You are on page 1of 18

Antimicrobial therapy for patients with severe sepsis and septic

shock: An evidence-based review


Pierre-Yves Bochud, MD; Marc Bonten, MD; Oscar Marchetti, MD; Thierry Calandra, MD, PhD

Objective: In 2003, critical care and infectious disease experts grades from A to E, with A being the highest grade. Pediatric
representing 11 international organizations developed manage- considerations to contrast adult and pediatric management are in
ment guidelines for antimicrobial therapy for patients with severe the article by Parker et al. on p. S591.
sepsis and septic shock that would be of practical use for the Conclusion: Since the prompt institution of therapy that is active
bedside clinician, under the auspices of the Surviving Sepsis against the causative pathogen is one of the most important predic-
Campaign, an international effort to increase awareness and tors of outcome, clinicians must establish a system for rapid admin-
improve outcome in severe sepsis. istration of a rationally chosen drug or combination of drugs when
Design: The process included a modified Delphi method, a con- sepsis or septic shock is suspected. The expanding number of
sensus conference, several subsequent smaller meetings of sub- antibacterial, antifungal, and antiviral agents available provides op-
groups and key individuals, teleconferences, and electronic-based portunities for effective empiric and specific therapy. However, to
discussion among subgroups and among the entire committee. minimize the promotion of antimicrobial resistance and cost and to
Methods:The modified Delphi methodology used for grading maximize efficacy, detailed knowledge of the likely pathogens and
recommendations built on a 2001 publication sponsored by the the properties of the available drugs is necessary for the intensivist.
International Sepsis Forum. We undertook a systematic review of (Crit Care Med 2004; 32[Suppl.]:S495–S512)
the literature graded along five levels to create recommendation

I n 2001, the International Sepsis the empirical management of patients ics; 6) need for empirical antifungal ther-
Forum published guidelines on with severe sepsis or septic shock. apy in patients with severe sepsis; 7)
the management of patients with Under the auspices of the Surviving analyses of the efficacy and safety of con-
severe sepsis and septic shock, in- Sepsis Campaign, a joint venture of the ventional vs. lipid formulations of am-
cluding an evidence-based review on an- International Sepsis Forum, the Society photericin B and of azoles and echinocan-
tibiotic therapy (1). The recommenda- of Critical Care Medicine, and the Euro- dins vs. amphotericin B for the treatment
tions on antibiotic therapy for sepsis pean Society of Intensive Care Medicine, of fungal sepsis; and 8) impact of antibi-
comprised sections on the epidemiology the International Sepsis Forum guide- otic cycling on morbidity and mortality of
of sepsis and the impact of appropriate lines have been updated and extended. sepsis.
antimicrobial therapy on outcome of The revised guidelines extend, but do not
Gram-negative, Gram-positive, and fun- entirely replace, the previously published Materials and Methods
gal sepsis and an evidence-based review of article (1), as several parts of the initial
monotherapy vs. combination therapy for document are still up-to-date. As for the Selection of Panel, Topics of Interest,
original publication, the revised guide- and Review Process. The panel was com-
lines were developed using an evidence- posed of three board-certified infectious
based review of the literature. The pro- diseases specialists, with expertise in the
From The Institute for Systems Biology, Seattle, cesses used by the panel of experts to fields of antimicrobial therapy, sepsis, in-
WA (PYB); the Department of Internal Medicine, Divi- select and review the relevant literature tensive care medicine, and infection con-
sion of Acute Internal Medicine and Infectious Dis-
eases, University Medical Center Utrecht, Utrecht, The
are outlined in the Material and Methods trol, who have been selected by the co-
Netherlands (MB); Infectious Diseases Service, Depart- section. The panel elected to focus on the chairs of the “Diagnosis and Management
ment of Internal Medicine, Centre Hospitalier Univer- following topics: 1) impact of appropriate of Infection in the Severe Sepsis” track of
sitaire Vaudois, Lausanne, Switzerland (OM, TC). antibiotic therapy on the outcome of sep- Surviving Sepsis Campaign. The panel-
Supported by grants from the Swiss Foundation for
Medical and Biological Grants to PYB (1121), from the
sis; 2) impact of pharmacokinetic and ists, chairpersons, and directors of the
Swiss National Science Foundation to PYB (81LA- pharmacodynamic principles on treat- Surviving Sepsis Campaign discussed the
65462) and to TC (3100 – 066972), the Bristol-Myers ment outcome; 3) need for prompt initi- specific scopes of the guidelines during a
Squibb Foundation, the Santos-Suarez Foundation for ation of antimicrobial therapy in patients 2-day meeting (June 27–28, 2003). Iden-
Medical Research, and the Leenaards Foundation. TC
is a recipient of a career award from the Leenaards
with severe sepsis and septic shock; 4) tified topics of interest were distributed
Foundation. analyses of the efficacy and toxicity of among the panelists based on individual
Copyright © 2004 by the Society of Critical Care monotherapy vs. combination therapy as expertise. Each panelist either alone or in
Medicine and Lippincott Williams & Wilkins empirical therapy; 5) indications for em- collaboration with colleagues wrote sev-
DOI: 10.1097/01.CCM.0000143118.41100.14 pirical use of anti-Gram-positive antibiot- eral sections of the guidelines, which

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S495


were assembled by the panel chair and tients with Gram-positive infections and Epidemiological Features of
reviewed and critiqued by the panelists severe sepsis. A Medline search using the Severe Sepsis and Septic
and track co-chairs. terms clinical trial and either linezolid or Shock
Data Source. Medline was used to quinupristin/dalfopristin and Synercid
search articles published between 1966 yielded 100 and 41 hits, respectively. Data on secular epidemiologic trends
and July 1, 2003. Medline searches and Selection of Articles. Abstracts of all of the etiological agents and most fre-
selections of articles were performed by articles satisfying the selection criteria quent sites of sepsis have not changed
each of the authors. Medical Subject were reviewed to exclude irrelevant stud- since the 2001 review and will, therefore,
Heading (MeSH; http://www.nlm.nih.gov/ ies. Review articles and articles on topics not be reviewed in detail again (1).
mesh/meshhome.html) terms used in the such as antibiotic prophylaxis, pharma- Microorganisms and Sites of Infec-
Medline search were sepsis (comprising cology, microbiology, oncology, hematol- tion. By the mid-1980s, the frequency of
the terms septicemia, sepsis syndrome, ogy, immunology, mediators of inflam- Gram-positive sepsis (mainly caused by
septic shock, bacteremia, fungemia, par- mation, allergy, catheter management, Staphylococcus aureus, coagulase-nega-
asitemia, and viremia), pneumonia (com-
animal studies, chronic infections, or tive staphylococci, enterococci, and
prising the terms bronchopneumonia,
specific infections (acquired immunode- streptococci) has equaled that of Gram-
pleuropneumonia, aspiration pneumonia,
ficiency syndrome, endocarditis, chronic negative sepsis (mainly caused by Enter-
bacterial pneumonia, viral pneumonia,
salmonellosis, viral infections in organ obacteriaceae, especially Escherichia coli
and Pneumocystis carinii pneumonia).
Since no ad hoc MeSH term exists for transplant patients, hemorrhagic fever, and Klebsiella pneumoniae, and by
intraabdominal infections, we used the viral hepatitis, parasitic infections, and Pseudomonas aeruginosa). Recent epide-
words and MeSH terms intraabdominal, malaria) were excluded if they did not miologic data in the United States and in
abdomen, abdominal infection, peritoni- fulfill the inclusion criteria. For the com- Europe indicated that Gram-positive bac-
tis, appendicitis, abdominal abscess to parison of monotherapy vs. combination teria have now surpassed Gram-negative
search for articles on intraabdominal in- therapy, articles on each of the three clin- bacteria as etiological agents of sepsis (9,
fections. The Medline search was then ical entities reviewed (i.e., sepsis, pneu- 10), confirming a trend observed in many
narrowed by using the MeSH terms anti- monia, and intraabdominal infections) recent sepsis studies (4, 5, 11–17). Fungi
infective agents (comprising the term were selected only if there was unequiv- account for about 5% of all cases of sep-
antibiotics, which was exploded to in- ocal evidence that patients had either sis, severe sepsis, and septic shock (1).
clude all classes of antibiotics and all clinically or microbiologically docu- Most cases of fungal sepsis are caused by
antibiotic names), clinical trials, or mented infections and if the study met at Candida species, which are the fourth
randomized controlled trials, further least one of the following criteria: 1) a most common bloodstream pathogens in
limiting the search to human studies definition of sepsis or severe sepsis con- all recent U.S. studies of nosocomial
and English literature. The MeSH key- sistent with the definition of the Consen- bloodstream infections and are associated
words agranulocytosis, antibiotic pro- sus Conference of the American College with the highest mortality (40%) of all
phylaxis, and ambulatory care were of Chest Physicians and the Society of bloodstream pathogens (18, 19). The in-
used to exclude studies on neutropenic Critical Care Medicine (7); 2) sepsis with cidence of fungal sepsis has increased
patients, antibiotic prophylaxis, and at least one organ dysfunction or sign of three-fold between 1979 and 2000 (10).
outpatient therapy. Additional articles hypoperfusion present in ⬎50% of the However, due to the paucity of published
were retrieved from references of arti- patients; or 3) an overall mortality data on national surveys of fungal infec-
cles identified by the Medline search ⬎10%. This cutoff for mortality was cho- tions, it is unclear whether the incidence
and of guidelines and review articles on sen to ensure that articles included a of fungal sepsis has increased in a similar
the following topics: sepsis, communi- substantial proportion of patients with fashion in other parts of the world. Dutch
ty-acquired pneumonia, ventilator- severe sepsis and not simply sepsis. To investigators reported a two-fold increase
associated pneumonia, and intraab-
ensure that articles on antimicrobial in the incidence of candidemia in the
dominal infections. Epidemiologic data
therapy had been properly selected by one early 1990s (20). In contrast, a Norwegian
were extracted from articles identified
of the authors (PYB), a random sample of survey also conducted in the early 1990s
by a Medline search using the keywords
20% of the articles identified by the Med- reported stable incidence of candidemia
epidemiology and sepsis and by a sys-
tematic review of 27 clinical trials of line search were examined by the panel and Candida species distribution (21).
antiinflammatory or mediator-targeted chair (TC). Overall agreement between Likewise, a recent survey of the incidence
therapies in patients with severe sepsis the two reviewers was 94% for articles on of candidemia in Switzerland has shown
and septic shock (2– 6). intraabdominal infections (␬ statistic of that the incidence of candidemia re-
For the section on empirical therapy 0.71) and 96% for articles on communi- mained stable between 1991 and 2000
for Gram-positive severe sepsis or septic ty-acquired pneumonia and ventilator- (median, 0.5 episodes/10,000 patient-
shock, a Medline search was performed associated pneumonia (␬ statistic of 0.81) days) (22). By decreasing order of fre-
using the terms empirical combined with corresponding to “substantial” and “al- quency, the predominant sites of infec-
either vancomycin or teicoplanin, which most perfect” agreements, respectively, tions in patients with severe sepsis and
revealed one and 33 hits, respectively. according to the criteria proposed by septic shock are the lungs, the blood-
Extension of the search to the terms em- Sackett et al. (8). Consensus was reached stream (i.e., without another identifiable
pirical, glycopeptide, and clinical trials between the two authors on the articles source of infection), the abdomen, the
yielded 20 hits. However, none of these for which there was disagreement regard- urinary tract, and the skin and soft tis-
articles included nonneutropenic pa- ing the inclusion criteria. sues.

S496 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


Need for Prompt Initiation of 18% in 1,454 patients who received appro- ally ⬍100 per trial), a majority of these
Antimicrobial Therapy in priate antibiotics (p ⬍ .0001) (28). Smaller patients with Gram-negative sepsis, and
Patients with Severe Sepsis and recent studies showed that the appropriate- did not include intent-to-treat analyses.
Septic Shock ness of the antibiotic regimen favorably in- Studies performed in the 1990s were
fluenced the outcome of patients infected larger, included patients with both Gram-
Rationale: Establishing vascular access with specific Gram-negative bacteria, such positive and Gram-negative sepsis, and in
and initiating aggressive fluid resuscita- as Enterobacter species (29), P. aeruginosa most instances, included an intent-to-treat
tion is the first priority when managing (30), and ceftazidime-resistant K. pneu- analysis. Given the relatively small number
patients with severe sepsis or septic moniae or E. coli (31). of studies available and the fact that the
shock. However, prompt infusion of anti- Fewer data have been published on the sepsis subset of studies included patients
microbial agents is also a logical strategy impact of appropriate antibiotic therapy with various types of infections, including
and may require additional vascular ac- in patients with Gram-positive sepsis community-acquired and ventilator-associ-
cess ports. Establishing a supply of pre- (32). Several studies evaluated the impact ated pneumonias and intraabdominal in-
mixed antibiotics in an emergency de- of appropriate antimicrobials in patients fections, all studies were pooled.
partment or critical care unit for such with severe infections due to Gram- Among the 79 studies identified, 67
urgent situations is an appropriate strat- negative and Gram-positive bacteria (33– met the inclusion criteria for severe sep-
egy for enhancing the likelihood that an- 44). In all but one study (42), appropriate sis and/or septic shock due to bacterial
timicrobial agents will be infused antibiotic therapy was associated with a infections and were grouped in four main
promptly. Staff should be cognizant that better outcome. categories: 1) studies comparing different
some agents require more lengthy infu- combination therapies, usually a ␤-lac-
sion time, whereas others can be rapidly Empirical Antimicrobial Therapy tam antibiotic or a fluoroquinolone plus
infused or even administered as a bolus. for Patients with Severe Sepsis an aminoglycoside (n ⫽ 9; sepsis, 4;
and Septic Shock pneumonia, 5) (54 – 62); 2) studies com-
Impact of Appropriate Antibiotic paring a monotherapy with a combina-
Therapy on Outcome of Sepsis The evidence-based review of the lit- tion therapy with an aminoglycoside (n ⫽
erature on empirical antimicrobial ther- 24; sepsis, 11; pneumonia, 7; intraab-
Recommendations: Initial empirical anti- apy for patients with severe sepsis and dominal infections, 6) (63– 86); 3) studies
infective therapy should include one or septic shock has focused on three clinical comparing single-agent therapies (n ⫽
more drugs that have activity against the entities: sepsis (i.e., primary or secondary 26; sepsis, 14; pneumonia, 11; intraab-
likely pathogens (bacterial or fungal) and bloodstream infections), pneumonia dominal infections, 1) (56, 87–111); and
that penetrate into the presumed source (both community-acquired and hospital- 4) miscellaneous studies (n ⫽ 8; pneu-
of the sepsis. The choice of drugs should acquired), and intraabdominal infections. monia, 7; sepsis, 1) (112–119).
be guided by the susceptibility patterns of Management guidelines and review arti-
microorganisms in the community and cles have been published recently on Monotherapy Vs. Combination
in the hospital. these topics (45–53), which should be Antibiotic Therapy
used as a supplement to the present arti-
Grade D Recommendation: Monotherapy is as ef-
cle, which focuses on clinical trials of
ficacious as combination therapy with a
Rationale: Early administration of appro- antimicrobial agents for patients with se-
␤-lactam and an aminoglycoside as em-
priate antibiotics reduces mortality in pa- vere sepsis or septic shock.
pirical therapy of patients with severe
tients with Gram-positive and Gram- A total of 2,065 articles (sepsis, 988;
sepsis or septic shock.
negative bacteremias. Retrospective studies community-acquired pneumonia and
Carbapenem: Grade B. Third- or
conducted in the 1960s and in the 1970s ventilator-associated pneumonia, 509; in-
fourth-generation cephalosporins: Grade
have shown that appropriate antimicrobial traabdominal infections, 393; Gram-
B. Extended-spectrum carboxypenicillins
therapy, defined as the use of at least one positive infections, 175) were identified
or ureidopenicillins combined with ␤-lac-
antibiotic active in vitro against the caus- using the Medline search strategy de-
tamase inhibitors: Grade E
ative bacteria, reduced the mortality of scribed in Material and Methods, of which
Gram-negative bacteremia when compared 1,464 did not meet our inclusion criteria Rationale. The use of antibiotic combina-
with patients receiving inappropriate ther- based on the content of the abstract (Fig. tions for the treatment of severe infec-
apy (23–26). In a landmark study of 173 1). A full text review was performed on tions relies on the following rationale: 1)
patients with Gram-negative bacteremia, the remaining 614 articles, of which 79 a combination of two antibiotics broad-
who were classified in three categories (13%) (sepsis, 37; community-acquired ens the antibacterial spectrum, which
based on the severity of the underlying dis- pneumonia and ventilator-associated may be important as treatment is usually
ease categories (i.e., rapidly fatal, ultimately pneumonia, 35; and intraabdominal in- initiated empirically in critically ill septic
fatal, and nonfatal), McCabe et al. (23) ob- fections, 7) met the inclusion criteria of patient; 2) combination of antibiotics
served that appropriate antibiotic therapy severe sepsis or septic shock. Thus, only a may exert additive or synergistic effects
reduced mortality from 48% to 22%. Four small proportion of the screened clinical against the infecting pathogen, resulting
subsequent studies that included larger trials of antibiotic therapy have included in enhanced antibacterial activity and
numbers of patients yielded similar results critically ill septic patients with high possibly better clinical response (120 –
(24 –27). In a recent prospective study of mortality. The overall mortality of the 122); 3) the use of a combination of an-
2,124 patients with Gram-negative bactere- patients in the selected studies was 18%. tibiotics may reduce the emergence of
mia, mortality was 34% in 670 patients Most studies performed before 1990 com- resistant bacteria (123) or of superinfec-
who received inappropriate antibiotics and prised a limited number of patients (usu- tions (124).

