27.01.2015 Views

Antimicrobial Treatment of Chronic Osteomyelitis - Lippincott ...

Antimicrobial Treatment of Chronic Osteomyelitis - Lippincott ...

Antimicrobial Treatment of Chronic Osteomyelitis - Lippincott ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

CLINICAL ORTHOPAEDICS AND RELATED RESEARCH<br />

Number 360, pp 47-65<br />

0 1999 <strong>Lippincott</strong> Williams & Wilkins, Inc.<br />

<strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong><br />

<strong>Chronic</strong> <strong>Osteomyelitis</strong><br />

Jon T. Mader, MD*j**>P$; Mark E. Shirtliff, BS**j$;<br />

Stephen C. Bergquist, MSIk and Jason Calhoun, MDY<br />

<strong>Chronic</strong> osteomyelitis has been a difficult<br />

problem for patients and the treating physicians.<br />

Appropriate antibiotic therapy is necessary<br />

to arrest osteomyelitis along with adequate<br />

surgical therapy. Factors involved in<br />

choosing the appropriate antibiotic(s) include<br />

infection type, infecting organism, sensitivity<br />

results, host factors, and antibiotic characteristics.<br />

Initially, antibiotics are chosen on the<br />

basis <strong>of</strong> the organisms that are suspected to be<br />

causing the infection. Once the infecting organism(s)<br />

is isolated and sensitivities are established,<br />

the initial antibiotic(s) may be modified.<br />

In selecting specific antibiotics for the<br />

treatment <strong>of</strong> osteomyelitis, the type <strong>of</strong> infection,<br />

current hospital sensitivity resistance<br />

patterns, and the risk <strong>of</strong> adverse reactions<br />

must be strongly appraised. Antibiotic classes<br />

used in the treatment <strong>of</strong> osteomyelitis include<br />

penicillins, p-lactamase inhibitors, cephalosporins,<br />

other p-lactams (aztreonam and imipenem),<br />

vancomycin, clindamycin, rifampin,<br />

aminoglycosides, fluoroquinolones, trimethoprim-sulfamethoxazole,<br />

metronidazole, and<br />

new investigational agents including teicoplanin,<br />

quinupristiddalfopristin, and oxazolidinones.<br />

Traditional treatments have used operative<br />

procedures followed by 4 to 6 weeks <strong>of</strong><br />

From the Divisions <strong>of</strong> *Infectious Diseases and **Marine<br />

Medicine, Marine Biomedical Institute, Departments<br />

<strong>of</strong> $Internal Medicine, §Microbiology and Immunology,<br />

IIPharmacy, and 9Orthopaedic Surgery,<br />

University <strong>of</strong> Texas Medical Branch, Galveston, TX.<br />

Reprint requests to Jon T. Mader, MD, Surgical Infectious<br />

Diseases Hyperbaric Facility, New Trauma Building,<br />

University <strong>of</strong> Texas Medical Branch, Galveston,<br />

TX 77555-1 115.<br />

parenteral antibiotics. Adjunctive therapy for<br />

treating chronic osteomyelitis may be achieved<br />

by using beads, spacers, or coated implants to<br />

deliver local antibiotic therapy andlor by using<br />

hyperbaric oxygen therapy (once per day for<br />

90-120 minutes at two to three atmospheres at<br />

100% oxygen).<br />

Based on etiologic considerations, bone infections<br />

traditionally have been classified as<br />

either hematogenous osteomyelitis, osteomyelitis<br />

secondary to a contiguous focus<br />

<strong>of</strong> infection, or chronic osteomyelitis.g* Contiguous<br />

focus osteomyelitis has been subdivided<br />

further into osteomyelitis in patients<br />

having normal vascularity and those having<br />

generalized vascular insufficiency. All etiologic<br />

classes <strong>of</strong> osteomyelitis may progress<br />

to a chronic disease process.<br />

Appropriate antibiotic therapy is necessary<br />

to arrest osteomyelitis along with adequate<br />

surgical therapy. Factors involved in<br />

choosing the appropriate antibiotic(s) include<br />

infection type, infecting organism,<br />

sensitivity results, host factors, and antibiotic<br />

characteristics. Initially, antibiotics are<br />

chosen on the basis <strong>of</strong> the organisms that are<br />

suspected to be causing the infection. Later,<br />

when an infecting organism is isolated and<br />

sensitivities are established, the initial antibiotic(s)<br />

may be modified.<br />

In hematogenous osteomyelitis one organism<br />

usually is responsible for the infec-<br />

47


48 Maderetal<br />

tion with Staphylococcus aureus being the<br />

most common isolate. In contrast to<br />

hematogenous osteomyelitis, more than one<br />

pathogen usually is isolated in contiguous<br />

focus osteomyelitis and chronic osteomyelitis.<br />

In contiguous focus osteomyelitis,<br />

Staphylococcus aureus is the<br />

most commonly isolated pathogen, but aerobic<br />

Gram negative rods and anaerobic organisms<br />

<strong>of</strong>ten are found.<br />

BACTERIAL ANTIBIOTIC<br />

RESISTANCE<br />

Beta-lactamase production primarily is mediated<br />

by plasmids. This enzyme is responsible<br />

for the resistance approximately 90% <strong>of</strong><br />

Staphylococcus aureus isolates to penicillin.<br />

Of the penicillins, the penicillinase resistant<br />

semisynthetic penicillins have become the antistaphylococcal<br />

drugs <strong>of</strong> choice because <strong>of</strong><br />

their stability in the presence <strong>of</strong> Staphylococcus<br />

aureus P-lactamase.<br />

Intrinsic resistance is chromosomally mediated<br />

and heterogeneous. This type <strong>of</strong> resistance<br />

is present in only a small percentage <strong>of</strong> a<br />

given Staphylococcus aureus inoculum. Methicillin<br />

resistant Staphylococcus aureus and<br />

Staphylococcus epidermidis are the significant<br />

examples <strong>of</strong> intrinsic resistance. These<br />

isolates are resistant to the semisynthetic<br />

penicillins and to the cephalosporins. The expression<br />

<strong>of</strong> this intrinsically resistant subpopulation<br />

can be enhanced by altering several<br />

cultural conditions such as the use <strong>of</strong> agar<br />

containing high salt concentrations and incubation<br />

at 30" C.77 Methicillin resistant Staphylococcus<br />

aureus and methicillin resistant<br />

Staphylococcus epidermidis are isolated frequently<br />

in large tertiary care hospitals and<br />

nursing homes.85<br />

Tolerance <strong>of</strong> organisms to antibiotics is understood<br />

poorly. It is an in vitro phenomenon<br />

characterized by resistance to the lethal action<br />

<strong>of</strong> a normally bactericidal drug. There is a<br />

marked discrepancy between the mean inhibitory<br />

concentration and mean bactericidal<br />

concentration <strong>of</strong> the bactericidal antibiotic to<br />

Clinical Orthopaedics<br />

and Related Research<br />

the organism. A functional definition <strong>of</strong> tolerance<br />

is a mean bactericidal concentration that<br />

is 32-fold or greater than the mean inhibitory<br />

concentration.76 The clinical significance <strong>of</strong><br />

tolerance is still controversial. One study suggested<br />

that seriously ill patients who are<br />

treated with an antimicrobial to which the infecting<br />

organism was tolerant did less well<br />

than patients treated with bactericidal therapy.53<br />

The existence <strong>of</strong> tolerance is another<br />

reason to perform mean inhibitory concentration<br />

and mean bactericidal concentration testing<br />

on all pathogens isolated from infected<br />

bone.<br />

<strong>Chronic</strong> osteomyelitis or infected joints<br />

with replacements are treated for 4 to 6 weeks<br />

or more. 1.6,29,36,38-41,45 These lengthy antibiotic<br />

regimens become even more problematic<br />

when the patient is infected with methicillin<br />

resistant Staphylococcus aureus, vancomycin<br />

resistant enterococcus, or other multiresistant<br />

bacterial species. In particular, because some<br />

methicillin resistant Staphylococcus aureus<br />

strains are resistant to all current antibiotics<br />

except vancomycin, these infections must be<br />

resolved with parenteral vancomycin treatment<br />

with its associated clinical toxi~ity.55,~~<br />

Also, it has been shown in the laboratory that<br />

the vancomycin resistance gene cluster found<br />

in Enterococcus faecalis can be transferred to<br />

Staphylococcus aureus via conjugation and<br />

can express high level resistance.53 Vancomycin<br />

resistance also has been seen in serial<br />

passaged Staphylococcus aureus laboratory<br />

isolates and has been found in the clinical<br />

setting in Japan, New Jersey, and Oregon.<br />

Therefore, vancomycin, the last chance antimicrobial<br />

for many strains <strong>of</strong> methicillin resistant<br />

Staphylococcus aureus, soon may become<br />

ineffective because <strong>of</strong> the development<br />

<strong>of</strong> resistant strains.<br />

SENSITIVITY TESTING<br />

Once the organism(s) is isolated, the specific<br />

antibacterial activity <strong>of</strong> various antibiotics<br />

can be determined by appropriate sensitivity<br />

techniques. The disk diffusion method is the


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 49<br />

most commonly used method for susceptibility<br />

testing. The diameter <strong>of</strong> a zone <strong>of</strong> inhibition<br />

around an antimicrobial impregnated paper<br />

disk relates approximately linearly to the<br />

antibiotic’s log, mean inhibitory concentration.<br />

Inhibition diameters are interpreted as<br />

signifying susceptibility, intermediate susceptibility,<br />

or resistance to each antimicrobial<br />

agent tested according to published criteria.54<br />

Standard procedures must be followed for<br />

these criteria to retain their validity. The interpretive<br />

criteria only apply to organisms that<br />

grow rapidly. The disk diffusion method is<br />

simple to perform and relatively inexpensive.<br />

However, the method provides only semiquantitative<br />

or qualitative data about the susceptibility<br />

<strong>of</strong> a given organism to a given antibiotic.<br />

Nonetheless, if the test is done<br />

carefully, it provides information that is clinically<br />

useful. Quantitative data are provided by<br />

methods that incorporate serial dilution <strong>of</strong> antibiotics<br />

in agar containing or broth culture<br />

media. Quantitative sensitivity testing by<br />

macro or microdilution techniques is a prerequisite<br />

for the determination <strong>of</strong> the least concentration<br />

<strong>of</strong> the antibiotic required to inhibit<br />

(mean inhibitory concentration) and kill<br />

(mean bactericidal concentration) the isolated<br />

organisms.14 Clinical prejudice demands selection<br />

<strong>of</strong> an antibiotic or antibiotic combination<br />

having a low mean inhibitory concentration<br />

and mean bactericidal concentration<br />

activity relative to its expected serum concentration.<br />

Quantitative testing is useful for selecting<br />

antibiotics for difficult aerobic infections<br />

and/or where therapy will be prolonged.<br />

Selection <strong>of</strong> the best antibiotic or antibiotic<br />

combination is crucial in these situations. It is<br />

reasonable to use quantitative testing to determine<br />

the best antibiotic therapy for osteomyelitis,<br />

septic joint infections, and prosthetic<br />

joint infections. Quantitative testing <strong>of</strong><br />

anaerobic organisms is not yet standardized.<br />

BACTERICIDAL LEVELS<br />

Peak and trough serum bacteriostatic and<br />

bactericidal levels described by Schlicter<br />

and MacLean** <strong>of</strong>ten are used to assess the<br />

bacteriostatic and bactericidal capabilities <strong>of</strong><br />

the treatment antibiotic(s). Initially, patient<br />

serum samples are obtained after dosing to<br />

obtain the peak and trough serum levels. The<br />

serum samples then are diluted serially and<br />

the dilution fractions are tested against an inoculum<br />

<strong>of</strong> the infecting bacterial species.<br />

Using this method, one can obtain an estimation<br />

<strong>of</strong> the antibiotic dose necessary to obtain<br />

adequate serum inhibitory and bactericidal<br />

antibiotic levels. These results are expressed<br />

as minimal inhibitory dilutions and minimum<br />

serum bactericidal dilutions. The interpretation<br />

criteria and significance <strong>of</strong> the data<br />

vary for different laboratories and a standard<br />

uniform system for minimal inhibitory dilutions<br />

and minimum serum bactericidal dilutions<br />

studies currently is needed.68J1.90 Most<br />

investigators strive for a peak minimum<br />

serum bactericidal dilution <strong>of</strong> 1:8 or greater<br />

(eightfold or higher dilution <strong>of</strong> patient’s<br />

serum still is capable <strong>of</strong> having a bactericidal<br />

effect on the infecting bacterial species or<br />

strain).74 In patients with osteomyelitis,<br />

serum bactericidal concentrations have been<br />

used to verify the likelihood <strong>of</strong> treatment<br />

success, especially when second choice antibiotics<br />

are necessary for treatment in patients<br />

with drug allergie~.3~ Minimum serum<br />

bactericidal dilutions also have been used to<br />

ensure the adequacy <strong>of</strong> oral antibiotic ther-<br />

In a typical patient with osteomyelitis<br />

where optimal antibiotics are selected by<br />

mean inhibitory concentration testing, the<br />

likelihood <strong>of</strong> success is governed by the adequacy<br />

<strong>of</strong> debridement surgery rather than by<br />

the adequacy <strong>of</strong> serumcidal levels.<br />

ANTIBIOTIC CHARACTERISTICS<br />

In selecting specific antibiotics for the treatment<br />

<strong>of</strong> osteomyelitis, the type <strong>of</strong> infection,<br />

current hospital sensitivity and resistance<br />

patterns, and the risk <strong>of</strong> adverse reactions<br />

must be appraised strongly. No one antibiotic<br />

or antibiotic combination can be expected<br />

to be effective in all clinical settings.


