13.11.2013 Views

Global Initiative for Chronic Obstructive Lung Disease - GOLD

Global Initiative for Chronic Obstructive Lung Disease - GOLD

Global Initiative for Chronic Obstructive Lung Disease - GOLD

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page A1<br />

<strong>Global</strong> <strong>Initiative</strong> <strong>for</strong> <strong>Chronic</strong><br />

<strong>Obstructive</strong><br />

<strong>Lung</strong><br />

<strong>Disease</strong><br />

GLOBAL STRATEGY FOR THE DIAGNOSIS,<br />

MANAGEMENT, AND PREVENTION OF<br />

CHRONIC OBSTRUCTIVE PULMONARY DISEASE<br />

UPDATED 2005


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page i<br />

GLOBAL INITIATIVE FOR<br />

CHRONIC OBSTRUCTIVE LUNG DISEASE<br />

GLOBAL STRATEGY FOR THE DIAGNOSIS, MANAGEMENT,<br />

AND PREVENTION OF CHRONIC OBSTRUCTIVE PULMONARY DISEASE<br />

Updated 2005 (Based on an April 1998 NHLBI/WHO Workshop)<br />

i


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page ii<br />

APRIL 1998 WORKSHOP PANEL<br />

<strong>Global</strong> Strategy <strong>for</strong> the Diagnosis, Management, and Prevention of<br />

<strong>Chronic</strong> <strong>Obstructive</strong> Pulmonary <strong>Disease</strong>: NHLBI/WHO Workshop<br />

National Heart, <strong>Lung</strong>, and Blood Institute: Claude Lenfant, MD<br />

World Health Organization: Nikolai Khaltaev, MD<br />

Romain Pauwels, MD, PhD, Chair<br />

Ghent University Hospital<br />

Ghent, Belgium<br />

Nicholas Anthonisen, MD<br />

University of Manitoba<br />

Winnipeg, Manitoba, Canada<br />

William C. Bailey, MD<br />

University of Alabama at Birmingham<br />

Birmingham, Alabama, US<br />

Peter J. Barnes, MD<br />

National Heart & <strong>Lung</strong> Institute<br />

London, UK<br />

A. Sonia Buist, MD<br />

Oregon Health Sciences University<br />

Portland, Oregon, US<br />

Peter Calverley, MD<br />

University Hospital, Aintree<br />

Liverpool, UK<br />

Tim Clark, MD<br />

Imperial College<br />

London, UK<br />

Leonardo Fabbri, MD<br />

University of Modena & Reggio Emilia<br />

Modena, Italy<br />

Yoshinosuke Fukuchi, MD<br />

Juntendo University<br />

Tokyo, Japan<br />

Lawrence Grouse, MD, PhD<br />

University of Washington<br />

Seattle, Washington, US<br />

James C. Hogg, MD<br />

St. Paul’s Hospital<br />

Vancouver, British Columbia, Canada<br />

Christine Jenkins, MD<br />

Concord Hospital<br />

Sydney, New South Wales, Australia<br />

Dirkje S. Postma, MD<br />

Academic Hospital Groningen<br />

Groningen, the Netherlands<br />

Klaus F. Rabe, MD<br />

Leiden University Medical Center<br />

Leiden, the Netherlands<br />

Scott D. Ramsey, MD, PhD<br />

University of Washington<br />

Seattle, Washington, US<br />

Stephen I. Rennard, MD<br />

University of Nebraska Medical Center<br />

Omaha, Nebraska, US<br />

Roberto Rodriguez-Roisin, MD<br />

University of Barcelona<br />

Barcelona, Spain<br />

Nikos Siafakas, MD<br />

University of Crete Medical School<br />

Heraklion, Greece<br />

Sean D. Sullivan, PhD<br />

University of Washington<br />

Seattle, Washington, US<br />

Wan-Cheng Tan, MD<br />

National University Hospital<br />

Singapore<br />

<strong>GOLD</strong> Staff<br />

Sarah DeWeerdt<br />

Editor<br />

Seattle, Washington, US<br />

Suzanne S. Hurd, PhD<br />

Scientific Director<br />

Bethesda, Maryland, US<br />

ii


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page iii<br />

ONSULTANT REVIEWERS<br />

(FOR 1998 WORKSHOP PANEL REPORT)<br />

Individuals<br />

Sherwood Burge (UK)<br />

Moira Chan-Yeung (Hong Kong)<br />

James Donohue (US)<br />

Nicholas J. Gross (US)<br />

Helgo Magnussen (Germany)<br />

Donald Mahler (US)<br />

Jean-Francois Muir (France)<br />

Mrigrendra Pandey (India)<br />

Peter Pare (Canada)<br />

Thomas Petty (US)<br />

Michael Plit (South Africa)<br />

Sri Ram (US)<br />

Harold Rea (New Zealand)<br />

Andrea Rossi (Italy)<br />

Maureen Rutten-van Molken (The Netherlands)<br />

Marina Saetta (Italy)<br />

Raj Singh (India)<br />

Frank Speizer (US)<br />

Robert Stockley (UK)<br />

Donald Tashkin (US)<br />

Ian Town (New Zealand)<br />

Paul Vermeire (Belgium)<br />

Gregory Wagner (US)<br />

Scott Weiss (US)<br />

Miel Wouters (The Netherlands)<br />

Jan Zielinski (Poland)<br />

Organizations<br />

American College of Chest Physicians<br />

Suzanne Pingleton<br />

American Thoracic Society<br />

Bart Celli<br />

William Martin<br />

Austrian Respiratory Society<br />

Friedriech Kummer<br />

Arab Respiratory Society<br />

Salem El Sayed<br />

Thoracic Society of Australia and New Zealand<br />

Alastair Stewart<br />

David McKenzie<br />

Peter Frith<br />

Australian <strong>Lung</strong> Foundation<br />

Robert Edwards<br />

Belgian Society of Pneumology<br />

Marc Decramer<br />

Jean-Claude Yernault<br />

British Thoracic Society<br />

Neil Pride<br />

Canadian Thoracic Society<br />

Louis-Philippe Boulet<br />

Kenneth Chapman<br />

Chinese Respiratory Society<br />

Nan-Shan Zhong<br />

Yuanjue Zhu<br />

Croatian Respiratory Society<br />

Neven Rakusic<br />

Davor Plavec<br />

Czech Thoracic Society<br />

Stanislav Kos<br />

Jaromir Musil<br />

Vladimir Vondra<br />

European Respiratory Society<br />

Marc Decramer (Belgium)<br />

French Speaking Pneumological Society<br />

Michel Fournier<br />

Thomas Similowski<br />

Hungarian Respiratory Society<br />

Pal Magyar<br />

Japanese Respiratory Society<br />

Yoshinosuke Fukuchi<br />

Latin American Thoracic Society<br />

Juan Figueroa (Argentina)<br />

Maria Christina Machado (Brazil)<br />

Ilma Paschoal (Brazil)<br />

Jose Jardim (Brazil)<br />

Gisele Borzone (Chile)<br />

Orlando Diaz (Chile)<br />

iii


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page iv<br />

Patricio Gonzales (Chile)<br />

Carmen Lisboa (Chile)<br />

Rogelio Perez Padilla (Mexico)<br />

Jorge Rodriguez De Marco (Uruguay)<br />

Maria Victorina Lopez (Uruguay)<br />

Roberto Lopez (Uruguay)<br />

Malaysian Thoracic Society<br />

Zin Zainudin<br />

Norwegian Thoracic Society<br />

Amund Gulsvik<br />

Ernst Omenaas<br />

Polish Phthisiopneumonological Society<br />

Michal Pirozynski<br />

Romanian Society of Pulmonary <strong>Disease</strong>s<br />

Traian Mihaescu<br />

Sabina Antoniu<br />

Singapore Thoracic Society<br />

Alan Ng<br />

Wei Keong<br />

Slovakian Pneumological and Phthisiological Society<br />

Ladislav Chovan<br />

Slovenian Respiratory Society<br />

Stanislav Suskovic<br />

South African Thoracic Society<br />

James Joubert<br />

Spanish Society of Pneumology<br />

Teodoro Montemayor Rubio<br />

Victor Sobradillo<br />

Swedish Society <strong>for</strong> Chest Physicians<br />

Kjell Larsson<br />

Sven Larsson<br />

Claes-Goran Lofdahl<br />

Swiss Pulmonary Society<br />

Philippe Leuenberger<br />

Erich Russi<br />

Thoracic Society of Thailand<br />

Ploysongsang Youngyudh<br />

Vietnam Asthma-Allergology and Clinical<br />

Immunology Association<br />

Nguyen Nang An<br />

iv


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page v<br />

PREFACE<br />

<strong>Chronic</strong> <strong>Obstructive</strong> Pulmonary <strong>Disease</strong> (COPD) is a<br />

major public health problem. It is the fourth leading<br />

cause of chronic morbidity and mortality in the United<br />

States 1 and is projected to rank fifth in 2020 as a worldwide<br />

burden of disease according to a study published by<br />

the World Bank/World Health Organization 2 . Yet, COPD<br />

fails to receive adequate attention from the health care<br />

community and government officials. With these concerns<br />

in mind, a committed group of scientists encouraged<br />

the US National Heart, <strong>Lung</strong>, and Blood Institute<br />

and the World Health Organization to <strong>for</strong>m the <strong>Global</strong><br />

<strong>Initiative</strong> <strong>for</strong> <strong>Chronic</strong> <strong>Obstructive</strong> <strong>Lung</strong> <strong>Disease</strong> (<strong>GOLD</strong>).<br />

Among <strong>GOLD</strong>’s important objectives are to increase<br />

awareness of COPD and to help the thousands of people<br />

who suffer from this disease and die prematurely from<br />

COPD or its complications.<br />

The first step in the <strong>GOLD</strong> program was to prepare a<br />

consensus Workshop Report, <strong>Global</strong> Strategy <strong>for</strong> the<br />

Diagnosis, Management, and Prevention of COPD. The<br />

<strong>GOLD</strong> Expert Panel, a distinguished group of health<br />

professionals from the fields of respiratory medicine,<br />

epidemiology, socioeconomics, public health, and health<br />

education, reviewed existing COPD guidelines, as well as<br />

new in<strong>for</strong>mation on pathogenic mechanisms of COPD as<br />

they developed a consensus document. Many recommendations<br />

will require additional study and evaluation as<br />

the <strong>GOLD</strong> program is implemented.<br />

Development of the Workshop Report was supported<br />

through educational grants from Altana, Andi-Ventis,<br />

AstraZeneca, Aventis, Bayer, Boehringer Ingelheim,<br />

Chiesi, GlaxoSmithKline, Merck, Sharp & Dohme,<br />

Mitsubishi Pharma, Nikken Chemicals, Novartis, Pfizer,<br />

Schering-Plough, and Zambon.<br />

Leonardo Fabbri, MD<br />

Modena, Italy<br />

Chair, <strong>GOLD</strong> Executive Committee<br />

REFERENCES<br />

1. National Heart, <strong>Lung</strong>, and Blood Institute. Morbidity &<br />

mortality: chartbook on cardiovascular, lung, and blood diseases.<br />

Bethesda, MD: US Department of Health and Human<br />

Services, Public Health Service, National Institutes of Health;<br />

1998. Available from: URL:<br />

www.nhlbi.nih.gov/nhlbi/seiin//other/cht-book/htm<br />

2. Murray CJL, Lopez AD. Evidence-based health<br />

policy-lessons from the <strong>Global</strong> Burden of <strong>Disease</strong> Study.<br />

Science 1996; 274:740-3.<br />

A major problem is the incomplete in<strong>for</strong>mation about the<br />

causes and prevalence of COPD, especially in developing<br />

countries. While cigarette smoking is a major known risk<br />

factor, much remains to be learned about other causes of<br />

this disease. The <strong>GOLD</strong> <strong>Initiative</strong> will bring COPD to the<br />

attention of governments, public health officials, health<br />

care workers, and the general public, but a concerted<br />

ef<strong>for</strong>t by all involved in health care will be necessary to<br />

control this major public health problem.<br />

I would like to acknowledge the expert panel that prepared<br />

the first workshop report, and the <strong>GOLD</strong> Science Committee<br />

<strong>for</strong> its work in preparation of the yearly updated volumes.<br />

We look <strong>for</strong>ward to our continued work with interested<br />

organizations and the <strong>GOLD</strong> National Leaders to meet<br />

the goals of the <strong>GOLD</strong> initiative.<br />

v


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page vi<br />

Methodology and Summary of New Recommendations (2005 Update)<br />

The <strong>GOLD</strong> Workshop Report, <strong>Global</strong> Strategy <strong>for</strong><br />

Diagnosis, Management and Prevention of COPD<br />

presented in 2001 was based on the scientific literature<br />

available until mid 2000.. To assure that the recommendations<br />

<strong>for</strong> management of COPD remained as current<br />

as possible, the <strong>GOLD</strong> Executive Committee established<br />

a Science Committee* to review published research and<br />

to post an updated report yearly on the <strong>GOLD</strong> website.<br />

The first (2003) and second (2004) updates were posted<br />

on the <strong>GOLD</strong> website (www.gold.copd.org) in July 2003<br />

and July 2004 respectively. This third update (July 2005)<br />

includes review of publications from January to<br />

December 2004. This will be the final update of the 2001<br />

document; a revision of the entire document has been<br />

implemented and is scheduled to be completed in 2006.<br />

Methods: The process used <strong>for</strong> the 2005 update, identical<br />

to that described <strong>for</strong> the previous updates, included a<br />

Pub Med search using fields established by the<br />

Committee: 1) COPD OR chronic bronchitis OR emphysema,<br />

All Fields, All Adult, 19+ years, only items with<br />

abstracts, Clinical Trial, Human, sorted by Authors; and<br />

2) COPD OR chronic bronchitis OR emphysema AND<br />

systematic, All fields, All adult, 19+ years, only items<br />

with abstracts, Human, sorted by Author. In addition,<br />

publications in peer review journals not captured by Pub<br />

Med could be submitted to individual members of the<br />

Committee providing an abstract and the full paper were<br />

submitted in (or translated into) English.<br />

All members of the Committee received a summary of<br />

citations and all abstracts. Each abstract was assigned<br />

to 2 Committee members (members were not assigned<br />

to a paper where he/she appears as an author), although<br />

any member was offered the opportunity to provide an<br />

opinion on any abstract. Members evaluated the abstract<br />

or, up to her/his judgment, the full publication, by answering<br />

specific written questions from a short questionnaire, and<br />

to indicate if the scientific data presented impacted on<br />

recommendations in the <strong>GOLD</strong> report. If so, the member<br />

was asked to specifically identify modifications that<br />

should be made. The <strong>GOLD</strong> Science Committee met<br />

on a regular basis to discuss each individual publication<br />

indicated by at least 1 member of the Committee to have<br />

an impact on COPD management, and to reach a<br />

consensus on the changes in the report. Disagreements<br />

were decided by vote.<br />

*Members: K. Rabe, Chair; P. Barnes, S. Buist, P. Calverley,<br />

L. Fabbri, Y. Fukuchi, W. MacNee, R. Rodriguez-Roisin, I. Zielinski<br />

<strong>GOLD</strong> Workshop Report (2005 Update):<br />

Summary of Recommendations<br />

Between January 1 and December 2004, 131 articles met<br />

the search criteria. Of these, 10 papers were identified to<br />

have an impact on the <strong>GOLD</strong> report. Of these, 5 papers<br />

confirmed an existing statement and were added as a<br />

reference:<br />

1. Page 68: Man WD, Mustfa N, Nikoletou D, Kaul S,<br />

Hart N, Rafferty GF, Donaldson N, Polkey MI, Moxham J.<br />

Effect of salmeterol on respiratory muscle activity during<br />

exercise in poorly reversible COPD. Thorax. 2004<br />

Jun;59(6):471-6.<br />

2. Page 68: O'Donnell DE, Fluge T, Gerken F, Hamilton<br />

A, Webb K, Aguilaniu B, Make B, Magnussen H. Effects<br />

of tiotropium on lung hyperinflation, dyspnoea and exercise<br />

tolerance in COPD. Eur Respir J. 2004 Jun;23(6):832-40.<br />

3. Page 68: Oostenbrink JB, Rutten-van Molken MP, Al<br />

MJ, Van Noord JA, Vincken W. One-year cost-effectiveness<br />

of tiotropium versus ipratropium to treat chronic obstructive<br />

pulmonary disease. Eur Respir J. 2004 Feb;23(2):241-9.<br />

4. Page 71: Spencer S, Calverley PM, Burge PS, Jones<br />

PW. Impact of preventing exacerbations on deterioration<br />

of health status in COPD. Eur Respir J. 2004<br />

May;23(5):698-702.<br />

5. Page 73: Wongsurakiat P, Maranetra KN, Wasi C,<br />

Kositanont U, Dejsomritrutai W, Charoenratanakul S.<br />

Acute respiratory illness in patients with COPD and the<br />

effectiveness of influenza vaccination: a randomized<br />

controlled study. Chest. 2004 Jun;125(6):2011-20.<br />

Five papers introduced in<strong>for</strong>mation that required a new<br />

statement to be added to the report:<br />

1. Page 67 – Add sentence: In a study of mild to moderate<br />

COPD patients at an out-patient clinic, patient education<br />

involving one four hour group session followed by one to<br />

two individual nurse- and physiotherapist-sessions<br />

improved patient outcomes and reduced costs in a 12-<br />

month follow-up.<br />

Reference: Gallefoss F. The effects of patient education<br />

in COPD in a 1-year follow-up randomised, controlled<br />

trial. Patient Educ Couns. 2004 Mar;52(3):259-66.<br />

vi


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page vii<br />

2. Page 70 - Add sentence: Twenty-one days of inhaled<br />

tiotropium, 18 mg/day as a dry powder does not reduce<br />

mucus clearance from the lungs.<br />

Reference: Hasani A, Toms N, Agnew JE, Sarno M,<br />

Harrison AJ, Dilworth P. The effect of inhaled tiotropium<br />

bromide on lung mucociliary clearance in patients with<br />

COPD. Chest. 2004 May;125(5):1726-34.<br />

3. Page 71 – Add sentence: Short-term treatment with<br />

a combined inhaled glucocorticosteroid and long-acting<br />

ß 2 -agonist resulted in greater control of lung function and<br />

symptoms than combined anticholinergic and short-acting<br />

b2-agonist.<br />

Reference: Donohue JF, Kalberg C, Emmett A, Merchant<br />

K, Knobil K. A short-term comparison of fluticasone<br />

propionate/ salmeterol with ipratropium bromide/albuterol<br />

<strong>for</strong> the treatment of COPD. Treat Respir Med.<br />

2004;3(3):173-81.<br />

4. Page 73 - Change paragraph on immunoregulators to<br />

read: Studies using an immunostimulator in COPD show<br />

a decrease in the severity and frequency of exacerbations 94<br />

(add new reference). However, additional studies to<br />

examine the long term effects of this therapy are required<br />

be<strong>for</strong>e regular use can be recommended (Evidence B).<br />

Reference: Li J, Zheng JP, Yuan JP, Zeng GQ, Zhong<br />

NS, Lin CY. Protective effect of a bacterial extract against<br />

acute exacerbation in patients with chronic bronchitis<br />

accompanied by chronic obstructive pulmonary disease.<br />

Chin Med J (Engl). 2004 Jun;117(6):828-34.<br />

An Appendix includes a report on outcome measures<br />

<strong>for</strong> COPD to encourage comments and input from the<br />

scientific community to prepare <strong>for</strong> the full revision of the<br />

report, scheduled to appear in mid-2006. The many<br />

individuals who participated in preparation of this report<br />

are listed in the document. The <strong>GOLD</strong> Science<br />

Committee is grateful to those who contributed to this<br />

report, particularly the work of Dr. Paul Jones, London,<br />

England and Dr. Alvar Agusti, Palma de Mallorca, Spain.<br />

The proposed modifications <strong>for</strong> the <strong>GOLD</strong> Workshop<br />

Report (Updated 2005) were approved by the <strong>GOLD</strong><br />

Executive Committee.<br />

The <strong>GOLD</strong> Workshop Report (Updated 2005), the <strong>GOLD</strong><br />

Executive Summary(Updated 2005), and the <strong>GOLD</strong><br />

Pocket Guide (Updated 2005) along with the complete list<br />

of references examined by the Committee are available<br />

on the <strong>GOLD</strong> website (www.goldcopd.org).<br />

5. Page 93 - Add sentence: Early outpatient pulmonary<br />

rehabilitation after hospitalization <strong>for</strong> COPD exacerbation<br />

results in exercise capacity and health status improvements<br />

at three months.<br />

Reference: Man WD, Polkey MI, Donaldson N, Gray BJ,<br />

Moxham J. Community pulmonary rehabilitation after<br />

hospitalisation <strong>for</strong> acute exacerbations of chronic obstructive<br />

pulmonary disease: randomised controlled study. BMJ.<br />

2004 Nov 20;329(7476):1209.<br />

A major new segment appears in Chapter 5-4 on antibiotics<br />

in treatment of COPD exacerbations (page 94). The<br />

material was prepared by the <strong>GOLD</strong> Science Committee<br />

which gratefully acknowledges the opportunity to review<br />

a statement on this topic prepared by the European<br />

Respiratory Society and provided to the Committee by<br />

Dr. William MacNee and Dr. Mark Woodhead. Prior to its<br />

release, the material was reviewed by Dr. Sanjay Sethi,<br />

State University of New York at Buffalo, Buffalo, New York,<br />

and Dr. Antonio Anzueto, University of San Antonio, San<br />

Antonio, Texas.<br />

vii


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page viii<br />

TABLE OF CONTENTS<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .1<br />

1. Definition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .5<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . . .6<br />

Definition . . . . . . . . . . . . . . . . . . . . . . . . . . .6<br />

Natural History . . . . . . . . . . . . . . . . . . . . . . .7<br />

Classification of Severity . . . . . . . . . . . . . . .7<br />

Variable Course of COPD . . . . . . . . . . . . . .8<br />

Scope of the Report . . . . . . . . . . . . . . . . . . .9<br />

Asthma and COPD . . . . . . . . . . . . . . . . . . .9<br />

Pulmonary Tuberculosis and COPD . . . . . .9<br />

References . . . . . . . . . . . . . . . . . . . . . . . . . .9<br />

2. Burden of COPD . . . . . . . . . . . . . . . . . . . . . . . . .11<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .12<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .12<br />

Epidemiology . . . . . . . . . . . . . . . . . . . . . . .12<br />

Prevalence . . . . . . . . . . . . . . . . . . . . . . . .12<br />

Morbidity . . . . . . . . . . . . . . . . . . . . . . . . . .14<br />

Mortality . . . . . . . . . . . . . . . . . . . . . . . . . .14<br />

Economic and Social Burden of COPD . . . .15<br />

Economic Burden . . . . . . . . . . . . . . . . . . .15<br />

Social Burden . . . . . . . . . . . . . . . . . . . . . .16<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .17<br />

3. Risk Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . .19<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .20<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .20<br />

Host Factors . . . . . . . . . . . . . . . . . . . . . . . .21<br />

Genes . . . . . . . . . . . . . . . . . . . . . . . . . . . .21<br />

Airway Hyperresponsiveness . . . . . . . . . . .21<br />

<strong>Lung</strong> Growth . . . . . . . . . . . . . . . . . . . . . . .21<br />

Exposures . . . . . . . . . . . . . . . . . . . . . . . . . .21<br />

Tobacco Smoke . . . . . . . . . . . . . . . . . . . .21<br />

Occupational Dusts and Chemicals . . . . . .22<br />

Outdoor and Indoor Air Pollution . . . . . . . .22<br />

Infections . . . . . . . . . . . . . . . . . . . . . . . . .22<br />

Socioeconomic Status . . . . . . . . . . . . . . . .23<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .23<br />

4. Pathogenesis, Pathology, and<br />

Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . .27<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .28<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .28<br />

Pathogenesis . . . . . . . . . . . . . . . . . . . . . . .28<br />

Inflammatory Cells . . . . . . . . . . . . . . . . . .29<br />

Inflammatory Mediators . . . . . . . . . . . . . . .30<br />

Differences Between Inflammation in<br />

COPD and Asthma . . . . . . . . . . . . . . . . .31<br />

Inflammation and COPD Risk Factors . . . . . . .32<br />

Proteinase-Antiproteinase Imbalance . . . . . . .32<br />

Oxidative Stress . . . . . . . . . . . . . . . . . . . .33<br />

Pathology . . . . . . . . . . . . . . . . . . . . . . . . . .33<br />

Central Airways . . . . . . . . . . . . . . . . . . . . .33<br />

Peripheral Airways . . . . . . . . . . . . . . . . . .34<br />

<strong>Lung</strong> Parenchyma . . . . . . . . . . . . . . . . . . .35<br />

Pulmonary Vasculature . . . . . . . . . . . . . . .36<br />

Pathophysiology . . . . . . . . . . . . . . . . . . . . .36<br />

Mucus Hypersecretion and<br />

Ciliary Dysfunction . . . . . . . . . . . . . . . . .36<br />

Airflow Limitation and<br />

Pulmonary Hyperinflation . . . . . . . . . . . .36<br />

Gas Exchange Abnormalities . . . . . . . . . . . .37<br />

Pulmonary Hypertension and<br />

Cor Pulmonale . . . . . . . . . . . . . . . . . . . .38<br />

Systemic Effects . . . . . . . . . . . . . . . . . . . .38<br />

Pathophysiology and the<br />

Symptoms of COPD . . . . . . . . . . . . . . . .38<br />

Pathology and Pathophysiology of<br />

Exacerbations . . . . . . . . . . . . . . . . . . . . . .39<br />

Pathology . . . . . . . . . . . . . . . . . . . . . . . . .39<br />

Pathophysiology . . . . . . . . . . . . . . . . . . . .39<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .39<br />

5. Management of COPD . . . . . . . . . . . . . . . . . . . . .45<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .46<br />

Component 1: Assess and Monitor <strong>Disease</strong> . . .47<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .47<br />

Initial Diagnosis . . . . . . . . . . . . . . . . . . . . . .47<br />

Assessment of Symptoms . . . . . . . . . . . . . . .47<br />

Medical History . . . . . . . . . . . . . . . . . . . . .49<br />

Physical Examination . . . . . . . . . . . . . . . .50<br />

Measurement of Airflow Limitation . . . . . . . .50<br />

Assessment of Severity . . . . . . . . . . . . . . .51<br />

Additional Investigations . . . . . . . . . . . . . .52<br />

Differential Diagnosis . . . . . . . . . . . . . . . .53<br />

Ongoing Monitoring and<br />

Assessment . . . . . . . . . . . . . . . . . . . . . . .53<br />

Monitor <strong>Disease</strong> Progression and<br />

Development of Complications . . . . . . . .54<br />

Monitor Pharmacotherapy and<br />

Other Medical Treatment . . . . . . . . . . . . .55<br />

Monitor Exacerbation History . . . . . . . . . .55<br />

Monitor Comorbidities . . . . . . . . . . . . . . . .55<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .56<br />

viii


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page ix<br />

Component 2: Reduce Risk Factors . . . . . . . . . .58<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .58<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .58<br />

Tobacco Smoke . . . . . . . . . . . . . . . . . . . . .58<br />

Smoking Prevention . . . . . . . . . . . . . . . . .58<br />

Smoking Cessation . . . . . . . . . . . . . . . . . .58<br />

Occupational Exposures . . . . . . . . . . . . . . .62<br />

Indoor/Outdoor Air Pollution . . . . . . . . . . . .62<br />

Regulation of Air Quality . . . . . . . . . . . . . .62<br />

Patient-Oriented Control . . . . . . . . . . . . . .62<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .63<br />

Component 3: Manage Stable COPD . . . . . . . . .65<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .65<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .65<br />

Education . . . . . . . . . . . . . . . . . . . . . . . . . .65<br />

Goals and Educational Strategies . . . . . . .66<br />

Components of an Education Program . . .66<br />

Cost Effectiveness of Education<br />

Programs <strong>for</strong> COPD Patients . . . . . . . . . .67<br />

Pharmacologic Treatment . . . . . . . . . . . . . .67<br />

Overview of the Medications . . . . . . . . . . .67<br />

Bronchodilators . . . . . . . . . . . . . . . . . . . . .67<br />

Glucocorticosteroids . . . . . . . . . . . . . . . . .71<br />

Pharmacologic Therapy by <strong>Disease</strong> Severity . .72<br />

Other Pharmacologic Treatments . . . . . . .73<br />

Non-Pharmacologic Treatment . . . . . . . . . . .74<br />

Rehabilitation . . . . . . . . . . . . . . . . . . . . . .74<br />

Oxygen Therapy . . . . . . . . . . . . . . . . . . . .76<br />

Ventilatory Support . . . . . . . . . . . . . . . . . .77<br />

Surgical Treatments . . . . . . . . . . . . . . . . . .78<br />

Special Considerations . . . . . . . . . . . . . . .79<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .80<br />

Component 4: Manage Exacerbations . . . . . .88<br />

Key Points . . . . . . . . . . . . . . . . . . . . . . . . . .88<br />

Introduction . . . . . . . . . . . . . . . . . . . . . . . . .88<br />

Diagnosis and Assessment of Severity . . . .88<br />

Medical History . . . . . . . . . . . . . . . . . . . . .88<br />

Assessment of Severity . . . . . . . . . . . . . . .89<br />

Home Management . . . . . . . . . . . . . . . . . . .89<br />

Bronchodilator Therapy . . . . . . . . . . . . . . .90<br />

Glucocorticosteroids . . . . . . . . . . . . . . . . .90<br />

Hospital Management . . . . . . . . . . . . . . . . .91<br />

Emergency Department or Hospital . . . . . .91<br />

Hospital Discharge and Follow-Up . . . . . . .93<br />

Antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . .94<br />

References . . . . . . . . . . . . . . . . . . . . . . . . .96<br />

6. Future Research . . . . . . . . . . . . . . . . . . . . . . . . . .99<br />

APPENDIX<br />

Outcomes and Markers in COPD . . . . . . . .103<br />

ix


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page x<br />

x


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 1<br />

INTRODUCTION<br />

<strong>Chronic</strong> <strong>Obstructive</strong> Pulmonary <strong>Disease</strong> (COPD) is<br />

a major cause of chronic morbidity and mortality<br />

throughout the world. Many people suffer from this<br />

disease <strong>for</strong> years and die prematurely from it or its<br />

complications. COPD is currently the fourth leading<br />

cause of death in the world 1 , and further increases in its<br />

prevalence and mortality can be predicted in the coming<br />

decades 2 . A unified international ef<strong>for</strong>t is needed to<br />

reverse these trends.<br />

The <strong>Global</strong> <strong>Initiative</strong> <strong>for</strong> <strong>Chronic</strong> <strong>Obstructive</strong> <strong>Lung</strong><br />

<strong>Disease</strong> (<strong>GOLD</strong>) is conducted in collaboration with the<br />

US National Heart, <strong>Lung</strong>, and Blood Institute (NHLBI)<br />

and the World Health Organization (WHO). Its goals are<br />

to increase awareness of COPD and decrease morbidity<br />

and mortality from the disease. <strong>GOLD</strong> aims to improve<br />

prevention and management of COPD through a concerted<br />

worldwide ef<strong>for</strong>t of people involved in all facets of health<br />

care and health care policy, and to encourage a renewed<br />

research interest in this highly prevalent disease.<br />

A nihilistic attitude toward COPD has arisen among<br />

some health care providers, due to the relatively limited<br />

success of primary and secondary prevention (i.e.,<br />

avoidance of factors that cause COPD or its progression),<br />

the prevailing notion that COPD is largely a self-inflicted<br />

disease, and disappointment with available treatment<br />

options. The <strong>GOLD</strong> project will work toward combating<br />

this nihilistic attitude by disseminating in<strong>for</strong>mation about<br />

available treatments, both pharmacologic and nonpharmacologic.<br />

Tobacco smoking is a major cause of COPD, as well as<br />

of many other diseases. A decline in tobacco smoking<br />

would result in substantial health benefits and a decrease<br />

in the prevalence of COPD and other smoking-related<br />

diseases. There is an urgent need <strong>for</strong> improved strategies<br />

to decrease tobacco consumption. However, tobacco<br />

smoking is not the only cause of COPD and may not<br />

even be the major cause in some parts of the world.<br />

Furthermore, not all smokers develop clinically significant<br />

COPD, which suggests that additional factors are<br />

involved in determining each individual's susceptibility.<br />

Thus, investigation of COPD risk factors and ways to<br />

reduce exposure to these factors is also an important<br />

area <strong>for</strong> future research. New research tools have<br />

recently revealed that inflammation plays a prominent<br />

role in COPD pathogenesis, but this inflammation is<br />

different than that involved in asthma. Further study of<br />

the molecular and cellular mechanisms involved in COPD<br />

pathogenesis should lead to effective treatments that<br />

slow or halt the course of the disease.<br />

<strong>GOLD</strong> WORKSHOP REPORT:<br />

GLOBAL STRATEGY FOR THE<br />

DIAGNOSIS, MANAGEMENT, AND<br />

PREVENTION OF COPD<br />

One strategy to help achieve <strong>GOLD</strong>'s objectives is to<br />

provide health care workers, health care authorities,<br />

and the general public with state-of-the-art in<strong>for</strong>mation<br />

about COPD and specific recommendations on the most<br />

appropriate management and prevention strategies.<br />

The <strong>GOLD</strong> Workshop Report, <strong>Global</strong> Strategy <strong>for</strong> the<br />

Diagnosis, Management, and Prevention of COPD, is<br />

based on the best-validated current concepts of COPD<br />

pathogenesis and the available evidence on the most<br />

appropriate management and prevention strategies.<br />

The Report has been developed by individuals with<br />

expertise in COPD research and patient care and extensively<br />

reviewed by many experts and scientific societies.<br />

It provides state-of-the-art in<strong>for</strong>mation about COPD <strong>for</strong><br />

pulmonary specialists and other interested physicians.<br />

The document will also serve as a source <strong>for</strong> the production<br />

of various communications during the implementation<br />

of the <strong>GOLD</strong> program, including a practical guide <strong>for</strong><br />

primary care physicians and a document <strong>for</strong> use in<br />

developing countries.<br />

The <strong>GOLD</strong> Report is not intended to be a comprehensive<br />

textbook on COPD, but rather to summarize the current<br />

state of the field. Each chapter starts with Key Points<br />

that crystallize current knowledge. The chapters on the<br />

Burden of COPD and Risk Factors demonstrate the<br />

global importance of COPD and the various causal<br />

factors involved. The chapter on Pathogenesis,<br />

Pathology, and Pathophysiology documents the current<br />

understanding of, and remaining questions about, the<br />

mechanism(s) that lead to COPD, as well as the<br />

structural and functional abnormalities of the lungs<br />

characteristic of the disease.<br />

A major part of the <strong>GOLD</strong> Workshop Report is devoted<br />

to the clinical Management of COPD and presents a<br />

management plan with four components: (1) Assess<br />

and Monitor <strong>Disease</strong>; (2) Reduce Risk Factors;<br />

(3) Manage Stable COPD; (4) Manage Exacerbations.<br />

INTRODUCTION 1


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 2<br />

Management recommendations are largely symptom<br />

driven and are presented according to the severity of<br />

the disease, using a simple classification of severity to<br />

facilitate the practical implementation of the available<br />

management options. Where appropriate, in<strong>for</strong>mation<br />

about health education <strong>for</strong> patients is included.<br />

The final chapter identifies critical gaps in knowledge<br />

requiring Further Research and provides a summary of<br />

proposed directions <strong>for</strong> the development of new therapeutic<br />

approaches.<br />

METHODS USED TO DEVELOP<br />

THIS REPORT<br />

In January 1997, COPD experts from several countries<br />

met in Brussels, Belgium to explore the development of a<br />

<strong>Global</strong> <strong>Initiative</strong> <strong>for</strong> <strong>Chronic</strong> <strong>Obstructive</strong> <strong>Lung</strong> <strong>Disease</strong>.<br />

Dr. Romain Pauwels served as Chair; representatives of<br />

the NHLBI and WHO attended. Participants agreed that<br />

the project was timely and important, and recommended<br />

the establishment of a panel with expertise on a wide<br />

variety of COPD-related topics to prepare an evidencebased<br />

document on diagnosis, management, and<br />

prevention of COPD. NHLBI and WHO staff, in concert<br />

with Dr. Pauwels, identified individuals from many regions<br />

of the world to serve on the Expert Panel, which included<br />

health professionals in the areas of respiratory medicine,<br />

epidemiology, pathology, socioeconomics, public health,<br />

and health education.<br />

The first step toward developing the Workshop Report<br />

was to review the multiple COPD guidelines already<br />

published. The NHLBI collected these guidelines and<br />

prepared a summary table of similarities and differences<br />

between the documents. Where agreement existed, the<br />

Expert Panel drew on these existing documents <strong>for</strong> use<br />

in the Workshop Report. Where major differences existed,<br />

the Expert Panel agreed to carefully examine the scientific<br />

evidence to reach an independent conclusion.<br />

In September 1997, several members of the Expert Panel<br />

met with a consultant to develop a comprehensive set<br />

of terms to build a database of COPD literature. The<br />

database and a computer program to search the world<br />

literature on COPD have been developed, and they will<br />

be placed on the Internet and cross-referenced with the<br />

Workshop Report to help keep the Report current as new<br />

literature is published.<br />

In April 1998, the NHLBI and WHO cosponsored a workshop<br />

to begin the development of the Report. Workshop<br />

participants were divided into three groups: definition and<br />

natural history, chaired by Dr. Sonia Buist; pathophysiology,<br />

risk factors, diagnosis, and classification of severity,<br />

chaired by Dr. Leonardo Fabbri; and management,<br />

chaired by Dr. Romain Pauwels. A table of contents<br />

was developed and writing assignments were made.<br />

The Panel agreed that clinical recommendations would<br />

require scientific evidence, or would be clearly labeled as<br />

"expert opinion." Each chapter would contain a set of the<br />

most current and representative references.<br />

In September 1998, the Panel met to evaluate its<br />

progress. Members reviewed a variety of evidence tables<br />

and chose to assign levels of evidence to statements<br />

using the system developed by the NHLBI (Figure A).<br />

Levels of evidence are assigned to management<br />

recommendations where appropriate in Chapter 5,<br />

Management of COPD, and are indicated in boldface<br />

type enclosed in parentheses after the relevant statement<br />

- e.g., (Evidence A). The methodological issues<br />

concerning the use of evidence from meta-analyses were<br />

carefully considered (e.g., a meta-analysis of a number<br />

of smaller studies considered to be evidence level B) 2 .<br />

The panel met in May 1999, September 1999, and May<br />

2000 in conjunction with meetings of the American<br />

Thoracic Society (ATS) and the European Respiratory<br />

Society (ERS). Symposia were held at these meetings to<br />

present the developing program and to solicit opinion and<br />

comments. The meeting in May 2000 was the final<br />

consensus workshop.<br />

After this workshop, the document was submitted <strong>for</strong><br />

review to individuals and medical societies interested in<br />

the management of COPD. The reviewers' comments<br />

were incorporated, as appropriate, into the final document<br />

by the Chair in cooperation with members of the<br />

Expert Panel. Prior to its release <strong>for</strong> publication, the<br />

Report was reviewed by the NHLBI and the WHO.<br />

A workshop was held in September, 2000 to begin<br />

implementation of the <strong>GOLD</strong> program.<br />

2 INTRODUCTION


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 3<br />

Figure A. Description of Levels of Evidence<br />

Evidence Category Sources of Evidence Definition<br />

A<br />

Randomized controlled<br />

trials (RCTs).<br />

Rich body of data.<br />

Evidence is from endpoints of well-designed RCTs that<br />

provide a consistent pattern of findings in the population<br />

<strong>for</strong> which the recommendation is made. Category A requires<br />

substantial numbers of studies involving substantial numbers<br />

of participants.<br />

B<br />

Randomized controlled<br />

trials (RCTs). Limited<br />

body of data.<br />

Evidence is from endpoints of intervention studies that<br />

include only a limited number of patients, posthoc or<br />

subgroup analysis of RCTs, or meta-analysis of RCTs.<br />

In general, Category B pertains when few randomized trials<br />

exist, they are small in size, they were undertaken in a<br />

population that differs from the target population of the<br />

recommendation, or the results are somewhat inconsistent.<br />

C<br />

Nonrandomized trials.<br />

Observational studies.<br />

Evidence is from outcomes of uncontrolled or nonrandomized<br />

trials or from observational studies.<br />

D<br />

Panel Consensus<br />

Judgment.<br />

This category is used only in cases where the provision of<br />

some guidance was deemed valuable but the clinical literature<br />

addressing the subject was deemed insufficient to justify<br />

placement in one of the other categories. The Panel<br />

Consensus is based on clinical experience or knowledge that<br />

does not meet the above-listed criteria.<br />

REFERENCES<br />

1. World Health Organization. World health report. Geneva: World Health Organization; 2000.<br />

Available from: URL: http://www.who.int/whr/2000/en/statistics.htm<br />

2. Murray CJL, Lopez AD. Evidence-based health policy - lessons from the <strong>Global</strong> Burden of <strong>Disease</strong> Study.<br />

Science 1996; 274:740-3.<br />

INTRODUCTION 3


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 4<br />

4 INTRODUCTION


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 5<br />

CHAPTER<br />

1<br />

DEFINITION


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 6<br />

CHAPTER 1: DEFINITION<br />

KEY POINTS:<br />

• COPD is a disease state characterized by airflow<br />

limitation that is not fully reversible. The airflow<br />

limitation is usually both progressive and associated<br />

with an abnormal inflammatory response of<br />

the lungs to noxious particles or gases.<br />

• The four-stage classification of COPD severity<br />

used throughout this report provides an<br />

educational tool and a general indication of the<br />

approach to management. This conceptual<br />

framework also emphasizes that COPD is usually<br />

progressive if exposure to the noxious agent is<br />

continued.<br />

• The characteristic symptoms of COPD are<br />

cough, sputum production, and dyspnea upon<br />

exertion.<br />

• <strong>Chronic</strong> cough and sputum production often<br />

precede the development of airflow limitation<br />

by many years and these symptoms identify<br />

individuals at risk of developing COPD.<br />

• The focus of this Workshop Report is primarily<br />

on COPD caused by inhaled particles and gases,<br />

the most common of which worldwide is tobacco<br />

smoke.<br />

• COPD can coexist with asthma, the other major<br />

chronic obstructive airway disease characterized<br />

by an underlying airway inflammation. However,<br />

the inflammation characteristic of COPD is distinct<br />

from that of asthma.<br />

• Pulmonary tuberculosis may affect lung function<br />

and symptomatology and, in areas where<br />

tuberculosis is prevalent, can lead to confusion<br />

in the diagnosis of COPD.<br />

DEFINITION<br />

For years, clinicians, physiologists, pathologists, and<br />

epidemiologists have struggled with the definitions of<br />

disorders associated with chronic airflow limitation,<br />

including chronic bronchitis, emphysema, chronic<br />

obstructive pulmonary disease (COPD), and asthma. The<br />

definitions of these terms variably emphasize structure<br />

and function and are often based on whether the term is<br />

used <strong>for</strong> clinical or research purposes. For example,<br />

epidemiologists have created terminology and criteria,<br />

based on functional status, that can be monitored in<br />

population-based studies or studies of physicians'<br />

diagnoses 1,2 .<br />

Based on current knowledge, a working definition of COPD<br />

is a disease state characterized by airflow limitation that<br />

is not fully reversible. The airflow limitation is usually both<br />

progressive and associated with an abnormal inflammatory<br />

response of the lungs to noxious particles or gases.<br />

Symptoms, functional abnormalities, and complications of<br />

COPD can all be explained on the basis of this underlying<br />

inflammation and the resulting pathology (Figure 1-1).<br />

Figure 1-1. Mechanisms Underlying Airflow<br />

Limitation in COPD<br />

Small airway disease<br />

INFLAMMATION<br />

Parenchymal destruction<br />

AIRFLOW LIMITATION<br />

The chronic airflow limitation characteristic of COPD is<br />

caused by a mixture of small airway disease (obstructive<br />

bronchiolitis) and parenchymal destruction (emphysema),<br />

the relative contributions of which vary from person to<br />

person. <strong>Chronic</strong> inflammation causes remodeling and<br />

narrowing of the small airways. Destruction of the lung<br />

parenchyma, also by inflammatory processes, leads to<br />

the loss of alveolar attachments to the small airways and<br />

decreases lung elastic recoil; in turn, these changes diminish<br />

the ability of the airways to remain open during expiration.<br />

Airflow limitation is measured by spirometry, as this is the<br />

most widely available, reproducible test of lung function.<br />

Many previous definitions of COPD have emphasized the<br />

terms "emphysema" and "chronic bronchitis," which are<br />

no longer included in the definition of COPD used in this<br />

report. Emphysema, or destruction of the gas-exchanging<br />

6 DEFINITION


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 7<br />

surfaces of the lung (alveoli), is a pathological term that<br />

is often (but incorrectly) used clinically and describes<br />

only one of several structural abnormalities present in<br />

patients with COPD. <strong>Chronic</strong> bronchitis, or the presence<br />

of cough and sputum production <strong>for</strong> at least 3 months in<br />

each of two consecutive years, remains a clinically and<br />

epidemiologically useful term. However, it does not<br />

reflect the major impact of airflow limitation on morbidity<br />

and mortality in COPD patients. It is also important to<br />

recognize that cough and sputum production may precede<br />

the development of airflow limitation; conversely, some<br />

patients develop significant airflow limitation without<br />

chronic cough and sputum production.<br />

NATURAL HISTORY<br />

COPD has a variable natural history and not all individuals<br />

follow the same course. However, COPD is generally a<br />

progressive disease, especially if a patient's exposure<br />

to noxious agents continues. If exposure is stopped, the<br />

disease may still progress due to the decline in lung<br />

function that normally occurs with aging. Nevertheless,<br />

stopping exposure to noxious agents, even after<br />

significant airflow limitation is present, can result in<br />

some improvement in function and will certainly slow or<br />

even halt the progression of the disease.<br />

Classification of Severity: Stages of COPD<br />

For educational reasons, a simple classification of disease<br />

severity into four stages is recommended (Figure 1-2).<br />

The staging is based on airflow limitation as measured by<br />

spirometry, which is essential <strong>for</strong> diagnosis and provides<br />

a useful description of the severity of pathological<br />

changes in COPD. Specific FEV 1 cut-points (e.g.,<br />

< 80% predicted) are used <strong>for</strong> purposes of simplicity:<br />

these cut-points have not been clinically validated.<br />

The impact of COPD on an individual patient depends<br />

not just on the degree of airflow limitation, but also on<br />

the severity of symptoms (especially breathlessness and<br />

decreased exercise capacity) and complications of the<br />

disease. The management of COPD is largely symptom<br />

driven, and there is only an imperfect relationship between<br />

the degree of airflow limitation and the presence of<br />

symptoms. The staging, there<strong>for</strong>e, is a pragmatic<br />

approach aimed at practical implementation and should<br />

only be regarded as an educational tool, and a very<br />

Figure 1-2. Classification of Severity of COPD<br />

Stage<br />

Characteristics<br />

0: At Risk • normal spirometry<br />

• chronic symptoms (cough, sputum production)<br />

I: Mild COPD • FEV 1 /FVC < 70%<br />

• FEV 1 ≥ 80% predicted<br />

• with or without chronic symptoms (cough, sputum production)<br />

II: Moderate COPD • FEV 1 /FVC < 70%<br />

• 50% ≤ FEV 1 < 80% predicted<br />

• with or without chronic symptoms (cough, sputum production)<br />

III: Severe COPD • FEV 1 /FVC < 70%<br />

• 30% ≤ FEV 1 < 50% predicted<br />

• with or without chronic symptoms (cough, sputum production)<br />

IV: Very Severe COPD • FEV 1 /FVC < 70%<br />

• FEV 1 < 30% predicted or FEV 1 < 50% predicted plus chronic respiratory<br />

failure<br />

Classification based on postbronchodilator FEV 1<br />

FEV 1 : <strong>for</strong>ced expiratory volume in one second; FVC: <strong>for</strong>ced vital capacity; respiratory failure: arterial partial pressure of oxygen (PaO 2 ) less than<br />

8.0 kPa (60 mm Hg) with or without arterial partial pressure of CO 2 (PaCO 2 ) greater than 6.7 kPa (50 mm Hg) while breathing air at sea level.<br />

DEFINITION 7


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 8<br />

general indication of the approach to management. All<br />

FEV 1 values refer to postbronchodilator FEV 1 .<br />

Although COPD is defined on the basis of airflow<br />

limitation, in practice the decision to seek medical help<br />

(and so permit the diagnosis to be made) is normally<br />

determined by the impact of a particular symptom on a<br />

patient's lifestyle. Thus, COPD may be diagnosed at any<br />

stage of the illness.<br />

The characteristic symptoms of COPD are cough,<br />

sputum production, and dyspnea upon exertion. <strong>Chronic</strong><br />

cough and sputum production often precede the<br />

development of airflow limitation by many years, although<br />

not all individuals with cough and sputum production go<br />

on to develop COPD. This pattern offers a unique<br />

opportunity to identify those at risk <strong>for</strong> COPD and<br />

intervene when the disease is not yet a health problem.<br />

A major objective of <strong>GOLD</strong> is to increase awareness<br />

among health care providers and the general public of<br />

the significance of these symptoms.<br />

Stage 0: At Risk - Characterized by chronic cough and<br />

sputum production. <strong>Lung</strong> function, as measured by<br />

spirometry, is still normal.<br />

Stage I: Mild COPD - Characterized by mild airflow<br />

limitation (FEV 1 /FVC < 70% but FEV 1 > 80% predicted)<br />

and usually, but not always, by chronic cough and sputum<br />

production. At this stage, the individual may not even be<br />

aware that his or her lung function is abnormal. This<br />

underscores the importance of health care providers<br />

doing spirometry in all smokers so that their lung function<br />

can be observed and recorded over time.<br />

Stage II: Moderate COPD - Characterized by worsening<br />

airflow limitation (50% < FEV 1 < 80% predicted), and<br />

usually the progression of symptoms with shortness of<br />

breath typically developing on exertion. This is the stage<br />

at which patients typically seek medical attention<br />

because of dyspnea or an exacerbation of their disease.<br />

may also lead to effects on the heart such as cor<br />

pulmonale (right heart failure). Clinical signs of cor<br />

pulmonale include elevation of the jugular venous pressure<br />

and pitting ankle edema. Patients may have Stage IV:<br />

Very Severe COPD even if the FEV 1 is > 30% predicted,<br />

whenever these complications are present. At this stage,<br />

quality of life is very appreciably impaired and exacerbations<br />

may be life threatening.<br />

Variable Course of COPD<br />

The common statement that only 15-20% of smokers<br />

develop clinically significant COPD is misleading. A<br />

much higher proportion develop abnormal lung function<br />

at some point if they continue to smoke. Not all individuals<br />

with COPD follow the classical linear course as outlined<br />

in the Fletcher and Peto diagram, which is actually the<br />

mean of many individual courses 3 .<br />

Figure 1-3 shows four examples of the various courses<br />

that individual COPD patients may follow. Panel A<br />

illustrates an individual who has cough and sputum<br />

production, but never develops abnormal lung function<br />

(as defined in this Report). Panel B illustrates an<br />

individual who develops abnormal lung function but who<br />

may never come to diagnosis. Panel C illustrates a<br />

person who develops abnormal lung function around<br />

age 50, then progressively deteriorates over about 15<br />

years and dies of respiratory failure at age 65. Panel D<br />

illustrates an individual who develops abnormal lung<br />

function in mid-adult life and continues to deteriorate<br />

gradually but never develops respiratory failure and does<br />

not die as a result of COPD.<br />

Figure 1-3. Examples of Individual Patient Histories<br />

Age<br />

Age<br />

Stage III: Severe COPD - Characterized by further<br />

worsening of airflow limitation (30% ≤ FEV 1 < 50% predicted),<br />

increased shortness of breath, and repeated<br />

exacerbations which have an impact on patients’ quality<br />

of life.<br />

Stage IV: Very Severe COPD - Characterized by severe<br />

airflow limitation (FEV 1 < 30% predicted) or the presence<br />

of chronic respiratory failure. Respiratory failure is<br />

defined as an arterial partial pressure of O 2 (PaO 2 ) less<br />

than 8.0 kPa (60 mmHg) with or without arterial partial<br />

pressure of CO 2 (PaCO 2 ) greater than 6.7 kPa (50 mm<br />

Hg) while breathing air at sea level. Respiratory failure<br />

A<br />

Age<br />

C<br />

B<br />

Age<br />

D<br />

8 DEFINITION


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 9<br />

SCOPE OF THE REPORT<br />

The focus of this Report is primarily on COPD caused by<br />

inhaled particles and gases, the most common of which<br />

worldwide is tobacco smoke. Poorly reversible airflow<br />

limitation associated with bronchiectasis, cystic fibrosis,<br />

tuberculosis, or asthma is not included except insofar as<br />

these conditions overlap with COPD.<br />

Asthma and COPD<br />

COPD can coexist with asthma, the other major chronic<br />

obstructive airway disease characterized by an underlying<br />

airway inflammation. Asthma and COPD have their major<br />

symptoms in common, but these are generally more<br />

variable in asthma than in COPD. The underlying chronic<br />

airway inflammation is also very different (Figure 1-4):<br />

that in asthma is mainly eosinophilic and driven by CD4 +<br />

T lymphocytes, while that in COPD is neutrophilic and<br />

characterized by the presence of increased numbers of<br />

macrophages and CD8 + T lymphocytes. In addition,<br />

airflow limitation in asthma is often completely reversible,<br />

either spontaneously or with treatment, while in COPD it is<br />

never fully reversible and is usually progressive if exposure<br />

to noxious agents continues. Finally, the responses to<br />

treatment of asthma and COPD are dramatically different,<br />

in terms of both the overall magnitude of the achievable<br />

response and the qualitative effects of specific treatments<br />

such as anticholinergics and glucocorticosteroids.<br />

However, there is undoubtedly an overlap between<br />

asthma and COPD. Individuals with asthma who are<br />

exposed to noxious agents that cause COPD may develop<br />

a mixture of "asthma-like" inflammation and "COPD-like"<br />

inflammation. There is also evidence that longstanding<br />

asthma on its own can lead to airway remodeling and<br />

partly irreversible airflow limitation. Asthma can usually<br />

be distinguished from COPD, but until the causal mechanisms<br />

and pathognomonic markers of these diseases are<br />

Figure 1-4. Asthma and COPD<br />

Asthma<br />

Sensitizing agent<br />

Asthmatic airway inflammation<br />

CD4 + T lymphocytes<br />

Eosinophis<br />

Completely<br />

reversible<br />

Airflow Limitation<br />

COPD<br />

Noxious agent<br />

COPD airway inflammation<br />

CD8 + T lymphocytes<br />

Macrophages Neutrophils<br />

Completely<br />

irreversible<br />

better understood it will remain difficult to differentiate the<br />

two diseases in some individual patients. Given the current<br />

state of medical and scientific knowledge, an attempt to<br />

determine an absolutely rigid definition of COPD or asthma<br />

is bound to end up in semantics.<br />

Pulmonary Tuberculosis and COPD<br />

In many developing countries both pulmonary tuberculosis<br />

and COPD are common. In countries where tuberculosis<br />

is very common, respiratory abnormalities may be too<br />

readily attributed to this disease. Conversely, where the<br />

rate of tuberculosis is greatly diminished, the possible<br />

diagnosis of this disease is sometimes overlooked.<br />

<strong>Chronic</strong> bronchitis/bronchiolitis and emphysema often<br />

occur as complications of pulmonary tuberculosis and<br />

are important contributors to the mixed lung function<br />

changes characteristic of tuberculosis 4 . The degree of<br />

obstructive airway changes 5 in treated patients with<br />

pulmonary tuberculosis increases with age, the amount<br />

of cigarettes smoked, and the extent of the initial<br />

tuberculous disease 6 . In patients with both diseases,<br />

COPD adds to the disability of pulmonary tuberculosis,<br />

and vice versa.<br />

There<strong>for</strong>e, in all subjects with symptoms of COPD, a<br />

possible diagnosis of tuberculosis should be considered,<br />

especially in areas where this disease is known to be<br />

prevalent. Investigations to exclude tuberculosis should<br />

be a routine part of COPD diagnosis, the intensity of the<br />

diagnostic procedures depending on the degree of<br />

suspicion. Chest radiograph and sputum culture are<br />

helpful in making the differential diagnosis.<br />

REFERENCES<br />

1. Samet JM. Definitions and methodology in COPD<br />

research. In: Hensley M, Saunders N, eds. Clinical epidemiology<br />

of chronic obstructive pulmonary disease. New<br />

York: Marcel Dekker; 1989. p. 1-22.<br />

2. Vermeire PA, Pride NB. A "splitting" look at chronic nonspecific<br />

lung disease (CNSLD): common features but<br />

diverse pathogenesis. Eur Respir J 1991; 4:490-6.<br />

3. Fletcher C, Peto R. The natural history of chronic airflow<br />

obstruction. BMJ 1977; 1:1645-8.<br />

4. Leitch AG. Pulmonary tuberculosis: clinical features. In:<br />

Crofton J, Douglas A, eds. Respiratory diseases. Ox<strong>for</strong>d:<br />

Blackwell Science; 2000. p. 507-27.<br />

5. Birath G, Caro J, Malmberg R, Simonsson BG. Airway<br />

obstruction in pulmonary tuberculosis. Scand J Resp Dis<br />

1966; 47:27-36.<br />

6. Snider GL, Doctor L, Demas TA, Shaw AR. <strong>Obstructive</strong> airway<br />

disease in patients with treated pulmonary tuberculosis.<br />

Am Rev Respir Dis 1971; 103:625-40.<br />

DEFINITION 9


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 10<br />

10 DEFINITION


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 11<br />

CHAPTER<br />

2<br />

BURDEN OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 12<br />

CHAPTER 2: BURDEN OF COPD<br />

KEY POINTS:<br />

• COPD prevalence and morbidity data that are<br />

available probably greatly underestimate the total<br />

burden of the disease because it is not usually<br />

recognized and diagnosed until it is clinically<br />

apparent and moderately advanced.<br />

• Prevalence, morbidity, and mortality vary<br />

appreciably across countries, but in all countries<br />

where data are available COPD is a significant<br />

health problem in both men and women.<br />

• The substantial increase in the global burden<br />

of COPD projected over the next twenty years<br />

reflects, in large part, the increasing use of<br />

tobacco worldwide, and the changing age<br />

structure of populations in developing countries.<br />

• Medical expenditures <strong>for</strong> treating COPD and<br />

the indirect costs of morbidity can represent a<br />

substantial economic and social burden <strong>for</strong><br />

societies and public and private payers<br />

worldwide. Nevertheless, very little economic<br />

in<strong>for</strong>mation concerning COPD is available.<br />

INTRODUCTION<br />

COPD is a leading cause of morbidity and mortality<br />

worldwide and results in an economic and social burden<br />

that is both substantial and increasing. COPD prevalence,<br />

morbidity, and mortality vary appreciably across countries<br />

and across different groups within countries, but in general<br />

are directly related to the prevalence of tobacco smoking.<br />

Most epidemiological studies have found that COPD<br />

prevalence, morbidity, and mortality have increased over<br />

time and are greater in men than in women. Very few<br />

studies have quantified the economic and social burden<br />

of COPD. In developed countries, the direct medical<br />

costs of COPD are substantial because the disease is<br />

both chronic and highly prevalent. In developing<br />

countries, the indirect cost of COPD from loss of work<br />

and productivity may be more important than the direct<br />

costs of medical care.<br />

EPIDEMIOLOGY<br />

Most of the in<strong>for</strong>mation available on COPD prevalence,<br />

morbidity, and mortality comes from developed countries.<br />

Even in these countries, accurate epidemiological data<br />

on COPD are difficult and expensive to collect.<br />

Prevalence and morbidity data greatly underestimate the<br />

total burden of COPD because the disease is usually not<br />

diagnosed until it is clinically apparent and moderately<br />

advanced. The imprecise and variable definitions of<br />

COPD have made it hard to quantify the morbidity and<br />

mortality of this disease in developed 1 and developing<br />

countries. Mortality data also underestimate COPD as a<br />

cause of death because the disease is more likely to be<br />

cited as a contributory than as an underlying cause of<br />

death, or may not be cited at all 2 .<br />

Prevalence<br />

Available estimates of COPD prevalence have been<br />

developed by determining either the proportion of the<br />

population that reports having respiratory symptoms<br />

and/or airflow limitation, or the proportion that reports<br />

having been diagnosed with COPD, chronic bronchitis, or<br />

emphysema by a physician. Each of these approaches<br />

will yield a different estimate, and may be useful <strong>for</strong><br />

different purposes. For example, studies that ask about<br />

the full range of COPD symptoms from early to advanced<br />

disease are useful to estimate the total societal burden of<br />

the disease. Data on doctor diagnoses of COPD are<br />

useful to estimate the prevalence of clinically significant<br />

disease that is of sufficient severity to require health<br />

services, and there<strong>for</strong>e is likely to incur significant costs.<br />

The population surveys necessary to develop accurate<br />

estimates of COPD prevalence are costly to do and<br />

there<strong>for</strong>e have not been conducted in many countries.<br />

Obtaining reliable prevalence data <strong>for</strong> COPD in each<br />

country should be a priority in order to alert those<br />

responsible <strong>for</strong> planning prevention services and health<br />

care delivery to the high prevalence and cost of the<br />

disease. The prevalence of COPD is likely to vary<br />

appreciably depending on the prevalence of risk<br />

factor exposure, age distribution, and prevalence of<br />

susceptibility genes in different countries.<br />

Until recently, virtually all population-based studies<br />

in developed countries showed a markedly greater<br />

prevalence and mortality of COPD among men<br />

12 BURDEN OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 13<br />

compared to women 3-6 . Gender-related differences in<br />

exposure to risk factors, mostly cigarette smoking,<br />

probably explain this pattern. In developing countries,<br />

some studies report a slightly higher prevalence of COPD<br />

in women than men. This likely reflects exposure to<br />

indoor air pollution from cooking and heating fuels<br />

(greater among women) as well as exposure to tobacco<br />

smoke (greater among men) 7-15 . Recent large populationbased<br />

studies in the US show a different pattern emerging,<br />

with the prevalence of COPD almost equal in men and<br />

women 16,17 . This likely reflects the changing pattern of<br />

exposure to the most important risk factor, tobacco<br />

smoke.<br />

Estimates based on self-report of respiratory symptoms.<br />

COPD prevalence data based on self-report of respiratory<br />

symptoms (chronic cough, sputum production, wheezing,<br />

and shortness of breath) include people at risk <strong>for</strong> COPD<br />

(Stage 0) as well as those with airflow limitation, and thus<br />

yield maximum prevalence estimates. These studies<br />

reveal sizable variations in the prevalence of respiratory<br />

symptoms depending on smoking status, age,<br />

occupational and environmental exposures, country or<br />

region, and, to a lesser extent, gender and race. The<br />

data also reveal appreciable variations over time, reflecting<br />

important temporal changes in populations' exposure to<br />

risk factors such as smoking, outdoor air pollution, and<br />

occupational exposures.<br />

The third National Health and Nutrition Examination<br />

Survey (NHANES 3) 16 , a large national survey conducted<br />

in the US between 1988 and 1994, included self-report<br />

questions about respiratory symptoms. The prevalence<br />

of respiratory symptoms varied markedly by smoking<br />

status (current>ex>never). Among white males, chronic<br />

cough was reported by 24% of smokers, 4.7% of exsmokers,<br />

and 4.0% of never smokers. The prevalence<br />

of chronic cough among white women was 20.6% in<br />

smokers, 6.5% in ex-smokers, and 5.0% in never smokers.<br />

There was a smaller gradient in the prevalence of chronic<br />

cough by race (white>black). The prevalence of sputum<br />

production was similar to that of chronic cough in these<br />

groups.<br />

Estimates based on the presence of airflow limitation.<br />

People may have respiratory symptoms such as cough<br />

and sputum production <strong>for</strong> many years be<strong>for</strong>e developing<br />

airflow limitation. Thus, COPD prevalence data based on<br />

the presence of airflow limitation provide a more accurate<br />

estimate of the burden of COPD that is, or probably soon<br />

will be, clinically significant. However, the use of different<br />

cut points to define airflow limitation makes comparing<br />

the results of different studies difficult.<br />

In the NHANES 3 study 16 , airflow limitation was defined<br />

as an FEV 1 /FVC < 70%. The prevalence of airflow<br />

limitation was lower than the prevalence of respiratory<br />

symptoms found in the same study, but both sets of data<br />

rein<strong>for</strong>ce the view that smoking is the most important<br />

determinant of COPD prevalence in developed countries.<br />

Among white males, airflow limitation was present in<br />

14.2% of current smokers, 6.9% of ex-smokers, and 3.3%<br />

of never smokers. Among white females, the prevalence<br />

of airflow limitation was 13.6% in smokers, 6.8% in exsmokers,<br />

and 3.1% in never smokers. Airflow limitation<br />

was more common among white smokers than among<br />

black smokers.<br />

Estimates based on physician diagnosis of COPD.<br />

COPD prevalence data based on physician diagnosis<br />

provide in<strong>for</strong>mation about the prevalence of clinically<br />

significant COPD that is of sufficient severity to prompt a<br />

visit to a physician. Few population-based prevalence<br />

surveys have been published to provide this in<strong>for</strong>mation,<br />

and available data are often confusing because asthma<br />

and COPD diagnoses are not separated, all age groups<br />

are considered together, or chronic bronchitis and<br />

emphysema are considered separately.<br />

In the UK the General Practice Research Database 18 ,<br />

which is based on 525 practices serving 3.4 million<br />

patients (6.4% of the total population of England and<br />

Wales), provides population-based data on physiciandiagnosed<br />

COPD (Figure 2-1). In 1997, the prevalence<br />

of COPD was 1.7% among men and 1.4% among<br />

women. Between 1990 and 1997, the prevalence<br />

increased by 25% in men and 69% in women. The<br />

prevalence of COPD among men plateaued in the mid-<br />

1990s, but continued to increase among women, reaching<br />

Figure 2-1. Prevalence (%) of Physician-Diagnosed<br />

COPD in the UK From 1990 to 1997 by Sex 18<br />

2.0<br />

1.5<br />

1.0<br />

0.5<br />

0.0<br />

1990<br />

1991<br />

1992<br />

Men<br />

1993<br />

1994<br />

1995<br />

1996<br />

Women<br />

1997<br />

Reprinted with permission from Soriano JR, Maier WC, Egger R, Visick G, Thakrar B, Sykes J,<br />

et al. Thorax 2000; 55:789-94. Copyright 2000 BMJ Publishing Group.<br />

BURDEN OF COPD 13


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 14<br />

in 1997 the level observed in men in 1990. The General<br />

Practice Research Database includes all ages and thus<br />

underestimates the true impact of COPD on older adults.<br />

The <strong>Global</strong> Burden of <strong>Disease</strong> Study. The WHO/World<br />

