in<br />

children with intrahepatic or extrahepatic causes<br />

of portal hypertension. 33 Endoscopic findings vary<br />

from mild involvement, including a snake-skin<br />

mosaic pattern of the mucosa, a fine pink speckling<br />

and superficial erythema (scarlatina-type<br />

rash), to a severe gastropathy, defined by cherry<br />

red spots with a diffuse confluence of reddened<br />

areas and a hemorrhagic appearance. These<br />

patterns seem to be specific for portal hypertensive<br />

gastropathy and have not been found in any of 500<br />

endoscopic examinations performed in children<br />

without hepatic disease. The fundus and corpus<br />

are usually involved, although antral involvement<br />

is occasionally observed. Histologically, portal<br />

hypertensive gastropathy is characterized by<br />

ectasia of mucosal capillaries and venules and by<br />

submucosal venous dilatation, with no acute or<br />

chronic inflammation.


Hemodynamic disturbances are thought to be<br />

involved in the pathogenesis of portal hypertensive<br />

gastropathy, because gastric perfusion alterations<br />

parallel the severity of mucosal involvement<br />

and previous endoscopic sclerotherapy of<br />

esophageal varices both in adults and children<br />

may exacerbate mucosal congestion and the severity<br />

of gastropathy. 34<br />

Zollinger–Ellison and pseudo-<br />

Zollinger–Ellison syndromes<br />

It is intuitive that excessive acid production can<br />

result in multiple erosions and frank ulcers in the<br />

stomach, duodenum and jejunum. Although rare,<br />

this condition may arise from a gastrinoma<br />

(Zollinger–Ellison syndrome (ZES)) or from antral<br />

gastrin cell hyperfunction with or without<br />

hyperplasia (pseudo-Zollinger–Ellison syndrome<br />

(PZES)).<br />

ZES is the most frequent ulcerogenic syndrome<br />

associated with increased gastrin levels and, in<br />

adults, represents the most common of the malignant<br />

islet cell tumors. 35 ZES is characteristically<br />

associated with increased levels of serum gastrin<br />

(hypergastrinemia), which, in turn, causes an overproduction<br />

of gastric acid and results in complicated<br />

ulcer disease. As would be expected, the<br />

most common presenting signs are abdominal<br />

pain, nausea, vomiting, hematemesis and melena.<br />

Diarrhea occurs less commonly in childhood than<br />

in adulthood. ZES can be sporadic or associated<br />

with multiple endocrine neoplasia type I (MEN I).<br />

ZES-associated MEN-I patients often become<br />

symptomatic in childhood and at an earlier age<br />

than those with the sporadic form. Thus, if<br />

patients show severe peptic ulceration, kidney<br />

stones, watery diarrhea and malabsorption with a<br />

positive family history of endocrinopathy, MEN-I<br />

and nephrolithiasis, ZES should be strongly<br />

suspected. The diagnosis of ZES is made if the<br />

patient shows an elevated fasting serum gastrin<br />

level, gastric acid hypersecretion and either a positive<br />

secretin test or a histologically proven gastrinoma.<br />

36 Typically, patients have fasting serum<br />

gastin levels of > 100 pg/ml, even though adult<br />

patients usually show a level greater than<br />

500 pg/ml, a level greater than 1000 pg/ml is nearly<br />

diagnostic of the disease. The basal acid output<br />

Zollinger–Ellison syndrome 101<br />

(BAO) is generally more than 10 mEq/h. The<br />

secretin provocative test (2units/kg, intravenously)<br />

has a high diagnostic value; in adults, a rise in the<br />

serum gastrin level of 200 pg/ml above the fasting<br />

concentration is considered a positive diagnosis.<br />

Endoscopic findings are characterized by multiple<br />

peptic ulcers, most of which are localized in the<br />

duodenum, but the stomach and jejunum are<br />

involved less commonly. Recent developments of<br />

both radiological and nuclear medicine studies<br />

have increased the capability for identifying<br />

neoplastic lesions in the majority of patients with<br />

ZES. Recommended imaging procedures are<br />

computed tomography (CT), magnetic resonance<br />

imaging (MRI) and somatostatin receptor scintigraphy<br />

(octreoscan); more recently, endoscopic ultrasound<br />

has been proposed as the most sensitive<br />

imaging modality. 37<br />

‘PZES’ is usually used for two distinct entities:<br />

antral G-cell hyperplasia, described as an<br />

increased number of G cells, and antral G-cell<br />

hyperfunction, in which hypergastrinemia occurs<br />

in the absence of detectable G-cell hyperplasia. 38<br />

The clinical manifestation of PZES varies from<br />

vague features, such as non-specific abdominal<br />

pain, vertigo, anemia and occult bleeding, to<br />

severe gastrointestinal bleeding. The differentiation<br />

between ZES and PZES is based on the<br />

response to provocative tests. PZES is characterized<br />

by exaggerated serum gastrin release in<br />

response to a feeding test, and unchanged or even<br />

depressed serum gastrin values after injection of<br />

secretin.<br />

Lymphocytic gastritis<br />

Lymphocytic gastritis is characterized by an<br />

increase of intraepithelial lymphocytes in surface<br />

and foveolar epithelium, together with a variable<br />

amount of inflammation in the lamina propria of<br />

the gastric mucosa, which ranges from a predominantly<br />

lymphocytic pattern to a mixed chronic<br />

active pattern. The generally accepted criterion for<br />

the diagnosis of lymphocytic gastritis is the<br />

finding of 25 or more lymphocytes per 100 epithelial<br />

cells. 39<br />

Lymphocytic gastritis is thought to be a histologic<br />

response of the gastric mucosa to several antigens.<br />

The main interest comes from its association with


102<br />

Other gastritides<br />

Table 7.2 Conditions associated with lymphocytic gastritis<br />

Varioliform gastritis<br />

Celiac disease<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection<br />

Ménétrier’s disease<br />

Drug administration (ticlopidine)<br />

seemingly diverse groups of disorders (Table 7.2),<br />

among them celiac disease, where this type of<br />

gastritis is very prevalent (see below).<br />

Chronic varioliform gastritis<br />

Chronic varioliform gastritis is a rare disorder of<br />

unknown origin, commonly affecting middle-aged<br />

and elderly men. To date, few cases have been<br />

reported in the pediatric literature. In children,<br />

clinical signs arise insidiously, are often subacute<br />

or chronic and include epigastric pain, nausea<br />

and vomiting, anorexia, weight loss, anemia,<br />

protein-losing enteropathy and, in some cases,<br />

peripheral eosinophilia and increased serum IgE<br />

levels. 40<br />

The endoscopic features of chronic varioliform<br />

gastritis are enlarged and thickened rugal folds<br />

bearing erosions and widespread small nodules<br />

frequently surmounted by an umbilicated central<br />

crater or small rounded erosions (aphthoid<br />

nodules). According to the topography of the<br />

lesions, three forms can be distinguished: diffuse,<br />

when the whole stomach is involved; corporeal,<br />

when the lesions are limited to the fundus and<br />

proximal body of the stomach; and antral, when<br />

they are present only in the antrum. Histology<br />

shows a focal or diffuse infiltrate of intraepithelial<br />

lymphocytes in the surface and foveolar epithelium.<br />

Throughout the lamina propria an inflammatory<br />

infiltrate consisting of IgE plasma cells,<br />

lymphocytes, neutrophils and eosinophils is<br />

observed.<br />

The etiology of chronic varioliform gastritis is still<br />

unsettled. An immunological mechanism related<br />

to food antigens is suggested by raised serum IgE<br />

levels and increased numbers of IgE staining<br />

plasma cells. However, owing to a decreased incidence<br />

of the disorder and to the fact that the histological<br />

pattern is highly reminiscent of celiac<br />

gastritis (see below), it has been suggested that<br />

chronic varioliform gastritis is only a crude endoscopic<br />

expression of a disease that has the characteristic<br />

features of lymphocytic intraepithelial<br />

infiltration. The macroscopic appearance might<br />

appear only at some periods in the evolution of<br />

the disease.<br />

Celiac gastritis<br />

An association between lymphocytic gastritis and<br />

celiac disease has been reported in both adults and<br />

children. Lymphocytic gastritis is found in up to<br />

45% of adults with celiac disease, with a range of<br />

prevalence from less than 10% to 45%. Wolberg et<br />

al identified ten of 22 adult patients with lymphocytic<br />

gastritis characterized by marked infiltration<br />

of the surface and superficial pit epithelium by<br />

lymphocytes, primarily T cells, with sparing of the<br />

deep glandular epithelium both in the antrum and<br />

in the body. The lamina propria showed an infiltrate<br />

of plasma cells, lymphocytes and rare<br />

neutrophils. 41 Recently, it has been shown that the<br />

pattern of involvement of the gastric mucosa is<br />

predictive of duodenal villous atrophy. Patients<br />

with corpus-predominant lymphocytic gastritis are<br />

unlikely to have duodenal pathology, whereas<br />

those with an antrum-predominant or a diffuse<br />

pattern have a 50% chance of coexistent villous<br />

atrophy. 42 In a study of 60 children with chronic<br />

gastritis, De Giacomo et al found lymphocytic


gastritis in nine of 25 children with celiac disease,<br />

but in none of 35 children without gluten-sensitive<br />

enteropathy. Children with celiac gastritis showed<br />

an average of 40 lymphocytes per 100 epithelial<br />

cells, compared with an average of three to five in<br />

control subjects or patients with H. <strong>pylori</strong>-associated<br />

gastritis. Interestingly, at endoscopy, all children<br />

showed resolution of the lymphocytic infiltrate<br />

after strict adherence to a gluten-free diet. 43<br />

Recently, some authors have suggested that celiac<br />

lymphocytic gastritis may represent an abnormal<br />

immunological response to gliadin. It is now<br />

recognized that there is a spectrum of gluteninduced<br />

intestinal changes, ranging from the<br />

classic ‘celiac’ lesions of total and/or subtotal<br />

villous atrophy to more subtle manifestations,<br />

such as an abnormal density and subtype distribution<br />

of intraepithelial lymphocytes in the small<br />

intestinal mucosa. It is conceivable that lymphocytic<br />

gastritis, such as lymphocytic colitis, is<br />

another manifestation of a mucosal immune<br />

response to a luminal antigen, which is maximally<br />

expressed in the small intestine. 44 Thus, it is<br />

believed that the presence of lymphocytic gastritis<br />

in dyspeptic children may be used as an indication<br />

to perform a small-bowel biopsy to rule out covert<br />

celiac disease.<br />

<strong>Helicobacter</strong> <strong>pylori</strong> lymphocytic gastritis<br />

Some authors have suggested that lymphocytic<br />

gastritis may represent an idiosyncratic immune<br />

response to some local antigen, such as H. <strong>pylori</strong>,<br />

although the relationship is still not clear. It has<br />

been shown that many cases of lymphocytic gastritis<br />

are associated with H. <strong>pylori</strong> infection, and that<br />

the eradication brings about significant reduction<br />

in the gastric intraepithelial lymphocytic infiltration<br />

and dyspeptic symptoms. On the other hand,<br />

in adults and pediatric series, H. <strong>pylori</strong> has been<br />

detected in a minority of patients with lymphocytic<br />

gastritis, whereas the infection was more<br />

frequent in control biopsy specimens.<br />

Crohn’s disease<br />

Although Crohn’s disease (CD) most commonly<br />

affects the terminal ileum and/or the colon,<br />

involvement of the upper gastrointestinal tract is<br />

Crohn’s disease 103<br />

frequently found in both adults and children.<br />

Symptoms such as epigastric pain, early satiety,<br />

nausea, vomiting, weight loss and, less frequently,<br />

hematemesis and melena are commonly reported<br />

in these patients and have previously been related<br />

to reflex inhibition of foregut motility secondary to<br />

inflammation and partial obstruction of the distal<br />

small bowel. It has been shown that macroscopic<br />

and/or histological abnormalities are found in up<br />

to 70% of adults and 90% of children with CD. 45<br />

Endoscopic findings include patchy or streaky<br />

mucosal reddening, edema, single or multiple<br />

nodularities, a cobblestone appearance, rigidity of<br />

the antrum wall, narrowing of the lumen, aphthoid<br />

erosions and linear or serpiginous ulcers (Figure<br />

7.2). Histological diagnosis of gastric CD is best<br />

achieved by the detection of epithelioid granulomas<br />

and giant cells in biopsy specimens, although<br />

focal, non-specific gastritis containing a mixed<br />

inflammatory cell infiltrate is observed in a high<br />

percentage of patients. Endoscopic and/or histological<br />

gastric involvement in CD may be present<br />

in the absence of upper gastrointestinal symptoms<br />

and may precede diagnostic findings in the small<br />

and large bowel. The prevalence of granulomas in<br />

bioptic specimens of adult patients is highly variable,<br />

ranging from 7 to 83%, whereas in children it<br />

is up to 40%. 46 Table 7.3 lists the most common<br />

causes of granulomatous gastritis in children. In<br />

Figure 7.2 Small pre-<strong>pylori</strong>c ulcers (arrows) in a 14year-old<br />

girl with Crohn’s disease.


104<br />

Other gastritides<br />

Table 7.3 Causes of granulomatous gastritis<br />

in children<br />

Systemic disease<br />

Crohn’s disease<br />

Chronic granulomatous disease<br />

Sarcoidosis<br />

Other vasculitides<br />

Infectious causes<br />

Tuberculosis<br />

<strong>Helicobacter</strong> <strong>pylori</strong><br />

Histoplasmosis<br />

Syphilis<br />

Parasites<br />

Isolated<br />

Idiopathic<br />

Foreign substances<br />

recent years a characteristic pattern of inflammation,<br />

the so-called focal enhanced gastritis, has<br />

caused great interest, because of the observation<br />

that it is quite common and is certainly seen with<br />

greater frequency than granulomas in both adults<br />

and children. It is found more commonly in the<br />

antrum than in the body and is characterized by at<br />

least one foveola/gland or small groups of foveolae/glands<br />

surrounded by infiltrated inflammatory<br />

cells that consist mainly of lymphocytes, monocytes<br />

and occasionally neutrophils, resembling<br />

focal inflammation observed in the intestinal and<br />

colonic mucosa of patients with CD.<br />

In adults, two prospective studies have evaluated<br />

the prevalence of focal enhanced gastritis in<br />

patients with CD compared with a control population.<br />

Histological focally enhanced gastritis was<br />

observed in more than half of CD patients, suggesting<br />

that it could be a diagnostic marker of the<br />

disease. 47,48 However, in both studies no patients<br />

with ulcerative colitis were included. Subsequently,<br />

Parente et al found, in an adult population,<br />

focally enhanced gastritis in 43% of H. <strong>pylori</strong>negative<br />

CD patients, 12% of ulcerative colitis<br />

(UC) patients and 19% of controls with no inflammatory<br />

bowel disease (IBD). The authors reported<br />

a specificity and positive predictive value of<br />

focally enhanced gastritis for CD of 84% and 71%,<br />

respectively. 49 These results have been confirmed<br />

in two pediatric studies. Sharif et al showed that<br />

focally enhanced gastritis was present in 65.1% of<br />

children with CD, 20.8% of children with UC and<br />

2.6% of children without IBD. 50 More recently,<br />

Kundhal et al found focally enhanced gastritis in<br />

52% of children with CD and 8% of patients with<br />

UC. 51 This suggested that, although this histological<br />

finding was highly suggestive of CD, it could<br />

not be seen as a specific diagnostic marker.<br />

Chronic granulomatous disease<br />

Chronic granulomatous disease is a rare group of<br />

inherited disorders characterized by impaired<br />

phagocyte oxidative metabolism caused by<br />

missing components or subunits of the NADPH<br />

oxidative complex, resulting in defective intracellular<br />

killing of catalase-positive micro-organisms.<br />

The most common form of chronic granulomatous<br />

disease is inherited as an X-linked recessive trait,<br />

although autosomal-type mutations have been<br />

described. The disease usually appears during the<br />

first two years of life with symptoms and signs of<br />

recurrent pyogenic infections, although milder<br />

forms of the disease have been described, with<br />

onset occurring in adulthood. Characteristic granulomas<br />

may develop in any organ system, including<br />

the skin, lungs, genitourinary, bone and<br />

reticoloendothelial systems. Gastrointestinal involvement<br />

has commonly been described.<br />

Narrowing of the gastric antrum, with signs of<br />

gastric outlet obstruction, such as vomiting,<br />

weight loss and epigastric pain, is a rare but<br />

distinctive feature of chronic granulomatous<br />

disease. 52 Endoscopic features are usually not<br />

specific, whereas histology shows mild edema of<br />

the lamina propria, chronic inflammatory cells<br />

and granulomas characterized by phagocytes,<br />

giant cells and lipid-containing histiocytes. Upper<br />

gastrointestinal series reveal gastric rugae hypertrophy<br />

and narrowing of the antral lumen. Recent<br />

developments of nuclear medicine studies have<br />

increased the capability of evaluating thickness<br />

and inflammatory involvement of the gastric wall.


Eosinophilic gastritis<br />

Eosinophilic gastritis is a component of<br />

eosinophilic gastroenteropathy, a rare disease<br />

characterized by prominent eosinophilic infiltration<br />

of the gastrointestinal tract. The cause is<br />

unknown and the mechanisms responsible for<br />

gastrointestinal infiltration by eosinophils remain<br />

poorly understood. Recently, it has been shown<br />

that the production of eotaxin, a protein with 73<br />

amino acid residues and a member of the chekines<br />

family, at the site of inflammation promotes<br />

recruitment and aggregation of eosinophils in the<br />

tissue by up-regulating integrins and enhancing<br />

eosinophilic adhesion to endothelial cells. Finally,<br />

it may contribute to tissue damage by stimulating<br />

the release of highly cytotoxic granular proteins. 53<br />

It has been suggested that food allergy might be a<br />

triggering factor in childhood. Associations<br />

between eosinophilic gastritis and parasitic infection<br />

of the stomach have been described.<br />

Any part of the gastrointestinal tract, from the<br />

esophagus to the rectum, can be affected, even<br />

though the stomach seems to be the most frequent<br />

site. 54 Eosinophilic infiltration may involve the<br />

mucosa, the muscularis propria and the serosa. On<br />

the basis of the predominant affected layer, the<br />

enteropathy has been classified into three different<br />

forms: mucosal, which produces clinical features<br />

of inflammatory diseases; submucosal, usually<br />

producing obstruction; and serosal, producing<br />

eosinophilic ascites. The clinical picture depends<br />

on the site and depth of the inflammatory involvement.<br />

Gastric involvement is commonly associated<br />

with abdominal pain, bloating, growth failure,<br />

weight loss, nausea and non-bilious vomiting<br />

secondary to gastric outlet obstruction. Gastric<br />

perforation has also been reported. Patients with<br />

associated small bowel involvement may develop<br />

protein-losing enteropathy, malabsorption and<br />

iron-deficiency anemia. In some patients, an<br />

elevated circulating eosinophilic count and raised<br />

serum IgE level can be found, although their clinical<br />

significance remains unclear.<br />

Endoscopy and biopsy are the main ways to establish<br />

the diagnosis. Endoscopic findings, when<br />

present, are non-specific and include erythema,<br />

antral nodular lesions, ulcers and, rarely, a<br />

narrowed lumen. In mucosal involvement, histology<br />

is diagnostic in more than 80% of patients,<br />

Eosinophilic gastritis 105<br />

revealing a striking eosinophilic infiltrate of the<br />

lamina propria and penetration of eosinophils into<br />

the epithelium. Although eosinophils may be a<br />

prominent inflammatory component in other types<br />

of gastritis, such as H. <strong>pylori</strong> gastritis and chemical<br />

gastropathy, the diagnosis can be made in almost<br />

all cases through multiple biopsies. Rarely,<br />

laparoscopy and full-thickness biopsies are needed<br />

to confirm the suspicions.<br />

Collagenous gastritis<br />

Collagenous gastritis is an extremely rare disorder<br />

of unknown etiology, To date, fewer than ten cases<br />

have been reported in the literature. The condition<br />

is characterized by deposition of a subepithelial<br />

collagen band greater than 10 µm in thickness. The<br />

disorder was originally described by Colletti and<br />

Trainer in a 15-year-old girl with refractory H.<br />

<strong>pylori</strong>-negative chronic gastritis. 55 It has been<br />

reported either as an isolated entity or with<br />

synchronous collagenous colitis, collagenous<br />

duodenitis, lymphocytic colitis or celiac disease.<br />

Clinical features, such as epigastric pain, vomiting,<br />

anorexia, postprandial fullness and weight<br />

loss are reported. Endoscopic findings include<br />

diffuse nodularity, patchy or diffuse erythema,<br />

erosions and frank ulcers with hemorrhage. The<br />

diagnosis relies on mucosal histology, usually of<br />

the fundus and corpus, sharing discontinuous<br />

subepithelial collagen deposition with entrapped<br />

capillaries and fibroblasts, in association with<br />

mild glandular atrophy and mixed inflammatory<br />

infiltrate in the lamina propria, including lymphocytes,<br />

neutrophils, degranulating eosinophils and<br />

mast cells. 56 Depending on the extension of the<br />

inflammatory infiltrate, the corresponding surface<br />

epithelium may show an increased number of<br />

intraepithelial lymphocytes and degenerative<br />

changes.<br />

Three major pathogenic theories have been<br />

proposed for the increased subepithelial collagen<br />

deposition: inflammatory origin, abnormality of<br />

the pericryptal collagen sheath and autoimmune<br />

injury. It has been suggested that an initial stimulus<br />

caused by an infective agent, drug or food allergen<br />

may damage the mucosal surface and, in<br />

susceptible individuals, determine subepithelial<br />

collagen deposition irrespective of the course of


106<br />

Other gastritides<br />

gastritis in the rest of the mucosa. However, this<br />

theory remains to be demonstrated.<br />

Proton pump inhibitor gastritis<br />

Omeprazole, a proton pump inhibitor (PPI) is now<br />

widely used for severe erosive esophagitis in both<br />

adults and children. Long-term omeprazole treatment<br />

is associated with increased levels of serum<br />

gastrin (hypergastrinemia) and gastric mucosal<br />

change, including parietal cell hypertrophy, an<br />

increased number of antral G cells and an<br />

increased number of argyrophil cells.<br />

Pashankar and Israel found gastric nodules or<br />

polyps in seven of 31 children receiving omeprazole<br />

for more than 6 months. 57 At histology,<br />

nodules, ranging from 2 to 4 mm, showed only<br />

mucosal edema and disappeared spontaneously<br />

during treatment, whereas gastric polyps, of<br />

hyperplastic or fundic gland types, persisted<br />

during ongoing therapy. Two different pathogenetic<br />

mechanisms for polyp formation have<br />

been suggested: it has been proposed that glandular<br />

obstructions and dilatation may be caused by<br />

increased viscosity of gland secretion, because of<br />

decreased gastric acid and fluid output; it has also<br />

been speculated that hypertrophy of parietal cells,<br />

induced by omeprazole therapy, may increase<br />

outflow resistance and induce dilatation of the<br />

gland.<br />

This gastropathy seems to be benign, but a careful<br />

endoscopic follow-up is suggested in children<br />

receiving long-term omeprazole therapy.<br />

Pernicious anemia<br />

Pernicious anemia is an autoimmune disorder<br />

caused by production of autoantibodies against the<br />

parietal cell antigen, H + /K + -ATPase, and against<br />

the parietal cell secretory product, intrinsic factor,<br />

resulting in the loss of parietal cells in the fundus<br />

and body of the stomach. The loss of these cells is<br />

associated with achlorhydria, vitamin B12 deficiency<br />

and megaloblastic anemia. 58<br />

Macroscopically, it is characterized by loss of<br />

mucosal folds and thinning of the gastric mucosa.<br />

Histology shows a marked infiltration into the<br />

submucosa by mononuclear cells, including<br />

autoantibody-containing plasma cells, T and B<br />

cells, which can extend into the lamina propria<br />

between the glands. Cellular infiltration of the<br />

mucosa is accompanied by degenerative change in<br />

parietal cells. In the fully established lesion, as<br />

mentioned, there is a marked reduction in number<br />

of gastric glands and the parietal cells are replaced<br />

by mucus-containing cells. Recently, studies in<br />

murine models of autoimmune gastritis have<br />

suggested that macrophages and CD4 + T cells<br />

might play a role in the pathogenesis of the disease<br />

and that the interaction between the Fas antigen on<br />

the target cells and its ligand on the effector cells<br />

might be responsible for target cell destruction. 59<br />

In children the ‘adult form’ of pernicious anemia<br />

has been reported in association with other<br />

autoimmune diseases, such as thyroiditis and<br />

diabetes mellitus, and with precancerous and<br />

malignant lesions. In childhood, pernicious<br />

anemia has been attributed to dietary lack of cobalamin,<br />

to absence of cobalamin in the ileum or to<br />

so-called juvenile pernicious anemia. The last is<br />

characterized by absent or very low levels of<br />

intrinsic factor in the gastric juice, in the absence<br />

of achlorhydria and the absence of intrinsic factor<br />

or H + /K + -ATPase autoantibodies. 58 It has been<br />

suggested that this defect could be due to abnormal<br />

synthesis and biological elaboration in the<br />

gastric lumen of intrinsic factor.<br />

Gastritis associated with other<br />

autoimmune disease<br />

Gastritis with or without atrophy has been<br />

described in association with several autoimmune<br />

diseases, such as connective tissue disorders,<br />

autoimmune thyroiditis and vitiligo. Histological<br />

gastritis has been reported in 25 of 27 diabetic<br />

children undergoing gastroscopy for upper<br />

gastrointestinal symptoms; only half of them had<br />

evidence of macroscopic involvement of the<br />

gastric mucosa, including erosion and ulcers. 60 A<br />

teenage girl with scleroderma and atrophic gastritis<br />

has been reported. 3<br />

Graft-versus-host disease<br />

Acute graft-versus-host disease (GVHD) begins 3 or<br />

4 weeks after transplantation with mucositis,


dermatitis, enteritis and hepatic dysfunction.<br />

Upper non-specific gastrointestinal symptoms,<br />

such as nausea, vomiting, bloating and food intolerance,<br />

occur in a large proportion of patients,<br />

even in the absence of lower gastrointestinal<br />

symptoms. 61 These symptoms seem to be more<br />

frequent than those arising from lower involvement<br />

and are thought to represent an early manifestation<br />

of GVHD. Endoscopic findings vary<br />

considerably, ranging from normal-appearing<br />

mucosa to exensive mucosal sloughing.<br />

Histological findings are diagnostic, including<br />

gastric epithelial cell apoptosis and a marked<br />

lymphocytic infiltrate in the lamina propria.<br />

Upper gastrointestinal GVHD appears to be highly<br />

responsive to immunosuppressive therapy.<br />

Clinical assessment<br />

Symptoms of gastritis and gastropathy are usually<br />

not specific. The most common presenting clinical<br />

features are upper gastrointestinal bleeding and<br />

abdominal pain, which can also characterize other<br />

clinical conditions (Table 7.4). In very young children,<br />

abdominal pain is usually poorly localized<br />

and in some cases may manifest as feeding refusal<br />

or irritability, whereas in older children and<br />

adolescents, the site of pain is more often defined<br />

and a mealtime relationship may be present.<br />

Rarely, acute abdominal pain, resulting from ulcer<br />

perforation, may dominate the clinical presentation.<br />

Gastric blood losses can be revealed by<br />

chronic signs such as iron deficiency anemia or,<br />

alternatively, by acute hemorrhage, with hematemesis<br />

and/or melena. Severe bleeding and perforation<br />

may occur, resulting in a high mortality rate.<br />

Diagnosis<br />

Both single- and double-contrast barium studies<br />

show poor sensitivity in documenting gastritis and<br />

gastropathy, although in some conditions they may<br />

be helpful. Double-contrast studies of the upper<br />

gastrointestinal tract can reveal an irregular<br />

mucosal profile, nodularity, ulceration, lumen<br />

narrowing or mass effect in almost 50% of children<br />

with Crohn’s disease. 62 A double-contrast upper<br />

gastrointestinal tract series in varioliform gastritis<br />

revealed multiple, well-defined, circular filling<br />

Clinical assessment 107<br />

defects, 3–10 mm in diameter, with central punctate<br />

flecks of barium reflecting central superficial<br />

ulceration, usually located on antral and/or gastric<br />

body hypertrophied rugae. 40 In patients with<br />

Ménétrier’s disease, upper gastrointestinal series<br />

identified the characteristic gastric ruga hypertrophy,<br />

localized predominantly in the fundus and<br />

proximal corpus. However, in the evaluation of<br />

children with suspected organic abdominal pain, a<br />

radiology study of the upper gastrointestinal tract<br />

is performed to exclude anatomic abnormalities<br />

such as malrotation, duodenal bands or antral<br />

web.<br />

Upper gastrointestinal endoscopy represents the<br />

first-line procedure for investigating gastric<br />

mucosal pathology. Endoscopic features have been<br />

previously discussed in each section. Although<br />

upper gastrointestinal endoscopy is the most reliable<br />

method for identifying the bleeding source,<br />

defining the lesions and localizing the anatomic<br />

site of mucosal damage, the ability to make a definitive<br />

diagnosis based on endoscopic appearance<br />

alone is limited, because of a poor correlation<br />

between endoscopic abnormalities and histological<br />

findings. The diagnosis and definition of gastritis<br />

and gastropathy are primarily established by<br />

histology. A sampling strategy dealing with<br />

number and size of specimens taken during<br />

endoscopy is recommended. Biopsy specimens<br />

should be taken from both endoscopically visible<br />

lesions and normal-appearing sites, adjacent to the<br />

lesions. Ideally, at least two specimens from each<br />

site should be taken. Visualization and knowledge<br />

of endoscopic and histological findings of other<br />

sites of the gastrointestinal tract (esophagus,<br />

duodenum and colon) can be helpful for a correct<br />

assessment of the gastric mucosa.<br />

Treatment<br />

Stress-associated gastric ulcer<br />

Controlled data on management and outcome of<br />

acute upper gastrointestinal bleeding in children<br />

are lacking. Generally, therapy should start by<br />

replacing the depleted blood volume with human<br />

albumin, saline or Ringer’s lactate. If patients have<br />

massive bleeding, blood transfusion should be<br />

started within a few hours. The use of a nasogastric<br />

tube to decompress the stomach, remove


108<br />

Other gastritides<br />

Table 7.4 Differential diagnosis of gastritis and gastropathy<br />

Upper abdominal pain<br />

Esophagitis<br />

Non-ulcer dyspepsia<br />

Cholelithiasis<br />

Cholecystitis<br />

Pyelonephritis<br />

Urolithiasis<br />

Ureteropelvic junction stricture<br />

Spinal cord lesion<br />

Colitis involving transverse colon<br />

Pancreatitis<br />

Gastrointestinal bleeding<br />

Non-gastrointestinal sources<br />

nasopharynx<br />

chest<br />

maternal blood<br />

Munchausen’s syndrome by proxy<br />

Gastrointestinal sources<br />

esophagitis<br />

Mallory–Weiss tear<br />

erosions/ulcerations<br />

hematobilia<br />

duodenitis<br />

structural lesions<br />

ectopic pancreas<br />

duplication<br />

polyp<br />

duplication cyst<br />

pancreatitis<br />

Vascular causes<br />

hemangioma (Klippel–Trenaunay–Weber syndrome, blue rubber bleb nevus<br />

syndrome)<br />

arteriovenous malformation<br />

connective tissue disorders<br />

bleeding diathesis<br />

gastric secretions and stop the bleeding with saline<br />

lavage is routinely recommended, although the<br />

efficacy of the gastric lavage has not been<br />

confirmed. PPIs have been shown to be effective in<br />

the treatment of active bleeding from gastric and<br />

duodenal ulcers in adults, and they are widely<br />

used in children as well, although scarce data are<br />

available. While in adults therapeutic endoscopy<br />

is effective in reducing further bleeding and lowering<br />

mortality, its efficacy in children has not been


formally addressed. However, it is conceivable that<br />

endoscopic treatment by an experienced endoscopist<br />

might be effective in the treatment of<br />

highly selected children with active gastrointestinal<br />

bleeding. Surgery should be performed when<br />

medical treatment has not stopped the bleeding.<br />

(See also Chapter 36).<br />

The use of prophylactic treatment remains controversial.<br />

Although in adults the results of a recently<br />

published meta-analysis indicated a decreased<br />

frequency of stress-associated gastrointestinal<br />

bleeding with prophylaxis using the H2-receptor<br />

antagonist, the prophylactic use of cimetidine in a<br />

pediatric intensive care unit has not been shown to<br />

be effective in preventing gastrointestinal bleeding<br />

episodes. 63,64 However, prophylactic treatment<br />

with PPIs or sucralfate can be suggested in patients<br />

with severe life-threatening illness.<br />

Other gastritis or gastropathy<br />

Two major issues confront clinicians using<br />

NSAIDs: prevention of induced ulcers, especially<br />

in high-risk groups, and their treatment. In adults,<br />

patients at high risk for gastrointestinal hemorrhage<br />

and perforation from aspirin and NSAIDs<br />

should be considered for prophylactic treatment.<br />

65,66 Risk factors include a history of gastrointestinal<br />

complications, age > 60 years, high dosage,<br />

concurrent use of corticosteroids and concurrent<br />

use of an anticoagulant. Several randomized<br />

controlled trials have shown that concomitant use<br />

of the PGE1 analog misoprostol is effective in<br />

preventing NSAID-related gastropathy. In a retrospective<br />

study, the use of misoprostol during<br />

NSAID therapy resolved symptoms in 82% of<br />

arthritic children with gastrointestinal complaints,<br />

while 18% had recurrence of symptoms after initial<br />

improvement. 67 However, no controlled trials are<br />

available in childhood and the concerns raised<br />

about its use do not support widespread prophylactic<br />

treatment at this time. PPIs are certainly a<br />

good alternative for prevention of NSAID-related<br />

complications. It has been shown in three large<br />

randomized controlled trials in adults that omeprazole<br />

significantly reduced the total number of<br />

NSAID-related ulcers when compared with placebo<br />

and ranitidine, and was also more effective than<br />

misoprostol in preventing duodenal ulcers and<br />

equally so in reducing gastric ulcers. 66<br />

Treatment 109<br />

NSAID-related ulcers may be treated effectively<br />

with any approved therapy for peptic ulcer<br />

disease. In patients who develop severe dyspeptic<br />

symptoms during NSAID use it is preferable to<br />

discontinue the therapy. A PPI is the drug of<br />

choice for a large ulcer or if NSAIDs must be<br />

continued. Despite the fact that several epidemiological<br />

studies have shown little interaction<br />

between H. <strong>pylori</strong> and NSAIDs in the development<br />

of ulcer disease, it has been shown that eradication<br />

of H. <strong>pylori</strong> before NSAID therapy can reduce the<br />

incidence of peptic disease, suggesting that the<br />

organism should be eradicated in any infected<br />

patients before they start NSAIDs. 66<br />

A new class of agents, the so-called selective COX-<br />

2 inhibitors, are thought to be associated with<br />

greater short-term and long-term gastrointestinal<br />

safety compared to regular NSAIDs (non-selective<br />

COX inhibitors). 17 These newly introduced drugs,<br />

by sparing the COX-1 enzyme and its physiological<br />

function, reduce PG-dependent inflammation<br />

while leaving protective gastric mucosal PG<br />

synthesis intact. Although in adulthood initial<br />

studies have shown that they are associated with<br />

fewer ulcer complications compared to nonselective<br />

NSAIDs and they have already been<br />

approved for arthritis treatment, prospective<br />

clinical data are still needed to establish the shortand<br />

long-term safety profiles in childhood.<br />

In patients with Ménétrier’s disease, treatment is<br />

generally supportive, with the majority of patients<br />

(about 90%) responding to a high-protein diet,<br />

diuretic drugs, intravenous albumin transfusion,<br />

and salt and fluid restriction. 25–27 Some patients<br />

can require an 8-week course of antisecretory<br />

agents (H2-receptor antagonist or PPIs). In rare<br />

cases, parenteral nutrition, due to enteral feeding<br />

intolerance, or gastric resection for controlling<br />

protracted hemorrhage, is necessary.<br />

In ZES, although the occurrence of peptic acid<br />

disease can be significantly reduced with an<br />

adequate control of gastric acid secretion, through<br />

appropriate doses of PPIs, surgical resection of a<br />

neoplastic lesion is recommended for any patient.<br />

In patients with PZES, antrectomy was usually<br />

suggested before the advent of antisecretory drugs.<br />

Currently, however, the treatment of choice has<br />

become the long-term maintenance of acid<br />

suppression by PPIs. 35


110<br />

Other gastritides<br />

Crohn’s disease gastritis usually responds to<br />

systemic anti-inflammatory therapies, even<br />

though PPIs are effective for symptomatic relief.<br />

Gastric-outlet obstruction in chronic granulomatous<br />

disease has been successfully managed with<br />

prolonged antimicrobial therapy, corticosteroids<br />

and immunosuppressive drugs, such as<br />

cyclosporin. 68 In eosinophilic gastritis, although<br />

an elemental amino acid-based diet occasionally<br />

appears helpful, corticosteroids remain the main-<br />

REFERENCES<br />

1. Yardley JH, Hendrix TR. Gastritis, gastropathy, duodenitis<br />

and associated ulcerative lesions. In Yamada T, eds.<br />

Textbook of Gastroenterology, 3rd edn. Philadelphia:<br />

Lippincot Williams & Wilkins, 1999: 1463–1499.<br />

2. Drumm B, Rhoad JM, Stringer DA et al. Peptic ulcer<br />

disease in children: etiology, clinical findings, and clinical<br />

course. Pediatrics 1988; 82: 410–414.<br />

3. Dohil R, Hassall E, Jevon G et al. Gastritis and gastropathy<br />

of childhood. J Pediatr Gastroenterol Nutr 1999; 29:<br />

378–394.<br />

4. Oliva MM, Lazenby AJ, Perman JA. Gastritis associated<br />

with Gastrosporillum hominis in children: comparison<br />

with <strong>Helicobacter</strong> <strong>pylori</strong> and review of the literature.<br />

Mod Pathol 1993; 6: 513–515.<br />

5. Norris CR, Marks SL, Eaton KA et al. Healthy cats are<br />

commonly colonized with ‘<strong>Helicobacter</strong> heilmannii’ that<br />

is associated with minimal gastritis. J Clin Microbiol<br />

1999; 37: 189–194.<br />

6. van Loon S, Bart A, den Hertog EJ et al. <strong>Helicobacter</strong><br />

heilmannii gastritis caused by cat to child transmission.<br />

J Pediatr Gastroenterol Nutr 2003; 36: 407–409.<br />

7. Subei I, Attar B, Schmitt G et al. Primary gastric tuberculosis:<br />

a case report and literature review. Am J<br />

Gastroenterol 1987; 82: 769–72.<br />

8. Bodmer T, Miltner E, Bermudez LE. Mycobacterium<br />

avium resists exposure to the acidic conditions the<br />

stomach. FEMS Microbiol Lett 2000; 182: 45–49.<br />

9. Howiler W, Goldberg HI. Gastroesophageal involvement<br />

in herpes simplex. Gastroenterology 1976; 70: 775–778.<br />

10. McGluggage WG, Fox JD, Baillie KEM et al. Varicella<br />

zoster gastritis in a bone marrow transplant recipient. J<br />

Clin Pathol 1994; 47: 1054–1056.<br />

11. Armstrong KL, Fraser DK, Faoagali JL. Gastrointestinal<br />

bleeding with influenza virus. Med J Aust 1991; 154:<br />

180–182.<br />

12. Alonso A, Daschner A, Moreno-Ancillo A. Anaphylaxis<br />

with Anisakis simplex in the gastric mucosa. N Engl J<br />

Med 1997; 337: 350–354.<br />

13. Doglioni C, De Boni M, Cielo R et al. Gastric giardiasis. J<br />

Clin Pathol 1992; 45: 964–967.<br />

14. Garone MA, Winston BJ, Lewis JH. Cryptosporidiosis of<br />

the stomach. Am J Gastroenterol 1986; 81: 465–470.<br />

15. Gabriel SE, Jaakkimainen L, Bombardier C. Risk for<br />

serious gastrointestinal complications related to use of<br />

nonsteroidal anti-inflammatory drugs. A meta-analysis.<br />

Ann Intern Med 1991; 115: 787–796.<br />

stay of therapy in patients. The use of sodium<br />

cromoglycate is very controversial, whereas<br />

montelukast, a selective leukotriene receptor<br />

antagonist, has been successfully used in a 13year-old<br />

girl with acute gastrointestinal disease. 69<br />

In celiac disease the lymphocytic infiltrate usually<br />

resolves after strict adherence to a gluten-free<br />

diet. 43 The standard treatment for pernicious<br />

anemia is regular monthly intramuscular injection<br />

of vitamin B12 to correct the vitamin deficiency. 58<br />

16. Wallace JL. Nonsteroidal anti-inflammatory drugs and<br />

gastroenteropathy: the second hundred years.<br />

Gastroenterology 1997; 112: 1000–1016.<br />

17. Vane JR. Towards a better aspirin. Nature 1994; 367:<br />

215–216.<br />

18. Ng PC, Brownlee KG, Dear PRF. Gastroduodenal perforation<br />

in preterm babies treated with dexamethasone for<br />

bronchopulmonary dysplasia. Arch Dis Child 1991; 66:<br />

1164–1166.<br />

19. Messer J, Reitman D, Sacks HS et al. Association of<br />

adrenocorticosteroid therapy and peptic-ulcer disease.<br />

N Engl J Med 1983; 309: 21–24.<br />

20. Peled N, Dagan O, Babyn P et al. Gastric-outlet obstruction<br />

induced by prostaglandin therapy in neonates. N<br />

Eng J Med 1992; 327: 505–510.<br />

21. Laine L, Weinstein WM. Histology of alcoholic hemorrhagic<br />

‘gastritis’: a prospective evaluation.<br />

Gastroenterology 1988; 94: 1254–1262.<br />

22. Trier JS, Szabo S, Allan CH. Ethanol-induced damage to<br />

mucosal capillaries of rat stomach. Ultrastructural<br />

features and effects of prostaglandin F2b and<br />

cysteamine. Gastroenterology 1987; 92: 13–22.<br />

23. Grjboski JD. Peptic ulcer disease in children. Med Clin<br />

North Am 1991; 75: 899–902.<br />

24. Chaibou M, Tucci M, Dugas MA et al. Clinically significant<br />

upper gastrointestinal bleeding acquired in a pediatric<br />

intensive care unit: a prospective study. Pediatrics<br />

1998; 102: 933–938.<br />

25. Occena RO, Taylor SF, Robinson CC et al. Association of<br />

cytomegalovirus with Ménétrier’s disease in childhood:<br />

report of two new cases with a review of literature. J<br />

Pediatr Gastroenterol Nutr 1993; 17: 217–224.<br />

26. Eisenstat DDR, Griffiths AM, Cutz E et al. Acute<br />

cytomegalovirus infection in a child with Ménétrier’s<br />

disease. Gastroenterology 1995; 109: 592–595.<br />

27. Kelly DG, Miller LJ, Malagelada J-R et al. Giant hypertrophic<br />

gastropathy (Ménétrier’s disease): pharmacologic<br />

effects on protein leakage and mucosal ultrastructure.<br />

Gastroenterology 1982; 83: 581–589.<br />

28. Bayerdörffer E, Ritter MM, Hatz R et al. Healing of<br />

protein losing hypertrophic gastropathy by eradication<br />

of <strong>Helicobacter</strong> <strong>pylori</strong> – Is <strong>Helicobacter</strong> <strong>pylori</strong> a pathogenic<br />

factor in Ménétrier’s disease. Gut 1994; 35:<br />

701–704.<br />

29. Sferra TJ, Pawel BR, Qualman SJ et al. Ménétrier’s<br />

disease of childhood: role of cytomegalovirus and


trasforming growth factor alpha. J Pediatr 1996; 128:<br />

213–219.<br />

30. Bechi P, Amorosi A, Mazzanti R et al. Reflux-related<br />

gastric mucosal injury is associated with increased<br />

mucosal histamine content in human. Gastroenterology<br />

1993; 104: 1057–1063.<br />

31. Dixon MF, O’Connor HJ, Axon ATR et al. Reflux gastritis:<br />

distinct histopathological entity? J Clin Pathol 1990;<br />

39: 524–530.<br />

32. Dixon MF, Mapstone NP, Neville PM et al. Bile reflux<br />

gastritis and intestinal metaplasia at the cardia. Gut<br />

2002; 51: 351–355.<br />

33. Hyams JS, Treem WR. <strong>Portal</strong> hypertensive gastropathy<br />

in children. J Pediatr Gastroenterol Nutr 1993; 17:<br />

13–18.<br />

34. Yachha SK, Ghoshal UC, Gupta R et al. <strong>Portal</strong> hypertensive<br />

gastropathy in children with extrahepatic portal<br />

venous obstruction: role of variceal obliteration by<br />

endoscopic sclerotherapy and <strong>Helicobacter</strong> <strong>pylori</strong> infection.<br />

J Pediatr Gastroenterol Nutr 1996; 23: 20–23.<br />

35. Pisegna JR. The effect of Zollinger-Ellison syndrome<br />

and neuropeptide-secreting tumors on the stomach.<br />

Curr Gastroenterol Rep 1999; 1: 511–517.<br />

36. Hirschowitz BI. Zollinger–Ellison syndrome: pathogenesis,<br />

diagnosis and management. Am J Gastroenterol<br />

1997; 92 (S): 44S–48S.<br />

37. Modlin IM, Tang LH. Approaches to the diagnosis of gut<br />

neuroendocrine tumors: the last word (today).<br />

Gastroenterology 1997; 112: 583–590.<br />

38. Annibale B, Bonamico M, Rindi G et al. Antral gastric<br />

cell hyperfunction in children: a functional and<br />

immunocytochemical report. Gastroenterology 1991;<br />

101: 1547–1551.<br />

39. Haot J, Hamichi L, Wallez L et al. Lymphocytic gastritis:<br />

a newly described entity: a retrospective/endoscopic<br />

and histological study. Gut 1988; 29: 1258–1264.<br />

40. Couper R, Laski B, Drumm B et al. Chronic varioliform<br />

gastritis in childhood. J Pediatr 1989; 115: 441–444.<br />

41. Wolberg R, Owen D, DelBuono L et al. Lymphocytic<br />

gastritis in patients with celiac sprue or spruelike<br />

intestinal disease. Gastroenterology 1990; 98: 310–315.<br />

42. Hayat M, Arora DS, Wyatt JI et al. The pattern of<br />

involvement of the gastric mucosa in lymphocytic<br />

gastritis is predictive of the presence of duodenal<br />

pathology. J Clin Pathol 1999; 52: 815–819.<br />

43. De Giacomo C, Gianatti A, Negrini R et al. Lymphocytic<br />

gastritis: a positive relationship with celiac disease. J<br />

Pediatr 1994; 124: 57–62.<br />

44. Verkarre V, Asnafi V, Lecomte T et al. Refractory coeliac<br />

sprue is a diffuse gastrointestinal disease. Gut 2002; 52:<br />

205–211.<br />

45. Schmidt-Sommerfeld, Kirschner B, Stephens J.<br />

Endoscopic and histologic findings in the upper<br />

gastrointestinal tract of children with Crohn’s disease. J<br />

Pediatr Gastroenterol Nutr 1990; 11: 448–454.<br />

46. Tobin JM, Sinha B, Ramani P et al. Upper gastrointestinal<br />

mucosal disease in pediatric Crohn disease and<br />

ulcerative colitis: a blinded, controlled study. J Pediatr<br />

Gastroenterol Nutr 2001; 32: 443–448.<br />

47. Oberhuber G, Puspok A, Oesterreicher C et al. Focally<br />

enhanced gastritis: a frequent type of gastritis in<br />

patients with Crohn’s disease. Gastroenterology 1997;<br />

112: 698–706.<br />

48. Meining A, Bayerdorffer E, Bastlein E et al. Focally<br />

inflammatory infiltrations in the gastric biopsy specimens<br />

are suggestive of Crohn’s disease. Scand J<br />

Gastroenterol 1997; 32: 813–818.<br />

References 111<br />

49. Parente F, Cucino C, Bollani S et al. Focal gastric inflammatory<br />

infiltrates in inflammatory bowel disease: prevalence,<br />

immunohistochemical characteristics, and diagnostic<br />

role. Am J Gastroenterol 2000; 95: 705–711.<br />

50. Sharif F, McDermott M, Path MRC et al. Focally<br />

enhanced gastritis in children with Crohn’s disease and<br />

ulcerative colitis. Am J Gastroenterol 2002; 97:<br />

1415–1420.<br />

51. Kundhal PS, Stormon MO, Zachos M et al. Gastral<br />

antral biopsy in the differentiation of pediatric colitides.<br />

Am J Gastroenterol 2003; 98: 557–561.<br />

52. Dikerman JD, Colletti RB, Tampas JP et al. Gastric outlet<br />

obstruction in chronic granulomatous disease of childhood.<br />

Am J Dis Child 1986; 140: 567–570.<br />

53. Daneshjoo R, Talley NJ. Eosinophilic gastroenteritis.<br />

Curr Gastroenterol Rep 2002; 4: 366–372.<br />

54. Kelly KJ. Eosinophilic gastroenteritis. J Pediatr<br />

Gastroenterol Nutr 2000; 30: S28–S35.<br />

55. Colletti RB, Trainer TD. Collagenous gastritis.<br />

Gastroenterology 1989; 97: 1552–1555.<br />

56. Pulimood AB, Ramakrishna BS, Mathan MM.<br />

Collagenous gastritis and collagenous colitis: a report<br />

with sequential histological and ultrastructural findings.<br />

Gut 1999; 44: 881–885.<br />

57. Pashankar DS, Israel DM. Gastric polyps and nodules in<br />

children receiving long-term omeprazole therapy. J<br />

Pediatr Gastroenterol Nutr 2002; 35: 658–662.<br />

58. Toh BH, van Driel IR, Gleeson PA. Pernicious anemia. N<br />

Engl J Med 1997; 337: 1441–1448.<br />

59. Nishio A, Katakai T, Oshima C et al. A possible involvement<br />

of Fas–Fas ligand signaling in the pathogenesis of<br />

murine autoimmune gastritis. Gastroenterology 1996;<br />

111: 959–967.<br />

60. Burghen GA, Murrell LR, Whitington GL et al. Acid<br />

peptic disease in children with type I diabetes mellitus:<br />

a complicating relationship. 1992; 146: 718–722.<br />

61. Weisdorf DJ, Snover DC, Haake R. Acute upper gastrointestinal<br />

graft-versus-host disease: clinical significance<br />

and response to immunosoppressive therapy. Blood<br />

1990; 76: 624–629.<br />

62. Ruuska T, Vaajalahati P, Arajarvi P et al. Prospective<br />

evaluation of upper gastrointestinal mucosal lesions in<br />

children with ulcerative colitis and Crohn’s disease. J<br />

Pediatr Gastroenterol Nutr 1994; 19: 181–186.<br />

63. Cook DJ, Reeve BK, Guyatt GH et al. Stress ulcer<br />

prophylaxis in critically ill patients: resolving discordant<br />

meta-analyses. JAMA 1996; 275: 308–314.<br />

64. Lacroix J, Nadeau D, Laberge S et al. Frequency of upper<br />

gastrointestinal bleeding in a pediatric intensive care<br />

unit. Crit Care Med 1992; 20: 35–42.<br />

65. Goldstein JL, Brown RD. NSAID-induced ulcers. Curr<br />

Treat Options Gastroenterol 2000; 3: 149–157.<br />

66. Lanza FL. A guideline for the treatment and prevention<br />

of NSAID-induced ulcers. Am J Gastroenterol 1998; 93:<br />

2037–2046.<br />

67. Gazarian M, Berkovitch M, Koren G et al. Experience<br />

with misoprostol therapy for NSAID gastropathy in children.<br />

Ann Rheum Dis 1995; 45: 277–280.<br />

68. Rash JR, Tang HB, Mayer L et al. Treatment of<br />

intractable gastrointestinal manifestations of chronic<br />

granulomatous disease with cyclosporine. J Pediatr<br />

1995; 126: 143–145.<br />

69. Neustrom MR, Friesen C. Treatment of eosinophilic<br />

gastroenteritis with montelukast. J Allergy Clin Immunol<br />

1999; 104: 506.


8<br />

Introduction<br />

HIV and the intestine<br />

Nigel C Rollins<br />

It was estimated that at the end of 2002 there were<br />

42 million people worldwide with HIV-1 infection,<br />

over 29 million of whom were living in sub-<br />

Saharan Africa. Despite some encouraging signs<br />

that the rate of new infections is stabilizing, there<br />

are still about 3 million children under 15 years of<br />

age living with HIV. As a case in point, approximately<br />

68000 children with HIV infection were<br />

born in South Africa in 2002, and AIDS-related<br />

complications now account for 60–80% of pediatric<br />

admissions there and 70% of hospital deaths.<br />

Gastrointestinal disease, mainly diarrhea, accounts<br />

for about a third of these admissions.<br />

Pathophysiology: transmission and<br />

progression of HIV disease<br />

The gut, more than any other organ in the body,<br />

plays a critical role in the vertical transmission<br />

and progression of HIV infection. This reflects not<br />

only its nutritional function but also its central<br />

activity in immune programming and regulation.<br />

More than half the body’s total lymphoid tissue is<br />

found in the bowel; as HIV infects lymphocytes<br />

with CD4 or other co-receptors, this makes the<br />

bowel an ideal target for primary infection and a<br />

site for viral replication and seeding.<br />

Intestinal lymphoid tissue is organized into<br />

distinct structures including the tonsils, Peyer’s<br />

patches (mainly in the ileum) and mesenteric<br />

lymph nodes, and is also diffusely spread<br />

throughout the mucosal epithelium and lamina<br />

propria. Activation of both the organized and<br />

diffuse elements of the gut immune system<br />

depends on presentation of antigens to T and B<br />

lymphocytes lying beneath the epithelium. M<br />

cells are specialized epithelium found in the follicle-associated<br />

epithelium (FAE) that overlies<br />

Peyer’s patches in the small intestine and<br />

lymphoid follicles in the rectum, and are also<br />

found in rich supply in the tonsils. These are<br />

capable of transporting macromolecules and<br />

micro-organisms, including viruses such as polio,<br />

to macrophages or possibly dendritic cells in the<br />

lamina propria. These latter cells are the professional<br />

antigen-presenting cells that facilitate<br />

presentation of antigens to activated and possibly<br />

non-activated T cells. However, intestinal epithelium<br />

itself can also generate antigen-specific<br />

responses and may be able to function as antigenpresenting<br />

cells. Complementing these responses<br />

are intraepithelial lymphocytes, interspersed<br />

throughout the mucosa, that produce cytokines<br />

which support humoral and cell-mediated<br />

responses. Finally, the lamina propria throughout<br />

the gut contains the bulk of the immune effector<br />

cells including T and B lymphocytes, plasma<br />

cells, macrophages, mast cells and some<br />

eosinophils and neutrophils.<br />

Primary infection with ingested HIV depends on<br />

the virus gaining access to the lamina propria,<br />

where, in early infection, it replicates primarily<br />

in lymphocytes. 1,2 The exact mechanism for entry<br />

to the lamina propria is not fully understood, but<br />

may be through a variety of routes, and is likely<br />

to be a complex process modified by viral characteristics,<br />

gut mucosal receptors and the factors<br />

that promote or impede the interaction.<br />

Direct passage through disruptions in mucosa or<br />

between immature mucosal junctions is possible.<br />

However, longitudinal gut permeability studies in<br />

children born to HIV-infected mothers do not<br />

support this mechanism. 3 Rather, increased<br />

permeability, without clinical symptoms, may be<br />

present soon after children become infected.<br />

113


114<br />

HIV and the intestine<br />

Recent work shows that gut mucosa does not<br />

express the familiar CD4 binding sites that the<br />

HIV-1 gp120 protein commonly uses for attaching<br />

to and thereby infecting mononuclear cells. 4<br />

Rather, early infection is dependent on HIV strains<br />

binding to alternative sites, namely the primary<br />

receptor galactosyl ceramide (GalCer) and the<br />

CCR5 co-receptor, which are expressed on upper<br />

gastrointestinal epithelium. 5 This is supported by<br />

the observation that HIV-1 isolates from acutely<br />

infected persons are predominantly R5 viruses, i.e.<br />

macrophage-tropic HIV that require CCR5 for cell<br />

entry (in contrast to R4 viruses, which are lymphocyte-tropic<br />

requiring the CXCR4 chemokine coreceptor).<br />

6 Once attached, the virus is probably<br />

translocated across the epithelial cell and<br />

presented to T lymphocytes that express the same<br />

CCR5 co-receptor. The lamina propria now<br />

becomes a potent site for viral replication. M cells,<br />

in mouse and rabbit ex vivo systems, can transport<br />

HIV-1 to mononuclear cells, but human M cells<br />

have not been shown to take up and transport HIV<br />

in the same way. 7<br />

The gut has a number of defense mechanisms<br />

against primary infection apart from maternal<br />

factors. The rapid passage of ingested amniotic<br />

fluid or maternal blood limits contact time with M<br />

cells expressed on the surface of the tonsils.<br />

Gastric acid would normally inhibit HIV in the<br />

stomach, but this is reduced in neonates and<br />

young infants. Mucin produced by goblet cells and<br />

the rapid turnover of the intestinal mucosa provide<br />

additional mechanical barriers to HIV attachment.<br />

However, mother-to-child transmission of HIV<br />

through breast feeding depends on a number of<br />

risk factors other than the susceptibility of the gut<br />

to HIV. Increased viral load in breast milk is correlated<br />

with high plasma viral load and decreased<br />

CD4 count in the mother, and is associated with<br />

higher transmission. 8–10 These conditions are<br />

found in the early stages of new infection or in<br />

advanced disease. It is not clear, however, whether<br />

it is cell-associated or cell-free virus that is more<br />

important for transmission. Other breast-milk<br />

factors such as HIV-1-specific IgG and secretory<br />

leukocyte protease inhibitor (SLPI) have HIV-1<br />

inhibitory activity in vitro. 11 SLPI acts on the target<br />

cell rather than the virus and inhibits internalization<br />

of HIV-1 rather than initial binding. 12<br />

Infant feeding practices may significantly influence<br />

the way in which these non-intestinal factors<br />

interact: exclusive breast feeding results in a<br />

greater volume of milk being ingested and hence<br />

increases the HIV load presented to the infant gut.<br />

However, it also increases the protective breastmilk<br />

factors reaching the infant and that promote<br />

the development and maintain the integrity of the<br />

mucosa and which may have direct anti-HIV<br />

effects. 13 Exclusive breast feeding is associated<br />

with fewer breast health problems, e.g. clinical<br />

and subclinical mastitis that are associated with<br />

increased breast-milk viral load. 14 Infant feeding<br />

practices also contribute to the establishment of<br />

different enteric microflora that might significantly<br />

affect the priming or responses of intestinal<br />

lymphoid cells or dendritic cell. 15–17 This could,<br />

theoretically, modify adherence or facilitate infection<br />

of HIV, although this interesting concept has<br />

not been explored to date. Innate responses such<br />

as lactoferrin and lysozymes that are secreted by<br />

exocrine glands onto mucosal surfaces are bactericidal,<br />

but their protective capacity in HIV is<br />

unknown. There is also renewed interest around<br />

secretory IgA and IgM that can inhibit transcytosis<br />

of HIV-1 across enterocytes in in vitro models. 18,19<br />

Passive immunization of macaque monkeys with<br />

monoclonal antibodies against the HIV-1 gp120<br />

protein, given either orally or applied locally,<br />

protected against mucosal challenges with HIV-<br />

1. 20 This raises the possibility of a vaccine strategy,<br />

similar to that used against hepatitis B, of combining<br />

active and passive immunization to induce<br />

effective mucosal protection against HIV-1.<br />

Once HIV infection is established the gut serves as<br />

a major reservoir for viral amplification. CD4<br />

lymphocytes in the lamina propria are infected<br />

and progressively depleted; 21 this precedes the<br />

eventual decrease in CD4 counts in the peripheral<br />

circulation and other sites. 22,23 IgA-secreting<br />

plasma cells, especially the IgA2 subclass, are lost<br />

early in disease, resulting in reduced secretory<br />

IgA, thereby predisposing the gut to infection with<br />

enteric pathogens. 24–27 Responses to oral vaccines<br />

may also be impaired, raising concern about the<br />

effectiveness of putative rotavirus vaccines in<br />

areas of high HIV seroprevalence. 28 Local cytokine<br />

responses have been extensively reported,<br />

although it is still not entirely clear whether they<br />

influence the development of AIDS or HIV


enteropathy or are simply the consequence of the<br />

latter. 29,30 These responses may be the consequence<br />

of opportunistic infections or the direct<br />

effect of HIV on the mucosa.<br />

Diarrhea<br />

Persistent and recurrent diarrheas are amongst the<br />

most frequent manifestations of HIV/AIDS in both<br />

children and adults, especially in developing<br />

countries, where diarrhea is associated with<br />

growth failure, weight loss and death. In Zaire,<br />

85% of adults admitted with persistent diarrhea<br />

had HIV infection. 31 Among children presenting to<br />

a primary health care facility in South Africa, a<br />

history of persistent diarrhea in the preceding 3<br />

months strongly predicted HIV infection (odds<br />

ratio 4.8; CI 2.5–9.3 and positive predictive value<br />

of 63%). In South Africa, the profile of children<br />

admitted with diarrheal diseases has changed,<br />

with increasing prevalence of persistent diarrhea<br />

and loss of seasonal peaks of acute diarrhea. HIVinfected<br />

children admitted to hospital with diarrhea<br />

have more severe symptoms than children<br />

uninfected by HIV; 32 they frequently have severe<br />

co-infections such as pneumonia, pseudomonal<br />

skin sepsis and tuberculosis (TB). These result in<br />

longer periods of admission and case fatality rates<br />

in the order of 27%. In Zaire, children with HIV<br />

and persistent diarrhea had an 11-fold increased<br />

risk of death compared with uninfected controls.<br />

Hence, in developing countries, HIV-related diarrheal<br />

diseases represent a major burden on both<br />

caregivers and health resources. The availability of<br />

antiretroviral drugs in North America and Europe<br />

has dramatically altered the natural history of HIV<br />

infection and has reduced the incidence of<br />

gastrointestinal disease due to opportunistic infections.<br />

33,34 This has shifted much of the scientific<br />

interest and research funding to other complications<br />

such as lipodystrophy.<br />

Although there is considerable knowledge of the<br />

pathophysiology and management of diarrhea in<br />

adults with AIDS, less is known about this<br />

problem in children. Opportunistic infections and<br />

fat malabsorption are common causes of prolonged<br />

and recurrent diarrhea in infected adults. 35,36<br />

Among children with HIV, carbohydrate intolerance<br />

is common and may follow acute infections<br />

Diarrhea 115<br />

with rotavirus or other common enteropathogens.<br />

37–39 In resource-poor settings, the<br />

optimal management of HIV-related diarrhea<br />

remains largely unexamined. Even after 25 years of<br />

the epidemic, fewer than 40% of countries with a<br />

high HIV seroprevalence have published national<br />

recommendations for the management of HIVassociated<br />

diarrhea. 40<br />

Mucosal structure and function in HIV-related<br />

diarrhea<br />

A number of intestinal mucosal structural and<br />

functional abnormalities have been described in<br />

HIV-infected children with diarrhea. Their pathogenesis<br />

is multifactorial and includes the direct<br />

effect of pathogens, other inflammatory processes,<br />

malnutrition and the direct effect of HIV itself.<br />

Variable degrees of villous atrophy, crypt hyperplasia<br />

and increased inflammatory cells in the<br />

lamina propria are found in the small intestine of<br />

children with diarrhea (Figure 8.1), although these<br />

can be present without symptoms, and are often<br />

diffusely spread throughout the upper duodenum<br />

and jejunum. 41,42 Even when the mucosa appears<br />

normal on conventional histology, short and irregular<br />

microvilli may be evident on electron<br />

microscopy and tubuloreticular inclusions may be<br />

seen in endothelial cells. 43 In the large bowel, loss<br />

Figure 8.1 Villous blunting with crypt hyperplasia and<br />

increased inflammatory cells in the lamina propria are<br />

evident in this small-intestinal biopsy of an HIV-infected<br />

child.


116<br />

HIV and the intestine<br />

of mucin-producing cells is accompanied by an<br />

increase in inflammatory cells, especially mast<br />

cells; crypt abscesses and apoptosis are also<br />

detected. Although up to 75% of adult HIV-related<br />

diarrhea has been attributed to enteric pathogens,<br />

causal agents have not been demonstrated with the<br />

same frequency in children. 44 This may be<br />

explained by the difficulty of obtaining appropriate<br />

specimens from small children or identifying<br />

pathogens such as microsporidia; in addition,<br />

alternative pathways of diarrhea may be involved,<br />

as seen in other causes of severe malnutrition.<br />

Carbohydrate malabsorption is commonly found<br />

in HIV-infected children with persistent diarrhea.<br />

37 This may be the consequence of preceding<br />

enteric infection but in most cases the precipitant<br />

is unknown. Intestinal biopsies in these children<br />

do not always demonstrate villous atrophy. 45,46 An<br />

abnormal breath H2 analysis in the presence of<br />

relatively normal mucosal architecture suggests a<br />

non-specific brush-border disorder rather than a<br />

specific enzyme deficiency. 37,46 Gut permeability,<br />

measured by the dual sugar absorption test, is<br />

increased, especially in patients with more<br />

advanced disease; 41 however, this may also be<br />

increased in asymptomatic patients and early in<br />

disease. 3,47 Furthermore, lactose malabsorption is<br />

not always associated with clinical symptoms<br />

such as diarrhea or growth failure. 48 Monosaccharide<br />

intolerance has been described in<br />

African children with HIV and severe diarrhea,<br />

and has an 87% positive predictive value for identifying<br />

HIV infection in children presenting with<br />

diarrhea. 49 It is associated with prolonged recovery,<br />

probably due to delayed regeneration of enterocytes<br />

and return of absorptive capacity. In<br />

adults, monosaccharide intolerance is associated<br />

with accelerated small bowel transit times, especially<br />

in protozoal infections such as cryptosporidiosis,<br />

50 but this is unlikely to be the sole<br />

explanation. Although monosaccharide intolerance<br />

does occur in HIV-uninfected children with<br />

severe malnutrition, in settings where HIV is<br />

highly prevalent, it can be useful as an indication<br />

for HIV testing.<br />

Other macronutrients and micronutrients may not<br />

be adequately absorbed or there may be increased<br />

losses in stools. Bile salt and fat malabsorption are<br />

associated with low-grade enteropathies in adult<br />

patients with AIDS, 51 but are not consistent find-<br />

ings in HIV-infected children. 39,52 Protein maldigestion<br />

and malabsorption are seen in children with<br />

severe carbohydrate malabsorption and indicate a<br />

profound disruption of normal gastrointestinal<br />

function and absorptive capacity. Enteric<br />

cytomegalovirus or bacterial infections, and occasionally<br />

intestinal Kaposi’s sarcoma, may present in<br />

adults as a protein-losing enteropathy. 53,54 In children,<br />

protein-losing enteropathy, suggested by<br />

increased fecal α1-antitrypsin, has also been<br />

reported, but specific enteric pathogens were not<br />

identified. 39 Iron malabsorption has been reported<br />

in children with symptomatic HIV with decreased<br />

D-xylose absorption, suggesting a general loss of<br />

absorptive capacity rather than a specific iron transport<br />

defect. 55 Vitamin A is absorbed even in the<br />

presence of severe diarrhea; 56 health-care personnel<br />

should aim, therefore, to provide vitamin A supplements,<br />

according to the WHO/UNICEF Integrated<br />

Management of Childhood Illnesses protocol, to all<br />

children presenting with persistent diarrhea at<br />

primary health-care facilities. 57<br />

Host responses and susceptibility<br />

The relationship between the risk and type of<br />

enteric infection and HIV-viral activity, or host<br />

immune competence as measured by CD4 counts,<br />

has been established in HIV-infected adults in<br />

developed countries, and to a limited extent in<br />

developing countries. The rapid and pronounced<br />

loss of CD4 T-helper cells in gut mucosa that<br />

precedes losses in the peripheral blood might<br />

explain the loss of local defenses to opportunistic<br />

infections. In Uganda, HIV-infected adults with<br />

lower CD4 counts were at greater risk of non-typhi<br />

salmonellae septicema and other opportunistic<br />

infections 58 (Figure 8.2).<br />

Children with reduced CD4 counts have more<br />

severe diarrhea, i.e. higher fluid losses and more<br />

prolonged disease; this is often due to cryptosporidiosis.<br />

45,59 Local gut immunity may also play<br />

an important role in both local susceptibility and<br />

disease progression. Total IgA in stool is decreased<br />

in symptomatic children with severe diarrhea;<br />

elevated serum IgA, which may be due to selective<br />

increase of the IgA1 subclass, is related to decreased<br />

CD4 counts. 26,60 These changes occur early in pediatric<br />

HIV infection and reflect the disturbance of the<br />

gut mucosal immune system.


HIV enteropathy<br />

Cases/1000 person-years<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

< 200 200–500 > 500<br />

CD4 + count<br />

HIV enteropathy is typified by weight loss, diarrhea<br />

and characteristic endoscopic and pathological<br />

appearances without any identifiable enteric<br />

pathogens. 61 These include reduced villous height,<br />

increased crypt length, focal enterocyte vacuolization,<br />

and hyporegeneration and dysmaturation of<br />

intestinal cells. 62,63 Several hypotheses have been<br />

proposed to explain these findings, including a<br />

mucosal immune reaction to HIV-infected cells 64<br />

or to signals from regulatory T cells, the effect of<br />

bacterial overgrowth or bile salt malabsorption,<br />

65,66 unidentified enteric pathogens, or direct<br />

small-intestinal damage by HIV itself. The latter<br />

hypothesis is supported by the finding of HIVinfected<br />

mononuclear cells in the gut, epithelial<br />

hypoproliferation, altered intestinal cell differentiation<br />

67 and reduced apoptotic lymphoid cells<br />

following the onset of antiretroviral therapy. 68<br />

Intestinal cell lines directly infected with HIV<br />

clade B strains have reduced brush-border enzyme<br />

activity and altered calcium responses, suggesting<br />

that direct infection of enterocytes with HIV may<br />

modify absorptive and secretory function. 69 Some<br />

of these effects may be due to activation of the<br />

HIV-1 transactivating factor protein gene (Tat)<br />

which is an early immediate regulatory gene that is<br />

essential for virus integration and protein expression.<br />

70 Circulating cytokines and lymphokines<br />

such as interleukin-1β and tumor necrosis factor-α<br />

may have a role in diarrhea due to bacteria, proto-<br />

Enteric infections in HIV-infected children 117<br />

Plasmodium falciparum malariae<br />

Tuberculosis<br />

Cryptococcus<br />

Non-typhi salmonellae<br />

Pneumococci<br />

Gram-negative bacteremia<br />

Figure 8.2 CD4 counts in HIV-infected adults presenting with specific infections in Uganda.<br />

zoa, cytomegalovirus or where there is no identifiable<br />

infection. 30,53 These may produce diarrhea<br />

through their secretory effects in the colon rather<br />

than by altering mucosal barriers. There are<br />

isolated reports of collagenous colitis in adults<br />

with chronic watery diarrhea, but none in children.<br />

71<br />

Enteric infections in HIV-infected<br />

children<br />

Surprisingly little is known about the patterns of<br />

enteric infection in HIV-infected children, timing<br />

of their acquisition, their individual effects on<br />

children’s health and nutrition, and their relationship<br />

to HIV activity and host immune status.<br />

There is reasonable consistency between the few<br />

studies that have been conducted in Africa<br />

describing the pathogens isolated in children<br />

presenting with HIV and diarrhea. 44,72 Enteric<br />

pathogens that commonly cause acute diarrhea in<br />

children uninfected by HIV also frequently result<br />

in diarrhea in HIV-infected children. 73 Adenovirus<br />

40/41 and astrovirus have been identified in children<br />

with persistent diarrhea, but their relative<br />

contribution to the burden of diarrheal disease in<br />

HIV infection is not known. 74–76 Only a few<br />

pathogens seem to be opportunistic in HIVinfected<br />

children with diarrhea. The most prevalent<br />

of these are Candida, Cryptosporidium


118<br />

HIV and the intestine<br />

parvum and cytomegalovirus (CMV); these usually<br />

cause oropharyngeal lesions and invasive lesions<br />

elsewhere in the gut or persistent diarrhea.<br />

Additional pathogens, which occasionally cause<br />

diarrhea or other symptoms in children with<br />

symptomatic HIV disease, may include herpes<br />

simplex virus and Mycobacterium avium intracellulare.<br />

Mycobacterium tuberculosis may be isolated<br />

from stools in both adults and children with no<br />

evidence of pulmonary TB. Perhaps surprisingly,<br />

there is little to suggest any association between<br />

shigellosis and HIV. One study from North<br />

America described an increase in prevalence in<br />

HIV-infected homosexuals; however, numerous<br />

reports from the African continent do not support<br />

this finding. It may be that the virulence of<br />

Shigella readily overcomes even the intact<br />

mucosal defense system and so the immunocompromised<br />

individual is not at any greater risk.<br />

Another possibility is that because Shigella must<br />

be taken up by enteric immune cells in order to be<br />

invasive, an immune system that is not competent<br />

may, paradoxically, be protective. Isospora belli, a<br />

significant enteric pathogen in HIV-infected<br />

adults 77–79 has not been commonly reported<br />

amongst HIV-infected children in Africa. 72,80 Little<br />

is known about the role of the Microsporidia spp<br />

and the newly recognized protozoan enteric<br />

pathogen Cyclospora cayetanensis in AIDS-associated<br />

diarrhea in children. 81–83 Laboratory identification<br />

of several of these infections may be very<br />

difficult, requiring special stains or techniques<br />

that are expensive or unavailable in most developing<br />

countries. In children, the proportion of diarrhea<br />

cases due to infective agents may, therefore,<br />

be underestimated.<br />

Cryptosporidium parvum<br />

C. parvum is a common cause of diarrhea in both<br />

HIV-infected and uninfected children, but in the<br />

immunocompromised child it results in prolonged<br />

disease and high fluid losses. 84–86 It may be<br />

responsible for up to 25% of persistent diarrhea in<br />

HIV-infected children in sub-Saharan Africa. 59,72<br />

Infection is usually through person-to-person<br />

contact, food contamination (e.g. unwashed raw<br />

vegetables or fruit) and contaminated water. Two<br />

distinct genotypes cause clinical disease in HIVinfected<br />

children: human and bovine (calf)<br />

types. 87 The clinical and epidemiological differ-<br />

ences between these genotypic variations is<br />

unclear, but the human genotype may be associated<br />

with a less severe clinical course in HIVinfected<br />

adults. Identifying C. parvum is difficult<br />

on direct microscopy; without experienced laboratory<br />

staff and appropriate stains, the diagnosis is<br />

easily missed. 88 In most developing countries,<br />

enzyme-linked immunosorbent assay (ELISA) or<br />

immunofluorescence methods to enhance identification<br />

rates are not routinely available. Shedding<br />

of the parasite is sporadic, and several stools<br />

should be examined to confirm or reject the diagnosis.<br />

Endoscopy and duodenal biopsy and aspirate<br />

examination further enhance the diagnostic<br />

yield. 45<br />

The most common site of infection is the duodenum,<br />

where it causes marked, though sometimes<br />

patchy, villous atrophy and reactive epithelial<br />

changes. Co-infection with CMV may exacerbate<br />

these responses. Other sites of infection include<br />

the stomach and colon (where cryptitis and apoptosis<br />

may occur) and the bile tract (which may be<br />

associated with pancreatitis). 89 Gut permeability<br />

may be markedly increased and fat malabsorption<br />

may be significant. 90,91 Protracted, or even<br />

intractable, secretory diarrhea with high fluid<br />

losses follows. It is often associated with weight<br />

loss, and up to 2–4% loss of body weight has been<br />

reported. 36 In HIV-infected children infection and<br />

a higher case fatality rate are more common in<br />

those with a lower CD4 count. 59<br />

In developing countries management is largely<br />

supportive to prevent severe dehydration and to<br />

recover lost weight through extended nutritional<br />

support. Several antimicrobial agents including<br />

spiromycin, parmomycin, azithromycin and nitazoxanide,<br />

and other approaches such as oral bovine<br />

immunoglobulin have been used with limited<br />

benefit. 92–94 The most effective treatment by far<br />

remains highly active antiretroviral treatment,<br />

which restores gut mucosal CD4 cells with eradication<br />

of opportunistic infections including C.<br />

parvum. 95<br />

Candida<br />

Oral thrush is one of the most characteristic signs in<br />

young children with HIV/AIDS, appearing as<br />

white–yellow plaques that cannot be easily scraped<br />

off the buccal mucosa. It is not, however, reliable or


specific as an indicator of HIV infection; recurrent<br />

episodes are better markers of HIV-infection in both<br />

children and adults. Thrush is often reported as a<br />

risk factor for breast-feeding transmission of HIV,<br />

presumably by inducing mucosal breaks facilitating<br />

viral entry, but the direction of causality is unclear<br />

from the available data. 96,97 Gut colonization<br />

follows oral infection with Candida species.<br />

Candidiasis of the mouth and esophagus is<br />

associated with poor appetite and weight loss.<br />

Esophageal inflammation is reported in up to 40%<br />

of children with AIDS and can be severe; 45,98 occasionally<br />

a necrotizing esophagitis which may bleed<br />

or even perforate can occur. The main differential<br />

diagnosis is herpes, CMV and Mycobacterium<br />

avium intracellulare. Candida may also produce<br />

inflammation and erosions in the gastric mucosa<br />

and the large and small intestines. The parasite can<br />

occasionally cause local abscesses, which may<br />

disseminate resulting in generalized candidiasis.<br />

Endoscopy and biopsy are generally required to<br />

diagnose invasive Candida, but in developing countries<br />

a high index of suspicion may be all that is<br />

available. Fluconazole is generally effective,<br />

although resistance does occur and higher doses are<br />

sometimes needed.<br />

Cytomegalovirus<br />

Gastrointestinal CMV infection in children most<br />

commonly presents with fever and persistent diarrhea.<br />

99,100 CMV-associated disease including retinitis,<br />

pneumonia, encephalitis and gastrointestinal<br />

involvement is estimated to affect up to 40% of<br />

adult AIDS patients, but is probably less frequent in<br />

children. 101 Symptomatic infection is more<br />

common and severe in HIV-infected children less<br />

than 12 months of age and with low age-corrected<br />

CD4 counts. 99 In children the colon is the most<br />

common site of gastrointestinal infection, followed<br />

by the small intestine and then the esophagus.<br />

Mucosal ulceration may sometimes result in<br />

massive hemorrhage and perforation of the large<br />

bowel, and strictures may follow CMV infection of<br />

the esophagus. 102 Diagnosis can be difficult. The<br />

virus may remain latent for long periods of time and<br />

neither serological tests nor positive urine cultures<br />

necessarily imply active disease. Children with<br />

chronic diarrhea, fever and no other identifiable<br />

pathogen should be investigated for CMV infection<br />

with lower gastrointestinal endoscopy and biopsy.<br />

Enteric infections in HIV-infected children 119<br />

CMV DNA quantification (CMV viral load) by polymerase<br />

chain reaction (PCR) can be used to decide<br />

when to offer prophylactic or active treatment.<br />

Prophylaxis and treatment with ganciclovir can<br />

prevent complications and significantly reduce<br />

morbidity, although the infection is not eradicated<br />

and relapses are still frequent. 103 Multiple pinpoint<br />

perforations or near perforations of the bowel may<br />

occur in the small bowel, ascending colon or<br />

both. 104 The prognosis is poor with or without<br />

ganciclovir.<br />

Rotavirus<br />

Studies in sub-Saharan Africa show that rotavirus is<br />

a common cause of diarrhea in HIV-infected children.<br />

105 It is not, however, found more commonly,<br />

shed for longer or associated with greater clinical<br />

severity than in children uninfected by HIV in<br />

similar settings; nor do children with more<br />

advanced HIV disease demonstrate any additional<br />

susceptibility. 106 Rotavirus was not associated with<br />

changes in viral load or CD4 counts in a small study<br />

in Malawi. 107 The pending availability of two new<br />

live oral rotavirus vaccines highlights the importance<br />

of understanding the relationship, and potential<br />

interaction, between these two viral infections.<br />

Clinical trials are necessary to ensure that any<br />

vaccine is safe and immunogenic in this population.<br />

Microsporidia spp<br />

Intestinal microsporidiosis is increasingly being<br />

reported in patients with HIV. 83,108 These intracellular<br />

protozoa are able to infect most animal species<br />

and, in humans, five genera (Enterocytozoon,<br />

Encephalitozoon, Septata, Pleistophora and<br />

Nosema) are associated with pulmonary, ocular,<br />

muscular, renal, intestinal and hepatic clinical<br />

disease. In Thailand, Enterocytozoon bieneusi was<br />

diagnosed in 25% and 15% of HIV-infected and<br />

uninfected children presenting with diarrhea,<br />

respectively. 81 In Uganda, however, it was found in<br />

17% of children of unknown HIV status attending<br />

hospital out-patient clinics for conditions other<br />

than diarrhea. 82 In contrast, microsporidiosis in<br />

HIV-infected adults produces chronic watery diarrhea<br />

and wasting, suggesting that responses in children<br />

may be quite different. 82,109 Villous atrophy<br />

and reduced brush-border enzyme activity may<br />

cause lactose intolerance. 110


120<br />

HIV and the intestine<br />

Diagnosis is made on stool microscopy using a<br />

modified trichrome stain, but this requires considerable<br />

technical skill to identify organisms<br />

correctly. Identification of parasites in stool or<br />

biopsy can be optimized by using PCR methods or<br />

electron microscopy on upper gastrointestinal<br />

biopsy. 111 Albendazole is an effective treatment for<br />

Encephalitozoon spp infections. 77<br />

Management of diarrhea and<br />

nutritional support<br />

HIV infection in children is often thought to be a<br />

rapidly and uniformly lethal disease. In Rwanda,<br />

however, 40% of children with perinatal infection<br />

survive for 5 years without antiretroviral treatment,<br />

and there are many children in sub-Saharan<br />

Africa with vertically acquired HIV infection<br />

attending schools and growing into adolescence.<br />

112 Optimizing the quality of life for these<br />

children is a necessary challenge, especially when<br />

the option of effective antiretroviral therapy is<br />

absent. The approaches outlined below are<br />

intended for developing countries.<br />

Diarrhea<br />

In most clinical settings in southern Africa it is not<br />

possible to investigate children presenting with<br />

recurrent or persistent diarrhea extensively. Using<br />

a syndromic approach for managing diarrhea in<br />

both HIV-infected and uninfected children is<br />

therefore appropriate. The WHO/UNICEF<br />

Integrated Management of Childhood Illnesses<br />

(IMCI) recommends that children with a history of<br />

persistent diarrhea and/or reported weight loss in<br />

the previous 3 months should be assessed for<br />

possible HIV infection. 113 Where there are no<br />

specific guidelines for HIV-infected children, the<br />

WHO guidelines for the management of severe<br />

malnutrition, including persistent diarrhea, are<br />

helpful.<br />

An approach for resource-poor countries would<br />

be:<br />

(1) Assess for and correct dehydration, hypoglycemia,<br />

hypothermia and electrolyte disturbances,<br />

especially K + deficiency;<br />

(2) Treat concurrent bacterial infections, e.g.<br />

pneumonia, Pneumocystis carinii pneumonia<br />

(PCP) and urinary tract infection, and exclude<br />

TB. This often means giving antibiotics empirically,<br />

e.g. cefuroxime, gentamycin and cotrimoxazole;<br />

(3) Start low lactose (< 3.2 g/kg) containing feeds<br />

such as F-75 (WHO) and porridge;<br />

(4) Provide vitamin A, zinc and multivitamins,<br />

including folate. See below for comment on<br />

zinc supplements;<br />

(5) If diarrhea persists then test stools for lactose<br />

intolerance using Clinitest ® tablets;<br />

(6) If Clinitest is positive then exclude all lactose<br />

from diet – use milks containing maltodextran<br />

as the main carbohydrate;<br />

(7) If Clinitest is negative then send repeat stools<br />

for routine microbiological assessment,<br />

including for Shigella, non-typhi Salmonella,<br />

Cryptosporidium, microsporidia and TB. Treat<br />

accordingly. Cryptosporidia may be shed intermittently;<br />

three stools should be sent to<br />

exclude diagnosis. Experienced staff are<br />

required to identify cryptosporidia and<br />

microsporidia on routine stool analysis.<br />

Prolonged fluid and nutritional support is<br />

often required for patients with cryp-<br />

(8)<br />

tosporidiosis;<br />

If no pathogen is identified then treat for bacterial<br />

overgrowth, e.g. cholestyramine for 5 days<br />

and neomycin for 3 days;<br />

(9) If diarrhea persists check for monosaccharide<br />

intolerance; test for glucose in stool using<br />

Clinitest or Glucostix ®. If positive, check for<br />

excessive intake of oral rehydration solution<br />

(ORS). Use maltodextran (glucose polymer)<br />

containing feeds and deliver milk by continuous<br />

slow infusion through a nasogastric tube.<br />

Use plain boiled water for oral rehydration<br />

rather than ORS. Only revert back to bolus<br />

feeds once the stool becomes formed again and<br />

glucose is not present. This takes longer in<br />

children who are more wasted or who have<br />

severe diarrhea;<br />

(10) Where resources are available, perform upper<br />

gastrointestinal endoscopy to increase the<br />

microbiological diagnostic yield. If visible or<br />

microscopic blood is present in stools then<br />

perform sigmoidoscopy for biopsy and culture;


(11) Where there is persistent pyrexia consider<br />

CMV colitis and exclude TB;<br />

(12) If no cause for persisting diarrhea is evident<br />

then consider antimotility drugs such as<br />

loperamide (HIV enteropathy is a diagnosis of<br />

exclusion);<br />

(13) Older children should be asked about abdominal<br />

pain and on these occasions underlying<br />

opportunistic infections such as TB,<br />

Cryptosporidium or CMV should be excluded;<br />

opiates such as codeine phosphate may be<br />

helpful;<br />

(14) Attention must be given in the weeks following<br />

discharge to restoring, as much as possible,<br />

any weight loss that has occurred. It is<br />

often very difficult to achieve this during<br />

hospitalization, and in busy hospitals significant<br />

weight gain cannot be used as a criterion<br />

for discharge (see Nutrition, below).<br />

Parenteral nutrition is not generally feasible in<br />

most developing countries because of the risk of<br />

systemic infection and metabolic complications.<br />

Terminal care<br />

When children are admitted with evidence of clinically<br />

advanced HIV infection such as diarrhea<br />

and severe wasting, clinicians and carers must<br />

consider what is reasonable and right for the child.<br />

The first admission with diarrhea often results in<br />

HIV testing. Many mothers discover for the first<br />

time that both she, and her child, are HIV-infected.<br />

In this situation it is appropriate to use all<br />

resources available to ensure that the child recovers<br />

and is able to go home. However, the treatment<br />

paradigm and objectives for care often change<br />

when the child has suffered several admissions,<br />

and the family has had some time to adapt to the<br />

diagnosis. In the absence of antiviral drugs, relieving<br />

discomfort becomes the overriding priority,<br />

and being less aggressive when complications<br />

such as concurrent severe bacterial infections<br />

intervene may be more caring for the child.<br />

In resourced settings<br />

While the same principles apply, the use of invasive<br />

diagnostic methods, i.e. upper and lower<br />

intestinal endoscopy, should be used early to<br />

Management of diarrhea and nutritional support 121<br />

determine whether a treatable enteric pathogen is<br />

present. Parenteral feeding may be a safe option if<br />

severe malabsorption is present. Antiretroviral<br />

drugs should be initiated if the child has not been<br />

previously treated.<br />

Micronutrients and HIV-related diarrhea<br />

Multiple micronutrient deficiencies have been<br />

reported in both HIV-infected adults and children<br />

with, or without, diarrhea. Biochemical indicators<br />

of vitamin and trace element status may be<br />

misleading and reflect redistribution or acutephase<br />

responses rather true body depletion. A few<br />

studies have shown that micronutrient supplementation<br />

in HIV-infected or exposed children is<br />

associated with improved morbidity or immune<br />

function. 114–116 In children uninfected with HIV,<br />

zinc supplementation reduces the incidence, duration<br />

and severity of acute and chronic diarrhea and<br />

promotes recovery of the mucosal lining. 117,118<br />

This effect has not so far been demonstrated in<br />

HIV-infected children.<br />

Vitamin A does not seem significantly to influence<br />

the course of acute or persistent diarrhea in uninfected<br />

children, but does decrease the severity<br />

and likelihood of recurrence. 119 HIV-infected children<br />

with persistent diarrhea should receive<br />

vitamin A (0–6 months, 50000IU; 6–12 months,<br />

100000 IU; >12 months, 200000IU daily for 2<br />

days), zinc sulfate or gluconate 2mg/kg for 2 weeks<br />

and multivitamin preparations including folate for<br />

2–4 weeks.<br />

Nutrition<br />

Much of the impact of diarrheal illnesses and<br />

repeated opportunistic infections on the health of<br />

HIV-infected children is mediated by its effect on<br />

nutrition. Loss of lean body tissue is consistently<br />

seen in adults and children with advanced disease<br />

and is a strong predictor of death. 120 Resting<br />

energy expenditure (REE) in adults increases by<br />

about 10% once they are infected with HIV, but<br />

this does not been seem to be the case in<br />

children. 53,121 However, even if REE increases<br />

modestly, total energy expenditure may not<br />

increase, because of inactivity. Rather, weight loss<br />

is most likely to be due to decreased energy intake,


122<br />

HIV and the intestine<br />

especially during the recovery period from opportunistic<br />

infections. Nutritional interventions,<br />

however, have generally only reversed weight loss<br />

through gain of fat rather than lean body tissue.<br />

There is work to suggest that different opportunistic<br />

infections may have differential effects on<br />

nutrition with some being capable of impairing<br />

anabolism and effective utilization of energy from<br />

food.<br />

WHO currently recommends that infected children<br />

should increase their overall energy intake by<br />

about 10% in order to maintain normal health,<br />

growth and activity. In chronic illness such as TB<br />

infection or chronic lung disease, energy intake<br />

should increase by 25–30%. During acute<br />

illnesses, particularly when recovering from acute<br />

weight loss, these requirements may increase to<br />

50–100% extra energy. Protein should represent<br />

about 10–15% of energy. These goals should<br />

ideally be achieved through dietary approaches<br />

rather than by specialized supplements which<br />

may not be available or affordable in most developing<br />

countries. 122<br />

Surgical aspects of HIV infection<br />

The prevalence of HIV disease among pediatric<br />

surgical patients in southern Africa has increased<br />

dramatically and is frequently considered as part<br />

of the differential diagnosis. Abdominal pain is not<br />

uncommon in adults with HIV, but there are no<br />

data on children. An underlying pathological<br />

cause can be identified in most cases 123 and<br />

include TB abdomen, cryptosporidia and CMV.<br />

There are four common surgical manifestations of<br />

gastrointestinal HIV disease: destructive lesions,<br />

opportunistic infections, primary peritonitis and<br />

tumors.<br />

Destructive lesions<br />

Rectovaginal fistulae are an extremely common<br />

presentation of HIV disease in female<br />

infants. 124,125 In the past this was treated with a<br />

defunctioning colostomy followed by closure of<br />

the fistula. Recurrence was frequent. Antiretroviral<br />

therapy may improve surgical results, but in the<br />

absence of such treatment it is generally better to<br />

leave the fistula alone. 126 In males, there is the less<br />

common presentation of a rectourethral fistula<br />

which always requires surgical repair.<br />

Occasionally stricture formation may follow sclerosis<br />

of the lower esophagus after repeated ulcerative<br />

disease such as CMV or invasive candidiasis.<br />

Opportunistic infections<br />

Intestinal perforations due to CMV are described<br />

above. 104 HIV occasionally presents as cancrum<br />

oris, where early treatment with penicillin and<br />

metronidazole may help, but excision and reconstruction<br />

may be required for large areas of fullthickness<br />

skin and tissue loss.<br />

Primary peritonitis<br />

This is sometimes seen in the older child who<br />

presents with a clinical picture suggestive of<br />

appendicitis. At surgery an odorless pus is found<br />

in the abdomen; pneumococcus is often isolated.<br />

Tumors<br />

Kaposi’s sarcoma in the gut may present with<br />

rectal bleeding, and other less common smoothmuscle<br />

tumors may rarely present as an intussusception<br />

and intestinal obstruction. 127,128<br />

Condyloma accuminata that may vary in size from<br />

isolated lesions to large pancake lesions can cover<br />

the perineum and genitalia. The more extensive<br />

lesions cause severe discomfort and problems with<br />

local hygiene. Isolated lesions may respond to<br />

podophyllin, but diathermy results in annular<br />

scarring and anal stenosis. Treatment with interferon<br />

may be helpful.<br />

Sialadenitis is a frequent complaint. Parotid size<br />

may increase and decrease intermittently, and pain<br />

may be due to superimposed bacterial infection,<br />

TB, bleeding into cysts or malignant change. Acute<br />

pain should be treated with antibiotics and analgesia;<br />

if the parotid continues to enlarge and is<br />

painful, then fine-needle or open biopsy should be<br />

performed to exclude TB or malignant change.<br />

Corticosteroids may be helpful.


REFERENCES<br />

1. McDougal JS, Mawle A, Cort SP et al. Cellular tropism<br />

of the human retrovirus HTLV-III/LAV. I. Role of T cell<br />

activation and expression of the T4 antigen. J Immunol<br />

1985; 135: 3151–3162.<br />

2. Lapenta C, Boirivant M, Marini M et al. Human intestinal<br />

lamina propria lymphocytes are naturally permissive<br />

to HIV-1 infection. Eur J Immunol 1999; 29:<br />

1202–1208.<br />

3. Rollins NC, Filteau SM, Coutsoudis A, Tomkins AM.<br />

Feeding mode, intestinal permeability, and neopterin<br />

excretion: a longitudinal study in infants of HIVinfected<br />

South African women. J Acquir Immune Defic<br />

Syndr 2001; 28: 132–139.<br />

4. Meng G, Wei X, Wu X et al. Primary intestinal epithelial<br />

cells selectively transfer R5 HIV-1 to CCR5+ cells. Nat<br />

Med 2002; 8: 150-156.<br />

5. Poles MA, Elliott J, Taing P et al. A preponderance of<br />

CCR5(+) CXCR4(+) mononuclear cells enhances<br />

gastrointestinal mucosal susceptibility to human<br />

immunodeficiency virus type 1 infection. J Virol 2001;<br />

75: 8390–8399.<br />

6. Wolinsky SM, Wike CM, Korber BT et al. Selective<br />

transmission of human immunodeficiency virus type-1<br />

variants from mothers to infants. Science 1992; 255:<br />

1134–1137.<br />

7. Amerongen HM, Weltzin R, Farnet CM et al.<br />

Transepithelial transport of HIV-1 by intestinal M cells:<br />

a mechanism for transmission of AIDS. J Acquir<br />

Immune Defic Syndr 1991; 4: 760–765.<br />

8. Dunn DT, Newell ML, Ades AE, Peckham CS. Risk of<br />

human immunodeficiency virus type 1 transmission<br />

through breastfeeding. Lancet 1992; 340: 585–588.<br />

9. Lewis P, Nduati R, Kreiss JK et al. Cell-free human<br />

immunodeficiency virus type 1 in breast milk. J Infect<br />

Dis 1998; 177: 34–39.<br />

10. Leroy V, Karon JM, Alioum A et al. Postnatal transmission<br />

of HIV-1 after a maternal short-course zidovudine<br />

peripartum regimen in West Africa. AIDS 2003; 17:<br />

1493–1501.<br />

11. Hocini H, Bomsel M. Infectious human immunodeficiency<br />

virus can rapidly penetrate a tight human epithelial<br />

barrier by transcytosis in a process impaired by<br />

mucosal immunoglobulins. J Infect Dis 1999; 179(Suppl<br />

3): S448–S453.<br />

12. McNeely TB, Dealy M, Dripps DJ et al. Secretory leukocyte<br />

protease inhibitor: a human saliva protein exhibiting<br />

anti-human immunodeficiency virus 1 activity in<br />

vitro. J Clin Invest 1995; 96: 456–464.<br />

13. Janoff EN, Scamurra RW, Sanneman TC et al. Human<br />

immunodeficiency virus type 1 and mucosal humoral<br />

defense. J Infect Dis 1999; 179(Suppl 3): S475–S479.<br />

14. Willumsen JF, Filteau SM, Coutsoudis A et al.<br />

Breastmilk RNA viral load in HIV-infected South<br />

African women: effects of subclinical mastitis and<br />

infant feeding. AIDS 2003; 17: 407–414.<br />

15. Fearon DT, Locksley RM. The instructive role of innate<br />

immunity in the acquired immune response. Science<br />

1996; 272: 50–53.<br />

16. Medzhitov R, Janeway CA Jr. Innate immunity: impact<br />

on the adaptive immune response. Curr Opin Immunol<br />

1997; 9: 4–9.<br />

17. Kalliomaki M, Salminen S, Arvilommi H et al.<br />

Probiotics in primary prevention of atopic disease: a<br />

randomised placebo-controlled trial. Lancet 2001; 357:<br />

1076–1079.<br />

References 123<br />

18. Devito C, Broliden K, Kaul R et al. Mucosal and plasma<br />

IgA from HIV-1-exposed uninfected individuals inhibit<br />

HIV-1 transcytosis across human epithelial cells. J<br />

Immunol 2000; 165: 5170–5176.<br />

19. Alfsen A, Iniguez P, Bouguyon E, Bomsel M. Secretory<br />

IgA specific for a conserved epitope on gp41 envelope<br />

glycoprotein inhibits epithelial transcytosis of HIV-1. J<br />

Immunol 2001; 166: 6257–6265.<br />

20. Veazey RS, Shattock RJ, Pope M et al. Prevention of<br />

virus transmission to macaque monkeys by a vaginally<br />

applied monoclonal antibody to HIV-1 gp120. Nat Med<br />

2003; 9: 343–346.<br />

21. Lim SG, Condez A, Lee CA et al. Loss of mucosal CD4<br />

lymphocytes is an early feature of HIV infection. Clin<br />

Exp Immunol 1993; 92: 448–454.<br />

22. Schneider T, Jahn HU, Schmidt W et al. Loss of CD4 T<br />

lymphocytes in patients infected with human immunodeficiency<br />

virus type 1 is more pronounced in the<br />

duodenal mucosa than in the peripheral blood. Berlin<br />

Diarrhea/Wasting Syndrome Study Group. Gut 1995; 37:<br />

524–529.<br />

23. Clayton F, Snow G, Reka S, Kotler DP. Selective depletion<br />

of rectal lamina propria rather than lymphoid<br />

aggregate CD4 lymphocytes in HIV infection. Clin Exp<br />

Immunol 1997; 107: 288–292.<br />

24. Kotler DP, Scholes JV, Tierney AR. Intestinal plasma cell<br />

alterations in acquired immunodeficiency syndrome.<br />

Dig Dis Sci 1987; 32: 129–138.<br />

25. Schneider T, Zippel T, Schmidt W et al. Secretory<br />

immunity in HIV infection. Pathobiology 1998; 66:<br />

131–138.<br />

26. Kakai R, Bwayo JJ, Wamola IA et al. Effect of human<br />

immunodeficiency virus on local immunity in children<br />

with diarrhoea. East Afr Med J 1995; 72: 699–702.<br />

27. Bard E, Laibe S, Clair S et al. Nonspecific secretory<br />

immunity in HIV-infected patients with oral candidiasis.<br />

J Acquir Immune Defic Syndr 2002; 31: 276–284.<br />

28. Chadwick EG, Chang G, Decker MD et al. Serologic<br />

response to standard inactivated influenza vaccine in<br />

human immunodeficiency virus-infected children.<br />

Pediatr Infect Dis J 1994; 13: 206–211.<br />

29. Olsson J, Poles M, Spetz AL et al. Human immunodeficiency<br />

virus type 1 infection is associated with significant<br />

mucosal inflammation characterized by increased<br />

expression of CCR5, CXCR4, and beta-chemokines. J<br />

Infect Dis 2000; 182: 1625–1635.<br />

30. Bode H, Schmitz H, Fromm M et al. IL-1beta and TNFalpha,<br />

but not IFN-alpha, IFN-gamma, IL-6 or IL-8, are<br />

secretory mediators in human distal colon. Cytokine<br />

1998; 10: 457–465.<br />

31. Colebunders R, Francis H, Mann JM et al. Persistent<br />

diarrhea, strongly associated with HIV infection in<br />

Kinshasa, Zaire. Am J Gastroenterol 1987; 82: 859–864.<br />

32. Chintu C, DuPont HL, Kaile T et al. Human immunodeficiency<br />

virus-associated diarrhea and wasting in<br />

Zambia: selected risk factors and clinical associations.<br />

Am J Trop Med Hyg 1998; 59: 38–41.<br />

33. Monkemuller KE, Call SA, Lazenby AJ, Wilcox CM.<br />

Declining prevalence of opportunistic gastrointestinal<br />

disease in the era of combination antiretroviral therapy.<br />

Am J Gastroenterol 2000; 95: 457–462.<br />

34. Nannini EC, Okhuysen PC. HIV1 and the gut in the era<br />

of highly active antiretroviral therapy. Curr<br />

Gastroenterol Rep 2002; 4: 392–398.


124<br />

HIV and the intestine<br />

35. Kotler DP, Francisco A, Clayton F et al. Small intestinal<br />

injury and parasitic diseases in AIDS. Ann Intern Med<br />

1990; 113: 444–449.<br />

36. Carbonnel F, Beaugerie L, Abou RA et al. Macronutrient<br />

intake and malabsorption in HIV infection: a comparison<br />

with other malabsorptive states. Gut 1997; 41:<br />

805–810.<br />

37. Yolken RH, Hart W, Oung I et al. Gastrointestinal<br />

dysfunction and disaccharide intolerance in children<br />

infected with human immunodeficiency virus. J Pediatr<br />

1991; 118: 359–363.<br />

38. Miller TL, Orav EJ, Martin SR et al. Malnutrition and<br />

carbohydrate malabsorption in children with vertically<br />

transmitted human immunodeficiency virus 1 infection.<br />

Gastroenterology 1991; 100: 1296–1302.<br />

39. The Italian Paediatric Intestinal/HIV Study Group.<br />

Intestinal malabsorption of HIV-infected children: relationship<br />

to diarrhoea, failure to thrive, enteric microorganisms<br />

and immune impairment. AIDS 1993; 7:<br />

1435–1440.<br />

40. Lepage P, Spira R, Kalibala S et al. Care of human<br />

immunodeficiency virus-infected children in developing<br />

countries. International Working Group on Motherto-Child<br />

Transmission of HIV. Pediatr Infect Dis J 1998;<br />

17: 581–586.<br />

41. Keating J, Bjarnason I, Somasundaram S et al. Intestinal<br />

absorptive capacity, intestinal permeability and jejunal<br />

histology in HIV and their relation to diarrhoea. Gut<br />

1995; 37: 623–629.<br />

42. Kotler DP, Reka S, Chow K, Orenstein JM. Effects of<br />

enteric parasitoses and HIV infection upon small<br />

intestinal structure and function in patients with AIDS.<br />

J Clin Gastroenterol 1993; 16: 10–15.<br />

43. Fontana M, Boldorini R, Zuin G et al. Ultrastructural<br />

changes in the duodenal mucosa of HIV-infected children.<br />

J Pediatr Gastroenterol Nutr 1993; 17: 255–259.<br />

44. Thea DM, St Louis ME, Atido U et al. A prospective<br />

study of diarrhea and HIV-1 infection among 429<br />

Zairian infants. N Engl J Med 1993; 329: 1696–1702.<br />

45. Miller TL, McQuinn LB, Orav EJ. Endoscopy of the<br />

upper gastrointestinal tract as a diagnostic tool for children<br />

with human immunodeficiency virus infection. J<br />

Pediatr 1997; 130: 766–773.<br />

46. Oktedalen O, Skar V, Dahl E, Serck-Hanssen A. Changes<br />

in small intestinal structure and function in HIVinfected<br />

patients with chronic diarrhoea. Scand J Infect<br />

Dis 1998; 30: 459–463.<br />

47. Pernet P, Vittecoq D, Kodjo A et al. Intestinal absorption<br />

and permeability in human immunodeficiency virusinfected<br />

patients. Scand J Gastroenterol 1999; 34: 29–34.<br />

48. Taylor C, Hodgson K, Sharpstone D et al. The prevalence<br />

and severity of intestinal disaccharidase deficiency<br />

in human immunodeficiency virus-infected<br />

subjects. Scand J Gastroenterol 2000; 35: 599–606.<br />

49. Rollins NC, Wittenberg DF, Coovadia HM. Trends in HIV<br />

prevalence outcome and monosaccharide intolerance in<br />

a paediatric diarrhoea unit in South Africa.<br />

Commonwealth Congress on Diarrhoea and<br />

Malnutrition, Karachi, Pakistan, 1997.<br />

50. Sharpstone D, Neild P, Crane R et al. Small intestinal<br />

transit, absorption, and permeability in patients with<br />

AIDS with and without diarrhoea. Gut 1999; 45: 70–76.<br />

51. Kotler D, Haroutiounian G, Greenberg R. Increased bile<br />

salt deconjugation in AIDS. Gastroenterology 1985; 88:<br />

1455.<br />

52. Sentongo TA, Rutstein RM, Stettler N et al. Association<br />

between steatorrhea, growth, and immunologic status in<br />

children with perinatally acquired HIV infection. Arch<br />

Pediatr Adolesc Med 2001; 155: 149–153.<br />

53. Sharpstone DR, Ross HM, Gazzard BG. The metabolic<br />

response to opportunistic infections in AIDS. AIDS<br />

1996; 10: 1529–1533.<br />

54. Laine L, Garcia F, McGilligan K et al. Protein-losing<br />

enteropathy and hypoalbuminemia in AIDS. AIDS 1993;<br />

7: 837–840.<br />

55. Guarino A, Tarallo L, Guandalini S et al. Impaired<br />

intestinal function in symptomatic HIV infection. J<br />

Pediatr Gastroenterol Nutr 1991; 12: 453–458.<br />

56. Rollins NC, Filteau SM, Elson I, Tomkins AM. Vitamin<br />

A supplementation of South African children with<br />

severe diarrhea: optimum timing for improving<br />

biochemical and clinical recovery and subsequent<br />

vitamin A status. Pediatr Infect Dis J 2000; 19: 284–289.<br />

57. Gove S. Integrated management of childhood illness by<br />

outpatient health workers: technical basis and overview.<br />

The WHO Working Group on Guidelines for Integrated<br />

Management of the Sick Child. Bull. World Health<br />

Organ 1997; 75(Suppl 1): 7–24.<br />

58. Brink AK, Mahe C, Watera C et al. Diarrhea, CD4 counts<br />

and enteric infections in a community-based cohort of<br />

HIV-infected adults in Uganda. J Infect 2002; 45:<br />

99–106.<br />

59. Rollins NC, Pent M, Kindra G et al. CD4 count, HIV<br />

viral load and enteric pathogens in HIV-infected children<br />

with persistent diarrhea in Durban, South Africa.<br />

Pediatric Academic Societies & American Academy of<br />

Pediatrics Joint Meeting, Boston, 2000: Poster abstr 1620.<br />

60. Quesnel A, Moja P, Blanche S et al. Early impairment of<br />

gut mucosal immunity in HIV-1-infected children. Clin<br />

Exp Immunol 1994; 97: 380–385.<br />

61. Kotler DP, Gaetz HP, Lange M et al. Enteropathy associated<br />

with the acquired immunodeficiency syndrome.<br />

Ann Intern Med 1984; 101: 421–428.<br />

62. Kelly P, Davies SE, Mandanda B et al. Enteropathy in<br />

Zambians with HIV related diarrhoea: regression modelling<br />

of potential determinants of mucosal damage. Gut<br />

1997; 41: 811–816.<br />

63. Zeitz M, Ullrich R, Schneider T et al. HIV/SIV enteropathy.<br />

Ann NY Acad Sci 1998; 859: 139–148.<br />

64. Batman PA, Miller AR, Forster SM et al. Jejunal<br />

enteropathy associated with human immunodeficiency<br />

virus infection: quantitative histology. J Clin Pathol<br />

1989; 42: 275–281.<br />

65. Wilcox CM, Waites KB, Smith PD. No relationship<br />

between gastric pH, small bowel bacterial colonisation,<br />

and diarrhoea in HIV-1 infected patients. Gut 1999; 44:<br />

101–105.<br />

66. Bjarnason I, Sharpstone DR, Francis N et al. Intestinal<br />

inflammation, ileal structure and function in HIV. AIDS<br />

1996; 10: 1385–1391.<br />

67. Delezay O, Yahi N, Tamalet C et al. Direct effect of type<br />

1 human immunodeficiency virus (HIV-1) on intestinal<br />

epithelial cell differentiation: relationship to HIV-1<br />

enteropathy. Virology 1997; 238: 231–242.<br />

68. Kotler DP. Characterization of intestinal disease associated<br />

with human immunodeficiency virus infection and<br />

response to antiretroviral therapy. J Infect Dis 1999;<br />

179(Suppl 3): S454–S456.<br />

69. Asmuth DM, Hammer SM, Wanke CA. Physiological<br />

effects of HIV infection on human intestinal epithelial<br />

cells: an in vitro model for HIV enteropathy. AIDS 1994;<br />

8: 205–211.<br />

70. Canani RB, Cirillo P, Mallardo G et al. Effects of HIV-1<br />

Tat protein on ion secretion and on cell proliferation in<br />

human intestinal epithelial cells. Gastroenterology 2003;<br />

124: 368–376.<br />

71. Otegbayo JA, Oluwasola AO, Akang EE. Collagenous<br />

colitis in an adult patient with chronic diarrhoea: case<br />

report. East Afr Med J 2001; 78: 272–274.


72. Chintu C, Luo C, Baboo S et al. Intestinal parasites in<br />

HIV-seropositive Zambian children with diarrhoea. J<br />

Trop Pediatr 1995; 41: 149–152.<br />

73. Powell KR. Guidelines for the care of children and<br />

adolescents with HIV infection. Approach to gastrointestinal<br />

manifestations in infants and children with HIV<br />

infection. J Pediatr 1991; 119: S34–S40.<br />

74. Yan Z, Nguyen S, Poles M et al. Adenovirus colitis in<br />

human immunodeficiency virus infection: an underdiagnosed<br />

entity. Am J Surg Pathol 1998; 22: 1101–1016.<br />

75. Pollok RC. Viruses causing diarrhoea in AIDS. Novartis<br />

Found Symp 2001; 238: 276–283.<br />

76. Orenstein JM, Dieterich DT. The histopathology of 103<br />

consecutive colonoscopy biopsies from 82 symptomatic<br />

patients with acquired immunodeficiency syndrome:<br />

original and look-back diagnoses. Arch Pathol Lab Med<br />

2001; 125: 1042–1046.<br />

77. Kelly P, Baboo KS, Wolff M et al. The prevalence and<br />

aetiology of persistent diarrhoea in adults in urban<br />

Zambia. Acta Trop 1996; 61: 183–190.<br />

78. Bini EJ, Weinshel EH, Gamagaris Z. Comparison of<br />

duodenal with jejunal biopsy and aspirate in chronic<br />

human immunodeficiency virus-related diarrhea. Am J<br />

Gastroenterol 1998; 93: 1837–1840.<br />

79. Sewankambo N, Mugerwa RD, Goodgame R et al.<br />

Enteropathic AIDS in Uganda. An endoscopic, histological<br />

and microbiological study. AIDS 1987; 1: 9–13.<br />

80. Pavia AT, Long EG, Ryder RW et al. Diarrhea among<br />

African children born to human immunodeficiency<br />

virus 1-infected mothers: clinical, microbiologic and<br />

epidemiologic features. Pediatr Infect Dis J 1992; 11:<br />

996–1003.<br />

81. Wanachiwanawin D, Chokephaibulkit K, Lertlaituan P<br />

et al. Intestinal microsporidiosis in HIV-infected children<br />

with diarrhea. Southeast Asian J Trop Med Public<br />

Health 2002; 33: 241–245.<br />

82. Tumwine JK, Kekitiinwa A, Nabukeera N et al.<br />

Enterocytozoon bieneusi among children with diarrhea<br />

attending Mulago Hospital in Uganda. Am J Trop Med<br />

Hyg 2002; 67: 299–303.<br />

83. Drobniewski F, Kelly P, Carew A et al. Human<br />

microsporidiosis in African AIDS patients with chronic<br />

diarrhea. J Infect Dis 1995; 171: 515–516.<br />

84. Enriquez EJ, Avila CR, Santos JI et al. Cryptosporidium<br />

infections in Mexican children: clinical, nutritional,<br />

enteropathogenic and diagnostic evaluations. Am J Trop<br />

Med Hyg 1997; 56: 254–257.<br />

85. Newman RD, Sears CL, Moore SR et al. Longitudinal<br />

study of Cryptosporidium infection in children in northeastern<br />

Brazil. J Infect Dis 1999; 180: 167–175.<br />

86. Cegielski JP, Ortega YR, McKee S et al. Cryptosporidium,<br />

enterocytozoon, and cyclospora infections in pediatric<br />

and adult patients with diarrhea in Tanzania. Clin Infect<br />

Dis 1999; 28: 314–321.<br />

87. Leav BA, Mackay MR, Anyanwu A et al. Analysis of<br />

sequence diversity at the highly polymorphic<br />

Cpgp40/15 locus among Cryptosporidium isolates from<br />

human immunodeficiency virus-infected children in<br />

South Africa. Infect Immun 2002; 70: 3881–3890.<br />

88. Mayer HB, Wanke CA. Diagnostic strategies in HIVinfected<br />

patients with diarrhea. AIDS 1994; 8:<br />

1639–1648.<br />

89. McGowan I, Hawkins AS, Weller IV. The natural history<br />

of cryptosporidial diarrhea in HIV-infected patients.<br />

AIDS 1993; 7: 349–354.<br />

90. Lima AA, Silva TM, Gifoni AM et al. Mucosal injury<br />

and disruption of intestinal barrier function in HIVinfected<br />

individuals with and without diarrhea and<br />

cryptosporidiosis in northeast Brazil. Am J Gastroenterol<br />

1997; 92: 1861–1866.<br />

References 125<br />

91. Guarino A, Castaldo A, Russo S et al. Enteric cryptosporidiosis<br />

in pediatric HIV infection. J Pediatr<br />

Gastroenterol Nutr 1997; 25: 182–187.<br />

92. Hicks P, Zwiener RJ, Squires J, Savell V. Azithromycin<br />

therapy for Cryptosporidium parvum infection in four<br />

children infected with human immunodeficiency virus.<br />

J Pediatr 1996; 129: 297–300.<br />

93. Amadi B, Mwiya M, Musuku J et al. Effect of nitazoxanide<br />

on morbidity and mortality in Zambian children<br />

with cryptosporidiosis: a randomised controlled trial.<br />

Lancet 2002; 360: 1375–1380.<br />

94. Greenberg PD, Cello JP. Treatment of severe diarrhea<br />

caused by Cryptosporidium parvum with oral bovine<br />

immunoglobulin concentrate in patients with AIDS. J<br />

Acquir Immune Defic Syndr Hum Retrovirol 1996; 13:<br />

348–354.<br />

95. Schmidt W, Wahnschaffe U, Schafer M et al. Rapid<br />

increase of mucosal CD4 T cells followed by clearance<br />

of intestinal cryptosporidiosis in an AIDS patient<br />

receiving highly active antiretroviral therapy.<br />

Gastroenterology 2001; 120: 984–987.<br />

96. Embree JE, Njenga S, Datta P et al. Risk factors for postnatal<br />

mother-child transmission of HIV-1. AIDS 2000;<br />

14: 2535–2541.<br />

97. Ekpini ER, Wiktor SZ, Satten GA et al. Late postnatal<br />

mother-to-child transmission of HIV-1 in Abidjan, Cote<br />

d’Ivoire. Lancet 1997; 349: 1054–1059.<br />

98. Haller JO, Cohen HL. Gastrointestinal manifestations of<br />

AIDS in children. Am J Roentgenol 1994; 162: 387–393.<br />

99. Chandwani S, Kaul A, Bebenroth D et al.<br />

Cytomegalovirus infection in human immunodeficiency<br />

virus type 1-infected children. Pediatr Infect Dis J 1996;<br />

15: 310–314.<br />

100. Ukarapol N, Chartapisak W, Lertprasertsuk N et al.<br />

Cytomegalovirus-associated manifestations involving<br />

the digestive tract in children with human immunodeficiency<br />

virus infection. J Pediatr Gastroenterol Nutr 2002;<br />

35: 669–673.<br />

101. Kitchen BJ, Engler HD, Gill VJ et al. Cytomegalovirus<br />

infection in children with human immunodeficiency<br />

virus infection. Pediatr Infect Dis J 1997; 16: 358–363.<br />

102. Kahn E, Greco MA, Daum S. Pathology of the gastrointestinal<br />

tract in pediatric AIDS. Surg Pathol 1994; 5:<br />

239–252.<br />

103. Wilcox CM, Straub RF, Schwartz DA. Cytomegalovirus<br />

esophagitis in AIDS: a prospective evaluation of clinical<br />

response to ganciclovir therapy, relapse rate, and longterm<br />

outcome. Am J Med 1995; 98: 169–176.<br />

104. Shaik AS, Hadley GP. Surgically significant CMV enterocolitis<br />

in children. 25th Biennial Congress of the<br />

South African Paediatric Association, and the South<br />

African Association of Paediatric Surgeons, 20–24 Oct,<br />

2002 (abstr).<br />

105. Oshitani H, Kasolo FC, Mpabalwani M et al. Association<br />

of rotavirus and human immunodeficiency virus infection<br />

in children hospitalized with acute diarrhea,<br />

Lusaka, Zambia. J Infect Dis 1994; 169: 897–900.<br />

106. Cunliffe NA, Gondwe JS, Kirkwood CD et al. Effect of<br />

concomitant HIV infection on presentation and outcome<br />

of rotavirus gastroenteritis in Malawian children. Lancet<br />

2001; 358: 550–555.<br />

107. Jere C, Cunliffe NA, Hoffman IF et al. Plasma HIV<br />

burden in Malawian children co-infected with<br />

rotavirus. AIDS 2001; 15: 1439–1442.<br />

108. Brasil P, Sodre FC, Cuzzi-Maya T et al. Intestinal<br />

microsporidiosis in HIV-positive patients with chronic<br />

unexplained diarrhea in Rio de Janeiro, Brazil: diagnosis,<br />

clinical presentation and follow-up. Rev Inst Med<br />

Trop Sao Paulo 1996; 38: 97–102.


126<br />

HIV and the intestine<br />

109. Hautvast JL, Tolboom JJ, Derks TJ et al. Asymptomatic<br />

intestinal microsporidiosis in a human immunodeficiency<br />

virus-seronegative, immunocompetent Zambian<br />

child. Pediatr Infect Dis J 1997; 16: 415–416.<br />

110. Schmidt W, Schneider T, Heise W et al. Mucosal abnormalities<br />

in microsporidiosis. AIDS 1997; 11: 1589–1594.<br />

111. van den Bergh Weerman MA, van Gool T, Eeftinck<br />

Schattenkerk JK, Dingemans KP. Electron microscopy as<br />

an essential technique for the identification of parasites<br />

in aids patients. Eur J Morphol 1993; 31: 107–110.<br />

112. Spira R, Lepage P, Msellati P et al. Natural history of<br />

human immunodeficiency virus type 1 infection in children:<br />

a five year prospective study in Rwanda.<br />

Pediatrics 1999; 104: 56.<br />

113. Horwood C, Liebeschuetz S, Blaauw D et al. Diagnosis<br />

of paediatric HIV infection in a primary health care<br />

setting using a clinical algorithm. Bull World Health<br />

Organ 2003; 81: 858–866.<br />

114. Coutsoudis A, Bobat RA, Coovadia HM et al. The effects<br />

of vitamin A supplementation on the morbidity of children<br />

born to HIV-infected women. Am J Public Health<br />

1995; 85: 1076–1081.<br />

115. Villamor E, Mbise R, Spiegelman D et al. Vitamin A<br />

supplements ameliorate the adverse effect of HIV-1,<br />

malaria, and diarrheal infections on child growth.<br />

Pediatrics 2002; 109: E6.<br />

116. Fawzi WW, Mbise R, Spiegelman D et al. Vitamin A<br />

supplements and diarrheal and respiratory tract infections<br />

among children in Dar es Salaam, Tanzania. J<br />

Pediatr 2000; 137: 660–667.<br />

117. Bahl R, Bhandari N, Saksena M et al. Efficacy of zincfortified<br />

oral rehydration solution in 6- to 35-month-old<br />

children with acute diarrhea. J Pediatr 2002; 141:<br />

677–682.<br />

118. Bhandari N, Bahl R, Taneja S et al. Substantial reduction<br />

in severe diarrheal morbidity by daily zinc supple-<br />

mentation in young north Indian children. Pediatrics<br />

2002; 109: E86.<br />

119. Faruque AS, Mahalanabis D, Haque SS et al. Doubleblind,<br />

randomized, controlled trial of zinc or vitamin A<br />

supplementation in young children with acute diarrhoea.<br />

Acta Paediatr 1999; 88: 154–160.<br />

120. Berhane R, Bagenda D, Marum L et al. Growth failure as<br />

a prognostic indicator of mortality in pediatric HIV<br />

infection. Pediatrics 1997; 100: E7.<br />

121. Arpadi SM, Cuff PA, Kotler DP et al. Growth velocity,<br />

fat-free mass and energy intake are inversely related to<br />

viral load in HIV-infected children. J Nutr 2000; 130:<br />

2498–2502.<br />

122. WHO Technical Advisory Group on Nutrition and<br />

HIV/AIDS. Nutrient Requirements for People Living with<br />

HIV/AIDS. Summary report of a Technical Consultation.<br />

Geneva, Switzerland: World Health Organisation, 2003.<br />

123. O’Keefe EA, Wood R, Van Zyl A, Cariem AK. Human<br />

immunodeficiency virus-related abdominal pain in<br />

South Africa. Aetiology, diagnosis and survival. Scand J<br />

Gastroenterol 1998; 33: 212–217.<br />

124. Borgstein ES, Broadhead RL. Acquired rectovaginal<br />

fistula. Arch Dis Child 1994; 71: 165–166.<br />

125. Oliver MJ. Spontaneously occurring rectovaginal fistulae<br />

in children and adults with HIV infection. East &<br />

Central Afr J Surg 1995; 1: 23–25.<br />

126. Wiersma R. HIV-positive African children with rectal<br />

fistulae. J Pediatr Surg 2003; 38: 62–64.<br />

127. Wang NC, Chang FY, Chou YY et al. Intussusception as<br />

the initial manifestation of AIDS associated with<br />

primary Kaposi’s sarcoma: a case report. J Formos Med<br />

Assoc 2002; 101: 585–587.<br />

128. Chadwick EG, Connor EJ, Hanson IC et al. Tumors of<br />

smooth-muscle origin in HIV-infected children. JAMA<br />

1990; 263: 3182–3184.


9<br />

Epidemiology and etiology<br />

Viral diarrhea<br />

Alfredo Guarino and Fabio Albano<br />

While infectious gastroenteritides are the second<br />

most common diseases in childhood worldwide,<br />

viruses are the most frequent agents of infectious<br />

diarrhea. Viral infections of the gastrointestinal<br />

tract cause 2 billion cases of diarrhea in children<br />

per year, resulting in 18 million hospitalizations<br />

and as many as 3 million deaths. 1<br />

Acute diarrhea is thus an enormous problem both<br />

in developing and in industrialized countries, but<br />

with two distinct consequences. In the former,<br />

enteric infections are extremely frequent, as the<br />

incidence of diarrhea is estimated at 3.8 episodes/<br />

child per year in infants and 2.1 episodes/child per<br />

year in children 1–4 years of age. 2 A cumulative<br />

incidence of 2.5–3 million deaths each year has<br />

been estimated, corresponding to 25% of all deaths<br />

in childhood up to 5 years of age. In the developing<br />

world, one out of 40 children will die because<br />

of diarrhea. 3<br />

In industrialized countries, on the other hand, the<br />

incidence of diarrhea is approximately 1–2<br />

episodes/child per year in subjects younger than 3<br />

years. The case/fatality ratio is far from the figures<br />

in poor countries, but not negligible. In the USA,<br />

150–300 infants or younger children die each year<br />

because of acute diarrhea, and substantial<br />

resources are needed for hospitalizations and<br />

medical visits. Diarrhea was associated with an<br />

annual average of 35 hospitalizations per 10000<br />

children younger than 5 years, corresponding to<br />

4% of all hospitalizations. 4 It was estimated that 1<br />

in 57 children would be hospitalized by 5 years of<br />

age for diarrhea-associated illness. The rate of outpatient<br />

visits was 943/10000 children, corresponding<br />

to 2% of all visits. Rotavirus is the leading<br />

agent of infectious diarrhea, being responsible for<br />

approximately 40% of all cases of diarrhea in the<br />

USA. 4 Similar figures have been recorded in<br />

Europe, 5 with rotavirus consistently playing a<br />

leading role in younger subjects. 6 The estimated<br />

average costs for an episode of diarrhea requiring<br />

hospital admission may be as high as US$ 2300.<br />

Costs for acute diarrhea are not negligible even in<br />

less severe cases, particularly when the so-called<br />

societal costs are considered. It has been estimated<br />

that the cost of an episode of diarrhea requiring an<br />

office visit in the USA averages approximately<br />

$300, 7 half of which is related to the loss of<br />

working days by the parents of sick children.<br />

During the past three decades, there has been a<br />

dramatic increase in the number of newly recognized<br />

etiological agents of gastroenteritis. Before<br />

1970, a pathogen could be identified in fewer than<br />

10% of patients hospitalized with diarrhea; the<br />

remaining 90% of cases represented a ‘diagnostic<br />

void’ consisting of various idiopathic, poorly<br />

defined conditions. Since 1970, more than 20<br />

different micro-organisms – bacteria, parasites and<br />

viruses – have been recognized as etiological<br />

agents, and most cases of gastroenteritis are now<br />

presumed to have an infectious etiology. Nevertheless,<br />

a pathogen is currently identified in only a<br />

small proportion of cases. Although numerous<br />

viruses have been identified in fecal samples of<br />

patients with diarrhea, causal relationships have<br />

been determined for relatively few (Table 9.1).<br />

Most children are infected with viruses belonging<br />

to four distinct families: rotaviruses, caliciviruses,<br />

astroviruses and enteric adenoviruses. Other<br />

viruses, such as the toroviruses, picornaviruses<br />

(the Aichi virus), and enterovirus 22, play a minor<br />

epidemiological role. Finally, selected viruses<br />

induce diarrhea only in children at risk. These<br />

include cytomegalovirus, Epstein–Barr virus and<br />

picobirnaviruses. Recent evidence suggests that<br />

127


128<br />

Viral diarrhea<br />

Table 9.1 Etiological role of viruses in childhood diarrhea<br />

Conclusively established Probable Possible in selected children<br />

Rotavirus Torovirus HIV<br />

Adenovirus Aichi virus Cytomegalovirus<br />

Astrovirus Enterovirus 22 Epstein–Barr virus<br />

Calicivirus Picobirnavirus<br />

HIV-1 virus may directly induce diarrhea through<br />

the production and release of TAT, its transactivating<br />

transfer factor. 8<br />

Enteric infections are usually associated with<br />

diarrhea and, less frequently, with vomiting.<br />

Diarrhea usually lasts a few days and resolves<br />

spontaneously without any major problem.<br />

However, in selected cases it may be severe, leading<br />

to dehydration, requiring hospital admission and<br />

possibly even resulting in fatal outcome.<br />

The impact of viral diarrhea is related to the<br />

specific strain, but occasionally also to the<br />

epidemiological setting, the host baseline features<br />

and the efficacy of early medical intervention<br />

(Table 9.2). Viral diarrhea may have a major impact<br />

in closed communities such as day-care centers<br />

and hospitals. It may be severe in malnourished or<br />

immunocompromized children, but also in children<br />

not belonging to groups at risk. Finally, diarrhea<br />

may become severe if rehydration is not initiated<br />

in the initial phases of the disease.<br />

Pathophysiology of viral diarrhea<br />

In the classic and simple view, the pathogenesis of<br />

diarrhea may be divided into osmotic and secretory<br />

(Figure 9.1). Viral diarrhea was originally<br />

believed to be caused by cell invasion and<br />

epithelial destruction by enteropathogenic agents,<br />

therefore being the result of endoluminal fluid<br />

accumulation osmotically driven by non-absorbed<br />

nutrients. It is now known that several mechanisms<br />

are responsible for diarrhea, depending on<br />

the specific agents and the host features. In<br />

addition, selected viruses possess multiple virulence<br />

pathways that act synergistically to induce<br />

diarrhea.<br />

The mechanisms of diarrhea induced by group A<br />

rotaviruses have been extensively investigated<br />

and provide a paradigm of the pathophysiology of<br />

viral diarrhea. Rotavirus has tissue- and cellspecific<br />

tropisms, infecting the mature enterocyte<br />

of the small intestine. The first step is virus<br />

binding to specific receptors located on the cell<br />

surface, the GM1 ganglioside. However, different<br />

rotavirus strains bind in either a sialic acid-dependent<br />

or an -independent fashion. 9 Most<br />

rotaviruses, including all human strains, infect<br />

polarized enterocytes through both the apical and<br />

the basolateral side, in a sialic acid-independent<br />

manner, suggesting the presence of different<br />

receptors. 10 The early stages of rotavirus binding<br />

involve the viral protein (VP)4 spike attachment<br />

and cleavage. This outer capsid protein contains<br />

ligand sequences for α2β1 and α4β1 integrins, and<br />

for complement receptor 4, located on the enterocyte<br />

surface. After binding, the rotavirus enters<br />

into the cell by a multistep process that requires<br />

both VP7 and VP4 proteins. The route of internalization<br />

remains controversial. Two mechanisms<br />

have been proposed: direct penetration through<br />

the cell membrane, which could be mediated by<br />

the VP5 cleavage product of VP4, or a receptorprimed<br />

calcium-dependent endocytosis.<br />

Infection of the villous enterocyte leads to cell<br />

lysis, compromising nutrient absorption and<br />

driving water into the intestinal lumen through an<br />

osmotic mechanism (Figure 9.1). However, the<br />

destruction of villus-tip cells induces a compensatory<br />

proliferation of crypt cells. These immature<br />

enterocytes physiologically maintain a secretive<br />

tone, thus contributing to diarrhea with ion secretion,<br />

as the result of the imbalance between<br />

absorptive villous and secretory crypt cells. Thus,<br />

the cytopathic action by rotavirus results in both<br />

osmotic and secretory diarrhea.


Pathophysiology of viral diarrhea 129<br />

Table 9.2 Epidemiological features and associated impact of viral diarrhea in specific settings<br />

Setting Features Impact Viruses<br />

Developing high frequency high mortality rotavirus<br />

countries<br />

Industrialized high frequency high costs all<br />

countries<br />

Day-care centers high frequency high costs for the society rotavirus + others<br />

Seasonal pattern major outbreaks winter/early spring rotavirus<br />

Food-related food poisoning massive attack rates calicivirus,<br />

transmission rotavirus,<br />

Aichi virus<br />

Age maximal incidence in increased severity in younger infants rotavirus<br />

children


130<br />

Viral diarrhea<br />

Figure 9.1 Diagram of osmotic and secretory mechanisms<br />

of viral diarrhea. The arrows indicate the water’s movements<br />

and their volumes. In osmotic diarrhea, water is driven into<br />

the intestinal lumen by the osmotic force of non-absorbed<br />

nutrients. In secretory diarrhea, ions are actively pumped<br />

within the lumen and are passively followed by water. From<br />

Guandalini S. Acute diarrhea. In Walzer WA et al. Pediatric<br />

Gastrointestinal Disease, 3rd edn. Hamilton, Ontario: BC<br />

Decker Inc. 2000, with permission.<br />

actin, the perturbation of cellular protein trafficking,<br />

the damage of tight-junctions, with disruption<br />

of cell–cell interaction and cytolysis. 16,17 This is<br />

reflected by the loss of epithelial integrity, as<br />

shown by the progressive decrease in tissue resistance<br />

measured in Caco-2 cell monolayers<br />

mounted in Ussing chambers 18 (Figure 9.2).<br />

In children with rotavirus infection, the onset of<br />

diarrhea is abrupt and occurs in the absence of<br />

histological changes, even if oral feeding is withdrawn,<br />

suggesting that a secretory pathway is<br />

responsible for diarrhea, at least in the initial<br />

phases of infection. A major advancement in the<br />

understanding of rotavirus pathophysiology came<br />

from the identification of the non-structural<br />

protein NSP4 as a viral enterotoxin, defined by its<br />

ability to cause fluid secretion, but not epithelial<br />

changes. 19<br />

NSP4 is a multifunctional virulence factor, as it<br />

possesses the following features (Figure 9.3):<br />

(1) It is released from infected cells; 20<br />

(2) It enters the cells through a specific receptor;<br />

21–24<br />

(3) It causes calcium-dependent chloride secretion,<br />

with an age-dependent pattern; 19<br />

Figure 9.2(a) Cytopathic effects induced by rotavirus (5<br />

PFU/cell) in a model based on the morphology of Caco-2<br />

cell monolayers. a, Uninfected Caco-2 cell monolayers; b,<br />

Caco-2 cells at 48 h post-infection: cellular vacuolization,<br />

opening of intracellular junction and spotting cell detachments<br />

are observed; c, Caco-2 cells at 96 h post-infection:<br />

extensive cellular detachment is observed with only a few<br />

picnotic cells yet present. From reference 18.


TEER (Ohm/cm 2 )<br />

800<br />

700<br />

600<br />

500<br />

400<br />

300<br />

200<br />

100<br />

0<br />

Virus-free control<br />

Virus 25 PFU/cell<br />

Virus 5 PFU/cell<br />

Virus 1 PFU/cell<br />

0 12 24 36 48<br />

Time (h)<br />

72 96<br />

Pathophysiology of viral diarrhea<br />

Figure 9.2(b) Cytopathic effects of rotavirus infection in a Caco-2 cell model, as measured by transepithelial electrical<br />

resistance (TEER). The decrease of TEER reflects progressive cell damage, which is shown in Figure 9.3. Increasing loads of<br />

virus induce an earlier and steeper fall of TEER, with a clear relationship with virus multiplicity. The TEER value decreases<br />

below a detectable level within approximately 36 h postinfection with 25 PFU/cell, 60–72 h post-infection with<br />

5 PFU/cell, and 96 h post-infection with 1 PFU/cell. From reference 18.<br />

Figure 9.3 Combined effects by NSP4 in the pathophysiology of rotavirus diarrhea. Rotavirus infects epithelial cells of the<br />

small intestine, replicates, and induces cell lysis. NSP4 is released by infected cells and functions as a Ca 2+-dependent<br />

enterotoxin triggering Cl - secretion. It decreases fluid and electrolyte transport by inhibiting Na–glucose symport SGLT1<br />

and, possibly Na–K ATPase. It also impairs disaccharidase expression. Furthermore, rotavirus and/or NSP4 may diffuse<br />

underneath the intestinal epithelium activating secretory reflexes in the enteric nervous system. Late during the infection,<br />

an inflammatory response in the lamina propria may be detected, and the production of inflammatory substances and<br />

cytokines may further contribute to the increase of intestinal permeability and diarrhea.<br />

131


132<br />

Viral diarrhea<br />

(4) It alters plasma membrane permeability and<br />

is cytotoxic; 25–27<br />

(5) It is sensitive to specific antibody, which<br />

prevents or reduces diarrhea; 28,29<br />

NSP4 is the only rotavirus gene product capable of<br />

eliciting intracellular calcium mobilization. 30<br />

NSP4 was demonstrated to stimulate a calciumdependent<br />

chloride secretion, in mouse small<br />

intestinal mucosa sheets mounted in Ussing chambers,<br />

suggesting that this enterotoxin triggers diarrhea<br />

in the early phase of infection in animal<br />

models. 14,31 NSP4 further contributes to diarrheal<br />

pathogenesis by directly altering enterocyte actin<br />

distribution and paracellular permeability. 32<br />

Finally, NSP4 plays a role in the inhibition of the<br />

Na + -dependent glucose transporter SGLT-1. 33<br />

Glucose absorption is impaired in rotavirus diarrhea<br />

as well as disaccharidase activities, whereas<br />

the Na/amino acid co-transporters are not<br />

involved.<br />

Rotavirus diarrhea may also have an inflammatory<br />

component. The induction of cytokines is important<br />

in developing an inflammatory and immune<br />

response, especially in intestinal infection caused<br />

by bacteria. In rotavirus infection, limited inflammation<br />

is detected by histological studies, suggest-<br />

Isc (µA)<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 1 2 3 6 12 24 36<br />

Hours after infection<br />

ing that cytokines are effective in inducing a host<br />

immune response to rotavirus diarrhea. However,<br />

it has been shown that the rotavirus-infected enterocyte<br />

activates NF-κB and the production of<br />

chemokines interleukin (IL)-8, Rantes and GRO-a,<br />

and of cytokines interferon (IFN)α and granulocyte/macrophage–colony-stimulating<br />

factor (GM-<br />

CSF). 34,35<br />

In conclusion, the primary target of rotavirus is the<br />

enterocyte, which is induced to secrete fluids and<br />

is subsequently destroyed. On the other hand, the<br />

enterocyte acts as a sensor to the mucosa with the<br />

production of viral and endogenous factors and<br />

the activation of other cell types including nerves.<br />

Thus, rotavirus-induced diarrhea is a multistep<br />

and multifactorial event, in which fluid secretion<br />

and cell damage are observed in sequence, as<br />

shown in an intestinal cell line-based experimental<br />

model (Figure 9.4). A summary of the multiple<br />

mechanisms involved in the rotavirus–intestine<br />

interaction is given in Table 9.3.<br />

Clinical signs and symptoms<br />

The predominant symptoms of vomiting and diarrhea<br />

are common to enteric infections, regardless<br />

of the causative role of more than 20 different<br />

Figure 9.4 Biphasic effect of rotavirus in Caco-2 cells. Rotavirus induces a biphasic response, in an in vitro model of<br />

infection in Caco-2 enterocytes mounted in Ussing chambers. An early secretion is evident in the first few hours of infection,<br />

with a peak at 2h post-infection, as shown by the increase in short circuit current (Isc, - -). Subsequently, rotavirus<br />

exerts a cytotoxic effect with a loss of tissue integrity, as demonstrated by the fall of transepithelial resistance<br />

(Ohm/cm 2 , - -) which is evident at 24h post-infection. The results suggest that rotavirus diarrhea is initially the result of an<br />

early secretory mechanism and of a subsequent osmotic pathway, due to cell damage and loss of functional absorptive<br />

surface, leading to nutrient malabsorption. From G. De Marco et al, unpublished data.<br />

600<br />

400<br />

200<br />

0<br />

Resistance (Ohm/cm 2)


Table 9.3 Pathogenesis of rotavirus diarrhea<br />

Pathophysiology of viral diarrhea<br />

Key process Pathway Consequences<br />

Villous cell destruction cytoskeleton disruption, cell lysis nutrient<br />

malabsorption,<br />

osmotic diarrhea<br />

Crypt cell proliferation compensatory secretory cell proliferation secretory diarrhea<br />

NSP4 enterotoxin increase in intracellular calcium, secretory diarrhea<br />

chloride secretion<br />

NSP4-induced inhibition of SGLT-1 osmotic diarrhea<br />

glucose malabsorption<br />

Neuromediated neurotransmitter microcirculation secretory diarrhea<br />

vascular ischemia impairment<br />

Inflammation NF-κB, IL-8, Rantes osmotic/secretory diarrhea<br />

microbial agents, including bacteria, parasites and<br />

viruses.<br />

Usually, viral diarrhea lasts for 3–5 days. In<br />

selected cases, viral diarrhea may be persistent<br />

and even become life-threatening. In a series of<br />

children with intractable diarrhea syndrome,<br />

rotavirus was detected in five out of 38 children<br />

who had no evident risk factors. 36<br />

The history may provide valuable clinical information.<br />

Diarrheal onset may be abrupt or<br />

progressive. The child’s age, admission to a daycare<br />

center, the time of year, exposure to diarrheal<br />

contacts, previous antibiotic courses, and ingestion<br />

of contaminated food such as eggs or water<br />

should be evaluated to define the origin of diarrhea.<br />

Risk factors for severe diarrhea include<br />

malnutrition, immune derangement and AIDS,<br />

and history of repeated episodes of diarrhea.<br />

Recent weaning from breast milk or introduction<br />

of feedings other than milk may be associated with<br />

a more severe course of the disease (Table 9.2).<br />

Dehydration is the key symptom to define the<br />

severity of the disease and the need for fluid<br />

replacement. The degree of dehydration should be<br />

evaluated at first observation and followed up to<br />

evaluate the ongoing losses and the efficacy of<br />

fluid intake to replace them. The gold standard for<br />

133<br />

determining dehydration is acute weight loss.<br />

Because a patient’s pre-illness weight is rarely<br />

known, an estimate of fluid deficiency is made on<br />

clinical grounds.<br />

Vomiting may be associated with diarrhea and<br />

induce additional fluid losses. In addition, vomiting<br />

may prevent oral rehydration, thus requiring<br />

parenteral fluid replacement.<br />

Body temperature may be elevated, as a consequence<br />

of dehydration or reflecting an inflammatory<br />

response. Abdominal pain is not frequent and<br />

suggests colonic involvement, but may also<br />

indicate a surgical problem.<br />

Stool characteristics should be carefully considered:<br />

large volumes of watery stools indicate<br />

small bowel involvement and are frequently<br />

associated with dehydration, whereas frequent<br />

outputs of a small amount of mucus or bloody<br />

stools are associated with colitis. In severe cases<br />

the entire intestine is involved.<br />

Colonic involvement is more often associated with<br />

bacterial rather than viral etiology. Some features<br />

may help in distinguishing viral from bacterial<br />

diarrhea (Table 9.4). 37 Viral diarrhea is more<br />

frequently associated with vomiting and dehydration.<br />

It affects younger children compared to


134<br />

Viral diarrhea<br />

Table 9.4 Main clinical features associated with the most frequent enteric pathogens (modified from<br />

reference 37)<br />

bacterial diarrhea. Selected features are more<br />

frequently associated with specific enteric viruses<br />

(Table 9.5). 38 However, signs and symptoms in the<br />

individual child do not reliably allow identification<br />

of specific etiological agents of gastroenteritis.<br />

Salmonella Campylobacter Giardia<br />

species species Rotavirus NLV lamblia<br />

Fever ++++* ++++* ++++* + +<br />

Blood in stool ++ ++++* + ++ +++<br />

Abdominal cramps ++++* ++ -* + +<br />

Vomiting + + ++++ * ++++* +<br />

>6 stools per day + ++++* ++ + +<br />

Duration of 7 7


disease. 39 Acute diarrhea may be a side-effect of<br />

various drugs including antibiotics. History and<br />

clinical evaluation may aid in the differential diagnosis.<br />

However, close follow-up is required.<br />

Generally, microbiological investigations are not<br />

necessary in children with acute gastroenteritis.<br />

Several studies have reported a low yield and<br />

cost/efficacy ratio per identified agent even in<br />

hospitalized children. Microbiological examination<br />

should be considered: in cases of persistent<br />

diarrhea; when a specific antimicrobial treatment<br />

is considered, such as for children belonging to a<br />

group at risk; when an intestinal infection must be<br />

excluded in order to support a different etiology;<br />

and to investigate an outbreak.<br />

The search for enteric viruses is made by several<br />

techniques, including culture in sensitive cells,<br />

electron microscopy, immune-based assays and<br />

molecular probes (Table 9.6). Virus culture is the<br />

gold standard, but it is cumbersome and the<br />

results are available only with delay, limiting its<br />

clinical applications. Immune-based methods are<br />

widely used, but a progressive increase in the use<br />

of polymerase chain reaction (PCR) techniques is<br />

leading to a shift of diagnostic techniques. The<br />

choice of a specific technique is based on the clin-<br />

Features of specific etiologies and virology<br />

135<br />

ical need, the availability of technical skills and<br />

the efficacy ratio. In most clinical institutions,<br />

immune-based assays are available to detect<br />

rotavirus and less frequently enteric adenovirus.<br />

The search for other viruses is generally available<br />

in reference centers or research institutions.<br />

However, trying to identify the etiology may not be<br />

clinically useful, as treatment of diarrhea is relatively<br />

independent of the responsible agent.<br />

Rather, it is the evaluation of the child’s clinical<br />

condition that provides information for case<br />

management.<br />

Features of specific etiologies and<br />

virology<br />

Rotaviruses<br />

Table 9.6 Etiological diagnosis of viral infections of the gastrointestinal tract<br />

Reovirus-like particles were first identified in 1973<br />

by electron microscopy in duodenal biopsies from<br />

children with acute diarrhea. 40 This led to a<br />

cascade of clinical and laboratory studies that have<br />

established the rotavirus as the single most<br />

common agent causing diarrhea in childhood.<br />

Viruses Antigen detection EM PCR Preferred test method<br />

Rotavirus EIA, latex agglutination ++++ RT-PCR EIA<br />

Calicivirus EIA + RT-PCR RT-PCR<br />

Astrovirus EIA + RT-PCR EIA<br />

RT-PCR<br />

Adenovirus EIA +++ RT-PCR EIA<br />

Cytomegalovirus PCR histology,<br />

immunohistochemistry<br />

Picornavirus EIA RT-PCR EIA<br />

Epstein–Barr virus PCR histology,<br />

immunohistochemistry<br />

EM, electron microscopy; PCR, polymerase chain reaction; EIA, enzyme immunoassay; RT, reverse transcriptase;<br />

+, detectable with low sensitivity by a skilled microscopist; +++, visible; ++++, easily visible


136<br />

Viral diarrhea<br />

The global illness and deaths caused by rotavirus<br />

in children were recently estimated by reviewing<br />

studies between 1985 and 2000. Rotavirus caused<br />

111 million episodes of gastroenteritis requiring<br />

home treatment, 25 million clinic visits, 2 million<br />

hospitalizations and 350–600000 deaths in children<br />

less than 5 years of age, worldwide. All children<br />

were expected to be infected with rotavirus<br />

within 5 years of age, one in five children were<br />

expected to need a clinic visit and one in 65 to be<br />

hospitalized. Finally, one out of 293 children<br />

would die because of rotavirus infection, 84% in<br />

the poorest countries. 41<br />

Also, in countries with high economic standards,<br />

rotaviruses are a major problem. In industrialized<br />

countries, the estimates were: a total of 7122000<br />

episodes of rotavirus gastroenteritis requiring only<br />

home care in children less than 5 years of age, a<br />

total of approximately 1781000 clinic visits and a<br />

total of 223000 rotavirus-associated hospitalizations.<br />

41 There are an estimated 3.5 million cases<br />

annually among children less than 5 years of age<br />

in the USA, leading to 500000 office visits, 50000<br />

hospitalizations, and approximately 20 deaths. 42<br />

These numbers translate into costs, resulting in<br />

US$ 1 billion/ year. 43<br />

Infection is widespread throughout the world.<br />

Rotavirus infection may occur repeatedly in<br />

humans from birth to old age. The first infection is<br />

predominant among children aged 6–24 months,<br />

although cases may occur in older children.<br />

<strong>Neonatal</strong> infections appear to be nosocomial in<br />

origin, because they are rarely seen in babies born<br />

at home or at village health centers.<br />

Approximately 90% of children in both developed<br />

and developing countries experience rotavirus<br />

infection by 3 years of age. The cumulative incidence<br />

of rotavirus illness by the age of 5 years<br />

approaches 0.8 episodes/child per year.<br />

Several factors are responsible for the high spreading<br />

of rotavirus. Rotavirus shedding averages 6<br />

days per episode, but may be persistent.<br />

Asymptomatic carriers contribute to infection<br />

spreading. Infected children develop a protective<br />

immunity and, although repeated infections generally<br />

occur in growing children, their clinical severity<br />

decreases. 44 Despite differences among studies<br />

in geographical areas, years and age groups, an<br />

increase in rotavirus cases is consistently reported<br />

in the winter months, with a peak in February to<br />

April. In tropical areas, rotaviruses are identified<br />

throughout the year.<br />

Transmission occurs through the fecal–oral route.<br />

However, droplet transmission has been suggested<br />

to explain the rapidity with which the rotavirus<br />

can spread through a community. 45<br />

Virology<br />

Rotavirus is a double-stranded RNA virus belonging<br />

to the Reoviridae family. The virion, 70–75 nm,<br />

is composed of a three-layered protein capsid that<br />

encloses 11 distinct segments of genomic RNA,<br />

each coding for a different capsid or non-structural<br />

protein. The internal core contains viral proteins<br />

(VP) 1, 2 and 3; the inner capsid contains VP4; the<br />

two outer capsid proteins encoded by genes 4 and<br />

7, namely VP4 and VP7, represent the only established<br />

neutralization antigens of the virus. The<br />

protective role of antibodies directed at these<br />

proteins has been confirmed both in experimental<br />

animal models and in humans. A possible role has<br />

been suggested for antibodies directed at the inner<br />

capsid protein VP6, which is not associated with<br />

in vitro neutralization. The non-structural proteins<br />

NSP1, NSP2 and NSP4 are virulence factors in<br />

mice.<br />

Rotavirus groups A–F have been described, but<br />

only groups A, B and C have been identified in<br />

humans. Most human infections are caused by<br />

group A rotaviruses that are classified into<br />

serotypes by a dual classification system based on<br />

neutralizing antigens on two outer capsid proteins,<br />

VP7 (G serotype) and VP4 (P serotype). To date, 10<br />

G types and almost as many P types have been<br />

identified in infected humans. There is great<br />

genetic diversity within each G and P type, as<br />

shown by gel electrophoretic analysis of gene<br />

patterns (electropherotypes). Epidemiological and<br />

molecular studies in many countries show<br />

complex changes from year to year in the serotypes<br />

and electropherotypes that cause diarrhea in children<br />

from the same geographical areas. 46,47 The<br />

majority of severe diseases have been caused by<br />

serotypes G1 to G4, P1A and P1B, worldwide. 48<br />

Epidemiological studies in Bangladesh, 49 Brazil, 50<br />

India, 51 Kenya 48 and the USA 52 show that other G<br />

and P types (G5 to G10, P2A and P8) can be


common and may be of emerging importance in<br />

communities. 53 Specific strains may express<br />

stronger virulence factors, which could be related<br />

to the severity of symptoms. More severe diseases<br />

may also be related to the reintroduction of strains<br />

in areas where they have been previously absent. 54<br />

Astroviruses<br />

Human astroviruses (HAstVs) were first identified<br />

in 1975 in an outbreak of diarrhea in infants, and<br />

were named astrovirus because of the distinctive<br />

five- or six-pointed stars seen at electron<br />

microscopy. 55<br />

Recent studies have established that astrovirus is<br />

the third most frequent cause of diarrhea in children<br />

and the second in selected settings. The<br />

reported infection rates depend on detection<br />

methods. These include electron microscopy (EM),<br />

enzyme immunoassay (EIA) or reverse transcriptase-polymerase<br />

chain reaction (RT-PCR). 56<br />

Incidence rates also depend on the population<br />

under study. The reported incidence of HAstV<br />

diarrhea ranges from 2% of children seeking<br />

medical care in Baltimore, to 17% of children with<br />

persistent diarrhea in Bangladesh. 57 Younger<br />

infants are at greater risk of developing diarrhea<br />

than older children. In a child-care center, attack<br />

rates among infants and toddlers ranged between<br />

11 and 89%. 58<br />

HAstV may be an important agent of diarrhea in<br />

immunocompromised hosts such as those infected<br />

with HIV 59 and bone marrow transplanted<br />

patients. 60,61 Current evidence supports foodborne,<br />

water-borne and person-to-person transmission.<br />

The incidence peaks in winter months in<br />

temperate climates.<br />

Virology<br />

HAsVs are non-enveloped, single-stranded RNA<br />

viruses with a distinctive star appearance and a<br />

smooth particle edge. The genomic organization is<br />

unique among positive stranded RNA and<br />

warrants classification of astroviruses as a separate<br />

family, the Astroviridae. 62 Eight antigenic types<br />

have been identified. The complete genomic<br />

sequence of types 1 and 2 and the sequence of the<br />

Features of specific etiologies and virology<br />

137<br />

capsid gene of types 3, 4, 5, 6, 7 and 8 have been<br />

obtained. 63<br />

HAstV-1 is the dominant genotype followed by<br />

HAstV-2. It is not known whether infection with<br />

one serotype confers protection against subsequent<br />

infection with other serotypes.<br />

Caliciviruses<br />

In 1972, the ‘Norwalk agent’ was discovered in<br />

fecal specimens during a gastroenteritis outbreak<br />

in an elementary school in Norwalk, Ohio. 64,65<br />

This was the first discovered viral agent of<br />

gastroenteritis in humans using EM. Subsequently,<br />

a large number of other small round structured<br />

viruses (SRSVs) were detected. These agents<br />

appeared morphologically indistinguishable from<br />

the Norwalk virus by EM, but they were antigenically<br />

distinct by immune EM. SRSVs were discovered<br />

during investigations of gastroenteritis<br />

outbreaks and were named for the investigation<br />

site (e.g. Bristol, Hawaii, Snow Mountains, etc.). In<br />

addition, a morphologically distinct SRSV, defined<br />

as ‘classic human calicivirus’, was first described<br />

in the UK, 66 and the prototype strain – the Sapporo<br />

agent – was subsequently identified in Japan. 67<br />

This rather confusing classification system of<br />

these viruses has been recently revised and is now<br />

based on genetic sequences of the viruses and of<br />

their genomic organization. All viruses belong to<br />

the family Caliciviridae, and fall into two provisionally<br />

named genera: ‘Norwalk-like viruses’<br />

(NLV) and ‘Sapporo-like viruses’ (SLV).<br />

Molecular epidemiological studies showed that<br />

caliciviruses are the main cause of non-bacterial<br />

gastroenteritis outbreaks causing over 90% of<br />

outbreaks of acute, non-bacterial gastroenteritis in<br />

the USA. 68 They are spread by the fecal–oral route,<br />

but outbreaks are often caused by contaminated<br />

food or water. 69 Contaminated surfaces carry calicivirus<br />

in detectable amounts, thereby providing<br />

another important substrate for explosive<br />

outbreaks. 70 Droplets or person-to-person transmission<br />

explain outbreaks in which other spreading<br />

routes cannot be identified. 71 Infections rates<br />

increase in cold seasons, 72 almost disappearing in<br />

the warm summer months. NLVs have been found<br />

in 5–20% of stool specimens from sporadic cases


138<br />

Viral diarrhea<br />

of diarrhea using RT-PCR. 73 SLVs cause predominantly<br />

diarrheal diseases, while NLVs cause<br />

‘winter vomiting disease’ in young children. 74<br />

Virology<br />

Caliciviruses are single-stranded, positive sense<br />

RNA viruses closely related to picornaviruses.<br />

Based on morphology, typical and atypical caliciviruses<br />

have been described. SLVs have the<br />

typical calicivirus morphology, with a six-pointed<br />

star appearance similar to those of many animal<br />

caliciviruses. In contrast, the surface structure of<br />

NLVs is rather smooth, leading to the designation<br />

‘small round structured viruses’.<br />

The family Caliciviridae encompasses four distinct<br />

virus genera. Two, the NLVs and SLVs, contain the<br />

human calicivirus (previously referred to as small<br />

round structured viruses) and the classic human<br />

caliciviruses, respectively. Phylogenetic analyses<br />

of sequences in the RNA polymerase and the<br />

capsid regions of the genome revealed that<br />

currently identified NLVs and SLVs can be divided<br />

into genogroups and genetic clusters. The NLV<br />

genus includes two genogroups (I and II) and at<br />

least 15 genetic clusters. 75 The SLV genus has<br />

fewer members, with a total of 4–5 genetic clusters.<br />

Enteric adenoviruses<br />

There are six different groups of adenovirus, but<br />

only group F, including serotypes 40 and 41, are<br />

referred to as enteric adenoviruses. Both cause<br />

endemic diarrhea and outbreaks in hospitals,<br />

orphanages and day-care centers.<br />

The enteric adenoviruses are non-enveloped particles,<br />

containing double-stranded DNA. 76<br />

These viruses infect children more than adults;<br />

more than 50% of children are seropositive by<br />

years 3–4 of life. 77 Enteric adenoviruses are<br />

detected in 1.5–4% of children with diarrhea and<br />

are isolated throughout the year with no specific<br />

seasonal distribution. 78,79 Transmission is fecal–<br />

oral. The incubation lasts from 3 to 10 days.<br />

Adenovirus diarrhea lasts for 6–9 days and may be<br />

associated with vomiting and fever. Stools are<br />

watery, without blood or fecal leukocytes. EM<br />

initially was used to detect enteric adenoviruses,<br />

but commercial enzyme-linked immunosorbent<br />

assays are now widely available.<br />

Toroviruses<br />

Toroviruses include the Breda virus of cattle and<br />

Berne virus of horses, 80–82 and were first documented<br />

in 1984 by EM in humans with gastroenteritis.<br />

83 These enveloped RNA viruses contain a<br />

tightly coiled tubular nucleocapsid that generally<br />

assumes a ‘donut’ torus shape in the virion. 80<br />

Toroviruses were classified on the basis of the<br />

Berne virus genome sequence as members of the<br />

family Coronaviridae, which together with the<br />

family Arteriviridae are now classified in the order<br />

Nidovirales. 84–86<br />

Study of torovirus infections in calves indicates<br />

that Breda viruses infect differentiating crypt cells,<br />

expecially in the large intestine. 81,82<br />

The incidence of torovirus infection remains relatively<br />

constant throughout the year. Early studies<br />

indicated that toroviruses infect school-age children,<br />

and are responsible for nosocomial infections,<br />

more frequently in immunocompromised<br />

patients. 87<br />

The clinical manifestations of torovirus infection<br />

are similar to those of rotavirus or astrovirus,<br />

except that children with torovirus infection had<br />

less vomiting and bloodier diarrhea. 83,87 Symptom<br />

duration is similar to that of rotavirus infection,<br />

but toroviruses more frequently induce persistent<br />

diarrhea. 87 Further epidemiological studies are<br />

needed to determine its frequency in the<br />

community.<br />

Picornaviruses (the Aichi virus)<br />

In 1989, a cytopathic small round virus was<br />

isolated from a patient with oyster-associated<br />

gastroenteritis. 88 Genetic analyses of this virus,<br />

named Aichi virus, led to its classification into the<br />

Picornavirus family. It is distinct from any other<br />

genus such as the enterrhino-, cardio-, aptho-,<br />

hepato- and parechovirus group. 89 The Aichi virus<br />

is presently an unassigned species in the<br />

Picornavirus family. 90 Recently, it has been


proposed to assign the Aichi virus to a new genus<br />

named Kobuvirus. 91<br />

The Aichi viruses cause acute gastroenteritis<br />

outbreaks. The main clinical symptoms are diarrhea<br />

(59%), abdominal pain (83%), nausea (92%),<br />

vomiting (71%) and fever (58%). 92<br />

Coronaviruses<br />

Coronaviruses are large enveloped single-stranded<br />

RNA viruses related to toroviruses. These viruses<br />

are a well-documented cause of gastroenteritis in<br />

animals and of the common cold in humans.<br />

Coronaviruses have been identified in the stools of<br />

children with diarrhea, but their role as a cause of<br />

diarrhea is unknown. They have been detected<br />

more commonly in the diarrheal stools of older<br />

children and young adults. The prolonged virus<br />

excretion makes it difficult to assess their etiological<br />

role. Recently, coronavirus mutants have been<br />

implicated in the severe acute respiratory<br />

syndrome (SARS). 93<br />

Picobirnaviruses<br />

These viruses have icosahedral symmetry with<br />

triangulation number (T) equal to 3. 94 They have<br />

been detected in diarrheic as well as non-diarrheic<br />

animals, and occasionally in children and in<br />

humans with HIV infection. Their etiological role<br />

in immunocompetent children is unknown.<br />

Treatment<br />

For the cornerstone of treatment of acute diarrhea,<br />

i.e. the restoration of fluid and electrolyte homeostasis<br />

95,96 see Chapter 39.<br />

For children hospitalized with severe rotavirus<br />

diarrhea, passive immune treatment should be<br />

considered. Several studies have shown that oral<br />

administration of human serum immunoglobulin<br />

is associated with an effective antiviral effect.<br />

Human immunoglobulin, including pooled<br />

gammaglobulin, bovine colostrums, or human<br />

milk, may decrease the frequency and duration of<br />

diarrhea. 97 Human immunoglobulin was initially<br />

used to treat children with life-threatening<br />

Treatment<br />

139<br />

rotavirus diarrhea. 98 In a double-blind, placebocontrolled<br />

study oral administration of human<br />

serum immunoglobulin, in a single bolus of<br />

300 mg/kg body weight, resulted in faster recovery<br />

and early discharge of immunocompetent children<br />

admitted to hospital with severe rotavirus diarrhea,<br />

without adverse effects 99 (Figure 9.5). The<br />

same treatment was given to children with AIDS<br />

and severe rotavirus infection, and led to rapid<br />

remission of diarrhea and permanent clearance of<br />

the virus. 100 We currently give human immunoglobulin<br />

to children hospitalized with severe<br />

rotavirus infection and to patients at risk of a poor<br />

outcome. 101<br />

Human immunoglobulin, although expensive, is<br />

widely available. Its efficacy might be related to<br />

high titers of specific neutralizing antibodies to<br />

rotavirus. Specific titers are consistently detected<br />

in preparations commercially available for intravenous<br />

use, because of the high frequency of<br />

rotavirus infection and the consequent widespread<br />

immune response. The efficacy of immunoglobulin<br />

is probably related to dose- and time-related<br />

direct neutralization of rotavirus, which prevents<br />

enterocyte infection and cell death. 18 Preliminary<br />

experimental evidence indicates that immunoglobulin<br />

prevents both the cytotoxic and the enterotoxic<br />

effect induced by rotavirus in human enterocytes.<br />

102 Thus, immunoglobulin should be<br />

administered orally early in the course of the<br />

disease for maximum efficacy.<br />

This information may be of practical relevance,<br />

while awaiting the development of safer vaccines<br />

that will hopefully be effective, for normal or<br />

compromised immune function.<br />

Vaccine development<br />

Rotavirus<br />

A review of the global prevalence of rotavirus<br />

disease was published in 1985, showing that<br />

rotavirus accounted for 6% of diarrhea episodes<br />

and 20% of deaths caused by diarrhea in children<br />

less than 5 years of age in developing countries. 103<br />

The incidence of rotavirus disease was similar in<br />

both industrialized and developing countries,<br />

suggesting that adequate control may not be<br />

achieved by improvements in water supply,


140<br />

Stool output/24 h<br />

Viral diarrhea<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 1 2 3 4 5<br />

Days<br />

Figure 9.5 Stool frequency (stools/24 h) in children hospitalized because of Rotavirus treated with human serum<br />

immunoglobulin administered per oral route in a single dose of 300 mg/kg body weight (•) and control children (o). Values<br />

are means ± SEM. *p


though a rotavirus vaccine was licensed, correlates<br />

of protective immunity remain relatively unclear.<br />

‘Norwalk-like viruses’<br />

Some human study volunteers never become<br />

infected after challenge with NLV, perhaps reflecting<br />

the recent finding that NLVs recognize human<br />

histoblood group antigens as receptors, defining<br />

host susceptibility. Immunity to NLV infections is<br />

believed to be short lived, as volunteers were reinfected<br />

with the same strain within months of<br />

previous challenge.<br />

Conclusions/summary<br />

Table 9.7 Live, attenuated, oral rotavirus vaccines currently in human trials (modified from<br />

reference 105)<br />

Product Company Concept<br />

LLR Lanzhou Institute of Biological monovalent lamb strain (P[12]G10)<br />

Products (China)<br />

Rotateq Merck (USA) WC-3 based multivalent human-bovine<br />

reassortant<br />

Rotarix (89-12) GlaxoSmithKline (Belgium) monovalent human strain (P[8]G1)<br />

UK-reassortant vaccine Wyeth Ayerst/NIH (USA) UK-based multivalent human–bovine<br />

reassortant<br />

RV3 University of Melbourne (Australia) neonatal strain (P[6]G3)<br />

116E Bharat Biotech (India) neonatal strain (P[11]G9)<br />

I321 Bharat Biotech (India) neonatal strain (P[11]G10)<br />

Self-assembled recombinant capsid proteins of<br />

Norwalk virus (rNV) into VLPs have been tested as<br />

oral immunogens. 106 Preclinical studies in mice<br />

and phase I studies in healthy adult volunteers<br />

have shown that these VLPs are immunogenic in<br />

the absence of adjuvant, and high doses (250mg)<br />

delivered orally were safe and elicited antibody<br />

responses in all volunteers. The positive results<br />

obtained with oral delivery of rNV VLPs have stimulated<br />

new vaccine developments, including the<br />

expression of rNV in tobacco and potato tubers. 107<br />

RNV VLPs expressed in potato tubers were also<br />

immunogenic when given to volunteers orally. 108<br />

The protective efficacy of these candidate vaccines<br />

in human volunteers is currently under study.<br />

Conclusions/summary<br />

141<br />

Viral diarrhea still represents a major threat to<br />

childhood health, worldwide. It has an immense,<br />

but distinct, impact in poor and rich countries,<br />

being responsible for a substantial number of<br />

deaths in the former and of heavy money loss in<br />

the latter. Rotavirus is the leading agent and its<br />

pathophysiology involves multiple mechanisms,<br />

several of which are triggered by NSP4 enterotoxin.<br />

Diarrhea is the hallmark of viral diarrhea<br />

and is usually self-limiting. However, it may run a<br />

severe course and have a fatal outcome in children<br />

with malnutrition or immune impairment and in<br />

those who have no access to rehydration.<br />

Diagnosis is usually based on clinical grounds, and<br />

investigations are not necessary. Treatment is<br />

based on carbohydrate and electrolyte solution<br />

administered through the oral or parenteral route.<br />

Selected probiotics may be effective in reducing<br />

the duration of symptoms, whereas, in severe<br />

cases in which rotavirus is involved, oral administration<br />

of human serum immunoglobulin may be<br />

effective. Efforts towards vaccine development<br />

have been hampered by withdrawal of antirotavirus<br />

vaccine, because of its association with<br />

intussusception, but novel vaccines are currently<br />

under investigation.


142<br />

Viral diarrhea<br />

REFERENCES<br />

1. Taterka JA, Cuff CF, Rubin DH. Viral gastrointestinal<br />

infections. Gastroenterol Clin North Am 1992; 21:<br />

303–330.<br />

2. Kosek M, Bern C, Guerrant RL. The magnitude of the<br />

global burden of diarrhoeal disease, as estimated from<br />

studies published between 1992 and 2000. Bull World<br />

Health Organ 2003; 81:197–204.<br />

3. Glass RI, Bresee J, Jiang B et al. Gastroenteritis viruses:<br />

an overview. Novartis Found Symp 2001; 238: 5–25.<br />

4. Zimmermann CM, Bresee JS, Parashar UD et al. Cost of<br />

diarrhea-associated hospitalizations and outpatient<br />

visits in an insured population of young children in the<br />

United States. Pediatr Infect Dis J 2001; 20: 14–19.<br />

5. Fruhwirth M, Karmaus W, Moll-Schuler I et al. A<br />

prospective evaluation of community acquired gastroenteritis<br />

in paediatric practices: impact and disease<br />

burden of rotavirus infection. Arch Dis Child 2001; 84:<br />

393–397.<br />

6. de Wit MAS, Koopmans MPG, van der Blij JF et al.<br />

Hospital admission for rotavirus in the Netherlands.<br />

Clin Infect Dis 2000; 31: 698–704.<br />

7. Avendano P, Matson DO, Long J et al. Costs associated<br />

with office visits for diarrhea in infants and toddlers.<br />

Pediatr Infect Dis J 1993; 12: 897–902.<br />

8. Berni Canani R, Cirillo P, Mallardo G et al. Effects of<br />

HIV-1 Tat protein on ion secretion and on cell proliferation<br />

in human intestinal epithelial cells.<br />

Gastroenterology 2003; 124: 368–376.<br />

9. Ciarlet M, Estes MK. Human and most animal rotavirus<br />

strains do not require the presence of sialic acid on the<br />

cell surface for efficient infectivity. J Gen Virol 1999; 80:<br />

943–948.<br />

10. Ciarlet M, Crawford SE, Estes MK. Asymmetric infection<br />

of epithelial cell lines of sialic-acid-dependent<br />

rotaviruses. Gastroenterology 2000; 118: A101.<br />

11. Reynolds D, Hall G, Debney T et al. Pathology of natural<br />

rotavirus infection in clinically normal calves. Res Vet<br />

Sci 1985; 38: 264–269.<br />

12. Collins J, Benfield D, Duimstra J. Comparative virulence<br />

of two porcine group A rotavirus isolates in gnotobiotic<br />

pigs. Am J Vet Res 1989; 50: 827–835.<br />

13. Osborne MP, Haddon SJ, Worton KJ et al. Rotavirusinduced<br />

changes in microcirculation of intestinal villi<br />

of neonatal mice in relation to the induction and persistence<br />

of diarrhea. J Pediatr Gastroenterol Nutr 1991; 12:<br />

111–120.<br />

14. Lundgren O, Timar Peregrin A, Persson K et al. Role of<br />

the enteric nervous system in the fluid and electrolyte<br />

secretion of rotavirus diarrhea. Science 2000; 287:<br />

491–495.<br />

15. Ruiz MC, Cohen J, Michelangeli F. Role of Ca 2+ in the<br />

replication and pathogenesis of rotavirus and other viral<br />

infections. Cell Calcium 2000; 28: 137–149.<br />

16. Obert G, Peiffer I, Servin AL. Rotavirus-induced structural<br />

and functional alterations in tight junctions of<br />

polarized intestinal Caco-2 cell monolayers. J Virol<br />

2000; 74: 4645–4651.<br />

17. Brunet JP, Cotte-Laffitte J, Linxe C et al. Rotavirus infection<br />

induces an increase in intracellular calcium<br />

concentration in human intestinal epithelial cells: role<br />

in microvillar actin alteration. J Virol 2000; 74:<br />

2323–2332.<br />

18. Guarino A, Casola A, Bruzzese E et al. Human serum<br />

immunoglobulin counteracts rotaviral infection in Caco-<br />

2 cells. Pediatr Res 1996; 40: 881–887.<br />

19. Ball JM, Tiam P, Zeng CQ-Y et al. Age-dependent diarrhoea<br />

is induced by a viral nonstructural glycoprotein.<br />

Science 1996; 272: 101–104.<br />

20. Zhang M, Zeng CQ-Y, Morris AP et al. A functional<br />

NSP4 enterotoxin peptide secreted from rotavirus<br />

infected cells. J Virol 2000; 74: 11663–11670.<br />

21. Au KS, Chan WK, Burns JW et al. Receptor activity of<br />

rotavirus nonstructural glycoprotein NS28. J Virol 1989;<br />

63: 4553–4562.<br />

22. Meyer JC, Bergmann CC, Bellamy AR. Interaction of<br />

rotavirus cores with nonstructural glycoprotein NS28.<br />

Virology 1989; 171: 98–107.<br />

23. Taylor JA, O’Brien JA, Lord VJ et al. The RER-localized<br />

rotavirus intracellular receptor: a truncated purified<br />

soluble form is multivalent and binds virus particles.<br />

Virology 1993; 194: 807–814.<br />

24. Bergmann CC, Maass D, Poruchynsky MS et al.<br />

Topology of the non-structural rotavirus receptor glycoprotein<br />

NS28 in the rough endoplasmic reticulum.<br />

EMBO J 1989; 8: 1695–1703.<br />

25. Tian P, Hu Y, Schilling WP et al. The nonstructural<br />

glycoprotein of rotavirus affects intracellular calcium<br />

levels. J Virol 1994; 68: 251–257.<br />

26. Tian P, Estes MK, Hu Y et al. The rotavirus nonstructural<br />

glycoprotein NSP4 mobilizes Ca2+ from the endoplasmic<br />

reticulum. J Virol 1995; 69: 5763–5772.<br />

27. Newton K, Meyer JC, Bellamy AR et al. Rotavirus<br />

nonstructural glycoprotein NSP4 alters plasma<br />

membrane permeability in mammalian cells. J Virol<br />

1997; 71: 9458–9465.<br />

28. Richardson SC, Grimwood K, Bishop RF. Analysis of<br />

homotypic and heterotypic serum immune responses to<br />

rotavirus proteins following primary rotavirus infection<br />

by using the radioimmunoprecipitation technique. J<br />

Clin Microbiol 1993; 31: 377–385.<br />

29. Johansen K, Hinkula J, Espinoza F et al. Humoral and<br />

cell-mediated immune responses to the NSP4 enterotoxin<br />

of rotavirus. J Med Virol 1999; 59: 369–377.<br />

30. Estes MK, Morris AP. A viral enterotoxin. A new mechanism<br />

of virus-induced pathogenesis. Adv Exp Med Biol<br />

1999; 473: 73–82.<br />

31. Salim AF, Phillips AD, Walker-Smith JA et al. Sequential<br />

changes in small structure and function during<br />

rotavirus infection in neonatal rats. Gut 1996; 36:<br />

231–238.<br />

32. Ciarlet M, Estes MK. Rotaviruses and calicivirus infections<br />

of the gastrointestinal tract. Curr Opin<br />

Gastroenterol 2001; 17: 10–16.<br />

33. Halaihel N, Lievin V, Alvarado F et al. Rotavirus infection<br />

impairs intestinal brush-border membrane Na + -<br />

solute cotransport activities in young rabbits. Am J<br />

Physiol Gastrointest Liver Physiol 2000; 279: G587–G596.<br />

34. Casola A, Estes MK, Crawford SE et al. Rotavirus infection<br />

of cultured intestinal epithelial cells induces secretion<br />

of CXC and CC chemokines. Gastroenterology 1998;<br />

114: 947–955.<br />

35. Rollo EE, Kumar KP, Reich NC et al. The epithelial cell<br />

response to rotavirus infection. J Immunol 1999; 163:<br />

4442–4452.<br />

36. Guarino A, Spagnuolo MI, Russo S et al. Etiology and<br />

risk factors of severe and protracted diarrhea. J Pediatr<br />

Gastroenterol Nutr 1995; 20: 173–178.<br />

37. de Wit MAS, Koopmans MPG, Kortbeek LM et al.<br />

Etiology of gastroenteritis in sentinel general practices<br />

in the Netherlands. Clin Infect Dis 2001; 33: 280–288.


38. Pang XL, Honma S, Nakata S et al. Human caliciviruses<br />

in acute gastroenteritis of young children in the<br />

community. J Infect Dis 2000; 181: S288–S294.<br />

39. Capano G, Guandalini S, Guarino A et al. Enteric infections,<br />

cow’s milk protein intolerance and parenteral<br />

infections in 118 consecutive cases of acute diarrhea in<br />

children. Eur J Pediatr 1984; 142: 281–285.<br />

40. Bishop RF, Davidson GP, Holmes IH et al. Virus particles<br />

in epithelial cells of duodenal mucosa from children<br />

with viral gastroenteritis. Lancet 1973; 2: 1281–1283.<br />

41. Parashar UD, Hummelmann E, Brese JS et al. Global<br />

illness and deaths caused by Rotavirus disease in children.<br />

Emerg Infect Dis 2003; 9: 565–572.<br />

42. Glass RI, Kilgore PE, Holman RC et al. The epidemiology<br />

of rotavirus diarrhea in the United States: surveillance<br />

and estimates of disease burden. J Infect Dis 1996;<br />

174: S5–11.<br />

43. Ho MS, Glass RI, Pinsky PF et al. Rotavirus as a cause of<br />

diarrheal morbidity and mortality in the United States. J<br />

Infect Dis 1988; 158: 1112–1116.<br />

44. Velasquez FR, Matson DO, Calva JJ et al. Rotavirus<br />

infection in infants as protection against subsequent<br />

infections. N Engl J Med 1996; 335: 1022–1028.<br />

45. Prince DS, Astry C, Vonderfecht S et al. Aerosol transmission<br />

of experimental rotavirus infection. Pediatr<br />

Infect Dis 1986; 5: 218–222.<br />

46. Bishop RF. Natural history of rotavirus infection. In AZ<br />

Kapikian, ed. Viral Infections of the Gastrointestinal<br />

Tract, 2nd edn. New York: Marcel Dekker, 1994;<br />

131–167.<br />

47. Gentsch JR, Woods PA, Ramachandran M et al. Review<br />

of G and P typing results from a global collection of<br />

rotavirus strains: implications for vaccine development.<br />

J Infect Dis 1996; 174: S30–S36.<br />

48. Nakata S, Gatheru Z, Ukae S et al. Epidemiological<br />

study of the G serotype distribution of group A<br />

rotaviruses in Kenya from 1991 to 1994. J Med Virol<br />

1999; 58: 296–303.<br />

49. Unicomb LE, Podder JR, Gentsch PA et al. Evidence of<br />

high-frequency genomic reassortment of group A<br />

rotavirus strains in Bangladesh: emergence of type G9<br />

in 1995. J Clin Microbiol 1999; 37: 1885–1891.<br />

50. Santos N, Lima RCC, Pereira CFA et al. Detection of<br />

rotavirus types G8 and G10 among Brazilian children<br />

with diarrhea. J Clin Microbiol 1998; 36: 2727–2729.<br />

51. Aijas S, Gowda K, Jagannath HV et al. Epidemiology of<br />

symptomatic human rotaviruses in Bangladore and<br />

Mysore, India from 1988 to 1994 as determined by electropherotype,<br />

subgroup and serotype analysis. Arch<br />

Virol 1996; 141: 715–726.<br />

52. Ramachandran M, Gentsch JR, Parashar UD et al.<br />

Detection and characterization of novel rotavirus strains<br />

in the United States. J Clin Microbiol 1998; 36:<br />

3223–3229.<br />

53. Gouvea V, Santos N. Rotavirus serotype G5: an emerging<br />

cause of epidemic childhood diarrhea. Vaccine 1999; 17:<br />

1291–1292.<br />

54. Cascio A, Vizzi E, Alaimo C et al. Rotavirus gastroenteritis<br />

in Italian children: can severity of symptoms be<br />

related to the infecting virus? Clin Infect Dis 2001; 32:<br />

1126–1132.<br />

55. Madeley CR, Cosgrove BP. Virus in infantile gastroenteritis.<br />

Lancet 1975; 2: 451–452.<br />

56. Glass RI, Noel J, Mitchell D et al. The changing<br />

epidemiology of astrovirus-associated gastroenteritis: a<br />

review. Arch Virol Suppl 1996; 12: 287–300.<br />

57. Unicomb LE, Banu NN, Azim T et al. Astrovirus infection<br />

in association with acute, persistent and nosocomial<br />

diarrhea in Bangladesh. Pediatr Infect Dis J 1998;<br />

17: 611–614.<br />

References 143<br />

58. Mitchell DK, Monroe SS, Jiang X et al. Virologic<br />

features of an astrovirus diarrhea outbreak in a day care<br />

center revealed by reverse transcriptase-polymerase<br />

chain reaction. J Infect Dis 1995; 172: 1437–1444.<br />

59. Grohmann GS, Glass RI, Pereira HG et al. Enteric<br />

viruses and diarrhea in HIV-infected patients. Enteric<br />

Opportunistic Infections Working Group. N Engl J Med<br />

1993; 329: 14–20.<br />

60. Cox GJ, Matsui SM, Lo RS et al. Etiology and outcome<br />

of diarrhea after marrow transplantation: a prospective<br />

study. Gastroenterology 1994; 107: 1398–1407.<br />

61. Cubitt WD, Mitchell DK, Carter MJ et al. Applications of<br />

electronmicroscopy, enzyme immunoassay, a RT-PCR to<br />

monitor an outbreak of astrovirus type 1 in a paediatric<br />

bone marrow transplant unit. J Med Virol 1999; 57:<br />

313–321.<br />

62. Wyatt RG, Dolin R, Blacklow NR et al. Comparison of<br />

three agents of acute infectious nonbacterial gastroenteritis<br />

by cross-challenge in volunteers. J Infect Dis<br />

1974; 129: 709–714.<br />

63. Monroe SS, Jiang B, Stine SE et al. Subgenomic RNA<br />

sequence of human astrovirus supports classification of<br />

Astroviridae as a new family of RNA viruses. J Virol<br />

1993; 67: 3611–3614.<br />

64. Adler JL, Zickl R. Winter vomiting disease. J Infect Dis<br />

1969; 119: 668–673.<br />

65. Kapikian AZ, Wyatt RG, Dolin R et al. Visualization by<br />

immune electron microscopy of a 27nm particle associated<br />

with acute infectious nonbacterial gastroenteritis. J<br />

Virol 1972; 10: 1075–1081.<br />

66. Madeley CR, Cosgrove BP. 28 nm particles in faeces in<br />

infantile gastroenteritis. Lancet 1975; 2: 451–452.<br />

67. Chiba S, Sakuma Y, Kogasaka R et al. An outbreak of<br />

gastroenteritis associated with calicivirus in an infant<br />

home. J Med Virol 1979; 4: 249–254.<br />

68. Fankhauser RI, Noel JS, Monroe SS et al. Molecular<br />

epidemiology of ‘Norwalk-like viruses’ in outbreaks of<br />

gastroenteritis in the United States. J Infect Dis 1998;<br />

178: 1571–1578.<br />

69. Lopman BA, Reacher MH, van Duijnhoven Y et al. Viral<br />

gastroenteritis outbreaks in Europe, 1995–2000. Emerg<br />

Infect Dis 2003; 9: 90–96.<br />

70. Brown DWG. The pattern and burden of disease due to<br />

human calicivirus infections in the UK (abstract S1-2).<br />

In Proceedings of the International Workshop on Human<br />

Caliciviruses, Atlanta GA, 29–31 March 1999. Atlanta:<br />

Centers for Disease Control and Prevention,1999.<br />

71. Sawyer LA, Murphy JJ, Kaplan JE et al. 25 to 30 nm<br />

virus particle associated with a hospital outbreak of<br />

acute gastroenteritis with evidence for airborne transmission.<br />

Am J Epidemiol 1988; 127: 1261–1271.<br />

72. Mounts AW, Ando T, Koopmans M et al. Cold weather<br />

seasonality of gastroenteritis associated with Norwalklike<br />

viruses. J Infect Dis 2000; 181: S284–S287.<br />

73. Jiang Xi, Pickering LK. Update on caliciviruses and<br />

human acute gastroenteritis. Pediatr Infect Dis J 2002;<br />

21: 1069–1070.<br />

74. Zahorsky J. Hyperemesis hiemis or the winter vomiting<br />

disease. Arch Pediatr 1929; 46: 391.<br />

75. Green J, Vinje J, Gallimore CI et al. Capsid protein<br />

diversity among Norwalk-like viruses. Virus Genes 2000;<br />

20: 227–236.<br />

76. Favier AL, Schoehn G, Jaquinod M et al. Structural<br />

studies of human enteric adenovirus type 41. Virology<br />

2002; 293: 75–85.<br />

77. Kotloff KL, Losonsky GA, Morris JG et al. Enteric adenovirus<br />

infection and childhood diarrhea: an epidemiologic<br />

study in three clinical settings. Pediatrics 1989; 84:<br />

219–225.


144<br />

Viral diarrhea<br />

78. Soares CC, Volotao EM, Albuquerque MC et al.<br />

Prevalence of enteric adenoviruses among children with<br />

diarrhea in four Brazilian cities. J Clin Virol 2002; 23:<br />

171–177.<br />

79. Waters V, Ford-Jones EL, Petric M et al. Etiology of<br />

community-acquired pediatric viral diarrhea: a prospective<br />

longitudinal study in hospitals, emergency departments,<br />

pediatric practices and child care centers during<br />

the winter rotavirus outbreak, 1997 to 1998. The<br />

Pediatric Rotavirus Epidemiology Study for<br />

Immunization Study Group. Pediatr Infect Dis J 2000;<br />

19: 843–848.<br />

80. Weiss M, Horzinek MC. The proposed family,<br />

Toroviridae: agents of enteric infections. Arch Virol<br />

1987; 92: 1–15.<br />

81. Woode GN, Reed DE, Runnels PL et al. Studies with an<br />

unclassified virus isolated from diarrheic calves. Vet<br />

Microbiol 1982; 7: 221–240.<br />

82. Woode GN, Saif LJ, Quesanda M et al. Comparative<br />

studies on three isolates of Breda virus of calves. Am J<br />

Vet Res 1985; 46: 1003–1010.<br />

83. Beards GM, Green J, Hall C et al. An enveloped virus in<br />

stools of children and adults with gastroenteritis resembles<br />

the Breda virus of calves. Lancet 1984; 1:<br />

1050–1052.<br />

84. Snijder E, Horzinek MC. Toroviruses: replication, evolution<br />

and comparison with other members of the coronavirus-like<br />

family. J Gen Virol 1993; 74: 2305–2316.<br />

85. Cavanagh D, Horzinek M. Genus Torovirus assigned to<br />

the Coronaviridae. Arch Virol 1993; 128: 395–396.<br />

86. Cavanaugh D. Nidovirales: a new order comprising<br />

Coronaviridae and Arteriviridae. Arch Virol 1997; 142:<br />

629–633.<br />

87. Jamieson FB, Wang EEL, Bain C et al. Human Torovirus:<br />

a new nosocomial gastrointestinal pathogen. J Infect Dis<br />

1998; 178: 1263–1269.<br />

88. Yamashita T, Kobayashi S, Sakae K et al. Isolation of<br />

cytophathic small round viruses with BS-C-1 cells from<br />

patient with gastroenteritis. J Infect Dis 1991; 164:<br />

954–957.<br />

89. Yamashita T, Sakae K, Tsuzuki H et al. Complete<br />

nucleotide sequence and genetic organization of Aichi<br />

virus, a distinct member of Picornaviridae associated<br />

with acute gastroenteritis in humans. J Virol 1998; 72:<br />

8408–8412.<br />

90. King AMQ, Brown F, Christian P et al. Picornaviridae.<br />

In van Regenmortel MHV, Fauquet CM, Bishop DHL, et<br />

al., eds. Virus Taxonomy: Seventh Report of the<br />

International Committee on Taxonomy of Viruses. San<br />

Diego: Academy Press, 2000: 657–678.<br />

91. King AMQ, Brown F, Christian P et al. Picornavirus<br />

taxonomy: a modified species definition and proposal<br />

for three new genera. XIth International Congress of<br />

Virology, Sidney, Australia, 1999.<br />

92. Yamashita T, Ito M, Tsuzuki H et al. Identification of<br />

Aichi virus infection by measurement of immunoglobu-<br />

lin responses in a enzyme linked immunosorbent assay.<br />

J Clin Microbiol 2001; 39: 4178–4180.<br />

93. Stohr K. A multicentre collaboration to investigate the<br />

cause of severe acute respiratory syndrome. Lancet<br />

2003; 361: 1730–1733.<br />

94. Chandra R. Picobirnavirus, a novel group of undescribed<br />

viruses of mammals and birds: a minireview.<br />

Acta Virol 1997; 41: 59–62.<br />

95. Guarino A, Albano F, Guandalini S et al. Oral rehydration<br />

solution: toward a real solution. J Pediatr<br />

Gastroenterol Nutr 2001; 33: S2–S12.<br />

96. Guarino A, Albano F. Guidelines for the approach to<br />

outpatient children with acute diarrhoea. Acta Paediatr<br />

2001; 90: 1087–1095.<br />

97. Guarino A, Berni Canani R, Russo S. Developments in<br />

the treatment of rotaviral gastroenteritis: oral therapy<br />

with immunoglobulins and prospects for a vaccine. Clin<br />

Immunother 1995; 3: 476–484.<br />

98. Guarino A, Guandalini S, Albano F et al. Enteral<br />

immunoglobulin for treatment of protracted Rotaviral<br />

diarrhea. Pediatr Infect Dis J 1991; 10: 612–614.<br />

99. Guarino A, Berni Canani R, Russo S et al. Oral<br />

immunoglobulins for treatment of acute rotaviral<br />

gastroenteritis. Pediatrics 1994; 93: 12–16.<br />

100. Guarino A, Russo S, Castaldo A et al. Passive<br />

immunotherapy for Rotavirus-induced diarrhoea in<br />

children with HIV infection. AIDS 1996; 10: 1176–1178.<br />

101. Guarino A, Albano F, Berni Canani R et al. HIV, fatal<br />

rotavirus infection, and treatment options. Lancet 2002;<br />

359: 74.<br />

102. De Marco G, Bruzzese E, Di Nardo G et al. Rotavirus<br />

induces a galanin-dependent chloride secretion which<br />

is inhibited by human immunoglobulin in a Caco-2<br />

experimental model. J Pediatr Gastroenterol Nutr 2003;<br />

36: 521(abstr).<br />

103. De Zoysa I, Feachem RG. Interventions for the control of<br />

diarrhoeal disease among young children: rotavirus and<br />

cholera immunization. Bull World Health Organ 1985;<br />

62: 569–583.<br />

104. Rennels MB. The rotavirus vaccine story: a clinical<br />

investigator’s view. Pediatrics 2000; 106: 123–125.<br />

105. Cunliffe NA, Bresee JS, Hart CA. Rotavirus vaccines:<br />

development, current issues and future prospects. J<br />

Infect 2002; 45: 1–9.<br />

106. Ball JM, Graham DY, Opekun AR et al. Recombinant<br />

Norwalk-like particles given orally to volunteers: phase<br />

I study. Gastroenterology 1999; 1117: 40–48.<br />

107. Mason H, Ball JM, Shi J et al. Expression of Norwalk<br />

virus capsid protein in transgenic tobacco and potato<br />

and its oral immunogenicity in mice. Proc Nat Acad Sci<br />

USA 1996; 93: 5335–5340.<br />

108. Tacket C, Mason H, Losonsky G et al. Human immune<br />

responses to a novel Norwalk virus vaccine delivered in<br />

transgenic potatoes. J Infect Dis 2000; 182: 302–305.


10<br />

Bacterial infections<br />

Alessio Fasano<br />

Enteric infections exact a heavy toll on human<br />

populations, particularly among children. Despite<br />

the explosion of knowledge on the pathogenesis of<br />

enteric diseases experienced during the past<br />

decade, the number of diarrheal episodes and<br />

childhood deaths reported worldwide remains of<br />

apocalyptic dimensions. In the next 15s, a child<br />

somewhere in the world will die from diarrhea.<br />

Worldwide, it is estimated that 6–60 billion cases<br />

of gastrointestinal illness occur annually, the vast<br />

majority being severe enough and affecting so<br />

many unprivileged populations to pose a serious<br />

global health burden. In overpopulated developing<br />

countries, poor sanitation and hygiene, unsafe<br />

water supplies and limited education contribute to<br />

the propagation of diarrheal diseases. In industrialized<br />

nations, diarrhea had been thought of as<br />

more of an inconvenience rather than a serious<br />

cause of illness, but recent attention to food contamination<br />

by pathogens such as Salmonella and<br />

Escherichia coli, in combination with a growing<br />

population of immunosuppressed patients, has led<br />

to increased recognition of their impact. The situation<br />

is further complicated by the recent escalation<br />

of international terrorism that is raising the<br />

risk of epidemics of enteric pathogens beyond the<br />

boundaries of natural endemic areas.<br />

The appreciation of these multifaceted aspects of<br />

the problem is pivotal for full comprehension of<br />

the threat that diarrheal diseases pose to public<br />

health and for appropriate allocation of resources<br />

and efforts to tackle them. However, there are a<br />

series of obstacles that have historically hampered<br />

this process, including non-standardized definitions<br />

of disease and symptoms; failure to identify a<br />

causative agent in many, if not most, cases of<br />

disease; failure to report episodes to health authorities;<br />

and the existence of incompatible reporting<br />

systems. Most of all, the lack of long-term commit-<br />

ment of rich countries to finance sanitation<br />

campaigns to treat and prevent diarrheal diseases<br />

in endemic areas has been a key limiting factor in<br />

tackling the burden of gastrointestinal diseases.<br />

Nevertheless, bacterial genome sequencing and<br />

better understanding of the pathogenic mechanisms<br />

involved in the onset of diarrhea, particularly<br />

concerning the elaboration of toxin (Figure<br />

10.1), are finally leading to preventive interventions,<br />

such as enteric vaccines, that may have<br />

a significant impact on the magnitude of this<br />

human plague.<br />

The majority of acute infectious diarrheal illnesses<br />

in both children and adults in developing countries<br />

are due to bacteria (Table 10.1). The risk<br />

factors for transmission of these pathogens are<br />

higher in these countries, owing to increased exposure<br />

to the organisms (by contact with feces, water,<br />

food and fomites) and increased susceptibility of<br />

the host due to malnutrition.<br />

Cholera<br />

Of all enteric pathogens, Vibrio cholerae is responsible<br />

for the most rapidly fatal diarrheal disease in<br />

humans. 1 Although cholera is rare in developed<br />

countries, it remains a major cause of diarrheal<br />

morbidity and mortality in many parts of the<br />

developing world. 2 However, with the occurrence<br />

of both natural (e.g. earthquakes) and humangenerated<br />

calamities (such as ethnic wars), the<br />

spreading of cholera infection in refugee camps,<br />

where sanitary conditions resemble those in<br />

cholera endemic areas, represents a significant<br />

threat worldwide. Vibrios (from the Greek for<br />

‘comma’) are single, short, curved Gram-negative<br />

rods, with a single, long, polar flagellum that<br />

allows for the organism’s characteristic motility.<br />

145


146<br />

Bacterial infections<br />

Figure 10.1 Enterocyte intracellular signaling leading to intestinal secretion. Four main pathways seem to be involved in<br />

the intestinal secretion of water and electrolytes: cAMP, cGMP, Ca and the cytoskeleton. These pathways are activated by<br />

several enteric pathogens, either directly or through the elaboration of enterotoxic products. CT, cholera toxin; LT, heatlabile<br />

enterotoxin; TDH, thermostable direct hemolysin; C.D., Clostridium difficile; EAST1, enteroaggregative Escherichia<br />

coli heat stable toxin 1; STa, heat stable toxin a; AC, adenylate cyclase; GC, guanylate cyclase; CM, calmodulin; PKC,<br />

protein kinase C; ZOT, Zonula Occludens Toxin; EGF-r, epidermal growth factor receptor; ECM, extracellular matrix.<br />

Table 10.1 Percentage of identified bacterial pathogens in symptomatic patients from industrialized<br />

and developing countries<br />

Agent Industrialized countries (%) Developing countries (%)<br />

Vibrio cholerae


Vibrio cholerae O1 is transmitted by the fecal–oral<br />

route and is spread through contaminated food<br />

and water, with a period of incubation of a few<br />

hours to 5 days. The vast majority of subjects<br />

infected remain asymptomatic or experience mild<br />

disease with watery stools, rare nausea or vomiting,<br />

and no significant dehydration. Stools are<br />

classically described as ‘rice water’ due to the<br />

presence of mucus in clear stools. In cholera<br />

gravis, profuse watery diarrhea and vomiting lead<br />

to massive fluid and electrolyte loss that can occur<br />

at a rate of 1 litre/h, and can reach a total volume<br />

loss of 100% of body weight. Vibrios can be easily<br />

identified from the stool by Gram stain.<br />

Cholera has been a recognized human plague for<br />

over two millennia. The study of this disease has<br />

spanned from ancient Greece, and the Roman<br />

Empire, to the famous John Snow’s tracing of an<br />

outbreak to a water pump in Broad Street, London,<br />

in 1854. Thirty years later Robert Koch proposed<br />

that the agent responsible for cholera produced ‘a<br />

special poison’ acting on the intestinal epithelium<br />

and that the symptoms of cholera could be<br />

‘regarded essentially as a poisoning’. Considerable<br />

time elapsed before the existence of this hypothesized<br />

toxin was demonstrated in 1959. Ten years<br />

later, cholera toxin (CT) was purified to homogeneity.<br />

It was only recently, however, that we<br />

learned that vibrios produce a variety of other<br />

extracellular products that enable these microorganisms<br />

to activate different intracellular signaling<br />

in the host mammalian cells, all leading to<br />

diarrhea. One of the most intriguing new toxins<br />

discovered in V. cholerae is zonula occludens toxin<br />

(Zot), 3 an enterotoxin that increases the intestinal<br />

permeability by interacting with a mammalian cell<br />

receptor, with subsequent activation of intracellular<br />

signaling leading to the disassembly of the<br />

intercellular tight junctions (Figure 10.2).<br />

The introduction of oral rehydration solutions<br />

(ORS) decreased the mortality from this illness<br />

from over 50% to less than 1%. The use of antibiotics<br />

has limited indications, but has been<br />

demonstrated to reduce the volume and duration<br />

of diarrhea by half and reduce the duration of<br />

excretion to one day. Tetracycline (500 mg/dose,<br />

given four times per day) is the most widely used<br />

antibiotic, but large outbreaks of tetracycline-resistant<br />

organisms have been reported. Furazolidone<br />

(1.25mg/kg four times per day), trimethoprim<br />

Cholera 147<br />

(TMP, 5mg/kg twice per day), sulfamethoxazole<br />

(SMX, 25mg/kg twice per day), and erythromycin<br />

(10mg/kg three times per day) have been<br />

suggested for children.<br />

Both killed whole-cell and live attenuated cholera<br />

vaccines have been proposed as a preventive<br />

intervention for cholera. 3 A large double-blind<br />

field trial of the killed vaccine showed 85% efficacy<br />

for a period of 4–6 months, dropping to 50%<br />

over 3 years of follow-up. 4 A locally produced<br />

killed vaccine in Vietnam provided a 66% protection<br />

against El Tor cholera during an outbreak<br />

occurring 8–10 months after vaccination. 5 A<br />

genetically engineered attenuated cholera vaccine<br />

(CVD 103-HgR), obtained by deleting the active<br />

subunit of cholera toxin (see below) from a V.<br />

cholerae O1 classic biotype, was well tolerated<br />

when administered to volunteers. This vaccine<br />

elicited a high level of protection (82–100%)<br />

against homologous challenge with a strain of the<br />

same biotype. 6 Protection across biotypes was also<br />

observed, albeit to a lesser extent, 7 lasting for at<br />

least 6 months after a single oral dose. 8 Live attenuated<br />

V. cholerae O139 vaccines have been developed,<br />

with promising preliminary results. 9,10<br />

Non-01 Vibrio species<br />

V. parahaemolyticus, V. fluvialis, V. mimicus, V.<br />

hollisae, V. furnissii and V. vulnificus cause<br />

sporadic cases of gastroenteritis, and are present<br />

in coastal and estuarine areas throughout the<br />

world. 11 Virtually all cases of non-O1 Vibrio infections<br />

in the USA are associated with eating raw<br />

shellfish, 12 and the gastroenteritis can range from<br />

a mild illness to profuse, watery diarrhea comparable<br />

to that seen in epidemic cholera. Diarrhea,<br />

abdominal cramps and fever are the most<br />

common symptoms, with nausea, vomiting and<br />

bloody stools occurring less frequently. 11 As with<br />

V. cholerae O1, the mainstay of therapy for diarrheal<br />

disease is oral rehydration. In cases of<br />

septicemia (which typically occur in immunocompromised<br />

patients), supportive care and<br />

correction of shock are essential interventions<br />

associated with the antibiotic treatment (tetracycline).<br />

In countries such as the USA non-O1 infections<br />

can be prevented by not eating raw or undercooked<br />

seafood, particularly during the warm<br />

months.


148<br />

Bacterial infections<br />

Figure 10.2 Proposed zonula occludens toxin (Zot) intracellular signaling leading to the opening of intestinal tight junctions.<br />

Zot interacts with a specific surface receptor (1) whose distribution within the intestine varies. The protein is then<br />

internalized and activates phospholipase C (2) that hydrolyzes phosphatidyl inositol (PPI) (3) to release inositol 1,4,5-tris<br />

phosphate (IP3) and diacylglycerol (DAG) (4). Protein kinase Cα (PKCα) is then activated (5), either directly (via DAG) (4) or<br />

through the release of intracellular Ca ++ (via IP3) (4a). PKCα catalyzes the phosphorylation of target protein(s), with subsequent<br />

polymerization of soluble G actin in F actin (7). This polymerization causes the rearrangement of the filaments of<br />

actin and the subsequent displacement of proteins (including ZO1) from the junctional complex (8). As a result, intestinal<br />

tight junctions become looser.<br />

Salmonella<br />

Salmonella typhi and S. paratyphi are Gramnegative,<br />

motile bacilli that colonize only humans.<br />

Therefore, disease is acquired through close<br />

personal contact or through the ingestion of water<br />

or food contaminated with human excrement.<br />

Typhoid fever continues to represent a global health<br />

problem, with more than 12.5 million annual cases,<br />

and subequatorial countries reporting mortality<br />

rates of up to 32% despite antibiotic treatment. 13 In<br />

the USA, substantial progress has been made in<br />

their eradication, with better sanitation, foodhandling<br />

and water treatment. These bacteria cause<br />

systemic illnesses characterized by fever, gastrointestinal<br />

symptoms and occasionally psychosis,<br />

confusion or rose spots on the trunk. 14 The incubation<br />

period varies between 5 and 21 days (depending<br />

on the inoculum ingested), and chills,<br />

headache, cough, weakness and muscle pain are<br />

frequent prodromes. Most symptoms resolve by the<br />

4th week without antimicrobial treatment.<br />

However, some patients relapse with high fever,<br />

abdominal pain from inflammation of Peyer’s<br />

patches, and intestinal microperforation followed<br />

by secondary bacteremia with normal enteric flora.<br />

The definitive diagnosis of enteric fever requires the<br />

isolation of S. typhi or S. paratyphi from blood,<br />

stool, urine, rose spots, bone marrow, or gastric or<br />

enteric secretions. Chloramphenicol is the treatment<br />

of choice and has been shown to reduce the<br />

duration of fever and mortality.


In contrast to S. typhi, the cases of infection with<br />

non-typhoidal salmonellae infections have been<br />

increasing in the developed world. Patients as<br />

higher risk for infection include those with<br />

immunodeficiencies, age younger than 3 months,<br />

alterations in intestinal defenses (achlorhydria,<br />

antacids, rapid gastric emptying post-gastrectomy),<br />

impaired reticuloendothelial function,<br />

(sickle cell and hemolytic anemias) and ingestion<br />

of antibiotics to which the organism was resistant.<br />

Reservoirs include a wide range of domestic and<br />

wild animals, including poultry, swine, cattle,<br />

rodents and reptiles. S. enteriditis is the leading<br />

reported cause of food-borne disease outbreaks in<br />

the USA, with eggs and contaminated raw fruits<br />

and vegetables identified as vehicles. 15 Transmission<br />

from person to person and from pets has<br />

also been reported. The incubation period is<br />

6–48 h, after which fever, headache, vomiting,<br />

abdominal pain and watery diarrhea (which may<br />

contain blood, mucus, and leukocytes) occur for<br />

about 1 week. Severe extraintestinal infections can<br />

range from life-threatening sepsis to focal infections<br />

in the meninges, bones and lungs. The<br />

micro-organism is easily isolated from fresh stools<br />

or blood culture. Antimicrobials are not indicated<br />

to treat asymptomatic carriage or uncomplicated<br />

infections in the normal host, as they may prolong<br />

excretion or induce relapse. Although efficacy is<br />

unproven, it is common clinical practice to administer<br />

oral or parenteral antibiotics to high-risk<br />

patients or to those who have an extraintestinal<br />

focus of infection. Increasing resistance to<br />

commonly used antibiotics is seen, so the choice<br />

of regimens should be guided by susceptibility<br />

data. Suggested therapies include TMP–SMX<br />

(Bactrim ® , Septra ® , Sulfatrim ® ), ampicillin<br />

(10–20% of isolates in the USA are resistant), cefotaxime,<br />

ceftriaxone or chloramphenicol.<br />

Hygienic practices for preventing food-borne<br />

transmission is the most efficient prevention for<br />

non-typhoidal Salmonella infections, since the<br />

vast majority of outbreaks and sporadic cases<br />

result from culinary practices that allow the organisms<br />

to survive and multiply in food. Parents<br />

should be instructed to avoid serving food containing<br />

raw or undercooked eggs and meat (especially<br />

poultry). Food should be thawed in the refrigerator,<br />

microwave, or under cold water but not at room<br />

temperature, because surface bacteria begin to<br />

multiply when the outer layers warm. Eggs should<br />

Shigella 149<br />

be cooked until both the yolk and the white are<br />

firm, and meats must reach an internal temperature<br />

of at least 74°C (165°F). Frequent hand<br />

washing is important. High-risk pets (especially<br />

chicks, ducklings and reptiles) are not advisable<br />

for young children.<br />

An extremely problematic situation is the management<br />

of an infected child who is attending day<br />

care. Excretion can go on for weeks and create a<br />

hardship to working parents if the child must be<br />

excluded from day care. While the decision to<br />

admit such a child must be made in concert with<br />

day care and public health officials, it is generally<br />

recommended that the infected children be<br />

excluded from day care if they are symptomatic or<br />

if adequate hygiene cannot be ensured. There is no<br />

vaccine to prevent non-typhoidal salmonellosis.<br />

Enteric fever<br />

The definitive diagnosis of enteric fever requires<br />

the isolation of S. typhi or S. paratyphi from the<br />

patient. Cultures of blood, stool, urine, rose spots,<br />

bone marrow and gastric and enteric secretions<br />

may all be useful in establishing the diagnosis.<br />

Chloramphenicol has been the treatment of choice<br />

since its introduction, given its low costs and its<br />

high efficiency after oral administration.<br />

Treatment with chloramphenicol reduced typhoid<br />

fever mortality from approximately 20 to 1% and<br />

reduced the duration of the fever from 14–28 days<br />

to 3–5 days. 16<br />

The most effective attenuated vaccine for typhoid<br />

fever currently available, Ty21a, has proved to be<br />

free of adverse reactions in large-scale efficacy<br />

field trials involving almost 600000 pediatric<br />

subjects. 17 When administered as a liquid suspension,<br />

Ty21a protected both young (82% vaccine<br />

efficacy) and older children (69% vaccine<br />

efficacy). 17 Currently, there are three new-generation<br />

attenuated vaccines, genetically engineered<br />

by deleting different pathogenic factors, that are<br />

undergoing extensive phase II trials.<br />

Shigella<br />

Kiyoshi Shiga first isolated Shigella dysenteriae<br />

type 1 during a severe dysentery epidemic in Japan


150<br />

Bacterial infections<br />

in 1896, when more than 90000 cases were<br />

described with a mortality rate approaching<br />

30%. 18 Shigellae are Gram-negative, non-lactose<br />

fermenting, non-motile bacilli, with S. sonnei the<br />

main type found in industrialized countries, and<br />

S. flexneri and S. dysenteriae predominating in<br />

underdeveloped countries. Humans are the only<br />

natural hosts and transmission occurs by<br />

fecal–oral contact. The low infectious inoculum<br />

(as few as ten organisms) 19 renders shigellae<br />

highly contagious. Shigella causes 250 million<br />

cases of diarrhea and 650 000 deaths worldwide. 20<br />

In the USA, S. dysenteriae infection is seen almost<br />

exclusively among travelers. After an incubation of<br />

1–4 days, shigellosis begins with fever, headache,<br />

malaise, anorexia and occasional vomiting and<br />

watery diarrhea with progression to dysentery<br />

within hours to days. Unusual extraintestinal<br />

manifestations may occur, including hemolytic<br />

uremic syndrome (HUS) in children and thrombotic<br />

thrombocytopenic purpura in adults. Most<br />

episodes of shigellosis in otherwise healthy individuals<br />

resolve within 7 days. Shigellae are<br />

extremely fastidious and are best isolated from<br />

fresh stool rapidly inoculated onto selective<br />

culture plates incubated immediately at 37 o C.<br />

Appropriate antibiotics (ampicillin or TMP–SMX)<br />

given for 5 days significantly decrease the duration<br />

of fever, diarrhea, intestinal protein loss and<br />

pathogen excretion. However, Shigella strains that<br />

are resistant to one or both drugs have been<br />

identified. Several promising Shigella mutants<br />

with deletions in virulence genes have entered<br />

clinical trials as oral vaccine candidates.<br />

Campylobacter<br />

These organisms are small, microaerophilic,<br />

spiral-shaped Gram-negative organisms that enjoy<br />

a widespread reservoir in the intestines of both<br />

wild and domestic animals. 21 It is the most<br />

frequently identified bacterial cause of diarrhea in<br />

the USA. Common vehicles are poultry, unpasteurized<br />

milk and contaminated water. 22–24<br />

After an incubation of 3–6 days, enteritis begins<br />

abruptly with cramps and watery diarrhea, which<br />

may progress to blood-containing stools. The<br />

abdominal pain may mimic appendicitis. Diarrhea<br />

usually lasts 4–5 days, with the duration of fecal<br />

excretion 1 month. The micro-organism can be<br />

identified only from stool. Indications for anti-<br />

biotics remain controversial, with some studies<br />

showing a shortened course of diarrhea, and others<br />

no clear benefit. It is advisable to reserve antibiotics<br />

for patients in high-risk groups with severe<br />

symptoms. Erythromycin remains the drug of<br />

choice. Campylobacter vaccine development has<br />

proceeded cautiously, because of concerns about<br />

post-exposure arthritis or Guillain–Barré syndrome.<br />

However, a monovalent, formalininactivated,<br />

C. jejuni whole-cell vaccine with a<br />

mucosal adjuvant has entered human trials.<br />

Yersinia<br />

Yersinia enterocolitica and Y. pseudotuberculosis<br />

are two important human enteropathogens distributed<br />

widely in the environment, with swine<br />

serving as the major reservoir. The incubation<br />

period is 3–7 days, with food-borne transmission<br />

the suspected route for most infections. Yersinia’s<br />

preference for cool temperatures makes this<br />

pathogen more common in Northern Europe,<br />

Scandinavia, Canada, the USA and Japan, where it<br />

is responsible for up to 8% of sporadic diarrhea<br />

episodes. Yersinia enterocolitis occurs most often<br />

in children younger than 5 years 25 and is characterized<br />

by fever, vomiting, exudative pharangitis,<br />

cervical adenitis, abdominal pain and watery<br />

diarrhea, which may contain blood. 26,27 Diarrhea<br />

typically lasts for 14–22 days but fecal excretion<br />

may persist for 7 weeks or longer. Abdominal<br />

complications include appendicitis, pseudoappendicitis,<br />

diffuse ulceration of the intestine<br />

and colon, intestinal perforation, peritonitis,<br />

ileocecal intussusception, toxic megacolon,<br />

cholangitis and mesenteric vein thrombosis.<br />

Bacteremic spread may result in abscess formation<br />

and granulomatous lesions in the liver, spleen,<br />

lungs, kidneys and bone, as well as mycotic<br />

aneurysms, meningitis and septic arthritis. Infection<br />

can also be associated with immunopathological<br />

sequela including reactive arthritis, uveitis,<br />

Reiter’s syndrome and erythema nodosum. 28 It<br />

may be isolated from stool or pharyngeal exudate<br />

on commonly used selective media, and appears as<br />

gram-negative colonies after 48h of growth at<br />

25–28 o C. Most uncomplicated cases resolve<br />

without treatment, which is reserved for patients<br />

with severe symptoms, extraintestinal infections<br />

and immunocompromised hosts. Production of<br />

β-lactamases generally renders all but third-


generation cephalosporins, aztreonam and<br />

imipenem ineffective. Treatment is generally 2–6<br />

weeks, with an initial intravenous antibiotic<br />

(third-generation cephalosporin often in combination<br />

with aminoglycosides), followed by an oral<br />

agent to which the clinical isolate is sensitive.<br />

Escherichia coli<br />

An extremely heterogeneous group of micro-organisms,<br />

Escherichia coli encompasses almost all<br />

features of possible interactions between intestinal<br />

microflora and the host, ranging from a role of<br />

mere harmless presence to that of a highly pathogenic<br />

organism. In fact, the E. coli species is made<br />

up of many strains that profoundly differ from<br />

each other in terms of biological characteristics<br />

and virulence properties. 29<br />

Escherichia coli is a Gram-negative, lactosefermenting<br />

motile bacillus of the family<br />

Enterobacteriaceae. Currently, 171 somatic (O) and<br />

56 flagellar (H) antigens are recognized. Six<br />

distinct categories of E. coli are currently considered<br />

enteric pathogens (based on either outbreak<br />

data or volunteer studies) (Table 10.2). The diagnosis<br />

of diarrheagenic E. coli relies on isolation<br />

from stool and subsequent differentiation from<br />

commensal E. coli either by using genetic probes<br />

or by phenotypic assays. With the exception of E.<br />

coli O157:H7, assays for detection are not routinely<br />

available in clinical laboratories.<br />

Enteropathogenic E. coli<br />

This was the first group of E. coli serotype shown<br />

to be pathogens for humans and has been responsible<br />

for devastating outbreaks of nosocomial<br />

neonatal diarrhea and infant diarrhea in virtually<br />

every corner of the globe. Strains of enteropathogenic<br />

E. coli (EPEC) are distinguished from other E.<br />

coli strains by their ability to induce a characteristic<br />

attaching and effacing lesion in the smallintestinal<br />

enterocytes and by their inability to<br />

produce Shiga toxins. Between the 1940s and the<br />

1960s, EPEC was associated with infant diarrhea<br />

in summertime and nursery outbreaks of diarrhea<br />

in the USA and other industrialized countries.<br />

Since then, it has become extremely uncommon in<br />

industrialized countries, although it is<br />

occasionally reported in child-care settings. 30<br />

Escherichia coli 151<br />

Table 10.2 Pathogenic Escherichia coli<br />

Enteropathogenic E. coli (EPEC)<br />

Enterotoxigenic E. coli (ETEC)<br />

Enteroinvasive E. coli (EIEC)<br />

Enterohemorrhagic E. coli (EHEC)<br />

Diffusely adherent E. coli (DAEC)<br />

Enteroaggregative E. coli (EAggEC)<br />

However, EPEC persists as an important cause of<br />

infantile diarrhea in many developing countries. 31<br />

In nursery outbreaks, transmission was thought to<br />

occur via the hands of caregivers and via fomites.<br />

In less developed countries, contaminated formula<br />

and weaning foods have been incriminated.<br />

Volunteer studies and epidemiological observations<br />

suggest that the infective dose for EPEC is<br />

high (approximately 10 9 colony-forming units<br />

(CFU)). 32 EPEC causes a self-limited watery diarrhea<br />

with a short incubation period (6–48h). There<br />

may be associated fever, abdominal cramps and<br />

vomiting, and EPEC is a leading cause of persistent<br />

diarrhea (lasting 14 days or longer) in children in<br />

developing countries. 33 Although few data exist to<br />

guide antibiotic therapy of EPEC diarrhea, administration<br />

of appropriate antibiotics seems to diminish<br />

morbidity and mortality. A 3-day course of<br />

oral, non-absorbable antibiotics such as colistin or<br />

gentamicin (if available) has been shown to be<br />

effective. 34 Some clinicians also advocate the use<br />

of oral neomycin; however, this drug causes diarrhea<br />

in about 20% of people. In a placebocontrolled<br />

trial among Ethiopian infants with<br />

severe EPEC diarrhea, TMP–SMX and mecillinam<br />

resulted in significant clinical and bacteriological<br />

cure rates by the third day, as compared with<br />

placebo. 35 Strategies for the prevention of EPEC<br />

infection include efforts to improve social and<br />

economic conditions in developing countries,<br />

efforts to encourage breast feeding and prevention<br />

of nosocomial infections.<br />

Enterotoxigenic E. coli<br />

Strains of enterotoxigenic E. coli (ETEC) are an<br />

important cause of diarrheal disease in humans


152<br />

Bacterial infections<br />

and animals worldwide. The clinical importance<br />

of these micro-organsims was first outlined in the<br />

1970s by epidemiological studies in India that<br />

identified them as a major cause of endemic diarrhea.<br />

36 Their pathogenicity is related to the<br />

elaboration of one or more enterotoxins that are<br />

either heat stable (ST) or heat labile (LT) (see<br />

Pathogenesis section) without invading or damaging<br />

intestinal epithelial cells. Together with<br />

rotavirus, ETEC is the leading cause of dehydrating<br />

diarrheal disease among weaning<br />

infants in the developing world. These children<br />

experience 2–3 episodes of ETEC diarrhea in each<br />

of the first 2 years of life. This represents over 25%<br />

of all diarrheal illness 37 and results in an estimated<br />

700000 deaths each year. 38 In industrialized countries,<br />

ETEC does not contribute to endemic<br />

disease, but is notorious for being the leading<br />

agent of travelers’ diarrhea, accounting for about<br />

half of all episodes. 39 Transmission occurs by<br />

ingestion of contaminated food and water, with<br />

peaks during the warm, wet season. Like EPEC,<br />

ETEC requires a relatively high inoculum 40 and a<br />

short incubation period (14–30h). The cardinal<br />

symptom is watery diarrhea, sometimes with<br />

associated fever, abdominal cramps and vomiting.<br />

In its most severe form, ETEC can cause choleralike<br />

purging, even in adults. The illness is typically<br />

self-limited, lasting for less than 5 days and with<br />

few cases persisting beyond 3 weeks. Infection<br />

with ETEC has also been associated with shortand<br />

long-term adverse nutritional consequences in<br />

infants and children.<br />

Most diarrheal illnesses due to ETEC are selflimited<br />

and do not require specific antimicrobial<br />

therapy. Empirical therapy is reserved for those<br />

whose diarrhea is moderate to severe despite rehydration<br />

and supportive measures. Antibiotic<br />

regimens that have been efficacious in clinical<br />

trials, shortening the duration of illness by 1–2<br />

days, include doxycycline, TMP–SMX, ciprofloxacin,<br />

quinolones, and furazolidone. 41 In the<br />

past, the drug of choice for children has been<br />

TMP–SMX; however, except in Central Mexico, 42 a<br />

large proportion of ETEC is now resistant. An alternative<br />

regimen for children is furazolidone.<br />

Prevention of ETEC infection is based on avoiding<br />

contaminated vehicles. Although antibiotics are<br />

effective as prophylactic agents, their use is not<br />

recommended. Some experts advocate the use of<br />

bismuth subsalicylate to diminish the risk of<br />

travelers’ diarrhea. 43 The development of vaccines<br />

against ETEC has received a great deal of attention<br />

because of its disease burden. Oral vaccines for<br />

ETEC are being developed by five different<br />

strategies, including killed whole cells, toxoids,<br />

purified fimbriae, living attenuated strains and<br />

live carrier strains elaborating ETEC antigens. A<br />

killed whole-cell Vibrio cholerae vaccine given<br />

with cholera toxin B (CTB) provided 67%<br />

protection against LT-producing ETEC diarrhea for<br />

3 months. 44 A formalin-inactivated whole-cell oral<br />

vaccine consisting of ETEC strains bearing<br />

colonization factor antigens (CFAs) in combination<br />

with CTB has entered field trial. 45<br />

Enteroinvasive E. coli<br />

This group consists of invasive E. coli strains that<br />

are genetically, biochemically and clinically nearly<br />

identical to Shigella. This section will serve only to<br />

highlight relevant characteristics that distinguish<br />

this pathogen. Strains of enteroinvasive E. coli<br />

(EIEC) are endemic in developing countries, where<br />

they exhibit similar epidemiology to Shigella and<br />

cause an estimated 1–5% of diarrheal episodes<br />

among patients visiting treatment centers. 46 The<br />

occurrence of EIEC in industrialized countries is<br />

limited to rare food-borne outbreaks. 47 From<br />

volunteer studies, it appears that the infectious<br />

inoculum contains more organisms than that<br />

required to cause shigellosis. 48 Like Shigella, EIEC<br />

can produce dysentery, but watery diarrhea is<br />

more common. 49 The rare episodes for which<br />

treatment is desired are treated with antibiotics<br />

recommended for shigellosis. The same general<br />

preventive measures used for Shigella infections<br />

apply to EIEC-associated diarrhea.<br />

Enterohemorrhagic E. coli<br />

These E. coli strains produce either one or both<br />

phage-encoded potent cytotoxins termed Shigalike<br />

toxin I (SLT I) (which is neutralized by antisera<br />

to Shiga toxin produced by S. dysenteriae type<br />

1) or Shiga-like toxin II (SLT II) (which is not<br />

neutralized) and can cause diarrhea or HUS. E. coli<br />

O157:H7 is the prototypic (but not the exclusive)<br />

enterohemorrhagic E. coli (EHEC) serotype, since it<br />

is the predominant SLT-producing E. coli, the one<br />

most commonly associated with HUS in North


America and the type most readily identified in<br />

stool specimens. 50 In 1982, a multistate outbreak<br />

of hemorrhagic colitis that was linked to the<br />

consumption of hamburgers at the same fast-food<br />

restaurant led to the identification of EHEC. 51 The<br />

causative organism was E. coli O157:H7, a serotype<br />

not previously recognized as a human pathogen.<br />

Soon after, Canadian investigators uncovered an<br />

association between O157:H7 and other SLTproducing<br />

strains of E. coli and HUS. 52 EHEC is<br />

now recognized as a global health problem; in<br />

1996, an outbreak in Japan linked to eating radish<br />

sprouts affected over 6000 persons. 53 One the most<br />

severe EHEC outbreaks in the USA took place in<br />

New York State in 1999, with more than 1000<br />

ascertained cases, two HUS-related casualties, and<br />

eight children in dialysis because of renal failure.<br />

Most of the infected individuals attended a fair<br />

whose underground water supply was contaminated<br />

by cow manure from a nearby cattle<br />

barn. The predominant mode of transmission is<br />

ingestion of contaminated, undercooked ground<br />

beef. However, the spectrum of vehicles is widening<br />

to include raw fruits (including apple juice)<br />

and vegetables, 54,55 raw milk, 56 processed meats, 57<br />

and drinking 58 or swimming 59 in contaminated<br />

water. The uncooked food vehicles are usually<br />

contaminated with manure from infected animals<br />

during growth or processing. Person-to-person<br />

transmission is the mode of spread in day-care<br />

outbreaks, where secondary transmission rates of<br />

22% have been reported. 60<br />

EHEC also causes sporadic diarrhea. Isolation from<br />

stools of unselected patients is low (


154<br />

Bacterial infections<br />

a risk factor for the development of HUS and<br />

should be avoided.<br />

Prevention of E. coli O157:H7 is a complex<br />

process. From a public health standpoint, control<br />

measures at the level of farms, slaughterhouses<br />

and processing plants can decrease the risk of<br />

colonization of cattle and contamination of beef.<br />

Since these procedures are unlikely to achieve<br />

complete success, regulations governing proper<br />

processing and cooking of contaminated foods are<br />

also required. Advice to consumers should include<br />

recommending complete avoidance of raw foods of<br />

animal origin. Hamburger should be cooked until<br />

no pink remains and until the juices are clear.<br />

Because of the severity of disease, there has been a<br />

recent focus on vaccine development for EHEC<br />

infection. Efforts have concentrated on three<br />

approaches: parenteral toxoids and live oral carrier<br />

strains elaborating the B subunit of Shiga toxin; 74<br />

vaccines expressing the adhesin intimin, designed<br />

to prevent intestinal colonization; 75 and a<br />

parenteral O157 polysaccharide protein conjugate.<br />

76<br />

Diffusely adhering E. coli<br />

Until recently, diffusely adhering E. coli (DAEC)<br />

was considered a non-pathogenic E. coli, since<br />

early studies failed to find an association between<br />

this micro-organism and diarrheal disease. 77–79<br />

However, more recent studies have demonstrated<br />

such an association, particularly in children older<br />

than 2 years of age. A community-based<br />

case–control study in southern Mexico revealed<br />

that DAEC was significantly associated with diarrhea<br />

in children less than 6 years of age. 80<br />

Prospective cohort studies in Chile 81 and<br />

Bangladesh 81 also demonstrated a diarrheagenic<br />

role for DAEC that peaked in the 48–60-month age<br />

group. 81 This micro-organism was more frequently<br />

isolated from cases of prolonged diarrhea, 82 and it<br />

showed a seasonal pattern similar to that of ETEC,<br />

occurring more frequently in the warm season. 81<br />

The gastrointestinal symptoms that characterize<br />

DAEC infection are practically indistinguishable<br />

from those caused by ETEC, with self-limiting<br />

watery diarrhea rarely associated with vomiting<br />

and abdominal pain. The diagnosis is mainly<br />

based on the DNA probe technique and on the<br />

pattern of adherence of the micro-organism to HE-<br />

2 cells. Given the technical challenge of both<br />

assays, their use is limited to epidemiological<br />

surveys rather than the diagnosis of single individuals.<br />

Enteroaggregative E. coli<br />

Enteroaggregative E. coli (EAggEC) are diarrheagenic<br />

E. coli defined by a characteristic aggregating<br />

pattern of adherence to HEp-2 cells and the<br />

intestinal mucosa. They have been particularly<br />

associated with cases of persistent diarrhea in the<br />

developing world. It has been hypothesized that<br />

the aggregating pattern of adherence may be a<br />

result of non-specific, possibly hydrophobic interaction,<br />

and therefore, not all organisms meeting<br />

the definition of EAggEC may be pathogenic in<br />

humans. Moreover, since epidemiological studies<br />

have not uniformly implicated EAggEC as pathogenic,<br />

some investigators have questioned the<br />

virulence of all EAggEC isolates. Volunteer studies<br />

performed to address both of these questions 82<br />

confirmed that at least some EAggEC strains are<br />

genuine human pathogens but that virulence is not<br />

uniform among isolates. More recently, EAggEC<br />

pathogenicity has also been proven in several<br />

outbreaks.<br />

From the earliest epidemiological reports, EAggEC<br />

was most prominently associated with persistent<br />

cases of pediatric diarrhea (i.e. lasting ≥ 14<br />

days), 84 a condition that represents a disproportionate<br />

share of diarrheal mortality. On the Indian<br />

subcontinent, several independent studies have<br />

demonstrated the importance of EAggEC in pediatric<br />

diarrhea. 85 These studies include hospitalized<br />

patients with persistent diarrhea, 78 outpatients<br />

visiting health clinics, 85 and cases of<br />

sporadic diarrhea detected by household surveillance.<br />

77 In Fortaleza, Brazil, Fang et al have demonstrated<br />

a consistent association between EAggEC<br />

and persistent diarrhea; 86 in this area, EAggEC<br />

accounts for more cases of persistent diarrhea than<br />

all other causes combined. 86 EaggEC have been<br />

implicated as a cause of sporadic diarrhea in other<br />

developing countries (including Mexico, Chile,<br />

Bangladesh, Congo and Iran) as well as in industrialized<br />

countries such as Germany and<br />

England. 87 Besides being responsible for sporadic<br />

cases of diarrhea, EAggEC has also been associated


with outbreaks in India, 88 Serbia, 89 Japan 90 and the<br />

UK. 69<br />

The clinical features of EAggEC diarrhea are<br />

becoming increasingly well defined in outbreaks,<br />

in sporadic cases and in the volunteer model.<br />

Typically, illness is manifested by a watery,<br />

mucoid, secretory diarrheal illness with low-grade<br />

fever and little or no vomiting. 77,91 However, in<br />

epidemiological studies, grossly bloody stools<br />

have been reported in up to one-third of patients<br />

with EAggEC diarrhea. 92 This phenomenon may<br />

well be strain-dependent. In volunteers infected<br />

with EAggEC strain 042, diarrhea was mucoid, of<br />

low volume, and notably, without occult blood or<br />

fecal leukocytes; all patients remained afebrile. In<br />

such volunteers, the incubation period of the<br />

illness ranged from 8 to 18 h. 82<br />

Perhaps even more significant than the association<br />

of EAggEC with diarrhea are the recent data from<br />

Brazil that link EAggEC with growth retardation in<br />

infants. 92 In this study, the isolation of EAggEC<br />

from the stools of infants was associated with a<br />

low z-score for height and/or weight, irrespective<br />

of the presence of diarrheal symptoms. Given the<br />

high prevalence of asymptomatic EAggEC excretion<br />

in many areas, 84,93 such an observation may<br />

imply that the contribution of EAggEC to the<br />

human disease burden is significantly greater than<br />

is currently appreciated. Colonization of EAggEC<br />

is detected by the isolation of E. coli from the<br />

stools of patients and the demonstration of the<br />

aggregative pattern in the HEp-2 assay. Implication<br />

of EAggEC as the cause of the patient’s disease<br />

must be cautious, given the high rate of asymptomatic<br />

colonization in many populations. 84,93 If no<br />

other organism is implicated in the patient’s<br />

illness and EAggEC is isolated repeatedly, then<br />

EAggEC should be considered a potential cause of<br />

the patient’s illness. A DNA-fragment probe has<br />

proven highly specific in the detection of EAggEC<br />

strains. A polymerase chain reaction (PCR) assay<br />

using primers derived from the aggregative probe<br />

sequence shows similar sensitivity and specificity.<br />

94<br />

The optimal management of EAggEC infection has<br />

not been studied. Acute diarrhea is apparently<br />

self-limiting; however, more persistent cases may<br />

benefit from antibiotic and/or nutritional therapy.<br />

Given the high rate of antibiotic resistance among<br />

Clostridium difficile 155<br />

EAggEC, 95 susceptibility testing is recommended<br />

when available.<br />

Clostridium difficile<br />

Even though Clostridium difficile is now recognized<br />

as the single most common cause of bacterial<br />

diarrhea in hospitalized patients, its role as a<br />

pathogen had not been established as recently as<br />

the late 1970s. C. difficile has the ability to become<br />

established in the gastrointestinal tract once the<br />

natural microflora have been modified by antibiotic<br />

therapy. The organism causes intestinal<br />

disease ranging from mild diarrhea to fatal<br />

pseudomembranous colitis (PMC). While C. difficile<br />

is associated with almost all cases of PMC,<br />

only 25% of antibiotic-associated diarrheas are<br />

due to this pathogen. C. difficile is a Gram-positive<br />

anaerobe that forms spores, making this microorganism<br />

very difficult to remove from the hospital<br />

environment. Unlike some toxigenic clostridia,<br />

the production of spores is not associated with<br />

toxin production. C. difficile spreads from patient<br />

to patient 96 and tends to persist in the environment<br />

because of the formation of spores. The<br />

micro-organism is not only present in the infected<br />

patient and soiled linens but can be isolated from<br />

bookshelves, curtains and floors of rooms of<br />

infected patients where it can persist for as long as<br />

5 months. 96–98 The organism is spread primarily by<br />

health-care workers; up to 60% of personnel<br />

attending patients infected with C. difficile in one<br />

study had the organism on their hands. 96 The<br />

isolation of C. difficile toxins from the feces of<br />

asymptomatic normal-term neonates and (in<br />

higher proportion) those admitted into neonatal<br />

intensive care units, 99 further support the concept<br />

of the nosocomial spreading of the infection.<br />

Several outbreaks of C. difficile infection have<br />

been reported in the USA and throughout the<br />

world, and the incidence continues to rise.<br />

Whether this increase represents a true increment<br />

or represents an increased awareness of the<br />

disease is not clear at this stage. Infections with C.<br />

difficile range in severity from asymptomatic forms<br />

to clinical syndromes, such as severe diarrhea,<br />

PMC, and toxic megacolon, and can even lead to<br />

death. 100 The onset of symptomatic forms usually<br />

begins several days after antibiotic therapy is


156<br />

Bacterial infections<br />

started up to 2 months following cessation of treatment.<br />

Diarrhea and abdominal cramps are usually<br />

the first symptoms, followed by the development<br />

of fever and chills in severe cases. Mild forms of<br />

colitis, with bloody stools and mucus, particularly<br />

if they are preceded by antibiotic treatment,<br />

should be considered suspicious for C. difficile<br />

infection. Clinical microbiologists face an array of<br />

methods and commercial tests when considering<br />

what procedure to use for the detection of C. difficile<br />

and its toxins. Culturing of the organism, latex<br />

agglutination, tissue culture assay and enzymelinked<br />

immunosorbent assay (ELISA) are all used<br />

as aids for the diagnosis of C. difficile infection.<br />

In many instances, C. difficile disease is selflimiting,<br />

and the patient may respond simply to<br />

the withdrawal of the offending antibiotic. In more<br />

severe forms, particularly if complicated by PMC,<br />

antibiotic treatment with either oral vancomycin<br />

101 (5–10mg/kg, maximum 500mg, given<br />

every 6h for 7 days) or metronidazole 102<br />

(5–10 mg/kg, maximum 500mg, given every 8h for<br />

7 days) is recommended. Despite pharmacological<br />

treatment, the rate of relapse is significant (up to<br />

40–50% of cases). In these complicated cases, the<br />

use of probiotics, particularly Lactobacillus GG 103<br />

and Saccharomyces boulardii, 104 has been associated<br />

with a significant eradication of C. difficile<br />

and a substantial decrease in the recurrence of the<br />

infection.<br />

Evaluation of diarrheal diseases<br />

The patient’s history, signs and symptoms should<br />

direct the diagnostic evaluation of the patient with<br />

acute infectious diarrhea. With a history of travel<br />

to developing nations, E. coli, Salmonella, Shigella,<br />

Campylobacter, cholera, Entamoeba histolytica,<br />

and Giardia lamblia should be high on the differential.<br />

Vomiting after the ingestion of fast foods,<br />

canned products, or raw seafood and meats should<br />

prompt the clinician to look for toxin-producing<br />

enteropathogens associated with food poisoning,<br />

such as Staphylococcus aureus and Bacillus cereus,<br />

as well as hepatitis A, parasites (tapeworms,<br />

flukes, trichinae), Salmonella and E. coli. The<br />

hospitalized patient may experience diarrhea not<br />

only from C. difficile, but also from procedures and<br />

medications (such as antibiotics, antacids, and<br />

medications with a high osmolality). The immunocompromised<br />

host presents a special challenge in<br />

the work-up of acute diarrhea, in the face of<br />

polypharmacy and malabsorpsion due to enteropathy<br />

and pancreatic insufficiency (see Chapter 8).<br />

With potential multiple pathogens, gastrointestinal<br />

endoscopy with biopsy and aspiration of fluid and<br />

fecal contents may give the highest yield of diagnosis<br />

in the immunocompromised patient.<br />

Direct examination of the stool for the presence of<br />

mucus, blood, or leukocytes may be helpful in<br />

classifying infectious agents. For example,<br />

profusely watery stools without mucus, blood or<br />

leukocytes are characteristic of cholera,<br />

Salmonella, ETEC, C. parvum, Giardia and most<br />

viral agents. The presence of mucus, blood and<br />

leukocytes in the stool are more consistent with<br />

inflammatory infections such as Campylobacter,<br />

EIEC, Shigella, C. difficile, Yersinia, Entamoeba<br />

and cytomegalovirus.<br />

General guidelines for treatment of<br />

diarrheal diseases<br />

As outlined in the various sections of this chapter<br />

(see also Chapter 37), the treatment of the majority<br />

of infectious diarrheal episodes is supportive, with<br />

ORS representing the cardinal intervention to<br />

minimize life-threatening dehydration, particularly<br />

in young children. 105 Breast feeding should<br />

be continued, as it may confer protection.<br />

Intravenous rehydration should be reserved for<br />

high-risk patients who are unable to tolerate enterals<br />

due to recurrent vomiting or diminished<br />

mental status. The use of antimotility agents, such<br />

as bismuth subsalicylate (Pepto-Bismol ® ), loperamide<br />

(Imodium ® ), and atropine sulfate with diphenoxylate<br />

hydrochloride (Lomotil ® ) should be<br />

discouraged, owing to the possible risk of salicylate<br />

intoxication, ileus, toxic megacolon, bowel<br />

perforation, and HUS in subjects infected with<br />

EHEC. Probiotics, such as Lactobacillus GG, have<br />

been recently shown to be effective both in the<br />

prevention, and in the treatment of viral<br />

(rotavirus) and antibiotic-associated (C. difficile)<br />

diarrheas, as well documented by two recent metaanalyses.<br />

106,107


REFERENCES<br />

1. Barua D, Greenough WB III. Cholera, 1st edn. New<br />

York: Plenum Publishing, 1992.<br />

2. Wachsmuth IK, Blake PA, Olsvik O. Vibrio cholerae and<br />

Cholera: Molecular and Perspectives, 1st edn.<br />

Washington, DC: American Society for Microbiology,<br />

1994.<br />

3. Nataro JP, Levine MM. Enteric bacterial vaccines,<br />

Salmonella, Shigella, cholera, Escherichia coli. In Ogra<br />

P et al, eds. Mucosal Immunology, 2nd edn. San Diego:<br />

Academic Press, 1999: 851–866.<br />

4. Clemens JD, Sack DA, Harris JR et al. Field trial of oral<br />

cholera vaccines in Bangladesh. Lancet. 1986: 2:<br />

124–127.<br />

5. Trach DD, Clemens JD, Ke NT et al. Field trial of a<br />

locally produced, killed, oral cholera vaccine in<br />

Vietnam. Lancet 1997; 349: 231–235.<br />

6. Levine MM, Kaper JB, Herrington D et al. Safety,<br />

immunogenicity, and efficacy of recombinant live oral<br />

cholera vaccines, CVD103 and CVD103-HgR. Lancet<br />

1988; 2: 467–470.<br />

7. Suharyono PM, Simanjuntak C, Witham N et al. Safety<br />

and immunogenicity of single-dose live oral cholera<br />

vaccine CVD103-HgR in 5–9-year-old Indonesian children.<br />

Lancet 1992; 340: 689–694.<br />

8. Tacket CO, Losonsky G, Nataro JP et al. Onset and duration<br />

of protective immunity in challenged volunteers<br />

after vaccination with live oral cholera vaccine<br />

CVD103-HgR. J Infect Dis 1992; 166: 837–841.<br />

9. Tacket CO, Losonsky G, Nataro JP et al. Initial clinical<br />

studies of CVD112 Vibrio cholerae O139 live oral<br />

vaccine: safety and efficacy against experimental challenge.<br />

J Infect Dis 1995; 172: 883–886.<br />

10. Coster TS, Killeen KP, Waldor MK et al. Safety,<br />

immunogenicity, and efficacy of live attenuated Vibrio<br />

cholerae O139 vaccine prototype. Lancet 1995; 345:<br />

949–952.<br />

11. West PA, Brayton PR, Twilley RR, et al. Numerical<br />

tazonom of nitrogen-fixing ‘decarboxylase-negative’<br />

Vibrio species isolated from aquatic environments. Int J<br />

Syst Bacteriol 1985; 35: 198–205.<br />

12. Morris JG Jr, Wilson R, Davis BR et al. Non-O group 1<br />

Vibrio cholerae gastroenteritis in the United States:<br />

clinical, epidemiologic and laboratory characteristics of<br />

sporadic cases. Ann Intern Med 1981; 94: 656–658.<br />

13. Edelman R, Levine MM. Summary of an international<br />

workshop on typhoid fever. Rev Infect Dis 1986; 8:<br />

329–349.<br />

14. Hoffman TA, Ruiz CJ, Counts GW. Water-borne typhoid<br />

fever in Dade County, FL: clinical and therapeutic<br />

evaluations of 105 bacteremic patients. Am J Med 1975;<br />

59: 481–487.<br />

15. Centers for Disease Control and Prevention:<br />

Surveillance for foodborne-disease outbreaks, United<br />

States, 1988–1992. Morbid Mortal Weekly Rep 1996; 45:<br />

1S–66S.<br />

16. Woodward TE, Smadel JE, Ley HL et al. Preliminary<br />

report on the beneficial effect of chloromycetin in the<br />

treatment of typhoid fever. Ann Intern Med 1948; 29:<br />

131–134.<br />

17. Levine MM, Ferreccio C, Cryz S, Ortiz E. Comparison<br />

of enteric-coated capsules and liquid formulation of<br />

Ty21 a typhoid vaccine in randomized controlled field<br />

trial. Lancet 1990; 336: 891–894.<br />

18. Shiga K. Ueber den Dysenteriebacillus (Bacillus<br />

dysenteriae). Zentralbl Bakteriol Parasit Kde Abt I Orig<br />

1898; 24: 817–824.<br />

References 157<br />

19. DuPont HL, Levine MM, Hornick RB, Formal SB.<br />

Inoculum size in shigellosis and implications for<br />

expected mode of transmission. J Infect Dis 1989; 159:<br />

1126–1128.<br />

20. Institute of Medicine. The prospects for immunizing<br />

against Shigella spp. In New Vaccine Development.<br />

Establishing Priorities: Diseases of Importance in<br />

Developing Countries, vol 2. Washington DC: National<br />

Academy Press, 1986: 329–337.<br />

21. Blaser MJ, Reller LB. Campylobacter enteritis. N Engl J<br />

Med 1981; 305:1444–1452.<br />

22. Deming MS, Tauxe RV, Blake PA et al. Campylobacter<br />

enteritis at a university: transmission from eating<br />

chicken and from cats. Am J Epidemiol 1987; 126:<br />

526–534.<br />

23. Wood RC, Macdonald KL, Osterholm MT.<br />

Campylobacter enteritis outbreaks associated with<br />

drinking raw milk during youth activities: a 10-year<br />

review of outbreaks in the United States. JAMA 1992;<br />

268: 3228–3230.<br />

24. Mentzing LO. Waterborne outbreaks of Campylobacter<br />

enteritis in central Sweden. Lancet 1981; 2: 352–354.<br />

25. Metchock B, Londway DR, Carter GP et al. Yersinia<br />

enterocolitica: a frequent seasonal stool isolate from<br />

children at an urban hospital in the southeast United<br />

States. J Clin Microbiol 1991; 29: 2868–2869.<br />

26. Marks MI, Pai CH, Lafleur L et al. Yersinia enterocolitica<br />

gastoenteritis: a prospective study of clinical,<br />

bacteriologic and epidemiologic features. J Pediatr 1980;<br />

96: 26–31.<br />

27. Tacket CO, Davis BR, Carter GP et al. Yersinia<br />

enterocolitica pharyngitis. Ann Intern Med 1983; 99:<br />

40–42.<br />

28. Cover TL. Yersinia enterocolitica and Yersinia<br />

pseudotuberculosis. In Blaser MJ et al, eds. Infections of<br />

the Gastrointestinal Tract. New York: Raven Press, 1995:<br />

811–823.<br />

29. Levine MM. Escherichia coli that cause diarrhea:<br />

enterotoxigenic, enteropathogenic, enteroinvasive and<br />

enteroadherent. J Infect Dis 1987; 155: 377–389.<br />

30. Paulozzi LJ, Johnson KE, Kamahele LM et al. Diarrhea<br />

associated with adherent enteropathogenic Escherichia<br />

coli in infant and toddler center, Seattle, Washington.<br />

Pediatrics 1986; 77: 296–300.<br />

31. Donnenberg MS, Kaper JB. Enteropathogenic<br />

Escherichia coli. Infect Immun 1992; 60: 3953–3961.<br />

32. Levine MM, Bergquist EJ, Nalin DR et al. Escherichia<br />

coli strains that cause diarrhoea but do not produce<br />

heat-labile or heat-stable enterotoxins and are noninvasive.<br />

Lancet 1978; 1: 1119–1122.<br />

33. Clausen CR, Christie DL. Chronic diarrhea in infants<br />

caused by adherent enteropathogenic, Escherichia coli. J<br />

Pediatr 1982; 100: 358–361.<br />

34. Nelson JD. Duration of neomycin for enteropathogenic<br />

Escherichia coli diarrheal disease: a comparative study<br />

of 113 cases. Pediatrics 1971; 48: 248–258.<br />

35. Thoren AL. Antibiotic sensitivity of enteropathogenic<br />

Escherichia coli to mecillinam,<br />

trimethoprim–sulfamethoxazole and other antibiotics.<br />

Acta Pathol Microbiol Scand 1980; 88: 265–268.<br />

36. Gorbach SL, Banwell JG, Chatterjee BD et al. Acute<br />

undifferentiated human diarrhea in the tropics, vol 1.<br />

Alterations in intestinal microflora. J Clin Invest 1971;<br />

50: 881–889.<br />

37. Black RE, Merson MH, Huq I et al. Incidence and<br />

severity of rotavirus and Escherichia coli diarrhoea in


158<br />

Bacterial infections<br />

rural Bangladesh: implications for vaccine development.<br />

Lancet 1981; 1: 141–143.<br />

38. Institute of Medicine. New Vaccine Development:<br />

Establishing Priorities, vol 2, Diseases of Importance in<br />

Developing Countries. Washington, DC: National<br />

Academy Press, 1986.<br />

39. Sack RB. Travelers’ diarrhea: microbiologic bases for<br />

prevention and treatment. Rev Infect Dis 1990; 12:<br />

59S–63S.<br />

40. Black RE, Levine MM, Clements ML et al. Treatment of<br />

experimentally induced enterotoxigenic Escherichia coli<br />

diarrhea with trimethoprim, trimethoprim–sulfamethoxazole<br />

or placebo. Rev Infect Dis 1982; 4: 540–545.<br />

41. DuPont HL, Ericsson CD. Prevention and treatment of<br />

traveler’s diarrhea. N Engl J Med 1993; 328: 1821–1827.<br />

42. Bandres JC, Mathewson JJ, Ericsson CD, Dupont HL.<br />

Trimethoprim/sulfamethoxazole remains active against<br />

enterotoxigenic Escherichia coli and Shigella species in<br />

Guadalajara, Mexico. Am J Med Sci 1992; 303: 289–291.<br />

43. Wolfe MD. Protection of travelers. Clin Infect Dis 1997;<br />

25: 177–184.<br />

44. Peltola H, Siitonen A, Kyronseppa H et al. Prevention of<br />

travellers’ diarrhoea by oral B-subunit/whole-cell<br />

cholera vaccine. Lancet 1991; 338: 1285–1289.<br />

45. Svennerholm AM, Ahren C, Jertborn M. Vaccines<br />

against enterotoxogenic Escherichia coli infections: I.<br />

Oral inactivated vaccines against enterotoxigenic<br />

Escherichia coli. In Levine MM et al, eds. New<br />

Generation Vaccines, 2nd edn. New York: Marcel<br />

Dekker, 1997: 865–873.<br />

46. Echeverria P, Sethabutr O, Pitarangsi C. Microbiology<br />

and diagnosis of infections with Shigella and enteroinvasive<br />

Escherichia coli. Rev Infect Dis 1991; 13:<br />

220S–225S.<br />

47. Macdonald KL, Eidson M, Strohmeyer C et al. A<br />

multistate outbreak of gastrointestinal illness caused by<br />

enterotoxigenic Escherichia coli in imported semisoft<br />

cheese. J Infect Dis 1985; 151: 716–720.<br />

48. DuPont HL, Formal SB, Hornick RB et al. Pathogenesis<br />

of Escherichia coli diarrhea. N Engl J Med 1971; 285:<br />

1–9.<br />

49. Snyder JD, Wells JG, Yashuck et al. Outbreak of invasive<br />

Escherichia coli gastroenteritis on a cruise ship. Am J<br />

Trop Med Hyg 1984; 33: 281–284.<br />

50. Tarr PI. Escherichia coli O157:H7 clinical, diagnostic<br />

and epidemiological aspects of human infection. Clin<br />

Infect Dis 1995; 20: 9–10.<br />

51. Riley LW, Remis RS, Helgerson SD et al. Hemorrhagic<br />

colitis associated with a rare Escherichia coli serotype.<br />

N Engl J Med 1983; 308: 681–685.<br />

52. Karmali MA, Petric M, Lim C et al. The association<br />

between idiopathic hemolytic uremic syndrome and<br />

infection by verotoxin-producing Escherichia coli. J<br />

Infect Dis 1985; 151: 775–782.<br />

53. Centers for Disease Control and Prevention. Isolation of<br />

E. coli 157:H7 from sporadic cases of hemorrhagic<br />

colitis, United States. Morbid Mortal Weekly Rep 1997;<br />

46: 700–704.<br />

54. Besser RE, Lett SM, Weber JT et al. An outbreak of<br />

diarrhea and hemolytic uremic syndrome from<br />

Escherichia coli O157:H7 in fresh-pressed apple cider.<br />

JAMA 1993; 269: 2217–2220.<br />

55. Watanabe H, Guerrant RL. Summary: Nagasaki<br />

enterohemorrhagic Escherichia coli meeting and<br />

workshop. J Infect Dis 1997; 176: 247–249.<br />

56. Keene WE, Hedberg K, Herriott DE et al, A prolonged<br />

outbreak of Escherichia coli O157:H7 infections caused<br />

by commercially distributed raw milk. J Infect Dis 1997;<br />

176: 815–818.<br />

57. Tilden J Jr, Young W, McNamara AM et al. A new route<br />

of transmission for Escherichia coli: infection from dry<br />

fermented salami. Am J Public Health 1996; 86:<br />

1142–1145.<br />

58. Swerdlow DL, Woodruff BA, Brady RC et al. A<br />

waterborne outbreak in Missouri of Escherichia coli<br />

O157:H7 associated with bloody diarrhea and death.<br />

Ann Intern Med 1992; 117: 812–819.<br />

59. Keene W, McAnulty JM, Hoesly FC et al. A swimmingassociated<br />

outbreak of hemorrhagic colitis caused by<br />

Escherichia coli O157:H7 and Shigella sonnei. N Engl J<br />

Med 1994; 331: 579–584.<br />

60. Belongia EA, Osterholm MT, Soler JT et al.<br />

Transmission of Escherichia coli O157:H7 infection in<br />

Minnesota child day-care facilities. JAMA 1993; 269:<br />

883–888.<br />

61. Slutsker L, Ries AA, Greene KD et al. Escherichia coli<br />

O157:H7 diarrhea in the United States: clinical and<br />

epidemiologic features. Ann Intern Med 1997; 126:<br />

505–513.<br />

62. Macdonald KL, O’Leary MJ, Cohen ML et al. Escherichia<br />

coli O157:H7, an emerging gastrointestinal pathogen:<br />

results of a one-year, prospective, population-based<br />

study. JAMA 1988; 59: 3567–3570.<br />

63. Griffin PM et al. Large outbreak of Escherichia coli<br />

O157:H7 infections in the western United States: the<br />

big picture. In Karmali MA, Goglio AG, eds. Recent<br />

Advances in Verocytotoxin-producing Escherichia coli<br />

Infection. Amsterdam: Elsevier Science BV, 1994: 7–12.<br />

64. Griffen PM, Ostroff SM, Tauxe RV et al. Illnesses<br />

associated with Escherichia coli O157:H7 infections: a<br />

broad clinical spectrum. Ann Intern Med 1988; 109:<br />

705–712.<br />

65. Pai Ch, Ahmed N, Lior H et al. Epidemiology of<br />

sporadic diarrhea due to verocytotoxin-producing<br />

Escherichia coli: a two-year prospective study. J Infect<br />

Dis 1988; 157: 1054–1057.<br />

66. Boyce TG, Swerdlow DL, Griffin PM. Escherichia coli<br />

O157:H7 and the hemolytic-uremic syndrome. N Engl J<br />

Med 1995; 333: 364–368.<br />

67. Uc A, Mitros FA, Kao SC, Sanders KD.<br />

Pseudomembranous colitis with Escherichia coli<br />

O157:H7. J Pediatr Gastroenterol Nutr 1997; 24:<br />

590–593.<br />

68. Pavia AT, Nichols CR, Green DP et al. Hemolytic-uremic<br />

syndrome during an outbreak of Escherichia coli<br />

O157:H7 infections in institutions for mentally retarded<br />

persons: clinical and epidemiologic observations. J<br />

Pediatr 1990; 116: 544–551.<br />

69. Smith HR, Cheasty T, Rowe B. Enteroaggregative<br />

Escherichia coli and outbreaks of gastroenteritis in UK.<br />

Lancet 1997; 350: 814–815.<br />

70. Cimolai N, Carter JE, Morrison BJ, Anderson JD. Risk<br />

factors for the progression of Escherichia coli O157:H7<br />

enteritis to hemolytic-uremic sydrome. J Pediatr 1990;<br />

116: 589–592.<br />

71. Tarr PI, Neill MA, Clausen CR et al. Escherichia coli<br />

O157:H7 and the hemolytic uremic syndrome:<br />

importance of early cultures in establishing the etiology.<br />

J Infect Dis 1990; 162: 553–556.<br />

72. Proulx F, Turgeon JP, Delage G et al. Randomized,<br />

controlled trial of antibiotic therapy for Escherichia coli<br />

O157:H7 enteritis. J Pediatr 1992; 121: 299–303.<br />

73. Carter AO, Borczyk AA, Carlson JA et al. A severe<br />

outbreak of Escherichia coli O157:H7-associated<br />

hemorrhagic colitis in a nursing home. N Engl J Med<br />

1987; 317: 1496–1500.<br />

74. Bosworth BT, Samuel JE, Moon HW et al. Vaccination<br />

with genetically modified Shiga-like toxin IIe prevents<br />

edema disease in swine. Infect Immun 1996; 64: 55–60.


75. Butterton JR, Ryan ET, Acheson DW, Calderwood SB.<br />

Coexpression of the B subunit of Shiga toxin 1 and<br />

EaeA from enterohemorrhagic Escherichia coli in Vibrio<br />

cholerae vaccine strains. Infect Immun 1997; 65:<br />

2127–2135.<br />

76. Konadu E, Robbins JB, Shiloach J et al. Preparation,<br />

characterization and immunological properties in mice<br />

of Escherichia coli O157 O-specific polysaccharideprotein<br />

conjugate vaccines. Infect Immun 1994; 62:<br />

5048–5054.<br />

77. Bhan MK, Raj P, Levine MM et al. Enteroaggregative<br />

Escherichia coli associated with persistent diarrhea in a<br />

cohort of rural children in India. J Infect Dis 1989; 159:<br />

1061–1064.<br />

78. Bhan MK, Khoshoo V, Sommerfelt H et al.<br />

Enteroaggregative Escherichia coli and Salmonella<br />

associated with nondysenteric persistent diarrhea.<br />

Pediatr Infect Dis J 1989; 8: 499–502.<br />

79. Cravioto A, Tello A, Navarro A et al. Association of<br />

Escherichia coli Hep-2 adherence patterns with type and<br />

duration of diarrhoea, Lancet 1991; 337: 262–264.<br />

80. Giron JA, Jones T, Millan-Velasco F et al. Diffuseadhering<br />

Escherichia coli (DAEC) as a putative cause of<br />

diarrhea in Mayan children in Mexico. J Infect Dis 1991;<br />

163: 507–513.<br />

81. Levine MM, Ferreccio C, Prado V et al. Epidemiologic<br />

studies of Escherichia coli diarrheal infections in a low<br />

socioeconomic level peri-urban community in Santiago,<br />

Chile. Am J Epidemiol 1993; 138: 849–69.<br />

82. Baqui AH, Sack RB, Black RE et al. Enteropathogens<br />

associated with acute and persistent diarrhea in<br />

Bangladeshi children < 5 years of age. J Infect Dis 1992;<br />

166: 792–796.<br />

83. Nataro JP, Deng Y, Cookson S et al. Heterogeneity of<br />

enteroaggregative Escherichia coli virulence<br />

demonstrated in volunteers. J Infect Dis 1995; 171:<br />

465–468.<br />

84. Henry FJ, Udoy AS, Wanke CA, Aziz KMA.<br />

Epidemiology of persistent diarrhea and etiologic agents<br />

in Mirzapur, Bangladesh. Acta Paediatr Suppl 1996; 381:<br />

27–31.<br />

85. Bhatnagar S, Bhan MK, Sommerfelt H et al.<br />

Enteroaggregative Escherichia coli may be a new<br />

pathogen causing acute and persistent diarrhea. Scand J<br />

Infect Dis 1993; 25: 579–583.<br />

86. Fang GD, Lima AA, Martins CV et al. Etiology and<br />

epidemiology of persistent diarrhea in northeastern<br />

Brazil: a hospital-based, prospective, case–control study.<br />

J Pediatr Gastroenterol Nutr 1995; 21: 137–144.<br />

87. Nataro JP. Enteroaggregative Escherichia coli. Alpe Adria<br />

Microbiol J 1998; 7: 265–273.<br />

88. Pai M, Kang G, Ramakrishna BS et al. An epidemic of<br />

diarrhoea in south India caused by enteroaggregative<br />

Escherichia coli. Indian J Med Res 1997; 106: 7–12.<br />

89. Cobeljic M, Miljkovic-Selimovic B, Paunovic-Todosijevic<br />

D et al. Enteroaggregative Escherichia coli associated<br />

with an outbreak of diarrhoea in a neonatal nursery<br />

ward. Epidemiol Infect 1996; 117: 11–16.<br />

90. Itoh Y, Nagano I, Kunishima M, Ezaki T. Laboratory<br />

investigation of enteroaggregative Escherichia coli O<br />

References 159<br />

untypeable:H10 associated with a massive outbreak of<br />

gastrointestinal illness. J Clin Microbiol 1997; 35:<br />

2546–2550.<br />

91. Paul M, Tsukamoto T, Ghosh AR et al. The significance<br />

of enteroaggregative Escherichia coli in the etiology of<br />

hospitalized diarrhoea in Calcutta, India and the<br />

demonstration of a new honey-combed pattern of<br />

aggregative adherence. FEMS Microbiol Lett 1994; 117:<br />

319–326.<br />

92. Steiner TS, Lima AAM, Nataro JP, Guerrant RL.<br />

Enteroaggregative Escherichia coli produce intestinal<br />

inflammation and growth impairment and cause<br />

interleukin-8 release from intestinal epithelial cells. J<br />

Infect Dis 1998; 177: 88–96.<br />

93. Bouzari S, Jafari A, Farhoudi-Moghaddam AA et al.<br />

Adherence of non-enteropathogenic Escherichia coli to<br />

HeLa cells. J Med Microbiol 1994; 40: 95–97.<br />

94. Schmidt H, Knop C, Franke S et al. Development of PCR<br />

for screening of enteroaggregative Escherichia coli. J<br />

Clin Microbiol 1995; 33: 701–705.<br />

95. Yamamoto T, Echeverria P, Yokota T. Drug resistance and<br />

adherence to human intestines of enteroaggregative<br />

Escherichia coli. J Infect Dis 1992; 165: 744–749.<br />

96. McFarland LV, Mulligan ME, Kwok RYY, Stamm WE.<br />

Nosocomial acquisition of Clostridium difficile infection.<br />

N Engl J Med 1989; 320: 204–210.<br />

97. Kaatz GW, Gitlin SD, Schaberg DR et al. Acquisition of<br />

Clostridium difficile from the hospital environment. Am<br />

J Epidemiol 1988; 127: 1289–1294.<br />

98. McFarland LV, Surawicz CM, Stamm WE. Risk factors<br />

for Clostridium difficile carriage and C. difficileassociated<br />

diarrhea in a cohort of hospitalized patients.<br />

J Infect Dis 1990; 162: 678–684.<br />

99. Donta ST, Myers MG. Clostridium difficile toxin in<br />

asymptomatic neonates, J Pediatr 1982; 100: 431–434.<br />

100. Borriello SP. Pathogenesis of Clostridium difficile<br />

infection. J Antimicrob Chemother 1998; 41: 13–19.<br />

101. Batts DH, Martin D, Holmes R et al. Treatment of<br />

antibiotic-associated Clostridium difficile diarrhea with<br />

oral vancomycin. J Pediatr 1980; 97: 151–153.<br />

102. Bolton RP, Culshaw MA. Maecal metronidazole<br />

concentrations during oral and intravenous therapy for<br />

antibiotic colitis due to Clostridium difficile. Gut 1986;<br />

27: 1169–1172.<br />

103. Gorbach SL, Chang T-W, Foldin B. Successful treatment<br />

of relapsing Clostridium difficile colitis with<br />

Lactobacillus GG. Lancet 1987; 2: 1519.<br />

104. Surawicz CM, Elmer GW, Speelman P et al. Prevention<br />

of antibiotic-associated diarrhea by Saccharomyces<br />

boulardii: a prospective study. Gastroenterology 1989;<br />

96: 981–988.<br />

105. Guandalini S. The treatment of acute diarrhea in the<br />

third millennium: a pediatrician’s perspective. Acta<br />

Gastroenterol Belgn 2002; 65: 33–6.<br />

106. Huang JS, Bousvaros A, Lee JW et al. Efficacy of<br />

probiotic use in acute diarrhea in children: a metaanalysis.<br />

Dig Dis Sci 2002; 47: 2625–2634.<br />

107. D’Souza AL, Rajkumar C, Cooke J, Bulpitt CJ. Probiotics<br />

in prevention of antibiotic associated diarrhoea: metaanalysis.<br />

BMJ 2002; 324:1361–1366.


11<br />

Introduction<br />

Intestinal parasites<br />

David Brewster<br />

The parasites of humans are classified into<br />

five major divisions: Protozoa (amebae, flagellates,<br />

ciliates, sporozoans, coccidia, microsporidia);<br />

Platyhelminths (cestodes, trematodes), Acanthocephela<br />

(thorny headed worms); Nematodes<br />

(roundworms) and Arthropods (insects, spiders,<br />

mites, ticks). Geohelminths are a sub-group of soiltransmitted<br />

intestinal nematodes with similar<br />

epidemiological characteristics. They include<br />

Strongyloides, hookworm, Ascaris and Trichuris. 1<br />

In this chapter, we focus on the common protozoa,<br />

nematodes and trematodes which affect the intestine<br />

(Table 11.1). Out of the large numbers of children<br />

infected by these parasites in the developing<br />

world, most are asymptomatic and do not present<br />

to the health services for treatment.<br />

The World Health Organization (WHO) estimates<br />

that some 3.5 billion people worldwide are<br />

infected by intestinal parasitic and protozoan<br />

infections, including 350 million with morbidity<br />

from ascariasis, 220 million from trichuris and 150<br />

million from hookworm. 2 A nationwide survey of<br />

intestinal parasites in China during 1988–92 on<br />

1.5 million people reported a prevalence of 62.6%,<br />

of whom 43.3% had multiple parasites. The five<br />

common parasites were Ascaris lumbricoides<br />

(47.0%), Enterobius vermicularis (26.4%), Trichuris<br />

trichiura (18.8%), Giardia lamblia (2.5%) and<br />

Entamoeba histolytica (0.9%). 3 In sub-Saharan<br />

African schoolchildren, on the other hand, estimates<br />

of the prevalence of helminth infections<br />

were: hookworms 32.1%, Ascaris 30.2%, Trichuris<br />

29.5% and Schistosoma mansoni 14.0%. 4 These<br />

prevalence rates have been fairly stable over the<br />

past 50 years with only a modest decrease in<br />

ascariasis. However, higher rates have been<br />

reported, such as surveys reporting that only 14%<br />

of Tanzanian and 37% of Ghanaian children were<br />

free from worm infections. 5 Intestinal parasites<br />

also occur in industrialized countries, with a third<br />

of 5792 fecal specimens positive for parasites in<br />

the USA in 2000, peaking between July and<br />

October. 6<br />

The peak age for intestinal parasites tends to be<br />

school-age children but overt sickness from them<br />

is the exception. A large Tanzanian morbidity<br />

survey of schoolchildren found that no sign or<br />

symptom was significantly associated with any<br />

helminth infection, only blood in stool and<br />

hepatomegaly with Schistosoma mansoni. 7 The<br />

major determinant of morbidity from parasitic<br />

diseases is the intensity of infection, but this is<br />

rarely reported in children. A Zairean study found<br />

that the highest mean intensities (measured as<br />

eggs per gram of feces (epg)) of Ascaris and<br />

Trichuris were among 2–4-year-olds, whereas<br />

school-age children excreted the most hookworm<br />

ova. 8 Using disability-adjusted life years lost<br />

(DALY), WHO estimated that intestinal helminthiasis<br />

caused 39 million DALYs, with 57% attributed<br />

to hookworm, 27% to ascariasis and 16% to<br />

trichurasis. 2<br />

The most common symptoms of intestinal parasites<br />

are diarrhea and/or failure to thrive from<br />

anorexia and malabsorption. The protozoa Giardia<br />

lamblia and Cryptosporidium parvum are common<br />

causes of diarrhea in children, but other protozoa<br />

such as Cyclospora cayetanensis, Entamoeba<br />

histolytica, and Microsporidia species may also<br />

cause diarrhea, and these include Isospora belli in<br />

immunodeficient (e.g. HIV-seropositive) patients.<br />

Only five helminthic parasites are associated with<br />

diarrheal disease: Strongyloides stercoralis,<br />

Trichuris trichiura, Schistosoma mansoni,<br />

Trichinella spiralis and Capillaria philippinensis. 9<br />

Strictly intraluminal worms (e.g. Ascaris lumbricoides)<br />

only rarely affect intestinal function<br />

161


162<br />

Table 11.1 Classification of intestinal parasites<br />

Intestinal parasites<br />

Infection Other name Symptoms Transmission Alternative treatments Treatment of choice<br />

PROTOZOA<br />

Intestinal amebae<br />

Entamoeba histolytica amebiasis dysentery fecal–oral metronidazole<br />

Entamoeba dispar asymptomatic fecal–oral nil<br />

Flagellates<br />

Giardia lamblia (duodenale giardiasis chronic diarrhea, fecal–oral metronidazole tinidazole<br />

or intestinalis) malabsorption<br />

Ciliates<br />

Balantidium coli asymtomatic fecal–oral nil<br />

Coccidia<br />

Cryptosporidium parvum cryptosporidiosis persistent diarrhea fecal–oral nitazoxanide*<br />

Cyclospora cayetanensis cyclosporiasis diarrhea fecal–oral co-trimoxazole<br />

Isospora belli isosporiasis diarrhea with AIDS fecal–oral co-trimoxazole<br />

NEMATODES<br />

Ascaris lumbricoides roundworm intestinal obstruction fecal–oral levamisole, pyrantel albendazole †<br />

Enterobius vermicularis pinworm, threadworm nocturnal anal pruritis fecal–oral levamisole, pyrantel albendazole †<br />

Ancylostoma duodenale hookworm iron deficiency anemia percutaneous levamisole, pyrantel albendazole †<br />

Necator americanus hookworm iron deficiency anemia percutaneous levamisole, pyrantel albendazole †<br />

Strongyloides stercoralis strongyloidiasis diarrhea percutaneous albendazole ivermectin<br />

Trichuris trichiura whipworm, trichuriasis dysentry, rectal fecal–oral levamisole, pyrantel albendazole †<br />

prolapse (rare)<br />

CESTODES<br />

Hymenolepis nana dwarf tapeworm asymptomatic fecal–oral praziquantel nitazoxanide*<br />

TREMATODES<br />

Schistosoma mansoni bilharzia melena, portal percutaneous oxamniquine praziquantel<br />

hypertension<br />

Fasciolopsis buski giant intestinal fluke percutaneous praziquantel<br />

*Nitazoxanide is the drug of choice (if available) where treatment is indicated; †or mebendazole


adversely when the intensity of infection is high.<br />

Infections such as giardiasis, cryptosporidiosis and<br />

severe trichuriasis may contribute to growth<br />

failure via malabsorption, so their resolution or<br />

treatment should lead to catch-up growth.<br />

Malnutrition appears to predispose children to an<br />

acute diarrheal syndrome in strongyloidiasis,<br />

which may then exacerbate the nutritional status<br />

through anorexia and enteropathy. 10 Owing to the<br />

prolonged carriage of many parasites, only<br />

Strongyloides and Cryptosporidium have a consistently<br />

significant association with diarrheal<br />

disease when compared to non-diarrheal controls<br />

(Table 11.2).<br />

Giardiasis<br />

Introduction<br />

Giardia lamblia (also called intestinalis or duodenale)<br />

is a flagellate protozoan with two stages:<br />

motile trophozoites (Figure 11.1) and cysts.<br />

Life cycle<br />

Infection may result from ingesting cysts, which<br />

release trophozoites in the upper small intestine,<br />

attaching to the mucosa by a ventral disc. Giardia<br />

trophozoites undergo significant biological<br />

changes to survive outside the host by<br />

differentiating into infective cysts, which is<br />

promoted by exposure to conjugated bile salts and<br />

Figure 11.1 Giardia lamblia trophozoite. Courtesy of UK<br />

Tropical International Health.<br />

Table 11.2 Parasites in diarrhea in children: case–control studies<br />

Guinea-Bissau 16 Bangladesh 207 Five Countries †208 Tropical Australia 10 South Africa 209 Total<br />

Cases Controls Cases Controls Cases Controls Cases Controls Cases Controls Cases Controls<br />

n 1219 511 814 814 3640 3279 290 85 373 371 6336 5060<br />

Giardia 233 128 7 23 109 98 9 5 24 22 6.0% 5.5%<br />

Cryptosporidium* 69 11 11 3 20 1 9 2 4.0% 1.0%<br />

Strongyloides* 54 15 21 2 5.0% 2.9%<br />

Entamoeba 50 31 5 1 11 3 0 0 1 0 1.1% 0.7%<br />

Ascaris 7 1 0 0 40 53 2.5% 5.6%<br />

Hookworm 31 28 1 0 2.1% 4.7%<br />

Trichuris 5 2 22 4 13 23 2.1% 3.0%<br />

Hymenolepis 2 0 2 4 1 3 0.3% 0.7%<br />

Giardiasis 163<br />

*Significantly associated with diarrhea, odds ratio 4.4 (95% CI 2.6–7.6) for Cryptosporidium and 1.8 (95% CI 1.0–3.3) for Strongyloides<br />

†China, India, Mexico, Myanmar, Pakistan


164<br />

Intestinal parasites<br />

low levels of cholesterol. 11,12 A specific proteolytic<br />

event caused by a constitutively expressed<br />

membrane-associated dipeptidyl peptidase IV is<br />

necessary for encystation. 13 The incubation period<br />

is 1–2 weeks.<br />

Epidemiology<br />

Giardia lamblia is one of the most common parasitic<br />

infections in humans with a prevalence of<br />

2–5% in industrialized countries, and high transmission<br />

rates in day-care center outbreaks among<br />

12–36-month-old children who are not toilet<br />

trained. 14,15 In developing countries, prevalences<br />

of 20–30% occur with up to 60% of children<br />

infected at some time during childhood. Since<br />

cysts are excreted for prolonged periods, community<br />

studies often find higher rates of infection in<br />

children without diarrhea than in diarrheal<br />

cases. 16 Giardia accounts for about 5% of travelers’<br />

diarrhea and is more common in children with<br />

immunodeficiency, although not HIV infection.<br />

The main routes of transmission are water, food<br />

and fecal–oral. Infection may occur after the ingestion<br />

of only 10–100 cysts and unfiltered water has<br />

been a common source of outbreaks in developed<br />

countries.<br />

A Brazilian 4-year cohort study of 157 children<br />

from an urban slum documented substantial<br />

morbidity from giardiasis, with a similar frequency<br />

between diarrheal and non-diarrheal cases (9.7%<br />

vs. 7.4%), an association with persistent diarrhea<br />

(20.6% of episodes) and recurrent or relapsing<br />

infections (46%) and poorer nutritional status for<br />

those with symptomatic infections. 17 A study of a<br />

rural Egyptian cohort of 152 infants documented<br />

4.5 episodes per child-year with a mean duration<br />

of excretion of 7.2 weeks, and reduced risk in<br />

breast-fed infants, particularly for symptomatic<br />

illness. 18 A Kenyan cohort study of 84 rural children<br />

aged 10–28 months reported a 44.7% prevalence,<br />

an estimated incidence of 2.8 episodes per<br />

year and a duration of 75.3 days per child for giardiasis.<br />

19<br />

Pathophysiology<br />

Despite intensive investigations, the exact mechanisms<br />

by which Giardia causes diarrhea and<br />

malabsorption are still unclear. There is evidence<br />

of a secretory mechanism, malabsorption,<br />

decreased brush-border surface area and interference<br />

with intraluminal digestion. Another possible<br />

mechanism for diarrhea is small-bowel bacterial<br />

overgrowth, but mucosal invasion is uncommon.<br />

Duodenal morphology may be normal, but partial<br />

or even total villus atrophy may occur, as well as a<br />

mucosal inflammatory response with increased<br />

intraepithelial lymphocytes. Giardia parasites are<br />

able to adhere to the gut mucosa and may cause a<br />

direct effect on intestinal function by disrupting<br />

the brush border and its enzymatic system.<br />

The effect of giardiasis on intestinal structure and<br />

function is quite variable, but animal and in vitro<br />

studies have documented villus atrophy, malabsorption,<br />

increased paracellular permeability and<br />

reduced brush-border enzymes. 20,21 Giardia<br />

trophozoites are known to produce glycosidase<br />

enzymes which may affect the barrier function of<br />

mucins secreted by mucosal epithelial cells. 22 An<br />

Italian study of 61 children with symptomatic giardiasis<br />

documented lactose malabsorption on<br />

breath testing, resulting in intolerance to cows’<br />

milk but not to yogurt. 23 However, a breath hydrogen<br />

study in asymptomatic Mexican children with<br />

giardiasis did not find significant carbohydrate<br />

malabsorption. 24 Another study found only<br />

modest effects of Giardia treatment on biopsy<br />

changes, absorptive studies and bile salt conjugation,<br />

suggesting that the parasite’s effect was<br />

synergistic with other organisms in contributing to<br />

tropical enteropathy. 25<br />

A Danish study of 29 children with chronic diarrhea<br />

from giardiasis documented enteropathy in<br />

20, vitamin B12 malabsorption in nine, folate<br />

malabsorption in only five, no evidence of smallbowel<br />

bacterial overgrowth (SBBO) or iron malabsorption<br />

and more severe manifestations in those<br />

who acquired giardiasis overseas. 26 A study of 210<br />

urban Nepali children found no association<br />

between helminth infection and growth or intestinal<br />

permeability, but the eight children with giardiasis<br />

had significantly higher mean permeability<br />

ratios (L : M ratio 0.43) than for either helminth<br />

infected (0.27) or uninfected (0.25). 27 A Canadian<br />

study in a mouse model showed that Giardia<br />

trophozoites increase small-intestinal permeability,<br />

mediated by a myosin light chain kinasedependent<br />

cytoskeletal effect on tight junctions. 28<br />

This disrupted enterocyte barrier function was<br />

T-cell independent and did not affect the stomach


or colon. It is still unclear whether it is these<br />

effects or other mechanisms that cause the T-celldependent<br />

shortening of the brush-border<br />

microvilli with reduced disaccharidases and<br />

impaired absorptive function.<br />

The immune response to Giardia involves both<br />

humoral and cellular mechanisms. 29 Humans<br />

produce IgG, IgM, IgA and IgE responses including<br />

cytotoxic antibodies and an important intestinal<br />

IgA response to acute infection. Nevertheless,<br />

there is still no direct evidence that antibodies<br />

control infection. A Gambian study of six children<br />

with giardiasis, persistent diarrhea and malnutrition<br />

who failed to clear infection after treatment<br />

had raised only Giardia-specific IgM antibodies, so<br />

poor serum and secretory IgA response may be a<br />

marker for ineffective parasite clearance. 30<br />

Children with agammaglobulinemia, but not HIVinfected<br />

children, are susceptible to giardiasis and<br />

it is difficult to eradicate. Breast feeding provides<br />

protection from symptomatic disease, but not from<br />

infection. 31<br />

An association between giardiasis and allergy has<br />

been suggested based on the hypothesis that<br />

increased antigen penetration through damaged<br />

intestinal mucosa enhances sensitization to food<br />

antigens. One study documented allergy symptoms<br />

in 70% of children with giardiasis who had<br />

mean IgE levels of 1194 IU/ml compared to 43%<br />

and 822 IU/ml, respectively, for controls. 32 The<br />

giardiasis group also had higher levels of specific<br />

serum IgE antibodies to food allergens compared<br />

both to controls and to other parasitic infections,<br />

whereas IgE responses to house dust mite were<br />

similar.<br />

Clinical features<br />

The clinical manifestations of Giardia infection<br />

vary from asymptomatic passage of cysts to<br />

chronic diarrhea with malabsorption and weight<br />

loss. The usual clinical syndrome is characterized<br />

by watery diarrhea, foul-smelling stools, bloating<br />

and abdominal cramps. Only about half of patients<br />

develop symptoms following ingestion of cysts,<br />

with 15% passing cysts asymptomatically and the<br />

remainder showing no trace of infection. Although<br />

older children may be asymptomatic, infection<br />

early in life usually causes acute symptoms with<br />

watery diarrhea, anorexia, abdominal distension<br />

Giardiasis 165<br />

and foul stools which persist untreated and may<br />

result in malabsorption. The course of giardiasis is<br />

frequently prolonged and, although many eventually<br />

resolve without treatment, some go on to<br />

syndromes of chronic diarrhea or frequent<br />

relapses. The severity of disease may be determined<br />

by strain-dependent virulence factors in the<br />

parasite, as well as age, nutritional status and<br />

immunocompetence of the host. 33 Young children<br />

are more likely to be severely affected, with failure<br />

to thrive, hypokalemia and malabsorption (fat,<br />

vitamins A and B12, protein and lactose). Children<br />

in the developing world with chronic diarrhea and<br />

malnutrition often have giardiasis, but it is not<br />

always clear how much Giardia is contributing to<br />

the illness, since they are often co-infected with<br />

other enteric pathogens. The results of a small<br />

Venezuelan trial suggested that the therapeutic<br />

control of giardiasis could be important in<br />

programs to combat anemia in children living in<br />

settings with high prevalence. 34<br />

Diagnosis<br />

The diagnosis of giardiasis relies upon stool<br />

microscopy finding trophozoites or cysts. The<br />

sensitivity of a single stool is only 50–70% but<br />

increases to 90% if three stools are examined.<br />

Commercial assays for Giardia antigen are more<br />

sensitive and use polyclonal or monoclonal antibody<br />

directed against cyst or trophozoite antigens,<br />

usually as an enzyme-linked immunosorbent<br />

assay (ELISA) or immunofluorescent assay. These<br />

antigen detection assays are most useful for population<br />

screening such as children at day-care<br />

centers. Duodenal aspirates or biopsies are invasive,<br />

so are not justified exclusively to diagnose<br />

giardiasis, but when done for other reasons are<br />

reliable, particularly in the immunocompromised<br />

subject.<br />

Prevention<br />

Giardia is waterborne and cysts are highly resistant<br />

to chlorine and ozone, so filtration provides<br />

the best protection against transmission through<br />

tap water. 35 Hygiene measures to prevent<br />

fecal–oral transmission need to focus on young<br />

children. The drug treatment of choice for symptomatic<br />

disease (not just cyst excretors) is tinidazole<br />

(see p.183).


166<br />

Intestinal parasites<br />

Ascariasis<br />

Introduction<br />

Ascaris lumbricoides is a large, 15–35cm long<br />

white roundworm that is specific to humans.<br />

Ascaris is one of 63 species of nematode infecting<br />

humans, and the adult roundworm has a biologically<br />

inert surface, so the main antigenic stimuli to<br />

the host are excretory and secretory antigens from<br />

the orifices. A. suum is the related pig species<br />

which may migrate through human tissues in the<br />

larval stage, 36 but cannot complete its life cycle in<br />

man.<br />

Life cycle<br />

The female Ascaris worm produces about 200000<br />

eggs/day, which embryonate in soil in about 3<br />

weeks to become infectious, but are sensitive to<br />

excessive heat, drying and sunlight. Ingested eggs<br />

moult to form larvae (0.2mm long) which penetrate<br />

blood vessels or lymphatics and travel to the<br />

lung via the portal vein or thoracic duct to<br />

pulmonary capillaries and alveoli, where they<br />

moult, grow in size, migrate up the airways and<br />

down the esophagus, and mature and reproduce in<br />

the small intestine. Adult Ascaris are large white<br />

roundworms (females 2–4cm long) which, unlike<br />

hookworm and Trichuris, do not attach to the<br />

mucosa but live free in the small-intestinal lumen.<br />

The duration of the life cycle from ingestion of<br />

Ascaris ova to new egg excretion in feces by the<br />

mature worm takes about 2 months. Adult worms<br />

are expelled from the intestine after about 18<br />

months.<br />

Epidemiology<br />

Ascariasis is one of the most prevalent infections<br />

in the world affecting approximately 1400 million<br />

people (23% of the world population) with 59<br />

million (mostly children) at risk of morbidity,<br />

including 95% of the population of Africa and 45%<br />

of inhabitants of Central and South America. 37<br />

Highest prevalences are found in countries where<br />

sanitation is deficient, and children have higher<br />

mean worm loads than adults. There is some<br />

evidence of a predisposition to high infestation of<br />

Ascaris worms due to a genetic or immunological<br />

predisposition. Morbidity is related to worm load,<br />

which is indirectly assessed by egg concentration<br />

per gram (epg) of feces (e.g. light infection,<br />

< 5000epg, heavy infection, > 50000epg).<br />

Community prevalence levels greater than 70% are<br />

associated with high worm burdens, likelihood of<br />

morbidity and rapid reinfection, so warrant antihelminthic<br />

control programs (see below). 38<br />

Geophagy (eating soil) is a risk factor for ascariasis<br />

in African schoolchildren. 39,40<br />

Immunology of helminths<br />

An important principle of helminth immunology<br />

is that very few responses elicited by infection are<br />

likely to be functionally protective and some have<br />

severe pathological consequences for the host.<br />

Immune responses can also be divided into innate<br />

(immediate response on exposure with little<br />

memory and broad molecular specificity not<br />

involving lymphoid cells) and adaptive (lymphocyte-mediated<br />

and antigen-specific with<br />

memory). 41 Although immune responses in the<br />

intestinal mucosa are similar to systemic<br />

responses, an important difference is the predominance<br />

of IgA antibodies.<br />

Immune responses against helminth invasion<br />

rarely eliminate them directly. Effective immunity<br />

against reinfection is also difficult to demonstrate<br />

for helminth infections, since worms persist in the<br />

face of specific immune responses, although agerelated<br />

declines in infection rates do occur in<br />

endemic areas. However, detailed epidemiological<br />

and animal model studies have demonstrated that<br />

acquired protective immunity is important for<br />

control of hookworm, Trichuris and schistosomiasis.<br />

41 Ascaris produces the lipid-binding protein<br />

ABA-1 which can stimulate IgE antibodies, high<br />

levels of which correlate with protection against<br />

Ascaris. 42<br />

The traditional view of the host immune response<br />

was that with time it reduced their fecundity and<br />

survival via mechanisms such as induction of<br />

intestinal mast cells and eosinophils. More recent<br />

evidence suggests that other indirect host<br />

responses may result in parasite starvation or<br />

depletion of energy sources for defending itself<br />

against the host’s immune responses, leading to<br />

reduced fecundity or survival. 43 However, there is<br />

also a cost to the host from mounting an immune


esponse to the parasite, particularly on nutritional<br />

status in children. For example, zinc deficiency<br />

may prolong nematode survival in the host by its<br />

known effect on mucosal immune function. 44 In<br />

addition, many helminths release immunomodulatory<br />

molecules which inactivate components of<br />

the host’s immune response, but this may also<br />

predispose the host to other infections such as<br />

tuberculosis (TB) or AIDS. There is also evidence<br />

that ascariasis impairs immune responses to<br />

vaccines in that albendazole treatment increased<br />

the seroconversion rate and antibody titers after<br />

cholera immunization, mediated by suppression of<br />

interleukin (IL)-2 responses. 45,46 Moreover, treatment<br />

of helminths in HIV-infected individuals was<br />

associated with significant reductions in viral<br />

load. 47<br />

Immune-mediated inflammation occurs in most<br />

parasitic infections, particularly involving IgE<br />

antibodies, eosinophils, cytokines and T cells, but<br />

there are important differences between helminths<br />

and protozoa. First, although protozoa may be<br />

phagocytosed by macrophages, cell-mediated<br />

immunity against helminths involves attachment<br />

to its surface with antibody or complementmediated<br />

mechanisms and release of mediators<br />

which damage its surface. Second, whereas protozoa<br />

tend to elicit T-helper lymphocytes with activation<br />

of macrophages, cytokines (IL-12) and<br />

specific IgG antibodies (Th1 responses), helminths<br />

elicit Th2 responses with IL-4 stimulating IgE antibodies<br />

and inflammatory cells (e.g. eosinophils<br />

and mast cells). Thus, compared to controls,<br />

cytokine responses in Ascaris-infected subjects<br />

involved mononuclear cell IL-4 and IL-5 stimulation<br />

with no difference in IL-10 and interferon-γ<br />

responses, consistent with a highly polarized Th2<br />

cytokine response. 48<br />

Helminth infections are characterized by<br />

increased mast cells and eosinophils. The key<br />

function of IL-5, which is produced by activated<br />

CD4 T cells, is control of eosinophils, although<br />

early differentiation of eosinophils is controlled by<br />

granulocyte-macrophage–colony-stimulating<br />

factor (GM-CSF) and IL-3. It is now recognized that<br />

eosinophils, together with antibody and complement,<br />

have a role in the killing of infective larval<br />

stages of most helminths, but not of adult worms.<br />

Recent studies in mice suggest that the effect of IL-<br />

5 and eosinophils differs for each parasite species,<br />

Ascariasis 167<br />

but they do have a role in host protection which is<br />

not yet clearly defined. 49,50<br />

IgE antibodies are an important component of<br />

host-protective immune responses against<br />

helminthic parasites, including both parasitespecific<br />

IgE and non-specific polyclonal IgE mediated<br />

by IL-4. Excessive polyclonal stimulation of<br />

IgE synthesis by helminths, which is prevented by<br />

anthelminthic treatment, suppresses allergic reactivity<br />

by saturating mast cell receptors and inhibiting<br />

specific IgE antibodies (e.g. to inhaled antigens<br />

such as house dust mite) and skin test reactivity. 51<br />

Compared to urban children, rural Kenyan children’s<br />

total IgE and Ascaris-specific IgE antibodies<br />

were much higher, and evidence of atopy (skin<br />

testing, bronchial hyper-reactivity) were much<br />

lower. 52 A Venezuelan study examined two groups<br />

of children with comparable living conditions and<br />

Ascaris prevalence but with very different prevalences<br />

of allergic disease. 53 Those in the atopic<br />

group had an intrinsic propensity for specific over<br />

polyclonal IgE responses to the parasite and had<br />

significantly lower intensities of infection than the<br />

non-atopic group, suggesting that the atopic state<br />

conferred a selective evolutionary advantage.<br />

Immune responses are useful as a means of diagnosing<br />

infection by ELISA tests. The major drawback<br />

of antibody-based tests is that a positive test<br />

does not prove current infection in an endemic<br />

area, although use of particular classes of antibody<br />

(e.g. IgG4, IgM) may improve the specificity.<br />

Newer diagnostic tests by antigen-capture ELISA<br />

for coproantigens in feces are becoming available,<br />

which are highly sensitive and specific.<br />

Clinical features<br />

Ascaris infection is not associated with mucosal<br />

damage, increased intestinal permeability or<br />

lactose malabsorption, 54,55 since 85% of infected<br />

individuals have light infections which remain<br />

asymptomatic. Heavy infection (ingestion of<br />

> 2000 eggs) may induce a pneumonitis from<br />

migrating pulmonary larvae, with cough, wheeze,<br />

eosinophilia and transient patchy infiltrates which<br />

may be difficult to differentiate from pneumonia,<br />

asthma or bronchitis. This syndrome of tropical<br />

pulmonary eosinophilia (Loeffler’s) is rarely<br />

recognized clinically in children with Ascaris or


168<br />

Intestinal parasites<br />

hookworm, but is more often symptomatic with<br />

filariasis or toxocara infections. 56<br />

The most common clinical feature of ascariasis is<br />

intestinal obstruction from a bolus of worms,<br />

which occurs in 0.2% of infections in children,<br />

accounting for 72% of all complications of<br />

Ascaris. 57 The mean age of cases is under 5 years<br />

and the case fatality rate about 5.7%. Surgical<br />

management can invariably be avoided with experience<br />

with this syndrome, and daily nasogastric<br />

administration of anthelminthics with supportive<br />

therapy until the bolus is passed. Worms are often<br />

vomited or passed in stool on presentation of<br />

febrile children with severe malaria or bacterial<br />

infections, which invariably prompts comments<br />

from health workers about them ‘abandoning a<br />

sinking ship’. Fever, acute phase reactants and<br />

certain irritants (e.g. carbon tetrachloride, a former<br />

hookworm therapy) are associated with worm<br />

expulsion or aberrant migration. Less frequent<br />

complications of migrating Ascaris worms are<br />

biliary colic, pancreatitis, appendiceal abscess and<br />

appearance through a surgical abdominal wound.<br />

Diagnosis<br />

The diagnosis of ascariasis is based upon identification<br />

of the characteristic eggs on microscopy of<br />

stool or identification of the adult worm passed<br />

spontaneously or after treatment. Eggs are plentiful<br />

in feces since each female produces a mean of<br />

200 000 daily, although the correlation between<br />

eggs per gram of feces and intensity of infection is<br />

imperfect.<br />

Prevention<br />

If human feces are used as fertilizer for growing<br />

vegetables, they need to be stored at 30°C for 40<br />

days to ensure destruction of ova. A number of<br />

reports have examined environmental risk factors<br />

for ascariasis. A Brazilian study, for example, gave<br />

the following relative risks with 95% CI for ascariasis<br />

intensity: over crowding 2.2 (1.0–4.5), poor<br />

household water availability 2.4 (1.1–5.0), poor<br />

hygiene 2.4 (1.0–5.6), less than 4 years’ schooling<br />

5.9 (2.6–14.3) and no recent anthelminthic treatment<br />

2.0 (1.0–4.0). 58 A Kenyan study found that<br />

household overcrowding and the absence of<br />

latrines increased the risk of hookworm infection,<br />

whereas a lack of soap and other household children<br />

under 5 years increased the risk of Ascaris<br />

infection. 59 Since the lack of latrines and soap<br />

usage were identified as risk factors for infection,<br />

helminth control interventions should focus on<br />

these. Mass chemotherapy campaigns for<br />

geohelminths is discussed below.<br />

Hookworm<br />

Introduction<br />

The two major species of hookworms are<br />

Ancylostoma duodenale and Necator americanus<br />

(Figure 11.2). They will be considered together<br />

here, because they have similar life cycles and<br />

disease. However, Ancylostoma tends to be more<br />

virulent than Necator, and adult females are larger<br />

(10–13 vs. 9–11 mm), survive for a shorter time in<br />

the host (1 vs. 4 years), produce more eggs<br />

(10 000–30 000 vs. 5–10 000/day), cause greater<br />

blood loss (0.2 vs. 0.02ml/day) and their larvae can<br />

rest dormant and infect via oral ingestion (unlike<br />

Necator).<br />

Life cycle<br />

The gravid female hookworm produces about<br />

5000–30 000 eggs/day (60µm long) in feces, which<br />

require a moist shady environment to hatch into<br />

Figure 11.2 Necator americanus hookworm. Courtesy of<br />

UK Tropical International Health.


habditiform larvae (300µm long) which grow to<br />

become infective larvae (600µm) and enter the<br />

host’s venules or lymphatics, usually when walked<br />

upon with bare feet. The larvae then migrate into<br />

the lungs and ascend up the respiratory tract and<br />

descend to the small intestine, where they attach<br />

and mature in the jejunum. The time from infection<br />

to egg production is almost 2 months.<br />

Epidemiology<br />

Hookworm is probably the second most prevalent<br />

intestinal parasite after ascariasis, with 1200<br />

million people infected worldwide (two-thirds by<br />

Necator), including 90–130 million with morbidity.<br />

Necator predominates in Central and South<br />

America (New World), and Ancylostoma in India,<br />

China, North Africa and tropical Australia, but<br />

mixed infections occur in many regions of Asia,<br />

Africa, Central America and the South Pacific.<br />

Ancylostoma extends into more temperate regions<br />

than Necator (e.g. North Africa, China and Europe)<br />

because its ova are more resistant to cold. In an<br />

urban Nigerian study of 862 schoolchildren with<br />

hookworm, 72.0% had Necator alone, 4.5%<br />

Ancylostoma alone and 23.4% both species of<br />

hookworm, but with Necator in higher numbers. 60<br />

Unlike Ascaris and Trichuris, hookworm transmission<br />

is closely associated with rural farming rather<br />

than urban slums. There are several other species<br />

of dog and cat helminths (e.g. Toxocara species)<br />

which can cause eosinophilic enteritis, cutaneous<br />

larva migrans or viscera larva migrans in humans.<br />

For example, studies in tropical Australia have<br />

described eosinophilic enteritis with abdominal<br />

pain caused by Ancylostoma caninum, a dog hookworm.<br />

61<br />

Pathophysiology<br />

Hookworms attach to the mucosa with teeth<br />

(Ancylostoma) or cutting plates (Necator) and<br />

release anticoagulant polypeptides and neutrophil<br />

inhibitory factor which down-regulate the inflammatory<br />

response of the host. They also release<br />

hydrolytic enzymes which damage mucosal capillaries<br />

and exacerbate the blood loss. Hookworm<br />

larvae release Ancylostoma-secreted proteins<br />

(ASPs) during the early stages of infection, which<br />

are highly antigenic. The indirect evidence of<br />

Hookworm 169<br />

reduced hookworm burdens in adults suggests an<br />

effect of host immunity, but humoral antibodies<br />

and cell-mediated immune responses correlate<br />

poorly with resistance to infection. The possibility<br />

of a hookworm vaccine to ASPs is under investigation.<br />

62<br />

Clinical features<br />

Hookworm larvae entering skin can result in a<br />

papulovesicular rash at the site of entry (ground<br />

itch) or cutaneous larvae migrans for animal hookworms.<br />

Although eosinophilia accompanies the<br />

larval migration phase, pneumonitis is mild and<br />

rarely recognized in children. The main morbidity<br />

from hookworm is iron deficiency anemia, particularly<br />

with heavy infections. It was not uncommon<br />

in Caribbean and Pacific islands to have<br />

school children presenting in congestive heart<br />

failure with loud hemic murmurs and hemoglobin<br />

levels of


170<br />

Prevention<br />

Intestinal parasites<br />

Measures to prevent hookworm include ceasing<br />

the use of human feces as fertilizer, use of toilets,<br />

wearing shoes and generally improving living<br />

standards. Mass treatment programs with albendazole<br />

have a transient effect, but need to be<br />

combined with improved sanitation and health<br />

education to prevent high reinfection rates. 64 Iron<br />

supplementation has no effect on either reinfection<br />

rates or helminth intensities in children. 65 In<br />

high prevalence areas of hookworm infection and<br />

schistosomiasis, regular mass deworming<br />

campaigns with albendazole and praziquantel are<br />

effective in reducing anemia rates. 66,67<br />

Trichuriasis<br />

Introduction<br />

Trichuris trichiuria, meaning ‘hairy tail’, is actually<br />

a misnomer, since it is the proximal end that is<br />

hairlike. The popular name is whipworm, with the<br />

whip as the long thin pharynx (stichostome) and<br />

the whip handle as the posterior end with reproductive<br />

organs and intestine (Figure 11.3).<br />

Life cycle<br />

A mature female worm produces up to 20 000<br />

eggs/day, which are 50 µm long and not infectious<br />

until the larval stage develops in the soil over 2–4<br />

weeks. Warm damp soil is ideal for embryonation,<br />

Figure 11.3 Trichuris trichiura worms attached to the<br />

colon. Courtesy of UK Tropical International Health.<br />

and environmental exposure is associated with<br />

poor hygiene practices. Once ingested, larvae<br />

penetrate the epithelium of the mucosal crypt in<br />

the cecum, where they moult and the hair-like<br />

stichosome remains attached while the broader<br />

distal end extends into the lumen. The adult worm<br />

is 4 cm long and survives for 1–2 years in the host.<br />

Epidemiology<br />

Trichuriasis is a very common infestation with an<br />

estimated 1049 million cases worldwide, including<br />

114 million preschool and 233 million schoolage<br />

children. 68 Most of these infections are asymptomatic,<br />

but children suffer greater morbidity than<br />

adults. There appears to be a genetic or immunological<br />

predisposition to heavy infection, which<br />

may be HLA group-mediated. 69 There also appears<br />

to be some epidemiological ‘synergism’ between<br />

ascariasis and trichuriasis in that high-intensity<br />

infections occur together more often than by<br />

chance. 70<br />

Pathophysiology<br />

With severe infection, the cecal mucosa becomes<br />

inflamed and edematous, but there are only minor<br />

changes in histology (increased IgM lamina<br />

propria plasma cells and calprotectin-secreting<br />

cells, crypt epithelial cell proliferation, distended<br />

goblet cells). 71 The cytokine response in colonic<br />

mucosa involves both interferon-γ (Th1) and IL-4<br />

(Th2), but results in neither worm expulsion nor<br />

obvious protection from reinfection. There may be<br />

a protein-losing enteropathy proportional to the<br />

worm burden. 72 There is an acute-phase response<br />

with intense trichuriasis, including increases in<br />

plasma tumor necrosis factor (TNF)-α, C-reactive<br />

protein (CRP), globulins, viscosity and fibronectin<br />

with decreased insulin-like growth factor (IGF)-<br />

I 73–75 . There is also development of an IgEmediated<br />

immediate hypersensitivity reaction in<br />

the colon, 76 usually, but not always, with systemic<br />

eosinophilia.<br />

Clinical features<br />

Most infections in children are light (< 20 adult<br />

worms) and asymptomatic, with symptoms developing<br />

in < 10% of infected children. Occult blood


in feces is uncommon even with heavy infestations,<br />

although even light infections incite a local<br />

inflammatory response involving eosinophils and<br />

neutrophils in the colon. 77,78 With heavy infestations,<br />

frequent watery or mucous stools occur,<br />

sometimes with frank blood. Rectal prolapse can<br />

occur with heavy infestations. 79,80 A very small<br />

minority of heavily infected children (>200<br />

worms in the rectum, colon and often terminal<br />

ileum) develop the Trichuris dysentery<br />

syndrome, 81,82 characterized by chronic dysentery,<br />

stunting, anemia and finger clubbing.<br />

Diagnosis<br />

The diagnosis is based on finding eggs on stool<br />

microscopy, with more than 10 000 epg generally<br />

associated with heavy infection, although there is<br />

considerable individual variation in eggs to worm<br />

load. Trichuris dysentery in children must be<br />

differentiated from amebic and bacilliary causes.<br />

Most cases of rectal prolapse in malnourished children<br />

are not due to trichuriasis.<br />

Prevention<br />

The use of proper latrines, good hygiene with<br />

handwashing and washing of vegetables will interrupt<br />

the life cycle. Overcrowded urban slums with<br />

limited water supply and heavily fecally contaminated<br />

soil for growing vegetables place children at<br />

particular risk. Mass chemotherapy is highly effective,<br />

but reinfection occurs rapidly in this setting.<br />

Cryptosporidiosis<br />

Introduction<br />

The protozoa Cryptosporidium parvum, Isospora<br />

belli, Cyclospora cayetanensis and Sarcocystis<br />

hominis all belong to the group of intestinal<br />

coccidial infections, which cause diarrhea. They<br />

have come into prominence in recent years due to<br />

causing severe and protracted diarrhea in AIDS or<br />

cell-mediated immune deficiency and infecting<br />

piped water supplies due to the chlorine resistance<br />

of oocysts. The opportunistic protozoal infections<br />

which affect HIV-infected patients include<br />

Cryptosporidium, Microsporidia, Cyclospora and<br />

Cryptosporidiosis 171<br />

Isospora belli. Cryptosporidium also causes persistent<br />

diarrhea and proximal small-intestinal<br />

enteropathy in children with normal immune<br />

function. 10,83<br />

Life cycle<br />

Cryptosporidiosis is transmitted by a 3–6µm acidfast<br />

thick-walled oocyst shed in stool which<br />

encysts in the lumen of the small bowel after<br />

ingestion and produces four sporozoites. These<br />

penetrate the microvillus border and develop into<br />

trophozoites, which cause disease and can reproduce<br />

asexually, releasing eight merozoites, which<br />

invade nearby cells and redevelop into trophozoites<br />

to continue the infection. The life cycle is<br />

completed by asexual reproduction of trophozoites<br />

to schizonts, which develop progressively into<br />

male microgametes or female macrogametes,<br />

combining to form zygotes and then oocysts,<br />

which pass into the stool. After ingestion of<br />

oocysts, encystation takes place in the upper small<br />

intestine and sporozoites invade the absorptive<br />

epithelial cells causing inflammation, partial<br />

villus atrophy, malabsorption and diarrhea. About<br />

20% of oocysts are thin-walled and can autoinfect<br />

the host, which explains how a small number of<br />

ingested oocysts can cause severe disease, especially<br />

in immunocompromised patients. Fecal<br />

oocysts can survive for at least 2 days, and the<br />

incubation period from cyst ingestion to diarrhea<br />

in humans is 2–14 days. The major difference from<br />

Plasmodium species (malaria) is that Cryptosporidium<br />

completes its life cycle in a single host<br />

without the mosquito vector.<br />

Epidemiology<br />

Cryptosporidium causes diarrheal disease in children<br />

of developing countries, in travelers and in<br />

immunocompromised patients; there have been<br />

waterborne outbreaks in developed countries.<br />

Transmission is from person to person and from<br />

animals to people by ingestion of fecally contaminated<br />

food or water. C. parvum infects numerous<br />

mammals, including man and cattle, and although<br />

there is a human-specific form, animal forms may<br />

also cause disease in humans. Oocysts are highly<br />

infectious, with ingestion of a median of 132 cysts<br />

found to be infectious in adult volunteers. Infected


172<br />

Intestinal parasites<br />

AIDS patients excrete millions of oocysts daily.<br />

Person-to-person transmission is common, occurring<br />

in 19% of infected children and is especially<br />

common in day-care center settings. Oocysts are<br />

relatively resistant to conventional chlorination<br />

and filtering, but 1-µm filters, heating (72°C for a<br />

minute) and freezing are effective.<br />

Cryptosporium has been reported to occur in 6.1%<br />

of diarrheal cases vs. 1.5% of controls in developing<br />

world subjects compared to 2.2% vs. 0.2%,<br />

respectively, in the developed world. For AIDS<br />

patients, these rates increase to 24% vs. 5%,<br />

respectively, in the developing world and 14% vs.<br />

0% in the developed world. A Peruvian periurban<br />

study found a peak cryptosporidiosis incidence of<br />

0.42 episodes/year in 1-year-olds (mean childhood<br />

incidence of 0.20), with the rainy season and lack<br />

of toilets as risk factors. 84 In Zambian periurban<br />

children, cryptosporidial infection appeared to be<br />

predominantly waterborne with a prevalence<br />

among diarrheal cases of 18%, and no association<br />

with home animals, nutritional status or parental<br />

education. 85<br />

In community studies of children in Guinea-<br />

Bissau, Cryptosporidium was found in 5.8–7.4% of<br />

diarrheal episodes and 2.0% without diarrhea,<br />

with peak prevalences in the early rainy season<br />

(17.6% in May), in young children (e.g. 12.6% in<br />

infants < 6 months) and with persistent diarrhea<br />

(15%). 86–88 Additional risk factors were pigs and<br />

dogs in the household, prolonged storage of<br />

cooked food and male sex, but not impaired immunity.<br />

89 According to the authors, the epidemiology<br />

of infection was consistent with a small infective<br />

dose, airborne transmission and slow development<br />

of protective immunity. In Kuwait, C. parvum<br />

oocysts were found in 10% (51 cases) of childhood<br />

diarrhea, but more older children were affected<br />

(73% > 2 years). 90 A Gambian study of 1200 children<br />

documented Cryptosporidium in 8.8% of diarrheal<br />

cases vs. 2.8% of controls (p < 0.001), with<br />

peak incidence during the rainy season and among<br />

infants (6–11 months). 91 A Mexican study of 403<br />

children with diarrhea had a 6.4% prevalence of<br />

Cryptosporidium, which was associated with<br />

young age, malnutrition and lack of breast<br />

feeding. 92 A Brazilian case–control study showed<br />

that children who acquired symptomatic cryptosporidiosis<br />

before 1 year of age had a higher<br />

burden of subsequent diarrheal disease. 93<br />

A major waterborne cryptosporidiosis outbreak in<br />

Milwaukee, USA in 1993 increased IgG antibody<br />

responses from 15% to 82% over a 5-week period<br />

in the area served by the water treatment plant,<br />

demonstrating that C. parvum infection had been<br />

much more widespread than was previously<br />

appreciated from stool testing. 94 During this<br />

outbreak, Cryptosporidium, as the sole pathogen,<br />

was identified in stools from only 23% of affected<br />

children suggesting that stool tests for<br />

Cryptosporidium were insensitive early in the<br />

course of illness. 95 Although secondary transmission<br />

undoubtedly took place in child-care facilities,<br />

the presence of children with asymptomatic<br />

Cryptosporidium infections did not result in an<br />

increased risk of diarrhea. 96<br />

Pathophysiology<br />

It is paradoxical that Cryptosporidium can produce<br />

such profound inflammatory mucosal damage<br />

without invasion. It has no known cytotoxins or<br />

enterotoxins, but some of the damage may be<br />

cytokine-mediated (TNF-α, IL-8), contributing to<br />

the malabsorption. C. parvum sporozoites produce<br />

a mucin-like surface glycoprotein which may help<br />

in parasite attachment to enterocytes. 22 Animal<br />

studies have revealed that cryptosporidiosis<br />

impairs glucose-stimulated sodium absorption at<br />

the villus without affecting cyclic AMP-mediated<br />

chloride secretion by the crypt epithelium. 97,98<br />

The inflammatory response in the lamina propria<br />

does not appear to enhance active transport<br />

processes such as chloride secretion, but diarrhea<br />

is related to loss of villus epithelium and glucosefacilitated<br />

sodium transport.<br />

Severe disease occurs in agammaglobulinemia, so<br />

humoral immunity provides important protection,<br />

but cell-mediated immunity is also important in<br />

clearing infection. An antibody response to<br />

Cryptosporidium develops in the ileal mucosa<br />

which is responsible for termination of the infection.<br />

99 Interferon-γ, produced by the Th1 subset of<br />

CD4 intraepithelial lymphocytes, is also important<br />

in combating cryptosporidiosis, as are TNF-α and<br />

IL-1β but without any synergistic effect. 100,101 The<br />

mechanisms of action of interferon-γ in vitro are<br />

prevention of penetration of host enterocytes by<br />

the parasite and retardation of development of<br />

intracellular parasites, which were independent of


nitric oxide-mediated killing of parasites, although<br />

interferon-γ may also up-regulate nitric oxide<br />

synthesis in enterocytes. 100<br />

Parasite expulsion and immune-mediated enteropathic<br />

changes appear to be mediated by T-cell<br />

function, dependent upon TNF, inducible nitric<br />

oxide synthase (iNOS) and IL-4 cytokines (Th2<br />

responses). 102,103 Thus, Th2 cytokines (e.g. IL-4)<br />

are involved in protection but may also produce a<br />

pathological response in the intestinal mucosa,<br />

which had been attributed to Th1 cytokines, and<br />

does not occur in TNF-α- and interferon-γ-deficient<br />

subjects. Both severe intestinal inflammation<br />

with enteropathy and protective immune<br />

responses to intestinal parasites are dependent<br />

upon IL-4, but immune protection can be acquired<br />

without enteropathy. 102 A neonatal mouse model<br />

of cryptosporidiosis documented increased<br />

inducible nitric oxide (NO) production with infection,<br />

which was worsened by NO inhibition and<br />

antioxidant administration, suggesting a protective<br />

role for NO. 104,105 The severity of nutrient malabsorption,<br />

villus atrophy and abnormal intestinal<br />

permeability with Cryptosporidium infection in<br />

AIDS is proportional to the number of infecting<br />

organisms. 106<br />

Clinical features<br />

Cryptosporidial infection causes watery diarrhea<br />

with low-grade fever, vomiting and often cramps,<br />

severe dehydration and hypokalemia. Among<br />

Aboriginal children in Darwin, it was found in the<br />

stool of 7.4% of admissions with diarrheal disease<br />

with a mean age of 12.2 (9.6–15.5) months and<br />

mean admission serum potassium level of 2.7<br />

(2.4–3.0) mmol/l. It was associated with the most<br />

severe and prolonged mucosal damage and inflammation<br />

on permeability and NO testing, but<br />

showed less lactose intolerance than rotavirus. 10 A<br />

retrospective hospital study of 109 cases in<br />

London, 92% had watery and offensive stools with<br />

blood and mucus only occasionally, 50% had<br />

persistent diarrhea (> 14 days, and 33% > 21<br />

days), 51% had vomiting, 21% had significant<br />

abdominal pain and 23% were underweight. 83<br />

Biopsies in nine cases with prolonged diarrhea and<br />

failure to thrive showed a mild to moderate<br />

enteropathy with Cryptosporidium adhering to the<br />

villus epithelium, reduced villus height, reduced<br />

disaccharidases and increased intraepithelial<br />

Cryptosporidiosis 173<br />

lymphocytes. Key risk factors for cryptosporidiosis<br />

in this European setting were travel to a developing<br />

country and itinerant parents living in caravan<br />

sites. There have been two community growth<br />

studies in Peru and Guinea-Bissau, both suggesting<br />

an adverse long-term effect of cryptosporidiosis on<br />

growth. 107,108<br />

Cryptosporidium causes more prolonged and<br />

severe diarrhea, which may be fulminant, in<br />

immunocompromised patients. In an Italian study<br />

of HIV-infected children with cryptosporidiosis,<br />

the median duration of diarrhea was 32 days<br />

compared to only 4 days for other causes. 109 It was<br />

also associated with loss of 5–30% of body weight.<br />

A Peruvian hospital case–control study found an<br />

association between C. parvum infection and<br />

malnutrition, and high nosocomial spread in the<br />

hospital context. 110 In a follow-up study, they<br />

documented that a first symptomatic infection<br />

resulted in a mean (with 95% CI) of 342 g<br />

(167–517) weight deficit whereas asymptomatic<br />

infection (which was twice as common) led to a<br />

162g (27–297) deficit during the first month of<br />

infected cases compared to uninfected nondiarrheal<br />

cases. 111<br />

Diagnosis<br />

Cryptosporidiosis is diagnosed by finding oocysts<br />

in stool using an acid-fast stain. This is sensitive<br />

with more than 10 000 cysts/g in diarrheal cases,<br />

but 50 times less sensitive without diarrhea.<br />

Immunofluorescent and ELISA techniques are<br />

more sensitive, and PCR may be even more sensitive<br />

for detecting low numbers of oocysts in stool<br />

specimens. 92 Serological testing for cryptosporidial<br />

antibodies is useful for epidemiological<br />

surveys, but not for clinical diagnosis in endemic<br />

areas. A Peruvian study has questioned whether<br />

cryptosporidiosis is caused by a single species by<br />

identifying both human (in 81% of children) and<br />

zoonotic genotypes of Cryptosporidium (bovine,<br />

dog, C. meleagridis and felis) in HIV-negative children,<br />

with only duration of oocyst shedding longer<br />

in the human genotype. 112<br />

Prevention<br />

The high infectivity and ubiquitous oocysts in the<br />

environment make prevention by water, hygiene


174<br />

Intestinal parasites<br />

and sanitation programs very difficult, indeed<br />

impossible, in the developing world, where up to<br />

95% of children in some areas have positive serology<br />

by the age of 2 years. Precautions for travelers<br />

include washing hands, boiling water, avoiding<br />

animals, proper cooking of food, peeling fruit and<br />

avoiding uncooked food in contact with unboiled<br />

water (e.g. salads).<br />

Strongyloidiasis<br />

Introduction<br />

Although not a major cause of morbidity worldwide,<br />

the nematode Strongyloides stercoralis is<br />

unique in its ability to persist indefinitely within<br />

the host through autoinfection, and cause disseminated<br />

disease with the prolonged use of corticosteroids<br />

or other causes of immunosuppression.<br />

Life cycle<br />

Adult females are about 2.5mm in length and are<br />

attached to the lamina propria of the duodenum or<br />

proximal jejunum (Figure 11.4). Their eggs, which<br />

hatch into rhabditiform larvae (250µm long) and<br />

pass via feces into the external environment,<br />

where they moult into the infective filariform<br />

larvae (550µm long) which infect by penetrating<br />

the skin, like hookworms (see above) or become<br />

tissue-penetrating infective larvae by penetrating<br />

the colonic wall or perianal skin, becoming reestablished<br />

as mature female worms in the small<br />

Figure 11.4 Strongyloides stercoralis lava. Courtesy of<br />

UK Tropical International Health.<br />

intestine (autoinfection). Larvae are sensitive to<br />

drying, excessive heat (> 40°C) and cold (< 8°C),<br />

but may survive for about 2 weeks in soil.<br />

Rhabditiform larvae in feces may also moult in<br />

moist warm soil to form free-living adult males<br />

and females, who mate. The females produce eggs<br />

which develop into infective larvae to continue<br />

their life cycle in man. The time from skin penetration<br />

to mature worms producing eggs is approximately<br />

28 days.<br />

Epidemiology<br />

S. stercoralis is present in virtually all tropical and<br />

subtropical regions, but estimates of worldwide<br />

prevalence vary widely (3–100 million) with the<br />

best estimate probably around 30 million people in<br />

70 countries. 113 It is known to be prevalent among<br />

children in Cambodia, Laos, India (Bihar), Guinea-<br />

Bissau, Kenya, Sierra Leone, Brazil and the<br />

Caribbean. A large study in a Peruvian Amazon<br />

community found a 19.5% prevalence of stool<br />

larvae, including 25% in preschool children who<br />

also had high rates of other parasites. 114<br />

Strongyloidiasis accounted for 7.8% of 269 acute<br />

diarrheal admissions in Australian Aboriginal children<br />

in Darwin, with a mean age of 23.3<br />

(18.1–30.1) months, which was significantly older<br />

than the mean age of 12.7 (11.8–13.7) months for<br />

other diarrheal admissions. 10<br />

Prevalence rates vary with climate, geographical<br />

region, environmental conditions, soil characteristics<br />

and socioeconomic status, but also with modifiable<br />

risk factors such as quality of housing,<br />

hygiene standards and high population density. In<br />

endemic areas, most affected individuals have a<br />

low intensity of infection, but a few are heavily<br />

infected. Household aggregation of infection has<br />

been documented in Bangladesh, with risk factors<br />

being older age (7–10 years), lack of a home latrine<br />

and low socioeconomic status. 115,116 S. fuelleborni<br />

is a more virulent infection affecting young children<br />

in Papua New Guinea, which may be transmitted<br />

via breast milk. 117<br />

Pathophysiology<br />

Malabsorption was recognized as a feature of<br />

strongyloidiasis in a 1965 Jamaican study using<br />

iron, folate and xylose absorption tests along with


jejunal biopsies. 118 SBBO and abnormal intestinal<br />

permeability to 51 Cr-ethylenediaminetetra-acetic<br />

acid (EDTA) has also been documented in<br />

Brazilian adults with strongyloidiasis and symptoms<br />

of abdominal pain, diarrhea and weight<br />

loss. 119,120 The suggested mechanisms for abnormal<br />

permeability were enhanced mucus secretion,<br />

increased enterocyte turnover with impaired paracellular<br />

barrier function, and protein-losing<br />

enteropathy. The latter was documented by means<br />

of increased fecal α1-antitrypsin excretion in 17%<br />

(5/29) of Gambian children with persistent diarrhea<br />

and malnutrition, including two of six with<br />

Strongyloides and two of 17 Giardia cases. 121<br />

Small-bowel biopsy studies have reported normal<br />

histology in some mild infections, but most show<br />

thickening of the bowel wall with edema or fibrosis<br />

in what has been described as catarrhal, edematous<br />

or ulcerative enteritis. 122 In severe cases of<br />

autoinfection, larvae hatching from deposited eggs<br />

burrow through the lumen causing superficial<br />

damage to the mucosa with excess mucus production,<br />

larvae in the lymphatics lead to a granulomatous<br />

lymphangitis, and parasites in the submucosa<br />

cause edematous and atrophic changes with fibrosis.<br />

As with other intestinal nematodes,<br />

Strongyloides infection elicits Th2-dependent antibodies.<br />

Immunoglobulin responses to S. stercoralis<br />

indicate that IgA affects larval output, IgE regulates<br />

autoinfection and IgG4 blocks IgE<br />

responses. 123 These immune responses appear to<br />

offer little protective immunity, and the relative<br />

contributions of genetic versus environmental<br />

factors to heavy infections is still unclear. Preexisting<br />

malnutrition (wasting) is a risk factor for<br />

hospitalization with the acute diarrheal syndrome<br />

compared to other causes of diarrhea, increasing<br />

the risk 6.5-fold even after adjusting for confounding<br />

factors. 10<br />

Clinical features<br />

As with hookworm, larval migration may affect the<br />

lungs (eosinophilic pneumonitis) or skin (‘ground<br />

itch’ on the foot or ‘larva currens’ on the buttocks)<br />

but these are not usually recognized in children.<br />

Larvae from species infecting other mammals may<br />

penetrate the skin of humans, causing only local<br />

irritation, but cannot complete their life cycle. The<br />

most common manifestation of S. stercoralis infec-<br />

Strongyloidiasis 175<br />

tion in children is an acute diarrheal illness with<br />

foul stools with a typical musty odor which can be<br />

recognized by experienced staff in high-prevalence<br />

areas. Severe dehydration is uncommon, but<br />

hypokalemia and malabsorption commonly occur.<br />

Eosinophilia (5–15% of total white blood cell<br />

(WBC) count) is a common but not universal<br />

finding. A syndrome of partial intestinal obstruction<br />

with strongyloidiasis has been described in<br />

Aboriginal children in the Northern Territory of<br />

Australia. 124,125<br />

S. fuelleborni in infants is associated with abdominal<br />

swelling, ascites, pleural effusions and a high<br />

mortality. 126 In contrast to hookworm, with which<br />

it is commonly associated in infants in Papua New<br />

Guinea, the intensity of infection peaks by about<br />

12 months of age. 117,127 A similar S. fuelleborni<br />

species affects monkeys and humans in central<br />

Africa, causing less virulent disease with fever,<br />

lymphadenitis, abdominal pain and eosinophilia.<br />

41<br />

Disseminated strongyloidiasis (hyperinfection)<br />

occurs with impaired cell-mediated immunity, as<br />

in children treated with prolonged courses of<br />

steroids (although not from short courses of<br />

steroids for asthma) or malignancy (e.g.<br />

lymphoma, leukemia) or on immunosuppressive<br />

drugs (except cyclosporin, which is antiparasitic).<br />

Eradication of Strongyloides is essential before<br />

immunosuppressive therapy is commenced.<br />

Disseminated infection is always a serious complication<br />

with high mortality, usually affecting the<br />

bowel, lungs and central nervous system (CNS)<br />

and often accompanied by sepsis. Although<br />

strongyloidiasis is not a common opportunistic<br />

infection in AIDS, there is an increased frequency<br />

of larvae with HTLV-I infection, possibly due to<br />

suppression of the host IgE response. 128<br />

Diagnosis<br />

The diagnosis is established by identification of<br />

larvae on stool microscopy, which is very reliable<br />

with acute diarrhea, but less reliable with chronic<br />

or asymptomatic infection, because rhabditiform<br />

larva excretion is irregular and the parasite load is<br />

often low. In this situation, a single stool examination<br />

may detect larvae in only 30% of cases of<br />

latent infections, although this can be increased to


176<br />

Intestinal parasites<br />

50% with three stool specimens or various concentration<br />

techniques. There are a number of culture<br />

methods which are particularly useful if no larvae<br />

are found on stool microscopy in the face of clinical<br />

suspicion. The agar plate technique has a 96%<br />

sensitivity, which identifies larval tracks<br />

(Strongyloides larvae lash like a whip, whereas<br />

hookworm larvae move like a snake). 41,129 In an<br />

immunocompromised child with suspected overwhelming<br />

infection, rapid diagnosis can be made<br />

from a duodenal aspirate, although larvae are<br />

abundant with hyperinfection so diagnosis is less<br />

difficult than with latent infection. S. fuelleborni<br />

infection can be diagnosed from abundant eggs<br />

shed in the feces.<br />

Owing to the unreliability of stool microscopy and<br />

the importance of detecting even low levels of<br />

infection, various serological tests are available.<br />

An ELISA test for IgG is available, but there is still<br />

controversy about its reliability, because of its low<br />

specificity and positive predictive value. 130,131 The<br />

main problems with antibody tests are differentiating<br />

current from past infection, cross-reactivity<br />

with other helminth infections and false negatives<br />

on presentation of acute diarrheal disease in children<br />

(when stool is more reliable). Improved diagnostic<br />

tests are being developed using more<br />

specific antigens and specialized techniques,<br />

which have better potential to detect a risk<br />

of hyperinfection with immunosuppressive<br />

therapy. 129<br />

Prevention<br />

Disposal of human excreta, wearing of shoes, treatment<br />

of cases and improved hygiene reduce the<br />

risk of transmission of strongyloidiasis in communities.<br />

Regular mass chemotherapy programs<br />

against all geohelminths (e.g. albendazole) may<br />

have a modest impact on strongyloidiasis, but less<br />

than for the other helminths.<br />

Amebiasis<br />

Introduction<br />

Recent molecular and immunological techniques<br />

have demonstrated two distinct species of<br />

Entamoeba that are morphologically identical. E.<br />

histolytica is pathogenic, causing symptomatic<br />

disease in 10% of infections whereas E. dispar<br />

causes only asymptomatic colonization. In addition<br />

to the E. histolytica strain, other risk factors for<br />

invasive disease are interaction with bacterial<br />

flora, host genetic susceptibility, malnutrition,<br />

male sex, young age and immunodeficiency.<br />

Entamoeba coli and E. hartmanni are also nonpathogenic<br />

morphologically distinct members of<br />

the genus.<br />

Life cycle<br />

The E. histolytica life cycle consists of an infective<br />

cyst (10–15µm in diameter) and an invasive<br />

trophozoite (10–60µm in diameter). Cysts are<br />

resistant to chlorination, gastric acidity and desiccation<br />

and can survive in a moist environment for<br />

several weeks. For transmission of E. histolytica,<br />

cysts are ingested from fecally contaminated food<br />

and water and encystation occurs in the intestine,<br />

transforming the cyst into eight trophozoites.<br />

Mucosal invasion by trophozoites leads to colonic<br />

ulcers with migration of parasites to the liver via<br />

the portal vein.<br />

Epidemiology<br />

It is estimated that 500 million people are infected<br />

with E. histolytica in the world with 50 million per<br />

year developing invasive disease and 50000–<br />

100000 deaths. The case/fatality rate is 1 per<br />

500–1000 diagnosed cases, but in a hospital<br />

setting, the case/fatality ratio in children is higher,<br />

with 9% mortality and 27% morbidity. Liver<br />

abscess and extra-abdominal amebiasis has a<br />

much higher mortality rate of 10–40% or up to<br />

90% for cerebral amebiasis.<br />

The epidemiology of amebiasis has been complicated<br />

by the difficulty of identifying two genetically<br />

identical but morphologically distinct<br />

species, E. histolytica and E. dispar. It is estimated<br />

that 10% of the population is colonized by<br />

Entamoeba but 90% of these are with nonpathogenic<br />

E. dispar. Although E. dispar distribution<br />

is worldwide, E. histolytica infection occurs<br />

predominantly in Central and South America,<br />

Africa and the Indian subcontinent. Prevalence


studies based on stool parasites measure predominantly<br />

E. dispar, whereas serological surveys<br />

reflect the incidence of E. histolytica, since E.<br />

dispar does not cause seroconversion.<br />

A Bangladeshi study of urban children found the<br />

prevalence of E. histolytica to be 4.2% compared to<br />

6.5% for E. dispar with the highest prevalence of E.<br />

histolytica infection in children with diarrhea aged<br />

6–14 years. 132 A Mexican national survey reported<br />

an 8.4% seropositivity to E. histolytica, peaking at<br />

11% in the 5–9-year-old age group, with 4.2% of<br />

diarrheal cases due to E. histolytica infection, and<br />

6.5% with E. dispar. 132 Children 6–14 years of age<br />

with diarrhea had the highest incidence of E.<br />

histolytica infection (8%), whereas rural asymptomatic<br />

children had a 1% prevalence of E. histolytica<br />

and 7% prevalence of E. dispar. Shigella<br />

(dysenteriae and flexnerii) were also more frequent<br />

in children with diarrhea who also had Entamoeba<br />

infection. A West African study in school children<br />

found a ratio of E. histolytica : E.dispar of 1 : 46<br />

(compared to 1 : 3–9 in Asian studies), suggesting<br />

that the vast majority of Entamoeba infections in<br />

this region are not pathogenic. 133 A Brazilian study<br />

documented a 10.6% colonization rate for E.<br />

histolytica (excluding E. dispar), affecting predominantly<br />

those of poor socioeconomic status,<br />

hygiene and nutritional status, but most infections<br />

were asymptomatic and self-limited (clearing<br />

within 30–45 days) and reinfection rates were<br />

low. 134<br />

Pathophysiology<br />

Amebic disease occurs when trophozoites invade<br />

colonic tissue, which is initiated by prior adherence<br />

to mucins lining the surface of the large<br />

bowel followed by enzymatic destruction of the<br />

basement membrane and underlying tissue. The<br />

resulting inflammatory response contributes<br />

further to tissue destruction. The stages of amebic<br />

infection include: adherence to bacteria (especially<br />

Escherichia coli) and intestinal epithelial<br />

cells via galactose-binding lectin, which protects<br />

the parasite by blocking complement; activation of<br />

virulence factors in parasites such as cysteine<br />

proteinases; stimulation of intestinal epithelial<br />

cells to produce cytokines and inflammatory mediators<br />

by activation of NF-κB; the resulting release<br />

of chemokines leading to neutrophil influx; medi-<br />

Amebiasis 177<br />

ators greatly contributing to the destruction of host<br />

tissues by lytic necrosis and apoptosis, enabling<br />

amebic invasion. 135<br />

Cell-mediated immunity limits the extent of invasive<br />

amebiasis and protects the host from recurrence,<br />

including trophozoite killing by activated<br />

macrophages and cytotoxic lymphocytes.<br />

Cytokines such as TNF-α and interferon-γ<br />

contribute to immunocompetent cell activation.<br />

Innate resistance to infection in children is linked<br />

to the absence of serum anti-trophozoite IgG,<br />

which is inherited, whereas acquired resistance is<br />

linked to intestinal IgA and serum antibodies to a<br />

parasite lectin. 136,137<br />

The initial colonic lesion in amebiasis is a small<br />

intraglandular ulcer of only 1 mm, which extends<br />

only to the muscularis mucosa. Margins may be<br />

hyperemic with slight edema of the surrounding<br />

mucosa. The next stage involves deeper buttonhole<br />

ulcers which are up to 1 cm in diameter and<br />

extend into the sub-mucosa. In young children this<br />

can progress to a fulminant necrotizing colitis<br />

associated with transmural necrosis. Amebomas<br />

are uncommon in children but involve the formation<br />

of granulation tissue with a fibrous outer wall,<br />

which may result in stricture or obstruction to the<br />

lumen. Dissemination to the liver occurs in a high<br />

proportion of patients with fulminant disease.<br />

Clinical features<br />

The carrier state is the most common form of<br />

amebic infestation with all E. dispar infections and<br />

up to 90% of E. histolytica infections remaining<br />

asymptomatic with only cysts in the feces. Amebic<br />

dysentery is the most common form of symptomatic<br />

disease, with gradual onset of symptoms<br />

over 3 or 4 weeks after infection with increasingly<br />

severe diarrhea with abdominal pain such that an<br />

acute abdomen is often suspected. Stools contain<br />

blood and mucus, fever occurs in about half, and a<br />

small proportion develop abdominal distension<br />

with dehydration. Occasionally, young children<br />

present in a more fulminant manner with intussusception,<br />

perforation, peritonitis or necrotizing<br />

colitis. Rarely, children with amebic dysentery<br />

may present with an abdominal mass that has an<br />

‘apple-core’ appearance on radiographs, similar to<br />

colonic carcinoma. Amebic liver abscess and


178<br />

Intestinal parasites<br />

extra-abdominal amebiasis are much less common<br />

in children than in adults.<br />

Diagnosis<br />

The diagnosis of amebiasis should be considered<br />

in any child with blood or mucus in the stools,<br />

particularly if associated with abdominal pain or<br />

distension. Stool microscopy will yield cysts in<br />

< 30% of infected individuals, owing to the intermittent<br />

nature of cyst shedding, although multiple<br />

examinations increase the diagnostic yield to<br />

60–70% with amebic colitis but to under half of<br />

amebic hepatic abscesses.<br />

Serology can be helpful with invasive amebiasis<br />

since asymptomatic infections with E. dispar do<br />

not usually elicit a serological response. The gold<br />

standard remains culture or iso-enzyme analysis,<br />

but stool antigen detection tests are now commercially<br />

available which are more reliable than<br />

microscopy. A PCR-based amplification technique<br />

is now available for stool samples; infecting<br />

isolates are genetically diverse, with differences<br />

between those associated with intestinal and<br />

hepatic disease. 138 The major problem with serological<br />

tests is that they remain positive for years<br />

after an episode of amebiasis, so they may not be<br />

useful in endemic areas.<br />

Wet preparations of material aspirated or scraped<br />

from the base of ulcers on colonoscopy can be<br />

examined for motile trophozoites and tested for E.<br />

histolytica antigen. The appearance of amebic<br />

colitis resembles inflammatory bowel disease with<br />

a granular, friable and diffusely ulcerated mucosa.<br />

It is preferable for biopsy specimens to be taken<br />

from the edge of ulcers, but the sensitivity of this<br />

method of diagnosis appears variable.<br />

Prevention<br />

Prevention requires interruption of the fecal–oral<br />

spread of the infectious cyst stage of the parasite<br />

by improved hygiene, sanitation and water<br />

treatment. This is of course very difficult in the<br />

developing world. Current efforts towards a<br />

vaccine are focusing on the adherence lectin,<br />

which may prevent colonization. This would lead<br />

to elimination of the parasites if effective, since<br />

humans are the only significant reservoir of E.<br />

histolytica infection.<br />

Other parasites<br />

Hymenolepiasis<br />

Hymenolepis nana is the dwarf tapeworm and the<br />

only human tapeworm that does not require an<br />

intermediate host. Nevertheless, rodent strains for<br />

H. nana are infectious for humans including pet<br />

rodents such as rats, mice and hamsters. It occurs<br />

worldwide, with high childhood prevalences<br />

(> 10%) reported from Argentina, Peru, Brazil,<br />

Egypt, Pakistan and Zimbabwe. 41 Transmission is<br />

mostly fecal–oral, either from person to person or<br />

in food and water, with high rates in children from<br />

orphanages. 139<br />

Adult worms are only 15–45 mm in length with<br />

the spherical/ovoid eggs measuring 30–45µm in<br />

diameter, which do not survive beyond 11 days<br />

and are sensitive to heat and drying. In children<br />

who ingest eggs, the larval stage develops in the<br />

intestinal villi in 4–5 days, then break into the<br />

lumen and develop into adult worms, which<br />

produce ova in the feces within a month or less.<br />

The larval stage of H. nana is a cyst-like structure<br />

about 250 µm in diameter and is called a cysticercoid.<br />

As for other intestinal parasites, transmission<br />

is enhanced by poor hygiene and overcrowding,<br />

but control programs have found mass treatment<br />

more effective than improved hygiene. 41<br />

Most infected children are asymptomatic. High<br />

worm burdens of over 3000 may be associated<br />

with abdominal pain, loose stools and growth<br />

retardation. Disseminated larval infection has<br />

been described but is rare. Diagnosis is based on<br />

the characteristic eggs in stool. A single dose of<br />

praziquantel 20mg/kg is effective. Niclosamide<br />

60–80mg/kg per day for 5 days or paromomycin<br />

45mg/kg per day for 7 days are alternatives, but<br />

compliance is more difficult to ensure. Retreatment<br />

10 days later may be necessary to ensure<br />

eradication of cysticercoids in tissues.<br />

Enterobiasis<br />

Enterobius vermicularis is the common pinworm<br />

or threadworm of the large bowel. Unlike most<br />

other helminthic infection, enterobiasis may be<br />

more common in temperate, developed countries<br />

than in poor, tropical, developing countries. For<br />

example, rates in Australian schoolchildren have


een reported as 43% compared to 12% in India.<br />

However, it is worldwide in distribution and is<br />

almost exclusively transmitted person to person.<br />

The organism is a small white roundworm with<br />

the adult female worm 9–12mm long with the<br />

male worm only 2.5mm long and rarely observed.<br />

The worm’s lifespan is 5–13 weeks for females and<br />

only 7 weeks for males, but reinfection is common.<br />

The oval eggs laid by the gravid female on the anus<br />

are 50 x 25µm and the diagnosis is based on observation<br />

of these typical eggs from sticky tape<br />

applied to the perianal skin and then examined<br />

under a microscope. Adult pinworms or their eggs<br />

are rarely found in the feces, so this tape test for<br />

demonstrating pinworm eggs on the perianal skin<br />

is the most reliable technique.<br />

Humans are the only host for E. vermicularis, and<br />

worms inhabit the lumen of the cecum and appendix.<br />

The gravid adult female migrates to the perianal<br />

skin where she deposits her eggs resulting in<br />

intense anal pruritus, itching of the perianal skin<br />

leads to the contamination of the fingernails with<br />

the infective eggs. Each mature female worm can<br />

produce up to 11000 eggs. Most infections are<br />

asymptomatic, but perianal pruritus is the most<br />

common symptom of enterobiasis. Eosinophilia is<br />

uncommon, since the organism does not generally<br />

invade tissues. The incubation period is 3–4<br />

weeks.<br />

There is no definitive evidence that pinworms<br />

cause appendicitis, since most pinworms encountered<br />

in the appendix are an incidental finding,<br />

occurring in as many normal as inflamed appendices<br />

after surgery. However, it is impossible to<br />

exclude the possibility that chronic inflammatory<br />

changes due to local pinworm infection may<br />

contribute to some cases of appendicitis. Large<br />

numbers of larval pinworms have been shown to<br />

cause eosinophilic enterocolitis in an adult. 140 The<br />

most common site of infection (86.5%) is the<br />

lumen of the appendix, where the worms invoke<br />

no histological reaction. Other sites of infection<br />

include the abdominal and pelvic peritoneum and<br />

female genital tract. 141 Little girls sometimes<br />

present with ectopic pinworm infection of the<br />

vagina, which can cause hysterical reactions.<br />

The treatment of choice is albendazole or mebendazole,<br />

but ivermectin, pyrantel and levamisole<br />

are also effective. Since reinfection is very<br />

common, it is wise to readminister the drugs 2<br />

Other parasites 179<br />

weeks later and treat all household members,<br />

particularly children. Prevention of enterobiasis<br />

rests largely on treating infected children, keeping<br />

nails short and possibly improving personal<br />

hygiene, although infection is not an indicator of<br />

poor hygiene.<br />

Schistosomiasis<br />

There are seven human species of this trematode,<br />

including S. haematobium which affects the renal<br />

tract and S. mansoni the gastrointestinal tract. It<br />

has been estimated that 220 million people in 74<br />

countries are infected by schistosomiasis, and 20<br />

million have severe disease. Eggs of S. mansoni are<br />

passed in the feces, and hatch in warm water; the<br />

ciliated larvae swim and penetrate fresh-water<br />

snails (Biomphalaria). After a sporocyst stage, by<br />

4–5 weeks of infection, the larval phase results in<br />

thousands of cercariae (300 x 60µm) which penetrate<br />

human skin in water and enter peripheral<br />

lymphatics or veins and are carried to the lungs<br />

and mature in portal vessels. Adult worms migrate<br />

to the liver and mesenteric veins, where they may<br />

survive for 2–5 years or longer, producing eggs<br />

25–28 days after cercarial infection.<br />

Eggs cause granuloma formation resulting in localized<br />

colitis and hepatitis. The acute phase of S.<br />

mansoni infection may cause allergic symptoms<br />

(Katayama syndrome) which are rarely recognized<br />

in children. Most chronic infections are light and<br />

asymptomatic, but with heavy infections up to half<br />

of the eggs become trapped in the mucosa and<br />

submucosa of the colon, resulting in granulomatous<br />

reactions with significant blood loss. The<br />

host’s inflammatory reaction to eggs carried to the<br />

liver in the portal veins leads to portal hypertension.<br />

Severe disease with hepatosplenomegaly<br />

affects about 10% of S. mansoni cases in endemic<br />

areas, taking 5–15 years to develop.<br />

Diagnosis is based on finding eggs in the feces, but<br />

stool concentration methods and numerous<br />

immunological techniques (ELISA, immunoblotting)<br />

are more sensitive for milder infections.<br />

Treatment is with praziquantel 40 mg/kg as a<br />

single dose. Prevention involves avoidance of<br />

water sources containing cercariae and promotion<br />

of latrine use. Control programs for schistosomiasis<br />

involve mass chemotherapy, destruction of<br />

snails, environmental sanitation, prevention of


180<br />

Intestinal parasites<br />

water contact, health education and the future<br />

development of vaccines.<br />

Fasciolopsiasis<br />

The giant intestinal fluke Fasciolopsis buski is a<br />

class of flatworm called trematodes, which<br />

includes schistosomes. It is estimated that more<br />

than 40 million people have food-borne trematode<br />

infections. Those affecting the liver and lungs will<br />

not be considered here. The adult worm of F. buski<br />

measures up to 7.5cm in length and attaches to the<br />

mucosa of the proximal small intestine. It has a<br />

lifespan of about 6 months and begins producing<br />

eggs about 3 months after infection. These are<br />

excreted in the stool and measure 130µm in<br />

length. Up to 25000 eggs may be excreted daily.<br />

After several weeks in fresh water, the larvae hatch<br />

from the eggs and penetrate snails, where they<br />

undergo further development. Cercariae emerge<br />

from the snail after 1–2 months and encyst on a<br />

wide variety of aquatic vegetation. When humans<br />

ingest these raw, they develop infection, which<br />

lodges in the duodenum. The severity of symptoms<br />

correlates with the number of parasites, but<br />

heavy infections may cause nausea, vomiting,<br />

abdominal pain, edema, eosinophilia and poor<br />

nutrition. The diagnosis of fasciolopsiasis is based<br />

upon stool microscopy identifying the eggs. These<br />

are indistinguishable from those of the liver fluke,<br />

Fasciola hepatica. However, the intestinal fluke<br />

has a short lifespan and infection does not persist<br />

beyond 9 months from the time of departure from<br />

and endemic area.<br />

The treatment of choice is praziquantel 25 mg/kg<br />

three times daily for 1 day. The prognosis is generally<br />

excellent, except for heavy infection in children<br />

with intestinal obstruction of edematous<br />

malnutrition. The infection can be prevented by<br />

cooking aquatic vegetation or immersing plants or<br />

nuts in boiling water. The use of human feces as<br />

fertilizer in aquaculture is a major cause of human<br />

infection.<br />

Trichostrongylus<br />

Species of Trichostrongylus (wireworms) mostly<br />

affect ruminants, but they affect humans in close<br />

proximity to cattle, particularly in Africa,<br />

Australia, India, Iran and South America. The<br />

small adult worms (5–10 mm long) attach to the<br />

wall of the duodenum and jejunum after ingestion,<br />

and excrete eggs similar to those of the hookworm.<br />

Light infections are asymptomatic, but abdominal<br />

pain, diarrhea and eosinophilia may occur with<br />

heavy infections. Treatment is as for hookworm,<br />

and prevention involves thorough washing or<br />

cooking of vegetables.<br />

Parastrongylus costaricensis<br />

This nematode was discovered in Costa Rica in<br />

1971 and appears to be mainly confined to Latin<br />

America and the Caribbean. The natural host is the<br />

cotton rat but adult worms (20mm long) have been<br />

found in the cecum of children with appendicitislike<br />

illnesses with eosinophilia. Infection occurs<br />

by ingesting infected slugs. Diagnosis before<br />

surgery is based upon eosinophilia, radiology or<br />

serology, but larvae are not excreted in stool and<br />

no chemotherapy is effective.<br />

Cyclospora cayetanensis<br />

Cyclospora has emerged recently as an important<br />

protozoal pathogen causing diarrheal disease. 142 It<br />

appears to be endemic in most parts of the<br />

Americas, Africa and Asia, with infections occurring<br />

in travelers, as well as waterborne and foodborne<br />

outbreaks. 143–145 Children in tropical areas<br />

appear to have a high prevalence, and the disease<br />

peaks in the hot rainy season. 146,147 A Venezuelan<br />

study documented prevalences in adult AIDS<br />

patients and children with diarrhea of 9.8% vs.<br />

5.3%, which was more common than S. stercoralis<br />

(4.2% vs. 1.5%, respectively) but less common<br />

than C. parvum (35.2% vs. 9.8%, respectively) and<br />

had a peak prevalence in children between 2 and 5<br />

years of age. 148 Other childhood diarrheal prevalences<br />

have been similar in Nepal and Guatemala,<br />

but higher in Peru and Haiti (up to 18–20%). The<br />

incidence in Peruvian periurban children was<br />

0.21–0.28 episodes/child-years. 84 A Haitian study<br />

found a prevalence of 11% in AIDS diarrhea, with<br />

symptoms identical to isosporiasis and cryptosporidiosis.<br />

149<br />

Since other pathogens are often found, it is difficult<br />

to characterize the illness exactly, but<br />

prolonged watery diarrhea with weight loss is


typical, often associated with fever. Mucosal biopsies<br />

show inflammatory changes with partial villus<br />

atrophy with crypt hyperplasia. 150 Stool oocytes<br />

are sparse in infected immunocompetent children<br />

with diarrhea (


182<br />

Intestinal parasites<br />

fumarate reductase. It is poorly soluble in water,<br />

but well absorbed with a fatty meal. It is rapidly<br />

metabolized in the liver to the active form, albendazole<br />

sulfoxide, which has a serum half-life of<br />

8–9 h, and is excreted by the kidneys. It is usually<br />

very well tolerated as a single dose or daily for 3<br />

days, with gastrointestinal symptoms (e.g. pain,<br />

diarrhea, nausea or vomiting) in only 1.3% of<br />

courses. 156 Worm migration is uncommon with<br />

albendazole treatment, but prolonged therapy may<br />

cause alopecia, reversible marrow suppression or<br />

hepatocellular damage. Albendazole should be<br />

avoided in pregnancy and in children under 6<br />

months of age.<br />

Albendazole 400mg (200mg if


Levamisole<br />

Levamisole is an immune stimulant which is effective<br />

against Ascaris and hookworm, and may be<br />

more effective for intestinal obstruction from<br />

roundworms, since it acts by paralyzing the<br />

myoneural junction of the worm. The dosage is<br />

3mg/kg as a single dose.<br />

Ivermectin<br />

Ivermectin has broad-spectrum activity against<br />

helminths and filariasis, but is the drug of choice<br />

against strongyloidiasis. Ivermectin is well<br />

absorbed orally, accumulating in adipose tissue,<br />

metabolized in the liver, highly protein bound<br />

with a serum half-life of 12 h and excreted in stool.<br />

It is generally well tolerated, with occasional<br />

abdominal distension, chest tightness or wheezing.<br />

In a Tanzanian study of children, ivermectin<br />

was found to be more effective than albendazole<br />

for curing Strongyloides, equally effective for<br />

Ascaris, less effective for Trichuris and ineffective<br />

against hookworm infections. 163 Another African<br />

study of ivermectin in intestinal nematode infections<br />

found little effect on either prevalence or<br />

intensity of N. americanus hookworm or Trichuris,<br />

and only a modest effect on Ascaris. 164<br />

Ivermectin 200µg/kg as a single dose has a<br />

reported cure rate of 83% for Strongyloides. In<br />

complicated or disseminated infection, ivermectin<br />

should be repeated on days 2, 15 and 16 to<br />

decrease relapse. In immunosuppressed patients,<br />

treatment is not always successful, and may need<br />

to be repeated at monthly intervals, or a longer<br />

course given. An adult open-label study of 60<br />

patients with strongyloidiasis compared singledose<br />

ivermectin with albendazole for 3 days and<br />

found higher cure rates with the former (83% vs.<br />

38%). 165 Another adult study comparing ivermectin<br />

with thiabendazole documented high cure<br />

rates for both drugs but the adverse effects were<br />

much greater for thiabendazole (e.g. disorientation,<br />

fatigue, nausea and anorexia). 166 A comparative<br />

trial in 301 children from Zanzibar with S.<br />

stercoralis larvae in stool had cure rates of 82.9%<br />

with single-dose ivermectin compared to only<br />

45.0% with 3 days of albendazole. 163 Disseminated<br />

disease has a high mortality, so may require ivermectin<br />

over 3–4 weeks.<br />

Metronidazole<br />

Drug treatment 183<br />

Metronidazole is used for giardiasis and amebiasis.<br />

It is activated by reduction of its 5-nitro group, is<br />

concentrated in anaerobic organisms and interacts<br />

with DNA to cause microbial death. It is not well<br />

tolerated, with anorexia and gastrointestinal upset<br />

occurring after several days’ treatment. 167 The<br />

dose of metronidazole has been controversial, but<br />

a computer simulation study using the conventional<br />

30 mg/kg per day regime calculated that<br />

steady state was reached at 24mg/kg per day in<br />

rehabilitated children and at 12mg/kg per day in<br />

severely malnourished children using the twice<br />

daily dosage. 168 Patients with immunodeficiency<br />

may require treatment for 6–8 weeks for giardiasis.<br />

The American Academy of Pediatrics and Centers<br />

for Disease Control recommend that asymptomatic<br />

Giardia infections not be treated. 169,170<br />

Asymptomatic cyst excretors, even at day-care<br />

centers, do not warrant treatment. 170,171<br />

Amebic colitis or dysentery should be treated with<br />

metronidazole plus a luminal agent such as diloxanide<br />

furoate, paromomycin or iodoquinol.<br />

Although improvement usually occurs within 3–4<br />

days of treatment, metronidazole should be<br />

continued for a minimum of 10 days to eliminate<br />

intestinal colonization with risk of relapse.<br />

Treatment of asymptomatic cyst passers is inappropriate<br />

for E. dispar, but is warranted for E.<br />

histolytica in developed countries but probably not<br />

practical in most of the developing world where<br />

the disease in endemic. The dosage for treatment<br />

of acute amebic colitis is metronidazole<br />

35–50mg/kg per day in three divided doses for 10<br />

days. The intraluminal agents are used for eradicating<br />

cyst passage. The dosages are: iodoquinol<br />

30–40mg/kg per day in three divided oral doses for<br />

20 days, or diloxanide furoate 20mg/kg per day in<br />

three divided doses, or paromomycin 25–35mg/kg<br />

per day in three divided doses for 7 days.<br />

Tinidazole<br />

Tinidazole is another 5-nitroimidazole with a<br />

similar mechanism of action to that of metronidazole<br />

but a convenient single dosage. A Cochrane<br />

review of 34 trials on drugs for treating giardiasis<br />

concluded that tinidazole 50mg/kg in a single dose<br />

had a higher clinical cure rate than a short course


184<br />

Intestinal parasites<br />

of treatment with metronidazole, albendazole or<br />

secnidazole. 172<br />

Nitazoxanide<br />

Nitazoxanide is a new broad-spectrum antimicrobial<br />

agent with activity against nematodes, trematodes,<br />

anaerobic bacteria and protozoal parasites<br />

such as Cryptosporidium. It is metabolized in<br />

blood to tizoxanide, which inhibits the key<br />

enzyme pyruvate ferredoxin oxidoreductase of<br />

target organisms, and is excreted in urine and<br />

feces. Adverse effects tend to affect the gastrointestinal<br />

tract, but appear to be mild and transient.<br />

173 A 3-day course of 100–200mg 12-hourly<br />

in children (adults 500mg) is effective against giardiasis,<br />

amebiasis, Blastocystis hominis,<br />

Balantidium coli, Isospora belli, Ascaris, Trichuris,<br />

hookworm and Hymenolepis nana. 168 In view of<br />

this wide spectrum of action, single-dose therapy<br />

in combination with other drugs is under investigation<br />

for community treatment programs.<br />

Two Egyptian randomized placebo-controlled<br />

trials of diarrheal subjects have shown that nitazoxanide<br />

was associated with resolution of the<br />

diarrhea within 7 days in about 80% of cases of<br />

giardiasis, amebiasis or cryptosporidiosis<br />

compared to about 40% in the placebo group. 174,175<br />

A Mexican study of 275 children with helminth or<br />

protozoan infections documented a higher (but<br />

non-significant) parasite eradication rate with 3<br />

days of nitazoxanide (79%) than with 3 days of<br />

mebendazole plus quifamide (74%). 176 Another<br />

Mexican study has documented its in vitro effectiveness<br />

against trophozoites of E. histolytica and<br />

Giardia. 177 Nitazoxanide was best for eradication<br />

of H. nana (90–100%) and worst for giardiasis<br />

(56–74%). Comparative studies of Peruvian children<br />

with ascariasis, trichuriasis, hymenolepiasis<br />

and giardiasis reported cure rates for nitazoxanide<br />

of 89%, 89%, 82% and 85%, respectively, compared<br />

with 91%, 58%, 96% and 75–80% for albendazole,<br />

praziquantel (H. nana) or metronidazole<br />

(Giardia), respectively. 178,179 A recent Zambian<br />

trial of 3 days of nitazoxanide (vs. placebo) in 100<br />

children with cryptosporidiosis showed clinical<br />

response rates of 56 vs. 23% and parasitological<br />

response rates of 52 vs. 14% in HIV-seronegative<br />

children, but no significant response in HIVseropositive<br />

cases. 180<br />

Paromomycin<br />

Paromomycin is a poorly absorbed aminoglycoside<br />

which reaches high concentrations in the intestine,<br />

but absorbed drug is excreted renally and is<br />

potentially ototoxic and nephrotoxic. It has been<br />

used for cryptosporidial diarrhea in AIDS patients,<br />

requiring continuous maintenance therapy, but<br />

appears now to be ineffective. 181,182<br />

Mass community anthelminthic<br />

treatment<br />

UNICEF, the World Bank and WHO have promoted<br />

routine mass anthelminthic treatment programs as<br />

a cost-effective intervention, and school-based<br />

programs are popular. 183–185 Although most<br />

studies report prevalence, reducing the intensity of<br />

the worm burden is the major aim of control<br />

programs in children, who have the highest<br />

burden. Albendazole and praziquantel have broadspectrum<br />

anthelminthic activity against ascariasis,<br />

trichuriasis, enterobiasis, hookworm, giardiasis,<br />

strongyloidiasis and schistosomiasis at relatively<br />

low cost and with low rates of resistance to these<br />

agents. Mass treatment protocols may reduce the<br />

risk of drug resistance by targeting school-age and<br />

preschool children, by only repeating treatment at<br />

intervals greater than the nematode generation<br />

time, by combining anthelminthic drugs in control<br />

programs to reduce or delay selection for resistance,<br />

and by monitoring drug resistance to benzimadoles<br />

using DNA probes. 2 Deworming programs<br />

were held back for young children because of<br />

concerns about their safety in children under 2<br />

years of age (as well as in pregnancy and lactation).<br />

However, a recent WHO Informal Consultation<br />

that children over 12 months of age should be<br />

included in deworming campaigns using praziquantel<br />

and albendazole/mebendazole on the basis<br />

of improved safety data and risk–benefit analysis.<br />

186<br />

The impact of helminth infections on growth and<br />

development is a controversial but important<br />

issue. A Cochrane systematic review of 30<br />

randomized trials involving 15000 children on the<br />

effects of antihelminthic treatment of endemic<br />

communities on growth and cognitive performance<br />

was reported in 2000. 187,188 It found only


modest weight gain of 0.1kg (0.04–0.17) after a<br />

year of follow-up and no differences in cognitive<br />

performance, so concluded that the evidence for<br />

routine anthelminthic treatment to improve<br />

growth and cognitive performance was unconvincing.<br />

Critics of the review pointed out that poorly<br />

designed trials may have failed to document an<br />

effect, that short-term treatment cannot assess the<br />

long-term benefits of regular treatment and that<br />

the review hid the greatest effect of anthelminthic<br />

treatment on growth and development in children<br />

with heavy parasitic burdens.<br />

Since the Cochrane review, there has been a study<br />

of 614 children 12–48 months at baseline, in a<br />

community of high parasite intensity in Tanzania<br />

who were randomized to single-dose mebendazole<br />

500 mg 3-monthly vs. placebo and followed for 12<br />

months. It reported modest reductions in prevalence<br />

of Trichuris (75 vs. 58%), Ascaris (49 vs. 27%)<br />

and hookworm (66 vs. 60%), but the effect on<br />

worm intensity was more significant, reducing<br />

Ascaris from 126 to 12 eggs per gram of feces (epg),<br />

Trichuris from 511 to 88 epg and hookworm from<br />

198 to 119 epg. 189 A large randomized mass treatment<br />

trial of school children with albendazole and<br />

praziquantel in China, Kenya and the Philippines<br />

documented significant reductions (p < 0.0001) in<br />

mean egg counts at 45 days for hookworm (97 to<br />

27), Ascaris (5903 to 626) and Trichuris (233 to<br />

161), but the effects were limited by rapid reinfection<br />

(Ascaris) and single-dose albendazole ineffectiveness<br />

(Trichuris). 190 The most important shortterm<br />

benefits at 6 months from treatment were<br />

modest rises in hemoglobin level and reductions<br />

in hepatomegaly (Kenya only) in the children<br />

treated with praziquantel, but catch-up growth did<br />

not occur in treated children compared to controls.<br />

Another study in the low-intensity setting of<br />

Bangladesh followed 123 children aged 2–5 years<br />

for 12 months with 2-monthly mebendazole and<br />

showed reduced prevalences of Trichuris (65 to<br />

9%), Ascaris (78 to 8%) and hookworm (4 to 0%)<br />

while associated with an increase in giardiasis (19<br />

to 49%), but failed to document improved growth<br />

or improvements in intermediate variables (intestinal<br />

permeability, plasma albumin, α1-antichymotrypsin).<br />

191 The increase in giardiasis with mebendazole<br />

was found in an earlier Bangladeshi<br />

study, 192 but could be prevented by substituting<br />

albendazole, which has anti-giardial activity.<br />

Mass community anthelminthic treatment 185<br />

Confirmation of the limited effect of helminths on<br />

growth has come from indirect calorimetry, which<br />

showed no treatment effect on energy metabolism<br />

with low-level hookworm, Ascaris, Trichuris or<br />

Strongyloides infections. 193 A Malaysian study<br />

found that intestinal helminths did not contribute<br />

to poor school attendance. 194<br />

A 9-year cohort study of 119 children in a<br />

Brazilian shantytown showed that, in this setting,<br />

the burden of diarrheal disease and helminth<br />

infections in children under 2 years were independently<br />

associated with stunting by 2–7 years of<br />

age, even controlling for confounders. 195 Thus,<br />

early childhood helminth infections were associated<br />

with a 4.6-cm shortfall by age 7 years,<br />

compared to 3.6cm attributed to the mean 9.1<br />

episodes of diarrhea before the age of 2 years.<br />

There is also indirect evidence that intestinal<br />

nematodes affect productivity in adults through<br />

both disease-related morbidity and ill-health in<br />

childhood. 196 A Zairian study of 358 moderately<br />

malnourished preschool children randomized<br />

them to either vitamin A, mebendazole 500 mg<br />

3-monthly or no treatment. 197 The vitamin Adeficient<br />

children showed significant catch-up<br />

growth on vitamin A treatment, but deworming<br />

did not improve growth, because only 12 of 123 in<br />

the mebendazole group had ascariasis of low<br />

intensity, making the claim in the title of the<br />

article misleading.<br />

A South African randomized trial of 428 schoolchildren<br />

with a high prevalence of Trichuris, hookworm<br />

and schistosomiasis found that combination<br />

treatment with 3 days of albendazole and singledose<br />

praziquantel 6-monthly and iron supplementation<br />

weekly for 10 weeks reduced worm prevalence<br />

and increased hemoglobin levels, but did not<br />

improve growth. 198 A Kenyan study showed that<br />

albendazole was more effective than mebendazole<br />

in reducing worm burdens in schoolchildren. 199<br />

However, a Tanzanian study of 2294 schoolchildren<br />

from a very high prevalence setting showed<br />

that single-dose albendazole vs. mebendazole<br />

reduced the parasite burden of Ascaris by 97% vs.<br />

97%, of Trichuris by 73% vs. 82%, and of hookworm<br />

by 98% vs. 82%, respectively. 200 Moreover,<br />

the high reinfection rate in this setting meant that<br />

the impact of chemotherapy was short-lived, so 4monthly<br />

treatment would be necessary to reduce<br />

long-term morbidity. 64 In South African school-


186<br />

Intestinal parasites<br />

children, a single dose of albendazole and praziquantel<br />

reduced prevalence rates of Ascaris,<br />

Trichuris and S. haematobium from 29.5 to 4.7%,<br />

51.9 to 38.0% and 22.3 to 3.3%, respectively.<br />

Invasive parasites which are known to cause<br />

malabsorption, weight loss or prolonged diarrhea,<br />

such as Giardia, Cryptosporidium and<br />

Strongyloides, are the most likely to affect growth,<br />

so specific studies have examined this in community<br />

settings. Giardia is the most prevalent intestinal<br />

protozoan parasite, and certainly can cause<br />

persistent diarrhea with malabsorption, weight<br />

loss and mucosal damage, but the key public<br />

health question is whether the high rates of infection<br />

without overt clinical symptoms contribute to<br />

poor growth of preschool children in the developing<br />

world. Lunn and colleagues examined this<br />

question in 60 infants in The Gambia where both<br />

giardiasis and poor growth are known to be highly<br />

prevalent. 201,202 Giardia-specific plasma immunoglobulins<br />

were used for diagnosis and did not<br />

explain the observed growth faltering. Giardia<br />

infection was not associated with diarrhea in this<br />

context, but mild infections may have caused<br />

minimal abnormalities in intestinal permeability<br />

and α1-antichymotrypsin, an acute-phase reactant<br />

protein. They concluded that giardiasis was<br />

unlikely to be a major cause of the poor growth of<br />

rural Gambian infants. Rapid reinfection after<br />

treatment was also documented for giardiasis in<br />

Egypt and Peru, where 98% of children were reinfected<br />

within 6 months of tinidazole treatment<br />

and stool excretion lasted a mean of 3.2<br />

months. 203,204<br />

REFERENCES<br />

1. Booth M, Bundy DA, Albonico M et al. Associations<br />

among multiple geohelminth species infections in<br />

schoolchildren from Pemba Island. Parasitology 1998;<br />

116: 85–93.<br />

2. Anonymous. Prevention and Control of Schistosomiasis<br />

and Soil-Transmitted Helminthiasis. WHO Technical<br />

Report Series 912. Geneva: World Health Organization.<br />

2002: 1–57.<br />

3. Yu SH, Xu LQ, Jiang ZX et al. Nationwide survey of<br />

human parasite in China. Southeast Asian J Trop Med<br />

Public Health 1994; 25: 4–10.<br />

4. Brooker S, Rowlands M, Haller L et al. Towards an atlas<br />

of human helminth infection in sub-Saharan Africa: the<br />

Finally, studies have shown that control of intestinal<br />

parasitic infections is more than a question of<br />

mass chemotherapy, but is influenced by social<br />

and cultural factors that affect human behavior,<br />

such as treatment seeking and promiscuous defecation.<br />

205,206<br />

Conclusion<br />

The most prevalent intestinal parasites associated<br />

with morbidity in children are the protozoa<br />

Cryptosporidium, Giardia and Entameoba histolytica,<br />

and the helminths Trichuris, Ascaris, hookworm<br />

and Strongyloides. Although most infected<br />

children remain asymptomatic, some children<br />

with high intensity of infection have manifestations<br />

of severe and persistent enteropathy, particularly<br />

with strongyloidiasis, cryptosporidiosis, giardiasis<br />

and trichuriasis. In spite of reductions in<br />

overall child mortality rates over recent decades,<br />

intestinal parasites remain highly prevalent in<br />

children in the developing world with no evidence<br />

of decreases in either global prevalence or morbidity,<br />

despite effective treatment being available.<br />

Recent advances in molecular biology and<br />

immunology of parasites are improving diagnostic<br />

techniques and open the way for the development<br />

of vaccines to control them. New vaccines and<br />

mass chemotherapy programs appear to be more<br />

effective approaches in the control of intestinal<br />

parasites in the short-term than improvements in<br />

hygiene, sanitation and living standards.<br />

use of geographical information systems (GIS). Parasitol<br />

Today 2000; 16: 303–307.<br />

5. The Partnership for Child Development. The health and<br />

nutritional status of schoolchildren in Africa: evidence<br />

from school-based health programmes in Ghana and<br />

Tanzania. Trans R Soc Trop Med Hyg 1998; 92: 254–261.<br />

6. Amin OM. Seasonal prevalence of intestinal parasites in<br />

the United States during 2000. Am J Trop Med Hyg 2002;<br />

66: 799–803.<br />

7. Booth M, Mayombana C, Machibya H et al. The use of<br />

morbidity questionnaires to identify communities with<br />

high prevalences of schistosome or geohelminth infections<br />

in Tanzania. Trans R Soc Trop Med Hyg 1998; 92: 484–490.


8. Tshikuka JG, Gray DK, Scott M, Olela KN. Relationship<br />

of childhood protein–energy malnutrition and parasite<br />

infections in an urban African setting. Trop Med Int<br />

Health 1997; 2: 374–382.<br />

9. Genta RM. Diarrhea in helminthic infections. Clin Infect<br />

Dis 1993; 16(Suppl 2): S122–S129.<br />

10. Kukuruzovic RH, Robins Browne RM, Anstey N,<br />

Brewster DR. Enteric pathogens, intestinal permeability<br />

and nitric oxide production in childhood diarrheal<br />

disease. Pediatr Infect Dis J 2002; 21: 730–739.<br />

11. Gillin FD, Reiner DS, Gault MJ et al. Encystation and<br />

expression of cyst antigens by Giardia lamblia in vitro.<br />

Science 1987; 235: 1040–1043.<br />

12. Lujan HD, Mowatt MR, Byrd LG, Nash TE. Cholesterol<br />

starvation induces differentiation of the intestinal<br />

parasite Giardia lamblia. Proc Natl Acad Sci USA 1996;<br />

93: 7628–7633.<br />

13. Touz MC, Nores MJ, Slavin I et al. Membrane-associated<br />

dipeptidyl peptidase IV is involved in encystationspecific<br />

gene expression during Giardia differentiation.<br />

Biochem J 2002; 364: 703–710.<br />

14. Thompson SC. Giardia lamblia in children and the<br />

child care setting: a review of the literature. J Paediatr<br />

Child Health 1994; 30: 202–209.<br />

15. Nuchprayoon S, Siriyasatien P, Kraivichian K et al.<br />

Prevalence of parasitic infections among Thai patients<br />

at the King Chulalongkorn Memorial Hospital, Bangkok,<br />

Thailand. J Med Assoc Thai 2002; 85 (Suppl 1):<br />

S415–S423.<br />

16. Molbak K, Wested N, Hojlyng N et al. The etiology of<br />

early childhood diarrhea: a community study from<br />

Guinea-Bissau. J Infect Dis 1994; 169: 581–587.<br />

17. Newman RD, Moore SR, Lima AA et al. A longitudinal<br />

study of Giardia lamblia infection in north-east<br />

Brazilian children. Trop Med Int Health 2001; 6:<br />

624–634.<br />

18. Mahmud MA, Chappell CL, Hossain MM et al. Impact<br />

of breast-feeding on Giardia lamblia infections in<br />

Bilbeis, Egypt. Am J Trop Med Hyg 2001; 65: 257–260.<br />

19. Chunge RN, Nagelkerke N, Karumba PN et al.<br />

Longitudinal study of young children in Kenya:<br />

intestinal parasitic infection with special reference to<br />

Giardia lamblia, its prevalence, incidence and duration,<br />

and its association with diarrhoea and with other<br />

parasites. Acta Trop Basel 1991; 50: 39–49.<br />

20. Nain CK, Dutt P, Vinayak VK. Alterations in enzymatic<br />

activities of the intestinal mucosa during the course of<br />

Giardia lamblia infection in mice. Ann Trop Med<br />

Parasitol 1991; 85: 515–522.<br />

21. Buret AG, Mitchell K, Muench DG, Scott KG. Giardia<br />

lamblia disrupts tight junctional ZO-1 and increases<br />

permeability in non-transformed human small intestinal<br />

epithelial monolayers: effects of epidermal growth<br />

factor. Parasitology 2002; 125: 11–19.<br />

22. Hicks SJ, Theodoropoulos G, Carrington SD, Corfield<br />

AP. The role of mucins in host-parasite interactions. Part<br />

I. Protozoan parasites. Parasitol Today 2000; 16:<br />

476–481.<br />

23. Pettoello MM, Guandalini S, Ecuba P et al. Lactose<br />

malabsorption in children with symptomatic Giardia<br />

lamblia infection: feasibility of yogurt supplementation.<br />

J Pediatr Gastroenterol Nutr 1989; 9: 295–300.<br />

24. Moya-Camarena SY, Sotelo N, Valencia ME. Effects of<br />

asymptomatic Giardia intestinalis infection on<br />

carbohydrate absorption in well-nourished Mexican<br />

children. Am J Trop Med Hyg 2002; 66: 255–259.<br />

25. Jove S, Fagundes-Neto U, Wehba J et al. Giardiasis in<br />

childhood and its effects on the small intestine. J<br />

Pediatr Gastroenterol Nutr 1983; 2: 472–7.<br />

References 187<br />

26. Hjelt K, Paerregaard A, Krasilnikoff PA. Giardiasis:<br />

haematological status and the absorption of vitamin B12<br />

and folic acid. Acta Paediatr 1992; 81: 29–34.<br />

27. Goto R, Panter-Brick C, Northrop-Clewes CA et al. Poor<br />

intestinal permeability in mildly stunted Nepali children:<br />

associations with weaning practices and Giardia<br />

lamblia infection. Br J Nutr 2002; 88: 141–149.<br />

28. Scott KG, Meddings JB, Kirk DR et al. Intestinal<br />

infection with Giardia spp. reduces epithelial barrier<br />

function in a myosin light chain kinase-dependent<br />

fashion. Gastroenterology 2002; 123: 1179–1190.<br />

29. Roberts Thomson IC, Mitchell GF, Anders RF et al.<br />

Genetic studies in human and murine giardiasis. Gut<br />

1980; 21: 397–401.<br />

30. Char S, Cevallos AM, Yamson P et al. Impaired IgA<br />

response to Giardia heat shock antigen in children with<br />

persistent diarrhoea and giardiasis. Gut 1993; 34: 38–40.<br />

31. Walterspiel JN, Morrow AL, Lourdes Guerrero M et al.<br />

Secretory anti-Giardia lamblia antibodies in human<br />

milk: protective effect against diarrhea. Pediatrics 1994;<br />

93: 28–31.<br />

32. Di Prisco MC, Hagel I, Lynch NR et al. Association<br />

between giardiasis and allergy. Ann Allergy Asthma<br />

Immunol 1998; 81: 261–265.<br />

33. Cevallos AM, Carnaby S, James M, Farthing MJ. Small<br />

intestinal injury in a neonatal rat model of giardiasis is<br />

strain dependent. Gastroenterology 2002; 109: 766–773.<br />

34. Jimenez JC, Rodriguez N, Di Prisco MC et al.<br />

Haemoglobin concentrations and infection by Giardia<br />

intestinalis in children: effect of treatment with<br />

secnidazole. Ann Trop Med Parasitol 1999; 93: 823–827.<br />

35. Lane S, Lloyd D. Current trends in research into the<br />

waterborne parasite Giardia. Crit Rev Microbiol 2002;<br />

28: 123–147.<br />

36. Matsuyama W, Mizoguchi A, Iwami F et al. [A case of<br />

pulmonary infiltration with eosinophilia caused by<br />

Ascaris suum]. Nihon Kokyuki Gakkai Zasshi 1998; 36:<br />

208–12.<br />

37. de Silva NR, Chan MS, Bundy DA. Morbidity and<br />

mortality due to ascariasis: re-estimation and sensitivity<br />

analysis of global numbers at risk. Trop Med Int Health<br />

1997; 2: 519–528.<br />

38. Hall A, Holland C. Geographical variation in Ascaris<br />

lumbricoides fecundity and its implications for<br />

helminth control. Parasitol Today 2000; 16: 540–544.<br />

39. Geissler PW, Mwaniki D, Thiong F, Friis H. Geophagy as<br />

a risk factor for geohelminth infections: a longitudinal<br />

study of Kenyan primary schoolchildren. Trans R Soc<br />

Trop Med Hyg 1998; 92: 7–11.<br />

40. Saathoff E, Olsen A, Kvalsvig JD, Geissler PW. Geophagy<br />

and its associates with geohelminth infection in rural<br />

schoolchildren from northern KwaZulu-Natal, South<br />

Africa. Trans R Soc Trop Med Hyg 2002; 96: 485–90.<br />

41. Muller R. Worms and Human Disease. 2nd edn.<br />

Wallingford, UK: CABI Publishing, 2002.<br />

42. Kennedy MW. The nematode polyprotein allergens/<br />

antigens. Parasitol Today 2000; 16: 373–380.<br />

43. Viney M. How do host immune responses affect<br />

nematode infections? Trends Parasitol 2002; 18: 63–66.<br />

44. Scott ME, Koski KG. Zinc deficiency impairs immune<br />

responses against parasitic nematode infections at<br />

intestinal and systemic sites. J Nutr 2000; 130:<br />

1412S–1420S.<br />

45. Cooper PJ, Chico M, Sandoval C et al. Human infection<br />

with Ascaris lumbricoides is associated with<br />

suppression of the interleukin-2 response to<br />

recombinant cholera toxin B subunit following<br />

vaccination with the live oral cholera vaccine CVD 103-<br />

HgR. Infect Immun 2001; 69: 1574–1580.


