Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Send Orders of Reprints at reprints@benthamscience.

org
Current Pharmaceutical Design, 2013, 19, 209-222 209

Mechanisms of -lactam Resistance Among Pseudomonas aeruginosa

Daniel J. Wolter1 and Philip D. Lister2,*

1
Department of Pediatrics, University of Washington, Seattle, WA; 2Department of Medical Microbiology and Immunology, Creigh-
ton University School of Medicine, 2500 California Plaza, Omaha, NE 68178, USA

Abstract: Treatment of serious P. aeruginosa infections becomes more challenging with each passing year. As this pathogen acquires
more transferrable resistance mechanisms and continues to rapidly adapt and emerge resistant during the course of antimicrobial therapy,
we face the growing threat of pan-resistance. This review has focused on those mechanisms that directly impact the future of -lactam
antibiotics, including the production of -lactamases, porin-mediated resistance, and/or the overexpression of RND efflux pumps. With
the pipeline of new anti-pseudomonal agents diminishing, it is essential that novel therapeutic strategies be explored. These include tar-
geting biofilm formation and maintenance, virulence factors, and resistance mechanisms. Furthermore, we must continue to search for ef-
fective antibacterial combinations to not only prevent further emergence of resistance but also treat resistant strains already in the envi-
ronment.

Keywords: Pseudomonas aeruginosa, -lactam, resistance.

INTRODUCTION For the subsets of nosocomial pneumonia, health-care associated


Antibacterial resistance is an increasing threat to the treatment pneumonia, and ventilator associated pneumonia, P. aeruginosa is
of gram-positive and gram-negative pathogens. Among the gram- the second most common pathogen identified [17, 18]. In addition,
negatives, P. aeruginosa has historically been one of our most dif- P. aeruginosa is a leading cause of infections among patients in the
ficult therapeutic challenges. This pathogen is not only armed with intensive care unit [11, 12].
an impressive arsenal of chromosomally-encoded resistance Selection of appropriate antibacterial therapy is essential to
mechanisms, but has the propensity to acquire a wide range of re- optimizing the treatment of serious infections with P. aeruginosa
sistance genes from its environment. [19, 20]. The initial challenge with P. aeruginosa centers on its
The focus of this review will be on the diversity of resistance intrinsic antibiotic resistance that is not observed in most other
mechanisms that threaten the clinical use of -lactams for P. aeru- gram-negative bacilli. This inherent resistance is due to low perme-
ginosa infections, including -lactamases, porin-mediated resis- ability of its outer membrane [21] and intrinsic basal expression of
tance, and efflux-mediated resistance. For some of these mecha- chromosomal resistance mechanisms [22]. Complicating selection
nisms, P. aeruginosa has the ability to alter production and/or activ- of appropriate therapy is the propensity for P. aeruginosa to de-
ity and emerge resistant during the course of treating a patient. Fur- velop resistance to multiple classes of antibacterial agents. Multi-
thermore, with the cooperative effects of multiple mechanisms, the drug resistant strains are isolated with alarming frequency, and
threat of pan--lactam resistant P. aeruginosa is a growing concern. these strains develop a multi-drug resistant phenotype through the
acquisition of resistance genes on mobile genetic elements and/or
CLINICAL SIGNIFICANCE AND THERAPEUTIC CHAL- through mutational events influencing the expression and/or activity
LENGE OF PSEUDOMONAS AERUGINOSA of chromosomal resistance mechanisms [22]. An even greater con-
P. aeruginosa is a ubiquitous microbial pathogen that can be cern is the propensity for P. aeruginosa to emerge resistant during
isolated from a wide variety of environmental sources including the course of therapy, leading to clinical failure and potentially
plants, animals, and humans. As a nosocomial threat, P. aeruginosa doubling the length of hospital stay and the cost of patient care [23].
has been isolated from respiratory therapy equipment, antiseptics, This review will focus specifically on mechanisms of -lactam
sinks, medicines, and hydrotherapy pools [1]. Community reser- resistance among P. aeruginosa. Figure 1 summarizes the general
voirs include swimming pools, humidifiers, hot tubs, and contact mechanisms by which P. aeruginosa expresses resistance to this
lens solution [2-4]. Although P. aeruginosa is not normally a part antibacterial class. -lactam antibiotics enter the periplasmic space
of the normal flora, a significant proportion of hospitalized patients of P. aeruginosa by direct penetration across the outer membrane or
can become colonized [1], especially if protective barriers of skin or through porins located in the outer membrane (Fig. 1A). Once in
mucous membranes have been breached or if the patient is im- the periplasmic space, -lactams exert their antibacterial effect by
munocompromised [3, 5] Patient populations at particular risk are binding to and inactivating the penicillin-binding proteins involved
those on mechanical ventilators, patients with catheters, surgical in cell wall biosynthesis. Although mutational changes impacting
patients, patients with severe burn wounds, and seriously ill patients the affinity of penicillin-binding proteins for -lactams may play a
in the intensive care unit [6-12]. In addition, patients for whom the role in resistance for some strains, the primary mechanisms of -
normal protective flora has been disrupted by antimicrobial therapy lactam resistance are 1) production of an inactivating enzyme (-
are also at increased risk for colonization [6, 13, 14]. lactamase), 2) porin-mediated resistance, and 3) efflux-pump medi-
Although P. aeruginosa can cause community-acquired infec- ated resistance (Fig. 1B).
tions, the majority of serious infections are nosocomial. According -LACTAMASES OF Pseudomonas aeruginosa
to the CDC National Nosocomial Infections Surveillance System
(NNISS), P. aeruginosa ranks fifth among nosocomial pathogens The production of drug-inactivating enzymes is the most com-
for overall frequency of isolation in U.S. hospitals [15, 16]. mon mechanism of -lactam resistance among gram-negative bac-
teria. Molecular and genetic analyses of clinical isolates have
*Address correspondence to this author at the Department of Medical Mi-
shown that P. aeruginosa can produce a diversity of -lactamases,
crobiology and Immunology, Creighton University School of Medicine, some of which are encoded by chromosomal genes and others on
USA; Tel: 402-280-1224; E-mail: plister@cnm.edu mobile genetic elements. Although the -lactamases produced by P.