Bank <strong>Global</strong> Burden of <strong>Disease</strong> Study 19,20 used data from<br />

both published and unpublished studies to estimate the<br />

prevalence of various diseases in different countries and<br />

regions around the world (Figure 2-2). Where few data<br />

<strong>for</strong> a region were available, experts made in<strong>for</strong>med<br />

estimates. Where no in<strong>for</strong>mation was available, preliminary<br />

estimates were derived from data from other regions that<br />

were believed to have similar epidemiological patterns.<br />

Using this approach, the worldwide prevalence of COPD<br />

in 1990 was estimated at 9.34/1,000 in men and<br />

7.33/1,000 in women. However, these estimates include<br />

all ages and underestimate the true prevalence of COPD<br />

in older adults.<br />

Figure 2-2. COPD Around the World (All Ages) 19,20<br />

Region or Country 1990 Prevalence per 1,000<br />

Males/Females<br />

Established Market Economies 6.98/3.79<br />

Formerly Socialist Economies of Europe 7.35/3.45<br />

India 4.38/3.44<br />

China* 26.20/23.70<br />

Other Asia and Islands 2.89/1.79<br />

Sub-Saharan Africa 4.41/2.49<br />

Latin America and Caribbean 3.36/2.72<br />

Middle Eastern Crescent 2.69/2.83<br />

World 9.34/7.33<br />

*The prevalence of COPD in China reported in this study has been<br />

questioned based on recent publications from China 21 - see text.<br />

Given the striking dearth of population-based data on<br />

COPD prevalence in many countries of the world, the<br />

values listed in Figure 2-2 should not be viewed as very<br />

precise. Nevertheless, some general patterns emerge.<br />

The prevalence of COPD is highest in countries where<br />

cigarette smoking has been, or still is, very common,<br />

while the prevalence is lowest in countries where smoking<br />

is less common, or total tobacco consumption per capita<br />

is still low. The lowest COPD prevalence among men<br />

(2.69/1,000) was found in the Middle Eastern Crescent<br />

(a group of 36 countries in North Africa and the Middle<br />

East) and the lowest prevalence among women<br />

(1.79/1,000) was found in the region referred to as "Other<br />

Asia and Islands" (a group of 49 countries and islands,<br />

the largest of which is Indonesia and which includes<br />

Papua New Guinea, Nepal, Vietnam, Korea, Hong Kong,<br />

and many small island countries). Except in the Middle<br />

Eastern Crescent, the prevalence of COPD is higher<br />

among men than among women.<br />

The <strong>Global</strong> Burden of <strong>Disease</strong> study reported a<br />

significantly higher prevalence of COPD in China than in<br />

most of the other regions (26.20/1,000 among men and<br />

23.70/1,000 among women). A more recent survey<br />

conducted in three regions of China (Northern: Beijing;<br />

Northeast: Liao-Ning; and South-Mid: HuBei) in persons<br />

older than 15 years estimated the prevalence of COPD at<br />

4.21/1,000 among men and 1.84/1,000 among women 21 .<br />

Morbidity<br />

Morbidity includes physician visits, emergency<br />

department visits, and hospitalizations. COPD databases<br />

<strong>for</strong> these outcome parameters are less readily<br />

available and usually less reliable than mortality databases.<br />

The limited data available indicate that morbidity<br />

due to COPD increases with age and is greater in men<br />

than women 17,22,23 .<br />

In the UK, general practice consultations <strong>for</strong> COPD<br />

during one year ranged from 4.17/1,000 in 45- to<br />

64-year-olds to 8.86/1,000 in 65- to 74-year-olds to<br />

10.32/1,000 in 75- to 84-year-olds. These rates are<br />

2 to 4 times the equivalent rates <strong>for</strong> chest pain due to<br />

ischemic heart disease 24 .<br />

In 1994, according to statistics from the UK Office of<br />

National Statistics 25 , there were 203,193 hospital<br />

admissions in Northern Ireland, Scotland, Wales, and<br />

England <strong>for</strong> COPD; the average length of hospital stay<br />

among those admitted <strong>for</strong> a COPD diagnosis was<br />

9.9 days.<br />

US data indicate that in 1997 there were 16.365 million<br />

(60.6/1,000) ambulatory care visits <strong>for</strong> COPD and<br />

448,000 (1.66/1,000) hospitalizations <strong>for</strong> which COPD<br />

was the first-listed discharge diagnosis 23 . Hospitalization<br />

rates <strong>for</strong> COPD increased with age and were higher<br />

among men than among women. These data should<br />

be interpreted cautiously, however, because the ICD-9<br />

codes <strong>for</strong> COPD that were in use in 1997, 490-492 and<br />

494-496, include "bronchitis not specified as acute or<br />

chronic." There<strong>for</strong>e, the data <strong>for</strong> ambulatory care visits<br />

are likely to have been inflated by inclusion of visits <strong>for</strong><br />

acute bronchitis 16 .<br />

Mortality<br />

Of all of the descriptive epidemiological data <strong>for</strong> COPD,<br />

mortality data are the most readily available, and<br />

probably the most reliable. (The World Health<br />

Organization publishes mortality statistics <strong>for</strong> selected<br />

causes of death annually <strong>for</strong> all WHO regions 26 ;<br />

14 BURDEN OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 15<br />

additional in<strong>for</strong>mation is available from the WHO<br />

Evidence <strong>for</strong> Health Policy Department 27 .) However,<br />

inconsistent use of terminology <strong>for</strong> COPD causes<br />

problems that do not arise <strong>for</strong> many other diseases.<br />

For example, prior to about 1968 and the Eighth<br />

Revision of the ICD, the terms "chronic bronchitis" and<br />

"emphysema" were used extensively. During the<br />

1970s, the term "COPD" increasingly replaced those<br />

terms in the US and some but not all other countries,<br />

making comparisons of COPD mortality in different<br />

countries very difficult. However, the situation has<br />

improved with the Ninth and Tenth Revisions of the<br />

ICD, in which deaths from COPD or chronic airways<br />

obstruction are included in the broad category of<br />

"COPD and allied conditions" (ICD-9 codes 490-496<br />

and ICD-10 codes J42-46).<br />

The age-adjusted death rates <strong>for</strong> COPD by race and<br />

sex in the US from 1960 to 1996 by ICD code are<br />

shown in Figure 2-3 17 . COPD death rates are very<br />

low among people under age 45 in the US, but then<br />

increase with age, and COPD becomes the fourth or<br />

fifth leading cause of death among those over 45 17 , a<br />

pattern that reflects the cumulative effect of cigarette<br />

smoking 28 . Although appreciable variations in mortality<br />

across developed countries <strong>for</strong> both genders have been<br />

reported 29 , these differences should be interpreted<br />

cautiously. Differences between countries in death<br />

certification, diagnostic practices, the structure of<br />

health care systems, and life expectancy have an<br />

appreciable impact on reported mortality rates.<br />

Figure 2-3. Age-Adjusted* Death Rates <strong>for</strong><br />

<strong>Chronic</strong> <strong>Obstructive</strong> Pulmonary <strong>Disease</strong> by<br />

Race and Sex, US 1960-1996 17<br />

ECONOMIC AND SOCIAL<br />

BURDEN OF COPD<br />

Because COPD is highly prevalent and can be severely<br />

disabling, direct medical expenditures and the indirect<br />

costs of morbidity and premature mortality from COPD<br />

can represent a substantial economic and social burden<br />

<strong>for</strong> societies and public and private insurance payers<br />

worldwide. Nevertheless, very little quantitative in<strong>for</strong>mation<br />

concerning the economic and social burden of COPD<br />

is available in the literature today.<br />

Economic Burden<br />

Cost of illness studies provide insight into the economic<br />

impact of a disease. Some countries attempt to separate<br />

economic burden into disease-attributable direct and indirect<br />

costs. The direct cost is the value of health care<br />

resources devoted to diagnosis and medical management<br />

of the disease. Indirect costs reflect the monetary<br />

consequences of disability, missed work and school,<br />

premature mortality, and caregiver or family costs resulting<br />

from the illness. Data on these topics from developing<br />

countries are not available, but data from the US and<br />

some European countries provide an understanding of<br />

the economic burden of COPD in developed countries.<br />

United States. Figure 2-4 compares the estimated costs<br />

of various lung disorders in the US in 1993. In 1993, the<br />

annual economic burden of COPD in the US was estimated<br />

at $23.9 billion 17 , including $14.7 billion in direct expenditures<br />

<strong>for</strong> medical care services, $4.7 billion in indirect morbidity<br />

costs, and $4.5 billion in indirect costs related to premature<br />

mortality. With an estimated 15.7 million cases of COPD<br />

in the US 30 , the estimated direct cost of COPD is $1,522<br />

per COPD patient per year.<br />

Rate/100,000 Population<br />

ICD/7<br />

ICD/8<br />

ICD/9<br />

Figure 2-4. Direct and Indirect Costs of<br />

<strong>Lung</strong> <strong>Disease</strong>s, 1993 (US $ Billions) 17<br />

White Male<br />

Black Male**<br />

Condition<br />

Total<br />

Cost<br />

Direct<br />

Medical<br />

Cost<br />

Mortality-<br />

Related<br />

Indirect<br />

Cost<br />

Morbidity-<br />

Related<br />

Indirect<br />

Cost<br />

Total<br />

Indirect<br />

Cost<br />

White Female<br />

Black Female**<br />

1960 62 64 66 68 70 72 74 76 78 80 82 84 86 88 90 92 94 1996<br />

*Age-adjusted to the 2000 standard **Nonwhite from 1960 to 1967.<br />

COPD 23.9 14.7 4.5 4.7 9.2<br />

Asthma 12.6 9.8 0.9 0.9 2.8<br />

Influenza 14.6 1.4 0.1 13.1 13.2<br />

Pneumonia 7.8 1.7 4.6 1.5 6.1<br />

Tuberculosis 1.1 0.7 - - 0.4<br />

<strong>Lung</strong> Cancer 25.1 5.1 17.1 2.9 20.0<br />

BURDEN OF COPD 15


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 16<br />

In a US study 31 of COPD-related illness costs based on<br />

the 1987 National Medical Expenditure Survey, per capita<br />

expenditures <strong>for</strong> inpatient hospitalizations of COPD<br />

patients ($5,409 per hospitalization) were 2.7 times the<br />

expenditures <strong>for</strong> patients without COPD ($2,001 per<br />

hospitalization). In 1992, under Medicare, the US<br />

government health insurance program <strong>for</strong> individuals<br />

over 65, annual per capita expenditures <strong>for</strong> people with<br />

COPD ($8,482) were nearly 2.5 times higher than annual<br />

expenditures <strong>for</strong> people without COPD ($3,511) 32 .<br />

United Kingdom. In 1996, the direct cost of COPD in<br />

the UK was approximately £846 million (about US $1.393<br />

billion) or £1,154 (about US $1,900) per person per year,<br />

according to data from the National Health Service (NHS)<br />

Executive 33 . Pharmaceutical expenditures <strong>for</strong> COPD and<br />

allied conditions accounted <strong>for</strong> 11.0% of the total expenditures<br />

<strong>for</strong> prescription medications. Only 2% of total<br />

primary care expenditures were <strong>for</strong> COPD-related visits.<br />

In 1996, lost work productivity, disability, and premature<br />

mortality from COPD in the UK accounted <strong>for</strong> an estimated<br />

24 million days of work lost. The indirect cost of the<br />

disease was estimated at £600 million (about US $960<br />

million) <strong>for</strong> attendance and disability living allowance and<br />

£1.5 billion (about US $2.4 billion) to employers <strong>for</strong> work<br />

absence and reduced productivity 24 .<br />

The Netherlands. In 1993, the direct cost of COPD in<br />

the Netherlands was estimated to exceed US $256 million,<br />

or US $813 per patient per year. Assuming constant costs<br />

and treatment patterns, the direct cost is expected to reach<br />

US $410 million per year by 2010. In 1993 inpatient<br />

hospitalizations accounted <strong>for</strong> 57% of the total direct cost<br />

of COPD, and medications accounted <strong>for</strong> an additional<br />

23%. The indirect cost of COPD in the Netherlands was<br />

not available 34 .<br />

Sweden. The direct cost of COPD-related medical care<br />

in Sweden was estimated at 1.085 billion SEK (about US<br />

$179.4 million) in 1991. The estimated indirect cost of<br />

COPD was an additional 1.699 billion SEK (about US<br />

$280.8 million) 35 .<br />

Comparison of different countries. Figure 2-5<br />

provides data on the economic burden of COPD in four<br />

countries with Western styles of medical practice and<br />

social or private insurance structures. The data are<br />

standardized to equivalent year on a per capita basis.<br />

After adjusting to a common base year and population,<br />

the costs of COPD were relatively similar. The remaining<br />

variability in across-country estimates of economic<br />

burden can be partly explained by several factors,<br />

including: disease prevalence and demographics, particularly<br />

smoking patterns; the type and usage patterns of<br />

health care and non-health care services among patients<br />

with COPD; the relative prices of health care services;<br />

employment and wage rates; and the availability of<br />

medical prevention strategies and treatments <strong>for</strong> COPD.<br />

Similar data from developing countries are not available.<br />

Home care. Individuals with COPD frequently receive<br />

professional medical care in their homes. In some<br />

countries, national health insurance plans provide coverage<br />

<strong>for</strong> oxygen therapy, visiting nursing services, rehabilitation,<br />

and even mechanical ventilation in the home, although<br />

coverage <strong>for</strong> specific services varies from country to<br />

country 36 .<br />

Any estimate of direct medical expenditures <strong>for</strong> home<br />

care underrepresents the true cost of home care to society,<br />

because it ignores the economic value of the care provided<br />

to those with COPD by family members. In developing<br />

countries especially, direct medical costs may be less<br />

important than the impact of COPD on workplace and<br />

home productivity. Because the health care sector<br />

might not provide long-term supportive care services <strong>for</strong><br />

severely disabled individuals, COPD may <strong>for</strong>ce two<br />

individuals to leave the workplace - the affected individual<br />

and a family member who must now stay home to care<br />

<strong>for</strong> the disabled relative. Since human capital is often the<br />

most important national asset <strong>for</strong> developing countries,<br />

COPD may represent a serious threat to their economies.<br />

Social Burden<br />

Figure 2-5. Four-Country Comparison<br />

of COPD Direct and Indirect Costs<br />

Country (ref) Year Direct Cost<br />

(US$ Millions)<br />

Indirect Cost<br />

(US$ Millions)<br />

Total<br />

(US$ Millions)<br />

Per Capita*<br />

(US$)<br />

UK 33 1996 778 3,312 4,090 65<br />

The Netherlands 34 1993 256 N/A N/A N/A #<br />

Sweden 35 1991 179 281 460 60<br />

US 1 1993 14,700 9,200 23,900 87<br />

* Per capita valuation based on 1993 population estimates from the United<br />

Nations Population Council and expressed in 1993 US dollars.<br />

# The authors did not provide estimates of indirect costs.<br />

Since mortality offers a limited perspective on the human<br />

burden of a disease, it is desirable to find other measures<br />

of disease burden that are consistent and measurable<br />

across nations. The World Bank/WHO <strong>Global</strong> Burden of<br />

<strong>Disease</strong> Study 19 designed a method to estimate the fraction<br />

of mortality and disability attributable to major diseases<br />

and injuries using a composite measure of the burden of<br />

each health problem, the Disability-Adjusted Life Year<br />

(DALY). The DALYs <strong>for</strong> a specific condition are the sum<br />

16 BURDEN OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 17<br />

Figure 2-6. Leading Causes of Disability-Adjusted Life Years<br />

(DALYs) Lost Worldwide, 1990 and 2020 (Projected) 19,20<br />

<strong>Disease</strong><br />

or Injury<br />

of years lost because of premature mortality and years<br />

of life lived with disability, adjusted <strong>for</strong> the severity of<br />

disability.<br />

The leading causes of DALYs lost worldwide in 1990<br />

and 2020 (projected) are shown in Figure 2-6. In 1990,<br />

COPD was the twelfth leading cause of DALYs lost in the<br />

world, responsible <strong>for</strong> 2.1% of the total. According to the<br />

projections, COPD will be the fifth leading cause of<br />

DALYs lost worldwide in 2020, behind ischemic heart<br />

disease, major depression, traffic accidents, and cerebrovascular<br />

disease. This substantial increase in the<br />

global burden of COPD projected over the next twenty<br />

years reflects, in large part, the increasing use of tobacco<br />

worldwide and the changing age structure of populations<br />

in developing countries.<br />

REFERENCES<br />

Rank<br />

1990<br />

Percent of<br />

Total<br />

DALYs<br />

Rank<br />

2020<br />

Percent of<br />

Total<br />

DALYs<br />

Lower respiratory infections 1 8.2 6 3.1<br />

Diarrheal diseases 2 7.2 9 2.7<br />

Perinatal period conditions 3 6.7 11 2.5<br />

Unipolar major depression 4 3.7 2 5.7<br />

Ischemic heart disease 5 3.4 1 5.9<br />

Cerebrovascular disease 6 2.8 4 4.4<br />

Tuberculosis 7 2.8 7 3.1<br />

Measles 8 2.6 25 1.1<br />

Road traffic accidents 9 2.5 3 5.1<br />

Congenital anomalies 10 2.4 13 2.2<br />

Malaria 11 2.3 19 1.5<br />

COPD 12 2.1 5 4.1<br />

Trachea, bronchus, lung cancer 33 0.6 15 1.8<br />

Excerpted with permission from Murray CJL, Lopez AD. Science 1999; 274:740-3.<br />

Copyright 1999 American Association <strong>for</strong> the Advancement of Science.<br />

1. Pride NB, Vermeire P, Allegra L. Diagnostic labels applied<br />

to model case histories of chronic airflow obstruction.<br />

Responses to a questionnaire in 11 North American and<br />

Western European countries. Eur Respir J 1989; 2:702-9.<br />

2. Mannino DM, Brown C, Giovino GA. <strong>Obstructive</strong> lung disease<br />

deaths in the United States from 1979 through 1993.<br />

An analysis using multiple-cause mortality data. Am J<br />

Respir Crit Care Med 1997; 156:814-8.<br />

3. Buist AS, Vollmer WM. Smoking and other risk factors. In:<br />

Murray JF, Nadel JA, eds. Textbook of respiratory<br />

medicine. Philadelphia: WB Saunders; 1994. p. 1259-87.<br />

4. Thom TJ. International comparisons in COPD mortality. Am<br />

Rev Respir Dis 1989; 140:S27-34.<br />

5. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking,<br />

changes in smoking habits, and rate of decline in FEV 1 :<br />

new insight into gender differences. Eur Respir J 1994;<br />

7:1056-61.<br />

6. Feinleib M, Rosenberg HM, Collins JG, Delozier JE,<br />

Pokras R, Chevarley FM. Trends in COPD morbidity and<br />

mortality in the United States. Am Rev Respir Dis 1989;<br />

140:S9-18.<br />

7. Chen JC, Mannino MD. Worldwide epidemiology of chronic<br />

obstructive pulmonary disease. Current Opinion in<br />

Pulmonary Medicine 1999; 5:93-9.<br />

8. Dossing M, Khan J, al-Rabiah F. Risk factors <strong>for</strong> chronic<br />

obstructive lung disease in Saudi Arabia. Respiratory Med<br />

1994; 88:519-22.<br />

9. Dennis R, Maldonado D, Norman S, Baena E, Martinez G.<br />

Woodsmoke exposure and risk <strong>for</strong> obstructive airways disease<br />

among women. Chest 1996; 109:115-9.<br />

10. Perez-Padilla R, Regalado U, Vedal S, Pare P, Chapela R,<br />

Sansores R, et al. Exposure to biomass smoke and chronic<br />

airway disease in Mexican women. Am J Respir Crit<br />

Care Med 1996; 154:701-6.<br />

11. Behera D, Jindal SK. Respiratory symptoms in Indian women<br />

using domestic cooking fuels. Chest 1991; 100:385-8.<br />

12. Amoli K. Bronchopulmonary disease in Iranian housewives<br />

chronically exposed to indoor smoke. Eur Respir J 1998;<br />

11:659-63.<br />

13. Pandey MR. Prevalence of chronic bronchitis in a rural<br />

community of the Hill Region of Nepal. Thorax 1984;<br />

39:331-6.<br />

14. Pandey MR. Domestic smoke pollution and chronic bronchitis<br />

in a rural community of the Hill Region of Nepal.<br />

Thorax 1984; 39:337-9.<br />

15. Samet JM, Marbury M, Spengler J. Health effects and<br />

sources of indoor air pollution. Am Rev Respir Dis 1987;<br />

136:1486-508.<br />

16. National Center <strong>for</strong> Health Statistics. Current estimates from<br />

the National Health Interview Survey, United States, 1995.<br />

Washington, DC: Department of Health and Human<br />

Services, Public Health Service, Vital and Health Statistics;<br />

1995. Publication No. 96-1527.<br />

17. National Heart, <strong>Lung</strong>, and Blood Institute. Morbidity & mortality:<br />

chartbook on cardiovascular, lung, and blood diseases.<br />

Bethesda, MD: US Department of Health and Human<br />

Services, Public Health Service, National Institutes of Health;<br />

1998. Available from: URL:<br />

www.nhlbi.nih.gov/nhlbi/seiin/other/cht-book/htm<br />

18. Soriano JR, Maier WC, Egger P, Visick G, Thakrar B, Sykes<br />

J, et al. Recent trends in physician diagnosed COPD in<br />

women and men in the UK. Thorax 2000; 55:789-94.<br />

19. Murray CJL, Lopez AD. Evidence-based health policy -<br />

lessons from the <strong>Global</strong> Burden of <strong>Disease</strong> Study. Science<br />

1996; 274:740-3.<br />

BURDEN OF COPD 17


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 18<br />

20. Murray CJL, Lopez AD, eds. The global burden of disease: a<br />

comprehensive assessment of mortality and disability from<br />

diseases, injuries and risk factors in 1990 and projected to<br />

2020. Cambridge, MA: Harvard University Press; 1996.<br />

21. Xian Sheng Chen. Analysis of basic data of the study on<br />

prevention and treatment of COPD. Chin J Tuber Respiratory<br />

Dis 1998; 21:749-52 (with English abstract).<br />

22. Higgins MW, Thom T. Incidence, prevalence, and mortality:<br />

intra- and inter-country differences. In: Hensley M, Saunders<br />

N, eds. Clinical epidemiology of chronic obstructive pulmonary<br />

disease. New York: Marcel Dekker; 1989. p. 23-43.<br />

23. National Center <strong>for</strong> Health Statistics. National hospital interview<br />

survey. Vital and health statistics, series 10 (issues from<br />

1974 to 1995).<br />

24. Calverley PMA. <strong>Chronic</strong> obstructive pulmonary disease: the<br />

key facts. London: British <strong>Lung</strong> Foundation; 1998.<br />

25. Office of National Statistics. Mortality statistics (revised)<br />

1994, England and Wales. London: Her Majesty’s Stationery<br />

Office; 1996.<br />

26. World Health Organization. World health statistics annual<br />

1995. Geneva: World Health Organization; 1995.<br />

27. World Health Organization, Geneva. Available from: URL:<br />

www.who.int<br />

28. Renzetti AD, McClement JH, Litt BD. The Veterans<br />

Administration Cooperative Study of Pulmonary Function. III:<br />

Mortality in relation to respiratory function in chronic obstructive<br />

pulmonary disease. Am J Med 1966; 41:115-29.<br />

29. Incalzi RA, Fuso L, De Rosa M, Forastiere F, Rapiti E,<br />

Nardecchia B, et al. Co-morbidity contributes to predict mortality<br />

of patients with chronic obstructive pulmonary disease.<br />

Eur Respir J 1997; 10:2794-800.<br />

30. Singh GK, Matthews TJ, Clarke SC. Annual summary of<br />

births, marriages, divorces, and deaths: United States, 1994.<br />

Monthly Vital Statistics Report 14 (13). National Center <strong>for</strong><br />

Health Statistics, Hyattsville, MD.<br />

31. Sullivan SD, Strassels S, Smith DH. Characterization of the<br />

incidence and cost of COPD in the US. Eur Respir J 1996;<br />

9:S421.<br />

32. Grasso ME, Weller WE, Shaffer TJ, Diette GB, Anderson GF.<br />

Capitation, managed care, and chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med 1998; 158:133-8.<br />

33. National Health Service Executive. Burdens of disease: a discussion<br />

document. London: Department of Health; 1996.<br />

34. Rutten-van Molken MP, Postma MJ, Joore MA, Van<br />

Genugten ML, Leidl R, Jager JC. Current and future medical<br />

costs of asthma and chronic obstructive pulmonary disease<br />

in the Netherlands. Respir Med 1999; 93:779-87.<br />

35. Jacobson L, Hertzman P, Lofdahl C-G, Skoogh B-E, Lindgren<br />

B. The economic impact of asthma and COPD in Sweden<br />

1980 and 1991. Respir Med 2000; 94:247-55.<br />

36. Fauroux B, Howard P, Muir JF. Home treatment <strong>for</strong> chronic<br />

respiratory insufficiency: the situation in Europe in 1992. The<br />

European Working Group on Home Treatment <strong>for</strong> <strong>Chronic</strong><br />

Respiratory Insufficiency. Eur Respir J 1994; 7:1721-6.<br />

18 BURDEN OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 19<br />

CHAPTER<br />

3<br />

RISK FACTORS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 20<br />

CHAPTER 3: RISK FACTORS<br />

KEY POINTS:<br />

• Risk factors <strong>for</strong> COPD include both host factors<br />

and environmental exposures, and the disease<br />

usually arises from an interaction between these<br />

two types of factors.<br />

• The host factor that is best documented is a rare<br />

hereditary deficiency of alpha-1 antitrypsin. Other<br />

genes involved in the pathogenesis of COPD<br />

have not yet been identified.<br />

• The major environmental factors are tobacco<br />

smoke, occupational dusts and chemicals<br />

(vapors, irritants, fumes), and indoor/outdoor air<br />

pollution.<br />

INTRODUCTION<br />

The identification of risk factors is an important step<br />

toward developing strategies <strong>for</strong> prevention and treatment<br />

of any disease. Identification of cigarette smoking as an<br />

important risk factor <strong>for</strong> COPD has led to the incorporation<br />

of smoking cessation programs as a key element of<br />

COPD prevention, as well as an important intervention<br />

<strong>for</strong> patients who already have the disease. However,<br />

although smoking is the best-studied COPD risk factor, it<br />

is not the only one. Further studies of other risk factors<br />

could lead to similar powerful interventions.<br />

Much of the evidence concerning risk factors <strong>for</strong> COPD<br />

comes from cross-sectional epidemiological studies that<br />

identify associations rather than cause-and-effect relationships.<br />

Although several longitudinal studies (which<br />

are capable of revealing causal relationships) of COPD<br />

have followed groups and populations <strong>for</strong> up to 20 years,<br />

none of them has monitored the progression of the disease<br />

through its entire course. Thus, current understanding of<br />

risk factors <strong>for</strong> COPD is in many respects incomplete.<br />

Figure 3-1 provides a summary of risk factors <strong>for</strong> COPD.<br />

The division into "Host Factors" and "Exposures" reflects<br />

the current understanding of COPD as resulting from an<br />

interaction between the two types of factors. Thus, of two<br />

people with the same smoking history, only one may<br />

develop COPD due to differences in genetic predisposition<br />

to the disease, or in how long they live. Risk factors <strong>for</strong><br />

COPD may also be related in more complex ways. For<br />

example, gender may influence whether a person takes<br />

up smoking or experiences certain occupational or environmental<br />

exposures; socioeconomic status may be<br />

linked to a child's birth weight; longer life expectancy will<br />

allow greater lifetime exposure to risk factors; etc.<br />

Understanding the relationships and interactions among<br />

risk factors is a crucial area of ongoing investigation.<br />

Host Factors<br />

Exposures<br />

Figure 3-1. Risk Factors <strong>for</strong> COPD<br />

• Genes (e.g., alpha-1<br />

antitrypsin deficiency)<br />

• Airway Hyperresponsiveness<br />

• <strong>Lung</strong> Growth<br />

• Tobacco Smoke<br />

• Occupational Dusts and<br />

Chemicals<br />

• Indoor and Outdoor Air Pollution<br />

• Infections<br />

• Socioeconomic Status<br />

The best-documented host factor is a severe hereditary<br />

deficiency of alpha-1 antitrypsin. The major environmental<br />

factors are tobacco smoke, occupational dusts and<br />

chemicals (vapors, irritants, fumes), and indoor and outdoor<br />

air pollution. However, it is very difficult to demonstrate that<br />

a given risk factor is sufficient to cause the disease.<br />

Data are not available to determine whether the increasing<br />

prevalence of respiratory symptoms and the accelerated<br />

rate of lung function decline that occur with age reflect<br />

the cumulative exposure to respiratory particles, irritants,<br />

fumes, vapors, etc., or host-related phenomena such as<br />

the loss of elastic recoil of lung tissue and stiffening of<br />

the chest wall. The field of normal lung aging has been<br />

only minimally explored and more work is required.<br />

The role of gender as a risk factor <strong>for</strong> COPD remains<br />

unclear. In the past, most studies showed that COPD<br />

prevalence and mortality were greater among men than<br />

women 1-4 . More recent studies 5,6 from developed countries<br />

show that the prevalence of the disease is almost equal<br />

in men and women, which probably reflects changing<br />

patterns of tobacco smoking. Some studies have in fact<br />

suggested that women are more susceptible to the<br />

effects of tobacco smoke than men 4,7 . This is an important<br />

question given the increasing rate of smoking among<br />

women in both developed and developing countries.<br />

The role of nutritional status as an independent risk factor<br />

20 RISK FACTORS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 21<br />

<strong>for</strong> the development of COPD is unclear. Malnutrition and<br />

weight loss can reduce respiratory muscle strength and<br />

endurance, apparently by reducing both respiratory muscle<br />

mass and the strength of the remaining muscle fibers 8 .<br />

The association of starvation and anabolic/catabolic<br />

status with the development of emphysema has been<br />

shown in experimental studies in animals 9 .<br />

HOST FACTORS<br />

Genes<br />

It is believed that many genetic factors increase (or<br />

decrease) a person's risk of developing COPD. Studies<br />

have demonstrated an increased risk of COPD within<br />

families with COPD probands. Some of this risk may be<br />

due to shared environmental factors, but several studies<br />

in diverse populations also suggest a shared genetic risk 10,11 .<br />

The genetic risk factor that is best documented is a<br />

severe hereditary deficiency of alpha-1 antitrypsin 12-14 , a<br />

major circulating inhibitor of serine proteases. This rare<br />

hereditary deficiency is a recessive trait most commonly<br />

seen in individuals of Northern European origin.<br />

Premature and accelerated development of panlobular<br />

emphysema and decline in lung function occur in both<br />

smokers and nonsmokers with the severe deficiency,<br />

although smoking increases the risk appreciably. There<br />

is considerable variation between individuals in the<br />

extent and severity of the emphysema and the rate of<br />

lung function decline. Although alpha-1 antitrypsin<br />

deficiency is relevant to only a small part of the world's<br />

population, it illustrates the interaction between host<br />

factors and environmental exposures leading to COPD.<br />

In this way, it provides a model <strong>for</strong> how other genetic risk<br />

factors are thought to contribute to COPD.<br />

Exploratory studies have revealed a number of candidate<br />

genes that may influence a person's risk of COPD,<br />

including ABO secretor status 15,16 , microsomal epoxide<br />

hydrolase 17 , glutathione S-transferase 18 , alpha-1 antichymotrypsin<br />

19 , the complement component GcG 20 , cytokine<br />

TNF- 21 , and microsatellite instability 22 . However, when<br />

several studies of a given trait are available, the results<br />

are often inconsistent. Several of these genes are<br />

thought to be involved in inflammation, and there<strong>for</strong>e are<br />

related to potential pathogenic mechanisms of COPD.<br />

Airway Hyperresponsiveness<br />

Asthma and airway hyperresponsiveness, identified as<br />

risk factors that contribute to the development of COPD,<br />

are complex disorders related to a number of genetic and<br />

environmental factors. The relationship between asthma/<br />

airway hyperresponsiveness and increased risk of<br />

developing COPD was originally described by Orie and<br />

colleagues 23 and termed the "Dutch hypothesis."<br />

Asthmatics, as a group, experience a slightly accelerated<br />

loss of lung function 24,25 compared to non-asthmatics, as<br />

do smokers with airway hyperresponsiveness compared<br />

to normal smokers 26 . How these trends are related to the<br />

development of COPD is unknown, however. Airway<br />

hyperresponsiveness may also develop after exposure to<br />

tobacco smoke or other environmental insults and thus<br />

may be a result of smoking-related airway disease.<br />

<strong>Lung</strong> Growth<br />

<strong>Lung</strong> growth is related to processes occurring during<br />

gestation, birth weight, and exposures during childhood 27-31 .<br />

Reduced maximal attained lung function (as measured by<br />

spirometry) may identify individuals who are at increased<br />

risk <strong>for</strong> the development of COPD 32 .<br />

EXPOSURES<br />

It may be helpful conceptually to think of a person's<br />

exposures in terms of his or her total burden of inhaled<br />

particles (Figure 3-2). Each type of particle, depending<br />

on its size and composition, may contribute a different<br />

weight to the risk, and the total risk will depend on the<br />

integral of the inhaled exposures. Of the many inhalational<br />

exposures that people may encounter over a lifetime, only<br />

tobacco smoke 2,33-39 and occupational dusts and chemicals<br />

(vapors, irritants, and fumes) 40,41 are known to cause<br />

COPD on their own. Tobacco smoke and occupational<br />

exposures also appear to act additively to increase a<br />

person's risk of developing COPD.<br />

Tobacco Smoke<br />

Cigarette smoking is by far the most important risk factor<br />

<strong>for</strong> COPD and the most important way that tobacco<br />

contributes to the risk of COPD. Cigarette smokers have<br />

a higher prevalence of respiratory symptoms and lung<br />

function abnormalities, a greater annual rate of decline in<br />

FEV 1 , and a greater COPD mortality rate than nonsmokers.<br />

These differences between cigarette smokers and nonsmokers<br />

increase in direct proportion to the quantity of<br />

smoking. Pipe and cigar smokers have greater COPD<br />

morbidity and mortality rates than nonsmokers, although<br />

their rates are lower than those <strong>for</strong> cigarette smokers 33 .<br />

Other types of tobacco smoking popular in various<br />

countries are also risk factors <strong>for</strong> COPD, although their<br />

risk relative to cigarette smoking has not been reported.<br />

Age at starting to smoke, total pack-years smoked, and<br />

current smoking status are predictive of COPD mortality.<br />

RISK FACTORS 21


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 22<br />

Figure 3-2. Total Burden of Inhaled Particles<br />

Figure 3-3. Interaction of Smoking<br />

and Occupational Exposures 41<br />

Cigarette Smoke<br />

Occupational Dusts<br />

& Chemicals<br />

Environmental Tobacco<br />

Smoke (ETS)<br />

Indoor/Outdoor<br />

Air Pollution<br />

Not all smokers develop clinically significant COPD,<br />

which suggests that genetic factors must modify each<br />

individual's risk. Although it is unclear what percentage<br />

of smokers develop the disease, the commonly cited<br />

figure of 15-20% is likely an underestimate because<br />

COPD is both underdiagnosed and underappreciated.<br />

Passive exposure to cigarette smoke (also known as<br />

environmental tobacco smoke or ETS) may also contribute<br />

to respiratory symptoms and COPD by increasing the<br />

lungs' total burden of inhaled particles and gases 2,42,43 .<br />

Smoking during pregnancy may also pose a risk <strong>for</strong> the<br />

fetus, by affecting lung growth and development in utero<br />

and possibly the priming of the immune system 32,44 .<br />

Occupational Dusts and Chemicals<br />

Occupational dusts and chemicals (vapors, irritants,<br />

and fumes) can also cause COPD when the exposures<br />

are sufficiently intense or prolonged, such as those<br />

experienced by miners in many countries. These<br />

exposures can both cause COPD independently of<br />

cigarette smoking and increase the risk in the presence<br />

of concurrent cigarette smoking (Figure 3-3) 41 . Exposure<br />

to coal dust alone in sufficient doses can produce airflow<br />

limitation 45,46 .<br />

Exposure to particulate matter, irritants, organic dusts,<br />

and sensitizing agents can cause an increase in airway<br />

hyperresponsiveness 47 , especially in airways already<br />

damaged by other occupational exposures, cigarette<br />

smoke, or asthma. There is some evidence from<br />

community studies that a combination of dust exposure<br />

Excerpted with permission from Kaufmann F, Drouet D, Lellouch J, Brille D. International<br />

Journal of Epidemiology 1979; 8:201-12. Copyright 1979 Ox<strong>for</strong>d University Press.<br />

and gas or fume exposure may have an additive effect<br />

on the risk of COPD 48-50 .<br />

Indoor and Outdoor Air Pollution<br />

High levels of urban air pollution are harmful to individuals<br />

with existing heart or lung disease. The role of outdoor<br />

air pollution in causing COPD is unclear, but appears to<br />

be small when compared with that of cigarette smoking.<br />

The relative effect of short-term, high peak exposures<br />

and long-term, low-level exposures is a question yet to<br />

be resolved.<br />

Over the past two decades, air pollution in most cities in<br />

developed countries has decreased appreciably. In<br />

contrast, air pollution has increased markedly in many<br />

cities in developing countries. Although it is not clear<br />

which specific elements of ambient air pollution are<br />

harmful, there is some evidence that particles found in<br />

polluted air will add to a person's total inhaled burden.<br />

Indoor air pollution from biomass fuel has been implicated<br />

as a risk factor <strong>for</strong> the development of COPD. This exposure<br />

is greatest in regions where biomass fuel is used <strong>for</strong><br />

cooking and heating in poorly vented dwellings, leading<br />

to high levels of particulate matter in indoor air 51-61 .<br />

Infections<br />

A history of severe childhood infection has been associated<br />

with reduced lung function and increased respiratory<br />

symptoms in adulthood 32 . There are several possible<br />

22 RISK FACTORS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 23<br />

explanations <strong>for</strong> this association (which are not mutually<br />

exclusive). There may be an increased diagnosis of<br />

severe infections in children who have underlying airway<br />

hyperresponsiveness, itself considered a risk factor <strong>for</strong><br />

COPD. Viral infections may be related to another factor,<br />

such as birth weight, that is related to COPD.<br />

HIV infection has been shown to accelerate the onset of<br />

smoking-induced emphysema; HIV-induced pulmonary<br />

inflammation may play a role in this process 62-66 .<br />

Socioeconomic Status<br />

There is evidence that the risk of developing COPD is<br />

inversely related to socioeconomic status 65 . It is not clear,<br />

however, whether this pattern reflects exposures to<br />

indoor and outdoor air pollutants, crowding, poor nutrition,<br />

or other factors that are related to low socioeconomic<br />

status 60,66 .<br />

REFERENCES<br />

1. Feinleib M, Rosenberg HM, Collins JG, Delozier JE,<br />

Pokras R, Chevarley FM. Trends in COPD morbidity and<br />

mortality in the United States. Am Rev Respir Dis 1989;<br />

140:S9-18.<br />

2. Buist AS, Vollmer WM. Smoking and other risk factors. In:<br />

Murray JF, Nadel JA, eds. Textbook of respiratory medicine.<br />

Philadelphia: WB Saunders; 1994. p. 1259-87.<br />

3. Thom TJ. International comparisons in COPD mortality. Am<br />

Rev Respir Dis 1989; 140:S27-34.<br />

4. Xu X, Weiss ST, Rijcken B, Schouten JP. Smoking,<br />

changes in smoking habits, and rate of decline in FEV 1 :<br />

new insight into gender differences. Eur Respir J 1994;<br />

7:1056-61.<br />

5. National Center <strong>for</strong> Health Statistics. Current estimates<br />

from the National Health Interview Survey, United States,<br />

1995. Washington, DC: Department of Health and Human<br />

Services, Public Health Service, Vital and Health Statistics;<br />

1995. Publication No. 96-1527.<br />

6. National Heart, <strong>Lung</strong>, and Blood Institute. Morbidity &<br />

mortality: chartbook on cardiovascular, lung, and blood<br />

diseases. Bethesda, MD: US Department of Health and<br />

Human Services, Public Health Service, National<br />

Institutes of Health; 1998. Available from:<br />

URL: www.nhlbi.nih.gov/nhlbi/seiin/other/cht-book/html<br />

7. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey<br />

WC, Buist AS, et al. Effects of smoking intervention and<br />

the use of an inhaled anticholinergic bronchodilator on the<br />

rate of decline of FEV 1 . The <strong>Lung</strong> Health Study. JAMA<br />

1994; 272:1497-505.<br />

8. Wilson DO, Rogers RM, Wright EC, Anthonisen NR. Body<br />

weight in chronic obstructive pulmonary disease. The<br />

National Institutes of Health Intermittent Positive-Pressure<br />

Breathing Trial. Am Rev Respir Dis 1989; 139:1435-8.<br />

9. Sahebjami H, Vassallo CL. Influence of starvation on<br />

enzyme-induced emphysema. J Appl Physiol 1980; 48:284-8.<br />

10. Silverman EK, Speizer FE. Risk factors <strong>for</strong> the development<br />

of chronic obstructive pulmonary disease. Med Clin<br />

North Am 1996; 80:501-22.<br />

11. Chen Y. Genetics and pulmonary medicine. 10: Genetic<br />

epidemiology of pulmonary function. Thorax 1999; 54:818-24.<br />

12. Laurell CB, Eriksson S. The electrophoretic alpha-1 globulin<br />

pattern of serum in alpha-1 antitrypsin deficiency. Scand<br />

J Clin Lab Invest 1963; 15:132-40.<br />

13. Hubbard RC, Crystal RG. Antiproteases. In: Crystal RB,<br />

West JB, Barnes PJ, Cherniack NS, Weibel ER, eds. The<br />

lung: scientific foundations. New York: Raven Press; 1991.<br />

p. 1775-87.<br />

14. McElvaney NG, Crystal RG. Inherited susceptibility of the<br />

lung to proteolytic injury. In: Crystal RG, West JB, Weibel<br />

ER, Barnes PJ, eds. The lung: scientific foundations.<br />

Philadelphia: Lippincott-Raven; 1997. p. 2537-53.<br />

15. Khoury MJ, Beaty TH, Newill CA, Bryant S, Cohen BH.<br />

Genetic-environmental interactions in chronic airways<br />

obstruction. Int J Epidemiol 1986; 15:65-72.<br />

16. Cohen BH, Bias WB, Chase GA, Diamond EL, Graves CG,<br />

Levy DA, et al. Is ABH nonsecretor status a risk factor <strong>for</strong><br />

obstructive lung disease? Am J Epidemiol 1980; 111:285-91.<br />

17. Smith CA, Harrison DJ. Association between polymorphism<br />

in gene <strong>for</strong> microsomal epoxide hydrolase and susceptibility<br />

to emphysema. Lancet 1997; 350:630-3.<br />

18. Harrison DJ, Cantlay AM, Rae F, Lamb D, Smith CA.<br />

Frequency of glutathione S-transferase M1 deletion in<br />

smokers with emphysema and lung cancer. Hum Exp<br />

Toxicol 1997; 16:356-60.<br />

19. Faber JP, Poller W, Olek K, Baumann U, Carlson J,<br />

Lindmark B, et al. The molecular basis of alpha 1-antichymotrypsin<br />

deficiency in a heterozygote with liver and lung<br />

disease. J Hepatol 1993; 18:313-21.<br />

20. Schellenberg D, Pare PD, Weir TD, Spinelli JJ, Walker BA,<br />

Sand<strong>for</strong>d AJ. Vitamin D binding protein variants and the risk<br />

of COPD. Am J Respir Crit Care Med 1998; 157:957-61.<br />

21. Huang SL, Su CH, Chang SC. Tumor necrosis factor-alpha<br />

gene polymorphism in chronic bronchitis. Am J Respir Crit<br />

Care Med 1997; 156:1436-9.<br />

22. Siafakas NM, Tzortzaki EG, Sourvinos G, Bouros D,<br />

Tzanakis N, Kafatos A, et al. Microsatellite DNA instability in<br />

COPD. Chest 1999; 116:47-51.<br />

23. Orie NGM, Sluiter HJ, De Vreis K, Tammerling K, Wikop J.<br />

The host factor in bronchitis. In: Orie NGM, Sluiter HJ, eds.<br />

Bronchitis, an international symposium. Assen, Netherlands:<br />

Royal Vangorcum; 1961. p. 43-59.<br />

24. Peat JK, Woolcock AJ, Cullen K. Rate of decline of lung<br />

function in subjects with asthma. Eur J Respir Dis 1987;<br />

70:171-9.<br />

25. Lange P, Parner J, Vestbo J, Schnohr P, Jensen G. A 15-<br />

year follow-up study of ventilatory function in adults with<br />

asthma. N Engl J Med 1998; 339:1194-200.<br />

RISK FACTORS 23


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 24<br />

26. Tashkin DP, Altose MD, Connett JE, Kanner RE, Lee WW,<br />

Wise RA. Methacholine reactivity predicts changes in lung<br />

function over time in smokers with early chronic obstructive<br />

pulmonary disease. The <strong>Lung</strong> Health Study Research<br />

Group. Am J Respir Crit Care Med 1996; 153:1802-11.<br />

27. Morgan WJ. Maternal smoking and infant lung function.<br />

Further evidence <strong>for</strong> an in utero effect [editorial; comment].<br />

Am J Respir Crit Care Med 1998; 158:689-90.<br />

28. Hagstrom B, Nyberg P, Nilsson PM. Asthma in adult life - is<br />

there an association with birth weight? Scand J Prim Health<br />

Care 1998; 16:117-20.<br />

29. Svanes C, Omenaas E, Heuch JM, Irgens LM, Gulsvik A.<br />

Birth characteristics and asthma symptoms in young adults:<br />

results from a population-based cohort study in Norway. Eur<br />

Respir J 1998; 12:1366-70.<br />

30. Todisco T, de Benedictis FM, Iannacci L, Baglioni S, Eslami<br />

A, Todisco E, et al. Mild prematurity and respiratory functions.<br />

Eur J Pediatr 1993; 152:55-8.<br />

31. Stein CE, Kumaran K, Fall CH, Shaheen SO, Osmond C,<br />

Barker DJ. Relation of fetal growth to adult lung function in<br />

South India. Thorax 1997; 52:895-9.<br />

32. Tager IB, Segal MR, Speizer FE, Weiss ST. The natural history<br />

of <strong>for</strong>ced expiratory volumes. Effect of cigarette smoking<br />

and respiratory symptoms. Am Rev Respir Dis 1988;<br />

138:837-49.<br />

33. US Surgeon General. The health consequences of smoking:<br />

chronic obstructive pulmonary disease. Washington, DC:<br />

US Department of Health and Human Services; 1984.<br />

Publication No. 84-50205.<br />

34. Higgins MW, Thom T. Incidence, prevalence, and mortality:<br />

intra- and inter-country differences. In: Hensley M, Saunders<br />

N, eds. Clinical epidemiology of chronic obstructive pulmonary<br />

disease. New York: Marcel Dekker; 1989. p. 23-43.<br />

35. Sherrill DL, Lebowitz MD, Burrows B. Epidemiology of<br />

chronic obstructive pulmonary disease. Clin Chest Med<br />

1990; 11:375-87.<br />

36. Auerbach O, Hammond EC, Garfinkel L, Benante C. Relation<br />

of smoking and age to emphysema. Whole-lung section<br />

study. N Engl J Med 1972; 286:853-7.<br />

37. Burrows B, Knudson RJ, Cline MG, Lebowitz MD.<br />

Quantitative relationships between cigarette smoking and<br />

ventilatory function. Am Rev Respir Dis 1977; 115:195-205.<br />

38. Lebowitz MD, Burrows B. Quantitative relationships between<br />

cigarette smoking and chronic productive cough. Int J<br />

Epidemiol 1977; 6:107-13.<br />

39. Higgins MW, Keller JB, Becker M, Howatt W, Landis JR,<br />

Rotman H, et al. An index of risk <strong>for</strong> obstructive airways disease.<br />

Am Rev Respir Dis 1982; 125:144-51.<br />

40. Becklake MR. Occupational exposures: evidence <strong>for</strong> a<br />

causal association with chronic obstructive pulmonary disease.<br />

Am Rev Respir Dis 1989; 140:S85-91.<br />

41. Kauffmann F, Drouet D, Lellouch J, Brille D. Twelve years<br />

spirometric changes among Paris area workers. Int J<br />

Epidemiol 1979; 8:201-12.<br />

42. Leuenberger P, Schwartz J, Ackermann-Liebrich U, Blaser<br />

K, Bolognini G, Bongard JP, et al. Passive smoking exposure<br />

in adults and chronic respiratory symptoms (SAPALDIA<br />

Study). Swiss Study on Air Pollution and <strong>Lung</strong> <strong>Disease</strong>s in<br />

Adults, SAPALDIA Team. Am J Respir Crit Care Med 1994;<br />

150:1222-8.<br />

43. Dayal HH, Khuder S, Sharrar R, Trieff N. Passive smoking in<br />

obstructive respiratory disease in an industrialized urban<br />

population. Environ Res 1994; 65:161-71.<br />

44. Holt PG. Immune and inflammatory function in cigarette<br />

smokers. Thorax 1987; 42:241-9.<br />

45. US Centers <strong>for</strong> <strong>Disease</strong> Control and Prevention. Criteria<br />

<strong>for</strong> a recommended standard: occupational exposure to<br />

respirable coal mine dust. Morgantown, WV: National<br />

Institute of Occupational Safety and Health; 1995.<br />

Publication No. 95-106.<br />

46. Heppleston AG. Prevalence and pathogenesis of pneumoconiosis<br />

in coal workers. Environ Health Perspect 1988;<br />

78:159-70.<br />

47. Niewoehner DE. Anatomic and pathophysiological correlations<br />

in COPD. In: Baum GL, Crapo JD, Celli BR, Karlinsky<br />

JB, eds. Textbook of pulmonary diseases. Philadelphia:<br />

Lippincott-Raven; 1998. p. 823-42.<br />

48. Bakke S, Baste V, Hanoa R, Gulsvik A. Prevalence of<br />

obstructive lung disease in a general population: relation to<br />

occupational title and exposure to some airborne agents.<br />

Thorax 1991; 46:863-70.<br />

49. Humerfelt S, Gulsvik A, Skjaerven R, Nilssen S, Kvale G,<br />

Sulheim O, et al. Decline in FEV 1 and airflow limitation related<br />

to occupational exposures in men of an urban community.<br />

Eur Respir J 1993; 6:1095-103.<br />

50. Humerfelt S, Eide GE, Gulsvik A. Association of years of<br />

occupational quartz exposure with spirometric airflow limitation<br />

in Norwegian men aged 30-46 years. Thorax 1998;<br />

53:649-55.<br />

51. Chen JC, Mannino MD. Worldwide epidemiology of chronic<br />

obstructive pulmonary disease. Current Opinion in<br />

Pulmonary Medicine 1999; 5:93-9.<br />

52. Perez-Padilla R, Regalado U, Vedal S, Pare P, Chapela R,<br />

Sansores R, et al. Exposure to biomass smoke and chronic<br />

airway disease in Mexican women. Am J Respir Crit Care<br />

Med 1996; 154:701-6.<br />

53. Dossing M, Khan J, al-Rabiah F. Risk factors <strong>for</strong> chronic<br />

obstructive lung disease in Saudi Arabia. Respiratory Med<br />

1994; 88:519-22.<br />

54. Behera D, Jindal SK. Respiratory symptoms in Indian<br />

women using domestic cooking fuels. Chest 1991; 100:385-8.<br />

55. Amoli K. Bronchopulmonary disease in Iranian housewives<br />

chronically exposed to indoor smoke. Eur Respir J 1998;<br />

11:659-63.<br />

56. Dennis R, Maldonado D, Norman S, Baena E, Martinez G.<br />

Woodsmoke exposure and risk <strong>for</strong> obstructive airways disease<br />

among women. Chest 1996; 109:115-9.<br />

57. Pandey MR. Prevalence of chronic bronchitis in a rural community<br />

of the Hill Region of Nepal. Thorax 1984; 39:331-6.<br />

24 RISK FACTORS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 25<br />

58. Pandey MR. Domestic smoke pollution and chronic bronchitis<br />

in a rural community of the Hill Region of Nepal. Thorax<br />

1984; 39:337-9.<br />

59. Samet JM, Marbury M, Spengler J. Health effects and<br />

sources of indoor air pollution. Am Rev Respir Dis 1987;<br />

136:1486-508.<br />

60. Tao X, Hong CJ, Yu S, Chen B, Zhu H, Yang M. Priority<br />

among air pollution factors <strong>for</strong> preventing chronic obstructive<br />

pulmonary disease in Shanghai. Sci Total Environ 1992;<br />

127:57-67.<br />

61. Smith KR. National burden of disease in India from indoor<br />

air pollution. Proc Natl Acad Sci USA 2000; 97:13286-93.<br />

62. Diaz PT, Clanton TL, Pacht ER. Emphysema-like pulmonary<br />

disease associated with human immunodeficiency virus<br />

infection. Ann Intern Med 1992; 116:124-8.<br />

63. Diaz PT, King MA, Pacht ER, Wewers MD, Gadek JE,<br />

Nagaraja HN, et al. Increased susceptibility to pulmonary<br />

emphysema among HIV-seropositive smokers. Ann Intern<br />

Med 2000; 132:369-72.<br />

64. Diaz PT, King MA, Pacht ER, Wewers MD, Gadek JE, Neal<br />

D, et al. The pathophysiology of pulmonary diffusion impairment<br />

in human immunodeficiency virus infection. Am J<br />

Respir Crit Care Med 1999; 160:272-7.<br />

65. Prescott E, Lange P, Vestbo J. Socioeconomic status, lung<br />

function and admission to hospital <strong>for</strong> COPD: results from<br />

the Copenhagen City Heart Study. Eur Respir J 1999;<br />

13:1109-14.<br />

66. Strachan DP. Epidemiology: a British perspective. In:<br />

Calverley PMA, Pride NB, eds. <strong>Chronic</strong> obstructive pulmonary<br />

disease. London: Chapman and Hall; 1995. p. 47-67.<br />

RISK FACTORS 25


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 26<br />

26 RISK FACTORS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 27<br />

CHAPTER<br />

4<br />

PATHOGENESIS,<br />

PATHOLOGY, AND<br />

PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 28<br />

CHAPTER 4: PATHOGENESIS, PATHOLOGY,<br />

AND PATHOPHYSIOLOGY<br />

KEY POINTS:<br />

• Exposure to inhaled noxious particles and gases<br />

causes inflammation of the lungs that can lead to<br />

COPD if the normal protective and/or repair<br />

mechanisms are overwhelmed or defective.<br />

• Exacerbations of COPD are associated with an<br />

increase in airway inflammation.<br />

• Although inflammation is important in both<br />

diseases, the inflammatory response in COPD is<br />

markedly different from that in asthma.<br />

• In addition to inflammation, two other processes<br />

thought to be important in the pathogenesis of<br />

COPD are an imbalance of proteinases and<br />

antiproteinases in the lung, and oxidative stress.<br />

• Pathological changes characteristic of COPD are<br />

found in the central airways, peripheral airways,<br />

lung parenchyma, and pulmonary vasculature.<br />

• The peripheral airways become the major site of<br />

airways obstruction in COPD. The structural<br />

changes in the airway wall are the most important<br />

cause of the increase in peripheral airways<br />

resistance in COPD. Inflammatory changes such<br />

as airway edema and mucus hypersecretion also<br />

contribute to airway narrowing.<br />

• Most common in COPD patients is the<br />

centrilobular <strong>for</strong>m of emphysema, which involves<br />

dilatation and destruction of the respiratory<br />

bronchioles.<br />

• Physiological changes characteristic of the<br />

disease include mucus hypersecretion, ciliary<br />

dysfunction, airflow limitation, pulmonary hyperinflation,<br />

gas exchange abnormalities, pulmonary<br />

hypertension, and cor pulmonale, and they<br />

usually develop in this order over the course of<br />

the disease.<br />

• The irreversible component of airflow limitation is<br />

primarily due to remodeling of the small airways.<br />

Parenchymal destruction (emphysema) also<br />

contributes but plays a smaller role.<br />

• In advanced COPD, peripheral airways obstruction,<br />

parenchymal destruction, and pulmonary vascular<br />

abnormalities reduce the lung's capacity <strong>for</strong> gas<br />

exchange, producing hypoxemia and, later on,<br />

hypercapnia. Inequality in the ventilation/perfusion<br />

ratio (V A /Q) is the major mechanism behind<br />

hypoxemia in COPD.<br />

• Pulmonary hypertension develops late in the<br />

course of COPD. It is the major cardiovascular<br />

complication of COPD and is associated with a<br />

poor prognosis.<br />

• COPD is associated with systemic inflammation<br />

and skeletal muscle dysfunction that may<br />

contribute to limitation of exercise capacity and<br />

decline of health status.<br />

INTRODUCTION<br />

Inhaled noxious particles and gases that lead to COPD<br />

cause lung inflammation, induce tissue destruction,<br />

impair the defense mechanisms that serve to limit the<br />

destruction, and disrupt the repair mechanisms that may<br />

be able to restore tissue structure in the face of some<br />

injuries. The results of lung tissue damage are mucus<br />

hypersecretion, airway narrowing and fibrosis, destruction<br />

of the parenchyma (emphysema), and vascular changes.<br />

In turn, these pathological changes lead to airflow limitation<br />

and the other physiological abnormalities characteristic of<br />

COPD.<br />

Much of the in<strong>for</strong>mation concerning the pathogenesis<br />

of COPD comes from studies in experimental animals or<br />

in vitro systems. These experimental systems are limited<br />

as they differ from human disease in a number of<br />

respects. Studies in human subjects of the pathogenesis,<br />

pathology, and pathophysiology of COPD are often limited<br />

by patient selection, small numbers of subjects, and limited<br />

access to the relevant tissue. There<strong>for</strong>e, an evidencebased<br />

perspective on these topics is in many respects<br />

incomplete.<br />

PATHOGENESIS<br />

COPD is characterized by chronic inflammation throughout<br />

the airways, parenchyma, and pulmonary vasculature.<br />

The intensity and cellular and molecular characteristics of<br />

the inflammation vary as the disease progresses. Over<br />

28 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 29<br />

time, inflammation damages the lungs and leads to the<br />

pathologic changes characteristic of COPD.<br />

In addition to inflammation, two other processes thought<br />

to be important in the pathogenesis of COPD are an<br />

imbalance of proteinases and antiproteinases in the lung,<br />

and oxidative stress. These processes may themselves<br />

be consequences of inflammation, or they may arise from<br />

environmental (e.g., oxidant compounds in cigarette<br />

smoke) or genetic (e.g., alpha-1 antitrypsin deficiency)<br />

factors. Figure 4-1 details the interactions between these<br />

mechanisms. The multiplicity of cells and mediators<br />

thought to be involved in the pathogenesis of COPD is<br />

presented schematically in Figure 4-2.<br />

Inflammatory Cells<br />

COPD is characterized by an increase in neutrophils,<br />

macrophages, and T lymphocytes (especially CD8 + ) in<br />

various parts of the lung (Figure 4-3). There may also be<br />

an increase in eosinophils in some patients, particularly<br />

during exacerbations. These increases are brought about<br />

by increases in inflammatory cell recruitment, survival,<br />

and/or activation. Many studies reveal a correlation<br />

between the number of inflammatory cells of various<br />

types in the lung and the severity of COPD 1-10 .<br />

Figure 4-3. Sites of Inflammatory<br />

Cell Increases in COPD<br />

Anti-oxidants<br />

Figure 4-1. Pathogenesis of COPD<br />

Oxidative stress<br />

Noxious particles<br />

and gases<br />

<strong>Lung</strong> Inflammation<br />

Antiproteinases<br />

Proteinases<br />

Large Airways<br />

Small Airways<br />

Parenchyma<br />

• Macrophages<br />

• T lymphocytes (especially CD8 + )<br />

• Neutrophils (severe disease only)<br />

• Eosinophils (in some patients)<br />

• Macrophages<br />

• T lymphocytes (especially CD8 + )<br />

• Eosinophils (in some patients)<br />

• Macrophages<br />

• T lymphocytes (especially CD8 + )<br />

• Neutrophils<br />

COPD pathology<br />

Repair mechanisms<br />

Pulmonary Arteries • T lymphocytes (especially CD8 + )<br />

• Neutrophils<br />

Figure 4-2. Cells and Mediators Involved<br />

in the Pathogenesis of COPD<br />

CELLS<br />

Macrophages<br />

Neutrophils<br />

CD8 + lymphocytes<br />

Eosinophils<br />

Epithelial cells<br />

Fibroblasts<br />

Printed with permission of Dr. Peter J. Barnes.<br />

MEDIATORS<br />

LTB4<br />

IL-8, GRO-1<br />

MCP-1, MIP-1<br />

GM-CSF<br />

4<br />

Endothelin<br />

Substance P<br />

-1<br />

PROTEINASES<br />

Neutrophil elastase<br />

Cathepsins<br />

Proteinase-3<br />

MMPs<br />

EFFECTS<br />

Mucus hypersecretion<br />

Fibrosis<br />

Alveolar wall<br />

destruction<br />

Neutrophils. Increased numbers of activated neutrophils<br />

are found in sputum and bronchoalveolar lavage (BAL)<br />

fluid of patients with COPD 4,5,8,9 , although the role of<br />

neutrophils in COPD is not yet clear. Neutrophils are<br />

also increased in smokers without COPD 11 . However,<br />

neutrophils are little increased in airway and parenchyma<br />

tissue sections, which may reflect their rapid transit<br />

through these parts of the lung. Induced sputum studies<br />

also show an increase in myeloperoxidase (MPO)<br />

and human neutrophil lipocalin, indicating neutrophil<br />

activation 12 . Exacerbations of COPD are characterized by<br />

a marked increase in the number of neutrophils in BAL<br />

fluid 13 . Neutrophils secrete several proteinases, including<br />

neutrophil elastase (NE), neutrophil cathepsin G, and<br />

neutrophil proteinase-3, which may contribute to<br />

parenchymal destruction and chronic mucus hypersecretion.<br />

Macrophages. Increased numbers of macrophages are<br />

present in the large and small airways and lung<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 29