188<br />

Intestinal parasites<br />

46. Cooper PJ, Chico ME, Losonsky G et al. Albendazole<br />

treatment of children with ascariasis enhances the<br />

vibriocidal antibody response to the live attenuated oral<br />

cholera vaccine CVD 103-HgR. J Infect Dis 2000; 182:<br />

1199–1206.<br />

47. Wolday D, Mayaan S, Mariam ZG et al. Treatment of<br />

intestinal worms is associated with decreased HIV<br />

plasma viral load. J Acquir Immune Defic Syndr 2002;<br />

31: 56–62.<br />

48. Cooper PJ, Chico ME, Sandoval C et al. Human infection<br />

with Ascaris lumbricoides is associated with a<br />

polarized cytokine response. J Infect Dis 2000; 182:<br />

1207–1213.<br />

49. Behm CA,.Ovington KS. The role of eosinophils in<br />

parasitic helminth infections: insights from genetically<br />

modified mice. Parasitol Today 2000; 16: 202–209.<br />

50. Meeusen EN, Balic A. Do eosinophils have a role in the<br />

killing of helminth parasites? Parasitol Today 2000; 16:<br />

95–101.<br />

51. Lynch NR, Hagel I, Perez M et al. Effect of anthelmintic<br />

treatment on the allergic reactivity of children in a<br />

tropical slum. J Allergy Clin Immunol 1993; 92: 404–411.<br />

52. Perzanowski MS, Ng’ang’a LW, Carter MC et al. Atopy,<br />

asthma, and antibodies to Ascaris among rural and<br />

urban children in Kenya. J Pediatr 2002; 140: 582–588.<br />

53. Lynch NR, Hagel IA, Palenque ME et al. Relationship<br />

between helminthic infection and IgE response in<br />

atopic and nonatopic children in a tropical<br />

environment. J Allergy Clin Immunol 1998; 101:<br />

217–221.<br />

54. Gendrel D, Richard Lenoble D, Kombila M et al.<br />

Influence of intestinal parasitism on lactose absorption<br />

in well-nourished African children. Am J Trop Med Hyg<br />

1992; 46: 137–140.<br />

55. Raj SM, Sein KT, Anuar AK, Mustaffa BE. Effect of<br />

intestinal helminthiasis on intestinal permeability of<br />

early primary schoolchildren. Trans R Soc Trop Med Hyg<br />

1996; 90: 666–669.<br />

56. Chitkara RK, Sarinas PS. Dirofilaria, visceral larva<br />

migrans, and tropical pulmonary eosinophilia. Semin<br />

Respir Infect 1997; 12: 138–148.<br />

57. de Silva NR, Guyatt HL, Bundy DA. Morbidity and<br />

mortality due to Ascaris-induced intestinal obstruction.<br />

Trans R Soc Trop Med Hyg 1997; 91: 31–36.<br />

58. Carneiro FF, Cifuentes E, Tellez-Rojo MM, Romieu I.<br />

The risk of Ascaris lumbricoides infection in children as<br />

an environmental health indicator to guide preventative<br />

activities in Caparao and Alto Caparo, Brazil. Bull World<br />

Health Organ 2002; 80: 40–46.<br />

59. Olsen A, Samuelsen H, Onyango-Ouma W. A study of<br />

risk factors for intestinal helminth infections using<br />

epidemiological and anthropological approaches. J<br />

Biosoc Sci 2001; 33: 569–584.<br />

60. Adenusi AA. The distribution of Necator americanus<br />

and Ancylostoma duodenale among schoolchildren in<br />

Lagos, Nigeria. Trans R Soc Trop Med Hyg 1997; 91: 270.<br />

61. Walker NI, Croese J, Clouston AD et al. Eosinophilic<br />

enteritis in northeastern Australia. Pathology,<br />

association with Ancylostoma caninum, and<br />

implications. Am J Surg Pathol 1995; 19: 328–37.<br />

62. Sen L, Ghosh K, Bin Z et al. Hookworm burden<br />

reductions in BALB/c mice vaccinated with<br />

recombinant Ancylostoma secreted proteins (ASPs) from<br />

Ancylostoma duodenale, Ancylostoma caninum and<br />

Necator americanus. Vaccine 2000; 18: 1096–1102.<br />

63. Yu SH, Jiang ZX, Xu LQ. Infantile hookworm disease in<br />

China. A review. Acta Trop 1995; 59: 265–270.<br />

64. Albonico M, Smith PG, Ercole E et al. Rate of<br />

reinfection with intestinal nematodes after treatment of<br />

children with mebendazole or albendazole in a highly<br />

endemic area. Trans R Soc Trop Med Hyg 1995; 89:<br />

538–541.<br />

65. Olsen A, Nawiri J, Friis H. The impact of iron<br />

supplementation on reinfection with intestinal<br />

helminths and Schistosoma mansoni in western Kenya.<br />

Trans R Soc Trop Med Hyg 2000; 94: 493–499.<br />

66. Guyatt HL, Brooker S, Kihamia CM et al. Evaluation of<br />

efficacy of school-based anthelmintic treatments against<br />

anemia in children in the United Republic of Tanzania.<br />

Bull World Health Organ 2001; 79: 695–703.<br />

67. Beasley NM, Tomkins AM, Hall A et al. The impact of<br />

population level deworming on the haemoglobin levels<br />

of schoolchildren in Tanga, Tanzania. Trop Med Int<br />

Health 1999; 4: 744–750.<br />

68. Stephenson LS, Holland CV, Cooper ES. The public<br />

health significance of Trichuris trichiura. Parasitology<br />

2000; 121 (Suppl): S73–S95.<br />

69. Brown CJ, McCloskey DJ, Bundy DA, Navarrete CV.<br />

Parasitic infection with Trichuris trichiura influences<br />

plasma levels of soluble HLA class I. Hum Immunol<br />

1999; 60: 1067–1071.<br />

70. Needham C, Kim HT, Hoa NV et al. Epidemiology of<br />

soil-transmitted nematode infections in Ha Nam<br />

Province, Vietnam. Trop Med Int Health 1998; 3:<br />

904–912.<br />

71. MacDonald TT, Choy MY, Spencer J et al.<br />

Histopathology and immunohistochemistry of the<br />

caecum in children with the Trichuris dysentery<br />

syndrome. J Clin Pathol 1991; 44: 194–199.<br />

72. Cooper ES, Whyte Alleng CA, Finzi Smith JS et al.<br />

Intestinal nematode infections in children: the<br />

pathophysiological price paid. Parasitology 1992; 104<br />

(Suppl): S91–103.<br />

73. Cooper ES, Ramdath DD, Whyte Alleng C et al. Plasma<br />

proteins in children with Trichuris dysentery syndrome.<br />

J Clin Pathol 1997; 50: 236–240.<br />

74. Duff EM, Anderson NM, Cooper ES. Plasma insulin-like<br />

growth factor-1, type 1 procollagen, and serum tumor<br />

necrosis factor alpha in children recovering from<br />

Trichuris dysentery syndrome. Pediatrics 1999; 103: e69.<br />

75. MacDonald TT, Spencer J, Murch SH et al.<br />

Immunoepidemiology of intestinal helminthic<br />

infections. 3. Mucosal macrophages and cytokine<br />

production in the colon of children with Trichuris<br />

trichiura dysentery. Trans R Soc Trop Med Hyg 1994; 88:<br />

265–268.<br />

76. Cooper ES, Spencer J, Whyte-Alleng CA et al.<br />

Immediate hypersensitivity in colon of children with<br />

chronic Trichuris trichiura dysentery. Lancet 1991; 338:<br />

1104–1107.<br />

77. Raj SM. Fecal occult blood testing on Trichuris-infected<br />

primary school children in northeastern peninsular<br />

Malaysia. Am J Trop Med Hyg 1999; 60: 165–166.<br />

78. Kaur G, Raj SM, Naing NN. Trichuriasis: localized<br />

inflammatory responses in the colon. Southeast Asian J<br />

Trop Med Public Health 2002; 33: 224–228.<br />

79. Kamath KR. Severe infection with Trichuris trichiura in<br />

Malaysian children. A clinical study of 30 cases treated<br />

with stilbazium iodine. Am J Trop Med Hyg 1973; 22:<br />

600–605.<br />

80. Mathan VI,.Baker SJ. Whipworm disease. Intestinal<br />

structure and function of patients with severe Trichuris<br />

trichuria infestation. Am J Dig Dis 1970; 15: 913–918.<br />

81. Gilman RH, Davis C, Fitzgerald F. Heavy Trichuris<br />

infection and amoebic dysentery in Orang Asli children.<br />

A comparison of the two diseases. Trans R Soc Trop Med<br />

Hyg 1976; 70: 313–316.<br />

82. Noorizan AM, Mahendra RS. Trichuris dysentery<br />

syndrome: evidence that it may be underdiagnosed in<br />

Kelantan. Med J Malaysia 2001; 56: 53–57.


83. Phillips AD, Thomas AG, Walker-Smith JA.<br />

Cryptosporidium, chronic diarrhea and the proximal<br />

small intestinal mucosa. Gut 1992; 33: 1057–1061.<br />

84. Bern C, Ortega Y, Checkley W et al. Epidemiologic<br />

differences between cyclosporiasis and<br />

cryptosporidiosis in Peruvian children. Emerg Infect Dis<br />

2002; 8: 581–585.<br />

85. Nchito M, Kelly P, Sianongo S et al. Cryptosporidiosis in<br />

urban Zambian children: an analysis of risk factors. Am<br />

J Trop Med Hyg 1998; 59: 435–437.<br />

86. Molbak K, Hojlyng N, Ingholt L et al. An epidemic<br />

outbreak of cryptosporidiosis: a prospective community<br />

study from Guinea Bissau. Pediatr Infect Dis J 1990; 9:<br />

566–570.<br />

87. Molbak K, Aaby P, Hojilyng N, Da Silva APJ. Risk<br />

factors for Cryptosporidium diarrhea in early childhood:<br />

a case–control study from Guinea-Bissau, West Africa.<br />

Am J Epidemiol 1994; 139: 734–740.<br />

88. Molbak K, Hojlyng N, Gottschau A et al.<br />

Cryptosporidiosis in infancy and childhood mortality in<br />

Guinea Bissau, west Africa. Br Med J 1993; 307: 417–420.<br />

89. Molbak K, Lisse IM, Hojlyng N, Aaby P. Severe<br />

cryptosporidiosis in children with normal T-cell<br />

subsets. Parasite Immunol 1994; 16: 275–277.<br />

90. Iqbal J, Hira PR, Al Ali F, Philip R. Cryptosporidiosis in<br />

Kuwaiti children: seasonality and endemicity. Clin<br />

Microbiol Infect 2001; 7: 261–266.<br />

91. Adegbola RA, Demba E, De Veer G, Todd J.<br />

Cryptosporidium infection in Gambian children less<br />

than 5 years of age. J Trop Med Hyg 1994; 97: 103–107.<br />

92. Javier Enriquez F, Avila CR, Ignacio Santos J et al.<br />

Cryptosporidium infections in Mexican children:<br />

clinical, nutritional, enteropathogenic, and diagnostic<br />

evaluations. Am J Trop Med Hyg 1997; 56: 254–257.<br />

93. Agnew DG, Lima AA, Newman RD et al.<br />

Cryptosporidiosis in northeastern Brazilian children:<br />

association with increased diarrhea morbidity. J Infect<br />

Dis 1998; 177: 754–760.<br />

94. McDonald AC, MacKenzie WR, Addiss DG et al.<br />

Cryptosporidium parvum-specific antibody responses<br />

among children residing in Milwaukee during the 1993<br />

waterborne outbreak. J Infect Dis 2001; 183: 1373–1379.<br />

95. Cicirello HG, Kehl KS, Addiss DG et al.<br />

Cryptosporidiosis in children during a massive<br />

waterborne outbreak in Milwaukee, Wisconsin: clinical,<br />

laboratory and epidemiologic findings. Epidemiol Infect<br />

1997; 119: 53–60.<br />

96. Cordell RL, Thor PM, Addiss DG et al. Impact of a<br />

massive waterborne cryptosporidiosis outbreak on child<br />

care facilities in metropolitan Milwaukee, Wisconsin.<br />

Pediatr Infect Dis J 1997; 16: 639–644.<br />

97. Moore R, Tzipori S, Griffiths JK et al. Temporal changes<br />

in permeability and structure of piglet ileum after sitespecific<br />

infection by Cryptosporidium parvum.<br />

Gastroenterology 1995; 108: 1030–1039.<br />

98. Capet C, Kapel N, Huneau JF et al. Cryptosporidium<br />

parvum infection in suckling rats: impairment of<br />

mucosal permeability and Na(+)-glucose cotransport.<br />

Exp Parasitol 1999; 91: 119–125.<br />

99. Wyatt CR, Brackett EJ, Savidge J. Evidence for the<br />

emergence of a type-1-like immune response in<br />

intestinal mucosa of calves recovering from<br />

cryptosporidiosis. J Parasitol 2001; 87: 90–95.<br />

100. Pollok RC, Farthing MJ, Bajaj-Elliott M et al. Interferon<br />

gamma induces enterocyte resistance against infection<br />

by the intracellular pathogen Cryptosporidium parvum.<br />

Gastroenterology 2001; 120: 99–107.<br />

101. Hayward AR, Chmura K, Cosyns M. Interferon-gamma<br />

is required for innate immunity to Cryptosporidium<br />

parvum in mice. J Infect Dis 2000; 182: 1001–1004.<br />

References 189<br />

102. Lawrence CE, Paterson JC, Higgins LM et al. IL-4regulated<br />

enteropathy in an intestinal nematode<br />

infection. Eur J Immunol 1998; 28: 2672–2684.<br />

103. Garside P, Kennedy MW, Wakelin D, Lawrence CE.<br />

Immunopathology of intestinal helminth infection.<br />

Parasite Immunol 2000; 22: 605–612.<br />

104. Leitch GJ, He Q. Reactive nitrogen and oxygen species<br />

ameliorate experimental cryptosporidiosis in the<br />

neonatal BALB/c mouse model. Infect Immun 1999; 67:<br />

5885–5891.<br />

105. Leitch GJ, He Q. Arginine-derived nitric oxide reduces<br />

fecal oocyst shedding in nude mice infected with<br />

Cryptosporidium parvum. Infect Immun 1994; 62:<br />

5173–5176.<br />

106. Goodgame RW, Kimball K, Ou CN et al. Intestinal<br />

function and injury in acquired immunodeficiency<br />

syndrome-related cryptosporidiosis. Gastroenterology<br />

1995; 108: 1075–1082.<br />

107. Checkley W, Epstein LD, Gilman RH et al. Effects of<br />

Cryptosporidium parvum infection in Peruvian children:<br />

growth faltering and subsequent catch-up growth. Am J<br />

Epidemiol 1998; 148: 497–506.<br />

108. Molbak K, Andersen M, Aaby P et al. Cryptosporidium<br />

infection in infancy as a cause of malnutrition: a<br />

community study from Guinea-Bissau, West Africa. Am<br />

J Clin Nutr 1997; 65: 149–152.<br />

109. Guarino A, Castaldo A, Russo S et al. Enteric cryptosporidiosis<br />

in pediatric HIV infection. J Pediatr<br />

Gastroenterol Nutr 1997; 25: 182–187.<br />

110. Sarabia Arce S, Salazar Lindo E, Gilman RH et al.<br />

Case–control study of Cryptosporidium parvum infection<br />

in Peruvian children hospitalized for diarrhea:<br />

possible association with malnutrition and nosocomial<br />

infection. Pediatr Infect Dis J 1990; 9: 627–631.<br />

111. Checkley W, Gilman RH, Epstein LD et al.<br />

Asymptomatic and symptomatic cryptosporidiosis: their<br />

acute effect on weight gain in Peruvian children. Am J<br />

Epidemiol 1997; 145: 156–163.<br />

112. Xiao L, Bern C, Limor J et al. Identification of 5 types of<br />

Cryptosporidium parasites in children in Lima, Peru. J<br />

Infect Dis 2001; 183: 492–497.<br />

113. Genta RM. Global prevalence of strongyloidiasis: critical<br />

review with epidemiologic insights into the prevention<br />

of disseminated disease. Rev Infect Dis 1989; 11:<br />

755–767.<br />

114. Egido JM, De Diego JA, Penin P. The prevalence of<br />

enteropathy due to strongyloidiasis in Puerto<br />

Maldonado (Peruvian Amazon). Braz J Infect Dis 2001;<br />

5: 119–123.<br />

115. Clyne M, Labigne A, Drumm B. <strong>Helicobacter</strong> <strong>pylori</strong><br />

requires an acidic environment to survive in the<br />

presence of urea. Infect Immun 1995; 63: 1669–1673.<br />

116. Quiros A, Quiros E, Gonzalez I et al. <strong>Helicobacter</strong> <strong>pylori</strong><br />

seroepidemiology in risk groups. Eur J Epidemiol 1994;<br />

10: 299–301.<br />

117. Barnish G, Ashford RW. Strongyloides cf. fuelleborni<br />

and hookworm in Papua New Guinea: patterns of<br />

infection within the community. Trans R Soc Trop Med<br />

Hyg 1989; 83: 684–688.<br />

118. Milner PF, Irvine RA, Barton CJ et al. Intestinal<br />

malabsorption in Strongyloides stercoralis infestation.<br />

Gut 1965; 6: 574–581.<br />

119. Sipahi AM, Damiao AO, Simionato CS et al. Small<br />

bowel bacterial overgrowth in strongyloidiasis.<br />

Digestion 1991; 49: 120–124.<br />

120. Werneck-Silva AL, Sipahi AM, Damiao AO et al.<br />

Intestinal permeability in strongyloidiasis. Braz J Med<br />

Biol Res 2001; 34: 353–357.<br />

121. Sullivan PB, Lunn PG, Northrop Clewes CA, Farthing<br />

MJ. Parasitic infection of the gut and protein-losing


190<br />

Intestinal parasites<br />

enteropathy. J Pediatr Gastroenterol Nutr 1992; 15:<br />

404–407.<br />

122. Burke JA. Strongyloidiasis in childhood. Am J Dis Child<br />

1978; 132: 1130–1136.<br />

123. Atkins NS, Conway DJ, Lindo JF et al. L3 antigenspecific<br />

antibody isotype responses in human<br />

strongyloidiasis: correlations with larval output. Parasite<br />

Immunol 1999; 21: 517–526.<br />

124. Walker AC, Blake G, Downing D. A syndrome of partial<br />

intestinal obstruction due to Strongyloides stercoralis.<br />

Med J Aust 1976; 1: 47–48.<br />

125. Walker-Smith JA, McMillan B, Middleton AW et al.<br />

Strongyloidiasis causing small-bowel obstruction in an<br />

Aboriginal infant. Med J Aust 1969; 2: 1263–1265.<br />

126. Vince JD, Ashford RW, Gratten MJ, Bana-Koiri J.<br />

Strongyloides species infestation in young infants of<br />

Papua, New Guinea: association with generalized<br />

oedema. P N G Med J 1979; 22: 120–127.<br />

127. Barnish G, Barker J. An intervention study using<br />

thiabendazole suspension against Strongyloides<br />

fuelleborni-like infections in Papua New Guinea. Trans R<br />

Soc Trop Med Hyg 1987; 81: 60–63.<br />

128. Robinson RD, Lindo JF, Neva FA et al.<br />

Immunoepidemiologic studies of Strongyloides stercoralis<br />

and human T lymphotropic virus type I infections<br />

in Jamaica. J Infect Dis 1994; 169: 692–696.<br />

129. Siddiqui AA, Berk SL. Diagnosis of Strongyloides<br />

stercoralis infection. Clin Infect Dis 2001; 33:<br />

1040–1047.<br />

130. Sato Y, Kobayashi J, Shiroma Y. Serodiagnosis of<br />

strongyloidiasis. The application and significance. Rev<br />

Inst Med Trop Sao Paulo 1995; 37: 35–41.<br />

131. Gyorkos TW, Genta RM, Viens P, MacLean JD.<br />

Seroepidemiology of Strongyloides infection in the<br />

Southeast Asian refugee population in Canada. Am J<br />

Epidemiol 1990; 132: 257–264.<br />

132. Haque R, Faruque AS, Hahn P et al. Entamoeba<br />

histolytica and Entamoeba dispar infection in children<br />

in Bangladesh. J Infect Dis 1997; 175: 734–736.<br />

133. Heckendorn F, N’Goran EK, Felger I et al. Speciesspecific<br />

field testing of Entamoeba spp. On an area of<br />

high endemicy. Trans R Soc Trop Med Hyg 2002; 96:<br />

521–528.<br />

134. Braga LL, Gomes ML, Da Silva MW et al. Household<br />

epidemiology of Entamoeba histolytica infection in an<br />

urban community in northeastern Brazil. Am J Trop Med<br />

Hyg 2001; 65: 268–271.<br />

135. Stanley SL. Pathophysiology of amoebiasis. Trends<br />

Parasitol 2001; 17: 280–285.<br />

136. Haque R, Duggal P, Ali IM et al. Innate and acquired<br />

resistance to amebiasis in Bangladeshi children. J Infect<br />

Dis 2002; 186: 547–552.<br />

137. Haque R, Ali IM, Petri WA Jr. Prevalence and immune<br />

response to Entamoeba histolytica infection in preschool<br />

children in Bangladesh. Am J Trop Med Hyg 1999; 60:<br />

1031–1034.<br />

138. Ayeh-Kumi PF, Ali IM, Lockhart LA et al. Entamoeba<br />

histolytica: genetic diversity of clinical isolates from<br />

Bangladesh as demonstrated by polymorphisms in the<br />

serine-rich gene. Exp Parasitol 2001; 99: 80–88.<br />

139. Sirivichayakul C, Radomyos P, Praevanit R et al.<br />

Hymenolepis nana infection in Thai children. J Med<br />

Assoc Thai 2000; 83: 1035–1038.<br />

140. Liu LX, Chi J, Upton MP, Ash LR. Eosinophilic colitis<br />

associated with larvae of the pinworm Enterobius<br />

vermicularis. Lancet 1995; 346: 410–412.<br />

141. Sinniah B, Leopairut J, Neafie RC et al. Enterobiasis: a<br />

histopathological study of 259 patients. Ann Trop Med<br />

Parasitol 1991; 85: 625–635.<br />

142. Hoge CW, Shlim DR, Rajah R et al. Epidemiology of<br />

diarrheal illness associated with coccidian-like<br />

organism among travellers and foreign residents in<br />

Nepal. Lancet 1993; 341: 1175–1179.<br />

143. Ho AY, Lopez AS, Eberhart MG et al. Outbreak of<br />

cyclosporiasis associated with imported raspberries,<br />

Philadelphia, Pennsylvania, 2000. Emerg Infect Dis<br />

2002; 8: 783–788.<br />

144. Katz D, Kumar S, Malecki J et al. Cyclosporiasis<br />

associated with imported raspberries, Florida, 1996.<br />

Public Health Rep 1999; 114: 427–438.<br />

145. Quintero-Betancourt W, Peele ER, Rose JB.<br />

Cryptosporidium parvum and Cyclospora cayetanensis: a<br />

review of laboratory methods for detection of these<br />

waterborne parasites. J Microbiol Methods 2002; 49:<br />

209–224.<br />

146. Bern C, Hernandez B, Lopez MB et al. The contrasting<br />

epidemiology of Cyclospora and Cryptosporidium among<br />

outpatients in Guatemala. Am J Trop Med Hyg 2000; 63:<br />

231–235.<br />

147. Eberhard ML, Nace EK, Freeman AR et al. Cyclospora<br />

cayetanensis infections in Haiti: a common occurrence<br />

in the absence of watery diarrhea. Am J Trop Med Hyg<br />

1999; 60: 584–586.<br />

148. Chacin-Bonilla L, Estevez J, Monsalve F, Quijada L.<br />

Cyclospora cayetanensis infections among diarrheal<br />

patients from Venezuela. Am J Trop Med Hyg 2001; 65:<br />

351–354.<br />

149. Pape JW, Verdier RI, Boncy M et al. Cyclospora infection<br />

in adults infected with HIV. Clinical manifestations,<br />

treatment, and prophylaxis. Ann Intern Med 1994; 121:<br />

654–657.<br />

150. Connor BA, Shlim DR, Scholes JV et al. Pathologic<br />

changes in the small bowel in nine patients with<br />

diarrhea associated with a coccidia-like body. Ann<br />

Intern Med 1993; 119: 377–382.<br />

151. Rigo CR,.Franco RM. [Comparison between the modified<br />

Ziehl–Neelsen and acid-fast-trichrome methods for fecal<br />

screening of Cryptosporidium parvum and Isospora<br />

belli]. Rev Soc Bras Med Trop 2002; 35: 209–214.<br />

152. Sherchand JB,.Cross JH. Emerging pathogen Cyclospora<br />

cayetanensis infection in Nepal. Southeast Asian J Trop<br />

Med Public Health 2001; 32 (Suppl 2): 143–150.<br />

153. Rizk H, Soliman M. Coccidiosis among malnourished<br />

children in Mansoura, Dakahlia Governorate, Egypt. J<br />

Egypt Soc Parasitol 2001; 31: 877–886.<br />

154. Madico G, McDonald J, Gilman RH et al. Epidemiology<br />

and treatment of Cyclospora cayetanensis infection in<br />

Peruvian children. Clin Infect Dis 1997; 24: 977–981.<br />

155. Hoge CW, Shlim DR, Ghimire M et al. Placebocontrolled<br />

trial of co-trimoxazole for Cyclospora<br />

infections among travellers and foreign residents in<br />

Nepal. Lancet 1995; 345: 691–693.<br />

156. Horton J. Albendazole: a review of anthelmintic efficacy<br />

and safety in humans. Parasitology 2000; 121 (Suppl):<br />

S113–S132.<br />

157. Forrester JE, Bailar JC III, Esrey SA et al. Randomised<br />

trial of albendazole and pyrantel in symptomless<br />

trichuriasis in children. Lancet 1998; 352: 1103–1108.<br />

158. Montresor A, Stoltzfus RJ, Albonico M et al. Is the<br />

exclusion of children under 24 months from<br />

anthelmintic treatment justifiable? Trans R Soc Trop<br />

Med Hyg 2002; 96: 197–199.<br />

159. Zulu I, Veitch A, Sianongo S et al. Albendazole<br />

chemotherapy for AIDS-related diarrhea in Zambia –<br />

clinical, parasitological and mucosal responses. Aliment<br />

Pharmacol Ther 2002; 16: 595–601.<br />

160. Belizario VY, Amarillo ME, de Leon WU et al. A<br />

comparison of the efficacy of single doses of


albendazole, ivermectin, and diethylcarbamzine alone<br />

or in combinations against Ascaris and Trichuris spp.<br />

Bull World Health Organ 2003; 81: 35–42.<br />

161. Hall A, Nahar Q. Albendazole as a treatment for<br />

infections with Giardia duodenalis in children in<br />

Bangladesh. Trans R Soc Trop Med Hyg 1993; 87: 84–86.<br />

162. Ermis B, Aslan T, Beder L, Unalacak M. A randomized<br />

placebo-controlled trial of mebendazole for halitosis.<br />

Arch Pediatr Adolesc Med 2002; 156: 995–998.<br />

163. Marti H, Haji HJ, Savioli L et al. A comparative trial of a<br />

single-dose ivermectin versus three days of albendazole<br />

for treatment of Strongyloides stercoralis and other soiltransmitted<br />

helminth infections in children. Am J Trop<br />

Med Hyg 1996; 55: 477–481.<br />

164. Behnke JM, Pritchard DI, Wakelin D et al. Effect of<br />

ivermectin on infection with gastro-intestinal<br />

nematodes in Sierra Leone. J Helminthol 1994; 68:<br />

187–195.<br />

165. Datry A, Hilmarsdottir I, Mayorga Sagastume R et al.<br />

Treatment of Strongyloides stercoralis infection with<br />

ivermectin compared with albendazole: results of an<br />

open study of 60 cases. Trans R Soc Trop Med Hyg 1994;<br />

88: 344–345.<br />

166. Gann PH, Neva FA, Gam AA. A randomized trial of<br />

single- and two-dose ivermectin versus thiabendazole<br />

for treatment of strongyloidiasis. J Infect Dis 1994; 169:<br />

1076–1079.<br />

167. Misra PK, Kumar A, Agarwal V, Jagota SC. A comparative<br />

clinical trial of albendazole versus metronidazole in<br />

children with giardiasis. Indian Pediatr 1995; 32:<br />

779–782.<br />

168. Lares Asseff I, Cravioto J, Santiago P, Perez Ortiz B. A<br />

new dosing regimen for metronidazole in malnourished<br />

children. Scand J Infect Dis 1993; 25: 115–121.<br />

169. Addiss DG, Juranek DD, Spencer HC. Treatment of<br />

children with asymptomatic and nondiarrheal Giardia<br />

infection. Pediatr Infect Dis J 1991; 10: 843–846.<br />

170. Ish Horowicz M, Korman SH, Shapiro M et al.<br />

Asymptomatic giardiasis in children. Pediatr Infect Dis J<br />

1989; 8: 773–779.<br />

171. Pickering LK, Engelkirk PG. Giardia lamblia. Pediatr<br />

Clin North Am 1988; 35: 565–577.<br />

172. Zaat JO, Mank T, Assendelft WJ. Drugs for treating<br />

giardiasis. Cochrane Database Syst Rev 2000;<br />

CD000217.<br />

173. Gilles HM, Hoffman PS. Treatment of intestinal parasitic<br />

infections: a review of nitazoxanide. Trends<br />

Parasitol 2002; 18: 95–97.<br />

174. Rossignol JF, Ayoub A, Ayers MS. Treatment of diarrhea<br />

caused by Giardia intestinalis and Entamoeba histolytica<br />

or E. dispar: a randomized, double-blind, placebocontrolled<br />

study of nitazoxanide. J Infect Dis 2001; 184:<br />

381–384.<br />

175. Rossignol JF, Ayoub A, Ayers MS. Treatment of diarrhea<br />

caused by Cryptosporidium parvum: a prospective<br />

randomized, double-blind, placebo-controlled study of<br />

Nitazoxanide. J Infect Dis 2001; 184: 103–106.<br />

176. Davila-Gutierrez CE, Vasquez C, Trujillo-Hernandez B,<br />

Huerta M. Nitazoxanide compared with quinfamide and<br />

mebendazole in the treatment of helminthic infections<br />

and intestinal protozoa in children. Am J Trop Med Hyg<br />

2002; 66: 251–254.<br />

177. Cedillo-Rivera R, Chavez B, Gonzalez-Robles A et al. In<br />

vitro effect of nitazoxanide against Entamoeba<br />

histolytica, Giardia intestinalis and Trichomonas<br />

vaginalis trophozoites. J Eukaryot Microbiol 2002; 49:<br />

201–208.<br />

178. Ortiz JJ, Chegne NL, Gargala G, Favennec L.<br />

Comparative clinical studies of nitazoxanide,<br />

albendazole and praziquantel in the treatment of<br />

References 191<br />

ascariasis, trichuriasis and hymenolepiasis in children<br />

from Peru. Trans R Soc Trop Med Hyg 2002; 96: 193–196.<br />

179. Ortiz JJ, Ayoub A, Gargala G et al. Randomized clinical<br />

study of nitazoxanide compared to metronidazole in the<br />

treatment of symptomatic giardiasis in children from<br />

Northern Peru. Aliment Pharmacol Ther 2001; 15:<br />

1409–1415.<br />

180. Amadi B, Mwiya M, Musuku J et al. Effect of nitazoxanide<br />

on morbidity and mortality in Zambian children<br />

with cryptosporidiosis: a randomised controlled trial.<br />

Lancet 2002; 360: 1375–1380.<br />

181. Bissuel F, Cotte L, Rabodonirina M et al. Paromomycin:<br />

an effective treatment for cryptosporidial diarrhea in<br />

patients with AIDS. Clin Infect Dis 1994; 18: 447–449.<br />

182. Hewitt RG, Yiannoutsos CT, Higgs ES et al.<br />

Paromomycin: no more effective than placebo for<br />

treatment of cryptosporidiosis in patients with<br />

advanced human immunodeficiency virus infection.<br />

AIDS Clinical Trial Group. Clin Infect Dis 2000; 31:<br />

1084–1092.<br />

183. Anonymous. World Development Report 1993: Investing<br />

in Health. New York: Oxford University Press, 1993.<br />

184. World Health Organization. Guidelines for the<br />

Evaluation of Soil-transmitted Helminthiasis and<br />

Schistosomiasis at Community Level: a Guide for<br />

Managers of Control Programmes. Geneva: WHO, 1998.<br />

185. Brooker S, Marriot H, Hall A et al. Community<br />

perception of school-based delivery of anthelmintics in<br />

Ghana and Tanzania. Trop Med Int Health 2001; 6:<br />

1075–1083.<br />

186. Allen HE, Crompton DW, de Silva N et al. New policies<br />

for using antihelmintics in high risk groups. Trends<br />

Parasitol 2002; 18: 381–382.<br />

187. Dickson R, Awasthi S, Williamson P et al. Effects of<br />

treatment for intestinal helminth infection on growth<br />

and cognitive performance in children: systematic<br />

review of randomised trials. Br Med J 2000; 320:<br />

1697–1701.<br />

188. Dickson R, Awasthi S, Demellweek C, Williamson P.<br />

Anthelmintic drugs for treating worms in children:<br />

effects on growth and cognitive performance. Cochrane<br />

Database Syst Rev 2000; CD000371.<br />

189. Stoltzfus RJ, Kvalsvig JD, Chwaya HM et al. Effects of<br />

iron supplementation and anthelmintic treatment on<br />

motor and language development of preschool children<br />

in Zanzibar: double blind, placebo controlled study. Br<br />

Med J 2001; 323: 1389–1393.<br />

190. Olds GR, King C, Hewlett J et al. Double-blind placebocontrolled<br />

study of concurrent administration of<br />

albendazole and praziquantel in schoolchildren with<br />

schistosomiasis and geoheminths. J Infect Dis 1999; 179:<br />

996–1003.<br />

191. Northrop-Clewes CA, Rousham EK, Mascie-Taylor CN,<br />

Lunn PG. Anthelmintic treatment of rural Bangladeshi<br />

children: effect on host physiology, growth, and<br />

biochemical status. Am J Clin Nutr 2001; 73: 53–60.<br />

192. Rousham EK. An increase in Giardia duodenalis<br />

infection among children receiving periodic<br />

anthelmintic treatment in Bangladesh. J Trop Pediatr<br />

1994; 40: 329–333.<br />

193. Stettler N, Schutz Y, Jequier E. Effect of low-level<br />

pathogenic helminth infection on energy metabolism in<br />

Gambian children. Am J Trop Med Hyg 1998; 58:<br />

476–479.<br />

194. Raj SM, Sein KT, Anuar AK, Mustaffa BE. Effect of<br />

intestinal helminthiasis on school attendance by early<br />

primary schoolchildren. Trans R Soc Trop Med Hyg<br />

1997; 91: 131–132.<br />

195. Moore SR, Lima AA, Conaway MR et al. Early<br />

childhood diarrhea and helminthiases associated with


192<br />

Intestinal parasites<br />

long-term linear growth faltering. Int J Epidemiol 2001;<br />

30: 1457–1464.<br />

196. Guyatt HL. Do intestinal nematodes affect productivity<br />

in adults? Parasitol Today 2000; 16: 153–158.<br />

197. Donnen P, Brasseur D, Dramaix M et al. Vitamin A<br />

supplementation but not deworming improves growth<br />

of malnourished preschool children in eastern Zaire. J<br />

Nutr 1998; 128: 1320–1327.<br />

198. Taylor M, Jinabhai CC, Couper I et al. The effect of<br />

different anthelmintic treatment regimens combined<br />

with iron supplementation on the nutritional status of<br />

schoolchildren in KwaZulu-Natal, South Africa: a<br />

randomized controlled trial. Trans R Soc Trop Med Hyg<br />

2001; 95: 211–216.<br />

199. Muchiri EM, Thiong’o FW, Magnussen P, Ouma JH. A<br />

comparative study of different albendazole and<br />

mebendazole regimens for the treatment of intestinal<br />

infections in school children of Usigu Division, Western<br />

Kenya. J Parasitol 2001; 87: 413–418.<br />

200. Albonico M, Renganathan E, Bosman A et al. Efficacy of<br />

a single dose of mebendazole on prevalence and<br />

intensity of soil-transmitted nematodes in Zanzibar.<br />

Trop Geogr Med 1994; 46: 142–146.<br />

201. Lunn PG, Erinoso HO, Northrop-Clewes CA, Boyce SA.<br />

Giardia intestinalis is unlikely to be a major cause of the<br />

poor growth of rural Gambian infants. J Nutr 1999; 129:<br />

872–877.<br />

202. Sullivan PB, Marsh MN, Phillips MB et al. Prevalence<br />

and treatment of giardiasis in chronic diarrhea and<br />

malnutrition. Arch Dis Child 1991; 66: 304–306.<br />

203. Sullivan PS, DuPont HL, Arafat RR et al. Illness and<br />

reservoirs associated with Giardia lamblia infection in<br />

rural Egypt: the case against treatment in developing<br />

world environments of high endemicity. Am J Epidemiol<br />

1988; 127: 1272–1281.<br />

204. Gilman RH, Marquis GS, Miranda E et al. Rapid<br />

reinfection by Giardia lamblia after treatment in a<br />

hyperendemic Third World community. Lancet 1988; 1:<br />

343–345.<br />

205. Rousham EK. Perceptions and treatment of intestinal<br />

worms in rural Bangladesh: local differences in<br />

knowledge and behaviour. Soc Sci Med 1994; 39:<br />

1063–1068.<br />

206. Muller M, Sanchez RM, Suswillo RR. Evaluation of a<br />

sanitation programme using eggs of Ascaris lumbricoides<br />

in household yard soils as indicators. J Trop Med Hyg<br />

1989; 92: 10–16.<br />

207. Albert MJ, Faruque AS, Faruque SM et al. Case–control<br />

study of enteropathogens associated with childhood<br />

diarrhea in Dhaka, Bangladesh. J Clin Microbiol 1999;<br />

37: 3458–3464.<br />

208. Huilan S, Zhen LG, Mathan MM et al. Etiology of acute<br />

diarrhea among children in developing countries: a<br />

multicentre study in five countries. Bull World Health<br />

Organ 1991; 69: 549–555.<br />

209. Loening WE, Coovadia YM, van den Ende J. Aetiological<br />

factors of infantile diarrhea: a community-based study.<br />

Ann Trop Paediatr 1989; 9: 248–255.


12<br />

Post-infectious persistent<br />

diarrhea in developing<br />

countries<br />

Zulfiqar A Bhutta<br />

Global significance of diarrheal<br />

disorders and persistent diarrhea<br />

Despite considerable advances in the understanding<br />

and management of diarrheal disorders in<br />

childhood, they are still responsible for a major<br />

burden of childhood deaths globally, with an estimated<br />

2.5 million deaths. 1 In an estimate of the<br />

global burden of diarrheal disorders in 1980 the<br />

World Health Organization (WHO) calculated that<br />

there were over 700 million episodes of diarrhea<br />

annually in children under 5 years of age in developing<br />

countries (excluding China), with approximately<br />

4.6 million deaths. 2 Reviews in the early<br />

1990s suggested that diarrheal disorders still<br />

accounted for almost a third of all admissions to<br />

health facilities in developing countries, 3 with an<br />

estimated incidence of diarrheal disorders at<br />

around 2.6 episodes/child per year. More recent<br />

reviews of studies published in the past 10 years<br />

indicate that, while global mortality may have<br />

reduced, the incidence remained unchanged at<br />

about 3.2 episodes/child year. 4,5 These findings<br />

indicate the continuing need to focus on prevention<br />

and management of childhood diarrhea in<br />

developing countries.<br />

It is recognized that most diarrheal disorders form<br />

a continuum, with the majority of cases resolving<br />

within the first week of the illness. However, a<br />

smaller proportion of diarrheal illnesses fail to<br />

resolve and may persist for longer than 2 weeks.<br />

Persistent diarrhea has been defined as an episode<br />

that begins acutely but lasts for 14 days or longer.<br />

It has been shown to identify children with a<br />

substantially increased diarrheal burden and leads<br />

to the majority of all diarrhea-related deaths. 6 In a<br />

prospective study of diarrheal disorders in north<br />

India, persistent diarrhea accounted for only 5% of<br />

all diarrheal episodes, but the case/fatality rate for<br />

persistent diarrhea was 14% in comparison with<br />

0.7% for episodes of shorter duration. 7 In a similar<br />

prospective study of diarrheal episodes in the<br />

community in and around Lahore (Pakistan),<br />

persistent diarrhea accounted for 8–18% of all<br />

diarrheal episodes but 54% of all diarrheal<br />

deaths. 8 Figure 12.1 indicates the relative proportion<br />

of diarrheal episodes from several prospective<br />

community-based studies in developing countries.<br />

7,9–11<br />

It is important to emphasize that the bulk of the<br />

epidemiological information on the relationship of<br />

acute diarrheal disorders to persistent diarrhea is<br />

derived from studies undertaken over 10–15 years<br />

ago, and that there is a paucity of recent data on<br />

this subject, especially from areas non-endemic for<br />

HIV. 5 However, it is evident from studies in HIVendemic<br />

areas that chronic enteropathy and<br />

persistent diarrhea is a common manifestation of<br />

advancing HIV infection and AIDS.<br />

Pathogenesis of persistent diarrhea<br />

Although a close relationship between diarrheal<br />

disease and malnutrition has long been recognized,<br />

12 this has been challenged. 13 The relationship<br />

of persistent diarrhea to malnutrition is less<br />

controversial, as the disorder is commonly seen in<br />

association with significant malnutrition. In a<br />

verbal autopsy study of diarrheal deaths in<br />

Bangladesh, Fauveau et al 14 found that almost half<br />

the deaths were in malnourished children with<br />

persistent diarrhea, and the relative risk of dying<br />

with persistent diarrhea and severe malnutrition<br />

was 17-fold higher than in children with lower<br />

degrees of malnutrition.<br />

There are several reasons for malnutrition both to<br />

predispose to persistent diarrhea and to follow it.<br />

193


194<br />

Post-infectious persistent diarrhea in developing countries<br />

80<br />

60<br />

40<br />

20<br />

% of episodes 100<br />

0<br />

Figure 12.1 Distribution of diarrheal episodes in community-based studies. From references 7 and 9–11.<br />

These range from achlorhydria with increased risk<br />

of small-bowel contamination, systemic immune<br />

deficiency, intestinal and pancreatic enzyme deficiency<br />

and altered intestinal mucosal repair mechanisms<br />

following an infectious insult. An independent<br />

relationship has also been demonstrated<br />

between cutaneous anergy and the subsequent risk<br />

of development of persistent diarrhea. 15 There has<br />

been much interest in the possibility that such<br />

transient immune deficiency may also be a marker<br />

of concomitant micronutrient deficiency. 16–18 The<br />

most striking example of the critical role that the<br />

immune system plays in the pathogenesis of<br />

persistent diarrhea is the relationship between<br />

HIV/AIDS and persistent diarrhea. This is exemplified<br />

by the host of studies linking persistent<br />

diarrhea with cryptosporidiosis 19 and other parasitic<br />

infections 20,21 in Africa and Asia.<br />

A clear understanding of alterations in intestinal<br />

morphology and physiology is crucial towards the<br />

development of interventional strategies, but there<br />

has been little progress in our understanding of<br />

this problem in developing countries. This has<br />

been largely due to a paucity of studies formally<br />

evaluating intestinal biopsy findings in representative<br />

populations. A wide variety of pathological<br />

changes have been described after persistent diarrhea,<br />

ranging from near-normal appearance to<br />

mucosal flattening, crypt hypertrophy and<br />

lymphocytic infiltration of the mucosa. 22 Recent<br />

electron microscopic studies of the intestinal<br />

1–7 days 8–14 days 15–21 days > 22 days<br />

mucosa in persistent diarrhea have revealed<br />

patchy villous atrophy and intraepithelial lymphocytic<br />

infiltration 23 as well as severe mucosal<br />

damage and villous atrophy. 24<br />

Poor intestinal repair is regarded as a key component<br />

of the abnormal mucosal morphology.<br />

However, the exact factors underlying this ineffective<br />

repair process and continuing injury are<br />

poorly understood (Figure 12.2). The end result of<br />

this mucosal derangement is poor absorption of<br />

luminal nutrients, as well as increased permeability<br />

of the bowel to abnormal dietary or microbial<br />

antigens. 25–27 Alterations of intestinal permeability<br />

in early childhood may reflect changes in intestinal<br />

mucosal maturation 28 and may be affected by<br />

concomitant enteric infections. 29<br />

Risk factors for persistent diarrhea<br />

It is important to recognize the major risk factors<br />

for development of persistent diarrhea, as appropriate<br />

case management of acute diarrhea is key to<br />

the prevention of prolonged episodes.<br />

Specific pathogens<br />

Peru (1989)<br />

Bangladesh (1982)<br />

India (1989)<br />

Bangladesh (1992)<br />

Pakistan (1993)<br />

The association of specific bacterial and viral<br />

infections with persistent diarrhea has been the<br />

subject of considerable debate. 30,31 Evidence from


Figure 12.2 Mechanisms and effects of enteropathy of malnutrition and prolonged diarrhea.<br />

Bangladesh suggests that recurrent bouts of infection<br />

with pathogens such as Shigella lead to<br />

prolongation of the duration of successive diarrheal<br />

episodes. Although several studies have<br />

identified an association between persistent diarrhea<br />

and enteroaggregative Escherichia coli in the<br />

small bowel, this is by no means pathognomonic, 32<br />

nor is there a particular pattern of small-bowel<br />

microbial colonization or overgrowth seen in most<br />

cases. In parts of Africa endemic for HIV an association<br />

of persistent diarrhea with cryptosporidiosis<br />

and other pathogens 19–21,33 is well recognized,<br />

but may represent a manifestation of immunodeficiency.<br />

Malnutrition<br />

Increased macromolecular<br />

absorption and permeability<br />

Increased protein energy and<br />

micronutrient requirement for<br />

repair<br />

Protein energy malnutrition<br />

Persistent diarrhea is commonly seen in association<br />

with significant malnutrition, and the relationship<br />

may be bi-directional. It is widely recognized<br />

that diarrheal episodes, especially if<br />

invasive, may become prolonged in malnourished<br />

children. The recent evidence of micronutrient<br />

deficiencies, especially of zinc and vitamin A in<br />

malnourished children with persistent diarrhea,<br />

Prolonged small-intestinal mucosal injury<br />

and ineffective repair<br />

Risk factors for persistent diarrhea 195<br />

Immunological and inflammatory<br />

mucosal reaction (cytokine activation<br />

and altered growth factors)<br />

Decreased brush-border enzymes<br />

and transport<br />

Nutrient malabsorption<br />

Bacterial overgrowth, infection and<br />

possible translocation<br />

indicates impaired immunological mechanisms for<br />

clearing infections as well as ineffective mucosal<br />

repair mechanisms. From initial studies indicating<br />

the potential benefit of zinc supplementation on<br />

reducing the risk of prolonged diarrhea, 34 the<br />

evidence of the benefit of zinc supplements in<br />

children with persistent diarrhea was equivocal.<br />

35,36 However, a recent meta-analysis of zinc<br />

supplementation in diarrheal illnesses indicated a<br />

significant reduction in duration and severity of<br />

diarrheal illnesses. 37 Thus, zinc deficiency may<br />

significantly contribute to the prolongation of<br />

mucosal injury and delayed intestinal repair<br />

mechanisms.<br />

Dietary risk factors<br />

While many children with persistent diarrhea are<br />

lactose-intolerant, the role of specific dietary allergies<br />

in inducing and perpetuating enteropathy of<br />

malnutrition is unclear. Several studies have highlighted<br />

the high risk of prolonged diarrhea with<br />

lactation failure and early introduction of artificial<br />

feeds in developing countries. In particular, the<br />

administration of unmodified cow’s or buffalo’s


196<br />

Post-infectious persistent diarrhea in developing countries<br />

milk is associated with prolongation of diarrhea,<br />

suggesting the potential underlying role of milk<br />

protein enteropathy. 26,38<br />

Inappropriate management of acute diarrhea<br />

The association of prolongation of diarrhea with<br />

starvation and inappropriately prolonged administration<br />

of parenteral fluids has been recognized<br />

for over half a century. Continued breast feeding is<br />

important; unnecessary food withdrawal, and<br />

replacement of luminal nutrients, especially breast<br />

milk, with non-nutritive agents is a major factor in<br />

prolonging mucosal injury after diarrhea. In particular,<br />

blanket administration of antibiotics and antimotility<br />

agents and semi-starvation diets should be<br />

avoided in cases of prolonged diarrhea. 39,40 While<br />

parenteral nutrition has been occasionally life<br />

saving in selected cases in developing countries, 41<br />

it is clearly an impractical option for most of the<br />

developing world. There is now clear evidence<br />

supporting the enteral route for nutritional rehabilitation<br />

of malnourished children with persistent<br />

diarrhea. 12 Starvation has been shown to<br />

have deleterious effects on the intestinal mucosa, 42<br />

with a reduction in the nutritive transporters for<br />

glutamine and arginine. 43 It is therefore imperative<br />

that malnourished children with persistent<br />

diarrhea should receive enteral nutrition during<br />

their period of rehabilitation.<br />

The aforementioned risk factors highlight the<br />

importance of recognizing that optimal management<br />

of diarrheal episodes is key to the prevention<br />

of persistent diarrhea. It is thus necessary that,<br />

given the close relationship between diarrheal<br />

disorders and malnutrition, persistent diarrhea be<br />

widely recognized as a nutritional disorder, 44,45<br />

and optimal nutritional rehabilitation be considered<br />

as the cornerstone of its management. 12<br />

Principles of management of<br />

persistent diarrhea<br />

In general the management of persistent diarrhea<br />

in malnourished children represents a blend of the<br />

principles of management of diarrhea and malnutrition.<br />

Associated malnutrition may be quite<br />

severe in affected children, necessitating rapid<br />

nutritional rehabilitation, often in hospital. Given<br />

the chronicity of the disorder, prolonged hospitalization<br />

may be problematic in developing countries<br />

and, whenever possible, ambulatory or homebased<br />

therapy must be supported.<br />

The following represent the basic principles of<br />

management of persistent diarrhea. A suggested<br />

therapeutic approach is indicated in Figure 12.3.<br />

Rapid resuscitation, antibiotic therapy and<br />

stabilization<br />

Most children with persistent diarrhea and associated<br />

malnutrition are not severely dehydrated,<br />

and oral rehydration may be adequate. However,<br />

acute exacerbations and associated vomiting may<br />

require brief periods of intravenous rehydration<br />

with Ringer’s lactate. Acute electrolyte imbalance<br />

such as hypokalemia and severe acidosis may<br />

require correction. More importantly, associated<br />

systemic infections (bacteremia, pneumonia and<br />

urinary tract infection) are well recognized in<br />

severely malnourished children with persistent<br />

diarrhea and a frequent cause of early mortality.<br />

Children must be screened for these at admission.<br />

Almost 30–50% of malnourished children with<br />

persistent diarrhea may have an associated<br />

systemic infection requiring resuscitation and<br />

antimicrobial therapy. 46,47 In severely ill children<br />

requiring hospitalization, it may be best to cover<br />

with parenteral antibiotics at admission (usually<br />

ampicillin and gentamicin) while awaiting the<br />

results of cultures. It should be emphasized that<br />

there is little role for oral antibiotics in persistent<br />

diarrhea, as in most cases the original bacterial<br />

infection triggering the prolonged diarrhea has<br />

disappeared by the time the child presents. One<br />

possible exception may be adjunctive therapy for<br />

cryptosporidiosis in children with HIV and persistent<br />

diarrhea. 48<br />

Oral rehydration therapy<br />

This is the preferred mode of rehydration and<br />

replacement of on-going losses. While in general<br />

the standard WHO oral rehydration solution (ORS)<br />

is adequate, recent evidence indicates that hypoosmolar<br />

rehydration fluids 49,50 as well as cerealbased<br />

oral rehydration fluids may be advantageous<br />

in malnourished children. In general replacing<br />

each stool with about 50–100 ml ORS is safe.


Recovery<br />

Parental guidance to sustain feeding at home<br />

Follow-up<br />

Figure 12.3 Therapeutic approach to the management of persistent diarrhea.<br />

Enteral feeding and diet selection<br />

Irrespective of the cellular mechanisms and structural<br />

alterations in malnourished children with<br />

persistent diarrhea, the end result is one of altered<br />

brush-border and luminal enzymes, with con-<br />

Principles of management of persistent diarrhea 197<br />

Persistent diarrhea<br />

(diarrhea ≥ 14 days with malnutrition<br />

Assessment, resuscitation and early stabilization<br />

Intravenous and/or oral rehydration (hypo-osmolar)<br />

Treat electrolyte imbalance<br />

Screen and treat associated systemic infections<br />

Continued breast feeding<br />

Reduce lactose load by<br />

• Milk–cereal (usually rice-based) diet or<br />

• Replacement of milk with yogurt<br />

Micronutrient supplementation<br />

(zinc, vitamin A, folate)<br />

Growth monitoring Failure to recover<br />

Continued or recurrent diarrhea<br />

Poor weight gain<br />

Reinvestigate for infections<br />

Comminuted chicken diet or green banana diet<br />

Elemental feeds<br />

Continued diarrhea and dehydration<br />

Reinvestigate in the framework of the so-called<br />

'intractable diarrhea of infancy'<br />

Intravenous hyperalimentation<br />

sequent malabsorption. Despite the aforementioned<br />

alterations in digestive and absorptive<br />

mechanisms, analysis of studies of metabolic<br />

balance in children with persistent diarrhea indicates<br />

that satisfactory carbohydrate, protein and fat<br />

absorption can take place on a variety of diets. 12,38


198<br />

Post-infectious persistent diarrhea in developing countries<br />

It is exceedingly rare to find persistent diarrhea in<br />

exclusively breast-fed infants, and with the exception<br />

of situations where persistent diarrhea accompanies<br />

perinatally acquired HIV infection, breast<br />

feeding must be continued. Most children with<br />

persistent diarrhea are not lactose intolerant,<br />

although administration of a lactose load exceeding<br />

5g/kg per day is associated with higher purging<br />

rates and treatment failure. In general therefore<br />

withdrawal of milk and replacement with specialized<br />

(and expensive) lactose-free formulations is<br />

unnecessary. Alternative strategies for reducing<br />

the lactose load in malnourished children with<br />

persistent diarrhea include the addition of milk to<br />

cereals as well as replacement of milk with<br />

fermented milk products such as yogurt. These<br />

dietary interventions have now been extensively<br />

evaluated in several studies in South Asia, and<br />

found to be of equivalent efficacy to expensive<br />

formulations. 51,52<br />

Rarely, when dietary intolerance precludes the<br />

administration of cow’s milk-based formulations<br />

or milk, it may be necessary to administer specialized<br />

milk-free diets such as a comminuted or<br />

blenderized chicken-based diet or an elemental<br />

formulation. 53 It must be pointed out that,<br />

although effective in some settings, 54 the latter are<br />

unaffordable in most developing countries. In<br />

addition to rice–lentil formulations such as<br />

khitchri, the addition of green banana or pectin to<br />

the diet 55 has been shown to be effective in the<br />

treatment of persistent diarrhea.<br />

The usual energy density of any diet used for the<br />

therapy of persistent diarrhea should be around<br />

1kcal/g, aiming to provide an energy intake of a<br />

minimum of 100kcal/kg per day, and a protein<br />

intake of 2–3g/kg per day. In selected circumstances<br />

when adequate intake of energy-dense<br />

food is problematic, the addition of amylase to the<br />

diet through germination techniques may also be<br />

helpful.<br />

Micronutrient supplementation<br />

It is now widely recognized that most malnourished<br />

children with persistent diarrhea have associated<br />

deficiencies of micronutrients including<br />

zinc, iron and vitamin A. This may be a consequence<br />

of poor intake and continued enteral<br />

losses, and requires replenishment during<br />

therapy. 56 While the evidence supporting zinc<br />

administration in children with persistent diarrhea<br />

is persuasive, it is likely that these children<br />

have multiple micronutrient deficiencies.<br />

Concomitant vitamin A administration to children<br />

with persistent diarrhea has been shown to<br />

improve outcome 57,58 especially in HIV-endemic<br />

areas. 59 It is therefore important to ensure that all<br />

children with persistent diarrhea and malnutrition<br />

receive an initial dose of 100000U of vitamin A<br />

and a daily intake of at least 3–5mg/kg per day of<br />

elemental zinc. While the association of significant<br />

anemia with persistent diarrhea is well recognized,<br />

iron replacement therapy is best initiated<br />

only after recovery from diarrhea has started and<br />

the diet is well tolerated.<br />

Follow-up and nutritional rehabilitation in<br />

community settings<br />

Given the high rates of relapse in most children<br />

with persistent diarrhea, it is important to address<br />

the underlying risk factors and institute preventive<br />

measures. These include appropriate feeding<br />

(breast feeding, complementary feeding) and close<br />

attention to environmental hygiene and sanitation.<br />

This poses a considerable challenge in communities<br />

deprived of basic necessities such as clean<br />

water and sewage disposal.<br />

In addition to the preventive aspects, the challenge<br />

in most settings is to develop and sustain a form of<br />

dietary therapy using inexpensive, home-available<br />

and culturally acceptable ingredients that can be<br />

used to manage children with persistent diarrhea.<br />

Given that the majority of cases of persistent diarrhea<br />

occur in the community and that parents are<br />

frequently hesitant to seek institutional help, there<br />

is a need to develop and implement inexpensive<br />

and practical home-based therapeutic measures. 12<br />

Recent data indicate that it may be entirely feasible<br />

to do so in community settings. 60,61<br />

Conclusion<br />

Most of the knowledge and tools needed to prevent<br />

diarrhea-associated mortality in developing countries<br />

and especially persistent diarrhea are


available. These require concerted and sustained<br />

implementation in public health programs. 62<br />

Given the emerging evidence of the long-term<br />

impact of childhood diarrhea on developmental<br />

REFERENCES<br />

1. Black RE, Morris SS, Bryce J. Where and why are 10<br />

million children dying every year? Lancet 2003; 361:<br />

2226–2234.<br />

2. Snyder JD, Merson MH. The magnitude of the global<br />

problem of acute diarrheal disease: a review of active<br />

surveillance data. Bull World Heath Organ 1982; 60:<br />

605–613.<br />

3. Bern C, Martines J, Zoysa ID, Glass RI. The magnitude<br />

of the global problem of diarrheal disease: a ten year<br />

update. Bull World Health Organ 1992; 70: 705–714.<br />

4. Yusufzai M, Bhutta ZA. In Bhutta ZA, ed. Contemporary<br />

Issues in Childhood Diarrhea and Malnutrition. Oxford:<br />

Karachi, 1997.<br />

5. Kosek M, Bern C, Guerrant RL. The global burden of<br />

diarrheal disease as estimated from studies published<br />

between 1990 and 2000. Bull World Health Organ 2003;<br />

81; 197–204.<br />

6. Moore SR, Lima AA, Schorling JB et al. Changes over<br />

time in the epidemiology of diarrhea and malnutrition<br />

among children in an urban Brazilian shantytown, 1989<br />

to 1996. Int J Infect Dis 2000; 4: 179–186.<br />

7. Bhan MK, Bhandari N, Sazawal S et al. Descriptive<br />

epidemiology of persistent diarrhea among young<br />

children in rural north India. Bull World Health Organ<br />

1989; 67: 281–288.<br />

8. Khan SR, Jalil F, Zaman S et al. Early child health in<br />

Lahore, Pakistan: X. Mortality. Acta Paediatr Suppl 1993;<br />

390: 109–117.<br />

9. Black RE, Brown KH, Becker S, Yunus M. Longitudinal<br />

studies of infectious diseases and physical growth of<br />

children in rural Bangladesh. I. Patterns of morbidity.<br />

Am J Epidemiol 1982; 115: 305–314.<br />

10. Lanata CF, Black RE, Gilman RH et al. Epidemiologic,<br />

clinical, and laboratory characteristics of acute vs<br />

persistent diarrhea in periurban Lima, Peru. J Pediatr<br />

Gastroenterol Nutr 1991; 12: 82–88.<br />

11. Mahmud A, Jalil F, Karlberg J, Lindblad BS. Early child<br />

health in Lahore, Pakistan: VII. Diarrhea. Acta Paediatr<br />

Suppl 1993; 390: 79–85.<br />

12. Bhutta ZA, Hendricks KM. Nutritional management of<br />

persistent diarrhea in childhood: a perspective from the<br />

developing world. J Pediatr Gastroenterol Nutr 1996; 22:<br />

17–37.<br />

13. Briend A. Is diarrhea a major cause of malnutrition<br />

among under-fives in developing countries? A review of<br />

available evidence. Eur J Clin Nutr 1990; 44: 611–628.<br />

14. Fauveau V, Henry FJ, Briend A et al. Persistent diarrhea<br />

as a cause of childhood mortality in rural Bangladesh.<br />

Acta Paediatr Suppl 1992; 381: 12–14.<br />

15. Baqui AH, Black RE, Sack RB et al. Malnutrition, cellmediated<br />

immune deficiency and diarrhea: a<br />

community-based longitudinal study in rural<br />

Bangladeshi children. Am J Epidemiol 1993; 137:<br />

355–365.<br />

References 199<br />

outcomes, 63 it is imperative that due emphasis is<br />

placed on prompt recognition and appropriate<br />

management of persistent diarrhea.<br />

16. Azim T, Ahmad SM, Sefat-E-Khuda et al. Immune<br />

response of children who develop persistent diarrhea<br />

following rotavirus infection. Clin Diagn Lab Immunol<br />

1999; 6: 690–695.<br />

17. Raqib R, Mia SM, Qadri F et al. Innate immune<br />

responses in children and adults with Shigellosis. Infect<br />

Immun 2000; 68: 3620–3629.<br />

18. Taniguchi K, Rikimaru T, Yartey JE et al. Immunological<br />

background in children with persistent diarrhea in<br />

Ghana. Pediatr Int 1999; 41: 162–167.<br />

19. Amadi B, Kelly P, Mwiya M et al. Intestinal and<br />

systemic infection, HIV, and mortality in Zambian children<br />

with persistent diarrhea and malnutrition. J Pediatr<br />

Gastroenterol Nutr 2001; 32: 550–554.<br />

20. Tumwine JK, Kekitiinwa A, Nabukeera N et al.<br />

Enterocytozoon bieneusi among children with diarrhea<br />

attending Mulago Hospital in Uganda. Am J Trop Med<br />

Hyg 2002; 67: 299–303.<br />

21. Chokephaibulkit K, Wanachiwanawin D, Tosasuk K et<br />

al. Intestinal parasitic infections among human<br />

immunodeficiency virus-infected and<br />

-uninfected children hospitalized with diarrhea in<br />

Bangkok, Thailand. Southeast Asian J Trop Med Public<br />

Health 2001; 32: 770–775.<br />

22. Sullivan PB, Marsh MN, Mirakian R et al. Chronic<br />

diarrhea and malnutrition – histology of the small<br />

intestinal lesion. J Pediatr Gastroenterol Nutr 1991; 12:<br />

195–203.<br />

23. Shiner M, Nichols VN, Barrish JP, Nichols BL.<br />

Pathogenesis of small-intestinal mucosal lesions in<br />

chronic diarrhea of infancy: II. An electron microscopic<br />

study. J Pediatr Gastroenterol Nutr 1990; 11: 464–480.<br />

24. Fagundes-Neto U, De Martini-Costa S, Pedroso MZ,<br />

Scaletsky IC. Studies of the small bowel surface by<br />

scanning electron microscopy in infants with persistent<br />

diarrhea. Braz J Med Biol Res 2000; 33: 1437–1442.<br />

25. Manuel PD. The role of cow’s milk protein intolerance<br />

in chronic diarrhea in a developing community. In<br />

Walker-Smith JA, McNeish AS, eds. Diarrhea and<br />

Malnutrition in Childhood. London: Butterworth, 1986:<br />

193–199.<br />

26. Lunn PG, Northrop-Clewes CA, Downes RM. Chronic<br />

diarrhea and malnutrition in the Gambia: studies on<br />

intestinal permeability. Trans R Soc Trop Med Hyg 1991;<br />

85: 8–11.<br />

27. Sullivan PB, Lunn PG, Northrop-Clewes C et al.<br />

Persistent diarrhea and malnutrition – the impact of<br />

treatment on small bowel structure and permeability. J<br />

Pediatr Gastroenterol Nutr 1992; 14: 208–215.<br />

28. Jakobsson I. Intestinal permeability in children of<br />

different ages and with different gastrointestinal<br />

diseases. Pediatr Allergy Immunol 1993; 4(Suppl 3):<br />

33–39.


200<br />

Post-infectious persistent diarrhea in developing countries<br />

29. Holm S, Lindberg T, Gothefors L. Macromolecular<br />

absorption during and after gastroenteritis in children.<br />

Acta Pediatr Scand 1992; 81: 585–588.<br />

30. Okeke IN, Nataro JP. Enteroaggregative Escherichia coli.<br />

Lancet Infect Dis 2001; 1: 304–313.<br />

31. Unicomb LE, Banu NN, Azim T et al. Astrovirus<br />

infection in association with acute, persistent and<br />

nosocomial diarrhea in Bangladesh. Pediatr Infect Dis J<br />

1998; 17: 611–614.<br />

32. Fraser D, Dagan R, Porat N et al. Persistent diarrhea in a<br />

cohort of Israeli Bedouin infants: role of enteric<br />

pathogens and family and environmental factors. J<br />

Infect Dis 1998; 178: 1081–1088.<br />

33. Germani Y, Minssart P, Vohito M et al. Etiologies of<br />

acute, persistent, and dysenteric diarrheas in adults in<br />

Bangui, Central African Republic, in relation to human<br />

immunodeficiency virus serostatus. Am J Trop Med Hyg<br />

1998; 59: 1008–1014.<br />

34. Sazawal S, Black RE, Bhan MK et al. Zinc supplementation<br />

in young children with acute diarrhea in India. N<br />

Engl J Med 1995; 333: 839–844.<br />

35. Bhutta ZA, Nizami SQ, Isani Z. Zinc supplementation<br />

during persistent diarrhoea of childhood in Pakistan.<br />

Pediatrics 1999; 103: e42.<br />

36. Roy SK, Tomkins AM, Mahalanabis D et al. Impact of<br />

zinc supplementation on persistent diarrhoea in<br />

malnourished Bangladeshi children. Acta Paediatr 1998;<br />

87: 1235–1239.<br />

37. Bhutta ZA, Bird SM, Black RE et al. Therapeutic effects<br />

of oral zinc in acute and persistent diarrhea in children<br />

in developing countries: pooled analysis of randomized<br />

controlled trials. Am J Clin Nutr 2000; 72: 1516–1522.<br />

38. Bhutta ZA, Molla AM, Isani Z et al. Nutrient absorption<br />

and weight gain in persistent diarrhoea: comparison of a<br />

traditional rice–lentil–yogurt–milk diet with soy<br />

formula. J Pediatr Gastroenterol Nutr 1994; 18: 45–52.<br />

39. Sodemann M, Jakobsen MS, Molbak K et al. Episodespecific<br />

risk factors for progression of acute diarrhoea to<br />

persistent diarrhoea in west African children. Trans R<br />

Soc Trop Med Hyg 1999; 93: 65–68.<br />

40. Mahmud MA, Hossain MM, Huang DB et al.<br />

Sociodemographic, environmental and clinical risk<br />

factors for developing persistent diarrhoea among<br />

infants in a rural community of Egypt. J Health Popul<br />

Nutr 2001; 19: 313–319.<br />

41. Vaidya U, Bhave S, Pandit A. Parenteral nutrition in the<br />

management of severe protracted diarrhea. Indian J<br />

Pediatr 1993; 60: 19–24.<br />

42. Illig KA, Ryan CK, Hardy DJ et al. Total parenteral<br />

nutrition-induced changes in gut mucosal function:<br />

atrophy alone is not the issue. Surgery 1992; 112:<br />

631–637.<br />

43. Sarac TP, Souba WW, Miller JH et al. Starvation induces<br />

differential small bowel luminal amino acid transport.<br />

Surgery 1994; 116: 679–686.<br />

44. Lo CW, Walker WA. Chronic protracted diarrhea in<br />

infancy: a nutritional disease. Pediatrics 1983; 72:<br />

786–800.<br />

45. Lima AA, Moore SR, Barboza MS Jr et al. Persistent<br />

diarrhea signals a critical period of increased diarrhea<br />

burdens and nutritional shortfalls: a prospective cohort<br />

study among children in northeastern Brazil. J Infect Dis<br />

2000; 181: 1643–1651.<br />

46. Bhutta ZA, Nizami SQ, Thobani S. Factors determining<br />

recovery during nutritional therapy of persistent<br />

diarrhoea: the impact of diarrhoea severity and<br />

intercurrent infections. Acta Paediatrica 1997; 86:<br />

796–802.<br />

47. Alam NH, Faruque AS, Dewan N et al. Characteristics of<br />

children hospitalized with severe dehydration and<br />

persistent diarrhoea in Bangladesh. J Health Popul Nutr<br />

2001; 19: 18–24.<br />

48. Amadi B, Mwiya M, Musuku J et al. Effect of<br />

nitazoxanide on morbidity and mortality in Zambian<br />

children with cryptosporidiosis: a randomised<br />

controlled trial. Lancet 2002; 360: 1375–1380.<br />

49. Sarker SA, Mahalanabis D, Alam NH et al. Reduced<br />

osmolarity oral rehydration solution for persistent<br />

diarrhea in infants: a randomized controlled clinical<br />

trial. J Pediatr 2001; 138: 532–538.<br />

50. Dutta P, Mitra U, Dutta S et al. Hypo-osmolar oral<br />

rehydration salts solution in dehydrating persistent<br />

diarrhoea in children: double-blind, randomized,<br />

controlled clinical trial. Acta Paediatr 2000; 89:<br />

411–416.<br />

51. Bhutta ZA, Molla AM, Isani Z et al. Dietary<br />

management of persistent diarrhoea: comparison of a<br />

traditional rice–lentil based diet with soy formula.<br />

Pediatrics 1991; 88: 1010–1018.<br />

52. Ashraf H, Ahmed S, Fuchs GJ, Mahalanabis D.<br />

Persistent diarrhoea: associated infection and response<br />

to a low lactose diet. J Trop Pediatr 2002; 48: 142–148.<br />

53. International Working Group on Persistent Diarrhea.<br />

Evaluation of the efficacy of an algorithm for the<br />

treatment of persistent diarrhea: a multicentre study.<br />

Bull World Health Organ 1996; 74: 479–489.<br />

54. Amadi B. Role of food antigen elimination in treating<br />

children with persistent diarrhea and malnutrition in<br />

Zambia. J Pediatr Gastroenterol Nutr 2002; 34 (Suppl 1):<br />

S54–S56.<br />

55. Rabbani GH, Teka T, Zaman B et al. Clinical studies in<br />

persistent diarrhea: dietary management with green<br />

banana or pectin in Bangladeshi children.<br />

Gastroenterology 2001; 121: 554–560.<br />

56. Mahalanabis D, Bhan MK. Micronutrients as adjunct<br />

therapy of acute illness in children: impact on the<br />

episode outcome and policy implications of current<br />

findings. Br J Nutr 2001; 85 (Suppl 2): S151–S158.<br />

57. Rahman MM, Vermund SH, Wahed MA et al.<br />

Simultaneous zinc and vitamin A supplementation in<br />

Bangladeshi children: randomised double blind<br />

controlled trial. BMJ 2001; 323: 314–318.<br />

58. Khatun UH, Malek MA, Black RE et al. A randomized<br />

controlled clinical trial of zinc, vitamin A or both in<br />

undernourished children with persistent diarrhea in<br />

Bangladesh. Acta Paediatr 2001; 90: 376–380.<br />

59. Villamor E, Mbise R, Spiegelman D et al. Vitamin A<br />

supplements ameliorate the adverse effect of HIV-1,<br />

malaria, and diarrheal infections on child growth.<br />

Pediatrics 2002; 109: E6.<br />

60. Bhandari N, Bahl R, Saxena M et al. Prognostic factors<br />

for persistent diarrhoea managed in a community<br />

setting. Indian J Pediatr 2000; 67: 739–745.<br />

61. Valentiner-Branth P, Steinsland H, Santos G et al.<br />

Community-based controlled trial of dietary<br />

management of children with persistent diarrhea:<br />

sustained beneficial effect on ponderal and linear<br />

growth. Am J Clin Nutr 2001; 73: 968–974.<br />

62. Jones GR, Steketee RW, Black RE, Bhutta ZA, Morris SS<br />

and the Bellagio Child Survival Study Group. How<br />

many child deaths can we prevent this year? Lancet<br />

2003; 362: 65–71.<br />

63. Niehaus MD, Moore SR, Patrick PD et al. Early<br />

childhood diarrhea is associated with diminished<br />

cognitive function 4 to 7 years later in children in a<br />

northeast Brazilian shantytown. Am J Trop Med Hyg<br />

2002; 66: 590–593.


13<br />

Introduction<br />

Small-bowel bacterial<br />

overgrowth<br />

Mauro Batista de Morais and Ulysses Fagundes-Neto<br />

Small-bowel bacterial overgrowth (SBBO) is characterized<br />

by the colonization of the small intestine<br />

by bacteria that are normally found only in the<br />

colonic microbiota.<br />

In the literature, SBBO has received many names<br />

such as ‘the blind loop syndrome’, 1 ‘stagnant loop<br />

syndrome’, 2 ‘small intestinal stasis syndrome’, 3<br />

and the ‘contaminated small bowel syndrome’. 4<br />

Initially, SBBO descriptions predominated in<br />

patients with surgical or anatomical abnormalities,<br />

and later SBBO was recognized in children who<br />

did not present anatomical abnormalities, in situations<br />

such as protein–energy malnutrition, acute<br />

diarrhea, persistent diarrhea and environmental<br />

enteropathy. These findings characterize SBBO as<br />

an important public health problem, occurring<br />

especially in the pediatric population in lessdeveloped<br />

countries. SBBO can cause asymptomatic<br />

intestinal malabsorption and/or chronic<br />

diarrhea and may be associated with protein–<br />

energy malnutrition and stunting.<br />

Etiology<br />

SBBO is a syndrome that can cause functional and<br />

morphological abnormalities of the digestive tract,<br />

resulting in a disturbance in the homeostasis of the<br />

bacterial flora in the small intestine and a consequent<br />

imbalance of the mechanisms that control<br />

the flora. 5,6 Therefore, for better understanding of<br />

the development of SBBO, the microflora that is<br />

normally found in the digestive tract and its regulatory<br />

mechanisms are briefly described.<br />

It is generally accepted that the upper digestive<br />

tract (stomach, duodenum, jejunum) is sterile, as<br />

found in 70% of the population, or may present a<br />

scarce microflora made up of facultative microorganisms,<br />

predominantly Gram-positive bacteria.<br />

The concentration of bacteria (Streptococcus,<br />

Lactobacillus, aerobes and diphtheroids) and yeast<br />

is normally no greater than 10 4 colonies/ml in the<br />

fluid of the small bowel. These micro-organisms<br />

come from the oral cavity, colonizing the upper<br />

small intestine after surviving the gastric juice.<br />

The terminal portion of the ileum comprises a<br />

transition zone and the microbiota begins to<br />

change with the appearance of Gram-negative<br />

bacteria, such as coliforms and bacteroids. The<br />

ileocecal valve acts as a true barrier, separating the<br />

Gram-positive species, which predominate in the<br />

upper small intestine, from the Gram-negative<br />

species, which inhabit the colon, where the anaerobic<br />

bacterial population represents the main<br />

portion of colonic microflora. Bacteroids, anaerobic<br />

Lactobacilli and Clostridium spp are the main<br />

components of the colonic flora and exist in<br />

concentrations ranging between 10 8 and 10 11<br />

colonies/ml, surpassing the facultative or aerobic<br />

microflora in a proportion of 1000–10000:1. Table<br />

13.1 shows the microflora of various segments of<br />

the digestive tract. 5–7<br />

The maintenance of the intestinal microbiota with<br />

the characteristics mentioned above depends upon<br />

the action of various regulatory mechanisms, such<br />

as diet, digestive tube motility, gastric acidity, the<br />

intestinal immunological system and the integrity<br />

of the ileocecal valve. 5,6<br />

Diet type<br />

Animals raised in sterile environmental conditions,<br />

including feed, are different from animals<br />

raised under normal conditions that present digestive<br />

tract colonization. Animals free of bacteria<br />

201


202<br />

Small-bowel bacterial overgrowth<br />

Table 13.1 Microflora of the digestive tract<br />

Concentration of<br />

Site micro-organisms/ml Description<br />

Stomach, duodenum,


intestinal pseudo-obstruction syndrome, which<br />

is associated with profound abnormalities of<br />

intestinal motility, SBBO is frequent. 10<br />

Intestinal mucosal immunity<br />

Intestinal mucosal immune responses are different<br />

from those that occur systemically. One of<br />

the fundamental differences is the production of<br />

immunoglobulin A (IgA), which is secreted in<br />

the intestine. IgA is the main class of antibodies<br />

produced in the intestine, as illustrated by the<br />

larger number of IgA-producing cells: a proportion<br />

of 20 for each IgG-producing cell and three<br />

for each producing IgM. Secretory IgA differs<br />

from serum IgA, as it is in dimeric form, created<br />

by the secretory piece, which confers resistance<br />

against proteolysis in the intestinal lumen.<br />

Secretory IgA can impede the adherence of<br />

micro-organisms to the surface of enterocytes, an<br />

important control mechanism to prevent SBBO.<br />

It is important to emphasize that newborns have<br />

a smaller population of plasma cells in the<br />

lamina propria, which explains their reduced<br />

ability to respond to local antigenic stimuli. At<br />

this stage of life, the colostrum and mother’s<br />

milk are rich in secretory IgA and play an important<br />

role in the newborn’s defense mechanism.<br />

5,7,11<br />

Ileocecal valve<br />

This valve acts as a barrier separating Grampositive<br />

bacteria that inhabit the proximal small<br />

intestine from the Gram-negative bacteria and<br />

anaerobes that comprise the colonic bacterial<br />

flora. In the absence of the ileocecal valve due to<br />

intestinal resection, a retrograde colonization of<br />

the small intestine with bacteria that are<br />

normally found only in the colon is observed. 6<br />

SBBO develops when an alteration of one or<br />

more of the intestinal mucosa regulatory mechanisms<br />

occurs. Theoretically, bacteria responsible<br />

for SBBO may come either from the environment,<br />

by the ingestion of contaminated food or<br />

water, or from the colonic microflora itself, as a<br />

result of proximal colonization due to intestinal<br />

motility disturbances or ileocecal valve functional<br />

insufficiency.<br />

Pathophysiology<br />

Pathophysiology 203<br />

In patients with SBBO, functional and/or morphological<br />

alterations can be observed, owing to the<br />

capacity of these bacteria to promote deconjugation<br />

of bile salts, such as 7α-dehydroxylation,<br />

which transforms them into secondary bile salts.<br />

The deconjugation of bile salts causes a reduction<br />

in the capacity to form mixed micelle, which is<br />

important in the process of lipid digestion and<br />

absorption, because the concentration in the<br />

intestinal lumen does not achieve a critical micellar<br />

concentration. Therefore, lower solubility of<br />

fats in the diet causing steatorrhea is observed. On<br />

the other hand, the secondary bile acids (deoxycholic<br />

and lithocholic) have a large capacity to<br />

provoke lesions in the intestinal mucosa, causing<br />

partial villous atrophy that may be associated with<br />

the secretion of electrolytes and malabsorption of<br />

glucose. 5,12,13<br />

Patients with SBBO also present reduced absorption<br />

of carbohydrates from the diet by:<br />

(1) Metabolism of carbohydrates by the SBBO<br />

bacteria, which generates short-chain fatty<br />

acids (acetic, propionic and butyric acids)<br />

and gases, such as hydrogen, which, after<br />

diffusion through the intestinal mucosa, pass<br />

through to the circulatory system and reach<br />

the lungs, where they are eliminated in<br />

respiration.<br />

(2) Reduction in the activity of disaccharidases<br />

due to alterations in the ultrastructure of the<br />

small intestine.<br />

Consequently, the unabsorbed carbohydrates<br />

remain in the intestinal lumen in their original<br />

form or in the form of short-chain fatty acids as a<br />

result of bacterial fermentation, generating<br />

osmotic pressure that promotes the secretion of<br />

water into the intestinal lumen. 5,11<br />

Evidence also exists of a reduction in protein<br />

digestion and absorption from the diet due to<br />

intraluminal protein catabolism as well as a reduction<br />

in the transport and uptake of amino acids by<br />

the intestinal mucosa. 5,11<br />

SBBO may also cause malabsorption of liposoluble<br />

vitamins A and D accompanied by steatorrhea.<br />

Reduction in the absorption of vitamin B12 may


204<br />

Small-bowel bacterial overgrowth<br />

Table 13.2 Probable effects of bacterial overgrowth<br />

Intraluminal Mucosal Systemic<br />

Bile salt deconjugation Deficiency of disaccharidases Absorption of bacterial toxins<br />

and antigens<br />

Decrease of the biliary Enterocyte damage Hepatic inflammation<br />

salt ‘pool’<br />

Steatorrhea Inflammation Immune complex formation<br />

Vitamin B12 malabsorption Protein loss Vasculitis<br />

Production of short-chain Bleeding Polyarthritis<br />

fatty acids<br />

occur, owing to the consumption of vitamins by<br />

bacteria present in the intestinal lumen, especially<br />

Bacteroides. 5,11<br />

Systemic effects of SBBO are associated with the<br />

absorption of antigens and bacterial products<br />

through the injured intestinal mucosa. 6 Table 13.2<br />

lists the effects of an overgrowth of bacteria on its<br />

host.<br />

Diagnosis<br />

Research to characterize the digestive tract<br />

microflora has developed mainly from the 1960s<br />

onward. The micro-organisms of the digestive tube<br />

flora, made up of aerobic and anaerobic bacteria,<br />

protozoa and even some viruses, form a complex<br />

ecosystem. The terms ‘indigenous flora’ and<br />

‘microbiota’ are used as synonyms for normal flora<br />

and refer to micro-organisms that can be found in<br />

normal individuals. Therefore, in order to establish<br />

a diagnosis for SBBO, it is necessary to define<br />

the location, the count and the type of bacteria<br />

found. In general, studies regarding normal flora<br />

have been carried out by analyzing fluid collected<br />

by intubation of the Treitz angle region. Dickman<br />

et al 14 studied the intestinal fluid of 22 healthy<br />

individuals, finding 10 3 bacteria/ml in 16 of them<br />

and 10 4 bacteria/ml in two individuals. In another<br />

study, Thadepalli et al 15 found up to 10 4 bacteria/ml<br />

in six of 28 individuals. Fagundes-Neto et<br />

al 16 evaluated the small-bowel microflora in chil-<br />

dren who were not undernourished or presenting<br />

diarrhea, and found similar numbers of bacteria.<br />

Many authors 17–27 characterize SBBO based on<br />

bacterial count, without taking into consideration<br />

the family or genus of these micro-organisms.<br />

According to this quantitative principle, various<br />

cut-off points can be found in the literature:<br />

(1) ≥ 10 4 bacteria/ml; 17–21<br />

(2) ≥ 10 5 bacteria/ml; 22–25<br />

(3) ≥ 10 6 bacteria/ml. 26,27<br />

Other authors 28,29 have used criteria that take into<br />

consideration the family as well as the number of<br />

the bacteria, characterizing SBBO when ≥10 3<br />

coliforms and aerobic or anaerobic enterobacteria/ml<br />

are found in the intestinal fluid.<br />

Finally, there is a group of authors that considers<br />

SBBO to be the presence, in any concentration, of<br />

aerobic or anaerobic bacteria of the colonic<br />

microflora. 16,30–33<br />

In spite of the lack of consensus on the interpretation<br />

of results, the intestinal fluid culture is<br />

considered the gold standard test for the diagnosis<br />

of SBBO. 6,22,28 However, the invasive character of<br />

duodenal intubation and the high cost of aerobic<br />

and anaerobic microbiological study have driven<br />

research into lower cost, non-invasive methods, of<br />

which the hydrogen breath test has shown itself to<br />

be the most important.


The basic principle of the hydrogen breath test is<br />

the fermentation of carbohydrates by bacteria<br />

present in the intestinal lumen, which generates,<br />

among other products, hydrogen. After it diffuses<br />

through the intestinal mucosa and reaches the<br />

lungs through the circulatory system, the hydrogen<br />

is eliminated in exhaled air. Normally,<br />

fermenting bacteria are found only in the colon:<br />

so, when an unabsorbed carbohydrate reaches the<br />

colon, it is fermented and the elimination of<br />

hydrogen through the breath occurs about 10 min<br />

after it reaches the large intestine. Under these<br />

circumstances, the carbohydrate must travel the<br />

entirety of the small intestine for it to begin to<br />

produce hydrogen. This orocecal transit time<br />

varies according to the type of carbohydrate and its<br />

concentration in the administered solution. The<br />

lactulose solution employed in the hydrogen<br />

breath test for SBBO diagnosis presents a mean<br />

orocecal transit time of approximately 90 min.<br />

When SBBO is present, bacterial fermentation<br />

occurs prematurely, while still in the small intestine,<br />

elevating the concentration of hydrogen in<br />

the breath in the samples collected in the 60 min<br />

following the ingestion of the carbohydrate.<br />

Ideally, when the unabsorbed carbohydrate<br />

reaches the colon, it will produce a second peak of<br />

hydrogen corresponding to its fermentation by the<br />

Hydrogen (ppm)<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Normal<br />

Diagnosis 205<br />

colonic flora. Thus, after the ingestion of a carbohydrate<br />

that is not absorbed, such as lactulose, in<br />

individuals with bacterial overgrowth, two peaks<br />

in hydrogen production are expected: the first is an<br />

early peak, generated by the bacteria of the SBBO,<br />

while the second is a late peak, due to the normal<br />

production of hydrogen in the colon, as illustrated<br />

in Figure 13.1. The other carbohydrate used in the<br />

hydrogen breath test is glucose. As glucose is, in<br />

ordinary circumstances, completely absorbed by<br />

the small intestine, there should be no delayed<br />

peak, except if the patient presents intestinal<br />

malabsorption of glucose associated with<br />

SBBO. 19,34–36<br />

Most studies relating the hydrogen breath test to<br />

the culture of intestinal fluid have been carried out<br />

in adults. However, common criteria were not used<br />

for the interpretation of intestinal fluid cultures,<br />

which is considered the gold standard for the characterization<br />

of SBBO. The main points of these<br />

studies are summarized in Table 13.3. 22,26,35,37–39<br />

To our knowledge, only six studies have been<br />

carried out in children. Davidson et al 40 studied<br />

nine children aged 2–34 months, and characterized<br />

SBBO as the presence of more than 10 4 bacteria/ml<br />

in duodenal fluid. Lactose, sucrose and<br />

lactulose were used as substrates. SBBO was deter-<br />

Bacterial overgrowth<br />

(double peak)<br />

0 50 100 150 200 250 300<br />

Time (min)<br />

Non-producer<br />

Figure 13.1 Schematic representation of possible results of the hydrogen breath test with lactulose.


206<br />

Small-bowel bacterial overgrowth<br />

Table 13.3 Studies in adults evaluating the diagnostic performance of the hydrogen breath test for<br />

the diagnosis of small-bowel bacterial overgrowth<br />

Authors and Number of Criteria for Sensitivity Specificity<br />

reference patients Probe bacterial overgrowth* (%) (%)<br />

King and Toskes 26 20 lactulose 10g >10ppm 61 41<br />

Kerlin and Wong 37 45 glucose 50g >12ppm 93 78<br />

Corazza et al 35 77 lactulose 12g >10ppm 68 44<br />

Riordan et al 38 42 lactulose 10g >16ppm 20 75<br />

Riordan et al 22 28 lactulose 10g >10ppm 16 70<br />

MacMahon et al 39 30 glucose 50g >10ppm 75 30<br />

*Increment of breath hydrogen concentration before colonic peak<br />

mined by the presence of a double peak. Boissieu<br />

et al 41 studied cultures of intestinal fluid and the<br />

hydrogen breath test after administering glucose to<br />

five children. A study carried out by Khin-Maung<br />

et al, 27 with 19 children aged from 3 to 5 years,<br />

considered SBBO to be a concentration of more<br />

than 10 6 bacteria/ml in the enteral fluid. After<br />

ingestion of 10g of lactulose, an increase of more<br />

than 10ppm in samples collected after 20, 40 and<br />

60min was considered indicative of SBBO. Only<br />

two of the children with positive cultures<br />

presented a hydrogen breath test indicative of<br />

SBBO. Furthermore, three of the nine children<br />

with positive cultures did not present a positive<br />

hydrogen breath test indicative of SBBO.<br />

Marcelino 42 carried out a study in our institution<br />

and observed that only three of 18 unweaned<br />

infants, with acute or persistent diarrhea and<br />

SBBO, according to an intestinal fluid culture of<br />

the small intestine, presented a rise in breath<br />

hydrogen of more than 20ppm after the ingestion<br />

of 10g of lactulose. It should be pointed out that<br />

almost 80% of the unweaned infants studied were<br />

not hydrogen producers and many presented<br />

secondary lactose malabsorption.<br />

Guno et al 43 studied SBBO in 31 infants with ages<br />

ranging from 2 to 24 months suffering from<br />

protein–energy malnutrition. The culture of the<br />

small-bowel fluid revealed that SBBO was present<br />

in ten of the 31 patients. The sensitivity and specificity<br />

of the hydrogen breath test using lactulose<br />

was 72% and 90%, respectively, while the scores<br />

for the hydrogen breath test using glucose were<br />

55% and 90%, respectively. The authors did not<br />

include criteria for interpretation of the breath test.<br />

Silva 33 compared the results of the culture of<br />

aerobic and anaerobic organisms in the intestinal<br />

fluid of 31 children, with ages ranging from 6<br />

months to 16 years, with the increase in breath<br />

hydrogen after the ingestion of 10g of lactulose.<br />

SBBO was characterized by the presence of any<br />

bacterium from the colonic flora in the duodenum,<br />

and was found in 16 (51.6%) of the 31 children.<br />

Given the minimum increase of 10ppm in breath<br />

hydrogen concentration in the samples collected<br />

up to 60min, the sensitivity of the hydrogen test<br />

was 75% and specificity 60%.<br />

Analysis of the available information shows that<br />

both in adults and in children, irrespective of the<br />

carbohydrate that is used – lactulose or glucose –<br />

the occurrence of false-positive and false-negative<br />

results in the hydrogen breath test in relation to<br />

culture requires cautious application of this test in<br />

researching SBBO in a individual patient.<br />

Another utilization of the hydrogen breath test is<br />

the analysis of groups of individuals in order to<br />

obtain information about the bacterial flora of the


Increment of the concentration of H 2 (ppm)<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

0 20 40 60 80 100 120 140 160 180 200<br />

digestive tract in varying environmental conditions.<br />

The results obtained by Silva, 33 with two<br />

groups of children (15 with SBBO and 16 without<br />

SBBO) can, in this light, be reanalyzed differently.<br />

Figure 13.2 presents the median values of<br />

increases in hydrogen on expiration. It can be seen<br />

that the SBBO groups produced a greater amount<br />

of hydrogen after 60min, and that there was a<br />

statistically significant difference in hydrogen<br />

increases at 120, 150 and 180min of the test. The<br />

data suggest two possibilities: that individuals<br />

with SBBO may not present a double peak of<br />

hydrogen; or that the colonic flora of children with<br />

SBBO has a greater hydrogen-producing capacity.<br />

This is based on the observation that, at the beginning<br />

of the test, the difference in hydrogen production<br />

in expired breath in SBBO patients was lower<br />

than after 60min of the test. This second aspect<br />

was further analyzed using the area under the<br />

curve as the basis for calculation. In the fasting<br />

sample until 60min, the 16 patients with SBBO<br />

presented a median hydrogen production rate<br />

(720 ppm/min) not statistically different from the<br />

15 patients without SBBO (923 ppm/min). In turn,<br />

from 60 to 180min this parameter was signifi-<br />

Time (min)<br />

Clinical presentation 207<br />

Figure 13.2 Median of the increment of the concentration of hydrogen in the expired air of 16 patients with bacterial<br />

overgrowth (unbroken line) and in 15 patients without bacterial overgrowth (dotted line) according to culture of the intestinal<br />

fluid. Differences at 15min, p = 0.98; 30 min, p = 0.90; 45min, p = 0.58; 60min, p = 0.26; 90 min, p = 0.075; 120 min,<br />

p= 0.028; 150min, p = 0.031; 180 min, p =0.007.<br />

cantly higher (p=0.017) in SBBO patients<br />

(6943 ppm/min) compared to patients without<br />

SBBO (3074ppm/min).<br />

Clinical presentation<br />

SBBO may be asymptomatic or may present with<br />

chronic diarrhea associated with malabsorption of<br />

macronutrients. SBBO may thus lead to or aggravate<br />

protein–energy malnutrition. 5,6 In children,<br />

SBBO has also been linked to recurrent abdominal<br />

pain that responded to antimicrobial treatment. 41<br />

Several disorders may predispose to the development<br />

of SBBO (Table 13.4). It is therefore crucial<br />

that in every child diagnosed with SBBO a careful<br />

work-up be performed, aimed at ruling out these<br />

underlying conditions.<br />

We will focus here on SBBO associated with acute<br />

or persistent diarrhea and with environmental<br />

factors, given their high incidence and prevalence,<br />

particularly in young infants and in children living<br />

in unfavorable environments.


208<br />

Small-bowel bacterial overgrowth<br />

Table 13.4 Clinical conditions associated<br />

with small-bowel bacterial overgrowth<br />

Anatomic abnormalities<br />

Congenital intestinal obstruction<br />

Acquired strictures and stenosis<br />

(e.g. Crohn’s disease)<br />

Intestinal fistula<br />

Abnormalities of intestinal motility<br />

Pseudo-obstruction syndrome<br />

Scleroderma<br />

Damage of the myenteric plexus<br />

Post-surgery complications<br />

Stasis of the afferent loop<br />

Gastroenterostomy<br />

Enteroenterostomy<br />

Colectomy or jejunoileal anastomosis<br />

Without anatomic abnormalities<br />

Carbohydrate malabsorption<br />

Immunological deficiency<br />

Giardia infection<br />

Acute and persistent diarrhea<br />

Environmental enteropathy<br />

Small-bowel bacterial overgrowth in acute and<br />

persistent diarrhea<br />

In developing countries, these two conditions<br />

account for a major proportion of deaths in the<br />

early years of life, and it should be emphasized<br />

that mortality due to persistent infectious diarrhea<br />

is much greater than that due to its acute<br />

phase. 44,45 SBBO does occur in children with acute<br />

diarrhea and is considered one of the factors<br />

leading to its perpetuation. In the 1970s, Fagundes-<br />

Neto et al 46 and Albert et al 47 showed a raised incidence<br />

of SBBO in children with acute diarrhea. A<br />

later study by Penny et al 48 showed that SBBO was<br />

more frequent in children with persistent diarrhea<br />

(80%) than in acute diarrhea (40%). In a study<br />

carried out in Cuba, Cristia et al 49 observed a<br />

similar frequency of SBBO in unweaned infants<br />

who had been hospitalized with acute and<br />

persistent diarrhea. From a functional point of<br />

view, Coello Ramirez and Lifschitz 50 found a<br />

correlation between SBBO and malabsorption of<br />

carbohydrates.<br />

Studies in Brazil 31,32,42 and in other countries 48<br />

have shown an association between SBBO and<br />

severe acute diarrhea or persistent diarrhea caused<br />

by classic enteropathogenic Escherichia coli.<br />

SBBO can thus be present in both acute and<br />

persistent diarrhea, aggravating the clinical manifestations<br />

of patients, and, in conjunction with<br />

other factors, prolonging its course. Being aware of<br />

this possibility is obviously necessary for proper<br />

and timely diagnostic and therapeutic interventions.<br />

Small-bowel bacterial overgrowth in<br />

environmental enteropathy<br />

In the 1960s, several studies in tropical countries<br />

showed that adults without clinical gastrointestinal<br />

manifestations presented histological changes<br />

in the small intestine and reduction of D-xylose<br />

absorption capacity, compared to healthy adults<br />

living in developed countries. 51,52 With regard to<br />

morphology, the abnormalities observed in the<br />

small intestine were reduced height of the intestinal<br />

villi and increased lymphoplasmocytic infiltrate<br />

in the lamina propria. The disorder was<br />

initially named tropical enteropathy. However, the<br />

possibility of a spontaneous normalization of the<br />

defect in D-xylose absorption after a change of<br />

environment, as observed in Indians and<br />

Pakistanis who moved to New York, 53 its close link<br />

with unfavorable environmental conditions, as<br />

well as the occurrence of this disorder in nontropical<br />

regions, led in the 1980s to a redefinition<br />

of this clinical condition as ‘environmental<br />

enteropathy’ by Fagundes-Neto et al. 54,55<br />

Environmental enteropathy can thus be defined as<br />

a set of unspecific morphological and functional<br />

abnormalities of the small intestine that are potentially<br />

reversible with a change in environmental<br />

conditions. From the clinical standpoint, it may be<br />

asymptomatic or associated with chronic diarrhea<br />

or with recurrent bouts of diarrhea. Stunted<br />

growth and protein–energy malnutrition often<br />

occur, as a result of the combination of malabsorption<br />

and inadequate nutrition, related to under-


privileged socioeconomic conditions. Environmental<br />

enteropathy may be associated with. SBBO.<br />

The high prevalence of SBBO in this disorder was<br />

confirmed by studies on slum-dwelling infants in<br />

the city of São Paulo, 56,57 including 40 unweaned<br />

infants without diarrhea but presenting blunted Dxylose<br />

absorption and histological abnormalities<br />

of the small intestinal mucosa. SBBO was found in<br />

61.2% of such infants. Study of the intestinal ultrastructure<br />

in these patients showed several abnormalities,<br />

such as decreased number and fusion of<br />

microvilli, cytoplasmic vacuolization and derangement<br />

of the mitochondria and endoplasmic reticulum.<br />

Taken together, these data 56,57 showed that<br />

exposure to unsuitable environmental conditions<br />

can damage the intestinal digestive–absorptive<br />

functions.<br />

Use of the hydrogen breath test with lactulose to<br />

characterize SBBO has enabled research into<br />

environmental enteropathy in larger numbers of<br />

children, leading to a social vision of the threat<br />

that SBBO represents to the infant population.<br />

Pereira et al 58 studied 340 children under 5 years<br />

of age and found SBBO in 27.2% of this population<br />

in a town in Australia.<br />

In Brazil, several studies have been performed<br />

using the hydrogen breath test with lactulose to<br />

characterize SBBO associated with environmental<br />

enteropathy. One such study 59 involved 83 schoolchildren<br />

who lived in a rural area, an urban area<br />

and a slum (favela) area of a city in the interior of<br />

São Paulo State, south-east Brazil. SBBO was<br />

detected in 7.2% of the children investigated. In<br />

the same study, the proportion of SBBO in the<br />

slum-dwelling children (18.2%) was statistically<br />

higher than that of the non-slum-dwelling<br />

children, in whom SBBO was not identified. These<br />

data thus demonstrated an association between<br />

unsuitable environmental conditions and SBBO. 39<br />

Two studies performed in 5–10-year-old children<br />

are also of interest. The first 60 was carried out in<br />

Indian children living in a reservation in Mato<br />

Grosso do Sul and found SBBO in 11.5% of the 252<br />

children studied. The second 61 compared 50 slumdwelling<br />

children with 50 control children who<br />

lived in domiciles with adequate food and<br />

environmental conditions and came from families<br />

with a solid socioeconomic background. SBBO<br />

was investigated by means of the hydrogen breath<br />

Treatment 209<br />

test with lactulose (10g) on one day and then with<br />

glucose (50g) on the next. After excluding nonhydrogen-producing<br />

children, SBBO (increase of<br />

20ppm in hydrogen concentration in the first hour<br />

after ingestion of lactulose) was observed in 50.0%<br />

(23/46) of slum-dwelling children and in 2.2%<br />

(1/46) of control children. The hydrogen test with<br />

glucose did not enable characterization of greater<br />

frequency of SBBO in slum-dwelling children.<br />

Figure 13.3 displays the curves based on the<br />

average concentrations of hydrogen in exhaled air,<br />

showing that production of hydrogen in slumdwelling<br />

children was greater than in the control<br />

group, indirectly indicating a greater quantity of<br />

lactulose-fermenting bacteria. On the other hand,<br />

the glucose test did not draw a distinction between<br />

the groups. The average (±SD) of the z-scores of<br />

weight for age (-0.45±0.95) and height for age<br />

(-0.66±1.05) of the slum-dwelling children were<br />

lower than those of the control group (+0.39 ±0.97<br />

and +0.18±0.84, respectively); the difference was<br />

statistically significant and showed an association<br />

between SBBO and protein–energy malnutrition. 61<br />

The evidence presented shows that unfavorable<br />

environmental conditions with lack of basic<br />

sanitation or inadequate supply of treated water<br />

can prompt the consumption of contaminated<br />

water and food that cause episodes of acute<br />

infectious diarrhea. This chronic state of environmental<br />

aggression may then cause long-lasting<br />

abnormalities of intestinal function that by inducing<br />

environmental enteropathy, ends up by<br />

negatively impacting the health of a large portion<br />

of the world population.<br />

Treatment<br />

Treatment of SBBO depends fundamentally on the<br />

patient’s general characteristics, especially on the<br />

presence of those clinical conditions listed in<br />

Table 13.3. Obviously, when possible, one should<br />

treat the predisposing conditions, improving<br />

factors to control bacterial flora.<br />

In patients with clinical conditions associated<br />

with SBBO in whom treatment of the underlying<br />

disease does not always produce satisfactory<br />

results, such as in intestinal pseudo-obstruction<br />

syndrome and ileocecal valve resection, use of<br />

antibiotics is indicated.


210<br />

Small-bowel bacterial overgrowth<br />

Hydrogen (ppm)<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

Figure 13.3 Median of the concentration of hydrogen in the expired air in the children living in a slum (n = 50) and<br />

controls (n = 50) after the administration of 10 g of lactulose and 50 g of glucose on different days.<br />

Boissieu et al 41 observed the disappearance of<br />

symptoms attributed to SBBO, such as chronic<br />

abdominal pain and chronic diarrhea, after antibiotic<br />

therapy.<br />

Lichtman 6 reviewed the literature in 2000 on the<br />

use of antibiotics in the treatment of SBBO. Most<br />

reports dealt with small groups of patients, with<br />

different underlying diseases predisposing<br />

towards SBBO; there were no controlled studies or<br />

series with a sufficient number of patients.<br />

Therefore, based on his own experience and in<br />

view of the scanty evidence from the literature,<br />

this author recommended the use of antibiotics<br />

effective against bacteroids, such as metronidazole,<br />

chloramphenicol and tetracycline. In children,<br />

an initial course of metronidazole for 2–4<br />

weeks is considered the first choice.<br />

In our practice, we normally prescribe courses of<br />

metronidazole and trimethoprim–sulfamethoxazole.<br />

In the case of SBBO associated with severe acute<br />

diarrhea and with persistent diarrhea, a doubleblind<br />

placebo-controlled study was carried out by<br />

our group to assess the effect of oral polymyxin for<br />

7 days on the clinical course and on the proximal<br />

0 20 40 60 80 100 120 140 160 180 200<br />

Time (min)<br />

lactulose (slum children)<br />

lactulose (controls)<br />

glucose (controls)<br />

glucose (slum children)<br />

small-intestine fluid culture in 25 hospitalized<br />

infants. 32 Both groups were on the same basic and<br />

dietary treatments; pre-treatment rates of SBBO<br />

were 61.5% in the polymyxin group and 71.4% in<br />

the placebo group. Both groups had a satisfactory<br />

clinical course. SBBO, however, persisted after<br />

treatment in a high proportion of patients: 76.9%<br />

of those given polymyxin and 57.1% on the<br />

placebo (NS). However, in the group treated with<br />

polymyxin a reduced need for other antibiotics for<br />

suspected systemic infections was found (p<br />

= 0.08).<br />

Recently, growing attention has been given to the<br />

possible use of probiotics in a variety of gastrointestinal<br />

disorders, including SBBO, and some<br />

preliminary evidence of possible efficacy is<br />

beginning to emerge. In fact, the risk of bacterial<br />

translocation in experimental short-bowel syndrome,<br />

a condition characterized by frequent<br />

episodes of SBBO, has been found to be reduced<br />

by the administration of Bifidobacterium lactis in<br />

rats. 62 Furthermore, two strains of lactobacilli<br />

(Lactobacillus casei and L. acidophilus strains<br />

cerela) have been found useful in the treatment of<br />

SBBO-related chronic diarrhea. 63


When SBBO is associated with environmental<br />

enteropathy, there is no evidence that antibiotics<br />

can help control it; in addition, the chronic nature<br />

of the process with possible recurrences clearly<br />

discourages this type of treatment. However,<br />

environmental enteropathy shows spontaneous<br />

REFERENCES<br />

1. Ellis H, Smith ADM. The blind loop syndrome. Monogr<br />

Surg Sci 1977; 4: 193–197.<br />

2. Gorbach SL, Tabaqchali S. Bacteria, bile and small<br />

bowel. Gut 1969; 10: 963–972.<br />

3. Ament MF, Shimoda SS, Sanders DP. Phatogenesis of<br />

steatorrhea in three cases of small intestinal stasis<br />

syndrome. Gastroenterology 1972; 63: 728.<br />

4. Gracey M. The contaminated small bowel symdrome:<br />

pathogenesis, diagnosis, and treatment. Am J Clin Nutr<br />

1979; 32: 234–243.<br />

5. Fagundes Neto U. Enteropatia Ambiental uma<br />

Conseqüência do Fracasso das Políticas de Saúde<br />

Pública. Rio de Janeiro: Livraria e Editora Revinter Ltda;<br />

1996.<br />

6. Lichtman SN. Bacterial overgrowth. In Walker WA,<br />

Durie PT, Hamilton JR et al. Pediatric Gastrointestinal<br />

Disease, 3rd edn. Ontario: BC Decker, 2000: 569–581.<br />

7. Araya M, Figueroa G. Flora residente intestinal.<br />

Funciones fisiológicas y alterações. Rev Chil Pediatr<br />

1985; 56: 490–496.<br />

8. Morais TB, Morais MB, Sigulem DM. Bacterial<br />

contamination of the lacteal contents of feeding bottles in<br />

metropolitan. Bull World Health Organ 1998; 76: 173–181.<br />

9. Dixon JM. The fate of bacteria in the small intestine. J<br />

Path Bact 1960; 79: 131–139.<br />

10. Ament EM, Vargas J. Diagnóstico e tratamento da<br />

síndrome da pseudo-obstrução intestinal crônica na<br />

criança. In Fagundes Neto U, Wehba J, Pena FJ, eds.<br />

Gastrenterologia Pediátrica, 2nd edn. Rio de Janeiro:<br />

MEDSI, 1991: 349–368.<br />

11. King CE, Tokes PP. Small intestine bacterial overgrowth.<br />

Gastroenterology 1979; 76: 1035–1055.<br />

12. Simon GL, Gorbach SL. Intestinal flora in health and<br />

disease. Gastroenterology 1984; 86: 174–193.<br />

13. Kocoshis AS, Scheletewitz K, Lovelace G, Laine AR.<br />

Duodenal bile acids among children: keto derivatives<br />

and aerobic small bowel bacterial overgrowth. J Pediatr<br />

Gastroenterol Nutr 1987; 6: 686–696.<br />

14. Dickman MD, Chappelka AR, Schaelder RW. The<br />

microbial ecology of the upper small bowel. AJG 1976;<br />

65: 57–62.<br />

15. Thadepalli H, Ann Lou SM, Bach VT et al. Microflora of<br />

the human small intestine. A J Surge 1979; 138:<br />

845–850.<br />

16. Fagundes Neto U, Reis MHL, Webha J et al. Small bowel<br />

bacterial flora in normal and in children with acute<br />

diarrhea. Arq Gastroenterol São Paulo 1980; 17:<br />

103–108.<br />

17. Drasar BS, Shiner M. Studies on the intestinal flora:<br />

part II. Bacterial flora of the small intestine in patients<br />

with gastrointestinal disorders. Gut 1969; 10: 812–819.<br />

References 211<br />

regression once appropriate environmental conditions<br />

are restored. It is obvious, therefore, that the<br />

mainstay for the approach to this socially relevant<br />

problem has to be through creating suitable living<br />

conditions for such a wide proportion of the<br />

world’s children.<br />

18. Isaacs PET, Kim YS. The contaminated small bowel<br />

syndrome. Am J Med 1979; 67: 1049–1057.<br />

19. Davidson GP, Butler RN. Breath analysis. In Walker WA,<br />

Durie PD, Hamilton JR et al., eds. Pediatric<br />

Gastrointestinal Disease, 3rd edn. Ontario: BC Decker,<br />

2000: 1529–1537.<br />

20. Kirsch M. Bacterial overgrowth. Am J Clin Nutr 1990;<br />

85: 231–237.<br />

21. Suarez L, Perdomo M, Escobar H. Microflora bacteriana<br />

y ecosistema intestinal isiopatologia del intestino<br />

delgado contaminado. Diarréia aguda. Meio ambiente en<br />

Espana. GEN 1994; 48: 61–64.<br />

22. Riordan SM, Mciver CJ, Walker BM et al. The lactulose<br />

breath hydrogen test and small intestinal bacterial overgrowth.<br />

Am J Gastroenterol 1996; 91: 1795–1803.<br />

23. Bouhnik Y, Alain S, Attar A et al. Bacterial populations<br />

contaminating the upper gut in patients with small<br />

intestinal bacterial overgrowth syndrome. Am J<br />

Gastroenterol 1999; 94: 1328–1331.<br />

24. Attar A, Flourié B, Rambaud JC et al. Antibiotic efficacy<br />

in small intestinal bacterial overgrowth-related chronic<br />

diarrhea: a crossover, randomized trial. Gastroenterology<br />

1999; 117: 794–797.<br />

25. Bardhan PK, Feger A, Kogon M et al. Urinary choloyl-<br />

PABA excretion in diagnosing small intestinal bacterial<br />

overgrowth. Dig Dis Sci 2000; 45: 474–479.<br />

26. King CE, Toskes PP. Comparison of the 1-Gram [ 14 C]<br />

xylose, 10-Gram lactulose-H2, and 80-Gram glucose-H2<br />

breath test in patients with small intestine bacterial<br />

overgrowth. Gastroenterology 1986; 91: 1447–1451.<br />

27. Khin-Maung U, Tin-Ay, Ku-Tin M et al. In vitro<br />

hydrogen production by enteric bacteria cultured from<br />

children with small bowel bacterial overgrowth. J<br />

Pediatr Gastroenterol Nutr 1992; 14: 192–197.<br />

28. Rumessen JJ, Gudmand-Hoyer E, Bachmann E, Justesen<br />

T. Diagnosis of bacterial overgrowth of small intestine:<br />

comparison of the 14 C D-xylose breath test and jejunal<br />

cultures in 60 patients. Scand J Gastroenterol 1985; 20:<br />

1267–1275.<br />

29. Farfán GF, Augusto CY, Raúl RL, Tello RC.<br />

Sobrepoblacion bacteriana del intestino delgado y<br />

diarrea cronica: estudo clínico y bacteriológico de 40<br />

casos. Diagnostico 1991; 28: 41–47.<br />

30. Challacombe DN, Richardson MJ, Andersoon CM.<br />

Bacterial microflora of the upper gastrointestinal tract in<br />

infants without diarrhoea. Arch Dis Child 1974; 49:<br />

264–269.<br />

31. Cruz AS, Fagundes Neto U. Influência da Escherichia<br />

coli enteropatogênica clássica sobre a proliferação bacteriana<br />

no intestino delgado na diarréia aguda e persistente<br />

do lactente. Rev Ass Med Brasil 1996; 42: 89–94.


212<br />

Small-bowel bacterial overgrowth<br />

32. Tahan S. O efeito de um antimicrobiano na microbiota<br />

duodenal e na evolução clínica de lactentes<br />

hospitalizados por diarréia aguda e persistente: um<br />

ensaio clínico duplo-cego randomizado. Thesis,<br />

Universidade Federal de São Paulo, 2000.<br />

33. Silva NS. Cultura de bactérias aeróbias e anaeróbias e<br />

teste do hidrogênio no ar expirado no diagnóstico de<br />

sobrecrescimento bacteriano no intestino delgado.<br />

Thesis, Universidade Federal de São Paulo, 2001.<br />

34. Levitt MD, Bond JH. Volume, composition, and source<br />

of intestinal gas. Gastroenterology 1970; 59: 921–929.<br />

35. Corazza G, Menozzi GM, Strocchi A et al. The diagnosis<br />

of small bowel bacterial overgrowth. Gastroenterology<br />

1990; 98: 302–309.<br />

36. Perman JA. Clinical application of breath hydrogen<br />

measurements. Can J Physiol Phamacol 1991; 69:<br />

111–115.<br />

37. Kerlin P, Wong L. Breath hydrogen testing in bacterial<br />

overgrowth of the small intestine. Gastroenterology<br />

1988; 95: 982–988.<br />

38. Riordan SM, Mciver CJ, Bolin TD, Duncombe VM.<br />

Fasting breath hydrogen concentrations in gastric and<br />

small-intestinal bacterial overgrowth. Scand J<br />

Gastroenterol 1995; 30: 252–257.<br />

39. MacMahon M, Gibbons N, Mullins E et al. Are<br />

hydrogen breath tests valid in the elderly? Gerontology<br />

1996; 42: 40–45.<br />

40. Davidson GP, Robb TA, Kirubakaran CP. Bacterial<br />

contamination of the small intestine as an important<br />

cause of chronic diarrhea and abdominal pain:<br />

diagnosis by breath hydrogen test. Pediatrics 1984; 74:<br />

229–235.<br />

41. Boissieu D, Chaussain M, Badoual J et al. Small-bowel<br />

bacterial overgrowth in children with chronic diarrhea,<br />

abdominal pain, or both. J Pediatr 1996; 128: 203–207.<br />

42. Marcelino RT. Teste do hidrogênio no ar expirado no diagnóstico<br />

do sobrecrescimento bacteriano do intestino<br />

delgado. Thesis, Universidade Federal de São Paulo, 1995.<br />

43. Guno MJV, Nolasco ET, Rogacion JM et al. Small bowel<br />

bacterial overgrowth in severely malnourished filipino<br />

children using breath hydrogen tests. J Pediatr<br />

Gastrentrol Nutr 2000; 31(Suppl 2): 240.<br />

44. World Health Organization. Persistent diarrhoea in<br />

children in developing countries. Report of a WHO<br />

Meeting. Bull World Health Organ 1988; 66: 709–717.<br />

45. World Health Organization. Evalution of an algorithm<br />

for the treatment of persistent diarrhea: a multicentre<br />

study. Bull World Health Organ 1996; 74: 479–489.<br />

46. Fagundes Neto U, Toccalino H, Dujovney F. Stool<br />

bacterial aerobic overgrowth in the small intestine of<br />

children with acute diarrhoea. Acta Paediatr Scand<br />

1976; 65: 609–615.<br />

47. Albert MJ, Bhat P, Rojand D et al. Jejunal microbial flora<br />

of Southern India infants in health and with acute<br />

gastroenteritis. J Med Microbiol 1978; 11: 43–44.<br />

48. Penny ME, Silva DGH, Mcneish AS. Bacterial<br />

contamination of the small intestine of infants with<br />

enteropathogenic Escherichia coli and other enteric<br />

infections: a factor in the aetiology of persistent<br />

diarrhoea? Br Med J 1986; 292: 1223–1225.<br />

49. Cristia YG, Arbelo TF, Vivanca MVP et al. Estudos de la<br />

microflora intestinal in niños com diarreia aguda y<br />

persistent. GEN 1994; 48: 236–244.<br />

50. Coello-Ramirez P, Lifschitz F. Enteric microflora and<br />

carbohydrate intolerance in infants with diarrhea.<br />

Pediatrics 1972; 49: 233–242.<br />

51. Sprinz H, Sribhibhadh R, Gangarosa et al. Biopsy of<br />

small bowel of Thai people. Am J Clin Pathol 1962; 38:<br />

43–51.<br />

52. Klipstein FA. Recent advances in tropical<br />

malabsorption. Scand J Gastroenterol Suppl 1970; 6:<br />

93–114.<br />

53. Gerson CD, Kent TH, Saha JR et al. Recovery of smallintestinal<br />

structure and function after residence in the<br />

tropics. II. Studies in Indians and Pakistanis living in<br />

New York City. Ann Intern Med 1971; 75: 41–48.<br />

54. Fagundes-Neto U, Viaro T, Wehba J et al. Enteropatia<br />

tropical: na infância: uma síndrome decorrente da<br />

contaminação ambiental (enteropatia ambiental). J<br />

Pediatr (Rio de J) 1983; 54: 313–319.<br />

55. Fagundes-Neto U, Viaro T, Wehba J et al. Tropical<br />

enteropathy (environmental enteropathy) in early<br />

childhood: a syndrome caused by contaminated<br />

environment. J Trop Pediatr 1984; 30: 204–209.<br />

56. Fagundes Neto U, Martins MCV, Lima FMLS et al.<br />

Symptomatic environmental enteropathy in infancy:<br />

bacterial proliferation, functional and morphological<br />

alterations of the small bowel. Rev Hosp Säo Paulo Esc<br />

Paul Med 1992; 4: 64–70.<br />

57. Gusmão RHP, Martins MCV, Gusmão SRB, Fagundes<br />

Neto U. Enteropatia ambiental: estudo ultra-estrutural<br />

da mucosa jejunal de criancas assintomaticas<br />

[Ambiental intestinal diseases: ultrastructure of the<br />

jejunum mucosa of asymptomatic children]. J Pediatr<br />

(Rio de J) 1993; 69: 21–26.<br />

58. Pereira SP, Khin-Maung U, Bolin TD et al. A pattern of<br />

breath hydrogen excretion suggesting small bowel<br />

bacterial overgrowth in Burmese village children. J<br />

Pediatr Gastroenterol Nutr 1991; 13: 32–38.<br />

59. Reis JC, Morais MB, Fagundes-Neto U. Teste do H2 no ar<br />

expirado na avaliação de absorção de lactose e<br />

sobrecrescimento bacteriano no intestino delgado de<br />

escolares. Arq Gastroenterol 1999; 36: 169–176.<br />

60. Alves GMS, Morais MB, Fagundes-Neto U. Estado<br />

nutricional e teste do hidrogênio no ar expirado com<br />

lactose e lactulose em crianças indígenas terenas. J<br />

Pediatr (Rio de J) 2002; 78: 113–119.<br />

61. Reis JC. Utilização da glicose e lactulose no diagnóstico<br />

de sobrecrescimento bacteriano no intestino delgado por<br />

meio do teste do hidrogênio no ar expirado. Thesis,<br />

Universidade Federal de São Paulo, 2002.<br />

62. Eizaguirre I, Urkia NG, Asensio AB et al. Probiotic<br />

supplementation reduces the risk of bacterial<br />

translocation in experimental short bowel syndrome. J<br />

Pediatr Surg 2002; 37: 699–702.<br />

63. Gaon D, Garmendia C, Murrielo NO et al. Effect of<br />

Lactobacillus strains (L. casei and L. acidophillus strains<br />

cerela) on bacterial overgrowth-related chronic diarrhea.<br />

Med (Buenos Aires) 2002; 62: 159–163.


14<br />

Introduction<br />

Functional abdominal pain<br />

and other functional bowel<br />

disorders<br />

Miguel Saps and Carlo Di Lorenzo<br />

The classic definition of recurrent abdominal pain<br />

has been based on the work of Apley and Naish, 1<br />

who described children who presented with intermittent<br />

episodes of abdominal pain occurring for<br />

at least 3 months without any identifiable cause<br />

and interfering with daily activities. The term<br />

‘chronic abdominal pain’ is often used interchangeably<br />

with ‘recurrent abdominal pain’.<br />

Chronic abdominal pain is abdominal pain that is<br />

continuous, persistent, or intermittent over a<br />

period of a few months. The pain may wax and<br />

wane, with some days being better than others. 2<br />

On occasions, relatively short asymptomatic<br />

periods may be interposed with ‘painful periods’,<br />

but the episodes of wellness rarely last long, generating<br />

a condition that profoundly distresses the<br />

daily life of children and their families. It is important<br />

to emphasize that recurrent abdominal pain<br />

represents a description and not a diagnosis. Many<br />

conditions can cause abdominal pain that is recurrent,<br />

but in clinical practice most children and<br />

adolescents presenting with this symptom have a<br />

functional disorder without any evidence of<br />

organic disease. They are considered to have ‘functional<br />

abdominal pain’. In this chapter we discuss<br />

conditions associated with functional alterations<br />

of the gastrointestinal (GI) system, called functional<br />

bowel disorders, and we only briefly<br />

describe those organic diseases causing abdominal<br />

pain (in the section dedicated to the differential<br />

diagnosis).<br />

Epidemiology<br />

Chronic abdominal pain is one of the most<br />

common pediatric complaints, 3 accounting for<br />

2–4% all of pediatric office visits. 4 Symptoms<br />

consistent with irritable bowel syndrome (IBS),<br />

one of the most common functional bowel disorders,<br />

occur in 14% of all high-school students and<br />

6% of all middle-school students. 5 IBS is more<br />

prevalent than other medical conditions such as<br />

hypertension, asthma and diabetes that tend to<br />

receive greater medical attention. 6 As many<br />

patients with IBS do not seek medical attention,<br />

assessing the prevalence of IBS is often difficult.<br />

Current estimates suggest that only one in four<br />

patients with symptoms of IBS seek medical care<br />

for their symptoms. 7 Population-based studies of<br />

adults have shown IBS-type symptoms in 13–15%<br />

of adult females, with abdominal pain reported as<br />

the most common complaint. 8 Reports from Kenya,<br />

Poland, Russia, India and Pakistan reveal that,<br />

although the relative frequency of an organic etiology<br />

for chronic pain may differ in various regions<br />

of the world, functional abdominal pain is probably<br />

a universal problem. 9–12 A Malaysian study in<br />

1500 schoolchildren found evidence of chronic<br />

abdominal pain in 10% of children, concluding<br />

that, in spite of differences in diet, customs and<br />

culture, the overall prevalence of this entity was<br />

similar across regions. 13 A Danish study also<br />

demonstrated that 15% of children aged 9–12<br />

years had recurrent abdominal pain. 14<br />

Quality of life in patients with IBS is substantially<br />

poorer than in the general population or in those<br />

suffering from asthma or migraine. 15 Costs related<br />

to functional bowel disorders are also enormous,<br />

with direct and indirect medical expenses associated<br />

with IBS being estimated as up to US$ 30<br />

billion a year, comparable to those of asthma,<br />

stroke, hypertensive disease, migraine and arthritis.<br />

16–18 IBS also has a high impact on a patient’s<br />

productivity, as a result of missed working days. 19<br />

A recent survey of adult patients suffering from<br />

213


214<br />

Functional abdominal pain and other functional bowel disorders<br />

IBS showed that 15% believed that IBS would stop<br />

them from finding a long-term partner, 16% had<br />

turned down a date in fear of the embarrassment<br />

that may result from intimacy, and 17% stated that<br />

they had a relationship ending as a consequence of<br />

IBS. 20 The survey also revealed that 12% of<br />

respondents had to give up working, while 35% of<br />

employed respondents reported having lost work<br />

time due to IBS. In addition to the direct and indirect<br />

costs, IBS patients are affected by intangible<br />

costs and social stigma. In 14% of cases, IBS symptoms<br />

were not accepted as a valid reason for<br />

absence by their employers. One-third of the<br />

patients with IBS had refrained from applying for<br />

promotions for positions involving multiple meetings<br />

and presentations. 20<br />

Physiology of the gastrointestinal<br />

pain response<br />

Visceral pain perception involves a complex<br />

pathway of peripheral and central nervous structures<br />

that encode, relay and modify the afferent<br />

stimulus.<br />

The enteric nervous system (ENS) is organized<br />

into two major plexuses providing the intrinsic<br />

innervation of the gut. The plexuses and the<br />

nerves connecting them constitute a continuous<br />

network around the circumference of the GI tract<br />

and along its length. The myenteric plexus, also<br />

known as Auerbach’s plexus, is situated between<br />

the external longitudinal and internal circular<br />

muscle layers, and the submucosal plexus<br />

(Meissner’s plexus) lies between the circular<br />

muscle layer and the mucosa. The myenteric<br />

plexus is larger and projects fibers primarily to the<br />

smooth muscle of the gut controlling motility.<br />

Meanwhile, the submucosal plexus projects into<br />

the mucosa and submucosa and includes more<br />

sensory cells and the neurons that control gland<br />

secretion. Although the two intestinal plexuses are<br />

separated spatially, interconnections bind the two<br />

networks into a functionally unified nervous<br />

system. The characteristics of this mesh of sensory<br />

fibers, interneurons and motor neurons, enables<br />

this mini-brain or ‘gut brain’ to integrate the<br />

sensory information, organize the motor and secretory<br />

responses and influence the luminal absorption,<br />

producing a functional state that is adapted to<br />

the well-being of the individual. Although the ENS<br />

receives input from the central and autonomic<br />

nervous systems, it can function independently.<br />

The ENS performs most of its functions in the<br />

absence of central nervous system (CNS) control,<br />

locally integrating the information of intrinsic<br />

afferent fibers (for example, luminal distension<br />

and chemical stimuli) with efferent axons, resulting<br />

in motor reflexes or secretory or absorptive<br />

responses.<br />

The extrinsic nervous system consists of afferent<br />

and efferent fibers connecting the ENS with the<br />

CNS. This communication allows the CNS continuously<br />

to integrate the information from the<br />

gastrointestinal tract with incoming information<br />

from other organs and from the environment, in<br />

order to initiate an adequate response. Under<br />

physiological conditions, most of these processes<br />

do not reach the level of conscious perception. 21<br />

However, sensations that trigger a particular<br />

behavior, including hunger, satiety and need to<br />

defecate, reach the cortex. The constant influence<br />

of the CNS on the ENS through activation of a<br />

subset of vagal and sacral parasympathetic fibers<br />

is exemplified by the relation between psychological<br />

stress and gastrointestinal response, manifested<br />

clinically by the occurrence of vomiting or<br />

diarrhea in patients experiencing a stressful event.<br />

The ENS and the brain use multiple neurotransmitters<br />

for chemical signaling and exchanging of<br />

inhibitory or excitatory information. They include<br />

excitatory neurotransmitters such as acetylcholine<br />

and substance P, and gut inhibitory neurotransmitters<br />

such as nitric oxide, ATP, vasoactive intestinal<br />

peptide (VIP), cholecystokinin, enkephalins, calcitonin<br />

gene-related peptide (CGRP), norepinephrine<br />

(noradrenaline), epinephrine (adrenaline) and<br />

others. Other neurotransmitters such as serotonin<br />

(5-hydroxytryptamine; 5-HT) and histamine have<br />

more complex effects. Ninety-five per cent of the<br />

total body 5-HT lies within the gut. Of the total gut<br />

5-HT, 90% is found in the granules of the enteroendocrine<br />

cells, and 10% in the neurons of the myenteric<br />

plexus. 22,23 5-HT plays a role in regulating GI<br />

motility and intestinal secretion. 22 5-HT receptors<br />

appear to participate in mucosal sensory processing<br />

within the gut. Distension and stroking of<br />

mechanosensitive receptors in the enteroendocrine<br />

cells triggers the release of 5-HT. 24 There<br />

are at least seven main classes of 5-HT receptor


and 22 subclasses that can be differentiated on the<br />

basis of structure and function. Four classes have<br />

been reported in the human GI tract (5-HT1, 5-HT2,<br />

5-HT3 and 5-HT4). 25 Serotonin action is complex,<br />

with mixed effects ranging from smooth muscle<br />

contraction via cholinergic nerves, to relaxation<br />

through stimulation of inhibitory nitric oxidereleasing<br />

neurons. Higher levels of serotonin are<br />

present in diarrhea-predominant IBS. 26 The ENS<br />

has the ability to modulate signal transduction by<br />

enhancing or inhibiting the activation of nociceptors<br />

through alteration in smooth muscle tone and<br />

contractile activity. Visceral pain may be modulated<br />

also at the CNS level by emotional or cognitive<br />

factors, providing a rationale for the use of<br />

centrally acting agents or cognitive behavioral<br />

treatments in functional bowel disorders.<br />

Neuroimaging studies have provided information<br />

on differences in brain processing of visceral<br />

stimuli between normal individuals and those<br />

suffering from IBS, revealing an increased activity<br />

at the level of the anterior cingulated cortex,<br />

prefrontal cortex, insular cortex and thalamus (the<br />

areas associated with emotional responses) in<br />

patients with IBS compared to asymptomatic individuals.<br />

27–29<br />

Pathophysiology<br />

Several hypotheses have been put forward to<br />

explain the cause of functional recurrent abdominal<br />

pain. We examine them in the following<br />

sections.<br />

Visceral hyperalgesia<br />

The visceral hyperalgesia hypothesis proposes that<br />

greater sensitivity of visceral afferent pathways or<br />

central amplification of visceral input lead to an<br />

enhanced perception of visceral stimuli. There is<br />

evidence that the pain and discomfort of IBS might<br />

be due to hyperalgesia and allodynia of the gut.<br />

While in hyperalgesia a painful stimulus is<br />

perceived as even more painful, in allodynia a<br />

non-painful stimulus becomes painful. 30 A<br />

noxious stimulus applied to a particular area of the<br />

gut may sensitize primary afferent fibers and nociceptors<br />

of adjacent areas, causing painful sensations<br />

with a low-intensity stimulus, resulting in<br />

primary hyperalgesia.<br />

Pathophysiology 215<br />

Most IBS patients experience rectal discomfort at<br />

lower intraluminal volumes or pressures 31,32 and<br />

have diminished tolerance to intestinal gas. 33<br />

Trimble et al 31 found that patients presenting with<br />

one functional bowel disorder frequently had<br />

additional symptoms referable to other parts of the<br />

digestive system, suggesting that enhanced<br />

visceral nociception may be a pan-intestinal<br />

phenomenon. For example, it has been reported<br />

that in addition to the features of rectal hyperalgesia,<br />

IBS patients have a decreased sensory threshold<br />

to balloon distension of the esophagus.<br />

Children with functional abdominal pain exhibited<br />

generalized visceral hyperalgesia, whereas<br />

IBS patients had rectal but not gastric hyperalgesia.<br />

32 Different GI symptoms were reproduced by<br />

stimulation of the predominant site of hyperalgesia,<br />

providing a physiological explanation of symptoms<br />

in children who have distinct phenotypic<br />

presentations.<br />

Dysmotility<br />

In addition to a greater intestinal sensitivity,<br />

patients with functional bowel disorders may<br />

display abnormal motility. Various types of motor<br />

disturbances have been documented in IBS, apparently<br />

reflecting dysfunction at one or more levels<br />

of the brain–gut axis. 34 Although the pathophysiology<br />

of IBS is commonly attributed to dysfunction<br />

of the large intestine, evidence exists to incriminate<br />

the small bowel as well. 35 Postprandial motor<br />

dysfunction in the small bowel appears to be more<br />

prevalent among IBS patients who exhibit underlying<br />

visceral hypersensitivity in the fasting state.<br />

Abdominal cramping has been associated with the<br />

passage of high-amplitude contractions through<br />

the ileocecal region. 36 Bloating has been explained<br />

by an abnormal transit and pooling of gas in<br />

conjunction with gut hypersensitivity. 33<br />

Manometric studies have demonstrated postprandial<br />

antral hypomotility in children and adults<br />

with functional dyspepsia. 37 However, not all<br />

studies have demonstrated differences between<br />

patients and control subjects. 38,39 Motility changes<br />

in IBS are neither specific nor predictable and do<br />

not serve as a diagnostic marker or as an aid to the<br />

selection of treatment. 40 It has been suggested<br />

than, rather than having a persistent motility<br />

abnormality, patients with functional bowel disorders<br />

exhibit an abnormal motor response to a<br />

variety of physiological stimuli. 41


216<br />

Functional abdominal pain and other functional bowel disorders<br />

Brain–gut interaction<br />

The shortcomings of isolated experimental or<br />

observational models in explaining the complex<br />

nature of functional bowel disorders have led<br />

research to focus on the alterations in the communications<br />

between the CNS and the GI tract, hence<br />

the term ‘brain–gut’ interaction. 6,42 There are<br />

multiple examples of brain–gut interaction, the<br />

most common being the subjects who, under<br />

emotionally stressful situations, develop diarrhea,<br />

nausea or vomiting. Anger and aggression increase<br />

colonic motility, while hopelessness results in<br />

decreased motility. 43 The brain–gut model links<br />

alterations in peripheral sensory afferent communication<br />

from the gut (e.g. visceral hyperalgesia) to<br />

CNS processing of the sensory stimuli and its efferent<br />

signaling to the gut. In IBS patients multiple<br />

studies have shown that both gut and brain show<br />

an exaggerated responsiveness to different stimuli.<br />

Patients with IBS have significantly greater electroencephalogram<br />

(EEG) abnormalities than<br />

controls. 44 Dynamic brain imaging technologies<br />

such as positron emission tomography (PET) and<br />

functional magnetic resonance imaging (fMRI)<br />

have recently been applied to the study of the<br />

gut–brain axis in order to identify the areas of the<br />

brain activated by visceral sensations. Studies<br />

with these techniques have suggested an abnormal<br />

cerebral processing of visceral stimuli in patients<br />

with functional bowel disorders. 27,28<br />

Inflammation<br />

There is evidence that IBS can occur following a<br />

gastrointestinal infection resulting in transient<br />

inflammation. Gwee et al reported that 20–25% of<br />

patients admitted to the hospital for bacterial<br />

gastroenteritis developed symptoms consistent<br />

with IBS in the first 3 months. 45 Rectal biopsies<br />

prospectively obtained during and after acute<br />

gastroenteritis from patients who developed postinfectious<br />

IBS and a control group, showed that<br />

the former group exhibited significantly greater<br />

expression of interleukin (IL)-1β mRNA. 46 A recent<br />

study examining full-thickness biopsies from the<br />

jejunum of patients with severe IBS revealed<br />

inflammation and neuronal degeneration in the<br />

myenteric plexus, suggesting a possible pathogenetic<br />

role of inflammation. 47 Animal studies also<br />

seem to indicate that inflammation may produce<br />

persistent neuromuscular gut dysfunction. 48 Mild<br />

mucosal inflammation may perturb neuromuscular<br />

function also at remote non-inflamed sites. The<br />

gut dysfunction may persist even after reduction of<br />

the mucosal inflammation. Substances that<br />

mediate these changes are not fully understood,<br />

but there is growing recognition of the role of serotonin<br />

as a sensitizing agent.<br />

Immunity<br />

The mucosal immune system mediates the clinical<br />

impact of stress and other psychological factors on<br />

the gut. Vagal afferents can be activated by products<br />

of mast cell degranulation, resulting in sensitization<br />

of silent nociceptors. Mast cell mediators<br />

may be released in response to luminal macromolecules,<br />

a phenomenon that could explain<br />

brain–immune system interactions within the gut.<br />

A descending input by the vagus nerve may also<br />

reciprocally affect mast cell degranulation, resulting<br />

in local effects on secretomotor activity.<br />

Stressors<br />

Stressful events have long been believed to be<br />

important in the development of symptoms in<br />

functional bowel disorders. 49 Physiological reactions<br />

to stressors should be considered as attempts<br />

by the body to adapt – a natural coping mechanism,<br />

in which, if stressors are not too extreme or<br />

long standing, the subject is usually successful in<br />

reaching a homeostatic state. However, at times<br />

the body loses the capacity to adapt and deleterious<br />

behavioral responses may arise. Chronic exposure<br />

to threat is associated with alterations in the<br />

autonomic outflow, resulting in activation of the<br />

hypothalamic–hypopituitary–adrenal axis with<br />

alteration in pain modulation. Corticotropin<br />

releasing hormone seems to be the hormonal mediator<br />

of the stress response. Intracerebroventricular<br />

injection of corticotropin releasing factor, which<br />

mimics the responses to stress in animals, exacerbates<br />

nociceptive responses associated with<br />

increased release of histamine. 50 In humans, possibly<br />

as a primitive response to danger, stress<br />

induces delayed gastric emptying, slower smallbowel<br />

activity and accelerated colonic transit. 51–54


These alterations may presumably cause diarrhea,<br />

constipation or abdominal distension depending<br />

on the predominant abnormality.<br />

Genetics<br />

A recent study showed a significant association<br />

between subjects with abdominal pain or bowel<br />

disturbances and first-degree relatives with IBS<br />

and dyspepsia. 55 Twin studies have shown a 17%<br />

concordance for IBS in monozygotic patients with<br />

only 8% concordance in dizygotic twins. Although<br />

these data suggest a specific role for heredity in the<br />

development of IBS, the same study showed a<br />

higher correlation of IBS with parental symptoms,<br />

suggesting that social learning from the patient’s<br />

environment has an equal or greater influence. 56<br />

Biopsychosocial model<br />

The biopsychosocial model 57 provides a framework<br />

to integrate the biological and psychosocial<br />

processes, in an attempt to understand the underlying<br />

pathophysiological mechanisms determining<br />

disease susceptibility, and to explain the clinical<br />

variability and outcome among individuals.<br />

The biopsychosocial model, proposed as an<br />

alternative to the traditional biomedical model,<br />

conceptualizes the general state of health as resulting<br />

from the integration of medical and psychosocial<br />

factors. To understand this model, one<br />

should differentiate between disease, which is the<br />

abnormality of the structure and/or function of<br />

organs and tissues (physical component), and<br />

illness, defined as the patient’s perception of<br />

health and bodily dysfunction (psychological<br />

component). In Engel’s model, 57 illness and<br />

disease result from interactions at the cellular,<br />

tissue, interpersonal and environmental levels<br />

resulting in a clinical outcome. The biopsychosocial<br />

model assumes that genetic influences on<br />

disease susceptibility and behavior result in a<br />

biological and psychosocial predisposition that<br />

will influence later psychosocial experiences,<br />

physiological functioning, or susceptibility to a<br />

pathological condition. This particular background<br />

is affected by physical and environmental<br />

exposures such as infection, food intolerance and<br />

social exposures including friends, family and<br />

community to influence the patient’s attitude<br />

Pathophysiology 217<br />

towards illness. 58 Stress acting on a vulnerable GI<br />

tract leads to an imbalance in the system, resulting<br />

in an alteration of the brain–gut axis. Multiplicity<br />

of stressors reinforces and up-regulates the<br />

response. It is well recognized that some IBS<br />

patients report initiation or exacerbation of symptoms<br />

at times of stress, trauma and major loss. 59<br />

Traumatic early life events such as child abuse may<br />

predispose to functional bowel disorders. 60 There<br />

have been reports of a greater prevalence of sexually<br />

and physically abusive experiences in individuals<br />

with IBS than in patients with organic<br />

gastrointestinal disorders and non-patient populations.<br />

61 The interaction of the previously described<br />

subsystems with psychosocial modifiers (concurrent<br />

psychiatric diagnosis, life stress, social<br />

support, coping mechanisms) affects the behavior<br />

of the individual, the biological nature of the<br />

condition and, ultimately, the clinical outcome.<br />

New or uncontrollable threatening situations may<br />

result in emotional and physiological arousal.<br />

Psychosocial factors may affect the end result<br />

(clinical presentation and outcome) acting on gut<br />

physiology, modulating symptoms experience, and<br />

influencing health behavior and therapeutic interventions<br />

(Figure 14.1). The relative contribution of<br />

the medical and psychosocial factors varies among<br />

patients. This model should be considered when<br />

Figure 14.1 Proposed pathophysiological model for<br />

functional bowel disorders.


218<br />

Functional abdominal pain and other functional bowel disorders<br />

planning therapy, as failure to link the disease and<br />

illness components will reduce the likelihood of<br />

an effective treatment.<br />

Functional bowel disorders<br />

presenting with abdominal pain<br />

Irritable bowel syndrome<br />

IBS is the most common painful functional<br />

gastrointestinal disorder in children, with symptoms<br />

arising with similar incidence in both<br />

genders. 5,62 Because there are no known biochemical<br />

or structural markers for IBS, the diagnosis is<br />

based on typical symptoms with the aid of negative<br />

results of a limited diagnostic evaluation.<br />

Despite their limited validation, the most widely<br />

accepted criteria for definition of all functional<br />

bowel disorders in children are the Rome II criteria<br />

(Table 14.1). The criteria for IBS require abdominal<br />

pain associated with bowel movements or<br />

with change in stool frequency or stool characteristics.<br />

63,64 Patients with IBS may be classified into<br />

those with a predominance of diarrhea, those who<br />

tend to have constipation, and those whose symptoms<br />

alternate from diarrhea to constipation.<br />

Functional dyspepsia<br />

According to Rome II criteria, functional dyspepsia<br />

is persistent or recurrent pain or discomfort<br />

centered in the upper abdomen. Functional<br />

dyspepsia has two presentations – ulcer-like and<br />

dysmotility-like – although considerable variation<br />

and overlap occur between the two entities.<br />

Symptoms include upper abdominal pain or<br />

discomfort, bloating, belching, early satiety,<br />

nausea, retching, or vomiting. The diagnosis is<br />

based on symptoms, as there are no biologic<br />

markers for this condition. 65 A controversial issue<br />

is the requirement for a negative esophagogastroduodenoscopy<br />

to reach the diagnosis (Table 14.1).<br />

Dyspeptic symptoms are frequent in children. An<br />

Italian school study in children 6–19 years of age<br />

revealed symptoms of dyspepsia in 45% of children.<br />

66 Although the link between <strong>Helicobacter</strong><br />

<strong>pylori</strong> infection and the development of chronic<br />

abdominal pain is controversial, there seems to be<br />

evidence of an increased risk of dyspepsia in adult<br />

patients infected with H. <strong>pylori</strong>. 67 This link has not<br />

been clearly defined in children, where it remains<br />

unknown whether H. <strong>pylori</strong>-induced gastritis<br />

(when not associated with peptic ulcer) is responsible<br />

for clinical symptoms. 68<br />

Functional abdominal pain<br />

Although the terminology seems confusing, it<br />

should be noted that the term ‘functional abdominal<br />

pain’ has been considered by the Rome II<br />

committee members as one of the categories of<br />

abdominal pain, being an entity by itself (Table<br />

14.1). This group comprises patients with pain<br />

that is usually located in the periumbilical region<br />

and is not consistently related to eating, defecation,<br />

menses, or exercise. Patients may have associated<br />

headache, dizziness, light-headedness,<br />

nausea and vomiting. As with other forms of pain<br />

of functional origin, the diagnosis is clinical.<br />

Abdominal migraine<br />

In some children with a personal and family<br />

history of migrainous headache, the abdominal<br />

pain may be acute, severe and non-colicky, and<br />

often associated with pallor and anorexia. This is<br />

sometimes referred to as abdominal migraine.<br />

Children with abdominal migraine are completely<br />

healthy between attacks, but suffer a feeling of<br />

intense misery during attacks, interrupting their<br />

activities, and seeking out a quiet, dark room. Pain<br />

is usually periumbilical, incapacitating and poorly<br />

defined. Bouts of pain can last from a few hours up<br />

to 2–3 days. Peak prevalence is at 10 years of age,<br />

declining rapidly thereafter, although occasionally<br />

it may persist into adulthood. 69<br />

In contrast to children, adults with migraine<br />

headaches usually do not have abdominal pain. 70<br />

Follow-up studies have shown an evolution of<br />

children with abdominal migraine into adults with<br />

migraine headaches, 71 and the episodes of abdominal<br />

pain have been considered a prodrome to<br />

migraine headaches. The clinical features of<br />

abdominal migraine and cyclic vomiting<br />

syndrome (recurrent, sudden, self-limiting<br />

episodes of nausea, vomiting and lethargy) show<br />

considerable similarity; treatment of the two<br />

conditions often utilizes similar pharmacological<br />

agents. 72


Functional bowel disorders presenting with abdominal pain 219<br />

Table 14.1 Rome II criteria for functional bowel disorders associated with abdominal pain or<br />

discomfort (from reference 74)<br />

Functional dyspepsia<br />

In children mature enough to provide an accurate pain history, at least 12 weeks, which need not be consecutive,<br />

within the preceding 12 months of:<br />

(1) Persistent or recurrent pain or discomfort centered in the upper abdomen (above the umbilicus); and<br />

(2) No evidence (including upper endoscopy) that organic disease is likely to explain the symptoms; and<br />

(3) No evidence that dyspepsia is exclusively relieved by defecation or associated with the onset of a change in stool<br />

frequency or stool form.<br />

Irritable bowel syndrome<br />

In children old enough to provide an accurate pain history, at least 12 weeks, which need not be consecutive, in the<br />

preceding 12 months of:<br />

(1) Abdominal discomfort or pain that has two out of three features:<br />

(a) Relieved with defecation; and/or<br />

(b) Onset associated with a change in frequency of stool; and/or<br />

(c) Onset associated with a change in form (appearance) of stool; and<br />

(2) There are no structural or metabolic abnormalities to explain the symptoms.<br />

The following symptoms also support a diagnosis of irritable bowel syndrome:<br />

(a) Abnormal stool frequency defined as more than three bowel movements per day or fewer than three bowel<br />

movements per week;<br />

(b) Abnormal stool form (lumpy/hard or loose/watery);<br />

(c) Abnormal stool passage (straining, urgency, or feeling of incomplete evacuation);<br />

(d) Passage of mucus with stool;<br />

(e) Bloating or feeling of abdominal distension.<br />

Functional abdominal pain:<br />

At least 12 weeks of:<br />

(1) Nearly continuous abdominal pain in a school-aged child or adolescent; and<br />

(2) No or only occasional relation of pain with physiological events (e.g. eating, menses, or defecation); and<br />

(3) Some loss of daily functioning; and<br />

(4) The pain is not feigned (e.g. malingering); and<br />

(5) The patient has insufficient criteria for other functional gastrointestinal disorders that would explain the<br />

abdominal pain.<br />

Abdominal migraine:<br />

In the preceding 12 months:<br />

(1) Three or more paroxysmal episodes of intense, acute midline, abdominal pain lasting 2 h to several days, with<br />

intervening symptom-free intervals lasting weeks to months; and<br />

(2) No evidence of absence of metabolic, gastrointestinal and central nervous system structural or biochemical<br />

diseases; and<br />

(3) Two of the following features:<br />

(a) Headache during episodes;<br />

(b) Photophobia during episodes;<br />

(c) Family history of migraines;<br />

(d) Headache confined to one side only;<br />

(e) An aura or warning period consisting of either visual disturbances, sensory symptoms, or motor<br />

abnormalities.


220<br />

Functional abdominal pain and other functional bowel disorders<br />

Differential diagnosis<br />

A number of clinical features (‘red flags’) are<br />

commonly considered as orienting towards an<br />

organic etiology, although definitive proof for their<br />

predictive value is scant. Elements that have been<br />

classically associated with a greater likelihood of<br />

an organic condition are listed in Table 14.2. The<br />

presence of an isolated symptom (such as isolated<br />

abdominal pain) is usually thought to be consistent<br />

with a functional disorder, while multiple<br />

symptoms (such as abdominal pain with weight<br />

loss, or vomiting, or diarrhea) are more likely to be<br />

due to an organic condition. There are almost<br />

endless causes of chronic organic abdominal pain<br />

in children. The most common are listed in Table<br />

14.3.<br />

Peptic disease<br />

Pain related to peptic ulcer disease is usually<br />

epigastric and non-radiating, but may also be<br />

generalized or periumbilical. It occasionally<br />

Table 14.2 Chronic abdominal pain ‘red<br />

flags’: signs and symptoms suggesting an<br />

organic etiology<br />

Age


Table 14.3 Causes of chronic abdominal pain<br />

Functional<br />

Irritable bowel syndrome<br />

Functional abdominal pain syndrome<br />

Dyspepsia<br />

Abdominal migraine<br />

Constipation<br />

Organic<br />

Gastrointestinal<br />

esophagitis<br />

gastritis<br />

duodenitis<br />

peptic ulcer<br />

eosinophilic gastroenteritis<br />

malrotation<br />

cysts (duplication or mesenteric)<br />

celiac disease<br />

parasites<br />

hernias<br />

tumors<br />

foreign body<br />

intussusception<br />

inflammatory bowel disease<br />

Hepatobiliary<br />

chronic hepatitis<br />

cholelithiasis<br />

cholecystitis<br />

choledochal cyst<br />

sphincter of Oddi dysfunction<br />

Pancreatic<br />

pancreatitis<br />

pseudocyst<br />

Carbohydrate intolerance<br />

In children with chronic abdominal pain,<br />

increased flatulence and bloating, the diagnosis of<br />

carbohydrate intolerance should be considered.<br />

The breath hydrogen test following a lactose or<br />

fructose load will confirm the diagnosis. Exclusion<br />

of these offending agents may improve the symptoms.<br />

On the other hand, lactose intolerance has a<br />

very high prevalence in the general population and<br />

Differential diagnosis 221<br />

Respiratory<br />

infection, tumor or inflammation vicinity of<br />

diaphragm<br />

Genital<br />

hematocolpos<br />

endometriosis<br />

mittelschmertz<br />

tumor<br />

Urinary<br />

ureteropelvic junction obstruction<br />

recurrent pyelonephritis<br />

recurrent cystitis<br />

hydronephrosis<br />

nephrolithiasis<br />

Metabolic<br />

porphyria<br />

diabetes<br />

lead poisoning<br />

Hematological<br />

angioedema<br />

collagen vascular disease<br />

sickle cell disease<br />

Musculoskeletal<br />

trauma<br />

inflammation<br />

infection<br />

tumor<br />

Psychiatric<br />

conversion reaction<br />

symptoms after a lactose load develop in only a<br />

few of the self-reported milk-intolerant subjects. 79<br />

In a large sample of American patients, lactose<br />

malabsorption was found in 21–25% of IBS<br />

patients, 81 a prevalence considered comparable to<br />

that in the general American population.<br />

Lebenthal et al 84 found a similar prevalence of<br />

lactase deficiency in children with recurrent<br />

abdominal pain and in children of a control group<br />

of similar ethnic background. Moreover, a lactose-


222<br />

Functional abdominal pain and other functional bowel disorders<br />

free elimination diet resolved symptoms in a<br />

similar percentage of patients in the lactose<br />

absorbers and in the lactose malabsorbers. As a<br />

result of the high independent prevalence of both<br />

conditions in the general population, the presence<br />

of chronic abdominal pain and lactose intolerance<br />

in one patient may be merely coincidental. Thus,<br />

the recommendation of an exclusion diet should<br />

be made with reasonable expectations, as it may be<br />

helpful in the resolution of symptoms in only a<br />

limited number of patients.<br />

Inflammatory bowel disease<br />

Occult IBD was found in 1% of adult patients 80<br />

and in 3–4% of pediatric patients 62 evaluated for<br />

IBS. Some patients may complain for months or<br />

years of vague abdominal pain and intermittent<br />

diarrhea before being diagnosed as having IBD.<br />

The presentation of the abdominal pain is variable,<br />

depending on the site of bowel involvement.<br />

Terminal ileum and cecal disease in the setting of<br />

Crohn’s disease is often associated with right lower<br />

quadrant discomfort and tenderness. Diagnosis is<br />

frequently delayed, with an average lag in diagnosis<br />

of approximately 7 months. 82 A decrease in<br />

growth preceded the onset of any symptoms by at<br />

least 1 year in 24 of 50 patients with Crohn’s<br />

disease. 83<br />

Chronic constipation<br />

IBS with constipation predominance and chronic<br />

constipation present many descriptive similarities.<br />

65 However, constipation in combination with<br />

abdominal pain has a wide differential diagnosis<br />

and constipation should not be judged as causing<br />

abdominal pain without consideration of alternative<br />

diagnoses. In chronic constipation physical<br />

examination may reveal a fecal mass in the left<br />

lower quadrant and suprapubic region. Examination<br />

of the perineum should include assessment<br />

of the lower back, sacrum and site of the<br />

anus. Anal examination may reveal the presence of<br />

a fissure or a sentinel skin tag indicative of a<br />

fissure. Rectal examination may reveal the presence<br />

of a dilated rectum containing a hard fecal<br />

mass. A flat plate radiograph may help in the diagnosis<br />

in cases where obesity precludes an appropriate<br />

abdominal examination.<br />

Gallstones<br />

Cholelithiasis is considered uncommon in infancy,<br />

childhood and adolescence, with a prevalence<br />

ranging from 0.2 to 0.5%. 84 In older children,<br />

obesity, ileal disease and a family history of childhood<br />

gallstones have been associated with<br />

cholelithiasis. 85 Children with gallstones may<br />

have colicky or unspecific pain or remain asymptomatic.<br />

The pain is mostly located in the right<br />

upper quadrant or in the epigastrium, and may<br />

radiate to the back or right shoulder. Nausea and<br />

vomiting are present in more than 50% of cases.<br />

Fatty food intolerance is not commonly reported in<br />

children. Other biliary tree pathologies such as<br />

choledochal cyst may present with abdominal<br />

pain. Presentation is age dependent, with jaundice<br />

prevailing in children and abdominal pain in<br />

adults. 86<br />

Parasitic infections<br />

Positive fecal ova and parasite tests were found in<br />

only 2% of adult patients with IBS. 80 Giardia<br />

lamblia, Dientamoeba fragilis, Cryptosporidium<br />

and Blastocystis hominis may be found in patients<br />

presenting with abdominal pain, often accompanied<br />

by anorexia, abdominal distension and<br />

diarrhea. Blastocystis hominis is most likely to be<br />

non-pathogenic in the immunocompetent human<br />

host. 87<br />

Small-bowel bacterial overgrowth<br />

Symptoms of IBS may overlap with those of<br />

small-intestinal bacterial overgrowth (SBBO).<br />

Pimentel et al, 88 in a prospective study, showed<br />

that 78% of IBS patients had SBBO diagnosed by<br />

a lactulose hydrogen breath test, and that the<br />

eradication of the overgrowth improved diarrhea<br />

and abdominal pain. A study of IBS patients with<br />

SBBO demonstrated significant motility abnormalities,<br />

89 leading the authors to conclude that<br />

dysmotility was the pathogenic mechanism<br />

linking SBBO to IBS. Another controlled study<br />

looking at the effects of antibiotics on IBS<br />

patients with alterations of the duodenal and<br />

colonic flora has also shown a significant improvement<br />

of symptoms in the group of patients<br />

receiving antibiotics. 90


Celiac disease<br />

Patients with celiac disease may present with<br />

symptoms mimicking other conditions. 91 A recent<br />

study has shown that celiac disease patients were<br />

initially diagnosed as having IBS in 37% of cases. 92<br />

In this study, only 32% of adults with celiac<br />

disease were underweight, and only 50% reported<br />

frequent diarrhea and weight loss. Anemia was<br />

present in 67% of the cases. Patients frequently<br />

presented with abdominal pain and bloating or gas<br />

which are common clinical manifestations of functional<br />

bowel disorder. The Dutch College of<br />

General Practitioners states that celiac disease<br />

should be added to the differential diagnosis of<br />

IBS. 93 A Canadian study revealed that, prior to<br />

being diagnosed as celiac disease, 37% of respondents<br />

consulted four or more family doctors. 94 An<br />

Israeli study in 270 consecutive patients who<br />

underwent endoscopy for abdominal pain demonstrated<br />

celiac disease in one out of 23 of the<br />

patients. 95<br />

Genitourinary disorders<br />

Pyelonephritis and obstructive conditions such as<br />

ureteral or pelviureteric junction obstruction may<br />

present with recurrent cramping abdominal pain,<br />

despite normal physical examination and urinalysis.<br />

96 Hematuria can be present in the setting of<br />

urinary tract infections, abuse, trauma, Henoch–<br />

Schönlein purpura, or renal stones. Dysuria associated<br />

with abdominal pain can represent a sign of<br />

pyelonephritis, abuse, trauma, or a sexually transmitted<br />

disease. 97 In adolescent girls, a history of<br />

mid-lower abdominal pain was found to have low<br />

sensitivity but high specificity for gynecological<br />

diseases. 98 Gynecological pathology such as<br />

ovarian cysts, congenital uterine abnormalities<br />

and endometriosis should also be considered in<br />

the differential diagnosis of abdominal pain.<br />

Hematocolpos 99 due to imperforate hymen may<br />

present with periodic lower abdominal pain and<br />

urinary retention. Endometriosis may begin 3–4<br />

years after menarche. Clinically it may manifest as<br />

cyclic abdominal pain, nausea, vomiting, constipation<br />

or diarrhea. 100<br />

Congenital anomalies<br />

Malrotation presents usually early in life, with<br />

85% of all cases of midgut volvulus occurring in<br />

Differential diagnosis 223<br />

the first year. In neonates, symptoms are usually<br />

dramatic with sudden onset of bilious vomiting<br />

and a visibly seriously ill patient. Older infants<br />

may present with episodes of colicky abdominal<br />

pain. In one series, 20% of cases of malrotation in<br />

patients over 1 year of age presented with chronic<br />

abdominal pain. 101 These patients often have<br />

vague, long-standing abdominal complaints with<br />

or without emesis. The pain is often postprandial<br />

and may be accompanied by bilious emesis and<br />

diarrhea or evidence of malabsorption or proteinlosing<br />

enteropathy associated with bacterial overgrowth.<br />

Duplications of the alimentary tract are<br />

uncommon congenital abnormalities. The clinical<br />

presentations may be vague and diverse, depending<br />

on the location of the duplication. 102<br />

Presenting signs and symptoms include abdominal<br />

mass, vomiting, decreased oral intake, gastrointestinal<br />

bleeding, periumbilical tenderness and<br />

abdominal distension.<br />

Musculoskeletal pain<br />

Pain related to trauma is usually well localized and<br />

sharp in nature, and may be exacerbated by movement.<br />

Patients usually are able to recall a history of<br />

trauma or strain, but occasionally that history<br />

cannot be elicited. The diagnosis of costochondritis<br />

should be considered in adolescent patients<br />

complaining of chest or upper abdominal pain.<br />

Costochondritis pain originates in the anterior<br />

chest wall, from where it may radiate into the<br />

chest, back, or abdomen. Pain is reproducible by<br />

palpating the affected costal cartilage. 103<br />

Miscellaneous<br />

Familial Mediterranean Fever (FMF) is a hereditary<br />

disease with an autosomal recessive transmission<br />

that primarily affects populations of non-<br />

Ashkenazi Jews, Armenians, Arabs and Turks. 104 A<br />

typical attack consists of fever and inflammation<br />

of serous membranes lasting from 1 to 4 days.<br />

Between attacks, FMF patients are asymptomatic<br />

and appear healthy. The frequency of the attacks<br />

varies from weekly to one every 3–4 months or<br />

less. Severe abdominal pain similar to acute peritonitis<br />

is present in 95% of patients. Other symptoms<br />

may include chest pain, arthritis and rash.<br />

Attacks may be triggered by physical and<br />

emotional stress, menstruation and a high-fat diet.<br />

There are no specific laboratory tests for FMF,


224<br />

Functional abdominal pain and other functional bowel disorders<br />

although a genetic diagnosis might become available<br />

in the foreseeable future. During attacks,<br />

acute-phase reactants may be elevated.<br />

Diagnostic testing<br />

In patients with no alarm symptoms, the Rome<br />

criteria have a positive predictive value of approximately<br />

98%, with additional diagnostic tests<br />

providing a yield of 2% or less. 105 When needed,<br />

the exclusion of an organic condition can be<br />

accomplished by utilizing inexpensive, non-invasive<br />

and easily available diagnostic tests such as<br />

complete blood cell count, erythrocyte sedimentation<br />

rate, chemistry panel, liver and thyroid function<br />

studies, urine analysis and stool examination<br />

for blood, ova and parasites. Need for other diagnostic<br />

tests should be based on history and physical<br />

examination findings. The physician should<br />

avoid the lure of having to ‘rule out’ an organic<br />

disease at all cost. Performing multiple tests may<br />

provide results that often are unrelated to the<br />

presenting symptom or have no clinical relevance<br />

(such as a mildly elevated sedimentation rate).<br />

Repeating tests to confirm the serendipitous findings<br />

may further increase anxiety and undermine<br />

the clinical diagnosis of functional bowel disorder.<br />

One could use time as the physician’s ally, assuring<br />

the patient that no test is necessary at this<br />

point but if further symptoms present or the<br />

current symptom worsens the physician will not<br />

hesitate to proceed with further work-up.<br />

Blood and stool studies<br />

Hamm et al 80 studied 1452 patients with an established<br />

diagnosis of IBS and found that screening<br />

tests showed a low incidence of thyroid dysfunction,<br />

ova and parasite infestation, or colonic<br />

pathology. The authors concluded that limited<br />

detection rates, added costs and the inconvenience<br />

of these tests made the routine use of endoscopy,<br />

radiography, thyroid function tests, fecal ova and<br />

parasite determination and the lactose hydrogen<br />

breath test questionable in the diagnostic evaluation<br />

of established IBS patients. In accordance<br />

with these results, Tolliver et al 106 performed fecal<br />

ova and parasite determinations in 196 patients<br />

with a possible diagnosis of IBS, and found no<br />

evidence of infection in any of them. In the same<br />

study, complete blood cell count, sedimentation<br />

rate, serum chemistries, thyroid profile and urinalysis<br />

were normal or yielded no useful clinical<br />

information.<br />

A study designed to investigate the prevalence of<br />

elevated antiendomysial antibody titers in children<br />

with recurrent abdominal pain compared<br />

with healthy children found no association<br />

between abdominal pain and celiac disease. 107 The<br />

study showed that 1% of patients in each group<br />

had positive celiac disease antibodies. An adult<br />

investigation studied serum antibody testing for<br />

celiac disease in patients with IBS symptoms and<br />

a control group, followed by upper endoscopy in<br />

positive cases. The study revealed that 4.6% of<br />

patients in the group with possible IBS had positive<br />

antibodies in comparison with 0.67% in the<br />

control group, suggesting that testing for celiac<br />

disease may be one of the few cost-effective evaluations<br />

in patients with IBS. 108<br />

Endoscopic studies<br />

A study investigating the presence of gastroesophageal<br />

reflux in children with recurrent<br />

abdominal pain concluded that pathological<br />

gastroesophageal reflux is a frequent finding in<br />

such children. 109 Treatment of gastroesophageal<br />

reflux in this group of patients resulted in<br />

resolution or improvement of abdominal pain in<br />

71% of cases. Another study evaluating findings<br />

on endoscopic examinations in 62 Indonesian<br />

children with recurrent abdominal pain revealed<br />

pathological abnormalities including esophagitis,<br />

erosions and duodenitis in 50% of the patients. 110<br />

In the absence of peptic ulcers, it is unclear how<br />

much these pathological findings contribute to the<br />

patients’ symptoms. Endoscopy and biopsy<br />

performed in children evaluated for dyspepsia<br />

demonstrated that most children did not have<br />

significant mucosal disease. Inflammation without<br />

evidence of peptic ulceration was found in 38% of<br />

the patients with H. <strong>pylori</strong> being identified in only<br />

five cases. 111 Follow-up at 6 months to 2 years<br />

revealed that most subjects improved, regardless<br />

of the cause of dyspepsia.<br />

Ultrasound<br />

The diagnostic yield of a sonographic examination<br />

of the abdomen in children presenting with func-


tional bowel disorders seems to be extremely low.<br />

Yip et al, in a retrospective evaluation of 644 ultrasound<br />

studies performed in children with the diagnosis<br />

of recurrent abdominal pain found abnormalities<br />

in only ten children, concluding that only<br />

children who have abdominal pain with atypical<br />

clinical features should receive sonographic<br />

screening. 112 In another study, the evaluation of 57<br />

patients with chronic abdominal pain by abdominal<br />

and/or pelvic sonography revealed only one<br />

case of ovarian cyst that later resolved spontaneously.<br />

113 Stordal et al 114 studied 44 children<br />

with recurrent abdominal pain without finding<br />

any abnormalities on ultrasound that could be<br />

related to the symptoms.<br />

Intraesophageal pH monitoring<br />

The diagnostic yield of esophageal pH monitoring<br />

in children presenting with chronic abdominal<br />

pain is controversial. A study of 44 children<br />

presenting with recurrent abdominal pain demonstrated<br />

gastroesophageal reflux in 25% of cases. 114<br />

No studies have compared outcomes between children<br />

who had pH monitoring studies and those<br />

who did not. The inconvenience associated with<br />

this test and its cost preclude its use at least in the<br />

initial evaluation of chronic abdominal pain.<br />

Lactose hydrogen breath test<br />

This test is often used to diagnose lactose intolerance<br />

in patients with functional bowel disorders,<br />

but the cause–effect relationship between lactose<br />

intolerance and symptoms has been questioned. 81<br />

Lactose intolerance is discussed in more detail in a<br />

previous section of this chapter.<br />

Treatment<br />

There is no uniformly successful treatment or cure<br />

for functional bowel disorders. Once the diagnosis<br />

has been made, it is essential to emphasize the<br />

benign aspects of the history, physical examination<br />

and laboratory tests in order effectively to<br />

reassure the patient and the family of their significance.<br />

Initial treatment of functional pain is based<br />

on reassurance and establishing an effective<br />

physician–patient–family relationship. Alleviating<br />

Treatment 225<br />

symptoms is one of the main goals of caring for<br />

patients with functional bowel disorders, but a<br />

rational management of these disorders is often<br />

challenging, owing to the lack of objective diagnostic<br />

criteria and unclear pathogenesis. As a<br />

consequence, there are no specific, universally<br />

effective therapies. 115<br />

Reassurance<br />

It is of great importance to assure the family and<br />

the patient that the physician believes that the<br />

symptoms are ‘real’ and that an organic or progressive<br />

disease is not present. An extensive explanation<br />

of the nature of the disorder should be given,<br />

discussing the problem as a common diagnosis<br />

and not just an exclusion of an organic disease. A<br />

comprehensive but easily understandable description<br />

of the nature of this group of disorders should<br />

be attempted. Comparisons with other common<br />

and benign entities such as headaches or muscle<br />

cramps may help. The family and the patient<br />

should be encouraged to ask questions and share<br />

their concerns, which should be addressed in<br />

depth to avoid fears and misconceptions.<br />

The main goal of the therapy is to re-establish a<br />

normal daily life for the patient and the family.<br />

The family should be discouraged from reinforcing<br />

the symptoms by allowing the child to miss school<br />

and leisure activities. Patients with perceived low<br />

self-worth and academic competence may find the<br />

relief of responsibility as a benefit of the pain experience;<br />

116 meanwhile, patients with adequate<br />

perception of their self-worth may find it discouraging.<br />

Fordyce and others have suggested that<br />

positive attention from others may serve as a<br />

secondary gain, transforming the painful experience<br />

into a rewarded activity that in turn could<br />

reinforce symptoms, leading to further disability.<br />

117,118 However, negative attention to pain in<br />

children with low self-esteem has been associated<br />

with increased pain behavior, possibly by creating<br />

affective distress that may further contribute to<br />

somatic symptoms. 116 Thus, the parents’ attitude<br />

towards the pain experience should be balanced,<br />

showing support and understanding, but being<br />

aware that excessive attention to the painful experience<br />

and missing activities may allow some<br />

patients (especially those with low self-worth) to<br />

develop a sick role, perpetuating the symptoms.


226<br />

Functional abdominal pain and other functional bowel disorders<br />

Behavior alternative to assuming the sick role<br />

should be encouraged and rewarded. Patients<br />

should be encouraged to discuss perceived triggering<br />

factors. Psychosocial stressors at home or<br />

school should be addressed. At school, strict toilet<br />

times or issues relating to social embarrassment to<br />

attend the restrooms should be discussed. It is<br />

often useful to communicate with the school nurse<br />

or teacher in order to address these issues.<br />

Owing to the high index of symptomatic success<br />

with reassurance, medications are not necessary<br />

for every patient with functional abdominal pain.<br />

Drug therapy should be recommended only for<br />

patients with symptoms interfering with satisfactory<br />

quality of life.<br />

Diet<br />

A detailed dietary history may identify factors that<br />

patients may feel as aggravating or provoking the<br />

symptoms. Food intolerance was perceived as a<br />

problem by 20% in an unselected UK population<br />

who responded to a questionnaire, but with<br />

controlled challenge the prevalence was slightly<br />

higher than 1%. 119 Food-induced symptoms are<br />

common reports among IBS patients, with 20–65%<br />

attributing their symptoms to adverse food reactions.<br />

120,121 In a study of 200 IBS patients, 120 the<br />

effect of an exclusion diet was evaluated, with a<br />

symptomatic improvement in almost 50% of<br />

patients, indicating that a significant proportion of<br />

IBS patients could benefit from therapeutic dietary<br />

manipulation. However, such intervention is still<br />

controversial because the observed response rate<br />

replicates the average placebo response rate in IBS<br />

trials. 40 Within IBS patients the subgroup of<br />

patients with diarrhea-predominant symptoms<br />

seems to benefit the most by a trial of exclusion<br />

diet. Among those with abdominal pain with or<br />

without diarrhea, lactose, or excessive fructose or<br />

sorbitol intake may induce symptoms. The avoidance<br />

of gas-forming foods such as legumes,<br />

complex carbohydrates, lactose and fructose may<br />

provide symptomatic relief in some patients.<br />

High-fiber diets have long been used in adult IBS<br />

patients but the data in children are still preliminary<br />

and accomplishing a substantial increase in<br />

fiber consumption may be difficult. 122,123 As fibers<br />

decrease the whole-gut transit time, fiber-enriched<br />

diets may be more useful in the subgroup of<br />

patients with constipation. 124,125 Fiber may also<br />

decrease intraluminal pressures, reducing wall<br />

tension and pain. 126 In committed families<br />

wishing to increase dietary fibers, the change<br />

should be attempted gradually, as the excess of<br />

undigested carbohydrates in the colon results in<br />

fermentation with consequent increase of gas,<br />

aggravating IBS symptoms. 127 Frequently, nonpharmacological<br />

strategies alone fail to bring<br />

complete relief to IBS patients, necessitating<br />

pharmacotherapy (Table 14.4).<br />

Laxatives<br />

Patients with severe constipation may find relief<br />

by combining fiber with a laxative. It is our preference<br />

to use polyethylene glycol or a senna derivative,<br />

but other laxatives may be used according to<br />

the practitioner’s preference. Lactulose should be<br />

avoided, as the increase of gas production derived<br />

from its use may trigger pain.<br />

Anticholinergic and antidiarrheal medications<br />

Some patients with diarrhea seem to benefit from<br />

an antidiarrheal preparation such as loperamide or<br />

diphenoxylate. Studies in adults 6 and anecdotal<br />

experience seem to demonstrate that some patients<br />

find relief by using anticholinergics such as<br />

hyoscyamine, 128 dicyclomine or others that may<br />

modify intestinal tone and motility. These agents<br />

are best used on a sporadic basis, whenever the<br />

symptoms are present much like analgesics are<br />

used for headaches. When giving medications for<br />

pain, the high placebo response rate should be<br />

considered, as several preparations may work in<br />

the short term, only to relapse after a variable<br />

period of time. 129<br />

Tricyclic antidepressants<br />

An additional option for treating chronic abdominal<br />

pain is the use of tricyclic antidepressants<br />

(TCA). TCA are used at smaller doses<br />

(0.2–0.4 mg/kg per day, 5–50mg/day) than needed<br />

for treatment of clinical depression. The analgesic<br />

effects of TCA and other antidepressants are independent<br />

of their effects on depression, and this<br />

information should be shared with the family and<br />

the patient. The beneficial effect of the TCA starts


Table 14.4 Drugs approved for treatment of IBS and scientific<br />

evidence<br />

3–7 days after the beginning of the treatment,<br />

while it takes 2–3 weeks for the onset of the antidepressant<br />

effects. 130 Relief of chronic pain with<br />

the use of antidepressants has been documented in<br />

the absence of any measurable antidepressant<br />

response, both in depressed patients 131 and in<br />

patients without clinical depression. In addition to<br />

its action on noradrenergic and serotoninergic<br />

receptors, the TCA have antimuscarinic and antihistaminic<br />

effects. Thus, these agents are especially<br />

effective in diarrhea-predominant<br />

patients 132 and those with disturbed sleep, when<br />

slowing intestinal transit and the side-effects of<br />

sleepiness may be of therapeutic value. The<br />

medication is best administered at bedtime. Other<br />

side-effects such as undesirable weight gain and<br />

the possibility of cardiac arrhythmias, although<br />

rare at such low doses, demand caution when<br />

prescribing these drugs. Electrocardiogram (EKG)<br />

monitoring can be performed at the practitioner’s<br />

discretion. Amitriptyline, although probably more<br />

effective, has greater sedative and anticholinergic<br />

effects than imipramine. 133 It is recommended that<br />

the medication be started at low doses, increasing<br />

the dose progressively as needed to achieve a full<br />

dose in weeks. 134 Other antidepressant drugs, such<br />

as selective serotonin reuptake inhibitors (SSRIs)<br />

are also being used in the relief of chronic pain. 135<br />

Selective serotonin re-uptake inhibitors<br />

Scientific<br />

USA Canada evidence<br />

Dicyclomine yes yes<br />

Propantheline yes<br />

Hyoscymine yes yes<br />

Hyoscine/atropine yes yes<br />

Peppermint oil yes yes<br />

Tegaserod yes yes yes<br />

Alosetron yes<br />

(restricted)<br />

yes<br />

Source: physicians’ desk reference 2001 (USA)<br />

Compendium of Pharmaceuticals and Specialties (Canada)<br />

SSRIs, such as paroxetine, fluoxetine, or sertraline,<br />

also seem to have therapeutic value in relieving<br />

Treatment 227<br />

symptoms in adult patients with functional bowel<br />

disorders. 128 SSRIs have become the most<br />

frequently prescribed antidepressant medications,<br />

owing to their favorable side-effect profile. 136<br />

Despite the growing popularity of SSRIs, there are<br />

few controlled studies of their efficacy in managing<br />

chronic pain syndromes. The effects of TCAs<br />

and SSRIs in the GI tract are different, with the<br />

TCAs slowing intestinal transit and SSRIs increasing<br />

motility in the small intestine. 39,137 Thus, a<br />

patient in whom the main symptom is constipation<br />

may benefit most from an initial trial of an<br />

SSRI, whereas a patient with increased bowel<br />

frequency may benefit from an antidepressant<br />

with anticholinergic properties. Recent reviews<br />

concluded that, although SSRIs may be effective,<br />

in most circumstances TCAs should remain the<br />

first-line antidepressant agents for chronic pain. 131<br />

Serotonin receptor antagonists<br />

There has been much recent interest in clinical<br />

gastrointestinal pharmacology focused on 5-HT3<br />

and 5-HT4 receptors. Such receptors have been<br />

shown to be involved in diverse sensory and motor<br />

regulatory processes in the GI tract. The 5-HT3<br />

receptor has a role in modulating colonic motility<br />

and visceral pain, increasing the threshold for<br />

sensation and discomfort, slowing colonic transit<br />

and improving stool consistency. 138 A number of<br />

selective 5-HT3 antagonists have been developed<br />

including ondansetron, granisetron, tropisetron


228<br />

Functional abdominal pain and other functional bowel disorders<br />

renzapride and zacopride. Ondansetron was the<br />

first 5-HT3 to be evaluated for its effects on the gut.<br />

It demonstrated some benefits in diarrhea-predominant<br />

IBS, but no improvement in abdominal<br />

pain. Similarly, no reduction in pain was seen with<br />

granisetron. This modest efficacy led to the search<br />

for a 5-HT3 with greater potency. Alosetron, a<br />

newer 5-HT3 receptor antagonist, has greater<br />

potency than ondansetron, and good bioavailability.<br />

139 Treatment with alosetron has led to<br />

significant relief of abdominal pain and discomfort<br />

in women with diarrhea-predominant IBS. Though<br />

generally safe, its use has been associated with<br />

severe constipation and ischemic colitis. It is<br />

currently available in the USA as part of a limited<br />

access program.<br />

5-HT4 agonists such as tegaserod and prucalopride,<br />

have been developed for patients with IBS<br />

and constipation. Tegaserod has demonstrated efficacy<br />

in the short-term relief of abdominal pain and<br />

discomfort in adult women with constipationpredominant<br />

IBS 140 and is commercially available<br />

for this indication. Adverse events, particularly<br />

loose stools, are compatible with an exaggerated<br />

pharmacological response to tegaserod and are<br />

most common during the first 2 days of therapy.<br />

Alternative and complementary therapy<br />

Despite the interventions described above, some<br />

patients will continue to experience symptoms,<br />

suggesting that current treatments that target the<br />

predominant symptom are only partially effective,<br />

presumably because they do not resolve the underlying<br />

cause of functional bowel disorder. 6 The<br />

large number of patients in whom these therapies<br />

fail has prompted an interest in alternative therapies<br />

such as diet supplements, probiotics and<br />

ancient therapeutic modalities such as Chinese<br />

medicine.<br />

Peppermint oil (Mentha piperita), which is<br />

commonly found in many over-the-counter preparations<br />

for IBS, has long been recognized as a spasmolytic<br />

agent that relaxes GI smooth muscle,<br />

relieving pain. Placebo-controlled studies have<br />

shown an overall improvement in IBS patients<br />

who used peppermint oil. 141,142 A double-blind<br />

clinical trial in Chinese medicine demonstrated<br />

that herbal therapy was effective in the management<br />

of symptoms related to IBS. 143 Natural and<br />

herbal medications are not without adverse effects,<br />

and patients should not take these products<br />

without medical supervision. A variety of other<br />

herbal preparations have been studied with different<br />

methodologies, resulting in mixed results.<br />

More well-designed, controlled trials must be<br />

performed to identify other complementary therapies,<br />

with validation of the safety and efficacy of<br />

their use. 73<br />

Another alternative therapeutic strategy for<br />

patients with significant pain is to use hypnotherapy<br />

or psychotherapy. 144–146 Hypnotherapy<br />

has been shown to be effective in the treatment not<br />

only of gastrointestinal symptoms but also of<br />

urological, sexual and psychological symptoms<br />

that are often associated features of IBS in<br />

adults. 147 Effective psychological treatments<br />

include cognitive–behavioral interventions, dynamic<br />

or interpersonal psychotherapy and stress<br />

management. In a review of published psychological<br />

trials, Talley et al found methodological<br />

problems in all the studies, concluding that the<br />

efficacy of psychological treatment for IBS could<br />

not yet be established. 148<br />

Despite the fact that alterations of enteric flora<br />

may play a role in IBS, convincing evidence for a<br />

pathogenic role of bacterial overgrowth or for a<br />

beneficial effect of probiotic therapy is still scant.<br />

A review of the therapeutic role of probiotics<br />

concluded that further studies are needed to identify<br />

particular subgroups of patients with IBS who<br />

could benefit from their use. 149 More recently,<br />

however, a very encouraging randomized, doubleblind<br />

and placebo-controlled study in adults with<br />

diarrhea-predominant IBS showed efficacy for the<br />

probiotic preparation ‘VSL#3’. 150 These findings<br />

will of course have to be reproduced in children.<br />

In chronic cases of refractory pain, referral to<br />

specialized treatment centers for an interdisciplinary<br />

pain management approach may be the most<br />

efficient method of treating disability.<br />

Natural history<br />

Functional abdominal pain is not always a benign<br />

condition with a satisfactory outcome. Long-term<br />

psychiatric disorders have been identified in<br />

patients suffering from functional abdominal pain<br />

in childhood. 151 Children with abdominal pain do


not necessarily continue to experience physical<br />

symptoms in adulthood but may have an increased<br />

risk of adult psychiatric disorders. 152<br />

Future trends<br />

The key to revealing the mechanisms and improving<br />

therapy of functional bowel disorders lies in<br />

the collaborative efforts among basic scientists,<br />

clinical investigators, physicians and behavioralists.<br />

Progress in better understanding of the<br />

sensory mediators and the causes of visceral<br />

REFERENCES<br />

1. Apley J, Naish N. Recurrent abdominal pain: a field<br />

survey of 1000 school children. Arch Dis Child 1958; 33:<br />

165–170.<br />

2. Russell, G, Abu-Arafeh, Ishaq S et al. Abdominal<br />

migraine: evidence for existence and treatment options.<br />

Paediatric Drugs. 2002; 4: 1–8.<br />

3. Alfven G. The covariation of common psychosomatic<br />

symptoms among children from socioeconomically<br />

differing residential areas: an epidemiologic study. Acta<br />

Paediatr 1993; 82: 484–487.<br />

4. Starfield B, Hoekelman RA, McCormick M et al. Who<br />

provides health care to children and adolescents in the<br />

United States? Pediatrics 1984; 74: 991–997.<br />

5. Hyams JS, Burke G, Davis PM et al. Abdominal pain<br />

and irritable bowel syndrome in adolescents: a<br />

community-based study. J Pediatr 1996; 129: 220–226.<br />

6. Camilleri M, Choi M. Review article: irritable bowel<br />

syndrome Aliment Pharmacol Ther 1997; 11: 3–15.<br />

7. Drossman DA, Thompson WG. The irritable bowel<br />

syndrome: review and a graduated multicomponent<br />

treatment approach. Ann Intern Med 1992; 116:<br />

1009–1016.<br />

8. Heaton KW, O’Donnell LJD, Braddon FEM et al.<br />

Symptoms of irritable bowel syndrome in a British<br />

urban community: consulters and nonconsulters.<br />

Gastroenterology 1992; 102: 1962–1967.<br />

9. Cross AW. Recurrent abdominal pain and duodenal<br />

ulcers in Kenyan children. East Afr Med J 1977; 54:<br />

548–551.<br />

10. Skorochodzki J, Oldak E, Taraszkiewicz F et al.<br />

Frequency of giardiasis in children with chronic<br />

abdominal pain coming from North-East Poland. Przegl<br />

Epidemiol 1998; 52: 309–315.<br />

11. Dutta S, Mehta M, Verma IC. Recurrent abdominal pain<br />

in Indian children and its relation with school and<br />

family environment. Indian Pediatr 1999; 36: 917–920.<br />

12. Hafeez A, Ali S, Hassan M. Recurrent abdominal pain<br />

and <strong>Helicobacter</strong> <strong>pylori</strong> infection in children. J Pak Med<br />

Assoc 1999; 49: 112–114.<br />

13. Boey C, Yap S, Goh KL. The prevalence of recurrent<br />

abdominal pain in 11- to 16-year-old Malaysian schoolchildren.<br />

J Paediatr Child Health 2000; 36: 114–116.<br />

Future trends 229<br />

afferent dysfunction should lead to treatments that<br />

reduce the visceral perception or reflex motor<br />

responses that lead to symptoms. We should<br />

continue to pursue investigations looking for<br />

biological markers of this group of disorders.<br />

Education of patients and physicians on the nature<br />

and therapy of this group of conditions, in<br />

conjunction with early identification of psychosocial<br />

variables and development of better<br />

therapies, are fundamental strategies in order to<br />

reduce patient suffering and the elevated costs to<br />

society associated with functional bowel disorders.<br />

14. Lundby L, Sandbaek A, Juul S. Recurrent abdominal<br />

pain in schoolchildren 9–12 years of age. Ugeskr Laeger<br />

1990; 152: 2851–2854.<br />

15. Frank L, Kleinman L, Rentz A et al. Health-related<br />

quality of life associated with irritable bowel syndrome:<br />

comparison with other chronic diseases. Clin Ther 2002;<br />

24: 675–689.<br />

16. Drossman DA, Li Z, Andruzzi E et al. US householder<br />

survey of functional gastrointestinal disorders. Dig Dis<br />

Sci 1993; 38: 1569–1580.<br />

17. Talley NJ, Gabriel SE, Harmsen WS et al. Medical costs<br />

in community subjects with irritable bowel syndrome.<br />

Gastroenterology 1995; 109: 1736–1741.<br />

18. Martin R, Barron JJ, Zacker C. Irritable bowel syndrome:<br />

toward a cost-effective management approach. Am J<br />

Manag Care 2001; 7: S268–S275.<br />

19. Hahn BA, Yan S, Strassels S. Impact of irritable bowel<br />

syndrome on quality of life and resource use in the<br />

United States and United Kingdom. Digestion 1999; 60:<br />

77–81.<br />

20. Silk DB. Impact of irritable bowel syndrome on personal<br />

relationships and working practices. Eur J Gastroenterol<br />

Hepatol 2001; 13: 1327–1332.<br />

21. Andrews PLR. Modulation of visceral afferent activity<br />

as a therapeutic possibility for gastro-intestinal<br />

disorders. In Read NW, ed. Irritable Bowel Syndrome.<br />

London: Blackwell Scientific, 1991: 91–121.<br />

22. Talley NJ. Serotoninergic neuroenteric modulators.<br />

Lancet 2001; 358: 2061–2068.<br />

23. Spiller RC. Effects of serotonin on intestinal secretion<br />

and motility. Curr Opin Gastroenterol 2001; 17: 99–103.<br />

24. Kellum JM, Albuquerque FC, Stoner MC et al. Stroking<br />

human jejunal mucosa induces 5-HT release and Clsecretion<br />

via afferent neurons and 5-HT4 receptors. Am J<br />

Physiol 1999; 277: G515–G520.<br />

25. De Ponti F, Tonini M. Irritable bowel syndrome: new<br />

agents targeting serotonin receptor subtypes. Drugs<br />

2001; 61: 317–332.<br />

26. Bearcroft CP, Perrett D, Farthing MJG. Postprandial<br />

plasma 5-hydroxytryptamine in diarrhea predominant<br />

irritable bowel syndrome: a pilot study. Gut 1998; 42:<br />

42–46.


230<br />

Functional abdominal pain and other functional bowel disorders<br />

27. Silverman DHS, Munakata JH, Ennes H et al. Regional<br />

cerebral activity in normal and pathological perception<br />

of visceral pain. Gastroenterology 1997; 112:<br />

64–72.<br />

28. Mertz M, Morgan V, Tanner G et al. Regional cerebral<br />

activation in irritable bowel syndrome and control<br />

subjects with painful and non-painful rectal distention.<br />

Gastroenterology 2000; 118: 842–848.<br />

29. Read NW. Rectal distention: from sensation to feeling.<br />

An up-to-date evaluation of brain-imaging in IBS<br />

patients and interpretation in terms of psychophysical<br />

factors of higher brain function. Gastroenterology 2000;<br />

118: 972–974.<br />

30. Merskey H, Bogduk N. Classification of Chronic Pain,<br />

2nd edn. Seattle: IASP Press, 1994.<br />

31. Trimble KC, Farouk R, Pryde A et al. Heightened<br />

visceral sensation in functional gastrointestinal disease<br />

is not site-specific. Evidence for a generalized disorder<br />

of gut sensitivity. Dig Dis Sci 1995; 40: 1607–1613.<br />

32. Di Lorenzo C, Youssef NN, Sigurdsson L et al. Visceral<br />

hyperalgesia in children with functional abdominal<br />

pain. J Pediatr 2001; 138: 838–843.<br />

33. Serra J, Azpiroz F, Malagelada JR. Impaired transit and<br />

tolerance of intestinal gas in the irritable bowel<br />

syndrome. Gut 2001; 48: 14–19.<br />

34. Kellow JE, Phillips SF, Miller LJ et al. Dysmotility of the<br />

small intestine in irritable bowel syndrome. Gut 1988;<br />

29: 1236–1243.<br />

35. Kellow JE, Phillips SF. Altered small bowel motility in<br />

irritable bowel syndrome is correlated with symptoms.<br />

Gastroenterology 1987; 92: 1885–1893.<br />

36. Kellow JE, Delvaux M, Azpiroz F et al. Principles of<br />

applied neurogastroenterology: physiology/motilitysensation.<br />

Gut 1999; 45 (Suppl): 17–24.<br />

37. Di Lorenzo C, Hyman PE, Flores AF et al Antroduodenal<br />

manometry in children and adults with severe nonulcer<br />

dyspepsia. Scand J Gastroenterol 1994; 29:<br />

799–806.<br />

38. Latimer P, Sarna S, Campbell D et al. Colonic motor and<br />

myoelectrical activity: a comparative study of normal<br />

subjects, psychoneurotic patients, and patients with<br />

irritable bowel syndrome. Gastroenterology 1981; 80:<br />

893–901.<br />

39. Gorard DA, Libby GW, Farthing MJ. Ambulatory small<br />

intestinal motility in diarrhoea predominant irritable<br />

bowel syndrome. Gut 1994; 35: 203–210.<br />

40. Camilleri M, Heading RC, Thompson WG. Clinical<br />

perspectives, mechanisms, diagnosis and management<br />

of irritable bowel syndrome. Aliment Pharmacol Ther<br />

2002; 16: 1407–1430.<br />

41. Van Ginkel R, Voskuijl WP, Benninga MA et al.<br />

Alterations in rectal sensitivity and motility in childhood<br />

irritable bowel syndrome. Gastroenterology 2001;<br />

120: 31–38.<br />

42. Mayer EA, Gebhart GF. Basic and clinical aspects of<br />

visceral hyperalgesia. Gastroenterology 1994; 107:<br />

271–293.<br />

43. Welgan P, Meshkinpour H, Beeler M. Effect of anger on<br />

colon motor and myoelectric activity in irritable bowel<br />

syndrome. Gastroenterology 1988; 94: 1150–1156.<br />

44. Nomura T, Fukudo S, Matsuoka H et al. Abnormal electroencephalogram<br />

in irritable bowel syndrome. Scand J<br />

Gastroenterol 1999; 34: 478–484.<br />

45. Gwee KA, Graham JC, McKendrick MW et al.<br />

Psychometric scores and persistence of irritable bowel<br />

after infectious diarrhea. Lancet 1996; 20: 150–153.<br />

46. Gwee KA, Collins SM, Read NW et al. Increased rectal<br />

mucosal expression of interleukin 1beta in recently<br />

acquired post-infectious irritable bowel syndrome. Gut<br />

2003; 52: 523–526.<br />

47. Tornblom H, Lindberg G, Nyberg B et al. Full-thickness<br />

biopsy of the jejunum reveals inflammation and enteric<br />

neuropathy in irritable bowel syndrome.<br />

Gastroenterology 2002; 123: 1972–1979.<br />

48. Barbara G, Vallance BA, Collins SM et al. Persistent<br />

intestinal neuromuscular dysfunction after acute nematode<br />

infection in mice. Gastroenterology 1997; 113:<br />

1224–1232.<br />

49. Boey CC, Goh KL. Psychosocial factors and childhood<br />

recurrent abdominal pain. J Gastroenterol Hepatol 2002;<br />

17: 1250–1253.<br />

50. Gue M, Del Rio-Lacheze C, Eutamene H et al. Stressinduced<br />

visceral hypersensitivity to rectal distension in<br />

rats: role of CRF and mast cells. Neurogastroenterol<br />

Motil 1997; 9: 271–279.<br />

51. Beglinger C, Degen L. Role of thyrotrophin releasing<br />

hormone and corticotrophin releasing factor in stress<br />

related alterations of gastrointestinal motor function.<br />

Gut 2002; 51 (Suppl): 145–149.<br />

52. Whitehead WE, Crowell MD, Robinson JC et al. Effects<br />

of stressful life events on bowel symptoms: subjects<br />

with irritable bowel syndrome compared with subjects<br />

without bowel dysfunction. Gut 1992; 33: 825–30.<br />

53. Drossman DA, Sandler RS, McKee DC et al. Bowel<br />

patterns among subjects not seeking health care. Use of<br />

a questionnaire to identify a population with bowel<br />

dysfunction. Gastroenterology 1982; 83: 529–534.<br />

54. Kumar D, Wingate DL. The irritable bowel syndrome: a<br />

paroxysmal motor disorder. Lancet 1985; 2: 973–977.<br />

55. Locke GR 3rd, Zinsmeister AR, Talley NJ et al. Familial<br />

association in adults with functional gastrointestinal<br />

disorders. Mayo Clin Proc 2000; 75: 907–912.<br />

56. Levy RL, Jones KR, Whitehead WE et al. Irritable bowel<br />

syndrome in twins: heredity and social learning both<br />

contribute to etiology. Gastroenterology 2001; 121:<br />

799–804.<br />

57. Engel GL. The need for a new medical model: a challenge<br />

for biomedicine. Science 1977; 196: 129–136.<br />

58. Drossman DA. Gastrointestinal illness and the biopsychosocial<br />

model. Psychosom Med 1998; 60: 258–267.<br />

59. Leserman J, Li Z, Hu Y et al. How multiple types of<br />

stressors impact on health. Psychosom Med 1998; 60:<br />

175–181.<br />

60. Drossman DA, Talley NJ, Leserman J et al. Sexual and<br />

physical abuse and gastrointestinal illness. Ann Intern<br />

Med 1995; 123: 782–794.<br />

61. Delvaux M, Denis P, Allemand H. Sexual abuse is more<br />

frequently reported by IBS patients than by patients<br />

with organic digestive diseases or controls. Results of a<br />

multicentre inquiry, French Club of Digestive Motility.<br />

Eur J Gastroenterol Hepatol 1997; 9: 345–352.<br />

62. Hyams JS, Treem WR, Justinich CJ et al.<br />

Characterization of symptoms in children with recurrent<br />

abdominal pain: resemblance to irritable bowel<br />

syndrome. J Pediatr Gastroenterol Nutr 1995; 20:<br />

209–214.<br />

63. Drossman DA. The functional gastrointestinal disorders<br />

and the Rome II process. Gut 1999; 45: II1–II5.<br />

64. Thompson WG, Longstreth GF, Drossman DA et al.<br />

Functional bowel disorders and functional abdominal<br />

pain. Gut 1999; 45: II43–II47.<br />

65. Hyams J, Colletti R, Faure C et al. Functional gastrointestinal<br />

disorders: Working Group Report of the First<br />

World Congress of Pediatric Gastroenterology,<br />

Hepatology, and Nutrition. J Pediatr Gastroenterol Nutr<br />

2002; 35: S110–S117.<br />

66. De Giacomo C, Valdambrini V, Lizzoli F et al. A<br />

population-based survey on gastrointestinal tract<br />

symptoms and <strong>Helicobacter</strong> <strong>pylori</strong> infection in children<br />

and adolescents. <strong>Helicobacter</strong> 2002; 7: 356–363.


67. Jaakkimainen RL, Boyle E, Tudiver F. Is <strong>Helicobacter</strong><br />

<strong>pylori</strong> associated with non-ulcer dyspepsia and will<br />

eradication improve symptoms? A meta-analysis. BMJ<br />

1999; 319: 1040–1044.<br />

68. Hyams J, Hyman PE. Recurrent abdominal pain. J<br />

Pediatr 1999; 135: 401–402.<br />

69. Dignan F, Abu-Arafeh I, Russell G. The prognosis of<br />

childhood abdominal migraine. Arch Dis Child 2001; 84:<br />

415–418.<br />

70. Lanzi G, Zambrino CA, Balottin U et al. Periodic<br />

syndrome and migraine in children and adolescents.<br />

Ital J Neurol Sci 1997; 18: 283–288.<br />

71. Blau JN, MacGregor EA. Is abdominal pain a feature of<br />

adult migraine? Headache 1995; 35: 207–209.<br />

72. Abu-Arafeh I, Russell G. Cyclical vomiting syndrome in<br />

children: a population-based study. J Pediatr<br />

Gastroenterol Nutr 1995; 21: 454–458.<br />

73. Haas L, McClain C, Varilek G et al. Complementary and<br />

alternative medicine and gastrointestinal diseases. Curr<br />

Opin Gastroenterol 2000; 16: 188–196.<br />

74. Rasquin-Weber A, Hyman PE, Cucchiara S et al.<br />

Childhood functional gastrointestinal disorders. Gut<br />

1999; 45: II60–II68.<br />

75. Frank F, Stricker T, Stallmach T et al. <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection in recurrent abdominal pain. J Pediatr<br />

Gastroenterol Nutr 2000; 31: 424–427.<br />

76. Bode G, Rothenbacher D, Brenner H et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> and abdominal symptoms: a population-based<br />

study among preschool children in southern Germany.<br />

Pediatrics 1998; 101: 634–637.<br />

77. Splawski JB. <strong>Helicobacter</strong> <strong>pylori</strong> and nonulcer dyspepsia:<br />

is there a relation? J Pediatr Gastroenterol Nutr 2002;<br />

34: 274–277.<br />

78. Mergener K, Baillie J. Chronic pancreatitis. Lancet 1997;<br />

350: 1379–1385.<br />

79. Carroccio A, Montalto G, Cavera G et al. Lactose<br />

intolerance and self-reported milk intolerance:<br />

relationship with lactose maldigestion and nutrient<br />

intake. Lactase Deficiency Study Group. J Am Coll Nutr<br />

1998; 17: 631–636.<br />

80. Hamm LR, Sorrells SC, Harding JP et al. Additional<br />

investigations fail to alter the diagnosis of irritable<br />

bowel syndrome in subjects fulfilling the Rome criteria.<br />

Am J Gastroenterol 1999; 94: 1279–1282.<br />

81. Lebenthal E, Rossi TM, Nord KS et al. Recurrent<br />

abdominal pain and lactose absorption in children.<br />

Pediatrics 1981; 67: 828–832.<br />

82. Heikenen JB, Werlin SL, Brown CW et al. Presenting<br />

symptoms and diagnostic lag in children with<br />

inflammatory bowel disease. Inflamm Bowel Dis 1999;<br />

5: 158–160.<br />

83. Kanof ME, Lake AM, Bayless TM. Decreased height<br />

velocity in children and adolescents before the<br />

diagnosis of Crohn’s disease. Gastroenterology 1988; 95:<br />

1523–1527.<br />

84. Toscano E, Trivellini V, Andria G. Cholelithiasis in<br />

Down syndrome. J Arch Dis Child 2001; 85: 242–243.<br />

85. Wesdorp I, Bosman D, de Graaff A et al. Clinical<br />

presentations and predisposing factors of cholelithiasis<br />

and Sludge in Children. J Pediatr Gastroenterol Nutr<br />

2000; 31: 411–417.<br />

86. de Vries JS, de Vries S, Aronson DC et al. Choledochal<br />

cysts: age of presentation, symptoms, and late<br />

complications related to Todani’s classification. J Pediatr<br />

Surg 2002; 37: 1568–1573.<br />

87. Herwaldt BL, de Arroyave KR, Wahlquist SP et al.<br />

Multiyear prospective study of intestinal parasitism in a<br />

cohort of Peace Corps volunteers in Guatemala. J Clin<br />

Microbiol 2001; 39: 34–42.<br />

References 231<br />

88. Pimentel M, Chow EJ, Lin HC. Eradication of small<br />

intestinal bacterial overgrowth reduces symptoms of<br />

irritable bowel syndrome. Am J Gastroenterol 2000; 95:<br />

3503–3506.<br />

89. Pimentel M, Soffer EE, Chow EJ et al. Lower frequency<br />

of MMC is found in IBS subjects with abnormal lactulose<br />

breath test, suggesting bacterial overgrowth. Dig Dis<br />

Sci 2002; 47: 2639–2643.<br />

90. Yakovenko E, Grigoriev P, Tcherenchimediin S et al. Is<br />

irritable bowel syndrome (IBS) related to altered gut<br />

flora? Gut 1997; 41: 123A.<br />

91. Guandalini S. Celiac disease. School Nurse News 2003;<br />

20: 24–27.<br />

92. Zipser RD, Patel S, Yahya KZ et al. Presentations of<br />

adult celiac disease in a nationwide patient support<br />

group. Dig Dis Sci 2003; 48: 761–764.<br />

93. Kleibeuker JH. The Dutch College of General<br />

Practitioners’ ‘Irritable bowel syndrome’ standard; reaction<br />

from the field of gastroenterology. Ned Tijdschr<br />

Geneeskd 2002; 146: 790–791.<br />

94. Cranney AB, Zarkadas M, Graham ID et al. The<br />

Canadian celiac health survey – The Ottawa chapter<br />

pilot. BMC Gastroenterol 2003; 3: 8.<br />

95. Yassin K, Lachter J, Suissa A et al. Undiagnosed celiac<br />

disease in adults unmasked by endoscopy. Harefuah<br />

2003; 142: 14–16.<br />

96. Byrne WJ, Arnold WC, Stannard MW et al.<br />

Ureteropelvic junction obstruction presenting with<br />

recurrent abdominal pain: diagnosis by ultrasound.<br />

Pediatrics 1985; 76: 934–937.<br />

97. McDonald JA. Abdominal pain in the adolescent female.<br />

CPEM 2002; 3: 33–44.<br />

98. Yamamoto W. The relationship between abdominal pain<br />

regions and specific diseases: an epidemiologic<br />

approach to clinical practice. J Epidemiol 1997; 7:<br />

27–32.<br />

99. Kumar A, Mittal M, Prasad S et al. Haematocolpos – an<br />

uncommon cause of lower abdominal pain in adolescent<br />

girls. J Indian Med Assoc 2002; 100: 240–241.<br />

100. Tsenov D. Endometriosis in adolescence – characteristic<br />

features. Akush Ginekol 2000; 40: 24–26.<br />

101. Sprigland N, Brandt ML, Yazbeck S. Malrotation<br />

presenting beyond the neonatal period. J Pediatr Surg<br />

1990; 25: 1139–1142.<br />

102. Otter ML, Marks CG, Cook MG. An unusual<br />

presentation of intestinal duplication with a literature<br />

review. Dig Dis Sci 1996; 41: 627–629.<br />

103. Brown RT. Costochondritis in adolescents. J Adolesc<br />

Health Care 1981; 1: 198–201.<br />

104. Ben-Chetrit E, Levy M. Familial mediterranean fever.<br />

Lancet 1998; 351: 659–664.<br />

105. Olden KW. Diagnosis of irritable bowel syndrome.<br />

Gastroenterology 2002; 122: 1701–1141.<br />

106. Tolliver BA, Herrera JL, DiPalma JA. Evaluation of<br />

patients who meet clinical criteria for irritable bowel<br />

syndrome. Am J Gastroenterol 1994; 89: 176–178.<br />

107. Fitzpatrick KP, Sherman PM, Ipp M et al. Screening for<br />

celiac disease in children with recurrent abdominal<br />

pain. J Pediatr Gastroenterol Nutr 2001; 33: 250–252.<br />

108. Wahnschaffe U, Ullrich R, Riecken EO et al. Celiac<br />

disease-like abnormalities in a subgroup of patients<br />

with irritable bowel syndrome. Gastroenterology 2001;<br />

121: 1329–1338.<br />

109. van der Meer SB, Forget PP, Kuijten RH et al.<br />

Gastroesophageal reflux in children with recurrent<br />

abdominal pain. Acta Paediatr 1992; 81: 137–140.<br />

110. Soeparto P. Endoscopic examinations in children with<br />

recurrent abdominal pain. Paediatr Indones 1989; 29:<br />

221–227.


232<br />

Functional abdominal pain and other functional bowel disorders<br />

111. Hyams JS, Davis P, Sylvester FA et al. Dyspepsia in<br />

children and adolescents: a prospective study. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 413–418.<br />

112. Yip WC, Ho TF, Yip YY et al. Value of abdominal<br />

sonography in the assessment of children with<br />

abdominal pain. J Clin Ultrasound 1998; 26: 397–400.<br />

113. Schmidt RE, Babcock DS, Farrell MK. Use of abdominal<br />

and pelvic ultrasound in the evaluation of chronic<br />

abdominal pain. Clin Pediatr (Phila) 1993; 32: 147–150.<br />

114. Stordal K, Nygaard EA, Bentsen B. Organic<br />

abnormalities in recurrent abdominal pain in children.<br />

Acta Paediatr 2001; 90: 638–642.<br />

115. Sperling MS, McQuaid KR. Rational medical therapy of<br />

functional GI disorders. In KW Olden, ed. Handbook of<br />

Functional Gastrointestinal Disorders. New York: Marcel<br />

Dekker, 1996: 269–328.<br />

116. Walker LS, Claar RL, Garber J. Social consequences of<br />

children’s pain: when do they encourage symptom<br />

maintenance? J Pediatr Psychol 2002; 27: 689–698.<br />

117. Fordyce WE. The cognitive/behavioral perspective on<br />

clinical pain. In JD Loeser, KJ Egan, eds. Managing the<br />

Chronic Pain Patient. New York: Raven Press, 1989:<br />

51–64.<br />

118. Fishbain DA, Rosomoff HL, Cutler RB et al. Secondary<br />

gain concept: a review of the scientific evidence. Clin J<br />

Pain 1995; 11: 6–21.<br />

119. Young E, Stoneham MD, Petruckevitch A et al. A population<br />

study of food intolerance. Lancet 1994; 343:<br />

1127–1130.<br />

120. Nanda R, James R, Smith H et al. Food intolerance and<br />

the irritable bowel syndrome. Gut 1989; 30: 1099–1104.<br />

121. Dainese R, De Galliani EA, Lazzari F et al. Discrepancies<br />

between reported food intolerance and sensitization test<br />

findings in irritable bowel syndrome patients. Am J<br />

Gastroenterol 1999; 94: 1892–1897.<br />

122. Aggett PJ, Agostoni C, Axelsson I et al. Nondigestible<br />

carbohydrates in the diets of infants and young children:<br />

a commentary by the ESPGHAN committee on nutrition.<br />

J Pediatr Gastroenterol Nutr 2003; 36: 329–337.<br />

123. Saldanha LG. Fiber in the diet of US children: results of<br />

national surveys. Pediatrics 1995; 96: 994–997.<br />

124. Hillemeier C. An overview of the effects of dietary fiber<br />

on the gastrointestinal transit. Pediatrics 1995; 96:<br />

997–999.<br />

125. Roma E, Adamidis D, Nikolava R et al. Diet and chronic<br />

constipation in children: the role of fiber. J Pediatr<br />

Gastroenterol Nutr 1999; 28: 169–174.<br />

126. Malcolm A, Phillips SP, Camilleri M et al.<br />

Pharmacological modulation of rectal tone alters<br />

perception of distention in humans. Am J<br />

Gastroenterology 1997; 92: 2073–2079.<br />

127. Anderson JW, Smith BM, Gustafson NJ. Health benefits<br />

and practical aspects of high fiber diets. Am J Clin Nutr<br />

1994; 59: 1242S–1247S.<br />

128. Wald A. Irritable bowel syndrome. Curr Treat Options<br />

Gastroenterol 1999; 2: 13–19.<br />

129. Spiller RC. Problems and challenges in the design of<br />

irritable bowel syndrome clinical trials: experience from<br />

published trials. Am J Med 1999; 107: 91S–97S.<br />

130. Hameroff SR, Weiss JL, Lerman JC et al. Doxepin effects<br />

on chronic pain and depression: a controlled study. J<br />

Clin Psychiatry 1984; 45: 45–52.<br />

131. Fishbain D. Evidence-based data on pain relief with<br />

antidepressants. Ann Med 2000; 32: 305–316.<br />

132. Greenbaum DS, Mayle JE, Vanegeren LE et al. Effects of<br />

desipramine on irritable bowel syndrome compared<br />

with atropine and placebo. Dig Dis Sci 1987; 32:<br />

257–266.<br />

133. Tura B, Tura SM. The analgesic effect of tricyclic antidepressants.<br />

Brain Res 1990; 518: 19–22.<br />

134. Hyams JS, Hyman PE. Recurrent abdominal pain and<br />

the biopsychosocial model of medical practice. J Pediatr<br />

1998; 133: 473–478.<br />

135. Jung, AC, Staiger T, Sullivan M. The efficacy of selective<br />

serotonin reuptake inhibitors for the management<br />

of chronic pain. J Gen Int Med 1997; 12: 384–389.<br />

136. Olfson M, Klerman G. Trends in prescription of antidepressants<br />

by office-based psychiatrists. Am J Psychiatry<br />

1993; 150: 571–577.<br />

137. Gorard DA, Libby GW, Farthing MJ. Effect of a tricyclic<br />

antidepressant on small intestinal motility in health and<br />

diarrhea-predominant irritable bowel syndrome. Dig Dis<br />

Sci 1995; 40: 86–95.<br />

138. Camilleri M, Chey WY, Mayer EA. A randomized<br />

controlled clinical trial of the serotonin type 3 receptor<br />

antagonist alosetron in women with diarrhea-predominant<br />

irritable bowel syndrome. Arch Intern Med 2001;<br />

161: 1733–1740.<br />

139. Gunput MD. Review article: clinical pharmacology of<br />

alosetron. Aliment Pharmacol Ther 1999; 13 (Suppl 2):<br />

70–76.<br />

140. Wagstaff A, Frampton J, Croom K. Tegaserod: a review of<br />

its use in the management of irritable bowel syndrome<br />

with constipation in women. Drugs 2003; 63:<br />

1101–1120.<br />

141. Pittler MH, Ernst E. Peppermint oil for irritable bowel<br />

syndrome: a critical review and meta-analysis. Am J<br />

Gastroenterol 1998; 93: 1131–1135.<br />

142. Kline RM, Kline JJ, Di Palma J et al. Enteric-coated, pHdependent<br />

peppermint oil capsules for the treatment of<br />

irritable bowel syndrome in children. J Pediatr 2001;<br />

138: 125–128.<br />

143. Bensoussan A, Talley NJ, Hing M et al. Treatment of irritable<br />

bowel syndrome with Chinese herbal medicine: a<br />

randomized controlled trial. JAMA 1998; 280:<br />

1585–1989.<br />

144. Whorwell PJ, Prior A, Faragher EB. Controlled trial of<br />

hypnotherapy in the treatment of severe refractory irritable-bowel<br />

syndrome. Lancet 1984; 2: 1232–1234.<br />

145. Lea R, Houghton LA, Calvert EL et al. Gut-focused<br />

hypnotherapy normalizes disordered rectal sensitivity<br />

in patients with irritable bowel syndrome. Aliment<br />

Pharmacol Ther 2003; 17: 635–642.<br />

146. Whitehead WE, Crowell MD. Psychologic considerations<br />

in the irritable bowel syndrome. Gastroenterol Clin<br />

North Am 1991; 20: 249–267.<br />

147. Houghton LA, Whorwell PJ. Symptomatology, quality of<br />

life and economic features of irritable bowel syndrome<br />

– the effect of hypnotherapy. Aliment Pharmacol Ther<br />

1996; 10: 91–95.<br />

148. Talley NJ, Owen BK, Boyce P et al. Psychological treatments<br />

for irritable bowel syndrome: a critique of<br />

controlled clinical trials. Am J Gastroenterol 1996; 91:<br />

277–286.<br />

149. Gionchetti P, Rizzello F, Campieri M. Probiotics in<br />

gastroenterology. Curr Opin Gastroenterol 2002; 18:<br />

235–239.<br />

150. Kim HJ, Camiller M, McKinzie S et al. A randomized<br />

controlled trial of a probiotic, VSL#3, on gut transit and<br />

symptoms in diarrhoea-predominant irritable bowel<br />

syndrome. Aliment Pharmacol Ther 2003; 17: 895–904.<br />

151. Campo JV, Di Lorenzo C, Chiappetta L et al. Adult<br />

outcomes of pediatric recurrent abdominal pain: do<br />

they just grow out of it? Pediatrics 2001; 108: E1.<br />

152. Hotopf M, Carr S, Mayou R et al. Why do children have<br />

chronic abdominal pain, and what happens to them<br />

when they grow up? Population based cohort study. BMJ<br />

1998; 316: 1196–1200.


15<br />

Introduction<br />

Disorders of sucking and<br />

swallowing<br />

Erasmo Miele and Annamaria Staiano<br />

The development of feeding skills is an extremely<br />

complex process influenced by multiple anatomic,<br />

neurophysiological, environmental, social and<br />

cultural factors. This entire process is dynamic<br />

because of ongoing growth and development.<br />

Functional feeding skills, which depend on the<br />

integrity of anatomic structures, undergo change<br />

based on neurological maturation and experimental<br />

learning.<br />

There are a variety of neurological, neuromuscular<br />

conditions in children and in infants that can<br />

impair the physiological phases of sucking and<br />

swallowing and cause disorders of feeding and<br />

dysphagia.<br />

In recent years, there has been an increase in<br />

infant swallowing disorders as a result of<br />

improved survival rates for infants born prematurely<br />

or with life-threatening medical disorders.<br />

Disorders of feeding and swallowing in children<br />

are serious and potentially fatal problems.<br />

Aspiration due to dysphagia may lead to severe<br />

pulmonary disease, and impaired oral and pharyngeal<br />

function may rapidly result in failure to<br />

thrive. Prompt evaluation of swallowing disorders<br />

is therefore critical.<br />

The differential diagnosis of dysphagia in children<br />

is widespread. The diagnostic work-up can be<br />

extremely difficult and exhaustive in many cases.<br />

Because of this complexity, multidisciplinary team<br />

evaluations should be conducted.<br />

Successful rehabilitation of children with swallowing<br />

disorders requires knowledge of the parameters<br />

of normal and abnormal swallowing plus<br />

skill in the integration of a variety of essential therapeutic<br />

techniques.<br />

Epidemiology and etiology<br />

Data on the incidence of swallowing disorders are<br />

lacking, because in clinical practice, disorders of<br />

swallowing are often considered in the general<br />

context of a feeding disorder. Feeding is a complex<br />

process that involves a number of phases in addition<br />

to the act of swallowing, including the recognition<br />

of hunger (appetite), the acquisition of food<br />

and the ability to bring the food to the mouth. 1 The<br />

estimated prevalence of feeding problems in the<br />

pediatric population ranges from 25 to 35% in<br />

normally developing children, and from 40 to 93%<br />

in children with developmental delay. 2,3<br />

Disorders of sucking and swallowing may be<br />

caused by multiple etiological factors that may<br />

interfere with the child’s ability to coordinate<br />

swallowing and breathing maneuvers and may be<br />

manifested as a unique set of symptoms. Potential<br />

causes are in three broad categories: immaturity,<br />

delay, or a defect in neuromuscular control; an<br />

anatomic abnormality of the aerodigestive tract;<br />

and/or systemic illness. The magnitude of the<br />

dysfunction depends on the balance between the<br />

extent of the structural or functional abnormality<br />

and the child’s compensatory adaptations. 4<br />

Disorders associated with sucking and swallowing<br />

difficulties are listed in Table 15.1. 5<br />

Pathophysiology<br />

The fetus is capable of swallowing amniotic fluid<br />

in utero, indicating that the motor program for<br />

swallowing functions before gestation is complete.<br />

However, oral feeding is not initiated in preterm<br />

infants before 32 weeks of postconceptional age,<br />

partly because the coordination of sucking,<br />

233


234<br />

Disorders of sucking and swallowing<br />

Table 15.1 Differential diagnosis in dysphagia (adapted from reference 5)<br />

Prematurity<br />

Upper airway obstruction<br />

Nasal and nasopharyngeal<br />

cohanal atresia, stenosis, septal deflections and abscess, infections, tumors, sinusitis<br />

Oropharynx<br />

defects of lips and alveolar processes, cleft lip or palate, hypopharyngeal stenosis, craniofacial syndromes or<br />

sequences (e.g. Cruzon, Treacher–Collins syndrome, Pierre Robin sequence)<br />

Laryngeal<br />

laryngeal cleft and cyst, laryngomalacia, subglottic stenosis and paralysis<br />

Congenital defects of the larynx, trachea and esophagus<br />

Laryngotracheoesophageal cleft<br />

Tracheoesophageal fistula with associated esophageal atresia<br />

Esophageal anomalies (e.g. strictures, webs)<br />

Vascular anomalies<br />

aberrant right subclavian artery<br />

double aortic arch<br />

right aortic arch with left ligamentum<br />

Acquired anatomic defects<br />

Trauma<br />

external trauma, intubation, endoscopic, foreign body<br />

Chemical ingestion<br />

Neurological disorders<br />

Central nervous system<br />

trauma<br />

hypoxia and anoxia<br />

cortical atrophy, hypoplasia, agenesis<br />

infections (meningitis, brain abscess)<br />

Peripheral nervous system disease<br />

Trauma<br />

Congenital defects<br />

Neuromuscular disorders<br />

Guillain–Barré syndrome<br />

Poliomyelitis (bulbar paralysis)<br />

Myasthenia gravis<br />

Myotonic muscular dystrophy<br />

Anatomic and functional defects<br />

Crycopharyngeal dysfunction<br />

Esophageal achalasia<br />

Esophageal spasm<br />

Paralysis of the esophagus<br />

Associated atresia-tracheoesophageal fistula, nerve defect<br />

Peptic and eosinophilic esophagitis<br />

Riley-Day syndrome (Dysautonomia)<br />

Brain stem compression (e.g. Chiari malfomation, tumor)


swallowing and respiration is not established. 6<br />

Even at 34 weeks, the minute ventilation during<br />

sucking decreases more than that of infants at<br />

36–38 weeks. Therefore, the co-ordination<br />

between swallowing and breathing is not yet fully<br />

organized at 34 weeks of postconceptional age. 7,8<br />

Anatomic structures, which are essential to<br />

competent feeding skills, undergo growth that<br />

changes their physical relationship to one another<br />

and consequently affects their function. The swallowing<br />

mechanism, by which food is transmitted<br />

to the stomach and digestive organs, is a complex<br />

action involving 26 muscles and five cranial<br />

nerves. The neurophysiological control involves<br />

sensory afferent nerve fibers, motor efferent fibers,<br />

paired brainstem swallowing centers, and suprabulbar<br />

neural input. Structural integrity is essential<br />

to the development of normal feeding and<br />

swallowing skills. 9<br />

Deglutition is generally divided into phases of<br />

swallowing, based on anatomic and functional<br />

characteristics: pre-oral, pharyngeal and<br />

esophageal. 10,11<br />

Anatomic considerations<br />

An understanding of the anatomy of the pharynx<br />

is essential to a thorough understanding of the<br />

swallowing process. The anatomy changes during<br />

Anatomic considerations 235<br />

development. The tongue, the soft palate and the<br />

arytenoid mass (arytenoid cartilage, false vocal<br />

cords and true vocal cords) are larger relative to<br />

their surrounding chambers when compared with<br />

the adult. 12 In the infant, the tongue lies entirely<br />

within the oral cavity, resulting in a small oropharynx.<br />

12,13 In addition, a sucking pad, composed of<br />

densely compacted fatty tissue that further<br />

reduces the size of the oral cavity, stabilizes the<br />

lateral walls of the oral cavity. The larynx lies high<br />

in the infant, and the tip of the epiglottis extends<br />

to and may overlap the soft palate. These anatomic<br />

relationships are ideal for the normal infant<br />

feeding pattern of sucking or suckling feeding a<br />

breast or a bottle in a recumbent position. 14 In the<br />

infant, the larynx sits high in the neck at the level<br />

of vertebrae C1 to C3, allowing for the velum,<br />

tongue and epiglottis to approximate, thereby<br />

functionally separating the respiratory and digestive<br />

tracts. This separation allows the infant to<br />

breathe and feed safely. By age of 2–3 years, the<br />

larynx descends, decreasing the separation of the<br />

swallowing and digestive tracts 2,15 (Figure 15.1).<br />

Development and normal swallowing<br />

function<br />

The newborn infant is reflex bound and automatically<br />

makes certain oral motor movements. For<br />

example, the newborn’s automatic-phasic bite<br />

Figure 15.1 (a) The adult oropharynx. The phases of swallowing are labeled. 1, preparatory phase; 2, oral phase; 3,<br />

pharyngeal phase; 4, esophageal phase. (b) The infant oropharynx. Note that the infant oral cavity providing little space for<br />

manipulation of the food bolus. The larynx is elevated so that the epiglottis almost touches the soft palate. The tongue is<br />

entirely within the oral cavity, with no oral pharynx. Reproduced with permission from reference 15.


236<br />

Disorders of sucking and swallowing<br />

pattern, although it may look like chewing, actually<br />

starts liquid flowing into the mouth with its<br />

pump-like action. The rooting reflex elicited by<br />

stroking the side of the mouth and resulting in the<br />

head turning towards the source of stimulation, is<br />

a food-seeking response. Over time, these reflexive<br />

movements of the newborn are gradually refined<br />

and incorporated into more voluntary feeding<br />

patterns. 14<br />

Therefore, by increasing the intraoral space, the<br />

infant begins to suppress reflexive suckling<br />

patterns and starts to use voluntary sucking<br />

patterns. In contrast to suckling, true sucking<br />

involves a raising and lowering of the body of the<br />

tongue with increased use of intrinsic musculature.<br />

Most infants complete the gradual transition<br />

from suckling to true sucking by 9 months of age.<br />

This is considered a critical step in the development<br />

of oral skills that will permit handling of<br />

thicker textures and spoon-feeding. 16<br />

As with sucking, chewing patterns emerge gradually<br />

during infancy. Between birth and 5 months of<br />

age, a phasic bite-release pattern develops. Jaw<br />

opening and closing begins as a reflex and evolves<br />

into a volitionally controlled bite. True chewing<br />

develops as activity of the tongue, cheeks and jaws<br />

co-ordinate to participate in the breakdown of<br />

solid food. The eruption of the deciduous teeth<br />

between the ages of 6 and 24 months provides a<br />

Table 15.2 Phases of normal deglutition<br />

Phase Activities Time<br />

chewing surface and increased sensory input to<br />

facilitate the development of chewing. 16<br />

The concept of a ‘critical period’ is relevant to<br />

feeding development. A critical period is a fairly<br />

well-delineated period of the time during which a<br />

specific stimulus must be applied in order to<br />

produce a particular action. After such a critical<br />

period a particular behavior pattern can no longer<br />

be learned. The ‘sensitive period’ is the optimal<br />

time for the application of a stimulus. After the<br />

sensitive period, it is more difficult to learn a<br />

specific pattern of behavior. 17<br />

Current knowledge of the swallowing mechanism<br />

is derived mainly from radiographic studies,<br />

which have been in use since the early 1900s.<br />

Plain films of the pharynx were replaced in the<br />

1930s by cineradiography, which, in the 1970s,<br />

was subsequently replaced by videofluoroscopy.<br />

Videofluoroscopy permits instant analysis of bolus<br />

transport, aspiration and pharyngeal function. 18<br />

Using this descriptive method deglutition can be<br />

divided into four phases: the oral preparatory<br />

phase, oral voluntary phase, pharyngeal phase,<br />

and esophageal phase (Table 15.2). 19<br />

The oral preparatory phase occurs after food is<br />

placed into the mouth. The food is prepared for<br />

pharyngeal delivery by mastication and mixing<br />

with saliva. This is a highly co-ordinated activity<br />

Pre-oral phase food introduced into the oral cavity varies; depends on substance<br />

(voluntary)<br />

Oral phase bolus formation and passage to Less than 1s<br />

(voluntary/involuntary) the pharynx<br />

Pharyngeal phase respiration ceases; pharyngeal 1s or less<br />

(involuntary) peristalsis; epiglottis closes; larynx<br />

closes, elevates and draws forward.<br />

UES relaxes<br />

Esophageal phase esophageal peristalsis; opening of 8–20s<br />

(involuntary) lower esophageal sphincter<br />

UES, upper esophageal sphincter


that is rhythmic and controlled to prevent injury to<br />

the tongue. The tongue is elevated towards the<br />

palate by the combined actions of the digastric,<br />

genioglossus, geniohyoid and mylohyoid muscles.<br />

Intrinsic tongue muscles produce both the initial<br />

depression in the dorsum that receives the food<br />

and the spreading action that distributes the food<br />

throughout the oral cavity. The buccinator muscles<br />

hold food between the teeth in dentulous infants<br />

and help to generate suction in neonates. In this<br />

phase, the soft palate is against the tongue base<br />

secondary to contraction of the palatoglossus<br />

muscles, which allows nasal breathing to<br />

continue. 2,20<br />

During, the oral propulsive phase, the bolus is<br />

propelled into the oropharynx. The oral phase is<br />

characterized by elevation of the tongue and a<br />

posterior sweeping or stripping action produced<br />

mainly by the action of styloglossus muscles. This<br />

propels the bolus into the pharynx and triggers the<br />

‘reflex swallow’. The receptors for this reflex are<br />

thought to be at the base of the anterior pillars, but<br />

there is evidence that others exist in the tongue<br />

base, epiglottis and pyriform fossae. Sensory<br />

impulses for the reflex are conducted through the<br />

afferent limbs of cranial nerves V, IX and X to the<br />

swallowing center. Oral transit time is less than<br />

1s. 2,21<br />

The pharyngeal phase of deglutition is the most<br />

complex and critical. The major component of the<br />

pharyngeal phase is the reflex swallow. This<br />

results from motor activity stimulated by cranial<br />

nerves IX and X. The reflex swallow may be triggered<br />

by a voluntary oral phase component or any<br />

stimulation of the afferent receptor in and around<br />

the anterior pillar. 2 Bolus passage through the<br />

pharynx is accompanied by soft palate elevation,<br />

lingual thrust, laryngeal elevation and descent<br />

upper esophageal sphincter (UES) relaxation and<br />

pharyngeal constrictor peristalsis. The pharyngeal<br />

phase commences as the bolus head is propelled<br />

past the tongue pillars and finishes as the bolus<br />

tail passes into the esophagus. 21 Once it begins, the<br />

pharyngeal phase is very quick, (1 s or less). 2 It is<br />

characterized biomechanically by the operation of<br />

three valves and several propulsive mechanisms.<br />

The larynx closes and the palate elevates to<br />

disconnect the respiratory tract. The UES opens to<br />

expose the esophagus. At the completion of the<br />

pharyngeal phase, the airway valves (larynx,<br />

Development and normal swallowing function 237<br />

palate) open, and the UES closes so that respiration<br />

can resume. 21<br />

Pharyngeal bolus transit occurs in two phases: an<br />

initial thrust phase and a mucosal clearance<br />

phase. 22 Bolus thrust, which propels most of the<br />

bolus into the esophagus, is provided by lingual<br />

propulsion, laryngeal elevation and gravity. The<br />

tongue has been linked to a piston, pumping the<br />

bolus though the pharynx. 23 Patients with tongue<br />

impairment cannot generate large bolus driving<br />

forces despite an intact pharyngeal constrictor<br />

mechanism. 24 Laryngeal elevation creates a negative<br />

post-crycoid pressure to suck the oncoming<br />

bolus towards the esophagus, and the elevated<br />

larynx holds the pharyngeal lumen open to minimize<br />

pharyngeal resistance. 23<br />

As the bolus enters the pharynx and is stripped<br />

inferiorly by the combined effects of gravity, the<br />

negative pressure mentioned above and the<br />

sequential contractions of the pharyngeal constrictors,<br />

the soft palate moves against the posterior<br />

pharyngeal wall to close off the nasopharyngeal<br />

port. The bolus divides around the epiglottis,<br />

combines and passes through the crycopharyngeal<br />

muscle, or upper esophageal sphincter. 2<br />

The UES is the high-pressure zone located<br />

between the pharynx and the cervical esophagus.<br />

The physiological role of this sphincter is to<br />

protect against reflux of food into the airways as<br />

well as to prevent entry of air into the digestive<br />

tract. 25 Posteriorly and laterally the cricopharyngeus<br />

muscle is a definitive component of the UES.<br />

The crycopharyngeus has many unique characteristics:<br />

it is tonically active, has a high degree of<br />

elasticity, does not develop maximal tension at<br />

basal length and is composed of a mixture of slowand<br />

fast-twitch fibers, with the former predominating.<br />

These features enable the crycopharyngeus<br />

to maintain a resting tone and yet be able to stretch<br />

open by distracting forces, such as a swallowed<br />

bolus and hyoid and laryngeal excursion. The<br />

crycopharyngeus muscle, however, constitutes<br />

only the lower one-third of the entire highpressure<br />

zone. The thyropharyngeus muscle<br />

accounts for the remaining upper two-thirds of the<br />

UES.<br />

The UES function is controlled by a variety of<br />

reflexes that involve afferent inputs to the<br />

motorneurons innervating the sphincter. 25 Based,


238<br />

Disorders of sucking and swallowing<br />

on functional studies, it is believed that the major<br />

motor nerve of the crycopharyngeus muscle is the<br />

pharyngoesophageal nerve. Vagal efferents probably<br />

reach the muscle by the pharyngeal plexus,<br />

using the pharyngeal branch of the vagus. 26 The<br />

superior laryngeal nerve may also contribute to<br />

motor control of the crycopharyngeus muscle. 1<br />

Sensory information from the UES is probably<br />

provided by the glossopharyngeal nerve and the<br />

sympathetic nervous system. There is probably<br />

little or no contribution by the sympathetic<br />

nervous system to crycopharyngeal control. 26<br />

The relaxation phase begins as the genioglossus<br />

and suspensory muscles pulls the larynx anteriorly<br />

and superiorly. The bolus is carried into the<br />

esophagus by a series of contraction waves that are<br />

a continuation of the pharyngeal stripping action. 2<br />

Proposed functions of the UES include prevention<br />

of esophageal distension during normal breathing<br />

and protection of the airway against aspiration<br />

following an episode of acid reflux. 1,26 Qualitative<br />

abnormalities of the UES have been documented<br />

in infants with reflux disease. 27<br />

The esophageal phase occurs as the bolus is<br />

pushed through the esophagus to the stomach by<br />

esophageal peristalsis. Esophageal transit time<br />

varies from 8 to 20s. 14<br />

Dysphagia<br />

Dysphagia is an impairment of swallowing involving<br />

any structures of the upper gastrointestinal<br />

tract from the lips to the lower esophageal sphincter.<br />

28 Dysphagia in children is generally classified<br />

as either oral dysphagia (abnormal preparatory or<br />

oral phase) or pharyngeal dysphagia (abnormal<br />

pharyngeal phase).<br />

Oral dysphagia is seen most commonly in children<br />

with neurodevelopmental disorders. Infants with<br />

oral dysphagia often have an impaired oral<br />

preparatory phase. These children typically<br />

demonstrate poor lingual and labial co-ordination,<br />

resulting in anterior substance loss and a poor<br />

labial seal for sucking or removing food from a<br />

spoon. Other abnormal patterns include jaw thrust<br />

and tongue thrust on presentation of food. Oral<br />

dysphagia may also involve the oral phase of swallowing.<br />

Children with impaired oral phase function<br />

often have difficulty in co-ordinating the<br />

‘suck, swallow, breathe’ pattern of early oral<br />

intake, resulting in diminished endurance during<br />

oral feeds. Apraxia of the oral swallow as well as<br />

reduction of oral sensation are also common.<br />

Other deficits include reduced bolus formation<br />

and transport, abnormal holding patterns, incomplete<br />

tongue-to-palate contact and repetitive<br />

lingual pumping. 14<br />

Oropharyngeal dysphagia results from either<br />

oropharyngeal swallowing dysfunction or<br />

perceived difficulty in the process of swallowing.<br />

Major categories of dysfunction are: an inability or<br />

excessive delay in initiation of pharyngeal swallowing;<br />

aspiration of ingestate; nasopharyngeal<br />

regurgitation; and residue of ingestate within the<br />

pharyngeal cavity after swallowing. Each of these<br />

categories of dysfunction can be mechanically<br />

subcategorized using fluoroscopic and/or manometric<br />

data. 18<br />

Clinical signs and symptoms<br />

Clinical signs and symptoms of sucking and swallowing<br />

disorders in infants and in children are<br />

listed in Table 15.3.<br />

Complications<br />

Malnutrition<br />

In the severely affected child with impaired swallowing,<br />

poor oral and/or pharyngeal function may<br />

lead to decreased energy intake as a consequence<br />

of prolonged feeding time and the inability to<br />

ingest adequate volumes, and malnutrition may<br />

result. 1 Malnutrition has many adverse effects.<br />

The most significant effects are on behavior and<br />

immune status. Malnutrition negatively influences<br />

immune status. This leads to recurrent infections<br />

that increase caloric requirements but decrease<br />

intake, leading to a worsening nutritional status. In<br />

addition, malnutrition causes behavioral apathy,<br />

weakness and anorexia, which can all profoundly<br />

affect feeding and secondarily, nutritional status.<br />

Thus, although malnutrition is often a direct result<br />

of poor feeding skills, it can also have a<br />

compounding, and even perpetuating, effect on<br />

feeding problems in children. 14


Table 15.3 Clinical signs and symptoms of dysfunctional sucking and swallowing<br />

Clinical signs<br />

Failure to thrive<br />

Meal-time distress<br />

Refusing food<br />

Nasal regurgitation<br />

Wet or hoarse voice<br />

Drooling<br />

Spitting<br />

Vomiting<br />

Gastroesophageal or pharyngeal reflux<br />

Symptoms<br />

Oral–tactile hypersensitivity<br />

Feeling of obstruction<br />

Sialorrea<br />

Sialorrea (excessive drooling) is defined as the<br />

unintentional loss of saliva and other oral contents<br />

from the mouth. Drooling usually occurs in<br />

patients with neurological disease complicated by<br />

abnormalities of the oral phase of deglutition.<br />

Clinical complications of drooling include soaking<br />

of clothes, offensive odors, macerated skin and, if<br />

‘posterior’ drooling occurs, aspiration. 29<br />

Respiratory complications<br />

Respiratory complications of swallowing disorders<br />

include apnea and bradycardia, choking episodes,<br />

chronic or recurrent pneumonia, bronchitis and<br />

atelectasis. 30<br />

Apnea and bradycardia may result from stimulation<br />

of laryngeal chemoreceptors without evidence<br />

of aspiration or as a consequence of hypoxemia.<br />

Hypoxemia may result from the effects of direct<br />

aspiration on gas exchange, from apnea triggered<br />

by laryngeal and nasopharyngeal chemoreceptors,<br />

or in patients with compromised lung function as<br />

a result of a normal decrease in minute ventilation<br />

that occurs with suckle feeding. 31–33 Symptoms<br />

such as chronic recurrent coughing, choking and<br />

postprandial congestion or wheezing generally<br />

indicate the occurrence of aspiration. Infants,<br />

Odynophagia<br />

Atypical chest pain<br />

Complications 239<br />

Respiratory manifestations<br />

Coughing<br />

Choking<br />

Stridor<br />

Change in respiration pattern after swallowing<br />

Apnea and bradycardia (predominantly in infants)<br />

Noisy breathing after feeding<br />

Chronic recurrent wheezing<br />

Chronic recurrent bronchitis, pneumonia and<br />

atelectasis<br />

especially premature infants, appear to be at<br />

increased risk of respiratory disease from dysfunctional<br />

swallowing. 4 Clinical manifestations of<br />

dysfunctional sucking and swallowing in infants<br />

are primarily apnea and bradycardia during<br />

feeding, although chronic or recurrent respiratory<br />

problems (congestion, cough, wheezing) are also<br />

seen. 30 Congested or noisy breathing during and<br />

following feeding is also a common complaint of<br />

parents of infants with dysfunctional swallowing.<br />

Dysphagia can also be an important but underrecognized<br />

cause of chronic or recurrent bronchitis,<br />

asthma and pneumonia in infants. 4<br />

Respiratory disease secondary to dysphagia in an<br />

older child is typically seen in a neurologically<br />

impaired host. 34,35 Apnea and bradycardia are<br />

uncommon in an older child. Bronchitis, pneumonia,<br />

atelectasis and recurrent wheezing are more<br />

likely to be seen in this population. Feeding and<br />

swallowing evaluation should be considered in<br />

those with central nervous system (CNS) injury<br />

affecting cranial nerve function and difficult-tocontrol<br />

chronic or recurrent bronchitis, wheezing,<br />

pneumonia, or asthma. Tracheobronchomalacia, a<br />

complication of chronic inflammation of the major<br />

airways, occurs commonly. Dysfunctional swallowing<br />

is also encountered in children with a<br />

tracheostomy. The tracheostomy may interfere<br />

with normal laryngeal function during swallowing<br />

and predispose to aspiration. 28


240<br />

Disorders of sucking and swallowing<br />

Aspiration may also occur in children with disorders<br />

of swallowing after an episode of gastroesophageal<br />

reflux; also acid reflux may result in<br />

bronchospasm, pneumonia or apnea. 36,37 The most<br />

obvious sign that a person may have aspirated is<br />

the post-swallow cough, but in the swallowingimpaired<br />

child other more insidious indicators<br />

may be present. ‘Silent aspiration’ with no clinical<br />

signs can account for over half of all cases of radiologically<br />

defined aspiration. 28,38<br />

Diagnosis<br />

Feeding disorders and dysphagia in infants and in<br />

children can be both physiological and behavioral<br />

in nature. 39 The evaluation of feeding and<br />

swallowing dysfunction is best performed as a<br />

multidisciplinary process with co-ordinated input<br />

from a variety of team members, including<br />

pediatricians, pediatric gastroenterologists, developmental<br />

pediatricians, speech–language pathologists,<br />

occupational therapists, and pediatric<br />

dietitians. 40 The goals of this evaluation include<br />

Oropharyngeal<br />

dysphagia<br />

Structural Functional<br />

Videoendoscopic<br />

swallow study<br />

Examination under<br />

anesthesia<br />

Surgery<br />

the following: ascertaining whether oropharyngeal<br />

dysphagia is likely, and identifying the etiology;<br />

identifying structural etiologies of oropharyngeal<br />

dysfunction; ascertaining the functional integrity<br />

of the oropharyngeal swallow; evaluating the risk<br />

of aspiration pneumonitis; and determining<br />

whether the pattern of dysphagia is amenable to<br />

therapy. 41 The investigation and management of<br />

swallowing disorders are summarized in the<br />

Figure 15.2.<br />

History<br />

History<br />

Physical Examination<br />

Clinical swallow<br />

examination<br />

Videofluoroscopic<br />

swallow study Videoendoscopic<br />

swallow study<br />

A comprehensive history, obtained from individuals<br />

directly involved in caring for the child (e.g.<br />

parents, feeding specialist) is essential in evaluating<br />

children with swallowing disorders. The evaluation<br />

begins with a focused feeding history,<br />

including current diet, textures, route and time of<br />

administration, modifications and feeding position.<br />

Medical co-morbidites that may affect swallowing<br />

need to be investigated.<br />

Neurological<br />

evaluation<br />

Management plan<br />

Videofluoroscopic<br />

swallow study<br />

Esophageal<br />

dysphagia<br />

Structural Functional<br />

Esophageal manometry<br />

Upper GI series<br />

Upper GI endoscopy<br />

Upper GI series<br />

Upper GI endoscopy<br />

Reduce risk Optimize hydration/nutrition<br />

Non-oral feeds Modify consistency Modify posture Supplements and dietary advice<br />

Figure 15.2 Flow chart for the investigation and management of dysphagia in children. Adapted from reference 28. GI,<br />

gastrointestinal.


The child’s caregivers should also be questioned<br />

regarding associated symptoms such as oral aversion,<br />

weak sucking, irritable behavior, gagging and<br />

choking, and disruptions in breathing or apnea.<br />

Postural or positional change during feeding may<br />

be reported in children with dysphagia.<br />

Odynophagia and emesis may be related to<br />

pharyngeal and/or esophageal disorders. A history<br />

of recurrent pneumonia may indicate chronic aspiration;<br />

a history of stridor in relation to feeding<br />

may indicate a glottic or subglottic abnormality<br />

contributing to feeding disorders. Determining<br />

whether these symptoms occur before, during or<br />

after the swallow helps localize the affected<br />

phase. 16,17<br />

In addition, nutritional and psychological assessment<br />

should be evaluated. Many patients with<br />

swallowing disorders have a concurrent illness<br />

that may increase metabolic needs. Psychological<br />

assessments help to identify behavioral and<br />

parental factors that may be contributing to a<br />

feeding disorder. Psychosomatic causes of dysphagia<br />

should be considered in adolescents with<br />

dysphagia. 2,42,43<br />

Physical and clinical evaluation<br />

The aims of physical examination in dysphagic<br />

patients are: to identify underlying systemic or<br />

metabolic disease when present; to localize the<br />

neuroanatomic level and severity of a causative<br />

neurological lesion when present; and to detect<br />

adverse sequelae such as aspiration pneumonia or<br />

nutritional deficiency. 18<br />

The physical examination views the whole child<br />

and specifically focuses on the upper aerodigestive<br />

tract, beginning with an examination for structural<br />

and functional abnormalities. Oral cavity<br />

anatomic abnormalities, such as ankyloglossia,<br />

cleft lip or palate, or macroglossia, need to be<br />

excluded. 2 The palatal gag is perhaps the most<br />

commonly assessed reflex and should be evaluated.<br />

29 A hyperactive gag can result in significant<br />

feeding difficulties; in the past an absent gag reflex<br />

was viewed as an indication to stop oral<br />

feeding. 4,34<br />

It is critical that observation of the feeding process<br />

be included. 40 This part of the examination is best<br />

performed in conjunction with a feeding and swal-<br />

Diagnosis 241<br />

lowing specialist, such as a speech–language<br />

pathologist or an occupational therapist. This<br />

examination includes assessments of posture,<br />

positioning, patient motivation, oral function, efficiency<br />

of oral intake and clinical signs of safety.<br />

During the feeding trial, the presence of abnormal<br />

movements such as jaw thrust, tongue thrust,<br />

tonic bite reflex and jaw clenching are noted. A<br />

variety of therapeutic positions, techniques and<br />

adaptive feeding utensils may be used. 1,16<br />

A variety of assessment scales may be used to<br />

detail and quantitate results of the swallowing<br />

evaluation. However, all assessments are based on<br />

similar observation of feeding structure and function.<br />

44<br />

Usually, a careful developmental, medical and<br />

feeding history provides clues to the diagnosis that<br />

guide the selection of further diagnostic tests. Only<br />

after all reasonable physical causes have been<br />

ruled out should a feeding or swallowing disorder<br />

be attributed to a purely behavioral cause. 2<br />

Diagnostic tests<br />

Radiographic assessment<br />

Videofluoroscopy represents the gold standard for<br />

evaluation of children with swallowing disorders.<br />

A videofluoroscopic swallow study is ideally<br />

performed by a consultant radiologist and specialist<br />

speech and language therapist. 45 A series of<br />

swallows of varied volumes and consistencies of<br />

contrast material are imaged in a lateral projection,<br />

and framed to include the oropharynx, palate,<br />

proximal esophagus and proximal airway. Studies<br />

are recorded on videotape to permit instant replay,<br />

in slow motion if necessary, and examination of<br />

both the presence and mechanism of the swallowing<br />

dysfunction. The videofluoroscopic study<br />

provides evidence of all four categories of oropharyngeal<br />

swallowing disorders: inability or excessive<br />

delay in initiation of pharyngeal swallowing;<br />

aspiration of ingestate; nasopharyngeal regurgitation;<br />

and residue of ingestate within the pharyngeal<br />

cavity after swallowing. Furthermore, the<br />

procedure allows for testing of the efficacy of<br />

compensatory dietary modifications, postures,<br />

swallowing maneuvers and facilatory techniques<br />

in correction of observed dysfunction. Generally,<br />

the videofluoroscopic evaluation is completed by


242<br />

Disorders of sucking and swallowing<br />

esophagography to evaluate the esophageal phase<br />

of deglutition (Figure 15.3). 18<br />

Ultrasonography<br />

Ultrasound imaging has been used to a limited<br />

extent in the assessment of oral phase dysphagia.<br />

Figure 15.3 Lateral fluoroscopic projection of an infant<br />

showing contrast material in the valleculas, pyriform<br />

sinuses, laryngeal vestibule and esophagus.<br />

Using a transducer positioned in the submental<br />

region, ultrasonography allows observation of the<br />

motion of structures in the oral cavity such as the<br />

tongue and floor of the mouth during feeding and<br />

deglutition, but lacks sensitivity in visualizing<br />

pharyngeal motion and for determining whether<br />

aspiration has occurred. Ultrasonography represents<br />

the only method of imaging that can study<br />

infants during breast feeding, and may be particularly<br />

useful in distinguishing an infant’s inability<br />

to attach from maternal factors contributing to<br />

feeding difficulties. 16 Unfortunately, laryngeal<br />

penetration and aspiration are not easily detected,<br />

because of the shadows cast by the laryngeal structures<br />

(Figure 15.4). 4,46,47<br />

Pharyngeal manometry<br />

Intraluminal manometry, performed using a<br />

transnasally positioned manometric assembly, can<br />

quantify the strength of pharyngeal contraction,<br />

the completeness of UES relaxation and the relative<br />

time of these two events. Most studies have<br />

indicated that manometry of the UES and pharynx<br />

provides useful information, primarily in patients<br />

who have symptoms of oropharyngeal dysfunction.<br />

Figure 15.4 Transverse ultrasound scan of the larynx at the level of the vocal cords. During swallowing the vocal cords<br />

(white arrow) adduct and the glottis closes (black arrow).


The co-ordination of muscle activity at various<br />

levels can be obtained by simultaneous recording<br />

of pressure in the pharynx, at the level of the<br />

crycopharyngeus, and in the esophagus.<br />

Anatomical references are not avalaible with this<br />

technique (Figure 15.5). 27,48<br />

Fiberoptic endoscopic examination<br />

Pediatric fiberoptic endoscopic examination is a<br />

relatively new diagnostic method to complement<br />

the current armamentarium of techniques for evaluating<br />

dysphagia and/or aspiration. The procedure<br />

is performed by passing a flexible laryngoscope<br />

into the oropharynx after anesthetizing the nares<br />

and nasopharynx. 49 It provides the ability to diagnose<br />

many of the laryngeal disorders that may<br />

affect the child, while at the same time evaluating<br />

the swallowing mechanism itself. The procedure<br />

involves five components: assessment of the<br />

anatomy as it affects swallowing; evaluation of<br />

movement and sensation of critical structures;<br />

assessment of secretion management; direct<br />

assessment of swallowing function for food and for<br />

liquid; and the patient’s response to therapeutic<br />

maneuvers. In experienced hands, this test can be<br />

performed in children with minimal discomfort.<br />

50,51<br />

Figure 15.5 Upper esophageal sphincter (UES) motility in<br />

a control child. The pressure is recorded in the pharynx, in<br />

the UES, and in the cervical esophagus. Note that the<br />

onset of UES relaxation precedes pharyngeal contraction,<br />

which is terminated before the return of UES pressure to<br />

resting values. UES relaxation is complete to the level of<br />

the esophageal resting pressure (dashed line). Contraction<br />

of the UES continues into the cervical esophagus as the<br />

primary wave (dots) of swallowing. From reference 27.<br />

Treatment options 243<br />

Scintigraphy<br />

Scintigraphy is a radionuclide evaluation using<br />

technetium-99m-labeled sulfur colloid mixed in<br />

the infant’s formula. It has been proposed as an<br />

alternative and perhaps more sensitive way of<br />

quantifying aspiration, transit times, gastroesophageal<br />

reflux and pharyngeal residue. Based on<br />

a case report, the radionuclide salivagram has also<br />

been used to document aspiration of saliva. The<br />

major limitations of this technique are the poor<br />

definition of the anatomy and the poor sensitivity<br />

for detecting aspiration during swallowing in<br />

known aspirators. At present, the use of this technique<br />

in pediatric patients is limited. 51,52<br />

Treatment options<br />

Optimal management strategies are critical for<br />

infants and children with feeding and swallowing<br />

problems. The management of swallowing<br />

dysfunction involves a team approach. Individuals<br />

involved in addition to the medical team include a<br />

swallowing expert (speech–language pathologist or<br />

occupational therapist), a nutritionist and the<br />

family. Since swallowing abnormalities arise from<br />

a diverse group of underlying disorders, management<br />

techniques must be individualized. This<br />

heterogeneity is also reflected in the fact that<br />

patients have different potentials for recovery. 1<br />

Although total oral feeding may not be a realistic<br />

goal, it is the universal hope of caregivers.<br />

Professionals are obliged to point out prerequisites<br />

for oral feeding and to discuss the probability that<br />

an individual child may reach the goal. These<br />

management decisions are typically made on the<br />

basis of clinical observations and assessments. In<br />

addition, important information is obtained<br />

through an instrumental assessment by videofluoroscopic<br />

swallow study. A methodical videofluoroscopic<br />

swallowing study defines the anatomy of<br />

the oropahrynx; detects dysfunction as evident by<br />

aspiration, poor clearance, or poor control of the<br />

bolus; determines the mechanism responsible for<br />

the dysfunction; and examines the short-term<br />

effects of the therapeutic strategies designed to<br />

eliminate or compensate for that dysfunction. 53<br />

Management decisions may incorporate nutritive<br />

recommendations, medical and surgical decisions,<br />

position guidelines, oral–motor swallowing practice<br />

and behavioral intervention. 54


244<br />

Disorders of sucking and swallowing<br />

The clinical and instrumental evaluation of children<br />

with sucking and swallowing disorders<br />

should allow for the recognition of treatable<br />

anatomic or inflammatory lesions.<br />

A child may refuse to eat even if his anatomic<br />

abnormality has been corrected, because of<br />

learned aversion to feeding. Behavior therapy can<br />

often overcome this type of conditioned food<br />

refusal. 2,55<br />

Various therapeutic approaches may improve the<br />

efficiency and safety of feeding. Management techniques<br />

involve devising compensatory strategies to<br />

minimize swallowing-related complications. 56<br />

These include changing the textures of foods;<br />

pacing of feeding; changing the bottle or utensils;<br />

and changing the alignment of the head, neck and<br />

body when feeding (Table 15.4). 49<br />

Frequently, children with severe anatomic disorders<br />

but normal neurological function develop<br />

their own adaptive strategies to allow for safe oral<br />

feeding. Unfortunately, many children with<br />

feeding disorders have non-correctable neurological<br />

or anatomic abnormalities that make oral<br />

feeding difficult or unsafe. Some patients cannot<br />

obtain adequate nutrition by mouth because of a<br />

risk for aspiration. Thus, supplying a portion of<br />

the patient’s nutrition by nasogastric or gastrostomy<br />

feeding may be beneficial. 2 For those children<br />

who have been intubated, management<br />

includes teaching techniques that will facilitate<br />

the transition from non-oral to oral feeding.<br />

However, there is little evidence that non-oral<br />

feeding reduces or eliminates the risk of aspiration.<br />

57–59<br />

The strongest evidence-based recommendation<br />

that can be made pertains to diet modification.<br />

Furthermore, the literature provides reasonable<br />

evidence of the plausibility of swallowing therapy<br />

but minimal evidence of efficacy. Nonetheless,<br />

although no hard evidence supports its efficacy,<br />

the available data are inconclusive and swallowing<br />

therapy has not been proved to be ineffective.<br />

Thus, the current weight of opinion, combined<br />

with the convincing demonstration of biological<br />

plausibility for specific techniques and the consistency<br />

of low-grade evidence, is the basis for recommending<br />

that swallowing therapy should be used.<br />

Large-scale randomized, controlled trials are<br />

needed to clarify the current recommendations. 18<br />

Prognosis<br />

The prognosis depends on underlying conditions<br />

that predispose to impaired sucking and swallowing.<br />

However, the early recognition of feeding<br />

problems, the diagnosis of underlying disorders<br />

and appropriate intervention improve outcomes<br />

for the child and the family.<br />

Table 15.4 Swallowing strategies for<br />

pediatric dysphagia<br />

Behavioral training<br />

Dietary modification<br />

thickened liquids<br />

thin liquids<br />

Proper intrabolus placement<br />

modification of feeding utensils and bolus<br />

presentation<br />

Swallowing exercises<br />

supraglottic swallow<br />

supersupraglottic swallow<br />

effortful swallow<br />

Mendelsohn maneuver<br />

Modification of body tone, posture, seating and<br />

positioning<br />

head tilt<br />

chin tuck<br />

head rotation<br />

lying on the side, elevation<br />

Suckle-feeding-valved feeding bottle<br />

Crycopharyngeal myotomy<br />

Facilitatory techniques<br />

biofeedback<br />

thermal stimulation<br />

gustatory stimulation<br />

Provision of alternative means of enteral nutrition<br />

nasogastric feeding<br />

gastrostomy tube (surgical or endoscopic)


REFERENCES<br />

1. Tuchman DN. Disorders of deglutition. In Walker WA,<br />

Durie PR, Walker-Smith JA, Watkins JB, eds. Pediatric<br />

Gastrointestinal Disease, 3rd edn. Hamilton, Ontario,<br />

Canada: BC Decker, 2000: 277–288.<br />

2. Rudolph CD, Link DT. Feeding disorders in infants and<br />

children. Pediatr Clin North Am 2002; 49: 97–112.<br />

3. Del Giudice E, Staiano A, Capano G et al.<br />

Gastrointestinal manifestations in children with<br />

cerebral palsy. Brain Dev 1999; 21: 307–311.<br />

4. Loughlin GM, Lefton-Greif MA. Dysfunctional<br />

swallowing and respiratory disease in children. Adv<br />

Pediatr 1994; 41: 135–162.<br />

5. Cohen SR. Difficulty with swallowing. In Bluestone CD,<br />

Stool SE, Arjona SK, eds. Pediatric Otolaryngology.<br />

Philadelphia: WB Saunders, 1983.<br />

6. Wolff Ph. The serial organization of sucking in the<br />

young infant. Pediatrics 1968; 42: 943–956.<br />

7. Mizuno K, Ueda A. The maturation and coordination of<br />

sucking, swallowing, and respiration in preterm infants.<br />

J Pediatr 2003; 142: 36–40.<br />

8. Mathew OP. Science of bottle-feeding. J Pediatr 1991;<br />

119: 511–519.<br />

9. Derkay CS, Schecter GL. Anatomy and physiology of<br />

pediatric swallowing disorders. Otolaryngol Clin North<br />

Am 1998; 31: 397–404.<br />

10. Miller AJ. Deglutition. Physiol Rev 1982; 62: 129–184.<br />

11. Morrell RM. The neurology of swallowing. In Groher<br />

ME, ed. Dysphagia and Management. Boston:<br />

Butterworth, 1984: 3.<br />

12. Bosma JF. Postnatal ontogeny of performances of the<br />

pharynx, larynx, and mouth. Am Rev Respir Dis 1985;<br />

131: S10–S15.<br />

13. Tuchman DN. Dysfunctional swallowing in the pediatric<br />

patient: clinical considerations. Dysphagia 1988; 2:<br />

203–208.<br />

14. Stevenson RD, Allaire JH. The development of normal<br />

feeding and swallowing. Pediatr Clin North Am 1991;<br />

38: 1439–1453.<br />

15. Rudolph CD. Diagnosis and management of children<br />

with feeding disorders. In Hyman P, Di Lorenzo C, eds.<br />

Gastrointestinal Motility Disorders in Children. New<br />

York: Academy Professional Information Services, 1994:<br />

33–54.<br />

16. Darrow DH, Harley CM. Evaluation of swallowing<br />

disorders in children. Otolaryngol Clin North Am 1998;<br />

31: 405–418.<br />

17. Illingworth RS, Lister J. The critical or sensitive period,<br />

with special reference to certain feeding problems in<br />

infants and in children. J Pediatr 1964; 65: 839–843.<br />

18. Cook IJ, Kahrilas PJ. AGA technical review on<br />

management of oropharyngeal dysphagia.<br />

Gastroenterology 1999; 116: 455–478.<br />

19. Logemann JA. Evaluation and Treatment of Swallowing<br />

Disorders. San Diego, CA: College Hill, 1983.<br />

20. Dodds WJ, Stewart ET, Logemann JA. Physiology and<br />

radiology of the normal oral and pharyngeal phases of<br />

swallowing. Am J Roentgenol 1990; 154: 953–963.<br />

21. Mendelsohn M. New concepts in dysphagia<br />

management. J Otolaryngol 1993; 22 (Suppl 1): 1–24.<br />

22. McConnel FMS. Analisys of pressure generation and<br />

bolus transit during pharyngeal swallowing.<br />

Laryngoscope 1988; 98: 71–78.<br />

23. McConnel FM, Cerenko D, Mendelsohn MS. Dysphagia<br />

after total laryngectomy. Otolaryngol Clin North Am<br />

1988; 21: 721–726.<br />

References 245<br />

24. Curtis DJ, Cruess DF, Dachman AH. Normal erect<br />

swallowing. Normal function and incidence of variations.<br />

Invest Radiol 1985; 20: 717–726.<br />

25. Sivarao DV, Goyal RK. Functional anatomy and physiology<br />

of the upper esophageal sphincter. Am J Med 2000;<br />

108 (Suppl 4a): 27S–37S.<br />

26. Palmer ED. Disorders of the cricopharyngeus muscle: a<br />

review. Gastroenterology 1976; 71: 510–519.<br />

27. Staiano A, Cucchiara S, De Vizia B et al. Disorders of<br />

upper esophageal sphincter motility in children. J<br />

Pediatr Gastroenterol Nutr 1987; 6: 892–898.<br />

28. Leslie P, Carding PN, Wilson JA. Investigation and<br />

management of chronic dysphagia. BMJ 2003; 326:<br />

433–436.<br />

29. Myer CM. Sialorrea. Pediatr Clin North Am 1989; 36:<br />

1495–1500.<br />

30. Loughlin GM. Respiratory consequences of<br />

dysfunctional swallowing and aspiration. Dysphagia<br />

1989; 3: 126–310.<br />

31. Durand M, Leahy FN, MacCallum M et al. Effect of<br />

feeding on the chemical control of breathing in the<br />

newborn infant. Pediatr Res 1981; 15: 1509–1512.<br />

32. Thach BT. Maturation and transformation of reflexes<br />

that protect the laryngeal airway from liquid aspiration<br />

from fetal to adult life. Am J Med 2001; 111 (Suppl 8A):<br />

69S–77S.<br />

33. Hoekstra RE, Perkett EA, Dugan M, Knox GE. Follow-up<br />

of the very low birth weight infant (less than 1251<br />

grams). Minn Med 1983; 66: 611–613.<br />

34. Tuchman DN. Cough, choke, spitter: the evaluation of<br />

the child with dysfunctional swallowing. Dysphagia<br />

1989; 3: 111–116.<br />

35. Rogers BT, Arvedson J, Msall M, Demerath RR.<br />

Hypoxemia during oral feeding of children with severe<br />

cerebral palsy. Dev Med Child Neurol 1993; 35: 3–10.<br />

36. Berquist WE, Ament ME. Upper GI function in sleeping<br />

infants. Am Rev Respir Dis 1985; 131: S26–S29.<br />

37. Boyle JT, Tuchman DN, Altschuler SM et al.<br />

Mechanisms for the association of gastroesophageal<br />

reflux and bronchospasm. Am Rev Respir Dis 1985; 131:<br />

S16–S20.<br />

38. Smith CH, Logemann JA, Colangelo LA et al. Incidence<br />

and patient characteristics associated with silent<br />

aspiration in the acute care setting. Dysphagia 1999;<br />

14: 1–7.<br />

39. Sonies BC. Swallowing disorders and rehabilitation<br />

techniques. J Pediatr Gastroenterol Nutr 1997; 25 (Suppl<br />

1): S32–S33.<br />

40. Kramer SS, Eicher PM. The evaluation of pediatric<br />

feeding abnormalities. Dysphagia 1993; 8: 215–224.<br />

41. American Gastroenterological Association. American<br />

Gastroenterological Association Medical Position<br />

Statement on Management of Oropharyngeal<br />

Dysphagia. Gastroenterology 1999; 116: 452–454.<br />

42. Kovar AJ. Nutrition assessment and management in<br />

pediatric dysphagia. Semin Speech Lang 1997; 18:<br />

39–49.<br />

43. Babbitt RL, Hoch TA, Coe DA et al. Behavioral<br />

assessment and treatment of pediatric feeding disorders.<br />

J Dev Behav Pediatr 1994; 15: 278–291.<br />

44. Gisel EG, Alphonce E, Ramsay M. Assessment of<br />

ingestive and oral praxis skills: children with cerebral<br />

palsy vs. controls. Dysphagia 2000; 15: 236–244.<br />

45. Ekberg O, Olsson R, Bulow M. Radiologic evaluation of<br />

dysphagia. Abdom Imag 1999; 24: 444.


246<br />

Disorders of sucking and swallowing<br />

46. Bu’Lock F, Woolridge MW, Baum JD. Development of coordination<br />

of sucking, swallowing and breathing:<br />

ultrasound study of term and preterm infants. Dev Med<br />

Child Neurol 1990; 32: 669–678.<br />

47. Rudolph CD. Feeding disorders in infants and children.<br />

Pediatrics 1994; 125: S116–S124.<br />

48. Hila A, Castell JA, Castell DO. Pharyngeal and upper<br />

esophageal sphincter manometry in the evaluation of<br />

dysphagia. J Clin Gastroenterol 2001; 33: 355–361.<br />

49. Broniatowski M, Sonies BC, Rubin JS et al. Current<br />

evaluation and treatment of patients with swallowing<br />

disorders. Otolaryngol Head Neck Surg 1999; 120:<br />

464–473.<br />

50. Langmore SE, Schatz K, Olsen N. Fiberoptic endoscopic<br />

examination of swallowing safety: a new procedure.<br />

Dysphagia 1988; 2: 216-219.<br />

51. Hamlet SL, Mutz J, Patterson, R, Jones L. Pharyngeal<br />

transit time: assessment with videofluoroscopic and<br />

scintigraphic techniques. Dysphagia 1989; 4: 4–7.<br />

52. Silver KH, Nostrand DV. Scintigraphic detection of<br />

salivary aspiration: description of a new diagnostic<br />

technique and case reports. Dysphagia 1992; 7: 45–49.<br />

53. Logemann JA. Role of the modified barium swallow in<br />

management of patients with dysphagia. Otolaryngol<br />

Head Neck Surg 1997; 116: 335–338.<br />

54. Arvedson JC. Management of pediatrics dysphagia.<br />

Otolaryngol Clin North Am 1998; 31: 453–476.<br />

55. Babbitt RL, Hoch TA, Coe DA et al. Behavioral<br />

assessment and treatment of pediatric feeding disorders.<br />

J Dev Behav Pediatr 1994; 15: 278–291.<br />

56. Helfrich-Miller KR, Rector KL, Straka JA. Dysphagia: its<br />

treatment in the profoundly retarded patient with<br />

cerebral palsy. Arch Phys Rehabil 1986; 67: 520–525.<br />

57. Croghan JE, Burke EM, Caplan S, Denman S. Pilot study<br />

of 12-month outcomes of nursing home patients with<br />

aspiration on videofluoroscopy. Dysphagia 1994; 9:<br />

141–146.<br />

58. Groher ME. Bolus Management and aspiration<br />

pneumonia in patient with pseudobulbar dysphagia.<br />

Dysphagia 1987; 1: 215–216.<br />

59. Shaker R, Easterling C, Kern M et al. Rehabilitation of<br />

swallowing by exercise in tube-fed patients with<br />

pharyngeal dysphagia secondary to abnormal UES<br />

opening. Gastroenterology 2002; 122: 1314–1321.<br />

Additional educational resource<br />

Resource center: resources for normal swallowing and<br />

swallowing disorders (www.dysphagia.com). This multidisciplinary<br />

website has a wealth of information on<br />

dysphagia, references to texts, archives and links to other<br />

related sites; it is user-friendly and comprehensive.


16<br />

Epidemiology<br />

Constipation and encopresis in<br />

childhood<br />

Jan Taminiau and Marc Benninga<br />

Constipation is a common disorder in Western<br />

societies. Survey estimates of the prevalence of<br />

constipation in childhood vary between 2 and<br />

15%. The average prevalence of constipation<br />

increases from childhood (2%) exponentially to<br />

15% in adults. It is estimated that a general<br />

practitioner will see 20–30 children with constipation<br />

per year in an average practice of 2500<br />

patients. Around 3% of a pediatrician’s practice<br />

consists of children with chronic constipation. For<br />

the pediatric gastroenterologist, consultation rates<br />

vary between 10 and 20%. Thus, a substantial<br />

number of children have constipation, 1 and this<br />

problem is of paramount practical importance for<br />

physicians.<br />

Normal physiology<br />

The Rome II criteria are currently used to define<br />

childhood constipation based on the presenting<br />

symptom profile. Two subgroups, namely functional<br />

constipation and functional fecal retention,<br />

are distinguished. Functional constipation is<br />

defined as follows: in infants and children at least<br />

2 weeks of scybalous, pebble-like hard stools for a<br />

majority of stools; or firm stools two or fewer<br />

times/week; and no evidence of structural or metabolic<br />

disease. 2<br />

Functional fecal retention in contrast, is defined as<br />

follows: from infancy to 16 years old, a history of<br />

at least 12 weeks of passage of large-diameter<br />

stools at intervals, fewer than two times/week; and<br />

retentive posturing, avoiding defecation by<br />

purposefully contracting the pelvic floor. As pelvic<br />

floor muscles fatigue, the child uses the gluteal<br />

muscles, squeezing the buttocks together. The<br />

main difference between functional constipation<br />

and functional fecal retention is the occurrence of<br />

retentive posturing in the latter. Retentive posturing<br />

is the behavioral withholding of stool during<br />

the sensation of urge, which, according to the<br />

authors of the Rome II criteria, mostly results from<br />

painful defecation.<br />

However, the Rome II criteria exclude a very<br />

common and major symptom of constipation,<br />

namely fecal soiling (if substantial called encopresis),<br />

which is not included in the definition.<br />

Therefore, to study constipation in children and<br />

define suitable end-points in pediatric research, a<br />

different definition of constipation has been used<br />

in children over 4 years of age. Pediatric constipation<br />

is thus diagnosed if at least two out of the four<br />

following criteria are fulfilled two or fewer bowel<br />

movements/week without laxatives; two or more<br />

soiling episodes/week; periodic passage of a very<br />

large amount of stool once every 7–30 days; or<br />

palpable abdominal or rectal mass on physical<br />

examination. Using this last definition, a small<br />

group of children (between 10 and 20%) do not<br />

fulfill these criteria, but still appear constipated,<br />

having a low frequency of defecation with production<br />

of firm hard stools with retentive posturing or<br />

painful experience. This last group is difficult to<br />

study, but will usually be included by the consulting<br />

physician for treatment efforts. 3<br />

Functional non-retentive fecal soiling<br />

The Rome II group defined functional nonretentive<br />

fecal soiling as follows: once a week, or<br />

more, for the preceding 12 weeks, in a child older<br />

than 4 years (who failed to be toilet trained), a<br />

history of defecation into places and at times inappropriate<br />

to the social contacts; in the absence of<br />

247


248<br />

Constipation and encopresis in childhood<br />

structural or inflammatory disease; and in the<br />

absence of signs of fecal retention. Many studies<br />

performed in encopretic children assumed that<br />

these children were constipated. Therefore, the<br />

new definition of encopresis or functional nonretentive<br />

fecal soiling states that this is a separate<br />

entity. Although in the current literature sometimes<br />

a difference is made between soiling (loss of<br />

small amounts of feces due to overflow) and encopresis<br />

(normal bowel movement) the terms are<br />

used interchangeably. Sometimes the term solitary<br />

encopresis is used in a research environment; the<br />

children who presented for constipation with<br />

soiling and encopresis (around one in three)<br />

proved to have solitary encopresis or functional<br />

non-retentive fecal soiling without any sign of<br />

constipation, and needed a different treatment<br />

strategy. 4<br />

Clinical presentation<br />

The functional diagnosis of constipation<br />

Constipation, in approximately 5–10% of the<br />

patients, is a symptom of an underlying disorder.<br />

In the remaining 90–95% of the patients constipation<br />

is idiopathic. This label implies that no clear<br />

underlying mechanism explains the defecation<br />

problem. The differential diagnosis is large, but it<br />

is highly unlikely that one of these diagnoses is<br />

missed on clinical grounds, and therefore investigations<br />

should be limited and approached<br />

cautiously. The main presenting symptoms of<br />

constipation in children are a combination of a<br />

low defecation frequency and soiling or encopresis.<br />

The latter occurs several times a day and, in<br />

severe fecal retention, also at night. If specifically<br />

asked, most children will report production of a<br />

large amount of stools, which might clog the toilet<br />

with the frequency of once a week or once a month.<br />

Often, the evacuation of such large amounts of<br />

stool is preceded by an increase of soiling<br />

frequency. At physical examination in constipated<br />

children, the rectal fecal impaction might be<br />

palpated with firm abdominal scybala, but usually<br />

manual palpation of a rectal examination with<br />

abdominal palpation as well identifies the large<br />

fecal mass. 5 Table 16.1 reports the prevalence of<br />

symptoms associated with constipation.<br />

Table 16.I The reported symptomatology of<br />

childhood constipation<br />

Clinical features Percentages<br />

Bowel history<br />

Infrequent defecation 80–100<br />

Abdominal pain 10–64<br />

Vomiting 8–10<br />

Anorexia or poor appetite 10–47<br />

Abdominal distension 0–61<br />

Passage of large stool 45–75<br />

Passage of hard stool 58–100<br />

Painful defecation 50–90<br />

Psychological problems 20–62<br />

Urinary problems 5–43<br />

Fecal incontinence 35–96<br />

Positive family history 9–49<br />

Physical examination<br />

Fissures and rectal bleeding 5–55<br />

Rectal prolapse 0–3<br />

Abdominal mass 30–71<br />

Rectal mass 28–100<br />

Percentages are adapted from references<br />

Defecation frequency<br />

The average neonate has a defecation frequency of<br />

between one and four times daily. Ninety-nine per<br />

cent of newborns pass the first stool within 48h.<br />

However, newborns with low birth weights or<br />

premature infants might have a delayed passage of<br />

the first stool. Eighty per cent of infants weighing<br />

less than 2500g pass meconium within the first<br />

24 h of life. If birth weight declines to 1000–1500 g,<br />

the first stool will be passed after 48h in 22–60%<br />

of children. This is still within the physiological<br />

range. In large cohort studies in normal children,<br />

the stool frequency varied greatly from once daily<br />

to once every other day. Therefore, a frequency of<br />

less than three times a week is considered abnormal,<br />

but other signs of constipation are necessary


to decide on the clinical symptomatology of constipation.<br />

6<br />

Normal anatomy and physiology of<br />

anorectal function<br />

The most distal part of the gastrointestinal tube is<br />

formed by the anal sphincter complex, and is<br />

responsible for maintaining fecal continence. This<br />

sphincter complex is embedded in the striated<br />

pelvic floor, where the puborectalis muscle joins<br />

the upper end of the external anal sphincter with a<br />

sling. The anal sphincter complex therefore<br />

consists of a smooth muscle component of the<br />

internal anal sphincter and a striated muscle<br />

component of the external anal sphincter and the<br />

puborectalis muscle. The internal anal sphincter is<br />

tonically contracted and generates 85% of the anal<br />

resting pressure, keeping the anal canal closed at<br />

rest. It is not under voluntary control, and is innervated<br />

by the enteric nervous system. Stimulation<br />

of mechanoreceptors in the rectal wall and the<br />

sigmoid activates intramural inhibitory neurons,<br />

leading to relaxation of the internal anal sphincter,<br />

called the anorectal inhibition reflex. 7 This occurs<br />

after presentation of stool or gas to the anal canal.<br />

The external anal sphincter is innervated by the<br />

pudendal nerve and is under voluntary control.<br />

This muscle is to some degree contracted, but on<br />

demand it might be contracted or relaxed upon the<br />

decision to defecate or to have it postponed. The<br />

puborectalis muscle ends from the pubic bone,<br />

loops around the caudal part of the rectum at the<br />

junction with the anal canal and ends at the os<br />

pubis. This muscle is tonically contracted, maintains<br />

and forms the anorectal angle and keeps the<br />

rectum and anus closed during episodes of<br />

increased abdominal pressure. The pelvic floor<br />

and gluteus muscles can assist the anal sphincter<br />

complex in contraction. Sensation arising from the<br />

anorectal area is crucial for detecting the presence,<br />

volume and consistency of feces and gas in the<br />

rectum. Mucosal and intramuscular receptors are<br />

involved. These nerve endings detect the nature of<br />

the anal contents, discriminating between gas,<br />

liquids and solid feces. Intramural stretch and/or<br />

pressure receptors detect the degree of rectal filling<br />

and produce sensations of desire to defecate and<br />

urgency. Receptors located in the pelvic floor<br />

Normal anatomy and physiology of anorectal function 249<br />

and/or the pelvis detect increases in intraabdominal<br />

pressure, causing changes in the anal<br />

sphincter complex, leading to expulsion of rectal<br />

contents. These receptors also activate compensatory<br />

reflexes to increase anal sphincter pressure<br />

and ensure fecal continence. Sensations arising<br />

from the anorectal area are transported by afferent<br />

neural pathways to the spine via ascending nerves<br />

in the spinal cord (the spinal thalamic tract). The<br />

information is transported to the thalamus. This<br />

sensory information is transferred to the limbic<br />

and somatosensory areas of the cerebellum, where<br />

sensations such as flatus and the desire to defecate<br />

will be perceived. 8<br />

Normal anorectal function depends on the<br />

complex interplay between the different anorectal<br />

and pelvic anatomic structures. The main task of<br />

the anorectum is to assure continuous fecal continence.<br />

Defecation might be considered as a controlled<br />

episode of incontinence at a socially acceptable<br />

moment. Normal defecation is a complex mechanism<br />

depending on normal sensation and motility<br />

of the anorectal area, pelvic floor, sigmoid and<br />

descending colon, and also involving the<br />

abdominal and respiratory musculature. Psychological<br />

factors have a major impact on these<br />

pathways.<br />

Fecal continence is produced by different mechanisms.<br />

9 First, the caudal end of the rectum is<br />

closed by the anal sphincter complex, creating an<br />

anal sphincter resting pressure of around<br />

40 mmHg, to which the internal sphincter<br />

contributes 85%, and the external anal sphincter<br />

15%. A reflex of the external anal sphincter<br />

increases this pressure when the intra-abdominal<br />

pressure increases acutely and counteracts imminent<br />

loss of feces. This is probably an important<br />

mechanism in normal daily life, since coughing,<br />

laughing, bending, sneezing, etc. instantly<br />

increase intra-abdominal pressure. This function<br />

of the anal sphincter for keeping the gate closed is<br />

supported by rectal motility, which is directed<br />

antegrade, to keep the rectum empty, transporting<br />

the feces back to the sigmoid and away from the<br />

anal canal. A third mechanism takes care of continence<br />

by a sensation in the cranial part of the anal<br />

sphincter. These receptors are located in the area<br />

of the skin close to the surface of the anus.


250<br />

Constipation and encopresis in childhood<br />

Transsection studies have shown that sensation is<br />

not lost, emphasizing the distal location of this<br />

sensation. Triggering of these receptors in the anal<br />

canal by feces or gas will result in the sensation of<br />

imminent loss, and gives the person the ability to<br />

prevent this loss of feces and gas by contracting<br />

the pelvic floor muscles. When defecation is not<br />

desired, the external sphincter complex and the<br />

pelvic floor muscles remain contracted until the<br />

rectal wall has adapted to the increased rectal<br />

volume; when the intrarectal pressure decreases,<br />

the sensation of urge will disappear. In addition,<br />

retrograde contraction of the caudal part of the<br />

rectum starts to occur, and transports the feces<br />

back into the sigmoid colon. The interplay of all<br />

these mechanisms starts when feces or gas enters<br />

the rectum, owing to increased propulsive activity<br />

of the colon after ingestion of a meal. Filling the<br />

rectal vault, these feces lead to an increase of<br />

intrarectal pressure and triggering of receptors<br />

located in the rectal wall. This induces the inhibition<br />

reflex, leading to a decrease of anal sphincter<br />

pressure (internal sphincter), which is more<br />

pronounced when rectal filling increases. Above a<br />

certain threshold in rectal pressure, the perception<br />

of urge occurs. At that time, due to a reflex triggered<br />

by this sensation of urge, the external anal<br />

sphincter complex contracts for a short time,<br />

preventing immediate loss of feces, thus creating<br />

time to consider whether the pelvic floor has to be<br />

contracted to stop imminent defecation or to<br />

permit the defecation process to continue.<br />

Defecation occurs when there is a difference in<br />

pressure in the rectum on the one hand (rectal<br />

contractions and straining), and the pelvic floor on<br />

the other hand (relaxation of the anal sphincter<br />

complex and pelvic floor, and flattening of the<br />

anorectal angle). 10<br />

Training is essential when a child tries to defecate<br />

without a sensation of urge (e.g. during toilet training).<br />

During the process of toilet training the child<br />

starts to obtain control of these complex mechanisms<br />

in the defecation process. The will of the<br />

child seems to be a crucial factor in this process.<br />

Aberrations in these complex interacting mechanisms<br />

might lead to clinical signs and symptoms of<br />

constipation and fecal incontinence. The pathophysiological<br />

mechanisms of these clinical entities<br />

are largely unknown. In the past decades, only<br />

little progress has been made in resolving these<br />

mechanisms. 11<br />

Colonic propagated contractions are tonic and<br />

phasic and propel the luminal contents to the<br />

distal colon and rectum. Non-propagated contractions<br />

move fecal material antegrade and retrograde<br />

and mix a shift of the fecal contents over short<br />

distances. High-amplitude propagated contractions<br />

(HAPCs) are a pattern of colonic motility<br />

originating in the proximal colon and proceeding<br />

to the distal sigmoid colon, and are associated with<br />

mass movements. They have an amplitude of at<br />

least 100mmHg, duration of


muscular (impaired contractility) and neural<br />

(uncoordinated activity) mechanisms may lead to<br />

impairment of propulsion of fecal contents leading<br />

to slow-transit constipation. 12 Mechanisms underlying<br />

these entities might be an impairment of<br />

neural transmitter function, such as nitric oxide<br />

(NO) or substance P, or aberrations in the interstitial<br />

cells of Cajal. However, this entity remains<br />

extremely difficult to explain and might be<br />

secondary to the effect of stasis.<br />

In addition to neuromuscular dysfunction, constipation<br />

can also result from massive fecal retention<br />

in the rectum. It has been shown that voluntary<br />

retention of feces can cause a delay in colonic<br />

transit in healthy volunteers. Similarly, massive<br />

fecal retention in children can also inhibit colonic<br />

transit and thus indirectly leads to prolonged<br />

transit time. In several children, slow-transit<br />

constipation after treatment changes to outlet<br />

obstruction with only a delay in the rectosigmoid<br />

region.<br />

An alternative mechanism leading to constipation<br />

is the so-called outlet obstruction or abnormalities<br />

in the dynamics underlying defecation. In these<br />

children, the delay in colonic transit involves the<br />

rectum. This mechanism is found in approximately<br />

40–60% of constipated children. It should be noted<br />

that 50% of constipated children, all presenting<br />

with the same symptomatology, have normal<br />

colonic transit time. 13–15 This observation strongly<br />

suggests that abnormal transit times are secondary<br />

events in children with constipation. It is also<br />

supported by normalization of transit time in slowtransit<br />

constipation after successful treatment.<br />

Another alternative mechanism might be abnormal<br />

sphincter function. 16,17 Under normal circumstances,<br />

the pressure generated by the anal sphincter<br />

complex should drop during an attempt to<br />

defecate, allowing expulsion of fecal contents. As<br />

stated above, in 40% of children with constipation,<br />

colonic transit is delayed in the rectum, and it was<br />

suggested that this abnormal obstruction is the<br />

result of abnormal defecation dynamics. However,<br />

using anorectal manometry, a paradox for contraction<br />

in the anal sphincter complex is observed in<br />

more than 50% of constipated children. This<br />

contraction of the anal sphincter complex might<br />

lead to fecal accumulation and constipation. This<br />

abnormality might be the underlying pathophysio-<br />

Pathophysiology of functional constipation 251<br />

logical mechanism of childhood constipation,<br />

reflecting the observed stool withholding behavior/retentive<br />

posturing. Possible suggested causes<br />

leading to this behavior might be pain, resulting<br />

from the previous production of a large, hard stool;<br />

anal fissures; primary behavioral mechanisms; not<br />

taking time to visit the toilet; and avoiding other<br />

toilets, mainly at school. However, abnormal defecation<br />

dynamics have not only been observed in<br />

fecal retention and constipation, but similarly in<br />

children with functional non-retentive fecal<br />

soiling. In addition, a large study, comparing<br />

conventional treatment with additional biofeedback<br />

training in constipated children, showed that<br />

biofeedback training could normalize the aberrant<br />

sphincter contraction, but, it did not lead to a<br />

larger success rate in children receiving additional<br />

biofeedback training. 1 These observations argue<br />

against a major contribution of abnormal defecation<br />

dynamics to the development of constipation<br />

in children.<br />

As explained previously, different types of nerve<br />

endings, giving rise to sensations of flatus, urge to<br />

defecate and pain, sense the arrival of fecal material<br />

into the rectum. This sensory information is<br />

important for initiating the defecation process.<br />

Abnormalities in rectal sensation are believed to<br />

play an important role in the pathogenesis of<br />

constipation. Children usually report that they do<br />

not feel the sensation or urge to defecate. Several<br />

studies have investigated rectal sensitivity in children<br />

with constipation. In these studies, rectal<br />

sensation is determined by inflation of a rectal<br />

balloon, measuring the volume at which the sensation<br />

of urge is perceived. These studies showed<br />

impaired rectal sensation in a subgroup of patients<br />

with constipation. In the majority of children,<br />

however, rectal sensation was normal and present<br />

after insufflation of only 20ml of air in the balloon.<br />

Rectal volumes are obviously age-dependent, with<br />

a barostat (an insufflatable rectal balloon that can<br />

keep the volume constant while varying the pressure,<br />

or vice versa) showing that one subgroup<br />

needs higher pressures with a normal volume to<br />

feel urge, but children in another subgroup need<br />

higher volumes with a normal pressure to feel<br />

urge.<br />

Abnormal rectal sensation does not seem to be an<br />

important pathophysiological entity. In fact, a


252<br />

Constipation and encopresis in childhood<br />

number of studies in children with constipation<br />

have documented a normal anal resting pressure,<br />

as well as a normal maximal rectal squeeze pressure.<br />

To date, no consistent abnormalities in motility<br />

testing have been recorded in children with constipation.<br />

There were no differences in motility parameters<br />

between children with long-standing<br />

constipation and those who have had constipation<br />

for only a short period. Furthermore, there were no<br />

indications of neuromuscular damage due to<br />

abnormalities caused by constipation or its<br />

therapy.<br />

Therefore, it is fair to conclude that, there is no<br />

current explanation of the cause(s) of constipation<br />

or suggested pathophysiological mechanisms. All<br />

assumed mechanisms have been found to be<br />

normal in the majority of children with constipation<br />

with a mainly uniform clinical presentation.<br />

Children with functional non-retentive fecal<br />

soiling, in studies on its pathophysiology, show<br />

normal anal sphincter resting pressure, normal<br />

maximal squeeze pressure and normal rectal<br />

sensation. Also, colonic transit time measurements<br />

were normal in all children with this entity. 18–20<br />

Medical history<br />

Symptoms of constipation in most children start in<br />

more than 38–65% before the age of 6 months, and<br />

some have even described bowel problems in 40%<br />

of children in the first months of life. Stool habits<br />

might change, owing to stress factors and change<br />

in nutrition, for instance change from breast<br />

feeding to formula feeding. Specific questions<br />

regarding the presence of soiling and encopresis<br />

should be asked, and also regarding the frequency<br />

and whether it is present at night-time. These children<br />

produce an enormous amount of stool once<br />

every 7–30 days, which is almost never spontaneously<br />

presented as a symptom during a first<br />

doctor’s visit. It must also be asked whether this<br />

enormous amount of stool has been produced just<br />

before the visit to the doctor (the rectum might<br />

then be empty for rectal examination). Questions<br />

about retentive posturing should be asked, but<br />

might be difficult to answer; nevertheless, the<br />

parents might notice whether stools are produced<br />

with pain and also if any of these symptoms<br />

started after a period in which defecation and<br />

stools were completely normal. When the<br />

frequency is less than three times a week, soiling<br />

and encopresis are present, and large amounts of<br />

stool are occasionally produced, the diagnosis of<br />

functional constipation is likely. When a child has<br />

a painful stressful production of hard stools<br />

without soiling and encopresis and retentive<br />

posturing, even at an almost normal frequency,<br />

constipation will still be considered probable by<br />

the physician, and a therapeutic attempt should be<br />

initiated. 21–23<br />

In developed societies most children are toilet<br />

trained by the age of 2–3 years. There is no relation<br />

between failed toilet training and the development<br />

of constipation or functional non-retentive fecal<br />

soiling. Before the age of 4 years, the diagnosis of<br />

constipation can be made only on low or decreased<br />

frequency of defecation, production of occasional<br />

voluminous stools, production of hard stools with<br />

discomfort, pain and/or retentive posturing. In<br />

infants, the diagnosis of constipation can be<br />

inferred when they cry before, during and after<br />

defecation, and this consists of firm, hard stools.<br />

Behavioral problems occur often, but they are<br />

almost invariably secondary to the social consequences<br />

and depend on individual styles of<br />

coping. Such problems typically improve considerably<br />

once the defecation difficulties are successfully<br />

treated. However, poor social behavior, or an<br />

inadequate parent–child relationship may be associated<br />

with the continuation of defecation problems.<br />

Urinary problems, diurnal and also nocturnal<br />

enuresis, are secondary to the defecation<br />

problems and usually improve after successful<br />

treatment.<br />

Physical examination<br />

Abdominal masses should be gently looked for by<br />

palpation in the lower abdominal quadrants and<br />

suprapubic area. The stool mass might be palpable<br />

extending from the pelvis, but smaller scybala<br />

might also be detected. Usually, a bimanual palpation<br />

through a rectal examination with one hand<br />

combined with abdominal palpation with the<br />

other hand, clearly defines the rectal mass of stool<br />

the best. At the first visit a rectal examination<br />

should be attempted in all children who present


with constipation or non-functional fecal retention.<br />

Only in 10–15% of children is the fear of<br />

examination too high, when this procedure has to<br />

be omitted. Otherwise, it is well perceived and<br />

gives the essential information about the rectal<br />

fecal mass. Anal fissures can be clearly visualized<br />

by spreading the anus, and during the examination<br />

anatomical abnormalities should be inspected,<br />

checking the anal position in the perineum and its<br />

asymmetry: an ectopic anus can in fact be the<br />

cause of constipation. The inspection of the lower<br />

lumbar and sacral spine areas should be focused<br />

on dimpling, surgical scars and hair tufting. Also,<br />

tendon reflexes should be evoked. Usually, the<br />

school-aged child has stools or soiling in his<br />

underpants and one should be aware of the<br />

production of a large amount of stool just prior to<br />

the visit to the physician. That might interfere<br />

with the examination results. Fissures are considered<br />

to be related to the initiation of constipation<br />

only in very young infants; an abdominal and/or<br />

rectal mass is found in 30–100%, depending on the<br />

investigator. 24<br />

Differential diagnosis<br />

Over 90% of pediatric patients with constipation<br />

have functional constipation or non-retentive<br />

functional fecal soiling. Only in a very small<br />

number of children is a distinct etiology established,<br />

and these cases are not easily overlooked.<br />

Changed dietary habits might decrease the intake<br />

of fluids and food. The change from breast milk to<br />

formula feeding as an initiating point is highly<br />

suggestive of constipation.<br />

A careful perianal examination will disclose an<br />

imperforate anus, or an abnormal anteriorly<br />

displaced anus, as well as other congenital<br />

anatomic and structural defects.<br />

Spinal abnormalities are indicated by abnormalities<br />

of the skin, and also growth impairment and a<br />

combination of fecal and urinary incontinence or<br />

constipation.<br />

Metabolic disturbances are related to severe dehydration,<br />

as is seen in diabetes insipidus, hypocalcemia<br />

and renal tubular acidosis, and are not<br />

easily overlooked.<br />

Examinations 253<br />

Hypothyroidism is screened for in the neonatal<br />

period and might still occur later, but is not easily<br />

overlooked as a symptom complex in a child.<br />

Hirschsprung’s disease is suspected after a delayed<br />

production of meconium beyond 48h after birth,<br />

and continuing defecation problems usually with<br />

distended abdomen and poor appetite, failure to<br />

gain weight and vomiting. Also, the history would<br />

often document episodes of acute, severe enterocolitis<br />

with explosive diarrhea. Clinically, the diagnosis<br />

is made by insertion of a catheter through the<br />

anus in the distended part, after which decompression<br />

follows (see Chapter 17).<br />

Also, Hirschsprung’s disease might involve a very<br />

small segment of the anorectum and present as<br />

constipation. In practice, this is seen only on<br />

extremely rare occasions. We have not made this<br />

diagnosis in our practice for decades.<br />

Recently, magnetic resonance imaging (MRI) of the<br />

pelvis and distal colon has been proposed in order<br />

to detect small abnormalities, cysts, tumors or<br />

tethered cord which may be the basis of constipation<br />

without any other neurological symptom.<br />

This approach, however, has not been performed<br />

in a large population, and is currently awaiting<br />

some protocol definitions. 25<br />

Examinations<br />

Plain abdominal X-ray<br />

On a plain abdominal radiograph some amount of<br />

stool is always visible. The simple report of ‘stools<br />

in the colon’ should therefore not be regarded as<br />

indicative of constipation. Barr et al tried to<br />

develop a scoring system to define constipation.<br />

Although this scoring system is used to diagnose<br />

constipation in children with chronic abdominal<br />

pain as the only symptom, the method is subject to<br />

large intra- and interobserver variability. 26,27<br />

Colonic transit times are measured by the Metcalf<br />

method. Swallowed radio-opaque tiny rings allow<br />

the intestinal transit to be measured. This test<br />

shows normal transit times in half of the constipated<br />

children and is therefore only useful to diagnose<br />

slow-transit constipation, a clinical entity<br />

usually apparent with night-time soiling and resis


254<br />

Constipation and encopresis in childhood<br />

tance to treatment. The upper limit of colonic<br />

transit is 63h. A colonic transit time delayed to<br />

more than 100h is defined as slow-transit constipation.<br />

A palpable rectal mass is always found.<br />

The method can also be used to support the diagnosis<br />

of functional non-retentive fecal soiling, but<br />

it has no place in the general management of children<br />

with constipation.<br />

Barium enema<br />

A barium enema performed in the unprepared<br />

colon might delineate the transition zone in<br />

Hirschsprung’s disease in young infants, but is of<br />

limited value, as the transition zone is often difficult<br />

to see and might only be of value to the operating<br />

surgeon preoperatively after the diagnosis<br />

has been made.<br />

Defecography<br />

Defecography uses a relatively high radiation<br />

exposure and is difficult and unreliable in children.<br />

Anorectal manometry<br />

Anorectal manometry is a relatively safe, minor<br />

invasive technique. With an open perfusion<br />

system, pressures may be assessed in anal sphincter<br />

muscles at rest, at squeezing and during straining.<br />

During defecation, the pelvic pressure or<br />

rectal pressure increases, owing to abdominal<br />

muscle contraction, and the anal canal pressure<br />

decreases, creating a difference to allow defecation.<br />

A rectal balloon might be inflated to assess<br />

the rectal anal inhibitory reflex against relaxation<br />

of the internal anal sphincter. In the clinical<br />

setting, except for Hirschsprung’s disease, the<br />

majority of children have normal anorectal pressures.<br />

Rectal sensitivity after inflation of a balloon is<br />

normal. Children who have abnormal defecation<br />

dynamics, in an attempt to defecate the balloon,<br />

will contract the anal sphincters at the same time<br />

that they contract the abdominal musculature.<br />

This can be shown on a screen and children can<br />

learn to relax instead of having this paradoxical<br />

contraction during defecation of the anal muscula-<br />

ture. The training by reinforcement is called<br />

biofeedback training.<br />

Anorectal manometry is normal in children with<br />

functional constipation or functional non-fecal<br />

retention, and therefore this should not be used. 1<br />

Colonic manometry<br />

Colonic manometry 28 requires colonoscopic placement<br />

of the catheter with motility recordings,<br />

which are measured the day after placement.<br />

Colonic motility can be tested over hours, possibly<br />

with bisacodyl administration to enable propagated<br />

contractions to be endured. This test is<br />

reserved for children with intractable constipation.<br />

Some investigators can differentiate children<br />

with functional fecal retention from those with no<br />

neuropathy or myopathy of the colon with this<br />

technique. The technique is still used in a few<br />

centers; however, its value in the diagnostic<br />

process of constipation has to be assessed.<br />

Treatment<br />

Only a few studies, most of them with small<br />

patient numbers, have been performed to evaluate<br />

treatment for children with defecation disorders.<br />

Treatment of constipation is mainly based on<br />

empirical experience, rather than on placebocontrolled,<br />

randomized studies. The main reason<br />

for a consultation is interference with social activities<br />

of the child and its family because of the defecation<br />

problems (soiling and encopresis). They<br />

should be educated and the problem should be<br />

demystified. 29 It should be stressed that the prevalence<br />

of constipation and soiling is quite common.<br />

The relationship between fecal impaction and<br />

overflow diarrhea should be explained with the<br />

help of drawings, and the involuntary nature of the<br />

loss of feces in the underwear made clear. The<br />

objective of this is to make parents start to feel<br />

more comfortable and decrease the feeling of<br />

shame in the child. The child and parents almost<br />

invariably accept a positive non-accusatory<br />

approach with relief. Organic versus functional<br />

disease should be explained clearly, once the<br />

history and physical examination have been evaluated<br />

and the diagnosis is made with confidence.<br />

It should be stressed that there is no need for


further investigation. The therapy is stressful and<br />

might be long lasting. The child is the only one<br />

who is responsible for completing the treatment.<br />

Behavioral therapy (see below) is not generally<br />

recommended; if the treatment is successful, the<br />

behavior abnormalities usually normalize too. It is<br />

helpful to advise that a diary is kept to follow and<br />

gain insight into the therapeutic progress; it might<br />

also motivate the child. The child has to fill in the<br />

diary himself, and that enhances responsibility. It<br />

might also be linked to a reward system. 30–33<br />

Another simple general measure is toilet training<br />

to normalize defecation. It should be explained<br />

that the sensation of defecation must be felt by the<br />

child and, usually from the age of 4, will be admitted<br />

by the child when it is explained well. The<br />

treatment of the child should involve an instruction<br />

to attempt to defecate three times a day for<br />

5 min after each meal. This should stimulate<br />

straining actively and the child should be able to<br />

place his feet on a footrest or on the ground (the<br />

younger ones usually need a footrest). This is<br />

important, to flatten the anorectal angle, facilitating<br />

fecal expulsion. Using this approach, the child<br />

will be forced to focus on its bowel function. The<br />

emptying of the rectum will reduce the risk of fecal<br />

soiling during the rest of the day. This approach is<br />

only successful in 50% of children with severe<br />

constipation referred to a tertiary hospital, without<br />

any added pharmacological treatment.<br />

Fluid intake<br />

Fluid intake of children with constipation has<br />

been found to be slightly lower than normal, 33 and<br />

so is energy intake, estimated in a prospective<br />

study to be around 100kcal/day lower. Fiber intake<br />

of children with constipation was also slightly<br />

lower than controls, but in 6 months after reinforcement<br />

we found that it could be increased in<br />

some by only around 9g while it was actually<br />

decreased in others up to 16g. The total amount<br />

remained less than 20g and no child achieved<br />

30g/day. There was no relation between fiber<br />

intake, colonic transit times and success in treatment.<br />

Therefore, while we would still recommend<br />

increasing fiber intake in constipated children, one<br />

might not expect much success in children from<br />

this intervention alone. 34<br />

Pharmacotherapy<br />

Treatment 255<br />

No double-blind randomized study on oral or<br />

rectal laxatives has been performed in children<br />

with constipation, except for the use of cisapride. 35<br />

The effect of lactulose, one of the most widely<br />

used stool softeners, has not been investigated. An<br />

evidence-based treatment cannot be constructed.<br />

Almost all advice concerning the use of oral or<br />

rectal laxatives is currently based on clinical<br />

empirical experience.<br />

The major aim of medical therapy is two-fold: to<br />

remove fecal impaction; and to prevent its recurrence<br />

by avoiding prolonged rectal distension.<br />

Removing fecal impaction<br />

The most convenient approach in the majority of<br />

cases is by daily enemas, better administered after<br />

returning from school, as it might take some time<br />

to induce defecation. 36 The effect is mainly due to<br />

a sudden increase in rectal filling, which leads to a<br />

strong rectal contraction and reflex relaxation of<br />

the internal anal sphincter, often followed by a<br />

bowel movement. The advantage of an enema<br />

strategy is a direct effect on overflow incontinence<br />

and relief for the child and parents. Enemas have<br />

the effect of hyperosmosis and increased fluids in<br />

the colon; furthermore, they lubricate the feces<br />

and distend the colon. Enemas used in children<br />

may contain dioctyl sodium sulfosuccinate and<br />

sorbitol or phosphate. Also, oil enemas and tap<br />

water enemas can be used. Hyperphosphate<br />

enemas in children have some risk when the<br />

enema administration is not followed by defecation<br />

and might lead, through retention, to hyperphosphatemia,<br />

subsequent hypocalcemia, hypokalemia<br />

and dehydration. 37,38 Also, hypocalcemia<br />

might lead to tetany and cardiac abnormalities in<br />

children, which in practice it is mainly seen in<br />

treatment of Hirschsprung’s disease prior to<br />

surgery. Enemas should contain a sufficient<br />

amount of fluid: under 2 years of age around 60ml<br />

and above 2 years of age 120ml. Usually, evacuation<br />

is achieved within 3 days with consecutive<br />

daily enemas. Otherwise, it should be continued<br />

until the fecal mass has been removed successfully.<br />

When soiling relapses or the defecation<br />

frequency does not normalize with adequate treatment<br />

with oral laxatives, enemas are added to


256<br />

Constipation and encopresis in childhood<br />

long-term treatment on an individual basis,<br />

usually 2–3 times weekly. When this procedure<br />

for severe fecal impaction is unsuccessful, and<br />

also in children who are treated for constipation<br />

and have recurrent large impaction, intermittent<br />

nasogastric lavage treatment might be tried. A safe<br />

and efficient method to clean the intestine is the<br />

balanced electrolyte solution of non-resorbable<br />

polyethylene glycol. This solution is often well<br />

taken by the child and, if he cannot swallow the<br />

required quantity, nasogastric tubes might be used.<br />

The recommended volume varies between 14 and<br />

40ml/kg per h, not to exceed 1litre/h, and treatment<br />

should be continued until clear fluid is<br />

excreted through the rectum, an effect usually<br />

reached within 24h. This treatment gives some<br />

temporary relief, but is not a substitute for other<br />

treatment protocols.<br />

Preventing recurrence of fecal impaction<br />

The remaining initial treatment after desimpaction<br />

is achieved involves osmotic laxatives (lactulose,<br />

lactitol) at a dose of 1–6g/kg body weight per day.<br />

The main function is to loosen stool consistency,<br />

and enhance the rectal sensation and urge of the<br />

child to defecate. The dose might be increased<br />

without any danger up to 20–40g daily. Sideeffects<br />

are initially bloating, flatulence and<br />

increase of abdominal pain, but these symptoms<br />

usually disappear after the first 1–2 weeks.<br />

Therapy should be continued for at least 3–6<br />

months until constipation has disappeared; the<br />

dose should be titrated individually. If this treatment<br />

is insufficient, recently polyethylene glycol<br />

has been shown to be effective in treating constipation<br />

in children at a dose of 0.8g/kg per day. It<br />

also functions as an osmotic laxative. 39,40 It has<br />

fewer side-effects in terms of flatulence and<br />

abdominal pain, and its tasteless nature increases<br />

compliance. The effects are comparable to those of<br />

lactulose.<br />

Another laxative is mineral oil, an emollient agent,<br />

at a dose of 1–3ml/kg per day, whose main function<br />

is to keep the rectal walls lubricated.<br />

Disadvantages are anal oil seepage with coloring of<br />

the underwear, which is not removable with<br />

washing, and the risk of aspiration and chemical<br />

pneumonia in very young children, and in children<br />

with cerebral palsy or other causes of mental<br />

retardation. Milk of magnesia is a relatively non-<br />

absorbable laxative, and can be given at a dose of<br />

1ml/kg per day. The treatment might be started at<br />

a higher dose, up to 3ml/kg per day. Prokinetic<br />

agents, such as cisapride at a dose of 0.2mg/kg<br />

body weight three times daily, have some transient<br />

effect on constipation, but this drug is no longer<br />

available.<br />

Stimulant laxatives such as bisacodyl, sodium<br />

picosulfate and senna alkaloids should be used<br />

with caution and have no advantage over osmotic<br />

laxatives. Biofeedback training has been shown to<br />

normalize defecation dynamics, but has no place<br />

in management, because it does not influence therapeutic<br />

outcome. Surgical colostomies for antegrade<br />

enemas have been successfully tried, but<br />

also have their complications; at present, indications<br />

are difficult to establish.<br />

In functional non-retentive fecal soiling, biofeedback<br />

training showed a minor effect on treatment<br />

outcome. 41 In this form of constipation, the main<br />

treatment is laxatives. Daily enemas in the<br />

morning might help to have a clean day, which<br />

might stimulate the defecation behavior. Oral laxatives<br />

usually worsen soiling and encopresis and<br />

should be avoided in these children with functional<br />

non-retentive fecal soiling.<br />

Biofeedback training compared to conventional<br />

treatment<br />

In a large cohort of 200 children who had conventional<br />

treatment and one arm of biofeedback training<br />

added, they did learn to normalize their defecation<br />

dynamics, but this normalization had no<br />

influence on outcome of treatment. Only in functional<br />

non-retentive fecal soiling did biofeedback<br />

have a modest effect in a sample of 100 tested children.<br />

Prognosis<br />

In a cohort of more than 400 constipated children,<br />

follow-up was obtained by annual telephone<br />

contact in more than 95% of children. Mean<br />

follow-up was 5 years (range 1–8). The cumulative<br />

percentage of children who were successfully<br />

treated during follow-up was 60% in 1 year,<br />

increasing to 80% in 8 years (Figure 16.1).<br />

Interestingly, successful treatment was more


Figure 16.1 Outcome with and without laxatives.<br />

frequent in children without encopresis, and in<br />

children with an age at onset of defecation difficulty<br />

older than 4 years. In the group of successfully<br />

treated children, approximately 50%<br />

remained symptom free during the follow-up<br />

period, while the other half experienced at least<br />

REFERENCES<br />

1. van der Plas RN, Benninga MA, Büller HA et al.<br />

Biofeedback training in treatment of childhood<br />

constipation: a randomized, controlled study. Lancet<br />

1996; 348: 776–780.<br />

2. Rasquin-Weber A, Human PE, Cuccchiara S et al.<br />

Childhood functional gastrointestinal disorders. Gut<br />

1999; 45(Suppl 2): 60–68.<br />

3. Loening-Baucke V. Constipation in children.<br />

Gastroenterology 1993; 105: 1557–1564.<br />

4. Benninga MA, Buller HA, Heymans HS et al. Is<br />

encopresis always the result of constipation? Arch Dis<br />

Child 1994; 71: 186–193.<br />

5. Loening-Baucke V. Factors determining outcome in<br />

children with chronic constipation and fecal soiling.<br />

Gut 1989; 30: 999–1006.<br />

6. Weaver LT, Lucas A. Development of bowel habit in<br />

preterm infants. Arch Dis Child 1993; 68: 317–320.<br />

References 257<br />

one period of relapse. The relapses occurred more<br />

frequently in boys than in girls. In a subset of children<br />

aged 16 years and older, constipation was still<br />

present in around 30%, thus continuing into<br />

adulthood. 42<br />

7. Wald A. Colonic and anorectal motility testing in<br />

clinical practice. Am J Gastroenterol 1994; 89:<br />

2109–2115.<br />

8. Carlstedt A, Nordgren S, Fasth S et al. Sympathetic<br />

nervous influence on the internal anal sphincter and<br />

rectum in man. Int J Colorect Dis 1988; 3: 90–95.<br />

9. Sarna SK. Physiology and pathophysiology of colonic<br />

motor activity (1). Dig Dis Sci 1991; 36: 827–862.<br />

10. Benninga MA, Wijers OB, Hoeven van der CW et al.<br />

Manometry, profilometry, and endosonography: normal<br />

physiology and anatomy of the anal canal in healthy<br />

children. J Pediatr Gastroenterol Nutr 1994; 18: 68–77.<br />

11. Di Lorenzo C, Flores AF, Hyman PE. Age–related<br />

changes in colon motility. J Pediatr 1995; 127: 593–596.<br />

12. Wheatly JM, Hutson JM, Chow CW et al. Slow-transit<br />

constipation in childhood. J Pediatr Surg 1999; 34:<br />

829–832.


258<br />

Constipation and encopresis in childhood<br />

13. Corazziari E, Cucchiara S, Staiano A et al.<br />

Gastrointestinal transit time, frequency of defecation,<br />

and anorectal manometry in healthy and constipated<br />

children. J Pediatr 1985; 106: 379–382.<br />

14. Benninga MA, Büller HA, Tytgat GNJ et al. Colonic<br />

transit time in constipated children: does pediatric<br />

slow-transit constipation exist? J Pediatr Gastroenterol<br />

Nutr 1996; 23: 241–251.<br />

15. Metcalf AM, Phillips SF, Zinsmeister AR et al.<br />

Simplified assessment of segmental colonic transit.<br />

Gastroenterology 1987; 92: 40–47.<br />

16. Loening-Baucke VA, Younoszai MK. Abnormal anal<br />

sphincter response in chronically constipated children.<br />

J Pediatr 1982; 100: 213–218.<br />

17. Read NW, Timms JM. Pathophysiology of constipation.<br />

Acta Gastroenterol Belg 1987; 50: 393–404.<br />

18. Steffen R, Loening-Baucke V. Constipation and<br />

encopresis. In Wycki R, Hyams JS, eds. Pediatric<br />

Gastrointestinal Disease, 2nd edn. Philadelphia: WB<br />

Saunders, 1999: 43–50.<br />

19. Cucchiara S, Coremans G, Staiano A et al.<br />

Gastrointestinal transit time and anorectal manometry<br />

in children with fecal soiling. J Peditar Gastroenterol<br />

Nutr 1984; 3: 545–550.<br />

20. Borowitz SM, Sutphen J, Ling W, Cox DJ. Lack of<br />

correlation of anorectal manometry with symptoms of<br />

chronic childhood constipation and encopresis. Dis<br />

Colon Rectum 1996; 39: 400–405.<br />

21. van der Plas RN, Benninga MA, Redekop WK et al. How<br />

accurate is the recall of bowel habits in children with<br />

defecation disorders? Eur J Pediatr 1997; 156: 178–181.<br />

22. Loening-Baucke V. Constipation in early childhood:<br />

patient characteristics, treatment, and longterm follow<br />

up. Gut 1993; 34: 1400–1404.<br />

23. Loening-Baucke V. Urinary incontinence and urinary<br />

tract infection and their resolution with treatment of<br />

chronic constipation of childhood. Pediatrics 1997; 100:<br />

228–232.<br />

24. Meunier P, Mollard P, Marechal JM. Physiopathology of<br />

megarectum: the association of megarectum with<br />

encopresis. Gut 1976; 17: 224–227.<br />

25. Andrade R, Fortunato C, Nurko S. Spinal cord abnormalities<br />

in children with constipation. Gastroenterology<br />

2002; 122: A315.<br />

26. Benninga MA, Büller HA, Staalman CR et al. Defecation<br />

disorders in children, colonic transit time versus the<br />

Barr-score. Eur J Pediatr 1995; 154: 277–284.<br />

27. Barr RG, Levine MD, Wilkinson RH, Mulvihill D.<br />

Chronic and occult stool retention: a clinical tool for its<br />

evaluation in school-aged children. Clin Pediatr (Phila)<br />

1979; 18: 674, 676.<br />

28. Di Lorenzo C, Flores AF, Reddy SN, Hyman PE. Use of<br />

colonic manometry to differentiate causes of intractable<br />

constipation in children. J Pediatr 1992; 120: 690–695.<br />

29. van der Plas RN, Benninga MA, Taminiau JA, Büller<br />

HA. Treatment of defecation problems in children: the<br />

role of education, demystification and toilet training.<br />

Eur J Pediatr 1997; 156: 689–692.<br />

30. Lowery SP, Srour JW, Whitehead WE, Schuster MM.<br />

Habit training as treatment of encopresis secondary to<br />

chronic constipation. J Pediatr Gastroenterol Nutr 1985;<br />

4: 397–401.<br />

31. Roberson G, Meshkinpour H, Vandenberg K et al. Effects<br />

of exercise on total and segmental colon transit. J Clin<br />

Gastroenterol 1993; 16: 300–303.<br />

32. Levine MD, Mazonson P, Bakow H. Behavioral symptom<br />

substitution in children cured of encopresis. Am J Dis<br />

Child 1980; 134: 663–667.<br />

33. Stark LJ, Spirito A, Lewis AV, Hart KJ. Encopresis:<br />

behavioral parameters associated with children who fail<br />

medical management. Child Psychiatry Hum Dev 1990;<br />

20: 169–179.<br />

34. Mooren GC, van der Plas RN, Bossuyt PM et al. Het<br />

verband tussen inname van voedingsvezels en<br />

chronische obstipatie bij kinderen. Ned Tijdschr<br />

Geneesk 1996; 140: 2036–2039.<br />

35. Nurko S, Garcia-Arnada JA, Guerrero VY, Worona LB.<br />

Treatment of intractable constipation in children:<br />

experience with cisapride. J Pediatr Gastroenterol Nutr<br />

1996; 22: 38–44.<br />

36. Price KJ, Elliott TM. What is the rule of stimulant<br />

laxatives in the management of childhood constipation<br />

and soiling? (Cochrane Review). Cochrane Database<br />

Syst Rev 2001; 3: CD002040.<br />

37. Martin RR, Lisehora GR, Braxton M Jr, Barcia PJ. Fatal<br />

poisoning from sodium phosphate enema: case report<br />

and experimental study. JAMA 1987; 257: 2190–2192.<br />

38. Sotos JF, Cutler EA, Finkel MA, Doody D. Hypocalcemic<br />

coma following two pediatric phosphate enemas.<br />

Pediatrics 1977; 60: 305–307.<br />

39. Loening-Baucke V. Polyethylene glycol without electrolytes<br />

for children with constipation and encopresis. J<br />

Ped Gastroent Nutr 2002; 34: 372–377.<br />

40. Youssef NN, Peters JM, Hnederson W et al. Dose<br />

response of PEG 3350 for the treatment of childhood<br />

fecal impaction. J Pediatr 2002; 141: 410–415.<br />

41. van Ginkel R, Benninga MA, Blommaart PJ et al. Lack of<br />

benefit of laxatives as adjunctive therapy for functional<br />

nonretentive fecal soiling in children. J Pediatr 2000;<br />

137: 808–813.<br />

42. van Ginkel R, Reitsma JB, Buller HA et al. Childhood<br />

constipation: longitudinal follow-up beyond puberty.<br />

Gastroenterology 2003; 125: 357–363.


17<br />

Introduction<br />

Hirschsprung’s disease and<br />

intestinal neuronal dysplasias<br />

Annamaria Staiano, Lucia Quaglietta and Renata Auricchio<br />

Hirschsprung’s disease is a heterogeneous genetic<br />

disorder, resulting from an anomaly of the enteric<br />

nervous system of neural crest cell origin, characterized<br />

by the absence of parasympathetic intrinsic<br />

ganglionic cells in the submucosal and myenteric<br />

plexuses. It is regarded as the consequence of the<br />

premature arrest of the craniocaudal migration of<br />

vagal neural crest cells in the hindgut, between the<br />

5th and 12th weeks of gestation, to form the<br />

enteric nervous system, and is therefore considered<br />

a neurocristopathy. 1<br />

Epidemiology<br />

Hirschsprung’s disease occurs in approximately 1<br />

of 5000 live births, and with a male predominance<br />

of 4 : 1. It is generally sporadic, although in 3–7%<br />

of cases a genetic transmission has been reported. 1<br />

The risk for short-segment disease is 5% in brothers<br />

and 1% in sisters of index cases; for longsegment<br />

disease the risk is 10%, regardless of sex. 2<br />

Etiology<br />

Hirschsprung’s disease is characterized by the<br />

congenital absence of ganglionic cells in the submucosal<br />

and myenteric plexuses in the distal bowel,<br />

and variable proportion of the colon proximally. The<br />

embryonic disorder is a lack of the craniocaudal<br />

migration, differentiation and maturation of neuroblasts<br />

from the neural crests. The earlier the migration<br />

ceases, the longer the aganglionic segment will be.<br />

The aganglionic segment is permanently contracted,<br />

causing dilatation of its proximal tract. 3<br />

Hirschsprung’s disease may be classified according<br />

to the length of the aganglionic segment: the<br />

classic form (short segment in 70–75% of cases) is<br />

limited to the rectum and sigmoid colon; the long<br />

segment, or subtotal colonic disease (10–15%),<br />

generally involves the bowel up to the splenic<br />

flexure; total colonic aganglionosis (TCA; 3–6%)<br />

may extend to involve a variable amount of the<br />

short bowel; and total intestinal aganglionosis is<br />

sometimes associated with intestinal malrotation<br />

or volvulus. 4 Ultrashort-segment aganglionosis is<br />

considered a functional alteration, without any<br />

detectable histological finding. Although longer<br />

aganglionic segments tend to produce more<br />

dramatic symptoms, some patients with even<br />

short-segment disease deteriorate rapidly. 5<br />

Pathophysiology<br />

The hallmark of diagnosis is the absence of<br />

ganglionic cells from the myenteric and submucosal<br />

plexuses, as seen on a full-thickness or<br />

suction (mucosal–submucosal) biopsy of the<br />

rectum. Proximal contents fail to enter the unrelaxed,<br />

aganglionic segment. The lack of nonadrenergic–non<br />

cholinergic (NANC) inhibitory<br />

innervation is responsible for a tonic contraction<br />

of the affected segment, with absence of peristalsis<br />

and proximal dilatation of the gut. 6<br />

Morphologically, ganglionic cells are absent from<br />

the narrowed segment and for some distance<br />

(usually 1–5cm) into the dilated segment. The<br />

pattern of nerve fibers is also abnormal; they are<br />

hypertrophic with abundant, thickened bundles.<br />

Specific stains for acetylcholinesterase are used to<br />

highlight the abnormal morphology. 7,8<br />

259


260<br />

Hirschsprung’s disease and intestinal neuronal dysplasias<br />

In recent years, new insights into the pathophysiology<br />

of Hirschsprung’s disease have been gained.<br />

There is growing evidence suggesting that the<br />

disease might be the expression of a genetic alteration,<br />

as reported in the genetic section of this<br />

chapter.<br />

It has also been suggested that abnormal expression<br />

of the muscular neural cell adhesion molecule<br />

is likely to be associated with an arrest in the<br />

craniocaudal migration of neural cells to their<br />

most distal location. 9 Furthermore, the lack of<br />

nitric oxide (NO)-producing nerve fibers in the<br />

aganglionic intestine probably contributes to the<br />

inability of the smooth muscle to relax, thereby<br />

causing lack of peristalsis. 10 In addition, in the<br />

aganglionic segments, interstitial cells of Cajal are<br />

scarce and their network appears to be disrupted. 11<br />

Clinical signs/symptoms<br />

In the newborn, symptoms may appear during the<br />

first hours of life with failure to pass meconium, or<br />

in the first week with a picture of intestinal<br />

obstruction. However, the delay in passage of<br />

meconium is not constant, and a percentage of<br />

children still presents late or with complications<br />

despite a history of problems since birth.<br />

Enterocolitis, the most common complication, is<br />

always severe and is an important cause of mortality<br />

in these young patients.<br />

In infants and children, the presentation is often<br />

less dramatic and may not mimic acute intestinal<br />

obstruction (Figure 17.1). Severe constipation and<br />

recurrent fecal impaction are more common.<br />

Physical examination reveals a distended<br />

abdomen and a contracted anal sphincter and<br />

rectum in most children. The rectum is devoid of<br />

stools, except in cases of short-segment aganglionosis.<br />

As the finger is withdrawn, there may be<br />

an explosive discharge of foul-smelling liquid<br />

stools, with decompression of the proximal normal<br />

bowel.<br />

Complications<br />

Over the past four decades, enterocolitis has been<br />

a major cause of morbidity and mortality in infants<br />

and children with Hirschsprung’s disease. The<br />

mean incidence is 25%, but the range is great<br />

(from 17 to 50%) and may be differently estimated,<br />

depending on the manner in which it is diagnosed.<br />

Mortality rates range from 0 to 33%, probably<br />

reflecting differences in the diagnostic criteria. 12<br />

Mortality also appears to be associated with other<br />

factors, such as trisomy 21. The classic clinical<br />

manifestations described for enterocolitis include<br />

abdominal distension, explosive diarrhea, vomiting,<br />

fever, lethargy, rectal bleeding and shock. 13<br />

Abdominal radiographs show the intestinal ‘cutoff’<br />

sign in the rectosigmoidal region with absence<br />

of air distally. Other common findings are smallbowel<br />

dilatation in 74% of patients and multiple<br />

air–fluid levels. 14 Because of the risk of perforation,<br />

contrast enema should not be performed in<br />

the presence of clinical enterocolitis.<br />

Postoperative enterocolitis has been associated<br />

with a fairly high rate of mortality in several series.<br />

In fact, when examining the deaths related to<br />

Figure 17.1 Severe abdominal distension in an infant<br />

with Hirschsprung’s disease.


Hirschsprung’s disease, several groups found that<br />

approximately 50% of deaths resulted from<br />

complications directly related to an enterocolitis<br />

episode. 15,16<br />

Rectal washouts should be the initial approach in<br />

the care of a child, regardless of age, who presents<br />

with enterocolitis. Along with washouts, intravenous<br />

antibiotics or oral metronidazole (in mild<br />

cases) should be used. Should the disease process<br />

fail to improve, or the infant’s condition deteriorate,<br />

the performance of a leveling colostomy<br />

should be considered. 15,16<br />

Diagnosis with differential<br />

For the diagnosis of Hirschsprung’s disease, the<br />

subject’s history is very important. The crucial<br />

elements to obtain are: the age of the appearance of<br />

symptoms; whether the passage of meconium has<br />

been normal or delayed; and whether the child<br />

presented with episodes of functional intestinal<br />

obstruction. In addition, a functional (idiopathic)<br />

megacolon must be ruled out. A clinical comparison<br />

between functional and congenital megacolon<br />

is shown in Table 17.1. When the history (early<br />

onset of constipation, absence of fecal soiling)<br />

and/or the physical examination (empty rectal<br />

ampulla) suggests an organic cause, anorectal<br />

manometry should be performed.<br />

Table 17.1 Differentiating types of megacolon in children<br />

Functional Colonic<br />

fecal neuromuscular<br />

Signs and symptoms retention disorders<br />

Soiling common rare<br />

Obstructive symptoms rare common<br />

Large-caliber stools common rare<br />

Stool-withholding behavior common rare<br />

Enterocolitis never possible<br />

Associated upper-gastrointestinal symptoms never common<br />

Symptoms from birth rare common<br />

Localization of stools rectum rectal and<br />

extrarectal<br />

Diagnosis with differential 261<br />

Anorectal manometry evaluates the response of<br />

the internal anal sphincter to inflation of a balloon<br />

in the rectal ampulla. 17 When the rectal balloon is<br />

inflated, there is normally a reflex relaxation of the<br />

sphincter. The rectoanal inhibitory reflex is absent<br />

in patients with Hirschsprung’s disease; there is no<br />

relaxation, or there may even be paradoxical<br />

contraction of the internal anal sphincter (Figure<br />

17.2). Anorectal manometry is particularly useful<br />

when the aganglionic segment is short and the<br />

results of radiological or pathological studies are<br />

equivocal.<br />

Barium enema is helpful in the assessment of a<br />

transition zone between aganglionic and<br />

ganglionic bowel, and in giving an estimation of<br />

the length of an aganglionic segment. Demonstration<br />

of the transition zone is easier if no effort<br />

is made to cleanse the bowel (Figure 17.3). In the<br />

newborn, dilatation of the proximal ganglionic<br />

bowel may not have developed and radiological<br />

diagnosis may be more difficult. The sensitivity<br />

and specificity for recognition of a transition zone<br />

have been reported to be 80% and 76%, respectively.<br />

18 The barium enema may not show a transition<br />

zone in cases of total colonic Hirschsprung’s<br />

disease, or may be indistinguishable from cases of<br />

functional constipation when ultrashort-segment<br />

Hirschsprung’s disease is present.<br />

Nevertheless, the diagnosis is based on<br />

histological evidence. Since the mid-1970s,


262<br />

Hirschsprung’s disease and intestinal neuronal dysplasias<br />

(a) (b)<br />

Figure 17.2 Anorectal manometry in a 2-month-old boy with functional constipation (a). Note that the distension of a<br />

rectal balloon with air for 1s produces a decrease of anal pressure (rectosphincter reflex). (b) Anorectal manometry in a<br />

3-month-old boy with Hirschsprung’s disease. Distension of a rectal balloon with air for 1s produces no decrease of anal<br />

pressure.<br />

Figure 17.3 Barium enema showing a long, narrowed<br />

segment in a child with Hirschsprung’s disease.<br />

demonstration of acetylcholinesterase activity in<br />

mucosal biopsies has allowed the non-invasive<br />

suction rectal biopsy technique to become the<br />

most reliable diagnostic method for<br />

aganglionosis. 7,8 The histological diagnosis is<br />

based on the demonstration of the total absence of<br />

ganglionic cells in the affected segment of the<br />

intestine, with an overgrowth of large nerve trunks<br />

in the intermuscular and submucosal zones<br />

(Figures 17.4 and 17.5). 19 Two small samples of<br />

rectal mucosa and submucosa, taken using the<br />

suction rectal biopsy technique, are sufficient for<br />

diagnosis. The two pieces must be taken not less<br />

than 2cm above the dentate line, to avoid the<br />

physiological hypoganglionic zone, and not more<br />

than 5cm above the dentate line, to avoid missing<br />

the diagnosis of a short-segment disease. Acetylcholinesterase<br />

activity in the normal colon shows<br />

only a few fibers in the lamina propria and muscularis<br />

mucosae; in Hirschsprung’s disease there is<br />

an increase in thick, knotted acetylcholinesterasepositive<br />

nerve fibers in the muscularis mucosae<br />

and lamina propria, and hypertrophied nerve<br />

trunks in the submucosa.<br />

The hyperplastic nerve trunks in the lamina<br />

propria, submucosa and muscularis propria are<br />

both adrenergic and cholinergic. Thus, these are<br />

extrinsic nerve fibers that are hyperplastic, owing


Figure 17.4 Rectal suction biopsy in a child with<br />

functional constipation. Note the presence of clusters of<br />

neurons in the submucosa and acetylcholinesterase<br />

activity showing only a few wispy fibers.<br />

Figure 17.5 Intense acetylcholinesterase activity in a<br />

patient with Hirschsprung’s disease. Note the absence of<br />

neurons and the increase in thick knotted nerve fibers in<br />

the muscularis mucosae and lamina propria. In addition,<br />

hypertrophied nerve trunks are visible in the submucosa.<br />

to the lack of intrinsic nerve cell bodies with<br />

which they can synapse. There is evidence that the<br />

obstruction can be explained on the ground of loss<br />

of NANC nerves, especially vasoactive intestinal<br />

polypeptide (VIP)-storing nerves, and an increase<br />

in sympathetic nerves containing neuropeptide<br />

Y. 20 Neuropeptide Y exerts strong contractile<br />

effects on the rectum. 21 These effects, together<br />

with the loss of VIP, are a more convincing expla-<br />

Treatment options 263<br />

nation for the loss of peristaltic activity and the<br />

contracted segment in Hirschsprung’s disease.<br />

Treatment options<br />

Treatment of Hirschsprung’s disease consists of<br />

resecting the aganglionic segment of the rectum<br />

and colon, pulling down normally innervated<br />

bowel and anastomosing this bowel at the anorectal<br />

region, while preserving the sphincter muscle.<br />

The past decade has seen an evolution in the surgical<br />

management of Hirschsprung’s disease. The<br />

previous gold standard of two- or three-stage pullthrough<br />

with a preliminary stoma has slowly been<br />

replaced by a one-stage approach in many<br />

centers. 22–24 More recently, minimally invasive<br />

approaches to the one-stage pull-through have<br />

become popular. These consist of pull-throughs<br />

utilizing laparoscopic abdominal and pelvic mobilization<br />

of the rectum and the transanal Soave<br />

procedure, which does not include any intraabdominal<br />

dissection. 25–30 The one-stage approach,<br />

either by laparotomy or by combined<br />

laparoscopy and transanal dissection, has been<br />

advocated even in the newborn period.<br />

The results of the one-stage approach in small<br />

infants appear to be at least as favorable as those in<br />

which a staged procedure with a colostomy was<br />

used. Recently, the use of the one-stage definitive<br />

procedure for small infants with Hirschsprung’s<br />

disease has increased. One-stage pull-through<br />

procedures using laparoscopy appear to be associated<br />

with shorter hospital stays, shorter time until<br />

full feeding is reached and superior cosmetic<br />

results. 31–32<br />

Follow-up<br />

Some patients with Hirschsprung’s disease<br />

continue to have problems postoperatively; this<br />

may be because of residual disease or association<br />

with neuronal dysplasia. Investigations include<br />

barium studies to delineate strictures or leaks, and<br />

further biopsy to exclude residual aganglionic<br />

bowel. If the definitive operation fails because of<br />

an impassable stricture, disruption or residual<br />

disease, further secondary surgery may be necessary<br />

and a different operation may then lead to an<br />

acceptable result.


264<br />

Hirschsprung’s disease and intestinal neuronal dysplasias<br />

Short- and long-term prognosis<br />

Data are accumulating to indicate that<br />

Hirschsprung’s disease, a disorder once known<br />

exclusively to involve an aganglionic segment of<br />

distal colon, also affects motor function in other<br />

parts of the gut. The variability in manifestations<br />

could reflect the heterogeneity of basic genetic<br />

defects now recognized as being responsible for<br />

the phenotypic expression of Hirschsprung’s<br />

disease. Abnormalities in esophageal motility are<br />

common, and duodenal motor dysfunction is<br />

present in 48% of patients. 5<br />

Miele et al have reported a systematic study of<br />

various aspects of gastrointestinal motor function<br />

in children with Hirschsprung’s disease long after<br />

removal of the aganglionic colonic segment,<br />

observing gastrointestinal symptoms, including<br />

vomiting, distension and poor growth, long after<br />

surgery. 33 Abnormalities in duodenal motor activity<br />

have also been observed in these children<br />

shortly after operation. 34<br />

Intestinal neuronal dysplasia<br />

Intestinal neuronal dysplasia (IND) or hyperganglionosis,<br />

a condition that clinically resembles<br />

Hirschsprung’s disease, was first described by<br />

Meier-Ruge in 1971. 7 It is often associated with<br />

Hirschsprung’s disease and may cause failure of<br />

clinical improvement after resectional pullthrough<br />

surgery. In 1983, Fadda et al classified IND<br />

into two clinically and histologically distinguished<br />

subtypes, called types A and B. Type A occurs in<br />

less than 5% of cases and is characterized by<br />

congenital aplasia or hypoplasia of the sympathetic<br />

innervation, presenting acutely in the<br />

neonatal period with episodes of intestinal<br />

obstruction, diarrhea and bloody stools. Type B is<br />

clinically indistinguishable from Hirschsprung’s<br />

disease: it is characterized by a malformation of<br />

the parasympathetic submucous plexus, and<br />

accounts for more than 95% of cases of isolated<br />

IND. 35<br />

The incidence of isolated IND varies from 0.3 to<br />

40% of all suction rectal biopsies. 36 The incidence<br />

varies considerably among different countries;<br />

some investigators have reported that 25–35% of<br />

patients with Hirschsprung’s disease have associ-<br />

ated IND. 35,37 However, others have rarely encountered<br />

IND in association with Hirschsprung’s<br />

disease. 38 Part of this discrepancy may be due to<br />

the persisting confusion over the essential diagnostic<br />

criteria.<br />

For a long time, IND has been diagnosed on the<br />

basis of four histological criteria applied to acetylcholinesterase-stained<br />

suction rectal biopsies. In<br />

1991, on the recommendations of a working party<br />

(the Consensus of German Pathologists), Borchard<br />

et al published diagnostic criteria for IND using a<br />

suction rectal biopsy specimen. These comprised<br />

two obligatory criteria: hyperplasia of the submucosal<br />

plexus and an increase in acetylcholinesterase-positive<br />

nerve fibers in the adventitia<br />

around submucosal blood vessels. Two<br />

additional criteria might be used: neuronal heterotopia<br />

and increased acetylcholinesterase-positive<br />

nerve fibers in the lamina propria. 39 However,<br />

concern has been expressed about whether intestinal<br />

neuronal dysplasia can be safely diagnosed by<br />

mucosal and submucosal alterations alone,<br />

without myenteric plexus abnormalities.<br />

Submucosal hyperganglionosis may reflect a<br />

normal age-related phenomenon due to immaturity,<br />

with clinical and histochemical normalization<br />

after the first year of life. Furthermore, it has been<br />

reported that most of the patients with submucosal<br />

IND have a spontaneous clinical improvement,<br />

which is sometimes associated with histological<br />

normalization. 40,41<br />

To date, submucosal intestinal neuronal dysplasia<br />

has been reported in several disorders such as<br />

intestinal malformations, meconium plug<br />

syndrome, cystic fibrosis, gastroschisis, <strong>pylori</strong>c<br />

stenosis and inflammatory processes involving the<br />

gut. The high frequency of histological ‘abnormalities’<br />

in young infants may represent a normal<br />

variant of postnatal development rather than a<br />

pathological process. Investigations using more<br />

refined and morphometric methods in rectal specimens<br />

from infants and children without bowel<br />

disease are needed to define the normal range for<br />

different ages. 41<br />

Patients with IND have been subjected to multiple<br />

types of treatment; however, the majority of<br />

patients with IND can be treated conservatively. If<br />

bowel symptoms persist after at least 6 months of<br />

conservative treatment, internal sphincter myectomy<br />

should be considered. The rapid acetyl-


cholinesterase technique has been found to be of<br />

great value in determining the extent of IND intraoperatively.<br />

42<br />

Genetic aspects<br />

Hirschsprung’s disease<br />

HSCR occurs as an isolated trait in 70% of<br />

patients, and is associated with chromosomal<br />

abnormality in 12% of cases, trisomy 21 being by<br />

far the most frequent (>90%). Additional congenital<br />

anomalies are found in 18% of cases, and<br />

include gastrointestinal malformation, cleft palate,<br />

polydactyly, cardiac septal defects and craniofacial<br />

anomalies. The higher rate of associated anomalies<br />

in familial cases than in isolated cases (39% vs.<br />

21%) strongly suggests syndromes with Mendelian<br />

inheritance. 43 Isolated HSCR appears to be a multifactorial<br />

malformation with low, sex-dependent<br />

penetrance, variable expression according to the<br />

length of the aganglionic segment, and a suggestion<br />

of involvement of one or more gene(s) with<br />

low penetrance. 44 These parameters must be taken<br />

into account for accurate evaluation of the recurrence<br />

risk in relatives. Segregation analyses<br />

suggested an oligogenic mode of inheritance in<br />

isolated HSCR. With a relative risk as high as 200,<br />

HSCR is an excellent model for the approach to<br />

common multifactorial diseases.<br />

A large number of chromosomal anomalies have<br />

been described in HSCR patients. Free trisomy 21<br />

(Down’s syndrome) is by far the most frequent,<br />

involving 2–10% of ascertained HSCR cases.<br />

Syndromes associated with HSCR can be classified<br />

as: pleiotropic neurocristopathies; syndromes with<br />

HSCR as a mandatory feature; and occasional association<br />

with recognizable syndromes. The neural<br />

crest is a transient and multipotent embryonic<br />

structure that gives rise to neuronal, endocrine and<br />

paraendocrine, craniofacial, conotruncal heart and<br />

pigmentary tissues. Neurocristopathies encompass<br />

tumors, malformations and single or multifocal<br />

abnormalities of the tissues mentioned above in<br />

various combinations. Multiple endocrine neoplasia<br />

type 2 (MEN 2) and Waardenburg syndrome are<br />

the most frequent neurocristopathies associated<br />

with HSCR. 45<br />

Waardenburg syndrome, an autosomal dominant<br />

condition, is by far the most frequent condition<br />

Genetic aspects 265<br />

combining pigmentary anomalies and sensorineural<br />

deafness, resulting from the absence of<br />

melanocytes of the skin and the stria vascularis of<br />

the cochlea. The combination of HSCR with<br />

Waardenburg syndrome defines the WS4 type<br />

(Shah–Waardenburg syndrome). Indeed, homozygous<br />

mutations of the endothelin pathway and<br />

heterozygous SOX10 mutations have been identified<br />

in WS4 patients with central nervous system<br />

involvement including seizures, ataxia and<br />

demyelinating peripheral and central neuropathies.<br />

46<br />

A wide spectrum of additional isolated anomalies<br />

have been described among HSCR cases with an<br />

incidence of sporadic types varying from 5 to<br />

30%. 47,48 No constant pattern is observed. These<br />

anomalies include distal limb, sensorineural, skin,<br />

gastrointestinal, central nervous system, genital,<br />

kidney and cardiac malformations, and facial<br />

dysmorphic features.<br />

These data highlight the importance of a careful<br />

assessment by a clinician trained in dysmorphology<br />

for all newborns diagnosed with HSCR.<br />

Skeletal X-ray and cardiac and urogenital echographic<br />

survey should be systematically performed.<br />

The observation of one additional<br />

anomaly to HSCR should prompt chromosomal<br />

studies.<br />

Molecular genetics<br />

Eight genes are known to be involved in HSCR in<br />

humans, namely the proto-oncogene RET (RET),<br />

glial cell line-derived neurotrophic factor (GDNF),<br />

neurturin (NTN), endothelin B receptor (EDNRB),<br />

endothelin 3 (EDN3), endothelin converting<br />

enzyme 1 (ECE1), SOX10 and SIP1 genes. RET and<br />

EDNRB signaling pathways were considered<br />

biochemically independent. However, an HSCR<br />

patient heterozygous for weak hypomorphic mutations<br />

in both RET and EDNRB has recently been<br />

reported. 49 Each mutation was inherited from a<br />

healthy parent. Sox10, otherwise, is involved in<br />

cell lineage determination and could be responsible<br />

of the reduced expression of EDNRB in the<br />

dom mouse.<br />

The RET signaling pathway<br />

The first observation was about an interstitial deletion<br />

of chromosome 10q11.2 in patients with TCA


266<br />

Hirschsprung’s disease and intestinal neuronal dysplasias<br />

and mental retardation. 50 The proto-oncogene RET,<br />

identified as disease-causing in MEN 2 and<br />

mapping to 10q11.2, was regarded as a good candidate<br />

gene, owing to the concurrence of MEN 2A<br />

and HSCR in some families and the expression in<br />

neural crest-derived cells. Consequently, RET gene<br />

mutations were identified in HSCR patients. 51<br />

Expression and penetrance of a RET mutation is<br />

variable and sex dependent within HSCR families<br />

(72% males and 51% females). Over 80 mutations<br />

have been identified including large deletions<br />

encompassing the RET gene, microdeletions and<br />

insertions, nonsense, missense and splicing mutations.<br />

52,53 Haploinsufficiency is the most likely<br />

mechanism for HSCR mutations. Biochemical<br />

studies showed variable consequences of some<br />

HSCR mutations (misfolding, failure to transport<br />

the protein to the cell surface, abolished biological<br />

activity).<br />

Despite extensive mutation screening, a RET mutation<br />

is identified in only 50% of familial and<br />

15–20% of sporadic HSCR cases. 54 However, most<br />

families, with a few exceptions, are compatible<br />

with linkage at the RET locus. 55<br />

Mutations in the RET ligand, such as GDNF,<br />

GFRA1-4, NTN, persephin (PSPN) and artemin<br />

(ARTN), may occur, but are not sufficient to lead to<br />

HSCR.<br />

The endothelin signaling pathway<br />

A susceptibility locus for HSCR in 13q22 was<br />

suggested for three main reasons: a significant lod<br />

score at 13q22 in a large inbred Old Order<br />

Mennonite community with multiple cases of<br />

HSCR; de novo interstitial deletion of 13q22 in<br />

several patients with HSCR; and synteny between<br />

the murine locus for piebald-lethal, a model of<br />

aganglionosis, and 13q22 in humans. Subsequently,<br />

an EDNRB missense mutation was identified<br />

in the Mennonite kindred. 56,57 Both EDNRB<br />

and EDN3 were screened in a large series of<br />

isolated HSCR patients, and EDNRB mutations<br />

were identified in approximately 5% of the<br />

patients. It is worth mentioning that the penetrance<br />

of EDN3 and EDNRB heterozygous mutations<br />

was incomplete in those HSCR patients, de novo<br />

mutations have not hitherto been observed and<br />

short HSCR (S-HSCR) is largely predominant. 58<br />

SOX10<br />

The last de novo mouse model for WS4 in humans<br />

is dominant megalon (Dom). The Dom gene is<br />

SOX10, a member of the sex-determining factor<br />

(SRY)-like, high mobility group (HMG) DNA<br />

binding proteins. Subsequently, heterozygous<br />

SOX10 mutations have been identified in familial<br />

and isolated patients with WS4 (including de novo<br />

mutation) with high penetrance. 59<br />

Intestinal neuronal dysplasia<br />

Studies have been performed to investigate the<br />

potential role of HSCR-associated RET, GDNF,<br />

EDNRB and EDN3 genes in the development of<br />

IND. They demonstrated that only three RET<br />

mutation were detected in patients with HSCR, no<br />

mutation in this gene was observed in IND and<br />

mixed HSCR/IND patients, HSCR and HSCR/IND<br />

patients showed over-representation of a specific<br />

RET polymorphism in exon 2, while IND patients<br />

exhibited a significantly lower frequency of the<br />

same polymorphism comparable with that of<br />

controls. These findings may suggest that IND is<br />

genetically different from HSCR.<br />

A homozygous mutation of the EDNRB gene in spotting<br />

lethal (sl/sl) rats leads to the HSCR phenotype<br />

with long segmented aganglionosis. The heterozygous<br />

(+/sl) EDNRB-deficient rats revealed more<br />

subtle abnormalities of the enteric nervous system:<br />

the submucous plexus was characterized by a significantly<br />

increased ganglionic size and density, and<br />

the presence of hypertrophied nerve fiber strands,<br />

resembling the histopathological criteria for IND.<br />

Other animal models, such as Ncx/Hox11L.1-deficient<br />

mice, suggest that many other genes could be<br />

involved in the pathogenesis of IND. 60


REFERENCES<br />

1. Taviras S, Pachinis V. Development of the mammalian<br />

enteric system. Curr Opin Genet Dev 1999; 9: 321.<br />

2. Lyonnet S, Bolino A, Pelet A et al. A gene for<br />

Hirschsprung disease maps to the proximal long arm of<br />

chromosome 10. Nat Genet 1993; 4: 346–350.<br />

3. Gershon MD, Chalazonitis A, Rothman TP. From neural<br />

crest to bowel: development of the enteric nervous<br />

system. J Neurobiol 1993; 24: 199–214.<br />

4. Badner JA, Sieber Wk, Garver KL et al. A genetic study<br />

of Hirschsprung’s disease. Am J Hum Genet 1990; 46:<br />

568–580.<br />

5. Staiano A, Corazziari E, Andreotti MR, Clouse RE.<br />

Esophageal motility in children with Hirschsprung’s<br />

disease. Am J Dis Child 1991; 145: 310–313.<br />

6. Larsson LT, Shen Z, Ekbland E et al. Lack of neuronal<br />

nitric oxide synthase in nerve fibers of aganglionic<br />

intestine: a clue to Hirschsprung’s disease. J Pediatr<br />

Gastroenterol Nutr 1995; 20: 49–53.<br />

7. Meier-Ruge W. Uber Ein Erkrankungsbild des colon mit<br />

Hirschsprung-Symptomatik. Vehr Dtsch Ges Pathol 1971;<br />

55: 506–510.<br />

8. Lake BD, Puri P, Nixon HH, Claireaux AE.<br />

Hirschsprung’s disease. An appraisal of histochemically<br />

demonstrated acetylcholinesterase activity in suction<br />

rectal biopsy specimens as an aid to diagnosis. Arch<br />

Path Lab Med 1978; 26: 288–291.<br />

9. Romanska HM, Bishop AE, Brereton RJ et al. Increased<br />

expression of muscular neural cell adhesion molecule<br />

in congenital aganglionosis. Gastroenterology 1993; 105:<br />

1104–1109.<br />

10. Vanderwinden JM, De Laet MH, Schiffmann SN et al.<br />

Nitric oxide synthase distribution in the enteric nervous<br />

system of Hirschsprung’s disease. Gastroenterology<br />

1993; 105: 969–973.<br />

11. Vanderwinden JM, Rumessen JJ, Liu H et al. Interstitial<br />

cells of cajal in human colon and in Hirschsprung’s<br />

disease. Gastroenterology 1996; 111: 901–910.<br />

12. Coran AG, Teitelbaum DH. Recent advances in management<br />

of Hirschsprung’s disease. Am J Surg 2000; 180:<br />

382–387.<br />

13. Bill JAH, Chapman ND. The enterocolitis of<br />

Hirschsprung’s disease: its natural history and treatment.<br />

Am J Surg 1962; 103: 70–74.<br />

14. Elhalaby EA, Coran AG, Blane CE et al. Enterocolitis<br />

associated with Hirschsprung’s disease: a clinical–radiological<br />

characterization based on 168 patients. J Pediatr<br />

Surg 1995; 30: 1023–1027.<br />

15. Swenson O, Fisher JH. Hirschsprung’s disease during<br />

infancy. Surg Clin North Am 1956; 36: 115–122.<br />

16. Marty TL, Matlak ME, Hendrickson M et al. Unexpected<br />

death from enterocolitis after surgery for Hirschsprung’s<br />

disease. Pediatrics 1995; 96: 118–121.<br />

17. Loening-Baucke V. Modulation of abnormal defecation<br />

dynamics by biofeedback treatment in chronically<br />

constipated children with encopresis. J Pediatr 1990;<br />

116: 214–222.<br />

18. Taxman TI, Yulish BS, Rothstein FC. How useful is<br />

barium enema in diagnosis of infantile Hirschsprung’s<br />

disease? Am J Dis Child 1986; 140: 881–884.<br />

19. Aldridge RT, Campbell PE. Ganglion cells distribution<br />

in the normal rectum and anal canal: a basis for diagnosis<br />

of Hirschsprung’s disease by anorectal biopsy. J<br />

Pediatr Surg 1968; 3: 475–489.<br />

20. Milla PJ. Regulatory gut peptides in intestinal neuronal<br />

dysplasia and Hirschsprung’s disease. In<br />

Hadziselimovic F, Herzog B, eds. Falk Symposium 65.<br />

References 267<br />

Inflammatory Bowel Disease and Morbus Hirschsprung.<br />

Dordrecht: Kluwer Academic Publishers, 1992: 239–250.<br />

21. Hellstrom PM, Lundberg JM, Holfelt T, Goldstein M.<br />

Neuropeptide Y, peptide YY and sympathetic control of<br />

rectal tone and anal canal pressure in the cat. Scand J<br />

Gastroenterol 1989; 24: 231–243.<br />

22. Langer JC, Fitzgerald PG, Winthrop AL et al. One vs two<br />

stage Soave pull-through for Hirschsprung’s disease in<br />

the first year of life. J Pediatr Surg 1996; 31: 33–37.<br />

23. Pierro A, Fasoli L, Kiely EM et al. Staged pull-through<br />

for rectosigmoid Hirschsprung’s disease is not safer<br />

than primary pull-through. J Pediatr Surg 1997; 32:<br />

505–509.<br />

24. Hackam DJ, Superina RA, Pearl RH. Single stage repair<br />

of Hirschsprung’s disease: a comparison of 109 patients<br />

over 5 years. J Pediatr Surg 1997; 32: 1028–1031.<br />

25. Jona JZ, Cohen RD, Georgeson KE et al. Laparoscopic<br />

pull-through procedure for Hirschsprung’s disease.<br />

Semin Pediatr Surg 1998; 7: 228–231.<br />

26. Smith BM, Stainer RB, Lobe TE. Laparoscopic Duhamel<br />

pull-through procedure for Hirschsprung’s disease in<br />

Childhood. J Laparoendosc Surg 1994; 4: 273–276.<br />

27. Curran TJ, Raffensperger JG. Laparoscopic Swenson<br />

pull-through: a comparison with the open procedure. J<br />

Pediatr Surg 1996; 31: 1155–1156.<br />

28. De La Torre- Mondragon L, Ortega-Salgado JA.<br />

Transanal endorectal pull-through for Hirschsprung’s<br />

disease. J Pediatr Surg 1998; 33: 1283–1286.<br />

29. Langer JC, Minkes RK, Mazziotti MV et al. Transanal<br />

one-stage Soave procedure for infants with<br />

Hirschsprung’s disease. J Pediatr Surg 1999; 34:<br />

148–152.<br />

30. Albanese CT, Jennings RW, Smith B et al. Perineal onestage<br />

pull-through for Hirschsprung’s disease. J Pediatr<br />

Surg 1999; 34: 377–380.<br />

31. Langer JC, Scifert M, Mikes RK. One stage Soave pullthrough<br />

for Hirschsprung’s disease. A comparison of the<br />

transanal and open approach. J Pediatr Surg 2000; 35:<br />

820–822.<br />

32. Teeraratkul S. Transanal one stage endorectal pullthrough<br />

for Hirschsprung’s disease in infants and children.<br />

J Pediatr Surg 2003; 38: 184–187.<br />

33. Miele E, Tozzi A, Staiano A et al. Persistence of abnormal<br />

gastrointestinal motility operation for<br />

Hirschsprung’s disease. Am J Gastroenterol 2000; 95:<br />

1226–1230.<br />

34. Di Lorenzo C, Flores AF, Reddy SN et al. Small bowel<br />

neuropathy in symptomatic children after surgery for<br />

Hirschsprung’s disease. Gastroenterology 1997; 112:<br />

783A.<br />

35. Fadda B, Meier WA, Meier-Ruge W et al. Neuronale<br />

intestinale Dysplasie: Eine Kritische 10-Jahres-Analyse<br />

Klinischer und Bioptischer Diagnostik. Z Kinderchir<br />

1983; 38: 305–311.<br />

36. Smith VV. Isolated intestinal neuronal dysplasia: a<br />

descriptive pattern or a distinct clinicopathological<br />

entity? In Hadziselimomic F, Herzog B, eds.<br />

Inflammatory Bowel Disease and Morbus Hirschsprung.<br />

Dordrecht, The Netherlands: Kluwer Academic, 1992:<br />

203–214.<br />

37. Kobayashi H, Hirakawa H, Surana R et al. Intestinal<br />

neuronal dysplasia is a possible cause of persistant<br />

bowel symptoms after pull-through operation for<br />

Hirschsprung’s disease. J Pediatr Surg 1995; 30:<br />

253–259.


268<br />

Hirschsprung’s disease and intestinal neuronal dysplasias<br />

38. Fadda B, Pistor G, Meier-Ruge W et al. Symptoms,<br />

diagnosis and therapy of neuronal intestinal dysplasia<br />

masked by Hirschsprung’s disease. J Pediatr Surg 1987;<br />

2: 76–80.<br />

39. Borchard F, Meier-Ruge W, Wiebecke B et al.<br />

Innervationsstrunger des Dickdarms – Klassifikation<br />

und Diagnostik. Pathologe 1991; 12: 171–174.<br />

40. Cord-Udy CL, Smith VV, Ahmed S et al. An evaluation<br />

of the role of suction rectal biopsy in the diagnosis of<br />

intestinal neuronal dysplasia. J Pediatr Gastroenterol<br />

Nutr 1997; 24: 1–6.<br />

41. Koletzko S, Jesh I, Faus- Kebler T et al. Rectal biopsy for<br />

diagnosis of intestinal neuronal dysplasia in children: a<br />

prospective study on interobserver variation and<br />

clinical outcome. Gut 1999; 44: 856–861.<br />

42. Kobayashi H, O’Briain S, Hirakawa H et al. A rapid<br />

tecnique for acetylcholinesterase staining. Arch Pathol<br />

Lab Med 1994; 118: 1127–1129.<br />

43. Brooks AS, Breuning MH, Meijers C. Spectrum of<br />

phenotypes associated with Hirschsprung disease: an<br />

evaluation of 239 patients from a single institution. The<br />

Third International Meeting: Hirschsprung Disease and<br />

the Correlated Neurocristopathies. France: Evian, 1998<br />

44. Puffenberger EG, Kauffman ER, Bolk S et al. Identity-bydescent<br />

and association mapping of a recessive gene for<br />

Hirschsprung disease on human chromosome 13q22.<br />

Hum Mol Genet 1994; 3: 1217–1225.<br />

45. Decker RA, Peacock ML, Watson P. Hirschsprung disease<br />

in MEN 2A: increased spectrum of RET exon 10 genotypes<br />

and strong genotype–<br />

phenotype correlation. Hum Mol Genet 1998; 7:<br />

129–134.<br />

46. Edery P, Attie T, Amiel J et al. Mutation of the<br />

endothelin-3 gene in the Waardenburg–Hirschsprung<br />

disease (Shah–Waardenburg syndrome). Nat Genet<br />

1996; 12: 442–444.<br />

47. Sarioglu A, Tanyel FC, Buyukpamukcu N, Hicsonmez<br />

A. Hirschsprung-associated congenital anomalies. Eur J<br />

Pediatr Surg 1997; 7: 331–337.<br />

48 Auricchio A, Griseri P, Carpentieri ML et al. Double<br />

heterozygosity for a RET substitution interfering with<br />

splicing and an EDNRB missense mutation in<br />

Hirschsprung disease. Am J Hum Genet 1999; 64:<br />

1216–1221.<br />

49. Martucciello G, Biocchini M, Dodero P et al. Total<br />

colonic aganglionosis associated with interstitial<br />

deletion of the long arm of chromosome 10. Pediatr Surg<br />

Int 1992; 7: 308.<br />

50. Edery P, Lyonnet S, Mulligan LM et al. Mutations of the<br />

RET proto-oncogene in Hirschsprung’s disease. Nature<br />

1994; 367: 378–380.<br />

51. Angrist M, Bolk S, Thiel B et al. Mutation analysis of<br />

the RET receptor tyrosine kinase in Hirschsprung<br />

disease. Hum Mol Genet 1995; 4: 821–830.<br />

52. Seri M, Yin L, Barone V et al. Frequency of RET<br />

mutations in long- and short-segment Hirschsprung<br />

disease. Hum Mutat 1997; 9: 243–249.<br />

53. Attie T, Pelet A, Edery P et al. Diversity of RET protooncogene<br />

mutations in familial and sporadic<br />

Hirschsprung disease. Hum Mol Genet 1995; 4:<br />

1381–1386.<br />

54. Bolk S, Pelet A, Hofstra RM et al. A human model for<br />

multigenic inheritance: phenotypic expression in<br />

Hirschsprung disease requires both the RET gene and a<br />

new 9q31 locus. Proc Natl Acad Sci USA 2000; 97:<br />

268–273.<br />

55. Borrego S, Ruiz A, Saez ME et al. RET genotypes<br />

comprising specific haplotypes of polymorphic variants<br />

predispose to isolated Hirschsprung disease. J Med<br />

Genet 2000; 37: 572–578.<br />

56. Kiss P, Orsztovics M. Association of 13q deletion and<br />

Hirschsprung’s disease. J Med Genet 1989; 26: 793–794.<br />

57. Puffenberger EG, Hosoda K, Washington SS et al. A<br />

missense mutation of endothelin-B receptor gene in<br />

multigenic Hirschsprung’s disease. Cell 1994; 30:<br />

1257–1266.<br />

58. Auricchio A, Casari G, Staiano A, Ballabio A.<br />

Endothelin-B receptor mutations in patients with<br />

isolated Hirschsprung disease from a non-inbred<br />

population. Hum Mol Genet 1996; 5: 351–354.<br />

59. Southard-Smith EM, Angrist M, Eleison JS et al. The<br />

Sox10 (Dom) mouse: modeling the genetic variation of<br />

Waardenburg–Shah (WS4) syndrome. Genom Res 1999;<br />

9: 215–225.<br />

60. Yamataka A, Datano M, Kobayashi H et al. Intestinal<br />

neuronal displasia-like pathology in Ncx/Hox11L.1<br />

deficient mice. J Pediatr Surg 2001; 36: 1293–1296.<br />

Additional educational resources<br />

www.1uphealth.com/health/hirschsprung_disease_info.htm<br />

www.bdid.com/cam.htm<br />

www.anatomy.med.unsw.edu.au/ebl/embryo/OMIM


18<br />

Introduction<br />

Chronic intestinal<br />

pseudo-obstruction in<br />

childhood<br />

Peter J Milla<br />

Chronic intestinal pseudo-obstruction (CIP) is a<br />

clinical syndrome characterized by signs of intestinal<br />

obstruction without mechanical occlusion of<br />

the gut. A number of other names have been used<br />

to described the disorder, including adynamic<br />

bowel, Hirschsprung’s disease, megacystis, microcolon<br />

hypoperistalsis syndrome, visceral neuropathies<br />

and visceral myopathies. Initially, it was<br />

thought that CIP was a single disease entity<br />

without clearly defined pathology. Over the past<br />

20 years, however, numerous reports have<br />

appeared which show that this is not the case. 1–4<br />

CIP is a heterogeneous disorder associated with a<br />

wide variety of pathologies, some intrinsic to the<br />

gut, others multisystem disorders involving the gut<br />

or altering the environment within which the gut<br />

operates. The conditions cause disordered intestinal<br />

motor activity and may present acutely or<br />

chronically. The most common forms of pseudoobstruction<br />

are acute in nature and occur as postoperative<br />

ileus or the ileus that is associated with<br />

electrolyte imbalance and metabolic disorder.<br />

Acute pseudo-obstruction episodes have also been<br />

reported in association with food-sensitive intestinal<br />

disease such as celiac disease and cow’s milk<br />

protein intolerance, together with drugs such as<br />

vincristine. CIP was originally thought to involve<br />

only the small intestine, but it is now realized that<br />

it may be restricted to one region of the gut, such<br />

as in achalasia or Hirschsprung’s disease, or it may<br />

be present diffusely throughout the gastrointestinal<br />

tract.<br />

Clinical presentation<br />

A child may present with either the primary or the<br />

secondary effects of the underlying disease.<br />

However, the clinical symptoms are variable and<br />

often non-specific. The location of the affected<br />

bowel, diffuse or regional, seems a more important<br />

determinant of the clinical presentation than<br />

the underlying disease. A child may be obstructed<br />

or complain of severe constipation depending<br />

upon whether the small intestine and colon or just<br />

the colon is affected. In addition, those with<br />

urinary tract involvement may initially present<br />

with acute or chronic urinary retention. These<br />

effects quite clearly are the result of the ability of<br />

the underlying disease of the neuromusculature of<br />

the gut to produce ordered motor activity.<br />

However, the effects of the underlying disease<br />

may also be secondary, such as the consequences<br />

of bacterial overgrowth, fecal impaction or adhesional<br />

obstruction associated with previous<br />

surgery. In neuropathic disorders the consequences<br />

of denervation may not only be on motor<br />

activity but also on secretomotor control and<br />

sensation. These effects may result in diarrhea or<br />

visceral hyperalgesia.<br />

Although initial reports of CIP were in adults, it is<br />

now apparent that it probably occurs more<br />

commonly in infants. Indeed, when the presentation<br />

and mortality of cases published in the literature<br />

are considered, it is found to occur most<br />

commonly in children, with high morbidity and<br />

mortality. Considering these data 5 together with a<br />

large series from one center, 1 the conditions in<br />

childhood present most commonly during infancy,<br />

either in the neonatal period, or under the age of 1<br />

year, during which time the highest mortality rates<br />

are suffered. In most cases an underlying abnormality<br />

of either smooth muscle or of enteric nerves<br />

is found when adequately sought. The clinical<br />

manifestation at presentation depends on both age<br />

and the type and extent of the condition affecting<br />

the neuromusculature of the gut.<br />

269


270<br />

Chronic intestinal pseudo-obstruction in childhood<br />

Before birth<br />

CIP may first be recognized before birth, either as<br />

part of a routine antenatal ultrasound scan or in<br />

the investigation of a mother with polyhydramnios.<br />

On abdominal ultrasound examination the<br />

fetus may have either dilated loops of bowel or a<br />

distended bladder, or both.<br />

Birth and the neonatal period<br />

The majority of children with CIP present either at<br />

birth or in the neonatal period. 1 In all series<br />

approximately half the infants have symptoms at<br />

birth or within the first few days of life. In those<br />

who present at birth, the labor and delivery are<br />

frequently difficult, owing to an already distended<br />

abdomen. After birth there is abdominal distension,<br />

failure to pass meconium and bilious<br />

vomiting. The abdominal distension is due to<br />

swallowed air, which distends and dilates the<br />

small bowel, but is not passed further through the<br />

gut. Contrast studies may show the presence of a<br />

microcolon or a short small intestine or, in approximately<br />

34%, a malrotation. In some there may be<br />

a specific clinical syndrome of a congenitally short<br />

small intestine, <strong>pylori</strong>c stenosis and a malrotation.<br />

6,7 In addition to gastrointestinal symptoms,<br />

there may also be failure to pass urine, megacystis<br />

and hydroureter or hydronephrosis.<br />

Incomplete bladder emptying often results in<br />

recurrent urinary tract infection, and this may<br />

completely overshadow the gastrointestinal<br />

symptoms.<br />

Later infancy<br />

After the neonatal period, although infants may<br />

present with an acute obstruction, the symptoms<br />

are often intermittent or slowly progressive.<br />

Initially some infants may appear completely<br />

healthy, taking breast feeds normally, but then<br />

suddenly develop an episode of obstruction<br />

following what appears to be an intercurrent<br />

enteric infection. In these infants persistence of<br />

vomiting for more than 7 days warrants further<br />

investigation in a patient who has been thought to<br />

have had acute gastroenteritis, as they may have<br />

developed an acute myositis, which is potentially<br />

treatable. 8 In other such infants, who have fed<br />

normally from birth, episodes of obstruction begin<br />

only when more complex foods are introduced<br />

into the diet and attempts are made at weaning.<br />

Urinary symptoms due to involvement of the<br />

urinary tract may continue to present for the first<br />

time during infancy.<br />

Childhood<br />

In later childhood the initial presentation may<br />

continue to indicate obstruction, but may simply<br />

be constipation, which becomes intractable.<br />

Severe abdominal pain may also occur, owing to<br />

distension of the bowel or as part of visceral hyperalgesia.<br />

In those with a distended abdomen, bowel<br />

sounds may be totally absent or very markedly<br />

reduced. If high-pitched bowel sounds are present,<br />

or if there is visible peristalsis, the distension of<br />

the gut is more likely to be secondary to a mechanical<br />

rather than a functional obstruction.<br />

Investigation<br />

In order to understand the pseudo-obstructive<br />

disorder and plan rational treatment, there are<br />

three steps in diagnosis: definition of the presence<br />

of functional obstruction. definition of the areas<br />

involved and the physiology of the affected areas;<br />

and delineating the disease process causing the<br />

functional obstruction.<br />

CIP is due to disordered motor activity, and this<br />

results from disturbance of the control mechanisms<br />

of motor activity and disease of the smooth<br />

muscle coats. This may occur as a consequence of<br />

primary disease of the gut motor apparatus or<br />

because of involvement of the neuromusculature<br />

of the gut secondary to disease, which either<br />

affects the gut as part of a multisystem disease or<br />

is primarily elsewhere. It is useful to consider that<br />

the obstruction may be caused at different levels of<br />

control of gut motor activity: the end-organ smooth<br />

muscle; the enteric nervous system; the humoral<br />

environment provided by gut endocrine cells<br />

or immunocytes; or the extrinsic innervation.<br />

Disruption at one or all of these levels results in<br />

the lack of effective co-ordinated propulsive<br />

movement. Initial investigations were designed to<br />

demonstrate this.