50 Maderetal<br />

Antibiotic factors that may lead to the decreased<br />

activity <strong>of</strong> antibiotics include pH,<br />

the presence <strong>of</strong> purulent material, and decreased<br />

blood flow.56 It has been shown that<br />

the aminoglycosides such as gentamicin, tobramycin,<br />

amikacin, and netilmicin are less<br />

active under anaerobic, acidic, and hypercapnic<br />

conditions. Although agents may diffuse<br />

into infected tissue, the conditions present<br />

in the area may reduce significantly the<br />

ability <strong>of</strong> these antibiotics to eradicate sensitive<br />

organisms.71.96<br />

There are also organism factors that must<br />

be considered in the treatment <strong>of</strong> osteomyelitis.<br />

The most common organism involved<br />

in osteomyelitis is Staphylococcus<br />

aureus, which has three recognized types <strong>of</strong><br />

staphylococcal resistance: P-lactamase production,<br />

intrinsic resistance, and tolerance.79<br />

ANTIBIOTICS<br />

The initial choice <strong>of</strong> antibiotics for Gram<br />

positive, Gram negative, and anaerobic organisms<br />

are shown in Tables 1, 2, and 3. The<br />

initial antibiotic regimen is modified, if necessary,<br />

by the culture and sensitivity results.<br />

Penicillins<br />

The penicillin class <strong>of</strong> antibiotics frequently<br />

is used for the treatment <strong>of</strong> osteomyelitis.<br />

The penicillins can be divided into general<br />

groups on the basis <strong>of</strong> their antibacterial activity.<br />

Overlap exists among the groups, but<br />

the differences within a group are usually <strong>of</strong><br />

a pharmacologic nature, although one compound<br />

within a group may be more active<br />

than another. The major penicillin groups <strong>of</strong><br />

interest to an orthopaedic surgeon are natural<br />

penicillins, aminopenicillins, penicillinase<br />

resistant penicillins, antipseudomonal penicillins,<br />

and extended spectrum penicillins.<br />

Penicillin G is the major natural penicillin.<br />

Penicillin G has a half life <strong>of</strong> 30 to 60 minutes,<br />

but it can be combined with procaine or<br />

benzathine to make a repository penicillin.<br />

This antimicrobial negatively interacts with<br />

erythromycin and tetracyclines to reduce an-<br />

Clinical Orthopaedics<br />

and Related Research<br />

timicrobial effectiveness. Penicillin is the<br />

drug <strong>of</strong> choice for the treatment <strong>of</strong> Streptococcus<br />

pyogenes and Streptococcus agalactiae.<br />

However, Streptococcus pneumoniae<br />

continues to become more resistant to penicillin.<br />

Currently, Streptococcus pneumoniae<br />

has an intermediate resistance <strong>of</strong> 28% and a<br />

high level resistance <strong>of</strong> 16% to penicillin. In<br />

addition, penicillin has a good anaerobic<br />

spectrum <strong>of</strong> activity except for the Bacteroides<br />

fragilis group. Penicillin G is a drug<br />

<strong>of</strong> choice for the treatment <strong>of</strong> Clostridia perfringens.<br />

The parenteral penicillinase resistant<br />

penicillins include methicillin, nafcillin, and<br />

the isoxazolyl penicillins (including cloxacillin,<br />

dicloxacillin, flucoxacillin, and oxacillin).<br />

These drugs are resistant to staphylococcal<br />

P-lactamase, and are used when<br />

methicillin sensitive Staphylococcus aureus<br />

is present or suspected. The semisynthetic<br />

penicillins are also active against Streptococcus<br />

pyogenes and Streptococcus pneumoniae,<br />

but they have no activity against Enterococcus<br />

species or Gram negative bacilli.<br />

The most active parenteral semisynthetic<br />

penicillins are nafcillin and oxacillin. Nafcillin<br />

and oxacillin may cause interstitial<br />

nephritis, leukopenia, and reversible hepatic<br />

dysfunction.29.66 Methicillin is associated<br />

with the greatest potential for producing interstitial<br />

nephritis.IO4 Cloxacillin and dicloxacillin<br />

are the oral semisynthetic penicillins<br />

<strong>of</strong> choice.<br />

The major aminopenicillins include ampicillin<br />

and amoxicillin. Ampicillin may be<br />

given parenterally or orally, whereas amoxicillin<br />

is only an oral agent. The antibacterial<br />

activity <strong>of</strong> the aminopenicillins is similar.<br />

They are not stable to P-lactamase, and are<br />

less active than penicillin G against Streptococcus<br />

pyogenes and Streptococcus agalactiae.<br />

They are the antibiotics <strong>of</strong> choice for the<br />

treatment <strong>of</strong> Enterococcus species (Enterococcus<br />

faecalis, Enterococcus faecium).55 The<br />

aminopenicillins are also active against many<br />

highly susceptible Gram negative rods, such<br />

as Escherichia coli and Proteus mirabilis.


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 51<br />

TABLE 1. Gram Positive Organisms: Initial Choice <strong>of</strong> Antibiotics for Therapy<br />

(Adult Doses)<br />

Organism First Choice Antibiotics Alternative Antibiotics<br />

Methicillin sensitive<br />

Staphylococcus aureus or<br />

Coagulase negative<br />

Staphylococcus species<br />

Methicillin Resistant<br />

Staphylococcus aureus or<br />

Coagulase negative<br />

Staphylococcus species<br />

Group A streptococcus<br />

Streptococcus pyogenes<br />

Group B streptococcus<br />

Streptococcus agalactiae<br />

Penicillin Sensitive<br />

Streptococcus pneurnoniae<br />

Intermediate Penicillin Resistance<br />

Streptococcus pneurnoniae<br />

Penicillin Resistant<br />

Streptococcus pneurnoniae<br />

Enterococcus species<br />

Nafcillin 2 g every 6 hours or<br />

Clindarnycin 900 rng every 8 hours<br />

Nafcillin 2 g every 6 hours or<br />

Clindarnycin 900 rng every 8 hours<br />

Vancornycin 1 g every 12 hours<br />

Vancornycin 1 g every 12 hours or<br />

Clindarnycin 900 ever 8 hours**<br />

Penicillin G 2 rnU every 4 hours<br />

Penicillin G 2 rnU every 4 hours<br />

Penicillin G 2 rnU every 4 hours<br />

Cefotaxirne 1 g every 6 hours<br />

Vancornycin 1 g every 12 hours<br />

L-Ofloxacin 500 rng daily<br />

Ampicillin 1 g every 6 hourst<br />

Vancornycin 1 g every 12 hours<br />

Cefazolin<br />

Vancornycin<br />

Cefazolin<br />

Vancornycin<br />

SXT* or Minocycline k Rifarnpin<br />

SXT* or Minocycline k Rifarnpin<br />

Clindamycin, Cefazolin, Vancornycin<br />

Clindarnycin, Cefazolin, Vancornycin<br />

Erythromycin, Clindarnycin<br />

Erythromycin, Clindarnycin<br />

Sparfloxacin<br />

Ampicillin-Sul bactarn<br />

*Sulfamethoxazole-Trimethoprim<br />

**Dose every 8 hours if sensitive to Clindamycin<br />

+In a serious Enterococcus species infection ampicillin + Sulbactam plus an aminoglycoside is used<br />

Ticarcillin is an antipseudomonal penicillin.<br />

Ticarcillin has a p-lactam ring and is<br />

susceptible to P-lactamase <strong>of</strong> Gram positive<br />

and Gram negative organisms. Ticarcillin<br />

has a Gram negative spectrum <strong>of</strong> activity<br />

similar to ampicillin, but is more active than<br />

ampicillin against Pseudomonas species, Enterobacter<br />

species, Serratia species, and certain<br />

strains <strong>of</strong> the Bacteroides fragilis group.<br />

Ticarcillin has poor activity against Klebsiella<br />

species.57 Side effects include sodium<br />

loading and bleeding problems because <strong>of</strong><br />

platelet dysfunction.59<br />

The extended spectrum penicillins include<br />

mezlocillin and piperacillin. These<br />

penicillins have an antibacterial spectrum<br />

similar to ticarcillin. In vitro, these antibiotics<br />

are active against Enterococcus<br />

species, Streptococcus species and they inhibit<br />

the majority <strong>of</strong> Klebsiella species. They<br />

are also more active than ticarcillin against<br />

Haemophilus influenza and the Bacteroides<br />

fragilis group.19367 These drugs act in synergy<br />

with the aminoglycosides against Pseudomonas<br />

aeruginosa and most <strong>of</strong> the Enterobacteriaceae.<br />

They have the same side effects<br />

as ticarcillin, except they cause less<br />

sodium loading and bleeding dysfunction.<br />

P-lactamase Inhibitors<br />

Clavulanic acid, sulbactam, and tazobactam<br />

are potent inhibitors <strong>of</strong> P-lactamase produced<br />

by Gram positive and Gram negative<br />

organisms.58 Beta-lactamase <strong>of</strong> Gram positive<br />

species are exoenzymes. Clavulanic<br />

acid, sulbactam, and tazobactam have been<br />

shown to inhibit P-lactamase for numerous<br />

clinically important Gram positive organisms<br />

including Staphylococcus aureus and<br />

Staphylococcus epidermidis.70 Beta-lacta-


52 Maderetal<br />

Clinical Orthopaedics<br />

and Related Research<br />

TABLE 2. Gram Negative Organisms: Initial Choice <strong>of</strong> Antibiotics for Therapy<br />

(Adult Doses)<br />

Organism Antibiotics <strong>of</strong> First Choice Alternative Antibiotics<br />

