Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

International Journal of Antimicrobial Agents 29 Suppl.

3 (2007) S33–S41
www.ischemo.org

Non-fermentative Gram-negative bacteria


D.A. Enoch*, C.I. Birkett, H.A. Ludlam
Clinical Microbiology & Public Health Laboratory, Addenbrooke’s Hospital, Hills Road, Cambridge CB2 2QW, UK

Abstract
Over the past decade, non-fermenting Gram-negative bacteria have emerged as important opportunistic pathogens in the increasing
population of patients who are immunocompromised by their disease or medical treatment. These bacteria are assisted by their ubiquitous
distribution in the environment and have a propensity for multiple, intrinsic or acquired drug resistance. The infections that they cause now
pose significant problems in terms of treatment and infection control, whilst the commonly observed rapid emergence of bacterial resistance
to new antimicrobial compounds raises concerns regarding the clinical lifespan of these agents. Studies are urgently required to assess
whether combination therapy can improve the long-term utility of new drugs in the treatment of patients infected with non-fermenters.
© 2007 Elsevier B.V. and the International Society of Chemotherapy. All rights reserved.
Keywords: Non-fermenters; Pseudomonas aeruginosa; Stenotrophomonas maltophilia; Burkholderia cepacia; Acinetobacter baumannii

1. Introduction The European Antimicrobial Resistance Surveillance Sys-


tem (EARSS) provides data on the prevalence and
Non-fermenting Gram-negative bacteria (“non-fermenters”) spread of resistance for seven major invasive bacte-
are widespread in the environment and are an increasing ria, including P. aeruginosa (but not A. baumannii,
cause of serious infections in hospital practice, primarily B. cepacia complex and S. maltophilia), in Europe
affecting the expanding population of patients immuno- (http://www.earss.rivm.nl/), illustrating the wide geograph-
compromised by disease or by medical and surgical ical variation in antimicrobial resistance. The British
treatments. Many species are notable for their resistance Society for Antimicrobial Chemotherapy (BSAC) Resis-
to multiple antibiotics and the facility with which they tance Surveillance Programme (http://www.bsacsurv.org)
may acquire further resistances. Acquisition of resistance provides local data for the UK and Irish Republic. In
may also promote their proliferation in hospitals, leading this paper, we review recent data from these programmes
to significant antibiotic treatment and infection control and other literature for P. aeruginosa, A. baumannii,
challenges. There is a pressing need for novel antimicrobials S. maltophilia and B. cepacia complex.
to address the problem caused by these organisms.
Non-fermenters comprise numerous species belonging
to many genera. This review will focus on the four 2. The organisms
species most often causing significant problems in hospital 2.1. Pseudomonas aeruginosa
practice, namely Pseudomonas aeruginosa, Acinetobacter
baumannii, Stenotrophomonas maltophilia and members of Pseudomonas aeruginosa is widely distributed in the
environment. It is a leading cause of nosocomial infections,
the Burkholderia cepacia complex, discussing the in vitro
being responsible for 10% of all hospital-acquired infec-
activity of the drugs available to treat infections caused by
tions (HAIs) [2]. Infection is usually opportunistic and as-
these organisms.
sociated with invasive devices, mechanical ventilation, burn
Surveillance programmes demonstrate the increasing
wounds or surgery. Cross-infection from other colonised
problem of antimicrobial resistance globally. The SEN-
patients occurs and is encouraged by suppression of hospital
TRY Program was established in 1997 to measure the
patients’ normal flora with broad-spectrum antibiotics to
predominant pathogens and their antimicrobial resistance
which this organism is widely resistant. The organism also
patterns over a broad network of sentinel hospitals
has a propensity to colonise and infect the lungs of cystic
in the USA, Canada, South America and Europe [1].
fibrosis (CF) patients and is virtually ineradicable in this
* Corresponding author. D.A. Enoch. setting.
Tel.: +44 1223 257 035; fax: +44 1223 242 775. Pseudomonas aeruginosa has an impressive armamen-
E-mail: david.enoch@addenbrookes.nhs.uk (D.A. Enoch). tarium of virulence factors, allowing it to combat host

0924-8579/$ – see front matter © 2007 Elsevier B.V. and the International Society of Chemotherapy. All rights reserved.
S34 D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41

defences. These include the flagellum, pili, alginate produc-


tion, mucoid phenotypes in CF, lipopolysaccharide, type III
secretion factors, secreted proteases, oxidative factors and
toxins [3]. Quorum sensing may regulate expression of viru-
lence factors [4]. The emergence and gradual dissemination
of metallo-b-lactamases, with their ability to hydrolyze all
b-lactams except aztreonam, has further compromised the
activity of many agents traditionally used for the treatment
of P. aeruginosa infections [5], although it remains more
common for pan-resistance to arise by accumulation of
successive mutations affecting efflux, impermeability and
AmpC b-lactamase expression.
Susceptibility testing gives variable results for isolates
recovered from samples in patients with CF, and its role in
EARSS 2005 Report: http://www.rivm.nl/earss/Images/EARSS%202005_tcm61-34899.pdf
antibiotic choice has been questioned as patients commonly
Fig. 2. Proportion of invasive Pseudomonas aeruginosa isolates resistant
improve despite receiving apparently inappropriate ther- to carbapenems in 2005, from the European Antimicrobial Resistance
apy [6]. Outside this setting, laboratory-detected resistance Surveillance System (EARSS) (http://www.earss.rivm.nl). Carbapenem
is believed to be significant. resistance is high all over Europe, with only Norway and The Netherlands
reporting proportions of <5%. Reproduced with permission from
EARSS.
2.1.1. Trends in isolation and resistance
2002 (n=171) 2003 (n=185) 2004 (n=207) 2005 (n=197)
In the SENTRY Program, the numbers of isolates of 20.0
P. aeruginosa reported are increasing, although distinct 18.0
differences exist by geographic region and site of infection. 16.0
SENTRY data reported that multidrug-resistant (resistant
14.0
to piperacillin, ceftazidime, imipenem and gentamicin)
12.0
% resistance