Radiology and transit studies<br />

Plain abdominal X-rays and abdominal ultrasound<br />

examinations may show the presence of large<br />

distended loops of gut, but it is difficult using such<br />

studies to define the area of the gut involved.<br />

Conventional contrast radiography will delineate<br />

anatomical abnormalities and together with<br />

studies by radioisotope and computed tomography<br />

(CT) scanning, may provide a measure of transit<br />

time and/or demonstrate disordered peristalsis.<br />

Limited descriptions of transit can be obtained<br />

using breath hydrogen or radio-opaque pellets and<br />

plain abdominal X-ray for whole bowel transit<br />

times. All of these methods provide a limited<br />

description of the disease but no clues to the<br />

nature of the disorder.<br />

Surface electrogastrography<br />

Electrogastrography (EGG) is defined as the<br />

recording of myoelectric activity of the smooth<br />

muscle of the stomach by means of electrodes<br />

attached to the abdominal skin surface. EGG was<br />

first devised by Alvarez in the 1920s using<br />

mechanical means of recording from a string<br />

galvanometer. The advent of powerful personal<br />

computers and the development of signal processing<br />

algorithms has allowed such data to be objectively<br />

analyzed. A great advantage of the methodology<br />

is that it is non-invasive and readily detects<br />

disturbance of gastric antral slow waves and, by<br />

suitable positioning, duodenal slow waves. This<br />

method has been used in patients with diffuse CIP<br />

to detect abnormal myoelectric activity. Persistent<br />

entral dysrhythmias have been found in the fasting<br />

state in both myopathic and neuropathic disorders.<br />

9<br />

Manometry<br />

Motor activity may be studied by measurement of<br />

intraluminal pressure changes and known<br />

manometry. This is helpful in delineating both the<br />

extent of the disordered motility and possibly the<br />

disease processes causing the disorder. In patients<br />

with suspected functional obstruction at least<br />

three areas of the gastrointestinal tract should be<br />

studied: the esophagus, the upper small intestine<br />

and the left colon, as the disease process may not<br />

Investigation 271<br />

be restricted to any one of these areas. In these<br />

areas swallow-induced peristalsis, fasting small<br />

intestinal motor activity and the gastrocolonic<br />

response to food or the induction of highamplitude<br />

propagated contraction by bisacodyl<br />

can be used as tests of the integrity of the enteric<br />

nervous system and of the contractile activity of<br />

the smooth muscle.<br />

Esophagus<br />

Swallow-induced peristalsis and the associated<br />

relaxation of the lower esophageal sphincter can<br />

be studied using a Dent sleeve assembly modified<br />

for use in infants or young children. 10 Particular<br />

attention should be paid to the nature of the<br />

primary peristaltic sequence and whether<br />

secondary peristalsis occurs in response to reflux.<br />

The presence of tertiary contractions and the<br />

amplitude and form of the contractile waves<br />

should also be noted, as should the behavior of the<br />

lower esophageal sphincter.<br />

Small intestine<br />

Most studies of intestinal motor activity in children<br />

with CIP have utilized small intestinal<br />

manometry. The cyclical nature of fasting small<br />

intestinal motor activity is determined by the<br />

inherent activity of the enteric nervous system.<br />

This intrinsic property can be used to test the<br />

integrity of the enteric nervous system and<br />

whether extrinsic nervous modulation is present.<br />

Disruption of fasting activity and the establishment<br />

of postprandial activity results from the<br />

humorally mediated responses to food and clarifies<br />

whether enteroenteric responses are intact. In<br />

addition, these responses can be further tested by<br />

utilizing the motilin agonists erythromycin and<br />

somatostatin. The algorithm that the author uses to<br />

test motor functioning in pseudo-obstruction is<br />

shown in Figure 18.1. Some studies have shown<br />

that myopathic processes produce low-amplitude<br />

poorly propagated contractions5,11 whereas neuropathic<br />

processes are associated with contractions<br />

of normal amplitude which are bizarre in wave<br />

form, abnormally propagated and, in phase 3 activity<br />

ill formed. 11,12 In addition, clustered phasic<br />

activity in phase 2 is often present. Disturbance of<br />

the neuroendocrine environment can also be<br />

shown where there are changes in frequency of


272<br />

Chronic intestinal pseudo-obstruction in childhood<br />

Radiology<br />

Transit study<br />

Rectal suction biopsy<br />

Electrogastrography<br />

Antroduodenal manometry<br />

+ve<br />

+ve<br />

Figure 18.1 Investigation of intestinal pseudo-obstruction.<br />

slow-wave activity and in frequency of phase 3<br />

contractions. In conditions where there is<br />

increased secretion of catecholamines such as<br />

hyperthyroidism, ganglioneuroma and pheochromocytoma<br />

there is increased frequency of slowwave<br />

rhythm; in preterm infants and hypothyroidism,<br />

there is decreased slow-wave<br />

frequency.<br />

Colon<br />

It is often useful to record simultaneously leftsided<br />

colonic motor activity and post-prandial<br />

small intestinal motility as a feed will also induce<br />

increased rectosigmoid contractions. In addition,<br />

the response of the descending colon to bisacodyl<br />

Plain abdominal X-ray<br />

Contrast study<br />

Hirschsprung's disease surgery<br />

Ileostomy and<br />

histology<br />

Antroduodenal manometry<br />

-ve<br />

Radio-opaque pellet transit study<br />

Rectal biopsy<br />

Full thickness<br />

-ve +ve<br />

Colonic Ileostomy<br />

manometry or colostomy<br />

Ileostomy and histology<br />

in inducing a high-amplitude propagated contraction<br />

can be demonstrated, and also whether this<br />

propagated contraction halts in the sigmoid colon.<br />

Thus, information regarding smooth muscle, local<br />

enteric nerves and those involved in the gastrocolonic<br />

response and colocolonic reflexes can be<br />

obtained. 13<br />

Etiology<br />

The disease processes, which result in CIP, affect<br />

the control mechanisms of intestinal motility. The<br />

disorders and disease may primarily be of the<br />

intrinsic enteric nerves with or without involvement<br />

of the extrinsic autonomic nerves, the


smooth muscle cells themselves or of the tumoral<br />

and endocrine environment. Good examples of the<br />

effect of disturbance of the endocrine environment<br />

are the ileus associated with vasoactive intestinal<br />

polypeptide-secreting tumors such as ganglioneuromas,<br />

and the constipation caused by<br />

hypothyroidism. These conditions will not be<br />

considered further here. A variety of diseases and<br />

drugs, which are listed in Table 18.1, may secondarily<br />

cause CIP. Such secondary disorders are<br />

much more common in adult life than in childhood.<br />

Primary disease of the gut neuromusculature<br />

may be due to disease of the enteric nerves or<br />

the smooth muscle cells or, at least theoretically, of<br />

the interstitial cells of Cajal. Surprisingly, there are<br />

virtually no descriptions of the primary pathology<br />

of interstitial cells of Cajal, which is said to occur<br />

in piebaldism, but there have been descriptions of<br />

alterations of these cells in hypertrophic <strong>pylori</strong>c<br />

stenosis and Hirschsprung’s disease. 14,15 Definite<br />

diagnosis of neuromuscular disease requires<br />

careful study of the full thickness of the gastrointestinal<br />

wall in optimally orientated pieces of gut.<br />

Table 18.1 Miscellaneous conditions<br />

causing intestinal pseudo-obstruction<br />

Endocrine disorders<br />

Hypothyroidism<br />

Hypoparathyroidism<br />

Pheochromocytoma<br />

Carcinoid<br />

Ganglioneuroblastoma/neuroma<br />

Metabolic disorders<br />

Uremia<br />

Porphyria<br />

Amyloidosis<br />

Drugs<br />

Antidepressants and anti-anxiety drugs<br />

Anticholinergic agents<br />

Opiates<br />

Anticonvulsive cytotoxic drugs<br />

Toxic agents<br />

Alcohol in fetal alcohol syndrome<br />

Irradiation<br />

Etiology 273<br />

It is best carried out in centers that have experience<br />

of both the disease and the histopathological<br />

techniques required.<br />

Primary visceral myopathies<br />

Primary disorders of the intestinal smooth muscle<br />

coats may represent abnormalities in morphogenesis<br />

or intrinsic myocyte defects. 4 Abnormalities of<br />

morphogenesis of the muscle coats may result in<br />

the presence of an additional muscle coat or the<br />

absence of a muscle layer. This may occur<br />

diffusely throughout the gut or may only involve<br />

only a segment of the gut. Clinically, primary<br />

visceral myopathies occur as either familial<br />

genetic diseases with a defined mode of inheritance<br />

or, more often, as sporadic cases. Three types<br />

of familial visceral myopathy have been described<br />

in adults, which all initially presented in the<br />

second decade and should therefore be considered<br />

when symptoms begin during adolescence.<br />

Familial visceral myopathy with diffuse<br />

abnormal muscle layering<br />

This condition usually presents at birth, or shortly<br />

after, with functional obstruction. Investigation<br />

shows the presence of a short gut and a mid-gut<br />

malrotation. Three related males have been<br />

described with this condition (the index case, his<br />

half brother from the same mother, and his maternal<br />

uncle), suggesting an X-linked mode of inheritance.<br />

4,6 The most striking histological finding was<br />

an extra circular muscle layer with the myenteric<br />

enteric plexus embedded within it. None of the<br />

patients tolerated enteral feeding, all required<br />

long-term parenteral nutrition and all died with<br />

septic complications of parenteral nutrition. One<br />

further sporadic case has since been described,<br />

also in a male child, aged 5 years, who has done<br />

well since birth with a decompression ileostomy<br />

and mixed enteral and parenteral nutrition.<br />

Infantile visceral myopathy<br />

Most cases occur sporadically but some families<br />

have been described with either a dominant gene<br />

with variable expressivity or autosomal recessive<br />

mode of inheritance. Most affected children<br />

develop symptoms at birth or within the first year


274<br />

Chronic intestinal pseudo-obstruction in childhood<br />

of life often with severe constipation and<br />

abdominal distension. Failure to thrive and malnutrition<br />

are common. When recurrent episodes of<br />

functional obstruction occur, long-term parenteral<br />

nutrition is necessary. Usually the entire gastrointestinal<br />

tract is affected, and almost all patients<br />

have involvement of the urinary tract with megaureter<br />

and megacystis.<br />

Smith and Milla, 4 studying 27 patients with<br />

smooth muscle disease, identified five histological<br />

phenotypes in full-thickness biopsies from such<br />

children using routine microscopy, histochemistry,<br />

immunocytochemistry (with monoclonal antibodies<br />

against different neural and smooth muscle<br />

markers) and electron microscopy. In two of the 27<br />

patients, a severe myositis was found. This is<br />

considered separately. Twenty-five had a primary<br />

myopathic disorder which, on routine microscopy<br />

of paraffin sections of the smooth muscle, was<br />

abnormal in only 15. In the remaining patients<br />

myopathic changes would have been missed<br />

without application of other techniques, particularly<br />

electron microscopy.<br />

These authors, like others, found the presence of<br />

gross fibrosis of the muscle layers and profound<br />

atrophy of the smooth muscle cells similar to that<br />

seen in adults with familial and sporadic forms of<br />

visceral myopathy. They were able to relate these<br />

changes to intrinsic myocyte defects or changes in<br />

the extracellular matrix. A myopathy with<br />

increased autophagic activity could sometimes be<br />

detected using histochemistry with increased acid<br />

phosphatase activity in the smooth muscle cells<br />

due to active lysozymes. However, but often electron<br />

microscopy was required, as the condition<br />

appeared to be active only in the central portions<br />

of the smooth muscle cells. A further and distinct<br />

myopathic subtype was found where there was a<br />

so-called pink blush in the circular muscle layer<br />

with nuclear crowding, possibly due to a disorganization<br />

of the muscle cells. On electron<br />

microscopy, the smooth muscle cells appeared to<br />

be separated from each other by material secreted<br />

either by the muscle cells themselves or the extracellular<br />

matrix. In patients with diffuse disease the<br />

prognosis is poor, with about 40% of them dying<br />

during childhood from the complications of<br />

malnutrition or long-term parenteral nutrition. 1<br />

Those who tolerate some oral feeding generally<br />

survive into adult life.<br />

Megacystis microcolon hypoperistalsis<br />

syndrome<br />

This condition causes severe CIP and affects<br />

predominantly female infants. 5,16 Most cases<br />

present antenatally. Ultrasound scans show the<br />

presence of megacystis hydronephrosis and<br />

distended bowel. In addition, the proximal small<br />

bowel is often short and there is a malrotated midgut<br />

with microcolon located entirely on the left<br />

side of the abdomen. The myopathy causing the<br />

problem appears to be a degenerative condition of<br />

smooth muscle cells. Whilst most cases are<br />

sporadic, others appear to have been inherited in<br />

an autosomal recessive mode.<br />

After birth, the affected infants develop massive<br />

abdominal distension, which is partly due to<br />

distended bowel and partly due to a distended<br />

bladder. Patients require a decompression<br />

ileostomy and either a vesicostomy or frequent<br />

catheterization to ensure that the urinary tract is<br />

decompressed. Almost all children require longterm<br />

parenteral nutrition for survival and, if this<br />

becomes problematic, small intestinal transplantation.<br />

Megacystis microcolon hypoperistalsis<br />

syndrome must be differentiated from prune belly<br />

syndrome, which is due to early intrauterine<br />

urethral obstruction and affects predominantly<br />

male infants. They present with a dilated<br />

abdomen, constipation, megacystis and hydronephrosis.<br />

Sometimes there is an intestinal malrotation<br />

present, but there is no hypoperistalsis and<br />

no microcolon.<br />

Familial visceral myopathies types 1, 2 and 3<br />

These conditions usually present in later childhood<br />

and have a number of distinctive features.<br />

Familial visceral myopathy type 1 presents as a<br />

megaduodenum and is inherited as an autosomal<br />

dominant trait with a female predominance in<br />

frequency and severity. 17 The disease is characterized<br />

by foregut functional obstruction, although<br />

gastric emptying is often normal, and an elongated,<br />

redundant, usually dilated colon. Barium<br />

studies show an aperistaltic esophagus, sometimes<br />

normal gastric emptying but a flaccid and dilated<br />

duodenum with prolonged retention of barium.<br />

The bladder is affected in about half the cases, but<br />

is often asymptomatic. 17 Schuffler and Pope


showed that there was marked thinning of muscle<br />

coats and degeneration with vacuolation of smooth<br />

muscle cells and replacement by fibrous tissue. 18<br />

Some patients appear to be more mildly affected<br />

than others, and merely require dietary modification<br />

with either a low-fat, low-fiber, low-lactose<br />

diet or enteral feeding, together with the intermittent<br />

use of antibiotics to treat bacterial overgrowth.<br />

In patients with more severe disease surgical<br />

decompression of the foregut into the normal<br />

jejunum appears to be effective. 19<br />

Familial visceral myopathy type 2 is quite distinct<br />

and was first reported as oculogastrointestinal<br />

muscular dystrophy. Recently the mitochondrialdisease<br />

nature of the condition has resulted in it<br />

being renamed mitochondrial neurogastrointestinal<br />

encephalomyopathy. 20 It is inherited as an<br />

autosomal recessive trait and presents with external<br />

ophthalmoplegia with ptosis and diplopia, a<br />

cardiac conduction defect, mild muscular atrophy<br />

and dilatation of the entire gastrointestinal tract<br />

with scattered small-bowel diverticulae. Gastrointestinal<br />

symptoms with dyspepsia, retrosternal<br />

chest pain and weight loss may start during<br />

teenage years. In skeletal muscle biopsies a deficiency<br />

of cytochrome C oxidase has been demonstrated<br />

which results in the ragged red fibers<br />

typical of mitochondrial myopathies. Examination<br />

of the smooth muscle of the gut shows the presence<br />

of fibrosis and degeneration with vacuolation<br />

of the smooth muscle cells. Most patients have<br />

eventually required total parenteral nutrition.<br />

Familial visceral myopathy type 3 is much less<br />

well understood. The data are from of one family<br />

in which there were four siblings who presented<br />

with dilatation of the entire gastrointestinal tract.<br />

The condition appeared to be inherited in an autosomal<br />

recessive fashion. 21<br />

Systemic disorders involving<br />

intestinal smooth muscle<br />

Until recently the majority of myopathies involving<br />

multisystem disorders have been described in<br />

adults, but in whom the disease process has been<br />

in place for a number of years resulting in obvious<br />

damage to the intestinal smooth muscle coats,<br />

mostly with increased fibrosis. With modern physiological<br />

recording methods and histological<br />

Systemic disorders involving intestinal smooth muscle 275<br />

techniques available in young children, myopathies<br />

associated with systemic disorders have<br />

been increasingly recognized. It is important to<br />

identify these conditions, as medical treatment<br />

may be available for them which can result in<br />

symptomatic improvement and even prevention of<br />

progression of the disease process, if it is put in<br />

place before fibrosis of the muscle coat has<br />

occurred.<br />

Connective tissues disorders<br />

Scleroderma, systemic lupus erythematosus and<br />

even dermatomyositis may all affect the muscle<br />

coats of the bowel. However, the best understood<br />

of these is scleroderma, a systemic disease characterized<br />

by the excessive deposition of collagen by<br />

fibroblasts in the skin and in many internal organs.<br />

It is associated with prominent and often severe<br />

alteration of the microvasculature, the autonomic<br />

nervous system and the immune system. 22<br />

Gastrointestinal involvement with symptoms of<br />

clinical relevance occur in approximately 50% of<br />

patients with the systemic form of scleroderma.<br />

The esophagus is the most commonly affected part<br />

of the gastrointestinal tract, followed by the<br />

rectum and then the small intestine. The lesions of<br />

the muscle coat are similar throughout the gut<br />

with atrophy and fragmentation of the muscle<br />

coats, collagen infiltration and fibrosis in a later<br />

stage of the disease.<br />

Enteric myositis<br />

In Crohn’s disease, infiltration of the smooth<br />

muscle layer with lymphocytes has been found,<br />

but no clue as to whether this is part of the Crohn’s<br />

disease or an autoimmune phenomenon. Acquired<br />

myositis has been described in children in a<br />

number of reports. Two have presented with<br />

functional intestinal obstruction at 1 and 2.5 years<br />

of age. In these patients a dense lymphocyte infiltrate<br />

mainly of T cells was found along the large<br />

and small intestine. Both responded to immunosuppressive<br />

treatment. A further child with similar<br />

histological findings but in whom there was clear<br />

evidence of an autoimmune process has more<br />

recently been described. 8 In this patient features<br />

were noted that suggested that the intestinal<br />

muscle cells themselves were taking part in the


276<br />

Chronic intestinal pseudo-obstruction in childhood<br />

inflammatory response. The patient responded to<br />

prednisolone, azathioprine and cyclophosphamide,<br />

but was dependent upon steroids.<br />

A similar inflammatory cell infiltrate was described<br />

in 12 of 27 Bantu children from South Africa who<br />

had some sporadic form of an acquired degenerative<br />

enteric myopathy (V.V. Smith, personal communication).<br />

However, no information was given as to<br />

whether the children suffered from other autoimmune<br />

diseases or had autoantibodies present. Nor<br />

was there information about response to immunosuppressive<br />

treatment.<br />

Muscular dystrophy<br />

Gastrointestinal involvement may occur in a<br />

number of forms of muscular dystrophy including<br />

myotome muscular dystrophy and the dystrophinopathies<br />

Duchenne and Becker muscular<br />

dystrophy. Involvement of the smooth muscle of<br />

the gastrointestinal tract and bladder is well<br />

described in myotonic muscular dystrophy, and<br />

seemingly motor abnormalities can be found<br />

throughout the whole gastrointestinal tract. 23<br />

Duchenne and the milder variety Becker muscular<br />

dystrophy are due to abnormalities of dysrophin. 24<br />

The predominant effects are on skeletal and<br />

cardiac muscle. Abnormality of particularly the<br />

smooth muscle of the foregut also occurs, with<br />

impairment of gastric emptying and disordered<br />

proximal small intestinal motor activity demonstrable<br />

on manometry. Histological studies show<br />

that the intestinal smooth muscle cells become<br />

swollen, then destroyed, and progressively<br />

replaced by fat. 25<br />

Disorders of the enteric nervous<br />

system<br />

The enteric nervous system is arranged in the form<br />

of two major plexuses: the myenteric plexus,<br />

which is located between the longitudinal and<br />

circular muscle layers and primarily provides<br />

motor innervation to the muscle coats; and the<br />

submucous plexus, which lies in the submucosa<br />

between the circular muscle layer and the muscularis<br />

mucosae. 26 The submucous plexus is important<br />

in regulating secretion by the mucosa and<br />

providing sensory innervation of the mucosa. As a<br />

whole it is a collection of neurons derived from<br />

neural crest cells and has been referred to as the<br />

‘brain of the gut’ or the ‘little brain’. Whilst there is<br />

a two-way flow of information between the central<br />

nervous system and the enteric nervous system,<br />

often referred to as the ‘big brain’ and ‘little brain’,<br />

respectively, the little brain can function independently<br />

of the big brain. 27 The neurons of the<br />

enteric nervous system are grouped into small<br />

ganglia that are connected by bundles of nerve<br />

processes forming the two major nerve plexuses.<br />

Ganglia consist of tightly packed neurons, terminal<br />

bundles of nerve fibers and glial cells, which<br />

usually outnumber the enteric neurons. 26<br />

Normally, there are between five and seven<br />

neurons in a ganglion. There are numerous<br />

interneurons between the two plexuses and within<br />

the plexuses. Both are connected to the central<br />

autonomic neural network by parasympathetic<br />

and sympathetic nerves. Throughout the gastrointestinal<br />

tract there are also non-neural cells<br />

derived from the mesenchyme, the interstitial cells<br />

of Cajal, that generate and propagate slow waves. 28<br />

The interstitial cells are important modulators of<br />

communication between nerves and muscle. The<br />

enteric nervous system is particularly concerned<br />

with the propulsion of the gut contents in an<br />

ordered physiologically effective fashion, and the<br />

control of secretion by the gut. Both quantitative<br />

and qualitative changes in the enteric nervous<br />

system have been identified and these are<br />

described below.<br />

Primary visceral neuropathies<br />

Primary visceral neuropathies can be subdivided<br />

into familial neuropathies, where a distinct pattern<br />

of inheritance is known, and sporadic cases, in<br />

which there are distinctive clinical and morphological<br />

findings. In this chapter, Hirschsprung’s<br />

disease, which is clearly a primary visceral neuropathy,<br />

will not be discussed. A number of neuropathic<br />

motility disorders, including those that<br />

present with slow-transit constipation, remain<br />

unclassified, although most frequently degenerative<br />

changes are found in the enteric neurons.<br />

Familial visceral neuropathy without extraintestinal<br />

manifestations<br />

In this disorder it is mainly the colon and distal<br />

small intestine that are affected, and it is inherited


as an autosomal dominant trait. 39 The age of onset<br />

may differ within the same family, but in general<br />

symptoms develop after infancy. Most patients<br />

present with severe slow-transit constipation associated<br />

with abdominal distension and colicky<br />

pain. In about half of the patients severe episodes<br />

of functional obstruction have occurred which<br />

have required decompression. To date, there have<br />

been no descriptions of any associated abnormalities,<br />

such as extrinsic autonomic dysfunction. Fullthickness<br />

biopsies of the affected parts of the<br />

bowel have shown evidence of degeneration, both<br />

on silver staining and on routine hematoxylin-andeosin<br />

staining. No other abnormalities have been<br />

observed.<br />

Familial visceral neuropathy with <strong>pylori</strong>c stenosis, a<br />

short small intestine and malrotation<br />

This syndrome has been described in several families.<br />

An extended kindred from Sicily and<br />

Southern Italy has shown that it is inherited as an<br />

X-linked recessive trait through four generations.<br />

Linkage analysis assigns the genetic defect to a<br />

locus at XQ28. To date, the gene involved has not<br />

been identified. Affected infant boys usually<br />

present with functional obstruction in the neonatal<br />

period and most have died during the first year<br />

of life. Histology of the nerve plexuses has shown<br />

the presence of shrunken neurons with particular<br />

loss of argyrophilic neurons. However, apparent<br />

loss of argyrophilic neurons may be a normal<br />

finding at this age, as the acquisition of argyrophilia<br />

is associated with changes in the neurofilamentous<br />

content of the neuron and this occurs<br />

progressively during the first year of life.<br />

Familial visceral neuropathy with neuronal<br />

intranuclear inclusions<br />

This condition may be inherited as an autosomal<br />

dominant trait, as it was described in three siblings<br />

(two female, one male) and their father. 30 The<br />

patients developed symptoms during childhood<br />

and, in addition to pseud-obstruction, suffered<br />

from dysphagia, diarrhea and constipation. They<br />

became developmentally delayed, had autonomic<br />

dysfunction and ataxia, and developed dementia.<br />

A characteristic feature of the condition is the<br />

presence of eosinophilic intranuclear inclusions in<br />

the neurons of the myenteric plexus and in the<br />

central nervous system. 3 In some of the patients<br />

Disorders of the enteric nervous system 277<br />

the intranuclear inclusions could be seen within<br />

the ganglion cells of the submucous plexus on<br />

suction rectal biopsy. This appears to be a progressive<br />

degenerative condition affecting a widespread<br />

class of neurons in both the enteric and the central<br />

nervous systems.<br />

Familial visceral neuropathy with neurological<br />

involvement<br />

A number of families have been described in which<br />

there are least two affected siblings. The condition<br />

is probably inherited as an autosomal recessive<br />

disorder. The symptoms start in early childhood,<br />

with neuropathic dysmotility of the gut and<br />

involvement of the central nervous system. The<br />

central nervous system disorder appears to consist<br />

of progressive sensory and motor peripheral<br />

neuropathies, ophthalmoplegia and hearing loss. 31<br />

It seems highly likely that this is a mitochondrial<br />

disorder. On full-thickness biopsy, neurons in the<br />

myenteric plexus appear normal on conventional<br />

staining, but there is evidence of degeneration of<br />

argyrophobic C cells.<br />

Familial visceral neuropathy associated with<br />

multiple endocrine neoplasia<br />

The multiple endocrine neoplasia type 2<br />

syndromes may be associated with involvement of<br />

the gastrointestinal tract. These include multiple<br />

endocrine neoplasia (MEN) 2A, MEN 2B and<br />

isolated medullary thyroid carcinoma. These<br />

conditions are inherited as autosomal dominant<br />

traits and are as a consequence of mutation of the<br />

gene encoding the RET tyrosine kinase receptor. It<br />

is extremely important in the development of the<br />

enteric nervous system as RET null knockout mice<br />

do not develop neurons within the gut. 32 There is<br />

now good evidence to show that RET is important<br />

not only for colonization of the primitive gastrointestinal<br />

tract by neural crest cells but also for<br />

their migration down the length of the gut and their<br />

further differentiation. In MEN 2A and isolated<br />

medullary thyroid carcinoma, there is an abnormality<br />

of the RET gene between codons 619 and<br />

638, the cysteine-rich region of the gene, which<br />

results in the development of medullary thyroid<br />

carcinoma with or without pheochromocytoma. In<br />

about 5% of patients with MEN 2A, Hirschsprung’s<br />

disease is also present with pheochromocytoma. 33


278<br />

Chronic intestinal pseudo-obstruction in childhood<br />

In MEN 2B there is always involvement of the<br />

gastrointestinal tract, with transmural intestinal<br />

ganglioneuromatosis. In most patients, disorders<br />

of gastrointestinal motility are the first manifestations<br />

of the disease, but the presentation is variable<br />

both in severity and in time. Some patients<br />

present in early infancy, similarly to<br />

Hirschsprung’s disease, but others not until adult<br />

life. 34 There is nearly always colonic dysfunction<br />

present and the most usual presentations are<br />

either with chronic constipation, episodes of functional<br />

obstruction or as if this were Hirschsprung’s<br />

disease. In all the patients that the author has<br />

studied, evidence of medullary thyroid carcinoma<br />

has been present from very early on, either as<br />

clumps of malignant cells in situ within the<br />

thyroid gland or as an overt tumor. Frequently in<br />

those patients in whom there are only collections<br />

of cells in situ within the gland, screening investigations<br />

such as CT scanning of the thyroid and<br />

calcitonin determinations have not been helpful. It<br />

is only when there is a considerable mass of C cells<br />

present that the normal screening investigations<br />

become positive. It is for this reason, together with<br />

the fact that there is no satisfactory radiotherapeutic<br />

or chemotherapeutic treatment available, that<br />

prophylactic thyroidectomy is recommended. 35<br />

The characteristic histopathological findings are<br />

an increased density of nerve fibers and possibly<br />

ganglion cells in the submucosa and myenteric<br />

plexus with penetration of the hyperplastic nerve<br />

fibers into the mucosal area. The hyperplastic<br />

nerve fibers are accompanied by large ganglionic<br />

nodes containing numerous glial cells, with a<br />

normal to increased quantity of neurons. This<br />

abnormality of the enteric nervous system is<br />

present along the entire gastrointestinal tract and<br />

the hyperplastic neurons may be seen within the<br />

mouth or anal canal. 35<br />

Sporadic visceral neuropathies<br />

Neuropathic dysmotility may also be produced by<br />

having too few neurons within the enteric nervous<br />

systems (hypoganglionosis) as well too many<br />

(hyperganglionosis) or none at all (aganglionosis).<br />

The conditions that cause these states are nearly<br />

always congenital, although both hypoganglionosis<br />

and aganglionosis may result from acquired<br />

disorders in which there is destruction of neurons.<br />

As these conditions are defined by the numbers of<br />

neurons present, it is clear that a reliable means of<br />

assessing neuronal density is required. 36 Neuronal<br />

density is affected by the age of the patient, tissue<br />

freshness and intestinal dilatation as well as by the<br />

disease process. It is therefore important that a<br />

standardized technique for assessing neuronal<br />

density be used. A full-thickness sample of intestine<br />

at an appropriate site, needing to be greater<br />

than 1 cm in length, is required. Preferably<br />

neurons should be counted in sections cut longitudinally<br />

along the long axis of the bowel rather<br />

than transversely. 36 If sections are cut transversely<br />

they should be at least 30 µm apart to avoid counting<br />

each neuron more than once. There are few<br />

published studies, especially in children, but<br />

Smith reported a mean neuronal density of<br />

3.6 neurons/mm for the jejunum, 4.3/mm for the<br />

ileum and 7.7/mm for the colon, with no significant<br />

difference between transverse and longitudinal<br />

sections. 36<br />

Hypoganglionosis<br />

A reduced number of neurons in the myenteric<br />

plexus, so-called hypoganglionosis, is perhaps the<br />

most frequent diagnosis in children who present<br />

with functional obstruction due to a visceral<br />

neuropathy. Hypoganglionosis also occurs<br />

commonly in children with severe slow-transit<br />

constipation. Navarro et al3 reported hypoganglionosis<br />

of the myenteric plexus in 13 of 26<br />

patients who presented with functional obstruction.<br />

Most commonly the hypoganglionosis was<br />

confined to the distal segment of the colon, and<br />

diffuse disease was uncommon. Histologically the<br />

ganglia were smaller than normal, often infiltrated<br />

with collagen, and there was a paucity of neurons<br />

within the ganglia. Between the ganglia there were<br />

numerous thickened nerve fibers that stained<br />

strongly with acetylcholinesterase. Krishnamurthy<br />

et al described a further series of patients with<br />

hypoganglionosis37 in four of 26 children, but<br />

unlike those described by Navarro et al the nerve<br />

tracts did not strongly stain with cholinesterase.<br />

As in both of these series the majority of the children<br />

presented as infants, care must be taken in<br />

the interpretation of silver staining as during the<br />

first year of life there may be absence of silver


staining. In the Krishnamurthy group 37 there were<br />

19 children with a deficiency of argyrophilic<br />

neurons. When argyrophilic neurons were present,<br />

they were small and had few processes. It was<br />

suggested that these patients suffered from a defect<br />

in differentiation and maturation of neurons from<br />

primitive neuroblasts. As these processes continue<br />

after birth, this would explain why some infants<br />

during the first year of life may have apparent<br />

abnormalities of the myenteric plexus which are<br />

not the case. 38<br />

The problem of the diagnosis of hypoganglionosis<br />

is emphasized by the marked variability of the<br />

clinical course of such patients. Most will present<br />

in the newborn period with symptoms suggestive<br />

of Hirschsprung’s disease, but others become<br />

symptomatic only during their preschool years. In<br />

those in which the disorder is restricted to the<br />

colon, ileostomy or colonic resection and the pullthrough<br />

procedure relieves their symptoms, but<br />

those in whom the disease is diffuse remain<br />

dependent on parenteral nutrition for their<br />

survival.<br />

Hyperganglionosis<br />

Hyperganglionosis is characterized by an excess of<br />

intestinal neurons in the myenteric plexus with or<br />

without hyperplasia of nerve fibers. The condition<br />

may also affect the submucous plexus, but in<br />

some, the submucous plexus will be normal.<br />

Hyperganglionosis requires the presence of large<br />

ganglia containing more than seven ganglion cells<br />

at a greater density than the normal range for the<br />

region. There is also some evidence that infants,<br />

especially those during the neonatal period, have a<br />

higher neuronal density than that in older children.<br />

38,39 Therefore, the finding of hyperganglionosis<br />

in a very small infant with dysmotility<br />

requires caution in its interpretation.<br />

Marked hyperganglionosis is the hallmark of<br />

intestinal ganglioneuromatosis and MEN 2B. This<br />

is dealt with above and will not be considered<br />

further here. In view of the implications of MEN<br />

2B in those with severe hyperganglionosis and<br />

ganglioneuromatosis, mutations of the RET gene<br />

should be sought in all patients who have severe<br />

hyperganglionosis even if only the submucous<br />

plexus is involved.<br />

Disorders of the enteric nervous system 279<br />

Intestinal neuronal dysplasia<br />

Those with mild submucous hyperganglionosis<br />

have sometimes been reported to suffer from<br />

intestinal neuronal dysplasia, a term that has been<br />

used over the past 30 years to describe quantitative<br />

and qualitative abnormalities of enteric ganglia.<br />

The term was originally used to describe abnormalities<br />

of both the myenteric and submucous<br />

plexus. 40 However, over the past 20 years, diagnosis<br />

has largely been dependent on suction rectal<br />

biopsy and thus on changes in the submucous<br />

plexus and the mucosal innervation. The term has<br />

raised confusion and controversy among clinicians<br />

and pathologists. It appears to affect all age groups,<br />

although it is mostly seen in infants with chronic<br />

constipation and was first considered to be a<br />

developmental defect of the submucous plexus. 41<br />

Part of the problem has been the lack of agreed<br />

criteria, and the confusion has been further<br />

compounded by the use of the histological description<br />

as a clinical diagnosis. No studies have shown<br />

a correlation between morphological features of<br />

intestinal neuronal dysplasia and symptoms or<br />

long-term outcome. 42,43 Nevertheless, surgical<br />

procedures have been recommended on the basis<br />

of the histological diagnosis. In one prospective<br />

study of rectal biopsies, the interobserver variation<br />

between centers was enormous and close to that<br />

which that might occur by chance. In this study, 43<br />

377 biopsies from 108 children aged 4–15 years<br />

were assessed by three experienced pathologists<br />

for a number of agreed histological features and a<br />

final diagnosis. Complete concordance was<br />

obtained for the diagnosis of Hirschsprung’s<br />

disease, but in only 14% of the remainder was<br />

there concordance. Assessment of the clinical<br />

symptoms 1 year after biopsy, demonstrated that<br />

the diagnosis of intestinal neuronal dysplasia had<br />

no prognostic value for the outcome in these individuals.<br />

It seems, therefore, that intestinal<br />

neuronal dysplasia describes neither a specific<br />

histological nor a clinical entity, and remains<br />

controversial. This author can see very little use<br />

for the term until it is better defined. It should at<br />

the present time be avoided and certainly not used<br />

as a criterion for individual treatments.<br />

Acquired visceral neuropathies<br />

Damage to the enteric nervous system may occur<br />

from a variety of agents as well as being secondary


280<br />

Chronic intestinal pseudo-obstruction in childhood<br />

to a systemic disease. Secondary visceral<br />

neuropathies may occur at any age, before or after<br />

birth, but most often occur in adults compared to<br />

children.<br />

Infectious agents<br />

Infectious agents may damage enteric neurons<br />

either by direct invasion of the neuron or by<br />

involvement in an inflammatory process, often<br />

autoimmune, that the infectious agent provokes.<br />

The best example of this is Chagas’ disease, caused<br />

by infection with Trypanosoma cruzi, in which the<br />

inflammatory response to the parasite results in an<br />

autoimmune response in which antibodies to the<br />

muscarinic receptors on neurons is produced.<br />

Whilst the most common clinical presentation<br />

following such an infection is achalasia, other<br />

areas of the bowel may be affected, including the<br />

small and large intestines.<br />

Functional obstructive episodes have also been<br />

observed with a variety of other infectious agents,<br />

including acute Lyme disease and the<br />

neurotrophic viruses from the herpes virus family,<br />

including cytomegalovirus, varicella zoster virus,<br />

Epstein–Barr virus and herpes simplex virus type<br />

1. 44–46<br />

Chronic inflammation and autoimmune disease<br />

A number of patients have been reported with<br />

different non-infectious inflammatory diseases.<br />

Clearly, classical mucosal inflammatory conditions<br />

such as Crohn’s disease and necrotizing enterocolitis<br />

may result in damage to both enteric<br />

nerves and muscular structures of the bowel.<br />

However, other autoimmune diseases affecting the<br />

gut, such as celiac disease and ulcerative colitis,<br />

may also result in severe dysmotility. It is becoming<br />

apparent that the neuromusculature of the<br />

bowel may become involved in the inflammatory<br />

process.<br />

Lymphocytic ganglionitis associated with the presence<br />

of circulating enteric neuronal antibodies has<br />

been described both in association with small<br />

round cell carcinoma as a paraneoplastic<br />

syndrome and in isolation. 47 In both settings<br />

acquired progressive aganglionosis occurs as a<br />

result of a severe T-cell-mediated inflammatory<br />

ganglionitis of both enteric plexuses. In the<br />

isolated form of the disease the antibody produced<br />

is similar to the Hu-protein antibody found in the<br />

paraneoplastic syndrome, but instead of antibody<br />

staining being restricted to the nucleus, it is<br />

present in the cytoplasm. In both forms of the<br />

disease, episodes of functional obstruction are<br />

responsive to immunosuppressive treatment, with<br />

a consequent fall in the titers of antibodies<br />

produced.<br />

The enteric neuromusculature may also become<br />

involved in a variety of connective tissues disorders<br />

including systemic sclerosis, systemic lupus<br />

erythematosus and dermatomyositis.<br />

Miscellaneous conditions<br />

The enteric neuromusculature can be affected by a<br />

wide variety of other conditions or affected by<br />

toxic agents, drugs and irradiation. These are listed<br />

in Table 18.1.<br />

Treatment<br />

In all the primary and many of the secondary<br />

causes of functional obstruction, treatment is<br />

symptomatic and supportive unless the underlying<br />

disease process can be corrected by specific<br />

therapy. For example, celiac disease presenting<br />

with functional obstruction can be treated acutely<br />

with immunosuppression and then by a glutenfree<br />

diet. The slow-transit constipation that occurs<br />

with hypothyroidism will respond to thyroxin<br />

replacement therapy. Similarly, other forms of<br />

autoimmune disease which affect specifically the<br />

nerves and muscle of the bowel can be treated by<br />

appropriate immunosuppressive treatment. Where<br />

an isolated obstructing segment of gut can be<br />

discerned, surgery may bring very real benefit.<br />

This will be discussed further below.<br />

Nutritional therapy plays an important role, as the<br />

majority of children die either as a consequence of<br />

malnutrition or of sepsis associated with<br />

parenteral nutrition. Wherever possible the enteral<br />

route should be used in preference to parenteral<br />

nutrition, and even if parenteral nutrition is<br />

required it is beneficial if small volumes of enteral<br />

feed can be maintained. In small infants, human<br />

milk is the preferred milk source, but if this is not<br />

available a whey protein hydrolysate formula


should be used in preference to whole-proteinbased<br />

cow’s milk formula, as this empties faster<br />

from the stomach. In older children episodes of<br />

functional obstruction can often be managed with<br />

enteral feeding of protein hydrolysates where they<br />

have previously tolerated a low-fiber diet. If<br />

enteral feeding cannot provide sufficient nutrients<br />

for normal growth and development, then<br />

parenteral nutrition should be initiated before<br />

malnutrition develops. Experience of over 40<br />

patients with severe recurrent episodes of pseudoobstruction<br />

has shown that approximately half<br />

will require parenteral nutrition at some time<br />

during the course of their illness and, in a proportion,<br />

home parenteral nutrition may be required.<br />

As a consequence of bowel dilatation and the<br />

development of blind loop syndromes, bacterial<br />

overgrowth frequently develops and will further<br />

enhance malabsorption due to fermentation of<br />

nutrients. Intermittent treatment with appropriate<br />

antibiotics is the recommended course of action,<br />

and this often improves symptoms.<br />

The use of pharmaceutical agents to improve<br />

intestinal motility can be expected to be successful<br />

only if there is remaining function of enteric<br />

nerves and muscles for the drugs to have a beneficial<br />

effect. In general, in those with severe and<br />

recurrent episodes of functional obstruction,<br />

pharmaceutical agents play little role. Agents that<br />

have been tried include cisapride, domperidone,<br />

metoclopramide, erythromycin and octreotide.<br />

Ondansetron, a 5-hydroxytryptamine3 antagonist,<br />

may be helpful in those cases in which the emetic<br />

reflex is activated, but it does not appear to affect<br />

episodes of obstruction.<br />

REFERENCES<br />

1. Heney KES, Smith VV, Spitz L, Milla PJ. Chronic intestinal<br />

pseudo obstruction: treatment and long term follow<br />

up of 44 patients. Arch Dis Child 1999; 81: 21–27.<br />

2. Vargos J, Sachs P, Ament ME. Chronic intestinal pseudo<br />

obstruction in paediatrics. J Paediatr Gastroenterol Nutr<br />

1998; 7: 323–332.<br />

3. Navarro J, Sonsino E, Boige N et al. Visceral<br />

neuropathies responsible for chronic intestinal pseudo<br />

obstruction syndrome in pediatric practice: analysis of<br />

26 cases. J Pediatr Gastronenterol Nutr 1990; 11:<br />

179–195.<br />

References 281<br />

Surgical intervention should be limited to the<br />

placement of decompression stomas, feeding<br />

gastrostomies and the provision of full-thickness<br />

intestinal biopsies for a specific diagnosis. Bowel<br />

resection may help in patients where the disease is<br />

limited to a specific segment of the gut such as the<br />

colon, but for those children with diffuse disease<br />

other than the placement of a decompression<br />

ileostomy, resection does not play a part. Similarly<br />

in those in whom a decompression ileostomy has<br />

produced relief, but there is diffuse disease, the<br />

urge to re-establish connection with the defunctioned<br />

limb of the bowel should be resisted as this<br />

will only result in further episodes of obstruction. 1<br />

Wherever possible, unnecessary surgery should be<br />

avoided as it will only create adhesion and complications.<br />

A most difficult surgical decision is in an<br />

individual case defining whether an episode of<br />

obstruction is functional or is a consequence of<br />

mechanical obstruction caused by the presence of<br />

adhesions. Signs of peritonism, extreme dilatation<br />

and pain in association with a specific episode of<br />

obstruction points more towards a mechanical<br />

obstruction, than a functional obstruction and<br />

laparotomy may be necessary to relieve it.<br />

For those infants born with primary neuromuscular<br />

disease and totally dependent on parenteral<br />

nutrition, the only therapeutic options are<br />

home parenteral nutrition or small-intestinal<br />

transplantation. Small-intestinal transplantation<br />

should be reserved for those who have severe<br />

parenteral nutrition-related liver disease or those<br />

whose intravenous access has become unreliable<br />

and precarious.<br />

4. Smith VV, Milla PJ. Histological phenotypes of enteric<br />

smooth muscle disease causing functional intestinal<br />

obstruction in childhood. Histopathology 1997; 31:<br />

112–122.<br />

5. Milla PJ. Clinical features of intestinal pseudo obstruction<br />

in Children. In Kamm MA, Lennard Jones JE, eds.<br />

Constipation. Petersfield, UK: Wrightson Bio Medical<br />

Publishing, 1994: 251–258.<br />

6. Tanner MS, Smith V, Lloyd JK. Functional intestinal<br />

obstruction due to deficiency of argyrophil neurons in<br />

the myenteric plexus. Familial syndrome presenting


282<br />

Chronic intestinal pseudo-obstruction in childhood<br />

with short small bowel, malrotation and <strong>pylori</strong>c hypertrophy.<br />

Arch Dis Child 1976; 51: 837–841.<br />

7. Auricchio A, Brancholini V, Casari G et al. the locus for<br />

a novel syndromic form of intestinal pseudo obstruction<br />

maps to XQ28. Am J Hum Genet 1996; 58: 743–749.<br />

8. Ruuska TH, Karikoski R, Smith VV, Milla PJ. Acquired<br />

myopathic pseudo-obstruction may be due to autoimmune<br />

enteric leiomyositis. Gastroenterology 2002; 122:<br />

1133–1139.<br />

9. Devane SP, Ravelli AM, Bisset WM et al. Gastric enteral<br />

dysrhythmias in children with chronic idiopathic<br />

intestinal pseudoobstruction. Gut 1992; 33: 1477–1481.<br />

10. Omari TI, Benninga MA, Barnett CP et al.<br />

Characterization of esophageal body and lower<br />

esophageal motorfunction. J Pediatr 1999; 135: 517–521.<br />

11. Fell JM, Smith VV, Milla PJ. Infantile chronic idiopathic<br />

intestinal pseudo obstruction: the role of small intestinal<br />

manometry as a diagnostic tool and prognostic indicator.<br />

Gut 1996; 39: 306–311.<br />

12. Stanghellini V, Camilleri M, Malagelada JR. Chronic<br />

idiopathic pseudo obstruction; clinical and intestinal<br />

manometric findings. Gut 1987; 28: 5–12.<br />

13. Di Lorenzo C, Floros AF, Hyman PE. Age related changes<br />

in colon manometry. J Paediatr 1995; 127: 593–596.<br />

14. Vanderwinden JM, Liu H, De Laet MH, Vanderhaeghen<br />

JJ. Study of the interstitial cells of Cajal in infantile<br />

hypertrophic <strong>pylori</strong>c stenosis. Gastroenterology 1996;<br />

111: 279–288.<br />

15. Vanderwinden JM, Rumessen JJ, Liu H et al. Interstitial<br />

cells of Cajal in human colon and in Hirschsprung’s<br />

disease. Gastroenterology 1996; 111: 901–910.<br />

16. Berdon WE, Baker DH, Blanc WA et al. Megacystitismicrocolon-intestinal<br />

hypoperistalsis syndrome: a new<br />

cause of intestinal obstruction in the new born. Am J<br />

Roentgenol 1976; 126: 957–964.<br />

17. Schuffler MD, Low CW, Bill AH. Studies of idiopathic<br />

intestinal pseudo obstruction. 1. Hereditary hollow<br />

visceral myopathy: clinical and pathological studies.<br />

Gastroenterology 1977; 73: 327–338.<br />

18. Schuffler MD, Pope CE. Studies of idiopathic intestinal<br />

pseudo obstruction. Hereditary hollow visceral myopathy:<br />

family studies. Gastroenterology 1977; 73: 339–344.<br />

19. Shaw A, Shaffer HA, Anuras S. Familial visceral myopathy:<br />

the role of surgery. Am J Surg 1985; 150: 102–108.<br />

20. Mueller LA, Camilleri M, Emslie SA. Mitochondrial<br />

neurogastrointestinal encephalo-myopathy: manometric<br />

and diagnostic features. Gastroenterology 1999; 116:<br />

959–963.<br />

21. Anuras S, Mitros FA, Milano A et al. A familial visceral<br />

myopathy with dilatation of the entire gastrointestinal<br />

tract. Gastroenterology 1986; 90: 385–390.<br />

22. Sjogren RW. Gastrointestinal motility disorders in scleroderma.<br />

Arthritis Rheum 1994; 37: 1265–1282.<br />

23. Lenard HW, Goebel HH, Weigel W. Smooth muscle<br />

involvement in congenital myotonic dystrophy.<br />

Neuropeadiatrie 1976; 8: 42–52.<br />

24. Hoffman EP, Brown RH, Kumdel LM. Dystrophin: the<br />

protein product of the Duchenne muscular dystrophy<br />

locus. Cell 1987; 51: 919–928.<br />

25. Staiano A, Del Giudice E, Romano A et al. Upper<br />

gastrointestinal tract motility in children with progressive<br />

muscular dystrophy. J Paediatr 1992; 121: 720–724.<br />

26. Furness JB, Costa M. The Enteric Nervous System.<br />

Edinburgh: Churchill Livingstone,1987.<br />

27. Wood JD. Physiology of the enteric nervous system. In<br />

Johnson LR, ed. Physiology of the Gastrointestinal Tract,<br />

3rd edn. New York: Raven Press, 1994: 423–482.<br />

28. Sanders KM. A case for interstitial cells of Cajal as pace<br />

makers and mediators of neurotransmission in the<br />

gastrointestinal tract. Gastroenterology 1996; 111: 492–515.<br />

29. Mayer EA, Schuffler MD, Rotter JI et al. Familial visceral<br />

neuropathy with autosomal dominant transmission.<br />

Gastroenterology 1986; 91: 1528–1535.<br />

30. Barnett JL, McDonnell WM, Appelman HD, Dobbins<br />

WO. Familial visceral neuropathy with neuronal<br />

intranuclear inclusions: diagnosis by rectal biopsy.<br />

Gastroenterology 1992; 102: 684–691.<br />

31. Faber J, Fich A, Steinberg A et al. Familial intestinal<br />

pseudoobstruction dominated by a progressive neurologic<br />

disease at a young age. Gastroenterology 1987; 92:<br />

786–790.<br />

32. Schuchardt A, D’Agati V, Larsson-Blomberg L et al.<br />

Defect in the kidney and enteric nervous system of mice<br />

lacking the tyrosine kinase receptor RET. Nature 1994;<br />

367: 380–383.<br />

33. Cote CJ, Gaged RF. Lessons learned from the management<br />

of a rare genetic career. N Engl J Med 2003; 399:<br />

1566–1568.<br />

34. Eng C, Marsh DJ, Robinson BG et al. Germline, RET,<br />

Codon 918 mutation in apparently isolated intestinal<br />

ganglioneuromatosis. J Endocrinol Metab 1998; 83:<br />

4191–4194.<br />

35. Smith VV, Eng C, Milla PJ. Intestinal ganglioneuromatosis<br />

and multiple neoplasia type 2B: implications<br />

for treatment. Gut 1999; 45: 143–146.<br />

36. Smith VV. Intestinal neuronal density in childhood: a<br />

baseline for the objective assessment of hypo and hyper<br />

aganglionosis. Paediatr Pathol 1993; 13: 225–237.<br />

37. Krishnamurthy S, Heng Y, Shuffler ND. Chronic intestinal<br />

pseudo obstruction in infants and children caused<br />

by diverse abnormalities of the myenteric plexus,<br />

Gastroenterology 1993; 104: 1398–1408.<br />

38. Smith VV, Milla PJ. Argyrophilia in the developing<br />

human myenteric plexus. Br J Biomed Sci 1996; 53:<br />

287–283.<br />

39. Schoffield DE, Yunise J. Intestinal neuronal dysplasia. J<br />

Pediatr Gastroenterol Nutr 1991; 12: 182–189.<br />

40. Meier-Ruge WA, Gambazzi F, Kaufeler RE et al. The<br />

neuropathological diagnosis of neuronal intestinal<br />

dysplasia (NIDB). Eur J Paediatr Surg 1993; 4: 267–273.<br />

41. Smith VV. Isolated intestinal neuronal dysplasia: a<br />

descriptive histological pattern or a distinct clinicopathological<br />

entity. In Hadziseli Movic F, Herzog B, eds.<br />

Inflammatory Bowel Disease & Morbus Hirschsprung.<br />

Dordrecht: Kluwer, 1992: 203–214.<br />

42. Cord-Udy CL, Smith VV, Ahmed S et al. An evaluation<br />

of the role of suction biopsy in the diagnosis of intestinal<br />

neuronal dysplasia. J Pediatr Gastroenterol Nutr<br />

1997; 24: 1–6.<br />

43. Koletzko S, Jesch I, Faus-Kebetaler T et al. Rectal biopsy<br />

for diagnosis of intestinal neuronal dysplasia in children:<br />

a prospective multi-centre study on interobserver<br />

variation and clinical outcome. Gut 1999; 44: 853–861.<br />

44. Sonsino E, Mouy R, Foucaud P et al. Intestinal pseudo<br />

obstruction related to cytomegalovirus infection of the<br />

myenteric plexus. N Engl J Med 1984; 311: 196–197.<br />

45. Bruyn GA, Bots GT, Van Wijhe M et al. Chronic intestinal<br />

pseudoobstruction as a possible sequel to encephalitis.<br />

Am J Gastroenterol 1986; 81: 50–54.<br />

46. Chen JJ, Liou YM, Gershon M. Latent, lytic, reactive varicella<br />

zoster virus in the ENS of humans and guinea pigs:<br />

could intestinal singles be a hidden cause of gastrointestinal<br />

disease. Neurogastroenterol Motil 2004; in press.<br />

47. Smith VV, Gregson N, Foggensteiner L et al. Acquired<br />

intestinal agangliosis and circulating auto antibodies<br />

without neoplasia or other neural involvement.<br />

Gastroenterology 1997; 112: 1366–1371.


19<br />

Introduction<br />

Gastrointestinal and nutritional<br />

problems in the neurologically<br />

handicapped child<br />

Jan Taminiau and Marc Benninga<br />

The neurologically handicapped child almost<br />

invariably experiences nutritional problems. 1<br />

They more commonly present as malnutrition, but<br />

obesity and overnutrition can also be found.<br />

Parameters to assess malnutrition and overnutrition<br />

in the handicapped child have to be<br />

adjusted. Height is a proper parameter for growth<br />

and nutritional status, but difficult in children<br />

with malformations and spasticity. 2–4 Also, disproportionate<br />

development of the head, rump and<br />

extremities makes assessment of height as a parameter<br />

of nutritional status difficult. 5–7 Therefore,<br />

crown–rump length, width, crown–heel length,<br />

distal femoral length and distal arm length<br />

(Spender growth curve) have been developed to<br />

assess growth and to relate height to developmental<br />

abnormalities or to nutrition. 8 It is clear<br />

that the Quetelet index or body mass index (BMI)<br />

have to be used with care in these children.<br />

Different etiologies result in different growth<br />

abnormalities during development in childhood. 9<br />

Children with Down’s syndrome, which is a<br />

genetic chromosomal disorder, have height and<br />

bone development retardation from birth. The<br />

rump or sitting height is normal in these children.<br />

The height development becomes relatively<br />

shorter each year up to the age of 15. The skull<br />

development stops at the age of 3. The growth<br />

deficit is more pronounced in height than in width<br />

and they appear microcephalic when they get<br />

older.<br />

Children with multiple congenital anomalies, with<br />

specific histories of drug therapy, irradiation and<br />

viral infections early in gestation, and which could<br />

be considered as an environmental insult, have<br />

height curves that are normal until the age of 5 for<br />

both sexes, and then start to lag behind. By the age<br />

of 10, they slow down and seem to miss their<br />

adolescent growth spurt. Many children in this<br />

group become microcephalic, and usually there<br />

are more deficits in width than height.<br />

In children with hypoxia, severe or prolonged<br />

deprivation of oxygen at the time of birth, in<br />

essence cerebral palsy with motor function impairment,<br />

height and trunk measurements are normal<br />

for 10 years and then slow down, perhaps reflecting<br />

a failure of the adolescent growth spurt. Head<br />

circumferences lag behind over all ages. On the<br />

other hand, children whose neurological deficit is<br />

minimal brain damage (attention deficit hyperactivity<br />

disorder, children with school and learning<br />

problems and no definite impairment of motor<br />

function), have their heights and weights within<br />

normal ranges. Therefore, body weight and BMI<br />

(kg/m 2 ) are more appropriate for evaluation of<br />

malnutrition in these children.<br />

In a study on more than 2000 institutionalized<br />

children with a handicap in Tokyo, Japan, 3 height<br />

and weight were measured in four distinct groups.<br />

Groups were divided into deaf children; blind<br />

children; mentally retarded children, some of<br />

whom were completely ambulatory and 15% of<br />

whom needed crutches; and physically handicapped<br />

children, of whom 65% were non-ambulatory.<br />

Height more than 3 standard deviations<br />

below the mean was present in 2% of deaf children,<br />

10% of blind children, 15% of children with<br />

mental retardation and 45% of physically handicapped<br />

children. Underweight more than 2 standard<br />

deviations below the mean was present in 1%<br />

of deaf children, 4% of blind children, 5% of children<br />

with mental retardation and 24% of physically<br />

handicapped children. Overweight more<br />

than 2 standard deviations above the mean was<br />

present in 8% of deaf children, 7.5% of blind<br />

283


284<br />

Gastrointestinal and nutritional problems<br />

children, 17% of mentally retarded children and<br />

9% of physically handicapped children. 10 In a<br />

Finnish study of patients up to the age of 20, the<br />

BMI showed that underweight (BMI 32 kg/m 2 ). 11 Also, more<br />

than 20% of children being overweight was found<br />

in another survey of more than 1100 children. 12,13<br />

Although the studies report height and weight<br />

reduction of almost 1 standard deviation in large<br />

cohorts of handicapped children, it seems more<br />

appropriate to relate the chance of malnutrition to<br />

mental retardation (5%) or physical handicap<br />

(25%). A height reduction of more than 3 standard<br />

deviations occurs in 1% of deaf children, 5% of<br />

blind children, 2% of mentally retarded children<br />

and 6% of physically handicapped children. This<br />

proportion of stagnation in height development in<br />

chromosomal, toxic and hypoxic disorders was not<br />

easily explained by nutritional depletion. Also,<br />

disproportionate development in the head, trunk<br />

and extremities were not in keeping with nutritional<br />

problems.<br />

In the normal child, undernutrition or overnutrition<br />

is obviously undesirable. In the handicapped<br />

child, however, a proper nutritional status is of<br />

crucial importance, as it supports the ability to<br />

employ all nutrients necessary for normal body<br />

functions in biological, physiological and psychological<br />

ways. Undernourished handicapped children<br />

might not respond properly to intercurrent<br />

diseases and suffer unnecessarily. On the other<br />

hand, restoring a normal nutritional status results<br />

in a better quality of life in many.<br />

Assessment of nutritional status requires a proper<br />

follow-up of height, body weight and assessment<br />

of the standard deviation score. By so doing, negative<br />

changes are easily discovered and appropriate<br />

nutritional intervention can be initiated. Spender<br />

et al 8 showed that upper arm length and lower leg<br />

length were as appropriate as height measurements<br />

in these children and had the same technical<br />

intra- and interobserver errors as the measurements<br />

of height, usually difficult to obtain in these<br />

children. In practice, they are easier to perform<br />

and have a better compliance for regular follow-up<br />

measurements. Triceps skin fold as indication of<br />

body fat and arm circumference as indication of<br />

muscle mass, should be measured in conjunction<br />

with height and body weight with the highly<br />

variable muscle mass in these children. 14<br />

Appetite<br />

Undernutrition in children with a neurological<br />

handicap is mostly caused by a decreased appetite<br />

or intake. Increased requirements or increased<br />

losses are not reported. Eating problems or difficulties<br />

in chewing, swallowing disturbances,<br />

absent swallowing reflex, no co-ordination of oral<br />

and esophageal phases and choking of food into<br />

the nose are implicated. Also, rumination, hypersensitivity<br />

in the mouth area, food refusal or aversion<br />

contribute to eating disorders.<br />

Nutrition is further influenced negatively by<br />

changes in taste, dry mouth, epigastric pain,<br />

nausea, sedation and hypersalivation. Specifically,<br />

anti-epileptics cause gingival hyperplasia and<br />

anorexia; antidepressants such as lithium increase<br />

thirst and appetite; mental stimulants such as<br />

Ritalin ® decrease appetite; antacids might cause<br />

constipation; and aspirin and non-steroidal antiinflammatory<br />

drugs (NSAIDs) cause pyrosis and<br />

nausea and may lead to ulcerations in the gastrointestinal<br />

tract. In the Institutional Tokyo Study it<br />

was shown that around 10% of the blind needed<br />

assistance with eating, mostly because of some<br />

chewing problems, and around 20% had poor<br />

appetite. In the mentally retarded, around 25%<br />

needed assistance during eating, 2% had swallowing<br />

disorders, around 35% had chewing disorders<br />

and 20% had poor appetite. In the physically<br />

handicapped, around 65% needed assistance with<br />

eating, 20% had swallowing disorders, 40% had<br />

chewing disorders and 30% had poor appetite.<br />

Four per cent of the mentally retarded children<br />

had a voracious appetite, which was in keeping<br />

with their overweight tendency. 15,16<br />

Nutrient deficiencies<br />

Nutrient deficiencies further show a relationship<br />

between undernutrition and nutritional intake,<br />

supported by several studies. 17–20 The undernourished<br />

had a lower intake of energy, protein, fat,<br />

fiber and water. In general, the linoleic acid intake<br />

was insufficient in 39% and fiber intake overall<br />

was insufficient in 59%. The micronutrient intake<br />

showed deficiencies in vitamin B6 (32%), vitamin<br />

C (18%), nicotinic acid (71%), sodium (61%) and<br />

iron (84%). Deficient intake of vitamins A, B1, B2<br />

and B6 was related to the undernourished group;


nicotinic acid and vitamin C were deficient in the<br />

whole group. In the undernourished, low sodium<br />

intake was more pronounced and that was also<br />

seen to a minor extent in the whole group.<br />

Sodium, potassium, calcium and phosphate intake<br />

were deficient only in the undernourished. Interestingly,<br />

iron intake was not different between<br />

the undernourished and the well-nourished group,<br />

in keeping with the laboratory assessments. Iron<br />

intake did not correlate with serum hemoglobin,<br />

serum iron and vitamin C intake. Calcium intake<br />

and milk intake showed no correlation with serum<br />

iron level. Low use of antacids was not correlated<br />

with serum iron level. Despite iron supplementation,<br />

serum iron remained deficient. Anemia<br />

occurred in 9% and, low serum iron in 24%.<br />

Anemia was more common in the undernourished<br />

(83% vs. 50%) as was the use of iron medication<br />

(67% vs. 22%) and also medication for gastroesophageal<br />

reflux (25% versus 16%). The iron<br />

resorption was not influenced by low levels of<br />

vitamin C, or high levels calcium, milk or antacid<br />

intake. The more severe the anemia, the more<br />

reflux medication was given. It is suggested that<br />

the anemia and low iron level were more related to<br />

intestinal blood loss due to reflux esophagitis than<br />

to the nutritional variations.<br />

Nutritional intervention<br />

In this last study a nutritional intervention was<br />

undertaken; in the well-nourished group, the nutritional<br />

intervention was compared and followed. The<br />

multiply handicapped children had spastic tetraplegia<br />

(75%), epilepsy (84%), disturbed chewing (84%),<br />

regurgitation (50%) and choking (50%). The nutritional<br />

intervention consisted more in a team<br />

approach than in taking measures specifically<br />

directed to individual needs. Milk products were<br />

maximized to 750 ml a day and were taken with<br />

meals. In between snacks, the emphasis was more<br />

on fruit and vegetable mixtures and fruit mixtures.<br />

Fiber was added to cereals in the order of 30 g a day.<br />

Those who refused bread were given porridge based<br />

on a mixture of tube feeding powder (1kcal/ml),<br />

water and cereals. Special considerations to drinks<br />

were given: drinks were enriched with nutritional<br />

supplements. If necessary, tube feeding was added;<br />

30% needed additional tube feeding.<br />

Nutritional intervention 285<br />

This multidisciplinary approach and intervention<br />

lasted for 1 year and resulted in an increase of energy<br />

intake from 1349 kcal to 1598 kcal. The normally fed<br />

group changed from 1673 to 1683 kcal/day. Protein<br />

intake increased from 50 to 67 g and in the normally<br />

fed from 73 to 79 g/day. Carbohydrates increased in<br />

the undernourished from 170 to 212 g/day, and in<br />

the well nourished from 189 to 208 g/day. Fat<br />

increased from 52 to 54 g/day, in the well nourished<br />

it declined from 70 to 60 g/day. Linoleic acid went<br />

from 6 to 10 g/day, and in the well nourished 8 to<br />

9 g/day. Fiber increased from 11 to 26 g/day and in<br />

the well nourished from 18 to 31 g/day. Water<br />

increased from 1077 to 1167 ml/day, and in the well<br />

nourished from 1401 to 1437 ml/day. After 1 year,<br />

the BMI improved from 18.8 to 19.3 kg/m 2 , triceps<br />

skin fold did not change, 0.98 to 1.03 cm, and the<br />

upper arm muscle circumference increased from 19<br />

to 20.1 cm. Nutritional deficiencies (in percentage<br />

less than two-thirds of the daily recommended<br />

allowance) declined in vitamin B6 of 32 to 2%,<br />

nicotinic acid of 77 to 34%, vitamin C of 18 to 0%,<br />

sodium of 61 to 39%, and iron of 84 to 11%). The<br />

normalization of fiber intake reduced the necessity<br />

for stimulant suppositories for constipation by<br />

56%.<br />

Thus, nutritional intervention by a team approach<br />

seems to be beneficial in the majority, showing<br />

that appropriate feeding is helpful in overcoming<br />

nutritional problems. When tube feeding is necessary<br />

for longer periods, percutaneous endoscopic<br />

gastrostomy (PEG) has become a fashionable<br />

approach. In a 1.5-year prospective study, the<br />

change of nasogastric tube feeding to PEG tube<br />

feeding had an unsuspected side-effect in reducing<br />

nutritional problems, vomiting and respiratory<br />

infections between 30 and 40%. 21 Also, well-being<br />

was improved in 40%. Interestingly, vomiting,<br />

airway infections and also irritability diminished.<br />

The standard deviation score of weight for height<br />

improved from -3.8 to -1.8. An easier access via<br />

PEG tube feeding, as compared to the irritating<br />

tube in the nose, resulted in better daily nutrient<br />

intake than more traditional nasogastric tube<br />

feeding.<br />

These observations are in keeping with studies in<br />

energy expenditure of children or adolescents with<br />

severe disabilities. In a study in children with<br />

cerebral palsy, 19 Stallings et al measured basic


286<br />

Gastrointestinal and nutritional problems<br />

energy expenditure by indirect calorimetry and<br />

total energy expenditure by the doubly labeled<br />

water method. Physical activity including chronic<br />

spasticity of children with spastic quadriplegic<br />

cerebral palsy was estimated from the ratio of total<br />

energy expenditure to resting energy expenditure.<br />

Control children were those children with<br />

adequate weight, adequate fat-free mass and fat<br />

mass, as well as children with a lower fat-free mass<br />

and a lower body fat mass. Measurements were<br />

made by triceps skin folds and arm circumference.<br />

An interesting observation in children with severe<br />

quadriplegic cerebral palsy who were expected to<br />

have extra energy demands for involuntary muscular<br />

work showed lower total energy expenditure<br />

and lower resting energy expenditure compared to<br />

well-nourished children with this handicap and to<br />

controls. Resting energy expenditure and activity<br />

energy expenditure (estimated as total minus<br />

resting energy expenditure) were both diminished,<br />

thus not supporting the commonly held view that<br />

spastic children expend more energy because of<br />

their involuntary movements. In this study,<br />

reported dietary intake was compared to total<br />

energy expenditure measured by the doubly<br />

labeled water method. With this method, body<br />

weight before and after the test period measures<br />

exactly the number of calories expended and this<br />

can be compared with the intake. It was thus<br />

shown that the dietary intake reported was overestimated<br />

by 50% compared to the actual intake<br />

measured exactly by the total energy expenditure.<br />

This finding underlines the problems that these<br />

children face with feedings: meal duration is<br />

longer, frequently exceeding 45 min, exceeding the<br />

tolerance attention span of the patient and the<br />

available time of the caregiver. Oral motor abnormalities<br />

of varying degrees are observed in these<br />

patients. They often contribute to the symptoms<br />

and may lead to food loss. In all these children<br />

with growth failure and abnormal body composition,<br />

it was shown that the resting energy expenditure<br />

adjusted for fat-free mass was lower in the<br />

poorly nourished than in the adequately nourished<br />

group of children with severe spasticity. This<br />

suggested a basic metabolic response or adaptation<br />

to the poorly nourished condition.<br />

Gastroesophageal reflux (GER) symptoms such as<br />

vomiting, rumination and regurgitations are found<br />

in 20–30% of the intellectually disabled popula-<br />

tion. GER-related iron deficiency anemia and<br />

hematemesis are noted in 10–20% of patients. In<br />

the Netherlands and Belgium in a large cohort of<br />

more then 1500 patients, a randomly selected<br />

intellectually disabled population was tested with<br />

pH-metry during 24 h. A pathological pH test<br />

(defined as a duration of a pH of < 4 for more than<br />

4% of the measured time) was seen in 48% of<br />

cases. 22 These patients were subjected to<br />

endoscopy and reflux esophagitis was found in<br />

96%: 14% had grade I esophagitis, 33% had grade<br />

II, 39% had grade III and 13% had grade IV (Savary<br />

Miller classification). Barrett’s esophagus was<br />

found in 14% and peptic strictures in 4% of cases.<br />

This study was repeated for the group under the<br />

age of 14, and similar findings were seen. In fact,<br />

GER disease was found even in the absence of<br />

overt symptomatology.<br />

A possible explanation for the high prevalence of<br />

GER disease is the increased prevalence of hiatal<br />

hernia, found in 50% of elderly patients and 76%<br />

of intellectually disabled children. In the Dutch<br />

study, 46% had hiatal hernia and no relationship<br />

with rumination. There was some association with<br />

cerebral palsy, use of anticonvulsant drugs, scoliosis<br />

and restlessness. Anticholinergics and sedatives<br />

do decrease the lower esophageal sphincter<br />

pressure, which in turn might trigger more<br />

episodes of GER. Interestingly, dental erosions<br />

were seen more frequently in individuals with a<br />

pH lower than 4 for more than 6.3% of the<br />

measured time and with an IQ of less than 35.<br />

For the whole Dutch and Belgian group, the standardized<br />

morbidity ratio for esophageal carcinoma<br />

was 2.9; there is thus an increased risk of developing<br />

esophageal cancer, probably related to chronic<br />

GER disease and Barrett’s dysplasia. For the<br />

patients who underwent anti-reflux surgery, this<br />

was found to be effective in 38%, while no<br />

symptom relief was seen in the remaining 62%.<br />

H2-blockers were effective in 60% of patients and<br />

proton pump inhibitors were effective in 69% of<br />

patients. Surgery for the neurologically impaired<br />

children is known to have a significantly higher<br />

incidence of major complications than in intellectually<br />

normal patients with GER. It is in fact, a<br />

common notion that re-operation and relapses of<br />

GER are frequent. The mortality rate was<br />

8.8–9.4%. The postoperative complication rate was<br />

12–15%. In this large Dutch and Belgian cohort,


omeprazole was used in a dose of 40 mg once daily<br />

and was effective in 88% independently of the<br />

severity of the esophagitis. Ten per cent of patients<br />

had symptomatic relapse after decreasing the dose<br />

to 20 mg daily as maintenance treatment; however,<br />

all these patients became symptom-free again after<br />

increasing the dose to 40 mg daily. Endoscopically,<br />

the esophagus was healed at the end of the study. 23<br />

The occasional patient may need up to 60 mg of<br />

omeprazole: Hassal et al found that a dose of<br />

1–4 mg/kg per day was needed in a variety of<br />

esophagitis patients with hiatal hernia, mental<br />

retardation, repaired esophageal atresia and even<br />

in apparently healthy children with esophagitis. 24<br />

Keeping these children on a maintenance of<br />

omeprazole 20 mg daily resulted in a marked<br />

improvement of persistent vomiting, hematemesis,<br />

regurgitation, food refusal and iron deficiency<br />

anemia. The prevalence of <strong>Helicobacter</strong> <strong>pylori</strong><br />

infections was also tested and found to be around<br />

50% of children, and up to 83% in adults. No clear<br />

correlation (either direct or inverse) between H.<br />

<strong>pylori</strong> infection and GER symptoms was found.<br />

Gastroesophageal reflux and<br />

percutaneus endoscopic gastrostomy<br />

GER and aspiration in patients fed via a gastrostomy<br />

tube may be caused by relaxation of the<br />

lower esophageal sphincter (LES) secondary to<br />

gastric distension caused by rapid intragastric<br />

bolus feeding. When feeding rate was slowed, LES<br />

pressure did not diminish to incompetent levels of<br />

2 mmHg. In general, prospective studies have<br />

shown reduction of vomiting, pneumonia, restlessness<br />

and pain in more then 60% of patients. After<br />

PEG placement, 24-h pH monitoring improved,<br />

and histological reflux esophagitis normalized in<br />

these children. Anti-reflux surgery for PEG placement<br />

considerably increased the complications<br />

and failed to improve symptomatology. Thus, there<br />

does not seem to be a place for anti-reflux surgery<br />

unless symptoms progress after PEG placement, in<br />

GER and PEG 287<br />

spite of concomitant treatment with proton pump<br />

inhibitors.<br />

The improvement in nutritional status obtained<br />

through PEG feeding induced further improvement<br />

in reflux, as judged by 24-h pH studies,<br />

suggesting a relation between malnourishment<br />

and reflux symptoms. 25<br />

Constipation in mentally<br />

handicapped children<br />

The incidence of constipation was around 61% in<br />

a large cohort of mentally handicapped children<br />

in Dutch and Belgian institutions. Constipation<br />

was defined as bowel movements less than 3<br />

times a week. Eighty-eight per cent of the constipated,<br />

mentally handicapped, children used laxatives,<br />

in comparison to 40% of constipated<br />

controls whose constipation was easily<br />

controlled. A significant correlation was found<br />

between non-ambulancy, cerebral palsy, use of<br />

anticonvulsive medication or benzodiazepines on<br />

the one hand and use of H2-receptor antagonists<br />

or proton pump inhibitors on the other. Also, an<br />

IQ of less than 50 correlated with food refusal,<br />

while there was no correlation with age, gender,<br />

use of cisapride, motilium, untreated GER disease<br />

or vomiting. Laxatives used were contact laxatives<br />

in 45%, and osmotic agents such as lactulose and<br />

enemas in 14%. Manual evacuation of feces was<br />

necessary in 7% of patients. In patients with cerebral<br />

palsy, anal sphincter pressures were normal.<br />

Rectal sensation was reduced and rectal size was<br />

increased, possibly contributing to the constipation<br />

so commonly seen in these children.<br />

Cisapride did not influence colonic transit time,<br />

but slightly and non-significantly increased the<br />

frequency of defecation from 2.5 to four times<br />

weekly. Thus, in summary, constipation in<br />

mentally handicapped children is frequently<br />

encountered, but seems to respond to laxative<br />

treatment without major sequelae. 26,27


288<br />

Gastrointestinal and nutritional problems<br />

REFERENCES<br />

1. Eyman RK, Grossman HJ, Chaney RH et al. The life<br />

expectancy of profoundly handicapped people with<br />

mental retardation. N Engl J Med 1990; 323: 584–589.<br />

2. Amundson JA, Sherbondy A, Dyke van DC, Alexander<br />

R. Early identification and treatment necessary to<br />

prevent malnutrition in children and adolescents with<br />

severe disabilities. J Am Diet Assoc 1994; 94: 880–883.<br />

3. Miller F, Koreska J. Height measurement of patients with<br />

neuromuscular disease and contractures. Dev Med Child<br />

Neurol 1991; 33: 55–58.<br />

4. Garn S, Weir HF. Assessing the nutritional status of the<br />

mentally retarded. Am J Clin Nutr 1971; 24: 853–854.<br />

5. Roche AF. Growth assessment in abnormal children.<br />

Kidney Int 1978; 14: 369–377.<br />

6. Mosier HD, Grossman HJ, Dingman HF. Physical growth<br />

in mental defectives: a study in an institutionalised<br />

population. Pediatrics 1995; 36: 465–473.<br />

7. Rimmer JH, Kelly LE, Rosentswieg J. Accuracy of<br />

anthropometric equations for estimating body composition<br />

of mentally retarded adults. Am J Ment Def 1987;<br />

91: 626–632.<br />

8. Spender QW, Cronk CE, Charney EB, Stallings VA.<br />

Assessment of linear growth of children with cerebral<br />

palsy: use of alternative measures to height or length.<br />

Dev Med Child Neurol 1989; 31: 206–214.<br />

9. Pryor HB, Thelander HE. Growth deviations in handicapped<br />

children. Clin Pediatr 1967; 6: 501–512.<br />

10. Suzuki M, Saitoh S, Tasaki Y et al. Nutritional status<br />

and daily physical activity of handicapped students in<br />

Tokyo metropolitan schools for deaf, blind, mentally<br />

retarded, and physically handicapped individuals. Am J<br />

Clin Nutr 1991; 54: 1101–1111.<br />

11. Similä S, Niskanen P. Underweight and overweight<br />

cases among the mentally retarded. J Ment Def Res 1991;<br />

35: 160–164.<br />

12. Fox R, Rotatori A. Prevalence of obesity in mentally<br />

retarded adults. Am J Ment Retard 1982; 87: 228–230.<br />

13. Perry M. Treating obesity in people with learning<br />

disabilities. Nursing Times 1996; 92: 37–38.<br />

14. Cole HS, Lopez R, Epel R et al. Nutritional deficiencies<br />

in institutionalised mentally retarded and physically<br />

disabled individuals. Am J Ment Def 1985; 89: 552–555.<br />

15. Ault MM, Guy B, Rues J et al. Some educational implications<br />

for students with profound disabilities at risk<br />

for inadequate nutrition and the nontherapeutic effects<br />

of medication. Ment Retard 1994; 32: 200–205.<br />

16. Karle IP, Bleiler RE, Ohlson MA. Nutritional status of<br />

cerebral–palsied children. J Am Diet Assoc 1960; 38:<br />

22–26.<br />

17. Couriel JM, Bisset R, Miler R et al. Assessment of<br />

feeding problems in neurodevelopmental handicap: a<br />

team approach. Arch Dis Child 1993; 69: 609–613.<br />

18. Ellman G, Salfi M, Fong A et al. Vitamin B6 status<br />

measures of an institutionalised mentally retarded<br />

population. Am J Ment Def 1986; 91: 30–34.<br />

19. Stallings VA, Zemel BS, Davies JC et al. Energy expenditure<br />

of children and adolescents with severe disabilities:<br />

a cerebral palsy model. Am J Clin Nutr 1996; 64:<br />

627–634.<br />

20. Hordijk R. De waarde van diverse parameters<br />

(antropometrische en laboratoriumgegevens, nutriënten)<br />

bij het bepalen van de voedingstoestand van ernstig<br />

meervoudig gehandicapten. 1992<br />

21. Mathus-Vliegen EM, Koning H, Taminiau JA, Moorman<br />

Voestermans CG. Percutaneous endoscopic gastrostomy<br />

and gastrojejunostomy in psychomotor retarded<br />

subjects: a follow-up covering 106 patient years. J<br />

Pediatr Gastroenterol Nutr 2001; 33: 488–494.<br />

22. Bohmer CJ, Niezen-de Boer MC, Klinkenberg-Knol EC et<br />

al. Gastro-oesophageal reflux disease in institutionalised<br />

intellectually disabled individuals. Neth J Med 1997; 51:<br />

134–139.<br />

23. Clarisse JM, Böhmer CJ, Riet C et al. Omeprazole.<br />

Therapy of choice in intellectually disabled children.<br />

Arch Pediatr Adolesc Med 1998; 152: 1113–1118.<br />

24. Hassal E, Israel D, Shepherd R et al. Omeprazole for<br />

treatment of chronic erosive esophagitis in children: a<br />

multicenter study of efficacy, safety, tolerability and<br />

dose requirements. International Pediatric Omeprazole<br />

Study Group. J Pediatr 2000; 137: 800–807.<br />

25. Van Winckel M, Vander Stichele R, De Bacquer D,<br />

Bogaert M. Use of laxatives in institutions for the<br />

mentally retarded. Eur J Clin Pharmacol 1999; 54:<br />

965–969.<br />

26. Staiano A, del Giudice E, Simeone D et al. Cisapride in<br />

neurologically impaired children with chronic constipation.<br />

Dig Dis Sci 1996; 41; 870–874.<br />

27. Böhmer CJ, Taminiau JA, Klinkenberg-Knol EC,<br />

Meuwissen SG. The prevalence of constipation in institutionalized<br />

people with intellectual disability. J<br />

Intellect Disabil Res 2001; 45: 212–218.


20<br />

Introduction<br />

Cyclic vomiting syndrome<br />

Bhanu Sunku and B UK Li<br />

Cyclic vomiting syndrome (CVS) is a disorder of<br />

unknown etiology and pathogenesis characterized<br />

by recurrent, stereotypical episodes of vomiting<br />

with varying intervals of baseline or normal health<br />

in between. 1 Various recent articles and proceedings<br />

of two international conferences on CVS<br />

published in the past decade have defined this<br />

disorder in detail and proposed potential mechanisms<br />

and treatment. These publications and<br />

symposia have provided critical steps in recognizing<br />

and understanding a disorder that has been<br />

poorly recognized and commonly misdiagnosed.<br />

Typical misdiagnoses include gastroenteritis,<br />

gastroesophageal reflux, food poisoning, recurrent<br />

‘flu’ and eating disorders. 2 Although CVS can<br />

begin in infancy, the median age of onset in our<br />

cohort is 4.8 years of age. As a measure of misdiagnosis,<br />

the median interval from onset of symptoms<br />

to the proper recognition is 1.9 years, during<br />

which time the child has suffered through about<br />

15 or so episodes. Although the prevalence and<br />

incidence of CVS are unknown, current estimates<br />

in a school-based survey of Caucasian children<br />

5–15 years of age report a prevalence of 2%. 3<br />

Although CVS is clearly misdiagnosed, in our<br />

experience this figure appears to be excessive,<br />

perhaps because the study was based on a questionnaire<br />

that did not involve exclusionary testing<br />

and because milder cases that did not require<br />

medical intervention were detected. In any case, in<br />

our reported pediatric gastroenterology experience,<br />

CVS was second only to gastroesophageal<br />

reflux disease as a cause of recurrent vomiting. 4<br />

Similar to the gender profile in migraine<br />

headaches, there is a slight predominance of girls<br />

over boys (57:43). 5<br />

Although cyclic vomiting was first reported in<br />

France by Heberden in 1806, 6 Samuel Gee in<br />

England in 1882 is credited with the most accurate<br />

‘modern’ description. 7 In the past decade, greater<br />

recognition and mechanistic understanding of this<br />

disorder have been achieved. Prior to this recent<br />

period, an association with migraine headaches<br />

was noted as early as 1898 by Whitney 8 and in<br />

1904 by Rachford. 9 Recently, pathophysiological<br />

connections have been made with mitochondrial<br />

disease, autonomic dysfunction and the stress<br />

response. Current research, including our own, is<br />

focused on the identification of neuroendocrine<br />

mechanisms mediating vomiting in these patients,<br />

with a specific emphasis on the role of hypothalamic<br />

corticotropin releasing factor (CRF).<br />

Clinical patterns<br />

CVS is distinguished by recurrent, severe, discrete<br />

episodes of vomiting. Episodes are stereotypical in<br />

regards to time of onset, duration and symptomatology.<br />

This disorder is also distinguished by an<br />

on–off pattern with intervals of returning to<br />

completely normal or baseline health between<br />

episodes. 2 The duration of episodes is generally<br />

from hours to days, with a median duration of<br />

27 h. The median frequency of episodes is 4<br />

weeks. Because nearly 47% of all patients have<br />

regular intervals and the remaining have sporadic<br />

attacks at unpredictable intervals, ‘cyclic’ is a<br />

slight misnomer.<br />

The most common time of onset is during nighttime<br />

or early morning hours with 42% of patients<br />

having onset from 01.00 to 07.00. 10 Of patients,<br />

67% have a well-described prodrome that precedes<br />

the episodes of vomiting. The parents often<br />

characterize both the onset and the resolution as<br />

sudden. The warning period typically lasts a<br />

median of 0.5–1.5 h. Despite the similarities to<br />

migraines, these prodromes rarely have visual<br />

289


290<br />

Cyclic vomiting syndrome<br />

disturbances and are characterized by pre-vomiting<br />

autonomic symptoms of pallor, nausea,<br />

abdominal pain and lethargy. Many parents have<br />

noted a rapid ‘shut-off’ of symptoms from the end<br />

of vomiting to the point of resuming oral intake<br />

and near-normal function that generally lasts a<br />

median of 8h with a range of 0h to 1 week.<br />

The vomiting itself has been described as rapidfire<br />

in nature, peaking at an average of six times<br />

per hour or once every 10min during the worst<br />

episode. 4 The vomiting is typically projectile, and<br />

contains bile, mucus and occasionally blood. The<br />

latter generally occurs late in the episode as a<br />

result of prolapse gastropathy from repetitive<br />

vomiting which can also produce epigastric pain. 11<br />

Although a threshold of more than four emeses per<br />

hour at the peak originally identified 92% of those<br />

with CVS, we have detected increasing numbers<br />

who fit the consensus historical criteria with a<br />

lower intensity of vomiting of two to four times per<br />

hour. 4 These children can appear remarkably<br />

debilitated during episodes. They are often curled<br />

up into the fetal position from pain, which is often<br />

accompanied by pallor, listlessness and unresponsiveness,<br />

and therefore appear much more ill than<br />

children with gastroenteritis. 2<br />

Even though children return to baseline health<br />

between episodes and are well about 90% of the<br />

time, CVS can have a significant impact on the<br />

quality of life of affected children. 10 Over half<br />

(58%) of all affected patients require intravenous<br />

hydration during episodes and 19% require this<br />

with every episode. School-age children miss an<br />

Table 20.1 Clinical features<br />

average of 24 days of school per year, owing to<br />

episodes of vomiting. The high medical morbidity<br />

is reflected by the average annualized cost of<br />

management of $17000, including doctor visits,<br />

emergency department visits, in-patient hospitalizations,<br />

biochemical, radiographic, endoscopic<br />

testing and missed work by the parents. 12<br />

There are many symptoms that typically accompany<br />

vomiting during CVS episodes (Table 20.1).<br />

Abdominal pain, retching, anorexia and nausea<br />

are the most common gastrointestinal symptoms.<br />

The abdominal pain can be excruciating occasionally<br />

to the point of requiring narcotics and/or<br />

laparotomy.<br />

By the patient’s report, nausea is generally the<br />

most persistent and distressing symptom: it is<br />

minimally relieved by vomiting, often receding<br />

only while sleeping or with sedation. Typical<br />

behaviors (assuming a fetal position, social withdrawal,<br />

compulsive drinking, and avoiding lights<br />

and sounds) represent attempts to alleviate the<br />

nausea. 10<br />

The most common autonomic symptoms are<br />

lethargy and pallor. Other autonomic symptoms<br />

include fever, flushing, drooling, diarrhea,<br />

hypothermia and hypertension. Less than half of<br />

the patients have migraine features including<br />

headache (42%), photophobia (38%) and phonophobia<br />

(30%). However, these numbers are significantly<br />

higher than in those with the chronic<br />

vomiting pattern who primarily have upper<br />

gastrointestinal tract disorders. 13 Other symptoms<br />

• Median age of onset: 4.8 years<br />

• Female/male: 57/43<br />

• Vomiting: 6 emesis/h at peak, with bile (81%), mucus (68.6%) and blood (34%)<br />

• Average emesis per episode: 31<br />

• Duration of episodes: 27h<br />

• Typical time of onset: 01.00–07.00<br />

• Median interval between episodes: 4 weeks<br />

• Associated symptoms: lethargy (93%), nausea (82%), abdominal pain (81%), anorexia (81%), retching (79%),<br />

headache (42%), photophobia (38%), phonophobia (30%)<br />

• Associated signs: pallor (91%), social withdrawal (54%), fever (30%), diarrhea (30%), drooling (27%)


during episodes include sensory hypersensitivity<br />

and vertigo.<br />

The largest fraction (32%) have a seasonal clustering<br />

of episodes with more during the winter and<br />

fewer during the summer. Although this pattern<br />

correlates with the school year, we can only speculate<br />

that less school-related stress and less exposure<br />

to infections trigger fewer episodes. Winter<br />

holidays including Thanksgiving, Christmas and<br />

New Year can serve as a positive stress for some.<br />

Various stressors have been noted to precipitate<br />

episodes of CVS. In 76% of patients, the parents<br />

can identify a recurring trigger preceding the<br />

vomiting episodes. These triggers consist of<br />

psychological, infectious and physical stressors<br />

(Table 20.2). Stress and infections are the most<br />

common triggers (44% and 31%, respectively).<br />

Interestingly, two-thirds of the stress is positive<br />

rather than negative. Various infections can trigger<br />

episodes, especially chronic sinusitis. Other<br />

triggers include dietary (23%), physical exhaustion<br />

(24%), atopic symptoms (6%), motion sickness<br />

(12%) and menses (22% of menstruating girls).<br />

Evaluating the family history of patients with CVS<br />

is helpful in diagnosis because of the much higher<br />

rate of positive migraines (82%) compared to that<br />

obtained in those with chronic vomiting due to<br />

gastrointestinal disorders or to the general population<br />

(15%). 5 Also, a family history of food allergies<br />

or atopy can be a clue for food triggers of<br />

Table 20.2 Common triggers for episodes of cyclic vomiting syndrome<br />

Infections (urinary tract infection, streptococcal throat, flu, sinusitis, gastroenteritis)<br />

Positive stress (birthdays, holidays, vacations)<br />

Negative stress (school, family, deaths)<br />

Dietary intake (cheese, chocolate, caffeine, monosodium glutamate)<br />

Physical exhaustion (lack of sleep)<br />

Motion sickness (car rides, roller-coasters, air travel)<br />

Asthma<br />

Allergies (seasonal, inhalants, foods)<br />

Menses<br />

Surgery/anesthesia<br />

Weather (temperature or pressure changes)<br />

Clinical patterns 291<br />

attacks. In our series, we have found an unusually<br />

high percentage of patients with a family history of<br />

depression (40%) compared to the general population.<br />

How this plays a role in possible susceptibility<br />

to CVS of patients is currently unknown.<br />

Cyclic versus chronic patterns of vomiting<br />

An important clinical clue to the diagnosis of CVS<br />

is the pattern of vomiting. Based on a temporal<br />

pattern, children with recurrent vomiting can be<br />

delineated into cyclic and chronic groups (Figure<br />

20.1). The cyclic group has an intense, but<br />

intermittent pattern of vomiting with peak emeses<br />

of ≥ 4/h and 2 episodes per week. 13 The chronic<br />

group has a low-grade, daily pattern of emeses<br />

with 2 episodes per week 13<br />

(Table 20.3). These criteria are 92% sensitive and<br />

100% specific for identifying children with CVS. 2<br />

Two-thirds of all children with recurrent vomiting<br />

fit into the chronic or continuous pattern of vomiting.<br />

These children rarely appear acutely ill or<br />

become dehydrated. Conversely, the cyclical<br />

pattern is associated with more intense vomiting<br />

and affected children more often require intravenous<br />

hydration (62% vs. 18%) compared with<br />

the chronic group. 4<br />

These two patterns are important because both of<br />

these groups differ in symptom and diagnostic<br />

profile. In those with the cyclical vomiting pattern,


292<br />

Cyclic vomiting syndrome<br />

non-gastrointestinal disorders including neurological<br />

(including abdominal migraine), renal,<br />

endocrine and metabolic disorders predominate<br />

over gastrointestinal disorders by a ratio of 5 : 1. 2,13<br />

In contrast, in the chronic group, gastrointestinal<br />

disorders (mostly peptic disease) predominate<br />

over non-gastrointestinal causes of vomiting by a<br />

ratio of 7 : 1. 2,13 This implies the need to center the<br />

Emeses/day<br />

30<br />

20<br />

10<br />

0 0 30 60<br />

Days<br />

Figure 20.1 Temporal patterns of vomiting: cyclic versus<br />

chronic. The number of emeses is plotted over a 2-month<br />

period. The chronic pattern represented by a dashed line<br />

has low-grade, nearly daily episodes, whereas the cyclic<br />

pattern represented by the solid line has many emeses<br />

over a 1–2-day period that recurs every few weeks.<br />

(Adapted from reference 46).<br />

Table 20.3 Characteristics of chronic and cyclic vomiting<br />

diagnostic work-up on extraintestinal disorders in<br />

cyclic vomiting, and on upper gastrointestinal<br />

tract disorders in chronic vomiting. Consistent<br />

with proposed migraine association with CVS, the<br />

cyclic pattern of vomiting has a higher prevalence<br />

of family members with migraine headache (72%<br />

vs. 14%), associated headache (41% vs. 19%) and<br />

photophobia (18% vs. 4%). 4<br />

Differential diagnosis<br />

Differentiating a cyclic versus a chronic pattern of<br />

vomiting is the first step in narrowing the differential<br />

diagnosis (Table 20.4). Although the majority<br />

of patients (88%) with a cyclical pattern ultimately<br />

are diagnosed with CVS, the remaining<br />

12% have specific causes for vomiting found on<br />

diagnostic testing. The majority of disorders that<br />

can mimic CVS include non-gastrointestinal as<br />

well as gastrointestinal disorders.<br />

Of the gastrointestinal disorders, the most serious<br />

are anatomic anomalies of the gastrointestinal tract<br />

including malrotation with intermittent volvulus,<br />

which can cause ischemic necrosis. Although not<br />

typically cyclical, we have found a few children<br />

with eosinophilic esophagitis related to significant<br />

food allergies to mimic CVS. Lucarelli et al have<br />

described seven children with a positive radioallergosorbent<br />

test (RAST) to foods (milk, egg<br />

Chronic pattern Cyclic pattern<br />

Time of onset daytime night-time or early morning<br />

Number of emeses/h 2 episodes/week ≤2 episodes/week, typically<br />

2–4 weeks<br />

Family history of migraine uncommon (14%) common (82%)<br />

Ill-appearing no yes (pale, lethargic)<br />

Headaches infrequent (19%) frequent (41%)<br />

Photophobia infrequent (4%) frequent (18%)<br />

Vertigo infrequent (7%) frequent (24%)<br />

Intravenous hydration required uncommon (18%) common (62%)<br />

Esophagitis on EGD common (59%) uncommon (15%)<br />

EGD, esophagogastroduodenoscopy


Table 20.4 Differential diagnosis of cyclic vomiting<br />

white and soy) whose episodes diminished after<br />

specific food elimination. 14<br />

The most common extraintestinal cause is acute<br />

hydronephrosis resulting from proximal or distal<br />

ureteral obstruction. Metabolic causes include<br />

mitochondrial disorders (disorders of fatty acid<br />

oxidation, mitochondrial encephalopathy, lactic<br />

acid and stroke-like syndrome), urea cycle<br />

defects (partial ornithine transcarbamylase deficiency),<br />

organic acidurias (proprionic acidemia),<br />

aminoacidurias and porphyrin degradation<br />

disorders (acute intermittent porphyria). 15,16<br />

Chronic pattern Cyclic pattern<br />

Pathophysiology 293<br />

Gastrointestinal peptic injury (GERD esophagitis, anatomic (malrotation, volvulus,<br />

gastritis, duodenitis) duplication cyst)<br />

inflammatory bowel disease pseudo-obstruction<br />

celiac disease cholelithiasis/gallbladder dyskinesia<br />

chronic appendicitis<br />

pancreatitis<br />

eosinophilic gastroenteritis/esophagitis<br />

Infectious chronic sinusitis sinusitis/other infections may be<br />

a trigger<br />

giardiasis<br />

Genitourinary pyelonephritis, pregnancy acute hydronephrosis due to<br />

uretopelvic junction obstruction<br />

or stones<br />

Metabolic rare mitochondrial disorders (MELAS)<br />

organic acidemias<br />

aminoacidurias<br />

fatty acid oxidation defects<br />

urea cycle defects<br />

acute intermittent porphyria<br />

Endocrine adrenal hyperplasia Addison’s disease<br />

diabetic ketoacidosis<br />

pheochromocytoma<br />

Neurological Chiari malformation migraine (headaches/abdominal)<br />

subtentorial neoplasm abdominal epilepsy<br />

familial dysautonomia<br />

Psychiatric Münchausen-by-proxy Münchausen-by-proxy<br />

pscyhogenic vomiting anorexia nervosa<br />

bulimia nervosa<br />

CVS<br />

GERD, gastroesophageal reflux disease; CVS, cyclic vomiting syndrome<br />

Neurosurgical causes include various lesions of<br />

the subtentorial region including cerebellar<br />

medulloblastoma, brain stem glioma and Chiari<br />

malformation. 2<br />

Pathophysiology<br />

CVS is considered an idiopathic disorder because<br />

no etiopathogenesis has been documented.<br />

However, although no specific cause has been<br />

identified, several tenable hypotheses and associa-


294<br />

Cyclic vomiting syndrome<br />

tions have been raised to explain this unique<br />

disorder. The current concept is that CVS is a<br />

functional brain–gut disorder involving central<br />

neuroendocrine mediation and peripheral<br />

gastrointestinal manifestations.<br />

Migraines<br />

An association with migraines was identified over<br />

a century ago. 8,9 Previous reports demonstrated the<br />

association CVS has with migraine headaches, a<br />

positive family history of migraines, 5 and progression<br />

of clinical episodes of CVS to migraine<br />

headaches with advancing age. 5,17 In the absence<br />

of definitive diagnostic tests for migraines and<br />

CVS, a putative causal relationship is further<br />

supported by similar symptomatology (e.g. pallor,<br />

lethargy, nausea, photophobia, phonophobia) and<br />

positive responses in both groups to anti-migraine<br />

therapy.<br />

Data from our series of over 440 patients show that<br />

the majority (87%) of patients with CVS do have a<br />

migraine association based on either a positive<br />

family history or concomitant or subsequent<br />

development of migraines in the patient. The<br />

progression of cyclic vomiting in childhood to<br />

abdominal migraines and eventually migraine<br />

headaches in adulthood has been labeled by<br />

Barlow as the periodic syndrome. 18 Many studies<br />

have confirmed this observation including reports<br />

of adults with migraines whose headache symptoms<br />

started with recurrent vomiting. 19,20<br />

Compared with the 13% who have non-migraine<br />

CVS, these migraine-associated CVS patients<br />

generally have milder episodes with significantly<br />

fewer emeses/episode, more symptoms of abdominal<br />

pain, headache, photophobia and social withdrawal,<br />

a greater association with psychological<br />

stress and significantly higher response rates to<br />

anti-migraine therapy (79% vs. 36%). 5,17<br />

Among the anti-migraine drugs that achieve such<br />

a positive response is sumatriptan (52%), a selective<br />

1B/1D serotonin agonist. This action on serotonin<br />

receptors with similar rates of response to<br />

patients with migraine headaches suggests a<br />

central role of action presumably by decreasing<br />

cerebrovascular dilatation. Since less than half of<br />

patients with a migraine association actually have<br />

headaches, a family history of migraines and the<br />

development of headaches over time are important<br />

in supporting this etiological linkage. Abdominal<br />

migraine shares many of the clinical features of<br />

CVS with midline abdominal pain being the most<br />

consistent and distressing symptom during<br />

episodes. 13 Migraine headaches, abdominal<br />

migraine and CVS differ in their primary symptoms<br />

but all have similar rates of secondary symptoms<br />

of pallor, lethargy, anorexia and nausea.<br />

Recent studies on migraine headaches have identified<br />

the periaqueductal gray (PAG) matter as a site<br />

of dysfunction in migraine attacks. Previous work<br />

with electrode stimulation on PAG, 21 positron<br />

emission tomography (PET) scan data demonstrating<br />

increased blood flow, 22 and altered iron homeostasis<br />

23 all suggest PAG involvement in migraine<br />

patients. Welch et al have speculated that similar<br />

mechanisms could be involved in CVS: PAG<br />

dysfunction may fail to attenuate afferent signals<br />

for vomiting and other autonomic symptoms<br />

during attacks. 23 Until we have a clearer delineation<br />

of mechanisms involved in migraine and<br />

CVS, we cannot be certain whether the CVS<br />

patients who do not fit under the migraine<br />

umbrella have distinct or similar pathophysiologic<br />

cascades.<br />

Mitochondrial dysfunction<br />

An association with mitochondrial DNA (mtDNA)<br />

mutations has been noted in children with CVS.<br />

Although the precise functional defects are<br />

unknown, Boles and Willians have proposed that<br />

altered mitochondrial metabolism may be<br />

involved. 16 They reported a large mtDNA<br />

rearrangement associated with CVS and subsequently<br />

demonstrated nine mutations in the<br />

hypervariable region of the D-loop (control region)<br />

of the mtDNA. 16 Some migraines and cyclic vomiting<br />

appear to result from disordered energy metabolism<br />

in mitochondrial encephalopathy lactic<br />

acidosis and stroke-like (MELAS) syndrome. 24<br />

Since mtDNA is maternally derived, the findings<br />

of a matrilineal predominance of migraines (64%<br />

maternal side only vs. 16% paternal side) in children<br />

with CVS further suggested an association. 10<br />

Many of the affected children with identified<br />

mutations have a clinical presentation with developmental<br />

delay, poor growth and seizures.<br />

However, based on non-specific lactate elevations


and organic acid abnormalities (ethylmalonic<br />

acid), we suspect that there could be subtle underlying<br />

mitochondrial dysfunction evident during<br />

CVS episodes even in patients without identified<br />

mutations.<br />

Neuroendocrine dysfunction<br />

Activation of the hypothalamic–pituitary–adrenal<br />

(HPA) axis was first described in CVS by Wolfe and<br />

Adler 25 and Sato et al. 25,26 Stressors in the form of<br />

infectious disorders, psychological perturbations<br />

and physical triggers have been well documented<br />

as precipitants of CVS episodes. 2 Sato et al<br />

described elevated levels of adrenocorticotropic<br />

hormone (ACTH), antidiuretic hormone (ADH),<br />

cortisols, prostaglandin E2, and serum and urinary<br />

catecholamines during episodes of CVS. 27 This<br />

finding may partially explain the symptoms of<br />

hypertension and profound lethargy in this subset<br />

of patients.<br />

The role of CRF as a brain–gut mediator in foregut<br />

function has been extensively described in<br />

animals by Taché et al. 28 In animal models, these<br />

authors demonstrated convincingly that central<br />

CRF acting on CRF-R2 receptors stimulated the<br />

dorsal motor nucleus (proximal end) of the vagus<br />

and delayed gastric emptying. 28,29 Clinical CVS in<br />

humans is precipitated by parallel stimuli that<br />

augment CRF release in animals. Also, the same<br />

hormones in a subset of Sato’s group reflected a<br />

stimulated HPA axis presumably initiated by CRF<br />

release, and appeared to give rise to the prominent<br />

symptoms of hypertension, anorexia and delayed<br />

gastric emptying. Our current studies examining<br />

the role of CRF in CVS may not only elucidate the<br />

pathophysiological cascade but could also open<br />

potential therapeutic avenues involving CRF<br />

antagonists.<br />

Despite the coherent hypothesis of CRF as a main<br />

mediator of vomiting, there may be another layer<br />

of dysfunction needed to explain why CVS<br />

episodes tend to be prolonged for hours. This<br />

added dysfunction could explain why most<br />

stressed individuals simply have ‘butterflies in<br />

their stomach’ or a single emesis, whereas children<br />

with CVS vomit for days on end. This could represent<br />

a dysfunctional PAG area that permits afferent<br />

autonomic signals to persist unattenuated, or<br />

Potential subtypes of cyclic vomiting syndrome 295<br />

augmented HPA axis stimulation or CRF receptor<br />

sensitivity in CVS patients.<br />

Autonomic dysfunction<br />

Symptoms of fever, lethargy, pallor, flushing,<br />

drooling and diarrhea occur commonly during<br />

episodes of CVS and are mediated by the autonomic<br />

nervous system. There are several lines of<br />

evidence that support the role of autonomic<br />

dysfunction in CVS, including clinical parallels in<br />

familial dysautonomia, documented alterations in<br />

autonomic tone and evidence of dysmotility on<br />

electrogastrography. Children with familial dysautonomia<br />

(FD, also known as Riley–Day syndrome),<br />

can manifest episodes of autonomic crises which<br />

resemble a sympathetic storm including discrete<br />

episodes of vomiting associated with tachycardia,<br />

hypertension, diffuse sweating, and emotional<br />

lability. Given the similarities, there may be<br />

commonalities in autonomic dysfunction between<br />

CVS episodes and FD crises. Studies using single<br />

photon emission computed tomography (SPECT)<br />

(cerebral perfusion) have localized enhanced<br />

temporal lobe perfusion during symptoms of<br />

nausea and retching that support that region’s<br />

involvement in ictal vomiting. 30<br />

To et al have observed heightened sympathetic<br />

cardiovascular tone and diminished parasympathetic<br />

vagal modulation via spectral analysis of<br />

R–R interval variability in CVS patients compared<br />

to controls. 31 Rashed et al demonstrated similar<br />

adrenergic autonomic abnormalities between CVS<br />

and migraine patients by showing lowered<br />

postural adjustment ratios. 32 Although the significance<br />

is still controversial, there is some electrogastrographic<br />

evidence of baseline gastric<br />

dysmotility in children with CVS that may<br />

respond to medium-dose erythromycin acting like<br />

motilin to enhance gastric motility and prevent<br />

vomiting episodes. 33–35<br />

Potential subtypes of cyclic vomiting<br />

syndrome and common associations<br />

The vast majority (87%) of patients with CVS have<br />

a migraine association either based most<br />

commonly on a family history of migraine or, less


296<br />

Cyclic vomiting syndrome<br />

frequently, on the subsequent development of<br />

migraines in the affected child. Because these<br />

associated symptoms occurred less than half the<br />

time, we did not use headache, photophobia and<br />

phonophobia as specific criteria to determine who<br />

had migraine-associated CVS. When we examined<br />

the remainder with non-migraine-associated CVS<br />

(13%), they appeared to have longer episodes, had<br />

more emeses per episode and were far less likely to<br />

respond to anti-migraine therapy (79 vs. 36%). 5 At<br />

present, we cannot determine whether the nonmigraine<br />

CVS is pathophysiologically distinct or<br />

simply resides at the severe end of the same mechanistic<br />

spectrum.<br />

Are there more subtypes of CVS? Although we<br />

have been able to differentiate other subgroups<br />

based on precipitating events, clinical pattern and<br />

associated symptomatology, there are too few<br />

subjects in each to know whether there are significant<br />

differences. In Sato’s subtype, described<br />

patients have hypertension and profound lethargy<br />

to the point of inability to walk, talk or respond.<br />

We have found that these patients generally have<br />

more prolonged episodes (102h vs. 50h) and<br />

increased vomiting per episode (75 vs. 31 emeses).<br />

Several children with developmental and growth<br />

delay, seizures and mild lactic acidosis are<br />

suspected of having underlying mitochondrial<br />

enzymopathy from a mtDNA mutation. Indeed,<br />

nine patients with this clinical picture have been<br />

found to have mutations in the D-loop or control<br />

region of the mtDNA. 36 Others with CVS have<br />

been found to a have either a large rearrangement<br />

or a single point mutation (MELAS). 16,24<br />

There are several other clinical patterns that can<br />

be identified. Again, it is unclear whether they<br />

simply represent various inciting stimuli that<br />

initiate the same pathophysiological cascade, or<br />

whether they represent distinct effector pathways.<br />

We hope that delineation of clinical patterns into<br />

subgroups may ultimately point us towards potential<br />

treatment approaches. For instance, some<br />

appear to have episodes that occur after periods of<br />

fasting, often induced by illness. They appear to<br />

respond rapidly to intravenous glucose and are<br />

suspected of being heterozygote carriers of disorders<br />

of fatty acid oxidation. Others have a stable<br />

periodicity (e.g. 60 days) to their episodes independent<br />

of stress or infectious triggers. Stable periodicity<br />

has been observed in some postmenarchal<br />

girls who have episodes at the onset of their<br />

menses and often respond to birth control pills<br />

with a low estrogen dose. 2 Some respond to prokinetic<br />

agents, but because of the lack of motility<br />

studies, it is not known whether they have documented<br />

gastric dysmotility. 37 Finally, in a group of<br />

children, dietary triggers of their episodes can be<br />

identified. Some are initiated by typical migraine<br />

precipitants including cheese, chocolate and<br />

monosodium glutamate, whereas others are triggered<br />

by food allergies identified by RAST<br />

testing. 14<br />

Diagnostic evaluation<br />

At present, there are no specific laboratory<br />

changes to diagnose CVS, and the diagnosis<br />

depends on fulfilling key historical criteria (Table<br />

Table 20.5 Diagnostic criteria (from the First International Symposium on Cyclic Vomiting<br />

Syndrome)<br />

Essential criteria<br />

≥3 recurrent, severe, discrete episodes of vomiting<br />

Varying intervals of normal or baseline health between episodes<br />

Duration of episodes from hours to days<br />

No apparent cause for vomiting (negative laboratory, radiographic and endoscopic testing)<br />

Supportive criteria<br />

Stereotypical episodes are similar in regards to time of onset, intensity, duration, frequency and associated symptoms<br />

and signs<br />

Self-limited in that episodes will resolve if left untreated<br />

Associated symptoms and signs


20.5). In the absence of positive laboratory findings,<br />

most of the testing with recurrent vomiting is<br />

directed towards excluding other treatable causes.<br />

Potentially treatable underlying gastrointestinal,<br />

neurological, renal, metabolic and endocrine<br />

causes are not rare, and are found in 12% of those<br />

who present with a cyclic pattern of vomiting. In<br />

addition, 12% (many overlapping) have an identifiable<br />

surgical disorder. 2 The challenge is to determine<br />

what and how much testing should be done,<br />

because a ‘shot-gun’ approach can be costly, timeconsuming<br />

and invasive. We conducted a costdecision<br />

analysis and concluded that an upper<br />

gastrointestinal tract X-ray with a small bowel<br />

follow-through followed by 2 months of empiric<br />

anti-migraine therapy was the most cost-effective<br />

initial treatment strategy for CVS. 12 If no therapeutic<br />

response occurs, more systematic testing<br />

should be performed (Figure 20.2).<br />

The first step is to identify a recurrent pattern<br />

versus a single acute vomiting illness typical of<br />

gastroenteritis. The next is to distinguish between<br />

a low-grade, nearly daily chronic and explosive,<br />

intermittent cyclic pattern of vomiting. Once<br />

identified, the diagnostic evaluation involves<br />

evaluation for both gastrointestinal and nongastrointestinal<br />

causes. In our experience, the<br />

most potentially devastating causes are sought<br />

including anatomic anomalies of the gastrointestinal<br />

tract and renal hydronephrosis by upper<br />

gastrointestinal series with small bowel followthrough<br />

and renal ultrasound examination, respectively.<br />

Metabolic and endocrine testing must be<br />

performed before intravenous glucose and fluids<br />

are administered, because these can alter respective<br />

findings on metabolic screening and evaluation<br />

of the HPA axis metabolites. The endoscopy<br />

and the head magnetic resonance imaging (MRI)<br />

are reserved for those in whom therapy has failed<br />

or who have specific symptoms suggesting peptic<br />

or allergic gastrointestinal disease or intractable<br />

headaches, respectively. Laboratory testing and a<br />

proposed algorithm for evaluation are presented in<br />

Figure 20.2.<br />

Natural history and complications<br />

There are limited data on the natural history of<br />

CVS. From our data of over 440 patients, the<br />

median age of resolution of symptoms is 9.9 years<br />

Natural history and complications 297<br />

and one-third (28%) of the children have thus far<br />

undergone the transition from CVS into migraine<br />

headaches as they reached early adolescence. Our<br />

projection analysis estimates that 75% of patients<br />

will develop migraine headaches by the of age 18.<br />

Other long-term studies have shown that up to half<br />

of CVS patients will continue with CVS or<br />

migraine headaches. 17 Several studies have noted<br />

the mean duration of illness to be around 6<br />

years, 13,38 but in our cohort, the younger the age of<br />

onset, the longer the duration. Also, 5% of patients<br />

will progress through all three phases of periodic<br />

disease including CVS to abdominal migraine and<br />

finally to migraine headaches. 13<br />

Complications and medical morbidity include<br />

iatrogenic tests and interventions from the misdiagnoses<br />

that were often applied to recurrent<br />

vomiting. Most are mislabeled as gastroenteritis,<br />

gastroesophageal reflux and food poisoning, and<br />

are treated in urgent care settings. Some with<br />

severe pain, bilious vomiting and intractability<br />

have undergone inappropriate laparotomy, appendectomy,<br />

cholecystectomy and Nissen fundoplication.<br />

Others have been labeled with psychiatric<br />

disorders including bulimia and psychogenic<br />

vomiting, and have been hospitalized on psychiatric<br />

wards, and a few parents have been<br />

suspected of Münchausen-by-proxy. 39<br />

Complications can also occur from the frequent<br />

and often severe episodes of vomiting that occur<br />

with CVS. Dehydration and electrolyte disturbances<br />

are common and intravenous rehydration<br />

is required in 58% of patients, which can be<br />

compared to less than 1% in rotavirus infection.<br />

Hematemesis can occur towards the end of attacks<br />

and is usually related to prolapse gastropathy or<br />

Mallory–Weiss tears. 11 Although not common,<br />

frequent vomiting can lead to secondary peptic<br />

injury. Aspiration and growth failure fortunately<br />

seem to be uncommon occurrences.<br />

Treatment<br />

Current treatment for CVS can be divided into<br />

supportive (during episodes), prophylactic (to<br />

prevent episodes) (Table 20.6) and abortive<br />

therapy (to stop episodes) (Table 20.7). Other<br />

strategies for management of CVS include avoidance<br />

of identified triggers (e.g. dietary cheese),


298<br />

Cyclic vomiting syndrome<br />

Figure 20.2 Algorithm for diagnostic evaluation and acute management of recurrent (chronic and cyclic) vomiting.


Table 20.6 Prophylactic therapy<br />

Drug Side-effects<br />

Anti-migraine<br />

Amitryptyline (1–2mg/kg per day) qhs sedation, anticholinergic<br />

Propranalol (0.5–1mg/kg per day) bid or tid hypotension, bradycardia, fatigue<br />

Cyproheptadine (0.25–0.5mg/kg per day) bid or tid sedation, weight gain, anticholinergic<br />

Anticonvulsants<br />

Phenobarbital (2–3mg/kg per day) qd or bid sedation<br />

Valproate (500–1000mg ER qhs) somnolence, hepatotoxicity<br />

Carbamezapine (5–10mg/kg per day) bid sedation, anticholinergic<br />

Prokinetic<br />

Erythromycin (10–20mg/kg per day) qid gastric cramps in larger doses<br />

Treatment 299<br />

Birth control<br />

Norethindrone (1.5mg) ethinylestradiol (20–30µg) qd estrogen-related, nausea, abdominal pain<br />

qhs, at bed-time: bid, twice a day; tid, three times a day; qd, every day<br />

Table 20.7 Abortive therapy<br />

Drug Side-effects<br />

Anti-migraine<br />

Sumatriptan 20mg NAS and may repeat x1 or 25mg po x1 chest and neck burning (po form), headache<br />

Frovatriptan 2.5mg po and may repeat x1 coronary vasospasm, dizziness<br />

Rizatriptan 5–10mg po x1, may repeat q 2h arrythmias, chest pain<br />

Ketorolac 0.5–1mg/kg per dose q 6h iv/po GI bleeding, dyspepsia<br />

Antiemetic<br />

Ondansetron 0.3–0.4mg/kg per dose q 4–6h iv/po headache, drowsiness, dry mouth<br />

Granisetron 10µg/kg iv q 4–6h pancytopenia, headache<br />

Sedative<br />

Lorazepam 0.05–0.1mg/kg per dose q 6 h iv/po sedation, respiratory depression<br />

(useful adjunct to ondansetron)<br />

Chlorpromazine 0.5–1mg/kg per dose q 6h iv/po drowsiness, hypotension, seizures<br />

Diphenhydramine 1.25mg/kg per dose q 6h iv/po hypotension, sedation, dizziness<br />

(useful adjunct to chlorpromazine)<br />

NAS, intranasally ; po, orally; q, every; iv, intravenous; GI, gastrointestinal<br />

psychological interventions (e.g. stress management)<br />

and supportive care during attacks when all<br />

else fails. 40 Avoidance of known triggers,<br />

especially dietary, can reduce the episode<br />

frequency. In some cases, stress management<br />

strategies through the aid of a psychologist can<br />

attenuate the effects of positive or negative stressors.<br />

41<br />

Daily use of prophylactic medications is based on<br />

empiric therapy that is traditionally used to treat<br />

other disorders, including migraines, epilepsy,


300<br />

Cyclic vomiting syndrome<br />

gastrointestinal dysmotility and birth control.<br />

Although there are no evidence-based guidelines,<br />

we generally consider prophylaxis in children who<br />

have either frequent episodes, e.g. more than one<br />

episode per month, or severe episodes (prolonged<br />

for more than 3–5 days), debilitating (associated<br />

with hospitalization) or disabling (e.g. missing<br />

school days). 2 The goal of prophylaxis is to prevent<br />

attacks altogether, and if unsuccessful to at least to<br />

reduce the frequency, duration or intensity<br />

(number of emeses) of episodes.<br />

A family history of migraines is a strong indicator<br />

(79%) of a positive response to anti-migraine<br />

therapy. 5 In addition, associated symptoms of<br />

headache, photophobia and phonophobia should<br />

make one consider starting anti-migraine prophylaxis<br />

with propranolol, cyproheptadine, or<br />

amitriptyline with respective efficacy (more than<br />

50% reduction in frequency or severity of<br />

episodes) rates of 52%, 39% and 67%, respectively,<br />

based on our data. Propranolol is contraindicated<br />

in asthmatics, has side-effects of tiredness and can<br />

be monitored by a resting pulse decline of 15–20<br />

beats/min. Cyproheptadine can cause tiredness<br />

and an increase in appetite and weight. 4 Pizotifen,<br />

available in Europe, Canada and Australia, acts<br />

similarly but with fewer side-effects. 42<br />

Amitriptyline has been the most effective prophylactic<br />

agent in our experience, but it can cause<br />

arrhythmias, especially in those with a prolonged<br />

QTc interval. 43 Based on current recommendations,<br />

prior to starting amitriptyline, we evaluate<br />

all candidate patients by checking their QTc interval<br />

on electrocardiogram.<br />

Other prophylactic agents that have been used<br />

include phenobarbital, valproate, gabapentin and<br />

carbamazepine. 44 Although they are specifically<br />

indicated when spike and waves are noted on the<br />

electroencephalogram, they are increasingly used in<br />

both migraine and CVS prophylaxis. Erythromycin<br />

has been useful as a prokinetic agent 34 and low-dose<br />

estrogen birth control can be useful in teenage girls<br />

who have catemenial CVS. We also note that the<br />

efficacy rates in the open-label trials above must be<br />

interpreted cautiously, because of the large placebo<br />

effect (70%) that has been described from consultation<br />

alone prior to instituting therapy.<br />

Abortive therapy is medication taken at the onset<br />

of an episode or even earlier during the prodromal<br />

phase, ideally to abort the episode altogether or<br />

reduce the duration or severity of the episode.<br />

Abortive therapy should be considered for those<br />

who have sporadic episodes that occur less than<br />

once per month and who prefer not taking<br />

prophylaxis or those who have breakthrough<br />

episodes while on prophylaxis. 2,40 Because<br />

patients with intractable emesis are unable to<br />

tolerate oral medication, these medications<br />

usually have to be administered parenterally.<br />

The primary anti-migraine therapy includes 5-<br />

HT1B/1D agonists commonly used for migraine<br />

headaches. In our experience, success rates are<br />

higher in those children with migraine-associated<br />

CVS, when used early in the episode, and in those<br />

with episodes less than 24h. In our patients,<br />

sumatriptan administered via oral, subcutaneous<br />

or intranasal routes has a 51% efficacy rate, as<br />

compared with 65% in headaches. The sensation<br />

of substernal and neck burning rarely occurs with<br />

intranasal administration.<br />

Supportive care is used whenever the child is in a<br />

break-through episode that fails to respond to<br />

abortive therapy. Supportive care includes intravenous<br />

fluids, non-stimulating environment,<br />

antiemetics, sedation and analgesia. 2,40<br />

Intravenous fluids with high dextrose concentration<br />

(10%) and electrolytes have a 42% efficacy<br />

alone in our experience. A quiet, dark single room<br />

may be helpful. Analgesia is not routinely used in<br />

our experience but occasionally can include<br />

narcotic agents.<br />

Ondansetron, a 5-HT3 antagonist, is primarily<br />

used as an antiemetic with efficacy rates around<br />

62%. 45 More effective at the higher dosing of<br />

0.3–0.4mg/kg per dose, the side-effect profile has<br />

been excellent with few reports of drowsiness, dry<br />

mouth and headache. 10 It generally reduces both<br />

nausea and vomiting, but only rarely aborts the<br />

episode. The addition of a γ-aminobutyric acid<br />

(GABA) inhibitor, lorazepam, provides sedation<br />

that relieves intractable nausea. When all else<br />

fails, we use a combination of chlorpromazine<br />

and diphenhydramine primarily to provide sleep<br />

for relief from symptoms. In our experience,<br />

phenothiazine antiemetics (D2 antagonists), have<br />

poor (15%) efficacy in this disorder, even less than<br />

our placebo responses, suggesting that dopaminergic<br />

pathways are not involved.


Summary<br />

Recurrent vomiting is one of the most common<br />

causes for referral to the pediatric gastroenterologist.<br />

Improved recognition of CVS as a definable<br />

disorder with established criteria for diagnosis has<br />

come with time, and support of two recent scientific<br />

symposia. The Cyclic Vomiting Syndrome<br />

Association has been invaluable in providing<br />

family support for CVS patients as well as supporting<br />

research efforts in CVS. As work in understanding<br />

metabolic defects and the pathophysiological<br />

cascade(s) responsible for brain–gut<br />

interactions in CVS continues to define this disor-<br />

REFERENCES<br />

1. Li BUK. Cyclic vomiting syndrome and abdominal<br />

migraine. Int Semin Pediatr Gastroenterol 2000; 9: 1–9.<br />

2. Li BUK, Balint J. Cyclic vomiting syndrome: evolution<br />

in our understanding of a brain–gut disorder. In Adv<br />

Pediatr 2000; 47: 117–160.<br />

3. Abu-Arafeh I, Russel G. Cyclic vomiting syndrome in<br />

children: a population based study. J Pediatr<br />

Gastroenterol Nutr 1995; 21:454–458.<br />

4. Pfau BT, Li BUK, Murray RD et al. Differentiating cyclic<br />

from chronic vomiting patterns in children: quantitative<br />

criteria and diagnostic implications. Pediatr 1996; 97:<br />

364–368.<br />

5. Li B UK, Murray RD, Heitlinger LA. Is cyclic vomiting<br />

syndrome related to migraine? J Pediatrics 1999; 134:<br />

567–572.<br />

6. Heberden W. Commentaries on the History and Causes<br />

of Diseases, 3rd edn. London: Payne & Foss, 1806, cited<br />

by Hammond J. The late sequelae of recurrent vomiting<br />

of childhood. Dev Med Child Neurol 1974; 16: 15–22.<br />

7. Gee S. On fitful or recurrent vomiting. St Bartholemew<br />

Hosp Rev 1882; 18: 1–6.<br />

8. Whitney HB. Cyclic vomiting: a brief review of this<br />

affection as illustrated by a typical case. Arch Pediatr<br />

1898; 15: 839–845.<br />

9. Rachford BK. Recurrent vomiting. Arch Pediatr 1904; 21:<br />

881–891.<br />

10. Li BUK, Fleisher DR. Cyclic vomiting syndrome:<br />

features to be explained by a pathophysiologic model.<br />

Dig Dis Sci 1999; 44: 13S–18S.<br />

11. Shepherd HA, Harvey J, Jackson A et al. Recurrent<br />

retching and gastric mucosal prolapse: a proposed<br />

prolapse gastropathy syndrome. Dig Dis Sci 1984; 29:<br />

121–128.<br />

12. Olson AD, Li BUK. The diagnostic evaluation of<br />

children with cyclic vomiting: a cost-effectiveness<br />

assessment. J Pediatr 2002; 141: 724–728.<br />

13. Li BUK. Cyclic vomiting: the pattern and syndrome<br />

paradigm. J Pediatr Gastroenterol Nutr 1995; 21(Suppl<br />

1): S6–S10.<br />

14. Lucarelli S, Corrado G, Pelliccia A et al. Cyclic vomiting<br />

syndrome and food allergy/intolerance in seven<br />

Summary<br />

301<br />

der better, greater understanding will no doubt<br />

result in improved therapeutic strategies for<br />

patients suffering from CVS.<br />

Acknowledgments<br />

We would like to acknowledge Abid Kagalwalla for<br />

managing our CVS database and providing us with<br />

the appropriate data for this chapter. Dr Li was<br />

supported by funding from the Cyclic Vomiting<br />

Syndrome Association, Grant Healthcare<br />

Foundation and NINS.<br />

children: a possible association. Eur J Pediatr 2000; 159:<br />

360–363.<br />

15. Rinaldo P. Mitochondrial fatty acid oxidation disorders<br />

and cyclic vomiting syndrome. Dig Dis Sci 1999; 44:<br />

97S–102S.<br />

16. Boles RG, Williams JC. Mitochondrial disease and cyclic<br />

vomiting syndrome. Dig Dis Sci 1999; 44: 103S–107S.<br />

17. Symon DNK. Is cyclical vomiting an abdominal form of<br />

migraine in children? Dig Dis Sci 1999; 44: 23S–25S.<br />

18. Barlow CF. The periodic syndrome: cyclic vomiting and<br />

abdominal migraine. Clin Dev Med 1984; 91: 76–92.<br />

19. Lance JW, Anthony M. Some clinical aspects of<br />

migraine. Arch Neurol 1966; 15: 356–361.<br />

20. Salmon MA. The evolution of adult migraine through<br />

childhood migraine equivalents. In Lanzi G, Balottin U,<br />

Cernibori A, eds. Headache in Children and<br />

Adolescents. Amsterdam: Elvesier Science Publishers,<br />

1989: 27–32.<br />

21. Raskin NH, Hosobuchi Y, Lamb S. Headache may arise<br />

from perturbation of the brain. Headache 1987; 27:<br />

416–420.<br />

22. Weiller C, May A, Limmroth V et al. Brain stem<br />

activation in spontaneous human migraine attacks. Nat<br />

Med 1995; 1: 658–660.<br />

23. Welch KM, Nagesh V, Aurora SK. Periaqueductal gray<br />

matter dysfunction in migraine: cause or the burden of<br />

illness? Headache 2001; 41: 629–637.<br />

24. Hirano M, Pavlakis SG. Mitochondrial myopathy:<br />

encephalopathy, lactic acidosis, and strokelike episodes<br />

(MELAS): current concepts. J Child Neurol 1994; 9:<br />

4–13.<br />

25. Wolfe SM, Adler R. A syndrome of periodic hypothalamic<br />

discharge. Am J Med 1964; 36: 956–967.<br />

26. Sato T, Uchigata Y, Uwadana N et al. A syndrome of<br />

periodic adrenocorticotropin and vasopressin discharge.<br />

J Clin Endocrinol Metab 1982; 54: 517–522.<br />

27. Sato T, Igarashi M, Minami S et al. Recurrent attacks of<br />

vomiting, hypertension, and psychotic depression: a<br />

syndrome of periodic catecholamine and prostaglandin<br />

discharge. Acta Endocrinol 1988; 117: 189–197.


302<br />

Cyclic vomiting syndrome<br />

28. Taché Y, Martinez V, Million M et al. Stress and the<br />

gastrointestinal tract III. Stress–related alterations of gut<br />

motor function: role of brain corticotropin-releasing<br />

factor receptors. Am J Physiol-Gastrointest Liver Physiol<br />

2001; 280: G173–G177.<br />

29. Taché Y. Cyclic vomiting syndrome: the corticotropinreleasing-factor<br />

hypothesis. Dig Dis Sci 1999; 44:<br />

79S–86S.<br />

30. Axelrod FB, Zupanc M, Hilz MJ et al. Ictal SPECT<br />

during autonomic crisis in famial dysautonomia.<br />

Neurology 2000; 55: 122–125.<br />

31. To J, Issenman RM, Kamath MV et al. Evaluation of<br />

neurocardiac signals in pediatric patients with cyclic<br />

vomiting syndrome through power spectral analysis of<br />

heart rate variability. J Pediatr 1999; 135: 363–366.<br />

32. Rashed R, Abell TL, Familoni BO et al. Autonomic<br />

function in cyclic vomiting syndrome and classic<br />

migraine. Dig Dis Sci 1999; 44: 74S–78S.<br />

33. Chong SKF. Electrogastrography in cyclic vomiting<br />

syndrome. Dig Dis Sci 1999; 44: 64S–73S.<br />

34. Vanderhoof JA, Young R, Kaufmann SS et al. Treatment<br />

of cyclic vomiting syndrome in childhood with<br />

erythromycin. J Pediatr Gastroenterol Nutr 1993; 17:<br />

387–391.<br />

35. Wood JD. Enteric nervous control of motility in the<br />

upper gastrointestinal tract in defensive states. Dig Dis<br />

Sci 1999; 44: 44S–52S.<br />

36. Boles RG, Adams K, Ito M, Li BUK. Maternal<br />

inheritance in cyclic vomiting syndrome with<br />

neuromuscular disease. Am J Med Genet 2003; 120A:<br />

474–482.<br />

37. Abell TL, Chung HK, Malagelada JR. Idiopathic cyclic<br />

nausea and vomiting – a disorder of gastrointestinal<br />

motility? Mayo Clin Proc 1988; 63: 1169–1175.<br />

38. Fleisher DR, Matar M. The cyclic vomiting syndrome: a<br />

report of 71 cases and literature review. J Pediatr<br />

Gastroenterol Nutr 1993; 17: 361–369.<br />

39. Brown JB, Li BUK. Recurrent vomiting in children. Clin<br />

Perspect Gastroenterol 2002; 35–39.<br />

40. Li BUK. Cyclic vomiting syndrome. Curr Treat Options<br />

Gastroenterol 2000; 3: 395–402.<br />

41. Forbes D, Withers G, Silburn S et al. Psychological and<br />

social characteristics and precipitants of vomiting in<br />

children with cyclic vomiting syndrome. Dig Dis Sci<br />

1999; 44: 19S–22S.<br />

42. Symon DNK. Double-blind placebo-controlled trial of<br />

pizotifen syrup in the treatment of abdominal migraine.<br />

Arch Dis Child 1995; 72: 48–50.<br />

43. Prakash C, Clouse RE. Cyclic vomiting syndrome in<br />

adults: clinical features and response to tricyclic<br />

antidepressants. Am J Gastroenterol 1999; 94:<br />

2855–2859.<br />

44. Gokhale R, Huttenlocher PR, Brady L et al. Use of<br />

barbiturates in the treatment of cyclic vomiting during<br />

childhood. J Pediatr Gastroenterol Nutr 1997; 25: 64–67.<br />

45. Li BUK, Robbins JL, Vu KT et al. Morbidity and<br />

treatment in cyclic vomiting syndrome.<br />

Gastroenterology 1996; 110: 25A.<br />

46. Li BUK. Cyclic vomiting syndrome: new understanding<br />

of an old disorder. Contemp Pediatr 1996; 13: 49.


21<br />

Introduction<br />

Acute and chronic pancreatitis<br />

Michelle M Pietzak<br />

Pancreatitis commonly occurs in the pediatric<br />

population, but it mostly remains undiagnosed.<br />

The clinician treating acute cases must have a low<br />

threshold to test for the non-specific and common<br />

symptoms that occur with this inflammatory<br />

process. Often, the symptoms are dismissed as<br />

occurring from an acute gastroenteritis or viral<br />

infection.<br />

Pancreatitis can be defined as an inflammatory<br />

process of the pancreas which occurs in the clinical<br />

setting of characteristic abdominal and back<br />

pain, accompanied by elevations of the pancreatic<br />

enzymes. The different types of pancreatitis can<br />

be characterized into five main categories according<br />

to the timing of the illness, clinical symptoms,<br />

family history and radiographic findings (acute,<br />

chronic, hereditary, hemorrhagic and necrotic).<br />

Acute pancreatitis is usually a short, self-limited<br />

process characterized by the classic symptoms of<br />

nausea, vomiting, anorexia, epigastric pain and<br />

back pain. Levels of serum amylase and lipase are<br />

markedly elevated in the acute inflammatory<br />

response. Although a child may suffer from<br />

repeated bouts of acute pancreatitis, the symptoms<br />

and enzyme elevations should completely resolve<br />

between attacks. Pancreatic function remains<br />

intact and the pancreatic morphology is undisturbed.<br />

If the signs and symptoms of inflammation are<br />

progressive, this may lead to chronic pancreatitis.<br />

Chronicity may be accompanied by either temporary<br />

or permanent loss of both exocrine and<br />

endocrine function. The severe pain that accompanies<br />

this condition can be debilitating and lead<br />

to dependence upon narcotics. Within the gland<br />

itself, protein plugs may calcify, indicating<br />

advanced and chronic disease (Figure 21.1).<br />

Patients with hereditary pancreatitis, by definition,<br />

will have a family history of the disease. These<br />

diseases are autosomal dominant; affected family<br />

members will often present with recurrent bouts of<br />

acute pancreatitis starting in childhood.<br />

Hemorrhagic and necrotic pancreatitis are uncommon<br />

in the pediatric population, but are a significant<br />

cause of morbidity and mortality from the<br />

disease. A secondary bacterial infection can occur<br />

within the gland, leading to bacteremia, shock and<br />

resultant multi-organ system failure.<br />

Figure 21.1 Kidney, ureters and bladder radiograph with<br />

calcified pancreas of a 6-year-old female who presented to<br />

the emergency department with a 3-month history of<br />

abdominal pain and distension due to severe pancreatitis.<br />

Plain film revealed ascites and extensive pancreatic calcifications<br />

(arrows). Ascitic fluid was markedly hemorrhagic<br />

and the endoscopic retrograde cholangiopancreatogram<br />

was normal. Patient was later discovered to have two<br />

different mutations in the CFTR gene.<br />

303


304<br />

Acute and chronic pancreatitis<br />

Incidence and prevalence<br />

Although pancreatitis is seen more often in the<br />

adult rather than the pediatric population, it is<br />

likely to be underdiagnosed. Its true identification<br />

requires a high index of suspicion on the part of<br />

the clinician with a prompt evaluation before its<br />

resolution. In the literature, most publications on<br />

this disease in childhood are restricted to reports<br />

of either an isolated patient or small clusters of<br />

patients. Because of this, the true incidence and<br />

prevalence of the disease in this population are<br />

unknown. The most common pancreatic disorder<br />

in children is thought to be acute pancreatitis,<br />

with cystic fibrosis the second most common.<br />

Etiology and pathophysiology<br />

The clinical characteristics of acute pancreatitis<br />

follow a similar pattern, despite its varied causes.<br />

Damage occurs to the pancreatic acinar cell by an<br />

inciting event, leading to premature activation of<br />

the digestive enzymes while still within the cell.<br />

This inciting event may be infectious, traumatic,<br />

metabolic, drug-associated or related to an underlying<br />

anatomic anomaly. An inflammatory<br />

response occurs to these damaged cells, activating<br />

platelets and the complement system. Pro-inflammatory<br />

cytokines are released, which include<br />

nitric oxide, interleukin-1, platelet activating<br />

factor and tumor necrosis factor-α. 1 These inflammatory<br />

mediators, as well as the release of additional<br />

free radicals and other vasoactive<br />

substances, damage the gland directly. This can<br />

lead to edema, ischemia, necrosis and eventual<br />

loss of glandular tissue and atrophy. Systemic<br />

shock may occur in severe cases, as demonstrated<br />

by tachycardia, hypotension, hypoxia and adult<br />

respiratory distress syndrome (ARDS).<br />

While the majority of adult cases of pancreatitis<br />

are related to either alcohol consumption or gallstone<br />

disease, the causes of pediatric pancreatitis<br />

are more varied. In adolescent females, gallstone<br />

pancreatitis is not uncommon. However, young<br />

children with this disease have often been exposed<br />

to a recent trauma, infection, or medication as the<br />

inciting event. Recurrent pancreatitis in childhood<br />

can be attributed to hereditary pancreatitis, an<br />

anatomic variant of the pancreatic or biliary tree,<br />

or an underlying systemic or metabolic disorder.<br />

Despite exhaustive investigation, up to 25% of<br />

pediatric patients will not have an attributable<br />

cause for their acute pancreatitis. The conditions<br />

associated with acute and chronic pancreatitis in<br />

childhood are outlined in Table 21.1. 1–17<br />

Anatomic abnormalities<br />

Congenital defects in the pancreas are rare, but if<br />

left uncorrected they can lead to chronic pancreatitis.<br />

Pancreatic divisum is the most common<br />

anatomic variant. 18 It occurs when the dorsal and<br />

ventral pancreatic ducts fail to fuse during fetal<br />

development, directing flow primarily to the<br />

dorsal duct. 19 Some believe this variant to be a<br />

significant cause of recurrent pancreatitis, which<br />

requires either endoscopic or surgical correction.<br />

Others think that pancreatic divisum is a variant of<br />

normal, and that most people with this anatomy<br />

will not experience pancreatitis.<br />

Changes in anatomy which result in duodenal<br />

obstruction can also lead to pancreatitis. Reasons<br />

for this include strictures, tumors, duplications or<br />

diverticula of the duodenum or pancreas, and<br />

duodenal hematoma from either accidental or nonaccidental<br />

trauma (child abuse).<br />

Traumatic causes<br />

Trauma is possibly the most common cause of<br />

pancreatitis in childhood, and is probably underestimated.<br />

Motor vehicle and bicycle accidents can<br />

result in blunt trauma to the pancreas from seatbelts<br />

and handlebars. Findings on physical examination<br />

consistent with trauma, in the absence of a<br />

reliable history, should raise suspicion for child<br />

abuse. There are often other associated intraabdominal<br />

injuries in cases of significant blunt<br />

abdominal trauma. Duodenal hematoma and<br />

intestinal perforation are not uncommon.<br />

Infectious causes<br />

A variety of organisms account for a significant<br />

number of cases of pancreatitis worldwide, including<br />

bacteria, viruses and parasites. The Escherichia<br />

coli strain which produces verotoxin and is associated<br />

with hemolytic uremic syndrome can also<br />

cause pancreatitis. Viruses such as varicella and


Etiology and pathophysiology 305<br />

Table 21.1 Conditions associated with pancreatitis in the pediatric population (from references 1–17)<br />

Idiopathic<br />

Up to 25% of cases<br />

Anatomic<br />

Ampullary diverticulum<br />

Ampullary stenosis<br />

Annular pancreas<br />

Biliary tract malformations<br />

Choledochal cyst<br />

Choledochocele<br />

Choledochopancreaticoductal junction anomaly<br />

Cholelithiasis<br />

Common bile duct: absence or anomalous<br />

insertion<br />

Duodenal obstruction from diverticulum,<br />

hematoma, tumor or stricture<br />

Duodenal ulcer – perforated<br />

Duplication cyst of the common bile duct,<br />

duodenum, gastropancreatic area<br />

Gastric trichobezoar<br />

Pancreatic aplasia<br />

Pancreatic divisum<br />

Pancreatic duct: absence or anomalous insertion<br />

Pancreatic dysplasia<br />

Pancreatic heterotopy<br />

Pancreatic hypoplasia<br />

Pancreatic pseudocyst<br />

Pancreatic tumors<br />

Sclerosing cholangitis<br />

Sphincter of Oddi dysfunction<br />

Traumatic<br />

Blunt injury to the abdomen<br />

Burns<br />

Contrast from ductal imaging (endoscopic<br />

retrograde cholangiopancreatography,<br />

percutaneous transhepatic cholangiography)<br />

Head trauma<br />

Non-accidental trauma (child abuse)<br />

Radiation to the abdomen<br />

Surgical trauma<br />

Total body cast<br />

Infectious<br />

Ascaris lumbricoides (duct obstruction)<br />

Campylobacter fetus<br />

Clonorchis sinensis (duct obstruction)<br />

Coxsackie B virus<br />

Cryptosporidium<br />

Cytomegalovirus<br />

Echovirus<br />

Enterovirus<br />

Escherichia coli (verotoxin-producing)<br />

Hepatitis A virus<br />

Hepatitis B virus<br />

Human immunodeficiency virus (HIV)<br />

Influenza A virus<br />

Influenza B virus<br />

Legionnaire’s disease<br />

Leptospirosis<br />

Malaria<br />

Measles<br />

Mumps<br />

Mycobacteria<br />

Mycoplasma<br />

Rubella<br />

Rubeola<br />

Toxoplasma<br />

Typhoid fever<br />

Varicella<br />

Yersinia<br />

Hereditary/Metabolic/Systemic<br />

α1-Antitrypsin deficiency<br />

Anorexia nervosa<br />

Autoimmune disease<br />

Brain tumor<br />

Bulimia<br />

Collagen vascular disease<br />

Congenital partial lipodystrophy<br />

Crohn’s disease<br />

Cystic fibrosis<br />

Dehydration<br />

Dermatomyositis<br />

Diabetes mellitus (ketoacidosis)<br />

Glycogen storage disease types Ia and Ib<br />

Hemochromatosis<br />

Hemolytic–uremic syndrome<br />

Honoch–Schönlein purpura<br />

Hereditary pancreatitis<br />

Hyperalimentation<br />

Hypercalcemia<br />

Hyperlipidemia types I, IV and V<br />

Hyperparathyroidism<br />

Hypertriglyceridemia<br />

Hypothermia<br />

Hypovolemia<br />

Inborn errors of metabolism (organic academia,<br />

cytochrome-C oxidase deficiency)<br />

Juvenile tropical pancreatitis<br />

Kawasaki disease<br />

Liver disease<br />

Malnutrition<br />

Organic acidemias (homocystinuria, isovaleric<br />

acidemia, methylmalonic acidemia, maple<br />

syrup urine disease)<br />

Periarteritis nodosa<br />

Peritonitis<br />

Refeeding syndrome<br />

Renal failure with uremia<br />

Reye’s syndrome<br />

Sarcoidosis<br />

Sepsis<br />

Shock<br />

Systemic lupus erythematosus<br />

Transplantation (bone marrow, heart, kidney, liver,<br />

pancreas)<br />

Ulcerative colitis<br />

Vascular diseases<br />

Uremia<br />

Wilson’s disease


306<br />

Acute and chronic pancreatitis<br />

influenza B, associated with Reye’s syndrome,<br />

have also been implicated in causing acute pancreatitis<br />

in childhood. Parasites that are more<br />

common in developing areas, such as Ascaris and<br />

Clonorchis, can migrate into the biliary tree,<br />

leading to obstructive jaundice and pancreatitis.<br />

Left untreated, this obstruction can lead to severe<br />

portal hypertension, liver failure and death.<br />

Children with AIDS are at higher risk for developing<br />

acute pancreatitis. One Italian study in symptomatic<br />

HIV-infected children demonstrated<br />

pancreatic biochemical abnormalities in 15% of<br />

the patients. 20 However, the elevated enzymes did<br />

not correlate with clinical evidence of acute<br />

pancreatitis. Although an adult autopsy series<br />

showed pancreatic lesions in about 30% of AIDS<br />

patients, fewer than 10% had clinical symptoms. 21<br />

In children with HIV at autopsy, pancreatic<br />

involvement, defined as edema, inflammation,<br />

fibrosis, inspissated material and enlarged<br />

Langerhan’s islets, was frequent. 22 However,<br />

tumors and involvement of opportunistic infectious<br />

was rare. HIV-positive patients can develop<br />

elevated enzymes, owing to the HIV infection<br />

itself, or owing to other co-infections, such as<br />

cytomegalovirus (CMV), Toxoplasma, mycobacteria<br />

and Cryptosporidium. 17 Many of the pharmacological<br />

agents used to inhibit HIV can cause<br />

pancreatitis (see Medications). Hyperamylasemia<br />

without pancreatitis can occur from renal failure,<br />

AIDS-associated nephropathy, or salivary hyperamylasemia<br />

from parotid gland disease due to<br />

HIV. 23 Kaposi’s sarcoma and lymphoma can also<br />

affect the pancreas. 24<br />

Hereditary, metabolic and systemic diseases<br />

The most common inherited disease of the<br />

exocrine pancreas is thought to be cystic fibrosis<br />

(CF). 25 This autosomal recessive disease is caused<br />

by mutations in the CF transmembrane conductance<br />

regulator gene (CFTR). CFTR is located on<br />

the apical membrane of the epithelial cells that<br />

line the pancreatic ducts. The transporter<br />

promotes dilution and alkalinization of the pancreatic<br />

secretions and they flow through the ductular<br />

network. CF may be one of the most common<br />

causes of pancreatitis in childhood, but pancreatitis<br />

itself is a rare presenting feature of CF. 26 It is<br />

believed that up to 2% of individuals with CF<br />

experience pancreatitis as a result of ductal<br />

plugging due to mutant CFTR. 27 CF is the only<br />

known hereditary disease in which there can be<br />

both pancreatitis, and exocrine insufficiency in the<br />

absence of pancreatic inflammation. 28 However,<br />

those patients with CF and pancreatic insufficiency<br />

do not develop acute relapsing pancreatitis,<br />

presumably because of the loss of functional<br />

acinar tissue. 7 Even in the absence of lung disease,<br />

there appears to be a strong correlation between<br />

specific CFTR mutations and idiopathic and<br />

chronic pancreatitis. 29–31 Patients with recurrent<br />

pancreatitis without an obvious cause should be<br />

screened for CF with a sweat chloride test. CFTR<br />

mutational analysis is also commercially available.<br />

The role of CFTR in pancreatitis and other diseases<br />

of the pancreas is the subject of ongoing research.<br />

The second most common cause of chronic<br />

pancreatitis in childhood is believed to be hereditary<br />

pancreatitis. 32 The gene defects were reported<br />

in 1996, 33 and help explain some of the pathophysiology<br />

of the non-hereditary forms of both<br />

acute and chronic pancreatitis. 34,35 The two<br />

known types of hereditary pancreatitis are clinically<br />

similar, and involve different mutations<br />

within the same gene. Both types are autosomal<br />

dominant with 80% penetrance. 36 The majority of<br />

affected patients report symptoms before the age of<br />

15 years, with some symptomatic even before the<br />

age of 5 years. The gene involved is located on<br />

chromosome 7q35, and codes for cationic<br />

trypsinogen (PRSS1). Both mutations result in a<br />

form of trypsin that resists degradation by<br />

mesotrypsin and enzyme Y, 26 allowing trypsinogen<br />

to become activated to trypsin within the pancreas<br />

instead of within the duodenum. This leads to<br />

uncontrolled activation of other pancreatic<br />

enzymes within the acinar cell, resulting in<br />

autodigestion and inflammation. The attacks of<br />

acute pancreatitis are remarkably only intermittent.<br />

It is thought that this uncontrolled activation<br />

of other enzymes occurs only when trypsin<br />

exceeds the capacity of pancreatic secretory<br />

trypsin inhibitor, the ‘secondary brake’ within the<br />

pancreatic gland. Identification of those with<br />

hereditary pancreatitis is critical, as affected<br />

patients are at increased risk for pseudocysts,<br />

pancreatic adenocarcinoma, and exocrine and<br />

endocrine failure. 37<br />

Since the discovery that hereditary pancreatitis<br />

can be caused by mutations in PRSS1, researchers<br />

have been searching for other potential candidate


genes that may predispose patients to chronic<br />

pancreatitis. Given that the proposed mechanism<br />

for pancreatitis with PRSS1 mutations is decreased<br />

inactivation, one candidate gene is that encoding<br />

for pancreatic secretory trypsin inhibitor or serine<br />

protease inhibitor, Kazal type 1 (SPINK1). 38<br />

SPINK1 mutations have been reported in a wide<br />

phenotype of conditions, including familial,<br />

hereditary, tropical and idiopathic chronic pancreatitis.<br />

39–42 Whether or not SPINK1 mutations<br />

modify an already underlying genetic disease has<br />

yet to be elucidated. 38<br />

Pancreatitis has been a reported complication in<br />

children who have received heart, kidney, liver,<br />

Etiology and pathophysiology 307<br />

pancreas or bone marrow transplants. 8 Following<br />

liver transplantation, it can be life-threatening,<br />

and is associated with a higher risk for infectious<br />

peritonitis and emergency retransplantation. 9 As<br />

with autoimmune and collagen–vascular diseases,<br />

it is difficult to distinguish between the contributions<br />

of medications versus the primary disease<br />

process to the development of the pancreatitis.<br />

Medications<br />

Numerous medications and naturally occurring<br />

toxins have been reported as a cause of pancreatitis<br />

(Table 21.2). 1–4,6,17,23,43–45 Often, however, the<br />

Table 21.2 Drugs and toxins associated with pancreatitis (from references 1–4,6,17,23,43–45)<br />

Acetaminophen overdose<br />

Alcohol<br />

Amphetamines<br />

Anticoagulants<br />

Asparaginase<br />

Azathioprine<br />

Boric acid<br />

Bumetanide<br />

Calcium<br />

Carbamazepine<br />

Chlorthalidone<br />

Cholestyramine<br />

Cimetidine<br />

Cisplatin<br />

Clonidine<br />

Clozapine<br />

Corticosteroids<br />

Corticotropin<br />

Cyclophosphamide<br />

Cyproheptadine<br />

Cytarabine<br />

Cytosine arabinoside<br />

Diazoxide<br />

Didanosine<br />

Dideoxycinosine<br />

Dideoxycytidine<br />

Diphenoxylate<br />

Enalapril<br />

Erythromycin<br />

Estrogen<br />

Ethacrynic acid<br />

Furadantin<br />

Furosemide<br />

Heroin<br />

Histamine<br />

Indomethacin<br />

Isoniazid<br />

Lamivudine<br />

Meprobamate<br />

Mercaptopurine<br />

Mesalamine<br />

Methotrexate<br />

Methyldopa<br />

Metronidazole<br />

Non-steroidal anti-inflammatory drugs<br />

Nitrofurantoin<br />

Octreotide<br />

Opiates<br />

Oxyphenbutazone<br />

Organophosphates<br />

Paromomycin<br />

Penicillin<br />

Pentamidine<br />

Phenformin<br />

Piroxicam<br />

Procainamide<br />

Propoxyphene<br />

Propylthiouracil<br />

Ranitidine<br />

Rifampin<br />

Salicylates<br />

Scorpion venom<br />

Spider venom<br />

Sulfasalazine<br />

Sulfonamides<br />

Sulindac<br />

Tetracycline<br />

Thiazides<br />

Tretinoin<br />

Trimethoprim–sulfamethoxazole<br />

Valproic acid<br />

Vincristine<br />

Vitamin D


308<br />

Acute and chronic pancreatitis<br />

relationship between drugs and pancreatitis is<br />

more of association than causation. Classes of<br />

medications most likely to cause pancreatitis<br />

include antibiotics (erythromycin, metronidazole,<br />

nitrofurantoin, penicillin, rifampin), anticonvulsants<br />

(carmbamazepine, valproic acid), antihypertensives<br />

(clonidine, diazoxide, enalapril), antiinflammatories<br />

(corticosteroids, ibuprofen,<br />

indomethacin, sulfasalazine and acetaminophen<br />

when overdosed) and antineoplastic agents<br />

(asparaginase, azathioprine, mercaptopurine,<br />

cyclophosphamide, vincristine). Most of the drugs<br />

mentioned in this chapter have a proposed but<br />

unproven pathophysiology, and very few documented<br />

an established causal relationship to<br />

pancreatitis.<br />

Clinical signs and symptoms<br />

In the pediatric population, the classic signs of<br />

acute pancreatitis include nausea, vomiting,<br />

anorexia and abdominal pain. The pain is classically<br />

located in the epigastrium, with radiation to<br />

the back. However, the pain could also be located<br />

in the periumbilical region, right upper quadrant<br />

or lower chest. 46,47 Eating usually exacerbates the<br />

abdominal discomfort and emesis, which may<br />

progress to biliousness. In the review of systems,<br />

the physician should inquire about rashes, diarrhea,<br />

joint pain and other signs of vasculitis. A<br />

family history of pancreatitis should raise suspicion<br />

for hereditary and metabolic diseases (see<br />

Table 21.1).<br />

More common causes of acute abdominal pain in<br />

childhood may be differentiated from pancreatitis<br />

by a thorough physical examination. If fever is<br />

present, it is usually of low grade. However, tachycardia<br />

and hypotension may be present early in<br />

the course of the disease. Tachypnea with hypoxemia<br />

can indicate developing pulmonary edema.<br />

The child may be icteric and ill-appearing, with a<br />

distended abdomen and decreased bowel sounds,<br />

owing to an ileus, ascites or a mass from a pancreatic<br />

phlegmon or pseudocyst. While lying supine<br />

on the examination table, the patient may experience<br />

some relief of pain when the knees are drawn<br />

up to a flexed trunk. In advanced disease, where<br />

there has been pancreatic hemorrhage or necrosis,<br />

two signs may be present: Grey Turner sign, which<br />

is a blue discoloration of the flank; and Cullen’s<br />

sign, where there is blue discoloration around the<br />

pancreas. The child should be examined for physical<br />

findings of child abuse, especially if there is an<br />

unclear history of trauma to the abdomen.<br />

Evaluation<br />

Laboratory tests<br />

Routine blood tests can usually differentiate acute<br />

pancreatitis from the more common causes of<br />

abdominal pain in childhood. A complete blood<br />

cell count with white cell differential usually<br />

demonstrates leukocytosis with a left shift, and<br />

hemoconcentration from dehydration. Frequent<br />

findings in a routine chemistry panel include<br />

hyperglycemia and elevated levels of total bilirubin,<br />

alanine aminotransferase and aspartate<br />

aminotransferase. Anemia, azotemia, hypoalbuminemia,<br />

hypocalcemia and an elevated lactate<br />

dehydrogenase level suggest advanced disease<br />

with hemorrhage and pancreatic damage.<br />

The most frequently used laboratory tests to screen<br />

for acute pancreatitis are serum amylase and<br />

lipase. Amylase levels begin to rise within 2–12h,<br />

and peak at 12–72h after the initial pancreatic<br />

insult. 48 However, an isolated serum amylase level<br />

has a relatively low sensitivity (75–92%) and<br />

specificity (20–60%). 5 This is because normal<br />

amylase levels may be seen with pancreatitis, and<br />

hyperamylasemia can result from many diseases<br />

that are not of pancreatic origin (Table<br />

21.3). 5,23,48,49 If the amylase level is three to six<br />

times above the upper limit of normal for that<br />

laboratory, the specificity increases for pancreatitis,<br />

but at the expense of sensitivity. 5 More<br />

discriminatory are the measurements of serum<br />

isoamylase levels, which differentiate between<br />

enzymes of salivary and pancreatic origin.<br />

Isoamylase levels should be determined if parotitis<br />

from a viral infection is suspected (such as with<br />

HIV), in the presence of some cancers, and if<br />

ovarian disease is present (Table 21.3).<br />

After serum amylase, serum lipase is the test most<br />

frequently used to confirm acute pancreatitis.<br />

Lipase levels begin to increase 4–8h after the onset<br />

of symptoms, and also peak at about 24h. 50<br />

However, lipase levels remain elevated for a longer<br />

period of time than amylase levels, decreasing over


Table 21.3 Conditions other than pancreatitis associated with elevated serum amylase (from<br />

references 5,23,48,49)<br />

Abdominal aortic aneurysm (pancreatic amylase<br />

elevation only)<br />

Alcoholism<br />

Anorexia nervosa (salivary amylase elevation only)<br />

Appendicitis (pancreatic amylase elevation only)<br />

Biliary duct obstruction (parasite, stone, tumor)<br />

(pancreatic amylase elevation only)<br />

Biliary tract disease (pancreatic amylase elevation<br />

only)<br />

Bulimia (salivary amylase elevation only)<br />

Burns<br />

Cardiopulmonary bypass<br />

Choledocholithiasis (pancreatic amylase elevation<br />

only)<br />

Cirrhosis<br />

Cystic fibrosis<br />

Diabetic ketoacidosis<br />

Drugs<br />

Hepatitis<br />

Heroin addiction<br />

Intestinal infarction (pancreatic amylase elevation<br />

only)<br />

Intestinal obstruction (pancreatic amylase elevation<br />

only)<br />

Intestinal perforation (pancreatic amylase elevation<br />

only)<br />

8–14 days. Serum lipase levels have a reported<br />

clinical sensitivity of 86–100% and specificity of<br />

50–99%. 51 If the serum lipase level is greater than<br />

three times the upper limit of normal for that laboratory,<br />

sensitivity and specificity can be increased<br />

to 99–100%. 5 However, a significantly elevated<br />

lipase level, in the presence of a normal amylase<br />

level, has been reported in esophagitis, hypertriglyceridemia,<br />

renal insufficiency, acute cholecystitis<br />

and non-pancreatic sources of lipolytic<br />

enzymes due to malignant tumors. 52<br />

Clinical sensitivity for the diagnosis of acute<br />

pancreatitis increases to 94% by using serum<br />

amylase and lipase level determinations in parallel.<br />

5,52 It is important to note, however, that the<br />

degrees of elevation of the amylase and lipase<br />

levels in the plasma in no way reflect the severity<br />

of the pancreatic disease process itself. There are<br />

serum enzymes more sensitive than amylase<br />

which do correlate with disease severity, such as<br />

Evaluation 309<br />

Lung cancer (salivary amylase elevation only)<br />

Macroamylasemia<br />

Opiates<br />

Ovarian cyst (salivary amylase elevation only)<br />

Ovarian tumor (salivary amylase elevation only)<br />

Pancreatic duct obstruction (parasite, stone, tumor)<br />

(pancreatic amylase elevation only)<br />

Pancreatic tumor (pancreatic amylase elevation<br />

only)<br />

Parotitis (salivary amylase elevation only)<br />

Peptic ulcer – perforated (pancreatic amylase<br />

elevation only)<br />

Peritonitis (pancreatic amylase elevation only)<br />

Pneumonia (salivary amylase elevation only)<br />

Prostate tumors (salivary amylase elevation only)<br />

Renal insufficiency<br />

Renal transplant<br />

Ruptured ectopic pregnancy<br />

Salivary duct obstruction (salivary amylase<br />

elevation only)<br />

Salpingitis (salivary amylase elevation only)<br />

Trauma (to the head or abdomen)<br />

Viral infections (mumps) (salivary amylase elevation<br />

only)<br />

plasma immunoreactive cationic trypsin, pancreatic<br />

elastase I and phospholipase A2. 5 However,<br />

these tests are not readily available outside<br />

research centers.<br />

The diagnosis of chronic pancreatitis relies not<br />

only on clinical and radiographic findings (see<br />

next section), but also on tests of pancreatic function.<br />

53 The ‘gold standard’ for the assessment of<br />

pancreatic function involves direct testing for<br />

pancreatic insufficiency. This is accomplished via<br />

the administration of intravenous secretin or<br />

cholecystokinin, and measuring the levels of bicarbonate<br />

and pancreatic enzymes from the pancreatic<br />

ductal secretions. To perform this in a pediatric<br />

patient requires endoscopic intubation of the<br />

duodenum, with accurate placement of a catheter<br />

to collect the secretions, under appropriate anesthesia.<br />

If performed correctly, the sensitivity and<br />

specificity of this procedure for the diagnosis of<br />

chronic pancreatitis ranges from 90 to 100%. 54


310<br />

Acute and chronic pancreatitis<br />

Because of the challenges in performing and interpreting<br />

these examinations, they are usually available<br />

only in tertiary centers.<br />

Although not as accurate, non-invasive tests of<br />

pancreatic function are more readily available.<br />

Chronic pancreatitis can be demonstrated by<br />

decreased enzymes in the blood (amylase, lipase,<br />

isoamylase, immunoreactive trypsinogen) or stool<br />

(trypsin, pancreatic elastase I), or increased<br />

amounts of malabsorbed food products (primarily<br />

fat). A recent pediatric study showed that testing<br />

for fecal pancreatic elastase I compared favorably<br />

to the secretin–pancreozymin test, reporting 100%<br />

sensitivity and 96% specificity for pancreatic<br />

insufficiency due to CF. However, because of the<br />

generally poor negative predictive value of these<br />

tests, chronic pancreatitis cannot be excluded with<br />

certainty. False-positive results can be seen with<br />

bacterial overgrowth and other mucosal diseases of<br />

the small bowel.<br />

Radiographic studies<br />

Although the study of choice to delineate pancreatic<br />

changes is abdominal ultrasonography, 55 a<br />

plain abdominal radiograph (kidney, ureters and<br />

bladder; KUB) may demonstrate anomalies. Acute<br />

pancreatitis may result in an ileus, with either<br />

colonic distension or a ‘sentinel’ loop of dilated<br />

small bowel. Obscured psoas margins or a radiolu-<br />

Figure 21.2 Ultrasound demonstrating acute pancreatitis<br />

in a 9-year-old HIV-positive girl. The head and the body of<br />

the pancreas are decreased in echogenicity and diffusely<br />

enlarged.<br />

cent ‘halo’ around the left kidney are also suggestive<br />

of pancreatitis. Calcifications can be seen in<br />

the area of the pancreatic parenchyma and ductal<br />

system with chronic pancreatitis (Figure 21.1).<br />

Changes in pancreatic size, contour and texture<br />

are best appreciated with ultrasound. This modality<br />

is also excellent at identifying ascites,<br />

abscesses, pseudocysts, dilated ducts and gallstone<br />

disease (Figures 21.2–21.5).<br />

Computed tomography (CT), with oral and intravenous<br />

contrast, is useful in managing the complications<br />

of long-standing pancreatitis. 55 CT can<br />

provide more accurate guidance in the aspiration<br />

and drainage of an abscess, phlegmon or pseudocyst.<br />

Prior to any type of surgical intervention,<br />

such as necrostomy (surgical debridement for<br />

necrosis), CT may be utilized for further definition<br />

of the peripancreatic anatomy, and to rule out<br />

other complications, such as a portal vein thrombosis<br />

(Figures 21.6–21.15).<br />

KUB, ultrasound and CT are not adequate in clinical<br />

circumstances under which a detailed anatomy<br />

of the pancreatic and biliary tree is necessary, such<br />

as in chronic pancreatitis and recurrent attacks of<br />

acute pancreatitis. Cholangiopancreatography may<br />

be accomplished by endoscopic retrograde cholangiopancreatography<br />

(ERCP), magnetic resonance<br />

cholangiopancreatography (MRCP), or via a direct<br />

cholangiogram performed either percutaneously or<br />

Figure 21.3 Ultrasound demonstrating sludge in the<br />

gallbladder of the same patient as in Figure 21.2. There is<br />

no gallbladder wall thickening or pericholecystic fluid. No<br />

calculi were identified, but they could be obscured by the<br />

sludge.


Figure 21.4 Ultrasound demonstrating biliary ductal<br />

dilatation in a 7-year-old girl with chronic pancreatitis.<br />

The common bile duct is abnormally dilated, measuring<br />

1.2cm in diameter. This fusiform enlargement of the<br />

common bile duct, proximal to its bifurcation just at the<br />

hilus of the liver, is suggestive of a choledochal cyst.<br />

Figure 21.6 Computed tomography scan of acute<br />

pancreatitis in the abdomen of a 4-year-old boy. Fluid<br />

collections are seen near the pancreatic head and tail<br />

(arrows), consistent with inflammatory changes. There are<br />

areas of hypodensity within the pancreas, representative<br />

of acute pancreatitis.<br />

Evaluation 311<br />

Figure 21.5 Ultrasound of a pseudocyst in an 18-yearold<br />

female with chronic pancreatitis after a severe upper<br />

gastrointestinal bleed, respiratory failure and septic shock.<br />

The pancreas was normal in echogenicity, but a pseudocyst<br />

was seen in the body measuring<br />

2.8x1.9x1cm.<br />

Figure 21.7 Computed tomography scan demonstrating<br />

biliary ductal dilatation in the abdomen of the patient in<br />

Figure 21.4. There is extrahepatic bile duct and pancreatic<br />

duct dilatation (arrows). The pancreas has a normal<br />

density. Intrahepatic ductal dilatation was seen on other<br />

sections. The differential diagnosis in this patient includes<br />

choledochal cyst, Caroli’s disease and sclerosing<br />

cholangitis.


312<br />

Acute and chronic pancreatitis<br />

Figure 21.8 Computed tomography scan of the<br />

abdomen of a 16-year-old morbidly obese male with<br />

chronic pancreatitis. The pancreas demonstrates enhancements<br />

with focal lesions. The head was poorly visualized.<br />

The body and tail are enlarged, consistent with chronic<br />

pancreatitis. Endoscopic retrograde cholangiopancreatography<br />

demonstrated pancreatic divisum.<br />

Figure 21.10 Computed tomography (CT) scan of<br />

calcified chronic pancreatitis in the abdomen of a 14-yearold<br />

obese girl. The patient had complained of abdominal<br />

pain with nausea and vomiting for several months. She<br />

was found to have a hiatal hernia, and underwent laparoscopic<br />

fundoplication. The abdominal pain persisted,<br />

despite a repeat of her ‘slipped’ fundoplication, and this<br />

CT scan was ordered. There is fatty atrophy of the pancreas<br />

with multiple punctuate and small calcifications consistent<br />

with chronic pancreatitis. The pancreatic duct is dilated,<br />

measuring 1.2cm at the uncinate process. There is a calcification<br />

in the duct near the ampulla of Vater (arrow),<br />

which appears to be lodged in the common bile duct,<br />

causing proximal dilatation. There are several small<br />

pseudocysts in the upper abdomen. The largest is in the<br />

lesser sac just inferior to the stomach, measuring 3x4x<br />

3cm, with inflammation present. This patient remains<br />

dependent upon parenteral nutrition.<br />

Figure 21.9 Computed tomography scan of an atrophic<br />

pancreas in the same patient as in Figure 21.8, 1 year<br />

later. The pancreas is essentially absent, with only<br />

minimal tissue identified in the tail. A 4x3.6x3.8cm<br />

cystic mass is present in the region of the former body of<br />

the pancreas, consistent with a pseudocyst (arrow). This<br />

patient went on to develop cirrhosis with portal hypertension<br />

due to portal vein thrombosis. Esophageal, gastric,<br />

splenic and retroperitoneal varices were seen on other<br />

sections. Recanalization of the portal and umbilical veins<br />

was also visualized. Pancreatic atrophy rendered this<br />

patient dependent upon insulin and pancreatic enzyme<br />

replacements.<br />

Figure 21.11 Computed tomography scan of the<br />

abdomen in an 11-year-old girl with chronic pancreatitis<br />

undergoing treatment for acute lymphoblastic leukemia. A<br />

massive 19x5.5x8.5cm pseudocyst is seen in the left<br />

upper quadrant (arrows). There is edema and necrosis<br />

within the pancreatic tissue, and a smaller pseudocyst<br />

within the pancreatic head. There is free fluid within the<br />

abdomen and stranding within the mesentery of the left<br />

upper quadrant consistent with chronic inflammation. The<br />

liver shows fatty infiltration with a normal gallbladder.<br />

This pseudocyst resolved without surgical intervention.


Figure 21.12 Computed tomography scan of the<br />

abdomen in a 10-year-old girl with end-stage renal<br />

disease, found to have bloody drainage from her peritoneal<br />

dialysis catheter. There are diffuse inflammatory<br />

changes around the pancreas which extend up and<br />

around the lateral aspect of the stomach. The pancreas is<br />

low in attenuation and heterogeneous, suggestive of<br />

necrosis. There are multiple fluid collections in the region<br />

of the tail of the pancreas. There is a small amount of<br />

pericholecystic fluid surrounding the anterior wall of the<br />

gallbladder (arrow). Other sections delineated superior<br />

mesenteric and portal vein thrombosis. The kidneys did<br />

not concentrate or excrete contrast, consistent with the<br />

patient’s renal failure.<br />

Figure 21.14 Computed tomography scan of the<br />

abdomen of a teenage boy with recurrent pancreatitis. He<br />

had received high-dose steroids for treatment of non-<br />

Hodgkin’s lymphoma. The pancreas is edematous and<br />

there are multiple, small pseudocysts (arrows) which have<br />

enhancing rims. These findings are consistent with severe<br />

hemorrhagic pancreatitis. The liver is enlarged with low<br />

attenuation consistent with fatty infiltration.<br />

Evaluation 313<br />

Figure 21.13 Computed tomography scan of chronic<br />

pancreatitis with cystic changes in the same patient as in<br />

Figure 21.12, taken 5 months later, demonstrating<br />

pseudocyst formation with extensive cystic changes within<br />

the body and tail of the pancreas. Owing to an intracranial<br />

bleed, the patient’s nutritional status was maintained<br />

on an elemental liquid diet (appropriate for a patient with<br />

renal failure) via a gastrostomy tube.<br />

Figure 21.15 Repeat computed tomography scan of the<br />

abdomen of the patient in Figure 21.14, performed 8<br />

months later. There are areas of necrosis within the<br />

pancreatic body and tail (arrows). A pseudocyst is present<br />

at the pancreatic tail near the splenic hilum. Bones<br />

demonstrate osteopenia, also consistent with high-dose<br />

steroids.


314<br />

Acute and chronic pancreatitis<br />

Figure 21.16 Magnetic resonance imaging (MRI)<br />

demonstrating pancreatitis with ductal dilatation in a<br />

previously healthy 16-year-old female with a 1-month<br />

history of nausea and epigastric pain after meals. Initial<br />

ultrasound examination revealed pancreatic edema and<br />

ductal dilatation. The patient’s amylase and lipase levels<br />

continued to rise, despite the pancreatic rest. MRI<br />

revealed a dilated pancreatic duct (arrow) with increased<br />

signal intensity at the junction of the body and the tail,<br />

consistent with edema.<br />

intraoperatively. These studies may reveal<br />

cholelithiasis, anatomic malformations or biliary<br />

strictures.<br />

ERCP is the study of choice to visualize anatomic<br />

malformations, such as pancreas divisum or anomalous<br />

pancreaticobiliary duct junction. 15,18 ERCP<br />

may be difficult in very small children, owing to<br />

the large diameter of the endoscope required.<br />

ERCP, as opposed to MRCP, has the added benefit<br />

of being a therapeutic modality. Sphincterotomy,<br />

stent placement and stone removal can all be<br />

performed at the time of the initial diagnostic evaluation.<br />

56 Sphincter of Oddi manometry to look for<br />

elevated basal pressures consistent with dysfunction<br />

can also be useful, as these patients may<br />

benefit from endoscopic sphincterotomy. 15 These<br />

procedures should be undertaken only at centers<br />

that have significant experience both in performing<br />

the studies and in managing severe cases of<br />

childhood pancreatitis. A pediatric surgeon should<br />

be available at the center, as complications of<br />

ERCP can include duct perforation or a worsening<br />

of the pancreatitis from the contrast, leading to<br />

abscess, phlegmon or pseudocyst formation<br />

requiring surgical drainage. However, when<br />

performed by experienced endoscopists, therapeu-<br />

Figure 21.17 Magnetic resonance cholangiopancreatography<br />

demonstrating pancreatitis with ductal dilatation in<br />

the same patient as in Figure 21.16. Dilatation of the<br />

pancreatic duct (arrow), common bile duct, and the right<br />

and left main hepatic ducts (arrowheads) are seen, consistent<br />

with an impacted stone at the ampulla of Vater.<br />

tic ERCP in selected pediatric patients has been<br />

reported to have a lower rate of complications than<br />

in the adult population. 57<br />

MRCP should be reserved for patients who are too<br />

small or too clinically unstable to receive general<br />

anesthesia to undergo ERCP. This modality has<br />

proved useful in determining the presence of<br />

pancreaticobiliary disease, the level of biliary<br />

obstruction, and the presence of malignancy or<br />

bile duct calculi (Figures 21.16 and 21.17). 58,59 The<br />

quality of the MRCP images depend upon the<br />

ability of the patient to remain still (which may<br />

require light sedation in small children), and<br />

whether the MRCP equipment and computer<br />

programs are regularly updated. Interpretation also<br />

requires a skilled radiologist familiar with the<br />

various potential pancreaticobiliary tree anomalies.<br />

If an anatomic variant is suspected, and ERCP or<br />

MRCP are not possible, the next appropriate step is<br />

direct imaging of the pancreaticobiliary system<br />

with contrast, performed either percutaneously or<br />

intraoperatively. A percutaneous cholangiogram<br />

may be performed in the presence of dilated ducts,<br />

or through a biliary drain. Biliary drains may have<br />

been placed postoperatively from pancreatic surgical<br />

debridement or orthotopic liver transplant.<br />

This type of study requires a skilled interventional<br />

radiologist. In the absence of dilated ducts or a


iliary drain, transduodenal exploration with<br />

intraoperative pancreatography should be<br />

performed. Like ERCP, intraoperative imaging can<br />

likewise be a therapeutic modality at the time of<br />

initial diagnostic evaluation. If minor or major<br />

stenosis of papillae is found, surgical sphincteroplasty<br />

can be performed concurrently. 60<br />

Treatment<br />

The management of acute, uncomplicated pancreatitis<br />

in childhood is mainly supportive. The<br />

patient should be provided adequate hydration,<br />

pain relief and pancreatic rest. 55 The child should<br />

be made nil per os in severe cases, which will<br />

decrease the cephalic, gastric and intestinal phases<br />

of pancreatic secretion. Continuous or intermittent<br />

nasogastric suction may be required in cases with<br />

ileus or persistent emesis. If the child is expected<br />

to be without enteral feedings for more than 3<br />

days, parenteral nutrition should be initiated to<br />

prevent protein catabolism. Recent studies have<br />

suggested that nasojejunal feedings with an<br />

elemental diet, which bypass the duodenum<br />

completely and therefore do not potently stimulate<br />

pancreatic secretion, are just as safe as parenteral<br />

nutrition in adults with severe, acute pancreatitis.<br />

Nasojejunal feeds are also less costly, have fewer<br />

metabolic complications, and may shorten the<br />

length of stay. 61 Antibiotics are not normally indicated,<br />

unless there are signs of sepsis, necrotic<br />

pancreatitis, or multiorgan system failure.<br />

Histamine (H2) receptor antagonists may help to<br />

prevent stress ulceration by reducing duodenal<br />

acidification. Current research targeting the<br />

inflammatory cascade as described in the<br />

Pathophysiology section of this chapter may also<br />

lead to therapies that are beneficial for acute<br />

pancreatitis, regardless of the etiology.<br />

Adequate treatment of pain in acute, severe<br />

pancreatitis in childhood can be challenging. It<br />

may be difficult to relieve a child’s pain<br />

completely. Opiates have been reported to worsen<br />

symptoms by increasing spasms of the sphincter of<br />

Oddi. Meperidine (Demerol ®) is the analgesic of<br />

choice of all the pure opiate agonists for acute<br />

pancreatitis, because it produces the least increase<br />

in enterobiliary pressure. We have also used<br />

hydromorphone hydrochloride (Dilaudid ® ) as a<br />

continuous infusion in many children with severe,<br />

Treatment 315<br />

acute pancreatitis, as well as in chronic and<br />

complicated pancreatitis, with excellent pain<br />

control.<br />

Complications of chronic pancreatitis include<br />

pancreatic atrophy with resultant exocrine and/or<br />

endocrine insufficiency. Severe, prolonged cases<br />

may require insulin, pancreatic enzyme replacement<br />

and an elemental or low-fat diet to optimize<br />

absorption once enteral feedings are reinitiated.<br />

The major cause of mortality in pediatric patients<br />

with acute pancreatitis is septic complications.<br />

These are believed to arise from bacteria that have<br />

translocated across the intestinal epithelium and<br />

disseminated systemically via the mesenteric<br />

lymph nodes and lymphatics. This can result in<br />

pancreatic abscess, infected pseudocyst, or even<br />

necrosis of the gland. Evidence of infection within<br />

a defined area can be obtained with fine-needle<br />

aspiration under ultrasound or CT guidance. Gram<br />

stain and culture of the aspirate are clinically<br />

useful. Often, enteric organisms are recovered,<br />

such as Escherichia coli, Klebsiella species and<br />

other Gram-negative rods. 62 In the patient with<br />

necrotizing pancreatitis and organ failure, it is<br />

reasonable to initiate treatment with an antibiotic<br />

that has broad-spectrum activity against both<br />

aerobic and anaerobic bacteria. 55<br />

These infections necessitate surgical intervention.<br />

However, whether a sterile abscess, pseudocyst or<br />

necrosis requires operative management is still<br />

controversial. 55 Antibiotics and intensive care<br />

usually provide adequate support for the patient<br />

who has sterile necrosis. The presence of persistent<br />

ileus, bowel perforation, portal vein thrombosis<br />

and multisystem organ failure are ‘red flags’<br />

that indicate urgent surgical intervention. Infected<br />

pancreatic necrosis is an absolute surgical indication,<br />

requiring necrosectomy (surgical debridement).<br />

Necrosectomy is thought to stop the<br />

progression of the necrotizing process and resultant<br />

multi-organ failure. Debridement, rather than<br />

total or partial pancreatic resection, is preferred, as<br />

it preserves exocrine and endocrine function. It<br />

may be necessary for the patient to undergo multiple<br />

reoperations, or continuous lavage with<br />

catheters left in the retroperitoneum. Necrosectomy<br />

itself may cause further complications,<br />

such as sepsis, hemorrhage, wound infection and<br />

fistulas of the pancreas, intestine and biliary<br />

system.


316<br />

Acute and chronic pancreatitis<br />

Short- and long-term prognosis<br />

Most cases of acute pancreatitis in childhood are<br />

isolated and uncomplicated, persisting for usually<br />

less than a week, and rarely progress to chronic<br />

pancreatitis. The chances of surviving a severe or<br />

complicated course of pancreatitis are directly<br />

related to the other organ systems involved. Associated<br />

complications can affect virtually all organ<br />

systems (Table 21.4). 2–4,6,15,17–19,21,23,26,34,36,37,45,53,60–63<br />

The primary morbidity and mortality arise from<br />

septic shock, renal failure, respiratory failure and<br />

inflammatory masses of the pancreas.<br />

The APACHE-II and Ranson criteria, which have<br />

been developed to predict the outcome from acute<br />

pancreatitis in adults, are not as reliable in young<br />

children. Pediatric studies have shown a mortality<br />

rate of 5–17.5% from an initially mild presentation<br />

of acute pancreatitis to upwards of 80–100% from<br />

hemorrhagic pancreatitis or severe multisystem<br />

disorders. After a bout of necrotizing pancreatitis,<br />

exocrine and endocrine insufficiency are common.<br />

The degree of dysfunction correlates directly with<br />

the extent of parenchymal damage.<br />

For patients with traumatic pancreatitis, in the<br />

absence of complete duct transection, nonoperative<br />

management is believed to be safe, and<br />

there are usually no long-term complications. 63<br />

Identification of those with hereditary pancreatitis<br />

is critical, as affected family members are at<br />

increased risk for pseudocysts, pancreatic adenocarcinomas,<br />

and exocrine and endocrine failure.<br />

Table 21.4 Complications associated with pancreatitis (from references 2–4,6,15,17–19,21,23,26,34,<br />

36,37,45,53,60–63)<br />

Pancreatic<br />

Abscess<br />

Ascites<br />

Calculi<br />

Carcinoma<br />

Diabetes mellitus<br />

Duct strictures<br />

Exocrine insufficiency<br />

Fibrosis<br />

Fistula<br />

Necrosis<br />

Phlegmon<br />

Pseudocyst<br />

Gastrointestinal/hepatic<br />

Biliary obstruction<br />

Bowel infarction<br />

Gastritis<br />

Gastrointestinal fistula<br />

Hemorrhage<br />

Hepatic vein thrombosis<br />

Hepatorenal syndrome<br />

Ileus<br />

Jaundice<br />

Peptic ulcer<br />

<strong>Portal</strong> vein thrombosis<br />

Splenic vein varices<br />

Systemic/metabolic<br />

Acidosis<br />

Atelectasis<br />

Adult respiratory distress syndrome<br />

Disseminated intravascular coagulation<br />

Electrocardiographic changes<br />

Encephalopathy<br />

Fat emboli<br />

Fat necrosis<br />

Hyperglycemia<br />

Hyperkalemia<br />

Hypertriglyceridemia<br />

Hypoalbuminemia<br />

Hypocalcemia<br />

Hypotension<br />

Mediastinal abscess<br />

Pericardial effusion<br />

Pleural effusion<br />

Pneumonitis<br />

Psychosis<br />

Renal failure<br />

Renal vessel thrombosis<br />

Respiratory failure<br />

Sepsis<br />

Sudden death<br />

Thrombosis


Conclusion<br />

The clinician needs to have a high index of<br />

suspicion for pancreatitis in the child who<br />

presents with the non-specific but common symptoms<br />

of nausea, vomiting and abdominal pain. A<br />

thorough history that emphasizes recent infections,<br />

medications, trauma and any underlying<br />

medical condition may make the diagnosis clearer.<br />

The traditional use of enzyme testing alone (serum<br />

amylase and lipase) for the diagnosis of both acute<br />

and chronic pancreatitis may not be adequate,<br />

since the clinical specificity remains suboptimal.<br />

Unlike pancreatitis in adults, in which the majority<br />

of cases are due to either gallstone disease or<br />

alcoholism, this disease in childhood can be from<br />

a variety of disparate causes. No identifying factor<br />

is present in up to 25% of cases. Common known<br />

etiologies include infection, trauma, medications,<br />

abnormal anatomy and hereditary or systemic<br />

diseases. A family history of pancreatitis, espe-<br />

REFERENCES<br />

1. Karne S, Gorelick FS. Etiopathogenesis of acute pancreatitis.<br />

Surg Clin North Am 1999; 79: 699-710.<br />

2. Lerner A. Acute pancreatitis in children and adolescents.<br />

In Lebenthal E, ed. Gastroenterology and Nutrition<br />

in Infancy, 2nd edn. New York: Raven Press, 1989:<br />

897–906.<br />

3. Robertson MA, Durie PR. Pancreatitis. In Walker WA,<br />

Durie PR, Hamilton JS, Walker-Smith JA, Watkins JB,<br />

eds. Pediatric Gastrointestinal Disease, 2nd edn. St<br />

Louis: Mosby, 1996: 1436–1465.<br />

4. Roy CC, Silverman A, Alagille D. Pancreatitis and<br />

pancreatic tumors. In Roy CC, Silverman A, Alagille D,<br />

eds. Pediatric Clinical Gastroenterology, 4th edn. St.<br />

Louis: Mosby, 1995: 986–1004.<br />

5. Tietz NW. Support of the diagnosis of pancreatitis by<br />

enzyme tests – old problems, new techniques. Clin<br />

Chim Acta 1997; 257: 85–98.<br />

6. Weizman Z. Acute pancreatitis. In Wyllie R, Hyams JS,<br />

eds. Pediatric Gastrointestinal Disease. Philadelphia: WB<br />

Saunders, 1993: 873–879.<br />

7. Durie PR. Pancreatitis and mutations of the cystic<br />

fibrosis gene. N Engl J Med 1998; 339: 687–688.<br />

8. Werlin SL, Casper J, Antonson D et al. Pancreatitis associated<br />

with bone marrow transplantation in children.<br />

Bone Marrow Transplant 1992; 10: 65–69.<br />

9. Tissieres P, Simon L, Debray D et al. Acute pancreatitis<br />

after orthotopic liver transplantation in children: incidence,<br />

contributing factors and outcome. J Pediatr<br />

Gastroenterol Nutr 1998; 26: 315–320.<br />

Conclusion 317<br />

cially occurring at a young age, should prompt the<br />

physician to look for evidence of anatomic abnormalities<br />

or inherited biochemical defects.<br />

Ultrasound remains the initial radiographic study<br />

of choice in acute pancreatitis in childhood, to<br />

screen for infection, cholelithiasis, congenital<br />

abnormalities and biliary ductal dilatation. If the<br />

course of the pancreatitis is complicated or recurrent,<br />

further detailed imaging of the biliary system<br />

should be performed with ERCP, MRCP or percutaneous<br />

or intraoperative cholangiogram.<br />

Treatment of acute, uncomplicated pancreatitis in<br />

the pediatric population is basically supportive,<br />

and most children will recover without sequelae.<br />

Complications, when they occur, can involve<br />

every major organ system and be life-threatening.<br />

Children with severe or complicated pancreatitis<br />

should be managed at a center with expertise in<br />

pediatric biliary surgery and non-operative radiological<br />

and endoscopic interventions.<br />

10. Collins JE, Brenton DP. Pancreatitis and homocystinuria.<br />

J Inherit Metab Dis 1990; 13: 232–233.<br />

11. Kahler SG, Sherwood G, Woolf D et al. Pancreatitis in<br />

patients with organic acidemias. J Pediatr 1994; 124:<br />

239–243.<br />

12. Lévy P, Menzelxhiu A, Paillot B et al. Abdominal radiotherapy<br />

is a cause for chronic pancreatitis.<br />

Gastroenterology 1993; 105: 905–909.<br />

13. Keljo DJ, Sugerman KS. Pancreatitis in patients with<br />

inflammatory bowel disease. J Pediatr Gastroenterol Nutr<br />

1997; 25: 108–112.<br />

14. See Y, Martin K, Rooney M et al. Severe juvenile<br />

dermatomyositis complicated by pancreatitis. Br J<br />

Rheumatol 1997; 36: 912–916.<br />

15. Guelrud M, Morera C, Rodriguez M et al. Sphincter of<br />

Oddi dysfunction in children with recurrent pancreatitis<br />

and anomalous pancreaticobiliary union: an etiologic<br />

concept. Gastrointest Endosc 1999; 50: 194–199.<br />

16. Mattioli G, Buffa P, Pesce F et al. Pancreatitis caused by<br />

duodenal duplication. J Pediatr Surg 1999; 34: 645–648.<br />

17. Dassopoulos T, Ehrenpreis ED. Acute pancreatitis in<br />

human immunodeficiency virus-infected patients: a<br />

review. Am J Med 1999; 107: 78–84.<br />

18. Kamelmaz I, Elitsur Y. Pancreas divisum – the role of<br />

ERCP in children. W V Med J 1999; 95: 14–16.<br />

19. Lerner A, Branski D, Lebenthal E. Pancreatic diseases in<br />

children. Pediatr Clin North Am 1996; 43: 125–156.<br />

20. Carroccio A, Fontana M, Spagnuolo MI et al. Serum<br />

pancreatic enzymes in human immunodeficiency virus-


318<br />

Acute and chronic pancreatitis<br />

infected children. A collaborative study of the Italian<br />

Society of Pediatric Gastroenterology and Hepatology.<br />

Scand J Gastroenterol 1998; 33: 998–1001.<br />

21. Brivet FG, Naneau SH, Lemaigre GF et al. Pancreatic<br />

lesions in HIV-infected patients. Ballières Clin<br />

Endocrinol Metab 1994; 8: 859–877.<br />

22. Kahn E, Anderson VM, Greco A et al. Pancreatic disorders<br />

in pediatric acquired immune deficiency<br />

syndrome. Hum Pathol 1995; 26: 765–770.<br />

23. Aboulafia DM. Acute pancreatitis: a fatal complication<br />

of AIDS therapy. J Clin Gastroenterol 1997; 25: 640–645.<br />

24. Friedman SL. Kaposi’s sarcoma and lymphoma of the<br />

gut in AIDS. Ballières Clin Gastroenterol 1990; 4:<br />

455–475.<br />

25. Welsh MJ, Tsui LC, Boat TF et al. Cystic fibrosis. In<br />

Scriver CR, Beaudet AL, Sly WS, Valle D, eds. The<br />

Metabolic and Molecular Basis of Inherited Disease. New<br />

York: McGraw-Hill, 1995: 3799–3876.<br />

26. Dodge JA. Paediatric and hereditary aspects of chronic<br />

pancreatitis. Digestion 1998; 59 (Suppl 4): 49–59.<br />

27. Uomo G, Manes G, Rabitti PR. Role of hereditary<br />

pancreatitis and CFTR gene mutations in the aetiology<br />

of acute relapsing pancreatitis of unknown origin. How<br />

are they important? J Pancreas (Online) 2001; 2:<br />

368–372.<br />

28. Rosenstein MG, Cutting GR. The diagnosis of cystic<br />

fibrosis: a consensus statement. J Pediatr 1998; 132:<br />

589–595.<br />

29. Cohn JA, Friedman KJ, Noone PG et al. Relation<br />

between mutations of the cystic fibrosis gene and<br />

idiopathic pancreatitis. N Engl J Med 1998; 339:<br />

653–658.<br />

30. Cohn JA, Jowell PS. Are mutations in the cystic fibrosis<br />

gene important in chronic pancreatitis? Surg Clin North<br />

Am 1999; 79: 723–731.<br />

31. Sharer N, Schwarz M, Malone G et al. Mutations of the<br />

cystic fibrosis gene in patients with chronic<br />

pancreatitis. N Engl J Med 1998; 339: 645–652.<br />

32. Elitsur Y, Chertow BC, Jewell RD et al. Identification of<br />

a hereditary pancreatitis mutation in four West Virginia<br />

families. Pediatr Res 1998; 44: 927–930.<br />

33. Whitcomb DC, Gorry MC, Preston RA et al. A gene for<br />

hereditary pancreatitis maps to chromosome 7q35.<br />

Gastroenterology 1996; 110: 1975–1980.<br />

34. Gates LK, Ulrich CD II, Whitcomb DC. Hereditary<br />

pancreatitis. Surg Clin North Am 1999; 79: 711–722.<br />

35. Whitcomb DC. Hereditary pancreatitis: a model for<br />

understanding the genetic basis of acute and chronic<br />

pancreatitis. Pancreatology 2001; 1: 565–570.<br />

36. Perrault J. Hereditary pancreatitis. Gastroenterol Clin<br />

North Am 1994; 23: 743–752.<br />

37. Uretsky G, Goldschmiedt M, James K, Childhood<br />

pancreatitis. Am Fam Physician 1999; 59: 2507–2512.<br />

38. Pfützer RH, Whitcomb DC. SPINK1 mutations are<br />

associated with multiple phenotypes. Pancreatology<br />

2001; 1: 457–460.<br />

39. Pfützer RH, Barmada MM, Brunskill APJ et al.<br />

SPINK1/PSTI polymorphisms act as disease modifiers in<br />

familial and idiopathic chronic pancreatitis.<br />

Gastroenterology 2000; 119: 615–623.<br />

40. Chen JM, Mercier B, Audrezet MP et al. Mutational<br />

analysis of the human pancreatic secretory trypsin<br />

inhibitor (PSTI) gene in hereditary and sporadic chronic<br />

pancreatitis (letter). J Med Genet 2000; 37: 67–69.<br />

41. Rossi L, Pfützer RH, Parvin S et al. SPINK1/PSTI<br />

mutations are associated with tropical pancreatitis in<br />

Bangladesh: a preliminary report. Pancreatology 2001; 1:<br />

242–245.<br />

42. Teich N, Ockenga J, Hoffmeister A et al. Chronic<br />

pancreatitis associated with an activation peptide<br />

mutation that facilitates trypsin activation.<br />

Gastroenterology 2000; 119: 461–465.<br />

43. Sahu S, Saika S, Pai SK et al. L-Asparaginase (Leunase)<br />

induced pancreatitis in childhood acute lymphoblastic<br />

leukemia. Pediatr Hematol Oncol 1998; 15: 533–538.<br />

44. Sammett D, Greben C, Sayeed-Shah U. Acute<br />

pancreatitis caused by penicillin. Dig Dis Sci 1998; 43:<br />

1778–1783.<br />

45. Tobias JD, Capers C, Sims P et al. Necrotizing<br />

pancreatitis after 10 years of therapy with valproic acid.<br />

Clin Pediatr 1995; 34: 446–448.<br />

46. Oberlander TF, Rappaport LA. Recurrent abdominal<br />

pain during childhood. Pediatr Rev 1993; 14: 313-319.<br />

47. Stevenson RJ, Ziegler MM. Abdominal pain unrelated to<br />

trauma. Pediatr Rev 1993; 14: 302–311.<br />

48. Pieper-Bigelow C, Strocchi A, Levitt MD. Where does<br />

serum amylase come from and where does it go?<br />

Gastroenterol Clin North Am 1990; 19: 793–810.<br />

49. Murthy UK, DeGregorio F, Oates RP et al.<br />

Hyperamylasemia in patients with the acquired immunodeficiency<br />

syndrome. Am J Gastroenterol 1992; 87:<br />

332–336.<br />

50. Gwodz GP, Steinberg WM, Werner M et al. Comparative<br />

evaluation of the diagnosis of acute pancreatitis based<br />

on serum and urine enzyme assays. Clin Chim Acta<br />

1990; 187: 243–248.<br />

51. Agarwal N, Pitchumoni CS, Sivaprasad AV. Evaluating<br />

tests for acute pancreatitis. Am J Gastroenterol 1990; 85:<br />

356–360.<br />

52. Frank B, Gottlieb K. Amylase normal, lipase elevated: is<br />

it pancreatitis? A case series and review of the literature.<br />

Am J Gastroenterol 1999; 94: 463–469.<br />

53. Clain JE, Pearson RK. Diagnosis of chronic pancreatitis.<br />

Surg Clin North Am 1999; 79: 829–845.<br />

54. Heiji HA, Obertop H, Schmitz PIM et al. Evaluation of<br />

the secretin–cholecystokinin test for chronic<br />

pancreatitis by discriminant analysis. Scand J<br />

Gastroenterol 1986; 21: 35–42.<br />

55. Banks PA. Practice guidelines in acute pancreatitis. Am J<br />

Gastroenterol 1997; 92: 377–386.<br />

56. Samavedy R, Sherman S, Lehman GA. Endoscopic<br />

therapy in anomalous pancreatobiliary duct junction.<br />

Gastrointest Endoscop 1999; 50: 623–627.<br />

57. Guelrud M, Endoscopic therapy of pancreatic disease in<br />

children. Gastrointest Endoscop Clin North Am 1998; 8:<br />

195–219.<br />

58. Fulcher AS, Turner MA, Capps GW et al. Half-Fourier<br />

RARE MR cholangiopancreatography: experience in 300<br />

subjects. Radiology 1998; 207: 21–32.<br />

59. Hirohashi S, Hirohashi R, Uchida H et al. Pancreatitis:<br />

evaluation with MR cholangiopancreatography in<br />

children. Radiology 1997; 203: 411–415.<br />

60. O’Rourke RW, Harrison MR. Pancreas divisum and<br />

stenosis of the major and minor papillae in an 8 year<br />

old girl: treatment by dual sphincteroplasty. J Pediatr<br />

Surg 1998; 33: 789–791.<br />

61. Abou-Assi S, Craig K, O’Keefe SJ. Hypocaloric jejunal<br />

feeding is better than total parenteral nutrition in acute<br />

pancreatitis: Results of a randomized, comparative<br />

study. Am J Gastroenterol 2002; 97: 2255–2262.<br />

62. Luiten EJ, Hop WC, Lange JF et al. Differential prognosis<br />

of gram-negative versus gram-positive infected and<br />

sterile pancreatic necrosis: results of a randomized trial<br />

in patients with severe, acute pancreatitis treated with<br />

adjuvant selective decontamination. Clin Infect Dis<br />

1997; 25: 811–818.<br />

63. Holland AJ, Davey RB, Sparnon AL et al. Traumatic<br />

pancreatitis: long-term review of initial non-operative<br />

management in children. Paediatr Child Health 1999;<br />

35: 78–81.


22<br />

Introduction<br />

Food allergies<br />

Simon Murch<br />

During the past half century, allergies of all kinds<br />

have become much more common within the<br />

developed world. Dietary allergies are no exception,<br />

and up to 5% of children develop allergy to<br />

cow’s milk and other proteins (reviewed by<br />

Walker-Smith and Murch 1 and Wood 2 ). There has<br />

also been a change in patterns of presentation. As<br />

dietary exposures in infancy and early childhood<br />

have altered, previously unusual reactions to antigens<br />

such as peanut and sesame have become<br />

much more common. 3–5 There are important<br />

geographical differences, with different incidence<br />

of specific allergies varying from country to<br />

country. 5 This may relate to genetic differences in<br />

immune responses amongst different ethnic<br />

groups, or to local dietary customs – exemplified<br />

most strikingly by reports of anaphylaxis to birds’<br />

nest soup in Singaporean infants. 6 However, a<br />

broader context is provided by recent evidence<br />

from the direction of basic science, which highlights<br />

the importance of infectious exposures of<br />

the innate immune system in early life in inducing<br />

tolerance to dietary antigens (oral tolerance). This<br />

chapter attempts to encompass both the clinical<br />

aspects of food allergy and some of the relevant<br />

scientific background.<br />

Early life exposures, possibly even prenatal exposures,<br />

have changed substantially within countries<br />

of the developed world, in the past two generations.<br />

Birth in disadvantaged conditions within the<br />

developing world is protective against most forms<br />

of allergy, and predisposition to allergy may manifest<br />

only once a certain threshold of improved<br />

material conditions is passed. 7,8 Allergies occur<br />

because of breakdown of immunological tolerance.<br />

The immune system must differentiate between all<br />

foreign molecules, reacting only to potentially<br />

harmful pathogens while remaining unresponsive<br />

(tolerant) to the commensal bacterial flora and to<br />

foods. The role of the bacterial flora in establishing<br />

immune tolerance has only recently become<br />

apparent, and will be discussed in more detail<br />

later, together with evidence that manipulation of<br />

the bacterial flora may provide a novel approach to<br />

the management of dietary and other allergies. 8–11<br />

First, however, it is important to clarify the difference<br />

between food intolerance and food allergy<br />

(Table 22.1). As there may however be an overlap<br />

in the symptomatology, an accurate history and<br />

Table 22.1 Food intolerance reactions which may be confused with true allergy<br />

Direct toxic effects<br />

Bacterial contamination; heavy metals or toxins; some food additives<br />

Intolerance due to enzyme deficiency<br />

Inborn metabolic abnormalities (e.g. phenylketonuria, tyrosinemia, galactosemia) inducing adverse responses to<br />

specific dietary components. Enterocyte gene deficiencies (lactase deficiency, sucrase–isomaltase deficiency or<br />

glucose–galactose malabsorption) inducing diarrhea after ingestion of relevant sugars. Enteropathy causing downregulated<br />

expression of lactase and sucrase<br />

Symptoms due to pharmacological properties of foods<br />

For example tyramine contained in cheese or red wine; histamine in strawberries<br />

319


320<br />

Food allergies<br />

supportive investigations are needed for secure<br />

diagnosis of true food allergy.<br />

Food allergies – classification<br />

There are two major classes of food allergic reactions:<br />

IgE-mediated and non-IgE-mediated. IgEmediated<br />

allergies usually present soon after<br />

ingestion, and thus the causative antigen is often<br />

readily identifiable. In addition, there are usually<br />

supportive diagnostic tests such as skin prick tests.<br />

These reactions are those of type I hypersensitivity<br />

(Table 22.2). 12,13 They can be very severe, and may<br />

cause death through anaphylaxis in severe cases.<br />

Non-IgE-mediated allergies usually present later<br />

after ingestion, and the causative antigen may be<br />

more difficult to detect, particularly as tests for<br />

immediate allergies are often negative. These may<br />

include type IV hypersensitivity (Table 22.2), or<br />

may be caused by local eosinophil recruitment.<br />

They can be an important cause of morbidity,<br />

which may go unrecognized. The immunological<br />

basis of such reactions is discussed later.<br />

Food allergic reactions may also be divided clinically<br />

into quick-onset reactions, occurring within<br />

an hour of food ingestion, and slow-onset reac-<br />

Table 22.2 The classical forms of hypersensitivity (after reference 12)<br />

tions, taking hours or days. In general, quick onset<br />

symptoms are IgE-mediated and slow-onset symptoms<br />

are non-IgE-mediated. However, this is by no<br />

means invariable, and many children with clear<br />

quick-onset responses to foods have a low or even<br />

undetectable serum total IgE level and absent foodspecific<br />

IgE level. 14 It is also notable that mice<br />

totally deficient in IgE owing to gene knockout<br />

may still suffer anaphylaxis, as IgG1 bound to<br />

FCγRIII on mast cells may induce antigen-specific<br />

degranulation. 15,16 Such a phenomenon may<br />

explain why investigations such as skin prick tests<br />

may be positive in children despite undetectable<br />

specific IgE for that antigen.<br />

Differentiation between IgE-mediated and non-<br />

IgE-mediated reactions may therefore be difficult.<br />

The Melbourne Milk Allergy Study, conducted by<br />

Hill and colleagues, 17 identified three types of<br />

reaction in sensitized children – immediate reactions<br />

(rapid skin reactions with perioral erythema,<br />

facial angioedema and urticaria, some developing<br />

anaphylaxis), intermediate reactions (gastrointestinal<br />

symptoms such as vomiting or diarrhea occurring<br />

1–24h after ingestion) and delayed reactions<br />

(eczema flares or respiratory symptoms such as<br />

cough and wheeze, occurring between 1 and 5<br />

days after challenge). The volume of milk required<br />

Type I: anaphylactic or immediate hypersensitivity<br />

This occurs within minutes of exposure, as seen in quick-onset food allergy. The allergen binds to, and cross-links IgE<br />

(occasionally IgG1) on the mast cell surface, inducing its degranulation and release of vasoactive agents (histamine,<br />

tryptase, etc.) and cytokines (tumor necrosis factor-α). Responses to some antigens (classically peanut) are usually of<br />

this kind.<br />

Type II: cytotoxic hypersensitivity<br />

This reaction occurs when antibody binds to an epitope on the cell surface, then fixes complement, causing<br />

complement-mediated cell death. This is not a reaction usually described for food allergy, but complement activation<br />

can be detected in celiac disease.<br />

Type III: immune complex hypersensitivity<br />

In this type of reaction, antigen complexes with antibody (IgG or IgM) in the presence of antigen excess, to induce<br />

complement fixation and a consequent local inflammatory response, several hours after exposure to the antigen. The<br />

expression of Fc receptors for immunoglobulin appears to determine tissue damage. 13<br />

Type IV: delayed hypersensitivity or cell-mediated immunity<br />

This reaction is essentially mediated by T lymphocytes, with tissue damage also caused by macrophages responding<br />

to T-cell cytokines. The pattern of T-cell responses (Th1 or Th2) may determine overall immunopathology. The classic<br />

type IV reaction is a Th1 response, as in Crohn’s disease, while both Th1 and Th2 reactions occur in food allergy.


to elicit these symptoms increased between<br />

groups, and classic allergy tests such as skin prick<br />

tests were helpful only for the first group of early<br />

reactors. Knowledge of the time course and likely<br />

immunopathogenesis indicates that the early reactions<br />

are due to IgE responses and mast cell<br />

degranulation, the intermediate reactions follow<br />

eosinophil recruitment and the delayed responses<br />

are likely to relate to T-cell responses. 18 These<br />

concepts will be discussed later, in the sections on<br />

immunopathogenesis. The Melbourne group have<br />

also played an important role in the recognition of<br />

the increasing incidence of multiple food allergies,<br />

and of the role of food allergy in inducing a spectrum<br />

of symptoms not previously associated with<br />

allergy. 5 The role of food allergy in inducing<br />

visceral dysmotility syndromes such as infant<br />

colic, gastroesophageal reflux and recurrent<br />

abdominal pain will be discussed later.<br />

In a recent study of 121 children with allergy to<br />

two or more foods, including both those with IgEmediated<br />

and those with non-IgE-mediated allergies,<br />

14 children with early-onset symptoms had a<br />

significant overall increase in serum IgE compared<br />

to those with late-onset symptoms. However, 30%<br />

of those with early-onset symptoms had no elevation<br />

of IgE concentration above the normal range,<br />

while 10% of those with only delayed symptoms<br />

did have an elevated IgE concentration. Over 90%<br />

of those with early-onset symptoms additionally<br />

demonstrated late-onset symptoms. Although the<br />

groups differed in IgE responses, they shared,<br />

regardless of speed of reaction, a pattern of<br />

immune deviation, with increased serum IgG1 and<br />

circulating B cells, but reduced IgG2 and IgG4,<br />

CD8 cells and natural killer cells. IgA concentrations<br />

were at the low end of the normal range or<br />

below. This raises the question of whether the<br />

propensity to sensitize to food antigens is associated<br />

with minor immunodeficiency, a concept first<br />

suggested by Soothill, 19 who postulated that<br />

demonstrated maturational delay in IgA responses<br />

predisposed to food allergic sensitization. Further<br />

data in support of a link with a developmental<br />

delay in IgA maturation was provided by a population<br />

survey from Iceland, in which an IgA level<br />

at the lower end of the normal range was more<br />

predictive of allergic sensitization than was<br />

elevated IgE concentration, 20 and from other<br />

studies of food allergic infants, in which increased<br />

B cells and decreased CD8 cells and decreased IgA,<br />

Patterns of food allergic responses 321<br />

IgG2 and IgG4 were associated with milk allergies<br />

and food-sensitive colitis. 21,22 These data suggest<br />

that there may be a consistent pattern of minor<br />

immunodeficiency associated with the process of<br />

sensitization in early life, and that the manifestation<br />

of that sensitization (quick- or slow-onset)<br />

may depend on whether the child has inherited a<br />

tendency to high IgE production. Thus, a high IgE<br />

level may not cause food allergy per se, but may<br />

determine how that allergy is expressed. Most of<br />

the early literature on food allergy, however,<br />

understandably focuses on IgE and quick-onset<br />

responses.<br />

Later in the chapter, the role of infectious exposures<br />

in maturation of mucosal immune responses<br />

and immune tolerance will be explored. Whether<br />

food allergy can be fully explained by the Clean<br />

Child Hypothesis 7 remains uncertain, but there is<br />

substantial circumstantial evidence that these<br />

infectious exposures are probably an important<br />

contributory factor in the pathogenesis of allergies.<br />

Recent recognition of the role of infectious exposures<br />

in the generation of cells producing the regulatory<br />

cytokine transforming growth factor (TGF)β,<br />

and of the central requirement for TGF-β in both<br />

IgA responses and oral tolerance, may provide<br />

some explanation for the consistent links with<br />

slow IgA maturation. 8,19,23<br />

Patterns of food allergic responses<br />

Quick-onset symptoms<br />

These often follow the ingestion of a single food,<br />

such as egg, peanut or sesame. Within minutes the<br />

sufferer may notice tingling of the tongue, and<br />

there may be the rapid development of skin rash,<br />

urticaria or wheezing. One localized variant, the<br />

oral allergy syndrome, is often seen in older<br />

pollen-sensitized individuals and is characterized<br />

by lip tingling and mouth swelling after ingestion<br />

of certain fruits and vegetables. 24 More serious<br />

reactions, however, can occur at any age, and antigens<br />

such as peanut, tree nuts, fish and shellfish<br />

can cause exquisite sensitization from early childhood.<br />

Swelling of the mucous membranes of the<br />

mouth and upper airway (angioneurotic edema)<br />

can develop extremely quickly, and the airway<br />

may become compromised. In cases where


322<br />

Food allergies<br />

appropriate therapy is not available, life-saving<br />

tracheostomy has even been performed in the<br />

presence of gross laryngeal edema. Anaphylactic<br />

shock may also occur, with dramatic systemic<br />

hypotension accompanying the airway obstruction.<br />

A grading system for anaphylaxis has been<br />

suggested by Sampson, 25 with a severity score<br />

based on the worst symptom present (Table 22.3).<br />

Specific therapy for mild cases of immediate<br />

hypersensitivity would include antihistamines<br />

such as chlorpheniramine, together with inhaled<br />

bronchodilators as appropriate. It is notable that<br />

some patients can have a biphasic response, with<br />

a relatively modest initial reaction followed<br />

several hours later by a more profound and potentially<br />

life-threatening response, and therefore care<br />

should be taken in the assessment to ensure that<br />

adequate instructions and therapy are adminis-<br />

tered before allowing the patient home. There have<br />

indeed been several cases of anaphylactic death in<br />

patients discharged home after initial resuscitation,<br />

up to 6h after food ingestion, and it is therefore<br />

prudent to maintain observation for several<br />

hours if there are any clear risk factors, such as<br />

active asthma or a history of a severe reaction. 25,26<br />

The presence of wheezing on examination should<br />

suggest caution; a bronchodilator should certainly<br />

be prescribed, and in many cases a few days’<br />

course of oral prednisolone.<br />

More severe reactions should be assessed very<br />

rapidly, and any evidence of airway obstruction or<br />

systemic hypotension warrants the immediate use<br />

of intramuscular epinephrine (adrenaline) (Table<br />

22.3). Use of a preloaded syringe pen, such as a<br />

pediatric or adult EpiPen ® , prior to transfer to<br />

hospital, may be life-saving, and avoids potential<br />

Table 22.3 A grading for anaphylactic reactions, proposed by Sampson. 25 The severity score should<br />

be based on the most severe symptom in any domain. Symptoms in bold are absolute indications for<br />

the use of epinephrine (adrenaline)<br />

Gastrointestinal<br />

Grade Skin tract Respiratory Cardiovascular Neurological<br />

1 Localized itching, oral itching or<br />

flushing, urticaria, tingling, mild<br />

angioedema lip swelling<br />

2 Generalized itching, any of the above, nasal congestion change in<br />

flushing, urticaria, nausea and/or and/or sneezing activity level<br />

angioedema single vomiting<br />

episode<br />

3 Any of the above any of the above rhinorrhea, tachycardia change in<br />

plus repetitive marked congestion, (increase activity<br />

vomiting sensation of throat >15 beats/min) level plus<br />

itching or tightness anxiety<br />

4 Any of the above any of the above any of the above, any of the above, ‘light<br />

plus diarrhea hoarseness, ‘barky’ dysrhythmia headedness,’<br />

cough, difficulty and/or mild feeling of<br />

swallowing, hypotension ‘impending<br />

dyspnea, wheezing, doom’<br />

cyanosis<br />

5 Any of the above any of the above, any of the above, severe loss of<br />

loss of bowel respiratory arrest bradycardia consciousness<br />

control and/or hypotension<br />

or cardiac arrest


dosage errors at such a time of extreme stress. Any<br />

child thought to be at risk of an anaphylactic reaction<br />

to food should be prescribed such injection<br />

devices (at least two or three should be prescribed,<br />

so that all those caring for the child are equipped),<br />

and parents and carers should be trained in their<br />

use. Any severe immediate reaction to foods necessitates<br />

urgent transfer to an appropriate medical<br />

setting, such as an Accident and Emergency<br />

Department. Other treatments that may be needed<br />

include oxygen, intravenous hydrocortisone,<br />

chlorpheniramine and inhaled β-adrenergic bronchodilator<br />

therapy. 25 Supportive treatment for<br />

hypotension or cardiac dysrrhythmia may be<br />

required, and in the most severe cases the patient<br />

may need to be transferred to an intensive therapy<br />

unit.<br />

The true incidence of anaphylactic death due to<br />

food allergy is unknown. A recent UK report<br />

suggested a low incidence of 0.006 deaths per<br />

100000 children per year, 27 but this is thought to<br />

be a significant underestimate, as many cases were<br />

unlikely to be identified from the study of death<br />

certificates and clinical reporting alone. 25<br />

In the follow-up of a patient who has had an<br />

immediate hypersensitive response to food antigens,<br />

a decision needs to be taken about the level<br />

of prophylaxis required, and whether or not to<br />

prescribe an epinephrine pen. Clearly the severity<br />

of the first response will inform this decision, and<br />

it is probably better to err on the side of caution if<br />

foods such as peanuts are implicated, because of<br />

Table 22.4 Clinical manifestations in food allergic responses<br />

Patterns of food allergic responses 323<br />

their propensity for triggering particularly severe<br />

episodes. If there is doubt about the food involved,<br />

both skin prick tests and specific IgE radioallergosorbent<br />

test (RAST) may be very helpful.<br />

However, these should be postponed for several<br />

weeks after an episode of anaphylaxis, as they may<br />

be artifactually negative in the immediate aftermath<br />

of a severe reaction.<br />

Late-onset symptoms<br />

Slow-onset symptoms may be more insidious and<br />

their true allergic nature may not be recognized<br />

(Table 22.4). These may include failure to thrive or<br />

chronic diarrhea due to enteropathy or colitis,<br />

eczema, rhinitis, or rectal bleeding. 1,24 As these are<br />

likely to be mediated by T cells in a delayed hypersensitive<br />

reaction, they may not be so clearly<br />

linked to food ingestion. Children may, however,<br />

manifest both delayed and immediate-onset symptoms.<br />

Analysis of IgG subclasses and peripheral<br />

lymphocyte subsets may identify the child at<br />

increased risk of delayed food allergic reactions. 14<br />

Certain specific presentations can be recognized,<br />

as discussed below.<br />

Food protein-induced enterocolitis<br />

This disorder is most common in young infants<br />

below the age of 3 months, and usually presents<br />

with blood streaking of stools in the absence of<br />

marked weight loss or systemic upset. Anemia is<br />

1. Quick onset wheezing, urticaria, angioedema, rashes, vomiting, gastroesophageal reflux,<br />

anaphylaxis<br />

2. Late onset diarrhea, abdominal pain, allergic rhinitis, atopic eczema, food-sensitive enteropathy<br />

or colitis, rectal bleeding, constipation, protein-losing enteropathy<br />

3. Less clear responses irritable bowel syndrome, chronic fatigue, attention-deficit with hyperactivity,<br />

autistic symptoms<br />

4. Secondary general effects eosinophilia, iron deficiency anemia, hypoproteinemia<br />

For group 3 conditions, there are anecdotal reports of clinical improvement with dietary exclusions. However, there<br />

are as yet few properly validated studies, and it is likely that only a proportion of such patients will benefit. While<br />

many maintain an open-minded approach, it is important to ensure that any such diet is nutritionally adequate (joint<br />

management with dietitians is ideal), and that clinical responses to exclusion and challenge are sufficiently striking<br />

to justify continuing exclusion diets.


324<br />

Food allergies<br />

unusual, but an elevated platelet count<br />

(>450x10 9 /l ) is characteristic. Cow’s milk or soy<br />

proteins are the most common causative antigens,<br />

and it is common for such symptoms to occur in<br />

exclusively breast-fed infants, triggered by milk<br />

protein in the mother’s diet. 28 It is important to<br />

recognize that negative skin prick tests do not<br />

exclude this diagnosis, and indeed most cases are<br />

negative. 24 If colonoscopy is performed, the colitic<br />

changes are usually milder than with classic<br />

inflammatory bowel disease, and the macroscopic<br />

findings are dominated by loss of vascular pattern,<br />

prominent lymphoid follicles with a rim of perifol-<br />

(a) (b)<br />

(c) (d)<br />

licular erythema (red halo sign) and an easily traumatized<br />

mucosa. Histological changes include<br />

mononuclear cell infiltration, mucosal<br />

eosinophilia with evidence of degranulation and<br />

the presence of lymphoid follicles in the majority<br />

of colonic biopsies. If the ileum is visualized,<br />

lymphoid hyperplasia is usually seen. Recent data<br />

suggest a concordance between the endoscopic<br />

finding of ileocolonic lymphoid hyperplasia and<br />

food allergies (Figure 22.1), not restricted to those<br />

infants with allergic enterocolitis alone, 29 which<br />

may be associated with an increase in mucosal γδ<br />

T-cell infiltration. 30 See also Chapter 30.<br />

Figure 22.1 Lymphoid hyperplasia is an important endoscopic feature in food allergies. 29 This ileocolonoscopy was<br />

performed in a 2-year-old girl with multiple food allergies, and demonstrated ileal lymphonodular hyperplasia (a), with<br />

prominent reactive germinal centers on histology (b), together with multiple colonic lymphoid follicles (c – a typical follicle<br />

is arrowed). Histology of each biopsy in her colonic series demonstrated a lymphoid follicle (d).


Some infants manifest a form of food proteininduced<br />

enterocolitis that is more severe than the<br />

classic variant, and is less often induced by cow’s<br />

milk. 24 The histological response is more severe,<br />

with evidence of frank colitis including crypt<br />

abscesses, and the response to food challenge may<br />

include shock.<br />

Food protein enteropathy<br />

The most common and best described mucosal<br />

manifestation of food allergy is food-sensitive<br />

enteropathy, in which there is an immunologically<br />

mediated abnormality of the small intestinal<br />

mucosa, which may include excess lymphocyte<br />

infiltration, epithelial abnormality or architectural<br />

disturbance. This may often impair absorption and<br />

less commonly causes a frank malabsorption<br />

syndrome. This continues while the food remains<br />

in the diet and remits on an exclusion diet. This is<br />

best described for cow’s milk, and cow’s milksensitive<br />

enteropathy (CMSE) has been recognized<br />

for over two decades. 32 Enteropathy can also occur<br />

in response to other antigens, notably soy. The<br />

lesion is less severe than celiac disease, but is<br />

characterized by similar findings of crypt elongation<br />

and villus shortening, giving a reduction of<br />

overall crypt/villus ratio. There is usually an<br />

increase of mononuclear cell density within the<br />

mucosal lamina propria, often including prominent<br />

eosinophils, and the intraepithelial lymphocyte<br />

density is often increased (Figures 22.2 and<br />

22.3). The lesion is not associated directly with<br />

systemic IgE responses, and skin prick tests are<br />

often negative. Analysis of mucosal lymphocytes<br />

confirms excess T-cell activation, with either a Th1<br />

dominated or mixed Th1/Th2 response. 31,33,34 The<br />

consequences of this mucosal T-cell activation<br />

include reduction in brush-border disaccharidase<br />

expression, leading to impaired carbohydrate<br />

absorption, and a secondary reduction in pancreatic<br />

enzyme release, 35 both contributing to malabsorption.<br />

There is evidence that some children do<br />

not grow out of CMSE in early childhood, and an<br />

abnormal mucosa may be seen in later childhood.<br />

36<br />

In cases of diagnostic uncertainty, it may be necessary<br />

to confirm the return of mucosal abnormality<br />

by food challenge. This was a more common practice<br />

in the past, when the very existence of foodsensitive<br />

enteropathies other than celiac disease<br />

Patterns of food allergic responses 325<br />

was uncertain. Improvements in infant formulas<br />

have led to a change in the mucosal appearances of<br />

CMSE, so that celiac-like villous atrophy is now<br />

very rare in the developed world. Morphometry of<br />

recent cases of CMSE confirms a less severe lesion<br />

than archival biopsies from the 1980s. 14 This<br />

causes some histopathological difficulty, as there<br />

is no international consensus in the reporting of<br />

subtle lesions such as villous blunting or mild<br />

mucosal eosinophilia.<br />

Eczema/atopic dermatitis<br />

There seems little doubt that food-allergic<br />

responses may contribute to the clinical presentation<br />

of eczema in infancy, but considerable uncertainty<br />

about whether adults have anything to gain<br />

from exclusion diets. 37 There is evidence that children<br />

with eczema have elevated food-specific IgE<br />

in serum, and that food challenges of eczematous<br />

children induce increases in circulating eosinophil<br />

and mast cell products. 38 There is evidence of<br />

enhanced intestinal permeability in infants with<br />

eczema, potentially contributing to sensitization<br />

through bypass of enterocyte antigen-handling<br />

mechanisms. 39 The response within the skin is<br />

dominated by Th2 cytokines, and characterized by<br />

influx of both T lymphocytes, which express skinhoming<br />

markers such as the cutaneous lymphocyte<br />

antigen (CLA), and eosinophils. 37,40 The<br />

mechanisms by which skin-homing markers<br />

become expressed on gut sensitized cells remains<br />

uncertain, as expression of the gut-homing marker<br />

β7 integrin is likely to have been required for<br />

initial homing to the intestine. 1 There remains the<br />

intriguing possibility that bacterial products,<br />

particularly superantigens from staphylococci and<br />

streptococci, may specifically up-regulate CLA<br />

expression in an interleukin (IL)-12-dependent<br />

fashion, and thus promote skin homing of lymphocytes<br />

sensitized to dietary antigens. 41 Staphylococcus<br />

aureus may be an important specific<br />

complicating factor, as 24 and 28kD proteins<br />

within the toxins may directly induce an IgE-mediated<br />

response and thus contribute to allergic<br />

immunopathology. 42,43 Deficiency in innate<br />

immune responses, in particular production of βdefensins,<br />

may contribute to the propensity for<br />

superinfection with toxin-producing staphylococci<br />

in children with allergic eczema. 44 Therefore, an<br />

important consideration in the infant or young<br />

child with eczema is whether both the diet and the


326<br />

(a)<br />

Food allergies<br />

(b) (c)<br />

Figure 22.2 The relatively subtle appearances of food-sensitive enteropathy. Duodenal biopsy of the same patient as<br />

shown in Figure 22.1, showing minimal evidence of duodenal blunting, but with a patchy increase in mononuclear cell<br />

infiltration within the lamina propria (a). High-power views (b, c) show infiltration of eosinophils within the lamina propria<br />

and epithelium, together with a moderate increase in mononuclear cells.<br />

bacteriological status of the child have been<br />

optimized.<br />

A recent large study of skin prick reactivity to<br />

dietary antigen in infants with eczema identified<br />

positive skin prick tests for cow’s milk, egg or<br />

peanut in 22% of 6-month-old infants with<br />

eczema, compared to only 5% without eczema. 45<br />

At 1 year the incidences were 36% and 11%,<br />

respectively, giving a calculated attributable risk of<br />

65%. This supports data by Kjellman and<br />

Hattevig 46 that the development of infantile, but<br />

not later-onset eczema, is associated with IgE<br />

antibodies to food antigens. Whether the IgE<br />

antibodies are directly causative, or alternatively a<br />

marker of dietary sensitization in infants


(a) (b)<br />

predisposed to high IgE levels, remains uncertain.<br />

However, there is clear evidence that manipulation<br />

of the diet of the eczematous child may be beneficial.<br />

A double-blind study by Atherton et al in<br />

1978 47 found that two-thirds of 2–8-year-old children<br />

with atopic eczema were improved by exclusion<br />

diets. Subsequent studies have broadly<br />

confirmed this finding, although in most cases the<br />

response was not so striking, except in younger<br />

infants. 37 The dominant inducing antigens of food<br />

allergy-induced childhood eczema are eggs, milk,<br />

soy, and peanut in young infants, with the addition<br />

of wheat, tree nuts and fish in older infants. Most<br />

children tend to outgrow allergies to eggs, milk,<br />

wheat and soy, but tend to have persistent reactions<br />

to peanuts, tree nuts, shellfish and fish. The<br />

use of skin patch testing has suggested that multiple<br />

sensitizations are common in affected infants<br />

below 2 years, and thus exclusion of single antigens<br />

may not give satisfactory clinical responses. 48<br />

In addition to antigen exclusion, the use of probiotic<br />

treatment has been reported to improve the<br />

symptoms of chronic eczema in food-allergic<br />

infants. 49 Whether this is because of a direct effect<br />

on epithelial integrity, or competitive reduction of<br />

toxin-producing staphylococci is as yet unknown.<br />

Probiotics are considered in further depth later in<br />

this chapter, as well as in Chapter 32. For the<br />

Patterns of food allergic responses 327<br />

Figure 22.3 There may be an increase of intraepithelial lymphocytes in food-allergic children. This child with cow’s milksensitive<br />

enteropathy showed normal villous architecture, but increase in CD8 intraepithelial lymphocytes (a). The CD4<br />

cell population lies essentially within the lamina propria (b). Photomicrographs courtesy of Dr Franco Torrente.<br />

affected infant who is exclusively breast fed,<br />

maternal dietary exclusions can improve the<br />

eczema, but potentially at the cost of maternal<br />

nutritional inadequacy if multiple exclusions are<br />

required. 50 In addition, some infants may not<br />

respond completely to extensively hydrolyzed<br />

formulas, due to response to the residual milk antigens,<br />

and these may show improvement only with<br />

an amino acid formula. 51,52<br />

Allergic dysmotility<br />

The concept that food allergies might trigger<br />

intestinal dysmotility in pediatric patients is relatively<br />

new, but there is mounting evidence that<br />

dietary antigens (most commonly cow’s milk, soy<br />

or wheat) can induce gastroesophageal reflux or<br />

constipation. 8,53–55 Gastrointestinal investigation<br />

of children with delayed food allergic responses<br />

frequently uncovers a history of infant colic,<br />

gastroesophageal reflux and chronic abdominal<br />

pain. 5,14,24,51,52 Conversely, amongst infants<br />

presenting with colic, there is a significant incidence<br />

of underlying cow’s milk allergy. 56,57 There<br />

may additionally be evidence of small-intestinal<br />

enteropathy or allergic colitis. However, it is not<br />

uncommon for food allergy to be overlooked<br />

because ‘the investigations are negative’.


328<br />

Food allergies<br />

In infancy, food allergy-induced gastroesophageal<br />

reflux is often accompanied by a complex presentation,<br />

including low-grade enteropathy with<br />

secondary carbohydrate malabsorption, colic, irritability,<br />

eczema and prolonged viral infections. 5,8,14<br />

The infants may fail to thrive, and show foodaversive<br />

behavior, or weight gain may be acceptable.<br />

5,14 In many cases, reactions to hydrolysate<br />

formulas occur, and a therapeutic trial of an amino<br />

acid formula should be considered. 5,52,58,59<br />

Dermatographia may be a useful clinical pointer in<br />

an apparently colicky infant, as is a family history<br />

of allergies. A distinct pattern on 24-h pH testing<br />

has been reported to be associated with antigeninduced<br />

gastroesophageal reflux, in which there is<br />

gradual reduction in esophageal pH following a<br />

feed. 53 The frequent finding on esophageal biopsy<br />

is mucosal eosinophilia, which may be related to<br />

expression of the chemokine eotaxin in milkinduced<br />

gastroesophageal reflux. 55<br />

Even in the older child, milk responses may be a<br />

cause of continuing symptoms, including chronic<br />

abdominal pain, with endoscopic findings of<br />

lymphonodular hyperplasia. 60 There is some evidence<br />

that delayed allergic responses to milk in<br />

infancy are not always outgrown, and thus the<br />

diagnosis should be considered in the older child<br />

with an atopic history who presents with chronic<br />

abdominal pain. 36 There is also evidence that<br />

constipation can be triggered by delayed allergic<br />

responses to cow’s milk protein in older children.<br />

54 Once again, mucosal eosinophil infiltration<br />

was characteristic. Studies using anorectal manometry<br />

suggested that this pattern of constipation<br />

was caused by antigen-induced spasm of the anal<br />

spincter muscles, rather than a more widespread<br />

colonic dysmotility. 61 The use of radio-opaque<br />

markers may confirm the pattern of normal colonic<br />

transit with impaction within the rectum.<br />

Clinically the children often demonstrate avoidance<br />

posturing, leaning forward during defecation<br />

in an attempt to stop the large rectal hard mass<br />

from bearing down, while passing the softer stool<br />

from above. Clinical examination may be difficult,<br />

and even a sizeable acquired megarectum may be<br />

difficult to palpate. In cases of clinical doubt, a<br />

plain abdominal X-ray may be helpful (Figure<br />

22.4). One characteristic pattern is of fecal<br />

impaction within the rectum, together with abundant<br />

gas within the small intestine, related to<br />

malabsorption of carbohydrates plus constipationassociated<br />

small-bowel bacterial overgrowth.<br />

Figure 22.4 Plain abdominal X-ray of a child with<br />

non-IgE-mediated food allergies, demonstrating acquired<br />

megarectum with fecal impaction. Conventional constipation<br />

therapy was ineffective, and a cow’s milk- and wheatfree<br />

diet were required.<br />

Eosinophilic esophagitis and eosinophilic<br />

enteropathy<br />

Eosinophilic esophagitis and eosinophilic enterocolitis<br />

are emerging clinical entities, which<br />

overlap with non-IgE-mediated food allergy, but<br />

which may also occur without recognizable food<br />

responses. 62 While an apparently increasing cause<br />

of infant gastroesophageal reflux disease,<br />

eosinophilic esophagitis is also increasingly seen<br />

in older children, who often present with abdominal<br />

pain, dysphagia or vomiting, sometimes with<br />

associated loose stools. 63,64 In some cases,<br />

eosinophil recruitment is confined to the serosa,<br />

and may be missed on mucosal biopsies. In many<br />

cases, symptoms of esophagitis are severe and<br />

prolonged.


At endoscopy, the esophagus shows a distinctive<br />

finding of linear furrows or transverse rings, and<br />

the mucosa appears granular. 64 The mucosa is<br />

traumatized unusually easily. 65 Histological diagnosis<br />

is based on the detection of five or more<br />

eosinophils per high-powered field. Recent reports<br />

of endoscopic ultrasound examination show<br />

increased thickness of the esophageal wall. 66 The<br />

results of 24-h pH testing are variable, and novel<br />

techniques such as luminal impedance testing<br />

suggest that much non-acid reflux may occur. 67<br />

Skin prick testing is often negative, but a combination<br />

of skin prick and skin patch testing may be<br />

helpful in identifying cases of food-induced<br />

eosinophilic esophagitis. 68<br />

Later in the chapter, the potential mechanisms of<br />

eosinophil recruitment within the mucosa will be<br />

addressed. The eosinophil chemokine eotaxin is<br />

clearly important in this process, and indeed this<br />

chemokine was up-regulated within the basal<br />

esophageal epithelium in infants with milkinduced<br />

reflux, in comparison to those with<br />

simple mechanical gastroesophageal reflux. 55<br />

Management of eosinophilic esophagitis<br />

Particularly in younger infants, it is important to<br />

perform adequate dietary exclusions. In a significant<br />

minority of affected infants, symptoms are not<br />

adequately relieved using hydrolysate formulas,<br />

and an amino acid-based formula may be<br />

required. 58 Effective acid reduction therapy with<br />

ranitidine or omeprazole, together with a prokinetic<br />

such as domperidone, are frequently used<br />

in conjunction. 67 An important clinical finding is<br />

the rapid symptomatic worsening during viral<br />

illnesses, which is unresponsive to dietary exclusion.<br />

This often requires a temporary increase<br />

in antacid therapy.<br />

For the older child, dietary exclusions are often<br />

less helpful, but a therapeutic trial of a ‘few foods’<br />

diet may determine whether this is an avenue<br />

worth exploring. Topical corticosteroids such as<br />

inhaled fluticasone have proved effective in cases<br />

where diet has proved ineffective. 69 More recent<br />

evidence suggests that leukotriene antagonists<br />

such as montelukast may be helpful in corticosteroid-resistant<br />

eosinophilic esophagitis. 70<br />

Respiratory symptoms<br />

Testing for food allergies 329<br />

Respiratory allergies, particularly asthma, are<br />

increasingly common in young children. It is also<br />

well recognized that the immediate hypersensitive<br />

response to a food allergen includes respiratory<br />

symptoms such as wheeze (Table 22.4). What has<br />

been less clear is whether delayed responses to<br />

food antigens may include worsening of respiratory<br />

status in asthma, or can be implicated in<br />

upper airway disorders such as rhinitis. 71 Foodinduced<br />

wheezing appears most common in young<br />

infants. 5,17 The estimated prevalence of foodinduced<br />

wheezing is under 6% of the total population<br />

of children with asthma, but it occurs much<br />

more frequently in children with cow’s milk<br />

allergy (29%) or atopic dermatitis (17–27% 71 ).<br />

Whether antigen-induced gastroesophageal reflux<br />

can contribute to respiratory symptoms is an<br />

important consideration, and there is potentially<br />

important recent evidence suggesting that effective<br />

treatment of gastroesophageal reflux may significantly<br />

diminish the requirements for bronchodilator<br />

therapy in an unselected population of childhood<br />

asthmatics. 72 However, it is not clear how<br />

many children had straightforward mechanical<br />

gastroesophageal reflux and how many may have<br />

had an underlying allergic dysmotility.<br />

Testing for food allergies<br />

Food challenge testing<br />

The essential criterion for diagnosis of food allergy<br />

is a response to an elimination diet, and other<br />

diagnostic tests are secondary to this. If there is<br />

allergy to a single food, exclusion should induce<br />

relief of all symptoms, and restore normal growth.<br />

For secure diagnosis, a positive response to challenge<br />

with the food antigen is strongly supportive<br />

of the diagnosis of food allergy. This is not always<br />

practicable in routine practice, if diagnostic tests<br />

were positive at diagnosis, and many parents may<br />

refuse challenge if their child has improved<br />

dramatically. Insurance companies, however, may<br />

require evidence of positive food challenge for<br />

reimbursement.<br />

In addition to initial challenge for first diagnosis,<br />

subsequent challenge may be performed when it is<br />

reasonably likely that tolerance has been regained.


330<br />

Food allergies<br />

This is usually after 2 years, but often later,<br />

especially in multiply allergic children. The<br />

European Society for Paediatric Gastroenterology,<br />

Hepatology and Nutrition 73 made recommendations<br />

for challenge criteria for children<br />

with predominant gastrointestinal symptoms,<br />

suggesting that blinded challenge is not always<br />

necessary in younger children, but that smallbowel<br />

biopsy should be performed in cases with<br />

failure to thrive or diarrhea. Skin prick testing (see<br />

later) may provide more specific guidance for children<br />

with immediate reactions.<br />

In children with multiple food allergies, the<br />

response to elimination of single antigens is<br />

incomplete, and lengthy assessment with a very<br />

restricted diet is often required. Such situations<br />

may become complicated, particularly since the<br />

advent of Internet sites that implicate food allergies<br />

in an unfeasibly broad spectrum of disorders,<br />

and it may be difficult to persuade some parents to<br />

broaden their child’s diet. For the child who manifests<br />

the symptoms of only non-IgE-mediated<br />

allergy, it may become extraordinarily difficult to<br />

obtain a clear picture of the true state of current<br />

true allergy. This situation is complicated by the<br />

minor immunodeficiency often associated with<br />

multiple food allergies, and the exacerbating<br />

effects of intercurrent viral illnesses. 14,19,20 It is<br />

important not to perform food challenges while a<br />

child is systemically unwell. For cases of multiple<br />

sensitization, with delayed responses, it becomes<br />

logistically difficult to perform lengthy blinded<br />

challenges. Close teamwork with an experienced<br />

dietitian is essential in management of any such<br />

complicated cases.<br />

The open food challenge is the most common form<br />

of diagnostic challenge, and is performed either in<br />

the out-patient clinic or in a day ward, depending<br />

on the severity and type of reaction expected. 74<br />

Many centers will use a graded challenge in those<br />

children who manifest immediate reactions,<br />

initially placing a single drop of milk or other<br />

antigen on the skin, then on the lips, and then<br />

giving increasing amounts by mouth, leaving a<br />

period of several minutes between each stage. For<br />

the child with a history of severe reaction, it is<br />

mandatory that there be adequate medical supervision<br />

with the presence of appropriate resuscitation<br />

and drugs. For those with a history of anaphy-<br />

laxis, an intravenous line is usually inserted prior<br />

to the procedure.<br />

For the child who makes delayed reactions only,<br />

the test may have to be completed at home for<br />

logistic reasons. False-negative results have been<br />

reported if late reactions are not taken into<br />

account. In a study of 370 challenges in 242 children,<br />

five exhibited mild immediate reactions that<br />

manifested only on the second day at home. 75<br />

These reactions were subsequently confirmed by<br />

skin prick test and double-blind placebocontrolled<br />

food challenge, and the authors<br />

suggested supervised feeding of antigen on the<br />

second day to identify late reactors. Hill et al 17<br />

identified very late reactions (up to 3 days) which<br />

required up to 120ml to elicit. Recent data suggest<br />

that even later reactions may occur during cow’s<br />

milk challenges, with the onset of gastrointestinal,<br />

respiratory or cutaneous responses beyond 1 week<br />

after reintroduction. 76 Therefore, caution and<br />

judgement are needed to interpret food challenge<br />

tests, to prevent the missing of true late phenomena,<br />

but also to exclude over-reporting of symptoms<br />

by parents convinced that food allergy is<br />

responsible for all their child’s various symptoms.<br />

The ‘gold-standard’ challenge test, the doubleblind<br />

placebo-controlled food challenge is a<br />

cumbersome and time-consuming intervention,<br />

but is clearly necessary for assessment of cases<br />

with genuine uncertainty about sensitization.<br />

24,73,77 In cases of true uncertainty, the test can<br />

firmly establish tolerance or persistent reactivity<br />

in a way that no other test can. Its value is much<br />

lower when there are large numbers of foods to be<br />

tested, and the child makes delayed responses<br />

only. Normal day-to-day variation may then be<br />

overinterpreted, the situation can become stressed<br />

if performed on an in-patient basis, or intercurrent<br />

viral illness induces symptoms interpreted as due<br />

to the food allergy. This test requires extremely<br />

good teamwork between pediatrician, dietitian,<br />

pediatric nursing staff and the child’s family. No<br />

universally accepted protocol exists, 78 but in<br />

general the suspected allergen or placebo is given<br />

in disguised form, either by capsule or hidden in a<br />

liquid over a period of around 2h. The child<br />

should not be receiving antihistamines and should<br />

not have received the antigen for at least 2 weeks,<br />

and should be well. A total dose of around 10g is<br />

normally taken as sufficient, although this would


not pick up some of the delayed reactors identified<br />

by Hill et al, 17 Carroccio et al 76 or Caffarelli and<br />

Petroccione. 75 Nevertheless, for immediate allergies,<br />

the test is highly specific and of very high<br />

prognostic value, with a suggested false-positive<br />

rate below 1% and false-negative rate below 5%. 78<br />

Skin prick testing<br />

The skin prick test remains a cornerstone of the<br />

diagnosis of immediate allergic reactions, but is<br />

usually negative in those who manifest delayed<br />

reactions only. 14,74 This is performed by placing a<br />

drop of the potential antigen on the skin (usually a<br />

commercially obtained solution, although some<br />

units use freshly prepared foods) and introducing<br />

it into the skin through a puncture device such as<br />

a lancet. A positive control (histamine) and negative<br />

control (saline) are used at the same time, and<br />

the results read at 15–20min. The test is based on<br />

the induced degranulation of cutaneous mast cells<br />

by antigen binding and cross-linking of their<br />

surface IgE molecules. The size of wheal elicited<br />

determines whether the test is regarded as positive.<br />

Usually a wheal of 3mm or more is taken as a<br />

positive reading, although in young infants the test<br />

can be falsely negative, as the histamine-induced<br />

positive control is much smaller in infancy than<br />

later childhood, owing to the smaller numbers of<br />

cutaneous mast cells. 79 Recent data suggest that<br />

the size of wheal obtained may be an important<br />

predictor of reaction, and large wheals (>8mm)<br />

may be so highly predictive of sensitization that<br />

unnecessary food challenges can be avoided. 80,81<br />

The study by Sporik et al, 80 of 467 children aged<br />

from 1 month to 16 years, correlated the size of the<br />

wheal elicited to cow’s milk, egg or peanut with<br />

clinical outcome. Using the traditional cut-off<br />

value of 3mm was not found to be helpful in<br />

predicting a response to challenge, and many<br />

false-positive and false-negative results were<br />

obtained. However, using a cut-off value of 8mm<br />

for cow’s milk and peanut and 7mm for egg, they<br />

found no children with a negative response on<br />

challenge. Their calculations suggest that they<br />

could avoid clinical testing of 33% of milk-allergic,<br />

56% of egg-allergic and 68% of peanut-allergic<br />

patients. These are potentially very important<br />

results, but clearly do require local replication, as<br />

testing solutions and technique will vary from<br />

center to center, and a negative test will not rule<br />

Testing for food allergies 331<br />

out a delayed non-IgE-mediated response to an<br />

antigen.<br />

Roberts and Lack 79 have extrapolated from the<br />

data from Sporik et al to suggest that a more<br />

precise risk value can be obtained using the Fagan<br />

likelihood nomogram, 82 in which the risk of true<br />

reaction based on history is used together with the<br />

result of the skin prick to determine the overall<br />

likelihood of true sensitization. They provide an<br />

example where a child with low pre-test likelihood<br />

of peanut sensitization (headache and vomiting 4h<br />

after a peanut butter sandwich) would have a 0.2%<br />

likelihood of true peanut allergy with a 3mm<br />

wheal, rising to 5% with a 6mm wheal and >99%<br />

only at 10mm, in contrast to a child with a high<br />

pre-test likelihood (urticaria and wheeze on two<br />

occasions within minutes of accidental peanut<br />

exposure), who would have a 15% likelihood of<br />

true peanut allergy with a negative skin prick test,<br />

rising to 70% with a 3mm reaction and 96% with<br />

a 6mm reaction.<br />

Skin patch testing<br />

The use of the skin patch (atopy patch) test, in<br />

which the relevant antigen is maintained against<br />

the skin under a sealed patch for 48h, has been<br />

suggested to identify cases of non-IgE-mediated<br />

delayed allergy. A positive test is signaled by the<br />

finding of erythema and induration at 72h.<br />

Combination of patch testing with either skin<br />

prick testing or specific IgE testing has identified<br />

significant numbers of food-sensitized children<br />

who may have been negative on classic testing. 83,84<br />

Of children with delayed reactions to antigen, 89%<br />

were identified by patch test in one study. 83 This<br />

test has not become widely used and remains<br />

under evaluation. Recent reports do, however<br />

suggest that patch testing may be clinically useful,<br />

particularly in the presence of eczema. 85,86 There<br />

are some logistic difficulties, in that the test should<br />

be read only after 3 days, requiring a second clinic<br />

visit, and some children will not tolerate an occlusive<br />

dressing for so long. It is also potentially open<br />

to artifact due to lack of supervision, and our unit<br />

has had at least one case of factitious illness, in<br />

which irritants were introduced beneath the occlusive<br />

dressing by the mother. However, it appears<br />

that the test induces a specific T-cell response to<br />

dietary antigen, 87 and thus a properly performed<br />

patch test may be helpful in confirming delayed


332<br />

Food allergies<br />

food allergic responses in cases of diagnostic difficulty.<br />

Specific IgE testing<br />

Testing of specific IgE production to individual<br />

foods can be helpful. Formerly known as RAST<br />

testing, the technique is now usually based on<br />

enzyme-linked immunosorbent assay (ELISA) or<br />

the Pharmacia Cap system. It provides results that<br />

are complementary to skin prick testing, but at<br />

greater cost and with more delay. However, there is<br />

evidence that the information provided can be<br />

clinically predictive. Using the Pharmacia Cap<br />

system, positive predictive values of 95% were<br />

found for egg at 6kUA/l, milk at 32kUA/l, peanut<br />

at 15kUA/l and fish at 20kUA/l. 88 These tests are<br />

frequently reported on a semiquantitative scale of<br />

0 (no specific IgE) to 6 (high titers), with increasing<br />

likelihood of clinical relevance above level 3.<br />

However, there is wide variance between centers,<br />

and it is important to perform local audit to determine<br />

the clinical relevance of reported specific<br />

IgE. There is an increased likelihood of longlasting<br />

allergy in children with higher titers of<br />

specific IgE for milk, casein or β-lactoglobulin. 89<br />

Potentially important prognostic information may<br />

be allowed in future by study of the specific<br />

sequence of epitopes bound by food-specific IgE.<br />

Cooke and Sampson 90 identified two forms of IgE<br />

binding to ovomucoid (the dominant egg allergen).<br />

Binding to linear peptide sequences within the<br />

ovomucoid molecule was associated with longlasting<br />

allergy, whereas binding to discontinuous<br />

sequences, which are brought into apposition only<br />

by protein folding, was seen in those who outgrew<br />

their allergies. This may explain the phenomenon<br />

whereby some children will be tolerant of cooked<br />

egg, where the tertiary sequence has been<br />

disrupted in the cooking process, but still react to<br />

raw egg white. For cow’s milk, too, IgE reaction to<br />

linear sequences in casein, rather than the whey<br />

proteins α-lactalbumin and β-lactoglobulin,<br />

appears to be predictive of long-lasting milk<br />

allergy. IgE binding sites in specific sequences of<br />

αS1-casein, αS2-casein and κ-casein were identified<br />

in those whose allergy was not outgrown. 91,92 It is<br />

possible that this may provide the basis for future<br />

testing of milk-allergic infants, to identify those for<br />

whom immunotherapy may be necessary to<br />

prevent lifelong sensitization.<br />

In vitro testing<br />

In vitro tests for lymphocyte response remains so<br />

far a research tool, and simple proliferation assays<br />

have given quite unreliable results. However,<br />

analysis of cytokine production patterns, using<br />

single-cell techniques such as ELISPOT or flow<br />

cytometry, represents a promising approach for the<br />

future. Lymphocytes from food-allergic children<br />

produce a pattern of cytokines associated with Th2<br />

responses (IL-4, IL-5, IL-13) on antigen challenge,<br />

whereas those from healthy controls may make a<br />

more Th1-dominated response, with higher interferon-γ<br />

production. 93 However, studies have<br />

reported quite contradictory results when based<br />

on derived T-cell lines or clones, which may not<br />

represent true in vivo responses.<br />

A promising novel technique has recently been<br />

reported, in which peripheral blood mononuclear<br />

cells were tagged with a fluorescent molecule<br />

called carboxyfluorescein succinimidyl ester,<br />

before stimulation with peanut antigen. 94 This<br />

allowed simple identification of peanut-responding<br />

T cells by flow cytometry, and confirmed that<br />

T-cell responses to peanut in children with active<br />

allergies are indeed skewed towards Th2 (producing<br />

IL-4, IL-5 and IL-13, but not interferon-γ or<br />

TNF-α), in contrast to findings of a Th1-dominated<br />

response with high interferon-γ and tumor necrosis<br />

factor (TNF)-α in healthy controls or those who<br />

had outgrown their allergies. 94 The authors<br />

reported similar skewing in milk- and egg-allergic<br />

children compared to controls, suggesting that this<br />

is likely to be a common mechanism in IgEmediated<br />

food allergies. These findings imply that<br />

it is the host immune response, rather than a<br />

fundamental property of individual food antigens,<br />

that determines whether a Th2-skewed response<br />

occurs and allergy is induced.<br />

It should be noted that mucosal responses appear<br />

quite different from systemic responses, and there<br />

are as yet no validated tests that might help in the<br />

diagnosis of non-IgE-mediated food allergies<br />

(Figure 22.5). TNF-α production is stimulated<br />

within the mucosa by milk challenge of allergic<br />

patients, 38,95 which would not be expected from<br />

study of circulating T cells, 94 but may reflect its<br />

release from mucosal mast cells. Studies of<br />

mucosal T-cell responses agree in showing that<br />

Th1 responses are maintained in intraepithelial,


(a) (b)<br />

lamina propria and Peyer’s patch lymphocytes of<br />

food-allergic children. 23,31,96<br />

Specific food allergies<br />

Allergic responses to many food proteins have been<br />

described. The most common in childhood are to<br />

cows’ milk, soy, eggs and fish, with peanut allergy<br />

rapidly becoming more common. 1,3,4,24 However,<br />

intolerance to fruits, vegetable, meats, chocolate,<br />

Specific food allergies 333<br />

Figure 22.5 Mucosal IgE responses may be distinct from those in other sites. (a) IgE immunohistochemistry of the<br />

duodenal mucosa of a child with milk-induced dysmotility, but negative skin prick testing, a serum IgE below 5 kIU/l and<br />

no circulating specific IgE. Two types of IgE-positive cell are seen: round cells with a large nucleus and strong surface<br />

staining (IgE plasma cells) and irregularly shaped cells with intense granular staining (mast cells). (b) Confirmation that<br />

these granular cells are mast cells, as double staining for mast cell tryptase (red) and IgE (green) gives a resultant yellow<br />

color. Photomicrographs courtesy of Dr Franco Torrente.<br />

nuts, shellfish and cereals has been described. In<br />

adult life there is a different spectrum, with allergy<br />

to nuts, fruits and fish relatively more common<br />

than in childhood. However, as the dietary exposures<br />

of UK children broaden, the range of reported<br />

allergens also increases: thus sesame, kiwi fruit,<br />

mango, avocado and other allergies have increased<br />

in frequency. Such sensitization may depend as<br />

much on a combination of genetics, infectious<br />

challenges and timing and dose of exposure as on<br />

innate antigenicity of individual foodstuffs.


334<br />

Food allergies<br />

There are no consistent associations between any<br />

particular food and specific syndromes, although<br />

some foods are more likely than others to induce<br />

enteropathy, such as cow’s milk and soy, and<br />

others usually induce immediate hypersensitivity,<br />

such as peanut. The incidence of gastrointestinal<br />

food allergy is greatest in early life and appears to<br />

decrease with age. However, analogy with celiac<br />

disease, in which late-onset enteropathy is more<br />

likely to be clinically subtle, suggests that the<br />

increase in colonic salvage that occurs with age<br />

may mask true food-sensitive enteropathy.<br />

Although enteropathy can cause significant failure<br />

to thrive, complex multiple allergies may also<br />

occur in the presence of normal growth. 5<br />

Cow’s milk<br />

Cow’s milk allergy may commonly present either<br />

as an immediate response, including anaphylaxis,<br />

or with delayed responses within the gut or skin.<br />

CMSE was frequently diagnosed in the past<br />

because of failure to thrive following an episode of<br />

gastroenteritis, 97 and the consequent lactose intolerance<br />

often inappropriately managed with<br />

reduced-lactose formulas without cow’s milk<br />

antigen exclusion (still common practice in some<br />

developing world countries). Modern adapted<br />

formulas are now much less sensitizing, and the<br />

mucosal lesions less severe. While positive skin<br />

prick tests and milk-specific IgE may be seen in<br />

some children with enteropathy, they are much<br />

more frequent in children who have immediate<br />

reactions to milk. Cow’s milk colitis is most<br />

common in breast-fed babies, whose mothers are<br />

consuming milk in their diet, and usually presents<br />

with low-grade rectal bleeding. For reasons<br />

unknown, it is unusual for one infant to develop<br />

both cow’s milk enteropathy and colitis. Milkinduced<br />

dysmotility is discussed above.<br />

Egg<br />

By contrast to cow’s milk, egg allergy usually<br />

presents either as an acute hypersensitive<br />

response or with delayed respiratory or cutaneous<br />

reactions, with worsening of asthma or eczema.<br />

While vomiting may occur soon after ingestion, or<br />

diarrhea ensue after a few hours, there is little<br />

evidence that egg can induce small-intestinal<br />

enteropathy. 1 Skin prick tests, patch tests and<br />

specific IgE to egg are more often positive than for<br />

other antigens, 86 and may be predictive of time<br />

taken to outgrow egg allergy. About half of under-<br />

2-year-old children who develop egg allergy will<br />

tolerate it during 3 years of follow-up, with the size<br />

of skin prick reaction and specific IgE potentially<br />

predictive of those children who are unlikely to<br />

outgrow allergy. 79,98<br />

Soy<br />

Soy-based formulas have for some years been used<br />

in infants with cow’s milk allergy, although more<br />

commonly by general pediatricians than pediatric<br />

gastroenterologists. However, recommendation of<br />

soy milk use by the American Academy of<br />

Pediatrics Committee on Nutrition 99 may increase<br />

the use of soy in comparison to hydrolysates. Soybased<br />

formulas are as antigenic as cow’s milk<br />

formulas, 100 and the reported reactions span the<br />

range from anaphylaxis to enteropathy, eczema<br />

and respiratory symptoms. 1 A 30kD protein in soy<br />

may induce cross-reactivity to cow’s milk<br />

caseins, 101 potentially explaining the high incidence<br />

of soy intolerance in cow’s milk-allergic<br />

children. An important consideration is that antigenicity<br />

of soy-derived products is strongly influenced<br />

by methods of preparation, and thus children<br />

may react to some soy-based products and<br />

not others. 102 Unlike in antigens such as peanut<br />

and egg, skin prick testing is frequently not a good<br />

predictor of subsequent clinical reactions to soy. 79<br />

Patch testing may provide more clinically relevant<br />

information, particularly in a child with eczema. 85<br />

Wheat<br />

Acute allergic reactions to wheat are very uncommon,<br />

although wheat anaphylaxis has been<br />

described, 103 and an ω-gliadin has been characterized<br />

as the likely sensitizing antigen in children<br />

with immediate reactions. 104 By contrast to the<br />

relative rarity of immediate reactions, delayed<br />

hypersensitive reactions are common and clinically<br />

important. Celiac disease is particularly<br />

important, and affects at least 1% of European and<br />

North American populations. 105 Celiac disease is<br />

discussed in Chapter 27 in depth. However, it is<br />

important to recognize that a low IgA predisposes


to both celiac disease and food allergies. It is thus<br />

advisable that serological testing for celiac disease<br />

should be performed in all food-allergic patients at<br />

some stage during their diagnostic evaluation.<br />

There is increasing recognition that wheat products<br />

may play a disproportionate role in inducing<br />

intestinal dysmotility, such as gastroesophageal<br />

reflux and constipation. There are also reports that<br />

wheat and cow’s milk may induce behavioral<br />

effects, possibly because of their natural content of<br />

morphine-like exorphins such as β-casomorphine<br />

and gliadomorphine. 106,107 While this may also<br />

contribute to constipation, such a response would<br />

technically be an intolerance rather than a true<br />

allergy. However, further work is clearly needed in<br />

what is a poorly understood but potentially important<br />

area.<br />

Peanut<br />

Peanut allergy is concerning, because of its rising<br />

incidence and its propensity to induce severe<br />

anaphylaxis. 3,4,24,25 It is particularly important in<br />

childhood allergy as a cause of fatal anaphylactic<br />

reaction. 108 Even trace amounts of peanut can<br />

cause death in those severely sensitized. The surge<br />

in peanut hypersensitivity may relate to novel<br />

patterns of exposure, and there is recent evidence<br />

to suggest that percutaneous sensitization may be<br />

more important than simple ingestion. 108 Analysis<br />

of the Avon Longitudinal Study of Parents and<br />

Children identified that prenatal sensitization was<br />

extremely uncommon, but that peanut allergy was<br />

associated with intake of soy milk, rash over joints<br />

or a crusted oozing rash, and in particular use of<br />

skin creams for eczema that contained peanut<br />

oil. 109 In this study, cases were initially identified<br />

on the basis of a questionnaire, and then subsequently<br />

the diagnosis was confirmed by doubleblind<br />

placebo-controlled food challenge. From a<br />

studied cohort of just under 14000 children, 49<br />

were identified with a history of peanut allergy<br />

and the diagnosis confirmed in 23 of 36 children<br />

tested.<br />

T-cell responses appear important in determining<br />

sensitization to peanut, and indeed there has been<br />

one report of peanut anaphylaxis transferred to the<br />

recipient of a liver transplant. 110 Intriguingly,<br />

chimerism was noted in the skin, but not the<br />

Specific food allergies 335<br />

blood, of the recipient, which implies that<br />

lymphocyte homing had occurred. The T-cell<br />

response of peanut-allergic, but not tolerant, children<br />

is skewed towards Th2 cytokines, 94 suggesting<br />

that there is no innate property of peanuts that<br />

is responsible for allergic sensitization. However,<br />

the severity of peanut reactions does argue for<br />

some additional factor beyond simple Th2<br />

skewing, and it is thus notable that the peanutderived<br />

lectin peanut agglutinin is used by pathologists<br />

to identify germinal centers in lymphoid<br />

follicles. In a rodent model of food allergic sensitization,<br />

peanut agglutinin was notable amongst<br />

dietary antigens for its ability to induce high IgE<br />

responses. 111 Whether the presence of peanut<br />

agglutinin affects the level of response to the<br />

recognized sensitizing epitopes for humans is<br />

currently unknown. Similar evidence from this<br />

model that a wheat lectin, wheat germ agglutinin,<br />

has a modulating effect on ovalbumin responses,<br />

112 suggests that the presence of lectins<br />

within foods may contribute to immune sensitization<br />

events.<br />

In the UK, it is recommended that children from<br />

an atopic background should not be given peanut<br />

products until after the age of 6. 3,4 However, as<br />

most children may be sensitized early in life by<br />

non-classical routes, 109 this may not be as effective<br />

as was initially hoped. There is evidence that<br />

many children may outgrow peanut allergy, and<br />

that skin prick testing may identify those with a<br />

good chance of a successful peanut challenge (see<br />

above). For those with established allergy and a<br />

history of anaphylaxis, monoclonal anti-IgE<br />

therapy is a promising option. 113 This is discussed<br />

in more depth in the section on basic mechanisms<br />

later in the chapter.<br />

Multiple food allergy<br />

Many infants develop gastrointestinal and other<br />

symptoms related to a wide variety of foods. The<br />

condition of multiple food allergy 5 provides great<br />

challenges for the family, the child and the allergist.<br />

Reactions may be immediate or delayed, and<br />

do not differ significantly from those described<br />

above for individual foods. Affected children often<br />

have a family history of atopy, may have increased<br />

eosinophils in the peripheral blood, with elevated<br />

serum IgE and positive specific IgE, and skin tests


336<br />

Food allergies<br />

to specific foods. 8,14,24,51 Many cases appear to<br />

sensitize through maternally ingested antigens<br />

during exclusive breast feeding, with the small<br />

amounts of dietary proteins either sufficient to<br />

sensitize or insufficient to tolerize. A specific<br />

defect in oral tolerance for low-dose antigen has<br />

been postulated as a cause of this phenomenon, 114<br />

supported by recent data of a defective generation<br />

of Th3 cells within the mucosa of affected children.<br />

34 Affected children may show residual intolerance<br />

of hydrolysates. 5,14,52<br />

Recommendations for food allergen<br />

avoidance<br />

There remains controversy about the stringency of<br />

allergen avoidance required in food allergies,<br />

recently reviewed by Zeiger. 115 Two position statements<br />

have been published, by the American<br />

Academy of Pediatrics (AAP) 99 and a joint statement<br />

by the European Society for Pediatric<br />

Allergology and Clinical Immunology (ESPACI)<br />

and the European Society for Pediatric<br />

Gastroenterology, Hepatology, and Nutrition<br />

(ESPGHAN). 116 These statements deal with two<br />

areas of importance: the primary prevention of<br />

development of food allergies, and the treatment of<br />

the affected child.<br />

With respect to primary prevention, there are areas<br />

of clear concordance, and both statements support<br />

the limitation of primary prevention to high-risk<br />

infants only, and the use of hypoallergenic formulas<br />

(ideally extensively hydrolyzed) but not soy<br />

milk for bottle-fed high-risk infants. High risk is<br />

defined on family history grounds rather than any<br />

perinatal testing, although the AAP defines a positive<br />

family history as two first-degree relatives<br />

with atopic disease and the ESPACI/ESPGHAN<br />

definition requires only one. Neither recommend<br />

maternal exclusion diets during pregnancy and<br />

both recommend exclusive breast feeding (AAP 6<br />

months, ESPACI/ESPGHAN 4–6 months). While<br />

the European bodies do not recommend a maternal<br />

exclusion diet during lactation, the American body<br />

recommends exclusion of peanuts and nuts, and<br />

consideration of further exclusions. The European<br />

regulations are also less restrictive about the introduction<br />

of solid foods, suggesting introduction at 5<br />

months, rather than the much later introduction of<br />

cow’s milk and eggs suggested in the AAP report.<br />

For treatment of established food allergies, both<br />

statements recommend complete exclusion of the<br />

causative antigen, and show broad consensus in<br />

the management of a formula-fed cow’s milksensitized<br />

infant, with recommendation of an<br />

extensively hydrolyzed but not partially<br />

hydrolyzed formula. However, the AAP guidelines<br />

also suggest that soy is an alternative in this<br />

circumstance, which is not supported by the<br />

European guidelines. Both recommend an amino<br />

acid formula for the infant who is intolerant of<br />

hydrolysates. Neither support the use of unmodified<br />

goat’s or sheep’s milk. For the infant who<br />

becomes sensitized while breast fed, both statements<br />

concord in support of maternal exclusion of<br />

the relevant antigen, while the AAP further recommends<br />

weaning to an extensively hydrolyzed<br />

formula or soy milk. For the infant with concomitant<br />

malabsorption due to enteropathy, both<br />

recommend extensively hydrolyzed or amino acid<br />

formulas.<br />

An important recent report from the German<br />

Infant Nutrition Intervention (GINI) study group 117<br />

assessed 2252 at-risk infants, who were randomly<br />

assigned at birth to receive one of four blinded<br />

formulas, either cow’s milk-based, partially<br />

hydrolyzed whey, extensively hydrolyzed whey, or<br />

extensively hydrolyzed casein. The primary endpoint<br />

at 1 year of age was the presence of one or<br />

more of atopic dermatitis, gastrointestinal food<br />

allergy or urticaria. The drop-out rate was high, as<br />

865 remained exclusively breast fed for 4 months,<br />

304 left the study and 138 did not comply. Study<br />

of the 945 remaining treated infants showed a<br />

significant protective effect of extensively<br />

hydrolyzed casein compared to unmodified cow’s<br />

milk (9% vs. 16% had allergies) and atopic<br />

dermatitis was significantly reduced with extensively<br />

hydrolyzed casein or partially hydrolyzed<br />

whey formulas. The protective effect of<br />

hydrolysates was attenuated in those with a strong<br />

family history of atopy.<br />

The basic mechanisms of immune<br />

response to dietary antigen<br />

The intestine is an organ that shows the traces of<br />

evolutionary longevity, and indeed Cambrian<br />

period fossils from over 600 million years ago


show a recognizable gastrointestinal tract. 118 There<br />

is much current interest on the links between<br />

innate and adaptive immune responses, in particular<br />

pattern receptor molecules such as toll-like<br />

receptors and nod proteins that induce an immune<br />

response within innate cells, such as dendritic<br />

cells, that polarize subsequent T-cell<br />

responses. 119,120 Evidence that oral tolerance<br />

cannot be established normally in germ-free mice<br />

suggests that the normal flora plays an important<br />

role in the generation of tolorogenic lymphocytes<br />

and the prevention of food allergies. 121,122 The<br />

potential role for probiotics in prevention of food<br />

allergies in susceptible infants is thus likely to be<br />

based on the role of luminal bacteria in inducing a<br />

tolerant lymphocyte response. 8,9 Transgenic mice<br />

whose only T cells responded to ovalbumin were<br />

in fact entirely tolerant of ovalbumin feeds, unless<br />

innate immune responses to the flora were blocked<br />

using cyclo-oxygenase-2 antagonists, when foodsensitive<br />

enteropathy was induced. 123 Further<br />

study of dendritic cell populations within the<br />

intestine is likely to shed light on basic mechanisms<br />

of tolerance and sensitization. Genetic variation<br />

in receptors for bacterial products, such as<br />

toll-like receptors and nod proteins, is known to<br />

occur, and is likely to be related to allergic sensitizations,<br />

124 particularly as toll receptors are also<br />

expressed on mast cells. 125<br />

In addition to innate immune cells, the intestine<br />

also contains large numbers of primitive T and B<br />

cells, such as peritoneal B1 lymphocytes, γδ T<br />

cells, natural killer T cells and atypical CD8 cells<br />

with two α and no β chains. 9 Little is known about<br />

the role of these primitive lymphocyte types in<br />

food allergies, although it is intriguing that γδ-deficient<br />

mice have low mucosal IgA levels, 126 while<br />

their numbers are increased within the mucosa in<br />

food allergies. 30 There is also evidence that these<br />

types of T cell can react to lipid and glycolipid<br />

antigens, presented by non-classical MHC molecules<br />

such as CD1d, which are expressed by the<br />

enterocyte. 127–129 It is thus notable that IgE<br />

responses to β-lactoglobulin in a rodent model of<br />

sensitization were substantially enhanced when<br />

the animals were sensitized to whole milk than to<br />

β-lactoglobulin alone. 130<br />

The role of extrathymically derived T cells, which<br />

mature within the gut epithelium, is also unknown<br />

The basic mechanisms of immune response to dietary antigen 337<br />

but likely to be significant, particularly in infants<br />

born preterm. Given the numbers and situation of<br />

these cells, it is likely that further studies will<br />

unmask a relevant role in the induction of tolerance<br />

to dietary antigen. It is probably not coincidence<br />

that infants with a variety of immunodeficiencies<br />

have a high incidence of dietary<br />

sensitizations, chronic enteropathy and failure to<br />

thrive. 131 It is likely that T-cell responses play a<br />

large role in determining tolerance or sensitization<br />

to dietary antigen. There are differences in racial<br />

susceptibility to sensitization to individual antigens,<br />

which do not relate to simple early life exposure.<br />

5<br />

Antigen presentation by the epithelium<br />

One cell type that is likely to play a more significant<br />

role in food allergies than was previously<br />

recognized is the small-intestinal enterocyte,<br />

which separates the immune system from both<br />

dietary antigens and bacteria. Their role in antigen<br />

presentation and production of an array of<br />

cytokines has recently been recognized. 32,128,129,132<br />

An important role in induction of tolerance has<br />

thus been suggested. Recent data suggest that<br />

intestinal epithelium may directly induce a regulatory<br />

phenotype in CD8 cells. 133 Increased paracellular<br />

permeability, allowing transfer of antigen to<br />

the immune system without epithelial processing<br />

and presentation, may underlie the phenomenon<br />

of sensitization to food antigens during conditions<br />

such as rotavirus gastroenteritis. 32,97 Formal confirmation<br />

of the sensitizing role of excess paracellular<br />

permeability was provided in a transgenic<br />

mouse model where an intercellular adhesion<br />

molecule called cadherin was mutated, inducing<br />

severe enteropathy. 134<br />

Skewing of B cells towards IgE<br />

While non-IgE-mediated responses to dietary<br />

antigen may cause chronic symptomatology, IgEmediated<br />

mechanisms account for the majority of<br />

immediate hypersensitive reactions to foods.<br />

Transient IgE responses to foods are seen in<br />

normal children, so this is unlikely to be clinically<br />

relevant. 135 By contrast, high-level IgE responses<br />

are usually pathological and may be important in<br />

severe food allergies and anaphylaxis.


338<br />

Food allergies<br />

Production of IgE is favored by dominance of Th2<br />

responses, particularly due to IL-4 and IL-13 secretion.<br />

136 The receptors for IL-4 and IL-13 share a<br />

common α chain (IL-4Rα), mutations in which<br />

increased signaling is associated with increased<br />

atopy. 137,138 Intracellular signaling downstream of<br />

this receptor is mediated through Stat-6 (signal<br />

transducer and activator of transcription-6), and<br />

blockade of either IL-4Rα or Stat-6 appears promising<br />

as a therapeutic target for IgE-mediated allergic<br />

reactions. 139,140<br />

Lineage commitment of B cells towards IgE is<br />

favored by the presence of IL-4 and IL-13, and<br />

inhibited by Th1-associated cytokines. 136 This<br />

may potentially occur within the Peyer’s patches<br />

in circumstances of Th2-skewed local responses,<br />

and lead to generation of mucosal IgE-producing<br />

plasma cells without necessarily affecting the<br />

commitment of circulating B-cell populations.<br />

Mucosally produced IgE may also be transported<br />

into the lumen of the gut or airway by a mechanism<br />

distinct from secretory component-mediated<br />

IgA transport. 141,142 It is thus possible that a<br />

compartmentalized response may occur within the<br />

intestine, in which mucosal IgE responses may be<br />

elicited by dietary antigen, even if cutaneous IgE<br />

responses do not occur, leading to negative skin<br />

prick tests, and in the absence of circulating<br />

specific IgE.<br />

Mast cells and eosinophils in food allergies<br />

There are undoubted links between intestinal food<br />

allergic responses and the infiltration of both<br />

eosinophils and mast cells, which produce a<br />

variety of vasoactive and neuroactive mediators.<br />

Both cell types have been particularly implicated<br />

in dysmotility responses, and it is likely that these<br />

products may directly affect the function of enteric<br />

nerves. 143 During the food allergic response, both<br />

mast cell tryptase and cationic protein (ECP) are<br />

released into the lumen and may be detected in<br />

stools. 144–146 However, although mast cells and<br />

eosinophils produce a similar spectrum of mediators,<br />

their responses show a different time-course.<br />

Mast cells induce rapid responses through immediate<br />

degranulation of mediators stored in intracellular<br />

granules, whereas eosinophil responses are<br />

often delayed for several hours, as they are<br />

recruited from the peripheral circulation into<br />

tissues.<br />

Two molecules are very clearly implicated in<br />

mucosal eosinophilia – the chemokine eotaxin and<br />

the cytokine IL-5. Important studies by Rothenberg<br />

et al, using targeted gene deletion in mice, have<br />

clarified the relative contributions of both mediators.<br />

Mice were sensitized to ovalbumin, and then<br />

challenged with oral administration of ovalbumincoated<br />

beads. 147 Wild-type mice mounted an allergen-specific<br />

Th2 response, showing mucosal<br />

eosinophilia with increased circulating IgE and<br />

IgG1, while eotaxin-deficient mice had preserved<br />

systemic IgE and IgG1 responses but did not<br />

recruit eosinophils to the mucosa. By contrast,<br />

IL5-deficient mice had reduced circulating<br />

eosinophils. 147 In additional studies of gastric<br />

eosinophil recruitment, it was possible to confirm<br />

that antigen-coated beads induced delayed gastric<br />

emptying, and again eotaxin-deficient mice were<br />

protected. 148 Further studies from this group have<br />

identified the esophagus as an apparent target for<br />

eosinophil recruitment, either as a consequence of<br />

inhalation of aeroallergens such as aspergillus 149<br />

or in circumstances of excess systemic IL-5 expression.<br />

150 This targeting may presumably have some<br />

as yet unclear evolutionary basis, but these findings<br />

have potentially important clinical implications.<br />

First, there may be more than one factor<br />

inducing esophageal eosinophilia, and there may<br />

be only a partial response to antigen exclusion if<br />

the child is also responding to inhaled aspergillus.<br />

Second, an intercurrent systemic viral illness in an<br />

allergic child with a Th2-deviated immune response<br />

may promote esophageal eosinophilia in an<br />

antigen non-specific manner. The increased<br />

frequency of viral infections in food-allergic children,<br />

probably due to low immunoglobulins, CD8<br />

and natural killer cells, 14 may make this a clinically<br />

difficult scenario, as appropriate food exclusions<br />

may give an apparently poor clinical<br />

response.<br />

In adults, IL-5 mRNA is increased within the<br />

small-bowel mucosa of food-allergic patients, but<br />

not in atopic or non-allergic controls. 151 T cells<br />

from food antigen sensitized children produce IL-<br />

5 on food challenge, whereas those from tolerant<br />

children do not. 93,94 Thus, there is evidence for a<br />

final common pathway in the mucosal allergic<br />

response to dietary antigen, which is dependent on


up-regulation of IL-5 production and expression of<br />

the chemokine eotaxin.<br />

T-cell responses in oral tolerance<br />

The phenomenon of oral tolerance lies at the heart<br />

of food allergy, which by its nature implies a breakdown<br />

or failure of establishment of oral tolerance<br />

mechanisms. There has been much recent<br />

progress in the understanding of oral tolerance<br />

mechanisms. The dose of ingested antigen appears<br />

to be particularly important in determining how<br />

tolerance is established. 152 The bulk of dietary<br />

antigen is absorbed by enterocytes for nutritional<br />

purposes, and this antigen is presented by the<br />

epithelium in such a way that lymphocyte reactivity<br />

is suppressed and the lymphocytes become<br />

anergic. 128,129 The absent expression of co-stimulatory<br />

molecules by enterocytes and production of<br />

suppressor cytokines may both play a role,<br />

although little is known about these processes in<br />

human infancy and childhood. Food antigens have<br />

also been shown to induce apoptosis of antigenspecific<br />

lymphocytes in the Peyer’s patches of<br />

mice, but this has not yet been shown in<br />

humans. 153 More recent data suggest that a more<br />

complex state is induced in tolerogenic lymphocytes<br />

by food administration, in which pro-apoptotic<br />

and anti-apoptotic factors are simultaneously<br />

up-regulated while T-cell receptor signaling molecules<br />

are down-regulated. 154<br />

In contrast, tolerance to low doses of antigen<br />

requires uptake by the antigen-sampling M cells<br />

that overlie Peyer’s patches. This form of tolerance<br />

requires an active generation of suppressor<br />

lymphocytes within the Peyer’s patches, and in<br />

particular Th3 cells that produce the anti-inflammatory<br />

cytokine TGF-β. 8,155,156 Other regulatory Tcell<br />

populations that are likely to be very important<br />

in preventing food allergies include T<br />

regulator-1 (Tr1) cells, which produce IL-10, and<br />

CD4+CD25+ cells. 9,129,156 The transcription factor<br />

Foxp3 appears to be central in the commitment of<br />

naive T cells towards the regulatory pathway in<br />

mice. 157 Similar relevance in humans is suggested<br />

by the development of a multifocal inflammatory<br />

condition (IPEX syndrome) in infants with mutations<br />

in Foxp3. 158 Thus, Foxp3 may be a molecule<br />

that is of critical importance in maintaining oral<br />

and systemic tolerance. There are currently no<br />

The basic mechanisms of immune response to dietary antigen 339<br />

clear data to determine which regulatory cell type<br />

is most relevant in prevention of childhood food<br />

allergy. There is certainly evidence that a multiply<br />

exposed population of elderly circulating CD4<br />

+CD25+ cells inhibits milk responses in adult<br />

humans. 159 However, which would be unlikely to<br />

be the case in early life, when most T cells are<br />

initially of the naive phenotype? Analysis of circulating<br />

CD4+CD25+ in adults shows that the<br />

majority express the skin-homing marker CLA, but<br />

not the gut-homing β7 integrin, while cord blood<br />

CD25+CD4+ cells express neither CLA nor β7<br />

integrin. 160 Evidence of functional immaturity of<br />

the neonatal CD4+CD25+ cell population, at least<br />

in mice, is provided by data showing that adult<br />

CD4+CD25+ cells prevent an autoimmune<br />

response to the autoantigen myelin oligodendrocyte<br />

protein, whereas cord blood cells do not. 161<br />

This is likely to be functionally important in<br />

responses to ingested antigen, as neonatal animals<br />

show impaired low-dose oral tolerance and may<br />

even paradoxically sensitize. 162<br />

There are now extensive data to suggest that oral<br />

tolerance is not innate, and that the mechanisms<br />

are not present at birth but develop postnatally.<br />

The expression of a specific array of toll receptors<br />

on CD4+CD25+ regulatory T cells, and increase of<br />

their suppressor functions by bacterial<br />

lipopolysaccharide, 163 suggest that the early infectious<br />

exposures of the young infant are indeed<br />

likely to be important in the generation of oral<br />

tolerance and the prevention of allergy. The<br />

current data on human infants point to a particular<br />

role for TGF-β rather than IL-10 in the prevention<br />

of infant allergy. Study of spontaneous and<br />

cow’s milk-stimulated cytokine production of cord<br />

blood mononuclear cells identified a reduced TGFβ<br />

but not IL-10 response in children of allergic<br />

mothers. 164 The dominant cytokine abnormality in<br />

the mucosa of food-allergic children does not<br />

appear to be diminished Th1 or excess Th2<br />

responses, but reduced numbers of TGF-β secreting<br />

Th3 cells. 34 Other studies have confirmed that<br />

Th1 responses are normal or even increased<br />

within the mucosa in childhood food allergy, 31,33,34<br />

but that TGF-β responses are impaired: expression<br />

of TGF-β1 and its receptor are diminished within<br />

the mucosa in food-allergic enterocolitis, 165 while<br />

milk-reactive T-cell clones from milk-allergic children<br />

show a Th2-deviated response but with


340<br />

Food allergies<br />

minimal TGF-β production. 166 The factors<br />

involved in early life generation of TGF-β1-producing<br />

Th3 cells are thus likely to be of great importance<br />

in determining whether food-allergic sensitization<br />

occurs. Infectious exposures are certainly<br />

one such factor, and it is notable that the density of<br />

TGF-β-producing cells within the duodenal<br />

mucosa of infants in rural Gambia was an order of<br />

magnitude higher than in healthy UK infants. 167 It<br />

is likely that early-life immunomodulation of regulatory<br />

responses, rather than alteration of dietary<br />

exposures, will prove the way ahead in childhood<br />

food allergies.<br />

Future challenges and opportunities<br />

in food allergy<br />

There have been substantial recent advances in<br />

the basic science of food allergies. There has been<br />

a broadening of the concepts of food allergy, away<br />

from simple focus on IgE and towards a consideration<br />

of overall mucosal tolerance. Could a genetic<br />

tendency to high IgE responses simply make<br />

adverse immunological reactions to foods more<br />

noticeable? As many practitioners are uncomfortable<br />

without supporting diagnostic tests, non-IgEmediated<br />

allergy may remain a difficult and<br />

controversial clinical area. Absence of specific<br />

tests can also lead to overdiagnosis of allergies, or<br />

inappropriate blaming of non-specific symptoms<br />

on food allergy – as can be seen on large numbers<br />

of Internet sites.<br />

The advances in basic research, which encompass<br />

both gut inflammation and allergy because of<br />

shared tolerance mechanisms, may explain some<br />

of the recent demographic shifts in allergy.<br />

Inappropriate infectious priming of the nascent<br />

mucosal immune system may affect the development<br />

of normal gut tolerance. Handling of the<br />

newborn infant has been shown in one study to<br />

affect allergy in young adulthood. 168 There is also<br />

clear evidence that the early gut colonization of<br />

allergic infants differs from those without allergies.<br />

169 One study which thus demonstrated great<br />

promise was the recent placebo-controlled trial by<br />

Isolauri’s group in which neonatal administration<br />

of a probiotic organism (Lactobacillus GG) led to a<br />

50% reduction in the later development of eczema,<br />

although without alteration of systemic IgE<br />

responses at either 1 year or 4 years. 10,11 The use of<br />

probiotics at birth may be more effective than later,<br />

as it may allow stable long-term colonization,<br />

which does not occur if probiotics are administered<br />

even at the age of 10 months. 170 As interaction<br />

between bacterial exposures in early infancy<br />

and genetically determined responses in innate<br />

immune cells may determine whether an adequate<br />

tolerogenic response occurs, probiotics may represent<br />

an important new class of immunomodulators,<br />

particularly if used in early infancy. However,<br />

much work remains to be done to determine the<br />

dosage, timing and nature of the probiotics to be<br />

used. 9 An alternative approach to programming a<br />

Th1 or tolerant response is to use bacterial products,<br />

such as mycobacterial peptides. A suspension<br />

made from killed Mycobacterium vaccae<br />

inhibited airway eosinophilia in a murine model of<br />

allergy, through the induction of an allergenspecific<br />

regulatory T-cell response, dependent on<br />

TGF-β and IL-10. 171 Such therapy may offer a<br />

refined alternative to probiotic therapy, in which<br />

regulatory T-cell generation is the therapeutic goal.<br />

However, much safety information needs to be<br />

accumulated, and placebo-controlled trials<br />

completed, before such therapies can become<br />

more widely used.<br />

There are other exciting potential therapies for<br />

food allergies, some of which have been subject to<br />

clinical trials in humans. 172 The area of<br />

immunotherapy is well established for systemic<br />

allergies such as bee-sting allergy, and has<br />

depended on increasing the dose of antigen gradually<br />

until an allergy-suppressing Th1 response is<br />

made. For food allergies, the use of small peptides,<br />

foods with altered protein sequences, DNA immunizations<br />

and IgE-blocking agents represent future<br />

targets for immunotherapy. For small peptide<br />

vaccines, an antigenic peptide is sequenced and<br />

synthetic 10–20 amino acid portions are then<br />

produced, covering the entire protein sequence.<br />

While able to block IgE binding sites, they are not<br />

long enough to cross-link IgE on mast cells. Food<br />

proteins can also be engineered, which are able to<br />

bind to T cells but not mast cells. In recent studies,<br />

the major peanut proteins Ara h1, ara h2 and ara<br />

h3 have been purified, their T-cell and IgE-binding<br />

domains elicited, and mutations made in the IgEbinding<br />

domain. 173


While much of the data outlined above suggests<br />

that IL-5 responses may be critical in food allergies,<br />

the therapeutic credentials of a highly<br />

promising anti-IL-5 monoclonal antibody were<br />

dented by a study in asthmatic patients, in which<br />

peripheral eosinophilia was reduced, but without<br />

any significant modulation of the late bronchoconstrictor<br />

response in asthma. 174 The results of<br />

clinical trials in severe food allergy will be<br />

extremely interesting, provided that such agents<br />

remain in clinical development after such a<br />

setback. Similarly, a monoclonal antibody that<br />

blocks eotaxin chemoattraction may prevent the<br />

migration of eosinophils into the mucosa in<br />

response to food allergens.<br />

Relevant therapeutic antibodies that have reached<br />

clinical trials include two anti-IgE monoclonals.<br />

The humanized monoclonal anti-IgE rhu Mab E-<br />

25, which binds to the constant region of IgE, and<br />

thus prevents IgE binding to its high- or low-affinity<br />

receptors, has shown promising effects in allergic<br />

asthma. 175 In a potentially very important<br />

study, the humanized IgG1 anti-IgE monoclonal<br />

TNX-901 showed clear promise in the treatment of<br />

established peanut allergy. 113 Using a dose of<br />

450mg, given subcutaneously at 4-weekly intervals<br />

for 16 weeks, treated patients showed an<br />

increase in reaction threshold to peanut from 178<br />

to 2805g, essentially the difference between half a<br />

peanut and nine peanuts. A dose-dependent<br />

increase in reaction threshold was seen from<br />

150–450mg doses. This represents a clinically<br />

worthwhile increase in reaction threshold, and<br />

would substantially reduce the chances of inadvertent<br />

consumption of sufficient peanut to trigger<br />

anaphylaxis.<br />

In addition to such radical advances in therapy,<br />

one future challenge in the field of food allergy<br />

will be provided by the advent of genetically modified<br />

foods, which have so far an unknown propensity<br />

for causing allergic reactions. Genes may be<br />

Future challenges and opportunities in food allergy 341<br />

introduced into plants either through use of a<br />

bacterial vector or by direct physical methods,<br />

while alternatively naturally occurring genes may<br />

be silenced. 176 Important lessons have already<br />

been learned. In an attempt to increase the nutritional<br />

component of cattle feed, the brazil nut 2S<br />

albumen protein was introduced transgenically<br />

into soy. Unfortunately, this protein turned out to<br />

be a major brazil nut allergen, and indeed the<br />

transgenic soy was able to induce hypersensitivity<br />

in brazil nut-allergic patients. 177 Therefore, avoidance<br />

of similar adverse events for the future must<br />

be minimized by appropriate predictions.<br />

Recognition of potentially allergic molecules can<br />

be attempted, on the basis that many are large and<br />

heavily glycosylated molecules that are resistant to<br />

breakdown by proteolysis or digestion. 178<br />

However, some antigens, particularly fruit allergens,<br />

do not follow these rules. One suggestion has<br />

thus been to test proteins from genes considered<br />

for transgenic insertion by immunoassay against<br />

sera from a variety of allergic patients. 179<br />

Even if genetically modified foods do not reach the<br />

market place in large amounts, there still remains<br />

the major challenge of ensuring that modern food<br />

manufacturing processes do not leave food-allergic<br />

patients at risk of anaphylaxis. Taylor et al 180 have<br />

published an important call for the determination<br />

of the minimal doses of antigen required to trigger<br />

reactions, and for international legislation to<br />

ensure that food manufacturers do not exceed<br />

these doses. The dietary exposure of infants and<br />

young children have changed out of all recognition<br />

within developing countries in the past decades, at<br />

a time when their infectious exposures have also<br />

been altered in a way that evolution has not<br />

prepared them for. The challenge of preventing<br />

food-allergic deaths is one in which food manufacturers<br />

may have to work together with basic scientists,<br />

and in which microbiologists may have as<br />

much to offer as immunologists. These are interesting<br />

times.


342<br />

Food allergies<br />

REFERENCES<br />

1. Walker-Smith JA, Murch SH. Gastrointestinal food<br />

allergy. In Diseases of the Small Intestine in Childhood,<br />

4th edn. Oxford: Isis Medical Media, 1999: 205–234.<br />

2. Wood RA. The natural history of food allergy. Pediatrics<br />

2003; 111: 1631–1637.<br />

3. Ewan P. Clinical study of peanut and nut allergy in 62<br />

consecutive patients: new features and associations.<br />

BMJ 1996; 312: 1074–1078.<br />

4. Hourihane JO. Peanut allergy – current status and future<br />

challenges. Clin Exp Allergy 1997; 27: 1240–1246.<br />

5. Hill DJ, Hosking CS, Heine RG. Clinical spectrum of<br />

food allergy in children in Australia and South-East<br />

Asia: identification and targets for treatment. Ann Med<br />

1999; 31: 272–281.<br />

6. Goh DL, Chew FT, Chua KY et al. Edible ‘bird’s nest’induced<br />

anaphylaxis: an under-recognized entity? J<br />

Pediatr 2000; 137: 277–279.<br />

7. Rook GAW, Stanford JL. Give us this day our daily<br />

germs. Immunol Today 1998; 19: 113–116.<br />

8. Murch SH. The immunologic basis for intestinal food<br />

allergy. Curr Opin Gastroenterol 2000; 16: 552–557.<br />

9. Murch SH. Toll of allergy reduced by probiotics. Lancet<br />

2001; 357: 1057–1059.<br />

10. Kalliomaki M, Salminen S, Arvillomi H et al. Probiotics<br />

in primary prevention of atopic disease: a randomised<br />

placebo-controlled trial. Lancet 2001; 357: 1076–1079.<br />

11. Kalliomaki M, Salminen S, Poussa T et al. Probiotics<br />

and prevention of atopic disease: 4-year follow-up of a<br />

randomised placebo-controlled trial. Lancet 2003; 361:<br />

1869–1871.<br />

12. Gell PGH, Coombs RRA. Classification of allergic reactions<br />

responsible for hypersensitivity and disease. In<br />

Gell PGH, Coombs RRA, eds. Clinical Aspects of<br />

Immunol. Oxford: Blackwell, 1968: 575.<br />

13. Clynes R, Dumitru C, Ravetch JC. Uncoupling of<br />

immune complex formation and kidney damage in<br />

autoimmune glomerulonephritis. Science 1998; 279:<br />

1052–1054.<br />

14. Latcham F, Merino F, Lang A et al. A consistent pattern<br />

of minor immunodeficiency and subtle enteropathy in<br />

children with multiple food allergy. J Pediatr 2003; 43:<br />

39–47.<br />

15. Oettgen HC, Martin TR, Wynshaw-Boris A et al. Active<br />

anaphylaxis in IgE-deficient mice. Nature 1994; 370:<br />

367–370.<br />

16. Miyajima I, Dombrowicz D, Martin TR et al. Systemic<br />

anaphylaxis in the mouse can be mediated largely<br />

through IgG1 and FcgRIII. Assessment of the cardiopulmonary<br />

changes, mast cell degranulation and death<br />

associated with active or IgE- or IgG1-dependent passive<br />

anaphylaxis. J Clin Invest 1997; 99: 901–914.<br />

17. Hill DJ, Firer MA, Shelton MJ, Hosking CS.<br />

Manifestations of milk allergy in infancy: clinical and<br />

immunologic findings. J Pediatr 1986; 109: 270–276.<br />

18. Hill DJ, Ball G, Hosking CS, Wood PR. Gamma-interferon<br />

production in cow milk allergy. Allergy 1993; 48:<br />

75–80.<br />

19. Soothill JF, Stokes CR, Turner MW et al. Predisposing<br />

factors and the development of reaginic allergy in<br />

infancy. Clin Allergy 1976; 6: 305–319.<br />

20. Ludviksson BR, Elriksson TH, Ardal B et al. Correlation<br />

between serum immunoglobulin A concentrations and<br />

allergic manifestations in infants. J Pediatr 1992; 121:<br />

23–27.<br />

21. Järvinen KM, Aro A, Juntunen-Backman K,<br />

Suomalainen H. Large number of CD19 + /CD23 + B cells<br />

and small number of CD8 + T cells as early markers for<br />

cow’s milk allergy (CMA). Pediatr Allergy Immunol<br />

1998; 9: 139–142.<br />

22. Ojuawo A, St Louis D, Lindley KJ, Milla PJ. Non-infective<br />

colitis in infancy: evidence in favour of minor<br />

immunodeficiency in its pathogenesis. Arch Dis Child<br />

1997; 76: 345–348.<br />

23. Pérez-Machado MA, Ashwood P, Thomson MA et al.<br />

Reduced transforming growth factor-β1 producing T<br />

cells in the duodenal mucosa of children with food<br />

allergy. Eur J Immunol 2003; 33: 2307–2315.<br />

24. Sicherer SH. Clinical aspects of gastrointestinal food<br />

allergy in childhood. Pediatrics 2003; 111: 1609 –1616.<br />

25. Sampson HA. Anaphylaxis and emergency treatment.<br />

Pediatrics 2003; 111: 1601–1608.<br />

26. Pumphrey RS. Lessons for management of anaphylaxis<br />

from a study of fatal reactions. Clin Exp Allergy 2000;<br />

30: 1144–1150.<br />

27. Macdougall CF, Cant AJ, Colver AF. How dangerous is<br />

food allergy in childhood? The incidence of severe and<br />

fatal allergic reactions across the UK and Ireland. Arch<br />

Dis Child. 2002; 86: 236–239.<br />

28. Anveden HL, Finkel Y, Sandstedt B, Karpe B.<br />

Proctocolitis in exclusively breast-fed infants. Eur J<br />

Pediatr 1996; 155: 464–467.<br />

29. Kokkonen J, Karttunen TJ. Lymphonodular hyperplasia<br />

on the mucosa of the lower gastrointestinal tract in children:<br />

an indication of enhanced immune response? J<br />

Pediatr Gastroenterol Nutr 2002; 34: 42–46.<br />

30. Kokkonen J, Ruuska T, Karttunen TJ, Maki M.<br />

Lymphonodular hyperplasia of the terminal ileum associated<br />

with colitis shows an increase γδ+ T cell density<br />

in children. Am J Gastroenterol 2002; 97: 667–672.<br />

31. Hauer AC, Breese E, Walker-Smith JA, MacDonald TT.<br />

The frequency of cells secreting interferon-γ, interleukin-4,<br />

interleukin-5 and interleukin-10 in the blood<br />

and duodenal mucosa of children with cow’s milk<br />

hypersensitivity. Pediatr Res 1997; 42: 629–638.<br />

32. Walker-Smith JA, Murch SH. Enterocyte proliferation<br />

and functions. In Diseases of the Small Intestine in<br />

Childhood, 4th edn. Oxford: Isis Medical Media, 1999:<br />

29–43.<br />

33. Veres G, Westerholm-Ormio M, Kokkonen J et al.<br />

Cytokines and adhesion molecules in duodenal mucosa<br />

of children with delayed-type food allergy. J Pediatr<br />

Gastroenterol Nutr 2003; 37: 27–34.<br />

34. Pérez-Machado MA, Ashwood P, Torrente F et al.<br />

Spontaneous Th1 cytokine production by intraepithelial,<br />

but not circulating T cells, in infants with or<br />

without food allergies. Allergy 2004; 59: 346–353.<br />

35. Schappi MG, Smith VV, Cubitt D et al. Faecal elastase 1<br />

concentration is a marker of duodenal enteropathy. Arch<br />

Dis Child 2002; 86: 50–53.<br />

36. Kokkonen J, Haapalahti M, Laurila K et al. Cow’s milk<br />

protein-sensitive enteropathy at school age. J Pediatr<br />

2001; 139: 797–803.<br />

37. Burks W. Skin manifestations of food allergy. Pediatrics<br />

2003; 111: 1617–1624.<br />

38. Sampson HA, Broadbent KR, Bernhisel-Broadbent J.<br />

Spontaneous release of histamine from basophils and<br />

histamine-releasing factor in patients with atopic<br />

dermatitis and food hypersensitivity. N Engl J Med 1989;<br />

321: 228–232.<br />

39. Majamaa H, Isolauri E. Evaluation of the gut mucosal<br />

barrier: evidence for increased antigen transfer in children<br />

with atopic eczema. J Allergy Clin Immunol 1996;<br />

97: 985–990.


40. Abernathy-Carver KJ, Sampson HA, Picker LJ, Leung<br />

DY. Milk-induced eczema is associated with the expansion<br />

of T cells expressing cutaneous lymphocyte<br />

antigen. J Clin Invest 1995; 95: 913–918.<br />

41. Leung DY, Gately M, Trumble A et al. Bacterial superantigens<br />

induce T cell expression of the skin-selective<br />

homing receptor, the cutaneous lymphocyte-associated<br />

antigen, via stimulation of interleukin 12 production. J<br />

Exp Med 1995; 181: 747–753.<br />

42. Nissen D, Pedersen LJ, Skov PS et al. IgE-binding<br />

components of staphylococcal enterotoxins in patients<br />

with atopic dermatitis. Ann Allergy Asthma Immunol<br />

1997; 79: 403–408.<br />

43. Sohn MH, Kim CH, Kim WK et al. Effect of staphylococcal<br />

enterotoxin B on specific antibody production in<br />

children with atopic dermatitis. Allergy Asthma Proc<br />

2003; 24: 67–71.<br />

44. Ong PY, Ohtake T, Brandt C et al. Endogenous antimicrobial<br />

peptides and skin infections in atopic dermatitis.<br />

N Engl J Med 2002; 347: 1151–1160.<br />

45. Hill DJ, Sporik R, Thorburn J, Hosking CS. The association<br />

of atopic dermatitis in infancy with immunoglobulin<br />

E food sensitization. J Pediatr 2000; 137: 475–479.<br />

46. Kjellman B, Hattevig G. Allergy in early and late onset<br />

atopic dermatitis. Acta Pediatr 1994; 83: 229–231.<br />

47. Atherton DJ, Sewell M, Soothill JF et al. A double-blind<br />

controlled crossover trial of an antigen-avoidance diet<br />

in atopic eczema. Lancet 1978; 1: 401–403.<br />

48. Kekki OM, Turjanmaa K, Isolauri E. Differences in skinprick<br />

and patch-test reactivity are related to the heterogeneity<br />

of atopic eczema in infants. Allergy 1997; 52:<br />

755–759.<br />

49. Isolauri E, Arvola T, Sutas Y et al. Probiotics in the<br />

management of atopic eczema. Clin Exp Allergy 2000;<br />

30: 1604–1610.<br />

50. Isolauri E, Tabvanainen A, Peltola T, Arvola T. Breastfeeding<br />

of allergic infants. J Pediatr 1999; 134: 27–32.<br />

51. Hill DJ, Cameron DJ, Francis DE et al. Challenge confirmation<br />

of late-onset reactions to extensively hydrolyzed<br />

formulas in infants with multiple food protein intolerance.<br />

J Allergy Clin Immunol 1995; 96: 386–394.<br />

52. de Boissieu D, Dupont C. Time course of allergy to<br />

extensively hydrolyzed cow’s milk proteins in infants. J<br />

Pediatr 2000; 136: 119–120.<br />

53. Cavataio F, Iacono G, Montalto G et al. Clinical and pHmetric<br />

characteristics of gastroesophageal reflux<br />

secondary to cows’ milk protein allergy. Arch Dis Child<br />

1996; 75: 51–56.<br />

54. Iacono G, Cavataio F, Montalto G et al. Intolerance of<br />

cow’s milk and chronic constipation in children. N Engl<br />

J Med 1998; 339: 1100–1104.<br />

55. Butt AM, Murch SH, Ng C et al. Upregulated eotaxin<br />

expression and T cell infiltration in the basal<br />

oesophageal epithelium in cow’s milk-associated reflux<br />

esophagitis. Arch Dis Child 2002; 87: 124–130.<br />

56. Iacono G, Carroccio A, Montalto G et al. Severe infantile<br />

colic and food intolerance: a long-term prospective<br />

study. J Pediatr Gastroenterol Nutr 1991; 12: 332–335.<br />

57. Heine RG, Elsayed S, Hosking CS, Hill DJ. Cow’s milk<br />

allergy in infancy. Curr Opin Allergy Clin Immunol 2002;<br />

2: 217–225.<br />

58. Kelly KJ, Lazenby AJ, Rowe PC et al. Eosinophilic<br />

esophagitis attributed to gastroesophageal reflux:<br />

improvement with an amino acid-based formula.<br />

Gastroenterology 1995; 109: 1503–1512.<br />

59. Vanderhoof JA, Murray ND, Kaufman SS et al.<br />

Intolerance to protein hydrolysate infant formulas: an<br />

underrecognized cause of gastrointestinal symptoms in<br />

infants. J Pediatr 1997; 131: 741–744.<br />

References 343<br />

60. Kokkonen J, Tikkanen S, Karttunen TJ, Savilahti E. A<br />

similar high level of immunoglobulin A and<br />

immunoglobulin G class milk antibodies and increment<br />

of local lymphoid tissue on the duodenal mucosa in<br />

subjects with cow’s milk allergy and recurrent<br />

abdominal pains. Pediatr Allergy Immunol 2002; 13:<br />

129–136.<br />

61. Shah N, Lindley K, Milla P. Cow’s milk and chronic constipation<br />

in children. N Engl J Med 1999; 340: 891–892.<br />

62. Furuta GT. Eosinophilic esophagitis: an emerging<br />

clinicopathologic entity. Curr Allergy Asthma Rep 2002;<br />

2: 67–72.<br />

63. Khan S, Orenstein SR. Eosinophilic gastroenteritis:<br />

epidemiology, diagnosis and management. Paediatr<br />

Drugs 2002; 4: 563–570.<br />

64. Orenstein SR, Shalaby TM, Di Lorenzo C et al. The<br />

spectrum of pediatric eosinophilic esophagitis beyond<br />

infancy: a clinical series of 30 children. Am J<br />

Gastroenterol 2000; 95: 1422–1430.<br />

65. Vasilopoulos S, Murphy P, Auerbach A et al. The smallcaliber<br />

esophagus: an unappreciated cause of dysphagia<br />

for solids in patients with eosinophilic esophagitis.<br />

Gastrointest Endosc 2002; 55: 99–106.<br />

66. Fox VL, Nurko S, Teitelbaum JE et al. High-resolution<br />

EUS in children with eosinophilic ‘allergic’ esophagitis.<br />

Gastrointest Endosc 2003; 57: 30–36.<br />

67. Thomson MA. The pediatric esophagus comes of age. J<br />

Pediatr Gastroenterol Nutr 2002; 34: S40–45.<br />

68. Spergel JM, Beausoleil JL, Mascarenhas M, Liacouras<br />

CA. The use of skin prick tests and patch tests to identify<br />

causative foods in eosinophilic esophagitis. J Allergy<br />

Clin Immunol 2002; 109: 363–368.<br />

69. Teitelbaum JE, Fox VL, Twarog FJ et al. Eosinophilic<br />

esophagitis in children: immunopathological analysis<br />

and response to fluticasone propionate. Gastroenterology<br />

2002; 122: 1216–1225.<br />

70. Attwood SE, Lewis CJ, Bronder CS et al. Eosinophilic<br />

esophagitis: a novel treatment using Montelukast. Gut<br />

2003; 52: 181–185.<br />

71. James JM. Respiratory manifestations of food allergy.<br />

Pediatrics 2003; 111: 1625–1630.<br />

72. Khoshoo V, Le T, Haydel RM Jr et al. Role of gastroesophageal<br />

reflux in older children with persistent<br />

asthma. Chest 2003; 123: 1008–1013.<br />

73. The European Society for Paediatric Gastroenterology<br />

and Nutrition Group for the diagnostic criteria for food<br />

allergy. Diagnostic criteria for food allergy with predominantly<br />

intestinal symptoms. J Pediatr Gastroenterol Nutr<br />

1992; 14: 108–112.<br />

74. Bock SA. Diagnostic evaluation. Pediatrics 2003; 111:<br />

1638–1644.<br />

75. Caffarelli C, Petroccione T. False-negative food challenges<br />

in children with suspected food allergy. Lancet<br />

2001; 358: 1871–1872.<br />

76. Carroccio A, Montalto G, Custro N et al. Evidence of<br />

very delayed clinical reactions to cow’s milk in cow’s<br />

milk-intolerant patients. Allergy 2000; 55: 574–579.<br />

77. Sampson HA, Sicherer SH, Bimbaum AH. AGA technical<br />

review on the evaluation of food allergy in gastrointestinal<br />

disorders. American Gastroenterological<br />

Association. Gastroenterology 2001; 120: 1026–40.<br />

78. Sampson HA. Use of food challenge tests in children.<br />

Lancet 2001; 358: 1832–1833.<br />

79. Roberts G, Lack G. Getting more out of your skin prick<br />

tests. Clin Exp Allergy 2000; 30: 1495–1498.<br />

80. Sporik R, Hill DJ, Hosking CS. Specificity of allergen<br />

skin testing in predicting positive open food challenges<br />

to milk, egg and peanut in children. Clin Exp Allergy<br />

2000; 30: 1540–1546.


344<br />

Food allergies<br />

81. Hill DJ, Hosking CS, Reyes-Benito MLV. Reducing the<br />

need for food allergen challenges in young children:<br />

comparison of in vitro with in vivo tests. Clin Exp<br />

Allergy 2001; 31: 1031–1035.<br />

82. Fagan TJ. Nomogram for Bayes’s theorem. N Engl J Med<br />

1975; 293: 257.<br />

83. Isolauri E, Turjanmaa K. Combined skin prick and patch<br />

testing enhances identification of food allergy in infants<br />

with atopic dermatitis. J Allergy Clin Immunol 1996; 97:<br />

9–15.<br />

84. Roehr CC, Reibel S, Ziegert M et al. Atopy patch tests,<br />

together with determination of specific IgE levels,<br />

reduce the need for oral food challenges in children<br />

with atopic dermatitis. J Allergy Clin Immunol 2001;<br />

107: 548–553.<br />

85. Niggemann B, Reibel S, Wahn U. The atopy patch test<br />

(APT) – a useful tool for the diagnosis of food allergy in<br />

children with atopic dermatitis. Allergy 2000; 55:<br />

281–285.<br />

86. Stromberg L. Diagnostic accuracy of the atopy patch test<br />

and the skin-prick test for the diagnosis of food allergy<br />

in young children with atopic eczema/dermatitis<br />

syndrome. Acta Paediatr 2002; 91: 1044–1049.<br />

87. Wistokat-Wulfing A, Schmidt P, Darsow U et al. Atopy<br />

patch test reactions are associated with T lymphocytemediated<br />

allergen-specific immune responses in atopic<br />

dermatitis. Clin Exp Allergy 1999; 29: 513–521.<br />

88. Sampson HA, Ho DG. Relationship between foodspecific<br />

IgE concentrations and the risk of positive food<br />

challenges in children and adolescents. J Allergy Clin<br />

Immunol 1997; 100: 444–451.<br />

89. Sicherer SH, Sampson HA. Cow’s milk protein-specific<br />

IgE concentrations in two age groups of milk-allergic<br />

children and in children achieving clinical tolerance.<br />

Clin Exp Allergy 1999; 29: 507–512.<br />

90. Cooke SK, Sampson HA. Allergenic properties of<br />

ovomucoid in man. J Immunol 1997;159: 2026–2032.<br />

91. Vila L, Beyer K, Jarvinen KM et al. Role of<br />

conformational and linear epitopes in the achievement<br />

of tolerance in cow’s milk allergy. Clin Exp Allergy 2001;<br />

31: 1599–1606.<br />

92. Järvinen KM, Beyer K, Vila L et al. B-cell epitopes as a<br />

screening instrument for persistent cow’s milk allergy. J<br />

Allergy Clin Immunol 2002; 110: 293–297.<br />

93. Schade RP, Van Leperen-Van Dijk AG, Van Reijsen FC.<br />

Differences in antigen-specific T-cell responses between<br />

infants with atopic dermatitis with and without cow’s<br />

milk allergy: relevance of Th2 cytokines. J Allergy Clin<br />

Immunol 2000; 106: 1155–1162.<br />

94. Turcanu V, Maleki SJ, Lack G. Characterization of<br />

lymphocyte responses to peanuts in normal children,<br />

peanut-allergic children, and allergic children who<br />

acquired tolerance to peanuts. J Clin Invest 2003; 111:<br />

1065–1072.<br />

95. Heyman M, Darmon N, Dupont C et al. Mononuclear<br />

cells from infants allergic to cow’s milk secrete tumor<br />

necrosis factor α, altering intestinal function.<br />

Gastroenterology 1994; 106: 1514–1523.<br />

96. Nagata S, McKenzie C, Pender SL et al. Human Peyer’s<br />

patch T cells are sensitized to dietary antigen and<br />

display a Th cell type 1 cytokine profile. J Immunol<br />

2000; 165: 5315–5321.<br />

97. Kilby A, Walker-Smith JA, Wood CBS. Small intestinal<br />

mucosa in cow’s milk allergy. Lancet 1975; 1: 53.<br />

98. Boyano-Martinez T, Garcia-Ara C, Diaz-Pena JM, Martin-<br />

Esteban M. Prediction of tolerance on the basis of<br />

quantification of egg white-specific IgE antibodies in<br />

children with egg allergy. J Allergy Clin Immunol 2002;<br />

110: 304–309.<br />

99. American Academy of Pediatrics, Committee on<br />

Nutrition. Hypoallergenic<br />

infant formulas. Pediatrics 2000; 106: 346–349.<br />

100. Eastham EJ, Lichanco T, Pang K, Walker WA.<br />

Antigenicity of infant formulas and the induction of<br />

systemic immunological tolerance by oral feeding:<br />

cow’s milk versus soy milk. J Pediatr Gastroenterol Nutr<br />

1982; 1: 23–28.<br />

101. Rozenfeld P, Docena GH, Anon MC, Fossati CA.<br />

Detection and identification of a soy protein component<br />

that cross-reacts with caseins from cow’s milk. Clin Exp<br />

Immunol 2002; 130: 49–58.<br />

102. Franck P, Moneret Vautrin DA, Dousset B et al. The<br />

allergenicity of soybean-based products is modified by<br />

food technologies. Int Arch Allergy Immunol 2002; 128:<br />

212–219.<br />

103. Rudd P, Manuel PD, Walker-Smith JA. Anaphylactic<br />

shock in an infant after feeding with a wheat rusk. A<br />

transient phenomenon. Postgrad Med J 1981; 57:<br />

794–795.<br />

104. Palosuo K, Varjonen E, Kekki OM et al. Wheat omega-5<br />

gliadin is a major allergen in children with immediate<br />

allergy to ingested wheat. J Allergy Clin Immunol 2001;<br />

108: 634–638.<br />

105. Catassi C, Ratsch IM, Fabiani E et al. Coeliac disease in<br />

the year 2000: exploring the iceberg. Lancet 1994; 343:<br />

200–203.<br />

106. Knivsberg AM, Reichelt KL, Hoien T, Nodland M. A<br />

randomised, controlled study of dietary intervention in<br />

autistic syndromes. Nutr Neurosci 2002; 5: 251–261.<br />

107. Wakefield AJ, Puleston JM, Montgomery SM et al.<br />

Entero-colonic encephalopathy, autism and opioid<br />

receptor ligands. Aliment Pharmacol Therapeut 2002;<br />

16: 663–674.<br />

108. Sampson HA, Mendelson L, Rosen JP. Fatal and nearfatal<br />

anaphylactic reactions to food in children and<br />

adolescents. N Engl J Med 1992; 327: 380–384.<br />

109. Lack G, Fox D, Northstone K, Golding J, for the Avon<br />

Longitudinal Study of Parents and Children Study<br />

Team. Factors associated with the development of<br />

peanut allergy in childhood. N Engl J Med 2003; 348:<br />

977–985.<br />

110. Legendre C, Caillat-Zucman S, Samuel D et al. Transfer<br />

of symptomatic peanut allergy to the recipient of a<br />

combined liver-and-kidney transplant. N Engl J Med<br />

1997; 337: 822–824.<br />

111. Dearman RJ, Caddick H, Stone S et al. Characterization<br />

of antibody responses induced in rodents by exposure<br />

to food proteins: influence of route of exposure.<br />

Toxicology 2001; 167: 217–231.<br />

112. Watzl B, Neudecker C, Hansch GM et al. Dietary wheat<br />

germ agglutinin modulates ovalbumin-induced immune<br />

responses in Brown Norway rats. Br J Nutr 2001; 85:<br />

483–490.<br />

113. Leung DYM, Sampson HA, Yunginger JW et al, for the<br />

TNX-901 Peanut Allergy Study Group. Effect of anti-IgE<br />

therapy in patients with peanut allergy. N Engl J Med<br />

2003; 348: 986–993.<br />

114. Murch S. Diabetes and cows’ milk. Lancet 1996; 348: 1656.<br />

115. Zeiger RS. Food allergen avoidance in the prevention of<br />

food allergy in infants and children. Pediatrics 2003;<br />

111: 1662–1671.<br />

116. Host A, Koletzko B, Dreborg S et al. Joint Statement of<br />

the European Society for Paediatric Allergology and<br />

Clinical Immunology (ESPACI) Committee on<br />

Hypoallergenic Formulas and the European Society for<br />

Paediatric Gastroenterology, Hepatology and Nutrition<br />

(ESPGHAN) Committee on Nutrition. Arch Dis Child<br />

1999; 81: 80–84.


117. von Berg A, Koletzko S, Grubl A, Filipiak-Pittroff B,<br />

Wichmann HE, Bauer CP, Reinhardt D, Berdel D;<br />

German Infant Nutritional Intervention Study Group.<br />

The effect of hydrolyzed cow’s milk formula for allergy<br />

prevention in the first year of life: the German Infant<br />

Nutritional Intervention Study, a randomized doubleblind<br />

trial. J Allergy Clin Immunol 2003; 111: 533–540.<br />

118. Zhu D, Zhang X, Chen L. Reinterpretation of<br />

Yunnanozoon as the earliest known hemichordate.<br />

Nature 1996; 380: 428–430.<br />

119. Medzhitov R, Janeway CA Jr. Innate immunity: impact<br />

on the adaptive immune response. Curr Opin Immunol<br />

1997; 9: 4–9.<br />

120. Aderem A, Ulevitch RJ. Toll-like receptors in the<br />

induction of the innate immune response. Nature 2000;<br />

406: 782–787.<br />

121. Sudo N, Sawamura S, Tanaka K et al. The requirement<br />

of intestinal bacterial flora for the development of an<br />

IgE production system susceptible to oral tolerance<br />

induction. J Immunol 1997; 159: 1739–1745.<br />

122. Cebra JJ. Influences of microbiota on intestinal immune<br />

system development. Am J Clin Nutr 1999; 69 (Suppl):<br />

1046S–1051S.<br />

123. Newberry RD, Stenson WF, Lorenz RG. Cycloxygenase-<br />

2-dependent arachidonic acid metabolites are essential<br />

modulators of the immune response to dietary antigen.<br />

Nature Med 1999; 5: 900–906.<br />

124. Tlaskalova-Hogenova H, Tuckova L, Lodinova-<br />

Zadnikova R et al. Mucosal immunity: its role in<br />

defense and allergy. Int Arch Allergy Immunol 2002; 128:<br />

77–89.<br />

125. Supajatura V, Ushio H, Nakao A et al. Differential<br />

responses of mast cell Toll-like receptors 2 and 4 in<br />

allergy and innate immunity. J Clin Invest 2002; 109:<br />

1351–1359.<br />

126. Fujihashi K, McGhee JR, Kweon MN et al. γδ T cell-deficient<br />

mice have impaired mucosal immunoglobulin A<br />

responses. J Exp Med 1996; 183: 1929–1935.<br />

127. Ulrichs T, Porcelli SA. CD1 proteins: targets of T cell<br />

recognition in innate and adaptive immunity. Rev<br />

Immunogenet 2000; 2: 416–432.<br />

128. Mayer L. Mucosal immunity and gastrointestinal<br />

antigen processing. J Pediatr Gastroenterol Nutr 2000;<br />

30: S4–S12.<br />

129. Mayer L. Mucosal immunity. Pediatrics 2003; 111:<br />

1595–1600.<br />

130. Miller K, Meredith C, Selo I, Wal JM. Allergy to bovine<br />

beta-lactoglobulin: specificity of immunoglobulin E<br />

generated in the Brown Norway rat to tryptic and<br />

synthetic peptides. Clin Exp Allergy1999; 29:<br />

1696–1704.<br />

131. Fischer A. Primary immunodeficiency diseases: an<br />

experimental model for molecular medicine. Lancet<br />

2001; 357: 1863–1869.<br />

132. Eckmann L, Kagnoff MF, Fierer J. Epithelial cells secrete<br />

the chemokine interleukin-8 in response to bacterial<br />

entry. Infection Immun 1993; 61: 4569–4574.<br />

133. Allez M, Brimnes J, Dotan I, Mayer L. Expansion of<br />

CD8+ T cells with regulatory function after interaction<br />

with intestinal epithelial cells. Gastroenterology 2002;<br />

123: 1516–1526.<br />

134. Hermiston ML, Gordon JI. Inflammatory bowel disease<br />

and adenomas in mice expressing a dominant negative<br />

N-cadherin. Science 1995; 270: 1203–1207.<br />

135. Sigurs N, Hattevig G, Kjellman B et al. Appearance of<br />

atopic disease in relation to serum IgE antibodies in<br />

children followed from birth for 4 to 15 years. J Allergy<br />

Clin Immunol 1994; 94: 757–763.<br />

References 345<br />

136. Corry DB, Kheradmand F. Induction and regulation of<br />

the IgE response. Nature 1999; 402 (Suppl): B18–B23.<br />

137. Hershey GK, Friedrich MF, Esswein LA et al. The association<br />

of atopy with a gain-of-function mutation in the α<br />

subunit of the interleukin-4 receptos. N Engl J Med<br />

1997; 337: 1720–1725.<br />

138. Mitsuyasu H, Izuhara K, Mao XQ et al. Ile50val variant<br />

of IL4Rα upregulates IgE synthesis and associates with<br />

atopic asthma. Nature Genet 1998; 19: 119–120.<br />

139. Grunewald SM, Werthmann A, Schnarr B et al. An<br />

antagonistic IL-4 mutant prevents type I allergy in the<br />

mouse: inhibition of the IL-4/IL-13 receptor system<br />

completely abrogates humoral immune response to<br />

allergen and development of allergic symptoms in vivo.<br />

J Immunol 1998; 160: 4004–4009.<br />

140. Shimoda K, van Deursen J, Sangster MY et al. Lack of<br />

IL-4-induced T cell response and IgE class switching in<br />

mice with disrupted Stat6 gene. Nature 1996; 380:<br />

630–633.<br />

141. Ramaswamy K, Hakimi J, Bell RG. Evidence for an<br />

interleukin-4 inducible immunoglobulin E uptake and<br />

transport mechanism in the intestine. J Exp Med 1994;<br />

180: 1793–1803.<br />

142. Negrao-Correa D, Adams LS, Bell RG. Intestinal transport<br />

and catabolism of IgE: a major blood-independent<br />

pathway of IgE dissemination during a Trichinella<br />

spiralis infection of rats. J Immunol 1996; 157:<br />

4037–4044.<br />

143. Collins SM. The immunomodulation of enteric neuromuscular<br />

function: implications for motility and inflammatory<br />

disorders. Gastroenterology 1996; 111:<br />

1683–1699.<br />

144. Majamaa H, Miettinen A, Laine S, Isolauri E. Intestinal<br />

inflammation in children with atopic eczema: fecal<br />

eosinophil cationic protein and tumor necrosis factor-α<br />

as non-invasive indicators of food allergy. Clin Exp<br />

Allergy 1996; 26: 181–187.<br />

145. Kapel N, Matarazzo P, Haouchine D et al. Fecal tumor<br />

necrosis factor α, eosinophil cationic protein and IgE<br />

levels in infants with cows milk allergy and gastrointestinal<br />

manifestations. Clin Chem Lab Med 1998; 37:<br />

29–32.<br />

146. Santos J, Bayarri C, Saperas E et al. Characterisation of<br />

immune mediator release during the immediate<br />

response to segmental mucosal challenge in the<br />

jejunum of patients with food allergy. Gut 1999; 45:<br />

553–558.<br />

147. Hogan SP, Mishra A, Brandt EB et al. A critical role for<br />

eotaxin in experimental oral antigen-induced<br />

eosinophilic gastrointestinal allergy. Proc Natl Acad Sci<br />

USA 2000; 97: 6681–6686.<br />

148. Hogan SP, Mishra A, Brandt EB, et al. A pathological<br />

function for eotaxin and eosinophils in eosinophilic<br />

gastrointestinal inflammation. Nature Immunol 2001; 2:<br />

353–360.<br />

149. Mishra M, Hogan SP, Brandt EB, Rothenberg ME. An<br />

etiological role for aeroallergens and eosinophils in<br />

experimental esophagitis. J Clin Invest 2001; 107: 83–90.<br />

150. Mishra A, Hogan SP, Brandt EB, Rothenberg ME. IL-5<br />

promotes eosinophil trafficking to the esophagus. J<br />

Immunol 2002; 168: 2464–2469.<br />

151. Vandenzande LM, Wallaert B, Desreumaux P et al.<br />

Interleukin-5 immunoreactivity and mRNA expression<br />

in gut mucosa from patients with food allergy. Clin Exp<br />

Allergy 1999; 29: 652–659.<br />

152. Weiner HL. Oral tolerance: immune mechanisms and<br />

treatment of autoimmune diseases. Immunol Today<br />

1997; 18: 335–343.


346<br />

Food allergies<br />

153. Chen Y, Inobe J, Marks R et al. Peripheral deletion of<br />

antigen-reactive T cells in oral tolerance. Nature 1995;<br />

376: 177–180.<br />

154. Kaji T, Hachimura S, Ise W, Kaminogawa S. Proteome<br />

analysis reveals caspase activation in hyporesponsive<br />

CD4 T lymphocytes induced in vivo by the oral administration<br />

of antigen. J Biol Chem 2003; 278:<br />

27836–27843.<br />

155. Groux H, Powrie F. Regulatory T cells and inflammatory<br />

bowel disease. Immunol Today 1999; 20: 442–446.<br />

156. Strobel S. Oral tolerance, systemic immunoregulation,<br />

and autoimmunity. Ann NY Acad Sci 2002; 958: 47–58.<br />

157. Hori S, Nomura T, Sakaguchi S. Control of regulatory T<br />

cell development by the transcription factor Foxp3.<br />

Science 2003; 299: 1057–1061.<br />

158. Wildin RS, Smyk-Pearson S, Filipovich AH. Clinical and<br />

molecular features of the immunodysregulation, polyendocrinopathy,<br />

enteropathy, X linked (IPEX) syndrome. J<br />

Med Genet 2002; 39: 537–545.<br />

159. Taams LS, Vukmanovic-Stejic M, Smith J et al. Antigenspecific<br />

T cell suppression by human CD4+CD25+<br />

regulatory T cells. Eur J Immunol 2002; 32: 1621–1630.<br />

160. Iellem A, Colantonio L, D’Ambrosio D. Skin- versus gutskewed<br />

homing receptor expression and intrinsic CCR4<br />

expression on human peripheral blood CD4+CD25+<br />

suppressor T cells. Eur J Immunol 2003; 33: 1488–1496.<br />

161. Wing K, Lindgren S, Kollberg G et al. CD4 T cell activation<br />

by myelin oligodendrocyte glycoprotein is<br />

suppressed by adult but not cord blood CD25+ T cells.<br />

Eur J Immunol 2003; 33: 579–587.<br />

162. Miller A, Lider O, Abramsky O, Weiner HL. Orally<br />

administered myelin basic protein in neonates primes<br />

for immune responses and enhances experimental<br />

autoimmune encephalomyelitis in adult animals. Eur J<br />

Immunol 1994; 24: 1026–1032.<br />

163. Caramalho I, Lopes-Carvalho T, Ostler D et al.<br />

Regulatory T cells selectively express toll-like recptors<br />

and are activated by lipopolysaccharide. J Exp Med<br />

2003; 197: 403-411.<br />

164. Hauer AC, Rieder M, Griessl A et al. Cytokine production<br />

by cord blood mononuclear cells stimulated by<br />

cows milk protein in vitro: interleukin-4 and transforming<br />

growth factor β-secreting cells detected in the<br />

CD45RO T cell population in children of atopic<br />

mothers. Clin Exp Allergy 2003; 33: 615–623.<br />

165. Chung HL, Hwang JB, Park JJ, Kim SG. Expression of<br />

transforming growth factor β1, transforming growth<br />

factor type I and II receptors, and TNF-α in the mucosa<br />

of the small intestine in infants with food-protein<br />

induced enterocolitis syndrome. J Allergy Clin Immunol<br />

2002; 109: 150–154.<br />

166. Beyer K, Castro R, Birnbaum A et al. Human milkspecific<br />

mucosal lymphocytes of the gastrointestinal<br />

tract display a Th2 cytokine profile. J Allergy Clin<br />

Immunol 2002; 109: 707–713.<br />

167. Campbell DI, Murch SH, Lunn PG et al. Chronic T cellmediated<br />

enteropathy in rural West African children:<br />

Relationship with nutritional status and small bowel<br />

function. Pediatr Res 2003; 54: 306–311.<br />

168. Montgomery SM, Wakefield AJ, Morris DL et al. The<br />

initial care of newborn infants and subsequent hayfever.<br />

Allergy 2000; 55: 916–922.<br />

169. Björksten B, Sepp E, Julge K et al. Allergy development<br />

and the intestinal microflora during the first year of life.<br />

J Allergy Clin Immunol 2001; 108: 516–520.<br />

170. Heilig HG, Zoetendal EG, Vaughan EE et al. Molecular<br />

diversity of Lactobacillus spp. and other lactic acid<br />

bacteria in the human intestine as determined by<br />

specific amplification of 16S ribosomal DNA. Appl<br />

Environ Microbiol 2002; 68: 114–123.<br />

171. Zuany-Amorim C, Sawicka E, Manlius C et al.<br />

Suppression of airway eosinophilia by killed<br />

Mycobacterium vaccae-induced allergen-specific<br />

regulatory T-cells. Nature Med 2002; 8: 625–629.<br />

172. Nowak-Wegrzyn A. Future approaches to food allergy.<br />

Pediatrics 2003; 111: 1672–1680.<br />

173. Rabjohn P, Helm EM, Stanley JS et al. Molecular cloning<br />

and epitope analysis of the peanut allergen Ara h 3. J<br />

Clin Invest 1999; 103: 535–542.<br />

174. Leckie MJ, ten Brinke A, Khan J et al. Effects of an<br />

interleukin-5 blocking monoclonal antibody on<br />

eosinophils, airway hyper-responsiveness, and the late<br />

asthmatic response. Lancet 2000; 356: 2144–2148.<br />

175. Milgrom H, Fick RBJ, Su JQ et al. Treatment of allergic<br />

asthma with monoclonal anti-IgE antibody. rhuMAb-<br />

E25 Study Group. N Engl J Med 1999; 341: 1966–1973.<br />

176. BSACI Working Party: Lack G, Chapman M, Kalsheker<br />

N, King V, Robinson C, Venables K. Report on the<br />

potential allergenicity of genetically modified organisms<br />

and their products. Clin Exp Allergy 2002; 32:<br />

1131–1143.<br />

177. Nordlee JA, Taylor SL, Townsend JA et al. Identification<br />

of a Brazil-nut allergen in transgenic soybeans. N Engl J<br />

Med 1996; 334: 688–692.<br />

178. Taylor SL, Lehrer SB. Principles and characteristics of<br />

food allergens. Crit Rev Food Sci Nutr 1996; 36(S):<br />

91–118.<br />

179. Lack G. Clinical risk assessment of GM foods. Toxicol<br />

Lett 2002; 127: 337–340.<br />

180. Taylor SL, Hefle SL, Bindslev-Jensen C et al. Factors<br />

affecting the determination of threshold doses for<br />

allergenic foods: how much is too much? J Allergy Clin<br />

Immunol 2002; 109: 24–30.


23<br />

Introduction<br />

Crohn’s disease<br />

Qian Yuan and Harland S Winter<br />

New knowledge continues to impact on the pathogenesis<br />

and treatment of the chronic inflammatory<br />

intestinal disorder described by Dr Burrill Bernard<br />

Crohn in the early 1930s. 1 However, for over a<br />

century prior to this report physicians knew of the<br />

existence of a non-tuberculous ileitis. Characterized<br />

by transmural, chronic mucosal inflammation<br />

involving any portion of the gastrointestinal<br />

tract from the lips to the perineum, Crohn’s disease<br />

frequently involves the small intestine, especially<br />

the terminal ileum. Diarrhea, abdominal pain,<br />

malnutrition and growth delay are common<br />

features of disease that involves the small intestine.<br />

Rectal bleeding and extraintestinal manifestations<br />

are more commonly associated with colonic<br />

involvement. Although the clinical features of the<br />

disease are similar in adults and children, complications<br />

of chronic inflammation are most frequently<br />

seen in adults who by nature of their age,<br />

have had inflammation for a longer period of time.<br />

Growth retardation and delays in sexual maturation<br />

are issues faced primarily by children with<br />

Crohn’s disease.<br />

The diagnosis of Crohn’s disease remains a clinical–pathological<br />

diagnosis. Despite the availability<br />

of new serological tests, radiological and pathological<br />

features are needed to establish a diagnosis.<br />

Granulomata or chronic inflammation involving<br />

the small and large intestine will enable an experienced<br />

pathologist to establish the diagnosis with<br />

some certainty. However, if only colonic involvement<br />

is present, one may have difficulty distinguishing<br />

Crohn’s colitis from ulcerative colitis.<br />

The distribution of the inflammation, such as<br />

rectal sparing or predominantly right-sided<br />

disease, may favor Crohn’s over ulcerative colitis.<br />

Inflammatory bowel disease (IBD) is thought to<br />

result from inappropriate and ongoing activation<br />

of the mucosal immune system driven by the presence<br />

of normal luminal flora. 2 Despite many<br />

advances, the relationship between the environment<br />

and the immunogenetics of the host remains<br />

unclear. Crohn’s disease and ulcerative colitis are<br />

collectively referred to as inflammatory bowel<br />

diseases, and they are thought to be distinct entities<br />

with significant overlap in pathogenesis, clinical<br />

presentation and therapeutic management.<br />

Understanding the potential mechanism of<br />

mucosal immune dysregulation and developing<br />

effective therapies in children will not only<br />

improve our management of Crohn’s disease, but<br />

may also prevent the complications of chronic<br />

inflammation seen in adults with Crohn’s disease.<br />

This new knowledge will ultimately improve the<br />

life quality of all patients with Crohn’s disease.<br />

Epidemiology<br />

General incidence and prevalence<br />

A recent systematic review by Loftus et al 3<br />

analyzed the epidemiology and natural history of<br />

Crohn’s disease in population-based cohorts from<br />

North America. The prevalence rates of Crohn’s<br />

disease ranged from 26.0 to 198.5 cases per<br />

100000 persons. The incidence rates ranged from<br />

3.1 to 14.6 cases per 100000 person-years. Based<br />

on an estimate of 300 million people in North<br />

America, there are approximately 400000 to<br />

600000 patients living with Crohn’s disease and<br />

approximately 9000 to 44000 people are newly<br />

diagnosed in North America with Crohn’s disease<br />

each year. 3 There appears to be a slight female<br />

predominance, with the percentage of females<br />

with Crohn’s disease ranging from 48% to 66%. 3<br />

The prevalence and incidence of IBD is higher in<br />

North America and Northern Europe than in Asia,<br />

347


348<br />

Crohn’s disease<br />

Africa and southern Europe. 4–9 This geographic<br />

difference is also observed within individual<br />

countries including the USA. 10 Despite the low<br />

incidence and prevalence of IBD in Africa, 11<br />

similar rates for Crohn’s disease have been<br />

observed between African-American and Caucasian<br />

people. 12 Both Crohn’s disease and ulcerative<br />

colitis appear to have a higher incidence<br />

among the Jewish people. 13,14<br />

Incidence and prevalence in children with<br />

inflammatory bowel disease<br />

There is significantly less information about<br />

prevalence and incidence rates in children with<br />

Crohn’s disease. From a study in Sweden, 15 the<br />

incidence of Crohn’s disease in children appears to<br />

be increasing, from 2.4 per 100000 persons<br />

between 1990 and 1992 to 5.4 per 100000 persons<br />

between 1996 and 1998. Similar findings have also<br />

been observed by other investigators. 15–17 The<br />

peak incidence occurs between the ages of 15 and<br />

25 years, and about 25–30% of patients with<br />

Crohn’s disease develop the illness before the age<br />

of 20 years. 5 In a recent study performed in<br />

Wisconsin, 18 the incidence of Crohn’s disease was<br />

4.56 per 100000 children, more than twice the rate<br />

of ulcerative colitis (2.14 per 100000 children). An<br />

equal incidence occurred among all ethnic groups,<br />

and children from sparsely and densely populated<br />

counties were equally affected. 18 A report from the<br />

British Pediatric Surveillance Unit revealed an<br />

estimated incidence of IBD in the UK of 5.3 per<br />

100000 children under the age of 16, with Crohn’s<br />

disease being at least twice as common as ulcerative<br />

colitis. 19 The mean age at diagnosis was 11.8<br />

years (median 12.6 years) and 13% of cases<br />

occurred in children aged less than 10 years of<br />

age. 19 There was a median delay of 5 months from<br />

the onset of symptoms to a diagnosis; furthermore,<br />

25% of children experienced symptoms for more<br />

than 1 year prior to diagnosis. 19<br />

Etiology<br />

Genetic factors<br />

Recent studies in identifying susceptibility genes<br />

for human IBD have supported the hypothesis that<br />

genetic factors play an important role in the devel-<br />

opment of IBD. Several clinical observations with<br />

variations in incidence and prevalence among<br />

different populations, co-segregation of IBD in rare<br />

kindreds with various genetic disorders and familial<br />

aggregation of IBD have suggested that genetic<br />

factors contribute to an individual’s susceptibility<br />

to IBD. 2 First-degree relatives of an affected patient<br />

with IBD are 4–20 times more likely to develop the<br />

disease than are the background population. 20–23<br />

The absolute risk of IBD is approximately 7%<br />

among first-degree family members. 24 In addition,<br />

a family history of Crohn’s disease is associated<br />

with an earlier age of diagnosis in affected<br />

patient. 25–27 Observations in twin studies have<br />

strongly supported the role of genetic factors in the<br />

development of Crohn’s disease. 24,28 However, the<br />

absence of simple Mendelian inheritance suggests<br />

that multiple gene products contribute to a<br />

person’s risk of IBD. 2<br />

DNA linkage analyses have identified an area on<br />

chromosome 16, designated IBD1, with apparent<br />

linkage to Crohn’s disease but not ulcerative<br />

colitis. 29 Several other genomic loci have also been<br />

identified in patients with both Crohn’s disease<br />

and ulcerative colitis. 30–32 Detailed mapping analysis<br />

revealed that the NOD2 gene on chromosome<br />

16 was linked to Crohn’s disease. 33,34 The NOD2<br />

gene encodes a cytoplasmic protein expressed in<br />

macrophages known as CARD 15 (caspase activation<br />

and recruitment domain). CARD 15 is thought<br />

to function as a pattern-recognition receptor for<br />

bacterial lipopolysaccharide and to regulate<br />

nuclear factor-κB (NFκB) activation and<br />

macrophage apoptosis. 2 European and North<br />

American patients with Crohn’s disease, including<br />

those without a family history of IBD are more<br />

likely to have a variant of NOD2 than are persons<br />

without Crohn’s disease; 2 however, the number of<br />

individuals with Crohn’s disease who also have a<br />

variant of NOD2 remains very small. A more than<br />

20-fold increase in susceptibility to Crohn’s<br />

disease (ileal disease in particular) is observed in<br />

individuals homozygous for variant NOD2. 35–37 In<br />

children with Crohn’s disease, not only are<br />

NOD2/CARD15 variants associated with ileal<br />

disease, but they are also associated with lower<br />

weight percentiles at the time of diagnosis. 37<br />

Another putative locus on chromosome 5 has been<br />

identified in strong association with early-onset<br />

Crohn’s disease. 38,39 A list of putative genes identified<br />

for IBD is given in Table 23.1.


Table 23.1 Putative genes for inflammatory bowel disease (IBD)<br />

IBD Locus Chromosome Putative genes Reference<br />

CD<br />

CD, UC<br />

UC<br />

Environmental factors<br />

IBD1 16q12 NOD2 29<br />

IBD4 14q11 TCR α/δ complex 32, 164<br />

IBD5 5q31-33 IL-3, 4, 5, 13, and CSF-2 38<br />

IBD6 19p13 ICAM-1, C3, 38<br />

TBXA2R, LTB4H<br />

other 1p36 TNF-R family, CASP9 165<br />

other 7q MUC-3 30<br />

other 3p HGFR, EGFR, GNAI2 30<br />

IBD2 12q13 VDR, INF-γ 30<br />

CD, Crohn’s disease; UC, ulcerative colitis<br />

Despite the genetic association of Crohn’s disease,<br />

lack of total concordance among monozygotic<br />

twins indicates that additional factors may<br />

contribute to the pathogenesis of Crohn’s disease.<br />

Multiple environmental factors have been studied,<br />

but when critically evaluated, many do not<br />

provide consistent results. The evolving role of<br />

epigenetics may be significant in explaining some<br />

of these observations.<br />

Dietary component<br />

To date, no specific dietary component has been<br />

consistently identified as a cause for IBD. Diets<br />

high in refined sugar appear to be associated with<br />

the development of Crohn’s disease, 40 but not<br />

ulcerative colitis. 41 Decreased dietary consumption<br />

of fruits and vegetables may be associated<br />

with an increased risk of developing Crohn’s<br />

disease. 40 Increased dietary n-6 polyunsaturated<br />

fatty acids relative to n-3 polyunsaturated fatty<br />

acids also may be associated with Crohn’s<br />

disease. 42 Dietary components may alter intestinal<br />

flora, which then may modify the mucosal<br />

immune response in the susceptible host.<br />

Modified carbohydrate diets are used by many<br />

patients with Crohn’s disease and one should not<br />

disregard patients’ observations about the impact<br />

of specific foods on disease activity. As long as<br />

Etiology 349<br />

dietary restriction does not impact on nutritional<br />

well-being, patient observation will guide these<br />

therapies in the absence of randomized controlled<br />

studies.<br />

Breast feeding and perinatal exposures<br />

The data supporting the positive or negative association<br />

of breast feeding with IBD is not strong.<br />

Although studies on children with Crohn’s disease<br />

and their unaffected siblings suggest that children<br />

with Crohn’s disease are three to four times less<br />

likely to have been breast fed, 43 no apparent association<br />

existed between breast feeding and the<br />

development of ulcerative colitis. 44 Other data do<br />

not indicate an association between breast feeding<br />

and Crohn’s disease. 45 The negative association<br />

between breast feeding and Crohn’s disease may<br />

be related to early exposure to pathogens. Children<br />

with Crohn’s disease are three times more likely to<br />

have had a diarrheal illness in infancy. 43<br />

Furthermore, in a Swedish study, a four-fold<br />

increased risk of IBD was observed in patients who<br />

had a history of illness in early infancy. 46<br />

Cigarette smoking<br />

Cigarette smoking appears to have different effects<br />

on the development of Crohn’s disease and ulcerative<br />

colitis. Smoking doubles the risk for developing<br />

Crohn’s disease, 47,48 and cessation of smoking


350<br />

Crohn’s disease<br />

may decrease the risk of exacerbation in patients<br />

with established Crohn’s disease. 49 In contrast,<br />

smoking decreases the risk of developing ulcerative<br />

colitis. 48,50,51 Although current smokers have<br />

about a 40% lower risk of developing ulcerative<br />

colitis than non-smokers, former smokers are<br />

about 1.5 times more likely to develop ulcerative<br />

colitis than those individuals who have never<br />

smoked. 51<br />

Non-steroidal anti-inflammatory drugs<br />

Taking non-steroidal anti-inflammatory drugs<br />

(NSAIDs) has been associated with an increased<br />

risk for the development and exacerbation of<br />

IBD. 52–55 A retrospective study by Bonner et al<br />

failed to demonstrate an association between<br />

NSAID use and an increased risk for active IBD; 56<br />

however, there may be subsets of IBD patients who<br />

can tolerate NSAIDs with less likelihood of an<br />

exacerbation of disease. 57 Although the mechanisms<br />

of NSAID-associated gastric toxicity have<br />

been extensively studied, the mechanisms leading<br />

to intestinal damage are poorly understood, but<br />

may be related to damage of enterocyte mitochondria<br />

causing enhanced intestinal permeability, 58<br />

inhibition of cyclo-oxygenase (COX), 59 enterohepatic<br />

recirculation, 60 and formation of drug enterocyte<br />

adducts. 61,62<br />

Both COX-1 and COX-2 serve as constitutive and<br />

inducible enzymes in inflammation and cytoprotection.<br />

63 Although selective inhibition of COX-1<br />

or COX-2 is not ulcerogenic, the combined inhibition<br />

of COX-1 and COX-2 induced severe lesions in<br />

the stomach and small intestine in rats, suggesting<br />

an important role for COX-2 in the maintenance of<br />

gastrointestinal mucosal integrity. 64,65 While COX-<br />

2 selective inhibitors have been shown to cause<br />

less gastrointestinal injury than standard NSAIDs<br />

in healthy animals and humans, 66,67 their effect on<br />

pre-existing gastrointestinal inflammation is not<br />

completely clear. COX-1 expression is detected in<br />

both non-inflamed and inflamed gastrointestinal<br />

mucosa. In contrast, COX-2 is expressed in epithelial<br />

cells in the upper portions of the crypts as well<br />

as on the surface of colonic enterocytes in Crohn’s<br />

colitis and ulcerative colitis and in villous epithelial<br />

cells in Crohn’s ileitis. COX-2 is not detectable<br />

in the epithelium of the normal ileum or colon, 68,69<br />

but is up-regulated in the colonic mucosa in both<br />

experimental and human colitis 68 and appears to<br />

have a beneficial effect in healing experimental<br />

colitis. On the other hand, COX products are an<br />

important part of the inflammatory process and<br />

COX inhibition by agents such as mesalamine<br />

might be beneficial. 70<br />

Oral contraceptive drugs<br />

Data relating oral contraceptive drugs to the development<br />

of inflammatory bowel disease have been<br />

inconsistent. In two prospective studies, 47,71 oral<br />

contraceptive use has been associated with<br />

increased risk for developing both Crohn’s disease<br />

and ulcerative colitis. Women who currently use<br />

oral contraceptives are 2.5 times more likely to<br />

develop ulcerative colitis and 1.7 times more likely<br />

to develop Crohn’s disease. 47 Other case–control<br />

studies have not found a strong association<br />

between the use of oral contraceptives and the<br />

development of either Crohn’s disease72 or ulcerative<br />

colitis. 73<br />

Infectious agents<br />

Although available data do not convincingly<br />

incriminate a single, persistent pathogen as a<br />

universal cause of IBD, the role of infectious<br />

agents is still considered a strong possibility as a<br />

trigger for the development of IBD. Many infectious<br />

agents including Mycobacterium, Chlamydia,<br />

Listeria monocytogenes, cell-wall-deficient Pseudomonas<br />

species and reovirus have been proposed as<br />

the causative organism of Crohn’s disease.<br />

Paramyxovirus (measles virus) has been implicated<br />

etiologically in Crohn’s disease as a cause of<br />

granulomatous vasculitis and microinfarcts of the<br />

intestine. 74 However, the tissue evidence for<br />

persistent measles infection in patients with<br />

Crohn’s disease remains controversial. 75,76 One<br />

Swedish study demonstrated an increased incidence<br />

of Crohn’s disease but not ulcerative colitis<br />

among individuals born during measles<br />

epidemics; 77 however, two other studies from the<br />

UK failed to establish an association between the<br />

development of Crohn’s disease and birth during<br />

measles epidemics. 78,79 The relationship between<br />

measles vaccination and the development of<br />

Crohn’s disease has evoked even more acrimonious<br />

discussion. Although one initial study


eported that the relative risk for developing<br />

Crohn’s disease and ulcerative colitis was 3.0 and<br />

2.5, respectively, in children who received the<br />

measles vaccine, 80 subsequent studies failed to<br />

confirm these findings. 81,82 Mycobacterium paratuberculosis<br />

has been cultured from the bowel of<br />

patients with IBD, but definitive evidence to link a<br />

mycobacterium to Crohn’s disease is lacking. 83–85<br />

Commensal bacterial flora is an important etiological<br />

factor in IBD. Bacteria within the enteric<br />

lumen have a complex ecosystem that is in contact<br />

with the external environment. There are 10 12<br />

bacteria/g feces in the colon, of 400 different<br />

species, with anaerobes predominating. 86 The<br />

numbers of anaerobic bacteria and Lactobacillus<br />

are significantly decreased in patients with active,<br />

but not inactive, IBD. 87 Genetically altered animals<br />

that are susceptible to acquiring IBD do not<br />

express the phenotype when raised in a germ-free<br />

environment. 88,89 Furthermore, experimental<br />

colitis is attenuated when animals are treated with<br />

broad-spectrum antibiotics. 90 Further studies by<br />

Duchmann et al demonstrated that mucosal, but<br />

not peripheral blood mononuclear cells from<br />

patients with IBD proliferate when exposed to<br />

autologous intestinal bacteria. These data support<br />

the hypothesis that normal flora function as a<br />

modulator of ‘physiological inflammation’. 91,92<br />

Patients with IBD have increased numbers of<br />

surface-adherent and intracellular bacteria in the<br />

colonic epithelium, 93,94 and these commensal<br />

organisms may be playing a role in the development<br />

and maintenance of mucosal inflammation.<br />

Others factors<br />

Several studies have suggested that appendectomy<br />

may protect against the occurrence and severity of<br />

ulcerative colitis. 95–99 However, appendectomy<br />

does not affect the disease course of ulcerative<br />

colitis. 100 The relationship between appendectomy<br />

and the risk of developing Crohn’s disease is<br />

controversial. 99,100 In a large, retrospective cohort<br />

study, Andersson et al demonstrated that an<br />

increased risk of Crohn’s disease was found for<br />

more than 20 years after appendectomy. This<br />

increased risk was dependent on the patient’s<br />

gender, age and the diagnosis at operation (nonperforated<br />

vs. perforated appendicitis). 101 In T cell<br />

receptor (TCR)-α-deficient mice, appendectomy is<br />

protective against the development of colitis. 102<br />

Pathophysiology<br />

Pathophysiology 351<br />

Growing evidence suggests that inflammatory<br />

bowel disease is the result of a dysregulated<br />

immune response to common luminal flora. The<br />

intestinal inflammation of IBD may be viewed as<br />

an exaggeration of the ‘physiological’ inflammatory<br />

response always present in the normal lamina<br />

propria of the intestine and colon. 103 An intact<br />

mucosal barrier and regulatory mechanisms<br />

normally prevent the immune and inflammatory<br />

responses from causing tissue injury. 103 A defect in<br />

either mucosal barrier function, antigen processing<br />

or immunoregulation could result in a chronic<br />

inflammation, lymphocyte proliferation, cytokine<br />

release, neutrophil recruitment and tissue<br />

damage. 103 The factors that trigger activation of the<br />

immune system are unclear, but could be related<br />

to an intrinsic defect (either constitutive activation<br />

or the failure of down-regulatory mechanisms) or<br />

ongoing stimulation resulting from a change in the<br />

epithelial mucosal barrier. 2<br />

Data from studies on tissue samples from patients<br />

with IBD and experimental animal models of<br />

colitis have demonstrated unbalanced Th1 vs. Th2<br />

responses in the intestinal mucosa. 84,104,105 The<br />

mucosa of patients with established Crohn’s<br />

disease is dominated by CD4 + lymphocytes with a<br />

Th1 phenotype, characterized by the production of<br />

interferon-γ and interleukin (IL)-2; 2 whereas the<br />

mucosa in patients with ulcerative colitis may be<br />

dominated by CD4 + lymphocytes with an atypical<br />

Th2 phenotype, characterized by the production of<br />

transforming growth factor (TGF)-β and IL-5, but<br />

not IL-4. 2 CD4 + lymphocytes clearly play an<br />

important role in the pathogenesis of tissue<br />

damage in inflammatory bowel disease, particularly<br />

in Crohn’s disease. 84,106 Patients with disorders<br />

such as glycogen storage disease 1b, which<br />

are associated with abnormalities in neutrophil<br />

function may exhibit clinical manifestations of<br />

IBD. These observations suggest that the mechanisms<br />

leading to mucosal injury are not always<br />

lymphocyte dependent.<br />

Loss of tolerance towards the luminal commensal<br />

bacterial flora appears to be an important factor in<br />

the pathophysiology of IBD. Concomitant decrease<br />

of production of TGF-β and IL-10 by regulatory T<br />

lymphocytes may be partially responsible for the<br />

loss of tolerance to luminal flora and activation of


352<br />

Crohn’s disease<br />

Th1 cells. 107–109 Results from murine studies have<br />

shown that Th1 cytokines activate macrophages,<br />

which in turn, produce IL-12, IL-18 and macrophage<br />

migration inhibitor factor, thereby stimulating<br />

a Th1 response in a self-sustaining cycle. 110,111<br />

Activated macrophages produce a variety of potent<br />

proinflammatory cytokines including tumor<br />

necrosis factor (TNF)-α, IL-1 and IL-6. TNF-α has a<br />

variety of biological effects, including:<br />

macrophage activation and induction of protease,<br />

both of which can play a role in tissue destruction;<br />

112 up-regulation of adhesion molecule<br />

expression, facilitating recruitment of monocytes,<br />

lymphocytes and granulocytes; 113 enhancement of<br />

chloride secretion from intestinal epithelial<br />

cells; 114 increase of epithelial cell permeability; 115<br />

and enhancement of production of acute-phase<br />

reactants. 112 The inflammatory process and tissue<br />

destruction of the intestine are further promoted<br />

by generation of various proinflammatory mediators<br />

including other cytokines, chemokines,<br />

growth factors, arachidonic acid metabolites<br />

(prostaglandins and leukotrienes) and reactive<br />

oxygen metabolites such as nitric oxide. 2<br />

Activation of this destructive process results in the<br />

mucosal injury and clinical manifestations of IBD.<br />

Clinical signs and symptoms<br />

The initial clinical presentation of Crohn’s disease<br />

may be subtle, variable, non-specific and easily<br />

overlooked. Common symptoms include abdominal<br />

pain, diarrhea and/or weight loss. 103 A plateau<br />

in linear growth, delayed pubertal development,<br />

perianal lesions, fever, pallor, hematochezia and<br />

digital clubbing may also be present. 103 Crohn’s<br />

disease may affect any area of the gastrointestinal<br />

tract from the lips to the perianal area. In a study<br />

on Scottish children and adolescents with Crohn’s<br />

disease, 116 approximately 30% of patients had<br />

disease limited to the terminal ileum, 20% of<br />

patients had exclusive colonic involvement and<br />

50% of patients had both ileal and colonic involvement.<br />

In a more recent population-based study in<br />

Wisconsin, at the time of diagnosis isolated ileal<br />

disease was identified in 25% and ileocolonic<br />

involvement was found in 29%. About one-third of<br />

children had colonic involvement and 14% had<br />

disease in the upper gastrointestinal tract. 18 The<br />

inflammation associated with ulcerative colitis is<br />

limited to the colon, with the exception of mild<br />

inflammation in the ileum, termed ‘backwash<br />

ileitis’.<br />

The constellation of abdominal pain, diarrhea,<br />

poor appetite and weight loss represents the<br />

classic presentation of Crohn’s disease in children<br />

of any age group. 103 Frequently, the onset of pain is<br />

insidious and intermittent. Families and care<br />

providers often dismiss the symptoms as an acute<br />

illness, or a condition related to anxiety. In some<br />

individuals, anorexia may be the primary manifestation<br />

prompting an evaluation for anorexia<br />

nervosa or an eating disorder. Laboratory studies<br />

may suggest chronic inflammation, but up to 20%<br />

of children with IBD have a normal erythrocyte<br />

sedimentation rate. A summary of the prevalence<br />

of various clinical presentations from two separate<br />

studies is shown in Table 23.2. Abdominal pain is<br />

the most common single symptom at presentation,<br />

and may be periumbilical or localized to the right<br />

lower quadrant or to the lower abdomen. Grossly<br />

bloody diarrhea or extraintestinal manifestations<br />

signify colonic involvement. When diarrhea is<br />

predominant without visible blood or the patient<br />

has clubbing, one should suspect small-bowel<br />

involvement. Uncontrolled gastrointestinal hemorrhage<br />

is rare in Crohn’s disease, but may occur<br />

with an ileal ulcer.<br />

Clinical case 1<br />

A 6-year-old boy developed swollen lips<br />

(Figure 23.1) and painful ulcers in his mouth.<br />

He denied any history of abdominal pain, but<br />

his weight was decreased. An upper gastrointestinal<br />

series with small-bowel followthrough<br />

was normal. He had a history of anal<br />

fissures in the past and because of hard stools,<br />

he had been treated with stool softeners. An<br />

upper endoscopy and colonoscopy did not<br />

reveal evidence of inflammation, but a biopsy<br />

of the buccal mucosa demonstrated granulomatous<br />

cheilitis. He was treated with oral<br />

prednisone and metronidazole, with an initial<br />

improvement. He developed hepatitis while<br />

taking 6-mercaptopurine. Three infusions of<br />

infliximab were not effective. His perianal<br />

disease improved with a course of 20 hyperbaric<br />

oxygen sessions. Nutritional therapy<br />

with nightly nasogastric tube feedings for 2


Figure 23.1 Swollen lips in a child with Crohn’s disease<br />

involving the oral mucosa.<br />

years has contributed to his maintaining his<br />

weight and growth, but he has been unable to<br />

tolerate sufficient caloric intake to have catchup<br />

growth. Five years after his initial presentation,<br />

he has developed chronic inflammatory<br />

changes in the colon.<br />

Points of interest<br />

Clinical signs and symptoms 353<br />

(1) Crohn’s disease may present in any region of<br />

the gastrointestinal tract;<br />

(2) One-third of patients with Crohn’s disease do<br />

not respond to infliximab;<br />

(3) Nutritional therapy is important adjunctive<br />

therapy.<br />

Impaired linear growth and concomitant delay in<br />

sexual maturation are important clinical manifestations<br />

in children with Crohn’s disease, that may<br />

precede the onset of intestinal symptoms by 12–18<br />

months. 103 In a prospective study of children with<br />

IBD, 117 30% of children with Crohn’s disease had<br />

growth delay, which was defined as a fall in the<br />

height centile of more than 0.3 standard deviation<br />

per year, a growth velocity of less than 5cm per<br />

year, or a decrease in the growth velocity of ≥ 2cm<br />

compared with the preceeding year. About 50% of<br />

children had evidence of decreased height velocity<br />

prior to the diagnosis of Crohn’s disease. 118<br />

Delayed linear growth velocity and sexual maturation<br />

may be related to a chronically insufficient<br />

dietary intake and/or an increased energy requirements<br />

related to chronic inflammation or<br />

fever. 118,119 Anorexia occurs in 30% of children<br />

with Crohn’s disease and may be related to upper<br />

gastrointestinal tract involvement. 120 Studies in a<br />

rat colitis model demonstrated that TNF-α inhibited<br />

maturation of growth plate chondrocytes, 121<br />

suggesting that some proinflammatory cytokines<br />

Table 23.2 Prevalence of clinical presentations in Crohn’s disease from two studies (references 103<br />

and 166)<br />

Clinical symptoms Percentage (n=386) 103 Percentage (n=40) 166<br />

Abdominal pain 86 95<br />

Weight loss 80 80<br />

Diarrhea 78 77<br />

Blood in the stool 49 60<br />

Perianal lesions 44 *<br />

Fevers 38 *<br />

Growth failure * 30<br />

Mouth ulcers 28 *<br />

Arthralgia/arthritis 17 *<br />

Skin lesions 8 *<br />

*Prevalence was not specified


354<br />

Crohn’s disease<br />

may be a factor in the growth delay observed in<br />

children with Crohn’s disease.<br />

Anemia is present in 25–85% of patients with<br />

Crohn’s disease, 122 many of whom have anemia of<br />

chronic disease. Iron deficiency anemia is microcytic<br />

and may result from gastrointestinal blood<br />

loss, malabsorption of iron in the duodenum and<br />

jejunum because of inflammation and/or a lack of<br />

adequate oral intake. Folate deficiency causes a<br />

megaloblastic anemia, but frequently folate deficiency<br />

is associated with iron deficiency as well,<br />

and the indices may not be macrocytic. Folate<br />

deficiency is most commonly due to nutritional<br />

causes, but patients who take sulfasalazine are at<br />

increased risk for developing folate deficiency. For<br />

that reason, sulfasalazine is always prescribed<br />

with folate supplementation. Vitamin B12 is<br />

absorbed in the ileum; deficiencies are usually<br />

related to lack of absorption, related to inflammatory<br />

changes, fibrosis or resection. Because many<br />

years may be required to deplete vitamin B12 stores<br />

in an individual who has a normal reserve, the<br />

development of deficiency is often insidious.<br />

Children with ileal disease should be monitored<br />

for vitamin B12 deficiency by measuring serum<br />

levels. Clinical manifestations of the disease –<br />

anemia, dermatitis, cheilitis, decreased serum<br />

transaminases, peripheral neuritis, irritability and<br />

posterior column signs – do not develop for many<br />

months.<br />

Clinical case 2<br />

A 13-year-old girl was referred for evaluation of<br />

lower abdominal pain, diarrhea and a perirectal<br />

abscess. She had had intermittent abdominal<br />

pain from infancy but about 1 year prior to evaluation<br />

noted an increase in frequency and<br />

intensity. Her pediatrician recommended a<br />

lactose-restricted diet and the pain completely<br />

resolved. Two months prior to her referral to a<br />

pediatric gastroenterologist, she developed a<br />

perirectal abscess that was drained by a<br />

surgeon. The abscess healed slowly. She<br />

reported having 4–5 loose stools daily, but<br />

never noticed any blood. Her appetite was good<br />

and her rate of growth was not changed. There<br />

was no family history of IBD but her mother has<br />

Sjögren’s syndrome, a maternal second cousin<br />

has juvenile dermatomyositis and her maternal<br />

grandfather has rheumatoid arthritis. Her physical<br />

examination was normal with the exception<br />

of a well-healed surgical incision about<br />

4cm from the anus. Her weight was 25% for<br />

age; her height was 50% for age. A complete<br />

blood count (CBC), erythrocyte sedimentation<br />

rate (ESR) and liver function tests were normal.<br />

An upper gastro-intestinal radiograph with<br />

small-bowel follow-through was read as<br />

normal. Over the next 10 weeks she continued<br />

to experience intermittent crampy abdominal<br />

pain and occasional loose stools. She noted<br />

some blood on the paper after wiping and the<br />

perianal abscess would drain intermittently.<br />

However, over this period of time she continued<br />

to gain weight. An ileocolonoscopy<br />

revealed a normal-appearing ileum and right<br />

colon with multiple aphthoid lesions in the left<br />

colon and erythema in the rectum. Biopsies<br />

identified an active and chronic ileitis with<br />

granulomas but no evidence for ulceration, as<br />

well as microscopic changes of chronic and<br />

active inflammatory changes in the left colon<br />

with erosions.<br />

She was started on mesalamine and metronidazole.<br />

She began to feel better after 1 week of<br />

therapy, but then had recurrence of crampy<br />

abdominal pain with nausea and increased<br />

frequency of stooling up to seven loose bowel<br />

movements daily. Two weeks after starting<br />

therapy, prednisone 20mg twice a day was<br />

prescribed. Metronidazole was discontinued<br />

because of dysesthesia in her hands. After 3<br />

weeks of taking prednisone, she was quite<br />

Cushingoid, but she was passing three formed<br />

bowel movements daily and the perianal<br />

abscess was not draining. She began to taper<br />

the dose of prednisone by 5mg/day every week.<br />

When she reached 30mg of prednisone daily,<br />

she began to have increased frequency of stooling<br />

and crampy abdominal pain. Because of<br />

concerns for steroid dependence, she was<br />

started on 50mg daily of 6-mercaptopurine<br />

after it was checked that her thiopurine methyltransferase<br />

(TPMT) activity was in the normal<br />

range. A CBC and differential, liver function<br />

tests and amylase were checked weekly for 3<br />

weeks and then monthly. She tolerated the 6mercaptopurine<br />

well and was able to discontinue<br />

prednisone after 5 months. She was maintained<br />

on mesalamine and 6-mercaptopurine.


After 18 months of therapy, she began to<br />

develop intermittent abdominal pain and diarrhea.<br />

Her symptoms increased despite therapy<br />

with metronidazole and ciprofloxacin. She<br />

began to lose weight. Infliximab was started<br />

and 1 week after the initial infusion, she began<br />

to improve. She completed a second infusion 2<br />

weeks after the first and a third infusion 6<br />

weeks after the second infusion. She is now<br />

maintained on 6-mercaptopurine and an infliximab<br />

infusion every 8 weeks and remains<br />

without gastrointestinal symptoms.<br />

Points of interest<br />

(1) The onset of Crohn’s disease is often insidious;<br />

(2) Radiographic studies in the early stages of<br />

Crohn’s disease may appear normal;<br />

(3) Mucosal biopsy of normal-appearing endoscopic<br />

areas may provide evidence for inflammatory<br />

bowel disease;<br />

(4) Mesalamine with or without antibiotics may<br />

be effective therapy in some children with<br />

new-onset mild-to-moderate Crohn’s disease,<br />

but therapy should be changed if there is not<br />

a response within 2–4 weeks;<br />

(5) The inability to tolerate a taper of corticosteroids<br />

is an indication to begin 6-mercaptopurine;<br />

(6) Therapy with 6-mercaptopurine may take at<br />

least 3 months to become effective;<br />

(7) When 6-mercaptopurine fails to control<br />

symptoms of Crohn’s disease, infliximab<br />

should be considered. About two-thirds of<br />

adults will have an initial response to infliximab.<br />

Perianal lesions may be the first presenting feature<br />

of Crohn’s disease. Perianal disease associated<br />

with Crohn’s disease is frequently mild with small<br />

perianal skin tags or anal fissures, but more severe<br />

problems such as perianal fistulae and abscesses<br />

may develop. Fortunately, less than 5% of children<br />

with Crohn’s disease will develop a highly destructive<br />

form of perianal disease with recurrent<br />

abscesses and fistulae (Figure 23.2) involving the<br />

genitalia, and often resulting in rectal strictures.<br />

123,124 As illustrated in Case 2, the patient<br />

Clinical signs and symptoms 355<br />

was thought to have a perirectal abscess, but after<br />

surgical intervention, the area did not heal. With<br />

the exception of the first 2 years of life, children<br />

with persistent perianal abscess, fissure or fistula<br />

that does not respond to topical treatment and<br />

antibiotics should be evaluated for Crohn’s disease<br />

prior to the initiation of surgical therapy. Approximately<br />

one-third of children with Crohn’s disease<br />

can have significant perianal abnormalities during<br />

the course of their disease. 123 The most common<br />

perianal problem is skin tags which may become<br />

quite large. Some may become inflamed and<br />

painful, but for many adolescents they become a<br />

source of embarrassment. Perianal inflammatory<br />

disease may respond to antibiotics that have good<br />

anaerobic coverage, such as metronidazole, but<br />

intravenous administration is frequently required<br />

to achieve maximal healing. Non-inflamed skin<br />

Figure 23.2 Perianal fistula in a child with Crohn’s<br />

disease. Arrow marks the opening of the fistula. The<br />

erythema is commonly seen because of the excoriation to<br />

the skin caused by the drainage onto the perineum.


356<br />

Crohn’s disease<br />

tags usually do not resolve with medical treatment,<br />

and the use of topical tacrolimus or corticosteroids<br />

has not proved to be beneficial in reducing the size<br />

of the skin tags. Immunomodulators or corticosteroids<br />

may be necessary to control mucosal<br />

inflammation which may be a contributing factor<br />

to the growth of perianal skin tags.<br />

Extraintestinal manifestations are usually associated<br />

with colonic involvement of Crohn’s disease,<br />

and may precede intestinal symptoms by many<br />

months. The commonly involved organs are joints,<br />

skin, liver, eye and bone; the manifestations are<br />

listed in Table 23.3. 125,126 At least one extraintestinal<br />

manifestation is present in about 25–35% of<br />

adults with IBD. 125<br />

Complications<br />

Malnutrition<br />

Weight loss and malnutrition are the most prevalent<br />

nutritional disturbances in patients with<br />

IDB. 103 Approximately 85% of children with<br />

Crohn’s disease have a history of weight loss at<br />

initial diagnosis. 127 Malnutrition is mainly due to<br />

decreased intake caused by either primary<br />

anorexia from proinflammatory cytokines, or<br />

intestinal inflammation. An active inflammatory<br />

process, especially associated with fever, may<br />

further increase the body’s caloric consumption.<br />

Table 23.3 Extraintestinal manifestations of Crohn’s disease<br />

Estimates suggest that an increase in 1ºC increases<br />

the metabolic rate by 7%. For this reason, children<br />

with Crohn’s disease who experience recurrent<br />

febrile episodes often need to receive additional<br />

calories.<br />

As the result of the location of chronic intestinal<br />

inflammation in Crohn’s disease, a variety of<br />

micronutrient deficiencies may occur. These deficiencies<br />

include water-soluble vitamins (mainly<br />

folic acid and vitamin B12), fat-soluble vitamin D,<br />

and minerals (iron, copper and zinc). 103 Deficiency<br />

of these micronutrients may be subclinical or associated<br />

with specific clinical signs and symptoms<br />

(Table 23.4). The following factors may create or<br />

potentiate specific nutrient deficiencies: inadequate<br />

intake, the inflammatory/immunological<br />

response, poor absorption, increased nutrient<br />

demand for tissue repair or concurrent usage of<br />

specific medications. The severity of nutrient deficiency<br />

is often associated with the duration of<br />

disease and the degree of Crohn’s disease activity.<br />

Hepatobiliary and pancreatic complications<br />

Bile salts are absorbed in the distal ileum via an<br />

enterohepatic circulation. Malabsorption of bile<br />

salts caused by ileal inflammation or resection<br />

results in increased colonic bile salts and diarrhea.<br />

Bile salt malabsorption also may predispose<br />

towards gallstone formation. Agents that bind bile<br />

acids may be beneficial in these clinical situations.<br />

Presentations Percentage Reference<br />

Joint<br />

arthritis/arthralgia 2*, 15 (adult data) 126<br />

Skin<br />

erythema nodosum 1*, 8–15 (adult data) 18, 125<br />

pyoderma gangrenosum 0.5*, 1.3 (adult data)<br />

Eye<br />

episcleritis 125, 126<br />

uveitis<br />

orbital myositis<br />

Hepatobiliary involvement 3* 18<br />

PSC


Table 23.4 Micronutrient deficiencies<br />

Complications 357<br />

Clinical<br />

Micronutrient Prevalence presentations Laboratory test RDA Reference<br />

Water-soluble vitamins<br />

Vitamin B12 20–60% macrocytic plasma vitamin B12 1.5–3µg for 168<br />

anemia, level children; 169<br />

neuropathy 3µg for adults 170<br />

Vitamin C * scurvy (ecchymoses, plasma vitamin C 40–45mg for 171<br />

gingival bleeding, level children 172<br />

petechiae, 60–125mg for 173<br />

hyperkeratosis,<br />

arthralgia, poor<br />

wound healing<br />

adults 174<br />

Folate 34–54% macrocytic anemia plasma folate level 200–400µg for 127<br />

children; 168<br />

400µg for adults 175<br />

Thiamine * beriberi-like erythrocyte thiamine 0.9–1.5mg for 122<br />

(vitamin B1) symptoms transketolase (ETKA), children; 169<br />

(neuropathy, or blood thiamine 1–1.4mg for<br />

cardiomyopathy, concentration, or adults<br />

encephalopathy, transketolase urinary<br />

gastrointestinal<br />

symptoms)<br />

thiamine excretion<br />

Nicotinic acid * pellagra ? plasma nicotinic 12–20mg for 122<br />

(dermatitis, acid level children and 169<br />

diarrhea and adults 176<br />

dementia) 177<br />

Riboflavin (B2) * photophobia, plasma riboflavin 0.4mg for infants; 122<br />

angular level 1.1–1.8 mg for 169<br />

stomatitis, children and adults 177<br />

dermatitis 178<br />

Pyridoxine * neuropathy, plasma pyrodoxal-5- 0.9–2mg for 122<br />

(vitamin B6) glossitis, phophate (PLP) level, children; 169<br />

dermatitis or erythrocyte<br />

transaminase activity,<br />

or urinary 4-pyridoxic<br />

acid excretion, or<br />

urinary excretion of<br />

xanthurenic acid<br />

2mg for adults 178<br />

Fat-soluble vitamins<br />

Vitamin A 11% xerophthalmia plasma retinol level 2500–4000IU 171<br />

(dry eyes and skin, for children; 172<br />

blindness) 4000–5000IU 179<br />

for adults 180<br />

181<br />

Vitamin D 75% osteomalacia, rickets plasma vitamin D level 400IU for 122<br />

children and adults 182<br />

Vitamin E * neuromuscular plasma vitamin E level 9–15IU for 171<br />

disorders, hemolysis children and 30IU 172<br />

and anemia for adults 173<br />

Vitamin K * bleeding diathesis 20–80µg for<br />

children; 65–80µg<br />

for adults<br />

168<br />

continued


358<br />

Crohn’s disease<br />

Table 23.4 continued Micronutrient deficiencies<br />

Clinical<br />

Micronutrient Prevalence presentations Laboratory test RDA Reference<br />

Minerals<br />

Calcium 13% osteopenia, rickets plasma calcium level 800–1200mg 168<br />

for children<br />

and adults<br />

173<br />

Copper * skin and hair plasma copper level 0.4–0.6mg for 183<br />

abnormalities infants; 1.5–3mg<br />

for children and<br />

adults<br />

Iron 39–81% microcytic anemia plasma iron level 10–18mg for 168<br />

children and adults 184<br />

Magnesium 14–33% myopathy plasma magnesium 200–400mg for 122<br />

level children; 168<br />

300–350mg for 169<br />

adults 185<br />

186<br />

187<br />

Selenium * cardiomyopathy, plasma selenium level 20-50 µg for 173<br />

encephalopathy, children; 188<br />

immune dysfunction 55–70 µg for 189<br />

adults 190<br />

191<br />

Zinc 40–50% growth retardation, plasma zinc level 10–15 µg 122<br />

alopecia, dysgeusia, for children; 168<br />

acrodermatitis, 15mg for adults 192<br />

impaired wound<br />

healing<br />

193<br />

* Not specified<br />

RDA, recommended daily allowance<br />

Pancreatitis can occur as a result of inflammation<br />

in the duodenum or by medications such as<br />

azathioprine, 6-mercaptopurine or 5-aminosalicylic<br />

acid. 103 In some patients, the cause of pancreatitis<br />

is unknown and may be recurrent. Pancreatic<br />

insufficiency and carbohydrate intolerance are<br />

rare in Crohn’s disease, but a trial of pancreatic<br />

enzyme supplementation may be beneficial in<br />

treating inflammation.<br />

Nephrolithiasis<br />

Patients with Crohn’s disease have an increased<br />

risk of developing calcium oxalate and uric acid<br />

renal stones as a result of chronic steatorrhea and<br />

diarrhea. Under normal circumstances, free<br />

dietary calcium binds to oxalate in the intestinal<br />

lumen and is eliminated in the stool. In patients<br />

with steatorrhea, increased luminal fatty acids<br />

competitively bind free dietary calcium and leave<br />

oxalate free to be absorbed. The increased oxalate<br />

absorption results in hyperoxaluria. Dehydration<br />

and metabolic acidosis potentiate the formation of<br />

uric acid stones.<br />

Thromboembolic events<br />

Venous and arterial thromboembolism can result<br />

from hypercoaguability caused by thrombocytosis,<br />

hyperfibrinogenemia, elevated factor V and factor<br />

VII, and depression of antithrombin III. 128,129 A<br />

retrospective study by Talbot et al in 7199 adult<br />

patients with either chronic ulcerative colitis or<br />

Crohn’s disease demonstrated that thrombo-


embolic complications developed in 92 (1.3%) of<br />

these patients. 130 An additional four patients had<br />

cutaneous vasculitis, and 17 had an arteritis-associated<br />

diagnosis. Of the thromboembolic complications,<br />

61 were deep vein thromboses or<br />

pulmonary emboli. 130 Peripheral arterial thrombosis,<br />

coronary thrombosis, and mesenteric and<br />

portal vein thrombosis were predominantly postsurgical<br />

complications, but 77% of peripheral<br />

venous thromboses occurred spontaneously. 130,131<br />

In isolated case reports, cerebrovascular accidents<br />

have occurred in children with Crohn’s disease<br />

with seizures being the primary presenting<br />

symptom. 132–134 Retinal vascular disease can also<br />

occur in patients with Crohn’s disease. 129,135,136<br />

Children with clinical evidence of a hypercoaguable<br />

state should be evaluated for underlying<br />

causes. Dehydration and indwelling catheters are<br />

the main risk factors that should be avoided.<br />

Metabolic bone disease<br />

Decreased bone density is an important and probably<br />

underdiagnosed complication in children with<br />

Crohn’s disease. Multiple factors, including inflammatory<br />

cytokines that inhibit osteoclast activity,<br />

vitamin D deficiency and chronic systemic steroid<br />

therapy, may contribute to osteopenia or osteoporosis.<br />

Therefore, in children with Crohn’s<br />

disease, it is crucial to assess bone density and<br />

Table 23.5 Bisphosphonate preparations<br />

Complications 359<br />

decrease the risk factors for osteoporosis such as<br />

lack of physical exercise, chronic steroid use, lack<br />

of hormonal therapy of chronic secondary amenorrhea<br />

in adolescent girls, and deficient calcium and<br />

vitamin D supplementation. 103 Bisphosphonates<br />

are synthetic analogs of inorganic pyrophosphate<br />

that inhibit bone resorption (Table 23.5). In a recent<br />

randomized study of 84 adult patients with Crohn’s<br />

disease and osteopenia/osteoporosis, the efficacy of<br />

ibandronate and sodium fluoride as adjuncts to<br />

calcium and vitamin D treatment was evaluated. 137<br />

The results showed that both ibandronate and<br />

sodium fluoride were effective, safe and well tolerated<br />

in inducing an increase in lumbar bone<br />

density. 137 Experience is very limited with bisphosphonate<br />

therapy in children who have metabolic<br />

bone disease associated with Crohn’s disease.<br />

Results from ongoing multicenter trials of bisphosphonate<br />

in children with Crohn’s disease will<br />

provide valuable information.<br />

Local intestinal complications<br />

Intestinal obstruction, severe hemorrhage, perforation,<br />

fistulae, intra-abdominal abscesses and toxic<br />

megacolon can occur as a result of chronic intestinal<br />

inflammation. Mucosal ulceration may result in<br />

bleeding and perforation depending on the site and<br />

size of the ulcer. Fibrosis, a complication of inflammation,<br />

may result in stricture formation that<br />

Preparations Potency within generation Potency across generation<br />

First generation low<br />

Etidronate<br />

Second generation high<br />

Pamidronate<br />

Clodronate<br />

Tiludronate low<br />

Third generation high<br />

Zolendronate<br />

Ibandronate<br />

Risedronate<br />

Olpadronate<br />

Alendronate<br />

Neridronate<br />

Incadronate low high


360<br />

Crohn’s disease<br />

predisposes the patient to fistula, abscess, obstruction<br />

and perforation. In contrast to fibrosis, which<br />

usually accompanies Crohn’s disease, perforation<br />

and toxic megacolon are unusual and most<br />

commonly associated with ulcerative colitis. These<br />

conditions may require surgical intervention.<br />

Diagnosis<br />

The list of conditions that should be considered in<br />

the differential diagnosis for Crohn’s disease is<br />

extensive, and is related to the various clinical<br />

presentations of Crohn’s disease (Table 23.6). A<br />

high index of clinical suspicion is crucial if the<br />

clinician is to make a diagnosis in the early phases<br />

of the illness. Delay in growth is often a clue that<br />

the presenting symptoms are not caused by an<br />

acute illness.<br />

Table 23.6 Differential diagnosis of Crohn’s disease<br />

Symptoms Differential diagnosis<br />

Clinical suspicion for Crohn’s disease<br />

It is important to rule out Crohn’s disease in any<br />

child with recurrent abdominal pain, chronic<br />

diarrhea, weight loss, blood in stools, growth delay,<br />

pubertal delay, unexplained anemia and perianal<br />

disease. Growth failure can be an important initial<br />

clue in suspecting Crohn’s disease, since approximately<br />

half the children with Crohn’s disease have<br />

a delay in height velocity prior to obvious intestinal<br />

manifestation. 118 For patients with abdominal pain<br />

or growth delay, accompanying anorexia, change in<br />

diet, diarrhea or extraintestinal manifestations<br />

should raise the possibility of Crohn’s disease. If<br />

fecal leukocytes are found in a patient with<br />

hematochezia, inflammatory/infectious causes<br />

should be considered. Although 20% of patients<br />

with Crohn’s disease may have a normal ESR, if it<br />

is abnormal, this non-specific marker of inflammation<br />

may have relevance.<br />

Constitutional<br />

recurrent fever, malaise, pallor collagen vascular disease, infection, malignancy, especially<br />

lymphoma<br />

Intestinal<br />

abdominal pain lactose intolerance, constipation, peptic ulcer disease, <strong>Helicobacter</strong><br />

<strong>pylori</strong>, irritable bowel syndrome or psychosocial stress<br />

diarrhea infectious colitis/enteritis (Salmonella, Shigella, Campylobacter,<br />

Yersinia); immunodeficiency (HIV, primary immune deficiency)<br />

heme-positive stool/hematochezia ulcerative colitis, infectious colitis including Shigella, Salmonella,<br />

Campylobacter, Escherichia coli O157:H7 and amebiasis,<br />

Clostridium difficile colitis, CMV colitis, vasculitis, Henoch–<br />

Schönlein purpura; juvenile polyp, Meckel’s diverticulum, fissure,<br />

hemorrhoid<br />

fever, acute severe abdominal pain appendicitis, diverticulitis, intestinal perforation<br />

perianal disease histiocytosis, immunodeficiency<br />

Abnormal liver profile viral hepatitis, toxin, cholelithiasis<br />

Abnormal pancreatic enzymes idopathic pancreatitis, familial pancreatitis, cystic fibrosis<br />

Growth failure celiac disease, cystic fibrosis, endocrinopathy (thyroid, adrenal,<br />

pituitary, especially growth hormone), anorexia nervosa,<br />

Pubertal delay anorexia nervosa; bulimia; chromosomal abnormality<br />

Arthritis juvenile rheumatoid arthritis, acute rheumatic fever<br />

CMV, cytomegalovirus


Findings on physical examination such as pallor<br />

and abdominal tenderness are most often not<br />

specific for Crohn’s disease, but abdominal mass,<br />

aphthoid oral ulcers, erythema nodosum,<br />

pyoderma gangrenosum, digital clubbing, arthritis,<br />

or perianal skin tags are highly suggestive of<br />

Crohn’s disease.<br />

Laboratory studies<br />

A number of laboratory tests can offer useful and<br />

supportive information in facilitating diagnosis,<br />

although they are generally non-specific. An initial<br />

screening evaluation with CBC with differential,<br />

platelet count, ESR, serum albumin, stool hemoccult<br />

test, and stool culture are often obtained.<br />

Serum iron studies and serum levels of vitamins A,<br />

E, B12 and folate can be helpful in assessing specific<br />

nutritional deficiencies.<br />

Serological markers<br />

Several serological markers have been reported to<br />

facilitate the diagnosis of IBD and in distinguishing<br />

between Crohn’s disease and ulcerative colitis.<br />

Although these tests may have benefit in some clinical<br />

situations, the diagnosis remains to be determined<br />

by clinical–pathological criteria. Atypical<br />

perinuclear antineutrophil cytoplasmic antibodies<br />

(atypical P-ANCA, i.e. not directed against<br />

myeloperoxidase) can be detected in 60–80% of<br />

adult patients with ulcerative colitis and 10–20% of<br />

adult patients with Crohn’s disease. 138–140 In children<br />

with IBD, a positive test for P-ANCA and a<br />

negative test for anti-Saccharomyces cerevisiae<br />

antibody (ASCA) are indicative of increased likelihood<br />

of ulcerative colitis rather than Crohn’s<br />

disease (sensitivity 57%, specificity 97%). 139<br />

Conversely, a negative P-ANCA test and a positive<br />

ASCA test in children with IBD are suggestive of<br />

Crohn’s disease (sensitivity 49%, specificity 97%).<br />

The relatively low sensitivities of these serological<br />

markers for establishing a diagnosis of Crohn’s<br />

disease and ulcerative colitis limit their widespread<br />

clinical utility. 103 Antibody against Escherichia coli<br />

outer membrane porin (anti-OmpC antibody) has<br />

been reported as a potential serological marker for<br />

Crohn’s disease. It is a curious finding that the<br />

markers that are associated with Crohn’s disease<br />

are related to a host response to luminal organisms<br />

Diagnosis 361<br />

(i.e. yeast and bacteria). As experience increases in<br />

the pediatric population with the relationship<br />

between phenotype and serological markers, they<br />

may become more clinically relevant.<br />

Radiographic studies<br />

Barium upper gastrointestinal series with smallbowel<br />

follow-through is a useful tool in diagnosing<br />

gastroduodenal and ileal involvement of Crohn’s<br />

disease (Figure 23.3). Radiographic features in<br />

patients with Crohn’s disease include narrowing of<br />

the lumen of the small intestine or colon with<br />

nodularity and ulceration, a ‘string’ sign when<br />

luminal narrowing becomes more advanced or<br />

with severe spasm, a cobblestone appearance, fistulae<br />

and abscess formation, and separation of bowel<br />

loops, a manifestation reflecting the transmural<br />

inflammation and bowel wall thickening. Antral<br />

narrowing and segmental structuring of the duodenum<br />

can be seen with gastroduodenal Crohn’s<br />

disease.<br />

Air-contrast barium enema may be helpful in<br />

detecting colonic lesions such as ulcers, strictures<br />

and fistulae, but these colonic radiographs have<br />

been largely replaced by colonoscopy. In specific<br />

clinical situations, such as a distal stricture that<br />

does not permit visual inspection of the more proximal<br />

colon, virtual colonoscopy may be of benefit.<br />

Inflammation limited to the colon may make it<br />

difficult to distinguish ulcerative colitis from<br />

Crohn’s colitis; however, radiographic disease in<br />

the small intestine, stomach or esophagus strongly<br />

supports a diagnosis of Crohn’s disease.<br />

Computerized tomography (CT) scans have become<br />

extremely valuable in the assessment of the child<br />

with established Crohn’s disease who presents with<br />

abdominal pain or fever. The use of oral, rectal and<br />

or intravenous contrast increases the likelihood of<br />

finding a fistula, stricture or abscess. For the child<br />

with perianal disease, a careful CT scan with<br />

narrow cuts may identify a small perirectal abscess.<br />

Scintigraphy<br />

Scintigraphy with the 99mTc hexamethyl, propylene<br />

amino oxime (HMPAO)-labeled leukocyte scan<br />

(99mTc white blood cell (WBC) scan) has been


362<br />

Crohn’s disease<br />

(a) (b)<br />

(c)<br />

(d)<br />

Figure 23.3 (a) Axial computerized tomography (CT)<br />

images with intravenous and oral contrast revealing<br />

abnormal mucosal thickening of the cecum with<br />

abnormal enhancement. Numerous enlarged mesenteric<br />

lymph nodes are present (arrow). (b) This 15-year-old<br />

child with Crohn’s disease developed a large abscess in<br />

the anterior abdominal wall (arrow).(c) Markedly<br />

narrowed terminal ileum with ulcerations seen on a<br />

barium study (arrowhead). The same small bowel loop is<br />

seen on a coronal reformatted CT image (white arrow).<br />

(d) Small fistula tracks are seen extending from a<br />

thickened, inflamed terminal ileum to the ascending<br />

colon (arrow). (Courtesy of Dr Sudha Anupindi).


used as an alternative, non-invasive diagnostic test<br />

to determine the extent and distribution of inflammation<br />

in children with IBD. 142–144 In one study, 141<br />

the result of the 99mTc WBC scan correlated with<br />

histological findings on endoscopic and colonoscopic<br />

biopsies in 128 of 137 children. The sensitivity<br />

and specificity were 90% and 97%, respectively.<br />

141 Nevertheless, the value of this study for<br />

diagnosis is limited.<br />

Endoscopic studies and histologic features<br />

Compared with the continuous distribution of<br />

ulcerative colitis, Crohn’s disease is characteristically<br />

segmental, with areas of sparing<br />

throughout the intestinal tract; the terminal ileum<br />

is the most commonly affected site. 103 Data from a<br />

study in 389 children and adolescents with Crohn’s<br />

disease 103 revealed that 29% of patients had<br />

involvement of the terminal ileum with or without<br />

cecal disease, 9% had more isolated proximal (ileal<br />

or jejunal) disease, 42% had ileocolonic inflammation<br />

and 20% had only colonic involvement.<br />

Endoscopic examination is an important diagnostic<br />

tool for Crohn’s disease and colonoscopy is the<br />

most effective test to determine whether the colon<br />

is affected. Crohn’s disease often spares the<br />

rectosigmoid region, but this pattern of involvement<br />

may also be seen in early ulcerative colitis,<br />

especially in young patients. The colonoscopic<br />

features of Crohn’s disease range from subtle focal<br />

aphthoid ulcerations adjacent to areas of normal<br />

appearing mucosa (Figure 23.4) to diffuse areas of<br />

edema and ulceration that create a polypoid<br />

mucosa and give a cobblestone appearance (Figure<br />

23.5). Discontinuous colonic involvement with<br />

intervening normal appearing mucosa, often<br />

referred to as skip areas, is a key feature of Crohn’s<br />

disease. Deep linear ulcerations may evolve into<br />

mucosal bridges with relatively normal-appearing<br />

mucosa traversing ulcers (Figure 23.6). Nonspecific<br />

gastritis and duodenitis may be seen in<br />

patients with Crohn’s disease on esophagogastroduodenoscopy,<br />

but histological findings are often<br />

most helpful in establishing a diagnosis.<br />

Mucosa that appears grossly normal may reveal abnormalities<br />

on histological examination. Edema and<br />

an increase in mononuclear cell density in the<br />

lamina propria are relatively non-specific findings. 103<br />

Diagnosis 363<br />

Figure 23.4 Focal aphthous lesion in Crohn’s disease<br />

with surrounding area of erythema (arrow).<br />

Figure 23.5 Cobblestone appearance (arrow) and<br />

mucosal ulcerations with mucopurulent exudate of the<br />

colon in Crohn’s disease. (Courtesy of Dr Esther J. Israel).<br />

In the early phases of Crohn’s disease, microscopic<br />

changes may resemble an infectious colitis with<br />

infiltration of the crypts by polymorphonuclear<br />

leukocytes (cryptitis or crypt abscesses), and distortion<br />

of crypt architecture 103 (Figure 23.7). Focal


364<br />

Crohn’s disease<br />

Figure 23.6 Ulceration with mucosal bridging (arrows)<br />

caused by undermining ulcerations of the colon in<br />

Crohn’s disease. (Courtesy of Dr Esther J. Israel).<br />

(a)<br />

Figure 23.8 (a) Focal ileitis in Crohn’s disease (arrow)<br />

(low-power view). (b) Ileitis in Crohn’s disease (highpower<br />

view) demonstrating neutrophilic infiltrate into the<br />

mucosa (arrow). (Courtesy of Dr Gregory Lauwers).<br />

ileitis is characteristic of Crohn’s disease (Figure<br />

23.8). The presence of fibrosis and histiocytic<br />

proliferation in the submucosa suggests Crohn’s<br />

disease. The pathological hallmark of Crohn’s<br />

inflammation is focal inflammation or transmural<br />

extension involving all layers of the bowel wall.<br />

Non-necrotizing granulomas are seen in 60% of<br />

Figure 23.7 Active colitis in Crohn’s disease (arrow) with<br />

area of relatively preserved mucosa without mucin depletion.<br />

(Courtesy of Dr Gregory Lauwers).<br />

(b)<br />

surgical specimens and 20–40% of mucosal biopsies<br />

145,146 (Figure 23.9). Microscopic focal enhancing<br />

lesions in the stomach (Figure 23.10) were<br />

thought to be indicative of Crohn’s disease, but<br />

they can be seen in ulcerative colitis as well.<br />

However, they are supportive of a chronic inflammatory<br />

process.