Acinetobacter species<br />

Enterobacter sDecies<br />

Escherichia coli<br />

Haemophilus influenza<br />

Klebsiella species<br />

Proteus mirabilis<br />

Proteus vulgaris<br />

Proteus rettgeri or<br />

Morganella morganii<br />

Neisseria gonorrhea<br />

Providencia species<br />

Pseudomonas aeruginosa<br />

Serratia marcescens<br />

Ceftazidime 1 g every 8 hours<br />

Cefotaxime 1 g every 8 hours; Mezlocillin;<br />

Ceftazidime<br />

Ampicillin 1 g every 6 hours; Gentamicin; SXT*<br />

Cefotaxirne 1 g every 8 hours; Ampicillinsulbactam;<br />

SXT*<br />

Cefazolin 2 g every 8 hours: Cefotaxime<br />

Ampicillin 1 g every 6 hours; Gentarnicin<br />

Cefotaxirne 2 g every 8 hours<br />

Ceftriaxone 125 mg<br />

Cefotaxime 2 g intravenously every 8 hours<br />

Gentamicin 1.67 mg/kg every 8 hours<br />

Ceftazidirne* 2 g every 8 hours or Cipr<strong>of</strong>loxacin*<br />

400 mg every 12 hours Piperacillin* 3 g<br />

every 6 hours<br />

Cefotaxime 2 g every 8 hours<br />

Gentamicin, lmipenem<br />

L-Ofloxacin, Gentamicin<br />

Cefazolin, L-Ofloxacin<br />

Ampicillin,** L-Ofloxacin<br />

L-Ofloxacin, Gentamicin<br />

L-Ofloxacin, Cefazolin<br />

Mezlocillin, L-Ofloxacin, or<br />

Gentamicin<br />

Doxycycl i ne, L-Of loxac i n,<br />

Ampicillin<br />

SXT* Tobramycin<br />

Ticarcillin Clavulanic Acid<br />

Ticarcillin Clavulanic Acid<br />

Tobramycin, lrnipenern<br />

Ofloxacin, Gentamicin<br />

'Sulfamethoxazole-Trimethoprim<br />

"Nonp-lactarnase producing strain <strong>of</strong> Haernophilus influenzae<br />

tNonpenicillinase producing strain <strong>of</strong> Neisseria gonorrhea<br />

*In a serious infection should be used with an aminoglycoside-Gentamicin or Tobramycin 5 mglkg per day every 8 hours<br />

mase <strong>of</strong> Gram negative and most anaerobic<br />

organisms is situated in the periplasmic<br />

space and is chromosome and plasmid induced.73<br />

Clavulanic acid, sulbactam, and<br />

tazobactam will inhibit P-lactamase <strong>of</strong> many<br />

Gram negative organisms including most Escherichia<br />

coli, Klebsiella species, and Bac-<br />

teroides species. Currently, clavulanic acid is<br />

commercially available with amoxicillin<br />

(AugmentinB Smithkline Beecham, Philadelphia,<br />

PA), and ticarcillin (Timentin@,<br />

Smithkline Beecham). Sulbactam is available<br />

with ampicillin (UnasynB, Pfizer Inc,<br />

New York, NY). Tazobactam is combined<br />

TABLE 3. Anaerobic Organisms: Initial Choice <strong>of</strong> Antibiotics for<br />

Therapy (Adult Doses)<br />

Organism Antibiotic <strong>of</strong> First Choice Alternative Antibiotics<br />

Bacteroides fragilis group Clindamycin 900 rng every 8 hours Ampicillin-sulbactam,<br />

Metronidazole 500 rng every 8 hours Ticarcillin-clavulanic acid<br />

Prevotella species Clindamycin 900 mg every 8 hours Ampicillin-sulbactam,<br />

Cefotetan<br />

Peptostreptococcus species<br />

Metronidazole 500 mg every 8 hours<br />

Penicillin G 2 mU every 4 hours<br />

Ticarcillin-clavulanic acid<br />

Clindarnycin, Metronidazole<br />

Cefotetan<br />

Clostridiurn species Clindarnycin 900 mg every 8 hours Metronidarole, Penicillin


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 53<br />

with piperacillin (Zosyna), Wyeth-Ayerst<br />

Laboratories, Philadelphia, PA). The p-lactam<br />

inhibitors enhance the Gram positive<br />

coverage and to a lesser extent the Gram<br />

negative spectrum <strong>of</strong> these antibiotics.<br />

Cephalosporins<br />

The cephalosporins have been divided into<br />

first, second, third, and fourth generation<br />

agents. The first generation cephalosporins<br />

include cephalothin, cephapirin, cephradine,<br />

and cefazolin, and are active against Staphylococcus<br />

aureus, Staphylococcus epidermidis,<br />

and streptococcus species. They have<br />

limited Gram negative activity, but are active<br />

against Eschericia coli, Klebsiella species,<br />

and Proteus mirabilis. The first generation<br />

cephalosporins are safe antibiotics, but occasionally<br />

are associated with the production<br />

<strong>of</strong> allergic reactions, drug eruptions,<br />

phlebitis, and diarrhea. Cefazolin is the first<br />

generation cephalosporin most widely used<br />

by the orthopaedic community for the treatment<br />

<strong>of</strong> staphylococcal infections, including<br />

osteomyelitis. Large amounts <strong>of</strong> p-lactamase<br />

produced by Staphylococcus aureus ( lo9 organisms<br />

per gram tissue) will inactivate<br />

cefazolin.17 However, high numbers <strong>of</strong><br />

Staphylococcus aureus are not the norm in<br />

staphylococcal osteomyelitis where lo5 or<br />

less organisms per gram <strong>of</strong> bone usually are<br />

found. Cefazolin has a longer half life and<br />

higher serum concentration than the other<br />

first generation cephalosporins.36 The remainder<br />

<strong>of</strong> the first generation cephalosporins<br />

is comparable. They are all more stable<br />

to P-lactamase than they are to cefazolin.<br />

There are many second generation<br />

cephalosporins, but the major ones include<br />

cefamandole, cefoxitin, cefotetan, cefuroxime,<br />

ceforanide, and cefonicid. The second<br />

generation cephalosporins have somewhat<br />

increased activity against Gram negative organisms<br />

as compared with the first generation,<br />

but are less active than the third generation<br />

agents. Cefoxitin and cefotetan are<br />

more active than the other first or second<br />

generation cephalosporins against the anaer-<br />

obes, especially the Bacteroides fragilis<br />

group.35<br />

The major third generation cephalosporins<br />

include: cefotaxime, ceftriaxone, ceftizoxime,<br />

cefoperazone, and ceftazidime. The<br />

third generation cephalosporins are generally<br />

less active than the first generation<br />

cephalosporins against Gram positive organisms,<br />

but are more active against the enterobacteriaceae.95<br />

Cefotaxime, ceftriaxone, and<br />

ceftizoxime and are third generation<br />

cephalosporins with similar antibacterial activity.<br />

They are highly resistant to p-lactamase,<br />

and have activity against Gram positive<br />

organisms with the exception <strong>of</strong> the<br />

Enterococcus species. They have good activity<br />

against most Gram negative organisms<br />

except for Pseudomonas aeruginosa. Cefotaxime,<br />

ceftizoxime, and ceftriaxone have<br />

half lives <strong>of</strong> 1.1, 1.7, and 8 hours, respectively.<br />

Ceftazidime is similar in activity to cefotaxime,<br />

ceftizoxime, and ceftriaxone against<br />

the Enterobacteriaceae, but it has superior<br />

activity against Pseudomonas aeruginosa. It<br />

inhibits approximately 9 1 % <strong>of</strong> the Pseudomonas<br />

aeruginosa strains found in the University<br />

<strong>of</strong> Texas Medical Branch Hospitals.<br />

However, this percentage may differ in other<br />

hospitals. Ceftazidime is the cephalosporin<br />

<strong>of</strong> choice for the treatment <strong>of</strong> sensitive<br />

Pseudomonas aeruginosa.60 For serious<br />

Pseudomonas aeruginosa infections ceftazidime<br />

should be combined with an aminoglycoside<br />

or quinolone.84 Ceftazidime is half<br />

as active against Gram positive organisms as<br />

is cefotaxime, ceftizoxime, and ceftriaxone.<br />

The fourth generation cephalosporins are<br />

represented by cefepime. Cefepime has excellent<br />

activity against aerobic Gram positive<br />

organisms including methicillin sensitive<br />

Staphylococcus aureus and Gram negative<br />

organisms including Pseudomonas aeruginosa.<br />

In vitro data suggest increased activity<br />

<strong>of</strong> cefepime against multiresistant Enterobacter<br />

species. Similar to other cephalosporins,<br />

cefepime has no activity against Enterococcus<br />

species.


54 Maderetal<br />

Other b-lactam Antibiotics<br />

Aztreonam is a monocyclic p-lactam antibiotic,<br />

which is active against most Enterobacteriaceae<br />

and Pseudomonas aeruginosa.93<br />

Aztreonam has no appreciable antibacterial<br />

activity against aerobic Gram positive or<br />

anaerobic bacteria. The drug must be given<br />

parenterally. No major adverse reactions<br />

have been reported. It also <strong>of</strong>fers low liability<br />

<strong>of</strong> cross sensitivity in patients allergic to<br />

penicillin or cephalosporin.<br />

Imipenem is an antimicrobial agent belonging<br />

to the p-lactam class <strong>of</strong> antibiotics.<br />

Biochemically they are carbapenems. Imipenem<br />

has excellent in vitro activity against<br />

aerobic Gram positive organisms including<br />

Staphylococcus aureus, Staphylococcus epidermidis,<br />

Streptococcal species, and Enterococcus<br />

species. Imipenem has excellent<br />

Gram negative activity including the Enterobacteriaceae<br />

and Pseudomonas aeruginosa.<br />

Imipenem also inhibits most anaerobic<br />

species, including the Bacteroides fragilis<br />

group.61 Side effects include seizure activity.<br />

Resistance to Pseudomonas aeruginosa may<br />

develop during therapy and fungal superinfection<br />

may occur.<br />

Vancomy cin<br />

Vancomycin has excellent activity against<br />

Staphylococcus aureus, Staphylococcus epidermidis,<br />

and the Enterococcus species. It is<br />

the antibiotic <strong>of</strong> choice in individuals who are<br />

unable to tolerate either the penicillins or the<br />

cephalosporins.23 Vancomycin is also the antibiotic<br />

<strong>of</strong> choice for the treatment <strong>of</strong> methicillin<br />