P. aeruginosa bloodstream isolates were most prevalent


10.0
in Latin America (12.0−17.6%; average 15.0%), fol-
8.0
lowed by Europe (5.1−14.2%; average 9.3%) and North
6.0
America (1.6−2.5%; average 2.1%) between 1997 and
2001 [1] (Fig. 1). Multidrug resistance in bloodstream 4.0

infections saw a steady increase between 1997 and 2001, 2.0

with resistance even to the carbapenems widespread in 0.0


Ceftazidime Gentamicin Imipenem Ciprofloxacin
Europe (Fig. 2). The UK situation is better: the BSAC
Fig. 3. British Society for Antimicrobial Chemotherapy bacteraemia
bacteraemia survey reported relatively stable or declining surveillance data for Pseudomonas aeruginosa from 2002–2005 [7] for
rates of resistance among P. aeruginosa isolates between the UK and Republic of Ireland.
2002 and 2005, except possibly to ciprofloxacin (Fig. 3).
North American surveillance studies report a rise in
aminoglycoside [8], carbapenem [9] and fluoroquinolone [8]
Europe (1152) Latin America (608) North America (1550) resistance. By 2005, US studies reported ceftazidime and
20.0
imipenem MIC50 values (minimum inhibitory concentration
17.6 for 50% of the organisms) ranging from <1 mg/L to
16.0
14.3 14.2 14.5 >32 mg/L [10,11] and from 0.06 mg/L to 16 mg/L [10],
% MDR Resistance

15.0
12.0 respectively, with this great diversity perhaps reflecting
10.1 10.4
10.0
9.5 different patient groups or the effects of local outbreak
strains with more or less resistance. In CF, MIC50 values
5.1 for ceftazidime and imipenem ranged from 0.25 mg/L to
5.0
2.5
1.6 2.1 2.0 2.0 >512 mg/L and 0.5 mg/L to >512 mg/L, respectively [12].
0.0
Reported rates of multidrug-resistant P. aeruginosa from the
1997 1998 1999 2000 2001 SENTRY Program varied from 1.1% to 31.2% according to
Fig. 1. SENTRY Program data for multidrug-resistant (MDR) Pseu- geographic location [13].
domonas aeruginosa from bloodstream isolates (1997–2001). Adapted
from ref. [1]. Whilst multidrug resistance remained relatively stable 2.2. Acinetobacter baumannii
between 1997 and 2001 in North America, it increased from 12.0 to 17.6%
of P. aeruginosa isolates in Latin America over the same time period and Acinetobacter baumannii is also widely distributed in the
more than doubled in Europe. environment. It is generally non-pathogenic in healthy
D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41 S35

Cumulative no. hospitals affected


individuals, except possibly as an agent of wound infection 80

in military casualties from the Middle East [14,15]. Over the 70


last decade it has become an increasing cause of serious 60
opportunistic infection, particularly ventilator-associated 50
pneumonia and invasion of burns wounds, in susceptible 40
patients in Intensive Care Units (ICUs). Virulence factors 30
are limited, consistent with the organism’s limited invasive 20
potential. It produces no cytotoxins and its lipopolysaccha-
10
ride is of uncertain endotoxigenic potential. Its enhanced
0
survival is due to a combination of bacteriocin production,

Q3

Q3
Q4

Q4

Q2
0

4
Q2
Q3
Q4

Q2

Q4

Q2

Q2
Q3

Q3
Q4
the presence of a capsule and prolonged viability under

200

200
200

200

200
dry conditions [16]. The population structure is highly
S.E. clone OXA-23 clone 1 OXA-23 clone 2
clonal, with single strains commonly affecting multiple
Fig. 5. Carbapenem-resistant Acinetobacter clones in the UK (2000–2004).
patients in a unit or hospital. Indeed, a few lineages Reproduced from ref. [23]. These data illustrate the rise of carbapenem-
of A. baumannii have achieved “epidemic” status as resistant clones in the UK over a 5-year period between 2000 and 2005.
nosocomial pathogens, spreading to cause outbreaks of There was no evidence of widely disseminated clones at the beginning in
infections in many hospitals and countries [17]. Many of 2000, whereas by 2004 the SE clone and the OXA-23 clone affected ca.
40 hospitals in the UK.
these outbreaks have involved strains that were multidrug
resistant, including to carbapenems and amikacin, with OXA-type carbapenemases and class B metalloenzymes
colistin and tigecycline sometimes representing the only (IMP or VIM family), whilst PER extended-spectrum
remaining therapeutic options [17,18]. enzymes cause resistance to third-generation cephalosporins
in many isolates in Turkey [24].
2.2.1. Trends in isolation and resistance Multidrug-resistant isolates of A. baumannii have in-
BSAC surveillance data illustrate resistance trends in creased significantly in the UK since 2002, becoming most
A. baumannii for bacteraemias only since 2001 [7]. In prevalent in hospitals in London and southeast England [19,
2005, >30% of bacteraemia isolates were resistant to 20]. Pulsed-field gel electrophoresis has shown that many of
gentamicin and piperacillin/tazobactam [7] (Fig. 4). Many these isolates belong to three clones that possess the non-
isolates from non-bacteraemic sources are even more metallo OXA-23 or -51 carbapenemases as the predominant
resistant [19,20]. Similar trends have been reported in other mechanism for their carbapenem resistance. All three
countries [21]. A recent study from Greece reported >94% exhibit resistance to b-lactams (including carbapenems),
resistance to ceftazidime, amikacin, piperacillin/tazobactam fluoroquinolones and many aminoglycosides, although they
and imipenem, whilst colistin MICs ranged from 0.25 mg/L vary in susceptibility to amikacin. The most prevalent
to >1024 mg/L and those of tigecycline from 0.12 mg/L clones, each recorded from ca. 40 hospitals, are the SE
to 4 mg/L [22]. In the USA, ceftazidime and amikacin clone and the OXA-23 clone l; their spread since 2000 is
MICs range from <0.06 mg/L to 128 mg/L and <0.5 mg/L illustrated in Fig. 5 [23].
to >64 mg/L, respectively. Several mechanisms for car- Risk factors for acquisition of multidrug-resistant A. bau-
bapenem resistance have been described, namely class D mannii include presence on an ICU or burns unit, large
hospital size (>500 beds) [25] and previous exposure to
2002 (n=28) 2003 (n=38) 2004 (n=41) 2005 (n=43) antibiotics [25,26], notably carbapenems [27]. Immunosup-
45.0
pression, emergency admission, respiratory failure or me-
40.0 chanical ventilation, invasive procedures, urinary catheteri-
35.0 sation and recent surgery are further risk factors [25–27].
30.0
Attributable mortality is hard to define, since A. bauman-
nii tends only to cause infection in very vulnerable patients
% resistance