Figure 23.9 Focal granuloma (arrow) in the colon of a<br />

child with Crohn’s disease. (Courtesy of Dr Gregory<br />

Lauwers).<br />

Treatment<br />

Management of children with Crohn’s disease<br />

includes medical therapy, potential surgical intervention<br />

as well as nutritional and psychiatric<br />

support. Education of the patient and the family is<br />

a critical aspect of care that should not be overlooked.<br />

The IBD Notebook that is available through<br />

the North American Society for Pediatric<br />

Gastroenterology, Hepatology and Nutrition is a<br />

valuable resource empowering patients to understand<br />

and control their disease. The general<br />

guiding principles for treatment are to control<br />

inflammation and achieve normal growth while<br />

maintaining a high quality of life. For patients with<br />

mild-to-moderate disease, initial therapy with 5aminosalicylic<br />

acid (5-ASA) and antibiotics may<br />

induce remission. For patients with more active<br />

disease, corticosteroids may provide a more rapid<br />

response. Initial treatment with 6-mercaptopurine<br />

seems to decrease steroid dependency, but most<br />

clinicians reserve immunomodulator therapy for<br />

patients who fail to maintain a remission.<br />

Nutrition support<br />

Nutritional therapy is a necessary and essential<br />

aspect of therapy for all children with Crohn’s<br />

disease; achieving normal growth potential is one<br />

of the major therapeutic challenges. Nutrition has<br />

been used as a primary therapy for children with<br />

Treatment 365<br />

Figure 23.10 Focal enhancing lesions of the stomach in<br />

a patient with Crohn’s disease. (Courtesy of Dr Gregory<br />

Lauwers).<br />

Crohn’s disease, but when compared to corticosteroids,<br />

children treated with nutritional therapy<br />

relapsed sooner. Although the impact on linear<br />

growth was less in the nutrition group, compliance<br />

with nutritional regimens is more difficult.<br />

Suboptimal intake related to anorexia or fear of<br />

symptoms with eating, stool losses of nutrients<br />

caused by inflammation, ulceration or resection,<br />

increased nutritional requirements secondary to<br />

fever or increased metabolic requirements, treatment<br />

with corticosteroids that inhibit insulin-like<br />

growth factor-1, or circulating cytokines that delay<br />

linear growth are all relevant factors that impair<br />

growth. Medical or surgical therapy may correct or<br />

in some situations exacerbate some of the causes of<br />

undernutrition, but finding a tolerable nutritional<br />

supplement is the key to successful nutritional<br />

therapy.<br />

Careful plotting of weight, height and rate of<br />

growth, and calculation of the body mass index<br />

(BMI) (weight in kg/(height in m) 2 ) is a critical part<br />

of the evaluation that enables one to assess when<br />

growth delay began. Anthropometric measurements<br />

such as triceps skin-fold thickness and midarm<br />

circumference provide an estimation of body<br />

fat and muscle mass. CT scans and magnetic resonance<br />

imaging (MRI) may also be beneficial in<br />

assessing body composition. Bioelectric impedance<br />

analysis (BIA) and total body electrical<br />

conductance (TOBEC) assess total body water and<br />

fat mass, but have not been validated in the


366<br />

Crohn’s disease<br />

pediatric IBD population. Dual energy X-ray<br />

absorptiometry (DEXA) scanning of the spine is a<br />

valuable tool to assess bone mineralization.<br />

Daily energy requirements are increased in children<br />

with Crohn’s disease, and during a flare of<br />

disease energy requirements increase while energy<br />

generation decreases. Catch-up growth is possible,<br />

but may require daily dietary energy intakes over<br />

130% of the recommended values for ideal body<br />

weight. In practical terms, this corresponds to<br />

intakes ranging between 60 and 75kcal/kg per day.<br />

Daily protein intake should approximate 3g/kg,<br />

but the requirements of specific proteins such as<br />

glutamine is not substantiated. Amino acid-based,<br />

hydrolyzed or intact protein diets appear to be<br />

equally beneficial although there is a theoretical<br />

immunological benefit to giving protein that is less<br />

antigenic. In practice this does not seem to be<br />

beneficial for most patients. Total amount of<br />

protein rather than composition appears most relevant.<br />

Growing children with Crohn’s disease who are<br />

treated with corticosteroids stimulate osteoclastic<br />

bone resorption, inhibit osteoblast proliferation,<br />

decrease calcitriol synthesis and do not absorb<br />

calcium efficiently. These processes result in<br />

decreased bone mineralization. Patients who are at<br />

greatest risk for decreased bone mineralization<br />

seem to be those who take more than 7.5mg/day,<br />

have a greater than 5g lifetime cumulative dose or<br />

more than 1 year lifetime exposure. For a growing<br />

adolescent 1300mg/day of calcium is recommended<br />

along with 400IU/day of vitamin D.<br />

Other minerals and vitamins should be replaced if<br />

deficient. Zinc is decreased with inflammation<br />

and serum zinc may not reflect intracellular stores.<br />

Many patients with Crohn’s disease take zinc along<br />

with selenium and vitamin E as an antioxidant<br />

cocktail without strong supportive evidence for<br />

efficacy. Folate requirements may increase in<br />

active Crohn’s disease, but the folic acid requirement<br />

for children with Crohn’s disease is not<br />

known. Many clinicians recommend a dose of<br />

1mg daily, but the dose is not based on sound<br />

evidence. Vitamin B12 deficiency may need to be<br />

treated with subcutaneous injections as ileal<br />

absorption is often impaired. Release in 2003 of a<br />

nasal form of vitamin B12 may permit children<br />

who require vitamin B12 to avoid painful injections.<br />

5-Aminosalicylic acid drugs<br />

The anti-inflammatory effects of this class of<br />

medication results from the inhibitory effect of 5-<br />

ASA on leukotriene biosynthesis. Effective delivery<br />

of these drugs is achieved by various structural<br />

modifications that permit the medication to reach<br />

the area of inflamed mucosa. Sulfasalazine, a<br />

prototype of this class of drug, consists of 5-ASA<br />

linked to sulfapyridine via an azo bond. The sulfa<br />

moiety serves as a carrier to facilitate delivery of<br />

therapeutically active 5-ASA to the colon, which is<br />

achieved by bacterial cleavage of the diazo bond<br />

and release of the 5-ASA in the intestinal lumen. A<br />

number of different 5-ASA preparations have been<br />

developed based on a similar strategy. A summary<br />

of the different chemical modifications of 5-ASA<br />

preparations, and their release mechanisms are<br />

listed in Table 23.7. The dosage and site of effect as<br />

well as side-effects are listed in Tables 23.8 and<br />

23.9, respectively. A profile of release within the<br />

gastrointestinal tract of the different 5-ASA preparations<br />

is shown in Figure 23.11. The medication<br />

should be selected that targets the areas of inflammation.<br />

The dose ranges between 30–60 mg/kg per<br />

day, but some clinicians use higher doses.<br />

Pediatric dosing should not exceed the adult<br />

recommended dose. Topical 5-ASA preparations<br />

(enema or suppository) are used more commonly<br />

in ulcerative colitis or in those patients with<br />

Crohn’s disease who do not have rectal sparing.<br />

Corticosteroids<br />

Systemic corticosteroid treatment remains an<br />

important and effective therapy for children with<br />

Crohn’s disease, but side-effects often limit their<br />

use. The anti-inflammatory effect of corticosteroids<br />

is related to inhibition of cell-mediated<br />

immunity, decreased cytokine production,<br />

decreased capillary permeability, impaired<br />

neutrophil and monocyte chemotaxis and stabilization<br />

of lysosomal membranes. 147,148 Prednisone<br />

or prednisolone is commonly used in children<br />

with Crohn’s disease who have active mucosal<br />

inflammation that does not respond to initial<br />

therapy with 5-ASA and antibiotics. Prednisone is<br />

a synthetic glucocorticoid of intermediate potency,<br />

and is converted to its active form, prednisolone,<br />

in the liver. 103 Prednisone or prednisolone is<br />

administrated orally or intravenously at 1–2 mg/kg


Table 23.7 Chemical modification and release mechanisms of 5-aminosalicylic acid (5-ASA)<br />

preparations<br />

Preparation Trade name Chemical modification Release mechanism<br />

Table 23.8 Dosage and site of effect of 5-aminosalicylic acid preparations<br />

Preparation Trade name Dosage Site of effect<br />

Sulfasalazine Azulfidine C: 40–60 mg/kg per day ÷ BID. colon<br />

For maintenance 30mg/kg per day ÷<br />

QID (max 2g/day)<br />

A: 3-4 g/day ÷ QID<br />

For maintenance 2g/day ÷ QID<br />

Olsalazine Dipentum C: not available colon<br />

A: 1.5–3g/day ÷ BID – QID<br />

Balsalazide Colazal C: not available colon<br />

A: 6.75g/day ÷ TID.<br />

For maintenance 3–4 g/day ÷ BID<br />

Treatment 367<br />

Sulfasalazine Azulfidine azo bond to sulfapyridine bacterial cleavage<br />

Olsalazine Dipentum 5-ASA attaches to 5-ASA bacterial cleavage<br />

Ipsalazide azo bond to inert carrier bacterial cleavage<br />

Balsalazide Colazal azo bond to inert carrier bacterial cleavage<br />

Mesalamine/mesalazine Asacol attach to acrylic-based resins pH-dependent release (pH >7)<br />

Mesalamine/mesalazine Salofalk, Claversal attach to acrylic-based resins pH-dependent release (pH >5.6)<br />

Mesalamine/mesalazine Pentasa coated with ethyl cellulose time and moisture-dependent<br />

release<br />

Mesalamine/mesalazine Rowasa, Canasa uncoated direct local effect (rectum to left<br />

enema/suppository colon)<br />

Mesalamine Asacol C: 25–50 mg/kg per day ÷ BID – TID distal ileum or right colon<br />

A: 2.4–4.8g/day ÷ TID – QID<br />

Mesalamine Salofalk, C: not available mid- to small bowel<br />

Claversal A: 2.4–4g/day ÷ TID – QID<br />

Mesalamine Pentasa C: 25–50mg/kg per day ÷ BID – TID small bowel and colon<br />

A: 4g/day ÷ TID – QID<br />

Mesalamine Rowasa enema, C: not available rectum and left colon<br />

enema/suppository Canasa suppository A: 4g enema nightly; 500mg<br />

suppository BID<br />

C, children; A, adults; BID, twice a day; QID, four times a day; TID, three times a day


368<br />

Crohn’s disease<br />

Table 23.9 Side-effects of 5-aminosalicylic acid preparations<br />

Preparation Side-effect<br />

Sulfasalazine (mainly related to sulfa moiety) dose-dependent (nausea, vomiting, headaches, hemolysis)<br />

hypersensitivity (fever, rashes, Stevens–Johnson syndrome,<br />

agranulocytosis, pulmonary fibrosis, hepatotoxicity)<br />

male fertility impairment<br />

folate deficiency<br />

Mesalamine/mesalazine acute pancreatitis<br />

Olsalazine diarrhea<br />

Figure 23.11 Gastrointestinal release profile of 5-aminosalicylic acid preparations. The<br />

diagram of gradient indicates increased release.<br />

per day to a maximum of 40–60 mg/day, and gradually<br />

tapered after establishment of remission. A<br />

typical adolescent patient who required corticosteroids<br />

might start on 40mg of prednisone once<br />

daily (or 2mg/kg per day, whichever was less). If<br />

the clinical response is suboptimal, the dose may<br />

be divided twice a day. Splitting the dose further<br />

has little proven efficacy and increases adrenal<br />

suppression. Although some clinicians may<br />

increase the dose to a maximum of 60 mg/day for<br />

those patients who do not respond, there is no<br />

evidence that higher doses are beneficial and they<br />

are associated with increased untoward events.<br />

Systemic corticosteroid therapy is generally not<br />

used as a maintenance regimen. The adverse effects<br />

of systemic corticosteroid therapy are summarized<br />

in Table 23.10. Topical intrarectal corticosteroids<br />

(hydrocortisone enemas or foam) are also available<br />

for those patients with rectal disease.<br />

Oral budesonide is designed for release in the<br />

ileum, cecum and right colon and is usually<br />

prescribed at 9mg/day initially for an adult for 8<br />

weeks and then tapered by 3-mg increments.<br />

Budesonide is not approved by the Food and Drug<br />

Administration (FDA) for use in children. Because<br />

of its high first-pass hepatic metabolism, budesonide<br />

has significant local effect with much less<br />

systemic effect. 149 The incidence of overall adverse<br />

effects is less than with prednisolone as noted in<br />

clinical trials with adult patients with Crohn’s<br />

disease. 150 However, long-term use of oral budesonide<br />

may still cause significant systemic glucocorticoid<br />

effects, since budesonide has an affinity<br />

for the glucocorticoid receptor 50–100 times more<br />

than that of prednisone. 2 Experience with pediatric<br />

patients with Crohn’s disease has demonstrated<br />

that growth failure remains an active issue<br />

in long-term treatment with budesonide, because


Table 23.10 Adverse effect of systemic corticosteroid therapy<br />

Common Rare Long term<br />

of either direct suppression of linear growth or an<br />

inability to control disease activity fully. 151<br />

Immunoregulatory agents<br />

Azathioprine and 6-mercaptopurine<br />

Growing evidence supports the use of the<br />

immunoregulatory agents azathioprine and 6mercaptopurine<br />

in treating Crohn’s disease as effective<br />

therapy to maintain control of intestinal<br />

inflammation. These agents are particularly useful<br />

in patients who are steroid-dependent. 6-<br />

Mercaptopurine is a purine analog capable of interfering<br />

with DNA and RNA synthesis by competing<br />

with endogenous purines. Azathio-purine is a prodrug<br />

that exerts its immunosuppressive effect<br />

through its metabolite 6-mercaptopurine. 6-<br />

Mercaptopurine undergoes extensive transformations<br />

to form cytotoxic intracellular metabolites<br />

responsible for the immunosuppressive effect.<br />

Studies suggest that the intracellular levels of the<br />

metabolite 6-thioguanine (6-TG) correlate with efficacy,<br />

and the intracellular levels of the metabolite<br />

6-methyl mercaptopurine (6-MMP) with hepatotoxicity.<br />

152 Genetic polymorphisms of thiopurine<br />

methyltransferase (TPMT), an enzyme that catabolizes<br />

6-mercaptopurine to 6-MMP, controls the<br />

baseline enzymatic activity of TPMT. One in 300<br />

subjects has very low enzyme activity or none at<br />

all, and 11% of the population has intermediate<br />

enzyme activity. Inherited low TPMT activity<br />

appears to be a risk factor for acute bone marrow<br />

failure by leaving more 6-mercaptopurine available<br />

for conversion to cytotoxic 6-TG.<br />

In a prospective, double-blind, placebo-controlled<br />

pediatric trial, 55 children (mean age of 13 years)<br />

Treatment 369<br />

Acne seizure growth failure<br />

Moon facies pseudotumor cerebri nephrolithiasis (hypercalciuria)<br />

Hirsutism psychosis osteopenia<br />

Cutaneous striae myopathy osteoporosis<br />

aseptic necrosis of femoral head diabetes<br />

cataracts<br />

increased intraocular pressure<br />

secondary hyperparathyroidism<br />

with newly diagnosed Crohn’s disease were<br />

randomly assigned to receive prednisone plus<br />

either 6-mercaptopurine (1.5mg/kg per day) or<br />

placebo. 153 Markowitz et al found that the duration<br />

of steroid use and the cumulative steroid dose was<br />

significantly decreased in the group that received<br />

steroids plus 6-mercaptopurine. Although the rate<br />

of remission achieved in both groups was similar<br />

(89%), relapse was less frequent in the group that<br />

received 6-mercaptopurine. 153 Base on these findings,<br />

6-mercaptopurine should be considered as<br />

part of the initial treatment prescribed for children<br />

with newly diagnosed moderate-to-severe Crohn’s<br />

disease activity who are started on corticosteroids.<br />

153 The dosages, adverse effects, and monitoring<br />

parameters for azathioprine and 6-mercaptopurine<br />

are listed in Table 23.11. The monitoring<br />

of 6-TG and 6-MMP remains controversial, as<br />

discussed by Dubinsky and Griffiths. 154<br />

Methotrexate<br />

Methotrexate exerts its cytotoxic effect by interfering<br />

with thymidine synthesis, by inhibiting the<br />

conversion of the folic acid to its active form<br />

tetrahydrofolate. Intramuscular administration of<br />

methotrexate weekly has proved effective in inducing<br />

and maintaining remission in adult patients<br />

with Crohn’s disease who are either steroid-refractory<br />

or -dependent, 155 or in pediatric patients with<br />

Crohn’s disease intolerant or refractory to azathioprine/6-mercaptopurine<br />

treatment. 156 Common<br />

adverse effects of methotrexate include nausea and<br />

abnormal liver serum transaminase levels.<br />

Concerns about possible hepatic fibrosis may limit<br />

the long-term use of methotrexate.


370<br />

Crohn’s disease<br />

Table 23.11 6-Mercaptopurine and azathioprine therapy<br />

Dosage<br />

Azathioprine 6-Mercaptopurine Potential adverse effects Monitoring program<br />

1.5–2.5mg/kg per day 1–2mg/kg per day fever TPMT level before therapy<br />

pancreatitis<br />

rash<br />

arthralgia weekly for 4 weeks, then<br />

nausea every 3months: CBC with<br />

vomiting differential, liver function<br />

diarrhea, leukopenia tests, amylase, lipase<br />

thrombocytopenia<br />

opportunistic infection<br />

hepatitis consider: monitoring 6-TG<br />

malignancy and 6-MMP<br />

TPMT, thiopurine methyltransferase; CBC, complete blood count; 6-TG, 6-thioguanine; 6-MMP, 6-methyl<br />

mercaptopurine<br />

Anti-tumor necrosis factor-α agents<br />

TNF-α is a pro-inflammatory cytokine produced by<br />

activated macrophages, and is thought to play an<br />

important role in the pathogenesis and clinical<br />

manifestations of IBD. Several reagents have been<br />

developed as antagonists of TNF-α in the treatment<br />

of Crohn’s disease. Infliximab (Remicade ® ) is a<br />

chimeric monoclonal antibody composed of a<br />

complement-fixing ‘human’ IgG1 constant region<br />

and a mouse-derived antigen-binding variable<br />

region, and binds directly to soluble TNF-α. 2<br />

Infliximab is efficacious as therapy in about twothirds<br />

of adult patients with moderately to<br />

severely active Crohn’s disease resistant to corticosteroid<br />

and azathioprine/6-mercaptopurine<br />

therapy. However, about half of those who respond<br />

initially will have a sustained response over the<br />

first year. Infliximab is also effective in patients<br />

with fistulae that are refractory to antibiotic and/or<br />

azathioprine/6-mercaptopurine treatment. 157–159<br />

Infliximab is administered intravenously with a<br />

recommended regimen for treatment of fistulae of<br />

three initial doses at 0, 2 and 6 weeks, followed by<br />

repeat infusions at 2-month intervals, for patients<br />

who demonstrate response to the initial doses. 160<br />

Although this regimen is not FDA approved, most<br />

patients with Crohn’s disease including those<br />

without fistulae are treated thus. Common sideeffects<br />

associated with infliximab therapy include<br />

fever, rash, vomiting and chest pain. 160 Rarely, a<br />

serious hypersensitivity reaction may also occur<br />

with repeat infusion. Reactivation of tuberculosis<br />

associated with infliximab therapy has been<br />

reported as a potential lethal complication. 160<br />

Therefore, all patients must receive tuberculosis<br />

screening prior to initiation of therapy. Lymphoma<br />

has developed in a small number of patients<br />

receiving infliximab, although a causal relationship<br />

has not been established. 2,160<br />

In a retrospective open-labeled pediatric study, 19<br />

children with Crohn’s disease (mean age 14.4<br />

years) received 1–3 infusions of infliximab<br />

(5mg/kg per dose) over a 12-week period for corticosteroid-resistant<br />

(n=7) or corticosteroid-dependent<br />

disease (n=12). 161 Significant initial<br />

improvement (first 4 weeks after infusion) was<br />

noted in all subjects, with pediatric Crohn’s<br />

disease activity index (PCDAI) values decreasing<br />

significantly (42.1±13.7 to 10.0±5.6, p


with Crohn’s disease (aged 8–17 years) were<br />

randomized to receive a single infusion of infliximab<br />

1mg/kg (n=6), 5mg/kg (n=7), or 10mg/kg<br />

(n=8). 162 Improvement in the PCDAI, ESR and Creactive<br />

protein (CRP) was observed with all infliximab<br />

doses, beginning at week 1. All treated<br />

patients experienced approximately 50% improvement<br />

in the PCDAI by week 2, and by week 12, the<br />

PCDAI remained approximately 30% improved<br />

from baseline. 162 During the study, all 21 patients<br />

(100%) achieved a clinical response, and ten<br />

patients (48%) achieved clinical remission; there<br />

were no infusion reactions in any of the treated<br />

patients. 162 Appropriately powered pediatric<br />

studies of infliximab are needed to establish safety<br />

and efficacy. 163<br />

Antibiotics<br />

Antibiotics are often used to treat newly diagnosed<br />

or relapsed patients with Crohn’s disease or<br />

patients with perianal fistulae/abscesses. There are<br />

no randomized clinical trials to support efficacy in<br />

the pediatric population. Commonly used antibiotics<br />

and their dosage and side-effects are listed<br />

in Table 23.12.<br />

Surgery<br />

The most common reasons for surgery are<br />

intractable symptoms despite medical therapy,<br />

obstruction, intra-abdominal abscess, fistula<br />

refractory to medical therapy, or intractable<br />

hemorrhage. In patients with a well-defined region<br />

Table 23.12 Commonly used antibiotics for Crohn’s disease<br />

Follow-up management 371<br />

of disease that has not been progressive over time,<br />

surgical resection can be a highly effective therapy.<br />

The potential benefits of surgery include rapid<br />

resolution of symptoms, enhanced growth and<br />

pubertal development, and improved quality of<br />

life. Postoperative recurrence of disease is a significant<br />

risk. Data from adults have indicated that the<br />

incidence of clinical recurrence following resection<br />

is about 10% per year.<br />

Supportive measures<br />

Antibiotic Dosage Side-effects<br />

Psychological support and psychiatric counseling<br />

are important adjunctive therapies to cope with<br />

chronic disease. Children with Crohn’s disease<br />

may have to face delayed sexual maturation and<br />

short stature. Missing school impacts on social<br />

maturation and relationships. Psychiatric and<br />

psychological support is a critical aspect of a pediatric<br />

IBD program; anti-anxiety or antidepressant<br />

medications may be beneficial. Long-term therapy<br />

should be considered for any child having difficulty<br />

in school or with interpersonal relationships.<br />

Follow-up management<br />

The PCDAI is a useful monitoring tool for disease<br />

activity, but it is time-consuming to use in a<br />

routine clinical setting. 194–198 A numerical score<br />

(0–100) based on general well-being, clinical<br />

symptoms and objective measurements of weight,<br />

height and clinical examination, as well as laboratory<br />

tests provides an objective means to follow<br />

disease activity and response to therapy (Table<br />

Metronidazole 30–50mg/kg per day, TID metallic taste, disulfiram reaction with alcohol,<br />

peripheral neuropathy, potential birth defect<br />

Ciprofloxacin 20–30mg/kg per day, BID skin rash, headache, nausea, vomiting, renal<br />

failure, seizures, arthropathy<br />

Augmentin 20–40mg/kg per day, TID skin rash, nausea, vomiting, diarrhea, hypersensitivity<br />

reactions, seizures, interstitial nephritis<br />

TID, three times a day; BID, twice a day


372<br />

Crohn’s disease<br />

23.13). A score of 0–10 indicates inactive disease,<br />

11–30 mild-to-moderate disease, and over 30 moderate-to-severe<br />

disease. Signs of pubertal development<br />

and nutritional assessment should be an important<br />

part of ongoing care. Annual ophthalmological<br />

examination is important for all patients who have<br />

been exposed to corticosteroids. In addition to<br />

dietary supplement of vitamin D and calcium to<br />

prevent development of osteopenia and osteoporosis,<br />

regular bone density study using DEXA is recommended<br />

for patients on prolonged corticosteroid<br />

therapy. There are no proven therapeutic interventions<br />

in children with Crohn’s disease that have been<br />

shown to prevent recurrence of disease or maintain<br />

remission. Nevertheless, immunomodulator therapy<br />

and 5-ASA are often used in this role.<br />

Table 23.13 Pediatric Crohn’s Disease Activity Index (PCDAI) (adapted from reference 194)<br />

History (recall, 1 week) Score<br />

Abdominal pain<br />

None 0<br />

Mild (brief, does not interfere with activities 5<br />

Moderate (daily, prolonged, nocturnal, interfering with activities)<br />

Stools (per day)<br />

10<br />

0–1 (liquid, no blood) 0<br />

1–2 (semi-formed with small amount of blood)<br />

or 2–5 (liquid)<br />

5<br />

≥6 (liquid or gross bleeding or nocturnal diarrhea)<br />

Patient functioning, general well-being (recall, 1 week)<br />

10<br />

No limitation of activities, well 0<br />

Occasional difficulty in maintaining age-appropriate activities 5<br />

Frequent limitation of activity<br />

Laboratory tests<br />

Hematocrit (%)<br />

33 0<br />

28–32 2.5<br />

34 0<br />

29–33 2.5<br />


Prognosis<br />

Crohn’s disease is a chronic inflammatory process<br />

that cannot be cured by current medical therapy or<br />

by resection. Nevertheless, children with Crohn’s<br />

disease can manage their disease and achieve their<br />

goals. General Eisenhower had Crohn’s disease<br />

and he was elected President of the United States.<br />

Athletes with Crohn’s disease perform effectively<br />

and children should be encouraged to pursue their<br />

dreams. Education, recognition of early manifestations<br />

of disease and compliance will enable chil-<br />

REFERENCES<br />

1. Haubich WS. Crohn of Crohn’s disease. Gastroenterology<br />

1999; 116: 1034.<br />

2. Podolsky DK. Inflammatory bowel disease. N Engl J<br />

Med 2002; 347: 417–429.<br />

3. Loftus EV, Jr, Schoenfeld P, Sandborn WJ. The epidemiology<br />

and natural history of Crohn’s disease in population-based<br />

patient cohorts from North America: a<br />

systematic review. Aliment Pharmacol Ther 2002; 16:<br />

51–60.<br />

4. Moum B, Vatn MH, Ekbom A et al. Incidence of Crohn’s<br />

disease in four counties in southeastern Norway, 1990-<br />

93. A prospective population-based study. The<br />

Inflammatory Bowel South-Eastern Norway (IBSEN)<br />

Study Group of Gastroenterologists. Scand J<br />

Gastroenterol 1996; 31: 355–361.<br />

5. Shivananda S, Lennard-Jones J, Logan R et al. Incidence<br />

of inflammatory bowel disease across Europe: is there a<br />

difference between north and south? Results of the<br />

European Collaborative Study on Inflammatory Bowel<br />

Disease (EC-IBD). Gut 1996; 39: 690–697.<br />

6. Ranzi T, Bodini P, Zambelli A et al. Epidemiological<br />

aspects of inflammatory bowel disease in a north Italian<br />

population: a 4-year prospective study. Eur J<br />

Gastroenterol Hepatol 1996; 8: 657–661.<br />

7. Tragnone A, Corrao G, Miglio F et al. Incidence of<br />

inflammatory bowel disease in Italy: a nationwide<br />

population-based study. Gruppo Italiano per lo Studio<br />

del Colon e del Retto (GISC). Int J Epidemiol 1996; 25:<br />

1044–1052.<br />

8. Tragnone A, Hanau C, Bazzocchi G, Lanfranchi GA.<br />

Epidemiological characteristics of inflammatory bowel<br />

disease in Bologna, Italy – incidence and risk factors.<br />

Digestion 1993; 54: 183–188.<br />

9. Manousos ON, Koutroubakis I, Potamianos S et al. A<br />

prospective epidemiologic study of Crohn’s disease in<br />

Prognosis 373<br />

dren to reach their potential and maximize benefit<br />

from medical therapy.<br />

Acknowledgments<br />

We would like to thank Dr Gary J. Russell for critical<br />

reading of the manuscript, Dr Gregory Lauwers<br />

for contributing the histology photographs, Dr<br />

Esther Israel for endosopic photographs and Dr<br />

Sudha Anupindi for providing the radiographic<br />

figures.<br />

Heraklion, Crete. Incidence over a 5-year period. Scand<br />

J Gastroenterol 1996; 31: 599–603.<br />

10. Sonnenberg A, McCarty DJ, JacobsenSJ. Geographic<br />

variation of inflammatory bowel disease within the<br />

United States. Gastroenterology 1991; 100: 143–149.<br />

11. Mayberry J, Mann R. Inflammatory bowel disease in<br />

rural sub-Saharan Africa: rarity of diagnosis in patients<br />

attending mission hospitals. Digestion 1989; 44:<br />

172–176.<br />

12. Kurata JH, Kantor-Fish S, Frankl H et al. Crohn’s disease<br />

among ethnic groups in a large health maintenance<br />

organization. Gastroenterology 1992; 102: 1940–1948.<br />

13. Calkins BM, Lilienfeld AM, Garland CF, Mendeloff AI.<br />

Trends in incidence rates of ulcerative colitis and<br />

Crohn’s disease. Dig Dis Sci 1984; 29: 913–920.<br />

14. Roth MP, Petersen GM, McElree C et al. Geographic<br />

origins of Jewish patients with inflammatory bowel<br />

disease. Gastroenterology 1989; 97: 900–904.<br />

15. Askling J, Grahnquist L, Ekbom A, Finkel Y. Incidence<br />

of paediatric Crohn’s disease in Stockholm, Sweden.<br />

Lancet 1999; 354: 1179.<br />

16. Barton JR, Gillon S, Ferguson A. Incidence of inflammatory<br />

bowel disease in Scottish children between 1968<br />

and 1983; marginal fall in ulcerative colitis, three-fold<br />

rise in Crohn’s disease. Gut 1989; 30: 618–622.<br />

17. Armitage E, Drummond H, Ghosh S, Ferguson A.<br />

Incidence of juvenile-onset Crohn’s disease in Scotland.<br />

Lancet 1999; 353: 1496–1497.<br />

18. Kugathasan S, Judd RH, Hoffman RG et al.<br />

Epidemiologic and clinical characteristics of children<br />

with newly diagnosed inflammatory bowel disease in<br />

Wisconsin: a statewide population-based study. J<br />

Pediatrics 2003; 143: 525–531.<br />

19. Jenkins, H. R. Inflammatory bowel disease. Arch Dis<br />

Child 2001; 85: 435–437.


374<br />

Crohn’s disease<br />

20. Roth MP, Petersen GM, McElreeC et al. Familial empiric<br />

risk estimates of inflammatory bowel disease in<br />

Ashkenazi Jews. Gastroenterology 1989; 96: 1016–1020.<br />

21. Monsen U, Brostrom O, Nordenvall B et al. Prevalence<br />

of inflammatory bowel disease among relatives of<br />

patients with ulcerative colitis. Scand J Gastroenterol<br />

1987; 22: 214–218.<br />

22. Fielding JF. The relative risk of inflammatory bowel<br />

disease among parents and siblings of Crohn’s disease<br />

patients. J Clin Gastroenterol 1986; 8: 655–657.<br />

23. Yang H, McElree C, Roth MP et al. Familial empirical<br />

risks for inflammatory bowel disease: differences<br />

between Jews and non-Jews. Gut 1993; 34: 517–524.<br />

24. Orholm M, Munkholm P, Langholz E et al. Familial<br />

occurrence of inflammatory bowel disease. N Engl J Med<br />

1991; 324: 84–88.<br />

25. Lee JC, Lennard-Jones JE. Inflammatory bowel disease<br />

in 67 families each with three or more affected firstdegree<br />

relatives. Gastroenterology 1996; 111: 587–596.<br />

26. Polito JM 2nd, Childs B, Mellits ED et al. Crohn’s<br />

disease: influence of age at diagnosis on site and clinical<br />

type of disease. Gastroenterology 1996; 111:<br />

580–586.<br />

27. Colombel JF, Grandbastien B, Gower-Rousseau C et al.<br />

Clinical characteristics of Crohn’s disease in 72 families.<br />

Gastroenterology 1996; 111: 604–607.<br />

28. Tysk C, Lindberg E, Jarnerot G, Floderus-Myrhed B.<br />

Ulcerative colitis and Crohn’s disease in an unselected<br />

population of monozygotic and dizygotic twins. A study<br />

of heritability and the influence of smoking. Gut 1988;<br />

29: 990–996.<br />

29. Hugot JP, Laurent-Puig P, Gower-Rousseau C et al.<br />

Mapping of a susceptibility locus for Crohn’s disease on<br />

chromosome 16. Nature 1996; 379: 821–823.<br />

30. Satsangi J, Parkes M, Louis E et al. Two stage genomewide<br />

search in inflammatory bowel disease provides<br />

evidence for susceptibility loci on chromosomes 3, 7<br />

and 12. Nat Genet 1996; 14: 199–202.<br />

31. Brant SR, Fu Y, Fields CT et al. American families with<br />

Crohn’s disease have strong evidence for linkage to<br />

chromosome 16 but not chromosome 12.<br />

Gastroenterology 1998; 115: 1056–1061.<br />

32. Ma Y, Ohmen JD, Li Z et al. A genome-wide search<br />

identifies potential new susceptibility loci for Crohn’s<br />

disease. Inflamm Bowel Dis 1999; 5: 271–278.<br />

33. Hugot J, Chamaillard PM, Zouali H et al.Association of<br />

NOD2 leucine-rich repeat variants with susceptibility to<br />

Crohn’s disease. Nature 2001; 411: 599–603.<br />

34. Ogura Y, Bonen DK, Inohara N et al. A frameshift mutation<br />

in NOD2 associated with susceptibility to Crohn’s<br />

disease. Nature 2001; 411: 603–606.<br />

35. Cuthbert AP, Fisher SA, Mirza MM et al. The contribution<br />

of NOD2 gene mutations to the risk and site of<br />

disease in inflammatory bowel disease. Gastroenterology<br />

2002; 122: 867–874.<br />

36. Lesage S, Zouali H, Cezard JP et al. CARD15/NOD2<br />

mutational analysis and genotype–phenotype correlation<br />

in 612 patients with inflammatory bowel disease.<br />

Am J Hum Genet 2002; 70: 845–857.<br />

37. Tomer G, Ceballos C, Concepcion E, Benkov KJ.<br />

NOD2/CARD15 variants are associated with lower<br />

weight at diagnosis in children with Crohn’s disease.<br />

Am J Gastroenterology 2003; 98: 2479–2484.<br />

38. Rioux JD, Silverberg MS, Daly MJ et al. Genomewide<br />

search in Canadian families with inflammatory bowel<br />

disease reveals two novel susceptibility loci. Am J Hum<br />

Genet 2000; 66: 1863–1870.<br />

39. Rioux JD, Daly MJ, Silverberg MS et al. Genetic variation<br />

in the 5q31 cytokine gene cluster confers susceptibility<br />

to Crohn disease. Nat Genet 2001; 29: 223–228.<br />

40. Thornton JR, Emmett PM, Heaton KW. Diet and Crohn’s<br />

disease: characteristics of the pre-illness diet. Br Med J<br />

1979; 2: 762–764.<br />

41. Jarnerot G, Jarnmark I, Nilsson K. Consumption of<br />

refined sugar by patients with Crohn’s disease, ulcerative<br />

colitis, or irritable bowel syndrome. Scand J<br />

Gastroenterol 1983; 18: 999–1002.<br />

42. Shoda R, Matsueda K, Yamato S, Umeda N.<br />

Epidemiologic analysis of Crohn disease in Japan:<br />

increased dietary intake of n-6 polyunsaturated fatty<br />

acids and animal protein relates to the increased incidence<br />

of Crohn disease in Japan. Am J Clin Nutr 1996;<br />

63: 741–745.<br />

43. Koletzko S, Sherman P, Corey M et al. Role of infant<br />

feeding practices in development of Crohn’s disease in<br />

childhood. Br Med J 1989; 298: 1617–1628.<br />

44. Koletzko S, Griffiths A, Corey M et al. Infant feeding<br />

practices and ulcerative colitis in childhood. Br Med J<br />

1991; 302: 1580–1581.<br />

45. Gilat T, Hacohen D, Lilos P, Langman MJ. Childhood<br />

factors in ulcerative colitis and Crohn’s disease. An<br />

international cooperative study. Scand J Gastroenterol<br />

1987; 22: 1009–1024.<br />

46. Ekbom A, Adami HO, Helmick GC et al. Perinatal risk<br />

factors for inflammatory bowel disease: a case–control<br />

study. Am J Epidemiol 1990; 132: 1111–1119.<br />

47. Vessey M, Jewell D, Smith A et al. Chronic inflammatory<br />

bowel disease, cigarette smoking, and use of oral<br />

contraceptives: findings in a large cohort study of<br />

women of childbearing age. Br Med J (Clin Res Ed) 1986;<br />

292: 1101–1103.<br />

48. Calkins BM. A meta-analysis of the role of smoking in<br />

inflammatory bowel disease. Dig Dis Sci 1989; 34:<br />

1841–1845.<br />

49. Cosnes J, Beaugerie L, Carbonnel F, Gendre JP. Smoking<br />

cessation and the course of Crohn’s disease: an intervention<br />

study. Gastroenterology 2001; 120: 1093–1099.<br />

50. Harries AD, Jones L, Heatley RV, Rhodes J. Smoking<br />

habits and inflammatory bowel disease: effect on nutrition.<br />

Br Med J (Clin Res Ed) 1982; 284: 1161.<br />

51. Boyko EJ, Koepsell TD, Perera DR, Inui TS. Risk of<br />

ulcerative colitis among former and current cigarette<br />

smokers. N Engl J Med 1987; 316: 707–710.<br />

52. Tanner AR, Raghunath AS. Colonic inflammation and<br />

nonsteroidal anti-inflammatory drug administration. An<br />

assessment of the frequency of the problem. Digestion<br />

1988; 41: 116–120.<br />

53. Kaufmann HJ, Taubin HL. Nonsteroidal anti-inflammatory<br />

drugs activate quiescent inflammatory bowel<br />

disease. Ann Intern Med 1987; 107: 513–516.<br />

54. Evans JM, McMahon AD, Murray FE et al. Non-steroidal<br />

anti-inflammatory drugs are associated with emergency<br />

admission to hospital for colitis due to inflammatory<br />

bowel disease. Gut 1997; 40: 619–622.<br />

55. Felder JB, Korelitz BI, Rajapakse R et al. Effects of nonsteroidal<br />

anti-inflammatory drugs on inflammatory<br />

bowel disease: a case–control study. Am J Gastroenterol<br />

2000; 95: 1949–1954.<br />

56. Bonner GF, Walczak PA, Kitchen L, Bayona M. Tolerance<br />

of nonsteroidal antiinflammatory drugs in patients with<br />

inflammatory bowel disease. Am J Gastroenterol 2000;<br />

95: 1946–1948.<br />

57. O’Brien J. Nonsteroidal anti-inflammatory drugs in<br />

patients with inflammatory bowel disease. Am J<br />

Gastroenterol 2000; 95: 1859–1861.<br />

58. Bjarnason I, Hayllar J, MacPherson AJ, Russell AS. Side<br />

effects of nonsteroidal anti-inflammatory drugs on the<br />

small and large intestine in humans. Gastroenterology<br />

1993; 104: 1832–1847.


59. Wallace JL. Nonsteroidal anti-inflammatory drugs and<br />

gastroenteropathy: the second hundred years.<br />

Gastroenterology 1997; 112: 1000–1016.<br />

60. Reuter BK, Davies NM, Wallace JL. Nonsteroidal antiinflammatory<br />

drug enteropathy in rats: role of permeability,<br />

bacteria, and enterohepatic circulation.<br />

Gastroenterology 1997; 112: 109–117.<br />

61. Pumford NR, Myers TG, Davila JC et al.<br />

Immunochemical detection of liver protein adducts of<br />

the nonsteroidal antiinflammatory drug diclofenac.<br />

Chem Res Toxicol 1993; 6: 147–150.<br />

62. Atchison CR, West AB, Balakumaran A et al. Drug enterocyte<br />

adducts: possible causal factor for diclofenac<br />

enteropathy in rats. Gastroenterology 1996; 119:<br />

1537–1547.<br />

63. Halter F, Tarnawski AS, Schmassmann A, Peskar BM.<br />

Cyclooxygenase 2–implications on maintenance of<br />

gastric mucosal integrity and ulcer healing: controversial<br />

issues and perspectives. Gut 2001; 49: 443–453.<br />

64. Wallace JL, McKnight W, Reuter BK, Vergnolle N.<br />

NSAID-induced gastric damage in rats: requirement for<br />

inhibition of both cyclooxygenase 1 and 2.<br />

Gastroenterology 2000; 119: 706–714.<br />

65. Tanaka A, Hase S, Miyazawa T, Takeuchi K. Up-regulation<br />

of cyclooxygenase-2 by inhibition of cyclooxygenase-1:<br />

a key to nonsteroidal anti-inflammatory druginduced<br />

intestinal damage. J Pharmacol Exp Ther 2002;<br />

300: 754–761.<br />

66. Silverstein FE, Faich G, Goldstein JL et al.<br />

Gastrointestinal toxicity with celecoxib vs nonsteroidal<br />

anti-inflammatory drugs for osteroarthritis and rheumatoid<br />

arthritis: the CLASS study: a randomized<br />

controlled trial. Celecoxib long-term arthritis safety<br />

study. J Am Med Assoc 2000; 284: 1247–1255.<br />

67. Watson DJ, Harper SE, ZhaoPL et al. Gastrointestinal<br />

tolerability of the selective cyclooxygenase-2 (COX-2)<br />

inhibitor rofecoxib compared with nonselective COX-1<br />

and COX-2 inhibitors in osteoarthritis. Arch Intern Med<br />

2000; 160: 2998–3003.<br />

68. Singer II, Kawka DW, Schloemann S et al.<br />

Cyclooxygenase 2 is induced in colonic epithelial cells<br />

in inflammatory bowel disease. Gastroenterology 1998;<br />

115: 297–306.<br />

69. Handel J, Nielsen OH. Expression of cyclooxygenase-2<br />

mRNA in active inflammatory bowel disease. Am J<br />

Gastroenterol 1997; 92: 1170–1173.<br />

70. Bantel H, Berg C, Vieth M et al. Mesalazine inhibits<br />

activation of transcription factor NF-kappaB in inflamed<br />

mucosa of patients with ulcerative colitis. Am J<br />

Gastroenterol 2000; 95: 3452–3457.<br />

71. Logan RF. Inflammatory bowel disease incidence: up,<br />

down or unchanged? Gut 1998; 42: 309–311.<br />

72. Lashner BA, Kane SV Hanauer SB. Lack of association<br />

between oral contraceptive use and Crohn’s disease: a<br />

community-based matched case-control study.<br />

Gastroenterology 1989; 97: 1442–1447.<br />

73. Lashner BA, Kane SV, Hanauer SB. Lack of association<br />

between oral contraceptive use and ulcerative colitis.<br />

Gastroenterology 1990; 99: 1032–1036.<br />

74. Wakefield AJ, Ekbom A, Dhillon AP et al. Crohn’s<br />

disease: pathogenesis and persistent measles virus<br />

infection. Gastroenterology 1995; 108: 911–916.<br />

75. Wakefield AJ, Pittilo RM, Sim R et al. Evidence of<br />

persistent measles virus infection in Crohn’s disease. J<br />

Med Virol 1993; 39: 345–353.<br />

76. Haga Y, Funakoshi O, Kuroe K et al. Absence of measles<br />

viral genomic sequence in intestinal tissues from<br />

Crohn’s disease by nested polymerase chain reaction.<br />

Gut 1996; 38: 211–215.<br />

References 375<br />

77. Ekbom A, Wakefield AJ, Zack M, Adami HO. Perinatal<br />

measles infection and subsequent Crohn’s disease.<br />

Lancet 1994; 344: 508–510.<br />

78. Thompson NP, Pounder RE, Wakefield AJ. Perinatal and<br />

childhood risk factors for inflammatory bowel disease: a<br />

case–control study. Eur J Gastroenterol Hepatol 1995; 7:<br />

385–390.<br />

79. Haslam N, Mayberry JF, Hawthorne AB et al. Measles,<br />

month of birth, and Crohn’s disease. Gut 2000; 47:<br />

801–803.<br />

80. Thompson NP, Montgomery SM, Pounder RE, Wakefield<br />

AJ. Is measles vaccination a risk factor for inflammatory<br />

bowel disease? Lancet 1995; 345: 1071–1074.<br />

81. Feeney M, Ciegg A, Winwood P, Snook J. A case–control<br />

study of measles vaccination and inflammatory bowel<br />

disease. The East Dorset Gastroenterology Group. Lancet<br />

1997; 350: 764–766.<br />

82. Davis RL, Kramarz P, Bohlke K et al.<br />

Measles–mumps–rubella and other measles-containing<br />

vaccines do not increase the risk for inflammatory<br />

bowel disease: a case–control study from the Vaccine<br />

Safety Datalink project. Arch Pediatr Adolesc Med 2001;<br />

155: 354–359.<br />

83. Lisby G, Andersen J, Engbaek K, Binder V.<br />

Mycobacterium paratuberculosis in intestinal tissue<br />

from patients with Crohn’s disease demonstrated by a<br />

nested primer polymerase chain reaction. Scand J<br />

Gastroenterol 1994; 29: 923–929.<br />

84. Fiocchi C. Inflammatory bowel disease: etiology and<br />

pathogenesis. Gastroenterology 1998; 115: 182–205.<br />

85. Shanahan F. Crohn’s disease. Lancet 2002; 359: 62–69.<br />

86. Ardizzone S, Bianchi Porro G. Inflammatory bowel<br />

disease: new insights into pathogenesis and treatment. J<br />

Intern Med 2002; 252: 475–496.<br />

87. Fabia R, Ar’Rajab A, Andersson ML et al. Impairment of<br />

bacterial flora in human ulcerative colonic and experimental<br />

colitis in the rat. Digestion 1993; 54: 248–255.<br />

88. Contractor NV, Bassiri H, Reya T et al. Lymphoid hyperplasia,<br />

autoimmunity, and compromised intestinal<br />

intraepithelial lymphocyte developement in colitis-free<br />

gnotobiotic IL-12-deficient mice. J Immunol 1998; 160:<br />

385–394.<br />

89. Taurog JD, Richardson JA, Croft JT et al. The germfree<br />

state prevents development of gut and joint inflammatory<br />

disease in HLA-B27 transgenic rats. J Exp Med<br />

1994; 180: 2359–2364.<br />

90. Sartor R. Microbial factors in the pathogenesis of<br />

Crohn’s disease, ulcerative colitis, and experimental<br />

intestinal inflammation. In Kirsner JB, ed. Inflammatory<br />

Bowel Disease. Philadelphia: WB Saunders, 2000: 153.<br />

91. Duchmann R, Kaiser I, Hermann E et al. Tolerance<br />

exists towards resident intestinal flora but is broken in<br />

active inflammatory bowel disease. Clin Exp Immunol<br />

1995; 102: 448–455.<br />

92. Duchmann R, May E, Heike M et al. T cells specificity<br />

and cross reactivity towards enterobacteria, bacteroides,<br />

bifidobacterium, and antigens from resident intestinal<br />

flora in humans. Gut 1999; 44: 812–818.<br />

93. Darfeuille-Michaud A, Neut C, Barnich N et al. Presence<br />

of adherent Escherichia coli strains in ileal mucosa of<br />

patients with Crohn’s disease. Gastroenterology 1998;<br />

115: 1405–1413.<br />

94. Swidsinski A, Ladhoff A, Pernthaler A et al. Mucosal<br />

flora in inflammatory bowel disease. Gastroenterology<br />

2002; 122: 44–54.<br />

95. Rutgeerts P, D’Haens G, Hiele M, et al. Appendectomy<br />

protects against ulcerative colitis. Gastroenterology<br />

1994; 106: 1251–1253.<br />

96. Sandler RS. Appendicectomy and ulcerative colitis.<br />

Lancet 1998; 352: 1797–1798.


376<br />

Crohn’s disease<br />

97. Andersson RE, Olaison G, Tysk C, Ekbom A.<br />

Appendectomy and protection against ulcerative colitis.<br />

N Engl J Med 2001; 344: 808–814.<br />

98. Cosnes J, Carbonnel F, Beaugerie L et al. Effects of<br />

appendicectomy on the course of ulcerative colitis. Gut<br />

2002; 51: 803–807.<br />

99. Radford-Smith GL, Edwards JE, Purdie DM et al.<br />

Protective role of appendicectomy on onset and severity<br />

of ulcerative colitis and Crohn’s disease. Gut 2002; 51:<br />

808–813.<br />

100. Selby WS, Griffin S, Abraham N, Solomon MJ.<br />

Appendectomy protects against the development of<br />

ulcerative colitis but does not affect its course. Am J<br />

Gastroenterol 2002; 97: 2834–2838.<br />

101. Andersson RE, Olaison G, Tysk C, Ekbom A.<br />

Appendectomy is followed by increased risk of Crohn’s<br />

disease. Gastroenterology 2003; 124: 40–46.<br />

102. Mizoguchi A, Mizoguchi E, Chiba C, Bhan AK. Role of<br />

appendix in the development of inflammatory bowel<br />

disease in TCR-a mutant mice. J Exp Med 1996; 184:<br />

707–715.<br />

103. Griffiths AM, Buller HB. Inflammatory bowel disease. In<br />

Walker WA, Durie PR, Hamilton JR et al., eds. Pediatric<br />

Gastrointestinal Disease. Ontario, BC: Decker, 2000: 613.<br />

104. Monteleone I, Vavassori P, Biancone L et al.<br />

Immunoregulation in the gut: success and failures in<br />

human disease. Gut 2002; 50(Suppl 3): III60–III64.<br />

105. De Winter H, Cheroutre H, Kronenberg M. Mucosal<br />

immunity and inflammation. II. The yin and yang of T<br />

cells in intestinal inflammation: pathogenic and protective<br />

roles in a mouse colitis model. Am J Physiol 1999;<br />

276: G1317–G1321.<br />

106. MacDonald TT, Monteleone G, Pender SL. Recent developments<br />

in the immunology of inflammatory bowel<br />

disease. Scand J Immunol 2000; 51: 2–9.<br />

107. Singh B, Read S, Asseman C et al. Control of intestinal<br />

inflammation by regulatory T cells. Immunol Rev 2001;<br />

182: 190–200.<br />

108. Toms C, Powrie F. Control of intestinal inflammation by<br />

regulatory T cells. Microbes Infect 2001; 3: 929–935.<br />

109. Read S, Powrie F. CD4(+) regulatory T cells. Curr Opin<br />

Immunol 2001; 13: 644–649.<br />

110. Micallef MJ, Ohtsuki T, Kohno K et al. Interferongamma-inducing<br />

factor enhances T helper 1 cytokine<br />

production by stimulated human T cells: synergism<br />

with interleukin-12 for interferon-gamma production.<br />

Eur J Immunol 1996; 26: 1647–1651.<br />

111. Matsui K, Yoshimoto T, Tsutsui H et al.<br />

Propionibacterium acnes treatment diminishes CD4+<br />

NK1.1+ T cells but induces type I T cells in the liver by<br />

induction of IL-12 and IL-18 production from Kupffer<br />

cells. J Immunol 1997; 159: 97–106.<br />

112. Dinarello CA. Interleukin-1 and tumor necrosis factor:<br />

effector cytokines in autoimmune diseases. Semin<br />

Immunol 1992; 4: 133–145.<br />

113. Mackay F, Loetscher H, Stueber D et al. Tumor necrosis<br />

factor alpha (TNF-alpha)-induced cell adhesion to<br />

human endothelial cells is under dominant control of<br />

one TNF receptor type, TNF-R55. J Exp Med 1993; 177:<br />

1277–1286.<br />

114. Schmitz H, Fromm M, Bode H et al. Tumor necrosis<br />

factor-alpha induces Cl- and K+ secretion in human<br />

distal colon driven by prostaglandin E2. Am J Physiol<br />

1996; 271: G669–G674.<br />

115. Mullin JM, Snock KV. Effect of tumor necrosis factor on<br />

epithelial tight junctions and transepithelial permeability.<br />

Cancer Res 1990; 50: 2172–2176.<br />

116. Barton JR, Ferguson A. Clinical features, morbidity and<br />

mortality of Scottish children with inflammatory bowel<br />

disease. Q J Med 1990; 75: 423–439.<br />

117. Motil KJ, Grand RJ, Davis-Kraft L et al. Growth failure in<br />

children with inflammatory bowel disease: a prospective<br />

study. Gastroenterology 1993; 105: 681–691.<br />

118. Kanof ME, Lake AM, Bayless TM. Decreased height<br />

velocity in children and adolescents before the diagnosis<br />

of Crohn’s disease. Gastroenterology 1988; 95:<br />

1523–1527.<br />

119. Cezard JP, Touati G, Alberti C et al. Growth in paediatric<br />

Crohn’s disease. Horm Res 2002; 58 (Suppl 1): 11–15.<br />

120. Lenaerts C, Roy CC, Vaillancourt M et al. High incidence<br />

of upper gastrointestinal tract involvement in children<br />

with Crohn disease. Pediatrics 1989; 83: 777–781.<br />

121. Ballinger A. Fundamental mechanisms of growth failure<br />

in inflammatory bowel disease. Horm Res 2002; 58<br />

(Suppl 1): 7–10.<br />

122. Zurita VF, Rawls DE, Dyck WP. Nutritional support in<br />

inflammatory bowel disease. Dig Dis Sci 1995; 13: 92–107.<br />

123. Markowitz J, Grancher K, Rosa J et al. Highly destructive<br />

perianal disease in children with Crohn’s disease. J<br />

Pediatr Gastroenterol Nutr 1995; 21: 149–153.<br />

124. Tolia V. Perianal Crohn’s disease in children and adolescents.<br />

Am J Gastroenterol 1996; 91: 922–926.<br />

125. Greenstein AJ, Janowitz HD, Sachar DB. The extraintestinal<br />

complications of Crohn’s disease and ulcerative<br />

colitis: a study of 700 patients. Medicine (Baltimore)<br />

1976; 55: 401–412.<br />

126. Hyams JS. Extraintestinal manifestations of inflammatory<br />

bowel disease in children. J Pediatr Gastroenterol<br />

Nutr 1994; 19: 7–21.<br />

127. Seidman E, LeLeiko N, Ament M et al. Nutritional<br />

issues in pediatric inflammatory bowel disease. J Pediatr<br />

Gastroenterol Nutr 1991; 12: 424–438.<br />

128. Bernstein CN, Blanchard JF, Houston DS, Wajda A. The<br />

incidence of deep venous thrombosis and pulmonary<br />

embolism among patients with inflammatory bowel<br />

disease: a population-based cohort study. Thromb<br />

Haemost 2001; 85: 430–434.<br />

129. Markowitz RL, Ment LR, Gryboski JD. Cerebral thromboembolic<br />

disease in pediatric and adult inflammatory<br />

bowel disease: case report and review of the literature. J<br />

Pediatr Gastroenterol Nutr 1989; 8: 413–420.<br />

130. Talbot RW, Heppell J, Dozois RR, Beart RWJ. Vascular<br />

complications of inflammatory bowel disease. Mayo<br />

Clin Proc 1986; 61: 140–145.<br />

131. Sanghavi P, Paramesh A, Dwivedi A, et al. Mesenteric<br />

arterial thrombosis as complication of Crohn’s disease.<br />

Dig Dis Sci 1999; 46: 2344–2346.<br />

132. Gormally SM, Bourke W, Kierse B et al. Isolated cerebral<br />

thrombo-embolism and Crohn’s disease. Eur J Pediatr<br />

1995; 154: 815–818.<br />

133. Akobeng A, Miller V, Thomas AG. Epilepsy and Crohn’s<br />

disease in children. J Pediatr Gastroenterol Nutr 1998; 26:<br />

458–460.<br />

134. Ertem D, Ozguven E, Acar Y et al. Thromboembolic<br />

complications in children with Crohn’s disease. J<br />

Pediatr Gastroenterol Nutr 1999; 28: 540–541.<br />

135. Garcia-Diaz M, Mira M, Nevado L et al. Retinal vasculitis<br />

associated with Crohn’s disease. Postgrad Med J 1995;<br />

71: 170–172.<br />

136. Greenfield SM, Teare JP, Whitehead MW, Thompson RP.<br />

Amaurosis fugax, Crohn’s disease and the cardiolipin<br />

antibody. Lupus 1993; 2: 271–273.<br />

137. van Tirpitz C, Klaus J, Steinkamp M et al. Therapy of<br />

osteoporosis in patients with Crohn’s disease: a randomized<br />

study comparing sodium fluoride and ibandronate.<br />

Aliment Pharmacol Ther 2003; 17: 807–816.<br />

138. Winter HS, Landers CJ, Winkelstein A et al. Antineutrophil<br />

cytoplasmic antibodies in children with ulcerative<br />

colitis. J Pediatr 1994; 125: 707–711.


139. Ruemmele FM, Targan SR, Levy G et al. Diagnostic accuracy<br />

of serological assays in pediatric inflammatory<br />

bowel disease. Gastroenterology 1998; 115: 822–829.<br />

140. Terjung B, Spengler U, Sauerbruch T, Worman HJ.<br />

“Atypical p-ANCA” in IBD and hepatobiliary disorders<br />

react with a 50-kilodalton nuclear envelope protein of<br />

neutrophils and myeloid cell lines. Gastroenterology<br />

2000; 119: 310–322.<br />

141. Charron M, del Rosario FJ, Kocoshis SA. Pediatric inflammatory<br />

bowel disease: assessment with scintigraphy with<br />

99mTc white blood cells. Radiology 1999; 212: 507–513.<br />

142. Alberini JL, Badran A, Freneaux E et al. Technetium-99m<br />

HMPAO-labeled leukocyte imaging compared with<br />

endoscopy, ultrasonography, and contrast radiology in<br />

children with inflammatory bowel disease. J Pediatr<br />

Gastroenterol Nutr 2001; 32: 278–286.<br />

143. Charron M, Di Lorenzo C, Kocoshis S. Are 99mTc leukocyte<br />

scintigraphy and SBFT studies useful in children<br />

suspected of having inflammatory bowel disease? Am J<br />

Gastroenterol 2000; 95: 1208–1212.<br />

144. Charron M, del Rosario JF, Kocoshis S. Use of technetium-tagged<br />

white blood cells in patients with Crohn’s<br />

disease and ulcerative colitis: is differential diagnosis<br />

possible? Pediatr Radiol 1998; 28: 871–877.<br />

145. Surawicz CM, Meisel JL, Ylvisaker T et al. Rectal biopsy<br />

in the diagnosis of Crohn’s disease: value of multiple<br />

biopsies and serial sectioning. Gastroenterology 1981; 80:<br />

66–71.<br />

146. Chong SK, Blackshaw AJ, Boyle S et al. Histological diagnosis<br />

of chronic inflammatory bowel disease in childhood.<br />

Gut 1985; 26: 55–59.<br />

147. Brattsand R, Linden M. Cytokine modulation by glucocorticoids:<br />

mechanisms and actions in cellular studies.<br />

Aliment Pharmacol Ther 1996; 10: 81–90.<br />

148. Zimmerman MJ, Jewell DP. Cytokines and mechanisms of<br />

action of glucocorticoids and aminosalicylates in the<br />

treatment of ulcerative colitis and Crohn’s disease.<br />

Aliment Pharmacol Ther 1996; 10: 93–98.<br />

149. Brattsand R. Overview of newer glucocorticoid preparations<br />

for inflammatory bowel disease. Can J Gastroenterol<br />

1990; 4: 407–414.<br />

150. Rutgeerts P, Lofberg R, Malchow H et al. A comparison of<br />

budesonide with prednisolone for active Crohn’s disease.<br />

N Engl J Med 1994; 331: 842–845.<br />

151. Kundhal P, Zachos M, Holmes JL, Griffiths AM.<br />

Controlled ileal release budesonide in pediatric Crohn’s<br />

disease: efficacy and effect on growth. J Pediatr<br />

Gastroenterol Nutr 2001; 33: 75–80.<br />

152. Cuffari C, Theoret Y, Latour S, Seidman G. 6-<br />

Mercaptopurine metabolism in Crohn’s disease.<br />

Correlation with efficacy and toxicity. Gut 1996; 39:<br />

401–406.<br />

153. Markowitz J, Grancher K, Kohn N et al. A multicenter<br />

trial of 6-mercaptopurine and prednisone in children<br />

with newly diagnosed Crohn’s disease. Gastroenterology<br />

2000; 119: 895–902.<br />

154. Dubinsky MC, Griffiths AM. Contorversies in IBD: monitoring<br />

of AZA/6-MP treatment in children with IBD.<br />

Inflamm Bowel Dis 2003; 9: 386–388.<br />

155. Feagan BG, Fedorak RN, Irvine EJ et al. A comparison of<br />

methotrexate with placebo for the maintenance of remission<br />

in Crohn’s disease. N Engl J Med 2000; 342:<br />

1627–1632.<br />

156. Mack DR, Young R, Kaufman SS et al. Methotrexate in<br />

patients with Crohn’s disease after 6-mercaptopurine. J<br />

Pediatr 1998; 132: 830–835.<br />

157. Targan SR, Hanauer SB, van Deventer SJ et al. A shortterm<br />

study of chimeric monoclonal antibody cA2 to<br />

tumor necrosis factor alpha for Crohn’s disease. N Engl J<br />

Med 1997; 337: 1029–1035.<br />

References 377<br />

158. Present DH, Rutgeerts P, Targan S et al. Infliximab for<br />

the treatment of fistulas in patients with Crohn’s<br />

disease. N Engl J Med 1999, 340: 1398–1405.<br />

159. Cohen RD, Tsang JF, Hanauer SB. Infiximab in Crohn’s<br />

disease: first anniversary clinical experience. Am J<br />

Gastroenterol 2000; 95: 3469–3477.<br />

160. Hanauer SB, Feagan BG, Lichtenstein GR et al.<br />

Maintenance infliximab for Crohn’s disease: the<br />

ACCENT I randomised trial. Lancet 2002; 359:<br />

1541–1549.<br />

161. Hyams JS, Markowitz J, Wyllie R. Use of infliximab in<br />

the treatment of Crohn’s disease in children and adolescents.<br />

J Pediatr 2000; 137: 192–196.<br />

162. Baldassano R, Braegger CP, Escher JC et al. Infliximab<br />

(REMICADE) therapy in the treatment of pediatric<br />

Crohn’s disease. Am J Gastroenterol 2003; 98: 833–838.<br />

163. Cezard JP, Nouaili N, Talbotec C et al. A prospective<br />

study of the efficacy and tolerance of a chimeric antibody<br />

to tumor necrosis factors (Remicade) in severe<br />

pediatric Crohn’s disease. J Pediatr Gastroenterol Nutr<br />

2003; 36: 632–636.<br />

164. Duerr RH, Barmada MM, Zhang L et al. High-density<br />

genome scan in Crohn disease shows confirmed linkage<br />

to chromosome 14q11-12. Am J Hum Genet 2000; 66:<br />

1857–1862.<br />

165. Cho JH, Nicolae DL, Gold LH et al. Identification of<br />

novel susceptibility loci for inflammatory bowel disease<br />

on chromosomes 1p, 3q, and 4q: evidence for epistasis<br />

between 1p and IBD1. Proc Natl Acad Sci USA 1998; 95:<br />

7502–7507.<br />

166. Gryboski JD. Crohn’s disease in children 10 years old<br />

and younger: comparison with ulcerative colitis. J<br />

Pediatr Gastroenterol Nutr 1994; 18: 174–182.<br />

167. Keljo DJ, Sugerman KS. Pancreatitis in patients with<br />

inflammatory bowel disease. J Pediatr Gastroenterol Nutr<br />

1997; 25: 108–112.<br />

168. Perkal MF, Seashore JH. Nutrition and inflammatory<br />

bowel disease. Gastroenterol Clin North Am 1989; 18:<br />

567–578.<br />

169. Harries AD, Jones LA, Heatley RV, Rhodes J.<br />

Malnutrition in inflammatory bowel disease: an anthropometric<br />

study. Hum Nutr Clin Nutr 1982; 36: 307–313.<br />

170. Behrend C, Jeppesen PB, Mortensen PB. Vitamin B12<br />

absorption after ileorectal anastomosis for Crohn’s<br />

disease: effect of ileal resection and time span after<br />

surgery. Eur J Gastroenterol Hepatol 1995; 7: 397–400.<br />

171. Fernandez-Banares F, Abad-Lacruz A, Xiol X et al.<br />

Vitamin status in patients with inflammatory bowel<br />

disease. Am J Gastroenterol 1989; 84: 744–748.<br />

172. Kuroki F, Iida M, Tominaga M et al. Multiple vitamin<br />

status in Crohn’s disease. Correlation with disease activity.<br />

Dig Dis Sci 1993; 38: 1614–1618.<br />

173. Geerling BJ, Badart-Smook A, Stockbrugger RW,<br />

Brummer RJ. Comprehensive nutritional status in<br />

patients with long-standing Crohn disease currently in<br />

remission. Am J Clin Nutr 1998; 67: 919–926.<br />

174. Jacob RM. Vitamin C. In Shils Olson J, Shike M et al.<br />

eds. Modern Nutrition in Health and Disease.<br />

Philadelphia: Lippincott, 2000: 467.<br />

175. Steger GG, Mader RM, Vogelsang H et al. Folate absorption<br />

in Crohn’s disease. Digestion 1994; 55: 234–238.<br />

176. Abu-Qurshin R, Naschitz JE, Zuckermann E et al.<br />

Crohn’s disease associated with pellagra and increased<br />

excretion of 5-hydroxyindolacetic acid. Am J Med Sci<br />

1997; 313: 111–113.<br />

177. Food and Nutrition Board, Institute of Medicine. Dietary<br />

Reference Intakes. Thiamine, Riboflavin, Niacin, Vitamin<br />

B6, Folate, Vitamin B12, Pantothenic Acid, Biotin, and<br />

Choline. Washington, DC: National Academy Press,<br />

1989.


378<br />

Crohn’s disease<br />

178. Food and Nutrition Board, National Research Council.<br />

In Recommended Dietary Allowances. Washington, DC:<br />

National Academy Press, 1989.<br />

179. Main AN, Mills PR, Russell RI et al. Vitamin A deficiency<br />

in Crohn’s disease. Gut 1983; 24: 1169–1175.<br />

180. Schoelmerich J, Becher MS, Hoppe-Seyler P et al. Zinc<br />

and vitamin A deficiency in patients with Crohn’s<br />

disease is correlated with activity but not with localization<br />

or extent of the disease. Hepatogastroenterology<br />

1985; 32: 34–38.<br />

181. Janczewska I, Bartnik W, Butruk E et al. Metabolism of<br />

vitamin A in inflammatory bowel disease.<br />

Hepatogastroenterology 1991; 38: 391–395.<br />

182. Leichtmann GA, Bengoa JM, Bolt MJ, Sitrin MD.<br />

Intestinal absorption of cholecalciferol and 25-hydroxycholecalciferol<br />

in patients with both Crohn’s disease and<br />

intestinal resection. Am J Clin Nutr 1991; 54: 548–552.<br />

183. Goldschmid S, Graham M. Trace element deficiencies in<br />

inflammatory bowel disease. Gastroenterol Clin North<br />

Am 1989; 18: 579–587.<br />

184. Heatley RV. Assessing nutritional state in inflammatory<br />

bowel disease. Gut 1986; 27 (Suppl 1): 61–66.<br />

185. Sjogren A, Floren CH, Nilsson A. Evaluation of magnesium<br />

status in Crohn’s disease as assessed by intracellular<br />

analysis and intravenous magnesium infusion.<br />

Scand J Gastroenterol 1988; 23: 555–561.<br />

186. Hessov I, Hasselblad C, Fasth S, Hulten L. Magnesium<br />

deficiency after ileal resections for Crohn’s disease.<br />

Scand J Gastroenterol 1983; 18: 643–649.<br />

187. Galland L. Magnesium and inflammatory bowel disease.<br />

Magnesium 1988; 7: 78–83.<br />

188. Loeschke K, Konig A, Trebert Haeberlin S, Lux F. Low<br />

blood selenium concentration in Crohn disease. Ann<br />

Intern Med 1987; 106: 908.<br />

189. Hinks LJ, Inwards KD, Lloyd B, Clayton B. Reduced<br />

concentrations of selenium in mild Crohn’s disease. J<br />

Clin Pathol 1988; 41: 198–201.<br />

190. Ringstad J, Kildebo S, Thomassen Y. Serum selenium,<br />

copper, and zinc concentrations in Crohn’s disease and<br />

ulcerative colitis. Scand J Gastroenterol 1993; 28:<br />

605–608.<br />

191. Rannem T, Ladefoged K, Hylander E et al. Selenium<br />

status in patients with Crohn’s disease. Am J Clin Nutr<br />

1992; 56: 933–937.<br />

192. Kelly DG, Fleming CR. Nutritional considerations in<br />

inflammatory bowel diseases. Gastroenterol Clin North<br />

Am 1995; 24: 597–598.<br />

193. Hendricks KM, Walker WA. Zinc deficiency in inflammatory<br />

bowel disease. Nutr Rev 1988; 46: 401–408.<br />

194. Hyams JS, Ferry GD, Mandel FS et al. Development and<br />

validation of a pediatric Crohn’s disease activity index. J<br />

Pediatr Gastroenterol Nutr 1991; 12: 439–447.<br />

195. Hyams JS, Mandel F, Ferry GD et al. Relationship of<br />

common laboratory parameters to the activity of Crohn’s<br />

disease in children. J Pediatr Gastroenterol Nutr 1992;<br />

14: 216–222.<br />

196. Otley A, Loonen H, Parekh N et al. Assessing activity of<br />

pediatric Crohn’s disease: which index to use?<br />

Gastroenterology 1999; 116: 527–531.<br />

197. Loonen HJ, Griffiths AM, Merkus MP, Derkx HH. A critical<br />

assessment of items on the Pediatric Crohn’s Disease<br />

Activity Index. J Pediatr Gastroenterol Nutr 2003; 36:<br />

90–95.<br />

198. Kundhal PS, Critch JN, Zachos M et al. Pediatric Crohn<br />

Disease Activity Index: responsive to short-term change.<br />

J Pediatr Gastroenterol Nutr 2003; 36: 83–89.


24<br />

Introduction<br />

Indeterminate colitis<br />

Barbara S Kirschner<br />

The majority of patients with chronic inflammatory<br />

bowel disease (IBD) are diagnosed with either<br />

ulcerative colitis or Crohn’s disease on the basis of<br />

established clinical, endoscopic, histological and<br />

radiological criteria. 1 However, in 5–23% of<br />

patients with chronic colitis, a definitive diagnosis<br />

of ulcerative colitis or Crohn’s disease cannot be<br />

established because the initial colonoscopic and<br />

histological features overlap between ulcerative<br />

colitis and Crohn’s disease. 2–6 While the condition<br />

of most of these patients eventually evolves into<br />

patterns consistent with ulcerative colitis or<br />

Crohn’s disease, approximately 20–60% retain the<br />

diagnosis of indeterminate colitis over periods as<br />

long as 5–10 years after the diagnosis. 3,5,7–11 This<br />

latter observation suggests that indeterminate<br />

colitis may constitute a separate category within<br />

the spectrum of IBD. This chapter reviews the clinical<br />

features of indeterminate colitis, focusing<br />

primarily on the pediatric population.<br />

Table 24.1 Prevalence of indeterminate colitis (IC)<br />

Epidemiological aspects of<br />

indeterminate colitis<br />

Most children with indeterminate colitis are evaluated<br />

because of recurrent abdominal pain and<br />

diarrhea, with a smaller number noting hematochezia.<br />

12 In young children (less than 5 years of<br />

age), failure to thrive is more prominent than seen<br />

in ulcerative colitis. 13 Hassan et al noted no difference<br />

in gender, age at diagnosis or types of<br />

symptom at presentation among the 38 newly<br />

diagnosed children with ulcerative colitis, indeterminate<br />

colitis and Crohn’s disease. 14<br />

The prevalence of indeterminate colitis in reported<br />

series of patients with IBD varies among centers<br />

from 5 to 23% 2–6 (Table 24.1). In our pediatric<br />

population of 428 children actively followed with<br />

IBD, 49 (11.4%) were diagnosed with indeterminate<br />

colitis 4 (Table 24.2). In 42.9% of those with<br />

indeterminate colitis, the histology ‘favored ulcerative<br />

colitis’ but these patients also had features of<br />

Total no. of Age Indeterminate Peak age (years)<br />

IBD patients group colitis patients IC UC<br />

Gupta et al 4 420 pediatric 51 (11.9%) 10 —<br />

Heikenen et al 5 91 pediatric 9 (10%) 7.8 9.7<br />

Hildebrand et al 3 132 pediatric 36 (27%) — —<br />

Stewenius et al 15 — — — 10–19 20–29<br />

Meucci et al 10 1113 adult 50 (4.6%) — —<br />

Shivananda et al 2 2201 adult 116 (5.3%) — —<br />

IBD, inflammatory bowel disease; UC, ulcerative colitis<br />

379


380<br />

Indeterminate colitis<br />

Table 24.2 Indeterminate colitis: histological<br />

features in 49 pediatric patients at the<br />

University of Chicago (from reference 4)<br />

‘Favor ulcerative colitis’: 43% – except for the<br />

presence of:<br />

Focal colitis<br />

Colonic granulomas adjacent to ruptured crypts<br />

Gastroduodenal inflammation (<strong>Helicobacter</strong> <strong>pylori</strong><br />

negative)<br />

Anal fissure<br />

‘Favor Crohn’s disease’: 18% – except for findings<br />

of:<br />

Absence of granulomas<br />

No small-bowel X-ray features of Crohn’s disease<br />

No distinguishing features: 39%<br />

Crohn’s disease including areas of focal colitis,<br />

focal gastric or duodenal inflammation, anal<br />

fissures or isolated granulomas adjacent to<br />

ruptured crypts. Features ‘favoring Crohn’s<br />

disease’ were present in 20.4% of children with<br />

indeterminate colitis, none of whom had granulomas,<br />

radiological evidence of small-bowel Crohn’s<br />

disease or perianal findings. Endoscopic and histological<br />

findings of IBD without distinguishing<br />

features of ulcerative colitis or Crohn’s disease<br />

were present in 36.7% of our patients with indeterminate<br />

colitis.<br />

Heikenen et al noted a similar prevalence of indeterminate<br />

colitis (10%) in a pediatric population<br />

of IBD. 5 These authors noted that children with<br />

indeterminate colitis were diagnosed at a younger<br />

age (7.8 years) than those with either ulcerative<br />

colitis (9.7 years) or Crohn’s disease (11.4 years).<br />

Similarly, in Sweden, the peak age range at diagnosis<br />

was younger (10–19 years) for patients with<br />

indeterminate colitis in comparison with ulcerative<br />

colitis (20–29 years). 15 A particularly high<br />

prevalence of indeterminate colitis (23%) has been<br />

reported from one pediatric center in Sweden. 3<br />

Criteria for histological diagnosis<br />

In establishing the diagnosis of indeterminate<br />

colitis, it is essential to exclude other causes of<br />

colitis such as infections (Clostridium difficile,<br />

Yersinia, Mycobacterium tuberculosis, Entamoeba<br />

histolytica, Escherichia coli 0157:H7 or other verocytotoxin-producing<br />

strains), drugs (non-steroidal<br />

anti-inflammatory drugs (NSAIDs)), Behçet’s<br />

disease, malignancy, vasculitis and other identifiable<br />

causes of colitis.<br />

Riddell stated that to differentiate indeterminate<br />

colitis lesions from Crohn’s disease lesions such as<br />

submucosal and subserosal lymphoid aggregates<br />

away from areas of ulceration, non-necrotizing<br />

granulomas and skip areas should be absent. 16<br />

This is particularly true where there is nonspecific<br />

ileal involvement or gastritis and special<br />

stains for Helibacter <strong>pylori</strong> are negative. The most<br />

frequently used diagnostic study for distinguishing<br />

indeterminate colitis from Crohn’s disease (in<br />

addition to ileocolonoscopy with biopsy) is the<br />

small-bowel X-ray.<br />

The endoscopic and histological findings differ<br />

from classic ulcerative colitis in that there may be<br />

relative rectal sparing, focal inflammation or deep<br />

fissuring ulceration. The latter is most common in<br />

patients with fulminant colitis. It is also important<br />

to recognize that some medications, such as corticosteroids<br />

or aminosalicylic acid preparations,<br />

may change the diffuse histological appearance in<br />

ulcerative colitis to a more focal appearance. 6,16<br />

Thus, slides from the original or pretreatment<br />

colonoscopy should be reviewed when considering<br />

a diagnosis of indeterminate colitis.<br />

Although the above criteria would appear to allow<br />

differentiation between Crohn’s disease and ulcerative<br />

colitis on the basis of pathology, Farmer et al<br />

documented disparity among pathologists reviewing<br />

cases of colonic IBD. 17 The diagnoses of<br />

gastrointestinal pathologists differed from that of<br />

the referring institution in 45% of surgical<br />

specimens and 54% of biopsy specimens. Of 70<br />

cases initially diagnosed with ulcerative colitis, 30<br />

(43%) were changed to Crohn’s disease or indeterminate<br />

colitis; in contrast, 17% of cases initially<br />

diagnosed with Crohn’s disease were changed to<br />

ulcerative colitis or indeterminate colitis.<br />

The performance of upper gastrointestinal<br />

endoscopy and biopsy (EGD) identifies some<br />

patients with Crohn’s disease whose colonoscopy<br />

biopsies were indeterminate. 12,18 Kundhal et al<br />

reported that, while diffuse non-specific gastritis<br />

occurred with similar frequency in children with


either Crohn’s disease or ulcerative colitis (92% vs.<br />

75%), focal antral gastritis was significantly more<br />

common in Crohn’s disease than ulcerative colitis<br />

(52% vs. 8%). 12 The authors defined focal inflammation<br />

as ‘localized inflammation of the gastric<br />

pits, glands, or foveolae by mononuclear and polymorphonuclear<br />

leukocytes bordering directly on<br />

uninflamed mucosa’. This is similar to the statements<br />

previously published by Riddell. 16<br />

Granulomas in the stomach or duodenum<br />

provided evidence confirming the diagnosis of<br />

Crohn’s disease even in endoscopically normalappearing<br />

mucosa. 12 Hence, performing an EGD<br />

should be strongly considered during the initial<br />

evaluation of patients for IBD. In order to further<br />

categorize our patients with indeterminate colitis<br />

further, an X-ray study of the upper gastrointestinal<br />

tract with small-bowel follow-through was<br />

performed in all patients to exclude the possibility<br />

of Crohn’s disease. EGD with biopsy may be particularly<br />

useful in those children whose symptoms<br />

(such as nausea, vomiting, early satiety) suggest<br />

gastroduodenal Crohn’s disease. 18<br />

Serologic markers and defining<br />

inflammatory bowel disease<br />

categories<br />

We are currently evaluating the role of perinuclear<br />

anti-neutrophil cytoplasmic antibodies (P-<br />

ANCA) and anti-Saccharomyces cerevisiae antibodies<br />

(ASCA) in identifying patients with<br />

ulcerative colitis and Crohn’s disease previously<br />

diagnosed with indeterminate colitis. 4 While P-<br />

ANCA was positive in 68% of those favoring ulcerative<br />

colitis, and ASCA was positive in 37% of<br />

those favoring Crohn’s disease, 86% of our patients<br />

with indeterminate colitis were both p-ANCA and<br />

ASCA negative.<br />

Recently, Joossens et al correlated serological<br />

markers with prospective follow-up evaluation in<br />

97 adult patients with indeterminate colitis. 8 After<br />

a mean follow-up of 6 years, 32% of the adult<br />

patients with indeterminate colitis were reclassified<br />

as having ulcerative colitis or Crohn’s disease,<br />

half of whom were positive for p-ANCA or ASCA.<br />

However, almost half of the patients with indeterminate<br />

colitis (48.5%) remained p-ANCA/ASCA<br />

negative and continued to have characteristics of<br />

Serologic markers and defining IBD categories 381<br />

indeterminate colitis even 10 years after the initial<br />

diagnosis.<br />

Radiological imaging<br />

In a further attempt to distinguish between the<br />

various forms of pediatric IBD, 99 m-technetium<br />

white cell scans and magnetic resonance imaging<br />

have been evaluated in children with indeterminate<br />

colitis, ulcerative colitis and Crohn’s<br />

disease. 18–20 The technetium scan had a sensitivity<br />

of only 76% compared with colonoscopy and<br />

biopsy. 19 Others found that conventional magnetic<br />

resonance imaging had a low sensitivity (40%) for<br />

detecting Crohn’s disease and did not correlate<br />

with the severity of inflammation. 20 However,<br />

gadolinium-enhanced magnetic resonance imaging<br />

in combination with oral polyethylene glycol<br />

(PEG) solution (used to distend the small bowel)<br />

may be more discriminating. 21 With the latter technique,<br />

increased wall thickness was noted in 26/26<br />

children with Crohn’s disease while those with<br />

indeterminate colitis and ulcerative colitis showed<br />

mild parietal contrast enhancement but not bowel<br />

wall thickening. Further confirmation of these<br />

observations is needed.<br />

Natural history<br />

With time, 50–72% of adult patients and 64% of<br />

pediatric patients with indeterminate colitis can<br />

be reclassified as having definite ulcerative colitis<br />

or Crohn’s disease during subsequent observation<br />

3–4,9-11,22 (Table 24.3). After a mean follow-up<br />

period of 14 months, one group reported that 33%<br />

of 36 patients with indeterminate colitis were<br />

reclassified as ulcerative colitis and 17% as<br />

Crohn’s disease. 9 Meucci et al reported that 37 of<br />

50 patients (74%) changed from indeterminate<br />

colitis to a definite diagnosis of ulcerative colitis or<br />

Crohn’s disease during follow-up with a cumulative<br />

probability of 80% within 8 years of diagnosis.<br />

10 In contrast, Wells et al followed 16 patients<br />

with indeterminate colitis for a mean of 10 years<br />

and observed that three were reclassified with<br />

ulcerative colitis, one with Crohn’s disease and the<br />

rest remained indeterminate. 11 The course of indeterminate<br />

colitis in 36 Swedish children after a<br />

mean follow-up of 4.6 years was analyzed by<br />

Hildebrand et al. 3 The findings were similar to


382<br />

Indeterminate colitis<br />

Table 24.3 Indeterminate colitis (IC): changes in diagnosis with time<br />

those as described above in adults: 21/36 (58%)<br />

were subsequently categorized as ulcerative<br />

colitis, 2/36 (6%) as Crohn’s disease and 13/36<br />

(36%) remaining as indeterminate colitis. In our<br />

report describing 49 children with indeterminate<br />

colitis, nine had undergone colectomy at a mean<br />

of 24 months after diagnosis. During a mean<br />

follow-up period of 42 months, 6/9 (66%) had a<br />

subsequent course, including repeat endoscopic<br />

examination consistent with ulcerative colitis,<br />

3/9 (33%) as indeterminate colitis and 1/9 (11%)<br />

as Crohn’s disease.<br />

Medical therapy<br />

The observation that the majority of patients<br />

with indeterminate colitis over time are reclassified<br />

as ulcerative colitis or Crohn’s disease<br />

makes it difficult to know whether indeterminate<br />

colitis represents a separate form of IBD. Perhaps<br />

because of the small number of patients with<br />

indeterminate colitis, the response to various<br />

drug regimens in this population has not been<br />

specifically addressed. 7 In our program the<br />

choice of therapeutic intervention is selected<br />

depending on the severity of symptoms, extent<br />

and severity of endoscopic and histological findings<br />

and laboratory parameters 23 (Table 24.4). For<br />

most patients, drug therapy is similar to that<br />

indicated for patients with ulcerative colitis of<br />

Diagnosis<br />

Initial Final Final Final<br />

IC IC (%) UC (%) CD (%)<br />

Hildebrand et al3 36 (pediatric) 36 58 6<br />

Moum et al9 36 (adult) 50 33 17<br />

Meucci et al10 50 (adult) 20 34 40<br />

Kangas et al22 6 (surgery) 50 — 50<br />

Wells et al11 16 (surgery) 75 19 6<br />

Gupta et al4 12 (surgery/pediatric) 33 50 17<br />

UC, ulcerative colitis; CD, Crohn’s disease<br />

comparable extent and severity. These include 5aminosalicylic<br />

acid (5-ASA) preparations for<br />

mild disease and corticosteroids and<br />

immunomodulatory therapy for moderate and<br />

severe disease. However, we are more likely to<br />

use metronidazole in this population, especially<br />

where there is extensive focal colitis or those<br />

‘favoring’ Crohn’s disease. Immunomodulatory<br />

agents, such as azathioprine or 6-mercaptopurine,<br />

are used in approximately 60% of our<br />

pediatric population with IBD, owing to the presence<br />

of steroid dependency, resistance or toxicity.<br />

24<br />

Many large series involving clinical trials of<br />

adult patients with IBD have included small<br />

numbers of patients with indeterminate colitis,<br />

although they have not specifically addressed<br />

the treatment of indeterminate colitis. 7 The<br />

response of adult patients with refractory ulcerative<br />

colitis and indeterminate colitis appears to<br />

show similar improvement to azathioprine/6mercaptopurine<br />

and cyclo-sporin as well as<br />

newer immunomodulatory agents such tacrolimus<br />

and thalidomide. 25–27 The role of infliximab<br />

in indeterminate colitis is as yet to be<br />

determined. However, favorable outcomes in<br />

pediatric patients with ulcerative colitis suggest<br />

that it could be considered in patients with indeterminate<br />

colitis who are not responding to<br />

conventional medications. 28


Table 24.4 Medical approach to the pediatric patient with indeterminate colitis<br />

5-Aminosalicylic acid preparations for mild disease<br />

Corticosteroids (moderate-to-severe disease severity)<br />

Metronidazole if histology favors Crohn’s disease<br />

Azathioprine (AZA) or 6-mercaptopurine (6-MP)<br />

Surgical treatment<br />

Considerable conflicting data have been published<br />

regarding complication rates following medical<br />

and surgical intervention in patients with indeterminate<br />

colitis versus ulcerative colitis. Some<br />

centers reported that indeterminate colitis was<br />

more refractory to medical interventions than<br />

ulcerative colitis, resulting in a greater relapse<br />

rate 29 and subsequent need for colectomy. 30<br />

Colectomy rates averaged 36.1/1000 person-years<br />

in patients with indeterminate colitis versus only<br />

7.5 for those with definite ulcerative colitis. 30 In<br />

contrast, Witte et al described similar response<br />

rates to medical intervention in indeterminate<br />

colitis and ulcerative colitis. 31 The European<br />

Collaborative Study on Inflammatory Bowel<br />

Disease (EC-IBD) reported ‘complete relief of<br />

complaints’ in 48% of ulcerative colitis patients<br />

versus 50% of those with indeterminate colitis; in<br />

addition, 37% of patients with ulcerative colitis<br />

‘improved’ versus 33% with indeterminate<br />

colitis. 31 Similarly, higher rates of pouch failure<br />

after ileal pouch–anal anastomosis (IPPA) 32,33 and<br />

colorectal cancer 34 have been reported by some<br />

groups, while others have reported lower rates of<br />

pouchitis for indeterminate colitis (29%) than<br />

Surgical treatment 383<br />

Consider measuring red blood cell 6-thioguanine levels to reduce the risk of toxicity if increasing the dose in<br />

refractory patients<br />

Change to AZA from 6-MP (or vice versa) for non-hypersensitivity side-effects (e.g. rash, arthralgia, headache)<br />

Methotrexate (parenterally) if intolerant or refractory to AZA/6-MP<br />

Cyclosporin intravenously or micro-emulsified oral formulation<br />

Tacrolimus (0.1–0.15mg/kg twice a day); monitor blood level<br />

Thalidomide (50–100mg/day in older children and adolescents)<br />

Remicade (at conventional dosing)<br />

ulcerative colitis (58%) and Crohn’s disease (72%).<br />

However, this latter group observed a greater<br />

frequency of fistulae after IPAA in patients with<br />

indeterminate colitis (26%) versus for ulcerative<br />

colitis (10%). 35 Post-IPAA complications resulting<br />

in pouch removal were higher for indeterminate<br />

colitis (19–28%) versus ulcerative colitis (0.4–8%)<br />

in some studies 32,33 but not in others. 35–37<br />

It is our practice to repeat colonoscopy, usually<br />

with concurrent EGD, during selected periods of<br />

relapse to assess whether histological changes<br />

consistent with ulcerative colitis or Crohn’s<br />

disease have developed. This is especially the case<br />

if colectomy and IPAA are being considered<br />

because of refractory disease. At this time, repeat<br />

small-bowel X-ray is usually obtained so that<br />

patients with ileal Crohn’s disease would be<br />

excluded and patients with persistent<br />

indeterminate colitis would be counseled regarding<br />

the potentially greater risk of pouch complications.<br />

Often, patients with indeterminate colitis<br />

undergo a multi-staged operative procedure<br />

consisting of a subtotal colectomy with temporary<br />

ileostomy and Hartmann pouch. In this way, the<br />

entire resected colon can be assessed to exclude<br />

Crohn’s disease prior to creating the IPAA. 36


384<br />

Indeterminate colitis<br />

REFERENCES<br />

1. Marion JF, Rubin TH, Present DH. Differential diagnosis<br />

of chronic ulcerative colitis and Crohn disease. In<br />

Kirsner JB, ed. Inflammatory Bowel Disease, 5th edn.<br />

Philadelphia: WB Saunders, 2000: 315–325.<br />

2. Shivananda S, Lennard-Jones J, Logan R et al. Incidence<br />

of inflammatory bowel disease across Europe: is there a<br />

difference between north and south? Results of the<br />

European Collaborative Study on Inflammatory Bowel<br />

Disease (EC-IBD). Gut 1996; 39: 690–697.<br />

3. Hildebrand H, Brydolf M, Holmquist L et al. Incidence<br />

and prevalence of inflammatory bowel disease in children<br />

in southwestern Sweden. Acta Paediatr 1994; 83:<br />

640–645.<br />

4. Gupta P, Hart J, Kirschner BS. Perinuclear antineutrophilic<br />

cytoplasmic antibodies (pANCA) and anti-<br />

Saccharomyces cerevisiae (ASCA) antibodies in children<br />

with indeterminate colitis (abstract). Gastroenterology<br />

2000; 118: 103, A687.<br />

5. Heikenen JB, Werlin SL, Brown CW et al. Presenting<br />

symptoms and diagnostic lag in children with inflammatory<br />

bowel disease. Inflamm Bowel Dis 1999; 5: 158–160.<br />

6. Kirschner BS, Heyman MB, Clemons T et al. The<br />

Pediatric IBD Consortium Database – initial demographic<br />

data. Gastroenterology 2002; 122: A–611.<br />

7. Geboes K, DeHertogh G. Indeterminate colitis. Inflamm<br />

Bowel Dis 2003; 9: 324–331.<br />

8. Joossens S, Reinisch W, Vermeire S et al. The value of<br />

serologic markers in indeterminate colitis: a prospective<br />

follow-up. Gastroenterology 2002; 122: 1242–1247.<br />

9. Moum B, Ekbom A, Vatn MH et al. Inflammatory bowel<br />

disease: re-evaluation of the diagnosis in a prospective<br />

population based study in south eastern Norway. Gut<br />

1997; 40: 328–332.<br />

10. Meucci G, Bortoli A, Riccioli FA et al. Frequency and<br />

clinical evolution of indeterminate colitis: a retrospective<br />

multi-centre study in northern Italy. GSMII (Gruppo<br />

di Studio per le Malattie Infiammatorie Intestinali). Eur J<br />

Gastroenterol Hepatol 1999; 11: 909–913.<br />

11. Wells AD, McMillan I, Price AB et al. Natural history of<br />

indeterminate colitis. Br J Surg 1991; 78: 179–181.<br />

12. Kundhal PS, Stormon MO, Zachos M et al. Gastric antral<br />

biopsy in the differentiation of pediatric colitides. Am J<br />

Gastroenterol 2003; 98: 557–561.<br />

13. Mamula P, Telega GW, Markowitz JE et al. Inflammatory<br />

bowel disease in children 5 years of age and younger.<br />

Am J Gastroenterol 2002; 97: 2005–2010.<br />

14. Hassan K, Cowan FJ, Jenkins HR. The incidence of<br />

childhood inflammatory bowel disease in Wales. Eur J<br />

Pediatr 2000; 159: 261–263.<br />

15. Stewenius J, Adnerhill I, Ekelund GR et al. Ulcerative<br />

colitis and indeterminate colitis in the city of Malmo,<br />

Sweden. A 25-year incidence study. Scand J<br />

Gastroenterol 1995; 30: 38–43.<br />

16. Riddell RH. Pathology of idiopathic inflammatory bowel<br />

disease. In Kirsner JB, ed. Inflammatory Bowel Disease,<br />

5th edn. Philadelphia: WB Saunders, 2000: 439–441.<br />

17. Farmer M, Petras RE, Hunt LE et al. The importance of<br />

diagnostic accuracy in colonic inflammatory bowel<br />

disease. Am J Gastroenterol 2000; 95: 3184–3188.<br />

18. Lenaerts C, Roy CC, Vaillancourt M et al. High incidence<br />

of upper gastrointestinal tract involvement in children<br />

with Crohn disease. Pediatrics 1989; 83: 777–781.<br />

19. Cucchiara S, Celentano L, deMagistris TM et al.<br />

Colonoscopy and technetium99m white cell scan in<br />

children with suspected inflammatory bowel disease. J<br />

Pediatr 1999; 135: 727–732.<br />

20. Durno CA, Sherman P, Williams T et al. Magnetic resonance<br />

imaging to distinguish the type and severity of<br />

pediatric inflammatory bowel diseases. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 170–174.<br />

21. Laghi A, Borrelli O, Paolantonio P et al. Contrast<br />

enhanced magnetic resonance imaging of the terminal<br />

ileum in children with Crohn disease. Gut 2000; 52:<br />

393–397.<br />

22. Kangas E, Matikainen M, Mattila J. Is ‘indeterminate<br />

colitis’ Crohn disease in the long-term follow-up? Int<br />

Surg 1994; 79: 120–123.<br />

23. Kirschner BS. Indeterminate colitis. Implications for<br />

management: medical approach. Inflamm Bowel Dis<br />

2000; 6: 516–517.<br />

24. Kirschner BS. The safety of azathioprine and 6-mercaptopurine<br />

in children and adolescents with inflammatory<br />

bowel disease. Gastroenterology 1998; 115: 813–821.<br />

25. Fraser AG, Orchard TR, Jewell DP. The efficacy of<br />

azathioprine for the treatment of inflammatory bowel<br />

disease: a 30-year review. Gut 2002; 50: 485–489.<br />

26. Fellerman K, Tanko Z, Herrlinger KR et al. Response of<br />

refractory colitis to intravenous or oral tacrolimus<br />

(FK506). Inflamm Bowel Dis 2002; 8: 317–324.<br />

27. Bariol C, Meagher AP, Vickers CR et al. Early studies on<br />

the safety and efficacy of thalidomide for symptomatic<br />

inflammatory bowel disease. J Gastroenterol Hepatol<br />

2002; 17: 135–139.<br />

28. Mamula P, Markowitz JE, Brown KA et al. Infliximab as a<br />

novel therapy for pediatric ulcerative colitis. J Pediatr<br />

Gastroenterol Nutr 2002; 34: 307–311.<br />

29. Stewenius J, Adnerhill I, Ekelund GR et al. Risk of<br />

relapse in new cases of ulcerative colitis and indeterminate<br />

colitis. Dis Colon Rectum 1996; 39: 1019–1025.<br />

30. Stewenius J, Adnerhill I, Ekelund GR et al. Operations in<br />

unselected patients with ulcerative colitis and indeterminate<br />

colitis. A long-term follow-up study. Eur J Surg<br />

1996; 162: 131–137.<br />

31. Witte J, Shivananda S, Lennard-Jones et al. Disease<br />

outcome in inflammatory bowel disease: mortality,<br />

morbidity and therapeutic management of 796-person<br />

inception cohort in the European Collaborative Study on<br />

Inflammatory Bowel Disease (EC-IBD). Scand J<br />

Gastroenterol 2000; 35: 1272–1277.<br />

32. Koltun WA, Schoetz DJJ Jr, Roberts PL et al.<br />

Indeterminate colitis predisposes to perineal complications<br />

after ileal pouch–anal anastomosis. Dis Colon<br />

Rectum 1991; 34: 857–860.<br />

33. McIntyre PB, Pemberton JH, Wolff BG et al.<br />

Indeterminate colitis. Long-term outcome in patients<br />

after ileal pouch–anal anastomosis. Dis Colon Rectum<br />

1995; 38: 51–54.<br />

34. Stewenius J, Adnerhill I, Ekelund GR et al. Incidence of<br />

colorectal cancer and all cause mortality in non-selected<br />

patients with ulcerative colitis and indeterminate colitis<br />

in Malmo, Sweden. Int J Colorectal Dis 1995; 10:<br />

117–122.<br />

35. Rudolph WG, Uthoff SM, McAuliffe TL et al.<br />

Indeterminate colitis: the real story. Dis Colon Rectum<br />

2002; 45: 1528–1534.<br />

36. Marcello PW, Schoetz DJ, Rogers PL et al. Evolutionary<br />

changes in the pathologic diagnosis after the ileoanal<br />

pouch procedure. Dis Colon Rectum 1997; 40: 263–269.<br />

37. Gramlich T, Delaney CP, Lynch AC et al. Pathological<br />

subgroups may predict complications but not late failure<br />

after ileal pouch–anal anastomosis for indeterminate<br />

colitis. Colorectal Dis 2003; 5: 315–319.


25<br />

Introduction<br />

Ulcerative colitis<br />

Leslie M Higuchi and Athos Bousvaros<br />

Ulcerative colitis and Crohn’s disease are the two<br />

most common forms of idiopathic inflammatory<br />

bowel disease (IBD). Ulcerative colitis differs from<br />

Crohn’s disease in that the inflammation in ulcerative<br />

colitis is confined to the mucosal layer of the<br />

colon. In contrast, Crohn’s disease is characterized<br />

by transmural inflammation in either a limited<br />

region or extensively in the bowel, and may<br />

involve any portion of the gastrointestinal tract<br />

from the mouth to the anus. The peak incidence of<br />

IBD occurs between the ages of 15 and 25 years,<br />

but ulcerative colitis may begin at any age. 1<br />

Approximately 20% of patients with ulcerative<br />

colitis present before the age of 20 years. 2 While<br />

children and adults develop similar symptoms,<br />

children often present with more extensive<br />

disease. 3 Clinicians caring for children and adolescents<br />

with ulcerative colitis must treat both the<br />

gastrointestinal and extraintestinal complications,<br />

optimize nutrition and linear growth, and address<br />

the psychosocial ramifications of the illness. Since<br />

the majority of the published studies investigating<br />

the natural history and treatment of ulcerative<br />

colitis are in adults, we refer primarily to these<br />

studies, with reference to pediatric studies where<br />

available.<br />

Epidemiology<br />

Most children with ulcerative colitis present<br />

between the ages of 10 and 18 years. However,<br />

ulcerative colitis in children under the age of 5<br />

years is well described. 1,4,5 Epidemiological<br />

studies primarily conducted in the US, GB, and<br />

Scandinavian countries, suggest that the incidence<br />

of ulcerative colitis in children ranges from 1.4 to<br />

4.3 cases/100000 population per year. 3,6–10 The<br />

incidence of ulcerative colitis in children has<br />

remained relatively stable 11 (Table 25.1). The<br />

majority of incidence data for Crohn’s disease and<br />

ulcerative colitis in the pediatric population originates<br />

from geographic regions with higher rates of<br />

IBD.<br />

Adult studies demonstrate that ulcerative colitis is<br />

more prevalent in North America, the UK, and<br />

Scandinavia and less common in southern Europe,<br />

Asia and Africa. 2 The data suggest a north–south<br />

gradient with higher incidence rates of both<br />

Crohn’s disease and ulcerative colitis in northern<br />

locations, even within individual countries. 12,13<br />

Ulcerative colitis is more common among Jewish<br />

than non-Jewish peoples, 14 but disease rates in<br />

people of Jewish origin vary by geographic region<br />

and parallel those of the general population. 15 The<br />

higher rates of IBD in individuals of Jewish origin<br />

across different countries support a common<br />

genetic predisposition; however, the geographic<br />

variation of IBD rates in Jews emphasizes that<br />

environmental factors (see below) influence the<br />

inherited risk.<br />

Etiology and pathogenesis<br />

The pathogenesis of ulcerative colitis is unknown.<br />

A widely accepted hypothesis suggests that, in the<br />

genetically susceptible individual, a combination<br />

of host and environmental factors leads to the<br />

initiation and perpetuation of an abnormal intestinal<br />

immune response, resulting in ulcerative<br />

colitis. 16 In support of this theory, colitis in<br />

animals occurs in a wide variety of genetically<br />

altered rodents, including: knockout mice for<br />

interleukin (IL)-2, IL-10 and T-cell receptor; and<br />

HLA-B27 transgenic rats. 17 Interestingly, most of<br />

these animal models do not develop colitis in<br />

385


386<br />

Ulcerative colitis<br />

germ-free environments. These findings suggest<br />

that multiple genes may contribute to the pathogenesis<br />

of IBD, and that interaction with the environment<br />

is essential.<br />

In humans, ulcerative colitis appears to have a<br />

non-Mendelian pattern of inheritance. Current<br />

evidence suggests that genes comntribute less to<br />

the risk in individuals with ulcerative colitis than<br />

in those with Crohn’s disease. In a Danish twin<br />

cohort study and a recently updated Swedish twin<br />

cohort study, the calculated pair concordance rates<br />

among monozygotic twins were 14–19% for ulcerative<br />

colitis and 50% for Crohn’s disease; and<br />

among dizygotic twins, 0–5% for ulcerative colitis<br />

and 0–4% for Crohn’s disease. 18,19 Although the<br />

concordance rates of disease are higher in monozygotic<br />

twins, the incomplete concordance suggests<br />

that non-genetic factors also contribute to the<br />

development of ulcerative colitis. First-degree relatives<br />

of patients with ulcerative colitis have a 9.5fold<br />

increase in risk of developing ulcerative<br />

colitis, compared to the general population. 20 The<br />

risk of developing Crohn’s disease or ulcerative<br />

colitis is increased in children of affected parents<br />

with IBD. In one study, the life-time risks for IBD<br />

in offspring ranged from 5–8% when one parent<br />

had Crohn’s disease, and from 2–5% when one<br />

parent had ulcerative colitis. 21 In another study of<br />

families with two or more affected family members<br />

with IBD, there was a high concordance for the<br />

diagnosis of ulcerative colitis or Crohn’s disease. 22<br />

Of 83 families in which the proband had ulcerative<br />

colitis, 72 relatives had ulcerative colitis (81%,<br />

72/89) and 17 had Crohn’s disease (19%, 17/89). 22<br />

Family linkage studies suggest that candidate<br />

genes may be present on chromosomes 3, 7 and 12,<br />

with the strongest association identified on chromosome<br />

12. 23 Associations with certain HLA<br />

alleles (especially DR2) have been described. 16,24,25<br />

However, no specific gene or protein has been<br />

identified. 17<br />

Environmental factors may contribute to the development<br />

of ulcerative colitis. However, most<br />

studies examining environmental risk factors have<br />

the limitations of retrospective, case–control<br />

methodology. One of the most consistent findings<br />

in multiple studies is the lower risk of ulcerative<br />

colitis among current smokers. Current smokers<br />

have approximately one-half the risk of developing<br />

ulcerative colitis compared to non-smokers. 26,27<br />

Table 25.1 Incidence of ulcerative colitis (UC) in children and adolescents (no. of new cases/<br />

100000 persons per year)<br />

First author Location Age range (years) Time period studied Incidence<br />

Barton, 1989 6 Scotland >5 and ≤ 16 for UC 1968–83<br />

1968 1.9<br />

1983 1.6<br />

Calkins, 1984 7 Baltimore, MD, USA


Several authors have suggested that exposure to<br />

infections in the perinatal period or early life may<br />

contribute to the development of ulcerative colitis.<br />

Individuals with Crohn’s disease or ulcerative<br />

colitis were more likely to have experienced a diarrheal<br />

illness during infancy, when compared to<br />

their unaffected siblings. 28 Appendectomy at a<br />

young age is associated with a lower risk of ulcerative<br />

colitis. 29–31 In a population-based study, the<br />

inverse relation between appendectomy and ulcerative<br />

colitis was observed only in those individuals<br />

who underwent surgery before the age of 20<br />

years. 29 Interestingly, the risk of ulcerative colitis<br />

was reduced only in patients who had an appendectomy<br />

performed for inflammatory conditions,<br />

such as appendicitis or mesenteric lymphadenitis,<br />

but not in patients who underwent appendectomy<br />

for non-specific abdominal pain. 29 These findings<br />

may suggest that the inflammatory condition that<br />

preceded the appendectomy, rather than the<br />

appendectomy itself, may be inversely related to<br />

the development of ulcerative colitis later in life. 29<br />

Some published reports suggest that non-steroidal<br />

anti-inflammatory drugs (NSAIDs) may precede<br />

the onset of IBD, lead to a reactivation of quiescent<br />

IBD, or exacerbate already active IBD in<br />

humans. 32–35 In an adult case–control study,<br />

Felder et al compared the use of NSAIDs in 60<br />

patients hospitalized for exacerbations of IBD,<br />

either ulcerative colitis or Crohn’s disease, to that<br />

of 62 out-patients with irritable bowel<br />

syndrome. 36 At least 31% of the IBD patients (35%<br />

of the ulcerative colitis patients) who used<br />

NSAIDs within 1 month of symptoms developed<br />

the onset or an exacerbation of IBD, whereas only<br />

2% of the irritable bowel patients experienced<br />

aggravation of their symptoms after similar<br />

NSAID use. In a retrospective review of initial<br />

office visit records of adult out-patients with<br />

either Crohn’s disease (112 initial visits) or ulcerative<br />

colitis (80 initial visits), Bonner et al did not<br />

find any association between NSAID use and<br />

active IBD. 37 Also, another short report suggested<br />

the possible exacerbation of IBD activity associated<br />

with the use of celecoxib, a selective cyclooxygenase<br />

(COX)-2 inhibitor, in two patients, one<br />

with ulcerative colitis and one with Crohn’s<br />

disease. 35 Studies suggest that both standard<br />

NSAIDs and selective COX-2 inhibitors can exacerbate<br />

colitis in animal models of colonic inflammation.<br />

38,39 It has been postulated that NSAIDs<br />

Etiology and pathogenesis 387<br />

may worsen colitis by inhibiting the synthesis of<br />

colonic prostaglandins that may exert anti-inflammatory<br />

effects in the setting of colonic inflammation.<br />

40 Further study is needed to clarify any true<br />

association of NSAID therapy with the onset of<br />

ulcerative colitis, to identify those individuals at<br />

most risk, and to determine the safety profile of<br />

COX-2 inhibitors in comparison to standard<br />

NSAIDs in reference to potential colonic mucosal<br />

injury. In general, given the current data, it is<br />

advisable for children with ulcerative colitis to<br />

avoid NSAID therapy if there are other reasonable<br />

alternatives for pain control (e.g. acetaminophen).<br />

In patients with arthritis, alternative anti-inflammatory<br />

medications (e.g. sulfasalazine) should be<br />

considered.<br />

Given the constant intestinal exposure to numerous<br />

luminal dietary antigens, it seems reasonable<br />

to postulate a relationship between diet and ulcerative<br />

colitis. The data on the association of breast<br />

feeding and ulcerative colitis are inconclusive,<br />

with one published study suggesting a protective<br />

effect, 41 and two other studies showing no<br />

effect. 28,42 Ecological and case–control studies<br />

have examined the association between certain<br />

foods and ulcerative colitis, 43,44 but at present,<br />

there is no definitive evidence linking diet to the<br />

development of ulcerative colitis. Methodological<br />

issues of these retrospectively designed dietary<br />

studies, such as the collection of pre-illness<br />

dietary information, remain a problem in the<br />

interpretation of findings. As an example, the<br />

recalled dietary intake may reflect changes in the<br />

diet secondary to the effects of the disease itself,<br />

rather than the individual’s dietary habits preceding<br />

the onset of disease. The association between<br />

oral contraceptive use and ulcerative colitis<br />

remains controversial. 26,45<br />

Current evidence suggests that colitis results<br />

when the intestinal mucosal immune system in<br />

patients with inflammatory bowel disease reacts<br />

inappropriately to intestinal bacteria. Evidence<br />

supporting this hypothesis includes the fact that<br />

most animal models of colitis do not develop<br />

disease in germ-free environments, the reports of<br />

efficacy of antibiotics in the treatment of colitis<br />

and pouchitis, and the seasonal variations in the<br />

onset of ulcerative colitis. However, no specific<br />

infectious agent has consistently been associated<br />

with exacerbations of ulcerative colitis. 17,46


388<br />

Ulcerative colitis<br />

Activated cells of the mucosal immune system are<br />

present in the bowel of both Crohn’s disease and<br />

ulcerative colitis patients. There is some evidence<br />

that Crohn’s disease and ulcerative colitis are characterized<br />

by different mucosal immune responses,<br />

suggesting that different subsets of T cells may be<br />

involved. Crohn’s disease is characterized by<br />

increased secretion of the cytokines IL-2, interferon-γ,<br />

tumor necrosis factor (TNF)-α, and IL-12,<br />

suggesting a Th1 pattern of cytokine response. 47,48<br />

In contrast, in ulcerative colitis, the lamina propria<br />

T cells secrete increased amounts of IL-5 and<br />

possibly IL-13 and there is increased production<br />

by B cells of IgG1, suggesting a Th2 pattern of<br />

cytokine response. 46,29 Such differences may<br />

explain why anti-TNF therapies, such as infliximab,<br />

have thus far failed to show the same degree<br />

of efficacy in ulcerative colitis as in Crohn’s<br />

disease. Patients with active ulcerative colitis also<br />

have increased levels of the chemokine IL-8 in<br />

biopsy specimens as well as in rectal<br />

dialysates. 50–52 IL-8 and other chemokines facilitate<br />

the recruitment and transmigration of<br />

neutrophils across mucosal surfaces. 53,54 These<br />

neutrophils in turn produce prostaglandins and<br />

leukotrienes, which cause pain, diarrhea and<br />

further intestinal inflammation. 55 Thus, alterations<br />

in function of immune system cells, particularly<br />

regulatory CD4 lymphocytes, probably<br />

result in aberrant responses to bacterial antigens,<br />

antibody production and cytotoxicity against gut<br />

epithelium, and recruitment of inflammatory cells<br />

into the colonic crypts.<br />

Clinical signs and symptoms<br />

The typical symptoms of ulcerative colitis include<br />

rectal bleeding, diarrhea and abdominal pain. The<br />

presentation can vary depending upon the extent<br />

of colonic involvement and the severity of inflammation.<br />

The colon in ulcerative colitis is inflamed<br />

in a diffuse, continuous distribution, extending<br />

from the rectum proximally. By convention, ulcerative<br />

colitis is classified according to the extent of<br />

disease into the following three subgroups: proctitis<br />

(disease limited to the rectum), left-sided colitis<br />

(disease extending to the sigmoid or descending<br />

colon, but not past the splenic flexure) and pancolitis<br />

(disease extending past the splenic flexure).<br />

Proctitis may present with tenesmus, urgency and<br />

the passage of formed or semi-formed stool with<br />

blood and mucus. 56 In contrast, pancolitis or leftsided<br />

disease may present with bloody diarrhea<br />

and significant abdominal pain. The majority of<br />

patients will present with a history of symptoms<br />

for several weeks; however, some will present with<br />

a more acute clinical picture.<br />

Although adults and children with ulcerative<br />

colitis can present with similar symptoms, there<br />

are differences in the clinical presentation of these<br />

two populations. Studies suggest that children<br />

with ulcerative colitis present with more extensive<br />

colonic involvement than adults with ulcerative<br />

colitis. 3,4,10 In a Danish study of 80 patients aged<br />

less than 15 years and 1081 patients aged 15 years<br />

or more with ulcerative colitis, the younger group<br />

had more extensive disease at diagnosis compared<br />

to the older group diagnosed with ulcerative<br />

colitis. 3 Of the ulcerative colitis patients younger<br />

than 15 years, 29% had pancolitis and 25% had<br />

proctitis; in contrast, of the ulcerative colitis<br />

patients 15 years or older, 16% had pancolitis and<br />

46% had proctitis. 3 Gryboski examined 38 children<br />

diagnosed with ulcerative colitis at ≤ 10 years<br />

old and reported 71% with pancolitis, 13% with<br />

left-sided colitis and 6% with proctitis. 4<br />

Approximately 5% of children with ulcerative<br />

colitis have evidence of delayed linear growth<br />

and/or weight loss at diagnosis, although growth<br />

failure is much less frequent than in children with<br />

Crohn’s disease 4,57 (Table 25.2).<br />

Table 25.2 Symptoms at initial presentation<br />

of ulcerative colitis in children and<br />

adolescents (from references 3, 4, 9, 57, 300)<br />

Clinical symptoms Range (%)<br />

Rectal bleeding 75–98<br />

Diarrhea 71–91<br />

Abdominal pain 44–92<br />

Weight loss 13–74<br />

Arthralgia/arthritis 5–9<br />

Fever 3–34<br />

Growth restriction 4–5


Children with ulcerative colitis may present with<br />

varying degrees of disease severity. Approximately<br />

50% of children with ulcerative colitis will present<br />

with a mild form of disease, characterized by an<br />

insidious onset of diarrhea and rectal bleeding,<br />

without abdominal pain or systemic symptoms<br />

such as fever. In these patients, disease may be<br />

confined to the distal colon. 58,59 Disease of moderate<br />

severity is seen in 30% of children with ulcerative<br />

colitis and is characterized by a more acute<br />

presentation with bloody diarrhea, tenesmus and<br />

urgency; systemic symptoms including low-grade<br />

fever, abdominal tenderness, weight loss and mild<br />

anemia may be present. 58,59 Approximately 10% of<br />

children will present with a severe form of ulcerative<br />

colitis. 58,59 Characteristic findings in severe<br />

disease include six or more bloody stools per day,<br />

fever, weight loss, anemia, hypoalbuminemia and<br />

diffuse abdominal tenderness on physical examination.<br />

58–60 A very small percentage of children<br />

will present initially with extraintestinal symptoms<br />

or manifestations, without obvious intestinal<br />

symptoms. 59 These extraintestinal manifestations<br />

of IBD may include axial or peripheral arthritis,<br />

erythema nodosum, pyoderma gangrenosum, or<br />

primary sclerosing cholangitis (see Extraintestinal<br />

manifestations, p.394).<br />

Diagnosis and differential<br />

Differential diagnosis<br />

The diagnosis of ulcerative colitis is established by<br />

the information gathered from a detailed symptom<br />

and family history, physical examination, and a<br />

combination of laboratory, radiological, endoscopic<br />

and histological findings. It is important to<br />

exclude other etiologies, such as an infectious<br />

process, and to distinguish ulcerative colitis from<br />

Crohn’s disease. Colonic inflammation is typically<br />

characterized by bloody diarrhea with abdominal<br />

cramping. The differential diagnosis of colitis<br />

depends upon the age of the child at the time of<br />

evaluation. In infancy, necrotizing enterocolitis,<br />

Hirschsprung’s enterocolitis and allergic colitis are<br />

common. In contrast, in the older child and<br />

adolescent, enteric infection and IBD are the most<br />

common diagnoses. Causes of colitis are listed in<br />

Table 25.3. In patients with painless rectal bleeding,<br />

other conditions (Meckel’s diverticulum,<br />

Diagnosis and differential 389<br />

Table 25.3 Differential diagnosis of colitis<br />

Infectious etiologies<br />

Campylobacter<br />

Salmonella<br />

Shigella<br />

Escherichia coli 0157: H7 and other<br />

enterohemorrhagic E. coli<br />

Clostridium difficile<br />

Aeromonas<br />

Plesiomonas<br />

Entamoeba histolytica<br />

Cytomegalovirus<br />

Herpes simplex virus<br />

Yersinia<br />

Tuberculosis<br />

HIV and HIV-related opportunistic infections<br />

Other<br />

Ulcerative colitis<br />

Crohn’s disease<br />

Henoch–Schönlein purpura<br />

Hemolytic uremic syndrome<br />

Intestinal ischemia<br />

Intussusception<br />

Allergic colitis (primarily in infancy)<br />

Hirschsprung’s enterocolitis (primarily in infancy)<br />

polyp) should be considered. In addition to details<br />

of the clinical presentation, the history should<br />

include family history, recent antibiotic therapy,<br />

infectious exposures, growth and sexual development<br />

and the presence of extraintestinal manifestations<br />

of ulcerative colitis. Physical examination<br />

should include assessment of height, weight and<br />

body mass index; abdominal distension, tenderness,<br />

or mass; extraintestinal manifestations (e.g.<br />

aphthous stomatitis, pyoderma gangrenosum,<br />

uveitis or arthritis); and fecal blood on rectal<br />

examination, perianal abnormalities (e.g. fistulae,<br />

fissures or tags). Findings on physical examination<br />

may help to distinguish ulcerative colitis from<br />

Crohn’s disease; for example, pronounced growth<br />

failure or a perianal abscess strongly suggests the<br />

diagnosis of Crohn’s disease. A severely ill child<br />

with ulcerative colitis may have tachycardia,<br />

orthostatic hypotension, fever, or dehydration.


390<br />

Ulcerative colitis<br />

Such findings in the presence of abdominal distension<br />

and a concerning abdominal examination,<br />

may herald a fulminant presentation of ulcerative<br />

colitis with increased risk of developing toxic<br />

megacolon.<br />

Laboratory assessment<br />

Initial laboratory evaluation should include appropriate<br />

blood tests, stool for occult blood, Clostridium<br />

difficile toxin assay and stool cultures. A<br />

complete blood cell count with differential may<br />

reveal a leukocytosis with or without left shift,<br />

anemia, or thrombocytosis. Thrombocytosis, hypoalbuminemia<br />

and elevated erythrocyte sedimentation<br />

rate (ESR) or C-reactive protein (CRP) may<br />

indicate increased disease activity. 61–63 The presence<br />

of anemia with low mean corpuscular<br />

volume (MCV), wide red cell distribution width<br />

(RDW) and low iron levels may indicate an irondeficient<br />

anemia secondary to ongoing fecal blood<br />

losses or the anemia of chronic disease. Children<br />

with significant mucosal inflammation may have<br />

normal laboratory test results. In a study of children<br />

with ulcerative colitis or Crohn’s colitis, 13 of<br />

36 patients with ulcerative colitis (36%) had<br />

normal blood test results, including seven of 28<br />

ulcerative colitis patients with macroscopic findings<br />

(obvious abnormalities) on colonoscopy, and<br />

12 of 31 ulcerative colitis patients with histological<br />

moderate or severe chronic inflammation. 63 Stool<br />

examination should rule out possible enteric infections<br />

(see Table 25.3). In sexually active patients,<br />

rectal cultures for gonorrhea should be considered.<br />

In ulcerative colitis, Gram stain or methylene blue<br />

stain of stools may identify leukocytes.<br />

It has been proposed that certain serum antibodies<br />

may be helpful for screening for IBD and discriminating<br />

ulcerative colitis from Crohn’s disease. 64,65<br />

Perinuclear antineutrophil cytoplasmic antibodies<br />

(P-ANCA) are seen in 60–80% of adults with<br />

ulcerative colitis compared to 10–27% of<br />

adults with Crohn’s disease. 64,66,67 Similarly, anti-<br />

Saccharomyces cerevisiae antibodies (ASCA) are<br />

commonly found in individuals with Crohn’s<br />

disease but are rarely seen in ulcerative colitis. In<br />

a study of 173 children, ASCA yielded a sensitivity<br />

of 55% and specificity of 95% for Crohn’s<br />

disease, and ANCA had a sensitivity of 57% and<br />

specificity of 92% for ulcerative colitis. 64 In a<br />

study of 128 pediatric patients undergoing evaluation<br />

for IBD, Dubinsky et al utilized modified cutoff<br />

values to optimize the sensitivity of the ASCA<br />

and ANCA assays. For the combination of ASCA<br />

and P-ANCA, the sensitivity of detecting IBD<br />

increased to 81% with the modified values<br />

compared to the 69% with standard cut-off values;<br />

however, this was accompanied by an increase in<br />

false-positive rates among the children without<br />

IBD. 65 An overlap of ASCA and P-ANCA positive<br />

serology between patients with Crohn’s disease or<br />

ulcerative colitis remains. In particular, P-ANCA<br />

tends to test positive in the serum of patients with<br />

Crohn’s disease who exhibit ulcerative colitis<br />

features. 64,68 The value of these tests to supplement<br />

the routine diagnostic tests in IBD is a<br />

subject under study.<br />

Endoscopic and radiographic evaluation<br />

Evaluation with colonoscopy and ileoscopy with<br />

biopsies and a barium upper gastrointestinal series<br />

with small-bowel follow-through should be<br />

performed to diagnose ulcerative colitis, determine<br />

the extent and severity of ulcerative colitis presentation<br />

and distinguish ulcerative colitis from<br />

Crohn’s disease or a non-IBD diagnosis. In patients<br />

with severe colitis, a limited flexible sigmoidoscopy<br />

examination with minimal air insufflation<br />

may be prudent to avoid increased risk of a<br />

full colonoscopy (perforation, hemorrhage, toxic<br />

dilatation). In order to establish the extent of<br />

disease involvement by colonoscopy, we recommend<br />

biopsies from the terminal ileum and each<br />

segment of the colon, even if there are no visible<br />

findings at a particular level of the colon. In ulcerative<br />

colitis, typical findings seen by the endoscopist<br />

include a diffuse, continuous process starting<br />

at the rectum and extending more proximally<br />

into the colon. However, in children with ulcerative<br />

colitis, rectal sparing has been reported. 69,70<br />

The colonic mucosa often appears edematous,<br />

erythematous and friable, with minute surface<br />

erosions and ulcerations (Figure 25.1). Larger,<br />

deeper ulcerations with associated exudate may<br />

develop in more severe disease. With more chronic<br />

(longstanding) ulcerative colitis, pseudopolyps<br />

may be present. (Figure 25.2). In contrast, in<br />

Crohn’s disease, colonoscopy may reveal focal<br />

ulcerations (aphthous lesions) with intervening<br />

areas of normal-appearing mucosa (skip lesions).


Figure 25.1 Severe colitis at initial presentation of ulcerative<br />

colitis. The colonoscopy demonstrates a featureless<br />

colon with loss of vascular pattern and hemorrhage.<br />

In severe or chronic Crohn’s disease, linear ulcerations,<br />

nodularity (cobblestoning) and strictures or<br />

stenoses may be present. In general, the ulcerations<br />

in Crohn’s disease are deeper and focal<br />

versus the diffuse, superficial ulcerations typical<br />

of ulcerative colitis (Table 25.4). 64–74<br />

An upper gastrointestinal series with small-bowel<br />

follow-through should be performed to look for<br />

any evidence of abnormality of the terminal ileum<br />

or more proximal gastrointestinal tract, and the<br />

presence of fistulae, which would suggest Crohn’s<br />

disease. The only exception is the finding of ulcerative<br />

colitis-associated ‘backwash ileitis’, which<br />

should be distinguished from terminal ileal<br />

disease of Crohn’s disease. With ‘backwash ileitis’,<br />

the ileum appears patulous and inflamed, involves<br />

only the distal ileal segment and is associated with<br />

pancolitis; there should be no evidence of extensive<br />

ulcerations or stricturing, as seen with<br />

Crohn’s disease. 75<br />

Although by definition, the disease of ulcerative<br />

colitis is confined to the colon, children with<br />

Diagnosis and differential 391<br />

Figure 25.2 Colonic pseudopolyps present in a 16-yearold<br />

girl with an 8-year history of ulcerative colitis. The<br />

patient had persistent symptoms of diarrhea and rectal<br />

bleeding despite chronic corticosteroid and 6-mercaptopurine<br />

therapy, and subsequently underwent colectomy.<br />

Crohn’s disease or ulcerative colitis can have<br />

inflammation of the upper gastrointestinal<br />

tract. 76–79 In a controlled, blinded study of children<br />

with ulcerative colitis or Crohn’s disease,<br />

biopsies from the esophagus, gastric antrum and<br />

duodenum revealed histological evidence of<br />

esophagitis, gastritis and duodenitis in patients<br />

with either ulcerative colitis or Crohn’s disease.<br />

Esophagitis occurred in 50%, gastritis in 69% and<br />

duodenitis in 23% of patients with ulcerative<br />

colitis. In contrast to ulcerative colitis patients,<br />

those with Crohn’s disease had a higher prevalence<br />

of esophagitis, gastritis and duodenitis (72%, 92%<br />

and 33%, respectively). 76 Granulomas of the upper<br />

gastrointestinal tract were seen in 40% of the<br />

patients with Crohn’s disease and duodenal cryptitis<br />

was noted in 26% of patients with Crohn’s<br />

disease; none of these lesions was seen in the biopsies<br />

of patients with ulcerative colitis. 76 In another<br />

study, the presence of focally enhanced gastritis in<br />

children did not reliably differentiate between<br />

Crohn’s disease and ulcerative colitis. 79 Except for<br />

the presence of granulomas of Crohn’s disease, it<br />

may be difficult to distinguish between ulcerative


392<br />

Ulcerative colitis<br />

Table 25.4 Typical endoscopy and histopathology findings – ulcerative colitis (UC) vs. Crohn’s<br />

disease (from references 69–74, 84)<br />

Characteristic Ulcerative colitis Crohn’s disease<br />

Endoscopy findings diffuse continuous involvement focal lesions/disease interspersed<br />

extending from the rectum with normal-appearing mucosa (skip lesions)<br />

colitis and Crohn’s disease by the appearance of<br />

upper gastrointestinal lesions. Some pediatric<br />

centers routinely perform upper endoscopy in<br />

addition to colonoscopy under general anesthesia<br />

in their initial evaluation of children with<br />

suspected IBD, in order to determine the extent and<br />

severity of upper gastrointestinal inflammation.<br />

Evaluation of plain abdominal radiographs and<br />

abdominal/pelvic computerized tomography (CT)<br />

scans may aid in the assessment for complications<br />

of ulcerative colitis, including toxic megacolon,<br />

perforation or stricture. A plain abdominal radiograph<br />

may demonstrate thumbprinting, loss of<br />

haustral patterns, colonic dilatation (i.e. toxic<br />

megacolon), obstruction, or pneumoperitoneum<br />

(i.e. perforation of the bowel). 80–82 CT is not typi-<br />

rectum usually involved* rectal sparing possible<br />

diffuse, superficial, minute aphthous lesions often surrounded by<br />

ulcerations; deeper ulcerations normal-appearing mucosa; deep ‘collar button’<br />

in severe disease ulcers; linear or serpiginous ulcerations<br />

strictures very rare strictures, often occurring in terminal ileum<br />

pseudopolyps<br />

Histopathology findings no granulomas † granulomas (36%)<br />

diffuse chronic inflammation focal chronic inflammation,<br />

limited to the mucosa †† ; crypt transmural inflammation<br />

abscesses<br />

with or without architectural<br />

distortion**<br />

* In children, ‘rectal sparing’ has been seen in UC69,70 † Giant cell reactions can occur around damaged crypts and spilled mucin. This must be distinguished from ‘true’<br />

granulomas, which by definition, are not seen in UC<br />

†† Deeper layers of the colon may be involved in fulminant UC disease<br />

** In children with UC, initial colonic biopsies at time of diagnosis are less likely to show architectural distortion<br />

than biopsies from adults74 cally performed in the initial assessment for ulcerative<br />

colitis, but may demonstrate diffuse bowel<br />

wall thickening, marked rectal wall thickening and<br />

perirectal fibrofatty proliferation. 75 A barium<br />

enema should not be performed during active<br />

ulcerative colitis as this predisposes to toxic megacolon.<br />

83 Prior to the routine use of colonoscopy for<br />

the diagnosis of ulcerative colitis, published<br />

barium enema findings of ulcerative colitis<br />

included mucosal granularity, superficial ulcerations,<br />

thickened and nodular haustral folds<br />

(secondary to inflammation and edema) and<br />

colonic shortening. 81 With longstanding ulcerative<br />

colitis, the colon becomes featureless and markedly<br />

shortened 81 and strictures may form, which may<br />

require colonoscopy for further evaluation for<br />

possible cancer. 75


(a) (b)<br />

Pathology of ulcerative colitis<br />

In active ulcerative colitis, typical findings on<br />

histopathology include a diffuse inflammatory<br />

cell infiltrate of the lamina propria mostly with<br />

plasma cells, lymphocytes and neutrophils, but<br />

mast cells and eosinophils are also seen 73 (Figure<br />

25.3a). Neutrophils invade the epithelium of the<br />

crypts, leading to cryptitis, crypt abscess formation<br />

and goblet cell mucin depletion (Figure<br />

25.3b). The inflammatory infiltrate is typically<br />

confined to the mucosa, but in severe ulcerative<br />

colitis, ulceration may extend into the submucosa<br />

and deeper layers. 73 In quiescent (inactive) ulcerative<br />

colitis, the inflammatory infiltrate may<br />

diminish, but signs of chronic colitis (architectural<br />

distortion, crypt branching and shortening,<br />

reduction in the number of crypts and separation<br />

of crypts) can persist. 73 In children with ulcerative<br />

colitis, signs of chronic colitis (e.g. architectural<br />

distortion) are not always seen. 74<br />

Histological differentiation of ulcerative colitis<br />

from Crohn’s disease can be difficult. The histological<br />

hallmark of Crohn’s disease is the noncaseating<br />

granuloma, which may be found in up to<br />

36% of children with Crohn’s disease. 84 However,<br />

a giant cell reaction mimicking a granuloma can<br />

Diagnosis and differential 393<br />

Figure 25.3 (a) Low-power view of a colonic biopsy from a patient with active ulcerative colitis. Note the increased<br />

lamina propria inflammatory infiltrate, crypt abscesses and crypt architectural distortion. The crypts are irregular in shape<br />

and placement and do not descend to the level of the muscularis mucosae. (b) High-power view of colonic crypts in a<br />

patient with active ulcerative colitis, demonstrating neutrophilic infiltration in the crypt and a crypt abscess. (Courtesy of<br />

Jonathan Glickman, MD, Department of Pathology, Children’s Hospital, Boston).<br />

occur around damaged crypts and spilled mucin.<br />

These ‘mucin granulomas’ must be distinguished<br />

from true granulomas, which, by definition, are<br />

not seen in ulcerative colitis. 73 Histological skip<br />

areas, rectal sparing, focal inflammation and transmural<br />

inflammation also suggest the diagnosis of<br />

Crohn’s disease. How-ever, in children, histological<br />

skip areas may occur both at initial presentation<br />

of ulcerative colitis and as a result of<br />

therapy. 69,85 In addition, patients with ulcerative<br />

colitis can have histological evidence of upper<br />

gastrointestinal inflammation similar to that in<br />

Crohn’s disease. 76,77,79 Given the overlap of<br />

histopathology findings in ulcerative colitis and<br />

Crohn’s disease, it may be difficult to distinguish<br />

between these two diagnoses if granulomas are not<br />

present.<br />

If a clinician cannot reliably distinguish between<br />

Crohn’s disease and ulcerative colitis based on the<br />

available clinical, radiographic and endoscopic<br />

data, an interim diagnosis of ‘indeterminate colitis’<br />

may be given until the patient can be more clearly<br />

classified in the future. The prevalence of indeterminate<br />

colitis in adults and children with IBD is<br />

estimated to be 10–20%. 10,86 Approximately onethird<br />

of these patients will later be classified as<br />

ulcerative colitis or Crohn’s disease. 87


394<br />

Ulcerative colitis<br />

Extraintestinal manifestations<br />

Approximately 25–35% of patients with IBD<br />

develop extraintestinal symptoms. 88,89 Extraintestinal<br />

manifestations of IBD may occur before,<br />

during, or after the development of gastrointestinal<br />

symptoms and may appear after surgical removal<br />

of diseased bowel. 88,90–93 The clinical activity of<br />

the extraintestinal manifestations may or may not<br />

correlate with the activity of intestinal inflammation.<br />

Joint manifestations (arthropathy) occur in 5–20%<br />

of children with ulcerative colitis. 4,90 These can be<br />

classified into two main clinical forms: a peripheral<br />

arthropathy and an axial arthropathy (e.g.<br />

ankylosing spondylitis). 88,90 Patients with IBD<br />

develop peripheral arthropathies in approximately<br />

5–20% of cases. 88–90,94 The peripheral arthropathy<br />

is generally asymmetrical, non-deforming and<br />

migratory, affecting mostly the large joints of the<br />

lower extremities including the knees, ankles and<br />

hips. Less commonly, the upper limb joints or<br />

hands are affected. 88,90,94 Small joints of the hands<br />

and feet are generally spared. 90<br />

Exacerbations of peripheral joint disease seem to<br />

parallel increased activity of bowel disease in<br />

ulcerative colitis or Crohn’s disease. 95 Orchard et<br />

al, in a study of 976 adults with ulcerative colitis,<br />

suggested that there may be two subtypes of<br />

peripheral arthropathy: a pauciarticular form with<br />

fewer than five swollen joints, and a polyarticular<br />

form with more than five joints. Of these two<br />

subtypes, the pauciarticular arthropathy is more<br />

likely to be correlated with exacerbations of bowel<br />

disease. 93<br />

Axial arthropathies associated with HLA B27<br />

occur in 1–4% of patients. 88–90,94 Ankylosing<br />

spondylitis associated with IBD runs a course<br />

independent of the activity of bowel disease, and<br />

may progress to permanent deformity. 88,90,94 In<br />

addition to the two main forms of joint manifestations,<br />

individuals with ulcerative colitis can<br />

develop isolated arthritis involving large joints,<br />

including the sacroiliac joints, hips and shoulders.<br />

94<br />

Pyoderma gangrenosum and erythema nodosum<br />

are the two main skin manifestations associated<br />

with ulcerative colitis and Crohn’s disease.<br />

Pyoderma gangrenosum occurs in


aminotransferase (ALT) occur in 12% of children<br />

with ulcerative colitis and appear to be related to<br />

medications or disease activity. 106 Persistent ALT<br />

elevations suggest the presence of primary sclerosing<br />

cholangitis (PSC) or autoimmune chronic<br />

hepatitis. 106 Among children with ulcerative<br />

colitis, 3.5% develop sclerosing cholangitis and<br />


396<br />

Ulcerative colitis<br />

precipitating factors, including narcotic agents for<br />

pain or antidiarrheal effects, anticholinergic<br />

agents, drugs that decrease motility, or antidepressants<br />

with significant anticholinergic<br />

effects. 121,128,129 A barium enema or colonoscopy<br />

may cause distension that can further impair the<br />

colonic wall blood supply and may increase the<br />

mucosal uptake of bacterial products. 122 Barium<br />

enema examinations have been reported in proximity<br />

to the development of toxic megacolon. 83,129<br />

The early discontinuation or rapid tapering of<br />

steroids or 5-aminosalicylic acid (5-ASA) may<br />

contribute to the development of toxic megacolon.<br />

121,122 Electrolyte abnormalities, such as<br />

hypokalemia, have been observed in the setting of<br />

toxic megacolon, although it is not clear whether<br />

this finding is a causative factor or secondary to<br />

the illness itself. 121 Along with colonic dilatation,<br />

patients with toxic megacolon present with<br />

systemic findings, including fever, tachycardia,<br />

leukocytosis and anemia. 124 A decrease in the<br />

number of stools may herald the onset of toxic<br />

Figure 25.4 Toxic megacolon in a teenager with fulminant<br />

ulcerative colitis. There is a massively dilated loop of<br />

transverse colon in the upper quadrants, with a paucity of<br />

bowel gas in the remainder of the abdomen. This patient<br />

developed the megacolon despite corticosteroid therapy,<br />

and subsequently underwent emergent surgery for a<br />

colonic perforation. (Courtesy of Carlo Buonomo, MD,<br />

Department of Radiology, Children’s Hospital, Boston).<br />

megacolon. With progressive disease, these individuals<br />

can develop dehydration, mental status<br />

changes, electrolyte disturbances, hypotension<br />

and increasing abdominal distension and tenderness,<br />

with or without signs of peritonitis. 122,124<br />

Abdominal X-ray reveals colonic dilatation, most<br />

frequently involving the transverse colon, sometimes<br />

accompanied by inflammatory changes<br />

including an absent or markedly edematous haustral<br />

pattern 130 (Figure 25.4). In two series of adults<br />

with toxic megacolon, the diameter of the colon<br />

varied, with a range of 5.8–16cm. 125,129 Because<br />

the transverse colon is the most anterior portion of<br />

the colon, air will tend to accumulate in this<br />

segment of the colon when the patient is in the<br />

supine position; however, with repositioning of<br />

the patient, the colonic air will redistribute, filling<br />

other segments of the bowel. 131<br />

The management of toxic megacolon is detailed in<br />

multiple reviews elsewhere. 121,122 If toxic megacolon<br />

is present, surgical consultation is essential,<br />

and the patient will probably require a colectomy.<br />

Some authors have utilized medical management<br />

of this condition; however, this requires very close<br />

monitoring to avoid complications. 132 Early surgical<br />

intervention is indicated in the setting of failed<br />

medical therapy with progressive colonic dilatation,<br />

worsening systemic toxicity, perforation or<br />

uncontrolled hemorrhage. 122<br />

Both benign and malignant colonic strictures can<br />

develop in longstanding ulcerative colitis. 75,133,134<br />

Benign strictures present most commonly in the<br />

rectum and the sigmoid, are due to smooth-muscle<br />

hypertrophy and are thought to be potentially<br />

reversible. 133 Colonic strictures should be evaluated<br />

for possible malignancy, but the majority of<br />

strictures in ulcerative colitis are benign. 75,133,134<br />

There is an increased risk of dysplasia and colon<br />

cancer in patients with longstanding ulcerative<br />

colitis, which is addressed later in this chapter (see<br />

Prognosis and follow-up, p.407).<br />

Treatment options<br />

The treatment goals for children with ulcerative<br />

colitis are the control of active disease and induction<br />

of remission, the long-term maintenance of<br />

remission, and provision of education and


psychosocial support for the patient and family.<br />

The initial treatment of ulcerative colitis is<br />

medical, with surgery reserved for patients with<br />

severe disease, patients with medically refractory<br />

disease, or patients who develop adverse effects of<br />

medical therapy.<br />

Knowledge of the extent and severity of disease<br />

involvement will enable the clinician to choose<br />

the appropriate therapy for each individual<br />

patient. Distal ulcerative colitis (left-sided ulcerative<br />

colitis or proctitis) is characterized by involvement<br />

limited to the area distal to the splenic<br />

flexure, and potentially may be treated with<br />

topical agents (e.g. aminosalicylate or hydrocortisone<br />

enemas or suppositories). Extensive ulcerative<br />

colitis is defined by involvement extending<br />

proximally to the splenic flexure and requires<br />

systemic therapies with or without additional<br />

topical agents. Severity of disease is usually simple<br />

to ascertain, and can be determined by assessing<br />

stool frequency and consistency, abdominal pain,<br />

nocturnal diarrhea, hematocrit, albumin level and<br />

feeding intolerance. Separate published disease<br />

severity criteria for adults and children have been<br />

developed by Truelove and Witts, 135 and Werlin<br />

and Grand, 60 respectively (Tables 25.5 and 25.6).<br />

Other clinical scoring systems have been utilized<br />

in clinical trials to quantify disease severity of<br />

Treatment options 397<br />

ulcerative colitis, but are less useful for day-to-day<br />

clinical management. 136–139 Typically, severe<br />

colitis requires hospitalization and administration<br />

of either intravenous corticosteroids or other<br />

immunosuppressive agents (e.g. cyclosporin).<br />

Many patients with severe colitis will require<br />

colectomy.<br />

Induction therapy<br />

Table 25.5 Severity of ulcerative colitis in adults* (from reference 135)<br />

Characteristic Mild Severe<br />

Grossly bloody diarrhea ≤ 4 stools per day ≥ 6 stools per day<br />

(≤ small amounts of blood)<br />

Mild-to-moderate colitis<br />

In the child with mild-to-moderate ulcerative<br />

colitis, with no or only minimal systemic signs<br />

Fever none mean evening temperature >37.5ºC, or<br />

temperature ≥37.8ºC, at least 2 out of 4 days<br />

Tachycardia none >90beats/min<br />

Anemia not severe; hemoglobin present; hemoglobin ≤75% of normal value<br />

essentially normal<br />

Erythrocyte ≤30mm/h >30mm/h<br />

sedimentation rate<br />

* Moderate severe – intermediate between severe and mild<br />

Table 25.6 Characteristics of severe colitis<br />

in children* (from reference 60)<br />

Grossly bloody diarrhea, ≥ 5 stools per a day<br />

Oral temperature >37.8ºC during the first hospital day<br />

Tachycardia (pulse ≥ 90)<br />

Anemia (hematocrit ≤ 30%)<br />

Hypoalbuminemia (serum albumin ≤ 3.0g/100ml)<br />

Toxic megacolon<br />

* Patients need to fulfill four of the first five criteria<br />

or the 6th criterion alone


398<br />

Ulcerative colitis<br />

Table 25.7 Aminosalicylate agents* (from references 325, 326)<br />

Oral preparations Dosage form Mechanism of release Site of delivery<br />

Azo-bond<br />

sulfasalazine 500-mg tablet bacterial cleavage of colon<br />

(Azulfidine) azo bond<br />

olsalazine 250-mg capsule bacterial cleavage of colon<br />

(Dipentum) azo bond<br />

balsalazide 750-mg capsule bacterial cleavage of colon<br />

(Colazal/Colazide) azo bond<br />

Delayed-release<br />

mesalamine 400mg/800mg pH-dependent breakdown distal ileum<br />

(Asacol) tablets (pH >7) to colon<br />

mesalamine (Salofalk/ 250mg/500mg pH-dependent breakdown ileum to colon<br />

Mesasal/Claversal) tablets (pH >6)<br />

Sustained-release<br />

mesalamine 250mg/500mg/ ethylcellulose-controlled small intestine<br />

(Pentasa) 1000mg tablets time-release to colon<br />

Rectal preparations<br />

mesalamine suppository 400mg/500mg/ rectum<br />

(Canasa – 500mg) 1000mg<br />

mesalamine enema 1g/4g; 60-ml/ rectum to splenic<br />

(Rowasa – 4g/60ml) 100-ml suspension flexure<br />

* Availability of different preparations and dosage forms varies between different markets/countries<br />

(such as elevated ESR or mild anemia), aminosalicylates<br />

(ASAs) (e.g. sulfasalazine, olsalazine,<br />

mesalamine, balsalazide) are usually the first line<br />

of therapy (Tables 25.7 and 25.8). ASAs have<br />

multiple immunological effects. Potential mechanisms<br />

of action include the inhibition of the<br />

synthesis of leukotriene B4, a potent chemotactic<br />

and chemokinetic agent, and the inhibition of the<br />

activation of nuclear transcription factor κB (NFκB),<br />

an important mediator of the immune<br />

response in inflammatory processes. 140–142 Controlled<br />

studies suggest that currently available<br />

ASAs are superior to placebo for induction of<br />

remission and prevention of relapse. 143–146<br />

However, there do not appear to be any differences<br />

in efficacy between the older agent, sulfasalazine,<br />

and newer ASA drugs. 146,147 There are very few<br />

trials of ASA therapy in children with ulcerative<br />

colitis. One pediatric multicenter, randomized,<br />

double-blind study compared the efficacy and<br />

safety of olsalazine (30mg/kg per day; maximum<br />

2g/day) to sulfasalazine (60mg/kg per day;<br />

maximum 6g/day) in the treatment of mild-tomoderate<br />

ulcerative colitis. The findings demonstrated<br />

clinical remission after 3 months in 79% of<br />

the sulfasalazine-treated children, in comparison<br />

to 39% of the olsalazine-treated children. 148 The<br />

authors suggested the low dosage of olsalazine as a<br />

possible explanation for the difference in efficacy<br />

between sulfasalazine and osalazine. 148<br />

Potential advantages of the non-sulfa ASA agents<br />

include better tolerance compared to sulfasalazine<br />

147,149 and the availability of a non-sulfa


Table 25.8 Medical therapies for ulcerative colitis<br />

Medication Dosage Major side-effects<br />

ASA agent for sulfa-sensitive individuals. In adultonset<br />

ulceratimve colitis, balsalazide at higher<br />

doses (6.75g/day) may provide a faster improvement<br />

in active, mild-to-moderate ulcerative colitis<br />

than lower doses of balsalazide (2.25g/day) or mesalamine<br />

(2.4g/day). 150,151 Although some studies<br />

Treatment options 399<br />

Sulfasalazine 50–75mg/kg per day PO divided nausea, headaches, diarrhea,<br />

qid, bid or tid (maximum 6g/day) photosensitivity, hypersensitivity reaction,<br />

adult dose: 3–4g/day pancreatitis, azoospermia, hemolytic<br />

divided bid or tid anemia, neutropenia<br />

Mesalamine<br />

oral formulation 50–75mg/kg per day PO divided qid, nausea, headaches, diarrhea, pancreatitis,<br />

tid, or bid (may vary according to<br />

preparation) (maximum 6g/day);<br />

adult dose: 3–4g/day divided qid,<br />

tid, or bid<br />

nephritis, pericarditis, pleuritis<br />

enema formulation 2–4g PR q 12–24 h<br />

suppository formulation 500mg PR q 12–24 h<br />

Corticosteroids<br />

intravenous or 1–2mg/kg per day of prednisone or numerous; including Cushing’s syndrome,<br />

oral formulation equivalent, IV or PO, divided q 12 to growth suppression, immunosuppression,<br />

24h (maximum 60 mg/day) hypertension, hyperglycemia, increased<br />

enema formulation 50–100mg of hydrocortisone PR qhs appetite, osteoporosis, aseptic<br />

suppository formulation 25mg of hydrocortisone acetate PR qhs necrosis (hip), cataracts<br />

Azathioprine* 1.5–2.5mg/kg per day PO qd nausea, emesis, immunosuppression,<br />

hepatotoxicity, pancreatitis, myelosuppression<br />

6-Mercaptopurine* 1.0–2.0mg/kg per day PO qd nausea, emesis, immunosuppression,<br />

hepatotoxicity, pancreatitis, myelosuppression<br />

Cyclosporin induction regimen for fulminant colitis: nephrotoxicity, hypertension, headache,<br />

initial dose, 4mg/kg per day IV hirsutism, nausea, emesis, diarrhea,<br />

continuous or bid; maintenance oral tremor, hypomagnesemia, hyperkalemia,<br />

dose varies according to oral preparation hepatotoxicity, seizures, gingival<br />

hyperplasia, possibly lymphoproliferative<br />

disorder<br />

PO, orally; bid, twice a day; tid, three times a day; qid, four times a day; PR, per rectum; q, every; qd, every day; IV,<br />

intravenously; qhs, at bedtime<br />

* Thiopurine methyltransferase (TPMT) genotype determines metabolism and blood levels of metabolites; may help to<br />

determine risk of myelosuppression and optimal dosage of azathioprine or 6-mercaptopurine<br />

suggest an advantage of azo-bond ASA agents (e.g.<br />

balsalazide) in comparison to delayed-release ASA<br />

agents (e.g. mesalamine), the overall data suggest<br />

equivalence between the different oral ASAs,<br />

when comparable amounts of the ASA are released<br />

at the site of disease activity. 152


400<br />

Ulcerative colitis<br />

Common side-effects of sulfasalazine include<br />

headache, nausea and fatigue, which improve<br />

with reduction of the dose 152 (Table 25.8). The<br />

sulfa moiety can cause hypersensitivity reactions<br />

resulting in rash, fever, hepatitis, hemolytic<br />

anemia, bone marrow suppression and pneumonitis.<br />

152,153 Other side-effects include neutropenia,<br />

oligospermia, pancreatitis and the exacerbation of<br />

colitis. 60,154 Folic acid supplementation is recommended,<br />

given that sulfasalazine impairs the<br />

absorption of folic acid and may lead to<br />

anemia. 152 To decrease side-effects, sulfasalazine<br />

is started at a dose of 10–20mg/kg per day and<br />

gradually increased to the full dose (50–75mg/kg<br />

per day) over 5–7 days. Mesalamine and the other<br />

non-sulfa ASA agents have also been associated<br />

with adverse reactions, including pancreatitis,<br />

hepatitis, nephritis, exacerbation of colitis, and<br />

pneumonitis. 152,155,156<br />

In addition to medical therapy, a low-residue diet<br />

(no popcorn, nuts, seeds, raw fruits and vegetables)<br />

and avoidance of spicy foods during acute<br />

symptoms has been anecdotally reported to reduce<br />

symptoms during a flare-up of disease. 157<br />

Moderate-to-severe colitis<br />

Children with moderate disease are usually<br />

managed with oral corticosteroids (usually<br />

1mg/kg per day, up to 60mg/day of prednisone) as<br />

out-patients. In an uncontrolled study of 20 children<br />

with active ulcerative colitis (three with<br />

mild, 13 with moderate, four with severe activity),<br />

85% of the children achieved clinical remission<br />

with combination therapy of corticosteroids (oral<br />

prednisolone for pancolitis or topical prednisolone<br />

for distal colitis) and mesalamine<br />

(20–40mg/kg per day). 158 Reassessment with<br />

colonoscopy at 8 weeks demonstrated complete<br />

endoscopic remission in 40% and full histological<br />

remission in only 15%, suggesting that clinical<br />

remission may not correlate with endoscopic or<br />

histological remission. 158 Potential short-term<br />

complications of steroid therapy in patients with<br />

ulcerative colitis include increased appetite,<br />

weight gain, fluid retention, mood swings, hyperglycemia,<br />

hypertension, insomnia, acne and facial<br />

swelling.<br />

Complications of long-term steroid therapy<br />

(usually of more than 3 months) include growth<br />

restriction, osteopenia with compression fractures,<br />

aseptic necrosis of the hip, and cataracts. 152,159<br />

Given these reasons and the suppression of the<br />

hypothalamic–pituitary–adrenal axis, corticosteroids<br />

should be tapered shortly after remission is<br />

achieved. A standard taper utilized by the authors<br />

is reduction by 5mg/week of prednisone down to<br />

20mg/day, and then a more gradual taper on alternate<br />

days, aiming for 10mg every other day with<br />

further taper and cessation if remission is maintained.<br />

159 Budesonide, a steroid with a high firstpass<br />

metabolism and fewer systemic side-effects,<br />

is available in capsule form for treatment of ileocecal<br />

Crohn’s disease. 160 In Europe and Canada,<br />

there is an enema form of budesonide for treatment<br />

of distal colitis. 161 Unfortunately, there is no<br />

available oral budesonide preparation shown to<br />

treat patients with pancolitis effectively. 162<br />

Children with severe disease (e.g. more than five<br />

bowel movements/day, liquid bloody stools, or<br />

severe pain with defecation, anemia and hypoalbuminemia)<br />

require intravenous corticosteroids<br />

(methylprednisolone at 40–60mg/day, divided<br />

into two doses/day, approximately 1–2mg/kg per<br />

day) and hospitalization for further evaluation,<br />

observation and management. 157,163 Rectal corticosteroids<br />

or 5-ASA agents may be used as an<br />

adjunctive therapy with parenteral corticosteroids<br />

for patients with severe tenesmus. 152 Intravenous<br />

fluid for rehydration and correction of electrolyte<br />

imbalances should be provided. Blood transfusions<br />

and albumin infusions may be required. If<br />

bowel rest is indicated, the child may require<br />

central venous access for parenteral nutrition<br />

support. In addition to high-dose steroids, empiric<br />

antibiotics are sometimes used in severe colitis,<br />

although the efficacy of antibiotics has not been<br />

proven. 164–167 Assessment for response to intensive<br />

medical therapy includes resolution of fever,<br />

tachycardia, abdominal tenderness and macroscopic<br />

blood per rectum. Stools should be<br />

decreasing in frequency, but may still be<br />

unformed. Once the child shows significant<br />

improvement, diet is advanced to a low-residue<br />

diet, intravenous methylprednisolone is switched<br />

to oral prednisone, and similar parameters for<br />

steroid wean are followed as outlined above. The<br />

optimal duration of intravenous corticosteroid<br />

therapy is unclear, but most children will respond<br />

within 7–10 days.


If the child has not responded to steroid therapy<br />

after 7–10 days, the options of surgery or more<br />

intensive immunosuppression (intravenous cyclosporin<br />

or oral tacrolimus) should be considered<br />

and discussed with the family. Intensive immunosuppression<br />

should not be started if surgery is<br />

believed imminent, such as in a septic patient, a<br />

patient with toxic megacolon, or a patient with a<br />

suspected perforation. Intravenous or oral<br />

cyclosporin therapy has successfully induced<br />

remission in children with steroid-refractory<br />

ulcerative colitis. 168–170 The exact dosage of<br />

cyclosporin varies in different protocols, depending<br />

upon whether intravenous or oral medication<br />

is utilized. 136,169–171 In a study of 14 children<br />

treated with oral cyclosporin therapy for severe<br />

active colitis, unresponsive to high-dose intravenous<br />

steroids, 80% achieved clinical remission<br />

within 2–9 days; however, the majority of children<br />

who initially responded to cyclosporin eventually<br />

required colectomy within a year. 170 In these<br />

studies, most of the children who improved with<br />

cyclosporin induction relapsed or underwent<br />

colectomy within a year. 169,170 Cyclosporin may be<br />

most useful in delaying emergency colectomy at a<br />

time when the child’s health status is most<br />

compromised (high-dose steroids, anemia, hypoalbuminemia,<br />

poor nutrition) and may allow time<br />

to improve the child’s health and mental status in<br />

preparation for future ‘elective’ colectomy. 170 A<br />

recent review outlines specific recommendations<br />

on the initiation and monitoring of cyclosporin<br />

therapy in children with IBD. 171<br />

Oral tacrolimus can also induce remission of<br />

disease activity in children with severe active<br />

colitis. 172 If either cycloporin or tacrolimus is<br />

utilized as induction therapy, the aim should be<br />

the transition of patients off these potentially toxic<br />

agents and onto a maintenance medication (e.g. 6mercaptopurine,<br />

azathioprine) over a 3–6-month<br />

period. Azathioprine or 6-mercaptopurine may be<br />

effective in preventing relapse after cyclosporininduced<br />

remission in children with ulcerative<br />

colitis. 173 Children on cyclosporin or tacrolimus<br />

should receive prophylaxis for Candida and<br />

Pneumocystis carinii, and have careful monitoring<br />

of electrolytes, blood glucose, renal function,<br />

blood pressure and neurological status.<br />

The timing of colectomy in a child who is not<br />

responding to intravenous corticosteroids or other<br />

Treatment options 401<br />

immunosuppression can be difficult. It should be<br />

emphasized that, even if a child responds to<br />

immunosuppression, such as cyclosporin, there is<br />

a high likelihood that he/she will require a colectomy<br />

within a year. 170,172 If immunosuppression<br />

with cyclosporin is used and the child does not<br />

respond to these medications within 10–14 days,<br />

surgery should be strongly recommended.<br />

Close monitoring of patients with severe colitis for<br />

the development of fulminant colitis and associated<br />

complications including hemorrhage, toxic<br />

megacolon and perforation is essential, using<br />

serial abdominal examinations, complemented by<br />

serial abdominal films or other imaging as necessary.<br />

Physical examination should look for<br />

evidence of worsening abdominal tenderness,<br />

distension and hypoactive bowel sounds; these<br />

may herald the development of toxic megacolon.<br />

The appearance of persistent abdominal pain and<br />

distension, diffuse abdominal tenderness and<br />

rebound, fever and tachycardia are worrisome<br />

signs of an acute abdomen and may signal the<br />

need for emergency surgical intervention. Opiates<br />

and loperamide should be avoided, given the<br />

increased risk of developing toxic megacolon. 128<br />

Steroid therapy may mask the typical symptoms<br />

and signs of perforation, but these may be detected<br />

by serial upright abdominal films (see Complications,<br />

p.395).<br />

Left-sided colitis/proctitis<br />

Topical ASA or topical corticosteroids are effective<br />

in the treatment of proctitis, proctosigmoiditis, or<br />

left-sided ulcerative colitis174–176 (see Tables 25.7<br />

and 25.8). To be effective, topical therapy must<br />

reach the most proximal extent of the disease<br />

activity. Mesalamine enemas or suppositories are<br />

effective as first-line therapy/maintenance therapy<br />

for mild or moderately active left-sided ulcerative<br />

colitis or proctitis, respectively. 152 Rectal mesalamine<br />

may be superior to oral mesalamine in<br />

the treatment of active ulcerative proctitis. 177<br />

Mesalamine enemas may be superior to rectal<br />

corticosteroids178 and are also effective in treating<br />

distal colitis that is unresponsive to oral ASAs or<br />

corticosteroids. 179,180 Combination therapy with<br />

oral and topical mesalamine is more effective than<br />

one agent alone in the treatment of mild-to-moderate<br />

distal colitis. 181 Mesalamine suppositories<br />

spread to the upper rectum, and enemas and foams


402<br />

Ulcerative colitis<br />

can reach the splenic flexure or into the distal<br />

transverse colon. 179,180<br />

Corticosteroid suppositories or enemas also can be<br />

used as first-line induction therapy in patients<br />

with mild or moderately active ulcerative proctitis<br />

or left-sided ulcerative colitis. 152 Rectal administration<br />

of hydrocortisone or prednisolone permits<br />

more direct delivery of steriods to distal ulcerative<br />

colitis sites; however, as with oral steroid therapy,<br />

prolonged treatment with topical steroids may<br />

induce systemic steroid side-effects, including<br />

adrenal suppression. 176,180 Topical agents such as<br />

budesonide enemas, may induce remission in<br />

distal colitis with fewer systemic steroid sideeffects.<br />

161,182 In a randomized, double-blind,<br />

placebo-controlled trial, budesonide enema<br />

therapy (2.0mg or 8.0mg/enema, once per<br />

evening) effectively induced remission, with<br />

significant improvement in sigmoidoscopy and<br />

histopathology scores, compared to placebo in<br />

adults with active distal ulcerative colitis/proctitis.<br />

161 However, in another controlled trial, less<br />

frequent dosing of twice weekly budesonide<br />

enemas (2.0mg/enema) was not superior to<br />

placebo in the maintenance of remission. 183 Some<br />

evidence suggests that ASA enemas may be<br />

superior to hydrocortisone enemas. 175,178,180<br />

Cyclosporin-A enemas have been used in the treatment<br />

of severe refractory distal ulcerative<br />

colitis, 184 but were not found to be effective in a<br />

placebo-controlled trial for mildly to moderately<br />

active left-sided ulcerative colitis. 185<br />

Maintenance therapy<br />

5-Aminosalicylic acid agents<br />

Medication for the prevention of relapse after<br />

induction of remission is often started before the<br />

induction therapy is discontinued. The clinician<br />

usually aims towards a transition of the patient<br />

from corticosteroids onto sulfasalazine or another<br />

ASA agent. Multiple studies of adults with ulcerative<br />

colitis demonstrate the effectiveness of<br />

sulfasalazine and other ASA agents in preventing<br />

relapse. 147 Mesalamine is well tolerated in the<br />

long-term treatment of children with IBD, with the<br />

principal adverse event being exacerbation of diarrhea.<br />

186 Sulfasalazine and newer ASAs are all<br />

effective in maintaining remission in ulcerative<br />

colitis. 149 Balsalazide, at a higher dose (6g/day),<br />

may be more effective in preventing relapses of<br />

ulcerative colitis in adults, compared to lowerdose<br />

balsalazide (3g/day) and mesalamine<br />

(1.5g/day). 187 Topical mesalamine can effectively<br />

prevent relapse of active distal ulcerative colitis, 188<br />

but because of the rectal route of administration,<br />

patients may prefer oral mesalamine for maintenance<br />

therapy. The combination of oral and topical<br />

5-ASA therapy may be more effective in preventing<br />

relapse than oral 5-ASA therapy alone, especially<br />

for distal disease. 189 Some authors suggest<br />

that 5-ASA agents may reduce the risk of colorectal<br />

cancer. 190–192 Children with ulcerative colitis<br />

require years of maintenance therapy. The exact<br />

duration that an ulcerative colitis patient in remission<br />

should remain on maintenance therapy is<br />

unclear, and there are no formal guidelines on<br />

when or whether maintenance therapy should be<br />

discontinued. The risk of discontinuing maintenance<br />

medication is the possibility of relapse. In<br />

addition, maintenance with 5-ASA or other agents<br />

may reduce the risk of colorectal neoplasia. 193<br />

6-Mercaptopurine and azathioprine<br />

The immunomodulatory drugs azathioprine and<br />

its metabolite 6-mercaptopurine can reduce<br />

disease activity and allow the withdrawal of<br />

steroid therapy in children with steroid-dependent<br />

ulcerative colitis. 194,195 In a small, open-label trial<br />

of children with Crohn’s disease or ulcerative<br />

colitis, six of nine patients with ulcerative colitis<br />

reduced their steroid use by at least 75% with<br />

azathioprine therapy. 194 In another series of 16<br />

children with severe, steroid-dependent or steroidrefractory<br />

ulcerative colitis, 6-mercaptopurine or<br />

azathioprine therapy allowed the discontinuation<br />

of steroid use in 75%, and 67% remained without<br />

steroid therapy for 3–65 months. 195 Given their<br />

steroid-sparing effects194,195 and reasonable tolerance<br />

by children with IBD, 196 azathioprine and 6mercaptopurine<br />

offer an alternative maintenance<br />

treatment of IBD in children. In a study of 95 children<br />

with either Crohn’s disease or ulcerative<br />

colitis, only 18% required discontinuation of the<br />

medication; the majority of side-effects responded<br />

to dose reduction, or improved spontaneously. 196<br />

Side-effects reported include aminotransferase<br />

elevations or hepatitis, pancreatitis, bone marrow<br />

depression, hypersensitivity reactions and recurrent<br />

infections; 196,197 children should be moni-


tored for the development of these complications.<br />

Some clinicians have utilized 6-mercaptopurine<br />

metabolite levels to monitor responsiveness,<br />

compliance and potential toxicity. 198.199 Evaluation<br />

for thiopurine methyltransferase genetic<br />

polymorphism should be obtained prior to the<br />

institution of 6-mercaptopurine or azathioprine<br />

therapy as it can identify those children at higher<br />

risk for drug toxicity 200 and guide the clinician to<br />

prescribe lower doses of azathioprine and 6mercaptopurine,<br />

if appropriate.<br />

Given the high relapse rate with withdrawal of 6mercaptopurine<br />

in adults with ulcerative colitis, 201<br />

the majority of children requiring azathioprine or<br />

6-mercaptopurine to suppress disease activity will<br />

probably require long-term maintenance therapy<br />

with these agents. There are no good data in ulcerative<br />

colitis addressing the question of when (or<br />

whether) immunomodulators should be discontinued<br />

after a patient has entered remission. Most<br />

patients are continued on the medication for<br />

several years if they respond to 6-mercaptopurine<br />

or azathioprine. At the present time, evidence does<br />

not suggest any definitive increased risk of malignancy<br />

secondary to long-term use of azathioprine<br />

in adults. 202–204 However, one paper has identified<br />

a slightly increased risk of Epstein–Barr virusassociated<br />

lymphoma in a large cohort of patients<br />

treated with long-term 6-mercaptopurine or<br />

azathioprine. 205<br />

Methotrexate<br />

Methotrexate appears to be less effective for the<br />

treatment of ulcerative colitis than for Crohn’s<br />

disease. 206,207 Although open-label trials in adults<br />

with ulcerative colitis suggested a benefit of<br />

methotrexate in the induction of remission, 137,208 a<br />

double-blind, randomized trial failed to show any<br />

advantage of methotrexate in the induction or<br />

maintenance of remission in adults with chronic<br />

active ulcerative colitis in comparison to<br />

placebo. 209 Another study of two different dosages<br />

of parenteral methotrexate in adults with IBD<br />

showed a remission rate of approximately 20%. 210<br />

While methotrexate can be useful in treating children<br />

with Crohn’s disease intolerant to 6-mercaptopurine,<br />

there are currently no published studies<br />

describing its efficacy in children with ulcerative<br />

colitis. 211<br />

Other therapies<br />

Treatment options 403<br />

Despite the potential role of infectious agents in<br />

the pathogenesis of ulcerative colitis, 17 the use of<br />

antibiotic therapy in the treatment of ulcerative<br />

colitis remains controversial. There is a lack of<br />

consistent evidence of the effectiveness of antibiotics<br />

in the induction and maintenance of remission<br />

in ulcerative colitis. 165,212–215 In one study,<br />

oral tobramycin therapy improved short-term clinical<br />

and histological outcomes, 212 but there was no<br />

advantage in the prevention of relapse compared<br />

to placebo. 213 In two studies of active ulcerative<br />

colitis, the addition of 10–14 days of oral or intravenous<br />

ciprofloxacin to corticosteroid therapy did<br />

not improve rates of remission. 167,216 In another<br />

placebo-controlled study, the administration of 6<br />

months of treatment with ciprofloxacin, in addition<br />

to standard therapy with prednisone and<br />

mesalamine, resulted in a greater clinical response<br />

compared to placebo; however, this advantage was<br />

not sustained after the cessation of<br />

ciprofloxacin. 217 Empiric broad-spectrum antibiotics<br />

are often administered in the setting of severe<br />

active ulcerative colitis, 60,164 especially if there is<br />

concern for potential fulminant colitis or toxic<br />

megacolon. 122<br />

In open, uncontrolled trials, infliximab (chimeric<br />

monoclonal antibody to TNF-α) therapy resulted<br />

in clinical improvement in adults and children<br />

with ulcerative colitis. 218–222 However, the clinical<br />

response of infliximab therapy may not be<br />

sustained. 223 In an open-label study of nine children<br />

and adolescents with moderate-to-severe<br />

ulcerative colitis, seven children (77%) showed a<br />

decrease in disease activity measured by the<br />

Physician Global Assessment, and corticosteroid<br />

therapy was discontinued in six children (66%). 222<br />

A small, double-blind, placebo-controlled clinical<br />

trial of infliximab in 11 adults with severe, active<br />

steroid-refractory ulcerative colitis suggested a<br />

clinical benefit for patients treated with one dose<br />

of infliximab (5mg, 10mg or 20mg/kg per dose),<br />

compared to those who received placebo. 224 In this<br />

study, four of eight ulcerative colitis patients<br />

receiving infliximab improved, compared to none<br />

of three receiving placebo. Because of poor recruitment,<br />

the trial was terminated prematurely and no<br />

statistical analysis was performed. In a larger,<br />

randomized, double-blind, placebo-controlled<br />

study of 43 adults with moderately active gluco-


404<br />

Ulcerative colitis<br />

corticoid-resistant ulcerative colitis, there was no<br />

significant difference in remission rate or sigmoidoscopic<br />

score between the infliximab-treated<br />

group (5mg/kg per dose, at weeks 0 and 2) and the<br />

placebo group. 225 Thus, the therapeutic benefit of<br />

infliximab in ulcerative colitis remains unclear at<br />

this time, and the treatment does carry a risk of<br />

infusion reactions, antinuclear antibody formation<br />

and opportunistic infections. 226–230<br />

Studies evaluating the effectiveness of mycophenolate<br />

mofetil therapy for ulcerative colitis show<br />

mixed results and may suggest increased sideeffects<br />

compared to other immunomodulatory<br />

agents such as azathioprine. 231–233<br />

Probiotics have been studied in the maintenance<br />

of remission in adults with ulcerative colitis. 234–236<br />

In an open-label trial of the probiotic VSL no. 3,<br />

performed in adults with inactive ulcerative<br />

colitis, 75% of patients remained in remission<br />

during the 12-month study, 234 but no controlled<br />

trials with VSL no. 3 for maintenance therapy for<br />

ulcerative colitis have been performed. In two<br />

randomized controlled comparison trials, nonpathogenic<br />

E. coli strains and mesalamine maintained<br />

similar rates of remission in adults with<br />

quiescent ulcerative colitis. 235,236 There are no<br />

formal studies on the effectiveness of probiotics in<br />

children with ulcerative colitis.<br />

In adults with ulcerative colitis, transdermal nicotine<br />

therapy, combined with mesalamine or corticosteroids,<br />

may result in clinical improvement,<br />

but is associated with unwanted side-effects. 237,238<br />

Transdermal nicotine therapy does not appear to<br />

be effective in the maintenance of remission of<br />

ulcerative colitis. 239 An open-label trial of nicotine<br />

enema therapy in adults with ulcerative colitis<br />

showed improvement in symptoms of urgency and<br />

stool frequency, and sigmoidoscopic and histological<br />

scores, 240 but controlled studies are needed to<br />

determine true efficacy.<br />

Several placebo-controlled studies suggested a<br />

benefit of adjunctive therapy with fish oil supplementation<br />

containing eicosapentaenoic acid, a<br />

potent inhibitor of leukotriene B4 synthesis, in the<br />

treatment of active ulcerative colitis. 241–243<br />

However, fish oil supplementation did not appear<br />

to show any benefit in maintenance therapy. 243,244<br />

Unfortunately, given the large number of capsules<br />

required for daily therapy and the intolerance to<br />

unwanted fishy odor, patients’ compliance has<br />

been suboptimal.<br />

Randomized, controlled trials in adults with active<br />

distal ulcerative colitis suggest that therapy with<br />

topical short-chain fatty acid (SCFA) preparations<br />

result in clinical symptomatic improvement, but<br />

there is no statistically significant advantage in<br />

comparison to placebo. 245–247 In one double-blind,<br />

placebo-controlled, 6-week trial of rectal SCFA,<br />

103 patients with distal ulcerative colitis were<br />

entered and those on SCFA had larger, but statistically<br />

non-significant, reductions in every component<br />

of their clinical and histological activity<br />

scores. 246<br />

In several open-label trials, adults with steroidresistant<br />

ulcerative colitis showed a clinical<br />

response to heparin therapy; 248–250 however, a<br />

controlled trial did not show any advantage of<br />

heparin treatment for moderate-to-severe ulcerative<br />

colitis, compared to corticosteroid therapy. 251<br />

In an open-label pilot study, daclizumab (humanized<br />

anti-IL-2R antibody; CD25) resulted in clinical<br />

and endoscopic improvement in adults with<br />

refractory ulcerative colitis; 252 further study is<br />

needed to determine the effectiveness of this novel<br />

therapy.<br />

Nutritional therapy<br />

The importance of nutrition in the management of<br />

ulcerative colitis is extensively reviewed elsewhere.<br />

253,254 Children with ulcerative colitis can<br />

develop nutritional deficits with poor oral intake<br />

secondary to symptoms; and thus, promotion of<br />

continued good nutritional intake is essential for<br />

appropriate healing and nutritional repletion. 255<br />

Enteral nutrition is preferred to total parenteral<br />

nutrition when possible. In contrast to Crohn’s<br />

disease, enteral nutrition is not an effective<br />

primary therapy for active ulcerative colitis. 152,171<br />

Studies suggest no advantage of total nutritional<br />

support and bowel rest in addition to conventional<br />

medical therapy alone in the treatment of ulcerative<br />

colitis. 256,257 In a randomized, controlled trial<br />

of corticosteroid therapy combined with either<br />

polymeric enteral nutrition or total parenteral<br />

nutrition, adults with moderate or severe ulcerative<br />

colitis showed no differences in remission<br />

rate, need for colectomy, or changes in anthropo-


metric parameters. 258 Total parenteral nutrition is<br />

often utilized for nutritional repletion in severe<br />

ulcerative colitis, especially if the patient develops<br />

severe cramps and diarrhea when challenged with<br />

enteral nutrition.<br />

Surgical therapy<br />

In the majority of cases, medical therapy remains<br />

the first-line treatment for ulcerative colitis.<br />

However, colectomy may be required for patients<br />

with severe or medically refractory disease, or to<br />

prevent colon cancer. Since the inflammation in<br />

ulcerative colitis is limited to the colon, colonic<br />

resection will most often result in resolution of<br />

symptoms. However, colectomy is not without<br />

potential complications, such as the development<br />

of pouchitis in patients who undergo ileoanal<br />

anastomosis. 259,260 It is important to consider<br />

timely surgical intervention in the appropriate<br />

setting to avoid complications of ulcerative colitis.<br />

Indications for colectomy in a patient with ulcerative<br />

colitis include fulminant colitis or a complication<br />

of colitis, such as massive hemorrhage, perforation,<br />

or toxic megacolon; medical therapy<br />

failure; steroid dependency, which may lead to<br />

undesired side-effects; and the presence of colonic<br />

dysplasia. 261 Prepubertal children may experience<br />

catch-up growth after colectomy for ulcerative<br />

colitis. In one series, 11 of 18 children increased<br />

their median height velocity from 3.85cm/year<br />

preoperatively to 7.35cm/year postoperatively. 262<br />

As medical treatment for ulcerative colitis was<br />

developed, the frequency of colectomy decreased.<br />

At one center, a retrospective review of children<br />

and adolescents with ulcerative colitis revealed a<br />

decrease in the frequency of colectomy from<br />

48.9% (between 1955 and 1964) to 26.2% (between<br />

1965 and 1974). 263<br />

There are no early predictors to help determine<br />

who will proceed to colectomy. Hyams et al, in a<br />

retrospective review, reported that the 5-year<br />

colectomy rate in patients with mild disease at<br />

presentation was 8%, compared to 26% in patients<br />

with moderate-to-severe disease at presentation. 264<br />

In another retrospective review of 73 children with<br />

ulcerative colitis between the ages of 1 and 18<br />

years, the combination of steroid dependency and<br />

pancolitis was associated with an increased need<br />

Treatment options 405<br />

for colectomy. 265 Seventy-three per cent of the<br />

children with steroid-dependent pancolitis<br />

required colectomy within 3 years of diagnosis. 265<br />

Except in the setting of emergency colectomy, a<br />

complete evaluation should be performed to<br />

ensure that there is no evidence of Crohn’s disease<br />

prior to colectomy. If there is evidence suggesting<br />

the possibility of Crohn’s disease, the patient and<br />

family need to be informed of the potential for<br />

postoperative recurrence, and the relative<br />

contraindications of ileoanal pull-through procedures<br />

in patients with known Crohn’s disease. The<br />

authors recommend an upper gastrointestinal<br />

series with small-bowel follow-through and upper<br />

gastrointestinal endoscopy, in addition to a full<br />

colonoscopy with ileoscopy, if there is no clinical<br />

contraindication. Prior endoscopies and pathology<br />

reports should be carefully reviewed to establish<br />

that there is no evidence of Crohn’s disease.<br />

The surgical options available for ulcerative colitis<br />

are reviewed in detail elsewhere. 261 The ileal<br />

pouch–anal canal anastomosis (IPAA) is the operation<br />

most commonly performed in the majority of<br />

patients with ulcerative colitis. The IPAA removes<br />

the entire colon and the rectal mucosa, avoids<br />

permanent ileostomy, and preserves anorectal<br />

function. Several types of ileal pouches can be<br />

constructed, including the J-shaped, S-shaped, Wshaped<br />

and the lateral–lateral pouch. 261 The Jpouch<br />

design is now most commonly used for the<br />

IPAA operation. 266,267 If the rectal mucosa is in<br />

good condition, many centers use the two-stage<br />

operative approach. During the first stage, a subtotal<br />

colectomy of the cecum to proximal rectum,<br />

the removal of the distal rectal and proximal anal<br />

mucosa, and the formation of the ileal pouch are<br />

performed. In this initial stage, a diverting loop<br />

ileostomy is performed in order to allow the pouch<br />

to heal. The second stage involves closure of the<br />

loop ileostomy with restoration of fecal flow to the<br />

pouch. Surgeons at some surgical centers also<br />

complete the IPAA in one stage, without the loop<br />

ileostomy; 268 however, this would not be the<br />

procedure of choice in patients receiving highdose<br />

corticosteroids. 267 Some authors 261 believe<br />

that the omission of the diverting ileostomy may<br />

increase the risk of anastomotic leaks and prolong<br />

recovery; 269 thus, candidate patients for the onestage<br />

IAPP should be selected carefully. 270


406<br />

Ulcerative colitis<br />

If the patient presents for emergency surgical intervention,<br />

such as with fulminant colitis, a threestage<br />

operative approach is often utilized. At the<br />

time of acute presentation, a subtotal colectomy is<br />

performed with formation of a rectal stump (the<br />

so-called Hartman pouch) and Brooke ileostomy.<br />

After the first operation, the rectal mucosa is<br />

treated with topical therapies (e.g. hydrocortisone,<br />

aminosalicylates) to induce mucosal healing. At<br />

the second operation, the distal rectal and proximal<br />

anal mucosa is removed and the ileal pouch is<br />

created. The ileostomy is reversed at the third<br />

operation.<br />

The potential complications of IPAA include<br />

small-bowel obstruction, pelvic sepsis, anastomotic<br />

leak, fecal incontinence, pouchitis, strictures<br />

or fistulae. 261,270,271 The development of<br />

fistulae raises the suspicion of Crohn’s disease. 261<br />

In one series of children aged 9–16 who underwent<br />

proctocolectomy with IPAA, 12/29 (41%) of<br />

patients with ulcerative colitis developed early<br />

complications (wound infection, early bowel<br />

obstruction, prolonged fever). 272 Late complications<br />

(bowel obstruction, pouch fistula) occurred<br />

in 11/29 (38%) and pouchitis developed in 9/29<br />

(31%) of the children with ulcerative colitis.<br />

Median follow-up was 4 years (range 6 months to<br />

9 years). In this same study, daytime continence<br />

was noted in 100% and night-time continence in<br />

93%. The median frequency of bowel movements<br />

was four in 24h, and 7% of patients had night-time<br />

bowel movements.<br />

Pouchitis, or inflammation of the newly created<br />

reservoir, is the most significant chronic complication<br />

in ulcerative colitis patients undergoing IPAA;<br />

as many as 44–53% of children and young adults<br />

with ulcerative colitis and ileoanal anastomosis<br />

will develop pouchitis on long-term followup.<br />

259,260 The etiology of pouchitis is unknown,<br />

but theories involve the role of genetic susceptibility,<br />

fecal stasis, bacterial overgrowth, disruption of<br />

the balance of luminal bacteria, nutritional deficiencies,<br />

ischemia and IBD recurrence. 273<br />

Symptoms of pouchitis include diarrhea, rectal<br />

bleeding, abdominal cramping, urgency and<br />

incontinence of stool, malaise and fever. 261,273,274<br />

Patients with ulcerative colitis who undergo IPAA<br />

develop pouchitis more commonly than patients<br />

with familial polyposis who undergo the same<br />

procedure. 275 Pouchitis may occur more frequently<br />

Figure 25.5 Chronic active inflammation of an ileal<br />

J-pouch (pouchitis) in a patient with a history of ulcerative<br />

colitis. Note that both limbs of the ileal reservoir are<br />

erythematous with exudate.<br />

in children and adults with primary sclerosing<br />

cholangitis (PSC)-associated ulcerative colitis<br />

276,277 and in individuals with extraintestinal<br />

manifestations of ulcerative colitis. 273 Laboratory<br />

studies may demonstrate anemia and an elevated<br />

ESR. The definitive diagnosis is established by<br />

flexible endoscopy of the pouch with biopsies<br />

(Figure 25.5). In some patients, a contrast enema<br />

may be useful in identifying fistulae.<br />

Broad-spectrum antibiotics are usually the firstline<br />

treatment for pouchitis. 273,278 Metronidazole is<br />

the most commonly used antibiotic, but alternative<br />

therapies include ciprofloxacin, amoxicillin–<br />

clavulanic acid, erythromycin and tetracycline.<br />

274,279 If there is no improvement with antibiotics,<br />

other options include mesalamine enemas<br />

and steroid enemas or oral therapy with<br />

mesalamine, sulfasalazine or steroids. 274,280,281<br />

Other therapies examined include cyclosporin<br />

enemas, SCFA enemas, butyrate suppositories and<br />

glutamine suppositories. 184,274,282,283<br />

Probiotic therapy may prevent the onset of acute<br />

pouchitis after ileostomy closure 284 and effectively


maintain remission after chronic pouchitis. 285 A<br />

double-blind, placebo-controlled study evaluated<br />

the efficacy of a probiotic preparation VSL no. 3,<br />

(containing 5 x 10 11 per gram of viable lyophilized<br />

bacteria of four strains of lactobacilli, three strains<br />

of bifidobacteria, and one strain of Streptococcus<br />

salivarius subsp. thermophilus), compared with<br />

placebo in maintenance of remission of chronic<br />

pouchitis in 40 patients in clinical and endoscopic<br />

remission. Three patients (15%) in the VSL no. 3<br />

group had relapses within the 9-month follow-up<br />

period, compared with 20 (100%) in the placebo<br />

group. 285 In another double-blind, placebocontrolled<br />

study performed by the same authors in<br />

40 patients, VSL no. 3, administered immediately<br />

after ileostomy closure for 1 year, effectively<br />

reduced the onset of acute pouchitis in the VSL no.<br />

3 group (10%) in comparison to the placebo group<br />

(40%). 284<br />

Several studies have suggested that the risk of<br />

dysplasia in the ileal pouch appears to be<br />

low 286,287 and may be associated with chronic<br />

pouchitis. 288 The development of adenocarcinoma<br />

has been reported in the ileal pouch. 289 In a followup<br />

study (mean of 5 years) of 76 children and<br />

adolescents with ulcerative colitis who had an<br />

IPAA, no dysplasia was identified in screening<br />

pouch biopsy specimens. 286 The authors<br />

cautioned that the long-term risk of development<br />

of dysplasia is not yet known and recommended<br />

screening of the pouch for dysplasia. In the rare<br />

patient who has undergone an ileorectal anastomosis<br />

without rectal mucosectomy, surveillance of<br />

the rectum should be performed to screen for<br />

rectal cancer.<br />

Psychosocial support<br />

The social impact of ulcerative colitis on the lives<br />

of children with the disease needs to be considered.<br />

Ulcerative colitis often has its onset in<br />

adolescence, a time when body image issues are<br />

paramount. Children and teenagers with IBD may<br />

often experience anxiety over the diagnosis of a<br />

chronic disease, the need for invasive procedures<br />

and the uncertainty of the future. In addition,<br />

there may be struggles with parents about proper<br />

diet, and ‘medication fatigue’ from having to take<br />

more than ten pills per day. Social, school-related<br />

and extracurricular activities may be affected and<br />

Prognosis and follow-up 407<br />

may need appropriate modification. For example, a<br />

self-limited reduction of physical education activities<br />

and permission for special bathroom privileges<br />

may be needed.<br />

Previous studies report an increased risk of psychiatric<br />

and behavioral issues in children with IBD<br />

including depression, anxiety and low selfesteem.<br />

290–294 Burke and colleagues reported that<br />

children are at increased risk for depression as<br />

early as the time of diagnosis. 290 The physician<br />

caring for these patients needs to discuss the above<br />

issues openly with the patient and family, and be<br />

alert to the possibility that a patient may develop<br />

anxiety or depressive symptoms. More recently,<br />

two patient-generated, disease-specific, healthrelated<br />

quality of life (HRQoL) questionnaires for<br />

children with IBD have been validated: the<br />

IMPACT and Impact-II questionnaires. 295,296<br />

Impact-II is a modified version of the IMPACT<br />

questionnaire. The use of these instruments may<br />

provide important information to improve the care<br />

of children with IBD. Referral to an educational<br />

support group, such as those sponsored by the<br />

Crohn’s and Colitis Foundation of America<br />

(www.ccfa.org), may be helpful for patients and<br />

their famililies.<br />

Prognosis and follow-up<br />

Whether children with ulcerative colitis are<br />

treated medically or surgically, they have an excellent<br />

long-term prognosis and good quality of life.<br />

The majority of children with ulcerative colitis<br />

respond to medical therapy. In one American<br />

retrospective study of 171 children ranging in age<br />

from 1.5 to 17.7 years, diagnosed with ulcerative<br />

colitis between 1967 and 1994, 43% had mild<br />

disease at presentation and 57% had moderate or<br />

severe ulcerative colitis. With treatment, 70% of all<br />

the children were in remission within 3 months of<br />

diagnosis and, by 6 months, 90% of the children<br />

with mild disease and 81% of the children with<br />

moderate-to-severe ulcerative colitis experienced<br />

resolution of their symptoms. In each yearly<br />

follow-up period, approximately 55% of the children<br />

were symptom free, 38% experienced chronic<br />

intermittent symptoms and 7% had continuous<br />

symptoms. 264 In this same study, children with<br />

mild ulcerative colitis at presentation had a lower


408<br />

Ulcerative colitis<br />

frequency of colectomy than children with moderate-to-severe<br />

ulcerative colitis (9% vs. 26% at 5<br />

years, respectively). Patients with left-sided<br />

disease had comparable rates of colectomy at 5<br />

years to patients with pancolitis. 264 However, the<br />

authors cautioned that the number of patients in<br />

each subgroup of disease extent was small, and a<br />

true difference in colectomy rates may have been<br />

missed secondary to insufficient power of the<br />

study. 264 Another study of 80 children diagnosed<br />

with ulcerative colitis before the age of 15 years<br />

reported a slightly higher rate of remission of<br />

60–70% in each of the first 10 years, after the year<br />

of diagnosis. 3 In the same study, the cumulative<br />

probability of colectomy was 6% after 1 year and<br />

29% after 20 years. 3<br />

Limited distal ulcerative colitis (proctitis or proctosigmoiditis)<br />

diagnosed in adults and children<br />

can extend further to involve more proximal colon<br />

with time. 297–300 In a retrospective study of 38 children<br />

(mean age 11.6 years; range 4.2–17.7 years)<br />

diagnosed with ulcerative colitis proctitis between<br />

1975 and 1994, 29% of the children developed<br />

extension of disease involvement beyond the<br />

rectosigmoid. 299 Another retrospective evaluation<br />

of 85 patients under 21 years of age, diagnosed<br />

with ulcerative colitis proctosigmoiditis between<br />

1958 and 1983, demonstrated the extension of<br />

disease to the descending colon in 20% and to the<br />

hepatic flexure or beyond in another 38% of the<br />

patients. 300 The extension of disease may warrant<br />

a change in medical therapy to control disease<br />

activity and may have implications for an<br />

increased risk of potential colorectal cancer in the<br />

future. 301<br />

Women with ulcerative colitis generally experience<br />

similar rates of fertility to those of the general<br />

population; however, an increased risk of preterm<br />

births has been reported in some studies. 302–304 In<br />

contrast, women with ulcerative colitis who<br />

undergo proctocolectomy with IPAA may experience<br />

reduced fertility. 305<br />

Individuals with longstanding ulcerative colitis<br />

are at increased risk of developing colorectal<br />

cancer. 301,306,307 Risk of colorectal cancer increases<br />

with more extensive colonic involvement and<br />

longer duration of disease since diagnosis.<br />

301,306–309 Therefore, children with ulcerative<br />

colitis will potentially be at risk for colorectal<br />

cancer over a longer period of time, compared to<br />

individuals with adult-onset ulcerative colitis. In a<br />

population-based cohort study in Sweden, Ekbom<br />

et al reported standardized incidence ratios for<br />

colorectal cancer of 1.7 for ulcerative colitis proctitis,<br />

2.8 for left-sided ulcerative colitis and 14.8<br />

for ulcerative colitis pancolitis. 307 The absolute<br />

risk (cumulative risk) of colorectal cancer 35 years<br />

after diagnosis was 30% for patients with pancolitis<br />

at diagnosis for the entire cohort (adults and<br />

children), but 40% for individuals diagnosed with<br />

pancolitis at less than 15 years of age. 307<br />

Individuals with PSC and ulcerative colitis have a<br />

higher risk of developing colorectal neoplasia<br />

compared to those with ulcerative colitis alone,<br />

and of developing cholangiocarcinoma. 310–314<br />

Ursodeoxycholic acid (UDCA) may decrease the<br />

risk for developing colorectal dysplasia or cancer<br />

in patients with ulcerative colitis and PSC. 315<br />

Other risk factors reported for colorectal cancer in<br />

patients with ulcerative colitis include a family<br />

history of colorectal cancer 316 and the occurrence<br />

of backwash ileitis. 317 In contrast, some retrospective<br />

studies suggest that 5-ASA agents may provide<br />

a protective effect in the development of colorectal<br />

cancer in patients with ulcerative colitis, but not<br />

all studies support these findings. 190,191,193,318 A<br />

retrospective, case–control study suggested a risk<br />

reduction of colorectal cancer in ulcerative colitis<br />

with folate supplementation, but the findings were<br />

not statistically significant. 319 The role of pharmacological<br />

therapy and vitamin supplements in the<br />

development of colorectal cancer in patients with<br />

ulcerative colitis remains controversial. 320 Further<br />

prospective studies are needed to evaluate the<br />

potential risk reduction (preventive) effects of<br />

folate and 5-ASA agents. 192,321,322<br />

There are no randomized controlled trials examining<br />

the effectiveness of surveillance colonoscopy<br />

for dysplasia and colorectal cancer in patients with<br />

ulcerative colitis. 323,324 The reported general<br />

current practice includes surveillance colonoscopy<br />

every 1–2 years beginning at 8 years after<br />

the diagnosis of disease for pancolitis and 15 years<br />

in those with left-sided colitis. 324 However, it may<br />

be prudent to perform the initial surveillance<br />

colonoscopy beginning at 8–10 years after diagnosis<br />

in all patients with ulcerative colitis, in order to<br />

reassess the true extent of disease. 324 The consensus<br />

recommendations suggest that biopsy specimens


should be taken every 10cm in all four quadrants<br />

and additional biopsies should be performed for<br />

strictures and mass lesions. 324 It is not clear<br />

whether surveillance practice should differ for<br />

patients with younger-onset ulcerative colitis.<br />

There are no published formal recommendations<br />

to guide surveillance colonoscopy in children. 323<br />

Conclusions<br />

Ulcerative colitis in children presents in a similar<br />

manner to that in adults, although children often<br />

have more extensive disease at diagnosis. Once the<br />

diagnosis is established by colonoscopy, the first<br />

line of induction treatment in mild disease is<br />

usually an aminosalicylate, with corticosteroids<br />

REFERENCES<br />

1. Chong SK, Blackshaw AJ, Morson BC et al. Prospective<br />

study of colitis in infancy and early childhood. J Pediatr<br />

Gastroenterol Nutr 1986; 5: 352–358.<br />

2. Mendeloff AI, Calkins BM. The epidemiology of idiopathic<br />

inflammatory bowel disease. In Kirsner JB,<br />

Shorter RG, eds. Inflammatory Bowel Disease.<br />

Philadelphia: Lea & Febiger, 1988: 3–34.<br />

3. Langholz E, Munkholm P, Krasilnikoff PA et al.<br />

Inflammatory bowel diseases with onset in childhood.<br />

Clinical features, morbidity, and mortality in a regional<br />

cohort. Scand J Gastroenterol 1997; 32: 139–147.<br />

4. Gryboski JD. Ulcerative colitis in children 10 years old<br />

or younger. J Pediatr Gastroenterol Nutr 1993; 17: 24–31.<br />

5. Mamula P, Telega GW, Markowitz JE et al. Inflammatory<br />

bowel disease in children 5 years of age and younger.<br />

Am J Gastroenterol 2002; 97: 2005–2010.<br />

6. Barton JR, Gillon S, Ferguson A. Incidence of inflammatory<br />

bowel disease in Scottish children between 1968<br />

and 1983; marginal fall in ulcerative colitis, three-fold<br />

rise in Crohn’s disease. Gut 1989; 30: 618–622.<br />

7. Calkins BM, Lilienfeld AM, Garland CF et al. Trends in<br />

incidence rates of ulcerative colitis and Crohn’s disease.<br />

Dig Dis Sci 1984; 29: 913–920.<br />

8. Lindberg E, Lindquist B, Holmquist L et al.<br />

Inflammatory bowel disease in children and adolescents<br />

in Sweden, 1984–1995. J Pediatr Gastroenterol Nutr<br />

2000; 30: 259–264.<br />

Conclusions 409<br />

reserved for moderate-to-severe disease. If a<br />

patient responds to induction therapy, the maintenance<br />

agent most commonly used is aminosalicylates<br />

or 6-mercaptopurine. In severe steroidunresponsive<br />

colitis, the clinician is faced with the<br />

challenge of recommending either colectomy or<br />

more aggressive immunosuppression (e.g. cyclosporin).<br />

Even if a patient responds to cyclosporin,<br />

there is a high likelihood of proceeding to colectomy<br />

within a year. Surgery will eradicate the<br />

disease, but patients may develop chronic pouchitis<br />

requiring additional medical treatment. In addition<br />

to medical care of these patients, nutritional<br />

counseling, monitoring of growth, and psychosocial<br />

support are essential. If the medical team can<br />

give proper care and support, the patient’s longterm<br />

prognosis is usually excellent.<br />

9. Olafsdottir EJ, Fluge G, Haug K. Chronic inflammatory<br />

bowel disease in children in western Norway. J Pediatr<br />

Gastroenterol Nutr 1989; 8: 454–458.<br />

10. Bentsen BS, Moum B, Ekbom A. Incidence of inflammatory<br />

bowel disease in children in southeastern Norway:<br />

a prospective population-based study 1990–94. Scand J<br />

Gastroenterol 2002; 37: 540–545.<br />

11. Askling J, Grahnquist L, Ekbom A et al. Incidence of<br />

paediatric Crohn’s disease in Stockholm, Sweden.<br />

Lancet 1999; 354: 1179.<br />

12. Sonnenberg A, McCarty DJ, Jacobsen SJ. Geographic<br />

variation of inflammatory bowel disease within the<br />

United States. Gastroenterology 1991; 100: 143–149.<br />

13. Shivananda S, Lennard-Jones J, Logan R et al. Incidence<br />

of inflammatory bowel disease across Europe: is there a<br />

difference between north and south? Results of the<br />

European Collaborative Study on Inflammatory Bowel<br />

Disease (EC-IBD). Gut 1996; 39: 690–697.<br />

14. Acheson ED. The distribution of ulcerative colitis and<br />

regional enteritis in United States veterans with particular<br />

reference to the Jewish religion. Gut 1960; 1:<br />

291–293.<br />

15. Gilat T, Grossman A, Fireman Z et al. Inflammatory<br />

bowel disease in Jews. Front Gastrointest Res 1986; 11:<br />

141–145.<br />

16. Fiocchi C. Inflammatory bowel disease: etiology and<br />

pathogenesis. Gastroenterology 1998; 115: 182–205.


410<br />

Ulcerative colitis<br />

17. Hendrickson BA, Gokhale R, Cho JH. Clinical aspects<br />

and pathophysiology of inflammatory bowel disease.<br />

Clin Microbiol Rev 2002; 15: 79–94.<br />

18. Orholm M, Binder V, Sorensen TI et al. Concordance of<br />

inflammatory bowel disease among Danish twins.<br />

Results of a nationwide study. Scand J Gastroenterol<br />

2000; 35: 1075–1081.<br />

19. Halfvarson J, Bodin L, Tysk C et al. Inflammatory bowel<br />

disease in a Swedish twin cohort: a long-term follow-up<br />

of concordance and clinical characteristics.<br />

Gastroenterology 2003; 124: 1767–1773.<br />

20. Orholm M, Munkholm P, Langholz E et al. Familial<br />

occurrence of inflammatory bowel disease. N Engl J Med<br />

1991; 324: 84–88.<br />

21. Yang H, McElree C, Roth MP et al. Familial empirical<br />

risks for inflammatory bowel disease: differences<br />

between Jews and non-Jews. Gut 1993; 34: 517–524.<br />

22. Annese V, Andreoli A, Astegiano M et al. Clinical<br />

features in familial cases of Crohn’s disease and ulcerative<br />

colitis in Italy: a GISC study. Italian Study Group<br />

for the Disease of Colon and Rectum. Am J Gastroenterol<br />

2001; 96: 2939–2945.<br />

23. Satsangi J, Parkes M, Louis E et al. Two stage genomewide<br />

search in inflammatory bowel disease provides<br />

evidence for susceptibility loci on chromosomes 3, 7<br />

and 12. Nature Genet 1996; 14: 199–202.<br />

24. Farrell RJ, Peppercorn MA. Ulcerative colitis. Lancet<br />

2002; 359: 331–340.<br />

25. Papadakis KA, Targan SR. Current theories on the<br />

causes of inflammatory bowel disease. Gastroenterol<br />

Clin North Am 1999; 28: 283–296.<br />

26. Vessey M, Jewell D, Smith A et al. Chronic inflammatory<br />

bowel disease, cigarette smoking, and use of oral<br />

contraceptives: findings in a large cohort study of<br />

women of childbearing age. Br Med J (Clin Res Ed) 1986;<br />

292: 1101–1103.<br />

27. Calkins BM. A meta-analysis of the role of smoking in<br />

inflammatory bowel disease. Dig Dis Sci 1989; 34:<br />

1841–1854.<br />

28. Koletzko S, Griffiths A, Corey M et al. Infant feeding<br />

practices and ulcerative colitis in childhood. BMJ 1991;<br />

302: 1580–1581.<br />

29. Andersson RE, Olaison G, Tysk C et al. Appendectomy<br />

and protection against ulcerative colitis. N Engl J Med<br />

2001; 344: 808–814.<br />

30. Duggan AE, Usmani I, Neal KR et al. Appendicectomy,<br />

childhood hygiene, <strong>Helicobacter</strong> <strong>pylori</strong> status, and risk<br />

of inflammatory bowel disease: a case control study. Gut<br />

1998; 43: 494–498.<br />

31. Feeney MA, Murphy F, Clegg AJ et al. A case–control<br />

study of childhood environmental risk factors for the<br />

development of inflammatory bowel disease. Eur J<br />

Gastroenterol Hepatol 2002; 14: 529–534.<br />

32. Evans JM, McMahon AD, Murray FE et al. Non-steroidal<br />

anti-inflammatory drugs are associated with emergency<br />

admission to hospital for colitis due to inflammatory<br />

bowel disease. Gut 1997; 40: 619–622.<br />

33. Kaufmann HJ, Taubin HL. Nonsteroidal anti-inflammatory<br />

drugs activate quiescent inflammatory bowel<br />

disease. Ann Intern Med 1987; 107: 513–516.<br />

34. Rampton DS, Sladen GE. Relapse of ulcerative proctocolitis<br />

during treatment with non-steroidal anti-inflammatory<br />

drugs. Postgrad Med J 1981; 57: 297–299.<br />

35. Bonner GF. Exacerbation of inflammatory bowel disease<br />

associated with use of celecoxib. Am J Gastroenterol<br />

2001; 96: 1306–1308.<br />

36. Felder JB, Korelitz BI, Rajapakse R et al. Effects of nonsteroidal<br />

antiinflammatory drugs on inflammatory<br />

bowel disease: a case–control study. Am J Gastroenterol<br />

2000; 95: 1949–1954.<br />

37. Bonner GF, Walczak M, Kitchen L et al. Tolerance of<br />

nonsteroidal antiinflammatory drugs in patients with<br />

inflammatory bowel disease. Am J Gastroenterol 2000;<br />

95: 1946–1948.<br />

38. Reuter BK, Asfaha S, Buret A et al. Exacerbation of<br />

inflammation-associated colonic injury in rat through<br />

inhibition of cyclooxygenase-2. J Clin Invest 1996; 98:<br />

2076–2085.<br />

39. Wallace JL, Keenan CM, Gale D et al. Exacerbation of<br />

experimental colitis by nonsteroidal anti-inflammatory<br />

drugs is not related to elevated leukotriene B4 synthesis.<br />

Gastroenterology 1992; 102: 18–27.<br />

40. Wallace JL. Nonsteroidal anti-inflammatory drugs and<br />

gastroenteropathy: the second hundred years.<br />

Gastroenterology 1997; 112: 1000–1016.<br />

41. Corrao G, Tragnone A, Caprilli R et al. Risk of inflammatory<br />

bowel disease attributable to smoking, oral<br />

contraception and breastfeeding in Italy: a nationwide<br />

case–control study. Cooperative Investigators of the<br />

Italian Group for the Study of the Colon and the<br />

Rectum (GISC). Int J Epidemiol 1998; 27: 397–404.<br />

42. Gilat T, Hacohen D, Lilos P et al. Childhood factors in<br />

ulcerative colitis and Crohn’s disease. An international<br />

cooperative study. Scand J Gastroenterol 1987; 22:<br />

1009–1024.<br />

43. The Epidemiology Group of the Research Committee of<br />

Inflammatory Bowel Disease in Japan. A case–control<br />

study of ulcerative colitis in relation to dietary and<br />

other factors in Japan. The Epidemiology Group of the<br />

Research Committee of Inflammatory Bowel Disease in<br />

Japan. J Gastroenterol 1995; 30: 9–12.<br />

44. Jarnerot G, Jarnmark I, Nilsson K. Consumption of<br />

refined sugar by patients with Crohn’s disease, ulcerative<br />

colitis, or irritable bowel syndrome. Scand J<br />

Gastroenterol 1983; 18: 999–1002.<br />

45. Lashner BA, Kane SV, Hanauer SB. Lack of association<br />

between oral contraceptive use and ulcerative colitis.<br />

Gastroenterology 1990; 99: 1032–1036.<br />

46. Bouma G, Strober W. The immunological and genetic<br />

basis of inflammatory bowel disease. Nat Rev Immunol<br />

2003; 3: 521–533.<br />

47. Plevy S. The immunology of inflammatory bowel<br />

disease. Gastroenterol Clin North Am 2002; 31: 77–92.<br />

48. Sartor RB. Current concepts of the etiology and pathogenesis<br />

of ulcerative colitis and Crohn’s disease.<br />

Gastroenterol Clin North Am 1995; 24: 475–507.<br />

49. Fuss IJ, Neurath M, Boirivant M et al. Disparate CD4+<br />

lamina propria (LP) lymphokine secretion profiles in<br />

inflammatory bowel disease. Crohn’s disease LP cells<br />

manifest increased secretion of IFN-gamma, whereas<br />

ulcerative colitis LP cells manifest increased secretion<br />

of IL-5. J Immunol 1996; 157: 1261–1270.<br />

50. Izzo RS, Witkon K, Chen AI et al. Interleukin-8 and<br />

neutrophil markers in colonic mucosa from patients<br />

with ulcerative colitis. Am J Gastroenterol 1992; 87:<br />

1447–1452.<br />

51. Daig R, Andus T, Aschenbrenner E et al. Increased<br />

interleukin 8 expression in the colon mucosa of patients<br />

with inflammatory bowel disease. Gut 1996; 38:<br />

216–222.<br />

52. Keshavarzian A, Fusunyan RD, Jacyno M et al.<br />

Increased interleukin-8 (IL-8) in rectal dialysate from<br />

patients with ulcerative colitis: evidence for a biological<br />

role for IL-8 in inflammation of the colon. Am J<br />

Gastroenterol 1999; 94: 704-712.<br />

53. MacDermott RP. Chemokines in the inflammatory bowel<br />

diseases. J Clin Immunol 1999; 19: 266–272.<br />

54. Luster AD. Chemokines – chemotactic cytokines that<br />

mediate inflammation. N Engl J Med 1998; 338:<br />

436–445.


55. Wallace JL. Prostaglandin biology in inflammatory<br />

bowel disease. Gastroenterol Clin North Am 2001; 30:<br />

971–980.<br />

56. Rao SS, Holdsworth CD, Read NW. Symptoms and stool<br />

patterns in patients with ulcerative colitis. Gut 1988; 29:<br />

342–345.<br />

57. Heikenen JB, Werlin SL, Brown CW et al. Presenting<br />

symptoms and diagnostic lag in children with inflammatory<br />

bowel disease. Inflamm Bowel Dis 1999; 5:<br />

158–160.<br />

58. Motil KJ, Grand RJ. Ulcerative colitis and Crohn disease<br />

in children. Pediatr Rev 1987; 9: 109–120.<br />

59. Grand RJ, Homer DR. Approaches to inflammatory<br />

bowel disease in childhood and adolescence. Pediatr<br />

Clin North Am 1975; 22: 835–850.<br />

60. Werlin SL, Grand RJ. Severe colitis in children and<br />

adolescents: diagnosis. Course, and treatment.<br />

Gastroenterology 1977; 73: 828–832.<br />

61. Sachar DB, Smith H, Chan S et al. Erythrocytic sedimentation<br />

rate as a measure of clinical activity in<br />

inflammatory bowel disease. J Clin Gastroenterol 1986;<br />

8: 647–650.<br />

62. Macfarlane PI, Miller V, Wells F et al. Laboratory assessment<br />

of disease activity in childhood Crohn’s disease<br />

and ulcerative colitis. J Pediatr Gastroenterol Nutr 1986;<br />

5: 93–96.<br />

63. Holmquist L, Ahren C, Fallstrom SP. Relationship<br />

between results of laboratory tests and inflammatory<br />

activity assessed by colonoscopy in children and<br />

adolescents with ulcerative colitis and Crohn’s colitis. J<br />

Pediatr Gastroenterol Nutr 1989; 9: 187–193.<br />

64. Ruemmele FM, Targan SR, Levy G et al. Diagnostic<br />

accuracy of serological assays in pediatric inflammatory<br />

bowel disease. Gastroenterology 1998; 115: 822–829.<br />

65. Dubinsky MC, Ofman JJ, Urman M et al. Clinical utility<br />

of serodiagnostic testing in suspected pediatric inflammatory<br />

bowel disease. Am J Gastroenterol 2001; 96:<br />

758–765.<br />

66. Duerr RH, Targan SR, Landers CJ et al. Anti-neutrophil<br />

cytoplasmic antibodies in ulcerative colitis. Comparison<br />

with other colitides/diarrheal illnesses. Gastroenterology<br />

1991; 100: 1590–1596.<br />

67. Winter HS, Landers CJ, Winkelstein A et al. Antineutrophil<br />

cytoplasmic antibodies in children with<br />

ulcerative colitis. J Pediatr 1994; 125: 707–711.<br />

68. Rutgeerts P, Vermeire S. Serological diagnosis of inflammatory<br />

bowel disease. Lancet 2000; 356: 2117–2118.<br />

69. Markowitz J, Kahn E, Grancher K et al. Atypical<br />

rectosigmoid histology in children with newly diagnosed<br />

ulcerative colitis. Am J Gastroenterol 1993; 88:<br />

2034–2037.<br />

70. Bousvaros A, Glickman JN, Farraye FA et al. Pediatric<br />

patients with untreated ulcerative colitis may present<br />

initially with unusual morphologic findings.<br />

Gastroenterology 2002; 122: A-11.<br />

71. Seidman EG. Role of endoscopy in inflammatory bowel<br />

disease. Gastrointest Endosc Clin North Am 2001; 11:<br />

641–657, vi.<br />

72. Chutkan RK, Scherl E, Waye JD. Colonoscopy in inflammatory<br />

bowel disease. Gastrointest Endosc Clin North<br />

Am 2002; 12: 463–483, viii.<br />

73. Domizio P. Pathology of chronic inflammatory bowel<br />

disease in children. Baillières Clin Gastroenterol 1994; 8:<br />

35–63.<br />

74. Washington K, Greenson JK, Montgomery E et al.<br />

Histopathology of ulcerative colitis in initial rectal<br />

biopsy in children. Am J Surg Pathol 2002; 26:<br />

1441–1449.<br />

75. Horton KM, Jones B, Fishman EK. Imaging of the<br />

inflammatory bowel diseases. In Kirsner JB, ed.<br />

References 411<br />

Inflammatory Bowel Diseases, 5th edn. Philadelphia:<br />

WB Saunders, 2000: 479–500.<br />

76. Tobin JM, Sinha B, Ramani P et al. Upper gastrointestinal<br />

mucosal disease in pediatric Crohn disease and<br />

ulcerative colitis: a blinded, controlled study. J Pediatr<br />

Gastroenterol Nutr 2001; 32: 443–448.<br />

77. Kaufman SS, Vanderhoof JA, Young R et al.<br />

Gastroenteric inflammation in children with ulcerative<br />

colitis. Am J Gastroenterol 1997; 92: 1209–1212.<br />

78. Ruuska T, Vaajalahti P, Arajarvi P et al. Prospective evaluation<br />

of upper gastrointestinal mucosal lesions in children<br />

with ulcerative colitis and Crohn’s disease. J<br />

Pediatr Gastroenterol Nutr 1994; 19: 181–186.<br />

79. Sharif F, McDermott M, Dillon M et al. Focally enhanced<br />

gastritis in children with Crohn’s disease and ulcerative<br />

colitis. Am J Gastroenterol 2002; 97: 1415–1420.<br />

80. Scotiniotis I, Rubesin SE, Ginsberg GG. Imaging modalities<br />

in inflammatory bowel disease. Gastroenterol Clin<br />

North Am 1999; 28: 391–421, ix.<br />

81. Aideyan UO, Smith WL. Inflammatory bowel disease in<br />

children. Radiol Clin North Am 1996; 34: 885–902.<br />

82. Langmead L, Rampton DS. Plain abdominal radiographic<br />

features are not reliable markers of disease<br />

extent in active ulcerative colitis. Am J Gastroenterol<br />

2002; 97: 354–359.<br />

83. Koudahl G, Kristensen M. Toxic megacolon in ulcerative<br />

colitis. Scand J Gastroenterol 1975; 10: 417–421.<br />

84. Chong SK, Blackshaw AJ, Boyle S et al. Histological<br />

diagnosis of chronic inflammatory bowel disease in<br />

childhood. Gut 1985; 26: 55–59.<br />

85. Odze R. Diagnostic problems and advances in inflammatory<br />

bowel disease. Mod Pathol 2003; 16: 347–358.<br />

86. Hildebrand H, Fredrikzon B, Holmquist L et al. Chronic<br />

inflammatory bowel disease in children and adolescents<br />

in Sweden. J Pediatr Gastroenterol Nutr 1991; 13: 293–297.<br />

87. Joossens S, Reinisch W, Vermeire S et al. The value of<br />

serologic markers in indeterminate colitis: a prospective<br />

follow-up study. Gastroenterology 2002; 122: 1242–1247.<br />

88. Hyams JS. Extraintestinal manifestations of inflammatory<br />

bowel disease in children. J Pediatr Gastroenterol<br />

Nutr 1994; 19: 7–21.<br />

89. Greenstein AJ, Janowitz HD, Sachar DB. The extraintestinal<br />

complications of Crohn’s disease and ulcerative<br />

colitis: a study of 700 patients. Medicine (Baltimore)<br />

1976; 55: 401–412.<br />

90. Lindsley CB, Schaller JG. Arthritis associated with<br />

inflammatory bowel disease in children. J Pediatr 1974;<br />

84: 16–20.<br />

91. Mir-Madjlessi SH, Taylor JS, Farmer RG. Clinical course<br />

and evolution of erythema nodosum and pyoderma<br />

gangrenosum in chronic ulcerative colitis: a study of 42<br />

patients. Am J Gastroenterol 1985; 80: 615–620.<br />

92. Wilschanski M, Chait P, Wade JA et al. Primary sclerosing<br />

cholangitis in 32 children: clinical, laboratory, and<br />

radiographic features, with survival analysis.<br />

Hepatology 1995; 22: 1415–1422.<br />

93. Orchard TR, Wordsworth BP, Jewell DP. Peripheral<br />

arthropathies in inflammatory bowel disease: their articular<br />

distribution and natural history. Gut 1998; 42:<br />

387–391.<br />

94. Das KM. Relationship of extraintestinal involvements in<br />

inflammatory bowel disease: new insights into autoimmune<br />

pathogenesis. Dig Dis Sci 1999; 44: 1–13.<br />

95. Passo MH, Fitzgerald JF, Brandt KD. Arthritis associated<br />

with inflammatory bowel disease in children.<br />

Relationship of joint disease to activity and severity of<br />

bowel lesion. Dig Dis Sci 1986; 31: 492–497.<br />

96. Levitt MD, Ritchie JK, Lennard-Jones JE et al. Pyoderma<br />

gangrenosum in inflammatory bowel disease. Br J Surg<br />

1991; 78: 676–678.


412<br />

Ulcerative colitis<br />

97. Callen JP. Pyoderma gangrenosum. Lancet 1998; 351:<br />

581–585.<br />

98. Finkel SI, Janowitz HD. Trauma and the pyoderma<br />

gangrenosum of inflammatory bowel disease. Gut 1981;<br />

22: 410-412.<br />

99. Requena L, Requena C. Erythema nodosum. Dermatol<br />

Online J 2002; 8: 4.<br />

100. Su CG, Judge TA, Lichtenstein GR. Extraintestinal manifestations<br />

of inflammatory bowel disease. Gastroenterol<br />

Clin North Am 2002; 31: 307–327.<br />

101. Rychwalski PJ, Cruz OA, Alanis-Lambreton G et al.<br />

Asymptomatic uveitis in young people with inflammatory<br />

bowel disease. J AAPOS 1997; 1: 111–114.<br />

102. Hofley P, Roarty J, McGinnity G et al. Asymptomatic<br />

uveitis in children with chronic inflammatory bowel<br />

diseases. J Pediatr Gastroenterol Nutr 1993; 17: 397–400.<br />

103. Orchard TR, Chua CN, Ahmad T et al. Uveitis and<br />

erythema nodosum in inflammatory bowel disease: clinical<br />

features and the role of HLA genes.<br />

Gastroenterology 2002; 123: 714–718.<br />

104. Tripathi RC, Kipp MA, Tripathi BJ et al. Ocular toxicity<br />

of prednisone in pediatric patients with inflammatory<br />

bowel disease. Lens Eye Toxic Res 1992; 9: 469–482.<br />

105. Tripathi RC, Kirschner BS, Kipp M et al. Corticosteroid<br />

treatment for inflammatory bowel disease in pediatric<br />

patients increases intraocular pressure. Gastroenterology<br />

1992; 102: 1957–1961.<br />

106. Hyams J, Markowitz J, Treem W. Characterization of<br />

hepatic abnormalities in children with inflammatory<br />

bowel disease. Inflamm Bowel Dis 1995; 1: 27.<br />

107. Roberts EA. Primary sclerosing cholangitis in children. J<br />

Gastroenterol Hepatol 1999; 14: 588–593.<br />

108. Raj V, Lichtenstein DR. Hepatobiliary manifestations of<br />

inflammatory bowel disease. Gastroenterol Clin North<br />

Am 1999; 28: 491–513.<br />

109. Lorusso D, Leo S, Mossa A et al. Cholelithiasis in<br />

inflammatory bowel disease. A case–control study. Dis<br />

Colon Rectum 1990; 33: 791–794.<br />

110. Markowitz RL, Ment LR, Gryboski JD. Cerebral thromboembolic<br />

disease in pediatric and adult inflammatory<br />

bowel disease: case report and review of the literature. J<br />

Pediatr Gastroenterol Nutr 1989; 8: 413–420.<br />

111. Talbot RW, Heppell J, Dozois RR et al. Vascular complications<br />

of inflammatory bowel disease. Mayo Clin Proc<br />

1986; 61: 140–145.<br />

112. Schapira M, Henrion J, Ravoet C et al.<br />

Thromboembolism in inflammatory bowel disease. Acta<br />

Gastroenterol Belg 1999; 62: 182–186.<br />

113. Koutroubakis IE. Unraveling the mechanisms of thrombosis<br />

in inflammatory bowel disease. Am J Gastroenterol<br />

2001; 96: 1325–1327.<br />

114. Higuchi LM, Joffe S, Neufeld EJ et al. Inflammatory<br />

bowel disease associated with immune thrombocytopenic<br />

purpura in children. J Pediatr Gastroenterol Nutr<br />

2001; 33: 582–587.<br />

115. Giannadaki E, Potamianos S, Roussomoustakaki M et al.<br />

Autoimmune hemolytic anemia and positive Coombs<br />

test associated with ulcerative colitis. Am J<br />

Gastroenterol 1997; 92: 1872–1874.<br />

116. Gokhale R, Favus MJ, Karrison T et al. Bone mineral<br />

density assessment in children with inflammatory<br />

bowel disease. Gastroenterology 1998; 114: 902–911.<br />

117. Clark JH, Fitzgerald JF, Bergstein JM. Nephrolithiasis in<br />

childhood inflammatory bowel disease. J Pediatr<br />

Gastroenterol Nutr 1985; 4: 829–834.<br />

118. Keljo DJ, Sugerman KS. Pancreatitis in patients with<br />

inflammatory bowel disease. J Pediatr Gastroenterol Nutr<br />

1997; 25: 108–112.<br />

119. Huang C, Lichtenstein DR. Pancreatic and biliary tract<br />

disorders in inflammatory bowel disease. Gastrointest<br />

Endosc Clin North Am 2002; 12: 535–559.<br />

120. Jewell DP, Caprilli R, Mortensen N et al. Indications and<br />

timing of surgery for severe ulcerative colitis.<br />

Gastroenterol Int 1991; 4: 161–164.<br />

121. Present DH. Toxic megacolon. Med Clin North Am 1993;<br />

77: 1129–1148.<br />

122. Sheth SG, LaMont JT. Toxic megacolon. Lancet 1998;<br />

351: 509–513.<br />

123. Greenstein AJ, Barth JA, Sachar DB et al. Free colonic<br />

perforation without dilatation in ulcerative colitis. Am J<br />

Surg 1986; 152: 272–275.<br />

124. Jalan KN, Sircus W, Card WI et al. An experience of<br />

ulcerative colitis. I. Toxic dilation in 55 cases.<br />

Gastroenterology 1969; 57: 68–82.<br />

125. Fazio VW. Toxic megacolon in ulcerative colitis and<br />

Crohn’s colitis. Clin Gastroenterol 1980; 9: 389–407.<br />

126. Grieco MB, Bordan DL, Geiss AC et al. Toxic megacolon<br />

complicating Crohn’s colitis. Ann Surg 1980; 191: 75–80.<br />

127. Mourelle M, Casellas F, Guarner F et al. Induction of<br />

nitric oxide synthase in colonic smooth muscle from<br />

patients with toxic megacolon. Gastroenterology 1995;<br />

109: 1497–1502.<br />

128. Brown JW. Toxic megacolon associated with loperamide<br />

therapy. JAMA 1979; 241: 501–502.<br />

129. Norland CC, Kirsner JB. Toxic dilatation of colon (toxic<br />

megacolon): etiology, treatment and prognosis in 42<br />

patients. Medicine (Baltimore) 1969; 48: 229–250.<br />

130. Halpert RD. Toxic dilatation of the colon. Radiol Clin<br />

North Am 1987; 25: 147–155.<br />

131. Kramer P, Wittenberg J. Colonic gas distribution in toxic<br />

megacolon. Gastroenterology 1981; 80: 433–437.<br />

132. Present DH, Wolfson D, Gelernt IM et al. Medical<br />

decompression of toxic megacolon by ‘rolling’. A new<br />

technique of decompression with favorable long-term<br />

follow-up. J Clin Gastroenterol 1988; 10: 485–490.<br />

133. Caroline DF, Evers K. Colitis: radiographic features and<br />

differentiation of idiopathic inflammatory bowel<br />

disease. Radiol Clin North Am 1987; 25: 47–66.<br />

134. Gumaste V, Sachar DB, Greenstein AJ. Benign and<br />

malignant colorectal strictures in ulcerative colitis. Gut<br />

1992; 33: 938–41.<br />

135. Truelove SC, Witts LJ. Cortisone in ulcerative colitis.<br />

Final report on a therapeutic trial. BMJ 1955; 2: 1041.<br />

136. Lichtiger S, Present DH, Kornbluth A et al. Cyclosporine<br />

in severe ulcerative colitis refractory to steroid therapy.<br />

N Engl J Med 1994; 330: 1841–1845.<br />

137. Kozarek RA, Patterson DJ, Gelfand MD et al.<br />

Methotrexate induces clinical and histologic remission<br />

in patients with refractory inflammatory bowel disease.<br />

Ann Intern Med 1989; 110: 353–356.<br />

138. Harvey RF, Bradshaw JM. A simple index of Crohn’sdisease<br />

activity. Lancet 1980; 1: 514.<br />

139. Kane SV, Sandborn WJ, Rufo PA et al. Fecal lactoferrin<br />

is a sensitive and specific marker in identifying intestinal<br />

inflammation. Am J Gastroenterol 2003; 98:<br />

1309–1314.<br />

140. Leichtner AM. Aminosalicylates for the treatment of<br />

inflammatory bowel disease. J Pediatr Gastroenterol Nutr<br />

1995; 21: 245–252.<br />

141. Wahl C, Liptay S, Adler G et al. Sulfasalazine: a potent<br />

and specific inhibitor of nuclear factor kappa B. J Clin<br />

Invest 1998; 101: 1163–1174.<br />

142. Bantel H, Berg C, Vieth M et al. Mesalazine inhibits<br />

activation of transcription factor NF-kappaB in inflamed<br />

mucosa of patients with ulcerative colitis. Am J<br />

Gastroenterol 2000; 95: 3452–3457.


143. Dick AP, Grayson MJ, Carpenter RG et al. Controlled<br />

trial of sulfasalazine in the treatment of ulcerative<br />

colitis. Gut 1964; 5: 437–442.<br />

144. Sninsky CA, Cort DH, Shanahan F et al. Oral<br />

mesalamine (Asacol) for mildly to moderately active<br />

ulcerative colitis. A multicenter study. Ann Intern Med<br />

1991; 115: 350–355.<br />

145. Sutherland LR, May GR, Shaffer EA. Sulfasalazine<br />

revisited: a meta-analysis of 5-aminosalicylic acid in the<br />

treatment of ulcerative colitis. Ann Intern Med 1993;<br />

118: 540–549.<br />

146. Gisbert JP, Gomollon F, Mate J et al. Role of 5-aminosalicylic<br />

acid (5-ASA) in treatment of inflammatory bowel<br />

disease: a systematic review. Dig Dis Sci 2002; 47:<br />

471–488.<br />

147. Sutherland LR, Roth DE, Beck PL, Alternatives to<br />

sulfasalazine: a meta-analysis of 5-ASA in the treatment<br />

of ulcerative colitis. Inflamm Bowel Dis 1997; 3: 65–78.<br />

148. Ferry GD, Kirschner BS, Grand RJ et al. Olsalazine<br />

versus sulfasalazine in mild to moderate childhood<br />

ulcerative colitis: results of the Pediatric<br />

Gastroenterology Collaborative Research Group Clinical<br />

Trial. J Pediatr Gastroenterol Nutr 1993; 17: 32–38.<br />

149. Barden L, Lipson A, Pert P. Mesalazine in childhood<br />

inflammatory bowel disease. Aliment Pharmacol Ther<br />

1989; 3: 597.<br />

150. Levine DS, Riff DS, Pruitt R et al. A randomized, double<br />

blind, dose–response comparison of balsalazide (6.75 g),<br />

balsalazide (2.25 g), and mesalamine (2.4 g) in the treatment<br />

of active, mild-to-moderate ulcerative colitis. Am J<br />

Gastroenterol 2002; 97: 1398–1407.<br />

151. Green JR, Lobo AJ, Holdsworth CD et al. Balsalazide is<br />

more effective and better tolerated than mesalamine in<br />

the treatment of acute ulcerative colitis. The Abacus<br />

Investigator Group. Gastroenterology 1998; 114: 15–22.<br />

152. Hanauer SB. Inflammatory bowel disease. N Engl J Med<br />

1996; 334: 841–848.<br />

153. Boyer DL, Li BU, Fyda JN et al. Sulfasalazine-induced<br />

hepatotoxicity in children with inflammatory bowel<br />

disease. J Pediatr Gastroenterol Nutr 1989; 8: 528–532.<br />

154. Garau P, Orenstein SR, Neigut DA et al. Pancreatitis<br />

associated with olsalazine and sulfasalazine in children<br />

with ulcerative colitis. J Pediatr Gastroenterol Nutr 1994;<br />

18: 481–485.<br />

155. Paerregaard A, Krasilnikoff PA. Pancreatitis in a child<br />

after rectal administration of 5-aminosalicylic acid.<br />

Inflamm Bowel Dis 1997; 3: 20.<br />

156. Sturgeon JB, Bhatia P, Hermens D et al. Exacerbation of<br />

chronic ulcerative colitis with mesalamine.<br />

Gastroenterology 1995; 108: 1889–1893.<br />

157. Kirschner BS. The medical management of inflammatory<br />

bowel disease in children. In Kirsner JB, ed.<br />

Inflammatory Bowel Disease. Philadelphia: Saunders,<br />

2000: 578–597.<br />

158. Beattie RM, Nicholls SW, Domizio P et al. Endoscopic<br />

assessment of the colonic response to corticosteroids in<br />

children with ulcerative colitis. J Pediatr Gastroenterol<br />

Nutr 1996; 22: 373–379.<br />

159. Truhan AP, Ahmed AR. Corticosteroids: a review with<br />

emphasis on complications of prolonged systemic<br />

therapy. Ann Allergy 1989; 62: 375–391.<br />

160. Greenberg GR, Feagan BG, Martin F et al. Oral budesonide<br />

for active Crohn’s disease. Canadian<br />

Inflammatory Bowel Disease Study Group. N Engl J Med<br />

1994; 331: 836–841.<br />

161. Hanauer SB, Robinson M, Pruitt R et al. Budesonide<br />

enema for the treatment of active, distal ulcerative<br />

colitis and proctitis: a dose-ranging study. US<br />

References 413<br />

Budesonide enema study group. Gastroenterology 1998;<br />

115: 525–532.<br />

162. Hanauer SB, New steroids for IBD: progress report. Gut<br />

2002; 51: 182–183.<br />

163. Bousvaros A. Immunosuppression. In Walker WA, Durie<br />

PR, Hamilton JR, Walker-Smith JA, Watkins JB, eds.<br />

Pediatric Gastrointestinal Disease. Hamilton, Ontario,<br />

Canada: BC Decker, 2000: 1769–1794.<br />

164. Peppercorn MA. Are antibiotics useful in the management<br />

of nontoxic severe ulcerative colitis? J Clin<br />

Gastroenterol 1993; 17: 14–17.<br />

165. Chapman RW, Selby WS, Jewell DP. Controlled trial of<br />

intravenous metronidazole as an adjunct to corticosteroids<br />

in severe ulcerative colitis. Gut 1986; 27:<br />

1210–1212.<br />

166. Mantzaris GJ, Hatzis A, Kontogiannis P et al.<br />

Intravenous tobramycin and metronidazole as an<br />

adjunct to corticosteroids in acute, severe ulcerative<br />

colitis. Am J Gastroenterol 1994; 89: 43–46.<br />

167. Mantzaris GJ, Petraki K, Archavlis E et al. A prospective<br />

randomized controlled trial of intravenous ciprofloxacin<br />

as an adjunct to corticosteroids in acute, severe ulcerative<br />

colitis. Scand J Gastroenterol 2001; 36: 971–974.<br />

168. Treem WR, Davis PM, Hyams JS. Cyclosporine treatment<br />

of severe ulcerative colitis in children. J Pediatr<br />

1991; 119: 994–997.<br />

169. Benkov KJ, Rosh JR, Schwersenz AH et al. Cyclosporine<br />

as an alternative to surgery in children with inflammatory<br />

bowel disease. J Pediatr Gastroenterol Nutr 1994;<br />

19: 290–294.<br />

170. Treem WR, Cohen J, Davis PM et al. Cyclosporine for<br />

the treatment of fulminant ulcerative colitis in children.<br />

Immediate response, long-term results, and impact on<br />

surgery. Dis Colon Rectum 1995; 38: 474–479.<br />

171. Escher JC, Taminiau JA, Nieuwenhuis EE et al. Treatment<br />

of inflammatory bowel disease in childhood: best available<br />

evidence. Inflamm Bowel Dis 2003; 9: 34–58.<br />

172. Bousvaros A, Kirschner BS, Werlin SL et al. Oral<br />

tacrolimus treatment of severe colitis in children. J<br />

Pediatr 2000; 137: 794–799.<br />

173. Ramakrishna J, Langhans N, Calenda K et al. Combined<br />

use of cyclosporine and azathioprine or 6-mercaptopurine<br />

in pediatric inflammatory bowel disease. J Pediatr<br />

Gastroenterol Nutr 1996; 22: 296–302.<br />

174. Marshall JK, Irvine EJ. Rectal aminosalicylate therapy<br />

for distal ulcerative colitis: a meta-analysis. Aliment<br />

Pharmacol Ther 1995; 9: 293–300.<br />

175. Marshall JK, Irvine EJ. Rectal corticosteroids versus<br />

alternative treatments in ulcerative colitis: a meta-analysis.<br />

Gut 1997; 40: 775–781.<br />

176. Mulder CJ, Tytgat GN. Review article: topical corticosteroids<br />

in inflammatory bowel disease. Aliment<br />

Pharmacol Ther 1993; 7: 125–130.<br />

177. Gionchetti P, Rizzello F, Venturi A et al. Comparison of<br />

oral with rectal mesalazine in the treatment of ulcerative<br />

proctitis. Dis Colon Rectum 1998; 41: 93–97.<br />

178. Cohen RD, Woseth DM, Thisted RA et al. A meta-analysis<br />

and overview of the literature on treatment options<br />

for left-sided ulcerative colitis and ulcerative proctitis.<br />

Am J Gastroenterol 2000; 95: 1263–1276.<br />

179. Allgayer H. Sulfasalazine and 5-ASA compounds.<br />

Gastroenterol Clin North Am 1992; 21: 643–658.<br />

180. Sutherland LR. Topical treatment of ulcerative colitis.<br />

Med Clin North Am 1990; 74: 119–131.<br />

181. Safdi M, DeMicco M, Sninsky C et al. A double-blind<br />

comparison of oral versus rectal mesalamine versus<br />

combination therapy in the treatment of distal ulcerative<br />

colitis. Am J Gastroenterol 1997; 92: 1867–1871.


414<br />

Ulcerative colitis<br />

182. Danielsson A, Lofberg R, Persson T et al. A steroid<br />

enema, budesonide, lacking systemic effects for the<br />

treatment of distal ulcerative colitis or proctitis. Scand J<br />

Gastroenterol 1992; 27: 9–12.<br />

183. Lindgren S, Lofberg R, Bergholm L et al. Effect of budesonide<br />

enema on remission and relapse rate in distal<br />

ulcerative colitis and proctitis. Scand J Gastroenterol<br />

2002; 37: 705–710.<br />

184. Winter TA, Dalton HR, Merrett MN et al. Cyclosporin A<br />

retention enemas in refractory distal ulcerative colitis<br />

and ‘pouchitis’. Scand J Gastroenterol 1993; 28:<br />

701–704.<br />

185. Sandborn WJ, Tremaine WJ, Schroeder KW et al. A<br />

placebo-controlled trial of cyclosporine enemas for<br />

mildly to moderately active left-sided ulcerative colitis.<br />

Gastroenterology 1994; 106: 1429–1435.<br />

186. D’Agata ID, Vanounou T, Seidman E. Mesalamine in<br />

pediatric inflammatory bowel disease: A 10-year experience.<br />

Inflamm Bowel Dis 1996; 2: 229–235.<br />

187. Kruis W, Schreiber S, Theuer D et al. Low dose<br />

balsalazide (1.5g twice daily) and mesalazine (0.5g<br />

three times daily) maintained remission of ulcerative<br />

colitis but high dose balsalazide (3.0g twice daily) was<br />

superior in preventing relapses. Gut 2001; 49: 783–789.<br />

188. d’Albasio G, Paoluzi P, Campieri M et al. Maintenance<br />

treatment of ulcerative proctitis with mesalazine<br />

suppositories: a double-blind placebo-controlled trial.<br />

The Italian IBD Study Group. Am J Gastroenterol 1998;<br />

93: 799–803.<br />

189. d’Albasio G, Pacini F, Camarri E et al. Combined therapy<br />

with 5-aminosalicylic acid tablets and enemas for maintaining<br />

remission in ulcerative colitis: a randomized<br />

double-blind study. Am J Gastroenterol 1997; 92:<br />

1143–1147.<br />

190. Eaden J, Abrams K, Ekbom A et al. Colorectal cancer<br />

prevention in ulcerative colitis: a case–control study.<br />

Aliment Pharmacol Ther 2000; 14: 145–153.<br />

191. Pinczowski D, Ekbom A, Baron J et al. Risk factors for<br />

colorectal cancer in patients with ulcerative colitis: a<br />

case–control study. Gastroenterology 1994; 107:<br />

117–120.<br />

192. Ekbom A, Kornfield D. Sulfasalazine use as a preventative<br />

factor in colorectal cancer in ulcerative colitis<br />

patients: a review. Inflamm Bowel Dis 1996; 2: 276.<br />

193. Ryan BM, Russel MG, Langholz E et al.<br />

Aminosalicylates and colorectal cancer in IBD: a not-so<br />

bitter pill to swallow. Am J Gastroenterol 2003; 98:<br />

1682–1687.<br />

194. Verhave M, Winter HS, Grand RJ. Azathioprine in the<br />

treatment of children with inflammatory bowel disease.<br />

J Pediatr 1990; 117: 809–814.<br />

195. Kader HA, Mascarenhas MR, Piccoli DA et al.<br />

Experiences with 6-mercaptopurine and azathioprine<br />

therapy in pediatric patients with severe ulcerative<br />

colitis. J Pediatr Gastroenterol Nutr 1999; 28: 54–58.<br />

196. Kirschner BS. Safety of azathioprine and 6-mercaptopurine<br />

in pediatric patients with inflammatory bowel<br />

disease. Gastroenterology 1998; 115: 813–821.<br />

197. Present DH, Meltzer SJ, Krumholz MP et al. 6-<br />

Mercaptopurine in the management of inflammatory<br />

bowel disease: short- and long-term toxicity. Ann Intern<br />

Med 1989; 111: 641–649.<br />

198. Cuffari C, Theoret Y, Latour S et al. 6-Mercaptopurine<br />

metabolism in Crohn’s disease: correlation with efficacy<br />

and toxicity. Gut 1996; 39: 401–406.<br />

199. Dubinsky MC, Lamothe S, Yang HY et al.<br />

Pharmacogenomics and metabolite measurement for 6mercaptopurine<br />

therapy in inflammatory bowel disease.<br />

Gastroenterology 2000; 118: 705–713.<br />

200. Lennard L, Van Loon JA, Weinshilboum RM.<br />

Pharmacogenetics of acute azathioprine toxicity: relationship<br />

to thiopurine methyltransferase genetic polymorphism.<br />

Clin Pharmacol Ther 1989; 46: 149–154.<br />

201. George J, Present DH, Pou R et al. The long-term<br />

outcome of ulcerative colitis treated with 6-mercaptopurine.<br />

Am J Gastroenterol 1996; 91: 1711–1714.<br />

202. Fraser AG, Orchard TR, Robinson EM et al. Long-term<br />

risk of malignancy after treatment of inflammatory<br />

bowel disease with azathioprine. Aliment Pharmacol<br />

Ther 2002; 16: 1225–1232.<br />

203. Korelitz BI, Mirsky FJ, Fleisher MR et al. Malignant<br />

neoplasms subsequent to treatment of inflammatory<br />

bowel disease with 6-mercaptopurine. Am J<br />

Gastroenterol 1999; 94: 3248–3253.<br />

204. Connell WR, Kamm MA, Dickson M et al. Long-term<br />

neoplasia risk after azathioprine treatment in inflammatory<br />

bowel disease. Lancet 1994; 343: 1249–1252.<br />

205. Dayharsh GA, Loftus EV Jr, Sandborn WJ et al.<br />

Epstein–Barr virus-positive lymphoma in patients with<br />

inflammatory bowel disease treated with azathioprine<br />

or 6-mercaptopurine. Gastroenterology 2002; 122: 72–77.<br />

206. Feagan BG, Rochon J, Fedorak RN et al. Methotrexate for<br />

the treatment of Crohn’s disease. The North American<br />

Crohn’s Study Group Investigators. N Engl J Med 1995;<br />

332: 292–297.<br />

207. Schroder O, Stein J, Low dose methotrexate in inflammatory<br />

bowel disease: current status and future directions.<br />

Am J Gastroenterol 2003; 98: 530–537.<br />

208. Paoluzi OA, Pica R, Marcheggiano A et al. Azathioprine<br />

or methotrexate in the treatment of patients with<br />

steroid-dependent or steroid-resistant ulcerative colitis:<br />

results of an open-label study on efficacy and tolerability<br />

in inducing and maintaining remission. Aliment<br />

Pharmacol Ther 2002; 16: 1751–1759.<br />

209. Oren R, Arber N, Odes S et al. Methotrexate in chronic<br />

active ulcerative colitis: a double-blind, randomized,<br />

Israeli multicenter trial. Gastroenterology 1996; 110:<br />

1416–1421.<br />

210. Egan LJ, Sandborn WJ, Tremaine WJ et al. A randomized<br />

dose–response and pharmacokinetic study of<br />

methotrexate for refractory inflammatory Crohn’s<br />

disease and ulcerative colitis. Aliment Pharmacol Ther<br />

1999; 13: 1597–1604.<br />

211. Mack DR, Young R, Kaufman SS et al. Methotrexate in<br />

patients with Crohn’s disease after 6-mercaptopurine. J<br />

Pediatr 1998; 132: 830–835.<br />

212. Burke DA, Axon AT, Clayden SA et al. The efficacy of<br />

tobramycin in the treatment of ulcerative colitis.<br />

Aliment Pharmacol Ther 1990; 4: 123–129.<br />

213. Lobo AJ, Burke DA, Sobala GM et al. Oral tobramycin in<br />

ulcerative colitis: effect on maintenance of remission.<br />

Aliment Pharmacol Ther 1993; 7: 155–158.<br />

214. Gilat T, Suissa A, Leichtman G et al. A comparative<br />

study of metronidazole and sulfasalazine in active, not<br />

severe, ulcerative colitis. An Israeli multicenter trial. J<br />

Clin Gastroenterol 1987; 9: 415–417.<br />

215. Dickinson RJ, O’Connor HJ, Pinder I et al. Double blind<br />

controlled trial of oral vancomycin as adjunctive treatment<br />

in acute exacerbations of idiopathic colitis. Gut<br />

1985; 26: 1380–1384.<br />

216. Mantzaris GJ, Archavlis E, Christoforidis P et al. A<br />

prospective randomized controlled trial of oral<br />

ciprofloxacin in acute ulcerative colitis. Am J<br />

Gastroenterol 1997; 92: 454–456.<br />

217. Turunen UM, Farkkila MA, Hakala K et al. Long-term<br />

treatment of ulcerative colitis with ciprofloxacin: a<br />

prospective, double-blind, placebo-controlled study.<br />

Gastroenterology 1998; 115: 1072–1078.


218. Chey WY, Hussain A, Ryan C et al. Infliximab for refractory<br />

ulcerative colitis. Am J Gastroenterol 2001; 96:<br />

2373–2381.<br />

219. Su C, Salzberg BA, Lewis JD et al. Efficacy of anti-tumor<br />

necrosis factor therapy in patients with ulcerative<br />

colitis. Am J Gastroenterol 2002; 97: 2577–2584.<br />

220. Kohn A, Prantera C, Pera A et al. Anti-tumour necrosis<br />

factor alpha (infliximab) in the treatment of severe<br />

ulcerative colitis: result of an open study on 13 patients.<br />

Dig Liver Dis 2002; 34: 626–630.<br />

221. Serrano MS, Schmidt-Sommerfeld E, Kilbaugh TJ et al.<br />

Use of infliximab in pediatric patients with inflammatory<br />

bowel disease. Ann Pharmacother 2001; 35:<br />

823–828.<br />

222. Mamula P, Markowitz JE, Brown KA et al. Infliximab as<br />

a novel therapy for pediatric ulcerative colitis. J Pediatr<br />

Gastroenterol Nutr 2002; 34: 307–311.<br />

223. Actis GC, Bruno M, Pinna-Pintor M et al. Infliximab for<br />

treatment of steroid-refractory ulcerative colitis. Dig<br />

Liver Dis 2002; 34: 631–634.<br />

224. Sands BE, Tremaine WJ, Sandborn WJ et al. Infliximab<br />

in the treatment of severe, steroid-refractory ulcerative<br />

colitis: a pilot study. Inflamm Bowel Dis 2001; 7: 83–88.<br />

225. Probert CS, Hearing SD, Schreiber S et al. Infliximab in<br />

moderately severe glucocorticoid resistant ulcerative<br />

colitis: a randomised controlled trial. Gut 2003; 52:<br />

998–1002.<br />

226. Keane J, Gershon S, Wise RP et al. Tuberculosis associated<br />

with infliximab, a tumor necrosis factor alphaneutralizing<br />

agent. N Engl J Med 2001; 345: 1098–1104.<br />

227. Farrell RJ, Alsahli M, Jeen YT et al. Intravenous hydrocortisone<br />

premedication reduces antibodies to infliximab<br />

in Crohn’s disease: a randomized controlled trial.<br />

Gastroenterology 2003; 124: 917–924.<br />

228. Baert F, Noman M, Vermeire S et al. Influence of<br />

immunogenicity on the long-term efficacy of infliximab<br />

in Crohn’s disease. N Engl J Med 2003; 348: 601–608.<br />

229. Crandall WV, Mackner LM. Infusion reactions to infliximab<br />

in children and adolescents: frequency, outcome<br />

and a predictive model. Aliment Pharmacol Ther 2003;<br />

17: 75–84.<br />

230. Sandborn WJ, Hanauer SB. Infliximab in the treatment<br />

of Crohn’s disease: a user’s guide for clinicians. Am J<br />

Gastroenterol 2002; 97: 2962–2972.<br />

231. Orth T, Peters M, Schlaak JF et al. Mycophenolate<br />

mofetil versus azathioprine in patients with chronic<br />

active ulcerative colitis: a 12-month pilot study. Am J<br />

Gastroenterol 2000; 95: 1201–1207.<br />

232. Fellermann K, Steffen M, Stein J et al. Mycophenolate<br />

mofetil: lack of efficacy in chronic active inflammatory<br />

bowel disease. Aliment Pharmacol Ther 2000; 14: 171–176.<br />

233. Skelly MM, Logan RF, Jenkins D et al. Toxicity of<br />

mycophenolate mofetil in patients with inflammatory<br />

bowel disease. Inflamm Bowel Dis 2002; 8: 93–97.<br />

234. Venturi A, Gionchetti P, Rizzello F et al. Impact on the<br />

composition of the faecal flora by a new probiotic<br />

preparation: preliminary data on maintenance treatment<br />

of patients with ulcerative colitis. Aliment Pharmacol<br />

Ther 1999; 13: 1103–1108.<br />

235. Kruis W, Schutz E, Fric P et al. Double-blind comparison<br />

of an oral Escherichia coli preparation and<br />

mesalazine in maintaining remission of ulcerative<br />

colitis. Aliment Pharmacol Ther 1997; 11: 853–858.<br />

236. Rembacken BJ, Snelling AM, Hawkey PM et al. Nonpathogenic<br />

Escherichia coli versus mesalazine for the<br />

treatment of ulcerative colitis: a randomised trial.<br />

Lancet 1999; 354: 635–639.<br />

237. Pullan RD, Rhodes J, Ganesh S et al. Transdermal nicotine<br />

for active ulcerative colitis. N Engl J Med 1994; 330:<br />

811–815.<br />

References 415<br />

238. Sandborn WJ, Tremaine WJ, Offord KP et al.<br />

Transdermal nicotine for mildly to moderately active<br />

ulcerative colitis. A randomized, double-blind, placebocontrolled<br />

trial. Ann Intern Med 1997; 126: 364–371.<br />

239. Thomas GA, Rhodes J, Mani V et al. Transdermal nicotine<br />

as maintenance therapy for ulcerative colitis. N<br />

Engl J Med 1995; 332: 988–992.<br />

240. Green JT, Thomas GA, Rhodes J et al. Nicotine enemas<br />

for active ulcerative colitis – a pilot study. Aliment<br />

Pharmacol Ther 1997; 11: 859–863.<br />

241. Stenson WF, Cort D, Rodgers J et al. Dietary supplementation<br />

with fish oil in ulcerative colitis. Ann Intern Med<br />

1992; 116: 609–614.<br />

242. Aslan A, Triadafilopoulos G. Fish oil fatty acid supplementation<br />

in active ulcerative colitis: a double-blind,<br />

placebo-controlled, crossover study. Am J Gastroenterol<br />

1992; 87: 432–437.<br />

243. Hawthorne AB, Daneshmend TK, Hawkey CJ et al.<br />

Treatment of ulcerative colitis with fish oil supplementation:<br />

a prospective 12 month randomised controlled<br />

trial. Gut 1992; 33: 922–928.<br />

244. Greenfield SM, Green AT, Teare JP et al. A randomized<br />

controlled study of evening primrose oil and fish oil in<br />

ulcerative colitis. Aliment Pharmacol Ther 1993; 7:<br />

159–166.<br />

245. Scheppach W. Treatment of distal ulcerative colitis with<br />

short-chain fatty acid enemas. A placebo-controlled<br />

trial. German–Austrian SCFA Study Group. Dig Dis Sci<br />

1996; 41: 2254–2259.<br />

246. Breuer RI, Soergel KH, Lashner BA et al. Short chain<br />

fatty acid rectal irrigation for left-sided ulcerative<br />

colitis: a randomised, placebo controlled trial. Gut 1997;<br />

40: 485–491.<br />

247. Vernia P, Marcheggiano A, Caprilli R et al. Short-chain<br />

fatty acid topical treatment in distal ulcerative colitis.<br />

Aliment Pharmacol Ther 1995; 9: 309–313.<br />

248. Evans RC, Wong VS, Morris AI et al. Treatment of corticosteroid-resistant<br />

ulcerative colitis with heparin – a<br />

report of 16 cases. Aliment Pharmacol Ther 1997; 11:<br />

1037–1040.<br />

249. Torkvist L, Thorlacius H, Sjoqvist U et al. Low molecular<br />

weight heparin as adjuvant therapy in active ulcerative<br />

colitis. Aliment Pharmacol Ther 1999; 13:<br />

1323–1328.<br />

250. Folwaczny C, Wiebecke B, Loeschke K. Unfractioned<br />

heparin in the therapy of patients with highly active<br />

inflammatory bowel disease. Am J Gastroenterol 1999;<br />

94: 1551–1555.<br />

251. Panes J, Esteve M, Cabre E et al. Comparison of heparin<br />

and steroids in the treatment of moderate and severe<br />

ulcerative colitis. Gastroenterology 2000; 119: 903–908.<br />

252. Van Assche G, Dalle I, Noman M et al. A pilot study on<br />

the use of the humanized anti-interleukin-2 receptor<br />

antibody daclizumab in active ulcerative colitis. Am J<br />

Gastroenterol 2003; 98: 369–376.<br />

253. Motil KJ, Grand RJ. Nutritional management of inflammatory<br />

bowel disease. Pediatr Clin North Am 1985; 32:<br />

447–469.<br />

254. Burke A, Lichtenstein GR, Rombeau JL. Nutrition and<br />

ulcerative colitis. Baillières Clin Gastroenterol 1997; 11:<br />

153–174.<br />

255. Kleinman RE, Balistreri WF, Heyman MB et al.<br />

Nutritional support for pediatric patients with inflammatory<br />

bowel disease. J Pediatr Gastroenterol Nutr 1989;<br />

8: 8–12.<br />

256. Dickinson RJ, Ashton MG, Axon AT et al. Controlled<br />

trial of intravenous hyperalimentation and total bowel<br />

rest as an adjunct to the routine therapy of acute colitis.<br />

Gastroenterology 1980; 79: 1199–1204.


416<br />

Ulcerative colitis<br />

257. McIntyre PB, Powell-Tuck J, Wood SR et al. Controlled<br />

trial of bowel rest in the treatment of severe acute<br />

colitis. Gut 1986; 27: 481–485.<br />

258. Gonzalez-Huix F, Fernandez-Banares F, Esteve-Comas M<br />

et al. Enteral versus parenteral nutrition as adjunct<br />

therapy in acute ulcerative colitis. Am J Gastroenterol<br />

1993; 88: 227–232.<br />

259. Durno C, Sherman P, Harris K et al. Outcome after<br />

ileoanal anastomosis in pediatric patients with ulcerative<br />

colitis. J Pediatr Gastroenterol Nutr 1998; 27:<br />

501–507.<br />

260. Perrault J, Berry R, Greseth J et al. Pouchitis in young<br />

patients after the ileal-pouch anal anastomosis. Inflamm<br />

Bowel Dis 1997; 3: 181–185.<br />

261. Dozois RR, Kelly KA, The surgical management of ulcerative<br />

colitis. In Kirsner JB, ed. Inflammatory Bowel<br />

Disease. Philadelphia: WB Saunders, 2000: 626–657.<br />

262. Nicholls S, Vieira MC, Majrowski WH et al. Linear<br />

growth after colectomy for ulcerative colitis in childhood.<br />

J Pediatr Gastroenterol Nutr 1995; 21: 82–86.<br />

263. Michener WM, Farmer RG, Mortimer EA. Long-term<br />

prognosis of ulcerative colitis with onset in childhood<br />

or adolescence. J Clin Gastroenterol 1979; 1: 301–305.<br />

264. Hyams JS, Davis P, Grancher K et al. Clinical outcome of<br />

ulcerative colitis in children. J Pediatr 1996; 129: 81–88.<br />

265. Falcone RA Jr, Lewis LG, Warner BW. Predicting the<br />

need for colectomy in pediatric patients with ulcerative<br />

colitis. J Gastrointest Surg 2000; 4: 201–206.<br />

266. Kettlewell MGW. Recent advances in surgical therapy. In<br />

Tytgat GNJ, Bartelsman JFWM, van Deventer SJH, eds.<br />

Inflammatory Bowel Diseases. Boston: Kluwer Academic<br />

Publishers, 1995: 508–516.<br />

267. Farouk R, Pemberton JH. Surgical options in ulcerative<br />

colitis. Surg Clin North Am 1997; 77: 85–94.<br />

268. Mowschenson PM, Critchlow JF. Outcome of early<br />

surgical complications following ileoanal pouch operation<br />

without diverting ileostomy. Am J Surg 1995; 169:<br />

143–145; discussion 145–146.<br />

269. Cohen Z, McLeod RS, Stephen W et al. Continuing<br />

evolution of the pelvic pouch procedure. Ann Surg<br />

1992; 216: 506–511; discussion 511–512.<br />

270. Fonkalsrud EW, Thakur A, Beanes S. Ileoanal pouch<br />

procedures in children. J Pediatr Surg 2001; 36:<br />

1689–1692.<br />

271. Telander RL, Spencer M, Perrault J et al. Long-term<br />

follow-up of the ileoanal anastomosis in children and<br />

young adults. Surgery 1990; 108: 717–723; discussion<br />

723–725.<br />

272. Rintala RJ, Lindahl HG. Proctocolectomy and J-pouch<br />

ileo-anal anastomosis in children. J Pediatr Surg 2002;<br />

37: 66–70.<br />

273. Mahadevan U, Sandborn WJ. Diagnosis and management<br />

of pouchitis. Gastroenterology 2003; 124:<br />

1636–1650.<br />

274. Sandborn WJ. Pouchitis following ileal pouch–anal<br />

anastomosis: definition, pathogenesis, and treatment.<br />

Gastroenterology 1994; 107: 1856–1860.<br />

275. Kartheuser AH, Parc R, Penna CP et al. Ileal pouch–anal<br />

anastomosis as the first choice operation in patients<br />

with familial adenomatous polyposis: a ten-year experience.<br />

Surgery 1996; 119: 615–623.<br />

276. Faubion WA Jr, Loftus EV, Sandborn WJ et al. Pediatric<br />

‘PSC-IBD’: a descriptive report of associated inflammatory<br />

bowel disease among pediatric patients with PSC. J<br />

Pediatr Gastroenterol Nutr 2001; 33: 296–300.<br />

277. Penna C, Dozois R, Tremaine W et al. Pouchitis after<br />

ileal pouch-anal anastomosis for ulcerative colitis<br />

occurs with increased frequency in patients with associ-<br />

ated primary sclerosing cholangitis. Gut 1996; 38:<br />

234–239.<br />

278. Gionchetti P, Amadini C, Rizzello F et al. Treatment of<br />

mild to moderate ulcerative colitis and pouchitis.<br />

Aliment Pharmacol Ther 2002; 16 (Suppl 4): 13–19.<br />

279. Shen B, Achkar JP, Lashner BA et al. A randomized clinical<br />

trial of ciprofloxacin and metronidazole to treat<br />

acute pouchitis. Inflamm Bowel Dis 2001; 7: 301–305.<br />

280. Miglioli M, Barbara L, Di Febo G et al. Topical administration<br />

of 5-aminosalicylic acid: a therapeutic proposal<br />

for the treatment of pouchitis. N Engl J Med 1989; 320:<br />

257.<br />

281. Sambuelli A, Boerr L, Negreira S et al. Budesonide<br />

enema in pouchitis – a double-blind, double-dummy,<br />

controlled trial. Aliment Pharmacol Ther 2002; 16:<br />

27–34.<br />

282. de Silva HJ, Ireland A, Kettlewell M et al. Short-chain<br />

fatty acid irrigation in severe pouchitis. N Engl J Med<br />

1989; 321: 1416–1417.<br />

283. Wischmeyer P, Pemberton JH, Phillips SF. Chronic<br />

pouchitis after ileal pouch–anal anastomosis: responses<br />

to butyrate and glutamine suppositories in a pilot study.<br />

Mayo Clin Proc 1993; 68: 978–981.<br />

284. Gionchetti P, Rizzello F, Helwig U et al. Prophylaxis of<br />

pouchitis onset with probiotic therapy: a double-blind,<br />

placebo-controlled trial. Gastroenterology 2003; 124:<br />

1202–1209.<br />

285. Gionchetti P, Rizzello F, Venturi A et al. Oral bacteriotherapy<br />

as maintenance treatment in patients with<br />

chronic pouchitis: a double-blind, placebo-controlled<br />

trial. Gastroenterology 2000; 119: 305–309.<br />

286. Sarigol S, Wyllie R, Gramlich T et al. Incidence of<br />

dysplasia in pelvic pouches in pediatric patients after<br />

ileal pouch–anal anastomosis for ulcerative colitis. J<br />

Pediatr Gastroenterol Nutr 1999; 28: 429–434.<br />

287. Thompson-Fawcett MW, Marcus V, Redston M et al.<br />

Risk of dysplasia in long-term ileal pouches and<br />

pouches with chronic pouchitis. Gastroenterology 2001;<br />

121: 275–281.<br />

288. Veress B, Reinholt FP, Lindquist K et al. Long-term<br />

histomorphological surveillance of the pelvic ileal<br />

pouch: dysplasia develops in a subgroup of patients.<br />

Gastroenterology 1995; 109: 1090–1097.<br />

289. Heuschen UA, Heuschen G, Autschbach F et al.<br />

Adenocarcinoma in the ileal pouch: late risk of cancer<br />

after restorative proctocolectomy. Int J Colorectal Dis<br />

2001; 16: 126–130.<br />

290. Burke PM, Neigut D, Kocoshis S et al. Correlates of<br />

depression in new onset pediatric inflammatory bowel<br />

disease. Child Psychiatry Hum Dev 1994; 24: 275–283.<br />

291. Szajnberg N, Krall V, Davis P et al. Psychopathology and<br />

relationship measures in children with inflammatory<br />

bowel disease and their parents. Child Psychiatry Hum<br />

Dev 1993; 23: 215–232.<br />

292. Engstrom I. Mental health and psychological functioning<br />

in children and adolescents with inflammatory<br />

bowel disease: a comparison with children having other<br />

chronic illnesses and with healthy children. J Child<br />

Psychol Psychiatry 1992; 33: 563–582.<br />

293. Engstrom I, Lindquist BL. Inflammatory bowel disease<br />

in children and adolescents: a somatic and psychiatric<br />

investigation. Acta Paediatr Scand 1991; 80: 640–647.<br />

294. Loonen HJ, Grootenhuis MA, Last BF et al. Quality of<br />

life in paediatric inflammatory bowel disease measured<br />

by a generic and a disease-specific questionnaire. Acta<br />

Paediatr 2002; 91: 348–354.<br />

295. Otley A, Smith C, Nicholas D et al. The IMPACT questionnaire:<br />

a valid measure of health-related quality of


life in pediatric inflammatory bowel disease. J Pediatr<br />

Gastroenterol Nutr 2002; 35: 557–563.<br />

296. Loonen HJ, Grootenhuis MA, Last BF et al. Measuring<br />

quality of life in children with inflammatory bowel<br />

disease: the impact-II (NL). Qual Life Res 2002; 11:<br />

47–56.<br />

297. Langholz E, Munkholm P, Davidsen M et al. Changes in<br />

extent of ulcerative colitis: a study on the course and<br />

prognostic factors. Scand J Gastroenterol 1996; 31:<br />

260–266.<br />

298. Meucci G, Vecchi M, Astegiano M et al. The natural<br />

history of ulcerative proctitis: a multicenter, retrospective<br />

study. Gruppo di Studio per le Malattie<br />

Infiammatorie Intestinali (GSMII). Am J Gastroenterol<br />

2000; 95: 469–473.<br />

299. Hyams J, Davis P, Lerer T et al. Clinical outcome of<br />

ulcerative proctitis in children. J Pediatr Gastroenterol<br />

Nutr 1997; 25: 149–152.<br />

300. Mir-Madjlessi SH, Michener WM, Farmer RG. Course<br />

and prognosis of idiopathic ulcerative proctosigmoiditis<br />

in young patients. J Pediatr Gastroenterol Nutr 1986; 5:<br />

571–575.<br />

301. Ekbom A. Risk factors and distinguishing features of<br />

cancer in IBD. Inflamm Bowel Dis 1998; 4: 235–243.<br />

302. Kane S. Inflammatory bowel disease in pregnancy.<br />

Gastroenterol Clin North Am 2003; 32: 323–340.<br />

303. Baird DD, Narendranathan M, Sandler RS. Increased<br />

risk of preterm birth for women with inflammatory<br />

bowel disease. Gastroenterology 1990; 99: 987–994.<br />

304. Norgard B, Fonager K, Sorensen HT et al. Birth<br />

outcomes of women with ulcerative colitis: a nationwide<br />

Danish cohort study. Am J Gastroenterol 2000; 95:<br />

3165–3170.<br />

305. Ording Olsen K, Juul S, Berndtsson I et al. Ulcerative<br />

colitis: female fecundity before diagnosis, during<br />

disease, and after surgery compared with a population<br />

sample. Gastroenterology 2002; 122: 15–19.<br />

306. Devroede GJ, Taylor WF, Sauer WG et al. Cancer risk<br />

and life expectancy of children with ulcerative colitis. N<br />

Engl J Med 1971; 285: 17–21.<br />

307. Ekbom A, Helmick C, Zack M et al. Ulcerative colitis<br />

and colorectal cancer. A population-based study. N Engl<br />

J Med 1990; 323: 1228–1233.<br />

308. Brostrom O, Lofberg R, Nordenvall B et al. The risk of<br />

colorectal cancer in ulcerative colitis. An epidemiologic<br />

study. Scand J Gastroenterol 1987; 22: 1193–1199.<br />

309. Gilat T, Fireman Z, Grossman A et al. Colorectal cancer<br />

in patients with ulcerative colitis. A population study in<br />

central Israel. Gastroenterology 1988; 94: 870–877.<br />

310. Broome U, Lofberg R, Veress B et al. Primary sclerosing<br />

cholangitis and ulcerative colitis: evidence for increased<br />

neoplastic potential. Hepatology 1995; 22: 1404–1408.<br />

311. Brentnall TA, Haggitt RC, Rabinovitch PS et al. Risk and<br />

natural history of colonic neoplasia in patients with<br />

primary sclerosing cholangitis and ulcerative colitis.<br />

Gastroenterology 1996; 110: 331–338.<br />

References 417<br />

312. Shetty K, Rybicki L, Brzezinski A et al. The risk for<br />

cancer or dysplasia in ulcerative colitis patients with<br />

primary sclerosing cholangitis. Am J Gastroenterol 1999;<br />

94: 1643–1649.<br />

313. Jayaram H, Satsangi J, Chapman RW. Increased colorectal<br />

neoplasia in chronic ulcerative colitis complicated<br />

by primary sclerosing cholangitis: fact or fiction? Gut<br />

2001; 48: 430–434.<br />

314. Soetikno RM, Lin OS, Heidenreich PA et al. Increased<br />

risk of colorectal neoplasia in patients with primary<br />

sclerosing cholangitis and ulcerative colitis: a metaanalysis.<br />

Gastrointest Endosc 2002; 56: 48–54.<br />

315. Pardi DS, Loftus EV Jr, Kremers WK et al.<br />

Ursodeoxycholic acid as a chemopreventive agent in<br />

patients with ulcerative colitis and primary sclerosing<br />

cholangitis. Gastroenterology 2003; 124: 889–893.<br />

316. Askling J, Dickman PW, Karlen P et al. Family history as<br />

a risk factor for colorectal cancer in inflammatory bowel<br />

disease. Gastroenterology 2001; 120: 1356–1362.<br />

317. Heuschen UA, Hinz U, Allemeyer EH et al. Backwash<br />

ileitis is strongly associated with colorectal carcinoma<br />

in ulcerative colitis. Gastroenterology 2001; 120:<br />

841–847.<br />

318. Lindberg BU, Broome U, Persson B. Proximal colorectal<br />

dysplasia or cancer in ulcerative colitis. The impact of<br />

primary sclerosing cholangitis and sulfasalazine: results<br />

from a 20-year surveillance study. Dis Colon Rectum<br />

2001; 44: 77–85.<br />

319. Lashner BA, Heidenreich PA, Su GL et al. Effect of<br />

folate supplementation on the incidence of dysplasia<br />

and cancer in chronic ulcerative colitis. A case–control<br />

study. Gastroenterology 1989; 97: 255–259.<br />

320. Judge TA, Lewis JD, Lichtenstein GR. Colonic dysplasia<br />

and cancer in inflammatory bowel disease. Gastrointest<br />

Endosc Clin North Am 2002; 12: 495–523.<br />

321. Bernstein CN, Eaden J, Steinhart AH et al. Cancer<br />

prevention in inflammatory bowel disease and the<br />

chemoprophylactic potential of 5-aminosalicylic acid.<br />

Inflamm Bowel Dis 2002; 8: 356–361.<br />

322. van Hogezand RA, Eichhorn RF, Choudry A et al.<br />

Malignancies in inflammatory bowel disease: fact or<br />

fiction? Scand J Gastroenterol Suppl 2002: 48–53.<br />

323. Griffiths AM, Sherman PM. Colonoscopic surveillance<br />

for cancer in ulcerative colitis: a critical review. J<br />

Pediatr Gastroenterol Nutr 1997; 24: 202–210.<br />

324. Winawer S, Fletcher R, Rex D et al. Colorectal cancer<br />

screening and surveillance: clinical guidelines and<br />

rationale. Update based on new evidence.<br />

Gastroenterology 2003; 124: 544–560.<br />

325. Hanauer SB, Kane S. The pharmacology of anti-inflammatory<br />

drugs in inflammatory bowel disease. In Kirsner<br />

JB, ed. Inflammatory Bowel Diseases. Philadelphia: WB<br />

Saunders, 2000: 510–518.<br />

326. Makins RJ, Cowan RE. 5-amino-salicylate in the<br />

management of inflammatory bowel disease. Colorectal<br />

Dis 2001; 3: 218–222.


26<br />

Introduction<br />

Vasculitides<br />

Salvatore Cucchiara and Osvaldo Borrelli<br />

Vasculitides is a general term for a group of<br />

diseases characterized by inflammation within or<br />

around the wall of blood vessels, with alteration of<br />

the vascular blood flow and deranged integrity of<br />

the vessels, leading to ischemia and necrosis of the<br />

dependent organs. Blood vessels of all sizes may<br />

be affected, from the largest vessel in the body (the<br />

aorta) to the smallest vessels in the skin (capillaries).<br />

The size of the affected blood vessels varies<br />

according to the specific type of vasculitis. 1–3 The<br />

symptoms of vasculitides depend on the blood<br />

vessels involved in the inflammatory process.<br />

However, vasculitides are a systemic illness, and<br />

Table 26.1 Classification of the vasculitic syndromes<br />

Systemic necrotizing vasculitis<br />

polyarteritis nodosa (PAN) (classic PAN, microscopic polyangiitis)<br />

allergic angiitis and granulomatosis of Churg–Strauss<br />

polyangiitis overlap syndrome<br />

patients have fever, weight loss, fatigue, a rapid<br />

pulse, and diffuse aches and pains. In some cases,<br />

identifying the source and underlying cause of the<br />

pain is extremely challenging. In addition to these<br />

diffuse, poorly localized systemic symptoms,<br />

vasculitides may involve virtually every organ<br />

system in the body.<br />

It should be stressed that, although several specific<br />

entities are identified, different vasculitides may<br />

also have overlapping features. Table 26.1 reports<br />

a classification that takes into account the heterogeneity<br />

of vasculitides, and also the fact that some<br />

syndromes are mainly systemic, leading to<br />

progressive organ system dysfunction, whereas<br />

Wegener’s granulomatosis<br />

Temporal arteritis<br />

Takayasu’s arteritis<br />

Henoch–Schönlein purpura<br />

Predominantly cutaneous vasculitis (hypersensitivity vasculitis)<br />

exogenous stimuli proven or suspected (drug-induced, serum sickness and serum sickness-like reactions,<br />

associated with infectious diseases)<br />

endogenous antigens likely to be involved (associated with neoplasms, with connective tissue diseases, with other<br />

underlying diseases, or with congenital deficiencies of the complement system)<br />

Other vasculitic syndromes<br />

Kawasaki disease<br />

isolated central nervous system vasculitis<br />

thromboangiitis obliterans (Buerger’s disease)<br />

Behçet’s syndrome<br />

miscellaneous vasculitis<br />

419


420<br />

Vasculitides<br />

others are predominantly localized to the skin and<br />

rarely cause dysfunction of vital organs. Organ<br />

systems that can be affected by the vasculitic<br />

process are: skin, joints, lungs, kidneys, gastrointestinal<br />

tract, blood, sinuses, nose and ears, eyes,<br />

brain and nerves. In general, however, any type of<br />

systemic vasculitis can affect the gastrointestinal<br />

tract. For practical clinical work, vasculitides can<br />

also be differentiated into ‘primary’ and ‘secondary’.<br />

Primary vasculitides are not associated with<br />

systemic entities and the type of the disease<br />

depends on the caliber of the involved vessels.<br />

Secondary vasculitides are associated with underlying<br />

diseases, mainly rheumatic or connective<br />

tissue diseases, malignancy, infections, drugs and<br />

toxic agents. Rheumatic and autoimmune<br />

diseases, a cause of intestinal vasculitides, are<br />

systemic lupus erythematosus (SLE), rheumatoid<br />

arthritis, systemic sclerosis, dermatomyositis,<br />

primary biliary cirrhosis and autoimmune hepatitis.<br />

However, the most common infectious diseases<br />

underlying vasculitides are bacterial (streptococci,<br />

borrelia, mycobacteria, mycoplasms), viral (hepatitis,<br />

cytomegalovirus, herpes, HIV) and fungal.<br />

Vasculitides involving the gastrointestinal tract are<br />

part of a systemic process, although signs and<br />

symptoms may initially be limited. The onset of<br />

clinical manifestations may be acute and severe<br />

(mesenteric infarction) or chronic, due to mesenteric<br />

ischemia. 4 Acute intestinal vasculitides<br />

present as a rapidly evolving disorder, with a clinical<br />

picture mimicking embolic or thrombotic<br />

ischemia: intensive abdominal pain, signs of peritonitis<br />

and ileus. Chronic intestinal vasculitides<br />

can mimic each type of gastrointestinal disorder<br />

and require a detailed diagnostic procedure. The<br />

main symptoms of chronic vasculitides involving<br />

the intestine include abdominal pain, weight loss,<br />

nausea, vomiting, diarrhea, hematochezia and<br />

tenesmus. Endoscopy of both the upper and lower<br />

gastrointestinal tract often shows signs of inflammation<br />

such as petechiae, erosions and small<br />

ulcerations. Affected patients may also present<br />

acute episodes of small-bowel obstruction<br />

secondary to strictures, resembling Crohn’s disease<br />

or intussusceptions, or with massive gastrointestinal<br />

bleeding, secondary to aneurysm formation.<br />

4 In addition to mesenteric ischemia, systemic<br />

vasculitides can also cause ischemic damage to the<br />

liver, pancreas, gallbladder and, less commonly,<br />

esophagus or stomach.<br />

Epidemiology<br />

Certain forms of vasculitis occur more commonly<br />

in specific age groups. Vasculitides in childhood<br />

are dominated by Kawasaki disease and<br />

Henoch–Schönlein purpura, whereas primary<br />

systemic vasculitides, as well as conditions that<br />

can be complicated by a vasculitic process (e.g.<br />

SLE and juvenile dermatomyositis) are rarely<br />

reported in children. Furthermore, certain ethnic<br />

groups are at higher risk than others. The incidence<br />

of Kawasaki disease in children younger<br />

than 5 years doubled from 4.0 per 100000 in<br />

1991–92 to 8.1 per 100000 in 1999–2000, whereas<br />

for Henoch–Schönlein purpura the annual incidence<br />

is about 20 per 100000. 5 Recent data from<br />

the West Midlands of the UK indicated an incidence<br />

of 0.24 per 100000 for primary systemic<br />

vasculitides including cases of polyarteritis<br />

nodosa (PAN), microscopic polyangiitis, Wegener’s<br />

granulomatosis and Behçet’s disease. 6<br />

Pathogenesis<br />

Most of the vasculitides are thought to be mediated<br />

by immunopathogenetic mechanisms, although<br />

the evidence for this is mainly indirect. Three<br />

potential mechanisms of vessel damage in vasculitides<br />

have been suggested. 7<br />

Pathogenetic immune-complex formation<br />

The mechanism of tissue damage in immune<br />

complex diseases is derived from the occurrence of<br />

antigen–antibody complexes formed in antigen<br />

excess and deposited in vessel walls, whose<br />

permeability is increased by several vasoactive<br />

amines. The deposition of complexes determines<br />

the activation of complement components, mainly<br />

C5a, that recruit neutrophils. Neutrophils infiltrate<br />

the vessel wall, phagocytose the immune<br />

complexes and release enzymes that damage the<br />

vessel wall. When the process becomes subacute<br />

or chronic, an infiltrate of mononuclear cells<br />

predominates. The causal role of deposition of<br />

immune complexes in the wall of vessels has not<br />

been clearly defined in most vasculitides. Indeed,<br />

many patients with vasculitides do not show<br />

circulating or deposited immune complexes.


Antineutrophil cytoplasmic antibodies<br />

These are antibodies directed against certain<br />

proteins of the neutrophil cytoplasm, and are<br />

detected in a high percentage of patients with<br />

systemic vasculitides (particularly Wegener’s granulomatosis),<br />

microscopic polyangiitis and in those<br />

with necrotizing and crescentic glomerulonephritis.<br />

Two main categories of antineutrophil cytoplasmic<br />

antibodies (ANCA) are recognized, based<br />

on different targets: cytoplasmic ANCA (c-ANCA)<br />

have a diffuse cytoplasmic staining pattern at<br />

immunofluorescence microscopy, when<br />

proteinase-3 (located in neutrophil azurophilic<br />

granules) is the c-ANCA antigen; and perinuclear<br />

ANCA (p-ANCA) which have a perinuclear or<br />

nuclear staining pattern when the targets are<br />

myeloperoxidase, elastase, cathepsin G, lactoferrin,<br />

lysozyme or bactericidal/permeability-increasing<br />

protein. It is not clear why patients with<br />

certain vasculitides develop ANCA, but, once<br />

ANCA are present, they can contribute to the<br />

pathogenesis of the vasculitides. It is conceivable<br />

that when neutrophils are primed by tumor necrosis<br />

factor (TNF)-α or interleukin (IL)-1, intracytoplasmatic<br />

proteinase-3 translocates to the cell<br />

membrane, where it interacts with extracellular<br />

ANCA. The neutrophils degranulate and reactive<br />

oxygen species are delivered, causing tissue<br />

damage. Tanslocation of proteinase-3 close to the<br />

cell membrane also occurs in the endothelial cells<br />

upon priming with TNF-α, IL-1, or interferon<br />

(IFN)γ, thus making them susceptible to interaction<br />

with ANCA and consequent tissue damage<br />

due to complement-mediated cytotoxicity or antibody-dependent<br />

cellular toxicity. Although these<br />

in vitro data are attractive as explanations, there<br />

are no conclusive data that ANCA play a direct<br />

role in the pathogenesis of vasculitides, as they<br />

may represent epiphenomena.<br />

Pathogenic T-lymphocyte responses and<br />

granuloma formation<br />

Vascular endothelial cells can express HLA class II<br />

molecules following activation by cytokines such<br />

as IFNγ. They can take part in immunological reactions,<br />

such as interaction with CD4+ lymphocytes,<br />

similarly to antigen-presenting macrophages. They<br />

can also secrete IL-1, which is a powerful inducer<br />

of adhesion molecules such as endothelial leuko-<br />

Clinical manifestations 421<br />

cyte adhesion molecule 1 (ELAM-1) and vascular<br />

cell adhesion molecule 1 (VCAM-1) with consequent<br />

adhesion of leukocytes to the endothelial<br />

cells in the blood vessel wall. There is no strong<br />

evidence that other mechanisms, such as direct<br />

cellular cytotoxicity or antibodies directed against<br />

vessel components or antibody-dependent cellular<br />

cytotoxicity, have a causal contribution to the<br />

pathogenesis of vasculitides.<br />

Many aspects of the pathogenesis of vasculitides<br />

remain unclear. Other variables certainly implicated<br />

in the development of vasculitides are:<br />

genetic predisposition; defects of the regulatory<br />

factors of the immune responses to certain antigens;<br />

the ability of the reticuloendothelial system<br />

in clearing circulating complexes; the size and<br />

physicochemical properties of immune complexes;<br />

the degree of turbulence of blood flow; and the<br />

intravascular hydrostatic pressure in different<br />

vessels.<br />

Clinical manifestations<br />

It is clinically useful to distinguish between acute<br />

and chronic intestinal vasculitides, although both<br />

may have a severe course with deleterious effects.<br />

Acute intestinal vasculitis is a rapidly evolving<br />

disease, which presents with intense abdominal<br />

pain, followed by signs of peritonitis and ileus,<br />

and progressive cardiovascular shock syndrome<br />

with high lethality. It is often difficult to distinguish<br />

mesenteric ischemia due to vasculitides<br />

clinically from embolic or thrombotic ischemia in<br />

patients complaining of severe abdominal pain. It<br />

is therefore necessary to collect the patient’s<br />

history carefully, as well as clinical and laboratory<br />

parameters, in order to reach the diagnosis and<br />

begin treatment. Standard emergency laboratory<br />

values, serum concentration of lactate and coagulation<br />

parameters as well as disease-specific values<br />

must be measured. Table 26.2 gives a number of<br />

tests that are useful in establishing a link between<br />

‘vasculitis’ and an underlying disease. Every<br />

patient suspected of having ischemia should<br />

receive an abdominal ultrasonography including<br />

Doppler examination of the mesenteric arteries<br />

and an X-ray of the abdomen for assessing ileus,<br />

intestinal edema and perforation. If findings of<br />

mesenteric ischemia are evident or suspected,


422<br />

Vasculitides<br />

Table 26.2 Clinical features that raise suspicion of vasculitides<br />

General clinical feature Signs or presenting disorders Type of vasculitis<br />

Constitutional symptoms fever, fatigue, malaise,<br />

anorexia, weight loss<br />

any type of vasculitis<br />

Polymyalgia rheumatica proximal muscle pain with giant cell arteritis; less commonly<br />

morning stiffness other vasculitides<br />

Non-destructive oligoarthritis joint swelling, warmth, painful polyarteritis, Wegener’s<br />

range of motion granulomatosis,<br />

Churg–Strauss vasculitis<br />

Skin lesions livedo reticularis, necrotic lesions, polyarteritis, Wegener’s<br />

ulcers, nodules, digital tip infarcts granulomatosis, Churg–Strauss<br />

vasculitis, hypersensitivity<br />

vasculitis<br />

palpable purpura any type of vasculitis except<br />

giant cell arteritis and<br />

Takayasu’s arteritis<br />

Multiple mononeuropathy injury to two or more separate polyarteritis, Takayasu’s<br />

(mononeuritis multiplex) peripheral nerves (e.g. patient arteritis; less commonly,<br />

presents with both right foot Churg–Strauss vasculitis<br />

drop and left wrist drop) and Wegener’s granulomatosis<br />

angiography and subsequent surgery should be<br />

planned.<br />

The diagnosis of chronic vasculitis should be<br />

considered in young children and adolescents<br />

with systemic symptoms and evidence of single<br />

and/or multiorgan specific dysfunction. In these<br />

cases, common complaints are fatigue, weakness,<br />

fever, arthralgia and abdominal pain. Common<br />

signs include fever, hypertension, neurological<br />

dysfunction and renal abnormalities with an<br />

active urine sediment (containing red cells, other<br />

cellular and granular casts). The above findings<br />

are neither sensitive nor specific for vasculitides,<br />

and the diagnosis is often delayed because the<br />

clinical manifestations of the vasculitides mimic<br />

those of other more common disorders. In addition,<br />

although each type of vasculitis usually has a<br />

characteristic pattern of organ involvement, a<br />

single approach to the diagnosis is difficult, owing<br />

glomerulonephritis microscopic polyangiitis,<br />

Wegener’s granulomatosis,<br />

cryoglobulinemia,<br />

Churg–Strauss vasculitis,<br />

Henoch–Schönlein purpura<br />

to the variability of the manifestations of the<br />

vasculitides. All children suspected of having<br />

vasculitides should undergo a complete history<br />

and physical examination and certain basic laboratory<br />

tests (Figure 26.1). Both upper and lower<br />

intestinal endoscopy frequently reveal signs of<br />

mucosal inflammation, such as multiple<br />

petechiae, friability, erosions and small ulcers. A<br />

number of serological markers, such as antinuclear<br />

antibodies (ANA) and ANCA are associated with<br />

specific types of vasculitis, but tissue biopsy is<br />

often required to establish the diagnosis.<br />

History<br />

Diagnostic work-up starts with a detailed patient<br />

history. Postprandial abdominal pain that usually<br />

begins 30–60min after meals is a predominant<br />

symptom. Other aspects of the history include


whether the patient has recently been ill or taken<br />

medications or drugs, has a history of hepatitis<br />

(virus C), or has been diagnosed with any disorder<br />

known to be associated with vasculitis. Knowing<br />

the propensity of certain disorders to occur in<br />

specific ages, gender, or ethnic groups can be<br />

useful in making the diagnosis. Table 26.2 outlines<br />

clinical features that raise the suspicion of<br />

vasculitis.<br />

Physical examination<br />

I<br />

Patient with symptoms and<br />

signs suggesting vasculitis<br />

History and clinical features Laboratory tests<br />

II<br />

III<br />

Determine category of<br />

the vasculitis syndrome<br />

Treatment<br />

Establish diagnosis<br />

A careful physical examination helps in determining<br />

the extent of vascular lesions, the distribution<br />

of affected organs and the presence of additional<br />

Yes<br />

Biopsy<br />

Determine pattern and<br />

extent of the disease<br />

Search for antigens Search for underlying disease<br />

Antigen removal<br />

Treat vasculitis<br />

Treat underlying disease<br />

Treat vasculitis<br />

Figure 26.1 Suggested algorithm for the approach to patients with suspected vasculitis.<br />

No<br />

Clinical manifestations 423<br />

Angiogram if appropriate<br />

Re-evaluation<br />

disease processes. The type and extent of organ<br />

involvement in vasculitis can be helpful in determining<br />

the specific type of vasculitides and the<br />

degree of urgency in initiating treatment. The clinical<br />

features in a given patient can be used to<br />

discern the size of vessels affected by vasculitis<br />

(Table 26.3). Table 26.4 reports the frequency of<br />

intestinal involvement in different vasculitides.<br />

Incomplete stenosis of one of the major vessels<br />

(e.g. mesenteric artery) may cause changes in the<br />

Doppler ultrasound flow pattern, as well as typical<br />

bruits at abdominal auscultation. In three series<br />

with a total of 351 patients, approximately onethird<br />

had gastrointestinal manifestations. 8 In a<br />

large series of patients with PAN or Churg–Strauss<br />

syndrome, for example, the following frequency of


424<br />

Vasculitides<br />

Table 26.3 Clues for identifying the type of vessel involvement in vasculitides<br />

Most commonly associated<br />

Clinical feature Vessels most likely to be affected systemic vasculitis<br />

Cutaneous<br />

Palpable purpura post-capillary venules any type of vasculitis except giant cell<br />

arteritis and Takayasu’s arteritis<br />

Skin ulcers arterioles to small arteries polyarteritis, Churg–Strauss vasculitis<br />

Wegener’s granulomatosis,<br />

hypersensitivity vasculitis<br />

Gangrene in an small to medium-sized arteries polyarteritis, Churg–Strauss vasculitis,<br />

extremity Wegener’s granulomatosis<br />

Gastrointestinal tract<br />

Abdominal pain or small to medium-sized arteries Henoch–Schönlein purpura,<br />

mesenteric ischemia polyarteritis, Churg–Strauss vasculitis<br />

Gastrointestinal bleeding capillaries to medium-sized Henoch–Schönlein purpura,<br />

arteries polyarteritis, Churg–Strauss vasculitis<br />

Renal<br />

Glomerulonephritis capillaries microscopic polyangiitis, Wegener’s<br />

granulomatosis, cryoglobulinemia,<br />

Churg–Strauss vasculitis,<br />

Henoch–Schönlein purpura<br />

Ischemic renal failure small to medium-sized arteries polyarteritis, Takayasu’s arteritis; less<br />

commonly, Churg–Strauss vasculitis,<br />

Wegener’s granulomatosis<br />

Pulmonary<br />

Pulmonary hemorrhage capillaries; less commonly microscopic polyangiitis, Wegener’s<br />

small to medium-sized arteries granulomatosis<br />

Pulmonary infiltrate small to medium-sized arteries Churg–Strauss vasculitis,<br />

or cavities microscopic polyangiitis<br />

Neurological<br />

Peripheral neuropathy small arteries polyarteritis, Churg–Strauss vasculitis,<br />

Wegener’s granulomatosis,<br />

cryoglobulinemia<br />

Stroke small, medium-sized or giant cell arteritis, SLE-associated<br />

large arteries vasculitis<br />

SLE, systemic lupus erythematosus<br />

gastrointestinal symptoms and findings was noted:<br />

abdominal pain (25%), gastrointestinal bleeding<br />

(7%), peritonitis (4%), intestinal infarction (2%),<br />

pancreatitis (2%), duodenal ulcer (2%) and cholecystitis<br />

(1%), while ischemic hepatitis, gastritis<br />

and esophagitis occurred more rarely (


urea nitrogen, creatinine, liver enzymes, erythrocyte<br />

sedimentation rate (ESR), hepatitis serological<br />

markers and chest radiograph. Additional tests<br />

that may be required are cerebrospinal fluid analysis,<br />

central nervous system imaging, pulmonary<br />

function testing, and blood and tissue culture.<br />

These tests assess the extent of organ involvement<br />

and may suggest, confirm, or exclude additional<br />

diagnostic considerations. More specific laboratory<br />

tests may be indicated, depending upon the<br />

diagnosis being considered (Table 26.5).<br />

Imaging studies<br />

Imaging studies, such as computed tomography<br />

(CT) scanning or barium studies, are frequently<br />

normal in patients with symptomatic intestinal<br />

vasculitides and, when abnormal, reveal nonspecific<br />

abnormalities such as a thickened edematous<br />

bowel wall, unless a mesenteric infarction has<br />

occurred. Angiography may be useful in diagnosing<br />

small and medium vessel vasculitides, such as<br />

PAN. Nuclear medicine scanning using white<br />

blood cell markers such as indium-111 may localize<br />

bowel inflammation in vasculitis, but its clinical<br />

utility is uncertain.<br />

Endoscopy<br />

Endoscopy should be performed with great<br />

caution in patients with vasculitides because of an<br />

Clinical manifestations 425<br />

Table 26.4 Frequency of intestinal involvement in different vasculitides (from references 24, 38–47)<br />

Type of vasculitis Frequency of intestinal involvement (%)<br />

Primary vasculitis<br />

Polyarteritis nodosa (PAN) (classic PAN, microscopic polyangiitis) 30–50<br />

Churg–Strauss syndrome 25–50<br />

Behçet’s disease up to 30<br />

Takayasu’s arteritis up to 15<br />

Wegener’s granulomatosis 5–10<br />

Giant cell arteritis 1<br />

Secondary vasculitis<br />

Henoch–Schönlein purpura 50–90<br />

Systemic lupus erythematosus 50<br />

Rheumatoid arthritis vasculitis 10<br />

elevated risk of perforation of an edematous and<br />

ischemic bowel. In these cases, magnetic resonance<br />

imaging (MRI) of the intestinal tract can be<br />

used. As demonstrated recently, the two techniques<br />

show a good correlation in detecting<br />

inflammation of the intestinal mucosa. 9<br />

Tissue biopsy<br />

Biopsy examination of the involved tissue is<br />

almost always necessary for diagnosis. Obviously,<br />

specimens are more likely to yield a diagnosis<br />

when they are of adequate size and obtained from<br />

an involved area of an organ. The histological findings<br />

evolve as lesions progress and may vary from<br />

acute inflammation to healing and repair. The<br />

biopsy findings in the various vasculitides are<br />

discussed below (see Specific disorders). The decision<br />

to obtain a tissue biopsy is based upon the<br />

overall clinical assessment of the patient and<br />

complications associated with the procedure. As<br />

an example, the skin, which characteristically is<br />

involved in children with leukocytoclastic vasculitis,<br />

is easily accessible for biopsy, and skin biopsy<br />

in this setting has low morbidity. However, in this<br />

scenario, the biopsy adds little, if any, new information.<br />

The biopsy findings of leukocytoclastic<br />

vasculitis (inflammation of the small blood vessels<br />

most prominent in the post-capillary venules) are<br />

relatively non-specific, unless accompanied by<br />

vascular IgA deposition, which establishes a diag-


426<br />

Vasculitides<br />

Table 26.5 Laboratory parameters useful in the diagnostic approach to vasculitis<br />

Laboratory parameters Purpose of interpretation<br />

Routine tests (including complete blood cell evaluate for hematologic, renal and other<br />

count, liver enzymes, creatinine, urinalysis) organ involvement<br />

Blood cultures rule out infection<br />

ESR, C-reactive protein high values suggest active inflammatory disease<br />

Eosinophilia Churg–Strauss syndrome<br />

Anti-nuclear antibodies screen for SLE and Sjögren’s syndrome<br />

Anti-double-stranded DNA antibodies suggest SLE<br />

Rheumatoid factor very high titers in rheumatoid arthritis, Sjögren’s<br />

syndrome and cryoglobulinemia-associated vasculitis<br />

Complement (C3, C4, CH50) low complement levels suggest consumption by<br />

immune complexes, which are commonly found in<br />

SLE and cryoglobulinemia<br />

Cytoplasmic anti-neutrophil cytoplasmic<br />

(anti-proteinase-3) antibodies<br />

Wegener’s granulomatosis<br />

Perinuclear anti-neutrophil cytoplasmic<br />

antibodies (anti-myeloperoxidase)<br />

panarteritis nodosa, microscopic polyangiitis<br />

Creatine phosphokinase elevation suggests myositis, which can occur<br />

in many vasculitis syndromes<br />

Anti-glomerular basement membrane rule out Goodpasture’s syndrome, which can<br />

mimic vasculitis and cause pulmonary<br />

hemorrhage and glomerulonephritis<br />

Circulating immune complexes immune complex vasculitides<br />

HLA B51 suggests Behçet’s syndrome<br />

HLA B8 suggests SLE<br />

ESR, erythrocyte sedimentation rate; SLE, systemic lupus erythematosus<br />

nostic of Henoch–Schönlein purpura. 10 However,<br />

the diagnosis of Henoch–Schönlein purpura in<br />

children is usually made on clinical grounds, and<br />

biopsy may not be necessary.<br />

Arteriography<br />

Arteriograms can identify and characterize<br />

vasculitides of large and medium-sized arteries<br />

and giant cell arteritis with an aortic arch<br />

syndrome, whereas they are not helpful in the<br />

evaluation of vasculitides involving small vessels,<br />

because these are not visible with routine<br />

angiograms. Angiographic abnormalities may not<br />

be pathognomonic, but usually support a diagno-<br />

sis when combined with other clinical data. The<br />

decision to perform angiography in a patient with<br />

suspected vasculitis depends on the overall clinical<br />

judgement and assessment of the risk for<br />

complications associated with the procedure.<br />

Arteriography alone is usually performed to<br />

confirm the diagnosis of Takayasu’s arteritis: the<br />

arteriography changes tend to be most pronounced<br />

in the region of the aortic arch and its primary<br />

branches, which are relatively inaccessible for<br />

biopsy. In the patient with suspected PAN without<br />

an obvious area for biopsy, a mesenteric angiogram<br />

should be planned, particularly in the presence of<br />

abdominal pain: angiograms of mesenteric or renal<br />

arteries may show aneurysms, occlusions and


vascular wall irregularities. Magnetic resonance<br />

arteriography of the aorta and great vessels including<br />

some major branches such as the coronaries<br />

and renal arteries may be an alternative to contrast<br />

arteriography in children. When the two techniques<br />

were compared in ten children with<br />

various vascular lesions (age range: 1 month to 16<br />

years), excellent correlation was found between<br />

them. 11<br />

Specific disorders<br />

Primary vasculitides<br />

Polyarteritis nodosa<br />

PAN affects medium and small-sized arteries and<br />

can cause a wide spectrum of gastrointestinal<br />

symptoms and signs, which are mainly due to<br />

visceral ischemia and are detected in about twothirds<br />

of patients. Common intestinal features are:<br />

epigastric pain, nausea, anorexia, mucosal ulcerations<br />

(Figure 26.2), bleeding and diarrhea. The<br />

most severe intestinal findings are infarction,<br />

perforation, pneumatosis intestinalis and<br />

pseudomembranous colitis. 4 Survival after bowel<br />

Figure 26.2 A large (thick arrow) and a small (thin<br />

arrow) ulceration in a 14-year-old boy with polyarteritis<br />

nodosa at the level of the descending colon. The large<br />

ulceration is surrounded by hyperemic and friable<br />

mucosa.<br />

Specific disorders 427<br />

infarction requiring surgery is poor. Typical<br />

aneurysmal dilatation of small and medium-sized<br />

arteries on arteriogram will provide a definite diagnosis.<br />

In a series of 16 patients with severe PAN,<br />

five developed an abdominal crisis related to the<br />

disease: at laparotomy, all had evidence of mesenteric<br />

arteritis with infarcted bowel and intestinal<br />

perforation. 12 In another series of 165 patients<br />

with PAN or Churg–Strauss syndrome, 31% of<br />

deaths occurring at the follow-up were attributable<br />

to gastrointestinal disease (bleeding, peritonitis<br />

and pancreatitis). 12 Vasculitis of the hepatic arteries<br />

may cause elevated levels of transaminases,<br />

bilirubin and alkaline phosphatase.<br />

Treatment with corticosteroids and cyclophosphamide<br />

has relieved symptoms and improved the<br />

overall survival of these patients. 13 Adequate<br />

therapy for patients with PAN related to hepatitis<br />

B virus infection or hairy cell leukemia may<br />

require treatment of these underlying disorders. 12<br />

The tendency of PAN to involve large vessels has<br />

an important long-term implication. Some patients<br />

with PAN have gastrointestinal symptoms and<br />

other manifestations at a time when the disease is<br />

clinically inactive, owing to effective immunosuppressive<br />

therapy. In this setting, healing of<br />

inflamed vessels has led to progressive narrowing<br />

of the vascular bed.<br />

Churg–Strauss syndrome<br />

The Churg–Strauss syndrome (also called ‘allergic<br />

angiitis and granulomatosis’) is a multisystemic<br />

disease characterized by hypereosinophilia,<br />

pulmonary infiltration, extravascular granulomas<br />

and necrotizing vasculitis affecting small and<br />

medium-sized arteries and veins. 12 Involvement of<br />

the intestinal vasculature ranges from 25 to 50%<br />

and any intestinal organ can be affected with<br />

features such as ulcerations, bleeding, ileus, perforation<br />

and cholecystitis. Interestingly, microscopic<br />

polyangiitis resembles Churg–Strauss syndrome<br />

for the spectrum of gastrointestinal features as well<br />

as for its similar response to corticosteroids and<br />

cyclophosphamide. 14 Intestinal histology can<br />

reveal both eosinophilic infiltrates around the<br />

vessels and eosinophilic gastroenteritis. The<br />

syndrome commonly occurs in middle-aged adults<br />

and is rare in children. A 15-year-old girl with<br />

multiple colonic ulcers has recently been<br />

described. This patient also had vasculitic involve-


428<br />

Vasculitides<br />

ment of the major organs including migratory<br />

pulmonary infiltrates, myocarditis and central<br />

nervous system disease with hemiparesis. 15<br />

Wegener’s granulomatosis<br />

Wegener’s granulomatosis is characterized by a<br />

granulomatous (peri)vascular inflammation of the<br />

upper and lower airways combined with glomerulonephritis.<br />

It appears to be a disorder restricted to<br />

Caucasians, since only 3% of patients with<br />

Wegener’s granulomatosis are of other ethnic<br />

groups. Interestingly, although c-ANCA is considered<br />

a specific marker, it is not yet an accepted<br />

criterion for the diagnosis of Wegener’s granulomatosis.<br />

Gastrointestinal involvement seems to be<br />

rare, mostly producing granulomatous colitis and<br />

gastritis. 4 An unusual presentation of Wegener’s<br />

granulomatosis has been reported in a 16-year-old<br />

girl with symptoms and signs suggesting an<br />

inflammatory bowel disease (diarrhea, fever,<br />

weight loss, abdominal pain, arthralgias and<br />

mouth ulcers). However, biopsy specimens of<br />

rectal mucosa, oro- and nasopharynx, and skin<br />

conclusively demonstrated the vasculitic lesion of<br />

Wegener’s granulomatosis. 16<br />

Behçet’s disease<br />

Behçet’s disease is a necrotizing vasculitis of<br />

unknown etiology primarily affecting young adult<br />

males. The predominant features are recurrent oral<br />

and genital ulcerations (more than three times per<br />

year) in combination with other signs, such as<br />

uveitis, iritis, retinal vasculitis, skin lesions<br />

(erythema nodosum, acneiform or papulopustulous<br />

lesions, pseudofolliculitis) and pathergy<br />

(papulopustulous lesions 24–48h after cutaneous<br />

injection of 0.9% sodium chloride). A strong<br />

genetic component is thought to be part of the<br />

disorder, owing to the association with HLA B51. 17<br />

The disease has the highest incidence in Turkey<br />

(up to 3000 per million), and in southern<br />

Mediterranean and Asiatic regions. Gastrointestinal<br />

involvement is found frequently and<br />

includes major intestinal arteries as well as upper<br />

and lower parts of the gut. Oral lesions resemble<br />

aphthous ulcers commonly observed in Crohn’s<br />

disease and may be single or multiple; esophageal<br />

ulcerations and erosions are found frequently at<br />

endoscopy (Figure 26.3), although they do not<br />

Figure 26.3 Mucosa of the distal esophagus in an 8year-old<br />

girl with Behçet’s disease. Diffuse hyperemia,<br />

erosions and ulcerations are evident, together with an<br />

open cardia.<br />

cause bleeding or perforation. Intestinal involvement<br />

at the level of both small and large bowel can<br />

resemble Crohn’s disease; bleeding, abdominal<br />

pain and perforation may result. 18,19 The optimal<br />

management of Behçet’s disease is uncertain, since<br />

the disease is rare and symptoms tend to wax and<br />

wane. The mucocutaneous lesions can be treated<br />

with topical or intralesional corticosteroids, or<br />

systemic colchicine, dapsone, thalidomide, or<br />

steroids. Systemic disease is typically treated with<br />

corticosteroids and immunosuppressive drugs<br />

such as azathioprine, cyclosporin, or cyclophosphamide.<br />

Interestingly, systemic Behçet’s disease<br />

has been successfully treated with anti-TNF-α<br />

agents such as thalidomide and monoclonal antibodies<br />

(infliximab). 20<br />

Takayasu’s arteritis<br />

Takayasu’s arteritis is a rare chronic vasculitis of<br />

unknown etiology that predominantly affects the<br />

aorta and its primary branches. 21,22 It characteristically<br />

affects young women, with an age of onset<br />

usually between 10 and 40 years (this is one of the<br />

major criteria for diagnosis). The prevalence is<br />

greatest in Asians. Gastrointestinal features result


from involvement of the large branches of the<br />

abdominal aorta such as the celiac trunk and the<br />

upper mesenteric artery. Although not a major<br />

feature, mesenteric ischemia can occur, leading to<br />

abdominal pain, diarrhea and gastrointestinal<br />

hemorrhage. An association of Takayasu’s arteritis<br />

with ulcerative colitis has also been reported. 23<br />

Severe cerebral, intestinal and coronary circulatory<br />

disturbances as well as an insufficient blood<br />

supply in both arms have been described in a 9month-old<br />

infant with extensive inflammatory<br />

lesions in the large arterial trunks originating at<br />

the aorta. 24<br />

Giant cell arteritis and polymyalgia rheumatica<br />

These two closely related vasculitides mainly<br />

affect the vessels of the shoulder girdle and the<br />

intra- and extra-cranial arteries; however, involvement<br />

of mesenteric vessels can also occur. 25<br />

Disseminated fibrinoid necrosis of small vessels<br />

can result in visceral ischemia, abdominal pain,<br />

nausea, anorexia, weight loss, bleeding and perforation.<br />

Hepatic artery involvement can determine<br />

disturbances of liver function and hepatitis with<br />

fibrin-ring granulomas. 26<br />

Kawasaki’s disease<br />

Also called mucocutaneous lymph node<br />

syndrome, Kawasaki’s disease is a multisystem<br />

vasculitis affecting children under the age of 5<br />

years. The principal symptoms are high fever for<br />

more than 5 days and resistance to antibiotics;<br />

polymorphonuclear cell exanthema, skin lesions<br />

in peripheral extremities (reddening, indurative<br />

edema, membranous desquamation), oral lesions<br />

(pharyngitis, enanthema, ‘strawberry tongue’),<br />

bilateral conjunctive congestion and acute cervical<br />

lymphadenopathy. In addition to these symptoms,<br />

gastrointestinal complications can determine deterioration<br />

of the overall condition of the patient.<br />

Intestinal features during the acute phase are<br />

abdominal pain, vomiting and diarrhea; whereas<br />

small-bowel obstruction has been reported as a<br />

consequence of acquired ischemic stricture. 27,28<br />

The vasculitic process underlying intestinal<br />

involvement in Kawasaki’s disease includes four<br />

stages: intensive perivasculitis, focal panvasculitis,<br />

ongoing inflammation and granulation and<br />

stenosis with aneurysms. The spectrum of<br />

Specific disorders 429<br />

gastroenterological complications also includes<br />

hepatitis, cholangitis, gallbladder hydrops and<br />

pancreatic ductitis.<br />

Secondary vasculitides<br />

Henoch–Schönlein purpura and leukocytoclastic<br />

vasculitis<br />

Henoch–Schönlein purpura is a generalized<br />

vasculitis of small to medium-sized vessels which<br />

characteristically occurs in children of all ages. It<br />

is secondary to an immune process that generates<br />

circulating immune complexes containing large<br />

amounts of IgA. Patients classically present lower<br />

extremity purpura, arthritis and acute glomerulonephritis.<br />

The gastrointestinal tract is frequently<br />

involved and intramural hemorrhages, bloody<br />

diarrhea or colic may result; erythema, friability<br />

and ulcerations of the gastrointestinal mucosa can<br />

be observed at endoscopy (Figure 26.4). Intestinal<br />

obstruction and intussusception have also been<br />

described. Gastrointestinal signs may have a recurrent<br />

course and may precede the onset of the characteristic<br />

purpuric rash, 29,30 which rarely may<br />

even be lacking. The clinical spectrum of gastro-<br />

Figure 26.4 Diffuse hyperemia and congestion with<br />

small erosions in a 6-year-old girl with Henoch–Schönlein<br />

purpura at the level of the gastric antrum.


430<br />

Vasculitides<br />

intestinal involvement in Henoch–Schönlein<br />

purpura ranges from a dull periumbilical pain to<br />

severe courses with intestinal perforation and<br />

necrosis of the bile ducts. Ultrasonography and CT<br />

provide useful information to support the diagnosis,<br />

which is commonly strongly suspected on<br />

clinical grounds. Confirmation of the diagnosis<br />

requires evidence by immunofluorescence microscopy<br />

of tissue deposition in the skin or kidney of<br />

IgA. Biopsy of the skin lesions reveals inflammation<br />

of the small blood vessels, called leukocytoclastic<br />

vasculitis (see below) that is most prominent<br />

in the post-capillary venules. The prognosis<br />

of this disease is good, except for cases with severe<br />

renal involvement. The efficacy of corticosteroids<br />

and immunosuppressants (azathioprine,<br />

cyclophosphamide) in the treatment of<br />

Henoch–Schönlein purpura is debated, especially<br />

when the kidneys are involved. 31 Some cases of<br />

Henoch–Schönlein purpura have been successfully<br />

treated by plasmapheresis. 32 Little experience<br />

concerning treatment of Henoch–Schönlein<br />

purpura during pregnancy exists, although corticosteroids<br />

and plasmapheresis have been used in<br />

these cases. 31<br />

Leukocytoclastic vasculitis<br />

Leukocytoclastic vasculitis is a disorder that<br />

mainly affects middle-aged patients (especially<br />

women), and has characteristic skin histopathological<br />

features revealing the presence of vascular<br />

and perivascular infiltration of polymorphonuclear<br />

leukocytes with formation of nuclear dust<br />

(leukocytoclasis), extravasation of erythrocytes<br />

and fibrinoid necrosis of the vessel walls. 7 This<br />

process is dynamic; and a biopsy of a lesion taken<br />

too early or too late in its evolution may not reveal<br />

these findings. The picture of leukocytoclastic<br />

vasculitis can occur in any vasculitic syndrome<br />

but may also occur in non-vasculitic diseases such<br />

as neutrophilic dermatoses, at the base of leg<br />

ulceration, or in some insect bite reactions. Careful<br />

clinical–pathological correlation is necessary. 7<br />

Immuno-fluorescent staining may reveal<br />

immunoglobulins (IgG, IgM) and complement<br />

components (C3, C4) deposited on the skin basement<br />

membrane, which are suggestive of immune<br />

complex deposition. 12<br />

Vasculitides in connective tissue diseases<br />

The term connective tissue disease includes<br />

numerous entities such as SLE, systemic sclerosis,<br />

polymyositis, dermatomyositis or Sjögren’s<br />

syndrome. Most clinical signs of these disorders<br />

are attributed to a small-vessel vasculitis, and<br />

gastrointestinal clinical problems most frequently<br />

occur in SLE.<br />

Systemic lupus erythematosus<br />

The vasculitis associated with SLE involves small<br />

and medium-sized vessels, and affects the<br />

gastrointestinal tract in up to 50% of patients.<br />

Lower abdominal pain secondary to mesenteric<br />

vasculitis is generally an insidious symptom that<br />

may be intermittent for months prior to the development<br />

of an acute abdomen with nausea, vomiting,<br />

diarrhea, bleeding and fever. Risk factors for<br />

the development of mesenteric vasculitis are<br />

peripheral vasculitis and central nervous system<br />

involvement. Patients with an acute presentation<br />

may also have mesenteric thrombosis and infarction,<br />

often in association with antiphospholipid<br />

antibodies. Mesenteric vasculitis is a potentially<br />

life-threatening disorder. In addition to the possible<br />

development of necrotic segments of bowel,<br />

patients may have septic complications and bowel<br />

perforation. In a series published in 1982, 15 of<br />

140 patients with SLE who required hospitalization<br />

developed a disease-related abdominal event<br />

and 11 underwent laparotomy. 33 The majority of<br />

the patients in this study had prodromic signs,<br />

such as insidious onset of intermittent, lower<br />

quadrant cramping and abdominal pain, which<br />

were present for an average of 34 days prior to the<br />

hospitalization. The presence of these symptoms<br />

in patients with SLE should raise suspicion of<br />

possible mesenteric vasculitis, thereby permitting<br />

early diagnosis and treatment. Interestingly,<br />

abdominal features such as pain, rectal bleeding<br />

by mucosal ulcers and protein-losing enteropathy<br />

due to increased mucosal permeability have been<br />

described as sole and initial manifestations in<br />

patients with SLE. 34,35 Chronic intestinal pseudoobstruction<br />

of the myogenic type has also been<br />

described to complicate the course of SLE. 36<br />

Simple laboratory tests may be helpful in making<br />

the diagnosis or in evaluating a flare-up. However,<br />

the diagnosis is mostly dependent on the historical


and physical findings. Antinuclear antibody, an<br />

antibody to nucleosomal DNA–histone complexes,<br />

is highly sensitive but not specific; anti-doublestranded<br />

DNA is more specific for SLE. This test<br />

may correlate with the degree of disease activity.<br />

Antiphospholipid antibodies are present in 30% of<br />

SLE patients.<br />

Antiphospholipid antibody syndrome<br />

The antiphospholipid antibody syndrome (APS),<br />

also known as Hughes syndrome, is a disorder<br />

characterized by multiple different antibodies that<br />

are associated with both arterial and venous<br />

thrombosis. There are three primary classes of<br />

antibodies associated with the APS: anticardiolipin<br />

antibodies; the lupus anticoagulant; and<br />

antibodies directed against specific molecules<br />

including a molecule known as β2-glycoprotein-1.<br />

The APS can occur in patients with SLE or as an<br />

isolated disorder. There are two main classifications<br />

of the APS: if there is an underlying autoimmune<br />

disorder, such as SLE, the patient is said to<br />

have secondary APS; if the patient has no known<br />

underlying autoimmune disorder, it is termed<br />

primary APS. Of the APS patients, over half of<br />

them have the primary form. In persons with SLE,<br />

around 30% will develop the APS. In general, anticardiolipin<br />

antibodies occur approximately five<br />

times more often than the lupus anticoagulant in<br />

patients with the APS. Approximately 10% of<br />

patients with an initial presentation of primary<br />

APS will eventually be diagnosed with an autoimmune<br />

disorder such as SLE or a mixed connective<br />

tissue disorder. The APS represents a hypercoagulable<br />

state, which can be associated with a variety<br />

of clinical features including arterial and venous<br />

thrombosis, livedo reticularis and spontaneous<br />

abortions. The exact mechanism by which the<br />

antiphospholipid antibodies and anticardiolipin<br />

antibodies induce the thrombophilic state is not<br />

known. Gastrointestinal manifestations of APS are<br />

due to ischemia involving any part of the intestinal<br />

tube, and resulting in bleeding, abdominal pain,<br />

acute abdomen, esophageal necrosis with perforation,<br />

or giant gastric ulceration. 37,38<br />

Juvenile dermatomyositis and juvenile polymyositis<br />

These are uncommon disorders. Juvenile dermatomyositis<br />

has an incidence of 2–3 per 100000 per<br />

Specific disorders 431<br />

year. The clinical manifestations resemble those of<br />

adult dermatomyositis and polymyositis and<br />

include symmetric proximal muscle weakness,<br />

myalgias, muscle tenderness, fever and rash (the<br />

latter commonly detected in juvenile dermatomyositis).<br />

In addition, calcinosis is seen in onethird<br />

of patients. Functional outcomes have<br />

become good with modern treatments, but the<br />

diseases remain chronic in a large number of children<br />

and sequelae are often seen. Affected children<br />

typically present prodromic non-specific<br />

signs such as malaise, muscle aching, fever and<br />

irritability that are often interpreted as a viral<br />

illness. A change in gait may be a clue to muscle<br />

disease at this stage. 39 Gastrointestinal tract<br />

involvement can also occur, and includes mucosal<br />

ulcerations due to vascular occlusion. Affected<br />

patients may present with abdominal pain,<br />

gastrointestinal bleeding, or perforation. 39 In a<br />

recently revised series of 25 patients aged between<br />

1.5 and 15 years, dysphagia and abdominal pain<br />

were present in 32% and 40% of cases, respectively.<br />

40 A case of atonic esophagus and gastroparesis<br />

has been described in a child with juvenile<br />

dermatomyositis. 41 Juvenile dermatomyositis can<br />

usually be diagnosed on a clinical basis in the<br />

child with a characteristic rash and proximal<br />

muscle weakness. The diagnosis of juvenile<br />

dermatomyositis can be more difficult with the<br />

lack of cutaneous manifestations. Certain laboratory<br />

features such as elevated muscle enzyme<br />

levels, increase in von Willebrand’s factor 8<br />

antigen (vWF) and autoantibodies are frequently<br />

present in both disorders: they provide confirmatory<br />

evidence of the disease and a means of following<br />

patients. Electromyography can assist in<br />

making the diagnosis in equivocal cases (e.g. those<br />

without a typical rash or with normal muscle<br />

enzyme levels). Muscle biopsy may also be necessary<br />

in these instances, and is often the definitive<br />

test. The mainstays of therapy are corticosteroids;<br />

some children require additional immunosuppressive<br />

agents because of corticosteroid resistance or<br />

intolerance.<br />

Rheumatoid vasculitis<br />

A small-vessel vasculitis can complicate the<br />

course of rheumatoid arthritis. Small arteries in<br />

the peripheral organs are usually affected, resulting<br />

in gastrointestinal vasculitis in up to 10% of


432<br />

Vasculitides<br />

the patients with rheumatoid arthritis. 42 In<br />

contrast to PAN, vasculitis in rheumatoid arthritis<br />

shows few signs of inflammation, but intensive<br />

intimal proliferation and vascular occlusion. 43 The<br />

gastrointestinal symptoms of rheumatoid vasculitis<br />

range from slight recurrent abdominal pain to<br />

severe bleeding, intestinal ischemia and necrosis. 4<br />

It should be observed that, in rheumatoid arthritis,<br />

abdominal pain is not necessarily due to vasculitis,<br />

but can arise from mucosal lesions such as<br />

ulcers or bleeding, as a consequence of the administration<br />

of anti-inflammatory drugs.<br />

Thromboangiitis obliterans (Winiwarter–Buerger’s<br />

disease)<br />

A non-atherosclerotic, segmental, inflammatory<br />

disorder, Winiwarter–Buerger’s disease most<br />

commonly affects the small and medium-sized<br />

arteries, veins and nerves of the extremities. 4 This<br />

condition is distinguished from other forms of<br />

vasculitis by the highly cellular and inflammatory<br />

thrombus with relative sparing of the blood vessel<br />

wall. ESR and serum C-reactive protein are usually<br />

normal; commonly measured autoantibodies (e.g.<br />

antinuclear antibody and rheumatoid factor) are<br />

normal, as are other serological tests such as circulating<br />

immune complexes, complement levels and<br />

cryoglobulins, despite an immune reaction in the<br />

arterial intima. Winiwarter–Buerger’s disease<br />

usually begins with ischemia of the distal small<br />

arteries and veins, followed by more proximal arterial<br />

involvement as the disease advances. Patients<br />

usually note ischemia of the digits, which may<br />

manifest as claudication of the feet, legs, hands or<br />

arms. 44 The disease can progress to include<br />

ischemic ulcerations of the fingers or toes, with<br />

accompanying ischemic pain at rest. Buerger’s<br />

disease should be suspected in passive or active<br />

smokers who present with distal ischemia of the<br />

hands and/or feet. Obliteration of the intestinal<br />

vasculature can occur with abdominal angina,<br />

resulting in bowel infarction that requires immediate<br />

surgery. 45,46 A definitive diagnosis is made<br />

when the histopathology identifies the acute phase<br />

lesion in a patient with a clinical history of<br />

smoking. Typical arteriographic findings help to<br />

exclude proximal atherosclerotic disease and to<br />

meet the criteria for thromboangiitis obliterans.<br />

Treatment and follow-up<br />

Childhood vasculitides, particularly Kawasaki<br />

disease and PAN, require prompt recognition,<br />

because they can be life-threatening in the absence<br />

of appropriate therapy. The diagnosis can be laborious<br />

and cumbersome, since the clinical features<br />

of the various disorders overlap. Furthermore,<br />

although the disorders usually have a characteristic<br />

pattern of organ involvement, each organ<br />

system can be affected. Available treatments are<br />

helpful, particularly in the acute phase. When<br />

chronic therapy is required, the adverse effects of<br />

drugs and secondary infections are clinically relevant.<br />

Mortality data suggest that early deaths in<br />

vasculitides are due to active disease, whereas late<br />

deaths mainly result from the complications of<br />

therapy. The treatment of vasculitides depends<br />

upon the nature and severity of the disorder, and<br />

may include antihistamines, corticosteroids, nonsteroidal<br />

anti-inflammatory drugs, or cytotoxic<br />

agents. Patients with systemic vasculitis usually<br />

need at least corticosteroid therapy. As an<br />

example, glucocorticoid therapy is used for the<br />

treatment of vasculitis accompanying connective<br />

tissue disease. In most patients, the dose can be<br />

reduced and gradually discontinued, but some<br />

patients require chronic long-term administration<br />

of low-dose corticosteroids. Patients with rapidly<br />

progressive vasculitic diseases, such as PAN, are<br />

likely to require therapy consisting of a combination<br />

of a cytotoxic drug (usually cyclophosphamide)<br />

and corticosteroids. Both oral and pulse<br />

cyclophosphamide are used, depending upon the<br />

physician’s experience and the disease severity.<br />

After 1–2 months of combined therapy, the corticosteroid<br />

dose may be reduced if symptoms<br />

improve; therapy is usually continued for 6–12<br />

months to decrease the risk of relapse.<br />

Azathioprine and methotrexate have been used in<br />

less severe forms of vasculitides and as maintenance<br />

therapy, after remission has been induced<br />

by cyclophosphamide. Corticosteroid therapy,<br />

even in low doses, can produce substantial toxicity.<br />

The risk is greater as the dose increases, with<br />

complications ranging from minor skin bruising to<br />

life-threatening infections and fractures. Some of<br />

these complications can be prevented with appropriate<br />

patient monitoring. Some manifestations of


vasculitis such as neuropathy can take months to<br />

recede, even if the underlying vasculitis has<br />

abated, whereas others may be permanent.<br />

Patients in whom a chronic course is expected may<br />

become discouraged about their lack of rapid<br />

improvement and possible poor outcome. These<br />

patients may benefit from extra support and<br />

encouragement. Once the treatment has been initiated,<br />

patient monitoring is based on most of the<br />

procedures used during the diagnostic approach.<br />

REFERENCES<br />

1. Langford CA. Vasculitis. J Allergy Clin Immunol 2003;<br />

111 (2 Suppl): S602–S612.<br />

2. Jennette JC, Falk RJ. Do vasculitis categorization systems<br />

really matter? Curr Rheumatol Rep 2000; 2: 430–438.<br />

3. Yalcindag A, Sundel R. Vasculitis in childhood Curr<br />

Opin Rheumatol 2001; 13: 422–427.<br />

4. Müller-Ladner U. Vasculitides of the gastrointestinal<br />

tract. Best Pract Res Clin Gastroenterol 2001; 15: 59–82.<br />

5. Gardner-Medwin JM, Dolezalova P, Cummins C et al.<br />

Incidence of Henoch–Schönlein purpura, Kawasaki<br />

disease, and rare vasculitides in children of different<br />

ethnic origins. Lancet 2002; 360: 1197–1202.<br />

6. Watts RA, Scott DGI. Epidemiology of the vasculitides.<br />

Curr Opin Rheumatol 2003; 15: 11–16.<br />

7. Weyand CM, Goronzy JJ. Multisystem interactions in<br />

the pathogenesis of vasculitis. Curr Opin Rheumatol<br />

1997; 9: 3–11.<br />

8. Guillevin L, Du LT, Godeau P et al. Clinical findings and<br />

prognosis of polyarteritis nodosa and Churg–Strauss<br />

angiitis: a study of 165 patients. Br J Rheumatol 1988;<br />

27: 258–264.<br />

9. Laghi A, Borrelli O, Paolantonio P et al. Contrast<br />

enhanced magnetic resonance imaging of the terminal<br />

ileum in children with Crohn’s disease. Gut 2003; 52:<br />

393–397.<br />

10. Koutkia P, Mylonakis E, Rounds S et al. Leucocytoclastic<br />

vasculitis: an update for the clinician. Scand J<br />

Rheumatol 2001; 30: 315–322.<br />

11. Katayama H, Shimizu T, Tanaka Y et al. Threedimensional<br />

magnetic resonance angiography of<br />

vascular lesions in children. Heart Vessels 2000; 15: 1–6.<br />

12. Ozen S. The spectrum of vasculitis in children. Best<br />

Pract Res Clin Rheumatol 2002; 16: 411–425.<br />

13. Guillevin L, Pagnoux C. When should immunosuppressants<br />

be prescribed to treat systemic vasculitides. Intern<br />

Med 2003; 42: 313–317.<br />

14. Noth I, Strek ME, Leff AR. Churg–Strauss syndrome.<br />

Lancet 2003; 361: 587–594.<br />

15. Lin TL, Wang CR, Liu MF et al. Multiple colonic ulcers<br />

caused by Churg–Strauss syndrome in a 15-year-old<br />

girl. Clin Rheumatol 2001; 20: 362–364.<br />

16. Sokol RJ, Farrell MK, McAdams AJ. An unusual<br />

presentation of Wegener’s granulomatosis mimicking<br />

inflammatory bowel disease. Gastroenterology 1984; 87:<br />

426–432.<br />

References 433<br />

There are two main long-term concerns: most<br />

forms of vasculitis (except for drug-induced hypersensitivity<br />

vasculitis) can relapse; and vascular<br />

injury during the acute phase can heal with scarring<br />

and narrowing of the affected vessels, leading<br />

to signs of ischemia that do not reflect recurrent<br />

active disease. 47 Therefore, prolonged monitoring<br />

is indicated, even though it may be difficult to<br />

determine whether a change in findings results<br />

from the disease, the medications, or scarring.<br />

17. Bang D. Clinical spectrum of Behçet’s disease. J<br />

Dermatol 2001; 28: 610–613.<br />

18. Terrin G, Borrelli O, Di Nardo G et al. A child with<br />

aphthae and diarrhoea. Lancet 2002; 359: 316.<br />

19. Geboes K. Crohn’s disease, ulcerative colitis or<br />

indeterminate colitis – how important is it to<br />

differentiate? Acta Gastroenterol Belg 2001; 64: 197–200.<br />

20. Rozenbaum M, Rosner I, Portnoy E. Current therapy for<br />

Behçet’s disease. Am J Ther 2002; 9: 465–470.<br />

21. Lupi-Herrera E, Sanchez-Torres G, Marcushamer J et al.<br />

Takayasu’s arteritis. Clinical study of 107 cases. Am<br />

Heart J 1997; 93: 94–103.<br />

22. Johnston SL, Lock RJ, Gompels MM, Takayasu arteritis:<br />

a review. J Clin Pathol 2002; 55: 481–486.<br />

23. Sakhuja V, Gupta KL, Bhasin DK et al. Takayasu’s<br />

arteritis associated with idiopathic ulcerative colitis.<br />

Gut 1990; 31: 831–833.<br />

24. Westphal J, Lobbecke F, Mietens C. Arteritis of the large<br />

vessels originating at the aorta (Takayasu’s arteritis) in<br />

childhood. Klin Padiatr 1976; 188: 570–577.<br />

25. Salvarani C, Cantini F, Boiardi L et al. Polymyalgia<br />

rheumatica and giant-cell arteritis. N Engl J Med 2002;<br />

347: 261–271.<br />

26. De Bayser L, Roblot P, Ramassamy A et al. Hepatic<br />

fibrin-ring granulomas in giant cell arteritis.<br />

Gastroenterology 1993; 105: 272–273.<br />

27. Mele T, Evans M. Intestinal obstruction as a<br />

complication of Kawasaki disease. J Pediatr Surg 1996;<br />

31: 985–986.<br />

28. Beiler HA, Schmidt KG, von Herbay A et al. Ischemic<br />

small bowel strictures in a case of incomplete Kawasaki<br />

disease. J Pediatr Surg 2001; 36: 648–650.<br />

29. Saulsbury FT. Epidemiology of Henoch–Schönlein<br />

purpura. Cleve Clin J Med 2002; 69 (Suppl 2): SII87–89.<br />

30. Esaki M, Matsumoto T, Nakamura S et al. GI involvement<br />

in Henoch–Schönlein purpura. Gastrointest<br />

Endosc 2002; 56: 920–923.<br />

31. Dillon MJ. Henoch–Schönlein purpura (treatment and<br />

outcome). Cleve Clin J Med 2002; 69 (Suppl 2):<br />

SII121–123.<br />

32. Hattori M, Ito K, Konomoto T et al. Plasmapheresis as<br />

the sole therapy for rapidly progressive<br />

Henoch–Schönlein purpura nephritis in children. Am J<br />

Kidney Dis 1999; 33: 427–433.


434<br />

Vasculitides<br />

33. Zizic TM, Classen JN, Stevens MB. Acute abdominal<br />

complications of systemic lupus erythematosus and<br />

polyarteritis nodosa. Am J Med 1982; 73: 525–31.<br />

34. Yuasa S, Suwa A, Hirakata M et al. A case of lupus<br />

erythematosus presenting with rectal ulcers as the<br />

clinical manifestation of disease. Clin Exp Rheumatol<br />

2002; 20: 407–410.<br />

35. Northcott KA, Yoshida EM, Streinbrecher UP. Primary<br />

protein-losing enteropathy in anti-double-stranded DNA<br />

disease: the initial and sole clinical manifestation of<br />

occult systemic lupus erythematosus? Clin Gastroenterol<br />

2001; 33: 340–341.<br />

36. Perlemuter G, Chaussade S, Wechsler B et al. Chronic<br />

intestinal pseudo-obstruction in systemic lupus erythematosus.<br />

Gut 1998; 43: 117–122.<br />

37. Love PE, Santoro SA. Antiphospholipid antibodies:<br />

anticardiolipin and the lupus anticoagulant in systemic<br />

lupus erythematosus (SLE) and non-SLE disorders. Ann<br />

Intern Med 1990; 112: 682–698.<br />

38. Mihas AA. Gastrointestinal bleeding and intestinal<br />

ischemia associated with anticardiolipin antibodies. Dig<br />

Dis Sci 1995; 40: 1039–1040.<br />

39. Ramanan AV, Feldman BM. Clinical outcomes in<br />

juvenile dermatomyositis. Curr Opin Rheumatol 2002;<br />

14: 658–662.<br />

40. Shehata R, Al-Mayouf S, Al-Dalaan A et al. Juvenile<br />

dermatomyositis: clinical profile and disease course in<br />

25 patients. Clin Exp Rheumatol 1999; 17: 115–118.<br />

41. Laskin BL, Choyke P, Keenan et al. Novel gastrointestinal<br />

tract manifestations in juvenile dermatomyositis.<br />

J Pediatr 1999; 135: 371–374.<br />

42. Babian M, Nesef Sand Soloway G. Gastrointestinal<br />

infarction as a manifestation of rheumatoid vasculitis.<br />

Am J Gastroenterol 1998; 93: 119–120.<br />

43. Achkar AA, Stanson AW, Johnson CM et al. Rheumatoid<br />

vasculitis manifesting as intra-abdominal hemorrhage.<br />

Mayo Clin Proc 1995; 70: 565–569.<br />

44. Olin, JW, Thromboangiitis obliterans (Buerger’s disease).<br />

N Engl J Med 2000; 343: 864–869.<br />

45. Adem C, Benamouzig R, Royer I et al. Buerger’s disease<br />

or thromboangiitis obliterans revealed by an enteric<br />

ischemia. Case report and literature review.<br />

Gastroenterol Clin Biol 2002; 26: 409–411.<br />

46. Schellong SM, Bernhards J, Ensslen F et al. Intestinal<br />

type of thrombangitis obliterans (Buerger’s disease). J<br />

Intern Med 1994; 235: 69–73.<br />

47. Gonzales-Gay MA, Garcia Porrua C. Systemic vasculitides.<br />

Best Pract Res Clin Rheumatol 2002; 16: 833–845.


27<br />

Introduction<br />

Celiac disease<br />

Stefano Guandalini<br />

Important advances have occurred in the past few<br />

years in our understanding of celiac disease, its<br />

pathogenesis, its manifestations, its complications<br />

and its treatment. This fascinating condition is<br />

now regarded as a true autoimmune disorder, triggered<br />

by a well-known autoantigen, and affecting<br />

primarily the small intestine, where it progressively<br />

leads to severe villous blunting. However, it<br />

has clinical implications that reach far beyond the<br />

gut.<br />

Celiac disease occurs only in genetically susceptible<br />

individuals, and is triggered by the ingestion of<br />

gliadins contained in wheat, rye and barley. Wheat<br />

gluten and similar alcohol-soluble proteins in<br />

other grains (called prolamines) cause – when<br />

ingested – the intestinal damage in individuals<br />

who are genetically predisposed. Indeed, almost<br />

100% of celiacs are positive for either HLA DR3 (or<br />

DR5/DR7), or HLA DR4. 1 Although gluten is<br />

clearly the major environmental factor inducing<br />

celiac disease, other less known agents may be at<br />

play in modulating its onset, including the amount<br />

and type of gluten, and the utilization and duration<br />

of breast feeding. 2,3 An intriguing relationship<br />

between birth time and prevalence of celiac<br />

disease also suggests the influence of other environmental<br />

factors, such as infections. 4<br />

Epidemiology<br />

The estimated prevalence of celiac disease varies<br />

according to the method of defining the condition.<br />

Earlier investigations that included only florid<br />

cases of celiac disease with overt gastrointestinal<br />

manifestations found an overall low but highly<br />

variable incidence throughout several European<br />

countries. With the availability of tests for malab-<br />

sorption and of the pediatric peroral biopsy techniques,<br />

an increased incidence of celiac disease, to<br />

about 1:500, was reported in studies in Europe.<br />

Subsequently, sensitive serological tests have<br />

made it possible also to detect minimally symptomatic<br />

or even asymptomatic cases with the typical<br />

mucosal changes (thus fulfilling the definition of<br />

celiac disease). When these types of tool, and in<br />

particular the anti-endomysium antibodies, are<br />

utilized for screening studies, the prevalence of<br />

celiac disease soars to the current estimates of<br />

approximately 1:130 to 1:300 of the general<br />

European population. 5–8<br />

Until recently it was believed that celiac disease<br />

was a rare disorder in the USA, and rates of prevalence<br />

of lower than 1:10000 were often quoted.<br />

However, a recent multicenter investigation<br />

throughout the USA that screened more than<br />

13000 individuals with anti-endomysium antibodies<br />

and/or human tissue transglutaminase antibodies,<br />

found a prevalence of 1:133, 9 i.e. identical<br />

to that found in Europe. It is clear, however, that<br />

the current rates of diagnosis of celiac disease in<br />

the USA are well below those in Europe, perhaps<br />

owing to an overall later onset of the disease and<br />

more frequent extraintestinal manifestations. This,<br />

in turn, may be linked to various factors, with the<br />

more prolonged and more widespread use of breast<br />

feeding being a likely cause of postponing the<br />

onset of celiac disease. 2 The prevalence of celiac<br />

disease in other areas of the world has been less<br />

studied. Data are, however, available from Latin<br />

America, North Africa, the Near and Middle East<br />

and Northwest India: in all these areas celiac<br />

disease has been reported, and where prevalence<br />

data were sought, they do not differ significantly<br />

from those indicated above. In some ethnicities,<br />

such as in the Saharawi population, celiac disease<br />

has been found in as many as 5% of the general<br />

population. 10 Thus, it is fair to assume that celiac<br />

435


436<br />

Celiac disease<br />

disease constitutes one of the most common genetically<br />

induced chronic diseases. However, celiac<br />

disease is considered extremely rare or non-existing<br />

in people with African, Chinese or Japanese<br />

descent.<br />

Pathophysiology<br />

Celiac disease is an autoimmune disorder. The<br />

initial event in the pathogenesis of the celiac<br />

lesion is thought to be abnormal permeability,<br />

allowing the entry of gliadin peptides not entirely<br />

degraded by the intraluminal and brush-border<br />

bound-peptidases. Interestingly, the initial ‘theory’<br />

on celiac disease pathogenesis was the enzymatic<br />

one: for many years, research focused on ‘the<br />

missing enzyme’, unable properly to digest gluten<br />

and thus creating an indigestible, toxic fragment.<br />

However, there is no missing peptidase in celiac<br />

disease; simply, the most toxic amongst the fractions<br />

of gliadin that have been shown to be<br />

harmful to the celiac mucosa are remarkably resistant<br />

to digestion by gastric, pancreatic and<br />

mucosa-associated enzymes. 11 Under normal<br />

circumstances, the intestinal epithelium would act<br />

as a barrier to the passage of such macromolecules,<br />

but in celiac disease, a well-documented loosening<br />

of the intestinal tight junctions, 12 possibly even<br />

triggered by gliadin itself, 13 leads to increased<br />

permeability to macromolecules. The role of<br />

altered expression of zonulin, a novel human<br />

protein that induces tight junction disassembly<br />

and a subsequent increase in intestinal permeability,<br />

has been suggested. Zonulin expression was<br />

found to be elevated in intestinal tissues from<br />

patients with florid celiac disease. 14<br />

There are two pathways involved in the pathogenesis<br />

of celiac disease: an early one, involving<br />

mainly the innate immune system; and a subsequent<br />

one, involving T cells. Soon after reaching<br />

the serosal side of the intestinal epithelium, the<br />

toxic gliadin peptides elicit an early response that<br />

causes crucial modifications of the mucosal<br />

microenvironment that precede and prime the<br />

subsequent involvement of the pathogenic T cells.<br />

A recent elegant study by Maiuri et al 15 brings<br />

important new information on this early response,<br />

by showing that a non-immunodominant gliadin<br />

fragment can activate the innate immune system,<br />

affecting the in situ T-cell recognition of dominant<br />

gliadin epitopes. Although more than 50 epitopes<br />

have been identified, the dominant α-gliadin T-cell<br />

epitope appears to be a single tissue transglutaminase-modified<br />

peptide. 16<br />

During the first phase, there is a marked increase<br />

of HLA-DR expression on both the epithelium and<br />

the adjacent lamina propria macrophages; this is<br />

followed by overexpression of intercellular adhesion<br />

molecule 1 (ICAM-1). Finally, CD8 + T cells<br />

invaded epithelial cells (intraepithelial lymphocytes).<br />

About 95% of all celiac patients belong to the DR3<br />

(or DR5/DR7 heterozygous) genotype and express<br />

the DQ2 α,β-heterodimer, encoded by DQA1*0501/<br />

DQB1*0201, while almost all of the remaining 5%<br />

are DR4 and show the DQ8 α,β-heterodimer, encoded<br />

by DQA1*0301/DQB1*0302. This strong<br />

association implies that the adaptive branch of the<br />

immune system, and in particular CD4 + T<br />

lymphocytes, must play a crucial role in the pathogenesis.<br />

In fact, DQ2 and DQ8 molecules are<br />

located on the surface of antigen-presenting cells<br />

and bind peptides to be presented to CD4 + T<br />

lymphocytes. The DQ2 or DQ8 molecules<br />

expressed by the antigen-presenting cells (mostly<br />

macrophages) possess unique peptide-binding<br />

properties. They typically bind peptides of 12–20<br />

amino acid residues, with a core region of nine<br />

amino acids being bound to the peptide-binding<br />

groove on the HLA class-II molecule via interactions<br />

between side chains of amino acids and<br />

pockets of the binding groove. The autoantigen in<br />

celiac disease, a major target of all autoantibodies<br />

such as anti-endomysium and anti-reticulin, has<br />

been found to be the ubiquitous enzyme tissue<br />

transglutaminase. This enzyme, among other functions,<br />

selectively converts glutamine residues<br />

within gluten to glutamic acid. Such deamidation<br />

increases the negative charges on the gliadin<br />

peptides, 17 thus resulting in a strong enhancement<br />

of DQ binding and T-cell recognition.<br />

From a morphological point of view, small-bowel<br />

mucosal damage occurs as a result of gradual<br />

changes from normal mucosa to overt mucosal<br />

atrophy with crypt hyperplasia. It is crucial to<br />

understand that these changes occur in a progressive<br />

manner, so that the ‘flat’ mucosa, for a long<br />

time thought to be the necessary key finding in<br />

celiac disease, is only the last stage in a continuum<br />

of morphological changes. At lest three degrees of


change are recognized, as classically described by<br />

Marsh. 18 The infiltrative (type 1) lesion, seen in<br />

the latent phase, is characterized by morphologically<br />

normal mucosa and is not usually associated<br />

with gastrointestinal symptoms. Initial changes<br />

seen include an increase in the number of intraepithelial<br />

lymphocytes, followed by infiltration of<br />

the lamina propria with plasma cells and lymphocytes.<br />

The hyperplastic (type 2) lesion is similar to<br />

type 1, but with elongation of crypts, owing to a<br />

marked increase in undifferentiated crypt cells.<br />

The destructive (type 3) lesion is synonymous<br />

with villous atrophy, the original lesion described<br />

in celiac disease.<br />

The mechanisms leading to such changes have<br />

been the matter of extensive research in past years,<br />

and are currently still largely obscure. Several<br />

hypotheses coexist, each supported by experimental<br />

evidence. The ultimate agent responsible for<br />

Pathophysiology 437<br />

villous flattening appears to be the enterocyte<br />

expression of FAS (the prototype of the ‘death<br />

receptor’) and apoptosis. Indeed, the element that<br />

differentiates patchy lesions from areas with<br />

normal histology is the specific lack of expression<br />

of FAS in morphologically normal mucosa. 19 It is<br />

also thought 20 that interleukin (IL)-15 may be<br />

involved in both the proliferating and the atrophic<br />

epithelial phases of celiac disease. In an organ<br />

culture model, IL-15 was in fact able to induce<br />

proliferation in crypts and FAS expression in enterocytes.<br />

In the active phase of celiac disease, a<br />

specific increase of intraepithelial lymphocytes<br />

expressing CD94 has been demonstrated, 21 possibly<br />

induced by IL-15. In essence, it would appear<br />

that intraepithelial lymphocytes migrate into the<br />

epithelium to induce apoptosis of the enterocytes.<br />

A scheme of the main events currently thought to<br />

occur in the mucosa of celiac patients is shown in<br />

Figure 27.1.<br />

Figure 27.1 Scheme of the epithelial damage in celiac disease. After gluten is digested into gliadin peptides, they enter<br />

through an abnormally increased intestinal permeability and reach the serosal side, where they interact with tissue transglutaminase<br />

(tTG). Among several effects of this interaction is the deamidation of such peptides, a process that strongly<br />

enhances their affinity for the HLA-DQ2 or DQ8 heterodimer expressed at the surface of the antigen-presenting cell (APC).<br />

The peptide is thus presented to the T lymphocyte. This initiates a chain of events, including release of toxic lymphokines<br />

such as interleukin (IL)-2 and IL-15 that have a direct damaging effect on the epithelium. Interaction with B lymphocytes<br />

also leads (by unclear mechanisms) to the production of antibodies against the enzyme tTG (utilized in the diagnostic<br />

process). The reduced activation of the epithelium-protective peptide transforming growth factor (TGF)-β further contributes<br />

to the damage of the epithelium.


438<br />

Celiac disease<br />

Clinical presentations<br />

One of the most important advances in the past<br />

decade has been the understanding that, although<br />

characterized by intestinal damage, celiac disease<br />

may present with signs and symptoms not necessarily<br />

related to the gastrointestinal tract.<br />

Moreover, it may well remain asymptomatic, or<br />

oligosymptomatic, for many years or indeed for<br />

life.<br />

Thus, we have come to distinguish four forms of<br />

celiac disease:<br />

(1) Typical;<br />

(2) Atypical (or more exactly ‘extraintestinal’);<br />

(3) Silent;<br />

(4) Latent.<br />

Table 27.1 reports the corresponding definitions,<br />

and Figure 27.2 illustrates the so-called ‘celiac<br />

iceberg’ that depicts the relation between HLA<br />

status, duodenal morphology and clinical expression<br />

in these different forms. Interestingly, there is<br />

evidence from many epidemiological investigations<br />

that the less clinically evident conditions<br />

(silent, latent and oligosymptomatic forms) are<br />

much more common that the clinically overt ones.<br />

Genetic<br />

susceptibility<br />

DR3-DQ2<br />

DR5/7-DQ2<br />

DR4-DQ8<br />

Clinically overt celiac disease<br />

Silent celiac disease<br />

Potential celiac disease<br />

Healthy individuals<br />

Figure 27.2 The ‘celiac iceberg’. See text for details.<br />

Table 27.1 Definitions of the various forms<br />

of celiac disease<br />

Typical<br />

Gastrointestinal signs/symptoms predominate:<br />

diarrhea<br />

vomiting<br />

failure to thrive<br />

anorexia<br />

constipation<br />

recurrent abdominal pain<br />

Atypical or extraintestinal<br />

Gastrointestinal signs/symptoms are minimal or<br />

absent. Most common signs/symptoms are:<br />

fatigue<br />

malaise<br />

anemia<br />

Silent<br />

No signs/symptoms. Gluten-dependent duodenal<br />

mucosal changes consistent with celiac disease<br />

Latent<br />

No signs/symptoms. Duodenal mucosa normal.<br />

Gluten-dependent changes with or without<br />

symptoms to appear later in time<br />

Manifest<br />

mucosal<br />

lesion<br />

Normal<br />

mucosa


It should be noted that the factors ultimately<br />

responsible in any single individual for the expressivity<br />

of the disease (i.e. developing typical vs.<br />

extraintestinal vs. silent, etc.) are not known. It is,<br />

however, known that age influences the prevalence<br />

of the various forms, as schematically indicated<br />

in Figure 27.3.<br />

The typical form of celiac disease presents with<br />

gastrointestinal symptoms typically between 6 and<br />

24 months of age. Symptoms begin at various time<br />

intervals after the introduction of gluten in the<br />

diet, also depending on its amount and on other<br />

environmental factors such as the concomitant<br />

presence of breast feeding, occurrence of viral<br />

enteritis, etc. Affected infants have poor appetite,<br />

chronic diarrhea, abdominal distension, muscle<br />

wasting and failure to thrive. Vomiting is also<br />

common. As spontaneous avoidance of glutencontaining<br />

foods does not occur, symptoms may<br />

progress to serious malnutrition, even cachexia, if<br />

diagnosis is delayed. Behavioral changes are also<br />

often found: celiac infants and young children<br />

become less talkative, more irritable, avoid<br />

company and are sad. In the most severely affected<br />

infants, the clinical picture of the so-called ‘celiac<br />

crisis’ may develop. This acute syndrome, now<br />

rarely seen, is characterized by explosive watery<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Atypical or extraintestinal celiac disease 439<br />

diarrhea often followed by no passage of stools,<br />

impressive abdominal distension, dehydration,<br />

hypotension and lethargy. This severe picture is<br />

accompanied by profound electrolyte abnormalities<br />

including dangerously low potassium levels.<br />

Atypical or extraintestinal celiac<br />

disease<br />

In recent years an increasing number of patients<br />

have been diagnosed without typical gastrointestinal<br />

manifestations at an older age. 22,23 It is<br />

currently believed that almost 50% of patients<br />

with newly diagnosed celiac disease, and up to<br />

two-thirds of the adults, do not present with<br />

gastrointestinal symptoms and have either atypical<br />

or silent celiac disease. Table 27.2 reports the main<br />

presentations of ‘atypical’ or ‘extraintestinal’ celiac<br />

disease.<br />

Dermatitis herpetiformis<br />

1–2 2–5 5–12 12–18 >18<br />

Age (years)<br />

This variant of celiac disease presents with a<br />

blistering skin rash involving elbows, knees and<br />

buttocks associated with dermal granular<br />

Diarrhea<br />

Vomiting<br />

Failure to<br />

thrive;<br />

constipation<br />

Irritability<br />

Short stature<br />

Anemia<br />

Fatigue<br />

Minor GI symptoms<br />

Others<br />

Figure 27.3 Celiac disease presentations vary with age. Most gastrointestinal (GI) signs and symptoms are more common<br />

in early years, while anemia, fatigue and minor GI symptoms predominate later in life.


440<br />

Celiac disease<br />

Table 27.2 Main presentations of so-called<br />

‘atypical’ (or ‘extraintestinal’) celiac disease<br />

Dermatitis herpetiformis<br />

Permanent enamel hypoplasia<br />

Iron-deficient anemia resistant to oral iron intake<br />

Short stature, delayed puberty<br />

Chronic hepatitis with hypertransaminasemia<br />

Primary biliary cirrhosis<br />

Arthritis<br />

Osteopenia/osteoporosis<br />

Epilepsy with occipital calcifications<br />

Primary ataxia, white-matter focal lesions<br />

Psychiatric disorders<br />

Infertility of women<br />

immunoglobulin (Ig) A deposits. Rash as well as<br />

mucosal morphology improve on a gluten-free<br />

diet. 24 Previously considered a skin disease occurring<br />

often concomitantly with celiac disease, but<br />

overall a rare occurrence, it is now thought to be a<br />

common although often unrecognized problem in<br />

celiac patients. 25<br />

Dental enamel hypoplasia<br />

Dental enamel defects may be the only presenting<br />

manifestation of celiac disease. 26 These patients<br />

may have no or minimal gastrointestinal symptoms.<br />

Iron-deficiency anemia<br />

Iron-deficiency anemia, resistant to oral iron<br />

supplementation, has been found to be the most<br />

common extraintestinal manifestation of celiac<br />

disease in some studies, and often its primary clinical<br />

manifestation. 23,27 In one study 5% of all<br />

patients with anemia had celiac disease, and the<br />

prevalence rose to 8.5% when only patients with<br />

microcytic anemia resistant to iron therapy were<br />

considered. 28 Recently, screening with serology for<br />

celiac disease a large number of adult patients who<br />

were found to have either folate or iron deficiency<br />

(in the absence of gastrointestinal manifestations)<br />

detected 11% positive. 29 In spite of these well-<br />

known data, general screening of this type of<br />

patient is still rarely conducted.<br />

Short stature and delayed puberty<br />

Short stature may be the only manifestation of<br />

celiac disease. 30 As many as 8–10% of children<br />

with ‘idiopathic’ short stature may have celiac<br />

disease that can be detected on serologic testing. 31<br />

Adolescents with untreated celiac disease may<br />

have delayed onset of menarche. 32<br />

Chronic hepatitis and hypertransaminasemia<br />

Elevated transaminase levels are a frequent finding<br />

in untreated patients with celiac disease. In the<br />

majority of cases liver enzymes normalize on a<br />

gluten-free diet. 33 As many as 9% of patients with<br />

elevated transaminase levels of unclear etiology<br />

may have silent celiac disease. 34 Liver biopsies in<br />

these patients showed non-specific reactive<br />

hepatitis. Transaminases returned to normal on a<br />

gluten-free diet. Very recently, celiac disease has<br />

been described as causing severe liver disease,<br />

indeed hepatic failure, that was further shown to<br />

be responsive to a gluten-free diet. 35<br />

Arthritis and arthralgia<br />

Arthritis can be a common extraintestinal manifestation<br />

in adults with celiac disease including<br />

those on a gluten-free diet. 36 Of children with juvenile<br />

chronic arthritis, 2–3% may have celiac<br />

disease. 37<br />

Osteopenia/osteoporosis<br />

Patients with celiac disease are at high risk for<br />

developing low bone mineral density and osteoporosis.<br />

38,39 This has been found even for asymptomatic<br />

celiac disease patients detected at screening,<br />

40 whose reduced bone mineral density<br />

improved on a gluten-free diet. As a result of this<br />

condition, celiac disease patients are known to<br />

have an increased incidence of fractures. 41 The<br />

British Society of Gastroenterology recommended<br />

measuring bone mineral density in all patients<br />

with celiac disease. 42 Recently however, a survey<br />

conducted in the UK in 244 adult patients and 169


controls showed no statistically increased prevalence<br />

of fractures, and concluded that screening<br />

for bone mineral density in all celiac patients may<br />

not be warranted. 43 Thus, this issue is still open.<br />

Bone mineral density improves in the majority of<br />

patients on a gluten-free diet. 44–46 The pathogenesis<br />

of osteoporosis is complex, and only partially<br />

understood. It seems clear that poor absorption of<br />

calcium and/or vitamin D is not the most important<br />

factor. Recently, some evidence has been<br />

provided that an autoimmune aggression of the<br />

bone matrix may take place in celiac disease. 47<br />

Neurological problems<br />

Celiac disease can be the underlying cause of idiopathic<br />

cerebellar ataxia. 48 Celiac disease may also<br />

be associated with occipital calcifications and<br />

intractable epilepsy. These patients can be resistant<br />

to anti-seizure medicines but can benefit from<br />

a gluten-free diet if started soon after the onset of<br />

seizures. 49 Interestingly, a new neurological<br />

presentation of celiac disease has recently been<br />

described: focal brain white-matter lesions leading<br />

to multiple neurological manifestations such as<br />

seizures, muscle hypotonia and mild ataxia. 50<br />

Psychiatric disorders<br />

Although in recent years a large number of behavioral<br />

problems and disorders, such as autism,<br />

attention deficit hyperactivity disorder, etc., have<br />

been thought to be caused by celiac disease, there<br />

is no evidence to date that this is the case. 51 Celiac<br />

disease nevertheless can certainly present with<br />

Associated diseases 441<br />

some psychiatric disorders, such as depression 52<br />

and anxiety. 53,54 These conditions can be severe,<br />

but will usually respond to a gluten-free diet.<br />

Infertility<br />

Celiac disease may be responsible for unexplained<br />

infertility in women. 55 Ciacci et al 56 reported that<br />

the relative risk of abortion in women affected by<br />

celiac disease was 8.9 times higher than in healthy<br />

subjects, and that a gluten-free diet reduced the<br />

relative risk of abortion. Of interest, a recent study<br />

showed that not only fetuses from celiac mothers,<br />

but also those from celiac fathers suffered adverse<br />

effects in pregnancy, resulting in lower birth<br />

weight and perhaps shorter duration of pregnancy.<br />

57<br />

Associated diseases<br />

In addition to being responsible for various<br />

presentations, as described above, celiac disease is<br />

also known to be more commonly associated with<br />

a number of other diseases. Although the mechanisms<br />

of such links are still largely obscure, it is<br />

important to be aware of such associations in order<br />

to detect celiac disease as early as possible.<br />

Table 27.3 lists the main medical disorders that are<br />

associated with celiac disease. It is evident that<br />

many disorders are also of the autoimmune type.<br />

The association of celiac disease with autoimmune<br />

conditions is well established. 58 Recently, even<br />

autoimmune myocarditis was added to the list of<br />

such disorders. 59 An important multicenter study<br />

Table 27.3 Conditions associated with an increased prevalence of celiac disease<br />

Condition Approximate prevalence of celiac disease (%)<br />

Insulin-dependent diabetes mellitus 6%<br />

Thyroiditis 4%<br />

Sjögren’s syndrome and other connective tissue diseases 5%<br />

Primary biliary cirrhosis 3%<br />

Down’s syndrome 12%<br />

Williams’ syndrome 6%<br />

Turner’s syndrome 6%<br />

First-degree relatives of celiac patients 8–10%


442<br />

Celiac disease<br />

established a strong positive correlation between<br />

age at diagnosis of celiac disease and prevalence of<br />

autoimmune disorders such as type 1 diabetes,<br />

thyroiditis and alopecia, 60 thus suggesting that it is<br />

indeed the presence of celiac disease that acts as a<br />

trigger for the development of other autoimmune<br />

conditions. 61<br />

Type 1 (insulin-dependent) diabetes<br />

Up to 8% of patients with type-1 or insulin-dependent<br />

diabetes mellitus have been found to have<br />

typical features of celiac disease on duodenal<br />

biopsy. However, it is thought that the ‘real’ percentage<br />

is higher, as studies of such patients in serial<br />

screening over a period of years have documented<br />

that many individuals who initially had negative<br />

serological tests eventually developed positive tests<br />

and characteristic intestinal changes. 62 Typically<br />

(90% of cases), diagnosis of diabetes precedes by<br />

years that of celiac disease, most commonly<br />

presenting with mild to only moderate gastrointestinal<br />

symptoms. 63 As some of these symptoms are<br />

also seen in patients with diabetes (e.g. bloating or<br />

diarrhea), the diagnosis of celiac disease may be<br />

missed, unless screening is performed. Laboratory<br />

abnormalities such as anemia, and iron and folate<br />

deficiency are also common.<br />

It is still debated whether or not diabetic individuals<br />

found to be celiacs at screening (i.e. essentially<br />

asymptomatic) need to be put on a gluten-free diet.<br />

So far there is no convincing evidence that the diet<br />

has any obvious effect on diabetes. Indeed, the same<br />

considerations apply to all of the other associated<br />

autoimmune conditions. However, the following<br />

two considerations seem to suggest that adherence<br />

to a gluten-free diet should be recommended. First,<br />

a gluten-free diet has been shown in some cases to<br />

help improve glycemic control and improve<br />

gastrointestinal symptoms in patients with both<br />

conditions. Second, it can be argued that complications<br />

of untreated celiac disease known to be<br />

prevented by adherence to gluten-free diet (including<br />

osteopenia, infertility and malignancy – all<br />

documented occurrences in diabetic individuals)<br />

may be prevented by the diet in asymptomatic<br />

celiacs. As a consequence, the case for screening a<br />

type 1 diabetic for celiac disease seems well<br />

founded.<br />

Down’s syndrome<br />

Perhaps the best documented and most widely<br />

reported association of celiac disease with a nonautoimmune<br />

disorder is that with Down’s<br />

syndrome. The prevalence of Down’s syndrome in<br />

celiac disease, as assessed by screening methods,<br />

has been found to be between 5 and 12% in studies<br />

from both Europe 64–66 and more recently also<br />

North America. 9,67,68 Unlike patients with type-1<br />

diabetes, the majority with Down’s syndrome and<br />

celiac disease have other gastrointestinal symptoms,<br />

such as abdominal bloating, intermittent<br />

diarrhea, anorexia and failure to thrive. However,<br />

in a large multicenter study, it was found that<br />

about one-third of all Down’s syndrome patients<br />

with celiac disease had no gastrointestinal symptoms;<br />

66 the patients with celiac disease as a group,<br />

more commonly than their counterparts, had<br />

anemia, low serum iron and calcium, and a<br />

tendency to be mildly stunted in height and<br />

weight. 66 It seems highly desirable that patients<br />

with Down’s syndrome be screened for celiac<br />

disease and, whenever found positive, they should<br />

begin the gluten-free diet. As celiac disease may<br />

start at any age, patients with Down’s syndrome<br />

who test negative for serological markers (see<br />

below) would have to be re-tested again and again.<br />

To avoid this, since celiac disease occurs only in<br />

specific HLA haplotypes (see Epidemiology,<br />

p.435), an algorithm based on first determining the<br />

HLA haplotypes (thus leaving out of the re-screening<br />

process all those with the HLA haplotypes<br />

inconsistent with celiac disease) has been<br />

proposed. 69 An analogous strategy might be<br />

applied to screen for celiac disease patients with<br />

the rarer Williams’ syndrome, where an increased<br />

incidence of celiac disease has been reported. 70<br />

Complications<br />

Untreated, celiac disease can lead to a number of<br />

complications. Some, such as chronic liver<br />

disease, osteopenia/osteoporosis, infertility or<br />

psychiatric disorders, have been discussed under<br />

clinical presentations. Other important complications<br />

include hyposplenism, non-responsive celiac<br />

disease (including refractory sprue) and malignancy.


Hyposplenism<br />

Often seen in older patients, less commonly in<br />

children, this condition is detected by the presence<br />

of Howell–Jolly bodies and thrombocytosis 71<br />

and confirmed by imaging techniques.<br />

Non-responsive celiac disease<br />

Uncommonly, patients who have been diagnosed<br />

with celiac disease either fail to respond to the<br />

diet, continuing to present the same symptoms<br />

and signs, or they soon relapse, after a brief apparent<br />

improvement. This condition can be defined as<br />

‘non-responsive celiac disease’ with initial or<br />

subsequent failure of what appears to be a strict<br />

gluten-free diet to improve symptoms and/or to<br />

restore normal intestinal architecture and function<br />

in patients who have celiac-like enteropathy. This<br />

syndrome, which includes true refractory sprue,<br />

can be due to a number of causes, which are<br />

reported in Table 27.4.<br />

It should be stressed that, in any population of<br />

celiacs reported to fail to respond to a gluten-free<br />

diet, whether pediatric or adult, by far the most<br />

common cause remains the continued ingestion of<br />

gluten, often inadvertent. Thus, a meticulous<br />

search for hidden sources of gluten is the recommended<br />

first-line investigative approach.<br />

Table 27.4 Main causes of apparently nonresponsive<br />

celiac disease (‘refractory sprue’)<br />

Continued ingestion of gluten<br />

Incorrect diagnosis, e.g.<br />

Crohn’s disease<br />

autoimmune enteropathy<br />

eosinophilic gastroenteritis<br />

giardiasis<br />

cow’s milk protein allergic enteropathy<br />

Presence of a concomitant food allergy<br />

Lactose intolerance<br />

secondary to reduced absorptive surface<br />

(transient)<br />

adult-type lactase deficiency (permanent)<br />

Irritable bowel syndrome<br />

Pancreatic insufficiency<br />

Complications 443<br />

Second, an erroneous diagnosis of celiac disease is<br />

to be suspected. Particular care should be used in<br />

all cases to rule out other conditions that also<br />

present with morphological changes in the duodenal<br />

mucosa (see Table 27.4 for some examples).<br />

Among them, Crohn’s disease is a growing<br />

concern.<br />

Third, particularly in pediatric cases of celiac<br />

disease apparently unresponsive to a gluten-free<br />

diet, a concomitant food allergy, such as to milk or<br />

egg protein, must be sought. Indeed, the high<br />

prevalence of cow’s milk protein allergy makes the<br />

coincidental overlapping of the two conditions a<br />

not uncommon occurrence.<br />

Also, in cases with overt gastrointestinal symptoms,<br />

lactase deficiency is commonly present at<br />

diagnosis, as a result of the reduction of the<br />

absorptive area and hence of enough available<br />

enzymatic activity. Under these circumstances, it<br />

is not surprising that for some time a secondary<br />

lactose intolerance causes symptoms such as<br />

abdominal bloating, gassiness and diarrhea.<br />

Unless lactase deficiency is the result of the<br />

permanent loss of this enzymatic activity (‘adulttype’<br />

or ‘late-onset’ lactase deficiency), the<br />

problem is self-limited and within a few weeks in<br />

the majority of patients lactose can be reintroduced<br />

in the diet.<br />

Irritable bowel syndrome, whose recently<br />

proposed association with celiac disease in a<br />

proportion of cases 72 has spurred much debate, is<br />

not uncommonly found in celiac disease patients,<br />

mostly teenagers or young adults. Clearly, this<br />

remains a diagnosis of exclusion, and needs to be<br />

supported by evidence of lack of any problem with<br />

adequate weight gain or maintenance. Once irritable<br />

bowel syndrome is diagnosed in a celiac<br />

patient, obviously the gluten-free diet must be<br />

maintained, and any of the currently available<br />

modalities of management can be utilized.<br />

Pancreatic insufficiency, found also in patients<br />

who do not have an underlying important condition<br />

of malabsorption and malnutrition, 73 has been<br />

shown in newly diagnosed celiac patients to cause<br />

failure of an adequate response to the diet, particularly<br />

in terms of growth, 74 and can of course be<br />

treated with adequate pancreatic enzyme supplementation.<br />

75


444<br />

Celiac disease<br />

Refractory sprue<br />

A condition of persistence of severe symptoms<br />

(typically malabsorption), high serum autoantibodies<br />

and enteropathy in spite of a strict glutenfree<br />

diet and in the documented absence of any of<br />

the disorders mentioned above, is rare, and has<br />

never been described in pediatric age. It is termed<br />

refractory sprue. In a multicenter study conducted<br />

in France, it has been shown in the majority of<br />

cases to be characterized by abnormal monoclonal<br />

intraepithelial T lymphocytes expressing a cytoplasmic<br />

CD3 chain (CD3c), lacking CD3 and CD8<br />

surface expression, and showing T cell receptor-γ<br />

gene rearrangements. 76 These authors suggested<br />

that refractory sprue associated with an aberrant<br />

clonal population of intraepithelial lymphocytes<br />

may be classified as cryptic enteropathy-associated<br />

T-cell lymphoma. The same group subsequently<br />

developed an immunohistochemical technique<br />

that would allow rapid identification of this<br />

condition. 77 Refractory sprue is a difficult condition<br />

to treat, and multiple aggressive immunosuppressive<br />

regimens have been suggested,<br />

ranging from cyclosporine to infliximab. 78–83<br />

Malignancy<br />

The development of malignancy, particularly<br />

lymphoma of the small bowel, but also carcinoma<br />

of any segment of the esophagus and stomach, is<br />

the most serious complication to affect patients<br />

with celiac disease. 84 The association has been<br />

known for about 40 years, yet many questions are<br />

still unanswered. In particular, it is unclear why<br />

only some celiac patients develop malignancy.<br />

Development of malignancy is the main explanation<br />

for the well-documented shorter life<br />

expectancy of celiac patients who are either<br />

untreated, or diagnosed very late in the course of<br />

their disease, especially when presenting severe<br />

malabsorption. 85<br />

Diagnosis<br />

The European Society of Paediatric<br />

Gastroenterology, Hepatology and Nutrition<br />

(ESPGHAN) established the criteria for diagnosis<br />

of celiac disease more than 30 years ago. 86 These<br />

original criteria, which have been widely followed<br />

by both adult and pediatric gastroenterologists for<br />

more than two decades, stated that diagnosis of<br />

celiac disease would require:<br />

(1) The presence of compatible symptoms and<br />

demonstration of a structurally abnormal<br />

small-intestinal mucosa when taking a diet<br />

containing gluten;<br />

(2) A clear clinical response to a gluten-free diet;<br />

(3) Documentation of unequivocal improvement<br />

of the villous structure after having taken a<br />

gluten-free diet for 1 year;<br />

(4) Deterioration of the mucosa and reappearance<br />

of symptoms during challenge with<br />

gluten.<br />

With increasing availability of serological tests for<br />

diagnosis, and following a report of an Italian<br />

multicenter investigation in more than 3000 celiac<br />

children, 87 ESPGHAN in 1990 proposed new diagnostic<br />

criteria. According to them, the diagnosis of<br />

celiac disease can be established on a definitive<br />

basis when the characteristic changes of the<br />

duodenal mucosa are found in a child with signs<br />

and/or symptoms consistent with celiac disease,<br />

provided that a full and unequivocal clinical<br />

remission after withdrawal of gluten is seen, associated<br />

with the disappearance of circulating antibodies.<br />

These criteria are currently universally<br />

utilized, and it can be seen that, although the<br />

emerging role of serology testing was recognized<br />

and taken into crucial consideration, diagnosis<br />

still clearly relies on intestinal biopsy findings. 88<br />

The role of the serological markers of celiac<br />

disease will be discussed later, but it should be<br />

clear that they must be considered an important<br />

means of screening and of monitoring compliance,<br />

offering only supportive evidence for the diagnosis.<br />

89<br />

Duodenal biopsy<br />

Currently, most of the bioptic samples are obtained<br />

from the duodenal mucosa during endoscopy.<br />

Endoscopic changes described in untreated celiac<br />

patients include scalloping, a mosaic pattern and<br />

flattening or paucity of the folds. 90 However, these<br />

findings are not specific 91 and occur only infrequently<br />

in the pediatric age. Adequate biopsy<br />

specimens can be obtained at endoscopy or with a


suction biopsy tube. Specimens should be<br />

obtained from the distal duodenum. Obtaining<br />

multiple biopsies is crucial for a correct diagnosis.<br />

In fact, the old teaching that celiac disease results<br />

in a continuum of lesions has recently been proved<br />

incorrect. The typical flattening of villi can be<br />

patchy, with blunted villi juxtaposed to normal<br />

mucosa. 92 The diagnosis therefore can be easily<br />

missed if at least four or five bioptic samples are<br />

not obtained. Histological examination of involved<br />

areas confirms the loss of normal villous structure<br />

with severe shortening up to complete absence<br />

(flat mucosa) of the villi. The intestinal crypts are<br />

markedly elongated and hyperplastic. Unlike the<br />

well-differentiated, mature absorptive cells, the<br />

undifferentiated crypt cells are markedly increased<br />

in number in untreated celiac disease, and this<br />

accounts for the obvious lengthening of crypts.<br />

The cellularity of the lamina propria is increased<br />

and the thickness of the mucosa is overall also<br />

increased. The cellular infiltrate consists largely of<br />

plasma cells and lymphocytes. Another typical<br />

(but not pathognomonic) change that occurs in the<br />

small intestinal mucosa of untreated celiac<br />

patients is the increase in the number of intraepithelial<br />

lymphocytes, particularly T cells expressing<br />

the γδ receptors. 93 This change (Figure 27.4) is<br />

considered an early and subtle sign of celiac<br />

disease and, if supported by concordant serology<br />

and clinical findings, can be considered sufficient<br />

to finalize the diagnosis. Indeed, it was shown that<br />

these patients would benefit from a gluten-free<br />

diet. 94 Some patients may also have only minor<br />

morphometric changes on jejunal biopsies with<br />

changes in mean surface/volume ratio without<br />

gross histological abnormalities. 95,96 As it can be<br />

seen therefore, it is imperative that an experienced<br />

pathologist carefully review intestinal biopsies for<br />

intraepithelial lymphocytes and morphometric<br />

changes in a patient with positive celiac antibodies<br />

before labeling serology results as false positive.<br />

Furthermore, to complicate matters, some studies<br />

have demonstrated that patients with totally<br />

normal histology but positive celiac serology who<br />

are labeled as false positive may develop typical<br />

celiac morphological changes on follow-up. 97,98<br />

Thus, development of serum antibodies seems to<br />

precede, at least in some well-documented cases,<br />

that of the morphological lesions. For this reason,<br />

it appears wise to continue to follow up patients<br />

who have high levels of anti-endomysium or tissue<br />

Diagnosis 445<br />

transglutaminase antibodies, even with a normal<br />

mucosa, and to repeat the endoscopy if clinical<br />

signs or symptoms consistent with celiac disease<br />

appear.<br />

Serology<br />

Serologic testing for celiac disease has included<br />

testing for food protein-directed antibodies<br />

(antigliadin (AGA)) and for an autoantibody (antiendomysium).<br />

99 It should be recognized that, in<br />

spite of high potential in clinical settings, particularly<br />

for screening purposes, the sensitivity and<br />

specificity of serology have varied widely, in part<br />

because of the lack of standardized protocols and<br />

of reference sera. Inter-laboratory variations have<br />

been wide, particularly for AGA, as pointed out by<br />

Figure 27.4 Morphologically normal villus with<br />

increased intraepithelial lymphocytes. In the early stage of<br />

the changes induced by celiac disease, an otherwise<br />

normal villus can be seen to have an increased intraepithelial<br />

infiltration of lymphocytes.


446<br />

Celiac disease<br />

multicenter studies performed both in Europe 100<br />

and in the USA. 101 AGA have been widely utilized<br />

since the early 1980s, having soon become widely<br />

available and inexpensive. Two classes are<br />

currently measured: IgG and IgA. The former, even<br />

though considered of high sensitivity (85–98% in<br />

most large series from Europe), has been repeatedly<br />

shown to be extremely non-specific. Indeed,<br />

AGA-IgG can be found in about 30% of a control<br />

population, and thus their positive value is of little<br />

use. AGA-IgA, on the other hand, are known to be<br />

generally much more specific (95–100%), but are<br />

unfortunately also less sensitive (sensitivities<br />

reported range between 70 and 92%). The value of<br />

measuring AGA for screening purposes is therefore<br />

at best doubtful. However, this class of antibodies<br />

maintains its usefulness in monitoring the compliance<br />

to the gluten-free diet in an already established<br />

celiac patient, as it is known that AGA are<br />

more prone to reappear in the presence of even<br />

minimal dietary transgressions, where antiendomysium<br />

antibodies are less sensitive. 102 In<br />

practice, when screening a patient for celiac<br />

disease, one should always obtain total IgA, along<br />

with anti-endomysium antibodies or tissue transglutaminase<br />

antibodies (see below for anti-tissue<br />

transglutaminase antibodies). If a condition of<br />

total IgA deficiency is found (a condition affecting<br />

approximately 3% of celiac patients 103 ), then antiendomysium<br />

antibodies or tissue transglutaminase<br />

antibodies should be sought, as, in IgA-deficient<br />

subjects, IgG- and specifically IgG1-anti-endomysium<br />

or IgG-tissue transglutaminase antibodies<br />

become reliable indicators of celiac disease. 104<br />

The anti-endomysium antibodies are detected by<br />

assessing the immunofluorescence of sections of<br />

monkey esophageal or human umbilical cord<br />

smooth muscle on exposure to sera from patients<br />

being tested. Although the test relies on subjective<br />

operator assessment of fluorescence, its specificity<br />

in detecting untreated celiac disease is extremely<br />

high. A recent study conducted with strict criteria<br />

by the European working group on Serological<br />

Screening for Celiac Disease showed that antiendomysium<br />

antibodies had, among seven laboratories,<br />

a remarkable mean specificity of 99%<br />

(93.9–99.9%), while the mean sensitivity proved to<br />

be 90% (82.7–92.5%). 105 Also, the inter-laboratory<br />

reliability proved to be quite good for this test,<br />

unlike the poor reproducibility of AGA. This assay<br />

is relatively costly, and its utilization of monkey<br />

esophagus further limits its use for the screening of<br />

large populations.<br />

Tissue transglutaminase antibodies<br />

In 1997 Dieterich et al identified tissue transglutaminase<br />

as the autoantigen of celiac disease. 106 In a<br />

subsequent study by the same group, an enzymelinked<br />

immunosorbent assay (ELISA) for guineapig<br />

IgA tissue transglutaminase antibodies was<br />

found to be 98.1% sensitive and 94.7% specific in<br />

patients with biopsy-proved celiac disease. 107<br />

Since then, many studies have confirmed the high<br />

sensitivity and specificity of these antibodies in<br />

the diagnosis of celiac disease. Sensitivity and<br />

specificity have been subsequently further<br />

improved by utilizing the human antigen instead<br />

of the guinea pig antigen. 108–110 A simple noninvasive<br />

immunological dot blot assay based on<br />

recognition of recombinant human transglutaminase<br />

has also recently been proposed as a practical,<br />

reliable alternative to both the immunofluorescence-based<br />

anti-endomysium test and tissue<br />

transglutaminase ELISA for the diagnosis of celiac<br />

disease. 111<br />

In summary, even though currently anti-endomysium<br />

antibody tests can still be considered more<br />

specific, it is likely that serological (or whole<br />

blood) tests based on human tissue transglutaminase<br />

antibodies will become the gold standard for<br />

celiac disease screening in the very near future,<br />

eventually replacing the semiquantitative,<br />

observer-dependent, costly and time-consuming<br />

anti-endomysium antibody test.<br />

Currently, however, it seems that anti-tissue transglutaminase<br />

antibodies are less specific than antiendomysium<br />

antibodies for celiac disease. 112<br />

Treatment<br />

Total lifelong avoidance of gluten ingestion is the<br />

cornerstone treatment for celiac disease. Wheat,<br />

rye and barley are the grains containing toxic<br />

peptides. For a long time, oats were also considered<br />

toxic, and their elimination from the diet was<br />

recommended. However, during the past few years<br />

a growing body of scientific evidence obtained<br />

from in vitro studies as well as from clinical inves-


tigations, particularly in adults 113–115 but also<br />

recently in children, 116 allows us to conclude that<br />

oats are indeed totally safe. This makes sense<br />

based on the genetics of the grains, showing that<br />

oats are genetically entirely unrelated to the group<br />

of wheat, rye and barley. 117 Indeed, a recent in<br />

vitro study looking at the specificity of tissue transglutaminase<br />

further supported the lack of toxicity<br />

of oats. 118 However, because of uncontrolled<br />

harvesting and milling procedures, cross-contamination<br />

of oats with gluten is still a concern, which<br />

will have to be addressed in the context of a more<br />

general policy concerning labeling of food certified<br />

to be gluten-free.<br />

Often in the initial phases of dietary treatment,<br />

lactose is also eliminated. This has its basis in the<br />

lactase deficiency that is thought to accompany<br />

the flat mucosa. 119 However, it should be pointed<br />

out that, as we have seen, today most new celiacs<br />

are diagnosed in the absence of overt malabsorptive<br />

symptoms, and in these circumstances clinically<br />

significant lactose malabsorption or intolerance<br />

is rarely seen. Furthermore, even in cases<br />

with obvious malabsorption, the recovery of<br />

lactase activity is typically fast, so that the use of a<br />

lactose-free diet even in these cases must be on a<br />

short-term basis only. The possibility of an association<br />

between celiac disease and milk protein<br />

allergy has been repeatedly raised in the past, but<br />

it is now clear that the two conditions may simply<br />

coexist as a result of a statistical association, so<br />

that again there is no need to recommend avoidance<br />

of ‘dairy products’.<br />

REFERENCES<br />

1. Sollid LM. Molecular basis of celiac disease. Annu Rev<br />

Immunol 2000; 18: 53–81.<br />

2. Hernell O, Ivarsson A, Persson LA. Coeliac disease:<br />

effect of early feeding on the incidence of the disease.<br />

Early Hum Dev 2001; 65 Suppl: S153–S160.<br />

3. Ivarsson A, Hernell O, Stenlund H, Persson LA. Breastfeeding<br />

protects against celiac disease. Am J Clin Nutr<br />

2002; 75: 914–921.<br />

4. Ivarsson A, Hernell O, Nystrom L, Persson LA. Children<br />

born in the summer have increased risk for coeliac<br />

disease. J Epidemiol Community Health 2003; 57: 36–39.<br />

References 447<br />

A commonly asked question is ‘how strict must the<br />

diet be’? This implies that there could be a threshold,<br />

below which ‘some gluten’ might be tolerated.<br />

We know that the morphological damage is doserelated,<br />

120 and that amounts of gluten as low as<br />

100mg/kg per day may induce damage at the clinical,<br />

histological or laboratory level. 121 Thus, there<br />

is no scientific answer to the question of how low<br />

a dose is safe. In addition, tolerance is highly variable<br />

between celiac individuals, making predictions<br />

in single cases impossible. This is the basis of<br />

the only reasonable dietetic recommendation for<br />

celiac patients: ‘zero tolerance’. Currently, there is<br />

wide availability of resources to assist celiac<br />

patients and their families: recipe books, newsletters<br />

from support groups that continuously update<br />

lists of ‘safe’ products and hospital-affiliated<br />

specific programs. Clearly, however, a close interaction<br />

between families on the one hand and the<br />

dietitian–doctor team on the other is necessary to<br />

achieve optimal compliance. This need is especially<br />

important during adolescence, a time when<br />

dietetic compliance can be difficult, 122–124 and<br />

requires attention to possible dietetic deficiencies,<br />

124 but also in adults, who commonly end up<br />

by only partially adhering to the diet. 125 It is<br />

indeed important to realize that a close adherence<br />

to the gluten-free diet is associated with improved<br />

quality of life, both for patients with symptomdetected<br />

celiac disease and for those – mostly<br />

asymptomatic – in whom the diagnosis was<br />

reached as a result of a screening program. 126<br />

5. Ascher H, Krantz I, Kristiansson B. Increasing incidence<br />

of coeliac disease in Sweden. Arch Dis Child 1991; 66:<br />

608–611.<br />

6. Catassi C, Ratsch IM, Fabiani E et al. Coeliac disease in<br />

the year 2000: exploring the iceberg. Lancet 1994; 343:<br />

200–203.<br />

7. Maki M, Kallonen K, Lahdeaho ML, Visakorpi JK.<br />

Changing pattern of childhood coeliac disease in<br />

Finland. Acta Paediatr Scand 1988; 77: 408–412.<br />

8. Kolho KL, Farkkila MA, Savilahti E. Undiagnosed<br />

coeliac disease is common in Finnish adults. Scand J<br />

Gastroenterol 1998; 33: 1280–1283.


