You are on page 1of 6

INVITED ARTICLE REVIEWS OF ANTI-INFECTIVE AGENTS

Louis D. Saravolatz, Section Editor

Treatment of Acinetobacter Infections


Joel Fishbain1 and Anton Y. Peleg2,3,4
1
Division of Infectious Diseases, St John Hospital and Medical Center, Detroit, Michigan, and 2Division of Infectious Diseases, Massachusetts General Hospital
and Beth Israel Deaconess Medical Center, Boston, Massachusetts; 3Alfred Hospital and 4Department of Microbiology, Monash University, Melbourne, Australia

Acinetobacter baumannii remains an important and difficult-to-treat pathogen whose resistance patterns result in significant
challenges for the clinician. Despite the prevalence and interest in A. baumannii infections, there is relatively limited well-
controlled scientific data to help the clinician select optimal empirical and subsequent targeted therapy for a variety of
infections. We will review the currently available antimicrobial agents and discuss the clinical data supporting the use of the
various agents.

INTRODUCTION mechanisms is beyond the scope of this article but can be re-
viewed in Peleg et al [4]. Degradation enzymes against b-lac-
The challenges of treating multidrug-resistant bacteria continue
tams, modification enzymes against aminoglycosides, altered
to be at the forefront of the clinician’s practice in caring for
binding sites for quinolones, and a variety of efflux mecha-
hospitalized patients. Acinetobacter baumannii has proven to be
nisms and changes in outer membrane proteins have been re-
an increasingly important and demanding species in health care–
ported. Essentially, any and all of these elements can be com-
associated infections. The drug-resistant nature of the pathogen
and its unusual and unpredictable susceptibility patterns make bined to result in a highly drug-resistant, and at times pan-
empirical and therapeutic decisions even more difficult. resistant, opportunistic pathogen [4].
The association of A. baumannii with pneumonia, bactere- Attributable mortality has been difficult to assess for an or-
mia, wound infections, urinary tract infections, and meningitis ganism that appears to be as much a colonizer as it is a true
has been well described [1]. Risk factors associated with col- pathogen. Although one cannot argue against the pathogenic-
onization or infection (which can be difficult to distinguish) ity of an organism that is identified in bloodstream infections,
include prolonged hospitalization, intensive care unit admis- A. baumannii is often identified in the sputum samples of pa-
sion, recent surgical procedures, antimicrobial agent exposure, tients with mechanical ventilation. This quandary of patho-
central venous catheter use, prior hospitalization, nursing home genicity has added to the difficulty of treating these highly
residence, and local colonization pressure on susceptible pa- resistant organisms, because many therapeutic strategies are
tients [1–3]. associated with significant toxicity. The in-hospital attributable
The ability of A. baumannii to survive for extended periods mortality appears to be 8%–23%, but for the intensive care
on environmental surfaces is notorious and is likely important unit, it was found to be 10%–43% [5, 6]. Although most stud-
for transmission within the health care setting. Multidrug re- ies did not demonstrate a statistically significant difference be-
sistance is common with health care–associated A. baumannii tween case patients and control subjects, all studies did show
infections. The impressive number of acquired mechanisms of a higher mortality among the case patients. Despite the lack of
resistance makes selection of an appropriate empirical anti- consistent statistical support for the findings, one has to assume
microbial agent exceedingly difficult. The diversity of resistance that A. baumannii is at least as important as other pathogens
isolated from patients. The decision to treat on the basis of a
Received 13 January 2010; accepted 9 March 2010; electronically published 26 May 2010. clinical culture result remains in the hands of the clinician, and
Reprints or correspondence: Dr Joel Fishbain, Div of Infectious Diseases, St John Hospital criteria for decision-making in this regard will not be reviewed
and Medical Ctr, 19251 Mack Ave, Ste 333, Grosse Pointe Woods, Michigan 48236 (Joel
.Fishbain@stjohn.org). in this article. We will endeavor to discuss the therapeutic op-
Clinical Infectious Diseases 2010; 51(1):79–84 tions using available clinical data (Table 1). The reader is en-
 2010 by the Infectious Diseases Society of America. All rights reserved.
1058-4838/2010/5101-0013$15.00
couraged to review the in vitro and animal studies for addi-
DOI: 10.1086/653120 tional information.