25.0
and its properties may be clone-specific; nevertheless, rates
20.0
up to 20% [28] and >61% [29] have been claimed in
15.0
bacteraemia. Its capacity to acquire resistance to multiple
10.0 antimicrobial agents has made this organism a significant
5.0 challenge in many healthcare institutions.
0.0
Gentamicin Pip/tazo Imipenem Ciprofloxacin 2.3. Burkholderia cepacia complex
Fig. 4. British Society for Antimicrobial Chemotherapy bacteraemia
Members of B. cepacia complex are well recognised,
surveillance data for Acinetobacter baumannii from 2002–2005 [7]. The
data show increasing rates of resistance, with levels reaching over 30% for if uncommon, nosocomial pathogens. They are more
gentamicin, piperacillin/tazobactam (Pip/tazo) and ciprofloxacin and over important in CF, where the spread of epidemic strains
5% for imipenem by 2005. has presented significant infection control problems, both
S36 D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41

in hospital units and among the CF community as a antibiotics [39]. Nosocomial transmission of S. maltophilia
whole [30]. is rare and most infections are with unique strains.
A number of potential virulence factors have been Stenotrophomonas maltophilia is associated with signif-
identified, although not all of these have been proven to icant morbidity and mortality in immunocompromised
affect the pathogenesis of human disease [31]. They in- patients [39]. Putative, but poorly understood, virulence
clude intrinsic antimicrobial resistances (e.g. to polymyxins factors include a range of exoenzymes such as DNase,
and aminoglycosides), the ability to form biofilms, pili, RNase, fibrinolysin, lipases, hyaluronidase, protease and
the cenocepacia (pathogenicity) island [32], production of elastase, the ability to survive and then multiply in medical
exopolysaccharide, haemolysin, melanin and extracellular solutions and the ability to adhere to prosthetic materials.
proteases, flagella, lipopolysaccharide, siderophore produc- Bacteraemias related to central venous catheters and
tion, type III secretion factors and the ability to invade and nosocomial pneumonia are the two most common man-
survive inside respiratory cells and macrophages. ifestations of true infection due to S. maltophilia, al-
Burkholderia multivorans (genomovar II) and Burkholde- though most isolates originating from the respiratory tract
ria cenocepacia (genomovar III) account for the majority represent colonisation rather than infection. Recovery of
of infecting strains in Europe and the USA, and iso- the organism from this site, or from urinary catheters,
lation of B. cepacia complex isolates from respiratory should not ordinarily prompt antibiotic treatment. In rare
CF samples is accepted as a poor prognostic marker [33]. cases, S. maltophilia is associated with skin and soft-tissue
A fulminant pneumonic and septicaemic process called infections, endocarditis, meningitis, urinary tract infections,
“cepacia syndrome” may occur in 10% of colonised intra-abdominal infections and ophthalmological syndromes
CF patients, more commonly with B. cenocepacia than related to applied or implanted foreign materials.
B. multivorans [32]. Patients with B. cenocepacia had a Mortality rates of 10−60% in patients with bacteraemia
shorter survival than patients with P. aeruginosa (P = 0.01) due to S. maltophilia have been reported [40,41], although
in a recent study but there was no difference in survival attributable mortality is again hard to define in the types
between P. aeruginosa and B. multivorans [32]. Some of patient sufficiently ill to be affected by this species.
centres decline lung transplantation to patients colonised The majority of isolates remain sensitive to co-trimoxazole,
with B. cepacia complex [34]. although toxicity or intolerance remains the major problem
Burkholderia pseudomallei is prevalent in Southeast Asia in some patients, for whom ticarcillin/clavulanic acid may
and the Northern Territory of Australia both as a soil be considered as an alternative. Fluoroquinolones may be
organism and as a primary pathogen, often in patients with useful as adjuncts to co-trimoxazole or ticarcillin/clavulanic
underlying diabetes mellitus. It is associated with 30−40% acid therapy [42].
mortality in septicaemic disease, even when treated with
appropriate antibiotics, although with much lower mortality 2.4.1. Trends in isolation and resistance
rates for pulmonary disease. Tigecycline has an MIC90 The number of isolates of S. maltophilia reported are
of 2 mg/L [35] and may be an alternative therapy to increasing worldwide, although reports tend to be from
ceftazidime for melioidosis, although clinical trials are individual institutions and associated with the introduction
needed to support this hypothesis. of carbapenem use [43]. This increase may reflect a change
in patient types rather than a change in epidemiology.
2.3.1. Trends in isolation and resistance Among ‘uncommonly isolated’ non-enteric Gram-nega-
A recent review of HAIs related to contaminated substances tive bacilli, recent SENTRY data report S. maltophilia
reported B. cepacia to be a significant contaminant of to comprise 59.2%, with a roughly even distribution
substances other than blood products [36]. SENTRY data between respiratory and blood isolates (49% and 41%,
reported B. cepacia to comprise 7.7% of “uncommonly respectively) [11]. Distinct differences exist in the frequency
isolated” non-enteric Gram-negative bacilli between 1997 by geographic region and site of infection [43].
and 2003 [11]. In the USA, resistance ranges from 9.3% for Co-trimoxazole resistance varies widely. Betriu et al. [44,
co-trimoxazole to 48% for imipenem [11]. Among patients 45] reported that 2% of Spanish isolates of S. maltophilia
with CF, resistance to imipenem was >84% in Europe were resistant, whereas an Italian study reported 80.9%
and the USA [12,37]. MIC50 values for piperacillin ranged resistance [46]. The highest levels of resistance are seen in
from 4 mg/L [38] to 256 mg/L [12] for multidrug-resistant CF isolates, with 84% resistance to co-trimoxazole, 50%
CF isolates. to ticarcillin/clavulanic acid, 74% to colistin and 11% to
doxycycline [47]. A recent surveillance study reported that
2.4. Stenotrophomonas maltophilia >40% of pneumonia isolates from the USA are resistant
Independent risk factors for infection with S. maltophilia to b-lactams and ciprofloxacin [48]. A further problem –
include chronic obstructive pulmonary disease, neutropenia, and caution – when reading surveillance data is that the
the presence of a central venous catheter, prolonged MICs of b-lactams vary with the medium [49]; those of
hospitalisation and previous therapy with broad-spectrum aminoglycosides vary with the test temperature [49].
D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41 S37