448<br />

Celiac disease<br />

9. Fasano A, Berti I, Gerarduzzi T et al. Prevalence of<br />

celiac disease in at-risk and not at-risk groups in the<br />

United States. Arch Intern Med 2003; 163: 286–292.<br />

10. Catassi C, Tarsch IM, Gandolfi L et al. Why is coeliac<br />

disease endemic in the people of the Sahara? Lancet<br />

1999; 354: 647–648.<br />

11. Shan L, Molberg O, Parrot I et al. Structural basis for<br />

gluten intolerance in celiac sprue. Science 2002; 297:<br />

2275–2279.<br />

12. Schulzke JD, Bentzel CJ, Schulzke I et al. Epithelial<br />

tight junction structure in the jejunum of children with<br />

acute and treated celiac sprue. Pediatr Res 1998; 43:<br />

435–441.<br />

13. Clemente M, De Virgiliis S, Kang JS et al. Early effects<br />

of gliadin on enterocyte intracellular signalling involved<br />

in intestinal barrier function. Gut 2003; 52: 218–223.<br />

14. Fasano A, Not T, Wang W et al. Zonulin, a newly<br />

discovered modulator of intestinal permeability, and its<br />

expression in coeliac disease. Lancet 2001; 355:<br />

1518–1519.<br />

15. Maiuri L, Ciacci C, Ricciardelli I et al. Association<br />

between innate response to gliadin and activation of<br />

pathogenic T cells in coeliac disease. Lancet 2003; 362:<br />

30–37.<br />

16. Anderson RP, Degano P, Godkin AJ et al. In vivo antigen<br />

challenge in celiac disease identifies a single<br />

transglutaminase-modified peptide as the dominant Agliadin<br />

T-cell epitope. Nature Med 2000; 6: 337–342.<br />

17. Wucherpfennig K. Insights into autoimmunity gained<br />

from structural analysis of MHC–peptide complexes.<br />

Curr Opin Immunol 2001; 13: 650–656.<br />

18. Marsh MN. Gluten, major histocompatibility complex,<br />

and the small intestine. A molecular and immunobiologic<br />

approach to the spectrum of gluten sensitivity<br />

(‘celiac sprue’). Gastroenterology 1992; 102: 330–354.<br />

19. Maiuri L, Ciacci C, Raia V et al. FAS engagement drives<br />

apoptosis of enterocytes of coeliac patients. Gut 2001;<br />

48: 418–424.<br />

20. Maiuri L, Ciacci C, Auricchio S et al. Interleukin 15<br />

mediates epithelial changes in celiac disease.<br />

Gastroenterology 2000; 119: 996–1006.<br />

21. Jabri B, de Serre NP, Cellier C et al. Selective expansion<br />

of intraepithelial lymphocytes expressing the HLA-Especific<br />

natural killer receptor CD94 in celiac disease.<br />

Gastroenterology 2000; 118: 867–879.<br />

22. Visakorpi JK, Maki M. Changing clinical features of<br />

coeliac disease. Acta Paediatr Suppl 1994; 83: 10–13.<br />

23. Bottaro G, Cataldo F, Rotaolo N et al. The clinical<br />

pattern of subclinical/silent celiac disease: an analysis<br />

on 1026 consecutive cases. Am J Gastroenterol 1999; 94:<br />

691–696.<br />

24. Andersson H, Mobacken H. Dietary treatment of<br />

dermatitis herpetiformis. Eur J Clin Nutr 1992; 46:<br />

309–315.<br />

25. Collin P, Reunala T. Recognition and management of the<br />

cutaneous manifestations of celiac disease: a guide for<br />

dermatologists. Am J Clin Dermatol 2003; 4: 13–20.<br />

26. Aine L. Coeliac-type permanent-tooth enamel defects.<br />

Ann Med 1996; 28: 9–12.<br />

27. Mody RJ, Brown PI, Wechsler DS. Refractory iron<br />

deficiency anemia as the primary clinical manifestation<br />

of celiac disease. J Pediatr Hematol Oncol 2003; 25:<br />

169–172.<br />

28. Corazza GR, Valentini RA, Andreani ML et al.<br />

Subclinical coeliac disease is a frequent cause of irondeficiency<br />

anaemia. Scand J Gastroenterol 1995; 30:<br />

153–156.<br />

29. Howard MR, Turnbull AJ, Morley P et al. A prospective<br />

study of the prevalence of undiagnosed coeliac disease<br />

in laboratory defined iron and folate deficiency. J Clin<br />

Pathol 2002; 55: 754–757.<br />

30. Stenhammar L, Fallstrom SP, Jansson G et al. Coeliac<br />

disease in children of short stature without gastrointestinal<br />

symptoms. Eur J Pediatr 1986; 145: 185–186.<br />

31. Tumer L, Hasanoglu A, Aybay C. Endomysium<br />

antibodies in the diagnosis of celiac disease in shortstatured<br />

children with no gastrointestinal symptoms.<br />

Pediatr Int 2001; 43: 71–73.<br />

32. Smecuol E, Maurino E, Vazquez H et al. Gynaecological<br />

and obstetric disorders in coeliac disease: frequent<br />

clinical onset during pregnancy or the puerperium. Eur<br />

J Gastroenterol Hepatol 1996; 8: 63–89.<br />

33. Bardella MT, Fraquelli M, Quatrini et al. Prevalence of<br />

hypertransaminasemia in adult celiac patients and<br />

effect of gluten-free diet. Hepatology 1995; 22: 833–836.<br />

34. Volta U, De Franceschi L, Lari F et al. Coeliac disease<br />

hidden by cryptogenic hypertransaminasaemia. Lancet<br />

1998; 352: 26–29.<br />

35. Kaukinen K, Halme L, Collin P et al. Celiac disease in<br />

patients with severe liver disease: gluten-free diet may<br />

reverse hepatic failure. Gastroenterology 2002; 122:<br />

881–888.<br />

36. Lubrano E, Ciacci C, Ames PR et al. The arthritis of<br />

coeliac disease: prevalence and pattern in 200 adult<br />

patients. Br J Rheumatol 1996; 35: 1314–1318.<br />

37. Lepore L, Martelossi S, Pennesi M et al. Prevalence of<br />

celiac disease in patients with juvenile chronic arthritis.<br />

J Pediatr 1996; 129: 311–313.<br />

38. Kemppainen T, Kroger H, Janatuinen E et al.<br />

Osteoporosis in adult patients with celiac disease. Bone<br />

1999; 24: 249–255.<br />

39. Meyer D, Stavropolous S, Diamond B et al. Osteoporosis<br />

in a north american adult population with celiac<br />

disease. Am J Gastroenterol 2001; 96: 112–119.<br />

40. Mustalahti K, Collin P, Sievanen H et al. Osteopenia in<br />

patients with clinically silent coeliac disease warrants<br />

screening. Lancet 1999; 354: 744–745.<br />

41. Vasquez H, Mazure R, Gonzalez D et al. Risk of fractures<br />

in celiac disease patients: a cross-sectional, case-control<br />

study. Am J Gastroenterol 2000; 95: 183–189.<br />

42. Scott E, Gaywood I, Scott B. Guidelines for osteoporosis<br />

in coeliac disease and inflammatory bowel disease. Gut<br />

2000; 46(Suppl 1): 1–8.<br />

43. Thomason K, West J, Logan RF et al. Fracture experience<br />

of patients with coeliac disease: a population<br />

based survey. Gut 2003; 52: 518–522.<br />

44. Kalayci A, Kansu A, Girgin N et al. Bone mineral<br />

density and importance of a gluten-free diet in patients<br />

with celiac disease in childhood. Pediatrics 2001; 108:<br />

E89.<br />

45. Kemppainen T, Kroger H, Janatuinen E et al. Bone<br />

recovery after a gluten-free diet: a 5-year follow-up<br />

study. Bone 1999; 25: 355–360.<br />

46. Sategna-Guidetti C, Grosso SB, Grosso S et al. The<br />

effects of 1-year gluten withdrawal on bone mass, bone<br />

metabolism and nutritional status in newly-diagnosed<br />

adult coeliac disease patients. Aliment Pharmacol Ther<br />

2000; 14: 35–43.<br />

47. Sugai E, Chernavsky A, Pedreira S et al. Bone-specific<br />

antibodies in sera from patients with celiac disease:<br />

characterization and implications in osteoporosis. J Clin<br />

Immunol 2002; 22: 353–362.<br />

48. Hadjivassiliou M, Grunewald R, Sharrack B et al.<br />

Gluten ataxia in perspective: epidemiology, genetic<br />

susceptibility and clinical characteristics. Brain 2003;<br />

126: 685–691.<br />

49. Gobbi G, Bouquet F, Greco L et al. Coeliac disease,<br />

epilepsy, and cerebral calcifications. The Italian


Working Group on Coeliac Disease and Epilepsy [see<br />

comments]. Lancet 1992; 340: 439–443.<br />

50. Kieslich M, Errazuriz G, Posselt HG et al. Brain whitematter<br />

lesions in celiac disease: a prospective study of<br />

75 diet-treated patients. Pediatrics 2001; 108: E21.<br />

51. Black C, Kaye J, Jick H. Relation of childhood gastrointestinal<br />

disorders to autism: nested case-control study<br />

using data from the UK General Practice Research<br />

Database. BMJ 2002; 325: 419–421.<br />

52. Ciacci C, Iavarone A, Mazzacca G, De Rosa A.<br />

Depressive symptoms in adult coeliac disease. Scand J<br />

Gastroenterol 1998; 33: 247–250.<br />

53. Addolorato G, Capristo E, Ghittoni G et al. Anxiety but<br />

not depression decreases in coeliac patients after oneyear<br />

gluten-free diet: a longitudinal study. Scand J<br />

Gastroenterol 2001; 36: 502–506.<br />

54. Carta MG, Hardoy MC, Boi MF et al. Association<br />

between panic disorder, major depressive disorder and<br />

celiac disease: a possible role of thyroid autoimmunity. J<br />

Psychosom Res 2002; 53: 789–793.<br />

55. Collin P, Vilska S, Heinonen PK et al. Infertility and<br />

coeliac disease. Gut 1996; 39: 382–384.<br />

56. Ciacci C, Cirillo M, Auriemma G et al. Celiac disease<br />

and pregnancy outcome. Am J Gastroenterol 1996; 91:<br />

718–722.<br />

57. Ludvigsson JF, Ludvigsson J. Coeliac disease in the<br />

father affects the newborn. Gut 2001; 49: 169–175.<br />

58. Petaros P, Martelossi S, Tommasini A et al. Prevalence of<br />

autoimmune disorders in relatives of patients with<br />

celiac disease. Dig Dis Sci 2002; 47: 1427–1431.<br />

59. Frustaci A, Cuoco L, Chimenti C et al. Celiac disease<br />

associated with autoimmune myocarditis. Circulation<br />

2002; 105: 2611–2618.<br />

60. Ventura A, Magazzu G, Greco L. Duration of exposure to<br />

gluten and risk for autoimmune disorders in patients<br />

with celiac disease. SIGEP Study Group for<br />

Autoimmune Disorders in Celiac Disease.<br />

Gastroenterology 1999; 117: 297–303.<br />

61. James M, Scott B. Coeliac disease: the cause of the<br />

various associated disorders? Eur J Gastroenterol<br />

Hepatol 2001; 13: 1119–1121.<br />

62. Barera G, Bonfanti R, Viscardi M et al. Occurrence of<br />

celiac disease after onset of type 1 diabetes: a 6-year<br />

prospective longitudinal study. Pediatrics 2002; 109:<br />

833–838.<br />

63. Holmes G. Coeliac disease and type 1 diabetes mellitus:<br />

the case for screening. Diabet Med 2001; 18: 169–177.<br />

64. Carlsson A, Axelsson I, Borulf S et al. Prevalence of IgAantigliadin<br />

antibodies and IgA-antiendomysium antibodies<br />

related to celiac disease in children with Down<br />

syndrome. Pediatrics 1998; 101: 272–275.<br />

65. Gale L, Wimalaratna H, Brotodiharjo A, Duggan JM.<br />

Down’s syndrome is strongly associated with coeliac<br />

disease. Gut 1997; 40: 492–496.<br />

66. Bonamico M, Mariani P, Danesi HM et al. Prevalence<br />

and clinical picture of celiac disease in italian down<br />

syndrome patients: a multicenter study. J Pediatr<br />

Gastroenterol Nutr 2001; 33: 139–143.<br />

67. Book L, Hart A, Black J et al. Prevalence and clinical<br />

characteristics of celiac disease in Downs syndrome in a<br />

US study. Am J Med Genet 2001; 98: 70–74.<br />

68. Zachor DA, Mroczek-Musulman E, Brown P. Prevalence<br />

of celiac disease in Down syndrome in the United<br />

States. J Pediatr Gastroenterol Nutr 2000; 31: 275–279.<br />

69. Csizmadia C, Mearin ML, Oren A et al. Accuracy and<br />

cost-effectiveness of a new strategy to screen for celiac<br />

disease in children with Down syndrome. J Pediatr<br />

2000; 137: 756–761.<br />

References 449<br />

70. Giannotti A, Tiberio G, Castro M et al. Coeliac disease<br />

in Williams syndrome. J Med Genet 2001; 38: 767–768.<br />

71. O’Grady JG, Steens FM, Harding B et al. Hyposplenism<br />

and gluten-sensitive enteropathy: natural history, incidence,<br />

and relationship to diet and small bowel<br />

morphology. Gastroenterology 1984; 87: 1326–1331.<br />

72. Wahnschaffe U, Ullrich R, Riecken EO, Schulzke JD.<br />

Celiac disease-like abnormalities in a subgroup of<br />

patients with irritable bowel syndrome.<br />

Gastroenterology 2001; 121: 1329–1338.<br />

73. Carroccio A, Iacono G, Montalto G et al. Pancreatic<br />

insufficiency in celiac disease is not dependent on<br />

nutritional status. Dig Dis Sci 1994; 39: 2235–2242.<br />

74. Carroccio A., Iacono G, Lerro P et al. Role of pancreatic<br />

impairment in growth recovery during gluten-free diet<br />

in childhood celiac disease. Gastroenterology 1997; 112<br />

1839–1844.<br />

75. Carroccio A, Iacono G, Montalto G et al. Pancreatic<br />

enzyme therapy in childhood celiac disease. A doubleblind<br />

prospective randomized study. Dig Dis Sci 1995;<br />

40: 2555–2560.<br />

76. Cellier C, Delabesse E, Helmer C et al. Refractory sprue,<br />

coeliac disease, and enteropathy-associated T-cell<br />

lymphoma. French Coeliac Disease Study Group. Lancet<br />

2000; 356: 203–208.<br />

77. Patey-Mariaud De Serre N, Cellier C, Jabri B et al.<br />

Distinction between coeliac disease and refractory<br />

sprue: a simple immunohistochemical method.<br />

Histopathology 2000; 37: 70–77.<br />

78. Longstreth GF. Successful treatment of refractory sprue<br />

with cyclosporine. Ann Intern Med 1993; 119:<br />

1014–1016.<br />

79. Mandal A, Mayberry J. Elemental diet in the treatment<br />

of refractory coeliac disease. Eur J Gastroenterol Hepatol<br />

2001; 13: 79–80.<br />

80. Maurino E, Niveloni S, Chernavsky A et al.<br />

Azathioprine in refractory sprue: results from a prospective,<br />

open-label study. Am J Gastroenterol 2002; 97:<br />

2595–2602.<br />

81. Gillett HR, Amott ID, McIntyre M et al. Successful<br />

infliximab treatment for steroid-refractory celiac<br />

disease: a case report. Gastroenterology 2002; 122:<br />

800–805.<br />

82. Mulder CJ, Wahab PJ Meijer JW et al. A pilot study of<br />

recombinant human interleukin-10 in adults with<br />

refractory coeliac disease. Eur J Gastroenterol Hepatol<br />

2001; 13: 1183–1188.<br />

83. Wahab PJ, Crusius JB, Meijer JW et al. Cyclosporin in<br />

the treatment of adults with refractory coeliac disease –<br />

an open pilot study. Aliment Pharmacol Ther 2000; 14:<br />

767–774.<br />

84. Holmes GK. Coeliac disease and malignancy. Dig Liver<br />

Dis 2002; 34: 229–237.<br />

85. Corrao G, Corazza GR, Bagnardi V et al. Mortality in<br />

patients with coeliac disease and their relatives: a<br />

cohort study. Lancet 2001; 358: 356–361.<br />

86. Meeuwisse G. Round table discussion. Diagnostic criteria<br />

in coeliac disease. Acta Paediatr Scand 1970; 59:<br />

461–463.<br />

87. Guandalini S, Ventura A, Ansaldi N et al. Diagnosis of<br />

coeliac disease: time for a change? Arch Dis Child 1989;<br />

64: 1320–4; discussion 1324–5.<br />

88. Working Group of European Society of Paediatric<br />

Gastroenterology and Nutrition. Revised criteria for<br />

diagnosis of coeliac disease. Arch Dis Child 1990; 65:<br />

909–911.<br />

89. Guandalini S, Gupta P. Do you still need a biopsy to<br />

diagnose celiac disease? Curr Gastroenterol Opin 2001;<br />

3: 385–391


450<br />

Celiac disease<br />

90. Jabbari M, Wild G, Goresky CA et al. Scalloped valvulae<br />

conniventes: an endoscopic marker of celiac sprue.<br />

Gastroenterology 1988; 95: 1518–1522.<br />

91. Shah VH, Rotterdam H, Kotler DP et al. All that scallops<br />

is not celiac disease. Gastrointest Endosc 2000; 51:<br />

717–720.<br />

92. Maiuri L, Ciacci C, Raia V et al. FAS engagement drives<br />

apoptosis of enterocytes of coeliac patients. Gut 2001;<br />

48: 418–424.<br />

93. Iltanen S, Hol, K, Ashom M et al. Changing jejunal<br />

gamma delta T cell receptor (TCR)-bearing intraepithelial<br />

lymphocyte density in coeliac disease. Clin Exp<br />

Immunol 1999; 117: 51–55.<br />

94. Arranz E, Ferguson A. Intestinal antibody pattern of<br />

celiac disease: occurrence in patients with normal<br />

jejunal biopsy histology. Gastroenterology 1993; 104:<br />

1263–1272.<br />

95. Corazza G, Valentini RA, Frisoni et al. Gliadin immune<br />

reactivity is associated with overt and latent enteropathy<br />

in relatives of celiac patients. Gastroenterology 1992;<br />

103: 1517–1522.<br />

96. Vazquez H, Cabanne A, Sugai E et al. Serological<br />

markers identify histologically latent coeliac disease<br />

among first-degree relatives. Eur J Gastroenterol Hepatol<br />

1996; 8: 15–21.<br />

97. Collin P, Helin H, Maki M et al. Follow-up of patients<br />

positive in reticulin and gliadin antibody tests with<br />

normal small-bowel biopsy findings. Scand J<br />

Gastroenterol 1993; 28: 595–598.<br />

98. Niveloni S, Pedreira S, Sugai E et al. The natural history<br />

of gluten sensitivity: report of two new celiac disease<br />

patients resulting from a long-term follow-up of nonatrophic,<br />

first-degree relatives. Am J Gastroenterol 2000;<br />

95: 463–468.<br />

99. Lerner A, Kumar V, Iancu TC. Immunological diagnosis<br />

of childhood coeliac disease: comparison between<br />

antigliadin, antireticulin and antiendomysial antibodies.<br />

Clin Exp Immunol 1994; 95: 78–82.<br />

100. Volta U, Lazzari R, Guidetti CS et al. Multicenter study<br />

on the reproducibility of antigliadin (AGA) and antiendomysial<br />

antibodies (EmA) in celiac sprue screening.<br />

The Tenue Club Group. J Clin Gastroenterol 1994; 19:<br />

81–82.<br />

101. Murray JA, Herlein J, Mitros F et al. Serologic testing for<br />

celiac disease in the United States: results of a multilaboratory<br />

comparison study. Clin Diagn Lab Immunol<br />

2000; 7: 584–587.<br />

102. Troncone R, Mayer M, Spagnuolo F et al. Endomysial<br />

antibodies as unreliable markers for slight dietary transgressions<br />

in adolescents with celiac disease. J Pediatr<br />

Gastroenterol Nutr 1995; 21: 69–72.<br />

103. Cataldo F, Marino V, Ventura A et al. Prevalence and<br />

clinical features of selective immunoglobulin A deficiency<br />

in coeliac disease: an Italian multicentre study.<br />

Gut 1998; 42: 362–365.<br />

104. Cataldo F, Lio D, Marino V et al. IgG(1) antiendomysium<br />

and IgG antitissue transglutaminase (anti-tTG) antibodies<br />

in coeliac patients with selective IgA deficiency.<br />

Working Groups on Celiac Disease of SIGEP and Club<br />

del Tenue. Gut 2000; 47: 366–369.<br />

105. Stern M. Comparative evaluation of serologic tests for<br />

celiac disease: a European initiative toward standardization.<br />

Working Group on Serologic Screening for Celiac<br />

Disease. J Pediatr Gastroenterol Nutr 2000; 31: 513–519.<br />

106. Dieterich W, Ehnis T, Bauer M et al. Identification of<br />

tissue transglutaminase as the autoantigen of celiac<br />

disease. Nature Med 1997; 3: 797–801.<br />

107. Dieterich W, Laag E, Schopper H et al. Autoantibodies<br />

to tissue transglutaminase as predictors of celiac<br />

disease. Gastroenterology 1998; 115: 1317–1321.<br />

108. Seissler J, Boms S, Wohlrab U et al. Antibodies to<br />

human recombinant tissue transglutaminase measured<br />

by radioligand assay: evidence for high diagnostic sensitivity<br />

for celiac disease. Horm Metab Res 1999; 31:<br />

375–379.<br />

109. Sblattero D, Berti I, Trevisiol C et al. Human recombinant<br />

tissue transglutaminase ELISA: an innovative diagnostic<br />

assay for celiac disease. Am J Gastroenterol 2000;<br />

95: 1253-1257.<br />

110. Hansson T, Dahlbom I, Hall J et al. Antibody reactivity<br />

against human and guinea pig tissue transglutaminase<br />

in children with celiac disease. J Pediatr Gastroenterol<br />

Nutr 2000; 30: 379–384.<br />

111. Baldas V, Tommasini A, Trevisiol C et al. Development<br />

of a novel rapid non-invasive screening test for coeliac<br />

disease. Gut 2000; 47: 628–631.<br />

112. Carroccio A, Vitale G, Di Prima L et al. Comparison of<br />

anti-transglutaminase ELISAs and an anti-endomysial<br />

antibody assay in the diagnosis of celiac disease: a<br />

prospective study. Clin Chem 2002; 48: 1546–1550.<br />

113. Janatuinen EK, Kemppainen TA, Julkunen RJ et al. No<br />

harm from five year ingestion of oats in coeliac disease.<br />

Gut 2002; 50: 332–335.<br />

114. Storsrud S, Olsson M, Arvidsson Lenner R et al. Adult<br />

coeliac patients do tolerate large amounts of oats. Eur J<br />

Clin Nutr 2003; 57: 163–169.<br />

115. Hardman CM, Garioch JJ, Leonard JN et al. Absence of<br />

toxicity of oats in patients with dermatitis<br />

herpetiformis. N Engl J Med 1997; 337: 1884–1887.<br />

116. Hoffenberg EJ, Haas J, Drescher A et al. A trial of oats in<br />

children with newly diagnosed celiac disease. J Pediatr<br />

2000; 137: 361–366.<br />

117. Devos KM, Gale MD. Comparative genetics in the<br />

grasses. Plant Mol Biol 1997; 35: 3–15.<br />

118. Vader LW, de Ru A, van der Wal Y et al. Specificity of<br />

tissue transglutaminase explains cereal toxicity in celiac<br />

disease. J Exp Med 2002; 195: 643–649.<br />

119. Srinivasan U, Jones E, Weir DG et al. Lactase enzyme,<br />

detected immunohistochemically, is lost in active celiac<br />

disease, but unaffected by oats challenge. Am J<br />

Gastroenterol 1999; 94: 2936–2941.<br />

120. Catassi C, Rossini M, Ratsch IM et al. Dose dependent<br />

effects of protracted ingestion of small amounts of<br />

gliadin in coeliac disease children: a clinical and jejunal<br />

morphometric study. Gut 1993; 34: 1515–1519.<br />

121. Laurin P, Wolving M, Falth-Magnusson K. Even small<br />

amounts of gluten cause relapse in children with celiac<br />

disease. J Pediatr Gastroenterol Nutr 2002; 34: 26–30.<br />

122. Fabiani E, Catassi C, Villari A et al. Dietary compliance<br />

in screening-detected coeliac disease adolescents. Acta<br />

Paediatr Suppl 1996; 412: 65–67.<br />

123. Mayer M, Greco L, Troncome R et al. Compliance of<br />

adolescents with coeliac disease with a gluten free diet.<br />

Gut 1991; 32: 881–885.<br />

124. Mariani P, Viti MG, Monturori M et al. The gluten-free<br />

diet: a nutritional risk factor for adolescents with celiac<br />

disease? J Pediatr Gastroenterol Nutr 1998; 27: 519–523.<br />

125. Ciacci C, Cirillo M, Cavallaro R, Mazzacca G. Long-term<br />

follow-up of celiac adults on gluten-free diet: prevalence<br />

and correlates of intestinal damage. Digestion<br />

2002; 66: 178–185.<br />

126. Mustalahti K, Lohiniemi S, Collin P et al. Gluten-free<br />

diet and quality of life in patients with screen-detected<br />

celiac disease. Eff Clin Pract 2002; 5: 105–113.


28<br />

Introduction<br />

Protein-losing enteropathy<br />

Jorge Amil Dias and Eunice Trindade<br />

A number of pathological conditions cause excessive<br />

leakage of protein into the gastrointestinal<br />

tract. This common mechanism has been called<br />

protein-losing enteropathy (PLE) although the site<br />

of protein leakage may also be located in the<br />

stomach.<br />

The major clinical features of these diseases are<br />

edema and hypoalbuminemia, as the intestinal<br />

loss outweighs protein synthesis, therefore causing<br />

reduced oncotic pressure in the vascular space.<br />

This mechanism is different from maldigestion or<br />

malabsorption where nutrients escape intestinal<br />

absorption, owing to enzyme deficiencies, or<br />

mucosal lesions.<br />

Historically, the pioneer work of Albright et al 1 in<br />

1949 showed that hypoalbuminemia in PLE was<br />

caused by excess catabolism of intravenous<br />

albumin rather than abnormal synthesis. Later,<br />

Citrin et al 2 demonstrated that the gastrointestinal<br />

tract was the site of the protein loss. Usually, the<br />

digestive tract (stomach or intestine) is the sole<br />

source of protein loss in PLE, although nephrotic<br />

syndrome may coexist in some cases. 3<br />

Albumin is a water-soluble molecule with molecular<br />

weight of 60000Da. This protein acts as the<br />

major component of plasma oncotic pressure and<br />

is also a transporter for various substances, such as<br />

bilirubin, ions, metals or hormones.<br />

Synthesis of albumin occurs in the liver at a rate of<br />

approximately 150mg/kg per day (higher in the<br />

first year of life: 180–300mg/kg per day). In the<br />

absence of liver disease, synthesis depends mostly<br />

on protein intake, although hormones such as<br />

cortisol, thyroid hormone and insulin, also affect<br />

the rate of synthesis. Approximately one-third of<br />

the body albumin pool circulates in the intravas-<br />

cular space. In health, less than 10% of daily<br />

albumin degradation occurs through the gastrointestinal<br />

tract. However, in conditions causing PLE<br />

there is a marked leakage of protein into the bowel<br />

lumen, without an equivalent increase in hepatic<br />

synthesis. Reasons for increased loss of protein<br />

include lymphatic obstruction, mucosal inflammation<br />

and ulceration. When the loss of protein<br />

exceeds the limited hepatic adaptation there is a<br />

decrease in the vascular pool that leads to hypoalbuminemia<br />

and edema.<br />

The loss of protein in the gastrointestinal tract is<br />

independent of molecular weight and includes<br />

albumin, immunoglobulins and ceruloplasmin;<br />

this is different from abnormal losses from the<br />

kidneys, where molecular weight determines<br />

which proteins are lost. In fact, in PLE there are<br />

also losses of minerals, such as iron, copper and<br />

calcium, of lipids and even of cells, such as<br />

lymphocytes. However, most of these proteins are<br />

readily digested in the gut and the resulting amino<br />

acids are reabsorbed by the intact segments of<br />

bowel and reused for protein synthesis. The<br />

plasma levels of different proteins in PLE depend<br />

on the adaptative capacity to increase synthesis.<br />

As mentioned before, there is little response from<br />

the liver in albumin production and the same<br />

occurs with IgG, IgM or IgA. On the other hand,<br />

the plasma levels of proteins with increased<br />

turnover, such as IgE or insulin, may be sustained<br />

despite intestinal loss.<br />

In conditions affecting the lymphatic system, such<br />

as intestinal lymphangiectasia, there may also be<br />

lymphocyte losses leading to lymphopenia. 4–6<br />

Signs and symptoms of conditions associated with<br />

PLE may reflect the cause for hypoproteinemia.<br />

However, the common link among these diseases<br />

is the loss of proteins causing hypoalbuminemia<br />

and edema. As mentioned before, albumin is the<br />

451


452<br />

Protein-losing enteropathy<br />

major element for oncotic pressure but there may<br />

be loss of other proteins. Although decrease in<br />

immunoglobulins, transferrin or ceruloplasmin<br />

may be documented by biochemical assay, rarely<br />

are there clinical manifestations from these abnormalities.<br />

If lymphatic obstruction is the major<br />

cause of PLE then malabsorption of fat may occur.<br />

Investigations<br />

Assessing protein loss from the gastrointestinal<br />

tract is not easy, because proteins secreted into the<br />

gut are readily degraded and reabsorbed.<br />

Therefore, alternative methods have been<br />

designed. One approach makes use of proteins<br />

tagged with radioactive labels injected intravenously.<br />

Several markers were used in the past<br />

(iodine, 7,8 chromium, 9,10 iron 11 and niobium 12 )<br />

bound to several proteins, usually albumin, but<br />

also dextran or ceruloplasmin. Assessment of<br />

protein loss can be made by plasma clearance or<br />

stool content of the marker. However, these studies<br />

apart from demanding several days’ collections of<br />

stool or blood, had other drawbacks. It was<br />

required that the marker should not alter the metabolic<br />

pathway of the protein, that it should not be<br />

reabsorbed from the intestine and that it should<br />

not be excreted by the glands into the digestive<br />

fluids. The radiolabels used fulfill most of these<br />

aims, but calculations of catabolic rate are<br />

complex and stools free of urine must be collected<br />

for several days. These requirements, and the<br />

questionable use of radioactive elements, made<br />

these methods unpopular in pediatrics.<br />

The other approach to diagnosis made use of a<br />

protein resistant to digestive proteolysis. Crossley<br />

and Elliott 13 suggested α1-antitrypsin for this<br />

purpose. This protein has a molecular weight of<br />

50000Da, similar to albumin, and is resistant to<br />

proteolysis, therefore fulfilling the required needs.<br />

Several studies compared this method with the<br />

radiolabel tests and confirmed that it gave reliable<br />

results. 14–16 However, α1-antitrypsin may be<br />

degraded in the stomach by acid, and therefore has<br />

limited value in the assessment of gastric losses.<br />

The method has been widely accepted for clinical<br />

purposes. The contamination of collected stools<br />

with urine is not a problem, as the protein is not<br />

eliminated in urine. For exact assessment of α1-<br />

antitrypsin enteric loss, 24–72h of stool collection<br />

may be required. Clearance is calculated by the<br />

formula:<br />

(fecal α1-antitryspin concentration)x(24-h stool volume)<br />

Clearance=<br />

(serum α1-antitryspin concentration)<br />

However, it has been proposed that random<br />

sample determination also gives reliable results for<br />

clinical use. 17,18 Magazzu et al also showed that<br />

stools may be heat-dried rather than lyophilized,<br />

with comparable results, thus adding further<br />

simplicity to the method. The normal value of α1antitrypsin<br />

is below 3–4mg/g dry stools. 14,17 In<br />

some conditions there may be overestimation of<br />

protein loss from the assessment of α1-antitrypsin:<br />

in normal newborns, α1-antitrypsin may be increased<br />

in stools probably owing to meconium<br />

clearance of the gut 19 ; gross bleeding into the<br />

gastrointestinal tract may lead to a measurable<br />

amount of the protein in the stools. 16<br />

If the protein exudation comes from abnormal<br />

lymphatics, as in intestinal lymphangiectasia,<br />

imaging techniques may help locate the lesion.<br />

Several methods, such as lymphangiography,<br />

contrast radiology and computed tomography (CT)<br />

have been used. This will be discussed below<br />

under specific conditions.<br />

Diseases causing protein-losing<br />

enteropathy<br />

The main causes are shown in Table 28.1.<br />

Diseases related to lymphatics<br />

Intestinal lymphangiectasia<br />

This disease affects males and females equally and<br />

may present from birth, although the mean age is<br />

11 years. Even prenatal diagnosis has been<br />

reported. 20 Most cases are sporadic.<br />

Several rare diseases and syndromes have associated<br />

dyplasia of lymphatics leading to PLE. These<br />

include nephrotic syndrome 3 and other rare conditions<br />

(Table 28.2). 21–23 Familial clustering suggests<br />

genetic etiology at least in some cases with early<br />

onset of manifestations. 8<br />

Patients have abnormally dilated lymphatics in the<br />

mucosa, submucosa or subserosa that cause


Table 28.1 Main causes of protein-losing<br />

enteropathy<br />

Related to lymphatics<br />

Primary<br />

intestinal lymphangiectasia<br />

Secondary<br />

cardiac disease<br />

constrictive pericarditis<br />

congestive heart failure<br />

cardiomyopathy<br />

post-Fontan procedure<br />

obstruction of lymphatics<br />

vena cava obstruction<br />

tuberculosis<br />

sarcoidosis<br />

radiation therapy<br />

retroperitoneal fibrosis or tumor<br />

Related to mucosa<br />

Gastroenteritis<br />

Crohn’s disease<br />

Celiac disease<br />

Parasitosis (e.g. Giardia)<br />

Ménétrier’s disease<br />

Eosinophilic gastritis<br />

Graft-versus-host disease<br />

Langerhan’s histiocytosis<br />

Vasculitides<br />

Systemic lupus erythematosus<br />

Connective diseases<br />

Henoch–Schönlein purpura<br />

Vena cava obstruction<br />

Related to defective cellular synthesis<br />

Deficiency of enterocyte heparan sulfate<br />

Congenital disorders of glycosylation<br />

leakage of lymphatic fluid and cells into the gut,<br />

thus causing PLE with hypoproteinemia,<br />

hypogammaglobulinemia and lymphopenia, especially<br />

depletion of CD4 T cells. 5 This may lead to<br />

abnormal delayed hypersensitivity skin tests or<br />

reduced allograft rejection tests, 4,24 but these<br />

patients do not appear to be more sensitive to<br />

infections. Hyposplenism, thymic hypoplasia<br />

and neutrophil dysfunction have also been<br />

reported. 25–27<br />

Signs and symptoms include edema that may be<br />

asymmetric, variable diarrhea and steatorrhea<br />

Diseases causing protein-losing enteropathy 453<br />

Table 28.2 Uncommon causes of proteinlosing<br />

enteropathy<br />

Noonan’s syndrome 21<br />

Congenital glaucoma<br />

Peliosis hepatis<br />

Charcot–Marie–Tooth syndrome<br />

Klippel–Trénaunay–Weber syndrome<br />

Hennekam syndrome 22<br />

Hypobetalipoproteinemia 23<br />

related to impaired fat absorption by abnormal<br />

lymphatics. Leakage of fluid into other body cavities<br />

may also cause chylous effusions such as<br />

ascites that may lead to intestinal adhesions and<br />

obstruction. Reversible blindness has been<br />

reported, due to macular edema. Tetany secondary<br />

to hypocalcemia can occur and is usually associated<br />

with severe steatorrhea. Growth restriction<br />

reflects the increased loss of protein and other<br />

elements. The presence of a lymphatic–venous<br />

anastomosis may also cause gastrointestinal bleeding.<br />

28<br />

Examination of patients may be unremarkable<br />

except for edema and malnutrition. The coexistence<br />

of hypoalbuminemia, edema, lymphopenia<br />

and steatorrhea may suggest the diagnosis. Tetany<br />

may also be present, related to hypocalcemia. Fatsoluble<br />

vitamins may be deficient.<br />

Demonstration of intestinal protein loss can be<br />

made from increased α1-antitrypsin in stools. A<br />

number of imaging techniques have been used to<br />

demonstrate the effects of abnormal intestinal<br />

lymphatics or to show localization of segmental<br />

disease. Lymphangiography may show abnormal<br />

lymphatics, absence of the thoracic duct or<br />

drainage of contrast into the bowel, but this procedure<br />

is difficult to perform in children. Contrast<br />

radiology with barium may reveal thickening of<br />

intestinal folds, or flocculation of barium due to<br />

intestinal hypersecretion. The use of radiolabeled<br />

Tc-99m incorporated in to a microcolloid may<br />

show localized accumulation of the marker on<br />

scintigraphy. CT may also show signs suggestive of<br />

intestinal lesions causing PLE. 29–31 The use of CT<br />

may even allow identification of segmental<br />

lymphangiectasia, as recently reported. 32


454<br />

Protein-losing enteropathy<br />

When the proximal intestine is involved,<br />

endoscopy may show an abnormal mucosal<br />

pattern with scattered white plaques (Figure<br />

28.1a). Diagnosis may then be confirmed from<br />

biopsy specimens showing subepithelial dilated<br />

lacteals (Figure 28.1b). Although a small intestinal<br />

biopsy would reveal these typical features, there<br />

are some limitations: the use of a biopsy capsule<br />

may miss the lesion, owing to its focal distribution,<br />

making endoscopic guidance preferable when the<br />

diagnosis is suspected; and normal histology does<br />

not preclude the diagnosis in patients on a low-fat<br />

diet or with deeper lesions not available to superficial<br />

mucosal biopsy.<br />

Treatment When disease is confined to a<br />

segment of intestine, surgery may be curative by<br />

resecting the affected part or allowing anastomosis<br />

of abnormal lymphatics to the circulatory system.<br />

Unfortunately, surgical treatment is not available<br />

(a) (b)<br />

for the majority of patients and reducing the<br />

lymphatic flow may decrease the leak into the gut.<br />

This may be achieved by reducing the intake of<br />

long-chain fatty acids. Medium-chain triglycerides<br />

(6–10 atoms of carbon) are absorbed and passed<br />

directly into the portal vein and bypass the<br />

lymphatic system. Therefore, part of the dietary<br />

lipid may be given using medium-chain triglycerides<br />

as oil or in formulas. It must be kept in<br />

mind that medium-chain triglycerides do not<br />

contain essential fatty acids and some long-chain<br />

fatty acids must be ingested regularly. The diet<br />

should contain high protein to circumvent ongoing<br />

minor losses.<br />

Dietary treatment must be permanent and guided<br />

by clinical symptoms. As mentioned above, other<br />

losses must be born in mind, and supplementation<br />

with calcium and vitamins, both water-soluble and<br />

fat-soluble must be provided. Octreotide has<br />

Figure 28.1 Intestinal lymphangiectasia. (a) Endoscopic view of duodenum showing numerous dilated lymphatics within<br />

the mucosa. (b) Histology of the small intestine of the same patient with a dilated lymphatic along the villus axis and<br />

normal surface epithelium. (Courtesy of F. Pereira).


shown effects in reducing the thoracic duct lymph<br />

flow and fluid loss in the enteric vasculature. 33<br />

This effect has been used to treat patients with<br />

considerable success. 34<br />

In the rare occurrence of gastrointestinal bleeding,<br />

the beneficial effect of the low-fat diet may be<br />

enough to reduce the vascular pressure causing<br />

hemorrhage. 28 In adults antiplasmin therapy was<br />

reported in patients with persisting bleeding 35 but<br />

this has not been used in children.<br />

Protein-losing enteropathy secondary to<br />

increased lymphatic pressure<br />

PLE may arise from increased lymphatic pressure<br />

that causes leakage of fluid and protein into the<br />

bowel. This has been widely known after the<br />

Fontan operation for complex congenital cardiac<br />

disease. In this procedure the systemic venous<br />

circulation is directly connected to the pulmonary<br />

veins, bypassing the abnormal heart; the left<br />

ventricle is the only pumping mechanism for<br />

circulation. Therefore, there is an overall increase<br />

in venous pressure and lymphatic pressure<br />

causing congestion and leakage of fluid. PLE has<br />

been reported in 2.5–10% of patients. 36 The pathophysiology<br />

of PLE in this situation is not fully<br />

known, although mechanisms have been proposed,<br />

such as raised central venous pressure and<br />

increase in mesenteric vascular resistance. 37 A<br />

longer cardiopulmonary bypass time and single<br />

right ventricle anatomy may be risk factors. 38<br />

The first symptoms may occur weeks to years after<br />

the operation and severity is variable determining<br />

morbidity and mortality. 39 Common signs and<br />

symptoms are similar to other forms of PLE.<br />

Edema is the most frequent; half of affected<br />

patients have diarrhea and one-third also have<br />

steatorrhea. Investigations reveal hypoproteinemia,<br />

hypoalbuminemia, hypogammaglobulinemia,<br />

increased α1-antitrypsin and fat in stools. There<br />

may also be dilatation of intestinal lymphatics<br />

similar to intestinal lymphangiectasia. 8 Immunological<br />

alterations have also been described,<br />

although these may be quantitative rather than<br />

qualitative. 40<br />

The therapeutic strategy in affected patients<br />

depends on the severity of each case. Diuretics,<br />

albumin infusions and nutritional supplementa-<br />

Diseases causing protein-losing enteropathy 455<br />

tion with protein and medium-chain triglycerides<br />

may provide symptomatic relief. Conversion<br />

enzyme inhibitors, e.g. captopril, reduce afterload<br />

pressure and facilitate hemodynamic improvement.<br />

Stabilization of intestinal membranes with<br />

high doses of steroids 41,42 or high molecular<br />

heparin 43–45 showed improvement in protein loss.<br />

Various vascular surgical options including<br />

cardiac transplantation have also been used. 46,47<br />

Other cardiac and vascular diseases causing elevation<br />

of systemic pressure may lead to PLE:<br />

constrictive pericarditis, restrictive cardiomyopathy,<br />

congenital pulmonary stenosis, ventricular<br />

septal defect, tricuspid regurgitation in rheumatic<br />

disease 48 and vena cava obstruction. 49<br />

Protein-losing enteropathy related to the<br />

mucosa<br />

Diseases causing extensive lesions of the gastric or<br />

intestinal mucosa may damage the mucosal barrier<br />

and allow leakage of protein. Common intestinal<br />

infections such as rotavirus gastroenteritis, giardiasis<br />

or shigellosis usually cause mild protein<br />

loss that can be documented 50–52 but the magnitude<br />

of the loss is proportional to the severity of<br />

the mucosal lesion. Crohn’s disease also causes<br />

PLE although the level of stool α1-antitrypsin does<br />

not correlate with disease activity. 53,54<br />

Patients with untreated celiac disease also may<br />

have PLE. 55 Langerhans histiocytosis usually does<br />

not involve the intestine but in rare instances may<br />

cause severe lesions in the mucosa with considerable<br />

loss of albumin (Figure 28.2). 56,57<br />

In food allergy there may be involvement of the<br />

stomach, causing PLE. 58 In these patients there are<br />

the common features of increased protein loss –<br />

edema, microcytic anemia due to iron loss, hypoalbuminemia,<br />

hypogammaglobulinemia – plus<br />

eosinophilia. Digestive symptoms may be mild and<br />

particularly related to the ingestion of the offending<br />

foods. Other signs and symptoms of allergy,<br />

such as eczema, asthma and rhinitis, as well as a<br />

positive family history may help in directing the<br />

diagnosis. Gastric and intestinal biopsy may show<br />

an eosinophilic infiltrate of the lamina propria.<br />

Patients usually recover on a restricted diet,<br />

usually cow’s milk-free.


456<br />

Protein-losing enteropathy<br />

Figure 28.2 Langerhan’s histiocytosis. Endoscopy of the<br />

duodenum revealing multiple mucosal erosions.<br />

Ménétrier’s disease<br />

Ménétrier’s disease is a particular form of gastropathy<br />

causing PLE. It is usually caused by<br />

cytomegalovirus infection, although there may be<br />

an underlying abnormality of regulation of gastric<br />

epithelial growth, as suggested by animal models<br />

and clinical evidence. 59,60 Most cases occur before<br />

10 years of age. Clinical manifestations include<br />

digestive symptoms – vomiting, abdominal pain<br />

and edema. Laboratory investigations show mild<br />

anemia and hypoalbuminemia without protein-<br />

(a) (b)<br />

uria. As mentioned above, the use of stool<br />

α1-antitrypsin to demonstrate digestive protein<br />

loss may not be used in this case because of the<br />

rapid inactivation in the acidic gastric environment.<br />

In this case it may be necessary to use radiolabeled<br />

proteins. Ultrasonographic and radiological<br />

patterns have been described to reveal the<br />

thickened gastric folds but endoscopy is the<br />

method of choice as it reveals abnormal inflamed<br />

folds covered with thick mucus and allows diagnostic<br />

biopsy (Figure 28.3).<br />

Vasculitides<br />

Among uncommon causes of PLE, syndromes due<br />

to vasculitic processes (vasculitides; see also<br />

Chapter 26) have been described. Systemic lupus<br />

erythematosus (SLE) involves various organs, but<br />

the digestive system is seldom involved. However,<br />

various cases have been reported starting in<br />

childhood. PLE may occur before the diagnosis of<br />

SLE or present years later. 61,62 The likely pathophysiology<br />

of PLE is probably immunological<br />

involving cytokines rather than mechanical<br />

disruption of lymphatics. 63,64 Clinical manifestations<br />

are similar to other forms of PLE, namely<br />

abdominal pain, diarrhea, edema and hypoalbuminemia.<br />

Diagnosis rests on demonstration of<br />

increased protein loss. Intestinal biopsy may show<br />

lymphangiectasia. Interestingly, these patients<br />

Figure 28.3 Ménétrier’s disease. (a) Endoscopic view of the stomach showing inflamed mucosa of thickened folds<br />

covered by a thick layer of viscous mucus. (b) Histology of the stomach of the same patient. There is marked elongation<br />

and tortuosity of the gastric pits with cystic dilatation of some glands. (Courtesy of F. Carneiro).


may respond to steroids without relapse after stopping<br />

treatment. In severe cases pulses of cyclophosphamide,<br />

65 plasmapheresis 66 and octreotide 67<br />

have been used.<br />

PLE has also been associated with<br />

Henoch–Schönlein purpura in as many as 1.3% of<br />

cases. 68,69 It may be underdiagnosed, especially in<br />

severe multivisceral cases.<br />

Protein-losing enteropathy related to defective<br />

cellular synthesis<br />

Recent advances in molecular biology have uncovered<br />

the etiology of many previously unclarified<br />

conditions and allowed reclassification of<br />

some diseases. 70<br />

The epithelial expression of sulfated glycosaminoglycans<br />

(GAGs) seems to be essential in the<br />

maintenance of albumin homeostasis. This expression<br />

may be compromised in inflammatory<br />

states. Congenital defects may cause absence of<br />

synthesis, 71 and congenital glycosylation defects<br />

may lead to mislocation of sulfated GAGs. 72 This<br />

group of diseases must also be considered in otherwise<br />

unexplained PLE, as the histology may be<br />

unspecific.<br />

Deficiency of enterocyte heparan sulfate<br />

Deficiency of enterocyte heparan sulfate is a rare<br />

cause of congenital severe PLE described by<br />

Murch et al71 in three patients with malabsorption,<br />

secretory diarrhea and normal intestinal histology.<br />

There was almost complete absence of sulfated<br />

GAGs in the basolateral membrane of enterocytes<br />

despite normal distribution in vascular cells and<br />

in the lamina propria. The pathophysiology is<br />

probably similar to that of congenital nephrotic<br />

syndrome, where protein loss is due to decreased<br />

negative charges of heparan sulfate. 73 The reduced<br />

expression of sulfated GAGs allows abundant loss<br />

of albumin and water due to the reduced electrostatic<br />

interaction between anion sites of GAG and<br />

arginine residues within the albumin molecules.<br />

74,75<br />

Congenital disorders of glycosylation<br />

Congenital disorders of glycosylation are multisystemic<br />

diseases with variable clinical expression<br />

Diseases causing protein-losing enteropathy 457<br />

and several subtypes. The common feature is the<br />

alteration of N-glycosylation which may be defective<br />

in several points. This is a fundamental<br />

mechanism that regulates protein folding and<br />

allows intracellular quality control. 76,77 Most<br />

secretory and membrane proteins require glycosylation<br />

for proper folding and subsequent transport<br />

via the secretory pathway. In congenital disorders<br />

of glycosylation there is underglycosylation of<br />

several proteins including antithrombin III, factor<br />

IX or protein C, leading to cytoplasmic retention of<br />

the abnormal protein. The common screening test<br />

is identification of carbohydrate transferrin deficiency<br />

by isoelectric focusing analysis of serum<br />

transferrin. It is likely that the high turnover rate of<br />

epithelial cells may render them particularly<br />

vulnerable to this type of disease. 72 PLE is<br />

common in congenital disorders of glycosylation<br />

type 1b (mutation of phosphomannose isomerase)<br />

and type 1c (mutation of α-1,3-glucosyltransferase).<br />

72,78 Patients with type 1b have severe<br />

diarrhea, vomiting and PLE. Intestinal biopsy<br />

shows partial villous atrophy, and the enterocyte<br />

endoplasmic reticulum is distended and engorged<br />

with precipitated abnormal protein. Mannose is<br />

effective in the treatment of these patients,<br />

correcting both symptoms and biochemical abnormalities.<br />

79,80<br />

Patients with congenital disorders of glycosylation<br />

type 1c have variable clinical expression and may<br />

present features of types 1a and 1b. Westphal et al<br />

described one patient with recurrent PLE after<br />

acute gastroenteritis starting at the age of 3<br />

months. Intestinal biopsies showed marked reduction<br />

of epithelial heparan sulfate in the basolateral<br />

membrane during exacerbations and normalization<br />

in remission. 72 It is probable that increased<br />

cellular turnover during exacerbations cause<br />

intracellular retention rather than defective<br />

synthesis. The authors speculated that the already<br />

inefficient glycosylation was overwhelmed by the<br />

increased epithelial turnover in gastroenteritis,<br />

effectively leading to complete loss of heparan<br />

sulfate in the small-intestinal epithelium. Heparin<br />

may have a role in the stabilization of the cellular<br />

membrane.


458<br />

Protein-losing enteropathy<br />

REFERENCES<br />

1. Albright F, Bartter FC, Forbes AP. The fate of human<br />

serum albumin administered intravenously to a patient<br />

with idiopathic hypoalbuminemia and hypoglobulinemia.<br />

Trans Assoc Am Phys 1949; 62: 204<br />

2. Citrin Y, Sterling K, Halsted J. Mechanisms of hypoproteinemia<br />

associated with giant hypertrophy of gastric<br />

mucosa. N Engl J Med 1957; 257: 906–912<br />

3. de Sousa JS, Guerreiro O, Cunha A et al. Association of<br />

nephrotic syndrome with intestinal lymphangiectasia.<br />

Arch Dis Child 1968; 43: 245–248.<br />

4. Weiden PL, Blaese RM, Strober W et al. Impaired<br />

lymphocyte transformation in intestinal lymphangiectasia:<br />

evidence for at least two functionally<br />

distinct lymphocyte populations in man. J Clin Invest<br />

1972; 51: 1319–1325.<br />

5. Fuss IJ, Strober W, Cuccherini BA et al. Intestinal<br />

lymphangiectasia, a disease characterized by selective<br />

loss of naive CD45RA+ lymphocytes into the gastrointestinal<br />

tract. Eur J Immunol 1998; 28: 4275–4285.<br />

6. Arato A, Savilahti E, Balogh L. The distribution of<br />

lymphocyte subpopulations in an infant with primary<br />

intestinal lymphangiectasia. Acta Paediatr Hung 1992;<br />

32: 309–317.<br />

7. Gordon RSJ. Exudative enteropathy: abnormal<br />

permeability of the gastrointestinal tract demonstrable<br />

with labelled polyvinylpyrrolidone. Lancet 1959; 1:<br />

325–326.<br />

8. Waldmann TA. Protein-losing gastroenteropathies. In<br />

Haubrich WS, Kalser MA, Roth, JL et al., eds. Bockus<br />

Gastroenterology, vol 4. Philadelphia: WB Saunders,<br />

1985: 1814–1837.<br />

9. van Tongeren JH, Reichert WJ. Demonstration of<br />

protein-losing gastroenteropathy. The quantitative<br />

estimation of gastrointestinal protein loss, using<br />

51 Cr-labelled plasma proteins. Clin Chim Acta 1966; 14:<br />

42–48.<br />

10. Waldmann TA, Wochner RD, Strober W. The role of the<br />

gastrointestinal tract in plasma protein metabolism.<br />

Studies with 51Cr-albumin. Am J Med 1969; 46:<br />

275–285.<br />

11. Jarnum S, Westergaard H, Yssing M et al. Quantitation<br />

of gastrointestinal protein loss by means of FE59labeled<br />

iron dextran. Gastroenterology 1968; 55:<br />

229–241.<br />

12. Jeejeebhoy KN, Jarnum S, Singh B et al. 95Nb-labelled albumin for the study of gastrointestinal albumin loss.<br />

Scand J Gastroenterol 1968; 3: 449–457.<br />

13. Crossley JR, Elliott RB. Simple method for diagnosing<br />

protein-losing enteropathies. Br Med J 1977; 1: 428–429.<br />

14. Bernier JJ, Florent C, Desmazures C et al. Diagnosis of<br />

protein-losing enteropathy by gastrointestinal clearance<br />

of alpha1-antitrypsin. Lancet 1978; 2: 763–764.<br />

15. Hill RE, Hercz A, Corey ML et al. Fecal clearance of<br />

alpha 1-antitrypsin: a reliable measure of enteric protein<br />

loss in children. J Pediatr 1981; 99: 416–418.<br />

16. Thomas DW, Sinatra FR, Merritt RJ. Random fecal<br />

alpha-1-antitrypsin concentration in children with<br />

gastrointestinal disease. Gastroenterology 1981; 80:<br />

776–782.<br />

17. Magazzu G, Jacono G, Di Pasquale G et al. Reliability<br />

and usefulness of random fecal alpha 1-antitrypsin<br />

concentration: further simplification of the method. J<br />

Pediatr Gastroenterol Nutr 1985; 4: 402–407.<br />

18. Catassi C, Cardinali E, D’Angelo G et al. Reliability of<br />

random fecal alpha 1-antitrypsin determination on<br />

nondried stools. J Pediatr 1986; 109: 500–502.<br />

19. Keller KM, Knobel R, Ewe K. Fecal alpha 1-antitrypsin<br />

in newborn infants. J Pediatr Gastroenterol Nutr 1997;<br />

24: 271–275.<br />

20. Schmider A, Henrich W, Reles A et al. Isolated fetal<br />

ascites caused by primary lymphangiectasia: a case<br />

report. Am J Obstet Gynecol 2001; 184: 227–228.<br />

21. Vallet HL, Holtzapple PG, Eberlein WR et al. Noonan<br />

syndrome with intestinal lymphangiectasis. A metabolic<br />

and anatomic study. J Pediatr 1972; 80: 269–274.<br />

22. Van Balkom ID, Alders M, Allanson J et al.<br />

Lymphedema-lymphangiectasia-mental retardation<br />

(Hennekam) syndrome: a review. Am J Med Genet 2002;<br />

112: 412–421.<br />

23. Dobbins WO 3rd. Hypo-beta-lipoproteinemia and<br />

intestinal lymphangiectasia. A new syndrome of malabsorption<br />

and protein-losing enteropathy. Arch Intern<br />

Med 1968; 122: 31–38.<br />

24. Strober W, Wochner RD, Carbone PP et al. Intestinal<br />

lymphangiectasia: a protein-losing enteropathy with<br />

hypogammaglobulinemia, lymphocytopenia and<br />

impaired homograft rejection. J Clin Invest 1967; 46:<br />

1643–1656.<br />

25. Foster PN, Bullen AW, Robertson DA et al. Development<br />

of impaired splenic function in intestinal lymphangiectasia.<br />

Gut 1985; 26: 861–864.<br />

26. Sorensen RU, Halpin TC, Abramowsky CR et al.<br />

Intestinal lymphangiectasia and thymic hypoplasia. Clin<br />

Exp Immunol 1985 59: 217–226.<br />

27. Bolton RP, Cotter KL, Losowsky MS. Impaired<br />

neutrophil function in intestinal lymphangiectasia. J<br />

Clin Pathol 1986; 39: 876–880.<br />

28. Perisic VN, Kokai G. Bleeding from duodenal lymphangiectasia.<br />

Arch Dis Child 1991; 66: 153–154.<br />

29. Fakhri A, Fishman EK, Jones B et al. Primary intestinal<br />

lymphangiectasia: clinical and CT findings. J Comput<br />

Assist Tomogr 1985; 9: 767–770.<br />

30. Puri AS, Aggarwal R, Gupta RK et al. Intestinal<br />

lymphangiectasia: evaluation by CT and scintigraphy.<br />

Gastrointest Radiol 1992; 17: 119–121.<br />

31. Stevens RL, Jones B, Fishman EK. The CT halo sign: a<br />

new finding in intestinal lymphangiectasia. J Comput<br />

Assist Tomogr 1997; 21: 1005–1007.<br />

32. Yang DM, Jung DH. Localized intestinal lymphangiectasia:<br />

CT findings. Am J Roentgenol 2003; 180:<br />

213–214.<br />

33. Nakabayashi H, Sagara H, Usukura N et al. Effect of<br />

somatostatin on the flow rate and triglyceride levels of<br />

thoracic duct lymph in normal and vagotomized dogs.<br />

Diabetes 1981; 30: 440–445.<br />

34. Ballinger AB, Farthing MJ. Octreotide in the treatment<br />

of intestinal lymphangiectasia. Eur J Gastroenterol<br />

Hepatol 1998; 10: 699–702.<br />

35. MacLean JE, Cohen E, Weinstein M. Primary intestinal<br />

and thoracic lymphangiectasia: a response to antiplasmin<br />

therapy. Pediatrics 2002; 109: 1177–1180.<br />

36. Mertens L, Hagler DJ, Sauer U et al. Protein-losing<br />

enteropathy after the Fontan operation: an international<br />

multicenter study. PLE Study Group. J Thorac<br />

Cardiovasc Surg 1998; 115: 1063–1073.<br />

37. Rychik J, Spray TL. Strategies to treat protein-losing<br />

enteropathy. Semin Thorac Cardiovasc Surg Pediatr Card<br />

Surg Annu 2002; 5: 3–11.<br />

38. Powell AJ, Gauvreau K, Jenkins KJ et al. Perioperative<br />

risk factors for development of protein-losing<br />

enteropathy following a Fontan procedure. Am J Cardiol<br />

2001; 88: 1206–1209.


39. Marino BS. Outcomes after the Fontan procedure. Curr<br />

Opin Pediatr 2002; 14: 620–626.<br />

40. Cheung YF, Tsang HY, Kwok JS. Immunologic profile of<br />

patients with protein-losing enteropathy complicating<br />

congenital heart disease. Pediatr Cardiol 2002 23:<br />

587–593.<br />

41. Rychik J, Piccoli DA, Barber G. Usefulness of corticosteroid<br />

therapy for protein-losing enteropathy after<br />

the Fontan procedure. Am J Cardiol 1991; 68: 819–821.<br />

42. Zellers TM, Brown K. Protein-losing enteropathy after<br />

the modified fontan operation: oral prednisone<br />

treatment with biopsy and laboratory proved<br />

improvement. Pediatr Cardiol 1996; 17: 115–117.<br />

43. Donnelly JP, Rosenthal A, Castle VP et al. Reversal of<br />

protein-losing enteropathy with heparin therapy in<br />

three patients with univentricular hearts and Fontan<br />

palliation. J Pediatr 1997; 130: 474–478.<br />

44. Kelly AM, Feldt RH, Driscoll DJ et al. Use of heparin in<br />

the treatment of protein-losing enteropathy after fontan<br />

operation for complex congenital heart disease. Mayo<br />

Clin Proc 1998; 73: 777–779.<br />

45. Facchini M, Guldenschuh I, Turina J et al. Resolution of<br />

protein-losing enteropathy with standard high<br />

molecular heparin and urokinase after Fontan repair in<br />

a patient with tricuspid atresia. J Cardiovasc Surg<br />

(Torino) 2000; 41: 567–570.<br />

46. Rychik J. Management of protein-losing enteropathy<br />

after the Fontan procedure. Semin Thorac Cardiovasc<br />

Surg Pediatr Card Surg Annu 1998; 1: 15–22.<br />

47. Holmgren D, Berggren H, Wahlander H et al. Reversal of<br />

protein-losing enteropathy in a child with Fontan<br />

circulation is correlated with central venous pressure<br />

after heart transplantation. Pediatr Transplant 2001; 5:<br />

135–137.<br />

48. Nelson DL, Blaese RM, Strober W et al. Constrictive<br />

pericarditis, intestinal lymphangiectasia, and reversible<br />

immunologic deficiency. J Pediatr 1975; 86: 548–554.<br />

49. Lee WS, John P, McKiernan P et al. Inferior vena cava<br />

occlusion and protein-losing enteropathy after liver<br />

transplantation in children. J Pediatr Gastroenterol Nutr<br />

2002; 34: 413–416.<br />

50. Maki M, Harmoinen A, Vesikari T et al. Faecal excretion<br />

of alpha-1-antitrypsin in acute diarrhoea. Arch Dis Child<br />

1982; 57: 154–156.<br />

51. Sutton DL, Kamath KR. Giardiasis with protein-losing<br />

enteropathy. J Pediatr Gastroenterol Nutr 1985 4: 56–59.<br />

52. Bennish ML, Salam MA, Wahed MA. Enteric protein<br />

loss during shigellosis. Am J Gastroenterol 1993; 88:<br />

53–57.<br />

53. Grill BB, Hillemeier AC, Gryboski JD. Fecal alpha 1antitrypsin<br />

clearance in patients with inflammatory<br />

bowel disease. J Pediatr Gastroenterol Nutr 1984; 3:<br />

56–61.<br />

54. Karbach U, Ewe K, Bodenstein H. Alpha 1-antitrypsin, a<br />

reliable endogenous marker for intestinal protein loss<br />

and its application in patients with Crohn’s disease. Gut<br />

1983; 24: 718–723.<br />

55. Dinari G, Rosenbach Y, Zahavi I et al. Random fecal<br />

alpha 1-antitrypsin excretion in children with intestinal<br />

disorders. Am J Dis Child 1984; 138: 971–973.<br />

56. Boccon-Gibod LA, Krichen HA, Carlier-Mercier LA et al.<br />

Digestive tract involvement with exudative enteropathy<br />

in Langerhans cell histiocytosis. Pediatr Pathol 1992; 12:<br />

515–524.<br />

57. Santos-Machado TM, Cristofani LM, Almeida MT et al.<br />

Disseminated Langerhans’ cell histiocytosis and massive<br />

protein-losing enteropathy. Braz J Med Biol Res 1999;<br />

32: 1095–1099.<br />

References 459<br />

58. Waldmann TA, Wochner RD, Laster L et al. Allergic<br />

gastroenteropathy. A cause of excessive gastrointestinal<br />

protein loss. N Engl J Med 1967; 276: 762–769.<br />

59. Dempsey PJ, Goldenring JR, Soroka CJ et al. Possible<br />

role of transforming growth factor alpha in the<br />

pathogenesis of Menetrier’s disease: supportive<br />

evidence from humans and transgenic mice.<br />

Gastroenterology 1992; 103: 1950–1963.<br />

60. Sferra TJ, Pawel BR, Qualman SJ et al. Menetrier disease<br />

of childhood: role of cytomegalovirus and transforming<br />

growth factor alpha. J Pediatr 1996; 128: 213–219.<br />

61. Molina JF, Brown RF, Gedalia A et al. Protein losing<br />

enteropathy as the initial manifestation of childhood<br />

systemic lupus erythematosus. J Rheumatol 1996; 23:<br />

1269–1271.<br />

62. Tsukahara M, Matsuo K, Kojima H. Protein-losing<br />

enteropathy in a boy with systemic lupus<br />

erythematosus. J Pediatr 1980; 97: 778–780.<br />

63. Edworthy SM, Fritzler MJ, Kelly JK et al. Protein-losing<br />

enteropathy in systemic lupus erythematosus associated<br />

with intestinal lymphangiectasia. Am J Gastroenterol<br />

1990; 85: 1398–1402.<br />

64. Yazici Y, Erkan D, Levine DM et al. Protein-losing<br />

enteropathy in systemic lupus erythematosus: report of<br />

a severe, persistent case and review of pathophysiology.<br />

Lupus 2002; 11: 119–123.<br />

65. Lee CK, Han JM, Lee KN et al. Concurrent occurrence of<br />

chylothorax, chylous ascites, and protein-losing<br />

enteropathy in systemic lupus erythematosus. J<br />

Rheumatol 2002; 29: 1330–1333.<br />

66. Garcia-Consuegra J, Merino R, Alonso A et al. Systemic<br />

lupus erythematosus: a case report with unusual<br />

manifestations and favourable outcome after plasmapheresis.<br />

Eur J Pediatr 1992; 151: 581–582.<br />

67. Ossandon A, Bombardieri M, Coari G et al. Protein<br />

losing enteropathy in systemic lupus erythematosus:<br />

role of diet and octreotide. Lupus 2002; 11: 465–466.<br />

68. Reif S, Jain A, Santiago J et al Protein losing enteropathy<br />

as a manifestation of Henoch–Schonlein purpura,<br />

Acta Paediatr Scand 1991; 80: 482–485.<br />

69. Barajas DF, Bravo MB, Palomino UN et al. Familial<br />

hypouricemia due to an isolated tubular defect of urate<br />

reabsorption. Pediatr Nephrol 1993; 7: 83–85.<br />

70. Murch SH. Toward a molecular understanding of<br />

complex childhood enteropathies. J Pediatr<br />

Gastroenterol Nutr 2002; 34 (Suppl 1): S4–S10.<br />

71. Murch SH, Winyard PJ, Koletzko S et al. Congenital<br />

enterocyte heparan sulphate deficiency with massive<br />

albumin loss, secretory diarrhoea, and malnutrition.<br />

Lancet 1996; 347: 1299–1301.<br />

72. Westphal V, Murch S, Kim S et al. Reduced heparan<br />

sulfate accumulation in enterocytes contributes to<br />

protein-losing enteropathy in a congenital disorder of<br />

glycosylation. Am J Pathol 2000; 157: 1917–1925.<br />

73. Vernier RL, Klein DJ, Sisson SP et al. Heparan sulfaterich<br />

anionic sites in the human glomerular basement<br />

membrane. Decreased concentration in congenital<br />

nephrotic syndrome. N Engl J Med 1983; 309:<br />

1001–1009.<br />

74. Powers MR, Blumenstock FA, Cooper JA et al. Role of<br />

albumin arginyl sites in albumin-induced reduction of<br />

endothelial hydraulic conductivity. J Cell Physiol 1989;<br />

141: 558–564.<br />

75. Kanwar YS, Linker A, Farquhar MG. Increased<br />

permeability of the glomerular basement membrane to<br />

ferritin after removal of glycosaminoglycans (heparan<br />

sulfate) by enzyme digestion. J Cell Biol 1980; 86:<br />

688–693.


460<br />

Protein-losing enteropathy<br />

76. Aebi M, Hennet T. Congenital disorders of glycosylation:<br />

genetic model systems lead the way. Trends Cell<br />

Biol 2001; 11: 136–141.<br />

77. Helenius A, Aebi M. Intracellular functions of N-linked<br />

glycans. Science 2001; 291: 2364–2369.<br />

78. de Lonlay P, Seta N, Barrot S et al. A broad spectrum of<br />

clinical presentations in congenital disorders of<br />

glycosylation I: a series of 26 cases. J Med Genet 2001;<br />

38: 14–19.<br />

79. Freeze HH. Disorders in protein glycosylation and<br />

potential therapy: tip of an iceberg? J Pediatr 1998; 133:<br />

593–600.<br />

80. Harms HK, Zimmer KP, Kurnik K et al. Oral mannose<br />

therapy persistently corrects the severe clinical<br />

symptoms and biochemical abnormalities of phosphomannose<br />

isomerase deficiency. Acta Paediatr 2002; 91:<br />

1065–1072.


29<br />

Introduction<br />

Short-bowel syndrome<br />

Olivier Goulet<br />

Short-bowel syndrome is characterized by a state<br />

of malabsorption following extensive resection of<br />

the small bowel. It is a functional rather than an<br />

anatomical definition. The resection results in a<br />

state of insufficient nutritive supply requiring artificial<br />

nutrition. Until 20 years ago, the prognosis<br />

after extensive bowel resection was poor, especially<br />

in the neonatal period. The onset of<br />

parenteral nutrition (PN) and enteral feeding in the<br />

daily practice has transformed the outcome during<br />

the past two decades. 1–4 These methods allow<br />

infants and children with short-bowel syndrome to<br />

grow normally during the long period required for<br />

adaptation of the remaining small intestine. In<br />

addition to the requirement for PN, the child with<br />

intestinal failure from short-bowel syndrome may<br />

benefit from other established medical and surgical<br />

interventions, intended to improve the function<br />

of the remaining gut. 5 This chapter reviews<br />

the pathophysiology of short-bowel syndrome and<br />

describes the principles of its medical and surgical<br />

management.<br />

Consequences of intestinal resection<br />

The functional consequences of short-bowel<br />

syndrome depend on the length, surface and site of<br />

the resected small intestine (Table 29.1). The cause<br />

of resection and the age of the patient at the time<br />

at which surgery was carried out also influence the<br />

capacity of the remaining gut function and potential<br />

for adaptation. At birth, the small-bowel length<br />

is 250±40cm, and the increase in length is<br />

maximal during the first year of life. 6 The smallbowel<br />

length doubles during the last trimester of<br />

gestation, suggesting that a short-bowel remnant<br />

does not have the same prognosis in a preterm<br />

infant as in a full-term baby. 7,8 The same difference<br />

exists between neonates and older children.<br />

Despite this potential for growth and other factors<br />

determining the outcome, it is classical to consider<br />

three levels after small-bowel resection. According<br />

to the length of the small intestine measured along<br />

the anti-mesenteric border at surgery, short resection<br />

leaves more than 100–150cm of small intestine,<br />

large resection leaves between 40 and 100cm<br />

and massive resection less than 40cm. In all cases,<br />

one should consider the age of the patient at the<br />

time or resection, the portion of small bowel<br />

resected and the functional integrity of the remaining<br />

small intestine.<br />

Resection of the jejunum<br />

Both the transit time and the direct contact of<br />

intraluminal nutrients with the jejunal epithelium<br />

determine the malabsorption syndrome following<br />

Table 29.1 Consequences of intestinal<br />

resection<br />

Malabsorption (nutrients, vitamins, minerals) 1–3<br />

Gastric acid hypersecretion 4<br />

Pancreatic insufficiency 4<br />

Water and electrolyte losses 1–4<br />

Hyperoxaluria 1–3<br />

Biliary lithiasis 1, 3, 5<br />

Liver disease 1, 3, 5<br />

Bone disease 5<br />

The numbers refer to the following: 1, disruption of<br />

the enterohepatic cycle; 2, reduction of the absorptive<br />

surface; 3, bacterial overgrowth; 4, gastric acid<br />

hypersecretion; 5, long-term parenteral nutrition<br />

461


462<br />

Short-bowel syndrome<br />

extensive jejunal resection. The resulting malabsorption<br />

concerns all nutrients as well as minerals,<br />

electrolytes, trace elements and most of the vitamins.<br />

Severe diarrhea following extensive jejunal<br />

resection is associated with steatorrhea and creatorrhea.<br />

The degree of malabsorption is proportional<br />

to the length of jejunum resected and will be<br />

compensated, to some extent, by the ileum and/or<br />

by the process of adaptation in response to loss of<br />

intestinal surface.<br />

Resection of the ileum<br />

Despite the fact that, normally, most nutrients are<br />

absorbed in the proximal jejunum, the residual<br />

ileum is able to adapt and to assume the role of<br />

macronutrient absorption. However, the specialized<br />

cells of the terminal ileum, where vitamin B12/<br />

intrinsic factor receptors are located and where<br />

bile salts are reabsorbed, cannot be replaced by<br />

jejunal hypertrophy. Thus, the ileum has specific<br />

functions which the jejunum cannot substitute. In<br />

addition, resection of the distal ileum usually<br />

includes the ileocecal valve (ICV). Finally, ileal<br />

resection impairs vitamin B12 absorption which<br />

can cause macrocytic anemia and neuropathy.<br />

Malabsorption of bile salts is responsible for<br />

specific complications:<br />

(1) Secretory diarrhea may be severe and is<br />

related to the presence in excess of bile salts<br />

within the colon, causing injury to the<br />

colonic mucosa. The consequences are<br />

proportional to the concentration and dehydroxylation<br />

of bile salts into the lumen<br />

(deoxycholic and chenodeoxycholic acids). 9<br />

(2) The decrease of bile salt reabsorption by the<br />

ileum reduces their circulating pool and leads<br />

to lipid malabsorption and steatorrhea. 10<br />

Unabsorbed fatty acids are then hydroxylated<br />

by colonic bacteria increasing mucosal injury<br />

and secretory diarrhea. Because of the<br />

decreased pool of bile salts, fat-soluble<br />

vitamins (A, D, E and K) are also prone to be<br />

malabsorbed.<br />

(3) Cholelithiasis seems to be the direct consequence<br />

of the reduced concentration of bile<br />

salts in bile. 11,12 Resection of the terminal<br />

ileum, by disrupting the enterohepatic circu-<br />

lation of biliary acids, increases the lithogenicity<br />

of bile. Premature infants are especially<br />

exposed to cholelithiasis because of<br />

their low production of conjugated bile<br />

acids. 13<br />

(4) Hyperoxaluria with formation of renal stones<br />

results from loss of the ileum and subsequent<br />

lipid malabsorption. 14 This complication<br />

became rare with the increased use of diets<br />

rich in medium-chain triglycerides (MCTs)<br />

(see below).<br />

Finally, extensive loss of the ileum reduces transit<br />

time by suppressing the so-called ‘ileal brake’. It<br />

has been shown that the ileum has a greater potential<br />

for adaptation than the jejunum. 15 In<br />

addition, the ileum is the site of release of enteric<br />

hormones such as enteroglucagon, which are<br />

essential in the process of adaptation after extensive<br />

resection (see below).<br />

Ileocecal valve resection<br />

The resection of the ICV decreases transit time<br />

(ileocecal brake) and allows colonic bacteria to<br />

enter and populate the small intestine. Bacterial<br />

overgrowth may negatively impact on digestion<br />

and nutrient absorption, as bacteria compete for<br />

nutrients with enterocytes. Thus, ICV resection<br />

represents an additional major cause of malabsorption<br />

of nutrients, water and electrolytes, dehydroxylation<br />

of bile salts, mucosal injury and<br />

motility disorders. 16,17 The lack of the ICV appears<br />

greatly to influence the period required to achieve<br />

intestinal autonomy following efficient smallbowel<br />

adaptation. 18–20 In addition, ICV resection<br />

increases the risk for sepsis of intestinal origin.<br />

This occurred three times more frequently in<br />

infants without the ICV than in those with an<br />

intact cecum. 1<br />

Associated disorders<br />

Beside the anatomical impairment and its consequences,<br />

massive small-bowel resection may be<br />

aggravated by several associated disorders.<br />

(1) Gastric acid hypersecretion occurs in 50% or<br />

pediatric patients with short-bowel syndrome


while hypergastrinemia is inconstant. 21 Acid<br />

hypersecretion occurs early after resection and<br />

depends on the extent of the resection.<br />

Hypersecretion is transitory, but it increases<br />

with enteral feeding, leading to a larger<br />

amount or intestinal fluid loss. Gastric acid<br />

hypersecretion, by reducing the duodenal pH,<br />

decreases the activity of pancreatic enzymes<br />

such as amylase and lipase, which, in turn,<br />

increases fat malabsorption.<br />

(2) Gastric emptying of liquids is more rapid<br />

following jejunal resection, although intestinal<br />

transit may still remain normal, because of<br />

the braking effect of the ileum. 22 The loss of<br />

inhibition on gastric emptying and intestinal<br />

transit in children without colon is related to<br />

a significant decrease in peptide YY, glucagonlike<br />

peptide I (GLP-I) and neurotensin. 23<br />

Peptide YY is normally released from L cells<br />

in the ileum and colon when stimulated by fat<br />

or bile salts. These cells are missing in<br />

patients who have undergone distal ileal and<br />

colonic resection. Rapid gastric emptying may<br />

contribute to fluid losses in children with<br />

short-bowel syndrome.<br />

(3) Alteration in gut motor activity may be<br />

observed, especially in case of prenatal smallbowel<br />

malformations (atresia) or severe postnatal<br />

pathology such as extensive necrotizing enterocolitis.<br />

They contribute together with repeated<br />

surgical procedures, by increasing plastic peritonitis,<br />

to impair motor activity leading to bacterial<br />

overgrowth and the above-mentioned complications.<br />

These particular patients are at highest<br />

risk of developing rapid and severe liver disease<br />

that impairs the intestinal adaptation process<br />

and may require combined liver–intestine transplantation<br />

(see below).<br />

(4) Colonic and/or gastric resection is sometimes<br />

associated with small-bowel resection. The<br />

former aggravates water and electrolyte losses<br />

while the latter, by altering duodenal contents,<br />

further decreases intestinal absorption.<br />

Finally, the main factors determining the outcome<br />

of short-bowel syndrome are the length of the<br />

intestinal remnant, whether this includes the<br />

ileum or not, the conservation of the ICV and the<br />

degree of injury or the functional integrity of the<br />

remaining small intestine.<br />

Small-bowel adaptation after extensive intestinal resection 463<br />

Small-bowel adaptation after<br />

extensive intestinal resection<br />

The goal of intestinal adaptation after massive<br />

small-bowel resection is to develop the ability to<br />

withdraw artificial nutrition thanks to a compensatory<br />

increase in the mucosal surface area and<br />

absorption capacity. Adaptation is a slow process<br />

accompanied by a gradual increase in the<br />

absorption capacity of nutrients, electrolytes and<br />

minerals. 24,25 Soon after resection, the physiological<br />

process of adaptation of the remaining<br />

small-bowel develops. 26 This comprises muscular<br />

hypertrophy (increased bowel diameter and wall<br />

thickness) and hyperplasia of the intestinal<br />

mucosa. This mucosal hyperplasia is characterized<br />

by an increased number of enterocytes per unit of<br />

small-bowel length, an increased rate of enterocyte<br />

proliferation and an increased villous height and<br />

crypt depth. 27 In animals, it was demonstrated that<br />

epithelial hyperplasia following gut resection<br />

resulted in increased mucosal mass, including<br />

higher mucosal wet weight, higher protein content<br />

as well as higher DNA and RNA content per unit<br />

of bowel length. 28 The complex regulation of gut<br />

mucosal growth involves a multitude of factors<br />

including hormonal mediators such as<br />

enteroglucagon, glucagon-like peptides, neurotensin,<br />

peptide YY, growth hormone and insulinlike<br />

growth factor (IGF). 28–32 Additionally, oral or<br />

enteral feeding stimulates the release of<br />

enterotrophic hormones such as gastrin, cholecystokinin<br />

(CCK) and neurotensin, which may<br />

further improve the process of gut adaptation. 29–32<br />

Intraluminal substrates and nutrients, provided by<br />

oral or enteral feeding, are essential for achieving<br />

intestinal adaptation after extensive resection.<br />

Intraluminal nutrients stimulate the adaptation<br />

of the intestinal mucosa through several<br />

mechanisms:<br />

(1) Direct contact of intraluminal nutrients with<br />

intestinal cells; 33,34<br />

(2) Trophic effects of gastrointestinal secretions<br />

enhanced by food; 35,36<br />

(3) Release of several trophic hormones secreted<br />

by the gastrointestinal tract, mainly enteroglucagon.<br />

37<br />

The postulated influence of CCK and secretin<br />

might be an indirect action via the stimulation of


464<br />

Short-bowel syndrome<br />

pancreatic secretions that could lead to the release<br />

of enteroglucagon. 38 In turn, enteroglucagon stimulates<br />

the cell turnover, motility and absorptive<br />

capacity of the small intestine. 39<br />

Glutamine (Gln) is the most important circulating<br />

amino acid. Many findings have emphasized the<br />

role of Gln for the metabolism of enterocytes<br />

suggesting the importance of Gln as a substrate<br />

during PN in patients with short-bowel<br />

syndrome. 40–43<br />

Ornithine-α-ketoglutarate has been shown to<br />

improve mucosal trophicity and to enhance<br />

intestinal adaptation in a rat model of extensive<br />

intestinal resection. 44 Ornithine-α-ketoglutarate is<br />

a precursor of glutamine and could also act by an<br />

increased production of polyamines by the enterocytes.<br />

Short-chain fatty acids (SCFA) are produced from<br />

the fermentation of fibers by the colonic<br />

microflora. SCFA could be energetic substrates for<br />

the topical nutrition of intestinal cells. 45,46 In rats<br />

with short-bowel syndrome, pectin, a watersoluble,<br />

non-cellulose dietary fiber, was shown to<br />

enhance jejunal and colonic mucosal adaptation<br />

when added to the enteral diet 47 or PN mixture. 48<br />

The question as to whether patients with shortbowel<br />

syndrome would benefit from diets<br />

enriched in pectin remains unanswered.<br />

Besides nutrients, other factors seem to play an<br />

important role in the mucosal adaptation process:<br />

(1) Growth-stimulating peptide has been shown<br />

to be synthesized in the rat proximal intestine<br />

after small-bowel resection; 49<br />

(2) Epidermal growth factor (EGF), produced in<br />

the salivary glands and duodenal cells, is a<br />

trophic substance for the gastrointestinal<br />

tract. 50–52 EGF is a polypeptide able to stimulate<br />

[ 3 H] thymidine incorporation into<br />

intestinal DNA when given at pharmacological<br />

doses to mice. 53 ln addition, EGF<br />

infusion in rats is associated with a rise in<br />

galactose as well as in glycine absorption; 54<br />

(3) Polyamines (spermine, spermidine), whose<br />

synthesis is dependent on ornithine decarboxylase<br />

activity, greatly enhance intestinal<br />

cell turnover and protein synthesis; 55,56<br />

(4) Exogenous prostaglandin (16,16-dimethyl<br />

prostaglandin E1) has been shown to stimulate<br />

mucosal hyperplasia in the gastric<br />

antrum and in the jejunum. 57,58<br />

Thus, adaptation of the remaining small bowel is<br />

dependent on exogenous and endogenous factors,<br />

emphasizing the usefulness of early enteral<br />

feeding after resection. It is hoped that in the<br />

future the utilization of exogenous trophic<br />

peptides will further enhance and optimize<br />

adaptation of the remaining intestine.<br />

Clinical management of short-bowel<br />

syndrome<br />

Initial surgery<br />

Every time intestinal resection is performed for an<br />

acute event such as midgut volvulus, the initial<br />

surgery should aim at saving as much intestinal<br />

tissue as possible. In some situations, a so-called<br />

‘second-look procedure’ can be performed 2–3<br />

days later if the patient’s condition permits it. The<br />

intestinal segments which have not recovered<br />

from the initial procedure can then be resected. In<br />

the case of intestinal atresia with a very dilated<br />

proximal loop, performing an enterostomy is<br />

indicated in order to avoid intestinal stasis and<br />

subsequent bacterial overgrowth. If an end-to-end<br />

anastomosis is performed, the proximal intestinal<br />

loop should undergo tapering anti-mesenteric<br />

enteroplasty.<br />

When small-bowel resection includes the terminal<br />

ileum it is recommended that a gallbladder resection<br />

be performed. 12 We perform this whenever<br />

an extensive ileum resection is made in order to<br />

prevent the occurrence of cholelithiasis.<br />

The question as to whether a gastrostomy should<br />

be performed at the initial phase depends on the<br />

experience of the team. Nevertheless, if long-term<br />

enteral nutrition is planned, a gastrostomy is better<br />

for the patient’s comfort. Extensive small-bowel<br />

resection will require fluid replacement after<br />

surgery (see below) and, in a number of patients,<br />

long-term PN. Therefore, a permanent indwelling<br />

central venous catheter must be placed, such as a<br />

Broviac or Hickman catheter.


Medical therapy<br />

Whatever the etiology of the short-bowel<br />

syndrome (Table 29.2), it is customary and informative<br />

to consider three phases in the clinical<br />

course after extensive resection.<br />

The first phase follows small-bowel resection and<br />

is associated with massive losses of water and<br />

electrolytes. Severe diarrhea is increased by<br />

gastric hypersecretion. During this period, total<br />

PN is required in association with small amounts<br />

of substrates provided by the enteral route, orally<br />

or by continuous gastric infusion, as soon as<br />

intestinal transit has recovered. During the<br />

second phase, when intestinal transit is permanently<br />

re-established, intestinal function improves<br />

as a result of progressive adaptation of the<br />

remaining small bowel. PN allows the short<br />

bowel to develop and to become functional<br />

without exceeding its capacities. This period can<br />

take several months or years before the stage of<br />

maximal adaptation is reached. The third phase<br />

starts when intestinal function is sufficient to<br />

absorb nutrients, enabling PN to be withdrawn.<br />

All calories are provided by the oral route. Oral<br />

intake can then be further liberalized, both in<br />

volume and in variety, according to the tolerance<br />

of the patient. This long period requires special<br />

management and follow-up.<br />

Phase 1<br />

The major goals of medical therapy are to compensate<br />

for the intestinal losses while attempting to<br />

reduce them and to achieve nutritional repletion.<br />

Replacement solutions not uncommonly require a<br />

Table 29.2 Etiology of short-bowel syndrome<br />

Prenatal <strong>Neonatal</strong> Postnatal<br />

Clinical management of short-bowel syndrome 465<br />

very high sodium concentration replacement,<br />

often as high as 80–100mmol/l, in order to maintain<br />

fluid and electrolyte homeostasis. The<br />

requirements of massive intravenous fluids and<br />

the necessity to avoid multiple peripheral<br />

perfusions necessitate the early insertion of a<br />

central venous catheter. Such vascular access<br />

allows the safe replacement of volumes of fluid<br />

according to the evaluation of intestinal losses.<br />

Water, sodium and potassium requirements are<br />

also increased. When losses stabilize and the clinician<br />

becomes familiar with the management of<br />

the infant, fluid and electrolyte losses can be<br />

added to the PN solution and administered<br />

through a single infusion device. Patients with<br />

proximal enterostomy require special trace<br />

element supplementation. Zinc deficiency can<br />

lead to a decreased activity of zinc-dependent<br />

intestinal metalloenzymes such as alkaline phosphatase,<br />

leucine aminopeptidase and other intestinal<br />

disaccharidases. 59 On the other hand, PN<br />

provides adequate nutritional supply to the child,<br />

allowing optimal growth. Such nutritional support<br />

includes the use of 1.5–2.5g/kg per day of amino<br />

acids through a pediatric solution, a caloric intake<br />

consisting of 70–80% of non-protein energy<br />

substrates such as dextrose and 20–30% energy<br />

provided by a 20% intravenous fat emulsion.<br />

Maintenance amounts of vitamins and trace<br />

elements are added to the PN solution by using<br />

commercially available preparations. Calcium,<br />

phosphate and magnesium are also added to the<br />

solution according to the patient’s needs and the<br />

stability of the solution.<br />

During the early phases of therapy, serum electrolytes,<br />

glucose, urea and calcium should be<br />

Atresia (unique or multiple) Midgut volvulus Midgut volvulus (malrotation,<br />

Apple peel syndrome (midgut or segmental) bands or tumor)<br />

Midgut volvulus (malrotation) Necrotizing enterocolitis Complicated intussusception<br />

Segmental volvulus (with Arterial thrombosis Arterial thrombosis<br />

omphalomesenteric duct or Venous thrombosis Inflammatory bowel disease<br />

intra-abdominal bands) Post-trauma resection<br />

Abdominal wall defects Extensive angioma<br />

Gastroschisis > omphalocele<br />

Extensive Hirschsprung’s disease


466<br />

Short-bowel syndrome<br />

measured daily. When the patient’s condition and<br />

the intestinal losses become stable, blood monitoring<br />

can be less frequent. During the first stage after<br />

resection, H2-receptor blocking agents (ranitidine)<br />

should be given intravenously to inhibit gastric<br />

hypersecretion: 15–20mg/kg per day of ranitidine<br />

added to the PN solution, the drug being delivered<br />

as a continuous infusion. H2-receptor-blocking<br />

agents, by increasing duodenal pH, also improve<br />

the digestion and absorption of nutrients.<br />

The end of the phase of management is considered<br />

to be accomplished when the patient has recovered<br />

from the surgical procedure and is stable<br />

on PN with controlled intestinal losses and<br />

motility.<br />

Phase 2<br />

This phase of management is marked by the<br />

initiation of enteral and/or oral feeding and the<br />

gradual cycling of PN. As supported by the<br />

mechanisms of adaptation, there is a clear advantage<br />

in providing early gradual amounts of<br />

nutrients into the residual intestine. Indeed, intraluminal<br />

nutrition is mandatory for stimulating<br />

mucosal hyperplasia. The absence of luminal<br />

nutrients slows the intestinal hyperplasia process<br />

even when the appropriate amount of calories is<br />

given by PN. 60<br />

Phase 3<br />

The transition between total PN and full<br />

enteral/oral feeding is achieved during phase 2<br />

either in hospital or, better, on home PN when<br />

necessary. Phase 3 begins with the discontinuation<br />

of artificial nutrition. When the patient receives no<br />

more than two or three perfusions per week, PN<br />

discontinuation is attempted. If both protein–<br />

energy intake and intestinal function are adequate,<br />

normal weight gain and growth velocity must be<br />

achieved. If the weight gain remains correct after 3<br />

months without PN, the central venous catheter is<br />

removed. If not, PN should be restarted for a few<br />

weeks or months. In about 80–90% of cases, the<br />

first attempt to stop PN completely is successful.<br />

When all the necessary calories are provided by<br />

oral feeding, oral intake can be further liberalized<br />

in both volume and variety. In a very small<br />

number of neonatal cases of short-bowel syndrome<br />

(less than 5% in our experience) eating disorders<br />

may be a serious problem. After eliminating all<br />

organic causes, reinforcement of psychological<br />

support is mandatory.<br />

Mode of feeding<br />

The mode of administration of feeding varies<br />

among different groups. Oral feeding seems to<br />

have several advantages, especially in neonates.<br />

Oral feeding is more physiological, by stimulating<br />

salivary secretion containing EGF, gallbladder<br />

motility and gastrointestinal secretions. In addition,<br />

oral feeding reduces PN-related liver disease.<br />

Early initiation of oral feeding, by allowing the<br />

infant to learn how to suck and swallow, is in addition<br />

the best way to prevent anorexia secondary to<br />

the absence of suckling and long-term hospitalization.<br />

The use of breast milk has the advantage of<br />

containing additional non-nutritive factors such as<br />

prostaglandins or EGF and it seems logical, despite<br />

the fact that it contains lactose. Oral feeding<br />

increases stool volume output and frequency even<br />

when a semi-elemental diet is used. Intestinal<br />

tolerance can be determined by the volume of the<br />

stools, their consistency, the number of bowel<br />

movements, the presence or absence of reducing<br />

substances and pH (


tain no fiber, have an osmolality below<br />

310mOsm/kg, and contain substrates that are<br />

rapidly transferred across the intestine without<br />

leaving intraluminal residues. A dilute concentration<br />

is gradually increased from 0.6 to 1kcal/ml<br />

according to digestive tolerance (stool losses,<br />

reducing substances). It is also important to avoid<br />

excessive fluid administration concurrently with<br />

PN which may overload the patient with fluid.<br />

CEF enhances absorption by permitting total saturation<br />

of the transporters in the gut 24h a day.<br />

Bolus feeding through the nasogastric or gastrostomy<br />

tube is not tolerated and should be avoided.<br />

Adapted CEF subsequently decreases the PN<br />

requirements and reduces liver injury. The recent<br />

development of new portable infusion devices and<br />

backpacks permits older children, who are able to<br />

walk independently, to have reasonable mobility<br />

as they grow and develop physically. If the child is<br />

managed by using CEF, oral feeding should be<br />

maintained. Introduction of solid feedings by 6<br />

months of age is also important to help the child<br />

learn to eat and swallow at an appropriate age,<br />

again to avoid feeding-aversion behavior.<br />

Type of diet<br />

The choice of an appropriate diet for CEF remains<br />

controversial. During the past decades, special<br />

diets were first used; each of the constituents<br />

could be modified independently. An important<br />

improvement has been the introduction of semielemental<br />

diets containing protein hydrolysates,<br />

oligosaccharides and a mixture of MCTs and longchain<br />

triglycerides (LCTs).<br />

Protein in the diet<br />

It was demonstrated that the absorption of amino<br />

acids is more rapid and more efficient when given<br />

in the form of short peptides than in the form of<br />

free amino acids. 61–63 In addition to the quality of<br />

the protein, digestion and intestinal absorption of<br />

protein hydrolysates depends on the type of<br />

hydrolysate used: α-lactalbumin is better than<br />

casein. 64,65 Amino acid-based formulas have been<br />

used in pediatric patients with short-bowel<br />

syndrome and are well accepted by the infants. 65,66<br />

Their beneficial effects have to be demonstrated by<br />

further studies.<br />

Clinical management of short-bowel syndrome 467<br />

Energetic substrates: carbohydrates and lipids<br />

Because of the increased turnover of enterocytes in<br />

the small bowel, lactase activity appears to be<br />

depressed in patients with short-bowel syndrome.<br />

Oligosaccharides provide low osmolality and<br />

allow the amount of carbohydrates to be increased.<br />

The behavior of MCTs, their split products in the<br />

intestinal lumen and their absorption characteristics<br />

are mainly due to their greater water solubility.<br />

67 MCTs are hydrolyzed more rapidly than<br />

LCTs in the small intestine by pancreatic lipase;<br />

they are converted almost exclusively into free<br />

fatty acids and glycerol and reach the liver directly<br />

via the portal circulation. Thus, even in the case of<br />

pancreatic insufficiency, MCTs may be absorbed<br />

intact. Moreover, MCTs have been shown to be<br />

absorbed, to some extent, within the stomach and<br />

duodenum. The unabsorbed fraction is then<br />

absorbed within the proximal part of the jejunum.<br />

The excessive content in the diet of MCTs can lead<br />

to osmotic diarrhea as a result of rapid hydrolysis<br />

of the MCTs’ bulk. 68 Studies in animals with<br />

enterectomy have shown that MCTs are not as<br />

effective as LCTs in stimulating mucosal hyperplasia.<br />

69 LCTs appear to be potentially more<br />

trophic than other nutrients for the mucosal mass.<br />

LCTs stimulate biliary and pancreatic secretions,<br />

which are themselves trophic factors. In addition,<br />

LCTs are a substantial source of linolenic and<br />

linoleic acids, which are essential substrates for<br />

endogenous prostaglandin synthesis. 70,71 Most of<br />

the formulas containing MCTs also include up to<br />

50% of lipid in the form of LCTs. LCTs stimulate<br />

biliary and pancreatic secretions, which promote<br />

increased intestinal motility; when LCTs are in<br />

excess in the intestinal lumen, they may be<br />

hydroxylated by bacteria and reverse the rate of<br />

water and electrolyte absorption, resulting in<br />

aggravation of malabsorption. Under these conditions,<br />

oral therapy with cholestyramine (0.5–2g/kg<br />

per day) may be helpful, and supplementation<br />

with essential fatty acids is crucial. Thus, an<br />

intake of 2–4g/kg per day of lipids is recommended,<br />

depending on the absorption capacity<br />

and digestive tolerance.<br />

Dietary fibers<br />

It is possible for an intact colon to absorb daily<br />

energy from dietary fiber. 45,46 The role of SCFAs<br />

has been demonstrated in animals. 47,48 In patients


468<br />

Short-bowel syndrome<br />

with a preserved colon, a reasonable amount of<br />

dietary fiber may be progressively included in oral<br />

feeding. Because SCFAs are supposed to stimulate<br />

sodium and water absorption, infants and children<br />

might be expected to have a decreased stool output<br />

and sodium losses. However, this is not always<br />

observed in clinical practice. Therefore, soluble<br />

non-starch polysaccharides and some starches<br />

should be given, according to digestive tolerance<br />

and especially stool output as well as abdominal<br />

distension due to intracolonic fermentation of<br />

fiber by an abundant and modified intestinal<br />

bacterial microflora. In patients without a colon or<br />

with reduced colon length without an ICV, dietary<br />

fiber should be reduced or even avoided.<br />

Bacterial overgrowth<br />

Bacterial overgrowth is a frequent complication,<br />

causing mucosal injury, malabsorption and bacterial<br />

translocation. In addition, bacterial overgrowth<br />

exacerbates hepatotoxicity related to PN. 72–80<br />

Indeed, deconjugation of bile acids by bacteria<br />

facilitates their reabsorption, leading to hepatotoxicity.<br />

Bacterial overgrowth is likely to occur in the<br />

case of ICV resection, poor motility of a dilated<br />

small-bowel segment, or when a tight anastomosis<br />

is present. After bacterial overgrowth has been<br />

confirmed by intestinal flora analysis (fecal and<br />

duodenal) and the hydrogen breath test, it is<br />

mandatory to stop it. Antimicrobial therapy as the<br />

combination of metronidazole and colimycin three<br />

times a day may be helpful. The efficiency of the<br />

antimicrobial therapy can be assessed by the clinical<br />

improvement or by the intestinal tolerance to<br />

food and mainly by performing stool balance<br />

studies. The use of broad-spectrum antimicrobial<br />

therapy is, in our opinion, contraindicated,<br />

because of the high risk of emergence of multiresistant<br />

bacteria and the anti-physiological effect<br />

on colonic flora. In the case of bacterial<br />

overgrowth, the administration of Saccharomyces<br />

boulardii may be effective. Whenever possible,<br />

performing an intestinal tapering procedure or<br />

resecting a tight anastomosis may be required to<br />

obtain the disappearance of bacterial overgrowth<br />

(see below). The use of prokinetic drugs for<br />

enhancing gut motor activity is strictly contraindicated<br />

in these conditions.<br />

Parenteral nutrition cycling<br />

Besides enteral feeding, phase 2 is characterized<br />

by the cycling of PN. As soon as the metabolic and<br />

nutritional status or the patient permits it, cyclic<br />

PN should be started. Cyclic infusion of nutrients<br />

allows the periods of feeding and fasting to be<br />

alternated, which induces a more physiological<br />

regulation of metabolism by insulin and glucagon;<br />

it prevents hyperinsulinism which is responsible<br />

for fat accumulation within the liver and adipose<br />

tissue. 81,82 Furthermore, cyclic PN favors a more<br />

normal physical activity whose importance in<br />

terms of nutrition and psychology has been<br />

demonstrated. The tolerance of this mode of<br />

nutrient delivery has been well documented in<br />

children. In addition, cyclic PN is the most appropriate<br />

condition for home PN whose development,<br />

since 1980, has dramatically improved the wellbeing<br />

and quality of life of patients requiring longterm<br />

PN. 83,84 Cyclic PN can be progressively<br />

achieved by slowly decreasing the infusion rate,<br />

followed by complete discontinuation of nutrient<br />

infusion for several hours per day. Long-term PN<br />

has to be adapted in order to avoid complications<br />

related to excessive or insufficient intakes. 85–88<br />

Adaptation to fluid losses<br />

Extensive small-bowel resection is associated with<br />

gastric hypersecretion. The H2 antagonists and<br />

proton pump inhibitors are useful in reducing<br />

gastric fluid secretion, and therefore will also<br />

reduce fluid losses. However, absorption of orally<br />

dosed medications may be impaired, and either<br />

large doses, oral medication, or intravenous delivery<br />

may be required. Although fluid losses are<br />

decreased, macronutrient and electrolyte absorption<br />

are not affected by H2 antagonists and proton<br />

pump inhibitors.<br />

Reduction of fluid loss<br />

Fluid losses may require chronic control with<br />

hypomotility agents. Synthetic opiate derivatives<br />

such as loperamide may be beneficial in delaying<br />

gastric emptying and decreasing gastrointestinal<br />

motor activity. They allow the transit time to


decrease and enhance intestinal transport.<br />

Nevertheless, loperamide must be used very carefully<br />

and should be avoided whenever a motility<br />

disorder leads to intestinal distension with subsequent<br />

bacterial overgrowth. In patients with ileal<br />

resection and watery stools, cholestyramine may<br />

bind bile acids and reduce biliary colitis and diarrhea.<br />

However, cholestyramine causes fat malabsorption<br />

and increases the risk of fat-soluble<br />

vitamin deficiency. Somatostatin analogs may be<br />

helpful for short-term use. 89 The mechanism of<br />

action is unclear, but octreotide may be useful in<br />

slowing intestinal transit time and increasing<br />

water and sodium absorption. However, octreotide<br />

reduces splanchnic protein synthesis, thereby<br />

reducing mucosal protein incorporation and<br />

villous growth rate, and may impair postresectional<br />

intestinal adaptation. There is also an<br />

increased risk for cholelithiasis in the patient<br />

group already predisposed to this problem. Use in<br />

children has not been well studied and there are<br />

some concerns regarding the suppression of<br />

growth hormone in this population already at risk<br />

of poor growth.<br />

Prevention of dehydration<br />

In a child on cyclic PN, because of reduced water<br />

intake and persistence of intestinal losses, one<br />

should be careful when restarting PN. It is<br />

important to increase the infusion rate progressively<br />

during the first hour of PN, because the<br />

child may be mildly dehydrated. Especially infants<br />

with an extremely short-bowel or with a smallbowel<br />

stoma have obligatory losses of water and<br />

electrolytes even when fasting. The volume of<br />

effluent rises in proportion to the amount of distal<br />

intestine removed. Since the sodium concentration<br />

of the effluent is almost constant at about<br />

90mEq/l or greater, the sodium losses correlate<br />

directly with the volume of effluent and are<br />

increased by oral feeding. Despite large amounts of<br />

sodium given parenterally, fasting periods of cyclic<br />

PN should be carefully adapted. Patients with less<br />

than 60cm of jejunum remaining between the<br />

duodenojejunal flexure and the stoma lose more<br />

fluid and sodium from the stoma than they can<br />

take in orally and may be at risk for developing<br />

negative water and sodium balance. Those<br />

patients with severe diarrhea are also subject to<br />

magnesium deficiency. Oral rehydration solutions<br />

Clinical management of short-bowel syndrome 469<br />

with adequate sodium concentration can be given<br />

to maximize water and salt absorption. Such<br />

patients should be discouraged from drinking large<br />

quantities of water or diluted sodium solutions.<br />

Complications of short-bowel syndrome and<br />

parenteral nutrition<br />

Long-term PN exposes the patient to several<br />

complications, including catheter-related sepsis<br />

and thrombosis, bone disease and liver impairment.<br />

72–80 Daily catheter care and prevention of<br />

septic complications are essential (see Chapter 33).<br />

Liver function tests should be performed on a<br />

regular basis because of the risk of cholestasis and<br />

liver injury. If initial surgery did not include a gallbladder<br />

resection, ultrasonography aiming to look<br />

for sludge or cholelithiasis should be repeated<br />

every 3 months. In addition, the phosphorus and<br />

calcium status should be assessed and, if necessary,<br />

supplies should be given to prevent PNrelated<br />

bone disease.<br />

Liver disease is certainly the most frequent and the<br />

most severe complication in patients with shortbowel<br />

syndrome. Premature babies and/or smallfor-gestational-age<br />

babies with severe necrotizing<br />

enterocolitis are at specially high risk of developing<br />

liver disease and, rapidly, end-stage liver<br />

failure, because of the combination of prematurity,<br />

sub-occlusion, Gram-negative sepsis and repeated<br />

catheter-related sepsis. In general, liver disease is<br />

mostly related to the impaired small intestine and<br />

is further aggravated by inadequate PN.<br />

The main factors related to liver disease<br />

These are thought to be:<br />

(1) Disruption of the enterohepatic cycle (ileal<br />

disease or resection);<br />

(2) Intestinal stasis with consecutive intraluminal<br />

bacterial overgrowth and/or translocation<br />

(endotoxinemia); 74–76<br />

(3) Recurrent catheter-related sepsis; 90<br />

(4) Prematurity itself, which might be an associated<br />

factor; 78<br />

(5) Inadequate PN intakes such as continuous PN<br />

infusion with excessive glucose intake (hyper-


470<br />

Short-bowel syndrome<br />

insulinism) and subsequent steatosis or inadequate<br />

amino acid supply. 81<br />

Management for prevention or resolution of liver<br />

injury<br />

This should include:<br />

(1) Stimulation of the enterobiliary axis by ingestion<br />

of LCTs or breast milk, or by injection<br />

of CCK analogs; 38<br />

(2) Suppression of intraluminal bacterial overgrowth<br />

caused by intestinal stasis, by giving<br />

metronidazole and/or performing tapering<br />

enteroplasty; 91<br />

(3) The use of ursodesoxycholic acid (10–20mg/kg<br />

per day) which might contribute to the decrease<br />

of liver injury; 92<br />

(4) PN intake, which should be adapted in terms of:<br />

(a) limiting glucose intakes to reduce hepatic<br />

fat accumulation; 93<br />

(b) decreasing aluminium content of parenteral<br />

nutrition solution; 94<br />

(c) using appropriate type and amount of intravenous<br />

fat emulsion which provides essential<br />

fatty acids and reduces glucose load; 95<br />

(d) using the new pediatric adapted amino<br />

acid solutions which provide appropriate<br />

amino acids as well as taurine; 96<br />

(e) performing cyclic parenteral nutrition that<br />

contributes towards reducing hyperinsulinism<br />

and reducing liver steatosis. 81<br />

Transition to parenteral nutrition weaning<br />

When oral feeding and home PN are commenced<br />

in combination, the amounts and/or rate of PN<br />

infusion are progressively decreased. In our experience,<br />

the combination of oral feeding and cyclic<br />

PN allows home PN to be achieved in the best<br />

conditions. The combination of CEF and cyclic PN<br />

seems to us less logical, since nutritional support<br />

would take place only during the nocturnal period.<br />

If not, the patient would be dependent on artificial<br />

nutritional support for 24h a day. In the past 20<br />

years we have treated, in hospital and/or by home<br />

PN, over 200 patients, mainly infants, with shortbowel<br />

syndrome. The duration of stage 2 depends<br />

on several factors, which include: the length of the<br />

remaining total intestine, conservation of the ICV<br />

and the functional capacity of the remaining small<br />

intestine. The slow transition from total PN to full<br />

oral feeding requires time, during which the nutritional<br />

status has to be maintained at the optimal<br />

level. In this regard, home PN is the best tool both<br />

to maintain nutritional status and to achieve bowel<br />

adaptation. This is a physiological anabolic process<br />

which requires optimal nutritional status and liver<br />

function. The delay in achieving intestinal autonomy<br />

(PN weaning) depends on residual smallbowel<br />

length and ICV preservation (see below). 1–3<br />

By using stool balance analysis we have shown that<br />

most patients who have been weaned from PN<br />

remain with a certain degree of malabsorption that<br />

is compensated by relative hyperphagia. 97<br />

Conversely, trying to wean an infant with shortbowel<br />

syndrome from PN too rapidly leads to the<br />

risk of metabolic complications and failure to<br />

thrive. 97,98<br />

Long-term complications<br />

The loss of ileum can lead to hyperoxaluria and<br />

subsequent renal stone formation. Oxaluria evaluation,<br />

as well as ultrasonography of the kidney, must<br />

be repeated every 6 months. Prevention of oxalic<br />

lithiasis includes a restricted diet containing LCTS,<br />

the addition of calcium to the diet and elimination<br />

from the diet of foods containing large amounts of<br />

oxalic acid. 99,100<br />

In the case of resection of the terminal ileum,<br />

vitamin B12 supplementation is required. An intramuscular<br />

dose of 1mg every 6 months is recommended.<br />

Because of poor absorption of fat-soluble<br />

vitamins (A, D, E and K) follow-up and supplementation<br />

are necessary. Nutritional evaluation<br />

must also include research of zinc and biotin deficiency.<br />

After an extensive small-bowel resection<br />

and even after adaptation, one must take into<br />

account the possibility that the medications administered<br />

orally will not be absorbed at optimal levels.<br />

This becomes particularly important in the case of<br />

pediatric infections requiring antibiotics. On the<br />

other hand, unjustified use of antibiotics may lead<br />

to severe and protracted intestinal microflora<br />

impairment. Thus, antibiotic therapy should whenever<br />

possible be avoided, and if necessary be<br />

performed by a pediatric gastroenterologist.


D-Lactic acidosis<br />

D-Lactic acidosis is a classical disorder following<br />

extensive small-bowel resection with preservation<br />

of the colon. Lactobacilli and other bacteria,<br />

including Clostridium perfringens and Streptococcus<br />

bovis, when present, ferment non-absorbed<br />

carbohydrate to D-lactic acid, which cannot be<br />

metabolized by D-lactate dehydrogenase. These<br />

organisms may proliferate in an acidic environment<br />

that may be promoted by the metabolism of<br />

unabsorbed carbohydrate to SCFAs. A metabolic<br />

acidosis may develop in the absence of hepatic<br />

dysfunction if significant absorption of D-lactic<br />

acid occurs. 101 Clinical symptoms include, in the<br />

rare and severe form, encephalopathy, headache,<br />

ataxia and dysarthria. D-Lactate concentrations<br />

will be increased in blood and urine (L-lactate<br />

concentration, is usually measured when serum<br />

lactate concentration is normal). The mechanism<br />

for the neurological symptoms is unknown.<br />

Thiamine deficiency should be excluded. 102,103<br />

Spontaneous resolution is possible while treatments<br />

include oral metronidazole, neomycin,<br />

vancomycin (for 10–14 days) and avoidance of<br />

‘refined’ carbohydrates. 104,105 Saccharomyces<br />

boulardii may be used in these circumstances. 106<br />

Finally, chronic metabolic acidosis may cause<br />

impaired growth in some children with shortbowel<br />

syndrome.<br />

Long-term follow-up and growth monitoring<br />

During the long-term follow-up, weight gain and<br />

growth velocity must be regularly monitored.<br />

Dietary intake should be adapted to digestive tolerance,<br />

the child’s preference and growth. Whenever<br />

failure to thrive occurs, it is important to perform<br />

a dietetic evaluation of the nutrient intake, stool<br />

balance studies and investigations for bacterial<br />

overgrowth. 80 Bone mineral mass has to be<br />

assessed by dual X-ray absorptiometry. 107 At this<br />

point one should discuss the opportunity for<br />

restarting artificial nutrition by enteral feeding or<br />

in some cases PN. We performed a retrospective<br />

study to analyze the outcome, the prognosis<br />

factors and the long-term growth of children after<br />

extensive small-bowel resection in the neonatal<br />

period. 108 The study included 87 children who<br />

had undergone extensive neonatal small-bowel<br />

resection, followed up over a mean 15-year period.<br />

The overall survival was 89.7%, depending on the<br />

Clinical management of short-bowel syndrome 471<br />

date of birth. The duration of PN dependency<br />

varies according to the intestinal length and the<br />

presence of the ICV (Figure 29.1). All patients who<br />

remained PN dependent had less than 40cm of<br />

small bowel and/or the absence of the ICV. A group<br />

of patients with a mean smal-bowel length of<br />

35±19cm, resection of the ICV in 50% of cases<br />

and a PN duration of 47.4±23.8 months had<br />

significant decrease in height and weight gain<br />

within the 4 years after cessation of PN, requiring<br />

a new period of enteral or parenteral feeding.<br />

Finally, the largest group included patients with a<br />

mean small-bowel length of 57±19cm, the presence<br />

of the ICV in 81% of cases and a PN duration<br />

of 16.1±11.4 months. After PN weaning, they<br />

grew normally with normal puberty and final<br />

height as expected from genetic target height. PN<br />

duration is clearly influenced by the length of<br />

residual small bowel and the absence of the ICV.<br />

With good anatomic prognosis factors and short<br />

duration of initial PN, normal long-term growth<br />

may be predicted. Conversely, poor anatomical<br />

factors and protracted initial PN require careful<br />

monitoring of growth and may sometimes require<br />

nutritional support to be restarted. Patients permanently<br />

dependent on PN might be candidates for<br />

intestinal transplantation.<br />

% Children PN-dependent<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

0 12 24 36 48 96<br />

PN duration (months)<br />

< 40 cm, ICV –<br />

≥ 40 cm, ICV –<br />

< 40 cm, ICV +<br />

≥ 40 cm, ICV +<br />

Figure 29.1 Parenteral nutrition (PN) duration according<br />

to anatomical factors. Note that 100% of children with<br />

small-bowel length 40–80cm and the presence of<br />

ileocecal valve (ICV) are weaned from PN within 1 year.<br />

Conversely, a large percentage of children with


472<br />

Short-bowel syndrome<br />

Long-term outcome and management<br />

of ‘unadapted’ short small bowel<br />

Since the review published in 1972 by Wilmore,<br />

the prognosis of neonatal short-bowel syndrome<br />

has dramatically changed. 108–113 More than 90% of<br />

infants and children now survive after extensive<br />

small-bowel resection. 1–3 After neonatal extensive<br />

resection, the residual small intestine can adapt,<br />

even when its remaining length below the ligament<br />

or Treitz is shorter than 15cm, the ICV being<br />

left intact. In older children having undergone<br />

extensive resection, the criterion for length is<br />

different. Adequate adaptation to ensure normal<br />

growth without artificial nutrition requires at least<br />

40cm below the Treitz angle measured at the time<br />

of resection. 114,115 In neonatal short-bowel<br />

syndrome, by separating our series of patients into<br />

two groups according to PN duration (48 months), assessments of factors such as<br />

functional capacity of the remaining short bowel<br />

or bacterial overgrowth emerge as being essential<br />

(Table 29.3). 116 Thus, a small number of patients,<br />

because of motility disorders or an especially short<br />

small bowel, will adapt only very slowly, if at all.<br />

In such patients, different medical and/or surgical<br />

approaches have been proposed.<br />

Pharmacological enhancement of intestinal<br />

adaptation<br />

Growth hormone and insulin-like growth factor-I<br />

A number of studies have indicated that pituitary<br />

hormones modulate small-bowel growth in<br />

rodents. 117–119 In hypophysectomized rat pups, the<br />

rate of growth of the small intestine was<br />

decreased. 117 Administration of growth hormone<br />

(GH) to hypophysectomized rats restored the<br />

growth deficit of the small bowel. GH also stimulated<br />

small-bowel growth in intact animals. 120–122<br />

Although GH receptors have been detected in rat<br />

intestinal mucosal cells, 123 the effects of GH on the<br />

gut may be mediated through IGF-I, since both type<br />

I and type II receptors for IGF have been demonstrated<br />

in rat 124 and rabbit gastrointestinal epithelium.<br />

125 IGF-I is synthesized in the rat small intestine<br />

in a GH-dependent manner. 126 IGF-I treatment<br />

increased small-bowel mass in rats after 80% smallbowel<br />

resection or in rats with mucosal atrophy<br />

due to glucocorticoid treatment. 127<br />

In humans, it was shown that GH enhanced amino<br />

acid uptake by the small intestine ex vivo 128 and<br />

promoted crypt cell proliferation in cultured<br />

explants of duodenal mucosa. 129 Both open and<br />

randomized clinical trials have now been<br />

conducted in humans. 130–134 Their results are<br />

controversial and do not allow conclusions to be<br />

drawn on a long-term benefit in patients with<br />

short-bowel syndrome. GH administration<br />

(0.5IU/kg per day or 0.024mg/kg per day) alone for<br />

8 weeks had no effect on the absorptive capacity of<br />

energy, protein, or fluid in ten patients (four with<br />

colon in continuity). 134<br />

In infants with short-bowel syndrome, recombinant<br />

human GH (rhGH) has been used for a 10-day<br />

period at a dose of 0.3IU/kg per day. Reported data<br />

include only significant weight gain during rhGH<br />

treatment and a tendency to increase polyamine<br />

Table 29.3 Factors predicting parental nutrition duration to be >48 months in infants with neonatal<br />

short-bowel syndrome, by univariate analysis<br />

>48months


concentration in the small-bowel mucosa. 135<br />

Recently, we performed a small, pilot, open-labeled<br />

trial in eight children with short-bowel syndrome<br />

with more than 3 years of PN dependency, still<br />

receiving >50% of their protein–<br />

energy requirements from PN. After 3 months of<br />

rhGH treatment (0.6IU/kg per day) all the patients<br />

were weaned from PN. One-year follow-up indicated<br />

that 25% had been definitively PN weaned,<br />

50% had decreased PN requirements and 25% did<br />

not change their PN dependency. 136 More<br />

prolonged and perhaps earlier use of rhGH in<br />

infants or children with short-bowel syndrome<br />

might be helpful for future management.<br />

Glucagon-like-peptide-2<br />

Glucagon-like peptide-2 (GLP-2) has been proposed<br />

for enhancing intestinal adaptation in patients with<br />

short-bowel syndrome. 137,138 GLP-2 is secreted from<br />

L cells of the ileum as well as from pancreatic A<br />

cells. It was shown that postprandial serum GLP-2<br />

concentration was not unexpectedly depressed in<br />

patients who have had extensive small-bowel<br />

resection, especially including the ileum. 139 The<br />

relative lack of jejunal hypertrophy following ileal<br />

resection might be partly related to the resection of<br />

GLP-2-producing L cells. GLP-2 was used in a clinical<br />

setting (400µg subcutaneously, twice a day for<br />

35 days) was administered in a small, pilot, openlabeled<br />

trial to four patients with short-bowel<br />

syndrome without residual colon who required<br />

total PN and in four patients with sufficient residual<br />

jejunum where total PN was not required. 140<br />

Jejunal villous height and crypt depth tended to<br />

increase, energy absorption tended to increase and<br />

fecal wet weight decreased, indicating increased<br />

fluid absorption. Regarding its origin (ileum) and<br />

its physiological effect, GLP-2 might be the most<br />

logical medical approach for early management of<br />

the patient with short-bowel syndrome with ileal<br />

resection. GLP-2 is not commercially available, but<br />

might be helpful in the near future if a genetically<br />

engineered analog of GLP-2 is developed.<br />

Surgical treatment of the short-bowel<br />

syndrome<br />

Under some circumstances, surgical procedures<br />

can slow down intestinal transit or increase the<br />

Conclusion 473<br />

area of absorption. Several surgical procedures<br />

have been proposed for decreasing intestinal transit<br />

time. The construction of a valve or a sphincter<br />

with sutures and synthetic material, the denervation<br />

segments of the intestine, and the intussusception<br />

of intestinal loops have all been experimentally<br />

assessed. 141–145 The clinical experience is,<br />

however, more limited. Reversing segments of<br />

intestine aims at creating an antiperistaltic segment<br />

acting as a ‘physiological valve’ by causing retrograde<br />

peristalsis. Experimental results are conflicting,<br />

because the ideal length of the antiperistaltic<br />

segment has not been clearly established. 146,147<br />

Pediatric patients having undergone such a procedure<br />

have been reported. 148 Most of the time,<br />

transit time is slowed down and absorption<br />

increased. A serious limitation of this procedure is<br />

that patients with very short remaining small<br />

bowel may not be able to afford the sacrifice of a<br />

10-cm segment for reversal. Therefore, this procedure<br />

has never been used by our team.<br />

Interposition of a colonic segment has also been<br />

proposed both in isoperistaltic and in antiperistaltic<br />

fashions. Clinical results are poor, and show<br />

little benefit. 149–151 Intestinal tapering and lengthening<br />

have been performed in selected patients<br />

with dilated bowel segments. 152–157 Marked<br />

dilatation of the proximal intestine may occur<br />

secondary to chronic obstruction or adaptation.<br />

This situation leads to stasis and bacterial overgrowth<br />

which further aggravates malabsorption.<br />

Intestinal tapering and lengthening has the theoretical<br />

interest of not only tapering the dilated<br />

segment but also of using the divided intestine to<br />

increase in length. The procedure should be<br />

applied with caution, since there is a risk or ‘jeopardizing’<br />

the divided segments. Long-term patency<br />

and function have been shown to occur in some<br />

patients. 158–160 Finally, in cases or non-adapted<br />

small bowel despite long-term trials, the only alternative<br />

is a small-bowel transplantation.<br />

Conclusion<br />

Infants with short-bowel syndrome from birth or<br />

early life should adapt, thanks to the physiological<br />

process of adaptation. Everything has to be done to<br />

promote intestinal adaptation and digestive<br />

autonomy, especially by using the digestive tract<br />

and the oral route as soon and as much as possible.


474<br />

Short-bowel syndrome<br />

Unfortunately, too many infants have complications<br />

related to unadapted PN and become<br />

cholestatic with a course of end-stage liver disease<br />

requiring liver transplant. Some pediatric patients<br />

were reported to have received an isolated liver<br />

transplantation for end-stage liver disease compli-<br />

REFERENCES<br />

1. Goulet OJ, Revillon Y, Jan D et al. <strong>Neonatal</strong> short bowel<br />

syndrome. J Pediatr 1991; 119: 18–23.<br />

2. Georgeson KE, Breaux CW Jr. Outcome and intestinal<br />

adaptation in neonatal short–bowel syndrome. J Pediatr<br />

Surg 1992; 27: 344–350.<br />

3. Sondheimer JM, Cadnapaphornchai M, Sontag M, Zerbe<br />

GO. Predicting the duration of dependence on<br />

parenteral nutrition after neonatal intestinal resection. J<br />

Pediatr 1998; 132: 80–84.<br />

4. Warner BW, Chaet MS. Non transplant surgical options<br />

for the management of the short bowel syndrome. J<br />

Pediatr Gastroenterol Nutr 1993; 17: 1–12.<br />

5. Bianchi A. Autologous gastro-intestinal reconstruction.<br />

Semin Pediatr Surg 1995; 4: 54–59.<br />

6. Bryant J. Observations upon the growth and length of<br />

the human intestine. Am J Med Sci 1924; 167: 499–520.<br />

7. Siebert JR. Small intestine length in infants and<br />

children. Am J Dis Child 1980; 134: 593–595.<br />

8. Touloukian RJ, Smith GJW. Normal intestinal length in<br />

preterm infants. J Pediatr Surg 1983; 18; 720–723.<br />

9. Mitchell JE, Breuer RI, Zuckerman L. The colon<br />

influences ileal resection diarrhea. Dig Dis Sci 1980; 25:<br />

33–41.<br />

10. Cosnes J, Gendre JP, Le Quintrec Y. Role of the<br />

ileocaecal valve and site of intestinal resection in<br />

malabsorption after extensive small-bowel resection.<br />

Digestion 1978; 18: 329–336.<br />

11. Mashako MNK, Cezard JP, Boige N et al. The effect of<br />

artificial feeding on cholestasis, gallbladder sludge and<br />

lithiasis in infants: correlation with plasma<br />

cholecystokinin levels. Clin Nutr 1991; 10: 320–327.<br />

12. Pellerin D, Bertin P, Nihoul-Fekete C, Ricour C.<br />

Cholelithiasis and ileal pathology in children. J Pediatr<br />

Surg 1975; 10: 35–41.<br />

13. Ohkohchi N, Andoh T, Izumi U et al. Disorder of bile<br />

acid metabolism in children with short bowel<br />

syndrome. J Gastroenterol 1997; 32: 472–479.<br />

14. Modigliani R, Labayle D, Aymes C et al. Evidence for<br />

excessive absorption of oxalate by the colon in enteric<br />

hyperoxaluria. Scand J Gastroenterol 1978; 13: 187–192.<br />

15. Dowling RH, Booth CC. Structural and functional<br />

changes following small intestinal resection in the rat.<br />

Clin Sci 1967; 32: 139–149.<br />

16. Gracey M. Intestinal microflora and bacterial overgrowth<br />

in early life. J Pediatr Gastroenterol Nutr 1982; 1:<br />

13–22.<br />

cating short-bowel syndrome. 161,162 By using<br />

appropriate long-term PN and home PN, only a few<br />

patients with an extremely short bowel will not<br />

become autonomous and will require isolated<br />

intestinal transplantation. 163–167<br />

17. King CE, Toskes PP. Small intestine bacterial overgrowth.<br />

Gastroenterology 1979; 76: 1035–1055.<br />

18. Carbonnel F, Cosnes J, Chevret et al. The role of<br />

anatomic factors in nutritional autonomy after extensive<br />

small bowel resection. J Parenter Enteral Nutr 1996; 20:<br />

275–280.<br />

19. Nightingale JMD, Lennard-Jones JE, Gertner DJ et al.<br />

Colonic preservation reduces need for parenteral<br />

therapy, increases incidence of renal stones, but does<br />

not change high prevalence of gallstones in patients<br />

with a short bowel. Gut 1992; 33: 1493–1497.<br />

20. Nordgaard I, Hansen BS, Mortensen PB. Importance of<br />

colonic support for energy absorption as small-bowel<br />

failure proceeds. Am J Clin Nutr 1996; 64: 222–231.<br />

21. Strauss E, Gerson CD, Yalow RS. Hypersecretion of<br />

gastrin associated with short bowel syndrome.<br />

Gastroenterology 1974; 66: 175–180.<br />

22. Nightingale JMD, Kamm MA, van der Sijp JRM et al.<br />

Disturbed gastric emptying in the short bowel<br />

syndrome. Evidence for a colonic brake. Gut 1993; 34:<br />

1171–1176.<br />

23. Nightingale JMD, Kamm MA, van der Sijp JRM.<br />

Gastrointestinal hormones in short bowel syndrome.<br />

Peptide YY may be the colonic brake to gastric emptying.<br />

Gut 1996; 39: 267–272.<br />

24. Bury KD. Carbohydrate digestion and absorption after<br />

massive resection of the small intestine. Surg Gynecol<br />

Obstet 1972; 135: 177–187.<br />

25. Urban E, Pena M. In vivo calcium transport by rat small<br />

intestine after massive small bowel resection. Am J<br />

Physiol 1974; 226. 1304–1308.<br />

26. Kurkchubasche AG, Rowe MI, Smith SD. Adaptation in<br />

short-bowel syndrome: reassessing old limits. J Pediatr<br />

Surg 1993; 28: 1069–1071.<br />

27. Sacks AI, Warwick GJ, Barnard JA. Early proliferative<br />

events following intestinal resection in the rat. J Pediatr<br />

Gastroenterol Nutr 1995; 21: 158–164.<br />

28. Sukhotnik I, Siplovich L, Shiloni E et al. Intestinal<br />

adaptation in short-bowel syndrome in infants and<br />

children: a collective review. Pediatr Surg Int 2002; 18:<br />

258–263.<br />

29. Booth IW, Lander AD. Short bowel syndrome. Baillière’s<br />

Clin Gastroenterol 1998; 12: 739–773.<br />

30. Jeppesen PB, Mortensen AP. Enhancing bowel<br />

adaptation in short bowel syndrome. Curr Gastroenterol<br />

Rep 2002; 4: 338–347.


31. Wilmore DW. Growth factors and nutrients in the short<br />

bowel syndrome. J Parenter Enteral Nutr 1999; 23:<br />

117–120.<br />

32. Ziegler TR, Mantell MP, Chow JC et al. Gut adaptation<br />

and the insulin growth factor system: regulation by glutamine<br />

and IGF-1 administration. Am J Physiol 1996;<br />

277: G866–875.<br />

33. Dowling RH. The influence of luminal nutrition on<br />

intestinal adaptation after small bowel resection and bypass.<br />

In Dowling RH, Rieckcn EO, eds. Intestinal<br />

Adaptation. New York: Schattauer Verlag, 1974: 35–46.<br />

34. Weser E, Heller R, Taxil T. Stimulation of mucosal<br />

growth in the rat ileum by bile and pancreatic<br />

secretions after jejunal resection. Gastroenterology 1977;<br />

73: 524–529.<br />

35. Feldman El, Dowling RH, McNaughton J et al. Effects of<br />

oral versus intravenous nutrition on intestinal adaptation<br />

after small bowel resection in the dog.<br />

Gastroenterology 1976; 70: 712–719.<br />

36. Williamson RCN. Intestinal adaptation mechanisms of<br />

control. N Engl J Med 1978; 298: 1444–1450.<br />

37. Drucker DJ. Biological actions and therapeutic potential<br />

of the glucagon–like peptides. Gastroenterology 2002;<br />

122: 531–44.<br />

38. Ling PR, Sheikh M, Boyce P et al. Cholecystokinin<br />

(CCK) secretion in patients with severe short bowel<br />

syndrome (SSBS). Dig Dis Sci 2001; 46: 859–864.<br />

39. Sigalet DL, Martin GR. Hormonal therapy for short<br />

bowel syndrome. J Pediatr Surg 2000; 35: 360–64.<br />

40. Souba W, Klimberg S, Plumley D et al. The role of glutamine<br />

in maintaining a healthy gut and supporting the<br />

metabolic response to injury and infection. J Surg Res<br />

1990; 48: 383–391.<br />

41. Byrne TA, Morrissey TB, Nattakom TV et al. Growth<br />

hormone, glutamine, and a modified diet enhance nutrient<br />

absorption in patients with severe short bowel<br />

syndrome. J Parenter Enteral Nutr 1995; 19: 296–302.<br />

42. Beaugerie L, Carbonnel F, Hecketsweiler B et al. Effects<br />

of an isotonic oral rehydration solution, enriched with<br />

glutamine, on fluid and sodium absorption in patients<br />

with a short-bowel. Aliment Pharmacol Ther 1997; 11:<br />

741–746.<br />

43. Scolapio JS. Effect of growth hormone, glutamine, and<br />

diet on body composition in short bowel syndrome: a<br />

randomized, controlled study. J Parenter Enteral Nutr<br />

1999; 23: 309–312.<br />

44. Dumas F, De Bandt JP, Colomb V et al. Enteral ornithine<br />

alpha-ketoglutarate enhances intestinal adaptation to<br />

massive resection in rats. Metabolism 1998; 47:<br />

1366–1371.<br />

45. Nordgaard I, Hansen BS, Mortensen PB. Colon as a<br />

digestive organ in patients with short bowel. Lancet<br />

1994; 343: 373–376.<br />

46. Briet F, Flourie B, Achour L et al. Bacterial adaptation in<br />

patients with short bowel and colon in continuity.<br />

Gastroenterology 1995; 109: 1446–1453.<br />

47. Koruda MJ, Rolandelli RH, Settle RG et al. The effect of<br />

a pectin-supplemented elemental diet on intestinal<br />

adaptation to massive small bowel resection. J Parenter<br />

Enteral Nutr 1986; 10: 343–350.<br />

48. Koruda MJ, Rolandelli RH, Settle RG et al. Effect of<br />

parenteral nutrition supplemented with short-chain<br />

fatty acids on adaptation to massive small bowel resection.<br />

Gastroenterology 1988; 95: 715–720.<br />

49. Grey VL, Morin CL. Evidence for a growth-stimulating<br />

fraction in the rat after small bowel resection.<br />

Gastroenterology 1985; 89: 1305–1312.<br />

50. Weawer LT, Walker WA. Epidermal growth factor and<br />

the developing human gut. Gastroenterology 1988; 94:<br />

845–847.<br />

References 475<br />

51. Menard D, Arsenault P, Pothier P. Biologic effects or<br />

epidermal growth factor in human fetal jejunum.<br />

Gastroenterology 1988; 94: 656–663.<br />

52. Sham J, Martin G, Meddings JB, Sigalet DL. Epidermal<br />

growth factor improves nutritional outcome in a rat<br />

model of short bowel syndrome. J Pediatric Surg 2002;<br />

37: 765–769.<br />

53. Schwartz MZ, Storozuk RB. Epidermal growth factor<br />

enhances small intestine function. Gastroenterology<br />

1985; 88: 158–60.<br />

54. Malo C, Ménard D. Influence of epidermal growth factor<br />

on the development or suckling mouse intestinal<br />

mucosa. Gastroenterology 1982; 83: 28–35.<br />

55. Luk GD, Yang P. Distribution or polyamines and their<br />

biosynthetic enzymes in intestinal adaptation. Am J<br />

Physiol 1988; 254: G194–G200.<br />

56. Buts JP, De Keyser N, Kolanowski J et al. Maturation of<br />

villus and crypt cell functions in rat small intestine:<br />

role or dietary polyamines. Dig Dis Sciences 1993; 38:<br />

1091–1098.<br />

57. Helander HF, Johansson C, Blom H, Uribe A. Trophic<br />

actions of E2 prostaglandins in the rat gastrointestinal<br />

mucosa. A quantitative morphologic study.<br />

Gastroenterology 1985; 89: 1393–1399.<br />

58. Vanderhoof JA, Euler AR, Park JHY, Grandjean CJ.<br />

Augmentation of mucosal adaptation following massive<br />

small bowel resection by 16,16-dimethyl-PG E2 in the<br />

rat. Digestion 1987; 36: 213–219.<br />

59. Vanderhoof JA, Park JHY, Grandjean CJ. Effect of zinc<br />

deficiency on mucosal hyperplasia following 70% resection.<br />

Am J Clin Nutr 1986; 44: 670–677.<br />

60. Ford WDA, Boelhouwcr RU, King WWK et al. Total<br />

parenteral nutrition inhibits intestinal adaptive hyperplasia<br />

in young rats reversal by feeding. Surgery 1983;<br />

96: 527–534.<br />

61. Grimble GK. Rees RG, Keohane PP et al. Effects of<br />

peptide chain length on absorption of egg protein<br />

hydrolysates in the normal human jejunum.<br />

Gastroenterology 1987; 92: 136–142.<br />

62. Silk DBA, Fairclough PD, Clark ML et al. Uses of a<br />

peptide rather than a free amino-acid nitrogen source in<br />

chemically defined elemental diets. J Parenter Enteral<br />

Nutr 1980; 4: 548–553.<br />

63. Fairelough PD, Hegarty JE, Silk DBA, Clark ML.<br />

Comparison of the absorption of two protein<br />

hydrolysates and their effects on water and electrolyte<br />

movements in the human jejunum. Gut 1980: 21:<br />

829–834.<br />

64. Poullain MG, Cezard JP, Roger l, Mendy F. Effect of<br />

whey proteins, their oligopeptide hydrolysates and free<br />

amino acid mixtures on growth and nitrogen in fed and<br />

starved rats. J Parenter Enteral Nutr 1989; 13: 382–386.<br />

65. Bines J, Francis D, Hill D. Reducing parenteral requirement<br />

in children with short bowel syndrome: impact of<br />

an amino acid-based complete infant formula. J Pediatr<br />

Gastroenterol Nutr 1998; 26: 123–128.<br />

66. Andorsky DJ, Lund DP, Lillehei CW et al. Nutritional<br />

and other postoperative management of neonates with<br />

short bowel syndrome correlates with clinical<br />

outcomes. J Pediatr 2001; 139: 27–33.<br />

67. Bach AC, Babayan VK. Medium-chain triglycerides: an<br />

update. Am J Clin Nutr 1982; 36: 950–962.<br />

68. Jeppesen PB, Mortensen PB. The influence of a<br />

preserved colon on the absorption of medium chain fat<br />

in patients with small bowel resection. Gut 1998; 43:<br />

478–483.<br />

69. Vanderhoof JA, Grandjean CJ, Burkley KT et al. Effect of<br />

high-percentage medium-chain triglyceride diet on<br />

mucosal adaptation following massive bowel resection<br />

in rats. J Parenter Enteral Nutr 1984; 8: 685–689.


476<br />

Short-bowel syndrome<br />

70. Grey VL, Garofalo C, Greenberg GR, Morin CL. The<br />

adaptation of the small intestine after resection in<br />

response to free fatty acids. Am J Clin Nutr 1984; 40:<br />

1235–1242.<br />

71. Hart MH, Grandjean CL, Park JHY et al. Essential fatty<br />

acid deficiency and post-resection mucosal adaptation<br />

in the rat. Gastroenterology 1987; 94: 682–687.<br />

72. Goulet O, Richard S, Nezelof C et al. Parenteral<br />

nutrition associated liver disease in children with<br />

massive loss or intestine. Clin Nutr 1989; 8: 24 (abstr).<br />

73. Stanko RT, Nathan G, Mendelow H, Adibi SA.<br />

Development of hepatic cholestasis and fibrosis in<br />

patients with massive loss of intestine supported by<br />

prolonged parenteral nutrition. Gastroenterology 1987;<br />

92: 197–202.<br />

74. Wolf A, Pohlandt F. Bacterial infection: the main cause<br />

of acute cholestasis in newborn infants receiving shortterm<br />

parenteral nutrition. J Pediatr Gastroenterol Nutr<br />

1989; 8: 297–303.<br />

75. Sondheimer JM, Asturias E, Cadnapaphornchai M.<br />

Infection and cholestasis in neonates with intestinal<br />

resection and long-term parenteral nutrition. J Pediatr<br />

Gastroenterol Nutr 1998; 27: 131–137.<br />

76. Braxton C, Lowry SF. Editorial: parenteral nutrition and<br />

liver dysfunction – new insight? J Parenter Enteral Nutr<br />

1995; 19: 3–4.<br />

77. Kaufman SS. Prevention of parenteral nutritionassociated<br />

liver disease in children. Pediatr Transplant<br />

2002; 6: 37–42.<br />

78. Merritt RJ. Medical progress: cholestasis associated with<br />

total parenteral nutrition. J Pediatr Gastroenterol Nutr<br />

1986; 5: 9–22.<br />

79. Colomb V, Jobert-Giraud A, Lacaille F et al. Role of lipid<br />

emulsions in cholestasis associated with long-term<br />

parenteral nutrition in children. J Parenter Enteral Nutr<br />

2000; 24: 345–350.<br />

80. Kaufman SS, Loseke CA, Lupo JV et al. Influence of<br />

bacterial overgrowth and intestinal inflammation on<br />

duration of parenteral nutrition in children with short<br />

bowel syndrome. J Pediatr 1997; 131: 356–361.<br />

81. Goulet O. Parenteral nutrition in pediatrics – indications<br />

and perspectives. Acta Gastroenterol Belg 1999; 62:<br />

210–215.<br />

82. Salas JS, Dozio E, Goulet O et al. Energy expenditure<br />

and substrate utilization in the course of renutrition of<br />

malnourished children. J Parenter Enteral Nutr 1991; 15:<br />

288–293.<br />

83. Vargas JH, Ament ME, Berquist WE. Long-term home<br />

parenteral nutrition in pediatrics: ten years of experience<br />

in 102 patients. J Pediatr Gastroenterol Nutr 1987;<br />

6: 24–32.<br />

84. Colomb V, Goulet O, Ricour C. Home enteral and<br />

parenteral nutrition. Baillière’s Clin Gastroenterol 1998;<br />

122: 877–894.<br />

85. Moukarzel A, Song MK, Buchman AL et al. Excessive<br />

chromium intake in children receiving total parenteral<br />

nutrition. Lancet 1992; 339: 385–388.<br />

86. Buchman AL, Moukarzel AA. Metabolic bone disease<br />

associated with total parenteral nutrition. Clin Nutr<br />

2000; 19: 217–231.<br />

87. Schiano TD, Klang MG, Quesada E et al. Thiamine<br />

status in patients receiving long-term home parenteral<br />

nutrition. Am J Gastroenterol 1996; 91: 2555–2559.<br />

88. Edes TE, Walk BE, Thorton WH, Fritsche KL. Essential<br />

fatty acid sufficiency does not preclude fat-solublevitamin<br />

deficiency in short-bowel syndrome. Am J Clin<br />

Nutr 1991; 53: 499–502.<br />

89. Niv Y, Charash B, Sperber AD, Oren M. Effect of<br />

octreotide on gastrostomy, duodenostomy, and cholecys-<br />

tostomy effluents: a physiologic study of fluid and electrolyte<br />

balance. Am J Gastroenterol 1997; 92: 2107–2111.<br />

90. Colomb V, Fabeiro M, Dabbas M et al. Central venous<br />

catheter-related infections in children on long-term<br />

home parenteral nutrition: incidence and risk factors.<br />

Clin Nutr 2000; 19: 355–359.<br />

91. Capron JP, Gineston JL, Herve MA. Metronidazole in<br />

prevention of cholestasis associated with total<br />

parenteral nutrition. Lancet 1983; 1: 446–447.<br />

92. Spagnuolo MI, Iorio R, Vegnente A, Guarino A.<br />

Ursodeoxycholic acid for treatment of cholestasis in<br />

children on long-term total parenteral nutrition: a pilot<br />

study. Gastroenterology 1996; 111: 716–719.<br />

93. Goulet O, Duhamel JF. Ricour C. Nutritional problems.<br />

In Tinker J, Rapin M, eds. Care of the Critically Ill<br />

Patient. New York: Springer Verlag, 1992: 1415–1436.<br />

94. Advenier E, Landry C, Colomb V et al. Aluminium<br />

contamination of parenteral nutrition and aluminium in<br />

children on long-term parenteral nutrition. J Pediatr<br />

Gastroenterol Nutr 2003; 36: 448–453.<br />

95. Goulet O, de Potter S, Antebi H et al. Long-term efficacy<br />

and safety of a new olive oil-based intravenous fat<br />

emulsion in pediatric patients: a double-blind<br />

randomized study. Am J Clin Nutr 1999; 70: 338–345.<br />

96. Forchielli ML, Gura KM, Sandler R, Lo C. Aminosyn PF<br />

or Trophamine: which provides more protection from<br />

cholestasis associated with total parenteral nutrition? J<br />

Pediatr Gastroenterol Nutr 1995; 21: 374–382.<br />

97. Dabbas M, Jobert-Giraud A, Colomb V et al. Intestinal<br />

absorption in children with short bowel syndrome. Clin<br />

Nutr 1998; 17 (Suppl 1): 45 (abstr).<br />

98. Liefaard G, Heineman E, Molenaar JC, Tibboel D.<br />

Prospective evaluation of the absorptive capacity of the<br />

bowel after major and minor resections in the neonates.<br />

J Pediatr Surg 1995; 30: 388–391.<br />

99. Barilla DE, Notz C, Kennedy D, Pak CYC. Renal oxalate<br />

excretion following oral oxalate loads in patients with<br />

ileal disease and with renal and absorptive<br />

hypercalciurias. Effect of calcium and magnesium. Am J<br />

Med 1978; 64: 579–585.<br />

100. Rockwell GF, Campfield T, Nelson BC, Uden PC.<br />

Oxalogenesis in parenteral nutrition solution<br />

components. Nutrition 1999; 14: 836–839.<br />

101. Hudson M, Pocknee R, Mowat NAG. D-lactic acidosis in<br />

short bowel syndrome – an examination of possible<br />

mechanisms. Q J Med 1990; 74: 157–163.<br />

102. From the Centers for Disease Control and Prevention.<br />

Lactic acidosis traced to thiamine deficiency related to<br />

nation wide shortage of multivitamins for total<br />

parenteral nutrition – United States 1997. JAMA 1997;<br />

278: 109, 111.<br />

103. Kadakia SC. D-Lactic acidosis in a patient with<br />

jejunoileal bypass. J Clin Gastroenterol 1995; 20:<br />

154–156.<br />

104. Marteau P, Messing B, Arrigoni E et al. Do patients with<br />

short-bowel syndrome need a lactose-free diet?<br />

Nutrition 1997; 13: 13–16.<br />

105. Scolapio JS, Nguyen JH, Steers J, Ukleja A. Success with<br />

intestinal failure: from adaptation to transplantation.<br />

Dig Dis 1999; 17: 107–112.<br />

106. Herek O. Saccaromyces boulardii: a possible addition to<br />

the standard treatment and prophylaxis of enterocolitis<br />

in Hirschsprung’s disease? Pediatr Surg Int 2002; 18:<br />

567.<br />

107. Dellert SF, Farrell MK, Specker BL, Heubi JE. Bone<br />

mineral content in children with short bowel syndrome<br />

after discontinuation of parenteral nutrition. J Pediatr<br />

1998; 132: 516–519.


108. Goulet O, Baglin-Gobet S, Jais JP et al. Outcome and<br />

long-term growth after extensive small bowel resection<br />

in the neonatal period: a survey of 87 children. Eur J<br />

Pediatr Surg 2004; in press<br />

109. Leonberg BL, Chuang E, Eicher P et al. Long-term<br />

growth and development in children after home<br />

parenteral nutrition. J Pediatr 1998; 132: 461–466.<br />

110. Festen S, Brevoord JC, Goldhoorn GA et al. Excellent<br />

long-term outcome for survivors of apple peel atresia. J<br />

Pediatr Surg 2002; 37: 61–65.<br />

111. Dorney SFA, Ament MA, Berquist WE et al. Improved<br />

survival in very short small bowel of infancy with use<br />

of long-term parenteral nutrition. J Pediatr 1985; 107:<br />

521–525.<br />

112. Surana R, Quinn FMJ, Puri P. Short-gut syndrome:<br />

intestinal adaptation in a patient with 12 cm of<br />

jejunum. J Pediatr Gastroenterol Nutr 1994; 19: 246–249.<br />

113. Hancock BJ, Wiseman NE. Lethal short-bowel<br />

syndrome. J Pediatr Surg 1990; 25: 1131–1134.<br />

114. Ricour C, Duhamel JF, Arnaud-Battandier F et al. Enteral<br />

and parenteral nutrition in the short bowel syndrome in<br />

children. World J Surg 1985; 9: 310–315.<br />

115. Goulet O, Ricour C. The short bowel syndrome. In Buts<br />

JP, Sokal E, eds. Management of Digestive and Liver<br />

Disorders in Infants and Children. Elsevier, 1993:<br />

307–318.<br />

116. Goulet O, Revillon Y, Jan D et al. Which patients need<br />

small bowel transplantation for neonatal short bowel<br />

syndrome? Transplant Proc 1992; 24: 1058–1059.<br />

117. Taylor B, Murphy GM, Dowling RH. Pituitary hormones<br />

and the small bowel: effect of hypophysectomy on<br />

intestinal adaptation to small bowel resection in the rat.<br />

Eur J Clin Invest 1979; 9: 115–127.<br />

118. Scow RO, Hagan SN. Effect of testosterone propionate<br />

and growth hormone on growth and chemical<br />

composition of muscle and other tissues in<br />

hypophysectomized male rats. Endocrinology 1965; 77:<br />

852–858.<br />

119. Shulman DI, Shih Hu C, Duckett G, Lavallee-Grey M.<br />

Effects of short-term growth hormone therapy in rats<br />

undergoing 75% small intestinal resection. J Pediatr<br />

Gastroenterol Nutr 1992; 14: 3–11.<br />

120. Ulshen MA, Dowling RH, Fuller CR et al. Enhanced<br />

growth of small bowel in transgenic mice<br />

overexpressing bovine growth hormone.<br />

Gastroenterology 1993; 104: 973–980.<br />

121. Benhamou PH, Canarelli JP, Leroy C et al. Stimulation<br />

by recombinant human growth hormone of growth and<br />

development of remaining bowel after subtotal ileojejunectomy<br />

in rat. J Pediatr Gastroenterol Nutr 1994, 18:<br />

446–452.<br />

122. Vanderhoof JA, Kollman KA, Griffin S, Adrian TE.<br />

Growth hormone and glutamine do not stimulate<br />

intestinal adaptation following massive small bowel<br />

resection in the rat. J Pediatr Gastroenterol Nutr 1997;<br />

25: 327–331.<br />

123. Lobie PE, Briepohl S, Waters MJ. Growth hormone<br />

receptors in rat intestinal tract. Endocrinology 1990;<br />

126: 299–306.<br />

124. Laburthe M, Rouyer-Fessard C, Gammeltoft S. Receptors<br />

for insulin-like growth factors I and II in rat gastrointestinal<br />

epithelium. Am J Physiol 1988; 254:<br />

G457–462.<br />

125. Termanini B, Nardi RV, Finan TM. Insulin-like growth<br />

factor receptors in rabbit gastro-intestinal tract:<br />

characterization and autoradiographic localization.<br />

Gastroenterology 1990; 99: 51–60.<br />

126. Vanderhoof JA., McCusrer RH, Clark R et al. Truncated<br />

and native IGF-I enhance mucosal adaptation after<br />

References 477<br />

jejunoileal resection. Gastroenterology 1992; 102:<br />

1949–1956.<br />

127. Lemmey AB, Martin AA, Read TC et al. IGF-I and the<br />

truncated analogue des(1-3) 16F-I enhance growth in<br />

rats after gut resection. Am J Physiol 1991; 260:<br />

E213–E219.<br />

128. Inoue Y, Copeland EM, Souba WW. Growth hormone<br />

enhances amino-acid uptake by the human small<br />

intestine. Ann Surg 1994; 219: 715–724.<br />

129. Challacombe DN, Wheeler EE. The trophic action of<br />

human growth hormone duodenal mucosa cultured in<br />

vitro. J Pediatr Gastroenterol Nutr 1995; 21: 50–53.<br />

130. Byrne TA, Persiger RL, Young LS et al. A new treatment<br />

for patients with short bowel syndrome. Ann Surg 1995;<br />

222: 243–255.<br />

131. Scolapio JS, Camilleri M, Fleming CR et al. Effect of<br />

growth hormone, glutamine, and diet on adaptation in<br />

short-bowel syndrome: a randomized, controlled study.<br />

Gastroenterology 1997; 113: 1074–1081.<br />

132. Seguy D, Vahedi K, Crenn P et al. Growth hormone<br />

benefit in very short bowel patients a randomized<br />

controlled trial. Clin Nutr 1999.<br />

133. Szkudlarek J, Jeppesen P-B, Mortensen P-B. Effect of<br />

high dose growth hormone with glutamine and no<br />

change in diet on intestinal absoption in short bowel<br />

patients: a randomised, double blind, crossover, placebo<br />

controlled study. Gut 2000; 47: 199–205.<br />

134. Ellegard L, Bosaeus I, Nordgren S, Bengtsson BA. Lowdose<br />

recombinant human growth hormone increases<br />

body weight and lean body mass in patients with short<br />

bowel syndrome. Ann Surg 1997; 225: 88–96.<br />

135. Socha J, Ksiazyk J, Fogel WA et al. Is growth hormone a<br />

feasible adjuvant in the treatment of children after small<br />

bowel resection? Clin Nutr 1996; 15: 185–188.<br />

136. Dabbas-Tyan M, Colomb V, Rosilio M et al. Evaluation of<br />

the effect of recombinant human growth hormone<br />

(rhGH) treatment of children with short bowel<br />

syndrome. J Pediatr Gastroenterol Nutr 2000; 31:<br />

5165–5166.<br />

137. Drucker DJ. Gut adaptation and the glucagon-like<br />

peptides. Gut 2002; 50: 428–435.<br />

138. L’Heureux MC, Brubaker PL. Therapeutic potential of<br />

the intestinotropic hormone, glucagon-like peptide-2.<br />

Ann Med 2001; 33: 229–35.<br />

139. Jeppesen PB, Hartmann B, Hansen BS et al. Impaired<br />

meal stimulated glucagon-like peptide 2 response in<br />

ileal resected short bowel patients with intestinal<br />

failure. Gut 1999; 45: 559–563.<br />

140. Jeppesen PB, Hartmann B, Thulesen J et al. Glucagonlike<br />

peptide 2 improves nutrient absorption in shortbowel<br />

patients with no colon. Gastroenterology 2001;<br />

120: 806–815.<br />

141. Delaney HM, Parker JG, Gliedman ML. Experimental<br />

massive intestinal resection: comparison of surgical<br />

measures and spontaneous adaptation. Arch Surg 1970;<br />

101: 599–604.<br />

142. Persemlidis D, Kark AE. Antiperistaltic segments for the<br />

treatment or short bowel syndrome. Am J Gastroenterol<br />

1974; 62: 526–530.<br />

143. Stacchini A, Dido LJ, Primo ML et al. Artificial<br />

sphincter as surgical treatment for experimental<br />

massive resection of small intestine. Am J Surg 1982;<br />

143: 721–26.<br />

144. Ricotta J, Zuidcma FD, Gadacz RT, Sadri D.<br />

Construction of an ileocaecal valve and its role in<br />

massive resection of the small intestine. Surg Gynecol<br />

Obstet 1981; 152: 310–314.<br />

145. Waddell WR, Kern F, Halgrimson CG, Woodbury JJ. A<br />

simple jejunocolic ‘valve.’ For relief of rapid transit and


478<br />

Short-bowel syndrome<br />

the short bowel syndrome. Arch Surg 1970; 100:<br />

438–444.<br />

146. Hakami M, Moslehy A, Mosavy SH. Reversed jejunal<br />

segment used to treat the short bowel syndrome. Am<br />

Surg 1975; 41: 432–435.<br />

147. Panis Y, Messing B, Rivet P et al. Segmental reversal of<br />

the small bowel as an alternative to intestinal transplantation<br />

in patients with short bowel syndrome. Ann Surg<br />

1997; 225: 401–407.<br />

148. Trinkle JK, Bryant LR. Reversed colon segment in an<br />

infant with massive small bowel resection: a case<br />

report. J Ky Med Assoc 1967; 65: 1090–1091.<br />

149. Garcia VF, Templeton JM, Eichelberger MR et al. Colon<br />

interposition for the short bowel syndrome. J Pediatr<br />

Surg 1981; 16: 994–995.<br />

150. Glick PL, de Lorimier AA, Adzick NS, Harrison MR.<br />

Colon interposition: an adjuvant operation for short–gut<br />

syndrome. J Pediatr Surg 1984; 19: 719–725.<br />

151. Brolin RE. Colon interposition for extreme short bowel<br />

syndrome: a case report. Surgery 1986; 100: 576–580.<br />

152. Georgeson K, Halpin D, Figueroa R et al. Sequential<br />

intestinal lengthening procedures for refractory short<br />

bowel syndrome. J Pediatr Surg 1994; 29: 316–320;<br />

discussion 320–311.<br />

153. Thompson JS, Langnas AN, Pinch LW et al. Surgical<br />

approach to short-bowel syndrome. Experience in a<br />

population of 160 patients. Ann Surg 1995; 222:<br />

600–605; discussion 605–607.<br />

154. Devine RM, Kelly KA. Surgical therapy of the short<br />

bowel syndrome. Gastroenterol Clin North Am 1989; 18:<br />

603–618.<br />

155. Bianchi A. Intestinal loop lengthening a technique for<br />

increasing small intestinal length. J Pediatr Surg 1980;<br />

15: 145–151.<br />

156. Bianchi A. Longitudinal intestinal lengthening and<br />

tailoring: results in 20 children. J R Soc Med 1997; 90:<br />

429–432.<br />

157. Weber TR. Isoperistaltic bowel lengthening for short<br />

bowel syndrome in children. Am J Surg 1999; 178:<br />

600–604.<br />

158. Thompson JS, Pinch LW, Young R, Vanderhoof JA. Longterm<br />

outcome of intestinal lengthening. Transplant Proc<br />

2000; 32: 1242–1243.<br />

159. Waag KL, Hosie S, Wessel L. What do children look like<br />

after longitudinal intestinal lengthening? Eur J Pediatr<br />

Surg 1999; 9: 260–262.<br />

160. Figueroa-Colon R, Harris PR, Birdsong E et al. Impact of<br />

intestinal lengthening on the nutritional outcome for<br />

children with short bowel syndrome. J Pediatr Surg<br />

1996; 31: 912–916.<br />

161. Lawrence JP, Dun SP, Billmire DF et al. Isolated liver<br />

transplantation for liver failure in patients with short<br />

bowel syndrome. J Pediatr Surg 1994; 29: 751–753.<br />

162. Horslen SP, Sudan DL, Iyer KR et al. Isolated liver transplantation<br />

in infants with end-stage liver disease associated<br />

with short bowel syndrome. Ann Surg 2002; 235:<br />

435–439.<br />

163. Vanderhoof JA. Short bowel syndrome in children and<br />

small intestinal transplantation. Pediatr Clin North Am<br />

1996; 43: 533.<br />

164. Grant D. Intestinal transplantation: 1997 Report of the<br />

International Registry. Transplantation 1999; 15:<br />

1061–1064.<br />

165. Goulet O, Jan D, Lacaille F et al. Intestinal<br />

transplantation in children: preliminary experience in<br />

Paris. J Parenter Enteral Nutr 1999; 23: S121–125.<br />

166. Langnas AN, Sudan DL, Kaufman SS et al. Intestinal<br />

transplantation: a single center experience. Transplant<br />

Proc 2000; 32: 1228.<br />

167. Goulet O, Lacaille F, Jan D, Ricour C. Intestinal<br />

transplantation : indications, results and strategy. Curr<br />

Opin Clin Nutr Metab Care 2000; 3: 329–333.


30<br />

Introduction<br />

Lymphonodular hyperplasia<br />

Jorma Kokkonen<br />

Small lymphoid nodules inside the mucosa lining<br />

the gastrointestinal tract are normal endoscopic<br />

findings among children. 1 However, greatly<br />

enlarged lymphoid nodules or a mass covering<br />

most of the view area and described as lymphoid<br />

nodular hyperplasia (LNH) may be considered<br />

abnormal. The most typical sites for LNH are the<br />

bulb of the duodenum, the colon and the terminal<br />

ileum, where small nodules are visible in nearly<br />

all subjects. 2,3 Assessment by videoendoscope has<br />

made LNH now perhaps the most common finding<br />

seen in pediatric examinations. During the era of<br />

X-ray examinations and fiberoscopes only heavy<br />

accumulations of lymphoid tissue were diagnosed,<br />

most cases with minor lesions remaining undiagnosed.<br />

4 As we may examine only patients with<br />

long-term symptoms, for understandable reasons,<br />

we will never know the true prevalence of LNH in<br />

healthy subjects.<br />

The clinical significance of LNH has long<br />

remained obscure. It was previously considered as<br />

normal or age-related in pediatric patients diagnosed<br />

as infants or small children. 2,4 Even now it<br />

may be considered as a finding peculiar to children,<br />

being only rarely diagnosed in adults. 5 In<br />

earlier literature, intestinal LNH was a rare<br />

finding, but was clearly associated with congenital<br />

or acquired immunodeficiency states, including<br />

IgA deficiency. 6–8 It has also been seen in HIV<br />

subjects even before the development of AIDS. 9<br />

LNH may now be considered as a state indicating<br />

an ongoing or earlier up-regulated immune activity<br />

of the underlying mucosa. 10<br />

In a microscopic examination, a biopsy sample<br />

from the LNH site reveals follicles with or without<br />

germinal centers but usually no dense mononuclear<br />

or eosinophilic inflammation. LNH repre-<br />

sents a multiplication of Peyer’s patches, an accumulation<br />

of lymphoid cells specialized in handling<br />

antigens cross the mucosal barrier and regulation<br />

of the innate immune response. Recent evidence<br />

suggests that in subjects with LNH the local<br />

cytokine populations are skewed, showing similarities<br />

both with celiac disease and atopic allergy.<br />

Assessment<br />

To date there has been no objective method of<br />

measuring or evaluating the amount of intramural<br />

accumulations of lymphoid tissue downstream of<br />

the intestinal mucosa. Clusters of lymphoid nodules<br />

seen during an endoscopic examination may<br />

be considered significant. LNH may be considered<br />

as a hypernormal phenomenon, an enlargement<br />

and multiplication of normal lymphoid tissue<br />

(Figures 30.1a and b, 30.2a and b, 30.3a and b).<br />

Wakefield et al used a four-step classification to<br />

describe the prominence of LNH at the terminal<br />

ileum. 11 We have also used this practical grading,<br />

using the following description: grade 0, no<br />

lymphoid follicles; grade 1, mild – lymphoid follicles<br />

dispersed on the walls; grade 2, moderate,<br />

lymphoid follicles filling the walls; and grade 3,<br />

severe – terminal ileum massed with lymphoid<br />

tissue, the valve protruding. However, in some<br />

patients there are segments where the one wall<br />

may be massed with lymphoid tissue while the<br />

other wall has no visible lymphoid accumulation.<br />

The same classification may also be applied to the<br />

bulb of the duodenum and colon, where mostly<br />

only grades 0–2 are usually seen.<br />

A patchy and segmental appearance is a typical<br />

feature of LNH. In the foregut the nodules are most<br />

often seen in the bulb. Only in a quarter of cases<br />

479


480<br />

Lymphonodular hyperplasia<br />

(a) (b)<br />

(c)<br />

does the lesion continue to the descending duodenum.<br />

In the terminal ileum, LNH is usually<br />

restricted to an area of about 10–15 cm proximal to<br />

the valve. If massed just above the valve it usually<br />

becomes nodular and disappears entirely as it<br />

proceeds proximally in the ileum.<br />

LNH must be actively sought during the examination.<br />

The underlying mucosa is healthy in that<br />

there is usually no inflammation. The visible LNH<br />

bubbles may be hidden between the folds. During<br />

the videoendoscopic examination the lesion is<br />

Figure 30.1 Normally in a close endoscopic view of the<br />

bulb of the duodenum, the villous architecture is visible<br />

on a plain mucosa (a). In lymphoid nodular hyperplasia<br />

(LNH) small bubbles are dispersed along the walls covering<br />

otherwise normal mucosa (b). A biopsy sample from<br />

the site of the LNH reveals a lymphoid nodule with a<br />

germinal center but surrounded by normal villi (c).<br />

sought and assessed after the segments of the areas<br />

inspected are filled with air to the maximum possible<br />

and the nodularity is evaluated by sight. In the<br />

terminal ileum a protruding valve may reveal LNH<br />

inside the small intestine.<br />

Histology and immunohistochemistry<br />

A typical feature in the microscopic examination<br />

of a biopsy sample from a patient with LNH is an<br />

increased occurrence and enlargement of<br />

lymphoid follicles with or without germinal<br />

centers (Figures 30.1c, 30.2c, 30.3c). The centers<br />

are usually grossly enlarged and reactive as indicated<br />

by numerous tingle body macrophages. In<br />

advanced cases the outer margins of the T-cell<br />

zone may not be as well defined as in normal or<br />

mild cases, with the lymphoid compartment<br />

extending into the adjacent villi. There may also


(a) (b)<br />

(c)<br />

be disruption but not destruction of adjacent<br />

crypts, the number of which may be decreased.<br />

Typical cases have no villous atrophy, no slight<br />

villous abnormalities or mononuclear or even<br />

eosinophilic infiltration, as seen in celiac<br />

disease. 12 The histology of the colonic samples is<br />

quite similar, subepithelial apoptosis/debris being<br />

increased as well.<br />

LNH may also be associated with other disorders<br />

such as celiac disease or colitis, which have their<br />

Histology and immunohistochemistry 481<br />

Figure 30.2 On the healthy mucosa of the colon the<br />

vascular pattern is clear (a). Lymphoid nodular hyperplasia<br />

may cover the mucosa as a dense madras (b) or be<br />

sparsely dispersed. A biopsy sample in a histological<br />

examination reveals accumulation of lymphoid tissue (c).<br />

own characteristics. LNH seems to have a weak<br />

predilection for IgE-mediated diseases or IgEprone<br />

subjects; these may have excessive<br />

eosinophilic cells in the histology of their<br />

samples. 13 These patients may also show markers<br />

of atopic food allergy, namely positive skin prick<br />

tests or food-specific IgE class antibodies.<br />

However, these patients are a minority.<br />

An increased density of intraepithelial lymphocytes<br />

has been measured in the biopsy sample<br />

sites of LNH, especially γδ + T-cell receptor (TCR)bearing<br />

T cells. 14,15 Accordingly, the proportion of<br />

γδ + T cells of the total amount of CD3 + T cells, i.e.<br />

the γδ + /CD3 + ratio, is greater than in the controls.<br />

Another type of intraepithelial cell that is<br />

increased in LNH subjects is the cytotoxic TIA-1expressing<br />

cell. 16<br />

The rise in both types of intraepithelial cell seems<br />

to be more associated with the underlying disorder<br />

than with the LNH itself. Indeed, in recent reports<br />

the numbers of intraepithelial γδ + cells as well as


482<br />

Lymphonodular hyperplasia<br />

(a) (b)<br />

(c)<br />

TIA-1 presenting T cells have been observed to be<br />

high in patients with food allergy. 13,15 However, we<br />

found the increased densities in untreated cases,<br />

but low densities in the subjects on an elimination<br />

diet. These findings indicate that in the delayed<br />

and local types of food allergy, as in celiac disease,<br />

the increment of γ/δ + T cells is associated with the<br />

activity of the disease. 17,18 However, even after an<br />

Figure 30.3 On the mucosa of the terminal ileum there<br />

are nearly always tiny lymphoid nodules (a), but in states<br />

of lymphoid nodular hyperplasia they are highly enlarged<br />

(b) and may mass on the walls. The biopsy may be filled<br />

with lymphoid tissue as large nodules (c) but interspersed<br />

normal villi may be found.<br />

elimination diet, LNH seems to linger. Whether it<br />

is permanent or abates with time remains<br />

unknown.<br />

To date, the exact pathogenetic significance of γ/δ +<br />

and TIA-1 T cells still remains obscure. Being typically<br />

increased in untreated celiac disease and<br />

food allergy, they may have profound significance<br />

in the pathogenesis of these disorders.<br />

Pathophysiology<br />

The segmental or patchy appearance of LNH probably<br />

underlines the fact that the lesion develops as<br />

an in situ activation of the innate immune<br />

response leading to multiplication of the lymphoid<br />

tissue. As hyper-reactivity to food-borne antigens<br />

is the most common state underlying LNH, this<br />

suggests that the lesion develops at the contact<br />

between luminal allergens and the immunologically<br />

activated cells inside the mucosa. Because of<br />

the passage of food juice and the timing of its<br />

contact with the mucosa, the most vulnerable<br />

places may be the bulb of the duodenum and the


terminal ileum and colon, giving a reasonable<br />

explanation for the spread of the lesion in the<br />

gastrointestinal tract.<br />

In a recent study we found evidence to suggest<br />

that, in food allergy as in celiac disease, cellmediated<br />

immunity may be active and may cause<br />

the symptoms. However, we also found evidence<br />

showing that in LNH patients humoral immunity<br />

is also activated. The IgG and IgA class antibodies<br />

against milk proteins were increased in<br />

patients with LNH of the bulb of the duodenum. 19<br />

These antibodies may have no pathogenetic<br />

significance in the process, their role being<br />

simply secondary, owing to the increment of B<br />

cells in and around the lymphoid follicles and<br />

through the interaction between B and T<br />

cells. 20,21 However, the humoral response relating<br />

to LNH may be dichotomous. We also found<br />

lower-than-normal levels of IgA and IgG class<br />

antibodies against milk allergens in subjects<br />

reacting rapidly in a blind challenge test.<br />

Lacking villous atrophy and/or mononuclear<br />

inflammation, the symptoms in typical LNH<br />

cases may be due to an imbalance in cytokine<br />

cascades. LNH patients have more intraepithelial<br />

γδ-TCR + cells, which can produce a multiplicity<br />

of cytokines, including interleukin (IL)-2, interferon<br />

(IFN)-γ, tumor necrosis factor (TNF)-α, IL-4,<br />

IL-10 and transforming growth factor (TGF)-β,<br />

although to date there has been no detailed study<br />

of the subject. In infants with food allergy, Hauer<br />

et al showed an up-regulation of IFN-γ and, to a<br />

lesser extent, IL-4. 22 We found evidence in two<br />

experiments to show that, in patients with LNH<br />

and food allergy, the free secretion of IL-6 as<br />

measured in a biopsy sample was increased, as in<br />

celiac patients, but we failed to demonstrate any<br />

difference in the secretion of regulatory cytokines<br />

such as IL-10 and TGF-β. Examining the biopsy<br />

samples by a counter-polymerase chain reaction<br />

(PCR) method we found an up-regulation of<br />

chemokine receptor 4 (CCR-4) and IL-6 and<br />

down-regulation of IL-2 and IL-18 in LNH/food<br />

allergy cases (Paajanen et al, personal communication).<br />

Even in this study we saw no change in<br />

the regulatory cytokines as compared to the<br />

controls. At present, it seems possible that in<br />

LNH patients there is an imbalance in pre-allergic<br />

and pre-inflammatory cytokine function on the<br />

mucosa.<br />

Differential diagnosis 483<br />

Unlike patients with celiac disease, subjects with<br />

LNH and food allergy display no increased prevalence<br />

of the HLA DQ-2 antigen (A0501/B0201). 14<br />

HLA-DR seems to be only slightly up-regulated in<br />

the duodenal mucosa in these patients. There<br />

currently seems to be no evidence for genetic<br />

selection of LNH subjects. It seems clear that the<br />

process leading to an accumulation of lymphoid<br />

cells and tissue may take place in any subject with<br />

a temporary or permanent disorder in the protective<br />

mechanisms of the mucosal layer, leading to<br />

invasion of luminal antigens and up-regulation of<br />

the immune response in situ in any part of the<br />

gastrointestinal tract.<br />

Differential diagnosis<br />

In our series of 724 consecutive patients over 4<br />

years, LNH was diagnosed in 135 (19%) in all<br />

(Table 30.1). The incidence was highest among the<br />

patients with food allergy, being found in more<br />

than 50% of the definitely diagnosed cases. It was<br />

found equally among the treated and untreated<br />

cases. As in the other centers regularly using endoscopic<br />

examinations to diagnose celiac disease,<br />

LNH was found in 10% of cases. The proportion<br />

was the same in both main types of inflammatory<br />

bowel disease (IBD) as well as in patients in whom<br />

we could make no definite diagnosis, even after<br />

rigorous examination.<br />

LNH may be spread throughout the colon or as<br />

patches at any of the main segments. Among 301<br />

consecutive subjects undergoing colonoscopy, we<br />

found 36 children with diffuse disease and 101<br />

with segmental lesions. The transverse colon was<br />

the most common site for patchy lesions, followed<br />

by the cecum. Even in the colon the incidence of<br />

LNH was greatest among the patients with food<br />

allergy as diagnosed by history and a positive elimination–challenge<br />

test, rising to 77% of all cases.<br />

The patients also showed a 50% coincidence with<br />

diagnosed LNH in the foregut.<br />

Within the spectrum of IBD, a quarter of patients<br />

with colitis showed LNH, but only a few presented<br />

with Crohn’s disease. Colon et al found a similar<br />

association between LNH of the colon and IBD in<br />

13 subjects in a 10-year retrospective study in<br />

which patients were examined mostly by radiographic<br />

methods, while 84 had no underlying<br />

disorder. 4


484<br />

Lymphonodular hyperplasia<br />

Table 30.1 The presence of lymphoid nodular hyperplasia (LNH) on an<br />

endoscopic view in children examined for the first time with gastroduodenoscopy<br />

A new diagnostic entity showing LNH of the colon<br />

in half the patients is chronic constipation.<br />

Among these patients undergoing a challenge<br />

with cow’s milk, a quarter showed a positive<br />

response during the succeeding 4 weeks. The<br />

other definite disorders occasionally showing<br />

LNH on the mucosa of the colon are diabetes,<br />

rheumatoid arthritis, vasculitis and chronic<br />

aggressive hepatitis. Finally, it could be demonstrated<br />

in about 10% of cases without any final<br />

diagnostic conclusions (Table 30.2).<br />

Number of subjects LNH present<br />

n %<br />

Esophagogastroduodenoscopy 724 135 19<br />

untreated food allergy 96 47 49<br />

celiac disease 138 9 7<br />

IBD 76 10 13<br />

Colonoscopy 301 137 46<br />

untreated food allergy 71 55 77<br />

IBD 83 28 34<br />

Ileoscopy 204 105 51<br />

untreated food allergy 51 38 74<br />

IBD 64 20 31<br />

IBD, inflammatory bowel disease<br />

In the colonoscopic examinations lymphoid mass<br />

or LNH of the terminal ileum was diagnosed in<br />

two-thirds of subjects in whom this end-point was<br />

reached. The valve of Bauchini was markedly<br />

protruded inside the colon in a quarter, all with an<br />

abundance of lymphoid tissue inside the ileum.<br />

LNH of the terminal ileum was demonstrated in<br />

two-thirds of the subjects with defined food<br />

allergy, most having a mass of lymphoid tissue on<br />

the wall. It was seen in one in five with colitis,<br />

Table 30.2 Clinical association of endoscopically visible lymphoid nodular hyperplasia (LNH) of the<br />

gastrointestinal tract<br />

Level of gastrointestinal tract LNH present<br />

Foregut<br />

Stomach, normally no nodules <strong>Helicobacter</strong> <strong>pylori</strong> infection<br />

Upper small intestine, normally no nodules food allergy and celiac disease<br />

autoimmune disorders: diabetes, chronic aggressive<br />

hepatitis, rheumatoid arthritis, vasculitis. IgA deficiency,<br />

unknown<br />

Lower gastrointestinal tract<br />

Terminal ileum, normally small nodules dispersed food allergy<br />

evenly on the mucosa inflammatory bowel disease: right-sided or pancolitis<br />

Colon, normally small nodules seen in infants and autoimmune disorders: chronic aggressive hepatitis,<br />

small children rehumatoid arthritis, unknown


while in subjects with Crohn’s disease it was a<br />

surprisingly rare finding. In our series half the<br />

subjects with chronic constipation showed the<br />

lesion in the ileum. Wakefield et al reported an<br />

endoscopic LNH of the terminal ileum with histologically<br />

reactive follicular hyperplasia in 93% of<br />

children with developmental disorders, most<br />

having diarrhea and abdominal pains as well. 23<br />

The same group also provoked a discussion about<br />

the coincidence of the measles virus and especially<br />

the measles vaccine and reactive follicular<br />

hyperplasia and/or ileocolitis. 24,25 In their original<br />

work they found virus antigens in 95% of the children<br />

with developmental disorders and LNH<br />

using the cell reverse transcriptase (RT) PCR<br />

method compared with 11% of the controls.<br />

However, the significance of finding virus particles<br />

with an extremely sensitive methods from the<br />

lymphoid tissue of the gastrintestinal tract<br />

remains open. 26<br />

As stated above, LNH can be seen in association<br />

with congenital or aquired immunodeficiency<br />

states including IgA deficiency and HIV infections.<br />

7–10 It has also been seen in subjects even<br />

before the development of AIDS. Recently, the<br />

lesion has also been described also in patients<br />

with intestinal lymphomas. 27,28<br />

Food allergy<br />

Food allergy may indeed be considered the most<br />

common disease underlying LNH. 29–33 However,<br />

in most cases the question is not about IgE-mediated<br />

disease but a local, T-cell mediated immune<br />

response, an immunological state close to celiac<br />

disease and a delayed immune response against<br />

food-borne allergens. In small children we may<br />

see infiltrations of eosinophils in biopsy samples,<br />

but most of the children of school age have no<br />

increment of eosinophils and mononuclear cells.<br />

Two-thirds of subjects examined by both gastroduodenoscopy<br />

and colonoscopy, and shown by an<br />

elimination and challenge test to have food<br />

allergy, had LNH on the mucosa of the duodenum,<br />

ileum or colon. Only a few cases have had<br />

nodules spread all through the gastrointestinal<br />

tract. This finding confirms the view that food<br />

allergy in children may produce patchy or diffuse<br />

LNH in any part of the gastrointestinal tract. 34<br />

Symptoms<br />

Food allergy 485<br />

LNH in itself may not cause any symptoms, the<br />

underlying and altered immune response and<br />

skewing of the cytokine populations probably<br />

explaining the symptoms. As listed in Table 30.3,<br />

the most common symptoms in upper endoscopies<br />

have been abdominal pain, with or without ‘from<br />

table to toilet’ diarrhea. In the lower endoscopies<br />

the symptoms vary even more, from diarrhea/loose<br />

stools to constipation or blood in stools. A cascade<br />

of symptoms resembling irritable colon has<br />

perhaps been the most prominent seen in our<br />

patients. Indeed, at least in young adults with irritable<br />

colon, Simren et al found symptoms of food<br />

hypersensitivity but no general malabsorption in<br />

their patients, suggesting that some patients with<br />

this symptom cascade might have the same disease<br />

as we demonstrated in our subjects with LNH. 35<br />

Examining a group of school children who had<br />

milk allergy in their infancy and half of the study<br />

group with abdominal symptoms, we also found<br />

no signs of general malabsorption, but the subjects<br />

had lactose intolerance three times more often and<br />

the height of the symptom group was lower than in<br />

the controls. 36 Our interpretation is that the<br />

ongoing immune activity might disturb the epithelial<br />

cell layer with the net result described above.<br />

Table 30.3 Clinical symptoms of patients<br />

examined with gastroduodenoscopy and/or<br />

colonoscopy and having lymphoid nodular<br />

hyperplasia as an endoscopic finding<br />

Upper instestinal symptoms<br />

Recurrent abdominal pains<br />

Nausea/vomiting<br />

Eating disorders<br />

Growth restricted<br />

Anemia<br />

Heartburn/regurgitation<br />

Lower abdominal symptoms<br />

Loose stools, intermittent diarrhea<br />

From table-to-toilet diarrhea<br />

Constipation<br />

Blood in stool<br />

Toiletting difficulties<br />

Lactose intolerance


486<br />

Lymphonodular hyperplasia<br />

However, much investigation is still necessary to<br />

clarify the clinical significance of LNH.<br />

Treatment and prognosis<br />

As stated above LNH may be considered as an<br />

expression of an underlying immune response.<br />

There is no specific treatment. If the lesion is<br />

associated with another diagnosed disorder such<br />

as food allergy or IBD, these patients should be<br />

treated. In our clinical practice we have used<br />

budenoside in cases with LNH and a clinical<br />

picture of irritable colon but without definite<br />

association with food allergy. Most patients have<br />

shown a favorable response, but the approach still<br />

awaits controlled trials. Immunosupressive drugs<br />

in combination with other chemotherapy in<br />

patients with malignant lymphoma have been<br />

REFERENCES<br />

1. Bines JE, Winter HS. Lower endoscopy. In Walker WA,<br />

Durie PR, Hamilton JR, Walker-Smith J, Watkins JB, eds.<br />

Pediatric Gastroenterology. Philadelphia: Decker, 1991:<br />

256–269.<br />

2. Rossi T. Endoscopic examination of the colon in infancy<br />

and childhood. Pediatr Clin North Am 1988; 35:<br />

331–356.<br />

3. Kaplan B, Benson J, Rothstein F et al. Lymphonodular<br />

hyperplasia of the colon as a pathologic finding in children<br />

with lower intestinal bleeding. J Pediatr<br />

Gastroenterol Nutr 1984; 3: 704–708.<br />

4. Colon A, DiPalma J, Leftridge C. Intestinal lymphonodular<br />

hyperplasia of childhood: patterns of<br />

presentation. J Clin Gastroenterol 1991; 13: 163–166.<br />

5. Fine KD, Seidel RH, Do K. The prevalence, anatomic<br />

distribution, and diagnosis of colonic causes of chronic<br />

diarrhea. Gastrointest Endosc 2000; 51: 318–326.<br />

6. Kohler PF, Cook RD, Brown WR et al. Common variable<br />

hypogammaglobulinemia with T-cell nodular lymphoid<br />

interstitial pneumonitis and B-cell nodular lymphoid<br />

hyperplasia: different lymphocyte populations with a<br />

similar response to prednisone therapy. J Allergy Clin<br />

Immunol 1982; 70: 299–305.<br />

7. Jacobson KW, de Shazo RD. Selective immunoglobulin<br />

A deficiency associated with nodular lymphoid<br />

hyperplasia. J Allergy Clin Immunol 1979; 64: 516–521.<br />

8. Lai Ping So A, Mayer L. Gastrointestinal manifestations<br />

of primary immunodeficiency disorders. Semin<br />

Gastrointest Dis 1997; 8: 22–32.<br />

9. Levendoglu H, Rosen Y. Nodular lymphoid hyperplasia<br />

of gut in HIV infection. Am J Gastroenterol 1992; 87:<br />

1200–1202.<br />

reported to diminish the nodules on the gut<br />

mucosa. 28<br />

LNH seems to be anatomic tissue in the sense that<br />

at least in subjects with food allergy, the nodules<br />

remain, even after avoiding the triggering foodstuff.<br />

This conclusion may be judged from the fact<br />

that the nodules are seen nearly as often in children<br />

with treated food allergy as in untreated<br />

cases. Moreover, we have re-examined a group of<br />

children with an abundance of LNH on the<br />

mucosa of the bulb or colon, and saw an equal<br />

amount of lymphoid tissue to that seen before the<br />

treatment. However, the immune activity as<br />

measured by γ/δ + intraepithelial lymphocytes<br />

seemed to diminish during the treatment.<br />

In summary, LNH is a recently described<br />

condition. To date, there is only limited understanding<br />

of the phenomenon and its behavior.<br />

10. Kokkonen J, Karttunen T. Lymphonodular hyperplasia<br />

on the mucosa of the lower gastrointestinal tract in<br />

children: an indication of enhanced immune response?<br />

J Pediatr Gastroenterol Nutr 2002; 34: 42–46.<br />

11. Wakefield AJ, Anthony A, Murch SH et al. Enterocolitis<br />

in children with developmental disorders. Am J<br />

Gastroenterol 2000; 95: 2154–2156.<br />

12. Kokkonen J, Haapalahti M, Laurila K et al. Cow’s milk<br />

protein sensitive enteropathy at school age. J Pediatr<br />

2001; 139: 797–803.<br />

13. Kokkonen J, Ruuska T, Karttunen T et al. Mucosal<br />

pathology of the foregut associating with food allergy<br />

and recurrent abdominal pains in children. Acta Pediatr<br />

2001; 90: 16–21.<br />

14. Kokkonen J, Holm K, Karttunen T et al. Children with<br />

untreated food allergy express a relative increment in<br />

the density of duodenal g/d + T-cells. Scand J<br />

Gastroenterol 2000; 35: 1137–1142.<br />

15. Furlano RI, Anthony A, Day R et al. Colonic CD8 and<br />

gamma delta T-cell infiltration with epithelial damage<br />

in children with autism. J Pediatr 2001; 138: 366–372.<br />

16. Augustin M, Kokkonen J, Karttunen T. Increased<br />

densities of TIA-1 antigen bearing T-cells in duodenal<br />

samples of children with gastrointestinal food allergy. J<br />

Pediatr Gastroenterol Nutr 2001; 32: 11–18.<br />

17. Mäki M, Holm K, Collin P et al. Increase in<br />

gamma/delta T cell receptor bearing lymphocytes in<br />

normal small bowel mucosa in latent coeliac disease.<br />

Gut 1991; 32: 1412–1414.<br />

18. MacDonald T, Spencer J. Evidence for cell-mediated<br />

hypersensitivity as an important pathogenetic<br />

mechanism in food intolerance. Clin Exp Allergy 1995;<br />

25 (Suppl 1): 10–13.


19. Kokkonen J, Tikkanen S, Karttunen TJ et al. Similar high<br />

level of immunoglobulin A and immunoglobulin G class<br />

milk antibodies and increment of local lymphoid tissue<br />

on the duodenal mucosa in subjects with cow’s milk<br />

allergy and recurrent abdominal pains. Pediatr Allergy<br />

Immunol 2002; 13: 129–136.<br />

20. McGhee JR, Lamm ME, Strober W. Mucosal immune<br />

responses. An overview. In Ogra PL, Mestechy J, Lamm<br />

ME, Strober W, Bienenstock J, McGhee JR, eds. Mucosal<br />

Immunology. San Diego: Academic Press, 1999:<br />

485–506.<br />

21. Soothil JF, Stokes CR, Turner MW et al. Predisposing<br />

factors and development of reaginic allergy in infancy.<br />

Clin Allergy 1976; 6: 305–319.<br />

22. Hauer AC, Breese EJ, Walker-Smith JA et al. The<br />

frequency of cells secreting interferon-gamma and interleukin-4,<br />

-5, and -10 in the blood and duodenal mucosa<br />

of children with cow’s milk hypersensitivity. Pediatr Res<br />

1997; 42: 629–638.<br />

23. Wakefield AJ, Murch SH, Anthony A et al.<br />

Ileal–lymphoid–nodular hyperplasia, non-specific<br />

colitis, and pervasive developmental disorder in children.<br />

Lancet 1998; 351: 637–641.<br />

24. Ekbom A, Wakefield AJ, Zack M et al. Perinatal measles<br />

infection and subsequent Crohn’s disease. Lancet 1994;<br />

344: 508–510.<br />

25. Taylor B, Miller E, Farrington CP et al. Autism and<br />

measles, mumps, and rubella vaccine: no epidemiological<br />

evidence for a causal association. Lancet 1999;<br />

353: 2026–2029.<br />

26. Madsen KM, Hviid A, Vestergaard M et al. A<br />

population-based study of measles, mumps, and rubella<br />

vaccination and autism. N Engl J Med 2002; 347:<br />

1477–1482.<br />

References 487<br />

27. Matuchansky C, Touchard G, Lemaire M et al.<br />

Malignant lymphoma of the small bowel associated<br />

with diffuse nodular lymphoid hyperplasia. N Engl J<br />

Med 1985; 313: 166–171.<br />

28. Jonsson OT, Birgisson S, Reykdal S. Resolution of<br />

nodular lymphoid hyperplasia of the gastrointestinal<br />

tract following chemotherapy for extraintestinal<br />

lymphoma. Dig Dis Sci 2002; 47: 2463–2465.<br />

29. Gottrand F, Erkan T, Fabriaux J-P et al. Food-induced<br />

bleeding from lymphonodular hyperplasia of the colon.<br />

Am J Dis Child 1993; 147: 821–823.<br />

30. Beaoui M, Guezmir M, Hamdi M et al. Lymphoid<br />

hyperplasia of the intestine in children. 15 cases. Ann<br />

Pediatr (Paris) 1992; 39: 359–364.<br />

31. Kokkonen J, Karttunen T, Niinimäki A. Lymphonodular<br />

hyperplasia as a sign of food allergy in children. J<br />

Pediatr Gastroenterol Nutr 1999; 29: 57–62.<br />

32. Kokkonen J, Tikkanen S, Savilahti E. Residual intestinal<br />

disease after milk allergy in infancy. J Pediatr<br />

Gastroenterol Nutr 2001; 32: 156–161.<br />

33. Kokkonen J. Lymphonodular hyperplasia of the<br />

duodenal bulb indicates food allergy in children.<br />

Endoscopy 1999; 31: 464–468.<br />

34. Crowe S, Perdue M. Gastrointestinal food hypersensitivity:<br />

basic mechanisms of pathophysiology.<br />

Gastroenterology 1992; 103: 1075–1095.<br />

35. Simren M, Mansson A, Langkilde AM et al. Food-related<br />

gastrointestinal symptoms in the irritable bowel<br />

syndrome. Digestion 2001; 63: 108–115.<br />

36. Tikkanen S, Kokkonen J, Juntti H et al. Status of<br />

children with cow’s milk allergy in infancy by 10 years<br />

of age. Acta Paediatr 2000; 89: 1–7.


31<br />

Introduction<br />

Malnutrition<br />

Michael H N Golden<br />

Nutrition is the process by which dietary<br />

constituents are converted into, maintain and<br />

sustain the body in health. Nutrition and genetic<br />

coding are the determinants of the body’s development<br />

and composition; they modulate and control<br />

its function and enable it to resist disease.<br />

Nutrition is the major variable that determines the<br />

quality of the body – the ‘soil’ in which the ‘seeds’<br />

of disease germinate. The battle between the nutritional<br />

‘soil’ and the etiological ‘seed’ determines<br />

the course of most diseases. Both nutritional state<br />

and genetic endowment should be viewed separately<br />

from the agents of disease. They determine<br />

the internal environment in which the seeds of<br />

disease flourish or fail. Why should measles be a<br />

relatively minor exanthem in Europe and North<br />

America and a deadly killer in Africa, even in<br />

those with normal weight for age? Good nutrition<br />

gives ‘positive health’. There is a whole spectrum<br />

of deteriorating nutrition that gradually compromises<br />

physiological reserve and function. In severe<br />

malnutrition there has been dietary inadequacy<br />

severe enough to compromise every function of<br />

the body including its ability to resist disease<br />

agents. Just as good nutrition leads to positive<br />

health and resistance to disease, poor nutrition<br />

leads to ill health and susceptibility to many<br />

diseases. If a patient has HIV and dies from pneumonia<br />

we conceive of this as a death from HIV. If a<br />

malnourished child dies from pneumonia we<br />

conceive of this death as being due to the pneumonia<br />

itself. This is wrong. The commonest cause<br />

of immunodeficiency is nutritional deprivation –<br />

the underlying cause of death is really malnutrition.<br />

The stark differences between the perception<br />

of HIV and malnutrition as causes of death illustrate<br />

the neglect of nutrition; academic kudos,<br />

research effort and funding interest hardly exist.<br />

The nutritional state, by itself, alters the expression,<br />

course and response to treatment of all<br />

‘primary’ diagnoses. Notwithstanding the diagnostic<br />

label attached to a patient, the accompanying<br />

malnutrition is often the main reason for morbidity<br />

and mortality; and yet, it is frequently the most<br />

amenable to treatment. A clinical history of severe<br />

weight loss or anorexia is not just a diagnostic<br />

pointer making a debilitating disease more probable;<br />

it is a signal calling for active treatment of the<br />

malnutrition and modification of treatment regimens<br />

for the primary diagnosis. The soil needs<br />

care and consideration equal to that given to the<br />

seed. Up to half of all patients admitted to hospital<br />

suffer from anthropometric malnutrition; many<br />

more suffer from micronutrient malnutrition.<br />

If a malnourished patient with carcinoma is given<br />

radiotherapy, low antioxidant nutrient status often<br />

results in intolerable side-effects so that the treatment<br />

cannot continue and the patient dies – from<br />

carcinoma? Treatment has not just failed, it has<br />

been inadequate, if not wrong, because the nutrition<br />

of the patient has been neglected. Ensuring<br />

adequate nutrition of every patient is every pediatrician’s<br />

duty.<br />

Primary malnutrition is a condition of the dependent<br />

and vulnerable who rely on others for nourishment.<br />

It is seen most frequently in the young<br />

child, the elderly and groups such as prisoners and<br />

the mentally ill or disabled. Secondary malnutrition<br />

accompanies any disease that disturbs appetite,<br />

digestion, absorption or utilization of nutrients.<br />

In poor, technologically backward countries,<br />

malnutrition-associated disease is the major cause<br />

of death. More than half of all deaths have anthropometric<br />

malnutrition as the underlying cause; 1<br />

many more are compromised by micronutrient<br />

malnutrition, not associated with weight loss.<br />

Malnutrition stunts the physical and mental<br />

489


490<br />

Malnutrition<br />

development of the majority of the surviving population.<br />

Winston Churchill once said that he could<br />

think of ‘no better investment than to put good<br />

food in the mouths of babies’; that statement is<br />

true today.<br />

The term protein–energy malnutrition has been<br />

used to cover a number of clinical conditions in<br />

both adults and children, including: failure to<br />

thrive, marasmus, cachexia, phthisis, nutritional<br />

dwarfism, kwashiorkor and nutritional or famine<br />

edema. The large number of terms reflect the<br />

emphasis on particular clinical features. However,<br />

regardless of the clinical differences, most of the<br />

physiological, biochemical and body compositional<br />

features are common to all varieties of<br />

severe malnutrition. The principles of classification,<br />

investigation and management are the same<br />

for adults and children. The term protein–energy<br />

malnutrition is best avoided; it carries the false<br />

implication that protein and/or energy deficiencies<br />

are the direct cause of all these conditions.<br />

Nature of nutritional deficiency<br />

If a growing animal is given a diet devoid in folate<br />

there is a consumption of stores; the animal develops<br />

clinical signs characteristic of folate deficiency<br />

(and may die); and the concentration of folate in<br />

the tissues is markedly reduced. However, there is<br />

no effect upon growth or body weight until the<br />

illness is terminal.<br />

If a growing animal is given a diet devoid of<br />

protein there is an immediate cessation of growth<br />

and then loss of weight. When such an animal dies<br />

from protein deficiency, the concentration of<br />

protein in its major tissues is absolutely normal;<br />

the animal will have died from a nutrient deficiency<br />

without any direct evidence of depletion of<br />

the nutrient except for failure to grow or loss of<br />

weight.<br />

The absolute amount of the nutrient within the<br />

body in both folate and protein deficiency is less<br />

than normal, but with folate the reduced amount is<br />

contained within a normally sized body, whereas<br />

with protein the body size has contracted and the<br />

concentration maintained.<br />

There is clearly a fundamental and distinct difference<br />

between these two responses to deprivation<br />

of a single nutrient. Most nutrients can be classified<br />

into those that give a folate-like (type I) or a<br />

protein-like (type II) response (Table 31.1). The<br />

characteristics of the two types of deficiency are<br />

summarized in Table 31.2.<br />

Type I deficiency<br />

When one thinks of deficiency of an essential<br />

nutrient, one automatically considers a type I deficiency.<br />

The diet is deficient. The low intake leads<br />

to a reduction in the tissue concentration. The<br />

metabolic pathways that depend upon this nutrient<br />

are compromised. Characteristic clinical signs<br />

and symptoms develop. The diagnosis is conceptually<br />

straightforward – measure the nutrient<br />

concentration in a tissue, test the metabolic<br />

pathway where the defect lies, demonstrate an<br />

Table 31.1 Classification of nutrients<br />

according to whether the response to a<br />

deficiency is a reduced concentration in the<br />

tissues or a reduced growth rate. Energy<br />

does not fit into the classification. The type I<br />

can be looked upon as ‘functional’ nutrients,<br />

type II as the building blocks of tissue, and<br />

energy as the fuel that runs the system. The<br />

position of some essential nutrients, such as<br />

the essential fatty acids, is not clear<br />

Type I Type II<br />

Iron Nitrogen<br />

Iodine Sulfur<br />

Selenium Essential amino acids<br />

Copper Potassium<br />

Calcium Sodium<br />

Manganese Magnesium<br />

Thiamine Zinc<br />

Riboflavine Phosphorus<br />

Pyridoxine Chlorine<br />

Niacin Water<br />

Ascorbic acid<br />

Retinol<br />

Tocopherol<br />

Calciferol<br />

Folic acid<br />

Cobalamine<br />

Vitamin K


Table 31.2 Characteristics of the different types of nutritional deficiencies<br />

Type 1 Type II<br />

effect of replacing the nutrient in vitro or in vivo, or<br />

recognize the specific clinical signs.<br />

A critical feature of a type I deficiency is that there<br />

is no weight loss or growth failure. One cannot<br />

assume that all is well nutritionally from a growth<br />

chart. There may be one, several or multiple type I<br />

nutritional deficiencies and normal growth. Hence<br />

the devastation of measles in African children<br />

with a normal growth trajectory. Children in the<br />

West who exist on soft drinks, candy (sweets),<br />

crisps and other forms of ‘empty calories’ to<br />

become obese can have multiple micronutrient<br />

deficiencies. Obesity denotes only a chronic positive<br />

energy balance; it is a ‘fat storage disease’, not<br />

‘over-nutrition’ – it is just as likely to be due to<br />

‘under-nutrition’ from an unbalanced diet. Indeed,<br />

it is possible that nutritional deficiency is a cause<br />

of this fat storage disease. Recently in a besieged<br />

city in Angola there was a pellagra epidemic. Only<br />

the fat people developed pellagra (M.H. Golden,<br />

unpublished). To understand how this can occur is<br />

to understand the nature of nutritional deficiency.<br />

Type II deficiency<br />

The position with respect to the type II nutrients is<br />

different. Indeed, none of the strategies that can be<br />

used to diagnose a type I deficiency can be used to<br />

diagnose a deficiency of a type II nutrient. This<br />

gives rise to major difficulties, both conceptual<br />

and practical, when we try to understand, define,<br />

diagnose and treat these deficiencies. Much of the<br />

Nature of nutritional deficiency 491<br />

Tissue level variable Tissue level fixed<br />

Used in specific pathways Ubiquitously used<br />

Characteristic physical signs No characteristic signs<br />

Late or no growth response Immediate growth response<br />

Stored in the body No body store<br />

Buffered response Responds to daily input<br />

Not interdependent Control each other’s balance<br />

Little excretory control Sensitive physiological control<br />

Variable in breast milk with maternal status Fixed concentration in breast milk<br />

controversy that surrounds the definition of a deficiency,<br />

the signs and symptoms of deficiency, the<br />

requirements and the diagnosis of deficiency, of<br />

type II nutrients, stems from attempts to conceive<br />

of them as analogous to the type I nutrients.<br />

The type II nutrients are the fundamental building<br />

blocks of the tissue itself. The tissue cannot be<br />

sustained without the normal complement of these<br />

nutrients, so, with deficiency, the whole tissue is<br />

catabolized and all the components of the tissue<br />

are excreted. When the tissue is to be resynthesized<br />

all the components have to be supplied<br />

in a balanced way. These nutrients can be<br />

thought about as being interdependent, just as the<br />

essential amino acids are usually considered<br />

together under the rubric protein. They have characteristic<br />

ratios that vary over quite a narrow<br />

range. They are required to be absorbed in approximately<br />

the same ratios as occur in the body. Some,<br />

such as phosphorus, zinc and magnesium, have a<br />

low availability from the diet, so they should be<br />

present in higher concentration and available in<br />

chemical form in any diet designed to promote<br />

convalescence.<br />

As an understanding of these particular nutrients<br />

is integral to the problem of malnutrition, they will<br />

be considered in more detail.<br />

Common response<br />

The response to a deficiency – growth failure with<br />

a mild deficiency and weight loss with more


492<br />

Malnutrition<br />

profound deficiency – is the same for each type II<br />

nutrient. When nutritional growth failure or<br />

weight loss is seen, the particular nutrient that is<br />

lacking cannot be identified.<br />

Every animal experiment in feeding a diet lacking<br />

a type II nutrient results in progressive diminution<br />

or cessation of growth, then weight loss. This<br />

response has been observed consistently and<br />

universally in all species studied, in both acute<br />

and chronic experiments, with each of the type II<br />

nutrients. The response to a habitual mild deficiency<br />

is progressive stunting (with the body in<br />

proportion). The extent of the stunting is a function<br />

of the degree of shortfall of the nutrient and<br />

time. With an acute deficiency, in either the child<br />

or the adult, there is a loss of tissue leading to<br />

wasting. The balance between the severity of the<br />

deficiency and its duration will determine the relative<br />

amounts of stunting and wasting produced.<br />

Mild, chronic deficiencies are more common than<br />

severe, acute deficiencies and stunting is consequently<br />

more common than wasting.<br />

Type II nutrients are not stored in the body<br />

There is a common repertoire of metabolic<br />

changes and ‘reductive adaptations’ that occur in<br />

response to each type II deficiency. However,<br />

because whole tissue is being broken down, or at<br />

least there is no net synthesis, all the nutrients<br />

released from the catabolized tissue and are in the<br />

diet in excess relative to the deficient nutrient<br />

have to be excreted. These nutrients are not stored<br />

in the body. The balance between the various type<br />

II nutrients in the diet is thus very important. In<br />

the face of a deficiency of any one of them there is<br />

a negative balance for them all. For example, if<br />

potassium is omitted from a parenteral feeding<br />

regimen, the patient will loose nitrogen, zinc,<br />

phosphorus and magnesium from the body. He<br />

will not necessarily be in negative balance for the<br />

type 1 nutrients such as calcium, iron or folate<br />

whist he is losing weight.<br />

Dietary deficiency<br />

When a deficient diet is given, the body has mechanisms<br />

to avidly conserve and recycle the deficient<br />

nutrient to maintain itself. For this reason, it is<br />

extremely difficult to produce clinical signs of a<br />

deficiency of these nutrients in the non-growing<br />

animal by dietary means alone. There usually has<br />

to be a pathological loss of the nutrient from the<br />

body. This is commonly by diarrhea. The loss can<br />

be sufficiently severe to compromise intestinal<br />

function so that the deficiency and diarrhea<br />

become persistent. The treatment has to be a<br />

change of diet to replace all type II nutrients that<br />

have been lost. Zinc, phosphorus and magnesium<br />

are frequently not replaced adequately.<br />

Determinant of requirements<br />

The rate of weight gain is the main determinant of<br />

the dietary requirement for a type II nutrient. For<br />

example, children given a diet that supplied just<br />

enough energy for them to maintain their body<br />

weight, without growing, are able to remain in zinc<br />

balance, and maintain their plasma zinc concentration,<br />

with an intake of only 0.08mg Zn/kg per<br />

day. When the same children were gaining weight<br />

rapidly, their plasma zinc fell precipitously despite<br />

a ten-fold increase in the amount of zinc<br />

consumed (1mg Zn/kg per day); this limited lean<br />

tissue synthesis so that the children became obese<br />

without regaining muscle. They required an<br />

amount that far exceeded the recommended<br />

dietary allowance to maintain synthesis of new<br />

lean tissue. During convalescence from illness the<br />

recommended daily allowance (RDA) for type II<br />

nutrients is not sufficient. RDAs are set for normal<br />

individuals. Of course an increased nutrient<br />

density must be supplied for all type II nutrients in<br />

the right balance to make the new tissue.<br />

Children do not need a richer diet than adults<br />

This contradicts what most pediatricians, nutritionists<br />

and lay people believe. A diet that has a<br />

sufficiently low concentration of a type II nutrient<br />

to give clinical signs, other than growth failure,<br />

will affect the elderly first, then adults and lastly<br />

children. This is a consequence of the higher relative<br />

energy requirement of the child. Thus, the<br />

protein requirement of the child, for maintenance<br />

of body weight, is about the same as for an adult<br />

(0.6g/kg per day). However, the energy requirement<br />

for maintenance of the child is about 400kJ/kg per<br />

day whilst that for the adult is about 160kJ/kg per<br />

day. Hence, an adult must have a diet that supplies<br />

6% of the energy as protein, whereas a child


equires only about 2.4% of the energy as protein.<br />

Of course, for the child to grow normally, very<br />

much more protein is required. Interestingly, the<br />

increased requirement for normal growth brings<br />

the protein density up to that of the adult. We have<br />

evolved so that all the family can take the same<br />

diet. If the family has an adequate diet, it can be fed<br />

to the infant and child. It is not necessary to have<br />

special weaning foods. Nutritional requirements,<br />

when expressed as nutrient densities (per unit<br />

energy) are the same for infants, children, adolescents,<br />

adults, and the pregnant and lactating.<br />

In a community, we do not expect to find anything<br />

other than growth failure in children, unless they<br />

have diseases which cause a pathological loss of<br />

the nutrient. If the local diet is sufficiently deficient<br />

to give rise to other clinical signs, these<br />

should be seen first in the elderly and last in children.<br />

However, there is such a wide gap between<br />

the concentration that gives growth failure and<br />

that which gives any other feature of deficiency,<br />

that even where growth failure is common other<br />

clinical signs are rare.<br />

Anorexia<br />

Anorexia is common to a deficiency of each type II<br />

nutrient; this is corrected if the nutrient is<br />

supplied. Thus, on supplementation of the single<br />

missing component a patient will regain his<br />

appetite and then have an increased intake of the<br />

newly balanced diet. Dietary intake data are often<br />

wrongly interpreted as indicating ‘energy deficiency’.<br />

This is rarely the cause of a low dietary<br />

intake. Even in famine situations, the diet is<br />

normally so restricted that type II (and type I) deficiencies<br />

are usually present long before the total<br />

amount of food energy becomes limiting.<br />

Of course, there are other causes of anorexia, in<br />

particular liver disease, infection and psychiatric<br />

abnormality, but type II nutrient deficiency is very<br />

common. This is one reason why most people with<br />

malabsorption do not increase their intake to<br />

compensate for the amounts being lost in the<br />

stools. Refusal to eat a diet that leads to disordered<br />

metabolism is defensive. There is a metabolic preference<br />

for ‘consumption’ of one’s own tissues (a<br />

very good ‘diet’, to satisfy nutritional demands).<br />

This may have benefit in restoring metabolic<br />

balance, but in the long term it leads to severe<br />

malnutrition.<br />

Classification of severe malnutrition 493<br />

The anorexia is related to a balance of the type II<br />

nutrients. It is the relative surfeit of other nutrients,<br />

particularly amino acids, which need to be<br />

metabolized and excreted to prevent the toxicity<br />

that causes the anorexia. If the nutrient density of<br />

all the type II nutrients is low, but they are in<br />

balance, for example by adding lipid to the diet,<br />

the individual may not develop anorexia, but may<br />

become obese from the excess energy in the diet.<br />

They are still under-nourished.<br />

When a supplementary diet is given to children<br />

which does not contain all the nutrients required<br />

for new tissue synthesis, it is possible to unbalance<br />

the diet and make the malnutrition worse, by<br />

diluting the marginal nutrients in the basic diet to<br />

a deficient level.<br />

The type II nutrients have always given problems<br />

to clinicians and nutritionists because of the<br />

difficulty with diagnosis. Unfortunately, the nonspecificity<br />

of weight loss and lack of confirmatory<br />

tests of an inadequate intake have led these nutrients<br />

to be largely ignored by clinicians, and their<br />

nutritional importance to be grossly underestimated.<br />

The phosphorus requirement is set as a<br />

function of calcium not as a critical nutrient in its<br />

own right. Textbooks of nutrition hardly mention<br />

potassium, magnesium, sodium, sulfur or chlorine<br />

deficiency. Yet, as a group, type II nutrient deficiency<br />

is responsible for malnutrition in half the<br />

World’s children and for unrecognized problems of<br />

ill health in many others.<br />

Classification of severe malnutrition<br />

Any system of classification should have practical<br />

use, either to identify those individuals who<br />

require intervention or to examine the prevalence<br />

within a community so that preventive measures<br />

can be taken.<br />

The rest of this chapter is devoted to severe<br />

wasting and edematous malnutrition.<br />

Children<br />

Weight-for-height and height-for-age<br />

During childhood the most sensitive indicator of<br />

malnutrition (type II deficiency) is failure to


494<br />

Malnutrition<br />

achieve normal growth. The systems of classification<br />

compare the size of the child to a ‘normal’<br />

reference; the reference recommended by the<br />

World Health Organization (WHO) is the American<br />

NCHS standard. Other standards have been used;<br />

however, standards derived from elite healthy<br />

populations from around the world are all very<br />

close to NCHS values. The NCHS standards are<br />

derived almost exclusively from artificially fed<br />

infants. They have a higher early weight gain than<br />

exclusively breast-fed children, much of the additional<br />

tissue being adipose. Thus, healthy normal<br />

breast-fed infants ‘fall off’ the present NCHS standards<br />

so that infants may appear to be more<br />

malnourished than they are. New standards are<br />

being prepared by WHO for infants and young<br />

children.<br />

Normal growth is a continuous, balanced accretion<br />

of tissue in a predictable way that results in a<br />

steady, co-ordinated increase in height, weight and<br />

organ size. A mild insult which continues for a<br />

period of time results in slowing or cessation of<br />

growth. The child is normally proportioned but as<br />

time passes he falls further and further behind the<br />

actively growing child. Because of the shape of the<br />

normal growth curve the younger the child the<br />

more rapidly he will fall behind. If a normal child<br />

suddenly slows to half the normal growth rate, he<br />

will be twice his present age before he falls below<br />

70% of normal height for his age and can be diagnosed<br />

as severely stunted.<br />

A child who is exposed to a more severe insult will<br />

not only stop growing in height but will also lose<br />

weight. He becomes underweight for age much<br />

Table 31.3 The classification of severe malnutriton<br />

more quickly and is thin. The weight is low in relation<br />

to his height. Thus, within the group of children<br />

underweight for their age there are two<br />

conceptually different conditions: first, the<br />

stunted but normally proportioned (nutritional<br />

dwarfism); and second, the child of normal height<br />

who is thin and wasted (the original concept of<br />

marasmus). The two conditions can be differentiated<br />

by measuring the height as well as weight and<br />

age. Stunting is measured by the height of the<br />

child in relation to the height of a normal child of<br />

the same age (height-for-age) and wasting as the<br />

weight of the child in relation to a normal child of<br />

his same height, even though the normal child is<br />

usually chronologically younger (weight-forheight).<br />

The grades of stunting and wasting are<br />

shown in Table 31.3.<br />

Two methods of expressing the data are in<br />

common use. The simplest is to express the<br />

weight/height of the child as a percentage of the<br />

normal. The second is to express the deficit in<br />

terms of multiples of the standard deviation of the<br />

normal (NCHS) population. The result is called a<br />

Z-score. The Z-score is much more difficult to<br />

calculate, but it is relatively age independent and<br />

expresses stunting and wasting in the same relative<br />

units. Clearly, any child who has a Z-score<br />

within ± 2 is, by definition, normal. Children who<br />

are more than three standard deviations from the<br />

normal have severe stunting or wasting. In children<br />

over 6 months of age, a deficit of 5% in<br />

height-for-age or 10% in weight-for-height is<br />

approximately equal to one Z-score. In terms of<br />

admission to feeding programs the weight-forheight<br />

percentage is preferred. This is because it<br />

Normal Mild* Moderate* Severe*<br />

Weight-for-height 90–120% † 80–89% 70–79%


more closely relates to prognosis, is easier to<br />

understand and can be extended to the adolescent<br />

(Figure 31.1). 2<br />

The wasted child presents an immediate clinical<br />

problem where rehabilitation can lead to restoration<br />

of lost tissue and function. Correction of<br />

stunting, on the other hand, is more likely to<br />

depend upon public health measures designed to<br />

improve the circumstances of the family. This<br />

system, together with the presence or absence of<br />

edema, anorexia and complicating illness is the<br />

most useful and appropriate for deciding which<br />

individuals require intensive treatment. Because<br />

stunting is much more common than wasting,<br />

systems based upon weight-for-age are no longer<br />

used (for example, these include the Gomez and<br />

Wellcome classifications).<br />

Mid-upper-arm circumference<br />

Between the ages of 1 and 5 years there is very<br />

little change in a normal child’s arm circumference.<br />

This measurement thus gives a simple<br />

anthropometric measure of wasting which is<br />

almost age independent. Because stunting is<br />

common in most countries with a high prevalence<br />

Weight (kg)<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

Risk of death 1%<br />

2%<br />

5%<br />

10%<br />

Classification of severe malnutrition 495<br />

of severe malnutrition a chronological age of more<br />

than 1 year is unreliable, so the cut-off point is<br />

usually 75cm in height.<br />

Adult<br />

Height (cm)<br />

Body mass index<br />

The assessment of malnutrition in adults is<br />

conceptually similar to weight-for-height in children.<br />

Stunting in adults usually represents chronic<br />

undernutrition in childhood. It is irreversible. This<br />

may have implications for the pathogenesis of<br />

adult disease. However, as there is no therapeutic<br />

action that can be taken, adult stunting is mainly<br />

of theoretical interest.<br />

The object with adults is to classify their degree of<br />

‘thinness’. The most useful measure is the body<br />

mass index (BMI), defined as weight (kg) divided<br />

by the square of height (m): wt/(ht) 2 . Exactly the<br />

same index is used to define grades of obesity.<br />

Many sick adults cannot stand to have their height<br />

measured, and in those with kyphosis or scoliosis<br />

the measurement is not useful. In these patients<br />

there are several proxy measures for height that<br />

50 55 60 65 70 75 80 85<br />

Figure 31.1 Weight-for-height curves for 70% of the NCHS (blue) standard and minus 3 Z-score units (red). The other<br />

lines are calculated as ‘risk of death’ cut-off points from the data of Prudhon et al. 2 Note that the 70% line is smooth and<br />

more closely parallel to the risk of death lines. The children admitted by Z-score but excluded by per cent of the median<br />

have a very low mortality (2%), whereas those admitted by per cent of the median and excluded by Z-score are young and<br />

have a high mortality (18%).


496<br />

Malnutrition<br />

can be used. Although arm span is said to be equal<br />

to height, this is only so in those about 1.6m tall.<br />

Clinically, the most useful measurement is demispan.<br />

This is measured as the distance from the<br />

middle of the sternal notch to the tip of the middle<br />

finger in the coronal plane. Both sides should be<br />

measured; if there is a discrepancy the measurements<br />

should be repeated before taking the longest<br />

demi-span. Height (m) in both males and females<br />

can then be calculated from the formula:<br />

Height = 0.73 * (2 x demi-span) + 0.43<br />

Mid-upper-arm circumference<br />

As with children, the mid-upper-arm circumference<br />

can be used to grade the degree of body<br />

wasting in adults. It is particularly used for pregnant<br />

women where the weight is higher than in the<br />

non-pregnant state.<br />

Clinical features<br />

Severe malnutrition can broadly be divided into<br />

three clinical syndromes: marasmus, kwashiorkor<br />

and nutritional dwarfism. In primary malnutrition<br />

each of these conditions is associated with poverty,<br />

deprivation and infection; therefore, they often coexist<br />

in the same individual, just as other conditions<br />

which share etiological features (such as the<br />

various sexually transmitted diseases or helminthiasis)<br />

are frequently found in the same individual.<br />

Therefore, the clinical features often present a<br />

mixed picture.<br />

Marasmus<br />

The patient with classical marasmus has obviously<br />

lost weight with prominent ribs, zygoma and limb<br />

joints, gross loss of muscle mass particularly of the<br />

limb girdles – shoulders and buttocks – and almost<br />

absent subcutaneous fat. The bottom looks like<br />

‘baggy pants’ because the skin, which is thin and<br />

atrophic, lies in redundant folds.<br />

The textbooks describe the face as having a<br />

pinched look like an ‘old man’. This does occur but<br />

is uncommon. It is much more common to have a<br />

relatively normal-looking face (Figure 31.2). This<br />

is important, because one cannot tell whether a<br />

person is severely wasted by just looking at the<br />

face. Many cases are missed in busy out-patient<br />

departments because the children are not stripped<br />

and examined properly, or screened with weightfor-height.<br />

Kwashiorkor<br />

The clinical syndrome of kwashiorkor was first<br />

described by Spanish doctors in the West Indies<br />

and then by the French in Vietnam; it did not enter<br />

the English literature until described by Williams<br />

in 1933. 3<br />

The sine qua non of kwashiorkor is bilateral pitting<br />

edema. Typically, a child of 1 to 2 years with fine<br />

friable discolored hair develops a typical skin rash,<br />

edema and hepatomegaly. Kwashiorkor is an acute<br />

illness which comes on abruptly. The history of<br />

swelling, loss of appetite and mood change is of a<br />

few days only (Figure 31.3).<br />

Nutritional dwarfism<br />

On casual observation the nutritional dwarf<br />

appears perfectly normal. It is only when the age of<br />

the patient is known that the short stature<br />

becomes apparent. However, dental development<br />

is less retarded than height, so that these children’s<br />

facial shape is inappropriate for their size (Figure<br />

31.4).<br />

Edema<br />

The edema is usually dependent and periorbital.<br />

Small accumulations of fluid may be found at post<br />

mortem in the pericardium, pleura and peritoneum,<br />

but large effusions are uncommon; if<br />

serous effusion is present, associated conditions<br />

such as tuberculosis should be sought. Adults with<br />

kwashiorkor are more likely to have serous effusions.<br />

In severe cases the entire body and internal<br />

organs are edematous.<br />

The extent of sodium and water retention in the<br />

extracellular fluid is variable. It is almost always<br />

overestimated by clinicians. In mild edema of the<br />

feet only, the weight loss with resolution of edema<br />

is about 3% of body weight, with moderate edema<br />

5% and with severe edema 10%. There are


(a) (b)<br />

children with up to 30% or more recorded, but<br />

such cases are extremely rare.<br />

Circulation<br />

The retained sodium and water are not evenly<br />

distributed throughout the extracellular compartments.<br />

Depletion of the intravascular volume<br />

usually accompanies the enormous expansion of<br />

the interstitial space. This maldistribution gives<br />

rise to the anomalous statement that an edematous<br />

patient can be ‘dehydrated’. This is semantically<br />

incorrect and we should use the adjective ‘hypovolemic’<br />

to describe these patients. The use of the<br />

word ‘dehydration’ and the attendant concepts<br />

Clinical features 497<br />

Figure 31.2 (a) A severely wasted child from the Democratic Republic of Congo (DRC). This child did not have edema.<br />

His face appears fat, not unlike the facial shape of many children with kwashiorkor. (b) A severely wasted youth from DRC.<br />

Note that he does not have an ‘old man face’ – if he were clothed, you would not realize that he was wasted. Note also<br />

the displacement of his cartilaginous rib forwards. This is a sign of a longstanding low intake of vitamin C or copper.<br />

have led physicians to treat these patients, incorrectly,<br />

with rehydration solutions. In fact, the<br />

hypovolemia in edematous patients is a form of<br />

toxic shock and should not be treated with<br />

sodium-containing fluids.<br />

Hepatomegaly<br />

Hepatomegaly is frequently encountered in the<br />

West Indies. It is much less common in Africa or<br />

Asia, although it does occur. The liver may extend<br />

to the iliac brim. It is smooth, firm and not usually<br />

tender. The liver enlargement is due to fat accumulation,<br />

mainly as triglyceride. Up to half the wet<br />

weight of the liver can be fat, but it is usually


498<br />

Malnutrition<br />

Figure 31.3 A child with extensive skin lesions and<br />

edema from Uganda. Note that this child is receiving breast<br />

milk. The mother appears anthropometrically normal.<br />

much less than this. Signs of liver dysfunction such<br />

as petechiae or very slight hyperbilirubinemia are<br />

serious prognostic signs.<br />

Splenomegaly<br />

An enlarged spleen is very unusual in uncomplicated<br />

malnutrition. Where it occurs it is likely to<br />

be associated with particular infections such as<br />

malaria, kala azar or HIV, or a hemoglobinopathy.<br />

Anorexia<br />

Loss of appetite is a common feature of all forms of<br />

severe malnutrition. The most likely underlying<br />

causes are type II nutrient deficiency, infection<br />

and liver dysfunction. These patients nearly<br />

always have all three of these underlying causes<br />

compounding each other.<br />

Figure 31.4 These three Jamaican children are the same<br />

age. The one on the left is stunted. The one on the right is<br />

wasted and the one in the middle is normal. The wasted<br />

and stunted children have the same weight. The normal<br />

child is much heavier.<br />

Mood and behavior<br />

Classically the children are apathetic when left<br />

alone and complain when they are picked up. The<br />

passsivity can be profound, so that the children lie<br />

in one position for prolonged periods. These children<br />

can develop bed sores. Quite frequently catatonia<br />

can be demonstrated. If the limbs are raised<br />

then moved, the child will maintain each posture<br />

for prolonged periods. This passivity and the lack<br />

of crying is extremely serious. A good mother will<br />

find out why a child cries – hunger, thirst, cold,<br />

pain, unhappiness or being uncomfortable – and<br />

then put it right. If the child does not cry the<br />

mother thinks that none of these problems are<br />

affecting her child. The child is neglected. When a<br />

malnourished child is in the hospital the staff also<br />

neglect these children for the same reasons. This is<br />

why they should always be nursed together in a<br />

special unit.<br />

A series of stereotyped, self-stimulating repetitive<br />

movements may occur; this is typical of psychosocial<br />

deprivation. One of the most damaging forms<br />

of self-stimulation is rumination. When alone, the<br />

child regurgitates the last meal, to re-taste it, then<br />

re-swallows it. Inevitably some is lost. In less<br />

severely deprived children intentional regurgitation<br />

may be the only way in which the child can<br />

get attention from a busy mother; it then becomes<br />

a learned habit that leads to malnutrition.<br />

Rumination and attention-seeking regurgitation<br />

are frequently confused with vomiting from a


physical cause, so that many of these children<br />

undergo extensive gastrointestinal investigations.<br />

Skin<br />

In kwashiorkor there are skin changes that appear<br />

and progress like sunburn. The skin changes are<br />

acute, occurring over the course of a day or so. The<br />

skin first becomes darker in color, particularly over<br />

areas exposed to minor trauma or pressure. The<br />

superficial dermal layers then dry like thin parchment<br />

and split wherever they are stretched to<br />

reveal pale areas between the cracks (Figure 31.5).<br />

The dry cracked layer then peels off to leave<br />

hypopigmented extremely thin skin. If it is gently<br />

pinched between the fingers numerous small<br />

wrinkles appear, showing how thin and atrophic<br />

the epidermis is. The skin is very friable and ulcerates<br />

or macerates easily, particularly in the flexures,<br />

perineum and behind the ears. In severe<br />

cases it may appear as if the child has been burnt.<br />

Hair<br />

There is atrophy of the hair roots of the scalp. They<br />

cease to synthesize protein, the bulb shrinks and<br />

the hair may be plucked out easily and painlessly.<br />

The patients may go completely bald. The reduction<br />

in synthesis is also seen in chemotherapy.<br />

When this sign is present there are also low levels<br />

of hepatic export proteins, such as albumin and<br />

transferrin showing that the slowing of synthesis is<br />

general throughout the body.<br />

The hair itself becomes thin and straight and ‘lifeless’.<br />

In patients with naturally curly hair the curls<br />

may be lifted up by the new straight hair to give<br />

the appearance of trees with straight trunks and a<br />

canopy – the ‘forest sign’.<br />

The hair may change color to red, brown, gray or<br />

blond. The basis for this change in color is<br />

unknown. Hair color changes bear no relation to<br />

prognosis and should not be used to classify these<br />

children.<br />

Feminization<br />

Unlike the hair on the scalp, the eyelashes grow<br />

long and luxuriant. There may also be excessive<br />

Clinical features 499<br />

Figure 31.5 The stripping skin from this foot shows the<br />

different stages of the skin lesions of kwashiorkor.<br />

growth of lanugo hair. The face of little boys starts<br />

to look like girls. Breast enlargement is not uncommon;<br />

it occurs particularly in patients with gross<br />

hepatomegaly. The reason is not only the diminution<br />

of conjugation and excretion of steroid<br />

hormones by the liver. The small bowel bacterial<br />

overgrowth, that is almost universal in these children,<br />

deconjugate the conjugated steroids so that<br />

they are reabsorbed. These signs of feminization<br />

indicate longstanding chronic malnutrition with<br />

small-bowel overgrowth and fatty liver.<br />

Cheeks<br />

Fullness of the cheeks is commonly associated<br />

with edematous malnutrition (so-called jowls).<br />

The cause is unknown; it is not due to parotid<br />

enlargement.<br />

Although there is usually marked parotid atrophy,<br />

in some patients, particularly malnourished<br />

adults, there is painless parotid enlargement. This<br />

seems to be associated with particular geographical<br />

locations. For example, it is common in one of<br />

the Comoro islands off the African coast but not on<br />

a neighboring island. The reason is unknown.<br />

Bone<br />

There is nearly always an enlargement of the<br />

costochondral junctions giving a ‘rickety rosary’.<br />

The cause is not usually related to vitamin D


500<br />

Malnutrition<br />

deficiency. It is related to the very low available<br />

phosphorus and calcium in the diet. Harrison’s<br />

sulci and other chest deformities are not only due<br />

to the pliability of the bone, but also to repeated<br />

chest infections. Smoke pollution from cooking<br />

fires is very common in the developing world –<br />

the reason why chest infections are more frequent<br />

in the wet seasons is that the cooking is then done<br />

indoors. Chronic vitamin C (or copper) deficiency<br />

gives rise to dislocation of the costochondral junctions<br />

with the cartilaginous chest protruding<br />

forwards. This is the so-called ‘scorbutic rosary’.<br />

It is also common. Signs of acute scurvy are<br />

extremely uncommon. The sclera may be blue<br />

because the choroid pigment shows through,<br />

similar to that seen in congenital defects of collagen<br />

formation or structure. There is nearly always<br />

very marked osteopenia on X-ray. Despite the<br />

demineralization and collagen defects, and unlike<br />

osteitis fragilis, these children very rarely have<br />

fractures. The mothers must handle the children<br />

very gently indeed.<br />

Abdominal swelling<br />

The abdomen is usually protuberant. This is due<br />

to gas in the intestine rather than the enlarged<br />

liver. The gut gas is due to the very slow lowamplitude<br />

peristaltic waves. This is the main<br />

cause of the bacterial overgrowth. Frequently the<br />

abdominal wall is sufficiently thin for peristalsis<br />

to be easily visible. Bowel sounds are infrequent.<br />

In newly admitted patients there may be a succussion<br />

splash from the stomach or bowel.<br />

Inflammation<br />

The tympanic membranes are white and thickened<br />

(tympanosclerosis), usually without signs of<br />

inflammation. The tonsils are atrophic. It is<br />

unusual to find lymphadenopathy. When it occurs<br />

symmetrically it is usually associated with HIV<br />

disease. When asymmetrical it is usually tuberculosis.<br />

There is frequently oral, esophageal, gastric,<br />

colonic and perineal candidiasis.<br />

Pus does not form, even on open skin lesions.<br />

There is almost no inflammatory response. This is<br />

why fever is uncommon; when it occurs, it is<br />

usually due to a high environmental temperature.<br />

Similarly, lobar pneumonia does not usually<br />

occur in the absence of inhalation.<br />

Anemia<br />

Anemia is almost universal to some degree. It is<br />

due to multi-micronutrient deficiency and infection.<br />

Thus, there is frequent evidence of deficiencies<br />

of folic acid, copper, vitamin E, pyridoxine,<br />

riboflavin and vitamin C, all of which can cause<br />

anemia. Schistocytosis is not uncommon because<br />

of the fragility of the red cells. Malaria is also a<br />

common cause of anemia. In urban areas the<br />

blood content of lead is often high. The iron stores<br />

are normally replete and iron deficiency is not<br />

usually the cause of anemia in severe malnutrition.<br />

Other signs<br />

Angular stomatitis, lingual atrophy, follicular<br />

hyperkeratosis, vitamin A-deficient eye signs and<br />

other signs of specific type 1 nutrient deficiencies<br />

frequently occur.<br />

Pathophysiology<br />

The sequence of events that occur in any<br />

malnourished individual is shown in Figure 31.6.<br />

Although the defects reinforce each other in a<br />

cyclical way, the best starting point is the reduction<br />

in dietary intake. This can be due to psychiatric<br />

illness, the anorexia associated with liver<br />

disease, infection, neoplasia, drug intoxication or<br />

a type II nutrient deficiency, to famine or starvation,<br />

upper intestinal disease, malabsorption or<br />

other losses of nutrients from the body.<br />

Reduced mass<br />

A reduction in body mass is the most obvious<br />

abnormality clinically, and forms the basis for the<br />

various anthropometric classifications of malnutrition.<br />

As weight is lost, the absolute nutritional<br />

requirements are reduced simply on the basis of<br />

the decreased mass. There is also a relative reduction<br />

in requirement so that each gram of<br />

body tissue requires less energy. This reduces


equirement by about one-third and is achieved<br />

over weeks or months by metabolic adaptation.<br />

Efficient use and reduced work<br />

Type II nutrient deficiency Psychological<br />

Anorexia<br />

Infection<br />

Neoplasm<br />

Starvation<br />

Pathological loss<br />

Reduced mass<br />

Body composition<br />

changed<br />

Organs, tissue and chemical<br />

Small-bowel overgrowth<br />

Infection<br />

Figure 31.6 Schematic representation of the sequence of events in marasmus.<br />

The reduction in requirement comes about in two<br />

ways. First, nutrients are used more efficiently.<br />

However, we normally use our food quite efficiently,<br />

so that there is a relatively small absolute<br />

saving possible from increased efficiency. By far<br />

the most important adaptation is in the actual<br />

work performed by the whole body, its organs,<br />

tissues, cells, organelles and enzymatic machinery.<br />

In the well-nourished individual the metabolic<br />

capacity far exceeds the demand. Energy is used to<br />

maintain this excess capacity for metabolic work.<br />

The excess allows us to cope with rapid increases<br />

in demand for activity or imposed stress. Thus, we<br />

Reduced intake<br />

Efficient use<br />

Loss of reserve<br />

Tissue and functional<br />

capacity<br />

Loss of homeostasis<br />

Death<br />

Malabsorption<br />

Neglect<br />

Reduced requirement<br />

Reduced work<br />

Physiological and<br />

metabolic responses<br />

changed<br />

Specific deficiency<br />

Pathological losses<br />

Skin, intestine, kidney<br />

Pathophysiology 501<br />

can run at 30km/h achieving a cardiac output of<br />

20l/min, eat 70g protein and 10g sodium at one<br />

sitting, then fast for several days without untoward<br />

effects. In health, we maintain all the digestive,<br />

absorptive, hepatic and renal capacity to deal with<br />

feast or famine. This ‘physiological redundancy’ is<br />

why ‘unphysiological’ stress tests need to be used<br />

to diagnose disordered function at an early stage.<br />

These ‘reserves’ of tissue and functional capacity<br />

are nutritionally expensive to synthesize, replace<br />

and maintain; they are sacrificed in malnutrition<br />

(Table 31.4).<br />

Physiological and metabolic changes<br />

The reduction in work of the cells of the body<br />

leads to major changes in the physiological and<br />

metabolic responses of the body. Indeed, no physiological<br />

function has so far been studied in severe


502<br />

Malnutrition<br />

Table 31.4 Some changes in physiological function in malnourished children<br />

undernutrition and found to be ‘normal’. Some of<br />

the changes are listed in Table 31.5.<br />

At the level of the whole body, spontaneous activity<br />

is severely curtailed – apathy and passivity.<br />

Children no longer play or explore, and adults sit<br />

in a state of suspended animation, only moving<br />

when absolutely necessary.<br />

At the other end of the organizational spectrum is<br />

a fundamental adaptation: slowing of the sodium<br />

pump. Normally about one-third of basal energy<br />

requirements is consumed, pumping sodium out<br />

of cells and potassium back into the cells. This<br />

adaptation is at the cost of allowing the intracellular<br />

sodium concentration to rise and potassium to<br />

fall. The potassium lost from the cell cannot be<br />

accommodated in the extracellular fluid and is<br />

excreted. Slowing of the sodium pump also leads<br />

to a reduction in cellular electrical potential and<br />

delay in its restoration, hence there is a reduction<br />

in neuromuscular function and muscle rapidly<br />

fatigues. Other processes that depend upon a<br />

sodium gradient, such as amino acid and glucose<br />

transport, have a reduced capacity in malnutrition.<br />

Malnourished Recovered Difference (%recovered)<br />

Metabolic rate (kJ/kg 0.75 per day) 315 417 -24<br />

Sodium pump activity (/h) 3.62 4.94 -27<br />

Intracellular sodium (mM/kg DS) 169 109 +55<br />

Intracellular potassium (mM/kg DS) 341 387 -12<br />

Protein synthesis (g/kg per day) 4.0 6.3 -37<br />

Protein breakdown (g/kg per day) 3.7 6.4 -42<br />

Cardiac output (l/min per m 2 ) 4.77 6.90 -31<br />

Stroke volume (ml/beat per m 2 ) 44.1 53.0 -22<br />

Circulation time (s) 13.7 10.5 +30<br />

GFR (Cin – ml/min per m 2 ) 47.1 92.4 -41<br />

Renal blood flow (Cpah – ml/min per m 3 ) 249 321 -22<br />

H + excretion after NH4Cl (µEq/min) 10.4 28.4 -63<br />

Osmolal clearance rate (ml/min) 0.20 0.66 -70<br />

% Infused sodium excreted 22.3 48.7 -54<br />

Sodium excreted (% of sodium filtered)<br />

Normal ECF 0.50 1.23 -59<br />

Expanded ECF 0.82 11.07 -93<br />

Response to temperature change poikilotherm homeotherm —<br />

DS, dry stool; GFR, glomerular filtration rate; Cin, inulin clearance; Cpah, p-aminohippurate clearance;<br />

ECF, extracellular fluid<br />

A further one-third of basal energy requirement is<br />

used for the continual cycle of protein synthesis<br />

and breakdown in a process known as protein<br />

turnover. This is reduced by about 40%.<br />

There is a reduction in cardiac output due to both<br />

a lowered heart rate and a lowered stroke volume.<br />

The ventricular function curves (stroke work/pressure)<br />

are altered so that the point of maximum<br />

performance occurs at a lower mean pressure;<br />

these patients are very easily precipitated into<br />

heart failure.<br />

The maximum concentrating ability of the kidney<br />

is severely restricted. This means that one has to<br />

be very careful not to give a diet with a high renal<br />

solute load, and to ensure that sufficient water is<br />

taken. There is a very limited capacity to excrete<br />

free hydrogen ions, titratable acid and ammonia in<br />

response to an acid load. A common acid load<br />

comes from giving magnesium chloride as a<br />

dietary supplement. However, the most important<br />

renal abnormality is a particular and severe limitation<br />

of the ability to excrete sodium, particularly in<br />

response to an expanded extracellular fluid


volume. These abnormalities are shown in Figure<br />

31.7 and 31.8. With an expanded circulation it<br />

takes 10h for a malnourished child to excrete the<br />

sodium that a normal child can excrete in 20min.<br />

The motility of the whole intestine is reduced and<br />

small-intestinal transit time increased. This leads<br />

directly to small-bowel bacterial overgrowth and a<br />

distended abdomen. However, it also affects the<br />

pharyngeus and esophagus. These children very<br />

readily inhale food because of these neuromuscular<br />

abnormalities. They should never be force-fed<br />

or held recumbent whilst they are fed. Inhalation<br />

of the liquid diet, insufficient to obstruct the main<br />

bronchi, is probably the most common reason for<br />

pneumonia in the malnourished child. It is<br />

preventable.<br />

There is a reduction in gastric acid, bile and<br />

pancreatic enzyme production. The cellular<br />

enzymes and transport systems for nutrient<br />

absorption are compromised and the mucosa<br />

becomes flattened, but rarely to the degree found<br />

in primary intestinal disease such as gluten<br />

enteropathy; mitotic figures in the crypts become<br />

rare. That the defect is one of capacity and not a<br />

specific abnormality is demonstrated by absorption<br />

at low rates of presentation (perfusion) being<br />

Sodium excretion<br />

(% of sodium filtered)<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

Normal ECF<br />

Expanded ECF<br />

Figure 31.7 The fractional excretion rate of sodium<br />

(expressed as a per cent of filtered sodium) of<br />

malnourished (red) and normal patients (blue) who have a<br />

normal circulating volume and after expansion with a<br />

saline infusion. ECF, extracellular fluid.<br />

Pathophysiology 503<br />

relatively normal, whereas at high rates the digestive<br />

and absorptive capacity is overwhelmed.<br />

The malnourished patient becomes poikilothermic.<br />

Even a modest reduction to 21°C or elevation<br />

to 33°C in environmental temperature may lead to<br />

hypothermia or pyrexia, respectively. It is not<br />

unusual for malnourished children in the tropics<br />

to develop hypothermia; in temperate climates<br />

hypothermia is common in the elderly. They are<br />

qualitatively different from normal. Normal individuals<br />

increase their oxygen consumption in<br />

response to a cool environment to maintain body<br />

heat; malnourished patients reduce their oxygen<br />

consumption in response to a cool environment<br />

(Figure 31.9). They never shiver. Once they<br />

become cold the slowing metabolism means that<br />

they will not recover without some form of external<br />

heat.<br />

There are marked changes in the hormonal<br />

balance in malnutrition. Growth hormone levels<br />

are elevated with a low insulin concentration and<br />

a reduced insulin response to a test meal. Insulinlike<br />

growth factor (IGF)-I and II, catecholamine,<br />

glucagons, thyroxine and tri-iodothyronine, both<br />

free and bound, are all low; cortisol tends to be<br />

high. The response to injected hormones is diminished,<br />

with down-regulation of most receptors.<br />

Although there is glucose intolerance, glucose<br />

levels are generally lower than normal. The intolerance<br />

is partly due to liver dysfunction (thought<br />

to be due to slow initial phosphorylation of the<br />

monosaccharide) as the degree of galactose intolerance<br />

is similar to the degree of glucose intolerance.<br />

There is a very marked reduction in gluconeogenesis.<br />

The febrile, acute phase and inflammatory<br />

responses, and the immune system also partake in<br />

the reductive adaptation; they are either absent or<br />

severely blunted in seriously malnourished patients.<br />

Infection is normally recognized by the<br />

body’s response in terms of a fever, leukocytosis,<br />

pus formation, tachypnea, etc. When these<br />

responses do not occur, life-threatening infection<br />

may go completely unrecognized.<br />

Body composition<br />

There are changes in body composition. Most<br />

tissues contribute to the loss of weight, but, they


504<br />

Malnutrition<br />

Table 31.5 The main physiological changes in severe malnutrition and their implications for management<br />

Physiological change<br />

Cardiovascular system<br />

The heart is smaller and thinner than normal. The<br />

cardiac output and stroke volume are reduced. Saline<br />

infusion leads to greatly increased venous pressure.<br />

Overload of the heart readily leads to heart failure.<br />

Blood pressure is low. Renal perfusion is reduced. The<br />

circulation time is reduced. Plasma volume is usually<br />

normal and red cell volume reduced.<br />

Genitourinary system<br />

Glomerular filtration is reduced. The excretion of acid or<br />

of an osmolar load is greatly reduced. Urinary phosphate<br />

output is low. Sodium excretion is lower than<br />

normal. The kidney is physiologically unresponsive. An<br />

expanded intravascular or extracellular volume does not<br />

lead to increased sodium excretion. Urinary tract infection<br />

is common.<br />

Gastrointestinal system<br />

The stomach produces much less acid than normal. The<br />

motility of the whole intestine is reduced. The pancreas<br />

is atrophied and produces a reduced amount of digestive<br />

enzymes. The small-intestinal mucosa is atrophic with<br />

reduced levels of digestive enzymes. Absorption is<br />

reduced when a lot of substrate is given either from a<br />

high concentration or from large amounts of more dilute<br />

solutions.<br />

Liver<br />

There is a reduction in the synthesis of all hepatic export<br />

proteins. Abnormal metabolites of amino acids are<br />

produced. The ability of the liver to take up, metabolize<br />

and excrete toxins is severely limited. The energy<br />

production from substrates (such as galactose, fructose)<br />

is much slower than normal. The capacity for gluconeogensis<br />

is limited, leading to hypoglycemia with stress<br />

of infection. Biliary secretion is reduced. Limited output<br />

of bile salts. Jaundice is uncommon. Liver function tests<br />

unreliable, because intracellular enzyme levels very<br />

low. Indocyanine green uptake very abnormal.<br />

Diagnostic and therapeutic implication<br />

The children are vulnerable to both an increase and<br />

decrease in blood volume. Any decrease will further<br />

compromise tissue perfusion; an increase can easily<br />

produce acute heart failure. If dehydrated give restricted<br />

amounts of ReSoMal: do not give intravenous fluid<br />

unless in severe shock. Restrict blood transfusion to<br />

10ml/kg per day and cover with diuretic. Restrict the<br />

sodium intake from the diet and drugs. Give additional<br />

potassium, magnesium and phosphorus.<br />

Treat with diets that have a low renal solute load.<br />

Ensure that there is sufficient water in the diet. Do not<br />

overconcentrate the diet. Prevent further tissue breakdown<br />

(treat infection and give adequate energy –<br />

80–100kcal/kg per day) and do not give excess protein<br />

over and above that needed to restore tissue, to limit<br />

urea production. Protein should have balanced amino<br />

acids (high quality). Avoid salts that can give an acid<br />

load (e.g. magnesium chloride, high protein). There has<br />

to be adequate phosphorus in the diet to excrete acid<br />

equivalents (this is supplied in adequate amounts by<br />

cow’s milk, but is inadequate with soy substitutes).<br />

Only use diets in phase 1 that are hypo- or isotonic.<br />

Feed small amounts of diet often, to remain within<br />

absorptive capacity. If food is malabsorbed the first step<br />

is to increase the frequency and reduce the size of each<br />

feed (do not dilute the diet and give the same volume).<br />

The food is necessary to stimulate the intestine to<br />

regrow. Use antibiotics active against small-bowel<br />

bacterial overgrowth. Occasionally addition of pancreatic<br />

enzymes is useful. Persistent diarrhea nearly always<br />

responds to the diet alone.<br />

The child should not be given a large meal to metabolize<br />

at one time. The amount of protein should be<br />

within the capacity of the liver to metabolize it, but<br />

sufficient to stimulate synthesis of export proteins. This<br />

is much lower than the RDA. Prevention of catabolism<br />

is important. Drugs which depend upon hepatic<br />

disposal or are hepatotoxic should be given in reduced<br />

doses or not at all. Adequate carbohydrate should be<br />

given to prevent the necessity for gluconeogensis.<br />

continued


Table 31.5 Continued<br />

Immune system<br />

All aspects of immunity are diminished. Lymph glands,<br />

tonsils and the thymus are atrophic. Cell-mediated (T<br />

cell) immunity is particularly depressed. There is very<br />

little IgA in secretions. Complement components are<br />

low. Phagocytes do not kill ingested bacteria efficiently.<br />

Inflammatory response<br />

Tissue damage is not associated with inflammation;<br />

white cells do not migrate into areas of damage. The<br />

acute phase response is diminished.<br />

Endocrine system<br />

Insulin is reduced and there is glucose intolerance. IGF-<br />

I is very low, although growth hormone is high. Cortisol<br />

is usually high.<br />

Temperature regulation<br />

The children are poikilothermic. Both heat generation in<br />

the cold and sweating in the heat are impaired. The children<br />

become hypothermic in a cold environment and<br />

pyrexial in a hot environment.<br />

Cellular function<br />

The activity of the sodium pump is reduced (marasmus).<br />

The cell membranes are more leaky than normal (kwashiorkor).<br />

This leads to an increase in intracellular sodium<br />

and a decrease in intracellular potassium and magnesium.<br />

Protein synthesis is reduced.<br />

Metabolic rate<br />

The basal metabolic rate is reduced by about 30%. The energy<br />

expenditure due to activity of these children is very low.<br />

Body composition<br />

There is particular atrophy of skin, subcutaneous fat and<br />

muscle. Fat is lost from the orbit. There is atrophy of<br />

many glands including the sweat, lachrymal and salivary<br />

glands.<br />

ReSoMal; IGF, insulin-like growth receptor<br />

Pathophysiology 505<br />

Assume all malnourished children have infections and<br />

overgrowth of mucosal surfaces. Blind antibiotic treatment<br />

should be given on admission to all children.<br />

Treatment will have to continue until the improved<br />

nutritional state leads to improvement of the immune<br />

system. Physically separate acute admissions from<br />

recovering children. Give measles vaccine and vitamin<br />

A. Treat with an antifungal agent.<br />

Signs of infection are often absent even after careful<br />

examination. Localized infection such as lobar pneumonia<br />

is uncommon, generalized infection (bronchopneumonia)<br />

is common and may be present with no<br />

radiographic or other signs. Assessments of raised white<br />

cell count and fever are not necessary. Otitis usually<br />

does not give an inflamed or bulging ear. Urinary tract<br />

infection is normally symptomless. Hypoglycemia and<br />

hypothermia are both signs of severe infection.<br />

The endocrine system may not be able to respond<br />

appropriately to large meals. Give small frequent meals.<br />

Do not give steroids; they are already high.<br />

Maintain environment between 25 and 30ºC. Put a<br />

max–min thermometer in the ward. Cover the children<br />

with clothes and blankets. Children to sleep with their<br />

mothers. Keep windows closed at night. Dry children<br />

quickly and well after washing and clothe them. Cool<br />

fevered children with tepid (not cold) water.<br />

All the children need large doses of potassium and<br />

magnesium. Sodium intake should be restricted. During<br />

recovery the sodium has to come out of the cells and<br />

potassium has to go in; this easily leads to cardiac overload<br />

and hypokalemia. Reversal of the electrolyte<br />

abnormality should be gradual and should occur after<br />

the kidney has recovered. If used at all digoxin doses<br />

should be halved.<br />

Internal heat production is limited. Most metabolic<br />

processes are sluggish.<br />

Most signs of dehydration are unreliable: eyes may be<br />

sunken with loss of orbital fat. Atrophy leads to folds of<br />

skin. Skin, mouth and eyes are dry owing to gland<br />

atrophy. The children have limited reserves of energy.<br />

The respiratory muscles are easily exhausted.


506<br />

Malnutrition<br />

(a)<br />

Intracellular sodium (mmol/l)<br />

140<br />

120<br />

100<br />

80<br />

60<br />

40<br />

20<br />

0<br />

Malnourished<br />

Recovered<br />

Figure 31.8 The intracellular sodium (a) and potassium (b) before and after recovery from marasmus.<br />

(a)<br />

Core temperature (°C)<br />

37.5<br />

37.0<br />

36.5<br />

36.0<br />

35.5<br />

35.0<br />

-30 0 30 60 90 120 150<br />

Time (min)<br />

(b)<br />

Intracellular potassium (mmol/l)<br />

180<br />

160<br />

140<br />

120<br />

100<br />

Malnourished<br />

Recovered<br />

Figure 31.9 The body temperature response of exposure of malnourished (red) and recovered children (blue) to a low<br />

environmental temperature (a). The oxygen consumption (b) corresponding to (a). Note that, as the body temperature falls<br />

in the normal child, there is an increase in oxygen consumption, whereas in the malnourished child the oxygen consumption<br />

falls.<br />

(b)<br />

Metabolic rate (kcal/kg per 30 min)<br />

1.8<br />

1.5<br />

1.2<br />

0.9<br />

-30 0 30 60 90 120<br />

Time (min)


do not contribute equally. Subcutaneous fat may<br />

virtually disappear and muscle mass is often<br />

reduced by more than half. Skin and intestine are<br />

also disproportionately affected, whereas the<br />

viscera and the central nervous system are relatively<br />

well preserved.<br />

The chemical composition of the whole body is<br />

altered because of the changes in the relative size<br />

of the organs. Those that are most atrophic are<br />

those that are used least. Thus, although the<br />

gluteus muscle may shrivel, the external ocular<br />

muscles are almost normal. The viscera are<br />

preserved at the expense of muscle, fat and skin.<br />

However, where loss would be critical to survival,<br />

it is much less marked. For example, malnourished<br />

children in Tchad, in the Sahara desert, have good<br />

skin. In this environment if the skin lost its function<br />

the children would rapidly desiccate.<br />

However, there are also changes as a consequence<br />

of the reductive adaptations themselves. Critically,<br />

the reduction in activity of the sodium pump<br />

invariably leads to an increased total body sodium<br />

and reduced total body potassium, irrespective of<br />

the patient’s state of hydration or the serum electrolyte<br />

concentrations. When there is also edema,<br />

the malnourished patient also has an increased<br />

extracellular as well as intracellular sodium<br />

concentration. The reduction in the enzyme,<br />

soluble protein and RNA complement, with relative<br />

increase in structural protein, is accompanied<br />

by reduction in those trace elements and vitamins<br />

used as enzymatic co-factors. They are not<br />

retained in the tissues without the parent proteins.<br />

The proportionate reduction in each of these<br />

components of soft tissue is about the same. Of<br />

course, during reversal of the adaptation the<br />

deficits of all these components has to be made<br />

good before any weight gain could occur. Iron is an<br />

exception. There is an increased concentration of<br />

tissue iron in most forms of severe malnutrition<br />

(but not in patients with chronic blood loss from,<br />

say, intestinal helminthiasis).<br />

Loss of reserve<br />

The cost of both the reductive adaptations and the<br />

reduction in functional tissue mass is to dispense,<br />

to a greater or lesser extent, with the reserve capacity.<br />

The malnourished individual has a reduced<br />

Pathophysiology 507<br />

capacity to respond appropriately to the metabolic,<br />

environmental or infective stresses that do little<br />

harm to a well-nourished individual.<br />

Vicious cycles<br />

Superimposed upon the reduced ability to respond<br />

to metabolic and environmental changes are the<br />

pathological effects of the stresses themselves. The<br />

curtailment of the inflammatory and immune<br />

responses make infections ubiquitous. The atonic<br />

gut, achlorhydria, and poor secretion of IgA and<br />

bile salts combine to allow normal intestinal flora.<br />

Both bacteria and fungi overgrow the small intestine<br />

and stomach. These organisms directly<br />

damage the intestine, deconjugate bile salts and<br />

exacerbate any malabsorption. Unless they are<br />

suppressed, diarrhea may worsen when additional<br />

food is given. The gas produced and the intestinal<br />

stasis is the reason for abdominal swelling in<br />

malnutrition; such swelling is characteristic of<br />

small-bowel overgrowth.<br />

The diarrhea and repeated infections give rise, in<br />

turn, to specific nutrient deficiencies, particularly<br />

of mineral elements. When the integrity of the skin<br />

is breached from the burn-like lesions of severe<br />

childhood malnutrition, bed sores, fistulae and<br />

traumatic or surgical lesions which become indolent,<br />

blood, serum and heat can be lost in considerable<br />

amounts.<br />

The effect of the infections, small-bowel overgrowth,<br />

malabsorption, nutrient losses and<br />

thermal stress in the patient already adapted to<br />

malnutrition, is to exacerbate the anorexia and<br />

further reduce the intake. The cycle is now<br />

complete. The adaptations are reinforced and<br />

physiological function increasingly compromised.<br />

The debilitated patient reaches a self-perpetuating<br />

stage where increasing anorexia and organ<br />

dysfunction lead to rapid deterioration and death.<br />

Loss of homeostasis<br />

As the physiological and tissue reserve is whittled<br />

away and the effects of chronic infection and diarrhea<br />

deplete the patient, he becomes more and<br />

more ‘brittle’, like a diabetic patient losing homeostatic<br />

control of his blood sugar. However, unlike


508<br />

Malnutrition<br />

the diabetic, in the severely malnourished patient<br />

it is not just one organ or system that is functionally<br />

deranged but all of them. Finally, the patient<br />

simply cannot control his ‘milieu interieur’, everything<br />

slows down and stops.<br />

When treatment regimens are planned they must<br />

always work within the patient’s limited metabolic<br />

capacity whilst the reductive adaptations are<br />

reversed dietetically. Disequilibrium syndromes<br />

can occur during this reversal and lead to sudden<br />

unexpected death during early refeeding, even in<br />

previously obese patients who have been undergoing<br />

a prolonged period of energy restriction.<br />

Although the classification of marasmus is based<br />

entirely upon anthropometric criteria, sickness<br />

and death are related to the disordered physiology<br />

rather than the precise degree of wasting. There is<br />

not a close correspondence between the two.<br />

There are usually accompanying type I nutrient<br />

deficiencies and pathological losses. The difficulty<br />

of relying solely on anthropometry is illustrated by<br />

the patient with anorexia nervosa. She normally<br />

has had a very restricted amount of a high-quality<br />

diet and is abnormally active. Even though the<br />

anthropometry is similar, many of the physiological<br />

changes are different, and these patients can<br />

tolerate a much lower body mass before they<br />

become dysfunctional. Bulemic patients are more<br />

like those with severe malnutrition.<br />

Edematous malnutrition<br />

In community surveys about 80% of patients with<br />

edematous malnutrition are not wasted, and most<br />

are not stunted. Ecologically the condition is ‘overdispersed’<br />

following a negative binomial distribution,<br />

whereas marasmus follows a normal Poisson<br />

distribution. That means that kwashiorkor occurs<br />

in geographical pockets; one village may have no<br />

cases whereas another in the same region has more<br />

cases than would be expected. Marasmus and<br />

kwashiorkor do not share a distribution, even<br />

when they are prevalent in the same area. Some<br />

villages have predominantly marasmus and others<br />

kwashiorkor.<br />

Contrary to what is generally believed, about 25%<br />

of children with kwashiorkor are being breast fed,<br />

some exclusively. The type II nutrient concentra-<br />

tion in breast milk is defended by the mother. In<br />

contrast, the concentration of type I nutrients in<br />

breast milk varies with the mother’s intake and<br />

status. Samples of the breast milk, from mothers<br />

feeding children with kwashiorkor, are very low in<br />

antioxidants. These are all type I nutrients.<br />

Edematous malnutrition, unlike other forms of<br />

malnutrition, is an acute illness. A patient who<br />

appears quite well may, over the course of a few<br />

days, become extremely irritable, progress through<br />

the various stages of the skin lesions and suddenly<br />

‘blow up’ with edema. When they present they<br />

have almost complete retention of ingested<br />

sodium. Although they have a low blood pressure<br />

and signs of hypovolemia the sodium retention<br />

appears to be renal in origin, as it often does not<br />

recover with restoration of blood pressure and<br />

even over-expansion of the vascular volume.<br />

Although the level of aldosterone is high, it is<br />

unlikely to be the cause of the sodium retention, as<br />

it paradoxically increases when these patients<br />

commence diuresis to lose edema. Natriuretic<br />

peptides and their receptors have not been<br />

measured in kwashiorkor.<br />

Kwashiorkor belongs to the family of diseases that<br />

includes toxic shock syndrome, adult respiratory<br />

distress syndrome, multiple organ failure and sickcell<br />

syndrome. It also has similarities to patients<br />

with radiation injury, cytotoxic drug overdose and<br />

HIV infection in that they all lead to profound<br />

immuno-incompetence and abnormal glutathione<br />

metabolism.<br />

In edematous malnutrition, in contrast to marasmus,<br />

there is an increased excretion of urinary<br />

nitrate. Nitrate is the end product of nitric oxide<br />

metabolism and a measure of the whole body<br />

nitric oxide production rate. The circulating levels<br />

and urinary excretion of the leukotrienes are also<br />

specifically increased. There is always a reduction<br />

in cellular glutathione concentration, a key intermediate<br />

in protection of the body from free radicals.<br />

Measurement of the ratio of NADPH/NADP in<br />

cells from these children shows that they have a<br />

marked deficit in reducing equivalents.<br />

Furthermore, examination of the sulfhydryl groups<br />

on proteins shows that an unusually large proportion<br />

of them are oxidized. Incubation of normal<br />

red cells in vitro with peroxides reproduces the<br />

profile of membrane lipids found in kwashiorkor.


Although they also have a high intracellular<br />

sodium and a low potassium concentration, as in<br />

marasmus, this comes about by quite a different<br />

mechanism. Their cell membranes become leaky<br />

to sodium and potassium; it is this membrane leakiness<br />

that leads to the loss of intracellular potassium<br />

and an increase in intracellular sodium. In<br />

fact, the sodium pump is quite the opposite to that<br />

in marasmus; in kwashiorkor it is more active than<br />

normal and their cells have an increased complement<br />

of Na + K + ATPase. When cellular glutathione<br />

is reduced in normal cells experimentally, to the<br />

levels seen in edematous malnutrition, the leakiness<br />

of the membrane is reproduced and the same<br />

electrolyte abnormalities that are seen in kwashiorkor<br />

arise. In kwashiorkor, the cell membranes<br />

have been damaged by oxidation.<br />

In kwashiorkor there is also effacement of the<br />

podocytes of the glomerulus to resemble minimalchange<br />

nephrotic syndrome. However, there is no<br />

proteinuria. This is a morphological expression of<br />

a change in the surface anionic charge on the cell<br />

membranes. The defect can be corrected, in vitro,<br />

by perfusion with strongly anionic substances<br />

such as heparin. The loss of surface charge is due<br />

to the disruption of the glycosaminoglycans<br />

(GAGs) on the cell surface. Interestingly, these<br />

children do not get infections such as cholera<br />

which depend upon the organism attaching to<br />

surface complex carbohydrate receptors. Such<br />

receptor dysfunction may account for the abnormal<br />

renal sodium homeostasis, but at present there<br />

is no direct evidence for this speculation. The<br />

blood–brain barrier is dependent upon the<br />

integrity of surface GAGs. This may account for<br />

the fact that meningitis is uncommon in kwashiorkor<br />

(but not in marasmus). It also means that<br />

drugs that are neurotoxic if they enter the cerebrospinal<br />

fluid, but are normally excluded, should<br />

be used with extreme caution in edematous<br />

malnutrition. Ivermectin would be in this category.<br />

Protein synthesis is disrupted. Electron microscopy<br />

of various tissues shows a marked reduction<br />

of the protein synthetic machinery. Hepatic export<br />

proteins are not made at a sufficient rate to maintain<br />

their circulating concentrations. Reduction in<br />

lipoprotein synthesis may be sufficient to account<br />

for the vast accumulation of triglyceride in the<br />

liver although it is likely that other mechanisms<br />

are involved. For example, there are very few<br />

Edematous malnutrition 509<br />

peroxisomes seen in the cells and the abnormalities<br />

of NADPH biochemistry are likely to affect<br />

lipid metabolism profoundly.<br />

Most textbooks ascribe kwashiorkor to protein<br />

deficiency. However, this theory is now untenable.<br />

Protein deficiency would not account for the physiological<br />

and biochemical abnormalities. Further,<br />

experimental protein deficiency does not reproduce<br />

the clinical features of kwashiorkor in any<br />

species of animal. Being a type II nutrient, protein<br />

deficiency does give stunting and wasting. The<br />

edema can resolve completely on a very low<br />

protein diet with no change in plasma albumin<br />

level. In the community, no differences have been<br />

found in the protein contents of the diets of those<br />

who develop kwashiorkor and those who develop<br />

marasmus.<br />

How might poverty lead to kwashiorkor? First,<br />

poor people live crowded together, in a highly<br />

contaminated environment. They get many severe<br />

infections and small-bowel overgrowth. Indeed,<br />

epidemics of kwashiorkor follow epidemics of<br />

measles in the tropics, but not in the rich countries.<br />

Second, their diets are lacking in many of the<br />

type I nutrients that are crucial to protection from<br />

free radical damage, particularly: selenium,<br />

vitamin E, carotene, vitamin C, riboflavin, thiamine<br />

and nicotinic acid. From this list special<br />

mention should be made of selenium. Red cells<br />

become more dense as they age. Thus, by separating<br />

cells by their density, it is possible to look back<br />

in time to observe the selenium status during the<br />

evolution of the illness. Examination of the selenoenzyme,<br />

glutathione peroxidase, in age-fractionated<br />

cells shows that the children with kwashiorkor<br />

have become selenium deficient in the<br />

recent past. The areas of the world where kwashiorkor<br />

is common have mostly selenium-deficient<br />

soils.<br />

When an infection, toxin or drug gives rise to<br />

tissue damage, cytokine release, leukotriene<br />

synthesis and free radical production, these<br />

patients lack the ability to protect and repair their<br />

liver, skin and vessels from acute damage. These<br />

defenses depend upon an orchestra of type I nutrients<br />

collectively referred to as antioxidants. It is<br />

this acute oxidative damage that both causes the<br />

syndrome of kwashiorkor and accounts for its<br />

similarity to several other serious conditions<br />

usually found in intensive care units.


510<br />

Malnutrition<br />

Changes that occur during treatment<br />

During successful treatment the physiological and<br />

body compositional changes reverse. Unfortunately,<br />

the order in which normal function<br />

returns can endanger the patient. With increased<br />

intake of antioxidants the leak in the cell<br />

membrane of kwashiorkor is repaired. With the<br />

increase in dietary intake the slow pump of marasmus<br />

regains its function. The sodium inside the<br />

cell is exported. In malnutrition the intracellular<br />

sodium is about 70mmol/l cell water; the normal is<br />

about 30mmol/l cell water. If 40mmol/l were to be<br />

suddenly exported from the cell to be retained in<br />

the extracellular fluid at a concentration of<br />

135mmol/l, then there would be an increase of<br />

about 30% in the extracellular fluid volume. If this<br />

fluid were retained in the circulation it would<br />

precipitate volume overload and acute heart<br />

failure. In kwashiorkor, if the defect in the interstitial<br />

space is corrected, then edema fluid moves<br />

from the interstitial space to the vascular compartment;<br />

again, this leads to an additional expansion<br />

of the circulating volume in kwashiorkor.<br />

Although the children often present with hypovolemia,<br />

during early treatment the situation is<br />

completely reversed to a situation of hypervolemia.<br />

This is always associated with a fall in<br />

hemoglobin concentration. Indeed, the red cells<br />

function as a ‘marker’ of blood volume. The expansion<br />

of the total blood volume can be assessed by<br />

the magnitude of the fall in hemoglobin concentration.<br />

The liver enlarges with the expansion of<br />

the blood volume and there may be an increase in<br />

respiratory rate, distension of the neck veins,<br />

increased vascular shadows on chest X-ray and<br />

signs of pulmonary edema.<br />

The movement of sodium and water, to accumulate<br />

potentially in the vascular compartment,<br />

needs to take place after the kidney recovers its<br />

ability to excrete sodium. Where this does not<br />

happen the child is in great danger.<br />

The determining factors are thus:<br />

(1) The initial amount of edema fluid and increase<br />

in intracellular sodium;<br />

(2) The rate and degree of recovery of the tissue<br />

and cellular lesions;<br />

(3) The rate and degree of recovery of the renal<br />

lesions;<br />

(4) Sodium intake from the diet, oral rehydration<br />

fluid and intravenous fluid;<br />

(5) The additional expansion of the blood volume<br />

from a transfusion.<br />

Children with edema are more susceptible than<br />

those without edema, children with severe edema<br />

are much more susceptible than those with<br />

minimal edema, and children with marasmus from<br />

areas where kwashiorkor is also common are much<br />

more susceptible than marasmic children from<br />

areas where the dietary deficiencies that give rise<br />

to kwashiorkor are uncommon. This electrolytic<br />

dysequilibrium, leading to heart failure, accounts<br />

for the majority of deaths after admission and for<br />

the differences in mortality rates between different<br />

regions of the developing world.<br />

How can this be avoided? Even though antioxidant<br />

deficiency may be the immediate etiology of<br />

kwashiorkor, we should not treat the children with<br />

large amounts of antioxidants acutely. Modern<br />

diets (F75 and F100) contain significant amounts<br />

of the antioxidants, and the pathological changes<br />

reverse quite quickly in comparison to the diets<br />

that used to be used. If the staff do not understand<br />

the dangers of rapid reversal of the condition, fail<br />

to follow the protocol for the management of<br />

complications faithfully and do not guard against<br />

giving additional sodium or blood at this time,<br />

they should not use these treatments.<br />

As we do not understand the renal lesions, it is<br />

unclear how to ensure that they are corrected at an<br />

early stage. During the early phase of treatment,<br />

when the sodium pump is recovering and the<br />

excess intracellular sodium is being exported to<br />

the extracellular compartment, acute circulatory<br />

overload and sudden death can easily occur when<br />

the kidney recovers more slowly than the sodium<br />

pump.<br />

A low-sodium diet should always be used in the<br />

early stages. On admission the edge of the liver<br />

should be marked upon the skin with an indelible<br />

marker. Expansion and contraction of the liver is<br />

the easiest clinical way of assessing changes in<br />

blood volume (or changes in hematocrit, provided<br />

no one on the team will misinterpret this as<br />

anemia that requires transfusion).<br />

After admission diarrhea should be ignored unless<br />

it is associated with significant weight loss that is


not accounted for by resolution of edema, and<br />

contraction of the liver if it was initially enlarged.<br />

With treatment, great care should be taken that<br />

the weight, the respiratory rate or liver size does<br />

not increase excessively. If a blood transfusion is<br />

to be given, it must be given in the first 24h after<br />

admission and the diets should not be given<br />

simultaneously. After this window is passed a<br />

blood transfusion should not be given (unless the<br />

staff have the capacity to do a small exchange<br />

transfusion) for at least 2 weeks. A hemoglobin<br />

level that falls after admission should not be<br />

treated. Intravenous lines should not be established<br />

to give drugs. Quinine infusions should be<br />

avoided. Indwelling canulae are also dangerous<br />

because of the need to administer anticoagulants.<br />

Large doses of the sodium salts of drugs (antibiotics,<br />

antacids, etc.) should not be used.<br />

Heart failure is not easy to diagnose in this situation<br />

and many children are incorrectly diagnosed<br />

as having pneumonia. Persistent diarrhea, with<br />

which the child has already survived for several<br />

weeks, should not be treated acutely with rehydration<br />

fluids.<br />

Investigations<br />

A careful history and examination usually provide<br />

all the information required to treat these patients.<br />

The laboratory has a relatively minor role to play<br />

and is used for the identification and characterization<br />

of infection.<br />

Tuberculosis is common; its diagnosis presents<br />

special difficulties as the mantoux test is usually<br />

negative irrespective of the presence or absence of<br />

active disease. Acid-fast bacilli can sometimes be<br />

recovered from laryngeal aspirate obtained with a<br />

mucus extractor. A chest X-ray should be scrutinized<br />

carefully for small tuberculous lesions.<br />

Children with tuberculosis should not be transferred<br />

to a tuberculosis ward where the staff have<br />

not been trained and organized to manage malnutrition.<br />

The management of the malnutrition takes<br />

precedence (over the course of a few weeks,<br />

malnutrition has a much worse prognosis and<br />

higher mortality rate than tuberculosis); tuberculosis<br />

treatment can be given in the malnutrition unit.<br />

Investigations 511<br />

On the chest X-ray, infection causes much less<br />

shadowing than in well-nourished children and<br />

may even be absent in the presence of bronchopneumonia.<br />

The blood should be examined for<br />

malarial parasites if routine treatment is not being<br />

given to all severely malnourished patients.<br />

Testing for sickle cell disease is important in many<br />

communities. Hematocrit or hemoglobin may be<br />

helpful immediately on admission, although<br />

anemia is usually clinically obvious. Measurement<br />

of plasma constituents is unhelpful in<br />

management. Plasma concentrations bear no<br />

necessary relationship to whole body content.<br />

This is particularly true for potassium and<br />

sodium. Hyponatremia is frequently found and is<br />

a poor prognostic sign.<br />

Therapy should be based upon clinical criteria; it<br />

should be guided by frequent reappraisal of the<br />

direction of clinical progress and does not require<br />

laboratory data. Investigations are used for help<br />

with diagnosis, not to control treatment.<br />

Management<br />

The aims of treatment are to identify and treat all<br />

the life-threatening problems whilst reversing the<br />

pathological and physiological abnormalities and<br />

deficiencies safely, then to feed the patient so that<br />

weight is gained at an accelerated rate to ‘catch<br />

up’ towards normal. In children the aim is to start<br />

to stimulate mental development, to protect the<br />

patient from relapse and to secure continued<br />

normal development after discharge.<br />

There are two main criteria for admission to residential<br />

care: first, the presence of anorexia of<br />

more than a few days’ duration; and second, any<br />

child who does not respond immediately to a trial<br />

of out-patient management, and who has either<br />

edema, regardless of weight, or


512<br />

Malnutrition<br />

The acute phase<br />

The mothers have often visited traditional healers,<br />

waited until it is clear that help is needed and the<br />

expense justified, and then traveled long distances.<br />

The ill children have to be triaged and ‘fast<br />

tracked’ through registration and out-patient<br />

clinics. They should all be given 10% sugar water<br />

as soon as they arrive. They should then have their<br />

weight and height measured, have a brief history<br />

and examination taken and be admitted. They<br />

should not be washed or manipulated excessively.<br />

The caretaker (mother) should be admitted with<br />

the child. There should be low adult beds for the<br />

child and the mother to sleep together.<br />

We need to treat infections and start to reverse the<br />

physiological changes without the treatment overloading<br />

the limited capacity of the heart, kidney,<br />

intestine or liver. The therapeutic implications of<br />

the reductive physiological adaptations are given<br />

in Table 31.5.<br />

Dehydration<br />

Marasmus itself emulates all the signs of dehydration.<br />

It is almost impossible to make the diagnosis<br />

by examination. For this reason diagnoses of dehydration<br />

in the malnourished are always provisional.<br />

Only significant acute diarrhea needs treatment;<br />

children with persistent diarrhea should not<br />

be ‘rehydrated’. A history of recent sinking of the<br />

eyes with significant watery diarrhea should be<br />

present. If the patient has true dehydration then<br />

this has to be treated with rehydration fluid. The<br />

patients are very vulnerable to over-hydration and<br />

fatal heart failure is common during the early<br />

stages of treatment. Additionally, there is nearly<br />

always some element of ‘toxic shock’ arising from<br />

their infections and small-bowel overgrowth.<br />

These patients are quite different from ‘normal’<br />

persons with diarrhea or other types of dehydration.<br />

The management of diarrhea in the normal<br />

child intentionally and correctly gives excess fluid<br />

to ensure that all children receive adequate<br />

amounts. The kidney will readily excrete any<br />

excess that is given. This situation does not apply<br />

to the malnourished child. They are sensitive to<br />

sodium. For these reasons, oral rehydration fluids<br />

are given for a only short time in limited amounts.<br />

The normal solution used, ReSoMal, has a<br />

lower sodium (45mmol/l) and higher potassium<br />

(40mmol/l) content than that used for normally<br />

nourished patients (Table 31.6). The patients<br />

are also depleted in magnesium, zinc and phosphorus<br />

– solutions that contain these ions as well<br />

as the major electrolytes are particularly useful.<br />

Because of their difficulty with homeostasis, intravenous<br />

treatment is particularly dangerous and<br />

should be used only for unconscious patients with<br />

severe shock from dehydration.<br />

Rehydration is managed by taking serial weights.<br />

This gives an accurate, quick and reliable way of<br />

assessing rehydration. On admission, with even<br />

small amounts of weight gain, there should be<br />

definite and obvious clinical improvement if the<br />

diagnosis was correct. If there is weight gain<br />

without clinical improvement then the diagnosis<br />

was incorrect. Daily weights are taken from all<br />

patients under care; the dehydrating effect of diarrhea<br />

occurring after admission can then be<br />

assessed accurately from the weight change.<br />

Rehydration should never be given that increases<br />

the weight more than the pre-diarrheal weight.<br />

Dehydration denotes a lower than normal electrolyte<br />

and water content of the body. Edema<br />

denotes a higher than normal electrolyte and water<br />

content of the body. To say that an edematous<br />

patient is dehydrated is like diagnosing fever in a<br />

hypothermic patient – they are mutually exclusive<br />

diagnoses. That is not to say that a patient with<br />

kwashiorkor cannot be hypovolemic, indeed many<br />

are; but they are not dehydrated. The signs of<br />

‘dehydration’ are nearly always related to toxic (or<br />

cardiogenic) shock in the edematous patient.<br />

Shock<br />

There are three common types of shock in the<br />

severely malnourished: dehydration, toxic shock<br />

due to sepsis and cardiogenic shock. There are<br />

frequently elements of all three in the moribund<br />

malnourished child. They are very difficult to<br />

differentiate. Toxic shock is difficult to treat<br />

successfully. In toxic shock, the stress of endotoxemia<br />

is superimposed upon the other problems of<br />

severe malnutrition. The veins and capillaries<br />

dilate, the cardiac muscle is weakened and the<br />

blood pressure falls. The combined effects of the<br />

toxin, the metabolic changes produced by<br />

cytokines and low perfusion of the organs leads to<br />

increasing shock. Renal perfusion is reduced to a


Table 31.6 Composition of an oral<br />

rehydration solution suitable from<br />

malnourished children<br />

Component Concentration per liter<br />

Sodium 45mmol (45mEq)<br />

Potassium 40mmol (40mEq)<br />

Magnesium 3mmol (6mEq)<br />

Glucose 10g<br />

Sucrose 25g<br />

Osmolality 291mOsm<br />

Zinc 300µmol (19.5mg)<br />

Copper 45µmol (2.9mg)<br />

Selenium 0.6µmol (47µg)<br />

level where the kidney cannot excrete the endproducts<br />

of metabolism; the intestine fails to<br />

absorb and then secretes fluid, bowel movement<br />

diminishes and petechial bleeding occurs<br />

throughout the intestine; liver perfusion is<br />

reduced, gluconeogenesis becomes ineffective<br />

and the blood sugar falls; even the metabolic<br />

stress of processing dietary protein can give liver<br />

failure, and high-protein diets are dangerous;<br />

there is a progressive decrease of awareness<br />

because of poor cerebral perfusion, anoxia, electrolyte<br />

disturbance and hypoglycemia. The low<br />

perfusion of the tissues reduces the metabolic rate<br />

to a stage where there is insufficient heat to maintain<br />

body temperature. Incipient toxic shock<br />

needs to be recognized in its early stages and this<br />

progression prevented. Management depends<br />

upon the maintenance of cardiac output, the<br />

removal of the source of the toxin by treating<br />

infection, the prevention of hypoglycemia and<br />

hypothermia, the strict avoidance of any stress<br />

such as giving excess fluid or protein, the provision<br />

of nutrition and the correction of deficiencies.<br />

If the circulation is compromised, treatment<br />

is by plasma expansion.<br />

Diet<br />

The principle of dietary management in the acute<br />

stage is to give enough to prevent hypoglycemia<br />

and hypothermia, to prevent any further tissue<br />

catabolism and to allow the patient to begin to<br />

Management 513<br />

reassemble his cellular enzymes; this must be<br />

done within the capacity of the intestine, liver and<br />

other organs. However, at this stage, not only may<br />

the functional capacity be easily exceeded, but<br />

deficiencies and nutrient imbalances may be<br />

aggravated if the patient is given too much food. It<br />

is as important for the patient not to gain new<br />

tissue when he is in this state as it is to prevent<br />

the further loss of existing tissue. This is done by<br />

carefully controlling the amount of food that is<br />

given. Children are given not less than 80kcal/kg<br />

per day and not more than 100kcal/kg per day;<br />

infants can be given somewhat higher levels; in<br />

adults the intake should be 30–40kcal/kg per day.<br />

If less than 80kcal/kg per day is given to a child,<br />

he will continue to use his own tissues for food<br />

and will deteriorate; if more than 100kcal/kg per<br />

day is given, the child may develop a metabolic<br />

imbalance.<br />

Because the amount of food that the intestine,<br />

liver and kidney can handle is limited, to keep<br />

within the capacity of the organs the diet is<br />

divided into small portions given at frequent<br />

intervals (normally eight feeds per day). The more<br />

‘brittle’ the patient, the more restricted is his<br />

capacity and, thus, the smaller each feed has to be<br />

and the more frequently they are given. In the<br />

extremely ill patient, the diet is put into a nasogastric<br />

drip and given continuously; intravenous<br />

feeding can be used in specialized centers in rich<br />

countries, but the principles of management are<br />

exactly the same with these patients as with orally<br />

fed children in the tropics.<br />

In the less severely ill patient, the diet is given<br />

less frequently. Recently the success of day-care<br />

treatment has shown that uncomplicated cases<br />

can be fed during the day and allowed home at<br />

night.<br />

The diet should contain every essential nutrient<br />

(all the minerals, particularly potassium and<br />

magnesium, vitamins, protein and energy) the<br />

child needs to repair his tissues. If any one of the<br />

nutrients is not given in adequate amounts the<br />

child will not recover. He will suffer an acute<br />

deficiency of this nutrient as the energy intake<br />

increases. On the other hand, the diet should not<br />

contain a great excess of any nutrient as the<br />

excess will again cause a metabolic stress. The<br />

amounts of each of the nutrients that should be


514<br />

Malnutrition<br />

Table 31.7 The desirable nutrient intake (per kg body weight) from the diet during the<br />

catch-up phase of treatment. The diet is most easily formulated as a single diet of 1kcal/ml<br />

for transition phase and phase 2<br />

Per kg body weight/<br />

Nutrient Per kg body weight 100kcal<br />

Water 120–140ml 120–140ml<br />

Energy 420kJ 100kcal<br />

Protein 1–2g 1–2g<br />

Electrolytes<br />

Sodium 160mg<br />

Magnesium >0.6mmol >10mg<br />

Phosphorus 2.0mmol 60mg<br />

Calcium 2.0mmol 80mg<br />

Trace minerals<br />

Zinc 30µmol 2.0mg<br />

Copper 4.5µmol 0.3mg<br />

Selenium 60nmol 4.7µg<br />

Iodine 100nmol 12µg<br />

Water-soluble vitamins<br />

Thiamine 70µg 70µg<br />

Riboflavin 200µg 200µg<br />

Niacin 1000µg 1000µg<br />

Pyridoxine 70µg 70µg<br />

Cobalamin 100ng 100ng<br />

Folic acid 100µg 100µg<br />

Ascorbic acid 10mg 10mg<br />

Pantothenic acid 300µg 300µg<br />

Biotin 10µg 10µg<br />

Fat-soluble vitamins<br />

Retinol 150µg 150µg<br />

Calciferol 3µg 3µg<br />

Tocopherol 2.2mg 2.2mg<br />

Vitamin K 4µg 4µg<br />

Lipids<br />

Total lipid 25–55% energy<br />

N-6 fatty acids 4.5% energy<br />

N-3 fatty acids 0.5% energy<br />

used for treating malnourished patients are given<br />

in Table 31.7.<br />

Infections<br />

Because of the poor inflammatory response, the<br />

usual physical signs of infection in the malnourished<br />

are unobtrusive or absent; infection expresses itself<br />

as apathy, drowsiness, hypothermia, hypoglycemia<br />

and death. Nearly all malnourished children have<br />

infections; many have multiple infections. They<br />

have overgrowth of their small intestines with organisms<br />

normally present in the colon, and normally<br />

commensal organisms, such as Staphylococcus


epidermidis, become invasive. Infections are<br />

frequently the immediate cause of death.<br />

Early, effective treatment with antibiotics is critical<br />

in suppressing small-bowel overgrowth, pre-venting<br />

toxic shock, improving the nutritional response to<br />

feeding and preventing mortality. For these reasons<br />

blind unselective wide-spectrum antibiotic treatment<br />

is recommended for all patients with severe<br />

malnutrition. Oral amoxicillin is a good choice. It<br />

effectively suppresses the bowel overgrowth (this<br />

disturbance of the flora is why it sometimes<br />

provokes diarrhea in normal children) and is also<br />

active against most of the common organisms. Some<br />

clinicians believe that antibiotics should be<br />

prescribed only for clearly defined infection. This is<br />

to confuse prophylaxis with blind treatment. Often<br />

microbiologists advocate specific treatment of the<br />

organisms they manage to isolate and recognize as<br />

pathogenic; this can be dangerous in malnourished<br />

patients because of the multiple organisms, the<br />

need to suppress commensals and failure to sample<br />

most of the potential sites of infection.<br />

Measles<br />

This involves herpes and other systemic viral infections.<br />

The mortality rate of severely malnourished<br />

patients with measles is very high. To reduce the risk<br />

of cross-infection from a newly admitted patient<br />

who is incubating measles, it is recommended that<br />

measles vaccine be given to all malnourished children<br />

on admission. Children with severe malnutrition<br />

often have vitamin A deficiency. This is particularly<br />

associated with death from measles.<br />

Vitamin deficiency<br />

In regions where measles or vitamin A deficiency is<br />

known to occur, even if clinical vitamin A deficiency<br />

is uncommon, vitamin A should be<br />

routinely given to all malnourished children on<br />

admission. They should receive three doses: one<br />

each on the 1st, 2nd and 14th day. The patients are<br />

normally also folic acid deficient. All patients<br />

should receive a single dose of 5mg folic acid<br />

orally, on admission. It should not be repeated, as<br />

large doses of folic acid interfere with malaria treatment.<br />

Many patients are also deficient in<br />

riboflavin, ascorbic acid, pyridoxine, thiamine and<br />

the fat-soluble vitamins D, E and K. Where specific<br />

Management 515<br />

deficiencies are known to be common, the amounts<br />

can be increased in the therapeutic diet. However,<br />

for patients with clinical signs, therapeutic doses of<br />

the nutrients should be given. The commercial<br />

preparations of F75 and F100 have ample micronutrients<br />

and none need be given individually.<br />

Hypoglycemia<br />

All malnourished patients can develop hypoglycemia<br />

when they are fasted. However, this<br />

complication has been overemphasized in the past.<br />

In one series, one case of asymptomatic low blood<br />

sugar was found in 200 patients. The success of<br />

home, out-patient and day-care programs has<br />

confirmed that this is an uncommon complication.<br />

If there is concern about hypoglycemia then it can<br />

be prevented by giving frequent feeds throughout<br />

the day and night.<br />

Eyelid retraction is a sign of sympathetic overactivity<br />

that is frequently present in children who<br />

are actively attempting to maintain their blood<br />

sugar (or circulating volume in dehydration). If a<br />

child sleeps with its eyes open, that child should be<br />

awoken and given sugar water (10%). A low body<br />

temperature, lethargy, limpness and clouding of<br />

consciousness are other features of hypoglycemia;<br />

unlike normal persons, sweating and pallor do not<br />

usually occur. If hypoglycemia is suspected, treatment<br />

should be given immediately.<br />

If the patient is conscious or can be roused and will<br />

drink, give sugar in water or a formula feed by<br />

mouth, whichever is most quickly available, and<br />

stay beside the patient until he is alert.<br />

If the patient is losing consciousness, is unrousable<br />

or has convulsions, a bolus injection of 1ml/kg<br />

body weight of sterile 10% glucose should be given<br />

intravenously followed by nasogastric 10% sucrose<br />

to prevent recurrence.<br />

When the patient regains consciousness, an oral<br />

feed of a milk preparation or of sugar water is given<br />

immediately. This patient should then be given<br />

frequent oral feeds to prevent recurrence.<br />

Hypothermia<br />

The neutral temperature for malnourished patients is<br />

25–30°C. Staff, who are active and fully clothed,


516<br />

Malnutrition<br />

often find this temperature uncomfortably warm.<br />

They do not realize that a room temperature that is<br />

comfortable for themselves (20–24°C) is too cold<br />

for malnourished patients, particularly small,<br />

immobile, malnourished children. Young infants,<br />

those with marasmus, those with large areas of<br />

weeping skin and infected patients are particularly<br />

susceptible to hypothermia. Newly admitted patients<br />

should not be nursed near windows or other<br />

drafts. At night the mother should sleep in the<br />

same bed as the child. Little cages (cots or cribs)<br />

are used in many pediatric wards – they promote<br />

hypothermia, inhibit breast feeding and cause<br />

psychological stress for the child. The mother does<br />

not get adequate rest and this compromises her<br />

care of her child. Cots are for the benefit of the<br />

staff. The patients should be properly covered with<br />

hats, clothes and blankets. Washing should be kept<br />

to a minimum and done during the day; when<br />

patients are washed they must be immediately and<br />

carefully dried and not left wet. Hypothermia is a<br />

consequence of a low metabolic rate.<br />

When rectal temperature is below 35.5°C (94.9°F)<br />

or underarm temperature below 35°C (95°F) the<br />

patient should be warmed. This is best done by<br />

using the ‘kangaroo’ technique. The carer is the<br />

‘incubator’. The child is put on the mother’s bare<br />

chest and both are covered with a blanket; the<br />

mother is then given a hot drink to increase her<br />

skin blood flow. All hypothermic patients must<br />

also be treated for hypoglycemia.<br />

Severe anemia<br />

Deficiencies of the hematinics, folate, copper,<br />

vitamin E, ascorbic acid, pyridoxine and riboflavin<br />

are common. In urban areas blood levels of lead<br />

can be high. Malaria, hemoglobinopathies, favism<br />

and hookworm are regionally common. However,<br />

iron deficiency is not common, except in patients<br />

with chronic blood loss from intestinal helminths.<br />

The hemoglobin level usually increases rapidly<br />

during catch-up growth. Iron is added to the treatment<br />

only after transferrin has been resynthesized<br />

and bacteria are adequately treated. Moderately<br />

severe anemia is not otherwise treated.<br />

If the hemoglobin concentration is less than 40g/l<br />

on admission, then the patient can have a slow<br />

blood transfusion: 10ml/kg body weight of whole<br />

blood or packed cells over at least 3h. Anemia that<br />

is recognized after admission should not be<br />

treated. If the patient has heart failure secondary to<br />

anemia then the blood should be given as an<br />

exchange transfusion (an equal volume of blood is<br />

taken to that transfused; this is done by taking<br />

2.5ml/kg before transfusion and then at hourly<br />

intervals during the transfusion). It should be<br />

noted that anemia and heart failure often coexist.<br />

Heart failure that is due to the anemia is a highoutput<br />

failure, similar to beriberi, with warm<br />

peripheries and a wide pulse pressure. It is dangerous<br />

to transfuse a patient with anemia in heart<br />

failure, if the heart failure is due to other causes.<br />

Congestive heart failure and acute left heart failure<br />

These are common complications and causes of<br />

death. They occur as a complication of overhydration<br />

(especially when intravenous fluids are<br />

given), blood transfusion, plasma transfusion, or<br />

with formulae or drugs that have a high sodium<br />

content. It can also occur during early recovery<br />

when sodium effluxes from the cells and edema<br />

fluid is resorbed into the circulation at a faster rate<br />

than can be excreted by the kidney. Heart failure<br />

may be more common in areas where the diet is<br />

low in selenium.<br />

The important signs to look for are clinical deterioration<br />

with a gain in weight, an increasing respiratory<br />

rate or a rapid enlargement of the liver<br />

(Figure 31.10). Treatment should be started at this<br />

stage. However, heart failure has to be differentiated<br />

from inhalation, respiratory infection and<br />

toxic shock. The simplest way to make this differentiation<br />

is to examine the weight change.<br />

Respiratory distress with weight loss should be<br />

treated as pneumonia; with weight gain it should<br />

be treated as heart failure. The patient must be reexamined<br />

at very frequent intervals after any treatment<br />

is started so that, if there is not a rapid<br />

improvement, the diagnosis can be revised. The<br />

most useful signs are liver size and respiratory<br />

rate. Later signs are respiratory distress, rapid<br />

pulse, venous engorgement, cold hands and feet<br />

and a purple discoloration under the fingernails<br />

and tongue. Left heart failure often presents as<br />

sudden unexpected death.<br />

When heart failure is diagnosed, all oral intake and<br />

intravenous fluids should be stopped (the treatment<br />

of heart failure takes precedence over


feeding); the patient is fasted. This will ensure a<br />

negative fluid balance. If there is a danger of hypoglycemia<br />

then small amounts of 10% sugar water<br />

should be given orally. The fasting may also have<br />

the effect of allowing some sodium to return to the<br />

intracellular or interstitial compartments. It<br />

should be continued until the weight gained (a<br />

measure of the amount of fluid retention) since the<br />

start of symptoms has been re-lost, the respiratory<br />

rate has settled and the liver has returned to its<br />

admission size. A diuretic is given intravenously;<br />

the most appropriate diuretic is 1mg/kg of frusemide.<br />

Most malnourished patients do not respond<br />

to this diuretic; it is given because some<br />

patients do respond and this cannot be predicted.<br />

As these patients have a low total body potassium<br />

level, frequently hypokalemia, full doses of digitalis<br />

are never given; however, digitalis inhibits the<br />

sodium pump and allows sodium to re-enter the<br />

cell. A single dose of 5µg/kg digoxin should be<br />

given.<br />

If the patient continues to deteriorate, the physician<br />

is certain of the diagnosis of heart failure and<br />

heroic measures are definitely called for. The<br />

patient can then be given a 5ml/kg venesection.<br />

The blood should be kept sterile in the syringe,<br />

and the patient’s condition carefully watched. If he<br />

deteriorates, then the blood should be slowly<br />

returned; if there is marked improvement over the<br />

next 10min then the diagnosis was correct. No<br />

more than 10ml/kg should ever be taken. Although,<br />

in the right circumstances this is a highly<br />

effective treatment, it should be used only by those<br />

with considerable experience. In practice, unless<br />

the parent fully understands and trusts the staff,<br />

such treatment opens the staff to being accused of<br />

causing the death by bleeding the child.<br />

Experience with using agents that are used in<br />

other forms of heart failure have not been reported<br />

in heart failure associated with malnutrition.<br />

When heart failure is due to severe anemia then<br />

the treatment is by exchange transfusion, which is<br />

effectively transfusion and venesection combined.<br />

The amount of anemic blood withdrawn should<br />

exceed the amount of normal blood given.<br />

Transition phase<br />

By the end of the acute phase the patients should<br />

have all their complications treated, edema should<br />

Management 517<br />

Figure 31.10 An autopsy of a child with marasmickwashiorkor.<br />

Note the fatty liver, the presence of some<br />

subcutaneous fat, the greatly distended loops of bowel<br />

and the thinned transparent nature of the bowel wall.<br />

be resolving and treatment for infections started. A<br />

return of appetite is the sign that the patient is<br />

ready to progress to the transition phase. The<br />

appetite is used as a barometer of progress. Loss of<br />

appetite occurs when the patient’s metabolic<br />

processes cannot cope with the dietary intake.<br />

Great care must be taken not to overload the<br />

patient whilst he has a poor appetite – this is a<br />

warning that something is wrong metabolically.<br />

The diet is changed to a high-energy diet called<br />

F100, but it is given in restricted amounts (Table<br />

31.8). The volume given is the same as the diet<br />

given during the acute phase (F75), but as it is<br />

more concentrated the intake is increased by 25%.<br />

One would anticipate a weight gain on this diet of<br />

about 6g/kg per day. No other changes are made to<br />

the regimen.


518<br />

Malnutrition<br />

Because these patients may not be able to express<br />

thirst, their requirement for water may not be met<br />

from the formula alone. There is a considerable<br />

renal solute load from F100. High respiratory rates<br />

and fever, and infants and those treated in dry<br />

environments require extra water. This should be<br />

added to the formula, and the amount dispensed<br />

commensurately increased, if there is any doubt<br />

that all the children who require additional water<br />

will receive it.<br />

If any complications arise during the transition<br />

phase the children should be returned immediately<br />

to phase 1. No child should be force-fed or<br />

have a nasogastric tube in the transition phase.<br />

Milk intolerance<br />

Lactose or milk intolerance is seldom seen, even<br />

among severely malnourished patients. All patients<br />

should be started on the milk-based formulas.<br />

Intolerance is diagnosed only if copious watery<br />

diarrhea occurs with the introduction of milk feeds,<br />

it clearly improves when milk intake is reduced and<br />

it recurs again when the patient is challenged with<br />

milk a second time. In such cases, milk formulas<br />

can be partially or totally substituted by other liquid<br />

foods. Milk feeds should be reintroduced and their<br />

effects noted before a patient is discharged with a<br />

‘diagnosis’ of milk intolerance.<br />

Very occasionally other strategies are needed. The<br />

simplest is to ferment the milk to yoghurt. This is<br />

highly effective. Lactose- and milk-free formulas<br />

can be used but they are rarely available where<br />

severe malnutrition is common. Sometimes the<br />

addition of pancreatic enzymes (such as Pancrex<br />

V ® ) to the diet is beneficial, particularly in cases of<br />

severe kwashiorkor.<br />

Rehabilitation phase<br />

Together the acute and transition phases of treatment<br />

usually take about 7 days, but this is quite<br />

variable. The features that determine whether the<br />

patient has entered the rehabilitation phase are the<br />

complete loss of edema and a good appetite. In older<br />

patients with primary intestinal disease who<br />

require intravenous or nasogastric drip feeding,<br />

feelings of hunger and ‘well-being’ denote this<br />

phase of management.<br />

Table 31.8 A formula for use in<br />

malnourished children (F100)<br />

Ingredients Amount<br />

Dried skim milk 80g<br />

Sugar 50g<br />

Oil 60g<br />

Mineral mix to give the concentrations in<br />

Table 31.7<br />

Vitamin mix to give the concentrations in<br />

Table 31.7<br />

Water to 1000ml<br />

There is a great deal of flexibility in the way in<br />

which the rehabilitation phase of treatment is<br />

managed. There are now safe ready-to-use therapeutic<br />

formulas that can be used to continue the<br />

treatment at home. When well organized, with a<br />

home visitor and a good home environment, this<br />

approach is preferable.<br />

The return of appetite means that infections are<br />

under control and there is no major electrolyte<br />

imbalance or deficiency, even though the patient’s<br />

physiological responses are still abnormal and his<br />

capacity may be limited. At this stage the patient<br />

has resynthesized the cellular components needed<br />

to absorb and metabolize more food than is necessary<br />

for mere maintenance; he is ready to make new<br />

tissue. The single most important thing that determines<br />

the rate of recovery is the amount of the diet<br />

taken by the patient. The overriding principle of<br />

this phase is to feed the patient to appetite and to<br />

actively encourage him to eat.<br />

Moving from the transition and the rehabilitation<br />

phases is achieved simply by increasing the amount<br />

of the same diet that is given until the patient starts<br />

to refuse to finish the feed. Sufficient time must be<br />

spent with the patient to enable him to try to finish<br />

each feed; the attitude of the caretaker is crucial to<br />

success. The patient should never be left alone to<br />

‘take what he wants’ by himself – this is not what is<br />

meant by feeding to appetite.<br />

As the patient gains weight, his appetite and<br />

requirement steadily increase. Each day the weight,


the record of intake and the amount refused are<br />

plotted on a specially designed treatment chart.<br />

During rehabilitation most children and even adults<br />

take between 150 and 200kcal/kg per day.<br />

The formula should not be discontinued if visible<br />

fat appears in the stools, as a high proportion of<br />

fat is still absorbed. Fat malabsorption is,<br />

however, a frequent cause of the patient’s failing<br />

to gain weight at the expected rate. Fat content<br />

should be decreased only if it clearly produces<br />

watery diarrhea.<br />

It is appropriate to maintain the patients on the<br />

formula until they have achieved about 85%<br />

weight-for-height (children) or a BMI of 18kg/m 2<br />

(adults). At this time they usually signal their individual<br />

needs by reducing their appetites somewhat.<br />

They are then ready for discharge.<br />

With adults the general principles of giving sufficiently<br />

high intakes of energy, protein and all minerals<br />

and vitamins to enable them to gain weight<br />

rapidly must still be followed, despite the fact that a<br />

mixed diet is given. Many normal diets have a low<br />

energy density (in the tropics), and have insufficient<br />

protein, minerals and vitamins to sustain the rapid<br />

rates of growth. Further, the mixed diets often<br />

contain phytic acid and other anti-nutrients that<br />

markedly reduce the absorption of phosphorus,<br />

zinc, iron and calcium. To counter these effects,<br />

F100 should be given between meals of the mixed<br />

diet. For example, if a mixed diet is given three<br />

times daily the formula feed should also be given<br />

three times daily to make six ‘meals’ each day.<br />

Severely malnourished patients have a reduced<br />

iron-binding capacity; they are neither able to withhold<br />

iron from invading organisms nor to prevent<br />

the toxic effects of free iron itself. During the acute<br />

and intermediate phases of treatment iron should<br />

not be given, even in the presence of severe anemia.<br />

In the rehabilitation phase an iron supplement<br />

should be given.<br />

Assessing progress<br />

The patients should be weighed daily and their<br />

weight plotted on the special chart. It is useful to<br />

mark the target weight (BMI for adults) on the<br />

graph. They should gain weight at 5–20g/kg per day,<br />

usually about 10–15g/kg per day.<br />

Management 519<br />

Emotional and psychological stimulation<br />

At home patients, particularly children, are<br />

surrounded by familiar places and people. It is a<br />

major psychological trauma for a child to be separated<br />

from its mother, family and surroundings and<br />

to be closed into a cot with bars. The child needs to<br />

be with its mother and there should be adult beds<br />

for even very young patients so that the mother can<br />

sleep with the child. When the child is acutely ill<br />

the mother will sit by the bed. As he recovers he<br />

should be actively encouraged to interact with the<br />

other children. This is often discouraged by hospital<br />

staff on the grounds that it increases crossinfection<br />

and makes their job more difficult. This<br />

attitude, usually from senior staff, retards recovery<br />

and is much more damaging than the risk of crossinfection.<br />

The malnourished child needs affection and tender<br />

care from the very start of treatment, and interaction<br />

with other children when he becomes active. This<br />

requires patience and understanding by the hospital<br />

staff and the caretaker. When the children are over<br />

the acute and intermediate phases, they can be up<br />

and about for prolonged periods on large play mats.<br />

The risk of cross-infection is not increased substantially<br />

and the benefit for the children is much more<br />

important than the convenience of the staff.<br />

In hospital, it is not infrequent for ten different<br />

adults to interact with a patient over the course of a<br />

day, each one manipulating but rarely talking to or<br />

‘cuddling’ the patient. This is not good. It is particularly<br />

bad if the mother is not with the child. Each<br />

adult should talk, smile and laugh with the patient<br />

affectionately.<br />

Provision of child-oriented care, with affection and<br />

tenderness, is, of itself, insufficient. Severely<br />

malnourished children have delayed mental and<br />

behavioral development that requires treatment<br />

just as much as their delayed physical development.<br />

If such delays persist untreated they become<br />

the most serious long-term result of malnutrition.<br />

Psychological stimulation through play programs<br />

that start in hospital and continue after discharge<br />

can substantially reduce the mental retardation.<br />

The austerity of the traditional hospital should not<br />

be used for children in phase 2. Traditional buildings<br />

such as the child lives in at home are much<br />

more appropriate. They can often be erected in the<br />

grounds of a hospital or health center.


520<br />

Malnutrition<br />

There should be a large, safe, fenced area, with mats<br />

on the floor, where the children can play. Either<br />

mosquito nets or brightly colored mobiles should be<br />

over every bed. Hospital rooms should be brightly<br />

colored, with cheerful decorations that the children<br />

can relate to. Where possible the staff should not<br />

wear uniforms, or at least, have uniforms similar in<br />

design to standard maternal dress. Care is to be<br />

taken to avoid sensory deprivation; the child’s face<br />

should not be covered, they must be able to see and<br />

hear what is going on around them; they should not<br />

be wrapped or tied to prevent them moving around<br />

in the bed or on the floor.<br />

Toys must always be available. The mothers should<br />

be taught the importance of play and shown how to<br />

make the play materials. The toys should be safe,<br />

washable and appropriate for the child’s level of<br />

development.<br />

In hospital, one person should be in charge of organizing<br />

and running a play program. The play therapist<br />

should introduce new activities and play materials<br />

regularly; the activities should develop motor<br />

and language skills. It is useful to have a curriculum<br />

of activities and to play with each child in a structured<br />

way for 15–30min each day. This is in addition<br />

to the informal mixing of the children. The<br />

mothers should be taught the elements of the<br />

curriculum, at the appropriate level. It must be<br />

emphasized that the family should continue to<br />

make toys and play with their children after<br />

discharge, and indeed, throughout the whole period<br />

of development of their child. For this reason, teaching<br />

the mother is even more important than teaching<br />

the child. The mother should be intimately<br />

involved in the play therapy.<br />

Not only does increased physical activity encourage<br />

the development of the motor skills needed to<br />

explore the environment and play effectively but it<br />

may also enhance growth during nutritional rehabilitation.<br />

In immobile children, passive limb movements<br />

and allowing them to splash in a warm bath<br />

are helpful.<br />

Preparation for discharge<br />

There is not a sharp dividing line between the end<br />

of the rehabilitation phase and the preparation for<br />

discharge. The latter should be started during<br />

rehabilitation and completed after discharge.<br />

By the time of actual discharge the child needs to<br />

be ready for full integration into the family and<br />

community. As this is the environment that led to<br />

malnutrition in the first place, the family has to be<br />

equipped to cope with the patient to prevent recurrence.<br />

A child is usually considered ready for discharge<br />

when he reaches 85% of weight-for-height.<br />

Although these children are often referred to as<br />

‘recovered’, this is not usually true. Many have the<br />

underlying effects of chronic malnutrition with<br />

stunting in height and delayed mental development;<br />

these defects also require attention.<br />

However, management of these conditions<br />

depends upon long-term changes aimed at improving<br />

the resources, diet, hygiene, knowledge and<br />

skills of the family as a whole, and not upon care<br />

directed at the child alone.<br />

Attaining normal weight-for-height, of itself, is<br />

insufficient to discharge a child from the program.<br />

One of the problems with traditional hospital<br />

management is that anthropometric criteria alone<br />

are used for discharge, and follow-up is inadequate.<br />

Whenever possible, a home visit should be made.<br />

The child should be enrolled in a supplementary<br />

feeding program and followed there with regular<br />

weighing for 4–6 months after ‘recovery’. If there is<br />

no supplementary feeding program then community<br />

health workers or the local health clinic should<br />

follow these patients particularly. Where the patient<br />

is abandoned or the social and economic conditions<br />

at the home are hopeless, often because of absence<br />

of a caretaker, appeals should be made for foster<br />

homes or other forms of community support.<br />

The patient and caretaker have much to achieve and<br />

learn while they are in contact with the health staff.<br />

Education cannot be left to the last few days before<br />

the patient is discharged. Parents or caretakers<br />

should be taught about the causes of the patient’s<br />

malnutrition and how to prevent it in the future,<br />

whether the malnourished person is a child or<br />

infirm and elderly. For children, the parents need to<br />

know the consequences of malnutrition on future<br />

development and the steps that need to be taken,<br />

over a considerable time, to reverse any mental<br />

impairment and stunting in height. Practical<br />

instructions should be given on how to feed the<br />

child and continue nutritional rehabilitation at


home. As these families are the most vulnerable<br />

within a community, the opportunity can usefully<br />

be taken to give instruction on child feeding and<br />

rearing practices, family planning, personal<br />

hygiene, methods of income generation, sexually<br />

transmitted diseases and many other topics.<br />

Before discharge, children must be vaccinated in<br />

accordance with the local health regimen and provision<br />

made for booster doses to be given at the appropriate<br />

time.<br />

General considerations<br />

A child with severe malnutrition may indicate a<br />

serious problem in his household; the other children<br />

are also at risk. Therefore, nutritional and<br />

health education is not restricted to avoiding a<br />

recurrence of the index case, but should include the<br />

prevention or correction of nutritional problems of<br />

all the family members, especially young children,<br />

pregnant and lactating women and the elderly.<br />

The presence of children with severe malnutrition<br />

also suggests a high prevalence of malnutrition in<br />

that community. Perhaps a survey should be<br />

conducted. Most relief agencies determine where<br />

the malnourished live in order to plan their surveys<br />

and programs. Health promotion can include<br />

education and promotional programs for community<br />

leaders, local action groups and the community<br />

as a whole. Such programs focus on the psychological<br />

needs of vulnerable groups, promotion of breast<br />

feeding, appropriate use of weaning foods, nutritional<br />

alternatives using traditional foods, the<br />

absolute importance of dietary variety in order to<br />

get all the essential nutrients, personal and environmental<br />

hygiene, water management, adequate<br />

feeding practices during illness and convalescence,<br />

immunizations, early treatment of diarrhea, pneumonia<br />

and other diseases, income-generating strategies<br />

and adult literacy classes.<br />

Additional problems with management of<br />

malnourished patients<br />

Staff attitudes<br />

Sometimes a patient fails to respond because staff<br />

have a ‘feeling’ that their efforts for a particular<br />

patient are unlikely to succeed. This is particu-<br />

General considerations 521<br />

larly acute if they think that the child has HIV. In<br />

some places all failures are put down to HIV<br />

without any justification. Where such attitudes<br />

exist, the progress for the patient is poor; this in<br />

turn falsely confirms the ‘correctness’ of the original<br />

attitude and efforts are not made for future<br />

similar patients. This cause of failure-to-respond<br />

is among the most difficult to address, because it<br />

is usually based on the ‘experience’ of a longserving<br />

senior staff. The dependence of each<br />

patient’s well-being and survival on the attitudes<br />

of staff should be earnestly reviewed with them.<br />

Infections<br />

Atypical clinical manifestations of common<br />

infections occur in malnourished children.<br />

Infections with organisms that are not pathogenic<br />

in the well nourished are also common. The<br />

infections that are most commonly overlooked<br />

and interfere with a good nutritional response<br />

include: urinary tract infection, otitis media,<br />

tuberculosis, congenital syphilis, cytomegalovirus<br />

infection, hepatitis, AIDS, dengue, giardiasis,<br />

cryptosporidiosis and small-bowel bacterial<br />

overgrowth with secondary malabsorption.<br />

HIV is often thought to be a major factor in<br />

centers that have poor results. This is a false<br />

assumption. In one center in Burundi, patients<br />

with HIV had a 6% mortality whilst those<br />

without had a 5% mortality (difference not significant;<br />

Y. Grellety, personal communication). The<br />

length of phase 1 and total stay was only 1 day<br />

longer in the HIV-positive patients. A high prevalence<br />

of HIV is not a reason for failure; these children<br />

and adults should not be abandoned.<br />

Treating the malnutrition is one very positive<br />

thing that can be done for these patients. In retrospect,<br />

this result should not surprise, because the<br />

state of the immune system and infection is very<br />

similar in HIV disease and severe malnutrition.<br />

Specific nutrient deficiencies<br />

Many specific nutrient deficiencies impair the<br />

immune response. They can be the underlying<br />

cause of an infection that does not respond as<br />

expected to antibiotics. Occasionally patients may<br />

have a profound deficiency of one or more specific<br />

nutrients that are not adequately replaced by the


522<br />

Malnutrition<br />

therapeutic diet or nutrient supplements. Many<br />

commercial multivitamin-and-mineral preparations<br />

lack particular nutrients or do not contain<br />

sufficient amounts to treat a deficiency. They are<br />

generally designed for use as supplements by<br />

healthy individuals. Use of such preparations<br />

leads to overlooking a deficiency assumed to be<br />

‘covered’ by the treatment. The most frequently<br />

overlooked specific deficiencies include those of<br />

zinc, magnesium, copper, selenium, folic acid and<br />

vitamin E, although other micronutrients may be<br />

involved. Insufficient amounts of potassium,<br />

magnesium and phosphorus in the diet are common<br />

errors.<br />

Associated pathological conditions<br />

Malnutrition accompanies many congenital abnormalities,<br />

inborn errors of metabolism, tumors,<br />

immunological diseases and diseases of the major<br />

organs. All these conditions occur in areas where<br />

primary malnutrition is frequent, just as in rich<br />

countries, although they are less frequently recognized.<br />

Where primary malnutrition is prevalent<br />

children with secondary malnutrition are usually<br />

misdiagnosed as having primary malnutrition. As<br />

the prevalence of primary malnutrition falls the<br />

proportion of malnourished children who have an<br />

associated pathology rises. Failure to respond to<br />

treatment should lead to investigation of the major<br />

organ systems for primary pathology.<br />

Specific dermatosis of kwashiorkor<br />

Spontaneous resolution can be expected with<br />

improved nutrition. Atrophy of the skin in the<br />

perineum leads to severe napkin dermatitis, especially<br />

in children or the elderly with diarrhea or<br />

incontinence. The perineum should be left<br />

exposed to dry without napkins. A barrier such as<br />

zinc and castor oil ointment, petroleum jelly or<br />

paraffin gauze dressing (tulle gras) to raw areas<br />

helps to relieve pain and prevent infection. The<br />

management should be the same as that used for<br />

burns. The zinc supplement contained in the diet<br />

may be insufficient in these patients, as zinc deficiency<br />

almost always accompanies severe skin<br />

lesions. These patients should always be given<br />

systemic antibiotics.<br />

Candidiasis<br />

Most malnourished children have candidiasis. In<br />

the mouth it can be seen as whitish plaques.<br />

However, even when the mouth is free of lesions,<br />

it may occur in the esophagus, stomach and<br />

rectum as well as on any damp moist skin. In<br />

severe malnutrition systemic candidiasis with<br />

growth in the respiratory tract and blood also<br />

occurs. Oral nystatin suspension should be given<br />

to all patients with candidiasis. In addition,<br />

nystatin cream should be applied to any cutaneous<br />

lesions. Systemic candidiasis should be treated<br />

with drugs such as ketoconazole.<br />

Drug metabolism in malnourished patients<br />

Clearly the physiological and body compositional<br />

changes in the malnourished patient will alter the<br />

pharmocokinetics of many drugs. There has been<br />

very little research on this aspect of either primary<br />

malnutrition or even the malnutrition associated<br />

with neoplasia or intestinal disease. A patient with<br />

7% of body weight as fat will have a very different<br />

response to a fat-soluble drug from one with 40%<br />

of body weight as fat, and yet this is rarely taken<br />

into consideration in prescribing. Poor absorption<br />

from the intestine, disordered hepatic conversion<br />

of drugs, reduced renal clearance, increased bacterial<br />

deconjugation of drugs excreted in the bile,<br />

and alterations in receptors and enzyme targets for<br />

drugs each affect their efficacy and potential toxicity.<br />

The changes are sufficiently complex for the<br />

result of giving a new drug to be unpredictable.<br />

Nevertheless, these factors should be considered<br />

in the treatment of all malnourished patients and,<br />

where available, therapeutic drug monitoring<br />

should be used. Clearly the variables are potentially<br />

extremely important. For example, if a<br />

patient with malnutrition, secondary to neoplasm,<br />

has features of kwashiorkor, is depleted in antioxidant<br />

nutrients and has a low cellular glutathione<br />

level, then giving cytotoxic drugs or radiotherapy<br />

may damage the patient irretrievably.<br />

Unnecessary drugs should never be given during<br />

the acute stage. Unwanted toxic effects are more<br />

likely for most drugs. The list of drugs that do not<br />

need to be used is long. In particular, fever should<br />

be treated with tepid sponging, not with paraceta-


mol. Vomiting should not be treated with the usual<br />

antiemetics (promethazine, etc.) and diarrhea<br />

should never be treated with an antispasmodic.<br />

Many of the drugs used to treat tuberculosis are<br />

hepatotoxic. It is wise to use these drugs carefully<br />

during the first few days after admission.<br />

Conclusions<br />

The problems raised by malnutrition affect not<br />

only the individual but society as a whole, at<br />

medical, social, ethical, moral and political levels.<br />

REFERENCES<br />

1. Pelletier DL. The relationship between child anthropometry<br />

and mortality in developing countries: implications<br />

for policy, programs and future research. J Nutr<br />

1994; 124 (10 Suppl): 2047S–2081S.<br />

2. Prudhon C, Briend A, Laurier D et al. Comparison of<br />

weight- and height-based indices for assessing risk of<br />

Conclusions 523<br />

Malnutrition in its numerous guises, especially<br />

amongst children and the elderly, is the most<br />

common serious illness in the world today.<br />

Lessons learnt from the study and management of<br />

these children have relevance for malnourished<br />

individuals of all ages and with a wide variety of<br />

disorders. A clear understanding of the etiology<br />

and pathogenesis is a prerequisite for designing<br />

effective intervention and prevention programs.<br />

The legacy of childhood malnutrition is to be seen<br />

in adults who are stunted physically and<br />

mentally; it may lay the seeds for many of the<br />

chronic diseases of unknown etiology in adult<br />

life.<br />

death in severely malnourished children. Am J<br />

Epidemiol 1996; 144: 116–123.<br />

3. Williams CD. A nutritional disease of childhood associated<br />

with a maize diet. Arch Dis Child 1933; 8: 423–428.


32<br />

Biotherapeutic and<br />

nutraceutical agents<br />

Kirsi Laiho and Erika Isolauri<br />

The science of nutrition: from basic<br />

needs to specific health effects<br />

The role of the diet in health has changed as scientific<br />

knowledge has increased (Figure 32.1). The<br />

basic foundation lies in a healthy, balanced diet<br />

that follows the dietary recommendations and<br />

guidelines appropriate for age and life-stage with<br />

the aim of meeting the metabolic requirements and<br />

the needs for growth and development in children.<br />

The first goal of nutritional management is<br />

directed towards the prevention of direct dietrelated<br />

deficiencies, such as scurvy caused by low<br />

Basic<br />

diet<br />

Balanced<br />

diet<br />

Specific<br />

diet<br />

Figure 32.1 The steps of nutrition management.<br />

Nutraceuticals<br />

Dietary recommendations and guides for a balanced diet<br />

to meet metabolic requirements and growth and development<br />

vitamin C intake. The second target is the prevention<br />

of nutrition-related chronic diseases. For<br />

example, an unbalanced diet containing excessive<br />

amounts of saturated fatty acids has been shown to<br />

be associated with an increased risk of coronary<br />

heart disease. Furthermore, specific requirements<br />

for nutritional management are set by a variety of<br />

diseases necessitating carefully planned diets or<br />

usage of clinical dietary products. The current<br />

research interest is directed towards the invention<br />

of novel dietary compounds with specific effects<br />

in health promotion and management of diseases<br />

beyond the nutritional impact of food.<br />

Dietary management and risk reduction<br />

of specific diseases by means of diet with<br />

added or modified nutrients or compounds<br />

nutraceuticals<br />

Dietary management of specific diseases by<br />

means of clinical dietary products<br />

e.g. hydrolyzed infant formula for cow's milk allergy<br />

or gluten-free cereals for celiac disease<br />

Dietary management of specific diseases (e.g. diabetes mellitus)<br />

by means of ordinary food<br />

Balanced diet with additional benefit to reduce<br />

the risk of nutrition-related chronic diseases<br />

Increasing scientific knowledge<br />

525


526<br />

Biotherapeutic and nutraceutical agents<br />

Dietary recommendations and guidelines<br />

appropriate for age and life-stage<br />

Dietary recommendations and guidelines are set to<br />

direct dietary food and nutrient intake for maintenance<br />

of health. Specifically, dietary recommendations<br />

aim at prevention of nutrient deficiencies but<br />

also at reducing the risk of developing nutritionrelated<br />

chronic diseases.<br />

The dietary recommendations vary amongst<br />

regions so that many countries have formed their<br />

own recommendations. However, major international<br />

health-related organizations such as the<br />

Food and Agriculture Organization (FAO) and<br />

World Health Organization (WHO) have searched<br />

for more uniform recommendations. 1–3 The reader<br />

is advised to refer to these or national original<br />

reports for detailed description of the recommendations.<br />

Dietary guidelines for populations have<br />

been given both as recommended daily intakes<br />

(recommended daily allowances, recommended<br />

nutrient intakes, dietary reference values/intakes)<br />

of certain nutrients and as food guides (e.g. food<br />

pyramid) that direct the consumption of foods and<br />

food groups. Food guides may also include recommendations<br />

on eating behavior and food habits.<br />

The recommended nutrient intake is generally<br />

understood as the level of nutrients sufficient to<br />

meet the daily nutrient requirements of most individuals<br />

of a specific age and gender, based on an<br />

estimated average nutrient requirement plus two<br />

standard deviations above the mean. Specific<br />

recommendations are given for children and<br />

adults as well as for pregnant and lactating<br />

women. The recommendations can be deployed to<br />

provide guidance on appropriate dietary composition,<br />

for the assessment of dietary surveys, for<br />

outlining food policies or for food labeling<br />

purposes. The food guides, such as the<br />

well-known food pyramid, are more comprehensible<br />

and particularly helpful in nutrition counseling.<br />

The guides direct the use of foods or food<br />

products and aim to meet the nutrient intakes set<br />

by the recommended daily intake.<br />

Early nutrition and later consequences<br />

It is understood that dietary intake that satisfies<br />

nutritional demands and maintains a good nutritional<br />

status is important for the growth and devel-<br />

opment of children. However, the nutrition of the<br />

infant begins before birth, in utero, when the nutrition<br />

of the mother and also placental function<br />

regulate the growth and development of the fetus.<br />

The nutrition of the mother even before pregnancy<br />

may be important for the well-being of the fetus<br />

and child, as the nutrient stores that are being<br />

utilized during pregnancy and breast feeding are<br />

accumulated at an early stage. 4 As a good example,<br />

a sufficient availability of folic acid and the<br />

prophylactic supplementation of folic acid to the<br />

mother reduce the incidence of birth complications<br />

and neural tube defects. 5 Indication of an<br />

unbalanced nutrient intake (maternal hypercholesterolemia<br />

during pregnancy) induces changes<br />

in the fetus that determine the susceptibility of<br />

children to fatty-streak formation and subsequent<br />

atherosclerosis. 6<br />

Both epidemiological 7–9 and experimental 10,11<br />

studies suggest that several chronic diseases of<br />

later life, including coronary heart disease, hypertension<br />

and type 2 diabetes, are programmed<br />

during the fetal period. This theory on the fetal<br />

origins of adult disease suggests that alterations in<br />

fetal nutrition and endocrine status result in developmental<br />

adaptations that predispose individuals<br />

to cardiovascular, metabolic and endocrine<br />

disease in adult life. One of the first reports<br />

showed that those individuals who had had low<br />

birth weights had relatively high death rates from<br />

coronary heart disease in adult life. 9 The results<br />

have since been replicated in other populations<br />

and with other chronic lifestyle-related diseases.<br />

Also, the mechanisms have been searched for in<br />

experimental studies and the specific causal relations<br />

are actively being explored. The research is<br />

also directed towards identifying critical phases in<br />

which the impact of nutrition is most important<br />

for the risk of chronic diseases.<br />

Specific dietary requirements of disease<br />

A disease state may set specific requirements for<br />

energy and nutrients, the deficiency of which may<br />

contribute to the deterioration of nutritional status<br />

and growth failure in children (Figure 32.2). The<br />

reduced energy availability may originate from<br />

three main sources: reduced food intake due to<br />

poor appetite or symptoms of disease such as<br />

gastrointestinal complaints or poor utilization of


nutrients; increased losses, e.g. due to steatorrhea;<br />

or increased requirements, e.g. due to infections.<br />

On the other hand, physical activity may be<br />

reduced, owing to ill health, and compensates for<br />

increased energy requirements. Although<br />

cause–consequence relationships are difficult to<br />

prove, such events may result in exacerbation of<br />

the disease or in reduced survival. A careful monitoring<br />

of diet and nutritional status and intervention<br />

at an early stage are vital for the prevention, or<br />

halting the development, of this vicious cycle with<br />

possibly deleterious effects.<br />

Nutritional intervention includes provision of<br />

sufficient amounts of energy and nutrients, by<br />

increasing the amount of food eaten or the nutrient<br />

density of the diet, or by using clinical nutritional<br />

products to enhance the nutrient density, or by<br />

providing an additional source of energy, for<br />

example as snacks. In a range of diseases with<br />

known nutritional management, the specific products,<br />

such as hydrolyzed infant formulas for cow’s<br />

milk allergy, 12 low-protein and special amino acid<br />

products for phenylketonuria 13 or gluten-free<br />

products for celiac disease 14 are critical in the<br />

management of these patients. The next step might<br />

be the dietary management and risk reduction of<br />

specific diseases by means of diet with added or<br />

Intake,<br />

utilization<br />

Losses<br />

Energy deficit<br />

Weight loss, growth<br />

failure<br />

Deterioration of end-organ function<br />

e.g. gastrointestinal tract,<br />

immune defense<br />

Needs<br />

Figure 32.2 Possible causes for deterioration of<br />

nutritional status and growth failure in children.<br />

The science of nutrition: from basic needs to specific health effects 527<br />

modified nutrients or compounds, i.e. nutraceuticals.<br />

Food consumption and risk of chronic diseases<br />

The diet in Western societies has faced changes<br />

that reflect industrialization, urbanization,<br />

economic development and market globalization<br />

15 . The availability of a more diverse selection<br />

of foods has improved the nutritional status of<br />

populations. However, not all the changes in food<br />

consumption have led to beneficial effects with<br />

regard to health status. The impacts of changes in<br />

the diet are further enforced by alterations in<br />

lifestyle such as decreased physical activity and<br />

increased tobacco use. In a recent report by expert<br />

panels in WHO and FAO, the global and regional<br />

food consumption patterns and estimated trends<br />

from 1960 to 2030 and their impact upon the<br />

prevalence of chronic, non-communicable,<br />

diseases were discussed. 15 According to the report,<br />

it may be possible to reduce the risk of cardiovascular<br />

diseases, cancer, diabetes and obesity by<br />

changing dietary habits. Particularly important<br />

targets contributing to deaths from chronic<br />

diseases are obesity, high blood pressure, high<br />

cholesterol concentrations and low levels of physical<br />

activity. According to the WHO/FAO expert<br />

panel, the key changes in food consumption in<br />

recent decades and also in the next 15–30 years,<br />

are increased energy intake and increased<br />

consumption of fat and added sugar, greater saturated<br />

fat intake and reduced intakes of complex<br />

carbohydrates and fiber. 15 Intakes of fruits and<br />

vegetables have increased in some areas, whilst<br />

the recommended high average intakes have been<br />

reached only in a minority of the world’s population.<br />

15<br />

In light of the link between early nutrition with<br />

later consequences, the importance of pediatric<br />

nutrition is further emphasized. Furthermore,<br />

dietary habits are established at an early age, by<br />

the number and type of experiences with various<br />

foods and also by model learning from parents,<br />

friends, day-care centers and schools. 16<br />

A population study comparing data on serum<br />

cholesterol, anthropometric indices and dietary<br />

intakes in children aged 1–18 years of age from<br />

Japan, Spain and the USA showed the effects of


528<br />

Biotherapeutic and nutraceutical agents<br />

Westernization on blood cholesterol concentration.<br />

17 Both in Japan and in Spain the intake of<br />

total and saturated fat had increased substantially.<br />

In studies from Northern Europe, changes in food<br />

consumption and consequently in nutrient intakes<br />

towards the recommended intakes seemed to<br />

explain the serum cholesterol concentration which<br />

was reflected in a decline in coronary heart disease<br />

mortality. 18 It has also been shown that the unfavorable<br />

dietary habits, reflected in high intake of<br />

fat and saturated fatty acids contributing to the<br />

risk of coronary heart disease, may be adopted<br />

already in childhood. 19<br />

Cardiovascular diseases are a good example of<br />

diseases in which associations between food<br />

consumption, nutrient intake, biological markers<br />

of disease (serum cholesterol concentration) and<br />

risk of disease have been documented. It has also<br />

been shown that dietary interventions modifying<br />

food intake in adults 18 or in children 20 to resemble<br />

that recommended for healthy eating, result in<br />

beneficial changes in anthropometry and serum<br />

cholesterol concentration that are reflected in<br />

reduced risk of chronic Western diseases.<br />

Although fat intake in accordance with the recommended<br />

intake has been shown to be safe in terms<br />

of growth and neurological development, 21,22<br />

concerns have been raised over low fat intake<br />

(


long-chain polyunsaturated fatty acids, oligosaccharides<br />

and nucleotides have since been used in<br />

immunonutrition beyond formula-age. Even the<br />

pre-milk provided by the cow after delivery –<br />

bovine colostrum – has been suggested to be used<br />

in humans for modulation of the immune system,<br />

as reviewed by Solomons. 33<br />

At weaning, the infant gradually transfers to adulttype<br />

feeding. The transition period may be significant<br />

in terms of adopting dietary habits. Intakes of<br />

some nutrients, particularly vitamin E and zinc,<br />

may be low during dietary transition in early<br />

childhood and may actually decrease, despite<br />

increases in energy intake. 34<br />

The integrity of the gut barrier function –<br />

the target of protective nutrients<br />

The primary role of the gastrointestinal tract is<br />

digestion and absorption of nutrients to meet the<br />

metabolic requirements and the demands of<br />

normal growth and development. In addition to<br />

this, the intestinal mucosa provides a protective<br />

host defense against the constant presence in the<br />

gut lumen of antigens from food and microorganisms.<br />

An abrupt change in gut barrier function<br />

occurs at birth, when it is switched from<br />

processing amniotic fluid to digesting milk and<br />

when the intestinal colonization commences.<br />

Gut barrier<br />

Protection against potentially harmful agents is<br />

ensured by a number of factors, including saliva,<br />

gastric acid, peristalsis, mucus, intestinal proteolysis,<br />

intestinal microbiota and epithelial cell<br />

membranes with intercellular junctional<br />

complexes. 35 The surface of mucosal membranes<br />

is protected by a local adaptive immune system.<br />

The gut-associated lymphoid tissue represents the<br />

largest mass of lymphoid tissue in the human<br />

body. Consequently, it comprises an important<br />

element of the total immunological capacity of the<br />

host. The regulatory events of the intestinal<br />

immune response take place in different compartments:<br />

aggregated in follicles and Peyer’s patches,<br />

distributed within the mucosa and in the intestinal<br />

epithelium, as well as in secretory sites (reviewed<br />

The integrity of the gut barrier function 529<br />

in reference 24). IgA antibody production is abundant<br />

at mucosal surfaces. In contrast to IgA in<br />

serum, secretory IgA is present in dimeric or polymeric<br />

form. Secretory IgA is resistant to intraluminal<br />

proteolysis and does not activate inflammatory<br />

responses, which makes secretory IgA ideal for<br />

protecting mucosal surfaces. These elements in the<br />

gut are part of the common mucosal immune<br />

system, including the respiratory tract and<br />

lacrimal, salivary and mammary glands, and<br />

thereby an immune response initiated in the gutassociated<br />

lymphoid tissue can affect immune<br />

responses at other mucosal surfaces.<br />

There are specialized antigen transport mechanisms<br />

in the villous epithelium and particularly in<br />

Peyer’s patches, which are crucial in determining<br />

the subsequent immune responses to the antigen.<br />

35 In addition to the first line of gut defense –<br />

immune exclusion – there are specialized antigen<br />

transport mechanisms in the villous epithelium.<br />

Antigens are absorbed across the epithelial layer<br />

by transcytosis, and here the main degradative<br />

pathway entails lysosomal processing of the<br />

antigen. 36,37 A minor pathway allows the transport<br />

of unprocessed antigens. Peyer’s patches are<br />

covered by a unique epithelium. Antigen transport<br />

across this epithelium is characterized by rapid<br />

uptake and reduced degradation. In health, paracellular<br />

transfer of macromolecules is controlled<br />

by intact intercellular tight junctions, which<br />

prevent aberrant antigen absorption. The second<br />

line of host defense – immune elimination – is<br />

directed towards antigenic compounds penetrating<br />

the mucosa. Antigens are presented to subjacent<br />

T cells; these differentiate into various effector<br />

cells that mediate active immune regulation<br />

and promote the differentiation of IgAsecreting<br />

B cells. 38<br />

Initial signals for the maturation of the gut barrier<br />

functions are considered to stem from components<br />

of the innate immunity, which generates the necessary<br />

initial step for the targeted and specific function<br />

of the adaptive immune system.<br />

Gut microbiota<br />

The human gastrointestinal tract harbors a<br />

complex collection of micro-organisms, a specific<br />

microbiota for each person. 39 The generation of the


530<br />

Biotherapeutic and nutraceutical agents<br />

immunophysical regulation in the gut depends on<br />

the establishment of the indigenous microbiota.<br />

At birth the gastrointestinal tract of the newborn is<br />

sterile. The maternal intestinal microbiota is the<br />

first source of colonizing bacteria. Subsequently,<br />

the feeding practice and the home environment of<br />

the child influence the composition. Breast<br />

feeding encourages the growth of bifidobacteria,<br />

while formula-fed infants have a more complex<br />

microbiota with bifidobacteria, enterobacteria,<br />

lactobacilli, bacteroides, clostridia and streptococci.<br />

40 New molecular methods indicate that bifidobacteria<br />

can reach up to 90% of the total fecal<br />

microbiota in breast-fed infants. After weaning,<br />

the composition of the microbiota resembles that<br />

of the adult. Although bacteria are distributed<br />

throughout the intestine, the major concentration<br />

of microbes and metabolic activity can be found in<br />

the large intestine. From culture-based data, it is<br />

thought that at least 500 different microbial<br />

species exist, although on a quantitative basis<br />

10–20 genera probably predominate: Bacteroides,<br />

Lactobacillus, Clostridium, Fusobacterium, Bifidobacterium,<br />

Eubacterium, Peptococcus, Peptostreptococcus,<br />

Escherichia and Veillonella. 39<br />

The composition of the microbiota is associated<br />

with several disease states within the intestine and<br />

also beyond the gastrointestinal tract. Inflammation<br />

is accompanied by imbalance in the<br />

intestinal microbiota in such a way that the<br />

host–microbe interaction is disturbed and an<br />

immune response may be induced by resident<br />

bacteria. 41,42<br />

Modification of intestinal microbiota to increase<br />

the predominance of specific non-pathogenic<br />

bacteria and thereby to alter the intestinal milieu<br />

has been taken as an alternative to attain prophylactic<br />

or therapeutic effects in intestinal infectious<br />

conditions in childhood. Recent clinical and nutritional<br />

studies and characterization of the<br />

immunomodulatory potential of specific strains of<br />

the gut microbiota, beyond the effect on the<br />

composition of the microbiota, may lead to future<br />

applications also for allergic and inflammatory<br />

diseases. Such probiotics, acting as nutraceutical<br />

agents, need their mechanisms to be thoroughly<br />

clarified, either to control specific physiological<br />

processes in the evolution of disease in populations<br />

at-risk or to manage of specific diseases.<br />

Protective nutrients for the gastrointestinal<br />

tract<br />

The integrity of the intestinal mucosa and the<br />

structure of the villi are crucial for assimilation of<br />

nutrients from the gastrointestinal tract and for<br />

intestinal defense against invading pathogens and<br />

antigens. This is obvious in diseases such as celiac<br />

disease, where the structure of the intestinal villi is<br />

disturbed by dietary gluten thereby affecting nutrient<br />

assimilation and the nutritional status of the<br />

patients if an appropriate diet is not followed.<br />

However, despite an apparently normal histological<br />

presentation of the mucosa, the functionality of<br />

the enterocytes may be altered. It has been shown<br />

in malnourished patients that, despite a normal<br />

villous structure of the small intestine, the intestinal<br />

permeability may be increased. 43 Reduced<br />

integrity of the mucosa could even result in an<br />

outward diffusion of nutrients towards the lumen<br />

which would further prevent the absorption of<br />

nutrients.<br />

Increased permeability of the intestinal mucosa<br />

may also result in increased susceptibility to infections.<br />

Infections may further cause deterioration of<br />

the intestinal integrity, owing to increased cell<br />

turnover, and therefore increase nutrient requirements.<br />

Turnover of cells in the gastrointestinal<br />

mucosa is rapid and its integrity mainly depends<br />

on the production of new cells at a rate equal to<br />

that at which cells are lost. 44 Therefore, even a<br />

short-term deficit in the nutrient supply to the<br />

mucosa may affect the mucosal integrity and result<br />

in villous atrophy. Nutrition through the gastrointestinal<br />

tract is important in maintaining the<br />

mucosal structure and function; lack of nutrients<br />

may result in decreased villous height, increased<br />

permeability and decreased immunity.<br />

Probably the single most important nutrient to the<br />

mucosa is glutamine (both from luminal and<br />

vascular sides), which is consumed by replicating<br />

cells and affects the structure and function of the<br />

cells. Glutamine may be required in increased<br />

amounts by patients suffering from a catabolic<br />

insult such as injury or severe infection. In<br />

parenteral nutrition, glutamine improves nitrogen<br />

balance, supports gut function and alleviates catabolic<br />

demands upon muscle mass. However,<br />

whether glutamine is necessary for the preservation<br />

of normal intestinal morphology and function


in humans during parenteral nutrition is not clear,<br />

as reviewed by Buchman. 45 In another systematic<br />

review, in enteral feeding of critically ill patients,<br />

the immune-enhancing nutrients including glutamine,<br />

arginine, neucleotides and n-3 fatty acids<br />

reduced the appearance of infectious complications,<br />

but not the overall mortality. 46<br />

Vitamin A is also important for the differentiation<br />

of cells and may have a central role in mucosal<br />

function. Deficiency impairs innate immunity by<br />

diminishing the function of neutrophils,<br />

macrophages and natural killer cells as well as<br />

antibody-mediated responses by T helper (Th)<br />

cells. 47 Vitamin A deficiency compromises<br />

mucosal epithelial barriers in the gastrointestinal<br />

tract, particularly when complicated by infection.<br />

Most data concerning the effects of nutrients on<br />

the structure and function of the epithelium are<br />

derived from experimental studies, but it has also<br />

been shown in children that recovering from diarrhea<br />

may be faster when vitamin A supplement is<br />

received compared to placebo. 48<br />

Probiotics and prebiotics<br />

Recent demonstration that the gut microbiota is an<br />

important constituent of the intestine’s mucosal<br />

barrier has introduced new therapeutic strategies<br />

for fighting enteric infections and possibly other<br />

intestinal inflammatory conditions (Figure 32.3).<br />

Three approaches for improving intestinal<br />

integrity include the use of probiotics, prebiotics<br />

and symbiotics. Probiotics are ‘a live microbial<br />

food ingredient that is beneficial to health’. The<br />

prerequisites for probiotic action include survival<br />

in and adhesion to specific areas of the gastrointestinal<br />

tract and competitive exclusion of<br />

pathogens or harmful antigens. The application of<br />

probiotics has since been supplemented with the<br />

concept of prebiotics. A prebiotic is a ‘nondigestible<br />

food ingredient that beneficially affects<br />

the host by selectively stimulating the growth<br />

and/or activity of one or a limited number of bacteria<br />

in the colon that have the potential to improve<br />

host health’. The most commonly used prebiotics<br />

are carbohydrate substrates with the ability to<br />

promote the components of the normal intestinal<br />

microbiota that may evince a health benefit to the<br />

host. However, prebiotics can also be nonabsorbable<br />

substrates which stimulate the growth<br />

Increased<br />

intestinal<br />

permeability<br />

The integrity of the gut barrier function 531<br />

Antigen<br />

degradation<br />

Antigen<br />

absorbtion<br />

route/rate<br />

Antigen<br />

Probiotic<br />

therapy<br />

Balance of pathogenic/<br />

beneficial strains<br />

Microbiota<br />

Antigen<br />

presentation<br />

Balance of<br />

pro-/antiinflammatory<br />

cytokines<br />

Figure 32.3 Potential targets for probiotic therapy<br />

(adapted from reference 42).<br />

Inflammatory<br />

response<br />

of probiotics. When the two are applied together,<br />

the concept is defined as symbiotic.<br />

Recent research has expanded the definition of<br />

probiotics, by demonstrating that genetically engineered<br />

microbes and non-viable microbes may<br />

equally possess such potential. 42 However,<br />

normalization of the properties of unbalanced<br />

indigenous microbiota by specific strains forms<br />

the rationale of probiotic therapy. Oral introduction<br />

of probiotics has been shown to reinforce the<br />

various lines of gut defense: immune exclusion,<br />

immune elimination and immune regulation.<br />

Probiotics also stimulate non-specific host resistance<br />

to microbial pathogens and thereby aid in<br />

their eradication.<br />

So far, the best-documented clinical application is<br />

the treatment of acute diarrhea by specific probiotic<br />

bacteria. 49 The beneficial clinical effect in<br />

infantile diarrhea by probiotic therapy has been<br />

explained by stabilization of the indigenous microbiota,<br />

reduction in the duration of rotavirus shedding<br />

and reduction in increased gut permeability<br />

caused by rotavirus infection, together with a<br />

significant increase in cells secreting IgA against<br />

rotavirus. The probiotic performance of strains is<br />

such that different probiotic strains, even strains<br />

belonging to the same species, have different<br />

immunomodulatory effects. Hence, current probiotic<br />

research is directed towards identification of<br />

specific strains with potential to reduce the risk of<br />

diseases associated with gut barrier dysfunction.


532<br />

Biotherapeutic and nutraceutical agents<br />

Modern nutrition for children –<br />

allergic disease<br />

Allergic diseases, manifesting as atopic eczema,<br />

allergic rhinitis and asthma, are on the increase in<br />

industrialized countries, currently constituting the<br />

most common chronic diseases of childhood.<br />

Despite the pronounced hereditary element in<br />

allergic disease, genetic factors are unlikely to<br />

explain the increased emergence of the atopic-type<br />

immune responsiveness to environmental antigens.<br />

In contrast, changes in nutrition along with<br />

general environmental changes appear to shape<br />

the immune responder type of the host during a<br />

critical period of life. Specifically, the changes<br />

associated with Western lifestyle tend to favor the<br />

atopic Th 2-biased immune responder type.<br />

The specific processes that initiate the vicious<br />

cycle of allergic inflammation, remain unresolved.<br />

Recent advances, however, call into question a<br />

causal cascade of exposure to antigenic proteins,<br />

sensitization and hypersensitivity; the exposure to<br />

allergens sensitizing the host immune system may<br />

not necessarily induce allergic disease. Indeed,<br />

sensitization to indoor allergens such as house<br />

dust mite, as assessed by specific IgE, but not the<br />

level of exposure to these allergens, has been associated<br />

with asthma, wheeze and increased<br />

bronchial responsiveness. 50 In like manner, exposure<br />

to cow’s milk antigen during 3 days after<br />

birth, in 1533 breast-fed neonates, did not carry a<br />

risk of atopic disease during the first 5 years of<br />

age. 51,52 These data would imply that eradication<br />

of potential allergenic proteins from the early environment,<br />

including the diet, may not apply in<br />

allergy-prevention strategies.<br />

In contrast, one justifiable strategy against allergic<br />

disease may be based on the administration of<br />

tolerogenic gut-processed peptide fragments of a<br />

specific protein, and the use of specific dietary<br />

compounds such as fatty acids and antioxidants,<br />

or on providing a microbial stimulus for the immature<br />

immune system by means of cultures of beneficial<br />

live micro-organisms characteristic of the<br />

healthy human gut microbiota. 53 However, before<br />

such strategies can be developed, a better understanding<br />

will be necessary of the processes initiating<br />

and regulating the allergic inflammatory<br />

response. Further, a better understanding of the<br />

interaction between nutrients is required.<br />

Consequently, the properties of specific dietary<br />

compounds and their combinations, in an optimal<br />

food matrix, might be exploited in the development<br />

of specific prophylactic and therapeutic<br />

interventions.<br />

Elimination diets<br />

Elimination of potentially allergenic foods, e.g.<br />

cow’s milk, egg, wheat and fish, from the pregnant<br />

or breast-feeding mother’s or the infant’s diet has<br />

been a common approach in attempts to prevent<br />

allergic disease in high-risk infants. The approach<br />

of eliminating foods from the diet is perplexing, for<br />

two reasons. First, the benefits of elimination diets<br />

in prevention of allergic disease have been inconclusive.<br />

54 Especially in long-term prevention of<br />

allergic disease, elimination diets have proved<br />

unsuccessful. Dietary antigens may actually be<br />

protective from allergic disease as they may induce<br />

tolerance to antigens rather than sensitization. 52,55<br />

Second, besides allergenic proteins in the diet,<br />

other dietary factors might be associated with the<br />

development of allergic disease.<br />

Recent studies on the immunomodulatory properties<br />

of fatty acids and the antioxidant properties of<br />

certain nutrients including ascorbic acid, αtocopherol,<br />

β-carotene, selenium and zinc, have<br />

shown that food is not only a source of dietary<br />

antigens causing sensitization, but may also<br />

contain protective factors. Thus, elimination diets,<br />

i.e. eliminating specific offending antigens, should<br />

be reserved only to the management of proven<br />

food allergy. A better approach in the prevention of<br />

allergic disease might be to search for protective<br />

nutrients in the diet that may reinforce the immature<br />

immune defense of the infant.<br />

Modification of allergenic proteins<br />

The nutritional theories attracting current research<br />

interest relate to identification of active<br />

compounds which reinforce an anti-inflammatory,<br />

potentially tolerogenic type of immune response to<br />

assimilate allergenic challenges. Factors that influence<br />

the allergenicity of a protein include molecular<br />

complexity, solubility and stability, and its<br />

concentration. Heat treatment of cow’s milk<br />

proteins can affect the conformational epitopes of


allergens and facilitate their hydrolysis. To<br />

produce the least allergenic formulas for patients<br />

with documented cow’s milk allergy, cow’s milk<br />

proteins are modified by multiple enzymatic<br />

hydrolyses with progressive destruction of sequential<br />

epitopes. Such products are classified as<br />

partially hydrolyzed or extensively hydrolyzed<br />

formulas. However, enzymatic hydrolysis does not<br />

render the formula non-allergenic, as the optimal<br />

extent of hydrolysis is not known and traces of the<br />

original protein are detected in the hydrolysate. 56<br />

The approach may also be used in an attempt to<br />

reduce the risk of allergic disease. 57 Degradation of<br />

antigens is a necessary initial step in controlling<br />

inflammatory responsiveness to dietary antigens.<br />

55,58,59 Such processing has been linked to the<br />

potential to generate peptides with suppressive<br />

effects on lymphocyte proliferation in healthy<br />

subjects; systemic immune responses to gutprocessed<br />

antigens are preferentially directed<br />

towards suppression. The mechanisms of the<br />

preventive potential of specifically modified<br />

dietary proteins remain still poorly understood.<br />

Dietary lipids<br />

Increasing evidence has been provided to suggest<br />

that dietary lipids, especially long-chain polyunsaturated<br />

fatty acids (PUFA), and mediators<br />

synthesized from PUFA, regulate immune function<br />

and therefore contribute to the development and<br />

severity of the symptoms of allergic disease.<br />

Polyunsaturated n-6-series fatty acids derived from<br />

dietary linoleic acid (18:2, n-6) result in the production<br />

of eicosanoids, which are considered to<br />

have proinflammatory properties, whilst n-3-series<br />

fatty acids derived from dietary α-linolenic acid<br />

(18:3, n-3) appear to have less potent biological<br />

functions, even anti-inflammatory properties. 60<br />

Owing to the typically higher dietary intake of<br />

linoleic acid than α-linolenic acid in developed<br />

countries, it is thought that the metabolism of<br />

linoleic acid predominates.<br />

The most notable of the PUFA-derived mediators is<br />

arachidonic acid (20:4, n-6)-derived eicosanoid<br />

prostaglandin (PG)E2. PGE2 results in elevated IgE<br />

synthesis, owing to the induction of B-cell differentiation<br />

in the presence of interleukin IL-4. 61<br />

Indeed, the most frequently reported abnormality<br />

Modern nutrition for children – allergic disease 533<br />

in cell fatty acid composition of atopic patients has<br />

been an imbalance between series n-6 and n-3<br />

fatty acids, 62–64 predisposing the patients to the<br />

adverse effects of PGE2. Nevertheless, whether the<br />

observed alterations in the fatty acid composition<br />

of cells in the patients with atopic disease result<br />

from a primary defect that contributes to the onset<br />

of atopic disease or are a consequence of the atopic<br />

disease itself is currently poorly understood. This<br />

is especially intriguing as, despite their apparent<br />

proinflammatory role, n-6 fatty acids may also<br />

contribute to an anti-inflammatory intestinal environment,<br />

as antigen stimulation up-regulates PGE2<br />

production from arachidonic acid with ensuing<br />

suppression of antigen-specific T-cell proliferation<br />

in gut-associated lymphoid tissue. 65 However, the<br />

effects on dietary n-6 fatty acids may be counteracted<br />

by n-3 series fatty acids derived from dietary<br />

α-linolenic acid (18:3, n-3) or directly from marine<br />

food sources (eicosapentaenoic acid; 20:5, n-3 and<br />

docosahexaenoic acid; 22:6, n-3). Eicosanoids<br />

derived from α-linolenic acid appear to have a less<br />

potent biological, even anti-inflammatory, function<br />

compared to n-6-series fatty acids. The antiinflammatory<br />

properties of n-3 fatty acids arise<br />

from their capacity to inhibit the release of arachidonic<br />

acid from membrane phospholipids, thereby<br />

reducing the production of arachidonic acidderived<br />

eicosanoids whilst the synthesis of n-3<br />

fatty acid-derived eicosanoids increases. 66 Owing<br />

to the likely interactions between the nutrients,<br />

the overall fatty acid composition and the quantity<br />

of fat within the diet with respect to other nutrients<br />

may be crucial in the search for the optimal<br />

diet for prevention and management of allergic<br />

disease.<br />

Dietary antioxidants<br />

In atopic disease, inflammatory processes result in<br />

endogenously generated oxidative stress which<br />

dietary antioxidants, such as ascorbic acid, βcarotene,<br />

α-tocopherol, selenium and zinc may<br />

counteract. 67 Both cellular enzyme-based antioxidants<br />

and diet-derived antioxidants counteract<br />

oxidative stress and dampen the inflammatory<br />

response. 67 Dietary antioxidants may thus be<br />

important in implementing the ability of the individual<br />

to restrain the inflammatory response and<br />

in avoiding injury to tissues. Low concentrations


534<br />

Biotherapeutic and nutraceutical agents<br />

of β-carotene, ascorbate and α-tocopherol have<br />

been measured in plasma in wheezing illness,<br />

suggesting that antioxidant deficiencies may be<br />

associated with symptoms of allergic disease. 68<br />

However, at present the role of antioxidants in the<br />

onset of allergic disease remains obscure.<br />

Moreover, data accumulated to date on the specific<br />

antioxidant agents and their dose and mechanisms<br />

are insufficient to provide any recommendation on<br />

their use, either in the prevention or in the<br />

management of allergic disease.<br />

Probiotics in allergic diseases<br />

Probiotics represent one example of immunomodulatory<br />

compounds for the allergic host. The<br />

immunomodulatory potential of the healthy<br />

gastrointestinal microbiota, the source of specific<br />

probiotic strains, is possibly associated with two<br />

structural components of bacteria, the lipopolysaccharide<br />

portion of Gram-negative bacteria (endotoxin)<br />

and a specified CpG motif in bacterial<br />

DNA. 69,70 These structures activate immunomodulatory<br />

genes via Toll-like receptors present, for<br />

example on macrophages, and dendritic and<br />

intestinal epithelial cells. The net effect on intestinal<br />

epithelial cells is immunosuppressive by way<br />

of inhibition of the transcription factor NF-κB<br />

pathway. 71 In addition, specific strains of the gut<br />

microbiota have been shown to contribute to a Th<br />

cell population promoting oral tolerance induction,<br />

and to counter allergy by generation of antiinflammatory<br />

IL-10 and transforming growth<br />

factor (TGF)-β (reviewed in reference 41). The<br />

importance of the immunoregulatory potential of<br />

the gut microbiota is emphasized in the recent<br />

demonstration of cross-talk between the innate<br />

and the adaptive immune system; the nature of the<br />

initial immune response governs the homeostasis<br />

of the adaptive immune response.<br />

The compositional development of the gut microbiota<br />

differs between infants developing and those<br />

not developing atopic manifestations. Healthy<br />

infants harbor a natural predominance of bifidobacteria<br />

with specific strains present, while<br />

those later manifesting atopy present with a<br />

reduced ratio of bifidobacteria to clostridia 72 and<br />

with distinct bifidobacterial microbiota. 73 Further,<br />

intestinal Bifidobacterium species from allergic<br />

infants induce proinflammatory cytokine produc-<br />

tion, in contrast to those from healthy infants. 74<br />

Thus, the antiallergic potential of Bifidobacterium<br />

biota may be strain-specific.<br />

Probiotics may aid in degradation/structural modification<br />

of enteral antigens, normalization of the<br />

properties of aberrant indigenous microbiota and<br />

of gut barrier functions, regulation of the secretion<br />

of inflammatory mediators and direction of the<br />

development of the immune system during the<br />

critical period of life when the risk of allergic<br />

disease is heightened.<br />

Preliminary studies have revealed that cow’s milk<br />

casein, a common allergen in cow’s milk allergy,<br />

hydrolyzed with probiotic-derived enzymes<br />

suppresses lymphocyte proliferation and, more<br />

specifically, production of allergen-specific Th2<br />

cytokins. 58,75 In experiments conducted by von der<br />

Weid et al, 59 probiotics inhibited proliferation of T<br />

cells and reduced secretion of both Th1 and Th2<br />

cytokines whilst inducing the development of a<br />

population of T cells producing TGF-β and IL-10,<br />

reminiscent of tolerogenic Th3 cells. Furthermore,<br />

a specific probiotic strain has been demonstrated<br />

to suppress IgE responses and systemic anaphylaxis<br />

in a murine model of food allergy. 76<br />

Different probiotic strains appear to induce<br />

distinct and even opposing responses in murine<br />

dendritic cells and thus specific strains of the gut<br />

microbiota and probiotics may play a crucial role<br />

in determining the Th1/Th2/Th3-driving capacity<br />

of intestinal dendritic cells. 42 So far, clinical<br />

effects have been seen as a significant improvement<br />

in the clinical course of atopic eczema in<br />

infants given probiotic-supplemented elimination<br />

diets. The preventive potential of probiotics in<br />

atopic disease has been demonstrated in a doubleblind,<br />

placebo-controlled study. 77 Probiotics<br />

administered pre- and postnatally for 6 months to<br />

children at high risk of atopic diseases succeeded<br />

in reducing the prevalence of atopic eczema by<br />

half, as compared with that in infants receiving<br />

placebo, and the effect was recently shown to<br />

extend beyond infancy. 78<br />

The challenge in terms of prevention and management<br />

of allergic disease is to identify and clarify<br />

the mechanisms of action of the dietary factors<br />

that may be protective. Well-controlled intervention<br />

studies are required to address the potential<br />

effects of different dietary modifications and


supplementation to prevent and treat allergic<br />

disease or other chronic, mostly immunoinflammatory,<br />

diseases. In future, the most likely option<br />

may be the incorporation of probiotics and specific<br />

nutrients into the same products (nutraceuticals)<br />

to provide an optimal diet for individuals at risk.<br />

Unquestionably, the interactions between probiotics<br />

and nutrients need to be studied. However,<br />

nutraceuticals alone cannot resolve the challenge<br />

of atopic disease if the crucial role of the total<br />

composition of the overall diet is neglected.<br />

REFERENCES<br />

1. World Health Organization. Diet, Nutrition and the<br />

Prevention of Chronic Diseases, a report of a WHO study<br />

group. Technical report series no. 797. Geneva: World<br />

Health Organization, 1990.<br />

2. Food and Agriculture Organization. Fats and Oils in<br />

Human Nutrition. FAO food and nutrition paper no 57.<br />

Rome: Food and Agriculture Organization of the United<br />

Nations, 1993.<br />

3. World Health Organization. Trace Elements in Human<br />

Nutrition and Health. Geneva: World Health Organization,<br />

1996.<br />

4. King JC. Physiology of pregnancy and nutrient metabolism.<br />

Am J Clin Nutr 2000; 71: 1218–1225.<br />

5. Scholl TO, Johnson WG. Folic acid: influence on the<br />

outcome of pregnancy. Am J Clin Nutr 2000; 71:<br />

S1295–S1303.<br />

6. Napoli C, Glass CK, Witztum JL et al. Influence of<br />

maternal hypercholesterolaemia during pregnancy on<br />

progression of early atherosclerotic lesions in childhood:<br />

Fate of Early Lesions in Children (FELIC) study.<br />

Lancet 1999; 354: 1234–1241.<br />

7. Forsdahl A. Are poor living conditions in childhood and<br />

adolescence an important risk factor for arteriosclerotic<br />

heart disease? Br J Prev Soc Med 1977; 31: 91-95.<br />

8. Barker DJP, Osmond C. Early growth and death from<br />

cardiovascular disease in women. Br Med J 1993; 1:<br />

1077–1081.<br />

9. Osmond C, Barker DJP, Winter PD et al. Early growth<br />

and death from cardiovascular disease in women. Br<br />

Med J 1993; 307: 519–524.<br />

10. Langley SC, Jackson AA. Increased systolic blood pressure<br />

in adult rats induced by fetal exposure to maternal<br />

low protein diets. Clin Sci 1994; 86: 217–222.<br />

11. Desai M, Crowther N, Ozanne SE et al. Adult glucose<br />

and lipid metabolism may be programmed during fetal<br />

life. Biochem Soc Trans 1995; 23: 331–335.<br />

12. Isolauri E, Sütas Y, Mäkinen-Kiljunen S et al. Efficacy<br />

and safety of hydrolyzed cow milk and amino acidderived<br />

formulas in infants with cow milk allergy. J<br />

Pediatr 1995; 127: 550–557.<br />

13. Smith I, Beasley MG, Ades AE. Intelligence and quality<br />

of dietary treatment in phenylketonuria. Arch Dis Child<br />

1990; 65: 472–478.<br />

References 535<br />

Taken together, the direction of research should be<br />

focused towards searching for such dietary<br />

compounds that may have properties beyond the<br />

traditional nutritional effects, i.e. health promotion<br />

and risk reduction of a range of diseases. The<br />

research in allergic disease is achieving this extention<br />

by exploring novel prevention and management<br />

strategies. Consequently, research into allergic<br />

disease can be taken as a model for the<br />

development of nutritional management for other<br />

chronic immunoinflammatory diseases as well.<br />

14. Sategna-Guidetti C, Grosso SB, Grosso S et al. The<br />

effects of 1-year gluten withdrawal on bone mass, bone<br />

metabolism and nutritional status in newly-diagnosed<br />

adult coeliac disease patients. Aliment Pharmacol Ther<br />

2000; 14: 35–43.<br />

15. World Health Organization. Diet, Nutrition and the<br />

Prevention of Chronic Diseases, a report of a joint<br />

WHO/FAO expert consultation, 28 January–1 February<br />

2002. WHO technical report series, 916. Geneva: World<br />

Health Organization, 2003.<br />

16. Birch LL. Development of food preferences. Annu Rev<br />

Nutr 1999; 19: 41–62.<br />

17. Couch SC, Cross AT, Kida K et al. Rapid westernization<br />

of children's blood cholesterol in 3 countries: evidence<br />

for nutrient–gene interactions? Am J Clin Nutr 2000; 72:<br />

1266–1274.<br />

18. Pietinen P, Vartiainen E, Seppänen R et al. Changes in<br />

diet in Finland from 1972 to 1992: impact on coronary<br />

heart disease risk. Prev Med 1996; 25: 243–250.<br />

19. Räsänen L, Ahola M, Kara R et al. Atherosclerosis<br />

precursors in Finnish children and adolescents. VIII.<br />

Food consumption and nutrient intakes. Acta Paediatr<br />

Scand Suppl 1985; 318: 135–153.<br />

20. Simell O, Niinikoski H, Rönnemaa T et al. Special<br />

Turku coronary risk factor intervention project for<br />

babies (STRIP). Am J Clin Nutr 2000; 72: 1316–1331.<br />

21. Lagström H, Seppänen R, Jokinen E et al. Influence of<br />

dietary fat on the nutrient intake and growth of children<br />

from 1 to 5 years of age: the Special Turku Coronary<br />

Risk Factor Intervention Project. Am J Clin Nutr 1999;<br />

69: 516–523.<br />

22. Rask-Nissilä L, Jokinen E, Terho P et al. Effects of diet<br />

on the neurologic development of children at 5 years of<br />

age: the STRIP project. J Pediatr 2002; 140: 328–333.<br />

23. Uauy R, Mize CE, Castillo-Duran C. Fat intake during<br />

childhood: metabolic responses and effects on growth.<br />

Am J Clin Nutr 2000; 72: 1354–1360.<br />

24. Brandtzaeg P. Development of the mucosal immune<br />

system in humans. In Bindels JG, Goedhart AC, Visser<br />

HKA, eds. Recent Developments in Infant Nutrition.<br />

London: Kluwer Academic Publishers, 1996; 349–376.<br />

25. Gdalevich M, Mimouni D, David M et al. Breast-feeding<br />

and the onset of atopic dermatitis in childhood: a


536<br />

Biotherapeutic and nutraceutical agents<br />

systematic review and meta-analysis of prospective<br />

studies. J Am Acad Dermatol 2001; 45: 520–527.<br />

26. Gdalevich M, Mimouni D, Mimouni M. Breast-feeding<br />

and the risk of bronchial asthma in childhood: a<br />

systematic review with meta-analysis of prospective<br />

studies. J Pediatr 2001; 139: 261–266.<br />

27. Gillman MW, Rifas-Shiman SL, Camargo CAJ et al. Risk<br />

of overweight among adolescents who were breastfed as<br />

infants. JAMA 2001; 16: 2461–2467.<br />

28. von Kries R, Koletzko B, Sauerwald T et al. Does breastfeeding<br />

protect against childhood obesity? Adv Exp Med<br />

Biol 2000; 478: 29–39.<br />

29. Davis MK. Breastfeeding and chronic disease in childhood<br />

and adolescence. Pediatr Clin North Am 2001; 48:<br />

125–141.<br />

30. Michaelsen KM, Skafte L, Badsberg JH et al. Variation in<br />

macronutrients in human bank milk: influencing factors<br />

and implications for human milk banking. J Pediatr<br />

Gastroenterol Nutr 1990; 11: 229–239.<br />

31. Laiho K, Lampi A-M, Moilanen E et al. Breast milk fatty<br />

acids, eicosanoids and cytokines in mothers with and<br />

without allergic disease. Pediatr Res 2003; 53: 642–647.<br />

32. Rautava S, Kalliomäki M, Isolauri E. Probiotics during<br />

pregnancy and breastfeeding may confer immunomodulatory<br />

protection against atopic disease in the infant. J<br />

Allergy Clin Immunol 2002; 109: 119–121.<br />

33. Solomons NW. Modulation of the immune system and<br />

the response against pathogens with bovine colostrum<br />

concentrates. Eur J Clin Nutr 2002; 56: S24–S28.<br />

34. Picciano MF, Smiciklas-Wright H, Birch LL et al.<br />

Nutritional guidance is needed during dietary transition<br />

in early childhood. Pediatr 2000; 106: 109–114.<br />

35. Sanderson IR, Walker WA. Uptake and transport of<br />

macromolecules by the intestine: possible role in clinical<br />

disorders (an update). Gastroenterology 1993; 104:<br />

622–639.<br />

36. Ducroc R, Heyman M, Beaufrere B et al. Horseradish<br />

peroxidase transport across rabbit jejunum and Peyer’s<br />

patches in vitro. Am J Physiol 1983; 245: G54–G58.<br />

37. Isolauri E, Majamaa H, Arvola T et al. Lactobacillus<br />

casei strain GG reverses increased intestinal permeability<br />

induced by cow milk in suckling rats.<br />

Gastroenterology 1993; 105: 1643–1650.<br />

38. Brandtzaeg P. Development and basic mechanisms of<br />

human gut immunity. Nutr Rev 1998; 56: S5–S18.<br />

39. Salminen S, Bouley C, Boutron-Ruault M-C et al.<br />

Functional food science and gastrointestinal physiology<br />

and function. Br J Nutr 1998; 80: 47–71.<br />

40. Favier C, Vaughan E, de Vos W et al. Molecular monitoring<br />

of succession of bacterial communities in human<br />

neonates. Appl Environ Microbiol 2002; 68: 219–226.<br />

41. Isolauri E, Rautava S, Kalliomäki M et al. Role of probiotics<br />

in food hypersensitivity, Curr Opin Allergy Clin<br />

Immunol 2002; 2: 263–271.<br />

42. Isolauri E, Kirjavainen PV, Salminen S. Probiotics – a<br />

role in the treatment of intestinal infection and inflammation?<br />

Gut 2002; 50 (Suppl iii): 54–59.<br />

43. Welsh FKS, Farmery SM, MacLennan K et al. Gut<br />

barrier function in malnourished patients. Gut 1998; 42:<br />

396–401.<br />

44. Mathers JC. Nutrient regulation of intestinal proliferation<br />

and apoptosis. Proc Nutr Soc 1998; 57: 219–223.<br />

45. Buchman AL. Glutamine: commercially essential or<br />

conditionally essential? A critical appraisal of the<br />

human data. Am J Clin Nutr 2001; 74: 25–32.<br />

46. Heyland DK, Novak F, Drover JW et al. Should<br />

immunonutrition become routine in critically ill<br />

patients? A systematic review of the evidence. J Am Med<br />

Assoc 2001; 286: 944–953.<br />

47. Stephensen CB. Vitamin A, infection, and immune<br />

function. Annu Rev Nutr 2001; 21: 167–192.<br />

48. Thurnham DI, Northrop-Clewes CA, McCullough FS et<br />

al. Innate immunity, gut integrity, and vitamin A in<br />

Gambian and Indian infants. J Infec Dis 2000; 182:<br />

S23–2S8.<br />

49. Isolauri E. Probiotics for infectious diarrhoea. Gut 2003;<br />

52: 436–437.<br />

50. Lau S, Illi S, Sommerfeld C et al. Early exposure to<br />

house-dust mite and cat allergens and development of<br />

childhood asthma: a cohort study. Lancet 2000; 356:<br />

1392–1397.<br />

51. de Jong MH, Scharp-van der Linden VTM, Aalberse RC<br />

et al. Randomised controlled trial of brief neonatal<br />

exposure to cows' milk on the development of atopy.<br />

Arch Dis Child 1998; 79: 126–130.<br />

52. de Jong MH, Scharp-van der Linden VETM, Aalberse<br />

RC et al. The effect of brief neonatal exposure to cows'<br />

milk on atopic symptoms up to age 5. Arch Dis Child<br />

2002; 86: 365–369.<br />

53. Laiho K, Hoppu U, Ouwehand A et al. Probiotics–ongoing<br />

research on atopic individuals. Br J Nutr 2002; 88:<br />

S19–S27.<br />

54. Zeiger RS. Dietary manipulations in infants and their<br />

mothers and the natural course of atopic disease.<br />

Pediatr Allergy Immunol 1994; 5: 33–43.<br />

55. Barone KS, Reilly MR, Flanangan MP et al. Abrogation<br />

of oral tolerance by feeding encapsulated antigen. Cell<br />

Immunol 2000; 199: 65–72.<br />

56. Mäkinen-Kiljunen S, Sorva R. Bovine beta-lactoglobulin<br />

levels in hydrolysed protein formulas for infant feeding.<br />

Clin Exp Allergy 1993; 23: 287–291.<br />

57. Von Berg A, Koletzko S, Grubl A. The effect of<br />

hydrolyzed cow’s milk formula for allergy prevention in<br />

the first year of life: the German Infant Nutritional<br />

Intervention Study, a randomized double-blind trial. J<br />

Allergy Clin Immunol 2003; 111: 533–540.<br />

58. Sütas Y, Soppi E, Korhonen H. Suppression of lymphocyte<br />

proliferation in vitro by bovine caseins hydrolysed<br />

with Lactobacillus GG-derived enzymes. J Allergy Clin<br />

Immunol 1996; 98: 216–224.<br />

59. von der Weid T, Bulliard C, Schiffrin EJ. Induction by a<br />

lactic acid bacterium of a population of CD4+ T cells<br />

with low proliferative capacity that produce transforming<br />

growth factor beta and interleukin-10. Clin Diagn<br />

Lab Immunol 2001; 8: 695–701.<br />

60. Sellmayer A, Koletzko B. Long-chain polyunsaturated<br />

fatty acids and eicosanoids in infants–physiological and<br />

pathophysiological aspects and open questions. Lipids<br />

1999; 34: 199–205.<br />

61. Roper RL, Brown DM, Phipps P. Prostaglandin E2<br />

promotes B lymphocyte Ig isotype switching to IgE. J<br />

Immunol 1995; 154: 162–170.<br />

62. Biagi PL, Hrelia S, Celadon M et al. Erythrocyte<br />

membrane fatty acid composition in children with<br />

atopic dermatitis compared to age-matched controls.<br />

Acta Paediatr 1993; 82: 789–790.<br />

63. Leichsenring M, Kochsiek U, Paul K. (n-6)-Fatty acids in<br />

plasma lipids of children with atopic bronchial asthma.<br />

Pediatr Allergy Immunol 1995; 6: 209–212.<br />

64. Yu G, Duchén K, Björkstén B. Fatty acid composition in<br />

colostrum and mature milk from non-atopic and atopic<br />

mothers during the first 6 months of lactation. Acta<br />

Paediatr 1998; 87: 729–736.<br />

65. Newberry RD, Stenson WF, Lorenz RG. Cyclooxygenase-<br />

2-dependent arachidonic acid metabolites are essential<br />

modulators of the intestinal immune response to dietary<br />

antigen. Nature Med 1999; 5: 900–906.


66. Whelan J. Antagonistic effects of dietary arachidonic<br />

acid and n-3 polyunsaturated fatty acids. J Nutr 1996;<br />

126: 1086–1091.<br />

67. Greene LS. Asthma, oxidant stress, and diet. Nutrition<br />

1999; 15: 899–907.<br />

68. Bodner C, Godden D, Brown K et al. Antioxidant intake<br />

and adult-onset wheeze: a case–control study. Eur<br />

Respir J 1999; 13: 22–30.<br />

69. Hartmann G, Weiner GJ, Krieg AM. CpG DNA: a potent<br />

signal for growth, activation, and maturation of human<br />

dendritic cells. Proc Natl Sci USA 1999; 96: 9305–9319.<br />

70. Kranzer K, Bauer M, Lipford GB et al. CpGoligodeoxynucleotides<br />

enhance T-cell receptor-triggered<br />

interferon-gamma production and up-regulation of<br />

CD69 via induction of antigen-presenting cell-derived<br />

interferon type I and interleukin-12. Immunology 2000;<br />

99: 170–178.<br />

71. Neish AS, Gewirtz AT, Zeng H et al. Prokaryotic regulation<br />

of epithelial responses by inhibition of IkappaBalpha<br />

ubiquitination. Science 2000; 289: 1560–1563.<br />

72. Kalliomäki M, Kirjavainen P, Eerola E et al. Distinct<br />

patterns of neonatal gut microflora in infants in whom<br />

atopy was and was not developing. J Allergy Clin<br />

Immunol 2001; 107: 129–134.<br />

References 537<br />

73. Ouwehand AC, Isolauri E, He F et al. Differences in<br />

Bifidobacterium flora composition in allergic and<br />

healthy infants. J Allergy Clin Immunol 2001; 108:<br />

144–145.<br />

74. He F, Morita H, Hashimoto H. Intestinal Bifidobacterium<br />

species induce varying cytokine production. J Allergy<br />

Clin Immunol 2002; 109: 1035–1036.<br />

75. Pochard P, Gosset P, Grangette C. Lactic acid bacteria<br />

inhibit TH2 cytokine production by mononuclear cells<br />

from allergic patients. J Allergy Clin Immunol 2002; 110:<br />

617–623.<br />

76. Shida K, Takahashi R, Iwadate E. Lactobacillus casei<br />

strain Shirota suppresses serum immunoglobulin E and<br />

immunoglobulin G1 responses and systemic anaphylaxis<br />

in a food allergy model. Clin Exp Allergy 2002; 32:<br />

563–570.<br />

77. Kalliomäki M, Salminen S, Arvilommi H et al.<br />

Probiotics in primary prevention of atopic disease: a<br />

randomised, placebo-controlled trial. Lancet 2001; 357:<br />

1076–1079.<br />

78. Kalliomäki M, Salminen S, Poussa T et al. Probiotics<br />

and prevention of atopic disease – a 4-year follow-up of<br />

a randomised placebo-controlled trial. Lancet 2003; 361:<br />

1869–1871.


33<br />

Introduction<br />

Enteral nutrition<br />

Olivier Goulet and Virginie Colomb<br />

Enteral nutrition (EN) or enteral feeding is a technique<br />

for nutritional support which delivers a<br />

homogeneous, liquid nutritional admixture into<br />

the digestive tract by tube, into the stomach, or,<br />

more rarely in children, into the duodenum or the<br />

proximal jejunum. EN, has been used in pediatric<br />

patients for more than 30 years, in order to preserve<br />

nutritional status and normal growth, or to treat<br />

malnutrition when oral feedings cannot fill the<br />

protein–energy demand. EN is more physiological,<br />

usually safer, easier to administrate and less expensive<br />

than parenteral nutrition (PN). Therefore, EN<br />

should be preferred to PN in infants and children<br />

with malnutrition and/or nutritional risk, 1 when<br />

the intestinal tract is usable to provide nutrients.<br />

The physiological basis of continuous EN make it of<br />

great interest in pediatric patients with gastrointestinal<br />

(GI) disorders. 2,3 Nevertheless, this therapy<br />

is now widely used in a variety of extradigestive<br />

conditions.<br />

EN, like PN, is always initiated in the hospital, but<br />

may be performed at home in case of chronic<br />

disease, leading to long-term dependency. Home<br />

EN has enlarged the field of indications and<br />

improved the psychological tolerance to long-term<br />

nutritional therapy.<br />

Physiological basis of continuous<br />

enteral feeding<br />

Gastrointestinal motility<br />

In case of gastric administration of continuous<br />

enteral nutrition (CEN), a continuous gastric<br />

emptying related to infusion rate can be achieved,<br />

if the infusion rate, the caloric load and the osmolarity<br />

of the mixture are not excessive. When the<br />

infusion rate is below 3kcal/min, the gastric emptying<br />

rate increases with increasing caloric load, up<br />

to the same level as that of the infusion rate. 4 Thus,<br />

a steady state is achieved between the amount of<br />

nutrients delivered into the stomach, the gastric<br />

secretion volume and the gastric emptying rate.<br />

When the infusion rate is excessive and higher than<br />

the gastric emptying rate, the risk of vomiting<br />

increases. As the caloric load and/or osmolarity of<br />

the formula increase, the gastric emptying rate is<br />

reduced, to maintain a constant caloric load delivered<br />

to the duodenum; therefore, formulas with a<br />

concentration above 1kcal/ml should be used with<br />

caution. 5 The nature of the energy supply does not<br />

seem to play a role in gastric function, except for<br />

the type of triglyceride (long-chain triglycerides<br />

(LCTs) or medium-chain triglycerides (MCTs)). 6<br />

The effects of CEN on intestinal motility can be<br />

analyzed by manometry. Migrating complexes are<br />

observed in adult patients during CEN, as during<br />

the fasting state. 7 The type of formula influences<br />

jejunal motility according to the nature of triglycerides<br />

and/or the molecular weight of peptides and<br />

proteins. 8 Infusion of MCTs or short peptides<br />

within the duodenal lumen is associated with the<br />

persistence of migrating complexes, as during<br />

fasting. 9 Very few data are available about the<br />

changes of colonic motility induced by CEN; the<br />

continuous gastric infusion of the nutritive formula<br />

modifies the gastrocolic reflex.<br />

Gallbladder motility is maintained during EN, as<br />

assessed by ultrasonography. 10 The type of infused<br />

lipids (MCTs versus LCTs) influences gallbladder<br />

motility. 11 Biliary complications, such as sludge or<br />

cholelithiasis, are very rare during long-term CEN.<br />

539


540<br />

Enteral nutrition<br />

Digestive secretion and hormonal response<br />

Gastric secretion depends mostly on protein<br />

intake, and in case of elemental diet, on amino<br />

acid composition. 12 The secretory response is not<br />

influenced by carbohydrates, but is reduced by<br />

lipids. It has not been demonstrated whether or<br />

not the type of diet (i.e. elemental, semi-elemental<br />

or polymeric) modifies gastric acid secretion. 6<br />

Cholecystokinin secretion as well as pancreatic<br />

secretion are maintained during CEN. The amount<br />

of nitrogen infused within the jejunum correlates<br />

with the secretion of chymotrypsin and lipase. 13<br />

Secretory responses do not differ between an<br />

elemental or polymeric nitrogen supply. 14<br />

Gastrin secretion is also maintained during CEN,<br />

but its response to protein load is decreased.<br />

Gastric or duodenal CEN stimulate insulin secretion,<br />

depending on the type of infused nutrients.<br />

The lack of steatosis during CEN suggests that the<br />

insulin response is lower when carbohydrates are<br />

infused enterally as compared to parenterally. 15<br />

Effects of continuous enteral nutrition on<br />

mucosal trophism<br />

The effects of artificial nutrition on small-bowel<br />

mucosal trophism remain controversial. In experimental<br />

animal systems, CEN has been shown not<br />

to modify the absorption capacity of the proximal<br />

small bowel. In the distal small bowel and colon,<br />

however, enzymatic and functional capacity are<br />

decreased, despite a normal mucosal architecture;<br />

in addition, DNA as well as protein content are<br />

reduced. 16 These changes could be the consequence<br />

of the almost complete absorption of nutrients<br />

within the proximal part of the small bowel,<br />

leading to lack of stimulation of the distal segment.<br />

This suggests an ability of CEN to achieve bowel<br />

rest in the distal part of the bowel, providing efficient<br />

treatment for ileocolic inflammatory diseases.<br />

Effects of continuous enteral nutrition on<br />

energy expenditure<br />

The thermogenic effect of feeding is related to the<br />

increase of energy expenditure following ingestion<br />

of food. The increase in energy expenditure<br />

induced by CEN in normal subjects is lower than<br />

that for the same nutrient load as a bolus. 17,18<br />

Thus, the constant administration of energy<br />

substrates might reduce the energy storage and<br />

maintain homeostasis at a lower energy expenditure.<br />

19<br />

Finally, CEN by the slow and continuous administration<br />

of nutrients into the GI tract enables their<br />

optimal utilization to be achieved, despite intestinal<br />

illness. By changing the conditions of flow and<br />

of contact between the nutritive formula and the<br />

digestive tract, CEN may increase the capacity for<br />

intraluminal digestion and intestinal absorption.<br />

This feeding technique seems logical and efficient<br />

when the absorptive surface is reduced, e.g. in<br />

short-bowel syndrome, villous atrophy, enterocutaneous<br />

fistula or proximal enterostomy.<br />

Indications<br />

Indications for EN are different from indications<br />

for PN, since the use of EN as nutritional support<br />

is based on normal or at least partially preserved<br />

gut functions (Table 33.1).<br />

Digestive indications<br />

Since EN has a trophic effect on the intestinal<br />

mucosa, and helps maintain mucosal integrity, it<br />

plays an important role in the treatment of many<br />

digestive diseases, either replacing or completing<br />

oral feeding. Digestive diseases leading to an<br />

anatomical or functional reduction of the absorption<br />

capacity of the small bowel represent the first<br />

group; they include short-bowel syndrome,<br />

protracted diarrhea with villous atrophy and<br />

inflammatory bowel disease.<br />

Short-bowel syndrome<br />

Short-bowel syndrome (see Chapter 29) can be<br />

defined as malabsorption following small-intestinal<br />

resection. Prognosis after extensive intestinal<br />

resection has improved with the expanded use of<br />

PN over the past 20 years. 20–24 Following massive<br />

resection of the small intestine, the remaining<br />

small bowel undergoes an adaptative process characterized<br />

by epithelial hyperplasia (see Chapter


Table 33.1 Indications for enteral nutrition<br />

Digestive indications<br />

Short-bowel syndrome<br />

Protracted diarrhea of infancy<br />

Immunodeficiency<br />

Graft-versus-host disease<br />

Crohn’s disease<br />

<strong>Neonatal</strong> abdominal surgery<br />

gastroschisis<br />

omphalocele<br />

meconial ileus<br />

distal intestinal fistulae<br />

small-bowel resection<br />

necrotizing enterocolitis<br />

complicated Hirschsprung’s disease<br />

chronic intestinal pseudo-obstruction syndrome<br />

Other malabsorption syndromes<br />

cystic fibrosis<br />

cholestatic liver disease<br />

In children with normal intestinal function<br />

Eating disorders<br />

disorders of sucking and swallowing<br />

neurological impairment<br />

Esophageal diseases<br />

Hypermetabolic states<br />

head injury<br />

extensive burns<br />

cancer, AIDS, bone marrow transplantation<br />

renal failure<br />

congenital heart diseases<br />

chronic pulmonary diseases<br />

Inborn errors of metabolism<br />

glycogen storage diseases<br />

deficiencies of urea cycle<br />

Neonatology and prematurity<br />

Abnormal eating behavior<br />

anorexia nervosa<br />

failure to thrive<br />

29). 25 An early and optimal use of the enteral route<br />

is the condition of small-bowel adaptation.<br />

However, in children with short-bowel syndrome,<br />

oral feeding rarely permits a rapid reduction of the<br />

PN supply, because numerous patients have eating<br />

disorders instead of hyperphagia. In such conditions,<br />

CEN enables the delivery of nutrient to be<br />

increased to the digestive tract independently of<br />

the feeding behavior, with a known benefit on socalled<br />

intestinal adaptation, and added benefits on<br />

Indications 541<br />

the prevention of PN-associated liver disease. 25<br />

The delay for postoperative adaptation to occur<br />

depends on the length and quality of the residual<br />

bowel and on the presence of the ileocecal valve<br />

and colon. 21<br />

Severe protracted diarrhea of infancy<br />

A syndrome of intractable diarrhea of infancy was<br />

first described by Avery et al in 1968. Its definition,<br />

presentation and outcome have considerably<br />

changed during the past two decades. 26 This<br />

syndrome could now be defined as persistent diarrhea<br />

despite prolonged bowel rest requiring longterm<br />

total PN in children when no effective treatment<br />

is available. 27,28 According to that definition,<br />

EN in this circumstance is indicated and often<br />

effective. 29,30 In fact, the reduction of digestive<br />

secretions, villous atrophy and acquired brushborder<br />

disaccharidase deficiency lead to malabsorption<br />

and malnutrition. Most of the time, a<br />

short course of PN followed by protracted CEN<br />

provides control of the disease within 6–8<br />

weeks. 31,32 In a prospective study of CEN versus<br />

PN, Orenstein showed that the resolution of diarrhea<br />

was faster in the enterally fed group. 32<br />

The use of CEN in children and mainly infants<br />

with protracted diarrhea also presenting severe<br />

malnutrition may prove difficult. If the response to<br />

CEN is not good enough to provide rapidly<br />

adequate caloric supplies with resolution of diarrhea,<br />

PN should be started. Such decisions must<br />

be taken by experienced teams in specialized and<br />

well-staffed units.<br />

Severely malnourished infants with some types of<br />

particularly severe celiac disease, intolerance to<br />

cow’s milk proteins, protein hydrolysates, or<br />

specific malabsorption syndromes such as<br />

Anderson’s disease, may also benefit from CEN. 33<br />

Inflammatory bowel diseases<br />

Enteral feeding has been used for many years,<br />

particularly in Europe, not only to improve nutritional<br />

status and growth, but also to influence<br />

disease activity in patients with Crohn’s<br />

disease. 34–43 It has been shown that CEN using an<br />

elemental diet is as effective as a high dose of<br />

steroids in inducing remission in pediatric<br />

patients with Crohn’s disease involving the small


542<br />

Enteral nutrition<br />

intestine. The meta-analysis by Griffiths et al<br />

suggested that EN was inferior to steroid therapy. 44<br />

This was largely an analysis of adult studies and<br />

excluded several pediatric studies showing striking<br />

efficacy. More recent meta-analyses have shown an<br />

equivalent effect of EN and steroids in achieving<br />

remission. 45,46 In addition, it is clear that EN is<br />

advantageous in preserving growth while remission<br />

is achieved. There are no relevant data in children.<br />

However, in adults, studies on the comparative<br />

effect of elemental diets, protein hydrolysates<br />

and intact protein formulas have shown induced<br />

remission of polymeric diets in controlling Crohn’s<br />

disease involving the small bowel. How such treatment<br />

works in children with Crohn’s disease is not<br />

clear. Certainly, the increased energy intake<br />

provided by EN must be one factor. However, there<br />

is evidence that such treatment may have a specific<br />

anti-inflammatory effect. A reduction of cytokine<br />

production by isolated lamina propria lymphocytes<br />

following EN with a polymeric casein-based<br />

formula was shown to an extent equivalent to that<br />

produced by steroids or cyclosporin. 43 In addition,<br />

Fell et al have shown a massive reduction in mRNA<br />

transcripts for interleukin (IL)-1β using its derivative<br />

CT3211. 47 Removal of antigenic material, alteration<br />

in intestinal microflora, changes in gut<br />

hormone levels and the presence of bioactive transforming<br />

growth factor (TGF) β-1 in casein-based<br />

formulas may all play a role in the clinical success<br />

of EN. 48,49 A glutamine-enriched polymeric diet<br />

offered no advantage over a standard low-glutamine<br />

polymeric diet in the treatment of active<br />

Crohn’s disease. 50 EN may also be helpful in correction<br />

or maintenance of the nutritional state, especially<br />

during a relapse of Crohn’s disease. 45 EN is<br />

part of the preparation for a surgical procedure and<br />

may be useful during recovery, especially after<br />

intestinal resections or enterostomies.<br />

In case of severe digestive involvement during<br />

Schönlein–Henoch purpura, CEN can be used as<br />

nutritional support in the absence of occlusion. 51<br />

<strong>Neonatal</strong> abdominal surgery<br />

In neonatal abdominal surgery for congenital or<br />

acquired disease, CEN, usually combined with PN,<br />

offers prolonged nutritional support. This has<br />

transformed the prognosis in many conditions and<br />

is particularly important in the following situations:<br />

reduction of the absorptive surface with<br />

enterocutaneous fistulae or extensive intestinal<br />

resection; and functional disorders of gut motility,<br />

such as malfunctions of a duodenojejunal anastomosis,<br />

‘plastic’ peritonitis after repeated interventions,<br />

gastroschisis and omphalocele.<br />

Chronic intestinal pseudo-obstruction syndrome,<br />

with neonatal onset, is a special condition in which<br />

EN is rarely tolerated and usually does not allow<br />

PN and/or a surgical procedure such as ileostomy<br />

to be avoided. 52–54<br />

Other malabsorption syndromes<br />

Cystic fibrosis<br />

Failure to thrive is common in children with cystic<br />

fibrosis (CF), and results from several factors,<br />

including malabsorption from pancreatic insufficiency,<br />

bile salt abnormalities, anorexia, increased<br />

energy expenditure, protein catabolism from<br />

pulmonary infections and/or fever. 55 Even with an<br />

optimal approach to oral feeding, some patients fail<br />

to respond to conservative nutritional therapy. EN<br />

is proposed as a second-stage intervention and can<br />

help to restore and maintain nutritional status. 56–58<br />

EN is in most cases easily performed at home. It is<br />

generally performed during the night over 8–10h;<br />

CF patients are asked to eat and drink as much as<br />

possible during the day. Nasogastric tube feeding is<br />

generally used as a first step. The tube is passed<br />

every night, 1–2h after dinner, and removed in the<br />

early morning before physical therapy so that<br />

patients are not disturbed during the day for school<br />

attendance. In some children, a nasogastric tube<br />

becomes increasingly uncomfortable because of<br />

nausea, vomiting and nasal discomfort as a result of<br />

nasal polyposis or dislodgement during coughing<br />

in cases of pulmonary exacerbation. Thus, percutaneous<br />

endoscopic gastrostomy (PEG) has become<br />

the method of choice for performing long-term<br />

home nocturnal EN. A button device can be placed<br />

2–3 months after PEG tube placement, thus improving<br />

the physical and psychological tolerance of EN.<br />

It is important to look for glucose tolerance as well<br />

as for gastroesophageal reflux before starting EN.<br />

Psychological support of the patient is essential,<br />

since body image and self-esteem are frequently<br />

altered in CF patients, especially in cases of weight<br />

loss and/or failure to thrive. Nutritional support by<br />

EN, even recognized as necessary, is rightly considered<br />

by patients and/or parents as a new constraint.