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 30<br />

parenchyma of patients with COPD, as reflected in<br />

histopathology, BAL, bronchial biopsy, and induced<br />

sputum studies 2,4-6,9 . In patients with emphysema,<br />

macrophages are localized to sites of alveolar wall<br />

destruction 1 . Macrophages likely play an orchestrating<br />

role in COPD inflammation by releasing mediators<br />

such as tumor necrosis factor- (TNF-), interleukin<br />

8 (IL-8), and leukotriene B4 (LTB4), which promote<br />

neutrophilic inflammation.<br />

T lymphocytes. Histopathology and bronchial biopsy<br />

studies show an increase in T lymphocytes, especially<br />

CD8 + (cytotoxic) cells, throughout the lungs of patients<br />

with COPD 1,2,10,14 . Their role in COPD inflammation is<br />

not yet fully understood, but one way that CD8 + cells<br />

may contribute to COPD is by releasing per<strong>for</strong>in,<br />

granzyme-B, and TNF-, which can cause the cytolysis<br />

and apoptosis of alveolar epithelial cells 15 that may be<br />

responsible <strong>for</strong> the persistence of inflammation. An<br />

increased number of lymphocyte-like natural killer (NK)<br />

cells has also been reported in patients with severe<br />

COPD 3 .<br />

Eosinophils. The presence and role of eosinophils<br />

in COPD are uncertain. Some bronchial biopsy<br />

studies show eosinophils increased in the airways of<br />

some patients with stable COPD 6,16 . However, some<br />

of these patients may have had coexisting asthma, as<br />

other studies report no increase in eosinophils in<br />

COPD patients 2 . The levels of eosinophil cationic<br />

protein (ECP) and eosinophil peroxidase (EPO) in<br />

induced sputum are elevated in COPD, suggesting<br />

that eosinophils may be present but degranulated, and<br />

there<strong>for</strong>e no longer recognizable by light microscopy 12 .<br />

The high levels of neutrophil elastase (NE) often found<br />

in COPD may be responsible <strong>for</strong> this degranulation 17 .<br />

Most studies agree that airway eosinophils are<br />

increased during exacerbations of COPD 18,19 .<br />

Epithelial cells. Airway and alveolar epithelial cells<br />

are likely to be important sources of inflammatory<br />

mediators in COPD, though their role in inflammation<br />

in this disease has not yet been thoroughly studied.<br />

Exposure of nasal or bronchial epithelial cells from<br />

healthy volunteers to nitrogen dioxide (NO 2 ), ozone<br />

(O 3 ), and diesel exhaust particles results in significant<br />

synthesis and release of proinflammatory mediators,<br />

including eicosanoids, cytokines, and adhesion<br />

molecules 20 . The adhesion molecule E-selectin,<br />

involved in recruitment and adhesion of neutrophils, is<br />

up-regulated on airway epithelial cells in COPD patients 21 .<br />

Cultured human bronchial epithelial cells from COPD<br />

patients release lower levels of inflammatory mediators<br />

such as TNF- and IL-8 than similar preparations from<br />

nonsmokers or smokers without COPD, suggesting<br />

that some <strong>for</strong>m of down-regulation of inflammatory<br />

mediator release may occur in epithelial cells of individuals<br />

with COPD 20 .<br />

Inflammatory Mediators<br />

Activated inflammatory cells in COPD release a variety<br />

of mediators, including a spectrum of potent proteinases 22,23 ,<br />

oxidants 24 , and toxic peptides 25 . Many of the mediators<br />

thought to be important in the disease – notably LTB4 26 ,<br />

IL-8 4,27 , and TNF- 4,16 – are capable of damaging lung<br />

structures and/or sustaining neutrophilic inflammation.<br />

The damage induced by these moieties may further<br />

potentiate inflammation by releasing chemotactic<br />

peptides from the extracellular matrix 28 . Little is yet<br />

known about the specific role of these inflammatory<br />

mediators in COPD. Studies of the therapeutic use of<br />

selective mediator antagonists should identify the<br />

molecules relevant in COPD.<br />

Leukotriene B4 (LTB4). LTB4, a potent chemoattractant<br />

of neutrophils, is found at increased levels in the sputum<br />

of patients with COPD 26 . It is probably derived from<br />

alveolar macrophages, which secrete more LTB4 in<br />

patients with COPD. Several potent LTB4 receptor<br />

antagonists have been developed <strong>for</strong> clinical studies<br />

and should elucidate further the role of this mediator<br />

in COPD. So far there is no evidence that cysteinyl<br />

leukotrienes (LTC4, LTD4, LTE4) are involved in<br />

COPD. Selective antagonists of the cysteinyl<br />

leukotriene 1 receptor (CysLT 1 ) have proven helpful in<br />

patients with asthma and studies of these drugs in<br />

COPD patients are now underway. The role of the<br />

cysteinyl leukotriene 2 receptor (CysLT 2 ) in respiratory<br />

disease is as yet unknown 29 .<br />

Interleukin 8 (IL-8). IL-8, a selective chemoattractant<br />

of neutrophils that may be secreted by macrophages,<br />

neutrophils, and airway epithelial cells, is present at<br />

high concentrations in induced sputum and BAL<br />

fluid of patients with COPD 4,27 . IL-8 may play a<br />

primary role in the activation of both neutrophils and<br />

eosinophils in the airways of COPD patients and may<br />

serve as a marker in evaluating the severity of airway<br />

inflammation 27 .<br />

Tumor necrosis factor- (TNF-). TNF- activates<br />

the transcription factor nuclear factor-B (NF-B),<br />

which in turn activates the IL-8 gene in epithelial cells<br />

and macrophages (Figure 4-4). TNF- is present at<br />

high concentrations in sputum 4 and is detectable in<br />

bronchial biopsies 16 in patients with COPD. TNF-<br />

serum levels and production by peripheral blood<br />

monocytes are increased in weight-losing COPD<br />

patients, suggesting that this mediator may play a role in<br />

the cachexia of severe COPD 30 .<br />

30 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 31<br />

Figure 4-4. Interaction Between<br />

Macrophages, Neutrophils, and Epithelial Cells<br />

• Trans<strong>for</strong>ming growth factor-ß (TGF-ß) and epidermal<br />

growth factor (EGF) show increased expression in<br />

epithelial cells and submucosal cells (eosinophils and<br />

fibroblasts) in COPD patients 33 . These mediators<br />

may play a role in airway remodeling (fibrosis and<br />

narrowing) in COPD 34 .<br />

• Endothelin-1 (ET-1), a potent endothelium-derived<br />

vasoconstrictor peptide, is found at increased<br />

concentrations in induced sputum of patients with<br />

COPD 35 . Patients with severe COPD also have<br />

elevated plasma levels of ET-1, which is probably<br />

related to their chronic hypoxemia 36 .<br />

Cigarette smoke activates macrophages and epithelial<br />

cells to produce tumor necrosis factor- (TNF-),<br />

switching on the gene <strong>for</strong> interleukin-8 (IL-8), which<br />

recruits and activates neutrophils. This process occurs<br />

via the activation of the transcription factor nuclear factor-B<br />

(NF-B).<br />

Printed with permission of Dr. Peter J. Barnes.<br />

Others. Other inflammatory mediators that may be<br />

involved in COPD include the following:<br />

• Macrophage chemotactic protein-1 (MCP-1), a<br />

potent chemoattractant of monocytes, is increased<br />

in the BAL fluid of patients with COPD and smokers<br />

without COPD, but not in ex-smokers or nonsmokers 31 .<br />

Thus, MCP-1 may be involved in macrophage<br />

recruitment into the lungs in smokers.<br />

• Macrophage inflammatory protein-1ß (MIP-1ß) is<br />

increased in the BAL fluid of patients with COPD<br />

compared to smokers, ex-smokers, and nonsmokers 31 .<br />

Macrophage inflammatory protein-1 (MIP-1)<br />

shows increased expression in airway epithelial cells<br />

from COPD patients 3 compared to control smokers.<br />

• Granulocyte-macrophage colony stimulating factor<br />

(GM-CSF) is found at increased concentrations in<br />

the BAL fluid of patients with stable COPD and at<br />

markedly elevated levels during exacerbations 13 . The<br />

number of GM-CSF-immunoreactive macrophages is<br />

also increased in sputum of patients with COPD 32 .<br />

GM-CSF is important <strong>for</strong> neutrophil survival and may<br />

play a role in enhancing neutrophilic inflammation.<br />

• Neuropeptides, such as substance P, calcitonin<br />

gene-related peptide, and vasoactive intestinal<br />

peptide (VIP), have potent effects on vascular function<br />

and mucus secretion. An increased concentration<br />

of substance P is found in sputum of patients with<br />

chronic bronchitis 37 . One bronchial biopsy study<br />

showed an increase in VIP-immunoreactive nerves<br />

in the vicinity of submucosal glands in patients with<br />

chronic bronchitis, suggesting that this substance<br />

may play a role in mucus hypersecretion 38 . However,<br />

another study showed no significant differences in<br />

the number of nerves immunoreactive <strong>for</strong> substance<br />

P, calcitonin gene-related peptide, or VIP between<br />

COPD patients and healthy subjects 39 .<br />

• Complement. Activation of the complement pathway<br />

via generation of the potent chemotaxin C5a may<br />

play a significant role in the neutrophil accumulation<br />

seen in the lungs of patients with COPD 40 .<br />

Differences Between Inflammation<br />

in COPD and Asthma<br />

Although inflammation is important in both diseases,<br />

the inflammatory response in COPD is markedly different<br />

from that in asthma, as summarized in Figure 4-5.<br />

However, some patients with COPD also have asthma,<br />

and the inflammation in their lungs may show characteristics<br />

of both diseases.<br />

Since inflammation is a feature of COPD, it follows that<br />

anti-inflammatory therapies may have clinical benefit in<br />

controlling symptoms, preventing exacerbations, and<br />

slowing the progression of the disease. However, the<br />

inflammatory response in COPD appears to be poorly<br />

responsive to the glucocorticosteroids that are effective<br />

anti-inflammatory medications in asthma.<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 31


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 32<br />

Figure 4-5. Characteristics of Inflammation in COPD and Asthma<br />

COPD<br />

Asthma<br />

Cells • Neutrophils • Eosinophils<br />

• Large increase in macrophages • Small increase in macrophages<br />

• Increase in CD8 + T lymphocytes • Increase in CD4 + Th2 lymphocytes<br />

• Activation of mast cells<br />

Mediators • LTB4 • LTD4<br />

• IL-8<br />

• IL-4, IL-5<br />

• TNF-<br />

• (Plus many others)<br />

Consequences • Squamous metaplasia of epithelium • Fragile epithelium<br />

• Parenchymal destruction<br />

• Thickening of basement membrane<br />

• Mucus metaplasia<br />

• Mucus metaplasia<br />

• Glandular enlargement<br />

• Glandular enlargement<br />

Response to • Glucocorticosteroids have little or • Glucocorticosteroids inhibit<br />

Treatment no effect inflammation<br />

Inflammation and COPD Risk Factors<br />

The connection between cigarette smoke and inflammation<br />

has been most extensively studied 41-52 . Cigarette smoke<br />

activates macrophages and epithelial cells to produce<br />

TNF- and may also cause macrophages to release<br />

other inflammatory mediators, including IL-8 and LTB4 53,54 .<br />

Inflammation is present in the lungs of smokers without a<br />

diagnosis of COPD. This inflammation is similar to, but<br />

less intense than, the inflammation in the lungs of patients<br />

with COPD. For example, induced sputum studies show<br />

that smokers without COPD have a greater proportion of<br />

neutrophils in their lungs than age-matched nonsmokers,<br />

but a smaller proportion than COPD patients 4,9 . Thus, the<br />

inflammation characteristic of COPD is thought to represent<br />

an exaggeration of a normal, protective response to<br />

inhalational exposures.<br />

However, not all smokers develop COPD, and why the<br />

normal, protective inflammatory response becomes an<br />

exaggerated, harmful one in some smokers is poorly<br />

understood. Presumably the inflammation caused by<br />

cigarette smoking interacts with other host or environmental<br />

factors to produce the excess decline in lung function<br />

that results in COPD 55 . Inflammatory changes are also<br />

present in bronchial biopsies in ex-smokers, suggesting<br />

that the inflammatory response in COPD may persist even<br />

in the absence of continuous exposure to risk factors 56 .<br />

A number of studies have demonstrated that a variety of<br />

particulates (e.g., diesel exhaust, grain dust) can initiate<br />

respiratory tract inflammation 57-61 . It is likely that indoor air<br />

pollution derived from the burning of biomass fuels will<br />

prove to have similar effects.<br />

Proteinase-Antiproteinase Imbalance<br />

Laurell and Eriksson observed in 1963 that individuals<br />

with a hereditary deficiency of the serum protein alpha-1<br />

antitrypsin, which inhibits a number of serine proteinases<br />

such as neutrophil elastase, are at increased risk of<br />

developing emphysema 62 . Elastin, the target of neutrophil<br />

elastase, is a major component of alveolar walls, and<br />

elastin fragments may perpetuate inflammation by acting<br />

as potent chemotactic agents <strong>for</strong> macrophages and<br />

neutrophils. These observations led to the hypothesis<br />

that an imbalance between proteinases and endogenous<br />

antiproteinases results in lung destruction.<br />

Based on many observations, it now seems clear that an<br />

imbalance of proteinases and antiproteinases may involve<br />

either increased production or activity of proteinases, or<br />

inactivation or reduced production of antiproteinases.<br />

Often, the imbalance is a consequence of the inflammation<br />

induced by inhalational exposures. For example,<br />

macrophages, neutrophils, and airway epithelial cells<br />

release a combination of proteinases. The imbalance<br />

may also be caused by a decrease of antiproteinase<br />

activity by oxidative stress (itself a consequence of<br />

32 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 33<br />

inflammation), cigarette smoke 63,64 , and possibly other<br />

COPD risk factors.<br />

The concept has also been expanded to include additional<br />

proteinases and antiproteinases. While neutrophil elastase<br />

is likely to be the major proteinase involved in lung<br />

destruction in alpha-1 antitrypsin deficiency, it may not<br />

be involved in COPD caused by inhalational exposures.<br />

Additional proteinases that have been implicated in<br />

COPD include neutrophil cathepsin G, neutrophil<br />

proteinase-3, cathepsins released from macrophages<br />

(specifically cathepsins B, L, and S), and various matrix<br />

metalloproteinases (MMPs) 65 . These proteinases are<br />

capable of degrading elastin and also collagen, another<br />

main component of alveolar walls. Some proteinases,<br />

such as neutrophil elastase 66 and neutrophil proteinase-3 67 ,<br />

induce mucus secretion, and neutrophil elastase also<br />

produces mucus gland hyperplasia 68 . Thus, proteinases<br />

may be involved in mucus hypersecretion as well as<br />

parenchymal destruction. Antiproteinases thought to<br />

be involved in COPD include, in addition to alpha-1<br />

anti-trypsin, secretory leukoproteinase inhibitor (SLPI)<br />

and tissue inhibitors of MMPs (TIMPs).<br />

Oxidative Stress<br />

There is increasing evidence that an oxidant/antioxidant<br />

imbalance, in favor of oxidants, occurs in COPD. (The<br />

process is summarized in Figure 4-6.) Markers of<br />

oxidative stress have been found in the epithelial lining<br />

fluid, breath, and urine of cigarette smokers and patients<br />

with COPD. For example, hydrogen peroxide (H 2 O 2 )<br />

and nitric oxide (NO) are direct measures of oxidants<br />

generated by cigarette smoking or released from<br />

inflammatory leukocytes and epithelial cells. H 2 O 2 is<br />

increased in the breath of patients with stable COPD and<br />

during exacerbations 69 , and NO is increased in the breath<br />

during exacerbations of COPD 70 . A prostaglandin isomer,<br />

Figure 4-6. Increased Oxidative Stress in COPD<br />

Antiproteinases<br />

SLPI 1 -AT<br />

Proteolysis<br />

Mucus secretion<br />

ANTIOXIDANTS<br />

Glutathione<br />

Uric acid, bilirubin<br />

Vitamins C, E<br />

O - 2 , H 2 O 2<br />

OH, ONOO<br />

-<br />

Printed with permission of Dr. Peter J. Barnes.<br />

IL-8<br />

NF-B<br />

Neutrophil<br />

recruitment<br />

TNF <br />

Isoprostanes Plasma leak Bronchoconstriction<br />

isoprostane F 2 -III, which is <strong>for</strong>med by free radical<br />

peroxidation of arachidonic acid and believed to be an in<br />

vivo biomarker of lung oxidative stress, is increased in<br />

both breath condensates 71 and urine 72 in COPD patients<br />

compared to healthy controls and is increased even more<br />

during exacerbations.<br />

Oxidative stress contributes to COPD in a variety of<br />

ways. Oxidants can react with, and damage, a variety of<br />

biological molecules, including proteins, lipids, and nucleic<br />

acids, and this can lead to cell dysfunction or death,<br />

as well as damage to the lung extracellular matrix. In<br />

addition to directly damaging the lung, oxidative stress<br />

contributes to the proteinase-antiproteinase imbalance<br />

both by inactivating antiproteinases (such as alpha-1<br />

antirypsin and SLPI) and by activating proteinases (such<br />

as MMPs). Oxidants also promote inflammation, <strong>for</strong><br />

example by activating the transcription factor NF-B,<br />

which orchestrates the expression of multiple inflammatory<br />

genes thought to be important in COPD such as IL-8<br />

and TNF-. Finally, oxidative stress may contribute to<br />

reversible airway narrowing. H 2 O 2 constricts airway<br />

smooth muscle in vitro and isoprostane F 2 -III is a potent<br />

constrictor of human airways 73 .<br />

PATHOLOGY<br />

Pathological changes characteristic of COPD are found in<br />

the central airways, peripheral airways, lung parenchyma,<br />

and pulmonary vasculature 74 . The various lesions are a<br />

result of chronic inflammation in the lung, which in turn is<br />

initiated by the inhalation of noxious particles and gases<br />

such as those present in cigarette smoke. The lung has<br />

natural defense mechanisms and a considerable capacity<br />

to repair itself, but the working of these mechanisms may<br />

be affected by genetic traits (e.g., alpha-1 antitrypsin<br />

deficiency) or exposure to other environmental risk factors<br />

(e.g., infection, atmospheric pollution) 75 , as well as by the<br />

chronic nature of the inflammation and repeated nature of<br />

the injury.<br />

Central Airways<br />

The central airways include the trachea, bronchi, and<br />

bronchioles greater than 2-4 mm in internal diameter.<br />

In patients with chronic bronchitis, an inflammatory<br />

exudate of fluid and cells infiltrates the epithelium lining<br />

the central airways and associated glands and ducts 2,42 .<br />

The predominant cells in this inflammatory exudate are<br />

macrophages and CD8+ T lymphocytes 2,76 . <strong>Chronic</strong><br />

inflammation in the central airways is also associated<br />

with an increase in the number (metaplasia) of epithelial<br />

goblet and squamous cells; dysfunction, damage, and/or<br />

loss of cilia; enlarged submucosal mucus-secreting<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 33


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 34<br />

glands 77 ; an increase in the amount of smooth muscle<br />

and connective tissue in the airway wall 78 ; degeneration<br />

of the airway cartilage 79,80 ; and mucus hypersecretion.<br />

The mechanisms of mucus gland hypertrophy and goblet<br />

cell metaplasia have not yet been identified, but animal<br />

studies 81,82 show that irritants including cigarette smoke 83<br />

can produce these changes. The various pathological<br />

changes in the central airways (Figure 4-7) are responsible<br />

<strong>for</strong> the symptoms of chronic cough and sputum production,<br />

which identify people at risk <strong>for</strong> COPD and may continue<br />

to be present throughout the course of the disease.<br />

Thus, these pathological changes may be present either<br />

on their own or in combination with the changes in the<br />

peripheral airways and lung parenchyma described<br />

below.<br />

Figure 4-8. Pathological Changes<br />

of the Peripheral Airways in COPD<br />

Figure 4-7. Pathological Changes<br />

of the Central Airways in COPD<br />

Histological sections of peripheral airways from<br />

patients who are cigarette smokers. (A) is a<br />

nearly normal airway; (B) shows a plug of mucoid<br />

exudate in the lumen with little or no evidence of<br />

inflammation in the wall; (C) shows the presence of<br />

an inflammatory exudate in the wall and lumen of<br />

the airway; and (D) shows an airway with reduced<br />

lumen, structural reorganization of the airway wall,<br />

increased smooth muscle, and deposition of peribronchial<br />

connective tissue.<br />

Printed with permission of Dr. James C. Hogg and Stuart Greene.<br />

(A) shows a central bronchus from the lung of a cigarette<br />

smoker with normal lung function. Only small<br />

amounts of muscle are present and the epithelial<br />

glands are small. This contrasts sharply with a diseased<br />

bronchus (B), where the muscle appears as a<br />

thick bundle and the glands are enlarged. (C) shows<br />

these enlarged glands at a higher magnification. There<br />

is evidence of a chronic inflammatory process involving<br />

polymorphonuclear (arrowhead) and mononuclear<br />

cells, including plasma cells (arrow).<br />

Printed with permission of Dr. James C. Hogg and Stuart Greene.<br />

Peripheral Airways<br />

The peripheral airways include small bronchi and<br />

bronchioles that have an internal diameter of less than<br />

2 mm (Figure 4-8). The early decline in lung function<br />

in COPD is correlated with inflammatory changes in the<br />

peripheral airways, similar to those that occur in the<br />

central airways: exudate of fluid and cells in the<br />

airway wall and lumen, goblet and squamous cell<br />

metaplasia of the epithelium 43 , edema of the airway<br />

mucosa due to inflammation, and excess mucus in the<br />

airways due to goblet cell metaplasia.<br />

However, the most characteristic change in the peripheral<br />

airways of patients with COPD is airway narrowing.<br />

Inflammation initiated by cigarette smoking 45 and other<br />

risk factors 75 leads to repeated cycles of injury and<br />

repair of the walls of the peripheral airways. Injury is<br />

caused either directly by inhaled toxic particles and<br />

gases such as those found in cigarette smoke, or<br />

indirectly by the action of inflammatory mediators; this<br />

injury then initiates repair processes. Although airway<br />

repair is only partly understood, it seems likely that<br />

disordered repair processes can lead to tissue<br />

remodeling with altered structure and function.<br />

Cigarette smoke may impair lung repair mechanisms,<br />

thereby further contributing to altered lung structure 84-86 .<br />

Even normal lung repair mechanisms can lead to airway<br />

34 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 35<br />

remodeling because tissue repair in the airways, as<br />

elsewhere in the body, may involve scar tissue <strong>for</strong>mation.<br />

In any case, this injury-and-repair process results in a<br />

structural remodeling of the airway wall, with increasing<br />

collagen content and scar tissue <strong>for</strong>mation, that narrows<br />

the lumen and produces fixed airways obstruction 87 .<br />

The peripheral airways become the major site of airways<br />

obstruction in COPD, and direct measurements of<br />

peripheral airways resistance 88 show that the structural<br />

changes in the airway wall are the most important cause<br />

of the increase in peripheral airways resistance in COPD.<br />

Inflammatory changes such as airway edema and<br />

mucus hypersecretion also contribute to airway narrowing<br />

in COPD. So does loss of elastic recoil, but fibrosis of<br />

the small airways plays the largest role.<br />

Fibrosis in the peripheral airways, as elsewhere in the<br />

body, is characterized by the accumulation of mesenchymal<br />

cells (fibroblasts and myofibroblasts) and extracellular<br />

connective tissue matrix. Several cell types including<br />

mononuclear phagocytes and epithelial cells may produce<br />

mediators that drive this process. The mediators that<br />

drive the accumulation of these cells and of the matrix<br />

are incompletely defined, but it is likely that several<br />

mediators including TGF-ß, ET-1, Insulin-like growth<br />

factor-1, fibronectin, platelet-derived growth factor<br />

(PDGF), and others are involved 89 .<br />

Figure 4-9. Normal and Emphysematous <strong>Lung</strong>s<br />

Figure 4-10. Normal Respiratory Bronchioles<br />

and Centrilobular Emphysema<br />

(A) shows a photomicrograph of the pleural surface<br />

of a normal lung, with a secondary lobule defined by<br />

a connective tissue septum (solid arrow) and several<br />

terminal bronchioles (TB) filled with opaque material.<br />

(B) shows a low-power photomicrograph of a normal<br />

terminal bronchiole (TB) branching into a respiratory<br />

bronchiole (RB), which eventually end in alveolar<br />

ducts (AD). (C) is a schematic diagram of centrilobular<br />

emphysema and (D) shows the bronchographic<br />

appearance of this lesion (TB=terminal bronchiole;<br />

CLE=centrilobular emphysema).<br />

Printed with permission of Dr. James C. Hogg and Stuart Greene.<br />

Photomicrographs of paper-mounted whole lung sections<br />

prepared from (A) a normal lung, (B) a lung with<br />

mild centrilobular emphysema, and (C) a lung with<br />

severe panacinar emphysema. Note that the centrilobular<br />

<strong>for</strong>m affects mainly the upper lung regions whereas<br />

the panacinar <strong>for</strong>m is more apparent in the lower<br />

lung regions.<br />

Printed with permission of Dr. James C. Hogg and Stuart Greene.<br />

<strong>Lung</strong> Parenchyma<br />

The lung parenchyma includes the gas exchanging surface<br />

of the lung (respiratory bronchioles and alveoli) and the<br />

pulmonary capillary system (Figure 4-9). The most<br />

common type of parenchymal destruction in COPD<br />

patients is the centrilobular <strong>for</strong>m of emphysema (Figure<br />

4-10), which involves dilatation and destruction of the<br />

respiratory bronchioles 90 . These lesions occur more<br />

frequently in the upper lung regions in milder cases, but<br />

in advanced disease they may appear diffusely throughout<br />

the entire lung and also involve destruction of the pulmonary<br />

capillary bed. Panacinar emphysema, which extends<br />

throughout the acinus, is the characteristic lesion seen in<br />

alpha-1 antitrypsin deficiency and involves dilatation and<br />

destruction of the alveolar ducts and sacs as well as the<br />

respiratory bronchioles. It tends to affect the lower more<br />

than upper lung regions. Because this process usually<br />

affects all of the acini in the secondary lobule, it is also<br />

referred to as panlobular emphysema. The primary<br />

mechanism of lung parenchyma destruction, in both<br />

smoking-related COPD and alpha-1 antitrypsin deficiency,<br />

is thought to be an imbalance of endogenous proteinases<br />

and antiproteinases in the lung. Oxidative stress, another<br />

consequence of inflammation, may also contribute 91 .<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 35


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 36<br />

Figure 4-11. Pathological Changes of the Pulmonary<br />

Vasculature in COPD<br />

exercise and then at rest. As COPD worsens, greater<br />

amounts of smooth muscle, proteoglycans, and collagen 95<br />

further thicken the vessel wall. In advanced disease, the<br />

changes in the muscular arteries may be associated with<br />

emphysematous destruction of the pulmonary capillary bed.<br />

.<br />

PATHOPHYSIOLOGY<br />

Pathological changes in COPD lead to corresponding<br />

physiological abnormalities that usually become<br />

evident first on exercise and later also at rest.<br />

Physiological changes characteristic of the disease<br />

include mucus hypersecretion, ciliary dysfunction, airflow<br />

limitation, pulmonary hyperinflation, gas exchange<br />

abnormalities, pulmonary hypertension, and cor<br />

pulmonale, and they usually develop in this order<br />

over the course of the disease. In turn, various<br />

physiological abnormalities contribute to the<br />

characteristic symptoms of COPD – chronic cough<br />

and sputum production and dyspnea.<br />

Photomicrographs of small (A) and large (B) vessels<br />

in the lung of a heavy smoker with normal lung function,<br />

and small (C) and large (D) vessels in the lung of<br />

a patient with severe emphysema. Note that the smaller<br />

vessel has thicker walls (compare arrows in A and<br />

C) and that the larger vessel has a thicker media<br />

(compare arrows in B and D) in the patient with severe<br />

emphysema. (L=vessel lumen; magnification<br />

bars=100µ).<br />

Printed with permission of Dr. James C. Hogg and Stuart Greene.<br />

Pulmonary Vasculature<br />

Pulmonary vascular changes in COPD (Figure 4-11) are<br />

characterized by a thickening of the vessel wall that<br />

begins early in the natural history of the disease, when<br />

lung function is reasonably well maintained and pulmonary<br />

vascular pressures are normal at rest 92 . Endothelial<br />

dysfunction of the pulmonary arteries, which may be<br />

caused directly by cigarette smoke products 93 or indirectly<br />

by inflammatory mediators 14 , occurs early in COPD 94 .<br />

Since endothelium plays an important role in regulating<br />

vascular tone and cell proliferation, it is likely that<br />

endothelial dysfunction might initiate the sequence of<br />

events that results ultimately in structural changes.<br />

Thickening of the intima is the first structural change 92 ,<br />

followed by an increase in vascular smooth muscle and<br />

the infiltration of the vessel wall by inflammatory cells,<br />

including macrophages and CD8 + T lymphocytes 14 .<br />

These structural changes are correlated with an increase<br />

in pulmonary vascular pressure that develops first with<br />

Mucus Hypersecretion<br />

and Ciliary Dysfunction<br />

Mucus hypersecretion in COPD is caused by the<br />

stimulation of the enlarged mucus secreting glands<br />

and increased number of goblet cells by inflammatory<br />

mediators such as leukotrienes, proteinases, and<br />

neuropeptides. Ciliated epithelial cells undergo<br />

squamous metaplasia leading to impairment in<br />

mucociliary clearance mechanisms. These changes<br />

are usually the first physiological abnormalities to<br />

develop in COPD, and can be present <strong>for</strong> many years<br />

be<strong>for</strong>e any other physiological abnormalities develop.<br />

Airflow Limitation and<br />

Pulmonary Hyperinflation<br />

Expiratory airflow limitation is the hallmark physiological<br />

change of COPD. The airflow limitation characteristic<br />

of COPD is primarily irreversible, with a small reversible<br />

component. Several pathological characteristics<br />

contribute to airflow limitation and changes in pulmonary<br />

mechanics, as summarized in Figure 4-12. The<br />

irreversible component of airflow limitation is primarily<br />

due to remodeling 42,43,87,88,96,97 – fibrosis and narrowing – of<br />

the small airways that produces fixed airways obstruction<br />

and a consequent increase in airways resistance. The<br />

sites of airflow limitation in COPD are the smaller<br />

conducting airways, including bronchi and bronchioles<br />

less than 2 mm in internal diameter. In the normal lung,<br />

resistance of these smaller airways makes up a small<br />

percentage of the total airways resistance 88 . But in<br />

36 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 37<br />

Figure 4-12. Causes of Airflow Limitation in COPD<br />

Irreversible<br />

Reversible<br />

• Fibrosis and narrowing of airways<br />

• Loss of elastic recoil due to alveolar<br />

destruction<br />

• Destruction of alveolar support that<br />

maintains patency of small airways<br />

• Accumulation of inflammatory cells,<br />

mucus, and plasma exudate in bronchi<br />

• Smooth muscle contraction in peripheral<br />

and central airways<br />

• Dynamic hyperinflation during exercise<br />

patients with COPD the total lower airways resistance<br />

approximately doubles, and most of the increase is due<br />

to a large increase in peripheral airways resistance 88 .<br />

Although some have argued that a larger proportion of<br />

the total resistance should be attributed to peripheral airways<br />

in the normal lung, there is wide agreement that the<br />

peripheral airways become the major site of obstruction<br />

in COPD.<br />

Parenchymal destruction (emphysema) plays a smaller<br />

role in this irreversible component but contributes to<br />

expiratory airflow limitation and the increase in airways<br />

resistance in several ways. Destruction of alveolar<br />

attachments inhibits the ability of the small airways to<br />

maintain patency 98 . Alveolar destruction is also associated<br />

with a loss of elastic recoil of the lung 99,100 , which decreases<br />

the intra-alveolar pressure driving exhalation.<br />

Although both the destruction of alveolar attachments to<br />

the outer wall of the peripheral airways and the loss of<br />

lung elastic recoil produced by emphysema have been<br />

implicated in the pathogenesis of peripheral airways<br />

obstruction 98,100 , direct measurements of peripheral airways<br />

resistance 88 show that the structural changes in the<br />

airway wall are the most important cause of the increase<br />

in peripheral airways resistance in COPD.<br />

Airway smooth muscle contraction, ongoing airway<br />

inflammation, and intraluminal accumulation of mucus<br />

and plasma exudate may be responsible <strong>for</strong> the small<br />

part of airflow limitation that is reversible with treatment.<br />

Inflammation and accumulation of mucus and exudate<br />

may be particularly important during exacerbations 101 .<br />

Airflow limitation in COPD is best measured through<br />

spirometry, which is key to the diagnosis and management<br />

of the disease. The essential spirometric measurements<br />

<strong>for</strong> diagnosis and monitoring of COPD patients are the<br />

<strong>for</strong>ced expiratory volume in one second (FEV 1 ) and<br />

<strong>for</strong>ced vital capacity (FVC). As COPD progresses, with<br />

increased thickness of the airway wall, loss of alveolar<br />

attachments, and loss of lung elastic recoil, FEV 1 and<br />

FVC decrease. A decrease in the ratio of FEV 1 to FVC<br />

is often the first sign of developing airflow limitation.<br />

FEV 1 declines naturally with age, but the rate of decline<br />

in COPD patients is generally greater than that in normal<br />

subjects.<br />

With increasing severity of airflow limitation, expiration<br />

becomes flow-limited during tidal breathing. Initially, this<br />

occurs only during exercise, but later it is also seen at<br />

rest. In parallel with this, functional residual capacity (FRC)<br />

increases due to the combination of the decrease in the<br />

elastic properties of the lungs, premature airway closure,<br />

and a variable dynamic element reflecting the breathing<br />

pattern adopted to cope with impaired lung mechanics.<br />

As airflow limitation develops, the rate of lung emptying is<br />

slowed and the interval between inspiratory ef<strong>for</strong>ts does<br />

not allow expiration to the relaxation volume of the<br />

respiratory system; this leads to dynamic pulmonary<br />

hyperinflation. The increase in FRC can impair inspiratory<br />

muscle function and coordination, although the contractility<br />

of the diaphragm, when normalized <strong>for</strong> lung volume,<br />

seems to be preserved. These changes occur as the<br />

disease advances but are almost always seen first during<br />

exercise, when the greater metabolic stimulus to ventilation<br />

stresses the ability of the ventilatory pump to maintain<br />

gas exchange.<br />

Gas Exchange Abnormalities<br />

In advanced COPD, peripheral airways obstruction,<br />

parenchymal destruction, and pulmonary vascular<br />

abnormalities reduce the lung's capacity <strong>for</strong> gas<br />

exchange, producing hypoxemia and, later on, hypercapnia.<br />

The correlation between routine lung function tests and<br />

arterial blood gases is poor, but significant hypoxemia or<br />

hypercapnia is rare when FEV 1 is greater than 1.00 L 102 .<br />

Hypoxemia is initially only present during exercise, but as<br />

the disease continues to progress it is also present at rest.<br />

Inequality in the ventilation/perfusion ratio (V A /Q) is the<br />

major mechanism behind hypoxemia in COPD, regardless<br />

of the stage of the disease 103 . In the peripheral airways,<br />

injury of the airway wall is associated with VA /Q<br />

mismatching, as indicated by a significant correlation<br />

between bronchiolar inflammation and the distribution of<br />

ventilation. In the parenchyma, destruction of the lung<br />

surface area by emphysema reduces diffusing capacity<br />

and interferes with gas exchange 104 . High V A /Q units<br />

probably represent emphysematous regions with alveolar<br />

destruction and loss of pulmonary vasculature. The<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 37


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 38<br />

severity of pulmonary emphysema appears to be related<br />

to the overall inefficiency of the lung as a gas exchanger.<br />

This is reflected by the good correlation between the<br />

diffusing capacity of carbon monoxide per liter of alveolar<br />

volume (D Lco /V A ) and the severity of macroscopic<br />

emphysema. Reduced ventilation due to loss of elastic<br />

recoil in the emphysematous lung, together with the loss<br />

of the capillary bed and the generalized inhomogeneity<br />

of ventilation due to the patchy nature of these changes,<br />

leads to areas of V A /Q mismatching that result in arterial<br />

hypoxemia.<br />

The relationship between pulmonary vascular abnormalities<br />

and V A /Q relationships has been investigated in patients<br />

with mild COPD. The more severe the vessel wall damage<br />

is, the less the reversal of hypoxic vasoconstriction by<br />

oxygen 105 . This suggests that pathology in the pulmonary<br />

artery wall, particularly when it affects the intimal layer,<br />

may play a key role in determining the loss of vascular<br />

response to hypoxia that contributes to V A /Q mismatching.<br />

<strong>Chronic</strong> hypercapnia usually reflects inspiratory muscle<br />

dysfunction and alveolar hypoventilation.<br />

Pulmonary Hypertension and Cor Pulmonale<br />

Pulmonary hypertension develops late in the course of<br />

COPD (Stage IV: Very Severe COPD), usually after the<br />

development of severe hypoxemia (PaO 2 < 8.0 kPa or 60<br />

mm Hg) and often hypercapnia as well. It is the major<br />

cardiovascular complication of COPD and is associated<br />

with the development of cor pulmonale and with a poor<br />

prognosis 106 . However, even in patients with severe<br />

disease, pulmonary arterial pressure is usually only<br />

modestly elevated at rest, though it may rise markedly<br />

with exercise. Pulmonary hypertension in COPD is<br />

believed to progress rather slowly even if left untreated.<br />

Further studies are required to firmly establish the natural<br />

history of pulmonary hypertension in COPD.<br />

Factors that are known to contribute to the development<br />

of pulmonary hypertension in patients with COPD include<br />

vasoconstriction; remodeling of pulmonary arteries,<br />

which thickens the vessel walls and reduces the lumen;<br />

and destruction of the pulmonary capillary bed by<br />

emphysema, which further increases the pressure<br />

required to perfuse the pulmonary vascular bed.<br />

Vasoconstriction may itself have several causes, including<br />

hypoxia, which causes pulmonary vascular smooth muscle<br />

to contract; impaired mechanisms of endothelium-dependent<br />

vasodilation, such as reduced NO synthesis or release;<br />

and abnormal secretion of vasoconstrictor peptides (such<br />

as ET-1, which is produced by inflammatory cells). In<br />

advanced COPD, hypoxia plays the primary role in<br />

producing pulmonary hypertension, both by causing<br />

vasoconstriction of the pulmonary arteries and by<br />

promoting remodeling of the vessel wall (either by inducing<br />

the release of growth factors 107 or as a consequence of<br />

the mechanical stress that results from hypoxic<br />

vasoconstriction).<br />

Pulmonary hypertension is associated with the development<br />

of cor pulmonale, defined as "hypertrophy of the right<br />

ventricle resulting from diseases affecting the function<br />

and/or structure of the lungs, except when these pulmonary<br />

alterations are the result of diseases that primarily affect<br />

the left side of the heart, as in congenital heart disease."<br />

This is a pathological definition and the clinical diagnosis<br />

and assessment of right ventricular hypertrophy is difficult<br />

in life.<br />

The prevalence and natural history of cor pulmonale in<br />

COPD are not yet clear. Pulmonary hypertension and<br />

reduction of the vascular bed due to emphysema can<br />

lead to right ventricular hypertrophy and right heart failure,<br />

but right ventricular function appears to be maintained<br />

in some patients despite the presence of pulmonary<br />

hypertension 108 . Right heart failure is associated with<br />

venous stasis and thrombosis that may result in pulmonary<br />

embolism and further compromise the pulmonary<br />

circulation.<br />

Systemic Effects<br />

COPD is associated with systemic (i.e., extrapulmonary)<br />

effects, such as systemic inflammation and skeletal<br />

muscle dysfunction. Evidence of systemic inflammation<br />

includes the presence of systemic oxidative stress 109 ,<br />

abnormal concentrations of circulating cytokines 110 , and<br />

activation of inflammatory cells 111,112 . Evidence of skeletal<br />

muscle dysfunction includes the progressive loss of<br />

skeletal muscle mass and the presence of several<br />

bioenergetic abnormalities 113 . These systemic effects have<br />

important clinical consequences, as they contribute to the<br />

limitation of patients' exercise capacity and thus the<br />

decline of health status in COPD. The presence of these<br />

systemic effects appears to worsen a patient's prognosis 114 .<br />

Pathophysiology and the<br />

Symptoms of COPD<br />

<strong>Chronic</strong> cough and sputum production, sometimes labeled<br />

as chronic bronchitis, are a result of airway inflammation,<br />

which leads to mucus hypersecretion and dysfunction of<br />

the normal ciliary clearance mechanisms. Sputum is<br />

produced in COPD as a result of the inflammatory<br />

response, and contains plasma proteins exuded from the<br />

microvessels of the bronchial circulation, inflammatory<br />

cells, and small amounts of mucus from epithelial goblet<br />

cells. The volume of sputum produced overpowers clearance<br />

mechanisms, resulting in cough and expectoration.<br />

38 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 39<br />

Some pathological abnormalities, such as inflammation of<br />

the submucosal glands and hyperplasia of goblet cells,<br />

may contribute to chronic sputum production, although<br />

these pathological abnormalities are not present in all<br />

patients with this symptom.<br />

Dyspnea, an abnormal awareness of the act of breathing,<br />

usually reflects an imbalance between the neural drive to<br />

the respiratory muscles and the effectiveness of the<br />

resulting ventilation. Different individuals use different<br />

words to describe the feeling of breathlessness, which is<br />

also influenced by other factors such as mood. In COPD<br />

patients, dyspnea is mainly the result of impaired lung<br />

mechanics (increased airways resistance, decreased<br />

elastic recoil). It is only present on vigorous exercise in<br />

the early stages of disease but may be present at rest as<br />

the mechanical impairment becomes severe.<br />

PATHOLOGY AND<br />

PATHOPHYSIOLOGY OF<br />

EXACERBATIONS<br />

The progressive course of COPD is complicated by<br />

exacerbations that have many causes and occur with<br />

increasing frequency as the disease progresses.<br />

Pathology<br />

Distinguishing the pathology of these acute events from<br />

that of the underlying disease is difficult because patients<br />

experiencing an exacerbation are usually too ill to study.<br />

The limited evidence available suggests that mild COPD<br />

exacerbations are associated with increases of both<br />

neutrophils and eosinophils in sputum and biopsies, while<br />

severe COPD exacerbations are associated with an<br />

increase in sputum neutrophils and eosinophils 18,19 . At<br />

least in sputum, the changes in inflammatory cells during<br />

exacerbations of COPD are the same as those observed<br />

during exacerbations of asthma 115-119 . So far no study has<br />

been conducted examining the pathological abnormalities<br />

associated with fatal exacerbations of COPD, which can be<br />

considered the extreme end of the spectrum of severity.<br />

Pathophysiology<br />

Expiratory airflow is almost unchanged during mild<br />

exacerbations 18 , and only slightly reduced during severe<br />

exacerbations 120,121 . Although the pathophysiology of<br />

exacerbations is not fully understood, the primary<br />

physiological change in severe exacerbations is a further<br />

worsening of gas exchange, primarily produced by<br />

increased V A /Q inequality. As V A /Q relationships worsen,<br />

increased work of the respiratory muscles results in<br />

greater oxygen consumption, decreased mixed venous<br />

oxygen tension, and further amplification of gas<br />

exchange abnormalities 120 . Worsening of V A /Q relationships<br />

has several causes in exacerbations. Airway<br />

inflammation and edema, mucus hypersecretion, and<br />

bronchoconstriction may contribute to changes in the<br />

distribution of ventilation, while hypoxic constriction of<br />

pulmonary arterioles may modify the distribution of<br />

perfusion. Additional contributors to worsening gas<br />

exchange in exacerbations include abnormal patterns of<br />

breathing and fatigue of the respiratory muscles. These<br />

can cause further deterioration in blood gases and<br />

worsening of respiratory acidosis, leading to severe<br />

respiratory failure and death 120-123 . Alveolar hypoventilation<br />

also contributes to hypoxemia, hypercapnia, and<br />

respiratory acidosis. In turn, hypoxemia and respiratory<br />

acidosis promote pulmonary vasoconstriction, which<br />

increases pulmonary artery pressures and imposes an<br />

added load on the right ventricle.<br />

REFERENCES<br />

1. Finkelstein R, Fraser RS, Ghezzo H, Cosio MG. Alveolar<br />

inflammation and its relation to emphysema in smokers.<br />

Am J Respir Crit Care Med 1995; 152:1666-72.<br />

2. O'Shaughnessy TC, Ansari TW, Barnes NC, Jeffery PK.<br />

Inflammation in bronchial biopsies of subjects with chronic<br />

bronchitis: inverse relationship of CD8 + T lymphocytes with<br />

FEV 1 . Am J Respir Crit Care Med 1997; 155:852-7.<br />

3. Di Stefano A, Capelli A, Lusuardi M, Balbo P, Vecchio C,<br />

Maestrelli P, et al. Severity of airflow limitation is associated<br />

with severity of airway inflammation in smokers. Am J<br />

Respir Crit Care Med 1998; 158:1277-85.<br />

4. Keatings VM, Collins PD, Scott DM, Barnes PJ. Differences<br />

in interleukin-8 and tumor necrosis factor-alpha in induced<br />

sputum from patients with chronic obstructive pulmonary<br />

disease or asthma. Am J Respir Crit Care Med 1996;<br />

153:530-4.<br />

5. Thompson AB, Daughton D, Robbins RA, Ghafouri MA,<br />

Oehlerking M, Rennard SI. Intraluminal airway inflammation<br />

in chronic bronchitis. Characterization and correlation<br />

with clinical parameters. Am Rev Respir Dis 1989;<br />

140:1527-37.<br />

6. Lacoste JY, Bousquet J, Chanez P, Van Vyve T, Simony-<br />

Lafontaine J, Lequeu N, et al. Eosinophilic and neutrophilic<br />

inflammation in asthma, chronic bronchitis, and chronic<br />

obstructive pulmonary disease. J Allergy Clin Immunol<br />

1993; 92:537-48.<br />

7. Beeh KM, Beier J, Kommann O, Mander A, Buhl R. Longterm<br />

repeatability of induced sputum cells and inflammatory<br />

markers in stable, moderately severe COPD. Chest<br />

2003; 123:778-83.<br />

8. Stockley RA. Neutrophils and the pathogenesis of COPD.<br />

Chest 2002; 121:151S-155S.<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 39


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 40<br />

9. Peleman RA, Rytila PH, Kips JC, Joos GF, Pauwels RA.<br />

The cellular composition of induced sputum in chronic<br />

obstructive pulmonary disease. Eur Respir J 1999; 13:839-43.<br />

10. Saetta M, Di Stefano A, Turato G, Facchini FM, Corbino L,<br />

Mapp CE, et al. CD8 + T-lymphocytes in peripheral airways<br />

of smokers with chronic obstructive pulmonary disease.<br />

Am J Respir Crit Care Med 1998; 157:822-6.<br />

11. Roth MD, Arora A, Barsky SH, Kleerup EC, Simmons M,<br />

Tashkin DP. Airway inflammation in young marijuana and<br />

tobacco smokers. Am J Respir Crit Care Med 1998;<br />

157:928-37.<br />

12. Keatings VM, Barnes PJ. Granulocyte activation markers in<br />

induced sputum: comparison between chronic obstructive<br />

pulmonary disease, asthma, and normal subjects. Am J<br />

Respir Crit Care Med 1997; 155:449-53.<br />

13. Balbi B, Bason C, Balleari E, Fiasella F, Pesci A, Ghio R,<br />

et al. Increased bronchoalveolar granulocytes and granulocyte/macrophage<br />

colony-stimulating factor during exacerbations<br />

of chronic bronchitis. Eur Respir J 1997; 10:846-50.<br />

14. Peinado VI, Barbera JA, Abate P, Ramirez J, Roca J,<br />

Santos S, et al. Inflammatory reaction in pulmonary muscular<br />

arteries of patients with mild chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med 1999; 159:1605-11.<br />

15. Liu AN, Mohammed AZ, Rice WR, Fiedeldey DT,<br />

Liebermann JS, Whitsett JA, et al. Per<strong>for</strong>in-independent<br />

CD8(+) T-cell-mediated cytotoxicity of alveolar epithelial<br />

cells is preferentially mediated by tumor necrosis factoralpha:<br />

relative insensitivity to Fas ligand. Am J Respir Cell<br />

Mol Biol 1999; 20:849-58.<br />

16. Mueller R, Chanez P, Campbell AM, Bousquet J, Heusser<br />

C, Bullock GR. Different cytokine patterns in bronchial<br />

biopsies in asthma and chronic bronchitis. Respir Med<br />

1996; 90:79-85.<br />

17. Liu H, Lazarus SC, Caughey GH, Fahy JV. Neutrophil elastase<br />

and elastase-rich cystic fibrosis sputum degranulate<br />

human eosinophils in vitro.<br />

Am J Physiol 1999; 276:L28-34.<br />

18. Saetta M, Di Stefano A, Maestrelli P, Turato G, Ruggieri<br />

MP, Roggeri A, et al. Airway eosinophilia in chronic bronchitis<br />

during exacerbations. Am J Respir Crit Care Med<br />

1994; 150:1646-52.<br />

19. Saetta M, Di Stefano A, Maestrelli P, Turato G, Mapp CE,<br />

Pieno M, et al. Airway eosinophilia and expression of interleukin-5<br />

protein in asthma and in exacerbations of chronic<br />

bronchitis. Clin Exp Allergy 1996; 26:766-74.<br />

20. Mills PR, Davies RJ, Devalia JL. Airway epithelial cells,<br />

cytokines, and pollutants. Am J Respir Crit Care Med<br />

1999; 160:S38-43.<br />

21. Di Stefano A, Maestrelli P, Roggeri A, Turato G, Calabro S,<br />

Potena A, et al. Up-regulation of adhesion molecules in the<br />

bronchial mucosa of subjects with chronic obstructive bronchitis.<br />

Am J Respir Crit Care Med 1994; 149:803-10.<br />

22. McElvaney NG, Crystal RG. Proteinases and lung injury.<br />

In: Crystal RG, West JB, eds. The lung: scientific foundations.<br />

Philadelphia: Lippincott-Raven; 1997. p. 2205-18.<br />

23. Shapiro SD. Matrix metalloproteinase degradation of extracellular<br />

matrix: biological consequences. Curr Opin Cell<br />

Biol 1998; 10:602-8.<br />

24. Warren JS, Ward PA. Consequences of oxidant injury. In:<br />

Crystal RG, West JB, eds. The lung: scientific foundations.<br />

Philadelphia: Lippincott-Raven; 1997. p. 2279-88.<br />

25. Ganz T, Lehrer RI. Defensins. Curr Opin Immunol 1994;<br />

6:584-9.<br />

26. Hill AT, Bayley D, Stockley RA. The interrelationship of sputum<br />

inflammatory markers in patients with chronic bronchitis.<br />

Am J Respir Crit Care Med 1999; 160:893-8.<br />

27. Yamamoto C, Yoneda T, Yoshikawa M, Fu A, Tokuyama T,<br />

Tsukaguchi K, et al. Airway inflammation in COPD<br />

assessed by sputum levels of interleukin-8. Chest 1997;<br />

112:505-10.<br />

28. Senior RM, Griffin GL, Mecham RP. Chemotactic activity of<br />

elastin-derived peptides. J Clin Invest 1980; 66:859-62.<br />

29. Heise CE, O'Dowd BF, Figueroa DJ, Sawyer N, Nguyen T,<br />

Im DS, et al. Characterization of the human cysteinyl<br />

leukotriene 2 receptor. J Biol Chem 2000; 275:30531-6.<br />

30. de Godoy I, Donahoe M, Calhoun WJ, Mancino J, Rogers<br />

RM. Elevated TNF-alpha production by peripheral blood<br />

monocytes of weight-losing COPD patients. Am J Respir<br />

Crit Care Med 1996; 153:633-7.<br />

31. Capelli A, Di Stefano A, Gnemmi I, Balbo P, Cerutti CG,<br />

Balbi B, et al. Increased MCP-1 and MIP-1beta in bronchoalveolar<br />

lavage fluid of chronic bronchitics. Eur Respir J<br />

1999; 14:160-5.<br />

32. Hoshi H, Ohno I, Honma M, Tanno Y, Yamauchi K, Tamura<br />

G, et al. IL-5, IL-8 and GM-CSF immunostaining of sputum<br />

cells in bronchial asthma and chronic bronchitis. Clin Exp<br />

Allergy 1995; 25:720-8.<br />

33. Vignola AM, Chanez P, Chiappara G, Merendino A, Pace<br />

E, Rizzo A, et al. Trans<strong>for</strong>ming growth factor-beta expression<br />

in mucosal biopsies in asthma and chronic bronchitis.<br />

Am J Respir Crit Care Med 1997; 156:591-9.<br />

34. de Boer WI, van Schadewijk A, Sont JK, Sharma HS, Stolk<br />

J, Hiemstra PS, et al. Trans<strong>for</strong>ming growth factor beta1 and<br />

recruitment of macrophages and mast cells in airways in<br />

chronic obstructive pulmonary disease. Am J Respir Crit<br />

Care Med 1998; 158:1951-7.<br />

35. Chalmers GW, Macleod KJ, Sriram S, Thomson LJ,<br />

McSharry C, Stack BH, et al. Sputum endothelin-1 is<br />

increased in cystic fibrosis and chronic obstructive pulmonary<br />

disease. Eur Respir J 1999; 13:1288-92.<br />

36. Fujii T, Otsuka T, Tanaka S, Kanazawa H, Hirata K, Kohno<br />

M, et al. Plasma endothelin-1 level in chronic obstructive<br />

pulmonary disease: relationship with natriuretic peptide.<br />

Respiration 1999; 66:212-9.<br />

37. Tomaki M, Ichinose M, Miura M, Hirayama Y, Yamauchi H,<br />

Nakajima N, et al. Elevated substance P content in<br />

induced sputum from patients with asthma and patients<br />

with chronic bronchitis. Am J Respir Crit Care Med 1995;<br />

151:613-7.<br />

40 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 41<br />

38. Lucchini RE, Facchini F, Turato G, Saetta M, Caramori G,<br />

Ciaccia A, et al. Increased VIP-positive nerve fibers in the<br />

mucous glands of subjects with chronic bronchitis. Am J<br />

Respir Crit Care Med 1997; 156:1963-8.<br />

39. Chanez P, Springall D, Vignola AM, Moradoghi-Hattvani A,<br />

Polak JM, Godard P, et al. Bronchial mucosal immunoreactivity<br />

of sensory neuropeptides in severe airway diseases.<br />

Am J Respir Crit Care Med 1998; 158:985-90.<br />

40. Metcalf JP, Thompson AB, Gossman GL, Nelson KJ,<br />

Koyama S, Rennard SI, et al. Gcglobulin functions as a<br />

cochemotaxin in the lower respiratory tract. A potential<br />

mechanism <strong>for</strong> lung neutrophil recruitment in cigarette<br />

smokers. Am Rev Respir Dis 1991; 143:844-9.<br />

41. Wright JL, Lawson LM, Pare PD, Wiggs BJ, Kennedy S,<br />

Hogg JC. Morphology of peripheral airways in current<br />

smokers and ex-smokers. Am Rev Respir Dis 1983;<br />

127:474-7.<br />

42. Mullen JB, Wright JL, Wiggs BR, Pare PD, Hogg JC.<br />

Reassessment of inflammation of airways in chronic bronchitis.<br />

BMJ (Clin Res Ed) 1985; 291:1235-9.<br />

43. Cosio M, Ghezzo H, Hogg JC, Corbin R, Loveland M,<br />

Dosman J, et al. The relations between structural changes<br />

in small airways and pulmonary-function tests. N Engl J<br />

Med 1978; 298:1277-81.<br />

44. McLean KA. Pathogenesis of pulmonary emphysema. Am<br />

J Med 1958; 25:62-74.<br />

45. Niewoehner DE, Kleinerman J, Rice DB. Pathologic<br />

changes in the peripheral airways of young cigarette smokers.<br />

N Engl J Med 1974; 291:755-8.<br />

46. Auerbach O, Garfinkel L, Hammond EC. Relation of smoking<br />

and age to findings in lung parenchyma: a microscopic<br />

study. Chest 1974; 65:29-35.<br />

47. Auerbach O, Hammond EC, Garfinkel L, Benante C.<br />

Relation of smoking and age to emphysema. Whole-lung<br />

section study. N Engl J Med 1972; 286:853-7.<br />

48. Petty TL, Silvers GW, Stan<strong>for</strong>d RE, Baird MD, Mitchell RS.<br />

Small airway pathology is related to increased closing<br />

capacity and abnormal slope of phase III in excised human<br />

lungs. Am Rev Respir Dis 1980; 121:449-56.<br />

49. Ollerenshaw SL, Woolcock AJ. Characteristics of the<br />

inflammation in biopsies from large airways of subjects<br />

with asthma and subjects with chronic airflow limitation.<br />

Am Rev Respir Dis 1992; 145:922-7.<br />

50. Gadek JE, Fells GA, Crystal RG. Cigarette smoking<br />

induces functional antiproteinase deficiency in the lower<br />

respiratory tract of humans. Science 1979; 206:1315-6.<br />

51. Stone PJ, Calore JD, McGowan SE, Bernardo J, Snider<br />

GL, Franzblau C. Functional alpha 1-proteinase inhibitor in<br />

the lower respiratory tract of cigarette smokers is not<br />

decreased. Science 1983; 221:1187-9.<br />

52. Hunninghake GW, Crystal RG. Cigarette smoking and lung<br />

destruction. Accumulation of neutrophils in the lungs of cigarette<br />

smokers. Am Rev Respir Dis 1983; 128:833-8.<br />

53. Mio T, Romberger DJ, Thompson AB, Robbins RA, Heires<br />

A, Rennard SI. Cigarette smoke induces interleukin-8<br />

release from human bronchial epithelial cells. Am J Respir<br />

Crit Care Med 1997; 155:1770-6.<br />

54. Masubuchi T, Koyama S, Sato E, Takamizawa A, Kubo K,<br />

Sekiguchi M, et al. Smoke extract stimulates lung epithelial<br />

cells to release neutrophil and monocyte chemotactic<br />

activity. Am J Pathol 1998; 153:1903-12.<br />

55. Speizer FE, Tager IB. Epidemiology of chronic mucus<br />

hypersecretion and obstructive airways disease. Epidemiol<br />

Rev 1979; 1:124-42.<br />

56. Turato G, Di Stefano A, Maestrelli P, Mapp CE, Ruggieri<br />

MP, Roggeri A, et al. Effect of smoking cessation on airway<br />

inflammation in chronic bronchitis. Am J Respir Crit Care<br />

Med 1995; 152:1262-7.<br />

57. Li XY, Brown D, Smith S, MacNee W, Donaldson K. Shortterm<br />

inflammatory responses following intratracheal instillation<br />

of fine and ultrafine carbon black in rats Inhal Toxicol<br />

1999; 11:709-31.<br />

58. Monn C, Becker S. Cytotoxicity and induction of proinflammatory<br />

cytokines from human monocytes exposed to fine<br />

(PM2.5) and coarse particles (PM10-2.5) in outdoor and<br />

indoor air. Toxicol Appl Pharmacol 1999; 155:245-52.<br />

59. Salvi S, Blomberg A, Rudell B, Kelly F, Sandstrom T,<br />

Holgate ST, et al. Acute inflammatory responses in the airways<br />

and peripheral blood after short-term exposure to<br />

diesel exhaust in healthy human volunteers. Am J Respir<br />

Crit Care Med 1999; 159:702-9.<br />

60. Von Essen SG, O'Neill DP, McGranaghan S, Olenchock<br />

SA, Rennard SI. Neutrophilic respiratory tract inflammation<br />

and peripheral blood neutrophilia after grain sorghum dust<br />

extract challenge. Chest 1995; 108:1425-33.<br />

61. Von Essen SG, Robbins RA, Thompson AB, Ertl RF, Linder<br />

J, Rennard SI. Mechanisms of neutrophil recruitment to the<br />

lung by grain dust exposure [published erratum appears in<br />

Am Rev Respir Dis 1989; 139:1065]. Am Rev Respir Dis<br />

1988; 138:921-7.<br />

62. Laurell CB, Eriksson S. The electrophoretic alpha-1 globulin<br />

pattern of serum in alpha-1 antitrypsin deficiency. Scand<br />

J Clin Lab Invest 1963; 15:132-40.<br />

63. Carp H, Miller F, Hoidal JR, Janoff A. Potential mechanism<br />

of emphysema: alpha 1-proteinase inhibitor recovered from<br />

lungs of cigarette smokers contains oxidized methionine<br />

and has decreased elastase inhibitory capacity. Proc Natl<br />

Acad Sci U S A 1982; 79:2041-5.<br />

64. Cohen AB, James HL. Reduction of the elastase inhibitory<br />

capacity of alpha 1-antitrypsin by peroxides in cigarette<br />

smoke: an analysis of brands and filters. Am Rev Respir<br />

Dis 1982; 126:25-30.<br />

65. Shapiro SD. Elastolytic metalloproteinases produced by<br />

human mononuclear phagocytes. Potential roles in<br />

destructive lung disease. Am J Respir Crit Care Med 1994;<br />

150:S160-4.<br />

66. Sommerhoff CP, Nadel JA, Basbaum CB, Caughey GH.<br />

Neutrophil elastase and cathepsin G stimulate secretion<br />

from cultured bovine airway gland serous cells. J Clin<br />

Invest 1990; 85:682-9.<br />

67. Witko-Sarsat V, Halbwachs-Mecarelli L, Schuster A,<br />

Nusbaum P, Ueki I, Canteloup S, et al. Proteinase 3, a<br />

potent secretagogue in airways, is present in cystic fibrosis<br />

sputum. Am J Respir Cell Mol Biol 1999; 20:729-36.<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 41


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 42<br />

68. Christensen TG, Korthy AL, Snider GL, Hayes JA.<br />

Irreversible bronchial goblet cell metaplasia in hamsters<br />

with elastase-induced panacinar emphysema. J Clin Invest<br />

1977; 59:397-404.<br />

69. Dekhuijzen PN, Aben KK, Dekker I, Aarts LP, Wielders PL,<br />

van Herwaarden CL, et al. Increased exhalation of hydrogen<br />

peroxide in patients with stable and unstable chronic<br />

obstructive pulmonary disease. Am J Respir Crit Care Med<br />

1996; 154:813-6.<br />

70. Maziak W, Loukides S, Culpitt S, Sullivan P, Kharitonov SA,<br />

Barnes PJ. Exhaled nitric oxide in chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med 1998;<br />

157:998-1002.<br />

71. Montuschi P, Collins JV, Ciabattoni G, Lazzeri N, Corradi<br />

M, Kharitonov SA, et al. Exhaled 8-isoprostane as an in<br />

vivo biomarker of lung oxidative stress in patients with<br />

COPD and healthy smokers. Am J Respir Crit Care Med<br />

2000; 162:1175-7.<br />

72. Pratico D, Basili S, Vieri M, Cordova C, Violi F, Fitzgerald<br />

GA. <strong>Chronic</strong> obstructive pulmonary disease is associated<br />

with an increase in urinary levels of isoprostane F2alpha-<br />

III, an index of oxidant stress. Am J Respir Crit Care Med<br />

1998; 158:1709-14.<br />

73. Kawikova I, Barnes PJ, Takahashi T, Tadjkarimi S, Yacoub<br />

MH, Belvisi MG. 8-Epi-PGF2 alpha, a novel noncyclooxygenase-derived<br />

prostaglandin, constricts airways in vitro.<br />

Am J Respir Crit Care Med 1996; 153:590-6.<br />

74. Ciba Guest Symposium Report. Terminiology, definitions<br />

and classifications of chronic pulmonary emphysema and<br />

related conditions. Thorax 1959; 14:286-99.<br />

75. Pride NB, Burrows B. Development of impaired lung function:<br />

natural history and risk factors. In: Calverley PM,<br />

Pride NB, eds. <strong>Chronic</strong> obstructive lung disease. London<br />

and Glasgow: Chapman and Hall Medical; 1995. p. 69-71.<br />

76. Saetta M, Di Stefano A, Maestrelli P, Ferraresso A, Drigo<br />

R, Potena A, et al. Activated T-lymphocytes and<br />

macrophages in bronchial mucosa of subjects with chronic<br />

bronchitis. Am Rev Respir Dis 1993; 147:301-6.<br />

77. Reid L. Measurement of the bronchial mucous gland layer:<br />

a diagnostic yardstick in chronic bronchitis. Thorax 1960;<br />

15:132-41.<br />

78. Jamal K, Cooney TP, Fleetham JA, Thurlbeck WM. <strong>Chronic</strong><br />

bronchitis. Correlation of morphologic findings to sputum<br />

production and flow rates. Am Rev Respir Dis 1984;<br />

129:719-22.<br />

79. Haraguchi M, Shimura S, Shirato K. Morphometric analysis<br />

of bronchial cartilage in chronic obstructive pulmonary disease<br />

and bronchial asthma. Am J Respir Crit Care Med<br />

1999; 159:1005-13.<br />

80. Thurlbeck WM, Pun R, Toth J, Frazer RG. Bronchial cartilage<br />

in chronic obstructive lung disease. Am Rev Respir<br />

Dis 1974; 109:73-80.<br />

81. Snider GL. Parker B. Francis Lecture. Animal models of<br />

chronic airways injury. Chest 1992; 101:74-9S.<br />

82. Snider GL, Faling LJ, Rennard SI. <strong>Chronic</strong> bronchitis and<br />

emphysema. In: Murray JF, Nadel JA, eds. Textbook of respiratory<br />

medicine. Philadelphia: WB Saunders; 2000. p.<br />

1187-246.<br />

83. Rogers DF, Jeffery PK. Inhibition by oral N-acetylcysteine<br />

of cigarette smoke-induced "bronchitis" in the rat. Exp <strong>Lung</strong><br />

Res 1986; 10:267-83.<br />

84. Nakamura Y, Romberger DJ, Tate L, Ertl RF, Kawamoto M,<br />

Adachi Y, et al. Cigarette smoke inhibits lung fibroblast proliferation<br />

and chemotaxis. Am J Respir Crit Care Med<br />

1995; 151:1497-503.<br />

85. Osman M, Cantor JO, Roffman S, Keller S, Turino GM,<br />

Mandl I. Cigarette smoke impairs elastin resynthesis in<br />

lungs of hamsters with elastase-induced emphysema. Am<br />

Rev Respir Dis 1985; 132:640-3.<br />

86. Laurent P, Janoff A, Kagan HM. Cigarette smoke blocks<br />

cross-linking of elastin in vitro. Chest 1983; 83:63-5S.<br />

87. Matsuba K, Thurlbeck WM. The number and dimensions of<br />

small airways in emphysematous lungs. Am J Pathol 1972;<br />

67:265-75.<br />

88. Hogg JC, Macklem PT, Thurlbeck WM. Site and nature of<br />

airway obstruction in chronic obstructive lung disease. N<br />

Engl J Med 1968; 278:1355-60.<br />

89. Rennard SI. Inflammation and repair processes in chronic<br />

obstructive pulmonary disease. Am J Respir Crit Care Med<br />

1999; 160:S12-6.<br />

90. Leopold JG, Goeff J. Centrilobular <strong>for</strong>m of hypertrophic<br />

emphysema and its relation to chronic bronchitis. Thorax<br />

1957; 12:219-35.<br />

91. Repine JE, Bast A, Lankhorst I. Oxidative stress in chronic<br />

obstructive pulmonary disease. Oxidative Stress Study<br />

Group. Am J Respir Crit Care Med 1997; 156:341-57.<br />

92. Wright JL, Lawson L, Pare PD, Hooper RO, Peretz DI,<br />

Nelems JM, et al. The structure and function of the pulmonary<br />

vasculature in mild chronic obstructive pulmonary<br />

disease. The effect of oxygen and exercise. Am Rev Respir<br />

Dis 1983; 128:702-7.<br />

93. Sekhon HS, Wright JL, Churg A. Cigarette smoke causes<br />

rapid cell proliferation in small airways and associated pulmonary<br />

arteries. Am J Physiol 1994; 267:L557-63.<br />

94. Peinado VI, Barbera JA, Ramirez J, Gomez FP, Roca J,<br />

Jover L, et al. Endothelial dysfunction in pulmonary arteries<br />

of patients with mild COPD. Am J Physiol 1998; 274:L908-13.<br />

95. Riley DJ, Thakker-Varia S, Poiani GJ, Tozzi CA. Vascular<br />

remodeling. In: Crystal RG, West JB, Barnes PJ, Weibel<br />

ER, eds. The lung: scientific foundations. Philadelphia:<br />

Lippincott-Raven; 1977. p. 1589-97.<br />

96. Kuwano K, Bosken CH, Pare PD, Bai TR, Wiggs BR, Hogg<br />

JC. Small airways dimensions in asthma and in chronic<br />

obstructive pulmonary disease. Am Rev Respir Dis 1993;<br />

148:1220-5.<br />

97. Matsuba K, Wright JL, Wiggs BR, Pare PD, Hogg JC. The<br />

changes in airways structure associated with reduced<br />

<strong>for</strong>ced expiratory volume in one second. Eur Respir J<br />

1989; 2:834-9.<br />

98. Dayman H. Mechanics of airflow in health and emphysema.<br />

J Clin Invest 1951; 30:1175-90.<br />

99. Butler J, Caro C, Alkaler R, Dubois AB. Physiological factors<br />

affecting airway resistance in normal subjects and in<br />

patients with obstructive airways disease. J Clin Invest<br />

1960; 39:584-91.<br />

42 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 43<br />

100.Mead J, Turner JM, Macklem PT, Little JB. Significance of<br />

the relationship between lung recoil and maximum expiratory<br />

flow. J Appl Physiol 1967; 22:95-108.<br />

101.Burnett D, Stockley RA. Serum and sputum alpha 2<br />

macroglobulin in patients with chronic obstructive airways<br />

disease. Thorax 1981; 36:512-6.<br />

102.Lane DJ, Howell JB, Giblin B. Relation between airways<br />

obstruction and CO 2 tension in chronic obstructive airways<br />

disease. BMJ 1968; 3:707-9.<br />

103.Rodriguez-Roisin R, MacNee W. Pathophysiology of chronic<br />

obstructive pulmonary disease. In: Postma DS, Siafakas<br />

NM, eds. Management of chronic obstructive pulmonary<br />

disease. European Respiratory Monograph 1998; 3:107-26.<br />

104.McLean A, Warren PM, Gillooly M, MacNee W, Lamb D.<br />

Microscopic and macroscopic measurements of emphysema:<br />

relation to carbon monoxide gas transfer. Thorax 1992;<br />

47:144-9.<br />

105.Barbera JA, Riverola A, Roca J, Ramirez J, Wagner PD,<br />

Ros D, et al. Pulmonary vascular abnormalities and ventilation-perfusion<br />

relationships in mild chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med 1994;<br />