3. Antibiotics for treatment of infections due to 3.4. Penicillins with activity against non-fermenters
non-fermentative Gram-negative bacteria A number of semisynthetic penicillins, including ticarcillin
and piperacillin, have activity against non-fermenters. These
3.1. Aminoglycosides agents can be used in combination with the b-lactamase
Aminoglycosides are a long-established component of inhibitors clavulanic acid, tazobactam or sulbactam, al-
therapy for infections due to non-fermenters, although they though these do not inhibit all b-lactamases of these
should be seen as adjuncts rather than as sole agents [50]. species. Moreover, most resistance in P. aeruginosa is
Against P. aeruginosa they are generally combined with due to non-b-lactamase mechanisms, principally efflux, and
b-lactams, although the evidence for synergy is weak except b-lactamase inhibitors have no effect on this mechanism.
with penicillins. Bacterial resistance may occur owing Resistance rates are typically highest in isolates from
to reduced drug uptake, increased efflux pump activity CF patients [12] and in outbreaks of carbapenem-resistant
and enzymatic inactivation. This topic has been reviewed P. aeruginosa and A. baumannii [61]. Temocillin is used for
elsewhere [51]. They have no in vitro activity against the treatment of B. cepacia in CF, but otherwise has little
S. maltophilia and B. cepacia. Tobramycin is inherently the activity against non-fermenters [62].
most active of the group against P. aeruginosa, where most
resistance to aminoglycosides in general is mutational due 3.5. Cephalosporins
to impermeability or efflux [51]. Pseudomonas aeruginosa The cephalosporin with most potent activity against non-
isolates with the highest MICs were most commonly from fermenters is ceftazidime [63]. Its spectrum of activity
CF patients, with tobramycin MIC50 and MIC90 values of includes P. aeruginosa, S. maltophilia, A. baumannii,
8 mg/L and 64 mg/L for mucoid isolates and 16 mg/L and B. cepacia and B. pseudomallei, for which it is standard
256 mg/L for non-mucoid isolates, respectively [12]. therapy. Resistance occurs via a variety of mechanisms,
including hydrolysis by b-lactamases and efflux (e.g. by
upregulation of MexAB–OprM in P. aeruginosa) [64].
3.2. Co-trimoxazole
The combination of trimethoprim and sulphamethoxazole 3.6. Carbapenems
is active in vitro against many isolates of B. cepacia and
Carbapenems are b-lactams with exceptionally broad an-
S. maltophilia, but not P. aeruginosa. Acquired resistance
tibacterial spectra. Meropenem and imipenem/cilastatin are
is common in A. baumannii. Co-trimoxazole is the
active against non-fermenters. Ertapenem has no activity
treatment of choice for infections with S. maltophilia [52]
against these organisms and will not be discussed here [65].
and significant morbidity may occur in patients with
Resistance in Gram-negative organisms usually results from
co-trimoxazole-resistant S. maltophilia bacteraemia [53].
a combination of impaired drug entry, efflux and the
Despite the Committee for Safety of Medicines in the UK
presence of a b-lactamase. Diminished permeability is
advising against its general use, co-trimoxazole remains
often a result of loss of the outer membrane proteins
the standard therapy for these infections, and trimethoprim through which carbapenems enter the periplasmic space
therapy alone is not active. and is a well known mechanism for P. aeruginosa
where loss of OprD is associated with resistance [66].
3.3. Colistin Multidrug efflux systems (e.g. MexAB–OprM) are present
in P. aeruginosa and can excrete meropenem as well
The polymyxins are a group of five chemically differ- as penicillins, fluoroquinolones and cephalosporins [66],
ent bactericidal antibiotics (polymyxins A to E). Only although it is important to note that imipenem escapes this
polymyxin B and polymyxin E (colistin) have been used in type of mechanism [67]. Stenotrophomonas maltophilia is
clinical practice. Resistance may develop through mutation intrinsically resistant to carbapenems owing to production
and occurs of a result of alterations of the outer membrane. of inducible zinc-metalloenzyme (L-1 enzyme) with car-
Colistin has a wide spectrum of activity against Gram- bapenemase activity [68]. Meropenem is one of the most
negative organisms, including A. baumannii, P. aeruginosa active agents versus B. cepacia and some would regard it
and S. maltophilia, but is inactive against B. cepacia as the agent of choice.
owing to its unusual lipopolysaccharide structure. Although Doripenem is currently undergoing six phase III trials
resistance to polymyxin B has recently been reported in for a variety of conditions including complicated urinary
P. aeruginosa isolates from the USA and elsewhere [54], it tract infection, pneumonia and complicated intra-abdominal
remains rare and is almost exclusive to CF isolates. Recent infections. It has similar activity to meropenem and
clinical reports and reviews of experience with colistin imipenem against carbapenemase-negative A. baumannii
are encouraging, with side effects observed less often than strains, but no carbapenem retains activity against strains
expected from historical data [55–60]. with expression of OXA or metallo-carbapenemases [69].
S38 D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41

Table 1
Potency of tigecycline against non-fermentative Gram-negative bacteria (adapted from refs. [38,77])

Organism MIC a (mg/L) % Susceptible b % Resistant b


Range MIC50 MIC90

Pseudomonas aeruginosa (n = 1121) [77] 0.12 to >32 8 16 5.1 77.2


Acinetobacter spp. (n = 326) [77] 0.06−8 0.5 2 94.5 0.9
Burkholderia cepacia (n = 21) [38] 0.25−32 1 16 67.0 29.0
Stenotrophomonas maltophilia (n = 203) [77] 0.12−8 1 2 93.1 3.0
a MIC, minimum inhibitory concentration (MIC50/ 90 , MIC for 50% and 90% of the organisms, respectively).
b The tigecycline susceptible breakpoint was defined as 2 mg/L and the resistance breakpoint as 8 mg/L for comparison purposes
only [78]; these values are now accepted by the US Food and Drug Administration (FDA) but are one doubling dilution higher than
the values adopted for Enterobacteriaceae by the European Committee on Antimicrobial Susceptibility Testing (EUCAST)/European
Medicines Agency (EMEA), who presently have no values specific for non-fermenters.