resistant Staphylococcus aureus89 and<br />

Staphylococcus epidermidis.' Recent reports<br />

<strong>of</strong> vancomycin resistant Enterococcus species<br />

dictate increased vigilance and caution.52.88<br />

Vancomycin may be associated with nephrotoxicity<br />

or ototoxicity, especially when given<br />

concurrently with an aminoglycoside.85 When<br />

used as monotherapy, the end organ toxicity<br />

<strong>of</strong> vancomycin is minimal. A red man syndrome<br />

<strong>of</strong>ten is observed when vancomycin is<br />

administered in less than 1 hour.<br />

Clinical Orthopaedics<br />

and Related Research<br />

Clindamycin<br />

Clindamycin is one <strong>of</strong> the most active antibiotics<br />

against clinically significant anaerobic<br />

bacteria, particularly the B acteroides fragilis<br />

group. However, clindamycin is ineffective<br />

against 10% to 20% <strong>of</strong> clostridial species<br />

other than perfringens.92 In addition to its<br />

anaerobic activity, clindamycin is also effective<br />

against Staphylococcus aureus, Staphylococcus<br />

epidermidis, and the Streptococcus<br />

species. The half life <strong>of</strong> clindamycin is 2.4<br />

hours, clindamycin is ideally given every 8<br />

hours. Clindamycin has good penetration<br />

into most tissues including bone,65.91 and it<br />

penetrates well into abscesses. Clindamycin<br />

is relatively nontoxic, but may cause diarrhea<br />

and pseudomembranous colitis in a<br />

small percentage <strong>of</strong> patients.38<br />

Rifampin<br />

Rifampin exhibits bactericidal activity<br />

against various Gram positive and negative<br />

organisms. Rifampin is the most active antistaphylococcal<br />

agent known.78 However, rifampin<br />

has less activity than the aminoglycosides<br />

against most Gram negative bacteria.<br />

When rifampin is used alone for the treatment<br />

<strong>of</strong> bacterial infections, a rifampin resistant<br />

subpopulation rapidly develops.16 Expression<br />

<strong>of</strong> rifampin resistance can be<br />

lessened by the addition <strong>of</strong> a second effective<br />

antibiotic. Rifampin in combination<br />

with a semisynthetic penicillin has been used<br />

to treat methicillin sensitive Staphylococcus<br />

species osteomyelitis. Trimethoprim and sulfamethoxazole<br />

or minocycline plus rifampin<br />

have been used to treat methicillin resistant<br />

Staphylococcus species osteomyelitis. In a<br />

multicenter study, Norden et a164 has shown<br />

that the combination <strong>of</strong> rifampin and nafcillin<br />

was slightly superior to nafcillin alone.<br />

However, the results <strong>of</strong> the study only<br />

reached a 0.2 statistical significance. Side effects<br />

<strong>of</strong> rifampin include red discoloration <strong>of</strong><br />

body fluids, gastrointestinal complaints, hepatitis,<br />

and possibly mild immunosuppression.<br />

Rifampin induces liver enzyme activity


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 55<br />

resulting in the inactivation <strong>of</strong> numerous<br />

drugs, including verapamil, corticosteroids,<br />

quinidine, cyclosporin, oral anticoagulants,<br />

estrogens, and oral contraceptives. Drug regimens<br />

for patients must be monitored carefully<br />

with adjustments <strong>of</strong> drug doses when<br />

indicated.<br />

Aminoglycosides<br />

The aminoglycosides include gentamicin, tobramycin,<br />

amikacin, and netilmicin. The<br />

aminoglycosides are the standard to which<br />

other antibiotics are measured for the treatment<br />

<strong>of</strong> aerobic Gram negative infections.<br />

The aminoglycosides generally have poor<br />

activity against Gram positive organisms.<br />

Initially, they may be used for the treatment<br />

<strong>of</strong> Staphylococcus aureus, but resistance to<br />

the aminoglycoside may develop<br />

rapidly.62.IOi They have no effect against the<br />

Streptococcus species or anaerobes. The<br />

aminoglycosides have excellent activity<br />

against the Enterobacteriaceae and Pseudomonas<br />

aeruginosa. The aminoglycosides<br />

may be inactivated by enzymatic modification.<br />

Amikacin has fewer available sites than<br />

the other aminoglycosides for enzymatic inactivation.<br />

consequently, the percentage <strong>of</strong><br />

strains susceptible to amikacin is greater<br />

than for tobramycin, gentamicin, or netilmicin.80<br />

There is no evidence to support<br />

amikacin having greater or lesser activity<br />

than the other aminoglycosides. Toxicity <strong>of</strong><br />

the aminoglycosides includes nephrotoxicity<br />

and ototoxicity.<br />

Fiuoroquinolones<br />

The fluoroquinolones currently are being<br />

used to treat adult patients with orthopaedic<br />

infections including osteomyelitis. The<br />

quinolones are divided into four generations.<br />

The first generation quinolone, nalidixic<br />

acid, is not used to treat orthopaedic infections.<br />

The second generation quinolones include<br />

cipr<strong>of</strong>loxacin and <strong>of</strong>loxacin. Cipr<strong>of</strong>loxacin<br />

and <strong>of</strong>loxacin provide adequate serum, tissue,<br />

and urine concentrations. Cipr<strong>of</strong>loxacin<br />

and <strong>of</strong>loxacin have efficacy against most<br />

Gram negative organisms. Most streptococcal<br />

strains and anaerobic organisms are resistant<br />

to cipr<strong>of</strong>loxacin and <strong>of</strong>loxacin. Reports<br />

<strong>of</strong> resistance in some Staphylococcus aureus<br />

and Staphylococcus epidermidis strains dictate<br />

ca~tion.3~39.81 Cipr<strong>of</strong>loxacin is particularly<br />

advantageous in the treatment <strong>of</strong> Gram<br />

negative bone infections, which traditionally<br />

require prolonged parenteral antibiotic therapy.45<br />

Cipr<strong>of</strong>loxacin is the more active<br />

quinolone against Pseudomonas aeruginosa.<br />

The third generation includes lev<strong>of</strong>loxacin<br />

and sparfloxacin. L-<strong>of</strong>loxacin and<br />

sparfloxacin provide higher serum levels<br />

than either cipr<strong>of</strong>loxacin or <strong>of</strong>loxacin. These<br />