149:423-9.<br />

106.MacNee W. Pathophysiology of cor pulmonale in chronic<br />

obstructive pulmonary disease. Part two. Am J Respir Crit<br />

Care Med 1994; 150:1158-68.<br />

107.Knighton DR, Hunt TK, Scheuenstuhl H, Halliday BJ, Werb<br />

Z, Banda MJ. Oxygen tension regulates the expression of<br />

angiogenesis factor by macrophages. Science 1983;<br />

221:1283-5.<br />

108.Biernacki W, Flenley DC, Muir AL, MacNee W. Pulmonary<br />

hypertension and right ventricular function in patients with<br />

COPD. Chest 1988; 94:1169-75.<br />

109.Rahman I, Morrison D, Donaldson K, MacNee W. Systemic<br />

oxidative stress in asthma, COPD, and smokers. Am J<br />

Respir Crit Care Med 1996; 154:1055-60.<br />

110.Schols AM, Buurman WA, Staal van den Brekel AJ,<br />

Dentener MA, Wouters EF. Evidence <strong>for</strong> a relation between<br />

metabolic derangements and increased levels of inflammatory<br />

mediators in a subgroup of patients with chronic<br />

obstructive pulmonary disease. Thorax 1996; 51:819-24.<br />

111.Sauleda J, Garcia-Palmer FJ, Gonzalez G, Palou A, Agusti<br />

AG. The activity of cytochrome oxidase is increased in circulating<br />

lymphocytes of patients with chronic obstructive<br />

pulmonary disease, asthma, and chronic arthritis. Am J<br />

Respir Crit Care Med 2000; 161:32-5.<br />

112.Sauleda J, Garcia-Palmer F, Wiesner RJ, Tarraga S,<br />

Harting I, Tomas P, et al. Cytochrome oxidase activity and<br />

mitochondrial gene expression in skeletal muscle of<br />

patients with chronic obstructive pulmonary disease. Am J<br />

Respir Crit Care Med 1998; 157:1413-7.<br />

113.American Thoracic Society and European Respiratory<br />

Society. Skeletal muscle dysfunction in chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med 1999;<br />

159:S1-40.<br />

114.Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss<br />

is a reversible factor in the prognosis of chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med 1998;<br />

157:1791-7.<br />

115.Pizzichini MM, Pizzichini E, Efthimiadis A, Clelland L,<br />

Mahony JB, Dolovich J, et al. Markers of inflammation in<br />

induced sputum in acute bronchitis caused by Chlamydia<br />

pneumoniae. Thorax 1997; 52:929-31; discussion 6-7.<br />

116.Pizzichini E, Pizzichini MM, Gibson P, Parameswaran K,<br />

Gleich GJ, Berman L, et al. Sputum eosinophilia predicts<br />

benefit from prednisone in smokers with chronic obstructive<br />

bronchitis. Am J Respir Crit Care Med 1998; 158:1511-7.<br />

117.Maestrelli P, Saetta M, Di Stefano A, Calcagni PG, Turato<br />

G, Ruggieri MP, et al. Comparison of leukocyte counts in<br />

sputum, bronchial biopsies, and bronchoalveolar lavage.<br />

Am J Respir Crit Care Med 1995; 152:1926-31.<br />

118.Turner MO, Hussack P, Sears MR, Dolovich J, Hargreave<br />

FE. Exacerbations of asthma without sputum eosinophilia.<br />

Thorax 1995; 50:1057-61.<br />

119.Fahy JV, Kim KW, Liu J, Boushey HA. Prominent neutrophilic<br />

inflammation in sputum from subjects with asthma<br />

exacerbation. J Allergy Clin Immunol 1995; 95:843-52.<br />

120.Barbera JA, Roca J, Ferrer A, Felez MA, Diaz O, Roger N,<br />

et al. Mechanisms of worsening gas exchange during<br />

acute exacerbations of chronic obstructive pulmonary disease.<br />

Eur Respir J 1997; 10:1285-91.<br />

121.Seemungal TA, Donaldson GC, Bhowmik A, Jeffries DJ,<br />

Wedzicha JA. Time course and recovery of exacerbations<br />

in patients with chronic obstructive pulmonary disease. Am<br />

J Respir Crit Care Med 2000; 161:1608-13.<br />

122.Schmidt GA, Hall JB. Acute or chronic respiratory failure.<br />

Assessment and management of patients with COPD in<br />

the emergency setting. JAMA 1989; 261:3444-53.<br />

123.Rodriguez-Roisin R. Pulmonary gas exchange in acute respiratory<br />

failure. Eur J Anaesthesiol 1994; 11:5-13.<br />

PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY 43


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 44<br />

44 PATHOGENESIS, PATHOLOGY, AND PATHOPHYSIOLOGY


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 45<br />

CHAPTER<br />

5<br />

MANAGEMENT<br />

OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 46<br />

CHAPTER 5: MANAGEMENT OF COPD<br />

INTRODUCTION<br />

Management of Mild to Moderate COPD (Stages I and II)<br />

involves the avoidance of risk factors to prevent disease<br />

progression and pharmacotherapy as needed to control<br />

symptoms. Severe (Stage III) and very severe (Stage IV)<br />

disease often require the integration of several different<br />

disciplines, a variety of treatment approaches, and a<br />

commitment of the clinician to the continued support of<br />

the patient as the illness progresses. In addition to<br />

patient education, health advice, and pharmacotherapy,<br />

COPD patients may require specific counseling about<br />

smoking cessation, instruction in physical exercise,<br />

nutritional advice, and continued nursing support. Not all<br />

approaches are needed <strong>for</strong> every patient, and assessing<br />

the potential benefit of each approach at each stage of<br />

the illness is a crucial aspect of effective disease<br />

management.<br />

An effective COPD management plan includes four<br />

components: (1) Assess and Monitor <strong>Disease</strong>; (2)<br />

Reduce Risk Factors; (3) Manage Stable COPD; (4)<br />

Manage Exacerbations.<br />

While disease prevention is the ultimate goal, once<br />

COPD has been diagnosed, effective management<br />

should be aimed at the following goals:<br />

• Prevent disease progression.<br />

• Relieve symptoms.<br />

• Improve exercise tolerance.<br />

• Improve health status.<br />

• Prevent and treat complications.<br />

• Prevent and treat exacerbations.<br />

• Reduce mortality.<br />

These goals should be reached with minimal side effects<br />

from treatment, a particular challenge in COPD patients<br />

because they commonly have comorbidities. The extent<br />

to which these goals can be realized varies with each<br />

individual, and some treatments will produce benefits in<br />

more than one area. In selecting a treatment plan, the<br />

benefits and risks to the individual, and the costs, direct<br />

and indirect, to the individual, his or her family, and the<br />

community must be considered.<br />

Patients should be identified as early in the course of the<br />

disease as possible, and certainly be<strong>for</strong>e the end stage<br />

of the illness when disability is substantial. However, the<br />

benefits of community-based spirometric screening, of<br />

either the general population or smokers, are still unclear.<br />

Educating patients and physicians to recognize that<br />

cough, sputum production, and especially breathlessness<br />

are not trivial symptoms is an essential aspect of the<br />

public health care of this disease.<br />

Reduction of therapy once symptom control has been<br />

achieved is not normally possible in COPD. Further<br />

deterioration of lung function usually requires the<br />

progressive introduction of more treatments, both<br />

pharmacologic and non-pharmacologic, to attempt to limit<br />

the impact of these changes. Exacerbations of signs and<br />

symptoms, a hallmark of COPD, impair patients' quality<br />

of life and decrease their health status 1,2 . Appropriate<br />

treatment and measures to prevent further exacerbations<br />

should be implemented as quickly as possible.<br />

Important differences exist between countries in the<br />

approach to chronic illnesses such as COPD and in the<br />

acceptability of particular <strong>for</strong>ms of therapy. Ethnic<br />

differences in drug metabolism, especially <strong>for</strong> oral<br />

medications, may result in different patient preferences<br />

in different communities. Little is known about these<br />

important issues in relationship to COPD.<br />

REFERENCES<br />

1. O'Brien C, Guest PJ, Hill SL, Stockley RA. Physiological and<br />

radiological characterization of patients diagnosed with<br />

chronic obstructive pulmonary disease in primary care.<br />

Thorax 2000; 55:635-42.<br />

2. Seemungal TA, Donaldson GC, Bhowmik A, Jeffries DJ,<br />

Wedzicha JA. Time course and recovery of exacerbations in<br />

patients with chronic obstructive pulmonary disease. Am J<br />

Respir Crit Care Med 2000; 161:1608-13.<br />

46 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 47<br />

COMPONENT 1: ASSESS AND MONITOR DISEASE<br />

KEY POINTS:<br />

• Diagnosis of COPD is based on a history of<br />

exposure to risk factors and the presence of<br />

airflow limitation that is not fully reversible, with<br />

or without the presence of symptoms.<br />

• Patients who have chronic cough and sputum<br />

production with a history of exposure to risk<br />

factors should be tested <strong>for</strong> airflow limitation,<br />

even if they do not have dyspnea.<br />

• For the diagnosis and assessment of COPD,<br />

spirometry is the gold standard as it is the most<br />

reproducible, standardized, and objective way of<br />

measuring airflow limitation. FEV 1 /FVC < 70%<br />

and a postbronchodilator FEV 1 < 80% predicted<br />

confirms the presence of airflow limitation that is<br />

not fully reversible.<br />

• Health care workers involved in the diagnosis<br />

and management of COPD patients should have<br />

access to spirometry.<br />

• Measurement of arterial blood gas tensions<br />

should be considered in all patients with FEV 1 <<br />

40% predicted or clinical signs suggestive of<br />

respiratory failure or right heart failure.<br />

INITIAL DIAGNOSIS<br />

A diagnosis of COPD should be considered in any patient<br />

who has cough, sputum production, or dyspnea, and/or a<br />

history of exposure to risk factors <strong>for</strong> the disease (Figure<br />

5-1-1). The diagnosis is confirmed by spirometry. The<br />

presence of a postbronchodilator FEV 1 < 80% of the<br />

predicted value in combination with an FEV 1 /FVC < 70%<br />

confirms the presence of airflow limitation that is not fully<br />

reversible. Where spirometry is unavailable, the diagnosis<br />

of COPD should be made using all available tools.<br />

Clinical symptoms and signs, such as abnormal shortness<br />

of breath and increased <strong>for</strong>ced expiratory time, can be<br />

used to help with the diagnosis. A low peak flow is<br />

consistent with COPD, but has poor specificity since it<br />

can be caused by other lung diseases and by poor<br />

per<strong>for</strong>mance. In the interest of improving the diagnosis of<br />

COPD, every ef<strong>for</strong>t should be made to provide access to<br />

standardized spirometry.<br />

Assessment of Symptoms<br />

Although exceptions occur, the general patterns of<br />

symptom development in COPD are well established.<br />

The main symptoms among patients in Stage 0: At Risk<br />

and Stage I: Mild COPD are chronic cough and sputum<br />

production. These symptoms can be present <strong>for</strong> many<br />

years be<strong>for</strong>e the development of airflow limitation and<br />

are often ignored or discounted by patients. As airflow<br />

limitation develops in Stage II: Moderate COPD, patients<br />

often experience dyspnea, which may interfere with their<br />

Figure 5-1-1. Key Indicators <strong>for</strong><br />

Considering a Diagnosis of COPD<br />

Consider COPD, and per<strong>for</strong>m spirometry, if any of<br />

these indicators are present. These indicators are not<br />

diagnostic by themselves, but the presence of multiple<br />

key indicators increases the probability of a diagnosis<br />

of COPD. Spirometry is needed to establish a<br />

diagnosis of COPD.<br />

<strong>Chronic</strong> cough:<br />

Present intermittently or every day.<br />

Often present throughout the day;<br />

seldom only nocturnal.<br />

<strong>Chronic</strong> sputum Any pattern of chronic sputum<br />

production: production may indicate COPD.<br />

Dyspnea that is:<br />

History of<br />

exposure to<br />

risk factors,<br />

especially:<br />

Progressive (worsens over time).<br />

Persistent (present every day).<br />

Described by the patient as an<br />

“increased ef<strong>for</strong>t to breathe,”<br />

“heavi-ness,” “air hunger,” or “gasping.”<br />

Worse on exercise.<br />

Worse during respiratory infections.<br />

Tobacco smoke.<br />

Occupational dusts and chemicals.<br />

Smoke from home cooking and<br />

heating fuels.<br />

MANAGEMENT OF COPD 47


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 48<br />

daily activities. Typically, this is the stage at which they<br />

seek medical attention and are diagnosed with COPD.<br />

However, some patients do not experience cough, sputum<br />

production, or dyspnea in Stage I: Mild COPD or<br />

Stage II: Moderate COPD, and do not come to medical<br />

attention until their airflow limitation becomes more<br />

severe or their lung function is worsened acutely by a<br />

respiratory tract infection. As airflow limitation worsens<br />

and the patient enters Stage III: Severe COPD, the symptoms<br />

of cough and sputum production typically continue,<br />

dyspnea worsens, and additional symptoms heralding<br />

complications may develop. It is important to note that,<br />

since COPD may be diagnosed at any stage, any of the<br />

symptoms described below may be present in a patient<br />

presenting <strong>for</strong> the first time.<br />

Figure 5-1-2. Causes of <strong>Chronic</strong> Cough<br />

with a Normal Chest X-ray<br />

Intrathoracic<br />

• <strong>Chronic</strong> obstructive pulmonary disease<br />

• Bronchial asthma<br />

• Central bronchial carcinoma<br />

• Endobronchial tuberculosis<br />

• Bronchiectasis<br />

• Left heart failure<br />

• Interstitial lung disease<br />

• Cystic fibrosis<br />

Extrathoracic<br />

• Postnasal drip<br />

• Gastroesophageal reflux<br />

• Drug therapy (e.g., ACE inhibitors)<br />

Figure 5-1-3. Questionnaire <strong>for</strong> Assessing the<br />

Impact of Respiratory Symptoms 6<br />

WHEEZING<br />

Does your chest ever sound wheezing or whistling? Yes ❑<br />

No ❑<br />

IF YOU ANSWERED “YES” TO THIS QUESTION:<br />

Do you get this on most days – or nights? Yes ❑<br />

No ❑<br />

Have you ever had attacks of shortness of breath Yes ❑<br />

with wheezing? No ❑<br />

IF YOU ANSWERED “YES” TO THIS QUESTION:<br />

Is/was your breathing absolutely normal between Yes ❑<br />

attacks? No ❑<br />

CHEST ILLNESSES<br />

During the last three years have you had any chest Yes ❑<br />

illnesses which have kept you from your usual No ❑<br />

activities <strong>for</strong> as much as a week?<br />

IF YOU ANSWERED YES TO THIS QUESTION:<br />

Did you bring up phlegm more than usual during Yes ❑<br />

these illnesses? No ❑<br />

IF YOU ANSWERED YES TO THIS QUESTION:<br />

Have you had more than one illness like this in the Yes ❑<br />

past three years? No ❑<br />

Cough. <strong>Chronic</strong> cough, usually the first symptom of<br />

COPD to develop 1 , is often discounted by the patient as<br />

an expected consequence of smoking and/or environmental<br />

exposures. Initially, the cough may be intermittent,<br />

but later is present every day, often throughout the<br />

day, and is seldom entirely nocturnal. The chronic cough<br />

in COPD may be unproductive 2 . In some cases, significant<br />

airflow limitation may develop without the presence<br />

of a cough. Figure 5-1-2 lists some of the other causes<br />

of chronic cough in individuals with a normal chest X-ray.<br />

Sputum production. COPD patients commonly raise<br />

small quantities of tenacious sputum after coughing<br />

bouts. Regular production of sputum <strong>for</strong> 3 or more<br />

months in 2 consecutive years is the epidemiological definition<br />

of chronic bronchitis 3 , but this is a somewhat arbitrary<br />

definition that does not reflect the range of sputum<br />

production in COPD patients. Sputum production is often<br />

BREATHLESSNESS<br />

PLEASE TICK IN THE BOX THAT APPLIES TO YOU<br />

(ONE BOX ONLY)<br />

I only get breathless with strenuous exercise.<br />

I get short of breath when hurrying on the level or<br />

walking up a slight hill.<br />

I walk slower than people of the same age on the level<br />

because of breathlessness, or I have to stop <strong>for</strong> breath<br />

when walking on my own pace on the level.<br />

I stop <strong>for</strong> breath after walking about 100 yards or after a<br />

few minutes on the level.<br />

I am too breathless to leave the house or I am breathless<br />

when dressing or undressing.<br />

❑<br />

❑<br />

❑<br />

❑<br />

❑<br />

48 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 49<br />

difficult to evaluate because patients may swallow sputum<br />

rather than expectorate it, a habit subject to significant<br />

cultural and gender variation.<br />

Dyspnea. Dyspnea, the hallmark symptom of COPD, is<br />

the reason most patients seek medical attention and is a<br />

major cause of disability and anxiety associated with the<br />

disease. Typical COPD patients describe their dyspnea<br />

as a sense of increased ef<strong>for</strong>t to breathe, heaviness, air<br />

hunger, or gasping 4 . The terms used to describe dyspnea<br />

vary both by individual and by culture 5 . It is often possible<br />

to distinguish the breathlessness of COPD from that due<br />

to other causes by analysis of the terms used, although<br />

there is considerable overlap with descriptors of bronchial<br />

asthma. A simple way to quantify the impact of breathlessness<br />

on a patient’s health status is the British<br />

Medical Research Council (MRC) questionnaire (Figure<br />

5-1-3). This questionnaire relates well to other measures<br />

of health status 6 .<br />

Breathlessness in COPD is characteristically persistent<br />

and progressive. Even on “good days” COPD patients<br />

experience dyspnea at lower levels of exercise than<br />

unaffected people of the same age. Initially, breathlessness<br />

is only noted on unusual ef<strong>for</strong>t (e.g., walking or running<br />

up a flight of stairs) and may be avoided entirely by<br />

appropriate behavioral change (e.g., using an elevator).<br />

As lung function deteriorates, breathlessness becomes<br />

more intrusive, and patients may notice that they are<br />

unable to walk at the same speed as other people of the<br />

same age or carry out activities that require use of the<br />

accessory respiratory muscles (e.g., carrying grocery<br />

bags) 7 . Eventually, breathlessness is present during<br />

everyday activities (e.g., dressing, washing) or at rest,<br />

leaving the patient confined to the home.<br />

Wheezing and chest tightness. Wheezing and chest<br />

tightness are relatively non-specific symptoms that may<br />

vary between days, and over the course of a single day.<br />

These symptoms may be present in Stage I: Mild COPD,<br />

but are more characteristic of asthma or Stage III:<br />

Severe COPD and Stage IV: Very Severe COPD. Audible<br />

wheeze may arise at a laryngeal level and need not be<br />

accompanied by ausculatory abnormalities. Alternatively,<br />

widespread inspiratory or expiratory wheezes can be<br />

present on listening to the chest. Chest tightness often<br />

follows exertion, is poorly localized, is muscular in<br />

character, and may arise from isometric contraction of the<br />

intercostal muscles. An absence of wheezing or chest<br />

tightness does not exclude a diagnosis of COPD.<br />

Additional symptoms in severe disease. Weight loss<br />

and anorexia are common problems in advanced COPD 8 .<br />

Hemoptysis can occur during respiratory tract infections<br />

in COPD patients 9 . However, this can be a sign of other<br />

diseases (e.g., tuberculosis, bronchial tumors) and<br />

there<strong>for</strong>e should always be investigated. Cough syncope<br />

occurs due to rapid increases in intrathoracic pressure<br />

during attacks of coughing. Coughing spells may also<br />

cause ribfractures, which are sometimes asymptomatic.<br />

Psychiatric morbidity, especially symptoms of depression<br />

and/or anxiety, is common in advanced COPD 10 . Ankle<br />

swelling can be the only symptomatic pointer to the<br />

development of cor pulmonale.<br />

Medical History<br />

A detailed medical history of a new patient known or<br />

thought to have COPD should assess:<br />

• Patient’s exposure to risk factors, such as smoking<br />

and occupational or environmental exposures.<br />

• Past medical history, including asthma, allergy,<br />

sinusitis or nasal polyps, respiratory infections in<br />

childhood, other respiratory diseases.<br />

• Family history of COPD or other chronic respiratory<br />

disease.<br />

• Pattern of symptom development: COPD typically<br />

develops in adult life and most patients are<br />

conscious of increased breathlessness, more<br />

frequent “winter colds,” and some social restriction<br />

<strong>for</strong> a number of years be<strong>for</strong>e seeking medical help.<br />

• History of exacerbations or previous hospitalizations<br />

<strong>for</strong> respiratory disorder: Patients may be aware of<br />

periodic worsening of symptoms even if these<br />

episodes have not been identified as exacerbations<br />

of COPD.<br />

• Presence of comorbidities, such as heart disease<br />

and rheumatic disease, which may also contribute to<br />

restriction of activity.<br />

• Appropriateness of current medical treatments:<br />

For example, beta-blockers commonly prescribed<br />

<strong>for</strong> heart disease are usually contraindicated in<br />

COPD.<br />

• Impact of disease on patient’s life, including limitation<br />

of activity; missed work and economic impact; effect<br />

on family routines; feelings of depression or anxiety.<br />

• Social and family support available to the patient.<br />

• Possibilities <strong>for</strong> reducing risk factors, especially<br />

smoking cessation.<br />

MANAGEMENT OF COPD 49


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 50<br />

Physical Examination<br />

Though an important part of patient care, a physical<br />

examination is rarely diagnostic in COPD. Physical signs<br />

of airflow limitation are usually not present until significant<br />

impairment of lung function has occurred 11,12 , and their<br />

detection has a relatively low sensitivity and specificity.<br />

A number of physical signs may be present in COPD, but<br />

their absence does not exclude the diagnosis.<br />

Inspection.<br />

• Central cyanosis, or bluish discoloration of the mucosal<br />

membranes, may be present but is difficult to detect<br />

in artificial light and in many racial groups.<br />

• Common chest wall abnormalities, which reflect the<br />

pulmonary hyperinflation seen in COPD, include<br />

relatively horizontal ribs, “barrel- shaped” chest, and<br />

protruding abdomen.<br />

• Flattening of the hemi-diaphragms may be associated<br />

with paradoxical in-drawing of the lower rib cage on<br />

inspiration, reduced cardiac dullness, and widening<br />

xiphisternal angle.<br />

• Resting respiratory rate is often increased to more<br />

than 20 breaths per minute and breathing can be<br />

relatively shallow 12 .<br />

• Patients commonly show pursed-lip breathing, which<br />

may serve to slow expiratory flow and permit more<br />

efficient lung emptying.<br />

• COPD patients often have resting muscle activation<br />

while lying supine. Use of the scalene and sternocleidomastoid<br />

muscles is a further indicator of<br />

respiratory distress.<br />

• Ankle or lower leg edema can be a sign of right heart<br />

failure.<br />

Palpation and percussion.<br />

• These are often unhelpful in COPD.<br />

• Detection of the heart apex beat may be difficult due<br />

to pulmonary hyperinflation.<br />

• Hyperinflation also leads to downward displacement<br />

of the liver and an increase in the ability to palpate<br />

this organ without it being enlarged.<br />

Auscultation.<br />

• Patients with COPD often have reduced breath<br />

sounds, but this finding is not sufficiently characteristic<br />

to make the diagnosis 13 .<br />

Figure 5-1-4. Considerations in<br />

Per<strong>for</strong>ming Spirometry<br />

Preparation<br />

• Spirometers need calibration on a regular basis.<br />

• Spirometers should produce hard copy to permit<br />

detection of technical errors.<br />

• The supervisor of the test needs training in its effective<br />

per<strong>for</strong>mance.<br />

• Maximal patient ef<strong>for</strong>t in per<strong>for</strong>ming the test is<br />

required to avoid errors in diagnosis and management.<br />

Per<strong>for</strong>mance<br />

• Spirometry should be per<strong>for</strong>med using techniques that<br />

meet published standards 14 .<br />

• The expiratory volume/time traces should be smooth<br />

and free from irregularities.<br />

• The recording should go on long enough <strong>for</strong> a volume<br />

plateau to be reached, which may take more than 12<br />

seconds in severe disease.<br />

• Both FVC and FEV 1 should be the largest value<br />

obtained from any of 3 technically satisfactory curves<br />

and the FVC and FEV 1 values in these three curves<br />

should vary by no more than 5% or 100 ml, whichever<br />

is greater.<br />

Evaluation<br />

• Spirometry measurements are evaluated by comparison<br />

of the results with appropriate reference values based<br />

on age, height, sex, and race (e.g., see reference 14).<br />

• The presence of a postbronchodilator FEV 1 < 80%<br />

predicted together with an FEV 1 /FVC < 70% confirms<br />

the presence of airflow limitation that is not fully<br />

reversible.<br />

• In patients with FEV 1 > 80% predicted, FEV 1 /FVC <<br />

70% may be an early indicator of developing airflow<br />

limitation.<br />

• The presence of wheezing during quiet breathing is a<br />

useful pointer to airflow limitation. However, wheezing<br />

heard only after <strong>for</strong>ced expiration is of no diagnostic<br />

value.<br />

• Inspiratory crackles occur in some COPD patients but<br />

are of little help diagnostically.<br />

• Heart sounds are best heard over the xiphoid area.<br />

Measurement of Airflow Limitation<br />

(Spirometry)<br />

Spirometry measurements should be undertaken <strong>for</strong> any<br />

patient who may have COPD. To help identify individuals<br />

earlier in the course of the disease, spirometry should be<br />

50 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 51<br />

per<strong>for</strong>med <strong>for</strong> patients who have chronic cough and<br />

sputum production even if they do not have dyspnea.<br />

Although spirometry does not fully capture the impact of<br />

COPD on a patient’s health, it remains the gold standard<br />

<strong>for</strong> diagnosing the disease and monitoring its progression.<br />

It is the best standardized, most reproducible, and most<br />

objective measurement of airflow limitation available.<br />

Health care workers who care <strong>for</strong> COPD patients should<br />

have access to spirometry, which is useful in both diagnosis<br />

and periodic monitoring. Figure 5-1-4 summarizes some<br />

considerations that are crucial to achieving consistently<br />

accurate test results.<br />

Spirometry should measure the maximal volume of air<br />

<strong>for</strong>cibly exhaled from the point of maximal inspiration<br />

(<strong>for</strong>ced vital capacity, FVC) and the volume of air exhaled<br />

during the first second of this maneuver (<strong>for</strong>ced expiratory<br />

volume in one second, FEV 1 ), and the ratio of these two<br />

measurements (FEV 1 /FVC) should be calculated.<br />

Spirometry measurements are evaluated by comparison<br />

with reference values based on age, height, sex, and race<br />

(use appropriate reference values, e.g., see reference 14).<br />

Figure 5-1-5 shows a normal spirogram and a spirogram<br />

typical of patients with mild to moderate COPD. Patients<br />

with COPD typically show a decrease in both FEV 1 and<br />

FVC. The degree of spirometric abnormality generally<br />

reflects the severity of COPD (Figure 1-2). The presence<br />

of a postbronchodilator FEV 1 < 80% of the predicted<br />

value in combination with an FEV 1 /FVC < 70% confirms<br />

Figure 5-1-5. Normal Spirogram and Spirogram<br />

Typical of Patients with Moderate COPD<br />

FEV 1<br />

FVC<br />

FEV 1 /FVC<br />

the presence of airflow limitation that is not fully<br />

reversible. The FEV 1 /FVC on its own is a more sensitive<br />

measure of airflow limitation, and an FEV 1 /FVC < 70% is<br />

considered an early sign of airflow limitation in patients<br />

whose FEV 1 remains normal (≥ 80% predicted). This<br />

approach to defining airflow limitation is a pragmatic one<br />

in view of the fact that universally applicable reference<br />

values <strong>for</strong> FEV 1 and FVC are not available.<br />

Peak expiratory flow (PEF) is sometimes used as a measure<br />

of airflow limitation, but in COPD the relationship between<br />

PEF and FEV 1 is poor. PEF may underestimate the<br />

degree of airways obstruction in these patients 15 . If<br />

spirometry is unavailable, prolongation of the <strong>for</strong>ced<br />

expiratory time beyond 6 seconds is a crude, but useful,<br />

guide to the presence of an FEV 1 /FVC ratio < 50% 16,17 .<br />

The role of screening spirometry in the general population<br />

or in a population at risk <strong>for</strong> COPD is controversial. Both<br />

FEV 1 and FVC predict all-cause mortality independent of<br />

tobacco smoking, and abnormal lung function identifies a<br />

subgroup of smokers at increased risk <strong>for</strong> lung cancer.<br />

This has been the basis of an argument that screening<br />

spirometry should be employed as a global health<br />

assessment tool 18 . However, there are no data to indicate<br />

that screening spirometry is effective in directing<br />

management decisions or in improving COPD outcomes.<br />

Assessment of Severity<br />

Assessment of COPD severity is based on the patient’s<br />

level of symptoms, the severity of the spirometric<br />

abnormality, and the presence of complications such as<br />

respiratory failure and right heart failure (Figure 1-2).<br />

The use of specific spirometric cut-points (e.g.,<br />

FEV 1 /FVC


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 52<br />

that should be targeted <strong>for</strong> preventive intervention. Much<br />

depends on the success of convincing such people, as<br />

well as health care workers, that minor respiratory<br />

symptoms may be markers of future ill health.<br />

The severity of a patient’s breathlessness is important<br />

and can be gauged by the MRC scale (Figure 5-1-3).<br />

Arterial blood gases should be measured in all patients<br />

who have FEV 1 < 40% predicted or clinical signs of<br />

respiratory failure or right heart failure.<br />

Additional Investigations<br />

For patients diagnosed with Stage II: Moderate COPD<br />

and beyond, the following additional investigations may<br />

be useful.<br />

Bronchodilator reversibility testing. Generally per<strong>for</strong>med<br />

only once, at the time of diagnosis, this test is useful <strong>for</strong><br />

several reasons:<br />

• To help rule out a diagnosis of asthma.<br />

If FEV 1 returns to the predicted normal range after<br />

administration of a bronchodilator, the patient’s airflow<br />

limitation is likely due to asthma.<br />

• To establish a patient’s best attainable lung function at<br />

that point in time.<br />

• To gauge a patient’s prognosis.<br />

Some studies show that the postbronchodilator FEV 1 is<br />

a more reliable prognostic marker than pre-bronchodilator<br />

FEV 1<br />

22<br />

. In addition, the Intermittent Positive Pressure<br />

Breathing (IPPB) Study, a multicenter clinical trial,<br />

suggested that the degree of bronchodilator response<br />

is inversely related to the rate of FEV 1 decline in<br />

COPD patients 23 .<br />

• To assess potential response to treatment.<br />

Patients who show significant improvement in FEV 1<br />

after administration of a bronchodilator are more likely<br />

to benefit from treatment with bronchodilators and have<br />

a positive response to glucocorticosteroids. However,<br />

individual responses to bronchodilator tests are influenced<br />

by many factors, and failure of FEV 1 to change by an<br />

arbitrary amount on one day does not preclude a<br />

response on another. Moreover, even patients who do<br />

not show a significant FEV 1 response to a short-acting<br />

bronchodilator test may benefit symptomatically from<br />

long-term bronchodilator therapy.<br />

Figure 5-1-6. Bronchodilator Reversibility Testing<br />

Preparation<br />

• Tests should be per<strong>for</strong>med when patients are clinically<br />

stable and free from respiratory infection.<br />

• Patients should not have taken inhaled short-acting<br />

bronchodilators in the previous six hours, long-acting<br />

ß 2 agonists in the previous 12 hours, or sustainedrelease<br />

theophyllines in the previous 24 hours.<br />

Spirometry<br />

• FEV 1 should be measured be<strong>for</strong>e a bronchodilator is<br />

given.<br />

• The bronchodilator should be given by metered dose<br />

inhaler through a spacer device or by nebulizer to be<br />

certain it has been inhaled.<br />

• The bronchodilator dose should be selected to be<br />

high on the dose/response curve.<br />

• Suitable dosage protocols are 400 g ß 2 -agonist,<br />

80 g anticholinergic, or the two combined. FEV 1<br />

should be measured again 30-45 minutes after the<br />

bronchodilator is given.<br />

Results<br />

• An increase in FEV 1 that is both greater than 200 ml<br />

and 12% above the pre-bronchodilator FEV 1 is considered<br />

significant.<br />

Between-day reproducibility of spirometry in the same<br />

individual is approximately 178 ml 24 . Thus, an acute<br />

change that exceeds both 200 ml and 12% of the baseline<br />

measurement is unlikely to have arisen by chance.<br />

A protocol <strong>for</strong> bronchodilator reversibility testing is listed<br />

in Figure 5-1-6.<br />

Chest X-ray. A chest X-ray is seldom diagnostic in<br />

COPD unless obvious bullous disease is present, but it is<br />

valuable in excluding alternative diagnoses. Radiological<br />

changes associated with COPD include signs of hyperinflation<br />

(flattened diaphragm on the lateral chest film, and<br />

an increase in the volume of the retrosternal air space),<br />

52 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 53<br />

Diagnosis<br />

COPD<br />

Figure 5-1-7. Differential Diagnosis of COPD<br />

Asthma<br />

Congestive<br />

Heart Failure<br />

Bronchiectasis<br />

Tuberculosis<br />

Obliterative<br />

Bronchiolitis<br />

Diffuse<br />

Panbronchiolitis<br />

Suggestive Features*<br />

Onset in mid-life.<br />

Symptoms slowly progressive.<br />

Long smoking history.<br />

Dyspnea during exercise.<br />

Largely irreversible airflow limitation.<br />

Onset early in life (often childhood).<br />

Symptoms vary from day to day.<br />

Symptoms at night/early morning.<br />

Allergy, rhinitis, and/or eczema also<br />

present.<br />

Family history of asthma.<br />

Largely reversible airflow limitation.<br />

Fine basilar crackles on auscultation.<br />

Chest X-ray shows dilated heart,<br />

pulmonary edema.<br />

Pulmonary function tests indicate<br />

volume restriction, not airflow limitation.<br />

Large volumes of purulent sputum.<br />

Commonly associated with bacterial<br />

infection.<br />

Coarse crackles/clubbing on<br />

auscultation.<br />

Chest X-ray/CT shows bronchial<br />

dilation, bronchial wall thickening.<br />

Onset all ages.<br />

Chest X-ray shows lung infiltrate.<br />

Microbiological confirmation.<br />

High local prevalence of tuberculosis.<br />

Onset in younger age, nonsmokers.<br />

May have history of rheumatoid<br />

arthritis or fume exposure.<br />

CT on expiration shows hypodense<br />

areas.<br />

Most patients are male and nonsmokers.<br />

Almost all have chronic sinusitis.<br />

Chest X-ray and HRCT show diffuse<br />

small centrilobular nodular opacities<br />

and hyperinflation.<br />

*These features tend to be characteristic of the respective diseases,<br />

but do not occur in every case. For example, a person who has<br />

never smoked may develop COPD (especially in the developing<br />

world, where other risk factors may be more important than cigarette<br />

smoking); asthma may develop in adult and even elderly patients.<br />

hyperlucency of the lungs, and rapid tapering of the vascular<br />

markings. Computed tomography (CT) of the chest is not<br />

routinely recommended. However, when there is doubt<br />

about the diagnosis of COPD, high resolution CT (HRCT)<br />

might help in the differential diagnosis. In addition, if a<br />

surgical procedure such as bullectomy or lung volume<br />

reduction is contemplated, chest CT is helpful.<br />

Arterial blood gas measurement. In advanced COPD<br />

measurement of arterial blood gases is important. This<br />

test should be per<strong>for</strong>med in patients with FEV 1 < 40%<br />

predicted or with clinical signs suggestive of respiratory<br />

failure or right heart failure.<br />

Alpha-1 antitrypsin deficiency screening. In patients<br />

who develop COPD at a young age (< 45 years) or who<br />

have a strong family history of the disease, it may be<br />

valuable to identify coexisting alpha-1 antitrypsin deficiency.<br />

This could lead to family screening or appropriate<br />

counseling. A serum concentration of alpha-1 antitrypsin<br />

below 15-20 % of the normal value is highly suggestive<br />

of homozygous alpha-1 antitrypsin deficiency.<br />

Differential Diagnosis<br />

A major differential diagnosis is asthma. In some patients<br />

with chronic asthma, a clear distinction from COPD is not<br />

possible using current imaging and physiological testing<br />

techniques, and it is assumed that asthma and COPD<br />

coexist in these patients. In these cases, current<br />

management is similar to that of asthma. Other potential<br />

diagnoses are usually easier to distinguish from COPD<br />

(Figure 5-1-7).<br />

ONGOING MONITORING<br />

AND ASSESSMENT<br />

Visits to health care facilities will increase in frequency<br />

as COPD progresses. The type of health care workers<br />

seen, and the frequency of visits, will depend on the<br />

health care system. Ongoing monitoring and assessment<br />

in COPD ensures that the goals of treatment are being<br />

met and should include evaluation of: (1) exposure to<br />

risk factors, especially tobacco smoke; (2) disease<br />

progression and development of complications;<br />

(3) pharmacotherapy and other medical treatment;<br />

(4) exacerbation history; (5) comorbidities.<br />

Suggested questions <strong>for</strong> follow-up visits are listed in<br />

Figure 5-1-8. The best way to detect changes in<br />

symptoms and overall health status is to ask the same<br />

questions at each visit.<br />

MANAGEMENT OF COPD 53


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 54<br />

Monitor <strong>Disease</strong> Progression and<br />

Development of Complications<br />

COPD is usually a progressive disease. <strong>Lung</strong> function<br />

can be expected to worsen over time, even with the best<br />

available care. Symptoms and objective measures of<br />

airflow limitation should be monitored to determine when<br />

to modify therapy and to identify any complications that<br />

may develop. As at the initial assessment, follow-up<br />

visits should include a physical examination and<br />

discussion of symptoms, particularly any new or<br />

worsening symptoms.<br />

Pulmonary function. A patient’s decline in lung function<br />

is best tracked by periodic spirometry measurements.<br />

Useful in<strong>for</strong>mation about lung function decline is unlikely<br />

from spirometry measurements per<strong>for</strong>med more than<br />

once a year. Spirometry should be per<strong>for</strong>med if there is<br />

a substantial increase in symptoms or a complication.<br />

Other pulmonary function tests, such as flow-volume<br />

loops, diffusing capacity (D LCO ) measurements, and<br />

measurement of lung volumes are not needed in a<br />

routine assessment but can provide in<strong>for</strong>mation about the<br />

overall impact of the disease and can be valuable in<br />

resolving diagnostic uncertainties and assessing patients<br />

<strong>for</strong> surgery.<br />

Arterial blood gas measurement. Measurement of<br />

arterial blood gas tensions should be per<strong>for</strong>med in all<br />

patients with FEV 1 < 40% predicted or when clinical<br />

signs of respiratory failure or right heart failure are<br />

present. Respiratory failure is indicated by a<br />

PaO 2 < 8.0 kPa (60 mm Hg) with or without<br />

PaCO 2 > 6.7 kPa (50 mm Hg) in arterial blood gas<br />

measurements made while breathing air at sea level.<br />

Screening patients by pulse oximetry and assessing<br />

arterial blood gases in those with an oxygen saturation<br />

(SaO 2 ) < 92% may be a useful way of selecting patients<br />

<strong>for</strong> arterial blood gas measurement 27 . However, pulse<br />

oximetry gives no in<strong>for</strong>mation about CO 2 tensions.<br />

Several considerations are important to ensure accurate<br />

test results. Oxygen pressure in the inspired air (FiO 2 )<br />

should be measured, taking note if patient is using an<br />

O 2 -driven nebulizer. Changes in arterial blood gas tensions<br />

take time to occur, especially in severe disease.<br />

Thus, 20-30 minutes should pass be<strong>for</strong>e rechecking the<br />

gas tensions when the FiO 2 has been changed.<br />

Figure 5-1-8. Suggested Questions<br />

<strong>for</strong> Follow-Up Visits*<br />

Monitor exposure to risk factors:<br />

• Have you continued to stay off cigarettes?<br />

• If not, how many cigarettes per day are you smoking?<br />

• Would you like to quit smoking?<br />

• Has there been any change in your working<br />

environment?<br />

Monitor disease progression and development<br />

of complications:<br />

• How much can you do be<strong>for</strong>e you get short of breath?<br />

(Use an everyday example, such as walking up flights<br />

of stairs, up a hill, or on flat ground.)<br />

• Has your dyspnea worsened, improved, or stayed the<br />

same since your last visit?<br />

• Have you had to reduce your activities because of<br />

dyspnea or other symptoms?<br />

• Have any of your symptoms worsened since your<br />

last visit?<br />

• Have you experienced any new symptoms since your<br />

last visit?<br />

• Has your sleep been disrupted due to dyspnea or other<br />

symptoms?<br />

• Since your last visit, have you missed any work<br />

because of your symptoms?<br />

Monitor pharmacotherapy and other medical<br />

treatment:<br />

• What medications are you taking?<br />

• How often do you take each medication?<br />

• How much do you take each time?<br />

• Have you missed or stopped taking any regular doses<br />

of your medications <strong>for</strong> any reason?<br />

• Have you had trouble filling your prescriptions<br />

(e.g., <strong>for</strong> financial reasons, not on <strong>for</strong>mulary)?<br />

• Please show me how you use your inhaler.<br />

• Have you tried any other medicines or remedies?<br />

• Has your medication been effective in controlling<br />

your symptoms?<br />

• Has your medication caused you any problems?<br />

Monitor exacerbation history:<br />

• Since your last visit, have you had any episodes/times<br />

when your symptoms were a lot worse than usual?<br />

• If so, how long did the episode(s) last? What do you<br />

think caused the symptoms to get worse? What did you<br />

do to control the symptoms?<br />

*These questions are examples and do not represent a standardized<br />

assessment instrument. The validity and reliability of these questions<br />

have not been assessed.<br />

54 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 55<br />

Adequate pressure must be applied at the puncture site<br />

<strong>for</strong> at least one minute; failure to do so can lead to painful<br />

bruising.<br />

Clinical signs of respiratory failure or right heart failure<br />

include central cyanosis, ankle swelling, and an increase<br />

in the jugular venous pressure. Clinical signs of hypercapnia<br />

are extremely nonspecific outside of exacerbations.<br />

Assessment of pulmonary hemodynamics. Pulmonary<br />

hypertension is only likely to be important in patients who<br />

have developed respiratory failure. Measurement of pulmonary<br />

arterial pressure is not recommended in clinical<br />

practice as it does not add practical in<strong>for</strong>mation beyond<br />

that obtained from a knowledge of PaO 2 .<br />

Diagnosis of right heart failure or cor pulmonale.<br />

Elevation of the jugular venous pressure and the presence<br />

of pitting ankle edema are often the most useful<br />

findings suggestive of cor pulmonale in clinical practice.<br />

However, the jugular venous pressure is often difficult to<br />

assess in patients with COPD, due to large swings in<br />

intrathoracic pressure. Firm diagnosis of cor pulmonale<br />

can be made through a number of investigations, including<br />

radiography, electrocardiography, echocardiography,<br />

radionucleotide scintigraphy, and magnetic resonance<br />

imaging. However, all of these measures involve inherent<br />

inaccuracies of diagnosis.<br />

CT and ventilation-perfusion scanning. Despite the<br />

benefits of being able to delineate pathological anatomy,<br />

routine CT and ventilation-perfusion scanning are currently<br />

confined to the assessment of COPD patients <strong>for</strong> surgery.<br />

HRCT is currently under investigation as a way of visualizing<br />

airway and parenchymal pathology more precisely.<br />

Hematocrit. Polycythemia can develop in the presence<br />

of arterial hypoxemia, especially in continuing smokers 28 .<br />

Polycythemia can be identified by hematocrit > 55%.<br />

Respiratory muscle function. Respiratory muscle function<br />

is usually measured by recording the maximum inspiratory<br />

and expiratory mouth pressures. More complex<br />

measurements are confined to research laboratories.<br />

Measurement of expiratory muscle <strong>for</strong>ce is useful in<br />

assessing patients when dyspnea or hypercapnia is not<br />

readily explained by lung function testing or when peripheral<br />

muscle weakness is suspected. This measurement<br />

may improve in COPD patients when other measurements<br />

of lung mechanics do not (e.g., after pulmonary<br />

rehabilitation) 29,30 .<br />

Sleep studies. Sleep studies may be indicated when<br />

hypoxemia or right heart failure develops in the presence<br />

of relatively mild airflow limitation or when the patient has<br />

symptoms suggesting the presence of sleep apnea.<br />

Exercise testing. Several types of tests are available to<br />

measure exercise capacity, but these are primarily used<br />

in conjunction with pulmonary rehabilitation programs.<br />

Monitor Pharmacotherapy and Other<br />

Medical Treatment<br />

In order to adjust therapy appropriately as the disease<br />

progresses, each follow-up visit should include a discussion<br />

of the current therapeutic regimen. Dosages of various<br />

medications, adherence to the regimen, inhaler technique,<br />

effectiveness of the current regime at controlling symptoms,<br />

and side effects of treatment should be monitored.<br />

Monitor Exacerbation History<br />

During periodic assessments, health care workers should<br />

question the patient and evaluate any records of exacerbations,<br />

both self-treated and those treated by other<br />

health care providers. Frequency, severity, and likely<br />

causes of exacerbations should be evaluated. Increased<br />

sputum volume, acutely worsening dyspnea, and the<br />

presence of purulent sputum should be noted. Specific<br />

inquiry into unscheduled visits to providers, telephone<br />

calls <strong>for</strong> assistance, and use of urgent or emergency care<br />

facilities may be helpful. Severity can be estimated by<br />

the increased need <strong>for</strong> bronchodilator medication or glucocorticosteroids<br />

and by the need <strong>for</strong> antibiotic treatment.<br />

Hospitalizations should be documented, including the<br />

facility, duration of stay, and any use of critical care or<br />

intubation. The clinician then can request summaries of<br />

all care received to facilitate continuity of care.<br />

Monitor Comorbidities<br />

In treating patients with COPD, it is important to consider<br />

the presence of concomitant conditions such as bronchial<br />

carcinoma, tuberculosis, sleep apnea, and left heart failure.<br />

The appropriate diagnostic tools (chest radiograph,<br />

ECG, etc.) should be used whenever symptoms (e.g.,<br />

hemoptysis) suggest one of these conditions.<br />

MANAGEMENT OF COPD 55


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 56<br />

REFERENCES<br />

1. Georgopoulos D, Anthonisen NR. Symptoms and signs of<br />

COPD. In: Cherniack NS, ed. <strong>Chronic</strong> obstructive pulmonary<br />

disease. Toronto: WB Saunders; 1991. p. 357-63.<br />

2. Burrows B, Niden AH, Barclay WR, Kasik JE. <strong>Chronic</strong><br />

obstructive lung disease II. Relationships of clinical and<br />

physiological findings to the severity of airways obstruction.<br />

Am Rev Respir Dis 1965; 91:665-78.<br />

3. Medical Research Council. Definition and classification of<br />

chronic bronchitis <strong>for</strong> clinical and epidemiological purposes:<br />

a report to the Medical Research Council by their<br />

Committee on the Aetiology of <strong>Chronic</strong> Bronchitis. Lancet<br />

1965; 1:775-80.<br />

4. Simon PM, Schwartstein RM, Weiss JW, Fencl V,<br />

Teghtsoonian M, Weinberger SE. Distinguishable types of<br />

dyspnea in patients with shortness of breath. Am Rev<br />

Respir Dis 1990; 142:1009-14.<br />

5. Elliott MW, Adams L, Cockcroft A, MacRae KD, Murphy K,<br />

Guz A. The language of breathlessness. Use of verbal<br />

descriptors by patients with cardiopulmonary disease. Am<br />

Rev Respir Dis 1991; 144:826-32.<br />

6. Bestall JC, Paul EA, Garrod R, Garnham R, Jones PW,<br />

Wedzicha JA. Usefulness of the Medical Research<br />

Council (MRC) dyspnoea scale as a measure of disability<br />

in patients with chronic obstructive pulmonary disease.<br />

Thorax 1999; 54:581-6.<br />

7. Celli BR, Rassulo J, Make BJ. Dyssynchronous breathing<br />

during arm but not leg exercise in patients with chronic airflow<br />

obstruction. N Engl J Med 1986; 314:1485-90.<br />

8. Schols AM, Soeters PB, Dingemans AM, Mostert R,<br />

Frantzen PJ, Wouters EF. Prevalence and characteristics of<br />

nutritional depletion in patients with stable COPD eligible<br />

<strong>for</strong> pulmonary rehabilitation. Am Rev Respir Dis 1993;<br />

147:1151-6.<br />

9. Johnston RN, Lockhart W, Ritchie RT, Smith DH.<br />

Haemoptysis. BMJ 1960; 1:592-5.<br />

10. Calverley PM. Neuropsychological deficits in chronic<br />

obstructive pulmonary disease. Monaldi Archives <strong>for</strong> Chest<br />

<strong>Disease</strong> 1996; 51:5-6.<br />

11. Kesten S, Chapman KR. Physician perceptions and management<br />

of COPD. Chest 1993; 104:254-8.<br />

12. Loveridge B, West P, Kryger MH, Anthonisen NR. Alteration<br />

of breathing pattern with progression of chronic obstructive<br />

pulmonary disease. Am Rev Respir Dis 1986; 134:930-4.<br />

13. Badgett RC, Tanaka DV, Hunt DK, Jelley MJ, Feinberg LE,<br />

Steiner JF, et al. Can moderate chronic obstructive pulmonary<br />

disease be diagnosed by history and physical findings<br />

alone? Am J Med 1993; 94:188-96.<br />

14. Standardization of spirometry, 1994 update. Am J Respir<br />

Crit Care Med 1995; 152:1107-36.<br />

15. Kelly CA, Gibson GJ. Relation between FEV 1 and peak<br />

expiratory flow in patients with chronic obstructive pulmonary<br />

disease. Thorax 1988; 43:335-6.<br />

16. Lal S, Ferguson AD, Campbell EJM. Forced expiratory time;<br />

a simple test <strong>for</strong> airways obstruction. BMJ 1964; 1:814-7.<br />

17. Swanney MP, Jensen RL, Crichton DA, Beckert LE, Cardno<br />

LA, Crapo RO. FEV(6) is an acceptable surrogate <strong>for</strong> FVC<br />

in the spirometric diagnosis of airway obstruction and<br />

restriction. Am J Respir Crit Care Med 2000; 162:917-9.<br />

18. Ferguson GT, Enright PL, Buist AS, Higgins MW. Office<br />

spirometry <strong>for</strong> lung health assessment in adults: a consensus<br />

statement from the national lung health education program.<br />

Chest 2000; 117:1146-61.<br />

19. Kanner RE, Connett JE, Williams DE, Buist AS. Effects of<br />

randomized assignment to a smoking cessation intervention<br />

and changes in smoking habits on respiratory symptoms<br />

in smokers with early chronic obstructive pulmonary<br />

disease: the <strong>Lung</strong> Health Study. Am J Med 1999; 106:410-6.<br />

20. Lofdahl CG, Postma DS, Laitinen LA, Ohlsson SV, Pauwels<br />

RA, Pride NB. The European Respiratory Society study on<br />

chronic obstructive pulmonary disease (EUROSCOP):<br />

recruitment methods and strategies. Respir Med 1998;<br />

92:467-72.<br />

21. Peto R, Speizer FE, Cochrane AL, Moore F, Fletcher CM,<br />

Tinker CM, et al. The relevance in adults of airflow obstruction,<br />

but not of mucus hypersecretion, to mortality from<br />

chronic lung disease: results from twenty years of prospective<br />

observation. Am Rev Respir Dis 1983; 128:491-500.<br />

22. Hansen EF, Phanareth K, Laursen LC, KokJensen A,<br />

Dirksen A. Reversible and irreversible airflow obstruction<br />

as predictor of overall mortality in asthma and chronic<br />

obstructive pulmonary disease. Am J Respir Crit Care<br />

Med 1999; 159:1267-71.<br />

23. Anthonisen NR, Wright EC. Bronchodilator response in<br />

chronic obstructive pulmonary disease. Am Rev Respir<br />

Dis 1986; 133:814-9.<br />

24. Sourk RL, Nugent KM. Bronchodilator testing: confidence<br />

intervals derived from placebo inhalations. Am Rev Respir<br />

Dis 1983; 128:153-7.<br />

25. Reis AL. Response to bronchodilators. In: Clausen J, ed.<br />

Pulmonary function testing: guidelines and controversies.<br />

New York: Academic Press; 1982. p. 215-221.<br />

26. American Thoracic Society. <strong>Lung</strong> function testing: selection<br />

of reference values and interpretative strategies. Am Rev<br />

Respir Dis 1991; 144:1202-18.<br />

27. Roberts CM, Bugler JR, Melchor R, Hetzel ML, Spiro SG.<br />

Value of pulse oximetry <strong>for</strong> long-term oxygen therapy<br />

requirement. Eur Respir J 1993; 6:559-62.<br />

28. Calverley PM, Leggett RJ, McElderry L, Flenley DC.<br />

Cigarette smoking and secondary polycythemia in hypoxic<br />

cor pulmonale. Am Rev Respir Dis 1982; 125:507-10.<br />

56 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 57<br />

29. Dekhuijzen PNR, Folgering HT, van Herwaarden CLA.<br />

Target-flow inspiratory muscle training during pulmonary<br />

rehabilitation in patients with COPD. Chest 1991; 99:128-33.<br />

30. Heijdra YF, Dekhuijzen PN, van Herwaarden CLA,<br />

Forlgering H. Nocturnal saturation improves by target-flow<br />

inspiratory muscle training in patients with COPD. Am J<br />

Respir Crit Care Med 1996; 153:260-5.<br />

MANAGEMENT OF COPD 57


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 58<br />

COMPONENT 2: REDUCE RISK FACTORS<br />

KEY POINTS:<br />

• Reduction of total personal exposure to tobacco<br />

smoke, occupational dusts and chemicals, and<br />

indoor and outdoor air pollutants are important<br />

goals to prevent the onset and progression of<br />

COPD.<br />

• Smoking cessation is the single most effective -<br />

and cost effective - way in most people to reduce<br />

the risk of developing COPD and stop its progression<br />

(Evidence A).<br />

• Brief tobacco dependence counseling is effective<br />

(Evidence A) and every tobacco user should be<br />

offered at least this treatment at every visit to a<br />

health care provider.<br />

• Three types of counseling are especially effective:<br />

practical counseling, social support as part of<br />

treatment, and social support arranged outside of<br />

treatment (Evidence A).<br />

• Several effective pharmacotherapies <strong>for</strong> tobacco<br />

dependence are available (Evidence A), and at<br />

least one of these medications should be added<br />

to counseling if necessary and in the absence of<br />

contraindications (Evidence A).<br />

• Progression of many occupationally induced<br />

respiratory disorders can be reduced or controlled<br />

through a variety of strategies aimed at reducing<br />

the burden of inhaled particles and gases<br />

(Evidence B).<br />

INTRODUCTION<br />

Identification, reduction, and control of risk factors are<br />

important steps toward prevention and treatment of any<br />

disease. In the case of COPD, these factors include<br />

tobacco smoke, occupational exposures, and indoor and<br />

outdoor air pollution and irritants.<br />

Since cigarette smoking is the major risk factor <strong>for</strong> COPD<br />

worldwide, smoking prevention programs should be<br />

implemented and smoking cessation programs should<br />

be readily available and encouraged <strong>for</strong> all individuals<br />

who smoke. Reduction of total personal exposure to<br />

occupational dust, fumes, and gases and to indoor and<br />

outdoor air pollutants is also an important goal to prevent<br />

the onset and progression of COPD 1 .<br />

TOBACCO SMOKE<br />

Smoking Prevention<br />

Comprehensive tobacco control policies and programs<br />

with clear, consistent, and repeated nonsmoking<br />

messages should be delivered through every feasible<br />

channel, including health care providers, schools, and<br />

radio, television, and print media. National and local<br />

campaigns should be undertaken to reduce exposure to<br />

tobacco smoke in public <strong>for</strong>ums. Legislation to establish<br />

smoke-free schools, public facilities, and work environments<br />

should be encouraged by government officials, public<br />

health workers, and the public. Smoking prevention<br />

programs should target all ages, including young children,<br />

adolescents, young adults, and pregnant women.<br />

Physicians and public health officials should encourage<br />

smoke-free homes.<br />

The first exposure to cigarette smoke may begin in utero<br />

when the fetus is exposed to blood-borne metabolites<br />

from the mother 2,3 . Neonates and infants may be<br />

exposed passively to tobacco smoke in the home if a<br />

family member smokes. Children less than 2 years old<br />

who are passively exposed to cigarette smoke have an<br />

increased prevalence of respiratory infections, and are at<br />

a greater risk of developing chronic respiratory symptoms<br />

later in life 4 .<br />

Smoking Cessation<br />

Smoking cessation is the single most effective - and cost<br />

effective - way to reduce exposure to COPD risk factors.<br />

Quitting smoking can prevent or delay the development of<br />

airflow limitation or reduce its progression 5 . A statement<br />

by the WHO (Figure 5-2-1) 6 emphasizes the health and<br />

economic benefits to be gained from smoking cessation.<br />

All smokers - including those who may be at risk <strong>for</strong><br />

COPD as well as those who already have the disease -<br />

should be offered the most intensive smoking cessation<br />

intervention feasible.<br />

58 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 59<br />

Figure 5-2-1. World Health Organization Statement<br />

on Smoking Cessation 6<br />

Smoking cessation is a critical step toward substantially<br />

reducing the health risks run by current smokers,<br />

thereby improving world health. Tobacco has been<br />

shown to cause about 25 life-threatening diseases,<br />

or groups of diseases, many of which can be prevented,<br />

delayed, or mitigated by smoking cessation. As life<br />

expectancy increases in developing countries, the<br />

morbidity and mortality burden of chronic diseases<br />

will increase still further. This projected concentration<br />

of tobacco-related disease burden can be lightened<br />

by intensive ef<strong>for</strong>ts at smoking cessation. Studies<br />

have shown that 75-80% of smokers want to quit,<br />

while one-third have made at least three serious<br />

cessation attempts. Cessation ef<strong>for</strong>ts cannot be<br />

ignored in favor of primary prevention; rather, both<br />

ef<strong>for</strong>ts must be made in conjunction with one another.<br />

If only small portions of today's 1.1 billion smokers<br />

were able to stop, the long-term health and economic<br />

benefits would be immense. Governments,<br />

communities, organizations, schools, families and<br />

individuals are called upon to help current smokers<br />

stop their addictive and damaging habit.<br />

Smoking cessation interventions are effective in both<br />

genders, in all racial and ethnic groups, and in pregnant<br />

women. Age influences quit rates, with young people<br />

less likely to quit, but nevertheless smoking cessation<br />

programs can be effective in all age groups.<br />

International data on the economic impact of smoking<br />

cessation are strikingly consistent: investing resources in<br />

smoking cessation programs is cost effective in terms of<br />

medical costs per life year gained. Interventions that<br />

have been investigated include nicotine replacement with<br />

transdermal patch, counseling from physicians and other<br />

health professionals (with and without nicotine patch),<br />

self-help and group programs, and community-based<br />

stop-smoking contests. A review of data from a number<br />

of countries estimated the median societal cost of various<br />

smoking cessation interventions at $990 to $13,000 (US)<br />

per life year gained 7 . Smoking cessation programs are a<br />

particularly good value <strong>for</strong> the UK National Health<br />

Service, with costs from £212 to £873 (US $320 to<br />

$1,400) per life year gained 8 .<br />

The role of health care providers in smoking cessation.<br />

A successful smoking cessation strategy requires a<br />

multifaceted approach, including public policy, in<strong>for</strong>mation<br />

dissemination programs, and health education through<br />

the media and schools 9 . However, health care providers,<br />

including physicians, nurses, dentists, psychologists,<br />

pharmacists, and others, are key to the delivery of smoking<br />

cessation messages and interventions. Involving as<br />

many of these individuals as possible will help. Health<br />

care workers should encourage all patients who smoke<br />

to quit, even those patients who come to the health care<br />

provider <strong>for</strong> unrelated reasons and do not have symptoms<br />

of COPD or evidence of airflow limitation.<br />

Guidelines <strong>for</strong> smoking cessation entitled Treating<br />

Tobacco Use and Dependence: A Clinical Practice<br />

Guideline were published by the US Public Health<br />

Service 10 . The major conclusions are summarized in<br />

Figure 5-2-2.<br />

Figure 5-2-2. Public Health Service Report: Treating<br />

Tobacco Use and Dependence: A Clinical Practice<br />

Guideline - Major Findings and Recommendations 10<br />

1. Tobacco dependence is a chronic condition that<br />

warrants repeated treatment until long-term or<br />

permanent abstinence is achieved.<br />

2. Effective treatments <strong>for</strong> tobacco dependence exist<br />

and all tobacco users should be offered these<br />

treatments.<br />

3. Clinicians and health care delivery systems<br />

must institutionalize the consistent identification,<br />

documentation and treatment of every tobacco<br />

user at every visit.<br />

4. Brief tobacco dependence treatment is effective<br />

and every tobacco user should be offered at least<br />

brief treatment.<br />

5. There is a strong dose-response relation between the<br />

intensity of tobacco dependence counseling and its<br />

effectiveness.<br />

6. Three types of counseling were found to be especially<br />

effective: practical counseling, social support as part<br />

of treatment, and social support arranged outside of<br />

treatment.<br />

7. Five first-line pharmacotherapies <strong>for</strong> tobacco dependence<br />

- bupropion SR, nicotine gum, nicotine inhaler, nicotine<br />

nasal spray, and nicotine patch - are effective and at<br />

least one of these medications should be prescribed<br />

in the absence of contraindications.<br />

8. Tobacco dependence treatments are cost effective<br />

relative to other medical and disease prevention<br />

interventions.<br />

MANAGEMENT OF COPD 59


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 60<br />

Figure 5-2-3. Brief Strategies to Help the<br />

Patient Willing to Quit 10-13<br />

1. ASK: Systematically identify all tobacco users at<br />

every visit.<br />

Implement an office-wide system that ensures that,<br />

<strong>for</strong> EVERY patient at EVERY clinic visit, tobacco-use<br />

status is queried and documented.<br />

The Public Health Service Guidelines recommend a<br />

five-step program <strong>for</strong> intervention (Figure 5-2-3), which<br />

provides a strategic framework helpful to health care<br />

providers interested in helping their patients stop<br />

smoking 10-13 . The guidelines emphasize that tobacco<br />

dependence is a chronic disease (Figure 5-2-4) 10 and<br />

urge clinicians to recognize that relapse is common and<br />

reflects the chronic nature of dependence, not failure on<br />

the part of the clinician or the patient.<br />

2. ADVISE: Strongly urge all tobacco users to quit.<br />

In a clear, strong, and personalized manner, urge<br />

every tobacco user to quit.<br />

Figure 5-2-5.<br />

Stages of Change Model<br />

3. ASSESS: Determine willingness to make a quit<br />

Precontemplation<br />

attempt.<br />

Ask every tobacco user if he or she is willing to make<br />

a quit attempt at this time (e.g., within the next 30 days).<br />

4. ASSIST: Aid the patient in quitting.<br />

Help the patient with a quit plan; provide practical<br />

counseling; provide intra-treatment social support; help the<br />

Relapse<br />

Slipping Back<br />

Contemplation<br />

Recycling<br />

patient obtain extra-treatment social support; recommend<br />

Short-Term<br />

use of approved pharmacotherapy except in special<br />

Maintenance<br />

circumstances; provide supplementary materials.<br />

Sustained<br />

5. ARRANGE: Schedule follow-up contact.<br />

Maintenance<br />

Schedule follow-up contact, either in person or via<br />

telephone.<br />

Printed with permission of Dr. Peter M.A. Calverley.<br />

Figure 5-2-4. Tobacco Dependence<br />

• For most people, tobacco dependence results in<br />

true drug dependence comparable to dependence<br />

caused by opiates, amphetamines, and cocaine.<br />

• Tobacco dependence is almost always a chronic<br />

disorder that warrants long-term clinical intervention<br />

as do other addictive disorders. Failure to appreciate<br />

the chronic nature of tobacco dependence may<br />

impair the clinician's motivation to treat tobacco use<br />

consistently in a long-term fashion.<br />

• Clinicians must understand that this is a chronic<br />

condition comparable to diabetes, hypertension,<br />

or hyperlipidemia requiring simple counseling advice,<br />

support, and appropriate pharmacotherapy.<br />

• Relapse is common, which is the nature of dependence<br />

and not the failure of the clinician or the patient.<br />

Preparation<br />

Action<br />

Most individuals go through several stages be<strong>for</strong>e they<br />

stop smoking (Figure 5-2-5) 9 . It is often helpful <strong>for</strong> the<br />

clinician to assess a patient's readiness to quit in order to<br />

determine the most effective course of action at that time.<br />

The clinician should initiate treatment if the patient is<br />

ready to quit. For a patient not ready to make a quit<br />

attempt, the clinician should provide a brief intervention<br />

designed to promote the motivation to quit.<br />

Counseling. Counseling delivered by physicians and<br />

other health professionals significantly increases quit<br />

rates over self-initiated strategies 14 . Even a brief<br />

(3-minute) period of counseling to urge a smoker to quit<br />

results in smoking cessation rates of 5-10% 15 . At the very<br />

least, this should be done <strong>for</strong> every smoker at every<br />

health care provider visit 15,16 .<br />

However, there is a strong dose-response relationship<br />

between counseling intensity and cessation success 17,18 .<br />

Ways to make the treatment more intense include<br />

increasing the length of the treatment session, the<br />

number of treatment sessions, and the number of weeks<br />

over which the treatment is delivered. Counseling<br />

60 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 61<br />

sessions of 3 to 10 minutes result in cessation rates of<br />

around 12% 10 . With more complex interventions (<strong>for</strong><br />

example, controlled clinical trials that include skills training,<br />

problem solving, and psychosocial support), quit rates can<br />

reach 20-30% 17 . In one multicenter controlled clinical trial,<br />

a combination of physician advice, group support, skills<br />

training, and nicotine replacement therapy achieved a<br />

quit rate of 35% at one year and a sustained quit rate of<br />

22% at 5 years 5 .<br />

Both individual and group counseling are effective <strong>for</strong>mats<br />