It is unlikely that doripenem will prove to be superior to of this species, and the AcrAB pump found in Proteus
meropenem, but this requires additional clinical studies. mirabilis [72]. Published resistance rates for tigecycline
range from 58.5% to 97% for P. aeruginosa [38,73], 0%
3.7. Fluoroquinolones to 6% A. baumannii [74,75], 0% to 29% for B. cepacia
complex [38,76] and 0% to 3.5% for S. maltophilia [45,48].
In general, ciprofloxacin remains the most potent agent in
Table 1 [38,77] lists the MICs of tigecycline.
this class against non-fermenters and is particularly valued
and widely used. It is the only oral agent with activity
3.9. Combination therapy
against P. aeruginosa. Fluoroquinolones have been widely
used for the treatment of a wide variety of infections. The potential benefits of combination antibiotic therapy
Perhaps predictably, their use is increasingly compromised are well recognised. They include a broadened and more
by the widespread emergence of bacterial resistance [70]. reliable spectrum of activity, enhanced antibacterial activity
For the treatment of S. maltophilia, there is some clinical due to any synergistic or additive effect allowing lower
evidence that addition of moxifloxacin (more active than doses of toxic agents to be given, and prevention of the
ciprofloxacin against this species) to co-trimoxazole may be emergence of resistance. Enhanced in vitro activity has
therapeutically advantageous [42]. Resistance mechanisms been noted for a number of antimicrobial combinations
have recently been reviewed elsewhere [70] and mostly tested against A. baumannii. These include polymyxin B,
entail reduction in permeability, upregulation of efflux (also imipenem and rifampicin [79], polymyxin B, rifampicin
affecting b-lactams) and mutation of DNA gyrase and and sulbactam [80] and colistin and rifampicin [81]. There
topoisomerase IV genes. have been reports of some centres using tigecycline and
nebulised colistin against Acinetobacter pneumonia [82],
although studies are needed to evaluate this combination
3.8. Tigecycline
formally. Enhanced in vitro activity has also been noted
Tigecycline is a glycylcycline, a semisynthetic derivative for a number of antimicrobial combinations used against
of minocycline with a 9-t-butylglycylamido substitution. P. aeruginosa, and b-lactam/aminoglycoside combinations
Glycylcyclines act by binding to the bacterial 30S ribosomal remain the standard of care for severe infections. In
subunit, blocking entry of the aminoacyl-tRNA molecules vitro synergy was demonstrated with the combination
into the acceptor site of the ribosome, and are mostly of cephalosporins and fluoroquinolones [83], colistin and
considered to be bacteriostatic. The main determinants of ceftazidime [84] and a macrolide plus tobramycin [85]. Syn-
acquired resistance to tetracyclines, namely active efflux ergy was also demonstrated in vitro with the combination
and ribosomal protection, are overcome by steric hindrance of macrolides with ceftazidime (for B. cepacia) and co-
from the large R-group at position 9. Tigecycline has in trimoxazole (for S. maltophilia) for infecting organisms
vitro activity against a number of Gram-negative species, from patients with CF [85]. Conversely, no enhanced in
including A. baumannii (including many carbapenem- vitro antibacterial activity against P. aeruginosa was found
resistant isolates) and S. maltophilia. It lacks useful with combinations of ciprofloxacin and colistin [84].
activity against P. aeruginosa. Tigecycline is able to There are few clinical studies of combinations of
evade Tet-type efflux pumps (responsible for plasmid- antimicrobial agents for the treatment of multidrug-
mediated tetracycline resistance) but is susceptible to the resistant non-fermenters. Colistin was combined with a
chromosomal resistance–nodulation–cell division (RND) carbapenem in 19 of 25 patients, amikacin in 8 of
family of multidrug efflux pumps, including MexXY–OprM 25 patients, tobramycin (3), cefepime (3), quinolone (2),
of P. aeruginosa [71], which explains the intrinsic resistance ampicillin/sulbactam (3) and aztreonam (1 patient) in a
D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41 S39

retrospective non-controlled observational study of 25 crit- Role of funding source


ically ill patients with infection due to multidrug-resistant
The supplement has been funded by an unrestricted
P. aeruginosa or A. baumannii [86]. End-of-treatment
educational grant from Wyeth UK. Wyeth UK have had no
mortality was 21%. Ten patients with carbapenem-resistant
input into the content or editorial.
A. baumannii were treated with a combination of imipenem
and rifampicin in a recent prospective study [87]. Three
patients died, two because of therapeutic failure, and seven References
responded. High-level rifampicin resistance developed in [1] Jones RN. Global epidemiology of antimicrobial resistance among
7 of 10 patients. The authors suggested this combination community-acquired and nosocomial pathogens: a five-year summary
should not be used for the treatment of severe infection with from the SENTRY Antimicrobial Surveillance Program (1997–2001).