1873-4286/13 $58.00+.00 © 2013 Bentham Science Publishers


210 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

A.

O O O

OM

PG

PS
PBP PBP PBP PBP

CM

B.

O O O

X OM

PG

O PS
O
PBP PBP PBP

CM

Fig. (1). Mechanisms of -lactam Resistance Among P. aeruginosa. Panel A: Interactions of -lactams with “wild-type” susceptible P. aeruginosa. -
lactam molecules penetrate into the periplasmic space either by passing directly through the outer membrane or entering through specific porins, e.g. OprD for
carbapenems ( ). Once they enter the periplasmic space, -lactams can interact with their target penicillin binding proteins (PBP) located on the outside of
the cytoplasmic membrane. Panel B: Mechanisms of resistance to -lactams. The primary mechanisms of -lactam resistance include the production of -

lactamases ( ), decrease or loss of OprD porin in the outer membrane, and overproduction of RND efflux pumps ( )

aeruginosa can vary with respect to their spectra and efficiency of than penicillin and its relative resistance to inactivation by clavu-
hydrolysis, these characteristics alone do not define the resistance lanate and tazobactam [24]. The AmpC of P. aeruginosa is natu-
profiles associated with different enzymes. Instead, -lactamase- rally an inducible enzyme. In the absence of an inducing -lactam,
associated resistance in P. aeruginosa is also dependent upon the wild-type strains of P. aeruginosa produce low basal levels of
efficiency of drug penetration, ability of this pathogen to minimize AmpC and may be susceptible to the anti-pseudomonal penicillins,
drug accumulation in the periplasmic space (porin-mediated resis- penicillin-inhibitor combinations, cephalosporins and carbapenems
tance and/or extrusion by efflux pumps), and the cooperative effects if no other resistance mechanisms are operative [25]. However, the
of different -lactamases within the same cell. Therefore, it is not increased production of AmpC in P. aeruginosa can cause resis-
surprising that the resistance profile associated with a -lactamase tance to virtually all -lactams, except to the carbapenems [22, 26].
in one strain of P. aeruginosa may differ from the resistance profile Although data have suggested that AmpC plays a role in the intrin-
associated with that same -lactamase in a different strain. With sic level of susceptibility of P. aeruginosa to carbapenems [27-30],
this in mind, the following sections will summarize the different overproduction of AmpC does not significantly decrease P. aerugi-
families of -lactamases that have been detected in clinical isolates nosa susceptibility to carbapenems [27, 30-33]. Resistance to the
of P. aeruginosa and the general resistance threat they provide carbapenems usually requires the cooperation of additional resis-
when produced by this pathogen. The -lactamases have been cate- tance mechanisms, e.g. efflux pump overproduction, outer mem-
gorized based on the recently updated classification of Bush and brane porin decrease, and/or co-production of additional carbap-
Jacoby [24], and are summarized in Table 1. enem-hydrolyzing enzymes [22].
Group 1 Chromosomal AmpC Cephalosporinase. AmpC is a One pathway to overproduction of AmpC is through the re-
serine-based -lactamase belonging to molecular class C and is versible induction of ampC expression during exposure with certain
characterized by its more efficient hydrolysis of cephalosporins -lactams (cephamycins and carbapenems) and the -lactamase
Antibacterial Resistant Pseudomonas aeruginosa Current Pharmaceutical Design, 2013, Vol. 19, No. 2 211