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S497


in the imipenem plus netilmicin treat-
ment groups (65). However, nephrotoxic-
ity was significantly more frequent in the
aminoglycoside-containing treatment
arm. In three studies of severe sepsis or
intraabdominal sepsis, imipenem was
found to be at least as efficacious as and
less nephrotoxic (one study) than a com-
bination of gentamicin or tobramycin
and clindamycin or metronidazole (63,
81, 82).
Third-Generation and Fourth-Gener-
ation Cephalosporins. In ten prospective,
randomized, controlled studies, mono-
therapy with a third- or fourth-genera-
tion cephalosporin (cefotaxime, moxalac-
tam, cefoperazone, or ceftazidime) was as
effective as combination therapy with a
␤-lactam (usually an extended-spectrum
penicillin or a cephalosporin) and an ami-
noglycoside (67–71, 77, 78) or clindamy-
cin and an aminoglycoside (76, 83, 84). In
four of the ten studies, clinical success
Figure 1. Study flow. Studies were searched in Medline as indicated in Material and Methods. Abstracts rates were significantly higher in the
were reviewed, and studies that satisfied the inclusion criteria were selected for a full text review. monotherapy treatment groups (69, 76 –
MRGP, multiple-resistant Gram-positive bacteria.
78). However, it is difficult to draw firm
conclusions from these observations as
In the late 1960s and early 1970s, a otics, such as the extended-spectrum different ␤-lactams were used in the
series of retrospectives studies examined penicillins, third-generation or fourth- monotherapy and combination therapy
the potential benefits of combination generation cephalosporins, or the carbap- treatment groups in all but one study.
therapy (27, 125). Overall, combination enems, has substantially reduced the Mortality rates were similar in the mono-
therapy was not found to be superior to need for aminoglycoside-containing anti- therapy and combination therapy treat-
single-agent therapy, but some benefits biotic combinations. During the last two ment groups in nine of the ten studies. A
of antibiotic combinations were noted in decades, most antibiotic studies have, trend toward a lower mortality in the
subsets of patients, such as those with therefore, compared the efficacy and tox- monotherapy treatment arm was ob-
rapidly or ultimately fatal diseases. How- icity of single-agent antibiotic therapies served in one trial (77). As expected,
ever, the impact of these results for to- (i.e., monotherapies) with that of ␤-lac- nephrotoxicity was higher in the amin-
day’s practice is limited given the retro- tams paired with aminoglycosides (i.e., oglycoside-containing regimen in seven
spective nature of these studies and the combination therapies). Twenty-four of the ten studies, reaching statistical sig-
facts that it utilized antibiotics that studies of empirical monotherapy vs. nificance in five.
would no longer be considered appropri- combination therapy were identified by Extended-Spectrum Penicillins with
ate today and that it did not include mul- the Medline search. The results obtained Anti-Pseudomonas Activity. In four pro-
tivariate analyses. Subsequent studies with carbapenems, third-generation or spective, randomized, controlled studies,
evaluated the efficacy of different antibi- fourth-generation cephalosporins, and extended-spectrum carboxypenicillins (ti-
otic combinations, usually a ␤-lactam extended-spectrum penicillins with activ- carcillin) or ureidopenicillins (piperacil-
and an aminoglycoside, for the treatment ity against Pseudomonas species are lin) used either alone or in combination
of Gram-negative infections (54 –59, 61, shown in Table 1. with a ␤-lactamase inhibitor (clavulanic
62, 126). By and large, there were no Carbapenems. In five prospective, ran- acid or tazobactam) were found to be as
differences between the various antibiotic domized, controlled studies, mono- effective as amoxicillin-clavulanate, pip-
regimens in terms of clinical success, mi- therapy with imipenem-cilastatin or eracillin, piperacillin-tazobactam, or clin-
crobiological eradication, and mortality meropenem was shown to be as effective damycin combined with an aminoglyco-
rates. However, in one study of 204 pa- as a combination of a ␤-lactam (cefu- side (gentamicin, amikacin, or
tients with ventilator-associated pneumo- roxime, ceftazidime, or imipenem) and netilmicin) as empirical antibiotic ther-
nia, the clinical cure rate of piperacillin/ an aminoglycoside (gentamicin, amika- apy for patients with intraabdominal in-
tazobactam plus amikacin was superior cin, or netilmicin) (Table 1) (64 – 66, 74, fections, pneumonia, or neonatal sepsis
to that of ceftazidime plus amikacin (26/ 75). It is worth mentioning that the same (72, 79, 85, 86) (Table 1). However, as
51, 51%, vs. 23/64, 36%; p ⫽ .009), but ␤-lactam antibiotic (imipenem) was used noted for the carbapenem or cephalospo-
mortality was similar in the two treat- in both treatment arms in only one of rin studies, in three of these four studies,
ment groups (8/51, 16%, vs. 13/64, 20%; these studies, in which the success rates, different ␤-lactam antibiotics were used
p ⫽ .4) (60). the occurrence of superinfections, and of in the monotherapy and combination
Since the 1980s, the advent of broad- P. aeruginosa resistant to the carbap- therapy treatment groups. In a study of
spectrum and highly bactericidal antibi- enem were similar in the imipenem and 206 patients with intraabdominal sepsis,

S498 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


clinical success, and mortality and neph- even though the benefit of combination rates for the various cephalosporins were
rotoxicity rates were similar in patients therapy for the latter indication re- in the range of 65% to 85% in most
treated with piperacillin-tazobactam or mains controversial. Yet, the benefit studies, except for slightly lower response
with piperacillin-tazobactam plus amika- from additive or synergistic effects of rates for ceftriaxone and cefotaxime in
cin (86). antibiotic combination and the possible two smaller studies (101, 104).
Fluoroquinolones. One study showed prevention of emerging resistant bacte- Aminoglycosides, Monobactams, or
that monotherapy with ciprofloxacin was ria must be weighed against the risk of Fluoroquinolones. In the late 1970s and
as effective as therapy with a ␤-lactam increased toxicity. Indeed, aminoglyco- early 1980s, three studies compared the
and/or an aminoglycoside for the treat- side-containing regimens have been efficacy and safety of different aminogly-
ment of patients with documented Gram- shown repeatedly to increase the risk of cosides (gentamicin vs. tobramycin, gen-
negative sepsis (80). However, given that nephrotoxicity or ototoxicity. tamicin vs. amikacin, and netilmicin vs.
first-generation fluoroquinolones exhibit amikacin) as single-agent therapy for
suboptimal activities against Gram- Single-Agent Antibiotic Gram-negative sepsis (87– 89). Clinical
positive bacteria, this class of antibiotics Therapies responses rates, overall mortality, and
should not be used as empirical single- nephrotoxicity were similar among the
agent therapy in patients with severe sep- Recommendation: Third-generation and various aminoglycosides compared in
sis. fourth-generation cephalosporins, car- these studies. A high incidence of amin-
Comments. The evidence-based re- bapenems, and extended-spectrum car- oglycoside-induced nephrotoxicity and
view of the literature tends to suggest boxypenicillins or ureidopenicillins com- ototoxicity were noted in these studies,
that monotherapy with a broad-spectrum bined with ␤-lactamase inhibitors are which is a major concern in critically ill
␤-lactam antibiotic is as efficacious and equally effective as empirical antibiotic septic patients already at high risk of de-
less nephrotoxic than a combination of a therapy in patients with severe sepsis. veloping organ failures due to the circu-
␤-lactam and an aminoglycoside as em- Third- or fourth-generation cephalo- latory collapse associated with severe sep-
pirical therapy for critically ill patients sporins: Grade A. Carbapenem: Grade B. sis or septic shock. In two small studies,
presenting with severe sepsis or septic Extended-spectrum carboxypenicillins or aztreonam was found to be as effective
shock. However, monotherapy should not ureidopenicillins combined with ␤-lacta- and less toxic than aminoglycosides as
be regarded as a universal remedy to be mase inhibitors: Grade C. empirical monotherapy of patients with
used indiscriminately for several reasons. Carbapenems, Third-Generation, or severe Gram-negative sepsis (90, 100).
Compared with the abundant literature Fourth-Generation Cephalosporins. Six However, today, aminoglycosides or a
on empirical therapy for febrile neutro- prospective, randomized, clinical trials monobactam antibiotic such as aztreo-
penia, few studies have been conducted in (of which five included between 150 and nam should not be used as single-agent
patients with severe sepsis or septic 400 patients) were identified that investi- empirical therapy of severe sepsis because
shock. Moreover, many of the clinical tri- gated the efficacy and safety of a carbap- of their narrow antibacterial activity and
als reviewed here included ⬍200 pa- enem (imipenem or meropenem), a risk of toxicity (aminoglycosides).
tients, and the statistical power was, third-generation cephalosporin (ceftazi- More recently, five studies, of which
therefore, limited. Furthermore, rarely dime or cefotaxime combined with met- four enrolled only patients with severe
was the same ␤-lactam antibiotic used in ronidazole), a ureidopenicillin with a community-acquired or nosocomial
the control and experimental treatment ␤-lactamase inhibitor (piperacillin-ta- pneumonias, showed that fluoroquinolo-
groups, thus limiting the conclusions zobactam), or another carbapenem (Ta- nes (ciprofloxacin, four studies; levofloxa-
that could be drawn about the apparent ble 2) (92, 95–97, 99, 107, 111). In all but cin, one study) were as efficacious as
equivalent efficacy of the two regimens. one study, virtually identical clinical re- ␤-lactam antibiotics (imipenem in four
There is clearly a need for large, prospec- sponse rates and overall mortality were studies; various ␤-lactams with or with-
tive, randomized trials to assess the effi- observed in patients treated with either out an aminoglycoside in one study) for
cacy and toxicity of new antibiotics in imipenem or meropenem or control the treatment of patients with severe sep-
critically ill septic patients. ␤-lactam antibiotics. In a smaller clinical sis caused predominantly by Gram-
One should keep in mind that the trial of 94 patients with intraabdominal negative bacteria (Table 2) (56, 105, 106,
choice of empirical antibiotic therapy sepsis, the clinical response rate of pa- 109, 110). Of note, a large number of
depends on several factors related to tients treated with meropenem was supe- patients (ranging between 250 and 400)
the patient’s history (including drug in- rior to that of those treated with cefo- were randomized in three of the four
tolerance), underlying diseases, and taxime plus metronidazole (111). pneumonia studies. However, given the
susceptibility patterns of microorgan- In summary, as one might have antic- limited activity of first-generation fluoro-
isms of the hospital environment and ipated given the fairly comparable anti- quinolones (i.e., norfloxacin and cipro-
patient’s community. The initial selec- microbial spectrum of activity of most floxacin) against Gram-positive bacteria
tion of an empirical antimicrobial reg- third-generation and fourth-generation and the possibility of resistance among
imen should be broad enough to cover cephalosporins, the studies conducted Gram-negative bacteria, these antibiotics
all likely pathogens. Despite a lack of with these agents for the treatment of cannot be recommended as empirical
clearcut advantage, clinicians may still patients with severe sepsis and commu- monotherapy for patients with severe
prefer to rely initially on a ␤-lactam and nity-acquired or nosocomial (including sepsis or septic shock of unknown etiol-
aminoglycoside combination, especially ventilator-associated) pneumonia or mis- ogy, including patients with community-
in the context of high level of antibiotic cellaneous types of infections have acquired or healthcare-associated pneu-
resistance or when treating patients yielded very comparable results (Table 2) monia. The most recent, so-called
with suspected Pseudomonas infection, (91,93,94,98,101–104,108). Response respiratory fluoroquinolones (i.e., levo-

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S499


Table 1. Monotherapy versus combination antibiotic therapy as empirical treatment of severe sepsis and septic shock

Authors and Yr of Type of Experimental Control Success (%)