agents have excellent activity against Streptococcus<br />

species including penicillin intermediate<br />

and resistant Streptococcus pneumoniae.<br />

These agents are also active against atypical<br />

respiratory pathogens (Mycobacterium pneumoniae,<br />

Legionella species, and Chlamydia<br />

pneumoniae). These agents have efficacy<br />

against most Gram negative organisms. The<br />

fourth generation (trovafloxacin, grepafloxacin)<br />

quinolones have similar aerobic Gram<br />

positive and Gram negative coverage as the<br />

third generation quinolones. Unlike the third<br />

generation quinolones, the fourth generation<br />

quinolones have excellent anaerobic organism<br />

coverage.15.97 These agents have efficacy<br />

against most Gram negative organisms.<br />

Although second, third, and fourth generation<br />

quinolones are formulated for parenteral<br />

administration, the oral mode <strong>of</strong><br />

these quinolones provides excellent serum<br />

concentrations. Oral administration <strong>of</strong> these<br />

quinolones results in decreased length <strong>of</strong><br />

hospitalization and reduced treatment costs.<br />

In most cases, the patient is begun on the<br />

parenteral quinolone and switched to oral<br />

quinolone therapy unless the patient has a<br />

contraindication to oral antibiotic therapy.<br />

The switch to oral therapy usually occurs at<br />

1 to 2 days into therapy. Patients who have<br />

not completed puberty should not be given<br />

antimicrobial therapy with the quinolone<br />

class <strong>of</strong> antibiotics because <strong>of</strong> bone growth


56 Maderetal<br />

problems found in young beagle dogs. Toxicity<br />

to the quinolones is low. Gastrointestinal<br />

disturbances (nausea, vomiting, and dyspepsia)<br />

are the more commonly found side effects<br />

(2%-5%). Central nervous system reaction<br />

(1%-2%) may occur in the form <strong>of</strong><br />

headache, dizziness, tiredness, or insomnia.<br />

Moderate to severe phototoxicity may be<br />

manifested by some <strong>of</strong> the quinolones (lomefloxacin,<br />

sparfloxacin). Sparfloxacin causes<br />

prolongation <strong>of</strong> the Q-T interval. Rarely,<br />

Achilles tendon rupture may occur as a result<br />

<strong>of</strong> quinolone therapy.<br />

None <strong>of</strong> the quinolones have reliable Enterococcus<br />

species coverage. The current<br />

quinolones have variable Staphylococcus<br />

aureus and Staphylococcus epidermidis coverage,<br />

and resistance to the second generation<br />

quinolones is increasing.3<br />

Trimethoprim-sulfamethoxazole<br />

Trimethoprim-sulfamethoxazole is an antimetabolite<br />

composed <strong>of</strong> a fixed combination<br />

<strong>of</strong> a trimethoprim and sulfonamide. In<br />

vitro these agents are more active together<br />

than either agent is alone.6 Aerobic Gram<br />

negative bacteria including Escherichia coli,<br />

Proteus mirabilis, Haemophilas influenzae,<br />

and Stenotrophomonas maltophilia are consistently<br />

susceptible. In addition, Klebsiella<br />

pneumoniae, Enterobacter species, Serratia<br />

marcescens, indolepositive proteus, and nonaeruginosa<br />

Pseudomonas are also frequently<br />

susceptible. The principle targets <strong>of</strong><br />

trimethoprim-sulfamethoxazole are aerobic<br />

Gram negative organisms, but some Gram<br />

positive bacteria such as Staphylococcus aureus,<br />

Streptococcus pneumoniae, and Streptococcus<br />

pyogenes are <strong>of</strong>ten susceptible.102<br />

In some hospitals, the combination <strong>of</strong><br />

trimethoprim-sulfamethoxazole and rifampin<br />

may be effective for the oral treatment <strong>of</strong><br />

methicillin resistant Staphylococcus aureus<br />

and Staphylococcus epidermidis.100 Trimethoprim-sulfamethoxazole<br />

may be given<br />

either parenterally or orally. The combination<br />

is useful as suppressive therapy for osteomyelitis.<br />

Side effects include gastroin-<br />

Clinical Orthopaedics<br />

and Related Research<br />

testinal disturbances, serum sicknesslike<br />

syndrome, hemolytic anemia, and hypersensitivity<br />

reactions. Trimethoprim-sulfamethoxazole<br />

should not be administered during<br />

the last month <strong>of</strong> pregnancy.<br />

Metronidazole<br />

Metronidazole is a useful and inexpensive<br />

antibiotic for the treatment <strong>of</strong> anaerobic organisms.<br />

This antibiotic is a reducing compound<br />

that leads to the formation <strong>of</strong> toxic<br />

0, radicals. Toxic 0, radicals are lethal for<br />

strict anaerobic organisms because they<br />

lack the protective enzymes superoxide dismutase<br />

and catalase. Metronidazole is active<br />

against all anaerobic organisms except<br />

for actinomycetes and microaerophilic streptococci.75<br />

The drug is well absorbed and penetrates<br />

into tissues and abscesses. Side effects<br />

are rare, but include metallic taste,<br />

seizures, cerebellar dysfunction, disulfiram<br />

reaction with alcohol, and pseudomembranous<br />

colitis.<br />

Investigational Agents<br />

Several new antimicrobial agents are currently<br />

in clinical trials. They <strong>of</strong>fer much<br />

needed alternatives to currently available antimicrobials,<br />

particularly in the treatment <strong>of</strong><br />

infections caused by multiresistant Gram<br />

positive bacteria such as vancomycin resistant<br />

Enterococcus species and methicillin resistant<br />

Staphylococcus aureus.<br />

Teicoplanin is a glycopeptide antibiotic<br />

related to vancomycin but possessing several<br />

properties that make it clinically useful. Teicoplanin<br />

has a prolonged elimination half<br />

life <strong>of</strong> approximately 60 hours allowing one<br />

per day administration. The antimicrobial<br />

spectrum <strong>of</strong> teicoplanin includes Staphylococcal<br />

species and Streptococcus species including<br />

some methicillin resistant Staphylococcus<br />

aureus and vancomycin resistant<br />

Enterococcus species Type B organisms. Teicoplanin<br />

toxicity pr<strong>of</strong>ile is similar to vancomycin<br />

including reports <strong>of</strong> ototoxicity. Teicoplanin<br />

may be given by intramuscular or<br />

intravenous route.2.69


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 57<br />

Quinupristin and dalfopristin (Synercid 8<br />

RhGne-Poulenc Rorer Inc, Collegeville, PA),<br />

is a fixed combination <strong>of</strong> two streptogramins<br />

in a ration <strong>of</strong> 30:70 recently approved for use<br />

in the United States by the Food and Drug Administration.<br />

It possesses in vitro inhibitory<br />

and bactericidal activity against most Gram<br />

positive organisms including vancomycin resistant<br />

Enterococcus faecium.20 Quinupristiddalfopristin<br />

may have a role in the<br />

treatment <strong>of</strong> methicillin resistant Staphylococcus<br />

aureus, Group D enterococcus, and<br />

possible coagulase negative Staphylococcus<br />

species infections in patients who cannot receive<br />

vancomycin therapy. Adverse reactions<br />

seem to be mild and include self limited local<br />

reactions such as itching, pain and burning,<br />

vomiting, and diarrhea. Additional clinical<br />

experience is needed to define the role <strong>of</strong> this<br />

antibiotic in clinical practice.8.20<br />

New synthetic classes <strong>of</strong> antimicrobials,<br />

the oxazolidinones, currently are undergoing<br />

clinical trials. The analogs linezolid and<br />

eperezolid are representatives <strong>of</strong> this new<br />

class. These agents have bacteriostatic activity<br />

against numerous important organisms<br />

including methicillin resistant Staphylococcus<br />

aureus, penicillin resistant Streptococcus<br />

pneumoniae, and vancomycin resistant Enterococcus<br />

species.13 They seem to have efficacy<br />

when administered either orally or<br />

parenterally. Tongue discoloration and a folliculitis<br />

type rash are the commonly reported<br />

adverse effects.<br />

LENGTH OF THERAPY<br />

<strong>Osteomyelitis</strong> traditionally is treated with 4<br />

to 6 weeks <strong>of</strong> parenteral antibiotics after definitive<br />

debridement surgery. However, this<br />

time frame has no documented superiority<br />

over other time intervals. Because <strong>of</strong> failure<br />

rates <strong>of</strong> 20% in clinical studies, some authors<br />

advocate treatment with 6 to 8 weeks <strong>of</strong> intravenous<br />

therapy followed by a course <strong>of</strong> 3<br />

months or longer <strong>of</strong> oral therapy.40.98 In this<br />

era <strong>of</strong> resistance development, long duration<br />

antibiotic therapies must be scrutinized care-<br />

fully. There is no evidence that prolonged<br />

parenteral antibiotics will penetrate necrotic<br />

bone. Surgical debridement is necessary to<br />

ensure the physician that he or she is treating<br />

living vascularized bone. It takes approximately<br />

4 to 6 weeks for debrided bone to be<br />

protected by revascularized tissue.98 Because<br />

patient treatment failures are caused mostly<br />

by inadequate surgical debridement rather<br />

than the duration <strong>of</strong> antimicrobials, some<br />

clinicians advocate administering intravenous<br />

antimicrobials for as little as 2 weeks<br />

followed by 4 weeks <strong>of</strong> oral therapy. In cases<br />

<strong>of</strong> relapse, redebridement is advocated. This<br />

treatment method is based on the assumption<br />

that if 4 to 6 weeks <strong>of</strong> antibiotics fail to cure<br />

the disease, then longer treatment courses<br />

are unlikely to be curative unless the dead<br />

devitalized bone is removed.98<br />

TREATMENT MODALITIES<br />

Parenteral Versus Oral<br />

<strong>Osteomyelitis</strong> is a difficult problem for patients<br />

and the treating physicians. Flareups <strong>of</strong><br />

infection require multiple admissions, surgeries<br />

leading to pain, and lengthy antibiotic<br />

therapy with the associated administration<br />

problems and toxicities. The ultimate goal in<br />

management <strong>of</strong> osteomyelitis is to eradicate<br />

and prevent recurrence <strong>of</strong> infection.<br />

Four to 6 weeks <strong>of</strong> parenteral antibiotic<br />

administration after the last major debridement<br />

has become the standard length <strong>of</strong> antibiotic<br />

therapy. In the past, patients were<br />

hospitalized for the entire duration <strong>of</strong> antimicrobial<br />

treatment. Now home health and outpatient<br />

services use heparin locks, peripheral<br />

inserted central catheter lines, and implantable<br />

catheters, which allow parenteral<br />

antibiotic treatment outside the hospital setting.<br />

Although outpatient antibiotic therapy<br />

has decreased costs, 4 to 6 weeks <strong>of</strong> outpatient<br />

intravenous therapy is still expensive<br />

for the patient and healthcare systems.11,25,32,48<br />

An antibiotic treatment regimen<br />

that begins with parenteral therapy and<br />

ends with the self administration <strong>of</strong> oral an-


58 Maderetal<br />

tibiotic therapy would reduce significantly<br />

the cost <strong>of</strong> antibiotic administration.<br />

There is little difference in effectiveness<br />

between the intravenous and oral administration<br />

<strong>of</strong> an antibiotic as long as both routes<br />

provide adequate serum and bone concentrations.<br />

Oral antibiotic therapy has been used<br />

for treatment <strong>of</strong> childhood osteomyelitis. It<br />

is recommended that the patient initially receives<br />

1 to 2 weeks <strong>of</strong> parenteral antibiotic<br />

therapy before changing to an oral regimen.5.33,37,94<br />

The patient must be compliant<br />

and have close outpatient followup. In the<br />

child, serumcidal levels usually are used to<br />

monitor absorption and activity <strong>of</strong> the orally<br />

administered antibiotic.<br />

Currently 4 to 6 weeks <strong>of</strong> intravenous antibiotics<br />

and close followup is recommended<br />

for adult patients with 0steomyelitis.~8.63<br />

Comparable therapeutic experience with intravenous<br />

antibiotics followed by oral antibiotics<br />

and oral antibiotic therapy alone is not<br />

well described in adults. In a small series <strong>of</strong><br />

patients, oral cipr<strong>of</strong>loxacin and <strong>of</strong>loxacin<br />

have been shown to be safe and effective as<br />

parenteral antibiotics in the treatment <strong>of</strong><br />

chronic osteomyelitis caused by susceptible<br />

organisms.22,45<br />

Shirtliff et a187 retrospectively compared<br />

the clinical efficacy <strong>of</strong> 4 weeks <strong>of</strong> intravenous<br />

antibiotics versus 2 weeks <strong>of</strong> intravenous<br />

antibiotics followed by 4 weeks <strong>of</strong><br />

appropriate oral antibiotics. The patients<br />

were followed up at least 12 months after<br />

treatment for outcome determination. <strong>Osteomyelitis</strong><br />

was arrested in 16 <strong>of</strong> 19 patients<br />

in the group treated with 4 weeks <strong>of</strong> intravenous<br />

antibiotic therapy, resulting in an arrest<br />

rate <strong>of</strong> 84.3%. <strong>Osteomyelitis</strong> was arrested<br />

in 17 <strong>of</strong> 19 in the group treated with 2<br />

weeks <strong>of</strong> antibiotics followed by 4 weeks <strong>of</strong><br />

oral antibiotics, resulting in an arrest rate <strong>of</strong><br />

89.5%. The data were analyzed and the difference<br />

was not statistically significant (p ><br />

0.05, chi2 analysis). <strong>Treatment</strong> results for 4<br />

weeks <strong>of</strong> intravenous antibiotics versus 2<br />

weeks <strong>of</strong> intravenous antibiotics followed by<br />

4 weeks <strong>of</strong> oral antibiotics in the treatment <strong>of</strong><br />

Clinical Orthopaedics<br />

and Related Research<br />

long bone osteomyelitis are not significantly<br />

different in this study. This study was limited<br />

because <strong>of</strong> the small sizes <strong>of</strong> the groups and<br />

the variables between the groups.<br />

The major treatment variable between<br />

orally prescribed and intravenous antibiotics<br />

is patient compliance. Every treating physician<br />

must judge whether the patient will be<br />

compliant and will be able to independently<br />

complete the prescribed oral regimen. If the<br />

patient is not compliant, oral antibiotic therapy<br />

can be monitored in a direct observation<br />

oral dosing program. Increased reliance on<br />

oral antibiotic therapy for osteomyelitis will<br />

lead to a reduction <strong>of</strong> intravenous catheter<br />

associated infections.<br />

Because osteomyelitis is a surgical disease,<br />

complete surgical debridement and not<br />

a particular antibiotic regimen is the most<br />

important factor for a successful outcome.48.98<br />

<strong>Osteomyelitis</strong> is characterized by its ability to<br />

recur after long periods <strong>of</strong> quiescence and long<br />

term followup is required for these patients.<br />

Various authors have proposed the use <strong>of</strong> parenteral<br />

antibiotics for periods ranging from 14<br />

days to 6 weeks followed by a variable course<br />

<strong>of</strong> oral antibiotics for the treatment <strong>of</strong> osteornyelitis.9.11.22-45,48f~337<br />

Antibiotics work best<br />

when used in conjunction with adequate debridement,<br />

foreign body and dead bone removal,<br />

abscess drainage, and dead space<br />

obliteration.<br />

Local Therapy Through<br />

Antibiotic Beads<br />

In patients with osteomyelitis implant materials<br />

impregnated with antibiotics have been<br />

used to manage dead space created by debridement<br />

surgery. Beads are placed in local<br />

defects and spacers are used after the infected<br />

total joint prothesis is removed. Antibiotic<br />

beads provide high local concentrations<br />

<strong>of</strong> an antimicrobial agent(s) to the<br />

infected dead space. Thus, a high local concentration<br />

<strong>of</strong> antibiotic can be attained without<br />

exposing the patient to systemic toxic antibiotic<br />

levels, which could result in toxic<br />

side effects.


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 59<br />

Polymethylmethacrylate is an implant material<br />

that has been used successfully with numerous<br />

antibiotics, including vancomycin,<br />

clindamycin, tobramycin, and gentamicin.<br />

However, various problems have been associated<br />

with polymethylmethacrylate use.<br />

First, antibiotic impregnated polymethylmethacrylate<br />

requires a second surgery for its<br />

removal. Second, the implant produces local<br />

immune compromise by impairing natural<br />

luller, lymphocytic, and phagocytic cell activity.<br />

Polymethylmethacrylate implants also<br />

have been linked to decreasing the amount <strong>of</strong><br />

superoxide, a mediator <strong>of</strong> bacterial killing<br />

within phagocytic cell phagosome and reducing<br />

the amount <strong>of</strong> lymphocyte blastogenesis.7<br />

Normal phagocytic processes are devoted to<br />

the removal <strong>of</strong> the implant foreign material<br />

and polymethylmethacrylate particles use energy<br />

and resources <strong>of</strong> the immune system that<br />

normally would be used to fight infection.<br />

Third, polymethylmethacrylate beads usually<br />

provide local bactericidal levels <strong>of</strong> antibiotics<br />

for only 2 to 4 weeks. Once the level <strong>of</strong> antibiotics<br />

eluting from the implant has waned,<br />

there is an increased propensity for the overgrowth<br />

<strong>of</strong> antibiotic resistant organisms that<br />

were not eliminated by the original high concentration<br />

<strong>of</strong> antimicrobials. Finally, the antibiotics<br />

in the polymethylmethacrylate material<br />

<strong>of</strong>ten leech from the outer cortex <strong>of</strong><br />

implant, leaving behind a central core <strong>of</strong> unused<br />

antibiotics. Once the antibiotics have<br />

leeched from the implant cortex, the polymethylmethacrylate<br />

material provides a perfect<br />

substrate for additional bacterial colonization.<br />

Once colonized, many bacteria are<br />

able to synthesize a slime layer, termed the<br />

glycocalyx. This layer prevents the inward<br />

diffusion <strong>of</strong> numerous antimicrobials, allowing<br />

bacterial escape from the bactericidal and<br />

bacteriostatic effects <strong>of</strong> antimicrobial therapy.<br />

Also, the glycocalyx displays host antigenic<br />

properties, thereby allowing the bacteria<br />

to evade detection by the immune system<br />

<strong>of</strong> the host.<br />

New implant materials may be able to reduce<br />

or eliminate many <strong>of</strong> the problems as-<br />

sociated with the clinical standard <strong>of</strong> polymethylmethacrylate<br />

bead therapy for dead<br />

space management. Several studies have<br />

been performed that use alternative materials<br />

for implantation. Zhang et all05 showed that,<br />

in vitro, gentamicin containing high molecular<br />

weight biodegradable poly (D,L-lactide)<br />

cylinders provided a small initial burst followed<br />

by a gradual and sustained release <strong>of</strong><br />

gentamicin. Although this group did not test<br />

the eluted antibiotic against known osteomyelitic<br />

organisms, the detected gentamicin<br />

concentrations were sufficiently above<br />

minimum bactericidal concentrations for<br />

these pathogens. In another related in vitro<br />

model, Shinto et a186 described the ability <strong>of</strong><br />

gentamicin impregnated Ca hydroxyapatite<br />

biodegradable beads to deliver five times the<br />

minimum inhibitory concentrations for<br />

Staphylococcus species for at least 12<br />

weeks. Although in vivo models are lacking,<br />

there has been some research in this area.<br />

Garvin et a121 showed that polyglycolic<br />

beads loaded with gentamicin resulted in the<br />

effective treatment <strong>of</strong> tibia1 Staphylococcus<br />

aureus osteomyelitis in a canine model. In<br />

another study, Calhoun and Mader7 showed<br />

the efficacy <strong>of</strong> a biodegradable antibiotic implant<br />

composed <strong>of</strong> polylactic acid and<br />

poly(D1-lactide): co-glycolide combined<br />

with vancomycin. In the localized rabbit tibial<br />

Staphylococcus aureus osteomyelitis<br />

model, antibiotic impregnated, biodegradable<br />

implant treatment resulted in a significant<br />

reduction in infection when compared<br />

with treatment with systemic vancomycin.<br />

However, this study did not compare the efficacy<br />

<strong>of</strong> infection reduction with polymethylmethacrylate<br />

beads.<br />

Cripps et all0 showed vancomycin impregnated<br />

hydroxyapatite implant material<br />

had approximately equal efficacy in clearing<br />

Staphylococcus aureus osteomyelitis when<br />

compared with vancomycin polymethylmethacrylate<br />

beads in rabbits. Hydroxyapatite<br />

material impregnated with antibiotics<br />

may be better than polymethylmethacrylate<br />

beads and intravenous antibiotics in various


60 Maderetal<br />

ways. First, this material could provide bactericidal<br />

concentrations <strong>of</strong> antibiotics for the<br />

prolonged period necessary to treat completely<br />

the particular orthopaedic infection.<br />

Second, because the hydroxyapatite material<br />

is resorbed, there is no need for bead removal<br />

such as in the case <strong>of</strong> polymethylmethacrylate<br />

antibiotic impregnated beads.<br />

Third, variable resorbability from weeks to<br />

months may allow many types <strong>of</strong> infections<br />

to be treated. Fourth, the polymethylmethacrylate<br />

and polylactic acid material<br />

does not provide for a Ca source necessary<br />

for new bone formation in the repair process<br />

after infection. Finally, because the hydroxyapatite<br />

material is replaced slowly by new<br />

bone formation, the s<strong>of</strong>t tissue or bone defect<br />

may fill slowly with tissue eliminating additional<br />

need for reconstruction.<br />

BONE CONCENTRATIONS<br />

Studies quantifying the bone concentrations<br />

<strong>of</strong> the semisynthetic penicillins, first<br />

generation cephalosporins, clindamycin,<br />

lev<strong>of</strong>loxacin, and vancomycin have been performed.'O,4l342,65,9l<br />