<strong>for</strong> smoking cessation. Several particular items of<br />

counseling content seem to be especially effective,<br />

including problem solving, general skills training, and<br />

provision of intra-treatment support. The important<br />

elements in the support aspect of successful treatment<br />

programs are shown in Figure 5-2-6 9,10 . The common<br />

subjects covered in successful problem solving/skills<br />

training programs include:<br />

• Recognition of danger signals likely to be associated<br />

with the risk of relapse, such as being around other<br />

smokers, being under time pressure, getting into an<br />

argument, drinking alcohol, and negative moods.<br />

• Enhancement of skills needed to handle these<br />

situations, such as learning to anticipate and avoid a<br />

particular stress.<br />

• Basic in<strong>for</strong>mation about smoking and successful<br />

quitting, such as the nature and time course of<br />

withdrawal, the addictive nature of smoking, and the<br />

fact that any return to smoking, including even a single<br />

puff, increases the likelihood of a relapse.<br />

Pharmacotherapy. Numerous effective pharmacotherapies<br />

<strong>for</strong> smoking cessation now exist 9-11 (Evidence A), and<br />

pharmacotherapy is recommended when counseling is not<br />

sufficient to help patients quit smoking. Special consideration<br />

should be given be<strong>for</strong>e using pharmacotherapy in selected<br />

populations: people with medical contraindications, light<br />

smokers (fewer than 10 cigarettes/day), and pregnant<br />

and adolescent smokers.<br />

Nicotine replacement products. Numerous studies indicate<br />

that nicotine replacement therapy in any <strong>for</strong>m (nicotine<br />

gum, inhaler, nasal spray, transdermal patch, sublingual<br />

tablet, or lozenge) reliably increases long-term smoking<br />

abstinence rates 10,19 . Nicotine replacement therapy is<br />

more effective when combined with counseling and<br />

behavior therapy 20 , although nicotine patch or nicotine<br />

gum consistently increases smoking cessation rates<br />

regardless of the level of additional behavioral or<br />

Figure 5-2-6. Patient Support in<br />

Smoking Cessation Programs 9,10<br />

• Encourage the patient in the quit attempt.<br />

Indicate that effective cessation treatments are now<br />

available and, in fact, half of all people who smoked<br />

have now quit. Communicate your confidence in the<br />

patient's ability to quit.<br />

• Communicate care and concern. Ask how the<br />

patient feels about smoking and whether he/she<br />

wants to quit, expressing concern along with the<br />

ability and willingness to help. Be open to the<br />

patient's fears of quitting.<br />

• Encourage the patient to talk about the quitting<br />

process. Talk to the patient about the reasons<br />

he/she wants to quit, difficulty encountered while<br />

quitting, success the patient has achieved, and concerns<br />

and worries about quitting.<br />

• Provide basic in<strong>for</strong>mation about smoking, the<br />

risks of continuing, the benefits of quitting, and<br />

the techniques that optimize success. Outline the<br />

nature, symptoms, and time course of withdrawal<br />

and techniques <strong>for</strong> dealing with withdrawal.<br />

psychosocial interventions. Medical contraindications to<br />

nicotine replacement therapy include unstable coronary<br />

artery disease, untreated peptic ulcer disease, and recent<br />

myocardial infarction or stroke 9 . Specific studies to date<br />

do not support the use of nicotine replacement therapy<br />

<strong>for</strong> longer than 8 weeks 21 , although some patients may<br />

require extended use to prevent relapse.<br />

All <strong>for</strong>ms of nicotine replacement therapy are significantly<br />

more effective than placebo. Every ef<strong>for</strong>t should be<br />

made to tailor the choice of replacement therapy to the<br />

individual's culture and lifestyle to improve adherence.<br />

The patch is generally favored over the gum because it<br />

requires less training <strong>for</strong> effective use and is associated<br />

with fewer compliance problems.<br />

No data are available to help clinicians tailor nicotine<br />

patch regimens to the intensity of cigarette smoking. In<br />

all cases it seems generally appropriate to start with the<br />

higher dose patch. For most patches, which come in<br />

three different doses, patients should use the highest<br />

dose <strong>for</strong> the first four weeks and drop to progressively<br />

lower doses over an eight-week period. Where only two<br />

doses are available, the higher dose should be used <strong>for</strong><br />

the first four weeks and the lower dose <strong>for</strong> the second<br />

four weeks.<br />

MANAGEMENT OF COPD 61


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 62<br />

When using nicotine gum, the patient needs to be advised<br />

that absorption occurs through the buccal mucosa. For<br />

this reason, the patient should be advised to chew the<br />

gum <strong>for</strong> a while and then put the gum against the inside<br />

of the cheek to allow absorption to occur and prolong<br />

the release of nicotine. Continuous chewing produces<br />

secretions that are swallowed, results in little absorption,<br />

and can cause nausea. Acidic beverages, particularly<br />

coffee, juices, and soft drinks, interfere with the absorption<br />

of nicotine. Thus, the patient needs to be advised that<br />

eating or drinking anything except water should be avoided<br />

<strong>for</strong> 15 minutes be<strong>for</strong>e and during chewing. Although<br />

nicotine gum is an effective smoking cessation treatment,<br />

problems with compliance, ease of use, social acceptability,<br />

risk of developing temporo-mandibular joint symptoms,<br />

and unpleasant taste have been noted. In highly dependent<br />

smokers, the 4 mg gum is more effective than the 2 mg<br />

gum 22 .<br />

Other pharmacotherapy. The antidepressants bupropion 23<br />

and nortriptyline have also been shown to increase longterm<br />

quit rates 9,19,24 . Although more studies need to be<br />

conducted with these medications, a randomized<br />

controlled trial with counseling and support showed quit<br />

rates at one year of 30% with sustained-release bupropion<br />

alone and 35% with sustained-release bupropion plus<br />

nicotine patch 23 . The effectiveness of the antihypertensive<br />

drug clonidine is limited by side effects 19 .<br />

OCCUPATIONAL EXPOSURES<br />

Although it is not known how many individuals are at risk<br />

of developing respiratory disease from occupational<br />

exposures in either developing or developed countries,<br />

many occupationally induced respiratory disorders can be<br />

reduced or controlled through a variety of strategies aimed<br />

at reducing the burden of inhaled particles and gases 25 :<br />

●<br />

●<br />

●<br />

Implement and en<strong>for</strong>ce strict, legally mandated control<br />

of airborne exposure in the workplace.<br />

Initiate intensive and continuing education of exposed<br />

workers, industrial managers, health care workers,<br />

primary care physicians, and legislators.<br />

Educate workers and policymakers on how cigarette<br />

smoking aggravates occupational lung diseases and<br />

why ef<strong>for</strong>ts to reduce smoking where a hazard exists<br />

are important.<br />

The main emphasis should be on primary prevention,<br />

which is best achieved by the elimination or reduction of<br />

exposures to various substances in the workplace.<br />

Secondary prevention, achieved through surveillance and<br />

early case detection, is also of great importance. Both<br />

approaches are necessary to improve the present situation<br />

and to reduce the burden of lung disease.<br />

INDOOR AND OUTDOOR<br />

AIR POLLUTION<br />

Individuals experience diverse indoor and outdoor<br />

environments throughout the day, each of which has its<br />

own unique set of air contaminants. Although outdoor<br />

and indoor air pollution are generally thought of separately,<br />

the concept of total personal exposure may be more<br />

relevant <strong>for</strong> COPD. Reducing the risk from indoor and<br />

outdoor air pollution requires a combination of public<br />

policy and protective steps taken by individual patients.<br />

Regulation of Air Quality<br />

At the national level, achieving a set level of air quality<br />

should be a high priority; this goal will normally require<br />

legislative action. Details on setting and maintaining air<br />

quality goals are beyond the scope of this document.<br />

Understanding health risks posed by local air pollution<br />

sources may be difficult and requires skills in community<br />

health, toxicology, and epidemiology. Local physicians may<br />

become involved through concerns about the health of their<br />

patients or as advocates <strong>for</strong> the community's environment.<br />

Patient-Oriented Control<br />

The health care provider should consider susceptibility<br />

(including family history and exposure to indoor/outdoor<br />

pollution) <strong>for</strong> each individual patient.<br />

●<br />

●<br />

●<br />

Patients should be counseled concerning the nature<br />

and degree of their susceptibility. Those who are at<br />

high risk should avoid vigorous exercise outdoors<br />

during pollution episodes.<br />

If various solid fuels are used <strong>for</strong> cooking and heating,<br />

adequate ventilation should be encouraged.<br />

Persons with severe COPD should monitor public<br />

announcements of air quality and should stay indoors<br />

when air quality is poor.<br />

62 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 63<br />

●<br />

●<br />

●<br />

The use of medication should follow the usual clinical<br />

indications; therapeutic regimes should not be adjusted<br />

because of the occurrence of a pollution episode<br />

without evidence of worsening of symptoms or function.<br />

Respiratory protective equipment has been developed<br />

<strong>for</strong> use in the workplace in order to minimize exposure<br />

to toxic gases and particles. However, under most<br />

circumstances, health care providers should not suggest<br />

respiratory protection as a method <strong>for</strong> reducing the<br />

risks of ambient air pollution.<br />

Air cleaners have not been shown to have health<br />

benefits, whether directed at pollutants generated by<br />

indoor sources or at those brought in with outdoor air.<br />

REFERENCES<br />

1. Samet J, Utell MJ. Ambient air pollution. In: Rosenstock L,<br />

Cullen M, eds. Textbook of occupational and environmental<br />

medicine. Philadelphia: WB Saunders; 1994. p. 53-60.<br />

2. Jeffery PK. Cigarette smoke-induced damage of airway<br />

mucosa. In: Chretien J, Dusser D, eds. Environmental<br />

impact on the airways: from injury to repair. <strong>Lung</strong> biology<br />

in health and disease. Vol. 93. New York: Marcel Dekker;<br />

1996. p. 299-354.<br />

3. Helms PJ. <strong>Lung</strong> growth: implications <strong>for</strong> the development of<br />

disease [editorial]. Thorax 1994; 49:440-1.<br />

4. Colley JR, Holland WW, Corkhill RT. Influence of passive<br />

smoking and parental phlegm on pneumonia and bronchitis<br />

in early childhood. Lancet 1974; 2:1031-4.<br />

5. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey<br />

WC, Buist AS, et al. Effects of smoking intervention and<br />

the use of an inhaled anticholinergic bronchodilator on the<br />

rate of decline of FEV 1 . The <strong>Lung</strong> Health Study. JAMA<br />

1994; 272:1497-505.<br />

6. World Health Organization. Tobacco free initiative:<br />

policies <strong>for</strong> public health. Geneva: World Health<br />

Organization; 1999. Available from: URL:<br />

www.who/int/toh/worldnottobacco99<br />

7. Tengs TO, Adams ME, Pliskin JS, Safran DG, Siegel JE,<br />

Weinstein MC, et al. Five-hundred life-saving interventions<br />

and their cost-effectiveness. Risk Anal 1995; 15:369-90.<br />

8. Parrott S, Godfrey C, Raw M, West R, McNeill A. Guidance<br />

<strong>for</strong> commissioners on the cost effectiveness of smoking<br />

cessation interventions. Health Educational Authority.<br />

Thorax 1998; 53 (Suppl 5 Pt 2): S1-38.<br />

9. Fiore MC, Bailey WC, Cohen SJ. Smoking cessation: in<strong>for</strong>mation<br />

<strong>for</strong> specialists. Rockville, MD: US Department of<br />

Health and Human Services, Public Health Service,<br />

Agency <strong>for</strong> Health Care Policy and Research and Centers<br />

<strong>for</strong> <strong>Disease</strong> Control and Prevention; 1996. AHCPR<br />

Publication No. 96-0694.<br />

10. The Tobacco Use and Dependence Clinical Practice<br />

Guideline Panel, Staff, and Consortium Representatives.<br />

A clinical practice guideline <strong>for</strong> treating tobacco use and<br />

dependence. JAMA 2000; 283:244-54.<br />

11. American Medical Association. Guidelines <strong>for</strong> the diagnosis<br />

and treatment of nicotine dependence: how to help<br />

patients stop smoking. Washington, DC: American Medical<br />

Association; 1994.<br />

12. Glynn TJ, Manley MW. How to help your patients stop<br />

smoking. A National Cancer Institute manual <strong>for</strong> physicians.<br />

Bethesda, MD: US Department of Health and<br />

Human Services, Public Health Service, National Institutes<br />

of Health, National Cancer Institute; 1990. NIH Publication<br />

No. 90-3064.<br />

13. Glynn TJ, Manley MW, Pechacek TF. Physician-initiated<br />

smoking cessation program: the National Cancer Institute<br />

trials. Prog Clin Biol Res 1990; 339:11-25.<br />

14. Baillie AJ, Mattick RP, Hall W, Webster P. Meta-analytic<br />

review of the efficacy of smoking cessation interventions.<br />

Drug and Alcohol Review 1994; 13:157-70.<br />

15. Wilson DH, Wakefield MA, Steven ID, Rohrsheim RA,<br />

Esterman AJ, Graham NM. "Sick of smoking": evaluation of<br />

a targeted minimal smoking cessation intervention in general<br />

practice. Med J Aust 1990; 152:518-21.<br />

16. Britton J, Knox A. Helping people to stop smoking: the new<br />

smoking cessation guidelines [editorial]. Thorax 1999;<br />

54:1-2.<br />

17. Kottke TE, Battista RN, DeFriese GH, Brekke ML.<br />

Attributes of successful smoking cessation interventions in<br />

medical practice. A meta-analysis of 39 controlled trials.<br />

JAMA 1988; 259:2883-9.<br />

18. Ockene JK, Kristeller J, Goldberg R, Amick TL, Pekow PS,<br />

Hosmer D, et al. Increasing the efficacy of physician-delivered<br />

smoking interventions: a randomized clinical trial. J<br />

Gen Intern Med 1991; 6:1-8.<br />

19. Lancaster T, Stead L, Silagy C, Sowden A. Effectiveness of<br />

interventions to help people stop smoking: findings from<br />

the Cochrane Library. BMJ 2000; 321:355-8.<br />

20. Schwartz JL. Review and evaluation of smoking cessation<br />

methods: United States and Canada, 1978-1985.<br />

Bethesda, MD: National Institutes of Health; 1987. NIH<br />

Publication No. 87-2940.<br />

21. Fiore MC, Smith SS, Jorenby DE, Baker TB. The effectiveness<br />

of the nicotine patch <strong>for</strong> smoking cessation. A metaanalysis.<br />

JAMA 1994; 271:1940-7.<br />

22. Sachs DP, Benowitz NL. Individualizing medical treatment<br />

<strong>for</strong> tobacco dependence [editorial; comment]. Eur Respir J<br />

1996; 9:629-31.<br />

23. Tashkin D, Kanner R, Bailey W, Buist S, Anderson P, Nides<br />

M, Gonzales D, Dozier G, Patel MK, Jamerson B. Smoking<br />

cessation in patients with chronic obstructive pulmonary<br />

disease: a double-blind, placebo-controlled, randomised<br />

trial. Lancet 2001: 19;357 (9268): 1571-5<br />

MANAGEMENT OF COPD 63


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 64<br />

24. Jorenby DE, Leischow SJ, Nides MA, Rennard SI,<br />

Johnston JA, Hughes AR, et al. A controlled trial of sustained-release<br />

bupropion, a nicotine patch, or both <strong>for</strong><br />

smoking cessation. N Engl J Med 1999; 340:685-91.<br />

25. The COPD Guidelines Group of the Standards of Care<br />

Committee of the BTS. BTS guidelines <strong>for</strong> the management<br />

of chronic obstructive pulmonary disease. Thorax<br />

1997; 52 Suppl 5:S1-28.<br />

64 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 65<br />

COMPONENT 3: MANAGE STABLE COPD<br />

KEY POINTS:<br />

• The overall approach to managing stable COPD<br />

should be characterized by a stepwise increase in<br />

treatment, depending on the severity of the disease.<br />

• For patients with COPD, health education can play<br />

a role in improving skills, ability to cope with illness,<br />

and health status. It is effective in accomplishing<br />

certain goals, including smoking cessation<br />

(Evidence A).<br />

• None of the existing medications <strong>for</strong> COPD have<br />

been shown to modify the long-term decline in lung<br />

function that is the hallmark of this disease<br />

(Evidence A). There<strong>for</strong>e, pharmacotherapy <strong>for</strong><br />

COPD is used to decrease symptoms and/or<br />

complications.<br />

• Bronchodilator medications are central to the<br />

symptomatic management of COPD (Evidence A).<br />

They are given on an as-needed basis or on a<br />

regular basis to prevent or reduce symptoms.<br />

• The principal bronchodilator treatments are<br />

ß 2 -agonists, anticholinergics, theophylline, and a<br />

combination of these drugs (Evidence A).<br />

• Regular treatment with long-acting bronchodilators<br />

is more effective and convenient than treatment with<br />

short-acting bronchodilators, but more expensive<br />

(Evidence A).<br />

• The addition of regular treatment with inhaled<br />

glucocorticosteroids to bronchodilator treatment is<br />

appropriate <strong>for</strong> symptomatic COPD patients with an<br />

FEV1 < 50% predicted (Stage III: Severe COPD<br />

and Stage IV: Very Severe COPD) and repeated<br />

exacerbations (Evidence A).<br />

• <strong>Chronic</strong> treatment with systemic glucocorticosteroids<br />

should be avoided because of an unfavorable<br />

benefit-to-risk ratio (Evidence A).<br />

• All COPD patients benefit from exercise training<br />

programs, improving with respect to both exercise<br />

tolerance and symptoms of dyspnea and fatigue<br />

(Evidence A).<br />

• The long-term administration of oxygen (> 15 hours<br />

per day) to patients with chronic respiratory failure<br />

has been shown to increase survival (Evidence A).<br />

INTRODUCTION<br />

The overall approach to managing stable COPD should<br />

be characterized by a stepwise increase in treatment,<br />

depending on the severity of the disease. The step-down<br />

approach used in the chronic treatment of asthma is not<br />

applicable to COPD since COPD is usually stable and<br />

very often progressive. Management of COPD involves<br />

several objectives (see Chapter 5, Introduction) that<br />

should be met with minimal side effects from treatment.<br />

It is based on an individualized assessment of disease<br />

severity (Figure 5-3-1) and response to various therapies.<br />

Figure 5-3-1. Factors Affecting the Severity of COPD<br />

• Severity of symptoms<br />

• Severity of airflow limitation<br />

• Frequency and severity of exacerbations<br />

• Presence of one or more complications<br />

• Presence of respiratory failure<br />

• Presence of comorbid conditions<br />

• General health status<br />

• Number of medications needed to manage the disease<br />

The classification of severity (Figure 1-2) of stable COPD<br />

incorporates an individualized assessment of disease<br />

severity and therapeutic response into the management<br />

strategy. This classification is a guide that should help<br />

health care workers make decisions about the management<br />

of COPD in individual patients. Treatment depends on<br />

the patient’s educational level and willingness to apply<br />

the recommended management, on cultural and local<br />

conditions, and on the availability of medications.<br />

EDUCATION<br />

Although patient education is generally regarded as an<br />

essential component of care <strong>for</strong> any chronic disease, the<br />

role of education in COPD has been poorly studied.<br />

Assessment of the value of education in COPD may be<br />

difficult because of the relatively long time required to<br />

achieve improvements in objective measurements of lung<br />

function.<br />

Studies that have been done indicate that patient education<br />

alone does not improve exercise per<strong>for</strong>mance or lung<br />

function 1-4 (Evidence B), but it can play a role in improving<br />

skills, ability to cope with illness, and health status 5 .<br />

MANAGEMENT OF COPD 65


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 66<br />

These outcomes are not traditionally measured in clinical<br />

trials, but they may be most important in COPD where<br />

even pharmacologic interventions generally confer only a<br />

small benefit in terms of lung function.<br />

Patient education regarding smoking cessation has the<br />

greatest capacity to influence the natural history of<br />

COPD. Evaluation of the smoking cessation component<br />

in a long-term, multicenter study indicates that if effective<br />

resources and time are dedicated to smoking cessation,<br />

25% long-term quit rates can be maintained 6 (Evidence<br />

A). Education also improves patient response to<br />

exacerbations 7,8 (Evidence B). Prospective end-of-life<br />

discussions can lead to understanding of advance<br />

directives and effective therapeutic decisions at the end<br />

of life 9 (Evidence B).<br />

Ideally, educational messages should be incorporated<br />

into all aspects of care <strong>for</strong> COPD and may take place<br />

in many settings: consultations with physicians or other<br />

health care workers, home-care or outreach programs,<br />

and comprehensive pulmonary rehabilitation programs.<br />

Goals and Educational Strategies<br />

It is vital <strong>for</strong> patients with COPD to understand the nature<br />

of their disease, risk factors <strong>for</strong> progression, and their role<br />

and the role of health care workers in achieving optimal<br />

management and health outcomes. Education should be<br />

tailored to the needs and environment of the individual<br />

patient, interactive, directed at improving quality of life,<br />

simple to follow, practical, and appropriate to the intellectual<br />

and social skills of the patient and the caregivers.<br />

In managing COPD, open communication between patient<br />

and physician is essential. In addition to being empathic,<br />

attentive and communicative, health professionals should<br />

pay attention to patients’ fears and apprehensions, focus<br />

on educational goals, tailor treatment regimens to each<br />

individual patient, anticipate the effect of functional<br />

decline, and optimize patients’ practical skills.<br />

Several specific education strategies have been shown to<br />

improve patient adherence to medication and management<br />

regimens. In COPD, adherence does not simply refer to<br />

whether patients take their medication appropriately. It<br />

also covers a range of non-pharmacologic treatments -<br />

e.g., maintaining an exercise program after pulmonary<br />

rehabilitation, undertaking and sustaining smoking<br />

cessation, and using devices such as nebulizers, spacers,<br />

and oxygen concentrators properly.<br />

Components of an Education Program<br />

The topics that seem most appropriate <strong>for</strong> an education<br />

program include: smoking cessation; basic in<strong>for</strong>mation<br />

about COPD and pathophysiology of the disease; general<br />

approach to therapy and specific aspects of medical<br />

treatment; self-management skills; strategies to help<br />

minimize dyspnea; advice about when to seek help; selfmanagement<br />

and decision-making during exacerbations;<br />

and advance directives and end-of-life issues (Figure 5-3-2).<br />

Education should be part of consultations with health<br />

care workers beginning at the time of first assessment <strong>for</strong><br />

Figure 5-3-2. Topics <strong>for</strong> Patient Education<br />

Stage 0: At Risk<br />

• In<strong>for</strong>mation and advice about reducing risk factors<br />

Stage I: Mild COPD through Stage III: Severe COPD<br />

Above topic, plus:<br />

• In<strong>for</strong>mation about the nature of COPD<br />

• Instruction on how to use inhalers and other treatments<br />

• Recognition and treatment of exacerbations<br />

• Strategies <strong>for</strong> minimizing dyspnea<br />

Stage IV: Very Severe COPD<br />

Above topics, plus:<br />

• In<strong>for</strong>mation about complications<br />

• In<strong>for</strong>mation about oxygen treatment<br />

• Advance directives and end-of-life decisions<br />

COPD and continuing with each follow-up visit. The<br />

intensity and content of these educational messages<br />

should vary depending on the severity of the patient’s<br />

disease. In practice, a patient often poses a series of<br />

questions to the physician (Figure 5-3-3). It is important<br />

to answer these questions fully and clearly, as this may<br />

help make treatment more effective.<br />

There are several different types of educational programs,<br />

ranging from simple distribution of printed materials, to<br />

teaching sessions designed to convey in<strong>for</strong>mation about<br />

COPD, to workshops designed to train patients in specific<br />

skills (e.g., self-management, peak flow monitoring).<br />

Although printed materials may be a useful adjunct to<br />

other educational messages, passive dissemination of<br />

printed materials alone does not improve skills or health<br />

outcomes. Education is most effective when it is interactive<br />

and conducted in small workshops 4 (Evidence B)<br />

designed to improve both knowledge and skills.<br />

Behavioral approaches such as cognitive therapy and<br />

behavior modification lead to more effective selfmanagement<br />

skills and maintenance of exercise programs.<br />

66 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 67<br />

Figure 5-3-3. Examples of Patient Questions<br />

• What is COPD?<br />

• What causes COPD?<br />

• How will it affect me?<br />

• Can it be treated?<br />

• What will happen if my disease gets worse?<br />

• What will happen if I need to be admitted to the<br />

hospital?<br />

• How will I know when I need oxygen at home?<br />

• What if I do not wish to be admitted to intensive<br />

care <strong>for</strong> ventilation?<br />

Answers to these questions can be developed from this document<br />

and will depend on local circumstances. In all cases, it is important<br />

that answers are clear and use terminology that the patient understands.<br />

Cost Effectiveness of Education Programs<br />

<strong>for</strong> COPD Patients<br />

The cost effectiveness of education programs <strong>for</strong> COPD<br />

patients is highly dependent on local factors that influence<br />

the cost of access to medical services and that will vary<br />

substantially between countries. In one cost-benefit<br />

analysis of education provided to hospital inpatients with<br />

COPD 10 , an in<strong>for</strong>mation package resulted in increased<br />

knowledge of COPD and reduced use of health services,<br />

including reductions of hospital readmissions and general<br />

practice consultations. The education package involved<br />

training patients to increase knowledge of COPD,<br />

medication usage, precautions <strong>for</strong> exacerbations, and<br />

peak flow monitoring technique. However, this study<br />

was undertaken in a heterogeneous group of patients -<br />

65% were smokers and 88% were judged to have an<br />

asthmatic component to their disease - and these<br />

findings may not hold true <strong>for</strong> a “pure” COPD population.<br />

In a study of mild to moderate COPD patients at an outpatient<br />

clinic, patient education involving one four hour<br />

group session followed by one to two individual nurseand<br />

physiotherapist-sessions improved patient outcomes<br />

and reduced costs in a 12-month follow-up 220 .<br />

Although a healthy lifestyle is important, and should be<br />

encouraged, additional studies are needed to identify<br />

specific components of self-management programs that<br />

are effective 200 .<br />

PHARMACOLOGIC TREATMENT<br />

Overview of the Medications<br />

Pharmacologic therapy is used to prevent and control<br />

symptoms, reduce the frequency and severity of<br />

exacerbations, improve health status, and improve<br />

exercise tolerance. None of the existing medications <strong>for</strong><br />

COPD have been shown to modify the long-term decline<br />

in lung function that is the hallmark of this disease 6,11-13<br />

(Evidence A). However, this should not preclude ef<strong>for</strong>ts<br />

to use medications to control symptoms. Since COPD<br />

is usually progressive, recommendations <strong>for</strong> the<br />

pharmacological treatment of COPD reflect the following<br />

general principles:<br />

• There should be a stepwise increase in treatment,<br />

depending on the severity of the disease. (The<br />

step-down approach used in the chronic treatment<br />

of asthma is not applicable to COPD.)<br />

• Regular treatment needs to be maintained at the same<br />

level <strong>for</strong> long periods of time unless significant side<br />

effects occur or the disease worsens.<br />

• Treatment response of an individual patient is variable<br />

and should be monitored closely and adjusted frequently.<br />

The medications are presented in the order in which they<br />

would normally be introduced in patient care, based on<br />

the level of disease severity. However, each treatment<br />

regimen needs to be patient-specific as the relationship<br />

between the severity of symptoms and the severity of airflow<br />

limitation is influenced by other factors, such as the<br />

frequency and severity of exacerbations, the presence of<br />

one or more complications, the presence of respiratory<br />

failure, comorbidities (cardiovascular disease, sleep-related<br />

disorders, etc.), and general health status.<br />

Bronchodilators<br />

Medications that increase the FEV 1 or change other<br />

spirometric variables, usually by altering airway smooth<br />

muscle tone, are termed bronchodilators 14 , since the<br />

improvements in expiratory flow reflect widening of the<br />

airways rather than changes in lung elastic recoil. Such<br />

drugs improve emptying of the lungs, tend to reduce<br />

dynamic hyperinflation at rest and during exercise 15 ,<br />

and improve exercise per<strong>for</strong>mance. The extent of these<br />

changes, especially in moderate to severe disease, is<br />

not easily predictable from the improvement in FEV 1<br />

16,17<br />

.<br />

Regular bronchodilation with drugs that act primarily on<br />

airway smooth muscle does not modify the decline of<br />

function in mild COPD and, by inference, the prognosis<br />

of the disease 6 (Evidence B).<br />

Bronchodilator medications are central to the symptomatic<br />

management of COPD 18-21 (Evidence A). They are given<br />

either on an as-needed basis <strong>for</strong> relief of persistent or<br />

worsening symptoms, or on a regular basis to prevent or<br />

reduce symptoms. The side effects of bronchodilator<br />

therapy are pharmacologically predictable and dose<br />

dependent. Adverse effects are less likely, and resolve<br />

more rapidly after treatment withdrawal, with inhaled than<br />

MANAGEMENT OF COPD 67


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 68<br />

with oral treatment. However, COPD patients tend to be<br />

older than asthma patients and more likely to have<br />

comorbidities, so their risk of developing side effects is<br />

greater.<br />

When treatment is given by the inhaled route, attention to<br />

effective drug delivery and training in inhaler technique is<br />

essential. COPD patients may have more problems in<br />

effective coordination and find it harder to use a simple<br />

Metered Dose Inhaler (MDI) than do healthy volunteers<br />

or younger asthmatics. It is essential to ensure that inhaler<br />

technique is correct and to re-check this at each visit.<br />

Alternative breath-activated or spacer devices are available<br />

<strong>for</strong> most <strong>for</strong>mulations. Dry powder inhalers may be more<br />

convenient and possibly provide improved drug deposition,<br />

although this has not been established in COPD. In<br />

general, particle deposition will tend to be more central<br />

with the fixed airflow limitation and lower inspiratory flow<br />

rates in COPD 22,23 .<br />

Wet nebulizers are not recommended <strong>for</strong> regular treatment<br />

because they are more expensive and require appropriate<br />

maintenance 24 . A list of currently available inhaler devices<br />

is provided at http://www.goldcopd.org/inhalers/. The<br />

choice will depend on availability, cost, the prescribing<br />

physician, and the skills and ability of the patient.<br />

Dose-response relationships using the FEV 1 as the<br />

outcome are relatively flat with all classes of<br />

bronchodilators 18-21 . The dose-response relationships <strong>for</strong><br />

short-acting anticholinergics and ß 2 -agonists are shown<br />

in Figure 5-3-5 21 . Toxicity is also dose related.<br />

Increasing the dose of either a ß 2 -agonist or an anticholinergic<br />

by an order of magnitude, especially when<br />

given by a wet nebulizer, appears to provide subjective<br />

benefit in acute episodes 25 (Evidence B) but is not<br />

necessarily helpful in stable disease 26 (Evidence C).<br />

Inhaled ß 2 -agonists have a relatively rapid onset of<br />

bronchodilator effect although this is probably slower in<br />

COPD than in asthma. The bronchodilator effects of<br />

short-acting ß 2 -agonists usually wear off within 4 to 6<br />

hours 27,28 (Evidence A). For single-dose, as needed use<br />

in COPD, there appears to be no advantage in using levalbuterol<br />

over conventional nebulized bronchodilators 201 .<br />

Long-acting inhaled ß 2 -agonists, such as salmeterol and<br />

<strong>for</strong>moterol, show a duration of effect of 12 hours or more<br />

with no loss of effectiveness overnight or with regular use<br />

in COPD patients 29-32 (Evidence A). The long-acting<br />

inhaled anticholinergic, tiotropium, has a duration of<br />

action of more than 24 hours (Evidence A) 33-35 .<br />

All categories of bronchodilators have been shown to<br />

increase exercise capacity in COPD, without necessarily<br />

producing significant changes in FEV 1<br />

36,37,221,222<br />

(Evidence A).<br />

Regular treatment with long-acting bronchodilators is<br />

more effective and convenient than treatment with<br />

short-acting bronchodilators, but more expensive 34,38,39,223<br />

(Evidence A).<br />

Figure 5-3-5. Dose-Response<br />

Relationships <strong>for</strong> Short-acting Bronchodilators 21<br />

Figure 5-3-4. Bronchodilators in Stable COPD<br />

• Bronchodilator medications are central to symptom<br />

management in COPD.<br />

• Inhaled therapy is preferred.<br />

• The choice between ß 2 -agonist, anticholinergic,<br />

theophylline, or combination therapy depends on<br />

availability and individual response in terms of<br />

symptom relief and side effects.<br />

• Bronchodilators are prescribed on an as-needed or<br />

on a regular basis to prevent or reduce symptoms.<br />

• Long-acting inhaled bronchodilators are more<br />

effective and convenient, but more expensive.<br />

• Combining bronchodilators may improve efficacy<br />

and decrease the risk of side effects compared to<br />

increasing the dose of a single bronchodilator.<br />

Cumulative dose (µg)<br />

Open symbols - salbutamol (ß 2 -agonist)<br />

Closed symbols - ipratropium (anticholinergic)<br />

Squares - patients with asthma<br />

Triangles - patients with COPD<br />

Printed with permission from Higgins BG, Powell RM, Cooper S, Tattersfield AE. European<br />

Respiratory Journal 1991; 4:415-20. Copyright 1991 European Respiratory Society Journals, Ltd.<br />

68 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 69<br />

Drug<br />

2 -agonists<br />

Short-acting<br />

Fenoterol<br />

Salbutamol (albuterol)<br />

Terbutaline<br />

Long-acting<br />

Formoterol<br />

Salmeterol<br />

Anticholinergics<br />

Short-acting<br />

Ipratropium bromide<br />

Oxitropium bromide<br />

Long-acting<br />

Fenoterol/Ipratropium<br />

Salbutamol/Ipratropium<br />

Aminophylline<br />

Theophylline (SR)<br />

Beclomethasone<br />

Budesonide<br />

Fluticasone<br />

Triamcinolone<br />

Prednisone<br />

Methyl-prednisolone<br />

Figure 5-3-6. Commonly Used Formulations of Drugs used in COPD<br />

Inhaler<br />

(g)<br />

100-200 (MDI)<br />

100, 200 (MDI & DPI)<br />

400, 500 (DPI)<br />

4.5–12 (MDI & DPI)<br />

25-50 (MDI & DPI)<br />

20, 40 (MDI)<br />

100 (MDI)<br />

200/80 (MDI)<br />

75/15 (MDI)<br />

50-400 (MDI & DPI)<br />

100, 200, 400 (DPI)<br />

50-500 (MDI & DPI)<br />

100 (MDI)<br />

Formoterol/Budesonide 4.5/80, 160 (DPI)<br />

(9/320) (DPI)<br />

Salmeterol/Fluticasone 50/100, 250, 500 (DPI)<br />

25/50, 125, 250 (MDI)<br />

Systemic glucocorticosteroids<br />

10-2000 mg<br />

MDI=metered dose inhaler; DPI=dry powder inhaler<br />

Solution <strong>for</strong><br />

Nebulizer (mg/ml)<br />

1<br />

5<br />

–<br />

0.25-0.5<br />

1.5<br />

1.25/0.5<br />

0.75/4.5<br />

Oral<br />

0.05% (Syrup)<br />

5mg (Pill)<br />

Syrup 0.024%<br />

2.5, 5 (Pill)<br />

200-600 mg (Pill)<br />

100-600 mg (Pill)<br />

5-60 mg (Pill)<br />

4, 8, 16 mg (Pill)<br />

Vials <strong>for</strong> Injection<br />

(mg)<br />

0.1, 0.5<br />

0.2, 0.25<br />

Duration of Action<br />

(hours)<br />

Tiotropium 18 (DPI) 24+<br />

Combination short-acting 2 -agonists plus anticholinergic in one inhaler<br />

Methylxanthines<br />

Inhaled glucocorticosteroids<br />

0.2-0.4<br />

0.20, 0.25, 0.5<br />

40 40<br />

Combination long-acting 2 -agonists plus glucocorticosteroids in one inhaler<br />

4-6<br />

4-6<br />

4-6<br />

12+<br />

12+<br />

6-8<br />

7-9<br />

6-8<br />

6-8<br />

240 mg Variable, up to 24<br />

Variable, up to 24<br />

Regular use of a long-acting ß 2 -agonist 38 or a short- or<br />

long-acting anticholinergic improves health status 34,38,39 .<br />

Theophylline is effective in COPD, but due to its potential<br />

toxicity inhaled bronchodilators are preferred when available.<br />

All studies that have shown efficacy of theophylline in<br />

COPD were done with slow-release preparations. The<br />

classes of bronchodilator drugs commonly used in treating<br />

COPD, ß 2 -agonists, anticholinergics, and methylxanthines,<br />

are shown in Figure 5-3-6. The choice depends on the<br />

availability of medication and the patient’s response.<br />

ß 2 -agonists. The principal action of ß 2 -agonists is to<br />

relax airway smooth muscle by stimulating ß 2 -adrenergic<br />

receptors, which increases cyclic AMP and produces<br />

MANAGEMENT OF COPD 69


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 70<br />

functional antagonism to bronchoconstriction. Oral<br />

therapy is slower in onset and has more side effects than<br />

inhaled treatment 40 (Evidence A).<br />

Adverse effects. Stimulation of ß 2 -receptors can produce<br />

resting sinus tachycardia and has the potential to precipitate<br />

cardiac rhythm disturbances in very susceptible patients,<br />

although this appears to be a remarkably rare event with<br />

inhaled therapy. Exaggerated somatic tremor is troublesome<br />

in some older patients treated with higher doses of<br />

ß 2 -agonists, whatever the route of administration, and<br />

this limits the dose that can be tolerated. Although<br />

hypokalemia can occur, especially when treatment is<br />

combined with thiazide diuretics 41 , and oxygen consumption<br />

can be increased under resting conditions 42 , these metabolic<br />

effects show tachyphylaxis unlike the bronchodilator actions.<br />

Mild falls in PaO 2 occur after administration of both shortand<br />

long-acting ß 2 -agonists 43 , but the clinical significance<br />

of these changes is doubtful. Despite the concerns<br />

raised some years ago, further detailed study has found<br />

no association between ß 2 -agonist use and an accelerated<br />

loss of lung function or increased mortality in COPD.<br />

Anticholinergics. The most important effect of<br />

anticholinergic medications in COPD patients appears<br />

to be blockage of acetylcholine’s effect on M3 receptors.<br />

Current short-acting drugs also block M2 receptors and<br />

modify transmission at the pre-ganglionic junction,<br />

although these effects appear less important in COPD 44 .<br />

The long-acting tiotropium has a pharmacokinetic<br />

selectivity <strong>for</strong> the M3 and the M1 receptor 45 .<br />

The bronchodilating effect of short-acting inhaled<br />

anticholinergics lasts longer than that of short-acting<br />

ß 2 -agonists, with some bronchodilator effect generally<br />

apparent up to 8 hours after administration 27 (Evidence<br />

A). The long-acting inhaled anticholinergic, tiotropium,<br />

has a duration of action of more than 24 hours 33-35<br />

(Evidence A).<br />

Adverse effects. Anticholinergic drugs, such as ipratropium,<br />

oxitropium and tiotropium bromide, are poorly absorbed<br />

which limits the otherwise troublesome systemic effects<br />

seen with atropine. Extensive use of this class of inhaled<br />

agents in a wide range of doses and clinical settings has<br />

shown them to be very safe. The main side effect is dryness<br />

of the mouth. Twenty-one days of inhaled tiotropium, 18<br />

µg day as a dry powder, does not retard mucus clearance<br />

from the lungs 224 . Although occasional prostatic symptoms<br />

have been reported, there are no data to prove a true<br />

causal relationship. A bitter, metallic taste is reported<br />

by some patients using ipratropium. An unexpected<br />

small increase in cardiovascular events in COPD patients<br />

regularly treated with ipratropium bromide has been<br />

reported and requires further investigation 46 .<br />

Use of wet nebulizer solutions with a face mask has been<br />

reported to precipitate acute glaucoma, probably by a<br />

direct effect of the solution on the eye. Mucociliary<br />

clearance is unaffected by these drugs, and respiratory<br />

infection rates are not increased.<br />

Methylxanthines. Controversy remains about the exact<br />

effects of xanthine derivatives. They may act as nonselective<br />

phosphodiesterase inhibitors, but have also<br />

been reported to have a range of non-bronchodilator<br />

actions, the significance of which is disputed 47-51 . Data on<br />

duration of action <strong>for</strong> conventional, or even slow-release,<br />

xanthine preparations are lacking in COPD. Changes in<br />

inspiratory muscle function have been reported in<br />

patients treated with theophylline 47 , but whether this<br />

reflects changes in dynamic lung volumes or a primary<br />

effect on the muscle is not clear (Evidence B). All studies<br />

that have shown efficacy of theophylline in COPD were<br />

done with slow-release preparations. Theophylline is<br />

effective in COPD but, due to its potential toxicity, inhaled<br />

bronchodilators are preferred when available.<br />

Adverse effects. Toxicity is dose related, a particular<br />

problem with the xanthine derivatives because their<br />

therapeutic ratio is small and most of the benefit occurs<br />

only when near-toxic doses are given 49,50 (Evidence A).<br />

Methylxanthines are nonspecific inhibitors of all phosphodiesterase<br />

enzyme subsets, which explains their<br />

Figure 5-3-7. Drugs and Physiological Variables<br />

that Affect Theophylline Metabolism in COPD<br />

Increased<br />

• Tobacco smoking<br />

• Anticonvulsant drugs<br />

• Rifampicin<br />

• Alcohol<br />

Decreased<br />

• Old age<br />

• Arterial hypoxemia<br />

(PaO 2 < 6.0 kPa, 45<br />

mm Hg)<br />

• Respiratory acidosis<br />

• Congestive cardiac<br />

failure<br />

• Liver cirrhosis<br />

• Erythromycin<br />

• Quinolone antibiotics<br />

• Cimetidine<br />

(not ranitidine)<br />

• Viral infections<br />

• Herbal remedies<br />

(St. John’s Wort)<br />

70 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 71<br />

wide range of toxic effects. Problems include the<br />

development of atrial and ventricular arrhythmias (which<br />

can prove fatal) and grand mal convulsions (which can<br />

occur irrespective of prior epileptic history). More<br />

common and less dramatic side effects include<br />

headaches, insomnia, nausea, and heartburn, and<br />

these may occur within the therapeutic range of serum<br />

theophylline. Unlike the other bronchodilator classes,<br />

xanthine derivatives may involve a risk of overdose<br />

(either intentional or accidental).<br />

Theophylline, the most commonly used methylxanthine,<br />

is metabolized by cytochrome P450 mixed function<br />

oxidases. Clearance of the drug declines with age.<br />

Many other physiological variables and drugs modify<br />

theophylline metabolism; some of the potentially<br />

important interactions are listed in Figure 5-3-7.<br />

Combination bronchodilator therapy. Combining<br />

bronchodilators with different mechanisms and durations<br />

of action may increase the degree of bronchodilation <strong>for</strong><br />

equivalent or lesser side effects. A combination of a<br />

short-acting ß 2 -agonist and an anticholinergic produces<br />

greater and more sustained improvements in FEV 1<br />

than either drug alone and does not produce evidence<br />

of tachyphylaxis over 90 days of treatment 27,52,53<br />

(Evidence A).<br />

The combination of a ß 2 -agonist, an anticholinergic,<br />

and/or theophylline may produce additional improvements<br />

in lung function 27,51,54-56 and health status 27,57 . Increasing<br />

the number of drugs usually increases costs, and an<br />

equivalent benefit may occur by increasing the dose of<br />

one bronchodilator when side effects are not a limiting<br />

factor. Detailed assessments of this approach have not<br />

been carried out.<br />

Glucocorticosteroids<br />

The effects of oral and inhaled glucocorticosteroids in<br />

COPD are much less dramatic than in asthma, and their<br />

role in the management of stable COPD is limited to very<br />

specific indications. The use of glucocorticosteroids <strong>for</strong><br />

the treatment of acute exacerbations is described in<br />

Component 4: Manage Exacerbations.<br />

Oral glucocorticosteroids - short-term. Many existing<br />

COPD guidelines recommend the use of a short course<br />

(two weeks) of oral glucocorticosteroids to identify COPD<br />

patients who might benefit from long-term treatment with<br />

oral or inhaled glucocorticosteroids. This recommendation<br />

is based on evidence 58 that short-term effects predict<br />

long-term effects of oral glucocorticosteroids on FEV 1 ,<br />

and evidence that asthma patients with airflow limitation<br />

might not respond acutely to an inhaled bronchodilator<br />

but do show significant bronchodilation after a short<br />

course of oral glucocorticosteroids.<br />

There is mounting evidence, however, that a short course<br />

of oral glucocorticosteroids is a poor predictor of the longterm<br />

response to inhaled glucocorticosteroids in COPD 13,59 .<br />

For this reason, there appears to be insuffi-cient evidence<br />

to recommend a therapeutic trial with oralglucocorticosteroids<br />

in patients with Stage II: Moderate COPD, Stage<br />

III: Severe COPD, or Stage IV: Very Severe COPD and<br />

poor response to an inhaled bronchodilator.<br />

Oral glucocorticosteroids - long-term. Two retrospective<br />

studies 60,61 analyzed the effects of treatment with oral<br />

glucocorticosteroids on long-term FEV 1 changes in clinic<br />

populations of patients with moderate to very severe<br />

COPD. The retrospective nature of these studies, the lack<br />

of a true control group, and the imprecise definition of<br />

COPD are reasons <strong>for</strong> a cautious interpretation of the<br />

data and conclusions.<br />

A side effect of long-term treatment with systemic<br />

glucocorticosteroids is steroid myopathy 62-64 , which<br />

contributes to muscle weakness, decreased functionality,<br />

and respiratory failure in subjects with advanced COPD.<br />

In view of the well-known toxicity of long-term treatment<br />

with oral glucocorticosteroids, it is not surprising that no<br />

prospective studies have been per<strong>for</strong>med on the longterm<br />

effects of these drugs in COPD.<br />

There<strong>for</strong>e, based on the lack of evidence of benefit, and<br />

the large body of evidence on side effects, long-term<br />

treatment with oral glucocorticosteroids is not recommended<br />

in COPD (Evidence A).<br />

Inhaled glucocorticosteroids. Regular treatment with<br />

inhaled glucocorticosteroids does not modify the longterm<br />

decline of FEV 1 in patients with COPD 11-13,65 .<br />

However, regular treatment with inhaled glucocorticosteroids<br />

is appropriate <strong>for</strong> symptomatic COPD patients<br />

with an FEV 1 < 50% predicted (Stage III: Severe COPD<br />

and Stage IV: Very Severe COPD) and repeated<br />

exacerbations (<strong>for</strong> example, 3 in the last three years) 66-69<br />

(Evidence A). This treatment has been shown to reduce<br />

the frequency of exacerbations and thus improve health<br />

status 224 (Evidence A), and withdrawal from treatment<br />

with inhaled glucocorticosteroids can lead to exacerbations<br />

in some patients 202 . Inhaled glucocorticosteroid combined<br />

with a long-acting ß 2 -agonist is more effective than the<br />

individual components 66,68,69,203,204 (Evidence A). Shortterm<br />

treatment with a combined inhaled glucocorticosteroid<br />

and long-acting ß 2 -agonist resulted in greater control of<br />

lung function and symptoms than combined anticholinergic<br />

and short-acting ß 2 -agonist 225 .<br />

MANAGEMENT OF COPD 71


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 72<br />

Figure 5-3-8. Therapy at Each Stage of COPD<br />

Old 0: At Risk I: Mild II: Moderate III: Severe<br />

IIA<br />

IIB<br />

New 0: At Risk I: Mild II: Moderate III: Severe IV: Very Severe<br />

Characteristics • <strong>Chronic</strong> symptoms<br />

• Exposure to risk<br />

factors<br />

• Normal spirometry<br />

• FEV 1 /FVC < 70%<br />

• FEV 1 ≥ 80%<br />

• With or without<br />

symptoms<br />

• FEV 1 /FVC < 70%<br />

• 50% ≤ FEV 1 < 80%<br />

• With or without<br />

symptoms<br />

• FEV 1 /FVC < 70%<br />

• 30% ≤ FEV 1 < 50%<br />

• With or without<br />

symptoms<br />

• FEV 1 /FVC < 70%<br />

• FEV 1 < 30% or FEV 1 < 50%<br />

predicted plus chronic<br />

respiratory failure<br />

Avoidance of risk factor(s); influenza vaccination<br />

Add short-acting bronchodilator when needed<br />

Add regular treatment with one or more<br />

long-acting bronchodilators<br />

Add rehabilitation<br />

Add inhaled glucocorticosteroids<br />

if repeated exacerbations<br />

Add long-term<br />

oxygen if chronic<br />

respiratory<br />

failure<br />

Consider surgical<br />

treatments<br />

The dose-response relationships and long-term safety<br />

of inhaled glucocorticosteroids in COPD are not known.<br />

Only moderate to high doses have been used in long- term<br />

clinical trials. Two studies showed an increased incidence<br />

of skin bruising in a small percentage of the COPD<br />

patients 11,13 . One long-term study showed no effect of<br />

budesonide on bone density and fracture rate 11,70 , while<br />

another study showed that treatment with triamcinolone<br />

acetonide was associated with a decrease in bone<br />

density 65 . The efficacy and side effects of inhaled<br />

glucocorticosteroids in asthma are dependent on the dose<br />

and type of glucocorticosteroid 71 . This pattern can also<br />

be expected in COPD and needs documentation in this<br />

patient population. Treatment with inhaled glucocorticosteroids<br />

can be recommended <strong>for</strong> patients with more<br />

advanced COPD and repeated exacerbations.<br />

Pharmacologic Therapy by <strong>Disease</strong> Severity<br />

Figure 5-3-8 provides a summary of recommended<br />

treatment at each stage of COPD. For patients with few<br />

or intermittent symptoms (Stage I: Mild COPD), shortacting<br />

inhaled therapy as needed to control dyspnea or<br />

coughing spasms is sufficient. If inhaled bronchodilators<br />

are not available, regular treatment with slow-release<br />

theophylline should be considered.<br />

In patients with Stage II: Moderate COPD to Stage IV:<br />

Very Severe COPD) whose symptoms are not adequately<br />

controlled with as-needed short-acting bronchodilators,<br />

adding regular treatment with a long-acting inhaled<br />

bronchodilator is recommended (Evidence A). Regular<br />

treatment with long-acting bronchodilators is more effective<br />

and convenient than treatment with short-acting<br />

72 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 73<br />

bronchodilators, but more expensive (Evidence A). There<br />

is insufficient evidence to favor one or the other longacting<br />

bronchodilator. For patients who need additional<br />

symptom control, adding theophylline leads to additional<br />

benefits.<br />

Patients with Stage II: Moderate COPD to Stage IV:<br />

Very Severe COPD who are on regular short- or longacting<br />

bronchodilator therapy may also use a short-acting<br />

bronchodilator as needed.<br />

Some patients may request regular treatment with<br />

high-dose nebulized bronchodilators, especially if they<br />

have experienced subjective benefit from this treatment<br />

during an acute exacerbation. Clear scientific evidence<br />

<strong>for</strong> this approach is lacking, but one suggested option is<br />

to examine the improvement in mean daily peak<br />

expiratory flow recording during two weeks of treatment<br />

in the home and continue with nebulizer therapy if a<br />

significant change occurs 24 . In general, nebulized therapy<br />

<strong>for</strong> a stable patient is not appropriate unless it has been<br />

shown to be better than conventional dose therapy.<br />

In patients with a postbronchodilator FEV 1 < 50%<br />

predicted (Stage III: Severe COPD to Stage IV: Very<br />

Severe COPD) and a history of repeated exacerbations<br />

(<strong>for</strong> example, three in the last three years), regular<br />

treatment with inhaled glucocorticosteroids reduce<br />

frequency of exacerbations and improve health status.<br />

In these patients, regular treatment with an inhaled<br />

glucocorticosteroid should be added to regular<br />

bronchodilator treatment. <strong>Chronic</strong> treatment with oral<br />

glucocorticosteroids should be avoided.<br />

Other Pharmacologic Treatments<br />

Vaccines. Influenza vaccines can reduce serious<br />

illness 226 72, 205<br />

and death in COPD patients by about 50%<br />

(Evidence A). Vaccines containing killed or live, inactivated<br />

viruses are recommended 73 as they are more effective in<br />

elderly patients with COPD 74 . The strains are adjusted<br />

each year <strong>for</strong> appropriate effectiveness and should be<br />

given once (in autumn) or twice (in autumn and winter)<br />

each year. A pneumococcal vaccine containing 23<br />

virulent serotypes has been used, but sufficient data to<br />

support its general use in COPD patients are lacking 75-77<br />

(Evidence B).<br />

Alpha-1 antitrypsin augmentation therapy. Young<br />

patients with severe hereditary alpha-1 antitrypsin<br />

deficiency and established emphysema may be<br />

candidates <strong>for</strong> alpha-1 antitrypsin augmentation therapy.<br />

However, this therapy is very expensive, is not available<br />

in most countries, and is not recommended <strong>for</strong> patients<br />

with COPD that is unrelated to alpha-1 antitrypsin<br />

deficiency (Evidence C).<br />

Antibiotics. In several large-scale controlled studies 78-80 ,<br />

prophylactic, continuous use of antibiotics was shown<br />

to have no effect on the frequency of exacerbations in<br />

COPD. Another study examined the efficacy of winter<br />

chemoprophylaxis over a period of 5 years and concluded<br />

that there was no benefit 81 . Thus, on the present evidence,<br />

the use of antibiotics, other than <strong>for</strong> treating infectious<br />

exacerbations of COPD and other bacterial infections, is<br />

not recommended 82,83 (Evidence A).<br />

Mucolytic (mucokinetic, mucoregulator) agents<br />

(ambroxol, erdosteine, carbocysteine, iodinated glycerol).<br />

The regular use of mucolytics in COPD has been<br />

evaluated in a number of long-term studies with<br />

controversial results 84-86 . The majority showed no effect<br />

of mucolytics on lung function or symptoms, although<br />

some have reported a reduction in the frequency of<br />

exacerbations. A Cochrane collaborative review<br />

per<strong>for</strong>med a meta-analysis of all the available data,<br />

including that from a number of abstracts 87 . A statistically<br />

significant reduction in the number of episodes of chronic<br />

bronchitis was found in patients treated with mucolytics<br />

compared to those receiving placebo. However, these<br />

data are not easy to interpret, as the follow-up ranged<br />

from 2 to 6 months and the patients all had an FEV 1 ><br />

50% predicted. Although a few patients with viscous<br />

sputum may benefit from mucolytics 88,89 , the overall<br />

benefits seem to be very small. There<strong>for</strong>e, the widespread<br />

use of these agents cannot be recommended<br />

on the basis of the present evidence (Evidence D).<br />

Antioxidant agents. Antioxidants, in particular<br />

N-acetylcysteine, have been shown to reduce the<br />

frequency of exacerbations and could have a role in the<br />

treatment of patients with recurrent exacerbations 90-93<br />

(Evidence B). However, be<strong>for</strong>e their routine use can be<br />

recommended, the results of ongoing trials will have to<br />

be carefully evaluated.<br />

Immunoregulators (immunostimulators,<br />

immunomodulators). Studies using an immunostimulator<br />

in COPD show a decrease in the severity and frequency<br />

of exacerbations 94,227 . However, additional studies to<br />

examine the long term effects of this therapy are required<br />

be<strong>for</strong>e regular use can be recommended 95 (Evidence B).<br />

Antitussives. Cough, although sometimes a troublesome<br />

symptom in COPD, has a significant protective role 96 .<br />

Thus the regular use of antitussives is contraindicated in<br />

stable COPD (Evidence D).<br />

Vasodilators. The belief that pulmonary hypertension<br />

in COPD is associated with a poorer prognosis has<br />

provoked many attempts to reduce right ventricular<br />

afterload, increase cardiac output, and improve oxygen<br />

MANAGEMENT OF COPD 73


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 74<br />

delivery and tissue oxygenation. Many agents have<br />

been evaluated, including inhaled nitric oxide, but the<br />

results have been uni<strong>for</strong>mly disappointing. In patients<br />

with COPD, in whom hypoxemia is caused primarily by<br />

venti-lation-perfusion mismatching rather than by<br />

increased intrapulmonary shunt (as in noncardiogenic<br />

pulmonary edema), inhaled nitric oxide can worsen gas<br />

exchange because of altered hypoxic regulation of<br />

ventilation-perfusion balance 97,98 . There<strong>for</strong>e, based on<br />

the available evidence, nitric oxide is contraindicated in<br />

stable COPD.<br />

Respiratory stimulants. Almitrine bismesylate, a relatively<br />

specific peripheral chemoreceptor stimulant that increases<br />

ventilation at any level of CO 2 under hypoxemic conditions,<br />

has been studied in both stable respiratory failure and<br />

exacerbations. It improves ventilation-perfusion relationships<br />

by modifying the hypoxic vasoconstrictor response.<br />

Oral almitrine has been shown to improve oxygenation,<br />

but to a lesser degree than low doses of inspired O 2 .<br />

There is no evidence that almitrine improves survival or<br />

quality of life, and in large clinical trials it was associated<br />

with a number of significant side effects, particularly<br />

peripheral neuropathy 99-101 . There<strong>for</strong>e, on the present<br />

evidence almitrine is not recommended <strong>for</strong> regular use in<br />

stable COPD patients (Evidence B). The use of<br />

doxapram, a non-specific but relatively safe respiratory<br />

stimulant available as an intravenous <strong>for</strong>mulation, is not<br />

recommended in stable COPD (Evidence D).<br />

Narcotics (morphine). The use of oral and parenteral<br />

opioids are effective <strong>for</strong> treating dyspnea in COPD<br />

patients with advanced disease. There are insufficient<br />

data to conclude whether nebulized opioids are<br />

effective 102 . However, some clinical studies suggest that<br />

morphine used to control dyspnea may have serious<br />

adverse effects and its benefits may be limited to a few<br />

sensitive subjects 103-107 .<br />

Others. Nedocromil, leukotriene modifiers, and alternative<br />

healing methods (e.g., herbal medicine, acupunture,<br />

homeopathy) have not been adequately tested in COPD<br />

patients and thus cannot be recommended at this time.<br />

NON-PHARMACOLOGIC TREATMENT<br />

Rehabilitation<br />

The principal goals of pulmonary rehabilitation are to<br />

reduce symptoms, improve quality of life, and increase<br />

physical and emotional participation in everyday activities.<br />

To accomplish these goals, pulmonary rehabilitation<br />

covers a range of non-pulmonary problems that may not<br />

be adequately addressed by medical therapy <strong>for</strong> COPD.<br />

Such problems, which especially affect patients with<br />

Figure 5-3-9. The Cycle of Physical, Social, and<br />

Psychosocial Consequences of COPD<br />

COPD<br />

Dyspnea<br />

Depression<br />

Lack of Fitness<br />

Immobility<br />

Social Isolation<br />

Stage II: Moderate COPD, Stage III: Severe COPD,<br />

and Stage IV: Very Severe COPD, include exercise<br />

deconditioning, relative social isolation, altered mood<br />

states (especially depression), muscle wasting, and<br />

weight loss. These problems have complex interrelationships<br />

and improvement in any one of these interlinked<br />

processes can interrupt the “vicious circle” in COPD so<br />

that positive gains occur in all aspects of the illness<br />

(Figure 5-3-9).<br />

Figure 5-3-10. Benefits of Pulmonary Rehabilitation<br />

in COPD 5,110-120<br />

• Improves exercise capacity (Evidence A).<br />

• Reduces the perceived intensity of breathlessness<br />

(Evidence A).<br />

• Can improve health-related quality of life (Evidence A).<br />

• Reduces the number of hospitalizations and days in<br />

the hospital (Evidence A).<br />

• Reduces anxiety and depression associated with<br />

COPD (Evidence A).<br />

• Strength and endurance training of the upper limbs<br />

improves arm function (Evidence B).<br />

• Benefits extend well beyond the immediate period of<br />

training (Evidence B).<br />

• Improves survival (Evidence B).<br />

• Respiratory muscle training is beneficial, especially<br />

when combined with general exercise training<br />

(Evidence C).<br />

• Psychosocial intervention is helpful (Evidence C).<br />

74 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 75<br />

Pulmonary rehabilitation has been carefully evaluated in<br />

a large number of clinical trials; the various benefits are<br />

summarized in Figure 5-3-10 5,108-118 .<br />

Patient selection and program design. Although<br />

more in<strong>for</strong>mation is needed on criteria <strong>for</strong> patient<br />

selection <strong>for</strong> pulmonary rehabilitation programs, COPD<br />

patients at all stages of disease appear to benefit from<br />

exercise training programs, improving with respect to<br />

both exercise tolerance and symptoms of dyspnea and<br />

fatigue 119 (Evidence A). Data suggest that these<br />

benefits can be sustained even after a single pulmonary<br />

rehabilitation program 120-122 .<br />

Benefit does wane after a rehabilitation program ends,<br />

but if exercise training is maintained at home the patient’s<br />

health status remains above pre-rehabilitation levels<br />

(Evidence B). To date there is no consensus on whether<br />

repeated rehabilitation courses enable patients to sustain<br />

the benefits gained through the initial course.<br />

Ideally, pulmonary rehabilitation should involve several<br />

types of health professionals. Significant benefits can<br />

also occur with more limited personnel, as long as<br />

dedicated professionals are aware of the needs of each<br />

patient. Benefits have been reported from rehabilitation<br />

programs conducted in inpatient, outpatient, and home<br />

settings 112,113,123 . Considerations of cost and availability<br />

most often determine the choice of setting. The<br />

educational and exercise training components of<br />

rehabilitation are usually conducted in groups, normally<br />

with 6 to 8 individuals per class (Evidence D).<br />

The following points summarize current knowledge of<br />

considerations important in choosing patients:<br />

Functional status: Benefits have been seen in patients<br />

with a wide range of disability, although those who are<br />

chair-bound appear unlikely to respond even to home visiting<br />

programs 124 (Evidence A).<br />

Severity of dyspnea: Stratification by breathlessness<br />

intensity using the MRC questionnaire (Figure 5-1-3)<br />

may be helpful in selecting patients most likely to benefit<br />

from rehabilitation. Those with MRC grade 5 dyspnea<br />

may not benefit 124 (Evidence B).<br />

Motivation: Selecting highly motivated participants is<br />

especially important in the case of outpatient programs 121 .<br />

Smoking status: There is no evidence that smokers will<br />

benefit less than nonsmokers, but many clinicians<br />

believe that inclusion of a smoker in a rehabilitation<br />

program should be conditional on their participation in a<br />

smoking cessation program. Some data indicate that continuing<br />

smokers are less likely to complete pulmonary<br />

rehabilitation programs than nonsmokers 121 (Evidence B).<br />

Components of pulmonary rehabilitation programs.<br />

The components of pulmonary rehabilitation vary<br />

widely from program to program but a comprehensive<br />

pulmonary rehabilitation program includes exercise<br />

training, nutrition counseling, and education.<br />

Exercise training. Exercise tolerance can be assessed<br />

by either bicycle ergometry or treadmill exercise with the<br />

measurement of a number of physiological variables,<br />

including maximum oxygen consumption, maximum heart<br />

rate, and maximum work per<strong>for</strong>med. A less complex<br />

approach is to use a self-paced, timed walking test<br />

(e.g., 6-minute walking distance). These tests require at<br />

least one practice session be<strong>for</strong>e data can be interpreted.<br />

Shuttle walking tests offer a compromise: they provide<br />

more complete in<strong>for</strong>mation than an entirely self-paced<br />

test, but are simpler to per<strong>for</strong>m than a treadmill test 125 .<br />

Exercise training ranges in frequency from daily to<br />

weekly, in duration from 10 minutes to 45 minutes per<br />

session, and in intensity from 50% peak oxygen<br />

consumption (VO 2 max) to maximum tolerated 126 . The<br />

optimum length <strong>for</strong> an exercise program has not been<br />

investigated in randomized, controlled trials. Thus, the<br />

length depends on the resources available and usually<br />

ranges from 4 to 10 weeks, with longer programs<br />

resulting in larger effects than shorter programs 111 .<br />

Participants are often encouraged to achieve a<br />

predetermined target heart rate 127 , but this goal may have<br />

limitations in COPD. In many programs, especially those<br />

using simple corridor exercise training, the patient is<br />

encouraged to walk to a symptom-limited maximum, rest,<br />

and then continue walking until 20 minutes of exercise<br />

have been completed. Use of a simple wheeled walkingaid<br />

seems to improve walking distance and reduces<br />

breathlessness in severely disabled COPD patients<br />

(Evidence C) 206-208 . The minimum length of an effective<br />

rehabilitation program is two months; the longer the<br />

program continues, the more effective the results<br />

(Evidence B) 128-130 . However, as yet, no effective program<br />

has been developed to maintain the effects over time 131 .<br />

Many physicians advise patients unable to participate in<br />

a structured program to exercise on their own (e.g., walking<br />

20 minutes daily). The benefits of this general advice<br />

have not been tested, but it is reasonable to offer such<br />

advice to patients if a <strong>for</strong>mal program is not available.<br />

Some programs also include upper limb exercises,<br />

usually involving an upper limb ergometer or resistive<br />

training with weights. There are no randomized clinical<br />

trial data to support the routine inclusion of these<br />

exercises, but they may be helpful in patients with<br />

comorbidities that restrict other <strong>for</strong>ms of exercise and<br />

those with evidence of respiratory muscle weakness 132,133 .<br />

The addition of upper limb exercises or other strength<br />

MANAGEMENT OF COPD 75


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 76<br />

training to aerobic training is effective in improving strength,<br />

but does not improve quality of life or exercise tolerance 134 .<br />

Nutrition counseling. Nutritional state is an important<br />

determinant of symptoms, disability, and prognosis in<br />

COPD; both overweight and underweight can be a<br />

problem. Specific nutritional recommendations <strong>for</strong><br />

patients with COPD are based on expert opinion and<br />

some small randomized clinical trials 209 . Approximately<br />

25% of patients with Stage II: Moderate COPD to Stage<br />

IV: Very Severe COPD show a reduction in both their<br />

body mass index and fat free mass 135-137 . A reduction in<br />

body mass index is an independent risk factor <strong>for</strong><br />

mortality in COPD patients 138-140 (Evidence A).<br />

Health care workers should identify and correct the<br />

reasons <strong>for</strong> reduced calorie intake in COPD patients.<br />

Patients who become breathless while eating should be<br />

advised to take small, frequent meals. Poor dentition<br />

should be corrected and comorbidities (pulmonary sepsis,<br />

lung tumors, etc.) should be managed appropriately.<br />

Improving the nutritional state of weight-losing COPD<br />

patients can lead to improved respiratory muscle<br />

strength 141-143 . However, controversy remains as to<br />

whether this additional ef<strong>for</strong>t is cost effective 141,142 . Present<br />

evidence suggests that nutritional supplementa-tion alone<br />

may not be a sufficient strategy. Increased calorie intake<br />

is best accompanied by exercise regimes that have a<br />

nonspecific anabolic action. This approach has not been<br />

<strong>for</strong>mally tested in large numbers of subjects. Anabolic<br />

steroids in patients with COPD with weight loss increase<br />

body weight and lean body mass but have little or no<br />

effect on exercise capacity 144,145<br />

Education. Most pulmonary rehabilitation programs<br />

include an educational component, but the specific<br />

contributions of education to the improvements seen after<br />

pulmonary rehabilitation remain unclear.<br />

Assessment and follow-up. Baseline and outcome<br />

assessments of each participant in a pulmonary<br />

rehabilitation program should be made to quantify<br />

individual gains and target areas <strong>for</strong> improvement.<br />

Assessments should include:<br />

• Detailed history and physical examination.<br />

• Measurement of spirometry be<strong>for</strong>e and after a<br />

bronchodilator drug.<br />

• Assessment of exercise capacity.<br />

• Measurement of health status and impact of<br />

breathlessness.<br />

• Assessment of inspiratory and expiratory muscle<br />

strength and lower limb strength (e.g., quadriceps) in<br />

patients who suffer from muscle wasting.<br />

The first two assessments are important <strong>for</strong> establishing<br />

entry suitability and baseline status but are not used in<br />

outcome assessment. The last three assessments are<br />

baseline and outcome measures.<br />

Several detailed questionnaires <strong>for</strong> assessing health<br />

status are available, including some that are specifically<br />

designed <strong>for</strong> patients with respiratory disease (e.g.,<br />

<strong>Chronic</strong> Respiratory <strong>Disease</strong> Questionnaire 57 , St. George<br />