Publication Infection Therapy Therapy (Exp. vs. Control) p

Carbapenems vs. ␤-lactams or antianaerobes combined with an aminoglycoside


Solomkin et al. 1985 (63) SEPSIS Imipenem Gentamicin ⫹ clindamycin 28/37 (76) vs. 27/37 (73) 1.000
Poenaru et al. 1990 (81) ABDOM Imipenem Tobramycin ⫹ clindamycin — —
or metronidazole
Solomkin et al. 1990 (82) ABDOM Imipenem Tobramycin ⫹ clindamycin 67/81 (83) vs. 57/81 (70) .094
Mouton et al. 1990 (64) SEPSIS Imipenem Cefotaxime ⫹ amikacin 58/70 (83) vs. 54/70 (77) .527
a
Cometta et al. 1994 (65) SEPSIS Imipenem Imipenem ⫹ netilmycin 113/142 (80) vs. 119/138 (86) .155
Sieger et al. 1997 (74) HAP Meropenem Ceftazidime ⫹ amikacin 76/106 (72) vs. 62/105 (59) .061
Jaspers et al. 1998 (66) SEPSIS Meropenem Cefuroxime ⫹ gentamicin 27/39 (69) vs. 25/40 (63) .637
⫹/⫺ metronidazole
Alvarez-Lerma et al. 2001 (75) HAP Meropenem Ceftazidime ⫹ amikacin 47/69 (68) vs. 39/71 (55) .121
Third- or fourth-generation cephalosporins vs. ␤-lactams or antianaerobes combined with an aminoglycoside
Arich et al. 1987 (67) SEPSIS Cefotaxime Cefazolin ⫹ tobramycin 22/25 (88) vs. 17/22 (77) .446
Schentag et al. 1983 (83) ABDOM Moxalactam Tobramycin ⫹ clindamycin 37/49 (76) vs. 36/49 (73) 1.000
a
Oblinger et al. 1982 (68) SEPSIS Moxalactam Conventional therapy 33/38 (87) vs. 32/40 (80) .547
Mangi et al. 1988 (76) HAP Cefoperazone Clindamycin or cefazolin 41/46 (89) vs. 44/61 (72) .052
and gentamicin
Greenberg et al. 1994 (84) ABDOM Cefoperazone- Clindamycin ⫹ gentamicin 33/47 (70) vs. 15/29 (52) .143
sulbactam
b
Fernandez-Guerrero et al. 1991 (77) HAP Cefotaxime Combination therapy 217/275 (79) vs. 193/273 (71) .030
Croce et al. 1993 (78) HAP Ceftazidime or Ceftazidime or cefoperazone 22/39 (56) vs. 22/70 (31) .015
cephoperazone ⫹ gentamicin
Rubinstein et al. 1995 (69) SEPSIS Ceftazidime Ceftriaxone ⫹ tobramycin 227/306 (74) vs. 179/274 (65) .023
d
Extermann et al. 1995 (70) SEPSIS Ceftazidime “Best guess” combination 38/41 (93) vs. 28/30 (93) 1.000
McCormick et al. 1997 (71) SEPSIS Ceftazidime Mezlocilin ⫹ netilmicin 50/65 (77) vs. 48/63 (76) 1.000
Antipseudomonas penicillins vs. ␤-lactams or anti-anaerobes combined with an aminoglycoside
Fink 1991 (85) ABDOM Ticarcillin-clavulanate Gentamicin ⫹ clindamycin 15/20 (75) vs. 16/25 (64) .525
Hammerberg et al. 1989 (72) SEPSIS Piperacillin Ampicillin ⫹ amikacin — —
Speich et al. 1998 (79) CAP ⫹ HAP Piperacillin- Amoxiclav ⫹ netilmicin or 37/41 (90) vs. 36/43 (84) .521
tazobactam gentamicin
Dupont et al. 2000 (86) ABDOM Piperacillin- Piperacillin-tazobactam ⫹ 44/99 (44) vs. 55/105 (52) .266
tazobactam amikacin
Miscellaneous monotherapies versus ␤-lactams combined with an aminoglycoside
Korvick et al. 1992 (73) SEPSIS f
␤-lactam or ␤-lactam ⫹ aminoglycoside — —
aminoglycoside
alone
Manhold et al. 1998 (80) HAP Ciprofloxacin Ceftazidime ⫹ gentamicin — —

p, two-tailed Fisher’s exact test; ABDOM, intraabdominal infections; CAP, community-acquired pneumonia; HAP, hospital-acquired pneumonia
(ventilator-associated or not).
a
Conventional therapy included a ␤-lactam (i.e., penicillin, ampicillin, ticarcillin, nafcillin, cefamandole, cefoxitin) given either alone or in combination
with an aminoglycoside (tobramycin or amikacin); bcombination therapy comprised a ␤-lactam (a cephalosporin or a “broad-spectrum” penicillin) with an
aminoglycoside (gentamicin, tobramycin, or amikacin); cinfection-related mortality; dbest guess combination usually include a ␤-lactam with an
aminoglycoside (not specified); f␤-lactam included a cephalosporin, imipenem, or an extended-spectrum penicillin; aminoglycosides included gentamicin,
tobramycin, amikacin, or kanamycin.

floxacin, gatifloxacin, moxifloxacin, or spectrum penicillin) with or without an cocci, or ampicillin-resistant entero-
gemifloxacin) exhibit enhanced in vitro aminoglycoside. cocci) in the community or in the
activities against Gram-positive bacteria. hospital.
Numerous studies have shown that these Empirical Use of Anti-Gram-
agents are highly efficacious as single- Grade E
Positive Antibiotics in Patients
agent therapy of community-acquired with Severe Sepsis Rationale: Antimicrobial coverage of both
pneumonia in the outpatient or inpatient Gram-positive and Gram-negative bacte-
setting (48, 50, 127). When used for the Recommendation: Empirical use of gly- ria is mandatory in the empirical treat-
treatment of severe community-acquired copeptide antibiotics (vancomycin, teico- ment of critically ill patients with severe
or nosocomial infections in intensive care planin), oxazolidinones (linezolid), or
sepsis. Naturally, many broad-spectrum
unit (ICU) patients in whom Pseudomo- streptogramins (quinupristin/dalfopris-
antibiotics reliably cover both pathogens
nas infection is a concern, ciprofloxacin tin) in patients with severe sepsis or sep-
and, therefore, can be used for empirical
or the respiratory fluoroquinolones tic shock is justified in patients with hy-
should preferentially be combined with persensitivity to ␤-lactams or in therapy. The choice of empirical treat-
an anti-Pseudomonas antibiotic (i.e., a institutions with resistant Gram-positive ment, however, should depend on the lo-
carbapenem, a third- or fourth-genera- bacteria (i.e., methicillin-resistant staph- cal epidemiology of pathogens associated
tion cephalosporin, or an extended- ylococci, penicillin-resistant pneumo- with infections and their antimicrobial

S500 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


Table 1. Continued.

Mortality (%) (Exp. vs. Control) p Nephrotoxicity (%) (Exp. vs. Control) p

6/37 (16) vs. 4/37 (11) .736 1/37 (3) vs. 10/27 (27) .007
4/52 (8) vs. 9/52 (17) .235 — —

11/81 (14) vs. 14/81 (17) .664 — —


7/70 (10) vs. 7/70 (10) 1.000 1/105 (1) vs. 4/102 (4) .369
18/142 (13) vs. 13/138 (9) .448 0/158 (0) vs. 6/149 (4) .014
13/104 (13) vs. 23/107 (21) .100 0/104 (0) vs. 2/105 (2) .498
3/39 (8) vs. 4/40 (10) 1.000 2/39 (5) vs. 5/35 (13) .432

16/69 (23) vs. 20/71 (28) .564 0/69 (0) vs. 2/69 (3) .497

8/25 (32) vs. 5/22 (23) .530 0/25 (0) vs. 3/19 (14) .095
7/49 (14) vs. 6/49 (12) 1.000 6/49 (12) vs. 14/35 (29) .078
8/33 (24) vs. 9/32 (28) .783 3/41 (7) vs. 11/36 (23) .046
9/61 (15) vs. 13/71 (18) .645 6/61 (10) vs. 12/66 (15) .447

6/47 (13) vs. 3/29 (10) 1.000 0/47 (0) vs. 1/28 (3) .382

36/275 (13) vs. 52/273 (19) .063 0/275 (0) vs. 7/266 (3) .007
c
2/39 (5) vs. 7/70 (10) .485 5/39 (13) vs. 25/45 (36) .013

31/306 (10) vs. 33/274 (12) .508 0/306 (0) vs. 9/265 (3) .001
6/41 (15) vs. 4/30 (13) 1.000 — —
13/65 (20) vs. 9/63 (14) .484 2/65 (3) vs. 8/55 (13) .053

3/20 (15) vs. 5/25 (20) .716 1/20 (5) vs. 0/25 (0) .444
17/200 (9) vs. 27/196 (14) .110 50/200 (25) vs. 43/153 (22) .480
1/41 (2) vs. 6/43 (14) .110 0/44 (0) vs. 2/43 (4) .494

19/99 (19) vs. 22/105 (21) .862 3/99 (3) vs. 3/102 (3) 1.000

24/118 (20) vs. 20/112 (18) .738 — —

13/28 (46) vs. 6/23 (26) .158 — —

susceptibility. It is, therefore, essential to icillin-resistant S. pneumoniae is also in- tin) on a routine basis in all patients with
get frequent feedbacks from the microbi- creasing in many areas of the world. Yet, severe sepsis and septic shock?
ology laboratory on antibiotic susceptibil- the relevance of reduced susceptibility of Glycopeptides. To the best of our
ities trends. Major changes in the relative Streptococcus pneumoniae to penicillin knowledge, randomized trials comparing
importance of Gram-positive and Gram- on clinical outcome of patients treated empirical treatment with or without
negative infections among critically ill with ␤-lactam antibiotics is uncertain, Gram-positive coverage (including glyco-
patients have occurred during the last 10 especially for respiratory tract infections peptides) have never been performed in
yrs. While Gram-negative bacteria pre- (130). Clinical failures, however, have adult nonneutropenic patients and such
dominated until the middle 1980s, Gram- been reported in patients with bacterial an approach would probably be judged as
positive bacteria now account for at least meningitis caused by a penicillin-resis- unethical. None of the articles retrieved
one-half of the infections occurring in by the Medline search included a compar-
tant S. pneumoniae treated with ceftriax-
patients with severe sepsis (128, 129). ison of empirical treatment with or with-
one (131). Does this epidemiologic con-
Moreover, methicillin-resistant S. aureus out Gram-positive patients in nonneutro-
text justify the empirical use of
(MRSA) and methicillin-resistant Staph- penic adults with severe sepsis. Although
ylococcus epidermidis are responsible for glycopeptides (vancomycin or teicopla- the indiscriminate use of glycopeptides
a majority of staphylococcal infections in nin), oxazolidinones (linezolid), or for presumed Gram-positive infections in
some institutions. The frequency of pen- streptrogramins (quinupristin/dalfopris- patients with severe sepsis or septic shock

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S501


Table 2. Comparison of monotherapies as empirical treatment of severe sepsis and septic shock

Authors and Yr of Type of Experimental Success (%) Mortality (%)


Publication Infection Therapy Control Therapy (Exp. vs. Control) p (Exp. vs. Control) p

Carbapenems versus third-generation cephalosporins or carbapenems


Norrby et al. 1993 (92) SEPSIS Imipenem Ceftazidime 27 /197 (64) vs. 124/196 (63) .834 20/197 (10) vs. 31/196 (16) .101
Kempf et al. 1996 (111) ABDOM Meropenem Cefotaxime ⫹ 41 /43 (95) vs. 30/40 (75) .012 3/43 (7) vs. 5/40 (13) .473
metronidazole
Colardyn and Faulkner 1996 (95) SEPSIS Meropenem Imipenem 68 /90 (76) vs. 67/87 (77) .861 24/106 (23) vs. 17/98 (17) .385
Mehtar et al. 1997 (96) SEPSIS Meropenem Ceftazidime ⫹ 46 /48 (96) vs. 40/43 (93) .664 10/68 (15) vs. 11/63 (17) .812
metronidazole
Garau et al. 1997 (97) SEPSIS Meropenem Imipenem 55 /66 (83) vs. 49/67 (73) .208 22/76 (29) vs. 26/75 (35) .488
a
Jaccard et al. 1998 (107) HAP Imipenem Piperacillin-tazobactam 56 /79 (71) vs. 62/75 (83) .091 6/79 (8) vs. 7/75 (9) .777
Verwaest 2000 (99) SEPSIS Meropenem Imipenem 67 /87 (77) vs. 62/91 (68) .240 22/107 (21) vs. 14/105 (13) .201
Third- or fourth-generation cephalosporins vs. a third-generation cephalosporin
Mangi et al. 1988 (91) SEPSIS Cefoperazone Ceftazidime 48 /62 (77) vs. 54/63 (86) .256 23/62 (37) vs. 24/63 (38) 1.000
Reeves et al. 1989 (101) HAP Ceftriaxone Cefotaxime 12 /25 (48) vs. 19/26 (73) .089 2/25 (8) vs. 4/26 (15) .668
Mangi et al. 1992 (102) HAP Ceftriaxone Cefoperazone 35 /50 (70) vs. 48/60 (80) .269 12/50 (24) vs. 10/60 (17) .351
Thomas et al. 1992 (103) HAP Cefotaxime Ceftriaxone 8 /12 (67) vs. 10/15 (67) 1.000 1/12 (8) vs. 5/15 (33) .182
Norrby and Geddes 1993 (93) SEPSIS Cefpirome Ceftazidime 131 /176 (74) vs. 34/50 (68) .372 28/282 (10) vs. 10/80 (13) .536
Barckow et al. 1993 (104) CAP ⫹ HAP Cefepime Cefotaxime 27 /37 (73) vs. 10/18 (56) .231 13/37 (35) vs. 4/18 (22) .372
Schrank et al. 1995 (94) SEPSIS Cefepime Ceftazidime 11 /13 (85) vs. 12/15 (80) 1.000 1/13 (8) vs. 2/15 (13) 1.000
Norrby et al. 1998 (98) SEPSIS Cefpirome Ceftazidime 123 /188 (65) vs. 132/188 (70) .377 15/188 (8) vs. 23/188 (12) .231
Grossman et al. 1999 (108) CAP Cefepime Ceftriaxone 53 /67 (79) vs. 46/61 (75) .676 7/76 (9) vs. 7/75 (9) 1.000
Fluoroquinolones vs. ␤-lactams
b
Fink et al. 1994 (105) CAP ⫹ HAP Ciprofloxacin Imipenem 92 /202 (46) vs. 90/200 (45) .921 43/202 (21) vs. 38/200 (19) .620
Siami et al. 1995 (106) CAP ⫹ HAP Ciprofloxacin Imipenem 17 /24 (71) vs. 14 /21 (67) 1.000 2/24 (8) vs. 3/21 (14) .652
Krumpe et al. 1999 (56) SEPSIS Ciprofloxacin Several ␤-lactams 138 /166 (83) vs. 74/87 (85) .858 26/207 (13) vs. 13/99 (13) .857
Torres et al. 2000 (109) HAP Ciprofloxacin Imipenem 40 /57 (70) vs. 34/52 (65) .683 8/41 (20) vs. 4/34 (12) .529
West et al. 2003 (110) HAP Levofloxacin Imipenem followed by 135 /204 (66) vs. 143/206 (69) .526 38/220 (17) vs. 32/218 (15) .515
(IV then oral) oral ciprofloxacin

p, two-tailed Fisher’s exact test; CAP, community-acquired pneumonia; HAP, hospital-acquired pneumonia (ventilator-associated or not).
a
Infection-related mortality; bsame study as Snydam et al. 1994.