REVIEWS OF ANTI-INFECTIVE AGENTS • CID 2010:51 (1 July) • 79


Downloaded from https://academic.oup.com/cid/article-abstract/51/1/79/299253
by guest
on 27 March 2018
Table 1. Antimicrobial Agents for the Treatment of Acinetobacter Infections

a
Medication Dosage Route Toxicity Comments

Sulbactam (amp/sulb in 6 g per day IV Dermatologic, GI, nephritis Adjust for renal function; daily dose based
the United States) on sulbactam
Imipenem-cilastatin 500 mg every 6 h up to 1 g IV Phlebitis, GI, anaphylaxis, seizures, Extended infusions have been used, lim-
every 6–8 h nephritis ited data
Meropenem 500 mg to 1 g every 8 h IV GI, headache, dermatologic, hemato- Extended infusions have been used, lim-
logic, angioedema, seizure ited data
Doripenem 500 mg every 8 h IV Dermatologic, GI, anemia, anaphy- Consider extended infusion
laxis, seizure
Amikacin
Regimen 1 15 mg/kg daily IV Nephrotoxicity, ototoxicity, neuromus-
cular blockade
Regimen 2 30 mg IVent
Tobramycin
Regimen 1 4–7 mg/kg daily IV Nephrotoxicity, ototoxicity, neuromus- Peak and trough measurements as appro-
cular blockade priate for dosing regimen selected
Regimen 2 300 mg (1 ampule) twice daily IH Tobi inhaled formulation
Regimen 3 5– 20 mg IT/IVent Not for children Not FDA approved for this route of
administration
Colistin (colistimethate)
Regimen 1 5 mg/kg/day, 2–4 divided IV Nephrotoxicity and neurotoxicity Additional data still required on ideal
doses dosing
Regimen 2 1–3 million IU every 8 h IH Must be used immediately after re- 1 million IU is 80 mg of colistimethate. A
constitution to prevent accumula- variety of doses used in studies
tion of colistin–lung toxicity
Polymyxin B [25] 50,000 units daily (5 mg IT Meningeal irritation Has been used as intraventricular injec-
tion but not FDA labeled as approved
by this route
Polymyxin E (colistin [25]) 10 mg daily IT/IVent Meningeal irritation Has not been FDA approved for either
route of administration
Tigecycline 100 mg once then 50 mg ev- IV GI, shock, pancreatitis, anaphylaxis Avoid use in blood stream infections due
ery 12 h to large volume of distribution and low
mean maximum. steady-state levels
Minocycline 100 mg every 12 h IV GI, hepatic, dermatologic MIC90 lower than that of doxycycline; lim-
ited data on use in severe infections;
most active of all the tetracyclines [33]
(excluding tigecycline because of lack
of established breakpoints)

NOTE. Amp/sulb, ampicillin-sulbactam; FDA, US Food and Drug Administration; GI, gastrointestinal (eg, nausea, vomiting, and diarrhea); H, inhalational;
hepatic, jaundice and hepatitis; IT, intrathecal; IV, intravenous; IVent, intraventricular.
a
Based on adults with normal renal function.

SPECIFIC THERAPEUTIC OPTIONS of other agents when the infecting organisms were susceptible
to sulbactam in patients with A. baumannii pneumonia and
Sulbactam. Of the b-lactamase inhibitors, sulbactam pos-
blood stream infections [8–10]. A study from Israel identified
sesses the greatest intrinsic bactericidal activity against A. bau-
that treatment with ampicillin-sulbactam was the only statis-
mannii isolates [4]. Results of clinical investigations have doc-
tically significant variable associated with reduced mortality in
umented the efficacy of sulbactam (commercially available in
the United States in combination with ampicillin) in mild-to- patients with multidrug-resistant A. baumannii bloodstream
severe A. baumannii infections. Urban et al [7] have reported infection [11]. Mixed results have been reported for use of
one of the earliest experiences using ampicillin-sulbactam and sulbactam to treat A. baumannii meningitis, and this likely
observed that 9 of 10 patients who were seriously ill and re- relates to impaired drug penetration. The optimal dosage of
ceiving mechanical ventilation demonstrated clinical improve- sulbactam to treat serious A. baumannii infections is unknown,
ment using ampicillin-sulbactam at a dosage of 3 g of amicillin but most authors recommend at least 6 g per day in divided
and 1.5 g of sulbactam intravenously every 6 or 8 h. Sulbactam- doses for patients with normal renal function (Table 1).
containing regimens appeared to be comparable to regimens Whether higher dosages are more efficacious or reduce the risk