Respiratory Questionnaire 146 ), and there is increasing<br />

evidence that these questionnaires may be useful in a<br />

clinical setting. Health status can also be assessed by<br />

generic questionnaires, such as the Medical Outcomes<br />

Study Short Form (SF36) 147 , to enable comparison of<br />

quality of life in different diseases.<br />

Economic cost of rehabilitation programs. A<br />

Canadian study showing statistically significant improvements<br />

in dyspnea, fatigue, emotional health, and mastery<br />

found that the incremental cost of pulmonary rehabilitation<br />

was $11,597 (CDN) per person 148 . A study from the<br />

UK provided evidence that an intensive (6-week, 18-visit)<br />

multidisciplinary rehabilitation program was effective in<br />

decreasing use of health services 122 (Evidence B).<br />

Although there was no difference in the number of<br />

hospital admissions between patients with disabling<br />

COPD in a control group and those who participated in<br />

the rehabilitation program, the number of days the<br />

rehabilitation group spent in the hospital was significantly<br />

lower. The rehabilitation group had more primary-care<br />

consultations at the general practitioner’s premises than<br />

did the control group, but fewer primary-care home visits.<br />

Compared with the control group, the rehabilitation group<br />

also showed greater improvements in walking ability and<br />

in general and disease-specific health status.<br />

Oxygen Therapy<br />

Oxygen therapy, one of the principal non-pharmacologic<br />

treatments <strong>for</strong> patients with Stage IV: Very Severe<br />

COPD 88,149 , can be administered in three ways: long-term<br />

continuous therapy, during exercise, and to relieve acute<br />

dyspnea. The primary goal of oxygen therapy is to<br />

increase the baseline PaO 2 to at least 8.0 kPa (60 mm<br />

Hg) at sea level and rest, and/or produce an SaO 2 at<br />

least 90%, which will preserve vital organ function by<br />

ensuring adequate delivery of oxygen.<br />

The long-term administration of oxygen (> 15 hours per<br />

day) to patients with chronic respiratory failure has<br />

been shown to increase survival 150,152 . It can also have a<br />

beneficial impact on hemodynamics, hematologic<br />

characteristics, exercise capacity, lung mechanics, and<br />

mental state 151 . Continuous oxygen therapy decreased<br />

resting pulmonary artery pressure in one study 150 but not<br />

76 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 77<br />

in another study 152 . Several controlled prospective<br />

studies have shown that the primary hemodynamic effect<br />

of oxygen therapy is preventing the progression of<br />

pulmonary hypertension 153,154 . Long-term oxygen therapy<br />

improves general alertness, motor speed, and hand grip,<br />

although the data are less clear about changes in quality<br />

of life and emotional state. The possibility of walking<br />

while using some oxygen devices may help to improve<br />

physical conditioning and have a beneficial influence on<br />

the psychological state of patients 155 .<br />

Long-term oxygen therapy is generally introduced in<br />

Stage IV: Very Severe COPD <strong>for</strong> patients who have:<br />

• PaO 2 at or below 7.3 kPa (55 mm Hg) or SaO 2 at or<br />

below 88%, with or without hypercapnia (Evidence A); or<br />

• PaO 2 between 7.3 kPa (55 mm Hg) and 8.0 kPa<br />

(60 mm Hg), or SaO 2 of 89%, if there is evidence of<br />

pulmonary hypertension, peripheral edema<br />

suggesting congestive cardiac failure, or polycythemia<br />

(hematocrit > 55%) (Evidence D).<br />

A decision about the use of long-term oxygen should be<br />

based on the waking PaO 2 values. The prescription<br />

should always include the source of supplemental oxygen<br />

(gas or liquid), method of delivery, duration of use, and<br />

flow rate at rest, during exercise, and during sleep.<br />

Oxygen therapy given during exercise increases walking<br />

distance and endurance, optimizing oxygen delivery to<br />

tissues and utilization by muscles. However, there are no<br />

data to suggest that long-term oxygen therapy changes<br />

exercise capacity per se. Where available, this treatment<br />

is usually restricted to patients who meet the criteria <strong>for</strong><br />

continuous oxygen therapy, or experience significant<br />

oxygen desaturation during exercise (Evidence C).<br />

Oxygen therapy reduces the oxygen cost of breathing<br />

and minute ventilation, a mechanism that although still<br />

disputed helps to minimize the sensation of dyspnea.<br />

This has led to the use of short burst therapy to control<br />

severe dyspnea such as occurs after climbing stairs.<br />

However, there is no benefit from using short burst<br />

oxygen <strong>for</strong> symptomatic relief be<strong>for</strong>e or after exercise 210,211<br />

(Evidence B).<br />

Cost considerations. Supplemental home oxygen is<br />

usually the most costly component of outpatient therapy<br />

<strong>for</strong> adults with COPD who require this therapy 156 . Studies<br />

of the cost effectiveness of alternative outpatient oxygen<br />

delivery methods in the US and Europe suggest that<br />

oxygen concentrator devices may be more cost effective<br />

than cylinder delivery systems 157,158 .<br />

Oxygen use in air travel. Although air travel is safe <strong>for</strong><br />

most patients with chronic respiratory failure who are on<br />

long-term oxygen therapy, patients should be instructed<br />

to increase the flow by 1-2 L/min during the flight 159 .<br />

Ideally, patients who fly should be able to maintain an<br />

inflight PaO 2 of at least 6.7 kPa (50 mm Hg). Studies<br />

indicate that this can be achieved in those with moderate<br />

to severe hypoxemia at sea level by supplementary<br />

oxygen at 3 L/min by nasal cannulae or 31% by Venturi<br />

facemask 160 . Those with a resting PaO 2 at sea level of<br />

> 9.3 kPa (70 mm Hg) are likely to be safe to fly without<br />

supplementary oxygen 159,161 , although it is important to<br />

emphasize that a resting PaO 2 > 9.3 kPa (70 mm Hg) at<br />

sea level does not exclude the development of severe<br />

hypoxemia when travelling by air (Evidence C). Careful<br />

consideration should be given to any comorbidity that<br />

may impair oxygen delivery to tissues (e.g., cardiac<br />

impairment, anemia). Also, walking along the aisle may<br />

profoundly aggravate hypoxemia 162 .<br />

Ventilatory Support<br />

Although both noninvasive ventilation (using either<br />

negative or positive pressure devices) and invasive<br />

(conventional) mechanical ventilation are essentially<br />

designed to manage and treat acute episodes of COPD,<br />

<strong>for</strong> years noninvasive ventilation has been applied in<br />

patients with Stage IV: Very Severe COPD and chronic<br />

respiratory failure. This has followed the successful use of<br />

noninvasive ventilation in other <strong>for</strong>ms of chronic<br />

respiratory failure due to chest wall de<strong>for</strong>mities and/or<br />

neuromuscular disorders. Several scientific studies have<br />

examined the use of ventilatory support and there is no<br />

convincing evidence that this therapy has a role in the<br />

management of stable COPD. It is possible that some<br />

patients with chronic hypercapnia may benefit from this<br />

<strong>for</strong>m of treatment, but no randomized controlled study<br />

has yet been reported.<br />

Noninvasive mechanical ventilation. This modality of<br />

ventilatory support is applied when endotracheal and<br />

nasotracheal ventilation are not needed, using either<br />

negative pressure ventilation (nPV) or noninvasive<br />

intermittent positive pressure ventilation (NIPPV).<br />

Noninvasive negative pressure ventilation (nPV). The use<br />

of tank respirators, cuirass, or poncho ventilation is largely<br />

of historical interest in COPD. Problems with patient<br />

com<strong>for</strong>t and limited access restrict future use of nPV 163,164 .<br />

When this treatment is used in chronic respiratory failure,<br />

some patients develop upper airway obstruction during<br />

sleep 165 . A comparison of domiciliary active and sham<br />

nPV in patients with chronic respiratory failure due to<br />

COPD showed no differences in shortness of breath,<br />

exercise tolerance, arterial blood gases, respiratory<br />

muscle strength, or quality of life between the two<br />

treatments 166 .<br />

MANAGEMENT OF COPD 77


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 78<br />

Noninvasive intermittent positive pressure ventilation<br />

(NIPPV). The role of NIPPV in chronic respiratory failure<br />

remains unsettled, although this is now the standard<br />

means of providing noninvasive ventilatory support in<br />

other instances of chronic respiratory failure not directly<br />

related to COPD. NIPPV can be delivered by different<br />

types of ventilators: volume-controlled, pressure-controlled,<br />

bilevel positive airway pressure, or continuous positive<br />

airway pressure. New devices with lower cost, greater<br />

ease of operation, and greater portability are constantly<br />

being developed 167 . Recent technical improvements have<br />

facilitated the use of NIPPV while reducing the possibility<br />

of air leaking through face or nasal masks.<br />

A study of NIPPV compared to conventional therapy in a<br />

population with end-stage COPD using a randomized,<br />

crossover design <strong>for</strong> a 3-month period found that the<br />

noninvasive approach is not well tolerated and is<br />

associated with marginal clinical and functional<br />

improvements 168 (Evidence B). Although preliminary<br />

studies have suggested that combining NIPPV with<br />

long-term oxygen therapy could be beneficial on certain<br />

outcome variables, based on a 12-month study 169 and a<br />

24-month study 170 in stable COPD patients with chronic<br />

respiratory failure, its widespread use cannot be<br />

advocated as yet 171 . However, compared with long-term<br />

oxygen therapy alone, the addition of NIPPV has some<br />

effect on carbon dioxide retention and improved<br />

shortness of breath 170 .<br />

Given this conflicting evidence, long-term NIPPV cannot<br />

be recommended <strong>for</strong> the routine treatment of patients<br />

with chronic respiratory failure due to COPD. Nonetheless,<br />

the combination of NIPPV with long-term oxygen<br />

therapy may be of some use in a selected subset of<br />

patients, particularly in those with pronounced daytime<br />

hypercapnia 172 .<br />

Invasive (conventional) mechanical ventilation.<br />

The appropriateness of using invasive (conventional)<br />

ventilation in end-stage COPD continues to be debated.<br />

There are no guidelines to define which patients will<br />

benefit.<br />

Surgical Treatments<br />

Bullectomy. Bullectomy is an older surgical procedure<br />

<strong>for</strong> bullous emphysema. By removing a large bulla that<br />

does not contribute to gas exchange, the adjacent lung<br />

parenchyma is decompressed. Bullectomy can be<br />

per<strong>for</strong>med thoracoscopically. In carefully selected<br />

patients, this procedure is effective in reducing dyspnea<br />

and improving lung function 173 (Evidence C).<br />

Bullae may be removed to alleviate local symptoms<br />

such as hemoptysis, infection, or chest pain, and to allow<br />

re-expansion of a compressed lung region. This is the<br />

usual indication in patients with COPD. In considering the<br />

possible benefit of surgery it is crucial to estimate the<br />

effect of the bulla on the lung and the function of the<br />

nonbullous lung. A thoracic CT scan, arterial blood gas<br />

measurement, and comprehensive respiratory function<br />

tests are essential be<strong>for</strong>e making a decision regarding<br />

suitability <strong>for</strong> resection of a bulla. Normal or minimally<br />

reduced diffusing capacity, absence of significant<br />

hypoxemia, and evidence of regional reduction in perfusion<br />

with good perfusion in the remaining lung are indications<br />

a patient will likely benefit from surgery 174 . However,<br />

pulmonary hypertension, hypercapnia, and severe<br />

emphysema are not absolute contraindications <strong>for</strong><br />

bullectomy. Some investigators have recommended that<br />

the bulla must occupy 50% or more of the hemithorax<br />

and produce definite displacement of the adjacent lung<br />

be<strong>for</strong>e surgery is per<strong>for</strong>med 175 .<br />

<strong>Lung</strong> volume reduction surgery (LVRS). LVRS is a<br />

surgical procedure in which parts of the lung are resected<br />

to reduce hyperinflation 176 , making respiratory muscles<br />

more effective pressure generators by improving their<br />

mechanical efficiency (as measured by length/tension<br />

relationship, curvature of the diaphragm, and area of<br />

apposition) 177,178 . In addition, LVRS increases the elastic<br />

recoil pressure of the lung and thus improves expiratory<br />

flow rates 179 .<br />

LVRS does not improve life expectancy but improves<br />

exercise capacity in patients with predominant upper lobe<br />

emphysema and a low post-rehabilitation exercise<br />

capacity 212 , and may improve global health status in<br />

patients with heterogeneous emphysema 213 .<br />

In some centers with adequate experience, perioperative<br />

mortality of LVRS has been reported to be less than 5%.<br />

Results have been reported following bilateral (upper<br />

parts) resection using median sternotomy 180,181 or<br />

video-assisted thoracoscopy (VATS) 182 . Most studies<br />

select patients with FEV 1 < 35% predicted, PaCO 2<br />

< 6.0 kPa (45 mm Hg), predominant upper lobe<br />

emphysema on CT scan, and a residual volume of ><br />

200% predicted. The average increase in FEV 1 following<br />

LVRS has ranged from 32% to 93%, and the decrease in<br />

TLC from 15% to 20% 180,183 . LVRS appears to improve<br />

exercise capacity as well as quality of life in some patients.<br />

There are reports of these effects lasting more than one<br />

year 180-182 . Patients with an FEV 1 < 20 % predicted<br />

and either homogeneous emphysema on HRCT or a<br />

DLCO < 20 % predicted are at high risk <strong>for</strong> death after<br />

LVRS and unlikely to benefit from the intervention 184 .<br />

78 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 79<br />

Hospital costs associated with LVRS in 52 consecutive<br />

patients 185 ranged from $11,712 to $121,829 (US).<br />

Hospital charges in 23 consecutive patients admitted <strong>for</strong><br />

LVRS at a single institution 186 ranged from $20,032 to<br />

$75,561 with a median charge of $26,669 (US). A small<br />

number of individuals incurred extraordinary costs<br />

because of complications. Advanced age was a significant<br />

factor leading to higher expected total hospital costs.<br />

Although there are some encouraging reports 187 , LVRS<br />

is still an experimental palliative surgical procedure 188 .<br />

Most results (Evidence C) reported to date are from<br />

uncontrolled studies; several large randomized multicenter<br />

studies are underway to investigate the effectiveness and<br />

cost of LVRS in comparison to vigorous conventional<br />

therapy 189 . Until the results of these controlled studies are<br />

known, LVRS cannot be recommended <strong>for</strong> widespread<br />

use.<br />

<strong>Lung</strong> transplantation. In appropriately selected patients<br />

with very advanced COPD, lung transplantation has<br />

been shown to improve quality of life and functional<br />

capacity 190-193 (Evidence C), although the Joint United<br />

Network <strong>for</strong> Organ Sharing in 1998 found that lung<br />

transplantation does not confer a survival benefit in<br />

patients with end-stage emphysema after two years 192 .<br />

Criteria <strong>for</strong> referral <strong>for</strong> lung transplantation include FEV 1<br />

< 35% predicted, PaO 2 < 7.3-8.0 kPa (55-60 mm Hg),<br />

PaCO 2 > 6.7 kPa (50 mm Hg), and secondary pulmonary<br />

hypertension 194,195 .<br />

<strong>Lung</strong> transplantation is limited by the shortage of donor<br />

organs, which has led some centers to adopt the single<br />

lung technique. The common complications seen in<br />

COPD patients after lung transplantation, apart from<br />

operative mortality, are acute rejection and bronchiolitis<br />

obliterans, CMV, other opportunistic fungal (Candida,<br />

Aspergillus, Cryptococcus, Carini) or bacterial<br />

(Pseudomonas, Staphylococcus species) infections,<br />

lymphoproliferative disease, and lymphomas 191 .<br />

Another limitation of lung transplantation is its cost.<br />

Hospitalization costs associated with lung transplantation<br />

have ranged from $110,000 to well over $200,000<br />

(US). Costs remain elevated <strong>for</strong> months to years after<br />

surgery due to the high cost of complications and the<br />

immunosuppressive regimens 196-199 that must be initiated<br />

during or immediately after surgery.<br />

Special Considerations<br />

Surgery in COPD. Postoperative pulmonary complications<br />

are as important and common as postoperative cardiac<br />

complications and, consequently, are a key component<br />

of increased risk of surgery in COPD patients. The principal<br />

potential factors contributing to the risk include smoking,<br />

poor general health status, age, obesity and COPD severity.<br />

A comprehensive definition of postoperative pulmonary<br />

complications should include only major pulmonary<br />

respiratory complications, namely lung infections,<br />

atelectasis and/or increased airflow obstruction, all<br />

potentially resulting in acute respiratory failure and<br />

aggravation of underlying COPD 214-219 .<br />

The incidence of increased risk of postoperative pulmonary<br />

complications in COPD patients may vary according to<br />

the definition of postoperative pulmonary complications<br />

and the severity of COPD, with relative ranges of the<br />

order of 2.7 and 4.7 214 . The surgical site is the most<br />

important predictor, and risk increases as the incision<br />

approaches the diaphragm. Upper abdominal and thoracic<br />

surgery represents the greatest risk, the latter being<br />

uncommon after interventions outside the thorax or<br />

abdomen. Most reports conclude that epidural or spinal<br />

anesthesia have a lower risk than with general anesthesia,<br />

although the results are not totally uni<strong>for</strong>m.<br />

Patient-risk factors are identified by careful history, physical<br />

examination, chest radiography, and pulmonary function<br />

tests. Although the value of pulmonary function tests<br />

remains contentious, there is consensus that all COPD<br />

candidates <strong>for</strong> lung resection should undergo a complete<br />

battery, including <strong>for</strong>ced spirometry with bronchodilator<br />

response, static lung volumes, diffusing capacity and<br />

arterial blood gases at rest. One theoretical rationale<br />

behind the assessment of pulmonary function measurement<br />

is the identification of COPD patients in whom the risk is<br />

so elevated that surgery should be contraindicated.<br />

Several studies in high risk COPD patients suggest that<br />

there is threshold beyond which the risk of surgery is<br />

prohibitive. The risk of postoperative respiratory failure<br />

appears to be in patients undergoing pneumonectomy<br />

with a preoperative FEV 1 < 2 L or 50% predicted and/or<br />

a DLCO < 50% predicted 218 . COPD patients at high risk<br />

due to poor lung function should undergo further lung<br />

function assessment, <strong>for</strong> example, regional distribution<br />

of perfusion and exercise capacity 219 . To prevent<br />

postoperative pulmonary complications, stable COPD<br />

patients clinically symptomatic and/or with limited exercise<br />

capacity should be treated, be<strong>for</strong>e surgery, intensely with<br />

all the measures already well established <strong>for</strong> stable<br />

COPD patients who are not about to have surgery.<br />

Surgery should be postponed if an exacerbation is present.<br />

Surgery in patients with COPD needs to be differentiated<br />

from that aimed to improve function and symptoms <strong>for</strong><br />

COPD. This includes bullectomy, lung volume reduction<br />

surgery and lung transplantation 219 .<br />

MANAGEMENT OF COPD 79


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 80<br />

REFERENCES<br />

1. Reis AL. Response to bronchodilators. In: Clausen J, ed.<br />

Pulmonary function testing: guidelines and controversies.<br />

New York: Academic Press, 1982.<br />

2. Janelli LM, Scherer YK, Schmieder LE. Can a pulmonary<br />

health teaching program alter patients' ability to cope with<br />

COPD? Rehabil Nurs 1991; 16:199-202.<br />

3. Ashikaga T, Vacek PM, Lewis SO. Evaluation of a community-based<br />

education program <strong>for</strong> individuals with chronic<br />

obstructive pulmonary disease. J Rehabil 1980; 46:23-7.<br />

4. Toshima MT, Kaplan RM, Ries AL. Experimental evaluation<br />

of rehabilitation in chronic obstructive pulmonary disease:<br />

short-term effects on exercise endurance and health status.<br />

Health Psychol 1990; 9:237-52.<br />

5. Celli BR. Pulmonary rehabilitation in patients with COPD.<br />

Am J Respir Crit Care Med 1995; 152:861-4.<br />

6. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey<br />

WC, Buist AS, et al. Effects of smoking intervention and<br />

the use of an inhaled anticholinergic bronchodilator on the<br />

rate of decline of FEV1. The <strong>Lung</strong> Health Study. JAMA<br />

1994; 272:1497-505.<br />

7. Stewart MA. Effective physician-patient communication and<br />

health outcomes: a review. CMAJ 1995; 152:1423-33.<br />

8. Clark NM, Nothwehr F, Gong M, Evans D, Maiman LA,<br />

Hurwitz ME, et al. Physician-patient partnership in managing<br />

chronic illness. Acad Med 1995; 70:957-9.<br />

9. Heffner JE, Fahy B, Hilling L, Barbieri C. Outcomes of<br />

advance directive education of pulmonary rehabilitation<br />

patients. Am J Respir Crit Care Med 1997; 155:1055-9.<br />

10. Tougaard L, Krone T, Sorknaes A, Ellegaard H. Economic<br />

benefits of teaching patients with chronic obstructive pulmonary<br />

disease about their illness. The PASTMA Group.<br />

Lancet 1992; 339:1517-20.<br />

11. Pauwels RA, Lofdahl CG, Laitinen LA, Schouten JP,<br />

Postma DS, Pride NB, et al. Long-term treatment with<br />

inhaled budesonide in persons with mild chronic obstructive<br />

pulmonary disease who continue smoking. European<br />

Respiratory Society Study on <strong>Chronic</strong> <strong>Obstructive</strong><br />

Pulmonary <strong>Disease</strong>. N Engl J Med 1999; 340:1948-53.<br />

12. Vestbo J, Sorensen T, Lange P, Brix A, Torre P, Viskum K.<br />

Long-term effect of inhaled budesonide in mild and moderate<br />

chronic obstructive pulmonary disease: a randomised<br />

controlled trial. Lancet 1999; 353:1819-23.<br />

13. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson<br />

JA, Maslen TK. Randomised, double blind, placebo controlled<br />

study of fluticasone propionate in patients with moderate<br />

to severe chronic obstructive pulmonary disease: the<br />

ISOLDE trial. BMJ 2000; 320:1297-303.<br />

14. Calverley PMA. Symptomatic bronchodilator treatment. In:<br />

Calverley PMA, Pride NB, eds. <strong>Chronic</strong> obstructive pulmonary<br />

disease. London: Chapman and Hall, 1995:419-445.<br />

15. Belman MJ, Botnick WC, Shin JW. Inhaled bronchodilators<br />

reduce dynamic hyperinflation during exercise in patients<br />

with chronic obstructive pulmonary disease. Am J Respir<br />

Crit Care Med 1996; 153:967-75.<br />

16. Berger R, Smith D. Effect of inhaled metaproterenol on<br />

exercise per<strong>for</strong>mance in patients with stable "fixed" airway<br />

obstruction. Am Rev Respir Dis 1988; 138:624-9.<br />

17. Hay JG, Stone P, Carter J, Church S, Eyre-Brook A,<br />

Pearson MG, et al. Bronchodilator reversibility, exercise<br />

per<strong>for</strong>mance and breathlessness in stable chronic obstructive<br />

pulmonary disease. Eur Respir J 1992; 5:659-64.<br />

18. Vathenen AS, Britton JR, Ebden P, Cookson JB, Wharrad<br />

HJ, Tattersfield AE. High-dose inhaled albuterol in severe<br />

chronic airflow limitation. Am Rev Respir Dis 1988; 138:850-5.<br />

19. Gross NJ, Petty TL, Friedman M, Skorodin MS, Silvers<br />

GW, Donohue JF. Dose response to ipratropium as a nebulized<br />

solution in patients with chronic obstructive pulmonary<br />

disease. A three-center study. Am Rev Respir Dis<br />

1989; 139:1188-91.<br />

20. Chrystyn H, Mulley BA, Peake MD. Dose response relation<br />

to oral theophylline in severe chronic obstructive airways<br />

disease. BMJ 1988; 297:1506-10.<br />

21. Higgins BG, Powell RM, Cooper S, Tattersfield AE. Effect<br />

of salbutamol and ipratropium bromide on airway calibre<br />

and bronchial reactivity in asthma and chronic bronchitis.<br />

Eur Respir J 1991; 4:415-20.<br />

22. Ericsson CH, Svartengren K, Svartengren M, Mossberg B,<br />

Philipson K, Blomquist M, et al. Repeatability of airway<br />

deposition and tracheobronchial clearance rate over three<br />

days in chronic bronchitis. Eur Respir J 1995; 8:1886-93.<br />

23. Kim CS, Kang TC. Comparative measurement of lung<br />

deposition of inhaled fine particles in normal subjects and<br />

patients with obstructive airway disease. Am J Respir Crit<br />

Care Med 1997; 155:899-905.<br />

24. Boe J, Dennis JH, O'Driscoll BR, Bauer TT, Carone M,<br />

Dautzenberg B, et al. European Respiratory Society<br />

Guidelines on the use of nebulizers. Eur Respir J 2001;<br />

18(1):228-42.<br />

25. O'Driscoll BR, Kay EA, Taylor RJ, Weatherby H, Chetty<br />

MC, Bernstein A. A long-term prospective assessment of<br />

home nebulizer treatment. Respir Med 1992; 86:317-25.<br />

26. Jenkins SC, Heaton RW, Fulton TJ, Moxham J.<br />

Comparison of domiciliary nebulized salbutamol and salbutamol<br />

from a metered-dose inhaler in stable chronic airflow<br />

limitation. Chest 1987; 91:804-7.<br />

27. COMBIVENT Inhalation Aerosol Study Group. In chronic<br />

obstructive pulmonary disease, a combination of ipratropium<br />

and albuterol is more effective than either agent alone.<br />

An 85-day multicenter trial. Chest 1994; 105:1411-9.<br />

28. van Schayck CP, Folgering H, Harbers H, Maas KL, van<br />

Weel C. Effects of allergy and age on responses to salbutamol<br />

and ipratropium bromide in moderate asthma and<br />

chronic bronchitis. Thorax 1991; 46:355-9.<br />

29. Ulrik CS. Efficacy of inhaled salmeterol in the management<br />

of smokers with chronic obstructive pulmonary disease: a<br />

single centre randomised, double blind, placebo controlled,<br />

crossover study. Thorax 1995; 50:750-4.<br />

30. Boyd G, Morice AH, Pouns<strong>for</strong>d JC, Siebert M, Peslis N,<br />

Craw<strong>for</strong>d C. An evaluation of salmeterol in the treatment of<br />

chronic obstructive pulmonary disease (COPD). Eur Respir<br />

J 1997; 10:815-21.<br />

80 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 81<br />

31. Cazzola M, Matera MG, Santangelo G, Vinciguerra A,<br />

Rossi F, D'Amato G. Salmeterol and <strong>for</strong>moterol in partially<br />

reversible severe chronic obstructive pulmonary disease: a<br />

dose-response study. Respir Med 1995; 89:357-62.<br />

32. Rossi A, Kristufek P, Levine BE, Thomson MH, Till D,<br />

Kottakis J, et al. Comparison of the efficacy, tolerability,<br />

and safety of <strong>for</strong>moterol dry powder and oral, slow-release<br />

theophylline in the treatment of COPD. Chest 2002;<br />

121:1058-69.<br />

33. van Noord JA, Bantje TA, Eland ME, Korducki L,<br />

Cornelissen PJ. A randomised controlled comparison of<br />

tiotropium nd ipratropium in the treatment of chronic<br />

obstructive pulmonary disease. The Dutch Tiotropium<br />

Study Group. Thorax 2000; 55:289-94.<br />

34. Vincken W, van Noord JA, Greefhorst AP, Bantje TA,<br />

Kesten S, Korducki L, et al. Improved health outcomes in<br />

patients with COPD during 1 yr's treatment with tiotropium.<br />

Eur Respir J 2002; 19:209-16.<br />

35. Casaburi R, Mahler DA, Jones PW, Wanner A, San PG,<br />

ZuWallack RL, et al. A long-term evaluation of once-daily<br />

inhaled tiotropium in chronic obstructive pulmonary disease.<br />

Eur Respir J 2002; 19:217-24.<br />

36. Ikeda A, Nishimura K, Koyama H, Izumi T. Bronchodilating<br />

effects of combined therapy with clinical dosages of ipratropium<br />

bromide and salbutamol <strong>for</strong> stable COPD: comparison<br />

with ipratropium bromide alone. Chest 1995; 107:401-5.<br />

37. Guyatt GH, Townsend M, Pugsley SO, Keller JL, Short HD,<br />

Taylor DW, et al. Bronchodilators in chronic air-flow limitation.<br />

Effects on airway function, exercise capacity, and<br />

quality of life. Am Rev Respir Dis 1987; 135:1069-74.<br />

38. Mahler DA, Donohue JF, Barbee RA, Goldman MD, Gross<br />

NJ, Wisniewski ME, et al. Efficacy of salmeterol xinafoate<br />

in the treatment of COPD. Chest 1999; 115:957-65.<br />

39. Dahl R, Greefhorst LA, Nowak D, Nonikov V, Byrne AM,<br />

Thomson MH, et al. Inhaled <strong>for</strong>moterol dry powder versus<br />

ipratropium bromide in chronic obstructive pulmonary disease.<br />

Am J Respir Crit Care Med 2001; 164:778-84.<br />

40. Shim CS, Williams MH, Jr. Bronchodilator response to oral<br />

aminophylline and terbutaline versus aerosol albuterol in<br />

patients with chronic obstructive pulmonary disease. Am J<br />

Med 1983; 75:697-701.<br />

41. Lipworth BJ, McDevitt DG, Struthers AD. Hypokalemic and<br />

ECG sequelae of combined beta-agonist/diuretic therapy.<br />

Protection by conventional doses of spironolactone but not<br />

triamterene. Chest 1990; 98:811-5.<br />

42. Uren NG, Davies SW, Jordan SL, Lipkin DP. Inhaled bronchodilators<br />

increase maximum oxygen consumption in<br />

chronic left ventricular failure. Eur Heart J 1993; 14:744-50.<br />

43. Khoukaz G, Gross NJ. Effects of salmeterol on arterial<br />

blood gases in patients with stable chronic obstructive pulmonary<br />

disease. Comparison with albuterol and ipratropium.<br />

Am J Respir Crit Care Med 1999; 160:1028-30.<br />

44. Barnes PJ. Bronchodilators: basic pharmacology. In:<br />

Calverley PMA, Pride NB, eds. <strong>Chronic</strong> obstructive pulmonary<br />

disease. London: Chapman and Hall, 1995:391-417.<br />

45. Disse B, Speck GA, Rominger KL, Witek TJ, Jr., Hammer<br />

R. Tiotropium (Spiriva): mechanistical considerations and<br />

clinical profile in obstructive lung disease. Life Sci 1999;<br />

64:457-64.<br />

46. Anthonisen NR, Connett JE, Enright PL, Manfreda J.<br />

Hospitalizations and mortality in the <strong>Lung</strong> Health Study.<br />

Am J Respir Crit Care Med 2002; 166:333-9.<br />

47. Aubier M. Pharmacotherapy of respiratory muscles. Clin<br />

Chest Med 1988; 9:311-24.<br />

48. Moxham J. Aminophylline and the respiratory muscles: an<br />

alternative view. Clin Chest Med 1988; 9:325-36.<br />

49. Murciano D, Auclair MH, Pariente R, Aubier M. A randomized,<br />

controlled trial of theophylline in patients with severe<br />

chronic obstructive pulmonary disease. N Engl J Med<br />

1989; 320:1521-5.<br />

50. McKay SE, Howie CA, Thomson AH, Whiting B, Addis GJ.<br />

Value of theophylline treatment in patients handicapped by<br />

chronic obstructive lung disease. Thorax 1993; 48:227-32.<br />

51. Taylor DR, Buick B, Kinney C, Lowry RC, McDevitt DG.<br />

The efficacy of orally administered theophylline, inhaled<br />

salbutamol, and a combination of the two as chronic therapy<br />

in the management of chronic bronchitis with reversible<br />

air-flow obstruction. Am Rev Respir Dis 1985; 131:747-51.<br />

52. The COMBIVENT Inhalation Solution Study Group.<br />

Routine nebulized ipratropium and albuterol together are<br />

better than either alone in COPD. Chest 1997; 112:1514-21.<br />

53. Gross N, Tashkin D, Miller R, Oren J, Coleman W, Linberg<br />

S. Inhalation by nebulization of albuterol-ipratropium combination<br />

(Dey combination) is superior to either agent<br />

alone in the treatment of chronic obstructive pulmonary<br />

disease. Dey Combination Solution Study Group.<br />

Respiration 1998; 65:354-62.<br />

54. van Noord JA, de Munck DR, Bantje TA, Hop WC, Akveld<br />

ML, Bommer AM. Long-term treatment of chronic obstructive<br />

pulmonary disease with salmeterol and the additive<br />

effect of ipratropium. Eur Respir J 2000; 15:878-85.<br />

55. ZuWallack RL, Mahler DA, Reilly D, Church N, Emmett A,<br />

Rickard K, et al. Salmeterol plus theophylline combination<br />

therapy in the treatment of COPD. Chest 2001; 119:1661-70.<br />

56. Bellia V, Foresi A, Bianco S, Grassi V, Olivieri D, Bensi G,<br />

et al. Efficacy and safety of oxitropium bromide, theophylline<br />

and their combination in COPD patients: a doubleblind,<br />

randomized, multicentre study (BREATH Trial).<br />

Respir Med 2002; 96:881-9.<br />

57. Guyatt GH, Berman LB, Townsend M, Pugsley SO,<br />

Chambers LW. A measure of quality of life <strong>for</strong> clinical trials<br />

in chronic lung disease. Thorax 1987; 42:773-8.<br />

58. Callahan CM, Dittus RS, Katz BP. Oral corticosteroid therapy<br />

<strong>for</strong> patients with stable chronic obstructive pulmonary<br />

disease. A meta-analysis. Ann Intern Med 1991; 114:216-23.<br />

59. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson<br />

JA. Prednisolone response in patients with chronic<br />

obstructive pulmonary disease: results from the ISOLDE<br />

study. Thorax. 2003 Aug;58(8):654-8.<br />

60. Postma DS, Peters I, Steenhuis EJ, Sluiter HJ. Moderately<br />

severe chronic airflow obstruction. Can corticosteroids slow<br />

down obstruction? Eur Respir J 1988; 1:22-6.<br />

MANAGEMENT OF COPD 81


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 82<br />

61. Postma DS, Steenhuis EJ, van der Weele LT, Sluiter HJ.<br />

Severe chronic airflow obstruction: can corticosteroids slow<br />

down progression? Eur J Respir Dis 1985; 67:56-64.<br />

62. Decramer M, de Bock V, Dom R. Functional and histologic<br />

picture of steroid-induced myopathy in chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med 1996;<br />

153:1958-64.<br />

63. Decramer M, Lacquet LM, Fagard R, Rogiers P. Corticosteroids<br />

contribute to muscle weakness in chronic airflow obstruction.<br />

Am J Respir Crit Care Med 1994; 150:11-6.<br />

64. Decramer M, Stas KJ. Corticosteroid-induced myopathy<br />

involving respiratory muscles in patients with chronic<br />

obstructive pulmonary disease or asthma. Am Rev Respir<br />

Dis 1992; 146:800-2.<br />

65. The <strong>Lung</strong> Health Study Research Group. Effect of inhaled<br />

triamcinolone on the decline in pulmonary function in<br />

chronic obstructive pulmonary disease: <strong>Lung</strong> Health Study<br />

II. N Engl J Med 2000; 343:1902-1909.<br />

66. Mahler DA, Wire P, Horstman D, Chang CN, Yates J,<br />

Fischer T, et al. Effectiveness of fluticasone propionate and<br />

salmeterol combination delivered via the Diskus device in<br />

the treatment of chronic obstructive pulmonary disease.<br />

Am J Respir Crit Care Med 2002; 166(8):1084-91.<br />

67. Jones PW, Willits LR, Burge PS, Calverley PM. <strong>Disease</strong><br />

severity and the effect of fluticasone propionate on chronic<br />

obstructive pulmonary disease exacerbations. Eur Respir J<br />

2003; 21:68-73.<br />

68. Calverley P, Pauwels R, Vestbo J, Jones P, Pride N,<br />

Gulsvik A, et al. Combined salmeterol and fluticasone in<br />

the treatment of chronic obstructive pulmonary disease: a<br />

randomised controlled trial. Lancet 2003; 361:449-56.<br />

69. Szafranski W, Cukier A, Ramirez A, Menga G, Sansores R,<br />

Nahabedian S, et al. Efficacy and safety of budesonide/<strong>for</strong>moterol<br />

in the management of chronic obstructive pulmonary<br />

disease. Eur Respir J 2003; 21:74-81.<br />

70. Johnell O, Pauwels R, Lofdahl CG, Laitinen LA, Postma<br />

DS, Pride NB, et al. Bone mineral density in patients with<br />

chronic obstructive pulmonary disease treated with budesonide<br />

Turbuhaler. Eur Respir J 2002; 19:1058-63.<br />

71. NHLBI/WHO workshop report: global strategy <strong>for</strong> asthma<br />

management and prevention. Washington, D.C.:<br />

Department of Health and Human Services, 2002:NIH<br />

Publication No. 02-2659.<br />

72. Nichol KL, Margolis KL, Wuorenma J, Von Sternberg T. The<br />

efficacy and cost effectiveness of vaccination against<br />

influenza among elderly persons living in the community. N<br />

Engl J Med 1994; 331:778-84.<br />

73. Edwards KM, Dupont WD, Westrich MK, Plummer WD, Jr.,<br />

Palmer PS, Wright PF. A randomized controlled trial of<br />

cold-adapted and inactivated vaccines <strong>for</strong> the prevention of<br />

influenza A disease. J Infect Dis 1994; 169:68-76.<br />

74. Hak E, van Essen GA, Buskens E, Stalman W, de Melker<br />

RA. Is immunising all patients with chronic lung disease in<br />

the community against influenza cost effective? Evidence<br />

from a general practice based clinical prospective cohort<br />

study in Utrecht, The Netherlands. J Epidemiol Community<br />

Health 1998; 52:120-5.<br />

75. Simberkoff MS, Cross AP, Al-Ibrahim M, Baltch AL,<br />

Geiseler PJ, Nadler J, et al. Efficacy of pneumococcal vaccine<br />

in high-risk patients. Results of a Veterans Administration<br />

Cooperative Study. N Engl J Med 1986; 315:1318-27.<br />

76. Williams JH, Jr., Moser KM. Pneumococcal vaccine and<br />

patients with chronic lung disease. Ann Intern Med 1986;<br />

104:106-9.<br />

77. Davis AL, Aranda CP, Schiffman G, Christianson LC.<br />

Pneumococcal infection and immunologic response to<br />

pneumococcal vaccine in chronic obstructive pulmonary<br />

disease. A pilot study. Chest 1987; 92:204-12.<br />

78. Francis RS, May JR, Spicer CC. Chemotherapy of bronchitis:<br />

influence of penicillin and tetracylcline administered daily,<br />

or intermittently <strong>for</strong> exacerbations. BMJ 1961; 2:979-985.<br />

79. Francis RS, Spicer CC. Chemotherapy in chronic bronchitis:<br />

influence of daily penicillin and teracycline on exacerbations<br />

and their cost. A report to the research committee<br />

of the British Tuberculosis Assoication by their <strong>Chronic</strong><br />

Bronchitis subcommittee. BMJ 1960; 1:297-303.<br />

80. Fletcher CM, Ball JD, Carstairs LW, Couch AHC, Crofton JM,<br />

Edge JR, et al. Value of chemoprophylaxis and chemotherapy<br />

in early chronic bronchitis. A report to the Medical Research<br />

Council by their Working Party on trials of chemotherpay in<br />

early chronic bronchitis. BMJ 1966; 1(5499):1317-1322.<br />

81. Johnston RN, McNeill RS, Smith DH, Dempster MB, Nairn<br />

JR, Purvis MS, et al. Five-year winter chemoprophylaxis <strong>for</strong><br />

chronic bronchitis. Br Med J 1969; 4:265-9.<br />

82. Isada CM, Stoller JK. <strong>Chronic</strong> bronchitis: the role of antibiotics.<br />

In: Niederman MS, Sarosi GA, Glassroth J, eds.<br />

Respiratory infections: a scientific basis <strong>for</strong> management.<br />

London: WB Saunders, 1994:621-33.<br />

83. Siafakas NM, Bouros D. Management of acute exacerbation<br />

of chronic obstructive pulmonary disease. In: Postma<br />

DS, Siafakas NM, eds. Management of chronic obstructive<br />

pulmonary disease. Sheffield: ERS Monograph, 1998:264-77.<br />

84. Allegra L, Cordaro CI, Grassi C. Prevention of acute exacerbations<br />

of chronic obstructive bronchitis with carbocysteine<br />

lysine salt monohydrate: a multicenter, double- blind,<br />

placebo-controlled trial. Respiration 1996; 63:174-80.<br />

85. Guyatt GH, Townsend M, Kazim F, Newhouse MT. A controlled<br />

trial of ambroxol in chronic bronchitis. Chest 1987;<br />

92:618-20.<br />

86. Petty TL. The National Mucolytic Study. Results of a randomized,<br />

double-blind, placebo-controlled study of iodinated<br />

glycerol in chronic obstructive bronchitis. Chest 1990; 97:75-83.<br />

87. Poole PJ, Black PN. Mucolytic agents <strong>for</strong> chronic bronchitis<br />

or chronic obstructive pulmonary disease. Cochrane<br />

Database Syst Rev 2000; 2:Available from URL<br />

www.update-software.com or www.updatusa.com.<br />

88. Siafakas NM, Vermeire P, Pride NB, Paoletti P, Gibson J,<br />

Howard P, et al. Optimal assessment and management of<br />

chronic obstructive pulmonary disease (COPD). The<br />

European Respiratory Society Task Force. Eur Respir J<br />

1995; 8:1398-420.<br />

89. American Thoracic Society. Standards <strong>for</strong> the diagnosis<br />

and care of patients with chronic obstructive pulmonary<br />

disease (COPD) and asthma. Adopted by the ATS Board<br />

of Directors, November 1986. Am Rev Respir Dis 1987;<br />

136:225-44.<br />

82 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 83<br />

90. Hansen NC, Skriver A, Brorsen-Riis L, Balslov S, Evald T,<br />

Maltbaek N, et al. Orally administered N-acetylcysteine<br />

may improve general well-being in patients with mild<br />

chronic bronchitis. Respir Med 1994; 88:531-5.<br />

91. British Thoracic Society Research Committee. Oral N-<br />

acetylcysteine and exacerbation rates in patients with<br />

chronic bronchitis and severe airways obstruction. Thorax<br />

1985; 40:832-5.<br />

92. Boman G, Backer U, Larsson S, Melander B, Wahlander L.<br />

Oral acetylcysteine reduces exacerbation rate in chronic<br />

bronchitis: report of a trial organized by the Swedish<br />

Society <strong>for</strong> Pulmonary <strong>Disease</strong>s. Eur J Respir Dis 1983;<br />

64:405-15.<br />

93. Rasmussen JB, Glennow C. Reduction in days of illness<br />

after long-term treatment with N- acetylcysteine controlledrelease<br />

tablets in patients with chronic bronchitis. Eur<br />

Respir J 1988; 1:351-5.<br />

94. Collet JP, Shapiro P, Ernst P, Renzi T, Ducruet T, Robinson<br />

A. Effects of an immunostimulating agent on acute exacerbations<br />

and hospitalizations in patients with chronic<br />

obstructive pulmonary disease. The PARI-IS Study<br />

Steering Committee and Research Group. Prevention of<br />

Acute Respiratory Infection by an Immunostimulant. Am J<br />

Respir Crit Care Med 1997; 156:1719-24.<br />

95. Anthonisen NR. OM-8BV <strong>for</strong> COPD. Am J Respir Crit Care<br />

Med 1997; 156:1713-4.<br />

96. Irwin RS, Boulet LP, Cloutier MM, Fuller R, Gold PM,<br />

Hoffstein V, et al. Managing cough as a defense mechanism<br />

and as a symptom. A consensus panel report of the<br />

American College of Chest Physicians. Chest 1998;<br />

114:133S-181S.<br />

97. Barbera JA, Roger N, Roca J, Rovira I, Higenbottam TW,<br />

Rodriguez-Roisin R. Worsening of pulmonary gas<br />

exchange with nitric oxide inhalation in chronic obstructive<br />

pulmonary disease. Lancet 1996; 347:436-40.<br />

98. Jones AT, Evans TW. NO: COPD and beyond. Thorax<br />

1997; 52 Suppl 3:S16-21.<br />

99. Watanabe S, Kanner RE, Cutillo AG, Menlove RL, Bachand<br />

RT, Jr., Szalkowski MB, et al. Long-term effect of almitrine<br />

bismesylate in patients with hypoxemic chronic obstructive<br />

pulmonary disease. Am Rev Respir Dis 1989; 140:1269-73.<br />

100. Bardsley PA, Howard P, DeBacker W, Vermeire P, Mairesse<br />

M, Ledent C, et al. Two years treatment with almitrine<br />

bismesylate in patients with hypoxic chronic obstructive airways<br />

disease. Eur Respir J 1991; 4:308-10.<br />

101. Winkelmann BR, Kullmer TH, Kneissl DG, Trenk D,<br />

Kronenberger H. Low-dose almitrine bismesylate in the<br />

treatment of hypoxemia due to chronic obstructive pulmonary<br />

disease. Chest 1994; 105:1383-91.<br />

102. Jennings AL, Davies AN, Higgins JP, Gibbs JS, Broadley<br />

KE. A systematic review of the use of opioids in the management<br />

of dyspnoea. Thorax 2002; 57:939-44.<br />

103. Eiser N, Denman WT, West C, Luce P. Oral diamorphine:<br />

lack of effect on dyspnoea and exercise tolerance in the<br />

"pink puffer" syndrome. Eur Respir J 1991; 4:926-31.<br />

104. Young IH, Daviskas E, Keena VA. Effect of low dose nebulised<br />

morphine on exercise endurance in patients with<br />

chronic lung disease. Thorax 1989; 44:387-90.<br />

105. Woodcock AA, Gross ER, Gellert A, Shah S, Johnson M,<br />

Geddes DM. Effects of dihydrocodeine, alcohol, and caffeine<br />

on breathlessness and exercise tolerance in patients<br />

with chronic obstructive lung disease and normal blood<br />

gases. N Engl J Med 1981; 305:1611-6.<br />

106. Rice KL, Kronenberg RS, Hedemark LL, Niewoehner DE.<br />

Effects of chronic administration of codeine and promethazine<br />

on breathlessness and exercise tolerance in<br />

patients with chronic airflow obstruction. Br J Dis Chest<br />

1987; 81:287-92.<br />

107. Poole PJ, Veale AG, Black PN. The effect of sustainedrelease<br />

morphine on breathlessness and quality of life in<br />

severe chronic obstructive pulmonary disease. Am J<br />

Respir Crit Care Med 1998; 157:1877-80.<br />

108. American Thoracic Society. Pulmonary rehabilitation-1999.<br />

Am J Respir Crit Care Med 1999; 159:1666-82.<br />

109. Fishman AP. Pulmonary rehabilitation research. Am J<br />

Respir Crit Care Med 1994; 149:825-33.<br />

110. Pulmonary rehabilitation: joint ACCP/AACVPR evidencebased<br />

guidelines. ACCP/AACVPR Pulmonary<br />

Rehabilitation Guidelines Panel. American College of<br />

Chest Physicians. American Association of Cardiovascular<br />

and Pulmonary Rehabilitation. Chest 1997; 112:1363-96.<br />

111. Lacasse Y, Wong E, Guyatt GH, King D, Cook DJ, Goldstein<br />

RS. Meta-analysis of respiratory rehabilitation in chronic<br />

obstructive pulmonary disease. Lancet 1996; 348:1115-9.<br />

112. Goldstein RS, Gort EH, Stubbing D, Avendano MA, Guyatt<br />

GH. Randomised controlled trial of respiratory rehabilitation.<br />

Lancet 1994; 344:1394-7.<br />

113. Wijkstra PJ, Van Altena R, Kraan J, Otten V, Postma DS,<br />

Koeter GH. Quality of life in patients with chronic obstructive<br />

pulmonary disease improves after rehabilitation at<br />

home. Eur Respir J 1994; 7:269-73.<br />

114. O'Donnell DE, McGuire M, Samis L, Webb KA. The impact<br />

of exercise reconditioning on breathlessness in severe<br />

chronic airflow limitation. Am J Respir Crit Care Med 1995;<br />

152:2005-13.<br />

115. Lake FR, Henderson K, Briffa T, Openshaw J, Musk AW.<br />

Upper-limb and lower-limb exercise training in patients with<br />

chronic airflow obstruction. Chest 1990; 97:1077-82.<br />

116. Ries AL, Ellis B, Hawkins RW. Upper extremity exercise<br />

training in chronic obstructive pulmonary disease. Chest<br />

1988; 93:688-92.<br />

117. Martinez FJ, Vogel PD, Dupont DN, Stanopoulos I, Gray A,<br />

Beamis JF. Supported arm exercise vs unsupported arm<br />

exercise in the rehabilitation of patients with severe chronic<br />

airflow obstruction. Chest 1993; 103:1397-402.<br />

118. Wijkstra PJ, Ten Vergert EM, van Altena R, Otten V, Kraan<br />

J, Postma DS, et al. Long term benefits of rehabilitation at<br />

home on quality of life and exercise tolerance in patients<br />

with chronic obstructive pulmonary disease. Thorax 1995;<br />

50:824-8.<br />

119. Berry MJ, Rejeski WJ, Adair NE, Zaccaro D. Exercise rehabilitation<br />

and chronic obstructive pulmonary disease stage.<br />

Am J Respir Crit Care Med 1999; 160:1248-53.<br />

MANAGEMENT OF COPD 83


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 84<br />

120. Foglio K, Bianchi L, Bruletti G, Battista L, Pagani M,<br />

Ambrosino N. Long-term effectiveness of pulmonary rehabilitation<br />

in patients with chronic airway obstruction. Eur<br />

Respir J 1999; 13:125-32.<br />

121. Young P, Dewse M, Fergusson W, Kolbe J. Improvements<br />

in outcomes <strong>for</strong> chronic obstructive pulmonary disease<br />

(COPD) attributable to a hospital-based respiratory rehabilitation<br />

programme. Aust N Z J Med 1999; 29:59-65.<br />

122. Griffiths TL, Burr ML, Campbell IA, Lewis-Jenkins V,<br />

Mullins J, Shiels K, et al. Results at 1 year of outpatient<br />

multidisciplinary pulmonary rehabilitation: a randomised<br />

controlled trial. Lancet 2000; 355:362-8.<br />

123. McGavin CR, Gupta SP, Lloyd EL, McHardy GJ. Physical<br />

rehabilitation <strong>for</strong> the chronic bronchitic: results of a controlled<br />

trial of exercises in the home. Thorax 1977; 32:307-11.<br />

124. Wedzicha JA, Bestall JC, Garrod R, Garnham R, Paul EA,<br />

Jones PW. Randomized controlled trial of pulmonary rehabilitation<br />

in severe chronic obstructive pulmonary disease<br />

patients, stratified with the MRC dyspnoea scale. Eur<br />

Respir J 1998; 12:363-9.<br />

125. Singh SJ, Morgan MD, Scott S, Walters D, Hardman AE.<br />

Development of a shuttle walking test of disability in<br />

patients with chronic airways obstruction. Thorax 1992;<br />

47:1019-24.<br />

126. Mahler DA. Pulmonary rehabilitation. Chest 1998;<br />

113:263S-268S.<br />

127. American College of Sports Medicine position stand. The<br />

recommended quantity and quality of exercise <strong>for</strong> developing<br />

and maintaining cardiorespiratory and muscular fitness<br />

in healthy adults. Med Sci Sports Exerc 1990; 22:265-74.<br />

128. Behnke M, Taube C, Kirsten D, Lehnigk B, Jorres RA,<br />

Magnussen H. Home-based exercise is capable of preserving<br />

hospital-based improvements in severe chronic<br />

obstructive pulmonary disease. Respir Med 2000; 94:1184-91.<br />

129. Finnerty JP, Keeping I, Bullough I, Jones J. The effectiveness<br />

of outpatient pulmonary rehabilitation in chronic lung<br />

disease: a randomized controlled trial. Chest 2001;<br />

119:1705-10.<br />

130. Green RH, Singh SJ, Williams J, Morgan MD. A randomised<br />

controlled trial of four weeks versus seven weeks<br />

of pulmonary rehabilitation in chronic obstructive pulmonary<br />

disease. Thorax 2001; 56:143-5.<br />

131. Ries AL, Kaplan RM, Myers R, Prewitt LM. Maintenance<br />

after pulmonary rehabilitation in chronic lung disease: a<br />

randomized trial. Am J Respir Crit Care Med 2003;<br />

167:880-8.<br />

132. Belman MJ, Botnick WC, Nathan SD, Chon KH. Ventilatory<br />

load characteristics during ventilatory muscle training. Am<br />

J Respir Crit Care Med 1994; 149:925-9.<br />

133. Lotters F, van Tol B, Kwakkel G, Gosselink R. Effects of<br />

controlled inspiratory muscle training in patients with<br />

COPD: a meta-analysis. Eur Respir J 2002; 20:570-6.<br />

134. Bernard S, Whittom F, Leblanc P, Jobin J, Belleau R,<br />

Berube C, et al. Aerobic and strength training in patients<br />

with chronic obstructive pulmonary disease. Am J Respir<br />

Crit Care Med 1999; 159:896-901.<br />

135. Schols AM, Soeters PB, Dingemans AM, Mostert R,<br />

Frantzen PJ, Wouters EF. Prevalence and characteristics of<br />

nutritional depletion in patients with stable COPD eligible<br />

<strong>for</strong> pulmonary rehabilitation. Am Rev Respir Dis 1993;<br />

147:1151-6.<br />

136. Engelen MP, Schols AM, Baken WC, Wesseling GJ,<br />

Wouters EF. Nutritional depletion in relation to respiratory<br />

and peripheral skeletal muscle function in out-patients with<br />

COPD. Eur Respir J 1994; 7:1793-7.<br />

137. Wilson DO, Rogers RM, Wright EC, Anthonisen NR. Body<br />

weight in chronic obstructive pulmonary disease. The<br />

National Institutes of Health Intermittent Positive-Pressure<br />

Breathing Trial. Am Rev Respir Dis 1989; 139:1435-8.<br />

138. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss<br />

is a reversible factor in the prognosis of chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med 1998;<br />

157:1791-7.<br />

139. Gray-Donald K, Gibbons L, Shapiro SH, Macklem PT,<br />

Martin JG. Nutritional status and mortality in chronic<br />

obstructive pulmonary disease. Am J Respir Crit Care Med<br />

1996; 153:961-6.<br />

140. Gorecka D, Gorzelak K, Sliwinski P, Tobiasz M, Zielinski J.<br />

Effect of long-term oxygen therapy on survival in patients<br />

with chronic obstructive pulmonary disease with moderate<br />

hypoxaemia. Thorax 1997; 52:674-9.<br />

141. Efthimiou J, Fleming J, Gomes C, Spiro SG. The effect of<br />

supplementary oral nutrition in poorly nourished patients<br />

with chronic obstructive pulmonary disease. Am Rev<br />

Respir Dis 1988; 137:1075-82.<br />

142. Rogers RM, Donahoe M, Costantino J. Physiologic effects<br />

of oral supplemental feeding in malnourished patients with<br />

chronic obstructive pulmonary disease. A randomized control<br />

study. Am Rev Respir Dis 1992; 146:1511-7.<br />

143. Whittaker JS, Ryan CF, Buckley PA, Road JD. The effects<br />

of refeeding on peripheral and respiratory muscle function<br />

in malnourished chronic obstructive pulmonary disease<br />

patients. Am Rev Respir Dis 1990; 142:283-8.<br />

144. Yeh SS, DeGuzman B, Kramer T. Reversal of COPD-associated<br />

weight loss using the anabolic agent oxandrolone.<br />

Chest 2002; 122:421-8.<br />

145. Weisberg J, Wanger J, Olson J, Streit B, Fogarty C, Martin<br />

T, et al. Megestrol acetate stimulates weight gain and ventilation<br />

in underweight COPD patients. Chest 2002;<br />

121:1070-8.<br />

146. Jones PW, Quirk FH, Baveystock CM. The St George's<br />

Respiratory Questionnaire. Respir Med 1991; 85 Suppl<br />

B:25-31; discussion 33-7.<br />

147. Ware JE, Jr., Sherbourne CD. The MOS 36-item short-<strong>for</strong>m<br />

health survey (SF-36). I. Conceptual framework and item<br />

selection. Med Care 1992; 30:473-83.<br />

148. Goldstein RS, Gort EH, Guyatt GH, Feeny D. Economic<br />

analysis of respiratory rehabilitation. Chest 1997; 112:370-9.<br />

149. American Thoracic Society. Standards <strong>for</strong> the diagnosis<br />

and care of patients with chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med 1995; 152:S77-121.<br />

150. Nocturnal Oxygen Therapy Trial Group. Continuous or<br />

nocturnal oxygen therapy in hypoxemic chronic obstructive<br />

lung disease: a clinical trial. Ann Intern Med 1980; 93:391-8.<br />

84 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 85<br />

151. Tarpy SP, Celli BR. Long-term oxygen therapy. N Engl J<br />

Med 1995; 333:710-4.<br />

152. Report of the Medical Research Council Working Party.<br />

Long term domiciliary oxygen therapy in chronic hypoxic<br />

cor pulmonale complicating chronic bronchitis and emphysema.<br />

Lancet 1981; 1:681-6.<br />

153. Weitzenblum E, Sautegeau A, Ehrhart M, Mammosser M,<br />

Pelletier A. Long-term oxygen therapy can reverse the progression<br />

of pulmonary hypertension in patients with chronic<br />

obstructive pulmonary disease. Am Rev Respir Dis<br />

1985; 131:493-8.<br />

154. Zielinski J, Tobiasz M, Hawrylkiewicz I, Sliwinski P,<br />

Palasiewicz G. Effects of long-term oxygen therapy on pulmonary<br />

hemodynamics in COPD patients: a 6-year<br />

prospective study. Chest 1998; 113:65-70.<br />

155. Petty TL. Supportive therapy in COPD. Chest 1998;<br />

113:256S-262S.<br />

156. Petty TL, O'Donohue WJ, Jr. Further recommendations <strong>for</strong><br />

prescribing, reimbursement, technology development, and<br />

research in long-term oxygen therapy. Summary of the<br />

Fourth Oxygen Consensus Conference, Washington, D.C.,<br />

October 15-16, 1993. Am J Respir Crit Care Med 1994;<br />

150:875-7.<br />

157. Pelletier-Fleury N, Lanoe JL, Fleury B, Fardeau M. The<br />

cost of treating COPD patients with long-term oxygen therapy<br />

in a French population. Chest 1996; 110:411-6.<br />

158. Heaney LG, McAllister D, MacMahon J. Cost minimisation<br />

analysis of provision of oxygen at home: are the drug tariff<br />

guidelines cost effective? BMJ 1999; 319:19-23.<br />

159. Gong H, Jr. Air travel and oxygen therapy in cardiopulmonary<br />

patients. Chest 1992; 101:1104-13.<br />

160. Berg BW, Dillard TA, Rajagopal KR, Mehm WJ. Oxygen<br />

supplementation during air travel in patients with chronic<br />

obstructive lung disease. Chest 1992; 101:638-41.<br />

161. Gong H, Jr., Tashkin DP, Lee EY, Simmons MS. Hypoxiaaltitude<br />

simulation test. Evaluation of patients with chronic<br />

airway obstruction. Am Rev Respir Dis 1984; 130:980-6.<br />

162. Christensen CC, Ryg M, Refvem OK, Skjonsberg OH.<br />

Development of severe hypoxaemia in chronic obstructive<br />

pulmonary disease patients at 2,438 m (8,000 ft) altitude.<br />

Eur Respir J 2000; 15:635-9.<br />

163. Muir JF. Pulmonary rehabilitation in chronic respiratory<br />

insufficiency. 5. Home mechanical ventilation. Thorax 1993;<br />

48:1264-73.<br />

164. Simonds AK. Negative pressure ventilation in acute hypercapnic<br />

chronic obstructive pulmonary disease. Thorax<br />

1996; 51:1069-70.<br />

165. Corrado A, Gorini M, Villella G, De Paola E. Negative pressure<br />

ventilation in the treatment of acute respiratory failure:<br />

an old noninvasive technique reconsidered. Eur Respir J<br />

1996; 9:1531-44.<br />

166. Shapiro SH, Ernst P, Gray-Donald K, Martin JG, Wood-<br />

Dauphinee S, Beaupre A, et al. Effect of negative pressure<br />

ventilation in severe chronic obstructive pulmonary disease.<br />

Lancet 1992; 340:1425-9.<br />

167. Hillberg RE, Johnson DC. Noninvasive ventilation. N Engl J<br />

Med 1997; 337:1746-52.<br />

168. Strumpf DA, Millman RP, Carlisle CC, Grattan LM, Ryan<br />

SM, Erickson AD, et al. Nocturnal positive-pressure ventilation<br />

via nasal mask in patients with severe chronic obstructive<br />

pulmonary disease. Am Rev Respir Dis 1991;<br />

144:1234-9.<br />

169. Casanova C, Celli BR, Tost L, Soriano E, Abreu J, Velasco<br />

V, et al. Long-term controlled trial of nocturnal nasal positive<br />

pressure ventilation in patients with severe COPD.<br />

Chest 2000; 118:1582-90.<br />

170. Clini E, Sturani C, Rossi A, Viaggi S, Corrado A, Donner<br />

CF, et al. The Italian multicentre study on noninvasive ventilation<br />

in chronic obstructive pulmonary disease patients.<br />

Eur Respir J 2002; 20:529-38.<br />

171. Elliott MW. Noninvasive ventilation in chronic ventilatory<br />

failure due to chronic obstructive pulmonary disease. Eur<br />

Respir J 2002; 20:511-4.<br />

172. Consensus conference report. Clinical indications <strong>for</strong> noninvasive<br />

positive pressure ventilation in chronic respiratory<br />

failure due to restrictive lung disease, COPD, and nocturnal<br />

hypoventilation. Chest 1999; 116:521-34.<br />

173. Mehran RJ, Deslauriers J. Indications <strong>for</strong> surgery and<br />

patient work-up <strong>for</strong> bullectomy. Chest Surg Clin N Am<br />

1995; 5:717-34.<br />

174. Hughes JA, MacArthur AM, Hutchison DC, Hugh-Jones P.<br />

Long term changes in lung function after surgical treatment<br />

of bullous emphysema in smokers and ex-smokers. Thorax<br />

1984; 39:140-2.<br />

175. Laros CD, Gelissen HJ, Bergstein PG, Van den Bosch JM,<br />

Vanderschueren RG, Westermann CJ, et al. Bullectomy <strong>for</strong><br />

giant bullae in emphysema. J Thorac Cardiovasc Surg<br />

1986; 91:63-70.<br />

176. Cooper JD, Trulock EP, Triantafillou AN, Patterson GA, Pohl<br />

MS, Deloney PA, et al. Bilateral pneumectomy (volume<br />

reduction) <strong>for</strong> chronic obstructive pulmonary disease. J<br />

Thorac Cardiovasc Surg 1995; 109:106-16; discussion<br />

116-9.<br />

177. Criner G, Cordova FC, Leyenson V, Roy B, Travaline J,<br />

Sudarshan S, et al. Effect of lung volume reduction surgery<br />

on diaphragm strength. Am J Respir Crit Care Med 1998;<br />

157:1578-85.<br />

178. Martinez FJ, de Oca MM, Whyte RI, Stetz J, Gay SE, Celli<br />

BR. <strong>Lung</strong>-volume reduction improves dyspnea, dynamic<br />

hyperinflation, and respiratory muscle function. Am J<br />

Respir Crit Care Med 1997; 155:1984-90.<br />

179. Fessler HE, Permutt S. <strong>Lung</strong> volume reduction surgery and<br />

airflow limitation. Am J Respir Crit Care Med 1998;<br />

157:715-22.<br />

180. Cooper JD, Patterson GA, Sundaresan RS, Trulock EP,<br />

Yusen RD, Pohl MS, et al. Results of 150 consecutive bilateral<br />

lung volume reduction procedures in patients with<br />

severe emphysema. J Thorac Cardiovasc Surg 1996;<br />

112:1319-29; discussion 1329-30.<br />

181. Russi EW, Stammberger U, Weder W. <strong>Lung</strong> volume reduction<br />

surgery <strong>for</strong> emphysema. Eur Respir J 1997; 10:208-18.<br />

182. Gelb AF, McKenna RJ, Jr., Brenner M, Schein MJ, Zamel<br />

N, Fischel R. <strong>Lung</strong> function 4 years after lung volume<br />

reduction surgery <strong>for</strong> emphysema. Chest 1999; 116:1608-15.<br />

MANAGEMENT OF COPD 85


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 86<br />

183. Brenner M, McKenna RJ, Jr., Gelb AF, Fischel RJ, Wilson<br />

AF. Rate of FEV1 change following lung volume reduction<br />

surgery. Chest 1998; 113:652-9.<br />

184. National Emphysema Treatment Trial Research Group.<br />

Patients at high risk of death after lung-volume-reduction<br />

surgery. N Engl J Med 2001; 345:1075-83.<br />

185. Elpern EH, Behner KG, Klontz B, Warren WH, Szidon JP,<br />

Kesten S. <strong>Lung</strong> volume reduction surgery: an analysis of<br />

hospital costs. Chest 1998; 113:896-9.<br />

186. Albert RK, Lewis S, Wood D, Benditt JO. Economic aspects<br />

of lung volume reduction surgery. Chest 1996; 110:1068-71.<br />

187. Geddes D, Davies M, Koyama H, Hansell D, Pastorino U,<br />

Pepper J, et al. Effect of lung-volume-reduction surgery in<br />

patients with severe emphysema. N Engl J Med 2000; 343:239-45.<br />

188. Benditt JO, Albert RK. Surgical options <strong>for</strong> patients with<br />

advanced emphysema. Clin Chest Med 1997; 18:577-93.<br />

189. The National Emphysema Treatment Trial Research Group.<br />

Rationale and design of The National Emphysema Treatment<br />

Trial: a prospective randomized trial of lung volume reduction<br />

surgery. Chest 1999; 116:1750-61.<br />

190. Trulock EP. <strong>Lung</strong> transplantation. Am J Respir Crit Care Med<br />

1997; 155:789-818.<br />

191. Theodore J, Lewiston N. <strong>Lung</strong> transplantation comes of age.<br />

N Engl J Med 1990; 322:772-4.<br />

192. Hosenpud JD, Bennett LE, Keck BM, Fiol B, Boucek MM,<br />

Novick RJ. The Registry of the International Society <strong>for</strong><br />

Heart and <strong>Lung</strong> Transplantation: fifteenth official report--<br />