should be avoided, their use is appropri- Gram-negative coverage, at least until namely the need for empirical coverage of
ate in severely ill patients in the following microbiological results are available. VRE and documented hypersensitivity to
circumstances: 1) endemic levels of Empirical use of either vancomycin or glycopeptides. The use of linezolid is ap-
MRSA; 2) documented hypersensitivity to teicoplanin has been common practice in proved for infections with VRE (including
␤-lactam antibiotics; and 3) treatment of the management of neutropenic patients bacteremia), community-acquired and
bacterial meningitis in areas with high with fever, in whom coagulase-negative nosocomial pneumonia caused by S.
levels of penicillin-resistance of S. pneu- staphylococci and viridans streptococci pneumoniae or S. aureus (including
moniae. Naturally, “endemic” and “high” are predominant pathogens. Although MRSA), and skin and soft tissue infec-
levels of resistance are subjective, and no some studies have suggested that the use tions caused by Gram-positive bacteria.
studies have been performed to deter- of glycopeptides at the initiation of em- Four randomized trials comparing the
mine the level of resistance above which pirical therapy might be beneficial, a re- efficacy of linezolid with alternative treat-
the empirical use of antibiotics active cent prospective, randomized, double- ment in adult nonneutropenic patients
against these resistant pathogens is war- blind trial failed to demonstrate clinical have been identified (Table 3). In an
ranted. There probably is an inverse rela- benefits (132). Prolonged courses of van- open-labeled randomized trial, San Pedro
tionship between the severity of sepsis comycin therapy have been associated and coworkers (136) compared linezolid
and the willingness to accept a risk not to with the development of vancomycin- (sequential intravenous-to-oral therapy)
cover an antibiotic-resistant pathogen resistance S. aureus, either through re- with ceftriaxone IV followed by oral cef-
empirically. However, the possible clini- duced cell wall permeability (133) or podoxime for patients hospitalized with
cal benefit associated with the empirical through acquisition of the vanA gene community-acquired pneumonia. Epi-
use of glycopeptides should be weighed from vancomycin-resistant enterococci sodes of community-acquired pneumonia
against the risks of selecting resistant mi- (VRE) (134). were primarily caused by S. pneumoniae,
croorganisms and of increased toxicity, Linezolid and Quinupristin/Dalfopris- and overall clinical cure rates were
especially when vancomycin is used in tin. Linezolid and quinupristin/dalfopris- higher for linezolid-treated patients, es-
combination with an aminoglycoside or tin are recently introduced antibiotics for pecially for patients with S. pneumoniae
other nephrotoxic agents. To further re- the treatment of Gram-positive infec- bacteremia (93.1% vs. 68.2%; p ⫽ .021).
duce the risk of emergence of vancomy- tions. Linezolid is a new oxazolidinone In two double-blind, randomized trials, a
cin-resistant staphylococci, empirical antibiotic with activity against staphylo- combination of linezolid and aztreonam
vancomycin therapy should be rapidly cocci (including MRSA), and Enterococ- was compared with vancomycin and az-
discontinued in patients in whom Gram- cus faecium and Enterococcus faecalis treonam for the treatment of patients
positive infections have been ruled out. (including strains resistant to vancomy- with hospital-acquired pneumonia (119,
Finally, it is rarely, if ever, appropriate to cin) (135). Indications for the empirical 137). Although equivalence of activity
use vancomycin alone as empirical ther- use of linezolid are similar to that of was found in both trials, a subsequent
apy since most cases require additional glycopeptides plus two other indications, retrospective analysis of the combined

S502 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


Table 3. Clinical studies of linezolid in patients with Gram-positive infections

Authors and Yr of Type of Experimental Control Success (%) Survival (%) p


Publication Infection Therapy Therapy (Exp. vs. Control) p (Exp. vs. Control)

Rubinstein et al. 2001 (119) HAP Linezolid ⫹ Vancomycin ⫹ 71/107 (66.4)a vs. 62/91 (68.1)a NS 36/203 (18)b vs. 49/193 (25) .067
aztreonam aztreonam 86/161 (53)b vs. 74/142 (52)b .908
Stevens et al. 2002 (139) Known/suspected MRSA Linezolid Vancomcyin 109/192 (57)b vs. 93/169 (55)b NS b
40/240 (17) vs. 30/220 (14) b
NS
infections
Documented MRSA 41/56 (73) vs. 38/52 (73) NS
infections
San Pedro et al. 2002 (136) CAP due to S. Linezolid Ceftriaxon/ 316/381 (83) vs. 280/366 (76) .04 366/381 (4) vs. 347/366 (5) NS
pneumoniae cefpodoxim
Wunderink et al. 2003 (137) HAP Linezolid ⫹ Vancomycin ⫹ 135/256 (53)b vs. 128/245 (52)b NS 257/321 (80)b vs. 241/302 (80)b NS
aztreonam aztreonam 114 /168 (68)a vs. 111/171 (65)a 145/157 (92)a vs. 150 /166 (90)a
Wunderink et al. 2003 (138) Documented SA HAP Linezolid ⫹ Linezolid ⫹ 70/136 (51.5)a vs. 59/136 (43.4)a NS 131/168 (78) vs. 121/171 (71) NS
(n ⫽ 339) aztreonam aztreonam
Documented MRSA HAP 36/61 (59.0)a vs. 22/62 (35.5)a ⬍.01 60/75 (80) vs. 54/85 (64) .025
(n ⫽ 160)
SA HAP diagnosed by 47/92 (51)a vs. 39/90 (43)a NS 86/109 (79) vs. 82/114 (72) NS
invasive procedure
(n ⫽ 223)
MRSA HAP diagnosed by 19/33 (58)a vs. 13/39 (33)a .04 34/40 (85) vs. 37/55 (67) .05
invasive procedure
(n ⫽ 95)

HAP, hospital-acquired pneumonia (ventilator-associated or not); MRSA, methicillin-resistant Staphylococcus aureus; SA, Staphylococcus aureus.
a
In the clinically evaluable population; bIntention-to-treat analysis.

dataset revealed that initial therapy with randomized clinical trials of quinupris- quinupristin/dalfopristin alone as em-
linezolid was associated with significantly tin/dalfopristin have been identified pirical therapy of severe sepsis or septic
better survival and clinical cure rates in (Table 4). Fagon and coworkers (118) shock, since Gram-negative coverage is
patients with nosocomial pneumonia due compared quinupristin/dalfopristin needed, at least until microbiological
to MRSA (138). Finally, in an open- with vancomycin (aztreonam was al- results become available.
labeled randomized trial, Stevens and co- lowed in both treatment arms) in pa-
workers (139) compared the efficacy of tients with Gram-positive nosocomial Modification of Empirical
linezolid with that of vancomycin for the pneumonia. The second study was a Antimicrobial Therapy Based on
treatment of patients with suspected combined report of two nearly identical Culture Results
MRSA. Linezolid and vancomycin had open-label randomized trials in which
similar efficacy, both in an intention-to- patients with Gram-positive skin and Recommendation: Modification of empir-
treat analysis and in a subgroup of pa- skin structure infections were treated ical antimicrobial therapy with the aim to
tients with documented MRSA infections. with quinupristin/dalfopristin or with restrict the number of antibiotics and
Most patients, however, had skin or soft one of three comparator antibiotics (ce- narrow the spectrum of antimicrobial
tissue infections and probably did not ful- fazolin, vancomycin, or oxacillin) (143). therapy is an important and responsible
fill the criteria of severe sepsis. No clini- In both studies, clinical cure rates of strategy for minimizing the development
cal data are available on the use of lin- quinupristin/dalfopristin and of control of resistant pathogens and for containing
ezolid for bacterial meningitis in areas of antibiotics were similar. However, only of costs.
high levels of penicillin-resistant S. pneu- a few patients fulfilled the criteria of
Grade E
moniae. severe sepsis in the latter study (143). A
Resistance to linezolid is based on spe- formal head-to-head comparison of lin- To the best of our knowledge no stud-
cific point mutations in the 23S ribo- ezolid and quinupristin/dalfopristin has ies have been performed in which pa-
somal RNA of the 50S subunit of the not been performed. Therefore, empir- tients were randomized either to con-
ribosome preventing binding of linezolid ical therapy with quinupristin/dalfo- tinue to receive empirical broad-
(135). Resistance development of VRE pristin should be limited to severely ill spectrum antibiotic therapy or to be
and S. aureus during therapy was associ- patients with a high likelihood of infec- switched to a narrow-spectrum antibiotic
ated with the presence of indwelling pros- tion caused by vancomycin-resistant E. regimen selected on the basis of suscep-
thetic devices and prolonged courses of faecium or MRSA and to patients with tibility data of the causative pathogen. In
antimicrobial therapy (140, 141). More- documented or presumed hypersensi- a before-after study design, Ibrahim and
over, nosocomial spread of linezolid- tivity to linezolid. The indiscriminate coworkers (144) assessed the effects of a
resistant VRE has been reported as well use of linezolid or of quinupristin/ clinical guideline on the appropriateness
(142). Therefore, empirical linezolid ther- dalfopristin for presumed Gram-posi- of empirical therapy in patients with ven-
apy should be rapidly discontinued in pa- tive infections in patients with severe tilator-associated pneumonia. The clini-
tients in whom Gram-positive infections sepsis or septic shock should be cal guideline consisted of empirical ther-
have been ruled out. avoided. The possible clinical benefit apy with imipenem, ciprofloxacin, and
Quinupristin/dalfopristin is an in- associated with the empirical use of vancomycin, followed by narrowing of
jectable streptogramin available for the these agents should be weighed against antibiotic therapy when a microbiological
treatment of Gram-positive infections the risks of selection of resistant micro- cause of infection was identified and dis-
caused by S. aureus (including MRSA) organisms, toxicity, and increased continuation of antibiotic therapy after 7
and E. faecium (including VRE), but it treatment costs. Finally, it is rarely, if days when the clinical condition allowed.
has no activity against E. faecalis. Two ever, appropriate to use linezolid or Implementation of the clinical guideline

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S503


Table 4. Clinical studies of quinupristin/dalfopristin in patients with Gram-positive infections

Authors and Yr of Type of Experimental Control Success (%) Survival (%)


Publication Infection Therapy Therapy (Exp. vs. Control) p (Exp. vs. Control) p

Fagon et al. 2000 (118) HAP Quinup/Dalfop ⫹ Vancomycin ⫹ 49/87 (56)a vs. 49/84 (58)a NS NS
aztreonam aztreonam
b b b b
65/150 (43) vs. 67/148 (45) NS 38/150 (25) vs. 116/148 (22)
Nichols et al. 1999 (143) Complicated Gram- Quinup/Dalfop Cefazolin or 68.2%a vs. 70.7% NS
positive skin and vancomycin
structure infections or oxacillin

HAP, hospital-acquired pneumonia (ventilator-associated or not).


a
In the clinically evaluable population.

was associated with higher appropriate- Grade C The “real-life” experience with antibi-
ness of empirical therapy and a reduction otic cycling is sparse, and interpretation
Rationale: Antibiotic cycling has been
in the mean duration of therapy. How- of findings is seriously hampered by nu-
ever, the number of patients in whom proposed as a strategy to minimize the
merous methodologic problems in study
antibiotics were narrowed was not re- development of antibiotic resistance and design. These included a nonstructured
ported. thus to improve the patient’s outcome. antibiotic cycling scheme (148), evalua-
In fact, there are arguments both in The rationale is that the cyclic exposure tion of changes in empirical therapy in a
favor and against such an approach. On to different classes of antibiotics should before-after design (149), use of multiple
the one hand, narrowing of the antimi- prevent emergence of resistance by ex- antibiotics for cycling and different cy-
crobial spectrum may reduce the risk of posing the microbial flora to homoge- cling intervals (148, 150), presence of im-
selecting resistant microorganisms and neous selective antibiotic pressure during portant confounding factors, such as re-
treatment costs. On the other hand, mi- a limited period of time. This concept is duction in the antibiotic use (151), or
crobiological documentation of the etiol- based on several assumptions. First, it is changes in infection control strategies
ogy of sepsis is lacking in a significant assumed that antibiotic-resistant micro- (152).
proportion of sepsis cases, making it dif- organisms have a growth disadvantage Clinical Data. The impact of antibiotic
ficult, if at all possible, to narrow the when the selective antibiotic pressure is cycling on a patient’s outcome has been
antibiotic coverage based on clinical cri- withdrawn. Therefore, development of re- analyzed in three studies. Kollef and co-
teria only. sistance during exposure to a given anti- workers (149) compared the effects of re-
biotic will be counterbalanced during pe- placing ceftazidime by ciprofloxacin in
Recommendations: The antimicrobial riods of nonexposure. Second, should
regimen should always be reassessed after the empirical treatment of suspected
resistance develop during one period, ex- Gram-negative infections in two 6-month
48 –72 hrs on the basis of microbiological posure to another class of antibiotics in
and clinical data with the aim of using a periods. The incidence of ventilator-
the following cycle will eliminate the re- associated pneumonia (VAP) decreased
narrow-spectrum antibiotic to recant the sistant microorganisms. For this to oc-
development of resistance, to reduce tox- from 11.6% to 6.7% (p ⫽ .028), as did the
cur, it presupposes the absence of cross- incidences of VAP and bacteremia due to
icity, and to reduce costs. Once a caus- resistance (i.e., that the mechanism of
ative pathogen is identified, there is no antibiotic-resistant Gram-negative bacte-
resistance be different among different ria (4% to 0.9%, p ⫽ .013, and 1.7% to
evidence that combination therapy is classes of antibiotics). However, besides
more effective monotherapy. The dura- 0.3%, p ⫽ .12, respectively). However,
these theoretical considerations, antibi- outcome parameters such as survival and
tion of therapy should typically be 7–10
otics are just one of many factors that length of stay remained unaffected. In
days and guided by clinical response.
have an impact on the development of another trial, Raymond and coworkers
Grade E resistance. For example, changes in the (152) compared a 1-yr period of nonpro-
numbers of patients introducing resistant tocol-driven antibiotic use with a subse-
Recommendations: If the presenting clini- microorganisms into the unit or changes quent 1-yr period of rotating empirical
cal syndrome is determined to be due to a in compliance with hygienic measures antibiotic assignment. Different classes of
noninfectious cause, antimicrobial therapy
will also affect the emergence of antibi- antibiotics were used for different indica-
should be stopped promptly to minimize
otic resistance. The effectiveness of infec- tions (pneumonia vs. peritonitis or sepsis
the development of resistant pathogens and
tion control programs can be influenced of unknown origin) with rotation of an-
superinfection with other pathogenic or-
by changes in the workload of healthcare tibiotic combinations after 4 months. In-
ganisms.
workers or understaffing, which both will fection rates with resistant Gram-positive
Grade E lead to more contacts between patients and Gram-negative bacteria significantly
and healthcare workers and less adher- decreased, survival in the ICU increased,
Antibiotic Cycling ence to hand hygiene and thus to more and antibiotic rotation was identified as
transmission of pathogens (145–147). an independent predictor for survival in
Recommendations: At the present time, Whether the theoretical benefits of anti- logistic regression analysis. Yet, other
there is insufficient evidence to recom- biotic cycling hold true in daily practice confounding factors were changed as
mend the use of antibiotic cycling as a can only be tested in studies in which well. An antibiotic surveillance team was
strategy to reduce the development of there is careful control of confounding instituted in the second half of the first
antibiotic resistance. variables. study period, and alcohol hand dispensers