There still are unresolved<br />

methodologic problems. The results <strong>of</strong> bone<br />

concentrations studies are provided in pg per<br />

gram and serum concentrations are in pg per<br />

mL. Most investigators use an elution technique<br />

to recover antibiotic from bone, and<br />

optimal extraction procedures that recover<br />

antibiotic completely from bone still have to<br />

be standardized for each antibiotic. Current<br />

methodology does not allow for the reliable<br />

distinction between cancellous or cortical<br />

bone antibiotic concentrations. Despite these<br />

problems an estimate <strong>of</strong> mean bone concentrations<br />

can be determined and evaluated.<br />

Vancomycin, clindamycin, nafcillin, cefazolin,<br />

and tobramycin bone concentrations<br />

have been determined using an identical elution<br />

technique. The reference curves were<br />

performed in bone powder suspensions. Simultaneous<br />

bone and serum concentrations<br />

were determined using antibiotic doses that<br />

provided optimal serum concentrations for<br />

Clinical Orthopaedics<br />

and Related Research<br />

each antibiotic. Clindamycin was found to<br />

have the greatest bone to serum ratio followed<br />

by vancomycin, nafcillin, tobramycin,<br />

and cefazolin (Table 4). The significance <strong>of</strong><br />

antibiotic bone concentrations is unclear, but<br />

clindamycin had the best results <strong>of</strong> any single<br />

antibiotic therapy (nafcillin, oxacillin,<br />

cephalothin, cefamandole, moxalactam, vancomycin,<br />

rifampin, trimethoprim, gentamicin)<br />

in eradicating experimental Staphylococcus<br />

aureus osteomyelitis.4'<br />

HYPERBARIC OXYGEN THERAPY<br />

The results <strong>of</strong> several open clinical trials<br />

have shown that adjunctive hyperbaric 0,<br />

therapy may be useful in the treatment <strong>of</strong><br />

chronic osteomyelitis.43 Morrey et a151 reported<br />

on 40 patients with chronic osteomyelitis<br />

who met all <strong>of</strong> the following criteria:<br />

the infection had persisted longer than<br />

1 month; at least one surgical debridement<br />

had been performed; at least 2 weeks <strong>of</strong> parenteral<br />

antibiotics had been administered;<br />

and all had been followed up for at least 1<br />

year after treatment. All patients had chronic<br />

refractory osteomyelitis with a recurrence <strong>of</strong><br />

this infection despite previous aggressive antibiotics<br />

and surgical treatment. After hyperbaric<br />

0, therapy, appropriate surgery, and<br />

treatment with antibiotics, 34 patients (85%)<br />

remained clinically free <strong>of</strong> disease, and six<br />

experienced recurrences <strong>of</strong> their osteomyelitis.<br />

Using the same criteria, Davis et<br />

all2 evaluated 38 patients who were treated<br />

with adjunctive hyperbaric 0,. Of these 38<br />

patients, 34 remained free <strong>of</strong> clinical signs <strong>of</strong><br />

osteomyelitis. Although the results <strong>of</strong> these<br />

clinical trials are encouraging, the adjunctive<br />

role <strong>of</strong> hyperbaric 0, in the treatment <strong>of</strong> osteomyelitis<br />

is difficult to assess because <strong>of</strong><br />

patient, surgical, organism, bone, and antibiotic<br />

variables.<br />

Animal studies performed in an experimental<br />

Staphylococcus aureus osteomyelitis<br />

model have shown that hyperbaric 0, administered<br />

under standard treatment conditions<br />

was as effective as cephalothin in eradicating


~~ ~ ~~<br />

Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> Osteomvelitis 61<br />

TABLE 4. Infected Bone Concentrations After Antibiotic Administration in<br />

Experimental Staphylococcus aureus <strong>Osteomyelitis</strong><br />

Antibiotic (dose) Infected Serum pg/mL Bone pg/g Percentage<br />

Clindamycin (70 rng/kg)<br />

Vancomycin (30 rng/kg)<br />

Nafcillin (40 mg/kg)<br />

Moxalactam (40 mg/kg)<br />

Tobrarnycin (5 rng/kg)<br />

Cefazolin (15 mg/kg)<br />

Cefazolin (5 mg/kg)<br />

Cephalothin (40 mg/kg)<br />

12.1 k0.6<br />

36.4 f 4.6<br />

21.8 f 4.6<br />

65.2 f 5.2<br />

14.3k 1.3<br />

67.2 f 2.6<br />

45.6 k 3.2<br />

34.8 f 2.8<br />

11.9k1.9<br />

05.3 f 0.8<br />

02.1 k 0.3<br />

06.2 f 0.7<br />

01.3 +_ 0.1<br />

04.1 f0.7<br />

02.6 k 0.2<br />

01.3 f 0.2<br />

98.3<br />

14.5<br />

9.6<br />

9.5<br />

9.1<br />

6.1<br />

5.7<br />

3.7<br />

Staphylococcus aureus from infected bone.46<br />

Osteomyelitic bone in this experimental<br />

model has decreased blood flow and greatly<br />

decreased partial pressure <strong>of</strong> 0,. Hyperbaric<br />

0, was found to restore intramedullary<br />

0, tensions to physiologic or<br />

supraphysiologic tensions, but did not<br />

acutely increase blood flow in osteomyelitic<br />

bone. Because in vitro hyperoxia<br />

does not directly affect this strain <strong>of</strong><br />

Staphylococcus aureus, hyperbaric 0, was<br />

effective in Staphylococcus aureus osteomyelitis<br />

because it increased intramedullary<br />

0, to tensions at which phagocytic killing<br />

may proceed more efficiently.44<br />

Whereas superoxide dismutase and catalase<br />

are among the enzymatic mechanisms<br />

used by aerobic bacteria to degrade toxic 0,<br />

radicals,26 anaerobic and many microaero:<br />

philic organisms lack these enzymes.50<br />

Therefore, anaerobic organisms are rendered<br />

sensitive to 0, radicals developed intracellularly<br />

and extracellularly during hyperbaric<br />

0, therapy. As a result, increased 0, tension<br />

is directly lethal to fastidious anaerobic organisms<br />

and to some microaerophilic organisms,<br />

but not aerobes.4 In addition, several<br />

clinical reports support the adjunctive role<br />

<strong>of</strong> hyperbaric 0, therapy in the treatment <strong>of</strong><br />

nonclostridial anaerobic infections.47>83.94 The<br />

role <strong>of</strong> hyperbaric 0, therapy in the treatment<br />

<strong>of</strong> infection secondary to Clostridial species<br />

is well validated.28 Also, in vivo studies have<br />

shown hyperbaric 0, to have an indirect<br />

killing mechanism on Clostridium perfringens<br />

mediated through the polymorphonuclear<br />

leukocytes. Thus, hyperbaric 0, provides<br />

the necessary substrate (0,) for the killing <strong>of</strong><br />

aerobic and probably anaerobic organisms by<br />

the polymorphonuclear leukocyte.<br />

The effects <strong>of</strong> hyperbaric 0, on antibiotic<br />

efficacy were shown in a Pseudomonas<br />

aeruginosa osteomyelitis model, in which<br />

hyperbaric 0, potentiated the aminoglycoside<br />

tobramycin.42372.66 Other aminoglycosides<br />

and antibiotics including vancomycin,<br />

the quinolone class <strong>of</strong> antibiotics, nitr<strong>of</strong>urantoin,<br />

and certain sulfonamides are far less active<br />

in hypoxic environments. Such conditions<br />

are found readily in ischemic tissues and<br />

in normal bone. Therefore, hyperbaric 0, therapy<br />

also may be beneficial by augmenting the<br />

effects <strong>of</strong> these antibiotics.<br />

Wound healing is a dynamic process that<br />

requires an adequate 0, tension to proceed.30.31<br />

In the ischemic or infected wound,<br />

hyperbaric 0, provides 0, to promote collagen<br />

production, angiogenesis, and ultimately<br />

wound healing.<br />

Hyperbaric 0, therapy <strong>of</strong>ten is used as adjunctive<br />

therapy in the treatment <strong>of</strong> posttraumatic<br />

osteomyelitis and chronic refractory osteomyelitis.<br />

Posttraumatic osteomyelitis <strong>of</strong>ten<br />

requires significant bone healing because<br />

trauma and the infective process can result in<br />

significant bony destruction. Besides the beneficial<br />

effects <strong>of</strong> direct inhibition <strong>of</strong> anaerobes,<br />

upregulation <strong>of</strong> polymorphonuclear in-


~~<br />

62 Maderetal<br />

Clinical Orthopaedics<br />

and Related Research<br />

tracellular killing, augmenting antibiotic activity,<br />

and maintaining muscle and skin<br />

flaps, hyperbaric 0, therapy can promote accelerated<br />

bone repair. The optimum hyperbaric<br />

0, treatment regimens are one to two<br />

treatments per day for 60 to 120 minutes at<br />

two to three atmospheres <strong>of</strong> pressure with<br />

100% 0,. Oxygenation below this level<br />

(such as occurs in infected bone) will result<br />

in slow bone healing because <strong>of</strong> inhibition <strong>of</strong><br />

fibroblast, osteoclast, osteoblast, and macrophage<br />

activity.103 When 0, levels are raised<br />

beyond optimum levels for a sustained period,<br />

fibroblast activity is highly upregulated,<br />

resulting in a thick collagenous deposition.49<br />

Many in vitro studies have reported<br />

an upregulated osteoclast activity caused by<br />

long exposures to 0, radicals including hydrogen<br />

peroxide associated with hyperoxygen<br />

growth conditions.18J4.’7 The end result<br />

<strong>of</strong> sustained hyperoxygenation is the development<br />

<strong>of</strong> a repair process that is rich in collagen<br />

and structurally weak. Therefore, maximal<br />

bone healing may be achieved when<br />

hyperbaric 0, treatment is provided within<br />

the optimal range <strong>of</strong> treatments for 90 to 120<br />

minutes at two to three atmospheres <strong>of</strong> pressure<br />

with 100% 0, - once daily.<br />

Acknowledgments<br />

The authors thank Michael Cripps, BS, Donna<br />

Milner Mader, BA, and Maureen D. Shirtliff, RN,<br />

for manuscript research and preparation.<br />

References<br />

1. Archer GL: <strong>Antimicrobial</strong> susceptibility and selection<br />

or resistance among Staphylococcus epidermidis<br />

isolates recovered from patients with<br />

infections <strong>of</strong> indwelling foreign devices. Antimicrob<br />

Agents Chemother 14:353-359, 1978.<br />

2. Babul N, Pasko M: Teicoplanin: A new glycopeptide<br />

antibiotic complex. Drug Intell Clin Pharm<br />

22:218-226, 1988.<br />

3. Blumberg HM, Rimland D, Carroll DJ, et al:<br />

Rapid development <strong>of</strong> cipr<strong>of</strong>loxacin resistance in<br />