1998. J Heart <strong>Lung</strong> Transplant 1998; 17:656-68.<br />

193. Annual report of the US scientific registry <strong>for</strong> transplant<br />

recipients and the Organ Procurement and Transplantation<br />

Network. Transplant data: 1988-1994. Washington, D.C.:<br />

Division of Transplantation, Health Resources and Services<br />

Administration, US Department of Health and Human<br />

Services, 1995.<br />

194. Hosenpud JD, Bennett LE, Keck BM, Edwards EB, Novick<br />

RJ. Effect of diagnosis on survival benefit of lung transplantation<br />

<strong>for</strong> end- stage lung disease. Lancet 1998; 351:24-7.<br />

195. Maurer JR, Frost AE, Estenne M, Higenbottam T, Glanville<br />

AR. International guidelines <strong>for</strong> the selection of lung transplant<br />

candidates. The International Society <strong>for</strong> Heart and<br />

<strong>Lung</strong> Transplantation, the American Thoracic Society, the<br />

American Society of Transplant Physicians, the European<br />

Respiratory Society. Transplantation 1998; 66:951-6.<br />

196. Ramsey SD, Patrick DL, Albert RK, Larson EB, Wood DE,<br />

Raghu G. The cost-effectiveness of lung transplantation. A<br />

pilot study. University of Washington Medical Center <strong>Lung</strong><br />

Transplant Study Group. Chest 1995; 108:1594-601.<br />

197. Al MJ, Koopmanschap MA, van Enckevort PJ, Geertsma A,<br />

van der Bij W, de Boer WJ, et al. Cost-effectiveness of lung<br />

transplantation in The Netherlands: a scenario analysis.<br />

Chest 1998; 113:124-30.<br />

198. van Enckevort PJ, Koopmanschap MA, Tenvergert EM,<br />

Geertsma A, van der Bij W, de Boer WJ, et al. Lifetime costs<br />

of lung transplantation: estimation of incremental costs.<br />

Health Econ 1997; 6:479-89.<br />

199. van Enckevort PJ, TenVergert EM, Bonsel GJ, Geertsma A,<br />

van der Bij W, de Boer WJ, et al. Technology assessment of<br />

the Dutch <strong>Lung</strong> Transplantation Program. Int J Technol<br />

Assess Health Care 1998; 14:344-56.<br />

200. Monninkhof E, van der Valk P, van der Palen J, van<br />

Herwaarden C, Zielhuis G. Effects of a comprehensive selfmanagement<br />

programme in patients with chronic obstructive<br />

pulmonary disease. Eur Respir J. 2003 Nov;22(5):815-20.<br />

201. Datta D, Vitale A, Lahiri B, ZuWallack R. An evaluation of<br />

nebulized levalbuterol in stable COPD. Chest. 2003<br />

Sep;124(3):844-9.<br />

202. van der Valk P, Monninkhof E, van der Palen J, Zielhuis G,<br />

van Herwaarden C. Effect of discontinuation of inhaled corticosteroids<br />

in patients with chronic obstructive pulmonary disease:<br />

the COPE study. Am J Respir Crit Care Med<br />

2002;166(10):1358-63.<br />

203. Hanania NA, Darken P, Horstman D, Reisner C, Lee B,<br />

Davis S, Shah T. The efficacy and safety of fluticasone propionate(250microg)/salmeterol(50microg)<br />

combined in the<br />

Diskus inhaler <strong>for</strong> the treatment of COPD. Chest. 2003<br />

Sep;124(3):834-43.<br />

204. Calverley PM, Boonsawat W, Cseke Z, Zhong N, Peterson S,<br />

Olsson H. Maintenance therapy with budesonide and <strong>for</strong>moterol<br />

in chronic obstructive pulmonary disease. Eur Respir<br />

J. 2003 Dec;22(6):912-9.<br />

205. Wongsurakiat P, Lertakyamanee J, Maranetra KN,<br />

Jongriratanakul S, Sangkaew S. Economic evaluation of<br />

influenza vaccination in Thai chronic obstructive pulmonary<br />

disease patients. J Med Assoc Thai. 2003 Jun;86(6):497-508.<br />

206. Honeyman P. Barr P. Stubbing DG. Effects of a walking aid on<br />

disability, oxygenation, and breathlessness in patients with<br />

chronic airflow limitation. Journal of Cardiopulmonary<br />

Rehabilitation. 16(1): 63-7, 1996<br />

207. Roomi J.Yohannes AM. Connolly MJ. The effect of walking aids<br />

on exercise capacity and oxygenation in elderly patients with<br />

chronic obstructive pulmonary disease. Age & Ageing.<br />

27(6):703-6, 1998.<br />

208. Yohannes AM, Connolly MJ. Early mobilization with walking aids<br />

following hospital admission with acute exacerbation of chronic<br />

obstructive pulmonary disease. Clin Rehabil. 2003 Aug;17(5):465-71.<br />

209. Steiner MC, Barton RL, Singh SJ, Morgan MD. Nutritional<br />

enhancement of exercise per<strong>for</strong>mance in chronic obstructive<br />

pulmonary disease: a randomised controlled trial. Thorax. 2003<br />

Sep;58(9):745-51.<br />

210. Nandi K, Smith AA, Craw<strong>for</strong>d A, MacRae KD, Garrod R, Seed<br />

WA, Roberts CM. Oxygen supplementation be<strong>for</strong>e or after submaximal<br />

exercise in patients with chronic obstructive pulmonary<br />

disease. Thorax. 2003 Aug;58(8):670-3.<br />

211. Lewis CA, Eaton TE, Young P, Kolbe J. Short-burst oxygen<br />

immediately be<strong>for</strong>e and after exercise is ineffective in nonhypoxic<br />

COPD patients. Eur Respir J. 2003 Oct;22(4):584-8.<br />

212. Fishman A, Martinez F, Naunheim K, Piantadosi S, Wise R, Ries<br />

A, Weinmann G, Wood DE; National Emphysema Treatment Trial<br />

Research Group. A randomized trial comparing lung-volumereduction<br />

surgery with medical therapy <strong>for</strong> severe emphysema.<br />

N Engl J Med. 2003 May 2;348(21):2059-73.<br />

213. Goldstein RS, Todd TR, Guyatt G, Keshavjee S, Dolmage TE,<br />

van Rooy S, Krip B, Maltais F, LeBlanc P, Pakhale S, Waddell<br />

TK. Influence of lung volume reduction surgery (LVRS) on<br />

health related quality of life in patients with chronic obstructive<br />

pulmonary disease. Thorax. 2003 May;58(5):405-10.<br />

214. Smetana GW. Preoperative pulmonary evaluation. N Engl J Med<br />

1999;340(12):937-44.<br />

86 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 87<br />

215. Trayner E, Jr., Celli BR. Postoperative pulmonary complications.<br />

Med Clin North Am 2001;85(5):1129-39.<br />

216. Weisman IM. Cardiopulmonary exercise testing in the preoperative<br />

assessment <strong>for</strong> lung resection surgery. Semin Thorac<br />

Cardiovasc Surg 2001;13(2):116-25.<br />

217. Bolliger CT, Perruchoud AP. Functional evaluation of the lung<br />

resection candidate. Eur Respir J 1998;11(1):198-212.<br />

218. Schuurmans MM, Diacon AH, Bolliger CT. Functional evaluation<br />

be<strong>for</strong>e lung resection. Clin Chest Med 2002;23(1):159-72.<br />

219. Celli BR, MacNee W. Standards <strong>for</strong> the diagnosis and treatment<br />

of patients with COPD: a summary of the ATS/ERS position<br />

paper. Eur Respir J 2004;23(6):932-46.<br />

220. Gallefoss F. The effects of patient education in COPD in a 1-<br />

year follow-up randomised, controlled trial. Patient Educ Couns.<br />

2004 Mar;52(3):259-66.<br />

221. Man WD, Mustfa N, Nikoletou D, Kaul S, Hart N, Rafferty GF,<br />

Donaldson N, Polkey MI, Moxham J. Effect of salmeterol on respiratory<br />

muscle activity during exercise in poorly reversible<br />

COPD. Thorax. 2004 Jun;59(6):471-6.<br />

222. O'Donnell DE, Fluge T, Gerken F, Hamilton A, Webb K, Aguilaniu<br />

B, Make B, Magnussen H. Effects of tiotropium on lung hyperinflation,<br />

dyspnoea and exercise tolerance in COPD. Eur Respir J.<br />

2004 Jun;23(6):832-40.<br />

223. Oostenbrink JB, Rutten-van Molken MP, Al MJ, Van Noord JA,<br />

Vincken W. One-year cost-effectiveness of tiotropium versus<br />

ipratropium to treat chronic obstructive pulmonary disease. Eur<br />

Respir J. 2004 Feb;23(2):241-9.<br />

224. Spencer S, Calverley PM, Burge PS, Jones PW. Impact of preventing<br />

exacerbations on deterioration of health status in COPD.<br />

Eur Respir J. 2004 May;23(5):698-702.<br />

225. Donohue JF, Kalberg C, Emmett A, Merchant K, Knobil K. A<br />

short-term comparison of fluticasone propionate/ salmeterol with<br />

ipratropium bromide/albuterol <strong>for</strong> the treatment of COPD. Treat<br />

Respir Med. 2004;3(3):173-81.<br />

226. Wongsurakiat P, Maranetra KN, Wasi C, Kositanont U,<br />

Dejsomritrutai W, Charoenratanakul S. Acute respiratory illness<br />

in patients with COPD and the effectiveness of influenza vaccination:<br />

a randomized controlled study. Chest. 2004<br />

Jun;125(6):2011-20.<br />

227. Li J, Zheng JP, Yuan JP, Zeng GQ, Zhong NS, Lin CY. Protective<br />

effect of a bacterial extract against acute exacerbation in patients<br />

with chronic bronchitis accompanied by chronic obstructive pulmonary<br />

disease. Chin Med J (Engl). 2004 Jun;117(6):828-34.<br />

MANAGEMENT OF COPD 87


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 88<br />

COMPONENT 4: MANAGE EXACERBATIONS<br />

KEY POINTS:<br />

• Exacerbations of respiratory symptoms requiring medical<br />

intervention are important clinical events in COPD.<br />

• The most common causes of an exacerbation are<br />

infection of the tracheobronchial tree and air pollution,<br />

but the cause of about one-third of severe exacerbations<br />

cannot be identified (Evidence B).<br />

• Inhaled bronchodilators (particularly inhaled ß 2 -<br />

agonists and/or anticholinergics), theophylline, and<br />

systemic, preferably oral, glucocorticosteroids are<br />

effective treatments <strong>for</strong> exacerbations of COPD<br />

(Evidence A).<br />

• Patients experiencing COPD exacerbations with<br />

clinical signs of airway infection (e.g., increased<br />

volume and change of color of sputum, and/or fever)<br />

may benefit from antibiotic treatment (Evidence B).<br />

• Noninvasive intermittent positive pressure ventilation<br />

(NIPPV) in exacerbations improves blood gases<br />

and pH, reduces in-hospital mortality, decreases the<br />

need <strong>for</strong> invasive mechanical ventilation and<br />

intubation, and decreases the length of hospital<br />

stay (Evidence A).<br />

INTRODUCTION<br />

COPD is often associated with exacerbations of symptoms<br />

1-4. In patients with Stage I: Mild COPD to Stage II:<br />

Moderate COPD, an exacerbation is associated with<br />

increased breathlessness, often accompanied by<br />

increased cough and sputum production, and may<br />

require medical attention outside of the hospital 5 . The<br />

need <strong>for</strong> medical intervention intensifies as the airflow<br />

limitation worsens. Exacerbations in Stage IV: Very<br />

Severe COPD are associated with acute respiratory failure,<br />

representing a significant burden on the health care<br />

system. Hospital mortality of patients admitted <strong>for</strong> an<br />

exacerbation of COPD is approximately 10%, and the<br />

long-term outcome is poor. Mortality reaches 40% in one<br />

year 6-9 , and is even higher (up to 59%) <strong>for</strong> patients older<br />

than 65 years 9 . These figures vary from country to country<br />

depending on the health care system and the availability<br />

of intensive care unit (ICU) beds.<br />

The most common causes of an exacerbation are infection<br />

of the tracheobronchial tree and air pollution 10 , but the<br />

cause of about one-third of severe exacerbations cannot<br />

be identified. The role of bacterial infections is controversial,<br />

but recent investigations with newer research techniques<br />

have begun to provide important in<strong>for</strong>mation.<br />

Bronchoscopic studies have shown that at least 50% of<br />

patients have bacteria in high concentrations in their lower<br />

airways during exacerbations 11 . However, a significant<br />

proportion of these patients also have bacteria colonizing<br />

their lower airways in the stable phase of the disease.<br />

There is some indication that the bacterial burden<br />

increases during exacerbations 11 , and that acquisition<br />

of strains of the bacteria that are new to the patient is<br />

associated with exacerbations 12 . Development of specific<br />

immune responses to the infecting bacterial strains, and<br />

the association of neutrophilic inflammation with bacterial<br />

exacerbations also support the bacterial causation of a<br />

proportion of exacerbations 13-16 . Conditions that may<br />

mimic an exacerbation include pneumonia, congestive<br />

heart failure, pneumothorax, pleural effusion, pulmonary<br />

embolism, and arrhythmia. Recommendations <strong>for</strong> use of<br />

antibiotics <strong>for</strong> COPD exacerbations are provided at the<br />

end of this chapter.<br />

DIAGNOSIS AND ASSESSMENT<br />

OF SEVERITY<br />

Medical History<br />

Increased breathlessness, the main symptom of an<br />

exacerbation, is often accompanied by wheezing and<br />

chest tightness, increased cough and sputum, change<br />

of the color and/or tenacity of sputum, and fever.<br />

Exacerbations may also be accompanied by a number<br />

of nonspecific complaints, such as malaise, insomnia,<br />

sleepiness, fatigue, depression, and confusion. A<br />

decrease in exercise tolerance, fever, and/or new radiological<br />

anomalies suggestive of pulmonary disease may<br />

herald a COPD exacerbation. An increase in sputum<br />

volume and purulence points to a bacterial cause, as<br />

does prior history of chronic sputum production 4,14 .<br />

Assessment of Severity<br />

Assessment of the severity of an exacerbation is based<br />

on the patient’s medical history be<strong>for</strong>e the exacerbation,<br />

symptoms, physical examination, lung function tests,<br />

arterial blood gas measurements, and other laboratory<br />

tests (Figure 5-4-1). Specific in<strong>for</strong>mation is required on<br />

88 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 89<br />

the frequency and severity of attacks of breathlessness<br />

and cough, sputum volume and color, and limitation<br />

of daily activities. When available, prior tests of lung<br />

function and arterial blood gases are extremely useful <strong>for</strong><br />

comparison with those made during the acute episode,<br />

as an acute change in these tests is more important than<br />

their absolute values. Thus, where possible, physicians<br />

should instruct their patients to bring the summary of<br />

their last evaluation when they come to the hospital with<br />

an exacerbation. In patients with very severe COPD, the<br />

most important sign of a severe exacerbation is a change<br />

in the alertness of the patient and this signals a need <strong>for</strong><br />

immediate evaluation in the hospital.<br />

<strong>Lung</strong> function tests. Even simple lung function tests<br />

can be difficult <strong>for</strong> a sick patient to per<strong>for</strong>m properly. In<br />

general, a PEF < 100 L/min or an FEV 1 < 1.00 L<br />

indicates a severe exacerbation 21-23 , except in patients<br />

with chronically severe airflow limitation. For instance,<br />

an FEV 1 of 0.75 L, or a PaO 2 /FiO 2 (FiO 2 = fractional<br />

concentration of oxygen in dry inspired gas) of 32 kPa<br />

(240 mm Hg) may be well tolerated by a subject with<br />

severe COPD who copes with these values in stable<br />

conditions, whereas they may reflect a severe<br />

exacerbation <strong>for</strong> a subject with slightly higher values, e.g.,<br />

an FEV 1 of 0.90 L or a PaO 2 /FiO 2 of 38 kPa (282 mm<br />

Hg) in stable conditions 24 .<br />

Figure 5-4-1. Medical History and Signs of<br />

Severity of Exacerbations of COPD<br />

Medical History<br />

• Duration of worsening or<br />

new symptoms.<br />

• Number of previous<br />

episodes<br />

(exacerbations/hospitalizations).<br />

• Present treatment regimen.<br />

Signs of Severity<br />

• Use of accessory respiratory<br />

muscles.<br />

• Paradoxical chest wall<br />

movements.<br />

• Worsening or new onset<br />

central cyanosis.<br />

• Development of peripheral<br />

edema.<br />

• Hemodynamic instability.<br />

• Signs of right heart failure.<br />

• Reduced alertness.<br />

Arterial blood gases. In the hospital, measurement of<br />

arterial blood gases is essential to assess the severity of<br />

an exacerbation. A PaO 2 < 8.0 kPa (60 mm Hg) and/or<br />

SaO 2 < 90% with or without PaCO 2 > 6.7 kPa, (50 mm Hg)<br />

when breathing room air indicate respiratory failure. In<br />

addition, a PaO 2 < 6.7 kPa (50 mm Hg), PaCO 2 > 9.3<br />

kPa (70 mm Hg), and pH < 7.30 point toward a life-threatening<br />

episode that needs critical management 25 .<br />

Chest X-ray and ECG. Chest radiographs<br />

(posterior/anterior plus lateral) are useful in identifying<br />

alternative diagnoses that can mimic the symptoms of an<br />

exacerbation. Although the history and physical signs<br />

can be confusing, especially when pulmonary hyperinflation<br />

masks coexisting cardiac signs, most problems are<br />

resolved by the chest X-ray and ECG. An ECG aids in<br />

the diagnosis of right heart hypertrophy, arrhythmias, and<br />

ischemic episodes. Pulmonary embolism can be very<br />

difficult to distinguish from an exacerbation, especially in<br />

severe COPD, because right ventricular hypertrophy and<br />

large pulmonary arteries lead to confusing ECG and<br />

radiographic results. Spiral CT scanning and angiography,<br />

and perhaps specific D-dimer assays, are the best<br />

tools presently available <strong>for</strong> the diagnosis of pulmonary<br />

embolism in patients with COPD, but ventilation-perfusion<br />

scanning is of no value. A low systolic blood pressure<br />

and an inability to increase the PaO 2 above 8.0 kPa<br />

(60 mm Hg) despite high-flow oxygen also suggest<br />

pulmonary embolism. If there are strong indications that<br />

pulmonary embolism has occurred, it is best to treat <strong>for</strong><br />

this along with the exacerbation.<br />

Other laboratory tests. The whole blood count may<br />

identify polycythemia (hematocrit > 55%) or bleeding.<br />

White blood cell counts are usually not very in<strong>for</strong>mative.<br />

The presence of purulent sputum during an exacerbation<br />

of symptoms is sufficient indication <strong>for</strong> starting empirical<br />

antibiotic treatment. Streptococcus pneumoniae,<br />

Hemophilis influenzae, and Moraxella catarrhalis are the<br />

most common bacterial pathogens involved in COPD<br />

exacerbations. If an infectious exacerbation does not<br />

respond to the initial antibiotic treatment, a sputum<br />

culture and an antibiogram should be per<strong>for</strong>med.<br />

Biochemical tests can reveal whether the cause of<br />

the exacerbation is an electrolyte disturbance(s)<br />

(hyponatremia, hypokalemia, etc.), a diabetic crisis, or<br />

poor nutrition (low proteins), and may suggest a<br />

metabolic acid-base disorder.<br />

HOME MANAGEMENT<br />

There is increasing interest in home care <strong>for</strong> end-stage<br />

COPD patients, although economic studies of home-care<br />

services have yielded mixed results. Four randomized<br />

clinical trials have shown nurse administered home care<br />

represents an effective and practical alternative to<br />

hospitalization in selected patients with exacerbations of<br />

COPD without acidotic respiratory failure. However, the<br />

exact criteria <strong>for</strong> home vs hospital treatment remains<br />

uncertain and will vary by health care setting 26-29 . A major<br />

outstanding issue is when to treat an exacerbation at<br />

home and when to hospitalize the patient.<br />

MANAGEMENT OF COPD 89


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 90<br />

Figure 5-4-2. Algorithm <strong>for</strong> the Management<br />

of an Exacerbation of COPD at Home<br />

Initiate or increase bronchodilator therapy<br />

Consider antibiotics<br />

Resolution or improvement<br />

of signs and symptoms<br />

Continue management<br />

Step down when possible<br />

Review long-term management<br />

The algorithm reported in Figure 5-4-2 may assist in<br />

the management of an exacerbation at home; a stepwise<br />

therapeutic approach is recommended 30-33 .<br />

Bronchodilator Therapy<br />

Home management of COPD exacerbations involves<br />

increasing the dose and/or frequency of existing<br />

bronchodilator therapy (Evidence A). If not already<br />

used, an anticholinergic can be added until the symptoms<br />

improve. In more severe cases, high-dose nebulizer<br />

therapy can be given on an as-needed basis <strong>for</strong> several<br />

days and if a suitable nebulizer is available. However,<br />

long-term use of nebulizer therapy after an acute episode<br />

is not routinely recommended.<br />

Glucocorticosteroids<br />

Reassess within hours<br />

No resolution or improvement<br />

Add oral corticosteroids<br />

Reassess within hours<br />

Worsening of signs/symptoms<br />

Refer to hospital<br />

Systemic glucocorticosteroids are beneficial in the<br />

management of exacerbations of COPD. They shorten<br />

recovery time and help to restore lung function more<br />

quickly 34-36 (Evidence A) and may reduce the risk of early<br />

relapse 73 . They should be considered in addition to<br />

bronchodilators if the patient’s baseline FEV 1 is < 50%<br />

predicted. A dose of 40 mg prednisolone per day <strong>for</strong> 10<br />

days is recommended (Evidence D). One large study<br />

indicates that nebulized budesonide may be an alternative<br />

to oral glucocorticosteroids in the treatment<br />

of nonacidotic exacerbations 37 .<br />

HOSPITAL MANAGEMENT<br />

The risk of dying from an exacerbation of COPD is<br />

closely related to the development of respiratory acidosis,<br />

the presence of significant comorbidities, and the need<br />

<strong>for</strong> ventilatory support 6 . Patients lacking these features<br />

are not at high risk of dying, but those with severe<br />

underlying COPD often require hospitalization in any case.<br />

Attempts at managing such patients entirely in the<br />

community have met with only limited success 38 , but<br />

returning them to their homes with increased social<br />

support and a supervised medical care package after<br />

initial emergency room assessment has been much more<br />

successful 39 . Several randomized controlled trials have<br />

confirmed that this is a safe alternative to hospitalization,<br />

although it probably only applies to about 25% of COPD<br />

admissions. Savings on inpatient expenditures 40 offset the<br />

additional costs of maintaining a community-based<br />

COPD nursing team. However, detailed cost-benefit<br />

analyses of these approaches are awaited.<br />

Figure 5-4-3. Indications <strong>for</strong> Hospital Assessment<br />

or Admission <strong>for</strong> Exacerbations of COPD*<br />

• Marked increase in intensity of symptoms,<br />

such as sudden development of resting dyspnea.<br />

• Severe background COPD.<br />

• Onset of new physical signs (e.g., cyanosis,<br />

peripheral edema).<br />

• Failure of exacerbation to respond to initial<br />

medical management.<br />

• Significant comorbidities.<br />

• Newly occurring arrhythmias.<br />

• Diagnostic uncertainty.<br />

• Older age.<br />

• Insufficient home support.<br />

Figure 5-4-4. Indications <strong>for</strong> ICU Admission of<br />

Patients with Exacerbations of COPD*<br />

• Severe dyspnea that responds inadequately to<br />

initial emergency therapy.<br />

• Confusion, lethargy, coma.<br />

• Persistent or worsening hypoxemia (PaO 2<br />

< 5.3 kPa, 40 mm Hg), and/or severe/worsening<br />

hypercapnia (PaCO 2 > 8.0 kPa, 60 mm Hg), and/or<br />

severe/worsening respiratory acidosis (pH < 7.25)<br />

despite supplemental oxygen and NIPPV.<br />

*Local resources need to be considered.<br />

90 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 91<br />

Figure 5-4-5. Management of Severe but<br />

Not Life-Threatening Exacerbations of COPD in the<br />

Emergency Department or the Hospital*<br />

• Assess severity of symptoms, blood gases, chest<br />

X-ray.<br />

• Administer controlled oxygen therapy and repeat<br />

arterial blood gas measurement after 30 minutes.<br />

• Bronchodilators:<br />

– Increase doses or frequency.<br />

– Combine ß 2 -agonists and anticholinergics.<br />

– Use spacers or air-driven nebulizers.<br />

– Consider adding intravenous aminophylline, if<br />

needed.<br />

• Add oral or intravenous glucocorticosteroids.<br />

• Consider antibiotics:<br />

– When signs of bacterial infection, oral or<br />

occasionally intravenous.<br />

• Consider noninvasive mechanical ventilation.<br />

• At all times:<br />

– Monitor fluid balance and nutrition.<br />

– Consider subcutaneous heparin.<br />

– Identify and treat associated conditions<br />

(e.g., heart failure, arrhythmias).<br />

– Closely monitor condition of the patient.<br />

*Local resources need to be considered.<br />

A range of criteria to consider <strong>for</strong> hospital assessment/<br />

admission <strong>for</strong> exacerbations of COPD are shown in<br />

Figure 5-4-3. Some patients need immediate admission<br />

to an intensive care unit (ICU) (Figure 5-4-4). Admission<br />

of patients with severe COPD exacerbations to intermediate<br />

or special respiratory care units may be appropriate if<br />

personnel, skills, and equipment exist to identify and<br />

manage acute respiratory failure successfully.<br />

Emergency Department or Hospital<br />

The first actions when a patient reaches the emergency<br />

department are to provide controlled oxygen therapy<br />

and to determine whether the exacerbation is life<br />

threatening. If so, the patient should be admitted to the<br />

ICU immediately. Otherwise, the patient may be<br />

managed in the emergency department or hospital as<br />

detailed in Figure 5-4-5.<br />

Controlled oxygen therapy. Oxygen therapy is the<br />

cornerstone of hospital treatment of COPD exacerbations.<br />

Adequate levels of oxygenation (PaO 2 > 8.0 kPa,<br />

60 mm Hg, or SaO 2 > 90%) are easy to achieve in<br />

uncomplicated exacerbations, but CO 2 retention can<br />

occur insidiously with little change in symptoms. Once<br />

oxygen is started, arterial blood gases should be checked<br />

30 minutes later to ensure satisfactory oxygenation<br />

without CO 2 retention or acidosis. Venturi masks are<br />

more accurate sources of controlled oxygen than are<br />

nasal prongs but are more likely to be removed by the<br />

patient.<br />

Bronchodilator therapy. Short-acting inhaled<br />

2 -agonists are usually the preferred bronchodilators <strong>for</strong><br />

treatment of exacerbations of COPD 30,31,41 (Evidence A). If<br />

a prompt response to these drugs does not occur, the<br />

addition of an anticholinergic is recommended, even<br />

though evidence concerning the effectiveness of this<br />

combination is controversial. Despite its wide-spread<br />

clinical use, the role of aminophylline in the treatment of<br />

exacerbations of COPD remains controversial. Most<br />

studies of aminophylline have demonstrated minor<br />

improvements in lung volumes without showing gas<br />

exchange deterioration 42,43 . In more severe exacerbations,<br />

addition of an oral or intravenous methylxanthine to the<br />

treatment can be considered. However, close monitoring<br />

of serum theophylline is recommended to avoid the side<br />

effects of these drugs 42,44-46 . Possible beneficial effects in<br />

lung function, and clinical endpoints, are modest and<br />

inconsistent, whereas adverse effects are significantly<br />

increased 74 .<br />

Glucocorticosteroids. Oral or intravenous glucocorticosteroids<br />

are recommended as an addition to<br />

bronchodilator therapy (plus eventually antibiotics<br />

and oxygen therapy) in the hospital management of<br />

exacerbations of COPD 35-36 (Evidence A). The exact<br />

dose that should be recommended is not known, but high<br />

doses are associated with a significant risk of side effects.<br />

Thirty to 40 mg of oral prednisolone daily <strong>for</strong> 10 to 14<br />

days is a reasonable compromise between efficacy and<br />

safety (Evidence D). Prolonged treatment does not result<br />

in greater efficacy and increases the risk of side effects.<br />

Ventilatory support. The primary objectives of<br />

mechanical support in patients with exacerbations in<br />

Stage IV: Very Severe COPD are to decrease mortality<br />

and morbidity and to relieve symptoms. Ventilatory<br />

support includes both noninvasive mechanical ventilation<br />

using either negative or positive pressure devices, and<br />

invasive (conventional) mechanical ventilation by<br />

oro/naso-tracheal tube or tracheostomy.<br />

MANAGEMENT OF COPD 91


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 92<br />

Figure 5-4-6. Indications and Relative<br />

Contraindications <strong>for</strong> NIPPV 47,56<br />

Selection criteria<br />

• Moderate to severe dyspnea with use of accessory<br />

muscles and paradoxical abdominal motion.<br />

• Moderate to severe acidosis (pH ≤ 7.35) and<br />

hypercapnia (PaCO 2 > 6.0 kPa, 45 mm Hg) 57 .<br />

• Respiratory frequency > 25 breaths per minute.<br />

Exclusion criteria (any may be present)<br />

• Respiratory arrest.<br />

• Cardiovascular instability (hypotension,<br />

arrhythmias, myocardial infarction).<br />

• Somnolence, impaired mental status,<br />

uncooperative patient.<br />

• High aspiration risk; viscous or copious secretions.<br />

• Recent facial or gastroesophageal surgery.<br />

• Craniofacial trauma, fixed nasopharyngeal<br />

abnormalities.<br />

• Burns.<br />

• Extreme obesity.<br />

Noninvasive mechanical ventilation. Noninvasive<br />

intermittent positive pressure ventilation (NIPPV) has<br />

been studied in many uncontrolled and five randomized<br />

controlled trials in acute respiratory failure 47,48 . The<br />

studies show consistently positive results with success<br />

rates of 80-85% 49 . Taken together they provide evidence<br />

that NIPPV increases pH, reduces PaCO 2 , reduces<br />

the severity of breathlessness in the first 4 hours of<br />

treatment, and decreases the length of hospital stay<br />

(Evidence A). More importantly, mortality - or its surrogate,<br />

intubation rate - is reduced by this intervention 50-53 .<br />

However, NIPPV is not appropriate <strong>for</strong> all patients, as<br />

summarized in Figure 5-4-6 49 .<br />

Invasive (conventional) mechanical ventilation. During<br />

exacerbations of COPD the events occurring within the<br />

lungs include bronchoconstriction, airway inflammation,<br />

increased mucous secretions, and loss of elastic recoil,<br />

all of which prevent the respiratory system from reaching<br />

its passive functional residual capacity at the end of<br />

expiration, enhancing dynamic hyperinflation 54 . As a<br />

result of these processes, an elastic threshold load,<br />

referred to as intrinsic or auto-positive end-expiratory<br />

pressure (PEEPi), is imposed on the inspiratory muscles<br />

at the beginning of inspiration and increases the work of<br />

breathing. For these reasons, patients who show<br />

impending acute respiratory failure and those with<br />

Figure 5-4-7. Indications <strong>for</strong><br />

Invasive Mechanical Ventilation<br />

• Severe dyspnea with use of accessory muscles<br />

and paradoxical abdominal motion.<br />

• Respiratory frequency > 35 breaths per minute.<br />

• Life-threatening hypoxemia (PaO 2 < 5.3 kPa,<br />

40 mm Hg or PaO 2 /FiO 2 < 200 mm Hg).<br />

• Severe acidosis (pH < 7.25) and hypercapnia<br />

(PaCO 2 > 8.0 kPa, 60 mm Hg).<br />

• Respiratory arrest.<br />

• Somnolence, impaired mental status.<br />

• Cardiovascular complications (hypotension,<br />

shock, heart failure).<br />

• Other complications (metabolic abnormalities,<br />

sepsis, pneumonia, pulmonary embolism,<br />

barotrauma, massive pleural effusion).<br />

• NIPPV failure (or exclusion criteria,<br />

see Figure 5-4-7).<br />

FiO 2 : Fractional concentration of oxygen in dry inspired gas.<br />

Figure 5-4-8. Factors Determining<br />

Benefit from Invasive Ventilation<br />

• Cultural attitudes toward chronic disability.<br />

• Expectations of therapy.<br />

• Financial resources (especially the provision<br />

of ICU facilities).<br />

• Perceived likelihood of recovery.<br />

• Customary medical practice.<br />

• Wishes, if known, of the patient.<br />

life-threatening acid-base status abnormalities and/or<br />

altered mental status despite aggressive pharmacologic<br />

therapy are likely to be the best candidates <strong>for</strong> invasive<br />

(conventional) mechanical ventilation. The indications <strong>for</strong><br />

initiating mechanical ventilation during exacerbations of<br />

COPD are shown in Figure 5-4-7, the first being the<br />

commonest and most important reason. Figure 5-4-8<br />

details the factors determining benefit from invasive<br />

ventilation. The three ventilatory modes most widely<br />

used are assisted-control ventilation, and pressure<br />

support ventilation alone or in combination with intermittent<br />

mandatory ventilation 55 .<br />

The use of invasive ventilation in end-stage COPD<br />

patients is influenced by the likely reversibility of the<br />

precipitating event, the patient’s wishes, and the availability<br />

92 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 93<br />

of intensive care facilities. Major hazards include the risk<br />

of ventilator-acquired pneumonia (especially when<br />

multi-resistant organisms are prevalent), barotrauma,<br />

and failure to wean to spontaneous ventilation. Contrary<br />

to some opinions, mortality among COPD patients with<br />

respiratory failure is no greater than mortality among<br />

patients ventilated <strong>for</strong> non-COPD causes.<br />

A review of a large number of North American COPD<br />

patients ventilated <strong>for</strong> respiratory failure indicated an in<br />

hospital mortality of 17-30% 58 . Further attrition over the<br />

next 12 months was particularly high among those<br />

patients who had poor lung function be<strong>for</strong>e ventilation<br />

(FEV 1 < 30% predicted), had a non-respiratory comorbidity,<br />

or were housebound. Patients who did not have a<br />

previously diagnosed underlying disease, had respiratory<br />

failure due to a potentially reversible cause (such as an<br />

infection), or were relatively mobile and not using longterm<br />

oxygen did surprisingly well with ventilatory support.<br />

When possible, a clear statement of the patient’s own<br />

treatment wishes - an advance directive or “living will” -<br />

makes these difficult decisions much easier to resolve.<br />

Weaning or discontinuation from mechanical ventilation<br />

can be particularly difficult and hazardous in patients with<br />

COPD. The most influential determinant of mechanical<br />

ventilatory dependency in these patients is the balance<br />

between the respiratory load and the capacity of the<br />

respiratory muscles to cope with this load 59 . By contrast,<br />

pulmonary gas exchange by itself is not a major difficulty<br />

in patients with COPD 60-62 . Weaning patients from the<br />

ventilator can be a very difficult and prolonged process<br />

and the best method remains a matter of debate 63,64 .<br />

Whether pressure support or a T-piece trial is used,<br />

weaning is shortened when a clinical protocol is adopted<br />

(Evidence A). Noninvasive ventilation has been applied<br />

to facilitate the weaning process in COPD patients with<br />

acute or chronic respiratory failure 58 . Compared with<br />

invasive pressure support ventilation, noninvasive<br />

intermittent positive pressure ventilation (NIPPV) during<br />

weaning shortened weaning time, reduced the stay in<br />

the intensive care unit, decreased the incidence of<br />

nosocomial pneumonia, and improved 60-day survival<br />

rates. Similar findings have been reported when NIPPV is<br />

used after extubation <strong>for</strong> hypercapnic respiratory failure 65<br />

(Evidence C).<br />

Other measures. Further treatments that can be used in<br />

the hospital include: fluid administration (accurate monitoring<br />

of fluid balance is essential); nutrition (supplementary<br />

when the patient is too dyspneic to eat); low molecular<br />

heparin in immobilized, polycythemic, or dehydrated<br />

patients with or without a history of thromboembolic<br />

disease; and sputum clearance (by stimulating coughing<br />

and low-volume <strong>for</strong>ced expirations as in home management).<br />

Manual or mechanical chest percussion and<br />

postural drainage may be beneficial in patients producing<br />

> 25 ml sputum per day or with lobar atelectasis.<br />

Hospital Discharge and Follow-Up<br />

Insufficient clinical data exist to establish the optimal<br />

duration of hospitalization in individual patients<br />

developing an exacerbation of COPD 1,66,67 . Consensus<br />

and limited data support the discharge criteria listed in<br />

Figure 5-4-9. Figure 5-4-10 provides items to include in<br />

a follow-up assessment 4 to 6 weeks after discharge from<br />

the hospital. Thereafter, follow-up is the same as <strong>for</strong><br />

stable COPD, including supervising smoking cessation,<br />

monitoring the effectiveness of each drug treatment, and<br />

monitoring changes in spirometric parameters 39 . Home<br />

visits by a community nurse may permit earlier discharge<br />

of patients hospitalized with an exacerbation of COPD,<br />

without increasing readmission rate 29,68-70 . Early outpatient<br />

pulmonary rehabilitation after hospitalization <strong>for</strong> COPD<br />

exacerbation results in exercise capacity and health<br />

status improvements at three months 75 .<br />

If hypoxemia developed during the exacerbation, arterial<br />

blood gases should be rechecked at discharge and at<br />

the follow-up visit. If the patient remains hypoxemic,<br />

long-term oxygen therapy should be instituted. Decisions<br />

about suitability <strong>for</strong> continuous domiciliary oxygen based<br />

on the severity of the acute hypoxemia during an<br />

exacerbation are frequently misleading.<br />

The opportunities <strong>for</strong> prevention of future exacerbations<br />

should be reviewed be<strong>for</strong>e discharge, with particular<br />

Figure 5-4-9. Discharge Criteria <strong>for</strong><br />

Patients with Exacerbations of COPD<br />

• Inhaled ß 2 -agonist therapy is required no more<br />

frequently than every 4 hrs.<br />

• Patient, if previously ambulatory, is able to walk<br />

across room.<br />

• Patient is able to eat and sleep without frequent<br />

awakening by dyspnea.<br />

• Patient has been clinically stable <strong>for</strong> 12-24 hrs.<br />

• Arterial blood gases have been stable <strong>for</strong> 12-24 hrs.<br />

• Patient (or home caregiver) fully understands correct<br />

use of medications.<br />

• Follow-up and home care arrangements have been<br />

completed (e.g., visiting nurse, oxygen delivery, meal<br />

provisions).<br />

• Patient, family, and physician are confident patient<br />

can manage successfully.<br />

MANAGEMENT OF COPD 93


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 94<br />

Figure 5-4-10. Follow-Up Assessment 4-6 Weeks<br />

After Discharge from Hospital <strong>for</strong><br />

Exacerbations of COPD<br />

• Ability to cope in usual environment.<br />

• Measurement of FEV 1 .<br />

• Reassessment of inhaler technique.<br />

• Understanding of recommended treatment regimen.<br />

• Need <strong>for</strong> long-term oxygen therapy and/or home<br />

nebulizer (<strong>for</strong> patients with very severe COPD).<br />

attention to future influenza vaccination plans, knowledge<br />

about current therapy including inhaler technique 71,72 , and<br />

how to recognize symptoms of exacerbations.<br />

Pharmacotherapy known to reduce the number of<br />

exacerbations should be considered. Social problems<br />

should be discussed and principal caregivers identified if<br />

the patient has a significant persisting disability.<br />

Antibiotics<br />

Randomised placebo controlled studies of antibiotic<br />

treatment in exacerbations of COPD have demonstrated<br />

a small beneficial effect of antibiotics on lung function 76 ,<br />

and one randomised controlled trial has provided evidence<br />

<strong>for</strong> a significant beneficial effect of antibiotics in COPD<br />

patients who presented with an increase in all three of the<br />

following cardinal symptoms: dyspnea, sputum volume,<br />

sputum purulence 77 . There was also some benefit in<br />

those patients with an increase in only two of these cardinal<br />

symptoms.<br />

A study on non-hospitalized patients with exacerbations<br />

of COPD showed a relationship between the purulence of<br />

the sputum and the presence of bacteria 78 , suggesting<br />

that these patients should be treated with antibiotics if<br />

they also have at least one of the other two cardinal<br />

symptoms (dyspnea or sputum volume). However,<br />

these criteria <strong>for</strong> exacerbations of COPD have not been<br />

validated in other studies. A study in COPD patients<br />

with exacerbations requiring mechanical ventilation<br />

(invasive and non-invasive) indicated that not giving<br />

antibiotics was associated with increased mortality and<br />

a greater incidence of secondary intra-hospital pneumonia 79 .<br />

• Patients with exacerbations of COPD with two of the<br />

cardinal symptoms, if increased purulence of sputum<br />

is one of the two symptoms. (Evidence C)<br />

• Patients with a severe exacerbation of COPD that<br />

requires invasive mechanical ventilation (invasive and<br />

non-invasive). (Evidence B)<br />

The predominant bacterial organisms recovered in the<br />

lower airways of patients with mild exacerbations are<br />

H. influenzae, S. pneumoniae and M. catarrhalis 11,80 .<br />

In contrast, studies in patients requiring mechanical<br />

ventilation with severe underlying COPD 81,82 have shown<br />

that other microorganisms, such as enteric gram negative<br />

bacilli and P. aeruginosa may be more frequent. Other<br />

studies have shown that the severity of the COPD is an<br />

important determinant of the type of microorganism 83,84 .<br />

In patients with mild COPD, S. pneumoniae is predominant.<br />

When the FEV 1 is lower, H. influenzae and M. catarrhalis<br />

are more frequent and P. aeruginosa may appear in<br />

patients with a more severe degree of airways obstruction<br />

(Figure 5-4-11). The risk factors <strong>for</strong> P. aeruginosa infection<br />

are recent hospitalisation, frequent administration of antibiotics<br />

(4 courses in the last year, very severe COPD (Stage IV),<br />

and isolation of P. aeruginosa during a previous<br />

exacerbation or colonization during a stable period 83,84 .<br />

There is no clear in<strong>for</strong>mation about when to use oral or<br />

intravenous route of administration in hospitalized<br />

patients. The route of administration depends on the<br />

ability of the patient to eat, and the pharmacokinetics of<br />

the antibiotic. The oral route is preferred. Otherwise,<br />

the IV route has to be used, switching to oral when there<br />

is clinical stabilization. Antibiotic treatment in patients<br />

with exacerbations of COPD should be maintained <strong>for</strong> 3<br />

to 10 days. Figure 5-4-12 provides recommended<br />

antibiotic treatment in exacerbations of COPD.<br />

Ten to thirty percent of COPD exacerbated patients do<br />

not respond to empiric antimicrobial treatment 80 . In such<br />

cases the patient should be re-evaluated <strong>for</strong> complications<br />

that can aggravate symptoms and mimic exacerbations<br />

(e.g., cardiac failure, pulmonary embolism, non-compliance<br />

with prescribed medications); microbiological reassessment<br />

of these patients is recommended.<br />

Based on the current available evidence, antibiotics<br />

should be given to:<br />

• Patients with exacerbations of COPD with three of<br />

the following cardinal symptoms: increased dyspnea,<br />

increased sputum volume, increased sputum purulence.<br />

(Evidence B)<br />

94 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 95<br />

Figure 5-4-11: Stratification of patients with COPD exacerbated <strong>for</strong> antibiotic<br />

treatment and potential microorganisms involved in each group<br />

Group a Definition b Microorganisms<br />

Group A: Patients not requiring<br />

hospitalization (Stage I: Mild COPD)<br />

Mild exacerbation<br />

H. influenzae<br />

S. pneumoniae<br />

M. catarrhalis<br />

Chlamydia pneumoniae c<br />

Viruses<br />

Group B: Patients admitted to hospital<br />

(Stages II-IV: Moderate to Very Severe<br />

COPD)<br />

Group C: Patients admitted to hospital<br />

(Stages II-IV: Moderate to Very Severe<br />

COPD)<br />

Moderate-severe exacerbation without<br />

risk factors <strong>for</strong> P. aeruginosa infection<br />

Moderate-severe exacerbation with risk<br />

factors <strong>for</strong> P. aeruginosa infection<br />

Group A plus:<br />

Enterobacteriaceae (K.pneumoniae,<br />

E. coli, Proteus, Enterobacter, etc)<br />

Group B plus:<br />

P. aeruginosa<br />

a. In some settings, patients with moderate to severe exacerbations may be treated as outpatients. In this case, patients may best be stratified into<br />

two groups: an uncomplicated group without any risk factors and a complicated group that has one or more ‘risk factors’ (co-morbidity, severe COPD,<br />

frequent exacerbations, antimicrobial use within last 3 months). The uncomplicated group: use Group A recommendations Figure 5-4-12.<br />

Complicated group: use Group B or C recommendations (oral therapy) Figure 5-4-12 217-19 .<br />

b. Severity refers to the exacerbation, though this is intertwined with the severity of the underlying COPD.<br />

c. Chlamydia pneumonia (or Chlamidophila pneumoniae) has not been confirmed as a cause of exacerbations in some areas (e.g., UK).<br />

Group A<br />

Group B<br />

Group C<br />

Oral Treatment<br />

(No particular order)<br />

Figure 5-4-12: Antibiotic treatment in exacerbations of COPD a,b<br />

Patients with only one cardinal symptom<br />

should not receive antibiotics<br />

If indication then:<br />

• ß-lactam (Ampicillin/Amoxicillin c )<br />

• Tetracycline<br />

• Trimethoprim/Sulfamethoxazole<br />

• ß-lactam/b-lactamase inhibitor<br />

(Co-amoxiclav)<br />

• Fluoroquinolones (Ciprofloxacin,<br />

Levofloxacin - high dose e )<br />

Alternative<br />

(No particular order)<br />

• ß-lactam/ß-lactamase inhibitor<br />

(Co-amoxiclav)<br />

• Macrolides (Azithromycin,<br />

Clarithromycin, Roxithromycin d )<br />

• Cephalosporins - 2nd or 3rd generation<br />

• Ketolides (Telithromycin)<br />

• Fluoroquinolones d (Gatifloxacin,<br />

Gemifloxacin, Levofloxacin,<br />

Moxifloxacin)<br />

Parental Treatment<br />

(No particular order)<br />

• ß-lactam/b-lactamase inhibitor<br />

(Co-amoxiclav, ampicillin/sulbactam)<br />

• Cephalosporins - 2nd or<br />

3rd generation<br />

• Fluoroquinolones d (Gatifloxacin,<br />

Levofloxacin, Moxifloxacin)<br />

• Fluoroquinolones (Ciprofloxacin,<br />

Levofloxacin - high dose e ) or<br />

• ß-lactam with P.aeruginosa activity<br />

a. All patients with symptoms of a COPD exacerbation should be treated with additional bronchodilators ± glucocorticosteroids.<br />

b. Classes of antibiotics are provided (with specific agents in parentheses). In countries with high incidence of S. pneumoniae resistant to penicillin,<br />

high dosages of Amoxicillin or Co-Amoxiclav are recommended. (See Table 1 <strong>for</strong> definition of Groups A, B, C.)<br />

c. This antibiotic is not appropriate in areas where there is increased prevalence of ß-lactamase producing H. influenzae and M. catarrhalis and/or of<br />

S. pneumoniae resistant to penicillin.<br />

d. Not available in all areas of the world.<br />

e. Dose 750 mgs effective against P. aeruginosa.<br />

MANAGEMENT OF COPD 95


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 96<br />

REFERENCES<br />

1. Regueiro CR, Hamel MB, Davis RB, Desbiens N, Connors<br />

AF, Jr., Phillips RS. A comparison of generalist and pulmonologist<br />

care <strong>for</strong> patients hospitalized with severe chronic<br />

obstructive pulmonary disease: resource intensity, hospital<br />

costs, and survival. SUPPORT Investigators. Study to<br />

Understand Prognoses and Preferences <strong>for</strong> Outcomes and<br />

Risks of Treatment. Am J Med 1998; 105:366-72.<br />

2. Gibson PG, Wlodarczyk JH, Wilson AJ, Sprogis A. Severe<br />

exacerbation of chronic obstructive airways disease: health<br />

resource use in general practice and hospital. J Qual Clin<br />

Pract 1998; 18:125-33.<br />

3. Warren PM, Flenley DC, Millar JS, Avery A. Respiratory<br />

failure revisited: acute exacerbations of chronic bronchitis<br />

between 1961-68 and 1970-76. Lancet 1980; 1:467-70.<br />

4. Anthonisen NR, Manfreda J, Warren CP, Hershfield ES,<br />

Harding GK, Nelson NA. Antibiotic therapy in exacerbations<br />

of chronic obstructive pulmonary disease. Ann Intern<br />

Med 1987; 106:196-204.<br />

5. Thompson AB, Mueller MB, Heires AJ, Bohling TL,<br />

Daughton D, Yancey SW, et al. Aerosolized beclomethasone<br />

in chronic bronchitis. Improved pulmonary function<br />

and diminished airway inflammation. Am Rev Respir Dis<br />

1992; 146:389-95.<br />

6. Connors AF, Jr., Dawson NV, Thomas C, Harrell FE, Jr.,<br />

Desbiens N, Fulkerson WJ, et al. Outcomes following acute<br />

exacerbation of severe chronic obstructive lung disease.<br />

The SUPPORT investigators (Study to Understand<br />

Prognoses and Preferences <strong>for</strong> Outcomes and Risks of<br />

Treatments). Am J Respir Crit Care Med 1996; 154:959-67.<br />

7. Kong GK, Belman MJ, Weingarten S. Reducing length of<br />

stay <strong>for</strong> patients hospitalized with exacerbation of COPD<br />

by using a practice guideline. Chest 1997; 111:89-94.<br />

8. Fuso L, Incalzi RA, Pistelli R, Muzzolon R, Valente S,<br />

Pagliari G, et al. Predicting mortality of patients hospitalized<br />

<strong>for</strong> acutely exacerbated chronic obstructive pulmonary<br />

disease. Am J Med 1995; 98:272-7.<br />

9. Seneff MG, Wagner DP, Wagner RP, Zimmerman JE,<br />

Knaus WA. Hospital and 1-year survival of patients admitted<br />

to intensive care units with acute exacerbation of chronic<br />

obstructive pulmonary disease. JAMA 1995; 274:1852-7.<br />

10. White AJ, Gompertz S, Stockley RA. <strong>Chronic</strong> obstructive<br />

pulmonary disease . 6: The aetiology of exacerbations of<br />

chronic obstructive pulmonary disease. Thorax 2003;<br />

58(1):73-80.<br />

11. Monso E, Ruiz J, Rosell A, Manterola J, Fiz J, Morera J,<br />

Ausina V. Bacterial infection in chronic obstructive pulmonary<br />

disease. A study of stable and exacerbated outpatients<br />

using the protected specimen brush. Am J Respir<br />

Crit Care Med 1995; 152(4 Pt 1):1316-20.<br />

12. Sethi S, Evans N, Grant BJ, Murphy TF. New strains of<br />

bacteria and exacerbations of chronic obstructive pulmonary<br />

disease. N Engl J Med 2002; 347(7):465-71.<br />

13. Sethi S, Wrona C, Grant BJ, Murphy TF. Strain-specific<br />

immune response to Haemophilus influenzae in chronic<br />

obstructive pulmonary disease. Am J Respir Crit Care Med<br />

2004; 169(4):448-53.<br />

14. Murphy TF, Brauer AL, Grant BJ, Sethi S. Moraxella<br />

catarrhalis in <strong>Chronic</strong> <strong>Obstructive</strong> Pulmonary <strong>Disease</strong>:<br />

Burden of <strong>Disease</strong> and Immune Response. Am J Respir<br />

Crit Care Med 2005; 172(2):195-9.<br />

15. Sethi S, Muscarella K, Evans N, Klingman KL, Grant BJ,<br />

Murphy TF. Airway inflammation and etiology of acute<br />

exacerbations of chronic bronchitis. Chest 2000;<br />

118(6):1557-65.<br />

16. White AJ, Gompertz S, Bayley DL, Hill SL, O'Brien C,<br />

Unsal I, Stockley RA. Resolution of bronchial inflammation<br />

is related to bacterial eradication following treatment of<br />

exacerbations of chronic bronchitis. Thorax 2003; 58(8):680-5.<br />

References 17 - 20 have been deleted.<br />

21. Emerman CL, Effron D, Lukens TW. Spirometric criteria <strong>for</strong><br />

hospital admission of patients with acute exacerbation of<br />

COPD. Chest 1991; 99:595-9.<br />

22. Emerman CL, Lukens TW, Effron D. Physician estimation<br />

of FEV1 in acute exacerbation of COPD. Chest 1994;<br />

105:1709-12.<br />

23. Emerman CL, Cydulka RK. Use of peak expiratory flow<br />

rate in emergency department evaluation of acute exacerbation<br />

of chronic obstructive pulmonary disease. Ann<br />

Emerg Med 1996; 27:159-63.<br />

24. Barbera JA, Roca J, Ferrer A, Felez MA, Diaz O, Roger N,<br />

et al. Mechanisms of worsening gas exchange during<br />

acute exacerbations of chronic obstructive pulmonary disease.<br />

Eur Respir J 1997; 10:1285-91.<br />

25. Emerman CL, Connors AF, Lukens TW, Effron D, May ME.<br />

Relationship between arterial blood gases and spirometry<br />

in acute exacerbations of chronic obstructive pulmonary<br />

disease. Ann Emerg Med 1989; 18:523-7.<br />

26. Davies L, Wilkinson M, Bonner S, Calverley PM, Angus<br />

RM. "Hospital at home" versus hospital care in patients<br />

with exacerbations of chronic obstructive pulmonary disease:<br />

prospective randomised controlled trial. BMJ 2000;<br />

321:1265-8.<br />

27. Ojoo JC, Moon T, McGlone S, Martin K, Gardiner ED,<br />

Greenstone MA, et al. Patients' and carers' preferences in<br />

two models of care <strong>for</strong> acute exacerbations of COPD:<br />

results of a randomised controlled trial. Thorax 2002;<br />

57:167-9.<br />

28. Skwarska E, Cohen G, Skwarski KM, Lamb C, Bushell D,<br />

Parker S, et al. Randomized controlled trial of supported<br />

discharge in patients with exacerbations of chronic<br />

obstructive pulmonary disease. Thorax 2000; 55:907-12.<br />

29. Hernandez C, Casas A, Escarrabill J, Alonso J, Puig-Junoy<br />

J, Farrero E, et al. Home hospitalisation of exacerbated<br />

chronic obstructive pulmonary disease patients. Eur Respir<br />

J 2003; 21:58-67.<br />

30. Siafakas NM, Vermeire P, Pride NB, Paoletti P, Gibson J,<br />

Howard P, et al. Optimal assessment and management of<br />

chronic obstructive pulmonary disease (COPD). The<br />

European Respiratory Society Task Force. Eur Respir J<br />

1995; 8:1398-420.<br />

31. American Thoracic Society. Standards <strong>for</strong> the diagnosis<br />

and care of patients with chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med 1995; 152:S77-121.<br />

32. Sachs FL. <strong>Chronic</strong> bronchitis. Clin Chest Med 1981; 2:79-89.<br />

96 MANAGEMENT OF COPD


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 97<br />

33. Celli BR. Current thoughts regarding treatment of chronic<br />

obstructive pulmonary disease. Med Clin North Am 1996;<br />

80:589-609.<br />

34. Thompson WH, Nielson CP, Carvalho P, Charan NB,<br />

Crowley JJ. Controlled trial of oral prednisone in outpatients<br />

with acute COPD exacerbation. Am J Respir Crit<br />

Care Med 1996; 154:407-12.<br />

35. Davies L, Angus RM, Calverley PM. Oral corticosteroids in<br />

patients admitted to hospital with exacerbations of chronic<br />

obstructive pulmonary disease: a prospective randomised<br />

controlled trial. Lancet 1999; 354:456-60.<br />

36. Niewoehner DE, Erbland ML, Deupree RH, Collins D,<br />

Gross NJ, Light RW, et al. Effect of systemic glucocorticoids<br />

on exacerbations of chronic obstructive pulmonary<br />

disease. Department of Veterans Affairs Cooperative Study<br />

Group. N Engl J Med 1999; 340:1941-7.<br />

37. Maltais F, Ostinelli J, Bourbeau J, Tonnel AB, Jacquemet<br />

N, Haddon J, et al. Comparison of nebulized budesonide<br />

and oral prednisolone with placebo in the treatment of<br />

acute exacerbations of chronic obstructive pulmonary disease:<br />

a randomized controlled trial. Am J Respir Crit Care<br />

Med 2002; 165:698-703.<br />

38. Shepperd S, Harwood D, Jenkinson C, Gray A, Vessey M,<br />

Morgan P. Randomised controlled trial comparing hospital<br />

at home care with inpatient hospital care. I: three month<br />

follow up of health outcomes. BMJ 1998; 316:1786-91.<br />

39. Gravil JH, Al-Rawas OA, Cotton MM, Flanigan U, Irwin A,<br />

Stevenson RD. Home treatment of exacerbations of chronic<br />

obstructive pulmonary disease by an acute respiratory<br />

assessment service. Lancet 1998; 351:1853-5.<br />

40. Soderstrom L, Tousignant P, Kaufman T. The health and<br />

cost effects of substituting home care <strong>for</strong> inpatient acute<br />

care: a review of the evidence. CMAJ 1999; 160:1151-5.<br />

41. The COPD Guidelines Group of the Standards of Care<br />

Committee of the BTS. BTS guidelines <strong>for</strong> the management<br />

of chronic obstructive pulmonary disease. Thorax<br />

1997; 52 Suppl 5:S1-28.<br />

42. Barbera JA, Reyes A, Roca J, Montserrat JM, Wagner PD,<br />

Rodriguez-Roisin R. Effect of intravenously administered<br />

aminophylline on ventilation/perfusion inequality during<br />

recovery from exacerbations of chronic obstructive pulmonary<br />

disease. Am Rev Respir Dis 1992; 145:1328-33.<br />

43. Mahon JL, Laupacis A, Hodder RV, McKim DA, Paterson<br />

NA, Wood TE, et al. Theophylline <strong>for</strong> irreversible chronic<br />

airflow limitation: a randomized study comparing n of 1 trials<br />

to standard practice. Chest 1999; 115:38-48.<br />

44. Lloberes P, Ramis L, Montserrat JM, Serra J, Campistol J,<br />

Picado C, et al. Effect of three different bronchodilators<br />

during an exacerbation of chronic obstructive pulmonary<br />

disease. Eur Respir J 1988; 1:536-9.<br />

45. Murciano D, Aubier M, Lecocguic Y, Pariente R. Effects of<br />

theophylline on diaphragmatic strength and fatigue in<br />

patients with chronic obstructive pulmonary disease. N<br />

Engl J Med 1984; 311:349-53.<br />

46. Emerman CL, Connors AF, Lukens TW, May ME, Effron D.<br />

Theophylline concentrations in patients with acute exacerbation<br />

of COPD. Am J Emerg Med 1990; 8:289-92.<br />

47. Lightowler JV, Wedzicha JA, Elliott MW, Ram FS. Non-invasive<br />

positive pressure ventilation to treat respiratory failure<br />

resulting from exacerbations of chronic obstructive pulmonary<br />

disease: Cochrane systematic review and metaanalysis.<br />

Bmj 2003; 326:185.<br />

48. Meyer TJ, Hill NS. Noninvasive positive pressure ventilation<br />

to treat respiratory failure. Ann Intern Med 1994; 120:760-70.<br />

49. Consensus conference report. Clinical indications <strong>for</strong> noninvasive<br />

positive pressure ventilation in chronic respiratory<br />

failure due to restrictive lung disease, COPD, and nocturnal<br />

hypoventilation. Chest 1999; 116:521-34.<br />

50. Bott J, Carroll MP, Conway JH, Keilty SE, Ward EM, Brown<br />

AM, et al. Randomised controlled trial of nasal ventilation<br />

in acute ventilatory failure due to chronic obstructive airways<br />

disease. Lancet 1993; 341:1555-7.<br />

51. Brochard L, Mancebo J, Wysocki M, Lofaso F, Conti G,<br />

Rauss A, et al. Noninvasive ventilation <strong>for</strong> acute exacerbations<br />

of chronic obstructive pulmonary disease. N Engl J<br />

Med 1995; 333:817-22.<br />

52. Kramer N, Meyer TJ, Meharg J, Cece RD, Hill NS.<br />

Randomized, prospective trial of noninvasive positive pressure<br />

ventilation in acute respiratory failure. Am J Respir<br />

Crit Care Med 1995; 151:1799-806.<br />

53. Plant PK, Owen JL, Elliott MW. Early use of non-invasive<br />

ventilation <strong>for</strong> acute exacerbations of chronic obstructive<br />

pulmonary disease on general respiratory wards: a multicentre<br />

randomised controlled trial. Lancet 2000; 355:1931-5.<br />

54. Rossi A, Gottfried SB, Higgs BD, Zocchi L, Grassino A,<br />

Milic-Emili J. Respiratory mechanics in mechanically ventilated<br />

patients with respiratory failure. J Appl Physiol 1985;<br />

58:1849-58.<br />

55. Esteban A, Anzueto A, Alia I, Gordo F, Apezteguia C,<br />

Palizas F, et al. How is mechanical ventilation employed in<br />

the intensive care unit? An international utilization review.<br />

Am J Respir Crit Care Med 2000; 161:1450-8.<br />

56. American Thoracic Society. International Consensus<br />

Conferences in Intensive Care Medicine: noninvasive positive<br />

pressure ventilation in acute respiratory failure. Am J<br />

Respir Crit Care Med 2001; 163(1):283-91.<br />

57. Plant PK, Owen JL, Elliott MW. Non-invasive ventilation in<br />

acute exacerbations of chronic obstructive pulmonary disease:<br />

long term survival and predictors of in-hospital outcome.<br />

Thorax 2001; 56:708-12.<br />

58. Keenan SP, Kernerman PD, Cook DJ, Martin CM,<br />

McCormack D, Sibbald WJ. Effect of noninvasive positive<br />

pressure ventilation on mortality in patients admitted with<br />

acute respiratory failure: a meta-analysis. Crit Care Med<br />

1997; 25:1685-92.<br />

59. Purro A, Appendini L, De Gaetano A, Gudjonsdottir M,<br />

Donner CF, Rossi A. Physiologic determinants of ventilator<br />

dependence in long-term mechanically ventilated patients.<br />

Am J Respir Crit Care Med 2000; 161:1115-23.<br />

60. Torres A, Reyes A, Roca J, Wagner PD, Rodriguez-Roisin<br />

R. Ventilation-perfusion mismatching in chronic obstructive<br />

pulmonary disease during ventilator weaning. Am Rev<br />

Respir Dis 1989; 140:1246-50.<br />

MANAGEMENT OF COPD 97


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 98<br />

61. Beydon L, Cinotti L, Rekik N, Radermacher P, Adnot S,<br />

Meignan M, et al. Changes in the distribution of ventilation<br />

and perfusion associated with separation from mechanical<br />

ventilation in patients with obstructive pulmonary disease.<br />

Anesthesiology 1991; 75:730-8.<br />

62. Nava S, Ambrosino N, Clini E, Prato M, Orlando G, Vitacca<br />

M, et al. Noninvasive mechanical ventilation in the weaning<br />

of patients with respiratory failure due to chronic obstructive<br />

pulmonary disease. A randomized, controlled trial. Ann<br />

Intern Med 1998; 128:721-8.<br />

63. Esteban A, Frutos F, Tobin MJ, Alia I, Solsona JF, Valverdu<br />

I, et al. A comparison of four methods of weaning patients<br />

from mechanical ventilation. Spanish <strong>Lung</strong> Failure<br />

Collaborative Group. N Engl J Med 1995; 332:345-50.<br />

64. Brochard L, Rauss A, Benito S, Conti G, Mancebo J, Rekik<br />

N, et al. Comparison of three methods of gradual withdrawal<br />

from ventilatory support during weaning from mechanical<br />

ventilation. Am J Respir Crit Care Med 1994; 150:896-903.<br />

65. Hilbert G, Gruson D, Portel L, Gbikpi-Benissan G,<br />

Cardinaud JP. Noninvasive pressure support ventilation in<br />

COPD patients with postextubation hypercapnic respiratory<br />

insufficiency. Eur Respir J 1998; 11:1349-53.<br />

66. Kessler R, Faller M, Fourgaut G, Mennecier B,<br />

Weitzenblum E. Predictive factors of hospitalization <strong>for</strong><br />

acute exacerbation in a series of 64 patients with chronic<br />

obstructive pulmonary disease. Am J Respir Crit Care Med<br />

1999; 159:158-64.<br />

67. Mushlin AI, Black ER, Connolly CA, Buonaccorso KM,<br />

Eberly SW. The necessary length of hospital stay <strong>for</strong><br />

chronic pulmonary disease. JAMA 1991; 266:80-3.<br />

68. Cotton MM, Bucknall CE, Dagg KD, Johnson MK,<br />

MacGregor G, Stewart C, et al. Early discharge <strong>for</strong> patients<br />

with exacerbations of chronic obstructive pulmonary disease:<br />

a randomized controlled trial. Thorax 2000; 55:902-6.<br />

69. Hughes SL, Weaver FM, Giobbie-Hurder A, Manheim L,<br />

Henderson W, Kubal JD, et al. Effectiveness of team-managed<br />

home-based primary care: a randomized multicenter<br />

trial. Jama 2000; 284:2877-85.<br />

70. Hermiz O, Comino E, Marks G, Daffurn K, Wilson S, Harris<br />

M. Randomised controlled trial of home based care of<br />

patients with chronic obstructive pulmonary disease. Bmj<br />

2002; 325:938.<br />

71. Stoller JK, Lange PA. Inpatient management of chronic<br />

obstructive pulmonary disease. Respir Care Clin N Am<br />

1998; 4:425-38.<br />

72. Peach H, Pathy MS. Follow-up study of disability among<br />

elderly patients discharged from hospital with exacerbations<br />

of chronic bronchitis. Thorax 1981; 36:585-9.<br />

73. Aaron SD, Vandemheen KL, Hebert P, Dales R, Stiell IG,<br />

Ahuja J, Dickinson G, Brison R, Rowe BH, Dreyer J, Yetisir<br />

E, Cass D, Wells G. Outpatient oral prednisone after<br />

emergency treatment of chronic obstructive pulmonary<br />

disease. N Engl J Med. 2003 Jun 26;348(26):2618-25.<br />

74. Barr RG, Rowe BH, Camargo CA. Methylxanthines <strong>for</strong><br />

exacerbations of chronic obstructive pulmonary disease.<br />

Cochrane Database Syst Rev. 2003;(2):CD002168.<br />

(Update of: Cochrane Database Syst Rev.<br />

2001;(1):CD002168.)<br />

75. Man WD, Polkey MI, Donaldson N, Gray BJ, Moxham J.<br />

Community pulmonary rehabilitation after hospitalisation<br />

<strong>for</strong> acute exacerbations of chronic obstructive pulmonary<br />

disease: randomised controlled study. BMJ. 2004 Nov<br />

20;329(7476):1209.<br />

76. Saint S, Bent S, Vittinghoff E, Grady D. Antibiotics in chronic<br />

obstructive pulmonary disease exacerbations. A metaanalysis.<br />

JAMA 1995; 273(12):957-60.<br />

77. Anthonisen NR, Manfreda J, Warren CP, Hershfield ES,<br />

Harding GK, Nelson NA. Antibiotic therapy in exacerbations<br />

of chronic obstructive pulmonary disease. Ann Intern<br />

Med 1987; 106(2):196-204.<br />

78. Stockley RA, O'Brien C, Pye A, Hill SL. Relationship of<br />

sputum color to nature and outpatient management of<br />

acute exacerbations of COPD. Chest 2000; 117(6):1638-45.<br />

79. Nouira S, Marghli S, Belghith M, Besbes L, Elatrous S,<br />

Abroug F. Once daily oral ofloxacin in chronic obstructive<br />

pulmonary disease exacerbation requiring mechanical ventilation:<br />

a randomised placebo-controlled trial. Lancet 2001;<br />

358(9298):2020-5.<br />

80. Pela R, Marchesani F, Agostinelli C, Staccioli D, Cecarini L,<br />

Bassotti C, Sanguinetti CM. Airways microbial flora in<br />

COPD patients in stable clinical conditions and during<br />

exacerbations: a bronchoscopic investigation. Monaldi Arch<br />

Chest Dis 1998;53(3):262-7.<br />

81. Soler N, Torres A, Ewig S, Gonzalez J, Celis R, El-Ebiary<br />

M, Hernandez C, Rodriguez-Roisin R. Bronchial microbial<br />

patterns in severe exacerbations of chronic obstructive pulmonary<br />

disease (COPD) requiring mechanical ventilation.<br />

Am J Respir Crit Care Med 1998;157(5 Pt 1):1498-1505.<br />

82. Fagon JY, Chastre J, Trouillet JL, Domart Y, Dombret MC,<br />

Bornet M, Gibert C. Characterization of distal bronchial<br />

microflora during acute exacerbation of chronic bronchitis.<br />

Use of the protected specimen brush technique in 54<br />

mechanically ventilated patients. Am Rev Respir Dis 1990;<br />

142(5):1004-8.<br />

83. Miravitlles M, Espinosa C, Fernandez-Laso E, Martos JA,<br />

Maldonado JA, Gallego M. Relationship between bacterial<br />

flora in sputum and functional impairment in patients with<br />

acute exacerbations of COPD. Study Group of Bacterial<br />

Infection in COPD. Chest 1999; 116(1):40-6.<br />

84. Eller J, Ede A, Schaberg T, Niederman MS, Mauch H,<br />

Lode H. Infective exacerbations of chronic bronchitis: relation<br />

between bacteriologic etiology and lung function.<br />

Chest 1998; 113(6):1542-8.<br />

85. Balter MS, La Forge J, Low DE, Mandell L, Grossman RF.<br />

Canadian guidelines <strong>for</strong> the management of acute exacerbations<br />

of chronic bronchitis. Can Respir J 2003; 10 Suppl<br />

B:3B-32B.<br />

86. Wilson R, Allegra L, Huchon G, Izquierdo JL, Jones P,<br />

Schaberg T, Sagnier PP. Short-term and long-term outcomes<br />

of moxifloxacin compared to standard antibiotic<br />

treatment in acute exacerbations of chronic bronchitis.<br />

Chest 2004; 125(3):953-64.<br />

87. Martinez FJ, Grossman RF, Zadeikis N, Fisher AC, Walker<br />

K, Ambruzs ME, Tennenberg AM. Patient stratification in<br />

the management of acute bacterial exacerbation of chronic<br />

bronchitis: the role of levofloxacin 750 mg. Eur Respir J<br />

2005; 25(6):1001-10<br />

98 FUTURE RESEARCH


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 99<br />

CHAPTER<br />

6<br />

FUTURE<br />

RESEARCH


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 100<br />

CHAPTER 6: FUTURE RESEARCH<br />

A better understanding of the molecular and cellular<br />

pathogenic mechanisms of COPD should lead to many<br />

new directions <strong>for</strong> both basic and clinical investigations.<br />