S504 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


were distributed at the start of the second Empirical Use of Anti-Fungal sepsis or septic shock, which does not
period. In the third study, Gruson and Agents in Patients with Severe justify the use of antifungal therapy on a
coworkers (151) investigated whether a Sepsis or Septic Shock routine basis, but only in selected subsets
new program of antibiotic use had an of septic patients at high risk for invasive
impact on the incidence of VAP caused by Recommendation: Empirical antifungal candidiasis.
antibiotic-resistant bacteria in a before- therapy should not be used on a routine
after study. In the second part of the basis in patients with severe sepsis or Treatment of Candidemia
study, ceftazidime and ciprofloxacin use septic shock, but it may be justified in
were restricted, antibiotics were rotated selected subsets of septic patients at high Recommendation: Azoles (fluconazole)
without favoring any one antibiotic, and risk for invasive candidiasis (155). and echinocandins (caspofungin) are as
each antibiotic prescription was deter- efficacious as and less toxic than ampho-
Grade E
mined by one of the two investigators. In tericin B deoxycholate for the treatment
addition to these measures, a policy of Rationale: Since the 1980s, fungi have of patients with candidemia. Albeit better
shorter duration of aminoglycoside ther- emerged worldwide as an increasingly tolerated, there is no evidence that lipid
apy was implemented and compliance frequent cause of nosocomial infections formulations of amphotericin B are supe-
with protocol was stimulated by daily in critically ill patients and are associated rior to amphotericin B deoxycholate for
meetings between of one of the two in- with significant morbidity and mortality the treatment of candidemia.
vestigators with critical care physicians, (156 –159). Candida was the fourth most
Azoles: Grade A; Echinocandins: Grade B;
weekly meetings with nurses to identify frequent cause of bloodstream infections
Lipid formulations of amphotericin B:
nosocomial problems, monthly evalua- in U.S. hospitals in the 1990s (18). About
Grade E
tion of antibiotic consumption, and one third of all episodes of candidemia
three-monthly evaluation of changes in occur in medical, surgical, or pediatric Rationale: Candidemia is associated with
resistance patterns. The rotation protocol ICUs (22, 160). Fungi were isolated from significant morbidity, prolonged hospital
included the variable use of a ␤-lactam 17% of the patients with ICU-acquired stay, long-term sequelae, and high crude
antibiotic (cefepime or piperacillin- infections in a 1-day point prevalence mortality rates (40% to 60%) (162, 163).
tazobactam or imipenem or ticarcillin study conducted in 1,417 ICUs in western Moreover, the presence of disseminated
during periods of at least 1 month) and European countries (129). However, it is infection is an independent prognostic
an aminoglycoside (amikacin or tobramy- unclear whether all these fungal isolates factor of fatal outcome in patients with
cin, netilmicin or isepamicin) for late- were the etiological agent of infections or candidiasis (164), and antifungal therapy
onset VAP. For early-onset VAP, prescrip- simply colonizing microorganisms. Clin- has been shown to reduce mortality (165,
tion was rotated on a monthly basis using ical manifestations of candidiasis are usu- 166). Therefore, recent management
either amoxicillin-clavulanic acid, cefo- ally not specific, and standard culture guidelines have recommended that all pa-
taxime, ceftriaxone, or cefpirome in com- techniques lack sensitivity. Detection of tients with candidemia be treated with
bination with an aminoglycoside or fos- Candida antigens (mannan and ␤-glu- antifungal agents (155, 167).
fomycin. Importantly, implementation of can), metabolites (arabinitol and eno- For decades, amphotericin B deoxy-
all these measures reduced overall anti- lase), or antibodies (anti-mannan) and cholate, a fungicidal compound targeting
biotic use by ⬎50%. In addition, antibi- amplification of fungal DNA by polymer- ergosterol in the cell membrane of a
otic resistance levels of isolated patho- ase chain reaction have been or are cur- broad spectrum of fungi, has been the
gens decreased, both for antibiotic use rently under investigation. The facts that agent of choice for the empirical therapy
that had been reduced (i.e., ciprofloxacin Candida infections are difficult to diag- of invasive fungal infections. However,
and ceftazidime) and for antibiotics use nose and associated with severe morbid- nephrotoxicity and infusion-related ad-
that had been increased (i.e., cefepime ity and high mortality are arguments in verse events, especially fever and rigors,
and piperacillin-tazobactam). Incidences favor of the empirical or preemptive use are frequent adverse events of conven-
of microbiologically confirmed VAP de- of antifungal agents in critically ill ICU tional amphotericin B therapy that have
creased from 22.1% to 15.7% (p ⬍ .01) patients at high risk of candidiasis. The limited its use.
among patients requiring mechanical likelihood of fungal sepsis is increased in In the late 1980s, the advent of azoles
ventilation for ⬎48 hrs, but ICU mortal- patients who have been treated with sev- constituted a major progress in the man-
ity did not change (40.6% to 37.2%; p ⫽ eral broad-spectrum antibiotics, who are agement of invasive mycoses. This class
NS). colonized with Candida at multiple sites, of drugs inhibits the synthesis of ergos-
In summary, none of these studies who have damaged physiologic barriers terol. Fluconazole and itraconazole be-
have taken into account the most impor- (i.e., recurrent gastrointestinal perfora- came available in the early 1990s. Differ-
tant factors contributing to the develop- tions or anastomotic leakages, acute ne- ent clinical studies compared the efficacy
ment of dynamics of antibiotic resistance crotizing pancreatitis, chemotherapy- of fluconazole and amphotericin B deoxy-
in the hospital settings (i.e., relative im- induced mucositis, vascular access cholate for the treatment of candidemia.
portance of introduction, cross-transmis- devices, and total parenteral nutrition), Azoles. In a multicenter study of 206
sion, and endogenous acquisition by se- or who are immunosuppressed (i.e., can- nonneutropenic patients with candi-
lective antibiotic pressure), precluding a cer patients with neutropenia, or hema- demia, fluconazole (400 mg/day) was
reliable assessment of the role played by topoietic stem cells or solid organ trans- shown to be as efficacious (success rates
antibiotic cycling. Furthermore, the op- plant recipients) (155, 161). However, were 72% and 79%, respectively) as and
timal choice of antibiotics and cycle in- recent epidemiologic studies and multi- better tolerated than amphotericin B de-
tervals remain to be determined (153, center trials have shown that fungi ac- oxycholate (0.5– 0.6 mg/kg/day) (168).
154). count for only 5% of all cases of severe Similar findings were made in a prospec-

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S505


tive observational candidemia study in azole was found to be at least as effective against resistant strains awaits the results
which 227 patients were treated with am- as fluconazole (182). Moreover, voricon- of a recently completed large multicenter
photericin B deoxycholate (median daily azole salvage therapy showed an overall study in patients with candidemia. Given
dose, 0.5– 0.7 mg/kg) and 67 patients treatment success rate of 55% and 61% the poor prognosis of fungal sepsis, clini-
with fluconazole (median daily dose, 200 in patients with refractory systemic or cians have shown great interest for the
mg) (165) and in several other smaller esophageal candidiasis, respectively use of combinations of antifungal agents
comparative studies (169 –171). Noncom- (183). A large multicenter study compar- of different classes for the treatment of
parative studies yielded similar success ing voriconazole with sequential use of critically ill patients with invasive myco-
rates for fluconazole (172–178). In sum- amphotericin B deoxycholate followed by ses. However, up to now, there have been
mary, fluconazole (400 mg/day) was fluconazole in nonneutropenic patients relatively few in vitro or in vivo studies of
found to be as effective as and better with candidemia has recently been com- combinations of antifungal agents. While
tolerated than amphotericin B deoxy- pleted, and the first results of that trial awaiting the results of prospective, ran-
cholate (used at a dose of 0.3–1.2 mg/kg/ indicate that voriconazole is as effective domized clinical trials demonstrating
day) for the treatment of candidemia or as and was better tolerated than ampho- that combinations of antifungals are su-
invasive candidiasis. Whether higher tericin B/fluconazole. Posaconazole and perior to and reasonably not more toxic
doses of fluconazole would be associated ravuconazole are two other triazoles in than treatment with single agents, the
with better response rates is unknown. In clinical development. undiscriminating use of these costly
a study of 65 patients with Candida albi- Echinocandins. Echinocandins are a treatment regimens should be discour-
cans bloodstream infections, clinical re- new class of antifungal agents that act by aged.
sponse rates were 60% and 83% for pa- inhibiting the synthesis of ␤-(1, 3)-D- Lipid Formulations of Amphotericin
tients treated with 5 or 10 mg/kg/day of glucan, a component of the fungal cell B. Nephrotoxicity and infusion-related
fluconazole (179). The favorable efficacy- wall (184). Caspofungin, the first repre- reactions are frequent adverse events of
toxicity profile of fluconazole led to a sentative of this new family of antifungal treatment with amphotericin B deoxy-
rapidly increasing use of this azole for agents, is active against Candida (C. al- cholate, and treatment interruptions be-
prophylaxis and treatment of invasive bicans and non-C. albicans) and Aspergil- cause of toxicity may have a negative im-
candidiasis. As anticipated, in the late lus species. In immunosuppressed, pact on efficacy. In noncomparative
1990s an increasing incidence of infec- mostly human immunodeficiency virus- studies, continuous infusion of ampho-
tions due to non-C. albicans species with positive patients with oropharyngeal and tericin B deoxycholate over 24 hrs has
reduced dose-dependent susceptibility esophageal candidiasis, caspofungin was been shown to limit infusion-related re-
(Candida glabrata) or intrinsic resistance observed to be as effective as and better actions and nephrotoxicity and did not
(Candida krusei) to azoles was observed tolerated than amphotericin B deoxy- seem to affect efficacy (188, 189). Yet, the
in some U.S. centers (18, 180). The use of cholate (185, 186). In a large multicenter influence of this mode of administration
high doses (800 –1200 mg) of fluconazole trial that included 239 patients (of whom of amphotericin B on treatment efficacy
has been advocated for the empirical 24 were neutropenic) with invasive can- remains to be demonstrated, as it results
treatment of candidemia when infections didiasis (80% of the patients had candi- in a marked alteration of drug pharma-
with azole-resistant Candida species are demia), caspofungin (70-mg loading cokinetics. Lipid formulations have been
suspected, but efficacy data from compar- dose, followed by 50 mg daily) was at least developed to improve the toxicity and
ative trials are lacking. However, for most as efficacious as and less toxic than am- possibly also the efficacy profile of am-
experts, amphotericin B remained clearly photericin B deoxycholate (0.6 –1.0 mg/ photericin B. Three lipid formulations
the first choice for empirical therapy of kg/day) (success rates, 73% vs. 62%; dis- with different pharmacologic properties
invasive candidiasis in the preechinocan- continuation for adverse events, 3% vs. are available: liposomal amphotericin B,
din area (155, 167). This may have 23%) (187). Of note, the success rates of amphotericin B lipid complex, and am-
changed since the advent of the echino- caspofungin against C. glabrata and C. photericin B colloidal dispersion. They
candins. krusei were comparable with those ob- have been primarily used for the treat-
Itraconazole, an azole with improved tained in azole-susceptible Candida spe- ment of cancer patients with neutropenia
in vitro activity against fluconazole- cies. Micafungin and anidulafungin are and persistent fever or with invasive as-
resistant Candida species, has been avail- two other echinocandins that are cur- pergillosis. In large, multicenter clinical
able since the early 1990s. However, a rently in clinical development. trials, the lipid formulations were shown
poor bioavailability of the oral formula- In summary, azoles (fluconazole) and to be as efficacious as and usually better
tion of itraconazole has been a major echinocandins (caspofungin) have been tolerated than amphotericin B deoxy-
limitation for its use in critically ill pa- shown to be as efficacious as and better cholate (190 –197). Yet, there were differ-
tients. Recently, an intravenous form of tolerated than amphotericin B deoxy- ences between the lipid preparations in
itraconazole has been developed, but cholate for the treatment of patients with terms of adverse events. Infusion-related
there are very few data on the efficacy of candidemia and invasive candidiasis. reactions were significantly less frequent
IV itraconazole for the treatment of can- Limited information is available on the with liposomal amphotericin B (5% to
didemia. efficacy of these agents in neutropenic 20%) than with amphotericin B lipid
Voriconazole, a member of the newest patients. Amphotericin B and caspofun- complex (40% to 80%) or colloidal dis-
generation of antifungal triazoles, has gin are the antifungal agents of choice for persion of amphotericin B (50% to 80%).
been shown to exhibit excellent in vitro the treatment of infections caused by flu- Therefore, liposomal amphotericin B has
and in vivo activities against Candida spe- conazole-resistant non-C. albicans spe- been used as the control treatment regi-
cies (181). In patients with oropharyngeal cies. The place of the newest triazole vori- men in most recent studies of empirical
and/or esophageal candidiasis, voricon- conazole with improved in vitro activity antifungal therapy for persistent fever in

S506 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


neutropenic cancer patients (193, 198). Recommendation: Third- or fourth- Recommendation: Grade C
Lipid formulations are considerably more generation cephalosporins: Grade A. Car-
10. Empirical antifungal therapy should
expensive than conventional amphoteri- bapenem: Grade B. Extended-spectrum
not be used on a routine basis in
cin B, which has had a negative impact carboxypenicillins or ureidopenicillins
patients with severe sepsis or septic
on their use. combined with ␤-lactamase inhibitors:
shock, but it may be justified in se-
There are few data on the direct com- Grade C
lected subsets of septic patients at
parison of the efficacy and toxicity of am-
5. Empirical use of glycopeptide antibi- high risk for invasive candidiasis
photericin B deoxycholate and the lipid (155).
otics (vancomycin, teicoplanin), ox-
formulations for the treatment of pa-
azolidinones (linezolid), or strepto- Recommendation: Grade E
tients with invasive candidiasis. Small
gramins (quinupristin/dalfopristin) in
noncomparative studies suggest that lipid
patients with severe sepsis or septic 11. Azoles (fluconazole) and echinocan-
formulations of amphotericin B are as
shock is justified in patients with hy- dins (caspofungin) are as efficacious
efficacious as but better tolerated than
persensitivity to ␤-lactams or in insti- as and less toxic than amphotericin B
conventional amphotericin B (199 –202). deoxycholate for the treatment of pa-
tutions with resistant Gram-positive
Their high costs, the paucity of clinical tients with candidemia. Albeit better
bacteria (i.e., methicillin-resistant
data, and the existence of alternative an- tolerated, there is no evidence that
tifungal therapies (azoles and echinocan- staphylococci, penicillin-resistant
pneumococci, or ampicillin-resistant lipid formulations of amphotericin B
dins) explain why the use of lipid formu- are superior to amphotericin B de-
lations has been limited in patients with enterococci) in the community or in
the hospital. oxycholate for the treatment of can-
invasive candidiasis (155). didemia.
Recommendation: Grade E
Recommendation: Azoles: Grade A. Echi-
Summary Recommendations 6. Modification of empirical antimicro- nocandins: Grade B. Lipid formulations
bial therapy with the aim to restrict of amphotericin B: Grade E.
1. Antibiotic therapy should be started the number of antibiotics and narrow
within the first hour of recognition of the spectrum of antimicrobial therapy
is an important and responsible strat-
REFERENCES
severe sepsis, after appropriate cul-
tures have been obtained. egy for minimizing the development 1. Bochud PY, Glauser M, Calandra T: Antibi-
of resistant pathogens and for contain- otics in sepsis. Intensive Care Med 2001;
Recommendation: Grade E ing costs. 27(14 Suppl):S33–S48
2. Initial empirical antiinfective therapy 2. Zeni F, Freeman B, Natanson C: Anti-
Recommendation: Grade E inflammatory therapies to treat sepsis and
should include one or more drugs that
septic shock: A reassessment. Crit Care Med
have activity against the likely patho- 7. The antimicrobial regimen should al-
1997; 25:1095–1100
gens (bacterial or fungal) and that ways be reassessed after 48 –72 hrs on 3. Brun-Buisson C, Doyon F, Carlet J, et al:
penetrate into the presumed source of the basis of microbiological and clini- Incidence, risk factors, and outcome of se-
sepsis. The choice of drugs should be cal data with the aim to use a narrow- vere sepsis and septic shock in adults: A
guided by the susceptibility patterns of spectrum antibiotic to prevent the de- multicenter prospective study in intensive
microorganisms in the community velopment of resistance, to reduce care units. JAMA 1995; 274:968 –974
and in the hospital. toxicity, and to reduce costs. Once a 4. Brun-Buisson C, Doyon F, Carlet J: Bacte-
causative pathogen is identified, there remia and severe sepsis in adults: A multi-
Recommendation: Grade D is no evidence that combination ther- center prospective survey in ICUs and wards
of 24 hospitals. Am J Respir Crit Care Med
3. Monotherapy is as efficacious as com- apy is more effective monotherapy. 1996; 154:617– 624
bination therapy with a ␤-lactam and The duration of therapy should typi- 5. Cohen J, Carlet J, for the INTERSEPT Study
an aminoglycoside as empirical ther- cally be 7–10 days and guided by clin- Group: An international, multicenter, pla-
apy of patients with severe sepsis or ical response. cebo-controlled trial of monoclonal anti-
septic shock. body to human tumor necrosis factor-␣ in
Recommendation: Grade E patients with sepsis. Crit Care Med 1996;
Recommendation: Carbapenem: Grade B. 24:1431–1440
8. If the presenting clinical syndrome is
Third- or fourth-generation cephalospo- 6. Sands KE, Bates DW, Lanken PN, et al:
determined to be due to a noninfec-
rins: Grade B. Extended-spectrum car- Epidemiology of sepsis syndrome in 8 aca-
tious cause, antimicrobial therapy demic medical centers. JAMA 1997; 278:
boxypenicillins or ureidopenicillins com-
should be stopped promptly to mini- 234 –240
bined with ␤-lactamase inhibitors: Grade
mize the development of resistant 7. Bone RC, Sibbald WJ, Sprung CL: The
E.
pathogens and superinfection with ACCP-SCCM consensus conference on sep-
4. Third-generation and fourth-genera- other pathogenic organisms. sis and organ failure. Chest 1992; 101:
tion cephalosporins, carbapenems, 1481–1483
Recommendation: Grade E 8. Sackett DL, Haynes RB, Guyatt GH, et al:
and extended-spectrum carboxypeni-
Clinical Epidemiology. Philadelphia, Lip-
cillins or ureidopenicillins combined 9. At the present time, there is insuffi-
pincott-Raven, 1991
with ␤-lactamase inhibitors are cient evidence to recommend the use 9. Marchetti O, Calandra T: Infections in neu-
equally effective as empirical antibi- of antibiotic cycling as a strategy to tropenic cancer patients. Lancet 2002; 359:
otic therapy in patients with severe reduce the development of antibiotic 723–725
sepsis. resistance. 10. Martin GS, Mannino DM, Eaton S, et al: The