methicillin-susceptible and -resistant Staphylococcus<br />

aureus. J Infect Dis 163:1279-85, 1991.<br />

4. Brown GL, Thompson PD, Mader JT, et al: Effects<br />

<strong>of</strong> hyperbaric oxygen upon Staphylococcus aureus,<br />

Pseudomonas aeruginosa, and Candida albicans.<br />

Aviat Space Environ Med 50:717-720, 1973.<br />

5. Bryson VJ, Connor JD, Leclere M, Gimmmona<br />

6.<br />

7.<br />

8.<br />

9.<br />

10.<br />

11.<br />

12.<br />

ST High dose oral dicloxacillin treatment <strong>of</strong><br />

acute staphylococcal osteomyelitis in childhood. J<br />

Pediatr 94:673-675, 1979.<br />

Bushby SRM: Synergy <strong>of</strong> Trimethoprim and Sulfonamides:<br />

History and Current Status. In Woodbine<br />

D (ed). Antibiotics and Antibiosis in<br />

Agriculture. London, Butterworths 64-8 1, 1977.<br />

Calhoun JH, Mader JT <strong>Treatment</strong> <strong>of</strong> osteomyelitis<br />

with a biodegradable antibiotic implant.<br />

Clin Orthop 341:206-214, 1997.<br />

Chant C, Rybak M: Quinupristinldalfopristin (RP<br />

59500): A new streptograinin antibiotic. Ann<br />

Pharmacother 29: 1022-1027, 1995.<br />

Cierny G, Mader JT Adult chronic osteomyelitis.<br />

Orthopedics 7: 1557-1564, 1984.<br />

Cripps M, Shirtliff ME, Mader JT: The <strong>Treatment</strong><br />

<strong>of</strong> osteomyelitis with a hydroxyapatite antibiotic<br />

implant in a rabbit model. 38th Interscience Conference<br />

on <strong>Antimicrobial</strong> Agents and Chemotherapy,<br />

San Diego 1998.<br />

David WH, Mark PM: Bacterial osteomyelitis in<br />

adults: Evolving considerations in diagnosis and<br />

treatment. Am J Med 101:550-561, 1996.<br />

Davis JC, Heckman JD, DeLee JC, et al: <strong>Chronic</strong><br />

non-hematogenous osteomyelitis treated with adjuvant<br />

hyperbaric oxygen. J Bone Joint Surg<br />

68A: 1210-1217, 1986.<br />

13. Dresser LD, Rybak MJ: The pharmacologic and<br />

bacteriologic properties <strong>of</strong> oxazolidinones, a new<br />

class <strong>of</strong> synthetic antimicrobials. Pharmacotherapy<br />

1k456-462, 1998.<br />

14. Ericcson HM, Sherris, JC: Antibiotic sensitivity<br />

testing. Report <strong>of</strong> an international collaborative<br />

study. Acta Pathol Microbiol Scand 227(Suppl<br />

B):1-90, 1971.<br />

15. Ernst ME, Ernst EJ, Klepser ME: Lev<strong>of</strong>loxacin<br />

and trovafloxacin: The next generation <strong>of</strong> fluoroquinolones<br />

Am J Health Syst Pharm 54:<br />

2569-2584, 1997.<br />

16.<br />

Faville RJ, Zaske DE, Kaplan EL: Staphylococcus<br />

aureus endocarditis: Combined therapy with vancomycin<br />

and rifampin. JAMA 240: 1963-1965,<br />

1978.<br />

17. Fong IW, Engelking ER, Kirby WMM: Relative<br />

inactivation by Staphylococcus aureus <strong>of</strong> eight<br />

cephalosporin antibiotics. Antimicrob Agents<br />

Chemother 9:939-944, 1976.<br />

18. Fraser JH, Helfrich MH, Wallace HM, Ralston<br />

SH: Hydrogen peroxide, but not superoxide, stimulates<br />

bone resorption in mouse calvarieae. Bone<br />

19:223-226, 1996.<br />

19. Fu KP, Neu HC: Piperacillin a new penicillin active<br />

against many bacterial resistant to other penicillins.<br />

Antimicrob Agents Chemother 13:<br />

358-367, 1987.<br />

20.<br />

Fuller RE, Drew RH, Perfect JR: <strong>Treatment</strong> <strong>of</strong><br />

vancomycin-resistant enterococci with a focus on<br />

quinupristin-dalfopristin. Pharrnacotherapy 16:<br />

584- 592,1996.<br />

21. Garvin KL, Miyano JA, Robinson D, et al: Polylactidelpolyglycolide<br />

antibiotic implants in the<br />

treatment <strong>of</strong> osteomyelitis: A canine model. J<br />

Bone Joint Surg 76A: 1500-1506, 1994.<br />

22. Gentry LO, Rodriguez GG: Oral cipr<strong>of</strong>loxacin


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 63<br />

compared with parenteral antibiotics in the treatment<br />

<strong>of</strong> osteomyelitis. Antimicrob Agents<br />

Chemother 34:40-43, 1990.<br />

23. Geraci JE, Hermans PE: Vancomycin. Mayo Clin<br />

Pr0~58:88-91, 1983.<br />

24. Goldhaber P: The effect <strong>of</strong> hyperoxia on bone resorption<br />

in tissue culture. AMA Archives Pathol<br />

66:635-64 1, 1958.<br />

25. Graham DR, Keldermans MM, Klemm LW, et al:<br />

Infectious complications among patients receiving<br />

home intravenous therapy with peripheral, central<br />

or peripherally placed central venous catheter. Am<br />

J Med 91:95S-lOOS, 1991.<br />

26. Gregory EM, Fridovich I: Induction <strong>of</strong> superoxide<br />

dismutase by molecular oxygen. J Bacteriol<br />

114:543-548, 1973.<br />

27. Hall TJ, Chambers TJ: Molecular aspects <strong>of</strong> osteoclast<br />

function. Inflamm Res 45: 1-9,1996.<br />

28. Hill GB, Osterhout S: Experimental effects <strong>of</strong> hyperbaric<br />

oxygen on selected clostridial species. In<br />

vitro studies. J Infect Dis 125:17-25,1972.<br />

29. Hodgkin UG: Antibiotics in the treatment <strong>of</strong><br />

chronic staphylococcal osteomyelitis. South Med J<br />

68:817-823, 1975.<br />

30. Hunt TK, Pai MP: The effect <strong>of</strong> varying ambient<br />

oxygen tensions on wound metabolism and collagen<br />

synthesis. Surg Gynecol Obstet 135:756-758,<br />

1972.<br />

31. Hunt TK, Zederfeldt B, Goldstick TK: Oxygen and<br />

healing. Am J Surg 118:521-525, 1969.<br />

32. Ingram C, Eron LJ, Goldberg RI: Antibiotic therapy<br />

<strong>of</strong> osteomyelitis in outpatients. Med Clin North<br />

Am 72:723-738, 1988.<br />

33. Jackson MA, Nelson JD: Etiology and medical<br />

management <strong>of</strong> acute suppurative bone and joint<br />

infections in pediatric patients. J Pediatr Orthop<br />

2:313-323, 1982.<br />

34. Jordan CW, Kawachi MM: Analysis <strong>of</strong> serum bactericidal<br />

activity in endocarditis, osteomyelitis, and<br />

other bacterial infections. Medicine 6049-61, 1981.<br />

35. Kass EH, Evans DA: Future prospects and past<br />

problems in antimicrobial therapy: The role <strong>of</strong> cefoxitin.<br />

Rev Infect Dis l:l-2, 1979.<br />

36. Kirby WMM, Regamey C: Pharmacokinetics <strong>of</strong><br />

cefazolin compared with other cephalosporins. J<br />

Infect Dis 128(Suppl):341-346, 1973.<br />

37. Kolyvas E, Ahronheim G, Marks MI, et al: Oral antibiotic<br />

therapy <strong>of</strong> skeletal infections in children.<br />

Pediatrics 655367-871, 1980.<br />

38. LeFrock JL, Klainer AS, Chen S, et al: The spectrum<br />

<strong>of</strong> colitis associated with lincomycin and clindamycin<br />

therapy. J Infect Dis 131(Suppl):108-115,<br />

1975.<br />

39. Lew DP, Waldvogel FA: Quinolones and osteomyelitis:<br />

State-<strong>of</strong>-the-art. Drugs 49(Suppl<br />

2):100-111,1995.<br />

40. Mader JT Fluoroquinolones in Bone and Joint Infections.<br />

In Sanders Jr WE, Sanders CC (eds). Fluoroquinolones<br />

in the <strong>Treatment</strong> <strong>of</strong> Infectious<br />

Diseases. Glenview, IL, Physicians & Scientists<br />

71-86,1990.<br />

41. Mader JT, Adams KR: Experimental <strong>Osteomyelitis</strong>.<br />

In Schlossberg D (ed). Orthopaedic Infection.<br />

Springer Verlag Publishers. New York 3948, 1987.<br />

42. Mader JT, Adams KR, Couch LA, et al: Potentiation<br />

<strong>of</strong> tobramycin by hyperbaric oxygen in experimental<br />

Pseudomonas aeruginosa osteomyelitis<br />

(Abstract I33 1). Abstracts <strong>of</strong> the 27th Interscience<br />

Conference on <strong>Antimicrobial</strong> Agents and<br />

Chemotherapy. Washington, DC, American Society<br />

for Microbiology 1987.<br />

43. Mader JT, Adams KR, Sutton TE: Infectious diseases:<br />

Pathophysiology and mechanisms <strong>of</strong> hyperbaric<br />

oxygen. J Hyperbaric Med 2:133-140, 1987.<br />

44. Mader JT, Brown GL, Guckian JC, et al: A mechanism<br />

for the amelioration by hyperbaric oxygen <strong>of</strong><br />

experimental staphylococcal osteomyelitis in rabbits.<br />

J Infect Dis 142:915-922, 1980.<br />

45. Mader JT, Cantrell IS, Calhoun JH: Oral<br />

cipr<strong>of</strong>loxacin compared with standard parenteral<br />

antibiotic therapy for chronic osteomyelitis in<br />

adults. J Bone Joint Surg 72A:104-110, 1990.<br />

46. Mader JT, Guckian JC, Glass DL, et al: Therapy<br />

with hyperbaric oxygen for experimental osteomyelitis<br />

due to Staphylococcus aureus in rabbits.<br />

J Infect Dis 138:312-318, 1978.<br />

47. Mader JT, Hulet WA, Reinarz JA: Potential value<br />

<strong>of</strong> hyperbaric oxygen in inoperable and intra-abdominal<br />

sepsis. 20th Interscience Conference on<br />

<strong>Antimicrobial</strong> Agents and Chemotherapy. New<br />

York 631, 1980.<br />

48. Mader JT, Ortiz M, Calhoun JH: Update on the diagnosis<br />

and management <strong>of</strong> osteomyelitis. Clin Podiatr<br />

Med Surg 13:701-724, 1996.<br />

49. Mainous EG: Osteogenesis enhancement utilizing<br />

hyperbaric oxygen therapy. HBO Review<br />

3: 181-185,1982.<br />

50. McCord JM, Keele BB, Fridovich I: An enzyme<br />

based theory <strong>of</strong> obligate anaerobiasis: The physiologic<br />

function <strong>of</strong> superoxide dismutase. Proc Natl<br />

Acad Sci 68:l02&1027,1971.<br />

5 1. Morrey BF, Dunn JM, Heimbach RD, et al: Hyperbaric<br />

oxygen and chronic osteomyelitis. Clin Orthop<br />

144:121-127,1979<br />

52. Murray BE The life and times <strong>of</strong> the Enterococcus.<br />

Clin Microbiol Rev 3:4M5, 1990.<br />

53. Musher DM, Fletcher T Tolerant Staphylococcus<br />

aureus causing vertebral osteomyelitis. Arch Intern<br />

Med 142:631-634, 1982.<br />

54. National Committee for Clinical Laboratory Standards:<br />

Methods for dilution antimicrobial susceptibility<br />

tests for bacteria that grow aerobically<br />

(Tentative Standard: M7-T). Villanova, PA 1983.<br />

55. Neu HC: Aminopenicillins: Clinical pharmacology<br />

and use in disease states. Int J Clin Pharmacol Biopharm<br />

11:132-144,1975.<br />

56. Neu HC: Clinical pharmacokinetics in preventive<br />

antimicrobial therapy. South Med J 7O(Suppl):<br />

14-24,1977.<br />

57. Neu HC: Carbenicillin and ticarcillin. Med Clin<br />

North Am 66:61-77, 1882.<br />

58. Neu HC, Fu KP: Clavulanic acid: Anovel inhibitor<br />

<strong>of</strong> Beta lactamase. Antimicrob Agents Chemother<br />

14:650-655, 1978.<br />

59. Neu HC, Gamey GG: Comparative in vitro activity<br />

and clinical pharmacology <strong>of</strong> ticarcillin and carbenicillin.<br />

Antimicrob Agents Chemother 8:<br />

457-462, 1975.