80 • CID 2010:51 (1 July) • REVIEWS OF ANTI-INFECTIVE AGENTS


Downloaded from https://academic.oup.com/cid/article-abstract/51/1/79/299253
by guest
on 27 March 2018
of resistance, or even whether ampicillin-sulbactam should be for longer treatment courses). There has been much debate on
used in combination with other agents (as recommended by the usefulness of these agents in pneumonia, but a detailed
others), is yet to be determined [8]. review of this issue is well beyond the scope of this article. One
Carbapenems. Despite the absence of randomized con- study by Gounden et al [20] demonstrated tobramycin’s com-
trolled trials, the carbapenems (imipenem, meropenem, or do- parative activity and toxicity in treating A. baumannii infections
ripenem) remain one of the most important therapeutic op- with that of colistin. In that study, 32 patients (retrospective
tions for serious infections caused by multidrug-resistant A. cohort) were found to have received therapy in each group.
baumannii. They have excellent bactericidal activity and sta- They found no statistically significant difference in intensive
bility toward a range of b-lactamases. Unfortunately, increasing care unit mortality (total in-house mortality favored colistin),
carbapenem resistance is creating therapeutic challenges, es- increase in serum creatinine over baseline, or time to micro-
pecially considering that most A. baumannii strains that are biologic clearance [20].
resistant to the carbapenems are also resistant to the majority The efficacy of inhaled antibiotics, including aminoglyco-
of other antibiotics (except the polymyxins or tigecycline). Sus- sides, outside the cystic fibrosis population is of increasing
ceptibility to the carbapenems ranges from 190% to as low as interest. Unfortunately, very little clinical data exists for inhaled
32% depending on the geographic region and the carbapenem aminoglycosides (outside of cystic fibrosis patients). Tobi (No-
tested [12–16]. Discordance in susceptibilities between the car- vartis Pharmaceuticals), a specific formulation of inhaled to-
bapenems is well described. Norskov-Lauritsen et al [13] re- bramycin, has been approved by the US Food and Drug Ad-
ported that 13% of isolates were resistant to imipenem, yet ministration (FDA) for pseudomonas infections in cystic
23% were resistant to meropenem. Conversely, Ikonomidis et fibrosis patients only. Hallal et al [21] investigated the utility
al [16] reported that 59% of isolates were meropenem suscep- of inhaled versus intravenous tobramycin, both in combination
tible, whereas only 32% were imipenem susceptible. A clear with an intravenous b-lactam, in a pilot randomized study
understanding of local susceptibility patterns and sufficient lab- (n p 10). All 5 patients in the inhaled tobramycin group sur-
oratory support to test the carbapenems of interest are required vived, but only 3 survived in the intravenous tobramycin group.
when deciding on treatment. One cannot equate imipenem It should be noted, however, that these 2 patients had higher
results with meropenem. The clinical impact of discordant re- multiple organ dysfunction scores [21]. Historically, aminogly-
sults is exemplified in the case report by Lesho et al [17]. They cosides have been used mostly in combination therapy, and at
describe a fatal case of A. baumannii pneumonia in which least for Pseudomonas aeruginosa, monotherapy appears to be
meropenem was administered on the basis of imipenem sus- inferior to other agents.
ceptibility, yet the isolate was later proven to be meropenem Polymyxins. Unfortunately, familiarity with this older class
resistant. Doripenem is a new carbapenem with equivalent in of antibiotic is now increasing in many parts of the world. The
vitro activity against A. baumannii isolates. There does not polymyxins include colistin or polymyxin E and polymyxin B,
appear to be any advantage of doripenem when compared with and this class of drug has been a savior for the treatment of
imipenem and meropenem, however [18]. Discordant results highly drug-resistant gram-negative bacteria. Colistin is most
between doripenem and imipenem or meropenem have been commonly used in the United States, and it is administered
reported against a few isolates of A. baumannii [18]. intravenously as a pro-drug known as colistimethate sodium
Aminoglycosides. Amikacin and tobramycin are the 2 (CMS). Colistin sulfate is used topically but, most importantly,
agents that appear to retain activity against many A. baumannii is also the form that should be used in the laboratory for
isolates. As with all antimicrobial agents and multidrug-resis- susceptibility testing. Current Clinical and Laboratory Stan-
tant pathogens, resistance is increasing, and susceptibility test- dards Institute breakpoints for colistin are ⭐2 mg/mL (suscep-
ing is required to ensure activity. A recent analysis of discordant tible) and ⭓4 mg/mL (resistant).
susceptibility testing by Akers et al [19] has generated some A wide range of observational studies have now been pub-
significant concerns about automated testing for aminoglyco- lished on the clinical efficacy and toxicity of colistin for treating
side activity against A. baumannii. The percentage of isolates modern day gram-negative bacteria [4]. Efficacy ranges from
with amikacin susceptibility was reported as 53% by Vitek 2 ∼55% to 180% depending on the study and appears to be equal
(bioMérieux), compared with only 17% by Etest (AB Biodisk) to that of other antibiotics in similar populations. Nephrotox-
and broth microdilution [19]. Results were similar for tobra- icity and neurotoxicity remain as key concerns for increasing
mycin, but gentamicin did not display any discordance. To- use in an era of multidrug-resistant pathogens [22]. Nephro-
bramycin maintained the highest overall susceptibility rates toxicity is a particular issue for those with preexisting renal
[19]. impairment, the elderly population, and those who receive con-
Aminoglycosides are not often used as single agents for treat- comitant nephrotoxins. Many authors who have reported on
ment, and the toxicity profiles often hinder their use (especially the use of colistin have been pleasantly surprised with its tol-