Thus, the usefulness of EN must be precisely<br />

explained in order for it to become part of clear<br />

global therapeutic management. EN is required in<br />

malnourished children with decreased growth<br />

velocity, especially when transplantation is<br />

planned. Adequate EN, using either an intact<br />

formula with pancreatic enzyme extract or a semielemental<br />

diet can be achieved through either nasogastric<br />

or PEG tubes. Finally, it should be stressed<br />

that in CF nutritional supplementation improves<br />

not only nutritional status but also respiratory function.<br />

58–60<br />

Cholestatic liver disease<br />

Mechanisms leading to protein–energy malnutrition<br />

in infants and children with chronic liver disease<br />

are incompletely known. 61–63 They include reduced<br />

biliary secretion and intraluminal bile concentration<br />

resulting in malabsorption of lipid and fatsoluble<br />

vitamins. Energy requirements are<br />

increased by different mechanisms including<br />

portosystemic shunting and ascites, abnormal intermediary<br />

metabolism and the energy demands of<br />

complications such as sepsis or variceal bleeding.<br />

Anorexia is common in children with chronic liver<br />

disease resulting from organomegaly, abdominal<br />

pressure effects of ascites, congested gastric<br />

mucosa, reduced motility from portal hypertension,<br />

central effects of unidentified toxins, dietary manipulations<br />

such as fluid restriction or use of unpalatable<br />

feeds. Several factors contribute to long-chain<br />

polyunsaturated fatty acid (PUFA) deficiency<br />

including low PUFA intake, malabsorption and<br />

disturbed metabolism of long-chain PUFAs. Finally,<br />

the interaction of growth hormone with insulin-like<br />

growth factor (IGF)-I and its binding proteins constitute<br />

an important mechanism linking nutrition and<br />

growth.<br />

The most common cause of cirrhosis in infants is<br />

biliary atresia. Since orthotopic liver transplantation<br />

(OLT) is the only effective treatment, the role<br />

of nutritional support preceding transplantation is<br />

of importance. 63,64 Nutritional support has a direct<br />

impact on survival and suitability as candidates for<br />

OLT, while the effect of preoperative nutritional<br />

support on outcome after OLT is a matter of<br />

debate. 65,66<br />

Oral food intake is most of the time insufficient in<br />

these children. EN providing MCT-rich feeds is<br />

Indications in children with normal intestinal function 543<br />

recommended and should be associated with an<br />

adequate fat-soluble vitamin supply. 67,68 Many<br />

currently available commercial nutritional products<br />

are suitable for infants with chronic liver<br />

disease but none are ideal for all situations.<br />

Additives are usually necessary in infants and<br />

children with persistent failure to thrive.<br />

Ordinarily, nutritional management can be carried<br />

out at home. However, two issues remain controversial.<br />

First, children with portal hypertension<br />

may have esophagal varices with the risk of bleeding<br />

due to the nasogastric tube; in this circumstance,<br />

the use of a silicone tube is mandatory and<br />

home EN is contraindicated. Second, the use of<br />

branched-chain amino acids to feed children with<br />

chronic encephalopathy is debated. 69<br />

Indications in children with normal<br />

intestinal function<br />

Eating disorders<br />

EN is required in cases of inability to eat normally,<br />

i.e. in those situations that are secondary to structural<br />

or functional abnormalities of the upper GI<br />

tract or neurological impairment of the processes<br />

involved in sucking and/or swallowing (see also<br />

Chapter 15).<br />

Esophageal diseases including esophageal atresia,<br />

fistula or stenosis, can result from sequelae of<br />

epidermolysis bullosa. These conditions often<br />

need EN, usually through gastrostomy or duodenal<br />

tubes. 70 The choice of feeding through a gastrostomy<br />

or a trans<strong>pylori</strong>c tube must be assessed<br />

according to the patient’s age, disease and condition.<br />

Children with chronic diseases inducing immaturity<br />

or inability to feed orally, especially with<br />

sucking and swallowing troubles as seen in neurologically<br />

impaired children, with neuromuscular<br />

chronic diseases or cerebral palsy also require EN,<br />

using gastrostomy tubes. 71–73<br />

Other indications<br />

Indications for EN have been extended in recent<br />

years to include several extraintestinal conditions.


544<br />

Enteral nutrition<br />

Hypermetabolic states<br />

Patients with acute hypermetabolic conditions,<br />

such as those resulting from head injury or burns,<br />

should receive an adequate nutrient supply, which<br />

should be given enterally in most cases. 74 EN has<br />

been used successfully in children with cancer,<br />

bone marrow transplantation or AIDS. 75–78<br />

All chronic diseases inducing both an increase in<br />

protein–caloric needs and anorexia, such as<br />

chronic renal failure 79,80 with or without hemodialysis<br />

awaiting transplantation, or congenital heart<br />

disease, 81,82 often induce malnutrition and failure<br />

to thrive. Children with these disorders benefit<br />

from EN, since improvement of nutritional status<br />

is expected to influence the outcome positively<br />

after transplantation.<br />

Inborn errors of metabolism<br />

EN is part of the treatment of numerous inherited<br />

metabolic diseases, which induce hypoglycemia<br />

(glycogen storage disease) 83–85 or increase protein<br />

catabolism, e.g. enzymatic deficiencies of the urea<br />

cycle. 86,87 CEN is required to avoid neurological<br />

consequences of these metabolic disorders.<br />

Premature infants<br />

EN is commonly used in premature infants, and in<br />

small-for-gestational age neonates with sucking<br />

and swallowing troubles. Mother’s milk can be<br />

given, sometimes into the duodenum rather than<br />

into the stomach in low-birth-weight infants (less<br />

than 1000g). All the factors discussed earlier must<br />

be carefully monitored in this high-risk group.<br />

Some physicians prefer to stop CEN in infants<br />

with respiratory disease, and to use total PN (see<br />

also Chapter 34).<br />

Miscellaneous conditions<br />

Miscellaneous indications for EN include primitive<br />

difficulties of eating behavior, including<br />

anorexia nervosa, 88–91 and chronic idiopathic<br />

failure to thrive. Failure to thrive may be due to a<br />

poor parent–child relationship leading to insufficient<br />

food intake. In such circumstances, tube<br />

feeding can be used to demonstrate that growth<br />

can be achieved if adequate nutrients are<br />

provided. 88,89 In older children and adolescents<br />

with anorexia nervosa, EN can be used according<br />

to the opinion and experience of the psychiatric<br />

team. 90,91<br />

Techniques<br />

Material<br />

Nasogastic tubes<br />

The route of EN administration should be individually<br />

tailored, depending on the underlying condition.<br />

The intragastric route of administration is the<br />

more used in children, since it is the more physiological<br />

route. It permits the action of salivary and<br />

gastric enzymes, the bactericidal action of gastric<br />

acid and better mixing with biliary and pancreatic<br />

juice. Therefore, the duodenal or jejunal route is<br />

used in few circumstances in children. For intragastric<br />

EN, nasogastric tubes or gastrostomy tubes<br />

may be used. Nasogastric tube feeding is the best<br />

initial approach to EN, to evaluate the tolerance of<br />

EN before placing a permanent gastrostomy tube,<br />

and/or when a brief period of EN support is anticipated.<br />

The nasogastric tube may be made of<br />

polyvinylchloride (PVC), polyurethane or silicone.<br />

The tube size is chosen according to the weight<br />

and age of the child, with an external diameter as<br />

small as possible. The nasogastric tube is inserted<br />

nasogastrically by using the nose–umbilicus<br />

distance as a reference point. Its position is<br />

routinely checked by epigastric auscultation<br />

during the injection of air and the aspiration of<br />

fluid with a pH less than 3. Duodenal or jejunal<br />

tube placement is more difficult; the patient<br />

should be placed in the right lateral position and,<br />

if necessary, after intramuscular injection of metoclopramide.<br />

The position of the distal end of the<br />

tube is then checked by X-rays. Careful nasal fixation<br />

of the tube is used to avoid displacement; it is<br />

taped to the upper lip, the ipsilateral cheek and the<br />

external ear. In some particular indications for EN,<br />

the feeding tube may also be introduced through<br />

the mouth, especially in premature babies. The<br />

placement of the nasogastric tube made of PVC is<br />

easier, but these tubes should be changed every<br />

2–4 days. Indeed, the tube becomes rigid if left<br />

longer, whereas the silicone or polyurethane tube<br />

may be used over 3-week periods or more. On the<br />

other hand, silicone and polyurethane tubes are<br />

more flexible and are more easily displaced by


vomiting; they are preferentially used for trans<strong>pylori</strong>c<br />

and long-term EN. When the child requires<br />

prolonged EN (more than 2 or 3 months) and when<br />

EN is well tolerated, a gastrostomy should be<br />

considered.<br />

Percutaneous gastrostromy<br />

The PEG technique has revolutionized the placement<br />

of enteric feeding tubes in children. It is now<br />

a widely used and well-tolerated technique in children.<br />

92–95 This relatively simple and fast procedure<br />

can be performed during upper-GI endoscopy in<br />

an endoscopy suite with the use of conscious sedation<br />

and local anesthesia or under general anesthesia.<br />

Several techniques have been developed<br />

with the common basic principle that the endoscope<br />

locates the site of tube placement from<br />

within the stomach. The transillumination of the<br />

light from the endoscope through the abdominal<br />

wall identifies the site of skin incision. The ideal<br />

site is on the greater curvature of the stomach with<br />

the stoma sited on the anterior abdominal wall,<br />

below the costal margin with consideration to the<br />

axis of bending and to the clothing. To reduce the<br />

risk of infection at the stoma site, perioperative<br />

antibiotic prophylaxis is recommended. The tube<br />

can be used within 12–24h, while post-insertion<br />

edema at the stoma site is closely monitored. Care<br />

should be taken not to pull the bolster too high.<br />

The initial PEG tube is changed after 2–3 months,<br />

by which time a good tract has formed. Buttonreplacement<br />

gastrostomy devices provide patients<br />

a cosmetic advantage in case of long-term EN. PEG<br />

is contraindicated only in a few patients with<br />

previous abdominal surgery, abnormal abdominal<br />

anatomy, or severe deformities of the chest and<br />

spine that modify the position of the stomach. In<br />

such cases, a surgical gastrostomy tube should be<br />

placed. The implantation of a jejunal feeding tube,<br />

via PEG, is a possible method for the treatment of<br />

inadequate oral feeding in patients who are<br />

affected by gastroesophageal reflux and is thus an<br />

alternative to fundoplication and drugs. 96,97<br />

Special PEG techniques have also been developed<br />

for ICU patients. 98<br />

EN may be delivered by bolus or pump-controlled<br />

techniques, but should not be administered by<br />

gravity in children. Pumps recommended for pediatric<br />

use have to provide clear flow rate display<br />

and alarms. Miniaturized and battery-powered<br />

Techniques 545<br />

pumps are specially designed for home and ambulatory<br />

EN.<br />

Nutrients<br />

Nitrogen The absorption of amino acids is more<br />

rapid and efficient when given in the form of short<br />

peptides rather than free amino acids. 99,100 In addition,<br />

the quality, in terms of digestion and intestinal<br />

absorption of protein hydrolysates, depends on<br />

the type of hydrolysate; for example, lactalbumin<br />

is superior to casein. 101,102 Thus, the initial<br />

formula should be based on polypeptides, with a<br />

lower osmolality, rather than on a mixture of free<br />

amino acids. Nitrogen needs vary depending on<br />

tolerance, and range from 350 to 500mg/kg per<br />

day, or even more in some highly catabolic situations.<br />

The nitrogen/calorie ratio must be considered;<br />

protein intakes should represent 10% of the<br />

energy intake, especially for premature infants. 103<br />

Carbohydrates Disaccharidase enzymatic activities<br />

are depressed in disease involving the small-intestine<br />

mucosa. Lactase appears to be the most sensitive<br />

to injury and the last of the disaccharidases to<br />

recover. In addition, certain drugs such as<br />

neomycin or colchicine depress the intestinal<br />

disaccharidases. Therefore, it is important to avoid<br />

dietary sources of lactose. Other disaccharides<br />

should also be omitted from the solution used for<br />

initial feeding, as their corresponding brushborder<br />

enzymatic activities are reduced. The only<br />

carbohydrate allowed during the initial days of EN<br />

should be glucose, but its high osmolality<br />

(5.5mOsmol/g) limits the rate of infusion, and thus<br />

the total amount given. The later addition of fructose<br />

may lead to an increase of disaccharidase<br />

activity, so making it easier to substitute maltose<br />

and sucrose for glucose. The subsequent introduction<br />

of low-osmolality oligosaccharides may allow<br />

the intake to reach 20 or even 24g/kg per day, especially<br />

in infants, without resulting in a final solution<br />

osmolality higher than 350mOsm/l.<br />

Lipids The behavior of MCTs within the intestinal<br />

lumen and their absorption characteristics are<br />

primarily due to their greater water solubility.<br />

104,105 MCTs are hydrolyzed faster than LCTs in<br />

the small intestine by pancreatic lipase; they are<br />

converted almost exclusively into free fatty acids<br />

and glycerol and directly reach the portal circulation<br />

and the liver. Nevertheless, in case of pancre-


546<br />

Enteral nutrition<br />

atic insufficiency, MCTs may be absorbed intact.<br />

The excessive use of a MCT-containing diet can<br />

lead to osmotic diarrhea as a result of their rapid<br />

hydrolysis. Dicarboxylic aciduria has been<br />

described in infants supplemented with MCT-rich<br />

formulas without any evidence of a deleterious<br />

effect. 106 The provision of essential fatty acids<br />

(EFA) must be considered since MCTs contain no<br />

EFA. Furthermore, MCTs decrease the absorption<br />

of LCTs; therefore, supplementation with linoleic<br />

acid must be provided. However, its addition to a<br />

formula based on MCTs may be insufficient to<br />

prevent EFA deficiency, thus making it necessary<br />

to provide EFA parenterally. Nevertheless, most<br />

formulas containing MCTs include up to 50% of<br />

lipid as LCTs. By stimulating biliary and pancreatic<br />

secretions, LCTs promote increased intestinal<br />

motility; LCTs in excess in the intestinal lumen,<br />

especially if they are hydroxylated by bacteria,<br />

reverse the rate of water and electrolyte absorption<br />

and increase malabsorption. In those conditions,<br />

the addition of cholestyramine associated with<br />

EFA supplementation may be appropriate. Finally,<br />

a lipid intake of 3–4g/kg per day may be achieved,<br />

depending on the absorption capacity and digestive<br />

tolerance.<br />

Other components In infants and children, the<br />

recommended energy intake varies from 70 to<br />

100kcal/kg per day, and water rarely exceeds<br />

80ml/kg per day. The recommendations for<br />

average supplies of vitamins and trace elements<br />

are shown in Tables 33.2 and 33.3. Competition<br />

between trace elements such as copper and zinc<br />

has been taken into account in these recommendations;<br />

the daily supplement should be higher in<br />

premature than in full-term infants.<br />

Choice of a formula<br />

Enteral feeding formulas are divided into several<br />

families. The choice of a formula is made according<br />

to numerous parameters, such as<br />

protein–caloric needs, digestive function and tolerance<br />

to fluid intake – all obviously dependent on<br />

the age and on the underlying disease.<br />

(1) In newborns and young infants with normal<br />

or nearly normal intestinal function, breast or<br />

humanized milk may be used, while commercial<br />

polymeric diets are available for older<br />

children. Blenderized diets can be prepared<br />

Table 33.2 Vitamin requirements<br />

Infants Children<br />

Vitamin A (µg) 300–750 450–1000<br />

Vitamin D (IU) 400–1000 200–2500<br />

Vitamin E (mg) 3–10 10–15<br />

Vitamin K (µg) 50–75 50–70<br />

Vitamin Bl (mg) 0.4–0.5 1.5–3<br />

Vitamin B2 (mg) 0.4–0.6 1.1–3.6<br />

Vitamin B5 (mg) 2–5 0.5–5<br />

Vitamin B6 (mg) 0.1–1.0 1.5–2<br />

Vitamin B12 (µg) 0.3–3 3–100<br />

Vitamin C (mg) 25–35 20–100<br />

Folic acid (µg) 20–80 100–500<br />

Biotin (µg) 35–50 150–300<br />

Niacin (mg) 6–8 5–40<br />

Table 33.3 Trace element requirements<br />

Infants Children<br />

Element (/kg/day) (/day)<br />

Iron (mg) 50 100–2500<br />

Zinc (mg) 100–250 1000–5000<br />

Copper (mg) 20–30 200–300<br />

Selenium (µg) 2–3 30–60<br />

Manganese (µg) 1–10 50–250<br />

Molybdenum (µg) 0.25–3 50–70<br />

Chrome (µg) 0.25–2 10–20<br />

lodine (µg) 1–5 50–100<br />

Fluoride (µg) 20 500–1000<br />

using food from the kitchen, but are usually<br />

replaced by polymeric formulas containing<br />

‘intact’ proteins with or without fibers, which<br />

are commercially available; they are indicated<br />

in most non-stressed patients, with<br />

normal gut function. These formulas contain<br />

1kcal/ml (standard) to 1.5kcal/ml (calorically<br />

dense).<br />

(2) In case of GI disease, the choice of the nutritive<br />

solution must take into account not only the<br />

child’s age and nutritional status but also the<br />

underlying digestive disease, e.g. the presence<br />

of anatomical and functional changes in the


intestine, whether due to an extensive reduction<br />

of the absorptive surface, to enteropathy<br />

or to pancreatic insufficiency. Limiting factors<br />

in such cases are impairment of gastric, biliary<br />

and pancreatic secretions, disturbances of the<br />

intestinal flora and malabsorption. The osmolarity<br />

of enteral formulas should be kept below<br />

320mOsmol/kg, nutrients should be rapidly<br />

transferred across the intestine and they<br />

should not leave an intraluminal residue.<br />

Substrates requiring intraluminal hydrolysis<br />

(proteins, starches, LCTs) should be excluded,<br />

as should potentially highly antigenic<br />

substrates (cow’s milk proteins, gluten and<br />

soy). Oligomeric or ‘semi-elemental’ diets are<br />

designed for use in patients with malabsorption,<br />

such as in short-bowel syndrome, CF,<br />

cholestasis or food allergy. These diets include<br />

dipeptides, tripeptides and a few free amino<br />

acids, combined with LCTs and MCTs, and<br />

carbohydrates including glucose polymers and<br />

maltodextrins. Powder formulas enable progressive<br />

adaptation of the concentration to the<br />

clinical condition (for example, to increase the<br />

concentration progressively from 0.65kcal/ml<br />

up to 1kcal/ml or more). Ready-to-use formulas<br />

usually contain 1kcal/ml.<br />

(3) Free amino acid-based formulas tend to have a<br />

higher osmolality. For instance, the amino<br />

acid-based formula Neocate ® specifically designed<br />

for infants under 1 year of age, has an<br />

osmolality of 360mOsm. The lipid component<br />

is a mixture of safflower, coconut and soy oils<br />

to provide essential and non-essential fatty<br />

acids. The carbohydrates are from corn syrup<br />

solids or maltodextrins, both derived from<br />

corn starch and differing in the size of glucose<br />

polymers. They are indicated especially in<br />

children with severe food allergy who do not<br />

tolerate even the peptide-based formulas.<br />

Children with severe malabsorption or shortbowel<br />

syndrome unable to tolerate peptidebased<br />

formulas (protein hydrolysates) might<br />

benefit from the free amino acid-based formulas.<br />

107,108<br />

(4) Special formulas are proposed for some indications<br />

such as renal failure, liver failure,<br />

pulmonary disease, diabetes and metabolic<br />

diseases. On the other hand, special formulas<br />

have been developed for preterm infants, who<br />

Techniques 547<br />

are characterized by the following factors:<br />

decreased energy and glycogen stores, limited<br />

gastric capacity, reduced intestinal peristalsis,<br />

reduced bile salt pool and delayed enzymatic<br />

development. The main differences in the<br />

composition of premature and term infant<br />

formulas include: partial substitution of<br />

oligosaccharides for lactose; increased protein<br />

content to support rapid growth; 60:40<br />

whey/casein ratio to increase essential amino<br />

acid intakes and protein digestibility; partial<br />

substitution of LCTs by MCTs; and adapted<br />

concentrations of vitamin E, calcium, phosphorus<br />

and iron.<br />

(5) Nutritional formulas in which each of the<br />

constituents is modified independently are<br />

mostly used in special conditions, e.g. selective<br />

malabsorption. In that case, glucose may be<br />

given initially as the calorie source, the amount<br />

being increased progressively and controlled<br />

according to the stool volume, pH and absence<br />

of reducing substances in the stools. In the first<br />

days of feeding, at least, a molar ratio of glucose<br />

and so-dium is maintained. Then, protein<br />

hydrolysates are gradually introduced according<br />

to digestive tolerance. The caloric enteral<br />

intake usually increases from 10–15 to<br />

70–80kcal/kg per day over the first week. In a<br />

second step, qualitative and quantitative<br />

changes are gradually made: fructose is added<br />

to the glucose, and then disaccharides are<br />

introduced, starting with maltose. Nitrogen and<br />

lipids are simultaneously increased. During<br />

this period, the introduction of oligosaccharides<br />

precede that of a commercially available<br />

semi-elemental diet.<br />

Regulation of intakes and rhythm of enteral<br />

nutritional delivery<br />

EN should be progressively introduced, depending<br />

on the child’s nutritional status and the indications<br />

for EN. In case of digestive disease, CEN can<br />

be used after a prolonged period of PN or simply<br />

after a brief phase of peripheral venous infusion.<br />

The first step includes the progressive reduction of<br />

the parenteral intake and the stepwise increase of<br />

EN according to the digestive tolerance. The semielemental<br />

diet (protein hydrolysates) can be used<br />

early, by progressively increasing the volume as


548<br />

Enteral nutrition<br />

well as the concentration, according to digestive<br />

tolerance and until optimal energy and nitrogen<br />

intakes are achieved. The water and sodium<br />

supply should be increased to compensate for the<br />

intestinal losses generally induced by the start of<br />

EN; the tolerance is estimated from the weight of<br />

the child, from the volume and osmolality of urine<br />

samples and from the plasma osmolality. The<br />

tolerance and needs are estimated from 24-h urine<br />

analysis, while attempting to maintain natriuresis<br />

of 2–3mmol/kg per day. At the same time, potassium<br />

intake is adjusted as a function of the nitrogen<br />

and energy intakes.<br />

The rhythm of EN delivery depends on the underlying<br />

disease. Intermittent feeding using a bolus is<br />

more physiological and well tolerated when the<br />

digestive function is normal. Continuous cyclic<br />

nocturnal EN is better tolerated in some patients<br />

who do not tolerate bolus feeding, and provides<br />

less interference with daytime oral intake.<br />

However, a continuous 24/24h rhythm of delivery<br />

is indicated in case of impaired digestive function.<br />

The weaning period varies from a few days to<br />

several weeks or months. Eating disorders can be<br />

avoided by the maintenance of sucking and swallowing<br />

functions during the period of CEN. It has<br />

also been demonstrated that non-nutritive sucking<br />

during CEN enhances growth and intestinal maturation<br />

in premature babies. 109,110 Weaning includes<br />

a period of continuous night-time feeding<br />

supplemented by several meals in the daytime<br />

until the latter account for 50% of the total intake.<br />

Oral feeding must be carefully increased because<br />

of the relatively low intestinal activity due to longterm<br />

CEN.<br />

Complications of enteral therapy<br />

These are rare but serious. Strict adherence to the<br />

procedure indicated and careful supervision are<br />

essential to prevent them. 111,112<br />

Gastrointestinal complications<br />

Vomiting and aspiration are the most threatening<br />

complications associated with tube feeding, at<br />

home as well as in the hospital, while its incidence<br />

seems to be low in children on home EN. 113<br />

Irregular flow rate of infusion, delayed gastric<br />

emptying due to the underlying disease or to the<br />

drugs, gastroesophageal reflux and tube placement<br />

or migration into the distal esophagus, behavioral<br />

vomiting and formula intolerance are risk factors<br />

for vomiting and aspiration. The possible preventive<br />

effect of a semirecumbent position has been<br />

discussed. 114<br />

Diarrhea is the most common complication of EN,<br />

and can occur in about 30% of patients. It might<br />

result in dehydration and hypoglycemia. It may be<br />

due to multiple causes, especially intraduodenal<br />

infusion, high osmolarity, excess or irregular infusion<br />

rate and bacterial contamination of the<br />

formula. Increased stool losses occur when the<br />

combined absorptive capacity of the small bowel<br />

and the salvaging capacity of the colon are<br />

exceeded.<br />

Mechanical complications<br />

Tube-related complications such as nasal trauma<br />

due to placement of a nasogastric tube, laryngeal<br />

ulceration or stenosis, esophageal stricture or<br />

perforation mainly if an introducer is used, gastric<br />

or duodenal perforation, intestinal bleeding and<br />

bowel intussusception are exceptional in children,<br />

but have been described with PVC tubes left for 8<br />

days or more. 115–117 Pyloric stenosis has been<br />

reported after prolonged duodenal feeding in<br />

premature babies; this is probably due both to the<br />

presence of the trans<strong>pylori</strong>c tube and to spasm of<br />

the pylorus caused by the direct infusion of lipid<br />

into the duodenum.<br />

Ear, nose and throat complications are frequent in<br />

children who receive EN through a nasogastric<br />

tube. The incidence of these complications must be<br />

taken into account when making a decision for a<br />

gastrostomy.<br />

Complications from PEG placement include worsening<br />

of gastroesophageal reflux, dumping<br />

syndrome (if the PEG is to close to the pylorus) and<br />

gastrocolic fistula. Local skin problems include<br />

infection, cellulitis, granulomas and leaking.<br />

Leaking of formula, which may be the consequence<br />

of lack of adequate inflation of the<br />

balloon, or to malfunction of the antireflux valve,<br />

induces a vicious cycle with enlarging stoma and<br />

skin problems.


Infectious complications<br />

EN tubes have been associated with outbreaks of<br />

antimicrobial-resistant organisms. The enteral<br />

feed administration sets become colonized externally<br />

by microbes grown from the enteral tube<br />

hub, and therefore serve as a reservoir of organisms<br />

that can be cross-transmitted. 118–125<br />

Adherence to standard precautions is critical<br />

when handling enteral feeding apparatuses.<br />

Bacterial contamination of the nutritive solution<br />

with enterotoxin-producing bacteria such as<br />

coliforms or Enterobacter cloacae, 122 at the time of<br />

preparation, storage or delivery to the patient,<br />

leads to gastroenteritis and/or septicemia. 123–125<br />

The relationship between bacterial contamination<br />

of enteral formula and diarrhea may be a matter of<br />

debate when EN is delivered into the stomach,<br />

because of the protective role of gastric acid, the<br />

risk of bowel contamination being theoretically<br />

higher when EN is delivered below the pylorus.<br />

Furthermore, in hospitalized patients, bacteriological<br />

monitoring of formula samples is mandatory;<br />

manipulation of the tubing and feed reservoirs<br />

must be careful. Ready-to-use commercial formulas<br />

are therefore recommended for home EN and<br />

in hospitals.<br />

Since necrotizing enterocolitis may occur in<br />

premature infants and neonates suffering from<br />

hypoxia and infections, the abdomen must be<br />

checked daily very carefully. Because of the risk of<br />

infectious complications and necrotizing enterocolitis,<br />

some teams prefer PN to this technique for<br />

premature babies weighing less than 1500g, who<br />

require respiratory assistance. Prevention of such<br />

complications requires the use of the gastric route<br />

as far as possible, strict limitation of duodenal<br />

infusions and safe preparation of solutions.<br />

Metabolic complications<br />

Metabolic complications of EN are rare when<br />

compared to those of PN. However, especially in<br />

severely malnourished children, water overload<br />

or electrolyte imbalance should be avoided by<br />

careful monitoring. In those particular patients,<br />

phosphatemia should be monitored and phosphate<br />

supplementation provided.<br />

Dehydration and hypernatremia may occur in<br />

case of diarrhea or vomiting, which may result<br />

Home enteral nutrition 549<br />

from a formula that is hyperosmolar or too<br />

concentrated. Hypoglycemia may be due to<br />

sudden discontinuation of infusion, especially in<br />

infants, in malnourished children or when EN is<br />

used to prevent hypoglycemia in metabolic<br />

diseases such as glycogenosis. In children on<br />

cyclic EN who do not eat orally, a progressive<br />

decrease in infusion rate is recommended to<br />

prevent hypoglycemia.<br />

Home enteral nutrition<br />

Indications<br />

Home EN is a logical alternative to long-term hospitalization<br />

when long-term EN support is necessary<br />

(more than 1 month) in a patient in stable clinical<br />

condition. Home EN has been shown to be an effective<br />

and safe method, compatible with the best<br />

possible quality of life. 126–129 Major cost savings<br />

induced by home EN as compared to hospitalization<br />

have been demonstrated. The importance of<br />

families’ teaching and medical follow-up to prevent<br />

somatic and psychological complications should<br />

not be underestimated.<br />

Prevalence and incidence<br />

The origins of home EN are much older than those<br />

of home PN. In North America, 20000 patients<br />

received home EN in 1985 and 150000 in 1992<br />

(about 500 per million population). Based on an<br />

estimated growth rate of 25% per year, about<br />

500000 patients may have been on home EN in<br />

the USA in 1997. 130,131 The British Association of<br />

Enteral and Parenteral Nutrition estimated that<br />

the number of patients on home EN was about 40<br />

per million population, about ten-fold the number<br />

of home PN patients in the same country at the<br />

same time. Children accounted for 30–40% of<br />

home EN patients in Britain, compared to about<br />

20% in the USA. 130,132,133 However, the lack of<br />

national registers in most European countries<br />

makes estimation difficult.<br />

Organization<br />

The quality of home EN programs depends on the<br />

organization of multidisciplinary nutrition


550<br />

Enteral nutrition<br />

support teams, based on a tight collaboration<br />

between the different professionals including<br />

physicians, home care nurses, dietitians, pharmacists<br />

and social workers. In some countries,<br />

pumps, disposable equipment and nutrients for<br />

home EN are mainly delivered by hospitals, while<br />

in others, such as the USA, where they are the<br />

most developed, home care companies participate<br />

in patients’ training and provide a delivery<br />

service and a 24-h emergency phone contact.<br />

With expansion of the home care industry, it is<br />

important to ensure that adequate standards of<br />

care are provided. The American Society of<br />

Parenteral and Enteral Nutrition and the British<br />

Association of Parenteral and Enteral Nutrition<br />

have produced such standards. 132,133<br />

Parents’ teaching<br />

Tolerance and efficacy of EN have to be demonstrated<br />

at the hospital before home EN is organized.<br />

As for home PN, home EN in children is feasible<br />

only when the family is highly motivated and able<br />

to deal with technical aspects. Parents’ teaching is<br />

based on a nutrition multidisciplinary hospital team<br />

including a physician, nurse, dietitian and pharmacist.<br />

Parents are taught not only about technical<br />

aspects but also about risks, complications and their<br />

prevention. The hospital team ensures a 24/24h<br />

phone contact and regular follow-up, in close cooperation<br />

with the general practitioner. The help of<br />

a community nurse may be required, especially in<br />

case of placement of a nasogastric tube.<br />

Results<br />

Quality of life<br />

Although home EN, like home PN, is usually<br />

considered by children and families as an<br />

improvement of their quality of life, psychological<br />

consequences of this technique have to be<br />

carefully estimated and prevented. The placement<br />

of a nasogastric tube is distressing to<br />

parents and children. A tube in situ for continuous<br />

24/24h infusion is a major problem, particularly<br />

in older children and adolescents, because it<br />

induces an unwelcome public interest. These<br />

problems are solved by the use of gastrostomy.<br />

Children and families may also suffer from the<br />

suppression of meals taken together, which are<br />

usually considered as an important time in family<br />

life. Adequate psychological preparation and<br />

follow-up improve the tolerance in children and<br />

parents of home EN. 134,135<br />

Outcome<br />

Outcome studies in home EN patients have been<br />

fewer than those in patients on home PN.<br />

Improvement of the nutritional status and low<br />

mortality rate (always related to major underlying<br />

diseases) are usually described in children on<br />

home EN. 136–138 However, few data have been<br />

given about the outcome of such pediatric<br />

patients in the long term.<br />

Cost and funding<br />

Home EN is far from being as expensive as home<br />

PN. The total cost per day of home EN was about<br />

$35 in the USA in 1992, when the cost per day of<br />

home PN was about $300. 131 The cost savings<br />

associated with providing home EN estimated by<br />

the British Association for Parenteral and Enteral<br />

Nutrition in 1994 were about 70%. 126 In most<br />

countries, patients on home EN are funded by the<br />

National Health Service, although in the USA<br />

they are mostly paid for by insurance companies.<br />

130,139


REFERENCES<br />

1. Sermet-Gaudelus I, Poisson-Salomon AS, Colomb V et<br />

al. Simple pediatric nutritional risk score to identify<br />

children at risk of malnutrition. Am J Clin Nutr 2000;<br />

72: 64–70.<br />

2. Ricour C, Duhamel JF, Nihoul-Fekete C. Nutrition<br />

entérale à débit constant chez l’enfant – techniques et<br />

indications dans 170 cas de 1969 à 1975. Arch Fr Pediatr<br />

1977; 34: 154–170.<br />

3. American Society for Parenteral and Enteral Nutrition<br />

(ASPEN). Board of Directors. Guidelines for the use of<br />

enteral nutrition in the adult patient. J Enteral Parent<br />

Nutr 1987; 11: 435–439.<br />

4. Sogni P, Vidon N, Huchet B, Bernier JP. Gastric and<br />

duodeno-pancreatic functions during naso-gastric<br />

feeding in man: effect of infusion rate. Gastroenterology<br />

1991; 98: 344 (abstr).<br />

5. Meyer JE. Motility of the stomach and gastroduodenal<br />

function. In Johnson LR, ed. Physiology of the<br />

Gastrointestinal Tract. New York: Raven Press, 1987:<br />

613–629.<br />

6. Hunt JN, Knox MT. A relation between the chain length<br />

of fatty acids and the slowing of gastric emptying. J<br />

Physiol 1968; 194: 327–336.<br />

7. Montoya ML, Carriere D, Hebuterne X et al. Motricité<br />

duodénale au cours de l’alimentation entérale à débit<br />

constant. Gastroenterol Clin Biol 1988; 12: 143 (abstr).<br />

8. Ducrotte P. Motricité digestive au cours de la nutrition<br />

entérale. Ann Gastroenterol Hepatol 1988; 24: 277–280.<br />

9. Ducrotte P, Chayvialle JA, Lerebours E et al. Effects of<br />

intravenous and intraduodenal fat on jejunal motility<br />

and on plasma cholecystokinin in man. Dig Dis Sci<br />

1988; 33: 558–564.<br />

10. Douard H, Cosnes J, Sebog A et al. Etude échographique<br />

de la motricité vésiculaire au cours de l’alimentation<br />

entérale continue exclusive. Gastroenterol Clin Biol<br />

1987; 11: 643–647.<br />

11. Pet I, Ladas S, Forgacs IC et al. Comparison of effects of<br />

ingested medium- and long-chain triglyceride on gallbladder<br />

volume and release of cholecystokinin and<br />

other gut peptides. Dig Dis Sci 1987; 32: 481–486.<br />

12. Taylor IL, Byrne WJ, Christie DL et al. Effect of individual<br />

L-amino acids on gastric acid secretion and serum<br />

gastric and pancreatic polypeptide release in humans.<br />

Gastroenterology 1982; 83: 273–275.<br />

13. Grant JP, Dabey MC, Crae J, Snyder PJ. Effect of enteral<br />

nutrition on human pancreatic secretions. J Parenter<br />

Enteral Nutr 1987; 11: 302–308.<br />

14. Vidon N, Chaussade S, Merite F et al. Inhibitory effect<br />

of high caloric load of carbohydrates or lipids on<br />

human pancreatic secretions: a jejunal brake. Am J Clin<br />

Nutr 1989; 50: 231–236.<br />

15. Freeman JB, Fairfull-Smith RJ. Current concepts of<br />

enteral feeding. In Advances in Enteral Feeding.<br />

Kingsport, TN: Year Book Medical Publishers, 1983; 16:<br />

75–112.<br />

16. Monn CL, Ling V, Bourassa D. Small intestinal and<br />

colonic changes induced by a chemically defined diet.<br />

Dig Dis Sci 1980; 25: 123–128.<br />

17. Heymsfield SB, Casper K, Grossman GD. Bioenergetic<br />

and metabolic response to continuous versus intermittent<br />

nasoenteric feeding. Metabolism 1987; 36: 570–575.<br />

18. Nacht CA, Schutz Y, Vernet O et al. Continuous versus<br />

single bolus enteral nutrition: comparison of energy metabolism<br />

in humans. Am J Physiol 1986; 251: E524–E529.<br />

19. Leblanc J, Cabanac M, Samson P. Reduced post prandial<br />

heat production with gavage as compared with meal<br />

References 551<br />

feeding in human subjects. Am J Physiol 1984; 246:<br />

E95–E101.<br />

20. Ricour C, Duhamel JF, Arnaud-Battandier F et al. Enteral<br />

and parenteral nutrition in the short bowel syndrome in<br />

children. World J Surg 1985; 9: 310–315.<br />

21. Goulet O, Revillon Y, Jan D et al. <strong>Neonatal</strong> short bowel<br />

syndrome. J Pediatr 1991; 119: 18–23.<br />

22. Galea MH, Holliday H, Carachi R, Kapila L. Short bowel<br />

syndrome: a collective review. J Pediatr Surg 1992; 27:<br />

592–596.<br />

23. Vanderhoof JA, Langnas A, Pinch L et al. Short bowel<br />

syndrome. J Pediatr Gastroenterol Nutr 1992; 14:<br />

359–370.<br />

24. Sondheimer J, Cadnapaphornchai M, Sontag M, Zerbe<br />

GO. Predicting the duration of dependence on<br />

parenteral nutrition after neonatal intestinal resection. J<br />

Pediatr 1998; 132: 80–84.<br />

25. Kaufman SS. Prevention of parenteral nutrition-associated<br />

liver disease in children. Pediatr Transplant 2002;<br />

6: 37–42.<br />

26. Avery GB, Villavicencio O, Lilly JR, Randolph JG.<br />

Intractable diarrhea of infancy. Pediatrics 1968; 41:<br />

712–722.<br />

27. Goulet O, Brousse N, Canioni D et al. Syndrome of<br />

intractable diarrhea with persistent villous atrophy in<br />

early childhood: a clinicopathological survey of 47<br />

cases. J Pediatr Gastroenterol Nutr 1998; 26: 151–161.<br />

28. Guarino A, Spagnulo MI, Russo S et al. Etiology and<br />

risk factors of severe and protracted diarrhea. J Pediatr<br />

Gastroenterol Nutr 1995; 20: 173–178.<br />

29. Brasseur D, Goyens P. Enteral nutrition for therapy of<br />

chronic diarrhea. Diarrheal Dis 1997; 38: 289–316.<br />

30. Guandalini S. Prolonged diarrhea: etiology and pathogenesis.<br />

Diarrheal Dis 1997; 38: 153–170.<br />

31. Weizman Z, Schmueli A, Deckelbaum RJ. Continuous<br />

nasogastric drip elemental feeding: alternative for<br />

prolonged parenteral nutrition in severe prolonged diarrhea.<br />

Am J Dis Child 1983; 137: 253–255.<br />

32. Orenstein SR. Enteral versus parenteral therapy for<br />

intractable diarrhea of infancy: a prospective, randomized<br />

trial. J Pediatr 1986; 109: 277–286.<br />

33. Vanderhoof J, Murray ND, Kaufman SS et al. Intolerance<br />

to protein hydrolysate infant formulas: an under recognized<br />

cause of gastrointestinal symptoms in infants. J<br />

Pediatr 1997; 131: 741–744.<br />

34. Morin CL, Roulet M, Roy CC, Lapointe N. Continuous<br />

elemental enteral alimentation in the treatment of children<br />

and adolescents with Crohn’s disease. J Parenter<br />

Enteral Nutr 1982; 6: 194–199.<br />

35. O’Morain C, Segal AW, Levi AJ. Elemental diet as<br />

primary treatment of acute Crohn’s disease: a controlled<br />

trial. BMJ 1984; 288: 1859–1862.<br />

36. Seidman EG, Roy CC, Weber AM, Morin CL. Nutritional<br />

therapy of Crohn’s disease in childhood. Dig Dis Sci<br />

1987; 32 (Suppl): 82S–88S.<br />

37. Sanderson IR, Udeen S, Davies PSW et al. Remission<br />

induced by an elemental diet in small bowel Crohn’s<br />

disease. Arch Dis Child 1987; 61: 123–127.<br />

38. Rigaud D, Cosnes J, Le Quintrec Y et al. Controlled trial<br />

comparing two types of enteral nutrition in treatment of<br />

active Crohn’s disease: elemental vs polymeric diet. Gut<br />

1991; 32: 1492–1497.<br />

39. Belli DC, Seidman E, Bouthillier L et al. Chronic intermittent<br />

elemental diet improves growth failure in children<br />

with Crohn’s disease. Gastroenterology 1988; 94:<br />

603–610.


552<br />

Enteral nutrition<br />

40. Thomas AG, Taylor F, Miller V. Dietary intake and nutritional<br />

treatment in childhood Crohn’s disease. J Pediatr<br />

Gastroenterol Nutr 1993; 17: 75–81.<br />

41. Ruuska T, Savilahti E, Maki M et al. Exclusive whole<br />

protein enteral diet versus prednisolone in the treatment<br />

of active Crohn’s disease in children. J Pediatr<br />

Gastroenterol Nutr 1994; 19: 175–180.<br />

42. Beattie RM, Schiffin EJS, Donnet-Hughes A et al.<br />

Polymeric nutrition as the primary therapy in children<br />

with small bowel Crohn’s disease. Aliment Pharmacol<br />

Ther 1994; 8: 609–615.<br />

43. Breese EJ, Michie CA, Nicholls SW et al. The effect of<br />

treatment of lymphokine-secreting cells in the intestinal<br />

mucosa of children with Crohn’s disease. Aliment<br />

Pharmacol Ther 1995; 9: 547–553.<br />

44. Griffiths AM, Ohlsson A, Sherrnan PM, Sutherland LR.<br />

Meta-analysis of enteral nutrition as a primary treatment<br />

of active Crohn’s disease. Gastroenterology 1995;<br />

108: 1056–1067.<br />

45. Heuschkel RB, Menache CC, Megerian JT, Baird AE.<br />

Enteral nutrition and corticosteroids in the treatment of<br />

active Crohn’s disease in children. J Pediatr<br />

Gastroenterol Nutr 2000; 31: 8–15.<br />

46. Zachos M, Tondeur M, Griffiths AM. Enteral nutritional<br />

therapy for inducing remission of Crohn’s disease.<br />

Cochrane Database Syst Rev 2001; 3: CD000542.<br />

47. Fell JM, Paintin M, Arnaud-Battandier F et al. Mucosal<br />

healing and a fall in mucosal pro-inflammatory cytokine<br />

mRNA induced by a specific oral polymeric diet in<br />

paediatric Crohn’s disease. Aliment Pharmacol Ther<br />

2000; 14: 281–289.<br />

48. Murch S, Walker-Smith JW. Nutrition in inflammatory<br />

bowel disease. Baillière’s Clin Gastroenterol 1998; 122:<br />

719–738.<br />

49. Akobeng AK, Miller V, Stanton J et al. Double-blind<br />

randomized controlled trial of glutamine-enriched polymeric<br />

diet in the treatment of active Crohn’s disease. J<br />

Pediatr Gastroenterol Nutr 2000; 30: 78–84.<br />

50. Ruemmele FM, Roy CC, Levy E, Seidman EG. Nutrition<br />

as primary therapy in pediatric Crohn’s disease: fact or<br />

fantasy? J Pediatr 2000; 136: 285–291.<br />

51. Colomb V, Goulet O, Gorski AM et al. Forme digestive<br />

sévère de purpura rhumatoïde. Etude rétrospective de<br />

19 observations chez l’enfant. Arch Fr Pediatr 1990; 47:<br />

9–12.<br />

52. Goulet O, Jobert-Giraud A, Michel JL et al. Chronic<br />

intestinal pseudoobstruction syndrome in pediatric<br />

patients. Eur J Pediatr Surg 1999; 9: 83–89.<br />

53. Faure C, Goulet O, Ategbo S et al. Chronic intestinal<br />

pseudoobstruction syndrome: clinical analysis, outcome<br />

and prognosis in 105 children. Dig Dis Sci 1999; 44:<br />

953–959.<br />

54. Heneyke S, Smith VV, Spitz L, Milla PJ. Chronic intestinal<br />

pseudo-obstruction : treatment and long term follow<br />

up of 44 patients. Arch Dis Child 1999; 81: 21–27.<br />

55. Turck D, Michaud L. Cystic fibrosis: nutritional consequences<br />

and management. Baillières Clin Gastroenterol<br />

1998; 12: 805–822.<br />

56. Bertrand JM, Morin CL, Lasalle R et al. Short term clinical,<br />

nutritional and functional effect of continuous<br />

elemental enteral alimentation in children with cystic<br />

fibrosis. J Pediatr 1984; 104: 41–46.<br />

57. Boland MP, Stoski D, Macdonald N et al. Chronic<br />

jejunostomy feeding with a non-elemental formula in<br />

undernourished patients with cystic fibrosis. Lancet<br />

1986; 1: 232–234.<br />

58. Shephard RW, Holt TL, Thomas BJ et al. Nutritional<br />

rehabilitation in cystic fibrosis, controlled studies of<br />

effects on nutritional and growth retardation, body<br />

protein turnover and course of pulmonary disease. J<br />

Pediatr 1986; 109: 788–794.<br />

59. Gaskin K, Allen J. Exocrine pancreatic disease including<br />

cystic fibrosis. In Walker A, Watkins J, Duggan C, eds.<br />

Nutrition in Pediatrics. Basic Science and Clinical<br />

Applications. London: BC Decker, 2003: 671–685.<br />

60. Steinkamp G, Von Der Hardt H. Improvement of nutritional<br />

status and lung function after long-term nocturnal<br />

gastrostomy feeding in cystic fibrosis. J Pediatr 1994;<br />

124: 244–249.<br />

61. Goulet OJ, De Ville De Goyet J, Otte JB, Ricour C.<br />

Preoperative nutritional evaluation and support for liver<br />

transplantation in children. Transplant Proc 1987; 19:<br />

3249–3255.<br />

62. Kaufmann SS, Murray ND, Wood RP et al. Nutritional<br />

support for the infant with extra hepatic biliary atresia.<br />

J Pediatr 1987; 110: 679–686.<br />

63. Protheroe SM, Kelly DA. Cholestasis and end-stage liver<br />

disease. Baillières Clin Gastroenterol 1998; 12: 823–841.<br />

64. Moreno LA, Gottrand F, Hoden S et al. Improvement of<br />

nutritional status in cholestatic children with supplemental<br />

nocturnal enteral nutrition. J Pediatr<br />

Gastroenterol Nutr 1991; 2: 213–216.<br />

65. Moukarzel AA, Najm I, Vargas J et al. Effect of nutritional<br />

status on outcome of orthotopic liver transplantation<br />

in pediatric patients. Transplant Proc 1990; 22:<br />

1560–1563.<br />

66. Campos AC, Matias JE, Coelho JC. Nutritional aspects of<br />

liver transplantation. Curr Opin Clin Nutr Metab Care<br />

2002; 5: 297–307.<br />

67. Sokal EM. Nutritional and medical care in chronic<br />

cholestasis. In Buts JP, Sokal EM, eds. Management of<br />

Digestive and Liver Disorders in Infants and Children.<br />

Amsterdam: Elsevier, 1993: 537–542.<br />

68. Kaufmann SS, Scrivner DJ, Murray ND et al. Influence<br />

of Portagen and Pregestimil on essential fatty acid status<br />

in infantile liver disease. Pediatrics 1992; 89: 151–154.<br />

69. Sokal EM, Baudoux MC, Collette E et al. Branched<br />

chain amino acids improve body composition and nitrogen<br />

balance in a rat model of extra-hepatic biliary<br />

atresia. Pediatr Res 1996; 40: 66–71.<br />

70. Haynes L, Atherton DJ, Ade-Ajayi N et al. Gastrostomy<br />

and growth in dystrophic epidermolysis bullosa. Br J<br />

Dermatol 1996; 134: 872–879.<br />

71. Fung EB, Samson-Fang L, Stallings VA et al. Feeding<br />

dysfunction is associated with poor growth and health<br />

status in children with cerebral palsy. J Am Diet Assoc<br />

2002; 102: 361–373.<br />

72. Samson-Fang L, Butler C, O'Donnell M. Effects of<br />

gastrostomy feeding in children with cerebral palsy: an<br />

AACPDM evidence report. Dev Med Child Neurol 2003;<br />

45: 415–426.<br />

73. Wales PW, Diamond IR, Dutta S et al. Fundoplication<br />

and gastrostomy versus image-guided gastrojejunal tube<br />

for enteral feeding in neurologically impaired children<br />

with gastresophageal reflux. J Pediatr Surg 2002; 37:<br />

407–412.<br />

74. Briassoulis G, Zavras N, Hatzis T. Malnutrition, nutritional<br />

indices, and early enteral feeding in critically ill<br />

children. Nutrition 2001; 17: 548–557.<br />

75. Donaldson SS, Jundt S, Ricour C et al. Radiation enteritis<br />

in children. Cancer 1975; 35: 1167–1178.<br />

76. Mauer AM, Burgess JB, Donaldson SS et al. Special<br />

nutrition needs of children with malignancies: a review.<br />

J Parenter Enteral Nutr 1990; 14: 315–324.<br />

77. Deswarte-Wallace J, Firouzbakhsh S, Finklestein JZ.<br />

Using research to change practice: enteral feedings for<br />

pediatric oncology patients. J Pediatr Oncol Nurs<br />

2001;18: 217–223.


78. Pietsch JB, Ford C, Whitlock JA. Nasogastric tube feedings<br />

in children with high-risk cancer: a pilot study. J<br />

Pediatr Hematol Oncol 1999; 21: 111–114, Comment<br />

88–90.<br />

79. Wasmer SJ, Abitbol C, Alexander S et al. Nutritional<br />

requirements for infants with renal failure. Am J Kidney<br />

Dis 1986, 7: 300–305.<br />

80. Kleinknecht C, Laouari D, Burtin M et al. Contribution<br />

of experimental studies on the nutritional management<br />

of children with chronic renal failure. Pediatr Nephrol<br />

1991; 5: 487–491.<br />

81. Salzer HR, Haschke F, Wimmer M et al. Growth and<br />

nutritional intake of infants with congenital heart<br />

disease. Pediatr Cardiol 1989; 10: 17–23.<br />

82. Bougle D, Iselin M, Kahyat A, Duhamel JF. Nutritional<br />

treatment of congenital heart disease. Arch Dis Child<br />

1986; 61: 799–801.<br />

83. Jouvet P, Jugie M, Rabier D et al. Combined nutritional<br />

support and continuous extracorporeal removal therapy<br />

in the severe acute phase of maple syrup urine disease.<br />

Intensive Care Med 2001; 27: 1798–1806.<br />

84. Greene HL, Slonim AE, Barr IA, Moran JR. Type 1 glycogen<br />

storage disease. Five years of management with<br />

nocturnal intragastric feeding. J Pediatr 1980; 96:<br />

590–595.<br />

85. Fernandes J, Alaupovic P, Wit JM. Gastric drip feeding<br />

in patients with glycogen storage disease type I: its<br />

effects on growth and plasma lipids and apolipoproteins.<br />

Pediatric Res 1989; 25: 327–331.<br />

86. Collins JE, Leornard JV. The dietary management of<br />

inborn errors of metabolism. Hum Nutr Appl Nutr 1985,<br />

39A: 255–272.<br />

87. Stanbury JB, Wyngaarden JB, Fredrickson DS et al. The<br />

Metabolic Basis of Inherited Disease, 5th edn. New York:<br />

McGraw Hill.<br />

88. Franck DA, Ziesel SH. Failure to thrive. Pediatr Clin<br />

North Am 1988; 35: 1187–1206.<br />

89. Bithoney WG, McJunkin JJ, Michalek J et al. The effect<br />

of a multidisciplinary team approach on weight gain in<br />

monoorganic failure–to-thrive children. J Dev Behav<br />

Pediatr 1991; 12: 254–258.<br />

90. Fosson A, Knibbs J, Bryant-Waugh R, Lask B. Early onset<br />

anorexia nervosa. Arch Dis Child 1987; 62: 114–118.<br />

91. de Tournemire R, Alvin P. Anorexia nervosa and severe<br />

malnutrition: nutritional practice guidelines in pediatrics.<br />

Arch Pediatr 2002; 9: 429–433.<br />

92. Mamel JJ. Percutaneous endoscopic gastrostomy. Am J<br />

Gastroenterol 1989; 84: 703–710.<br />

93. Ho CS, Yee ACN, McPherson R. Complications of surgical<br />

and percutaneous non endoscopic gastrostomy:<br />

review of 233 patients. Gastroenterology 1988, 95:<br />

1206–1210.<br />

94. Anderson C. Enteral feeding: a change in practice. J<br />

Child Health Care 2000; 4: 160–162.<br />

95. Segal D, Michaud L, Guimber D et al. Late-onset complications<br />

of percutaneous endoscopic gastrostomy in children.<br />

J Pediatr Gastroenterol Nutr 2001; 33: 495–500.<br />

96. Gauderer MW. Percutaneous endoscopic gastrostomy<br />

and the evolution of contemporary long-term enteral<br />

access. Clin Nutr 2002; 21: 103–110.<br />

97. Doede T, Faiss S, Schier F. Jejunal feeding tubes via<br />

gastrostomy in children. Endoscopy 2002; 34: 539–542.<br />

98. Carrillo EH, Heniford BT, Osborne DL et al. Bedside<br />

percutaneous endoscopic gastrostomy. A safe alternative<br />

for early nutritional support in critically ill trauma<br />

patients. Surg Endosc 1997; 11: 1068–1071.<br />

99. Grimble GK, Rees RG, Keohane PP et al. Effect of<br />

peptide chain length on absorption of egg protein<br />

hydrolysates in the normal human jejunum.<br />

Gastroenterology 1987; 92: 136–142.<br />

References 553<br />

100. Silk DBA, Fairclough PO, Clark ML et al Uses of a<br />

peptide rather than a free amino-acid nitrogen source in<br />

chemically defined elemental diets. J Parenter Enteral<br />

Nutr 1980; 4: 548–553.<br />

101. Fairclough PD, Hegarty JE, Silk DBA, Clark ML.<br />

Comparison of the absorption of two protein<br />

hydrolysates and their effects on water and electrolyte<br />

movements in the human jejunum. Gut 1980; 21:<br />

829–834.<br />

102. Poullain MG, Cezard JP, Roger L, Mendy F. Effect of<br />

whey proteins, their oligopeptide hydrolysates and free<br />

amino acid mixtures on growth and nitrogen in fed and<br />

starved rats. J Parenter Enteral Nutr 1989; 13: 382–386.<br />

103. Burrin DG, Stoll B. Key nutrients and growth factors for<br />

the neonatal gastrointestinal tract. Clin Perinatol 2002;<br />

29: 65–96.<br />

104. Bach AC, Babayan VK. Medium–chain triglycerides: an<br />

update. Am J Clin Nutr 1982; 36: 950–962.<br />

105. Ingenbleek Y. Les triglycérides à chaînes moyennes en<br />

nutrition clinique. Nutr Clin Metab 1989; 3: 3–15.<br />

106. Lima LAM, Gray OP, Losty H. Excretion of dicarboxylic<br />

acids following administration of medium chain triglycérides.<br />

J Parenter Enteral Nutr 1987; 11: 600–601.<br />

107. Bines J, Francis D, Hill D. Reducing parenteral requirement<br />

in children with short bowel syndrome: impact of<br />

an amino acid-based complete infant formula. J Pediatr<br />

Gastroenterol Nutr 1998; 26: 123–128.<br />

108. Andorsky DJ, Lund DP, Lillehei CW et al. Nutritional<br />

and other postoperative management of neonates with<br />

short bowel syndrome correlates with clinical<br />

outcomes. J Pediatr 2001; 139: 27–33.<br />

109. Berbaum JC, Pereira GR, Watkins JB, Peckham GJ. Non<br />

nutritive sucking during gavage feeding enhances<br />

growth and maturation in premature infants. Pediatrics<br />

1983; 71: 41–45.<br />

110. Widstrom AM, Marchini G, Matthiesen AS et al. Non<br />

nutritive sucking in tube-fed preterm infants: effects on<br />

gastric motility and gastric contents of somatostatin. J<br />

Pediatr 1988; 7: 517–523.<br />

111. American Gastroenterological Association Patient Care<br />

Committee. American Gastroenterological Association<br />

technical review on tube feeding for enteral nutrition.<br />

Gastroenterology 1995; 108: 1282–1301.<br />

112. Aspen Board of Directors and the Clinical Guidelines<br />

Task Force. Guidelines for the use of parenteral and<br />

enteral nutrition in adult and pediatric patients. J<br />

Parenter Enteral Nutr 2002; 26(1 Suppl): 1SA–138SA.<br />

113. Iyer PU. Nutritional support in the critically ill child.<br />

Indian J Pediatr 2002; 69: 405–410.<br />

114. Gorman SR, Armstrong G, Allen KR et al. Scarcity in<br />

the midst of plenty: enteral tube feeding complicated by<br />

scurvy. J Pediatr Gastroenterol Nutr 2002; 35: 93–95.<br />

115. Perez-Rodriguez J, Quero J, Frias EG, Omenaca F.<br />

Duodenal perforation in a neonate by a tube of silicone<br />

rubber during trans<strong>pylori</strong>c feeding. J Pediatr 1978; 92:<br />

113–114.<br />

116. Hand RW, Kempster M, Levy JH et al. Inadvertent transbronchial<br />

insertion of narrow bore feeding tubes into<br />

the pleural space. JAMA 1984; 251: 2396–2397.<br />

117. Boros SJ, Reynolds JW. Duodenal perforation: a complication<br />

of neonatal nasojejunal feeding. J Pediatr 1974;<br />

85: 107–108.<br />

118. Anderson KR, Norris DJ, Godfrey LB et al. Bacterial<br />

contamination of tube-feeding formulas. J Parenter<br />

Enteral Nutr 1984; 8: 673–678.<br />

119. Casewell MW, Cooper JE, Webster M. Enteral feeds contaminated<br />

with Enterobacter cloacae as a cause of septicaemia.<br />

BMJ 1981; 282: 973.<br />

120. De Leeuw IH, Vandewoude MF. Bacterial contamination<br />

of enteral diets. Gut 1986; 27: 56–57.


554<br />

Enteral nutrition<br />

121. Goulet O, Duhamel JF, Ricour C. Nutritional problems.<br />

In Tinker J, Zapol WM, eds. Care of the Critically Ill<br />

Patient. Berlin: Springer Verlag, 1992: 1415–1436.<br />

122. Patchell CJ, Anderton A, Macdonald A et al. Bacterial<br />

contamination of enteral feeds. Arch Dis Child 1994; 70:<br />

327–330.<br />

123. Mehall JR, Kite CA, Gilliam CH et al. Enteral feeding<br />

tubes are a reservoir for nosocomial antibiotic-resistant<br />

pathogens. J Pediatr Surg 2002; 37: 1011–1012.<br />

124. Matlow A, Wray R, Goldman C et al. Microbial contamination<br />

of enteral feed administration sets in a pediatric<br />

institution. Am J Infect Control 2003; 31: 49–53.<br />

125. Mehall JR, Kite CA, Saltzman DA et al. Prospective<br />

study of the incidence and complications of bacterial<br />

contamination of enteral feeding in neonates. J Pediatr<br />

Surg 2002; 37: 1177–1182.<br />

126. Greene HL, Helinek GL, Folk CC et al. Nasogastric tube<br />

feeding at home: a method for adjunctive nutritional<br />

support of malnourished patients. Am J Clin Nutr 1981;<br />

34: 1131–1138.<br />

127. Reeves-Garcia J, Heyman MB. A survey of complications<br />

of pediatric home enteral tube feedings and<br />

discussion of developmental and psychosocial issues.<br />

Nutrition 1988; 4: 375–377.<br />

128. Aiges H, Markowitz J, Rosa J, Daum F. Home nocturnal<br />

supplemental nasogastric feedings in growth–retarded<br />

adolescents with Crohn’s disease. Gastroenterology<br />

1989; 97: 905–910.<br />

129. Claris-Appiani A, Ardissino GL, Dacco V et al. Catch-up<br />

growth in children with chronic renal failure treated<br />

with long-term enteral nutrition. J Parenter Enteral Nutr<br />

1995; 19: 175–178.<br />

130. Elia M. An international perspective on artificial nutritional<br />

support in the community. Lancet 1995; 345:<br />

1345–1349.<br />

131. Howard L, Ament M, Fleming CR et al. Current use and<br />

clinical outcome of home parenteral and enteral nutrition<br />

therapies in the United States. Gastroenterology<br />

1995; 109: 355–365.<br />

132. Elia M. Home enteral nutrition: some general aspects<br />

and comparison between the USA and Britain. Nutrition<br />

1994; 10: 1–9.<br />

133. Parker T, Neale, Elia M. Home enteral tube feeding in<br />

East Anglia. Eur J Clin Nutr 1996; 50: 47–53.<br />

134. Holden CE, Puntis JWL, Charlton CPL, Booth IW.<br />

Nasogastric feeding at home: acceptability and safety.<br />

Arch Dis Child 1991; 66: 148–151.<br />

135. Holden CE, Macdonald A, Ward M et al. Psychological<br />

preparation for nasogastric feeding in children. Br J<br />

Nurs 1997; 6: 376–385.<br />

136. Ganga-Zanzou PS, Hankard R, Gottrand F et al.<br />

Nutrition entérale à domicile chez l’enfant et l’adolescent.<br />

Expérience à propos de 79 patients. Ann Pediatr<br />

1995; 42: 295–231.<br />

137. Liptak GS. Home care for children who have chronic<br />

conditions. Pediatr Rev 1997; 18: 271–273.<br />

138. Marin OE, Glassman MS, Schoew BT, Caplan DB. Safety<br />

and efficacy of percutaneous endoscopic gastrostomy in<br />

children. Am J Gastroenterol 1994; 98: 357–360.<br />

139. Crocker KS. Current status of home infusion therapy.<br />

Nutr Clin Pract 1992; 7: 256–263.


34<br />

Introduction<br />

Parenteral nutrition in infants<br />

and children<br />

Olivier Goulet and Virginie Colomb<br />

In the late 1960s in the USA, Dudrick and Wilmore<br />

demonstrated that long-term administration of<br />

hypertonic nutrient infusates through a catheter<br />

inserted into the superior vena cava was feasible,<br />

with immediate dilution of the hypertonic solution,<br />

preventing damage to the vein. 1,2 By infusing a<br />

hypertonic mixture of glucose, protein hydrolysate,<br />

electrolytes, minerals and vitamins, they proved<br />

that it was possible to achieve normal growth and<br />

development in an infant with multiple smallbowel<br />

atresia. 1,2 Indeed, parenteral nutrition (PN),<br />

since its introduction in clinical practice during the<br />

late 1960s has enabled many patients to survive and<br />

has reduced the incidence of malnutrition related to<br />

a variety of digestive and non-digestive diseases.<br />

With antibiotics, antitumoral chemotherapy and<br />

organ transplantation, PN can be considered as one<br />

of the most important therapeutic advances over the<br />

past 50 years since it has demonstrated its potential<br />

for changing the course of both medical and surgical<br />

patients.<br />

Technical aspects, psychological consequences of<br />

feeding ‘deprivation’ and a high rate of complications<br />

led to suspicion of this therapeutic strategy at<br />

the early phase of its development. However,<br />

continuous clinical and basic research has allowed<br />

improvements in its efficiency and reduction in the<br />

rate of complications. PN is now widely used in a<br />

variety of indications as reviewed below. PN<br />

provides macro- and micronutrients and calories<br />

directly through the systemic circulation. Although<br />

such delivery short circuits the portal circulation,<br />

macronutrients such as lipids, glucose and amino<br />

acids, and micronutrients, can be infused in high<br />

concentrations without causing harmful effects and<br />

these are sufficient for nutritional maintenance and<br />

normal growth in children. Nevertheless, in some<br />

situations, such as those encountered in neonatol-<br />

ogy, gastroenterology or hematology, PN could be<br />

better adapted to the clinical condition by providing<br />

specific nutrients. This chapter will briefly review<br />

the indications, modalities, intakes and complications<br />

of PN for infants, children and adolescents. A<br />

specific chapter in this book is dedicated to<br />

parenteral nutrition in the newborn. Many references<br />

to reviews are provided and the reader is<br />

invited to refer to these for further details.<br />

Indications of parenteral nutrition<br />

Digestive and extradigestive indications<br />

PN is indicated in children for prevention or treatment<br />

of malnutrition, whatever its cause, as soon as<br />

it becomes impossible to use the enteral route. The<br />

range of PN indications, related both to digestive<br />

and non-digestive diseases (Table 34.1), has greatly<br />

increased during the last few years. However,<br />

because of the iatrogenic risk of the technique, especially<br />

when its use is prolonged, each indication<br />

should be discussed while PN should be, as often as<br />

possible, performed in specialized units, such as<br />

intensive care, hematology–oncology or gastroenterology<br />

units.<br />

Gastrointestinal (GI) indications for PN can be<br />

subdivided into three main situations: malabsorption<br />

syndromes, 3–5 indications of bowel rest (e.g.<br />

Crohn’s disease), 6,7 and congenital or acquired<br />

neonatal pathology of the GI tract. Short-bowel<br />

syndrome is the oldest and one of the most classic<br />

indications for long-term PN. 8–13 Some congenital or<br />

acquired metabolic diseases, such as severe hepatic<br />

or renal failure with Gl complications may benefit<br />

from PN with appropriate amino acid solutions. An<br />

increasing area of use of PN is represented by the<br />

management of children with malignant disease.<br />

555


556<br />

Parenteral nutrition in infants and children<br />

Table 34.1 Indications for parenteral<br />

nutrition<br />

Digestive indications<br />

Malabsorption medical and/or surgical<br />

Protracted and intractable diarrhea<br />

Short-bowel syndrome<br />

Enterocutaneous fistula<br />

Proximal enterostomy<br />

Intestinal bacterial overgrowth<br />

Immune deficiency<br />

Indication of bowel rest<br />

Inflammatory bowel disease (Crohn’s disease,<br />

ulcerative colitis, unidentified colitis)<br />

Necrotizing enterocolitis<br />

Intestinal lymphangiectasy<br />

Acute pancreatitis<br />

Systemic disease: Schönlein–Henoch, periarteritis<br />

Radiation enteritis<br />

Congenital or acquired neonatal pathology of the<br />

GI tract<br />

Gastroschisis<br />

Omphalocele<br />

Meconial ileus<br />

Extensive small-bowel resection<br />

Necrotizing enterocolitis<br />

Complicated Hirschsprung’s disease<br />

Chronic intestinal pseudo-obstruction syndrome<br />

Extradigestive indications<br />

Neonatology: premature baby


is the only way in which PN can be delivered at<br />

home.<br />

Home parenteral nutrition<br />

Home PN is the only alternative to prolonged hospitalization<br />

for patients requiring long-term PN. 32–36<br />

Such patients certainly need to be managed by a<br />

specialized center, from which a home PN program<br />

can be organized. 37 Nutrition is generally mixed<br />

parenteral and enteral, but it may be exclusively<br />

parenteral, lasting for months or years. Indications<br />

include total or subtotal resection of the small<br />

bowel; chronic intestinal pseudo-obstruction<br />

syndromes; intractable diarrhea caused by refractory<br />

atrophy of the intestinal mucosa with severe<br />

and persistent malabsorption, either alone or in<br />

association with an immune deficiency; and some<br />

cases of inflammatory bowel diseases (IBDs), especially<br />

severe Crohn’s disease, either extensive<br />

and/or with multiple surgery, with growth retardation<br />

or that have not responded to other therapy.<br />

The objective of long-term PN in these cases is to<br />

ensure normal growth of the child while the inflammatory<br />

syndrome subsides or while waiting for the<br />

residual intestinal condition to become stable.<br />

Whatever the duration of PN and the prognosis of<br />

the disease, home PN offers the patients and their<br />

families the best possible quality of life. 37 In addition,<br />

home PN reduces the cost of care, when<br />

compared to prolonged hospitalization. 36 Cyclic<br />

infusion, adequate and appropriate provision of<br />

macro- and micronutrients, and improvements in<br />

central venous catheters (CVCs) and nutritional<br />

mixtures have all played a major role in making<br />

home PN possible. The development of an infrastructure<br />

for patient training and follow-up, a PNsolution<br />

compounding unit, and an efficient domiciliary<br />

delivery service are required to organize<br />

home PN. Over the past 25 years, more than 300<br />

children treated at Necker-Enfants Malades<br />

Hospital have been discharged on home PN. Most<br />

home PN indications were primary digestive<br />

diseases: short-bowel syndrome accounted for 47%<br />

of the indications, the other main indications being<br />

IBD (11%), chronic intestinal pseudo-obstruction<br />

syndrome (10%) and intractable diarrhea of infancy<br />

(8%). Fifty-four per cent of the patients have been<br />

weaned from PN, 26% are still on home PN, and<br />

16% have died. Survival rates at 2 and 10 years<br />

Parenteral nutrition in clinical practice 557<br />

were 97% and 81%, respectively. Most of the PNrelated<br />

complications were from infection, with<br />

about two CVC-related infection for 1000 home PN<br />

days (Colomb et al. Submitted for publication).<br />

However, the most life-threatening home PN-related<br />

complication is liver disease. The French results, as<br />

well as those of other teams (Boston), suggest that<br />

home PN is the best option for children in need of<br />

long-term PN.<br />

Ethical dilemmas may arise for children with extensive<br />

GI resection, microvillous atrophy or persistent<br />

villous atrophy who are never able to tolerate full<br />

enteral feeding. In such patients, in whom intestinal<br />

autonomy cannot be achieved, intestinal transplantation<br />

may represent the only alternative to lifelong<br />

PN. 38–41 Indications for intestinal transplantation<br />

are not only extreme short-bowel syndromes but<br />

also all situations in which the small intestine is<br />

unable to achieve nutritional requirements; this<br />

includes intractable diarrhea of infancy (chronic<br />

villous atrophy, microvillus inclusion disease) or<br />

severe motility disorders such as chronic intestinal<br />

pseudo-obstruction syndrome.<br />

Parenteral nutrition in clinical<br />

practice<br />

Vascular access<br />

Central venous access for the purpose of supplying<br />

parenteral nutrition to the pediatric age group<br />

requires a careful definition of the patient’s caloric<br />

need, estimated duration of therapy, and an assessment<br />

of available sites. Peripheral venous administration<br />

is the easiest and least hazardous method,<br />

requiring rigorous asepsis, especially in malnourished<br />

children. However, only iso-osmolar solutes,<br />

providing insufficient protein-energy intake, can be<br />

used because of the risk of superficial phlebitis. In<br />

some cases, a further limiting factor can be that<br />

access to superficial veins is restricted or impossible.<br />

Peripherally inserted central catheters offer a<br />

new technology for accessing central veins while<br />

obviating the risk of central vein access. 42<br />

Infusion into the superior vena cava with a CVC is<br />

often necessary. 43 However, the use of a CVC is<br />

accompanied by specific complications, such as<br />

thrombosis or sepsis. There are several routes of


558<br />

Parenteral nutrition in infants and children<br />

central vein access and there are a variety of<br />

catheters available for placement. Tunneled percutaneous<br />

placement of silicone rubber-cuffed<br />

catheters via the subclavian vein approach is the<br />

most commonly used technique, the size of the<br />

catheter being selected according to the child’s<br />

weight and age. 44–46 A CVC with a Dacron ® cuff,<br />

ensuring efficient anchoring, may be inserted either<br />

percutaneously or surgically. In each case the cutaneous<br />

and venous entry sites should be separated<br />

by a 5–10-cm subcutaneous tunnel; one must be<br />

careful since subcutaneous tissue is particularly<br />

thin in severely malnourished children. 43 The risks<br />

of such access catheters include the mechanical<br />

risks of placement, venous thrombosis of the access<br />

sites and, most importantly, catheter-related infections,<br />

either at the exit site or at the subcutaneous<br />

tunnel or pouch, or even generalized sepsis. The<br />

cutaneous exit site should be given meticulous<br />

daily care using iodinized disinfectants and be<br />

protected by an occlusive dressing, which is<br />

changed every day. Implantable devices for longterm<br />

vascular access are also used in children, especially<br />

in those with cancer requiring intravenous<br />

chemotherapy. 47 The exclusive use of these devices<br />

for long-term home PN is rare and, in our experience,<br />

not well accepted by the children. In longterm<br />

home PN patients who have received many<br />

CVCs, we create an arteriovenous fistula. 48 With a<br />

full knowledge of the spectrum of access techniques,<br />

access materials and risks, safe total<br />

parenteral nutrition can be safely delivered to the<br />

children in need.<br />

The use of a 0.22-mm antibacterial membrane filter<br />

between the catheter and the administration tubing<br />

during perfusion remains recommended. The<br />

choice of pump is important, satisfying three criteria:<br />

reliability at low flow rates; ease of use; and<br />

fitted with a safety alarm capable of signaling a<br />

change in flow rate, an air bubble or a blockage<br />

leading to increased pressure. Solutions for<br />

parenteral nutrition must be prepared under strict<br />

asepsis by using antibacterial filters under a laminar<br />

flow hood.<br />

Parenteral nutrition supplies<br />

After a period of development of the technique and<br />

of improvement in prevention of complications, PN<br />

is now widely used in a variety of indications in<br />

pediatric patients. Macro- and micronutrients given<br />

exclusively through the central vein for prolonged<br />

periods are sufficient for nutritional recovery and<br />

maintenance as well as long-term growth in children.<br />

One of the current objectives is to adapt the<br />

PN intakes to the clinical and nutritional situation.<br />

Indeed, it is essential that the composition of the<br />

intravenous feeding solution be adjusted according<br />

to age and disease as well as to the clinical and<br />

biological criteria and as a function of the progressive<br />

transition from parenteral to enteral feeding.<br />

Energy requirements<br />

Defining energy requirements is mandatory to<br />

achieve adequate energy intake and to avoid overfeeding<br />

or underfeeding. Basal metabolic rate (BMR)<br />

and resting energy expenditure (EE) may be used for<br />

achieving adequate energy intake. BMR is the EE of<br />

a recumbent patient in a thermoneutral environment<br />

after a 12–18h fast just when the individual<br />

has awakened, but before starting daily activities.<br />

BMR reflects the EE required for vital processes.<br />

Resting EE refers to the EE of a person at rest in a<br />

thermoneutral environment. BMR and resting EE do<br />

not usually differ by more than 10%. Equations<br />

may be used for calculating BMR or resting EE. 49–51<br />

Most of them are based on body weight, height, age<br />

and sex. None of them will predict energy requirements<br />

with acceptable precision for daily clinical<br />

use. 49 The resting EE of a stable hospitalized child<br />

requiring PN may be measured by using indirect<br />

calorimetry. 52 However, energy intake must cover<br />

total EE, that is the sum of requirements for basal<br />

metabolism, thermic effect of food, thermoregulation<br />

and physical activity. In older children, physical<br />

activity may account for a large proportion of<br />

total EE, especially when he is able to be on cyclic<br />

PN. On the other hand, requirements for a malnourished<br />

child who requires catch-up growth, or the<br />

requirements for a critically ill patient are a matter<br />

of debate. Some data are available in infants or children<br />

after surgery 53–55 or in an ICU. 56–60 It has been<br />

thought that aggression, such as surgery, inflammation<br />

state or sepsis, increases energy requirements<br />

proportionally to the severity of the illness.<br />

However, the increased EE is short-lived. Newborns<br />

have a 20% increase in resting EE after major<br />

surgery, but this elevation returns to baseline within<br />

12–24h. 53 Another study performed by Jaksic et al<br />

failed to show any difference in EE between non-


ventilated surgical neonates (gastro-schisis, atresia,<br />

volvulus) on postoperative day 16 (±12 SD)<br />

compared with infants on extracorporeal life<br />

support studied at 7±3 days of age. 54<br />

Finally, these studies suggest that postoperative or<br />

critically ill infants and children do not require<br />

much more than their resting EE. Overestimation of<br />

the energy requirements for the entire postoperative<br />

period, or for any protracted period of time, may<br />

result in overfeeding and subsequent metabolic<br />

impairment. In addition, there may be significant<br />

interindividual variations in EE.<br />

Measuring resting EE in critically ill infants and<br />

children is difficult because of the expensive equipment<br />

and the expertise required. Nevertheless,<br />

Pierro et al and White et al proposed formulas for<br />

estimating EE in intensive care patients. 61,62 On a<br />

daily clinical practice, energy intake may be<br />

adapted and monitored according to the patient’s<br />

weight gain, the absence of water overload and<br />

evidence of overfeeding. In premature babies and<br />

infants, supplies vary from 150kcal/kg per day to<br />

100kcal/kg per day, and are reduced to<br />

60–80kcal/kg per day in older children and adolescents.<br />

Energy sources<br />

Glucose<br />

Glucose is the unique carbohydrate used during<br />

total PN. Few data are available for establishing the<br />

optimal glucose intake for children receiving PN<br />

and thus avoiding overfeeding from glucose. From<br />

the estimation of glucose utilization by the brain,<br />

which varies according to age, it is possible to<br />

discuss two issues: the contribution of gluconeogenesis<br />

in the glucose supply; and the glucose oxidation<br />

capacity on total body level. 63 Gluconeogenesis<br />

provides a significant amount of glucose (even in<br />

preterm infants) and suggests that not all the<br />

glucose has to be provided exogenously. 64,65<br />

The rate of parenteral glucose delivery must be kept<br />

constant without exceeding the maximum rate of<br />

glucose oxidation (RGO), which differs strongly<br />

among patients according to age and clinical status.<br />

In critically burned children, the maximal RGO has<br />

been found to be 5mg/kg per min. 66 In appropriatefor-gestational-age<br />

preterm infants, the RGO does<br />

not exceed 6–7mg/kg per min (9.5g/kg per day), 67<br />

Parenteral nutrition in clinical practice 559<br />

while in term surgical infants or infants on longterm<br />

total PN, the maximal RGO is about 12mg/kg<br />

per min (18g/kg per day). 68–70 Data collected in<br />

stable adult patients on long-term PN show a<br />

maximal RGO much lower than in children and<br />

infants. 71,72 By excluding the preterm infants and by<br />

taking into account the brain to total body weight<br />

ratio, and according to glucose brain consumption,<br />

one could consider that maximal RGO is continuously<br />

decreasing from birth to adulthood. Thus, the<br />

rate of glucose administration should be adapted to<br />

age and clinical situation, e.g. premature babies,<br />

critically ill patients, severe malnutrition (Table<br />

34.2).<br />

Consequences of excessive glucose intake<br />

(Table 34.3)<br />

When glucose is administered in excess of the<br />

amount that can be directly oxidized for energy and<br />

glycogen production, the excess is directed to lipogenesis73,74<br />

and promotes fat deposition, which may<br />

be a nutritional goal in some clinical situations.<br />

This conversion probably accounts, in part, for the<br />

increase in EE observed with high rates of glucose<br />

infusion. 71 Excessive glucose intake is thought to<br />

increase CO2 production and minute ventilation but<br />

few relevant data are available to support this<br />

evidence. 75–77 Total energy delivery, as well as<br />

amino acid intake, are responsible for increased<br />

CO2 production and minute ventilation. 76,77<br />

Excessive glucose intake may also impair liver function,<br />

especially by inducing steatosis, while its<br />

Table 34.2 Recommended range of glucose<br />

intake for infants and children<br />

Glucose/kg body Day 1 Day 2 Day 3 Day 4<br />

weight per 24h<br />

Infant


560<br />

Parenteral nutrition in infants and children<br />

Table 34.3 Consequences of excessive<br />

glucose intake<br />

Hyperglycemia<br />

Increased CO2 production and minute ventilation<br />

Increased energy expenditure<br />

Lipogenesis and fat tissue deposition<br />

Steatosis and liver dysfunction (increased<br />

transaminases)<br />

Increased production of VLDL triglycerides<br />

VLDL, very low-density lipoprotein<br />

contribution to the development of cholestasis is<br />

not clearly established. 78,79 Studies in normal adult<br />

volunteers suggest that high carbohydrate feeding<br />

leads to an increase in total very low-density<br />

lipoprotein (VLDL) triglyceride secretion rate from<br />

de novo synthesis primarily due to stimulation of<br />

the secretion of preformed fatty acids. 80 The results<br />

imply that the liver derives all its energy from<br />

carbohydrate oxidation as opposed to fatty acid<br />

oxidation, such that fatty acids taken up by the liver<br />

are channeled into VLDL triglycerides. 80 Hepatic<br />

steatosis results when export of the VLDL triglycerides<br />

does not keep pace with production. 80,81<br />

It is obvious that PN is associated with an increased<br />

risk of infectious complications compared with<br />

enteral feeding or no nutritional support. 82–85<br />

Nevertheless, data suggest that hyperglycemia<br />

might be a risk factor for infection, 86–88 especially in<br />

critically ill patients. 89 These data have to be<br />

confirmed. Total PN may be associated with insulin<br />

resistance, due to both the substrate infusion and<br />

the underlying disease. 32 Therefore, particular<br />

attention must be paid to glucose tolerance (hyperglycemia,<br />

glucosuria) at the time of starting cyclic<br />

PN, when decreasing the duration of infusion may<br />

lead to excessive increase in the glucose rate of<br />

delivery. In patients on stable long-term total PN,<br />

glucosuria may reveal a stressful event, particularly<br />

infection, which impairs sensitivity to insulin.<br />

Recommendations for glucose administration<br />

during cyclic PN are provided in Table 34.4.<br />

Lipids<br />

Intravenous fat emulsions (IVFEs) are usually<br />

added to the PN regimen because they provide<br />

Table 34.4 Cyclic parenteral nutrition (PN)<br />

Cyclic PN is well tolerated and may be used from<br />

3–6 months of age<br />

Advantages of cyclic PN<br />

achieves insulin/glucagon balance<br />

improves protein synthesis<br />

prevention of liver disease<br />

physical and psychological<br />

home parenteral nutrition<br />

Hyperinsulinic response prevents hyperglycemia<br />

even with a rate of glucose infusion as high as<br />

1.4g/kg/h<br />

Safety recommendations<br />

use maximum rate of infusion


anabolism, because of adequate energy and nitrogen<br />

intakes, can lead to onset of essential fatty acid deficiency.<br />

A linoleic acid supply of about 2–3% of the<br />

total energy input is recommended in children on<br />

total PN. 98 A daily supply of 4% or 450mg of<br />

linoleic acid per 100kcal is often necessary to<br />

correct a pre-existing deficiency. IVFEs also include<br />

α-linolenic acid (C18:3 n-3). Although its function<br />

and requirements are still poorly defined, an intake<br />

of about 40–50mg/kg per 24h, equal to about 0.5%<br />

of the total energy intake, is normally provided.<br />

Administration of IVFEs is required as soon as<br />

possible in a patient on total PN, according to the<br />

clinical status. Two important issues have to be<br />

considered for IVFE administration: the rate of IVFE<br />

delivery is a major determinant of the rate of fat<br />

clearance from the bloodstream and the optimal<br />

glucose/lipid ratio for optimal energetic substrate<br />

utilization (oxidation) and avoidance of fat overload.<br />

Intravascular metabolism of lipids<br />

Clearance of lipid emulsions<br />

The composition of IVFEs results in a mixture of<br />

artificial chylomicrons having physicochemical<br />

characteristics very similar to those of the natural<br />

chylomicrons produced by enterocytes. They are<br />

hydrolyzed by lipoprotein lipase (LPL) in capillaries<br />

Parenteral nutrition in clinical practice 561<br />

Figure 34.1 Metabolism of intravenous fat emulsions. NEFA, non-essential fatty acids; TG, triglycerides; PL,<br />

phospholipids; LPL, lipoprotein lipase; CETP, cholesterol ester transferase.<br />

of adipose tissue and muscle. This step determines<br />

the rate at which fatty acids from the emulsion are<br />

transported into the tissue for storage or oxidation.<br />

Like many other key enzymes, LPL can be regulated<br />

both in amount and in activity. The amount of LPL<br />

at the capillary endothelium is under hormonal<br />

control, while its activity is regulated by the specific<br />

activator apo CII 99 (Figure 34.1).<br />

Lipid clearance tests have been developed in the<br />

past but are not used routinely in clinical practice.<br />

They allow understanding of the factors determining<br />

the elimination of exogenous fat emulsions from<br />

the circulation. 100 Patients with moderate trauma,<br />

for example after cholecystectomy, have an<br />

increased fractional elimination rate in comparison<br />

with healthy controls. Critically ill patients on the<br />

other hand have an elimination rate which is lower<br />

or close to that of healthy controls. 101<br />

The composition of an exogenous fat emulsion is<br />

also important in determining its elimination from<br />

the circulation. The emulsifying agent is one important<br />

factor in this respect. The egg yolk phospholipids<br />

that are mostly used, give emulsions that are<br />

eliminated very rapidly. IVFEs with high concentrations<br />

of phospholipids, i.e. 10% emulsions, should<br />

be avoided as they carry a higher risk of producing<br />

high serum levels of triglycerides, cholesterol and<br />

phospholipids than other emulsions, i.e. 20% and<br />

30% emulsions. 102,103


562<br />

Parenteral nutrition in infants and children<br />

Interaction between drugs and fat elimination has<br />

frequently been discussed. With this regard,<br />

heparin remains controversial. Heparin acts by activating<br />

and releasing LPL from the endothelial<br />

surface and, when given together with exogenous<br />

fat emulsions, causes a sharp increase in circulating<br />

LPL activity and free fatty acid levels. Sometimes<br />

this might be a positive effect, but in patients with<br />

already high free fatty acid levels an increase in<br />

lipolysis may lead to extremely high free fatty acid<br />

values. There is no place for routine administration<br />

of heparin together with the fat emulsion as<br />

proposed in the past. 104<br />

Interactions with lipoproteins<br />

Lipoproteins play a key regulatory role in the metabolism<br />

and elimination of exogenous lipids.<br />

Endogenous VLDL particles undergo degradation to<br />

LDL in plasma under the action of LPL. These particles<br />

contain apo CII which acts as an activator of LPL.<br />

Exogenous triglyceride particles are hand-led in the<br />

same way but have to acquire apo CII mainly from<br />

the high-density lipoprotein (HDL) fraction before<br />

they can be hydrolyzed. During the degradation of<br />

VLDL particles there is a continuous exchange of<br />

apolipoproteins and surface material. The serum<br />

HDL concentration is positively correlated to the fractional<br />

elimination rate of exogenous fat emulsions.<br />

Routine assessment of patients receiving IVFEs<br />

should include, before and during the course of<br />

treatment: fasting plasma triglycerides, total cholesterol,<br />

and plasma LDL and HDL cholesterol. A<br />

normal plasma triglyceride level does not mean that<br />

exogenous triglycerides are adequately used (oxidation)<br />

or stored (adipose tissue). Indeed, part of the<br />

exogenous lipid may be cleared by other mechanisms,<br />

especially capture by the reticuloendothelial<br />

system (RES). In contrast, high plasma triglyceride<br />

levels suggest impaired clearance related to excessive<br />

infusion rate and/or decreased LPL activity.<br />

Reduced LPL activity may be due to prematurity,<br />

malnutrition, acidosis, hyponatremia, hypoalbuminemia-related<br />

high free fatty acid plasma levels,<br />

high plasma phospholipid levels, or cytokines such<br />

as tumor necrosis factor-α (TNF-α).<br />

Contraindictions to the use of IVFEs<br />

Despite the advantages of IVFEs and the need to<br />

correct any essential fatty acid deficiency, there are<br />

restrictions to the administration of lipids to<br />

malnourished patients. The lower the capillary<br />

tissue mass (a situation found in preterm infants<br />

and malnourished patients), the slower is the rate of<br />

intravenous fat emulsion clearance. Therefore, the<br />

longer the infusion time, the less is the risk for the<br />

patient to develop hypertriglyceridemia. 105,106 In<br />

very malnourished patients, LPL activity reappears<br />

rapidly with the onset of anabolism. The substrate<br />

itself also stimulates LPL synthesis. It is therefore<br />

recommended that IVFEs should not be administered<br />

until a few days after the start of parenteral<br />

nutrition in severely malnourished patients.<br />

There may also be several classic contraindications<br />

to the use of IVFEs during the initial phase, such as<br />

sepsis, thrombocytopenia, disseminated intravascular<br />

coagulation, respiratory distress syndrome or<br />

metabolic acidosis. Furthermore, administration of<br />

IVFEs during neonatal jaundice must be handled<br />

carefully as there is a risk of displacing non-conjugated<br />

bilirubin from albumin with free fatty acids.<br />

Finally, inadequate clearance of IVFEs may result in<br />

RES overload as suggested by the so-called ‘fat overload<br />

syndrome’ (see below).<br />

Optimal glucose/fat ratio<br />

In the past, total PN for adults, children and infants<br />

provided most of the energy as glucose, although it<br />

was not precisely known how much of the intravenously<br />

administered glucose was oxidized.<br />

Glucose-based total PN has been shown to cause<br />

adverse effects related to glucose storage, particularly<br />

as fat. This might account for the extensive<br />

lipid deposition reported both in liver and adipose<br />

tissue. 107,108 In these conditions, fat infusion further<br />

increases fat deposition and may result in fat overloading.<br />

Thus, substitution of part of the glucose<br />

calories avoids the undesirable effects reported with<br />

glucose-based total PN. Studies performed in<br />

infants or neonates have assessed glucose and fat<br />

utilization. 109 By using five isocaloric total PN regimens<br />

differing in their glucose/lipid ratio, it was<br />

possible to assess for the optimal glucose infusion<br />

rate. 110 Fat infusion aiming at a significant contribution<br />

to the coverage of energy expenditure requires<br />

that glucose oxidation be equal to or lower than<br />

maximal oxidative glucose disposal. Hence, glucose<br />

infusion rates should not exceed 18g/kg per day. A<br />

study in malnourished infants and young children<br />

showed that the amount of infused lipid mmust be


adapted to lipid oxidation capacity. 110 There is a<br />

maximal lipid utilization rate of about 3.3–3.6g/kg<br />

per day. Above these values there is an increased<br />

risk of fat deposition secondary to the incomplete<br />

metabolic utilization of infused lipid. Pierro et al<br />

showed similar results in a short-term study<br />

performed in surgical neonates on total PN. 111<br />

Fat overload syndrome<br />

Clinical expression<br />

IVFEs have been thought to impair immune function,<br />

but no relevant data are available suggesting an<br />

impairment. 112 Acute clinical and biological expression<br />

was described as mimicking septic syndrome<br />

but related to macrophage activation. Symptoms<br />

include high fever, hepatosplenomegaly, jaundice,<br />

respiratory distress syndrome, bleeding, thrombocytopenia,<br />

disseminated intra-vascular coagulation,<br />

metabolic acidosis and hypoalbuminemia. 113 This<br />

so-called ‘fat overload syndrome’ is thought to be<br />

related to the capture of exogenous particles by the<br />

RES. Cessation of lipid administration is insufficient<br />

most of the time for improving a patient’s<br />

condition. Steroids may stop the process of<br />

macrophage activation. More recently we reported<br />

several cases of severe cholestasis apparently<br />

related to long-term administration of intravenous<br />

lipids. 114<br />

Mechanisms of lipid toxicity<br />

Although lipids are necessary in patients on PN,<br />

they are strongly suspected to be toxic in some<br />

cases. The metabolism of their oxidized fraction is<br />

relatively well known; far less is known about the<br />

Table 34.5 Recommendations for lipid<br />

administration<br />

Use of 20% lipid emulsion<br />

Maximal daily amount of 2–2.5g/kg (


564<br />

Parenteral nutrition in infants and children<br />

shown to be cleared more rapidly and are now<br />

widely used in adult and pediatric patients. 118–120<br />

Fatty acids from the hydrolysis of MCTs are the<br />

primary substrate for ketogenesis. Carnitine is<br />

needed to transport long-chain fatty acids into the<br />

mitochondria, but malnourished and seriously ill<br />

patients may be carnitine-depleted. An energy<br />

source able to bypass this route into the mitochondria<br />

would in theory, be useful for such patients.<br />

The MCT/long-chain triglyceride (LCT) lipid emulsion<br />

(B. Braun, Germany) contains 50% LCTs to<br />

avoid any possible side-effects from excessive<br />

amounts of MCTs and to provide essential fatty<br />

acids. Medium-chain fatty acids can enter the mitochondria<br />

by simple diffusion, independent of the<br />

carnitine enzyme, and produce an elevation of<br />

plasma ketones. 118,119 MCTs have been shown to<br />

improve nitrogen balance in postoperative patients<br />

but this positive effect remains controversial<br />

according to several other studies. 120–122 There have<br />

been several published studies in children using<br />

MCTs. 123–126 Their use in malnourished infants after<br />

15 days on total PN indicates that they are well<br />

tolerated and could provide advantages in terms of<br />

nitrogen metabolism by supplying the equivalent of<br />

25% non-protein energy intake. 124 MCT emulsions<br />

have also been used in home PN pediatric patients<br />

with beneficial effect in reducing cholestasis. 125<br />

Structured triglyceride emulsion<br />

To improve the safety and the efficiency of MCTcontaining<br />

fat emulsions, a structured triglyceride<br />

emulsion, containing both long-chain and mediumchain<br />

fatty acids bound to the same carbon skeleton,<br />

has been synthesized. Such structured triglyceride<br />

emulsions have been shown to improve<br />

nitrogen retention and muscle protein synthesis in<br />

animal models and in humans. 127,128 There is is no<br />

study currently available in pediatric patients.<br />

Fish oil-based emulsions<br />

Fish oil triglycerides are also now being considered<br />

for PN either as pure fish oil particles or incorporated<br />

into MCT/LCT emulsions. Fish oil is now<br />

proposed as a component of lipid emulsions. By<br />

providing a high amount of long-chain polyunsaturated<br />

fatty acids (PUFAs), they are reported to have<br />

anti-inflammatory effects. 129,130 There is a study<br />

currently being carried out in pediatric patients.<br />

Olive oil-based IVFEs<br />

It is well known that a high intake of monounsaturated<br />

fatty acids in the form of olive oil is associated<br />

with a lower incidence of cardiovascular morbidity.<br />

131 In patients dependent on long-term PN, the<br />

availability of intravenous fat emulsions containing<br />

lower amounts of unsaturated fatty acids could be<br />

of interest. 132,133 New 20% fat emulsions containing<br />

17% olive oil and only 3% soybean oil are now<br />

available. 133 It has been shown in a long-term pediatric<br />

study that olive oil-based emulsion preserves<br />

essential fatty acid status and fatty acid elongation<br />

and decreases total cholesterol. 133 The olive oil<br />

group showed better measurements of antioxidant<br />

activity against lipid peroxidation. The same olive<br />

oil- and vitamin E-enriched emulsion was recently<br />

reported as a valuable alternative for PN of preterm<br />

infants who are often exposed to oxidative stress,<br />

while their antioxidative defense is weak. 134<br />

Carnitine and α-tocopherol<br />

Carnitine plays a central role in metabolism especially<br />

for optimal oxidation of fatty acids. 135<br />

Carnitine, in the form of acylcarnitine transfers free<br />

fatty acids into mitochondria, where they are<br />

recombined with co-enzyme A (CoA) to form acyl-<br />

CoA. 135 Plasma levels of carnitine decrease rapidly<br />

in premature newborns and small-for-gestationalage<br />

neonates during the first days of life if no exogenous<br />

carnitine is provided. 136 Available studies did<br />

not assess the use of carnitine during long-term PN,<br />

a situation that is more likely to result in carnitine<br />

deficiency. Some authors have raised the question<br />

of carnitine supplementation for children on total<br />

PN. 137 As yet, in the absence of specific recommendations,<br />

a supplement of 2mg/kg per day may be<br />

advisable. 138 Supplementation with α-tocopherol as<br />

antioxidant, added to IVFEs at a dose of 0.6mg/g of<br />

unsaturated fatty acids is also recommended.<br />

Nitrogen intake<br />

Energy supply should be closely correlated with<br />

that of nitrogen. Nitrogen intake obviously depends<br />

on the age and degree of malnutrition. 140 In premature<br />

babies the intake varies from 400–500mg to<br />

650mg/kg per day. 141 In infants, the intake varies<br />

from 400mg 142 to 800mg/kg per day in the case of<br />

great nitrogen losses from the GI tract. In older chil-


dren, 300mg/kg per day is usually sufficient. Such<br />

intakes, which are higher than the growth requirement,<br />

cover excessive nitrogen losses induced by<br />

catabolism. Greater amounts are unsuitable,<br />

because with a constant caloric intake there is a<br />

negative correlation between nitrogen intake and<br />

the amount retained and the subsequent danger of<br />

hyperaminoacidemia, metabolic acidosis or isoosmolar<br />

coma. Such intakes may also be responsible<br />

for excessive urinary losses of calcium and<br />

phosphorus and subsequent bone mineralization<br />

impairment. 143 Finally, usual nitrogen/energy ratio<br />

varies around 1g of nitrogen for 200–250kcal.<br />

Nitrogen sources<br />

Nitrogen sources available for PN come from<br />

various mixtures of crystalline L-amino acids. They<br />

have been shown to be effective in clinical use,<br />

providing appropriate nitrogen utilization and<br />

retention. Amino acid solutions for use in PN are<br />

selected according to the following criteria: 144,145<br />

(1) Whether the solution contains all or only some<br />

of the naturally occurring amino acids;<br />

(2) Total amino acid nitrogen content;<br />

(3) Osmolality and electrolyte content;<br />

(4) Amount of essential amino acids per gram of<br />

total nitrogen (E/T);<br />

(5) Non-nitrogen nutrient content (glucose, fructose).<br />

New pediatric solutions have been developed<br />

which appear to be better suited for use in<br />

newborns, premature babies or malnourished<br />

infants (Primene, Clintec ® ; Vaminolac, Pharmacia<br />

® ). Such products have an E/T ratio greater than<br />

3 and differ from standard solutions in having a<br />

higher percentage of branched-chain amino acids;<br />

modified aromatic amino acid content (a one-third<br />

reduction in phenylalanine); modified sulfur amino<br />

acid content, with methionine reduced by 50% and<br />

an increase in cysteine content (both solutions also<br />

contain taurine, which is absent from standard solutions);<br />

and increased lysine content.<br />

Other sources of nitrogen<br />

Protein-energy malnutrition secondary to chronic<br />

disease, as well as acute illness such as injury or<br />

Parenteral nutrition in clinical practice 565<br />

infection, are associated with loss of body fat and<br />

skeletal muscle mass. The loss of body tissue may<br />

be minimal and of little consequence in patients<br />

with normal nutritional status and a brief, self-limiting<br />

illness lasting a few days. However, when the<br />

disease is prolonged and the patient is malnourished,<br />

a variety of clinical events may occur in association<br />

with the catabolic state. These alterations<br />

include immunosuppression, delayed wound<br />

healing and tissue repair, and loss of muscle<br />

strength. 146 The accelerated breakdown of body<br />

protein can be slowed by the administration of<br />

adequate quantities of energy, protein (amino acids)<br />

and other essential nutrients. However, measurements<br />

of body composition and substrate-flux<br />

studies indicate that it is extremely difficult to<br />

maintain or replenish body protein during catabolism.<br />

146 Thus, reducing the debility associated with<br />

the catabolic process could potentially enhance<br />

recovery and decrease the consequences of illness<br />

on protein retention and height–growth velocity.<br />

Because the efficacy of nutrition cannot be<br />

improved easily by quantitative modifications,<br />

attention has focused in recent years on qualitative<br />

improvments. The addition of specific amino acids<br />

or other sources of nitrogen to the feeding formulas<br />

might be logical in critically ill children (sepsis,<br />

burns, trauma) or in patients with chronic inflammatory<br />

processes such as severe Crohn’s disease.<br />

Glutamine<br />

Glutamine is the most abundant amino acid in the<br />

body. 147 However, glutamine is absent from the<br />

currently available amino acids solutions. Indeed,<br />

glutamine is considered as unstable in aqueous<br />

solutions and during heat sterilization with the<br />

formation of pyroglutamic acid and ammonia.<br />

Although glutamine is a non-essential amino acid,<br />

the nutritional requirement for this amino acid<br />

during catabolic illness may differ greatly from that<br />

during health. 147 During starvation or stress, the<br />

concentration of free glutamine in the intracellular<br />

amino acid pool of skeletal muscle rapidly<br />

decreases. Glutamine exported from the muscle is<br />

used primarily by visceral organs; in the kidney it<br />

serves as an ammonia donor; in the gastrointestinal<br />

tract it serves as a primary oxidizable fuel source for<br />

enterocytes and colonocytes. 148,149 Glutamine also<br />

supports other rapidly proliferating tissue, such as<br />

fibroblasts or lymphocytes. Glutamine-supple-


566<br />

Parenteral nutrition in infants and children<br />

mented total PN has been shown to preserve gut<br />

structure and to improve gut immune function in<br />

animal models. 150 Studies have shown the clinical<br />

benefits of glutamine-supplemented total PN in<br />

adult patients undergoing bone marrow transplantation.<br />

151–154 The use of glutamine-containing<br />

dipeptides is proposed because of the instability of<br />

free glutamine. Improved nitrogen balance has been<br />

shown in patients receiving alanyl-glutaminesupplemented<br />

total PN. 155 In addition, glutamine<br />

dipeptide-supplemented PN prevents intestinal<br />

atrophy and increased permeability in critically ill<br />

adult patients. 156<br />

Ornithine α-ketoglutarate<br />

Another source of nitrogen for patients on PN is<br />

represented by ornithine α-ketoglutarate (OKG).<br />

This is a salt formed with two molecules of<br />

ornithine and one molecule of α-ketoglutarate. OKG<br />

has been successfully used by the enteral and<br />

parenteral route in burn, traumatized and surgical<br />

patients and in chronically malnourished<br />

patients. 159,160 According to the situation, OKG<br />

administration decreases muscle protein catabolism<br />

and/or increases protein synthesis. The mechanism<br />

of action of OKG is not fully understood, but it was<br />

clearly demonstrated that it is a precursor of glutamine.<br />

162,163 In addition, the secretion of anabolic<br />

hormones (insulin, human growth hormone) and<br />

the synthesis of metabolites (polyamines, arginine,<br />

ketoacids) may be involved. 160,161 In prepubertal<br />

children on PN, administration of OKG (15g/day)<br />

reversed growth retardation and increased insulinlike<br />

growth factor I (IGF-I) plasma levels. 161<br />

Finally, it seems likely that, in the near future a<br />

more specific therapeutic approach to protein<br />

metabolism might be achieved. This prospect is of<br />

great importance for children with regard to the<br />

consequences of inadequate protein metabolism on<br />

growth velocity.<br />

Other components of PN solutions<br />

Water and electrolyte intake must be varied according<br />

to the age and condition of the child, needing<br />

adjustment, for example, if there are intestinal<br />

losses. Premature babies need 150–200ml water per<br />

kilogram body weight daily to maintain equilibrium;<br />

infants require 120–140ml/kg and older<br />

Table 34.6 Vitamin requirements<br />

Infants Children<br />

Vitamin A (µg) 300–750 450–1000<br />

Vitamin D (IU) 100-1000 200–2500<br />

Vitamin E (mg) 3–10 10–15<br />

Vitamin K (µg) 50–75 50–70<br />

Vitamin Bl (mg) 0.4–0.5 1.5–3<br />

Vitamin B2 (mg) 0.4–0.6 1.1–3.6<br />

Vitamin B5 (mg) 2–5 0.5–5<br />

Vitamin B6 (mg) 0.1–1.0 1.5–2<br />

Vitamin B12 (µg) 0.3–3 3–100<br />

Vitamin C (mg) 25–35 20–100<br />

Folic acid (µg) 20–80 100–500<br />

Biotin (µg) 35–50 150–300<br />

Niacin (mg) 6–8 5–40<br />

Table 34.7 Trace element requirements<br />

Element Infants Children<br />

(/kg/day) (/day)<br />

Iron (mg) 50 100–2500<br />

Zinc (mg) 100–250 1000–5000<br />

Copper (mg) 20–30 200–300<br />

Selenium (µg) 2–3 30–60<br />

Manganese (µg) 1–10 50–250<br />

Molybdenum (µg) 0.25–10 50–70<br />

Chrome (µg) 0.25–2 10–20<br />

lodine (µg) 1–5 50–100<br />

Fluoride (µg) 20 500–1000<br />

children 80–100ml/kg per day. All normal babies<br />

and children need 3–5mmol chloride, sodium and<br />

potassium per kilogram per day. When there are<br />

losses due to vomiting or gastric aspiration 8mmol<br />

sodium, 1mmol potassium, 6mmol H + ions and<br />

12mmol chloride should be added to each 100ml of<br />

water. In the case of an enterostomy, 15mmol<br />

sodium, 1mmol potassium, 10mmol of chloride<br />

and 5mmol bicarbonate should be added to each<br />

100ml of water.<br />

With a nitrogen intake of 400mg/kg per day and a<br />

calcium intake of 1.7mmol/kg per day, the daily<br />

requirement of phosphorus is 2.3mmol/kg for bone<br />

growth and nitrogen anabolism. If the phosphorus<br />

intake is lower or the intake of nitrogen and/or


calcium is higher, severe phosphorus depletion<br />

results. Magnesium requirements are usually satisfied<br />

by 1mmol/kg per day.<br />

Vitamin and trace element intakes are provided as a<br />

function of the intake of the respective nutrients<br />

and their anabolism. For infants and older children,<br />

adherence to the recommendations in Tables 34.6<br />

and 34.7 helps to prevent depletion or excess. These<br />

intakes should be adjusted in cases of catabolic<br />

stress, such as an infection or intestinal losses.<br />

All-in-one mixture 162–168<br />

Administration of IVFEs together with the glucose<br />

amino acids in a three-in-one mixture is now widely<br />

used in children. There are a number of potential<br />

advantages to PN admixtures but also some drawbacks.<br />

In addition to reducing cost, use of a PN<br />

admixture simplifies the patient’s life in that only a<br />

single infusion pump with less tubing and other<br />

accessories is needed. 162 There is also evidence that<br />

it reduces the risk of bacterial growth in intravenous<br />

fat emulsion even after 24h. 163<br />

Stability is directly related to the concentration of<br />

the components, and is dependent on pH and<br />

temperature. If the mixture is not compatible, the<br />

fat emulsion will break, forming an oil–water<br />

interphase that can be seen floating on top of the<br />

mixture. Because the admixture is a physical mix<br />

of naturally incompatible substances (oil and<br />

water, calcium and phosphorus), their utilization<br />

requires strict attention to pharmaceutical guidelines<br />

for preparation, storage and use. 164,165<br />

Particulate matter (mobile, undissolved substances<br />

such as precipitates of calcium and phosphorus<br />

that are unintentionally present in products)<br />

resulting from inappropriate preparation or storage<br />

can be life-threatening. To reduce the risk of<br />

precipitates reaching the patient an in-line filter<br />

should be used. This should be a 1.2µm aireliminating<br />

filter (in contrast to the 0.22µm aireliminating<br />

filter recommended for non-lipidcontaining<br />

PN). A PN admixture is considered<br />

inappropriate for administration if >0.4% of the<br />

total fat contains particles >5µm in size. 166 Some<br />

drugs are compatible with PN admixtures but not<br />

with traditional PN, and vice versa. 167<br />

It is possible to formulate total PN mixtures that<br />

have good short-term stability and that satisfy the<br />

Refeeding syndrome 567<br />

nutritional requirements of most patients. Several<br />

companies produce a three-in-one mixture with<br />

good long-term stability. Retention of long-term<br />

stability in a mixture containing electrolytes is more<br />

difficult, although advances are being made in this<br />

area. The development of complete total PN<br />

mixtures with long-term stability will be assisted by<br />

advances in our understanding of the fundamental<br />

physical chemistry of colloid mixtures, particularly<br />

the interaction between emulsions and amino acids.<br />

Indeed such a ternary solution may lead to lipid<br />

instability, and/or catheter obstruction. PN admixtures<br />

should only be used in patients who are clinically<br />

stable. Solutions may be stable when refrigerated<br />

and become unstable at room temperature.<br />

Changes in the formulation of PN admixtures are<br />

more costly than changes in traditional (two-in-one)<br />

PN because of the wastage of both the dextrose<br />

amino acid and its components, and the intravenous<br />

fat emulsion. A number of questions remain<br />

about PN admixtures. These include the appropriate<br />

dosing of some vitamins, the true risks related to<br />

particulate matter and fat-droplet size, and drug<br />

compatibilities.<br />

Refeeding syndrome<br />

Pathophysiology<br />

If PN is required because of severe malnutrition and<br />

inability of the GI tract to cover the protein-energy<br />

requirements, it has to be provided very carefully. 168<br />

Indeed, refeeding syndrome may be observed in<br />

severely malnourished patients receiving concentrated<br />

calories via PN. 169 This syndrome includes<br />

the metabolic and physiological consequences of<br />

the depletion, repletion, compartmental shifts and<br />

inter-relationships of the following: phosphorus,<br />

potassium, magnesium, glucose metabolism,<br />

vitamin deficiency and fluid resuscitation. 170,171<br />

The net effect of the hormonal and metabolic<br />

changes of starvation is to facilitate survival by a<br />

reduction in basal metabolic rate, conservation of<br />

protein and prolongation of organ function, despite<br />

the preferential catabolism of skeletal muscle tissue.<br />

Infants and children who have suffered from<br />

malnutrition for weeks or months will also experience<br />

a significant loss of visceral cell mass.<br />

Refeeding the malnourished child disrupts the<br />

adaptative state of semi-starvation.


568<br />

Parenteral nutrition in infants and children<br />

As refeeding is initiated there is a rapid reversal<br />

in insulin, thyroid and adrenergic endocrine<br />

systems. Basal metabolic rate increases and<br />

glucose becomes the predominant cellular fuel.<br />

The body immediately begins the process of<br />

rebuilding lost tissue. Anabolism is accompanied<br />

by a positive balance of intracellular minerals.<br />

As minerals shift to intracellular spaces,<br />

serum levels may plummet. Body fluid compartments<br />

redistribute as intracellular fluid<br />

increases; extracellular fluid may increase or<br />

decrease depending on the previous intake, the<br />

persistent digestive losses and the refeeding<br />

regimen. These rapid changes in metabolic<br />

status can create life-threatening complications,<br />

so the nutritional regimen must be chosen wisely<br />

and monitored closely. Several potential metabolic<br />

complications of the refeeding syndrome<br />

are listed in the Table 34.8.<br />

To reduce the risk of refeeding complications,<br />

several conditions are required at the initial<br />

phase of renutrition of severely malnourished<br />

infants and children.<br />

Table 34.8 Metabolic disorders associated with refeeding syndrome<br />

Prevention of water and sodium overload<br />

It is necessary to reduce the patient’s water and<br />

sodium intake, depending on the hydration state,<br />

to prevent water and sodium overload resulting<br />

from their excessive retention, accentuated by<br />

increased secretions of vasopressin and aldosterone.<br />

Early weight gain may be the consequence<br />

of fluid retention. The monitoring of water and<br />

electrolyte intake must include uncontrolled<br />

losses as well as those from the Gl tract. One of the<br />

difficulties in such situations is the need to take<br />

into account a third factor, such as intraperitoneal<br />

or intestinal fluid retention. Monitoring of body<br />

weight changes, urine collection, and assessment<br />

of blood and urinary electrolytes are essential.<br />

Oncotic pressure restoration<br />

The infusion of macromolecules aims to restore<br />

oncotic pressure and to minimize hemodynamic<br />

problems, worsened by a water–electrolyte imbalance.<br />

Albumin is the best infusate, but fresh-frozen<br />

Hypophosphatemia<br />

Cardiac: altered myocardial function, arrhythmia, congestive heart failure, sudden death<br />

Hematological: altered red blood cell morphology, hemolytic anemia, white blood cell dysfunction,<br />

thrombocytopenia, depressed platelet function, bleeding<br />

Hepatic: liver dysfunction<br />

Neuromuscular: acute areflexic paralysis, confusion, coma, cranial nerve palsies, diffuse sensory loss,<br />

Guillain–Barré-like syndrome, lethargy, paresthesias, rhabdomyolysis, seizures, weakness<br />

Respiratory: acute ventilatory failure<br />

Hypokalemia<br />

Cardiac: arrhythmias, cardiac arrest, increased digitalis sensitivity, orthostatic hypotension, ECG changes (T-wave<br />

flattening or inversion, U waves, ST segment depression)<br />

Gastrointestinal: constipation, ileus, exacerbation of hepatic encephalopathy<br />

Metabolic: glucose intolerance, hyporeftexia, paralysis, paresthesias, respiratory depression, rhabdomyolysis,<br />

weakness<br />

Renal: decreased urinary concentrating ability, polyuria and polydypsia, nephropathy with decreased glomerular<br />

filtration rate, myoglobinuria (secondary to rhabdomyolysis)<br />

Hypomagnesemia<br />

Cardiac: arrhythmias, tachycardia, torsade de pointes<br />

Gastrointestinal: abdominal pain, anorexia, diarrhea, constipation<br />

Neuromuscular: ataxia, confusion, fasciculations, hyporeftexia, irritability, muscle tremors, painful paresthesias,<br />

personality changes, positive Trousseau's sign, seizures, tetany, vertigo, weakness


plasma or blood may be required in cases of anemia<br />

and/or coagulation disorders. Artificial ventilation<br />

may be required in those, generally few, cases<br />

having a poor cardiorespiratory status.<br />

Intake of carbohydrates<br />

Constant administration of carbohydrate is required<br />

to maintain blood glucose homeostasis as the<br />

reserves are very low; parenteral administration of<br />

glucose requires care because of the risk of hyperglycemia<br />

with osmotic diuresis and hyperosmolar<br />

coma.<br />

Potassium repletion<br />

Correction of potassium depletion is of great importance,<br />

but should be achieved progressively with<br />

monitoring of renal and cardiac functions. It can be<br />

dangerous to try to correct the deficiency too<br />

rapidly at a stage where the capacity for fixing<br />

potassium remains low because of reduced protein<br />

mass; excessive intake leads to the cardiac risk of<br />

hyperkalemia.<br />

Body temperature monitoring<br />

It must be monitored, since protein-energy malnutrition<br />

(PEM) can lead to hypothermia, often associated<br />

with hypoglycemia and bradycardia. On the<br />

other hand, hyperthermia or excessive reheating<br />

increases water loss as well as energy expenditure.<br />

Maintenance of a stable body temperature between<br />

36°C and 37°C by appropriate warming techniques<br />

is essential. Careful daily nursing to prevent cutaneous<br />

lesions and musculotendinous retractions is<br />

also very important.<br />

Prevention of infection<br />

The infective, metabolic and GI problems must be<br />

constantly borne in mind during treatment of pediatric<br />

patients with severe PEM. The risk of infection,<br />

an expression of both specific and non-specific<br />

immunity depression, may jeopardize the prognosis<br />

and aggravate nutritional problems at any time.<br />

Clinical and paraclinical investigations must be<br />

performed repeatedly, to look for widening foci of<br />

infection (respiratory, GI, skeletal and urinary) and<br />

Refeeding syndrome 569<br />

for their systemic spread. When a localized or<br />

systemic infection is identified, specific treatment is<br />

urgently required. The routine use of antibiotics in<br />

the absence of bacteriological evidence in a<br />

malnourished child is inadvisable; antibiotics<br />

should only be given if sufficient indirect evidence<br />

points to the likelihood of infection. Active intestinal<br />

parasitosis should, of course, be vigorously<br />

treated. Evidence or suspicion of an infection is an<br />

essential factor in modifying water, electrolyte and<br />

nutrient intake and also in the choice of the feeding<br />

technique.<br />

Protein and energy intake<br />

It is difficult to suppress protein catabolism under<br />

these conditions of stress and low-energy intake.<br />

Excessive nitrogen intake may lead to hyperammonemia<br />

and/or acidosis by exceeding the renal<br />

clearance capacity for H + and phosphate ions. An<br />

intake of 0.5–1g/kg of parenteral amino acids or oral<br />

peptide is sufficient to maintain the plasma amino<br />

acid pool. The protein-energy deficiency and other<br />

related disorders must be corrected during the days<br />

following the initial period of stress. This type of<br />

correction should be made carefully and gradually<br />

since the deficits are profound and of longstanding.<br />

It is essential to provide both nitrogen and calories<br />

simultaneously and in the correct ratio. The<br />

increase in energy intake, if progressive, avoids<br />

acute episodes of sodium and water retention<br />

accompanied by oliguria and a fall in urinary<br />

sodium and potassium output. It is likely that these<br />

changes are carbohydrate dependent, since their<br />

occurrence and equally rapid reversal develop with<br />

changes in glucose rate of glucose infusion. This<br />

antinatriuretic effect of glucose appears to be<br />

similar to the antinatriuresis observed during<br />

feeding after a phase of experimental fasting. The<br />

insulin secreted induces sodium tubular reabsorption,<br />

and the alkalosis that develops might be due to<br />

increased tubular absorption of bicarbonate.<br />

Micronutrients<br />

The provision of appropriate nutrient solutions<br />

requires an understanding of the nutritional relationships<br />

between nutrients electrolytes, vitamins<br />

and trace elements. It is during this initial phase<br />

that any deficit due to incorrect intake will become


570<br />

Parenteral nutrition in infants and children<br />

apparent through either clinical or laboratory signs.<br />

These deficits can usually be prevented by giving<br />

them in the following proportions: 200–250kcal,<br />

nitrogen 1g, calcium 1.8mmol, phosphorus<br />

2.9mmol, magnesium 1.0mmol, potassium<br />

10mmol, sodium and chloride 7mmol, zinc 1.2mg.<br />

Similarly, it is essential to adapt the intake of<br />

copper, manganese, chromium, iron, iodine, cobalt<br />

and fluoride, and the group B vitamins in particular;<br />

also, the intakes of essential fatty acids, tocopherol<br />

and selenium.<br />

Adaptation of intake<br />

After the initial phase of renutrition, most complications<br />

can be prevented by careful supervision and<br />

the provision of appropriate intakes. It is essential<br />

that the infusion rate, body temperature, cardiac<br />

and respiratory function, urinary volume, twice<br />

daily weight and digestive output are continuously<br />

monitored. During the first 5 days and also when<br />

the osmotic load is increased, urine should be<br />

checked for osmolality, pH, glucose and protein.<br />

The plasma and urinary ion data, plus the calcium,<br />

phosphorus, magnesium, glucose and hematocrits<br />

should be obtained twice during the first week, and<br />

then once weekly; plasma proteins, albumin, bilirubin,<br />

alkaline phosphatase and transaminase values<br />

should be assessed routinely. These data, and<br />

knowledge of the patient’s state and age, should<br />

make it possible to progressively regulate and<br />

control the intake and avoid problems of overload<br />

or depletion.<br />

Complications of parenteral nutrition<br />

(Table 34.9)<br />

Catheter-related sepsis<br />

In hospital as well as at home, catheter-related<br />

sepsis is one of the most serious complications<br />

which can arise during parenteral nutrition. 172–175 A<br />

2-year prospective study of 185 CVCs showed a<br />

sepsis rate of 0.26%, with an overall incidence of<br />

one catheter-related sepsis per 278 days of total<br />

PN. 176 Systemic antibiotics provided sepsis control<br />

in 88% of the cases while CVC removal was<br />

required in the other cases. The factors significantly<br />

correlated with sepsis were: age (1–5 years); CVC<br />

Table 34.9 Complications during parenteral<br />

nutrition (PN)<br />

Infectious<br />

Local skin infections<br />

Catheter-related sepsis<br />

Complications of catheter-related sepsis:<br />

endocarditis, osteomyelitis<br />

Metabolic<br />

Water and/or sodium overload<br />

Hyperosmolar coma<br />

Excessive urinary losses<br />

Hyperglycemia with glycosuria–hypoglycemia<br />

Metabolic acidosis<br />

Hyperazotemia–hyperammonemia<br />

Hypokalemia<br />

Hypophosphatemia<br />

Hypercalcemia–hypercalciuria<br />

Hypertriglyceridemia–hypercholesterolemia<br />

Nutritional deficiencies<br />

Essential fatty acid deficiency<br />

Carnitine deficiency<br />

Trace element deficiency: Fe, Zn, Cu, Se, Mo, etc.<br />

Vitamin deficiency: A, E, B, B12, folates, etc.<br />

Long-term total PN<br />

Total PN liver disease<br />

Total PN bone disease<br />

Hematological and coagulation disorders<br />

type (surgically inserted CVCs were more frequently<br />

infected); local hemorrhage following CVC insertion;<br />

and local suppuration at the skin exit site.<br />

Prevention of catheter-related sepsis requires strict<br />

asepsis during both CVC insertion and changes of<br />

filter and infusion sets. Daily care of the CVC skin<br />

exit site is of great importance. All PN solutions<br />

must be prepared under a laminar flow hood and<br />

filtered. The care of the child should be undertaken<br />

by physicians and nurses who have been specifically<br />

trained in this technique. Fever or clinical<br />

signs suggestive of catheter-related sepsis should<br />

lead to a thorough search for a source of sepsis,<br />

together with a white blood cell count, C-reactive<br />

protein and coagulation tests. Samples for blood<br />

culture should be taken via the catheter and from a<br />

peripheral vein. If fever persists, antibiotic therapy<br />

should be started early and include antibiotics


against Staphylococcus. Coagulase-negative staphylococci<br />

(CoNS) is the most frequent blood culture<br />

isolate from neonates, infants and children with<br />

CVCs. 172,176 Clinicians vary in their management of<br />

suspected line sepsis, particularly that caused by<br />

CoNS. A vancomycin-containing regimen is widely<br />

used in infants and children suspected of having<br />

line sepsis. Procedures to prevent CoNS-positive<br />

blood cultures and to differentiate CoNS contaminants<br />

from pathogens are needed. For safely<br />

decreasing vancomycin use, clinical practice guidelines<br />

should be developed, implemented and evaluated.<br />

The guidelines should include optimal skin<br />

antisepsis and catheter disinfection before obtaining<br />

blood for culture, obtaining two blood cultures<br />

and using adjunctive tests and information to help<br />

differentiate contaminants from pathogens, and<br />

restriction on empiric vancomycin use.<br />

Both catheter removal and infection dissemination<br />

may be avoided by starting antibiotics soon after<br />

onset of fever. However, removal of the catheter is<br />

systematically considered in case of fungal infection,<br />

infection due to Staphylococcus aureus with<br />

cutaneous infection, or if the patient continues to<br />

deteriorate even when appropriate antibiotic<br />

therapy had been started. In neonates who were<br />

infected with Staphylococcus aureus or with nonenteric<br />

Gram-negative rods, it was shown that<br />

delayed removal of the CVC was associated with<br />

complicated bacteremia. 174<br />

Otherwise, removal of the catheter is considered<br />

when blood cultures remain positive after several<br />

days of optimal antibiotic treatment, or when the<br />

PN program is close to completion. With good technique,<br />

displacement or obstruction of the catheter,<br />

or thrombosis of large vessels are rare. Careful technique<br />

should also help prevent superficial venous<br />

thrombophlebitis and the risk of dissemination of<br />

septic emboli.<br />

Parenteral nutrition-related bone disease<br />

The so-called PN-related bone disease resembles<br />

rickets, with fractures of the limbs which are sometimes<br />

asymptomatic and are only discovered after<br />

routine X-ray examination. 177 The most constant<br />

laboratory features are an elevated alkaline phosphatase<br />

activity and hypercalciuria, with normal or<br />

subnormal levels of vitamin D metabolites and<br />

parathyroid hormone. Bone histology shows osteo-<br />

Complications of parenteral nutrition 571<br />

malacia-like changes with reduced mineralization<br />

and excess of osteoid tissue. The etiology of these<br />

bone lesions is probably multifactorial: excess<br />

vitamin D or disorders of its metabolism mean that<br />

it must be given very carefully on long-term<br />

parenteral nutrition. It is also possible to reduce the<br />

hypercalciuria by ensuring that the supplies of<br />

phosphorus, nitrogen and energy are properly<br />

balanced, while reducing the supply of amino acids,<br />

especially the sulfur-containing amino acids. 178<br />

Finally, it is necessary to ensure that the solutions<br />

used for children during long-term PN are not contaminated<br />

with alum-inum. 179,180 Prevention of this<br />

‘bone disease’ depends primarily on regular<br />

measurements of urinary calcium, which should<br />

not exceed 5mg/kg per 24h, and serum alkaline<br />

phosphatase activity. Measurement of bone mineral<br />

content is now widely performed using dual X-ray<br />

absorptiometry (DEXA) scan. 181<br />

Parenteral nutrition-related liver disease<br />

Liver disease is a major side-effect of long-term PN.<br />

Fifteen years after the first reports its pathogeny is<br />

not completely understood. 182 Disruption of bile<br />

acid enterohepatic circulation in case of ileal amputation,<br />

impairment in choleresis in the absence of<br />

oral feeding, bacterial overgrowth due to bowel<br />

obstruction, stasis and lack of ileocecal valvula, are<br />

patient-dependent factors thought to contribute to<br />

PN-associated cholestasis. 183 One mechanism might<br />

be an increase in bile concentration of lithocholic<br />

acid. 184 Bacterial infections were also proposed as a<br />

co-factor in PN-related cholestasis, since the sepsisassociated<br />

cholestasis has been well described, both<br />

in human and animal models. 185–187 Duration of PN<br />

is also a known risk factor. Qualitative aspects of PN<br />

are also a matter of debate. It was experimentally<br />

demonstrated that an excess in total energy delivered<br />

induces liver lesions, reversible when decreasing<br />

the energy supply. 188 The role of amino acids<br />

was suspected, either an excess or a lack of<br />

them, 189,190 and an excessive glucose supply might<br />

induce steatosis through the increase in de novo<br />

lipogenesis. 191 Hepatobiliary complications of PN<br />

are now well recognized and documented. Such<br />

liver involvement may result in some cases in endstage<br />

liver disease within a few months or years.<br />

Many PN-related and patient-related risk factors are<br />

involved in those hepatobiliary complications.<br />

Since the underlying digestive disease plays a


572<br />

Parenteral nutrition in infants and children<br />

prominent role, some pediatric patients requiring<br />

long-term PN are at high risk of developing liver<br />

disease. Short-bowel syndrome may be associated<br />

with disruption of bile acid enterohepatic circulation<br />

due to ileal amputation, impairment in<br />

choleresis when oral feeding is impossible, bacterial<br />

overgrowth due to bowel obstruction, stasis and<br />

lack of ileocecal valvula, which are all factors<br />

thought to contribute to PN-associated cholestasis.<br />

Recurrent septic episodes either catheter-related<br />

(Gram-positive bacteria) or digestive-related (Gramnegative<br />

sepsis from in-traluminal bacterial overgrowth)<br />

also contribute to liver injury. 192–197<br />

Prematurity itself might be an associated factor. In<br />

addition, inadequate PN may have an additional<br />

deleterious effect on the liver because of metabolic<br />

disorders or micronutrient overload. Indeed,<br />

continuous PN infusion with excessive glucose<br />

intake is associated with hyperinsulinism and<br />

subsequent steatosis. 144 Inad-equate amino acid<br />

supply is thought to be responsible for metabolic<br />

dysfunction of the liver. Excessive aluminum, iron,<br />

or chromium intake might be responsible for liver<br />

injury. 179,180,198,199<br />

The earliest and most sensitive, but not specific<br />

laboratory markers are serum alkaline phosphatase<br />

and γ-glutamyl transferase activities, while hyperbilirubinemia<br />

is the latest marker of cholestasis to<br />

appear. Steatosis is the first non-specific histological<br />

abnormality. Steatosis may result either from<br />

excessive glucose supply leading to lipogenesis, or<br />

from the deposition of exogenous IVFE. Clinical<br />

liver enlargement, confirmed by ultrasonography,<br />

may appear within a few days after PN onset.<br />

Cholestasis together with portal and periportal cell<br />

infiltration leads to fibrosis. This indicates severe<br />

liver disease, which can lead to cirrhosis and liver<br />

failure, but is fortunately rare if PN is performed<br />

correctly. More recently, the association of<br />

cholestasis with thrombocytopenia has been<br />

described in patients on PN, 114 but the link<br />

between liver disease and the RES overload needs<br />

to be better understood.<br />

Careful monitoring of hepatic function is extremely<br />

important in order to minimize factors responsible<br />

for liver disease. Some measures have been found to<br />

limit or reverse liver disease and may include:<br />

(1) The stimulation of the entero-biliary axis by<br />

ingestion of LCTs or breast milk, or by injection<br />

of cholecystokinin analogs; 200<br />

(2) The reduction of intraluminal bacterial overgrowth<br />

caused by intestinal stasis by giving<br />

metronidazole and/or performing tapering<br />

enteroplasty; 201<br />

(3) The use of ursodesoxycholic acid (10–20mg/kg<br />

per day) or tauroursodeoxycholic acid could<br />

contribute in decreasing liver injury; 202,203<br />

PN intake should be adapted by:<br />

(1) Limiting glucose intakes to reduce hepatic fat<br />

accumulation; 204–207<br />

(2) Using the appropriate type and amount of<br />

intravenous fat emulsion, which provides<br />

essential fatty acids, reduces glucose load and<br />

limits peroxidation; 133,196,208<br />

(3) Controlling the lipid supply and rate of delivery<br />

and/or stopping IVFEs as soon as thrombocytopenia,<br />

hyperbilirubinemia and/or jaundice<br />

appear; 114<br />

(4) Using the new pediatric-adapted amino acid<br />

solutions which provide appropriate amino<br />

acids as well as taurine; 144,209<br />

(5) Performing cyclic PN, which helps to reduce<br />

hyperinsulinism and liver steatosis; 25–32<br />

(6) Adapting the iron intake and decreasing<br />

aluminum content of the PN solution. 210,211<br />

When cholestasis occurs, biliary obstruction, infection<br />

or drug toxicity should be ruled out by appropriate<br />

investigations. Decrease in platelet count<br />

below 150 000/mm 3 associated with an increase in<br />

plasma transaminases, and a further increase in<br />

plasma bilirubin, should lead to strong suspicion of<br />

lipid toxicity when all other explanations are ruled<br />

out. Bone marrow aspiration, liver biopsy and<br />

temporary suspension or decrease in lipid infusion<br />

should be discussed. Lipid infusion should be<br />

stopped until normalization of bilirubin level and<br />

platelet count. Essential fatty acid deficiency must<br />

be carefully checked. In case of persistent cholestasis<br />

after a 2-month suspension of lipid infusion, an<br />

irreversible course of the liver disease should be<br />

suspected. Reintroduction of lipid emulsions<br />

should be attempted under strict supervision, below<br />

the previous dosage, beginning with 1 or 2 perfusions<br />

per week. Medium-chain-based emulsions<br />

(50% MCT) might help to minimize the LCT load<br />

and theoretically their hepatic deposition and longterm<br />

toxicity, but neither the current study nor


other clinical data have demonstrated any effects of<br />

MCT-based emulsions to prevent or to reverse longterm<br />

PN-associated cholestasis or hematological<br />

complications. Emulsions based on olive oil might<br />

also reduce the risk of lipid toxicity due to peroxidation,<br />

by decreasing the amount of linoleic acid.<br />

Conclusions<br />

Parenteral nutrition has become a widely used therapeutic<br />

option in clinical nutrition. One of the goals<br />

of PN is to have the same nutritional efficacy as that<br />

of normal oral feeding. Clinical research in the field<br />

REFERENCES<br />

1. Dudrick SJ, Wilmore DW, Vars HM, Rhoads JE. Long-term<br />

total parenteral nutrition with growth, development and<br />

positive nitrogen balance. Surgery 1968; 64: 134–142.<br />

2. Wilmore DM, Dudrick SJ. Growth and development of<br />

an infant receiving all nutrients by vein. JAMA 1968;<br />

203: 860–864.<br />

3. Goulet O, Brousse N, Canioni D et al. Syndrome of<br />

intractable diarrhoea with persistent villus atrophy in<br />

early childhood: a clinicopathological survey of 47<br />

cases. J Pediatr Gastroenterol Nutr 1998; 26: 151–161.<br />

4. Goulet O. Intractable diarrhea of infancy. In Walker A et<br />

al., eds. Pediatric Gastrointestinal Disease. Decker, 2000:<br />

762–772.<br />

5. Guarino A, Immacolata Spagnulo M, Russo S et al.<br />

Etiology and risk factors of severe and protracted diarrhea.<br />

J Pediatr Gastroenterol Nutr 1995; 20: 173–178.<br />

6. Lerebours E, Messing B, Chevalier B et al. An evaluation<br />

of total parenteral nutrition in the management of<br />

steroid-dependent and steroidresistant patients with<br />

Crohn’s disease. J Parent Ent Nutr 1988; 10: 274–278.<br />

7. Tsujikawa T, Andoh A, Fujiyama Y. Enteral and<br />

parenteral nutrition therapy for Crohn’s disease. Curr<br />

Pharm Des 2003; 9: 323–332.<br />

8. Goulet O, Ricour C. The short bowel syndrome. In Buts<br />

E, Sokal JP, eds. Management of Digestive and Liver<br />

Disorders in Infants and Children. Elsevier Sciences<br />

Publisher, 1993: 307–318.<br />

9. Goulet O, Revillon Y, Jan D et al. <strong>Neonatal</strong> short bowel<br />

syndrome. J Pediatr 1991; 119: 18–23.<br />

10. Sondheimer JM, Cadnapaphornchai M, Sontag M, Zerbe<br />

GO. Predicting the duration of dependence on<br />

parenteral nutrition after neonatal intestinal resection. J<br />

Pediatr 1998; 132: 80–84.<br />

11. Leonberg BL, Chuang E, Eicher P et al. Long-term<br />

growth and development in children after home<br />

parenteral nutrition. J Pediatr 1998; 132: 461–466.<br />

Conclusions 573<br />

of PN is necessary. This research has three main<br />

objectives: the reduction of PN-related complications;<br />

the study of the tolerance and efficacy of new<br />

substrates (lipids, amino acids, growth factors, etc);<br />

and the study of energy and/or protein metabolism<br />

during PN, especially in children in whom growth<br />

is the main goal of nutritional support.<br />

Parenteral nutrition should be reserved for appropriate<br />

indications and avoided each time the enteral<br />

route is able to ensure adequate nutritional intakes<br />

for correction of malnutrition and/or normal<br />

growth. The successful deployment of a multidisciplinary<br />

nutrition support team minimizes inappropriate<br />

prescription of parenteral nutrition.<br />

12. Dellert SF, Farrell MK, Specker BL, Heubi JE. Bone<br />

mineral content in children with short bowel syndrome<br />

after discontinuation of parenteral nutrition. J Pediatr<br />

1998; 132: 516–519.<br />

13. Festen S, Brevoord JC, Goldhoorn GA et al. Excellent<br />

long-term outcome for survivors of apple peel atresia. J<br />

Pediatr Surg 2002; 37: 61–65.<br />

14. Rickard KA, Godshall B, Loghmani ES. Integration of<br />

nutrition support into oncology treatment protocols for<br />

high and low nutritional risk children with Wilm’s<br />

tumor. Cancer 1989; 64: 491–509.<br />

15. Gomez-Almaguer D, Montemayor J, Gonzales–Llano O<br />

et al. Improvement in the nutritional status of children<br />

with standard-risk acute lymphoblastic leukemia is<br />

associated with better tolerance to continuation<br />

chemotherapy. Int J Pediatr Hematol Oncol 1995; 2:<br />

53–56.<br />

16. Colomb V. Nutrition en cancérologie : aspects pédiatriques.<br />

Nutr Clin Metabol 2001; 15: 325–334.<br />

17. Goulet O, De Ville De Goyet J, Otte JB, Ricour C.<br />

Preoperative nutritional evaluation and support for liver<br />

transplantation in children. Transplant Proc 1987; 19:<br />

3249–3255.<br />

18. Moukarzel AA, Najm I, Vargas J et al. Effect of nutritional<br />

status on outcome of orthotopic liver transplantation<br />

in pediatric patients. Transplant Proc 1990; 22:<br />

1560–1563.<br />

19. Sheperd RW, Chin SE, Cleghorn GJ et al. Malnutrition<br />

in children with chronic liver disease accepted for liver<br />

transplantation: clinical profile and effect in outcome. J<br />

Pediatr Health 1991; 27: 295–299.<br />

20. Roggero P, Catalotti E, Ulla L et al. Factors influencing<br />

malnutrition in children waiting for liver transplantation.<br />

Am J Clin Nutr 1997; 65: 1852–1857.<br />

21. Matuchansky C, Messing B, Jeejeebhoy et al. Cyclical<br />

parenteral nutrition. Lancet 1992; 340: 588–592.


574<br />

Parenteral nutrition in infants and children<br />

22. Just B, Messing B, Darmaun D et al. Comparison of<br />

substrate utilization by indirect calorimetry during<br />

cyclic and continuous total parenteral nutrition. Am J<br />

Clin Nutr 1990; 51: 107–111.<br />

23. Gramlich LM. Cyclic parenteral nutrition: considerations<br />

of carbohydrate and lipid metabolism. Nutr Clin<br />

Pract 1994; 9: 49–50.<br />

24. Colomb V, Charbit M, Goulet O et al. Renal function in<br />

young patients on long-term cyclic parenteral nutrition.<br />

Transplant Proc 1996; 28: 2785.<br />

25. MacFie J. Cyclic parenteral nutrition. Nutrition 1997; 13:<br />

46–48.<br />

26. Nicol JJ, Hoagland RL, Heitlinger LA. The prevalence of<br />

nausea and vomiting in pediatric patients receiving<br />

home parenteral nutrition. Nutr Clin Pract 1995; 10:<br />

189–192.<br />

27. Collier S, Crough J, Hendricks K, Caballero B. Use of<br />

cyclic parenteral nutrition in infants less than 6 months<br />

of age. Nutr Clin Pract 1994; 9: 65–68.<br />

28. Lienhardt A, Rakotoambinina B, Colomb V et al. Insulin<br />

secretion and sensitivity in children on cyclic total<br />

parenteral nutrition. JPEN J Parenter Enteral Nutr 1998;<br />

22: 382–386.<br />

29. Colomb V, Dabbas M, Goulet O et al. Prepubertal growth<br />

in children with long-term parenteral nutrition. Horm<br />

Res 2002; 58 (Suppl 1): 2–6.<br />

30. Beghin L, Michaud L, Hankard R et al. Total energy<br />

expenditure and physical activity in children treated<br />

with home parenteral nutrition. Pediatr Res 2003; 53:<br />

684–690.<br />

31. Hwang TL, Lue MC, Chen LL. Early use of cyclic TPN<br />

prevents further deterioration of liver functions for the<br />

TPN patients with impaired liver function.<br />

Hepatogastroenterology 2000; 47: 1347–1350.<br />

32. Vargas JH, Ament ME, Berquist WE et al. Long-term<br />

home parenteral nutrition in pediatrics: ten years of<br />

experience in 102 patients. J Pediatr Gastroenterol Nutr<br />

1987; 6: 24–32.<br />

33. Ricour C, Gorski AM, Goulet O et al. Home parenteral<br />

nutrition in children: 8 years of experience with 112<br />

patients. Clin Nutr 1990; 9: 65–71.<br />

34. Messing B, Lemann M, Landais P et al. Prognosis of<br />

patients with chronic intestinal failure receiving longterm<br />

home parenteral nutrition in France and Belgium.<br />

Gastroenterology 1995; 108: 1005–1010.<br />

35. Howard L, Ament M, Fleming R et al. Current use and<br />

clinical outcome of home parenteral and enteral nutrition<br />

therapies in the United States. Gastroenterology<br />

1995; 109: 355–365.<br />

36. Howard L, Hassan N. Home parenteral nutrition: 25<br />

years later. Clin Nutr 1998; 27: 418–512.<br />

37. Colomb V, Goulet O, Ricour C. Home enteral and<br />

parenteral nutrition. Baillière’s Clin Gastroenterol 1998;<br />

122: 877–894.<br />

38. Grant D. Intestinal transplantation: 1997 Report of the<br />

International Registry. Transplantation 1999; 15:<br />

1061–1064.<br />

39. Goulet O, Jan D, Lacaille F et al. Intestinal transplantation<br />

in children: preliminary experience in Paris. JPEN J<br />

Parenter Enteral Nutr 1999; 23: S121–S125.<br />

40. Goulet O, Lacaille F, Jan D, Ricour C. Intestinal transplantation:<br />

indications, results and strategy. Curr Opin<br />

Clin Nutr Metab Care 2000; 3: 329–333.<br />

41. Goulet O, Ruemmele F, Lacaille F, Colomb V. Intestinal<br />

failure. J Pediatr Gastroenterol Nutr 2003; (in press).<br />

42. Chung DH, Ziegler MM. Central venous catheter access.<br />

Nutrition 1998; 14: 119–123.<br />

43. Jan D, Goulet O. Accès vasculaires. In Ricour C, Ghisolfi<br />

J, Putet G, Goulet O, eds. Traité de Nutrition Pédiatrique.<br />

Paris: Maloine Editeur, 1993: 831–838.<br />

44. Shulman R, Pokorny W, Martin CG et al. Comparison of<br />

percutaneous and surgical placement of central venous<br />

catheter in neonates. J Pediatr Surg 1986; 21:348–350.<br />

45. Ahn WS, Kang JS. An easy technique for long term<br />

central venous catheterization and subcutaneous<br />

tunneling of the silastic catheter in neonates and<br />

infants. J Pediatr Surg 1986; 21: 344–347.<br />

46. Weber TR, West KW, Grosfeld JL. Broviac central venous<br />

catheterization in infants and children. Am J Surg 1983;<br />

l45: 202–204.<br />

47. Reinberg O, Beck D. A new implantable device for longterm<br />

vascular access in children. Eur J Pediatr Surg<br />

1992; 2: 183–185.<br />

48. Ricour C, Revillon Y, Bougle D et al. Nutrition parentérale<br />

par fistule artérioveineuse. Arch Fr Pediatr 1983;<br />

40: 457–460.<br />

49. Schofield W. Predicting basal metabolic rate, new standards<br />

and review of previous work. Hum Nutr Clin Nutr<br />

1985; 39 (Suppl 1); 5–41.<br />

50. Goulet O. Assessment of nutritional status in clinical<br />

practice. Baillière’s Clin Gastroenterol 1998; 12:<br />

645–669.<br />

51. Duro D, Rising R, Cole C et al. New equations for calculating<br />

the components of energy expenditure in infants.<br />

J Pediatr 2002; 140: 534–539.<br />

52. Goran MI, Shewchuk R, Gower BA. Longitudinal<br />

changes in fatness in white children: no effect of childhood<br />

energy expenditure. Am J Clin Nutr 1998; 67:<br />

309–316.<br />

53. Pierro A, Jones MO, Donnell SC. Total parenteral nutrition<br />

in surgical infants. Biochem Soc Trans 1998; 26:<br />

131–136.<br />

54. Jaksic T, Shew SB, Keshen TH et al. Do critically ill<br />

surgical neonates have increased energy expenditure? J<br />

Pediatr Surg 2001; 36: 63–67.<br />

55. Lloyd DA. Energy requirements of surgical newborn<br />

infants receiving parenteral nutrition. Nutrition 1998;<br />

14: 101–104.<br />

56. Garza JJ, Shew SB, Keshen TH et al. Energy expenditure<br />

in ill premature neonates. J Pediatr Surg 2002; 37:<br />

289–293.<br />

57. Coss-Bu JA, Klish WJ, Walding D et al. Energy metabolism,<br />

nitrogen balance, and substrate utilization in critically<br />

ill children 1–3. Am J Clin Nutr 2001; 74: 664–669.<br />

58. Koen FM, Joosten KFM, Verhoeven JJ, Hazelzet JA.<br />

Energy expenditure and substrate utilization in mechanically<br />

ventilated children. Nutrition 1999; 15: 444–448.<br />

59. Turi RA, Petros AJ, Eaton S et al. Energy metabolism of<br />

infants and children with systemic inflammatory<br />

response syndrome and sepsis. Ann Surg 2001; 233:<br />

581–587.<br />

60. White MS, Shepherd RW, McEniery JA. Energy expenditure<br />

measurements in ventilated critically ill children:<br />

within- and between-day variability. J Parenter Enteral<br />

Nutr 1999; 23: 300–304.<br />

61. Pierro A, Jones MO, Hammond P et al. A new equation<br />

to predict the resting energy expenditure of surgical<br />

infants. J Pediatr Surg 1994; 29: 1103–1108.<br />

62. White MS, Shepherd RW, McEniery JA. Energy expenditure<br />

in 100 ventilated, critically ill children: improving<br />

the accuracy of predictive equations. Crit Care Med<br />

2000; 28: 2307–2312.<br />

63. Kalhan SC, Kilic I. Carbohydrate as nutrient in the<br />

infant and child: range of acceptable intake. Europ J<br />

Clin Nutr 1999; 53: S94–S100.<br />

64. Denne SC, Karn CA, Wang J, Liechty EA. Effect of intravenous<br />

glucose and lipid on proteolysis and glucose<br />

production in normal newborns. Am J Physiol 1995;<br />

269: E361–E367.


65. Sunehag AL, Haymond MW, Schanler RJ et al.<br />

Gluconeogenesis in very low birth weight infants<br />

receiving total parenteral nutrition. Diabetes 1999; 48:<br />

791–800.<br />

66. Sheridan RL, Yu YM, Prelack K et al. Maximal<br />

parenteral glucose oxidation in hypermetabolic young<br />

children: a stable isotope study. J Parenter Enteral Nutr<br />

1998; 22: 212–216.<br />

67. Lafeber HN, Sulkers EJ, Chapman TE, Sauer PJ. Glucose<br />

production and oxidation in preterm infants during<br />

total parenteral nutrition. Pediatr Res 1990; 28: 153–157.<br />

68. Jones MO, Pierro A, Hammond P et al. Glucose utilization<br />

in the surgical newborn infant receiving total<br />

parenteral nutrition. J Pediatr Surg 1993; 28: 1121–1125.<br />

69. Nose O, Tipton JR, Ament ME. Effect of the energy<br />

source on changes in energy expenditure, respiratory<br />

quotient, and nitrogen balance during total parenteral<br />

nutrition in children. Pediatr Res 1987; 21: 538–541.<br />

70. Bresson JL, Narcy P, Putet G, Ricour C. Energy substrate<br />

utilization in infants receiving total parenteral nutrition<br />

with different glucose to fat ratios. Pediatr Res 1989; 25:<br />

645–648.<br />

71. Elwyn DH, Askanazi J, Kinney JM, Gump FE. Kinetics<br />

of energy substrates. Acta Chir Scand Suppl 1981; 507:<br />

209–219.<br />

72. Koretz RL, Lipman TO, Klein S. AGA Technical review<br />

on parenteral nutrition. Gastroenterology 2001; 121:<br />

970–1001.<br />

73. Robin AP, Carpentier YA, Askanazi J et al. Metabolic<br />

consequences of hypercaloric glucose infusions. Acta<br />

Chir Belg 1981; 80: 133–140.<br />

74. Bresson JL. Protein and energy requirements. Baillière’s<br />

Clin Gastroenterol 1998; 12: 631–646.<br />

75. Talpers SS, Romberger DJ, Bunce SB, Pingleton SK.<br />

Nutritionally associated increased carbon dioxide<br />

production. Excess total calories vs high proportion of<br />

carbohydrate calories. Chest 1992; 102: 551–555.<br />

76. Askanazi J, Weissman C, LaSala PA et al. Effect of<br />

protein intake on ventilatory drive. Anesthesiology 1984;<br />

60: 106–10.<br />

77. Rodriguez JL, Askanazi J, Weissman C et al. Ventilatory<br />

and metabolic effects of glucose infusions. Chest 1985;<br />

88: 512–518.<br />

78. Burke JF, Wolfe RR, Mullany CJ et al. Glucose requirements<br />

following burn injury. Parameters of optimal<br />

glucose infusion and possible hepatic and respiratory<br />

abnormalities following excessive glucose intake. Ann<br />

Surg 1979; 190: 274–285.<br />

79. Tulikoura I, Huikuri K. Morphological fatty changes and<br />

function of the liver, serum free fatty acids, and triglycerides<br />

during parenteral nutrition. Scand J Gastroenterol<br />

1982; 17: 177–185.<br />

80. Aarsland A, Chinkes D, Wolfe RR. Contributions of de<br />

novo synthesis of fatty acids to total VLDL-triglyceride<br />

secretion during prolonged hyperglycemia/hyperinsulinemia<br />

in normal man. J Clin Invest 1996; 98:<br />

2008–2017.<br />

81. Klein CJ, Stanek GS, Wiles CE 3rd. Overfeeding<br />

macronutrients to critically ill adults: metabolic complications.<br />

J Am Diet Assoc 1998; 98: 795–806.<br />

82. The Veterans Affairs Total Parenteral Nutrition<br />

Cooperative Study group. Perioperative total parenteral<br />

nutrition in surgical patients. N Engl J Med 1991; 325:<br />

525–532.<br />

83. Kudsk KA, Croce MA, Fabian TC et al. Enteral versus<br />

parenteral feeding. Effects on septic morbidity after<br />

blunt and penetrating abdominal trauma. Ann Surg<br />

1992; 215: 503–511.<br />

84. Moore FA, Feliciano DV, Andrassy RJ et al. Early enteral<br />

feeding, compared with parenteral, reduces postopera-<br />

References 575<br />

tive septic complications. The results of a metaanalysis.<br />

Ann Surg 1992; 216: 172–183.<br />

85. Cruccetti A, Pierro A, Uronen H, Klein N. Surgical<br />

infants on total parenteral nutrition have impaired<br />

cytokine responses to microbial challenge. J Pediatr Surg<br />

2003; 38: 138–142.<br />

86. Khaodhiar L, McCowen K, Bistrian B. Perioperative<br />

hyperglycemia, infection or risk? Curr Opin Clin Nutr<br />

Metab Care 1999; 2: 79–82.<br />

87. Engelich G, Wright DG, Hartshorn KL. Acquired disorders<br />

of phagocyte function complicating medical and<br />

surgical illnesses. Clin Infect Dis 2001; 33: 2040–2048.<br />

88. Bistrian BR. Hyperglycemia and infection: which is the<br />

chicken and which is the egg? J Parenter Enteral Nutr<br />

2001; 25: 180–181.<br />

89. Gore DC, Chinkes D, Heggers J. Association of hyperglycemia<br />

with increased mortality after severe burn<br />

injury. J Trauma 2001; 51: 540–544.<br />

90. Macfie J, Smith RC, Hill GL. Glucose or fat as a nonprotein<br />

energy source? A controlled clinical trial in<br />

gastroenterological patients requiring intravenous nutrition.<br />

Gastroenterology 1981; 80: 103–107.<br />

91. Pineault M, Chessex P, Bisaillon S, Brisson G. Total<br />

parenteral nutrition in the newborn: impact of the<br />

quality of infused energy on nitrogen metabolism. Am J<br />

Clin Nutr 1988; 47: 298–304.<br />

92. Bresson JL, Bader B, Rocchiccioli F et al. Protein-metabolism<br />

kinetics and energy-substrate utilization in infants<br />

fed parenteral solutions with different glucose-fat ratios.<br />

Am J Clin Nutr 1991; 54: 370–376.<br />

93. Lefkowitz JB, Evers AS, Elliott WJ, Needleman P.<br />

Essential fatty deficiency: a new look at an old problem.<br />

Prostaglandins Leukot Med 1986; 23: 123–127.<br />

94. Roesner M, Grant JP. Intravenous lipid emulsions. Nutr<br />

Clin Pract 1987; 1: 96–107.<br />

95. Goulet O, Duhamel JF, Ricour C. Nutritional problems.<br />

In Tinker J, Zapol W, eds. Care in the Critically Ill<br />

Patients, Berlin, Heidelberg, New York, Springer Verlag<br />

1992: 1415–1436.<br />

96. Adolph M. Lipid emulsions in parenteral nutrition. Ann<br />

Nutr Metabol 1999; 43: 1–13.<br />

97. Koletzko B. Lipid supply and metabolism in infancy.<br />

Curr Opin Clin Nutr Metab Care 1998; 1: 171–177.<br />

98. Bistrian BR. Clinical aspects of essential fatty acid<br />

metabolism: Jonathan Rhoads Lecture. JPEN J Parenter<br />

Enteral Nutr 2003; 27: 168–175.<br />

99. Olivecrona G, Olivecrona T. Clearance of artificial triacylglycerol<br />

particles. Curr Opin Clin Nutr Metab Care<br />

1998; 1: 143–151.<br />

100. Carpentier YA. Intravascular metabolism of fat emulsions:<br />

the Arvid Wretling lecture ESPEN 1988. Clin Nutr<br />

1989; 8: 115–125.<br />

101. Carpentier YA, Simoens C, Siderova V et al. Recent<br />

developments in lipid emulsions: relevance to intensive<br />

care. Nutrition 1997; 13: 735–785.<br />

102. Kao LC, Cheng MH, Warburton D. Triglycerides, free<br />

fatty acids, free fatty acids/albumin molar ratio, and<br />

cholesterol levels in serum of neonates receiving longterm<br />

lipid infusions: controlled trial of continuous and<br />

intermittent regimens. J Pediatr 1984; 104: 429–435.<br />

103. Haumont D, Deckelbaum RJ, Richelle M et al. Plasma<br />

lipid and plasma lipoprotein concentrations in low birth<br />

weight infants given parenteral nutrition with twenty or<br />

ten percent lipid emulsion. J Pediatr 1989; 115:<br />

787–793.<br />

104. Spear ML, Stahl GE, Hamosh M et al. Effect of heparin<br />

dose and infusion rate on lipid clearance and bilirubin<br />

binding in premature infants receiving intravenous fat<br />

emulsions. J Pediatr 1988; 112: 94–98.


576<br />

Parenteral nutrition in infants and children<br />

105. Haumont D, Richelle M, Deckelbaum RJ et al. Effect of<br />

liposomal content of lipid emulsions on plasma lipid<br />

concentrations in low birth weight infants receiving<br />

parenteral nutrition. J Pediatr 1992; 121: 759–763.<br />

106. Putet G. Lipid metabolism of the micropremie. Clin<br />

Perinatol 2000; 27: 57–69.<br />

107. Defronzo RA, Jacot E, Jequier E et al. The effect of<br />

insulin on the disposal of intravenous glucose. Diabetes<br />

1981; 30: 1000–1007.<br />

108. Stein PT, Mullen JL. Hepatic fat accumulation in man<br />

with excess parenteral glucose. Nutr Res 1985; 5:<br />

1347–1351.<br />

109. Bresson JL, Narcy P, Putet G et al. Energy substrate<br />

utilization in infants receiving total parenteral nutrition<br />

with different glucose to fat ratios. Pediatr Res 1989; 25:<br />

645.<br />

110. Salas J, Girardet JP, De Potter S et al. Glucose versus<br />

glucose-fat mixture in the course of total parenteral<br />

nutrition: effects on substrate utilization and energy<br />

metabolism in malnourished children. Clin Nutr 1991;<br />

10: 272–278.<br />

111. Pierro A, Carnielli V, Filler RM et al. Metabolism of<br />

intravenous fat emulsion in the surgical newborn. J<br />

Pediatr Surg 1989; 24: 95.<br />

112. Palmblad J. Intravenous lipid emulsions and host<br />

defense a critical review. Clin Nutr 1991; 10: 303–308.<br />

113. Goulet O, Girot R, Maier-Redelsperger M et al.<br />

Hematologic disorders following prolonged use of intravenous<br />

fat in children. JPEN 1986; 10: 284–288.<br />

114. Colomb V, Jobert-Giraud A, Lacaille F et al. Role of lipid<br />

emulsions in cholestasis associated with long-term<br />

parenteral nutrition in children. JPEN 2000; 24:<br />

345–350.<br />

115. Tanner A, Keyhani A, Reiner R et al. Proteolytic<br />

enzymes released by liver macrophages may promote<br />

hepatic injury in a rat model of hepatic damage.<br />

Gastroenterology 1981; 80: 647–654.<br />

116. Neuzil J, Darlow BA, Inder TE et al. Oxidation of<br />

parenteral lipid emulsion by ambient and phototherapy<br />

lights: potential toxicity of routine parenteral feeding. J<br />

Pediatr 1995; 126: 785–790.<br />

117. Clayton PT, Bowron A, Mills KA et al. Phytosterolemia<br />

in children with parenteral nutrition-cholestatic associated<br />

liver disease. Gastroenterology 1993; 105:<br />

1806–1813.<br />

118. Bach AC, Frey A, Lutz O. Clinical and experimental<br />

effects of medium-chain triglyceride based fat emulsions<br />

– A review. Clin Nutr 1989; 8: 223–235.<br />

119. Dennison AR, Ball M, Crowe PJ et al. The metabolic<br />

consequences of infusing emulsions containing<br />

medium-chain triglycerides for parenteral nutrition: a<br />

comparative study with conventional lipid. Ann Royal<br />

Coll Surg Engl 1986; 68: 119–122.<br />

120. Dennison AR, Ball M, Hands LJ et al. Total parenteral<br />

nutrition using conventional and medium-chain triglycerides:<br />

effects on liver functions tests, complement and<br />

nitrogen balance. JPEN 1988; 12: 15–19.<br />

121. Bach AC, Guiraud M, Gibault JP et al. Medium-chain<br />

triglycerides in septic patients on total parenteral nutrition.<br />

Clin Nutr 1988; 7: 157–163.<br />

122. Beaufrère B, Chassard D, Broussolle C et al. Effects of Dβ-hydrohyxybutyrate<br />

and long- and medium-chain<br />

triglycerides on leucine metabolim in humans. Am J<br />

Physiol 1992; 262: E268–E274.<br />

123. Lima LAM. <strong>Neonatal</strong> parenteral nutrition with medium<br />

chain triglycerides: rationale for research. J Parenter<br />

Enteral Nutr 1989; l3: 312–317.<br />

124. Bresson JL, Narcy P, Sachs C et al. Energy substrate<br />

competition: comparative study of LCT and MCT<br />

utilization during continuous TPN in infants. Clin Nutr<br />

1986; 5 (Suppl): 54(A).<br />

125. Goulet O, De Potter S, Postaire M et al. Long term total<br />

parenteral nutrition in children: utilization of medium<br />

chain triglycerides. Nutrition 1992; 8: 33–37.<br />

126. Rubin M, Moser A, Naor N et al. Effect of three intravenously<br />

administered fat emulsions containing different<br />

concentrations of fatty acids on the plasma fatty<br />

acids composition of premature infants. J Pediatr 1994;<br />

125: 596–602.<br />

127. Maiz A, Yamazaki K, Sobrado J et al. Protein metabolism<br />

during total parenteral nutrition in injured rats<br />

using medium-chain triglycerides. Metabolism 1984; 33:<br />

901–909.<br />

128. Chambrier C, Guiraud M, Gibault JP et al. Medium- and<br />

long-chain triacylglycerols in postoperative patients:<br />

structured lipids versus a physical mixture. Nutrition<br />

1999; 15: 274–277.<br />

129. Waitzberg DL, Lotierzo PH, Logullo AF et al. Parenteral<br />

lipid emulsions and phagocytic systems. Br J Nutr 2002;<br />

87 (Suppl 1): S49–S57.<br />

130. Mayer K, Gokorsch S, Fegbeutel C et al. Parenteral nutrition<br />

with fish oil modulates cytokine response in<br />

patients with sepsis. Am J Respir Crit Care Med 2003;<br />

167: 1321–1328.<br />

131. Louheranta AM, Porkkala-Sarataho EK, Nyssönen MK et<br />

al. Linoleic acid intake and susceptibility of very-lowdensity<br />

and low-density lipoproteins to oxidation in<br />

man. Am J Clin Nutr 1996; 63: 698–703.<br />

132. Brouwer CB, de Bruin TWA, Jansen H, Erkelens DW.<br />

Different clearance of intravenously administered olive<br />

oil and soybean-oil emulsions: role of hepatic lipase.<br />

Am J Clin Nutr 1993; 57: 533–539.<br />

133. Goulet O, de Potter S, Antebi H et al. Long-term efficacy<br />

and safety of a new olive oil-based intravenous fat<br />

emulsion in pediatric patients: a double-blind randomized<br />

study. Am J Clin Nutr 1999; 70: 338–345.<br />

134. Göbel Y, Koletzko B, Böhles HJ et al. Parenteral fat emulsions<br />

based on olive and soybean oils: a randomized<br />

clinical trial in preterm infants. J Pediatr Gastroenterol<br />

Nutr 2003; 37: 161–67.<br />

135. Borum PR. Should carnitine be added to parenteral<br />

nutrition solutions? Nutr Clin Prac 2000; 15: 153–154.<br />

136. Penn D, Schmidt-Sommerfeld E, Pascu F. Decreased<br />

tissue carnitine concentrations in newborn infants<br />

receiving total parenteral nutrition. J Pediatr 1981; 98:<br />

976–978.<br />

137. Rawcliffe PM, Giles P, Barlett S et al. Carnitine as a<br />

possible adjunct in parenteral feeding. Clin Nutr 1984;<br />

3: 141–145.<br />

138. Penn D, Ludwigs B, Schmidt-Sommerfied E, Pascu F.<br />

Effect of nutrition on tissue carnitine concentrations in<br />

infants of different gestational ages. Biol Neonate 1985;<br />

47: 130–135.<br />

139. Cairns PA, Stalker DJ. Carnitine supplementation of<br />

parenterally fed neonates. Cochrane Database Syst Rev<br />

2000; 4: CD000950.<br />

140. Pellett PL. Protein requirements in humans. Am J Clin<br />

Nutr 1990; 51: 723–737.<br />

141. Eason LB, Halata MS, Dweck HS. Parenteral nutrition in<br />

the newborn: a practical guide. Pediatr Clin North Am<br />

1982; 29: 1171–1190.<br />

142. Goulet O, De Potter S, Salas J et al. Leucine metabolism<br />

at graded amino-acid intakes in children receiving<br />

parenteral nutrition. Am J Physiol 1993; 265:<br />

E540–E546.<br />

143. Bengoa JM, Sitrin MD, Wood RJ, Rosenberg IH. Aminoacid<br />

induced hypercalciuria in patients on total<br />

parenteral nutrition. Am J Clin Nutr 1983; 38: 264–269.


144. Coran AG, Drongowski RA. Studies on the toxicity and<br />

efficacy of a new amino acid solution in pediatric<br />

parenteral nutrition. J Parenter Enter Nutr 1987; 11:<br />

368–377.<br />

145. Corriol. O. Les solutions injectables d’acides aminés<br />

pour nutrition parentérale. Critères de choix thérapeutiques.<br />

Nutr Clin Metabol 1987; 1: 17–30.<br />

146. Wolfe RR, Goodenough RD, Burke JF, Wolfe MM.<br />

Response of protein and urea kinetics in burn patients<br />

to different level of protein intake. Ann Surg 1983; 197:<br />

163–171.<br />

147. Neu J, DeMarco V, Li N. Glutamine: clinical applications<br />

and mechanisms of action. Curr Opin Clin Nutr Metab<br />

Care 2002; 5: 69–75.<br />

148. Hartmann F, Plauth M. Intestinal glutamine metabolism.<br />

Metabolism 1989; 38: 18–24.<br />

149. Hankard R, Goulet O, Ricour C et al. Glutamine metabolism<br />

in children with short bowel syndrome: a stable<br />

isotope study. Pediatr Res 1994; 36: 202–206.<br />

150. Calder PC. Glutamine and the immune system. Clin<br />

Nutr 1994; 13: 2–8.<br />

151. Schloerb PR, Amare M. Total parenteral nutrition with<br />

glutamine in bone marrow transplantation and other<br />

clinical applications. J Parent Enter Nutr 1993; 17:<br />

407–413.<br />

152. Young LS, Bye R, Scheltinga M et al. Patients receiving<br />

glutamine supplemented intravenous J Parenter Enteral<br />

Nutr 1993; 17: 422–427.<br />

153. Ziegler TR, Young LS, Benfell K et al. Clinical and metabolic<br />

efficacy of glutamine supplemented parenteral<br />

nutrition after bone marrow transplantation. Ann Int<br />

Med 1992; 116: 821–828.<br />

154. Furst P, Stehle P. The potential use of parenteral dipeptides<br />

in clinical nutrition. Nutr Clin Pract 1993; 8:<br />

106–114.<br />

155. Tremel H, Kienle B, Weilemann LS et al. Glutamine<br />

dipeptide supplemented parenteral nutrition maintains<br />

intestinal function in the critically ill. Gastroenterology<br />

1994; 107: 1595–1601.<br />

156. Jiang ZM, Cao JD, Zhu XG et al. The impact of glutamine<br />

dipeptide on nitrogen balance, intestinal permeability<br />

and clinical outcome of post operative patients.<br />

JPEN 1999; 23: S62–S66.<br />

157. Hammarqvist F, Wernerman J, Von Der Decken A,<br />

Vinnars E. Alpha ketoglutarate preserves protein<br />

synthesis and free glutamine in skeletal muscle after<br />

surgery. Surgery 1991; 109: 28–31.<br />

158. Wernerman J, Hammarkvist F, Ali MR, Vinnars E.<br />

Glutamine and ornithine a ketoglutarate but not<br />

branched chain amino acids reduce the loss of muscle<br />

glutamine after surgical trauma. Metabolism 1989; 38:<br />

63–66.<br />

159. Cynober LA. The use of alpha-ketoglutarate salts in clinical<br />

nutrition and metabolic care. Curr Opin Clin Nutr<br />

Metab Care 1999; 2: 33–37.<br />

160. Dumas F, De Bandt JP, Colomb V et al. Enteral ornithine<br />

alpha-ketoglutarate enhances intestinal adaptation to<br />

massive resection in rats. Metabolism 1998; 47:<br />

1366–1371.<br />

161. Moukarzel AA, Goulet O, Salas JS et al. Growth retardation<br />

in children receiving long-term total parenteral<br />

nutrition: effects of ornithine alpha-ketoglutarate. Am J<br />

Clin Nutr 1994; 60: 408–413.<br />

162. Rollins CJ, Elsberry VA, Pollack KA et al. Three-in-one<br />

parenteral nutrition: a safe and economical method of<br />

nutritional support for infants. J Parenter Enteral Nutr<br />

1990; 14: 290–294.<br />

163. Didier ME, Fischer S, Maki DG. Total nutrient admixtures<br />

appear safer than lipid emulsion alone as regards<br />

References 577<br />

microbial contamination: growth properties of microbial<br />

pathogens at room temperature. J Parenter Enteral Nutr<br />

1998; 22: 291–296.<br />

164. Hardy G, Ball P, McElroy B. Basic principles for<br />

compounding all-in-one parenteral nutrition admixtures.<br />

Curr Opin Clin Nutr Metab Care 1998; 1: 291–296.<br />

165. McKinnon BT. FDA safety alert: hazards of precipitation<br />

associated with parenteral nutrition. Nutr Clin Prac<br />

1996; 11: 59–65.<br />

166. Bethune K, Allwood M, Grainger C, Wormleighton C.<br />

Use of filters during the preparation and administration<br />

of parenteral nutrition: position paper and guidelines<br />

prepared by a British pharmaceutical nutrition group<br />

working party. Nutrition 2001; 17: 403–408.<br />

167. Trissel LA, Gilbert DL, Martinez JF et al. Compatibility<br />

of medications with 3-in-1 parenteral nutrition admixtures.<br />

J Parenter Enteral Nutr 1999; 23: 67–74.<br />

168. Havala T, Shronts E. Managing the complications associated<br />

with refeeding. Nutr Clin Prac 1990; 5: 23–29.<br />

169. SM, Kirby DF. The refeeding syndrome: a review. J<br />

Parenter Enter Nutr 1989; 14: 90–97.<br />

170. Crook MA, Hally V, Panteli J. The importance of the<br />

refeeding syndrome. Nutrition 2001; 17: 632–637.<br />

171. Alpers DH, Klein S. Refeeding the malnourished<br />

patient. Curr Opin Gastroenterol 1999; 15: 151–153.<br />

172. Colomb V, Fabeiro M, Dabbas M et al. Central venous<br />

catheter-related infections in children on long-term<br />

home parenteral nutrition: incidence and risk factors.<br />

Clin Nutr 2000; 19: 355–359.<br />

173. Rubin LG, Sanchez PJ, Siegel J et al. Pediatric<br />

Prevention Network. Evaluation and treatment of<br />

neonates with suspected late-onset sepsis: a survey of<br />

neonatologists’ practices. Pediatrics 2002; 110: 42.<br />

174. Benjamin DK Jr, Miller W, Garges H et al. Bacteremia,<br />

central catheters, and neonates: when to pull the line.<br />

Pediatrics 2001; 107: 1272–1276.<br />

175. Moukarzel AA, Haddad I, Ament ME et al. 230 patientyears<br />

of experience with home long-term parenteral<br />

nutrition in childhood: natural history and life of<br />

central venous catheters. J Pediatr Surg. 1994; 29:<br />

1323–1327.<br />

176. Goulet O, Larchet M, Gaillard JL et al. Catheter related<br />

sepsis during long-term parenteral nutrition in pediatric<br />

gastroenterology patients: a study of 185 consecutive<br />

central venous catheter. Clin Nutr 1990; 9: 73–78.<br />

177. Buchman AL, Moukarzel A. Metabolic bone disease<br />

associated with total parenteral nutrition. Clin Nutr<br />

2000; 19: 217–231.<br />

178. Hicks W, Hardy G. Phosphate supplementation for<br />

hypophosphataemia and parenteral nutrition. Curr Opin<br />

Clin Nutr Metabol Care 2001; 4: 227–233.<br />

179. Klein GL, Leichtner AM, Heyman MB. Aluminium in<br />

large and small volume parenterals used in total<br />

parenteral nutrition: response to the Food and Drug<br />

Administration notice of proposed rule by the North<br />

American Society for Pediatric Gastroenterology and<br />

Nutrition. J Pediatr Gastroenterol Nutr 1998; 27:<br />

457–460.<br />

180. Popinska K, Kierkus J, Lyszkowska M et al. Aluminium<br />

contamination of parenteral nutrition additives, amino<br />

acid solutions, and lipid emulsions. Nutrition 1999; 15:<br />

683–686.<br />

181. Dellert SF, Farrell MK, Specker BL, Heubi JE. Bone<br />

mineral content in children with short bowel syndrome<br />

after discontinuation of parenteral nutrition. J Pediatr<br />

1998; 132: 516–519.<br />

182. Quigley EMM, Marsh MN, Shaffer JL, Markin RS.<br />

Hepatobiliary complications of total parenteral nutrition.<br />

Gastroenterology 1993; 104: 286–301.


578<br />

Parenteral nutrition in infants and children<br />

183. Colomb V, Goulet O, Rambaud C et al. Long term<br />

parenteral nutrition in children: liver and gallbladder<br />

disease. Transplant Proc 1992; 24: 1054–1055.<br />

184. Fouin Fortunet H, Le Quernec L, Erlinger S et al.<br />

Hepatic alterations during total parenteral nutrition in<br />

patients with inflammatory bowel disease: a possible<br />

consequence of lithocholate toxicity. Gastroenterology<br />

1982; 82: 932–937.<br />

185. Beath SV, Davies P, Papadopoulou A et al. Parenteral<br />

nutrition-related cholestasis in postsurgical neonates:<br />

multivariate analysis of risk factors. J Pediatr Surg 1996;<br />

31: 604–606.<br />

186. Moseley RH. Sepsis-associated cholestasis.<br />

Gastroenterology 1997; 112: 302–305.<br />

187. Roelofsen H, Schoemaker B, Bakker C et al. Impaired<br />

hepatocanalicular organic anion transport in endotoxemic<br />

rats. Am J Physiol 1995; 269: G427–G434.<br />

188. Noel M, Chevenne D, Porquet D. Utility of insulin-like<br />

growth factor-I and its binding protein assays. Curr<br />

Opin Clin Nutr Metab Care 2001; 4: 399–405.<br />

189. Moss RL, Das JB, Ansari G, Raffensperger JG.<br />

Hepatobiliary dysfunction during total parenteral nutrition<br />

is caused by infusate, not the route of administration.<br />

J Pediatr Surg 1993; 28: 391–397.<br />

190. Belli DC, Fournier LA, Lepage G et al. Total parenteral<br />

nutrition-associated cholestasis in rats: comparison of<br />

different amino acid mixtures. JPEN 1987; 11: 67–73.<br />

191. Reif S, Tano M, Oliverio R. Total parenteral nutrition<br />

induced steatosis: reversal by parenteral lipid infusion.<br />

JPEN 1991; 15: 102–104.<br />

192. Wolf A, Pohlandt F. Bacterial infection: the main cause<br />

of acute cholestasis in newborn infants receiving shortterm<br />

parenteral nutrition. J Pediatr Gastroenterol Nutr<br />

1989; 8: 297–303.<br />

193. Moss RL, Das JB, Raffensperger JG. Total parenteral<br />

nurition-associated cholestasis: clinical and histopathological<br />

correlation. J Pediatr Surg 1993; 28: 1270–1275.<br />

194. Braxton C, Lowry SF. Editorial: parenteral nutrition and<br />

liver dysfunction-new insight? JPEN 1995; 19: 3–4.<br />

195. Kaufman SS, Loseke CA, Lupo JV et al. Influence of<br />

bacterial overgrowth and intestinal inflammation on<br />

duration of parenteral nutrition in children with short<br />

bowel syndrome. J Pediatr 1997; 131: 356–361.<br />

196. Sondheimer JM, Asturias E, Cadnapaphornchai M.<br />

Infection and cholestasis in neonates with intestinal<br />

resection and long-term parenteral nutrition. J Pediatr<br />

Gastroenterol Nutr 1998; 27: 131–137.<br />

197. Kaufman SS. Prevention of parenteral nutrition-associated<br />

liver disease in children. Pediatr Transplant 2002;<br />

6: 37–42.<br />

198. Ben Hariz M, Goulet O, De Potter S et al. Iron overload<br />

in children receiving prolonged parenteral nutrition. J<br />

Pediatr 1993; 123: 238–241.<br />

199. Moukarzel A, Song MK, Buchman AL et al. Excessive<br />

chromium intake in children receiving total parenteral<br />

nutrition. Lancet 1992; 339: 385–388.<br />

200. Ling PR, Sheikh M, Boyce P et al. Cholecystokinin<br />

(CCK) secretion in patients with severe short bowel<br />

syndrome (SSBS). Dig Dis Sci 2001; 46: 859–864.<br />

201. Capron JP, Gineston JL. Herve MA. Metronidazole in<br />

prevention of cholestasis associated with total<br />

parenteral nutrition. Lancet 1983; 1: 446–447.<br />

202. Spagnuolo MI, Iorio R, Vegnente A, Guarino A.<br />

Ursodeoxycholic acid for treatment of cholestasis in<br />

children on long-term total parenteral nutrition: a pilot<br />

study. Gastroenterology 1996; 111: 716–719.<br />

203. Heubi JE, Wiechmann DA, Creutzinger V et al.<br />

Tauroursodeoxycholic acid (TUDCA) in the prevention<br />

of total parenteral nutrition-associated liver disease. J<br />

Pediatr 2002; 141: 237–242.<br />

204. Salas JS, Dozio E, Goulet O et al. Energy expenditure<br />

and substrate utilization in the course of renutrition of<br />

malnourished children. J Parenter Enteral Nutr 1991; 15:<br />

288–293.<br />

205. Lienhardt A, Rakotoambinina B, Colomb V et al. Insulin<br />

secretion and sensitivity in children on cyclic total<br />

parenteral nutrition. JPEN J Parenter Enteral Nutr 1998;<br />

22: 382–386.<br />

206. Beghin L, Michaud L, Hankard R et al. Total energy<br />

expenditure and physical activity in children treated with<br />

home parenteral nutrition. Pediatr Res 2003; 53: 684–690.<br />

207. Shulman RJ, Phillips S. Parenteral nutrition in infants<br />

and children. J Pediatr Gastroenterol Nutr 2003; 36:<br />

587–607.<br />

208. Dahlstrom KA, Goulet O, Roberts RL et al. Lipid tolerance<br />

in children receiving long-term parenteral nutrition:<br />

a biochemical and immunologic study. J Pediatr<br />

1988; 113: 985–990.<br />

209. Forchielli ML, Gura KM, Sandler R, Lo C. Aminosyn PF<br />

or Trophamine: which provides more protection from<br />

cholestasis associated with total parenteral nutrition? J<br />

Pediatr Gastroenterol Nutr 1995: 21: 374–382.<br />

210. Ben Hariz M, Goulet O, Colomb V et al. Inappropriate<br />

iron intake in children on long-term parenteral nutrition:<br />

outcome after iron withdrawal. Clin Nutr 1997; 16:<br />

251–255.<br />

211. Advenier E, Landry C, Colomb V et al. Aluminum contamination<br />

of parenteral nutrition and aluminum<br />

loading in children on long-term parenteral nutrition. J<br />

Pediatr Gastroenterol Nutr 2003; 36: 448–453.


35<br />

Introduction<br />

Gastrointestinal problems of<br />

the newborn<br />

Moti M Chowdhury and Agostino Pierro<br />

The birth of a newborn in most cases marks a<br />

joyous occasion for a family. However, occasionally<br />

it is dampened by the tragedy of a congenital<br />

anomaly or illness within the first few weeks of<br />

life, a period that accounts for most childhood<br />

fatalities. In this chapter we endeavor to outline<br />

some of the acquired problems relating to the<br />

gastrointestinal (GI) tract. Whilst it is acknowledged<br />

that almost all acquired GI problems of<br />

childhood may present within the neonatal period,<br />

most are rare events in newborns (e.g. appendicitis,<br />

intussusception, inflammatory bowel disease),<br />

and it would be beyond the scope of this chapter<br />

to discuss all such problems exhaustively. In this<br />

chapter we focus instead on the four common GI<br />

problems encountered in the first few weeks<br />

of life: necrotizing enterocolitis (NEC), gastroesophageal<br />

reflux (GER), infantile hypertrophic<br />

<strong>pylori</strong>c stenosis (IHPS) and inguinal hernia.<br />

Necrotizing enterocolitis<br />

NEC is a disease predominantly of premature<br />

infants characterized by a variable degree of<br />

intestinal necrosis, which may or may not be<br />

complicated by perforation and fulminant sepsis.<br />

Epidemiology<br />

NEC is the most common surgical emergency in<br />

the neonatal period, 1 affecting 0.5% of all live<br />

births and 3–5% of low-birth-weight live births.<br />

Although primarily a disease of the preterm infant,<br />

6–13% are in full-term infants. 2–4 There is a male<br />

preponderance of 2:1. 3<br />

Etiology<br />

Whilst the precise mechanism of NEC remains<br />

unclear, a number of predisposing risk factors<br />

have been identified: low birth weight, prematurity,<br />

enteral feeds and sepsis.<br />

Low birth weight<br />

Low birth weight (


580<br />

Gastrointestinal problems of the newborn<br />

hyperosmolar feeds has the potential to contribute<br />

large quantities of hydrogen gas to the pneumatosis<br />

intestinalis.<br />

Sepsis<br />

Although a number of organisms (see Pathophysiology<br />

below) have been cultured in pathological<br />

specimens of NEC, many are from the bowel<br />

flora, suggesting a mechanism involving an imbalance<br />

between the host and the organism rather<br />

than direct virulence of the organism. 14,19<br />

In addition to these recognized risk factors, a<br />

number of other weak associations have been<br />

reported with the development of NEC (Table<br />

35.1).<br />

Pathophysiology<br />

The precise pathophysiology of NEC remains<br />

unclear, but it is considered to be multifactorial in<br />

origin – the culmination of microbial colonization<br />

and inflammatory insult with mucosal injury to an<br />

intestine that is compromised by hypoxic ischemia<br />

and/or immaturity. 1,20 Hypoxia and circulatory<br />

ischemia result in preferential distribution of the<br />

limited blood supply to vital organs such as the<br />

heart, brain and kidney, at the expense of compromising<br />

blood flow away from non-vital regions<br />

including the splanchnic circulation of the intesti-<br />

Table 35.1 Risk factors for development of<br />

necrotizing enterocolitis<br />

Low birth weight<br />

Prematurity<br />

Enteral feeds<br />

Sepsis<br />

Intrauterine growth restriction 11,188,189<br />

Fetal distress 190<br />

Exposure to antenatal or postnatal glucocorticoids 7<br />

Premature rupture of membranes 191,192<br />

Birth asphyxia 14<br />

Respiratory disease, 191 e.g. hyaline membrane disease<br />

Congenital heart disease, e.g. coarctation, patent<br />

ductus arteriosis 193<br />

Maternal cocaine abuse 194,195<br />

Umbilical artery catheterization 196<br />

nal tract (the Herring–Breur reflex). Intestinal<br />

ischemia and bacterial colonization stimulate proinflammatory<br />

mediators (including interferonγ,<br />

21,22 tumor necrosis factor (TNF)-α 23 and platelet<br />

activating factor (PAF) 24,25 ), which through multiple<br />

cascades may lead to variable degrees of coagulative<br />

and ischemic necrosis of the bowel. 20<br />

Furthermore free radicals released from inflammatory<br />

cascades following ischemia and reperfusion<br />

contribute to the tissue necrosis by causing lipid<br />

peroxidative damage to cellular membranes. 26<br />

Diffusion of free radicals and the effect of gasforming<br />

organisms within the gut wall result in the<br />

pathognomonic pneumatosis intestinalis that is<br />

seen on radiographs in 90% of cases. 27<br />

Transmural necrosis with bacterial translocation<br />

across the gut wall may result in perforation with<br />

peritonitis, bacteremia or septicemia. Up to 35% of<br />

patients with NEC have positive cultures, 28 with<br />

the most common implicated pathogens being<br />

Klebsiella, 29 Escherichia coli, 29 Clostridium difficile<br />

30,31 and Streptococcus faecalis. 32<br />

The increased propensity of premature infants to<br />

develop NEC injury is attributed to a number of<br />

additional factors. These include a deficient<br />

intestinal microbial host defense, poor development<br />

of the mucosal barrier, 33,34 an immature<br />

mucosa at greater susceptibility to hyperosmolar<br />

feeds 18 and an accentuated pro-inflammatory<br />

response. 35,36 If the intestinal injury is short-lived,<br />

the pathological course may be followed by<br />

healing with fibrosis, with or without a resulting<br />

stricture in the affected bowel. The disease most<br />

commonly involves the terminal ileum and<br />

ascending colon, but it may affect variable<br />

segments of the small and large intestine, being<br />

isolated (30%), multifocal (55%) or pan-intestinal<br />

(15%) (Figure 35.1). 37<br />

Clinical signs and symptoms<br />

The clinical course of NEC is variable, with twothirds<br />

of patients developing the condition within<br />

the first week of life. 38 The severity of the disease<br />

may be categorized clinically using the original<br />

Bell’s staging system for NEC 39 outlined below, a<br />

classification later modified by Walsh and Kliegman.<br />

40


(a) (b) (c)<br />

Complications<br />

Necrotizing enterocolitis 581<br />

Figure 35.1 Intraoperative illustration of (a) isolated necrotizing enterocolitis (NEC), (b) multifocal NEC, (c) pan-intestinal NEC.<br />

Stage I<br />

Suspected NEC. At the early stage, symptoms and<br />

signs are usually non-specific, e.g. irritability,<br />

fever, apneas and bradycardias, and poor toleration<br />

of feeds. Many of these children are more<br />

likely to be suffering from feeding intolerance<br />

commonly associated with prematurity or low<br />

birth weight, but nonetheless they should be<br />

regarded as at risk of developing more fulminant<br />

disease.<br />

Stage II<br />

Established NEC. These babies will have established<br />

clinical and radiographic signs of NEC with<br />

moderate–profound systemic illness, including<br />

metabolic acidosis and thrombocytopenia.<br />

Vomiting/aspirates are usually bilious or bloodstained<br />

whilst stools are likely to be blood-stained.<br />

Abdominal distension, abdominal wall erythema<br />

and edema are usual, and there may or may not be<br />

evidence of perforation clinically and/or radiologically.<br />

A clinically palpable intra-abdominal mass<br />

(reported in 11% 38 ) may indicate the presence of<br />

persistently dilated bowel loops or, if the disease is<br />

localized, an intraperitoneal abscess.<br />

Stage III<br />

Advanced NEC with systemic instability. As well as<br />

features of stage II disease, these babies have<br />

cardiovascular instability (hypotension), severe<br />

metabolic acidosis and coagulative derangements.<br />

There will usually be abdominal and radiological<br />

evidence of perforation, peritonitis and ascites<br />

with generalized sepsis.<br />

At least 10% 41 of NEC cases develop one or more<br />

of the following complications, with their incidences<br />

being significantly more common in<br />

preterm infants. 37<br />

(1) Metabolic disorders including metabolic<br />

acidosis and disseminated intravascular coagulation<br />

(DIC);<br />

(2) Perforation with peritonitis and septicemia;<br />

(3) Localized intraperitoneal abscess formation;<br />

(4) Healing by fibrosis with stricture formation<br />

(20% in term infants and 31% in preterm<br />

infants 37 );<br />

(5) Massive gut involvement that requires resection<br />

may result in short-bowel syndrome, 42,43<br />

with resultant failure to thrive or, worse still,<br />

pan-intestinal involvement that is incompatible<br />

with life;<br />

(6) Recurrence of the disease (9–14% 37 );<br />

(7) Infants with stage II or III disease are<br />

commonly at risk of developing psychomotor<br />

retardation, 44–49 although this may be more<br />

attributed to the high incidence of prematurity<br />

than a direct effect of NEC.<br />

Diagnosis with differential<br />

The differential diagnoses of NEC are outlined in<br />

Table 35.2. Investigations for diagnosis of NEC<br />

include the following.


582<br />

Gastrointestinal problems of the newborn<br />

Table 35.2 Differential diagnosis of<br />

necrotizing enterocolitis<br />

Feeding intolerance of prematurity<br />

Spontaneous intestinal perforation<br />

Malrotation/volvulus<br />

<strong>Neonatal</strong> sepsis with ileus<br />

Congenital intestinal obstruction (intestinal atresia,<br />

Hirschsprung’s disease)<br />

<strong>Neonatal</strong> pseudomembranous colitis (rare)<br />

<strong>Neonatal</strong> appendicitis (rare)<br />

Abdominal X-ray (Figure 35.2) may demonstrate<br />

dilated bowel loops, edematous bowel wall,<br />

ascites, intramural gas (pneumatosis intestinalis),<br />

portal vein gas and free air in the peritoneal cavity<br />

(pneumoperitoneum). Signs of a pneumoperitoneum<br />

include the ‘football sign’ (central collection<br />

of free air outlining the falciform ligament and<br />

umbilical arteries), localized gas in the right upper<br />

quadrant (the ‘right upper quadrant gas sign’), or<br />

gas on both sides of the bowel wall (‘Rigler’s sign’).<br />

The presence of a single/multiple loops of bowel<br />

retaining the same shape or position for 24–36h<br />

(‘fixed-loop sign’) may suggest full-thickness bowel<br />

necrosis.<br />

In metabolic acidosis, the hemoglobin level may<br />

fall, owing to hemorrhage and sepsis, whilst leukocyte<br />

and platelet counts 37,50 may be depressed,<br />

particularly in association with DIC. 37<br />

Bacteriological screening should include blood<br />

culture and collection of swabs from the nose,<br />

throat, umbilicus and rectum.<br />

Treatment options<br />

The initial management of NEC consists of stabilization<br />

of the infant and resting of the intestinal<br />

tract by means of discontinuation of enteral feeds<br />

for 10 days, total parenteral nutrition, decompression<br />

of the stomach by a nasogastric tube, fluid<br />

resuscitation, intravenous antibiotics for 7–14<br />

days and correction of any metabolic derangement.<br />

However, up to 50% of neonates with NEC<br />

Figure 35.2 Abdominal X-ray demonstrating<br />

pneumoperitoneum from perforated necrotizing<br />

enterocolitis.<br />

develop advanced disease that requires surgery. 51<br />

Indications for surgery include:<br />

(1) Clinical deterioration despite maximal<br />

medical treatment;<br />

(2) A pneumoperitoneum, indicating perforated<br />

NEC;<br />

(3) An abdominal mass with persistent intestinal<br />

obstruction or sepsis;<br />

(4) The development of an intestinal stricture.<br />

The options for surgery are between primary peritoneal<br />

drainage or laparotomy. However, the optimum<br />

choice between primary peritoneal drainage<br />

and laparotomy remains controversial,<br />

particularly in low-birth-weight infants (1000g who have<br />

no associated morbidities and are clinically stable,<br />

are preferentially treated by primary laparotomy. 52<br />

The principal surgical objectives of laparotomy in<br />

acute NEC are to control sepsis, to remove<br />

gangrenous bowel and to preserve as much bowel<br />

length as possible. 53–55 Within these objectives, a


Figure 35.3 An infant with a peritoneal drain (arrow)<br />

inserted into the right iliac fossa.<br />

number of options exist, including: resection with<br />

enterostomy; resection with primary anastomosis;<br />

proximal jejunostomy; the ‘clip and drop’ technique;<br />

or to ‘patch, drain and wait’. The option<br />

exercised is highly variable between surgeons<br />

9,56–59 and is often influenced by the site and<br />

extent of the disease. An algorithm of the authors’<br />

preferred surgical approach for the management of<br />

advanced NEC is illustrated in Figure 35.4.<br />

Prognosis<br />

The outcome from NEC is highly variable, with the<br />

postoperative complication and mortality rates<br />

ranging between 10 and 70%. These depend on:<br />

Stable<br />

Focal<br />

Resection and<br />

anastomosis<br />

Unstable<br />

Viable<br />

distal bowel<br />

Stoma Resection and Stoma ±<br />

anastomosis resection<br />

NEC findings at laparotomy<br />

Proximal 'Clip and<br />

enterostomy drop'<br />

(1) Stage of the disease; 14,49,60,61<br />

Gastroesophageal reflux 583<br />

(2) Extent of disease involvement, with<br />

the lowest survival in pan-intestinal disease;<br />

5,8,11,37,42,49,62–65<br />

(3) Birth weight; 5–10,14,49<br />

(4) Gestational age, 5,14,37,49,63,66,67 with 77% survival<br />

in term infants compared to 66% in<br />

preterm infants;<br />

(5) Presence of associated medical co-morbidities;<br />

7,49,68<br />

(6) Modality of operative treatment. This affects<br />

the postoperative complication rate for<br />

multifocal NEC (85% survival after resection<br />

and primary anastomosis compared to 50%<br />

with enterostomy alone) but not isolated<br />

NEC. 37<br />

Contrary to popular belief, the loss of the ileocecal<br />

valve is not associated with increased complication<br />

or mortality rates, 37,69,70 suggesting that these<br />

neonates adapt rapidly to the loss of the ileocecal<br />

valve.<br />

Gastroesophageal reflux<br />

GER is a physiological process characterized by<br />

the involuntary passage of gastric contents into the<br />

lower esophagus not induced by noxious stimuli.<br />

The phenomenon is considered as GER disease<br />

only when it causes the patient to be symptomatic<br />

or results in pathological complications.<br />

Multifocal Pan-intestinal Total intestinal<br />

gangrene<br />

Questionable distal bowel or<br />

bleeding at bowel dissection or<br />

unstable<br />

Proximal<br />

jejunostomy<br />

'Clip and<br />

drop'<br />

Consider withdrawal<br />

of teatment<br />

Figure 35.4 Algorithm of the authors’ preferred operative management of advanced necrotizing enterocolitis (NEC)<br />

(reproduced with permission from Seminars in Perinatology 2003).


584<br />

Gastrointestinal problems of the newborn<br />

Epidemiology<br />

Most newborns have some degree of GER, but the<br />

majority resolve spontaneously, with the prevalence<br />

decreasing to 18% in childhood. 71 The<br />

male/female ratio is 1.6. 72 The incidence of GER is<br />

highest in neurologically impaired children (70%),<br />

who comprise 44–67% of children undergoing<br />

anti-reflux surgery. 73–75<br />

Pathophysiology<br />

A number of physiological and anatomical factors<br />

normally contribute to prevent chronic reflux of<br />

gastric contents into the lower esophagus. The<br />

combination of esophageal motility and gravity<br />

facilitates esophageal clearance of refluxed material<br />

as well as of saliva, which is rich in bicarbonate,<br />

that coats the esophagus. These esophageal<br />

clearance mechanisms are usually developed by<br />

31 weeks’ gestation. 76 Other physiological barriers<br />

to GER include antral contractions facilitating<br />

gastric emptying, and the production of mucus,<br />

prostaglandin and epithelial growth factors, which<br />

help to prevent damage to the esophageal mucosa.<br />

Anatomically the length of the intra-abdominal<br />

esophagus, the phrenoesophageal ligaments, the<br />

gastric mucosal ‘rosette’ and the esophageal hiatus<br />

(which is a sling formed by the crura of the<br />

diaphragm causing a pinchcock effect) all<br />

contribute to a higher-pressure zone in the lower<br />

esophagus. This high-pressure zone forms the<br />

lower esophageal sphincter (LES), a physiological<br />

rather than a true anatomical sphincter. Pressures<br />

at this gastroesophageal junction (10–30mmHg)<br />

are greater than gastric luminal pressure<br />

(5mmHg), thereby preventing retrograde passage<br />

of gastric contents. In addition, the acute angle of<br />

His (made by the esophagus and the axis of the<br />

stomach) and the above physiological factors<br />

cumulatively contribute to limit the volume and<br />

frequency of gastric contents refluxing into the<br />

lower esophagus. Much of these anatomical<br />

features, however, are poorly developed in the first<br />

weeks of an infant’s life, predisposing it to a higher<br />

risk of GER within this period. For instance, the<br />

angle of His is obtuse in newborns and decreases<br />

only as the infant grows; also, the length of the<br />

intra-abdominal esophagus is shorter, only 1cm at<br />

birth, compared to 3cm by 3months of age. Other<br />

abnormalities that predispose to GER include<br />

disruption of the gastroesophageal junction (with<br />

resulting hiatus hernia), weakness or incompetence<br />

of the LES and poor clearance of acid from<br />

the esophagus. 77<br />

Etiology<br />

A mean intra-abdominal pressure of less than<br />

10mmHg is necessary for the LES to remain<br />

competent. GER is made more likely in groups<br />

with raised intra-abdominal pressure for example<br />

following repair of omphalocele (43% 78 ), congenital<br />

diaphragmatic hernia 79 and chronic respiratory<br />

infections.<br />

Neurologically impaired children have the highest<br />

incidence of GER (65–70% 80 ). This is due to a<br />

combination of poor esophageal and gastric motility<br />

(due to vagal nerve dysfunction), chronic<br />

supine positioning, abdominal spasticity,<br />

diaphragmatic flaccidity, scoliosis, retching and<br />

increased use of gastrostomy for feeding.<br />

Insertion of gastrostomy tubes has been reported to<br />

be associated with the development or worsening<br />

of pre-existing GER. The gastrostomy, which fixes<br />

the stomach to the anterior abdominal wall, potentially<br />

opens the angle of His 81 and lowers the LES<br />

pressure 82 thereby predisposing to GER.<br />

GER occurs in 30–80% of children treated for<br />

esophageal atresia, the incidence being related to<br />

the length of the atresia gap. The GER is attributed<br />

partly to poor esophageal motility in these patients<br />

and partly to a shortened esophagus. The shortened<br />

esophagus, from the original anomaly and<br />

compounded by the surgical repair, results in<br />

upward displacement of the gastroesophageal<br />

junction.<br />

Clinical signs and symptoms<br />

The infant with GER typically presents with<br />

vomiting or poor toleration of feeds, made worse at<br />

night during the supine position. Those with associated<br />

esophagitis may manifest clinically with<br />

Sandifer’s syndrome, a voluntary dystonic contortion<br />

of the head, neck and trunk. These movements<br />

have been shown to improve peristalsis in<br />

the lower esophagus. If GER is left untreated,


failure to thrive from calorie deprivation may<br />

ensue. Reflux of gastric contents into the airways<br />

may result in coughing and choking, and chronic<br />

aspiration may cause the infant to present with<br />

complications of GER, including laryngospasm<br />

with apneic and bradycardia spells (particularly<br />

during sleep), stridor or pneumonia.<br />

Complications<br />

(1) Failure to thrive secondary to calorie deprivation<br />

occurs in up to 20–52% of children. 83,84<br />

(2) Iron deficiency anemia, seen in 31%, 83 may<br />

indicate significant reflux esophagitis with<br />

blood loss.<br />

(3) Aspiration of gastric contents may cause<br />

laryngospasm and subsequently obstructive<br />

apneas and bradycardias (17% 84 ), particularly<br />

during sleep, or be complicated by pneumonia<br />

(in 31% 83 ) or subglottic stenosis.<br />

(4) Bronchopulmonary dysplasia (60% 84 ), bronchiectasis,<br />

bronchitis and asthma. The mechanism<br />

of these complications is not clear. One<br />

hypothesis is that reflux of gastric contents<br />

may stimulate vagal afferents in the distal<br />

esophagus which may cause a reflex vagovagal<br />

bronchoconstriction. 85,86<br />

(5) Persistent GER may result in esophagitis,<br />

which in turn may lead to sticture with<br />

dysphagia or, in older children, Barrett’s<br />

esophagus, a recognized pre-malignant condition.<br />

Diagnosis with differential<br />

The differential diagnoses of an infant with clinical<br />

features of GER are outlined in Table 35.3.<br />

Investigations used for diagnosis of GER include<br />

the following.<br />

Twenty-four-hour pH monitoring is currently the<br />

most sensitive and specific test available for diagnosing<br />

GER. Monitoring is performed for 24<br />

continuous hours, during which time the patient is<br />

fed only breast milk, formula or apple juice. The<br />

juice is preferable, as the alkaline content of milk<br />

feeds may neutralize the gastric acid reflux and<br />

thereby potentially produce a false-negative result.<br />

Gastroesophageal reflux 585<br />

Table 35.3 Differential diagnosis of<br />

gastroesophageal reflux<br />

Infantile hypertrophic <strong>pylori</strong>c stenosis<br />

Overfeeding/feeding disorders<br />

Malrotation<br />

Gastroenteritis<br />

<strong>Neonatal</strong> sepsis<br />

Neurological pathology (e.g. raised intracranial<br />

pressure from hydrocephalus)<br />

Laryngeal cleft<br />

Acid reflux is defined by pH 50% of the radioisotope in<br />

the stomach after 90min, in the absence of<br />

mechanical obstruction, indicates delayed gastric<br />

emptying.<br />

Esophagoscopy allows visualization of the gastroesophageal<br />

mucosa. However, only 40% of cases of<br />

GER will demonstrate unequivocal esophagitis.<br />

Endoscopy is therefore a poor tool for diagnosis of<br />

GER, and is more useful in the assessment of<br />

complications of reflux (e.g. esophagitis or stricture)<br />

and in obtaining biopsies (e.g. <strong>Helicobacter</strong><br />

<strong>pylori</strong> infections or development of Barrett’s<br />

esophagus).<br />

Esophageal manometry enables measurement of<br />

the LES tone. However, the LES tone is low at<br />

birth, rising to a mean pressure of 10–15mmHg by


586<br />

Gastrointestinal problems of the newborn<br />

Figure 35.5 Upper gastrointestinal contrast study<br />

demonstrating gastroesophageal reflux.<br />

6 months of age. Thus, sphincter tone will be<br />

physiologically low in neonates, and therefore has<br />

little application in diagnosing GER in this age<br />

group.<br />

Chest X-ray may identify pulmonary complications<br />

of GER.<br />

Lipid-laden alveolar macrophages in tracheal aspirates/bronchoalveolar<br />

lavage may indicate aspiration<br />

secondary to GER. However, the sensitivity<br />

and specificity for detecting GER are as low as 38%<br />

and 59%, respectively. 87 Furthermore, elevated<br />

levels of lipid-laden macrophages are found in a<br />

number of pulmonary disease without any<br />

evidence of aspiration. 88<br />

Treatment options<br />

The two main aims of treatment are to prevent the<br />

respiratory complications of GER and to improve<br />

the nutritional status of the child from resumption<br />

of normal feeding.<br />

Medical therapy<br />

Conservative measures for GER in infants are<br />

frequently advocated. They include the avoidance<br />

of medications that reduce LES tone (caffeine,<br />

theophylline, anticholinergics), dietary modifications<br />

(changing the feed pattern with use of<br />

frequent, small-volume feeds) and positioning<br />

maneuvers. However, many of these have no<br />

confirmed efficacy. Thickening of formula feeds<br />

(e.g. with carob bean gum, or rice flour) may<br />

reduce frank emesis but does not reduce GER<br />

measurably compared to placebo. 89–92 Furthermore,<br />

there are no quality data to support the<br />

opinion that more frequent but smaller-volume<br />

feedings reduce GER. 93 With respect to positioning<br />

maneuvers, positioning at a 60º head elevation<br />

increases GER compared to the prone position. 94<br />

However, the association of the prone position<br />

with sudden infant death syndrome has brought<br />

controversy with this maneuver. No significant<br />

difference has been found between the flat and<br />

head-elevation prone positions. 95<br />

Pharmacotherapy forms the main first-line treatment<br />

modality of GER. A wide spectrum of agents<br />

is now available (Table 35.4), aimed at decreasing<br />

acid secretion and increasing gastric emptying.<br />

Detailed descriptions of the pharmacological<br />

mechanisms of these agents can be found elsewhere.<br />

Surgical treatment<br />

Whilst medical therapy serves to decrease the acid<br />

content of the refluxate, other components of the<br />

refluxate (e.g. bile pepsin, trypsin) remain unaffected.<br />

Surgery therefore becomes indicated in<br />

infants who do not respond to medical treatment,<br />

or who experience apparent life-threatening events<br />

(e.g. laryngospasm and apneas), or have anatomical<br />

problems such as hiatus hernias or esophageal<br />

strictures. Operative options available for treatment<br />

of GER are Nissen’s fundoplication (used in<br />

64% 74 ) and Thal (34% 74 ), Toupet (1.5% 74 ), Dor,<br />

Boerema, Boix–Ochoa, Collis–Belsey and Hill<br />

procedures. Nissen’s fundoplication (Figure 35.6) 96<br />

is the most popular method, and involves a 360º<br />

wrap of the fundus of the stomach, brought round<br />

the posterior aspect of the esophagus. It controls<br />

GER by increasing the acuteness of the angle of His<br />

and lengthening the intra-abdominal esophagus,


Table 35.4 Pharmacotherapy of gastroesophageal reflux (GER)<br />

Antacids, e.g. Gaviscon ®<br />

thereby increasing the high-pressure zone at the<br />

LES. A concurrent gastrostomy is frequently<br />

placed, particularly in neurologically impaired<br />

children, those with feeding difficulties and poor<br />

weight gain, or slow gastric emptying. The Thal<br />

procedure 97 is an anterior fundoplication with a<br />

partial (180–270º) wrap of the fundus of the<br />

stomach to the intra-abdominal esophagus. The<br />

benefit of the Thal procedure is that it allows the<br />

patient to belch and release any bloating, which is<br />

not feasible with Nissen’s fundoplication.<br />

Fundoplication procedures may be performed by<br />

the open or laparoscopic method. Reliable data<br />

comparing outcomes between the two approaches<br />

are, however, limited in children. Non-randomized<br />

studies suggest that the laparoscopic approach<br />

offers better cosmesis and, if not favorable, is at<br />

Gastroesophageal reflux 587<br />

H2 blockers, e.g. ranitidine<br />

Proton pump inhibitors, e.g. omeprazole<br />

Prokinetic agents, e.g. dopamine antagonists (e.g. metoclopramide), postganglionic 5-HT4 agonist/acetylcholine<br />

agonists (e.g. cisapride), erythromycin, bethanecol or domperidone. Prokinetics increase LES tone and esophageal<br />

peristalsis thereby improving gastric emptying. However, concerns regarding the safety of cisapride on the<br />

myocardium, with reports of arrhythmias and prolonged QT interval, has restricted its clinical use<br />

γ-Aminobutyric acid (GABA) agonists, e.g. baclofen, are agents that inhibit GER episodes by inhibition of transient<br />

relaxations of the LES, 197,198 and are currently being considered for clinical trials<br />

LES, lower esophageal sphincter<br />

Figure 35.6 Nissen’s fundoplication 1, Mobilization of the fundus of the stomach by division of short gastric vessels. 2,<br />

Fundus folded round the posterior aspect of the esophagus. 3, A loose wrap formed, using interrupted sutures. 4, The<br />

completed stomach wrap. (Adapted with permission from reference 187).<br />

least comparable with respect to operative time,<br />

postoperative pain, time to feeds and complication<br />

rate. 98–101<br />

Recently, two further endoscopic techniques have<br />

been described for the treatment of GER. The first<br />

is endoscopic radiofrequency energy delivery to<br />

the gastroesophageal junction around the LES.<br />

Early data suggest that this can achieve resolution<br />

of symptoms of GER in 87% of patients, 102 but is<br />

associated with significant mortality and morbidity,<br />

including aspiration, pleural effusion and<br />

atrial fibrillation. 103 The second is endoscopic<br />

gastroplication, 104 which involves internally plicating<br />

the stomach on itself approximately 1cm<br />

below the gastroesophageal junction to alter the<br />

angle of His and thereby decrease reflux of gastric


588<br />

Gastrointestinal problems of the newborn<br />

contents. Early results indicate poorer outcomes<br />

compared to fundoplications, with 75% still<br />

requiring adjuvant medical treatment and 6%<br />

undergoing fundoplication due to therapeutic<br />

failure. 105 Furthermore, the data reported with<br />

both these procedures have so far been limited to<br />

adult populations, and their application in pediatrics<br />

remains unclear.<br />

Prognosis<br />

The prognosis of infants with GER is highly variable,<br />

depending on the presence of associated<br />

anomalies (e.g. esophageal atresia, congenital<br />

diaphragmatic hernia, neurological disorders),<br />

anatomical problems (e.g. hiatus hernia), prematurity<br />

of the infant, or the presence of established<br />

complications of GER. Approximately 80% of<br />

patients with symptomatic GER are treated<br />

successfully without surgery. Infants undergoing<br />

anti-reflux surgery demonstrate clinical improvement<br />

in 90%, if GER is isolated, compared to 64%<br />

of infants with associated anomalies. 106 Fundoplication<br />

is more effective in improving emesis<br />

(76%) and respiratory symptoms (66%) than affecting<br />

the nutritional status, with persistence of<br />

failure to thrive in 62% of patients.<br />

Complications following fundoplication repairs<br />

(Table 35.5) occur in up to 24%, with the neurologically<br />

impaired having up to six-fold increased<br />

incidence compared to those neurologically<br />

normal. 73–75,106–113 A repeat fundoplication is<br />

required in up to 24% of cases, due to either failure<br />

of the wrap (disruption, herniation or excessive<br />

tightness) or recurrence of GER. Recurrence of GER<br />

symptoms after fundoplication is more frequent in<br />

infants, particularly those with previous esophageal<br />

atresia repair (42%), with half of these requiring<br />

a second procedure. 106<br />

Infantile hypertrophic <strong>pylori</strong>c stenosis<br />

IHPS is the second most common condition in<br />

infancy that requires surgery. The treatment of<br />

pyloromyotomy was first described by Fredet in<br />

1907, and then my Ramstedt 4 years later. 114<br />

Table 35.5 Complications following<br />

fundoplication<br />

Disruption of the wrap with recurrence of GER<br />

(8–12%)<br />

Dysphagia from an excessively tight wrap (2–12%)<br />

Herniation of the wrap (2–10%)<br />

Gas bloating (4–10%) particularly with Nissen’s<br />

fundoplications<br />

Adhesive intestinal obstruction (2–10%)<br />

Poor esophageal clearance (in patients with<br />

esophageal atresia)<br />

Splenic injury (0.5%)<br />

Mortality (0–13%)<br />

GER, gastroesophageal reflux<br />

Epidemiology<br />

IHPS affects 1 in 500 Caucasian children and fewer<br />

than 1 in 1000 Asian and African infants. 115–118<br />

There is a male preponderance – a gender discrepancy<br />

that has been widening recently, reported<br />

between 4:1 and 10:1. 119–125 Firstborn males are<br />

most often affected, accounting for 40–60% of all<br />

cases.<br />

Etiology<br />

In spite of IHPS being one of the most common and<br />

earliest recognized surgical problems in infants, its<br />

precise etiology remains unclear. Two proposed<br />

risk factors are breast feeding and a positive family<br />

history. Twenty per cent of infants with IHPS have<br />

a positive family history. Although more common<br />

in males, a positive family history is four times as<br />

common from affected mothers than fathers. 126<br />

Overall, there is a 15–20-fold increase in risk of a<br />

child of an affected parent developing <strong>pylori</strong>c<br />

stenosis compared to the general population. 126,127<br />

It is also more common in multiple births. 128,129<br />

The incidence of Smith–Lemli–Opitz syndrome<br />

diagnosed in infants with IHPS is 150-fold higher<br />

than in the general population. 117 This, and other<br />

congenital gastrointestinal and urinary tract<br />

malformations that are more common with<br />

IHPS, 118 suggest a genetic role in its etiology. The


mode of inheritance is likely to be multifactorial,<br />

although evidence of reduced expression of the<br />

neuronal nitric oxide synthase (nNOS) gene, which<br />

is responsible for the smooth-muscle relaxant nitric<br />

oxide (NO), has been found. 130,131 The influence of<br />

breast feeding on IHPS is more controversial. A<br />

number of groups have found that infants with<br />

IHPS were twice as likely to have been breast<br />

fed. 116,132,133 However, Hitchcock et al 134 found no<br />

such link whilst Pisacane et al 135 found a protective<br />

effect from breast feeding.<br />

Pathophysiology<br />

Whilst the precise mechanism of IHPS remains<br />

unclear, a number of causative factors have been<br />

reported.<br />

First, is failure of relaxation of the pylorus. The<br />

circular layer of smooth muscle of the pylorus is<br />

stimulated by the myenteric plexus, whilst its<br />

relaxation is dependent on non-adrenergic, noncholinergic<br />

(NANC) inhibitory motor neurons.<br />

Depletion of these nerves (either primary absence<br />

or degeneration) in <strong>pylori</strong>c muscle may be the<br />

cause of the excessively contracted hypertrophic<br />

circular <strong>pylori</strong>c muscle. 136 This hypothesis is<br />

further supported by the demonstration of reduced<br />

expression of the nNOS gene (responsible for NO<br />

synthesis, a mediator of NANC inhibitory nerves)<br />

in animals models 130 and depleted nNOS mRNA in<br />

the <strong>pylori</strong>c muscle of patients with IHPS. 131 The<br />

influence of uncompensated pylorus contractility<br />

may also explain why neonates receiving<br />

erythromycin, which induces the activity of migrating<br />

motor complexes in the stomach 137 and<br />

increases <strong>pylori</strong>c contraction, 138 have up to a<br />

seven-fold increased risk 139,140 of developing IHPS.<br />

It is suggested that this marked increase in motility<br />

may lead to hypertrophy of the pylorus. 139<br />

Increased expression of insulin-like growth factor-<br />

Ι 141,142 and transforming growth factor-β 143 found<br />

in hypertrophic <strong>pylori</strong>c muscle, may indicate a<br />

hormonal role in smooth-muscle hypertrophy in<br />

IHPS.<br />

Increased amounts of extracellular matrix proteins,<br />

particularly collagen, have been reported within<br />

the circular muscle wall of the pylorus in IHPS.<br />

This may account for the hypertrophic wall of the<br />

pylorus. 144,145<br />

Infantile hypertrophic <strong>pylori</strong>c stenosis 589<br />

Hypertrophy of the circular smooth muscle wall of<br />

the pylorus up to four-fold results in <strong>pylori</strong>c<br />

luminal outflow obstruction. The infant vomits<br />

obstructed milk feeds and with it gastric acid<br />

contents (hydrochloric acid), resulting in dehydration<br />

with a hypochloremic, hypokalemic metabolic<br />

alkalosis.<br />

Clinical signs and symptoms<br />

The infant with IHPS typically presents with<br />

vomiting, usually at 3–6 weeks of age, although up<br />

to one-third start vomiting within the first week of<br />

life. 121,146 Vomiting may initially be effortless, but<br />

with time it becomes forceful, described as ‘projectile’.<br />

The vomitus is usually milk feeds, practically<br />

never bilious, and if severe may contain some<br />

‘coffee-ground’ flecks of altered blood from<br />

secondary gastritis in 15–20%. 133 The infant is<br />

frequently hungry for further feeds, despite the<br />

vomiting. Constipation is common. Jaundice may<br />

occur in 3%, and is attributed to the adverse effect<br />

of starvation on hepatic glucuronyl transferase<br />

activity. 133 Clinically, dehydration from severe<br />

vomiting, loss of weight and failure to thrive may<br />

ensue. Abdominal examination may reveal visible<br />

peristalsis of the obstructed pylorus (from left to<br />

right) in 75–95% of cases, and a <strong>pylori</strong>c ‘tumor’<br />

mass, felt like an ‘olive’, representing the hypertrophic<br />

pylorus, usually in the epigastrium or<br />

upper quadrants of the abdomen. Successful palpation<br />

of the <strong>pylori</strong>c tumor can be achieved in up to<br />

90% of cases, 147 and may be aided by maneuvers<br />

such as nasogastric decompression of the stomach<br />

(which is usually distended with gas swallowed by<br />

a hungry crying child), palpation through the<br />

midline gap between the two rectus muscles or at<br />

the lateral margins of the recti, flexing the hips, or<br />

allowing the child to suck on a passifier or a ‘test<br />

feed’ of dextrose solution (which should be aspirated<br />

back from the nasogastric tube).<br />

Complications<br />

(1) Dehydration and metabolic imbalance;<br />

(2) Intraoperative duodenal perforation. This may<br />

occur in 3–4%; 148,149<br />

(3) Postoperative complications. These include<br />

transient persistence of vomiting in 3–19% 122,148


590<br />

Gastrointestinal problems of the newborn<br />

(particularly those with a protracted preoperative<br />

history of vomiting); wound infection<br />

(1–4%); 148 or wound dehiscence (1.4%). 149<br />

With experience and earlier diagnosis, mortality<br />

fell from 59% in 1925 to almost nil by<br />

1975. 121,148,150<br />

Diagnosis with differential<br />

The differential diagnosis of a vomiting child in<br />

this age group is usually an overfed child or GER.<br />

Less commonly, the following should be considered:<br />

incarcerated inguinal hernia, malrotation of<br />

the bowel (particularly if vomiting is bilious),<br />

congenital causes of gastric outlet obstruction (e.g.<br />

webs or duplications), or medical conditions (e.g.<br />

sepsis, raised intracranial pressure).<br />

Over the past 30 years, increased reliance on<br />

imaging has decreased clinical diagnosis from up to<br />

90% of cases 147,151,152 to as little as 23%. 152<br />

Ultrasound diagnosis, which has a sensitivity and<br />

specificity for detecting IHPS of 95–100%, 153,154 is<br />

the most popular mode of imaging, but some<br />

groups continue to use upper GI contrast studies<br />

(Figure 35.7). At least one imaging study is now<br />

performed in 96%, with diagnosis by radiological<br />

means alone having increased to up to 65%. 155 The<br />

criteria for ultrasound diagnosis of IHPS (Figure<br />

35.8) in a term infant include a <strong>pylori</strong>c diameter of<br />

>11mm, muscle wall thickness of >3mm and<br />

<strong>pylori</strong>c canal length of >16mm. However, overreliance<br />

on imaging of a clinically obvious palpable<br />

tumor is cost-ineffective, and may delay treatment<br />

unnecessarily, particularly if a false-negative result<br />

is reported, 156 and should be discouraged.<br />

Infants with suspected IHPS should routinely<br />

undergo analysis of serum electrolytes and<br />

acid–base status, which will typically demonstrate<br />

a hypokalemic, hypochloremic metabolic acidosis,<br />

secondary to loss of gastric acid and dehydration<br />

from vomiting.<br />

Treatment options<br />

Initial management of IHPS is rehydration and<br />

correction of biochemical derangements. Various<br />

fluid regimens are advocated; one such is an infusion<br />

of 0.45% saline with dextrose solution (10%<br />

Figure 35.7 Upper gastrointestinal contrast study<br />

demonstrating narrowed gastric outlet from <strong>pylori</strong>c<br />

stenosis (‘string sign’).<br />

for neonates and 4% for older infants) with<br />

10mmol potassium chloride. Only after correction<br />

and adequate rehydration is the definitive<br />

surgery performed. The operation, Fredet–<br />

Ramstedt’s pyloromyotomy, may be performed by<br />

the open or laparoscopic method. Whilst the<br />

laparoscopic approach offers better cosmesis,<br />

clinical outcomes appear comparable. 157,158 The<br />

open method may be performed using a vertical<br />

midline, right upper quadrant transverse, or an<br />

umbilical incision. No difference in complications<br />

were found between the right upper quadrant<br />

and vertical incisions. 159 The hypertrophic<br />

<strong>pylori</strong>c muscle (Figure 35.9) is split longitudinally,<br />

releasing the mucosal tension within. The<br />

optimum timing of re-commencing feeding postoperatively<br />

following an uncomplicated pyloromyotomy<br />

remains controversial. Advocates of<br />

early feeding ad libitum immediately following<br />

recovery from anesthesia report shorter time to<br />

full feeds and shorter hospital stay. 160 Others,<br />

who report higher incidence of emesis from such


Figure 35.8 Ultrasound scan demonstrating <strong>pylori</strong>c<br />

stenosis.<br />

a regimen, favor delaying feeding for 6–12h. 161<br />

Feeding is usually delayed for 24h in those<br />

complicated with an intraoperative mucosal<br />

perforation.<br />

Inguinal hernia<br />

A hernia is defined as an abnormal protrusion of a<br />

viscus or part of a viscus through its coverings into<br />

an abnormal anatomical site. Inguinal hernia is the<br />

most common condition requiring surgery during<br />

childhood. Almost all are indirect, i.e. they pass<br />

through the internal inguinal ring and through the<br />

inguinal canal.<br />

Epidemiology<br />

Inguinal hernias affect one in 50 term male infants<br />

and up to 30% of premature infants. 162 There is a<br />

male preponderance, reported between 4:1 and<br />

9:1. 163–169 In males, 60% of hernias occur on the<br />

right, 30% occur on the left and 10% are bilateral. 170<br />

Bilateral herniais are more common in females and<br />

in premature infants (15–24%). 166,169,171,172 A contralateral<br />

hernia is found in 50–60% of infants<br />

Figure 35.9 Ramstedt’s pyloromyotomy.<br />

Inguinal hernia 591<br />

under 6 months of age, although the majority of<br />

these are not manifested clinically.<br />

Pathogenesis<br />

The processus vaginalis is a peritoneal diverticulum<br />

that passes through the internal inguinal ring<br />

at 3 months in utero. During the 7th month in utero,<br />

the testis descends from its intra-abdominal position<br />

at the urogenital ridge, into the scrotum, and<br />

as it does so it takes part of the processus with it<br />

into the scrotum. The processus is usually obliterated<br />

shortly before birth, leaving only the portion<br />

of the processus surrounding the testis, which is<br />

now called the tunica vaginalis. In 10–20% of the<br />

population the processus remains patent throughout<br />

life. 173 Failure of obliteration of the processus<br />

leaves a potential communicating sac that allows<br />

passage of peritoneal fluid (hydrocele) or abdominal<br />

contents (hernia) into the groin or scrotum. The<br />

hernial sac may contain bowel or, additionally in<br />

girls, ovary or Fallopian tube. The descent of the<br />

right testis and the subsequent obliteration of its<br />

processus vaginalis occur later than the left. For<br />

this reason right-sided hernias are twice as<br />

common. In girls, the canal of Nuck undergoes the<br />

same obliteration as the processus vaginalis in<br />

boys, but the obliteration is more likely to be<br />

complete, explaining the lower incidence of<br />

inguinal hernias in girls. If the hernia becomes irreducible<br />

(incarceration), the sac contents are at risk<br />

of injury. Incarceration of bowel within the hernia


592<br />

Gastrointestinal problems of the newborn<br />

is most common, and it may lead to the development<br />

of intestinal obstruction or strangulation.<br />

Etiology<br />

A number of patient populations have been identified<br />

with increased incidence of inguinal hernias.<br />

Premature infants have the highest incidence (up<br />

to 30% 162 ) of inguinal hernias, owing to the premature<br />

birth preceding full completion of the obliteration<br />

of the processus vaginalis. Cystic fibrosis<br />

patients have a 15% incidence of inguinal<br />

hernias. 174 This increased risk is thought to be<br />

related to an altered embryogenesis of the Wolffian<br />

duct structures that also leads to an absent vas<br />

deferens in male infants with cystic fibrosis.<br />

Increased incidence is also found in patients with<br />

connective tissue disorders (e.g. Ehlers–Danlos<br />

syndrome 175,176 ) and mucopolysaccharidosis<br />

(Hunter–Hurler syndrome 177 ); children with<br />

ventriculoperitoneal shunt; 178 children receiving<br />

chronic peritoneal dialysis; 179,180 and children<br />

with congenital dislocation of the hips. 181<br />

Clinical signs and symptoms<br />

The hernia is often noticed by parents, for example<br />

on changing the infant’s diaper, as a swelling,<br />

either in the groin (inguinal hernia) or scrotum<br />

(inguinoscrotal hernia; Figure 35.10). In most<br />

cases the infant is asymptomatic. The hernia may<br />

become prominent on straining or crying.<br />

However, a child in discomfort, or who is vomiting,<br />

particularly bile, should alert to the possibility<br />

of an obstructed or strangulated inguinal hernia.<br />

Features of intestinal obstruction include bilious<br />

vomiting, constipation and abdominal distension.<br />

A strangulated hernia will cause severe discomfort<br />

from ischemic bowel or testis and tenderness of<br />

hernial contents. It may cause passage of blood in<br />

the stools, with or without features of obstruction.<br />

Occasionally the only clinical evidence of a<br />

hernial sac may be a thickened spermatic cord<br />

compared to the contralateral cord.<br />

Complications<br />

Of infants with an inguinal hernia, 20–50% 182,183<br />

present with incarceration, the risk being highest<br />

Figure 35.10 Bilateral inguinoscrotal hernias.<br />

in the first 6 months of life. The main complications<br />

associated with incarceration (most<br />

frequently bowel contents) are intestinal obstruction<br />

or strangulation with testicular necrosis<br />

and/or bowel necrosis.<br />

Diagnosis with differential<br />

The primary differential diagnosis in boys is<br />

between a hernia and a hydrocele. The latter is<br />

cystic in consistency and easily transilluminates.<br />

Other differentials include an undescended or<br />

retractile testis, inguinal lymphadenopathy and<br />

rarely inguinal abscess. The ability to delineate the<br />

upper margin of the swelling distinguishes it from<br />

an inguinal hernia. The diagnosis is almost exclusively<br />

clinical; rarely, ultrasound confirmation<br />

may be used where there is reason for doubt. The<br />

presence of an intestinal obstruction complicating<br />

an incarcerated inguinal hernia may be confirmed<br />

by plain abdominal X-ray, which will demonstrate<br />

dilated loops of bowel.


Treatment options<br />

Surgery is indicated in all cases, because of the<br />

danger of incarceration and strangulation of<br />

hernial sac contents. The timing of surgery is<br />

determined by three factors: the age of the child,<br />

its clinical condition, and whether the hernia has<br />

become complicated. In infants, in whom strangulation<br />

is most common, the hernia operation<br />

should be performed at the earliest possible date,<br />

even if the infant is asymptomatic, provided the<br />

infant’s clinical status allows it. Premature, ventilated<br />

and critically ill infants with an uncomplicated<br />

hernia may have it repaired once they are<br />

stable. An irreducible hernia with or without<br />

strangulation or obstruction requires immediate<br />

surgery, irrespective of age. An incarcerated hernia<br />

that has been reduced successfully but with great<br />

difficulty may benefit from delayed repair of the<br />

hernia after 24–48h, to allow edema of bowel<br />

contents that is usually present to settle preoperatively.<br />

The hernia may be repaired by the open or laparoscopic<br />

method, although the optimal approach<br />

remains unproven. At surgery, if the hernia is<br />

uncomplicated, the PPV is mobilized and ligated at<br />

the internal ring. If the hernia is complicated by<br />

non-viable bowel within the sac, resection of the<br />

affected bowel with end-to-end anastomosis is<br />

performed. The decision as to whether the<br />

contralateral groin should be explored routinely<br />

under the same anesthetic remains controversial.<br />

163,169,184 This practice is generally not favored,<br />

REFERENCES<br />

1. Kliegman RM, Fanaroff AA. Necrotizing enterocolitis. N<br />

Engl J Med 1984; 310: 1093–1103.<br />

2. Wiswell TE, Robertson CF, Jones TA, Tuttle DJ.<br />

Necrotizing enterocolitis in full-term infants. A<br />

case–control study. Am J Dis Child 1988; 142: 532–535.<br />

3. Kliegman RM, Fanaroff AA. <strong>Neonatal</strong> necrotizing enterocolitis:<br />

a nine-year experience. Am J Dis Child 1981;<br />

135: 603–607.<br />

4. Leong GM, Drew JH. Necrotizing enterocolitis: a 15-year<br />

experience. Aust NZ J Obstet Gynaecol 1987; 27: 40–44.<br />

References 593<br />

as it would lead to unnecessary surgery in about<br />

80% and has the potential for damage to cord<br />

structures. However, the argument is less decisive<br />

in premature infants, who have a higher incidence<br />

of contralateral patent processus and risk of incarceration<br />

in the first 6 months of life. The laparoscopic<br />

approach to the repair of the hernia has the<br />

added benefit of allowing inspection of the abdominal<br />

exit of the contralateral internal inguinal ring<br />

during repair of the primary hernia, and avoids a<br />

potentially unnecessary contralateral groin exploration.<br />

Postoperative complication rates from inguinal<br />

hernia repair are low, particularly with uncomplicated<br />

hernias. Reported complications include:<br />

(1) Testicular atrophy by up to 25% volume, seen<br />

in 6% of cases. 185 The incidence of atrophy<br />

(15%) is highest after a period of incarceration<br />

and strangulation, and may be<br />

compounded by iatrogenic surgical trauma of<br />

testicular vessels;<br />

(2) Wound infection/hematoma (1–4% 165–169 );<br />

(3) Recurrence of the hernia (1–4% 165–168,182 );<br />

(4) Ascending testis, which may occur in up to<br />

2%, 167–169,186 owing to surgical adhesions<br />

restraining cord structures. These infants<br />

may require orchidopexy to return the testis<br />

to the scrotum;<br />

(5) Iatrogenic damage to cord structures, e.g. vas<br />

deferens (0–2%).<br />

5. Cikrit D, Mastandrea J, West KW et al. Necrotizing enterocolitis:<br />

factors affecting mortality in 101 surgical<br />

cases. Surgery 1984; 96: 648–655.<br />

6. Llanos AR, Moss ME, Pinzon MC et al. Epidemiology of<br />

neonatal necrotising enterocolitis: a population-based<br />

study. Paediatr Perinat Epidemiol 2002; 16: 342–349.<br />

7. Guthrie SO, Gordon PV, Thomas V et al. Necrotizing<br />

enterocolitis among neonates in the United States. J<br />

Perinatol 2003; 23: 278–285.


594<br />

Gastrointestinal problems of the newborn<br />

8. Kanto WP Jr, Wilson R, Ricketts RR. Management and<br />

outcome of necrotizing enterocolitis. Clin Pediatr (Phila)<br />

1985; 24: 79–82.<br />

9. Ricketts RR, Jerles ML. <strong>Neonatal</strong> necrotizing enterocolitis:<br />

experience with 100 consecutive surgical patients.<br />

World J Surg 1990; 14: 600–605.<br />

10. Narang A, Rao R, Bhakoo ON. <strong>Neonatal</strong> necrotizing<br />

enterocolitis an epidemiological study. Indian Pediatr<br />

1993; 30: 1207–1214.<br />

11. de Souza JC, da Motta UI, Ketzer CR. Prognostic factors<br />

of mortality in newborns with necrotizing enterocolitis<br />

submitted to exploratory laparotomy. J Pediatr Surg<br />

2001; 36: 482–486.<br />

12. McGuire W, Anthony MY. Donor human milk versus<br />

formula for preventing necrotising enterocolitis in<br />

preterm infants: systematic review. Arch Dis Child Fetal<br />

<strong>Neonatal</strong> Ed 2003; 88: F11–F14.<br />

13. Lucas A, Cole TJ. Breast milk and neonatal necrotising<br />

enterocolitis. Lancet 1990; 336: 1519–1523.<br />

14. Buch NA, Ahmad SM, Ali SW, Hassan HM. An epidemiological<br />

study of neonatal necrotizing enterocolitis.<br />

Saudi Med J 2001; 22: 231–237.<br />

15. Barlow B, Santulli TV, Heird WC et al. An experimental<br />

study of acute neonatal enterocolitis – the importance of<br />

breast milk. J Pediatr Surg 1974; 9: 587–595.<br />

16. Yoshioka H, Iseki K, Fujita K. Development and differences<br />

of intestinal flora in the neonatal period in breastfed<br />

and bottle-fed infants. Pediatrics 1983; 72: 317–321.<br />

17. Gibson GR, Wang X. Regulatory effects of bifidobacteria<br />

on the growth of other colonic bacteria. J Appl Bacteriol<br />

1994; 77: 412–420.<br />

18. Book LS, Herbst JJ, Atherton SO, Jung AL. Necrotizing<br />

enterocolitis in low-birth-weight infants fed an elemental<br />

formula. J Pediatr 1975; 87: 602–605.<br />

19. Walther FJ, Verloove-Vanhorick SP, Brand R, Ruys JH. A<br />

prospective survey of necrotising enterocolitis in very<br />

low birthweight infants. Paediatr Perinat Epidemiol<br />

1989; 3: 53–61.<br />

20. Kliegman RM. Models of the pathogenesis of necrotizing<br />

enterocolitis. J Pediatr 1990; 117: S2–S5.<br />

21. Ford H, Watkins S, Reblock K, Rowe M. The role of<br />

inflammatory cytokines and nitric oxide in the pathogenesis<br />

of necrotizing enterocolitis. J Pediatr Surg 1997;<br />

32: 275–282.<br />

22. Ford HR, Sorrells DL, Knisely AS. Inflammatory<br />

cytokines, nitric oxide, and necrotizing enterocolitis.<br />

Semin Pediatr Surg 1996; 5: 155–159.<br />

23. Morecroft JA, Spitz L, Hamilton PA, Holmes SJ. Plasma<br />

cytokine levels in necrotizing enterocolitis. Acta<br />

Paediatr Suppl 1994; 396: 18–20.<br />

24. Caplan MS, Hedlund E, Adler L, Hsueh W. Role of<br />

asphyxia and feeding in a neonatal rat model of necrotizing<br />

enterocolitis. Pediatr Pathol 1994; 14: 1017–1028.<br />

25. Muguruma K, Gray PW, Tjoelker LW, Johnston JM. The<br />

central role of PAF in necrotizing enterocolitis development.<br />

Adv Exp Med Biol 1997; 407: 379–382.<br />

26. Vannucci RC. Current and potentially new management<br />

strategies for perinatal hypoxic–ischemic encephalopathy.<br />

Pediatrics 1990; 85: 961–968.<br />

27. Kliegman RM, Fanaroff AA. <strong>Neonatal</strong> necrotizing enterocolitis<br />

in the absence of pneumatosis intestinalis. Am J<br />

Dis Child 1982; 136: 618–620.<br />

28. Kliegman RM. <strong>Neonatal</strong> necrotizing enterocolitis: implications<br />

for an infectious disease. Pediatr Clin North Am<br />

1979; 26: 327–344.<br />

29. Bell MJ, Shackelford P, Feigin RD et al. Epidemiologic<br />

and bacteriologic evaluation of neonatal necrotizing<br />

enterocolitis. J Pediatr Surg 1979; 14: 1–4.<br />

30. Lawrence G, Bates J, Gaul A. Pathogenesis of neonatal<br />

necrotising enterocolitis. Lancet 1982; 1: 137–139.<br />

31. Han VK, Sayed H, Chance GW et al. An outbreak of<br />

Clostridium difficile necrotizing enterocolitis: a case for<br />

oral vancomycin therapy? Pediatrics 1983; 71: 935–941.<br />

32. Dobson SR, Baker CJ. Enterococcal sepsis in neonates:<br />

features by age at onset and occurrence of focal infection.<br />

Pediatrics 1990; 85: 165–171.<br />

33. Shin CE, Falcone RA Jr, Stuart L et al. Diminished<br />

epidermal growth factor levels in infants with necrotizing<br />

enterocolitis. J Pediatr Surg 2000; 35: 173–176.<br />

34. Lawrence JP, Brevetti L, Obiso RJ et al. Effects of epidermal<br />

growth factor and Clostridium difficile toxin B in a<br />

model of mucosal injury. J Pediatr Surg 1997; 32:<br />

430–433.<br />

35. Nanthakumar NN, Fusunyan RD, Sanderson I, Walker<br />

WA. Inflammation in the developing human intestine: a<br />

possible pathophysiologic contribution to necrotizing<br />

enterocolitis. Proc Natl Acad Sci USA 2000; 97:<br />

6043–6048.<br />

36. Walker WA. Breast milk and the prevention of neonatal<br />

and preterm gastrointestinal disease states: a new<br />

perspective. Zhonghua Min Guo Xiao Er Ke Yi Xue Hui<br />

Za Zhi 1997; 38: 321–331.<br />

37. Fasoli L, Turi RA, Spitz L et al. Necrotizing enterocolitis:<br />

extent of disease and surgical treatment. J Pediatr Surg<br />

1999; 34: 1096–1099.<br />

38. Yu VY, Tudehope DI, Gill GJ. <strong>Neonatal</strong> necrotizing enterocolitis:<br />

1. Clinical aspects. Med J Aust 1977; 1:<br />

685–688.<br />

39. Bell MJ, Ternberg JL, Feigin RD et al. <strong>Neonatal</strong> necrotizing<br />

enterocolitis. Therapeutic decisions based upon<br />

clinical staging. Ann Surg 1978; 187: 1–7.<br />

40. Walsh MC, Kliegman RM. Necrotizing enterocolitis:<br />

treatment based on staging criteria. Pediatr Clin North<br />

Am 1986; 33: 179–201.<br />

41. Stevenson DK, Kerner JA, Malachowski N, Sunshine P.<br />

Late morbidity among survivors of necrotizing enterocolitis.<br />

Pediatrics 1980; 66: 925–927.<br />

42. Ricketts RR. Surgical treatment of necrotizing enterocolitis<br />

and the short bowel syndrome. Clin Perinatol 1994;<br />

21: 365–387.<br />

43. Grosfeld JL, Rescorla FJ, West KW. Short bowel<br />

syndrome in infancy and childhood. Analysis of<br />

survival in 60 patients. Am J Surg 1986; 151: 41–46.<br />

44. Chacko J, Ford WD, Haslam R. Growth and neurodevelopmental<br />

outcome in extremely-low-birth-weight<br />

infants after laparotomy. Pediatr Surg Int 1999; 15:<br />

496–499.<br />

45. Vohr BR, Wright LL, Dusick AM et al.<br />

Neurodevelopmental and functional outcomes of<br />

extremely low birth weight infants in the National<br />

Institute of Child Health and Human Development<br />

<strong>Neonatal</strong> Research Network, 1993–1994. Pediatrics<br />

2000; 105: 1216–1226.<br />

46. Simon NP, Brady NR, Stafford RL, Powell RW. The effect<br />

of abdominal incisions on early motor development of<br />

infants with necrotizing enterocolitis. Dev Med Child<br />

Neurol 1993; 35: 49–53.<br />

47. Voss M, Moore SW, van der Merwe, I, Pieper C.<br />

Fulminanting necrotising enterocolitis: outcome and<br />

prognostic factors. Pediatr Surg Int 1998; 13: 576–580.<br />

48. Grosfeld JL, Molinari F, Chaet M et al. Gastrointestinal<br />

perforation and peritonitis in infants and children:<br />

experience with 179 cases over ten years. Surgery 1996;<br />

120: 650–655.<br />

49. Stanford A, Upperman JS, Boyle P et al. Long-term<br />

follow-up of patients with necrotizing enterocolitis. J<br />

Pediatr Surg 2002; 37: 1048–1050.<br />

50. Ververidis M, Kiely EM, Spitz L et al. The clinical<br />

significance of thrombocytopenia in neonates with


necrotizing enterocolitis. J Pediatr Surg 2001; 36:<br />

799–803.<br />

51. Kosloske AM. Surgery of necrotizing enterocolitis. World<br />

J Surg 1985; 9: 277–284.<br />

52. Ein SH, Marshall DG, Girvan D. Peritoneal drainage<br />

under local anesthesia for perforations from necrotizing<br />

enterocolitis. J Pediatr Surg 1977; 12: 963–967.<br />

53. Albanese CT, Rowe MI. Necrotizing enterocolitis. In<br />

O’Neill JAJ, Rowe MI, Grosfeld JL et al., eds. Pediatric<br />

Surgery, vol 2. St Louis: Mosby, 1998: 1297–1332.<br />

54. Pierro A. Necrotizing enterocolitis: pathogenesis and<br />

treatment. Br J Hosp Med 1997; 58: 126–128.<br />

55. Fasoli L, Turi RA, Spitz L et al. Necrotizing enterocolitis:<br />

extent of disease and surgical treatment. J Pediatr Surg<br />

1999; 34: 1096–1099.<br />

56. Sparnon AL, Kiely EM. Resection and primary anastomosis<br />

for necrotizing enterocolitis. Pediatr Surg Int<br />

1987; 2: 101–104.<br />

57. Cooper A, Ross AJ III, O’Neill JA Jr, Schnaufer L.<br />

Resection with primary anastomosis for necrotizing<br />

enterocolitis: a contrasting view. J Pediatr Surg 1988; 23:<br />

64–68.<br />

58. Griffiths DM, Forbes DA, Pemberton PJ, Penn IA.<br />

Primary anastomosis for necrotising enterocolitis: a 12year<br />

experience. J Pediatr Surg 1989; 24: 515–518.<br />

59. Harberg FJ, McGill CW, Saleem MM et al. Resection<br />

with primary anastomosis for necrotizing enterocolitis. J<br />

Pediatr Surg 1983; 18: 743–746.<br />

60. Yaseen H, Kamaledin K, Al Umran K et al.<br />

Epidemiology and outcome of ‘early-onset’ vs ‘lateonset’<br />

necrotizing enterocolitis. Indian J Pediatr 2002;<br />

69: 481–484.<br />

61. Narang A, Rao R, Bhakoo ON. <strong>Neonatal</strong> necrotizing<br />

enterocolitis: a clinical study. Indian Pediatr 1993; 30:<br />

1417–1422.<br />

62. Kabeer A, Gunnlaugsson S, Coren C. <strong>Neonatal</strong> necrotizing<br />

enterocolitis. A 12–year review at a county hospital.<br />

Dis Colon Rectum 1995; 38: 866–872.<br />

63. Rowe MI, Reblock KK, Kurkchubasche AG, Healey PJ.<br />

Necrotizing enterocolitis in the extremely low birth<br />

weight infant. J Pediatr Surg 1994; 29: 987–990.<br />

64. Fasching G, Hollwarth ME, Schmidt B. Necrotizing enterocolitis<br />

in very-low-birth-weight infants. Pediatr Surg<br />

Int 1992; 7: 428–430.<br />

65. Beasley SW, Auldist AW, Ramanujan TM. The surgical<br />

management of neonatal necrotizing enterocolitis<br />

1975–1984. Pediatr Surg Int 1994; 1: 210–217.<br />

66. Chan K, Ohlsson A, Synnes A et al. Survival, morbidity,<br />

and resource use of infants of 25 weeks’ gestational age<br />

or less. Am J Obstet Gynecol 2001; 185: 220–226.<br />

67. Horwitz JR, Lally KP, Cheu HW et al. Complications<br />

after surgical intervention for necrotizing enterocolitis: a<br />

multicenter review. J Pediatr Surg 1995; 30: 994–998.<br />

68. Rivera-Moreno MA, Mercado-Arellano JA, Ulloa-<br />

Ricardez A, Franco-Gutierrez M. [Risk factors related to<br />

mortality in newborns with stage III necrosing enterocolitis].<br />

Gac Med Mex 1999; 135: 245–251.<br />

69. Ladd AP, Rescorla FJ, West KW et al. Long-term followup<br />

after bowel resection for necrotizing enterocolitis:<br />

factors affecting outcome. J Pediatr Surg 1998; 33:<br />

967–972.<br />

70. Georgeson KE, Breaux CW Jr. Outcome and intestinal<br />

adaptation in neonatal short–bowel syndrome. J Pediatr<br />

Surg 1992; 27: 344–348.<br />

71. Bagucka B, De Schepper J, Peelman M et al. Acid<br />

gastro–esophageal reflux in the 10 degrees-reversed-<br />

Trendelenburg-position in supine sleeping infants. Acta<br />

Paediatr Taiwan 1999; 40: 298–301.<br />

References 595<br />

72. El Mouzan MI, Abdullah AM, Al Sanie AM, Al Khalifah<br />

SI. Pattern of gastroesophageal reflux in children. Saudi<br />

Med J 2001; 22: 419–422.<br />

73. Rice H, Seashore JH, Touloukian RJ. Evaluation of<br />

Nissen fundoplication in neurologically impaired children.<br />

J Pediatr Surg 1991; 26: 697–701.<br />

74. Fonkalsrud EW, Ashcraft KW, Coran AG et al. Surgical<br />

treatment of gastroesophageal reflux in children: a<br />

combined hospital study of 7467 patients. Pediatrics<br />

1998; 101: 419–422.<br />

75. Inge TH, Carmeci C, Ohara LJ et al. Outcome of Nissen<br />

fundoplication using intraoperative manometry in children.<br />

J Pediatr Surg 1998; 33: 1614–1617.<br />

76. Omari TI, Barnett C, Snel A et al. Mechanisms of<br />

gastroesophageal reflux in healthy premature infants. J<br />

Pediatr 1998; 133: 650–654.<br />

77. Cadiot G, Bruhat A, Rigaud D et al. Multivariate analysis<br />

of pathophysiological factors in reflux oesophagitis.<br />

Gut 1997; 40: 167–174.<br />

78. Koivusalo A, Rintala R, Lindahl H. Gastroesophageal<br />

reflux in children with a congenital abdominal wall<br />

defect. J Pediatr Surg 1999; 34: 1127–1129.<br />

79. Rasheed K, Coughlan G, O’Donnell B. Congenital<br />

diaphragmatic hernia in the newborn. Outcome in 59<br />

consecutive cases over a ten year period (1980–1989). Ir<br />

J Med Sci 1992; 161: 16–17.<br />

80. Mollitt DL, Golladay ES, Seibert JJ. Symptomatic<br />

gastroesophageal reflux following gastrostomy in neurologically<br />

impaired patients. Pediatrics 1985; 75:<br />

1124–1126.<br />

81. Papaila JG, Vane DW, Colville C et al. The effect of<br />

various types of gastrostomy on the lower esophageal<br />

sphincter. J Pediatr Surg 1987; 22: 1198–1202.<br />

82. Jolley SG, Tunell WP, Hoelzer DJ et al. Lower esophageal<br />

pressure changes with tube gastrostomy: a causative<br />

factor of gastroesophageal reflux in children? J Pediatr<br />

Surg 1986; 21: 624–627.<br />

83. Reyes AL, Cash AJ, Green SH, Booth IW.<br />

Gastrooesophageal reflux in children with cerebral<br />

palsy. Child Care Health Dev 1993; 19: 109–118.<br />

84. St Cyr JA, Ferrara TB, Thompson TR et al. Nissen<br />

fundoplication for gastroesophageal reflux in infants. J<br />

Thorac Cardiovasc Surg 1986; 92: 661–666.<br />

85. Downing SE, Lee JC. Laryngeal chemosensitivity: a<br />

possible mechanism for sudden infant death. Pediatrics<br />

1975; 55: 640–649.<br />

86. Harned HS, Myracle J Jr, Ferreiro J. Respiratory suppression<br />

and swallowing from introduction of fluids into the<br />

laryngeal region of the lamb. Pediatr Res 1978; 12:<br />

1003–1009.<br />

87. Krishnan U, Mitchell JD, Tobias V et al. Fat laden<br />

macrophages in tracheal aspirates as a marker of reflux<br />

aspiration: a negative report. J Pediatr Gastroenterol Nutr<br />

2002; 35: 309–313.<br />

88. Knauer-Fischer S, Ratjen F. Lipid-laden macrophages in<br />

bronchoalveolar lavage fluid as a marker for pulmonary<br />

aspiration. Pediatr Pulmonol 1999; 27: 419–422.<br />

89. Bailey DJ, Andres JM, Danek GD, Pineiro-Carrero VM.<br />

Lack of efficacy of thickened feeding as treatment for<br />

gastroesophageal reflux. J Pediatr 1987; 110: 187–189.<br />

90. Khoshoo V, Fried M, Pencharz P. Incidence of gastroesophageal<br />

reflux with casein and whey-based formulas.<br />

J Pediatr Gastroenterol Nutr 1993; 17: 116–117.<br />

91. Orenstein SR, Magill HL, Brooks P. Thickening of infant<br />

feedings for therapy of gastroesophageal reflux. J Pediatr<br />

1987; 110: 181–186.<br />

92. Vandenplas Y, Hachimi-Idrissi S, Casteels A et al. A<br />

clinical trial with an ‘anti-regurgitation’ formula. Eur J<br />

Pediatr 1994; 153: 419–423.


596<br />

Gastrointestinal problems of the newborn<br />

93. Carroll AE, Garrison MM, Christakis DA. A systematic<br />

review of nonpharmacological and nonsurgical therapies<br />

for gastroesophageal reflux in infants. Arch Pediatr<br />

Adolesc Med 2002; 156: 109–113.<br />

94. Orenstein SR, Whitington PF, Orenstein DM. The infant<br />

seat as treatment for gastroesophageal reflux. N Engl J<br />

Med 1983; 309: 760–763.<br />

95. Orenstein SR. Prone positioning in infant gastroesophageal<br />

reflux: is elevation of the head worth the<br />

trouble? J Pediatr 1990; 117: 184–187.<br />

96. Nissen R. Gastropexy and fundoplication in surgical<br />

treatment of hiatal hernia. Am J Dig Dis 1961; 6:<br />

959–961.<br />

97. Thal AP. A unified approach to surgical problems of the<br />

esophagogastric junction. Ann Surg 1968; 168: 542–550.<br />

98. Martinez JM, Halverson A, Magnuson DK, Sackier JM.<br />

Laparoscopic versus open Nissen fundoplication:<br />

outcome of surgery in monozygotic twins. J<br />

Laparoendosc Adv Surg Tech A 1997; 7: 323–326.<br />

99. Esposito C, Garipoli V, De Pasquale M et al.<br />

Laparoscopic versus traditional fundoplication in the<br />

treatment of children with refractory gastro-oesophageal<br />

reflux. Ital J Gastroenterol Hepatol 1997; 29: 399–402.<br />

100. Mattioli G, Repetto P, Carlini C et al. Laparoscopic vs<br />

open approach for the treatment of gastroesophageal<br />

reflux in children. Surg Endosc 2002; 16: 750–752.<br />

101. Collins JB III, Georgeson KE, Vicente Y, Hardin WD Jr.<br />

Comparison of open and laparoscopic gastrostomy and<br />

fundoplication in 120 patients. J Pediatr Surg 1995; 30:<br />

1065–1070.<br />

102. Triadafilopoulos G, Dibaise JK, Nostrant TT et al.<br />

Radiofrequency energy delivery to the gastroesophageal<br />

junction for the treatment of GERD. Gastrointest Endosc<br />

2001; 53: 407–415.<br />

103. Katz PO. Gastroesophageal reflux disease: new treatments.<br />

Rev Gastroenterol Disord 2002; 2: 66–74.<br />

104. Filipi CJ, Lehman GA, Rothstein RI et al. Transoral, flexible<br />

endoscopic suturing for treatment of GERD: a<br />

multicenter trial. Gastrointest Endosc 2001; 53: 416–422.<br />

105. Rothestein RI, Pohl H Grove M et al. Endoscopic gastric<br />

plication for the treatment of GERD; two-year follow up<br />

results. Am J Gastroenterol 2001; 96: 53 (abstr).<br />

106. Kubiak R, Spitz L, Kiely EM et al. Effectiveness of<br />

fundoplication in early infancy. J Pediatr Surg 1999; 34:<br />

295–299.<br />

107. Weber TR. A prospective analysis of factors influencing<br />

outcome after fundoplication. J Pediatr Surg 1995; 30:<br />

1061–1063.<br />

108. Vane DW, Harmel RP Jr, King DR, Boles ET Jr. The effectiveness<br />

of Nissen fundoplication in neurologically<br />

impaired children with gastroesophageal reflux. Surgery<br />

1985; 98: 662–667.<br />

109. Dedinsky GK, Vane DW, Black T et al. Complications<br />

and reoperation after Nissen fundoplication in childhood.<br />

Am J Surg 1987; 153: 177–183.<br />

110. Pearl RH, Robie DK, Ein SH et al. Complications of<br />

gastroesophageal antireflux surgery in neurologically<br />

impaired versus neurologically normal children. J<br />

Pediatr Surg 1990; 25: 1169–1173.<br />

111. Spillane AJ, Currie B, Shi E. Fundoplication in children:<br />

experience with 106 cases. Aust NZ J Surg 1996; 66:<br />

753–756.<br />

112. Bensoussan AL, Yazbeck S, Carceller–Blanchard A.<br />

Results and complications of Toupet partial posterior<br />

wrap: 10 years’ experience. J Pediatr Surg 1994; 29:<br />

1215–1217.<br />

113. Dalla Vecchia LK, Grosfeld JL, West KW et al.<br />

Reoperation after Nissen fundoplication in children<br />

with gastroesophageal reflux: experience with 130<br />

patients. Ann Surg 1997; 226: 315–321.<br />

114. Benson CD. Infantile <strong>pylori</strong>c stenosis. Historical aspects<br />

and current surgical concepts. Prog Pediatr Surg 1970; 1:<br />

63–88.<br />

115. Kerr AM. Unprecedented rise in incidence of infantile<br />

hypertrophic <strong>pylori</strong>c stenosis. BMJ 1980; 281: 714–715.<br />

116. Webb AR, Lari J, Dodge JA. Infantile hypertrophic<br />

<strong>pylori</strong>c stenosis in South Glamorgan 1970–9. Effects of<br />

changes in feeding practice. Arch Dis Child 1983; 58:<br />

586–590.<br />

117. Schechter R, Torfs CP, Bateson TF. The epidemiology of<br />

infantile hypertrophic <strong>pylori</strong>c stenosis. Paediatr Perinat<br />

Epidemiol 1997; 11: 407–427.<br />

118. Applegate MS, Druschel CM. The epidemiology of<br />

infantile hypertrophic <strong>pylori</strong>c stenosis in New York<br />

State, 1983 to 1990. Arch Pediatr Adolesc Med 1995;<br />

149: 1123–1129.<br />

119. Stringer MD, Brereton RJ. Current management of infantile<br />

hypertrophic <strong>pylori</strong>c stenosis. Br J Hosp Med 1990;<br />

43: 266–272.<br />

120. Habbick BF, Khanna C, To T. Infantile hypertrophic<br />

<strong>pylori</strong>c stenosis: a study of feeding practices and other<br />

possible causes. CMAJ 1989; 140: 401–404.<br />

121. Tandoh JF, Hesse AA. Infantile hypertrophic <strong>pylori</strong>c<br />

stenosis in Ghana. West Afr J Med 1992; 11: 135–139.<br />

122. Graham DA, Mogridge N, Abbott GD et al. Pyloric stenosis:<br />

the Christchurch experience. NZ Med J 1993; 106:<br />

57–59.<br />

123. Haahr P, Nielsen JP. Infantile hypertrophic <strong>pylori</strong>c stenosis.<br />

A 25-year study from the county of Viborg. Ugeskr<br />

Laeger 2000; 162: 3456–3459.<br />

124. Tam PK, Chan J. Increasing incidence of hypertrophic<br />

<strong>pylori</strong>c stenosis. Arch Dis Child 1991; 66: 530–531.<br />

125. Jedd MB, Melton LJ III, Griffin MR et al. Trends in<br />

infantile hypertrophic <strong>pylori</strong>c stenosis in Olmsted<br />

County, Minnesota, 1950–1984. Paediatr Perinat<br />

Epidemiol 1988; 2: 148–157.<br />

126. Finsen VR. Infantile hypertrophic <strong>pylori</strong>c stenosis –<br />

unusual familial incidence. Arch Dis Child 1979; 54:<br />

720–721.<br />

127. Mitchell LE, Risch N. The genetics of infantile hypertrophic<br />

<strong>pylori</strong>c stenosis. A reanalysis. Am J Dis Child<br />

1993; 147: 1203–1211.<br />

128. Hicks LM, Morgan A, Anderson MR. Pyloric stenosis – a<br />

report of triplet females and notes on its inheritance. J<br />

Pediatr Surg 1981; 16: 739–740.<br />

129. Spitz L. Congenital hypertrophic <strong>pylori</strong>c stenosis in<br />

triplets. Arch Dis Child 1974; 49: 325.<br />

130. Huang PL, Dawson TM, Bredt DS et al. Targeted disruption<br />

of the neuronal nitric oxide synthase gene. Cell<br />

1993; 75: 1273–1286.<br />

131. Kusafuka T, Puri P. Altered messenger RNA expression<br />

of the neuronal nitric oxide synthase gene in infantile<br />

hypertrophic <strong>pylori</strong>c stenosis. Pediatr Surg Int 1997; 12:<br />

576–579.<br />

132. Jedd MB, Melton LJ III, Griffin MR et al. Factors associated<br />

with infantile hypertrophic <strong>pylori</strong>c stenosis. Am J<br />

Dis Child 1988; 142: 334–337.<br />

133. Dodge JA. Infantile hypertrophic <strong>pylori</strong>c stenosis in<br />

Belfast, 1957–1969. Arch Dis Child 1975; 50: 171–178.<br />

134. Hitchcock NE, Gilmour AI, Gracey M, Burke V. Pyloric<br />

stenosis in Western Australia, 1971–84. Arch Dis Child<br />

1987; 62: 512–513.<br />

135. Pisacane A, de Luca U, Criscuolo L et al. Breast feeding<br />

and hypertrophic <strong>pylori</strong>c stenosis: population based<br />

case–control study. BMJ 1996; 312: 745–746.<br />

136. Wattchow DA, Furness JB, Costa M. Distribution and<br />

coexistence of peptides in nerve fibers of the external


muscle of the human gastrointestinal tract.<br />

Gastroenterology 1988; 95: 32–41.<br />

137. Jadcherla SR, Klee G, Berseth CL. Regulation of migrating<br />

motor complexes by motilin and pancreatic<br />

polypeptide in human infants. Pediatr Res 1997; 42:<br />

365–369.<br />

138. Boiron M, Dorval E, Metman EH et al. Erythromycin<br />

elicits opposite effects on antro-bulbar and duodenal<br />

motility: analysis in diabetics by cineradiography. Arch<br />

Physiol Biochem 1997; 105: 591–595.<br />

139. Honein MA, Paulozzi LJ, Himelright IM et al. Infantile<br />

hypertrophic <strong>pylori</strong>c stenosis after pertussis prophylaxis<br />

with erythromcyin: a case review and cohort study.<br />

Lancet 1999; 354: 2101–2105.<br />

140. SanFilippo A. Infantile hypertrophic <strong>pylori</strong>c stenosis<br />

related to ingestion of erythromycine estolate: a report<br />

of five cases. J Pediatr Surg 1976; 11: 177–180.<br />

141. Ohshiro K, Puri P. Increased insulin-like growth factor-I<br />

mRNA expression in <strong>pylori</strong>c muscle in infantile hypertrophic<br />

<strong>pylori</strong>c stenosis. Pediatr Surg Int 1998; 13:<br />

253–255.<br />

142. Ohshiro K, Puri P. Increased insulin-like growth factor<br />

and platelet-derived growth factor system in the <strong>pylori</strong>c<br />

muscle in infantile hypertrophic <strong>pylori</strong>c stenosis. J<br />

Pediatr Surg 1998; 33: 378–381.<br />

143. Pueyo C, Ohshiro K, Puri P. Increased transforming<br />

growth factor beta-11 in infantile hypertrophic <strong>pylori</strong>c<br />

stenosis. Presented at the international symposium State<br />

of the Art in Pediatric Surgery, Dublin, Ireland 1997.<br />

144. Miyazaki E, Yamataka T, Ohshiro K et al. Active collagen<br />

synthesis in infantile hypertrophic <strong>pylori</strong>c stenosis.<br />

Pediatr Surg Int 1998; 13: 237–239.<br />

145. Cass DT, Zhang AL. Extracellular matrix changes in<br />

congenital hypertrophic <strong>pylori</strong>c stenosis. Pediatr Surg<br />

Int 1991; 6: 190–194.<br />

146. Andrassy RJ, Haff RC, Larsen GL. Infantile hypertrophic<br />

<strong>pylori</strong>c stenosis during the first week of life.<br />

Approaches to diagnosis, based on observations of a<br />

newborn whose vomiting began on the first day. Clin<br />

Pediatr (Phila) 1977; 16: 475–476.<br />

147. Sharma M, Jain SK, Pathania OP, Taneja SB. The Indian<br />

experience with hypertrophic <strong>pylori</strong>c stenosis. Clin<br />

Pediatr (Phila) 1990; 29: 566–568.<br />

148. Hulka F, Harrison MW, Campbell TJ, Campbell JR.<br />

Complications of pyloromyotomy for infantile hypertrophic<br />

<strong>pylori</strong>c stenosis. Am J Surg 1997; 173: 450–452.<br />

149. Zhang AL, Cass DT, Dubois RS, Cartmill T. Infantile<br />

hypertrophic <strong>pylori</strong>c stenosis: a clinical review from a<br />

general hospital. J Paediatr Child Health 1993; 29:<br />

372–378.<br />

150. Mitchell KG, Cachia SM. Infantile hypertrophic <strong>pylori</strong>c<br />

stenosis. Scott Med J 1981; 26: 245–249.<br />

151. Macdessi J, Oates RK. Clinical diagnosis of <strong>pylori</strong>c<br />

stenosis: a declining art. BMJ 1993; 306: 553–555.<br />

152. Hulka F, Campbell TJ, Campbell JR, Harrison MW.<br />

Evolution in the recognition of infantile hypertrophic<br />

<strong>pylori</strong>c stenosis. Pediatrics 1997; 100: E9.<br />

153. Hernanz-Schulman M, Sells LL, Ambrosino MM et al.<br />

Hypertrophic <strong>pylori</strong>c stenosis in the infant without a<br />

palpable olive: accuracy of sonographic diagnosis.<br />

Radiology 1994; 193: 771–776.<br />

154. van der Schouw YT, van der Velden MT, Hitge-Boetes C<br />

et al. Diagnosis of hypertrophic <strong>pylori</strong>c stenosis: value<br />

of sonography when used in conjunction with clinical<br />

findings and laboratory data. Am J Roentgenol 1994;<br />

163: 905–909.<br />

155. Poon TS, Zhang AL, Cartmill T, Cass DT. Changing<br />

patterns of diagnosis and treatment of infantile hyper-<br />

References 597<br />

trophic <strong>pylori</strong>c stenosis: a clinical audit of 303 patients.<br />

J Pediatr Surg 1996; 31: 1611–1615.<br />

156. Abbas AE, Weiss SM, Alvear DT. Infantile hypertrophic<br />

<strong>pylori</strong>c stenosis: delays in diagnosis and overutilization<br />

of imaging modalities. Am Surg 1999; 65: 73–76.<br />

157. Downey EC Jr. Laparoscopic pyloromyotomy. Semin<br />

Pediatr Surg 1998; 7: 220–224.<br />

158. Bufo AJ, Merry C, Shah R et al. Laparoscopic pyloromyotomy:<br />

a safer technique. Pediatr Surg Int 1998; 13:<br />

240–242.<br />

159. Hingston G. Ramstedt’s pyloromyotomy – what is the<br />

correct incision? NZ Med J 1996; 109: 276–278.<br />

160. Garza JJ, Morash D, Dzakovic A et al. Ad libitum feeding<br />

decreases hospital stay for neonates after pyloromyotomy.<br />

J Pediatr Surg 2002; 37: 493–495.<br />

161. Golladay ES, Broadwater JR, Mollitt DL. Pyloric stenosis<br />

– a timed perspective. Arch.Surg 1987; 122: 825–826.<br />

162. Harper RG, Garcia A, Sia C. Inguinal hernia: a common<br />

problem of premature infants weighing 1,000 grams or<br />

less at birth. Pediatrics 1975; 56: 112–115.<br />

163. Surana R, Puri P. Is contralateral exploration necessary<br />

in infants with unilateral inguinal hernia? J Pediatr Surg<br />

1993; 28: 1026–1027.<br />

164. Misra D, Hewitt G, Potts SR et al. Inguinal herniotomy<br />

in young infants, with emphasis on premature neonates.<br />

J Pediatr Surg 1994; 29: 1496–1498.<br />

165. Fung A, Barsoum G, Bentley TM et al. Inguinal<br />

herniotomy in young infants. Br J Surg 1992; 79:<br />

1071–1072.<br />

166. Harvey MH, Johnstone MJ, Fossard DP. Inguinal<br />

herniotomy in children: a five year survey. Br J Surg<br />

1985; 72: 485–487.<br />

167. Tam PK, Tsang TM, Saing H. Inguinal hernia in Chinese<br />

children. Aust NZ J Surg 1988; 58: 403–406.<br />

168. Yeung YP, Cheng MS, Ho KL, Yip AW. Day-case inguinal<br />

herniotomy in Chinese children: retrospective study.<br />

Hong Kong Med J 2002; 8: 245–248.<br />

169. Morecroft JA, Stringer MD, Higgins M et al. Follow-up<br />

after inguinal herniotomy or surgery for hydrocele in<br />

boys. Br J Surg 1993; 80: 1613–1614.<br />

170. Momoh JT. External hernia in Nigerian children. Ann<br />

Trop Paediatr 1985; 5: 197–200.<br />

171. Tackett LD, Breuer CK, Luks FI et al. Incidence of<br />

contralateral inguinal hernia: a prospective analysis. J<br />

Pediatr Surg 1999; 34: 684–687.<br />

172. Peevy KJ, Speed FA, Hoff CJ. Epidemiology of inguinal<br />

hernia in preterm neonates. Pediatrics 1986; 77:<br />

246–247.<br />

173. Devlin HB. Management of Abdominal Hernias. London:<br />

Butterworths, 1988: 75.<br />

174. Holsclaw DS, Shwachman H. Increased incidence of<br />

inguinal hernia, hydrocele, and undescended testicle in<br />

males with cystic fibrosis. Pediatrics 1971; 48: 442–445.<br />

175. Liem MS, van der GY, Beemer FA, van Vroonhoven TJ.<br />

Increased risk for inguinal hernia in patients with<br />

Ehlers–Danlos syndrome. Surgery 1997; 122: 114–115.<br />

176. McEntyre RL, Raffensperger JG. Surgical complications<br />

of Ehlers–Danlos syndrome in children. J Pediatr Surg<br />

1977; 12: 531–535.<br />

177. Coran AG, Eraklis AJ. Inguinal hernia in the<br />

Hurler–Hunter syndrome. Surgery 1967; 61: 302–304.<br />

178. Moazam F, Glenn JD, Kaplan BJ et al. Inguinal hernias<br />

after ventriculoperitoneal shunt procedures in pediatric<br />

patients. Surg Gynecol Obstet 1984; 159: 570–572.<br />

179. Tank ES, Hatch DA. Hernias complicating chronic<br />

ambulatory peritoneal dialysis in children. J Pediatr<br />

Surg 1986; 21: 41–42.<br />

180. Tsai TC, Huang FY, Hsu JC et al. Continuous ambulatory<br />

peritoneal dialysis complicating with abdominal


598<br />

Gastrointestinal problems of the newborn<br />

hernias in children. Zhonghua Min Guo Xiao Er Ke Yi<br />

Xue Hui Za Zhi 1996; 37: 263–265.<br />

181. Uden A, Lindhagen T. Inguinal hernia in patients with<br />

congenital dislocation of the hip. A sign of general<br />

connective tissue disorder. Acta Orthop Scand 1988; 59:<br />

667–668.<br />

182. Carneiro PM. Inguinal herniotomy in children. East Afr<br />

Med J 1990; 67: 359–364.<br />

183. Palmer BV. Incarcerated inguinal hernia in children.<br />

Ann R Coll Surg Engl 1978; 60: 121–124.<br />

184. McGregor DB, Halverson K, McVay CB. The unilateral<br />

pediatric inguinal hernia: should the contralateral side<br />

by explored? J Pediatr Surg 1980; 15: 313–317.<br />

185. Leung WY, Poon M, Fan TW et al. Testicular volume of<br />

boys after inguinal herniotomy: combined clinical and<br />

radiological follow–up. Pediatr Surg Int 1999; 15: 40–41.<br />

186. Surana R, Puri P. Iatrogenic ascent of the testis: an<br />

under–recognized complication of inguinal hernia operation<br />

in children. Br J Urol 1994; 73: 580–581.<br />

187. Spitz L. Nissen fundoplication. In Dudley H, Carter D,<br />

Russell RCG et al., eds. Rob and Smith’s Operative<br />

Surgery. London: Buttersworth, 1988: 240–246.<br />

188. Bell MJ. Peritonitis in the newborn – current concepts.<br />

Pediatr Clin North Am 1985; 32: 1181–1201.<br />

189. Dykes EH, Fitzgerald RJ, O’Donnell B. Surgery for<br />

neonatal necrotising enterocolitis in Ireland 1980–1985.<br />

Intensive Care Med 1989; 15 Suppl 1: S24–S26.<br />

190. Flores-Nava G, Joachin-Roy H, Rodriguez-Cueto G. [Risk<br />

factors in neonatal necrotizing enterocolitis]. Biol Med<br />

Hosp Infant Mex 1993; 50: 645–649.<br />

191. Martinez-Tallo E, Claure N, Bancalari E. Necrotizing<br />

enterocolitis in full-term or near-term infants: risk<br />

factors. Biol Neonate 1997; 71: 292–298.<br />

192. Kadalraja R, Patole SK, Muller R, Whitehall JS.<br />

Comparison of clinical characteristics and high-risk<br />

factors in Australian aboriginal and non-aboriginal<br />

neonates with necrotising enterocolitis. Int J Clin Pract<br />

2001; 55: 251–254.<br />

193. Leung MP, Chau KT, Hui PW et al. Necrotizing enterocolitis<br />

in neonates with symptomatic congenital heart<br />

disease. J Pediatr 1988; 113: 1044–1046.<br />

194. Sehgal S, Ewing C, Waring P et al. Morbidity of lowbirthweight<br />

infants with intrauterine cocaine exposure.<br />

J Natl Med Assoc 1993; 85: 20–24.<br />

195. Lopez SL, Taeusch HW, Findlay RD, Walther FJ. Time of<br />

onset of necrotizing enterocolitis in newborn infants<br />

with known prenatal cocaine exposure. Clin Pediatr<br />

(Phila) 1995; 34: 424–429.<br />

196. Rand T, Weninger M, Kohlhauser C et al. Effects of<br />

umbilical arterial catheterization on mesenteric hemodynamics.<br />

Pediatr Radiol 1996; 26: 435–438.<br />

197. Lidums I, Lehmann A, Checklin H et al. Control of transient<br />

lower esophageal sphincter relaxations and reflux<br />

by the GABA(B) agonist baclofen in normal subjects.<br />

Gastroenterology 2000; 118: 7–13.<br />

198. Zhang Q, Lehmann A, Rigda R et al. Control of transient<br />

lower oesophageal sphincter relaxations and reflux by<br />

the GABA(B) agonist baclofen in patients with gastrooesophageal<br />

reflux disease. Gut 2002; 50: 19–24.


36<br />

Introduction<br />

Enteral nutrition in preterm<br />

infants<br />

Mario De Curtis and Jacques Rigo<br />

Currently, there is little consensus among neonatologists<br />

on the optimal way to initiate, proceed<br />

with or maintain enteral feeding in preterm<br />

infants. Most routines in neonatal intensive care<br />

units derive more from an empirical approach<br />

than from controlled studies. Nevertheless, the<br />

enormous number of studies conducted in this<br />

field over the past few years have greatly<br />

contributed to our knowledge of the nutrition of<br />

preterm infants, even if many doubts still remain.<br />

The gastrointestinal tract of preterm infants<br />

and especially of very-low-birth-weight (VLBW)<br />

infants is immature at birth and initially incapable<br />

of performing full digestion and absorption.<br />

Immature gastrointestinal motility manifests itself<br />

in so-called ‘feeding intolerance’, which is characterized<br />

by delayed gastric emptying, abdominal<br />

distension, water stool passage or constipation. 1<br />

The advent of intensive care and the increased<br />

survival of premature infants led to the problem of<br />

necrotizing enterocolitis (NEC), which almost<br />

always occurred in infants who were fed.<br />

Consequently, enteral feeding was avoided as a<br />

strategy to prevent NEC. The introduction of<br />

enteral feeding was delayed, often for prolonged<br />

periods and parenteral nutritional was considered<br />

as the best nutrition route for VLBW infants in the<br />

first period of life.<br />

Enteral nutrition during the early<br />

adaptive period<br />

More recent studies have suggested that associating<br />

parenteral nutrition and early postnatal enteral<br />

feeding with small amounts of human milk or<br />

formula (called ‘minimal enteral feeding’,<br />

‘gastrointestinal priming’, ‘trophic feeding’ and<br />

‘hypocaloric feeding’) could help improve the<br />

development of the gastrointestinal tract, gut<br />

hormone release and gut motility. 2 Results of clinical<br />

trials in premature infants support the opinion<br />

that minimal enteral feeding has some clinical<br />

benefits, such as reducing the time to start full<br />

enteral feeding and length of hospitalization<br />

without increasing the risk of NEC. 3 The precise<br />

impact of minimal enteral feedings is still difficult<br />

to evaluate, because the studies carried out, aside<br />

from being based on a limited number of infants,<br />

considered different criteria of inclusion, feeding<br />

protocols and outcome measures. Thus, the metaanalysis<br />

is a less effective tool for reaching clear<br />

conclusions, whereas a larger trial with a single<br />

uniform protocol could clarify many of these<br />

issues.<br />

In VLBW infants, the preferred early feeding<br />

regimen is human milk, which can be initiated,<br />

according to clinical conditions, even on the day of<br />

birth, or the following days, in small quantities,<br />

e.g. 1–2ml colostrum every 6h, progressively<br />

increasing the volume and frequency. Feeds<br />

should be given with a continuous infusion pump<br />

or by gavage. A recent review showed that there<br />

was no significant difference in somatic growth,<br />

days to discharge or the incidence of NEC for<br />

infants fed by continuous versus intermittent<br />

bolus tube feeding. 4 In any case the tube should be<br />

aspirated regularly in order to determine feeding<br />

tolerance. Gastric residues are usually measured<br />

every 3h in infants aged less than 32 weeks and<br />

even more frequently in smaller and sick infants.<br />

The aspirate is reinfused to reduce the loss of<br />

enzymes and electrolytes. There is little rationale<br />

for the practice of diluting milk; the dilution does<br />

not promote maturation of gut motility. 5<br />

599


600<br />

Enteral nutrition in preterm infants<br />

The ideal rate of progression of milk volume given<br />

to VLBW infants is controversial. While a Cochrane<br />

Review has concluded that a rapid rate of<br />

advancing feeds was not associated with a higher<br />

risk of NEC, 6 a recent study suggested that rapid<br />

advancement of enteral feeding might increase the<br />

risk of NEC. 7 Although additional studies are<br />

needed, a small progressive increment of 10ml/kg<br />

per day after a few days of stabilization can be<br />

recommended.<br />

Nutrition during the intermediate and<br />

stable growth period<br />

Transition from the early adaptive to an intermediate<br />

and stable growing period is related to stable<br />

clinical and metabolic conditions and tolerance of<br />

minimal enteral feeding. During this period enteral<br />

feeding can be slightly increased, up to<br />

140–180ml/kg per day, whereas the amount of<br />

parenteral fluid is progressively reduced and interrupted<br />

as soon as 100–120ml/kg per day is well<br />

tolerated.<br />

Nutrient needs during enteral feeding<br />

Water<br />

Total fluid intake is related to ingested calorie and<br />

protein intake as well as to the renal solute load.<br />

During the early phase there is a progressive cornification<br />

of the epidermidis causing a significant<br />

reduction of perspiratio insensibilis and an<br />

improvement of renal function. 8 The goal of fluid<br />

administration is to replace water loss, maintain<br />

water and electrolyte homeostasis and provide<br />

extra water and electrolytes to build up new<br />

tissues. Renal solute load (RSL) plays a major role<br />

in the water balance of enterally fed preterm<br />

infants. It can be defined as water-soluble waste<br />

products excreted through the kidney. The excretion<br />

of these products requires a given amount of<br />

water, which affects the net water balance. As the<br />

RSL increases, the minimal urinary water excretion<br />

increases, owing to the relative immaturity of<br />

the premature kidney to concentrate urine. The<br />

RSL is closely related to the nitrogen and electrolyte<br />

content of the diet, and its potential RSL<br />

can be estimated according to the following<br />

formula, expressed in millimoles (mmol, or<br />

milliosmoles, mOsm): Potential RSL=N/28+Na+<br />

Cl+K+P, where N/28 indicates the excretion, in<br />

mg, of nitrogenous substances such as urea (urea<br />

contains two nitrogen atoms, atomic weight 14),<br />

Na is sodium, Cl is chloride, K is potassium and P<br />

is available phosphorus which is the same as total<br />

phosphorus in milk-based formulas. 9 The<br />

potential RSL of mature human milk is<br />

14mOsm/100kcal, 10 whereas that of currently<br />

available premature infant formulas is about<br />

26mOsm/100kcal, 11 Maximum amounts of solutes<br />

in a formula providing 81kcal/100ml has been<br />

estimated in non-growing preterm infants as<br />

32mOsm/100kcal, 12 taking into account a formula<br />

intake of 150ml/kg per day, skin and lung water<br />

loss of 75ml/kg per day and stool loss of 8–11ml/kg<br />

per day. 13 In such conditions, at least 64ml/kg per<br />

day are available for urinary excretion, considering<br />

that the great majority of preterm infants without<br />

renal disease can concentrate urine to<br />

600mOsm/l. 14<br />

Because the growing infant incorporates into new<br />

tissue a portion of the substances present in the<br />

diet (protein, potassium, phosphate, etc.), which<br />

in the non-growing subject would be presented to<br />

the kidney for excretion, an ‘osmolar equivalent of<br />

weight gain’ was removed from the calculated<br />

potential renal solute load. A theoretical estimate<br />

of the size of this sparing effect, based on the body<br />

composition of the newborn infant, in a study on<br />

relatively large premature infants (mean birth<br />

weight 1787 g and gestational age 33 weeks),<br />

suggested an osmolar equivalent of 0.9mmol/g<br />

weight gain and half the expected true osmolar<br />

load of adapted formulas. 15 In the stable growing<br />

period, the water requirement, accounting for<br />

security levels, is 130–160ml/kg per day.<br />

Protein<br />

The goal in estimating the protein requirements of<br />

preterm infants is to provide the quantity and<br />

quality of protein needed to obtain a growth<br />

similar to that observed in the fetus during the<br />

third trimester of intrauterine life, and to obtain an<br />

early postnatal catch-up growth, keeping in mind<br />

that the accretion of lean body mass needs to be<br />

considered more than absolute weight gain. 16<br />

Protein utilization is influenced by several factors<br />

such as quality and quantity of the protein supply,


dietary protein energy ratio, gut immaturity and<br />

nutritional status. Dietary protein needs for<br />

preterm infants have been estimated by two different<br />

methods. The first, the empirical approach,<br />

measures biochemical or physiological responses<br />

to graded intakes. The second method, the factorial<br />

approach, considers the requirements as the<br />

sum of the essential losses (e.g. urine, feces, skin),<br />

plus the amount incorporated into newly formed<br />

tissues.<br />

The empirical approach evaluates physiological<br />

and biochemical variables to determine minimum<br />

and maximum protein needs in the growing<br />

preterm infant. The measures used in these types<br />

of study are: anthropometry (weight, length, head<br />

circumference, skinfold thickness); nitrogen<br />

balance or retention; metabolic or chemical<br />

indices (plasma and urinary amino acids, serum<br />

albumin, total protein, immunoglobulin, retinal<br />

binding protein and transthyretin – most of which<br />

provide an indirect reflection of protein synthesis);<br />

indirect calorimetry, to evaluate energy balance<br />

and – in combination with nitrogen balance – the<br />

composition of weight gain; isotope studies of<br />

whole-body nitrogen kinetics (used as indirect<br />

measures of protein synthesis rates); body composition,<br />

by dual energy X-ray absorptiometry; and<br />

development assessment, studying the effect of<br />

quality and quantity of protein on neurological<br />

development through various neurometric tests<br />

such as the <strong>Neonatal</strong> Behavioral Assessment scale<br />

and the Bayley Scales of Infant Development. 17<br />

In the factorial approach, compositional analysis<br />

of fetal tissues has been a valuable source of data<br />

for our understanding of the nutrient needs of the<br />

fetus and, by extension, those of the growing<br />

preterm infant. Fetal accretion rates have been<br />

obtained from compositional analyses of aborted<br />

fetuses or stillborn infants. 18 From these data, the<br />

protein increment for growth has been estimated<br />

as approximatly 2.3g/kg per day, 19 but more recent<br />

data seem to indicate a value closer to 2.5g/kg per<br />

day. Protein requirement is thus calculated by<br />

adding to this value the obligatory losses, and the<br />

amount needed for the additional catch-up<br />

growth. In a large population of preterm infants<br />

receiving a controlled energy intake, the minimal<br />

protein supply necessary to obtain a zero nitrogen<br />

balance (to cover all nitrogen losses) was calculated<br />

to be 0.74g/kg per day. 20 A similar value was<br />

Nutrition during the intermediate and stable growth period 601<br />

estimated to cover the need for catch-up growth. 21<br />

Thus, the protein requirements in VLBW infants,<br />

so calculated, reach 3.8–4g/kg per day. The recommended<br />

dietary protein allowance needs to be<br />

adapted to the fractional absorption rate and<br />

protein efficiency as well as to individual<br />

variations.<br />

Nitrogen absorption and utilization were recently<br />

reviewed on 226 metabolic balances performed in<br />

recent years in preterm infants fed human milk,<br />

human milk fortifiers and various preterm formulas.<br />

22 Data are summarized in Table 36.1.<br />

Nitrogen absorption rate (absorbed/intake) differs<br />

significantly according to the feeding regimen. It<br />

was higher with powder whey-predominant<br />

protein preterm formulas (90.7%) than with<br />

human milk supplemented with fortifiers (82.7%),<br />

powder protein hydrolyzed formulas (84.3%) or<br />

ready-to-use liquid whey-predominant preterm<br />

formulas (86.0 %) (Table 36.1). Those differences<br />

result from the nature of the various types of<br />

protein supply. In human milk, non-nutritional<br />

protein content (lactoferrin, sIgA, lysozyme) or<br />

non-protein nitrogen content (oligosaccharides)<br />

are absorbed less than the nutritional protein<br />

content (whey proteins, caseins) and contribute to<br />

a significant part of fecal excretion. An interesting<br />

observation is the relatively lower absorption rate<br />

of ready-to-use liquid preterm formulas or protein<br />

hydrolyzed preterm formulas, where the technical<br />

process seems to impair nitrogen absorption in<br />

relation to heat treatment – inducing some Maillard<br />

reaction – or to preliminary hydrolysis, altering<br />

the physiological absorption process in the<br />

lumen or at the border of the gastrointestinal<br />

tract. 23<br />

The efficiency of protein gain, estimated by the<br />

ratio between retained and metabolizable nitrogen<br />

(absorbed), differs also according to the feeding<br />

regimen (Table 36.1). The highest values were<br />

obtained in preterm infants fed powder (77.7%)<br />

and liquid (77.5%) preterm formulas. It was significantly<br />

lower in those fed protein-hydrolyzed<br />

formulas (74.0%) and fortified human milk<br />

(72.1%). 22 The lower value obtained with fortified<br />

human milk may be related to non-protein nitrogen,<br />

which represents 20–25% of total nitrogen<br />

content of human milk, but still 13.5–17% of total<br />

nitrogen content of fortified human milk. As


602<br />

Enteral nutrition in preterm infants<br />

Table 36.1 Nitrogen balances according to feeding regimen in preterm infants (from reference 22)<br />

Powder whey- Liquid whey- Powder<br />

predominant predominant protein-<br />

Amount Fortified preterm preterm hydrolyzed<br />

(mg/kg per day) human milka formulasb formulasc formulasd (n=88) (n=49) (n=58) (n=31)<br />

Intake 517±86 d 522 ± 70d 506 ± 58 553 ± 56ab Fecal excretion 90±28 bc 49±19acd 71±28abd 87±26bc Absorbed 428±76 bd 474±75ac 434±52 bd 466±51 ac<br />

Urinary excretion 121±45 c 106±36 98±21ad 122±39 c<br />

Retained 307±56 bcd 368±57acd 337±46 ab 343±42 ab<br />

Absorption (%) 82.7±4.8bc 90.7±3.3acd 86.0±5.0ab 84.3±4.0b Net protein utilization* (%) 59.7±7.7bc 71.5±6.5acd 66.6±5.8abd 62.4±6.5bc Protein efficiency † (%) 72.1±7.6bc 77.7±6.4ad 77.5±4.4ad 74.0 ± 6.9bc * Nitrogen retention/nitrogen intake<br />

†† Nitrogen retention/nitrogen absorption<br />

abcd Values with like letters p < 0.05<br />

demonstrated for urea nitrogen, the contribution of<br />

this metabolizable non-protein nitrogen fraction to<br />

protein gain is lower than that of the α-lactalbumin<br />

or the casein content of human milk.<br />

Therefore, regarding the net protein utilization (N<br />

retained/N intake), preterm formulas appear to be<br />

more efficient than human milk with or without<br />

protein supplementation. However, the net protein<br />

utilization of formulas can be altered by some<br />

technical processes, such as those required for<br />

hydrolysis. 23 Thus, adapted for fractional absorption<br />

rate and protein efficiency, the recommended<br />

protein allowance could reach 4.5g/kg per day in<br />

extremely-low-birth-weight (ELBW) infants with<br />

significant catch-up growth requirements. 16<br />

The potential risk of such an aggressive nutritional<br />

strategy is a metabolic stress resulting from protein<br />

overload or unbalanced amino acid supply.<br />

Therefore, the most recent technologies will be<br />

necessary to improve nitrogen bioavailability,<br />

reduce Maillard reactions and provide the most<br />

balanced amino acid composition.<br />

The whey/casein ratio significantly influences<br />

individual amino acid intakes and plasma amino<br />

acid concentrations. Plasma threonine is increased<br />

and tryptophan relatively decreased in infants fed<br />

a whey-predominant formula, whereas methionine<br />

and aromatic amino acids are increased in those<br />

fed a casein-predominant formula. 24,25 The high<br />

plasma threonine concentration, observed in<br />

preterm infants fed whey-predominant formulas,<br />

is related to the glycomacropeptide obtained from<br />

casein by enzymatic casein precipitation of cow’s<br />

milk proteins. Acidic precipitation, by contrast,<br />

removes the glycomacropeptide rich in threonine<br />

from the soluble phase. 26 Today it is possible to<br />

design whey-predominant formulas with a lower<br />

threonine content. 27–29 In 14 preterm infants<br />

receiving either an enzymatic or an acidic wheypredominant<br />

formula, a significant reduction<br />

in plasma threonine concentration was observed<br />

in acidic whey-protein formula-fed infants<br />

(27.9±8.5µmol/dl), compared to those receiving<br />

conventional enzymatic whey-protein formula<br />

(37.5±8.4µmol/dl). All other plasma amino acid<br />

concentrations were similar, with the exception of<br />

valine, which was slightly reduced in acidic whey<br />

protein formula-fed infants. 27<br />

In addition to these technological treatments,<br />

performed to remove the glycomacropeptide and<br />

reduce the threonine content, the ratio between<br />

different cow’s milk protein contents was modified


to increase the relative percentage of α-lactoalbumin.<br />

Indeed, this protein fraction is naturally rich<br />

in tryptophan and helps normalize the low levels<br />

of this amino acid frequently observed in wheypredominant<br />

formula-fed infants. 30,31<br />

Formulas based on hydrolyzed proteins have been<br />

recently proposed for the feeding of preterm<br />

infants to reduce gastrointestinal problems such as<br />

delayed gastric emptying, abdominal distension,<br />

hard stools and feeding intolerance. 32,33 The technological<br />

processes necessary to perform hydrolysis<br />

and reduce protein antigenicity may, however,<br />

modify the amino acid content and/or amino acid<br />

bioavailability. 34 The use of a higher percentage of<br />

whey in protein-hydrolyzed formulas further<br />

worsens the plasma amino acid pattern previously<br />

observed with the use of whey-predominant<br />

formulas by increasing threonine and decreasing<br />

aromatic amino acid concentrations. 32 Moreover, a<br />

significant decrease of plasma histidine and tryptophan<br />

concentrations, probably due to a relative<br />

reduction in amino acid bioavailability, was also<br />

observed. Using a more appropriate technology,<br />

these formulas have been corrected for the threonine<br />

content and supplemented with histidine and<br />

tryptophan. 35–37<br />

Formulas do not contain any of the biologically<br />

active immune substances, nor enzymes,<br />

hormones or growth factors found in human milk.<br />

The long-term implications of this deficiency has<br />

not been determined. Several studies have been<br />

performed to evaluate the functional properties of<br />

non-nutritional compounds, such as nucleotides,<br />

polyamines, growth factors or prebiotics, which<br />

could potentially improve the quality of these new<br />

formulas, although no definitive conclusions are<br />

currently available. 17<br />

Considering the revised protein recommendations,<br />

large multicenter randomized studies need to evaluate<br />

the improvement in growth and in body<br />

composition, as well as the reduction in incidence<br />

of postnatal growth restriction expected from an<br />

aggressive nutritional policy in preterm infants.<br />

Further studies are also needed to identify more<br />

sensitive markers of protein toxicity, to determine<br />

the safety and efficacy of such a high protein<br />

supply and to evaluate the beneficial impact on<br />

short- and long-term growth and development.<br />

Nutrition during the intermediate and stable growth period 603<br />

Energy<br />

The energy demands for preterm infants depend<br />

on many factors including fetal and neonatal<br />

development, genetically determined metabolic<br />

rate, thermal environment, activity, sleep status,<br />

nutritional status, nutrient intake, body composition<br />

and occurrence of illness. Energy intake<br />

recommendation for preterm infants, calculated<br />

for 150ml/kg per day of formula, by different nutrition<br />

committees, range between 98 and 139kcal/kg<br />

per day, while the more recent recommendations<br />

range between 110 and 135kcal/kg per day. 11,17,38,39<br />

Energy retention, in premature infants, after the<br />

first 2–3 days of life, is about 85–90% of energy<br />

intake. The remainder is lost in stool and a small<br />

quantity as urea in the urine. Energy retained is in<br />

part stored in tissues as protein and mainly as fat,<br />

and in part lost as energy expenditure. After birth,<br />

fat deposition represents a higher proportion of<br />

weight gain than in utero. 40 Rapid accretion of<br />

body fat appears to be unavoidable, although it can<br />

be altered considerably by dietary manipulation of<br />

the protein/energy ratio. 40.41 A fat deposition of<br />

20–25% of weight gain has been proposed as a<br />

reasonable goal for preterm infants but there are,<br />

however, no long-term data to justify this limit. 42<br />

Protein/energy ratio<br />

Protein and energy needs are reciprocally limiting,<br />

the intake of one affecting the ability of the infant<br />

to assimilate the other. A suboptimal range of the<br />

protein/energy ratio leads to untoward consequences.<br />

43 If energy intake is inadequate, protein<br />

is used as an energy source and the nitrogen<br />

balance becomes less positive. Increasing the<br />

caloric intake will spare protein loss and improve<br />

nitrogen retention. If there is a surfeit of energy<br />

with limited protein intake, the protein retention<br />

reaches a plateau and the energy excess is used<br />

only for fat deposition. 43 Nevertheless, when the<br />

protein supply is satisfactory (close to 4.0g/kg per<br />

day), the effect of energy increase from 130 to<br />

155kcal/kg per day on protein retention appears to<br />

be minimal – about 0.1g of protein/kg per day. 44<br />

An additional issue concerns the partition of<br />

energy intake between fat and carbohydrates. It<br />

seems that carbohydrates are slightly more effective<br />

than fat in sparing protein oxidation. At the<br />

same two levels of energy supply the advantage of


604<br />

Enteral nutrition in preterm infants<br />

a diet containing 65% of non-protein energy as<br />

carbohydrate versus 35% on nitrogen retention<br />

represented about 0.15g of protein/kg per day in<br />

enterally fed low-birth-weight (LBW) infants. 44<br />

Analysis of various studies in preterm infants fed<br />

human milk, 45–51 human milk fortifiers 46,49,52–57<br />

and various preterm formulas 45,47,50–52,55,56,58–61<br />

allows the evaluation of the main determinant of<br />

weight gain, nitrogen retention and fat mass deposition.<br />

From these data, the major determinants of<br />

weight gain, lean body mass gain, fat accretion and<br />

protein retention may be determined using<br />

stepwise regression analysis. Protein intake and<br />

protein/energy ratio are the main determinants of<br />

weight gain. Protein intake is the only determinant<br />

of lean body mass gain in contrast to fat mass gain,<br />

which is positively related to energy intake and<br />

negatively to protein/energy ratio. 16 Protein gain<br />

and lean body mass gain increase significantly<br />

with protein intake without any additional significant<br />

influence of energy intake and protein/energy<br />

ratio. According to these data, as well as to the<br />

factorial approach, new recommendations for<br />

protein and protein/energy ratio can be suggested<br />

(Table 36.2) in relation to post-conceptional age<br />

and the need for catch-up growth. 16 Thus, according<br />

to this calculation, recommended intakes for<br />

preterm infants at 26–30 weeks’ postconceptional<br />

age should have 3.8–4.4g of protein/kg per day<br />

with a protein/energy ratio between 3.0 and<br />

3.3g/100kcal, according to their relative postnatal<br />

growth restriction. These values are in the range of<br />

the recent suggestion from an Expert Panel of the<br />

Life Sciences Research Office and the American<br />

Society for Nutritional Sciences who suggested<br />

3.4–4.3g of protein/kg per day with an energy<br />

intake of 120kcal/kg per day and a protein/energy<br />

ratio between 2.5 and 3.6g/100kcal. 17<br />

Fat<br />

Fat provides the major source of energy in growing<br />

preterm infants, representing approximately 50%<br />

of the non-protein energy content of human milk<br />

and current infant formulas. Although a mainly<br />

carbohydrate-based diet appears beneficial in<br />

terms of sparing protein oxidation, enhancing<br />

growth and protein accretion as well as to improve<br />

quiet sleep and influence the distribution of<br />

behavior activity states in VLBW infants, it is<br />

generally accepted that metabolizable long-chain<br />

fat should be at least equal to the fat deposition in<br />

growing tissues. 62 Accordingly, a fat intake of<br />

5.4–7.2g/kg per day (or 40–54% of energy content)<br />

has been recommended. 38<br />

The newborn’s capability of digesting triglycerides<br />

is not fully developed in premature infants. Lipid<br />

malabsorption is the result of low levels of pancreatic<br />

lipase and bile salts. The bile acid pool is only<br />

half the size of that of a full-term infant. In premature<br />

infants there is an alternative mechanism for<br />

digestion of dietary fat, represented by intragastric<br />

lipolysis, due to lingual and gastric lipases,<br />

secreted by the 25th week of gestation, that<br />

compensate for the low concentration of pancreatic<br />

lipase. Human milk provides additional<br />

Table 36.2 Revised recommended protein intake and protein/energy ratio for preterm infants<br />

according to post-conceptional age (PCA) and need for catch-up growth (from reference 16)<br />

Without need for With need for<br />

catch-up growth catch-up growth<br />

26–30 weeks’ PCA: 16–18g/kg per day 3.8–4.2g/kg per day 4.4g/kg per day<br />

LBM 14% protein retention PER: ±3.0 PER: ±3.3<br />

30–36 weeks’ PCA: 14–15g/kg per day 3.4–3.6g/kg per day 3.6–4.0g/kg per day<br />

LBM 15% protein retention PER: ±2.8 PER: ±3.0<br />

36–40 weeks’ PCA: 13g/kg per day LBM 2.8–3.2g/kg per day 3.0–3.4g/kg per day<br />

17% protein retention PER: 2.4–2.6 PER: 2.6–2.8<br />

LBM, lean body mass; PER, protein/energy ratio


lipases: lipoprotein lipase, bile salt-stimulated<br />

esterase and non-activated lipase, which improves<br />

intestinal lipolysis. 63<br />

Lipid digestion and absorption are also affected by<br />

fatty acid composition, the triglyceride structure<br />

and the calcium content of the milk. Fatty acid<br />

absorption is related to chain length and degree of<br />

unsaturation. Thus, medium-chain triglycerides<br />

(MCTs) with 8–10 carbon atoms are hydrolyzed<br />

more readily than long-chain triglycerides (LCTs),<br />

and fatty acids with more double bonds are<br />

absorbed more efficiently. In order to improve fat<br />

absorption, commercial formulas contain a significant<br />

quantity of MCTs that are more easily<br />

absorbed than LCTs and transported directly to the<br />

liver via the portal vein as non-esterified fatty<br />

acids. The use of MCTs instead of LCTs in preterm<br />

infant formulas can reduce the formation of<br />

calcium and magnesium soaps with unabsorbed<br />

long-chain saturated fatty acids and, thereby,<br />

increase calcium and magnesium absorption.<br />

However, it needs to be stressed that the energy<br />

content of MCTs corresponds to about 85% of that<br />

of LCTs. In addition, the use of MCTs in formulas<br />

induces an increase in plasma ketones as well as in<br />

urinary excretion of dicarboxylic acids, suggesting<br />

that MCT metabolism could be slightly limited. 64<br />

Moreover, since MCTs do not contain essential<br />

fatty acids, a high MCT intake can reduce the<br />

availability of the essential long-chain fatty acids<br />

necessary for normal growth and development. It<br />

is recommended that the maximum MCT intake in<br />

preterm formula be limited to 40–50% of total fat<br />

content. 17,38<br />

In human milk, long-chain fatty acids are preferentially<br />

positioned in the sn-2 position on the glycerol<br />

and are thus directly absorbed as monoglyceride.<br />

The use of formulas with palmitic acid<br />

with preferential esterification in the sn-2 position<br />

(β-palmitate) induces a minimal positive effect on<br />

energy balance but increases mineral absorption,<br />

leading to a significant reduction of insoluble<br />

calcium soap formation with free palmitic acid<br />

(16:0) and stearic acid (18:0). In preterm infants<br />

the rate of calcium absorption was markedly<br />

increased from 42% in a control formula to 57% in<br />

a formula containing 74% of palmitate esterified in<br />

the triglyceride sn-2 position (as β-palmitate). 65 It<br />

is noteworthy, however, that for commercial<br />

reasons, some preterm infant formulas available<br />

Nutrition during the intermediate and stable growth period 605<br />

today with β-palmitate contain lower proportions<br />

of this structured triglyceride than those evaluated<br />

in previous clinical trials.<br />

A fraction of fat must be provided as essential fatty<br />

acids; these are necessary to ensure optimal fatty<br />

acid composition and function of growing tissue<br />

and for normal eicosanoid synthesis. In preterm<br />

infants linoleic acid intakes of 3.2–12.8% energy<br />

intake have been recommended. 17<br />

Because humans cannot insert double bonds at the<br />

n-3 and n-6 positions, fatty acids with double<br />

bonds in these positions cannot be synthesized<br />

endogenously. Therefore, either specific n-3 and<br />

n-6 fatty acids or the precursor of each series such<br />

as linoleic acid (18:3n-3) and α-linolenic acid<br />

(18:2n-6) must be provided as a component of the<br />

diet. Both linoleic and α-linolenic acid are metabolized<br />

by a series of desaturation and elongation<br />

reactions to more unsaturated longer-chain fatty<br />

acids. Important metabolites of these two fatty<br />

acids include 20:5n-3 eicosapentaenoic acid (EPA),<br />

22:6n-3 docosahexaenoic acid (DHA) and 20:4n-6<br />

arachidonic acid (AA).<br />

The biochemistry and metabolism of the essential<br />

fatty acids is complex, involving differences in<br />

metabolism and competition between the various<br />

n-6 and n-3 fatty acids. The desaturase enzymes<br />

show substrate preference in the order n-3>n-<br />

6>n-9, and are inhibited by products of either the<br />

n-6 or n-3 fatty-acid series. 66,67 Consequently, the<br />

rate of desaturation of linoleic acid (18:2n-6) to<br />

arachidonic acid (20:4n-6) and of α-linolenic acid<br />

(18:3n-3) to docosahexaenoic acid (22:6n-3) depends<br />

on the quantities of linoleic acid (18:2n-6)<br />

and α-linolenic acid (18:3n-3) substrate and<br />

preformed arachidonic acid (20:4n-6, AA) and/or<br />

docosahexaenoic acid (22:6n-3, DHA) in the diet.<br />

Accordingly, a linoleic/α-linolenic acid ratio of 5-<br />

16:1 wt/wt is currently recommended. 17<br />

Eicosapentaenoic acid (20:5n-3, EPA), which is<br />

found predominantly in fish oil, may also inhibit<br />

synthesis of arachidonic acid (20:4n-6). Thus, the<br />

infant’s ability to maintain tissue arachidonic acid<br />

(20:4n-6, AA) levels may be lost if inappropriately<br />

high amounts of eicosapentaenoic acid (20:5n-3,<br />

EPA) or a high ratio of 22:6n-3 and 20:5n-3 to<br />

20:4n-6 are provided by the diet. 68 In addition, the<br />

rate of precursors and long-chain polyunsaturated<br />

fatty acids (PUFAs) oxidation for energy


606<br />

Enteral nutrition in preterm infants<br />

production differs for each fatty acid and may<br />

influence biological activities and their multiple<br />

interactions. 68<br />

Considering that endogenous synthesis of longchain<br />

PUFAs is limited in preterm infants, these<br />

fatty acids were considered as semi-essential or<br />

essential for preterm infants, and numerous<br />

studies evaluated the consequences of early<br />

neonatal deficiencies as well as the need of<br />

formula supplementation. Available sources of<br />

essential fatty acids are vegetal oils, whereas those<br />

of long-chain PUFAs include egg yolk lipid, phospholipid<br />

and triglyceride, fish oils and oils<br />

produced by single-celled organisms (microalgal<br />

and fungal oils).<br />

Several studies have suggested that the long-chain<br />

PUFAs supply in preterm infants has a beneficial<br />

effect on growth, visual and cognitive function, as<br />

well as on the immune system. Although some<br />

methodological limits of these studies do not allow<br />

definitive conclusions to be drawn, most of the<br />

formulas in Europe and the USA are currently<br />

supplemented with long-chain PUFAs. However,<br />

as membranes enriched with PUFAs are particularly<br />

susceptible to oxidative damage, concern<br />

over the use of long-chain PUFAs in infant formulas<br />

might suggest the need for a higher vitamin E<br />

intake.<br />

Several expert groups have recommended the<br />

routine addition of DHA and AA to LBW infant<br />

formulas. 17,38,69 Based on biochemical analyses,<br />

comparison with estimated intrauterine accretion<br />

rates, and the ranges of intakes with human milk<br />

feeding, a consensus group of experts on perinatal<br />

fatty-acid metabolism recently recommended the<br />

supplementation of formulas for preterm infants<br />

with DHA in the range of 0.35–0.5% of the fatty<br />

acids, and with AA in an AA/DHA ratio of 1.2–2<br />

wt/wt. 14,63<br />

Carbohydrates<br />

Carbohydrates represent approximately 40% of the<br />

energy intake of infants ingesting human milk or<br />

infant formulas. By virtue of their important<br />

biological effects, they improve nutritional status<br />

and function of the intestine and colon via the<br />

stimulatory effects of short-chain fatty acids on<br />

cell proliferation and ion absorption, stimulate<br />

insulin secretion and metabolism and growth and<br />

enhance the metabolic response to growth<br />

hormone and calcium absorption. Thus, they are<br />

essential to the overall health of the gastrointestinal<br />

tract and the fulfillment of energy requirements.<br />

The predominant carbohydrate in mammalian<br />

milk and in term infant formula is lactose; after<br />

digestion by lactase, lactose is absorbed as glucose<br />

and galactose, which utilize the same carrier<br />

mechanism. In the human fetus, intestinal lactase<br />

activity is measurable by 10–12 weeks of gestation.<br />

There is a gradual increase in lactase activity with<br />

advancing gestation, although the activity remains<br />

low until about 36 weeks of gestation, when it<br />

reaches the levels seen in full-term neonates. 70<br />

Based on the low lactase activity in early gestation<br />

and the estimated length of the bowel, it was<br />

calculated that a preterm infant weighing<br />

1300–1400g might be expected to have nearly<br />

50–70% of the lactose ingested passing unabsorbed<br />

into the colon 71 where, under the action of<br />

bacterial flora, it is transformed into short fatty<br />

acids and then absorbed. No difference in fecal<br />

carbohydrate loss was found in preterm infants fed<br />

formulas containing either 100% lactose or 50%<br />

lactose and 50% glucose. 72 Lactose not fully<br />

digested in many preterm and term infants could<br />

serve as a source of nutrition for beneficial bacterial<br />

flora in the colon. Through the process of<br />

fermentation, this not only facilitates colonic<br />

water and electrolyte absorption but also stimulates<br />

cell turnover of both the colon and the small<br />

intestine. In premature infants early exposure to<br />

lactose seems to induce intestinal lactase. 73<br />

Regarding the relationship between lactose intake<br />

and calcium absorption, data in human adults<br />

seem to indicate that, in lactose-tolerant subjects,<br />

lactose does not appear to have any beneficial<br />

effect on calcium bioavailability. 74 In lactoseintolerant<br />

subjects, such as preterm infants, there<br />

is a significant level of uncertainty. Whether<br />

lactose in the distal bowel increases or impairs<br />

calcium absorption in preterm infants has yet to be<br />

defined. 17<br />

Quantitatively, other than lactose, oligosaccharides<br />

are the largest carbohydrate component in<br />

mature human milk and represent the third largest<br />

solute load after lactose and fat. 75,76 The


concentration of oligosaccharides in human milk<br />

has been observed to change with the duration of<br />

lactation, being highest in the colostrum (about<br />

20–23g/l), about 20g/l on day 4 and about 13g/l on<br />

day 120 of lactation. 77 Many of the protective<br />

effects of human milk against diarrheal diseases,<br />

respiratory and ear infections, documented from a<br />

number of clinical epidemiological studies in<br />

developed and developing countries have been<br />

attributed to oligosaccharides.<br />

In preterm formula a significant part of the carbohydrate<br />

is given as glucose polymers. These sugars<br />

have the advantage of providing a higher caloric<br />

intake without a corresponding rise in osmolarity.<br />

Glucose polymers are pure carbohydrates prepared<br />

by controlled acid/enzyme hydrolysis of cornstarch.<br />

They consist of polymers of glucose of<br />

varying chain length, although the majority are of<br />

medium (6–10 glucose units) chain length with a<br />

small amount (usually


608<br />

Enteral nutrition in preterm infants<br />

Table 36.3 Mineral balance in very-low-birth-weight infants fed human milk or preterm formulas<br />

Calcium Calcium Calcium Phosphorus Phosphorus Phosphorus<br />

intake absorption retention intake absorption retention<br />

(mg/kg per (%) (mg/kg per (mg/kg per (%) (mg/kg per<br />

Diet Reference day) day) day) day)<br />

HM (n=10) 80 58±11 71±14 25±12 24±6 92±4 21±5<br />

HM + Ca+P (n=8) 80 90±11 73±13 63±12 62±5 93±2 53±4<br />

HM + HMF (n=7) 80 101±19 66±7 65±14 78±13 94±2 62±9<br />

HM + HMF2 (n=9) * 170±13 57±17 91±27 87±6 94±2 61±12<br />

PTF1 (n=39) 32 91±14 51±17 45±18 63±9 89±6 42±8<br />

PTF2 (n=12) * 103±16 43±18 41±22 64±8 92±3 40±7<br />

PTF3 (n=7) 80 118±10 65±14 68±11 72±3 92±2 60±6<br />

PTF4 (n=19) 32 120±21 44±16 48±22 70±9 78±12 43±11<br />

PTF5(n=8) * 120±19 46±18 50±20 75±12 88±6 45±10<br />

PTF6 (n=7) 83 135±4 46±13 59±17 88±3 91±5 53±7<br />

PTF7 (n=7) * 161±11 41±13 58±22 90±5 59±8 50±8<br />

PTF8 (n=6) 84 162±5 49±6 78±11 78±2 92±2 49±3<br />

PTF9 (n=6) 84 162±5 64±5 100±8 75±2 92±2 58±3<br />

PTF10 (n=9) 83 164±13 55±13 85±18 93±4 90±4 63±8<br />

PTF11 (n=8) * 172±11 34±12 54±25 95±6 60±6 51±10<br />

PTF12 (n=11) 85 225±6 36±6 76±13 120±4 64±5 64±5<br />

HM, human milk; HMF, human milk fortifier; PTF, preterm formula<br />

* Unpublished data<br />

calcium absorption is usually lower than with<br />

powder formulas. With the use of formulas with a<br />

well-absorbed fat blend of about 85%, the formation<br />

of a calcium soap is of minimal interest in<br />

clinical practice. Owing to the poor solubility of<br />

calcium salts, especially calcium phosphate, the<br />

calcium content measured in the formula could be<br />

significantly lower than the claimed value, and<br />

additional loss due to precipitation may occur<br />

before feeding. Therefore, the actual amount of<br />

calcium provided by feeding needs to be chemically<br />

measured in each infant during metabolic<br />

balance studies. As shown in Table 34.3, a calcium<br />

retention close to 90mg/kg per day could currently<br />

be expected in preterm infants fed preterm<br />

formula with a highly soluble calcium content.<br />

Nevertheless, these values are still lower than the<br />

values estimated during the last trimester of gesta-<br />

tion (100–120mg/kg per day) and still considered<br />

as the target mineral accretion for VLBW infants.<br />

Accordingly, it could be recommended that the<br />

calcium intake in preterm infants range between<br />

130 and 170mg/kg per day with a calcium/phosphorus<br />

ratio close to 1.8, as long as the calcium<br />

absorption rate is 45–60%.<br />

Iron<br />

Preterm infants require iron for erythropoiesis, brain<br />

development, muscle function and cardiac function.<br />

The symptoms of iron deficiency are due to tissuelevel<br />

losses of iron containing enzymes and<br />

iron–sulfur proteins, not just to anemia. Oski has<br />

estimated that, in the absence of iron supplementation<br />

and blood loss, the VLBW preterm infant has<br />

enough iron stores to last 2 months and becomes<br />

anemic before liver stores are completely depleted. 86


Table 36.4 Recommended enteral micromineral<br />

intakes (µg/kg per day) for preterm<br />

infants (from reference 87)<br />

Transitional<br />

period Stable/post-<br />

(0–14 days) discharge periods<br />

Iron 0 2000–4000<br />

Zinc 500–800 1000–2000*<br />

Copper 120 120–150<br />

Selenium 1.3 1.3–4.5<br />

Chromium 0.5 0.1–2.25<br />

Molybdenum 0.3 0.3–4.0<br />

Manganese 0.75 0.75–7.5<br />

Iodine 11–27 10–60<br />

*Post-discharge supplement of 0.5mg/kg per day for<br />

infants fed human milk<br />

Preterm infants need supplemental iron after 2<br />

weeks of age. The enteral route appears safest. Iron<br />

can be supplied by preterm formula, human milk<br />

fortifier or medicinal iron drops. The enteral dose<br />

for the preterm infant not receiving erythropoietin<br />

ranges from 2 to 4mg/kg per day depending on the<br />

degree of prematurity and the amount of uncompensated<br />

phlebotomy. The recent introduction of<br />

erythropoietin puts a greater stress on iron<br />

balance, forcing the infant to mobilize endogenous<br />

iron stores at a faster rate. For infants receiving this<br />

medication, an enteral dose of 6mg/kg per day is<br />

recommended. No study has demonstrated oxidative<br />

toxicity from enteral iron given at conventional<br />

doses in preterm infants.<br />

Studies are currently addressing the need for iron<br />

after discharge in preterm infants. Their iron needs<br />

will probably remain greater than those of term<br />

infants because of their more rapid relative rate of<br />

growth and therefore blood volume expansion.<br />

Given their lower endogenous iron stores, it would<br />

be prudent to screen these infants earlier (e.g. 2<br />

months post-discharge) than term infants.<br />

Other trace elements<br />

Trace elements contribute less than 0.01% of total<br />

body weight. They function as constituents of<br />

Nutrition during the intermediate and stable growth period 609<br />

metalloenzymes, cofactors for metal ion-activated<br />

enzymes, or components of vitamins, hormones<br />

and proteins. Because the fetus accumulates stores<br />

of trace elements, especially during the third<br />

trimester of pregnancy, premature infants have<br />

low stores at birth and are at risk of developing<br />

mineral deficiencies if intakes are inadequate for<br />

their growth requirements.<br />

Trace minerals with established physiological<br />

importance in humans include zinc, copper, selenium,<br />

manganese, chromium, molybdenum, fluoride<br />

and iodine. The recommended oral intakes for<br />

infants are listed in Table 36.4. 87 Potentially toxic<br />

trace minerals for pediatric patients are lead and<br />

aluminum.<br />

Oral vitamin requirements<br />

Vitamins are organic compounds that are essential<br />

for metabolic reactions but are not synthesized by<br />

the body. Vitamins are classified as water soluble<br />

or fat soluble, based on the biochemical structure<br />

and function of the compound. The recommended<br />

oral intakes of vitamins for infants are shown in<br />

Table 36.5. 11<br />

Water-soluble vitamins that cannot be formed by<br />

precursors (with the exception of niacin from tryptophan)<br />

and do not accumulate in the body (with<br />

the exception of vitamin B12) include B complex<br />

vitamins and vitamin C. They serve as prosthetic<br />

groups for enzymes involved in amino acid metabolism,<br />

energy production and nucleic acid synthesis.<br />

A daily intake is required to prevent deficiency.<br />

Excretion occurs in the urine and bile.<br />

Altered urinary losses due to renal immaturity<br />

during the first week of life predispose a preterm<br />

infant to vitamin deficiency or excess.<br />

Fat-soluble vitamins include vitamin A, D, E and<br />

K. These vitamins function physiologically in the<br />

conformation and function of complex molecules<br />

and membranes and are important for the development<br />

and function of highly specialized tissues.<br />

They can be built from precursors, are excreted<br />

with difficulty and accumulate in the body, and<br />

therefore they can produce toxicity. They are not<br />

required daily and deficient states develop slowly.<br />

Fat-soluble vitamins require carrier systems,<br />

usually lipoproteins, for solubility in blood, and<br />

intestinal absorption depends on fat absorption.


610<br />

Enteral nutrition in preterm infants<br />

Table 36.5 Recommended oral intake of vitamins for preterm infants (from reference<br />

11, American Academy of Pediatrics, Committee on Nutrition, 1998)<br />

AAPCON Consensus<br />

Recommendations Recommendations<br />

Vitamin (amount for premature infants for infants with<br />

per 100 kcal) (1998) VLBW (1993)<br />

Fat soluble<br />

Vitamin A (IU) 75–225 583–1250<br />

Vitamin D (IU) 270 125–333<br />

Vitamin E (IU) >1.1 5–10<br />

Vitamin K (µg) 4 6.66–8.33<br />

Water soluble<br />

Vitamin B6 (µg) >35 125–175<br />

Vitamin B12 (µg) >0.35 0.25<br />

Vitamin C (mg) 35 15–20<br />

Biotin (µg) >1.5 3–5<br />

Folic acid (µg) 33 21–42<br />

Niacin (mg) >0.25 3–4<br />

Pantothenate (mg) 0.3 1–1.5<br />

Riboflavin (µg) >60 200–300<br />

Thiamine (µg) >40 150–200<br />

AAPCON, American Academy of Pediatrics Committee on Nutrition; VLBW, very low birth weight<br />

Practical aspects of enteral nutrition<br />

Breast milk<br />

Benefit of human milk<br />

The American Academy of Pediatrics has acknowledged<br />

the advantages of human milk feeding with<br />

the statement that it is the preferred feeding for all<br />

infants, including those born preterm. 88 The benefits<br />

of human milk for gastrointestinal function,<br />

host defense and neurodevelopmental outcome are<br />

well known. The amino acid composition of<br />

human milk is suitable for the nutritional requirements<br />

of preterm infants who are, because of<br />

incomplete development of some amino acid<br />

metabolic pathways, at risk of developing a deficit<br />

or an overload of essential and semi-essential<br />

amino acids.<br />

Clinical studies performed in various parts of the<br />

world have shown that preterm infants fed human<br />

milk have fewer infections than those fed on<br />

formula. 89 This effect has been observed with both<br />

fresh and pasteurised human milk. There are<br />

studies showing a protective effect of breast milk<br />

on the incidence of NEC. A meta-analysis of<br />

randomized controlled trials was performed to<br />

determine whether enteral feeding with donor<br />

human milk, compared with formula, reduced the<br />

incidence of NEC in preterm infants. They found<br />

that feeding with donor human milk was also associated<br />

with a significantly reduced risk of NEC;<br />

infants who received donor human milk were<br />

three times less likely to develop NEC and four<br />

times less likely to have confirmed NEC. 90<br />

The protective effect of human milk has been<br />

attributed to several factors, such as macrophages,<br />

lymphocytes, sIgA, lysozyme, lactoferrin, oligosaccharides,<br />

nucleotides, cytokines, growth factors<br />

and enzymes. Immune defense can be provided by<br />

an interaction between these factors. Human milk<br />

can play a protective role through the enteromammary<br />

immune system. The mother’s presence in


the neonatal nursery, particularly the premature<br />

infant’s skin-to-skin contact with the mother, may<br />

induce the production of specific antibodies<br />

against the nosocomial pathogens present in the<br />

nursery. 88 Breast milk enhances the growth, motility<br />

and maturity of the gastrointestinal tract;<br />

compared to formulas it induces faster gastric<br />

emptying. 91<br />

Preterm infants with a birth weight below 1850g,<br />

fed human milk during hospitalization, showed<br />

better neurological development. At 7.5–8 years<br />

they showed a higher intelligence quotient<br />

compared to those fed formula, even after controlling<br />

for the mother’s education and social class. 92<br />

Higher mental developmental index scores were<br />

reported in VLBW (


612<br />

Enteral nutrition in preterm infants<br />

Table 36.6 Composition of various human milk fortifiers (expressed per gram of protein)<br />

BMF EHMF1 SHMF WHMF EHMF2 EHMF3<br />

Eoprotin FM-85 (Nutricia) (M-J) (Abbott) (Wyeth) (M-J) (M-J)<br />

(Milupa) (Nestlé) bag bag bag bag bag bag<br />

powder powder 1.5g 0.9g 0.9g 2g 0.8g 0.8g<br />

Protein (g) 1 1 1 1 1 1 1 1<br />

Fat (g) 0 0 0 0 0.36 0 0.58 0.91<br />

Carbohydrate (g) 3.48 4.3 2.86 3.9* 1.8 2.0 1.0 0.2<br />

Sodium (mg) 3.3 32.3 8.7 10.3 15.0 15.0 1.0 15.0<br />

Potassium (mg) 4.0 13.8 5.7 23.0 62.9 20.0 18.2 26.0<br />

Calcium (mg) 62 61 87 136 117 80 82 82<br />

Magnesium (mg) 3.5 2.4 8.6 1.5 7.0 2.0 0.9 0.9<br />

Phosphorus (mg) 42.3 40.7 58.4 66.8 67.2 40.0 41.0 45.0<br />

Chloride (mg) 24.9 22.2 10.1 26.0 37.9 14.0 8.2 12.0<br />

Energy (kcal) 31.7 21.4 14.3 20.7 14.0 13.0 12.7 12.7<br />

Osmolality (mOsm) 90 96 85 177 90 136 59—<br />

* Including the carbohydrate content of calcium glycerophosphate<br />

titatively. In general, they contain bovine whey<br />

protein (intact or hydrolyzed), carbohydrates<br />

(mainly or exclusively glucose polymers or<br />

maltodextrins), minerals and electrolytes such as<br />

sodium, calcium, phosphorus and magnesium and<br />

some also contain micronutrients (zinc and<br />

copper) and vitamins.<br />

The properties of human milk may be changed by<br />

nutrient fortification. For example, the addition of<br />

human milk fortifiers induces a rapid and clinically<br />

significant increase in osmolality, frequently<br />

above 400mOsm/kg H2O, a value at least 40%<br />

higher than that of human milk. 99 This is the result<br />

of the osmotic content of the fortifier but also of<br />

the rapid and continuous activity of human milk<br />

amylase on the dextrin content of fortifiers. Such<br />

an increase in osmolality may at least partly<br />

explain some minimal clinical disturbances, such<br />

as abdominal discomfort and delayed gastric<br />

emptying, but in some cases it might increase the<br />

risk of NEC in preterm infants. 100 Recently, in<br />

order to avoid these problems, fortifiers with<br />

whole proteins, reduced carbohydrate content and<br />

fat supplementation have been proposed. 101 The<br />

presence of fat in fortifiers has the advantage of<br />

increasing the energy intake of human milk<br />

without increasing osmolality values. However,<br />

this needs further technological studies, because<br />

the action of human milk lipase on triglycerides of<br />

fortifiers could produce instability in human milk.<br />

Many studies have evaluated growth, metabolic<br />

balance and weight gain composition in preterm<br />

infants fed fortified human milk. 49,98,102–108 The<br />

addition of multinutrient fortifiers to human milk<br />

resulted in short-term improvements in weight<br />

gain and increments in both length and head<br />

circumference growth during hospital stay. For<br />

ethical reasons, it is now unlikely that further<br />

studies evaluating fortification of human milk in<br />

comparison with no fortification will be<br />

conducted. Although the fortification of human<br />

milk improves the growth of preterm infants,<br />

growth both in general and referred to specific<br />

parameters (such as lean body mass, fat mass and<br />

bone mineral content) is significantly lower in<br />

fortified human milk-fed infants than in those fed<br />

preterm formula (Table 36.7). These differences<br />

could be related to the lower protein content but<br />

also to the reduction in metabolizable protein and<br />

energy available for new tissues synthesis.<br />

Questions have been raised as to whether the addition<br />

of bovine-derived human milk fortifiers can


affect feeding tolerance in premature infants. No<br />

differences in feeding tolerance were reported in a<br />

meta-analysis comparing premature infants fed<br />

fortified human milk or unfortified human milk. 109<br />

In contrast, fortifiers have recently been considered<br />

as risk factors for NEC in VLBW infants. 100<br />

When compared with premature infants fed<br />

preterm formula, infants fed exclusively fortified<br />

human milk had a significantly lower incidence of<br />

NEC and/or sepsis, had fewer positive blood<br />

cultures and required less antibiotic administration.<br />

89<br />

The effect of nutrient fortification on some of the<br />

general host defense properties of milk have been<br />

evaluated. Fortification did not affect the concentration<br />

of IgA in milk. 110 When fortified human<br />

milk was evaluated under simulated nursery<br />

conditions, bacterial colony counts were not<br />

significantly different after 20h of storage at refrigerator<br />

temperature, but increased from 20 to 24h<br />

when maintained at incubator temperature. 110<br />

Early use of a human milk fortifier up to 1.3g of<br />

protein/100ml is recommended for more immature<br />

or smaller preterm infants, beginning from the<br />

time when they are able to tolerate 50–70ml/kg per<br />

day of milk. Preference should be given to fortifiers<br />

with higher protein density and lower osmotic<br />

load. Additional studies are required to define the<br />

Practical aspects of enteral nutrition 613<br />

Table 36.7 Nutritional intake, growth and weight gain in preterm infants fed fortified human milk<br />

(HMF) and preterm formula (PTF) (from reference 16)<br />

Amount (/kg per day) HMF (n=48) PTF (n=86) p Value<br />

Volume intake (ml) 164±12 152±12


614<br />

Enteral nutrition in preterm infants<br />

Nevertheless, according to more recent data, a<br />

general profile in macronutrients could probably<br />

be suggested. Energy content could be slightly<br />

higher than at present (80–90kcal/100ml in order<br />

to provide 130–135kcal/kg per day with a volume<br />

intake close to 160ml/kg per day). According to the<br />

protein requirements, the protein content would<br />

represent 3.3g/100kcal, i.e. 2.6–3.0g/100ml. Wheypredominant<br />

protein with reduced glycomacropeptide<br />

and α-lactalbumin enrichment<br />

could be used to optimize the amino acid profile.<br />

Up to now the use of protein-hydrolyzed formulas<br />

have not been recommended. Their amino acid<br />

pattern is relatively unbalanced and the net<br />

protein utilization and protein efficiency are relatively<br />

impaired in comparison to whole protein, in<br />

contrast to the minimal clinical advantages<br />

observed, such as an improvement in gastric<br />

emptying or a reduced transit time. 32,111 The partition<br />

of the non-protein energy supply would favor<br />

the carbohydrate content up to 50–60% in view of<br />

its suggested benefits in sparing protein oxidation,<br />

enhancing growth and protein accretion as well as<br />

improving quiet sleep and influencing the distribution<br />

of behavioral activity states in VLBW infants.<br />

According to the relatively reduced intestinal<br />

lactase activity, the lactose content could be relatively<br />

reduced and substituted with glucose polymers<br />

with the characteristic of maintaining the<br />

low osmolality of the formulas. The remaining<br />

energy supply would be covered by fat with a fat<br />

blend carefully designed to reduce the long-chain<br />

saturated fatty acid content, and provide the essential<br />

fatty acids (linoleic and α-linolenic acid) in an<br />

appropriate ratio as well as the long-chain PUFA<br />

such as DHA and AA. In order to improve fat<br />

absorption, an important quota of fat could be<br />

given as MCT with a maximum of 30–40%. Highly<br />

metabolizable (50–60% absorption rate) calcium<br />

content would be limited to 100–120mg/100ml. A<br />

higher calcium supply does not seem useful.<br />

According to the expected nitrogen and calcium<br />

retention, the phosphorus content of this formula<br />

would represent 55–65mg/100ml, considering a<br />

phosphorus absorption close to 90%.<br />

Additional components of human milk, such as<br />

nucleotides, polyamine prebiotics and glutamine,<br />

are currently under investigation, and the results<br />

need to be available before their use in preterm<br />

formulas. 17<br />

Preterm formulas are available as powder or liquid<br />

in glass bottles or cans. Liquid formulas have the<br />

advantage that they provide sterile feeding thereby<br />

reducing possible infections reported in pre-term<br />

infants fed powder formulas such as the<br />

Enterobacter sakazakii. 112 Nevertheless, it worth<br />

remembering that with liquid formulas, there is<br />

frequent discordance between claimed and available<br />

content of several nutrients. For instance, part<br />

of the calcium content of liquid formula may<br />

precipitate on the wall of the container reducing<br />

the calcium truly available to the preterm infants.<br />

In addition, the heat treatment necessary to sterilize<br />

the liquid formulas reduces the bioavailability<br />

of various nutrients such as proteins, calcium or<br />

copper. The impaired bioavailability is directly<br />

related to the importance of heat treatment and is<br />

probably lower in the ultra heat treated (UHT)<br />

brickpack formula. In summary, powder formulas<br />

allowing adaptation of nutrient density and being<br />

of higher nutritional value, remain the formulas of<br />

choice for the feeding of VLBW infants when<br />

human milk is not available.<br />

Post-discharge nutrition in preterm infants<br />

Protein- and mineral-enriched formulas have been<br />

proposed to feed preterm infants after discharge<br />

with the aim of minimizing as much as possible,<br />

postnatal growth restriction during the early weeks<br />

of life, inducing early catch-up growth and reducing<br />

the adult adverse effects of early malnutrition.<br />

In most neonatal units, the discharge of VLBW<br />

infants usually occurs usually when premature<br />

infants reach a corrected gestational age of 35–36<br />

weeks and a weight of about 1800–2100g. By that<br />

time, they have accumulated energy, protein and<br />

mineral deficits and still present higher nutrient<br />

requirements than healthy infants appropriate for<br />

gestational term. The potential benefits of<br />

enriched formula on growth have been investigated<br />

following the first study of Lucas et al in<br />

1992 who introduced the concept of the postdischarge<br />

formula. 113<br />

Although the various studies differed in design<br />

and reported conflicting results, they allow a<br />

global analysis to be made. Comments follow:<br />

(1) Among the VLBW infants, not all preterm<br />

infants are at similar risk of later growth


failure. Small-for-gestational-age infants<br />

and those appropriate for gestational age<br />

developing postnatal growth restriction<br />

seem to be the population at higher risk,<br />

while infants appropriate for corrected age<br />

at the time of discharge from the hospital<br />

subsequently continue to maintain appropriate<br />

growth. 114<br />

(2) Energy intakes were the main determinant<br />

of milk volume intakes. Compared to regular<br />

(standard) formula, an increase in energy<br />

density had no influence on energy supply,<br />

in contrast to the increase in protein density<br />

that influences protein intake, and protein<br />

metabolism.<br />

(3) A positive effect of growth parameters was<br />

not observed in all studies, but the benefit of<br />

the nutrient-enriched formula on growth,<br />

when observed, was mainly seen during the<br />

early post-discharge period (between discharge<br />

and term), in infants with very low<br />

birth weights. 115,116 In addition, the growth<br />

benefit could be related to the preterm<br />

infants without a postnatal growth deficit at<br />

the time of discharge. 117<br />

(4) There is a strong gender influence on the<br />

result of diet manipulation during the postdischarge<br />

period. The main positive results<br />

are limited to boys.<br />

(5) The safety of prolonged high-protein intake<br />

during the early period of life in preterm<br />

infants after discharge has not been established,<br />

in contrast to the minimal benefits<br />

suggested by the various studies. Highprotein<br />

intake, by inducing an increase in<br />

biochemical parameters of nitrogen metabolism<br />

(urea, amino acids, ammonia), could<br />

enhance the development of several diseases,<br />

such as obesity and hypertension<br />

later in life. 118<br />

Since early nutrition should not be considered<br />

simply in terms of providing immediate nutritional<br />

needs but also for the biological effects with<br />

Practical aspects of enteral nutrition 615<br />

possibly lasting or lifelong significance, it seems<br />

prudent to suggest an enriched formula only in<br />

infants at risk of future growth failure. 119<br />

Mothers of infants at low risk of longitudinal<br />

growth restriction should be encouraged to feed<br />

them their own milk. Careful attention is necessary<br />

to evaluate the appropriateness of intakes<br />

and growth just before and in the first week after<br />

discharge. In early discharge a supplementation<br />

with fortified human milk or formula might be<br />

necessary in order to improve the nutrient<br />

supply in relation to the remaining state of<br />

prematurity.<br />

In infants with a low risk of longitudinal growth<br />

restriction, fed on formula at the time of discharge,<br />

a regular formula with relatively low protein<br />

density (2.2g/100kcal) should be provided with<br />

particular attention to its long-chain PUFAs,<br />

mineral and trace element content.<br />

Infants at high risk of longitudinal growth restriction,<br />

should be breast fed or should receive human<br />

milk supplemented with fortified human milk or<br />

formula, in order to improve the nutrient supply<br />

and promote catch-up growth. An evaluation of<br />

nutrient intakes and growth should be done<br />

frequently to optimize growth and biological parameters.<br />

In infants at high risk of longitudinal growth<br />

restriction, on formula at the time of discharge, a<br />

specific post-discharge formula with higher<br />

protein and mineral density but also adapted in<br />

long-chain PUFAs and trace elements should be<br />

provided. Intakes and growth would also be<br />

recorded weekly and thereafter monthly to ensure<br />

that nutrient supplementation is in agreement<br />

with growth and biological results.<br />

Further research should be initiated in order to<br />

determine the specific nutritional needs of growthrestricted<br />

preterm infants in order to design more<br />

specific post-discharge formulas and evaluate their<br />

effect on the long-term incidence of growth restriction<br />

as well as somatic and neurodevelopmental<br />

outcomes.


616<br />

Enteral nutrition in preterm infants<br />

REFERENCES<br />

1. Ziegler EE, Thureen PJ, Carlson SJ. Aggressive nutrition<br />

of the very low birthweight infant. Clin Perinatol 2002;<br />

29: 225–244.<br />

2. Lucas A, Bloom SR, Aynsley-Green A. Gut hormones<br />

and ‘minimal enteral feeding’. Acta Paediatr Scand<br />

1986; 75: 719–723.<br />

3. Tyson JE, Kennedy K. Minimal enteral nutrition in<br />

parenterally fed neonates. 1997.<br />

http://www.nichd.nih.gov/cochraneonatal/tyson//tyson.htm.<br />

4. Premji S, Chessel L. Continuous nasogastric milk<br />

feeding versus intermittent bolus milk feeding for<br />

premature infants less than 1500 grams, Cochrane<br />

Database Syst Rev 2003; 1: CD001819.<br />

5. Currao WJ, Cox C, Shapiro DL. Diluted formula for<br />

beginning the feeding of premature infants. Am J Dis<br />

Child 1988; 142: 730–731.<br />

6. Kennedy KA, Tyson JE, Chamnanvanakij S. Rapid<br />

versus slow rate of advancement of feedings for promoting<br />

growth and preventing necrotizing enterocolitis in<br />

parenterally fed low birthweight infants. Cochrane<br />

Database Syst Rev 2000; 2: CD001241.<br />

7. Berseth CL, Bisquera JA, Paje VU. Prolonging small<br />

feeding volumes early in life decreases the incidence of<br />

necrotizing enterocolitis in very low birth weight<br />

infants. Pediatrics 2003; 111: 529–534.<br />

8. Davies ID, Avner ED. Fluid and electrolyte management.<br />

In Fanaroff AA, Martin RJ, eds. <strong>Neonatal</strong>–Perinatal<br />

Medicine. 7th edn. St Louis, MO: Mosby, 2002: 619–627.<br />

9. Fomon SJ, Ziegler EE. Renal solute load and potential<br />

solute load in infancy. J Pediatr 1999; 134: 11–14.<br />

10. Fomon SJ, Ziegler EE. Renal solute load. In Nutrition of<br />

Normal Infants. St. Louis, MO: Mosby-Year Book, 1993:<br />

91–102.<br />

11. American Academy of Pediatrics, Committee on<br />

Nutrition. Nutritional needs of preterm infants. In<br />

Kleinman RE, ed. Pediatric Nutrition Handbook, 4th<br />

edn. Elk Grove Village, Il: American Academy of<br />

Pediatrics, 1998: 55–87.<br />

12. Raiten DJ, Talbot JM, Waters JH. Assessment of nutrient<br />

requirements for infant formulas. Prepared by the Life<br />

Sciences Research Office, 9650 Rockville Pike, Bethesda<br />

MD. J Nutr 1998; 128 (Suppl 11S).<br />

13. Parmar V, Abdulrazzaq YM, Brooke OG. Stool water in<br />

preterm neonates. Arch Dis Child 1986; 61: 1136–1137.<br />

14. Chevalier RL. Developmental renal physiology of the low<br />

birth weight pre-term newborn. J Urol 1996; 156: 714–719.<br />

15. De Curtis M, Senterre J, Rigo J. Renal solute load in<br />

preterm infants. Arch Dis Child 1990; 65: 357–360.<br />

16. Rigo J. Protein, amino acid and other nitrogen<br />

compounds. In Tsang RC, Uauy R, Koletzko et al., eds.<br />

Nutritional Needs of the Preterm Infant. Cincinnati, OH:<br />

Digital Educational Publishing, 2004: 43–76 (in press).<br />

17. Klein CJ. Nutrient requirements for preterm infant<br />

formulas. J Nutr 2002; 132: 1395S–1577S.<br />

18. Sparks JW. Human intrauterine growth and nutrient<br />

accretion. Semin Perinatol 1984; 8: 74–93.<br />

19. Nutrition Committee CPS. Nutrient needs and feeding of<br />

premature infants. Can Med Assoc J 1995; 152: 1765–1785.<br />

20. Zello GA, Menendez CE, Rafii M et al. Minimum protein<br />

intake for the preterm neonate determined by protein<br />

and amino acid kinetics Pediatric Res 2003; 53: 338–344.<br />

21. Embleton NE, Pang N, Cooke RJ. Postnatal malnutrition<br />

and growth retardation: an inevitable consequence of<br />

current recommendations in preterm infants? Pediatrics<br />

2001; 107: 270–273.<br />

22. Rigo J, Putet G, Picaud JC et al Nitrogen balance and<br />

plasma amino acids in the evaluation of protein sources<br />

for extremely low birthweight infants. In Ziegler EE,<br />

Lucas A, Moro GE, eds. Nutrition of the Very Low<br />

Birthweight Infant. 43rd Nestlè Nutrition Workshop,<br />

Nestec Ltd. Philadelphia: Vevey/Lippincott Williams &<br />

Wilkins, 1999: 139–153.<br />

23. Rudloff S, Lonnerdal B. Solubility and digestibility of<br />

milk proteins in infant formula exposed to different heat<br />

treatments. J Pediatr Gastroenterol Nutr 1992; 15: 25–33.<br />

24. Cooke R, Watson D, Werkman S et al. Effects of type of<br />

dietary protein on acid–base status, protein nutritional<br />

status, plasma levels of amino acids, and nutrient<br />

balance in the very low birth weight infant. J Pediatr<br />

1992; 121: 444–451.<br />

25. Rigo J, Senterre J. Optimal threonine intake for preterm<br />

infants fed on oral or parenteral nutrition. J Parenter<br />

Enteral Nutr 1980; 4: 15–17.<br />

26. Boehm G, Cervantes H, Georgi G et al. Effect of increasing<br />

dietary threonine intakes on amino acid metabolism<br />

of central nervous system and peripheral tissues in<br />

growing rats. Pediatr Res 1998; 44: 900–906.<br />

27. Rigo J, Boehm G, Georgi G et al. An infant formula free<br />

of glycomacropeptide prevents hyperthreoninemia in<br />

formula-fed preterm infants. J Pediatr Gastroenterol Nutr<br />

2001; 32: 127–130.<br />

28. Picaud JC, Rigo J, Normand S et al. Nutritional efficacy<br />

of preterm formula with a partially hydrolyzed protein<br />

source: a randomized pilot study. J Pediatr Gastroenterol<br />

Nutr 2001; 32: 555–561.<br />

29. Raïhïa NCR, Nesci AF, Cajozzo C et al. Protein quantity<br />

and quality in infant formula: closer to the reference. In<br />

Raïhä NCR, Rubaltelli FF, eds. Infant Formula: Closer to<br />

the Reference. Nestlé Nutrition Workshop Series, Nestec<br />

Ltd. Philadelphia: Vevey/Lippincott Williams & Wilkins,<br />

2002; 47: 53–69.<br />

30. Heine WE, Klein PD, Reeds PJ. The importance of alphalactoalbumin<br />

in infant nutrition. J Nutr 1991; 121: 277–283.<br />

31. Heine WE, Radke M, Wutzke KD et al. Alfa-lactalbuminenriched<br />

low-protein infant formulas: a comparison to<br />

breast milk feeding. Acta Paediatr 1996; 85: 1024–1028.<br />

32. Rigo J, Salle BL, Picaud J-C et al. Nutritional evaluation<br />

of protein hydrolysate formulas. Eur J Clin Nutr 1995;<br />

49: S26–S38.<br />

33. Mihatsh WA, Hogel J, Pohlandt F. Hydrolysed protein<br />

accelerates the gastrointestinal transport of formula in<br />

preterm infants. Acta Paediatr 2001; 90: 196–198.<br />

34. Lee YH. Food-processing approaches to altering allergenic<br />

potential of milk-based formula. J Pediatr 1992;<br />

121: S47–S50.<br />

35. Picaud JC, Rigo J, Normand S et al. Nutritional efficacy<br />

of preterm formula with a partially hydrolyzed protein<br />

source: a randomized pilot study. J Pediatr Gastroenterol<br />

Nutr 2001; 32: 555–561.<br />

36. Lajoie N, Gauthier SF, Pouliot Y. Improved storage<br />

stability of model infant formula by whey peptides fractions.<br />

J Agric Food Chem 2001; 49: 1999–2007.<br />

37. Mihatsh WA, Pohlandt F. Protein hydrolysate formula<br />

maintains homeostasis of plasma amino acids in preterm<br />

infants. J Pediatr Gastroenterol Nutr 1999; 29: 406–410.<br />

38. ESPGAN Committee on Nutrition. Comment on the<br />

content and composition of lipids in infant formulas.<br />

Acta Paediatr Scand 1991; 80: 887–896.<br />

39. Health Canada Health Protection Branch of Canada,<br />

Guidelines for the Composition and Clinical Testing of<br />

Formulas for Preterm Infants. Report of an Ad Hoc<br />

Expert Consultation to the Health Protection Branch,<br />

Health Canada. Ottawa, Ontario: Canadian Government<br />

Publishing Center, 1995.<br />

40. Bell EF. Diet and body composition of preterm infants.<br />

Acta Paediatr 1994; 405 (Suppl): 25–28.


41. Kashyap S, Heird WC. Protein requirements of low birth<br />

weight, very low birth weight and small for gestational<br />

age infants. In Raiha NCR ed. Protein Metabolism During<br />

Infancy. 33rd edn. Nestlé Nutrition Workshop Series,<br />

vol 33. New York: Raven Press, 1994: 133–155.<br />

42. Putet G. Energy. In Tsang RC, Lucas A, Uauy R, Zlotkin<br />

S, eds. Nutritional Needs of the Preterm Infant. New<br />

York: Williams & Wilkins, 1993: 15–28.<br />

43. Micheli JL, Schutz T. Protein. In Tsang RC, Lucas A,<br />

Uauy R, Zlotkin S, eds. Nutritional Needs of the Preterm<br />

Infant: Scientific Basis and Practical Guidelines.<br />

Baltimore, MD: Williams and Wilkins, 1993: 29–46.<br />

44. Kashyap S, Towers HM, Sahni R et al. Effects of quality<br />

of energy on substrate oxidation in enterally fed, lowbirth-weight<br />

infants. Am J Clin Nutr 2001; 74: 374–380.<br />

45. Pencharz PB, Farri L, Papageorgiou A. The effect of human<br />

milk and low-protein formulae on the rates of total body<br />

protein turnover and urinary 3-methyl-histidine excretion<br />

of preterm infants. Clin Sci 1983; 64: 611–616.<br />

46. Putet G, Picaud JC, Salle BL et al. Utilization and<br />

storage of energy. In Salle BL, Swyer PR, eds. Nutrition<br />

of the Low-Birthweight Infant. Nestlé Nutrition<br />

Workshop Series. Nestec Ltd., New York: Vevey/Raven<br />

Press, 1993; 32: 61–69.<br />

47. Whyte RK, Haslam R, Vlainic C et al. Energy balance and<br />

nitrogen balance in growing low-birthweight infants fed<br />

human milk or formula. Pediatr Res 1983; 17: 891–898.<br />

48. Kashyap S, Forsyth M, Zucker C et al. Effect of varying<br />

protein and energy intakes on growth and metabolic<br />

response in low birth weight infants. J Pediatr 1986;<br />

108: 955–963.<br />

49. Kashyap S, Schulze KF, Forsyth M et al. Growth, nutrient<br />

retention and metabolic response of low-birthweight<br />

infants fed supplemented and unsupplemented preterm<br />

human milk. Am J Clin Nutr 1990; 52: 254–262.<br />

50. Roberts SB, Lucas A. Energetic efficiency and nutrient<br />

accretion in preterm infants fed extremes of dietary<br />

intake. Clin Nutr 1987; 416: 105–113.<br />

51. Schultze KF, Stefanski M, Masterson J et al. Energy<br />

expenditure, energy balance and composition of weight<br />

gain in low birth weight infants fed diets of different<br />

protein and energy content. J Pediatr 1987; 110: 753–759.<br />

52. Reichman B, Chessex P, Verellen G et al. Dietary composition<br />

and macronutrient storage in preterm infants.<br />

Pediatrics 1983; 72: 322–328.<br />

53. Boehm G, Muller DM, Senger H et al. Nitrogen and fat<br />

balances in very low birth weight infants fed formula<br />

fortifier with human milk or bovine milk protein. Eur J<br />

Pediatr 1993; 152: 236–239.<br />

54. Schanler RJ, Abrams SA. Postnatal attainment of intrauterine<br />

macromineral accretion rate birth weight infants fed<br />

fortified human milk. J Pediatr 1995; 126: 441–447.<br />

55. Polberger S, Raihä NC, Juvonen P et al. Individualised<br />

protein fortification of human milk for preterm infants:<br />

comparison of ultrafiltrated human milk protein and a<br />

bovine whey fortifier. J Pediatr Gastroenterol Nutr 1999;<br />

29: 332–338.<br />

56. Reiss BB, Hall RT, Schanler RJ et al. Enhanced growth<br />

of preterm infants fed a new powdered human milk<br />

fortifier: a randomized, controlled trial. Pediatrics 2000;<br />

106: 581–588.<br />

57. Porcelli P, Schanler R, Greer F et al. Growth in human<br />

milk-fed very low birth weight infants receiving a new<br />

human milk fortifier. Ann Nutr Metab 2000; 44: 2–10.<br />

58. Putet G, Senterre J, Rigo G et al. Nutrient balance,<br />

energy utilization and composition of weight gain in<br />

very-low-birth-weight-infants fed pooled human milk or<br />

preterm formula. J Pediatr 1984; 105: 79–85.<br />

59. De Curtis M, Brooke OG. Energy and nitrogen balances<br />

in very low birth weight infants. Arch Dis Child 1987;<br />

62: 830–832.<br />

References 617<br />

60. Kashyap S, Schuylze KF, Ramakrishan R et al.<br />

Evaluation of a mathematical model for predicting the<br />

relationship between protein and energy intakes of lowbirth-weight<br />

infants and the rate and composition of<br />

weight gain. Pediatr Res 1994; 35: 704–712.<br />

61. Putet G, Senterre J, Rigo J et al. Energy balance and<br />

composition of body weight. Biol Neonate 1987; 52<br />

(Suppl 1): 17–24.<br />

62. Rodriguez M, Funkes S, Fink M et al. Plasma fatty acids<br />

and the [13C] linoleic acid metabolism in preterm<br />

infants fed a formula with medium-chain triglycerides. J<br />

Lipid Res 2003; 44: 41–48.<br />

63. Hamosh M, Henderson TR, Ellis LA et al. Digestive<br />

enzymes in human milk: stability at suboptimal storage<br />

temperatures. J Pediatr Gastroenterol Nutr 1997; 24: 38–43.<br />

64. Henderson MJ, Dear PRF. Dicarboxylic aciduria and<br />

medium chain triglyceride supplemented milk. Arch Dis<br />

Child 1990; 61: 610–611.<br />

65. Lucas A, Quinlan P, Abrams S et al. Randomised<br />

controlled trial of a synthetic triglyceride milk formula<br />

for preterm infants. Arch Dis Child Fetal <strong>Neonatal</strong> Ed.<br />

1997; 77: F178–F184.<br />

66. Koletzko B. Lipid supply and metabolism in infancy.<br />

Curr Opin Clin Nutr Metab Care 1998; 1: 171–177.<br />

67. Innis SM. The role of dietary n-6 and n-3 fatty acids in<br />

the developing brain. Dev Neurosci 2000; 22: 474–480.<br />

68. Koletzko BV, Innis SM. Lipids. In Tsang RC, Uauy R,<br />

Koletzko et al. eds. Nutritional Needs of the Preterm Infant.<br />

Cincinnati, OH: Digital Educational Publishing, 2004:<br />

43–76 (in press).<br />

69. Koletzko B, Agostoni C, Carlson SE et al. Long chain<br />

polyunsaturated fatty acids (LC-PUFA) and perinatal<br />

development. Acta Paediatr 2001; 90: 460–464.<br />

70. Auricchio S, Rubino A, Murset G. Intestinal glucosidase<br />

activity in the human embryo, fetus and newborn.<br />

Pediatrics 1965; 35: 944–954.<br />

71. Watkins JB. Developmental aspects of carbohydrate<br />

malabsorption in the premature infant. In Lifshitz F, ed.<br />

Carbohydrate Intolerance in Infancy. New York: Marcel<br />

Dekker, 1982: 61–73.<br />

72. Murray RD, Boutton TW, Klein PD et al. Comparative<br />

absorption of [ 13 C]glucose and [ 13 C]lactose by premature<br />

infants. Am J Clin Nutr 1990; 51: 59–66.<br />

73. Shulman RJ, Schanler RJ, Lau C et al. Early feeding,<br />

feeding intolerance, and lactase activity in preterm<br />

infants. J Pediatr 1998; 133: 645–659.<br />

74. Zittermann A, Bock P, Drummer C et al. Lactose does<br />

not enhance calcium bioavailability in lactose-tolerant,<br />

healthy adults. Am J Clin Nutr 2000; 71: 931–936.<br />

75. Brand Miller J, McVeagh P. Human milk oligosaccharides:<br />

130 reasons to breast-feed. Br J Nutr 1999; 82: 333–335.<br />

76. Coppa GV, Pierani P, Zampini L et al. Oligosaccharides<br />

in human milk during different phases of lactation. Acta<br />

Paediatr Suppl 1999; 430: 89–94.<br />

77. Coppa GV, Gabrielli O, Pierani P et al, Changes in carbohydrate<br />

composition in human milk over 4 months of<br />

lactation. Pediatrics 1993; 91: 637–641.<br />

78. Shulman RJ, Feste A, Ou C. Absorption of lactose,<br />

glucose polymers, or combination in premature infants.<br />

J Pediatr 1995; 127: 626–631.<br />

79. Siegel M, Kranz B, Lebenthal E. Effect of fat and carbohydrate<br />

composition on the gastric emptying of<br />

isocaloric feedings in premature infants.<br />

Gastroenterology 1985; 89: 785–790.<br />

80. Salle BL, Senterre J, Putet G. Calcium, phosphorus, magnesium<br />

and vitamin D requirements in premature infants. In<br />

Salle BL, Swyer PR, eds. Nutrition of the Low Birthweight<br />

Infant. Nestlé Nutrition Workshop Series, vol. 32, Nestec<br />

Ltd., New York: Vevey/Raven Press, 1993: 125–134.


618<br />

Enteral nutrition in preterm infants<br />

81. Salle B, Senterre J, Glorieux FH et al. Vitamin D metabolism<br />

in preterm infants. Biol Neonate 1987; 52: 119–130.<br />

82. Rigo J, De Curtis M, Salle BL et al. Bone mineral metabolism<br />

in the micropremie. Clin Perinatol 2000; 27: 147–170.<br />

83. Rigo J, Picaud JC, Lapillonne A et al. Metabolic balance<br />

and plasma AA concentrations in VLBW infants fed a<br />

new acidic whey hydrolysate preterm formula. J Pediatr<br />

Gastroenterol Nutr 1997; 24: 491A.<br />

84. Carnielli VP, Luijendijk IH, Goudoever JB et al. Feeding<br />

premature newborn infants palmitic acid in amounts<br />

and stereoisomeric position similar to that of human<br />

milk: effects on fat and mineral balance. Am J Clin Nutr<br />

1995; 61: 1037–1042.<br />

85. Cooke R, Hollis B, Conner C et al. Vitamin D and mineral<br />

metabolism in the very low birth weight infant receiving<br />

400 IU of vitamin D. J Pediatr 1990; 116: 423–428.<br />

86. Oski FA. Iron requirements of the premature infant. In<br />

Tsang RC, ed. Vitamin and Mineral Requirements in<br />

Preterm Infants. New York: Marcel Dekker, 1985; 9–21.<br />

87. Rao R, Georgieff M. Microminerals. In Tsang RC, Uauy<br />

R, Koletzko et al. eds. Nutritional Needs of the Preterm<br />

Infant. Cincinnati, OH: Digital Educational Publishing,<br />

2004: 43–76 (in press).<br />

88. American Academy of Pediatrics, Work Group on<br />

Breastfeeding. Breastfeeding and the use of human<br />

milk. Pediatrics 1997; 100: 1035–1039.<br />

89. Schanler RJ, Shulman RJ, Lau C. Feeding strategies for<br />

premature infants: beneficial outcomes of feeding fortified<br />

human milk vs preterm formula. Pediatrics 1998;<br />

103: 1150–1157.<br />

90. McGuire W, Antont MY. Donor human milk versus<br />

formula for preventing necrotising enterocolitis in<br />

preterm infants: systematic review. Arch Dis Child Fetal<br />

<strong>Neonatal</strong> Ed 2003; 88: F11–F14.<br />

91. Schanler RJ, Hurst NM, Lau C. The use of human milk<br />

and breastfeeding in premature infants. Clin Perinatol<br />

1999; 26: 379–398.<br />

92. Lucas A, Morley R, Cole J et al. A randomized multicentre<br />

study of human milk versus formula and later development<br />

in preterm infants. Arch Dis Child 1994; 70: F141–F146.<br />

93. Carlson SE, Werkman SH, Rhodes PG et al. Visual-acuity<br />

development in healthy preterm infants: effect of marineoil<br />

supplementation. Am J Clin Nutr 1993; 58: 35–42.<br />

94. Atkinson SA, Bryan MH. The effect of gestational stage<br />

at delivery on human milk components. In Jensen RG,<br />

ed. Handbook of Milk Composition. San Diego:<br />

Academic Press, 1995: 222–237.<br />

95. Atkinson SA. Human milk feeding of the micropremie.<br />

Clin Perinatol 2000; 27: 235–247.<br />

96. Linzell JL, Peaker M. Changes in colostrums composition<br />

and in permeability of mammary epithelium at about the<br />

time of parturition in the goat. J Physiol 1974; 243: 129–151.<br />

97. Anderson GH. The effect of prematurity on milk composition<br />

and its physiological basis. Fed Proc 1984; 43:<br />

2438–2442.<br />

98. Senterre J, Voyer M, Putet G et al. Nitrogen, fat and<br />

mineral balance studies in preterm infants fed bank<br />

human milk, a human milk formula, or a low-birthweight<br />

infant formula. In Baum JD, ed. Human Milk<br />

Processing, Fractionation, and the Nutrition of the Low<br />

Birth-Weight Baby. Nestlé Nutrition Workshop series,<br />

New York: Raven Press, 1983; 3: 102–111.<br />

99. De Curtis M, Candusso M, Pieltain C et al. Effect of<br />

fortification on the osmolality of human milk. Arch Dis<br />

Child Fetal <strong>Neonatal</strong> Ed 1999; 81: F141–F143.<br />

100. Hallstrom M, Koivisto AM, Janas M et al. Frequency of<br />

and risk factors for necrotizing enterocolitis in infants<br />

born before 33 weeks of gestation. Acta Paediatr 2003;<br />

92: 111–113.<br />

101. Porcelli P, Schanler R, Greer F et al. Growth in human<br />

milk-fed very low birth weight infants receiving a new<br />

human milk fortifier. Ann Nutr Metab 2000; 44: 2–10.<br />

102. Pieltain C, De Curtis M, Gerard P et al. Weight gain<br />

composition in preterm infants fed fortified human milk<br />

or preterm formula. Pediatr Res 2001; 49: 120–124.<br />

103. Putet G, Rigo J, Salle B et al. Supplementation of pooled<br />

human milk with casein hydrolysate: energy and nitrogen<br />

balance and weight gain composition in very-lowbirth-weight-infants.<br />

Pediatr Res 1987; 21: 458–461.<br />

104. Picaud JC, Putet G, Rigo J et al. Metabolic and energy<br />

balance in small-and-appropriate for gestational age in<br />

very low-birth-weight infants, Acta Paediatr Suppl 1994;<br />

405: 54–59.<br />

105. Rigo J, Senterre J, Putet G et al. Various human milk<br />

fortifiers in low birth weight infants fed pooled human<br />

milk: plasma and urinary amino acid concentrations. In<br />

Koletzko B, Okken A, Rey J, Salle B, Van Biervliet JP,<br />

eds. Recent Advances in Infant Feeding. New York:<br />

Georg Thiem Verlag, 1992: 164–170.<br />

106. Boehm G, Muller DM, Senger H et al. Nitrogen and fat<br />

balances in very low birth weight infants fed formula<br />

fortifier with human milk or bovine milk protein. Eur J<br />

Pediatr 1993; 152: 236–239.<br />

107. Schanler RJ, Abrams SA. Postnatal attainment of intrauterine<br />

macromineral accretion rate birth weight infants fed<br />

fortified human milk. J Pediatr 1995; 126: 441–447.<br />

108. Moody GJ, Schanler RJ, Lau C et al. Feeding tolerance in<br />

premature infants fed fortified human milk. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 408–412.<br />

109. Kuschel CA, Harding JE. Multicomponent fortified<br />

human milk for promoting growth in preterm infants.<br />

Cochrane Review 2001; 3:<br />

http://www.cochranelibrary.com/enter<br />

110. Jocson MAL, Mason EO, Schanler RJ. The effects of<br />

nutrient fortification and varying storage conditions on<br />

host defense properties of human milk. Pediatrics 1997;<br />

100: 240–243.<br />

111. Mihatsch WA, Hogel J, Pohlandt F. Hydrolysed protein<br />

accelerates the gastrointestinal transport of formula in<br />

preterm infants. Acta Paediatr 2001; 90: 1–3.<br />

112. Bar-Oz B, Preminger A, Peleg O et al. Enterobacter sakazakii<br />

infection in the newborn. Acta Paediatr 2001; 90: 356–358.<br />

113. Lucas A, Bishop NJ, King FJ et al. Randomised trial of<br />

nutrition for preterm infants after discharge. Arch Dis<br />

Child 1992; 67: 324–327.<br />

114. Rigo J, Boboli H, Franckart G et al. Surveillance de l’ancien<br />

prematuré: croissance et nutrition. Arch Pédiatr<br />

1998; 5: 449–453.<br />

115. Brunton JA, Saigal S, Atkinson SA. Growth and body<br />

composition in infants with bronchopulmonary dysplasia<br />

up to 3 months corrected age: a randomized trial of<br />

a high-energy nutrient-enriched formula fed after hospital<br />

discharge. J Pediatr 1998; 133: 340–345.<br />

116. Cooke RJ, McCormick K, Griffin IJ et al. Feeding preterm<br />

infants after hospital discharge: effect of dietary manipulation<br />

on nutrient intake and growth. Pediatr Res 1998;<br />

43: 355–360.<br />

117. De Curtis M, Pieltain C, Rigo J. Body composition in<br />

preterm infants fed standard or enriched formula after<br />

hospital discharge. Eur J Nutr 2002; 41: 177–182.<br />

118. Rolland-Cachera MF. Early adiposity rebound is not<br />

associated with energy or fat intake in infancy.<br />

Pediatrics 2001; 108: 218–219.<br />

119. De Curtis M, Pieltain C, Rigo J. Nutrition of preterm<br />

infants on discharge from hospital. In Raiha NCR,<br />

Rubaltelli FF, eds. Infant Formula: Closer to the<br />

Reference. Nestlè Nutrition Workshop Series, Nestec<br />

Ltd. Vevey/Lippincott Williams & Wilkins Philadelphia,<br />

2002; 47 (Suppl): 149–163.