64 Maderetal<br />

60. Neu HC, Labthavikul P: Antibacterial activity and<br />

beta-lactam stability <strong>of</strong> ceftazidime, an aminothiazolyl<br />

cephalosporin potentially active against<br />

Pseudomonas aeruginosa. Antimicrob Agents<br />

Chemother 21 : 11-18. 1982.<br />

61. Neu HC. Labthavikul P: Comparative in vitro<br />

evaluation <strong>of</strong> N-formimidoyl thienamycin against<br />

Gram-positive and Gram-negative aerobic and<br />

anaerobic species and its P-lactamase stability.<br />

Antimicrob Agents Chemother 21: 180-187,<br />

1982.<br />

62. Norden CW: Experimental osteomyelitis V. Therapeutic<br />

trials with oxacillin and sisomicin alone<br />

and in combination. J Infect Dis 137:<br />

155-160,1978.<br />

63. Norden CW: <strong>Osteomyelitis</strong>. In Mandell GL, Douglas<br />

RG, Bennett JE (eds). Principles and Practice<br />

<strong>of</strong> Infectious Diseases. Ed 4. New York, Churchill<br />

Livingstone 922-930, 1990.<br />

64. Norden CW, Bryant R, Palmer D, Montgomerie<br />

JZ, Wheat J: <strong>Chronic</strong> osteomyelitis caused by<br />

Staphylococcus aureus: Controlled clinical trial <strong>of</strong><br />

nafcillin therapy and nafcillin-rifampin therapy.<br />

South Med J 79:947-95 1, 1986.<br />

65. Norden CW, Shinners E, Niederriter K: Clindamycin<br />

treatment <strong>of</strong> experimental chronic osteomyelitis<br />

due to Staphylococcus aureus. J Infect<br />

Dis 153:956-959, 1986.<br />

66. Onorato lM, Axelrod JL: Hepatitis from intravenous<br />

high-dose oxacillin therapy. Ann Intern<br />

Med 89:497-500, 1978.<br />

67. Pany MF. Folta D: The in vitro activity <strong>of</strong> mezlocillin,<br />

against community hospital isolates in<br />

comparison to other penicillins and cephalosporins.<br />

J Antimicrob Chemother 11 (Suppl):97-102,1983.<br />

68. Pein FD, Vosti KL: Variation in performance <strong>of</strong><br />

the serum bactericidal test. Antimicrob Agent<br />

Chemother 6:300-333, 1974.<br />

69. Pryka RD, Rodvold KA, Rotschafer JC: Teicoplanin:<br />

An investigational glycopeptide antibiotic.<br />

Clin Pharm 7:647-658, 1988.<br />

70. Reading C, Hepbum P: The inhibition <strong>of</strong> staphylococcal<br />

beta-lactamase by clavulanic acid.<br />

Biochem J 179:67-76, 1979.<br />

71 Reller LB, Stratton CW: Serum dilution test for<br />

bactericidal activity. II. Standardization and correlation<br />

with antimicrobial assays and susceptibility<br />

tests. J Infect Dis 136:196-204, 1977.<br />

72. Reynolds AV, Hamilton-Miller JMT, Brumfitt W:<br />

Diminished effect <strong>of</strong> gentamicin under anaerobic<br />

and hypercapnic conditions. Lancet 1 :447-449,<br />

1976.<br />

73. Richmond MM, Sykes RB: The beta-lactamase <strong>of</strong><br />

Gram negative bacteria and their possible role.<br />

Adv Microb Physiol9:31-88, 1975.<br />

74. Rosenblatt JE: Laboratory tests used to guide antimicrobial<br />

therapy. Mayo Clin Proc 52:611-615,<br />

1977.<br />

75 Rosenblatt JE, Edson RS: Metronidazole. Mayo<br />

Clin Proc 58:154-157, 1983.<br />

76. Sabath LD: Staphylococcal to Penicillins and<br />

Cephalosporins. In Schlessinger D (ed). Microbiology.<br />

Washington, DC, American Microbiological<br />

Society 299, 1976.<br />

Clinical Orthopaedics<br />

and Related Research<br />

77. Sabath LD: Chemical and physical factors influencing<br />

methicillin resistance <strong>of</strong> Staphylococcus<br />

aureus and Staphylococcus epidermidis. J Antimicrob<br />

Chemother 3(Suppl C):47-51, 1977.<br />

78. Sabath LD, Garner C. Wilcox C, Findland M:<br />

Susceptibility <strong>of</strong> Staphylococcus aureus and<br />

Staphylococcus epidermidis to 65 antibiotics. Antimicrob<br />

Agents Chemother 9:962-969, 1976.<br />

79. Sabath LD, Wheeler N, Laveridire M, Blazevic D,<br />

Wilkinson BJ: A new type <strong>of</strong> penicillin resistance<br />

<strong>of</strong> Staphylococcus aureus. Lancet I :443447,<br />

1977.<br />

80. Sanders CC, Sanders WE, Goering RV: In vitro<br />

studies with SCH 21420 and SCH 22591: Activity<br />

in comparison with six other aminoglycosides and<br />

synergy with penicillin against enterococci. Antimicrob<br />

Agents Chemother 14:178-184. 1978.<br />

81. Schamberg DR, Dillon WI, Terpenning MS, et a]:<br />

Increasing resistance <strong>of</strong> enterococci to<br />

cipr<strong>of</strong>loxacin. Antimicrob Agents Chemother<br />

36:2533-2535, 1992.<br />

82. Schlicter JG, MacLean H: A method <strong>of</strong> determining<br />

the effective therapeutic level in the treatment<br />

<strong>of</strong> subacute bacterial endocarditis with penicillin.<br />

Am Heart J 34:209-211, 1947.<br />

83. Schreiner A, Tonjum S, Digranes A: Hyperbaric<br />

oxygen therapy in bacteroides infections. Acta<br />

Chir Scand 140:73-76, 1974.<br />

84. Scully BE, Neu HC: Clinical efficacy <strong>of</strong> ceftazidime.<br />

<strong>Treatment</strong> <strong>of</strong> serious infection due to<br />

multiresistant pseudomonas and other Gram- negative<br />

bacteria. Arch Intern Med 144:57-62, 1984.<br />

85. Sheftel TJ, Mader JT, Pennick JJ, Cierny G: Methicillin-resistant<br />

Staphylococcus aureus osteomyelitis.<br />

Clin Orthop 188:23 1-239, 1985.<br />

86. Shinto Y, Uchida A, Korkusaz F, Araki N, Ono K<br />

Calcium hydroxyapatite ceramic used as a drug<br />

delivery system for antibiotics. J Bone Joint Surg<br />

74Bz600-604, 1992.<br />

87. Shirtliff ME, Mohan D, Calhoun JH, Mader IT:<br />

Four week intravenous antibiotic therapy versus<br />

two week intravenous plus four week oral antibiotic<br />

therapy in the treatment <strong>of</strong> long bone osteomyelitis.<br />

Musculoskeletal Infection Society<br />

Annual Meeting, Snowmass, CO 1997.<br />

88. Shlaes DM, Binczewski B: Enterococcal resistance<br />

to vancomycin and related cyclic glycopeptide<br />

antibiotics. Eur J Clin Microbiol Infect Dis<br />

9: 106-1 10,1990.<br />

89. Sorrel1 TC, Packham DR, Shanker S, Foldes M,<br />

Munro R: Vancomycin therapy for methicillin-resistant<br />

Staphylococcus aureus. Ann Intern Med<br />

971344-350, 1982.<br />

90. Stratton CW, Reller LB: Serum dilution test for<br />

bactericidal activity. I. Selection <strong>of</strong> a physiologic<br />

diluent. J Infect Dis 136: 187-195, 1977.<br />

9 I . Summersgill JT, Schupp LG, Raff MJ: Comparative<br />

penetration <strong>of</strong> metronidazole, clindamycin,<br />

chloramphenicol, cefoxitin, ticarcillin, and moxalactam<br />

into bone. Antimicrob Agents Chemother<br />

211601-603,1982<br />

92. Sutter VL: In vitro susceptibility <strong>of</strong> anaerobes:<br />

Comparison <strong>of</strong> clindamycin and other antimicrobial<br />

agents. J Infect Dis 135(Suppl):7-12, 1977.


Number 360<br />

March, 1999 <strong>Antimicrobial</strong> <strong>Treatment</strong> <strong>of</strong> <strong>Osteomyelitis</strong> 65<br />

93. Sykes RB, Bonner DP, Bush K. Aztreonam: A synthetic<br />

monobactam active against aerobic Gramnegative<br />

bacteria. Antimicrob Agents Chemother<br />

21:85-92, 1982.<br />

94. Tetzl<strong>of</strong>f TR, McCracken GH, Nelson FD: Oral antibiotic<br />

therapy for skeletal infections in children.<br />

11. Therapy <strong>of</strong> osteomyelitis and suppurative<br />

arthritis. J Pediatr 92:485490, 1978.<br />

95. Thornsberry C: Review <strong>of</strong> in vitro activity <strong>of</strong> thirdgeneration<br />

cephalosporins and other newer Betalactam<br />

antibiotics against clinically important<br />

bacteria. Am J Med 79(Suppl 2A):14-20, 1985.<br />

96. Verklin RM, Mandell GL: Alteration <strong>of</strong> effectiveness<br />

<strong>of</strong> antibiotics by anaerobiosis. J Lab Clin<br />

Med 89:65-71, 1977.<br />

97. Wagstaff AJ, Balfour JA: Grepafloxacin. Drugs<br />

825-827,1997.<br />

98. Waldvogel FA, Med<strong>of</strong>f G, Swartz MN: <strong>Osteomyelitis</strong>:<br />

A review <strong>of</strong> clinical features, therapeutic<br />

consideration, and unusual aspect. N Engl J<br />

Med. 282:260-266,1970,<br />

99. Wallyn RJ, Gumbiner SH, Goldfein S, et al: <strong>Treatment</strong><br />

<strong>of</strong> anaerobic infections with hyperbaric oxygen.<br />

Surg Clin North Am 44:107-112, 1964.<br />

100. Ward TT, Winn RE, Hartstein AI, Sewell DL: Observations<br />

relating to an inter-hospital outbreak <strong>of</strong><br />

methicillin-resistant Staphylococcus aureus: Role<br />

<strong>of</strong> antimicrobial therapy in infection control. Infect<br />

Control 2:453-459, 1981.<br />

01. Wilson SG, Sanders CC: Selection and characterization<br />

<strong>of</strong> strains <strong>of</strong> Staphylococcus aureus displaying<br />

unusual resistance to aminoglycosides.<br />

Antimicrob Agents Chemother 10:519-525,<br />

1976.<br />

02. Wormser GP, Keusch GT: Trimethoprim-sulfamethoxazole<br />

in the United States. Ann Intern<br />

Med 91:420429,1979.<br />

103. Yablon IG, Cruess RL: The effect <strong>of</strong> hyperbaric<br />

oxygen on fracture healing in rats. 8:<br />

186-202,1968.<br />

104. Yow MD, Taber LH, Barrett FF, et al: A ten-year<br />

assessment <strong>of</strong> methicillin-associated side-effects.<br />

Pediatrics 58:329-334, 1976.<br />

105. Zhang X, Wyss UP, Pichora D, Goosen MP:<br />

Biodegradable controlled antibiotic release devices<br />

for osteomyelitis: Optimization <strong>of</strong> release<br />

properties. J Pharm Pharmacol 46:718-724,<br />

1994.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!