REVIEWS OF ANTI-INFECTIVE AGENTS • CID 2010:51 (1 July) • 81


Downloaded from https://academic.oup.com/cid/article-abstract/51/1/79/299253
by guest
on 27 March 2018
erability. As has been discussed elsewhere, dosing remains con- reported for serious infections [30]. However, because of the
fusing, because formulations differ between countries [23]. rapid movement of tigecycline into tissues after intravenous
Current parenteral formulations in the United States are avail- administration, we would recommend avoiding tigecycline use
able as either Coly-Mycin M Parenteral (Parkdale Pharmaceu- alone for A. baumannii bloodstream infections (especially if the
ticals) or generically via a variety of manufacturers. The rec- MIC is ⭓1 mg/mL) [31]. Mean maximum serum steady-state
ommended dosages are 2.5–5.0 mg/kg per day of colistin base concentrations are only 0.63 mg/mL because of the large volume
given in 2–4 divided doses (equivalent to 6.67–13.3 mg/kg per of distribution [4].
day of CMS) in those with normal renal function. Additional Tetracyclines. Minocycline and doxycycline are both avail-
work is still required to determine the ideal dosing of intra- able by intravenous infusion and minocycline is approved by
venous colistin to maintain efficacy and minimize toxicity. the FDA for use in acinetobacter infections. In vitro suscep-
Promising data are available for the use of inhaled CMS as tibility testing is required for each agent to demonstrate sus-
adjunctive treatment for pneumonia caused by multidrug-re- ceptibility. Tetracycline cannot be used as a surrogate marker,
sistant A. baumannii. However, more data are still required. because many tetracycline-resistant isolates prove to be sus-
Concerns about lung toxicity, drug distribution, alveolar pen- ceptible to minocycline [32]. As with the aminoglycosides, Ak-
etration, emergence of resistance, and selection for organisms ers et al [33] also reported discordant susceptibility results be-
inherently resistant to colistin are all justified and still need tween broth microdilution and Etest/disk diffusion among all
clarification [24]. According to a recent FDA health alert, those 3 agents tested. A study by Hawley et al [34] examined 142 A.
prescribing nebulized CMS should use it immediately after baumannii isolates from US soldiers. They found that the MIC90
preparation to prevent build up of the active colistin form, for minocycline was 4, compared with 8 for tigecycline and
which can be toxic to the lungs. 116 for doxycycline, thus supporting the concept of clinical
Although A. baumannii meningitis remains an uncommon use in A. baumannii infections [34]. Clinical data is surprisingly
health care–associated infection, its incidence is increasing, and limited for minocycline and doxycyline. One study demon-
it is often caused by multidrug-resistant organisms [25]. Car- strated success in 7 of 8 patients who received minocycline for
bapenems should be used to treat these infections if the or- wound infections, and another study found 6 of 7 patients with
ganism is susceptible, with or without an intrathecal or in- ventilator-associated pneumonia were successfully treated with
traventricular aminoglycoside. For carbapenem-resistant cases, one of these agents [32].
intravenous polymyxin (colistin or polymyxin B) plus an in-
trathecal or intraventricular polymyxin or aminoglycoside, with COMBINATION THERAPY
or without intravenous rifampin, would be recommended [25]. A significant amount of time and energy has been devoted to
Multiple case series and reports have now been published on studying combination therapy for the treatment of A. bau-
the favorable efficacy and toxicity profile of intraventricular/ mannii infection. Much of the current information is derived
intrathecal colistin [25, 26]. Chemical meningitis appears to be from in vitro or animal studies. There are surprisingly limited
uncommon. The dosing recommended by the Infectious Dis- data from comparative studies involving human A. baumannii
eases Society of America for adults is 10 mg daily of colistin infections. Petrosillo et al [35] produced a nice review on co-
or 5 mg daily of polymyxin B [27]. listin versus combination therapy. They found only 4 relevant
Tigecycline. Tigecycline is the first of a new class of anti- clinical studies, and only 1 study demonstrated any statistical
biotics known as the glycylcyclines. It is a semisynthetic deriv- significance with respect to mortality, favoring monotherapy.
ative of minocycline and inhibits the 30S ribosomal subunit. The heterogeneity of combinations and infections studied, as
Its advantage over other tetracycline antibiotics is its ability to well as the small numbers of patients involved, makes inter-
evade the traditional tetracycline-specific resistance mecha- pretation very difficult [35]. Bassetti et al [36] conducted a
nisms—tet(A-E) and tet(K) efflux pumps and the tet(O) and prospective noncomparative study of colistin-rifampin com-
tet(M) determinants that provide ribosomal protection—and bination involving critically ill patients with pneumonia and
therefore it has a broader spectrum of activity. Recent global bacteremia. They observed a clinical and microbiological re-
in vitro data suggest that the 90% minimum inhibitory con- sponse rate of 76% with a mortality rate of 21%. All findings
centration (MIC90) for carbapenem-resistant A. baumannii iso- are considered to be within previously reported ranges of treat-
lates is 2 mg/mL [28]. Susceptibility breakpoints from the Clin- ment outcomes. The noncomparative nature of the study makes
ical and Laboratory Standards Institute have not yet been any concrete conclusions difficult. Lee et al [37] reported suc-
determined. Despite tigecycline’s in vitro activity against A. cessful treatment with carbapenem-sulbactam combination
baumannii, clinical data remains limited. Studies are observa- therapy in 4 patients (1 of whom received meropenem and 3
tional and are often clouded by the effects of combination of whom received imipenem) despite all isolates showing re-
antibiotic therapy [29]. Favorable clinical responses have been sistance to both agents. Falagas et al [38] conducted a retro-