these organisms. Semin Respir Crit Care Med 2003;24:121−34.
[2] Aloush V, Navon-Venezia S, Seigman-Igra Y, Cabili S, Carmeli Y.
Multidrug-resistant Pseudomonas aeruginosa: risk factors and
clinical impact. Antimicrob Agents Chemother 2006;50:43−8.
4. Conclusions [3] Pier GB, Ramphal R. Pseudomonas aeruginosa. In: Mandell GL,
Bennett JE, Dolin R, editors. Principles and practice of infectious
diseases. Philadelphia, PA: Churchill Livingstone; 2005. p. 216.
Pseudomonas aeruginosa, A. baumannii, S. maltophilia and [4] Heurlier K, Denervaud V, Haas D. Impact of quorum sensing on fitness
members of the B. cepacia complex are an increasingly of Pseudomonas aeruginosa. Int J Med Microbiol 2006;296:93–102.
recognised cause of infection and are increasingly difficult [5] Walsh TR. The emergence and implications of metallo-beta-
to treat. lactamases in Gram-negative bacteria. Clin Microbiol Infect
Glycylcyclines demonstrate useful in vitro activity against 2005;11(Suppl. 6):2−9.
[6] Foweraker JE, Laughton CR, Brown DF, Bilton D. Phenotypic
many non-fermenters except P. aeruginosa, but clinical variability of Pseudomonas aeruginosa in sputa from patients with
trials are needed to determine their role in the treatment acute infective exacerbation of cystic fibrosis and its impact on
of infections due to these organisms. Of particular concern the validity of antimicrobial susceptibility testing. J Antimicrob
are early reports of the development of resistance and Chemother 2005;55:921−7.
[7] British Society for Antimicrobial Chemotherapy. Resistance Surveil-
treatment failure in two patients with carbapenem-resistant
lance Website: Bacteraemia. BSAC; 2006. http://www.bsacsurv.org/
A. baumannii bacteraemia being treated with tigecycline [accessed 30 January 2007].
for other conditions [88]. The two patients had received [8] Mutnick AH, Rhomberg PR, Sader HS, Jones RN. Antimicrobial
9 days and 16 days of treatment with tigecycline when usage and resistance trend relationships from the MYSTIC
bacteraemias were detected. The MICs were 4 mg/L and Programme in North America (1999–2001). J Antimicrob Chemother
2004;53:290−6.
16 mg/L, respectively, but these fell to 1 mg/L and 4 mg/L [9] Lautenbach E, Weiner MG, Nachamkin I, Bilker WB, Sheridan A,
after addition of phenyl-arginine-b-naphthylamide, a pump Fishman NO. Imipenem resistance among Pseudomonas aeruginosa
inhibitor, suggesting efflux as the mechanism of resistance. isolates: risk factors for infection and impact of resistance on clinical
Noting a mean peak serum concentration of only 0.63 mg/L, and economic outcomes. Infect Control Hosp Epidemiol 2006;27:
the authors suggest that tigecycline should not be used 893–900.
[10] Bouchillon SK, Hoban DJ, Johnson BM, Johnson JL, Hsiung A,
to treat bloodstream infections caused by organisms with Dowzicky MJ. In vitro activity of tigecycline against 3989 Gram-
MICs >1 mg/L [88], which corresponds with the European negative and Gram-positive clinical isolates from the United States
Medicines Agency (EMEA)/European Committee on Anti- Tigecycline Evaluation and Surveillance Trial (TEST Program; 2004).
microbial Susceptibility Testing (EUCAST) breakpoint. Diagn Microbiol Infect Dis 2005;52:173−9.
[11] Sader HS, Jones RN. Antimicrobial susceptibility of uncommonly
There are currently no other drugs, besides doripenem,
isolated non-enteric Gram-negative bacilli. Int J Antimicrob Agents
in phase II or III trials for the treatment of multidrug- 2005;25:95–109.
resistant non-fermenters. Novel efflux pump inhibitors have [12] Chen Y, Garber E, Zhao Q, et al. In vitro activity of doripenem
not yet entered human clinical trials, although they appear (S-4661) against multidrug-resistant gram-negative bacilli isolated
promising [89]. Therefore, currently available antibiotics from patients with cystic fibrosis. Antimicrob Agents Chemother
2005;49:2510−1.
require judicious and prudent use, following evidence- [13] Gales AC, Jones RN, Turnidge J, Rennie R, Ramphal R.
based trial data whenever possible. The role of combining Characterization of Pseudomonas aeruginosa isolates: occurrence
these antibiotics in combating the development of further rates, antimicrobial susceptibility patterns, and molecular typing in
antibiotic resistance and delivering superior clinical efficacy the global SENTRY Antimicrobial Surveillance Program, 1997–1999.
requires more rigorous evaluation than in the past. Clin Infect Dis 2001;32(Suppl. 2):S146−55.
[14] Jones A, Morgan D, Walsh A, et al. Importation of multidrug-resistant
Acinetobacter spp. infections with casualties from Iraq. Lancet Infect
Dis 2006;6:317−8.
[15] Davis KA, Moran KA, McAllister CK, Gray PJ. Multidrug-resistant
Acknowledgements Acinetobacter extremity infections in soldiers. Emerg Infect Dis 2005;
11:1218−24.
Editorial assistance during the preparation of this manuscript [16] Wendt C, Dietze B, Dietz E, Ruden H. Survival of Acinetobacter
was provided by Magus Strategic Communications Ltd. baumannii on dry surfaces. J Clin Microbiol 1997;35:1394−7.
S40 D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41