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S507


epidemiology of sepsis in the United States bacteremia. Arch Intern Med 1962; 110: tions: A risk factor for hospital mortality
from 1979 through 2000. N Engl J Med 92–100 among critically ill patients. Chest 1999;
2003; 348:1546 –1554 24. Freid MA, Vosti KL: The importance of un- 115:462– 474
11. Abraham E, Wunderink R, Silverman H, et derlying disease in patients with gram- 38. Ibrahim EH, Sherman G, Ward S, et al: The
al: Efficacy and safety of monoclonal anti- negative bacteremia. Arch Intern Med 1968; influence of inadequate antimicrobial treat-
body to human tumor necrosis factor ␣ in 121:418 – 423 ment of bloodstream infections on patient
patients with sepsis syndrome: A random- 25. Bryant RE, Hood AF, Hood CE, et al: Fac- outcomes in the ICU setting. Chest 2000;
ized, controlled, double-blind, multicenter tors affecting mortality of gram-negative 118:146 –155
clinical trial. JAMA 1995; 273:934 –941 rod bacteremia. Arch Intern Med 1971; 127: 39. Harbarth S, Ferriere K, Hugonnet S, et al:
12. Abraham E, Glauser MP, Butler T, et al: p55 120 –128 Epidemiology and prognostic determinants
tumor necrosis factor receptor fusion pro- 26. Young LS, Martin WJ, Meyer RD, et al: of bloodstream infections in surgical inten-
tein in the treatment of patients with severe Gram-negative rod bacteremia: Microbio- sive care. Arch Surg 2002; 137:1353–1359
sepsis and septic shock: A randomized con- logic, immunologic, and therapeutic con- 40. Hanon FX, Monnet DL, Sorensen TL, et al:
trolled multicenter trial. JAMA 1997; 277: siderations. Ann Intern Med 1977; 86: Survival of patients with bacteraemia in re-
1531–1538 456 – 471 lation to initial empirical antimicrobial
13. Abraham E, Anzueto A, Gutierrez G, et al: 27. Kreger BE, Craven DE, McCabe WR: Gram- treatment. Scand J Infect Dis 2002; 34:
Double-blind randomised controlled trial of negative bacteremia. IV. Re-evaluation of 520 –528
monoclonal antibody to human tumour ne- clinical features and treatment in 612 pa- 41. Harbarth S, Garbino J, Pugin J, et al: Inap-
crosis factor in treatment of septic shock. tients. Am J Med 1980; 68:344 –355 propriate initial antimicrobial therapy and
NORASEPT II Study Group. Lancet 1998; 28. Leibovici L, Paul M, Poznanski O, et al: its effect on survival in a clinical trial of
351(9107):929 –933 Monotherapy versus beta-lactam-aminogly- immunomodulating therapy for severe sep-
14. Cohen J, Abraham E: Microbiologic findings coside combination treatment for gram- sis. Am J Med 2003; 115:529 –535
and correlations with serum tumor necrosis negative bacteremia: A prospective, obser- 42. Zaragoza R, Artero A, Camarena JJ, et al:
factor-alpha in patients with severe sepsis vational study. Antimicrob Agents The influence of inadequate empirical anti-
and septic shock. J Infect Dis 1999; 180: Chemother 1997; 41:1127–1133 microbial treatment on patients with blood-
116 –121 29. Chow JW, Fine MJ, Shlaes DM, et al: Enter- stream infections in an intensive care unit.
15. Fisher CJ, Agosti JM, Opal SM, et al: Treat- obacter bacteremia: Clinical features and Clin Microbiol Infect 2003; 9:412– 418
ment of septic shock with the tumor necro- emergence of antibiotic resistance during 43. Leone M, Bourgoin A, Cambon S, et al:
therapy. Ann Intern Med 1991; 115:585–590 Empirical antimicrobial therapy of septic
sis factor receptor:Fc fusion protein. N Engl
30. Vidal F, Mensa J, Almela M, et al: Epidemi- shock patients: Adequacy and impact on the
J Med 1996; 334:1697–1702
ology and outcome of Pseudomonas aerugi- outcome. Crit Care Med 2003; 31:462– 467
16. Fisher CJ Jr, Opal SM, Dhainaut JF, et al:
nosa bacteremia, with special emphasis on 44. Garnacho-Montero J, Garcia-Garmendia JL,
Influence of an anti-tumor necrosis factor
the influence of antibiotic treatment: Anal- Barrero-Almodovar A, et al: Impact of ade-
monoclonal antibody on cytokine levels in
ysis of 189 episodes. Arch Intern Med 1996; quate empirical antibiotic therapy on the
patients with sepsis. Crit Care Med 1993;
156:2121–2126 outcome of patients admitted to the inten-
21:318 –327
31. Schiappa DA, Hayden MK, Matushek MG, et sive care unit with sepsis. Crit Care Med
17. Opal SM, Fisher CJ, Dhainaut JF, et al:
al: Ceftazidime-resistant Klebsiella pneu- 2003; 31:2742–2751
Confirmatory interleukin-1 receptor antag-
moniae and Escherichia coli bloodstream 45. Mandell LA, Campbell GD Jr: Nosocomial
onist trial in severe sepsis: A phase III, ran-
infection: A case-control and molecular ep- pneumonia guidelines: An international
domized, double-blind, placebo-controlled,
idemiologic investigation. J Infect Dis 1996; perspective. Chest 1998; 113(3 Suppl):
multicenter trial. Crit Care Med 1997; 25:
174:529 –536 188S–193S.
1115–1124
32. Caballero-Granado FJ, Cisneros JM, Luque 46. Mandell LA, Marrie TJ, Grossman RF, et al:
18. Edmond MB, Wallace SE, McClish DK, et al: R, et al: Comparative study of bacteremias Canadian guidelines for the initial manage-
Nosocomial bloodstream infections in caused by Enterococcus spp. with and with- ment of community-acquired pneumonia:
United States hospitals: A three-year analy- out high-level resistance to gentamicin. An evidence-based update by the Canadian
sis. Clin Infect Dis 1999; 29:239 –244 J Clin Microbiol 1998; 36:520 –525 Infectious Diseases Society and the Cana-
19. Pfaller MA, Jones RN, Messer SA, et al: Na- 33. Ispahani P, Pearson NJ, Greenwood D: An dian Thoracic Society. Clin Infect Dis 2000;
tional surveillance of nosocomial blood analysis of community and hospital-ac- 31:383– 421
stream infection due to species of Candida quired bacteraemia in a large teaching hos- 47. BTS guidelines for the management of
other than Candida albicans: Frequency of pital in the United Kingdom. Q J Med 1987; community acquired pneumonia in adults.
occurrence and antifungal susceptibility in 63:427– 440 Thorax 2001; 56(Suppl 4):IV1–IV64
the SCOPE Program. Surveillance and Con- 34. Leibovici L, Drucker M, Konigsberger H, et 48. Niederman MS, Mandell LA, Anzueto A, et
trol of Pathogens of Epidemiologic. Diagn al: Septic shock in bacteremic patients: Risk al: Guidelines for the management of adults
Microbiol Infect Dis 1998; 30:121–129 factors, features and prognosis. Scand J In- with community-acquired pneumonia: Di-
20. Voss A, Kluytmans JA, Koeleman JG, et al: fect Dis 1997; 29:71–75 agnosis, assessment of severity, antimicro-
Occurrence of yeast bloodstream infections 35. Weinstein MP, Towns ML, Quartey SM, et bial therapy, and prevention. Am J Respir
between 1987 and 1995 in five Dutch uni- al: The clinical significance of positive blood Crit Care Med 2001; 163:1730 –1754
versity hospitals. Eur J Clin Microbiol Infect cultures in the 1990s: A prospective com- 49. Memish ZA, Shibl AM, Ahmed QA: Guide-
Dis 1996; 15:909 –912 prehensive evaluation of the microbiology, lines for the management of community-
21. Sandven P, Bevanger L, Digranes A, et al: epidemiology, and outcome of bacteremia acquired pneumonia in Saudi Arabia: A
Constant low rate of fungemia in Norway, and fungemia in adults. Clin Infect Dis model for the Middle East region. Int J
1991 to 1996. J Clin Microbiol 1998; 36: 1997; 24:584 – 602 Antimicrob Agents 2002; 20(Suppl 1):
3455–3459 36. Leibovici L, Shraga I, Drucker M, et al: The S1–S12
22. Marchetti O, Bille J, Fluckiger U, et al: Ep- benefit of appropriate empirical antibiotic 50. Mandell LA, Bartlett JG, Dowell SF, et al:
idemiology of candidemia in Swiss tertiary treatment in patients with bloodstream in- Update of practice guidelines for the man-
care hospitals: Secular trends, 1991–2000. fection. J Intern Med 1998; 244:379 –386 agement of community-acquired pneumo-
Clin Infect Dis 2004; 38:311–320 37. Kollef MH, Sherman G, Ward S, et al: Inad- nia in immunocompetent adults. Clin In-
23. Mc Cabe WR, Jackson GG: Gram negative equate antimicrobial treatment of infec- fect Dis 2003; 37:1405–1433