82 • CID 2010:51 (1 July) • REVIEWS OF ANTI-INFECTIVE AGENTS


Downloaded from https://academic.oup.com/cid/article-abstract/51/1/79/299253
by guest
on 27 March 2018
spective cohort study involving patients who were treated with have shown benefits for extended-infusion b-lactams [42], pro-
colistin versus colistin-meropenem. They found no difference spective clinical trials are lacking. It must also be highlighted
in outcomes between the 2 groups; however, the number of that the pharmacokinetic and pharmacodynamic benefits of
monotherapy patients was small. Overall, there is far more in extended-infusion b-lactams attenuate in patients with increas-
vitro and in vivo data available than there is data from clinical ing renal impairment, which is a common comorbidity in pa-
studies, which makes any direct applicability of the data to tients with Acinetobacter infection who are hospitalized in in-
clinical care problematic. To summarize the extensive in vitro tensive care units. Finally, it is unknown whether the use of
information, the most significant data on combination therapy extended-infusion carbapenems will reduce the emergence of
pertains to colistin and rifampin or a carbapenem [4, 39]. antibiotic resistance in Acinetobacter; however, promising re-
Because of the lack of well-controlled comparative trials of sults exist for Pseudomonas aeruginosa [43].
combination therapy for A. baumannii infections, we cannot
make any specific recommendations related to the various SUMMARY
agents available for therapy.
Acinetobacter infections remain difficult to treat. The prevalence
of drug-resistant strains is increasing, and treatment options
PROLONGED INFUSION b-LACTAMS
are increasingly limited. Effective therapy remains likely when
Because of the emergence of broad-spectrum antibiotic resis- the organism is proven to be susceptible. Recent data would
tance in Acinetobacter species and the dearth of new antibac- suggest that, for some agents, a single testing method may result
terials, greater emphasis has been placed on optimizing the use in incorrect susceptibility results and lead the clinician to select
of currently available agents. Significant attention has been jus- a potentially ineffective agent. Because of some of the unusual
tifiably directed toward using pharmacokinetic and pharma- and potentially toxic options, one must maintain a high index
codynamic parameters to tailor antibacterial dosing. This log- of suspicion for A. baumannii infection and select empirical
ical approach accounts for characteristics of the drug, the therapy wisely. The challenge for the clinician is to combine
pathogen, and the host. Despite the long-term knowledge and local susceptibility patterns with the agents that are most likely
understanding of time-dependent versus concentration-depen- to be effective. The use of combination therapy, whether em-
dent antimicrobial activity, physicians are only recently mod- pirical or targeted, has yet to be demonstrated, and hopefully
ifying their antibiotic dosing in an attempt to incorporate these future studies will elucidate a greater number of definitive and