Improved methods of early detection, new approaches <strong>for</strong><br />

interventions through targeted pharmacotherapy, possible<br />

means to identify the “susceptible” smoker, and more<br />

effective means of managing exacerbations are needed.<br />

Some research recommendations and future program<br />

goals are provided to stimulate the ef<strong>for</strong>ts of investigators<br />

around the world. There are many additional avenues to<br />

explore.<br />

❥ Until there is a better understanding of the causal<br />

mechanisms of COPD, an absolutely rigid definition of<br />

COPD, and its relationship to other obstructive airways<br />

diseases, will remain controversial. The defining<br />

characteristics of COPD should be better identified.<br />

❥ The stages and natural history of COPD vary from one<br />

patient to another. The clinical utility of the four-stage<br />

classification of severity used in the <strong>GOLD</strong> Report needs<br />

to be evaluated.<br />

❥ Surrogate markers of inflammation, possibly derived<br />

from the analysis of sputum (cells, mediators, enzymes)<br />

or exhaled condensates (lipid mediators, reactive oxygen<br />

species, cytokines), that may predict the clinical usefulness<br />

of new management and prevention strategies <strong>for</strong> COPD<br />

need to be developed.<br />

❥ Longitudinal studies demonstrating the course of<br />

COPD are needed in a variety of populations exposed to<br />

various risk factors. Such studies would provide insight<br />

into the pathogenesis of COPD, identify additional genetic<br />

bases <strong>for</strong> COPD, and identify how genetic risk factors<br />

interact with environmental risk factors in specific patient<br />

populations. Factors that determine why some, but not<br />

all, smokers develop COPD need to be identified.<br />

❥ Data are needed on the use, cost, and relative distribution<br />

of medical and non-medical resources <strong>for</strong> COPD, especially<br />

in countries where smoking and other risk factors are<br />

prevalent. These data are likely to have some impact<br />

on health policy and resource allocation decisions. As<br />

options <strong>for</strong> treating COPD grow, more research will be<br />

needed to help guide health care workers and health<br />

budget managers regarding the most efficient and effective<br />

ways of managing this disease. Methods and strategies<br />

<strong>for</strong> implementation of COPD management programs in<br />

developing countries will require special attention.<br />

❥ While spirometry is recommended to assess and<br />

monitor COPD, other measures need to be developed<br />

and evaluated in clinical practice. Reproducible and<br />

inexpensive exercise-testing methodologies (e.g., stairclimbing<br />

tests) suitable <strong>for</strong> use in developing countries<br />

need to be evaluated and their use encouraged.<br />

Spirometers need to be developed that can ensure e<br />

conomical and accurate per<strong>for</strong>mance when a relatively<br />

untrained operator administers the test.<br />

❥ In<strong>for</strong>mation is needed about the cellular and molecular<br />

mechanisms involved in inflammation in stable COPD<br />

and exacerbations. Inflammatory responses in nonsmokers,<br />

ex-smokers, and smokers with and without COPD should<br />

be compared. The mechanisms responsible <strong>for</strong> the<br />

persistence of the inflammatory response in COPD should<br />

be investigated. Why inflammation in COPD is poorly<br />

responsive to glucocorticosteroids and what treatments<br />

other than glucocorticosteroids are effective in suppressing<br />

inflammation in COPD are research topics that could lead<br />

to new treatment modalities.<br />

❥ Standardized methods <strong>for</strong> tracking trends in COPD<br />

prevalence, morbidity, and mortality over time need to be<br />

developed so that countries can plan <strong>for</strong> future increases<br />

in the need <strong>for</strong> health care services in view of predicted<br />

increases in COPD. This need is especially urgent in<br />

developing countries with limited health care resources.<br />

❥ Since COPD is not fully reversible (with current<br />

therapies) and slowly progressive, it will become ever<br />

more important to identify early cases as more effective<br />

therapies emerge. Consensus on standard methods <strong>for</strong><br />

detection and definition of early disease need to be<br />

developed. Data to show whether or not screening is<br />

effective in directing management decisions in COPD<br />

outcomes are required.<br />

❥ Primary prevention of COPD is one of the major<br />

objectives of <strong>GOLD</strong>. Investigations into the most costeffective<br />

ways to reduce the prevalence of tobacco smoking<br />

in the general population and more specifically in young<br />

people are very much needed. Strategies to prevent people<br />

from starting to smoke and methods <strong>for</strong> smoking cessation<br />

require constant evaluation and improvement. Research<br />

is required to gauge the impact and reduce the risk from<br />

increasing air pollution, urbanization, recurrent childhood<br />

100 FUTURE RESEARCH


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 101<br />

infections, occupational exposures, and use of local<br />

cigarette equivalents. Programs designed to reduce<br />

exposure to biomass fuel in countries where this is used<br />

<strong>for</strong> cooking and domestic heating should be explored in an<br />

ef<strong>for</strong>t to reduce exposure and improve ventilation in homes.<br />

❥ The specific components of effective education <strong>for</strong><br />

COPD patients need to be determined. It is not known,<br />

<strong>for</strong> example, whether COPD patients should be given an<br />

individual management plan, or whether these plans are<br />

effective in reducing health care costs or improving the<br />

outcomes of exacerbations. Developing and evaluating<br />

effective tools <strong>for</strong> physician education concerning prevention,<br />

diagnosis, and management of COPD will be important in<br />

view of the increasing public health problem presented by<br />

COPD.<br />

❥ Studies are needed to determine whether education is<br />

an essential component of pulmonary rehabilitation. The<br />

cost effectiveness of rehabilitation programs has not been<br />

assessed and there is a need to assess the feasibility,<br />

resource utilization, and health outcomes of rehabilitation<br />

programs that are delivered outside the major teaching<br />

hospital setting. Criteria <strong>for</strong> selecting individuals <strong>for</strong><br />

rehabilitation should be evaluated, along with methods to<br />

modify programs to suit the needs of individual patients.<br />

❥ Collecting and evaluating data to classify COPD<br />

exacerbations by severity would stimulate standardization<br />

of this outcome measure that is so frequently used in<br />

clinical trials. Further exploration of the ethical principles<br />

of life support and greater insight into the behavioral<br />

influences that inhibit discussion of such intangible issues<br />

are needed, along with studies to define the needs of<br />

end-stage COPD patients.<br />

❥ There is a pressing need to develop drugs that control<br />

symptoms and prevent the progression of COPD. Some<br />

progress has been made and there are several classes of<br />

drugs that are now in preclinical and clinical development<br />

<strong>for</strong> use in COPD patients.<br />

Bronchodilators: Bronchodilators are the mainstay of<br />

symptomatic therapy and new short-acting and long-acting<br />

bronchodilators are anticipated. With the recognition that<br />

there are different subtypes of muscarinic receptors,<br />

there has been a search <strong>for</strong> more selective antagonists.<br />

Tiotropium bromide, a new drug in advanced clinical trials,<br />

is a quaternary ammonium compound like ipratropium<br />

bromide, but with the unique property of kinetic selectivity<br />

and very long duration of action. Selective phosphodiesterase<br />

type IV inhibitors might combine bronchodilator<br />

and anti-inflammatory activity.<br />

Mediator antagonists: Attention has largely focused<br />

on mediators involved in recruitment and activation of<br />

neutrophils, and reactive oxygen species. In this category<br />

are the LTB4 antagonists, lipoxygenase inhibitors,<br />

chemokine inhibitors, and TNF- inhibitors.<br />

Antioxidants: Oxidative stress is increased in patients<br />

with COPD, particularly during exacerbations. Oxidants<br />

are present in cigarette smoke and are produced<br />

endogenously by activated inflammatory cells, including<br />

neutrophils and alveolar macrophages, suggesting that<br />

antioxidants may be of use in therapy <strong>for</strong> COPD.<br />

Anti-inflammatory drugs: The limited value of glucocorticosteroids<br />

in reducing inflammation in COPD suggests<br />

that novel types of nonsteroidal anti-inflammatory treatment<br />

may be needed. There are several new approaches to<br />

anti-inflammatory treatment in COPD including, <strong>for</strong> example,<br />

phosphodiesterase inhibitors, transcription factor NF-B<br />

inhibitors, and adhesion molecule blockers.<br />

Proteinase inhibitors: There is compelling evidence<br />

that an imbalance between proteinases that digest elastin<br />

(and other structural proteins) and antiproteinases that<br />

protect against this digestion exists in COPD.<br />

Considerable progress has been made in identifying the<br />

enzymes involved in elastolytic activity in emphysema<br />

and in characterizing the endogenous antiproteinases<br />

that counteract this activity, including neutrophil elastase<br />

inhibitors, cathepsin G and proteinase 3 inhibitors, and<br />

matrix metalloproteinase inhibitors. Other serine proteinase<br />

inhibitors (serpins), such as elafin, may also be<br />

important in counteracting elastolytic activity in the lung.<br />

Mucoregulators: It may be important to develop drugs<br />

that inhibit the hypersecretion of mucus, without<br />

suppressing the normal secretion of mucus or impairing<br />

mucociliary clearance. There are several types of<br />

mucoregulatory drugs in development including tachykinin<br />

antagonists, sensory neuropeptide inhibitors, mediator<br />

and enzyme inhibitors, mucin gene suppressors, mucolytic<br />

agents, macrolide antibiotics, and purinoceptor blockers.<br />

Alveolar repair: A major mechanism of airway obstruction<br />

in COPD is loss of elastic recoil due to proteolytic<br />

destruction of the lung parenchyma. Thus, it seems<br />

unlikely that this obstruction can be reversed by drug<br />

therapy, although it might be possible to reduce the rate<br />

of progression by preventing the inflammatory and<br />

enzymatic disease processes. It is even possible that<br />

drugs might be developed to stimulate regrowth of alveoli.<br />

Retinoic acid increases the number of alveoli in rats and,<br />

remarkably, reverses the histological and physiological<br />

FUTURE RESEARCH 101


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 102<br />

changes induced by elastase treatment. The molecular<br />

mechanisms involved and whether this can be extrapolated<br />

to humans are not yet known. Several retinoic acid<br />

receptor subtype agonists have now been developed that<br />

may have a greater selectivity <strong>for</strong> this effect. Hepatocyte<br />

growth factor (HGF) has a major effect on the growth of<br />

alveoli in the fetal lung, and it is possible that in the future<br />

drugs might be developed that switch on responsiveness<br />

to HGF in adult lung or mimic the action of HGF.<br />

Route of delivery: Many inhalers that deliver bronchodilators<br />

have been optimized to deliver drugs to the<br />

respiratory tract in asthma. Methods to quickly and safely<br />

deliver medications to target sites of inflammation and<br />

tissue destruction in COPD need to be evaluated.<br />

102 FUTURE RESEARCH


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 103<br />

Note: This segment on Outcomes and Markers in COPD is presented by the <strong>GOLD</strong> Science<br />

Committee as an Abstract to the <strong>GOLD</strong> Workshop Report (updated 2005) as a “work in<br />

progress.” Comments may be sent to the <strong>GOLD</strong> Science Committee (shurd@prodigy.net).<br />

The <strong>GOLD</strong> Science Committee intends to include segments from this document in the full<br />

revision of the report, scheduled to appear in mid-2006.<br />

OUTCOMES AND MARKERS IN COPD<br />

TABLE OF CONTENTS<br />

PARTICIPANTS ................................................................................................................................................................. 2<br />

PREFACE.......................................................................................................................................................................... 3<br />

I. INTRODUCTION......................................................................................................................................................... 4<br />

II. TERMINOLOGY AND DEFINITIONS .......................................................................................................................... 4<br />

A. Clinical outcome.................................................................................................................................................. 4<br />

B Marker................................................................................................................................................................. 4<br />

C. Relationship between markers and outcomes.................................................................................................... 5<br />

D. Clinical outcomes, markers and modifiers in COPD........................................................................................... 6<br />

E Worked example ................................................................................................................................................. 6<br />

III. CLINICAL OUTCOMES ............................................................................................................................................... 6<br />

A. Mortality ............................................................................................................................................................. 6<br />

B. Symptoms and quality of life.............................................................................................................................. 7<br />

C. Exercise tolerance ............................................................................................................................................. 7<br />

D. Exacerbations and acute respiratory failure ...................................................................................................... 7<br />

E. Weight loss......................................................................................................................................................... 7<br />

F. Use of health care and non-health care resources ............................................................................................ 8<br />

IV. MARKERS.................................................................................................................................................................... 8<br />

A. Mortality .............................................................................................................................................................. 8<br />

B. Symptoms and health status............................................................................................................................... 8<br />

C. Exacerbations and acute respiratory failure ....................................................................................................... 9<br />

D. <strong>Lung</strong> function....................................................................................................................................................... 9<br />

E. Exercise capacity ................................................................................................................................................ 10<br />

F. Weight Loss ......................................................................................................................................................... 10<br />

G. Imaging ............................................................................................................................................................... 10<br />

H. Resource utilization............................................................................................................................................. 11<br />

I. Biomarkers .......................................................................................................................................................... 11<br />

J. Composite markers.............................................................................................................................................. 11<br />

V. SUMMARY AND CONCLUSIONS ............................................................................................................................... 11<br />

TABLE 1. PROPERTIES OF THE IDEAL MARKER ......................................................................................................... 12<br />

TABLE 2. OUTCOME MEASURES FOR COPD............................................................................................................... 13<br />

TABLE 3. MARKERS FOR STABLE COPD ...................................................................................................................... 14<br />

TABLE 4: MARKERS FOR EXACERBATIONS OF COPD .............................................................................................. 15<br />

REFERENCES.................................................................................................................................................................. 15<br />

OUTCOME AND MARKERS 103


<strong>GOLD</strong>_WR_05 8/18/05 5:19 PM Page 104<br />

OUTCOMES AND MARKERS IN COPD<br />

PARTICIPANTS<br />

EXECUTIVE COMMITTEE: L. Fabbri, Chair: A. Agusti, S. Buist, P. Calverley, S. Hurd, C. Jenkins, P. Jones, K. Rabe,<br />

R. Rodriguez-Roisin<br />

WORKING GROUPS: Provided materials to develop the report and invited to participate<br />

in review.<br />

A. Mortality: B. Celli, Chair, Boston, Massachusetts, US<br />

J. Vestbo, Hvidore, Denmark<br />

E. Wouters, Maastricht, The Netherlands<br />

T. Oga, Kyoto, Japan<br />

B. Exacerbation, respiratory failure: C. Jenkins, Chair, NSW Australia<br />

R. Rodriguez Roisin, Barcelona, Spain<br />

S. Sethi, Buffalo, New York, US<br />

J. A. Wedzicha, London, UK<br />

A. Rossi, Verona, Italy<br />

C. Symptoms, quality of life: S. Rennard, Chair, Omaha, Nebraska, US<br />

H. Schunemann, Buffalo, New York, US<br />

P. Jones, Tonbridge, Kent, UK<br />

D. <strong>Lung</strong> Function: L. Fabbri, Chair, Modena, Italy<br />

A. S. Buist, Portland, Oregon, US<br />

P. Sterk, Leiden, The Netherlands<br />

E. Exercise: P. Calverley, Chair, Liverpool, UK<br />

K. Nishimura, Kyoto, Japan<br />

Jose Albert Jardim, Sao Paulo, Brazil<br />

F. Imaging: K. Rabe, Chair, Leiden, The Netherlands<br />

G. Biomarkers: S. Rennard, Chair, Omaha, Nebraska, US<br />

A. G. Agusti, Palma de Mallorca, Spain<br />

H. Health Care Utilization: S. Sullivan, Seattle, Washington, US<br />

<strong>GOLD</strong> NATIONAL LEADERS: Invited to participate in review.<br />

104 OUTCOME AND MARKERS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 105<br />

OUTCOMES AND MARKERS IN COPD<br />

PREFACE<br />

The lack of generally accepted outcome measures in<br />

COPD (other than FEV1) that can be used as criteria<br />

<strong>for</strong> the evaluation of novel treatments and <strong>for</strong> approval<br />

of new medications greatly hinders research and clinical<br />

practice. This problem has been long recognized by<br />

research scientists, clinical investigators, industry<br />

representatives, and regulatory authorities worldwide.<br />

recommendations are expected to be incorporated into the<br />

management section of the revised <strong>GOLD</strong> guidelines that<br />

will be available by the end of 2006.<br />

Leonardo Fabbri, MD<br />

Chair, <strong>GOLD</strong> Executive Committee<br />

July 31, 2005<br />

In 2001, the <strong>Global</strong> <strong>Initiative</strong> <strong>for</strong> <strong>Chronic</strong> <strong>Obstructive</strong> <strong>Lung</strong><br />

<strong>Disease</strong> (<strong>GOLD</strong>) developed, and widely distributed,<br />

evidence-based guidelines <strong>for</strong> COPD therapy titled <strong>Global</strong><br />

Strategy <strong>for</strong> Diagnosis, Management and Prevention of<br />

COPD 1 . These guidelines have been updated each year<br />

2 to assure that recommendations <strong>for</strong> COPD treatment<br />

and management are based on current published literature.<br />

In 2004, under the leadership of Professor Romain<br />

Pauwels, <strong>GOLD</strong> convened an expert panel to review data<br />

on outcome measures and to identify those that could be<br />

used to evaluate management of COPD as described in<br />

the <strong>GOLD</strong> guidelines. The panel was also asked to<br />

provide guidance about additional research that may be<br />

needed to confirm the validity of the use of other<br />

outcome measures.<br />

With the <strong>GOLD</strong> guidelines as the foundation of this<br />

project, and the consultation and support of respected<br />

experts from several regions of the world, we hope that<br />

the recommendations provided in this report will stimulate<br />

discussion in the scientific community as well as additional<br />

research to fill the several gaps in knowledge. We strongly<br />

believe that the process initiated by this “working” document<br />

will eventually benefit clinical practice, clinical research<br />

concerning new COPD medications, regulatory decision<br />

making, and patient welfare.<br />

We are indebted to Dr. Romain Pauwels <strong>for</strong> initiation of<br />

this project. His untimely death occurred be<strong>for</strong>e he could<br />

have significant input into this report. We are grateful <strong>for</strong><br />

the expert consultation provided in particular by Dr. Paul<br />

Jones and Dr. Alvar Agusti who, along with other<br />

colleagues on the panel, provided the recommendations<br />

to the <strong>GOLD</strong> Executive Committee. During the 12-month<br />

period beginning July 1, 2005, this report will be included<br />

as an Appendix to the 2005 update of the <strong>Global</strong><br />

Strategy <strong>for</strong> Diagnosis, Management and Prevention of<br />

COPD and posted on the <strong>GOLD</strong> website:<br />

http://www.goldcopd.org <strong>for</strong> comments. The final<br />

OUTCOME AND MARKERS 105


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 106<br />

I. INTRODUCTION<br />

Patients with COPD are heterogeneous in terms of their<br />

clinical presentation, co-morbidities, underlying lung<br />

pathology, disease severity, and rate of disease progression.<br />

Thus it is highly unlikely that a single measure can<br />

accurately assess the severity of COPD, predict patient<br />

prognosis, and evaluate the effectiveness of therapy,<br />

thereby measuring all the dimensions of the disease 3 .<br />

Yet traditionally, the <strong>for</strong>ced expiratory volume in one<br />

second (FEV 1 ) has been used extensively as a global<br />

measurement <strong>for</strong> COPD. While the long term excessive<br />

decline of FEV 1 is the pathognomonic abnormality of<br />

COPD and may indeed relate to mortality, FEV 1 varies<br />

little over short periods of time, even during exacerbations,<br />

and it relates weakly to other clinical manifestations such<br />

as quality of life or exercise tolerance. It is clear, there<strong>for</strong>e,<br />

that other measures (besides FEV 1 ) need to be identified<br />

and validated to allow a more complete and clinically<br />

relevant assessment of patients with COPD 3 .<br />

The terms “clinical outcome” and “marker” have been<br />

increasingly used over the past few years in relation to<br />

COPD, yet considerable confusion and misuse of these<br />

terms exists. The goals of this document are to:<br />

• clarify the meaning of the terms “clinical outcome” and<br />

“marker” and suggest how these terms can contribute<br />

to COPD management described in <strong>GOLD</strong> 1 ;<br />

• review the available evidence supporting the use of<br />

different clinical outcomes/markers in the overall<br />

assessment of a treatment or intervention in COPD<br />

patients;<br />

• identify existing gaps of knowledge where further<br />

research is required.<br />

II. TERMINOLOGY AND DEFINITIONS<br />

A. Clinical Outcome<br />

A clinical outcome is a consequence of COPD<br />

experienced by the patient (symptoms, weight loss,<br />

exorcise intolerance, exacerbations, health care<br />

resource use, mortality).<br />

The meaning of the term “outcome” varies depending<br />

on the context. For instance, in clinical trials, the term<br />

“outcome variable” refers to the main variable of interest,<br />

irrespective of its nature or type (e.g., FEV 1 , FEV 1<br />

decline, exacerbations). In this document, “outcome” will<br />

be used in the context of the clinical assessment of<br />

COPD.<br />

B. Marker<br />

A marker is a measurement associated with one or<br />

more clinical outcomes.<br />

The term “marker” is used in a number of contexts:<br />

• Diagnostic marker - used as a dichotomous variable<br />

- present or absent. It may not be measured on a<br />

dichotomous scale, but is assigned to one of two<br />

states, based on ranges defined from experience;<br />

e.g., alpha 1 -antitrypsin level, or FEV 1 when used <strong>for</strong><br />

COPD diagnosis.<br />

• Marker of disease severity - describes different<br />

levels of disease severity by categorizing levels of<br />

the marker into predefined ranges, e.g., Body Mass<br />

Index (BMI), or FEV 1 as used in <strong>GOLD</strong> staging.<br />

• Marker of disease progression - used to assess<br />

the course of the disease (e.g., rate of decline in<br />

FEV 1 or rate of deterioration in health status).<br />

• Marker of treatment effect - used to measure<br />

response to treatment (e.g., dyspnea score, lean<br />

body mass, exercise capacity, health status, FEV 1 ,<br />

etc.). In clinical trials these markers are usually<br />

termed ‘outcome variables’.<br />

• Biomarker – a measurement of chemical or biological<br />

material that reflects a disease process, e.g. a biomarker<br />

<strong>for</strong> inflammation.<br />

• Surrogate marker – applies when one marker is<br />

used as a substitute <strong>for</strong> the marker of primary interest,<br />

e.g., High resolution computed tomography (HRCT)<br />

scan densitometry measurement as a surrogate<br />

marker <strong>for</strong> the presence of emphysema.<br />

Regardless of these different uses, the ideal marker<br />

should have the properties shown in Table 1.<br />

C. Relationship between markers and outcomes<br />

The relationship between markers and outcomes is not<br />

straight<strong>for</strong>ward. The following factors need to be<br />

considered:<br />

• A clinical outcome may have multiple markers (e.g.,<br />

BMI, FEV 1 and exercise capacity are all independent<br />

predictors of mortality).<br />

106 OUTCOME AND MARKERS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 107<br />

• The relationship between a marker and a clinical<br />

outcome may be altered by modifiers that are not<br />

directly related to the disease but may have a<br />

significant impact on its clinical outcomes. Modifiers<br />

in COPD include co-morbidity, level of social support,<br />

and ease of access to the health care system.<br />

• A small number of markers may be so well<br />

characterized and understood that they can substitute<br />

effectively <strong>for</strong> a clinical outcome and become an<br />

outcome of treatment in itself. For instance, in the<br />

treatment of cardiovascular disease a reduction in<br />

blood pressure (a marker) has become an accepted<br />

clinical outcome since it is known to reduce the<br />

probability of cardiovascular morbidity and mortality.<br />

• Occasionally a marker may be used both as a marker<br />

of disease severity and as a clinically relevant<br />

outcome in itself. One example, weight loss, is<br />

particularly prevalent among patients with severe<br />

COP 4 , but influences prognosis independently of the<br />

level of lung function impairment 5,6 . Thus, it is both<br />

an outcome that affects the patient (e.g., in terms<br />

of body image) and a marker of underlying disease<br />

activity.<br />

D. Clinical outcomes, markers and modifiers in COPD<br />

Tables 2-4 summarize currently accepted clinical outcomes,<br />

markers, and modifiers in COPD, and the potential use of<br />

different markers in patients with stable and exacerbated<br />

COPD. Once appropriately validated, some current<br />

markers (e.g., HRCT) may become clinical outcomes in<br />

themselves (as in the example of blood pressure cited<br />

above).<br />

While clinical trials in patients with COPD have mainly<br />

focused on changes in lung function as a marker of<br />

treatment effect and/or disease progression, the<br />

importance of measuring clinical outcomes such as<br />

symptoms, exacerbations, and health-related quality of<br />

life (and their associated markers) is gaining increasing<br />

recognition because these outcomes are important to<br />

patients. <strong>Lung</strong> function is only weakly related to these<br />

outcomes (i.e., it is a poor surrogate marker).<br />

E. Worked example<br />

A simple worked example of clinical outcomes, markers,<br />

surrogate markers, and modifiers:<br />

• Clinical outcome: exercise tolerance<br />

• Marker: exercise capacity measured in laboratory<br />

• Surrogate marker <strong>for</strong> exercise capacity: FEV 1<br />

• Modifier: co-morbidity (e.g., heart failure)<br />

The correlation between exercise capacity and FEV 1 is<br />

not always strong. This is often the case with surrogate<br />

markers, and, as a general rule, they should be used only<br />

when it is not possible to use the relevant marker. In a<br />

clinical trial, exercise capacity may be used as the primary<br />

outcome variable, because it is a valid marker of exercise<br />

tolerance (the clinical outcome of interest in this example).<br />

III. CLINICAL OUTCOMES<br />

A. Mortality<br />

Mortality as a clinical outcome <strong>for</strong> COPD can be measured,<br />

is meaningful, and is obviously clinically relevant to the<br />

disease process, in particular to the end-stage disease 7 .<br />

A drawback to using mortality as an outcome in COPD is<br />

that it is often not listed on the death certificate, or may<br />

only be listed as a contributory cause of death. However,<br />

in clinical studies, particularly those in which participants<br />

are followed over time and in which there is supporting<br />

clinical in<strong>for</strong>mation about markers of disease severity,<br />

mortality provides a very important clinical outcome<br />

measure. In studies that use COPD mortality as a clinical<br />

outcome, an adequate adjudication process <strong>for</strong> all deaths<br />

must be followed so that COPD (as either a primary or a<br />

contributory cause of death) is coded as accurately as<br />

possible 8 .<br />

B. Symptoms and quality of life<br />

The most frequent symptoms in COPD patients are<br />

dyspnea, cough, sputum production, and fatigue.<br />

Although fatigue is poorly specific <strong>for</strong> COPD, it has a<br />

very high prevalence but is rarely reported spontaneously<br />

by COPD patients 9 . Symptoms contribute significantly<br />

to other relevant clinical outcomes such as disability,<br />

restriction of normal daily activities, and emotional and<br />

social disturbances. In turn, symptoms impair quality of<br />

life, although the impact will be unique to each patient.<br />

C. Exercise tolerance<br />

The ability to exercise is significantly impaired in many<br />

COPD patients and is an important determinant of<br />

health-related quality of life 10,11 . as it impairs the ability to<br />

carry out daily activities. The ability of exercise to provoke<br />

breathlessness is used in the Medical Research Council<br />

(MRC) dyspnea scale to estimate symptom intensity 12 .<br />

It is difficult to make reliable measurements of a patient’s<br />

daily activity, so physiological measurements in the<br />

OUTCOME AND MARKERS 107


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 108<br />

exercise laboratory are used as markers of impaired<br />

activity. Exercise capacity is not only a marker of exercise<br />

tolerance, but also an important predictor of mortality 13,14 .<br />

D. Exacerbations and acute respiratory failure<br />

A COPD exacerbation is a sustained worsening of the<br />

patient’s condition from the stable state and beyond normal<br />

day-to-day variation that is acute in onset and may warrant<br />

additional treatment 15 . Patients with exacerbations of<br />

COPD typically present with increased breathlessness<br />

with or without cough, changes in sputum volume and<br />

purulence, wheezing, and chest tightness. Exacerbations<br />

are an important clinical outcome of the disease as well<br />

as an important marker of disease severity.<br />

Severe exacerbations may be accompanied by acute<br />

respiratory failure, defined as decreased arterial PO2<br />

(arterial deoxygenation) with or without increased arterial<br />

PCO2. Acute respiratory failure is a common clinical<br />

outcome in severe exacerbations of COPD. Acute<br />

respiratory failure is perceived by the patient as severe<br />

dyspnea often associated with agitation, confusion,<br />

tachycardia and sweating 15 . Mortality ranges between<br />

11% 16 and 20% 17 in patients needing mechanical ventilation.<br />

E. Weight loss<br />

Patients with moderate to severe COPD have a depletion<br />

of fat-free mass, particularly skeletal muscle, that is<br />

reflected by weight loss 18,19 . Weight loss is a predictor<br />

of mortality in patients with COPD, and survival may<br />

improve with an increase in body-mass index. In addition<br />

to the relation of low body weight and mortality, weight<br />

loss is an important determinant of impaired muscle<br />

strength, exercise capacity, exercise response, and health<br />

status, as well as increased morbidity (e.g., recurrent<br />

exacerbations and readmission to hospital) in patients<br />

with COPD 18,19 .<br />

F. Use of health care and non-health care resources<br />

Between 60 and 75 percent of medical expenditures <strong>for</strong><br />

COPD are a direct consequence of exacerbations 20-23 , so<br />

use of health care resources is an important outcome in<br />

COPD representing treatment failure and progression of<br />

disease 24 . In clinical trials, use of emergency treatment,<br />

alone or in combination with symptom and lung function<br />

data, is customarily used to characterize an exacerbation<br />

especially when the primary study outcome is reduction<br />

in the frequency or time to an exacerbation event.<br />

Emergency treatment data can be obtained from the<br />

patient or care-giver, and from clinical or billing records 25 .<br />

Data on preventive pharmacotherapy, diagnostic<br />

investigations, and clinical follow-up are required to<br />

supplement data on emergency treatments in order to<br />

provide a more comprehensive assessment of health<br />

resource use and costs. Assessment of patient and<br />

care-giver travel and waiting time, disability, absence from<br />

work, and productivity while at work comprise additional<br />

and important non-health care resource consumption<br />

measures in COPD 20 .<br />

IV. MARKERS<br />

A. Mortality<br />

To establish a precise cause of death is difficult <strong>for</strong> chronic<br />

diseases (including COPD) that often are associated with<br />

other diseases. While all-cause mortality in COPD can<br />

be assessed, death caused specifically by COPD heavily<br />

relies on accuracy of death certificate. Predictors of<br />

mortality from COPD include lung function (FEV 1 , <strong>for</strong>ced<br />

vital capacity [FVC], inspiratory capacity/total lung capacity<br />

[IC/TLC]), blood gases (both PaO2 and PaCO2), respiratory<br />

symptoms 26 , exercise capacity, BMI 6 , exacerbations and<br />

combinations 27,28 . Of these, lung function is a marker of<br />

all-cause mortality and is associated with COPD mortality<br />

even in the early stages of disease 7 .<br />

B. Symptoms and health status<br />

Dyspnea is currently the only COPD symptom that can<br />

be measured in a standardized manner, through the use<br />

of Borg or Visual Analogue Scales usually applied during<br />

laboratory exercise tests. The Borg scale has standardized<br />

descriptors that allow more direct comparisons between<br />

studies than Visual Analogue Scale scores. Other<br />

measures of dyspnea do not assess this symptom directly,<br />

as they quantify self-reported activity limitation in daily<br />

life. All dyspnea instruments listed in Table 3 (except the<br />

Transitional dyspnea index [TDI]) can distinguish between<br />

different degrees of breathlessness-induced disability,<br />

although the MRC scale is simplest and most widely<br />

used. The TDI and University of Cali<strong>for</strong>nia San Diego<br />

(UCSD) instruments can respond to changes with<br />

treatment but there are currently too few data to make<br />

an assessment as to whether they can track long-term<br />

disease progression 29 .<br />

Health-related quality of life is a clinical outcome of<br />

COPD that will be unique to each patient, so it cannot<br />

be quantified in a standardized manner. Instead, health<br />

status questionnaires are used as markers of the impact<br />

108 OUTCOME AND MARKERS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 109<br />

of the disease on patients’ health, daily life and sense of<br />

well-being. All three health status questionnaires (chronic<br />

respiratory questionnaire [CRQ], St. Georges respiratory<br />

questionnaire [SGRQ], 36 item short <strong>for</strong>m [SF-36]) can<br />

distinguish between different degrees of severity. The<br />

disease-specific CRQ and SGRQ are sensitive to<br />

treatment, but the generic SF-36 is not consistently<br />

responsive to worthwhile therapeutic effects. The SGRQ<br />

and SF-36 have been shown to be responsive to longterm<br />

disease progression, but similar evidence is not<br />

currently available <strong>for</strong> the CRQ 30 .<br />

C Exacerbations and acute respiratory failure<br />

Exacerbations of COPD are clinical outcomes when<br />

evaluating the impact of interventions. Exacerbation<br />

frequency and severity are also used as markers of<br />

COPD severity, progression, impact on quality of life,<br />

and mortality 16,31 . Exacerbations are more frequent and<br />

severe in advanced disease 32 , and exacerbation frequency<br />

is related to the decline in quality of life experienced<br />

by the COPD patient 30,33,34 . There are a few clinical<br />

classifications of exacerbation severity, mostly ranging<br />

from mild to life-threatening, and based essentially on<br />

either symptom-driven 35 or event-driven definitions 36 .<br />

Major difficulties are currently encountered in studies<br />

that use exacerbations as endpoint primary outcome <strong>for</strong><br />

assessing interventions because of the lack of a widely<br />

acknowledged definition of COPD exacerbations. In most<br />

studies, exacerbations have been defined on the basis<br />

of increase in symptoms, requiring patient perception<br />

and a reaction to this perception. Both vary significantly<br />

between patients and may be influenced by a number of<br />

modifiers, such as access to the health care system, and<br />

the presence or absence of family or social support.<br />

Definitions of exacerbations based on the need <strong>for</strong> therapy<br />

can be useful indicators of severity, but may also be<br />

insensitive in some settings given that exacerbation<br />

therapies vary throughout the world. Patients with COPD<br />

exacerbations do not always seek medical care and are<br />

increasingly using self-management strategies that may<br />

exclude visiting the primary care physician unless critically<br />

ill. When a hospital admission occurs, objective measures<br />

of severity may be included. Symptoms, volume and<br />

color of sputum, use of health resources, lung function,<br />

and blood gases may be considered the most relevant<br />

markers of COPD exacerbations (Table 4). Arterial pH<br />

and blood gases are the only proven markers of acute<br />

respiratory failure in COPD 37 . Oxygen saturation by pulse<br />

oximetry can also be used, although less accurately, and<br />

does not provide in<strong>for</strong>mation on arterial PCO 2 .<br />

D. <strong>Lung</strong> function<br />

<strong>Lung</strong> function may act as a marker <strong>for</strong> clinical outcomes<br />

such as symptoms, quality of life, exercise tolerance,<br />

health care utilization, and mortality 1,38 . However, lung<br />

function measures relate weakly to these clinical<br />

outcomes 30 . <strong>Lung</strong> function measures have been used<br />

extensively <strong>for</strong> the diagnosis and assessment of severity<br />

of COPD 1 ; FEV 1 is the most readily available and<br />

reproducible. Other parameters of lung function add<br />

little to FEV 1 , being either less reproducible or less<br />

sensitive 39,40 . Post-bronchodilator FEV 1 and FEV 1 /FVC<br />

are essential markers <strong>for</strong> the diagnosis and assessment<br />

of severity of COPD 1 . Because of its unimodal distribution<br />

and poor reproducibility, short-term reversibility testing<br />

to bronchodilator or glucocorticosteroids add little to<br />

baseline lung function <strong>for</strong> diagnosis 41,42 , prediction of<br />

disease progression 15 , or response to treatment 43 .<br />

Repeated measurements of FEV 1 over a period of time<br />

have been used to study the natural progression of<br />

disease 44,45 . Follow-up studies have shown that the annual<br />

decline in post-bronchodilator FEV 1 may be more<br />

reproducible than pre-bronchodilator as a parameter of<br />

lung function to assess progression 45,46 . Additional lung<br />

function measures such as lung volumes and capacities<br />

(residual volume [RV], functional residual capacity [FRC],<br />

inspiratory capacity [IC], and total lung capacity [TLC]),<br />

carbon monoxide diffusion capacity, and arterial blood<br />

gases may be helpful to assess severity (e.g., severe<br />

COPD, exacerbations), and to predict the response to<br />

specific treatments (e.g., lung volume reduction<br />

surgery) 19,47 , but have not been shown to add much to<br />

FEV 1 , being either less reproducible or less sensitive<br />

E. Exercise capacity<br />

Exercise capacity is a marker <strong>for</strong> clinical outcomes such<br />

as symptoms, quality of life, exercise tolerance, health<br />

care utilization, and mortality. Exercise capacity can be<br />

evaluated by making detailed physiological measurements<br />

in the exercise laboratory (minute ventilation, breathing<br />

pattern, oxygen consumption, carbon dioxide production,<br />

oxygen saturation, and oxygen pulse - all during exercise)<br />

or by using simpler field tests where the duration of<br />

exercise or the distance covered in a fixed time period is<br />

recorded (e.g., six-minute walking test). Measures of<br />

exercise capacity are close to the ideal marker (Table 1),<br />

as they have good validity, specificity, reliability,<br />

repeatability 48 , predictive ability 14 , discriminative ability<br />

and evaluative ability 47 .<br />

OUTCOME AND MARKERS 109


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 110<br />

F. Weight loss<br />

Weight loss is a marker <strong>for</strong> clinical outcomes such as<br />

symptoms, quality of life, exercise tolerance, health care<br />

utilization, and mortality. Serial measurements of body<br />

weight can disclose progressive involuntary weight loss,<br />

generally considered to be clinically relevant when it<br />

exceeds 5% over a month or 10% over 6 months 49 .<br />

Actual body weight can be related to the ideal bodyweight<br />

as derived from height, frame size, and gender.<br />

Nutritional depletion is generally and arbitrarily defined<br />

as body weight of less than 90% of the ideal 4 . Body<br />

weight can be corrected <strong>for</strong> body size by calculation of<br />

body-mass index; a value lower than 20 kg/m 2 is generally<br />

taken as abnormal 4,50 . The assessment of nutritional<br />

status according to body weight provides no qualitative<br />

in<strong>for</strong>mation on body tissues. A fat-free-mass index<br />

(fat-free mass in kg divided by height squared) of less<br />

than 16 kg/m 2 in men and 15 kg/m 2 in women is taken<br />

as an indicator of active body-tissue depletion 18,19 .<br />

G. Imaging<br />

Chest imaging can provide useful in<strong>for</strong>mation <strong>for</strong> diagnosis<br />

and disease severity 51 . The chest x-ray is seldom<br />

diagnostic in COPD, but is valuable in the differential<br />

diagnosis to exclude other diseases. A chest x-ray<br />

may also be helpful in phenotyping COPD in term of<br />

bronchiolitis or emphysema 52 . There is no evidence of the<br />

value of chest x-ray to assess disease progression or<br />

treatment effect. HRCT scan is progressively replacing<br />

the regular chest x-ray <strong>for</strong> differential diagnosis of COPD,<br />

phenotyping of COPD (bronchiolitis vs. emphysema), and<br />

assessment of COPD severity. Densitometric analysis of<br />

a CT-scan or HRCT scan be used to assess the presence<br />

and severity of emphysema 53 , and thus is potentially useful<br />

in assessing the progression of the disease, and the effect<br />

of specific treatments (e.g., retinoids) on progression of<br />

emphysema 54,55 .<br />

H. Resource utilization<br />

The presence and frequency of health care resource<br />

utilization are good markers of COPD exacerbations,<br />

disease severity, and progression of disease 22 . In general,<br />

and <strong>for</strong> patients with chronic disease specifically, increasing<br />

age and female gender are positively related to resource<br />

consumption and costs. The general health status question<br />

on the SF-36, when adjusted <strong>for</strong> age and gender, has<br />

been shown to predict mortality and health care resource<br />

utilization.<br />

I. Biomarkers<br />

COPD is characterized by chronic inflammation 56 .<br />

Inflammatory cells (e.g., T cells, neutrophils, and<br />

eosinophils), mediators (e.g., IL-8, TNFa, LTB4, proteases<br />

and antiproteases, C-reactive protein [CRP]), and<br />

components of exhaled air or condensate involved in this<br />

complex inflammatory cascade have been identified as<br />

potential biomarkers of the disease process, and have<br />

been examined as markers <strong>for</strong> diagnosis 57 , assessment of<br />

severity of stable COPD 58 , diagnosis/assessment of COPD<br />

exacerbations 59 , and evaluation of the effect of treatment 60,61 .<br />

However, no single or combination of biomarkers has yet<br />

been identified that can be reliably used in clinical practice<br />

in the diagnosis, staging, or monitoring of COPD.<br />

J. Composite markers<br />

Because COPD is a multi-system, multi-component<br />

disease 62 , there has been increased interest in the use<br />

of composite markers that reflect the overall effect of the<br />

disease. Two markers in particular may fulfill this function:<br />

exercise testing, which reflects cardiopulmonary and<br />

skeletal muscle function, and health status measurements,<br />

which assess a wide range of symptomatic effects of the<br />

disease. A composite score derived from four established<br />

clinical markers - BMI, MRC Dyspnea Grade, FEV 1 and<br />

6-minute walking distance (all included in Table 3) - has<br />

been created and validated (the BODE Index: Bodymass<br />

index (B), the degree of airflow obstruction (O),<br />

dyspnea (D), exercise capacity (E) 27 ). The components<br />

of this instrument are all markers of mortality, and this<br />

instrument validated against mortality proved to be a<br />

better predictor than FEV 1 alone.<br />

V. SUMMARY AND CONCLUSIONS<br />

Based on available evidence, definitions of clinical outcome<br />

and markers in COPD have been proposed. Most published<br />

investigations have concentrated on COPD as a respiratory<br />

disease, and particularly on respiratory symptoms and<br />

lung function, although more comprehensive approaches<br />

addressing health-related quality of life and exercise testing<br />

have recently appeared. The judicious use of many of the<br />

outcomes and markers described in this report should<br />

greatly enhance the development of new therapeutic<br />

strategies that may eventually contribute to improve the<br />

management of COPD. The increasing evidence that<br />

COPD is a multi-component, complex disease suggests the<br />

need <strong>for</strong> the identification and use of more comprehensive<br />

clinical outcomes and more accurate markers or biomarkers<br />

to assess disease severity, prognosis, and response to<br />

therapy. This should be an important goal <strong>for</strong> future<br />

clinical research investigations.<br />

110 OUTCOME AND MARKERS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 111<br />

TABLE 1. PROPERTIES OF THE IDEAL MARKER<br />

PROPERTY<br />

Validity<br />

Specificity<br />

Reliability<br />

Repeatability<br />

Predictive ability<br />

Discriminative ability<br />

Evaluative ability<br />

Simplicity<br />

Cost-effective<br />

DEFINITION<br />

Strong relationship to underlying disease mechanisms and well-being<br />

Absence of confounding effects of co-morbidities or other factors<br />

Per<strong>for</strong>ms consistently in different settings.<br />

Measurements do not change in the stable state<br />

Predicts clinical outcomes<br />

Identifies differences in severity between patients<br />

Sensitive to changes within patients.<br />

Routine or research procedure<br />

Savings from improved management offset the cost<br />

TABLE 2. OUTCOME MEASURE FOR COPD<br />

Outcome Clinical Relevance Markers Modifiers<br />

Mortality<br />

End of life<br />

FEV 1<br />

BMI<br />

MRC dyspnea<br />

Exercise capacity<br />

BODE<br />

PaO 2 , PaCO 2<br />

Exacerbations<br />

Health status<br />

Co-morbidity<br />

Age<br />

Social/family support<br />

Access to health care system<br />

Symptoms<br />

Quality of Life<br />

Health status<br />

Dyspnea<br />

Health status<br />

Dyspnea scales<br />

Co-morbidity<br />

Exacerbations<br />

Mortality<br />

Health status<br />

<strong>Lung</strong> function decline<br />

Weight loss<br />

Previous frequency<br />

FEV 1<br />

PaCO 2<br />

Bronchial colonization<br />

Inflammatory markers<br />

Co-morbidity<br />

Age<br />

Social/family support<br />

Access to health care system<br />

Exercise Tolerance<br />

Health status<br />

Exacerbations<br />

FEV 1<br />

PaO 2<br />

BMI<br />

Co-morbidity<br />

Age<br />

Weight Loss<br />

Survival<br />

Exercise tolerance<br />

Health status<br />

Exacerbations<br />

Weight<br />

PaO 2 ,PaCO 2<br />

DLCO<br />

CT-emphysema<br />

Co-morbidity<br />

Age<br />

Social/family support<br />

Health Care Utilization<br />

Health status<br />

Economic cost<br />

FEV 1<br />

PaO 2 ,PaCO 2<br />

BMI<br />

Exercise tolerance<br />

Health status<br />

Age<br />

Co-morbidity<br />

Active smoking<br />

Social/family support<br />

Access to health care system<br />

OUTCOME AND MARKERS 111


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 112<br />

112 OUTCOME AND MARKERS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 113<br />

FEV 1 [% pred]<br />

TABLE 4. MARKERS FOR EXACERBATIONS OF COPD<br />

Markers Diagnosis Assessment of<br />

Severity<br />

Assessment of<br />

Treatment<br />

Effects<br />

Predictor<br />

NO YES ? YES ? YES<br />

PaO 2 /PaCO 2 /SaO 2 (%) NO YES [?] YES ///////////<br />

Sputum volume/color YES YES YES ///////////<br />

Imaging<br />

Inflammatory markers /////////// /////////// /////////// YES ?<br />

///////// indicates insufficient evidence<br />

? (differential<br />

diagnosis)<br />

NO NO NO<br />

REFERENCES<br />

1. Pauwels RA, Buist AS, Calverley PM, Jenkins CR, Hurd<br />

SS. <strong>Global</strong> strategy <strong>for</strong> the diagnosis, management, and<br />

prevention of chronic obstructive pulmonary disease.<br />

NHLBI/WHO <strong>Global</strong> <strong>Initiative</strong> <strong>for</strong> <strong>Chronic</strong> <strong>Obstructive</strong> <strong>Lung</strong><br />

<strong>Disease</strong> (<strong>GOLD</strong>) Workshop summary. Am J Respir Crit<br />

Care Med 2001;163(5):1256-1276.<br />

2. Workshop Report: <strong>Global</strong> Strategy <strong>for</strong> Diagnosis,<br />

Management and Prevention of COPD - Updated 2004.<br />

Available from http://www.goldcopd.org 2004.<br />

3. Jones PW, Agusti A. Markers and outcomes in the assessment<br />

of chronic obstructive pulmonary disease. Eur Respir<br />

J 2005;submitted.<br />

4. Schols AM, Soeters PB, Dingemans AM, Mostert R,<br />

Frantzen PJ, Wouters EF. Prevalence and characteristics of<br />

nutritional depletion in patients with stable COPD eligible<br />

<strong>for</strong> pulmonary rehabilitation. Am Rev Respir Dis<br />

1993;147(5):1151-1156.<br />

5. Schols AM, Slangen J, Volovics L, Wouters EF. Weight loss<br />

is a reversible factor in the prognosis of chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med<br />

1998;157(6 Pt 1):1791-1797.<br />

6. Landbo C, Prescott E, Lange P, Vestbo J, Almdal TP.<br />

Prognostic value of nutritional status in chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med<br />

1999;160(6):1856-1861.<br />

7. Mannino DM, Doherty DE, Sonia Buist A. <strong>Global</strong> <strong>Initiative</strong><br />

on <strong>Obstructive</strong> <strong>Lung</strong> <strong>Disease</strong> (<strong>GOLD</strong>) classification of lung<br />

disease and mortality: findings from the Atherosclerosis<br />

Risk in Communities (ARIC) study. Respir Med 2005.<br />

8. Vestbo J. The TORCH (towards a revolution in COPD<br />

health) survival study protocol. Eur Respir J<br />

2004;24(2):206-210.<br />

9. Meek PM, Lareau SC, Anderson D. Memory <strong>for</strong> symptoms<br />

in COPD patients: how accurate are their reports? Eur<br />

Respir J 2001;18(3):474-481.<br />

10. Jones PW, Quirk FH, Baveystock CM, Littlejohns P. A selfcomplete<br />

measure of health status <strong>for</strong> chronic airflow limitation.<br />

The St. George's Respiratory Questionnaire. Am<br />

Rev Respir Dis 1992;145(6):1321-1327.<br />

11. Dowson LJ, Newall C, Guest PJ, Hill SL, Stockley RA.<br />

Exercise capacity predicts health status in alpha(1)-antitrypsin<br />

deficiency. Am J Respir Crit Care Med<br />

2001;163(4):936-941.<br />

12. Bestall JC, Paul EA, Garrod R, Garnham R, Jones PW,<br />

Wedzicha JA. Usefulness of the Medical Research Council<br />

(MRC) dyspnoea scale as a measure of disability in<br />

patients with chronic obstructive pulmonary disease.<br />

Thorax 1999;54(7):581-586.<br />

13. Bowen JB, Votto JJ, Thrall RS, Haggerty MC, Stockdale-<br />

Woolley R, Bandyopadhyay T, ZuWallack RL. Functional<br />

status and survival following pulmonary rehabilitation.<br />

Chest 2000;118(3):697-703.<br />

14. Oga T, Nishimura K, Tsukino M, Sato S, Hajiro T. Analysis<br />

of the factors related to mortality in chronic obstructive pulmonary<br />

disease: role of exercise capacity and health status.<br />

Am J Respir Crit Care Med 2003;167(4):544-549.<br />

15. Rodriguez-Roisin R. Toward a consensus definition <strong>for</strong><br />

COPD exacerbations. Chest 2000;117(5 Suppl 2):398S-401S.<br />

16. Connors AF, Jr., Dawson NV, Thomas C, Harrell FE, Jr.,<br />

Desbiens N, Fulkerson WJ, Kussin P, Bellamy P, Goldman<br />

L, Knaus WA. Outcomes following acute exacerbation of<br />

severe chronic obstructive lung disease. The SUPPORT<br />

investigators (Study to Understand Prognoses and<br />

Preferences <strong>for</strong> Outcomes and Risks of Treatments). Am J<br />

Respir Crit Care Med 1996;154(4 Pt 1):959-967.<br />

17. Breen D, Churches T, Hawker F, Torzillo PJ. Acute respiratory<br />

failure secondary to chronic obstructive pulmonary<br />

disease treated in the intensive care unit: a long term follow<br />

up study. Thorax 2002;57(1):29-33.<br />

18. Agusti AG, Noguera A, Sauleda J, Sala E, Pons J,<br />

Busquets X. Systemic effects of chronic obstructive pulmonary<br />

disease. Eur Respir J 2003;21(2):347-360.<br />

19. Wouters EF. Management of severe COPD. Lancet<br />

2004;364(9437):883-895.<br />

OUTCOME AND MARKERS 113


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 114<br />

20. Ramsey SD, Sullivan SD. The burden of illness and economic<br />

evaluation <strong>for</strong> COPD. Eur Respir J Suppl<br />

2003;41:29s-35s.<br />

21. Andersson F, Borg S, Jansson SA, Jonsson AC, Ericsson<br />

A, Prutz C, Ronmark E, Lundback B. The costs of exacerbations<br />

in chronic obstructive pulmonary disease (COPD).<br />

Respir Med 2002;96(9):700-708.<br />

22. Wouters EF. Economic analysis of the Confronting COPD<br />

survey: an overview of results. Respir Med 2003;97 Suppl<br />

C:S3-14.<br />

23. Miravitlles M, Murio C, Guerrero T, Gisbert R.<br />

Pharmacoeconomic evaluation of acute exacerbations of<br />

chronic bronchitis and COPD. Chest 2002;121(5):1449-1455.<br />

24. Strassels SA, Smith DH, Sullivan SD, Mahajan PS. The<br />

costs of treating COPD in the United States. Chest<br />

2001;119(2):344-352.<br />

25. Vollmer WM, Osborne ML, Buist AS. Uses and limitations<br />

of mortality and health care utilization statistics in asthma<br />

research. Am J Respir Crit Care Med 1994;149(2 Pt 2):<br />

S79-87; discussion S88-90.<br />

26. Nishimura K, Izumi T, Tsukino M, Oga T. Dyspnea is a better<br />

predictor of 5-year survival than airway obstruction in<br />

patients with COPD. Chest 2002;121(5):1434-1440.<br />

27. Celli BR, Cote CG, Marin JM, Casanova C, Montes de Oca<br />

M, Mendez RA, Pinto Plata V, Cabral HJ. The body-mass<br />

index, airflow obstruction, dyspnea, and exercise capacity<br />

index in chronic obstructive pulmonary disease. N Engl J<br />

Med 2004;350(10):1005-1012.<br />

28. Casanova C, Cote C, de Torres JP, Aguirre-Jaime A, Marin<br />

JM, Pinto-Plata V, Celli BR. Inspiratory-to-total lung capacity<br />

ratio predicts mortality in patients with chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med<br />

2005;171(6):591-597.<br />

29. Jadad A, Rizo C, Cubillos P, Stahl E. Measuring symptom<br />

response to pharmacological interventions in patients with<br />

COPD: a review of instruments used in clinical trials. Curr<br />

Med Res Opin 2004;20(12):1993-2005.<br />

30. Jones PW, Kaplan RM. Methodological issues in evaluating<br />

measures of health as outcomes <strong>for</strong> COPD. Eur Respir J<br />

Suppl 2003;41:13s-18s.<br />

31. Vestbo J, Prescott E, Lange P, Schnohr P, Jensen G. Vital<br />

prognosis after hospitalization <strong>for</strong> COPD: a study of a random<br />

population sample. Respir Med 1998;92(5):772-776.<br />

32. Jones PW, Willits LR, Burge PS, Calverley PM. <strong>Disease</strong><br />

severity and the effect of fluticasone propionate on chronic<br />

obstructive pulmonary disease exacerbations. Eur Respir J<br />

2003;21(1):68-73.<br />

33. Fan VS, Curtis JR, Tu SP, McDonell MB, Fihn SD. Using<br />

quality of life to predict hospitalization and mortality in<br />

patients with obstructive lung diseases. Chest<br />

2002;122(2):429-436.<br />

34. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson<br />

JA, Maslen TK. Randomised, double blind, placebo controlled<br />

study of fluticasone propionate in patients with moderate<br />

to severe chronic obstructive pulmonary disease: the<br />

ISOLDE trial. Bmj 2000;320(7245):1297-1303.<br />

35. Anthonisen NR, Manfreda J, Warren CP, Hershfield ES,<br />

Harding GK, Nelson NA. Antibiotic therapy in exacerbations<br />

of chronic obstructive pulmonary disease. Ann Intern<br />

Med 1987;106(2):196-204.<br />

36. Vestbo J. What is an exacerbation of COPD. Eur Respir J<br />

2004;13(88):6-13.<br />

37. Bach PB, Brown C, Gelfand SE, McCrory DC.<br />

Management of acute exacerbations of chronic obstructive<br />

pulmonary disease: a summary and appraisal of published<br />

evidence. Ann Intern Med 2001;134(7):600-620.<br />

38. Gross N. Outcome measures <strong>for</strong> COPD treatments, a critical<br />

evaluation. J of COPD 2004;1:141-157.<br />

39. Quanjer PH, Tammeling GJ, Cotes JE, Pedersen OF,<br />

Peslin R, Yernault JC. <strong>Lung</strong> volumes and <strong>for</strong>ced ventilatory<br />

flows. Report Working Party Standardization of <strong>Lung</strong><br />

Function Tests, European Community <strong>for</strong> Steel and Coal.<br />

Official Statement of the European Respiratory Society.<br />

Eur Respir J Suppl 1993;16:5-40.<br />

40. Celli BR, MacNee W. Standards <strong>for</strong> the diagnosis and<br />

treatment of patients with COPD: a summary of the<br />

ATS/ERS position paper. Eur Respir J 2004;23(6):932-946.<br />

41. Reid DW, Soltani A, Johns DP, Bish R, Williams TJ, Burns<br />

GP, Walters EH. Bronchodilator reversibility in Australian<br />

adults with chronic obstructive pulmonary disease. Intern<br />

Med J 2003;33(12):572-577.<br />

42. Calverley PM, Burge PS, Spencer S, Anderson JA, Jones<br />

PW. Bronchodilator reversibility testing in chronic obstructive<br />

pulmonary disease. Thorax 2003;58(8):659-664.<br />

43. Burge PS, Calverley PM, Jones PW, Spencer S, Anderson<br />

JA. Prednisolone response in patients with chronic obstructive<br />

pulmonary disease: results from the ISOLDE study.<br />

Thorax 2003;58(8):654-658.<br />

44. Traver GA, Cline MG, Burrows B. Predictors of mortality in<br />

chronic obstructive pulmonary disease. A 15-year follow-up<br />

study. Am Rev Respir Dis 1979;119(6):895-902.<br />

45. Fletcher C, Peto R. The natural history of chronic airflow<br />

obstruction. Br Med J 1977;1(6077):1645-1648.<br />

46. Anthonisen NR, Connett JE, Kiley JP, Altose MD, Bailey<br />

WC, Buist AS, Conway WA, Jr., Enright PL, Kanner RE,<br />

O'Hara P, et al. Effects of smoking intervention and the use<br />

of an inhaled anticholinergic bronchodilator on the rate of<br />

decline of FEV 1 . The <strong>Lung</strong> Health Study. JAMA<br />

1994;272(19):1497-1505.<br />

47. National Emphysema Treatment Trial Research Group.<br />

Patients at high risk of death after lung-volume-reduction<br />

surgery. N Engl J Med 2001;345(15):1075-1083.<br />

48. O'Donnell DE, Fluge T, Gerken F, Hamilton A, Webb K,<br />

Aguilaniu B, Make B, Magnussen H. Effects of tiotropium<br />

on lung hyperinflation, dyspnoea and exercise tolerance in<br />

COPD. Eur Respir J 2004;23(6):832-840.<br />

49. Anker SD, Negassa A, Coats AJ, Afzal R, Poole-Wilson<br />

PA, Cohn JN, Yusuf S. Prognostic importance of weight<br />

loss in chronic heart failure and the effect of treatment with<br />

angiotensin-converting-enzyme inhibitors: an observational<br />

study. Lancet 2003;361(9363):1077-1083.<br />

114 OUTCOME AND MARKERS


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 115<br />

50. Agusti AG, Sauleda J, Miralles C, Gomez C, Togores B,<br />

Sala E, Batle S, Busquets X. Skeletal muscle apoptosis<br />

and weight loss in chronic obstructive pulmonary disease.<br />

Am J Respir Crit Care Med 2002;166(4):485-489.<br />

51. Miniati M, Filippi E, Falaschi F, Carrozzi L, Milne EN,<br />

Sostman HD, Pistolesi M. Radiologic evaluation of emphysema<br />

in patients with chronic obstructive pulmonary disease.<br />

Chest radiography versus high resolution computed<br />

tomography. Am J Respir Crit Care Med 1995;151(5):1359-<br />

1367.<br />

52. Boschetto P, Miniati M, Miotto D, Braccioni F, De Rosa E,<br />

Bononi I, Papi A, Saetta M, Fabbri LM, Mapp CE.<br />

Predominant emphysema phenotype in chronic obstructive<br />

pulmonary. Eur Respir J 2003;21(3):450-454.<br />

53. Baldi S, Miniati M, Bellina CR, Battolla L, Catapano G,<br />

Begliomini E, Giustini D, Giuntini C. Relationship between<br />

extent of pulmonary emphysema by high-resolution computed<br />

tomography and lung elastic recoil in patients with<br />

chronic obstructive pulmonary disease. Am J Respir Crit<br />

Care Med 2001;164(4):585-589.<br />

54. Stolk J, Ng WH, Bakker ME, Reiber JH, Rabe KF, Putter H,<br />

Stoel BC. Correlation between annual change in health<br />

status and computer tomography derived lung density in<br />

subjects with alpha1-antitrypsin deficiency. Thorax<br />

2003;58(12):1027-1030.<br />

55. Shaker SB, Stavngaard T, Stolk J, Stoel B, Dirksen A.<br />

Alpha1-antitrypsin deficiency. 7: Computed tomographic<br />

imaging in alpha1-antitrypsin deficiency. Thorax<br />

2004;59(11):986-991.<br />

56. Saetta M, Turato G, Maestrelli P, Mapp CE, Fabbri LM.<br />

Cellular and structural bases of chronic obstructive pulmonary<br />

disease. Am J Respir Crit Care Med<br />

2001;163(6):1304-1309.<br />

57. Sutherland ER, Martin RJ. Airway inflammation in chronic<br />

obstructive pulmonary disease: comparisons with asthma.<br />

J Allergy Clin Immunol 2003;112(5):819-827; quiz 828.<br />

58. Pavord ID, Sterk PJ, Hargreave FE, Kips JC, Inman MD,<br />

Louis R, Pizzichini MM, Bel EH, Pin I, Grootendorst DC,<br />

Parameswaran K, Djukanovic R. Clinical applications of<br />

assessment of airway inflammation using induced sputum.<br />

Eur Respir J Suppl 2002;37:40s-43s.<br />

59. Qiu Y, Zhu J, Bandi V, Atmar RL, Hattotuwa K, Guntupalli<br />

KK, Jeffery PK. Biopsy neutrophilia, neutrophil chemokine<br />

and receptor gene expression in severe exacerbations of<br />

chronic obstructive pulmonary disease. Am J Respir Crit<br />

Care Med 2003;168(8):968-975.<br />

60. Gamble E, Grootendorst DC, Brightling CE, Troy S, Qiu Y,<br />

Zhu J, Parker D, Matin D, Majumdar S, Vignola AM,<br />

Kroegel C, Morell F, Hansel TT, Rennard SI, Compton C,<br />

Amit O, Tat T, Edelson J, Pavord ID, Rabe KF, Barnes NC,<br />

Jeffery PK. Antiinflammatory effects of the phosphodiesterase-4<br />

inhibitor cilomilast (Ariflo) in chronic obstructive<br />

pulmonary disease. Am J Respir Crit Care Med<br />

2003;168(8):976-982.<br />

61. Barnes PJ, Shapiro SD, Pauwels RA. <strong>Chronic</strong> obstructive<br />

pulmonary disease: molecular and cellular mechanisms.<br />

Eur Respir J 2003;22(4):672-688.<br />

62. Agusti AG. COPD, a multicomponent disease: implications<br />

<strong>for</strong> management. Respir Med 2005;99(6):670-682.<br />

OUTCOME AND MARKERS 115


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 116<br />

NOTES


<strong>GOLD</strong>_WR_05 8/18/05 12:56 PM Page 117<br />

Printing and distribution has been made possible by educational grants from:<br />

Visit the <strong>GOLD</strong> website at www.goldcopd.org

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!