37<br />

Introduction<br />

Parenteral nutrition in<br />

premature infants<br />

Jacques Rigo and Mario De Curtis<br />

Over the past decades there has been a dramatic<br />

increase in the survival of preterm infants, especially<br />

very-low-birth-weight (VLBW) infants.<br />

Prenatal steroids to enhance pulmonary maturation,<br />

the use of exogenous surfactant for the treatment<br />

of the respiratory distress syndrome, and<br />

better prenatal obstetric and postnatal neonatal<br />

intensive care have all played a major role in<br />

improving survival rate in these infants. 1–3<br />

With the major advances in life-support measures,<br />

nutrition has become one the most debated issues<br />

in the care of low-birth-weight infants; in this<br />

regard, several reports have shown the important<br />

effect of nutrition during the first period of life on<br />

early and late outcome. 4<br />

Although the general objective of a nutritional<br />

regimen for preterm infants is to support life and<br />

achieve a growth rate sufficient to fulfill the individual’s<br />

genetic potential, there are many controversies<br />

on how to attain this goal. In this chapter<br />

we discuss the most important features regarding<br />

the nutrition of preterm, and particularly VLBW<br />

infants, outlining the main aspects of parenteral<br />

nutrition.<br />

Nutritional requirements<br />

While the nutritional reference standard for fullterm<br />

newborns is an exclusively breast-fed infant,<br />

no similar standard is available for preterm<br />

infants. In the neonatal period, preterm infants<br />

and especially VLBW infants have greater nutritional<br />

needs in order to achieve optimal growth<br />

than at any other time in their life. Preterm birth is<br />

associated with decreased nutrient storage, and<br />

the clinical conditions after birth may increase<br />

nutrient needs.<br />

The recommended nutrient intakes of preterm<br />

infants are still a matter of debate, arising from the<br />

lack of consensus on the short- and long-term<br />

objectives for early nutrition. Current nutritional<br />

recommendations from various international<br />

committees (American Academy of Pediatrics,<br />

European Society for Pediatric Gastroenterology,<br />

Hepatology and Nutrition, Canadian Academy of<br />

Pediatrics) are based on healthy preterm infants<br />

and designed to provide postnatal nutrient retention<br />

during the ‘stable-growing’ period equivalent<br />

to the intrauterine gain of a normal fetus. They do<br />

not take into consideration the relatively long transitional<br />

period that induces the cumulative nutritional<br />

deficit and the need to obtain an early catchup<br />

growth. 5–7<br />

The postnatal use of the gastrointestinal tract,<br />

instead of placental transfer, and the metabolic<br />

changes that occur immediately after birth, do not<br />

allow provision of the quantitative or qualitative<br />

supply of all the nutrients needed to approximate<br />

fetal growth of the comparable corrected age.<br />

Additional nutritional deprivations can also arise<br />

from neonatal illnesses such as metabolic stress,<br />

infectious diseases, necrotizing enterocolitis or<br />

bronchopulmonary dysplasia, which often further<br />

decrease nutrient supply.<br />

In practice, the shorter the gestation of a neonate<br />

the more challenging are the influences of immaturity<br />

and the accompanying morbidity on nutritional<br />

supply during the early weeks of life. As a<br />

result of this cumulative nutritional deficit during<br />

early life, most growth parameters remain subnormal<br />

by the time the preterm infant reaches a<br />

corrected age of 40 weeks (Figure 37.1); this<br />

phenomenon worsens in the case of VLBW and<br />

619


620<br />

Z Score compared to Usher & McLean (SD)<br />

1.5<br />

0.5<br />

-0.5<br />

-1.5<br />

-2.5<br />

-3.5<br />

-4.5<br />

Parenteral nutrition in premature infants<br />

At birth<br />

At discharge (± 7 weeks)<br />

Body weight Body length<br />

Figure 37.1 Z score of body weight, body length<br />

determined in healthy very-low-birth-weight infants (birth<br />

weight 2g amino acids/kg per day) and the use of intravenous<br />

lipids (0.5–1.0g lipid/kg per day), from the<br />

first day of life, associated when necessary, with<br />

the use of insulin to improve carbohydrate tolerance<br />

during the early adaptive period. 10,11<br />

Nutrition during the ‘stable-growth’ period on PN,<br />

is given exceptionally, when preterm infants are<br />

recovering from severe gastrointestinal problems,<br />

to prevent or limit the use of the gastrointestinal<br />

tract. In most preterm infants, nutritional supplies<br />

are provided using the mother’s milk supplemented<br />

with human milk fortifiers or preterm<br />

formulas specially designed for VLBW infants.<br />

The objective of nutrition in this period is to minimize<br />

postnatal growth deficit, maximize longitudinal<br />

growth and induce catch-up growth.<br />

Although the optimal nutritional supply is still<br />

controversial, more recent studies suggest that, in<br />

order specifically to enhance lean mass growth<br />

and improve the early catch-up growth in preterm<br />

infants, the use of a high-protein regimen<br />

combined with a high protein-energy density<br />

regimen (4.0–4.4g of protein/kg per day with a<br />

protein/energy ratio up to 3.3g/100kcal) promotes


growth without metabolic stress in VLBW<br />

infants. 12,13<br />

Parenteral nutrition<br />

PN provides the infant’s requirements for growth<br />

and development when the infant’s size or clinical<br />

conditions preclude enteral feeding. PN in<br />

preterm infants is prescribed especially for those<br />

with respiratory distress, before starting enteral<br />

feeding, and can be increased slowly to avoid<br />

overloading the immature gastrointestinal tract<br />

while continuing to satisfy nutritional requirements.<br />

A recent report indicates that in the USA, even<br />

though over the last 15 years, there has been a<br />

decrease in the length of days of PN in VLBW<br />

infants, most of these VLBW infants receive<br />

parenterally delivered nutrients as their major<br />

source of nutrition for the first few days to weeks<br />

of life. 14<br />

Today PN is seen by many neonatologists as a<br />

means of achieving rapid nutrition and administering<br />

nutrients that could be delayed and/or<br />

insufficiently absorbed via the enteral route, especially<br />

in the presence of conditions considered to<br />

be hazardous to the intestine and thus increasing<br />

the risk for necrotizing enterocolitis. PN allows<br />

infusion of concentrated hypertonic nutrient solutions<br />

without excessive fluid intake (which is not<br />

well tolerated by most VLBW infants), through an<br />

indwelling catheter, the tip of which is in the<br />

superior vena cava just above the right atrium.<br />

This technique was first described by Wilmore<br />

and Dudrick in 1968. 15<br />

Nowadays, in ELBW infants, PN is usually administered<br />

through the umbilical vessels for a limited<br />

number of days, followed by the use of a silastic<br />

catheter introduced percutaneously from a peripheral<br />

vein. Less frequently, in bigger preterm<br />

infants, a catheter is inserted surgically into either<br />

the internal or external jugular vein and tunneled<br />

subcutaneously. Currently, the use of direct infusion<br />

via peripheral veins has become less common<br />

because of the relatively high osmolarity of the<br />

solution that can lead to endothelial damage,<br />

frequent interruption of continuous feeding and<br />

relative reduction of the nutritional supplies.<br />

Fluids<br />

Parenteral nutrition 621<br />

A number of physiological changes and adaptive<br />

processes that occur at birth, immediately or<br />

subsequently affect water and electrolyte metabolism.<br />

In particular, placental clearance and placental<br />

supply of fluids, electrolytes and nutrients are<br />

discontinued, and a considerable insensible water<br />

loss and infant thermoregulation begin to occur.<br />

Compensatory regulative processes (such as renal<br />

adaptation, start of oral intake) need time to counteract<br />

these effects on body water pools.<br />

Just after the birth, there is a fall in urinary<br />

output due to a decreased glomerular filtration<br />

rate. This relative oliguria, which may last for a<br />

variable period (hours or days), is mainly determined<br />

by the presence of conditions and<br />

diseases, such as respiratory distress syndrome.<br />

Oliguria is then followed by a diuretic phase,<br />

during which a contraction of extracellular fluid<br />

volume occurs. The end of this transitional<br />

period is usually characterized by a urine volume<br />

3% to


622<br />

Parenteral nutrition in premature infants<br />

enormous, especially when the baby is under a<br />

radiant warmer and is undergoing phototherapy.<br />

A reduction of insensible water loss can be<br />

obtained by increasing the humidity of incubators.<br />

Increased fluid loss occurs when the infant<br />

is placed under radiant warmers or undergoes<br />

phototherapy. 16 Fluid requirements are decreased<br />

with the use of double-walled incubators, heat<br />

shields or plastic blankets and high-ambient<br />

humidity. 17,18<br />

The following intermediate phase is characterized<br />

by a decrease in insensible water loss from the<br />

skin, the increasing maturation of the epidermis,<br />

and improved renal function. In this period fluid<br />

and electrolytes have to be increased in order to<br />

cover the requirements (120–150ml/kg per day)<br />

and to allow adequate growth. During this period<br />

the amount of parenteral fluid can be reduced and<br />

enteral feeding can be augmented. Infants with<br />

bronchopulmonary dysplasia, patent ductus arteriosus,<br />

renal failure and respiratory distress<br />

syndrome, are very vulnerable to fluid overload.<br />

Energy<br />

Energy must be supplied by nutrient intake to<br />

cover two major components, energy expenditure<br />

and growth. A caloric intake of 40–60kcal/kg per<br />

day approximates energy expenditure and is a<br />

reasonable goal for the maintenance requirements<br />

of premature infants for the first days after birth.<br />

The energy cost of gaining 1g of new tissue is<br />

about 5kcal. This value is influenced largely by<br />

quantity of fat deposition ranging between 20 and<br />

40% of postnatal weight gain in premature infants,<br />

according to the feeding regimen. 19 The lower the<br />

fat deposition, the lower is the cost of growth;<br />

thus, energy cost of weight gain during the third<br />

trimester of intrauterine life, 16–20g/kg per day,<br />

with 15% fat, is lower than that for postnatal<br />

weight gain accounting for a higher fat deposition,<br />

up to 40%.<br />

In contrast to what is generally proposed, the<br />

energy requirement in PN approximates to that of<br />

enteral nutrition. In fact, the energy content of<br />

amino acid solutions in PN is lower than that<br />

provided by the protein in enteral nutrition<br />

(human milk or formulas). The gross energy,<br />

measured by calorimetric bomb, provided by<br />

glucose used in PN is less than that provided by<br />

the carbohydrate content of formulas. In contrast,<br />

while the metabolizable energy of amino acid solutions<br />

and glucose, given by the parenteral route, is<br />

identical to total intake, the metabolizable energy<br />

of dietary protein and carbohydrates represents<br />

about 90% of the intake. 20,21 Consequently, the<br />

recommendation for energy intake during the<br />

stable-growing period in VLBW infants on PN<br />

corresponds to 110–135kcal/kg per day.<br />

Carbohydrates<br />

Glucose is the most widely used intravenous<br />

carbohydrate for neonates, because it is readily<br />

available to the brain. The monohydrate form of<br />

dextrose is used for intravenous solutions; each<br />

gram provides 3.7kcal upon complete oxidation. 20<br />

As the solution concentration increases, so does<br />

osmolarity, ranging from approximately<br />

255mOsm/l for a 5% solution to 1020mOsm/l for a<br />

20% solution. Many other non-glucose carbohydrates<br />

have been tried, but with limited success.<br />

The newborn infant is often in a transitional stage<br />

of glucose homeostasis and is, therefore, subject to<br />

hyper- or hypoglycemia. Although the definition<br />

and the long-term consequences of neonatal hypoglycemia<br />

remain controversial, plasma glucose<br />

concentration should be monitored and corrected<br />

if it falls below 35mg/dl (1.9mmol/l) during the<br />

first days of life. 22 In the first few days of life,<br />

VLBW infants are more susceptible to hyperglycemia<br />

often associated with glucosuria, which<br />

seems to be caused by a persistent endogenous<br />

hepatic glucose production secondary to an insensitivity<br />

of hepatocytes to insulin. 23–25 The definition<br />

of hyperglycemia varies but is generally set at<br />

a plasma level above 150mg/dl (8.3mmol/l).<br />

However the upper ‘safe’ limit of blood glucose<br />

concentration in the neonate is not well defined<br />

and, as for hypoglycemia, there is a great variation<br />

in terms of the diagnosis and management of<br />

hyperglycemia. The increase in serum osmolarity<br />

induced by the increase in blood glucose has been<br />

associated with higher mortality and a higher rate<br />

of intracranial hemorrhage. Glycosuria associated<br />

with hyperglycemia results from the relatively low<br />

renal threshold for glucose excretion in the<br />

preterm infant compared with the adult and may<br />

increase the risk of dehydration. Risks for hyper-


glycemia and glycosuria increase with decreasing<br />

gestation and birth weight, and is estimated to be<br />

between 20% and 86% during the first few days of<br />

life in preterm infants who survive their first<br />

week. 26,27<br />

If hyperglycemia is detected early through<br />

frequent glucose monitoring, plasma glucose<br />

levels can be decreased by reducing insensible<br />

water loss, glucose infusion rate and by providing<br />

exogenous insulin supply. Decreasing volume<br />

intake and glucose concentration reduces energy<br />

supply and consequently growth and nitrogen<br />

retention. Insulin administration helps control<br />

plasma glucose concentration, achieve increased<br />

energy intake and promote nitrogen retention and<br />

growth, although there is need for more data on its<br />

safety and long-term consequences as a growthpromoting<br />

agent. More recently, it has been<br />

proposed that high amino acid intake, 2–3g/kg per<br />

day from the first day, in addition to preventing<br />

catabolism and promoting anabolism, may have<br />

several other beneficial effects including decreasing<br />

the frequency and severity of neonatal hyperglycemia<br />

by stimulating endogenous insulin secretion<br />

and stimulating growth by enhancing the<br />

secretion of insulin and insulin-like growth<br />

factors. 28<br />

In practice, 6g/kg per day of intravenous glucose<br />

are generally well tolerated (4–5mg/kg per min)<br />

even on the first day of life in VLBW infants. If this<br />

intake is tolerated, it may be increased to 8, 10 and<br />

up to 12–18g/kg per day. If it is not tolerated,<br />

progression of glucose intake will be stopped and<br />

Parenteral nutrition 623<br />

insulin perfusion will be considered according to<br />

clinical and nutritional status with an initial dose<br />

of 0.05IU/kg per h. 29<br />

Protein<br />

In order to define the protein requirements for<br />

preterm infants more precisely, it has been<br />

suggested that lean body mass be taken into<br />

account instead of weight gain, also considering<br />

that preterm infants present a high fat deposition<br />

in the postnatal period. Table 37.1 shows the<br />

revised recommended protein intake and<br />

protein/energy ratio for preterm infants according<br />

to postconceptional age and the need for catch-up<br />

growth. 12 It should be stressed that these recommendations<br />

derive especially from studies on<br />

preterm infants greater than 1000g rather than on<br />

ELBW infants (


624<br />

Parenteral nutrition in premature infants<br />

therapy and by catabolic stress due to diseases or<br />

early neonatal surgery.<br />

Amino acid composition of solutions for<br />

parenteral nutrition<br />

Considerable improvements in parenteral amino<br />

acid solutions have been made since the 1960s<br />

when the source of intravenous protein was casein<br />

hydrolysate. The use of a crystalline amino acid<br />

mixture in the early 1980s helped to improve the<br />

nitrogen utilization whereas, more specific pediatric<br />

amino acid solutions were designed in the<br />

early 1990s with a high essential/non-essential<br />

amino acid ratio and conditionally essential amino<br />

acid content for use in preterm infants. Despite the<br />

new evidence, the reference optimal solution for<br />

PN has not yet been well defined because the<br />

optimal value for plasma amino acid concentrations<br />

in preterm infants is still a matter of debate.<br />

At least three different ‘gold standards’ have been<br />

proposed for premature infants: first, the amino<br />

Plasma concentration (µmol/100ml)<br />

65<br />

60<br />

55<br />

50<br />

45<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

5<br />

0<br />

GLN<br />

ASP<br />

LYS<br />

PRO<br />

GLY<br />

THR<br />

VAL<br />

TYR<br />

SER<br />

TAU<br />

acid concentrations from the umbilical cord<br />

obtained following fetal cord puncture or after<br />

birth; second, the amino acid concentrations of<br />

rapidly growing preterm infants receiving their<br />

mother’s milk or human milk supplemented with<br />

human milk proteins; and third, the amino acid<br />

concentrations of healthy breast-fed term infants.<br />

The amino acid concentrations observed in VLBW<br />

infants during the last trimester of gestation or in<br />

well-growing preterm infants fed an optimal intake<br />

of human milk protein appear to be safe. However,<br />

considering the large differences observed between<br />

fetal and postnatal values of some amino<br />

acids (THR, VAL, TYR, PHE, LYS and HIS), a<br />

combined reference has been proposed taking into<br />

account the mean ±1 SD of the values obtained in<br />

cord blood and in the postprandial serum of<br />

preterm infants fed human milk supplemented<br />

with human milk proteins (Figure 37.2). 31<br />

Nevertheless, despite the diverse composition of<br />

parenteral amino acid solutions used in pediatric<br />

care, nitrogen utilization does not change signifi-<br />

Figure 37.2 Plasma amino acid reference (adapted from reference 31). HMF, human milk fortified with human milk<br />

protein; Cord, cord blood concentrations.<br />

GLU<br />

HIS<br />

ORN<br />

LEU<br />

ARG<br />

HPR<br />

ASN<br />

ILE<br />

Cord<br />

HMF<br />

Reference<br />

PHE<br />

TRP<br />

CYS<br />

MET<br />

CIT<br />

ASP


cantly. 32,33 Nitrogen amino acid content is related to<br />

the amino acid composition and varies according to<br />

the solution, and is frequently lower than in enteral<br />

nutrition (160mg/g of protein). Therefore, it is probably<br />

preferable to consider nitrogen rather than<br />

amino acid intake in total PN.<br />

Analysis of a large number of nitrogen balances in<br />

preterm infants shows that a nitrogen retention of<br />

380mg/kg per day can be obtained with a nitrogen<br />

intake of 530mg/kg per day (Figure 37.3), 33–43 which<br />

corresponds to a net protein utilization (nitrogen<br />

retention/nitrogen intake) of 70%, a value quite<br />

similar to that recorded in oral nutrition (60–72%). 44<br />

These data indicate that the nitrogen requirement in<br />

PN is close to 95% of the enteral requirement, but<br />

corresponds to a similar amino acid versus protein<br />

requirement in parenteral and enteral nutrition.<br />

Parenterally fed preterm infants are at risk of toxicity<br />

from excessive amino acid intake, immature metabolic<br />

pathways, absence of first hepatic crossing and<br />

lack of small-intestinal metabolism. Therefore,<br />

optimal amino acid pattern for parenteral amino<br />

acid solutions in preterm infants still needs to be<br />

determined, and new solutions should be developed<br />

keeping in mind the following principles:<br />

Nitrogen retention (mg/kg per day)<br />

450<br />

400<br />

350<br />

300<br />

250<br />

200<br />

150<br />

100<br />

50<br />

0<br />

100<br />

Parenteral nutrition 625<br />

(1) The requirement of essential amino acids in<br />

VLBW infants is greater than in term<br />

neonates or older infants; 45<br />

(2) The activity of phenylalanine hydroxylase as<br />

well as tyrosine aminotransferase and 4dioxyphenylpyruvate<br />

dioxygenase is considered<br />

to be immature during the neonatal<br />

period; 46<br />

(3) The activity of cystathionase in neonates is<br />

low and cysteine synthesis from methionine<br />

is reduced; 47<br />

(4) Nutritional supply of taurine is probably<br />

necessary during the neonatal period.<br />

Although cysteine sulphinic activity has been<br />

demonstrated in preterm infants, taurine is<br />

probably considered as a conditionally essential<br />

amino acid in neonates; 48<br />

(5) In addition to cysteine and tyrosine, histidine<br />

and arginine are categorized as semi-essential<br />

amino acids in neonates; 49<br />

(6) Organ dysfunction tends to occur when some<br />

amino acids are administered in high<br />

(methionine) or low (arginine, glutamine)<br />

amounts. 50,51<br />

150 200 250 300 350 400 450 500 550<br />

Nitrogen intake (mg/kg per day)<br />

Figure 37.3 Relationship between nitrogen retention and nitrogen intake in parenterally fed preterm infants. 12,33–43


626<br />

Parenteral nutrition in premature infants<br />

Table 37.2 Composition of commercially available parenteral amino acid solutions 12<br />

Aminoplasmal- Aminosyn Aminosyn-PF Aminovemos FreAmine III Neophan MPF<br />

Aminoped N-Pad (FAO/WHO)<br />

(Pfrimmer-kabi) (Abbott) (Abbott) (Fresenios) (McGaw) (Cutter) General use<br />

Trp 4 1.6 1.8 1.8 1.5 2.1 1.3<br />

Ile 5.1 7.2 7.6 6.4 6.9 4.7 5.6<br />

Leu 7.6 9.4 11.9 10.7 9.1 10.7 12.5<br />

Val 6.1 8 6.5 7.1 6.6 5.5 4.5<br />

Lys 8.8 7.2 6.8 7.1 7.3 8.6 8.8<br />

Met 2 4 1.8 4.3 5.3 2 3.5<br />

Phe 3.1 4.4 4.3 4.6 5.6 4.1 9.4<br />

Thr 5.1 5.2 5.1 5.1 4 5.5 6.5<br />

His 4.6 3 3.1 4.1 2.8 3.2 6<br />

Arg 9.1 9.8 12.3 6.4 9.5 6.3 7.9<br />

Orn 0 0 0 0 0 0 0<br />

Gly 2 12.8 3.9 4.1 14 3.2 10.7<br />

Ala 15.9 12.8 7 7.1 7.1 9.7 6.2<br />

Glu 9.3 0 8.2 0 0 10.9 6.5<br />

Asp 6.6 0 5.3 0 0 6.3 3.8<br />

Pro 6.1 8.6 8.1 16.1 11.2 8.6 3.3<br />

Ser 2 4.2 5 9 5.9 5.8 2.2<br />

Tyr 1.1* 0.4 0.6 1.6* 0.7 0.3<br />

Cys 1 0 0.6 0.2 1.5 1<br />

Tau 0.3 0 0.7 0 0 0<br />

AAA 46% 50% 49% 51% 49% 46% 58%<br />

BCAA 19% 25% 26% 24% 23% 21% 23%<br />

Nitrogen 160 167 165 152 162 153 165<br />

To date, the normalization of plasma amnio acid<br />

concentrations in PN has never been achieved due<br />

to the poor solubility of free tyrosine and cystine<br />

and the absence of an adequate and safe metabolic<br />

substitute. 52<br />

Table 37.2 shows some commercially available<br />

parenteral amino acid solutions.<br />

Amino acids for special purposes<br />

Arginine<br />

Arginine is a precursor of nitric oxide; inadequate<br />

availability has been implicated in various neonatal<br />

Continued<br />

diseases, such as persistent pulmonary hypertension<br />

and necrotizing enterocolitis. Recent evidence<br />

suggests that the de novo arginine synthesis in the<br />

neonate relies on small-intestinal metabolism, and<br />

that dietary arginine requirements could be higher<br />

in the parentally fed infants. 53 Currently, arginine<br />

content varies widely in pediatric parenteral solutions<br />

suggesting that the dietary requirement in<br />

parenteral and oral nutrition must be elucidated.<br />

Glutamine<br />

Glutamine is the most abundant amino acid in the<br />

human body and the main one supplied to the fetus


Table 37.2 Continued<br />

through the placenta. Glutamine is thought to be an<br />

important fuel for rapidly dividing cells such as<br />

enterocytes and lymphocytes. It is a regulator of<br />

acid–base balance via ammonium, as well as an<br />

important precursor of nucleic acids, nucleotides<br />

amino sugars and protein. There is, however, little<br />

information on the role of glutamine in children<br />

and infants or as to whether glutamine supplementation<br />

is beneficial in preterm babies. In ELBW<br />

infants and during critical illnesses glutamine could<br />

be a conditionally essential amino acid. 54<br />

Reviewing the available studies in preterm infants<br />

for the Cochrane database, Tubman and<br />

Thompson 55 did not find evidence to support the<br />

Parenteral nutrition 627<br />

PED Pleamine-P Primene Travasol Trophamine VAMIN Infant<br />

(B. Braun) (Fuso, Japan) (Baxter) (Clintec) (Kendall–McGaw) (Kabivitrun)<br />

Trp 1 1.6 2 1.8 2 2.2<br />

Ile 2.8 10.5 6.7 6 8.2 4.8<br />

Leu 5 21 10 7.3 14 10.8<br />

Val 3.2 7.9 7.6 5.8 7.8 5.5<br />

Lys 9 6.3 11 5.8 8.2 8.6<br />

Met 1.6 2 2.4 4 3.3 2<br />

Phe 3.8 3.3 4.2 5.6 4.8 4.2<br />

Thr 6 3.2 3.7 4.2 4.2 5.5<br />

His 6 3.3 3.8 4.8 4.8 3.2<br />

Arg 4.4 13.2 8.4 11.5 12.1 6.3<br />

Orn 0 0 2.2 0 0 0<br />

Gly 9.8 2.6 4 10.3 3.7 3.2<br />

Ala 11.6 6.8 8 20.7 5.3 9.7<br />

Glu 19.6 1.1 10 0 5 10.9<br />

Asp 3.8 1.1 6 0 3.2 6.3<br />

Pro 5.4 7.9 3 6.8 6.8 8.6<br />

Ser 2 5.3 4 5 3.8 5.9<br />

Tyr 0.6 0.8 0.5 0.4 0.7* 0.8<br />

Cys 1.7 2 1.7 0 0.2 1.5<br />

Tau 0 0.2 0.6 0 0.3 0<br />

AAA 38% 59% 51% 45% 57% 47%<br />

BCAA 11% 39% 24% 19% 30% 21%<br />

Nitrogen 149 177 161 173 168 154<br />

All amino acid mixtures are in % amino acid content. Nitrogen expressed as mg/100 ml<br />

AAA, aromatic amino acid; BCAA, branched chain amino acid<br />

* Mixture of L-tyrosine and N-acetyl-L-tyrosine<br />

routine use of parenteral or enteral glutamine<br />

supplementation in preterm infants. A more<br />

recent, multicentric study on ELBW infants<br />

showed that parenteral glutamine supplementation<br />

can increase plasma glutamine concentrations,<br />

but the potential clinical effects of such glutamine<br />

supplementation remain to be elucidated. 56<br />

Intravenous lipids<br />

Intravenous lipid emulsions are important<br />

constituents of total PN as they provide energy and<br />

essential fatty acids to VLBW infants. Lipid<br />

emulsions consist of different oils, egg yolk


628<br />

Parenteral nutrition in premature infants<br />

phospholipids and glycerol. The fat particles in the<br />

emulsions resemble endogenously produced<br />

chylomicrons in terms of size, physicochemical<br />

properties and metabolism, and are hydrolyzed by<br />

lipoprotein lipase. The rate of infused triglyceride<br />

clearance is determined by the available lipase<br />

activity and by the uptake of unesterified fatty acid<br />

products related to the adipose tissue mass and/or<br />

fatty acid oxidation in muscles. Intravenous lipid<br />

emulsions provide high caloric, isotonic solutions<br />

of linoleic acid (18:2n-6) and linolenic acid (18:3n-<br />

3), as well as other fatty acids, and can also be<br />

given through peripheral lines. Traditionally, lipid<br />

infusions are prepared from soybean oil triglycerides<br />

emulsified with egg yolk phospholipids.<br />

Typical soybean oil contains about 45–55%<br />

linoleic acid (18:2n-6) and 6–9% linolenic acid<br />

(18:3n-3), but very little saturated or monounsaturated<br />

fat (Table 37.3). 57 There are considerable<br />

concerns over the effect of these emulsions on the<br />

composition of the fatty acids deposited in the<br />

developing tissues of preterm infants.<br />

Intravenous lipids play two separate roles in the<br />

PN of VLBW infants. One is the role of intravenous<br />

lipids as an energy substrate to be readily utilized<br />

by VLBW infants. There is some evidence that the<br />

supply of some of the energy as lipids is preferable<br />

over carbohydrate as the sole energy substrate. 58<br />

Amino acid oxidation and protein breakdown<br />

were significantly lower when lipids provided<br />

50% of the non-protein calories than when glucose<br />

alone served as the energy substrate. Advantages<br />

of lipids emulsions over concentrated glucose<br />

solutions also include their isotonicity and greater<br />

energy density, the latter meaning that less volume<br />

is required per calorie. This is particularly true for<br />

20% lipid emulsion, which is preferred over the<br />

10% emulsion because it leads to a more normal<br />

pattern of plasma phospholipid and cholesterol<br />

than the 10% intralipid. 59 This beneficial effect of<br />

the 20% emulsion is attributed to the lower phospholipid<br />

content (per gram of triglyceride),<br />

although for the lower phospholipid content, the<br />

20% emulsion delivers less long-chain polyunsat-<br />

Table 37.3 Commercial intravenous lipid emulsions currently used in preterm infants 57<br />

Soybean Soybean/MCT Olive/soybean Soybean/fish<br />

Product names Intralipid (Pharmacia) Lipofundin ClinOleic Lipovenous +<br />

Ivelip (Clintec) MCT(Braun) (Clintec) Omegavenös,<br />

Lipofundin (Braun) 9+1 parts mix<br />

Lipovenös (Fresenius) (Fresenius)<br />

Liposyn III (Abbott)<br />

Triglycerides (%) 10, 20 & 30 20 20 10<br />

Ratio of phospholipids/ 120, 60 & 40 60 60 66<br />

triglycerides (mg/g)<br />

Glycerol (%) 2.5 2.5 2.25 2.5<br />

kcal/ml 1.1–2.0 1.9 2.0 —<br />

Fatty acid composition (% wt/wt)<br />

Medium chain (8:0+10:0) n.d. 50 n.d. n.d.<br />

Palmitic (16:0) 9–11.2 5 13.5 10.8<br />

Stearic (18:0) 4–4.2 2 2.9 3.7<br />

Oleic (18:1n9) 20.4–26 12 59.5 19.9<br />

Linoleic (18:2n6) 52.4–54.5 27 18.5 49.7<br />

linolenic (18:3n3)<br />

linolenic (18:3n6)<br />

8–8.5 4 2 6.5<br />

Arachidonic (20:4n6) n.d.–0.2 n.d. 0.2 0.2<br />

Eicosapentaenoic (20:5n3) n.d. n.d. n.d. 2.4<br />

Docosahexaenoic (22:6n3) n.d.–0.1 n.d. 0.1 2.3<br />

MCT, medium-chain triglyceride, n.d. not determined


urated fatty acids (PUFAs) (contained in the phospholipid<br />

fraction) than the 10% emulsion. For this<br />

reason, it seems that, at least in the first days of<br />

life, until the lipid dose reaches 2.0g/kg per day,<br />

the 10% emulsion could be preferable over the<br />

20% emulsion. 10 The other role is as a source of<br />

essential fatty acids as well as long-chain PUFAs. It<br />

is well known that when preterm infants are maintained<br />

on lipid-free PN there is a rapid essential<br />

fatty acid deficiency. An essential fatty acid deficiency<br />

is known to interfere with normal lung<br />

surfactant synthesis, possibly further impairing<br />

pulmonary function in infants already at risk for<br />

respiratory problems. 60 Abnormalities in platelet<br />

function, which could have implications for clinical<br />

bleeding, have also been described. 61 Essential<br />

fatty acid deficiency is avoided by infusions of<br />

0.5–1.0g lipid per kg per day. 62 The importance of<br />

long-chain PUFAs for the development of the brain<br />

and the retina has also been recognized. Preterm<br />

infants are unable to form sufficient quantities of<br />

long-chain PUFAs from the respective precursor<br />

fatty acids (linoleic and α-linolenic acids) and thus<br />

depend on an exogenous source of long-chain<br />

PUFAs. 63,64 Intravenous lipid emulsions contain<br />

small amounts of these fatty acids as part of the egg<br />

phospholipid used as a stabilizer.<br />

More recently, new lipids emulsions containing a<br />

mixture of medium-chain and long-chain triglycerides<br />

(MCTs and LCTs), a mixture of MCTs, LCTs<br />

and fish oil, or based on olive and soybean oil, are<br />

receiving increasing attention. 65–70 All these solutions<br />

are designed to reduce the high linoleic and<br />

α-linolenic acid supplies provided by the classic<br />

soybean oil, improve the fatty acid composition of<br />

plasma lipids, reduce the risk of peroxidation and<br />

provide additional long-chain PUFAs with the use<br />

of fish oil. Infused MCTs are rapidly cleared, 65<br />

although studies of parenterally fed newborn<br />

infants have not confirmed a benefit of mixed<br />

MCT–long-chain fatty acid emulsions on protein<br />

accretion 66 and plasma fatty acid pattern. 67 The<br />

addition of fish oil to MCT and LCT solutions has<br />

not led to adverse effects, although a significant<br />

increase in plasma α-tocopherol and eicosapentaenoic<br />

acid in the high-density lipoprotein phospholipid<br />

68 was seen. The most promising appears<br />

to be the lipid emulsion based on olive and<br />

soybean oil. In preterm infants, it produces more<br />

physiological levels of linoleic and oleic acid and<br />

a better antioxidant status than the conventional<br />

soybean oil emulsion. 69,70<br />

Parenteral nutrition 629<br />

Concerns had been raised in some retrospective<br />

studies, about the potentially adverse effects of<br />

early lipid infusion in preterm infants on later<br />

outcomes, such as increased rates of chronic lung<br />

disease and mortality. 71,72 However, a meta-analysis<br />

of the five prospective controlled trials in lowbirth-weight<br />

infants on this issue has shown that<br />

early lipid infusion within the first 5 days of life is<br />

not associated with an increased risk of adverse<br />

outcomes. 73 There is no credible evidence of<br />

potentially adverse effects of intravenous lipid<br />

emulsions when these are properly used. Proper<br />

use includes slow and continuous infusion rate<br />

(3g/kg per day). 74 It<br />

has been suggested that triglyceride concentrations<br />

should be maintained at around 150–200mg/dl as<br />

an upper limit in VLBW infants, especially in<br />

unstable clinical situations.<br />

Considering that even a short delay in adding fat to<br />

the PN of preterm infants leads to biochemical<br />

essential fatty acid deficiency, infusion of lipids<br />

should commence within 24h of birth for infants<br />

>28 weeks’ gestation and >1000g. The dose<br />

should be increased, as tolerated, at a rate of 0.5g<br />

every 1–2 days to a maximum of 3g/kg per day. In<br />

all cases, the triglyceride infusion dose should be<br />

adjusted to maintain a serum lipid level not<br />

exceeding 200mg/dl. Infants


630<br />

Parenteral nutrition in premature infants<br />

Electrolytes, minerals and trace elements<br />

Sodium<br />

Sodium exists predominantly as an extracellular<br />

ion, and its requirements are unaltered by PN.<br />

The normal sodium requirement is assumed to be<br />

3mmol/kg per day. During the first week of life,<br />

infants of less than 28 weeks’ gestation often<br />

receive additional sodium supply from sources<br />

other than the parenteral solution, i.e. blood<br />

transfusion, bicarbonate or drugs, and lose more<br />

water than sodium. Therefore, to prevent hyperosmolar<br />

hypernatremia, some authors suggest<br />

very close monitoring of sodium intake during<br />

the first week of life, while others recommend the<br />

complete exclusion of sodium during the first few<br />

days in ELBW infants. Frequent monitoring and<br />

customization of serum sodium and water/<br />

sodium is mandatory especially in infants with<br />

congestive heart failure, acute renal failure or<br />

chronic diuretic therapy.<br />

Potassium<br />

The normal requirement for potassium for<br />

growing preterm infants is assumed to be<br />

1–2mmol/kg per day. It should be withheld in the<br />

first 3 days after birth in those who are extremely<br />

preterm because of the risk of developing nonoliguric<br />

hyperkalemia from immature distal<br />

tubular function. An adjustment of potassium<br />

intake is required if the infant is on diuretics or<br />

has poor urine output.<br />

Chloride<br />

The recommended intake is 2–3mmol/kg per day<br />

and the chloride maintenance intake must not be<br />

lower than 1mmol/kg per day.<br />

Calcium, phosphorus and magnesium<br />

Inadequate calcium and phosphorus intake has<br />

been associated with diminished bone mineralization<br />

in parenterally fed premature infants.<br />

This occurs when protein and energy are<br />

adequate for growth, but calcium and phosphorus<br />

are insufficient to sustain appropriate skeletal<br />

mineralization. 75–78<br />

Calcium and phosphorus cannot be provided<br />

through the same parenteral solution at concentrations<br />

needed to support in utero accretion, because<br />

of precipitation. The solubility of calcium and<br />

phosphorus in parenteral solutions depends on<br />

temperature, type and concentration of amino<br />

acid, dextrose concentration, pH of the calcium<br />

salt, the sequence of addition of calcium and phosphorus<br />

to the solution, calcium/phosphorus ratio<br />

and the presence of lipids. 79 More acidic pediatric<br />

amino acid solutions improve calcium and phosphorus<br />

solubility. With a range of fluid intake of<br />

120–150ml/kg per day, it is advisable to supply a<br />

calcium content of 50–60mg/dl (1.25–1.5mmol/l), a<br />

phosphorus content of 40–45mg/dl (1.25–1.5mmol/l)<br />

and a magnesium content of 5–7mg/dl (2.1–2.9mmol/l)<br />

corresponding to a calcium phosphorus ratio of 1.3:1<br />

by weight and 1:1 by molar ratio in the total PN<br />

solution. It must be underlined that this quantity<br />

of calcium provided by the parenteral route is<br />

about 60–75% of that deposited by the fetus during<br />

the last trimester of gestation (100–120mg/kg per<br />

day) but similar or higher than that obtained in<br />

enteral nutrition with the available preterm formulas.<br />

Administering sufficient amounts of calcium<br />

and phosphorus in PN solutions is no longer a<br />

problem in countries where organic phosphate<br />

preparations are available and the amounts of<br />

calcium and phosphorus available with PN could<br />

be theoretically higher than that absorbed using<br />

the enteral route. 80 However, information on bone<br />

mineralization of ELBW infants receiving what<br />

appears to be an adequate calcium and phosphate<br />

intake is still limited.<br />

The usual dose of magnesium is about 6 mg/kg per<br />

day (2.5mmol/kg per day), delivered as sulfate.<br />

This dose is rarely adjusted unless the infant has<br />

persistent hypocalcemia secondary to hypomagnesemia,<br />

or the infant has abnormally high magnesium<br />

levels due to maternal treatment with magnesium<br />

sulfate. Serum magnesium levels before<br />

magnesium supplementation with a PN solution<br />

should be checked in any small infant whose<br />

mother was treated for hypertension or preeclampsia.<br />

Trace elements<br />

Nine trace elements (or trace minerals) are nutritionally<br />

essential for humans: iron, zinc, copper,


selenium, molybdenum, chromium, manganese<br />

and iodine. Although trace elements make up a<br />

small fraction of the total mineral content of the<br />

human body, they play an important role in<br />

numerous metabolic pathways. The infant born<br />

prematurely is at increased risk of developing trace<br />

mineral deficiencies. Premature birth is associated<br />

with low stores at birth, because accretion of trace<br />

minerals takes place during the last trimester of<br />

pregnancy. Rapid postnatal growth, unknown<br />

requirements, and variable intake of trace minerals<br />

also put the preterm infant at risk for deficiencies.<br />

79<br />

During the early years of PN, it was thought that<br />

frequent plasma and/or blood transfusions<br />

provided the necessary trace minerals. Reports of<br />

zinc and copper deficiencies, even in infants who<br />

had received these transfusions, demonstrated the<br />

inadequacy of this approach and led to the availability<br />

of zinc and copper additives. The exact<br />

requirements of premature infants for most of the<br />

microminerals remain poorly defined because of<br />

the paucity of randomized controlled trials assessing<br />

their efficacy and safety. Recent studies have<br />

better delineated the range of suggested micromineral<br />

delivery to this population and Table 37.4<br />

shows the recommended enteral and parenteral<br />

intake of trace elements for preterm infants for the<br />

transition period from the first 2 weeks of life to<br />

the following stable and post-discharge period. 81<br />

Table 37.4 Recommended micromineral intakes for preterm infants on<br />

total parenteral nutrition 81<br />

Transitional period Stable/post-discharge<br />

(0–14 days) periods (>14 days)<br />

(µg/kg per day) (µg/kg per day)<br />

Iron 0 100–200<br />

Zinc 150 400<br />

Copper 0, 20 * 20 *<br />

Selenium 0–1.3 1.5–4.5<br />

Chromium 0–0.05 0.05–0.3<br />

Molybdenum 0 0.25–1.0 †<br />

Manganese 0, 0.75 * 1.0 *<br />

Iodine 0–1.0 1.0<br />

* Should be withheld when hepatic cholestasis is present<br />

† For long-term total parenteral nutrition only<br />

Parenteral nutrition 631<br />

Research concerning the parenteral requirements<br />

of these nutrients by infants is, of course, hindered<br />

by the difficulties in both measuring plasma<br />

concentrations of the nutrients using small<br />

volumes of plasma, and interpreting the physiological<br />

significance of plasma concentrations.<br />

Accurate studies on the retention of these nutrients<br />

are also notoriously difficult.<br />

Intravenous vitamins<br />

Table 37.5 shows the recommended parenteral<br />

intake of vitamins for preterm infants. These<br />

guidelines, suggested in the past years by different<br />

committees provide a theoretical recommendation<br />

to obtain the optimal intake which is, however,<br />

unachievable with current commercial preparations.<br />

The delivery of vitamins from parenteral<br />

solutions can be lower than intended. Fat-soluble<br />

vitamins may be adsorbed into the storage bag and<br />

delivering tubing and may alter when exposed to<br />

light, oxygen or heat. As much as 80% of available<br />

vitamin A and 30% of vitamins D and E are lost<br />

during administration due to adherence to tubing<br />

and photodegradation especially during phototherapy82<br />

.<br />

Therefore, significant destruction may occur<br />

within the newborn intensive care unit environment,<br />

where higher environmental temperatures<br />

and light levels are commonplace. Vitalipid ®


632<br />

Parenteral nutrition in premature infants<br />

Table 37.5 Recommended parenteral intake<br />

of vitamins for preterm infants (dose/kg per<br />

day) 81<br />

Consensus<br />

Vitamins recommendations<br />

Fat Soluble<br />

Vitamin A (IU) 700–1500<br />

with lung disease 1500–2800<br />

Vitamin D (IU) 40–160<br />

Vitamin E (IU) 3.5 (max = 7)<br />

Vitamin K (µg) 8–10 (300 at birth)<br />

Water soluble<br />

Vitamin B6 (µg) 150–200<br />

Vitamin B12 (µg) 0.3<br />

Vitamin C (mg) 15–25<br />

Biotin (µg) 5–8<br />

Folic acid (µg) 56<br />

Niacin (mg) 4–6.8<br />

Pantothenate (mg) 1–2<br />

Riboflavin (µg) 150–200<br />

Thiamin (µg) 200–350<br />

(Kabivitrum, Stockholm), a lipid preparation available<br />

in Europe, is an intravenous soybean oil<br />

emulsion that contains vitamins A, D, E and K,<br />

thus reducing adsorption into the plastic tubing.<br />

Further clinical studies are needed to assess the<br />

vitamin status of VLBW and ill infants who receive<br />

parenteral solutions for prolonged periods and to<br />

evaluate the effects of the peroxide load induced<br />

by photo-exposed intravenous multivitamin solutions.<br />

83<br />

Complications of parenteral nutrition<br />

The main complications of PN, as shown in Table<br />

37.6, can be metabolic, infective or catheterrelated.<br />

Several complications (e.g. electrolyte<br />

imbalance, hypoglycemia, hyperglycemia, hypo/<br />

hypercalcemia, hypophosphatemia) can be<br />

corrected and prevented by manipulating the<br />

components of the solution. Other potentially<br />

more serious complications (e.g. cholestasis) are<br />

less easily corrected.<br />

Table 37.6 Possible complications of<br />

parenteral nutrition<br />

Metabolic complications<br />

Cholestasis<br />

Electrolyte imbalance<br />

Essential fatty acid deficiency<br />

Hyperammonemia<br />

Hypertriglyceridemia<br />

Hypocalcemia/hypercalcemia<br />

Hypoglycemia/hyperglycemia<br />

Metabolic acidosis<br />

Metabolic bone disease<br />

Prerenal nitrogen<br />

Vitamin, mineral and trace elements deficiencies<br />

Infectious complications<br />

Bacterial<br />

Fungal<br />

Catheter-related complications<br />

Vascular perforation<br />

Embolism<br />

Improper insertion resulting in extravasation, pneumothorax,<br />

hemorrhage, pleural effusion or pericardial<br />

tamponade<br />

Local irritation, infiltration<br />

Thromboses<br />

Cholestasis<br />

Cholestasis is a well-known complication associated<br />

with PN. 84,85 The initial lesion seen histologically<br />

is intracellular and intracanicular cholestasis,<br />

followed by portal inflammation. With<br />

prolonged administration, portal fibrosis and ultimately<br />

cirrhosis may develop. Cholestasis induced<br />

by PN is associated, initially with the elevation of<br />

serum bile acids and γ-glutamyl-transpeptidase<br />

(GGTP) and then with direct hyperbilirubinemia<br />

and jaundice. Elevation of hepatic transaminases,<br />

SGOT and SGPT is a late finding. Premature<br />

infants receiving PN for prolonged periods are at<br />

high risk of developing cholestasis. 86<br />

Incidence of cholestasis represents about 50% in<br />

infants with a birth weight


een suggested that hepatic dysfunction may be<br />

caused by the overgrowth of the intestinal flora in<br />

static gut. Fasting is also linked to development of<br />

cholestasis. This complication is less frequent<br />

when enteral feeding, even hypocaloric, is started.<br />

Abnormal amino acid pattern and taurine deficiency<br />

in some amino acid mixtures used as part of<br />

PN have been considered as possible etiological<br />

factors, but currently used solutions contain<br />

amino acids (including taurine), designed to maintain<br />

normal infant serum amino acid profiles. 89<br />

Usually cholestasis resolves with discontinuation<br />

of PN but there are some reports of advanced liver<br />

disease in infants parenterally fed for several<br />

months.<br />

Several measures have been suggested to prevent<br />

cholestasis induced by total PN such as: use of<br />

minimal enteral feeding, early introduction of<br />

enteral feeds to stimulate bile flow, use of<br />

balanced parenteral solutions, and reducing lipid<br />

infusion. In newborns and young infants with<br />

total PN-associated cholestasis who require<br />

continued PN, limiting the protein intake to 2g/kg<br />

per day may help minimize amino acid-related<br />

hepatic toxicity. Ursodeoxycholic acid treatment<br />

seems to improve the course of PN-associated<br />

cholestasis not only in children and adults but<br />

also in premature infants, leading to an early<br />

sustained decrease in bilirubin levels after 2<br />

weeks of therapy. 90<br />

Infection<br />

Bacterial and/or fungal infection is another complication<br />

of PN. The most common isolated agents are<br />

Staphylococcus epidermidis, Staphylococcus aureus<br />

and Candida albicans. The incidence of sepsis as a<br />

Table 37.7 Precautions taken to minimize the risk of sepsis<br />

Complications of parenteral nutrition 633<br />

complication of PN increases as gestational age<br />

decreases and the duration of PN increases. Even<br />

if infection is often a catheter-related complication<br />

it seems that the composition of solutions<br />

may predispose to sepsis. Intravenous lipid have<br />

been associated with coagulase-negative staphylococcal<br />

bacteremia and Malessezia furfur<br />

fungemia. 91 Table 37.7 shows the main precautions<br />

that should be taken to minimize the risk of<br />

sepsis.<br />

Catheter-related complications<br />

Preparation of individual aliquots of parenteral nutrition solutions in the pharmacy<br />

Manipulations carried out in the ward to be avoided<br />

Central venous silastic catheters must be placed under strict aseptic conditions<br />

Skin exit side for catheter placed in area which can be meticulously cleansed<br />

Proper care of the site and all the connectors and essential tubings<br />

Addition of heparin (1 unit/ml) to the infusate for peripheral/central venous catheters<br />

The main complications associated with venous<br />

catheter are usually the result of: improper insertion<br />

or placement; bacterial or fungal colonization<br />

of the catheter; and vessel irritation and/or thrombosis.<br />

Peripheral Teflon ® catheters are prone to<br />

infiltration in a short time and to be colonized<br />

with bacteria at a rate of over 30% when they have<br />

been in place for more than 3 days.<br />

Central venous catheters have a lower incidence<br />

of infiltration and deliver higher concentrations of<br />

the solution, but can be associated with severe<br />

complications. Broviac ® catheters are associated<br />

with a higher incidence of infection (5–60%)<br />

and/or thrombosis in the neonatal period. At<br />

present, with the small silastic catheters introduced<br />

percutaneously, the incidence of sepsis<br />

and/or thrombosis is lower than with Broviac<br />

catheters. Thrombosis in these small-bore<br />

catheters can be minimized by adding heparin to<br />

the solution. In order to obtain a correct intravenous<br />

nutrition and avoid the high risks of<br />

complications, the patient must be closely monitored.<br />

Table 37.8 shows the main parameters that<br />

need to be checked.


634<br />

Parenteral nutrition in premature infants<br />

Table 37.8 Monitoring during parenteral nutrition<br />

Measurement of body weight daily and body length and head circumference weekly<br />

Initially, during grading up of parenteral nutrients or during periods of metabolic instability:<br />

strict fluid balance<br />

6–12 hourly urine/blood glucose<br />

daily plasma sodium, potassium, calcium, urea and acid–base<br />

When on full parenteral nutrition and during metabolic steady state:<br />

strict fluid balance<br />

12–24 hourly urine/blood glucose<br />

plasma sodium, potassium, calcium, urea and acid–base once/twice weekly<br />

Plasma magnesium, phosphorus, alkaline phosphatase, albumin, transaminases and bilirubin weekly<br />

Plasma triglycerides, amino acids, trace elements and ammonia not usually routinely monitored<br />

Screening for infection or coagulation defects as indicated<br />

Practical aspects of parenteral<br />

nutrition in VLBW infants<br />

Tailored or standard parenteral solutions<br />

Parenteral solutions can be prescribed using either<br />

of two formats: tailored or standard. 92 Tailored<br />

solutions are formulated specifically to meet the<br />

daily nutritional requirements of the individual<br />

patient, whereas standard solutions are designed<br />

to provide a formulation that meets most of the<br />

nutritional needs of the stable biochemical and<br />

metabolic parameters. Both of these methods have<br />

advantages and disadvantages associated with<br />

their use.<br />

Tailored solutions are based on the principle that<br />

no single parenteral regimen can be ideal for all<br />

patients, for a wide variety of pathological<br />

processes, all age groups, or for the same patient<br />

during a single disease. The main advantage of<br />

tailored solutions is flexibility. Each solution is<br />

formulated for an individual patient and can be<br />

modified when the patient’s nutritional needs and<br />

metabolic, electrolyte or clinical status changes.<br />

The disadvantage of these solutions is linked to the<br />

time involved in calculation and label preparation,<br />

which today is nevertheless diminished with the<br />

use of specific computer programs. These solutions<br />

should be prepared with strict aseptic tech-<br />

niques, possibly in the pharmacy, not in the ward,<br />

and stored in a refrigerator at 4°C. The solutions<br />

thus prepared are stable for 96h and should be<br />

allowed to reach room temperature slowly and not<br />

warmed before infusion.<br />

Standard solutions contain fixed amounts of each<br />

component per unit volume. In some hospitals<br />

there are several types of fixed solutions to cover<br />

the nutritional requirements of premature infants<br />

more adequately. The advantages of these solutions<br />

is that they include all the essential nutrients<br />

in fixed amounts, which eliminates the chances of<br />

inadvertent omission or overload. The major<br />

disadvantage of standard solutions is their lack of<br />

patient specificity and the need of minimal adjustment<br />

particularly during the first days of life. 92<br />

Nutrient intake<br />

Table 37.9 shows the composition of a ready-to-use<br />

parenteral solution for VLBW infants and Table<br />

37.10 the daily nutrient intake (kg/day) given by<br />

total PN according to the new practice of ‘aggressive<br />

nutrition’ proposed to minimize the interruption<br />

of nutrient intake induced by premature birth.<br />

In any case, nutrient intakes are always indicative<br />

and have to be modified according to each patient,<br />

his/her clinical picture, biochemical data and<br />

tolerance to nutrient intake. Thus, this standard<br />

solution could be diluted with free water accord-


ing to fluid requirement and natrium chloride<br />

could be adapted after a few days of life.<br />

The following suggestions could be useful in the<br />

management of a parenterally fed VLBW infant.<br />

(1) In the first days of life fluid intake should be<br />

increased if daily weight loss is >5%, total<br />

weight loss is >12–15%, serum sodium is<br />

Table 37.9 Composition of a ready-to-use<br />

binary parenteral solution (per 100 ml)<br />

adapted for very-low-birth-weight infants<br />

Glucose (g) 12.0<br />

Amino acid (g) 2.7<br />

Calcium (mg) 56<br />

Phosphorus (mg) 43<br />

Magnesium (mg) 4<br />

Sodium (mmol) 1.6<br />

Potassium (mmol) 1.5<br />

Chloride (mmol) 2.0<br />

Energy (kcal)* 55<br />

* Energy (kcal) = amino acids (g)x3.75+glucose x 3.75<br />

Practical aspects of parenteral nutrition in VLBW infants 635<br />

>150mmol/l, urinary osmolality is<br />

>350mOsm/l and if the infant is under<br />

phototherapy or managed in a radian incubator.<br />

In contrast, fluid intakes should be<br />

reduced if daily weight loss is


636<br />

Parenteral nutrition in premature infants<br />

energy intakes reach about 70kcal/kg per day<br />

and there is no enteral protein intake, the<br />

dose should be increased progressively to<br />

3.5–4.4g/kg per day.<br />

(4) Although many neonatal intensive care units<br />

start lipid infusion only after the first few<br />

days, it seems logical to start parenteral lipids<br />

REFERENCES<br />

1. Jobe A. Pulmonary surfactant therapy. N Engl J Med<br />

1993; 328: 861–868.<br />

2. NIH Consensus Conference. Effect of corticosteroids for<br />

fetal maturation on perinatal outcomes. J Assoc Med<br />

Assoc 1995; 273: 413–417.<br />

3. Hauth JC, Goldenberg RL, Andrews WW et al. Reduced<br />

incidence of preterm delivery with metronidazole and<br />

erythromycin in women with bacterial vaginosis. N Engl<br />

J Med 1995; 333: 1732–1736.<br />

4. Lucas A. Programming by early nutrition in man. In<br />

Bock GR, Whelan J, eds. The Childhood Environment<br />

and Adult Disease. Ciba Foundation Symposium 156.<br />

Chichester: Wiley 1991: 38–55.<br />

5. American Academy of Pediatrics. Nutritional needs of<br />

preterm infants. In Kleinman RE, ed. Pediatric Nutrition<br />

Handbook, 4th edn. Elk Grove Village: American<br />

Academy of Pediatrics 1998: 55–87.<br />

6. Committee on Nutrition of the preterm infant, European<br />

Society for Paediatric Gastroenterology and Nutrition.<br />

Nutrition and feeding of preterm infants. Acta Paediatr<br />

Scand 1987; 336 (Suppl): 1–14.<br />

7. Nutrition Committee CPS. Nutrient needs and feeding<br />

of premature infants. Can Med Assoc J 1995; 152:<br />

1765–1785.<br />

8. Embleton NE, Pang N, Cooke RJ. Postnatal malnutrition<br />

and growth retardation: an inevitable consequence of<br />

current recommendations in preterm infants? Pediatrics<br />

2001; 107: 270–273.<br />

9. Rigo J, De Curtis M, Pieltain C. Nutritional assessment<br />

and body composition of preterm infants. Semin<br />

Neonatol 2002; 6: 383–391.<br />

10. Ziegler EE, Thureen PJ, Carlson SJ. Aggressive nutrition<br />

of the very low birthweight infant. Clin Perinatol 2002;<br />

29: 225–244.<br />

11. Wilson DC, Cairns P, Halliday HL et al. Randomised<br />

controlled trial of an aggressive nutritional regimen in<br />

sick very low birthweight infants. Arch Dis Child 1997;<br />

77: 4F–11F.<br />

12. Rigo J. Protein, amino acid and other nitrogen<br />

compounds. In Tsang RC, Uauy R, Koletzko B, Zlotkin SH,<br />

Hansen JW. Nutritional Needs for the Preterm Infant. Digital<br />

Educational Publishing, Ohio Cincinnati, 2004: in press.<br />

13. Cooke RJ, Rigo J, Embleton ND, Ziegler EE. Nutrient<br />

balance and metabolic status in preterm infants fed two<br />

levels of dietary protein. Pediatr Res 2002; 4: 318A.<br />

14. Ehrenkranz RA, Younes N, Lemons JA et al.<br />

Longitudinal growth of hospitalized very low birth<br />

weight infants. Pediatrics 1999; 104: 280–289.<br />

during the first day in order to avoid a<br />

prolonged interruption of essential fatty acids<br />

and long-chain PUFAs. The starting dose of<br />

0.5–1g/kg per day should gradually be<br />

increased to 3–3.5g/kg per day. The lipid infusion<br />

should be adjusted to maintain a serum<br />

lipid level


30. Thureen PJ, Melara D, Fennessey V et al. Effect of low<br />

versus high intravenous amino acid intake on very low<br />

birth weight infants in the early neonatal period. Pediatr<br />

Res 2003; 53: 24–32.<br />

31. Rigo J. Azote et acides aminés. In Ricour C, Ghisolfi J,<br />

Putet G, Goulet O. eds. Traité de Nutrition Pédiatrique,<br />

Paris: Maloine, 1993: 852–866.<br />

32. Rigo J, Senterre J. Significance of plasma amino acid<br />

pattern in preterm infants. Biol Neonate 1987; 52: 41–49.<br />

33. Duffy B, Gunn T, Collinge J et al, The effect of varying<br />

protein quality and energy intake on the nitrogen<br />

metabolism of parenterally fed very low birthweight<br />

(


638<br />

Parenteral nutrition in premature infants<br />

mixture of medium chain, (MCT), soybean oil (LCT),<br />

and fish oil (FO). Pediatr Res 2002; 51: 1853A.<br />

69. Putet G. Lipid metabolism of the micropremie Clin<br />

Perinatol 2000; 27: 57–69.<br />

70. Gobel Y, Koletzko B, Bohles HJ et al, Parenteral fat emulsions<br />

based on olive and soybean oils: a randomised<br />

clinical trial in preterm infants. J Pediatr Gastroenterol<br />

Nutr 2003; 37:161–167.<br />

71. Hammerman C, Aramburo MJ. Decreased lipid intake<br />

reduces morbidity in sick premature neonates. J Pediatr<br />

1988; 113: 1083–1088.<br />

72. Cooke RWI. Factors associated with chronic lung disease<br />

in preterm infants. Arch Dis Child 1991; 66: 776–779.<br />

73. Wilson DC, Fox GF, Ohlsson A. Meta-analyses of effects<br />

of early or late introduction of intravenous lipids to<br />

preterm infants on mortality and chronic lung disease<br />

[abstract]. J Pediatr Gastroenterol Nutr 1998; 26: 599.<br />

74. Kao LC, Cheng MH, Warburton D. Triglycerides, free<br />

fatty acids, free fatty acids/albumin molar ratio, and<br />

cholesterol levels in serum of neonates receiving longterm<br />

lipid infusions: controlled trial of continuous and<br />

intermittent regimens. J Pediatr 1984; 104: 429–435.<br />

75. Rigo J, De Curtis M, Salle BL et al. Bone mineral metabolism<br />

in the micropremie. Clin Perinatol 2000; 27:<br />

147–170.<br />

76. Rigo J, De Curtis M, Nyamugabo K et al. Premature<br />

Bone. In Tzang and Bonjour, eds. Nutrition and Bone<br />

Development. 42° Nestlé Nutrition Workshop. Puebla,<br />

1997; 41: 83–97.<br />

77. Rigo J, De Curtis M, Nyamugabo K et al. Whole body<br />

calcium content in term and preterm neonates. Eur J<br />

Pediatr 1998; 157: 259–264.<br />

78. Rigo J, Nyamugabo K, De Curtis M et al. Bone mineralization<br />

during the first year of life. In Bindels JG, Goedhardt<br />

AC, Visser HKA, eds. Recent Development in Infant<br />

Nutrition. Tenth Nutricia Simposium Scheveningen, The<br />

Hague, The Nederlands, 1995: 98–111.<br />

79. Aggett PJ. Trace elements of the micropremie. Clin<br />

Perinatol 2000; 27: 119–129.<br />

80. Prinzivalli M, Ceccarelli S. Sodium D-fructose-1,6diphosphate<br />

vs. sodium monohydrogen phosphate in<br />

total parenteral nutrition: a comparative in vitro assess-<br />

ment of calcium phosphate compatibility. J Parenter<br />

Enteral Nutr 1999; 23: 326–332.<br />

81. Raghavendra R, Georgieff M. Microminerals. In Tsang<br />

RC, Uauy R, Koletzko B et al, eds. Nutritional Needs for<br />

the Preterm Infant. Cincinnati, Ohio: Digital Educational<br />

Publishing 2004: in press.<br />

82. Gilles J, Jones G, Penchaarz P. Delivery of vitamins A, D<br />

and E in parenteral nutrition solutions. J Parenter<br />

Enteral Nutr 1983; 7: 11–14.<br />

83. Laborie S, Lavoie JC, Chessex P. Increased urinary<br />

peroxides in newborn infants receiving parenteral nutrition<br />

exposed to light. J Pediatr 2000; 136: 628–632.<br />

84. Merritt RJ. Cholestasis associated with total parenteral<br />

nutrition. J Pediatr Gastroenterol Nutr 1986; 5: 9–22.<br />

85. Whitington P. Cholestasis associated with total parenteral<br />

nutrition in infants. Hepatology 1986; 5: 693–696.<br />

86. Beale EF, Nelson RM, Bucciarelli RL et al. Intrahepatic<br />

cholestasis associated with parenteral nutrition in<br />

premature infants. Pediatrics 1979; 64: 342–347.<br />

87. Beath SV, Davies P, Papadopoulou A et al. Parenteral<br />

nutrition-related cholestasis in postsurgical neonates:<br />

multivariate analysis of risk factors. J Pediatr Surg 1996;<br />

31: 604–606.<br />

88. Brown MR, Thunberg BJ, Golub L et al. Decreased<br />

cholestasis with enteral instead of intravenous protein<br />

in the very low-birth-weight infant. J Pediatr<br />

Gastroenterol Nutr 1989; 9: 21–27.<br />

89. Heird WC, Dell RB, Helms RA et al. Amino acid mixture<br />

designed to maintain normal plasma amino acid<br />

patterns in infants and children requiring parenteral<br />

nutrition. Pediatrics 1987; 80: 401–408.<br />

90. Levine A, Maayan A, Shamir R et al. Parenteral nutrition-associated<br />

cholestasis in preterm neonates: evaluation<br />

of ursodeoxycholic acid treatment. J Pediatr<br />

Endocrinol Metab 1999; 12: 549–553.<br />

91. Long JB, Keyserling HL. Catheter-related infection in<br />

infants due to an unusual yeast-Malessezia furfur.<br />

Pediatrics 1985; 76: 896–900.<br />

92. Poole RL, Kerner JA. Practical steps in prescribing intravenous<br />

feeding. In Yu VYH, MacMahon RA, eds.<br />

Intravenous Feeding of the Neonate. Edward Arnold<br />

1992: 259–264.


38<br />

Introduction<br />

Approach to gastrointestinal<br />

bleeding<br />

Samy Cadranel and Michèle Scaillon<br />

Gastrointestinal (GI) bleeding, an uncommon<br />

phenomenon in infants and children, is most often<br />

felt, by parents and caregivers, as very dramatic,<br />

needing emergency treatment, regardless of the<br />

importance of the bleeding. Adequate management<br />

of the situation – which can obviously be<br />

life-threatening – requires experience, common<br />

sense and good co-ordination from the nursing<br />

staff and physicians in charge.<br />

Epidemiology<br />

In the general population, bleeding from the upper<br />

GI tract occurs with a prevalence of 100 in 100000<br />

adults per year, whereas lower GI bleeding is five<br />

times less frequent. The vast majority of hospitalizations<br />

occur in patients with concurrent illness<br />

and advanced age. 1 The overall incidence seems<br />

lower in Europe, for instance 45 in 100000 as<br />

reported in a Dutch study. 2 In childhood, bleeding<br />

is less frequent but potentially serious, and the<br />

proportion of upper and lower intestinal bleeding<br />

appears to be roughly similar to that reported in<br />

the adult literature. 3 However, the etiology can be<br />

different, largely depending on the age of the child.<br />

There are no reliable quantitative data about the<br />

epidemiology of GI bleeding in ambulatory pediatric<br />

patients. 4 Children are usually in better<br />

condition, with the notable exception of the<br />

newborn period, the critically ill child in an intensive<br />

care unit (ICU) and children with portal<br />

hypertension and end-stage liver failure. 5 Many<br />

medications used in adults have not been standardized<br />

in children and the devices available for<br />

emergency endoscopic treatment may sometimes<br />

be too large for the child’s limited GI tract caliber.<br />

Despite important technical and training improvements<br />

in the management of acute bleeding in the<br />

adult patient, occurring during the past 30 years,<br />

the morbidity and mortality has remained unchanged,<br />

at around 6–7%. 6<br />

This finding is probably biased, reflecting only a<br />

considerable decrease of the less dramatic bleedings<br />

related to relapsing gastroduodenal ulcers due<br />

to the active eradication of <strong>Helicobacter</strong> <strong>pylori</strong>.<br />

During the same period, the systematic use of<br />

medications that control gastric acidity (such as<br />

anti-H2 blockers or protein pump inhibitors (PPIs))<br />

has succeeded in considerably reducing acute<br />

bleeding episodes in ICU patients (adults and children<br />

alike). 7<br />

Presentations and definitions<br />

GI hemorrhage may present in different fashions.<br />

Acute GI bleeding is obvious GI blood loss, and is<br />

often associated with hemodynamic compromise.<br />

Gross GI bleeding that occurs intermittently is<br />

defined as acute-recurrent bleeding. Occult blood<br />

loss is a chronic source of GI blood loss usually<br />

detected by either a positive fecal occult blood test<br />

or iron deficiency anemia.<br />

Regurgitations or vomiting of large quantities of<br />

red or digested brownish ‘coffee ground’ blood by<br />

mouth is called ‘hematemesis’; the source of the<br />

bleeding is located between the esophagus and the<br />

ligament of Treitz. This is not always a sign of GI<br />

bleeding, since it may sometimes be confused with<br />

‘hemoptysia’ which is triggered by cough, or with<br />

a bleeding from a nasopharyngeal origin. The<br />

brownish aspect of regurgitations can also be due<br />

to other causes such as cola or coffee drinks, sometimes<br />

misinterpreted by the family as blood.<br />

639


640<br />

Approach to gastrointestinal bleeding<br />

‘Melena’ describes black, tarry, foul-smelling<br />

stool, and its presence usually indicates an upper<br />

intestinal source. Rarely, a proximal colonic<br />

bleeding may present as melena. Melena associated<br />

with hematemesis again implies a voluminous<br />

upper GI source.<br />

‘Hematochezia’ (red blood passed per rectum)<br />

usually indicates a lower GI source, although<br />

voluminous blood loss from any location may<br />

present in this manner. In the unstable patient,<br />

presumption of an upper GI source dictates<br />

management, regardless of the color of blood per<br />

rectum.<br />

Streaks of fresh blood on the feces or after defecation<br />

are not a sign of a severe hemorrhage but<br />

of a benign light bleeding often overestimated by<br />

the family. In the pediatric patient, ‘occult’ bleeding<br />

is usually discovered during a work-up of<br />

side-ropenic anemia together with clinical manifestations<br />

of chronic blood loss: fatigue, pallor or<br />

lightheadedness.<br />

Practical approach<br />

Extensive and comprehensive descriptions of all<br />

possible situations generating GI bleeding (some<br />

of them very rare in childhood) and of the different<br />

causes, diagnostic procedures and treatments<br />

have been published elsewhere and especially in<br />

classical textbooks. 8–11 The purpose of the<br />

present chapter is to focus on a practical approach<br />

to GI bleeding in children and we have<br />

arbitrarily organized it in seven gradual steps<br />

(Figure 38.1).<br />

Is it a life-threatening emergency due to the<br />

blood loss?<br />

The symptoms such as pallor, agitation, sweating,<br />

tachycardia or low blood pressure (this is a severe<br />

but late complication) are usually very clear and<br />

should be evaluated accurately in order to initiate<br />

adequate resuscitation measures. Treating or<br />

preventing hypovolemia is the first step in stabilization.<br />

A secure large-bore intravenous access –<br />

or a central line for the monitoring of central<br />

venous pressure – is mandatory, since shock can<br />

develop at any time if uncontrolled bleeding<br />

1. Vital emergency? 2. Ongoing emergency?<br />

No Yes<br />

No<br />

Emergency measures<br />

Intravenous access<br />

Hemodynamic stabilization<br />

Clotting factors<br />

Cross-matching<br />

Laboratory<br />

3. Is it digestive?<br />

4. Upper or lower?<br />

5. Etiology and underlying factors<br />

6. Investigations<br />

7. Management and prevention<br />

Figure 38.1 Practical approach to gastrointestinal<br />

bleeding in children.<br />

persists. Blood should be drawn for blood typing<br />

and cross-matching. At this stage, extra blood<br />

samples for hematocrit, hemoglobin, coagulation,<br />

and red and white blood cell and platelet counts<br />

are necessary. A low mean corpuscular volume<br />

(MCV) suggests a chronic blood loss and an<br />

elevated blood urea nitrogen (BUN) due to the<br />

absorption of intestinal blood points mainly to<br />

upper GI bleeding. 12,13 It is also advisable, whenever<br />

possible, to initiate investigations for etiological<br />

factors (such as liver function tests) by<br />

drawing extra blood samples. Management by<br />

fluid and/or colloid infusion can then be started.<br />

Once the stabilization has been obtained these<br />

children must be admitted to an ICU, ideally<br />

connected with a pediatric surgery department.<br />

Is the bleeding still ongoing?<br />

Relevant familial or personal surgical or medical<br />

histories should be taken as soon as possible.<br />

Every effort should be made to obtain a complete<br />

list of all medications administered to the child.<br />

This may prove difficult to obtain from upset


parents: it is strongly recommended to use a<br />

locally adapted checklist of drugs, which should<br />

be available and ready in the emergency room.<br />

Invasive monitoring is not always necessary and<br />

should be reserved for such situations where an<br />

ongoing bleeding is obvious or whenever history<br />

or physical examination suggests a potential for<br />

re-bleeding. Early insertion of a large-bore nasogastric<br />

tube is helpful for assessing possible<br />

continuing bleeding from the upper GI tract. We<br />

use a double-lumen tube (e.g. Salem sump TM ,<br />

Sherwood Medical, St Louis, MO, USA), which<br />

enables decompression, aspiration and also<br />

lavage using saline as preparation for a diagnostic<br />

endoscopic procedure. Attempts to stop an<br />

ongoing bleeding with iced saline lavage, a classical<br />

technique 14,15 once considered as helpful is<br />

no longer recommended, since it is rarely useful<br />

and potentially harmful. 16–18<br />

Is it a gastrointestinal tract bleeding?<br />

Hemoptysis is rare in children and usually associated<br />

with respiratory symptoms. Blood swallowed<br />

during an unrecognized epistaxis can mimic GI<br />

bleeding and present as hematemesis but also as<br />

melena. Previous episodes of epistaxis should be<br />

recorded. A rapid examination of the nasal cavity<br />

can easily diagnose the source of the bleeding.<br />

Not all vomiting or stools stained in red are blood.<br />

Additives coloring food, drink mixes or medicines<br />

can be mistaken for blood. Tomatoes, beets, cherries<br />

and cranberries may look like blood when<br />

vomited or passed per rectum by a child with diarrhea.<br />

Bismuth, iron supplements, spinach and<br />

dark chocolate can give a dark coloration to stools,<br />

which may be mistaken for melena. The very dark<br />

feces of constipated children are usually easy to<br />

distinguish from melena. Diapers can acquire a<br />

pink discoloration if some time elapses prior to<br />

their disposal.<br />

The testing for occult blood is usually performed<br />

on a stool specimen by a Guaiac reaction. Guaiac<br />

is a colorless compound that turns blue in contact<br />

with substances with peroxidase activity (e.g.<br />

hemoglobin) and hydrogen peroxide. This test is<br />

easy, inexpensive and convenient, but the reaction<br />

is not specific for blood. Many substances with<br />

peroxidase activity may yield a false-positive<br />

Practical approach 641<br />

result. Ingested red meat, certain fruits and vegetables<br />

such as cantaloupes, radishes, bean sprouts,<br />

cauliflower, broccoli and grapes have enough<br />

peroxidase activity to cause a positive reaction.<br />

Oral iron preparations may also cause falsepositive<br />

reactions.<br />

In the newborn, suspected GI bleeding can be due<br />

to swallowed maternal blood and should be ruled<br />

out by the Apt test. The test is based on the denaturation,<br />

in an alkaline milieu, of adult hemoglobin<br />

to a yellow-brown solution of alkaline globin<br />

hematin whereas fetal hemoglobin, resists and is<br />

colored to a pink solution.<br />

Is it an upper or a lower gastrointestinal<br />

bleeding?<br />

Logically, hematemesis or melena indicates bleeding<br />

from the upper GI tract, while hematochezia<br />

indicates bleeding from a colonic source. However,<br />

this simple distinction can be confusing in the<br />

newborn and in infants. The transit time can be<br />

very quick and undigested red blood hematochezia<br />

(instead of digested melena) may be the only<br />

symptom of a hemorrhage occurring in the upper<br />

segment of the GI tract.<br />

Similarly, in some instances, melena is the only<br />

manifestation of colonic blood trapped during<br />

sufficient time to permit the degradation of hemoglobin<br />

by the intestinal flora. Small-intestinal<br />

bleeding manifests as either melena or hematochezia.<br />

Simultaneous mixed digested black and<br />

undigested fresh red blood suggests a Meckel’s<br />

diverticulum or a large upper GI bleeding.<br />

The nasogastric tube, used as a tool for monitoring<br />

the continuity of bleeding, can also be used as a<br />

first-step investigation to distinguish between an<br />

upper or a lower GI bleeding origin; blood-stained<br />

gastric fluid indicates an upper GI source.<br />

However, a clear or bilious stained fluid does not<br />

exclude a duodenal origin of the bleeding.<br />

Guaiac testing for occult bleeding from gastric<br />

aspirates is not reliable. Needless to say, a<br />

complete and oriented history taking can provide<br />

useful clues, such as recent medication with<br />

non-steroidal anti-inflammatory drugs (NSAIDs),<br />

previous ulcer, familial history of polyposis, or<br />

liver disease. Associated symptoms such as bloody


642<br />

Approach to gastrointestinal bleeding<br />

mucous diarrhea suggest a colonic or terminal<br />

small-bowel source.<br />

A careful physical examination looking for signs of<br />

portal hypertension, splenic or liver enlargement,<br />

cutaneous stigma of Peutz–Jeghers polyposis or<br />

syndromes associated with vascular abnormalities<br />

is essential. A thorough examination of the oral<br />

and nasal cavity as well as the anal region can rule<br />

out simple diagnoses.<br />

What are the etiology and underlying factors?<br />

GI bleeding occurs at all ages with similar etiologies<br />

and mechanisms. However, the frequency and<br />

presentation varies according to age (Table 38.1)<br />

and underlying factors.<br />

<strong>Neonatal</strong> period<br />

Bleeding from coagulation abnormalities due to<br />

vitamin K deficiency is becoming rare since this<br />

condition is corrected by systematic vitamin K<br />

supplementation at birth. 19,20<br />

The most frequent cause of upper GI bleeding in<br />

the neonatal period is the poorly understood<br />

‘neonatal esophagogastritis’ which affects acutely<br />

distressed newborns. The course is most often<br />

benign and self-limited. The availability of miniaturized<br />

fiberscopes has enabled the recognition of<br />

this condition as a cause of GI bleeding in a series<br />

of 14 out of 32 newborns published in 1989. 21 The<br />

same group reviewed their series of endoscopies<br />

performed in the neonatal period; 158 out of 219<br />

newborns presented with esophagitis. 22 Other<br />

authors reported findings in 17 full-term newborns<br />

and attributed the lesions to a traumatic origin,<br />

because of the distribution of the lesions (more<br />

severe in the upper esophagus), of the early onset<br />

(almost at birth) and the very rapid healing. 23<br />

Finnish authors drew attention to the presence of<br />

gastric lesions, highly prevalent in preterm infants<br />

in the ICU 24 and claimed that mechanical ventilation<br />

has the main risk factor, but factors included<br />

also the mode of delivery and hypotension after<br />

birth, which increase the risk of stress-induced<br />

gastric lesions. 25 Other French groups insisted on<br />

the extension of the lesions involving the esopha-<br />

Table 38.1 Main sources of gastrointestinal (GI) bleeding according to age and frequency<br />

Newborn 1 month to 2 years 2–16 years<br />

Upper GI bleeding<br />

Maternal blood (Apt test) esophagitis esophagitis<br />

Coagulation factors gastritis gastritis<br />

Esophagitis acute ulcer acute ulcer<br />

Gastritis Mallory–Weiss syndrome Mallory–Weiss syndrome<br />

Duodenitis varices varices<br />

Vascular malformation vascular abnormalities vascular abnormalities<br />

GI duplications GI duplications<br />

Lower GI bleeding<br />

Maternal blood (Apt test) anal fissures anal fissures<br />

Coagulopathy infectious colitis infectious colitis<br />

NEC vascular anomalies vascular anomalies<br />

Other enterocolitides allergic colitis polyp(s)<br />

Volvulus Meckel’s diverticulum IBD<br />

vasculitis<br />

Henoch–Schönlein purpura<br />

hemolytic uremic syndrome<br />

hemorrhoids<br />

IBD, inflammatory bowel disease


gus and the stomach, or the stomach and the<br />

duodenum, or the whole upper GI tract. 26<br />

A case–control study concluded that there was a<br />

possible protection by breast feeding. 27 A recent<br />

case–control multicenter study in France pointed<br />

out the possible role of antacid drugs used by<br />

mothers during the last month of pregnancy, and<br />

emphasized the possible protective role of early<br />

breast feeding against severe lesions. 28<br />

In the neonatal period, rectal bleeding (sometimes<br />

from an upper GI source) is infrequent and differs<br />

little from rectal bleeding in older infants.<br />

However, necrotizing enterocolitis occurs early,<br />

especially in the preterm infant and after the first<br />

oral feedings. 29 Besides bloody stools, symptomatology<br />

includes abdominal distension, decreased<br />

activity and feeding difficulties. Radiological signs<br />

such as pneumatosis intestinalis and air in the<br />

biliary system are pathognomonic. The condition<br />

is severe and may result in sepsis.<br />

Infancy<br />

In infants, esophagitis due to gastroesophageal<br />

reflux disease is the most frequent cause of upper<br />

GI bleeding. In addition, although infrequent, it is<br />

sometimes an early complication of hypertrophic<br />

<strong>pylori</strong>c stenosis. 30 Gastroduodenal ulcers are rare,<br />

since <strong>Helicobacter</strong> <strong>pylori</strong> infection, an etiological<br />

factor in peptic ulceration, is not a common<br />

finding in this age group, at least in industrialized<br />

regions. Furthermore, interferon (IFN)-γ secretion<br />

in the stomach of H. <strong>pylori</strong>-infected patients is<br />

lower in children than in adults, and this could<br />

protect children from development of severe<br />

gastroduodenal diseases such as ulcer disease. 31<br />

Rectal red blood mixed with mucus and loose<br />

stools suggests an infectious cause. Viral infection<br />

is seldom the cause of rectal bleeding. However,<br />

many known pathogenic bacteria such as<br />

Salmonella, Shigella, Campylobacter jejuni and<br />

some strains of Escherichia coli can cause acute<br />

rectal bleeding through invasive colonic lesions.<br />

The role of Clostridium difficile is more<br />

debated 32,33 whereas we have observed Yersinia<br />

enterocolitica-associated colonic lesions that<br />

closely resemble those observed in inflammatory<br />

bowel diseases (S. Cadranel, personal communication).<br />

34 Rectal bleeding can also occur through<br />

Practical approach 643<br />

colonic parasitic infections with Entamoeba<br />

histolytica and Dientamoeba fragilis. In this age<br />

group, cow’s milk is the most frequent cause of<br />

food allergy producing occult or even frank blood<br />

loss. It occurs mostly in formula-fed infants,<br />

although the same allergy can be induced indirectly<br />

through breast milk. Resolution of the symptoms<br />

usually occurs shortly after recognition of<br />

this condition and further avoidance of dairy products<br />

by the breast-feeding mother or switching to a<br />

non-allergenic formula.<br />

In this age group, because of potential severe<br />

complications and sequelae, intussusception<br />

needs early recognition and adequate management.<br />

34 It occurs more frequently in male infants<br />

(3:1) aged 3–24 months presenting with acute<br />

paroxysmal abdominal pain alternating with<br />

symptom-free periods and, later on, passage of<br />

‘currant jelly’ stools. An association between<br />

antibiotic use and primary idiopathic intussusception<br />

has been reported recently. 35<br />

Hirschsprung’s related enterocolitis may affect<br />

undiagnosed constipated infants, but also patients<br />

of any age who had undergone complicated operations.<br />

36<br />

Childhood<br />

In older children upper GI bleeding suggests an<br />

acid-related disease such as peptic esophagitis or<br />

gastritis.<br />

The Mallory–Weiss tear is a mucosal laceration at<br />

the level of the cardia following vomiting with or<br />

without symptoms of epigastric pain. 37–39 The<br />

bleeding is usually self-limited. Retching can also<br />

cause mild-to-moderate bleeding through superficial<br />

ecchymotic traumatic lesions of the fundic<br />

mucosa.<br />

In industrialized regions, upper GI bleeding due to<br />

<strong>Helicobacter</strong> <strong>pylori</strong>-associated gastritis or ulcer is<br />

seldom observed in children younger than 5<br />

years. 40 Nevertheless, systematic antral and fundic<br />

biopsies are recommended for histological study<br />

and microbiological culture. 41 Esophagitis is the<br />

most frequent cause of acute upper GI bleeding in<br />

children with brain damage. The complaints of<br />

these neurologically impaired children are often<br />

unrecognized during long periods of time result-


644<br />

Approach to gastrointestinal bleeding<br />

ing in chronic occult blood losses and esophageal<br />

stenosis. 42<br />

In children mentally retarded or with behavioral<br />

abnormalities, the possibility of ingesting foreign<br />

bodies as a source of upper GI bleeding should be<br />

considered.<br />

<strong>Portal</strong> hypertension and deficient coagulation<br />

factors complicating liver diseases are the most<br />

dramatic causes of upper GI bleeding. The<br />

adequate management of esophageal varices and<br />

preventive treatment of portal hypertension need<br />

close collaboration between a skilled pediatric<br />

gastroenterology staff and a well-equipped ICU. A<br />

common complication of portal hypertension is<br />

the typical gastropathy in which the gastric<br />

mucosa from portal hypertensive patients exhibits<br />

a typical vascular dilatation and congestion that<br />

seems more extensive after endoscopic sclerotherapy<br />

of varices. 43<br />

In pediatric ICUs the systematic prophylaxis with<br />

H2-receptor blockers and, more recently PPIs, has<br />

resulted in a dramatic decrease of peptic stress<br />

ulcers affecting mainly children with severe<br />

burns, with acute neurological diseases or undergoing<br />

major surgical procedures. Ventilator-associated<br />

pneumonia and upper airway colonization<br />

with Gram-negative bacilli does not seem to be<br />

aggravated by these drugs. 44<br />

The Dieulafoy’s lesion is an unusually large<br />

mucosal ulcer eroding a submucosal artery. This<br />

unusual lesion is observed in children who<br />

present with massive hematemesis (sometimes<br />

hematochezia) without the usually associated<br />

symptoms of ulcer. In most cases the lesion is<br />

found in the proximal stomach, but it can occur at<br />

any site of the GI tract. 45<br />

One of the most frequent causes of upper GI<br />

bleeding in young children used to be acetylsalicylic<br />

compounds. These can cause acute gastric<br />

erosions but also deep extensive ulcers. 46 Stenotic<br />

scarring may develop in the <strong>pylori</strong>c region as a<br />

result of the healing of such ‘kissing ulcers’.<br />

Recently, ibuprofen has become widely used in<br />

children in the USA and also in continental<br />

Europe as an analgesic and antipyretic drug. 47,48<br />

Its innocuousness is controversial and, along with<br />

all NSAIDs, should be avoided in children with<br />

portal hypertension. 49<br />

Other drugs potentially causing upper GI bleeding<br />

include anticoagulants and antimitotics used in<br />

oncological patients and inducing esophageal and<br />

gastric mucitis.<br />

Hematemesis was the main symptom reported in<br />

an epidemic of dengue hemorrhagic fever in children<br />

in India. 50<br />

Juvenile polyps are the most common cause of<br />

rectal bleeding in this age group and affect 3–4%<br />

of the population in Mexico. 51 Rectal bleeding<br />

from juvenile polyps is usually mild and selflimited,<br />

although the relapsing episodes can cause<br />

emotional trouble in the child and family. Polyps<br />

are to be found mainly in the rectum and<br />

sigmoid. 52 However, in our personal series, 20% of<br />

all juvenile polyps occurred in other parts of the<br />

colon, including the cecum. Retrieval of polyps<br />

for histological study is strongly recommended,<br />

especially in case of multiple polyps (10% in our<br />

series).<br />

Meckel’s diverticulum is a congenital anomaly<br />

occurring in 2% of the population. While it<br />

commonly remains clinically silent, it may also<br />

present at any age, typically with intermittent,<br />

painless frank hematochezia in children. In a<br />

series of 164 children undergoing laparotomy<br />

between 1970 and 1989, a Meckel’s diverticulum<br />

was discovered at laparotomy. There were 120<br />

boys and 44 girls with a mean age of 5.2 years<br />

(range 0–18 years). Forty-seven cases were asymptomatic,<br />

representing an incidental finding at<br />

laparotomy, 25 were resected and ectopic gastric<br />

mucosa was present in seven specimens (28%). Of<br />

the 117 symptomatic patients, 49 (42%) presented<br />

with bowel obstruction, 45 (38%) had rectal<br />

bleeding, 16 (14%) had diverticulitis and seven<br />

(6%) had umbilical pathology. 53 In another more<br />

recent review, no sex ratio differences were<br />

observed. 54<br />

Duplication cysts containing gastric mucosa that<br />

may ulcerate and bleed represent other causes of<br />

bleeding from the small intestine. Of special interest<br />

are antral duplications that cause hypergastrinemia,<br />

with consequent ulceration and bleeding.<br />

55 Other lesions as a source of bleeding from<br />

the small bowel include infectious enteritis,<br />

lymphonodular hyperplasia, 56 typhlitis, 57 Crohn’s<br />

disease and several vasculitides.


In Henoch–Schönlein purpura, gastrointestinal<br />

bleeding has been reported in 33% of cases. 58<br />

Hemolytic uremic syndrome is another vascular<br />

complication following several strains of Shigatoxin<br />

producing Escherichia coli. 59–61 Other<br />

causes of GI bleeding have been described, such<br />

as cytomegalovirus (CMV)-associated hematochezia<br />

in HIV-infected children 62 and also<br />

complications of graft-versus-host disease after<br />

bone marrow transplant 59–63 and post-chemotherapy<br />

neutro-penic enterocolitis in children with<br />

cancer. 64 Acute major gastrointestinal hemorrhage<br />

is uncommon in inflammatory bowel disease and<br />

most cases are due to Crohn’s disease without a<br />

predilection for site of involvement. 65 Although<br />

rare, bacterial overgrowth may present as a cause<br />

of lower GI bleeding. 66<br />

Vascular anomalies are a rare cause of bleeding in<br />

children. The symptoms vary according to the site<br />

and size of the bleeding lesion. Hemangiomas are<br />

proliferative lesions with a tendency to regress<br />

spontaneously, whereas vascular malformations<br />

are non-proliferative and do not regress. These rare<br />

causes of GI bleeding include Osler–Rendu–Weber<br />

disease and Klippel –Trenaunay, Turner’s and bluerubber<br />

bleb nevus syndromes.<br />

How should diagnostic investigations be used?<br />

The introduction in pediatrics of fiberoptic endoscopes,<br />

some 30 years ago, 67–69 has progressively<br />

but radically changed the investigation of GI<br />

bleeding. GI bleeding implies a mucosal lesion<br />

that will always be better detected by endoscopy<br />

than by any other ‘indirect visualization’ means.<br />

Biopsies for further study of microscopic lesions<br />

and, more recently, endoscopic treatment has<br />

become possible; endoscopy has logically become<br />

the first diagnostic step (see below). Consequently,<br />

contrast radiology is seldom – if ever –<br />

used in upper GI bleeding, since barium contrast,<br />

even with double contrast, is too insensitive to<br />

detect superficial mucosal lesions.<br />

Imaging<br />

Has the plain abdominal X-ray film become obsolete?<br />

The main advantage of this simple procedure<br />

is that it is available almost everywhere. Plain<br />

abdominal radiographs may provide useful infor-<br />

Practical approach 645<br />

mation in children with upper GI or rectal bleeding<br />

when pain or vomiting is present, and can<br />

clearly show bowel obstruction or perforation.<br />

Echography, with Doppler ultrasound, is one of the<br />

best diagnostic tools for visualizing the vascular<br />

system and evaluating the blood flow when portal<br />

hypertension is suspected. It can provide valuable<br />

information about vascular abnormalities.<br />

Echography is also able to detect bowel wall thickening,<br />

intussusceptions or, on some occasions,<br />

intraluminal masses. However, it can seldom be<br />

used as the first step in the investigation and has<br />

probably no utility as a first approach to a massive<br />

hematemesis.<br />

Computed tomography (CT) and magnetic resonance<br />

imaging (MRI) can be helpful for the detection<br />

of mass lesions or vascular malformations. An<br />

important limitation to these investigations is the<br />

fact that they require adequate sedation, in order<br />

to avoid unwanted movements of the child.<br />

The most popular form of scintigraphy is the<br />

‘Meckel’s scan’ using technetium-99m pertechnetate,<br />

which accumulates in functional gastric<br />

mucosa such as Meckel’s diverticulum; but also in<br />

other ectopic gastric mucosa, for instance in duplication<br />

cysts. However, this test is far from being<br />

accurate in all instances and as many as 20% falsepositive<br />

and 20% false-negative results are<br />

observed. 70<br />

The ‘bleeding scan’ is another scintigraphic technique<br />

that can be used to identify a bleeding site<br />

that cannot be reached by endoscopy. Technetium-<br />

99m-labeled red blood cells from a sample of the<br />

patient’s blood are re-injected into the bloodstream.<br />

Bleeding rates of 0.1ml/min are detectable<br />

through gamma camera images of the abdomen<br />

every 5min for the first hour and then at regular<br />

intervals for as long as 24h if needed. A minimum<br />

bleeding of 500ml is necessary to obtain a positive<br />

scan in cases of lower GI hemorrhage. 71–73<br />

Angiography detects active bleeding lesions or<br />

chronic recurrent blood losses if the estimated rate<br />

of bleeding exceeds 0.5ml/min. The disadvantages<br />

of this technique, besides its invasiveness requiring<br />

general anesthesia, are frequent false-negative<br />

results. Its main advantages are the correct identification<br />

of the bleeding site – for further surgery –<br />

and the possibility of using the catheter for therapy


646<br />

Approach to gastrointestinal bleeding<br />

either with selective infusion of vasopressin or<br />

embolization. 74,75<br />

Endoscopy<br />

The considerable progress achieved in the miniaturization<br />

of endoscopes and also in the field of<br />

sedation and anesthesia has substantially changed<br />

the role of endoscopy in the handling of GI bleeding<br />

in children.<br />

The availability of instruments that can be used at<br />

any age, ranging from the slimmest 6mm outer<br />

diameter ‘neonatoscopes’ to the standard 9mm<br />

gastroscopes, has enabled pediatric gastroenterology<br />

to deal more easily with the diagnosis of GI<br />

bleeding. Also, modern video-endoscopes facilitate<br />

teamwork and training. However, the use of therapeutic<br />

probes usually requires the 2.8mm operating<br />

channel diameter of the standard adult endoscope<br />

which is difficult to employ in children younger<br />

than 2 years, even under general anesthesia.<br />

Prompt upper endoscopy guarantees a more accurate<br />

result, since identification of the bleeding site<br />

can vary from 82% when endoscopy is performed<br />

within 24h to 48% if done over 72h of an upper GI<br />

bleeding. 8<br />

For bleeding of the lower GI tract, rigid endoscopes<br />

are seldom used and are replaced by the most<br />

‘user-friendly’ flexible instruments. Miniaturized<br />

colonoscopes of 10mm in outer diameter can be<br />

easily used in children older than 5 years, properly<br />

sedated, and under the control of a trained pediatric<br />

anesthesiologist. In younger children the<br />

various pediatric gastroscopes can be used, taking<br />

into account that they are not designed with the<br />

same characteristics of colonoscopes (more stiff<br />

and less handy). Colonoscopy is not always an<br />

easy procedure and a clean intestinal lumen is<br />

sometimes difficult to obtain. In non-emergency<br />

conditions, a plain abdominal X-ray film permits<br />

assessment of the degree of stool retention and<br />

starting with the necessary steps to void the colon<br />

from its fecal contents. We recommend, when<br />

possible, a strict diet without residues for 4–5 days<br />

prior to the colonoscopy and a semi-liquid diet on<br />

the last day. Many preparations combining enemas<br />

with large quantities of oral polyethyleneglycol<br />

(PEG) solution (sometimes administered through a<br />

nasogastric tube) have proved disappointing and<br />

poorly tolerated. We prefer, at least in children<br />

older than 6 years, an oral preparation consisting<br />

of the combination of sodium phosphate (acting as<br />

an osmotic laxative within 2h), and bisacodyl<br />

(acting within 6–8h) (Prepacol “ Delpharm,<br />

Bretigny-sur-Orge, France).<br />

Enteroscopy and wireless capsule video-endoscopy<br />

Modern video-endoscopes have a working length<br />

of 100cm, enabling, in experienced hands, a<br />

thorough and accurate investigation of the upper<br />

GI tract as far as the ligament of Treitz. From the<br />

opposite side, investigation of the entire colon and<br />

10–20cm of the terminal ileum is also possible.<br />

However, bleeding originating from the small<br />

bowel is not accessible to conventional endoscopic<br />

instruments. The development of a long ‘enteroscope’<br />

with easy maneuverability in order to<br />

explore the entire small bowel seems a futuristic<br />

dream; on some occasions peroperative endoscopy<br />

with the surgeon directing the progress of the<br />

instrument can be helpful.<br />

The wireless capsule endoscopy is currently the<br />

outstanding technical innovation in diagnostic<br />

gastrointestinal endoscopy. 76,77 Especially for<br />

small-bowel diseases, this new technique offers<br />

several potential advantages compared to traditional<br />

diagnostic tools. Capsule endoscopy is a<br />

painless procedure that can be performed as an<br />

ambulatory endoscopic examination. The first<br />

experimental studies showed good tolerance of the<br />

capsule endoscopy and the possibility of a<br />

complete visual investigation of the small bowel.<br />

Clinical studies have demonstrated possible fields<br />

of application in obscure chronic or intermittent<br />

GI blood loss and inflammatory bowel disease,<br />

with better results than with classical pushenteroscopy<br />

or radiological imaging. 78–80 The<br />

major risk of the procedure – intestinal obstruction<br />

by the capsule – may hinder its use in the diagnosis<br />

of polyps or tumors in the small bowel. It will<br />

probably progressively replace the cumbersome<br />

and disappointing push-enteroscopy. The capsule<br />

is relatively large but can be swallowed by children<br />

older than 8 years. 81,82<br />

Management and prevention<br />

Most GI bleedings are self-limited and need only<br />

close observation; others require medications or


even aggressive and invasive endoscopic management.<br />

Many treatments and techniques are theoretically<br />

possible, extrapolated from adult studies<br />

to the pediatric patient. The regimen implemented<br />

for a child should be individualized, taking into<br />

account the supposed or confirmed origin and<br />

etiology of the bleeding, the underlying medical<br />

condition and the age. Guidelines for pediatric<br />

situations need controlled trials in order to provide<br />

an evidence-based approach; thus, they are currently<br />

based on local competence and equipment.<br />

A local protocol should be available in order to<br />

improve the quality and efficacy of the management.<br />

83<br />

The two main and serious origins of clear upper GI<br />

bleeding are peptic and variceal.<br />

Peptic lesions<br />

Medical treatment is based on acid suppression<br />

and is often started empirically because a peptic<br />

origin is frequent in pediatrics (40% of the moderate<br />

or severe hematemesis in the series reported by<br />

Mougenot and Balquet). 84 Currently, H2-receptor<br />

antagonists and PPIs are widely used, and ranitidine<br />

and omeprazole are the most extensively<br />

studied molecules in each class, respectively. They<br />

can be given either intravenously or orally. The<br />

multiple-unit pellet system enables dispersion in<br />

water and an easier use in small children unable to<br />

swallow tablets. In bleeding peptic ulcers, the<br />

pharmacotherapy is aimed at improving the environment<br />

of the bleeding point by keeping the<br />

gastric pH above the proteolytic range for pepsin<br />

and curing the initial lesion. This kind of treatment<br />

is also indicated for preventing stress ulcers.<br />

Ranitidine Serum concentrations of ranitidine<br />

necessary to inhibit gastric acid secretion by at<br />

least 90% ranged between 40 and 60ng/ml in children<br />

aged 3–16 years. 85 The pharmacokinetics of<br />

ranitidine in critically ill children is variable. In a<br />

study in mechanically ventilated, critically ill children<br />

weighing at least 10kg, the proposed doses of<br />

ranitidine, needed to reach a gastric pH of >4 for<br />

stress ulcer prophylaxis, depended on the regimen<br />

by bolus or continuous infusion. The recommended<br />

bolus regimen is 1.5mg/kg administered<br />

every 8h. In continuous infusion, the recommended<br />

intravenous loading dose is 0.45mg/kg,<br />

followed by a continuous infusion of 0.15mg/kg<br />

Practical approach 647<br />

per hour. Thereafter, gastric pH should be monitored<br />

and the dose of ranitidine adjusted accordingly.<br />

86 Children with acute central nervous<br />

system injury or pediatric risk of mortality<br />

(PRISM) scores of >20 show a poor control of<br />

gastric pH. 87 In another study, no significant differences<br />

were observed, regarding the raising of<br />

gastric pH values above 4, between a bolus<br />

regimen with 1mg/kg in two doses 6h apart or a<br />

continuous infusion regimen of ranitidine bolus of<br />

0.15mg/kg followed by continuous infusion at<br />

0.15mg/kg per hour for 12h. Proposed doses are<br />

shown in Table 38.2. There was no correlation<br />

between illness severity scores and gastric pH<br />

values. 88 Smaller volumes can be perfused with<br />

bolus infusions, whereas continuous infusions of<br />

ranitidine often need a second line, since many<br />

drugs are incompatible with ranitidine; however,<br />

continuous infusion decreases the variability of<br />

gastric pH in children in an ICU. 87 Critically ill<br />

children with normal renal and hepatic function<br />

should be treated with a minimum 3mg/kg per day<br />

of intravenous ranitidine and the dose should be<br />

titrated to a gastric pH of ≥ 4. 89<br />

In full-term newborns, with a stable renal and<br />

hepatic function treated with extracorporeal<br />

membrane oxygenation, ranitidine administered<br />

as a single 2mg/kg dose over 10min is able, within<br />

90min after administration, to increase the intragastric<br />

pH to >5 and to maintain it at >4 for a<br />

minimum of 15h. 90<br />

In a study in 30 full-term newborns treated for<br />

bleeding erosions during the first 2 days of life, a<br />

rate of less than 0.2mg/kg per hour seemed to be<br />

advisable for continuous ranitidine infusion,<br />

whereas the 5mg/kg twice a day regimen could be<br />

considered adequate for oral therapy. 91 Preterm<br />

infants need significantly smaller doses of ranitidine<br />

to keep their intraluminal gastric pH over 4.<br />

The required optimal dose of ranitidine is<br />

0.5mg/kg body weight twice a day and that for fullterm<br />

infants 1.5mg/kg body weight three times a<br />

day. 92 In premature infants with bronchopulmonary<br />

dysplasia, infusion of 0.0625mg/kg per<br />

hour of ranitidine during dexamethasone administration<br />

is sufficient to increase and maintain the<br />

gastric pH above 4. 93 The doses of ranitidine<br />

should be adjusted in patients with severe renal<br />

failure even when undergoing regular hemodialysis.<br />

94,95 The bolus injections ought to be slow


648<br />

Approach to gastrointestinal bleeding<br />

Table 38.2 Proposed doses of ranitidine for critically ill newborns, infants and children<br />

Children<br />

References Bolus IV Continuous infusion Orally<br />

Lugo (2001) 86 1.5mg/kg tid loading dose 0.45mg/kg,<br />

followed by 0.15mg/kg/ per h<br />

Adults Marchant (1988) 139 loading dose 50mg,<br />

followed by 0.15mg/kg per h<br />

or 0.25mg/kg per h (high risk)<br />

Infants<br />

Full-term Kuusela (1998) 92 1.5mg/kg tid<br />

Fontana (1993) 91 0.2mg/kg per h 5mg/kg bid<br />

Wells (1998) 90 2mg/kg bid<br />

Preterm Kuusela (1998) 92 0.5mg/kg bid<br />

IV, intravenous; tid, three times a day; bid, twice a day<br />

because of the risk of bradycardia. The tolerance<br />

effect of ranitidine could lead, as reported in adults,<br />

to a rapid loss of antisecretory activity on days 2 and<br />

3. 96<br />

Oral doses reported in the literature vary from to<br />

6–10mg/kg per day, twice or three times a day<br />

during 8 weeks to cure esophagitis to 2.5–10mg/kg<br />

per day during 4–8 weeks for ulcers.<br />

Omeprazole The oral dose range of omeprazole for<br />

management of gastroesophageal reflux disease and<br />

acid-related diseases is 0.3–3.5mg/kg with a<br />

maximum of 80mg/day). Orally, 1mg/kg per day<br />

every 12 or 24h or simply tablets of 10mg under<br />

and 20mg above 20kg of weight are most often<br />

prescribed.<br />

In children aged 3 months to 19 years, the pharmacokinetics<br />

of intravenous doses from 36.9 to<br />

139mg/1.73m 2 were: systemic clearance of 0.23l/kg<br />

per hour; volume of distribution of 0.45l/kg; elimination<br />

half-life of 0.86h with a high individual variability.<br />

97<br />

In adults, after an initial bolus of 80mg of omeprazole,<br />

the infusion rate of 8mg/h is significantly<br />

superior to 40mg/6h. 96–98<br />

In nine adult patients with duodenal ulcers, a daily<br />

intravenous dose of 40mg omeprazole was not sufficient<br />

to keep the intragastric pH above 4 in all<br />

patients during the first day of treatment. After 5<br />

days, 1 single daily low dose of 10mg intravenously<br />

and 20mg orally were effective and dependable in<br />

reducing 24-h intragastric acidity. 99 In adults, the<br />

best regimen to raise the intragastric pH above<br />

levels alleged to allow hemostasis in patients with<br />

peptic ulcer bleeding (and subsequent healing of<br />

the ulcer) seems to be an initial bolus of omeprazole<br />

followed by a continuous infusion for 72h and then<br />

switching to a single oral dose. 100<br />

Prevention of stress ulcers or re-bleeding An evidence-based<br />

medicine review of published trials<br />

yields sufficient evidence to support the use of<br />

prophylactic acid suppression in critically ill<br />

patients with coagulopathies or in those who are<br />

receiving prolonged mechanical ventilation. Not<br />

enough data have accumulated to prove the superiority<br />

of intravenous PPIs to intravenous H2-receptor<br />

antagonists for the prophylaxis of clinically important<br />

stress ulcer bleeding. With respect to acute GI<br />

bleeding, intravenous PPI is significantly more<br />

effective than an intravenous H2-receptor antagonist<br />

in reducing the rate of re-bleeding after hemostasis<br />

in patients with bleeding peptic ulcer. 101 In adult<br />

patients, a recent placebo-controlled trial of highdose<br />

parenteral omeprazole after an initial endoscopic<br />

treatment of bleeding peptic ulcers demonstrated<br />

a substantial reduction in the risk of<br />

re-bleeding 102 and a meta-analysis showed a significant<br />

beneficial effect of acid-decreasing agents in<br />

lowering re-bleeding and surgery rates, but demonstrated<br />

no effect upon mortality. 103


<strong>Portal</strong> hypertension<br />

Esophageal variceal bleeding is one of the severe<br />

consequences of portal hypertension. However,<br />

the bleeding site may sometimes be otherwise than<br />

the cardiac regions, for instance at the level of a<br />

portoenterostomy anastomosis, in the rectal<br />

mucosa or in the gastric cavity as a consequence of<br />

a hypertensive gastropathy. The management<br />

becomes even more difficult, because of coagulation<br />

abnormalities, infection and cirrhotic<br />

encephalopathy needing their own specific treatments.<br />

Endotracheal intubation should be considered<br />

especially to protect against blood flow<br />

during hemesis.<br />

Variceal bleeding is the most common cause of<br />

severe GI bleeding in childhood. Biliary atresia<br />

and portal venous obstruction are the most<br />

common causes. Hemorrhages due to portal hypertension<br />

from pre-hepatic obstruction carry a better<br />

prognosis but are more precocious than those from<br />

an intrahepatic origin, as observed in cirrhosis or<br />

liver fibrosis. Varices in patients with biliary<br />

atresia are at high risk of bleeding and need careful<br />

preventive monitoring.<br />

A recent extensive literature search identified 13<br />

randomized control trials with a clear indication of<br />

the number of adult patients with ongoing bleeding<br />

and their clinical outcomes. Treatment of<br />

esophageal varices with band ligation appeared to<br />

be the most effective approach (91%), significantly<br />

(p


650<br />

Approach to gastrointestinal bleeding<br />

together with endoscopic variceal band ligation or<br />

sclerotherapy; all children surviving variceal<br />

hemorrhage should undergo secondary prophylaxis<br />

with band ligation. 116<br />

A randomized controlled trial in cirrhotic adults<br />

has shown that a bolus injection of somatostatin<br />

caused an immediate and marked decrease of the<br />

hepatic venous pressure gradient and azygos blood<br />

flow. The decrease seemed more pronounced<br />

compared to the effect of octreotide, probably<br />

owing to a nitric oxide-induced mechanism. 117<br />

Continuous somatostatin infusions of 500–250µg<br />

per hour have no systemic effects, but a more<br />

pronounced effect on splanchnic hemodynamics<br />

than the recommended 250µg/h. 118 In patients<br />

with active bleeding at endoscopy, the 500µg/h<br />

infusion dose achieved a higher rate of control of<br />

bleeding (82 vs. 60%, p


Sclerotherapy is used in extrahepatic as well as in<br />

intrahepatic liver diseases with a reported efficacy<br />

of 90%. 129–131 Secondary esophageal strictures<br />

occur in 5–20%, whereas deep ulcerations and<br />

perforations are rare. We recommend a semi-liquid<br />

diet during 2 days following sclerotherapy or band<br />

ligation. Coughing and vomiting should also be<br />

prevented.<br />

Elastic band ligation of varices is currently used in<br />

most pediatric gastroenterology centers with<br />

success. It is a quick procedure requiring greater<br />

endoscopic skill because of the relatively large<br />

device used to release the elastic band. 132–134<br />

Recent devices allow multiple elastic ligations,<br />

although it seems difficult (and probably dangerous)<br />

to ligate more than two or three varices at the<br />

same time. Only large varices deserve band ligation<br />

and the complete cure of esophageal varices<br />

combines initial band ligation with subsequent<br />

sclerotherapy. 135,136<br />

Endoscopic treatment of the lower GI tract is<br />

primarily polypectomy. On rare occasions other<br />

REFERENCES<br />

1. Longstreth GF. Epidemiology of hospitalization for acute<br />

upper gastrointestinal hemorrage: a population based<br />

study. Am J Gastroenterol 1995; 90: 206–210.<br />

2. Vreeburg EM, Snel P, de Bruijne JW et al. Acute upper<br />

gastrointestinal bleeding in the Amsterdam area: incidence,<br />

diagnosis, and clinical outcome. Am J<br />

Gastroenterol 1997; 92: 236–243.<br />

3. Stav K, Reif S. Gastrointestinal bleeding in children –<br />

etiology and diagnosis. Survey of patients in a Tel Aviv<br />

medical center, in the years 1990 to 1997. Harefuah<br />

2000; 138: 534–538.<br />

4. Fox V. Gastrointestinal bleeding in infancy and childhood.<br />

Gastroenterol Clin North Am 2000; 29: 37–67.<br />

5. Chaibou M, Tucci M, Dugas MA et al. Clinically significant<br />

upper GI bleeding in a pediatric intensive care<br />

unit. Pediatrics 1998; 102: 933–938.<br />

6. Chalasani N, Kahi C, Francois F et al. Improved patient<br />

survival after acute variceal bleeding: a multicenter,<br />

cohort study. Am J Gastroenterol 2003; 98: 653–659.<br />

7. Lacroix J, Nadeau D, Laberge S et al. Frequency of upper<br />

gastrointestinal tract bleeding in a pediatric intensive<br />

careunit. Crit Care Med 1992; 20: 35–42.<br />

References 651<br />

hemostatic techniques such as laser, sclerotherapy,<br />

band ligation and electrocautery have been used<br />

for vascular colonic anomalies. The polypectomy<br />

technique is similar to that used in the adult with<br />

the same snares and electrocautery units. The<br />

main differences reside in the fact that it is advisable<br />

to perform these procedures under general<br />

anesthesia, not only for the sake of pain relief but<br />

also for safety reasons, unnecessary movements<br />

being most undesirable and potentially dangerous.<br />

Although the vast majority of polyps are of the<br />

benign juvenile type, every effort in order to<br />

retrieve the polyp for histology studies is strongly<br />

recommended. 137 It is also advisable to limit the<br />

number of polyps removed at the same procedure<br />

to a maximum of ten.<br />

Aspirin and other NSAIDs have been used to<br />

reduce the number of polyps in patients with<br />

familial adenomatous polyposis. Cyclooxygenase-2<br />

inhibitors have now been approved<br />

by the Food and Drug Administration for adjuvant<br />

treatment of familial adenomatous polyposis. 138<br />

8. Mougenot JF, Duche M. Hemorragies digestives. In<br />

Navarro J, Schmitz J, eds. Gastroenterologie Pediatrique,<br />

2nd edn. Paris: Flammarion, 2000: 612–621.<br />

9. Faubion WA, Perrault J. Gastrointestinal bleeding. In<br />

Walker WA, Durie PR, Hamilton JR, Walker-Smith JA,<br />

Watkins JB, eds. Pediatric Gastrointestinal Disease, 3rd<br />

edn. Hamilton, Ontario: BC Decker, 2000: 164–178.<br />

10. Bacchini PL, Ruspaggiari C, Fornaroli F, Torroni F.<br />

L’endocopia digestiva in Urgenza. In De Angelis GL, ed.<br />

L’endoscopia digestiva in eta pediatrica e giovanile.<br />

Rome: EMSI, 2002: 111–113.<br />

11. Donatone JO, Ben R. Hemorragia Digestiva. In Morano J,<br />

ed. Pediatría. Buenos Aires: Atlante, 1997: 705–716.<br />

12. Richards RJ, Donica MB, Grayer D. Can the blood urea<br />

nitrogen/creatinine ratio distinguish upper from lower<br />

gastrointestinal bleeding? J Clin Gastroenterol 1990; 12:<br />

500–504.<br />

13. Stellato T, Rhodes RS, McDougal WS. Azotemia in<br />

upper gastrointestinal hemorrhage. A review. Am J<br />

Gastroenterol 1980; 73: 486–489.<br />

14. Moss G. Technique of iced saline lavage in upper GI<br />

hemorrhage. Am J Surg 1971; 122: 565–566.


652<br />

Approach to gastrointestinal bleeding<br />

15. Dusek JL. Iced gastric lavage slows bleeding in gastric<br />

hemorrhage. Crit Care Nurse 1984; 4: 8.<br />

16. Rana S. Gastric lavage in upper GI hemorrhage: should<br />

iced saline solution be used? J Assoc Physicians India<br />

1990; 38: 377.<br />

17. Andrus CH, Ponsky JL. The effects of irrigant temperature<br />

in upper GI hemorrhage: a requiem for iced saline<br />

lavage. Am J Gastroenterol 1987; 82: 1062–1064.<br />

18. Gilbert DA, Saunders DR. Iced saline lavage does not<br />

slow bleeding from experimental canine gastric ulcers.<br />

Dig Dis Sci 1981; 26: 1065–1068.<br />

19. Kelly D. Vitamin K-deficiency bleeding in neonates. J<br />

Pediatr Gastroenterol Nutr 1999; 29: 532.<br />

20. Cornelissen M, von Kries R, Loughnan P, Schubiger G.<br />

Prevention of vitamin K deficiency bleeding: efficacy of<br />

different multiple oral dose schedules of vitamin K. Eur<br />

J Pediatr 1997; 156: 126–130.<br />

21. de Boissieu D, Bargaoui K, Sakiroglu O et al.<br />

Esophagogastroduodenitis in the newborn. A propos of<br />

32 cases. Arch Fr Pediatr 1989; 46: 711–715.<br />

22. de Boissieu D, Dupont C, Barbet JP et al. Distinct<br />

features of upper GI endoscopy in the newborn. J<br />

Pediatr Gastroenterol Nutr 1994; 18: 334–338.<br />

23. Deneyer M, Goossens A, Pipeleers-Marichal M et al.<br />

Esophagitis of likely traumatic origin in newborns. J<br />

Pediatr Gastroenterol Nurtr 1992; 15: 81–84.<br />

24. Maki M, Ruuska T, Kuusela AL et al. High prevalence of<br />

asymptomatic esophageal and gastric lesions in preterm<br />

infants in intensive care. Crit Care Med 1993; 21:<br />

1817–1819.<br />

25. Kuusela AL, Maki M, Ruuska T, Laippala P. Stressinduced<br />

gastric findings in critically ill newborn<br />

infants: frequency and risk factors. Intensive Care Med<br />

2000; 26: 1501–1506.<br />

26. Ganga-Zandzou PS, Ategbo S, Michaud L et al. <strong>Neonatal</strong><br />

esophago-gastro-duodenoscopy. A propos of 123 examinations<br />

performed on 107 newborn infants. Arch Pediatr<br />

1997; 4: 320–324.<br />

27. Benhamou PH, Cheikh A, Francoual C et al. Possible<br />

protection by breast-feeding against severe esophageal<br />

and gastric lesions in the neonate. A case–control study.<br />

Biol Neonate 1998; 73: 337–339.<br />

28. Benhamou PH, Francoual C, Glangeaud MC et al. Risk<br />

factors for severe esophageal and gastric lesions in term<br />

neonates: a case–control study. J Pediatr Gastroenterol<br />

Nutr 2000; 31: 377–380.<br />

29. Kliegman RM, Fanaroff AA. Necrotizing enterocolitis. N<br />

Engl J Med 1984; 310: 1093–1103.<br />

30. Takeuchi S, Tamate S, Nakahira M, Kadowaki H.<br />

Esophagitis in infants with hypertrophic <strong>pylori</strong>c stenosis:<br />

a source of hematemesis. J Pediatr Surg 1993; 28: 59–62.<br />

31. Bontems P, Robert F, Van Gossum A et al. <strong>Helicobacter</strong><br />

<strong>pylori</strong> modulation of gastric and duodenal mucosal T<br />

cell cytokine secretions in children compared with<br />

adults. <strong>Helicobacter</strong> 2003; 8: 216–226.<br />

32. Donta ST, Myers MG. Clostridium difficile toxin in<br />

asymptomatic neonates. J Pediatr 1982; 15: 431–434.<br />

33. Sherertz RJ, Sarubbi FA. The prevalence of Clostridium<br />

difficile and toxin in a nursery population: a comparison<br />

between patients with necrotizing enterocolitis and an<br />

asymptomatic group. J Pediatr 1982; 100: 435–439.<br />

34. Ein SH, Alton D, Palder SB et al. Intussusception in the<br />

1990s: has 25 years made a difference? Pediatr Surg Int<br />

1997; 12: 374–376.<br />

35. Spiro DM, Arnold DH, Barone F. Association between<br />

antibiotic use and primary idiopathic intussusception.<br />

Arch Pediatr Adolesc Med 2003; 157: 54–59.<br />

36. Murthi GV, Raine PA. Preoperative enterocolitis is associated<br />

with poorer long-term bowel function after<br />

Soave–Boley endorectal pull-through for Hirschsprung<br />

disease. J Pediatr Surg 2003; 38: 69–72.<br />

37. Cannon RA, Lee G, Cox KL. Gastrointestinal hemorrhage<br />

due to Mallory–Weiss syndrome in an infant, J<br />

Pediatr Gastroenterol Nutr 1985; 4: 323–324.<br />

38. Lamiell JM, Weyandt TB. Mallory–Weiss syndrome in<br />

two children. J Pediatr 1978; 92: 583–584.<br />

39. Bak-Romaniszyn L, Malecka-Panas E, Czkwianianc E,<br />

Planeta-Malecka I. Mallory–Weiss syndrome in children.<br />

Dis Esophagus 1999; 12: 65–67.<br />

40. Rotenbacher D, Bode G, Brenner H. Dynamics of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection in early childhood in a<br />

high-risk group living in Germany: loss of infection<br />

higher than acquisition. Aliment Pharmacol Ther 2002;<br />

16: 1663–1668.<br />

41. De Giacomo C, Valdambrini V, Lizzoli F et al. A population-based<br />

survey on GI tract symptoms and<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection in children and adolescents.<br />

<strong>Helicobacter</strong> 2002; 7: 356–363.<br />

42. Norrashidah AW, Henry RL. Fundoplication in children<br />

with gastro-oesophageal reflux disease. J Pediatr Child<br />

Health 2002; 38: 156–159.<br />

43. Yachha SK, Ghoshal UC, Gupta R et al. <strong>Portal</strong> hypertensive<br />

gastropathy in children with extrahepatic portal<br />

venous obstruction: role of variceal obliteration by<br />

endoscopic sclerotherapy and <strong>Helicobacter</strong> <strong>pylori</strong>. J<br />

Pediatr Gastroenterol Nutr 1996; 23: 20–23.<br />

44. Lopriore E, Markhorst DG, Gemke RJ. Ventilator-associated<br />

pneumonia and upper airway colonisation with<br />

Gram negative bacilli: the role of stress ulcer prophylaxis<br />

in children. Intensive Care Med 2002; 28: 763–767.<br />

45. Stark ME, Gostout CJ, Balm RK. Clinical features and<br />

endoscopic management of Dieulafoy’s disease.<br />

Gastrointest Endosc 1992; 38: 545–550.<br />

46. Descos B, Hermier M, Foasso MF. Comparative study of<br />

4 groups of gastroduodenal ulcers in children. Arch Fr<br />

Pediatr 1984; 41: 399–404.<br />

47. Autret-Leca E. A general overview of the use of ibuprofen<br />

in paediatrics. Int J Clin Pract 2003; 135: 9–12.<br />

48. Lesko SM. The safety of ibuprofen suspension in children.<br />

Int J Clin Pract 2003; 135: 50–53.<br />

49. Litalien C, Jacqz-Aigrain E. Risks and benefits of nonsteroidal<br />

anti-inflammatory drugs in children: a comparison<br />

with paracetamol. Paediatr Drugs 2001; 3: 817–858.<br />

50. Aggarwal A, Chandra J, Aneja S et al. An epidemic of<br />

dengue hemorrhagic fever and dengue shock syndrome<br />

in children in Delhi. Indian Pediatr 1998; 35: 727–732.<br />

51. Cervantes-Bustamante R, Ramirez-Mayans J, Mata-<br />

Rivera N et al. Juvenile polyposis in Mexican children.<br />

Rev Gastroenterol Mex 2002; 67: 150–154.<br />

52. Mougenot JF, Baldassare ME, Mashako LM. Recto-colic<br />

polyps in the child. Analysis of183 cases. Arch Fr<br />

Pediatr 1989; 46: 245–248.<br />

53. St-Vil D, Brandt ML, Panic S et al. Meckel’s diverticulum<br />

in children: a 20-year review. J Pediatr Surg 1991;<br />

26: 1289–1292.<br />

54. Pinero A, Martinez-Barba E, Canteras M et al. Surgical<br />

management and complications of Meckel’s diverticulum<br />

in 90 patients. Eur J Surg 2002; 168: 8–12.<br />

55. Stephen TC, Bendon RW, Nagaraj HS, Sachdeva R.<br />

Antral duplication cyst: a cause of hypergastrinemia,<br />

recurrent peptic ulceration, and hemorrhage. J Pediatr<br />

Gastroenterol Nutr 1998; 26: 216–218.<br />

56. Kaplan B, Benson J, Rothstein F et al. Lymphonodular<br />

hyperplasia of the colon as a pathologic finding in children<br />

with lower gastrointestinal bleeding. J Pediatr<br />

Gastroenterol Nutr 1984; 3: 704–708.<br />

57. Meyerovitz MF, Fellows KE. Typhlitis: a cause of<br />

gastrointestinal hemorrhage in children. Am J<br />

Roentgenol 1984; 143: 833–835.<br />

58. Saulsbury FT. Henoch–Schönlein purpura in children.<br />

Report of 100 patients and review of the literature.<br />

Medicine (Baltimore) 1999; 78: 395–409.


59. Yoshioka K, Yagi K, Moriguchi N. Clinical features and<br />

treatment of children with hemolytic uremic syndrome<br />

caused by enterohemorrhagic Escherichia coli 0157:H7<br />

infection: experience of an outbreak in Sakai City, 1996.<br />

Pediatr Int 1999; 41: 223–227.<br />

60. Elliott EJ, Robins-Browne RM, O’Loughlin EV et al.<br />

Nationwide study of haemolytic uraemic syndrome:<br />

clinical, microbiological and epidemiological features.<br />

Arch Dis Child 2001; 85: 125–131.<br />

61. Misselwitz J, Karch H, Bielazewska M et al. Cluster of<br />

haemolytic–uremic syndrome caused by Shiga toxinproducing<br />

Escherichia coli 026:H11. Pediatr Infect Dis J<br />

2003; 22: 349–354.<br />

62. Ukarapol N, Chartapisak W, Lertprasertsuk N et al.<br />

Cytomegalovirus-associated manifestations involving<br />

the digestive tract in children with human immunodeficiency<br />

virus infection. J Pediatr Gastroenterol Nutr 2002;<br />

35: 669–673.<br />

63. Chirletti P, Caronna R, Arcese W et al. Gastrointestinal<br />

emergencies in patients with acute intestinal graftversus-host<br />

disease. Leuk Lymphoma 1998; 29: 129–137.<br />

64. Martinez Martinez L, Sastre Urgelles A, Ortega Martinez<br />

L et al. Neutropenic enterocolitis in children with<br />

cancer. An Esp Pediatr 1997; 46: 367–371.<br />

65. Pardi DS, Loftus EV Jr, Tremaine WJ et al. Acute major<br />

GI hemorrhage in inflammatory bowel disease.<br />

Gastrointest Endosc 1999; 49: 153–157.<br />

66. Gorski AM, Goulet O, Jehannin B et al. Digestive hemorrhage<br />

and bacterial overgrowth in children. Arch Fr<br />

Pediatr 1988; 45: 569–571.<br />

67. Cremer M, Rodesch P, Cadranel S. Fiberendoscopy of<br />

the GI tract in children. Experience with newly<br />

designed fiberscopes. Endoscopy 1974; 6: 18–19.<br />

68. Cadranel S, Rodesch P. Pediatric endoscopy in children:<br />

preparation and sedation. Gastroenterology 1977; 71:<br />

41–45.<br />

69. Cadranel S, Rodesch P, Peeters JP, Cremer M.<br />

Fiberendoscopy of the gastrointestinal tract in children.<br />

A series of 100 examinations. Am J Dis Child 1977; 131:<br />

41–45.<br />

70. Majd M. Radionuclide imaging in pediatrics. Pediatr<br />

Clin North Am 1985; 32: 1559–1579.<br />

71. Wesselhoeft CW, DeLuca FG, Luke M. Positive 99mTcpertechnetate<br />

scan in a child with intestinal arteriovenous<br />

malformation. Pediatr Surg 1986; 21: 71–72.<br />

72. Smith R, Copely DJ, Bolen FH. 99mTc RBC scintigraphy:<br />

correlation of gastrointestinal bleeding rates with scintigraphic<br />

findings. Am J Roentgenol 1987; 148: 869–874.<br />

73. Maeda M, Yamashiro Y. Diagnostic red blood cell scintigraphy<br />

in GI tract bleeding from an intestinal hemangioma.<br />

J Pediatr Gastroenterol Nutr 1986; 5: 987–989.<br />

74. Afshani E, Berger PE. Gastrointestinal tract angiography<br />

in infants and children. J Pediatr Gastroenterol Nutr<br />

1986; 5: 173–186.<br />

75. Meyerovitz MF, Fellows KE. Angiography in gastrointestinal<br />

bleeding in children. Am J Roentgenol 1984;<br />

143: 837–840.<br />

76. Rabenstein T, Krauss N, Hahn EG, Konturek P. Wireless<br />

capsule endoscopy – beyond the frontiers of flexible<br />

gastrointestinal endoscopy. Med Sci Monit 2002; 8:<br />

128–132.<br />

77. Fritscher-Ravens A, Swain CP. The wireless capsule:<br />

new light in the darkness. Dig Dis 2002; 20: 127–133.<br />

78. Costamagna G, Shah SK, Riccioni ME et al. A prospective<br />

trial comparing small bowel radiographs and video<br />

capsule endoscopy for suspected small bowel disease.<br />

Gastroenterology 2002; 123: 999–1005.<br />

79. Eliakim R, Fischer D, Suissa A et al. Wireless capsule<br />

video endoscopy is a superior diagnostic tool in<br />

comparison to barium follow-through and computerized<br />

References 653<br />

tomography in patients with suspected Crohn’s disease.<br />

Eur J Gastroenterol Hepatol 2003; 15: 363–367.<br />

80 Saurin JC, Delvaux M, Gaudin JL et al. Diagnostic value<br />

of endoscopic capsule in patients with obscure digestive<br />

bleeding: blinded comparison with video pushenteroscopy.<br />

Endoscopy 2003; 35: 576–584.<br />

81. Seidman EG. Wireless capsule video-endoscopy: an<br />

odyssey beyond the end of the scope. J Pediatr<br />

Gastroenterol Nutr 2002; 34: 333–334.<br />

82. Mallet E, Cron J, Stoller J. Wireless-capsule videoendoscopy:<br />

preliminary results in children. Arch Pediatr<br />

2003; 10: 244–245.<br />

83. Anwar RM, Dhanji A, Fish A, Singh S. Impact of protocol-based<br />

guidelines on the management and outcome<br />

of acute upper gastrointestinal hemorrhage in a district<br />

general hospital. Can J Gastroenterol 2003; 17: 97–100.<br />

84. Mougenot JF, Balquet P. Les hématémèses de l’enfant.<br />

Approche diagnostique actuelle. In Journées Parisiennes<br />

de Pédiatrie. Paris: Flammarion Médecine-Sciences,<br />

1978: 243–253.<br />

85. Blumer JL, Rothstein FC, Kaplan BS et al. Pharmacokinetic<br />

determination of ranitidine pharmacodynamics<br />

in pediatric ulcer disease. J Pediatr 1986; 108: 630.<br />

86. Lugo RA, Harrison AM, Cash J et al. Pharmacokinetics<br />

and pharmacodynamics of ranitidine in critically ill<br />

children. Crit Care Med 2001; 29: 759–764.<br />

87. Gedeit RG, Weigle CG, Havens PL, Werlin SL. Control<br />

and variability of gastric pH in critically ill children.<br />

Crit Care Med 1993; 21: 1850–1855.<br />

88. Osteyee JL, Banner W Jr. Effects of two dosing regimens<br />

of intravenous ranitidine on gastric pH in critically ill<br />

children. Am J Crit Care 1994; 3: 267–272.<br />

89. Harrison AM, Lugo RA, Vernon DD. Gastric pH control<br />

in critically ill children receiving intravenous ranitidine.<br />

Crit Care Med 1998; 26: 1433–1436.<br />

90. Wells TG, Heulitt MJ, Taylor BJ et al. Pharmacokinetics<br />

and pharmacodynamics of ranitidine in neonates<br />

treated with extracorporeal membrane oxygenation. J<br />

Clin Pharmacol 1998; 38: 402–407.<br />

91. Fontana M, Tornaghi R, Petrillo M et al. Ranitidine treatment<br />

in newborn infants: effects on gastric acidity and<br />

serum prolactin levels. J Pediatr Gastroenterol Nutr<br />

1993; 16: 406–411.<br />

92. Kuusela AL. Long-term gastric pH monitoring for determining<br />

optimal dose of ranitidine for critically ill<br />

preterm and term neonates. Arch Dis Child Fetal<br />

<strong>Neonatal</strong> Ed 1998; 78: 151–153.<br />

93. Kelly EJ, Chatfield SL, Brownlee KG et al. The effect of<br />

intravenous ranitidine on the intragastric pH of preterm<br />

infants receiving dexamethasone. Arch Dis Child 1993;<br />

69: 37–39.<br />

94. Garg DC, Baltodano N, Jallad NS et al. Pharmacokinetics<br />

of ranitidine in patients with renal failure. J Clin<br />

Pharmacol 1986; 26: 286–291.<br />

95. Garg DC, Baltodano N, Perez GO et al. Pharmacokinetics<br />

of ranitidine after intravenous administration in<br />

hemodialysis patients. Pharmacology 1985; 31: 189–193.<br />

96. Netzer P, Gaia C, Sandoz M et al. Effect of repeated<br />

injection and continuous infusion of omeprazole and<br />

ranitidine on intragastric pH over 72 hours. Am J<br />

Gastroenterol 1999; 94: 351–357.<br />

97. Jacqz-Aigrain E, Bellaich M, Faure C et al.<br />

Pharmacokinetics of intravenous omeprazole in children.<br />

Eur J Clin Pharmacol 1994; 47: 181–185.<br />

98. Kiilerich S, Rannem T, Elsborg L. Effect of intravenous<br />

infusion of omeprazole and ranitidine on twenty-fourhour<br />

intragastric pH in patients with a history of duodenal<br />

ulcer. Digestion 1995; 56: 25–30.<br />

99. Cederberg C, Thomson AB, Mahachai V et al. Effect of<br />

intravenous and oral omeprazole on 24-hour intragastric


654<br />

Approach to gastrointestinal bleeding<br />

acidity in duodenal ulcer patients. Gastroenterology<br />

1992; 103: 913–918.<br />

100. Hasselgren G, Keelan M, Kirdeikis P. Optimization of<br />

acid suppression for patients with peptic ulcer bleeding:<br />

an intragastric pH-metry study with omeprazole. Eur J<br />

Gastroenterol Hepatol 1998; 7: 601–606.<br />

101. Cash BD. Evidence-based medicine as it applies to acid<br />

suppression in the hospitalized patient. Crit Care Med<br />

2002; 30: S373–S378.<br />

102. Fasseas P, Leybishkis B, Rocca G. Omeprazole versus<br />

ranitidine in the medical treatment of acute upper<br />

gastrointestinal bleeding: assessment by early repeat<br />

endoscopy. Int J Clin Pract 2001; 55: 661–664.<br />

103. Selby NM, Kubba AK, Hawkey CJ. Acid suppression in<br />

peptic ulcer haemorrhage: a ‘meta-analysis’. Aliment<br />

Pharmacol Ther 2000; 14: 1119–1126.<br />

104. Gross M, Schiemann U, Muhlhofer A, Zoller WG. Metaanalysis:<br />

efficacy of therapeutic regimens in ongoing<br />

variceal bleeding. Endoscopy 2001; 33: 737–746.<br />

105. Zhang HB, Wong BC, Zhou XM. Effects of somatostatin,<br />

octreotide and pitressin plus nitroglycerine on systemic<br />

and portal haemodynamics in the control of acute<br />

variceal bleeding. Int J Clin Pract 2002; 56: 447–451.<br />

106. Tuggle DW, Bennett KG, Scott J, Tunell WP. Intravenous<br />

vaso-pressin gastrointestinal hemorrhage in children. J<br />

Pediatr Surg 1988; 23: 627–629.<br />

107. Ioannou GN, Doust J, Rockey DC. Systematic review:<br />

terlipressin in acute oesophageal variceal haemorrhage.<br />

Aliment Pharmacol Ther 2003; 17: 53–64.<br />

108. Ioannou G, Doust J, Rockey DC. Terlipressin for acute<br />

esophageal variceal hemorrhage. Cochrane Database<br />

Syst Rev 2003; CD002147<br />

109. Sadowski DC. Use of octreotide in the acute management<br />

of bleeding esophageal varices. Can J<br />

Gastroenterol 1997; 11: 339–343.<br />

110. Zellos A, Schwarz KB. Efficacy of octreotide in children<br />

with chronic gastrointestinal bleeding. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 442–446.<br />

111. Heikenen JB, Pohl JF, Werlin SL, Bucuvalas JC.<br />

Octreotide in pediatric patients. J Pediatr Gastroenterol<br />

Nutr 2002; 35: 600–609.<br />

112. Jenkins SA, Nott DM, Baxter JN. Pharmacokinetics of<br />

octreotide in patients with cirrhosis and portal hypertension;<br />

relationship between the plasma levels of the<br />

analogue and the magnitude and duration of the reduction<br />

in corrected wedged hepatic venous pressure. HPB<br />

Surg 1998; 11: 13–21.<br />

113 Corley DA, Cello JP, Adkisson W et al. Octreotide for<br />

acute esophageal variceal bleeding: a meta-analysis.<br />

Gastroenterology 2001; 120: 946–954.<br />

114. Siafakas C, Fox VL, Nurko S. Use of octreotide for the<br />

treatment of severe gastrointestinal bleeding in children.<br />

J Pediatr Gastroenterol Nutr 1998; 26: 356–359.<br />

115. Tauber MT, Harris AG, Rochiccioli P. Clinical use of the<br />

long acting somatostatin analogue octreotide in pediatrics.<br />

Eur J Pediatr 1994; 153: 304–310.<br />

116. McKiernan PJ. Treatment of variceal bleeding.<br />

Gastrointest Endosc Clin North Am 2001; 11: 789–812.<br />

117. Matrella E, Valatas V, Notas G et al. Bolus somatostatin<br />

but not octreotide reduces hepatic sinusoidal pressure<br />

by a NO-independent mechanism in chronic liver<br />

disease. Aliment Pharmacol Ther 2001; 15: 857–864.<br />

118. Cirera I, Feu F, Luca A et al. Effects of bolus injections<br />

and continuous infusions of somatostatin and placebo<br />

in patients with cirrhosis: a double-blind hemodynamic<br />

investigation. Hepatology 1995; 22: 106–111.<br />

119. Moitinho E, Planas R, Banares R et al. Variceal Bleeding<br />

Study Group. Multicenter randomized controlled trial<br />

comparing different schedules of somatostatin in the<br />

treatment of acute variceal bleeding. J Hepatol 2001; 35:<br />

712–718.<br />

120. Lui HF, Stanley AJ, Forrest EH et al. Primary prophylaxis<br />

of variceal hemorrhage: a randomized controlled<br />

trial. Gastroenterology 2002; 123: 735–744.<br />

121. Shashidhar H, Langhans N, Grand RJ. Propranolol in<br />

prevention of portal hypertensive hemorrhage in children:<br />

a pilot study. J Pediatr Gastroenterol Nutr 1999; 29:<br />

12–17.<br />

122. Rossle M, Haag K, Ochs A et al. The transjugular intrahepatic<br />

portosystemic stent–shunt procedure for<br />

variceal bleeding. N Engl J Med 1994; 330: 165–171.<br />

123. Heyman MB, LaBerge JM, Somberg KA et al.<br />

Transjugular intrahepatic portosystemic shunts (TIPS)<br />

in children. J Pediatr 1997; 131: 914–919.<br />

124. de Ville de Goyet J, Clapuyt P, Otte JB. Extrahilar<br />

mesenterico-left portal shunt to relieve extrahepatic<br />

portal hypertension after partial liver transplant.<br />

Transplantation 1992; 53: 231-232.<br />

125. Fuchs J, Warmann S, Kardorff R et al. Mesenterico-left<br />

portal vein bypass in children with congenital extrahepatic<br />

portal vein thrombosis: a unique curative approach.<br />

J Pediatr Gastroenterol Nutr 2003; 36: 213–216.<br />

126. Kato SG, Ozawa A, Ebina K et al. Endoscopic ethanol<br />

injection for treatment of bleeding peptic ulcer. Eur J<br />

Pediatr 1994; 153: 873–875.<br />

127. Noronha PA, Leist MH. Endoscopic laser therapy for GI<br />

bleeding from congenital lesions. J Pediatr Gastroenterol<br />

Nutr 1988; 7: 375–378.<br />

128. Fuster S, Costaguta A, Tobacco O. Treatment of bleeding<br />

gastric varices with tissue adhesive (Histoacryl) in children.<br />

Endoscopy 1998; 30: S39–S40.<br />

129. Hassall E, Berquist WE, Ament ME et al. Sclerotherapy<br />

for extrahepatic portal hypertension in childhood. J<br />

Pediatr 1989; 115: 69–74.<br />

130. Sokal EM, Van Hoorebeeck N, Van Obergh L et al.<br />

Upper GI tract bleeding in cirrhotic children candidates<br />

for liver transplantation. Eur J Pediatr 1992; 151:<br />

326–328.<br />

131. Stringer MD, Howard ER. Longterm outcome after injection<br />

sclerotherapy for oesophageal varices in children<br />

with extrahepatic portal hypertension. Gut 1994; 35:<br />

257–259.<br />

132. Yachha SK, Sharma BC, Kumar M et al. Endoscopic<br />

sclerotherapy for esophageal varices in children with<br />

extrahepatic portal venous obstruction: a follow up<br />

study. J Pediatr Gastroenterol Nutr 1997; 24: 49–52.<br />

133. Fox VL, Carr-Locke DL, Connors PJ et al. Endoscopic<br />

ligation of esophageal varices in children. J Pediatr<br />

Gastroenterol Nutr 1995; 20: 202–208.<br />

134. Cano J, Urruzuno P, Medinal E et al. Treatment of<br />

esophageal varices by endoscopic ligation in children.<br />

Eur J Pediatr Surg 1995; 5: 299–302.<br />

135. Saski T, Hasegawa T, Nakajima K et al. Endoscopic<br />

variceal ligation in the management of gastroesophageal<br />

varices in postoperative biliary atresia. J Pediat Surg<br />

1998; 33: 1628–1632.<br />

136. Price MR, Sartorelli KH, Karrer FM et al. Management<br />

of esophageal varices in children by endoscopic variceal<br />

ligation. J Pediatr Surg 1996; 31: 1056–1059.<br />

137. Latt TT, Nicholl R, Domizio P et al. Rectal bleeding and<br />

polyps. Arch Dis Child 1993; 69: 144–147.<br />

138. Giercksky KE. COX-2 inhibition and prevention of<br />

cancer. Best Pract Res Clin Gastroenterol 2001; 15:<br />

821–833.<br />

139. Marchant J, Summers K, McIsaac RL, Wood JR. A<br />

comparison of two ranitidine infusion regimens in critically<br />

ill patients. Aliment Pharmacol Ther 1988; 2:<br />

55–63.


39<br />

Introduction<br />

Approach to the child with<br />

acute diarrhea<br />

Hania Szajewska and Jacek Z Mrukowicz<br />

In this chapter we present a pragmatic, evidencebased<br />

approach to the child with acute diarrhea,<br />

which is a result of an extensive review of several<br />

diagnostic and treatment options that have been<br />

studied in children. We searched MEDLINE for<br />

relevant, good-quality observational studies on<br />

etiology, epidemiology, risk factors, clinical presentation,<br />

complications and diagnostic work-up.<br />

To evaluate treatments for acute diarrhea, we<br />

focused on data from meta-analyses, systematic<br />

reviews and randomized controlled clinical trials<br />

in MEDLINE, the Cochrane Library and the<br />

Cochrane Central Register of Controlled Trials.<br />

Finally, we searched MEDLINE for published<br />

evidence-based clinical practice guidelines developed<br />

by respected scientific societies or expert<br />

groups. We included conclusions from these documents<br />

in our chapter, because they contain recommendations<br />

based usually on a process of balancing<br />

benefit and harm of available approaches and<br />

interventions, and are commonly used to improve<br />

the quality of health care in many countries. In all<br />

cases the last search date was May 2003.<br />

Background<br />

Management of a child presenting with diarrhea is<br />

a logical chain of clinical decisions guided by<br />

answers to the following main questions.<br />

(1) Does the patient have acute diarrhea?<br />

(2) What is the presumed etiology of diarrhea<br />

(infectious vs. non-infectious)?<br />

(3) Should the patient be treated in an ambulatory<br />

setting or in the hospital?<br />

(4) Are stool cultures or any other laboratory<br />

tests required?<br />

(5) How should the patient be rehydrated: orally<br />

or intravenously?<br />

(6) How should the patient be managed nutritionally?<br />

(7) Should the patient receive any antimicrobial<br />

drug, and – if yes – which one is the most<br />

suitable?<br />

(8) Should the patient be treated with any other<br />

drug or preparation (antidiarrheal, antimotility,<br />

antiemetic)?<br />

(9) Are there any complications or specific clinical<br />

situations that require modification of the<br />

recommended approach?<br />

Below, we provide readers with the information<br />

required to answer those questions and make the<br />

best decisions for each patient.<br />

Definitions<br />

In published clinical practice guidelines and<br />

consensus documents, diarrhea is defined as a<br />

change in bowel movement for the individual<br />

child, characterized by an increase in the water<br />

content, volume and – usually – frequency of<br />

stools. 1,2 This definition also applies to exclusively<br />

breast-fed infants, in whom patterns of bowel<br />

habits vary widely from several soft or loose stools<br />

per day in one group of infants to one or two soft<br />

stools every 4–5 days in the other. For practical<br />

purposes and epidemiological investigations, diagnostic<br />

criteria in children – except for exclusively<br />

breast-fed infants – include a decrease in consistency<br />

(loose or liquid) and increase in frequency of<br />

bowel movements to ≥ 3 per 24h. 1,3 Since the<br />

increased water content in stools is the most<br />

important criterion (frequent passage of formed<br />

stools is not diarrhea), some authors advise to<br />

diagnose diarrhea when the volume of loose stools<br />

655


656<br />

Approach to the child with acute diarrhea<br />

exceeds 10g/kg of body weight per 24h in children<br />

and 200g/24h in adolescents and adults. 2 Although<br />

this definition is more precise and may be<br />

used in a research setting, it is impractical and<br />

difficult to incorporate into daily medical practice.<br />

Depending on the duration of the diarrhea, the<br />

illness may be considered acute, persistent, or<br />

chronic. Acute diarrhea is an episode of less than<br />

14 days in duration (usually a few days). Diarrhea<br />

lasting longer than 14 days is called persistent<br />

diarrhea, and longer than 30 days – chronic. 3 This<br />

categorization is somewhat arbitrary, but longer<br />

duration of diarrhea (>14 days) significantly<br />

increases the probability that symptoms are<br />

caused by some specific pathogens (see below) or<br />

the etiology of the disease is non-infectious, and<br />

the risk of a poor outcome is greater. Therefore, a<br />

more detailed diagnostic work-up towards a different<br />

etiology and modification of management are<br />

required. Persistent diarrhea, often of presumed<br />

infectious etiology, complicated by malnutrition<br />

and a high risk of serious non-intestinal infections,<br />

is a significant problem in developing countries. 1<br />

In this chapter we will discuss exclusively the<br />

management of acute diarrhea in immunocompetent<br />

children.<br />

Prevalence, incidence and disease burden<br />

Acute diarrhea is one of the most common<br />

diseases in children, and the second leading cause<br />

of morbidity and mortality worldwide. Every child<br />

encounters at least one diarrheal episode. 3–6 The<br />

attack rate in developed countries ranges from 1.2<br />

to 1.9 illnesses per person annually in the general<br />

population, and is higher in the first 2–3 years of<br />

life (2.5 illnesses per child per year to even five in<br />

those attending day-care centers). 3 The incidence<br />

is greater in developing countries, and in some<br />

tropical areas may reach even 6–10 episodes per<br />

child annually in children aged


Table 39.1 Etiological agents of acute infectious diarrhea in immunocompetent children<br />

Bacterial<br />

Viruses* Bacteria* enterotoxins* † Parasites*<br />

Inflammatory diarrhea, characterized by gross<br />

(dysentery) or occult blood and/or an increased<br />

number of leukocytes (or increased level of their<br />

marker lactoferrin) in the stool, is the result of<br />

intestinal wall invasion by Salmonella, Shigella,<br />

Campylobacter, Yersinia enterocolitica, hemorrhagic<br />

E. coli (O157:H7), enteroinvasive E. coli<br />

(EIEC) or Clostridium difficile. 1,3 This classification<br />

is helpful in planning selective microbiological<br />

testing and selecting patients who may benefit<br />

from empirical antibacterial therapy. 3,25 For a more<br />

comprehensive and specific discussion of infectious<br />

diarrheas, see Chapters 9–12.<br />

Complications<br />

Introduction 657<br />

Group A rotavirus ‡ Salmonella spp. †‡ Staphylococcus aureus Cryptosporidium<br />

(toxins A, B, C, D, E) parvum<br />

Caliciviruses and Campylobacter jejuni, ‡ Clostridium perfringens Giardia lamblia<br />

Norwalk virus Campylobacter coli (toxins A, C)<br />

Astrovirus Shigella spp. Bacillus cereus (heat-labile Blastocystis hominis<br />

and heat-stabile toxin)<br />

Adenovirus (type 40/41) Escherichia coli: EPEC, ‡‡ EHEC, Entamoeba<br />

EIEC, ETEC, ‡‡ EAggEC, DAEC histolytica ‡‡<br />

Group B rotavirus** Yersinia enterocolitica, Yersinia Balantidium coli<br />

Group C rotavirus** pseudotuberculosis<br />

Picobirnavirus** Clostridium difficile Isospora belli**<br />

Others (coronavirus, Vibrio cholerae ‡‡ Cyclospora spp.**<br />

norovirus, parvovirus,<br />

torovirus, Breda virus) ††<br />

Listeria monocytogenes † **<br />

Vibrio parahaemolyticus**<br />

Plesiomonas shigelloides**<br />

Aeromonas hydrophila**<br />

EPEC, enteropathogenic E. coli, EHEC, enterohemorrhagic E. coli (also known as Shiga-toxin-producing E. coli);<br />

EIEC, enteroinvasive E. coli; ETEC, enterotoxigenic E. coli; EaggEC, enteroaggregative E. coli; DAEC, diffusely<br />

adherent E. coli<br />

*In order of decreasing prevalence in the particular category in developed countries; † implicated in food-borne<br />

illness; ‡ common in developed countries; ** rare cause; †† unknown role in children; ‡‡ found in developing<br />

countries and endemic regions<br />

The main complications of acute diarrhea are<br />

dehydration (leading to hypovolemic shock in<br />

severe cases) and – especially in developing countries<br />

– malnutrition (particularly in persistent diarrhea).<br />

1 The mainstay of management is therefore<br />

adequate rehydration and early resumed, appropriate<br />

feeding. 1,26–28 In more severe cases, especially<br />

in malnourished children and when clear<br />

fluids, broth or cow’s milk were used instead of<br />

oral rehydration solution for initial treatment of<br />

dehydration, electrolyte abnormalities (hyper- or


658<br />

Approach to the child with acute diarrhea<br />

hyponatremia, hypokalemia) may be present. 28–30<br />

Hyponatremia is especially common in children<br />

with shigellosis and in severly malnourished children<br />

with edema. 1 In developed countries, as<br />

shown in the UK, electrolyte derangement is rare,<br />

with 1% of admissions having hypernatremia and<br />

no reports of hypokalemia or hyponatremia. 28<br />

Awareness of typical signs of hypernatremia,<br />

including doughy, velvety skin, dry and beefy red<br />

mucous membranes, muscular signs such as<br />

twitching and hyper-reflexia and central nervous<br />

system symptoms such as lethargy, confusion, irritability,<br />

rigidity, generalized convulsions and<br />

coma, is of particular importance, although there<br />

are no published studies on the reliability of these<br />

signs in diagnosing hypernatremia. In most moderate-to-severe<br />

cases of diarrhea there is some degree<br />

of metabolic acidosis. In malnourished children<br />

hypoglycemia may be a serious complication<br />

contributing to increased case fatality. 31,32 Persistent<br />

diarrhea is a serious complication in young<br />

children in developing countries, since it results in<br />

severe malnutrition and increased risk of<br />

death. 31,33,34 Pathogens such as EPEC, EAggEC,<br />

Shigella, Cryptosporidium and Giardia are significantly<br />

correlated with persistent infectious diarrhea.<br />

35,36 Some bacterial causes of diarrhea result<br />

in other serious long-term sequelae: EHEC (or<br />

Shiga-toxin-producing E. coli; STEC) infection<br />

may be followed by hemolytic uremic syndrome 37<br />

and C. jejuni infection by Guillain–Barré syndrome.<br />

38<br />

Diagnosis<br />

Clinical features<br />

Rapid onset of liquid stools with or without<br />

accompanying symptoms or signs such as nausea,<br />

vomiting, fever and abdominal pain comprise the<br />

clinical picture of acute diarrhea. As mentioned<br />

previously, for practical reasons, clinical<br />

syndromes of non-inflammatory and inflammatory<br />

diarrhea may be distinguished. 1,3,25 Watery diarrhea,<br />

sometimes with more prominent vomiting,<br />

and no blood or signs of an inflammatory response<br />

in the stool (leukocytes or lactoferrin) are features<br />

of the first diarrheal syndrome, whereas gross<br />

blood (dysentery) and an increased number of<br />

stool leukocytes and/or presence of lactoferrin,<br />

usually accompanied by fever and prominent,<br />

cramping abdominal pain and/or tenesmus, are<br />

characteristic of the second syndrome. The presence<br />

of gross blood or an inflammatory response in<br />

the stool significantly increases the chance for<br />

isolation of invasive enteric bacteria. 39–41<br />

Differential diagnosis<br />

If there are no signs and/or symptoms suggesting<br />

an alternative diagnosis (Table 39.2), an episode of<br />

acute diarrhea is considered presumably to be<br />

infectious. If symptoms and signs persist beyond<br />

10–14 days, the diagnosis should be revised, and a<br />

more detailed work-up is indicated focusing on<br />

complications and chronic conditions.<br />

Table 39.2 List of conditions that should be<br />

considered in the differential diagnosis of<br />

acute diarrhea in children<br />

Acute appendicitis<br />

Intussusception<br />

Hypertrophic <strong>pylori</strong>c stenosis<br />

Malrotation<br />

Hirschsprung’s disease (enterocolitis variant)<br />

Ileus<br />

Necrotizing enterocolitis<br />

Inflammatory bowel disease<br />

Food allergy or intolerance<br />

Malaria<br />

Measles (particularly in malnourished children)<br />

Sepsis<br />

Jejunal and ileal diverticula<br />

Diabetic ketoacidosis<br />

Staphylococcal toxic shock syndrome<br />

Celiac disease and other malabsorption syndromes<br />

Irritable bowel syndrome<br />

Pneumonia<br />

Meningitis<br />

Urinary tract infection (pyelonephritis)<br />

Acute otitis media, mastoiditis<br />

Drugs (laxatives, antibiotics, potassium salts,<br />

antineoplastic agents)<br />

Congenital adrenal hyperplasia<br />

Münchausen and Münchausen per<br />

proxy syndromes<br />

Radiotherapy<br />

Lead intoxication<br />

Mushroom poisoning (Amanita)


Estimation of dehydration<br />

Assessment of dehydration is an essential step in<br />

evaluation of a child presenting with diarrhea,<br />

since the degree of dehydration is the criterion for<br />

deciding upon the route of rehydration (oral or<br />

intravenous), thus determining also the need for<br />

hospital admission. The gold standard is to<br />

compare the current body weight of a patient with<br />

a recent measurement before the onset of the<br />

disease. Dehydration is usually quantified from<br />

the percentage of total body weight loss, and<br />

categorized as mild (10%). Unfortunately, this method is not<br />

always possible in practice, especially in infants<br />

and young children who grow relatively fast.<br />

Therefore, the World Health Organization (WHO)<br />

has proposed a set of clinical symptoms and signs,<br />

which helps to approximate the degree of dehydration<br />

(volume of fluid loss; Table 39.3) 1 and to<br />

decide about an appropriate treatment plan (see<br />

below). In one prospective cohort study performed<br />

in a developing country, prolonged ‘skinfold’ (skin<br />

pinch going back slowly), altered neurological<br />

status, sunken eyes and dry oral mucosa were<br />

found to correlate best with the percentage of<br />

dehydration in infants. 42 A study in American<br />

children reported the capillary refill time to be<br />

closely correlated with the degree of dehydration,<br />

43 although this was not confirmed subsequently<br />

by others. 42,44,45 In another small prospective<br />

study performed in hospitalized American<br />

children conventional laboratory blood tests were<br />

found to be poorly predictive of fluid deficits. 46<br />

Laboratory testing<br />

Microbiological testing<br />

The majority of episodes of acute infectious diarrhea<br />

in children are mild and resolve spontaneously<br />

without any specific antimicrobial treatment,<br />

hence no in-depth diagnostic testing is<br />

usually required for managing an individual case.<br />

Antimicrobials are beneficial only in a small,<br />

selected proportion of cases (see below), and<br />

empirical antibiotic treatment should generally be<br />

avoided in acute diarrhea. 3 Routine stool cultures<br />

in acute diarrhea have a very low yield (1.5–5.6%)<br />

and relatively high cost. 3 On the other hand,<br />

Table 39.3 Assessment for dehydration in a patient with diarrhea (from reference 1)<br />

Degree of Mouth and Total body Estimated fluid<br />

dehydration Condition Eyes Tears tongue Thirst Skin pinch weight loss (%) deficit (ml/kg)<br />

Laboratory testing 659<br />

None or mild well, alert normal present moist drinks goes back 100<br />

unconscious; sunken or not able slowly *<br />

floppy* and dry to drink*<br />

*‘Major’ signs; to classify a patient into a particular category, at least two signs must be present, including one ‘major’


660<br />

Approach to the child with acute diarrhea<br />

organism-specific diagnosis may have great public<br />

health importance, allowing for outbreak detection<br />

and control, and prevention of spread of infection<br />

in the community. Selective testing is therefore<br />

recommended, since it improves the yield and<br />

cost-effectiveness of stool cultures. 3<br />

In cases of bloody (dysenteric) or inflammatory<br />

(see below) community-acquired or traveler’s diarrhea<br />

(especially if accompanied by fever) testing<br />

for Salmonella, Shigella, Campylobacter, E. coli<br />

0157:H7 (and other STEC) and Clostridium difficile<br />

toxin A and B is recommended, the latter particularly<br />

if the diarrhea is severe and the child was<br />

treated with antibiotics during the few preceding<br />

weeks. History suggesting food-borne disease<br />

should also prompt appropriate stool cultures. 3<br />

In children most cases of nosocomial diarrhea are<br />

of viral (mainly rotaviral) or non-bacterial etiology.<br />

47–49 In older children with nosocomial diarrhea,<br />

Clostridium difficile may be more prevalent<br />

than in infants and toddlers. 50 Therefore, older<br />

children with nosocomial diarrhea (i.e. with onset<br />

after 3 days in hospital), especially when disease is<br />

severe, should be tested for Clostridium difficile<br />

toxins. Routine stool cultures for standard bacterial<br />

pathogens are discouraged for episodes occurring<br />

after the third day in hospital, since the yield<br />

is very low, except for epidemics of bloody diarrhea<br />

or suspected food-borne disease. 3,51<br />

In diarrhea that persists for longer than 10–14<br />

days, especially in an immunocompromised<br />

patient, testing for Giardia, Cryptosporidium,<br />

Cyclospora and Isospora belli should be considered.<br />

3 In almost all cases of pediatric diarrheal<br />

diseases a single stool test is diagnostic, regardless<br />

of the type of pathogen isolated. 3,52<br />

Fecal screening tests<br />

Initial screening for inflammatory diarrhea may be<br />

helpful in identifying patients requiring stool<br />

cultures or tests. One systematic review evaluated<br />

several methods for the identification of patients<br />

with inflammatory bacterial diarrhea: Wright’s or<br />

methylene blue stain of fresh stool for fecal leukocytes,<br />

a test for fecal occult blood, an immunoassay<br />

for fecal lactoferrin (a neutrophil marker)<br />

and a stain for fecal leukocytes combined with<br />

clinical data. It was concluded that fecal lactoferrin<br />

appeared to be the most accurate and useful<br />

diagnostic test for initial screening for inflammatory<br />

diarrhea. 53 Five subsequent studies confirmed<br />

the utility of lactoferrin testing to determine<br />

patients infected with invasive enteric<br />

pathogens. 54–58 Disadvantages of lactoferrin testing<br />

include its additional cost and false-positive<br />

results in breast-fed infants. 3 Lactoferrin is often<br />

negative, despite blood in stool, in STEC infection<br />

and Entamoeba histolytica colitis. 3<br />

Electrolyte measurement<br />

Serum electrolyte measurements are generally<br />

unnecessary, as most episodes of dehydration<br />

caused by diarrhea are isonatremic (see above).<br />

The American Academy of Pediatrics recommends<br />

that electrolyte levels should be measured in<br />

moderately dehydrated children whose histories<br />

are inconsistent with straightforward diarrheal<br />

episodes and in all severely dehydrated children.<br />

Furthermore, electrolytes should be measured in<br />

children with features of hypernatremic dehydration<br />

that can result from ingestion of hypertonic<br />

liquids or the loss of hypotonic fluids in the stool<br />

or urine. 27<br />

Management<br />

The main objectives in the therapeutic approach to<br />

a child with acute diarrhea are: to prevent or treat<br />

dehydration; to promote weight gain following<br />

rehydration; and to reduce the duration of diarrhea<br />

and quantity of stool output.<br />

Rehydration<br />

Oral versus intravenous rehydration in mild to<br />

moderate dehydration<br />

One systematic review of six randomized controlled<br />

trials of unequal methodological quality in<br />

371 children in developed countries with acute<br />

gastroenteritis, mostly with mild-to-moderate<br />

dehydration, found no significant difference<br />

between oral versus intravenous fluids in duration<br />

of diarrhea, duration of hospitalization, or weight<br />

gain at discharge. 59 Furthermore, patients treated


with oral rehydration therapy were not found to be<br />

at higher risk of iatrogenic hypernatremia or<br />

hyponatremia. Failure of oral treatment defined as<br />

the need for intravenous treatment was reported in<br />

3.6% of patients. Similar findings were reported in<br />

two additional randomized controlled trials not<br />

included in the systematic review. 60,61 In view of<br />

the evidence from these clinical trials, oral rehydration<br />

therapy is generally recommended to<br />

replace fluid and electrolyte losses caused by diarrhea<br />

in infants and children with mild-to-moderate<br />

dehydration. 1,27,28,62<br />

Oral versus intravenous rehydration in severe<br />

dehydration<br />

One randomized controlled trial in 470 children<br />

with severe acute gastroenteritis aged 1–18 months<br />

found that oral rehydration compared with intravenous<br />

therapy reduced the duration of diarrhea<br />

and increased weight gain at discharge, and was<br />

associated with fewer adverse effects. 63 Failure of<br />

oral treatment occurred in 0.4%. No significant<br />

differences in the death rate were found. Although<br />

oral rehydration may be used even in severe dehydration,<br />

intravenous therapy is generally recommended<br />

if the patient is more than 10% dehydrated,<br />

as well as in case of shock, failure of oral<br />

rehydration therapy, if the patient is unconscious<br />

or ileus is present. 27,28,62<br />

Nasogastric tube versus intravenous fluids<br />

One randomized controlled trial comparing nasogastric<br />

and intravenous methods of rehydration in<br />

young children with acute dehydration due to<br />

vomiting and/or diarrhea found that rehydration<br />

through a nasogastric tube was equally efficacious<br />

as intravenous rehydration, and was associated<br />

with fewer complications and was cost-effective. 64<br />

The nasogastric route of rehydration may be suitable,<br />

particularly outside facility-based treatment<br />

centers when intravenous fluids are difficult to<br />

administer.<br />

Scientific background for using oral rehydration<br />

solutions<br />

Oral rehydration therapy, hailed as potentially the<br />

most important medical discovery of the 20th<br />

century, 65 is the mainstay of treatment of acute<br />

Management 661<br />

diarrhea in patients of all ages provided they are<br />

able to drink and that the dehydration is not<br />

severe. 1,27,62,66 The effectiveness of oral rehydration<br />

solutions depends on co-transport of sodium<br />

ions and glucose (or other organic solutes) across<br />

the brush-border membranes of enterocytes, 67<br />

which results in passive absorption of water and<br />

other electrolytes. It is now well established that<br />

this glucose–sodium co-transport system remains<br />

largely intact during nearly all kinds of acute infectious<br />

gastroenteritis, irrespective of their cause.<br />

The integrity of this mechanism during any diarrheal<br />

disease makes oral rehydration therapy<br />

appropriate for treatment of diarrhea associated<br />

with dehydration. Today, a number of different<br />

oral rehydration solutions (ORSs) are available.<br />

The major differences in these solutions are<br />

related to sodium concentration and the source<br />

of carbohydrates that affects osmolality (Table<br />

39.4). 68<br />

Despite the proven efficacy of oral rehydration<br />

therapy, it remains underused. 68,69 The main<br />

reason for this is that an ORS neither reduces the<br />

frequency of bowel movements and fluid loss nor<br />

shortens the duration of illness, which decreases<br />

its acceptance. Parents, but also health-care professionals,<br />

demand safe, effective and inexpensive<br />

agents as an additional treatment that will visibly<br />

reduce the rate of stool loss and the duration of<br />

diarrhea. Such a product could, perhaps, be<br />

helpful in efforts to reduce the common practice of<br />

treating diarrhea with ineffective antidiarrheal<br />

Table 39.4 Composition of World Health<br />

Organization (WHO) and European Society<br />

for Paediatric Gastroenterology, Hepatology<br />

and Nutrition (ESPGHAN) oral rehydration<br />

solutions<br />

WHO ESPGHAN<br />

Sodium (mmol/l) 90 60<br />

Potassium (mmol/l) 20 20<br />

Chloride (mmol/l) 80 60<br />

Base (mmol/l) 30 (bicarbonate) 10 (citrate)<br />

Glucose (mmol/l) 111 74–111<br />

Osmolality (mOsm/l) 331 225–260


662<br />

Approach to the child with acute diarrhea<br />

drugs or unnecessary antibiotics. In the following<br />

section we briefly summarize the results of clinical<br />

trials of improved oral rehydration solutions.<br />

Reduced-osmolarity oral rehydration solutions<br />

There is a significant body of literature indicating<br />

that osmolality is a major factor in determining<br />

ORS efficacy. 70 The reduction of the osmolarity of<br />

the ORS may be achieved either by reducing the<br />

concentration of sodium and glucose or by replacing<br />

glucose with a complex carbohydrate.<br />

The optimum sodium concentration of an ideal<br />

ORS that would preserve and/or correct the electrolyte<br />

balance in a child with acute diarrhea<br />

without carrying the risk of inducing hypo- or<br />

hypernatremia is controversial. For many years,<br />

WHO has recommended the standard formulation<br />

of a glucose-based ORS with 90mmol/l of sodium<br />

and 111mmol/l of glucose and a total osmolarity<br />

of 311mmol/l. This original WHO standard ORS<br />

was designed for patients with cholera or choleralike<br />

(toxigenic) diarrhea associated with large<br />

stool sodium losses. However, in developed countries,<br />

where diarrheal diseases are rarely associated<br />

with large stool sodium losses, the use of<br />

WHO standard ORS has led to hypernatremia,<br />

especially in infants


Amylase resistant starch oral rehydration solutions<br />

A novel approach to enhancing the clinical effectiveness<br />

of ORSs is the addition of ingredients that<br />

utilize the absorptive capacity of the human colon.<br />

When undigested carbohydrates, such as an<br />

amylase-resistant starch or guar gum, reach the<br />

colon they are fermented into short-chain fatty<br />

acids, which induce colonic absorption of sodium<br />

and water from both the normal and the secreting<br />

colon. 84 A small randomized trial in 48 adolescents<br />

and adults with watery diarrhea due to Vibrio<br />

cholerae showed that amylase-resistant starch<br />

added to the standard WHO ORS reduced stool<br />

output. Furthermore, the mean duration of diarrhea<br />

was shorter in the amylase-resistant starch<br />

group than in the rice-flour group and the standard<br />

WHO ORS group. 85 In a similarly designed trial in<br />

150 children, partially hydrolyzed guar gum added<br />

to the standard WHO ORS compared with the<br />

control group substantially reduced the duration<br />

of diarrhea and modestly reduced stool output in<br />

acute non-cholera diarrhea in young children. In<br />

contrast, a recent multicenter study by the<br />

ESPGHAN in 144 boys with acute non-cholera<br />

diarrhea with mild-to-moderate dehydration<br />

showed that a mixture of non-digestible carbohydrates<br />

(soy polysaccharide 25%, α-cellulose 9%,<br />

gum arabic 19%, fructo-oligosaccharides 18.5%,<br />

inulin 21.5%, resistant starch 7%) was ineffective<br />

as an adjunct to oral rehydration therapy. 86 Further<br />

trials are therefore needed.<br />

Other fluids<br />

A recent survey suggested that in some countries<br />

fluids other than ORS are used for oral rehydration,<br />

including tea, cola drinks, fruit juices or<br />

chicken broth. 69 Such beverages contain inappropriate<br />

electrolyte (low sodium and potassium) and<br />

glucose concentrations, are very often hyperosmotic,<br />

and are potentially dangerous as treatments<br />

in a dehydrated child. Homemade ORS prepared<br />

by parents is also not an optimal treatment,<br />

because its preparation may involve significant<br />

errors in composition and osmolality. 87,88<br />

Recommendations on management of dehydration<br />

See Table 39.5 for a summary of practical recommendations<br />

for management of dehydration in<br />

children with acute diarrhea.<br />

Criteria for hospital admission<br />

Nutritional management 663<br />

No studies were found in which out-patient and<br />

in-patient management of children with acute<br />

diarrhea was compared. However, in some clinical<br />

practice guidelines experts recently reached<br />

consensus and specified indications for hospital<br />

admission. 1,28,89 Management in the hospital is<br />

generally required in: severely dehydrated children;<br />

children whose parents are unable to<br />

manage oral rehydration at home; patients intolerant<br />

of oral rehydration; in case of failure of treatment,<br />

e.g. worsening diarrhea and/or progression<br />

of dehydration despite oral rehydration therapy;<br />

bloody or persistent diarrhea in a severely<br />

malnourished child; and other concerns, e.g. diagnosis<br />

uncertain, potential for surgery, child at risk,<br />

child irritable or drowsy, or child younger than 2<br />

months.<br />

Nutritional management<br />

Early versus late feeding<br />

For many decades it has been an established practice<br />

that children with acute diarrhea should be<br />

fasted for 24–48h. 90,91 In various forms, these erroneous<br />

practices persist today in many communities.<br />

69 However, several controlled trials have<br />

clearly shown the beneficial effect of early feeding<br />

in malnourished 92 as well as in adequately nourished<br />

children. 93–102 Based on these studies, the<br />

ESPGHAN recently recommended, in line with<br />

AAP and WHO recommendations, that the optimal<br />

management of mild-to-moderately dehydrated<br />

children should consist of oral rehydration with an<br />

ORS over 3–4h, and rapid reintroduction of normal<br />

feeding thereafter. 1,26,27 Early feeding may<br />

decrease the intestinal permeability induced by<br />

infection, minimize protein and energy deficits,<br />

and thereby maintain growth, reduce functional<br />

and morphological hypotrophy associated with<br />

bowel rest and maintain disaccharidase activity.<br />

103,104<br />

Breast feeding<br />

For infants who are breast fed exclusively, continuation<br />

of breast feeding at all times results in


664<br />

Approach to the child with acute diarrhea<br />

Table 39.5 Practical approach to management of a child with acute diarrhea based on published<br />

clinical practice guidelines (references 1, 26–28)<br />

Management of dehydration<br />

Estimate severity of dehydration (see Table 39.3).<br />

In mildly to moderately dehydrated children, initial oral rehydration with an oral rehydradtion solution (ORS) given<br />

over 3–4h should be the preferred treatment of fluid and electrolyte losses caused by acute gastroenteritis<br />

In developed countries use of a hypotonic ORS (sodium 60mmol/l, glucose 74–111mmol/l), instead of WHO<br />

standard ORS, is recommended<br />

Suggested intake of the ORS depends on the degree of dehydration:<br />

mild dehydration (10% dehydration<br />

signs of shock<br />

unconsciousness<br />

presence of ileus<br />

failure of oral replacement therapy<br />

inability to tolerate an ORS<br />

Prevention of further dehydration could be accomplished by supplementing maintenance fluids with an ORS for<br />

ongoing losses (10ml/kg per watery stool or approximate volume of vomits)<br />

Nutritional management<br />

Rapid reintroduction of an age-appropriate normal diet (including solids) should take place after successful<br />

rehydration for 3–4h with an ORS is accomplished<br />

Routine use of a special formula is unjustified<br />

Routine use of a diluted formula is unjustified<br />

Continuation of breast feeding at all times is recommended<br />

Pharmacological therapy<br />

Antiemetics and antidiarrheal drugs should not be routinely given to infants and children with acute gastroenteritis<br />

Stool cultures and tests should be performed selectively<br />

Antimicrobial therapy is generally recommended for:<br />

shigellosis<br />

suspected cases of cholera<br />

symptomatic infection with invasive intestinal Entamoeba histolytica<br />

laboratory-proven symptomatic infection with Giardia intestinalis<br />

suspected infection with enteroinvasive bacteria in patients with an increased risk of invasive disease, including<br />

infants younger than 6 months and persons with malignant neoplasms, hemoglobinopathies, HIV infection or<br />

other immunosuppressive illness or therapy, chronic gastrointestinal tract disease, or severe colitis<br />

decreased stool output, and is generally recommended.<br />

105,106 Importantly, as demonstrated by the<br />

results of three randomized controlled trials 107–109<br />

and some observational studies, 110–113 breast<br />

feeding protects against diarrhea, thereby this<br />

practice in addition may decrease the risk of recurrent<br />

diarrheal disease.<br />

Lactose-containing versus reduced lactose or<br />

lactose-free feeds<br />

Recurring diarrhea has been attributed to lactose<br />

intolerance that can occur for a short period of<br />

time after gastroenteritis, because of mucosal<br />

damage and temporary lactase deficiency. One


systematic review of 13 randomized controlled<br />

trials has found that lactose-free feeds versus<br />

lactose-containing feeds reduced the duration of<br />

diarrhea in children with mild-to-severe dehydration.<br />

114 Subsequent trials found conflicting results.<br />

Two found that lactose-free versus lactose-containing<br />

feeds significantly reduced the duration of<br />

diarrhea, 115,116 and the other two found no significant<br />

difference. 117,118 In Europe, where lactose<br />

intolerance appears to be uncommon, the<br />

ESPGHAN recommends that the normal diet can<br />

be resumed without restriction of lactose intake in<br />

most cases. However, if diarrhea worsens on the<br />

reintroduction of standard milk or formula, stool<br />

pH and/or reducing substances should be checked<br />

and lactose content reduced only if the stool is<br />

acid and contains >0.5% reducing substances. 26<br />

Diluted or full-strength milk or formula<br />

There is concern that feeding full-strength formula<br />

or animal milk to infants aged less than 6 months<br />

with diarrhea may have adverse consequences,<br />

although one randomized controlled trial has<br />

shown that diluting formula or milk as opposed to<br />

using full-strength formula over 48h did not<br />

reduce complications. 119 More recently, one<br />

randomized, unblinded, controlled study demonstrated<br />

that, in infants aged 3–12 months with<br />

acute diarrhea, decreasing the volume of each feed<br />

and increasing the frequency of feedings to 12<br />

times per day (while maintaining the same total<br />

daily energy intake) speeded recovery, reduced<br />

fecal frequency and fecal weight, and increased<br />

weight gain during the recovery period. 120<br />

Recommendations on nutritional management<br />

See Table 37.5 for a summary of practical recommendations<br />

on nutritional management of a child<br />

with acute diarrhea.<br />

Pharmacological therapy<br />

Rational use of antibacterial drugs<br />

In the vast majority of children, acute infectious<br />

diarrhea is self-limited and antibacterial drugs are<br />

generally unnecessary, even when a bacterial<br />

Pharmacological therapy 665<br />

cause is suspected. WHO recommends routine use<br />

of antibacterial agents only for shigellosis,<br />

suspected cases of cholera, symptomatic infection<br />

with invasive intestinal Entamoeba histolytica and<br />

laboratory-proven symptomatic infection with<br />

Giardia intestinalis. 1 Although of unproven<br />

benefit, anitmicrobial therapy is generally recommended<br />

for suspected infection with enteroinvasive<br />

bacteria (e.g. Salmonella gastroenteritis) in<br />

patients with an increased risk of invasive disease,<br />

including infants younger than 6 months<br />

of age and persons with malignant neoplasms,<br />

hemoglobinopathies, HIV infection or other<br />

immunosuppressive illness or therapy, chronic<br />

gastrointestinal tract disease, or severe colitis. 121<br />

Furthermore, patients with bloody or inflammatory<br />

diarrhea accompanied by high fever, especially<br />

when the disease is moderate to severe, may<br />

benefit from empirical antibacterial treatment,<br />

modified subsequently on the basis of results of<br />

stool culture. 3 Antimicrobial treatment should be<br />

preceded by appropriate stool cultures or pathogen<br />

detection tests. One situation in which empirical<br />

antibiotics are commonly recommended without<br />

obtaining a fecal specimen is in cases of traveler’s<br />

diarrhea, in which ETEC or other bacterial<br />

pathogens are usually the likely cause. Prompt<br />

treatment with trimethoprim–sulfamethoxazole in<br />

children or fluorochinolone in adolescents and<br />

adults can reduce the duration of illness from 3–5<br />

days to less than 1–2 days. 3 Most of these episodes<br />

are self-limiting, and antimicrobial use should<br />

always be preceded by careful weighing of the<br />

benefits and potential harms of such treatment. 3<br />

Details of treatment of specific infections are<br />

summarized in Table 39.6. Because of changing<br />

patterns of antimicrobial resistance, recent local<br />

patterns are critical in making decisions about<br />

antimicrobial therapy. 3 Irrational prescribing of<br />

antibiotics may cause more harm than good (for<br />

example, it prolongs the excretion of Salmonella,<br />

leads to the emergence of drug resistance among<br />

some bacterial strains, and increases the risk of<br />

antibiotic-associated diarrhea and pseudomembranous<br />

colitis). 122,123<br />

Antiemetic drugs<br />

Vomiting is a common symptom in children with<br />

gastroenteritis. Metoclopramide, prochlorprezine<br />

and promethazine hydrochloride are effective


666<br />

Approach to the child with acute diarrhea<br />

Table 39.6 Drugs for treatment of enteric pathogens (adapted from reference 3)<br />

Pathogen Recommended treatment in immunocompetent patients<br />

Shigella species TMP–SMZ 5 and 25mg/kg, respectively, twice daily for 3 days (if susceptible)<br />

OR<br />

fluoroquinolone* for 5 days 185–190<br />

OR<br />

nalidixic acid 55mg/kg per day in four doses for 3 days 191<br />

OR<br />

ceftriaxone 100mg/kg per day in one or two doses 192<br />

OR<br />

azithromycin 189<br />

Non-typhi species not recommended routinely, 122,193,194 but advised if severe infection or in infants<br />

of Salmonella 8 years of age) 6mg/kg<br />

O1 or O139 OR<br />

TMP–SMZ 5 and 25mg/kg, respectively, twice daily for 3 days;<br />

OR<br />

single-dose fluoroquinolone* 223–227<br />

OR<br />

single-dose azithromycin (20mg/kg) 228<br />

Continued


Table 39.6 Continued<br />

Pathogen Recommended treatment in immunocompetent patients<br />

Toxigenic Clostridium offending antibiotic should be withdrawn if possible 229–231<br />

difficile metronidazole 30mg/kg per day in four doses for 10 days 232–234<br />

Giardia metronidazole 15mg/kg per day in three doses for 5 days235–237 OR<br />

tinidazole 50mg/kg single dose (maximum 2g) 238<br />

OR<br />

furazolidone 6mg/kg per day in four doses for 7–10 days<br />

OR<br />

paromomycin 25–35mg/kg per day in three doses for 7 days<br />

Cryptosporidium if severe, consider paromomycin 25–35mg/kg per day<br />

species in two or four doses for 7 days<br />

Isospora species TMP–SMZ 5 and 25mg/kg, respectively, twice daily for 7–10 days<br />

Cyclospora species TMP–SMZ 5 and 25mg/kg, respectively, twice daily for 7–10 days239,240 antiemetics. However, troublesome side-effects,<br />

including sedation and extrapyramidal reactions,<br />

occur frequently with standard doses. 124–126<br />

Therefore, these drugs should be avoided in young<br />

children with vomiting associated with acute diarrhea.<br />

Two recent studies have suggested that intravenous<br />

or oral ondansetron, a specific serotonin<br />

(5HT3) antagonist, decreases vomiting in children<br />

with gastroenteritis managed in the emergency<br />

room and reduces the need for intravenous fluid<br />

administration and hospital admission, and thus<br />

may have a valuable role as an antiemetic<br />

therapy. 127,128<br />

Antimotility or antiperistaltic drugs<br />

There is a wide variety of drugs that alter intestinal<br />

motility, including loperamide hydrochloride,<br />

diphenoxylate with atropine, tincture of opium,<br />

Pharmacological therapy 667<br />

Microsporidium species not determined<br />

Entamoeba histolytica metronidazole 35–50mg/kg per day in three doses for 7–10 days plus either diloxanide<br />

furoate 20mg/kg per day in three doses for 10 days<br />

OR<br />

paromomycin 25–35mg/kg per day in two or four doses for 7days241–243 TMP–SMZ trimethoprim–sulfamethoxazole<br />

*Fluoroquinolones (i.e. ciprofloxacin, ofloxacin, norfloxacin) are not approved for treatment of children in some<br />

countries<br />

† Antibiotics are most effective if given early in the course of illness<br />

camphorated tincture of opium and codeine.<br />

These drugs are widely used for the relief of symptoms<br />

during diarrheal episodes and are regarded as<br />

safe for adults and older children, 129,130 excluding<br />

those with bloody diarrhea or documented infection<br />

with Shiga toxin-producing E. coli (STEC). 3<br />

However, antimotility treatment should not be<br />

given for acute diarrhea in infants and young children,<br />

1,27,28 as numerous reports have documented<br />

severe and potentially life-threatening adverse<br />

effects, including lethargy, ileus, respiratory<br />

depression and coma. 131–134<br />

Adsorbents<br />

In several countries dioctahedral smectite, a<br />

natural adsorbent clay capable of adsorbing<br />

viruses, bacteria, bacterial toxins and other intestinal<br />

irritants, is commonly used for the treatment of


668<br />

Approach to the child with acute diarrhea<br />

acute infectious diarrhea. 69 Three small randomized<br />

trials in children 135–137 have suggested that<br />

smectite compared with placebo was associated<br />

with moderately reduced duration of diarrhea,<br />

although there was no significant effect on total<br />

stool output. Other adsorbents such as<br />

kaolin–pectin, fiber, attapulgite (anhydrous aluminum<br />

silicate) and activated charcoal have not<br />

been shown to be of value in the treatment of acute<br />

diarrhea in children. Adsorbents are not recommended<br />

in the routine treatment of acute diarrhea<br />

in children. 1,27,28<br />

Antisecretory drugs<br />

Bismuth subsalicylate, or other bismuth salt<br />

preparations, are common constituents of overthe-counter<br />

medications for diarrhea. Although<br />

the precise mechanism of their action remains<br />

unknown, antisecretory and antimicrobial properties<br />

have been suggested. 138–140 Three randomized<br />

controlled trials that compared bismuth subsalicylate<br />

with placebo in infants with acute watery diarrhea<br />

found that bismuth subsalicylate modestly<br />

reduced the duration and severity of diarrhea.<br />

141–143 In addition to harmless and temporary<br />

side-effects (e.g. darkening of the tongue and<br />

stool), salicylate toxicity from bismuth subsalicylate<br />

use in children has been reported. 144 The<br />

routine use of bismuth subsalicylate is not recommended<br />

in the management of children with acute<br />

diarrhea. 1,27,28<br />

Racecadotril (acetorphan), is a newer antisecretory<br />

drug that exerts its antidiarrheal effects by inhibiting<br />

intestinal enkephalinase, thus preventing the<br />

breakdown of endogenous opioids (enkephalins)<br />

in the gastrointestinal tract, thereby reducing the<br />

secretion of water and electrolytes into the gut. 145<br />

In two recent randomized placebo-controlled trials<br />

conducted in hospitalized children in developed<br />

and developing countries, racecadotril was effective<br />

in reducing the volume and frequency of stool<br />

output and in reducing the duration of diarrhea<br />

(particularly in children with rotavirus or E. coli<br />

diarrhea). 146,147 Although these data are interesting,<br />

the cost-effectiveness as well as safety needs<br />

to be defined before this drug is recommended in<br />

the routine management of children with acute<br />

diarrhea.<br />

Probiotics<br />

Probiotics are living micro-organisms that, upon<br />

ingestion in certain numbers, exert health benefits<br />

beyond their inherent general nutrition. 148 The<br />

most commonly used as probiotics are lactic acid<br />

bacteria, such as lactobacilli or bifidobacteria, and<br />

the non-pathogenic yeast Saccharomyces boulardii.<br />

The rationale for the use of probiotics is<br />

based on the assumption that they modify the<br />

composition of the colonic microflora and act<br />

against enteric pathogens. The evidence from two<br />

systematic reviews 149,150 and two subsequent<br />

randomized controlled trials 151,152 suggest a statistically<br />

significant effect and moderate clinical<br />

benefit of some probiotic strains in the treatment<br />

of acute gastroenteritis, mainly rotaviral, in infants<br />

and young children. Lactobacillus GG shows the<br />

most consistent effect. Other probiotic strains may<br />

also be effective, but should be evaluated in<br />

randomized controlled trials to prove their clinical<br />

utility, since not all products marketed as ‘probiotic’<br />

are effective. 153 So far, the beneficial effects of<br />

probiotics in acute diarrhea in children seem to be:<br />

moderate; strain-dependent; dose-dependent<br />

(greater for doses >10 10 –10 11 CFU); significant in<br />

watery diarrhea and viral gastroenteritis, but not<br />

existing in invasive (inflammatory) bacterial diarrhea;<br />

and more evident when treatment with<br />

probiotics is initiated early in the course of diarrhea.<br />

154<br />

Homeopathy<br />

The role of homeopathic remedies in the treatment<br />

of acute childhood diarrhea is still controversial. A<br />

recent meta-analysis from three randomized<br />

controlled clinical trials involving 242 children<br />

aged 6 months to 5 years has suggested that some<br />

homeopathic treatment decreases the duration of<br />

acute diarrhea in children. 155 The exact mechanism<br />

by which homeopathic remedies could have<br />

exerted their activity is unclear.<br />

Herbal medicine<br />

No systematic review or randomized controlled<br />

trial was found on herbal medicine for the treatment<br />

of acute diarrhea in children.


Micronutrients (zinc, folic acid)<br />

Zinc deficiency, which is common in young children<br />

in the developing world, is associated with<br />

impaired water and electrolyte absorption, 156–159<br />

decreased brush-border enzymes, 160–162 and impaired<br />

cellular and humoral immunity. 163–166 As<br />

intestinal losses of zinc are considerably increased<br />

during acute diarrhea, 167,168 a number of<br />

trials have evaluated the effect of zinc supplements<br />

on diarrheal diseases. The findings suggest<br />

that in developing countries zinc supplementation<br />

results in clinically important reductions in the<br />

duration and severity of acute diarrhea when given<br />

as an adjunct to oral rehydration therapy, 169–174 or<br />

mixed with an ORS. 175 Further work is needed to<br />

clarify whether zinc supplementation would also<br />

be of benefit in developed countries in children<br />

with diarrhea without pre-existing zinc deficiency.<br />

Previous reports have suggested that there is a<br />

therapeutic effect of folic acid in the treatment of<br />

acute diarrhea in children. 176 However, a recent<br />

well-designed double-blind randomized controlled<br />

trial in 106 males aged 6–23 months found no<br />

significant effect of folic acid compared with<br />

placebo in the treatment of acute watery diarrhea.<br />

177<br />

REFERENCES<br />

1. World Health Organization. The Treatment of Diarrhea.<br />

A Manual for Physicians and Other Senior Health<br />

Workers. WHO/CDR/95.3. Geneva: World Health<br />

Organization, 1995.<br />

2. Riedel BD, Ghishan FK. Acute diarrhea. In Walker WA,<br />

Durie PR, Hamilton JR, Walker-Smith JA, Watkins JB,<br />

eds. Pediatric Gastrointestinal Disease, 2nd edn. St.<br />

Louis: Mosby Year Book, 1996: 251–262.<br />

3. Guerrant RL, van Gilder T, Steiner TS et al. Practice<br />

guidelines for the management of infectious diarrhoea.<br />

Clin Inf Dis 2001; 32: 331–350.<br />

4. World Health Organization. Conquering Suffering,<br />

Enriching Humanity. Report of the Director-General.<br />

Geneva: World Health Organization, 1997.<br />

5. Bern C, Glass RI. Impact of diarrhoeal diseases worldwide.<br />

In Kapikian AZ, ed. Viral Infections of the<br />

Gastrointestinal Tract, 2nd edn. New York: Marcel<br />

Dekker, 1995.<br />

6. Bern C, Martines J, de Zoysa I, Glass RI. The magnitude<br />

of the global problem of diarrhoeal disease: a ten year<br />

update. Bull WHO 1992; 70: 705–714.<br />

Oral immunoglobulins<br />

References 669<br />

It has been claimed that oral administration of<br />

immunoglobulin-containing preparations of bovine<br />

colostrum from immunized cows, egg yolk<br />

immunoglobulin from immunized hens or pooled<br />

plasma-derived human intravenous immunoglobulins<br />

can provide passive immunity. 178 These<br />

preparations may inhibit intestinal viral adherence<br />

or viral replication and may have a role in the<br />

treatment of rotavirus infections. In fact, several<br />

randomized controlled trials have confirmed that<br />

orally administered immunoglobulins have<br />

reduced viral shedding and shortened the duration<br />

of rotavirus diarrhea. 179–182 No evidence was found<br />

that oral immunotherapy is effective in children<br />

with acute diarrhea caused by bacterial<br />

pathogens. 183,184 Except for intravenous<br />

immunoglobulins given orally in one study, 181<br />

none of the specific preparations mentioned above<br />

is available commercially.<br />

Recommendations for drug therapy<br />

See Table 39.5 for a summary of practical recommendations<br />

for drug therapy of a child with acute<br />

diarrhea.<br />

7. Glass RI, Lew JF, Gangarosa RE et al. Estimates of<br />

morbidity and mortality rates for diarrheal diseases in<br />

American children. J Pediatr 1991; 118: S27–S33.<br />

8. Kotloff KL, Winickoff JP, Ivanoff B et al. Global burden<br />

of Shigella infections: implications for vaccine development<br />

and implementation of control strategies. Bull<br />

WHO 1999; 77: 651–666.<br />

9. World Health Organization. Fighting Disease, Fostering<br />

Development. Report of the Director-General. Geneva:<br />

World Health Organization, 1996.<br />

10. Bresee JS, Glass RI, Ivanoff B et al. Current status and<br />

future priorities for rotavirus vaccine development,<br />

evaluation and implementation in developing countries.<br />

Vaccine 1999; 17: 2207–2222.<br />

11. Glass RI, Bresee JS, Parashar DU et al. First rotavirus<br />

vaccine licensed: is there really a need? Acta Paediatr<br />

1999; 426 (Suppl): 2–8.<br />

12. Phavichtir N, Catto-Smith AG. Acute gastroenteritis in<br />

children. What role for antibacterials? Pediatr Drugs<br />

2003; 5: 279–290.


670<br />

Approach to the child with acute diarrhea<br />

13. Huilan S, Zhen LG, Matham MM et al. Etiology of acute<br />

diarrhoea among children in developing countries: a<br />

multicentre study in five countries. Bull WHO 1991; 69:<br />

549–555.<br />

14. Gracey M. Diarrheal disease in perspective. In Gracey<br />

M, Walker-Smith JA, eds. Diarrheal Disease. Vol 38.<br />

Nestle Nutrition Workshop Series Nestec Ltd. and<br />

Lippincott-Raven Publishers, Philadelphia, 1991: 1–12.<br />

15. Caeiro JP, Mathewson JJ, Smith MA et al. Etiology of<br />

outpatient pediatric nondysenteric diarrhea: a multicenter<br />

study in the United States. Pediatr Infect Dis J 1999;<br />

18: 94–97.<br />

16. Caprioli A, Pezzella C, Morelli R et al. Enteropathogens<br />

associated with childhood diarrhea in Italy. Pediatr<br />

Infect Dis J 1996; 15: 876–883.<br />

17. Abu-Elyazeed R, Wierzba TF, Mourad AS et al.<br />

Epidemiology of enterotoxigenic E. coli diarrhea in a<br />

pediatric cohort in a periurban area of lower Egypt. J Inf<br />

Dis 1999; 179: 382–389.<br />

18. Kapikian AZ. Viral gastroenteritis. JAMA 1993; 269:<br />

627–630.<br />

19. Forbes D, Ee L, Camer-Pesci P, Ward PB. Faecal candida<br />

and diarrhea. Arch Dis Child 2001; 84: 328–331.<br />

20. Cohen MB. Etiology and mechanisms of acute infectious<br />

diarrhea in infants in the United States. J Pediatr<br />

1991; 118: S34–S39.<br />

21. Guandalini S, Pensabene L, Abu Zikri M et al.<br />

Lactobacillus GG administered in oral rehydration solution<br />

to children with acute diarrhea: a multicenter<br />

European trial. J Pediatr Gastroenterol Nutr 2000; 30:<br />

54–60.<br />

22. Gastanaduy AS, Begue RE. Acute gastroenteritis. Clin<br />

Pediatr 1999; 38: 1–12.<br />

23. Mrukowicz JZ, Krobicka B, Duplaga M et al.<br />

Epidemiology and impact of rotavirus diarrhoea in<br />

Poland. Acta Paediatr 1999; 426 (Suppl): 53–60.<br />

24. Chalmers RM, Salmon RL. Primary care surveillance for<br />

acute bloody diarrhea, Wales. Emerg Infect Dis 2000; 6:<br />

412–414.<br />

25. DuPont HL. Guidelines on acute infectious diarrhea in<br />

adults. The Practice Parameters Committee of the<br />

American College of Gastroenterology. Am J<br />

Gastroenterol 1997; 92: 1962–1975.<br />

26. Walker-Smith J, Sandhu B, Isolauri E et al.<br />

Recommendation for feeding in childhood gastroenteritis.<br />

J Pediatr Gastroenterol Nutr 1997; 24: 619–620.<br />

27. Provisional Committee on Quality Improvement,<br />

Subcommittee on Acute Gastroenteritis. Practice<br />

Parameter: the management of acute gastroenteritis in<br />

young children. Pediatrics 1996; 97: 424–435.<br />

28. Armon K, Stephenson T, MacFaul R et al. An evidence<br />

and consensus based guideline for acute diarrhea<br />

management. Arch Dis Child 2001; 85: 132–142.<br />

29. Abu-Ekteish F, Zahraa J. Hypernatraemic dehydration<br />

and acute gastroenteritis in children. Ann Trop Paediatr<br />

2002; 22: 245–249.<br />

30. Moritz ML, Ayus JC. The changing pattern of hypernatremia<br />

in hospitalized children. Pediatrics 1999; 104:<br />

435–439.<br />

31. Uysal G, Sokmen A, Vidinlisan S. Clinical risk factors<br />

for fatal diarrhea in hospitalized children. Indian J<br />

Pediatr 2000; 67: 329–333.<br />

32. Ahmed T, Ali M, Ullah MM et al. Mortality in severely<br />

malnourished children with diarrhoea and use of a<br />

standardised management protocol. Lancet 1999; 353:<br />

1919–1922.<br />

33. Victora CG, Huttly SR, Fuchs SC et al. International<br />

differences in clinical patterns of diarrhoeal deaths: a<br />

comparison of children from Brazil, Senegal,<br />

Bangladesh, and India. J Diarrhoeal Dis Res 1993; 11:<br />

25–29.<br />

34. World Health Organization. Diarrhoeal Disease Control.<br />

Persistent Diarrhea in Children. CCD/DDM/85.1. Geneva:<br />

World Health Organization, 1985.<br />

35. Shahid NS, Sack DA, Rahman M et al. Risk factors for<br />

persistent diarrhoea. BMJ 1988; 297: 1036–1038.<br />

36. World Health Organization. Persistent Diarrhea in<br />

Children in Developing Countries. Report of a WHO<br />

meeting. WHO/CCD/88.27. Geneva: World Health<br />

Organization, 1988.<br />

37. Wong CS, Jelacic S, Habeeb RL et al. The risk of the<br />

hemolytic–uremic syndrome after antibiotic treatment<br />

of Escherichia coli 0157:H7 infections. N Engl J Med<br />

2000; 342: 1930–1936.<br />

38. Nachamkin I, Allos BM, Ho T. Campylobacter species<br />

and Guillain–Barré syndrome. Clin Microbiol Rev 1998<br />

11: 555–567.<br />

39. Talan DA, Moran GJ, Newdow M et al. Etiology of<br />

bloody diarrhea among patients presenting to US emergency<br />

departments: prevalence of E. coli 0157:H7 and<br />

other enteropathogens. Clin Inf Dis 2001; 32: 573–580.<br />

40. Choi SW, Park CH, Silva TM et al. To culture or not to<br />

culture: fecal lactoferrin screening for inflammatory<br />

bacterial diarrhea. J Clin Microbiol 1996; 34: 928–932.<br />

41. Huicho L, Campos M, Rivera J et al. Fecal screening<br />

tests in the approach to acute infectious diarrhea: a<br />

scientific overview. Pediatr Infect Dis J 1996; 15:<br />

486–494.<br />

42. Duggan Ch, Refat M, Hashem M et al. How valid are<br />

clinical signs of dehydration in infants? J Pediatr<br />

Gastroenterol Nutr 1996; 22: 56–61.<br />

43. Saavedra JM, Harris GD, Li S et al. Capillary refilling<br />

(skin turgor) in the assessment of dehydration. Am J Dis<br />

Child 1991; 145: 296–298.<br />

44. Schriger DL, Baraff L. Defining normal capillary refill:<br />

variation with age, sex, and temperature. Ann Emerg<br />

Med 1988; 17: 932–935.<br />

45. Gorelick MH, Shaw KN, Baker MD. Effect of ambient<br />

temperature on capillary refill in healthy children.<br />

Pediatrics 1993; 92: 699–702.<br />

46. Teach SJ, Yates EW, Feld LG. Laboratory predictors of<br />

fluid deficit in acutely dehydrated children. Clin Pediatr<br />

1997; 36: 395–400.<br />

47. Ford-Jones EL, Mindorff CM, Gold R et al. The incidence<br />

of viral-associated diarrhea after admission to a<br />

pediatric hospital. Am J Epidemiol 1990; 131: 711–718.<br />

48. Szajewska H, Kotowska M, Mrukowicz J et al.<br />

Lactobacillus GG in prevention of diarrhea in hospitalized<br />

children. J Pediatr 2001; 138: 361–365.<br />

49. Mastretta E, Longo P, Laccisaglia A et al. Lactobacillus<br />

GG and breast feeding in the prevention of rotavirus<br />

nosocomial infection. J Pediatr Gastroenterol Nutr 2002;<br />

35: 527–531.<br />

50. Metropol SB, Luberti AA, De Jong AR. Yield from stool<br />

testing of pediatric inpatients. Arch Pediatr Adolesc Med<br />

1997; 151: 142–145.<br />

51. Craven D, Brick D, Morrisey A et al. Low yield of bacterial<br />

stool culture in children with nosocomial diarrhea.<br />

Pediatr Inf Dis J 1998; 17: 1040–1044.<br />

52. Church DL, Cadrain G, Kabani A et al. Practice guidelines<br />

for ordering stool cultures in a pediatric population.<br />

Alberta Children’s Hospital, Calgary, Alberta,<br />

Canada. Am J Clin Pathol 1995; 103: 149–153.<br />

53. Huicho L, Campos M, Rivera J et al. Fecal screening<br />

tests in the approach to acute infectious diarrhea: a<br />

scientific overview. Pediatr Infect Dis J 1996; 15:<br />

486–494.


54. Choi SW, Park CH, Silva TM et al. To culture or not to<br />

culture: fecal lactoferrin screening for inflammatory<br />

bacterial diarrhea. J Clin Microbiol 1996; 34: 928–932.<br />

55. Huicho L, Garaycochea V, Uchima N et al. Fecal lactoferrin,<br />

fecal leukocytes and occult blood in the diagnostic<br />

approach to childhood invasive diarrhea. Pediatr Inf<br />

Dis J 1997; 16: 644–647.<br />

56. Ruiz-Pelaez JG, Mattar S. Accuracy of fecal lactoferrin<br />

and other stool tests for diagnosis of invasive diarrhea<br />

at a Colombian pediatric hospital. Pediatr Inf Dis J 1999;<br />

18: 342–346.<br />

57. McIver CJ, Hansman G, White P et al. Diagnosis of<br />

enteric pathogens in children with gastroenteritis.<br />

Pathology 2001; 33: 353–358.<br />

58. Venkataraman S, Ramakrishna BS, Kang G et al. Faecal<br />

lactoferrin as a predictor of positive faecal culture in<br />

small Indian children with acute diarrhoea. Ann Trop<br />

Paediatr 2003; 23: 9–13.<br />

59. Gavin N, Merrick N, Davidson B. Efficacy of glucosebased<br />

oral rehydration therapy. Pediatrics 1996; 98:<br />

45–51.<br />

60. Singh M, Mahmoodi A, Arya LS et al. Controlled trial of<br />

oral versus intravenous rehydration in the management<br />

of acute gastroenteritis. Indian J Med Res 1982; 75:<br />

691–693.<br />

61. Oritiz A. Rehidratacion oral: experiencia en el manejo<br />

de pacientes con gastroenteritis aguda en la sala de<br />

emergencia hospital pediatrico. Bol Asoc Med P R 1990;<br />

82: 227–233.<br />

62. Murphy MS. Guidelines for managing acute gastroenteritis<br />

based on a systematic review of published<br />

research. Arch Dis Child 1998; 79: 279–284.<br />

63. Sharifi J, Ghavami F, Nowrouzi Z et al. Oral versus<br />

intravenous rehydration therapy in severe gastroenteritis.<br />

Arch Dis Child 1985; 60: 856–860.<br />

64. Nager AL, Wang VJ. Comparison of nasogastric and<br />

intravenous methods of rehydration in pediatric<br />

patients with acute dehydration. Pediatrics 2002; 109:<br />

566–572.<br />

65. Anonymous. Water with sugar and salt. Lancet 1978; 2:<br />

300–301.<br />

66. Avery ME, Snyder JD. Oral therapy for acute diarrhea.<br />

The underused simple solution. N Engl J Med 1990; 323:<br />

891–894.<br />

67. Schultz SA. Sodium coupled solute transport by small<br />

intestine: a status report. Am J Physiol 1977; 223:<br />

E249–E254.<br />

68. Guarino A, Albano F, Guandalini S, for the ESPGHAN<br />

Working Group on Acute Diarrhea. Oral rehydration<br />

solution: toward a real solution. J Pediatr Gastroenterol<br />

Nutr 2001; 33: S2–S12.<br />

69. Szajewska H, Hoekstra JH, Sandhu B et al. Management<br />

of acute gastroenteritis in Europe and the impact of the<br />

new recommendations: a multicenter study. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 522–527.<br />

70. Thillainayagam AV, Hunt JB, Farthing MJG. Enhancing<br />

clinical efficacy of oral rehydration therapy: is low<br />

osmolality the key? Gastroenterology 1998; 114:<br />

197–210.<br />

71. Bhargarva SK, Sachder HPS, Das Gupta B et al. Oral<br />

rehydration of neonates and young infants with dehydrating<br />

diarrhea: comparison of low and standard<br />

sodium content in oral rehydration solutions. J Pediatr<br />

Gastroenterol Nutr 1984; 3: 500–504.<br />

72. Abdalla S, Helmy N, El Essaily M et al. Oral rehydration<br />

for the low birth weight baby with diarrhoea. Lancet<br />

1984; 2: 818–819.<br />

73. Nalin DR, Hanland E, Ramlal A et al. Comparison of<br />

low and high sodium and potassium content in oral<br />

rehydration solutions. J Pediatr 1980; 97: 848–853.<br />

References 671<br />

74. Santosham M, Daum RS, Dillman L et al. Oral rehydration<br />

therapy of infantile diarrhoea. A controlled study<br />

of well-nourished children hospitalized in the United<br />

States and Panama. N Engl J Med 1982; 306: 1070–1076.<br />

75. Pizzaro D, Posada G, Villariceucis N et al. Oral rehydration<br />

in hypernatremic and hyponatremic diarrheal<br />

dehydration. Treatment with oral jejunal electrolyte<br />

solution. Am J Dis Child 1983; 137: 730–734.<br />

76. Bhargarva S, Sachder HPS, Das Gupta B et al. Oral<br />

therapy of neonates and young infants with dehydrating<br />

diarrhea: composition of low and standard sodium in<br />

oral rehydration solutions. J Pediatr Gastroenterol Nutr<br />

1986; 6: 416–422.<br />

77. International Study Group on reduced-osmolality ORS<br />

solutions. Multicentre evaluation of reduced-osmolarity<br />

oral rehydration salts solution. Lancet 1995; 345:<br />

282–285.<br />

78. Booth I, Cunha Ferreira R, Desjeux JF et al.<br />

Recommendations for composition of oral rehydration<br />

solutions from the children of Europe. Report of an<br />

ESPGAN working group. J Pediatr Gastroenterol Nutr<br />

1992; 14: 113–115.<br />

79. Kim Y, Hahn S, Garner P. Reduced osmolarity oral rehydration<br />

solution for treating dehydration caused by acute<br />

diarrhoea in children (Cochrane Review). The Cochrane<br />

Library, Issue 2. Oxford: Update Software, 2003.<br />

80. Fontaine O, Gore SM, Pierce NF. Rice-based oral rehydration<br />

solution for treating diarrhoea (Cochrane<br />

Review). The Cochrane Library, Issue 2. Oxford, 2003.<br />

81. Akbar MS, Baker KM, Aziz MA et al. A randomized<br />

double-blind clinical trial of a maltodextrin containing<br />

oral rehydration solution in acute infantile diarrhoea. J<br />

Diarrhoeal Dis Res 1991; 9: 33–37.<br />

82. Santos Ocampo PD, Bravo LC, Rogacion JM et al. A<br />

randomized double-blind clinical trial of a maltodextrin<br />

containing oral rehydration solution in acute infantile<br />

diarrhea. J Pediatr Gastroenterol Nutr 1993; 16: 23–28.<br />

83. Bhan MK, Mahalanabis D, Fontaine O, Pierce NF.<br />

Clinical trials of improved oral rehydration salt formulations:<br />

a review. Bull WHO 1994; 72: 945–955.<br />

84. Binder HJ, Mehta P. Short-chain fatty acids stimulate<br />

active sodium and chloride absorption in vitro in the rat<br />

distal colon. Gastroenterology 1989; 96: 989–996.<br />

85. Ramakrishna BS, Venkataraman S, Srinivasan P et al.<br />

Amylase-resistant starch plus oral rehydration solution<br />

for cholera. N Engl J Med 2000; 342: 308–313.<br />

86. Hoekstra JH, Szajewska H, Abu Zikri M et al. Oral rehydration<br />

solution with a mixture of non-digestible carbohydrates<br />

in the treatment of acute diarrhea. A multicenter<br />

randomised placebo controlled study on behalf of<br />

the ESPGHAN Working Group on Intestinal Infections. J<br />

Pediatr Gastroenterol Nutr 2004; in press.<br />

87. Santosham M, Foster S, Garrett S et al. Outpatient use<br />

of oral rehydration solutions in an Apache population:<br />

effects of instructions on preparation and contamination.<br />

J Pediatr Gastroenterol Nutr 1984; 3: 687–691.<br />

88. Snyder JD, Molla M, Cash RA. Home-based therapy for<br />

diarrhea. J Pediatr Gastroenterol Nutr 1990; 11: 438–447.<br />

89. Sandhu BK, for the European Society of Paediatric<br />

Gastroenterology, Hepatology and Nutrition Working<br />

Group on Acute Diarrhoea. Practical guidelines for the<br />

management of gastroenteritis in children. J Pediatr<br />

Gastroenterol Nutr 2001; 33: S36–S39.<br />

90. Powers GF. A comprehensive plan of treatment for the<br />

so-called intestinal intoxication of infants. J Dis Child<br />

1926; 32: 232–257.<br />

91. Goodburn E, Mattosinho S, Mongi P et al. Management<br />

of childhood diarrhoea by pharmacists and parents: is<br />

Britain lagging behind the Third World? BMJ 1991; 302:<br />

440–443.


672<br />

Approach to the child with acute diarrhea<br />

92. Brown KH, Gastanaduy AS, Saavedra JM et al. Effect of<br />

continued oral feeding on clinical and nutritional<br />

outcomes of acute diarrhoea in children. J Pediatr 1988;<br />

112: 191–200.<br />

93. Sandhu B, Isolauri E, Walker-Smith JA et al. Early<br />

feeding in childhood gastroenteritis. A multicentre<br />

study on behalf of the European Society of Paediatric<br />

Gastroenterology and Nutrition Working Group on<br />

Acute Diarrhea. J Pediatr Gastroenterol Nutr 1997; 24:<br />

522–527.<br />

94. Vesikari T, Isolauri E. Oral rehydration, rapid feeding<br />

and cholestyramine for treatment of acute diarrhoea. J<br />

Pediatr Gastroenterol Nutr 1986; 4: 366–374.<br />

95. Isolauri E, Vesikari T, Saha P et al. Milk versus no milk<br />

in rapid refeeding after acute gastroenteritis. J Pediatr<br />

Gastroenterol Nutr 1986; 5: 254–261.<br />

96. Placzek M, Walker-Smith JA. Comparison of two feeding<br />

regimens following acute gastroenteritis in infancy. J<br />

Pediatr Gastroenterol Nutr 1984; 3: 245–248.<br />

97. Dugdale A, Lovell S, Gibbs V et al. Refeeding after acute<br />

gastroenteritis: a controlled study. Arch Dis Child 1982;<br />

57: 76–79.<br />

98. Rees L, Brook CGD. Gradual reintroduction of full<br />

strength milk after acute gastroenteritis in children.<br />

Lancet 1979; 2: 770–771.<br />

99. Hoghton MAR, Mittal NK, Mahdi G et al. Continuous<br />

modified feeding in acute gastroenteritis. J Gen Pract<br />

1996; 46: 173–175.<br />

100. Dugdale A, Lovell S, Gibbs V et al. Refeeding after acute<br />

gastroenteritis: a controlled study. Arch Dis Child 1982;<br />

57: 76–78.<br />

101. Haque KN, Al-Frayh A, El-Rifai R. Is it necessary to<br />

regraduate milk after acute gastroenteritis in children?<br />

Trop Geogr Med 1983; 35: 369–373.<br />

102. Rajah R, Pettifor JM, Noormohamed M et al. The effect<br />

of feeding four different formulae on stool weights in<br />

prolonged dehydrating infantile gastroenteritis. J Pediatr<br />

Gastroenterol Nutr 1988; 7: 203–207.<br />

103. Isolauri E, Juntunen M, Wiren S et al. Intestinal permeability<br />

changes in acute gastroenteritis: effects of clinical<br />

factors and nutritional management. J Pediatr<br />

Gastroenterol Nutr 1989; 8: 466–473.<br />

104. Levine GM, Deren JJ, Steiger E et al. Role of oral intake<br />

in maintenance of gut mass and disaccharide activity.<br />

Gastroenterology 1974; 67: 975–982.<br />

105. Brown KH, Lake A. Appropriate use of human and nonhuman<br />

milk for the dietary management of children<br />

with diarrhoea. J Diarrhoeal Dis Res 1991; 9: 168–185.<br />

106. Khin MU, Nyunt-Nyunt W, Myokhin AJ et al. Effect of<br />

clinical outcome of breast feeding during acute diarrhoea.<br />

BM J 1985; 290: 587–589.<br />

107. Morrow AL, Guerrero ML, Shults J et al. Efficacy of<br />

home-based peer counselling to promote exclusive<br />

breastfeeding: a randomised controlled trial. Lancet<br />

1999; 353: 1226–1231.<br />

108. Kramer MS, Chalmers B, Hodnett ED et al. Promotion of<br />

breastfeeding intervention trials (PROBIT): a randomized<br />

trial in the Republic of Belarus. JAMA 2001; 285:<br />

413–420.<br />

109. Bhandari N, Bahl R, Mazumdar S et al. Effect of<br />

community-based promotion of exclusive breastfeeding<br />

on diarrhoeal illness and growth: a cluster randomised<br />

controlled trial. Lancet 2003; 361: 1418–1423.<br />

110. Cunningham A, Jelliffe D, Jelliffe E. Breastfeeding and<br />

health in the 1980s: a global epidemiologic review. J<br />

Pediatr 1991; 118: 659–666.<br />

111. Dewey K, Heinig M, Nommsen-Rivers L. Differences in<br />

morbidity between breastfed and formula fed infants. J<br />

Pediatr 1995; 126: 696–702.<br />

112. Howie P, Forsyth J, Ogston S et al. Protective effects of<br />

breastfeeding against infection. BMJ 1990; 300: 11–16.<br />

113. Oddy WH. Breastfeeding protects against illness and<br />

infection in infants and children: a review of the<br />

evidence. Breastfeed Rev 2001; 9: 11–18.<br />

114. Brown KH, Peerson JM, Fontaine O. Use of nonhuman<br />

milks in the dietary management of young children<br />

with acute diarrhea: a meta-analysis of clinical trials.<br />

Pediatrics 1994; 93: 17–27.<br />

115. Allen UD, McLeod K, Wang EE. Cow’s milk versus soybased<br />

formula in mild and moderate diarrhea: a<br />

randomized, controlled trial. Acta Paediatr 1994; 83:<br />

183–187.<br />

116. Fayad IM, Hashem M, Hussein A et al. Comparison of<br />

soy-based formulas with lactose and with sucrose in the<br />

treatment of acute diarrhea in infants. Arch Pediatr<br />

Adolesc Med 1999; 153: 675–680.<br />

117. Clemente YF, Tapia CC, Comino AL et al. Lactose-free<br />

formula versus adapted formula in acute infantile diarrhoea.<br />

An Esp Pediatr 1993; 39: 309–312.<br />

118. Lozano JM, Cespedes JA. Lactose vs. lactose free<br />

regimen in children with acute diarrhoea: a randomized<br />

controlled trial, Arch Latinoam Nutr 1994; 44: 6–11.<br />

119. Chew F, Penna FJ, Peret Filho LA et al. Is dilution of<br />

cows’ milk formula necessary for dietary management<br />

of acute diarrhoea in infants aged less than 6 months?<br />

Lancet 1993; 341: 194–197.<br />

120. Wan C, Phillips MR, Dibley MJ et al. Randomised trial<br />

of different rates of feeding in acute diarrhoea Arch Dis<br />

Child 1999; 81: 487–491.<br />

121. American Academy of Pediatrics. 2000 Red Book: Report<br />

of the Committee on Infectious Diseases, 25th edn. Elk<br />

Grove Village, IL: American Academy of Pediatrics,<br />

2000.<br />

122. Sirinavin S, Garner P. Antibiotics for treating salmonella<br />

gut infections (Cochrane Review). In The Cochrane<br />

Library, Issue 2. Oxford: Update Software, 2003.<br />

123. Periman P. Antibiotic-associated diarrhea. N Engl J Med<br />

2002; 346: 334–339.<br />

124. DeGrandi T, Simon JE. Promethazine-induced dystonic<br />

reaction. Pediatr Emerg Care 1987; 3: 91–92.<br />

125. Bateman DN, Darling WM, Boys R et al. Extrapyramidal<br />

reactions to metoclopramide and prochlorperazine. Q J<br />

Med 1989; 71: 307–311.<br />

126. Boulloche J, Mallet E, Mouterde O et al. Dystonic reactions<br />

with metoclopramide: is there a risk population?<br />

Helv Paediatr Acta 1987; 42: 425–432.<br />

127. Reeves JJ, Shannon MW, Fleisher GR. Ondansetron<br />

decreases vomiting associated with acute gastroenteritis:<br />

a randomized, controlled trial. Pediatrics 2002; 109: e62.<br />

128. Ramsook C, Sahagan-Carreon I, Kozinetz CA et al. A<br />

randomized clinical trial comparing oral ondansetron<br />

with placebo in children with vomiting from acute<br />

gastroenteritis. Ann Emerg Med 2002; 39: 397–403.<br />

129. Manatsathit S, Dupont HL, Farthing M et al. Working<br />

Party of the Program Committee of the Bangkok World<br />

Congress of Gastroenterology 2002. Guideline for the<br />

management of acute diarrhea in adults. J Gastroenterol<br />

Hepatol 2002; 17 (Suppl): S54–S71.<br />

130. Ericsson CD, Johnson PC. Safety and efficacy of<br />

loperamide. Am J Med 1990; 88 (Suppl 6A): 10–14.<br />

131. Bhutta TI, Tahir KI. Loperamide poisoning in children.<br />

Lancet 1990; 335: 363.<br />

132. Minton NA, Smith PGD. Loperamide toxicity in a child<br />

after a single dose. BMJ 1987; 294: 1383.<br />

133. Herranz J, Luzuriaga C, Sarralle R et al. Neurological<br />

symptoms precipitated by loperamide. An Esp Pediatr<br />

1980; 13: 1117–1120.


134. Schwartz RH, Rodriquez WJ. Toxic delirium possibly<br />

caused by loperamide. J Pediatr 1991; 118: 656–657.<br />

135. Gilbert B, Lienhardt A, Palomera S et al. [A study of the<br />

effectiveness of smectite versus placebo or loperamide<br />

in acute infantile diarrhea]. Ann Pediatr (Paris) 1991; 38:<br />

633–636.<br />

136. Madkour AA, Madina EMH, El-Azzouni OEZ et al.<br />

Smectite in acute diarrhea in children: a double-blind<br />

placebo-controlled clinical trial. J Pediatr Gastroenterol<br />

Nutr 1993; 17: 176–181.<br />

137. Lachaux A, Danzon A, Collet JP et al. Acute infantile<br />

diarrhea. Role of treatment with smectite as complement<br />

to rehydration. Randomised double-blind study.<br />

International Rev Pediatr 1986; 163: 29–31.<br />

138. Ericsson CD, Tannenbaum C, Charles TT. Antisecretory<br />

and antiinflammatory properties of bismuth subsalicylate.<br />

Rev Infect Dis 1990; 12 (Suppl 1): S16–S20.<br />

139. Ericsson CD, Evans DG, DuPont HL et al. Bismuth<br />

subsalicylate inhibits activity of crude toxins of<br />

Escherichia coli and Vibrio cholerae. J Infect Dis 1977;<br />

136: 693–696.<br />

140. Manhart MD. In vitro antimicrobial activity of bismuth<br />

subsalicylate and other bismuth salts. Rev Infect Dis<br />

1990; 12 (Suppl 1): S11–S15.<br />

141. Soriano-Brucher H, Avendano P, O’Ryan M et al.<br />

Bismuth subsalicylate in the treatment of acute diarrhea<br />

in children: a clinical study. Pediatrics 1991; 87:<br />

S18–S27.<br />

142. Figueroa-Quintanilla D, Salazar-Lindo E, Sack RB et al.<br />

A controlled trial of bismuth subsalicylate in infants<br />

with acute watery diarrheal disease. N Engl J Med 1993;<br />

328: 1653–1658.<br />

143. Chowdhury HR, Yunus M, Zaman K et al. The efficacy<br />

of bismuth subsalicylate in the treatment of acute diarrhoea<br />

and the prevention of persistent diarrhoea. Acta<br />

Paediatr 2001; 90: 605–610.<br />

144. Pickering LK, Feldman S, Ericsson CD et al. Absorption<br />

of salicylate and bismuth from a bismuth subsalicylatecontaining<br />

compound (Pepto-Bismol). J Pediatr 1981;<br />

99: 654–656.<br />

145. Primi MP, Bueno L, Baumer P et al. Racecadotril demonstrates<br />

intestinal antisecretory activity in vivo. Aliment<br />

Pharmacol Ther 1999; 13: 3–7.<br />

146. Salazar-Lindo E, Santisteban-Ponce J, Chea-Woo E et al.<br />

Racecadotril in the treatment of acute watery diarrhea<br />

in children. N Engl J Med 2000; 343: 463–467.<br />

147. Cézard JP, Duhamel JF, Meyer M et al. Efficacy and tolerability<br />

of racecadotril in acute diarrhea in children.<br />

Gastroenterology 2001; 120: 799–805.<br />

148. Guarner F, Schaafsma GJ. Probiotics. Int J Food Microbiol<br />

1998; 39: 237–238.<br />

149. Szajewska H, Mrukowicz J. Probiotics in the treatment<br />

and prevention of acute infectious diarrhea in infants<br />

and children: a systematic review of published randomized,<br />

double-blind, placebo controlled trials. J Pediatr<br />

Gastroenterol Nutr 2001; 33: S17–S25.<br />

150. Van Niel C, Feudtner C, Garrison MM et al.<br />

Lactobacillus therapy for acute infectious diarrhea in<br />

children: a meta-analysis. Pediatrics 2002; 109: 678–684.<br />

151. Rosenfeldt V, Michaelson KF, Jakobsen M et al. Effect of<br />

probiotic Lactobacillus strains in young children hospitalized<br />

with acute diarrhea. Pediatr Infect Dis J 2002; 21:<br />

411–416.<br />

152. Rosenfeldt V, Michaelson KF, Jakobsen M et al. Effect of<br />

probiotic Lactobacillus strains on acute diarrhea in a<br />

cohort of nonhospitalized children attending day-care<br />

centers. Pediatr Infect Dis J 2002; 21: 417–419.<br />

153. Kowalska-Duplaga K, Strus M, Heczko P et al. Lactobif ® ,<br />

a marketed probiotic product containing<br />

References 673<br />

Bifidobacterium ruminantium, was not effective in the<br />

treatment of acute rotavirus diarrhea in infants. J Pediatr<br />

Gastroenterol Nutr 2002; 34: 4 (ESPGHAN abstr).<br />

154. Szajewska H, Mrukowicz J. Probiotics and acute<br />

gastroenteritis in children. Critical review of published<br />

evidence. Ann Nestle 2003; 61: 66–78.<br />

155. Jacobs J, Jonas WB, Jimenez-Perez M et al. Homeopathy<br />

for childhood diarrhea: combined results and metanalysis<br />

from three randomized, controlled clinical trials.<br />

Pediatr Infect Dis J 2003; 22: 229–234.<br />

156. Ghisham FK. Transport of electrolytes, water and<br />

glucose in zinc deficiency. J Pediatr Gastroenterol Nutr<br />

1984; 3: 608–612.<br />

157. Patrick J, Golden BE, Golden MHN. Leucocyte sodium<br />

transport and dietary zinc in protein energy malnutrition.<br />

Am J Clin Nutr 1980; 33: 617–620.<br />

158. Patrick J, Michael J, Golden MN et al. Effect of zinc on<br />

leukocyte sodium transport in vivo. Clin Sci Mol Med<br />

1978; 54: 585–587.<br />

159. Roy SK, Behrens RH, Haider R et al. Impact of zinc<br />

supplementation on intestinal permeability in<br />

Bangladeshi children with acute diarrhea and persistent<br />

diarrhea syndrome. J Pediatr Gastroenterol Nutr 1992;<br />

15: 289–296.<br />

160. Elmes ME, Jones JG. Ultrastructural changes in the<br />

small intestines of zinc deficient rats. J Pathol 1980;<br />

130: 37–43.<br />

161. Gebhard RL, Karouani R, Prigge WF et al. Effect of<br />

severe zinc deficiency on activity of intestinal disaccharidases<br />

and 3-hydroxy-3-methylglutaryl coenzyme A<br />

reductase in the rat. J Nutr 1983; 113: 855–859.<br />

162. Jones PE, Peters TJ. Oral zinc supplements and nonresponsiveness<br />

coeliac syndrome: effect on jejunal<br />

morphology, enterocyte production, and brush border<br />

disaccharidase activities. Gut 1981; 22: 194–198.<br />

163. Cunningham-Rundles C, Cunningham-Rundles S,<br />

Garofalo J. Increased T lymphocyte function and<br />

thymopoietin following zinc repletion in man. Fed Proc<br />

1979; 38: 1222.<br />

164. Fenwick PK, Aggett PJ, McDonald D et al. Zinc deficiency<br />

and zinc depletion effect on the response of rats<br />

to infection with Trichinella spiralis. Am J Clin Nutr<br />

1996; 52: 166–172.<br />

165. Schlesinger L, Arevalo M, Arredondo S et al. Effects of a<br />

zinc fortified formula on immunocompetence and<br />

growth of malnourished infants. Am J Clin Nutr 1992;<br />

56: 491–498.<br />

166. Shankar AH, Prasad AS. Zinc and immune function: the<br />

biological basis of altered resistance to infection. Am J<br />

Clin Nutr 1998; 68: 447S–463S.<br />

167. Wolman SL, Anderson GH, Marliss EB et al. Zinc in<br />

total parenteral nutrition: requirements and metabolic<br />

effects. Gastroenterology 1979; 76: 458–467.<br />

168. Castillo-Duran C, Vial P, Uauy R. Trace mineral balance<br />

during acute diarrhea in infants. J Pediatr 1988; 113:<br />

452–457.<br />

169. Sachdev HPS, Mittal NK, Mittal SK et al. A controlled<br />

trial on utility of oral zinc supplementation in acute<br />

dehydrating diarrhea in infants. J Pediatr Gastroenterol<br />

Nutr 1988; 7: 877–881.<br />

170. Roy SK, Tomkins AM, Akramuzzaman SM et al.<br />

Randomized controlled trial of zinc supplementation in<br />

malnourished Bangladeshi children with acute diarrhea.<br />

Arch Dis Child 1997; 77: 196–200.<br />

171. Hidayat A, Achadi A, Sunoto, Soedarma SP. The effect<br />

of zinc supplementation in children under three years<br />

of age with acute diarrhea in Indonesia. Med J Indonesia<br />

1998; 7: 237–241.


674<br />

Approach to the child with acute diarrhea<br />

172. Faruque ASG, Mahalanabis D, Haque SS et al. Double<br />

blind, randomized controlled trial of zinc or vitamin A<br />

supplementation in young children with acute diarrhea.<br />

Acta Pediatr 1999; 88: 154–160.<br />

173. Sazawal S, Black RE, Bhan MK et al. Zinc supplementation<br />

in young children with acute diarrhea in India. N<br />

Engl J Med 1995; 333: 839–844.<br />

174. Dutta P, Mitra U, Datta A et al. Impact of zinc supplementation<br />

in malnourished children with acute diarrhoea.<br />

J Trop Pediatr 2000; 46: 259–263.<br />

175. Bahl R, Bhandari N, Saksena M et al. Efficacy of zincfortified<br />

oral rehydration solution in 6- to 35-month-old<br />

children with acute diarrhea. J Pediatr 2002; 141:<br />

677–682.<br />

176. Haffejee IE. Effect of oral folate on duration of acute<br />

infantile diarrhoea. Lancet 1988; 2: 334–335.<br />

177. Ashraf H, Rahman MM, Fuchs GJ et al. Folic acid in the<br />

treatment of acute watery diarrhoea in children: a<br />

double-blind, randomized, controlled trial. Acta<br />

Paediatr 1998; 87: 1113–1115.<br />

178. Bogstedt AK, Johansen K, Hatta H et al. Passive immunity<br />

against diarrhoea. Acta Paediatr 1996; 85: 125–128.<br />

179. Sarker SA, Casswall TH, Mahalanabis D et al.<br />

Successful treatment of rotavirus diarrhea in children<br />

with immunoglobulin from immunized bovine<br />

colostrums. Pediatr Infect Dis J 1998; 17: 1149–1154.<br />

180. Mitra AK, Mahalanabis D, Ashraf H et al. Hyperimmune<br />

cow colostrum reduces diarrhoea due to rotavirus: a<br />

double-blind, controlled clinical trial. Acta Paediatr<br />

1995; 84: 996–1001.<br />

181. Guarino A, Canani RB, Russo S et al. Oral immunoglobulins<br />

for treatment of acute rotaviral gastroenteritis.<br />

Pediatrics 1994; 93: 12–16.<br />

182. Sarker SA, Casswall TH, Juneja LR et al. Randomized,<br />

placebo-controlled, clinical trial of hyperimmunized<br />

chicken egg yolk immunoglobulin in children with<br />

rotavirus diarrhea. J Pediatr Gastroenterol Nutr 2001; 32:<br />

19–25.<br />

183. Casswall TH, Sarker SA, Faruque SM et al. Treatment of<br />

enterotoxigenic and enteropathogenic Escherichia coliinduced<br />

diarrhoea in children with bovine<br />

immunoglobulin milk concentrate from hyperimmunized<br />

cows: a double-blind, placebo-controlled, clinical<br />

trial. Scand J Gastroenterol 2000; 35: 711–718.<br />

184. Ashraf H, Mahalanabis D, Mitra AK et al. Hyperimmune<br />

bovine colostrum in the treatment of shigellosis in children:<br />

a double-blind, randomized, controlled trial. Acta<br />

Paediatr 2001; 90: 1373–1378.<br />

185. Bennish ML, Salam MA, Haider R et al. Therapy for<br />

shigellosis. II. Randomized, double-blind comparison of<br />

ciprofloxacin and ampicillin. J Infect Dis 1990; 162:<br />

711–716.<br />

186. Bhattacharya SK, Bhattacharya MK, Dutta P et al.<br />

Randomized clinical trial of norfloxacin for shigellosis.<br />

Am J Trop Med Hyg 1991; 45: 683–687.<br />

187. Bennish ML, Salam MA, Khan WA et al. Treatment of<br />

shigellosis: III. Comparison of one- or two-dose<br />

ciprofloxacin with standard 5-day therapy. A randomized,<br />

blinded trial. Ann Intern Med 1992; 117: 727–734.<br />

188. Bassily S, Hyams KC, el-Masry NA et al. Short-course<br />

norfloxacin and trimethoprim–sulfamethoxazole treatment<br />

of shigellosis and salmonellosis in Egypt. Am J<br />

Trop Med Hyg 1994; 51: 219–223.<br />

189. Khan WA, Seas C, Dhar U et al. Treatment of shigellosis:<br />

V. Comparison of azithromycin and ciprofloxacin.<br />

A double-blind, randomized, controlled trial. Ann Intern<br />

Med 1997; 126: 697–703.<br />

190. Tauxe RV, Puhr ND, Wells JG et al. Antimicrobial resistance<br />

of Shigella isolates in the USA: the importance of<br />

international travelers. J Infect Dis 1990; 162:<br />

1107–1111.<br />

191. Salam MA, Bennish ML. Antimicrobial therapy for<br />

shigellosis. Rev Infect Dis 1991; 13 (Suppl): 41.<br />

192. Eidlitz-Marcus T, Cohen YH, Nussinovitch M et al.<br />

Comparative efficacy of two- and five-day courses of<br />

ceftriaxone for treatment of severe shigellosis in children.<br />

J Pediatr 1993; 123: 822–824.<br />

193. Neill MA, Opal SM, Heelan J et al. Failure of<br />

ciprofloxacin to eradicate convalescent fecal excretion<br />

after acute salmonellosis: experience during an outbreak<br />

in health care workers. Ann Intern Med 1991; 114:<br />

195–199.<br />

194. Nelson JD, Kusmiesz H, Jackson LH et al. Treatment of<br />

Salmonella gastroenteritis with ampicillin, amoxicillin,<br />

or placebo. Pediatrics 1980; 65: 1125–1130.<br />

195. Pegues DA, Hohmann EL, Miller SI et al. Salmonella,<br />

including S. typhi. In Blaser MJ, Smith PD, Ravdin JI,<br />

Greenberg HB, Guerrant RL, eds. Infections of the<br />

Gastrointestinal Tract. New York: Raven Press, 1995:<br />

785–809.<br />

196. Mandal BK. Treatment of multiresistant typhoid fever.<br />

Lancet 1990; 336: 1383.<br />

197. Soe GB, Oversturf GD. Treatment of typhoid fever and<br />

other systemic salmonelloses with cefotaxime, ceftriaxone,<br />

cefoperazone and other newer cephalosporins. Rev<br />

Infect Dis 1987; 9: 719–736.<br />

198. Mandal BK, Ellis ME, Dunbar EM et al. Double-blind<br />

placebo-controlled trial of erythromycin in the treatment<br />

of clinical Campylobacter infection. J Antimicrob<br />

Chemother 1984; 13: 619–623.<br />

199. Salazar-Lindo E, Sack RB, Chea-Woo E et al. Early treatment<br />

with erythromycin of Campylobacter jejuni-associated<br />

dysentery in children. J Pediatr 1986; 109:<br />

355–360.<br />

200. DuPont HL. Treatment of travelers’ diarrhea with<br />

trimethoprim/sulfamethoxazole and with trimethoprim<br />

alone. N Engl J Med 1982; 307: 841–844.<br />

201. Ericsson CD, Johnson PC, DuPont HL et al.<br />

Ciprofloxacin or trimethoprim–sulfamethoxazole as<br />

initial therapy for travelers’ diarrhea. A placebocontrolled,<br />

randomized trial. Ann Intern Med 1987; 106:<br />

216–220.<br />

202. Mattila L, Peltola H, Siitonen A et al. Short-term treatment<br />

of traveler’s diarrhea with norfloxacin: a doubleblind,<br />

placebo-controlled study during two seasons.<br />

Clin Infect Dis 1993; 17: 779–782.<br />

203. Thoren A, Wolde-Mariam T, Stintzing G et al.<br />

Antibiotics in the treatment of gastroenteritis caused by<br />

enteropathogenic Escherichia coli. J Infect Dis 1980 141:<br />

27–31.<br />

204. Murphy GS, Bodhidatta L, Echeverria P et al.<br />

Ciprofloxacin and loperamide in the treatment of bacillary<br />

dysentery. Ann Intern Med 1993; 118: 582–586.<br />

205. Prado D, Lopez E, Liu H et al. Ceftibuten and trimethoprim–sulfamethoxazole<br />

for treatment of shigella and<br />

enteroinvasive Escherichia coli disease. Pediatr Infect<br />

Dis J 1992; 11: 644–647.<br />

206. Cimolai N, Morrison BJ, Carter JE. Risk factors for the<br />

central nervous system manifestations of gastroenteritisassociated<br />

hemolytic–uremic syndrome. Pediatrics 1992;<br />

90: 616–621.<br />

207. Pavia AT, Nichols CR, Green DP et al. Hemolytic–uremic<br />

syndrome during an outbreak of Escherichia coli<br />

0157:H7 infections in institutions for mentally retarded<br />

persons: clinical and epidemiologic observations. J<br />

Pediatr 1990; 116: 544–551.<br />

208. Cimolai N, Carter JE, Morrison BJ et al. Risk factors for<br />

the progression of Escherichia coli 0157:H7 enteritis to


hemolytic–uremic syndrome. J Pediatr 1990; 116:<br />

589–592.<br />

209. Ostroff SM, Kobayashi JM, Lewis JH. Infections with<br />

Escherichia coli 0157:H7 in Washington State: the first<br />

year of statewide disease surveillance. JAMA 1989; 262:<br />

355–359.<br />

210. Proulx F, Turgeon JP, Delage G et al. Randomized,<br />

controlled trial of antibiotic therapy for Escherichia coli<br />

O157:H7 enteritis. J Pediatr 1992; 121: 299–303.<br />

211. Carter AO, Borczyk AA, Carlson JA et al. A severe<br />

outbreak of Escherichia coli O157:H7-associated hemorrhagic<br />

colitis in a nursing home. N Engl J Med 1987;<br />

317: 1496–1500.<br />

212. Riley LW, Remis RS, Helgerson SD et al. Hemorrhagic<br />

colitis associated with a rare Escherichia coli serotype.<br />

N Engl J Med 1983; 308: 681–685.<br />

213. Brenden RA, Miller MA, Janda JM. Clinical disease<br />

spectrum and pathogenic factors associated with<br />

Plesiomonas shigelloides infections in humans. Rev<br />

Infect Dis 1988; 10: 303–316.<br />

214. Holmberg SD, Farmer JJD. Aeromonas hydrophila and<br />

Plesiomonas shigelloides as causes of intestinal infections.<br />

Rev Infect Dis 1984; 6: 633–639.<br />

215. Holmberg SD, Wachsmuth IK, Hickman-Brenner FW et<br />

al. Plesiomonas enteric infections in the United States.<br />

Ann Intern Med 1986; 105: 690–694.<br />

216. Nathwani D, Laing RB, Harvey G et al. Treatment of<br />

symptomatic enteric Aeromonas hydrophila infection<br />

with ciprofloxacin. Scand J Infect Dis 1991; 23: 653–654.<br />

217. Reinhardt JF, George WL. Plesiomonas shigelloides-associated<br />

diarrhea. JAMA 1985; 253: 3294–3295.<br />

218. Cover TL, Aber RC. Yersinia enterocolitica. N Engl J Med<br />

1989; 321: 16–24.<br />

219. Ostroff SM, Kapperud G, Lassen J et al. Clinical features<br />

of sporadic Yersinia enterocolitica infections in Norway.<br />

J Infect Dis 1992; 166: 812–817.<br />

220. Pai CH, Gillis F, Tuomanen E et al. Placebo-controlled<br />

double-blind evaluation of trimethoprim–sulfamethoxazole<br />

treatment of Yersinia enterocolitica gastroenteritis. J<br />

Pediatr 1984; 104: 308–311.<br />

221. Robins-Browne RM, Prpic JK. Effects of iron and desferrioxamine<br />

on infections with Yersinia enterocolitica.<br />

Infect Immun 1985; 47: 774–779.<br />

222. Scavizzi M. Yersinia enterocolitica. In Yu VL, Merlgan<br />

TC, Barriere SL, eds. Antimicrobial Therapy and<br />

Vaccines. Baltiomore: Williams & Wilkins, 1999:<br />

481–488.<br />

223. Alam AN, Alam NH, Ahmed T et al. Randomised<br />

double blind trial of single dose doxycycline for treating<br />

cholera in adults. BMJ 1990; 300: 1619–1621.<br />

224. Bhattacharya SK, Bhattacharya MK, Dutta P et al.<br />

Double-blind, randomized, controlled clinical trials of<br />

norfloxacin for cholera. Antimicrob Agents Chemother<br />

1990; 34: 939–940.<br />

225. Gotuzzo E, Seas C, Echevarria J et al. Ciprofloxacin for<br />

the treatment of cholera: a randomized, double-blind,<br />

controlled clinical trial of a single daily dose in<br />

Peruvian adults. Clin Infect Dis 1995; 20: 1485–1490.<br />

226. Khan WA, Begum M, Salam MA et al. Comparative trial<br />

of five antimicrobial compounds in the treatment of<br />

cholera in adults. Trans R Soc Trop Med Hyg 1995; 89:<br />

1036.<br />

References 675<br />

227. Khan WA, Bennish ML, Seas C et al. Randomised<br />

controlled comparison of single-dose ciprofloxacin and<br />

doxycycline for cholera caused by Vibrio cholerae 01 or<br />

0139. Lancet 1996; 348: 296–300.<br />

228. Khan WA, Saha D, Rahman A et al. Comparison of<br />

single-dose azithromycin and 12-dose, 3-day<br />

erythromycin for childhood cholera: a randomised,<br />

double-blind trial. Lancet 2002; 360: 1722–1727.<br />

229. Fekety R. Guidelines for the diagnosis and management<br />

of Clostridium difficile-associated diarrhea and colitis.<br />

American College of Gastroenterology Practice<br />

Parameters Committee. Am J Gastroenterol 1997; 92:<br />

739–750.<br />

230. Gerding DN, Johnson S, Peterson LR et al. Clostridium<br />

difficile-associated diarrhea and colitis. Infect Control<br />

Hosp Epidemiol 1995; 16: 459–477.<br />

231. Teasley DG, Olson MM, Gebhard RL et al. Prospective<br />

randomised trial of metronidazole versus vancomycin<br />

for Clostridium difficile-associated diarrhoea and colitis.<br />

Lancet 1983: 1043–1046.<br />

232. Olson MM, Shanholtzer CJ, Lee JT Jr, Gerding DN. Ten<br />

years of prospective Clostridium difficile-associated<br />

disease surveillance and treatment at the Minneapolis<br />

VA Medical Center, 19821991. Infect Control Hosp<br />

Epidemiol 1994; 15: 371–381.<br />

233. Teasley DG, Olson MM, Gebhard RL et al. Prospective<br />

randomised trial of metronidazole versus vancomycin<br />

for Clostridium difficile-associated diarrhoea and colitis.<br />

Lancet 1983; 2: 1043–1046.<br />

234. Wenisch C, Parschalk B, Hasenhundl M et al.<br />

Comparison of vancomycin, teicoplanin, metronidazole,<br />

and fusidic acid for the treatment of Clostridium<br />

difficile-associated diarrhea [published erratum appears<br />

in Clin Infect Dis 1996; 2: 423]. Clin Infect Dis 1996; 22:<br />

813–818.<br />

235. Bassily S, Farid Z, Mikhail JW et al. The treatment of<br />

Giardia lamblia infection with mepacrine, metronidazole<br />

and furazolidone. J Trop Med Hyg 1970; 73: 1518.<br />

236. Lerman SJ, Walker RA. Treatment of giardiasis: literature<br />

review and recommendations. Clin Pediatr (Phila)<br />

1982; 21: 409–414.<br />

237. Levi GC, de Avila CA, Amato NV. Efficacy of various<br />

drugs for treatment of giardiasis. A comparative study.<br />

Am J Trop Med Hyg 1977; 26: 564–565.<br />

238. Zaat JOM, Mank ThG, Assendelft WJJ. Drugs for treating<br />

giardiasis (Cochrane Review). In The Cochrane Library,<br />

Issue 2. Oxford: Update Software.<br />

239. Gerding DN, Johnson S, Peterson LR et al. Clostridium<br />

difficile associated diarrhea and colitis. Infect Control<br />

Hosp Epidemiol 1995; 16: 459–477.<br />

240. Madico G, McDonald J, Gilman RH et al. Epidemiology<br />

and treatment of Cyclospora cayetanensis infection in<br />

Peruvian children. Clin Infect Dis 1997; 24: 977–981.<br />

241. Rubidge CJ, Scragg JN, Powell SJ. Treatment of children<br />

with acute amoebic dysentery. Comparative trial of<br />

metronidazole against a combination of dehydroemetine,<br />

tetracycline, and diloxanide furoate. Arch Dis Child<br />

1970; 45: 196–197.<br />

242. Scott F, Miller MJ. Trials with metronidazole in amebic<br />

dysentery. JAMA 1970; 211: 118–120.<br />

243. Haque R, Huston CD, Hughes M et al. Amebiasis. N<br />

Engl J Med 2003; 348: 1565–1573.


40<br />

Introduction<br />

Approach to the child with<br />

acute abdomen<br />

Luigi Dall’Oglio, Paola De Angelis and Giovanni Federici di Abriola<br />

‘Acute abdomen’ is a loose concept, referring to a<br />

medical emergency involving signs and symptoms<br />

pertaining to the abdomen and whose therapeutic<br />

option is usually surgery.<br />

In this chapter we review the clinical signs and<br />

symptoms of various gastrointestinal disorders<br />

that have an acute onset and can progress dramatically<br />

to involve the parietal or visceral peritoneum.<br />

Our aim is to help the pediatrician realize<br />

when a gastrointestinal condition changes from an<br />

indolent to an acute course, and when a surgeon<br />

should be consulted. We also describe some<br />

simple but important strategies that are helpful in<br />

reaching a diagnosis without delay, and in<br />

adequately preparing patients for either surgery or<br />

for one of the newer effective and less invasive<br />

non-surgical treatments.<br />

Symptoms and signs<br />

The symptoms and signs of an acute abdomen vary<br />

widely depending on the patient’s age and the clinical<br />

course of the disease. An early, efficient and<br />

accurate diagnosis depends crucially on a meticulous<br />

review of the patient’s medical history, focusing<br />

on the symptoms, their time of onset, and their<br />

intensity and character. To avoid a misdiagnosis of<br />

other extraintestinal diseases, the history taking<br />

must include symptoms apparently unrelated to<br />

the digestive tract. For example, a history of excessive<br />

thirst, increased urination and lack of appetite<br />

preceding the abdominal pain, accompanied by<br />

deep sighing and rapid breathing, and severe<br />

dehydration, may suggest diabetic ketoacidosis<br />

mimicking an acute abdomen. 1<br />

The most important elements to evaluate in a<br />

patient with an acute abdomen are:<br />

(1) Abdominal pain;<br />

(2) The presence and characteristics of vomiting;<br />

(3) The presence and characteristics of fever;<br />

(4) The characteristics of the stools;<br />

(5) The findings from a detailed physical examination.<br />

Abdominal pain<br />

Whereas in older, co-operative children, close<br />

observation and a detailed diagnostic work-up of<br />

abdominal pain will usually identify various possible<br />

causes of an acute abdomen, in infants and<br />

newborns, abdominal pain is often a poorly<br />

specific symptom. Young infants with functional<br />

abdominal colic, at times very painful, need only<br />

be kept under clinical observation, whilst those<br />

with abdominal pain of organic origin should<br />

undergo aggressive diagnostic and therapeutic<br />

procedures. In newborns and infants, a true acute<br />

abdomen is rare and is generally related to congenital<br />

malformation. It should be suspected when the<br />

patient presents with abdominal tension, paradoxical<br />

diarrhea and vomiting, sometimes accompanied<br />

by a surprisingly rapid decline in general<br />

condition followed by shock. Circumstances such<br />

as these require therapy for shock, along with<br />

diagnostic investigations.<br />

In older children, a complete medical history and<br />

description of the symptoms can generally be<br />

obtained. The suspected diagnosis can then be<br />

confirmed with standard clinical examinations<br />

and laboratory tests.<br />

677


678<br />

Approach to the child with acute abdomen<br />

The assessment of abdominal pain should include<br />

the intensity of pain, the type (continuous or colic)<br />

and the site – whether the pain affects a specific<br />

site or the entire abdomen, and whether it<br />

irradiates to the back, chest, or shoulders. The<br />

chronological pattern of the pain, the rapidity of<br />

onset and the progression and duration of<br />

symptoms can be useful in the diagnosis; pain<br />

with sudden and severe onset is generally due to<br />

perforation or mesenteric infarction. In gastroenteritis,<br />

pain is self-limited; in appendicitis, or in<br />

other peritoneal inflammations, pain is progressive.<br />

Infants with bowel occlusion typically have<br />

evident colicky pain and even more evident postcolic<br />

seediness.<br />

As a general rule, visceral pain (noxious stimuli in<br />

the intestinal wall) is located in the epigastrium,<br />

mid-abdomen and hypogastrium; the pain is<br />

generally crampy, burning or gnawing in quality.<br />

Parietal pain (noxious stimuli in the parietal peritoneum)<br />

is generally more intense and more<br />

precisely localized to the site of the lesion, and it<br />

is often aggravated by movement or coughing. The<br />

onset of pain differs in the various diseases. For<br />

example, in patients with perforated ulcers or<br />

ruptured abscess, pain has a sudden (instantaneous)<br />

onset; in strangulated bowel and intussusception,<br />

it has a rapid onset (minutes); and in<br />

appendicitis, bowel obstruction, cholecystitis, and<br />

Meckel’s diverticulitis, it has a gradual onset (a few<br />

hours). 2 Close observation, absolutely crucial in<br />

infants to gain information on the pain, is also<br />

necessary in the older child. A patient lying still in<br />

bed, in the fetal position, neither moving nor<br />

speaking, with a distressed facial expression, is<br />

likely to have peritonitis; conversely, a patient who<br />

has short periods free of pain, recognizable by<br />

normal appearance and behavior, could have<br />

visceral pains due to gastroenteritis or bowel<br />

occlusion. 3 Intestinal volvulus generally starts<br />

with intense abdominal pain and, after possible<br />

vascular torsion, the most important symptom is<br />

the worsening of the general condition; vomiting<br />

and blood in stools are frequent, without other<br />

characteristic clinical indications.<br />

Vomiting<br />

An important symptom in clarifying the severity<br />

and type of disease, vomiting is an early sign of<br />

peritoneal inflammation or bowel obstruction.<br />

Initially, vomit is mixed with food; after minutes or<br />

hours, it can become bile-stained, as in intestinal<br />

obstruction. Vomiting can be highly dangerous,<br />

owing to the risk of aspiration; especially in<br />

newborn or in unconscious infants, a nasogastric<br />

tube must be inserted for adequate gastric suction.<br />

Fever<br />

An unspecific symptom, fever, when present, can<br />

be helpful in suspected peritonitis. As a general<br />

rule, a temperature that rises slowly and progressively<br />

in parallel with the abdominal signs<br />

indicates peritoneal infection. Conversely, a<br />

temperature that rises rapidly, despite acute<br />

abdominal pain, indicates other, and often selflimited,<br />

non-surgical diseases.<br />

Characteristics of the stools<br />

The rule that adults with acute abdomen usually<br />

have no passage of stools does not hold true in<br />

infants and young children. The important signs to<br />

note in infants are: mucousy or bloody diarrhea,<br />

and the time elapsing after the last bowel movement<br />

and flatus. Bloody diarrhea is a common<br />

symptom of bowel occlusion with vascular<br />

damage due to strangulation. Diarrhea often<br />

accompanies peritoneal inflammation in small<br />

children. Older children with pelvic abscess also<br />

often have repeated small bowel movements.<br />

Complete information about stool habits before the<br />

onset of the acute abdominal symptoms is useful.<br />

For instance, a history of alternating diarrhea and<br />

constipation may be an indication of aganglionosis,<br />

while a history of bloody and mucousy<br />

stools could raise a suspicion of inflammatory<br />

bowel disease (IBD).<br />

Findings from the physical exam of the<br />

abdomen<br />

A careful physical examination, looking for the<br />

various signs of extra-abdominal systemic disease,<br />

is of paramount importance. For example, cutaneous<br />

petechiae can raise a suspicion of<br />

Henoch–Schönlein purpura and possible bowel<br />

involvement with intussusception. The abdomen


must be meticulously inspected, looking for<br />

distension; visible veins; visible loops of bowel,<br />

with or without peristaltic waves; superficial nonabdominal<br />

breathing; and flexion of lower limbs<br />

over the abdomen. All these signs can be more or<br />

less evident, and can differ in intensity, depending<br />

on the patient’s age, the disease and its severity.<br />

The same problems arise during abdominal palpation.<br />

In a newborn, insufficient muscular mass<br />

and muscle activity makes rigidity from muscular<br />

guarding difficult to detect. The only way to obtain<br />

information from abdominal palpation in a child is<br />

to approach the child carefully, and begin palpation<br />

at the least tender site; light finger percussion<br />

over an area of tenderness often elicits the same<br />

information as deep palpation and is less disturbing<br />

to infants and children. 4 Abdominal palpation<br />

with the child lying on the side, with one or both<br />

legs on the abdomen itself, is a helpful maneuver<br />

for obtaining muscular relaxation, thus gaining<br />

more information. Percussion can be useful to<br />

elicit tympany from intraluminal abdominal gas,<br />

as in obstruction, or from extraluminal gas, as in<br />

intestinal perforation.<br />

Careful rectal examination yields a great deal of<br />

information about anal or rectal malformations,<br />

stools, gas, blood, mucus and pelvic tenderness or<br />

masses.<br />

Laboratory examination, imaging and<br />

other techniques<br />

Laboratory examination<br />

All patients with suspected acute abdomen should<br />

have a complete blood count with differential, Creactive<br />

protein (CRP) and urinalysis. Analysis of<br />

serum electrolytes, blood urea nitrogen, creatinine<br />

and glucose levels is useful in determining fluid<br />

and acid–base status, renal function and metabolic<br />

status. In newborns and infants the standard laboratory<br />

tests sometimes have poor sensitivity and<br />

specificity, but they are important for identifying<br />

pre-shock signs early, before the clinical status<br />

becomes critical.<br />

The patient’s clinical history and physical examination<br />

may indicate the need for special tests (e.g.<br />

liver function tests and amylase in patients with<br />

upper abdominal symptoms).<br />

Laboratory examination, imaging and other techniques 679<br />

Imaging<br />

Symptoms of acute abdomen generally arise from<br />

bowel occlusion or peritoneal inflammation. Plain<br />

abdominal X-rays and ultrasonography are the two<br />

most common imaging techniques. Plain abdominal<br />

X-rays visualize free air, as in perforation, and<br />

the possible presence of air–fluid levels, pathognomonic<br />

for occlusion. They also provide evidence<br />

of various pre-surgical problems, such as pneumatosis<br />

in necrotizing enterocolitis, and toxic<br />

megacolon which need to be recognized early, in<br />

order to avoid delaying surgery.<br />

Plain abdominal series must always be taken in<br />

supine, upright and side views; in patients unable<br />

to stand in this position, a lateral decubitus film<br />

with the left side down (or cross-table lateral) can<br />

be useful to identify abnormal gas distribution. A<br />

chest X-ray must be obtained to exclude possible<br />

respiratory disease; lung-base pneumonia is a<br />

well-known cause of acute abdominal symptoms,<br />

whereas acute abdominal disease can give rise to<br />

chest problems, as it does in pancreatitis or<br />

subphrenic abscess. No recently published trials<br />

have addressed the diagnostic value of plain<br />

series, but in accordance with Glasgow and<br />

Mulvihill, 3 ‘it is a readily available and inexpensive<br />

examination and in most circumstances<br />

should be obtained’.<br />

Ultrasound gives rapid, non-invasive, accurate,<br />

repeatable and inexpensive information. Unlike<br />

plain series, the ultrasound technique needs an<br />

experienced examiner who knows precisely which<br />

information to seek in the abdomen and how to<br />

look for it. Ultrasound allows accurate detection of<br />

localized or diffuse liquid and its density; it also<br />

defines the morphology of other structures,<br />

including the biliary and pancreatic tree. This<br />

technique accurately estimates the intestinal wall<br />

thickness, and visualizes peristaltic activity. In<br />

conjunction with color and pulsed Doppler<br />

imaging, ultrasound provides a new tool for diagnosing<br />

an acute abdomen, depending on the<br />

child’s age. 5 It has a well-known role in IBD and in<br />

other inflammatory diseases, including intestinal<br />

localizations in Henoch–Schönlein purpura. It also<br />

visualizes hydrostatic reduction of intussusception<br />

with saline enema. 6 The primary imaging<br />

procedure 7 in abdominal emergency in childhood<br />

is: plain X-ray film of the abdomen followed by


680<br />

Approach to the child with acute abdomen<br />

ultrasound. The findings from these initial studies<br />

dictate the need for further imaging.<br />

The role of computed tomography (CT) and<br />

magnetic resonance imaging (MRI) remains less<br />

clear. These techniques are expensive and, in<br />

uncooperative children, deep sedation is needed to<br />

obtain the clear images necessary for a meaningful<br />

assessment.<br />

In suspected biliary or pancreatic disease, MRI<br />

cholangiography gives full information about<br />

morphology 8,9 and, with secretin stimulus, about<br />

pancreatic activity. 10 Other invasive diagnostic<br />

procedures, including peritoneal lavage or diagnostic<br />

laparoscopy, are occasionally useful but are<br />

more often used in post-trauma acute abdomen.<br />

Digestive endoscopy is generally contraindicated<br />

in acute abdomen, although it may have a diagnostic<br />

or therapeutic role in special circumstances.<br />

These include decompression, reduction and<br />

sigmoidopexy of sigmoid volvulus, 11,12 or clip<br />

closure of cardia perforation in Mallory–Weiss<br />

tears, after dilatations 13,14 or after a lap Nissen<br />

procedure.<br />

General considerations about<br />

treatment<br />

A good strategy for avoiding misdiagnosis or surgical<br />

delay is to contact the surgeon as soon as an<br />

acute abdomen develops and maintain full and<br />

timely collaboration. While awaiting the surgeon,<br />

the pediatrician must complete the diagnostic<br />

work-up and start medical treatment. Recent<br />

advances in diagnosis and therapy mean that<br />

many patients who hitherto would have undergone<br />

surgery can now be treated medically.<br />

Surgical intervention must be scheduled only after<br />

the patient’s status has been fully stabilized and<br />

only if the non-surgical therapeutic strategy is<br />

unsuccessful. In doubtful circumstances, the pediatrician<br />

along with the surgeon should observe<br />

how the disease evolves clinically, in order to<br />

identify the correct timing for diagnostic procedures<br />

and to decide between conservative therapeutic<br />

options or surgical intervention. Should the<br />

diagnosis remain unclear and a life-threatening<br />

risk develop, then the surgeon may proceed to an<br />

exploratory laparotomy or, as in recent years and<br />

in selected cases, laparoscopy.<br />

The management of an acute abdomen also has to<br />

be cost-effective. Therefore, surgeons must be<br />

involved early in the evaluation phase of patients<br />

with an acute abdomen, and follow the patient<br />

during the observation period, so that unnecessary<br />

laboratory or diagnostic tests can be avoided and<br />

the costs reduced. 15,16 The goal in treating an acute<br />

abdomen is to reach an early diagnosis and to<br />

establish the correct surgical indications and<br />

timing to allow effective surgery.<br />

Unfortunately, these goals are not always<br />

achieved. Far more often, the clinical presentation<br />

and the indications for surgery are unclear. In<br />

these cases, the pediatrician must, in strict collaboration<br />

with the surgeon, do everything necessary<br />

to reach a diagnosis, and at the same time start<br />

supportive therapy before the patient’s general<br />

clinical status takes a downhill course. The following<br />

simple but essential procedures can help.<br />

Evaluation of fluid loss<br />

This is obtained by the use of nasogastric suction<br />

with a tube of adequate caliber. A simple report on<br />

the number of vomits and the presumed amount<br />

vomited does not give a precise evaluation of fluid<br />

loss. The evaluation requires adequate urine<br />

collection and diaper weight for fecal volume.<br />

Placement of an adequate intravenous line<br />

Especially in newborns and infants, a vein catheter<br />

must be installed in time to avoid difficult venous<br />

puncture under conditions of collapse. In patients<br />

whose clinical status is more severe, a central<br />

venous catheter can be useful, not only for infusion<br />

but also for measuring central venous pressure.<br />

This measurement is essential in ensuring a<br />

proper fluid intake.<br />

Consideration of pharmacological therapy<br />

Antibiotic therapy is always necessary in patients<br />

with clinically evident sepsis and is useful in those<br />

with well-diagnosed peritonitis before surgery. In<br />

other less clear circumstances, antibiotics might


hide the clinical evolution of the disease, thus<br />

delaying the definitive diagnosis and proper surgical<br />

therapy, and should therefore be avoided.<br />

The usefulness of H2-antagonists or proton pump<br />

inhibitors in the prevention of acute peptic disease<br />

is unclear in children and debated in adults.<br />

Persistent vomiting attempts can provoke prolapse<br />

of the gastric wall through the cardia, thus causing<br />

blood in the vomit, despite the absence of a<br />

specific gastric disease. 17,18 In patients whose clinical<br />

status worsens, 10 mg/kg per day of ranitidine<br />

or 1–1.5 mg/kg per day of omeprazole are useful in<br />

preventing acute peptic disease.<br />

Specific diseases<br />

Some specific diseases resulting in acute abdomen<br />

(such as appendicitis, intussusception, Meckel’s<br />

diverticulum, Hirschsprung’s disease or chronic<br />

intestinal pseudo-obstruction) are discussed in<br />

detail in their respective chapters. Here we focus<br />

only on some conditions that may atypically<br />

present the clinical picture of acute abdomen,<br />

offering some suggestions pertinent to their diagnosis<br />

and treatment.<br />

Foreign body ingestion<br />

Foreign body ingestion is rarely described as a<br />

cause of acute abdomen; children can ingest everything<br />

and these objects, 19 even button batteries, 20<br />

often pass naturally.<br />

Acute abdomen was described in patients who had<br />

ingested bezoars 21,22 especially in mentally<br />

retarded children, or in cases where multiple<br />

magnets were attracted to each other across the<br />

bowel wall, leading to pressure necrosis, perforation,<br />

fistula or intestinal obstruction. 23 Ingestion of<br />

pins has been described as a cause of gastric perforation.<br />

24<br />

In our experience of ingested foreign bodies, an<br />

acute abdomen occurred twice: duodenal perforation<br />

from a hairpin and sigmoid perforation from a<br />

fragment of glass from the top of a cola bottle. The<br />

first patient, who presented in the pre-endoscopy<br />

period, with the foreign bodies embedded in the<br />

third duodenal portion, died of pancreatitis and<br />

Specific diseases 681<br />

septic complications. In the second case, the<br />

foreign bodies passed naturally with a small<br />

amount of blood in the stools as a lucky warning<br />

sign, in a patient without abdominal pain; radiological<br />

evidence of free abdominal gas suggested<br />

the surgical direct suture of the 3-cm long sigmoid<br />

lesion, with a good outcome. Refer to Chapter 41<br />

for further information.<br />

Inflammatory bowel disease<br />

Acute surgical emergencies in children with IBD<br />

need a team approach, involving gastroenterologists<br />

and surgeons, avoiding an unnecessary or<br />

delayed surgical strategy. If the team chooses the<br />

correct time for surgery, acute abdomen from toxic<br />

colitis with megacolon, perforation, abdominal<br />

abscesses and bowel obstructions can have a good<br />

outcome, with morbidity and mortality rates of<br />

15% in childhood. 25 The goal of this team is to<br />

make sure that unduly prolonged medical therapy<br />

does not result in an emergency operation.<br />

Emergency surgery, not being planned in advance,<br />

may be performed by a surgical team to which the<br />

patient is unknown, and often involves a multistep<br />

operation.<br />

Toxic colitis<br />

Toxic colitis is an emergency life-threatening<br />

complication of IBD. Crohn’s disease was once<br />

thought to be secondary only to ulcerative colitis,<br />

but we now know that it may cause up to 50% of<br />

the cases. 26<br />

The pathophysiology in Crohn’s disease and<br />

ulcerative colitis is similar, with transmural vascular<br />

congestion, muscular disintegration with<br />

consequent colon atony and dilatation, and with<br />

very thin, deeply ulcerated walls. Histologically,<br />

the inflammation involves all layers of the colon,<br />

with myocyte degeneration, necrosis and the<br />

formation of granulation tissue, infiltrated by<br />

neutrophils, lymphocytes and plasma cells. 27 The<br />

etiology seems to be related to colonic muscle<br />

paralysis, secondary to the inflammatory infiltrate<br />

and destruction of the myenteric plexus. More<br />

recent case reports have described toxic megacolon<br />

involving nitric oxide, generated by<br />

macrophages and smooth muscle cells. 27,28 Once<br />

the diagnosis is suspected for worsening general


682<br />

Approach to the child with acute abdomen<br />

conditions, abdominal distension and tenderness,<br />

plain abdominal series may confirm the colonic<br />

distension and must be repeated every 12h in order<br />

to provide a close monitoring of its evolution.<br />

Medical treatment consists of antibiotics<br />

(ciprofloxacin, metronidazole), to avoid bacterial<br />

translocation through the bowel wall, high-dose<br />

intravenous corticosteroids and cyclosporin or,<br />

more recently, tacrolimus. A higher incidence of<br />

postoperative complications was described in nondeferrable<br />

surgery and high-dose corticosteroid<br />

therapy. 25,29,30 If the dilated colon persists, free<br />

perforation develops, or the clinical conditions fail<br />

to improve within 2 or 3 days, prompt surgery is<br />

indicated. 31 Mortality related to perforation may<br />

exceed 40%, but if surgery is completed before<br />

perforation, the mortality decreases to 2–8%. 27<br />

Perforation<br />

Perforation occurs in approximately 2% of patients<br />

with ulcerative colitis, and is generally associated<br />

with toxic colitis and megacolon. In a patient with<br />

toxic colitis without megacolon, Crohn’s disease<br />

must be suspected. 32 The diagnosis may be delayed<br />

because high-dose corticosteroids may mask the<br />

signs of peritonitis. The transmural damage in<br />

Crohn’s disease creates an inflammatory adhesion<br />

between the affected bowel segments and local<br />

structures with consequent sealed perforation. 32<br />

Owing to these adhesions, pneumoperitoneum is<br />

present in only 20% of patients with perforated<br />

Crohn’s disease, and in a smaller percentage of<br />

those with ileal perforations. 33,34 The surgical<br />

procedure in colon perforation with or without<br />

toxic colitis must be limited to total colectomy and<br />

terminal ileostomy, avoiding proctectomy because<br />

of the risk of blood loss, pelvic sepsis and pelvic<br />

nerve damage. 26 This strategy removes the diseased<br />

bowel and avoids anastomosis in critically ill<br />

patients. 35<br />

In Crohn’s disease, gastroduodenal perforations<br />

need debridement and primary repair; jejunal–ileal<br />

perforations require resection and primary anastomosis<br />

with associated intestinal diversion in unfavorable<br />

conditions. 36<br />

Intra-abdominal abscess<br />

Characteristic of Crohn’s disease, intra-abdominal<br />

abscess can give rise to an acute abdomen if the<br />

abscess ruptures into the peritoneal cavity. Often<br />

the abscesses are associated with a fistula,<br />

secondary to a distended loop of proximal bowel, to<br />

a distal stricture or to transmural ulceration of the<br />

diseased bowel. Ultrasound and CT are useful for<br />

identifying the type of abscess and its location.<br />

Because surgical drainage of an abscess leads to<br />

numerous severe complications, it should be<br />

avoided. The best options are medical treatment or<br />

percutaneous drainage guided by ultrasound or CT.<br />

If surgery cannot be deferred, and in patients with<br />

intestinal damage, it should aim only at draining<br />

the abscess and diverting the fecal stream, avoiding<br />

resection or other useless and dangerous maneuvers.<br />

37<br />

Intestinal obstruction<br />

Intestinal obstruction, generally in the small<br />

bowel, is the most common complication requiring<br />

surgical correction in Crohn’s disease, but is rarely<br />

related to acute abdomen; improved medical and<br />

nutritional treatment has now reduced the indications<br />

for surgical emergencies for bowel obstruction.<br />

If acute obstruction cannot be managed<br />

otherwise, surgery must be as conservative as<br />

possible, avoiding wide resection. In accordance<br />

with the conservative strategy for surgery in<br />

Crohn’s disease, the newer stricturoplasty techniques<br />

are providing increasingly good results and<br />

fewer complications than resective surgery, avoiding<br />

the risk of short-bowel syndrome due to<br />

repeated resections. 38–42<br />

Biliary and pancreatic diseases<br />

Biliary diseases<br />

Biliary diseases are uncommon in children and<br />

especially rare as a possible cause of acute<br />

abdomen. Suppurative cholecystitis or cholangitis,<br />

from malformations and cholelithiasis, must be<br />

suspected in patients who present with acute<br />

abdomen and pain in the right upper quadrant that<br />

irradiates to the right shoulder, and laboratory<br />

blood findings indicating biliary stasis. Also, jaundice<br />

is not an early symptom of biliary and pancreatic<br />

disease. Ultrasound, in skilled hands, can be<br />

useful in showing common bile duct or gallbladder<br />

dilatation with stones or biliary sludge; it will also<br />

identify choledochal malformations.


MRI cholangiography gives full information about<br />

morphology. 8,9 Endoscopic retrograde cholangiopancreatography<br />

(ERCP) can be helpful for<br />

morphological evaluation of the biliary and pancreatic<br />

ducts and for therapeutic options such as<br />

sphincterotomy, stone removal and drainage. 43–45<br />

Percutaneous transhepatic cholangiography,<br />

drainage and stenting of the biliary tree is another<br />

non-surgical diagnostic and therapeutic option.<br />

However, an urgent surgical strategy for biliary<br />

drainage must be considered if antibiotic, endoscopic<br />

or radiological treatment fails. Depending on<br />

the degree of inflammation, surgery generally<br />

involves simple biliary drainage through a cholecystotomy.<br />

In patients with choledochal malformation,<br />

definitive surgery must be postponed until the<br />

peritoneal infection has resolved, because of the<br />

high risk of complications (including anastomosis<br />

leak) in patients with active biliary peritonitis.<br />

Spontaneous perforation of the bile duct<br />

This is a rare pediatric cause of biliary peritonitis46,47<br />

that has been described in newborns and<br />

infants, frequently associated with a choledochal<br />

cyst or anomalous pancreaticobiliary union. 48,49 In<br />

the absence of a dilated biliary duct, diagnosis is<br />

difficult; sometimes, biliary peritonitis is an<br />

occasional finding at exploratory laparotomy. The<br />

presence of bile-like fluid at abdominal puncture<br />

nevertheless suggests this disorder. Patients<br />

without malformations or peritonitis should<br />

undergo primary repair, with biliary and abdominal<br />

drainage. Otherwise, the best choice is simple<br />

biliary and abdominal drainage, postponing definitive<br />

surgery.<br />

Acute pancreatitis<br />

Acute pancreatitis is a dramatic cause of acute<br />

abdomen; it is uncommon in children and, in the<br />

past, has been an occasional finding in acute<br />

abdomen operated upon for suspected appendicitis.<br />

50 In acute pancreatitis, abdominal pain,<br />

referred to the middle or epigastric abdomen, has<br />

an acute onset and is deep-seated, requiring<br />

adequate analgesic treatment; abdominal parietal<br />

tenderness is generally evident. A major decision in<br />

the management of severe acute pancreatitis is<br />

whether and when surgery is necessary for<br />

Infectious and parasitic disease 683<br />

pancreatic necrosis (see Chapter 21). ERCP, sphincterotomy<br />

and drainage can be useful as diagnostic<br />

and therapeutic options if hepatic enzyme<br />

abnormalities suggest biliary pancreatitis due to<br />

stones or sludge. 51,52 Somatostatin and octreotide<br />

are widely used but their efficacy is not<br />

proven. 53–55 The course of pancreatitis can be<br />

assessed by monitoring the patients’ general condition,<br />

fever, hematological variables (white blood<br />

cell (WBC) count, CRP, lactate dehydrogenase,<br />

calcium and albumin), and imaging findings (ultrasound,<br />

CT). 56 In patients with necrosis and, in<br />

particular, septic necrosis, surgical intervention<br />

must be considered for drainage of the peripancreatic<br />

space. 57<br />

Infectious and parasitic disease<br />

Typhoid perforation<br />

Typhoid perforation is a well-known surgical<br />

complication of Salmonella typhi infection, 58–61<br />

whose outcome is related to the number of perforations<br />

and to the severity of peritonitis. 58 This last<br />

point is obviously related to the timeliness of diagnosis.<br />

Because no specific warning signs are known for<br />

identifying a pre-perforation state, the diagnosis<br />

rests on close clinical observation. Unspecific laboratory<br />

indexes include elevated WBC count and<br />

CRP; plain abdominal series will usually identify<br />

pneumoperitoneum. Multiple therapeutic protocols<br />

and surgical strategies have been proposed,<br />

depending on the intensity of peritoneal involvement.<br />

Antibiotic therapy consists of aminoxide,<br />

metronidazole and the third-generation<br />

cephalosporins. 60 In patients without advanced<br />

peritonitis, and with a single perforated lesion,<br />

excellent results can be achieved by excising the<br />

edges followed by direct suture and, in multiple<br />

perforations, by segmental resection followed by<br />

end-to-end anastomosis. Patients presenting with<br />

severe peritonitis must be invariably treated by<br />

bowel diversion. 59 The most common postoperative<br />

complications are wound infection and enterocutaneous<br />

fistula. In a prospective study, the ideal<br />

treatment for typhoid enteric perforation was found<br />

to be resection anastomosis with copious peritoneal<br />

lavage. 62


684<br />

Approach to the child with acute abdomen<br />

Non-typhi Salmonella<br />

In this intestinal infection, toxic megacolon can<br />

occur. Patients with toxic megacolon have a toxic<br />

appearance, diarrhea, high fever (>39°C) and<br />

marked colon dilatation with maximal diameter<br />

>1.5 times the width of the vertebral body of the<br />

first lumbar vertebra (L1-VB). A retrospective<br />

analysis identified as the most significant factors<br />

associated with intestinal perforation: age >1 year,<br />

serum CRP >200mg/l; colon diameter >2.5 times<br />

the width of L1-VB; inadequate early hydration;<br />

and delay in inserting a rectal tube. 63<br />

Entamoeba histolytica<br />

Entamoeba histolytica is another infectious cause<br />

of colonic involvement; acute abdomen due to<br />

perforation is rarely reported and related to perforated<br />

appendicitis. The most frequent manifestations<br />

are acute intestinal symptoms and bleeding.<br />

64 In severely ill patients, with acute bleeding<br />

and general septic signs, misdiagnosis of fulminant-onset<br />

IBD could result in unnecessary colectomy.<br />

Intestinal tuberculosis<br />

This is now widespread and must be suspected in<br />

patients who present with perforation or occlusion-like<br />

symptoms, as well as in cases of<br />

suspected Crohn’s disease, given the similarities<br />

between these two conditions. Intestinal perforation<br />

and fistulae were described in 7% of tuberculosis<br />

patients in a large retrospective series. 65 The<br />

diagnosis of perforation, due to adhesion between<br />

the bowel loops, is not easy. In a retrospective<br />

series, most patients presented with non-specific<br />

clinical features; pneumoperitoneum on abdominal<br />

radiographs was present in only 48.3% of<br />

cases. 66 Surgery consists of resection, end-to-end<br />

anastomosis and peritoneal drainage.<br />

Intestinal parasites<br />

Intestinal parasites (besides Entamoeba histolytica)<br />

are rarely a cause of perforation or occlusion.<br />

Anisakiasis can be a surprising finding at surgery<br />

for bowel obstruction; these parasites can be<br />

suspected in children with acute abdomen caused<br />

by bowel obstruction, who have eaten raw or<br />

undercooked fish. Typical ultrasound findings in<br />

patients with suspected anisakiasis are ascites,<br />

small-bowel dilatation, focal edema of Kerckring<br />

folds, 67 markedly thickened bowel loops associated<br />

with luminal narrowing and decreased peristalsis.<br />

68 Cytological examination of a specimen<br />

obtained from ascites by abdominal puncture<br />

showed a dense infiltration of eosinophils. 68 If the<br />

foregoing ultrasound features are found, the diagnosis<br />

of intestinal anisakiasis must be considered,<br />

because medical treatment has a good outcome<br />

and will avoid unnecessary surgery.<br />

Mucormycosis<br />

Mucormycosis is an increasing concern in<br />

immunocompromised patients, in whom mortality<br />

exceeds 60%. The multiple mycotic localizations<br />

(intestinal wall, liver, kidneys, sinuses, lower<br />

respiratory tract, or skin) give a dramatic clinical<br />

picture with acute abdomen secondary to occlusion<br />

and multiple perforations. The last are due to<br />

vessel thrombosis. 69,70 The standard treatment is<br />

amphotericin B combined with surgical debridement<br />

if necessary, depending on occlusion or<br />

perforations. In our experience, in one patient with<br />

fourth-degree neuroblastoma, after the histological<br />

diagnosis of mucormycosis in the gastrointestinal<br />

tract (surgical treatment for multiple perforations)<br />

and the echographic detection of multiple mycotic<br />

localizations in the liver and kidneys, the treatment<br />

was high-dose amphotericin B. At the same<br />

time, to prevent the growth of hyphae, we avoided<br />

the development of anaerobiosis and/or acidosis,<br />

as well as hyperglycemia. This therapeutic strategy<br />

successfully prevents spreading of the infection<br />

and encourages complete healing. 70<br />

Acute abdomen caused by<br />

endoscopic and other diagnostic<br />

procedures<br />

Endoscopic procedures, even in skilled hands, can<br />

be dangerous and provoke complications sometimes<br />

resulting in acute abdomen. Many of these<br />

events can be prevented by using strictly pediatric<br />

instruments, by suspecting a complication before


it becomes irreversible and by instituting specific<br />

treatments without delay.<br />

Polypectomy<br />

Simple rules can help in preventing perforation<br />

that may become manifest during the procedure or<br />

some hours later. Delayed perforation is caused by<br />

deep scalding of the intestinal wall due to the use<br />

of an inappropriate blend of cutting and hemostatic<br />

current and excessively high current intensities.<br />

Incomplete snare closure with its tip hidden by the<br />

polyp can perforate the intestinal wall. Sometimes<br />

the perforation becomes evident during the procedure<br />

because of bleeding and, moreover, because<br />

insufflation of air fails to distend the bowel, thus<br />

causing abdominal tension with respiratory<br />

distress. In cases like this, the complication can be<br />

resolved by immediate surgical intervention with<br />

direct suture and drainage. Deep scalding can<br />

occur during multiple polypectomies or during<br />

resection of polyps with a large pedicle, especially<br />

those sited in the right colon or jejunum, where<br />

the wall is thinner than the gastric or left colon<br />

wall. Abdominal pain, fever, leukocytosis and<br />

abdominal wall tenderness can be evident even<br />

without perforation. In patients with radiological<br />

evidence of a large, increasing quantity of free<br />

abdominal gas, surgery might be necessary. If<br />

surgery is avoided, the lesion may still have a good<br />

outcome in 2 or 3 days, after treatment with broadspectrum<br />

antibiotics, metronidazole (22mg/kg per<br />

day) and strict observation with fasting.<br />

Colonoscopy in inflammatory bowel disease<br />

In IBD, an attempted total colonoscopy can be<br />

hazardous for the patient, owing to the fragility of<br />

the colonic wall. In our experience, in an infant<br />

with acute ulcerative colitis careful insufflation<br />

into the sigma caused perforation of the transverse<br />

colon. In order to prevent peritoneal infection, the<br />

patient must undergo emergency surgery to<br />

attempt a direct suture of the lesion; if the colon is<br />

extensively damaged a fecal diversion (ileostomy)<br />

may be required, with or without colectomy.<br />

In colonic strictures from Crohn’s disease, surgical<br />

resection or stricturoplasty is increasingly avoided<br />

Acute abdomen caused by endoscopic and other diagnostic procedures 685<br />

in favor of palliative therapy with pneumatic<br />

endoscopic dilatations. 71,72 Because of the wall<br />

thickness, perforations are rare and the surrounding<br />

scar tissue hinders the diffusion of gas and<br />

intestinal contents into the peritoneal cavity.<br />

Peritoneal involvement is therefore less severe but<br />

the diagnosis can be delayed. Helpful ways of<br />

preventing further peritoneal complications<br />

include diagnostic ultrasound or CT scans, and<br />

conservative treatment with antibiotic prophylaxis<br />

and strict observation.<br />

Percutaneous endoscopic gastrostomy<br />

To avoid complications related to percutaneous<br />

endoscopic gastrostomy (PEG), the operator must<br />

be familiar with its contraindications, relative and<br />

absolute. The most frequent cause of a PEG-related<br />

acute abdomen is air, gastric juice or food in the<br />

peritoneal cavity due to incomplete adhesion of<br />

the gastric to the abdominal wall. 73–75 During the<br />

procedure, especially in infants with bowel distension,<br />

inserting the needle can provoke an<br />

intestinal perforation that might require surgical<br />

intervention. 76 In psychiatric and neurologically<br />

impaired children, premature tube removal can<br />

provoke peritonitis requiring surgery for peritoneal<br />

drainage. In patients without peritonitis, surgical<br />

intervention can be avoided by carefully inserting<br />

a pigtail wire and replacing the drainage tube,<br />

under radiographic control, with a slimmer<br />

gastrostomy Foley catheter (Medical Innovation<br />

Corporation, Milpitas, CA; Bard Interventional<br />

Products, Tewksbury, MA, USA). During substitution<br />

of the PEG with a low-profile device, the<br />

gastric wall can become detached from the abdominal<br />

wall. In our experience, in a patient with<br />

cystic fibrosis, this complication developed after a<br />

sports injury to the abdomen, which caused<br />

dramatic invasion of food into the peritoneum and<br />

required open surgery.<br />

In certain clinical settings, such as peritoneal dialysis,<br />

this type of complication can be avoided by<br />

placing the gastrostomy surgically 77 or by using a<br />

special kit (Cliny, Create Medic Co, Ltd, Yokohama,<br />

Japan) which, under endoscopic control,<br />

allows adhesion between the gastric and abdominal<br />

wall. Previous abdominal surgery can expose<br />

patients to a high risk of gastrocolic fistula, and<br />

surgery is necessary only in cases of dramatic


686<br />

Approach to the child with acute abdomen<br />

disruption of the gastric or colonic wall, with peritoneal<br />

fecal contamination; otherwise antibiotic<br />

therapy and gastric drainage resolves the problem<br />

within days.<br />

Cardia and esophageal dilatations<br />

Pneumatic or semi-rigid endoscopic dilatations in<br />

achalasia, or congenital esophageal strictures, can<br />

provoke perforation of the cardia or of the abdominal<br />

esophagus and possible acute abdomen. In<br />

children with esophageal perforation and mediastinum<br />

involvement, current opinion favors the<br />

following standard management protocol: nasogastric<br />

and pharyngeal suction, wide-spectrum antibiotics,<br />

proton pump inhibitors, total parenteral<br />

nutrition and daily testing of CRP and WBCs.<br />

Endoscopic clip closure of the perforation and<br />

transesophageal drainage can be attempted to<br />

avoid surgery 13,14 (and D. Fregonese, personal<br />

communication). The main indications for surgery<br />

are worsening sepsis and insufficient drainage,<br />

with diffuse or localized multiple purulent collections.<br />

The general rule that purulent infection of<br />

the peritoneum contraindicates resections and<br />

reanastomosis is even more applicable in these<br />

cases. In cardia or abdominal esophageal perforation,<br />

an attempt to suture the lesion may require<br />

gastrostomy (in order to obtain full gastric<br />

drainage), jejunostomy (for early enteral nutrition)<br />

and multiple drainage in the subphrenic and<br />

posterior mediastinum on both sides of the<br />

abdomen. In non-responders, esophagostomy and<br />

esophagogastric disconnection can be life-saving<br />

maneuvers.<br />

Duodenal hematoma<br />

Symptomatic intramural duodenal hematoma<br />

secondary to duodenal biopsies obtained at<br />

endoscopy is a rare but sometimes dramatic complication<br />

leading to possible perforation, pancreatitis<br />

and occlusion-like symptoms. 78,79 The resulting<br />

obstruction is self-limited and typically<br />

resolves spontaneously within a week. Conversely,<br />

perforation and pancreatitis progress to acute<br />

abdomen. In a patient with suspected perforation,<br />

because the posterior duodenal wall lies in the<br />

retroperitoneal cavity, plain abdominal series<br />

cannot show free abdominal gas. Nor are abdomi-<br />

nal pain and muscular tenderness as obvious as<br />

they are in children with gastric or anterior<br />

duodenal wall perforation. Ultrasound shows the<br />

hematoma and, if present, improvement in pancreatic<br />

thickness, and a CT scan allows the diagnosis<br />

of perforation to be made. In patients with massive<br />

infected necrosis of the pancreatic parenchyma or<br />

retroperitoneal infection, surgical drainage is<br />

required.<br />

Endoscopic cholangiopancreatography<br />

Complications after ERCP are well known in children<br />

as in adults; their incidence, in diagnostic<br />

ERCP, is higher in children. 43 The therapeutic<br />

options are the same in both age groups. 44 Post-<br />

ERCP pancreatitis is also more frequent in children<br />

than in adults (8% vs. 3–7%). 45 Its main<br />

causes are papillary swelling owing to repeated<br />

cannulations; and pancreatic parenchymography<br />

due to excessive contrast injection. 80 A transient<br />

rise in serum amylase level, unaccompanied by<br />

pain or elevated WBC count or CRP level, is not<br />

diagnostic of pancreatitis. True pancreatitis with<br />

acute abdomen generally responds to conservative<br />

management; in our experience we have observed<br />

only one case of severe pancreatitis, which was<br />

conservatively managed but complicated by a<br />

pancreatic pseudocyst.<br />

Biliary tree or duodenal perforation is a rare<br />

complication that can be managed conservatively<br />

but needs strict clinical surveillance. In a newborn<br />

with suspected biliary hypoplasia we observed a<br />

post-ERCP duodenal perforation, near the papilla,<br />

that responded well to non-surgical therapy.<br />

Rectal suction biopsy<br />

This well-known diagnostic procedure is the reference<br />

standard in the diagnosis of Hirschsprung’s<br />

disease. The diagnostic role of rectal suction<br />

biopsy, first described by Noblett, 81 has improved<br />

with the availability of acetylcholinesterase staining.<br />

Complications of this procedure are bleeding<br />

and, especially in newborns, rectal perforation.<br />

Rectal perforation was described in 0.2% of 1340<br />

consecutive rectal suction biopsy procedures,<br />

including one case of buttock gangrene. 82 In our<br />

experience, we have observed one case of bleeding<br />

in a patient who received a blood transfusion, and


a case of rectal perforation in a patient with acute<br />

abdomen and pneumoperitoneum that needed<br />

laparotomy, surgical suture and drainage. If a deep<br />

suction biopsy specimen has to be taken below the<br />

REFERENCES<br />

1. Valerio D. Acute diabetic abdomen in childhood. Lancet<br />

1976; 10: 66–68.<br />

2. Way LW. Abdominal pain and the acute abdomen. In<br />

Sleisenger MH, Fortran JS, eds. Gastrointestinal Disease,<br />

2nd edn. Philadelphia: WB Saunders, 1978: 394–410.<br />

3. Glasgow RE, Mulvihill SJ. Abdominal pain, including<br />

the acute abdomen. In Feldman M, Scharschmidt BF,<br />

Sleisenger MH, eds. Gastrointestinal Disease, 6th edn.<br />

Philadelphia: WB Saunders, 1998: 80–89.<br />

4. Liebman WM, Thaler MM. Pediatric considerations of<br />

abdominal pain and the acute abdomen. In Sleisenger<br />

MH, Fortran JS. Gastrointestinal Disease, 2nd edn.<br />

Philadelphia: WB Saunders, 1978: 411–436.<br />

5. Babcock DS. Sonography of acute abdomen in the<br />

pediatric patient. J Ultrasound Med 2002; 21: 887–899.<br />

6. Rohrschneider WK, Troger J. Hydrostatic reduction of<br />

intussusception under US guidance. Pediatr Radiol<br />

1995; 25: 530–536.<br />

7. Carty HM. Paediatric emergences: non traumatic<br />

abdominal emergences. Eur Radiol 2002; 12: 2835–2848.<br />

8. Shimizu T, Suzuki R, Yamashiro Y et al. Magnetic<br />

resonance cholangiopancreatography in assessing the<br />

cause of acute pancreatitis in children. Pancreas 2001;<br />

22: 196–199.<br />

9. Hiroashi S, Hiroashi R, Uchida H et al. Pancreatitis:<br />

evaluation with MR cholangiopancreatography in<br />

children. Radiology 1997; 203: 411–415.<br />

10. Manfredi R, Lucidi V, Gui B et al. Idiopatic chronic<br />

pancreatitis in children: MR cholangiopancreatography<br />

after secretin administration. Radiology 2002; 224:<br />

675–682.<br />

11. Gonzalez RA, Lopez-Roses L, Lancho SA et al.<br />

Strangulated volvulus of the sigmoid colon with<br />

inviable mucosal appareance in a non-surgical patient.<br />

Endoscopic devolvulation. Rev Esp Enferm Dig 2002; 94:<br />

221–225.<br />

12. Pinedo G, Kirberg A. Percutaneous endoscopic<br />

sigmoidopexy in sigmoid volvulus with T-fasteners:<br />

report of two cases. Dis Colon Rectum 2001; 44:<br />

1867–1869.<br />

13. Hurlstone DP. Successful endoscopic haemoclipping in<br />

Mallory–Weiss syndrome with concurrent closure of<br />

esophageal perforation: further prospective evaluation<br />

of the technique is required. Scand J Gastroenterol 2002;<br />

37: 866.<br />

14. Cipolletta L, Bianco MA, Rotondano G. Endoscopic clipping<br />

of perforation following pneumatic dilatation of<br />

esophagojejunal anastomotic strictures. Endoscopy<br />

2000; 32: 720–722.<br />

15. Gill BD, Jenkins JR. Cost-effective evaluation and<br />

management of the acute abdomen. Surg Clin North Am<br />

1996; 76: 71–82.<br />

References 687<br />

peritoneal reflection, apart from the risk of perirectal<br />

septic cellulites, it poses no peritoneal problems.<br />

16. Wilkinson AH Jr. Acute abdomen. How to balance costs<br />

and quality care. Semin Pediatr Surg 1997; 6: 88–91.<br />

17. Bishop PR, Nowicki MJ, Parker PH Vomiting-induced<br />

hematemesis in children: Mallory–Weiss tear or<br />

prolapse gastropathy? J Pediatr Gastroenterol Nutr 2000;<br />

30: 436–441.<br />

18. Montes H, Dolfo W, Venezuela M et al. Coexistence of<br />

gastric mucosal prolapse and Mallory Weiss tear. J<br />

Gastroenterol Hepatol 2001; 16: 1172–1174.<br />

19. Panieri E, Bass DH. The management of ingested foreign<br />

bodies in children – a review of 663 cases. Eur J Emerg<br />

Med 1995; 2: 83–87.<br />

20. Litovitz T, Schmitz BF. Ingestion of cylindrical and<br />

button batteries: an analyisis of 2382 cases. Pediatrics<br />

1992; 89: 747–757.<br />

21. Burstein I, Steinberg R, Zer M. Small bowel obstruction<br />

and covered perforation in childhood caused by bizarre<br />

bezoars and foreign bodies. Isr Med Assoc J 2000; 2:<br />

129–131.<br />

22. Jiledar, Singh G, Mitra SK. Gastric perforation<br />

secondary to recurrent trichobezoar. Indian J Pediatr<br />

1996; 63: 689–691.<br />

23. Cauchi JA, Shawis RN. Multiple magnet ingestion and<br />

gastrointestinal morbidity. Arch Dis Child 2002; 87:<br />

539–540.<br />

24. Stricker T, Kellenberger CJ, Neuhaus TJ et al. Ingested<br />

pins causing parforation. Arch Dis Child 2001; 84:<br />

165–166.<br />

25. Fonkalsand EW, Loar N. Long term results after colectomy<br />

and endorectal pull-through procedure in<br />

children. Ann Surg 1992; 215: 57–62.<br />

26. Fazio VW. Toxic megacolon in ulcerative colitis and<br />

Crohn’s colitis. In Farmer RG, eds. Clinics in<br />

gastroenterology. Philadelphia: WB Saunders, 1980:<br />

389–407.<br />

27. Sheth SG, LaMont JT. Toxic megacolon. Lancet 1998;<br />

351: 509–12.<br />

28. Mourelle M, Casellas F, Guarner F et al. Induction of<br />

nitric oxide synthase in colonic smooth muscle from<br />

patients with toxic megacolon. Gastroenterology 1995;<br />

109: 1497–1502.<br />

29. Ferzoco SJ, Becker JM. Does aggressive medical therapy<br />

for acute ulcerative colitis result in a higher incidence<br />

of staged colectomy? Arch Surg 1994; 129: 420–423.<br />

30. Furst MB, Stromberg BV, Blatchford GJ et al. Colonic<br />

anastomoses: bursting strength after corticosteroid treatment.<br />

Dis Colon Rectum 1994; 37: 12–15.<br />

31. Berg DF, Bahadursingh AM, Kaminki DL et al. Acute<br />

surgical emergencies in inflammatory bowel disease.<br />

Am J Surg 2002; 184: 45–51.<br />

32. Nicholls RJ, Dozois RR. Surgery for ulcerative colitis,<br />

Crohn’s disease. In Nicholls RJ, Dozois RR, eds. Surgery


688<br />

Approach to the child with acute abdomen<br />

of the Colon and Rectum. New York: Churchill<br />

Livingstone, 1997: 593–644.<br />

33. Nordlinger B, Saint-Mark O. Free perforation. In<br />

Michelassi F, Milsom JW. Operative Strategies in<br />

Inflammatory Bowel Disease. New York: Springer-Verlag,<br />

1999: 369–373.<br />

34. Greenstein AJ, Mann D, Heimann T et al. Spontaneous<br />

free perforation and perforated abscess in 30 patients<br />

with Crohn’s disease. Ann Surg 1987; 205: 72–76.<br />

35. Tjandra JJ. Toxic colitis and perforation. In Michelassi F,<br />

Milsom JW, eds. Operative Strategies in Inflammatory<br />

Bowel Disease. New York: Springer-Verlag, 1999:<br />

234–245.<br />

36. Menguy R. Surgical management of free perforations of<br />

the small intestine complicating regional enteritis. Ann<br />

Surg 1972; 175: 178–189.<br />

37. Nichols RJ. Intraabdominal mass and/or abscess. In<br />

Michelassi F, Milsom JW, eds. Operative Strategies in<br />

Inflammatory Bowel Disease. New York: Springer-Verlag,<br />

1999: 360–368.<br />

38. Fazio V, Tiandra JJ. Strictureplasy for Crohn’s disease<br />

with multiple long strictures. Dis Colon Rectum 1993;<br />

36: 71–72.<br />

39. Michelassi F. Side-to side isoperistaltic strictureplasty<br />

for multiple Crohn’s strictures. Dis Colon Rectum 1996;<br />

39: 345–349.<br />

40. Dietz DW, Fazio VW, Laureti S et al. Strictureplasty in<br />

diffuse Crohn’s jejunoileitis: safe and durable. Dis Colon<br />

Rectum 2002; 45: 764–770.<br />

41. Oliva L, Wyllie R, Alexander F et al. The results of<br />

strictureplasty in pediatric patients with multifocal<br />

Crohn’s disease. J Pediatr Gastroenterol Nutr 1994; 18:<br />

306–310.<br />

42. Federici di Abriola G, De Angelis P, Dall’Oglio L, Di<br />

Lorenzo M. Strictureplasty: an alternative approach in<br />

long segment bowel stenosis in pediatric Crohn’s<br />

disease. J Pediatr Surg 2003; 38: 814–818.<br />

43. Prasil P, Laberge JM, Barkun A, Flageole H. Endoscopic<br />

retrograde cholangiopancreatography in children: a<br />

surgeon’s perspective. J Pediatr Surg 2001; 36: 733–735.<br />

44. Freeman ML, Nelson DB, Sherman S et al. Complication<br />

of biliary sphincterotomy. N Engl J Med 1996; 335:<br />

909–918.<br />

45. Fox VL, Werlin SL, Heyman MB et al. Endoscopic<br />

retrograde cholangiography in children. J Pediatr<br />

Gastroenterol Nutr 2000; 30: 335–342.<br />

46. Hasegawa T, Udatsu Y, Kamiyama M et al. Does a<br />

pancreatico-biliary maljunction play a role in spontaneous<br />

perforation of the bile duct in children? Pediatr<br />

Surg Int 2002; 18: 565–566.<br />

47. Kasat LS, Borwankar SS, Jain M, Naregal A.<br />

Spontaneous perforation of the extrahepatic bile duct in<br />

an infant. Pediatr Surg Int 2001; 17: 463–464.<br />

48. Ando K, Miyano T, Kohno S et al. Spontaneous<br />

perforation of choledochal cyst: a study of 13 cases. Eur<br />

J Pediatr Surg 1998; 8: 23–25.<br />

49. Shenoy VG, Jawale SA, Oak SN, Kulkarni BK.<br />

Anomalous pancreatobiliary union and chronic pancreatitis:<br />

rare presentation with biliary peritonitis. Pediatr<br />

Surg Int 2001; 17: 549–551.<br />

50. Tam PK, Saing H, Irving IM, Lister J. Acute pancreatitis<br />

in children. J Pediatr Surg 1985; 20: 58–60.<br />

51. Yachha SK, Chetri K, Saraswat VA et al. Management of<br />

childhood pancreatic disorders: a multidisciplinary<br />

approach. J Pediatr Gastroenterol Nutr 2003; 36:<br />

206–212.<br />

52. Zargar SA, Javid G, Khan BA et al. Endoscopic<br />

sphincterotomy in the management of bile duct stones<br />

in children. Am J Gastroenterol 2003; 98: 586–589.<br />

53. Nikou GC, Arnautis TP, Giamarellos-Bouboulis EJ et al.<br />

The significance of the dosage adjustment of octreotide<br />

in the treatment of acute pancreatitis of moderate<br />

severity. Hepatogastroenterology 2001; 48: 1754–1757.<br />

54. Uhl W, Buchler MW, Malfertheiner P et al. A<br />

randomized, double blind, multicentre trial of<br />

octreotide in moderate to severe acute pancreatitis. Gut<br />

1999; 45: 97–104.<br />

55. Paran H, Mayo A, Paran D et al. Octeotride treatment in<br />

patients with severe acute pancreatitis. Dig Dis Sci<br />

2000; 45: 2247–2251.<br />

56. DeBanto JR, Goday PS, Pedroso MR et al. Acute<br />

pancreatitis in children. Am J Gastroenterol 2002; 97:<br />

1726–1731.<br />

57. Jackson WD. Pancreatitis: etiology, diagnosis and<br />

management. Curr Opin Pediatr 2001; 13: 447–451.<br />

58. Onen A, Dokucu AI, Cigdem MK et al. Factors affecting<br />

morbidity in typhoid intestinal perforation in children.<br />

Pediatr Surg Int 2002; 18: 696–700.<br />

59. Mallick S, Klein JF. Management of typhoid perforation<br />

of the small bowel: a case series in Western French<br />

Guiana. Med Trop 2001; 61: 491–494.<br />

60. Kouame BD, Ouattara O, Dick RK et al. Diagnostic,<br />

therapeutic and prognostic aspects of intestinal typhoid<br />

perforations in children of Abidjan, Cote d’Ivoire. Bull<br />

Soc Pathol Exot 2001; 94: 379–382.<br />

61. Rahman GA, Abubakar AM, Johnson AW, Adeniran JO.<br />

Typhoid ileal perforation in Nigerian children: an<br />

analysis of 106 operative cases. Pediatr Surg Int 2001;<br />

17: 628–630.<br />

62. Shah AA, Wani KA, Wazir BS. The ideal treatment of<br />

the typhoid enteric perforation – resection anastomosis.<br />

Int Surg 1999; 84: 35–38.<br />

63. Chao HC, Chiu CH, Kong MS et al. Factors associated<br />

with intestinal perforation in children’s non-typhi<br />

Salmonella toxic megacolon. Pediatr Infect Dis J 2000;<br />

19: 1158–1162.<br />

64. Ciftci AO, Karnak I, Senocak ME et al. Spectrum of<br />

complicated intestinal amebiasis through resected<br />

specimens: incidence and outcome. J Pediatr Surg 1999;<br />

34: 1369–1373.<br />

65. Nagi B, Lal A, Kochhar R et al. Perforations and fistulae<br />

in gastrointestinal tuberculosis. Acta Radiol 2002; 43:<br />

501–506.<br />

66. Talwar S, Talwar R, Prasad P. Tuberculous perforations<br />

of the small intestine. Int J Clin Pract 1999; 53: 514–518.<br />

67. Ido K, Yuasa H, Ide M et al. Sonographic diagnosis of<br />

small intestinal anisakiasis. J Clin Ultrasound 1998; 26:<br />

125–130.<br />

68. Shirahama M, Konga T, Ishibashi H et al. Intestinal<br />

anisakiasis: US in diagnosis. Radiology 1992; 185:<br />

789–793.<br />

69. Herbrecht R, Letscher-Bru V, Bowden RA et al.<br />

Treatment of 21 cases of invasive mucormycosis with<br />

amphotericin B colloidal dispersion. Eur J Clin<br />

Microbiol Infect Dis 2001; 20: 460–466.<br />

70. Villani A, Vacca P, Onofri A, Cori M. Disseminated<br />

mucormycosis. A rare case in pediatric intensive care.<br />

Minerva Anesthesiol 1997; 63: 249–252.<br />

71. Legnani PE, Kornbluth A. Therapeutic options in the<br />

management of strictures in Crohn’s disease.<br />

Gastrointest Endosc Clin North Am 2002; 12: 589–603.<br />

72. Thomas-Gibson S, Brooker JC, Hayward CM, Shah SG.<br />

Colonoscopic balloon dilatation of Crohn’s strictures: a<br />

review of long-term outcomes. Eur J Gastroenterol<br />

Hepatol 2003; 15: 485–488.<br />

73. Segal D, Michaud L, Guimber D et al. Late-onset<br />

complications of percutaneous endoscopic gastrostomy


in children. J Pediatr Gastroenterol Nutr 2001; 33:<br />

495–500.<br />

74. Fox VL, Abel SD, Malas S et al. Complications following<br />

percutaneous endoscopic gastrostomy and subsequent<br />

catheter replacement in children and young adults.<br />

Gastrointest Endosc 1997; 45: 64–71.<br />

75. Pofahl WE, Ringold F. Management of early dislodgment<br />

of percutaneous endoscopic gastrostomy tubes. Surg<br />

Laparosc Endosc Percutan Tech 1999; 9: 253–256.<br />

76. Meng Boey CC, Goh KL, Sithasanan N, Goh DW. Jejunal<br />

perforation complicating PEG insertion in a child.<br />

Gastrointest Endosc 2002; 55: 607–608.<br />

77. Ledermann SE, Spitz L, Moloney J et al. Gastrostomy<br />

feeding in infants and children on peritoneal dialysis.<br />

Pediatr Nefrol 2002; 17: 246–250.<br />

78. Warngard O, Stenhammar L, Ascher H et al. Small<br />

bowel biopsy in Swedish paediatric clinics. Acta<br />

Paediatr 1996; 85: 240–241.<br />

References 689<br />

79. Guzman C, Bousvaros A, Buonuomo C, Nurko S.<br />

Intraduodenal hematoma complicating intestinal<br />

biopsy: case reports and review of the literature. Am J<br />

Gastroenterol 1998; 93: 2547–2550.<br />

80. Dall’Oglio L, De Angelis P, Federici di Abriola G et al. La<br />

colangiopancreatografia retrograda per via endoscopica<br />

in età pediatrica. In De Angelis GL, ed. L’endoscopia<br />

digestiva in età pediatrica e giovanile, 1st edn. Rome:<br />

Edizioni Medico Scientifiche Internazionali, 2002:<br />

95–109.<br />

81. Noblett HR. A rectal suction biopsy tube for use in the<br />

diagnosis of Hirschsprung’s disease. J Pediatr Surg 1969;<br />

4: 406–409.<br />

82. Teitelbaum DH, Coran AG, Weitzman JJ et al.<br />

Hirschsprung’s disease and related neuromuscular<br />

disorders of the intestine. In O’Neill JA, Rowe MI,<br />

Grosfeld JL et al. eds. Pediatric Surgery, 5th edn. St.<br />

Louis: Mosby-Year Book, 1998: 1381–1424.


41<br />

Introduction<br />

Management of ingested<br />

foreign bodies<br />

Yvan Vandenplas, Said Hachimi-Idrissi and Bruno Hauser<br />

The first report of a foreign body in a child dates<br />

from 1692 when Frederick the Great who, at the<br />

age of 4 swallowed a shoe buckle that passed<br />

without incident. 1 Accidental ingestion of a<br />

foreign body occurs frequently in children, and<br />

accounts for a significant number of emergency<br />

room visits. In general, accidental ingestion causes<br />

little or no morbidity. 2–4 In the majority of cases<br />

that come to medical attention, the ingestion was<br />

witnessed or is strongly suspected by the child’s<br />

caretaker. A significant number of accidental<br />

ingestions are likely to remain asymptomatic,<br />

since most objects pass without any incident.<br />

Sometimes the foreign object is discovered by accident<br />

in the stools 5 .<br />

Epidemiology<br />

Ingestion of foreign bodies is mainly a pediatric<br />

problem, since about 80% of cases occur in children<br />

between the ages of 6 and 36 months. 6–9 In<br />

our series of 325 children, the median age of the<br />

children was 2.8 years (Table 41.1). 6 According to<br />

reported series, the median age is less than 5 years.<br />

In adults, ingestion of foreign bodies may be intentional,<br />

as can be the case in adolescents.<br />

Intentional ingestion occurs most frequently in<br />

psychiatrically impaired individuals and prisoners.<br />

10 The largest series ingested by one individual<br />

was reported by Chalk and Foucour in 1928 and<br />

was an assortment of 2533 pins, needles and<br />

bobbins. 11 We had an adolescent girl who was a<br />

pencil-swallowing recidivist (unpublished).<br />

Amongst the miscellaneous foreign bodies<br />

observed in our series, one developmentally<br />

retarded child managed to ingest a large plastic bag<br />

(the type used in supermarkets); the plastic bag<br />

caused decreased food intake as single symptom. 6<br />

During recent years, a number of large pediatric<br />

series have been reported in the literature. The<br />

series of Weissberg included 70 patients (children<br />

and adults), 12 Kim et al reported 104 Korean pediatric<br />

cases, 13 and Olives report a series of 395 children.<br />

8 Athanassiadi et al reported a series of 400<br />

cases, 14 and Lam and et al reported as many as<br />

5240 ingested foreign bodies. 15 In the USA, about<br />

200000 accidental ingestions occur yearly in children<br />

and adolescents. 16 In 1997, there were more<br />

than 21000 emergency room visits by children in<br />

the USA for coin ingestions alone. 17 The incidence<br />

Table 41.1 Age distribution of 325 children<br />

with foreign body ingestion<br />

Age (years) No. of patients %<br />

0–1 36 11<br />

1–2 64 20<br />

2–3 78 24<br />

3–4 55 17<br />

4–5 27 8<br />

5–6 29 9<br />

6–7 12 4<br />

7–8 10 3<br />

8–10 6 2<br />

10–18 8 2<br />

Total 325 100<br />

Mean age 2.8 years (range 5 months to 18 years)<br />

Boys/girls, 58/42%<br />

691


692<br />

Management of ingested foreign bodies<br />

Table 41.2 Nature and number of foreign<br />

bodies ingested<br />

n %<br />

Coins 89 27<br />

Sharp objects (needles, pins, etc.) 51 16<br />

Batteries 43 13<br />

Toy parts 38 12<br />

Bones (fish, chicken) 38 12<br />

Large food bags 37 12<br />

Jewellery 19 6<br />

Miscellaneous 10 3<br />

Total 325 100<br />

and type of foreign body ingested depends on the<br />

geographic region (and reports depending on the<br />

specialty of the author). In our department, during<br />

a period of 15 years, 325 pediatric cases were seen<br />

because of ingestion of a foreign body or symptoms<br />

of acute esophageal obstruction. In general, coins<br />

are the most common foreign bodies ingested, and<br />

account for 27–70% of all cases 6,10,18-21 (Tables<br />

41.2 and 41.3, and Figure 41.1). History in determining<br />

coin denomination is unreliable. 22 In<br />

seaside regions, fish bones tend to be the most<br />

important. 23 The incidence of accidental ingestion<br />

of fish bones in China seems very high, with an<br />

incidence reaching 84%. 24 In the children of this<br />

series, fish bones accounted for 43% and coins for<br />

39%. 24 Other frequently ingested objects are sharp<br />

objects such as needles and pins, batteries, toy<br />

parts, chicken or fish bones. Each accounts for<br />

5–30%, depending on the series. 6,10,23 In adults,<br />

unintentional ingestions most frequently involve<br />

meat or fish bones. Gastric (tricho-) bezoars can be<br />

considered as a special group of foreign bodies,<br />

most of the time needing surgical removal. 25 In<br />

young infants, lactobezoars can cause gastric<br />

outlet obstruction. 26 Some candies contain a gel<br />

that may become compact in the intestinal lumen<br />

and cause obstruction. 27<br />

Table 41.3 Comparison between our data<br />

and the study reported by Olives 8<br />

In our series of 325 children, 12 children had<br />

ingested more than one foreign body. Ten children<br />

were ‘recidivists’, and were seen twice because of<br />

ingestion of a foreign body. 6 Overall, natural elimination<br />

occurred in 46% of the patients. 6<br />

Clinical signs and symptoms<br />

Location of the object<br />

Present study Olives 8<br />

Number of 325 395<br />

patients<br />

Age distribution 5 months 2 months<br />

to 18 years to 18 years<br />

Boy/girl ratio (%) 58/42 60/40<br />

Radio-opaque 60 64<br />

foreign body (%)<br />

Asymptomatic (%) 46 20<br />

Dysphagia, 24 59<br />

restrosternal pain (%)<br />

Coins most frequent most frequent<br />

(27%) (40%)<br />

Endoscopic 25 85<br />

removal (%)<br />

Success rate (%) 99 64<br />

Perforation (n) 0 0<br />

Most swallowed foreign bodies do not come to<br />

medical attention as they do not cause symptoms.<br />

Sometimes foreign bodies are discovered by coincidence,<br />

on a standard X-ray that was performed<br />

for other indications. 6 The majority of coin ingestions<br />

are asymptomatic. 6,23 The symptoms of<br />

obstruction caused by a foreign body depend on<br />

patient age, object size and its location. Symptoms<br />

in young children are non-specific and can include<br />

choking, drooling and poor feeding. There may be


Symptomatic<br />

endoscopy and<br />

• FB trapped in esophagus: push into stomach<br />

• attempt at removal<br />

(fasting if symptoms allow waiting time)<br />

Symptomatic<br />

endoscopy and<br />

• FB trapped in esophagus: push into stomach<br />

and in pharynx, esophagus: removal/push into<br />

stomach<br />

• attempt at removal<br />

(fasting if symptoms allow waiting time)<br />

Normal<br />

X-ray neck, chest (anterior–posterior, lateral)<br />

normal: wait and see<br />

FB: removal<br />

a time interval between ingestion and development<br />

of (respiratory) symptoms in infants. 10<br />

Children may alter their diet to consume liquids or<br />

a soft diet. Older children and teenagers with a<br />

foreign body in the esophagus typically complain<br />

of dysphagia, odynophagia and chest pain.<br />

Depending on the organization of medical care,<br />

approximately 5–10% of patients will have the<br />

foreign body located in the oropharynx, 20% in the<br />

esophagus, 60% in the stomach and 10% distal to<br />

the stomach. 28 Where fish bone ingestion is<br />

History of radio-opaque foreign body (FB)<br />

Fluoroscopy or X-ray neck, chest, stomach (abdomen)<br />

History of radiolucent swallowed foreign body<br />

Physical examination<br />

Clinical signs and symptoms 693<br />

Asymptomatic<br />

FB In pharynx, esophagus: removal/push into stomach<br />

FB in stomach: removal if sharp, battery, large (> 3–4 cm<br />

in length)<br />

(magnet probe vs. endoscopy)<br />

FB beyond stomach: wait and see if after 1–2 weeks, FB<br />

not observed in stools: control X-ray<br />

Asymptomatic<br />

X-ray with contrast (visualizes FB if trapped in<br />

esophagus, diagnostic possibly therapeutic)<br />

if FB remains in esophagus: endoscopy when fasting<br />

if FB not in esophagus: wait and see (observe stools) if<br />

after 1–2 weeks, FB not observed in stools: control<br />

X-ray<br />

History of choking spell (acute onset of severe onset cough, difficulty<br />

breathing, anxiety)<br />

Abnormal<br />

X-ray neck, chest (anterior–posterior, lateral)<br />

FB: removal<br />

Figure 41.1 Proposed recomendations for a child who has possibly swallowed a foreign body or presents the symptoms<br />

suggesting esophageal obstruction.<br />

common, a higher percentage of patients will have<br />

the foreign body located in the oropharynx. In<br />

general, foreign bodies in the oropharynx are<br />

symptomatic and cause dysphagia. 6–10 In our<br />

series, 22 of the 28 (79%) foreign bodies in the<br />

oropharynx were bones from fish or poultry. The<br />

rest were glass and there was one metal object. 6<br />

These foreign bodies were visible on physical<br />

examination of the oropharynx. 6 Additionally, it<br />

should be mentioned that there were 17 children<br />

who became symptomatic during the eating of fish<br />

or poultry, but with a negative inspection and no


694<br />

Management of ingested foreign bodies<br />

visualization of a bone on X-ray. Because symptoms<br />

persisted in these children, endoscopy was<br />

performed and a (small) laceration of the pharyngeal<br />

or esophageal mucosa could be observed in<br />

eight of them. 6<br />

In our experience, 54% of the children had transient<br />

symptoms immediately after the ingestion. 6<br />

In the patients in whom the ingestion was not<br />

witnessed (32 out of 176; 18%), the sudden onset<br />

of symptoms (such as acute and severe coughing,<br />

pain, etc.) and the circumstances of this sudden<br />

onset made the diagnosis of accidental ingestion<br />

suspected. Symptoms were in almost all cases pain<br />

or discomfort at the pharyngeal or retrosternal<br />

region, excessive saliva production, nausea and<br />

vomiting, acute coughing and difficulties in<br />

breathing. In 28 out of 325 patients (9%), the event<br />

was accompanied by more severe manifestations<br />

such as cyanosis or severe dysphagia. 6<br />

Approximately 60–70% of foreign bodies that<br />

become stuck in the esophagus are located in the<br />

proximal esophagus at the level of the upper<br />

esophageal sphincter. 10,28 Twenty per cent of the<br />

obstructions occur at the lower esophageal sphincter;<br />

the rest occur in the mid-esophagus at the level<br />

of the aortic notch. Patients with underlying<br />

esophageal disease are at greater risk for the<br />

foreign body being entrapped in the esophagus.<br />

Especially children with congenital esophageal<br />

atresia, tracheoesophageal fistula or fundoplication<br />

are at risk. Coins are relatively frequently<br />

trapped in the esophagus, although the reported<br />

incidence varies from 5 to 60%, mostly at the level<br />

of the upper or lower esophageal sphincter. 29,30<br />

The history of the child does not help to understand<br />

the pathophysiological reason of this trapping.<br />

Manometric measurements of the sphincters<br />

have not been performed in these patients.<br />

Respiratory symptoms such as wheezing, stridor<br />

and speech impairment may be the only symptoms<br />

of a foreign body in the esophagus. Respiratory<br />

symptoms can be the consequence of<br />

paraesophageal soft tissue swelling 10 . Isolated<br />

respiratory symptoms are more frequent in infants<br />

and toddlers.<br />

In our published series, the vast majority of the<br />

foreign bodies (196; 60%) were located in the<br />

stomach at the time of presentation. 6 Thirty-six<br />

(11%) foreign bodies were already located in the<br />

small intestine at first presentation. 6 All these,<br />

except three, were eliminated spontaneously.<br />

Three foreign bodies were very long sharp metal<br />

objects (screws of >5cm) located in the duodenum<br />

at presentation. In order to avoid complications,<br />

they were endoscopically removed under<br />

anesthesia. Gastric and intestinal foreign bodies<br />

may cause obstruction and perforation.<br />

Complications<br />

Yearly, approximately 1500 deaths occur in the<br />

USA secondary to foreign body ingestion 31 .<br />

Foreign bodies impacted in the oropharynx are<br />

usually sharp and cause symptoms, and may cause<br />

life-threatening complications such as retropharyngeal<br />

abscess and perforation. 23,32–35<br />

Perforation is one of the most dangerous complications,<br />

but occurs in less than 1% of cases. Sharp<br />

objects are associated with a much higher perforation<br />

rate than dull objects. 36 Pins can cause gastric<br />

perforation and migrate into the thorax. 36 The<br />

notion that ‘advancing points perforate and trailing<br />

points do not’ dates back to 1937, based on the<br />

evidence provided by analysis of 3266 accidental<br />

ingestions and inhalations. 37 A foreign body<br />

entrapped in the esophagus may initially be<br />

asymptomatic, but cause ulceration of the<br />

esophageal mucosa due to pressure necrosis in<br />

case of a dull object. Sharp objects may perforate,<br />

and cause aortic–esophageal fistula, mediastinitis,<br />

tracheo-esophageal fistula and bronchoesophageal<br />

fistula. 23 Esophageal diverticula or<br />

pseudodiverticula may develop secondary to pressure<br />

by a dull object.<br />

Death because of multiple perforations occurred<br />

within 2 months in an individual who had swallowed<br />

over 500 pins. 38 No child in our series or in<br />

Olives’ series presented with perforation. 6,8 In the<br />

series reported by Weissberg, 15 perforations<br />

occurred in 70 patients. 12 Differences and similarities<br />

between the reported series are likely to be<br />

related to the selection of the patients. 39<br />

Perforation can result in extraluminal migration of<br />

the foreign body. Perforations occur most<br />

frequently in the region of the ileocecal valve.<br />

Congenital malformations such as Meckel’s diverticulum<br />

or prior intestinal surgery are sites of


increased risk for foreign body entrapment and<br />

perforation. 28<br />

Foreign bodies only rarely cause jejunal, ileal or<br />

colic obstruction, although it has been reported to<br />

occur. 40 Obstruction of the appendix with a foreign<br />

body is extremely rare. 41<br />

Metallic foreign bodies such as coins that remain<br />

for several weeks in the stomach could cause toxicity,<br />

especially with zinc-predominant coins. 17<br />

Diagnosis and differential diagnosis<br />

Rapid diagnosis and treatment of foreign bodies<br />

trapped in the gastrointestinal tract will decrease<br />

morbidity and length of hospital stay. 42 A foreign<br />

body not discovered in all patients with symptoms<br />

suggests the presence of an object in the oropharynx.<br />

In the series of Olives, 11% of the swallowed<br />

foreign bodies were not found in the gastrointestinal<br />

tract. 8<br />

Oral fluids make coins move easily into the<br />

stomach. 30 Therefore, a 5-day observation at home<br />

is advised. 30 Whether X-ray is recommended in<br />

every child who is suspected of having swallowed<br />

a foreign body has been a topic of controversy for<br />

many years. 43 Between 60 and 90% of ingested<br />

foreign bodies are radio-opaque. 6,10,18 Although<br />

most foreign body ingestions remain asymptomatic,<br />

when ingestion is witnessed it is recommended<br />

to perform a roentgenogram because the<br />

object may be lodged in the esophagus and pose a<br />

risk to the patient even if asymptomatic. 28 An anterior–posterior<br />

X-ray demonstrates the location of a<br />

coin. A lateral radiograph may be helpful in case of<br />

ingestion of a sharp object such as a needle or a pin<br />

to exclude asymptomatic location in the airways.<br />

Since esophageal trapping of a foreign body may<br />

remain asymptomatic, and since chronic<br />

impaction may cause ulceration and necrosis of<br />

the mucosa, fluoroscopy or X-ray is advised.<br />

Unnecessary exposure to radiation is less harmful<br />

than the consequence of chronic impaction. A<br />

simple X-ray or fluoroscopy (smaller irradiation<br />

than X-ray) is much easier than the use of metal<br />

detectors. 44,45<br />

In 6% of our patients, the foreign body was radiolucent,<br />

with five cases with a fish or chicken bone<br />

Diagnosis and differential diagnosis 695<br />

that penetrated the esophageal mucosa and 16<br />

cases of impaction of a food bolus 6 . Three of these<br />

children were psychomotor retarded, and the<br />

other children had been operated upon previously<br />

because of an esophageal atresia and were known<br />

to have a residual esophageal stenosis. 6 Non-radioopaque<br />

foreign bodies represent a much more<br />

difficult diagnostic challenge than radio-opaque<br />

bodies. A negative radiographic analysis does not<br />

rule out the presence of a foreign body in the<br />

gastrointestinal tract. 42 Indeed, non-radio-opaque<br />

foreign bodies such as plastic bag clips seem to be<br />

relatively frequently swallowed, and may cause<br />

obstruction and perforation. 46 If a non-radioopaque<br />

foreign body is not observed in the stools<br />

after a period of 2 weeks, control investigations<br />

should be performed. We perform contrast X-ray to<br />

detect a gastric foreign body, because we consider<br />

this less invasive than endoscopy.<br />

Chronic unexplained respiratory disease necessitates<br />

the exclusion of a foreign body in the gastrointestinal<br />

tract or airways. This object can be radiolucent<br />

and therefore difficult to diagnose. In such<br />

a case, the majority of centers recommend diagnostic<br />

and immediate therapeutic bronchoscopy. 47<br />

Treatment options<br />

Over 80% of foreign body ingestions that come to<br />

medical attention are naturally eliminated spontaneously.<br />

Ten to twenty per cent require endoscopic<br />

removal, and less than 1% require surgery. 6<br />

Foreign bodies located in the oropharynx may be<br />

visible on physical examination, and if this is the<br />

case, the foreign body can be removed with the<br />

help of a Magill’s forceps or similar equipment. 48<br />

In our experience, the foreign body was impacted<br />

in 28 (9%) of the patients in the oropharynx on<br />

presentation, and could be extracted with a Magill<br />

forceps. 6<br />

Symptomatic patients who are unable to swallow<br />

their secretions and those with respiratory symptoms<br />

should undergo emergency endoscopy. 49 The<br />

first endoscopic extraction dates from 1972. 50<br />

Different forceps and baskets used for removal are<br />

shown in Figure 41.2. Sharp objects and batteries<br />

should be removed whenever possible. 21 Straight<br />

pins are the exception to the perforation risk,


696<br />

Management of ingested foreign bodies<br />

because the blunt-ended head passes first through<br />

the gastrointestinal tract. (However, sharp metal<br />

objects can also be removed safely with a magnet.)<br />

The incidence of endoscopic removal is as low as<br />

25% in our series, but is 77% and 85% in other<br />

series, 8,23 and with a success rate of 99% in our<br />

experience and that of Kim et al, 6,13 whereas<br />

Olives reported a failure rate of 36%. 8 The timing<br />

of the extraction of the body from the esophagus<br />

depends on the severity of the symptoms and the<br />

fasting state of the patient. 6 Sedation and anesthesia,<br />

or esophageal introduction of the endoscope or<br />

magnet probe, in half of our patients caused relaxation<br />

of the sphincter, resulting in a spontaneous<br />

passage of the foreign body into the stomach. If the<br />

patient was under anesthesia, the foreign body was<br />

removed. If the patient was only sedated,<br />

endoscopy was stopped and the management for<br />

gastric foreign bodies was applied (Figure 41.3). 6<br />

Intravenous administration of glucagons dilates<br />

the esophagus and has been successfully used in<br />

adults, but has not been effective in children. 51<br />

Figure 41.2 Alligator jaw forceps, tripod or pentapod<br />

forceps and baskets used for endoscopic removal.<br />

Figure 41.3 Foreign bodies that have been extracted from the gastrointestinal tract in our unit.


Esophageal extraction with a Foley catheter was<br />

already reported in 1966 by Bigler. 52 Smooth<br />

foreign bodies can be removed from the esophagus<br />

with a Foley catheter. 10 This technique has been<br />

reported to be 95% successful. 53 Fluoroscopically<br />

guided Foley catheter extraction of retained coins<br />

in pediatric patients who lack evidence of significant<br />

esophageal edema causing tracheal compromise<br />

is a safe and efficacious technique. 54<br />

However, the procedure has also been said to be<br />

potentially dangerous, because a foreign body can<br />

flip out with catheter removal, and cause immediate<br />

respiratory complications. 55<br />

If the foreign body is in the stomach at the moment<br />

of presentation, the recommended attitude is to<br />

‘wait and observe’, except for long sharp objects<br />

such as needles and pins (>3 to 4cm). 6 Gastric<br />

coins do not have to be extracted, since the majority<br />

of them will be eliminated spontaneously. 30<br />

However, extraction is recommended if, after<br />

several weeks, the object is still in the stomach. 6<br />

Also, administration of prokinetics has been<br />

reported in this indication, 7 although this is not<br />

recommended. Objects greater than 10cm in<br />

length in the stomach of an adult-size patient<br />

cannot pass the duodenum. 49 An ovoid object<br />

greater than 5cm in length or with a thickness<br />

greater than 2cm is unlikely to pass the pylorus. 56<br />

Objects of the size of toothbrushes 1 or pencils<br />

(own experience) should be removed from the<br />

stomach. Anecdotally, in one of our cases, when a<br />

child had swallowed a ring worth over $2000 in a<br />

jeweler’s shop, the ring was removed immediately.<br />

In our department, magnetic foreign bodies are<br />

removed with a magnet probe. 6 In the series we<br />

reported, extraction of batteries was attempted<br />

with a magnet probe, but failed in four cases out of<br />

36. 6 The largest series of battery ingestions<br />

reported was from a national survey registry<br />

reporting 2382 ingestions in 2320 patients. 57<br />

Button batteries from hearing aids are frequently<br />

ingested. Children imitate body piercing by using<br />

small powerful magnets across parts of their body<br />

including nose, ears, penis and tongue. 58 Some<br />

swallowed the magnets while attempting to use<br />

them, resulting in at least one near fatal complication.<br />

58 There have been 25 reported cases of batteries<br />

dislodged in the esophagus, causing develop-<br />

Treatment options 697<br />

ment of fistula, strictures and even death. 57<br />

Batteries impacted in the esophagus should be<br />

immediately removed. 49,59 Severe complications<br />

have occurred even with small batteries, and<br />

esophageal perforation was reported within 6h<br />

after ingestion. The caustic material in the battery,<br />

and perhaps discharge of current, may be responsible<br />

for the rapid development of severe complications.<br />

60 Batteries contain corrosive substances<br />

and may cause necrosis of the mucosa in case of<br />

leakage. 4,21,61 Ingestion of 12 small magnets<br />

caused perforation; the magnets were attracted to<br />

each other and caused pressure necrosis. 62 As a<br />

consequence, according to some authors, batteries<br />

cause a dilemma between a ‘wait-and-see’ attitude<br />

and an urgent laparoscopy. 59,63 Therefore, the<br />

development of a magnet probe is a useful tool,<br />

since it enables easy extraction in safe conditions.<br />

9,61,64,65 In our series, all gastric needles and<br />

pins could be removed with the magnet probe,<br />

except for two patients who had swallowed open<br />

safety pins. In these cases, endoscopic extraction<br />

under anesthesia was performed. 6<br />

Prevention is preferable to treatment. Modification<br />

of toy manufacturing practices and labeling<br />

standards may decrease the relative incidence<br />

of toy part ingestion. The US Consumer Product<br />

Safety Commission considers the toys within eggs<br />

to violate the small parts regulation of this<br />

Commission with respect to children under 3, and<br />

therefore these chocolate eggs are not allowed to<br />

be marketed in the USA, although no injuries<br />

involving these toys have been reported to date. 66<br />

Appropriate equipment and personnel for emergency<br />

resuscitation should be present when<br />

removing a foreign body without general anesthesia<br />

and intubation. 6<br />

Food impaction<br />

Impaction of food, usually meat, is the most<br />

common obstruction in adolescents and in<br />

toddlers after esophageal repair for congenital<br />

atresia. 67,68 Contrast radiography is not indicated<br />

because of the risk of aspiration. 49 If the symptoms<br />

are not too serious, tenderizers can be help. 67 Cola<br />

drinks in small amounts can also help in digesting<br />

impacted food (own experience).


698<br />

Management of ingested foreign bodies<br />

Cocaine ingestion<br />

This phenomenon occurs primarily in adolescents<br />

and young adults who act as a ‘mule’ and ingest<br />

cocaine-filled condoms as a method of smuggling.<br />

49 Absorption of 1–3g of cocaine following<br />

condom rupture is fatal, but one condom contains<br />

3–5g.<br />

Follow-up<br />

A telephone survey 2 weeks after ingestion of the<br />

foreign body did not reveal any unexpected<br />

complication. 6 In our experience, with a gastric<br />

foreign body that was not extracted, we advise the<br />

parents to observe the stools for 2 weeks. 6 If the<br />

foreign body was not observed in the stools, which<br />

was the case in as much as 62% of our patients, a<br />

control X-ray was performed. 6 In our series, in<br />

about two-thirds of the cases, the foreign body<br />

appeared to be eliminated spontaneously, despite<br />

being undetected by the parents. 6 This finding is<br />

in accordance with those of Macgregor and<br />

Ferguson stating that almost 50% of the foreign<br />

bodies were not recovered in the stools. 7 In about<br />

one-third, the foreign body was still present in the<br />

stomach. In that case, the foreign body was<br />

actively removed. 6 Kim et al 13 reported a spontaneous<br />

elimination of the foreign body in only 22%<br />

of cases, whereas we observed this phenomenon in<br />

almost half of our patients (46%). Amin et al<br />

reported a gastric passage of only 19% of ingested<br />

coins. 22 The transit time is variable and unpre-<br />

REFERENCES<br />

1. Kirk AD, Bowers BA, Moylan JA, Meyers WC.<br />

Toothbrush swallowing. Arch Surg 1988; 123: 382–384.<br />

2. Harris LS, Baker SP, Smith GA. Childhood asphyxiation<br />

by food: a national analysis and overview. J Am Med<br />

Assoc 1984; 251: 2231–2235.<br />

3. Litovitz T, Schmitz BF. Ingestion of cylindral and button<br />

batteries: an analysis of 2382 cases. Pediatrics 1992; 89:<br />

747–757.<br />

4. Vandenplas Y, de Pont S. Foreign bodies in the upper<br />

gastrointestinal tract. Acta Endosc 1994; 24: 363–370.<br />

dictable; the mean transit time in the series of children<br />

who presented at our emergency department<br />

was 3.8 days. 6,7,39<br />

Because gastric foreign bodies are frequently eliminated<br />

naturally, esophageal bougienage pushing a<br />

foreign body from the esophagus into the stomach<br />

has been recommended. 69 Whether esophageal<br />

extraction or passage into the stomach should be<br />

attempted depends largely on the local possibilities<br />

and on the condition of the patient, fasting or<br />

not. If, after a ‘reasonable long period’ such as 4–6<br />

weeks, the foreign body is still present in the<br />

stomach, endoscopic removal is proposed. 6<br />

Conclusions<br />

Only a minority of the accidental foreign body<br />

ingestions in children are witnessed by a<br />

bystander, and as a consequence it can be hypothesized<br />

that the majority of accidental ingestions<br />

remain asymptomatic. However, experience shows<br />

that esophageal trapping may be asymptomatic<br />

and that a foreign body can remain in the stomach<br />

for several weeks. Severe complications such as<br />

obstruction or perforation have been reported.<br />

Therefore, management should be balanced, not<br />

neglecting the small risk for severe morbidity but<br />

also avoiding over-investigation. In Figure 41.1, a<br />

practical attitude is proposed. These recommendations<br />

are limited to swallowed foreign bodies, and<br />

do not discuss foreign bodies in the respiratory<br />

tract.<br />

5. Bendig DW, Mackie GG. Management of smooth–blunt<br />

gastric foreign bodies in asymptomatic patients. Clin<br />

Pediatr 1990; 29: 642–645.<br />

6. Arana A, Hauser B, Hachimi-Idrissi S, Vandenplas Y.<br />

Management of ingested foreign bodies in childhood<br />

and review of the literature. Eur J Pediatr 2001; 160:<br />

458–472.<br />

7. Macgregor D, Ferguson J. Foreign body ingestion in children:<br />

an audit of transit time. J Accid Emerg Med 1998;<br />

15: 371–373.


8. Olives JP. Ingested foreign bodies. J Pediatr Gastroenterol<br />

Nutr 2000; 31 (Suppl 2): S188.<br />

9. Webb WA. Management of foreign bodies of the<br />

upper gastrointestinal tract. Gastroenterology 1988; 94:<br />

204–216.<br />

10. Macpherson RI, Hill JG, Othersen HB. Esophageal<br />

foreign bodies in children: diagnosis, treatment and<br />

complications. Am J Roentgenol 1996; 166: 919–924.<br />

11. Chalk SG, Foucour HO. Foreign bodies in the stomach:<br />

report of a case in which more than 2,500 foreign<br />

bodies were found. Arch Surg 1928; 16: 494–500.<br />

12. Weissberg D. Foreign bodies in the gastro-intestinal<br />

tract. S Afr J Surg 1991; 29: 150–153.<br />

13. Kim JK, Kim SS, Kil JI et al. Management of foreign<br />

bodies in the gastrointestinal tract: an analysis of 104<br />

cases in children. Endoscopy 1999; 31: 302–304.<br />

14. Athanassiadi K, Gerazounis M, Metaxas E, Kalantzi N.<br />

Management of esophageal foreign bodies: a retrospective<br />

review of 400 cases. Eur J Cardio-Thorac Surg 2002;<br />

21: 653–656.<br />

15. Lam HC, Woo JK, van Hasslet CA. Management of<br />

ingested foreign bodies: a retrospective review of 5240<br />

patients. J Laryngol Otol 2001; 115: 954–7.<br />

16. Litovitz TL, Keliin-Schwartz W, White S. 1999 Annual<br />

report of the American Association of Poison Control<br />

Centers Toxic Exposure Surveillance System. Am J<br />

Emerg Med 2000; 18: 517–574.<br />

17. Jefferson T. A thought for your pennies. J Am Med Assoc<br />

1999; 281: 122.<br />

18. Darrow DH, Holinger LD, Lemberg PS. Aerodigestive<br />

tract foreign bodies in the older child and adolescent.<br />

Ann Otol Rhinol Laryngol 1996; 105: 267–271.<br />

19. Campbell JB, Quattromani FL, Foley LC. Foley catheter<br />

removal of blunt esophageal foreign bodies. Experience<br />

with 100 consecutive children. Pediatr Radiol 1983; 13:<br />

116–118.<br />

20. Paul RI, Christoffel KK, Binns HJ, Jaffe DM. Foreign<br />

body ingestions in children: risk for complication varies<br />

with site on initial health care contact. Pediatric<br />

Practice Research Group. Pediatrics 1993; 91: 121–127.<br />

21. Webb WA. Management of foreign bodies of the upper<br />

gastrointestinal tract: update. Gastrointest Endosc 1995;<br />

41: 39–51.<br />

22. Amin MR, Buchinsky FJ, Gaughan JP, Szeremeta W.<br />

Predicting outcome in pediatric coin ingestion. Int J<br />

Pediatr Otorhinolaryngol 2001; 59: 201–206.<br />

23. Cheng W, Tam PK. Foreign body ingestion in children:<br />

experience with 1,265 cases. J Pediatr Surg 1999; 34:<br />

1472–1476.<br />

24. Nandi P, Ong GB. Foreign body in the oesophagus:<br />

review of 2,394 cases. Br J Surg 1978; 65: 5–9.<br />

25. De Backer A, van Nooten V, Vandenplas Y. Huge gastric<br />

trichobezoar in a 10-year old girl: case report with<br />

emphasis on endoscopy in diagnosis and therapy. J<br />

Pediatr Gastroenterol Nutr 1999; 28: 513–515.<br />

26. DuBose T, Southgate WM, Hill JG. Lactobezoars: a<br />

patient series and literature review. Clin Pediatr 2001;<br />

40: 603–606.<br />

27. Seidel JS, Gausche-Hill M. Lychee-flavored gel candies:<br />

a potentially lethal snack for infants and children. Arch<br />

Pediatr Adolesc Med 2002; 156: 1120–1122.<br />

28. Panieri E, Bass DH. The management of ingested foreign<br />

bodies in children – a review of 663 cases. Eur J Emerg<br />

Med 1995; 2: 83–87.<br />

29. Binder L, Anderson WA. Pediatric gastro-intestinal<br />

foreign body ingestions. Ann Emerg Med 1984; 13:<br />

112–117.<br />

30. Caravati EM, Bennett DL, McElwee NE. Pediatric coin<br />

ingestion. A prospective study on the utility of routine<br />

roentgenograms. Am J Dis Child 1989; 143: 549–551.<br />

References 699<br />

31. Schwartz GF, Polsky HS. Ingested foreign bodies of the<br />

gastrointestinal tract. Am Surg 1976; 42: 236–238.<br />

32. Bizakis JC, Segas J, Haralambas S. Retropharyngeal<br />

abscess associated with a swallowed bone. Am J<br />

Otolaryngol 1993; 14: 354–357.<br />

33. Gostout C, Bowyer B, Alhquist D. Mucosal malformation<br />

of the gastrointesinal tract. Clinical observations<br />

and results of the endoscopic neodymium-YAG laser<br />

therapy. Mayo Clin Proc 1988; 63: 894–899.<br />

34. Knight LC, Lesser TH. Fish bones in the throat. Arch<br />

Emerg Med 1989; 6: 13–16.<br />

35. Simic MA, Budakov BM. Fatal upper esophageal<br />

hemorrhage caused by a previously ingested chicken<br />

bone: case report. Am J Forensic Med Pathol 1998; 19:<br />

166–168.<br />

36. Stricker T, Kellenberger CJ, Neuhaus TJ et al . Ingested<br />

pins causing perforation. Arch Dis Child 2001; 84:<br />

165–166.<br />

37. Jackson C, Jackson CL. Disease of the Air and Food<br />

Passages of Foreign Body Origin. Philadelphia: WB<br />

Saunders, 1937.<br />

38. Henderson CT, Engel J, Schlesinger P. Foreign body<br />

ingestion: review and suggested guidelines for management.<br />

Endoscopy 1987; 19: 68–71.<br />

39. Hachimi-Idrissi S, Corne L, Vandenplas Y. Management<br />

of ingested foreign bodies in childhood: our experience<br />

and review of the literature. Eur J Emerg Med 1998; 5:<br />

461–463.<br />

40. Da Silva EJ, Golakai VK. Foreign body causing chronic<br />

subacute small bowel obstruction: an unusual case from<br />

Harare hospital. Centr Afr J Med 1998; 44: 16–18.<br />

41. Klinger PJ, Seeling MH, de Vault KR et al. Ingested<br />

foreign bodies within the appendix: a 100 year review<br />

of the literature. Dig Dis 1998; 16: 308–314.<br />

42. Messner AH. Pitfalls in the diagnosis of aerodigestive<br />

tract foreign bodies. Clin Pediatr 1998; 37: 359–365.<br />

43. Hodge D, Tecklenburg F, Fleisher G. Coin ingestion:<br />

does every child need a radiograph? Ann Emerg Med<br />

1985; 4: 443–446.<br />

44. Bassett KE, Schunk JE, Logan L. Localizing ingested<br />

coins with a metal detector. Am J Emerg Med 1999; 17:<br />

38–41.<br />

45. Seikel K, Primm PA, Elizondo BJ, Remley KL. Handheld<br />

metal detector localization of ingested metallic foreign<br />

bodies: accurate in any hands? Arch Pediatr Adolesc<br />

Med 1999; 153: 853–857.<br />

46. Newell KJ, Taylor B, Walton JC, Tweedie EJ. Plastic<br />

bread-bag clips in the gastrointestinal tract: report of 5<br />

cases and review of the literature. CMAJ 2000; 162:<br />

527–529.<br />

47. Dunn GR, Wardrop P, Lo S, Cowan DL. Management of<br />

suspected foreign body aspiration in children. Clin<br />

Otolaryngol 2002; 27: 384–387.<br />

48. Mahafza TM. Extracting coins from the upper end of the<br />

esophagus using a Magill forceps technique. Intern J<br />

Pediatr Otorhinolaryngol 2002; 62: 37–39.<br />

49. Wahbeh G, Wyllie R, Lay M. Foreign body ingestion in<br />

infants and children: location, location, location. Clin<br />

Pediatr 2002; 41: 633–640.<br />

50. Morrissey JF. Gastrointestinal endoscopy. Gastroenterology<br />

1972; 62: 1241–1268.<br />

51. Mehta D, Attia M, Quintana E, Cronan K. Glucagon use<br />

for esophageal coin dislodgment in children: a prospective,<br />

double-blind, placebo-controlled trial. Acad Emerg<br />

Med 2001; 8: 200–203.<br />

52. Bigler FC. The use of a Foley catheter for removal of<br />

blunt foreign bodies from the esophagus. J Thorac<br />

Cardiovasc Surg 1966; 51: 759–760.


700<br />

Management of ingested foreign bodies<br />

53. Campbell JB, Condon VR. Catheter removal of blunt<br />

esophageal foreign bodies in children. Survey of the<br />

Society for Pediatric Radiology. Pediatr Radiol 1989; 19:<br />

361–365.<br />

54. Harned RK 2nd, Strain JD, Hay TC, Douglas MR.<br />

Esophageal foreign bodies: safety and efficacy of Foley<br />

catheter extraction of coins. Am J Roentgenol 1997; 169:<br />

443–446.<br />

55. Myer CM III. Potential hazards of esophageal foreign<br />

body extraction. Pediatr Radiol 1991; 21: 97–98.<br />

56. Koch H. Operative endoscopy. Gastrointest Endosc 1977;<br />

24: 65–68.<br />

57. Litovitz T, Schimitz BF. Ingestion of cylindrical and<br />

button batteries: an analysis of 2,382 cases. Pediatrics<br />

1992; 89: 747–757.<br />

58. McCormick S, Brennan P, Yassa J, Shawis R. Children<br />

and mini-magnets: an almost fatal attraction. Emerg<br />

Med J 2002; 19: 71–73.<br />

59. Chan YL, Chang SS, Kao KL et al. Button battery ingestion:<br />

an analysis of 25 cases. Chang Gung Med J 2002;<br />

25: 169–174.<br />

60. McDermott VG, Taylor T, Wyatt JP. Orogastric magnet<br />

removal of ingested disc batteries. J Pediatr Surg 1995;<br />

30: 29–32.<br />

61. Volle E, Beyer P, Kaufmann HJ. Therapeutic approach to<br />

ingested button-type batteries. Magnetic removal of<br />

ingested button-type batteries. Pediatr Radiol 1989; 19:<br />

114–118.<br />

62. Cauchi JA, Shawis RN. Multiple magnet ingestion and<br />

gastrointestinal morbidity. Arch Dis Child 2002; 87:<br />

539–540.<br />

63. Namasivayam S. Button battery ingestion: a solution to<br />

management dilemma. Pediat Surg Int 1999; 15:<br />

383–384.<br />

64. Kuhns DW, Dire DJ. Button battery ingestion. Am Emerg<br />

Med 1989; 18: 293–300.<br />

65. McDermott VG, Taylor T, Wyatt JP. Oro-gastric magnet<br />

removal of ingested disc batteries. J Pediatr Surg 1995;<br />

30: 29–32.<br />

66. Kehrt R, Niggeman B, Klaue S, Wahn U. Small toys<br />

contained in chocolate eggs – good or bad surprise?<br />

Resp Med 2002; 96: 955–956.<br />

67. Ginsburg GG. Management of ingested foreign objects<br />

and food bolus impactions. Gastrointest Endosc 1995;<br />

41: 33–38.<br />

68. Mamel JJ, Weiss D, Pouagare M, Nord HJ. Endoscopic<br />

suction removal of food boluses from the upper<br />

gastrointestinal tract using Stiegmann–Goff friction-fit<br />

adaptor: an improved method for removal of food<br />

impactions. Gastrointest Endosc 1995; 41: 593–597.<br />

69. Calkins CM, Christianes KK, Sell LL. Cost analysis in<br />

the management of esophageal coins: endoscopy versus<br />

bougienage. J Pediatr Surg 1999; 34: 412–414.


42<br />

Introduction<br />

Medical aspects of intestinal<br />

transplantation<br />

Olivier Goulet<br />

Intestinal transplantation was first demonstrated<br />

to be technically feasible in humans in the early<br />

1960s. 1 The initial excitement, however, rapidly<br />

decreased when post-transplant rejection and<br />

severe sepsis resulted in high morbidity and<br />

mortality rates. 1 After extensive experimental<br />

reports, and despite very encouraging clinical<br />

results for heart and liver transplantation, the use<br />

of cyclosporin A did not enable, except in one<br />

child, 2 long-term survival after isolated smallbowel<br />

transplantation for short-bowel syndrome. 3<br />

In the early 1990s, two advances made intestinal<br />

transplantation a promising option for the treatment<br />

of end-stage intestinal failure: the combination<br />

with liver transplantation 4 and the development<br />

of FK506 (tacrolimus). 5 Recent advances in<br />

immunosuppressive treatment and the better<br />

monitoring and control of acute rejection have<br />

brought intestinal transplantation into the realm of<br />

standard treatment for intestinal failure. The<br />

results from the Intestinal Transplant Registry<br />

(www.intestinaltransplant.org) indicate that intestinal<br />

transplantation is currently an acceptable<br />

clinical modality for selected patients with irreversible<br />

intestinal failure. 6,7<br />

Irreversible intestinal failure<br />

Intestinal failure is a condition in which gastrointestinal<br />

function is insufficient to satisfy body<br />

nutrient and fluid requirements. Parenteral nutrition<br />

(PN) and home PN remain the mainstay of<br />

therapy for intestinal failure, whether it is partial<br />

or total, provisional or permanent. Since its introduction,<br />

PN has improved greatly, especially with<br />

the development of home PN. 8–12 Data from the<br />

European Registry indicate that the long-term<br />

survival of non-malignant patients on home PN is<br />

currently higher than the 1–3-year survival after<br />

isolated intestinal transplantation. 9 The North<br />

American Home Parenteral and Enteral Nutrition<br />

Registry reported 1- and 4-year survival rates of all<br />

children receiving PN at home as a result of shortbowel<br />

syndrome of 94 and 80%, respectively. 12<br />

The survival rates of patients with chronic intestinal<br />

pseudo-obstruction are inferior, being 87 and<br />

70%, respectively. Although long-term PN is associated<br />

with complications such as liver impairment,<br />

bone disease, vascular thrombosis and<br />

sepsis, innovative therapeutic modalities are<br />

required. Indeed, some patients develop complications<br />

while receiving standard therapy for intestinal<br />

failure and are considered for intestinal<br />

transplantation. Indications for intestinal transplantation<br />

involve mostly pediatric patients and<br />

may be divided into three main group: short-bowel<br />

syndrome, intestinal motility disorders and<br />

congenital diseases of the epithelial mucosa.<br />

Short-bowel syndrome<br />

Short-bowel syndrome (see also Chapter 29)<br />

caused by extensive resection of the small bowel<br />

has logically been the first indication for intestinal<br />

transplantation. After extensive resection of the<br />

small bowel, most neonates now survive and<br />

acquire gastrointestinal autonomy after a period<br />

depending on the extent and site of resection, the<br />

preservation of the ileocecal valve and the quality<br />

of the remaining gut. 13,14 Two groups are potential<br />

candidates for intestinal transplantation. Infants<br />

left with only a duodenum or at most 10cm of<br />

jejunum and no ileocecal valve will remain permanently<br />

dependent on PN. A small number of<br />

infants, approximately 10–15% in our experience,<br />

who have undergone massive intestinal resection<br />

701


702<br />

Medical aspects of intestinal transplantation<br />

in the neonatal period are at high risk of permanent<br />

intestinal failure when a stereotypical combination<br />

of features are present: small-bowel length<br />

of less than 30–40cm, absence of ileocecal valve,<br />

resection of some colon, minimal tolerance of<br />

enteral feeding within the first months after<br />

intestinal resection, dilated intestinal loop and<br />

bacterial overgrowth, and multiple surgical procedures.<br />

15 Older children or adolescents have a<br />

lesser degree of growth potential of the bowel<br />

compared with neonates who have extensive<br />

intestinal resection. Intestinal adaptation and<br />

intestinal absorption sufficient to meet growth<br />

requirements can only be obtained if the length of<br />

remaining small bowel is more than 50–60cm<br />

beyond the angle of Treitz. Some children do not<br />

become autonomous even after more than 10 years<br />

of home-based PN, and are thus candidates for<br />

intestinal transplantation. 16<br />

In patients with short-bowel syndrome on longterm<br />

PN, intestinal transplantation can only be<br />

envisaged once it has been formally shown that<br />

the remnant bowel cannot adapt. Surgical approaches<br />

such as lengthening of the small bowel, loop<br />

interposition or assembly of a ‘reverse’ intestinal<br />

loop should be attempted. 17–21<br />

Trophic factors such as recombinant human<br />

growth hormone (rhGH) might be helpful in some<br />

patients and might contribute to decreasing the<br />

need for intestinal transplantation in the near<br />

future. Studies have indicated that pituitary<br />

hormones modulate small-bowel growth in<br />

animals. 22–24 Clinical studies have provided<br />

controversial results. 25–30 Recent data in adults as<br />

well as in pediatric patients demonstrated that<br />

rhGH treatment sometimes allows improvement in<br />

intestinal function. 27–30 Glucagon-like peptide-2<br />

(GLP2) might, in the near future, be very helpful in<br />

enhancing gut mucosa trophicity and in reducing<br />

delay for achieving intestinal autonomy. 31<br />

Therefore, only a small number of patients with<br />

short-bowel syndrome are candidates for intestinal<br />

transplantation in the absence of life-threatening<br />

complications, especially progressive liver disease<br />

which raises another problem. The prevalence of<br />

complicated home PN-related liver disease<br />

increases with a longer duration of PN. This condition<br />

is one of the main causes of death in patients<br />

with permanent non-malignant intestinal failure. 32<br />

Some pediatric patients were reported to have<br />

received an isolated liver transplantation for endstage<br />

liver disease complicating short-bowel<br />

syndrome. 33–35 Some of them became intestinally<br />

autonomous after liver transplantation. However,<br />

it is important to stress that to be obliged to transplant<br />

the liver to achieve intestinal autonomy is<br />

clearly not the most logical or the easiest procedure.<br />

Thus, the primary aim must remain to<br />

prevent end-stage liver disease in short-bowel<br />

patients by using appropriate and long-term PN<br />

especially in the patients at high risk of liver<br />

disease.<br />

Intestinal motility disorders<br />

Motility disorders in childhood include total aganglionosis<br />

(extensive Hirschsprung’s disease) and<br />

chronic intestinal pseudo-obstruction syndrome.<br />

The first causes the same problems as short-bowel<br />

syndrome, with two main differences. The nonfunctioning<br />

colon is excluded and the ganglionic<br />

small bowel has motility disorders. Therefore,<br />

when normally innervated small bowel is shorter<br />

than 60cm the probability of long-term PN dependency<br />

is high. Logically, this situation requires a<br />

combined colon transplantation. 36,37 Chronic<br />

intestinal pseudo-obstruction syndrome (see<br />

Chapter 18) is a very heterogeneous condition with<br />

regard to clinical presentation, histopathological<br />

features, severity of motility disorders and<br />

outcome. 38–40 In our experience, 20–25% of<br />

patients will become dependent on long-term<br />

PN. 38 Recent data reported that intestinal transplantation<br />

or multivisceral transplantation have<br />

been performed in these patients, including some<br />

with associated urinary tract involvement. 41,42<br />

Munchäusen syndrome by proxy causing intestinal<br />

pseudo-obstruction must be recognized, even<br />

if difficult, and never justifies intestinal transplantation,<br />

as previously reported. 43<br />

Congenital enteropathies<br />

Two congenital intractable epithelial mucosal<br />

diseases are responsible for permanent intestinal<br />

failure and are currently recognized as requiring<br />

intestinal transplantation: microvillus inclusion<br />

disease and epithelial dysplasia (see Chapter 1).<br />

Both are autosomal recessive inherited disorders


with neonatal onset of severe watery diarrhea and<br />

total malabsorption. Microvillus inclusion disease<br />

involves the intracellular pathway of brush-border<br />

development, 44 whereas epithelial dysplasia is associated<br />

with abnormal enterocytes and basement<br />

membrane. 45 The primary inherited defect is not<br />

currently known for either of these diseases. Some<br />

children with one of these two mucosal diseases<br />

have undergone successful small-bowel transplantation<br />

in isolation or in combination with the liver. 46–52<br />

Clinical results after intestinal<br />

transplantation<br />

Immunosuppressive treatment<br />

The current immunosuppression we use is based on<br />

tacrolimus (Prograf ® ), steroids and monoclonal antibodies<br />

directed against interleukin-2 (IL-2) receptors<br />

(anti R-IL2 mAb). Tacrolimus is started intraoperatively<br />

to maintain whole blood levels using<br />

microparticle enzyme immunoassay technology,<br />

around 15–20ng/ml during the first month,<br />

10–15ng/ml during the following month and<br />

5–10ng/ml thereafter. Methylprednisolone is given<br />

as an initial bolus (20–25mg/kg over 6 hours), then<br />

2mg/kg per day during the first month and then<br />

progressively tapered to 0.5mg/kg every other day.<br />

Rapamycin (sirolimus) is now used in children, but<br />

follow-up is too short, even if encouraging results<br />

have been reported. 53 However, no randomized trial<br />

using this drug for intestinal transplant recipients is<br />

yet available. Azathioprine (Imuran ® ) is now rarely<br />

used and has mainly been replaced in some<br />

programs by mycophenolate mofetil. Caution is<br />

recommended with the use of mycophenolate<br />

mofetil because it has been shown to induce diarrhea.<br />

54 A new monoclonal antibody Campath ® was<br />

recently reported to improve survival after intestinal<br />

transplantation. 55<br />

Other procedures have been proposed for decreasing<br />

immunogenicity of the graft such as irradiation or<br />

for enhancing the natural microchimerism by infusing<br />

recipients with donor bone marrow during the<br />

perioperative period. 56 Combined bone marrow<br />

augmentation did not significantly improve graft<br />

survival 57 and was reported to favor the occurrence<br />

of graft-versus-host disease (GVHD) in animals. 58<br />

Clinical results after intestinal transplantation 703<br />

Survival after intestinal transplantation<br />

Current clinical results come from the data<br />

collected through the International Intestinal<br />

Transplant Registry (www.intestinaltransplant.<br />

org). To date, approximately 1000 intestinal transplantations<br />

have been performed throughout the<br />

world, mostly in the USA, Canada, France and the<br />

UK. The current available data from the registry<br />

include 989 transplantations in 923 patients from<br />

61 intestinal transplantation programs. Among<br />

recipients, 61% were children or adolescents. The<br />

transplants involved the isolated small-bowel<br />

transplantation with or without the colon (37%),<br />

the liver + small-bowel transplantation (50%) and<br />

multivisceral grafts (13%), including the stomach,<br />

pancreas, liver and small-bowel.<br />

The main indications in 563 children were shortbowel<br />

syndrome (63%), chronic intestinal pseudoobstruction<br />

syndrome (10%), severe intractable<br />

diarrhea (10%), total aganglionosis/Hirschsprung’s<br />

disease (7%), and in 360 adults: ischemia (23%),<br />

Crohn’s disease (14%), trauma (16%), desmoid<br />

tumor (9%) or tumors (8%). The main immunosuppression<br />

included tacrolimus (97%) and steroids in<br />

association with a variety of immunosuppressive<br />

agents including mycophenolate mofetil,<br />

rapamycin, azathioprine and IL-2 blockers.<br />

Overall 3-year patient survival is around 50–60%<br />

depending on several factors related to the type of<br />

transplanted organ (e.g. isolated intestine or combined<br />

with liver), the experience of the center, the<br />

immunosuppressive regimen including rapamycin<br />

or not, and the period from 1991 to now (Figure<br />

42.1). Post-transplant death was due mainly to<br />

infections (46%), multiorgan failure (2.5%) or<br />

lymphoma (10%). Full nutritional autonomy with<br />

complete discontinuation of PN has been achieved<br />

in 81% of survivors and partial recovery was documented<br />

in another 10% giving a total rehabilitation<br />

rate of 85% in survivors, 8% of the intestinal grafts<br />

being removed.<br />

It is clear from the intestinal transplantation<br />

registry and from individual programs that prognosis<br />

has improved during the past 10 years.<br />

Surgical procedures are well described and<br />

adapted to each organ combination. 59 Patient<br />

survival is associated with the type of organ transplanted,<br />

with better survival after small-bowel


704<br />

Medical aspects of intestinal transplantation<br />

% Survival<br />

1.0<br />

0.9<br />

0.8<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0.0<br />

0 1 2 3 4 5 6 7 8 9 10<br />

Figure 42.1 Actuarial graft survival according to the era of intestinal transplantation (data available from the International<br />

Intestinal Transplantation Registry, www.intestinaltransplant.org).<br />

transplantation. Nevertheless, these results must<br />

be interpreted with care because they represent<br />

the first 12 years’ experience of a large number of<br />

programs in children and adults, using different<br />

immunosuppressive regimens. The results from<br />

the largest of these centers reflect the current situation<br />

more closely. 60–65 In addition, programs that<br />

have performed at least ten transplants have better<br />

graft and patient survival rates compared with<br />

those that have performed less than ten. 7<br />

At the University Hospital Necker in Paris, 25<br />

isolated intestinal transplantations and 30<br />

combined intestine–liver transplantations were<br />

performed from November 1994 in 51 children<br />

(17 girls) ranging in age between 2.5 and 15 years<br />

(median 5 years). Associated right colon transplantation<br />

was performed 37 times (21<br />

intestine–liver transplants). All patients were on<br />

long-term PN for a median duration of 4.5 years<br />

(range 18 months–13 years) for congenital<br />

≥ 1998<br />

Years post-transplantation<br />

1992–1997<br />

≤ 1991<br />

enteropathy (n=18), short-bowel syndrome<br />

(n=14), extensive Hirschsprung’s disease (n=13)<br />

and intestinal pseudo-obstruction (n=6).<br />

Immunosuppression included tacrolimus, methylprednisolone,<br />

azathioprine and IL-2 blockers. With<br />

a follow-up ranging between 6 months and 8.5<br />

years, 36 patients are alive (patient survival<br />

70.5%; graft survival 65%). The following factors<br />

are significantly related to poor outcome (death or<br />

graft loss) age >7 years (p


Liver-induced immune tolerance<br />

It is currently difficult to analyze the difference in<br />

intestinal graft survival rates between isolated and<br />

combined liver–intestine transplant. In general, the<br />

clinical status of liver–small bowel recipients is<br />

poor at the time of transplantation and contributes<br />

to the high post-transplant rate of morbidity and<br />

mortality. This is suggested by the 1- and 2-year<br />

survival of patients, who have not undergone<br />

transplantation, being 30 and 22%, respectively,<br />

and the number of deaths on the waiting list. 66 On<br />

the other hand, the isolated small-bowel graft not<br />

only has the highest incidence of rejection, but also<br />

requires more intense immunosuppression to<br />

control it. It appears from our experience that the<br />

most encouraging results have been with combined<br />

small bowel–liver transplantation. Intestinal graft<br />

rejection was less frequent and less severe in small<br />

bowel–liver transplant recipients. Simultaneous<br />

liver grafting might reduce the risk of intestinal<br />

rejection as supported by experimental data 67,68<br />

and clinical results. 60,69<br />

The mechanism by which the liver induces tolerance<br />

is still under debate. The deletion of donor<br />

alloreactive T cells was initially suggested but<br />

remains controversial. 70 Other mechanisms such as<br />

anergy or the generation of regulatory cells might<br />

contribute to liver-induced tolerance, as suggested<br />

by the decreased expression of CD25 or CD8 T cells<br />

infiltrating tolerated intestinal graft and the reversal<br />

of tolerance by IL-2. 71 Currently, the role of<br />

liver-derived dendritic cells in inducing tolerance<br />

is stressed. 72 A better insight into the mechanisms<br />

involved in liver-induced tolerance is important in<br />

the design of immunosuppressive regimens for<br />

small bowel–liver transplant recipients, because<br />

there is experimental evidence that immunosuppressive<br />

drugs may inhibit the tolerogenic effect of<br />

the liver. 73,74<br />

Complications after intestinal<br />

transplantation<br />

The singularities of the gut may impede intestinal<br />

transplantation for several reasons:<br />

(1) The gut-associated lymphoid tissue can<br />

induce GVHD and may enhance allograft<br />

Complications after intestinal transplantation 705<br />

rejection and the subsequent risk of Gramnegative<br />

sepsis;<br />

(2) The secretion of lymphokines or the production<br />

of cytotoxic T cells in response to intraluminal<br />

pathogens may impede the induction of<br />

tolerance and thereby favor allograft rejection;<br />

(3) The need for heavy immunosuppressive<br />

treatment increases the risk of developing<br />

opportunistic infection and post-transplant<br />

lymphoproliferative disorders (PTLD).<br />

Graft-versus-host-disease<br />

GVHD has been extensively studied in animal<br />

models of intestinal transplantation. 1 In humans,<br />

despite the presence of circulating donor-derived<br />

lymphocytes during the first few weeks after transplantation,<br />

clinical signs of GVHD have rarely<br />

been reported. 75 GVHD is not therefore a major<br />

complication after intestinal transplantation, unlike<br />

graft rejection.<br />

Intestinal rejection<br />

Intestinal allograft rejection remains the major<br />

complication after intestinal transplantation. As a<br />

result of increased immunosuppressive treatment,<br />

graft rejection may further precipitate opportunistic<br />

infections that become additive factors in<br />

patient and graft losses. As rejection can occur<br />

rapidly and can be life-threatening, close monitoring<br />

is required. This has led to the development of<br />

numerous diagnostic methods, which have not<br />

been validated in human intestinal transplantation<br />

or have limited value. 76–78 Therefore, regular biopsies<br />

of the proximal and distal ends of the graft for<br />

histological or immunohistochemical analysis are<br />

required 79–83 (Figure 42.2). Clinical signs of rejection<br />

occur later than histological and immunohistochemical<br />

signs and correspond to a relatively<br />

advanced rejection process with marked histological<br />

lesions. Rejection and sepsis can be intimately<br />

related after small-bowel transplantation when<br />

rejection compromises normal intestinal barrier<br />

mechanisms and bacterial translocation results<br />

with consequent multiorgan failure. Graft rejection<br />

may be reversed by using methylprednisolone<br />

pulses. Severe exfoliative rejection leaves the<br />

intestine totally denuded of its mucosa and does<br />

not respond to methylprednisolone or to anti-


706<br />

Medical aspects of intestinal transplantation<br />

(a) (b)<br />

(c)<br />

lymphoglobulins or anti-CD3 mAb. Recovery may<br />

sometimes occur after exfoliative rejection but<br />

with a very high cost related to severe protracted<br />

protein-losing enteropathy during which albumin<br />

and hemoglobin replacement is needed and<br />

malnutrition may occur. In case of unresponsive<br />

exfoliative graft rejection, subepithelial fibrosis<br />

may appear, leading to severe hypomotility and<br />

subsequent intraluminal bacterial overgrowth. 81–83<br />

In our experience, we do consider that a rejection<br />

unresponsive to methylprednisolone pulses is a<br />

matter of discussing intestinal graft removal<br />

instead of dramatic increase of immunosuppression<br />

with the risk of infection and PTLD. This<br />

decision is difficult and is based on the specific<br />

experience of each program with the mandatory<br />

input of the pathologist.<br />

Infectious complications<br />

Clinical manifestations of allograft rejection are<br />

non-specific. It is obviously very important to<br />

Figure 42.2 (a) Mild intestinal graft rejection showing<br />

only pictures of apoptosis in the gland (HES x1200); (b)<br />

severe intestinal graft rejection showing epithelial blunting<br />

and disappearance of glands (HES x200); (c) severe<br />

‘exfoliative’ intestinal graft rejection with total villous<br />

atrophy, epithelium disappearance, absence of glands and<br />

T-cell infiltration (HES x800).<br />

differentiate other sources of potential intestinal<br />

allograft disease that may clinically mimic rejection,<br />

such as cytomegalovirus (CMV), adenovirus,<br />

Epstein–Barr virus (EBV) or other bacterial/viral<br />

enteritis. Viral infections are frequent, such as<br />

CMV primo-infection or reactivation, which is not<br />

always prevented by the use of pre-emptive treatment<br />

(ganciclovir). The diagnosis of CMV infection<br />

improved with the use of the polymerase<br />

chain reaction (PCR), which has been shown to be<br />

a sensitive method for the early detection of CMV<br />

infection in solid-organ and intestinal graft recipients.<br />

84 The incidence of CMV infection has been<br />

reported to be as high as 29% in pediatric recipients<br />

of intestinal grafts. 84 It is recommended to<br />

avoid using a seropositive graft in a seronegative<br />

recipient. Nevertheless, patients who are awaiting<br />

composite grafts are frequently too sick to await a<br />

CMV-seronegative donor. CMV prophylaxis is now<br />

well established with the wide use of ganciclovir. 85<br />

EBV infection in association with immunosuppressive<br />

drugs used for solid-organ transplantation<br />

can produce a spectrum of illnesses. Donor selection<br />

and the prevention of EBV infection remain<br />

unsolved problems. Indeed, a high incidence of<br />

EBV-induced PTLD has been reported. 86 The inci-


dence increases with the degree of immunosuppression.<br />

Because of the high rates of morbidity<br />

and mortality associated with EBV disease, a preemptive<br />

therapy based on serial monitoring of the<br />

EBV viral load is required. Quantitative EBV–PCR<br />

in the peripheral blood has recently enabled early<br />

diagnosis as well as the follow-up of patients with<br />

EBV infection. 87 This may allow the diagnosis of<br />

EBV infection before the development of PTLD.<br />

In the case of established EBV-related PTLD,<br />

therapeutic changes based on quantitative PCR are<br />

helpful in preventing the end phases of this<br />

disease. 84 In addition, in the case of documented<br />

PTLD, the use of anti-CD20 monoclonal antibodies<br />

has proved to be efficient in reversing the<br />

disease. 88<br />

Other severe life-threatening complications have<br />

been reported, such as diffuse adenovirus enterocolitis<br />

or hemophagocytosis. 89–91 The early identification<br />

of viral infections, based on the repeated<br />

use of appropriate methods, may help in avoiding<br />

the misdiagnosis of rejection leading to an unnecessary<br />

increase of immunosuppressive treatment<br />

with consequent exacerbation of the underlying<br />

infectious condition.<br />

Intestinal graft function<br />

Provided that the small intestine survives ischemia<br />

and reperfusion injury, long-term graft function is<br />

dependent on the effects of denervation, lymphatic<br />

disruption, immunosuppressive treatment, rejection<br />

and infection. These factors may explain<br />

impaired function of the intestinal graft and a<br />

delay in achieving intestinal autonomy.<br />

Intestinal motor function after intestinal<br />

transplantation<br />

The experimental study of small-bowel allografts<br />

provides a unique opportunity to develop insights<br />

into the impact of manipulation, preservation,<br />

ischemia/reperfusion injury, extrinsic denervation<br />

and immunological injury on intestinal function.<br />

However, very few data exist on these injuries to<br />

transplanted human small intestine. Ischemiareperfusion<br />

injury or intestinal manipulation<br />

evokes an inflammatory response within the<br />

intestinal muscularis that is associated with<br />

Intestinal graft function 707<br />

intestinal dysmotility. It was shown from human<br />

intestinal graft specimens obtained during transplantion<br />

that manipulation during organ harvesting<br />

initiates a functionally relevant molecular and<br />

cellular inflammatory response within the graft<br />

muscularis that is potentiated during the reperfusion<br />

period. 92<br />

The loss of extrinsic innervation of the small<br />

bowel severely impairs intestinal motor function.<br />

It has been shown in animal models that extrinsic<br />

denervation with maintenance of intestinal continuity<br />

only alters the regularity of the interdigestive<br />

myoelectric complex. 93 However, autotransplantation<br />

causes complete disruption of the orderly,<br />

sequential migration of the interdigestive myoelectric<br />

complex from the innervated to the denervated<br />

intestine, and food intake no longer inhibits the<br />

myoelectric complex. 94 Studies have observed the<br />

changes of the transplanted intestine itself in order<br />

to evaluate the effects of intestinal transplantation<br />

on the intrinsic nervous system. Taguchi et al<br />

found that the contractile properties of smallbowel<br />

smooth muscle and its sensitivity to drugs<br />

were unaltered by transplantation. 95 Although the<br />

excitatory response was comparable with that of<br />

normal intestine, the inhibitory response was<br />

modified by the loss of the extrinsic adrenergic<br />

inhibitory innervation. They also studied the<br />

temporary additional loss of intrinsic inhibition<br />

caused by prolonged graft storage, which recovers<br />

within 7–8 days after transplantation. 96 An<br />

increase was also shown of the intestinal contractile<br />

motility after intestinal transplantation, associated<br />

with a marked increase of non-adrenergic,<br />

non-cholinergic (NANC) neural components, with<br />

the dominant intrinsic neural component changing<br />

from cholinergic to NANC over 4 weeks. 97<br />

Autotransplantation as well as isotransplantation<br />

in dogs or rats has allowed the study of the reinnervation<br />

process after small-bowel transplantation.<br />

98,99 Reinnervation of the graft intestinal wall<br />

does occur but requires a prolonged period. The<br />

major route of reinnervation is along the arterial<br />

axis of the intestinal graft, not beyond the enteric<br />

anastomosis. 100 On the other hand, Taguchi et al<br />

studied immunohistochemically the distribution<br />

of neurons in a syngeneic model of transplantation<br />

in Lewis rats. Both the nitric oxide (NO) and<br />

peptidergic neurons markedly decreased just after<br />

transplantation, and the NO neurons recovered<br />

faster than peptidergic neurons suggesting an


708<br />

Medical aspects of intestinal transplantation<br />

important role in the adaptation process in the<br />

early period after transplantation. 101 After transplantation<br />

of the entire jejuno-ileum, isotransplanted<br />

rat intestine has qualitatively normal<br />

myoelectrical activity but decreased absorptive<br />

capacity. 102–104 If rejection is controlled and<br />

ischemic time is kept to a minimum, allografts of<br />

small intestine will exhibit a normal propagated<br />

motor myoelectric complex. In humans, the intrinsic<br />

motor activity of the small intestine is<br />

preserved for the most part, although there is some<br />

discoordination between proximal and distal<br />

elements in the fed state. 105,106<br />

Acute allograft rejection provokes intestinal paralysis<br />

before mucosal destruction is established. 107<br />

In animals, chronic rejection causes thickening of<br />

muscularis propria by both hyperplasia and hypertrophy<br />

accompanied by inflammatory cell infiltrate.<br />

108 Chronic allograft rejection severely<br />

impairs the enteric nerve with subsequent alteration<br />

of myoelectrical activity. 108–110 Functional<br />

impairment partially regresses after FK506<br />

rescue. 109 In clinical practice, chronic rejection is<br />

very rare but its expression is dominated by severe<br />

intestinal obstruction.<br />

Very few data regarding intestinal motility are<br />

available in humans. 106 Most knowledge comes<br />

from the observations of recovery of intestinal<br />

transit and contrast examination of the transplanted<br />

intestine. 111<br />

Absorptive function after intestinal<br />

transplantation<br />

Studies performed in animals have shown that<br />

intestinal tranates, lipids, glutamine, water and<br />

electrolytes, but there is no evidence in<br />

humans. 112–120 Feeding must resume as early as<br />

possible after transplantation because this ensures<br />

optimal mucosasplantation disturbs the absorption<br />

of carbohydrl trophicity and reduces gastrointestinal<br />

stasis, which causes intraluminal bacterial<br />

overgrowth. Clinical experience has demonstrated<br />

that because of water–electrolyte malabsorption,<br />

abnormal motility and impaired lymphatic<br />

drainage it may take several weeks to achieve<br />

normal intestinal transit and reduced stool<br />

volume. If the recipient has no colon, associated<br />

colon transplantation has physiological advan-<br />

tages in terms of water and electrolyte re-absorption,<br />

slowing intestinal transit and trophic factors,<br />

through colonic synthesis of short-chain fatty<br />

acids. Finally, it is currently considered that<br />

intestinal transplantation restores an enteral axis<br />

capable of ensuring digestion and absorption.<br />

Intestinal function sufficient to withdraw PN<br />

completely may be achieved with adequate nutritional<br />

management. In our experience, colon grafting<br />

does not impede survival after intestinal transplantation<br />

and improves the patient’s condition<br />

with early PN weaning. The colon water–electrolyte<br />

absorption capacity reduces the need for compensation<br />

by the parenteral route.<br />

Post-transplant practice and<br />

procedures<br />

Initial post-transplant period<br />

Total PN is administered continuously, with modifications<br />

made to the electrolyte and macronutrient<br />

content of the solution based upon serum and<br />

urine laboratory parameters. Hyperglycemia may<br />

result from initial high steroid doses and steroid<br />

recycles during early rejection episodes and/or<br />

from tacrolimus. Post-transplant pancreatitis may<br />

also affect glucose metabolism. Thus, insulin may<br />

be required in patients with severe hyperglycemia<br />

to maintain adequate caloric balance. Impaired<br />

renal function may require the limitation of nitrogen<br />

intake required for post-transplant wound<br />

healing and fluid restrictions may prohibit the<br />

ability to provide enough total PN volume to<br />

supply adequate calories. Use of intravenous lipid<br />

emulsion whenever possible according to metabolic<br />

tolerance and infectious state is recommended<br />

to achieve adequate calorie intake and<br />

essential fatty acid provision. The type of emulsion<br />

has never been evaluated in such situations<br />

and might be controversial. In our practice,<br />

we preferentially use medium-chain/long-chain<br />

triglyceride (MCT/LCT) emulsion without overpassing<br />

1.5g/kg per day.<br />

Initiation of enteral and oral feeding<br />

Continuous enteral feeding through a gastrostomy<br />

or, for others, from proximal jejunostomy is started


once intestinal motility occurs, generally within 1<br />

week. Enteral feeding is initiated and advanced<br />

slowly unless contraindicated, as in the presence<br />

of severe diarrhea or vomiting and, of course, in<br />

the presence of an acute graft rejection. In our<br />

experience and elsewhere, the patients initially<br />

receive a low-antigenic and low-fat formula and<br />

are subsequently transferred over a period of<br />

weeks and months to a more complex product<br />

with intact macronutrients. The use of an amino<br />

acid- versus a peptide-based enteral product<br />

remains a controversial issue in the non-transplant<br />

critically ill patient with impaired gastrointestinal<br />

function. 121 However, in the presence of carbohydrate<br />

and fat, nitrogen absorption has been<br />

reported to be greater with a 100% elemental free<br />

amino acid-protein source. 122 Moreover, the presence<br />

of free glutamine in powdered amino acidbased<br />

formulas has been shown to be beneficial in<br />

protecting gastrointestinal mucosa and reducing<br />

bacterial translocation. 123 Conversely, peptidebased<br />

elemental formulas are generally lower in<br />

osmolality which may make them more suitable<br />

for use in the presence of increased stool output.<br />

Finally, it is impossible to identify significant<br />

differences in outcome as measured by the length<br />

of time until the patient is completely weaned<br />

from total PN, length of hospital stay, ileostomy<br />

output or time until transition to oral intake alone,<br />

by the type of enteral formula used, peptide or<br />

hydrolyzed casein-based enteral formula versus an<br />

amino acid preparation. Post-transplant-impaired<br />

lymphatic drainage with either chylous ascites or<br />

intestinal lymphangiectasia, limits lipid intake.<br />

Low-fat formulas and MCT-rich formulas are<br />

recommended at initiation of enteral feeding. In<br />

our experience, children with inferior vena cava<br />

thrombosis are at high risk of chylous ascites and<br />

must be fed very slowly with MCT-rich formulas.<br />

More complex products with intact macronutrients<br />

are progressively introduced according to<br />

graft status, digestive tolerance and capacity of<br />

eating.<br />

Parenteral nutrition weaning<br />

PN weaning is a crucial objective, allowing the<br />

intestinal transplantation to be considered<br />

successful. Full nutritional autonomy with<br />

complete discontinuation of PN has been achieved<br />

in 81% of cases and partial recovery was docu-<br />

Post-transplant practice and procedures 709<br />

mented in another 11%, for a total rehabilitation<br />

rate of 92% in survivors. In the absence of major<br />

complications, recipients of an isolated intestinal<br />

allograft are weaned from PN relatively quickly, the<br />

median time ranging between 4 and 6 weeks 60,65<br />

Patients are weaned from PN as enteral feeding is<br />

progressively increased according to the tolerance<br />

of the child. It can take several weeks or months<br />

depending on the pre-transplant nutritional status,<br />

post-transplant complication rate, and intestinal<br />

graft recovery and function. In our experience,<br />

among 31 survivors after isolated (n=11) or<br />

combined intestine–liver transplantation (n=20),<br />

all patients were weaned from PN after a delay<br />

ranging from 4 weeks to 3 years. In the absence of<br />

major complications, recipients of an isolated<br />

intestinal allograft can be weaned from PN relatively<br />

quickly within 4–6 weeks. The longer time to<br />

achieve full enteral nutrition after small bowel–liver<br />

transplantation reflects a more prolonged recovery<br />

from the surgical procedure compared with isolated<br />

small-bowel transplantation.<br />

Eating disorders<br />

Chronically ill infants and children are at risk for<br />

oral aversion caused by the loss of the sucking or<br />

swallowing reflex in those maintained on total PN<br />

or tube feeding for an extended period of time<br />

without oral intake. Further oral-associated problems,<br />

including delayed speech and language<br />

development, may also result from this aversion.<br />

Pre-transplant management of eating disorders and<br />

early and adequate post-transplant stimulation of<br />

oral feeding can reduce food aversion. It is essential<br />

to insert a digestive access (gastrostomy or<br />

proximal jejunostomy) at the time of transplant<br />

surgery, especially in patients felt to have eating<br />

disorders. Full enteral feeding can be progressively<br />

achieved by using a polymeric diet. Psychological<br />

evaluation and assistance are of course required in<br />

each case. In our experience, all patients achieved<br />

oral feeding, sometimes up to 2 years after<br />

transplantation.<br />

Monitoring of intestinal function<br />

The D-xylose absorption test provides basic information<br />

regarding mucosal integrity, but is influenced<br />

by a number of factors, such as impaired


710<br />

Medical aspects of intestinal transplantation<br />

gastric emptying or accelerated intestinal transit.<br />

This test is never used in our clinical practice.<br />

Mucosal enzyme activities, especially disaccharidase<br />

activity, can be assayed from a biopsy specimen<br />

obtained from endoscopy. Enzyme levels<br />

correlate with mucosal injury due to allograft<br />

rejection or viral infection. 124 Thus, the relevance<br />

of this assessment for monitoring of intestinal<br />

function in a stable patient is a matter of debate.<br />

Recipients with an ileostomy, during the 2–3<br />

months following transplant procedure, or continent<br />

older patients, can benefit from stool analysis.<br />

We used to perform a stool balance as<br />

measured by a comparison of 72-h fecal output<br />

with oral or enteral intake. Since multiple variables<br />

may affect fat absorption, including intestinal<br />

transit, lymphatic integrity, exocrine pancreas<br />

function and mucosal injury, assessment of fat<br />

balance is very relevant. However, as most<br />

patients receive a mixture of LCTs and MCTs, the<br />

measurement of fat excretion by using the Van de<br />

Kamer assay is not relevant. It is thus preferable<br />

to use the Jeejeebhoy method that extracts and<br />

detects both LCTs and MCTs. 125 In our practice<br />

we also used to measure energy balance by<br />

performing bomb calorimetry analysis. Stool<br />

balance analysis in 20 PN-weaned transplanted<br />

children, showed that lipid absorption rate was<br />

decreased, ranging between 70 and 88%.<br />

Nutritional outcome after intestinal<br />

transplantation<br />

Besides the enteral or oral feeding approach,<br />

nutritional evaluation following intestinal transplantation<br />

is mandatory. However, few nutritional<br />

data are available in children after intestinal<br />

transplantation. 126–128 We studied 20 children<br />

(8.8±3.5 years) with at least 12 months followup<br />

(median 27 months). 128 PN was stopped<br />

110±84 days after grafting. They were on unrestricted<br />

oral feeding, with enteral supplementation<br />

in six patients with varying degrees of eating<br />

disorders. At 12 months post-transplant, body<br />

weight-for-age and height-for-age were decreased,<br />

being 89±13% and 93±5%, respectively. Body<br />

weight-for-height was increased (105±9%) with<br />

a fat body mass (FBM) of 20.1±4.3% of body<br />

weight. Growth velocity was decreased 6 months<br />

before and after grafting by 67±38% and<br />

57±46% of the normal for age, respectively, but<br />

was increased by 142±64% of the normal for age<br />

during the last 6 months of follow-up. Bone<br />

mineral density (BMD), measured by using dual<br />

X-ray absorptiometry (DEXA), was decreased in<br />

all children according to age. Nutritional parameters<br />

showed normal albumin and pre-albumin<br />

plasma levels as well as plasma tocopherol.<br />

Conversely, both vitamin A and RBP plasma<br />

levels were decreased, as well as serum iron and<br />

ferritine.<br />

Weight gain with excessive FBM and catch-up<br />

growth is observed after intestinal transplantation<br />

in most cases, in our experience. However, in<br />

some cases, there is evidence of severe inhibition<br />

of linear growth at the time of transplantation<br />

with no evidence of catch-up after transplantation.<br />

127,128 Growth velocity is severely altered<br />

both before and just after transplantation due to<br />

liver disease and high-dose steroid thereafter.<br />

Thus catch-up growth is delayed. A follow-up<br />

period of 6 months is too short to observe positive<br />

trends in height/length. Despite normal growth,<br />

BMD is low and some nutritional biological parameters<br />

are altered, such as vitamin A plasma<br />

levels. Monitoring should not be restricted to<br />

growth parameters but include body composition<br />

and biological parameters recording nutritional<br />

supplementation if required. Post-intestinal transplantation<br />

nutritional management should<br />

include BMD assessment on a longitudinal basis.<br />

Insufficient BMD may be due to previous longterm<br />

PN worsened with high-dose steroids.<br />

Potential candidates for intestinal<br />

transplantation<br />

In order to improve the risk–benefit ratio and costeffectiveness<br />

of intestinal transplantation it is<br />

mandatory to select appropriate candidates and to<br />

decide the adequate time for performing the procedure.<br />

The number of potential candidates for<br />

intestinal transplantation is very difficult to establish<br />

according to the heterogeneity of patients<br />

included in home-PN programs, and in general, to<br />

the mostly well-tolerated long-term PN. 8–12 The<br />

number of patients will never be as high as the<br />

number of kidney or liver transplant recipients.


However, intestinal transplantation is theoretically<br />

indicated for all patients permanently dependent<br />

on PN. Functional grafts lead to gastrointestinal<br />

autonomy (weaning off PN) while maintaining<br />

satisfactory nutritional status and normal growth<br />

in children. However, as PN is generally well tolerated,<br />

even for long periods, each indication for<br />

transplantation must be carefully weighed in<br />

terms of survival rate, morbidity and quality of<br />

life. 129–133 By excluding malignant disease and<br />

immune deficiency, survival rates of patients with<br />

chronic intestinal failure on long-term home PN<br />

remain higher than after intestinal transplantation,<br />

even with the most optimistic interpretation of<br />

available data. 8–12 Intestinal transplantation is<br />

often accompanied by numerous life-threatening<br />

complications such as those briefly reviewed<br />

above, leading to recurrent or long-term hospitalization<br />

and sometimes a poor outcome. An evaluation<br />

of the quality-of-life after intestinal transplantation<br />

among home PN was recently<br />

performed in adult patients using a quality-of-life<br />

instrument in the form of a self-administered questionnaire.<br />

131 Intestinal transplant recipients with<br />

functioning grafts reported a significant improvement<br />

in the quality of their life and function. This<br />

information is encouraging and should be used<br />

towards future advancement in intestinal transplantation.<br />

Thus, when long-term PN is effective and well<br />

tolerated, it can thus be used pending further<br />

progress in intestinal transplantation. In contrast,<br />

when PN has reached its limits, especially in<br />

those cases associated with extensive thrombosis,<br />

recurrent sepsis, severe metabolic disorders or<br />

advanced liver disease, intestinal transplantation<br />

must be undertaken.<br />

Intestinal transplantation: isolated or<br />

combined with liver<br />

Patients with irreversible intestinal failure and<br />

end-stage liver disease (liver cirrhosis and complicated<br />

portal hypertension) are certainly candidates<br />

for a life-saving procedure such as<br />

combined small bowel–liver transplantation.<br />

Patients with severe hepatic fibrosis are more<br />

difficult to manage. Repeated liver biopsies within<br />

6–12 months and careful assessment for portal<br />

Potential candidates for intestinal transplantation 711<br />

hypertension are mandatory. It is difficult to<br />

predict the liver damage related to the persistent<br />

need for PN or to rejection or infection during the<br />

first weeks or months after transplantation. In<br />

addition, it is difficult to assess the amount of<br />

functioning liver necessary to withstand the insult<br />

of portal diversion during the transplantation<br />

procedure. Those patients with severe hepatic<br />

fibrosis or cirrhosis are thus usually listed for<br />

small bowel–liver transplantation.<br />

Patients with irreversible intestinal failure and PN<br />

dependency without consistent liver disease must<br />

satisfy rigorous criteria to be considered as candidates<br />

for isolated small-bowel transplantation.<br />

They must fulfill at least one of the following<br />

criteria: extensive thrombosis impeding the<br />

ability to administer PN, recurrent life-threatening<br />

sepsis, severe metabolic disorders preventing<br />

nutritional requirements from being met, with<br />

consequent failure to thrive in children, underlying<br />

disease with high water–electrolyte losses<br />

with the risk of life-threatening dehydration in the<br />

case of PN disruption. Some patients with severe<br />

chronic intestinal pseudo-obstruction may be<br />

disabled because of chronic, massive gastrointestinal<br />

dilation refractory to stomal decompression<br />

or partial enterectomy. They might be considered<br />

for intestinal transplantation, although the<br />

usual indications, including progressive liver<br />

disease, the threatened loss of vascular access and<br />

recurring life-threatening sepsis, have not developed.<br />

Contraindications to intestinal transplantation<br />

Contraindications to intestinal transplantation do<br />

not differ from those pertaining to other solid<br />

organs. 134 According to the high morbidity and<br />

mortality rates after intestinal transplantation<br />

and the donor pool size, the concept that the<br />

patient must have the potential to derive an<br />

obvious benefit from the procedure must remain<br />

paramount. Other contraindications include<br />

congenital or acquired neurological disabilities,<br />

life-threatening extradigestive illnesses, congenital<br />

or acquired immune disorders, non-resectable<br />

malignancy, and insufficient vascular patency to<br />

guarantee easy central venous access for up to 6<br />

months after intestinal transplantation.