[17] Coelho JM, Turton JF, Kaufmann ME, et al. Occurrence of [37] Bonacorsi S, Fitoussi F, Lhopital S, Bingen E. Comparative in
carbapenem-resistant Acinetobacter baumannii clones at multiple vitro activities of meropenem, imipenem, temocillin, piperacillin,
hospitals in London and Southeast England. J Clin Microbiol and ceftazidime in combination with tobramycin, rifampin, or
2006;44:3623−7. ciprofloxacin against Burkholderia cepacia isolates from patients with
[18] Bogaerts P, Naas T, Wybo I, et al. Outbreak of infection cystic fibrosis. Antimicrob Agents Chemother 1999;43:213−7.
by carbapenem-resistant Acinetobacter baumannii producing the [38] Cheng NC, Hsueh PR, Liu YC, et al. In vitro activities of
carbapenemase OXA-58 in Belgium. J Clin Microbiol 2006;44: tigecycline, ertapenem, isepamicin, and other antimicrobial agents
4189−92. against clinically isolated organisms in Taiwan. Microb Drug Resist
[19] Turton JF, Kaufmann ME, Warner M, et al. A prevalent, multiresistant 2005;11:330−41.
clone of Acinetobacter baumannii in Southeast England. J Hosp Infect [39] Nseir S, Di Pompeo C, Brisson H, et al. Intensive care unit-acquired
2004;58:170−9. Stenotrophomonas maltophilia: incidence, risk factors, and outcome.
[20] Manuel RJ, Shin GY, Farrag N, Holliman R. Endemic carbapenem- Crit Care 2006;10:R143.
resistant Acinetobacter baumannii in a London hospital. J Antimicrob [40] Friedman ND, Korman TM, Fairley CK, Franklin JC, Spelman DW.
Chemother 2003;52:141−2. Bacteraemia due to Stenotrophomonas maltophilia: an analysis of 45
[21] Van Looveren M, Goossens H. Antimicrobial resistance of episodes. J Infect 2002;45:47−53.
Acinetobacter spp. in Europe. Clin Microbiol Infect 2004;10:684– [41] Pathmanathan A, Waterer GW. Significance of positive
704. Stenotrophomonas maltophilia culture in acute respiratory tract
[22] Souli M, Kontopidou FV, Koratzanis E, et al. In vitro activity of infection. Eur Respir J 2005;25:911−4.
tigecycline against multiple-drug-resistant, including pan-resistant, [42] British Society for Antimicrobial Chemotherapy. Stenotrophomonas
gram-negative and gram-positive clinical isolates from Greek maltophilia guideline. BSAC; 2005. http://www.bsac.org.uk/content_
hospitals. Antimicrob Agents Chemother 2006;50:3166−9. display.cfm?cit_id=258 [accessed 7 February 2007].
[23] Livermore DM, Woodford N, The beta-lactamase threat [43] Meyer E, Schwab F, Gastmeier P, Ruden H, Daschner FD. Is
in Enterobacteriaceae, Pseudomonas and Acinetobacter. Trends the prevalence of Stenotrophomonas maltophilia isolation and
Microbiol 2006;14:413−20. nosocomial infection increasing in intensive care units? Eur J Clin
[24] Urban C, Segal-Maurer S, Rahal JJ. Considerations in control Microbiol Infect Dis 2006;25:711−4.
and treatment of nosocomial infections due to multidrug-resistant [44] Betriu C, Rodriguez-Avial I, Sanchez BA, Gomez M, Alvarez J,
Picazo JJ. In vitro activities of tigecycline (GAR-936) against recently
Acinetobacter baumannii. Clin Infect Dis 2003;36:1268−74.
isolated clinical bacteria in Spain. Antimicrob Agents Chemother
[25] Cisneros JM, Rodriguez-Bano J, Fernandez-Cuenca F, et al. Risk-
2002;46:892−5.
factors for the acquisition of imipenem-resistant Acinetobacter
[45] Betriu C, Rodriguez-Avial I, Sanchez BA, Gomez M, Picazo JJ.
baumannii in Spain: a nationwide study. Clin Microbiol Infect
Comparative in vitro activities of tigecycline (GAR-936) and
2005;11:874−9.
other antimicrobial agents against Stenotrophomonas maltophilia. J
[26] Garcia-Garmendia JL, Ortiz-Leyba C, Garnacho-Montero J, et al.
Antimicrob Chemother 2002;50:758−9.
Risk factors for Acinetobacter baumannii nosocomial bacteremia in
[46] Bonfiglio G, Cascone C, Azzarelli C, Cafiso V, Marchetti F,
critically ill patients: a cohort study. Clin Infect Dis 2001;33:939−46.
Stefani S. Levofloxacin in vitro activity and time-kill evaluation
[27] del Mar Tomas M, Cartelle M, Pertega S, et al. Hospital outbreak
of Stenotrophomonas maltophilia clinical isolates. J Antimicrob
caused by a carbapenem-resistant strain of Acinetobacter baumannii:
Chemother 2000;45:115−7.
patient prognosis and risk-factors for colonisation and infection. Clin
[47] San Gabriel P, Zhou J, Tabibi S, Chen Y, Trauzzi M, Saiman L.
Microbiol Infect 2005;11:540−6.
Antimicrobial susceptibility and synergy studies of Stenotrophomonas
[28] Chen HP, Chen TL, Lai CH, et al. Predictors of mortality in
maltophilia isolates from patients with cystic fibrosis. Antimicrob
Acinetobacter baumannii bacteremia. J Microbiol Immunol Infect Agents Chemother 2004;48:168−71.
2005;38:127−36. [48] Fritsche TR, Sader HS, Stilwell MG, Dowzicky MJ, Jones RN.
[29] Robenshtok E, Paul M, Leibovici L, et al. The significance of Antimicrobial activity of tigecycline tested against organisms causing
Acinetobacter baumannii bacteraemia compared with Klebsiella community-acquired respiratory tract infection and nosocomial
pneumoniae bacteraemia: risk factors and outcomes. J Hosp Infect pneumonia. Diagn Microbiol Infect Dis 2005;52:187−93.
2006;64:282−7. [49] King A. Recommendations for susceptibility tests on fastidious
[30] Festini F, Buzzetti R, Bassi C, et al. Isolation measures for prevention organisms and those requiring special handling. J Antimicrob
of infection with respiratory pathogens in cystic fibrosis: a systematic Chemother 2001;48(Suppl. 1):77−80. Erratum: 2002;49:1050.
review. J Hosp Infect 2006;64:1−6. [50] Edson RS, Terrell CL. The aminoglycosides. Mayo Clin Proc 1999;
[31] Mahenthiralingam E, Urban TA, Goldberg JB. The multifarious, 74:519−28.
multireplicon Burkholderia cepacia complex. Nat Rev Microbiol [51] Poole K. Aminoglycoside resistance in Pseudomonas aeruginosa.
2005;3:144−56. Antimicrob Agents Chemother 2005;49:479−87.
[32] Jones AM, Dodd ME, Govan JR, et al. Burkholderia cenocepacia [52] Valdezate S, Vindel A, Loza E, Baquero F, Canton R. Antimicrobial
and Burkholderia multivorans: influence on survival in cystic fibrosis. susceptibilities of unique Stenotrophomonas maltophilia clinical
Thorax 2004;59:948−51. strains. Antimicrob Agents Chemother 2001;45:1581−4.
[33] De Soyza A, Archer L, Wardle J, et al. Pulmonary transplantation [53] Tsiodras S, Pittet D, Carmeli Y, Eliopoulos G, Boucher H, Harbarth S.
for cystic fibrosis: pre-transplant recipient characteristics in patients Clinical implications of Stenotrophomonas maltophilia resistant to
dying of peri-operative sepsis. J Heart Lung Transplant 2003;22: trimethoprim–sulfamethoxazole: a study of 69 patients at 2 university
764−9. hospitals. Scand J Infect Dis 2000;32:651−6.
[34] LiPuma JJ. Burkholderia cepacia complex: a contraindication to lung [54] Landman D, Bratu S, Alam M, Quale J. Citywide emergence
transplantation in cystic fibrosis? Transpl Infect Dis 2001;3:149−60. of Pseudomonas aeruginosa strains with reduced susceptibility to
[35] Thamlikitkul V, Trakulsomboon S. In vitro activity of tigecycline polymyxin B. J Antimicrob Chemother 2005;55:954−7.
against Burkholderia pseudomallei and Burkholderia thailandensis. [55] Garnacho-Montero J, Ortiz-Leyba C, Jimenez-Jimenez FJ, et al.
Antimicrob Agents Chemother 2006;50:1555−7. Treatment of multidrug-resistant Acinetobacter baumannii ventilator-
[36] Vonberg RP, Gastmeier P. Hospital-acquired infections related to associated pneumonia (VAP) with intravenous colistin: a comparison
contaminated substances. J Hosp Infect 2007;65:15−23. with imipenem-susceptible VAP. Clin Infect Dis 2003;36:1111−8.
D.A. Enoch et al. / International Journal of Antimicrobial Agents 29 Suppl. 3 (2007) S33–S41 S41