principles. practical options.
Clinical literature is emerging on the use of extended-infu-
sion b-lactams to treat gram-negative bacteria, especially with Acknowledgments
cefepime, piperacillin-tazobactam, and the carbapenems (mer- Potential conflicts of interest. A.P. has been a consultant for Abbott
openem, imipenem, and doripenem). One of the key advan- Molecular and Ortho-McNeil-Janssen. J.F.: no conflicts.
tages of extended-infusion b-lactams is the ability to achieve
drug concentrations above the MIC for a greater time for less- References
susceptible organisms, especially those with an MIC between 1. Maragakis LL, Perl TM. Acinetobacter baumannii: epidemiology, an-
4 mg/mL and 16 mg/mL. For example, Li et al [40] have shown timicrobial resistance, and treatment options. Clin Infect Dis 2008;
that the likelihood of achieving bactericidal target attainment 46(8):1254–1263.
2. Jang T, Lee S, Huang C, Lee C, Chen W. Risk factors and impact of
with meropenem, which equates to a T 1MIC of at least 40%, nosocomial acinetobacter baumannii blood stream infections in the
increases from 64% to 90% when the infusion time of 1 g is adult intensive care unit: a case-control study. J Hosp Infect 2009; 73:
increased from 0.5 h to 3 h for an organism with an MIC of 143–150.
3. Mahgoub S, Ahmed J, Glatt AE. Underlying characteristics of patients
4 mg/mL (susceptible breakpoint). Importantly, modeling stud- harboring highly resistant Acinetobacter baumannii. Am J Infect Con-
ies that use data from patients with ventilator-associated pneu- trol 2002; 30(7):386–390.
monia support such findings and have shown that, by increas- 4. Peleg AY, Seifert H, Paterson DL. Acinetobacter baumannii: emergence
of a successful pathogen. Clin Microbiol Rev 2008; 21(3):538–582.
ing the dose of meropenem to 2 g every 8 h and increasing
5. Falagas ME, Rafailidis PI. Attributable mortality of Acinetobacter bau-
the infusion time to 3 h, bactericidal target attainment can be mannii: no longer a controversial issue. Crit Care 2007; 11(3):134.
achieved for organisms with an MIC of 16 mg/mL (intermediate 6. Falagas ME, Bliziotis IA, Siempos II. Attributable mortality of Acine-
tobacter baumannii infections in critically ill patients: a systematic re-
breakpoint) [41]. Data are also available for extended-infusion
view of matched cohort and case-control studies. Crit Care 2006; 10(2):
imipenem. Both imipenem and meropenem have short half- R48.
lives (⭐4 h), and this limits their usefulness for infusions be- 7. Urban C, Go E, Mariano N, et al. Effect of sulbactam on infections
yond 3 h. Conversely, doripenem has a longer half-life [18] caused by imipenem-resistant Acinetobacter calcoaceticus biotype ani-
tratus. J Infect Dis 1993; 167(2):448–451.
and has been administered as an extended-infusion in licensing 8. Wood GC, Hanes SD, Croce MA, Fabian TC, Boucher BA. Comparison
studies. Despite modeling and retrospective clinical studies that of ampicillin-sulbactam and imipenem-cilastatin for the treatment of