S508 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


51. Mazuski JE, Sawyer RG, Nathens AB, et al: mixed flora infections. Am J Med 1985; 78: 76. Mangi RJ, Greco T, Ryan J, et al: Cefopera-
The surgical infection society guidelines on 85–91 zone versus combination antibiotic therapy
antimicrobial therapy for intra-abdominal 64. Mouton Y, Deboscker Y, Bazin C, et al: of hospital-acquired pneumonia. Am J Med
infections: An executive summary. Surg In- Prospective, randomized, controlled study 1988; 84:68 –74
fect 2002; 3:161–173 of imipenem-cilastatin versus cefotaxime- 77. Fernandez-Guerrero M, Gudiol F, Rodri-
52. Mazuski JE: The surgical infection society amikacin in the treatment of lower respira- guez-Torres A, et al: Nosocomial pneumo-
guidelines on antimicrobial therapy for in- tory tract infection and septicemia at inten- nia: Comparative multicentre trial between
tra-abdominal infections: Evidence for the sive care units. Presse Medicale 1990; 19: monotherapy with cefotaxime and treat-
recommendations. Surg Infect 2002; 607– 612 ment with antibiotic combinations. Infec-
3:175–233 65. Cometta A, Baumgartner JD, Lew D, et al: tion 1991; 19(Suppl 6):S320 –S325
53. Solomkin JS, Mazuski JE, Baron EJ, et al: Prospective randomized comparison of imi- 78. Croce MA, Fabian TC, Stewart RM, et al:
Guidelines for the selection of anti-infective penem monotherapy with imipenem plus Empiric monotherapy versus combination
agents for complicated intra-abdominal in- netilmicin for treatment of severe infec- therapy of nosocomial pneumonia in
fections. Clin Infect Dis 2003; 37:997–1005 tions in nonneutropenic patients. Antimi- trauma patients. J Trauma 1993; 35:
54. Klastersky J, Meunier-Carpentier F, Prevost crob Agents Chemother 1994; 38: 303–309
JM: Significance of antimicrobial synergism 1309 –1313 79. Speich R, Imhof E, Vogt M, et al: Efficacy,
for the outcome of gram negative sepsis. 66. Jaspers CA, Kieft H, Speelberg B, et al: safety, and tolerance of piperacillin/tazobac-
Am J Med Sci 1977; 273:157–167 Meropenem versus cefuroxime plus genta- tam compared to co-amoxiclav plus an ami-
55. Hanson B, Coppens L, Klastersky J: Com- micin for treatment of serious infections in noglycoside in the treatment of severe
parative studies of ticarcillin and mezlocil- elderly patients. Antimicrob Agents Che- pneumonia. Eur J Clin Microbiol Infect Dis
lin plus sisomicin in Gram-negative bacil- mother 1998; 42:1233–1238 1998; 17:313–317
lary bacteraemia and bronchopneumonia. 67. Arich C, Gouby A, Bengler C, et al: Com- 80. Manhold C, von Rolbicki U, Brase R, et al:
J Antimicrob Chemother 1982; 10:335–341 parison of the efficacy of cefotaxime alone Outbreaks of Staphylococcus aureus infec-
56. Krumpe PE, Cohn S, Garreltes J, et al: In- and the combination cefazolin-tobramycin tions during treatment of late onset pneu-
travenous and oral mono- or combination- in the treatment of enterobacterial septice- monia with ciprofloxacin in a prospective,
therapy in the treatment of severe infec- mia. Pathol Biol 1987; 35:613– 615 randomized study. Intensive Care Med
tions: Ciprofloxacin versus standard 68. Oblinger MJ, Bowers JT, Sande MA, et al: 1998; 24:1327–1330
antibiotic therapy. J Antimicrob Chemother Moxalactam therapy vs. standard antimicro- 81. Poenaru D, De Santis M, Christou NV: Imi-
1999; 43(Suppl A):117–128 bial therapy for selected serious infections. penem versus tobramycin–antianaerobe an-
57. Beaucaire G: Evaluation of the efficacy and Rev Infect Dis 1982; 4(Suppl):S639 –S649 tibiotic therapy in intra-abdominal infec-
safety of isepamicin compared with amika- 69. Rubinstein E, Lode H, Grassi C: Ceftazidime tions. Can J Surg 1990; 33:415– 422
cin in the treatment of nosocomial pneu- monotherapy vs. ceftriaxone/tobramycin for 82. Solomkin JS, Dellinger EP, Christou NV, et
monia and septicaemia. J Chemother 1995; serious hospital-acquired gram-negative in- al: Results of a multicenter trial comparing
7(Suppl 2):165–173 fections. Clin Infect Dis 1995; 20: imipenem/cilastatin to tobramycin/clinda-
58. Colardyn F: The efficacy and safety of ise- 1217–1228 mycin for intra-abdominal infections. Ann
pamicin and ceftazidime compared with 70. Extermann M, Regamey C, Humair L, et al: Surg 1990; 212:581–591
amikacin and ceftazidime in acute lower Initial treatment of sepsis in non-neutro- 83. Schentag JJ, Wels PB, Reitberg DP, et al: A
respiratory tract infection. J Chemother penic patients: Ceftazidime alone versus randomized clinical trial of moxalactam
1995; 7(Suppl 2):129 –135 ‘best guess’ combined antibiotic therapy. alone versus tobramycin plus clindamycin
59. Wolff M: Comparison of strategies using Chemotherapy 1995; 41:306 –315 in abdominal sepsis. Ann Surg 1983; 198:
cefpirome and ceftazidime for empiric 71. McCormick PA, Greenslade L, Kibbler CC, 35– 41
treatment of pneumonia in intensive care et al: A prospective randomized trial of 84. Greenberg RN, Cayavec P, Danko LS, et al:
patients. Antimicrob Agents Chemother ceftazidime versus netilmicin plus me- Comparison of cefoperazone plus sulbactam
1998; 42:28 –36 zlocillin in the empirical therapy of pre- with clindamycin plus gentamicin as treat-
60. Brun-Buisson C, Sollet JP, Schweich H, et sumed sepsis in cirrhotic patients. Hepatol- ment for intra-abdominal infections. J An-
al: Treatment of ventilator-associated pneu- ogy 1997; 25:833– 836 timicrob Chemother 1994; 34:391– 401
monia with piperacillin-tazobactam/amika- 72. Hammerberg O, Kurnitzki C, Watts J, et al: 85. Fink MP: Antibiotic therapy of intra-
cin versus ceftazidime/amikacin: A multi- Randomized trial using piperacillin versus abdominal sepsis in the elderly: Experience
center, randomized controlled trial. Clin ampicillin and amikacin for treatment of with ticarcillin and clavulanic acid. Surg
Infect Dis 1998; 26:346 –354 premature neonates with risk factors for Gynecol Obstet 1991; 172(Suppl):36 – 41
61. Alvarez-Lerma F, Insausti-Ordenana J, sepsis. Eur J Clin Microbiol Infect Dis 1989; 86. Dupont H, Carbon C, Carlet J: Monotherapy
Jorda-Marcos R, et al: Efficacy and tolera- 8:241–244 with a broad-spectrum beta-lactam is as ef-
bility of piperacillin/tazobactam versus 73. Korvick JA, Bryan CS, Farber B, et al: Pro- fective as its combination with an amino-
ceftazidime in association with amikacin for spective observational study of Klebsiella glycoside in treatment of severe generalized
treating nosocomial pneumonia in inten- bacteremia in 230 patients: Outcome for peritonitis: a multicenter randomized con-
sive care patients: A prospective random- antibiotic combinations versus mono- trolled trial. Antimicrob Agents Chemother
ized multicenter trial. Intensive Care Med therapy. Antimicrob Agents Chemother 2000; 44:2028 –2033
2001; 27:493–502 1992; 36:2639 –2644 87. Stone HH, Kolb LD, Geheber CE, et al:
62. Beaucaire G, Nicolas MH, Martin C, et al: 74. Sieger B, Berman SJ, Geckler RW, et al: Treatment of surgical infections with to-
Phare study: Comparative study of com- Empiric treatment of hospital-acquired bramcin. Am Surg 1975; 41:301–308
bined cefepime-amikacin versus ceftazi- lower respiratory tract infections with 88. Smith CR, Baughman KL, Edwards CQ, et
dime combined with amikacin in the treat- meropenem or ceftazidime with tobramy- al: Controlled comparison of amikacin and
ment of nosocomial pneumonias in cin: A randomized study. Crit Care Med gentamicin. N Engl J Med 1977; 296:
ventilated patients. Ann Fr Anesth Reanim 1997; 25:1663–1670 349 –353
1999; 18:186 –195 75. Alvarez-Lerma F: Efficacy of meropenem as 89. Bock BV, Edelstein PH, Meyer RD: Prospec-
63. Solomkin JS, Fant WK, Rivera JO, et al: monotherapy in the treatment of ventilator- tive comparative study of efficacy and tox-
Randomized trial of imipenem/cilastatin associated pneumonia. J Chemother 2001; icity of netilmicin and amikacin. Antimi-
versus gentamicin and clindamycin in 13:70 – 81 crob Agents Chemother 1980; 17:217–225

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S509


90. Gudiol F, Pallares R, Ariza X, et al: Com- cefotaxime in the treatment of lower respi- Effect of quinupristin/dalfopristin on the
parative clinical evaluation of aztreonam ratory tract infections. J Antimicrob Che- outcome of vancomycin-resistant Entero-
versus aminoglycosides in gram-negative mother 1993; 32(Suppl B):187–193 coccus faecium bacteraemia: Comparison
septicaemia. J Antimicrob Chemother 1986; 105. Fink MP, Snydman DR, Niederman MS, et with a control cohort. J Antimicrob Che-
17:661– 671 al: Treatment of severe pneumonia in hos- mother 1997; 39(Suppl A):145–151
91. Mangi RJ, Ryan J, Berenson C, et al: Ce- pitalized patients: Results of a multicenter, 117. Polk HC Jr, Livingston DH, Fry DE, et al:
foperazone versus ceftazidime monotherapy randomized, double-blind trial comparing Treatment of pneumonia in mechanically
of nosocomial pneumonia. Am J Med 1988; intravenous ciprofloxacin with imipenem- ventilated trauma patients: Results of a pro-
85:44 – 48 cilastatin. Antimicrob Agents Chemother spective trial. Arch Surg 1997; 132:
92. Norrby SR, Finch RG, Glauser M: Mono- 1994; 38:547–557 1086 –1092
therapy in serious hospital-acquired infec- 106. Siami GA, Wilkins WT, Bess DT, et al: Com- 118. Fagon J, Patrick H, Haas DW, et al: Treat-
tions: A clinical trial of ceftazidime versus parison of ciprofloxacin with imipenem in ment of gram-positive nosocomial pneumo-
imipenem/cilastatin. J Antimicrob Che- the treatment of severe pneumonia in hos- nia: Prospective randomized comparison of
mother 1993; 31:927–937 pitalised geriatric patients. Drugs 1995; quinupristin/dalfopristin versus vancomy-
93. Norrby SR, Geddes AM: Efficacy of cefpi- 49(Suppl 2):436 – 438 cin. Am J Respir Crit Care Med 2000; 161:
rome in the treatment of septicaemia. 107. Jaccard C, Troillet N, Harbarth S, et al: 753–762
Scand J Infect Dis 1993; 91:51–59 Prospective randomized comparison of imi- 119. Rubinstein E, Cammarata S, Oliphant T, et
94. Schrank JH Jr, Kelly JW, McAllister CK: penem-cilastatin and piperacillin-tazobac- al: Linezolid (PNU-100766) versus vanco-
Randomized comparison of cefepime and tam in nosocomial pneumonia or peritoni- mycin in the treatment of hospitalized pa-
ceftazidime for treatment of hospitalized tis. Antimicrob Agents Chemother 1998; 42: tients with nosocomial pneumonia: A ran-
patients with gram-negative bacteremia. 2966 –2972 domized, double-blind, multicenter study.
Clin Infect Dis 1995; 20:56 –58 108. Grossman RF, Campbell DA, Landis SJ, et Clin Infect Dis 2001; 32:402– 412
95. Colardyn F, Faulkner KL: Intravenous al: Treatment of community-acquired 120. Calandra T, Glauser MP: Immunocompro-
meropenem versus imipenem/cilastatin in pneumonia in the elderly: The role of mised animal models for the study of anti-
the treatment of serious bacterial infections cefepime, a fourth-generation cephalospo- biotic combinations. Am J Med 1986; 80:
in hospitalized patients. J Antimicrob Che- rin. J Antimicrob Chemother 1999; 43: 45–52
mother 1996; 38:523–537 549 –554 121. Klastersky J, Zinner SH: Synergistic com-
96. Mehtar S, Dewar EP, Leaper DJ, et al: A 109. Torres A, Bauer TT, Leon-Gil C, et al: Treat- binations of antibiotics in gram-negative
multi-centre study to compare meropenem ment of severe nosocomial pneumonia: A bacillary infections. Rev Infect Dis 1982;
and cefotaxime and metronidazole in the prospective randomised comparison of in- 4:294 –301
treatment of hospitalized patients with se- travenous ciprofloxacin with imipenem/ 122. De Jongh CA, Joshi JH, Thompson BW, et
rious infections. J Antimicrob Chemother cilastatin. Thorax 2000; 55:1033–1039 al: A double beta-lactam combination ver-
1997; 39:631– 638 110. West M, Boulanger BR, Fogarty C, et al: sus an aminoglycoside-containing regimen
97. Garau J, Blanquer J, Cobo L, et al: Prospec- Levofloxacin compared with imipenem/ as empiric antibiotic therapy for febrile
tive, randomised, multicentre study of cilastatin followed by ciprofloxacin in adult granulocytopenic cancer patients. Am J
meropenem versus imipenem/cilastatin as patients with nosocomial pneumonia: A Med 1986; 80:101–111
empiric monotherapy in severe nosocomial multicenter, prospective, randomized, 123. Gribble MJ, Chow AW, Naiman SC, et al:
infections. Eur J Clin Microbiol Infect Dis open-label study. Clin Ther 2003; 25: Prospective randomized trial of piperacillin
1997; 16:789 –796 485–506 monotherapy versus carboxypenicillin-
98. Norrby SR, Geddes AM, Shah PM: Random- 111. Kempf P, Bauernfeind A, Muller A, et al: aminoglycoside combination regimens in
ized comparative trial of cefpirome versus Meropenem monotherapy versus cefo- the empirical treatment of serious bacterial
ceftazidime in the empirical treatment of taxime plus metronidazole combination infections. Antimicrob Agents Chemother
suspected bacteraemia or sepsis. J Antimi- treatment for serious intra-abdominal in- 1983; 24:388 –393
crob Chemother 1998; 42:503–509 fections. Infection 1996; 24:473– 479 124. The EORTC International Antimicrobial
99. Verwaest C: Meropenem versus imipenem/ 112. Macfarlane JT, Finch RG, Ward MJ, et al: Therapy Cooperative Group: Ceftazidime
cilastatin as empirical monotherapy for se- Erythromycin compared with a combina- combined with a short or long course of
rious bacterial infections in the intensive tion of ampicillin and amoxycillin as initial amikacin for empirical therapy of gram-
care unit. Clin Microbiol Infect 2000; therapy for adults with pneumonia includ- negative bacteremia in cancer patients with
6:294 –302 ing Legionnaires’ disease. J Infect 1983; granulocytopenia. N Engl J Med 1987; 317:
100. Schentag JJ, Vari AJ, Winslade NE, et al: 7:111–117 1692–1698
Treatment with aztreonam or tobramycin 113. Shann F, Barker J, Poore P: Chloramphen- 125. Anderson ET, Young LS, Hewitt WL: Anti-
in critical care patients with nosocomial icol alone versus chloramphenicol plus pen- microbial synergism in the therapy of
gram-negative pneumonia. Am J Med 1985; icillin for severe pneumonia in children. gram-negative rod bacteremia. Chemother-
78:34 – 41 Lancet 1985; 2:684 – 686 apy 1978; 24:45–54
101. Reeves JH, Russell GM, Cade JF, et al: Com- 114. Gaillat J, Bru JP, Sedallian A: Penicillin 126. Nicolau DP, McNabb J, Lacy MK, et al: Con-
parison of ceftriaxone with cefotaxime in G/ofloxacin versus erythromycin/amoxicil- tinuous versus intermittent administration
serious chest infections. Chest 1989; 96: lin-clavulanate in the treatment of severe of ceftazidime in intensive care unit pa-
1292–1297 community-acquired pneumonia. Eur tients with nosocomial pneumonia. Int J
102. Mangi RJ, Peccerillo KM, Ryan J, et al: Ce- J Clin Microbiol Infect Dis 1994; 13: Antimicrob Agents 2001; 17:497–504
foperazone versus ceftriaxone monotherapy 639 – 644 127. Bartlett JG, Dowell SF, Mandell LA, et al:
of nosocomial pneumonia. Diagn Microbiol 115. Roson B, Carratala J, Tubau F, et al: Use- Practice guidelines for the management of
Infect Dis 1992; 15:441– 447 fulness of betalactam therapy for commu- community-acquired pneumonia in adults.
103. Thomas P, Daly S, Misan G, et al: Compar- nity-acquired pneumonia in the era of drug- Clin Infect Dis 2000; 31:347–382
ison of the efficacy and adverse effect profile resistant Streptococcus pneumoniae: A 128. Richards MJ, Edwards JR, Culver DH, et al:
of cefotaxime and ceftriaxone in ICU pa- randomized study of amoxicillin-clavu- Nosocomial infections in medical intensive
tients with susceptible infections. Diagn lanate and ceftriaxone. Microb Drug Resist care units in the United States. Crit Care
Microbiol Infect Dis 1992; 15:89 –97 2001; 7:85–96 Med 1999; 27:887– 892
104. Barckow D, Schwigon CD: Cefepime versus 116. Linden PK, Pasculle AW, McDevitt D, et al: 129. Vincent JL, Bihari DJ, Suter PM, et al: The