[56] Levin AS, Barone AA, Penco J, et al. Intravenous colistin as therapy [74] Pachon-Ibanez ME, Jimenez-Mejias ME, Pichardo C, Llanos AC,
for nosocomial infections caused by multidrug-resistant Pseudomonas Pachon J. Activity of tigecycline (GAR-936) against Acinetobacter
aeruginosa and Acinetobacter baumannii. Clin Infect Dis 1999;28: baumannii strains, including those resistant to imipenem. Antimicrob
1008−11. Agents Chemother 2004;48:4479−81.
[57] Linden PK, Kusne S, Coley K, Fontes P, Kramer DJ, Paterson D. [75] Seifert H, Stefanik D, Wisplinghoff H. Comparative in vitro
Use of parenteral colistin for the treatment of serious infection due activities of tigecycline and 11 other antimicrobial agents against
to antimicrobial-resistant Pseudomonas aeruginosa. Clin Infect Dis 215 epidemiologically defined multidrug-resistant Acinetobacter
2003;37:e154−60. baumannii isolates. J Antimicrob Chemother 2006;58:1099–100.
[58] Markou N, Apostolakos H, Koumoudiou C, et al. Intravenous colistin [76] Milatovic D, Schmitz FJ, Verhoef J, Fluit AC. Activities of
in the treatment of sepsis from multiresistant Gram-negative bacilli
the glycylcycline tigecycline (GAR-936) against 1,924 recent
in critically ill patients. Crit Care 2003;7:R78−83.
European clinical bacterial isolates. Antimicrob Agents Chemother
[59] Michalopoulos AS, Tsiodras S, Rellos K, Mentzelopoulos S,
2003;47:400−4.
Falagas ME. Colistin treatment in patients with ICU-acquired
[77] Sader HS, Jones RN, Stilwell MG, Dowzicky MJ, Fritsche TR.
infections caused by multiresistant Gram-negative bacteria: the
renaissance of an old antibiotic. Clin Microbiol Infect 2005;11: Tigecycline activity tested against 26,474 bloodstream infection
115−21. isolates: a collection from 6 continents. Diagn Microbiol Infect Dis
[60] Reina R, Estenssoro E, Saenz G, et al. Safety and efficacy of colistin 2005;52:181−6.
in Acinetobacter and Pseudomonas infections: a prospective cohort [78] Jones RN. Disk diffusion susceptibility test development for the
study. Intensive Care Med 2005;31:1058−65. new glycylcycline, GAR-936. Diagn Microbiol Infect Dis 1999;35:
[61] Jones RN, Deshpande LM, Bell JM, et al. Evaluation of 249−52.
the contemporary occurrence rates of metallo-beta-lactamases in [79] Yoon J, Urban C, Terzian C, Mariano N, Rahal JJ. In vitro double and
multidrug-resistant Gram-negative bacilli in Japan: report from the triple synergistic activities of polymyxin B, imipenem, and rifampin
SENTRY Antimicrobial Surveillance Program (1998–2002). Diagn against multidrug-resistant Acinetobacter baumannii. Antimicrob
Microbiol Infect Dis 2004;49:289−94. Agents Chemother 2004;48:753−7.
[62] Lekkas A, Gyi KM, Hodson ME. Temocillin in the treatment of [80] Tascini C, Menichetti F, Bozza S, Del Favero A, Bistoni F. Evaluation
Burkholderia cepacia infection in cystic fibrosis. J Cyst Fibros 2006; of the activities of two-drug combinations of rifampicin, polymyxin
5:121−4. B and ampicillin/sulbactam against Acinetobacter baumannii. J
[63] Andes DR, Craig WA. Cephalosporins. In: Mandell GL, Bennett Antimicrob Chemother 1998;42:270−1.
JE, Dolin R, editors. Principles and practice of infectious diseases. [81] Hogg GM, Barr JG, Webb CH. In-vitro activity of the combination
Philadelphia, PA: Churchill Livingstone; 2005. p. 20−1. of colistin and rifampicin against multidrug-resistant strains of
[64] Li XZ, Nikaido H, Poole K. Role of mexA–mexB–oprM in antibiotic Acinetobacter baumannii. J Antimicrob Chemother 1998;41:494−5.
efflux in Pseudomonas aeruginosa. Antimicrob Agents Chemother
[82] Taccone FS, Rodriguez-Villalobos H, De Backer D, et al. Successful
1995;39:1948−53.
treatment of septic shock due to pan-resistant Acinetobacter
[65] Livermore DM, Sefton AM, Scott GM. Properties and potential of
baumannii using combined antimicrobial therapy including
ertapenem. J Antimicrob Chemother 2003;52:331−44.
tigecycline. Eur J Clin Microbiol Infect Dis 2006;25:257−60.
[66] Kohler T, Michea-Hamzehpour M, Epp SF, Pechere JC. Carbapenem
activities against Pseudomonas aeruginosa: respective contributions [83] Fish DN, Choi MK, Jung R. Synergic activity of cephalosporins plus
of OprD and efflux systems. Antimicrob Agents Chemother 1999; fluoroquinolones against Pseudomonas aeruginosa with resistance to
43:424−7. one or both drugs. J Antimicrob Chemother 2002;50:1045−9.
[67] Mushtaq S, Ge Y, Livermore DM. Comparative activities [84] Gunderson BW, Ibrahim KH, Hovde LB, Fromm TL, Reed MD,
of doripenem versus isolates, mutants, and transconjugants of Rotschafer JC. Synergistic activity of colistin and ceftazidime
Enterobacteriaceae and Acinetobacter spp. with characterized beta- against multiantibiotic-resistant Pseudomonas aeruginosa in an
lactamases. Antimicrob Agents Chemother 2004;48:1313−9. in vitro pharmacodynamic model. Antimicrob Agents Chemother
[68] Howe RA, Wilson MP, Walsh TR, Millar MR. Susceptibility testing 2003;47:905−9.
of Stenotrophomonas maltophilia to carbapenems. J Antimicrob [85] Saiman L, Chen Y, Gabriel PS, Knirsch C. Synergistic activities of
Chemother 1997;40:13−7. macrolide antibiotics against Pseudomonas aeruginosa, Burkholderia
[69] Jones RN, Huynh HK, Biedenbach DJ. Activities of doripenem cepacia, Stenotrophomonas maltophilia, and Alcaligenes xylosoxi-
(S-4661) against drug-resistant clinical pathogens. Antimicrob Agents dans isolated from patients with cystic fibrosis. Antimicrob Agents
Chemother 2004;48:3136−40. Chemother 2002;46:1105−7.
[70] Ruiz J. Mechanisms of resistance to quinolones: target alterations, [86] Sobieszczyk ME, Furuya EY, Hay CM, et al. Combination therapy
decreased accumulation and DNA gyrase protection. J Antimicrob with polymyxin B for the treatment of multidrug-resistant Gram-
Chemother 2003;51:1109−17. negative respiratory tract infections. J Antimicrob Chemother 2004;
[71] Dean CR, Visalli MA, Projan SJ, Sum PE, Bradford PA. Efflux- 54:566−9.
mediated resistance to tigecycline (GAR-936) in Pseudomonas
[87] Saballs M, Pujol M, Tubau F, et al. Rifampicin/imipenem combination
aeruginosa PA01. Antimicrob Agents Chemother 2003;47:972−8.
in the treatment of carbapenem-resistant Acinetobacter baumannii
[72] Visalli MA, Murphy E, Projan SJ, Bradford PA. AcrAB multidrug
infections. J Antimicrob Chemother 2006;58:697–700.
efflux pump is associated with reduced levels of susceptibility
[88] Peleg AY, Potoski BA, Rea R, et al. Acinetobacter baumannii
to tigecycline (GAR-936) in Proteus mirabilis. Antimicrob Agents
Chemother 2003;47:665−9. bloodstream infection while receiving tigecycline: a cautionary report.
[73] Fritsche TR, Sader HS, Stilwell MG, Dowzicky MJ, Jones RN. J Antimicrob Chemother 2007;59:128−31.
Potency and spectrum of tigecycline tested against an international [89] Talbot GH, Bradley J, Edwards Jr JE, Gilbert D, Scheld M, Bartlett JG.
collection of bacterial pathogens associated with skin and soft tissue Bad bugs need drugs: an update on the development pipeline from
infections (2000–2004). Diagn Microbiol Infect Dis 2005;52:195– the Antimicrobial Availability Task Force of the Infectious Diseases
201. Society of America. Clin Infect Dis 2006;42:657−68.

You might also like