REVIEWS OF ANTI-INFECTIVE AGENTS • CID 2010:51 (1 July) • 83


Downloaded from https://academic.oup.com/cid/article-abstract/51/1/79/299253
by guest
on 27 March 2018
acinetobacter ventilator-associated pneumonia. Clin Infect Dis 2002; intrathecal colistin. A new case and a systematic review of the litera-
34(11):1425–1430. ture. Int J Infect Dis 2009 (Epub ahead of print).
9. Oliveira MS, Prado GV, Costa SF, Grinbaum RS, Levin AS. Ampicillin/ 27. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the
sulbactam compared with polymyxins for the treatment of infections management of bacterial meningitis. Clin Infect Dis 2004; 39(9):1267–
caused by carbapenem-resistant Acinetobacter spp. J Antimicrob Che- 1284.
mother 2008; 61(6):1369–1375. 28. Garrison MW, Mutters R, Dowzicky MJ. In vitro activity of tigecycline
10. Jellison TK, Mckinnon PS, Rybak MJ. Epidemiology, resistance, and and comparator agents against a global collection of gram-negative and
outcomes of Acinetobacter baumannii bacteremia treated with imipe- gram-positive organisms: tigecycline evaluation and surveillance trial
nem-cilastatin or ampicillin-sulbactam. Pharmacotherapy 2001; 21(2): 2004 to 2007. Diagn Microbiol Infect Dis 2009; 65(3):288–299.
142–48. 29. Karageorgopoulos DE, Kelesidis T, Kelesidis I, Falagas ME. Tigecycline
11. Smolyakov R, Borer A, Riesenberg K, et al. Nosocomial multi-drug re- for the treatment of multidrug-resistant (including carbapenem-resis-
sistant Acinetobacter baumannii bloodstream infection: risk factors and tant) Acinetobacter infections: a review of the scientific evidence. J An-
outcome with ampicillin-sulbactam treatment. J Hosp Infect 2003; 54(1): timicrob Chemother 2008; 62(1):45–55.
32–38. 30. Poulakou G, Kontopidou FV, Paramythiotou E, et al. Tigecycline in the
12. Fraenkel CJ, Ullberg M, Bernander S, et al. In vitro activities of three treatment of infections from multi-drug resistant gram-negative patho-
carbapenems against recent bacterial isolates from severely ill patients gens. J Infect 2009; 58(4):273–284.
at Swedish hospitals. Scand J Infect Dis 2006; 38(10):853–859. 31. Peleg AY, Potoski BA, Rea R, et al. Acinetobacter baumannii bloodstream
13. Norskov-Lauritsen N, Marchandin H, Dowzicky MJ. Antimicrobial sus- infection while receiving tigecycline: a cautionary report. J Antimicrob
ceptibility of tigecycline and comparators against bacterial isolates col- Chemother 2007; 59(1):128–131.
lected as part of the TEST study in Europe (2004–2007). Int J Antimicrob 32. Bishburg E, Bishburg K. Minocycline—an old drug for a new century:
Agents 2009; 34(2):121–130. emphasis on methicillin-resistant Staphylococcus aureus (MRSA) and
14. Jones RN, Deshpande L, Fritsche TR, Sader HS. Determination of Acinetobacter baumannii. Int J Antimicrob Agents 2009; 34(5):395–401.
epidemic clonality among multidrug-resistant strains of Acinetobacter 33. Akers KS, Mende K, Yun HC, et al. Tetracycline susceptibility testing
spp. and Pseudomonas aeruginosa in the MYSTIC Programme (USA, and resistance genes in isolates of Acinetobacter baumannii-Acineto-
1999–2003). Diagn Microbiol Infect Dis 2004; 49(3):211–216. bacter calcoaceticus complex from a U.S. military hospital. Antimicrob
15. Scott P, Deye G, Srinivasan A, et al. An outbreak of multidrug-resistant Agents Chemother 2009; 53(6):2693–2695.
Acinetobacter baumannii-calcoaceticus complex infection in the US mil- 34. Hawley JS, Murray CK, Griffith ME, et al. Susceptibility of acineto-
itary health care system associated with military operations in Iraq. bacter strains isolated from deployed U.S. military personnel. Anti-
Clin Infect Dis 2007; 44(12):1577–1584. microb Agents Chemother 2007; 51(1):376–378.
16. Ikonomidis A, Pournaras S, Maniatis AN, Legakis NJ, Tsakris A. Dis- 35. Petrosillo N, Ioannidou E, Falagas ME. Colistin monotherapy vs. com-