S510 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)


prevalence of nosocomial infection in inten- 143. Nichols RL, Graham DR, Barriere SL, et al: 157. Beck-Sagué CM, Jarvis WR, the National
sive care units in Europe: Results of the Treatment of hospitalized patients with Nosocomial Infections Surveillance System:
European Prevalence of Infection in Inten- complicated gram-positive skin and skin Secular trends in the epidemiology of nos-
sive Care (EPIC) Study. JAMA 1995; 274: structure infections: Two randomized, mul- ocomial fungal infections in the United
639 – 644 ticentre studies of quinupristin/dalfopristin States, 1980 –1990. J Infect Dis 1993; 167:
130. Yu VL, Chiou CC, Feldman C, et al: An versus cefazolin, oxacillin or vancomycin. 1247–1251
international prospective study of pneumo- J Antimicrob Chemother 1999; 44:263–273 158. Fisher-Hoch SP, Hutwagner L: Opportunis-
coccal bacteremia: Correlation with in vitro 144. Ibrahim EH, Ward S, Sherman G, et al: tic candidiasis: An epidemic of the 1980s.
resistance, antibiotics administered, and Experience with a clinical guideline for the Clin Infect Dis 1995; 21:897–904
clinical outcome. Clin Infect Dis 2003; 37: treatment of ventilator-associated pneumo- 159. Fraser VJ, Jones M, Dunkel J, et al: Candi-
230 –237 nia. Crit Care Med 2001; 29:1109 –1115 demia in a tertiary care hospital: Epidemi-
131. John CC: Treatment failure with use of a 145. Haley RW, Bregman DA: The role of under- ology, risk factors, and predictors of mor-
third-generation cephalosporin for penicil- staffing and overcrowding in recurrent out- tality. Clin Infect Dis 1992; 15:414 – 421
lin-resistant pneumococcal meningitis: breaks of staphylococcal infection in a neo- 160. Wenzel RP: Nosocomial candidemia: Risk
Case report and review. Clin Infect Dis natal special-care unit. J Infect Dis 1982; factors and attributable mortality. Clin In-
1994; 18:188 –193 145:875– 885 fect Dis 1995; 20:1531–1534
132. Cometta A, Kern WV, De Bock R, et al: 146. Fridkin SK, Pear SM, Williamson TH, et al: 161. Calandra T, Bille J, Schneider R, et al: Clin-
Vancomycin versus placebo for treating per- The role of understaffing in central venous ical significance of Candida isolated from
sistent fever in patients with neutropenic catheter-associated bloodstream infections. peritoneum in surgical patients. Lancet
cancer receiving piperacillin-tazobactam Infect Control Hosp Epidemiol 1996; 17: 1989; 2:1437–1440
monotherapy. Clin Infect Dis 2003; 37: 150 –158 162. Wey SB, Mori M, Pfaller MA, et al: Hospital-
382–389 147. Grundmann H, Hori S, Winter B, et al: Risk acquired candidemia: The attributable mor-
133. Smith TL, Pearson ML, Wilcox KR, et al: factors for the transmission of methicillin- tality and excess length of stay. Arch Intern
Emergence of vancomycin resistance in resistant Staphylococcus aureus in an adult Med 1988; 148:2642–2645
Staphylococcus aureus. N Engl J Med 1999; intensive care unit: Fitting a model to the 163. Gudlaugsson O, Gillespie S, Lee K, et al:
340:493–501 data. J Infect Dis 2002; 185:481– 488 Attributable mortality of nosocomial candi-
134. Weigel LM, Clewell DB, Gill SR, et al: Ge- 148. Gerding DN, Larson TA, Hughes RA, et al: demia, revisited. Clin Infect Dis 2003; 37:
netic analysis of a high-level vancomycin- Aminoglycoside resistance and aminoglyco- 1172–1177
resistant isolate of Staphylococcus aureus. side usage: Ten years of experience in one 164. Uzun O, Ascioglu S, Anaissie EJ, et al: Risk
Science 2003; 302:1569 –1571 hospital. Antimicrob Agents Chemother factors and predictors of outcome in pa-
135. Moellering RC: Linezolid: The first oxazo- 1991; 35:1284 –1290 tients with cancer and breakthrough candi-
lidinone antimicrobial. Ann Intern Med 149. Kollef MH, Vlasnik J, Sharpless L, et al: demia. Clin Infect Dis 2001; 32:1713–1717
2003; 138:135–142 Scheduled change of antibiotic classes: A 165. Nguyen MH, Peacock JE Jr, Tanner DC, et
136. San Pedro GS, Cammarata SK, Oliphant strategy to decrease the incidence of venti- al: Therapeutic approaches in patients with
TH, et al: Linezolid versus ceftriaxone/ lator-associated pneumonia. Am J Respir candidemia: Evaluation in a multicenter,
cefpodoxime in patients hospitalized for the Crit Care Med 1997; 156:1040 –1048 prospective, observational study. Arch In-
treatment of Streptococcus pneumoniae 150. Dominguez EA, Smith TL, Reed E, et al: A tern Med 1995; 155:2429 –2435
pneumonia. Scand J Infect Dis 2002; 34: pilot study of antibiotic cycling in a hema- 166. Nolla-Salas J, Sitges-Serra A, Leon-Gil C, et
720 –728 tology-oncology unit. Infect Control Hosp al: Candidemia in non-neutropenic criti-
137. Wunderink RG, Cammarata SK, Oliphant Epidemiol 2000; 21(1 Suppl):S4 –S8 cally ill patients: Analysis of prognostic fac-
TH, et al: Continuation of a randomized, 151. Gruson D, Hilbert G, Vargas F, et al: Rota- tors and assessment of systemic antifungal
double-blind, multicenter study of linezolid tion and restricted use of antibiotics in a therapy. Intensive Care Med 1997; 23:23–30
versus vancomycin in the treatment of pa- medical intensive care unit: Impact on the 167. Edwards JE Jr, Bodey GP, Bowden RA, et al:
tients with nosocomial pneumonia. Clin incidence of ventilator-associated pneumo- International Conference for the Develop-
Ther 2003; 25:980 –992 nia caused by antibiotic-resistant gram- ment of a Consensus on the Management
138. Wunderink RG, Rello J, Cammarata SK, et negative bacteria. Am J Respir Crit Care and Prevention of Severe Candidal Infec-
al: Linezolid vs vancomycin: Analysis of two Med 2000; 162:837– 843 tions. Clin Infect Dis 1997; 25:43–59
double-blind studies of patients with methi- 152. Raymond DP, Pelletier SJ, Crabtree TD, et 168. Rex JH, Bennett JE, Sugar AM, et al: A
cillin-resistant Staphylococcus aureus nos- al: Impact of a rotating empiric antibiotic randomized trial comparing fluconazole
ocomial pneumonia. Chest 2003; 124: schedule on infectious mortality in an in- with amphotericin B for the treatment of
1789 –1797 tensive care unit. Crit Care Med 2001; 29: candidemia in patients without neutrope-
139. Stevens DL, Herr D, Lampiris H, et al: Lin- 1101–1108 nia. N Engl J Med 1994; 331:1325–1330
ezolid versus vancomycin for the treatment 153. Fridkin SK: Routine cycling of antimicro- 169. Phillips P, Shafran S, Garber G, et al: Mul-
of methicillin-resistant Staphylococcus au- bial agents as an infection-control measure. ticenter randomized trial of fluconazole
reus infections. Clin Infect Dis 2002; 34: Clin Infect Dis 2003; 36:1438 –1444 versus amphotericin B for treatment of can-
1481–1490 154. Paterson DL, Rice LB: Empirical antibiotic didemia in non-neutropenic patients. Eur
140. Gonzales RD, Schreckenberger PC, Graham choice for the seriously ill patient: Are min- J Clin Microbiol Infect Dis 1997; 16:
MB, et al: Infections due to vancomycin- imization of selection of resistant organ- 337–345
resistant Enterococcus faecium resistant to isms and maximization of individual out- 170. Anaissie EJ, Vartivarian SE, Abi-Said D, et
linezolid. Lancet 2001; 357:1179 come mutually exclusive? Clin Infect Dis al: Fluconazole versus amphotericin B in
141. Tsiodras S, Gold HS, Sakoulas G, et al: 2003; 36:1006 –1012 the treatment of hematogenous candidiasis:
Linezolid resistance in a clinical isolate of 155. Rex JH, Walsh TJ, Sobel JD, et al: Practice A matched cohort study. Am J Med 1996;
Staphylococcus aureus. Lancet 2001; 358: guidelines for the treatment of candidiasis. 101:170 –176
207–208 Clin Infect Dis 2000; 30:662– 678 171. Driessen M, Ellis JB, Cooper PA, et al: Flu-
142. Herrero IA, Issa NC, Patel R: Nosocomial 156. Banerjee SN, Emori TG, Culver DH, et al: conazole vs. amphotericin B for the treat-
spread of linezolid-resistant, vancomycin- Secular trends in nosocomial primary ment of neonatal fungal septicemia: A pro-
resistant Enterococcus faecium. N Engl bloodstream infections in the United States, spective randomized trial. Ped Infect Dis J
J Med 2002; 346:867– 869 1980 –1989. Am J Med 1991; 91:86S– 89S 1996; 15:1107–1112

Crit Care Med 2004 Vol. 32, No. 11 (Suppl.) S511


172. van’t Wout JW, Mattie H, van Furth R: A emerging, or refractory fungal infections. 194. Wingard JR, White MH, Anaissie E, et al: A
prospective study of the efficacy of flucon- Clin Infect Dis 2003; 36:1122–1131 randomized, double-blind comparative trial
azole (UK-49,858) against deep-seated fun- 184. Denning DW: Echinocandin antifungal evaluating the safety of liposomal ampho-
gal infections. J Antimicrob Chemother drugs. Lancet 2003; 362:1142–1151 tericin B versus amphotericin B lipid com-
1988; 21:665– 672 185. Villanueva A, Arathoon EG, Gotuzzo E, et plex in the empirical treatment of febrile
173. Kujath P, Lerch K: Secondary mycosis in al: A randomized double-blind study of neutropenia. Clin Infect Dis 2000; 31:
surgery: treatment with fluconazole. Infec- caspofungin versus amphotericin for the 1155–1163
tion 1989; 17:111–117 treatment of candidal esophagitis. Clin In- 195. Ellis M, Spence D, de Pauw B, et al: An
174. Ikemoto H: A clinical study of fluconazole fect Dis 2001; 33:1529 –1535 EORTC international multicenter random-
for the treatment of deep mycoses. Diagn 186. Arathoon EG, Gotuzzo E, Noriega LM, et al: ized trial (EORTC number 19923) compar-
Microbiol Infect Dis 1989; 12(4 Suppl): Randomized, double-blind, multicenter ing two dosages of liposomal amphotericin
239S–247S study of caspofungin versus amphotericin B B for treatment of invasive aspergillosis.
175. Viscoli C, Castagnola E, Fioredda F, et al: for treatment of oropharyngeal and esoph- Clin Infect Dis 1998; 27:1406 –1412
Fluconazole in the treatment of candidiasis ageal candidiases. Antimicrob Agents Che- 196. Bowden R, Chandrasekar P, White MH, et
in immunocompromised children. Antimi- mother 2002; 46:451– 457 al: A double-blind, randomized, controlled
crob Agents Chemother 1991; 35:365–367 187. Mora-Duarte J, Betts R, Rotstein C, et al: trial of amphotericin B colloidal dispersion
176. Nolla-Salas J, Leon C, Torres-Rodriguez JM, Comparison of caspofungin and amphoter- versus amphotericin B for treatment of in-
et al: Treatment of candidemia in critically icin B for invasive candidiasis. N Engl J Med vasive aspergillosis in immunocompro-
ill surgical patients with intravenous flu- 2002; 347:2020 –2029 mised patients. Clin Infect Dis 2002; 35:
conazole. Clin Infect Dis 1992; 14:952–954 188. Eriksson U, Seifert B, Schaffner A: Compar- 359 –366
177. Fasano C, O’Keeffe J, Gibbs D: Fluconazole ison of effects of amphotericin B deoxy- 197. Leenders AC, Daenen S, Jansen RL, et al:
treatment of children with severe fungal cholate infused over 4 or 24 hrs: Random- Liposomal amphotericin B compared with
infections not treatable with conventional ised controlled trial. BMJ 2001; 322: amphotericin B deoxycholate in the treat-
agents. Eur J Clin Microbiol Infect Dis 579 –582 ment of documented and suspected neutro-
1994; 13:344 –347 189. Imhof A, Walter RB, Schaffner A: Continu- penia-associated invasive fungal infections.
178. Presterl E, Graninger W: Efficacy and safety ous infusion of escalated doses of ampho- Br J Haematol 1998; 103:205–212
of fluconazole in the treatment of systemic tericin B deoxycholate: An open-label obser- 198. Walsh TJ, Pappas P, Winston DJ, et al: Vori-
fungal infections in pediatric patients. Eur vational study. Clin Infect Dis 2003; 36: conazole compared with liposomal ampho-
J Clin Microbiol Infect Dis 1994; 13: 943–951 tericin B for empirical antifungal therapy in
347–351 190. Prentice HG, Hann IM, Herbrecht R, et al: A patients with neutropenia and persistent fe-
179. Graninger W, Presteril E, Schneeweiss B, et randomized comparison of liposomal versus ver. N Engl J Med 2002; 346:225–234
al: Treatment of Candida albicans fungae- conventional amphotericin B for the treat- 199. Bowden RA, Cays M, Gooley T, et al: Phase
mia with fluconazole. J Infect 1993; 26: ment of pyrexia of unknown origin in neu- I study of amphotericin B colloidal disper-
133–146 tropenic patients. Br J Haematol 1997; 98: sion for the treatment of invasive fungal
180. Nguyen MH, Peacock JE Jr, Morris AJ, et al: 711–718 infections after marrow transplant. J Infect
The changing face of candidemia: Emer- 191. Clark AD, McKendrick S, Tansey PJ, et al: A Dis 1996; 173:1208 –1215
gence of non-Candida albicans species and comparative analysis of lipid-complexed and 200. Noskin GA, Pietrelli L, Coffey G, et al: Am-
antifungal resistance. Am J Med 1996; 100: liposomal amphotericin B preparations in photericin B colloidal dispersion for treat-
617– 623 haematological oncology. Br J Haematol ment of candidemia in immunocompro-
181. Johnson LB, Kauffman CA: Voriconazole: A 1998; 103:198 –204 mised patients. Clin Infect Dis 1998; 26:
new triazole antifungal agent. Clin Infect 192. White MH, Bowden RA, Sandler ES, et al: 461– 467
Dis 2003; 36:630 – 637 Randomized, double-blind clinical trial of 201. Juster-Reicher A, Flidel-Rimon O, Amitay
182. Ally R, Schurmann D, Kreisel W, et al: A amphotericin B colloidal dispersion vs. am- M, et al: High-dose liposomal amphotericin
randomized, double-blind, double-dummy, photericin B in the empirical treatment of B in the therapy of systemic candidiasis in
multicenter trial of voriconazole and flu- fever and neutropenia. Clin Infect Dis 1998; neonates. Eur J Clin Microbiol Infect Dis
conazole in the treatment of esophageal 27:296 –302 2003; 22:603– 607
candidiasis in immunocompromised pa- 193. Walsh TJ, Finberg RW, Arndt C, et al: Lipo- 202. Lopez Sastre JB, Coto Cotallo GD, Fernan-
tients. Clin Infect Dis 2001; 33:1447–1454 somal amphotericin B for empirical therapy dez CB: Neonatal invasive candidiasis: A
183. Perfect JR, Marr KA, Walsh TJ, et al: Vori- in patients with persistent fever and neu- prospective multicenter study of 118 cases.
conazole treatment for less-common, tropenia. N Engl J Med 1999; 340:764 –771 Am J Perinatol 2003; 20:153–163

S512 Crit Care Med 2004 Vol. 32, No. 11 (Suppl.)

You might also like