712<br />

Medical aspects of intestinal transplantation<br />

Timing for referral and prevention of liver<br />

disease<br />

Intestinal failure management<br />

Unfortunately, few centers manage all the stages of<br />

intestinal failure from onset to intestinal transplantation,<br />

including the home-PN program. Beath<br />

et al135 reported a marked discrepancy in clinical<br />

status between children referred for intestinal<br />

transplantation from centers with and without<br />

nutritional care teams. It was shown that<br />

the mortality rate, death within 6 months of<br />

evaluation for transplantation, was 90% in children<br />

with short guts, 50% in those with mucosal<br />

disease and 40% in those with chronic intestinal<br />

pseudo-obstruction syndrome. 136 Factors impacting<br />

the survival of children with intestinal failure<br />

referred for intestinal transplantation have been<br />

studied in a series of 257 patients (mean age<br />

3.4±0.26 years) evaluated for intestinal transplantation.<br />

66 Only 82 (32%) underwent intestinal<br />

transplantation (68 small bowel–liver transplantation)<br />

with a mean waiting time of 10.1±1.3<br />

months. Of the 175 patients who were not transplanted,<br />

120 died. The main factors associated<br />

with poor prognosis were: age below 1 year, surgical<br />

disease, bridging fibrosis or cirrhosis, bilirubin<br />

levels of over 3 mg/dl and thrombocytopenia.<br />

Combined small bowel–liver transplantation<br />

It is sometimes difficult to decide on liver transplantation.<br />

Indeed, some PN-dependent patients<br />

with advanced liver dysfunction in the setting of<br />

short-bowel syndrome may experience functional<br />

and biochemical liver recovery which appears to<br />

parallel autologous gut salvage. 137 On the other<br />

hand, we reported consecutive cases of severe<br />

cholestasis that totally resumed after discontinuation<br />

of intravenous lipid administration. 138<br />

Histology is not always predictive of functional<br />

liver recovery. However, in our experience, in the<br />

absence of biological change and PN administration,<br />

the time taken to go from portal fibrosis to<br />

cirrhosis is approximately 12 months, similar to<br />

the waiting time. 139 Once cirrhosis has been established,<br />

survival at 1 year is only 30%. 66,135<br />

It is well established that patients referred for<br />

small bowel–liver transplantation are more debilitated,<br />

have multiple complications, and have<br />

prolonged stays in the intensive care unit. 140 It<br />

may explain the lower patient and graft survival<br />

rate compared with isolated small-bowel<br />

transplantation reported from several programs.<br />

60–62,64,65 It was suggested that early isolated<br />

small-bowel transplantation with such a short<br />

period of postoperative hospitalization could well<br />

prove to be cost-effective compared with the<br />

intensive use of resources that characterizes the<br />

short-bowel patient with liver failure. 141<br />

Financial considerations<br />

Financial issues are now considered in managing<br />

intestinal failure patients. Provision of basic home<br />

PN is associated with charges. Actual costs for<br />

total PN, including the pharmacist’s time for<br />

compounding, are different according to home-PN<br />

programs and countries. The costs for transplant<br />

evaluation, the transplant and postoperative care,<br />

and post-transplant follow-up are not currently<br />

available for comparison. 142,143<br />

Short-bowel syndrome<br />

Patients with short-bowel syndrome can lead a<br />

productive, lengthy, happy and useful life if<br />

educated and managed appropriately. It is possible<br />

to reduce or even eliminate PN requirements<br />

over time in many of these patients using the<br />

evidence-based techniques of dietary and PN<br />

management (see Chapter 29). Hormonal therapy<br />

may eventually be available to augment the<br />

intestinal adaptation process as this becomes<br />

better understood. For patients with irreversible<br />

intestinal failure who do require total PN, it is<br />

essential that the therapy be prescribed appropriately.<br />

PN is associated with several potentially<br />

serious complications, many of which can be<br />

prevented when both the patient and the caring<br />

professional have the appropriate expertise.<br />

Early referral for irreversible intestinal failure<br />

As pediatric patients represent almost two-thirds<br />

of the indications for small-bowel tranplantation,<br />

appropriate therapeutic strategies should be<br />

developed for all phases of intestinal failure. It is<br />

first necessary to recognize, as early as possible,<br />

the patient with irreversible intestinal failure,<br />

such as extreme short-bowel syndrome or congen-


ital disease of the intestinal mucosa. These<br />

patients should be referred at an early stage to<br />

multidisciplinary teams involved in small-bowel<br />

transplantation in optimal nutritional status. For<br />

other patients, attempts at achieving intestinal<br />

autonomy (PN weaning) require appropriate<br />

management as previously emphasized. Intestinal<br />

transplantation thus requires a strategy based on a<br />

long-term multidisciplinary approach, aimed at<br />

demonstrating the irreversibility of intestinal<br />

failure, to avoid the complications of long-term<br />

PN and to refer early selected patients requiring<br />

this procedure. 141–145<br />

Particular procedures<br />

Because of the reduced size of the donor pool,<br />

strategies have been developed to enhance it and<br />

to perform transplantation without delay to avoid<br />

death on the waiting list, if the patient is in need<br />

of a combined liver–intestine transplantation.<br />

Living related donors<br />

Intestinal grafts are usually obtained from sizematched<br />

brain-dead adults or children. The ABO<br />

identity is necessary to avoid hemolysis related to<br />

circulating antibodies. It is difficult to obtain HLA<br />

compatibility, mainly for logistical reasons (the graft<br />

poorly tolerates ischemia). However, the longest<br />

survival time before the advent of cyclosporin A<br />

was observed after intrafamilial transplantation.<br />

Long-term survival has since been obtained in<br />

intrafamilial transplantation with cyclosporin A or<br />

FK506 immunosuppression. 146–149 A unique case of<br />

a successful isolated intestinal transplantation<br />

without immunosuppressive treatment between<br />

identical twins was recently reported. 149 However,<br />

living-related intestinal transplantation should<br />

remain very limited.<br />

Reduced-size composite liver–intestinal<br />

allograft<br />

Strict donor selection to prevent factors that may<br />

adversely affect the intestinal graft and size-match-<br />

Particular procedures 713<br />

ing have limited donor availability. 66 These factors<br />

contribute to the long waiting time for pediatric<br />

patients. Reduced-sized orthotopic composite<br />

liver–intestinal allografts are technically feasible<br />

and may increase the donor pool. 150,151<br />

Multivisceral transplantation<br />

Multivisceral or ‘cluster’ transplantation of organs<br />

from the celiac region (liver, stomach, duodenum–pancreas<br />

and small bowel) was reported for<br />

the first time in 1989. From the Registry, it represents<br />

13% in children and 24% in adults of all<br />

intestinal grafts. This procedure raises specific<br />

technical problems, but immunosuppressive treatment<br />

is no different. The Miami group performed<br />

multivisceral transplantation in very small children<br />

as an alternative to en bloc liver–intestine<br />

transplantation due to its advantage in creating<br />

more room in the abdominal space. 152 They<br />

reported improved survival rates with an excellent<br />

functional outcome of the stomach and the<br />

pancreas.<br />

Conclusion<br />

During the past decade, intestinal transplantation<br />

has gone from being an experimental procedure to<br />

one that is being increasingly accepted and<br />

performed. Further improvements depend on<br />

several factors, including the early and adapted<br />

management of patients with intestinal failure,<br />

refractory or likely to become refractory, by trained<br />

medico-surgical teams, improvements of immunosuppression<br />

by using new-generation drugs or<br />

monoclonal antibodies targeting receptors<br />

involved in T-cell activation, and the better<br />

prevention of post-transplant infectious complications<br />

by using appropriate antiviral therapy. 153–156<br />

Constant progress in immunosuppression<br />

provides the hope of continuing to improve<br />

outcome after intestinal transplantation in selected<br />

patients. Indeed, when successful, intestinal transplantation<br />

allows patients surviving with their<br />

graft a recovery of intestinal function sufficient to<br />

discontinue a mostly long-term dependency on<br />

parenteral nutrition.


714<br />

Medical aspects of intestinal transplantation<br />

REFERENCES<br />

1. Schraut W. Current status of small bowel transplantation.<br />

Gastroenterology 1988; 94: 525–535.<br />

2. Goulet O, Révillon Y, Brousse N et al. Successful small<br />

bowel transplantation in an infant. Transplantation<br />

1992; 53: 940–943.<br />

3. Schroeder P, Goulet O, Lear P. Small bowel transplantation:<br />

European experience. Lancet 1990; 336: 110–111.<br />

4. Grant D, Wall W, Mimerault R. Successful small<br />

bowel–liver transplantation. Lancet 1990; 335: 181–184.<br />

5. Todo S, Tsakis A, Abu-Elmagd K et al. Cadaveric small<br />

bowel and small bowel–liver transplantation in<br />

humans. Transplantation 1992; 53: 369–376.<br />

6. Grant D. Intestinal Transplantation Registry on behalf of<br />

the current results of intestinal transplantation. Lancet<br />

1996; 347: 1801–1803.<br />

7. Grant D. Intestinal transplantation: 1997 Report of the<br />

International Registry. Transplantation 1999; 15:<br />

1061–1064.<br />

8. Colomb V, Goulet O, Ricour C. Home enteral and<br />

parenteral nutrition. Baillière’s Clin Gastroenterol 1998;<br />

122: 877–894.<br />

9. Messing B, Lemann M, Landais P et al. Prognosis of<br />

patients with chronic intestinal failure receiving longterm<br />

home parenteral nutrition in France and Belgium.<br />

Gastroenterology 1995; 108: 1005–1010.<br />

10. Howard L, Ament M, Fleming R et al. Current use and<br />

clinical outcome of home parenteral and enteral nutrition<br />

therapies in the United States. Gastroenterology<br />

1995; 109: 355–365.<br />

11. Howard L, Hassan N. Home parenteral nutrition: 25<br />

years later. Clin Nutr 1998; 27: 418–512.<br />

12. Howard L, Malone M. Current status of home parenteral<br />

nutrition in the United States. Transplant Proc 1996; 28:<br />

2691–2695.<br />

13. Goulet O, Baglin-Gobet S, Jais JP et al. Outcome and<br />

long-term growth after extensive small bowel resection<br />

in the neonatal period: a survey of 87 children. Eur J<br />

Pediatr Surg 2004; in press.<br />

14. Sondheimer JM, Cadnapaphornchai M, Sontag M, Zerbe<br />

GO. Predicting the duration of dependence on<br />

parenteral nutrition after neonatal intestinal resection. J<br />

Pediatr 1998; 132: 80–84.<br />

15. Goulet O, Révillon Y, Jan D et al. Which patients need<br />

small bowel transplantation for neonatal short bowel<br />

syndrome. Transplant Proc 1992; 24: 1058–1059.<br />

16. Goulet O, Ricour C. The short bowel syndrome. In Buts<br />

JP, Sokal EM, eds. Elsevier Science Publishers BV, 1993:<br />

307–318.<br />

17. Bianchi A. Longitudinal intestinal lengthening and<br />

tailoring: results in 20 children. J R Soc Med 1997; 90:<br />

429–432.<br />

18. Thompson JS, Langnas AN, Pinch LW et al. Surgical<br />

approach to short-bowel syndrome. Experience in a<br />

population of 160 patients. Ann Surg 1995; 222:<br />

600–605.<br />

19. Berger DL, Malt RA. Management of the short gut<br />

syndrome. Adv Surg 1996; 29: 43–57.<br />

20. Vanderhoof JA. Short bowel syndrome in children and<br />

small intestinal transplantation. Pediatr Clin North Am<br />

1996; 43: 533–546.<br />

21. Panis Y, Messing B, Rivet P et al. Segmental reversal of<br />

the small bowel as an alternative to intestinal transplantation<br />

in patients with short bowel syndrome. Ann Surg<br />

1997; 225: 401–407.<br />

22. Shulman DI, Shih Hu C, Duckett G, Lavallee-Grey M.<br />

Effects of short-term growth hormone therapy in rats<br />

undergoing 75% small intestinal resection. J Pediatr<br />

Gastroenterol Nutr 1992; 14: 3–11.<br />

23. Ulshen MA, Dowling RH, Fuller CR et al. Enhanced<br />

growth of small bowel in transgenic mice overexpressing<br />

bovine growth hormone. Gastroenterology 1993; 104:<br />

973–980.<br />

24. Benhamou PH, Canarelli JP, Leroy C et al. Stimulation<br />

by recombinant human growth hormone of growth and<br />

development of remaining bowel after subtotal ileojejunectomy<br />

in rat. J Pediatr Gastroenterol Nutr 1994; 18:<br />

446–452.<br />

25. Byrne TA, Persinger RL, Young LS et al. A new treatment<br />

for patients with short-bowel syndrome: growth<br />

hormone, glutamine and a modified diet. Ann Surg<br />

1995; 22: 243–255.<br />

26. Scolapio JS, Camilleri M, Fleming CR et al. Effect of<br />

growth hormone, glutamine, and diet on adaptation in<br />

short-bowel syndrome: a randomized, controlled study.<br />

Gastroenterology 1997; 113: 1074–1081.<br />

27. Seguy D, Vahedi K, Kapel N et al. Low-dose growth<br />

hormone in adult home PN-dependent short bowel<br />

patients: a positive study. Gastroenterology 2003; 124:<br />

293–302.<br />

28. Dabbas-Tyan M, Colomb V, Rosilio M et al. Evaluation of<br />

the effect of recombinant human growth hormone<br />

(rhGH) treatment of children with short bowel<br />

syndrome. J Pediatr Gastroenterol Nutr 2000; 31:<br />

5165–5166.<br />

29. Ellegard L, Bosaeus I, Nordgren S, Bengtsson BA. Lowdose<br />

recombinant human growth hormone increases<br />

body weight and lean body mass in patients with short<br />

bowel syndrome. Ann Surg 1997; 225: 88–96.<br />

30. Szkudlarek J, Jeppesen P-B, Mortensen P-B. Effect of<br />

high dose growth hormone with glutamine and no<br />

change in diet on intestinal absorption in short bowel<br />

patients: a randomised, double blind, crossover, placebo<br />

controlled study. Gut 2000; 47: 199–205.<br />

31. Jeppesen PB, Mortensen AP. Enhancing bowel adaptation<br />

in short bowel syndrome. Curr Gastroenterol Rep<br />

2002; 4: 338–347.<br />

32. Beath SV, Davies P, Papadopoulou A et al. Parenteral<br />

nutrition-related cholestasis in postsurgical neonates:<br />

multivariate analysis of risk factors. J Pediatr Surg 1996;<br />

31: 604–606.<br />

33. Lawrence J P, Dun SP, Billmire DF et al. Isolated liver<br />

transplantation for liver failure in patients with short<br />

bowel syndrome. J Pediatr Surg 1994; 29: 751–753.<br />

34. Horslen SP, Sudan DL, Iyer KR et al. Isolated liver transplantation<br />

in infants with end-stage liver disease associated<br />

with short bowel syndrome. Ann Surg 2002; 235:<br />

435–439.<br />

35. Hassan OK, Beath SV, McKlerman PJ et al. Difficult<br />

management choices for infants with short bowel<br />

syndrome and liver failure. J Pediatr Gastroenterol Nutr<br />

2002; 35: 216–219.<br />

36. Sharif K, Beath SV, Kelly DA et al. New perspective for<br />

the management of near-total or total intestinal aganglionosis<br />

in infants. J Pediatr Surg 2003; 38: 25–28.<br />

37. Révillon Y, Aigrain Y, Jan D et al. Improved quality of<br />

life by combined transplantation in Hirschsprung’s<br />

disease with a very long aganglionic segment. J Pediatr<br />

Surg 2003; 38: 422–424.<br />

38. Goulet O, Jobert-Giraud A, Michel JL et al. Chronic<br />

intestinal pseudoobstruction syndrome in pediatric<br />

patients. Eur J Pediatr Surg 1999; 9: 83–90.


39. Rudolph CD, Hyman PE, Altschuler SM et al. Diagnosis<br />

and treatment of chronic intestinal pseudoobstruction<br />

in children: report of consensus workshop. J Pediatr<br />

Gastroenterol Nutr 1997; 24: 102–112.<br />

40. Faure C, Goulet O, Ategbo S et al. Chronic intestinal<br />

pseudoobstruction syndrome: clinical analysis, outcome<br />

and prognosis in 105 children. Dig Dis Sci 1999; 44:<br />

953–959.<br />

41. Sigurdsson L, Reyes J, Kocoshis SA et al. Intestinal<br />

transplantation in children with chronic intestinal<br />

pseudo-obstruction. Gut 1999; 45: 570–574.<br />

42. Masetti M, Rodriguez MM, Thompson JF et al.<br />

Multivisceral transplantation for megacystis microcolon<br />

intestinal hypoperistalsis syndrome. Transplantation<br />

1999; 68: 228–232.<br />

43. Kosmach B, Tarbell S, Reyes J, Todos S. ‘Munchausen<br />

by proxy’ in a small bowel transplant recipient.<br />

Transplant Proc 1996; 28: 2790–2791.<br />

44. Phillips A, Schmitz J. Congenital microvillus atrophy: a<br />

clinico-pathological survey of 23 cases. J Pediatr<br />

Gastroenterol Nutr 1992; 14: 380–396.<br />

45. Goulet O, Kedinger M, Brousse N et al. Intractable diarrhea<br />

of infancy with epithelial and basement membrane<br />

abnormalities. J Pediatr 1995; 127: 212–219.<br />

46. Oliva MM, Perman JA, Saavedra JM et al. Successful<br />

intestinal transplantation for microvillous inclusion<br />

disease. Gastroenterology 1994; 106: 771–774.<br />

47. Lacaille F, Cuenod B, Colomb V et al. Combined liver<br />

and small bowel transplantation in a child with epithelial<br />

dysplasia. J Pediatr Gastroenterol Nutr 1998; 27:<br />

230–233.<br />

48. Sudan DL, Kaufman SS Shaw BW Jr et al. Isolated<br />

intestinal transplantation for intestinal failure. Am J<br />

Gastroenterol 2000; 95: 1506–1515<br />

49. Herzog D, Atkinson P, Grant D et al. Combined bowelliver<br />

transplantation in an infant with microvillus inclusion<br />

disease. J Pediatr Gastroenterol Nutr 1996; 22:<br />

405–408.<br />

50. Randak C, Langnas AN, Kaufman SS et al. Pretransplant<br />

management and small bowel-liver transplantation in<br />

an infant with microvillus inclusion disease. J Pediatr<br />

Gastroenterol Nutr 1998; 27: 333–337.<br />

51. Ruemmele F, Révillon Y, Jan D et al. New perspectives<br />

for children with microvillous inclusion disease: early<br />

small bowel transplantation. Transplantation 2004; 77:<br />

1024–1028.<br />

52. Paramesh AS, Fishbein T, Tschernia A et al. Isolated<br />

small bowel transplantation for tufting enteropathy. J<br />

Pediatr Gastroenterol Nutr 2003; 36: 138–140.<br />

53. Fishbein TM, Florman S, Gondolesi G et al. Intestinal<br />

transplantation before and after the introduction of<br />

sirolimus. Transplantation 2002; 73: 1538–1542.<br />

54. Berribi C, Loirat C, Jacqz-Aigrain E. Mycophenolate<br />

mofetil may induce apoptosis in duodenal villi. Pediatr<br />

Nephrol 2000; 14: 177–178.<br />

55. Tzakis AG, Kato T, Nishida S et al. Preliminary experience<br />

with campath 1H (C1H) in intestinal and liver<br />

transplantation. Transplantation 2003; 75: 1227–1231.<br />

56. Reyes J, Mazariegos GV, Bond GM et al. Pediatric intestinal<br />

transplantation: historical notes, principles and<br />

controversies. Pediatr Transplant 2002; 6: 193–207.<br />

57. Wekerle T, Kurtz J, Sykes. Mixed hematopoietic<br />

chimerism and transplantation tolerance: insights from<br />

experimental models. Curr Opin Organ Transplant 1999;<br />

4: 44–49<br />

58. Pirenne J, Gruessner AC, Beneditti E. Donor-specific<br />

unmodified bone marrow transfusion does not facilitate<br />

intestinal engraftment after bowel transplantation in a<br />

porcine model. Surgery 1997; 121: 79–88.<br />

References 715<br />

59. De Roover A, Langnas AN. Surgical methods of small<br />

bowel transplantation. Curr Opin Organ Transplant<br />

1999; 4: 335–342.<br />

60. Reyes J, Bueno J, Kocoshis S et al. Current status of<br />

intestinal transplantation in children. J Pediatr Surg<br />

1998; 33: 243–254.<br />

61. Farmer DG, McDiarmid SV, Smith C et al. Experience<br />

with combined liver–small intestine transplantation at<br />

the University of California, Los Angeles. Transplant<br />

Proc 1998; 30: 2533–2534.<br />

62. Atkison P, Williams S, Wall S, Grant D. Results of pediatric<br />

small bowel transplantation in Canada. Transplant<br />

Proc 1998; 30: 2521–2522.<br />

63. Nishida S, Levi D, Kato T et al. Ninety-five cases of<br />

intestinal transplantation at the University of Miami. J<br />

Gastrointest Surg 2002; 6: 233–239.<br />

64. Goulet O, Lacaille F, Colomb V et al. Intestinal transplantation<br />

in children: Paris experience. Transplant Proc<br />

2002; 34: 1887–1888.<br />

65. Iyer KR, Srinath C, Horslen S et al. Late graft loss and<br />

long-term outcome after isolated intestinal transplantation<br />

in children. J Pediatr Surg 2002; 37: 151–154.<br />

66. Bueno J, Ohwada S, Kocoshis S et al. Factors impacting<br />

the survival of children with intestinal failure referred for<br />

intestinal transplantation. J Pediatr Surg 1999; 34: 27–33.<br />

67. Sarnacki S, Révillon Y, Cerf-Bensussan N et al. Long<br />

term small bowel graft survival induced by spontaneously<br />

tolerated liver allograft in inbred rat strains.<br />

Transplantation 1992; 54: 383–385.<br />

68. Zhong R, He G, Sakai Y et al. Combined small bowel<br />

and liver transplantation in the rat: possible role of the<br />

liver in preventing intestinal allograft rejection.<br />

Transplantation 1991; 52: 550–552.<br />

69. Goulet O, Jan D, Lacaille F et al. Intestinal transplantation<br />

in children: preliminary experience in Paris. J<br />

Parenter Enter Nutr 1999; 23: S121–S125.<br />

70. Kamada N, Shinomiya T. Clonal deletion as the mechanism<br />

of abrogation of immunological memory following<br />

liver grafting in rats. Immunology 1985; 55: 85–90.<br />

71. Tu Y, Arima T, Flye MW. Rejection of spontaneously<br />

accepted rat liver allografts with recipient interleukin 2<br />

treatment or donor irradiation. Transplantation 1997;<br />

63: 177–181.<br />

72. Thomson AW. Are dendritic cells the key to liver transplant<br />

tolerance. Immunol Today 1999; 20: 27–32.<br />

73. Sarnacki S, Nakai H, Calise D et al. Decreased expression<br />

of CD25 on CD8+ recipient T cells in intestinal<br />

grafts tolerated after liver transplantation in rats. Gut<br />

1998; 43: 849–855.<br />

74. Bishop GA, Sharland AF, McCaughan GW. Highdose/activation<br />

associated tolerance model for allografts:<br />

lesson from spontaneous tolerance of transplanted<br />

livers. Curr Opin Organ Transplant 1999; 4: 58–64.<br />

75. Iwaki Y, Starzl TE, Yagihasni A et al. Replacement of<br />

donor lymphoid tissue in small bowel transplantation.<br />

Lancet 1991; 337: 818–819.<br />

76. Goulet O. Recent studies on small intestinal transplantation.<br />

Gastroenterology 1997; 13: 500–509.<br />

77. Kaufman SS, Wisecarver JL, Ruby EI et al. Correlation of<br />

mucosal disaccharidase activities with histology in evaluation<br />

of rejection following intestinal transplantation.<br />

Pediatr Transpl 1998; 2: 134–138.<br />

78. Goulet O, Brousse N, Révillon Y, Ricour C. Pathology of<br />

human intestinal transplantation. In Grant D, Wood<br />

RFM, eds. Small Bowel Transplantation. London:<br />

Edward Arnold, 1993: 112–120.<br />

79. Lee RG, Nakamura K, Tsamandas AC et al. Pathology of<br />

human intestinal transplantation. Gastroenterology<br />

1996; 110: 1820–1834.


716<br />

Medical aspects of intestinal transplantation<br />

80. Sigurdsson L, Reyes J, Todo S et al. Anatomic variability<br />

of rejection in intestinal allografts after pediatric intestinal<br />

transplantation. J Pediatr Gastroenterol Nutr 1998;<br />

27: 403–406.<br />

81. Nogushi Si S, Reyes J, Maeariegos GV, Parizhskaya M,<br />

Jaffe R. Pediatric intestinal transplantation: the resected<br />

allograft. Pediatr Dev Pathol 2002; 5: 3–21.<br />

82. Parizhskaya M, Redondo C, Demetris A et al. Chronic<br />

rejection of small bowel grafts: pediatric and adult<br />

study of risk factors and morphologic progression.<br />

Pediatr Dev Pathol 2003; 6: 240–250<br />

83. Wu T, Abu-Elmagd K, Nalesnik MA et al. A schema for<br />

histologic grading of small intestine allograft acute<br />

rejection. Transplantation 2003; 75: 1241–1248.<br />

84. Green M, Bueno J, Sigurdsson L et al. Unique aspects of<br />

the infectious complications of intestinal transplantation.<br />

Curr Opin Organ Transplant 1999; 4: 361–367.<br />

85. Patel R, Snydman DR, Rubin RH et al. Cytomegalovirus<br />

prophylaxis in solid organ transplant recipients.<br />

Transplantation 1996; 61: 1279–1289.<br />

86. Finn L, Reyes J, Bueno J, Yunis E. Epstein–Barr virus<br />

infection in children after transplantation of the small<br />

intestine. Am J Surg Pathol 1998; 22: 299–309.<br />

87. Green M, Reyes J, Jabbour N et al. Use of quantitative<br />

PCR to predict onset of Epstein–Barr viral infection<br />

and post-transplant lymphoproliferative disease after<br />

intestinal transplantation in children. Transplant Proc<br />

1996; 28: 2759–2760.<br />

88. Berney T, Delis S, Kato T et al. Successful treatment of<br />

posttransplant lymphoproliferative disease with<br />

prolonged rituximab treatment in intestinal transplant<br />

recipients. Transplantation 2002; 74: 1000–1006.<br />

89. Furukawa H, Kusne S, Sutton DA et al. Acute invasive<br />

sinusitis due to trichoderma longibrachiatum in a liver<br />

and small bowel transplant recipient. Clin Infect Dis<br />

1998; 26: 487–489.<br />

90. Muiesan P, Dhawan A, Wendon J et al.<br />

Hemophagocytosis: a potential complication in small<br />

bowel transplantation. Transplantation 1998; 66:<br />

794–796.<br />

91. Berho M, Torroella M, Viciana A et al. Adenovirus enterocolitis<br />

in human small bowel transplants. Pediatr<br />

Transplant 1998; 2: 277–282.<br />

92. Turler A, Kalff JC, Heeckt P et al. Molecular and functional<br />

observations on the donor intestinal muscularis<br />

during human small bowel transplantation.<br />

Gastroenterology 2002; 122: 1886–1897.<br />

93. Sarr MG, Kelly KA. Myoelectric activity of the autotransplantation<br />

canine jejuno-ileum. Gastroenterology<br />

1981; 81: 303–310.<br />

94. Sugitani A, Bauer AJ, Reynolds JC et al. The effects of<br />

small bowel transplantation on the morphology and<br />

physiology of intestinal muscle – a comparison of autografts<br />

versus allografts in dogs. Transplantation 1997;<br />

63: 186–194.<br />

95. Taguchi T, Zorychta E, Sonnino RE, Guttman FM. Small<br />

intestinal transplantation in the rat: effect on physiological<br />

properties of smooth muscle and nerves. J Pediatr<br />

Surg 1989; 24: 1258–1263.<br />

96. Taguchi T, Zorychta E, Sonnino RE, Guttman FM.<br />

Function of smooth muscle and nerve after small intestine<br />

transplantation in the rat: effect of storing donor<br />

bowel in Eurocollins. J Pediatr Surg 1989; 24: 634–638.<br />

97. Ishii H, Kusunoki M, Fujita S et al. Changes of intestinal<br />

motility after small bowel transplantation in the rat.<br />

Transplantation 1994; 57: 1149–1152.<br />

98. Kusunoki M, Ishii H, Nakao K et al. Long-term effects of<br />

small bowel transplantation on intestinal motility.<br />

Transplantation 1995; 60: 897–899.<br />

99. Lu G, Sarr MG, Szurszewski JH. Effect of extrinsic<br />

denervation in a canine model of jejunoileal autotransplantation<br />

on mechanical and electrical activity of<br />

jejunal circular smooth muscle. Dig Dis Sci 1997; 42:<br />

40–46.<br />

100. Suginati A, Reynolds JC, Tsuboi M, Todo S. Extrinsic<br />

intestinal reinnervation after canine small bowel autotransplantation.<br />

Surgery 1998; 123: 25–35.<br />

101. Taguchi T, Guo R, Masumoto K et al. Chronological<br />

change of distribution in nitric oxide and peptidergic<br />

neurons after rat small intestinal transplantation. J<br />

Pediatr Surg 1999; 34: 341–345.<br />

102. Murr MM, Miller VM, Sarr MG. Contractile properties<br />

of enteric smooth muscle after small bowel transplantation<br />

in rats. Am J Surg 1996; 171: 212–217.<br />

103. Hamada N, Hutson WR, Nakada K et al. Intestinal<br />

neuromuscular function after preservation and transplantation.<br />

J Surg Res 1996; 63: 460–466.<br />

104. Lu G, Sarr MG, Szurszewski JH. Effect of intrinsic<br />

denervation in a canine model of jejunoileal autotransplantation<br />

on mechanical and electrical activity of<br />

jejunal circular smooth muscle. Dig Dis Sci 1997; 42:<br />

40–46.<br />

105. Johnson CP, Sarna SK, Zhu YR et al. Effects of intestinal<br />

transplantation on postprandial motility and regulation<br />

of intestinal transit. Surgery 2001; 129: 6–14.<br />

106. Mousa H, Bueno J, Griffiths J et al. Intestinal motility<br />

after small bowel transplantation. Transplant Proc 1998;<br />

30: 2535–2536.<br />

107. Pernthaler H, Kreczy A, Plattner R et al. Myoelectric<br />

activity during small bowel allograft rejection. Dig Dis<br />

Sci 1994; 39: 1216–1221.<br />

108. Klaus A, Klima G, Margreiter R, Pernthaler H.<br />

Myoelectric activity during chronic small bowel allograft<br />

rejection in rats. Dig Dis Sci 2002; 47: 2506–2511.<br />

109. Kenneth KWL, Heeckt PF, Halfter WM et al. Functional<br />

impairment of enteric smooth muscle and nerves<br />

caused by chronic intestinal allograft rejection regresses<br />

after FK506 rescue. Transplantation 1995; 59: 159–164.<br />

110. Telford GL, Nemeth MA, Sarna SK et al. Myoelectric<br />

activity and absorptive capacity of rat small intestinal<br />

isografts. Dig Dis Sci 1996; 41: 1082–1087.<br />

111. Campbell WL, Abu-Elmagd K, Federle MP et al.<br />

Contrast examination of the small bowel in patients<br />

with small-bowel transplants: findings in 16 patients.<br />

Am J Roentgenol 1993; 161: 969–974.<br />

112. Yanchar NL, Riegel TM, Martin G et al. Tacrolimus [FK<br />

506] – its effects on intestinal glucose transport.<br />

Transplantation 1996; 61: 630–634.<br />

113. Oishi AJ, Inoue Y, Souba WW, Sarr MG. Alterations in<br />

carrier-mediated glutamine transport after a model of<br />

canine jejunal autotransplantation. Dig Dis Sci 1996; 41:<br />

1915–1924.<br />

114. Sun SC, Katz SM, Schechner RS et al. Effect of<br />

tacrolimus on hemodynamics and absorption of experimental<br />

small intestinal transplants. Transplantation<br />

1996; 61: 1447–1450.<br />

115. Sigalet DL, Kneteman NN, Fedorak RN et al. Small<br />

intestinal function following syngeneic transplantation<br />

in the rat. J Surg Res 1996; 61: 379–384.<br />

116. Kato Y, Hamada Y, Ito S et al. Epidermal growth factor<br />

stimulates the recovery of glucose absorption after small<br />

bowel transplantation. J Surg Res 1998; 80: 315–319.<br />

117. Kim J, Fryer J, Craig RM. Absorptive function following<br />

small intestinal transplantation. Dig Dis Sci 1998; 43:<br />

1925–1930.<br />

118. Kellersman R, Zhong R, Kiyochi H et al. Reconstruction<br />

of the intestinal lymphatic drainage after small bowel<br />

transplantation. Transplantation 2000; 69: 10–16.


119. Sigalet DL, Williams DC, Garola R et al. Impact of<br />

FK506 and steroids on adaptation after intestinal resection<br />

or segmental transplantation. Pediatr Transplant<br />

2000; 4: 12–20.<br />

120. Libsch KD, Zyromski NJ, Tanaka T et al. Role of extrinsic<br />

innervation in jejunal absorptive adaptation to<br />

subtotal small bowel resection: a model of segmental<br />

small bowel transplantation. J Gastrointest Surg 2002; 6:<br />

240–247.<br />

121. Tsiotos GG, Kendrick ML, Libsch K et al. Ileal absorptive<br />

adaptation to jejunal resection and extrinsic denervation:<br />

implications for living-related small bowel transplantation.<br />

J Gastrointest Surg 2001; 5: 217–224.<br />

122. Raimundo AH, Grimble GK, Rees RG et al. The effect of<br />

carbohydrate and fat on the absorption of amino acids<br />

and peptides in the normal human small intestine<br />

[abstract]. J Parenter Enteral Nutr 1989; 13 (Suppl): 9S.<br />

123. Van Der Hulst RRJ, Van Kreel BK, Von Meyenfeldt MF et<br />

al. Glutamine and the preservation of gut integrity.<br />

Lancet 1993; 341: 1363–1365.<br />

124. Kaufman SS, Lyden ER, Brown CR et al. Disaccharidase<br />

activities and fat assimilation in pediatric patients after<br />

intestinal transplantation. Transplantation 2000; 15:<br />

362–365.<br />

125. Caliari S, Vantini I, Sembenini C et al. Fecal fat<br />

measurement in the presence of long- and mediumchain<br />

triglycerides and fatty acids. Scand J Gastroenterol<br />

1996; 31: 863–867.<br />

126. Iyer K, Horslen S, Iverson A et al. Nutritional outcome<br />

and growth of children after intestinal transplantation. J<br />

Pediatr Surg 2002; 37: 464–466.<br />

127. Nucci AM, Barksdale EM Jr, Beserock N et al. Long-term<br />

nutritional outcome after pediatric intestinal transplantation.<br />

J Pediatr Surg 2002; 37: 460–463.<br />

128. Goulet O, Allegri A, Colomb V et al. Growth and nutritional<br />

status after intestinal transplantation in children.<br />

J Pediatr Gastroenterol Nutr 2000; 31 (Suppl 2): S165 [A]<br />

129. Brook G. Quality of life issues: parenteral nutrition to<br />

small bowel transplantation – a review. Nutrition 1998;<br />

14: 813–816.<br />

130. Rovera GM, Di Martini A, Schoen RE et al. Quality of<br />

life of patients after intestinal transplantation.<br />

Transplantation 1998; 66: 1141–1145.<br />

131. Tarbell SE, Kosmach B. Parenteral psychosocial<br />

outcomes in pediatric liver and/or intestinal transplantation:<br />

pretransplantation and the early postoperative<br />

period. Liver Transpl Surg 1998; 4: 378–387.<br />

132. Iyer K, Kaufman S, Sudan D et al. Long-term results of<br />

intestinal transplantation for pseudo-obstruction in<br />

children. J Pediatr Surg 2001; 36: 174–177.<br />

133. Sudan D, Iyer K, Horslen S et al. Assessment of quality<br />

of life after pediatric intestinal transplantation by<br />

parents and pediatric recipients using the child health<br />

questionnaire. Transplant Proc 2002; 34: 963–964.<br />

134. Kaufman S, Atkinson JB, Bianchi A et al. Indications for<br />

pediatric intestinal transplantation: a position paper of<br />

the American society of transplantation. Pediatr<br />

Transplant 2001; 5: 80–87.<br />

135. Beath SV, Booth IW, Murphy MS et al. Nutritional care<br />

and candidates for small bowel transplantation. Arch<br />

Dis Child 1995; 73: 348–350.<br />

136. Beath SV, Brook GA, Kelly DA et al. Demand for pediatric<br />

small bowel transplantation in the United<br />

Kingdom. Transplant Proc 1998; 30: 2531–2532.<br />

137. Iyer K, Horslen S, Torres C et al. Histology is not predictive<br />

of functional liver recovery in parenteral-nutrition<br />

associated liver dysfunction. Pediatr Transplant 2003; 7:<br />

69(A).<br />

References 717<br />

138. Colomb V, Jobert-Giraud A, Lacaille F et al. Role of lipid<br />

emulsions in cholestasis associated with long-term<br />

parenteral nutrition in children. J Parenter Enteral Nutr<br />

2000; 24: 345–350.<br />

139. Colomb V, Jobert A, Lacaille F et al. Parenteral nutritionassociated<br />

liver disease in children : natural history and<br />

prognosis. Clin Nutr 1999; 18 (Suppl 1): 36.<br />

140. Filston HC, Colombani PM. Preliminary experience with<br />

intestinal transplantation in infants and children.<br />

Pediatrics 1996; 97: 583–584.<br />

141. Goulet O, Ruemmele F, Lacaille F, Colomb V. Irreversible<br />

intestinal failure. J Pediatr Gastroenterol Nutr 2004; 38:<br />

250–269.<br />

142. Buchman AL, Scolapio J, Fryer J. AGA technical review<br />

on short bowel syndrome and intestinal transplantation.<br />

Gastroenterology 2003; 124: 1111–1134.<br />

143. Kaufman SS. Small bowel transplantation: selection<br />

criteria, operative techniques, advances in specific<br />

immunosuppression, prognosis. Curr Opin Pediatr 2001;<br />

13: 425–428.<br />

144. Fishbein TM, Shciano T, LeLeiko N et al. An integrated<br />

approach to intestinal failure. Results of a new program<br />

with total parenteral nutrition, bowel rehabilitation, and<br />

transplantation. J Gastrointest Surg 2002; 6: 554–562.<br />

145. Martin D, Ezzelarab M, Bond G et al. Patient profile and<br />

candidacy for intestinal transplantation at the<br />

University of Pittsburgh. Transplant Proc 2002; 34:<br />

1897–1898.<br />

146. Jaffe BM, Beck R, Flint L et al. Living-related small<br />

bowel transplantation in adults – a report of two<br />

patients. Transplant Proc 1997; 29: 1851–1852.<br />

147. Fujimoto Y, Uemoto S, Inomata Y et al. Living-related<br />

small bowel transplant: management of rejection and<br />

infection. Transplant Proc 1998; 30: 1149.<br />

148. Ding J, Guo CC, Li CN et al. Postoperative endoscopic<br />

surveillance of human living-donor small bowel transplantations.<br />

World J Gastroenterol 2003; 9: 595–598.<br />

149. Genton L, Raguso CA, Berney T et al. Four year nutritional<br />

follow up after living related small bowel transplantation<br />

between monozygotic twins. Gut 2003; 52:<br />

659–662.<br />

150. Reyes J, Fishbein T, Bueno J et al. Reduced-size orthotopic<br />

composite liver-intestinal allograft.<br />

Transplantation 1998; 66: 489–492.<br />

151. Bueno J, Abu-Elmagd K, Mazariegos G et al. Composite<br />

liver–small bowel allografts with preservation donor<br />

duodenum and hepatic biliary system in children. J<br />

Pediatr Surg 2000; 35: 291–295; discussion 295–296.<br />

152. Mittal N, Kato T, Miller B et al. Multivisceral transplantation<br />

in children – functional outcomes of the transplanted<br />

pancreas and stomach in en-bloc graft. Pediatr<br />

Transplant 2003; 7: 51(A).<br />

153. Abu-Elmagd K, Bond G. The current status and future<br />

outlook of intestinal transplantation. Minerva Chir<br />

2002; 57: 543–560.<br />

154. Sigalet DL, Thorne PC, Martin GR et al. Combined<br />

immunosuppression with cyclosporine, rapamycin, and<br />

mycophenolate mofetil controls rejection with minimal<br />

nutritional impact in experimental small intestinal<br />

transplantation. Transplant Proc 2002; 34: 1121–1123.<br />

155. Pirenne J, Koshiba T, Geboes K et al. Complete freedom<br />

from rejection after intestinal transplantation using a<br />

new tolerogenic protocol combined with low immunosuppression.<br />

Transplantation 2002; 73: 966–968.<br />

156. Starzl TE, Murase N, Abu-Elmagd K et al. Tolerogenic<br />

immunosuppression for organ transplantation. Lancet<br />

2003; 361: 1502–1510.


43<br />

Introduction<br />

Intussusception<br />

Adolfo Bautista Casasnovas<br />

Intussusception is a form of intestinal obstruction,<br />

due to the telescoping or prolapse of one section of<br />

the intestine (the intussusceptum) into an adjacent<br />

section (the intussuscipiens). In most cases, the<br />

terminal ileum telescopes into the colon. Intussusception<br />

occurs most frequently before the<br />

age of 2 years; the incidence is highest in the first<br />

year, during which it is the most frequent cause of<br />

intestinal obstruction.<br />

This disorder has been known since the time of<br />

Aristotle, but the first detailed description was that<br />

of Paul Barbette, who in 1676 proposed surgical<br />

treatment. 1 Until the mid-19th century, however,<br />

intussusception remained almost invariably fatal.<br />

The first successful surgical treatment (of a 2-yearold<br />

boy) was reported in 1873 by Jonathan<br />

Hutchinson. 2 In 1876, Hirschsprung described the<br />

treatment of a series of patients with hydrostatic<br />

enema, marking the start of a period of progress in<br />

treatment. 3 In 1905 Hirschsprung published a new<br />

series of 107 cases, with 35% mortality in patients<br />

treated by enema and 80% mortality in patients<br />

treated surgically.<br />

In 1948 Ravitch and McClure published their<br />

experience with barium contrast radiography, and<br />

established indications and contraindications for<br />

hydrostatic reduction. 4 Subsequent publications<br />

helped generalize the use of hydrostatic reduction,<br />

typically with high success rates (94%) and low<br />

mortality (0.02%). 5–7 In the 1980s, ultrasonography<br />

was introduced as an innocuous and reliable<br />

method for diagnosis.<br />

Current standard treatment practice is based on<br />

careful early diagnosis, generally by contrast radiography<br />

and/or ultrasonography. Treatment<br />

usually involves hydrostatic or pneumatic reduction,<br />

with surgery reserved for a specific small<br />

subset of cases. When this approach is followed,<br />

morbidity is less than 1% and mortality close to<br />

zero.<br />

Epidemiology<br />

Intussusception is the predominant cause of<br />

intestinal obstruction between 5 months and 5<br />

years of age, with incidence differing between<br />

countries and races. In the UK and USA, for<br />

example, reported mean incidences range from<br />

1.5–4.3 per 1000 liveborn children, 8–10 versus<br />

2.7–6.1 per 1000 in Spain. 11,12<br />

Prevalence is typically about three times higher in<br />

boys than in girls, and indeed about eight times<br />

higher in boys from age 4 years onwards.<br />

It can present at any age, but as already noted it is<br />

most common in the first 2 years of life, with incidence<br />

peaking between 3 and 12 months (mean<br />

7–8 months). Two-thirds of cases present in the<br />

first year of life.<br />

Some studies have reported different patterns of<br />

seasonal variation in incidence, but large-sample<br />

and long-series studies suggest that there is no<br />

consistent seasonal pattern. Certainly, however,<br />

the incidence clearly increases during epidemics<br />

of gastroenteritis, respiratory infection and adenovirus<br />

infection, and during periods of vaccination<br />

against rotavirus. 13–16<br />

719


720<br />

Intussusception<br />

Etiopathology<br />

Primary idiopathic intussusception<br />

Despite numerous publications, the etiology of<br />

intussusception remains poorly understood. In<br />

90% of cases there is no detectable organic lesion<br />

acting as the lead point. 17 The high incidence<br />

during the first year of life appears to be related to<br />

the abundant lymphoid tissue of the terminal<br />

ileum at this age, with lymphoid tissue hypertrophy<br />

being detected in most surgical interventions,<br />

and often apparent on ultrasonography. However,<br />

some authors have suggested that these<br />

adenopathies are a consequence, not a cause, of<br />

the intussusception. 18<br />

As noted, the most commonly intussuscepted<br />

section is the terminal ileum, telescoped into the<br />

colon (i.e. ileocolic intussusception). The intussusception<br />

extends a variable distance along the<br />

colon, and may even reach the rectum, where it<br />

can be detected by rectal palpation. Occasionally,<br />

ileoileal intussusceptions are observed, extending<br />

to the ileocecal valve.<br />

In ileocolic intussusception, the intussusceptum is<br />

a section of the proximal part of the ileum, which<br />

telescopes into the adjacent distal colon, causing<br />

compression of the mesentery and strangulation of<br />

mesenteric blood vessels, with rapid edema of the<br />

intestinal wall, leading to impairment of venous<br />

return, edema of surrounding tissues, hemorrhage,<br />

blood infiltration and intestinal obstruction. The<br />

venous engorgement and ischemia lead to mucus<br />

secretion and bleeding, resulting in the classic<br />

‘currant jelly’ stool. If the venous obstruction and<br />

ischemia continue, the patient will develop<br />

gangrene, intestinal perforation due to necrosis of<br />

the intussusceptum and peritonitis.<br />

Secondary/organic intussusception<br />

With increasing age of the patient, it is increasingly<br />

likely that the intussusception will have an<br />

organic origin. About 5–10% of intussusceptions<br />

are caused by an organic lesion. Above the age of 4<br />

years, this percentage increases to 50%.<br />

The most frequent organic cause is Meckel’s diverticulum,<br />

followed by enteric duplications, intesti-<br />

nal polyps (whether isolated or forming part of<br />

Peutz–Jeghers syndrome), hemangiomas, traumatic<br />

hematomas and, less frequently,<br />

Henoch–Schönlein purpura, foreign bodies and<br />

trichobezoars, Crohn’s disease, cystic fibrosis,<br />

leukemias, lymphomas, and intestinal lymphosarcomas.<br />

11,12,19<br />

Postoperative intussusception may appear after<br />

any type of surgery (thoracic, abdominal, otorhinolaryngological,<br />

etc.), although it is most frequent<br />

after surgery to the digestive tract. Such intussusceptions<br />

make up about 1% of the total, and are<br />

related to postoperative motility alterations and<br />

paralytic ileus, or anesthetic medication. They<br />

typically present in the first 15–20 days after<br />

surgery and are often difficult to diagnose, since<br />

they are generally ileoileal or jejunoileal, and not<br />

revealed by contrast enema. They are rarely diagnosed<br />

in the immediate postoperative period,<br />

during which the signs and symptoms of intussusception<br />

are typically masked by postoperative pain<br />

and other consequences of the surgery. Treatment<br />

is surgical: usually surgical reduction is sufficient,<br />

without any need for intestinal resection. 20,21<br />

Clinical manifestations<br />

Intussusception typically presents in thriving and<br />

otherwise healthy nursing infants, and starts with<br />

a sudden crisis of crying and screaming, with<br />

knees drawn up to the abdomen. After a few<br />

minutes these responses generally subside, and<br />

the child appears normal, but the crises then recur<br />

at regular intervals of 10–20min, and may often be<br />

dramatic in that the child cannot be calmed.<br />

Sometimes the child has suffered a similar crisis<br />

during the preceding days or months. Vomiting of<br />

undigested food typically occurs a short time after<br />

onset of the pain crises; bilious vomiting generally<br />

occurs only if the disorder is left untreated for a<br />

long period. Stools of normal appearance may be<br />

passed (i.e. feces already present distal to the<br />

intussusception), but at later stages (generally not<br />

before 8–12h after onset of the pain crises) the<br />

patient will discharge the characteristic blood-andmucus<br />

clots known as ‘currant jelly’ stools.<br />

It should be stressed that the ‘typical’ symptom<br />

triad of abdominal pain, vomiting and blood in


stools is rarely seen early. In other words,<br />

Ombredanne’s criterion (i.e. intussusception=ileal<br />

signs+blood in stools), which was widely used in<br />

the past, is in fact valid only for the late course.<br />

Occasionally, other symptoms (such as fever, diarrhea<br />

and prostration) may appear. Diarrhea may<br />

suggest gastroenteritis and delay diagnosis, as may<br />

constipation, which occurs in up to 20% of cases. 22<br />

Repeated occurrence of the typical crises over a<br />

period of days or weeks may indicate chronic<br />

intussusception with spontaneous reduction,<br />

presenting at intervals as acute crises.<br />

Patients may show lethargy (i.e. apathy, sleepiness,<br />

prostration) during the early stages. Lethargy most<br />

commonly occurs between the pain crises, but in<br />

some cases may be the first symptom noted, before<br />

the pain crises, possibly suggesting encephalitis.<br />

Indeed, intussusception initially presenting as<br />

lethargy alone is typically problematic for diagnosis,<br />

since lethargy is normally associated with<br />

structural, toxic or metabolic disorders of the<br />

central nervous system; in such cases, central<br />

nervous system-related diagnostic procedures are<br />

typically applied before it becomes clear that the<br />

problem is in fact intussusception, with consequent<br />

diagnostic delay. 23,24 In this connection, it<br />

has been suggested that intussusception be<br />

included in mnemotechnics for diagnosis of<br />

patients with coma, with the letter ‘i’ being used to<br />

indicate ‘insulin’ and ‘intussusception’. 25 Various<br />

hypotheses have been put forward to explain the<br />

lethargy arising in association with intussusception,<br />

including secretion of endogenous opioids<br />

and cytokines during the pain crises, absorption of<br />

toxins into the bloodstream and association with<br />

rapid dehydration and shock. In almost all cases in<br />

which lethargy is observed, some other symptoms<br />

are present, such as vomiting, abdominal distension,<br />

or irritability. It may also appear in association<br />

with other neurological symptoms such as<br />

convulsions, coma, pinpoint pupils, opisthotonos<br />

and hypotonia.<br />

Occasionally, intussusception is not diagnosed<br />

until rectal bleeding occurs. This occurs most<br />

commonly in patients aged more than 1 year.<br />

If untreated, the patient’s condition is likely to<br />

worsen rapidly, with dehydration and possible<br />

shock. Early diagnosis is thus essential.<br />

Physical examination 721<br />

Some years ago we reviewed the symptoms<br />

observed in cases of intussusception treated at our<br />

hospital over the period 1969–84. 11 The most<br />

common symptoms were cramping abdominal<br />

pain (87% of patients), rectal bleeding (82%) and<br />

vomiting (78%) (Table 43.1).<br />

Physical examination<br />

In the early stages physical examination is in most<br />

cases normal, with the infant typically appearing<br />

well-fed and healthy. During pain crises, and if the<br />

crying permits, hyperperistaltic noise may be<br />

heard, with the abdomen tense and painful to<br />

palpation. Between the crises the right lower quadrant<br />

may appear strangely empty if the intussusception<br />

is ileocecal, with a mass palpable in practically<br />

any part of the abdomen; this mass is<br />

typically sausage-shaped and curved, owing to the<br />

traction exerted by the mesentery on the intestine.<br />

In rectal examination, the examining finger may<br />

appear covered with bloody mucus or blood, but<br />

direct contact between the examining finger and<br />

the intussuscepted mass is rare. Rectal hemorrhage,<br />

together with fever and tachycardia,<br />

become increasingly likely over time. In the past,<br />

when intussusceptions were typically diagnosed<br />

late, the intussusceptum was often detectable on<br />

rectal examination, and indeed there are<br />

Table 43.1 Signs and symptoms observed in<br />

intussusception patients seen at the Santiago de<br />

Compostela Clinical Hospital over the period<br />

1969–84<br />

Signs and symptoms Patients<br />

n %<br />

Cramping abdominal pain 61 87<br />

Rectal bleeding 58 82.8<br />

Vomiting 55 78.5<br />

Abdominal distension 30 42.8<br />

Abdominal mass 27 35.8<br />

Respiratory illness 16 22.8<br />

Lethargy 12 17<br />

Gastroenteritis 10 14.3<br />

Previous intussusception 2 3


722<br />

Intussusception<br />

descriptions of prolapse through the anus, with<br />

consequent ischemic damage (an extremely severe<br />

sign, generally associated with severe illness). If<br />

the intussusception has remained untreated for a<br />

long period, the patient may present with symptoms<br />

of hypovolemic shock.<br />

Diagnosis<br />

All pediatricians are well aware that persistent<br />

crying and abdominal pain in a nursing infant,<br />

particularly if associated with vomiting, needs to<br />

be carefully evaluated to confirm or rule out intussusception.<br />

Cramping pain with vomiting is sufficient<br />

grounds for continuous monitoring.<br />

Complementary examinations should follow a<br />

systematic order. Initially, simple abdominal radiography<br />

and ultrasonography should be<br />

performed, which together will reveal intussusception<br />

and possible repercussions for intestinal<br />

transit. In the absence of contraindications, hydrostatic<br />

enema or pneumoenema should then be<br />

performed, to confirm the diagnosis and, when<br />

appropriate, to achieve reduction.<br />

Simple abdominal radiography<br />

This will reveal about 50% of intussusceptions,<br />

allowing evaluation of the degree of intestinal<br />

obstruction and exclusion of perforation due to<br />

pneumoperitoneum. The radiographic signs are<br />

not in themselves fully diagnostic, and a normal<br />

radiograph does not allow intussusception to be<br />

ruled out; nevertheless, in my opinion it should be<br />

performed routinely.<br />

The most characteristic radiographic findings in<br />

ileocolic intussusception (Figure 43.1) are as<br />

follows:<br />

(1) Reduced or absent intestinal gas in the right<br />

quadrant;<br />

(2) Air–fluid levels;<br />

(3) Diffuse radiolucent image, generally located<br />

in the right hypochondrium;<br />

(4) Circle sign: two concentric circles and fat<br />

density, superimposed over the kidney to the<br />

right of the vertebral column;<br />

(5) Convex mass effect with interruption of air at<br />

the level of the transverse colon;<br />

Figure 43.1 Simple abdominal radiograph showing<br />

abnormal distribution of intestinal gas, with subhepatic<br />

mass.<br />

(6) Obstruction of the small intestine. 26–28<br />

Ultrasonography<br />

Since the first report of the diagnosis of intussusception<br />

in adult patients by ultrasonography in<br />

1977, this technique has become the imaging<br />

method of choice, and nowadays emergency ultrasonography<br />

will detect most cases of intussusception,<br />

particularly if the abdomen is not greatly<br />

distended by gas. 29<br />

There have been numerous descriptions of the<br />

ultrasonographic signs of intussusception, which<br />

habitually show a donut or ‘target’ image, with a<br />

hypoechoic peripheral ring representing the<br />

edematous walls of the intussusceptum, and a<br />

hyperechoic central zone made up of areas of<br />

compressed mucus. In longitudinal section the<br />

hypoechoic walls on both sides of the hyperechoic<br />

center show a tubular ‘pseudokidney’ or ‘sandwich’<br />

appearance 30 (Figure 43.2).


Figure 43.2 Ultrasonogram showing typical<br />

‘pseudokidney’ image in subhepatic location, indicative<br />

of intussusception.<br />

Ultrasonograms are not entirely pathognomonic<br />

for intussusception, but most published series<br />

indicate sensitivity and negative predictive value<br />

of close to 100%. 31,32<br />

Ultrasonography also offers a number of other<br />

important advantages: it is fast, cheap, noninvasive,<br />

innocuous (no ionizing radiation) and<br />

capable of detecting intussusceptions that are<br />

difficult to detect by conventional clinical examination<br />

and radiography, notably ileoileal and postsurgical<br />

intussusceptions. Ultrasonography facilitates<br />

assessment of reducibility and of the<br />

feasibility of non-surgical reduction, in view of the<br />

existing amount of intraperitoneal liquid, the<br />

thickness of the intestinal wall and the degree of<br />

distension and peristalsis of the intussusceptum.<br />

Doppler sonography (Figure 43.3) facilitates<br />

assessment of the degree of vascularization or<br />

ischemia of the intussusceptum, which is important<br />

for treatment selection. 33,34<br />

Ultrasonography may also detect pathological lead<br />

points such as Meckel’s diverticulum, lymphomas<br />

of the small intestine, or cysts. 35 In addition, it<br />

allows the examiner to rule out other causes of<br />

abdominal pain, such as appendicitis, ovarian<br />

pathology or anomalies of the urinary tract.<br />

Since the introduction and widespread use of<br />

hydrostatic reduction under ultrasonographic<br />

Diagnosis 723<br />

Figure 43.3 Doppler sonogram showing good<br />

vascularization of the intussusceptum, so that enema<br />

reduction is not contraindicated.<br />

guidance, reduction success rates of up to 95.5%<br />

have been reported, 36 with reduction confirmed by<br />

disappearance of the characteristic image, saline<br />

and air reflux through the ileocecal valve and<br />

distension of the ileum by the saline. The principal<br />

advantages are lack of exposure to ionizing<br />

radiation and absence of significant complications.<br />

Indeed, the only complication reported to date is<br />

perforation, easily recognized during reduction,<br />

and with fewer associated risks than escape of<br />

barium into the abdominal cavity. In short, hydrostatic<br />

reduction under ultrasound guidance is as<br />

effective as, or more effective than other reduction<br />

techniques, and has the obvious advantage of<br />

avoiding radiation exposure. 37<br />

Barium enema<br />

As a purely diagnostic method, barium enema is<br />

nowadays of little value. It may be useful for<br />

confirming the diagnosis in doubtful cases,<br />

showing a concave image at the head of the barium<br />

column corresponding to the apex of the intussusceptum;<br />

the image may be delimited by barium<br />

between the intussusceptum and the intussuscipiens,<br />

and is clearer when some of the barium is<br />

evacuated from the colon distal to the intussusception<br />

(Figure 40.4). However, possible organic<br />

causes of intussusception are rarely detected by<br />

this method.


724<br />

Intussusception<br />

Figure 43.4 Diagnostic barium contrast enema:<br />

radiograph showing non-passage of barium beyond the<br />

level of the hepatic angle, and with barium extending<br />

between intussusceptum and intussuscipiens.<br />

Treatment<br />

Medical treatment<br />

Once intussusception has been diagnosed, a nasogastric<br />

tube should be inserted, and peripheral<br />

endovenous fluid therapy should be started. Blood<br />

tests, including coagulation and electrolytes,<br />

should be done. Routine antibiotic treatment is not<br />

recommended, and should be reserved for complicated<br />

cases. Sedation and analgesia are recommended<br />

(with drugs including diazepam, midazolam,<br />

meperidine, thiopental sodium, ketamine,<br />

phenobarbital sodium and atropine) to avoid pain,<br />

reduce spasm, shorten the time required for reduction,<br />

and increase the likelihood of success.<br />

Glucagon is not currently used, in view of its<br />

doubtful efficacy. 38,39<br />

We consider non-surgical reduction to be<br />

contraindicated if the patient shows poor general<br />

status, with signs of shock, dehydration or peritoneal<br />

irritation, or clinical, radiographic or ultrasonographic<br />

signs of perforation or advanced<br />

intestinal obstruction, or when the patient is<br />

attended a long time after the onset of intussusception,<br />

since the success rate of hydrostatic<br />

reduction declines with increasing time since<br />

onset. In such cases, the aim should be to recover<br />

good general status and then perform emergency<br />

surgery.<br />

Once successful reduction has been achieved, the<br />

patient should be maintained on observation for<br />

24h with nothing per os, and discharged after reintroduction<br />

and confirmed tolerance of oral<br />

feeding.<br />

Hydrostatic reduction<br />

The publication of Ravitch’s series in 1948 led to<br />

the widespread adoption of hydrostatic barium<br />

enema as a standard treatment method. 4 As noted,<br />

hydrostatic reduction under ultrasonographic<br />

guidance offers high diagnostic sensitivity together<br />

with high treatment success rates.<br />

Various contrast materials are available, the most<br />

widely used being barium sulfate, dissolved in<br />

physiological saline to avoid water intoxication<br />

and hyponatremia, or alternatively non-barium<br />

hydrosoluble contrasts (which have fewer negative<br />

effects) (Figures 43.5 and 43.6).<br />

It is important to note that the column height of<br />

the enema should be between 1 and 1.2m never<br />

exceeding 1.5m. The anus should not be fully<br />

occluded, permitting reflux of contrast medium if<br />

excessive pressure develops. The abdomen<br />

should not be manipulated during reduction.<br />

Each reduction attempt should not exceed<br />

10min, and if no progress is made the attempt<br />

should be abandoned. If progress is detected,<br />

further attempts may be made, at intervals of<br />

5–15min.<br />

Pneumatic reduction<br />

Intussusception reduction using pressurized air<br />

under radiographic guidance was described by<br />

Fiorito and Recalde Cuesta, 5 although apparently<br />

techniques of this type were traditionally used in<br />

China. 40<br />

Within the colon, pressurized air ‘pushes’ the<br />

intussusceptum, and at the same time enters into


Figure 43.5 Barium contrast radiograph showing an<br />

ileocecal intussusception in the transverse colon.<br />

the space between the intussusceptum and the<br />

intussuscipiens, facilitating reduction (Figure<br />

43.7). The reduction can be guided either by<br />

contrast radiography or by ultrasonography. 7,41<br />

Various devices for the introduction of air have<br />

been described, all including some sort of pump,<br />

a manometer and an intracolic pressure regulator.<br />

Pressure can be controlled with a sphygmomanometer<br />

to control pressure, and any sort of<br />

expulsion pump can be used (Richardson’s bulb,<br />

syringe pump, etc.). It has been shown that CO2<br />

is more readily absorbed than air in the intestinal<br />

lumen, but reduction efficacy does not seem to be<br />

any higher. 42<br />

With the child in the prone position, an 18–20 Fr<br />

Foley catheter is inserted into the rectum, with an<br />

initial pressure of 60mmHg, gradually increased<br />

to no more than 80–100mmHg in infants and<br />

120mmHg in older children. Intestinal perforation<br />

is thought not to be a result of excessive<br />

pneumatic pressure, but of inappropriate application<br />

of this technique in cases in which the intestine<br />

is ischemic or necrotic.<br />

Treatment 725<br />

Figure 43.6 Barium contrast radiograph showing an<br />

ileocecal intussusception in the ileocecal valve.<br />

Figure 43.7 Air contrast radiography of an ileocecal<br />

intussusception extending as far as the hepatic angle. The<br />

intussusception is visible as a radiopaque mass that<br />

prevents the passage of air.


726<br />

Intussusception<br />

We currently prefer this technique to hydrostatic<br />

reduction, since it is easier to perform, and permits<br />

ready visualization of the intussusceptum, does<br />

not absorb body heat and facilitates confirmation<br />

of successful reduction (indicated by passage of air<br />

into the small intestine). Furthermore, if perforation<br />

occurs this is less problematic than in hydrostatic<br />

reduction with barium contrast, and is easier<br />

to treat, since the pneumoperitoneum can be<br />

drained immediately after insertion of a largegauge<br />

needle. 41,43,44<br />

The success rate with hydrostatic or pneumatic<br />

reduction is 75–95%, with perforation occurring in<br />

0.11–2% of cases. 6,7,38,39<br />

Surgical treatment<br />

Surgery is indicated only in very specific circumstances:<br />

(1) Whenever there are signs of shock or peritonitis;<br />

(2) In patients in whom hydrostatic or pneumatic<br />

reduction has not been fully successful;<br />

(3) In patients with poor general status, dehydration<br />

or peritoneal irritation;<br />

(4) In patients with intussusceptions of the sigmoid<br />

or rectum;<br />

(5) When there are clinical, radiographic or sonographic<br />

signs of perforation or advanced<br />

intestinal obstruction;<br />

(6) When the intussusception is not diagnosed<br />

until a long time after onset;<br />

(7) In cases of recurrent intussusception, with<br />

two or three non-surgical reductions previously<br />

per-formed.<br />

Successful reduction does not rule out the existence<br />

of organic pathology, so that any residual<br />

defect is an indication for laparotomy. Patients<br />

older than 2–3 years have been the object of some<br />

controversy, since the possibility that a lead point<br />

exists is much higher; in our series, 50% of children<br />

aged more than 2 years had organic causes,<br />

and some surgeons argue that in these patients<br />

exploratory laparotomy should be performed, even<br />

after successful non-surgical reduction.<br />

Surgery has certain advantages: any organic<br />

pathology will invariably be detected, the intussusception<br />

reduction will be complete and the<br />

recurrence rate is somewhat lower than with nonsurgical<br />

reduction. However, it also has a higher<br />

financial/resources cost, longer hospitalization<br />

time and a higher rate of complications (up to 17%<br />

in some series). 45<br />

Comfortable access can be achieved by a right<br />

transverse incision. The affected intestine can<br />

generally be readily exposed, enabling the intussusceptum<br />

to be gently massaged out of the intussuscipiens,<br />

using classic ‘milking’ movements, and<br />

not pulling the intussusceptum. Reduction in fact<br />

often tends to be difficult in surgically treated<br />

cases, and resections are required rather often; this<br />

probably reflects the high success rates obtained<br />

with pneumatic and hydrostatic reduction, and<br />

the fact that only problematic cases are treated<br />

surgically.<br />

Once complete reduction has been achieved, the<br />

viability of the intestine should be assessed, to<br />

identify possible regions of necrosis or doubtful<br />

viability, and to detect possible organic causes.<br />

After reduction, the cecal region and terminal<br />

ileum show a highly characteristic appearance,<br />

with thickening of the intestinal walls, and in<br />

some cases presence of a hard edematous plaque at<br />

the level of the terminal ileum; this should not be<br />

confused with an organic cause and does not<br />

require any sort of surgical response, which indeed<br />

would only cause complications. When intestinal<br />

resection is necessary, this should be as conservative<br />

as possible, aiming to maintain the ileocecal<br />

valve. Appendectomy should be performed<br />

routinely if the edema of the ileocecal region<br />

permits, although some authors have questioned<br />

its utility. Surgical treatment does not exclude the<br />

possibility of recurrence, which in one study was<br />

reported in 3.2% of cases. 46<br />

Intestinal fixation techniques (e.g. fixation of the<br />

cecum, suture of the terminal ileum to the ascending<br />

colon, Noble’s plication) were in the past<br />

widely used to prevent recurrence, but are now<br />

generally regarded as being ineffective and unnecessary.<br />

Recurrent intussusception requires a personalized<br />

treatment program: characteristically, surgery is


not required in patients aged less than 2 years,<br />

when they show a second or third crisis over a<br />

short period of time. In older patients, radiographic<br />

and sonographic findings should be<br />

REFERENCES<br />

1. Barbette P. Oeuvres chirugiques at anatomiques. Geneva:<br />

Francois Miege, 1674: 522.<br />

2. Hutchinson J. A successful case of abdominal section<br />

for intussusception. Proc R Med Chir Soc 1873; 7:<br />

195–198.<br />

3. Hirschsprung H. Et Tilfaetde af suakut<br />

Tarminvagination. Hospitals-Tidende 1876; 3: 321–327.<br />

4. Ravitch MM, McClure RM. Reduction of intussusception<br />

by barium enema: a clinical and experimental<br />

study. Ann Surg 1948; 728: 904.<br />

5. Fiorito ES, Recalde Cuesta LA. Diagnosis and treatment<br />

of acute intestinal intussusception with controlled<br />

insufflation of air. Pediatrics 1959; 24: 241–244.<br />

6. YaXiong She. Traitement de I'invagination intestinale<br />

avec regard spécial sur la reduction par insufflation du<br />

clan. Expérience de 5110 cases. Chir Pédiatr 1982; 23:<br />

373–378.<br />

7. Guo J, Ma X, Zhou Q. Results of air pressure enema<br />

reduction of intussusception: 6396 cases in 13 years. J<br />

Pediatr Surg 1986; 21: 1201–1203.<br />

8. Stringer MD, Pablot SM, Brereton RJ. Paediatric intussusception.<br />

Br J Surg 1992; 79: 867–876.<br />

9. Smith IM. Incidence of intussusception and congenital<br />

hypertrophic <strong>pylori</strong>c stenosis in Edinburgh children.<br />

BMJ 1960; 1: 551–552.<br />

10. Shah BR, Laude TA. Intussusception. In Shah BR,<br />

Laude TA, eds. Atlas of Pediatric Clinical Diagnosis.<br />

Philadelphia: W.B. Saunders, 2000: 338–341.<br />

11. Bautista Casasnovas A, Varela Cives R, Nieto Vázquez<br />

B et al. Evaluación del tratamiento medico y quirúrgico<br />

de la invaginación intestinal en el niño. An Esp Pediatr<br />

1988; 29: 279–283.<br />

12. Eizaguirre I, Morras I, Jimenez J, Tovar J. Invaginación<br />

intestinal. Revisión de 110 casos. Rev Esp Pediatr 1985;<br />

41: 339–344.<br />

13. Bruce J, Huh YS, Cooney DR et al. Intussusception:<br />

evolution of current management. J Pediatr<br />

Gastroenterol Nutr 1987; 6: 663–674.<br />

14. Simonsen L, Morens D, Elixhauser A et al. Effect of<br />

rotavirus vaccination programme on trends in admission<br />

of infants to hospital for intussusception. Lancet<br />

2001; 358: 1224–1229.<br />

15. DiFiore JW. Intussusception. Semin Pediatr Surg 1999;<br />

84: 214–220.<br />

16. Meier DE, Coln CE, Rescorla FJ et al. Intussusception in<br />

children: international perspective. World J Surg 1996;<br />

20: 1035–1040.<br />

17. Ong NT, Beasley SW. The leadpoint in intussusception.<br />

J Pediatr Surg 1990; 25: 640–643.<br />

18. Luks FI, Yazbeck S, Brandt ML, Desjardins JG. Fievre<br />

transitoire associée a la réduction de I'invagination<br />

intestinale. Chir Pédiatr 1990; 37: 157–159.<br />

References 727<br />

assessed, since the higher frequency of organic<br />

causes at these ages obliges us to consider the need<br />

for exploratory laparotomy, which of course will<br />

not always detect any organic lesion.<br />

19. Ducharme JC, Perreault G, Cyr R, Medoux L. L’ invagination<br />

intestinale, 188 malades traites au course d’ une<br />

periode de 22 ans. Chir Pediatr 1982; 23: 23–27.<br />

20. de Vries S, Sleeboom C, Aronson DC. Postoperative<br />

intussusception in children. Br J Surg 1999; 86: 81–83.<br />

21. Linke F, Eble F, Berger S. Postoperative intussusception<br />

in childhood. Pediatr Surg Int 1998; 14: 175–177.<br />

22. Yazbeck S. Intestinal obstruction in infancy and childhood.<br />

In Roy CC, Silverman MA, Alagille D, eds.<br />

Pediatric Clinical Gastroenterology, 4th edn. Boston:<br />

Mosby, 1993: 106–110.<br />

23. Singer J. Altered consciousness as an early manifestation<br />

of intussusception. Pediatrics 1979; 64: 93–95.<br />

24. Villanueva Jeremias A, Bautista Casasnovas A. Niño de<br />

11 meses con letargia. Casos Clínicos Pediatría 2002; 1:<br />

10–15.<br />

25. McCabe JB, Singer JL, Love T, Roth R. A supplement to<br />

the mnemonic for coma. Pediatr Emerg Care 1987; 3:<br />

118–119.<br />

26. Ratcliffe JF, Fong S, Cheong I, O'Conell PO. The plain<br />

abdominal film in intussusception: the accuracy and<br />

incidence of radiographic signs. Pediatr Radiol 1992; 22:<br />

110–111.<br />

27. Bisset GS, Kirks DR. Intussusception in infants and<br />

children: diagnosis and treatment. Radiology 1998; 168:<br />

141–145.<br />

28. Daneman A, Alton DJ. Intussusception. Issues and<br />

controversies related to diagnosis and reduction. Radiol<br />

Clin North Am 1996; 34: 743–756.<br />

29. Weissberg DL, Scheible W, Leopold GR.<br />

Ultrasonographic appearance of adult intussusception.<br />

Radiology 1977; 124: 791–792.<br />

30. Bhisitkul DM, Listernick R, Shikolnik A et al. Clinical<br />

application of ultrasonography in the diagnosis of intussusception.<br />

J Pediatr 1992; 727: 182–186.<br />

31. Woo SK, Kim JS, Suh SI et al. Childhood intussusception:<br />

US-guided hydrostatic reduction. Radiology 1992;<br />

782: 77–80.<br />

32. Yoon CH, Kim HJ, Goo HW. Intussusception in children:<br />

US-guided pneumatic reduction initial experience.<br />

Radiology 2001; 218: 85–88.<br />

33. Mirilas P, Koumanidou C, Vakaki M et al. Sonographic<br />

features indicative of hydrostatic reducibility of intestinal<br />

intussusception in infancy and early childhood. Eur<br />

Radiol 2001; 11: 2576–2580.<br />

34. Britton I, Wilkinson AG. Ultrasound features of intussusception<br />

predicting outcome of air enema. Pediatr<br />

Radiol 1999; 29: 705–710.<br />

35. Navarro O, Dugougeat F, Kornecki A et al. The impact of<br />

imaging in the management of intussusception owing to<br />

pathologic lead points in children. A review of 43 cases.<br />

Pediatr Radiol 2000; 30: 594–603.


728<br />

Intussusception<br />

36. Wang GD, Liu SI. Enema reduction of intussusception<br />

by hydrostatic pressure under ultrasound guidance: a<br />

report of 377 cases. J Pediatr Surg 1988; 23: 814–818.<br />

37. Shehata S, El Kholi N, Sultan A, El Sahwi E.<br />

Hydrostatic reduction of intussusception: barium, air, or<br />

saline? Pediatr Surg Int 2000; 16: 380–382.<br />

38. Katz ME, Kolm P. Intussusception reduction 1991: an<br />

international survey of pediatric radiologists. Pediatr<br />

Radiol 1992; 22: 318–322.<br />

39. Meyer IS. The current radiologic management of intussusception:<br />

a survey and review. Pediatr Radiol 1992;<br />

22: 323–325.<br />

40. Jinzhe Z, Yenxia W, Linchi W. Rectal inflation reduction<br />

of intussusception in infants. J Pediatr Surg 1986; 21:<br />

30–32.<br />

41. Gu L, Zhu H, Wang S et al. Sonographic guidance of air<br />

enema for intussusception reduction in children.<br />

Pediatr Radiol 2000; 30: 339–342.<br />

42. Palder SB, Ein SH, Stringer DA, Alton D.<br />

Intussusception: barium or air? J Pediatr Surg 1991; 26:<br />

271–275.<br />

43. Heenan SD, Kyriou J, Fitzgerald M, Adam EJ. Effective<br />

dose at pneumatic reduction of paediatric intussusception.<br />

Clin Radiol 2000; 55: 811–816.<br />

44. Stringer DA, Ein SH. Pneumatic reduction: advantages,<br />

risks and indications. Pediatr Radiol 1990; 20: 475–477.<br />

45. Leonidas JC. Treatment of intussusception with small<br />

bowel obstruction: application of decision analysis. Am<br />

J Reotgenol 1985; 145: 665–669.<br />

46. Aubrespy P, Derlon S, Alessandrini P et al. Invagination<br />

intestinale aigue du neurrison et de l' enfant. Analyse<br />

de 125 observations traitées chirurgicalement. Chir<br />

Pédiatr 1983; 24: 392–395.


44<br />

Introduction<br />

Meckel’s diverticulum<br />

Richard G Azizkhan<br />

Meckel’s diverticulum is the most common<br />

congenital anomaly of the gastrointestinal tract<br />

and the most common cause of serious gastrointestinal<br />

bleeding in children. Anatomically, it is<br />

an outpouching on the antimesenteric border of<br />

the small bowel (Figure 44.1). Although first<br />

reported by Fabricus Hildanus in 1598, this abnormality<br />

derives its name from Johann Friedrich<br />

Meckel, who in 1809 detailed its embryological<br />

origin and identified it as a potential cause of<br />

disease. 1,2 Reports estimate that Meckel’s diverticulum<br />

affects 2% of the general population.<br />

However, it is usually clinically silent, identified<br />

only as an incidental finding during laparotomy,<br />

laparoscopy, or at autopsy. Estimates of the life-<br />

Figure 44.1 Operative view of a Meckel’s diverticulum.<br />

The distal tip had heterotopic gastric mucosa.<br />

time risk of complications occurring range from 4<br />

to 6%. 3–5 These complications are usually due to<br />

ectopic tissue within the diverticulum or to<br />

omphalomesenteric or mesodiverticular bands.<br />

The clinical presentation of Meckel’s diverticulum<br />

is diverse. Symptoms often mimic those associated<br />

with other more common abdominal disorders,<br />

making preoperative diagnosis quite difficult. This<br />

chapter focuses on the major complications of this<br />

anomaly, describing its pathogenesis, diagnosis<br />

and management. Relevant embryology, histology<br />

and epidemiology will set the stage for this discussion.<br />

Pathoembryology<br />

Meckel’s diverticulum is one of a spectrum of<br />

congenital anomalies of the midgut that are embryological<br />

remnants of the vitelline (omphalomesenteric)<br />

duct. During week 3 of gestation, the yolk sac<br />

communicates with the gut through this duct.<br />

Between weeks 5–9 of gestation, the duct is<br />

normally obliterated, as the functioning placenta<br />

replaces the yolk sac as the source of fetal nourishment.<br />

Failure of this normal obliteration<br />

process can result in a number of developmental<br />

anomalies, of which Meckel’s diverticulum is the<br />

most common. The diverticulum may remain<br />

completely attached to the abdominal wall at the<br />

umbilicus, may remain attached as a fibrous cord<br />

as the distal portion of the vitelline duct involutes,<br />

or may remain unattached and free within the<br />

peritoneal cavity. The last presentation is the most<br />

common, occurring in 74% of cases. 6 Other anomalies<br />

related to the disturbance of vitelline duct<br />

involution include persistence of a patent<br />

omphalomesenteric duct (Figure 44.2) or the<br />

development of an omphalomesenteric sinus or<br />

cyst. Such a cyst may form when both proximal<br />

729


730<br />

Meckel’s diverticulum<br />

Figure 44.2 Patent omphalomesenteric duct in a<br />

neonate. The mucosa can be visualized in the<br />

transected umbilical cord. The omphalomesenteric duct<br />

communicates from the distal ileum to the umbilicus.<br />

Figure 44.3 Drawing depicting an omphalomesenteric<br />

cyst attached to the umbilical region as well as to a<br />

fibrous band to the terminal ileum.<br />

and distal obliteration occurs while the central<br />

portion of the duct remains patent (Figure 44.3).<br />

The blood supply to Meckel’s diverticulum is<br />

derived from the paired vitelline arteries. The left<br />

vitelline artery involutes, whereas the right<br />

persists as the superior mesenteric artery. A<br />

remnant of the primitive right vitelline artery<br />

arises directly from the mesentery to supply the<br />

diverticulum. The blood supply is anatomically<br />

unique in that the vessels to the diverticulum pass<br />

over the serosa of the ileum, terminating on the<br />

antimesenteric rather than the mesenteric side of<br />

the bowel. Although these vessels usually terminate<br />

at the tip of the diverticulum, they may<br />

continue on to the abdominal wall. They may also<br />

persist as a fibrous cord connecting the ileum to<br />

the umbilicus after obliteration of the diverticulum.<br />

The tip of the diverticulum may remain<br />

attached to the base of the mesentery by a fibrous<br />

remnant of the vitelline vessels, creating a mesodiverticular<br />

band through which loops of small<br />

bowel may pass and become incarcerated.<br />

Histology<br />

Meckel’s diverticulum is usually located within<br />

100cm (3 feet) of the ileocecal junction on the<br />

antimesenteric border of the ileum. It is a true<br />

diverticulum, containing all layers of the intestinal<br />

wall. Heterotopic mucosa is found in up to<br />

55–60% of Meckel’s diverticula 7–9 and is found<br />

predominantly in symptomatic patients. 5,10–12<br />

Gastric and pancreatic tissue predominate, with<br />

corresponding incidences of 60–90% and<br />

5–16% 5,10–13 (Figure 44.4); both ectopic tissue<br />

types may be found together within the diverticulum.<br />

Although colonic, duodenal, jejunal, hepatic<br />

and endometrial ectopic tissues have been<br />

found, 11,14–17 these are rare and are not generally<br />

associated with complications.<br />

Epidemiology<br />

Of symptomatic Meckel’s diverticula, 50–60%<br />

occur in children younger than 2 years of age. A<br />

significant increase in the 1–2% incidence in the<br />

general population is seen in association with a<br />

number of other congenital anomalies. In a series<br />

of nearly 6000 pediatric autopsies, authors<br />

reported significant increases in the incidence of<br />

Meckel’s diverticulum in children born with a<br />

cleft palate, bicornuate uterus, annular pancreas,<br />

esophageal or anorectal atresia, or gross malformation<br />

of the central nervous or cardiovascular<br />

systems. 18 Meckel’s diverticulum is also more<br />

common in patients with Crohn’s disease 19 and in<br />

patients with omphalocele, approximately 25% of<br />

whom exhibit some form of omphalomesenteric


Figure 44.4 Histology of a Meckel’s diverticulum<br />

demonstrating gastric and ileal mucosa. The deep cleft<br />

is an ulcer separating the two types of mucosa.<br />

remnant. 20 Although rare, carcinoid tumors have<br />

been found to arise from the diverticulum. 21–23<br />

Other tumors have also been reported, but these<br />

have occurred primarily in adult patients. 7<br />

Clinical presentations<br />

Predisposing factors<br />

While heterotopic tissue within the diverticula is<br />

the most significant predisposing factor to complications,<br />

several other factors also appear to be<br />

associated with an increased likelihood of complications.<br />

The reported predominance of symptomatic<br />

Meckel’s diverticulum in males (ratios<br />

ranging from 2:1 to 5:1) indicates that gender may<br />

play a role. 11,13 Studies have also shown that diverticular<br />

length 11 and base diameter 24 are predispos-<br />

Clinical presentations 731<br />

ing factors. Long, narrow-based diverticula are<br />

thought to be more prone to obstruction or inflammation,<br />

whereas short, large-based diverticula are<br />

subject to foreign body entrapment. Whether or<br />

not age can be correlated with the incidence of<br />

complications is currently controversial. Although<br />

the risk of complications has historically been<br />

thought to decline with age, 3,11,24 two relatively<br />

recent studies have demonstrated an even age<br />

distribution in patients with complications. 13,26<br />

Complications<br />

The most common complications are gastrointestinal<br />

bleeding, intestinal obstruction and inflammation.<br />

Clinical presentation is generally associated<br />

with the presence of ectopic gastric mucosa within<br />

the diverticulum or fixation of omphalomesenteric<br />

duct remnants to the abdominal wall. The ectopic<br />

mucosa may cause ulceration, which can result in<br />

inflammation, bleeding, or perforation. Fixation of<br />

omphalomesenteric duct remnants to the abdominal<br />

wall may result in intestinal obstruction from<br />

torsion of small-bowel loops around the point of<br />

fixation (Figure 44.5). The Meckel’s diverticulum<br />

may be recognized on preoperative studies or at<br />

operation, or it may be found unexpectedly in a<br />

resected specimen. The likelihood of the presence<br />

of heterotopic mucosa in surgically resected<br />

Meckel’s diverticula depends on the clinical<br />

presentation. It is found in approximately 50% of<br />

all symptomatic patients who have gastrointestinal<br />

bleeding, obstruction or inflammation (diverticulitis).<br />

Gastrointestinal bleeding<br />

Gastrointestinal bleeding occurs in 22% of all children<br />

with Meckel’s diverticula and 40–60% of<br />

symptomatic children. 12,26 It is the most common<br />

presenting symptom in children younger than age<br />

4, and the mean age of presentation is 2.8 years. 12<br />

Painless rectal bleeding is the usual manifestation.<br />

Although the bleeding may be significant, it is<br />

often episodic. The color of blood may vary from<br />

bright red to dark red or maroon to black and tarry,<br />

and may even appear as the ‘currant jelly stool’<br />

typically associated with intussusception. 26<br />

Bleeding is sometimes massive, transfusions are<br />

often required and life-threatening hemorrhage


732<br />

Meckel’s diverticulum<br />

(a) (b)<br />

Figure 44.5 (a) Drawing depicting a segmental ileal volvulus around an omphalomesenteric band.<br />

(b) Intraoperative photograph showing a segmental ileal volvulus around a Meckel’s diverticulum and<br />

omphalomesenteric band.<br />

occasionally occurs. This requires rapid resuscitation<br />

and stabilization and early diagnosis and<br />

treatment.<br />

Bleeding is thought to occur as a result of peptic<br />

ulceration caused by secretion of hydrochloric<br />

acid from parietal cells within the ectopic gastric<br />

mucosa located in the diverticulum. In patients<br />

who show bleeding only, the prevalence of heterotopic<br />

gastric mucosa approaches 100%. 7 Unlike<br />

the stomach, the ileum cannot buffer the resulting<br />

low pH and is thus more prone to ulceration. The<br />

ulceration may be small, visible only to the pathologist.<br />

It is generally located at the base of the diverticulum,<br />

at the junction of ectopic gastric mucosa<br />

and normal ileal mucosa. It may, however, also be<br />

found within the ectopic gastric mucosa itself or<br />

on the mesenteric side of the ileum, opposite the<br />

diverticulum (Figure 44.6). Ectopic gastric mucosa<br />

and ulceration are identified in about 95%<br />

and 98%, respectively, in specimens resected for<br />

bleeding. 5<br />

Although <strong>Helicobacter</strong> <strong>pylori</strong> plays a major role in<br />

ulcerogenesis elsewhere, it does not appear to be a<br />

significant factor in the pathogenesis of bleeding<br />

and inflammatory complications from the ectopic<br />

gastric mucosa in Meckel’s diverticula, and has<br />

only rarely been isolated from specimens. 27–29<br />

Differential diagnosis includes inflammatory<br />

bowel disease, intestinal polyps, duplications and<br />

arteriovenous malformations. Peptic ulcer disease<br />

and variceal bleeding may also present in a similar<br />

fashion, but can be distinguished through bloody<br />

nasogastric aspirates or with upper endoscopy.<br />

Occult bleeding with anemia is an infrequent<br />

presentation of Meckel’s diverticulum.<br />

Intestinal obstruction<br />

Bowel obstruction in any previously healthy child<br />

merits a high index of suspicion for Meckel’s<br />

diverticulum. When this anomaly is present, the<br />

obstruction is usually due to intussusception<br />

(46%) or volvulus (24%). 7 In children with intussusception,<br />

Meckel’s diverticulum often serves as<br />

the lead point of the intussusception (Figure 44.7).<br />

The clinical presentation of intussusception<br />

includes vomiting, intermittent abdominal pain,<br />

bloody stools, a palpable lower abdominal mass<br />

and eventual progression to dehydration and


Figure 44.6 Drawing demonstrating two ileal ulcers<br />

on the mesenteric side of the ileum which may be seen<br />

in patients with bleeding related to heterotopic gastric<br />

mucosa in the Meckel’s diverticulum.<br />

lethargy. 7 Ultrasonography, as well as pneumatic<br />

or barium enema are useful in establishing the<br />

diagnosis of intussusception. Rarely, complete<br />

reduction is obtained with enema and if Meckel’s<br />

diverticulum is visualized by imaging, an elective<br />

diverticulectomy can be performed. Most patients,<br />

however, have incomplete reduction of their<br />

ileoileal/ileocolonic intussusception, and require<br />

surgery to reduce the intussusception and resect<br />

the Meckel’s diverticulum.<br />

Volvulus can occur through several mechanisms.<br />

It is usually an acute event, and if allowed to<br />

progress, may result in strangulation of the<br />

involved bowel. Vitelline remnants or the<br />

omphalomesenteric duct may attach to the abdominal<br />

wall and form an anchor around which the<br />

bowel twists. The intestine may herniate under<br />

mesodiverticular bands, kinking or obstructing<br />

(Figure 44.8). Rarely, a long diverticulum twists on<br />

itself (axial torsion), causing obstruction. Also<br />

rare, intestinal obstruction may result from incarceration<br />

of a Meckel’s diverticulum within a<br />

hernia sac (Littre’s hernia). In neonates, an<br />

inverted Meckel’s diverticulum has been associated<br />

with intestinal obstruction and microcolon. 30<br />

Clinical presentations 733<br />

Figure 44.7 Drawing demonstrating an ileo-ileal<br />

intussusception caused by a Meckel’s diverticulum.<br />

Resuscitation for bowel obstruction consists of<br />

intravenous hydration, correction of electrolytic<br />

abnormalities, administration of broad-spectrum<br />

antibiotics, intestinal decompression and definitive<br />

operation.<br />

Inflammation<br />

In contrast to bleeding and intestinal obstruction,<br />

diverticulitis occurs more commonly in adolescents<br />

and adults. It is generally indistinguishable<br />

from appendicitis and, as with appendicitis, failure<br />

to diagnose the diverticulitis promptly may<br />

lead to perforation, peritonitis and death. The role<br />

of the ectopic mucosa in this process is suggested<br />

by its high incidence in resected inflamed mucosa.<br />

Thus, when appendicitis is suspected but not<br />

found at operation, it is critical to inspect the distal<br />

ileum for a symptomatic Meckel’s diverticulum.<br />

Other clinical presentations<br />

Inflammation and perforation of a Meckel’s diverticulum<br />

by a foreign body in the gastrointestinal<br />

tract has been reported. 12,16 Although rarely seen


734<br />

Meckel’s diverticulum<br />

Figure 44.8 Herniation and obstruction of small<br />

intestine through a Meckel’s band adhering to the base<br />

of the ileal mesentery.<br />

in pediatric patients, malignancies, 9 stone formation<br />

within the diverticulum, and even parasitic<br />

infections have also been documented. 31,32<br />

Additionally, a recent report described the atypical<br />

presentation of recurrent urinary tract infections<br />

caused by an infected Meckel’s diverticulum that<br />

displaced the urinary bladder. 33<br />

Diagnostic evaluation<br />

Abdominal scintigraphy (Meckel’s scan) using<br />

Tc-99m pertechnetate is a well-established diagnostic<br />

technique to evaluate children with lower<br />

gastrointestinal bleeding and is considered the<br />

diagnostic procedure of choice when there is a<br />

suspicion of Meckel’s diverticulum (Figure 44.9).<br />

Reported results of a 10-year pediatric review of<br />

this type of scan revealed a sensitivity for detection<br />

of Meckel’s diverticulum of 85%, a specificity<br />

of 95% and accuracy of 90%. False-negative results<br />

occurred in 1.7% of cases, and false-positive<br />

results in only 0.05% of cases. 34<br />

The high affinity of Tc-99m pertechnetate for<br />

gastric mucosa is what makes this radiopharmaceutical<br />

such a valuable tool. Tc-99m pertechnetate<br />

is taken up and secreted by the tubular glands<br />

of the gastric mucosa. The remainder of the<br />

isotope is concentrated within the urinary bladder.<br />

Sequential 1-min anterior abdominal images are<br />

obtained for 30–60min. A positive scan shows<br />

abnormal uptake of isotope outside the stomach<br />

and urinary bladder. 35<br />

False-positive results have been reported with<br />

intestinal duplications, certain genitourinary<br />

anomalies, Barrett’s esophagus, ulcers, inflammatory<br />

bowel disease, intussusception, bowel<br />

obstruction, neoplasms and vascular lesions such<br />

as hemangiomas and arteriovenous malformations;<br />

these results are caused by non-specific<br />

accumulation of the isotope. False-negative Tc-<br />

99m pertechnetate scintigraphy may result from a<br />

small diverticulum or hemorrhage washing out the<br />

isotope. Also, residual contrast in the bowel from<br />

previous barium studies may hinder its detection,<br />

or tracer in the urinary bladder may obscure visualization<br />

of heterotopic gastric mucosa if it is<br />

located near the bladder. 35 Several pharmacological<br />

agents, including pentagastrin, histamine-2<br />

blockers (e.g. cimetidine), and glucagon have been<br />

used to enhance the diagnostic accuracy of the<br />

scan. Pentagastrin stimulates gastric mucosal<br />

uptake, histamine-2 blockers inhibit excretion of<br />

the isotope once it is taken up and glucagon<br />

inhibits peristalsis, thereby increasing retention of<br />

the isotope in the diverticulum. While some<br />

authors maintain that pharmacological enhancement<br />

is necessary only when results of a nonpharmacologically<br />

enhanced study are inconclusive,<br />

35 I feel that enhancement reduces the need<br />

for repeated radionuclide studies and therefore use<br />

it routinely. Fasting, nasogastric suctioning, and<br />

bladder catheterization may also increase the diagnostic<br />

yield of the scan.<br />

Although Tc-99m pertechnetate scintigraphy is the<br />

diagnostic procedure of choice, there are limitations<br />

to its use in clinical decision-making.<br />

According to a recent study, 36 in patients with<br />

gastrointestinal bleeding and a serum hemoglobin<br />

level of less than 11g/dl, the Meckel’s scan has a<br />

relatively low negative predictive value (0.74).<br />

Authors suggest that replacing the scan with early<br />

operative evaluation may be cost-effective,


Figure 44.9 Positive Tc-99m pertechnetate radionuclide scan at 60min demonstrating<br />

abnormal uptake of the radionuclide in the lower abdomen in a child with a Meckel’s<br />

diverticulum. Note the normal gastric uptake in the upper abdomen.<br />

especially in patients in whom there is a high<br />

index of suspicion for a bleeding Meckel’s diverticulum.<br />

The known advantages of minimally<br />

invasive laparoscopy have led others to advocate<br />

this operative approach as a means of identifying<br />

the presence of a bleeding Meckel’s diverticulum.<br />

They have suggested that it may replace the<br />

Meckel’s scan in the diagnostic evaluation of pediatric<br />

patients with clinically significant gastrointestinal<br />

bleeding and high suspicion for this<br />

disease process. 37,38<br />

Visceral angiography is rarely needed, but is occasionally<br />

useful as a diagnostic tool in the actively<br />

bleeding patient (more than 0.5–1ml/min) when<br />

the Meckel’s radionuclide scan is negative. In this<br />

instance, the angiogram may show extravasation of<br />

contrast from the ileocolic branch of the superior<br />

mesenteric artery. A superselective superior<br />

mesenteric artery angiogram may show a nonbranching,<br />

elongated artery originating from the<br />

ileal artery and a group of dilated tortuous vessels<br />

at the distal portion of this artery in patients with<br />

Meckel’s diverticulum. 39 This technique allows<br />

visualization of Meckel’s diverticulum, even in the<br />

absence of bleeding.<br />

Management<br />

Management 735<br />

Management of the asymptomatic patient with an<br />

incidentally discovered Meckel’s diverticulum has<br />

long been controversial. The debate focuses on the<br />

probability of the diverticulum becoming symptomatic<br />

in the future as opposed to the likelihood of<br />

complications associated with resection. To date,<br />

there are no definitive intraoperative anatomic or<br />

biological features that enable the surgeon to<br />

predict the probability of future complications.<br />

While predisposing factors for symptomatic<br />

Meckel’s diverticula have been identified, these<br />

factors are only of limited value to the surgeon in<br />

deciding whether or not to resect an asymptomatic<br />

diverticulum.<br />

In some clinical situations, there is a degree of<br />

consensus that helps in making an intraoperative<br />

judgment. Resection is clearly indicated in<br />

patients with a long, narrow-based Meckel’s diverticulum<br />

or in those in whom the diverticulum is<br />

palpably thickened at its terminal portion; in<br />

patients who have vitellointestinal remnants or<br />

abdominal wall attachments; and in patients with<br />

a history of unexplained abdominal pain. These


736<br />

Meckel’s diverticulum<br />

Figure 44.10 Drawing demonstrating the concept of segmental ileal resection and anastomosis.<br />

factors all increase the likelihood of bleeding,<br />

obstruction, or diverticulitis. Because Meckel’s<br />

diverticulum can become symptomatic at any age,<br />

the age of the patient is not the primary factor in the<br />

decision to resect the asymptomatic diverticulum.<br />

In general, this decision must be made after careful<br />

consideration of the risks and benefits for each<br />

patient. Owing to the risk of suture line disruption,<br />

an elective diverticulectomy is contraindicated if a<br />

Meckel’s diverticulum is identified during repair of<br />

gastroschisis. 7<br />

Symptomatic Meckel’s diverticulum must be<br />

resected, and resection should be preceded by<br />

prompt and aggressive fluid resuscitation and blood<br />

transfusion when necessary. In patients with intestinal<br />

obstruction and possible intestinal ischemia, the<br />

patient’s physiology should be stabilized to the<br />

degree possible prior to operation. When a laparotomy<br />

is performed, the diverticulum is generally<br />

approached through a transverse right lower quadrant<br />

incision. Visual inspection and palpation to<br />

identify the location of ectopic gastric mucosa and<br />

ulceration is critical. If one is confident that the<br />

ulceration is confined to the diverticulum, simple<br />

diverticulectomy may be performed with either<br />

sutures or a stapling device. To prevent luminal<br />

narrowing, especially with broader-based diverticula,<br />

the defect may be closed transversely in a<br />

Heineke–Mikulicz fashion. If the base of the diverticulum<br />

is too broad, or if ulceration is identified on<br />

the mesenteric side of the ileum opposite the diverticulum,<br />

segmental resection of the ileum containing<br />

the diverticulum with an end-to-end anastomosis<br />

is preferred (Figure 44.10). Care must be taken<br />

to separately ligate the blood supply to the Meckel’s<br />

diverticulum. When a right lower quadrant incision<br />

is used, an appendectomy should be concomitantly<br />

performed to prevent future diagnostic dilemmas.<br />

Laparoscopic resection performed either intracorporeally<br />

or extracorporeally has increasingly been<br />

reported. 40–42 With intracorporeal resection, the<br />

diverticulum is identified and grasped, and a diverticulectomy<br />

is performed using a stapling device if<br />

the diverticulum is deemed anatomically suitable<br />

for such an approach (Figure 44.11). The disadvantage<br />

of this technique with a bleeding Meckel’s<br />

diverticulum is the inability to exclude ulceration<br />

on the mesenteric side of the ileum opposite the<br />

diverticulum, which could result in continued<br />

bleeding after resection. Also, it is technically more<br />

difficult to perform a segmental small-bowel resection<br />

if indicated. With extracorporeal resection, the


Figure 44.11 Laparoscopic stapling and removal of a Meckel’s diverticulum.<br />

diverticulum is identified, grasped and brought out<br />

through the umbilical or other trocar site. Once the<br />

diverticulectomy or segmental small bowl resection<br />

is performed, the bowel is returned to the peritoneal<br />

cavity. The advantage of this technique is that it<br />

allows the surgeon to inspect and palpate the bowel,<br />

and to perform an intestinal anastomosis, easily.<br />

One limitation of laparoscopic resection is that it<br />

does not allow for the management of a giant<br />

Meckel’s diverticulum, which may be difficult to<br />

resect through small laparoscopic incisions.<br />

Although theoretical advantages of a laparoscopic<br />

approach include improved cosmesis and less postoperative<br />

pain, prospective studies that demonstrate<br />

these advantages are warranted.<br />

Outcomes<br />

Since the 1980s, incidental diverticulectomy for<br />

patients with asymptomatic Meckel’s diverticulum<br />

has been associated with morbidity and mortality<br />

rates ranging from 0–9% to 0–1%, respectively.<br />

13,24,26,43 In a comprehensive Mayo Clinic<br />

study including both children and adults, the<br />

overall rate of morbidity of incidental removal of<br />

Outcomes 737<br />

Meckel’s diverticulum was 2%, while the postoperative<br />

mortality rate was 1%. The risk of developing<br />

long-term complications by 20 years after an incidental<br />

diverticulectomy was only 2%. In view of the<br />

4–6% lifetime risk of complications 3–5 from a<br />

Meckel’s diverticulum, these combined findings<br />

suggest that incidental Meckel’s diverticulectomy is<br />

justified in select cases in which the anatomical<br />

features of the diverticulum put patients at higher<br />

risk for future complications.<br />

Reported morbidity and mortality rates for diverticulectomy<br />

in symptomatic patients vary widely,<br />

ranging between 0–18% and 0–9%, respectively.<br />

11,13,24,26,43 The Mayo Clinic study 13 reported a<br />

12% incidence of early postoperative complications;<br />

these were mainly wound infection (3%),<br />

prolonged ileus (3%) and anastomotic leak (2%).<br />

The mortality rate was 1.5%. The incidence of late<br />

postoperative complications during a 20-year<br />

follow-up was 7%. In our own institution, operation<br />

in symptomatic pediatric patients has yielded excellent<br />

long-term outcomes. The perioperative complication<br />

rate has been less than 6%, with the most<br />

common complications being wound infection and<br />

prolonged ileus. We have had no reported deaths in<br />

over a decade.


738<br />

Meckel’s diverticulum<br />

REFERENCES<br />

1. Meckel JF. Uber die divertikel am daemkanal. Archiv<br />

Physiol 1809; 9: 421–453.<br />

2. Meckel JF. Handbuch der Pathologischen Anatomie. vol 1.<br />

Leipzig, Germany: Reclam, 1812<br />

3. Soltero JM. Bill AH. The natural history of Meckel’s<br />

diverticulum and its relation to incidental removal. A<br />

study of 202 cases of diseased Meckel’s diverticulum<br />

found in King County, Washington, over a fifteen year<br />

period. Am J Surg 1976; 132: 168–173.<br />

4. Matsagas MI, Fatouros M, Koulouras B et al. Incidence,<br />

complication and management of Meckel’s diverticulum,<br />

Arch Surg 1995; 130: 143–146.<br />

5. St-Vil D, Brandt ML, Panic S et al. Meckel’s diverticulum<br />

in children: a 20-year review. J Pediatr Surg 1991; 26:<br />

1289–1292.<br />

6. Soderlund S. Meckel’s diverticulum: a clinical and histologic<br />

study. Acta Chir Scand Suppl 1959; 248: 213–233.<br />

7. Amoury RA, Snyder CL. Meckel’s diverticulum. In<br />

O’Neill JA, Rowe MI, Grosfeld JL et al. eds. Pediatric<br />

Surgery, 5th edn. St Louis, MO: Mosby; 1998:<br />

1173–1184.<br />

8. Haubrich WS, Schaffner F, Berk JE, Bockus HL.<br />

Gastroenterology, 5th edn, vol 2. Philadelphia: WB<br />

Saunders, 1995: 912–914.<br />

9. Yachouchy EK, Marano AF, Etienne JCF et al. Meckel’s<br />

diverticulum. J Am Coll Surg 2001; 192: 658–662.<br />

10. DeBartolo HM Jr, van Heerden JA. Meckel’s diverticulum.<br />

Ann Surg 1976: 183: 30–33.<br />

11. Mackay WC, Dineen PA. Fifty-year experience with<br />

Meckel’s diverticulum. Surg Gynecol Obstet 1983; 156:<br />

56–64.<br />

12. Vane DW, West KW, Grosfeld JL. Vitelline duct anomalies.<br />

Experience with 217 childhood cases. Arch Surg<br />

1987; 122: 542–547.<br />

13. Cullen JJ, Kelly KA, Moir CR et al. Surgical management<br />

of Meckel’s diverticulum. An epidemiologic, populationbased<br />

study. Ann Surg 1994; 220: 564–569.<br />

14. Yamaguchi M, Takeuchi S, Awazu S. Meckel’s diverticulum<br />

investigation of 600 patients in the Japanese literature.<br />

Am J Surg 1978; 136: 247–249.<br />

15. Weinstein EC, Cain JC, ReMine WH. Meckel’s diverticulum:<br />

55 years of clinical and surgical experience. JAMA<br />

1962; 182: 131–133.<br />

16. Moses WR. Meckel’s diverticulum. Report of two<br />

unusual cases. N Engl J Med 1947; 237: 118–122.<br />

17. Baker AL, Marshall SF. Meckel’s diverticulum: a report<br />

on ninety three cases. Am Surg 1955; 21: 1173–1181.<br />

18. Simms M, Corkery J. Meckel’s diverticulum: its association<br />

with congenital malformation and the significance<br />

of atypical morphology. Br J Surg 1980; 67: 216–219.<br />

19. Andreyev HJN, Owen RA, Thompson I et al. Association<br />

between Meckel’s diverticulum and Crohn’s disease: a<br />

retrospective review. Gut 1994; 35: 788–790.<br />

20. Nicol JW, MacKinlay GA. Meckel’s diverticulum in<br />

exomphalos minor. J R Coll Surg Edinb 1994; 39: 6–7.<br />

21. Moyana TN. Carcinoid tumors arising from Meckel’s<br />

diverticulum: a clinical, morphologic, and immunohistochemical<br />

study. Am J Clin Pathol 1989; 91: 52–56.<br />

22. Nies C, Zielke A, Hasse C et al. Carcinoid tumors of<br />

Meckel’s diverticula. Report of two cases and review of<br />

the literature. Dis Colon Rectum 1992; 35: 589–596.<br />

23. Silk YN, Douglass HO Jr, Penetrante R. Carcinoid tumor<br />

in Meckel’s diverticulum. Am Surg 1988; 54: 664–667.<br />

24. Leijonmarck CE, Bonman-Sandelin K, Frisell J et al.<br />

Meckel’s diverticulum in the adult. Br J Surg 1986; 73:<br />

146-149.<br />

25. Arnold JF, Pellicane JV. Meckel’s diverticulum: a ten-year<br />

experience. Am Surg 1997; 63: 354–355.<br />

26. Rutherford RB, Akers DR. Meckel’s diverticulum: a<br />

review of 148 pediatric patients, with special reference<br />

to the pattern of bleeding and to mesodiverticular vascular<br />

bands. Surgery 1966; 59: 618–626.<br />

27. Bemelman WA, Bosma A, Wiersma PH et al. Role of<br />

<strong>Helicobacter</strong> <strong>pylori</strong> in the pathogenesis of complications<br />

of Meckel’s diverticula. Eur J Surg 1993; 159: 171–175.<br />

28. Kumar S, Small P, Mohammed R. <strong>Helicobacter</strong> <strong>pylori</strong> and<br />

Meckel’s diverticulum. J R Coll Surg Edinb 1991; 36:<br />

225–226.<br />

29. Fich A, Talley NJ, Shorter RG et al. Does <strong>Helicobacter</strong><br />

<strong>pylori</strong> colonize the gastric mucosa of Meckel’s diverticulum?<br />

Mayo Clin Proc 1990; 65: 187–191.<br />

30. Donnelly LF, Johnson JF. Case report: inverted Meckel’s<br />

diverticulum with associated microcolon. Clin Radiol<br />

1988; 53: 226–227.<br />

31. Pujari BD, Deodhare SG. Ascarideal penetration of<br />

Meckel’s diverticulum. Int Surg 1978; 63: 113–114.<br />

32. Hangloo VK, Koul I, Safaya R et al. Primary ascaridial<br />

perforations of small intestine and Meckel’s diverticulum.<br />

Indian J Gastroenterol 1990; 9: 287–288.<br />

33. Oguzkurt ˘<br />

P, Arda S, Kayaselcuk F et al. Cystic Meckel’s<br />

diverticulum: a rare cause of cystic pelvic mass presenting<br />

with urinary symptoms. J Pediatr Surg 2001; 36:<br />

1855–1858.<br />

34. Sfakianakis GN, Conway JJ. Detection of ectopic gastric<br />

mucosa in Meckel’s diverticulum and in other aberrations<br />

by scintigraphy: II. Indications and methods – a 10year<br />

experience. J Nucl Med 1981; 22: 732–738.<br />

35. Emamian SA, Shalaby-Rana E, Majd M. The spectrum of<br />

heterotopic gastric mucosa in children detected by Tc-<br />

99m pertechnetate scintigraphy. Clin Nucl Med 2001; 26:<br />

529–535.<br />

36. Swaniker F, Soldes O, Hirschl RB. The utility of technetium<br />

99m pertechnetate scintigraphy in the evaluation<br />

of patients with Meckel’s diverticulum. J Pediatr Surg<br />

1999; 34: 760–765.<br />

37. Stylianos S, Stein J, Flanigan LM et al. Laparoscopy for<br />

diagnosis and treatment of recurrent abdominal pain in<br />

children. J Ped Surg 1996; 31: 1158–1160.<br />

38. Teitelbaum DH, Polley TZ Jr, Obeid F. Laparoscopic diagnosis<br />

and excision of Meckel’s diverticulum. J Pediatr<br />

Surg 1994; 29: 495–497.<br />

39. Okazaki M, Higashihara H, Saida Y et al. Angiographic<br />

findings of Meckel’s diverticulum: the characteristic<br />

appearance of the vitelline artery. Abdom Imaging 1993;<br />

18: 15–19.<br />

40. Echenique M, Dominguez A, Echenique I et al.<br />

Laparoscopic diagnosis and treatment of Meckel's diverticulum<br />

complicated by gastrointestinal bleeding. J<br />

Laparoendosc Surg 1993; 3: 145–148.<br />

41. Sanders LE. Laparoscopic treatment of Meckel’s diverticulum.<br />

Obstruction and bleeding managed with minimal<br />

morbidity. Surg Endosc 1995; 9: 724–727.<br />

42. Huang CS, Lin LH. Laparoscopic Meckel’s diverticulectomy<br />

in infants: report of three cases. J Pediatr Surg<br />

1993; 281: 1486–1489.<br />

43. Ludtke FE, Mende V, Kohler H et al. Incidence and<br />

frequency of complications and management of Meckel’s<br />

diverticulum. Surg Gynecol Obstet 1989; 169: 537–542.


45<br />

Introduction<br />

Acute appendicitis<br />

Adolfo Bautista Casasnovas<br />

Acute appendicitis is a common pediatric surgical<br />

disease requiring urgent attention. In many cases it<br />

shows characteristic clinical manifestations, but in<br />

others it may simulate several other syndromes,<br />

leading to delays in diagnosis and thus increased<br />

morbidity and mortality. It is the most frequent<br />

cause of emergency surgery in infancy, and one of<br />

the most frequent causes of abdominal pain.<br />

In 1731, Aymand reported in Philosophical<br />

Transactions of the Royal Society the first successful<br />

appendectomy, performed in a child with a<br />

very large inguinoscrotal hernia, the sac of which<br />

contained an appendix perforated by a needle and<br />

giving rise to a stercoral fistula. The intervention<br />

lasted 30min, and the patient survived.<br />

The term ‘appendicitis’ was coined in 1886 by the<br />

American pathological anatomist Reginald Fitz,<br />

who stressed the need for early diagnosis and<br />

surgery in cases of acute inflammation of the<br />

appendix. 1 In 1889, McBurney declared that ‘In<br />

every case the seat of greatest pain, determined by<br />

the pressure of one finger, has been very exactly<br />

between an inch and a half and two inches from<br />

the anterior spinous process of the ilium on a<br />

straight line drawn from that process to the<br />

umbilicus’. 2 Since that time this manifestation has<br />

been known as the McBurney sign, marking the<br />

start of a period of greater scientific rigor in the<br />

diagnosis and treatment of acute appendicitis,<br />

with early diagnosis and treatment being recognized<br />

as essential.<br />

Epidemiology<br />

The incidence of acute appendicitis in the USA<br />

has been estimated at about 11 cases per 10000<br />

people per year, rising to 23 per 10000 per year in<br />

the 10–19-year-old age group, with slightly higher<br />

incidence among boys. Incidence among<br />

preschool children is lower, but in this age group<br />

delayed diagnosis and complications are more<br />

frequent. 3,4 About a quarter of operated acute<br />

appendicitis cases are found to be perforated at<br />

surgery.<br />

In fact, only about 0.5–2% of cases of acute<br />

appendicitis appear in children aged less than 2<br />

years. Heredity seems to be important: children<br />

who have appendicitis are twice as likely to have<br />

a positive family history as children who present<br />

lower abdominal quadrant pain not due to appendicitis,<br />

and almost three times as likely to have a<br />

positive family history as surgical controls without<br />

abdominal pain. 5<br />

In recent decades, as a consequence of the development<br />

of new and more effective antibiotics, and<br />

of advances in postoperative treatment, a marked<br />

reduction in morbidity and mortality has been<br />

achieved, but early diagnosis continues to be the<br />

most important factor for a good prognosis. 6 To<br />

reduce the incidence of complications and perforated<br />

appendicitis, practitioners should err on<br />

the side of caution: it is considered acceptable for<br />

up to 10% of laparotomies for suspected acute<br />

appendicitis to prove unnecessary (i.e. normal<br />

appendix).<br />

Anatomy<br />

The development of the cecum and appendix<br />

commences in the cecal diverticulum on the<br />

antimesenteric side of the caudal end of the medial<br />

intestine, around the 5th week of gestation. During<br />

its descent, the cecoappendiceal complex may<br />

locate in a retrocecal or retrocolonic position<br />

739


740<br />

Acute appendicitis<br />

(about 75% of cases) or inferomedial to the cecum<br />

(about 20% of cases). The infant cecum is located<br />

in the right iliac fossa in about 55% of individuals.<br />

7 In newborn children it is funnel-shaped with<br />

the ‘base’ oriented upwards and the vertex continuous<br />

with the appendix. The proximal end of the<br />

appendix is narrower, and its muscle layers<br />

thicker, which explains why perforations occur<br />

most frequently at the tip. Both cecum and appendix<br />

are irrigated by the ileocecal trunk, the ileocolic<br />

artery. The arterial supply is terminal, so that<br />

thrombosis leads to rapid necrosis. The veins drain<br />

to the superior mesenteric vein, which is a<br />

common route for propagation of infections from<br />

the appendix to the liver.<br />

The lymphatics of the cecum and appendix drain<br />

into a group of five or six lymph nodes located in<br />

the area of the terminal ileum, so that infections of<br />

the appendix may reach the peritoneum or even<br />

the thorax. 8<br />

The cecum and appendix are innervated by the<br />

solar plexus via the superior mesenteric plexus,<br />

from thoracic sympathetic nerves IX–XI. The autonomic<br />

innervation is provided by a small number<br />

of ganglion cells irregularly distributed in the<br />

submucosa and muscular layers, without forming<br />

a plexus. 9<br />

Pathophysiology<br />

The function of the appendix is unknown: it<br />

possibly plays some immunological role in the<br />

identification of foreign proteins and bacteria, and<br />

in the manufacture of IgA. It has been demonstrated<br />

experimentally that mucus secretion<br />

(1–2ml/day) when drainage is blocked increases<br />

intraluminal pressure with consequent vascular<br />

compromise. The appendix shows independent<br />

irregular peristaltic activity, detectable on<br />

radiography. 9<br />

Obstruction is a fundamental factor in the development<br />

of acute appendicitis. The position of the<br />

blind sac of the appendix favors obstruction of the<br />

lumen (by foreign bodies, fecaliths, doubling of the<br />

lumen upon itself, lymphoid hyperplasia, infection<br />

or tumor), which increases intraluminal<br />

pressure leading to ischemia, bacterial invasion,<br />

necrosis and perforation. 10 In the early phases,<br />

activation of receptors in the intestinal wall leads<br />

to perception of pain in the periumbilical region;<br />

in later phases, pain is perceived in the right iliac<br />

fossa, as a result of irritation due to purulent<br />

secretion or contact between the appendix and the<br />

parietal peritoneum.<br />

The bacteriological causes of appendiceal infection/inflammation<br />

are highly varied. For some<br />

authors, infection almost always occurs via the<br />

blood, 11 while others consider that intraluminal<br />

bacteria are generally responsible. 12–14 The characteristic<br />

bacteria are those present in the human<br />

colon, predominantly anaerobic, including<br />

Escherichia coli, Enterococcus, Bacterioides<br />

fragilis, Pseudomonas, Klebsiella and Clostridium.<br />

The higher incidence of acute appendicitis at<br />

certain times of the year suggests that respiratory<br />

and/or digestive infections showing similar<br />

seasonal trends may predispose to lymphoid<br />

hyperplasia and resulting narrowing of the<br />

appendiceal lumen.<br />

The apparent etiological relationship with certain<br />

infections (whether exanthematous or not) probably<br />

reflects lymphoid hyperplasia and resulting<br />

inflammation and obstruction, as observed in<br />

measles, infectious mononucleosis and infections<br />

with HIV, adenovirus or coxsackievirus B. The<br />

incidence of acute appendicitis is abnormally high<br />

in patients with such infections, and with a higher<br />

incidence of perforation because of the difficulty<br />

of early diagnosis. 15,16<br />

Likewise, it has been suggested that intestinal<br />

parasites may contribute to appendicitis, although<br />

a study in the USA found that parasites were<br />

present in less than 10% of 100 extirpated appendices.<br />

17<br />

Carcinoid tumors may cause acute appendicitis in<br />

children, especially when close to the tip; in such<br />

cases simple appendectomy is generally sufficient<br />

treatment. 18<br />

Clinicopathological course<br />

The course of appendicitis – from the normal state<br />

to gangrene – follows a series of clinicopathological<br />

states, classifiable as follows:


Simple appendicitis<br />

The first clinical manifestation is congestion of the<br />

subserous blood vessels and inflammatory<br />

exudate, together with loss of the characteristic<br />

shininess of the serous layer. Parietal thickening<br />

and luminal obstruction are typically manifested<br />

by swelling of the appendix and particularly its<br />

tip, with appearance of fibrin. Microscopy reveals<br />

small intraluminal groupings of pyocytes, scaling<br />

plaques in the mucosa, cryptic abscesses and<br />

scarce polymorphonuclear lymphocytes in the<br />

lamina propria. At this stage the patient typically<br />

begins to feel periumbilical pain, which gradually<br />

shifts towards the right iliac fossa.<br />

Phlegmonous appendicitis<br />

Suppurative areas are seen on the wall of the<br />

appendix, and venous thromboses in the mesoappendix.<br />

Microperforations are observed, generally<br />

in large numbers. The appendix is typically<br />

covered with omentum. Microscopy reveals an<br />

inflammatory infiltrate, together with evident<br />

signs of thrombosis and necrosis.<br />

Ulceronecrotic appendicitis<br />

The appendix has become a soft, friable, purplish<br />

red structure, with very evident signs of necrosis,<br />

including in some cases macroscopic perforation<br />

and purulent exudate. 19<br />

Fibrin increases the adherence of the appendix to<br />

neighboring structures, above all in older children.<br />

Initially, the peritoneal cavity is seen to contain a<br />

clear sterile liquid, due to increased pressure<br />

within the lumen. Later, the cavity is invaded by<br />

fibrin, leukocytes and pathogens, forming an<br />

exudate which, when invaded by bacteria (from<br />

the intestine, or due to perforation of the appendix)<br />

leads to abdominal abscess formation, generally<br />

in the right iliac fossa, close to the cecum and<br />

usually foul-smelling; this typically occurs in the<br />

phlegmonous or necrotic stages of acute appendicitis.<br />

Diagnosis<br />

The incidence of acute appendicitis gradually<br />

increases from age 1 year onwards, peaking at<br />

Diagnosis 741<br />

around age 12 years, when the number of<br />

lymphatic follicles in the appendix is maximal,<br />

favoring its obstruction. 7 Appendicitis is rare<br />

during the first year of life, although an association<br />

has been reported between Hirschsprung’s disease<br />

and appendiceal perforation. 20<br />

Diagnosis of acute appendicitis may be easy or<br />

difficult, depending on the patient. All clinicians<br />

are familiar with the classic course of acute appendicitis,<br />

but early diagnosis is necessary to minimize<br />

complications and ensure successful treatment.<br />

Diagnosis can almost always be achieved on<br />

the basis of a brief history and physical exploration<br />

of the abdomen. Difficulties of diagnosis<br />

may arise with obese patients (particularly preadolescent<br />

girls), and with girls younger than 2<br />

years. Except in these two patient categories,<br />

analytical and imaging studies are not usually<br />

necessary, since they provide relatively little additional<br />

information.<br />

Clinical manifestations<br />

The classic sequence (fever, persistent abdominal<br />

pain and localized pain on palpation at<br />

McBurney’s point) starts with periumbilical pain,<br />

preceded by appetite loss in about 50–60% of children.<br />

Over a varying period of time, this pain gradually<br />

shifts to the right iliac fossa, with a corresponding<br />

increase in pain on palpation. The pain<br />

may be constant, colic or stabbing, or a dull ache.<br />

Characteristically the pain is implacable and is<br />

exacerbated by movements and pressure, making<br />

deambulation painful and difficult. 21<br />

The change in location of pain is an important<br />

diagnostic sign, related to parietal peritoneal irritation,<br />

which as it intensifies leads to disappearance<br />

of the periumbilical pain.<br />

Nausea and vomiting are also important for diagnosis.<br />

They appear after the onset of pain. Indeed,<br />

if vomiting appears before the onset of pain, other<br />

diagnoses (notably enteritis) should be considered.<br />

During the early stages of appendicitis, diarrhea<br />

simulating that occurring in the early stages of<br />

gastroenteritis is not infrequent; furthermore, in<br />

very advanced stages, diarrhea is characteristic.<br />

Urological symptoms (dysuria, urine retention)<br />

may be similarly misleading, even when urine<br />

analysis reveals high white blood cell (WBC)


742<br />

Acute appendicitis<br />

counts, given the proximity of the appendix to the<br />

bladder and the ureter. In other words, the clinician<br />

should not allow diarrhea or urological symptoms<br />

to distract from the possibility of acute<br />

appendicitis.<br />

A child with acute appendicitis typically walks<br />

bent over and slowly, taking care not to make<br />

brusque movements. Often, he or she finds it difficult<br />

or impossible to get up onto the examination<br />

table.<br />

Fever, which appears after pain and vomiting, is<br />

generally not very high; indeed, patients typically<br />

have low-grade fever. In many cases there is a<br />

marked difference (up to about 1°C) between axillary<br />

and rectal temperature. Early-onset high fever<br />

argues against acute appendicitis, but late-onset<br />

high fever with generalized abdominal pain<br />

suggests peritonitis, which is also accompanied by<br />

general malaise.<br />

Children with appendicitis are typically anorexic.<br />

When a child with abdominal pain shows strong<br />

appetite, appendicitis can generally be ruled out,<br />

although there are exceptions. Symptoms may be<br />

influenced by the anatomic location of the appendix.<br />

For example, a retrocecal appendix may cause<br />

flank pain or back pain. If the inflamed tip<br />

contacts the ureter, pain may be experienced in the<br />

inguinal region or testes, and may cause urinary<br />

symptoms. Pelvic appendicitis with the inflamed<br />

tip contacting the bladder may cause pollakiuria<br />

and/or dysuria, or ureteral compression with<br />

hydronephrosis (Figure 45.1).<br />

Approximately two-thirds of appendices are retrocecal<br />

or retrocolic. In some cases the appendix<br />

may cross the abdomen, with the tip extending<br />

into the other quadrant. In cases of incomplete<br />

rotation of the intestine, the appendix may be<br />

found in the right upper quadrant of the abdomen,<br />

or less frequently on the left side. 22<br />

In pre-weaning and young weaned infants, the<br />

diagnosis of acute appendicitis is more difficult,<br />

and from 24h after onset the risk of perforation<br />

increases considerably. 23 This is particularly problematic,<br />

given the still immature immunity of<br />

young infants. The omentum is also not fully<br />

developed, so that the effects of the appendicitis<br />

(e.g. fluid release into the peritoneal cavity) are<br />

typically less localized. Early diagnosis is thus<br />

Figure 45.1 Urography showing a mass in the right iliac<br />

fossa compressing the bladder and obstructing the ureter,<br />

giving rise to hydronephrosis, with an abscess secondary<br />

to undiagnosed acute appendicitis.<br />

imperative, and is complicated by the difficulty of<br />

clinical examination. Infants with acute appendicitis<br />

typically show vomiting and irritability, and<br />

draw up their legs to reduce pain. Other common<br />

manifestations include abdominal distension,<br />

diarrhea, lethargy and anorexia, together with<br />

fever. In 50% of cases an abdominal mass is<br />

detectable on palpation. 24<br />

In conclusion, all patients with abdominal pain,<br />

vomiting and moderate fever should be considered<br />

as suffering acute appendicitis until otherwise<br />

demonstrated.<br />

Physical examination<br />

Physical examination performed with empathy,<br />

patience and experience is more valuable than a<br />

well-equipped laboratory. 25 A good start for the


physical examination is to take the patient’s pulse,<br />

for at least 30s; indeed, the pulse rate may be a<br />

more useful diagnostic clue than temperature.<br />

Observations of features such as general condition,<br />

mucocutaneous wetness or dryness, posture, and<br />

frequency and type of respiration (in small children<br />

with diaphragmatic respiration, immobility<br />

of the diaphragm due to peritoneal irritation may<br />

lead to respiratory compromise) may all be useful<br />

for achieving the diagnosis. Excessively hasty and<br />

brusque examinations may of course lead to incorrect<br />

diagnosis. Inspection of the throat and rectal<br />

examination are uncomfortable for the patient, and<br />

should be left for last. A distended bladder is<br />

painful and may lead to false diagnosis; the child<br />

should therefore urinate before the examination.<br />

Abdominal auscultation should be performed<br />

routinely, and should precede palpation. The<br />

examiner should take advantage of the stethoscope<br />

to auscultate suspected areas. A silent abdomen<br />

suggests inflammatory pathology, while hyperperistaltism<br />

is more indicative of gastroenteritis or<br />

intestinal obstruction.<br />

If the patient is asleep, an initial abdominal palpation<br />

should be performed above the clothing, since<br />

a sleeping patient is easier to examine. Some<br />

maneuvers, such as slight bending of the hips,<br />

improve relaxation. In some patients, abdominal<br />

palpation may be so difficult that it necessitates<br />

sedation to suppress voluntary contractions. 17<br />

Palpation should always be first superficial and<br />

then deep. Involuntary reflex contractions and<br />

guarding are very important. To locate pain in<br />

response to deep palpation, the palpation should<br />

start in an area without pain, and the patient’s face<br />

should be watched for signs of discomfort.<br />

In addition to McBurney’s point, other palpation<br />

points have been described (Morris’s, Gray’s,<br />

Loeper’s, Lanz’s, Kelly’s, etc.), together with<br />

maneuvers aimed at localization of pain (psoas<br />

maneuver, obturator test, etc.). 10–13,17 In fact, the<br />

important thing to look for during examination is<br />

any localized area of abdominal pain.<br />

Rebound tenderness or Blumberg’s sign (i.e. pain<br />

felt on sudden release of steady pressure in the<br />

right iliac fossa region) simply reflects irritation of<br />

the parietal peritoneum of the inflamed appendix,<br />

and associated fluid secretion.<br />

Laboratory tests 743<br />

Rectal examination invariably causes discomfort<br />

to children, but is necessary for detection of possible<br />

pelvic abscess, right-sided pain, or pain on<br />

movement of the bladder.<br />

Laboratory tests<br />

WBC counts are of limited diagnostic value. It is<br />

increasingly clear that diagnosis of acute appendicitis<br />

is basically clinical. However, a WBC count<br />

may be useful to confirm the diagnosis. Acute<br />

appendicitis leads to an increased WBC count<br />

(generally between 10000 and 20000 per mm 3 ),<br />

with increased polymorphonuclear leukocytes,<br />

neutrophils and immature neutrophils. The WBC<br />

count will generally be higher in cases of peritonitis<br />

or perforation.<br />

Some patients with acute appendicitis may,<br />

however, show a normal or low WBC count. This<br />

may reflect retrocecal or occult appendicitis. In<br />

such cases toxic granulations should be looked for.<br />

Determination of C-reactive protein level helps<br />

improve diagnostic certainty, but only when pain<br />

has been present for over 12–14h. 26,27<br />

Experience indicates that neutrophilia is more<br />

decisive for diagnosis than leukocytosis. In doubtful<br />

cases it is useful to monitor WBCs, as long as<br />

the patient is not receiving antibiotics. 28,29<br />

Urine sediment analyses are useful for detecting<br />

diseases of the urinary tract, ranging from infection<br />

to lithiasis; such analyses can be considered<br />

more important than WBC counts. However, it<br />

should be borne in mind that irritation of the<br />

ureter or the bladder by a retrocecal or pelvic<br />

appendix may lead to the appearance of leukocytes<br />

or blood in the urine, confusing the diagnosis. 30,31<br />

Diagnostic imaging<br />

It is currently accepted that simple radiography is<br />

both inadequately sensitive and inadequately<br />

specific for diagnosis of acute appendicitis; in<br />

addition, the cost–benefit relationship is poor. It<br />

thus seems probably that we will see a decline in<br />

the use of simple radiography as a primary diagnostic<br />

approach in patients with acute pain in the<br />

right iliac fossa; however, it will probably remain


744<br />

Acute appendicitis<br />

useful for detecting secondary problems associated<br />

with inflammation. The signs most frequently<br />

encountered are a dilated cecum with air–fluid<br />

level, antalgic scoliosis of the right concavity,<br />

obscuration of the right psoas muscle, free peritoneal<br />

fluid, minimal pneumoperitoneum and<br />

appendiceal fecalith. 30 The presence of a fecalith is<br />

highly suggestive of acute appendicitis if clinical<br />

characteristics are observed; however, an appendicolith<br />

(Figure 45.2) is observed in only 10% of<br />

cases.<br />

Ultrasonography has constituted a significant<br />

advance in the diagnosis of acute appendicitis, in<br />

view of its rapidity, innocuity, sensitivity of<br />

85–90% and specificity of 92–96%. It is of particular<br />

value in adolescent and prepubertal girls, since<br />

it helps to identify other possible causes of abdominal<br />

pain. It should be the first approach in doubtful<br />

cases. It is worth mentioning that the examiner<br />

must be an expert in pediatric ultrasound. The<br />

Figure 45.2 Simple radiography of the abdomen in a<br />

patient with acute appendicitis, showing anomalous<br />

distribution of gas with scoliosis of the right concavity<br />

and an appendicolith in the right iliac fossa.<br />

normal appendix is not usually visualized on<br />

ultrasonography. The most characteristic signs are<br />

the presence of free liquid and a distended noncompressible<br />

tubular structure corresponding to<br />

the inflamed appendix, with a size greater than<br />

6mm. Other useful signs are pericecal inflammatory<br />

changes and an appendix located in the retrocecal<br />

position or in front of the psoas (Figure<br />

45.3). 32–34<br />

In recent years, abdominal computed tomography<br />

(CT) has become the most informative imaging<br />

technique in the study of patients with atypical<br />

manifestations, with sensitivity and specificity of<br />

98% according to Rao et al. 35 In the future, it seems<br />

likely that continued development of helical CT<br />

Figure 45.3 Ultrasonography of a patient with acute<br />

appendicitis, showing a distended non-compressible<br />

tubular structure, with size greater than 6mm, an<br />

appendicolith and surrounded by free fluid.


technology will make this the diagnostic method<br />

of choice. 35 It is accepted that the sensitivity and<br />

specificity of diagnosis by CT is greater than that<br />

of ultrasound, with the following criteria: fluidfilled<br />

tubular structure measuring >6mm in<br />

maximum diameter, fat stranding, abscess or<br />

phlegmon in adjacent tissue, appendicolith and<br />

focal cecal apical thickening. 32,36,37<br />

Recent studies suggest that imaging should be<br />

performed even in cases where the diagnosis of<br />

acute appendicitis is clinically likely, as the procedure<br />

would enable the detection of a high<br />

percentage of normal appendices and the detection<br />

of other diagnoses. 38,39<br />

Differential diagnosis<br />

The disorders most frequently confused with acute<br />

appendicitis are urinary diseases (infections,<br />

hydronephrosis, lithiasis), basal pneumonias,<br />

constipation and gastroenteritis. 17,22,30,40 It should<br />

here be stressed that repeated examination is of<br />

key importance for differential diagnosis, with<br />

careful monitoring of the patient during the first<br />

few hours of abdominal symptoms.<br />

Pollakiuria with dysuria and an increased pyocyte<br />

count in the urine suggests a urinary tract infection,<br />

but these signs may also be observed in<br />

pelvic appendicitis, or in appendicitis with the<br />

appendix close to the ureter or bladder. It may be<br />

helpful to note that flank pain is more commonly<br />

associated with renal infection, in which fever and<br />

leukocytosis may be observed despite minimal<br />

abdominal symptoms. Other urological pathologies<br />

to be borne in mind are obstructive disorders<br />

such as pyeloureteral or ureterovesical stenosis, as<br />

well as vesicoureteral reflux, all of which may<br />

cause acute abdominal pain. 41<br />

Infectious pathologies of the lower lobe of the right<br />

lung may cause abdominal pain and generalized<br />

abdominal muscular spasm, but such cases do not<br />

show localized hypersensitivity in the right iliac<br />

fossa; in addition, these children are typically<br />

drowsy and sleep a great deal, with high fever and<br />

sometimes cough. In rarer cases, the pneumonia is<br />

secondary to appendicitis. This occurs when the<br />

inflamed appendix is in the retrocecal or subhepatic<br />

location, and develops an abscess that invades<br />

Differential diagnosis 745<br />

the subphrenic area with pleural effusion and radiological<br />

images suggestive of pulmonary pathology.<br />

Constipation is common in older children, and<br />

may involve crises with abdominal pain, fever,<br />

vomiting and leukocytosis. There are not always<br />

clear antecedents of constipation. In these cases<br />

there is no migration of pain from the epigastric<br />

region to McBurney’s point, and in addition there<br />

are no signs of peritoneal irritation. Hard packed<br />

feces may be palpated through the abdominal wall.<br />

Gastroenteritis is probably the most common<br />

cause of acute abdominal pain, and is frequently<br />

confused with appendicitis. Early-onset vomiting<br />

accompanied by abdominal pain is the most reliable<br />

feature of gastroenteritis, while in appendicitis<br />

vomiting appears late after the onset of abdominal<br />

pain. Abundant foul-smelling diarrhea is<br />

likewise indicative of gastroenteritis, by contrast<br />

with the scant mucosal secretions appearing in<br />

pelvic appendicitis. Nevertheless, it should be<br />

stressed that frequent bowel movements do not<br />

rule out appendicitis. 42<br />

In pubertal girls, premenstrual syndrome and<br />

dysmenorrhea are common causes of unnecessary<br />

surgery, and their differential diagnosis is not<br />

always easy. Ovarian cysts can be diagnosed by<br />

rectal examination or ultrasonography; they cause<br />

acute pain when ruptured or torsioned, and the<br />

patient typically shows vomiting. In doubtful<br />

cases, laparoscopy is of key value.<br />

In primary peritonitis due to pneumococcus or<br />

streptococcus, the severe effects on general wellbeing<br />

call for emergency surgery; in any case,<br />

acute appendicitis and peritonitis are often not<br />

distinguishable without surgery.<br />

Differential diagnosis with respect to mesenteric<br />

lymphadenitis may be very difficult, because of<br />

the close clinical similarities with acute appendicitis.<br />

Criteria indicative of adenitis include<br />

initial high fever, severe spasmodic abdominal<br />

pain, absence of anorexia and, in some cases, a<br />

history of respiratory infection. As noted in the<br />

classic texts, careful monitoring of the patient is<br />

extremely important. 12<br />

Inflammatory bowel diseases, notably Crohn’s<br />

disease, may sometimes be first detected during


746<br />

Acute appendicitis<br />

acute appendicitis, but generally the patient will<br />

present with a prolonged history of abdominal<br />

pain, diarrhea attacks and growth restriction, in<br />

some cases with a positive family history.<br />

Patients with severe leukopenia due to cancer or<br />

chemotherapy may show invasive infection of the<br />

entire wall of the cecal region (i.e. typhlitis), with<br />

pain and guarding of the right iliac fossa. If there is<br />

no improvement on antibiotic treatment, or if<br />

symptoms are suggestive of irritation or perforation<br />

of the bowel, surgery should be performed.<br />

Other differential diagnoses to be taken into<br />

account are viral hepatitis (especially in the<br />

preicteric phase), as a result of lymphoid–<br />

appediceal involvement by viral infections (hepatitis<br />

B, Herpes, coxsackievirus, adenovirus); intussusception;<br />

viral exanthematous diseases; sickle<br />

cell anemia; cholecystitis; pancreatitis; omentum<br />

infarction; and duodenal ulcer (Table 45.1).<br />

Treatment<br />

The treatment of choice for acute appendicitis is of<br />

course early appendectomy. Rapid administration<br />

of intravenous fluid and electrolytes (to restore<br />

acid–base balance) is fundamental for a good<br />

course. Depending on the clinical characteristics<br />

(degree of dehydration, fasting status, presence of<br />

vomiting), the rehydration fluid may be physiological<br />

saline with glucose, or lactate Ringer’s solution,<br />

in either case with supplementary potassium<br />

if necessary. In cases of peritonitis, losses to the<br />

third space should be taken into account, since<br />

this will increase the rehydration requirement.<br />

Once surgical intervention has been decided upon,<br />

intravenous antibiotic treatment should be<br />

started. 43<br />

In cases of intestinal obstruction or distension, the<br />

insertion of a nasogastric tube facilitates decompression<br />

of the abdomen and minimizes the risk of<br />

aspiration during anesthesia induction.<br />

If the appendix is gangrenous with peritonitis and<br />

the patient is seriously ill, intensive reanimation<br />

will be required before appendectomy, with immediate<br />

intravenous administration of antibiotics<br />

together with rehydration. In some cases, a vesical<br />

drainage will be necessary as well as nasogastric<br />

Table 45.1 Main differential diagnosis of<br />

acute appendicitis<br />

Urological pathology<br />

Urinary tract infections<br />

Pneumonia<br />

Constipation<br />

Gastroenteritis<br />

Premenstrual syndrome<br />

Ovarian pathology<br />

Primary peritonitis<br />

Mesenteric adenitis<br />

Meckel’s diverticulitis<br />

Henoch–Schönlein purpura<br />

Viral hepatitis<br />

Inflammatory bowel disease<br />

Intussusception<br />

Typhlitis<br />

Viral exanthematous diseases<br />

Sickle cell anemia<br />

Cholecystisis<br />

Pancreatitis<br />

Omentum infarction<br />

Duodenal ulcer<br />

Abdominal migraine<br />

Acute porphyria<br />

Familial Mediterranean fever<br />

Hemolytic uremic syndrome<br />

drainage, but generally the patient can undergo<br />

surgery within 6–8h of the start of reanimation.<br />

The type of antibiotic therapy used varies among<br />

different centers. Some authors have recommended<br />

prophylactic administration of one dose<br />

of cefotoxin before surgery and three doses after<br />

surgery. 44 However, cefotoxin may be less effective<br />

than a combination of aminoglycosides, synthetic<br />

penicillin and metronidazole in the treatment of a<br />

perforated appendix. Other authors have<br />

suggested the use of piperacillin and tazobactam. 30<br />

Presurgery treatment with ampicillin, gentamycin<br />

and clindamycin has been successfully used in<br />

some countries since the 1970s. 45 In the Pediatric<br />

Surgery Service of our hospital (the Santiago de<br />

Compostela University Hospital in northwest


Spain), we currently use treatment with<br />

tobramycin plus metronidazole, with a low incidence<br />

(


748<br />

Acute appendicitis<br />

After surgery of any type, late adherences may be<br />

observed, but a large proportion of adherencerelated<br />

obstructions are secondary to surgery for<br />

acute appendicitis, even though they first present<br />

months or even years later. Adherences and infections<br />

of this type have also been implicated in the<br />

sterility observed in some girls after surgery for<br />

acute appendicitis; 53 however, other studies have<br />

not been able to detect any raised incidence of<br />

sterility in patients of this type. 54<br />

Summary<br />

The large series to date have suggested that mortality<br />

in acute appendicitis is about 1%, and rather<br />

REFERENCES<br />

1. Fitz RH. Perforating inflammation of the vermiform<br />

appendix with special reference to its early diagnosis<br />

and treatment. Am J Med Sci 1886; 7: 321–346.<br />

2. McBurney C. Experience with early operative interference<br />

in cases of diseases of the vermiform appendix. NY<br />

State J Med 1889; 50: 676–684.<br />

3. Harmatz P. Acute apendicitis. In Finberg L, Kleinman<br />

RE, 2nd edn. Saunder Manual of Pediatric Practice. New<br />

York: WB Saunders, 2002: 590–592.<br />

4. Bratton SL, Haberrrkern CM, Waldhausen JH. Acute<br />

appendictis risks of complications: age and medicaid<br />

insurance. Pediatrics 2000; 106: 75-78<br />

5. Gauderer MW, Crane MM, Green JA et al. Acute appendictis<br />

in children: the importance of family history. J<br />

Pediatr Surg 2001; 36: 1214–1217.<br />

6. Helmer KS, Robinson EK, Lally KP et al. Standardized<br />

patient care guidelines reduce infectious morbidity in<br />

appendectomy patients. Am J Surg 2002; 183: 608–613.<br />

7. Testut L, Latarjet A. Tratado de anatomía humana, vol<br />

IV, 4th ed. Barcelona: Salvat, 1964: 396–432.<br />

8. Skandalakis JE, Gray SW. Embryology for Surgeons, 2nd<br />

edn. Baltimore: Williams and Wilkins, 1994: 224–245.<br />

9. Morson BC, Dawson IMP. The normal appendix. In<br />

Morson BC, Dawson IMP, eds. Gastrointestinal<br />

Pathology. Oxford: Blackwell Scientific Publications,<br />

1972: 391–407.<br />

10. Drachter R, Gossmann JR. Chirurgie des Kindesalter, 3rd<br />

edn. Leipzig: Van FCW Vagel, 1930: 231–257.<br />

11. Ombredanne L. Precis clinique et opératoire de chirurgie<br />

infantile, 12th edn. Paris: Masson, 1925: 532–561.<br />

12. Gross RE. Appendicitis. In Gross RE, ed. The Surgery of<br />

Infancy and Childhood. Philadelphia: WB Saunders,<br />

1953: 253–280.<br />

13. Ein SH. Appendicitis. In Ashcraft KW, Murphy JP,<br />

Sharp RJ, Sigalet DL, Snyder CL, Eds. Pediatric Surgery,<br />

higher in children aged less than 2 years. 55,56 The<br />

incidence of postoperative complications has<br />

previously been reported to be about 5–10%. Over<br />

the past 25 years in our hospital, the incidence of<br />

complications has been about 0.5%. This low incidence<br />

is perhaps attributable to the use of efficient<br />

broad-spectrum antibiotics, to progress in pre- and<br />

postoperative care, and to improved anesthesia<br />

techniques.<br />

Efforts to reduce morbidity and mortality further<br />

in acute appendicitis will require improved early<br />

diagnosis on the basis of anamnesis and physical<br />

examination, improved preoperative care and<br />

well-trained surgeons.<br />

3rd edn. Philadelphia: WB Saunders Company, 2000:<br />

571–579.<br />

14. Rautio M, Saxen H, Siitonen A et al. Bacteriology of<br />

histopathologically defined appendicitis in children.<br />

Pediatr Infect Dis J 2000; 19: 1078–1083<br />

15. Lawrence KB, Waring GW. Acute appendicitis complicating<br />

the acute infectious diseases of childhood. N Engl<br />

J Med 1949; 247: 1–6.<br />

16. Pancharoen C, Ruttanamongkol P, Suwangool P et al.<br />

Measles-associated appendicitis: two case reports and<br />

literature review. Scand J Infect Dis 2001; 33: 632–633.<br />

17. Folkman J. Appendicitis. In Ravitch MM, Welch KJ,<br />

Benson CD, Aberdeen E, Randolph JG, eds. Pediatric<br />

Surgery, 3rd edn. Chicago: Year Book Medical<br />

Publishers, 1979: 1004–1009.<br />

18. Pelizzo G, La Riccia A, Bouvier R et al. Carcinoid<br />

tumors of the appendix in children. Pediatr Surg Int<br />

2001; 17: 399–402.<br />

19. Tsujy M, Puri P, Reen DJ. Characterization of the local<br />

inflammatory response in appendicitis. J Pediatr<br />

Gastroenterol Nutr 1993; 76: 43–48.<br />

20. Martin LW, Perring EW. <strong>Neonatal</strong> perforation of the<br />

appendix in association with Hirschsprung’s disease.<br />

Ann Surg 1967; 766: 799–802.<br />

21. Mason Cobb L, Lelli JL. Appendicitis. In Reisdorff EJ,<br />

Roberts MR, Wiegenstein JG, eds. Pediatric Emergency<br />

Medicine Philadelphia: WB Saunders, 1993: 314–321.<br />

22. Cloud DT. Apendicitis. In Holder TM, Ashcraft KW, eds.<br />

Tratado de Cirugía Pediátrica, 2nd edn. Madrid:<br />

Editorial Interamericana–McGraw Hill, 1995: 484–491.<br />

23. Brickman ID, Leon W. Acute appendicitis in childhood.<br />

Surgery 1960; 60: 1083–1089.<br />

24. Grosfffeld JL, Weinberger M, Clatworthy HW. Acute<br />

appendicitis in the first two years of life. J Pediatr Surg<br />

1973; 8: 285–293.


25. Raffensperger JC. El abdomen agudo en lactantes y<br />

niños, 2nd edn. Buenos Aires: Bernardes Ediciones,<br />

1969: 121–141.<br />

26. Wetzman M. Diagnostic utility of white blood cell and<br />

differential counts. Am J Dis Child 1973; 129:<br />

1183–1189.<br />

27. Gronroos JM, Gronoos P. Leucocyte count and C-reactive<br />

protein in the diagnosis of acute appendicitis. Br J Surg<br />

1999; 86: 501–504.<br />

28. Andersson RE, Hugander A, Ravn H. Repeated clinical<br />

and laboratory examinations in patients with an equivocal<br />

diagnosis of appendicitis. World J Surg 2000; 24:<br />

479–485.<br />

29. Schwartz MZ, Bulas D. Acute abdomen. Laboratory<br />

evaluation and imaging. Semin Pediatr Surg 1997; 6:<br />

65–73.<br />

30. Lund DP, Folkman J. Appendicitis. In Walker WA, Durie<br />

PR, Hamilton JR, Walker-Smith JA, Watkins JB, eds.<br />

Pediatric Gastrointestinal Disease: Pathophysiology,<br />

Diagnosis and Management, 3rd edn. Hamilton,<br />

Ontario: BC Decker, 2000: 821–829.<br />

31. Rothrock SG, Pagane J. Acute appendicitis in children:<br />

emergency department diagnosis and management. Ann<br />

Emerg Med 2000; 36: 39–51.<br />

32. Garcia Pena BM, Mandl KD, Kraus SJ. Ultrasonography<br />

and limited computed tomography in the diagnosis and<br />

management of appendicitis in children. JAMA 1999;<br />

282: 1041–1046.<br />

33. Crady SK, Jones JS, Wyn T, Luttenton CR. Clinical validity<br />

of ultrasound in children with suspected appendicitis.<br />

Ann Emerg Med 1993; 22: 1125–1129.<br />

34. Sivit JS, Applegate KE, Stallion A et al. Imaging evaluation<br />

of suspected appendicitis in a pediatric population:<br />

effectiveness of sonography versus CT. Am J Roentgenol<br />

2000; 175: 977–980.<br />

35. Rao PM, Rhea JT, Novelline RA et al. Helical CT<br />

combined with contrast material administered only<br />

through the colon for imaging of suspected appendicitis.<br />

Am J Roentgenol 1997; 169: 1275–1280.<br />

36. Kaiser S, Frenckner B, Jorulf HK. Suspected appendicitis<br />

in children: US and CT – a prospective randomized<br />

study. Radiology 2002; 223: 633–638.<br />

37. Lowe LH, Perez R Jr, Scheker LE et al. Appendicitis and<br />

alternate diagnoses in children: findings on unenhanced<br />

limited helical CT. Pediatr Radiol 2001; 31: 569–577.<br />

38. Rettenbacher T, Hollerweger A, Gritzmann N et al.<br />

Appendicitis: should diagnostic imaging be performed if<br />

the clinical presentation is highly suggestive of the<br />

disease? Gastroenterology 2002; 123: 992–998.<br />

39. Pena BM, Taylor GA, Fishman SJ, Mandl KD. Costs and<br />

effectiveness of ultrasonography and limited computed<br />

tomography for diagnosing appendicitis in children.<br />

Pediatrics 2000; 106: 672–676.<br />

References 749<br />

40. Hecker WCH. Zur Einweisung Diagnose Appendicitis in<br />

Kindesalter. Arztl Wschr 1954; 9: 1149–1153.<br />

41. Bertin P, Tovar JA, Pellerin D. L’appendicitis avant trois<br />

ans. A propos de 77 observations en 12 ans. Rev Pediatr<br />

1973; 9: 121–128.<br />

42. Ferre M. Chirurgie infantile de urgencie, 2nd edn. Paris:<br />

Masson, 1958: 146–159.<br />

43. Neilson IR, Laberge JM, Nguyen LT et al. Appendicitis<br />

in children: current therapeutic recomendations. J<br />

Pediatr Surg 1990; 25: 1113–1116.<br />

44. Kaiser AB. Antimicrobial prophylaxis in surgery. N Engl<br />

J Med 1986; 315: 1129.<br />

45. Gilbert SR, Emmens RW, Putnam TC. Appendicitis in<br />

children. Surg Gynecol Obstet 1985; 161: 261.<br />

46. Andersen BR, Kallehave FL, Andersen HK. Antibiotics<br />

versus placebo for prevention of postoperative infection<br />

after appendectomy. Cochrane Database Syst Rev 2001:<br />

3: CD001439.<br />

47. Shackelford RT. Surgery of the Alimentary Tract.<br />

Philadelphia: WB Saunders, 1955: 2562–2567.<br />

48. Reiertsen O, Larsen S, Trondsen E et al. Randomized<br />

controlled trial with sequential design of laparoscopic<br />

versus conventional appendicectomy. Br J Surg 1997;<br />

84: 842–847.<br />

49. Chung RS, Rowland DY, Li P, Diaz J. A meta-analysis of<br />

randomized controlled trials of laparoscopic versus<br />

conventional appendectomy. Am J Surg 1999; 177:<br />

250–256.<br />

50. Moir CR. Appendectomy: open and laparoscopic<br />

approaches. In Spitz L, Coran AC, eds. Pediatric<br />

Surgery. London: Chapman, 1995: 408–410.<br />

51. Meguerditchian AN, Prasil P, Cloutier R et al.<br />

Laparoscopic appendectomy in children: a favorable<br />

alternative in simple and complicated appendicitis. J<br />

Pediatr Surg 2002; 37: 695–698.<br />

52. Little DC, Custer MD, May BH et al. Laparoscopic<br />

appendectomy: an unnecessary and expensive procedure<br />

in children? J Pediatr Surg 2002; 37: 310–317.<br />

53. Puri P, McGuinness EP, Guiney EJ. Fertility following<br />

perforated appendicitis in girls. J Pediatr Surg 1989; 24:<br />

547–549.<br />

54. Andersson R, Lambe M, Bergstrom R. Fertility patterns<br />

after appendicectomy: historical cohort study. Br Med J<br />

1999; 318: 963–967.<br />

55. Flum DR, Koepsell T. The clinical and economic correlates<br />

of misdiagnosed appendicitis: nationwide analysis.<br />

Arch Surg 2002; 137: 799–804.<br />

56. Blomqvist PG, Andersson RE, Granath F et al. Mortality<br />

after appendectomy in Sweden, 1987–1996. Ann Surg<br />

2001; 233: 455–460.


46<br />

Introduction<br />

Vascular lesions of the<br />

gastrointestinal tract in<br />

childhood<br />

Steven R Allen and Richard G Azizkhan<br />

Over the years, various histopathological systems<br />

combined with descriptive vernacular have been<br />

used to describe vascular lesions. Changes in<br />

nomenclature and inconsistencies in terminology<br />

have led to confusion and misunderstanding<br />

among medical specialists, making it difficult to<br />

compare published literature describing pathology,<br />

treatment and outcomes. A comprehensive<br />

biological classification system proposed in 1982<br />

correlated endothelial and mast cell kinetics of<br />

vascular lesions with their clinical characteristics<br />

and natural history. 1 With minor modifications,<br />

this classification system is now universally<br />

accepted by those at the forefront of this specialized<br />

field and is used to classify all vascular<br />

lesions in infancy and childhood.<br />

According to the current system, there are two<br />

main types of vascular lesion: tumors and malformations.<br />

Within the category of vascular tumors,<br />

hemangiomas are by far the most common.<br />

Lesions in the two classifications differ in their<br />

clinical, radiological, histopathological and hemodynamic<br />

presentations. 2,3 However, both types of<br />

lesion can permeate any organ system. Although<br />

they generally present cutaneously, they can also<br />

develop in multiple anatomic sites, and often<br />

involve the extremities, thoracoabdominal walls<br />

and cavities, solid organs, hollow viscera and<br />

brain. 4 Clinical symptoms are diverse, ranging<br />

from minor cervicofacial manifestations to serious<br />

and potentially life-threatening symptoms that<br />

may be difficult to identify (e.g. pain, bleeding,<br />

mass effect and congestive heart failure). Because<br />

management approaches and prognostic assessment<br />

for various lesions are different, accurate,<br />

precise and consistent terminology is essential.<br />

To clarify long-held misconceptions and to ensure<br />

that readers share common terminology and a<br />

common knowledge base, we will first describe the<br />

key clinical and biological features that distinguish<br />

hemangiomas from vascular malformations.<br />

Our discussion will then focus on the clinical<br />

presentation, diagnosis and management of vascular<br />

lesions of the gastrointestinal tract. We will<br />

briefly describe similar cutaneous vascular<br />

lesions. Since they are often associated with<br />

gastrointestinal manifestations, familiarity with<br />

them provides a useful framework for establishing<br />

a correct diagnosis.<br />

Clinical differences between vascular<br />

tumors and malformations<br />

Hemangiomas are benign tumors. They are characterized<br />

by endothelial hyperplasia with multilamination<br />

of the basement membrane and increased<br />

numbers of mast cells. After a period of rapid postnatal<br />

proliferation beginning shortly after birth,<br />

hemangiomas typically undergo slow, spontaneous<br />

involution. As involution is completed, mast cells<br />

fall to normal levels. Involution occurs in 50% of<br />

lesions by the age of 5 and in 70% by the age of 7.<br />

Other lesions continue to regress until the ages of<br />

10–12. Because of this clinical course, most<br />

hemangiomas do not require treatment.<br />

In contrast to hemangiomas, vascular malformations<br />

are not neoplastic lesions. Rather, they result<br />

from errors of vascular morphogenesis, and<br />

usually exhibit normal endothelial turnover and<br />

normal numbers of mast cells. Typical microscopic<br />

findings show progressive ectasia of structurally<br />

abnormal blood vessels. The blood-filled channels<br />

751


752<br />

Vascular lesions of the gastrointestinal tract in childhood<br />

are lined by a thin, flat endothelium, overlying a<br />

thin basal lamina. Additionally, there is a paucity<br />

or absence of smooth muscle within the vessel<br />

walls of vascular malformations. Although present<br />

at birth, these lesions may not manifest until childhood,<br />

adolescence, or even adulthood. Unlike<br />

hemangiomas, malformations generally grow<br />

commensurately with the child. They encompass a<br />

wide spectrum of anomalies, appearing with<br />

unique presentation and varying pathophysiology.<br />

Malformations are subcategorized by their<br />

predominant channel type as capillary, venous,<br />

arterial, lymphatic, or some combination of these.<br />

Lesions that have an arterial component are<br />

termed ‘fast-flow’ malformations, while capillary,<br />

venous and lymphatic lesions are slow-flow<br />

malformations. Fast-flow malformations are associated<br />

with high-output cardiac states, cardiac<br />

failure and disproportionate growth of involved<br />

organs and extremities.<br />

Whereas proliferating hemangiomas rarely cause<br />

bone distortion or hypertrophy, slow-flow malformations<br />

are frequently associated with diffuse<br />

bone hypertrophy, distortion, or elongation. Fastflow<br />

malformations can cause destructive<br />

interosseous changes.<br />

Vascular malformations also show hematological<br />

dissimilarities with vascular tumors. Hemangioendothelioma<br />

and tufted angioma, both of which<br />

are rare types of vascular tumors, can cause<br />

platelet trapping, shortened platelet half-life and<br />

profound thrombocytopenia. Vascular malformations,<br />

particularly the venous type, induce a true<br />

intravascular coagulation defect with only mild<br />

thrombocytopenia and slightly decreased platelet<br />

survival (Table 46.1).<br />

In most cases, the type of vascular anomaly can be<br />

correctly diagnosed by correlating physical<br />

Table 46.1 Clinical differences between vascular tumors and malformations (modified from<br />

reference 5)<br />

Vascular tumors Vascular malformations<br />

Neoplasm Congenital abnormality<br />

Generally apparent during first few weeks of life May not become apparent until months or even<br />

years after birth<br />

Female/male ratio of 3:1 No gender predilection<br />

Rapid postnatal growth followed by slow involution Slow steady growth; sepsis, trauma, or hormonal<br />

change may cause rapid expansion; no involution<br />

Endothelial cell hyperplasia Normal endothelial cell turnover<br />

Increased mast cells Normal mast cell count<br />

Multilaminated basement membrane Normal basement membrane<br />

Infrequent mass effect on adjacent bone; Slow-flow: frequent bone hypertrophy, distortion,<br />

hypertrophy rare or elongations<br />

Fast-flow: destructive interosseous changes<br />

Hemangioendothelioma and tufted angioma Mild thrombocytopenia and only slightly<br />

associated with platelet trapping and decreased platelet survival<br />

thrombocytopenia<br />

Angiographic findings: well circumscribed, Angiographic findings: diffuse, no parenchyma<br />

intense lobular–parenchymal staining with equatorial Low-flow: phleboliths, ectatic channels<br />

vessels High-flow: enlarged, tortuous arteries with<br />

arteriovenous shunting<br />

Response to corticosteroids and antiangiogenic agents No response to corticosteroids or antiangiogenic<br />

agents


findings and case history. Nevertheless, deep cutaneous<br />

and visceral lesions can be difficult to identify<br />

without radiological evaluation. Also, overlapping<br />

of clinical characteristics of the two<br />

classifications does occur, and because distinctive<br />

histopathological features of hemangiomas diminish<br />

during their evolution from the proliferative to<br />

the involutive phase, their differentiation from<br />

vascular malformations becomes increasingly<br />

difficult. 5 Doppler ultrasonography is often used to<br />

distinguish between these two types of lesion and<br />

to detect arterial flow. Depending on clinical<br />

circumstances, computerized tomography,<br />

magnetic resonance imaging, blood cell scintigraphy<br />

and angiography may also be required to<br />

determine the type, location and extent of lesions.<br />

Common errors in nomenclature<br />

Fishman and Fox 6 describe several important<br />

errors and inconsistencies in nomenclature that<br />

are readily apparent in the literature and are<br />

important for clinicians to be aware of. The term<br />

‘hemangioma’ is ubiquitously misused to label a<br />

wide spectrum of vascular lesions with different<br />

etiologies and natural histories. Moreover, some<br />

authors add misleading and incorrect descriptors<br />

such as ‘capillary’ or ‘cavernous’. Lesions with<br />

large blood-filled spaces, often referred to as<br />

‘cavernous hemangiomas’ are generally not<br />

hemangiomas, but are rather venous malformations.<br />

Also important to note, the suffix ‘oma’ in<br />

modern medical parlance implies a neoplastic<br />

process with rapid cellular turnover. Since vascular<br />

malformations are non-neoplastic lesions with<br />

quiescent endothelium, terms that are often seen<br />

in the literature such as ‘lymphangioma’, ‘cystic<br />

hygroma’, and ‘angioma’ are misnomers that<br />

should be avoided. Lesions that are commonly<br />

referred to as ‘lymphangiomas’, should be classified<br />

as lymphatic malformations. Cystic hygromas<br />

are more accurately classified as cystic lymphatic<br />

malformations, usually of the head and neck.<br />

Hemangiomas<br />

Hemangiomas are the most common tumor in<br />

infancy and are more common in girls than in<br />

boys, with a ratio of 3:1. They range in appearance<br />

Hemangiomas 753<br />

from small, hardly noticeable birthmarks to large<br />

grotesque tumors that can destroy tissue, obstruct<br />

the airway, or impair vision. Cutaneous hemangiomas<br />

generally become apparent early in the<br />

neonatal period, beginning as a faint discoloration.<br />

During proliferation, they typically turn bright red<br />

or crimson, and may ulcerate and/or bleed. After<br />

spontaneous regression, the lesion may become<br />

unnoticeable or may leave a fibrofatty residuum<br />

without much color. 6 A hemangioma that has<br />

ulcerated during proliferation can, however, leave<br />

wrinkling, telangiectasia, a yellowish inelastic<br />

patch, or a scar. 5<br />

Gastrointestinal hemangiomas can occur anywhere<br />

in the gastrointestinal tract. Although the liver is<br />

the most common visceral site, specific and<br />

adequate discussion of hepatic lesions is beyond<br />

the scope of the present chapter. Unlike cutaneous<br />

hemangiomas, gastrointestinal hemangiomas are<br />

rarely seen. They become evident only when<br />

lesions are symptomatic, and are often associated<br />

with multifocal cutaneous lesions (Figure 46.1).<br />

Mild to life-threatening gastrointestinal bleeding<br />

may occur, the later requiring repeated blood transfusions.<br />

Since an intestinal hemangioma rarely<br />

Figure 46.1 A 2-month old infant with multiple<br />

cutaneous hemangiomas.


754<br />

Vascular lesions of the gastrointestinal tract in childhood<br />

bleeds after the proliferative phase has ended,<br />

such a lesion would not, however, be a likely cause<br />

of bleeding beyond the first year or two of age.<br />

Another possible presenting symptom, though<br />

even less commonly seen, is intestinal obstruction.<br />

Treatment for hemangiomas is based on the degree<br />

and severity of symptoms. A small isolated hemangioma<br />

of the stomach or colon can be banded<br />

endoscopically, although not without risk of perforation<br />

or increased hemorrhage. 7 Occasionally, a<br />

focal lesion may be amenable to treatment with<br />

other endoscopic techniques such as multipolar<br />

thermocoagulation or argon plasma coagulation.<br />

For lesions visible by endoscopy, intralesional<br />

injection with corticosteroid may also be considered.<br />

6 Indications for excision include complications<br />

such as intussusception or obstruction,<br />

uncontrollable ulceration with hemorrhage or<br />

perforation, or infection (Figure 46.2). A surgical<br />

approach is not indicated when there is diffuse<br />

multifocal involvement of the gastrointestinal<br />

tract. Such lesions are more appropriately treated<br />

with corticosteroids, interferon α-2a, or other<br />

antiangiogenic agents (Table 46.2).<br />

In summary, if symptoms can be managed with<br />

persistent and aggressive blood replacement and<br />

pharmacotherapy, these are often the most prudent<br />

treatment strategies. Bleeding will diminish and<br />

cease after lesions enter the involution phase,<br />

which in many cases can be accelerated with<br />

angiogenesis inhibitors. Studies have shown that<br />

oral corticosteroids are effective within 2 weeks in<br />

one-third of patients; they yield an equivocal stabilizing<br />

response in one-third; and are ineffective in<br />

the remainder. 8 Although interferon α-2a induces<br />

regression in almost all patients, this process is<br />

less rapid than with corticosteroids. 9 Also, a<br />

number of studies 10–12 have indicated the development<br />

of spastic diplegia resulting from neurotoxic-<br />

Table 46.2 Pharmacotherapy options for problematic hemangiomas<br />

Figure 46.2 Jejunoileal hemangioma associated with<br />

significant gastrointestinal hemorrhage in a 5-month-old<br />

infant.<br />

ity during the course of interferon therapy; this is<br />

more commonly seen in infants younger than 1<br />

year of age. Although the condition generally<br />

reverses with the termination of therapy, careful<br />

monitoring of neurodevelopmental status is<br />

required. In light of the potential side-effects,<br />

interferon therapy is used only when hemangiomas<br />

are unresponsive to steroids or are life<br />

threatening.<br />

Vascular malformations<br />

Dosage Duration<br />

Gastrointestinal vascular malformations parallel<br />

the four main malformation groups described<br />

earlier: capillary, venous, lymphatic and arteriovenous<br />

(and complex combined lesions). Severity<br />

varies greatly both among malformations within<br />

each clinical group and among the four general<br />

group classifications.<br />

Oral prednisone 3-4mg/kg per day (initial dosage) gradual tapering over several months<br />

Interferon α-2a 1–3 million units/m 2 per day 12–18 months<br />

Vincristine 0.05mg/kg weekly 4–12 weeks


Capillary malformations<br />

Capillary malformations of the skin include port<br />

wine stain (also known as nevus flammeus) and<br />

congenital telangiectasias. These are present at<br />

birth and remain throughout life. With aging, the<br />

surface of the red vascular stain becomes raised<br />

and studded with nodular lesions. Lesions can be<br />

localized or can occur extensively on the face,<br />

trunk or limbs, and can be isolated or associated<br />

with other congenital malformations or vascular<br />

anomalies. Sturge–Weber syndrome consists of<br />

facial capillary malformation in association with<br />

ipsilateral leptomeningeal and ocular anomalies.<br />

Malformations on the trunk or extremities may<br />

coexist with venous and lymphatic abnormalities<br />

(Klippel–Trénaunay syndrome) (Figure 46.3). They<br />

may also overlie a deep arteriovenous malformation<br />

anywhere in the body. A facial lesion may<br />

indicate a unilateral arteriovenous malformation<br />

of the retina and intracranial optic pathway<br />

(Wyburn–Mason’s syndrome).<br />

Symptomatic capillary malformations of the<br />

gastrointestinal tract are extremely rare in childhood.<br />

Patients with hereditary hemorrhagic telangiectasia<br />

(Osler–Weber–Rendu syndrome) may<br />

experience gastrointestinal bleeding, but this<br />

rarely occurs before adulthood 13 and is generally<br />

preceded by pulmonary manifestations, which<br />

include arteriovenous malformations and cerebrovascular<br />

lesions. Diagnosis is usually established<br />

by endoscopic evaluation. Treatment for identified<br />

Figure 46.3 Complex combined capillary venous and<br />

lymphatic malformation involving the left lower extremity<br />

and trunk of an infant with Klippel–Trénaunay syndrome.<br />

Vascular malformations 755<br />

bleeding sites can readily be accomplished by<br />

endoscopic laser photocoagulation.<br />

Two published case reports 14,15 describe an association<br />

between hereditary hemorrhagic telangiectasia<br />

and juvenile polyposis. These reports are,<br />

however, inconclusive as to the actual cause of<br />

bleeding, since colonic polyps themselves are<br />

highly vascular and thus a common source of<br />

bleeding in childhood.<br />

Venous malformations<br />

These slow-flow lesions appear as a faint blue<br />

patch or a soft blue mass. Because of their<br />

coloration and soft, spongy consistency, they are<br />

frequently mislabeled as ‘cavernous hemangiomas’.<br />

Venous malformations are not tumors, do<br />

not regress spontaneously and do not respond to<br />

antiangiogenesis inhibitors. Lesions can be localized<br />

or extensive, and can range from miniscule to<br />

massive and disfiguring. Phleboliths are pathognomonic<br />

and, if present, can easily distinguish a<br />

venous malformation from a hemangioma. While<br />

cutaneous and soft tissue lesions are often asymptomatic,<br />

they may cause pain; this is probably due<br />

to congestion and/or low-grade thrombosis.<br />

Venous malformations are the most commonly<br />

symptomatic vascular anomalies of the gastrointestinal<br />

tract in childhood. 7 They generally<br />

present with either chronic or acute upper or<br />

lower gastrointestinal bleeding, which is often not<br />

identified until the patient is found to be severely<br />

anemic. Less commonly, malformations of the gut<br />

may cause pain or obstruction.<br />

Gastrointestinal lesions appear in the varying<br />

patterns described below. 5 Diagnostic and management<br />

approaches are best tailored according to<br />

these patterns.<br />

Focal venous malformations<br />

Focal venous malformations can occur anywhere<br />

in the gastrointestinal tract. These lesions may<br />

vary in size and may be mucosal, mural, or transmural.<br />

Endoscopic ultrasound may be helpful in<br />

making this determination. A small focal lesion<br />

that is clearly not transmural may be treated nonoperatively.<br />

Such a lesion can be managed with<br />

endoscopic sclerotherapy using sclerosing agents


756<br />

Vascular lesions of the gastrointestinal tract in childhood<br />

such as absolute ethanol or 1% sodium tetradecyl<br />

sulfate, which are generally effective in obliterating<br />

abnormal venous channels. Because of the<br />

danger of systemic intravascular migration of the<br />

sclerosing agent, clinicians should consider<br />

performing pretreatment angiography or a direct<br />

intralesional contrast study to identify any anomalous<br />

or large vessels communicating with the<br />

lesion. Another treatment option for small<br />

mucosal venous lesions is endoscopic banding. 7<br />

However, since perforations can occur after endoscopic<br />

treatment of small, unsuspected transmural<br />

lesions, it is best to provide for surgical back-up<br />

should complications arise. 5 Surgical excision is<br />

required for large or transmural gastrointestinal<br />

venous malformations.<br />

Multifocal venous malformations (blue rubber bleb<br />

nevus syndrome)<br />

Blue rubber bleb nevus syndrome is a rare multifocal<br />

syndrome of cutaneous and gastrointestinal<br />

venous malformations. Cutaneous lesions can be<br />

found over the entire body surface and usually<br />

become apparent by 2 years of age. They typically<br />

vary in size from 1–2mm to 2cm, and range in<br />

number from a single lesion to several hundred<br />

lesions that grow in proportion to the growth of the<br />

child. Skin lesions are most commonly flat or<br />

minimally raised and are deep blue or purple.<br />

They are often most dense on the plantar aspect of<br />

the feet. 5 Lesions may be tender to palpation and<br />

may become spontaneously painful. They may be<br />

partially emptied of blood if firm pressure is<br />

applied. 16<br />

Gastrointestinal manifestations can occur<br />

anywhere from the mouth to the rectum and may<br />

become symptomatic years after the appearance of<br />

cutaneous lesions. The number of intestinal<br />

lesions in a patient generally tends to parallel the<br />

number of cutaneous lesions, varying from a few<br />

to several hundred. Large gastrointestinal lesions<br />

may cause intussusception, resulting in the onset<br />

of acute abdominal pain and vomiting. Oral and<br />

esophageal lesions may compress the airway.<br />

Blood loss tends to be most prominent from small<br />

bowel lesions. Almost all patients have multiple<br />

lesions in the liver, but lesions can be present in<br />

the subcutaneous tissues, muscles, or almost any<br />

organ. A total body survey can be obtained using<br />

Tc-99m-labeled red blood cell scintigraphy. 5<br />

Characteristically, there is pooling of the Tc-99m<br />

red blood cells within the lesions. Lesions have a<br />

pathognomonic endoscopic appearance, presenting<br />

as discrete purple, berry-like protuberances<br />

from several millimeters to several centimeters in<br />

size. Most lesions are sessile. A broad rim of<br />

normal mucosa is often at the base of the lesion<br />

and encircles its reddish blue apex. 5<br />

Patients inevitably develop chronic gastrointestinal<br />

bleeding and anemia, beginning early in<br />

infancy or in young adulthood. They eventually<br />

require ongoing transfusions and iron replacement<br />

supplemented with the administration of erythropoietin.<br />

Melena is common. Some patients also<br />

experience intermittent bouts of severe abdominal<br />

cramping secondary to recurrent small-bowel<br />

intussusceptions. 5,16<br />

Treatment varies depending on the degree of<br />

gastrointestinal tract involvement and the severity<br />

of symptoms. Patients with mild or moderate<br />

anemia usually do not require transfusions or<br />

surgical intervention. Acid reduction therapy with<br />

proton pump inhibitors is used to reduce bleeding<br />

from gastric lesions. Patients are advised to eat a<br />

diet rich in iron and to take iron supplements as<br />

well as multivitamins. Also, to reduce the chance<br />

of trauma and bleeding from lesions in the left<br />

colon, stool softeners are sometimes advised.<br />

While patients with moderate anemia require<br />

occasional blood transfusions, they are treated<br />

similarly. Additionally, endoscopic ablation may<br />

be used to treat problematic gastric or colonic<br />

lesions. Patients with persistent or severe anemia<br />

require multiple interventions over time, including<br />

frequent blood transfusions, endoscopic ablation<br />

and surgical excision of small-bowel lesions.<br />

Octreotide is used as a temporizing treatment<br />

method to stabilize patients prior to surgery. 16<br />

Although there have been several published<br />

reports of treatment with pharmacological therapy,<br />

there is no sustained efficacy in their use. For<br />

patients with life-threatening bleeding that necessitates<br />

repeated transfusions, Fishman and Fox 6<br />

advocate surgical excision. Through a combined<br />

endoscopic and open surgical approach, these<br />

authors inspect the entire gastrointestinal tract.<br />

Small-intestinal lesions are identified and<br />

resected. If superficial, lesions of the esophagus,<br />

stomach, duodenum, colon and rectum are eradicated<br />

by endoscopic band ligation. If not amenable


to this approach, such lesions are surgically<br />

excised. The authors have reported that, with up to<br />

a 7-year follow-up, none of the nine patients who<br />

had undergone surgery developed anemia or<br />

required a transfusion after hospital discharge. 6<br />

Diffuse venous malformations<br />

Diffuse venous malformations involve large adjacent<br />

segments of bowel and extraintestinal structures,<br />

including mesentery, retroperitoneum,<br />

pelvic space, muscles, subcutaneous tissue and<br />

skin. Diffuse upper visceral malformations may<br />

warrant treatment only if there is symptomatic<br />

portal hypertension. Selective portosystemic<br />

shunts may be beneficial in that they reduce<br />

portal–venous pressure in the malformation.<br />

Malformations of the lower gastrointestinal tract<br />

typically extend from the anorectum proximally,<br />

involving the left colon or the entire colon. Most of<br />

these lesions are transmural and may extend into<br />

the pelvic structures. Imaging studies often reveal<br />

thickening of the colonic and rectal wall.<br />

Colonoscopic examination reveals engorged<br />

purple mucosa with varix-like projections. These<br />

lesions often cause chronic hemorrhage that<br />

requires transfusions throughout life. Endoscopic<br />

therapy is often futile and may exacerbate hemorrhage<br />

and cause perforation. Depending on the<br />

extent of involvement, a variety of surgical options<br />

are used. These include partial or subtotal colectomy<br />

with end ileostomy or colostomy, or colectomy<br />

with anorectal mucosectomy and endorectal<br />

pull-through. 17 In most cases, full-thickness resection<br />

of the rectum should be avoided because of<br />

the risk of uncontrollable hemorrhage from the<br />

extrarectal pelvic venous malformation.<br />

Lymphatic malformations<br />

Lymphatic malformations (often incorrectly<br />

referred to as ‘lymphangiomas’) include a wide<br />

spectrum of anomalies that can vary from simple<br />

cutaneous lymphatic lesions to severe lymphatic<br />

malformations that involve different organs and<br />

anatomic regions. These malformations can be<br />

seen in any anatomic region but are more<br />

commonly seen in rich lymphatic areas, such as<br />

the neck, axilla, mediastinum, groin and retroperitoneum.<br />

They present in many forms, from tiny<br />

cutaneous or mucosal blebs to large channel or<br />

Vascular malformations 757<br />

multilocular lesions. There are two morphological<br />

types of lymphatic malformation: microcystic and<br />

macrocystic. The former present as clear, tiny<br />

cutaneous vesicles that permeate the skin and<br />

muscles; these vesicles are often firm and may give<br />

the impression of a brawny edema. Macrocystic<br />

lesions are large, soft, smooth, translucent masses<br />

under normal or bluish skin. Capillary or venous<br />

malformations are frequently seen in association<br />

with lymphatic malformations.<br />

Mesenteric cysts<br />

Mesenteric cysts are rare macrocystic lesions that<br />

are thought to be lymphatic malformations (Figure<br />

46.4). They may be malformations of the lacteals<br />

and major mesenteric lymphatic vessels, including<br />

the cisterna chyli, or may be more diffuse, involving<br />

retroperitoneum and mesentery. Lesions are<br />

occasionally associated with chylous ascites and<br />

protein-losing enteropathy. Although they can be<br />

found anywhere from the duodenum to the<br />

rectum, they most commonly occur in the mesentery<br />

of the small bowel; the retroperitoneum is the<br />

second most frequent site. 18 Children tend to<br />

present acutely, requiring urgent admission and<br />

surgical intervention. Complaint at presentation is<br />

commonly abdominal pain due to hemorrhage or<br />

obstruction. Intestinal obstruction may occur from<br />

volvulus or acute enlargement from hemorrhage<br />

into the cyst, or from compression of the adjacent<br />

bowel. Diagnosis is primarily made with ultrasonography,<br />

often followed by a computed tomography<br />

scan to confirm the cystic nature of the<br />

lesions (Figure 46.5). Ultrasonography also<br />

Figure 46.4 A mesenteric cyst seen at laparotomy in a<br />

4-year-old with partial intestinal obstruction.


758<br />

Vascular lesions of the gastrointestinal tract in childhood<br />

Figure 46.5 Computed tomography scan demonstrating<br />

a mesenteric cyst (asterisk).<br />

provides preoperative measurements and is valuable<br />

for postoperative monitoring. When lesions<br />

are localized, small portions of the intestine can be<br />

resected with a good prognosis. In children in<br />

whom there is a diffuse process involving most of<br />

the intestinal mesentery, unroofing of major cysts<br />

to allow drainage of lymphatic fluid into the<br />

abdomen provides long-term relief of symptoms.<br />

In some patients with unremitting chylous ascites,<br />

oversewing of leaking retroperitoneal lymphatics<br />

is reportedly effective. 19 When there is respiratory<br />

compromise from massive chylous ascites or when<br />

other efforts are unsuccessful, peritoneovenous<br />

shunting is useful, although it carries significant<br />

risks. 20<br />

Arteriovenous malformations<br />

Arteriovenous malformations are fast-flow lesions<br />

in which there are usually many abnormal<br />

communications between abnormal arteries and<br />

veins that bypass the normal capillary bed. The<br />

length of channels between the arteries and the<br />

veins can vary from millimeters to centimeters,<br />

and convoluted or cavernous abnormal vascular<br />

structures may be intercalated between the arterial<br />

and venous ends of the malformations. 6<br />

Arteriovenous malformations can be present in<br />

any part of the body, including the upper and<br />

lower extremities, the head and neck, the gut, the<br />

liver, the lungs and the central nervous system.<br />

Cutaneous lesions become evident in infancy or<br />

childhood and are often initially mistaken for<br />

hemangioma or port wine stain. Puberty, pregnancy<br />

and trauma tend to trigger expansion,<br />

causing lesions to exhibit the cutaneous signs of its<br />

fast-flow nature. The skin becomes red or<br />

purplish, a mass appears beneath the vascular<br />

stain and there is local warmth, thrill and bruit. 4<br />

This inevitably results in ischemic skin changes,<br />

ulceration, intractable pain and intermittent bleeding.<br />

In severe cases, lesions can result in highoutput<br />

cardiac failure.<br />

Symptomatic arteriovenous malformations of the<br />

gastrointestinal tract are extremely rare in childhood.<br />

Typically, patients develop massive hematochezia<br />

or chronic melena with iron deficiency<br />

anemia. A case of intestinal perforation secondary<br />

to an arteriovenous malformation has also been<br />

reported. 21 As with cutaneous arteriovenous<br />

malformations, some patients present with pain<br />

and, in severe cases, with high-output cardiac<br />

failure. Endoscopic examination of the bowel may<br />

reveal a pulsatile vascular lesion. Endoscopic<br />

ultrasonography demonstrates high flow through<br />

the lesion.<br />

Traditional angiography or magnetic resonance<br />

imaging help in localizing the arteriovenous<br />

malformation and are essential for diagnosis and<br />

treatment planning. Angiography frequently shows<br />

characteristic enlargement and an increased<br />

number of arteries; on the venous phase, there is<br />

early venous return and venous dilatation in the<br />

region of the malformation.<br />

Embolization alone may lead to intestinal necrosis<br />

and perforation. Embolization combined with<br />

surgical excision is, however, the treatment of<br />

choice. Amenability of lesions to complete excision<br />

depends upon their extent and location. Specific<br />

localization in the gastrointestinal tract before<br />

resection can be enhanced by tattooing during<br />

selective angiography. 22 When resection is possible,<br />

it is curative. Proximal vessel ligation alone or<br />

embolization combined with incomplete surgical<br />

resection may result in the re-establishment of<br />

additional feeding vessels, and lead to re-expansion<br />

of the arteriovenous malformation. 5<br />

Complex combined vascular malformations<br />

While various combinations of anomalous vessel<br />

channel types can occur, the most common gross


vascular abnormalities to affect the gastrointestinal<br />

tract are evidenced in Klippel–Trénaunay<br />

syndrome. This condition results from a combination<br />

of capillary, lymphatic and venous malformations<br />

and is characterized by purple capillary<br />

stains, limb hypertrophy, congenitally abnormal<br />

veins and lymphatic vesicles that often protrude<br />

through the skin. It is not uncommon for lesions to<br />

extend into the pelvis, involving both the bowel<br />

and the bladder. As with lower visceral diffuse<br />

venous malformations, these lesions typically<br />

extend from the anus proximally to involve part of<br />

or the entire colon (Figure 46.6). 5 Colonoscopic<br />

Figure 46.6 Computed tomography scan in a patient<br />

with Klippel–Trénaunay syndrome and profuse rectal<br />

bleeding demonstrating a complex venous malformation<br />

(V) adjacent to the rectum (R).<br />

REFERENCES<br />

1. Mulliken JB, Glowacki J. Hemangiomas and vascular<br />

malformations in infants and children: a classification<br />

based on endothelial characteristics. Plast Reconstr Surg<br />

1982; 69: 412–422.<br />

2. Martinez-Perez D, Fein NA, Boon LM et al. Not all<br />

hemangiomas look like strawberries: uncommon<br />

presentations of the most common tumor of infancy.<br />

Pediatr Dermatol 1995; 12: 1–6.<br />

3. Meyer JS, Hoffer FA, Barnes PD. Biological classification<br />

of soft-tissue vascular anomalies: MR correlation. Am J<br />

Roentgenol 1991; 157: 559–564.<br />

4. Mulliken JB, Fishman SJ. Vascular anomalies:<br />

hemangiomas and malformations. In O’Neill JA, Rowe<br />

MI, Grosfeld JL et al., eds. Pediatric Surgery, 5th edn. St<br />

Louis, MO: Mosby, 1998: 1939–1951.<br />

5. Azizkhan RG. Vascular lesions in childhood. In Fischer<br />

JE, Baker RJ, eds. Mastery of Surgery, 4th edn.<br />

Philadelphia, PA: Lippincott, Williams & Wilkins, 2001:<br />

389–403.<br />

Summary 759<br />

appearances differ, showing varying degrees of<br />

mural thickening and vascular discoloration.<br />

Patients with severe chronic anemia due to hemorrhage<br />

or with recurrent cellulitis of the buttock or<br />

thigh require therapeutic intervention. Such infections<br />

are sometimes attributed to the translocation<br />

of enteric organisms from the feces through the<br />

abnormal mucosal barrier into the malformation.<br />

These symptoms may be minimized or eliminated<br />

by partial colectomy with a diverting colostomy, or<br />

by colectomy with anorectal mucosectomy and a<br />

coloanal pull-through procedure. 23 Extensive<br />

perineal malformations may preclude performing<br />

the pull-through procedure. 5<br />

Summary<br />

Although bleeding is the most common symptom<br />

of both vascular tumors and vascular malformations,<br />

diagnostic and treatment approaches are<br />

entirely different. Understanding the biological<br />

classifications and natural history of these lesions<br />

is thus essential. In light of the varied etiologies<br />

and complexity of vascular lesions, particularly<br />

those with visceral components, an individualized<br />

and interdisciplinary approach is frequently<br />

required. The importance of using current nomenclature<br />

to ensure optimal communication and<br />

understanding cannot be overemphasized.<br />

6. Fishman SJ, Fox VL. Visceral vascular anomalies.<br />

Gastrointest Endosc Clin North Am 2001; 11: 813–834.<br />

7. Fishman SJ, Burrows PE, Leichtner AM et al.<br />

Gastrointestinal manifestations of vascular anomalies in<br />

childhood: varied etiologies require multiple<br />

therapeutic modalities. J Pediatr Surg 1998; 33:<br />

1163–1167.<br />

8. Enjolras O, Riche MC, Merland JJ et al. Management of<br />

alarming hemangiomas in infancy: a review of 25 cases.<br />

Pediatrics 1990; 85: 491–498.<br />

9. Ezekowitz RA, Mulliken JB, Folkman J. Interferon alfa-<br />

2a therapy for life-threatening hemangiomas of infancy.<br />

N Engl J Med 1992; 326: 1456–1463.<br />

10. Vesikari T, Nuutila A, Cantell K. Neurologic sequelae<br />

following interferon therapy of juvenile laryngeal<br />

papilloma. Acta Paediatr Scand 1988; 77: 619–622.<br />

11. Worle H, Maass E, Kohler B et al. Interferon alpha-2a<br />

therapy in haemangiomas of infancy: spastic diplegia as<br />

a severe complication. Eur J Pediatr 1999; 158: 344.


760<br />

Vascular lesions of the gastrointestinal tract in childhood<br />

12. Barlow CF, Priebe CJ, Mulliken JB et al. Spastic diplegia<br />

as a complication of interferon Alfa-2a treatment of<br />

hemangiomas of infancy. J Pediatr 1998; 132: 527–530.<br />

13. Guttmacher AE, Marchuk DA, White RI Jr. Hereditary<br />

hemorrhagic telangiectasia. N Engl J Med 1995; 333:<br />

918–924.<br />

14. Ballauff A, Koletzko S. Hereditary hemorrhagic<br />

telangiectasia with juvenile polyposis – coincidence or<br />

linked autosomal dominant inheritance? Z Gastroenterol<br />

1999; 37: 385–388.<br />

15. Inoue S, Matsumoto T, Iida M et al. Juvenile polyposis<br />

occurring in hereditary hemorrhagic telangiectasia. Am J<br />

Med Sci 1999; 317: 59–62.<br />

16. Andersen JM. Blue rubber bleb nevus syndrome. Curr<br />

Treat Options Gastroenterol 2001; 4: 433–440.<br />

17. Fishman SJ, Shamberger RC, Fox VL et al. Endorectal<br />

pull-through abates gastrointestinal hemorrhage from<br />

colorectal venous malformations. J Pediatr Surg 2000;<br />

35: 982–984.<br />

18. Kurtz RJ, Heimann TM, Holt J et al. Mesenteric and<br />

retroperitoneal cysts. Ann Surg 1986; 203: 109–111.<br />

19. Unger SW, Chandler JG. Chylous ascites in infants and<br />

children. Surgery 1983; 93: 455–461.<br />

20. Chang JHT, Newkirk J, Carlton G et al. Generalized<br />

lymphangiomatosis with chylous ascites – Treatment by<br />

peritoneo-venous shunting. J Pediatr Surg 1980; 15:<br />

748–750.<br />

21. Munn J, Hussain AN, Castelli MJ et al. Ileal perforation<br />

due to arteriovenous malformation in a premature<br />

infant. J Pediatr Surg 1990; 25: 701–703.<br />

22. Frémond B, Yazbeck S, Dubois J et al. Intestinal vascular<br />

anomalies in children. J Pediatr Surg 1997; 32: 873–877.<br />

23. Telander RL, Ahlquist D, Blaufuss MC. Rectal<br />

mucosectomy: a definitive approach to extensive<br />

hemangiomas of the rectum. J Pediatr Surg 1993; 22:<br />

379–381.


47<br />

Introduction<br />

The role of minimally invasive<br />

surgery in pediatric<br />

gastrointestinal disease<br />

Stig Somme and Donald C Liu<br />

Traditionally, large incisions have been considered<br />

necessary for adequate surgical exposure.<br />

However, these wounds frequently result in significant<br />

perioperative stress and postoperative pain<br />

and morbidity, not to mention poor cosmesis. The<br />

concept of avoiding large incisions to perform<br />

invasive surgery forms the basis for the revolution<br />

of minimally invasive surgery. In this chapter, we<br />

focus on and describe minimally invasive procedures<br />

that are performed in infants and children<br />

with gastrointestinal disease.<br />

Background<br />

With advances in microchip technology and<br />

micro-instrumentation, as well as the development<br />

of sophisticated video equipment with supreme<br />

optics, the stage was set for the rapid development<br />

of minimally invasive surgery in the form of intracavitary<br />

endoscopic surgery, better known as<br />

laparoscopy in adults. The first significant minimally<br />

invasive surgery performed in adults was<br />

the laparoscopic cholecystectomy. 1 This sentinel<br />

event in general surgery, however, had little initial<br />

impact on the practice of pediatric surgery in children<br />

with gastrointestinal disease.<br />

Several factors are thought to be responsible for<br />

the slow development of the practice of minimally<br />

invasive surgery in children. Unfortunately, physicians,<br />

particularly surgeons, typically have underappreciated<br />

postoperative pain and surgical stress<br />

in young children, who essentially are unable<br />

adequately to verbalize their discomfort.<br />

Furthermore, the advantages of smaller incisions<br />

were underestimated by pediatric surgeons,<br />

because they already use ‘small incisions’ in their<br />

patients. However, in reality, when incision length<br />

is compared with body size, pediatric incisions for<br />

open procedures are proportional to adult incisions<br />

for comparable procedures.<br />

The need to develop endoscopic techniques<br />

further deters their use in children. Minimally<br />

invasive surgery demands that the surgeon acquire<br />

not only an entirely new set of surgical skills, but<br />

often requires an entirely different way of thinking<br />

about common pediatric surgical problems. The<br />

lack of depth perception and tactile sensation<br />

required during endoscopic procedures necessitate<br />

major changes in operative technique. The operative<br />

choreography for minimally invasive surgery<br />

often deviates from the routine fixated in the mind<br />

of the pediatric surgeon from years of traditional<br />

training. Increasingly, however, reports and the<br />

literature are documenting the safety, efficacy and<br />

cost-effectiveness of minimally invasive surgery in<br />

children and are showing a rapid evolution in<br />

instrumentation and the performance of such<br />

procedures. 2,3<br />

Minimally invasive surgery in children has<br />

become a highlight and focus of many general<br />

pediatric surgery practices across the world. At the<br />

present time, the number of minimally invasive<br />

procedures being performed for pediatric gastrointestinal<br />

diseases is growing rapidly. Current<br />

practice techniques for various pediatric gastrointestinal<br />

diseases are listed in Table 47.1 and are<br />

categorized under laparoscopy.<br />

Technical considerations<br />

The key concepts behind laparoscopic surgery are<br />

to obtain safe access to the abdominal cavity and<br />

create an ‘operating dome’ through establishment<br />

of pneumoperitoneum. Small trocars ranging<br />

from 2 to 12mm in size are subsequently placed<br />

761


762<br />

The role of minimally invasive surgery in pediatric gastrointestinal disease<br />

Table 47.1 Pediatric gastrointestinal diseases treatable via minimally<br />

invasive surgery<br />

Disease Laparoscopy<br />

Achalasia Heller myotomy<br />

Appendicitis appendectomy<br />

Chronic abdominal pain diagnostic laparoscopy<br />

Constipation appendicostomy (ACE procedure)<br />

Crohn’s disease enterectomy<br />

Gallstone disease cholecystectomy<br />

Gastroesophageal reflux disease fundoplication<br />

Hirschsprung’s disease endorectal pull-through<br />

Imperforate anus anal reconstruction<br />

Pyloric stenosis pyloromyotomy<br />

Ulcerative colitis colectomy, ileoanal reconstruction<br />

strategically to facilitate placement of miniaturized<br />

endoscopic instruments to perform surgery.<br />

Initial access to the peritoneum via the umbilicus<br />

is generally obtained by two different methods:<br />

direct puncture via the Veress needle or direct<br />

vision through the Hasson technique. 4 The skin is<br />

bluntly grasped, a small incision through the<br />

dermis made and the spring-loaded Veress needle<br />

advanced into the abdominal cavity. Gas insufflation<br />

is started using low gas flow and pneumoperitneum<br />

achieved. The Hasson technique mandates<br />

direct vision of the fascia and peritoneum before a<br />

blunt trocar is passed into the abdominal cavity,<br />

and is the technique preferred for patients with a<br />

history of previous abdominal surgery. Some<br />

surgeons advocate the use of the Hasson technique<br />

on a routine basis because of some support in the<br />

literature for a lowered risk of major complications<br />

compared with the Veress needle technique. 5<br />

The intra-abdominal pressure is adjusted according<br />

to the size of the patient with intra-abdominal<br />

pressures ranging from 5 to 15mmHg. 6 For<br />

example, in infants of


Figure 47.1 Laparoscopic Nissen fundoplication: 360º<br />

fundic wrap.<br />

Figure 47.2 Laparoscopic pyloromyotomy. The pylorus is<br />

incised sharply with a sheathed blade and the <strong>pylori</strong>c<br />

muscle is then carefully split and spread along the hypertrophic<br />

pylorus.<br />

fundoplication. 8,9 Importantly, strict postoperative<br />

dietary regimen enforcement and manipulation<br />

appears to contribute significantly to the avoidance<br />

of the necessity of esophageal dilatation in a<br />

population expected to have varying degrees of<br />

dysphagia secondary to the nature of the operation.<br />

Results in preventing GERD thus far have<br />

been excellent with recurrence rate less than 10%<br />

at 5 years, similar to if not in some cases lower<br />

than historical controls obtained from the ‘open’<br />

operation. 8–10 Longer follow-up, however, would<br />

be needed to establish the rate of recurrence in<br />

children undergoing laparoscopic fundoplication<br />

for GERD.<br />

Pyloric stenosis 763<br />

It is likely that new treatment modalities, that are<br />

even less invasive, will be used to a larger extent in<br />

the treatment of GERD. One such technique that<br />

has recently been introduced is Stretta. 11 This<br />

utilizes an endoscopic device with several perpendicular<br />

probes that are launched by the operator<br />

under videoscopic guidance. The probes are<br />

advanced into the mucosa of the lower esophageal<br />

sphincter and an electrical current is applied<br />

through the probes. This creates fibrosis in the<br />

area that, in cases of mild-to-moderate GERD, is<br />

enough to obviate gastroesophageal reflux. Data<br />

from the adult patient population indicates that<br />

Stretta is safe and that it relieves the symptoms of<br />

GERD. 12,13 No published follow-up data exists for<br />

pediatric patients.<br />

Pyloric stenosis<br />

In infants with <strong>pylori</strong>c stenosis, laparoscopic<br />

pyloromyotomy is performed through three puncture<br />

wounds (approximately 2mm in length each)<br />

in the upper abdomen. The pylorus is incised<br />

sharply with a sheath blade, and the <strong>pylori</strong>c<br />

muscle is then carefully split and spread along the<br />

length of the hypertropic pylorus (Figure 47.2).<br />

Children typically are discharged the following<br />

day with complete resolution of symptoms.<br />

Several studies have demonstrated the safety and<br />

efficacy of this procedure, although improvements<br />

in relevant surgical outcome parameters such as<br />

operating time and length of hospital stay have not<br />

been demonstrated when compared to the open<br />

procedure. 14,15 However, cosmesis is excellent and<br />

can be compared with that of an open pyloromyotomy<br />

(Figure 47.3).<br />

Inflammatory bowel disease<br />

Several series reporting the results of laparoscopic<br />

procedures for the treatment of inflammatory<br />

bowel disease have clearly demonstrated the feasibility<br />

of minimally invasive surgery in the pediatric<br />

population. Laparoscopic colectomy was first<br />

described in 1991 and, since then, laparoscopic<br />

colorectal procedures in adults ranging from<br />

simple ileocolic resection for Crohn’s disease to<br />

complex total colectomy with ileal/anal reconstruction<br />

for ulcerative colitis have been well


764<br />

The role of minimally invasive surgery in pediatric gastrointestinal disease<br />

(a) (b)<br />

Figure 47.3 Children who have undergone laparoscopic pyloromyotomy (a) and (b) typically are discharged the<br />

following day with complete resolution of symptoms. Cosmesis is excellent (b) and can be compared with that of an open<br />

pyloromyotomy (a).<br />

described. 16–18 However, acceptance of minimally<br />

invasive surgery as a standard surgical technique<br />

for Crohn’s disease remains controversial.<br />

Theoretically, the gross inflammation and phlegmon<br />

characteristic of Crohn’s disease can simulate<br />

landmines to the inexperienced laparoscopic<br />

surgeon. Nevertheless, these complications, plus<br />

their severe sequelae, can be effectively addressed<br />

by the careful, experienced laparoscopic surgeon.<br />

This, and the fact that Crohn’s disease affects a<br />

predominately younger group of patients who are<br />

likely to require re-resection, lends support to the<br />

application of minimally invasive surgery in<br />

Crohn’s disease. The pan-enteric and recurrent<br />

nature of Crohn’s disease renders a surgical cure<br />

impossible and results in re-operation in 70–90%<br />

of all patients and multiple procedures in more<br />

than 30%. 19 Ileocolic disease comprises the majority<br />

of surgical morbidity and predicts a high probability<br />

for surgical intervention. Furthermore,<br />

patients with ileocolic disease have the highest<br />

risk of recurrence, contributing to >40% probability<br />

for re-operation at 15 years. Laparoscopic ileocecectomy,<br />

the most common minimally invasive<br />

surgery procedure in children with Crohn’s<br />

disease, can be performed without complications<br />

and with excellent cosmesis (Figures 47.4 and<br />

47.5). 16 Furthermore, in children with intraabdominal<br />

abscesses, minimally invasive surgery<br />

techniques can be used successfully to drain the<br />

abscess, lyse the adhesions and also ascertain that<br />

the involved bowel was healthy enough according<br />

to the surgeon’s judgement to warrant or not<br />

warrant resection, keeping in mind that bowel<br />

preservation is the hallmark of surgery for Crohn’s<br />

disease. 20 Thus, minimally invasive surgery with<br />

its advantages of decreased postoperative pain<br />

from small wound sites, low incidence of hernias,<br />

better cosmesis and decreased rate of adhesive<br />

small-bowel obstruction compared with traditional<br />

laparotomy seem ideal in this patient population,<br />

who more often than not will require reoperation<br />

in the future. Laparoscopy reduces the<br />

need for surgical procedures not related to disease<br />

in this population and preserves the abdominal<br />

wall should stomas be needed in the future. Also,<br />

tiny, nearly invisible, strategically placed scars<br />

improve the perception of cosmesis in this<br />

younger population with already significant body<br />

image issues.<br />

Children with acute ulcerative colitis are generally<br />

referred for surgery on an emergency basis after<br />

failing to respond to aggressive medical therapy,<br />

including high-dose steroids and other immunosuppressants.<br />

17 In these often acutely ill children,<br />

we favor a staged surgical approach highlighted by<br />

initial subtotal colectomy and ileostomy followed<br />

by completion proctectomy and ileoanal pouch<br />

reconstruction several months later. From a


(a) (b)<br />

technical standpoint, a mini-Pfannenstiel incision<br />

of 2–3cm is a critical addition to our procedure<br />

because it facilitates safer dissection and subsequent<br />

transection at the rectosigmoid junction<br />

and, in addition, facilitates removal of the entire<br />

specimen. Laparoscopic subtotal colectomy in<br />

several series has been shown to be a safe procedure<br />

with acceptable blood loss. 17,21–27 Subsequent<br />

proctectomy and ileoanal reconstruction can be<br />

successfully completed with excellent results.<br />

The most common criticism of laparoscopic<br />

intestinal surgery in patients with inflammatory<br />

bowel disease has been increased operative time<br />

Appendicitis 765<br />

Figure 47.4 (a) Laparoscopically dissected diseased terminal ileum of Crohn’s disease; (b) exteriorized diseased bowel<br />

segment after laparoscopic dissection.<br />

Figure 47.5 Excellent cosmesis following laparascopic<br />

colectomy for Crohn’s disease.<br />

with resultant increased morbidity due to length of<br />

surgery. With increasing operative experience, it is<br />

fair to assume that operating times will decrease as<br />

surgeons gain experience and will be comparable<br />

to traditional open surgery.<br />

Although larger numbers of patients, longer-term<br />

follow-up, and a prospective, randomized,<br />

controlled study are necessary to determine the<br />

advantages of minimally invasive surgery in children<br />

with inflammatory bowel disease, preliminary<br />

results from several studies have demonstrated<br />

at the very least that minimally invasive<br />

surgery is a safe and viable surgical option. We are<br />

of the opinion that, with improving technology,<br />

increased surgeon expertise and better selection of<br />

patient populations, minimally invasive surgery<br />

may well become the standard approach when<br />

surgery for inflammatory bowel disease becomes<br />

necessary.<br />

Appendicitis<br />

Appendicitis is the most common diagnosis in<br />

children that requires emergency surgical intervention.<br />

Several randomized studies have<br />

compared open versus laparoscopic appendectomy<br />

and the results show a slight, but significant,<br />

reduction in postoperative wound infections. 28,29<br />

However, all other parameters compared, including<br />

length of stay, need for re-exploration, cost and<br />

pain showed no significant difference between the


766<br />

The role of minimally invasive surgery in pediatric gastrointestinal disease<br />

two groups. Some studies have even indicated that<br />

the laparoscopic/minimally invasive surgery operation<br />

takes more time. 2,30 It is apparent that, in<br />

children with appendicitis, the outcome depends<br />

mainly on the severity of the inflammation and the<br />

stage of the disease progression at which the<br />

surgery is performed. It is clearly less important<br />

whether the procedure is performed via minimally<br />

invasive or open techniques. There are, however,<br />

some patients who are clearly better served with a<br />

minimally invasive approach, in particular when<br />

the preoperative diagnosis is in question. The<br />

laparoscope provides the capability of looking at<br />

other organ systems, e.g. ovaries, Fallopian tubes<br />

and even the gallbladder, to investigate various<br />

pathologies. Patients who often fall into this category<br />

are adolescent females or obese patients.<br />

In cases where the appendix has perforated, as<br />

documented via radiological studies, a decision<br />

can be made regarding the practice of ‘interval’<br />

appendectomy. 29 In these cases intravenous antibiotics<br />

or a combination of intravenous and oral<br />

antibiotics are initiated in the patient and carried<br />

out for 4–6 weeks, at which time the appendix is<br />

subsequently removed (‘interval appendectomy’)<br />

on an out-patient basis via minimally invasive<br />

surgery.<br />

In summary, laparoscopic appendectomy is a good,<br />

albeit sometimes unnecessary, option in uncomplicated<br />

appendicitis. In the cases that are difficult<br />

to diagnose, and where exposure would be difficult,<br />

minimally invasive surgery is the technique<br />

of choice, resulting in better cosmesis.<br />

Chronic abdominal pain of unknown<br />

origin<br />

In children with complaints of non-specific<br />

abdominal pain and where an extensive work-up<br />

including abdominal ultrasonography, computerized<br />

tomography, upper gastrointestinal series<br />

and/or upper and lower endoscopy cannot identify<br />

any pathology, laparoscopic exploration of the<br />

abdomen has been shown to be useful. 31 The<br />

laparoscope provides an optimal tool for visualizing<br />

the entire abdominal cavity. Particularly in<br />

females, the adnexa and uterus can be inspected<br />

and manipulated atraumatically. The bowel can be<br />

inspected in its entire intraperitoneal length. In<br />

patients with Mediterranean fever, laparoscopic<br />

evaluation of the abdomen and elective appendectomy<br />

have been found useful, to rule out perforated<br />

appendicitis as a cause for peritonitis. Some<br />

surgeons believe laparoscopic exploration for illdefined<br />

abdominal pain or abdominal sepsis<br />

provides the ability to identify the problem and in<br />

most cases also the possibility of immediate repair.<br />

Importantly, unusual causes of abdominal pain<br />

such as Meckel’s diverticulum and related pathology<br />

(diverticulitis), obstruction, intussusception<br />

secondary to various lead points including<br />

Meckel’s, perforated duodenal ulcer, or inflammatory<br />

bowel disease, notably Crohn’s disease, can be<br />

discovered at laparoscopy and addressed from<br />

both a diagnostic and therapeutic perspective.<br />

Furthermore, unnecessary large disfiguring incisions<br />

can be avoided with the same postoperative<br />

result.<br />

Laparoscopic exploration of the abdomen, notably<br />

the right lower quadrant, can be performed in a<br />

variety of techniques as described in the literature.<br />

These vary specifically between series and authors<br />

and can be performed by one-, two- or three-port<br />

technique. 28,29 In all cases these ports can be<br />

placed in strategic positions where they are ‘nearly<br />

invisible’. The positive effect on the development<br />

of future small-bowel obstruction secondary to<br />

adhesions cannot be overemphasized, nor can the<br />

theoretical affect of preserving fertility secondary<br />

to minimizing adhesions in a female child.<br />

Cholecystectomy<br />

Etiologies for gallbladder disease in children are<br />

many and have been extensively reviewed elsewhere<br />

in this text. When cholecystectomy<br />

becomes necessary, laparoscopic cholecystectomy<br />

can be performed safely and effectively with excellent<br />

cosmetic results. Laparoscopic cholecystectomy<br />

has for more than a decade been considered<br />

a standard of care for removal of the gallbladder in<br />

uncomplicated cases. 1 Important adjuncts to<br />

biliary track surgery including performance of<br />

intraoperative cholangiogram and common bile<br />

duct exploration for choledocholithiasis can also<br />

be performed via minimally invasive techniques.<br />

Laparoscopic cholecystectomy is generally performed<br />

through four ports (2–5mm) and the gallbladder<br />

is removed through the umbilicus. Both


automatic and single reloadable stapling devices<br />

are used to ligate the cystic duct and artery.<br />

Important considerations in laparoscopic cholecystectomy<br />

include consideration of potential<br />

aberrant anatomy of the hepatic arteries specifically<br />

the right hepatic artery coming off the superior<br />

mesenteric artery (10% of occasions).<br />

Sometimes, the superior mesenteric artery can be<br />

mistaken for the cystic artery, especially in a small<br />

child. The greatest complication that can occur in<br />

laparoscopic cholecystectomy is injury to the<br />

common bile duct, which can be devastating,<br />

notably in a small child where biliary reconstruction<br />

can be prohibitive. 1 Thus, it is important to<br />

identify clearly the common bile duct, cystic duct<br />

and cystic artery prior to ligation and division of<br />

any of these structures. If there is any doubt as to<br />

the anatomy of the biliary tract, an intraoperative<br />

cholangiogram should be performed and conversion<br />

to laparotomy considered to prevent a devastating<br />

common bile duct injury.<br />

Constipation<br />

Constipation in children is 90% from functional<br />

causes versus 10% from mechanical causes.<br />

Hirschsprung’s disease is the most common surgical<br />

cause of constipation in children. Endorectal<br />

pull-through for Hirschsprung’s disease was first<br />

described by Soave in 1963. 32 In this technique, a<br />

ganglionic segment of colon is pulled down to the<br />

muscular cuff in the rectal canal after transabdominal<br />

mucosectomy of the distal sigmoid colon. The<br />

technique was modified by Boley, who performed<br />

a coloanal anastomosis to complete the operation.<br />

33 Common to these techniques is the fact that<br />

the rectal mucosectomy was performed via laparotomy.<br />

Today, advancements in minimally invasive<br />

techniques and instrumentation permit surgeons<br />

to mobilize the colon and perform a pull-through<br />

operation by minimally invasive techniques.<br />

Furthermore, rectal mucosectomy, aganglionic<br />

colectomy and normal ganglionic pull-through are<br />

performed by some surgeons entirely through the<br />

anus without the aid of laparotomy or laparoscopy,<br />

a truly minimally invasive approach. 32,33<br />

A critical step in any minimally invasive surgery<br />

technique for Hirschsprung’s disease is transanal<br />

mucosectomy. 34 Transanal mucosectomy provides<br />

several advantages, as follows. In open cases, it<br />

Constipation 767<br />

allows for shorter operating times, because a<br />

mucosectomy can be performed simultaneously<br />

and even completed during laparotomy. Transanal<br />

mucosectomy in conjunction with laparoscopic<br />

techniques theoretically confers all the advantages<br />

of laparoscopy versus laparotomy, including<br />

reduced hospital stay, less postoperative ileus and<br />

better cosmesis. Favorable postoperative outcomes<br />

with an acceptable rate of constipation, strictures,<br />

no reported incontinence of stools and an acceptable<br />

incidence of enterocolitis, the most significant<br />

complication of Hirschsprung’s disease, have been<br />

reported by several authors.<br />

In cases were children have been deemed to have<br />

functional constipation and where no surgical<br />

causes can be found, few surgical options exist.<br />

One option, however, that has been shown to be<br />

effective is the appendicostomy enema or so-called<br />

ACE procedure. 35 In this technique, the appendix<br />

is used as a conduit for daily enemas to relieve<br />

symptoms of constipation. This procedure can be<br />

performed laparoscopically; the appendix is mobilized<br />

and brought out to the skin and used effectively<br />

as the entry site for enemas. The minimally<br />

invasive techniques used to complete the ACE<br />

procedure allow for easier postoperative recovery,<br />

less postoperative ileus and, importantly, better<br />

cosmesis, especially in children who already have<br />

self-image issues, such as children with spinal<br />

dysraphism syndromes.<br />

Ladd’s procedure<br />

As part of the work-up for recurrent emesis in children,<br />

an upper gastrointestinal series is performed<br />

generally to assess for gastroesophageal reflux<br />

disease. Occasionally during these studies,<br />

however, malrotation is found to be present and<br />

thought to be the etiology. In order to address<br />

emesis surgically in cases of malrotation, and<br />

importantly lower the future incidence of midgut<br />

volvulus, an elective Ladd’s procedure is<br />

performed. Surgeons today have described<br />

performing this procedure through minimally<br />

invasive techniques where division of Ladd’s<br />

bands, appendectomy and placement of the cecum<br />

in the left side of the abdomen can be performed<br />

laparoscopically. 36 Clearly, the advantages of<br />

performing this procedure via minimally invasive<br />

techniques are less postoperative pain, shorter


768<br />

The role of minimally invasive surgery in pediatric gastrointestinal disease<br />

time until recovery from the postoperative ileus<br />

and a much improved cosmetic result. A longer<br />

follow-up period will be necessary to compare the<br />

incidence of midgut volvulus in the two groups<br />

receiving the open versus minimally invasive<br />

procedure.<br />

Complications<br />

In addition to the complications that are specific<br />

for each procedure, there are complications<br />

uniquely associated with laparoscopic procedures<br />

in general. 5 The most feared complications are the<br />

major vascular injuries. These injuries are not<br />

common, with an incidence of 0.05% in the adult<br />

literature, but have a high mortality when they<br />

occur, ranging from 8 to 17%. The most common<br />

major complication is perforation of hollow-organ<br />

viscera during insertion of the Veress needle or<br />

trocar, the most commonly injured being the small<br />

bowel. Dissection-related hollow- or solid-organ<br />

injuries may also occur, though with lower<br />

frequency. Most safety studies have clearly demonstrated<br />

a higher incidence of complications early<br />

in the learning curve of the surgeon, with significant<br />

decrease coinciding with increasing experi-<br />

REFERENCES<br />

1. Miller TA. Laparoscopic cholecystectomy; passing<br />

fantasy or legitimate treatment option? Gastroenterology<br />

1990; 5: 1527–1529.<br />

2. Ure BM, Bax NM, van der Zee DC. Laparoscopy in<br />

infants and children: a prospective study on feasibility<br />

and the impact on routine surgery. J Pediatr Surg 2000;<br />

35: 1170–1173.<br />

3. Reissman P, Durst AL, Rivkind A et al. Elective<br />

laparoscopic appendectomy in patients with familial<br />

Mediterranean fever. World J Surg 1994; 18: 139–141.<br />

4. Champault G, Cazacu F, Taffinder N. Serious trocar<br />

accidents in laparoscopic surgery: a French survey of<br />

103,852 operations. Surg Laparosc Endosc 1996; 6:<br />

367–370.<br />

5. Esposito C, Mattioli G, Monguzzi GL et al.<br />

Complications and conversions of pediatric<br />

videosurgery: the Italian multicentric experience on<br />

1689 procedures. Surg Endosc 2002; 16: 795–798.<br />

6. Bax NM, van der Zee. DC. Trocar fixation during<br />

endoscopic surgery in infants and children. Surg Endosc<br />

1998; 12: 181–182.<br />

ence. Most complications do not result in conversion<br />

to laparotomy (


13. Richards WO, Houston HL, Torquati A et al. Paradigm<br />

shift in the management of gastroesophageal reflux<br />

disease. Ann Surg 2003; 37: 638–649.<br />

14. Caceres M, Liu D. Laparoscopic pyloromyotomy:<br />

redefining the advantages of a novel technique. JSLS<br />

2003; 7: 123–127.<br />

15. Campbell BT, McLean K, Barnhart DC et al. A<br />

comparison of laparoscopic and open pyloromyotomy at<br />

a teaching hospital. J Pediatr Surg 2002; 37: 1068–1071.<br />

16. Falk PM, Beart RW Jr, Wexner SD et al. Laparoscopic<br />

colectomy: a critical appraisal. Dis Colon Rectum 1993;<br />

36: 28–34.<br />

17. Jacobs M, Verdeja JC, Goldstein HS. Minimally invasive<br />

colon resection (laparoscopic colectomy). Surg Laparosc<br />

Endosc 1991; 1: 144–150.<br />

18. Hunter JG. The case for fellowships in gastrointestinal<br />

and laparoendoscopic surgery. Surgery 2002; 132:<br />

523–525.<br />

19. Lock MR, Farmer RG, Fazio VW et al. Recurrence and<br />

reoperation for Crohn’s disease. A role of disease<br />

location in prognosis. N Engl J Med 1981; 304:<br />

1586–1588.<br />

20. Geis WP, Kim HC. Use of laparoscopy in the diagnosis<br />

and treatment of patients with surgical abdominal<br />

sepsis. Surg Endosc 1995; 9: 178–182.<br />

21. Gurland BH, Wexner SD. Laparoscopic surgery for<br />

inflammatory bowel disease: results of the past decade.<br />

Inflamm Bowel Dis 2002; 8: 46–54.<br />

22. Chen HH, Wexner SD, Iroatulam AJN et al.<br />

Laparoscopic colectomy compares favorably with<br />

colectomy by laparotomy for reduction of postoperative<br />

ileus. Dis Colon Rectum, 2000; 43: 61–65.<br />

23. Duepree HJ, Senagore AJ, Delaney CP et al. Advantages<br />

of laparoscopic resection for ileocecal Crohn’s disease.<br />

Dis Colon Rectum 2002; 45: 605–610.<br />

24. Milsom JW, Bohm B, Hammerhofer KA et al. A<br />

prospective, randomized trial comparing laparoscopic<br />

versus conventional techniques in colorectal cancer<br />

surgery: a preliminary report. J Am Coll Surg 1998; 187:<br />

46–57.<br />

References 769<br />

25. Paik PS, Beart RW Jr. Laparoscopic colectomy. Surg Clin<br />

North Am 1997; 77: 1–3.<br />

26. Proctor ML, Langer JC, Gerstle JT et al. Is laparoscopic<br />

subtotal colectomy better than open subtotal colectomy<br />

in children? J Ped Surg 2002; 37: 706–708.<br />

27. Ramos JM, Beart RW Jr, Goes R et al. Role of<br />

laparoscopy in colorectal surgery: a prospective<br />

evaluation of 200 cases. Dis Colon Rectum 1995; 38:<br />

494–501.<br />

28. Morfesis FA, Ahmad F. Use of laparoscopy in the<br />

treatment of acute and chronic right lower quadrant<br />

pain. J Ky Med Assoc 1996; 94: 16–21.<br />

29. Nowzaradan Y, Barnes JP Jr, Westmoreland J, Hojabri M.<br />

Laparoscopic appendectomy: treatment of choice for<br />

suspected appendicitis. Surg Laparosc Endosc 1993; 3:<br />

411–416.<br />

30. Lujan Mompean JA, Robles Campos R, Parrilla Paricio P<br />

et al. Laparoscopic versus open appendicectomy: a<br />

prospective assessment. Br J Surg 1994; 81: 133–135.<br />

31. Schier F, Waldschmidt J. Laparoscopy in children with<br />

ill-defined abdominal pain. Surg Endosc 1994; 8: 97–99.<br />

32. Georgeson KE, Inge TH, Albanese CT. Laparoscopically<br />

assisted anorectal pull-through for high imperforate<br />

anus – a new technique. J Pediatr Surg 2000; 35:<br />

930–931.<br />

33. Georgeson KE, Fuenfer MM, Hardin WD. Primary<br />

laparoscopic pull-through for Hirschsprung’s disease in<br />

infants and children. J Pediatr Surg 1995; 30:<br />

1017–1021.<br />

34. Liu DC, Rodriguez J, Hill CB, Loe WA Jr. Transanal<br />

mucosectomy in the treatment of Hirschsprung’s<br />

disease. J Pediatric Surg 2000; 35: 235–238.<br />

35. Lynch AC, Beasley SW, Robertson RW, Morreau PN.<br />

Comparison of results of laparoscopic and open<br />

antegrade continence enema procedure. Pediatr Surg Int<br />

1999; 15; 343–346.<br />

36. Bass KD, Rothenberg SS, Chang JH Laparoscopic Ladd’s<br />

procedure in infants with malrotation. J Pediatr Surg<br />

1998; 33: 279–281.


48<br />

Introduction<br />

Polyps and other tumors of the<br />

gastrointestinal tract<br />

Warren Hyer and John Fell<br />

Gastrointestinal polyps in children most<br />

commonly present with rectal bleeding and may<br />

have potential for malignant change if part of a<br />

polyposis syndrome. This chapter reviews the<br />

polyposis syndromes, the genetics of these conditions,<br />

their malignant potential and their management<br />

algorithms. Other tumors of the gastrointestinal<br />

tract are discussed.<br />

Gastrointestinal polyps<br />

Histopathological classification<br />

Gastrointestinal polyps in children fall into two<br />

major categories: hamartomas and adenomas<br />

(Table 48.1). Solitary polyps in children are most<br />

Table 48.1 Polyps and polyposis syndromes<br />

seen in childhood<br />

Adenomatous polyposis syndromes<br />

familial adenomatous polyposis<br />

Turcot’s syndrome<br />

Hamartomatous polyps<br />

solitary juvenile polyp<br />

juvenile polyposis syndrome<br />

Peutz–Jeghers syndrome<br />

Bannayan–Riley–Ruvalcaba syndrome<br />

Gorlins’s syndrome<br />

Cowden’s syndrome<br />

Lymphoid nodular hyperplasia<br />

Inflammatory polyps<br />

Mixed polyposis syndrome<br />

commonly hamartomas, predominantly of the<br />

juvenile type. Of the familial syndromes, familial<br />

adenomatous polyposis is more common than<br />

juvenile polyposis or Peutz–Jeghers polyposis.<br />

Table 48.2 outlines the different histological<br />

features.<br />

Clinical management<br />

The most common manifestation of a large-bowel<br />

polyp is painless rectal bleeding. Other symptoms<br />

attributed to polyps include abdominal pain,<br />

altered bowel habit, or prolapse of the polyp or<br />

rectum. Some children are completely asymptomatic<br />

– their polyps may be found as part of a<br />

screening program for a familial polyposis<br />

syndrome.<br />

Diagnosis is made at full colonoscopy and<br />

polypectomy. Apart from biopsying for histology,<br />

polypectomy serves to remove the symptomatic<br />

polyp, e.g. source of bleeding or intussusception<br />

(Table 48.3).<br />

In order to plan management appropriately, the<br />

pediatrician treating a child with a gastrointestinal<br />

polyp must know the histopathological type, the<br />

number and site of polyps, and whether or not<br />

there is a family history of polyps or colorectal<br />

cancer. The clinician should direct his history and<br />

examination towards complications related to<br />

polyposis syndromes (Table 48.4).<br />

Juvenile polyp (solitary at diagnosis)<br />

Clinical presentation and diagnosis<br />

Children with a solitary juvenile polyp present at<br />

a mean age of 4 years with painless rectal bleeding<br />

or peranal polyp protrusion. Up to 40% of children<br />

771


772<br />

Polyps and other tumors of the gastrointestinal tract<br />

Table 48.2 Pathological features of polyps seen in children<br />

Macroscopic Microscopic<br />

Polyp type appearance appearance Dysplasia<br />

Juvenile pedunculated dilated cysts filled rare, approximately<br />

1–3cm in size, rarely with mucin, abundant


Table 48.4 History and examination in a child<br />

with possible gastrointestinal (GI) polyps<br />

History<br />

Nature of bleeding and frequency<br />

Painful or painless rectal bleeding<br />

History of GI obstructive symptoms<br />

Detailed family history, exploring early deaths or<br />

diagnosis of GI cancer<br />

Weight loss, anorexia (tumor)<br />

Learning difficulties (JPS)<br />

Examination<br />

Mucosal pigmentation (PJS)<br />

Dysmorphic features (JPS)<br />

Edema (hypoalbuminemia in infantile JPS)<br />

Extraintestinal manifestations of FAP – see Table<br />

45.6 – e.g. subcutaneous cysts, exostosis,<br />

congenital hypertrophy of the retinal pigmented<br />

epithelium<br />

Hepatic mass (FAP)<br />

Thyroid mass (FAP or Cowden’s syndrome)<br />

JPS, juvenile polyposis syndrome; PJS,<br />

Peutz–Jeghers syndrome; FAP, familial<br />

adenomatous polyposis<br />

with a juvenile polyp have multiple polyps; 60%<br />

are proximal to the rectosigmoid, confirming the<br />

need for full colonoscopy even if a polyp is found<br />

in the rectum. 1 Although the risk of developing<br />

malignancy in a solitary juvenile polyp is very<br />

small, such polyps should be removed even when<br />

discovered incidentally by endoscopic polypectomy.<br />

If a polyp is found to be solitary after full<br />

colonoscopy, and there is no relevant family<br />

history, endoscopic polypectomy is sufficient<br />

treatment.<br />

Complications and follow-up<br />

The risk of malignant change for a solitary juvenile<br />

polyp is almost negligible, although neoplastic<br />

change has been documented. 2 In patients<br />

with a solitary juvenile polyp, there appears to be<br />

no increased risk of colorectal cancer or of dying<br />

as a result of the polyp. 3<br />

If the polyp was solitary and the patient is<br />

discharged, parents must be aware that juvenile<br />

polyps may be the first feature of juvenile polyposis.<br />

If fresh symptoms arise, the child should<br />

Gastrointestinal polyps 773<br />

be reinvestigated. When, however, there is a positive<br />

family history, or when multiple juvenile<br />

polyps are found, the possibility of juvenile polyposis<br />

syndrome (JPS) is raised and a different<br />

management strategy should be employed.<br />

Juvenile polyposis syndrome<br />

Clinical signs and diagnosis<br />

JPS is a rare autosomal dominant condition characterized<br />

by the occurrence of multiple juvenile<br />

polyps in the gastrointestinal tract. In children,<br />

the most common presentation is at a mean age of<br />

9 years with rectal bleeding, anemia or prolapse<br />

of either the polyp or the rectum. A significant<br />

proportion of patients with juvenile polyposis<br />

have been reported to have other morphological<br />

abnormalities, including digital clubbing, macrocephaly,<br />

alopecia, cleft lip or palate, congenital<br />

heart disease, genitourinary abnormalities or<br />

mental retardation. 4<br />

Compared to patients with solitary juvenile<br />

polyps, patients with JPS have more polyps, and<br />

are more likely to have right-sided polyps (hence<br />

the need for full colonoscopy), polyps with<br />

adenomatous change and anemia. 5 As the condition<br />

progresses, the number of polyps rises to<br />

50–200. Polyps are found primarily in the colon,<br />

but also in the stomach and small intestine. 6 The<br />

number of polyps needed to make the diagnosis<br />

remains controversial (between three and five). 2,7<br />

Less commonly, the condition presents in infancy<br />

with anemia and hemorrhage, diarrhea, proteinlosing<br />

enteropathy and rectal bleeding. 8 In this<br />

scenario the entire gastrointestinal tract is<br />

usually affected and there may be mild dysmorphic<br />

features (e.g. macrocephaly, clubbing, nail<br />

dystrophy); the prognosis appears to be related to<br />

the severity and extent of gastrointestinal<br />

involvement. The course in such infants is fulminant<br />

and death frequently occurs before the age<br />

of 2 years in severe cases despite colectomy. 9<br />

Genetics of juvenile polyposis<br />

A family history is found in 20–50% of patients,<br />

the inheritance apparently being autosomal dominant.<br />

There is evidence for genetic heterogeneity<br />

in JPS. Subsets of families have mutations in the


774<br />

Polyps and other tumors of the gastrointestinal tract<br />

tumor-suppressor gene PTEN located at 10q23.3,<br />

but these patients may have features suggestive<br />

of other polyposis syndromes, e.g. Cowden’s<br />

syndrome. 10,11 A proportion of JPS patients<br />

(especially those without stigmata of other polyposis<br />

syndromes) do not have germline mutations<br />

in PTEN – instead they have mutations in<br />

SMAD4/DPC4 located at 18q21. 12 It would appear<br />

that germline SMAD4 mutations predispose to<br />

hamartomas and cancer through disruption of<br />

the transforming growth factor (TGF)-β signaling<br />

pathway. Multiple genetic alterations (including<br />

APC mutations and K-ras mutations) appear to<br />

play a role in the neoplastic transformation of<br />

juvenile polyps of JPS patients.<br />

Complications<br />

There is little doubt that juvenile polyposis is a<br />

premalignant condition. There is a 15% incidence<br />

of colorectal carcinoma occurring in<br />

patients under the age of 35 years, leading to a<br />

cumulative risk of colorectal cancer of 68% by<br />

the age of 60 years. 7 Neoplastic changes have<br />

been documented both in the polyps and in flat<br />

apparently normal colonic mucosa. Malignancies<br />

in the stomach, duodenum and pancreas have<br />

been described in adults.<br />

Treatment and follow-up<br />

Patients with features suggestive of JPS should<br />

have surveillance colonoscopy with random<br />

biopsies of polyps and flat mucosa every 2 years<br />

(see Management algorithm, Figure 48.1). When<br />

the number of polyps is small, endoscopic<br />

polypectomy and follow-up may suffice, but it is<br />

not clear whether endoscopic surveillance is<br />

adequate to prevent malignancy. When there are<br />

numerous polyps, or symptoms such as bleeding<br />

and diarrhea persist, prophylactic colectomy<br />

should be considered after adolescence. As yet,<br />

there are insufficient data to justify prophylactic<br />

colectomy solely for the risk of colorectal carcinoma.<br />

With a risk of polyps in close relatives, firstdegree<br />

relatives of patients with JPS should be<br />

screened by colonoscopy starting at age 12 years,<br />

even when the subject is asymptomatic. 13<br />

Diagnosis of juvenile polyposis made at<br />

colonoscopy and histology<br />

Screen first-degree<br />

relatives<br />

Adequate control of<br />

poyp number and<br />

size – continue<br />

screening<br />

Peutz–Jeghers syndrome<br />

Correct anemia and<br />

malnutrition if relevant<br />

Colonoscopy and polypectomy<br />

at least every 2 years*<br />

If unable to control symptoms,<br />

e.g. bleeding or diarrhea,<br />

or polyps are too numerous and<br />

difficult to control via endoscopy<br />

Consider surgery – ileorectal<br />

anastomosis or proctocolectomy<br />

*Consider upper gastrointestinal surveillance<br />

Figure 48.1 Management of juvenile polyposis in<br />

childhood.<br />

Clinical features and diagnosis<br />

Peutz–Jeghers syndrome (PJS) is a rare autosomal<br />

dominant condition in which gastrointestinal<br />

polyps occur in association with macular melanin<br />

pigmentation. A definitive diagnosis of PJS can be<br />

made if a person fulfills one of the following criteria:<br />

(1) Two or more PJS polyps in the gastrointestinal<br />

tract;<br />

(2) One PJS polyp in the gastrointestinal tract,<br />

together with either the classical PJS pigmentation,<br />

or a family history of PJS.<br />

Presumptive diagnosis can be made in those with<br />

a positive family history and typical PJS freckling.<br />

14<br />

Polyps arise primarily in the small bowel, and to a<br />

lesser extent in the stomach and colon.<br />

Pigmentation occurs in most, although not all,


Figure 48.2 Macular pigmentation of the lips in a child<br />

with Peutz–Jeghers syndrome.<br />

patients and is seen most frequently on the lips and<br />

buccal mucosa (Figure 48.2) and occasionally on the<br />

hands, feet and eyelids. Clinicians should be aware<br />

that buccal mucosa pigmentation can occur in the<br />

normal population. The primary concern to the<br />

pediatrician is the risk of small-bowel intussusception<br />

causing intestinal obstruction (Figure 48.3),<br />

vomiting and pain. In addition, intestinal bleeding<br />

leading to anemia can occur.<br />

Genetics of Peutz–Jeghers syndrome<br />

The gene for this condition has recently been identified.<br />

The mutated gene LBK1/STK11 located on<br />

chromosome 19p13.3 is associated with PJS and<br />

encodes a serine/threonine kinase. 15,16 It appears<br />

that STK11 is a tumor-suppressor gene that might<br />

act as a gatekeeper regulating the development of<br />

hamartomas and adenocarcinomas in PJS. 17,18<br />

Allelic imbalance (allele loss and loss of heterozygosity)<br />

has previously been reported in a number of<br />

PJS cases, suggesting the existence of a hamartoma–carcinoma<br />

sequence in tumorigenesis. 19<br />

There is marked inter- and intra-family phenotypic<br />

variability in PJS. Therefore, the availability of<br />

predictive genetic testing may have some value, but<br />

cannot determine the likely severity of phenotype. 20<br />

In addition, not all PJS patients have the demonstrated<br />

LKB1/STK11 gene, 21 there remain potential<br />

loci for alternative mutated genes predisposing to<br />

PJS. If the gene mutation is known for previous<br />

affected cases in the family, it might have a role in<br />

presymptomatic testing in those patients with no<br />

pigmentation (or even potential prenatal diagnosis).<br />

Gastrointestinal polyps 775<br />

Figure 48.3 Small-bowel barium study showing a<br />

massive Peutz–Jeghers syndrome polyp in the second part<br />

of the duodenum.<br />

Management and complications<br />

Controversy exists over the management of the<br />

young child with mid-gut polyps. There is a high<br />

re-operation rate after initial laparotomy for smallbowel<br />

obstruction; this rate might be reduced in<br />

skilled hands by intraoperative enteroscopy<br />

(possibly through a surgical enterotomy) to remove<br />

other polyps. Intraoperative small-bowel<br />

endoscopy picked up 38% more polyps at laparotomy<br />

(17 additional polyps) compared with external<br />

palpation and small-bowel transillumination22 (Figure 48.4). Surgery for small-bowel complications<br />

in children with PJS is common (65% of<br />

patients


776<br />

Polyps and other tumors of the gastrointestinal tract<br />

Figure 48.4 A 2-cm pedunculated polyp in<br />

Peutz–Jeghers syndrome at resection after intraoperative<br />

endoscopic surveillance.<br />

Only small<br />

(< 5 mm)<br />

polyps<br />

present<br />

Investigate relevant symptoms (e.g. pain or<br />

anemia by upper and lower endoscopy, and<br />

barium FT/wireless capsule endoscopy<br />

Counsel parents,<br />

regular follow-up.<br />

Re-image according<br />

to symptoms<br />

Polyps<br />

5–10 mm,<br />

evaluate<br />

according to<br />

number<br />

Diagnosis made by phenotype<br />

or family history of PJS plus<br />

symptoms<br />

children who are asymptomatic with small polyps<br />

(less than 1.0cm in size), the parents should be<br />

counseled about the risk of intussusception. 24,25<br />

In order to reduce the need for repeat barium<br />

contrast studies in children, several centers are<br />

piloting the use of the wireless endoscopy capsule.<br />

This has identified polyps missed at contrast<br />

study, and may prove ideal for small-bowel<br />

surveillance in the future.<br />

Follow-up<br />

The risk of neoplasia is well documented in young<br />

adults and includes development of unusual<br />

tumors such as Sertoli cell tumor of the ovary and<br />

testicular tumors in prepubescent boys. 26 A metaanalysis<br />

of the existing literature to assess the risk<br />

of cancer in PJS identified a relative risk for all<br />

cancers in PJS patients (aged 15–64) of 15.2<br />

compared to the normal population, with tumors<br />

reported throughout the gastrointestinal tract, and<br />

extraintestinal tumors of the lung, testis, breast,<br />

uterus, ovary and cervix. 27 Clinicians caring for<br />

Large polyp > 1.5 cm,<br />

counsel and resect by<br />

polypectomy<br />

with intraoperative<br />

small-bowel endoscopy<br />

Presents with mid-gut<br />

intussusception<br />

Depending on symptoms (e.g.<br />

anemia or pain), carry out<br />

upper and lower endoscopy<br />

and barium FT/wireless<br />

capsule endoscopy at<br />

intervals (e.g. every 2 years)<br />

Refer for exploratory<br />

laparotomy with<br />

intraoperative<br />

endoscopy and<br />

polypectomy<br />

Figure 48.5 Management of Peutz–Jeghers syndrome (PJS) in childhood. Barium FT, barium follow-through.


adolescents with PJS should be aware of unusual<br />

symptoms, e.g. those due to a feminizing testicular<br />

tumor, and have a low threshold for investigating<br />

potential malignancies. A recommended screening<br />

program for PJS patients after adolescence is<br />

shown in Table 48.5, but it is not clear that such a<br />

program will reduce morbidity or mortality.<br />

Familial adenomatous polyposis<br />

In children, gastrointestinal adenomas are almost<br />

always associated with hereditary adenomatous<br />

polyposis syndromes. Therefore, whenever a<br />

colorectal adenoma is found in a child, total<br />

colonoscopy with dye spray is mandatory. Familial<br />

adenomatous polyposis (FAP) is the most common<br />

of the adenomatous polyposis syndromes.<br />

Clinical features<br />

Patients with FAP typically develop multiple<br />

adenomas throughout the large bowel, usually<br />

more than 100 and sometimes more than 1000<br />

(Figure 48.6). Polyps begin to appear in childhood<br />

or adolescence and increase in number with age.<br />

By the fifth decade colorectal cancer is almost<br />

inevitable if colectomy is not performed. Adult<br />

patients with FAP are also at increased risk of<br />

malignancies of the duodenum, ampulla of Vater,<br />

Table 48.5 Program for screening for<br />

malignancies in Peutz–Jeghers syndrome after<br />

adolescence<br />

Annual investigations<br />

Hemoglobin<br />

Pelvic ultrasound, gynecological and breast examinations<br />

(in females)<br />

Testicular ultrasound examinations (in males)<br />

Two-yearly investigations<br />

Upper and lower endoscopy<br />

Small bowel contrast X-ray<br />

Gastrointestinal polyps 777<br />

thyroid and pancreas. Papillary carcinoma of the<br />

thyroid has been reported in adolescence. 28<br />

Children under 5 years of age may develop hepatoblastoma.<br />

29 It may be advisable to measure<br />

serum α-fetoprotein levels and/or carry out<br />

abdominal ultrasound examination in children at<br />

risk.<br />

There are three ways in which a patient with FAP<br />

may present to the pediatrician.<br />

(1) Most will be called for screening because of a<br />

positive family history.<br />

(2) Some will present with colorectal symptoms<br />

such as bleeding or diarrhea.<br />

(3) A minority present with extracolonic manifestations<br />

(Table 48.6). General pediatricians<br />

treating a child with an unusual lesion such<br />

as maxillary osteoma or a rare tumor such as<br />

hepatoblastoma should consider the possibility<br />

of FAP, particularly if there are multiple<br />

tumors or another sibling is affected. 30<br />

Genetics of familial adenomatous polyposis<br />

The prevalence of FAP is estimated at 1:10000. It<br />

is inherited as an autosomal dominant trait with<br />

high penetrance but with a variable age of onset.<br />

The rate of spontaneous mutations is relatively<br />

high – reported as 10–30%. 31<br />

Other investigations<br />

Cervical smear 3-yearly<br />

Breast mammography 5-yearly from age 25 years Figure 48.6 Colectomy specimen demonstrating<br />

carpeting with dense polyposis in an adolescent with<br />

familial adenomatous polyposis.


778<br />

Polyps and other tumors of the gastrointestinal tract<br />

Table 48.6 Extracolonic manifestations of familial adenomatous<br />

polyposis in children and young adults<br />

Site Examples<br />

Bone osteomas, mandibular and maxillary<br />

exostosis<br />

sclerosis<br />

Dental abnormalities impacted or supernumerary teeth<br />

unerupted teeth<br />

Connective tissue desmoid tumors<br />

excessive intra-abdominal adhesions<br />

fibroma<br />

subcutaneous cysts<br />

Eyes congenital hypertrophy of the retinal pigmented<br />

epithelium<br />

Central nervous system glioblastomas, e.g. Turcot’s syndrome<br />

Adenomas stomach<br />

duodenum<br />

small intestine<br />

adrenal cortex<br />

thyroid gland<br />

Carcinomas thyroid gland<br />

adrenal gland<br />

Liver hepatoblastoma<br />

The gene responsible for FAP, APC (adenomatous<br />

polyposis coli), is located on chromosome 5q21<br />

and appears to be a tumor-suppressor gene. 32 Most<br />

mutations are small deletions or insertions that<br />

result in the production of a truncated APC<br />

protein. In FAP a germline mutation inactivates<br />

one of the two APC alleles that underlie the predisposition<br />

to adenoma formation.<br />

Mutations are widely distributed throughout the 5′<br />

half of the gene, although two ‘hot spots’ are found<br />

at codons 1061 and 1309 (Figure 48.7). These<br />

account for around one-third of all mutations<br />

detected and are associated with a more severe<br />

phenotype. Other phenotype–genotype correlations<br />

have been observed. 33 A variant of FAP has<br />

been described which is characterized by fewer<br />

colonic polyps and a generally milder phenotype,<br />

so-called attenuated FAP. 34,35 These correlations<br />

are not absolute and there may be considerable<br />

intrafamilial variation suggesting that there are<br />

other factors involved in the pathogenesis of the<br />

disease. Some of the phenotypic variability seen in<br />

patients cannot be explained by the location of<br />

their APC mutation. Environmental factors and<br />

other genes – often termed modifier genes – may<br />

have critical effects on APC function and disease<br />

expression. 36<br />

More than 300 different germline mutations have<br />

been described; finding the mutation may be a<br />

formidable task. Families need to be aware that the<br />

mutation may be detected in only 60–80% of cases.<br />

It is only in these cases that a predictive test can be<br />

offered to individuals at risk.<br />

Diagnosis – interpretation of the genetic test and<br />

clinical screening in familial adenomatous polyposis<br />

In order to define which screening protocol is<br />

appropriate for a given family, the first step is to<br />

determine, where possible, which mutation is<br />

present in the FAP-affected index case. At this<br />

stage, if a mutation cannot be found, the genetic


Attenuated<br />

APC<br />

168 1578<br />

Common mutations:<br />

0 400 800<br />

Classical/Servere FAP<br />

testing is non-informative and it will not be possible<br />

to offer predictive testing to asymptomatic atrisk<br />

relatives (Figure 48.8).<br />

For the 60–80% in whom a mutation is detected,<br />

at-risk relatives can be tested. A negative test is<br />

considered accurate in excluding FAP and the<br />

subject should be considered to hold an average<br />

population risk for the subsequent development<br />

of adenomas and cancer. Such genotype-negative<br />

individuals can be discharged from follow-up.<br />

Those patients where the deletion has not been<br />

sequenced, but has been excluded by linkage<br />

analysis with intragenic markers alone (a less<br />

reliable method for determining the risk of carrying<br />

the APC gene mutation) should not be<br />

discharged and they should undergo endoscopic<br />

surveillance.<br />

A positive test confirms the diagnosis of FAP and<br />

patients should undergo endoscopic assessment.<br />

The diagnosis is confirmed by finding polyps at<br />

flexible sigmoidoscopy, histologically confirmed<br />

as adenomas (Figure 48.9). Affected individuals<br />

undergo annual flexible sigmoidoscopy from the<br />

age of 10–14 years until adenomas are found. 37–39<br />

Most gene-positive children will have had a full<br />

colonoscopy by the age of 16 years to determine<br />

polyp density and location, and degree of dysplasia.<br />

1061 1068 1309<br />

APC GENE chromosome 5q21<br />

Exon 15<br />

Codon number<br />

Figure 48.7 Structure of the APC gene. FAP, familial adenomatous polyposis.<br />

Family<br />

genotype<br />

known,<br />

positive gene<br />

Positive family history –<br />

genetic counseling and<br />

detailed family pedigree<br />

Family<br />

genotype<br />

not known<br />

Start annual sigmoidoscopy<br />

from age 10–14, until rectal<br />

adenomas are confirmed<br />

Once adenomas found,<br />

counsel regarding timing for<br />

colectomy; consider<br />

colonoscopy wtih or<br />

without dye spray<br />

Gastrointestinal polyps 779<br />

1200 1600 2000<br />

Family genotype<br />

known,<br />

negative gene<br />

test<br />

Discharge<br />

No adenomas found –<br />

continue annual<br />

sigmoidoscopy, add in<br />

5-yearly colonoscopy<br />

Colectomy with IRA Restorative proctocolectomy<br />

6–monthly<br />

sigmoidoscopy and<br />

remove polyps of > 5 mm<br />

Attenuated<br />

APC<br />

Annual pouch<br />

examination<br />

Figure 48.8 Management protocol for screening<br />

children and adolescents at risk of familial adenomatous<br />

polyposis. IRA, ileorectal anastomosis.


780<br />

Polyps and other tumors of the gastrointestinal tract<br />

Figure 48.9 Endoscopic appearance of colon showing<br />

occasional adenomas in a 10-year-old with familial<br />

adenomatous polyposis. Dye spray emphasizes small<br />

polyps down to < 0.5 mm.<br />

In families in which the genotype is not known,<br />

protocols vary. The approach at St Mark’s Hospital<br />

is to perform annual sigmoidoscopy in all firstdegree<br />

relatives until adenomas are found. In<br />

addition, from the age of 20 years, colonoscopy<br />

with dye spray is performed at 5-yearly intervals.<br />

The presence on indirect ophthalmoscopy of more<br />

than four pigmented ocular fundus lesions –<br />

congenital hypertrophy of the retinal pigment<br />

epithelium – carries a 100% positive predictive<br />

value for FAP in families at risk, particularly if the<br />

lesions are large. The absence of pigmentation,<br />

however, is of no predictive value.<br />

No patient should undergo screening for FAP<br />

without detailed counseling. It is essential that<br />

the individual being screened understand the<br />

nature of the test and its possible outcomes. 40<br />

Issues such as emotional, family, insurance and<br />

employment implications of a positive result<br />

should be discussed prior to testing and there<br />

should be a clear protocol for post-test management.<br />

41<br />

Controversy surrounds the issue of screening in<br />

childhood for a condition that will cause few<br />

problems before adolescence. 42 Many authorities<br />

feel that the child should be involved in the decision-making<br />

process, and the diagnosis be<br />

delayed until the child is old enough to contribute<br />

to the screening program – for example, from the<br />

age of 11 years onwards. 43,44 Some children<br />

understand the genetic screening and its consequences<br />

at a younger age, e.g. 8 years; each family<br />

situation should be considered individually.<br />

Severe dysplasia and even malignancy have been<br />

documented in children with FAP under the age<br />

of 12 years. Consequently, those children from<br />

families in which severe dysplasia or carcinomas<br />

have been found at a young age should undergo<br />

screening at an earlier age. 45 This is particularly<br />

so if the family has one of the mutations associated<br />

with a severe phenotype, e.g. 1309 in exon<br />

15. Those children with symptoms such as diarrhea<br />

or rectal bleeding/anaemia should be investigated<br />

early, as this may signify severe colonic<br />

disease. 38<br />

Duodenal and ampullary cancers are currently the<br />

most common cause of cancer deaths in adults<br />

who have had a colectomy for FAP. Upper endoscopic<br />

surveillance of the stomach, duodenum<br />

and periampullary region with a side-viewing<br />

endoscope is recommended after the age 20 years,<br />

unless the patient has symptoms such as upper<br />

abdominal pain, which warrant earlier investigation.<br />

46<br />

Management of familial adenomatous polyposis<br />

Colectomy is the only effective therapy that eliminates<br />

the inevitable risk of colorectal cancer. In<br />

the absence of severe dysplasia, colectomy is<br />

usually performed in the mid- to late teens or<br />

early twenties to accommodate work and school<br />

schedules. Some clinicians advocate colectomy<br />

before puberty so that the patient can adapt to life<br />

without a colon before adolescence – each case<br />

should be considered on its own merits. Almost<br />

all FAP screen-detected adolescents are asymptomatic<br />

and may not contemplate interruptions in<br />

their schooling or effects on relationships. The<br />

surgical option, therefore, must not only be carefully<br />

timed but also have low morbidity and an<br />

excellent functional result.


The timing of primary preventative surgery may<br />

be influenced by knowledge of the mutation site<br />

and the likely severity of the polyposis. For<br />

example, patients with a deletion at exon 15 at<br />

codon 1309 may be offered earlier surgery since<br />

this phenotype is characterized by a large number<br />

of polyps and a higher risk of cancer. 47 Even<br />

though a low polyp density suggests a lower risk<br />

of developing malignancy, 48 it is unsafe to delay<br />

surgery on the grounds of polyp density alone. 49<br />

Surgical options include subtotal colectomy with<br />

ileorectal anastomosis, or restorative proctocolectomy<br />

with ileo-anal anastomosis (pouch procedure).<br />

The ileorectal anastomosis is a low-risk<br />

operation with good functional results, but the<br />

rectum remains at risk of cancer. Six-monthly<br />

surveillance of the rectum is needed postoperatively;<br />

despite this, inexperience can result in<br />

early cancers being missed. 50 A pouch procedure<br />

removes the colorectal cancer risk almost<br />

completely, but is more complicated than an ileorectal<br />

anastomosis, carrying a higher morbidity<br />

and often requiring a temporary ileostomy. The<br />

pouch procedure may carry a higher risk of<br />

complications, reoperations, longer hospital<br />

stays, night evacuation and influence on later<br />

sexual function. 51,52 Other authors have differing<br />

experience and claim acceptable morbidity<br />

following pouch construction for FAP 53,54 or total<br />

colectomy with rectal mucosectomy and straight<br />

endorectal pull-through. 55 Pouch creation is associated<br />

with an as yet unknown risk of pouch<br />

neoplasia; the pouch should be examined regularly.<br />

56<br />

The advantages of a pouch with a lower risk of<br />

cancer must be compared against the higher operative<br />

morbidity; the patients and parents must be<br />

carefully counseled. Conversion to an ileoanal<br />

pouch can be carried out when the patient is<br />

much older. 57 Patients with a large number of<br />

rectal polyps, or those with high-risk genotypes,<br />

may be better with a pouch procedure in the first<br />

instance, owing to the greater risk of malignancy.<br />

58–60<br />

Desmoid disease<br />

Desmoids are locally aggressive but non-metastasizing<br />

myofibroblastic lesions which occur with<br />

Gastrointestinal polyps 781<br />

disproportionate high frequency in patients with<br />

FAP. Associated etiological factors include<br />

germline mutation, estrogens and surgical<br />

trauma. 61 Desmoids occur most commonly in the<br />

peritoneal cavity (Figure 48.10), and may infiltrate<br />

locally leading to small-bowel, ureteric or<br />

vascular obstruction. These lesions may progress<br />

rapidly or may resolve spontaneously, their<br />

unpredictable nature making them difficult to<br />

treat. 62 Attempted surgical resection carries a high<br />

morbidity and mortality (10–60%) and usually<br />

stimulates further growth. 63 Medical treatments<br />

including non-steroidal anti-inflammatory drugs<br />

(NSAIDs) and antiestrogens have limited success.<br />

Cytotoxic chemotherapy is used as a last resort.<br />

Pediatricians treating children with extraintestinal<br />

desmoid tumors should consider the possibility<br />

of FAP in the family.<br />

Other polyposis syndromes<br />

Infrequently, juvenile polyposis may occur as part<br />

of the Bannayan–Riley–Ruvalcaba syndrome.<br />

This is characterized by macrocephaly, intestinal<br />

juvenile polyposis, pigmentation of the genitalia,<br />

psychomotor delay in childhood and occasionally<br />

lipid storage myopathy. 64 Cowden’s disease is the<br />

Figure 48.10 Abdominal computed tomography scan<br />

showing a massive intra-abdominal desmoid tumor in a<br />

14-year-old with familial adenomatous polyposis.


782<br />

Polyps and other tumors of the gastrointestinal tract<br />

association of multiple hamartomas of the<br />

stomach, small intestine or colon with macrocephaly,<br />

fibrocystic disease and cancer of the<br />

breast and thyroid. Germline mutations in the<br />

tumor-suppressor gene PTEN have been found in<br />

Cowden’s and Bannayan–Riley–Ruvalcaba pedigrees.<br />

65 Gorlin’s syndrome is an autosomal dominant<br />

condition comprising upper gastrointestinal<br />

hamartomas and pink or brown macules in<br />

exposed areas such as the face and hands. In addition,<br />

patients may have frontal and parietal<br />

bossing, hypertelorism and variable skeletal<br />

abnormalities and intracranial calcification and<br />

are at risk of medulloblastoma.<br />

Turcot’s syndrome is characterized by concurrence<br />

of a primary brain tumor and multiple<br />

colorectal adenomas. Patients with a polyposis<br />

syndrome and neurological symptoms should<br />

undergo thorough neurological examination and<br />

investigation for possible brain tumor. The<br />

management of the colonic polyps in Turcot’s<br />

syndrome is the same as for FAP.<br />

In the vast majority of patients with polyposis, a<br />

clear-cut diagnosis can be made, e.g. FAP or PJS.<br />

However, there are some rare cases where distinctions<br />

cannot be made on histology – the mixed<br />

polyposis syndromes. 66<br />

Future chemoprevention<br />

Studies have suggested that NSAIDs may be<br />

protective against colon cancer. NSAIDs inhibit<br />

prostaglandin synthesis via their effects on cyclooxygenase<br />

(COX). Several trials have shown<br />

regression of adenomas using the NSAID sulindac,<br />

67,68 but widespread use of sulindac has been<br />

limited by concerns regarding gastrointestinal<br />

side-effects with prolonged administration and<br />

case reports of rectal cancer despite treatment. 69<br />

Clinical trials using selective COX-2 inhibitors<br />

have reported a reduction in the number of<br />

colorectal polyps. 70 It remains to be seen what<br />

role this agent, and also the non-cyclo-oxygenase<br />

metabolite of sulindac, sulfone-sulindac, may<br />

have in the future management of colorectal and<br />

duodenal adenomas. They may have a role prior<br />

to surgery in young patients, but sulindac administered<br />

before polyps developed in genotype-positive<br />

adolescents did not prevent the development<br />

of adenomas. 71 Primary chemoprevention with an<br />

NSAID may not prevent polyp formation.<br />

Intestinal neoplasia<br />

In pediatric practice, colonic carcinoma and<br />

lymphoma are very rare, yet an awareness of these<br />

conditions is still important since a delay in diagnosis<br />

adversely affects outcome.<br />

Carcinoma of the colon<br />

Colorectal cancer is a major cause of morbidity<br />

and mortality in adult practice but its incidence in<br />

younger individuals is very low, with fewer than<br />

200 pediatric cases reported in the literature by<br />

1994. 72 Compared to adults, where sporadic cases<br />

of cancer predominate, in children there is an<br />

over-representation of cases with pre-existing<br />

polyposis or colitis (10%). 73<br />

Pathogenesis<br />

Colorectal carcinoma is believed to result from<br />

several sequential genetic mutations known as the<br />

adenoma–carcinoma sequence. Although the<br />

pattern of mutation is not consistent in all colorectal<br />

cancers, it is proposed that progress to malignant<br />

transformation occurs following the accumulation<br />

of a combination of four or five defects<br />

including mutational activation of oncogenes and<br />

inactivation of tumor-suppressor genes. 74,75<br />

Pathology<br />

The pathology of colonic carcinoma in children<br />

when compared to adult cases is characterized by<br />

more right-sided disease, with 50% located in the<br />

cecum to transverse colon. 76 Colorectal cancers<br />

grow locally, eventually penetrating the bowel<br />

wall. They can spread to regional lymphatics and<br />

subsequently to distant lymph nodes, and also to<br />

the liver, lungs and vertebrae. Cases are staged<br />

according to the degree of local spread and the<br />

presence or absence of transmural penetration, the<br />

degree of lymph node involvement and presence<br />

of distant metastases. In a review of younger<br />

patients with colonic cancer, 82% of these cases


had either distant metastases, lymph node involvement,<br />

or transmural penetrating disease, significantly<br />

higher than in adult patients. 77,78<br />

The usual histological appearance of colorectal<br />

cancers in adults is of a moderate to well differentiated<br />

adenocarcinoma. In children the proportion<br />

of tumors with mucinous (30%) and signet-ring<br />

(10%) histological appearances, associated with a<br />

poorer prognosis, is greater, and together with the<br />

tendency towards a more advanced stage at<br />

presentation may account for the poorer outcome<br />

of the disease in children.<br />

Clinical presentation<br />

Abdominal pain is the main presenting symptom,<br />

occurring in over 90% of cases, although other<br />

symptoms – weight loss, vomiting, rectal bleeding<br />

and altered bowel habit – may also be present. The<br />

clinical presentation is influenced by the site of<br />

the lesion, with constipation, obstruction and<br />

bleeding more common with left-sided disease,<br />

whereas right-sided disease may not present with<br />

obstruction, for example, until the lesion is larger,<br />

and a mass is palpable.<br />

Diagnosis is by colonoscopy in most cases,<br />

although the lesion can also be identified by other<br />

imaging modalities such as ultrasound or magnetic<br />

resonance imaging. Although serological markers<br />

such as carcinoembryonic antigen (CEA) are available,<br />

their role in establishing this rare diagnosis<br />

in children has not been fully evaluated. 79<br />

Treatment and outcome<br />

Surgery is the main treatment for colonic carcinoma,<br />

with complete resection of the primary<br />

lesion and draining lymphatics being the primary<br />

objective. Complete resection is often not possible<br />

in children, owing to the advanced stage at presentation.<br />

Chemotherapy for metastatic disease has in<br />

general been disappointing, and the role of radiotherapy<br />

or combination therapies awaits further<br />

study in the pediatric age group.<br />

The prognosis for children with colon cancer is<br />

poor, with 5-year survival rates of 10–20%<br />

reported. 80 These poor results are due to a combination<br />

of unfavorable histology and delayed<br />

presentation.<br />

Lymphoma<br />

Intestinal neoplasia 783<br />

Compared to adults, in younger patients there is a<br />

significant difference in the preponderance of the<br />

various histological types of lymphoma and also in<br />

the frequency of disease at different intestinal<br />

sites.<br />

Epidemiology and classification<br />

In the developed world, lymphoma accounts for<br />

10% of all cancers in children under 15 years of<br />

age. 81 In central Africa there is a very high incidence<br />

of Burkitt’s lymphoma (small non-cleaved<br />

B-cell lymphoma), accounting for up to 50% of all<br />

childhood cancers, whilst in North Africa and the<br />

Middle East there is a high rate of immunoproliferative<br />

small intestinal disease (IPSID).<br />

Etiology<br />

Non-Hodgkin’s lymphoma is associated in some<br />

cases with immunodeficiency syndromes, both<br />

inherited (e.g. ataxic-telangiectasia) and acquired<br />

(e.g. HIV infection), and also with immune<br />

suppressive therapy (e.g. after organ transplantation).<br />

An infectious agent (Epstein–Barr virus) may<br />

play a role in disease pathogenesis in some cases.<br />

This appears particularly to be the case for<br />

endemic Burkitt’s lymphoma, although the<br />

geographical association with malaria infection<br />

implies that co-infection may be important in<br />

promoting B-cell activation as part of the process<br />

leading to malignant change. Other conditions that<br />

give rise to chronic mucosal inflammation have<br />

also been linked to lymphoma. T-cell non-<br />

Hodgkin’s lymphoma is associated with celiac<br />

disease, whilst both lymphoma and adenocarcinoma<br />

are associated with both Crohn’s disease,<br />

where the lesion is typically in the small bowel,<br />

and ulcerative colitis. 82,83<br />

Pathology<br />

Intra-abdominal non-Hodgkin’s lymphoma in children<br />

typically is of an undifferentiated histological<br />

type – only around 50% have a primary intestinal<br />

origin84 – most common intestinal primary sites<br />

are the distal ileum, cecum and appendix. Bonemarrow<br />

involvement occurs in up to 40% of undifferentiated<br />

lymphoma, whilst central nervous


784<br />

Polyps and other tumors of the gastrointestinal tract<br />

system involvement is uncommon. In endemic<br />

Burkitt’s lymphoma 60% have abdominal disease,<br />

60% have characteristic involvement of the<br />

mandible. 84<br />

Clinical presentation<br />

Disease may present with non-specific symptoms<br />

such as abdominal distension, nausea, vomiting,<br />

altered bowel habit and abdominal pain. Disease<br />

primarily in the intestinal tract, typically localized<br />

to the distal ileum and cecum in children, may<br />

present with a mass, an ileo-cecal intussusception,<br />

obstructive symptoms or bleeding.<br />

The diagnosis of non-Hodgkin’s lymphoma depends<br />

on histological examination with immunophenotyping<br />

and cytogenetics. Staging of disease with<br />

chest and abdominal imaging, lumbar puncture and<br />

bone-marrow examination are also required, and<br />

REFERENCES<br />

1. Mestre JR. The changing pattern of juvenile polyps. Am<br />

J Gastroenterol 1986; 81: 312–314.<br />

2. Giardiello FM, Hamilton SR, Kern SE et al. Colorectal<br />

neoplasia in juvenile polyposis or juvenile polyps. Arch<br />

Dis Child 1991; 66: 971–975.<br />

3. Nugent KO, Talbot IC, Hodgson SV et al. Solitary juvenile<br />

polyps: not a marker for subsequent malignancy.<br />

Gastroenterology 1993; 105: 698–700.<br />

4. Desai DC, Murday V, Phillips RKS. A survey of phenotypic<br />

features in juvenile polyposis. J Med Genet 1998;<br />

35: 476–481.<br />

5. Hoffenberg EJ, Sauaia A, Malttzman T et al.<br />

Symptomatic colonic polyps in childhood: not so<br />

benign. J Pediatr Gastroenteral Nutr 1999; 28: 175–181.<br />

6. Desai DC, Neale KF, Talbot IC et al. Juvenile polyposis.<br />

Br J Surg 1995; 82: 14–17.<br />

7. Jass JR, Williams CB, Bussey HJR et al. Juvenile polyposis<br />

– a precancerous condition. Histopathology 1988; 13:<br />

619–630.<br />

8. Sachatello CR, Hahn IS, Carrington CB. Juvenile<br />

gastrointestinal polyposis in a female infant: a report of<br />

a case and review of the literature of a recently recognised<br />

syndrome. Surgery 1974; 75: 107–114.<br />

9. Scharf GM, Becker JHR, Laage NJ. Juvenile gastrointestinal<br />

polyposis or the infantile Cronkhite–Canada<br />

syndrome. J Pediatr Surg 1986; 21: 953–954.<br />

10. Lynch ED, Ostermeyer EA, Lee MK et al. Inherited<br />

mutations in PTEN that are associated with breast<br />

cancer, cowden disease, and juvenile polyposis. Am J<br />

Hum Genet 1997; 61: 1254–1260.<br />

may allow treatment to start if there is a delay in<br />

undertaking a laparotomy.<br />

Treatment and outcome<br />

Surgical resection of local disease is indicated<br />

where it is possible, although such fully resectable<br />

disease accounts for only 60–75% of cases. 84 The<br />

high mortality associated with intestinal perforation<br />

in more advanced disease has led to debate as to the<br />

role of partial resection as opposed to simple biopsy<br />

to establish histological type. Subsequent<br />

chemotherapy is required in all cases. For more<br />

localized disease the overall outcome is relatively<br />

good. In the 30% of childhood non-Hodgkin’s<br />

lymphoma cases with localized disease, cure<br />

rates as high as 95% have been reported<br />

following surgical resection and combination<br />

chemotherapy. 84<br />

11. Woodford-Richens K, Bevan S, Churchman M et al.<br />

Analysis of genetic and phenotypic heterogeneity in<br />

juvenile polyposis. Gut 2000; 46: 656–660.<br />

12. Howe JR, Roth S, Ringold JC et al. Mutations in the<br />

SMAD4/DPC4 gene in juvenile polyposis. Science 1998;<br />

280: 1086–1088.<br />

13. Stemper TJ, Kent TH, Summers RW. Juvenile polyposis<br />

and gastrointestinal carcinoma. A study of a kindred.<br />

Ann Intern Med 1975; 83: 639–646.<br />

14. Tomlinson IPM, Houlston RS. Peutz Jeghers syndrome. J<br />

Med Genet 1997; 34: 1007–1011.<br />

15. Hemminki A, Markie D, Tomlinson I et al. A serine/threonine<br />

kinase gene defective in Peutz–Jeghers syndrome.<br />

Nature 1998; 391: 184–187.<br />

16. Jenne DE, Reimann H, Nezu J et al. Peutz–Jeghers<br />

syndrome is caused by mutations in a novel serine threonine<br />

kinase. Nature Genetics 1998; 18: 38–43.<br />

17. Entius MM, Keller JJ, Westerman AM et al. Molecular<br />

genetic alterations in hamartomatous polyps and carcinomas<br />

of the patients with Peutz Jeghers syndrome. J<br />

Clin Pathol 2001; 54: 126–131.<br />

18. Gruber SB, Entius MM, Petersen GM et al. Pathogenesis<br />

of adenocarcinoma in Peutz–Jeghers syndrome. Cancer<br />

Res 1998; 58: 5267–5270.<br />

19. Wang ZJ, Ellis I, Zauber P et al. Allelic imbalance at the<br />

LKB1 (STK11) locus in tumours from patients with<br />

Peutz–Jeghers’ syndrome provides evidence for a hamartoma–(adenoma)–carcinoma<br />

sequence. J Pathol 1999;<br />

188: 9–13.


20. McGarrity TJ, Kulin HE, Zaino RJ. Peutz–Jeghers<br />

syndrome. Am J Gastroenterol 2000; 95: 598–604.<br />

21. Olschwang S, Markie D, Seal S et al. Peutz–Jeghers<br />

disease: most, but not all, families are compatible with<br />

linkage to 19p13.3. J Med Genet 1998; 35: 42–44.<br />

22. Spigelman AD, Thomson JPS, Phillips RKS. Towards<br />

reducing the relaparotomy rate in Peutz Jeghers<br />

syndrome: the role of a peroperative small bowel<br />

endoscopy. Br J Surg 1990; 77: 301–302.<br />

23. Hyer W, Fell JM, Philp C, Phillips RK. Peutz–Jeghers<br />

syndrome – a case for luminal surveillance in childhood.<br />

J Pediatr Gastroenteral Nutr 2002; 34: P452.<br />

24. Pennazio M, Rossini FP. Small bowel polyps in<br />

Peutz–Jeghers syndrome: management by combined<br />

push enteroscopy and intra operative enteroscopy.<br />

Gastrointest Endosc 2000; 51: 304–308.<br />

25. Edwards DP, Stafforton R, Phillips RKS et al. The long<br />

term results of polyp clearance in Peutz Jeghers<br />

syndrome by intra operative endoscopy. Abstract of the<br />

Third Joint Meeting, Leeds Castle Polyposis Group.<br />

Venice, Italy, 26–28 April 2001.<br />

26. Young RH, Welch WR, Dickerson GR, Scully RE.<br />

Ovarian sex cord tumour with annular tubules: review<br />

of 74 cases including 27 with Peutz–Jeghers syndrome<br />

and four with adenoma malignum of the cervix. Cancer<br />

1982; 50: 1384–1402.<br />

27. Giardiello FM, Brensinger JD, Tersmette AC et al. Very<br />

high risk of cancer in familial Peutz Jeghers Syndrome.<br />

Gastroenterology 2000; 119: 1447–1453.<br />

28. Bulow C, Bulow S. Is screening for thyroid carcinoma<br />

indicated in familial adenomatous polyposis? The Leeds<br />

Castle Polyposis Group. Int J Colorectal Dis 1997; 12:<br />

240–242.<br />

29. Bala S, Wunsch PH, Ballhausen WG. Childhood hepatocellular<br />

adenoma in familial adenomatous polyposis:<br />

mutations in adenomatous polyposis coli gene and p53.<br />

Gastroenterology 1997; 112: 919–922.<br />

30. Clark SK, Pack K, Pritchard J et al. FAP presenting with<br />

childhood desmoid tumours. Lancet 1997; 349: 471.<br />

31. Rustin RB, Jagelman DG, McGannon E et al.<br />

Spontaneous mutation in familial adenomatous polyposis.<br />

Dis Colon Rectum 1990; 33: 52–55.<br />

32. Kinzler KW, Nilbert MC, Su LK et al. Identification of<br />

FAP locus genes from chromosome 5q21. Science 1991;<br />

253: 661–665.<br />

33. Nugent KP, Phillips RK, Hodgson SV et al. Phenotypic<br />

expression in familial adenomatous polyposis: partial<br />

prediction by mutation analysis. Gut 1994; 35:<br />

1622–1623.<br />

34. Brensinger JD, Laken SJ, Luce MC et al. Variable phenotype<br />

of familial adenomatous polyposis in pedigrees<br />

with 3’ mutation in the APC gene. Gut 1998; 43:<br />

548–552.<br />

35. Lynch HT, Smyrk T, McGinn T et al. Attenuated familial<br />

adenomatous polyposis (AFAP). A phenotypically and<br />

genotypically distinctive variant of FAP. Cancer 1995;<br />

76: 2427–2433.<br />

36. Houlston R, Crabtree M, Phillips R et al. Explaining<br />

differences in the severity of familial adenomatous polyposis<br />

and the search for modifier genes. Gut 2001; 48:<br />

1–5.<br />

37. Vasen HF. When should endoscopic screening in familial<br />

adenomatous polyposis be started? Gastroenterology<br />

2000; 118: 808–809.<br />

38. Church JM, McGannon, Burke C et al. Teenagers with<br />

familial adenomatous polyposis. Dis Colon Rectum<br />

2002; 45: 887–889.<br />

References 785<br />

39. Church J, Burke C, McGannon E. Predicting polyposis<br />

severity by proctoscopy. Dis Colon Rectum 2001; 44:<br />

1249–1254.<br />

40. Giardiello F. Genetic testing in hereditary colorectal<br />

cancer. JAMA 1997; 278: 1278–1281.<br />

41. Giardiello FM, Brensinger JD, Petersen GM et al. The<br />

use and interpretation of commercial APC gene testing<br />

for familial adenomatous polyposis. N Engl J Med 1997;<br />

336: 823–827.<br />

42. Harper P, Clarke A. Should we test children for ‘adult’<br />

genetic diseases? Lancet 1990; 335: 1205–1206.<br />

43. Kodish ED. Testing children for cancer genes: the rule of<br />

the earliest onset. J Pediatr 1999; 135: 390–395.<br />

44. Hyer W, Fell J. Screening for familial adenomatous polyposis.<br />

Arch Dis Child 2001; 84: 377–380.<br />

45. Eccles DM, Lunt PW, Wallis Y et al. An unusually<br />

severe phenotype for FAP. Arch Dis Child 1997; 77:<br />

431–435.<br />

46. Working Group on Endoscopic and Diagnostic<br />

Techniques. World Congress of Paediatric<br />

Gastroenterology, Hepatology and Nutrition, 2000:<br />

205–218.<br />

47. Caspari R, Friedl W, Mandl M et al. FAP: mutation at<br />

codon 1309 and early onset of colon cancer. Lancet<br />

1994; 343: 629–632.<br />

48. Debinski HS, Love S, Spigelman AD et al. Colorectal<br />

polyp counts and cancer risk in FAP. Gastroenterology<br />

1996; 110: 1028–1030.<br />

49. Mills SJ, Chapman PD, Burn J, Gunn A. Endoscopic<br />

screening and surgery for familial adenomatous polyposis:<br />

dangerous delays. Br J Surg 1997; 84: 74–77.<br />

50. Vasen HF, Van Duijvendijk P, Buskens E et al. Decision<br />

analysis in the surgical treatment of patients with familial<br />

adenomatous polyposis: a Dutch–Scandinavian<br />

collaborative study including 659 patients. Gut 2001;<br />

49: 231–235.<br />

51. Madden MV, Neale KF, Nicholls RJ et al. Comparison of<br />

morbidity and function after colectomy and ileo rectal<br />

anastomosis or restorative proctocolectomy for FAP. Br J<br />

Surg 1991: 78; 789–792.<br />

52. Bjork J, Akerbrabt H, Iselius L et al. Outcome of primary<br />

and secondary pouch–anal anastomosis and ileorectal<br />

anastomosis in patients with familial adenomatous<br />

polyposis. Dis Colon Rectum 2001; 44: 984–992.<br />

53. Parc YR, Moslein G, Dozois RR. Familial adenomatous<br />

polyposis: results after ileal pouch–anal anastomosis in<br />

teenagers. Dis Colon Rectum 2000; 43: 893–902.<br />

54. Soravio C, Klein L, Berk T et al. Comparison of ileal<br />

pouch–anal anstomosis and ileorectal anastomosis in<br />

patients with familial adenomatous polyposis. Dis Colon<br />

Rectum 1999; 42: 1028–1034.<br />

55. Shilyansky J, Lelli JL, Drongowski R, Coran A. Efficacy<br />

of the straight endorectal pull-through in the management<br />

of familial adenomatous polyposis – a 16 year<br />

experience. J Pediatr Surg 1997; 32: 1139–1143.<br />

56. Wu J, McGannon BSW, Church JM. Incidence of<br />

neoplastic polyps in the ileal pouch of patients with<br />

familial adenomatous polyposis after restorative proctocolectomy.<br />

Dis Colon Rectum 1998; 41: 552–557.<br />

57. Soravia C, O’Connor BI, Berk T et al. Functional<br />

outcome of conversion of ileorectal anastomosis to ileal<br />

pouch–anal anastomosis in patients with familial<br />

adenomatous polyposis and ulcerative colitis. Dis Colon<br />

Rectum 1999; 42: 903–907.<br />

58. Cetta F, Gori M, Baldi C et al. APC genotype, polyp<br />

number, and surgical options in familial adenomatous<br />

polyposis. Ann Surg 1999; 229: 445–446.


786<br />

Polyps and other tumors of the gastrointestinal tract<br />

59. Vasen HF, van der Luijt RB, Slors JF et al. Molecular<br />

genetic tests as a guide to surgical management of FAP.<br />

Lancet 1996; 348: 433–435.<br />

60. Wu JS, McGannon PP, Church JM. APC genotype, polyp<br />

number, and surgical options in familial adenomatous<br />

polyposis. Ann Surg 1988; 227: 57–62.<br />

61. Farmer KCR, Hawley PR, Phillips RKS. Desmoid disease.<br />

In Phillips RKS, Spigelman AD, Thomson JPS, eds. FAP<br />

and other Polyposis Syndromes. London: Edward<br />

Arnold, 1994: 128–142.<br />

62. Clark SK, Smith TG, Katz DE et al. Identification and<br />

progression of desmoid precursor lesion in patients<br />

with FAP. Br J Surg 1998; 85: 970–973.<br />

63. Clark SK, Phillips RK. Desmoids in FAP. Br J Surg 1996;<br />

83: 1494–1504.<br />

64. Gorlin RJ, Cohen MM Jr, Condon LM, Burke BA.<br />

Bannyan–Riley–Rulcaba syndrome. Am J Med Genet<br />

1992; 44: 307–314.<br />

65. Liaw D, Marsh D, Li J et al. Germline mutations of the<br />

PTEN gene in Cowden disease, an inherited breast and<br />

thyroid cancer syndrome. Nature Genet 1997; 16: 64–67.<br />

66. Whitelaw SC, Murday VA, Tomlinson IPM. Clinical and<br />

molecular features of the hereditary mixed polyposis<br />

syndrome. Gastroenterology 1997; 112: 327–324.<br />

67. Giardiello FM, Hamilton SR, Krush AJ, et al. Treatment<br />

of colonic and rectal adenomas with sulindac in familial<br />

adenomatous polyposis. N Engl J Med 1993; 328:<br />

1313–1316.<br />

68. Hawk E, Lubet R, Limberg P. Chemoprevention in<br />

hereditary colorectal cancer syndromes. Cancer 1999;<br />

86 (Suppl): 2551–2563.<br />

69. Niv Y, Fraser GM. Adenocarcinoma in the rectal<br />

segment in familial polyposis coli is not prevented by<br />

sulindac therapy. Gastroenterology 1994; 107: 854–857.<br />

70. Steinbach G, Lynch PM, Phillips RKS et al. The effect of<br />

celecoxib, a cyclooxygenase-2 inhibitor, in familial<br />

adenomatous polyposis. N Engl J Med 2000; 342:<br />

1946–1952.<br />

71. Giardiello FM, Yang VW, Hylind LM et al. Primary<br />

chemoprevention of familial adenomatous polyposis<br />

with sulindac. N Engl J Med 2002; 346: 1054–1058.<br />

72. Steele GD. The national cancer data base report on<br />

colorectal cancer. Cancer 1994; 74: 1979–1989.<br />

73. Chabalko JJ, Fraumeni JF. Colorectal cancer in children:<br />

epidemiologic aspects. Dis Colon Rectum 1975; 18: 1–3.<br />

74. Midgley R, Kerr D. Colorectal cancer. Lancet 1999; 353:<br />

391–399.<br />

75. Ho JW, Yuen ST, Chung LP et al. Distinct clinical<br />

features associated with microsatellite instability in<br />

colorectal cancers of young patients. Int J Cancer 2000;<br />

89: 356–360.<br />

76. Griffin PM, Liff JM, Greenberg RS, Clark WS.<br />

Adenocarcinomas of the colon and rectum in persons<br />

under 40 years of age: a population-based study.<br />

Gastroenterology 1991; 100: 1033–1040.<br />

77. Enker WE, Palovan E, Kirsner JB. Carcinoma of the<br />

colon in adolescents: a report of survival and an analysis<br />

of the literature. Am J Surg 1977; 133: 737–741.<br />

78. LaQuaglia MP, Heller G, Filippa DA et al. Prognostic<br />

factors and outcome in patients 21 years and under<br />

with colorectal carcinoma. J Pediatr Surg 1992; 27:<br />

1085–1090.<br />

79. Angel CA, Pratt CB, Rao BN et al. Carcinoembryonic<br />

antigen and carbohydrate 19-9 antigen as markers for<br />

colorectal carcinoma in children and adolescents.<br />

Cancer 1992; 69: 1487–1491.<br />

80. Rao BN, Prall CB, Fleming ID et al. Carcinoma in children<br />

and adolescents. Cancer 1985; 55: 1322–1326.<br />

81. Shad A, Magrath I. Malignant non-Hodgkin’s<br />

lymphomas in children. In Pizzo PA, Poplack DG, eds.<br />

Principles and Practice of Pediatric Oncology, 3rd edn.<br />

Philidelphia: Lippincott-Raven, 1997: 545–587.<br />

82. Shepherd NA, Hall PA, Williams GT et al. Primary<br />

malignant lymphoma of the large intestine complicating<br />

chronic inflammatory bowel disease. Histopathology<br />

1989; 15: 325–337.<br />

83. Greenstein AJ, Mullin GE, Strauchen JA et al.<br />

Lymphoma in inflammatory bowel disease. Cancer<br />

1992; 69: 1119–1123.<br />

84. LaQuaglia MP, Stolar CJ, Krailo M et al. The role of<br />

surgery in abdominal non-Hodgkin’s lymphoma: experience<br />

from the children’s cancer study group. J Pediatr<br />

Surg 1992; 27: 230–235.<br />

85. Goldsby RE, Carroll WL. The molecular biology of pediatric<br />

lymphomas. J Pediatr Hematol Oncol 1998; 20:<br />

282–296.


Index<br />

abdominal migraine 218, 219<br />

abdominal pain see acute<br />

abdomen; chronic abdominal pain;<br />

functional abdominal pain<br />

abdominal wall defects 25–27<br />

exomphalos 26<br />

gastroschisis 26<br />

treatment 26–27<br />

abscess, intra-abdominal 682<br />

acetylsalicylic acid, gastric injury<br />

97–98<br />

achalasia 61–69<br />

epidemiology 61<br />

etiology 61–62<br />

investigations 64–66<br />

manometry 65, 66<br />

radiography 64–65<br />

pathology 62<br />

pathophysiology 62–63<br />

signs and symptoms 63–64<br />

treatment 66–69<br />

endoscopic botulinum toxin<br />

injection 67<br />

esophageal dilatation 67<br />

esophagomyotomy 67–69<br />

medical treatment 66<br />

acid reflux 42, 43<br />

see also gastroesophageal reflux<br />

disease<br />

acid secretion 77<br />

acute abdomen 677–687<br />

caused by procedures 684–687<br />

imaging studies 679–680<br />

laboratory tests 679<br />

specific diseases 681–684<br />

biliary diseases 682–683<br />

foreign body ingestion 681<br />

infectious and parasitic<br />

disease 683–684<br />

inflammatory bowel disease<br />

681–682<br />

pancreatitis 683<br />

symptoms and signs 677–679<br />

abdominal pain 677–678<br />

fever 678<br />

stool characteristics 678<br />

vomiting 678<br />

treatment considerations<br />

680–681<br />

acute appendicitis see appendicitis<br />

acyclovir, esophagitis treatment<br />

34, 35, 36<br />

adenocarcinoma<br />

cardia 47<br />

esophagus 47<br />

adenomas 771<br />

see also gastrointestinal polyps<br />

adenoviruses 138<br />

with HIV infection 117<br />

adsorbents 667–668<br />

Aichi virus 138–139<br />

AIDS (acquired immunodeficiency<br />

syndrome) 114–115<br />

esophagitis and 29, 30<br />

see also HIV infection<br />

alanine aminotransferase (ALT)<br />

394–395<br />

albendazole 181–182<br />

mass community treatment<br />

184, 185–186<br />

albumin 451–452<br />

alcohol, gastric pathology and 98<br />

Algicon 52<br />

allergic diseases 532–535<br />

allergic angiitis and<br />

granulomatosis 427<br />

allergic esophagitis 46<br />

dietary antioxidants and<br />

533–534<br />

dietary lipid mediators 533<br />

elimination diets 532<br />

modification of allergenic<br />

proteins 532–533<br />

probiotics and 534–535<br />

see also food allergies<br />

alosetron 51<br />

α2ß1 integrin 8<br />

α6ß4 integrin 8<br />

amebiasis 176–178<br />

clinical features 177–178<br />

diagnosis 178<br />

epidemiology 176–177<br />

pathophysiology 177<br />

prevention 178<br />

treatment 183, 184<br />

see also Entamoeba histolytica<br />

amino acid oral rehydration<br />

solutions 662<br />

aminosalicylic acid (ASA) drugs<br />

Crohn’s disease treatment 366,<br />

367<br />

indeterminate colitis<br />

management 382<br />

side-effects 368<br />

ulcerative colitis treatment<br />

398–400, 402<br />

colorectal cancer and 408<br />

amoxicillin<br />

H. <strong>pylori</strong> eradication 86–87<br />

malnutrition management 515<br />

amphotericin B, esophagitis<br />

treatment 32, 33<br />

amylase resistant starch oral<br />

rehydration solutions 663<br />

anal sphincter complex 249<br />

anaphylaxis 320, 322–323<br />

treatment 322–323<br />

Ancylostoma duodenale 168<br />

epidemiology 169<br />

see also hookworm<br />

Ancylostoma-secreted proteins<br />

(ASPs) 169<br />

anemia<br />

Crohn’s disease 354<br />

iron deficiency 354, 440<br />

H. <strong>pylori</strong> infection and 81<br />

malnutrition 500<br />

management 516<br />

pernicious 106, 110<br />

angiography 645–646<br />

anisakiasis 97, 684<br />

annular pancreas 16, 17<br />

anorectal congenital abnormalities<br />

23–24<br />

clinical features 23<br />

outcome 24<br />

treatment 23–24<br />

anorexia 493, 498<br />

antacids 51–52<br />

anti-Saccharomyces cerevisiae<br />

antibody (ASCA) 361, 381, 390<br />

anti-tumor necrosis factor-α agents<br />

370–371<br />

antibiotic treatment<br />

acute abdomen 680–681<br />

appendicitis 746–747<br />

Crohn’s disease 371<br />

787


788<br />

Index<br />

diarrhea 665, 666–667<br />

Campylobacter 150, 666<br />

cholera 147, 666<br />

Clostridium difficile 156, 667<br />

E. coli 152, 153, 666<br />

persistent diarrhea 196<br />

Salmonella 148, 149, 666<br />

Shigella 150, 666<br />

Yersinia 150–151, 666<br />

esophagitis 37<br />

H. <strong>pylori</strong> eradication 86–88<br />

malnutrition 515<br />

pouchitis 406<br />

short-bowel syndrome 470<br />

small-bowel bacterial<br />

overgrowth 209–210, 468<br />

ulcerative colitis 403<br />

see also specific drugs<br />

anticholinergics, abdominal pain<br />

management 226<br />

antiemetic drugs 665–667<br />

anti-endomysium antibodies 446<br />

antifungal drugs, esophageal<br />

candidiasis 33<br />

see also specific drugs<br />

antigliadin antibodies (AGA)<br />

445–446<br />

antimotility agents 156, 667<br />

abdominal pain management<br />

226<br />

short-bowel syndrome<br />

management 468–469<br />

antineutrophil cytoplasmic<br />

antibodies (ANCA) 421<br />

cytoplasmic (c-ANCA) 421<br />

perinuclear (P-ANCA) 361, 381,<br />

390, 421<br />

antioxidants, role in allergic<br />

diseases 533–534<br />

antiphospholipid antibody<br />

syndrome (APS) 431<br />

antisecretory drugs 668<br />

apnea 239<br />

appendectomy 746–747<br />

inflammatory bowel disease and<br />

351<br />

ulcerative colitis risk 387<br />

laparoscopic 765–766<br />

appendicitis 739–748<br />

clinical manifestations 741–742<br />

complications 747–748<br />

diagnosis 741<br />

differential diagnosis<br />

745–746<br />

epidemiology 739<br />

imaging studies 743–745<br />

laboratory tests 743<br />

pathological course 740–741<br />

phlegmonous appendicitis<br />

741<br />

simple appendicitis 741<br />

ulceronecrotic appendicitis<br />

741<br />

pathophysiology 740<br />

physical examination 742–743<br />

treatment 746–747<br />

laparoscopic procedures<br />

765–766<br />

appendix anatomy 739–740<br />

appetite<br />

anorexia 493, 498<br />

handicapped children 284<br />

in malnutrition 493, 498, 517,<br />

518<br />

arginine 626<br />

arteriography 426–427<br />

arteriovenous malformations 758<br />

arthralgia 440<br />

arthritis 440<br />

arthropathy 394<br />

ascariasis 166–168<br />

clinical features 167–168<br />

diagnosis 168<br />

epidemiology 166<br />

immune responses 166–167<br />

prevention 168<br />

treatment 182, 183, 185–186<br />

Ascaris lumbricoides 166<br />

life cycle 166<br />

see also ascariasis<br />

aspiration 46, 240, 485<br />

evaluation 242, 243<br />

see also dysphagia<br />

astrovirus 137<br />

virology 137<br />

with HIV infection 117<br />

atopic dermatitis, food allergy and<br />

325–327<br />

atresia<br />

biliary 21<br />

choanal 8<br />

duodenal 16<br />

esophageal 13–14<br />

jejunoileal 17–19<br />

<strong>pylori</strong>c 8<br />

autoimmune disease<br />

chronic intestinal<br />

pseudo-obstruction and<br />

275–276, 280<br />

see also celiac disease<br />

azathioprine<br />

Crohn’s disease treatment 369<br />

ulcerative colitis management<br />

402–403<br />

vasculitis treatment 432<br />

B-cell lymphoma 89<br />

baclofen 51<br />

bacterial infections 145<br />

appendicitis 740<br />

diarrhea 145, 656, 657<br />

evaluation 156<br />

treatment guidelines 156<br />

see also specific bacteria<br />

esophagitis 37<br />

gastritides 95–97<br />

gastrointestinal bleeding and<br />

643<br />

in malnutrition 514–515<br />

see also small-bowel bacterial<br />

overgrowth (SBBO);<br />

specific infections<br />

Bannayan–Riley–Ruvalcaba<br />

syndrome 781<br />

barium enema<br />

constipation 254<br />

Crohn’s disease 361<br />

Hirschsprung’s disease 261, 262<br />

intussusception 723–724<br />

ulcerative colitis 392<br />

barium swallow 64–65, 361<br />

Barrett’s esophagus 40, 46, 47<br />

Becker muscular dystrophy 276<br />

Behçet’s disease 428<br />

benzimidazoles 181–182<br />

Bifidobacterium lactis 210<br />

bile duct perforation 683<br />

bile reflux gastropathy 100<br />

bile salt malabsorption 356<br />

short-bowel syndrome 462<br />

biliary atresia 21<br />

biofeedback training, constipation<br />

management 256<br />

biopsy<br />

celiac disease 444–445<br />

rectal suction biopsy<br />

complications 686–687<br />

vasculitides 425–426<br />

biopsychosocial model 217–218<br />

bismuth subsalicylate 668<br />

bisphosphonates 359<br />

bleeding see gastrointestinal<br />

bleeding<br />

blue rubber bleb nevus syndrome<br />

756<br />

body mass index (BMI) 495–496<br />

bone disease<br />

in malnutrition 499–500<br />

parenteral nutrition-related 571<br />

see also osteopenia; osteoporosis<br />

botulinum toxin, achalasia<br />

treatment 67<br />

bradycardia 239<br />

brain–gut interaction 216<br />

breast feeding<br />

as model for optimal growth<br />

and development 528–529<br />

diarrhea management 663–664<br />

persistent diarrhea 198<br />

gut microbiota and 530<br />

inflammatory bowel disease<br />

and 349<br />

necrotizing enterocolitis and<br />

579<br />

breast milk 610–615<br />

benefits of 610–611<br />

fortification of 611–613<br />

limits of 611<br />

brush-border abnormalities 2


udesonide<br />

Crohn’s disease treatment<br />

368–369<br />

ulcerative colitis treatment 400,<br />

402<br />

Burkitt’s lymphoma 783, 784<br />

Cag-A protein 78, 79<br />

calcium channel blockers,<br />

achalasia treatment 66<br />

calcium requirements, premature<br />

infants 607–608, 630<br />

caliciviruses 139–140<br />

Campylobacter 150, 666<br />

cancer see specific forms of cancer<br />

cancrum oris 122<br />

Candida (candidiasis)<br />

esophageal infection 29–32<br />

diagnosis 30–32<br />

management 30, 31, 32, 33<br />

predisposing conditions 30<br />

symptoms 30, 31<br />

HIV infection and 117, 118–119<br />

malnutrition and 522<br />

capillary malformations 755<br />

carbohydrate intolerance 221–222<br />

lactose intolerance 195, 198,<br />

221–222<br />

carbohydrate malabsorption<br />

in HIV infection 116<br />

in small-bowel bacterial<br />

overgrowth 203<br />

carbohydrates<br />

in enteral nutrition 545<br />

premature infants 606–607<br />

in parenteral nutrition 559–560,<br />

568–569<br />

premature infants 622–623<br />

carcinoma<br />

colonic 782–783<br />

gastric, H. <strong>pylori</strong> infection and<br />

88–89<br />

cardia perforation 686<br />

cardiovascular disease 528<br />

carnitine 564, 629<br />

catheter-related sepsis 570, 633<br />

CCR5 co-receptor 114<br />

CD4 counts, in HIV infection<br />

116–117<br />

CD-10 immunoreactivity 3–4<br />

cecum 739–740<br />

celiac disease 334–335, 435–447<br />

associated diseases 441–442<br />

Down’s syndrome 442<br />

type 1 diabetes 442<br />

atypical/extraintestinal celiac<br />

disease 439–441<br />

arthritis/arthralgia 440<br />

dental enamel hypoplasia 440<br />

dermatitis herpetiformis<br />

439–440<br />

hepatitis/hypertransaminasemia<br />

440<br />

infertility 441<br />

iron-deficiency anemia 440<br />

neurological problems 441<br />

osteopenia/osteoporosis<br />

440–441<br />

psychiatric disorders 441<br />

short stature/delayed puberty<br />

440<br />

clinical presentations 438–439<br />

complications 442–444<br />

hyposplenism 443<br />

malignancy 444<br />

non-responsive disease 443<br />

refractory sprue 444<br />

diagnosis 444–446<br />

duodenal biopsy 444–445<br />

serology 445–446<br />

tissue transglutaminase<br />

antibodies 446<br />

differential diagnosis 223<br />

epidemiology 435–436<br />

pathophysiology 436–437<br />

treatment 446–447<br />

celiac gastritis 102–103<br />

central venous catheters (CVCs)<br />

557–558<br />

cereal-based oral rehydration<br />

solutions 662<br />

cerebral palsy 283, 285–286<br />

Chagas’ disease 280<br />

chloramphenicol, typhoid fever<br />

treatment 148, 149<br />

choanal atresia 8<br />

cholecystectomy 766–767<br />

cholecystokinin (CCK) 463–464<br />

choledochal cyst 21<br />

cholelithiasis 222, 462, 682<br />

cholera 145–148<br />

treatment 147<br />

vaccine development 147<br />

cholera toxin (CT) 147<br />

cholestasis, with parenteral<br />

nutrition 571–572<br />

premature infants 632–633<br />

chronic abdominal pain 213<br />

causes of 221<br />

laparoscopic procedures 766<br />

see also functional abdominal<br />

pain<br />

chronic granulomatous disease<br />

104, 110<br />

chronic intestinal pseudoobstruction<br />

(CIP) 269–281,<br />

702<br />

clinical presentation 269–270<br />

enteric myositis 275–276<br />

enteric nervous system<br />

disorders 276–280<br />

primary visceral neuropathies<br />

276–278<br />

sporadic visceral<br />

neuropathies 278–280<br />

etiology 272–275<br />

Index 789<br />

familial visceral myopathies<br />

273, 274–275<br />

infantile visceral myopathy<br />

273–274<br />

megacystis microcolon<br />

hypoperistalsis syndrome<br />

274<br />

primary visceral myopathies<br />

273<br />

intestinal smooth muscle<br />

disorders 275<br />

investigation 270–272<br />

manometry 271–272<br />

radiography 271<br />

surface electrogastrography<br />

271<br />

muscular dystrophy 276<br />

treatment 280–281<br />

chronic varioliform gastritis 102<br />

Churg–Strauss syndrome 427–428<br />

cidofovir, esophagitis treatment<br />

35, 36<br />

cigarette smoking see smoking<br />

cisapride<br />

cardiac effects 51<br />

gastroesophageal reflux<br />

disease treatment 50–51<br />

clarithromycin, H. <strong>pylori</strong><br />

eradication 86–88<br />

cleft lip and palate 13<br />

cloaca 23<br />

Clostridium spp 201, 667<br />

C. difficile 155–156<br />

cocaine ingestion 698<br />

colectomy<br />

laparoscopic 763–764, 765<br />

ulcerative colitis treatment 401,<br />

405<br />

colitis<br />

amebic 183<br />

indeterminate 379–383, 393<br />

diagnostic criteria 380–381<br />

epidemiology 379–380<br />

medical therapy 382–383<br />

natural history 381–382<br />

serologic markers 381<br />

surgical treatment 383<br />

necrotizing 177<br />

pseudomembranous (PMC) 155<br />

toxic 681–682<br />

see also enterocolitis;<br />

inflammatory bowel disease<br />

(IBD); ulcerative colitis<br />

collagenous gastritis 105–106<br />

colon<br />

carcinoma 782–783<br />

strictures 396<br />

see also colitis<br />

colonoscopy<br />

acute abdomen and 685<br />

Crohn’s disease 363<br />

gastrointestinal bleeding<br />

investigation 646


790<br />

Index<br />

ulcerative colitis 390–392<br />

colorectal cancer 782–783<br />

ulcerative colitis and 408<br />

computed tomography (CT)<br />

appendicitis 744–745<br />

Crohn’s disease 361, 362<br />

pancreas evaluation 310,<br />

311–313<br />

ulcerative colitis 392<br />

condyloma accuminata 122<br />

congenital biliary dilatation 21<br />

congenital diaphragmatic hernia<br />

24–25<br />

classification 24–25<br />

clinical features 25<br />

treatment 25<br />

connective tissue disorders 275,<br />

430<br />

constipation 247<br />

chronic 222<br />

clinical presentation 248–249<br />

defecation frequency<br />

248–249<br />

differential diagnosis 253, 745<br />

functional 247, 248<br />

handicapped children 287<br />

investigations 253–254<br />

abdominal X-ray 253–254<br />

anorectal manometry 254<br />

barium enema 254<br />

colonic manometry 254<br />

defecography 254<br />

medical history 252<br />

pathophysiology 250–252<br />

physical examination 252–253<br />

prognosis 256–257<br />

slow-transit constipation<br />

250–252<br />

treatment 254–256<br />

biofeedback training 256<br />

fecal impaction prevention<br />

256<br />

fecal impaction removal 255<br />

fluid intake 255<br />

laparoscopic procedures 767<br />

pharmacotherapy 255<br />

continuous enteral nutrition see<br />

enteral nutrition<br />

coronaviruses 139<br />

corticosteroids<br />

Crohn’s disease treatment<br />

366–369<br />

gastric injury 98<br />

indeterminate colitis<br />

management 382<br />

ulcerative colitis management<br />

400, 401–402<br />

vasculitis treatment 432<br />

Henoch–Schönlein purpura<br />

430<br />

polyarteritis nodosa 427<br />

corticotropin releasing factor (CRF)<br />

216<br />

role in cyclic vomiting<br />

syndrome 295<br />

costochondritis 223<br />

Cowden’s disease 781–782<br />

cow’s milk allergy 334<br />

modification of allergenic<br />

proteins 532–533<br />

cow’s milk-sensitive enteropathy<br />

(CMSE) 195–196, 325<br />

COX-2 inhibitors 109, 387<br />

Crohn’s disease (CD) 275,<br />

347–373, 388<br />

clinical signs and symptoms<br />

352–356<br />

complications 356–360<br />

hepatobiliary complications<br />

356<br />

local intestinal complications<br />

359–360<br />

malnutrition 356, 357–358<br />

metabolic bone disease 359<br />

nephrolithiasis 358<br />

pancreatic complications 358<br />

thromboembolic events<br />

358–359<br />

diagnosis 360–365, 380–381<br />

clinical suspicion 360–361<br />

endoscopic studies 363–365,<br />

390–392<br />

laboratory studies 361<br />

radiographic studies 361<br />

scintigraphy 361–363<br />

serological markers 361<br />

epidemiology 347–348<br />

etiology 348–351<br />

environmental factors<br />

349–351<br />

genetic factors 348–349<br />

follow-up management 371–372<br />

gastric 103–104, 110<br />

pathophysiology 351–352<br />

prognosis 373<br />

treatment 365–371<br />

5-aminosalicylic acid (5-ASA)<br />

drugs 366<br />

antibiotics 371<br />

corticosteroids 366–369<br />

enteral nutrition 541–542<br />

immunoregulatory agents<br />

369–371<br />

laparoscopic procedures<br />

763–764<br />

nutritional therapy 365–366<br />

supportive measures 371<br />

surgery 371<br />

see also inflammatory bowel<br />

disease (IBD)<br />

cryptosporidiosis 171–174<br />

clinical features 173<br />

diagnosis 173<br />

epidemiology 171–172<br />

pathophysiology 172–173<br />

prevention 173–174<br />

treatment 184, 667<br />

Cryptosporidium parvum<br />

HIV infection and 117–118,<br />

171, 172<br />

life cycle 171<br />

transmission 171–172<br />

see also cryptosporidiosis<br />

cutaneous lymphocyte antigen<br />

(CLA) 325<br />

cyclic vomiting syndrome (CVS)<br />

289–301<br />

clinical patterns 289–292<br />

cyclic versus chronic patterns<br />

291–292<br />

complications 297<br />

diagnosis 296–297, 298<br />

differential diagnosis<br />

292–293<br />

natural history 297<br />

pathophysiology 293–295<br />

autonomic dysfunction 295<br />

migraines 294<br />

mitochondrial dysfunction<br />

294–295<br />

neuroendocrine dysfunction<br />

295<br />

subtypes 295–296<br />

treatment 297–300<br />

cyclophosphamide 432<br />

Cyclospora cayetanensis 118, 171,<br />

180–181<br />

cyclosporin 401<br />

cyst<br />

choledochal 21<br />

duplication 24, 644<br />

mesenteric 757–758<br />

cystic fibrosis (CF) 306<br />

enteral nutrition and 542–543<br />

inguinal hernia and 592<br />

cytomegalovirus (CMV)<br />

esophageal infection 34–35<br />

treatment 35, 36<br />

following intestinal<br />

transplantation 706<br />

gastric infection 97<br />

HIV infection and 118, 119<br />

cytoplasmic antineutrophil<br />

cytoplasmic antibodies (c-<br />

ANCA) 421<br />

defecation<br />

frequency 248–249<br />

normal anatomy and physiology<br />

249–250<br />

dehydration 133, 657–658<br />

estimation of 659<br />

prevention in short-bowel<br />

syndrome 469<br />

with malnutrition 512<br />

see also rehydration therapy<br />

dendritic cells 113<br />

dental enamel hypoplasia 440<br />

dermatitis herpetiformis 439–440


desmoid disease 781<br />

diabetes mellitus, celiac disease<br />

and 442<br />

diaphragmatic hernia see<br />

congenital diaphragmatic hernia<br />

diarrhea<br />

bacterial 145, 656, 657<br />

evaluation 156<br />

small-bowel bacterial<br />

overgrowth 208<br />

treatment guidelines 156<br />

see also specific bacterial<br />

infections<br />

complications 657–658<br />

definitions 655–656<br />

diagnosis 658–660<br />

clinical features 658<br />

dehydration estimation 659<br />

differential diagnosis 658<br />

electrolyte measurement 660<br />

fecal screening tests 660<br />

microbiological testing<br />

659–660<br />

epidemiology 193, 656<br />

etiology 656<br />

Hirschsprung’s enterocolitis 23<br />

HIV infection and 115–117, 194<br />

HIV enteropathy 117<br />

host responses and<br />

susceptibility 116–117<br />

management 120–121<br />

mucosal structure/function<br />

and 115–116<br />

inflammatory 656–657, 660<br />

intestinal parasites and<br />

161–163<br />

see also specific parasites<br />

malnutrition and 193–194, 195,<br />

657–658<br />

management approach 655,<br />

660–669<br />

hospital admission 663<br />

nutritional management<br />

663–665<br />

pharmacological therapy<br />

665–669<br />

rehydration 660–663<br />

non-inflammatory 656<br />

pathogenesis 656–657<br />

persistent see persistent diarrhea<br />

phenotypic 9<br />

short-bowel syndrome 462, 465<br />

viral 127–141, 656, 657<br />

clinical signs and symptoms<br />

132–134<br />

diagnosis 134–135<br />

epidemiology 127<br />

pathogens and virology<br />

135–139<br />

pathophysiology 128–132, 133<br />

treatment 139<br />

vaccine development<br />

139–141<br />

see also intractable diarrhea of<br />

infancy (IDI);<br />

protracted diarrhea of<br />

infancy (PDI)<br />

diet see nutrition<br />

dietary deficiency 492<br />

see also malnutrition<br />

dietary intervention<br />

functional abdominal pain 226<br />

handicapped children 285–287<br />

malnutrition 513–514, 517,<br />

518–519<br />

persistent diarrhea 197–198<br />

short-bowel syndrome 467–468<br />

see also nutritional therapy<br />

Dieulafoy’s lesion 644<br />

dimethicone 52<br />

dioctahedral smectite 667–668<br />

diverticulitis 733<br />

see also Meckel’s diverticulum<br />

domperidone 50<br />

Down’s syndrome 283<br />

celiac disease and 442<br />

drooling 239<br />

Duchenne muscular dystrophy<br />

276<br />

duodenum<br />

atresia 16<br />

hematoma 686<br />

obstruction 16–17<br />

classification 16–17<br />

treatment 17<br />

stenosis 16–17<br />

ulceration<br />

H. <strong>pylori</strong> and 76<br />

see also peptic ulcer disease<br />

(PUD)<br />

duplication cysts 24, 644<br />

duplications of the alimentary tract<br />

24, 223<br />

dysentery 149–150<br />

amebic 177<br />

treatment 183<br />

Trichuris dysentery syndrome<br />

171<br />

see also diarrhea<br />

dysmotility 215<br />

allergic 327–328<br />

see also visceral myopathy;<br />

visceral neuropathies<br />

dyspepsia<br />

functional 218, 219<br />

H. <strong>pylori</strong> infection and 80–81,<br />

218, 220<br />

dysphagia 238<br />

achalasia and 63–64<br />

anatomic considerations 235<br />

clinical signs and symptoms<br />

239<br />

complications 238–240<br />

malnutrition 238<br />

respiratory complications<br />

239–240<br />

Index 791<br />

sialorrea 239<br />

diagnosis 240–243<br />

fiberoptic endoscopy 243<br />

history 240–241<br />

pharyngeal manometry<br />

242–243<br />

physical evaluation 241<br />

radiographic assessment<br />

241–242<br />

scintigraphy 243<br />

ultrasonography 242<br />

differential diagnosis 233, 234<br />

epidemiology 233<br />

etiology 233<br />

pathophysiology 233–235<br />

prognosis 244<br />

treatment options 243–244<br />

enteral nutrition 543<br />

eating disorders see feeding<br />

disorders<br />

eczema, food allergy and 325–327<br />

edema, in malnutrition 496–497<br />

see also kwashiorkor<br />

egg allergy 334<br />

eicosapentaenoic acid (EPA) 605<br />

electrogastrography (EGG) 271<br />

electrolyte measurement 660<br />

encopresis 248<br />

endometriosis 223<br />

endoscopic retrograde cholangiopancreatography<br />

(ERCP)<br />

310–314, 683<br />

complications 686<br />

endoscopy<br />

abdominal pain investigation<br />

224<br />

achalasia 65<br />

botulinum toxin injection 67<br />

acute abdomen causation<br />

684–686<br />

Crohn’s disease diagnosis<br />

363–365, 390–392<br />

gastroesophageal reflux disease<br />

54<br />

gastrointestinal bleeding<br />

investigation 646<br />

lymphonodular hyperplasia<br />

479–480<br />

portal hypertension<br />

management 650–651<br />

swallowing disorder evaluation<br />

243<br />

ulcerative colitis evaluation<br />

390–392<br />

vasculitides 425<br />

endothelin signalling pathway 266<br />

energy expenditure (EE) 558–559<br />

enteral nutrition effects 540<br />

energy requirements 558–559<br />

premature infants 603, 622<br />

protein/energy ratio 603–604,<br />

623


792<br />

Index<br />

reduction in malnutrition<br />

500–501<br />

Entamoeba dispar 176–177<br />

Entamoeba histolytica 176, 667<br />

acute abdomen and 684<br />

epidemiology 176–177<br />

life cycle 176<br />

see also amebiasis<br />

enteral nutrition (EN) 539–550<br />

complications 548–549<br />

gastrointestinal complications<br />

548<br />

infectious complications 549<br />

mechanical complications<br />

548<br />

metabolic complications 549<br />

following intestinal<br />

transplantation 708–709<br />

home enteral nutrition 549–550<br />

indications 549<br />

organization 549–550<br />

prevalence/incidence 549<br />

results 550<br />

teaching parents 550<br />

in preterm infants 544, 599–615<br />

early adaptive period 599–600<br />

nutrient needs 600–610<br />

practical aspects 610–615<br />

indications 540–544<br />

cholestatic liver disease 543<br />

cystic fibrosis 542–543<br />

eating disorders 543<br />

hypermetabolic states 544<br />

inborn errors of metabolism<br />

544<br />

inflammatory bowel diseases<br />

541–542<br />

neonatal abdominal surgery<br />

542<br />

premature infants 544<br />

protracted diarrhea of infancy<br />

541<br />

short-bowel syndrome<br />

466–467, 540–541<br />

physiological basis 539–540<br />

digestive secretion and<br />

hormonal response 540<br />

effects on energy expenditure<br />

540<br />

effects on mucosal trophism<br />

540<br />

gastrointestinal motility 539<br />

techniques 544–548<br />

choice of formula 546–547<br />

intake regulation 547–548<br />

nasogastric tubes 544–545<br />

nutrients 545–546<br />

percutaneous gastrostomy<br />

545<br />

enteric fever 148, 149<br />

enteric myositis 275–276<br />

enteric nervous system (ENS)<br />

214–215<br />

enterobiasis 178–179<br />

Enterobius vermicularis 178–179<br />

enterocolitis<br />

food protein-induced 323–325<br />

Hirschsprung’s 23, 260<br />

see also colitis<br />

enterocyte heparan sulfate<br />

deficiency 457<br />

enterotoxins 152<br />

environmental enteropathy<br />

208–209<br />

management 211<br />

eosinophilic enteropathy 328–329<br />

eosinophilic esophagitis 46–47<br />

food allergy and 328–329<br />

management 329<br />

eosinophilic gastritis 105<br />

eosinophils, in food allergy 338–339<br />

eotaxin 105, 338–339<br />

epidermal growth factor (EGF)<br />

464, 466<br />

epidermolysis bullosa 8<br />

episcleritis 394<br />

epithelial dysplasia see intestinal<br />

epithelial dysplasia<br />

Epstein–Barr virus (EBV)<br />

esophageal infection 35<br />

following intestinal<br />

transplantation 706–707<br />

erythema nodosum 394<br />

erythromycin<br />

Campylobacter treatment 150<br />

gastroesophageal reflux disease<br />

treatment 50<br />

Escherichia coli 151–155, 666<br />

diffusely adhering (DAEC) 154<br />

enteroaggregative (EAggEC)<br />

154–155<br />

enterohemorrhagic (EHEC)<br />

152–153<br />

enteroinvasive (EIEC) 152<br />

enteropathogenic (EPEC) 151<br />

enterotoxigenic (ETEC) 151–152<br />

esomeprazole 53<br />

esophageal dilatation 67<br />

esophagitis<br />

allergic 46<br />

eosinophilic 46–47<br />

food allergy and 328–329<br />

management 329<br />

gastrointestinal bleeding and<br />

643<br />

reflux 40, 46, 643<br />

see also infectious esophagitis<br />

esophagomyotomy 67–69<br />

esophagus<br />

adenocarcinoma 47<br />

atresia 13–14<br />

Barrett’s 40, 46, 47<br />

perforation 686<br />

pH monitoring 47–48, 225, 585<br />

stenosis 46<br />

strictures 46<br />

ulceration 34–35, 37, 46<br />

variceal bleeding 649–651<br />

prevention 650<br />

see also achalasia<br />

exomphalos 26<br />

treatment 26–27<br />

famciclovir, esophagitis treatment<br />

36<br />

familial adenomatous polyposis<br />

(FAP) 777–781<br />

clinical features 777<br />

diagnosis 778–780<br />

genetics 777–778<br />

management 780–781<br />

familial dysautonomia (FD) 295<br />

familial Mediterranean fever (FMF)<br />

223–224<br />

fasciolopsiasis 180<br />

Fasciolopsis buski 180<br />

fat overload syndrome 562–563<br />

fats see lipids<br />

fecal continence 249–250<br />

fecal retention 251<br />

fecal impaction prevention 256<br />

fecal impaction removal<br />

255–256<br />

fecal screening tests 660<br />

fecal soiling 247–248<br />

feeding development 235–238<br />

critical period 236<br />

feeding disorders<br />

diagnosis 240–241<br />

history 240–241<br />

physical evaluation 241<br />

following intestinal<br />

transplantation 709<br />

gastroesophageal reflux disease<br />

and 43<br />

prognosis 244<br />

treatment options 243–244<br />

enteral nutrition 543<br />

see also dysphagia; sucking<br />

disorders<br />

feminization, in malnutrition 499<br />

fever 678<br />

fish oils<br />

in parenteral nutrition 563–564<br />

ulcerative colitis management<br />

404<br />

fluconazole 32, 33, 119<br />

fluid balance, premature infants<br />

600, 621–622, 635<br />

fluid retention, with parenteral<br />

nutrition 568<br />

folate deficiency 490<br />

Crohn’s disease 354<br />

malnutrition 515<br />

folic acid, diarrhea management<br />

669<br />

food allergies 319–341<br />

classification 320–321<br />

cow’s milk allergy 334


egg allergy 334<br />

elimination diets 532<br />

food allergen avoidance<br />

recommendations 336<br />

future challenges 340–341<br />

immune response mechanisms<br />

336–340<br />

antigen presentation by<br />

epithelium 337<br />

mast cells and eosinophils<br />

338–339<br />

skewing of B cells towards<br />

IgE 337–338<br />

T-cell responses in oral<br />

tolerance 339–340<br />

lymphonodular hyperplasia and<br />

482–483, 485<br />

modification of allergenic<br />

proteins 532–533<br />

multiple food allergy 335–336<br />

patterns of allergic responses<br />

321–329<br />

allergic dysmotility 327–328<br />

eczema/atopic dermatitis<br />

325–327<br />

eosinophilic enteropathy<br />

328–329<br />

eosinophilic esophagitis<br />

328–329<br />

food protein enteropathy 325<br />

food protein-induced<br />

enterocolitis 323–325<br />

quick-onset symptoms<br />

321–323<br />

respiratory symptoms 329<br />

peanut allergy 335<br />

soy allergy 334<br />

testing for 329–333<br />

food challenge testing<br />

329–331<br />

in vitro testing 332–333<br />

skin patch testing 331–332<br />

skin prick testing 331<br />

specific IgE testing 332<br />

treatment 322–323<br />

wheat allergy 334–335<br />

see also allergic diseases<br />

food impaction 697<br />

food intolerance 319–320<br />

foreign body ingestion 691–698<br />

acute abdomen and 681<br />

clinical signs and symptoms<br />

692–695<br />

complications 694–695<br />

location of object 692–694<br />

cocaine ingestion 698<br />

diagnosis 695<br />

epidemiology 691–692<br />

food impaction 697<br />

treatment options 695–698<br />

follow-up 698<br />

foscarnet, esophagitis treatment<br />

35, 36<br />

Fredet–Ramstedt’s pyloromyotomy<br />

590–591<br />

functional abdominal pain<br />

213–229<br />

diagnostic testing 224–225<br />

blood studies 224<br />

endoscopic studies 224<br />

intraesophageal pH<br />

monitoring 225<br />

lactose hydrogen breath test<br />

225<br />

stool studies 224<br />

ultrasound 224–225<br />

differential diagnosis 220–224<br />

epidemiology 213–214<br />

future trends 229<br />

natural history 228–229<br />

pathophysiology 215–218<br />

biopsychosocial model<br />

217–218<br />

brain–gut interaction 216<br />

dysmotility 215<br />

genetics 217<br />

immunity 216<br />

inflammation 216<br />

stressors 216–217<br />

visceral hyperalgesia 215<br />

physiology of gastrointestinal<br />

pain response 214–215<br />

treatment 225–228<br />

alternative and<br />

complementary therapy<br />

228<br />

anticholinergics 226<br />

antidiarrheal medications<br />

226<br />

diet 226<br />

laxatives 226<br />

reassurance 225–226<br />

selective serotonin re-uptake<br />

inhibitors 227<br />

serotonin receptor<br />

antagonists 227–228<br />

tricyclic antidepressants<br />

226–227<br />

functional bowel disorders 213,<br />

218–219<br />

abdominal migraine 218, 219<br />

dysmotility 215<br />

dyspepsia 218, 219<br />

see also irritable bowel<br />

syndrome (IBS)<br />

functional constipation see<br />

constipation<br />

functional non-retentive fecal<br />

soiling 247–248, 252<br />

fundoplication 587–588, 762–763<br />

fungal infections<br />

esophagitis 29–32<br />

gastritides 97<br />

G-cell hyperfunction 101<br />

G-cell hyperplasia 101<br />

Index 793<br />

galactosyl ceramide (GalCer) 114<br />

gallstones 222<br />

ganciclovir<br />

cytomegalovirus infection<br />

management with HIV 119<br />

esophagitis treatment 35, 36<br />

gastric acid secretion 77<br />

continuous enteral feeding and<br />

540<br />

gut microbiota regulation 202<br />

hypersecretion 101, 462–463<br />

gastric asthma 46<br />

gastric carcinoma, H. <strong>pylori</strong><br />

infection and 88–89<br />

gastric ulcers<br />

gastrointestinal bleeding and<br />

643, 644<br />

management 647–648<br />

H. <strong>pylori</strong> and 76<br />

see also peptic ulcer disease<br />

(PUD)<br />

gastrin 77<br />

hypergastrinemia 101<br />

gastritis 95, 96<br />

alkaline 100<br />

autoimmune disease and 106<br />

chemical 97<br />

classification 83<br />

clinical assessment 107<br />

collagenous 105–106<br />

diagnosis 82–84, 107, 108<br />

eosinophilic 105<br />

focal enhanced 104<br />

granulomatous 103–104<br />

H. <strong>pylori</strong>-associated 80–81,<br />

82–84, 103<br />

prognosis 88<br />

lymphocytic 101–103<br />

celiac 102–103<br />

chronic varioliform 102<br />

H. <strong>pylori</strong> 103<br />

proton pump inhibitor gastritis<br />

106<br />

treatment 107–110<br />

tuberculosis 95–97<br />

gastroenteritis 127, 135, 745<br />

see also diarrhea<br />

gastroesophageal reflux disease<br />

(GERD) 39–54<br />

clinical signs and symptoms<br />

43–45, 584–585<br />

complications 46–47, 585<br />

definitions 39<br />

differential diagnosis 47–48,<br />

585–586<br />

environmental factors 40<br />

epidemiology 39–40, 584<br />

etiology 584<br />

genetic factors 40<br />

handicapped children 286–287<br />

percutaneous endoscopic<br />

gastrostomy and 287<br />

neonatal 583–588


794<br />

Index<br />

pathophysiology 40–43, 584<br />

prognosis 588<br />

respiratory disease and 45–46,<br />

585<br />

treatment 48–54, 586–588<br />

antacids 51–52<br />

complications of nonintervention<br />

48–49<br />

endoscopic procedures 54<br />

H2-receptor antagonists<br />

52–53<br />

laparoscopic techniques<br />

762–763<br />

non-pharmacological/<br />

non-surgical therapies 49<br />

prokinetics 49–51<br />

proton pump inhibitors 53–54<br />

surgery 54, 586–588<br />

gastrointestinal bleeding 639–651<br />

confirmation of 641<br />

definitions 639–640<br />

diagnostic investigations<br />

645–646<br />

endoscopy 646<br />

enteroscopy 646<br />

imaging 645–646<br />

epidemiology 639<br />

etiology 642–645<br />

childhood 643–644<br />

infancy 643<br />

neonatal period 642–643<br />

management and prevention<br />

646–651<br />

peptic lesions 647–648<br />

portal hypertension 649–651<br />

Meckel’s diverticulum and<br />

731–732<br />

ongoing bleeding 640–641<br />

presentations 639–640<br />

severity 640<br />

upper versus lower<br />

gastrointestinal bleeding<br />

641–642<br />

with ulcerative colitis 395<br />

gastrointestinal duplications 24,<br />

223<br />

gastrointestinal motility see<br />

intestinal motility<br />

gastrointestinal pain response<br />

214–215<br />

see also functional abdominal<br />

pain<br />

gastrointestinal polyps 771–782<br />

chemoprevention 782<br />

desmoid disease 781<br />

familial adenomatous polyposis<br />

777–781<br />

histopathological classification<br />

771<br />

juvenile polyposis syndrome<br />

773–774<br />

juvenile polyps 644, 651,<br />

771–773<br />

management 771<br />

Peutz–Jeghers syndrome<br />

774–777<br />

gastropathies 95, 96<br />

bile reflux 100<br />

classification 83<br />

clinical assessment 107<br />

diagnosis 107, 108<br />

drug-induced 97–98<br />

corticosteroids 98<br />

NSAIDs 97–98, 109<br />

portal hypertensive 100–101<br />

stress-related 98–99<br />

treatment 107–109<br />

treatment 107–110<br />

gastroschisis 26<br />

treatment 26–27<br />

Gaviscon 52<br />

genitourinary disorders 223<br />

giant cell arteritis 429<br />

Giardia lamblia 163<br />

gastric infection 97<br />

life cycle 163–164<br />

giardiasis 163–165<br />

clinical features 165<br />

diagnosis 165<br />

epidemiology 164<br />

impact on growth and<br />

development 186<br />

pathophysiology 164–165<br />

prevention 165<br />

treatment 183, 184, 667<br />

gliadin peptides 436<br />

glucagon-like-peptide-2 (GLP-2)<br />

473<br />

glucose<br />

in parenteral nutrition 559–560,<br />

568–569<br />

consequences of excessive<br />

intake 559–560<br />

optimal glucose/fat ratio 562<br />

premature infants 622–623,<br />

635<br />

in preterm formula 607<br />

glutamine (Gln) 464, 565, 626–627<br />

intestinal mucosa protection<br />

530–531, 565<br />

gluten sensitivity see celiac disease<br />

glycosaminoglycans (GAGs) 457, 509<br />

glycosylation disorders 457<br />

Gorlin’s syndrome 782<br />

graft-versus-host-disease (GVHD)<br />

106–107, 705<br />

granulomas<br />

chronic granulomatous disease<br />

104, 110<br />

Crohn’s disease 364, 381, 393<br />

gastritis and 103–104<br />

mucin granulomas 393<br />

vasculitides 421<br />

growth 494<br />

intestinal parasite impact<br />

184–186<br />

nutrition relationships 491–494<br />

short-bowel syndrome and 471<br />

growth hormone 472–473<br />

gut barrier 529<br />

gut microbiota 201, 202, 529–530<br />

inflammatory bowel disease and<br />

351<br />

regulation<br />

diet type 201–202<br />

digestive tract motility<br />

202–203<br />

gastric acid secretion 202<br />

ileocecal valve 203<br />

intestinal mucosal immunity<br />

203<br />

see also probiotics; small-bowel<br />

bacterial overgrowth (SBBO)<br />

H2-receptor antagonists 647–648<br />

gastroesophageal reflux disease<br />

treatment 52–53<br />

see also specific drugs<br />

hair changes in malnutrition 499<br />

hamartomas 771<br />

see also gastrointestinal polyps<br />

handicapped children 283–287<br />

appetite 284<br />

constipation 287<br />

gastroesophageal reflux 286–287<br />

percutaneous endoscopic<br />

gastrostomy and 287<br />

malnutrition and 283, 284<br />

nutrient deficiencies<br />

284–285<br />

nutritional intervention<br />

285–287<br />

Hasson technique 762<br />

heart failure, with malnutrition<br />

516<br />

management 516–517<br />

heartburn 40, 45<br />

see also gastroesophageal reflux<br />

disease<br />

height-for-age 493–495<br />

<strong>Helicobacter</strong> heilmannii 95<br />

<strong>Helicobacter</strong> <strong>pylori</strong> infection<br />

73–90, 220<br />

associated diseases 80<br />

clinical aspects 80–81<br />

complications 81–82<br />

diagnosis 84–85<br />

dyspepsia and 80–81, 218, 220<br />

epidemiology 74–76<br />

socioeconomic factors 75<br />

gastroesophageal reflux disease<br />

and 42<br />

immunization 89–90<br />

lymphocytic gastritis 103<br />

peptic ulcer disease etiology<br />

76–77<br />

mechanisms 78–80<br />

prognosis 88–89<br />

transmission 75–76


treatment 85–88<br />

indications for 85–86<br />

virulence factors 78<br />

Heller myotomy 67–69<br />

helminth parasites<br />

drug treatment 181–184<br />

mass community<br />

anthelminthic treatment 184–186<br />

immune responses 166–167<br />

impact on growth and<br />

development 184–185<br />

see also specific parasites<br />

hemangiomas 751, 752, 753–754<br />

hematemesis 639, 644<br />

see also gastrointestinal bleeding<br />

hematochezia 640<br />

see also gastrointestinal bleeding<br />

hematocolpos 223<br />

hematoma, duodenal 686<br />

hemolytic uremic syndrome (HUS)<br />

150, 152–154<br />

hemorrhage see gastrointestinal<br />

bleeding<br />

Henoch–Schönlein purpura<br />

429–430, 457, 645<br />

hepatitis, celiac disease and 440<br />

hepatomegaly, in malnutrition<br />

497–498<br />

herbal medicines<br />

abdominal pain management<br />

228<br />

diarrhea management 668<br />

herpes simplex virus (HSV)<br />

esophageal infection 32–34<br />

treatment 34, 36<br />

HIV infection and 118<br />

hiatal hernia, reflux and 43<br />

high-amplitude propagated<br />

contractions (HPAPCs) 250<br />

Hirschsprung’s disease 21–23, 259,<br />

702<br />

clinical signs and symptoms 22,<br />

260<br />

complications 260–261<br />

diagnosis 22, 253, 261–263<br />

epidemiology 259<br />

etiology 259<br />

follow-up 263<br />

genetic aspects 265–266<br />

Hirschsprung’s enterocolitis 23,<br />

260<br />

pathophysiology 259–260<br />

prognosis 23, 264<br />

treatment 22–23, 263<br />

laparoscopic procedures 767<br />

HIV infection 113–122<br />

diarrhea 115–117<br />

HIV enteropathy 117<br />

host responses and<br />

susceptibility 116–117<br />

management 120–121<br />

mucosal structure/function<br />

and 115–116<br />

persistent diarrhea 194<br />

disease progression 114–115<br />

enteric infections 117–120<br />

Candida 118–119<br />

Cryptosporidium parvum 118<br />

cytomegalovirus 119<br />

Microsporidia spp 119–120<br />

rotavirus 119<br />

epidemiology 113<br />

esophagitis and 29, 30, 35–37<br />

malnutrition and 521<br />

management 120–122<br />

diarrhea 120–121<br />

in resourced settings 121<br />

micronutrients 121<br />

nutrition 121–122<br />

surgical aspects 122<br />

terminal care 121<br />

pancreatitis and 306<br />

transmission 113–114<br />

mother-to-child transmission<br />

114<br />

homeopathy, diarrhea management<br />

668<br />

hookworm 168–170<br />

clinical features 169<br />

diagnosis 169<br />

epidemiology 169<br />

life cycle 168–169<br />

pathophysiology 169<br />

prevention 170<br />

treatment 182, 183, 185<br />

5-HT (serotonin) 214–215<br />

5-HT receptors 214–215<br />

antagonists, abdominal pain<br />

management 227–228<br />

Hughes syndrome 431<br />

human astroviruses (HAstVs) 137<br />

human papillomavirus, esophageal<br />

infection 35<br />

hydrocortisone 402<br />

hydrogen breath test 204–207,<br />

209, 221, 225<br />

hydrostatic reduction,<br />

intussusception 724<br />

hymenolepiasis 178<br />

Hymenolepis nana 178<br />

hyperganglionosis 264, 279<br />

hypergastrinemia 101<br />

hyperinsulinemic hypoglycemia<br />

21<br />

hypermetabolic states<br />

enteral nutrition and 544<br />

hypernatremia 657–658<br />

hypersensitivity 320<br />

see also food allergies<br />

hypertransaminasemia 440<br />

hypertrophic <strong>pylori</strong>c stenosis see<br />

infantile hypertrophic<br />

<strong>pylori</strong>c stenosis (IHPS)<br />

hypnotherapy, abdominal pain<br />

management 228<br />

hypoalbuminemia 451–452<br />

Index 795<br />

hypoganglionosis 278–279<br />

hypoglycemia, in malnutrition<br />

515, 658<br />

hypokalemia 658<br />

hypomotility agents see<br />

antimotility agents<br />

hyponatremia 658<br />

hyposplenism 443<br />

hypothermia, with malnutrition<br />

503, 506, 515–516<br />

management 515–516<br />

hypoxia 283<br />

IgE responses in food allergy<br />

337–338<br />

specific IgE testing 332<br />

ileal pouch–anal anastomosis<br />

(IPPA) 383, 405–406<br />

ileocecal valve<br />

gut microbiota regulation 203<br />

resection 462<br />

ileum<br />

classification 17–19<br />

obstruction 17–19<br />

resection 462<br />

treatment 19<br />

immune complexes, pathogenetic<br />

420<br />

immune response<br />

gut microbiota regulation 203<br />

inflammatory bowel disease and<br />

351–352<br />

lymphonodular hyperplasia and<br />

482–483<br />

to Cryptosporidium 172–173<br />

to dietary antigens 336–340<br />

antigen presentation by<br />

epithelium 337<br />

mast cells and eosinophils<br />

338–339<br />

skewing of B cells towards<br />

IgE 337–338<br />

T-cell responses in oral<br />

tolerance 339–340<br />

to helminth parasites 166–167<br />

to Strongyloides 175<br />

immunization<br />

Campylobacter 150<br />

cholera 147<br />

enterohemorrhagic E. coli 154<br />

H. <strong>pylori</strong> 89–90<br />

HIV 114<br />

Norwalk-like viruses 141<br />

rotavirus 139–141<br />

typhoid fever 149<br />

immunoglobulin treatment<br />

diarrhea management 669<br />

rotavirus infection 139<br />

immunosuppressive treatment 703<br />

immunotherapy<br />

Crohn’s disease 369–371<br />

food allergy 340–341<br />

indeterminate colitis 382


796<br />

Index<br />

impedancometry 48<br />

indeterminate colitis see colitis<br />

induction therapy<br />

left-sided colitis/proctitis<br />

401–402<br />

mild-to-moderate colitis<br />

397–400<br />

moderate-to-severe colitis<br />

400–401<br />

infantile hypertrophic <strong>pylori</strong>c<br />

stenosis (IHPS) 14, 588–591<br />

clinical signs and symptoms<br />

589<br />

complications 589–590<br />

differential diagnosis 590<br />

epidemiology 588<br />

etiology 588–589<br />

pathophysiology 589<br />

treatment 590–591<br />

laparoscopic pyloromyotomy<br />

763, 764<br />

infectious esophagitis 29–37<br />

bacterial infections 37<br />

epidemiology 29<br />

fungal infections 29–32<br />

predisposing factors 29<br />

viral infections 32–37<br />

infertility, celiac disease and 441<br />

inflammation<br />

abdominal pain and 216<br />

chronic intestinal<br />

pseudo-obstruction and 276,<br />

280<br />

diverticulitis 733<br />

in malnutrition 500<br />

pancreatitis 304<br />

parasitic infection and 167<br />

Cryptosporidium 172–173<br />

see also allergic diseases;<br />

vasculitides<br />

inflammatory bowel disease (IBD)<br />

222, 347, 379<br />

acute abdomen and 681–682, 685<br />

enteral nutrition and 541–542<br />

epidemiology 347–348<br />

etiology 348–351<br />

environmental factors<br />

349–351<br />

genetic factors 348–349<br />

extraintestinal manifestations<br />

394–395<br />

laparoscopic procedures<br />

763–764<br />

lymphonodular hyperplasia and<br />

483<br />

pathophysiology 351–352<br />

radiological imaging 381<br />

serologic markers 381<br />

see also colitis; Crohn’s disease;<br />

ulcerative colitis<br />

infliximab<br />

Crohn’s disease treatment<br />

370–371<br />

ulcerative colitis treatment<br />

403–404<br />

inguinal hernia 591–593<br />

clinical signs and symptoms<br />

592<br />

complications 592<br />

differential diagnosis 592<br />

epidemiology 591<br />

etiology 592<br />

pathogenesis 591–592<br />

treatment 593<br />

insulin-like growth factor-I (IGF-I)<br />

472<br />

interleukin 5 (IL-5) 338–339<br />

intestinal epithelial dysplasia 6–9,<br />

702–703<br />

associated disorders 8<br />

clinical expression 6<br />

diagnosis 7–8<br />

histological features 6–7<br />

outcome 9<br />

pathophysiology 8<br />

transmission 8<br />

intestinal failure 701, 712–713<br />

management 712<br />

see also intestinal<br />

transplantation<br />

intestinal lymphangiectasia<br />

452–455<br />

treatment 454–455<br />

intestinal malrotation 15, 16,<br />

19–20, 223<br />

treatment 19–20<br />

intestinal microbiota see gut<br />

microbiota<br />

intestinal motility<br />

continuous enteral feeding and<br />

539–540<br />

dysmotility 215, 327–328<br />

gut microbiota regulation<br />

202–203<br />

malnutrition and 503<br />

see also visceral myopathy;<br />

visceral neuropathies<br />

intestinal neuronal dysplasia (IND)<br />

264–265, 279<br />

genetic aspects 266<br />

intestinal obstruction see<br />

obstructive lesions<br />

intestinal parasites 161–163<br />

abdominal pain and 222<br />

acute abdomen and 684<br />

drug treatment 181–186<br />

mass community<br />

anthelminthic treatment 184–186<br />

epidemiology 161<br />

impact on growth and<br />

development 184–186<br />

see also specific parasites<br />

intestinal perforation see<br />

perforation<br />

intestinal polyps see<br />

gastrointestinal polyps<br />

intestinal pseudo-obstruction see<br />

chronic intestinal<br />

pseudo-obstruction (CIP)<br />

intestinal resection 464<br />

consequences of 461–463<br />

associated disorders 462–463<br />

ileal resection 462<br />

ileocecal valve resection 462<br />

jejunal resection 461–462<br />

small-bowel adaptation<br />

463–464<br />

intestinal smooth muscle disorders<br />

275<br />

intestinal transplantation 701–713<br />

clinical results 703–705<br />

immunosuppressive<br />

treatment 703<br />

liver-induced immune<br />

tolerance 705<br />

survival 703–704<br />

combined intestinal and liver<br />

transplantation 711, 712<br />

reduced-size composite<br />

allograft 713<br />

complications 705–707<br />

graft-versus-host-disease 705<br />

infectious complications<br />

706–707<br />

intestinal rejection 705–706<br />

indications 701–703<br />

congenital enteropathies<br />

702–703<br />

intestinal motility disorders<br />

702<br />

short-bowel syndrome<br />

701–702, 712<br />

intestinal graft function<br />

707–708<br />

absorptive function 708<br />

monitoring 709–710<br />

motor function 707–708<br />

living related donors 713<br />

microvillus inclusion disease<br />

5–6<br />

multivisceral transplantation<br />

713<br />

nutritional outcome 710<br />

post-transplant procedures<br />

708–710<br />

eating disorders 709<br />

initial post-transplant period<br />

708<br />

initiation of enteral and oral<br />

feeding 708–709<br />

monitoring of intestinal<br />

function 709–710<br />

parenteral nutrition weaning<br />

709<br />

potential candidates 710–713<br />

contraindications 711<br />

timing for referral 712–713<br />

intractable diarrhea of infancy (IDI)<br />

1


causes 2<br />

classification 1–2<br />

outcomes 2<br />

see also intestinal epithelial<br />

dysplasia; microvillus<br />

inclusion disease<br />

intractable ulcerating enterocolitis<br />

1<br />

intravenous fat emulsions (IVFEs)<br />

560–564<br />

fat overload syndromes<br />

562–563<br />

fish oil-based emulsions<br />

563–564<br />

intravascular metabolism<br />

561–562<br />

medium-chain triglycerides 563<br />

olive oil-based emulsions 564<br />

premature infants 627–629<br />

structured triglyceride emulsion<br />

563<br />

intussusception 719–727<br />

clinical manifestations<br />

720–721<br />

diagnosis 722–724<br />

barium enema 723–724<br />

radiography 722<br />

ultrasonography 722–723<br />

epidemiology 719<br />

etiopathology 720<br />

physical examination 721–722<br />

primary idiopathic 720<br />

secondary/organic 720<br />

treatment 724–727<br />

hydrostatic reduction 724<br />

medical treatment 724<br />

pneumatic reduction<br />

724–726<br />

surgery 726–727<br />

iron deficiency anemia<br />

celiac disease 440<br />

Crohn’s disease 354<br />

H. <strong>pylori</strong> infection and 81<br />

iron requirements, premature<br />

infants 608–609<br />

irritable bowel syndrome (IBS)<br />

213–214, 218, 219<br />

celiac disease and 443<br />

pathophysiology<br />

biopsychosocial model<br />

217–218<br />

dysmotility 215<br />

genetics 217<br />

inflammation 216<br />

visceral hyperalgesia 215<br />

see also functional abdominal<br />

pain<br />

isosorbide dinitrate, achalasia<br />

treatment 66<br />

Isospora belli 171<br />

itraconazole, esophagitis treatment<br />

32, 33<br />

ivermectin 181, 183<br />

jejunum<br />

obstruction 17–19<br />

classification 17–19<br />

treatment 19<br />

resection 461–462<br />

juvenile dermatomyositis 431<br />

juvenile polymyositis 431<br />

juvenile polyposis syndrome<br />

773–774<br />

clinical signs and diagnosis 773<br />

complications 774<br />

genetics 773–774<br />

treatment and follow-up 774<br />

juvenile polyps 644, 771–773<br />

polypectomy 651, 773<br />

Kaposi’s sarcoma 122<br />

Katayama syndrome 179<br />

Kawasaki’s disease 429<br />

ketoconazole, esophagitis treatment<br />

32, 33<br />

Klippel–Trénaunay syndrome 755,<br />

759<br />

kwashiorkor 496, 508–509<br />

changes during treatment<br />

510–511<br />

specific dermatosis of 522<br />

see also malnutrition<br />

D-lactic acidosis 471<br />

Lactobacillus spp 201, 210, 668<br />

see also probiotics<br />

lactose 606<br />

lactose intolerance 195, 198,<br />

221–222<br />

diarrhea and 664–665<br />

in malnutrition 518<br />

Ladd’s bands 19<br />

Ladd’s procedure 767–768<br />

lamina propria 113–114<br />

Langerhan’s histiocytosis 455, 456<br />

lansoprazole 53<br />

laparoscopy see minimally invasive<br />

surgery<br />

laxatives<br />

abdominal pain management<br />

226<br />

constipation management 255,<br />

256<br />

leukocytoclastic vasculitis 430<br />

levamisole 181, 183<br />

linoleic acid 605<br />

α-linolenic acid 605<br />

lipids<br />

in enteral nutrition 545–546<br />

premature infants 604–606<br />

in parenteral nutrition 560–564<br />

clearance of lipid emulsions<br />

561<br />

fish oil-based emulsions<br />

563–564<br />

lipoprotein interactions<br />

561–562<br />

Index 797<br />

medium-chain triglycerides<br />

563<br />

olive oil-based emulsions<br />

564<br />

optimal glucose/fat ratio 562<br />

premature infants 627–629,<br />

636<br />

structured triglyceride<br />

emulsion 563<br />

lipoproteins 561<br />

in parenteral nutrition 561–562<br />

liver disease<br />

congenital abnormalities 21<br />

enteral nutrition and 543<br />

hepatitis, celiac disease and 420<br />

in malnutrition 497–498<br />

parenteral nutrition-related<br />

571–572, 632–633<br />

related to short-bowel syndrome<br />

469–470<br />

management 470<br />

long-chain triglycerides (LCTs)<br />

605, 629<br />

lower esophageal sphincter (LES)<br />

41, 62–63, 65, 584<br />

transient relaxations (TLESRs)<br />

41, 42–43<br />

lymphatic malformations 757–758<br />

lymphocytic gastritis 101–103<br />

celiac 102–103<br />

chronic varioliform 102<br />

H. <strong>pylori</strong> 103<br />

lymphoma 89, 783–784<br />

lymphonodular hyperplasia (LNH)<br />

479–486<br />

assessment 479–480<br />

classification 479<br />

differential diagnosis 483–485<br />

food allergy and 482–483, 485<br />

histology 480–481<br />

immunohistochemistry<br />

481–482<br />

pathophysiology 482–483<br />

prognosis 486<br />

symptoms 485–486<br />

treatment 486<br />

M cells 113, 114<br />

magnesium requirement,<br />

premature infants 630<br />

magnetic resonance<br />

cholangiopancreatography<br />

(MRCP) 310–314<br />

magnetic resonance imaging (MRI),<br />

inflammatory bowel disease<br />

381<br />

malabsorption<br />

bile salts 356<br />

carbohydrates 116, 203<br />

in small-bowel bacterial<br />

overgrowth 203–204,<br />

208–209<br />

Mallory–Weiss tear 643


798<br />

Index<br />

malnutrition 489–523<br />

classification 493–496<br />

adults 495–496<br />

children 493–495<br />

clinical features 496–500<br />

abdominal swelling 500<br />

anemia 500<br />

anorexia 498<br />

bone 499–500<br />

cheeks 499<br />

circulation 497<br />

edema 496–497<br />

feminization 499<br />

hair 499<br />

hepatomegaly 497–498<br />

inflammation 500<br />

mood/behavior 498–499<br />

skin 499<br />

splenomegaly 498<br />

Crohn’s disease 356<br />

micronutrient deficiencies<br />

357–358<br />

diarrhea and 193–194, 195,<br />

657–658<br />

drug metabolism and 522–523<br />

handicapped children 283, 284<br />

nutrient deficiencies<br />

284–285<br />

nutritional intervention<br />

285–287<br />

investigations 511<br />

kwashiorkor (edematous) 496,<br />

508–509<br />

changes during treatment<br />

510–511<br />

specific dermatosis of 522<br />

management 511–523<br />

acute phase 512–517<br />

emotional/psychological<br />

stimulation 519–520<br />

preparation for discharge<br />

520–521<br />

problems with 521–523,<br />

567–568<br />

progress assessment 519<br />

rehabilitation phase 518–519<br />

transition phase 517–518<br />

marasmus 496<br />

nutritional dwarfism 496<br />

pathophysiology 500–508<br />

body composition 503–507<br />

energy requirement reduction<br />

500–501<br />

loss of homeostasis 507–508<br />

loss of reserve 507<br />

physiological and metabolic<br />

changes 501–502,<br />

504–506<br />

reduced mass 500–501<br />

vicious cycles 507<br />

swallowing disorder and 238<br />

type I deficiency 490–491<br />

type II deficiency 491–493<br />

malrotation see intestinal<br />

malrotation<br />

maltodextrin-based oral<br />

rehydration solutions 662<br />

MALToma 89<br />

manometry<br />

anorectal<br />

constipation 254, 262<br />

Hirschsprung’s disease 261,<br />

262<br />

colonic<br />

chronic intestinal pseudoobstruction<br />

272<br />

constipation 254<br />

esophageal<br />

achalasia 65, 66<br />

chronic intestinal pseudoobstruction<br />

271<br />

gastroesophageal reflux<br />

disease 48, 585–586<br />

pharyngeal 242–243<br />

small intestine, chronic<br />

intestinal pseudo-obstruction<br />

272–273<br />

marasmus 496<br />

see also malnutrition<br />

mast cells, in food allergy 338–339<br />

measles<br />

Crohn’s disease and 350–351<br />

malnutrition and 515<br />

mebendazole 181–182<br />

mass community treatment 185<br />

Meckel’s diverticulum 20–21, 644,<br />

729–737<br />

clinical features 20<br />

complications 731–734<br />

gastrointestinal bleeding<br />

731–732<br />

inflammation 733<br />

intestinal obstruction<br />

732–733<br />

diagnosis 734–735<br />

epidemiology 730–731<br />

histology 730<br />

management 21, 735–737<br />

outcomes 737<br />

pathoembryology 729–730<br />

predisposing factors 731<br />

Meckel’s scan 645, 734–735<br />

meconium ileus 20<br />

medium-chain triglycerides (MCTs)<br />

545–546, 563, 605, 629<br />

megacolon 261<br />

toxic 395–396, 401, 681, 684<br />

megacystis microcolon<br />

hypoperistalsis syndrome 274<br />

melena 640<br />

see also gastrointestinal bleeding<br />

Ménétrier’s disease 99–100<br />

protein-losing enteropathy and<br />

456<br />

treatment 109<br />

6-mercaptopurine<br />

Crohn’s disease treatment 369<br />

ulcerative colitis management<br />

402–403<br />

mesalamine 401, 402<br />

mesenteric cysts 757–758<br />

metabolic bone disease<br />

with Crohn’s disease 359<br />

methotrexate<br />

Crohn’s disease treatment 369<br />

ulcerative colitis management<br />

403<br />

vasculitis treatment 432<br />

metoclopramide 665–667<br />

gastroesophageal reflux disease<br />

treatment 50<br />

metronidazole<br />

amebiasis treatment 183<br />

Clostridium difficile management<br />

156<br />

giardiasis treatment 183<br />

H. <strong>pylori</strong> eradication 87<br />

small-bowel bacterial<br />

overgrowth treatment 210<br />

microbiota see gut microbiota<br />

Microsporidia spp, HIV infection<br />

and 118, 119–120, 171<br />

microvillus inclusion disease<br />

(MVID) 2–6, 702–703<br />

clinical expression 2–3<br />

definitive treatment 5–6<br />

histological examination 3–4<br />

outcome 5<br />

pathophysiology 4–5<br />

transmission 5<br />

mid-upper-arm circumference 495,<br />

496<br />

migraine<br />

abdominal 218, 219<br />

cyclic vomiting syndrome and<br />

294, 295–296, 297<br />

treatment 299–300<br />

milk allergy 334<br />

modification of allergenic<br />

proteins 532–533<br />

see also lactose intolerance<br />

milk protein enteropathy 195–196,<br />

325<br />

mineral requirements, premature<br />

infants 607–609, 630–631<br />

minimally invasive surgery<br />

761–768<br />

appendicitis 765–766<br />

background 761<br />

cholecystectomy 766–767<br />

chronic abdominal pain 766<br />

complications 768<br />

constipation 767<br />

gastroesophageal reflux disease<br />

762–763<br />

inflammatory bowel disease<br />

763–765<br />

Ladd’s procedure 767–768<br />

<strong>pylori</strong>c stenosis 763


technical considerations<br />

761–762<br />

mitochondrial dysfunction<br />

294–295<br />

mouth, congenital abnormalities<br />

13<br />

mucormycosis 684<br />

multiple endocrine neoplasia<br />

277–278<br />

multiple food allergy 335–336<br />

multivisceral transplantation 713<br />

muscular dystrophy 276<br />

musculoskeletal pain 223<br />

Mycobacterium, HIV infection and<br />

118<br />

mycophenolate mofetil 703<br />

myenteric (Auerbach’s) plexus 214<br />

nasogastric tubes 544–545<br />

in rehydration therapy 661<br />

Necator americanus 168<br />

epidemiology 169<br />

see also hookworm<br />

necrotizing enterocolitis (NEC)<br />

177, 579–583, 599<br />

clinical signs and symptoms<br />

580–581<br />

complications 581<br />

diagnosis 581–582<br />

epidemiology 579<br />

etiology 579–580<br />

pathophysiology 580<br />

prognosis 583<br />

risk factors 580<br />

treatment 582–583<br />

neonatal esophagogastritis 642<br />

nephrolithiasis, Crohn’s disease<br />

and 358<br />

nifedipine, achalasia treatment 66<br />

Nissen’s fundoplication 54,<br />

586–587, 762–763<br />

nitazoxanide 184<br />

nitrates, achalasia treatment 66<br />

nitric oxide (NO) 63, 173, 589<br />

nitrogen intake<br />

enteral nutrition 545, 601<br />

parenteral nutrition 564–565,<br />

569, 625<br />

premature infants 601, 625<br />

nizatidine, gastroesophageal reflux<br />

disease treatment 52–53<br />

non-Hodgkin’s lymphoma 783–784<br />

non-organic failure to thrive<br />

(NOFTT) 43<br />

non-steroidal anti-inflammatory<br />

drugs (NSAIDs)<br />

gastric injury 97–98, 109<br />

treatment 109<br />

inflammatory bowel disease and<br />

350<br />

ulcerative colitis 387<br />

role in chemoprevention 782<br />

Norwalk agent 137<br />

Norwalk-like viruses (NLV)<br />

137–138<br />

vaccine development 141<br />

NSP4 protein 130–132<br />

nutrition 489, 525<br />

diet role in chronic diseases<br />

527–528<br />

dietary deficiency 492<br />

see also malnutrition<br />

dietary recommendations and<br />

guidelines 526<br />

dietary requirements 492<br />

specific dietary requirements<br />

of disease 526–527<br />

early nutrition and later<br />

consequences 526<br />

nutritional deficiency see<br />

malnutrition<br />

nutritional dwarfism 496<br />

see also malnutrition<br />

nutritional therapy 527<br />

Crohn’s disease 365–366<br />

diarrhea 663–665<br />

breast feeding 663–664<br />

diluted or full-strength milk<br />

or formula 665<br />

early versus late feeding 663<br />

lactose-containing versus<br />

lactose-free feeds 664–665<br />

ulcerative colitis 404–405<br />

see also dietary intervention<br />

obstructive lesions 15–19, 682<br />

Ascaris infection 168<br />

clinical features 15<br />

duodenum 16–17<br />

ileum 17–19<br />

investigations 15–16<br />

jejunum 17–19<br />

Meckel’s diverticulum 732–733<br />

see also chronic intestinal<br />

pseudo-obstruction (CIP);<br />

intussusception<br />

octreotide<br />

gastroesophageal reflux disease<br />

treatment 54<br />

intestinal lymphangiectasia<br />

treatment 454–455<br />

variceal bleeding management<br />

649–650<br />

olive oil, in parenteral nutrition 564<br />

omeprazole 648<br />

gastritis and 106<br />

gastroesophageal reflux disease<br />

treatment 53–54<br />

oncotic pressure restoration 568<br />

ondansetron 667<br />

gastroesophageal reflux disease<br />

treatment 51<br />

oral cavity 235<br />

congenital abnormalities 13<br />

oral contraceptives, inflammatory<br />

bowel disease and 350<br />

Index 799<br />

oral rehydration solutions (ORS)<br />

147, 156, 196, 469, 660–663<br />

amino acid ORS 662<br />

amylase resistant starch ORS<br />

663<br />

cereal-based ORS 662<br />

in malnutrition 512, 513<br />

maltodextrin-based ORS 662<br />

reduced-osmolarity ORS 662<br />

scientific background 661–662<br />

versus intravenous rehydration<br />

660–661<br />

ornithine α-keto glutarate (OKG)<br />

464, 565<br />

Osler–Weber–Rendu syndrome<br />

755<br />

osteopenia<br />

with celiac disease 440–441<br />

with Crohn’s disease 359<br />

with ulcerative colitis 395<br />

osteoporosis<br />

with celiac disease 440–441<br />

with Crohn’s disease 359<br />

oxamniquine 181<br />

pain see functional abdominal pain<br />

pancreas, congenital abnormalities<br />

21<br />

pancreatitis 303–317<br />

acute 303, 683<br />

associated conditions 305<br />

Crohn’s disease 358<br />

chronic 303<br />

clinical signs and symptoms<br />

308<br />

complications 316<br />

diagnosis 220, 308–315<br />

laboratory tests 308–310<br />

radiographic studies 310–315<br />

epidemiology 304<br />

etiology/pathophysiology<br />

304–308<br />

anatomic abnormalities 304<br />

hereditary, metabolic and<br />

systemic diseases 306–307<br />

infection 304–306<br />

medications 307–308<br />

traumatic causes 304<br />

hemorrhagic 303<br />

hereditary 303, 306–307<br />

necrotic 303<br />

prognosis 316<br />

treatment 315<br />

pangastritis 82<br />

pantoprazole 53<br />

parasites see intestinal parasites;<br />

specific parasites<br />

Parastrongylus costaricensis 180<br />

parenteral nutrition (PN) 1, 196,<br />

555–572, 701<br />

complications 469, 567–572,<br />

632–634<br />

adaptation of intake 570


800<br />

Index<br />

body temperature 569<br />

bone disease 571<br />

catheter-related sepsis 570,<br />

633<br />

glucose homeostasis 568–569<br />

infection 569, 633<br />

liver disease 571–572,<br />

632–633<br />

micronutrients 569–570<br />

oncotic pressure restoration<br />

568<br />

potassium depletion 569<br />

protein and energy intake<br />

569<br />

refeeding syndrome 567–568<br />

water and sodium overload<br />

568<br />

following intestinal<br />

transplantation 709<br />

in clinical practice 557–567<br />

all-in-one mixture 566–567<br />

energy requirements<br />

558–559<br />

energy sources 559–560<br />

fat overload syndrome<br />

562–563<br />

intravenous fat emulsions<br />

561–564<br />

nitrogen sources 564–565<br />

supplies 558<br />

vascular access 557–558<br />

in premature infants 619–636<br />

complications 632–634<br />

nutritional requirements<br />

619–621<br />

practical aspects 634–636<br />

indications 555–556<br />

intestinal epithelial dysplasia<br />

9<br />

microvillus inclusion disease<br />

5<br />

short-bowel syndrome 461,<br />

465, 466, 468, 471<br />

long-term parenteral nutrition<br />

556–557<br />

cyclic parenteral nutrition<br />

556–557<br />

home parenteral nutrition<br />

557<br />

weaning 470, 709<br />

paromomycin 184<br />

peanut allergy 335<br />

pediatric Crohn’s disease activity<br />

index (PCDAI) 370–372<br />

peppermint oil (Mentha piperita)<br />

228<br />

pepsinogens 77–78<br />

pepsins 77<br />

peptic disease (PD) 73, 220<br />

peptic ulcer disease (PUD)<br />

clinical aspects 80–81<br />

complications 81–82<br />

corticosteroids and 98<br />

diagnosis 82, 220<br />

epidemiology 74<br />

etiology 76–77<br />

gastrointestinal bleeding and<br />

643, 644<br />

management 647–648<br />

H. <strong>pylori</strong> and 73, 78–80<br />

pathogenesis 77–80<br />

acid and pepsinogen role<br />

77–78<br />

H. <strong>pylori</strong>-related mechanisms<br />

78–80<br />

prognosis 88<br />

percutaneous endoscopic<br />

gastrostomy (PEG) 545<br />

acute abdomen and 685–686<br />

handicapped children 287<br />

perforation 15, 682<br />

bile duct 683<br />

endoscopic procedures and<br />

684–686, 768<br />

following foreign body ingestion<br />

694–695<br />

typhoid perforation 683<br />

perianal lesions, Crohn’s disease<br />

355–356<br />

perinuclear antineutrophil<br />

cytoplasmic antibodies<br />

(P-ANCA) 361, 381, 390, 421<br />

periodic acid Schiff (PAS) staining<br />

3<br />

peritonitis, HIV infection and 122<br />

see also acute abdomen<br />

pernicious anemia 106, 110<br />

persistent diarrhea 193–211, 658<br />

HIV relationship 194<br />

management 196–198<br />

antibiotic therapy 196<br />

enteral feeding and diet<br />

selection 197–198<br />

follow-up and nutritional<br />

rehabilitation 198<br />

micronutrient<br />

supplementation 198<br />

oral rehydration therapy 196<br />

pathogenesis 193–194<br />

risk factors 194–196<br />

dietary risk factors 195–196<br />

inappropriate management<br />

196<br />

malnutrition 195<br />

specific pathogens 194–195<br />

small-bowel bacterial<br />

overgrowth 208<br />

see also diarrhea<br />

Peutz–Jeghers syndrome 774–777<br />

clinical features and diagnosis<br />

774–775<br />

follow-up 776–777<br />

genetics 775<br />

management and complications<br />

775–776<br />

Peyer’s patches 529<br />

pH monitoring, esophageal 47–48,<br />

225, 585<br />

pharynx 235<br />

phosphorus requirement,<br />

premature infants 630<br />

picobirnaviruses 139<br />

picornaviruses 138–139<br />

pinworm 178–179<br />

pirenzipine 53<br />

pneumatic reduction,<br />

intussusception 724–726<br />

polyarteritis nodosa (PAN) 427<br />

polyhydramnios, obstructive<br />

lesions and 15<br />

polymyalgia rheumatica 429<br />

polymyxin, small-bowel bacterial<br />

overgrowth treatment 210<br />

polypectomy 651, 771<br />

acute abdomen and 685<br />

polyps see gastrointestinal polyps<br />

polyunsaturated fatty acids (PUFA)<br />

533, 605–606, 628–629<br />

portal hypertension 649–651<br />

gastropathy 100–101<br />

management 649–651<br />

endoscopic treatment<br />

650–651<br />

prevention of bleeding 650<br />

potassium requirement, premature<br />

infants 630<br />

pouchitis 405, 406–407<br />

praziquantel 181<br />

mass community treatment<br />

184, 185–186<br />

prebiotics 531<br />

prednisolone<br />

Crohn’s disease treatment<br />

366–368<br />

ulcerative colitis treatment 400,<br />

402<br />

prednisone, Crohn’s disease<br />

treatment 366–368<br />

premature infants<br />

early adaptive period 599–600,<br />

620<br />

enteral nutrition 544, 599–615<br />

breast milk 610–613<br />

post-discharge nutrition<br />

614–615<br />

preterm formulas 613–614<br />

intermediate and stable growth<br />

period 600–610, 620–621<br />

necrotizing enterocolitis 579,<br />

599<br />

nutritional requirements<br />

600–610, 619–621<br />

carbohydrates 606–607,<br />

622–623<br />

energy requirements 603,<br />

622<br />

fluids 600, 621–622, 635<br />

iron 608–609<br />

lipids 604–606, 627–629, 636


minerals 607–609, 630–631<br />

protein 600–603, 623–624,<br />

635–636<br />

protein/energy ratio 603–604,<br />

623<br />

vitamins 609–610, 631–632<br />

parenteral nutrition 619–636<br />

complications 632–634<br />

practical aspects 634–636<br />

primary sclerosing cholangitis<br />

(PSC) 395<br />

probiotics 530, 531<br />

allergic disease management<br />

327, 340, 534–535<br />

diarrhea management 156, 210,<br />

531, 668<br />

inflammatory bowel syndrome<br />

management 228<br />

pouchitis management 406–407<br />

small-bowel bacterial<br />

overgrowth management 210<br />

ulcerative colitis management<br />

404<br />

processus vaginalis 591<br />

proctitis treatment 401–402<br />

prokinetics 49–51<br />

see also specific drugs<br />

prostaglandin E2 (PGE2) 533<br />

protein deficiency 490, 509, 565<br />

see also malnutrition<br />

protein requirements 492–493<br />

premature infants 600–603,<br />

623–627<br />

amino acid composition of<br />

parenteral nutrition<br />

solutions 624–626,<br />

635–636<br />

amino acids for special<br />

purposes 626–627<br />

protein/energy ratio 603–604,<br />

623<br />

protein-losing enteropathy (PLE)<br />

451–457<br />

causes 452–457<br />

defective cellular synthesis<br />

457<br />

increased lymphatic pressure<br />

455<br />

intestinal lymphangiectasia<br />

452–455<br />

Ménétrier’s disease 456<br />

mucosal lesions 455<br />

vasculitides 456–456<br />

investigations 452<br />

proton pump inhibitors (PPIs)<br />

gastric ulcer treatment 108,<br />

109, 647–648<br />

gastritis and 106<br />

gastroesophageal reflux disease<br />

treatment 53–54<br />

see also specific drugs<br />

protozoan infections 97<br />

see also specific protozoans<br />

protracted diarrhea of infancy<br />

(PDI) 1<br />

causes 2<br />

enteral nutrition and 541<br />

outcomes 2<br />

prucalopride 51<br />

prune belly syndrome 274<br />

pseudo-obstruction see chronic<br />

intestinal pseudo-obstruction<br />

(CIP)<br />

pseudo-Zollinger–Ellison syndrome<br />

(PZES) 101<br />

management 109<br />

pseudomembranous colitis (PMC)<br />

155<br />

psychotherapy, abdominal pain<br />

management 228<br />

pulmonary aspiration 46, 240<br />

punctiform keratitis 8<br />

pyelonephritis 223<br />

<strong>pylori</strong>c atresia 8<br />

<strong>pylori</strong>c stenosis see infantile<br />

hypertrophic <strong>pylori</strong>c stenosis<br />

(IHPS)<br />

pyloromyotomy 590–591<br />

laparoscopic 763, 764<br />

pyoderma gangrenosum 394<br />

pyrantel 181, 182<br />

rabeprazole 53<br />

racecadotril 668<br />

radiography<br />

achalasia 64–65<br />

acute abdomen 679<br />

appendicitis 743–744<br />

chronic intestinal<br />

pseudo-obstruction 271<br />

constipation 253–254<br />

Crohn’s disease 361<br />

foreign body ingestion 695<br />

gastrointestinal bleeding<br />

investigation 645<br />

intussusception 722<br />

necrotizing enterocolitis 582<br />

pancreatitis 310–315<br />

swallowing disorders 241–242<br />

ulcerative colitis 392<br />

ranitidine 647–648<br />

gastroesophageal reflux disease<br />

treatment 52–53<br />

rapamycin 703<br />

rectovaginal fistula, with HIV<br />

infection 122<br />

recurrent abdominal pain (RAP)<br />

80–81, 213<br />

see also functional abdomina<br />

pain<br />

reduced osmolarity oral<br />

rehydration solutions 662<br />

refeeding syndrome 567–568<br />

reflux esophagitis 40, 46, 643<br />

see also gastroesophageal reflux<br />

disease<br />

Index 801<br />

refractory sprue 444<br />

regurgitation 39, 44–45<br />

achalasia and 63–64<br />

epidemiology 39–40<br />

see also gastroesophageal reflux<br />

disease<br />

rehydration therapy 660–663<br />

oral versus intravenous<br />

rehydration 660–661<br />

see also oral rehydration<br />

solutions (ORS)<br />

renal solute load (RSL) 600<br />

respiratory disease<br />

food allergy and 329<br />

gastroesophageal reflux disease<br />

and 45–46, 585<br />

swallowing disorder<br />

complications 239–240<br />

RET signalling pathway 265–266<br />

rhesus rotavirus-tetravalent<br />

vaccine (RRV-TV) 140<br />

rheumatoid vasculitis 431–432<br />

rotavirus 135<br />

diarrhea and 127, 135–137<br />

pathophysiology 128–132,<br />

133<br />

with HIV infection 119<br />

epidemiology 136<br />

immunoglobulin treatment 139<br />

transmission 136<br />

vaccine development 139–141<br />

virology 136–137<br />

Salmonella 148–149, 666<br />

acute abdomen and 684<br />

enteric fever 149<br />

treatment 148, 149<br />

Sapporo-like viruses (SLV)<br />

137–138<br />

Sarcocystis hominis 171<br />

Schistosoma mansoni 179<br />

schistosomiasis 179–180<br />

treatment 185–186<br />

scintigraphy<br />

Crohn’s disease 361–363<br />

gastrointestinal bleeding<br />

investigation 645<br />

Meckel’s diverticulum 734–735<br />

swallowing evaluation 243<br />

sclerotherapy 649, 650–651<br />

secretory leukocyte protease<br />

inhibitor (SLPI) 114<br />

selective serotonin re-uptake<br />

inhibitors (SSRIs) 227<br />

sepsis<br />

catheter-related 570, 633<br />

necrotizing enterocolitis and<br />

580<br />

serotonin see 5-HT<br />

serum amylase evaluation 308, 309<br />

serum lipase evaluation 308–309<br />

severe diarrhea requiring<br />

parenteral nutrition 1


802<br />

Index<br />

Shiga-like toxins (SLT) 152–153<br />

Shigella 149–150, 666<br />

HIV infection and 118<br />

shock, in malnutrition 512–513<br />

short stature, celiac disease and<br />

440<br />

short-bowel syndrome 461–474,<br />

712<br />

clinical management 464–471<br />

bacterial overgrowth 468<br />

dehydration prevention 469<br />

diet type 467–468<br />

feeding mode 466–467,<br />

540–541<br />

fluid loss adaptation 468<br />

fluid loss reduction 468–469<br />

initial surgery 464<br />

medical therapy 465–466<br />

parenteral nutrition cycling<br />

468<br />

unadapted short small bowel<br />

472–473<br />

complications 469–471<br />

D-lactic acidosis 471<br />

liver disease 469–470<br />

long-term complications 470<br />

consequences of intestinal<br />

resection 461–462<br />

associated disorders 462–463<br />

ileal resection 462<br />

ileocecal valve resection 462<br />

jejunal resection 461–462<br />

etiology 465<br />

follow-up 471<br />

growth monitoring 471<br />

prognosis 472–473<br />

small-bowel adaptation after<br />

resection 463–464<br />

pharmacological<br />

enhancement 472–473<br />

surgical treatment 473, 701–702<br />

short-chain fatty acids (SCFA) 464<br />

ulcerative colitis management<br />

404<br />

sialadenitis 122<br />

sialorrea 239<br />

skin changes in malnutrition 499<br />

skin patch testing 331–332<br />

skin prick testing 331<br />

small-bowel adaptation 463–464<br />

small-bowel bacterial overgrowth<br />

(SBBO) 201–211, 468<br />

clinical presentation 207–209<br />

acute and persistent diarrhea<br />

208<br />

environmental enteropathy<br />

208–209<br />

diagnosis 204–207<br />

differential diagnosis 222<br />

effects of 204<br />

etiology 201–203<br />

in malnutrition 507, 514–515<br />

pathophysiology 203–204<br />

treatment 209–211, 468<br />

see also gut microbiota<br />

smectite 667–668<br />

Smith–Lemli–Opitz syndrome 588<br />

smoking, inflammatory bowel<br />

disease and 349–350<br />

ulcerative colitis 386<br />

sodium, in parenteral nutrition<br />

568<br />

premature infants 630<br />

somatostatin 649–650<br />

SOX10 gene 266<br />

soy allergy 334<br />

splenomegaly, in malnutrition 498<br />

stenosis<br />

esophageal 46<br />

small bowel 15<br />

duodenal 16–17<br />

jejunoileal 17–19<br />

see also infantile hypertrophic<br />

<strong>pylori</strong>c stenosis (IHPS)<br />

stomach<br />

bacterial infection 95–97<br />

see also <strong>Helicobacter</strong> <strong>pylori</strong><br />

infection<br />

chronic granulomatous disease<br />

104, 110<br />

congenital abnormalities 14<br />

Crohn’s disease 103–104<br />

fungal infection 97<br />

protozoan infection 97<br />

viral infection 97<br />

see also gastritis; gastropathies;<br />

peptic ulcer disease (PUD)<br />

stress ulcers 80, 98–99<br />

treatment 107–109<br />

stressors<br />

abdominal pain 216–217<br />

cyclic vomiting syndrome 291<br />

stricture<br />

colonic 396<br />

esophageal 46<br />

Strongyloides stercoralis 174<br />

life cycle 174<br />

strongyloidiasis 174–176<br />

clinical features 175<br />

diagnosis 175–176<br />

disseminated (hyperinfection)<br />

175<br />

epidemiology 174<br />

pathophysiology 174–175<br />

prevention 176<br />

treatment 183<br />

structured triglyceride emulsion<br />

563<br />

stunting 494–495<br />

Sturge–Weber syndrome 755<br />

submucosal (Meissner’s) plexus<br />

214<br />

sucking 236<br />

sucking disorders<br />

clinical signs and symptoms<br />

239<br />

epidemiology 233<br />

etiology 233<br />

pathophysiology 233–235<br />

sucralfate 54<br />

sudden infant death syndrome<br />

(SIDS), H. <strong>pylori</strong> infection and<br />

81–82<br />

sulfasalazine 366, 398–400, 402<br />

swallowing 236–238<br />

anatomic considerations 235<br />

development and 235–238<br />

swallowing disorders see<br />

dysphagia<br />

systemic lupus erythematosus<br />

(SLE) 430–431<br />

protein-losing enteropathy and<br />

456–457<br />

T-cell responses<br />

in food allergy 337, 339–340<br />

in vasculitides 421<br />

tacrolimus 5, 401, 703<br />

Takayasu’s arteritis 428–429<br />

tapeworm 178<br />

tegaserod 51<br />

Thal procedure 587<br />

threadworm 178–179<br />

thromboangiitis obliterans 432<br />

thromboembolism, inflammatory<br />

bowel disease and 358–359, 395<br />

tinidazole 183–184<br />

tobacco smoking see smoking<br />

toilet training 250, 255<br />

toroviruses 138<br />

total colonic aganglionosis (TCA)<br />

259<br />

toxic colitis 681–682<br />

toxic megacolon 395–396, 401,<br />

681, 684<br />

toxic shock 512–513<br />

tracheoesophageal fistula 13–14<br />

classification 13–14<br />

clinical features 14<br />

outcome 14<br />

treatment 14<br />

transanal mucosectomy 767<br />

transdermal nicotine therapy,<br />

ulcerative colitis 404<br />

transglutaminase 436<br />

antibody test 446<br />

transient lower esophageal<br />

sphincter relaxations (TLESRs)<br />

41, 42–43<br />

trauma, pancreatitis and 304<br />

Trichostrongylus 180<br />

trichuriasis 170–171<br />

clinical features 170–171<br />

diagnosis 171<br />

epidemiology 170<br />

life cycle 170<br />

pathophysiology 170<br />

prevention 171<br />

treatment 182, 185–186


Trichuris trichiuria 170<br />

see also trichuriasis<br />

tricorrhexis nodosa 9<br />

tricyclic antidepressants (TCA)<br />

226–227<br />

Trypanosoma cruzi 280<br />

tuberculosis<br />

gastritis 95–97<br />

intestinal 684<br />

tufting enteropathy see intestinal<br />

epithelial dysplasia<br />

tumor necrosis factor-α (TNF-α)<br />

370<br />

anti-TNF-α agents 370–371<br />

Turcot’s syndrome 782<br />

typhoid fever 148, 149<br />

vaccine 149<br />

typhoid perforation 683<br />

ulcerative colitis 385–409<br />

clinical signs and symptoms<br />

388–389<br />

complications 395–396<br />

diagnosis 389–393<br />

differential diagnosis<br />

389–390<br />

endoscopy 390–392<br />

laboratory tests 390<br />

radiography 392<br />

epidemiology 385, 386<br />

etiology/pathogenesis 385–388<br />

environmental factors<br />

386–387<br />

genetic factors 385–386<br />

extraintestinal manifestations<br />

394–395<br />

follow-up 408–409<br />

pathology 393<br />

prognosis 407–408<br />

severity 397<br />

treatment options 396–407<br />

induction therapy 397–402<br />

laparoscopic procedures<br />

764–765<br />

maintenance therapy<br />

402–403<br />

nutritional therapy 404–405<br />

psychosocial support 407<br />

surgical therapy 405–407<br />

see also colitis; inflammatory<br />

bowel disease (IBD)<br />

ulcers<br />

Behçet’s disease 428<br />

colonic<br />

amebiasis 177<br />

Crohn’s disease 363, 364,<br />

391<br />

see also ulcerative colitis<br />

duodenal 76<br />

esophageal 34–35, 37, 46<br />

gastric 76<br />

drug-induced 98<br />

stress ulcers 80, 98–99<br />

treatment 107–109<br />

see also peptic ulcer disease<br />

(PUD)<br />

ultrasound<br />

abdominal pain investigation<br />

224–225<br />

acute abdomen investigation<br />

679–680<br />

appendicitis 744<br />

gastrointestinal bleeding<br />

investigation 645<br />

intussusception 722–723<br />

pancreas evaluation 310–311<br />

swallowing disorders 242<br />

upper esophageal sphincter (UES)<br />

237–238<br />

manometric evaluation 242<br />

uveitis 394<br />

Vac-A cytotoxin 78<br />

vaccination see immunization<br />

variceal bleeding 649–651<br />

prevention 650<br />

varicella zoster virus (VZV),<br />

esophageal infection 35<br />

vascular lesions 751–759<br />

hemangiomas 753–754<br />

nomenclature errors 753<br />

tumors versus malformations<br />

751–753<br />

vascular malformations<br />

754–759<br />

arteriovenous malformations<br />

758<br />

capillary malformations 755<br />

complex combined<br />

malformations 758–759<br />

lymphatic malformations<br />

757–758<br />

venous malformations 755–757<br />

vasculitides 419–433<br />

classification 419<br />

clinical manifestations 421–422<br />

diagnosis 422–427<br />

arteriography 426–427<br />

endoscopy 425<br />

history 422–423<br />

imaging studies 425<br />

laboratory tests 424–425<br />

physical examination<br />

423–424<br />

tissue biopsy 425–426<br />

epidemiology 420<br />

follow-up 433<br />

frequency of intestinal<br />

involvement 425<br />

in connective tissue diseases<br />

430<br />

pathogenesis 420–421<br />

antineutrophil cytoplasmic<br />

antibodies 421<br />

pathogenetic immunecomplex<br />

formation 420<br />

Index 803<br />

pathogenic T-cell responses<br />

and granuloma formation<br />

421<br />

primary 427–429<br />

protein-losing enteropathy and<br />

456–457<br />

secondary 429–432<br />

treatment 432–433<br />

see also specific disorders<br />

vasoactive intestinal polypeptide<br />

(VIP) 63<br />

vasopressin 649<br />

venous malformations 755–757<br />

diffuse venous malformations<br />

757<br />

focal malformations 755–756<br />

multifocal malformations<br />

756–757<br />

Veress needle 762<br />

very-low-birth-weight (VLBW)<br />

infants see premature infants<br />

Vibrio spp 147<br />

V. cholerae 145–148, 666<br />

videofluoroscopy 241, 243<br />

villous atrophy<br />

diarrhea and 1, 194<br />

HIV infection 115<br />

intestinal epithelial dysplasia 6<br />

microvillus inclusion disease 3<br />

viral infections<br />

epidemiology 127<br />

esophagitis 32–37<br />

following intestinal<br />

transplantation 706–707<br />

gastritides 97<br />

see also diarrhea; specific viruses<br />

visceral hyperalgesia hypothesis<br />

215<br />

visceral myopathy<br />

familial 274–275<br />

with diffuse abnormal muscle<br />

layering 273<br />

infantile 273–274<br />

primary 273<br />

visceral neuropathies 276–280<br />

familial 276–278<br />

with multiple endocrine<br />

neoplasia 277–278<br />

with neurological<br />

involvement 277<br />

with neuronal intranuclear<br />

inclusions 277<br />

with <strong>pylori</strong>c stenosis, short<br />

intestine and malrotation<br />

277<br />

without extraintestinal<br />

manifestations 276–277<br />

sporadic 278–280<br />

acquired visceral<br />

neuropathies 279–280<br />

chronic inflammation/<br />

autoimmune disease 280<br />

hyperganglionosis 279


804<br />

Index<br />

hypoganglionosis 278–279<br />

infectious agents 280<br />

intestinal neuronal dysplasia<br />

279<br />

vitamin A<br />

deficiency in malnutrition 515<br />

diarrhea management 198, 531<br />

with HIV 121<br />

intestinal mucosa protection<br />

531<br />

vitamin B12<br />

deficiency in Crohn’s disease<br />

354<br />

supplementation following ileal<br />

resection 470<br />

vitamin requirements, premature<br />

infants 609–610, 631–632<br />

volvulus 15, 16, 19, 223<br />

treatment 19–20<br />

vomiting 678–679<br />

antiemetic treatment 665–667<br />

obstructive lesions and 15<br />

infantile hypertrophic <strong>pylori</strong>c<br />

stenosis 589<br />

with diarrhea 133<br />

see also cyclic vomiting<br />

syndrome (CVS)<br />

Waardenburg syndrome 265<br />

wasting 494–495<br />

water balance, premature infants<br />

600, 621–622<br />

Wegener’s granulomatosis 428<br />

weight-for height 493–495<br />

wheat allergy 334–335<br />

whipworm 170<br />

see also trichuriasis<br />

Winiwarter–Buerger’s disease 432<br />

wireless capsule video-endoscopy<br />

646<br />

wireworms 180<br />

Wyburn–Mason’s syndrome 755<br />

X-ray see radiography<br />

Yersinia 150–151, 666<br />

Z-score 494<br />

zinc supplementation, diarrhea<br />

and 195, 198, 669<br />

Zollinger–Ellison syndrome (ZES)<br />

101<br />

management 109<br />

zonula occludens toxin (Zot) 147,<br />

148<br />

zonulin 436

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!