cordance of meropenem versus imipenem activity against Acinetobacter bination therapy: evidence from microbiological, animal and clinical
baumannii. Int J Antimicrob Agents 2006; 28(4):376–377. studies. Clin Microbiol Infect 2008; 14(9):816–827.
17. Lesho E, Wortmann G, Moran K, Craft D. Fatal Acinetobacter bau- 36. Bassetti M, Repetto E, Righi E, et al. Colistin and rifampicin in the
mannii infection with discordant carbapenem susceptibility. Clin In- treatment of multidrug-resistant Acinetobacter baumannii infections.
fect Dis 2005; 41(5):758–759. J Antimicrob Chemother 2008; 61(2):417–420.
18. Paterson DL, Depestel DD. Doripenem. Clin Infect Dis 2009; 49(2): 37. Lee NY, Wang CL, Chuang YC, et al. Combination carbapenem-sul-
291–298. bactam therapy for critically ill patients with multidrug-resistant Aci-
19. Akers KS, Chaney C, Barsoumian A, et al. Aminoglycoside resistance netobacter baumannii bacteremia: four case reports and an in vitro
and susceptibility testing errors in Acinetobacter baumannii-calcoace- combination synergy study. Pharmacotherapy 2007; 27:1506–1511.
ticus complex. J Clin Microbiol 2010; 48(4):1132–1138. 38. Falagas ME, Rafailidis PI, Kasiakou SK, Hatzopoulou P, Michalopoulos
20. Gounden R, Bamford C, van Zyl-Smit R, Cohen K, Maartens G. Safe- A. Effectiveness and nephrotoxicity of colistin monotherapy vs. colis-
ty and effectiveness of colistin compared with tobramycin for multi- tin-meropenem combination therapy for multidrug-resistant Gram-
drug resistant Acinetobacter baumannii infections. BMC Infect Dis negative bacterial infections. Clin Microbiol Infect 2006; 12(12):1227–
2009; 9:26. 1230.
21. Hallal A, Cohn SM, Namias N, et al. Aerosolized tobramycin in the 39. Kiffer CR, Sampaio JL, Sinto S, et al. In vitro synergy test of meropenem
treatment of ventilator-associated pneumonia: a pilot study. Surg In- and sulbactam against clinical isolates of Acinetobacter baumannii. Di-
fect Larchmt 2007; 8(1):73–82. agn Microbiol Infect Dis 2005; 52(4):317–322.
22. Falagas ME, Kasiakou SK. Colistin: the revival of polymyxins for the 40. Li C, Kuti JL, Nightingale CH, Nicolau DP. Population pharmacoki-
management of multidrug-resistant gram-negative bacterial infections. netic analysis and dosing regimen optimization of meropenem in adult
Clin Infect Dis 2005; 40(9):1333–1341. patients. J Clin Pharmacol 2006; 46(10):1171–1178.
23. Li J, Nation RL, Turnidge JD, et al. Colistin: the re-emerging antibiotic 41. Jaruratanasirikul S, Sriwiriyajan S, Punyo J. Comparison of the phar-
for multidrug-resistant gram-negative bacterial infections. Lancet Infect macodynamics of meropenem in patients with ventilator-associated
Dis 2006; 6(9):589–601. pneumonia following administration by 3-hour infusion or bolus in-
24. Luyt CE, Combes A, Nieszkowska A, Trouillet JL, Chastre J. Aerosolized jection. Antimicrob Agents Chemother 2005; 49(4):1337–1339.
antibiotics to treat ventilator-associated pneumonia. Curr Opin Infect 42. Lodise TP Jr, Lomaestro B, Drusano GL. Piperacillin-tazobactam for
Dis 2009; 22(2):154–158. Pseudomonas aeruginosa infection: clinical implications of an extend-
25. Kim BN, Peleg AY, Lodise TP, et al. Management of meningitis due ed-infusion dosing strategy. Clin Infect Dis 2007; 44(3):357–363.
to antibiotic-resistant Acinetobacter species. Lancet Infect Dis 2009; 43. Tam VH, Schilling AN, Neshat S, Poole K, Melnick DA, Coyle EA.
9(4):245–255. Optimization of meropenem minimum concentration/MIC ratio to
26. Cascio A, Conti A, Sinardi L, et al. Post-neurosurgical multidrug- suppress in vitro resistance of Pseudomonas aeruginosa. Antimicrob
resistant Acinetobacter baumannii meningitis successfully treated with Agents Chemother 2005; 49(12):4920–4927.

84 • CID 2010:51 (1 July) • REVIEWS OF ANTI-INFECTIVE AGENTS


Downloaded from https://academic.oup.com/cid/article-abstract/51/1/79/299253
by guest
on 27 March 2018

You might also like