Table 1. -lactamases of P. aeruginosa

Functional Group Molecular Class Potential Resistance Impact Representative Enzymes

Group 1 C Penicillins Chromosomal AmpC Cephalosporinase


Penicillin-Inhibitor Combinations
Cephalosporins
Cephamycins
Monobactams

Group 1e C Penicillins Extended-Spectrum AmpC Cephalosporinase


Penicillin-Inhibitor Combinations
Cephalosporins
Cephamycins
Monobactams
Carbapenems

Group 2b A Penicillins TEM-1, TEM-2, SHV-1


Early cephalosporins

Group 2be A Penicillins TEM-4, TEM-21, TEM-24, TEM-42, TEM-116


Early cephalosporins
Extended-spectrum cephalosporins SHV-2, SHV-2ª, SHV-5, SHV-12
Monobactams
CTX-M-1, CTX-M-2, CTX-M-43

PER-1, PER-2

VEB-1, VEB-1a, VEB-1b, VEB-2

GES-1, GES-8, GES-9

BEL-1, BEL-2

Group 2c A Penicillins PSE-1, PSE-3, PSE-4, PSE-5


Ticarcillin-clavulanate
CARB-3, CARB-4

Group 2d D Penicillins OXA-1, OXA-2, OXA-3, OXA-4, OXA-5, OXA-


Penicillin-Inhibitor Combinations 6, OXA-10, OXA-13, OXA-20, OXA-46, OXA-
50, OXA-56,

LCR-1

Group 2de D Penicillins OXA-11, OXA-14 to OXA-19, OXA-28, OXA-31,


Penicillin-Inhibitor Combinations OXA-32, OXA-35, OXA-45, OXA-74, OXA-147,
OXA-161
Cephalosporins
Monobactams

Group 2df D Penicillins OXA-40, OXA-50-type


Penicillin-Inhibitor Combinations
Narrow-spectrum Cephalosporins
Carbapenems
212 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

(Table 1) Contd....

Functional Group Molecular Class Potential Resistance Impact Representative Enzymes

Group 2f A Penicillins KPC


Penicillin-Inhibitor Combinations
Cephalosporins GES-2, GES-5
Carbapenems

Group 3 B Penicillins IMP-1, IMP-2, IMP-4, IMP-6, IMP-7, IMP-9,


Penicillin-inhibitor combinations IMP-10, IMP-11, IMP-13, IMP-14, IMP-15, IMP-
16, IMP-18, IMP-22,
Cephalosporins
Cephamycins
VIM-1 to VIM-11, VIM-13, VIM-15 to VIM-18,
Carbapenems

SPM-1

GIM-1

inhibitor clavulanate [34-38]. Although several components of the (65%) overexpressed an extended-spectrum AmpC cepha-
ampC regulatory system have been identified and characterized, the losporinase [55].
precise pathway of induction remains unclear [22]. Induction of Group 2b -lactamases. Group 2b -lactamases are serine-
ampC expression provides P. aeruginosa with resistance to ce- based enzymes from molecular class A and include the common
foxitin, but alone does not provide resistance to the carbapenems. plasmid-encoded enzymes TEM-1, TEM-2, and SHV-1 [24]. These
The clinical significance of clavunate’s induction of ampC is com- -lactamases were first found in members of the Enterobacteriaceae
plex. Due to induction by clavulanate, Enterobacteriaceae that pro- [56, 57], and later identified in strains of P. aeruginosa [58]. Group
duce an inducible ampC are usually more susceptible to ticarcillin 2b -lactamases most readily hydrolyze the penicillins and the early
than ticarcillin-clavulanate [39]. By contrast, MICs of ticarcillin- cephalosporin antibiotics.
clavulanate against P. aeruginosa remain similar to those of ticar-
cillin alone despite the induction of ampC [36]. The reason for this Group 2be Extended-Spectrum -lactamases. Group 2be in-
discrepancy appears to relate to the concentration of clavulanate cludes a wide variety of serine-based extended-spectrum -
used in CLSI-recommended susceptibility assays (2 μg/ml), a con- lactamases (ESBLs) from molecular class A. The first ESBLs char-
centration which is too low to induce sufficient levels of AmpC that acterized were variants of the Group 2b SHV-1, TEM-1, and TEM-
would impact the potency of ticarcillin in the combination [36]. 2 -lactamases and were discovered in members of the Enterobacte-
However, pharmacodynamic studies have demonstrated that the riaceae [59-61]. These ESBLs are able to hydrolyze the penicillins
level of induction achieved with pharmacokinetically-relevant con- and early cephalosporins like the Group 2b enzymes, but also dem-
centrations of clavulanate is sufficient to negatively impact the onstrate an extension of their hydrolytic capabilities to include ex-
antibacterial activity of the combination [36]. tended-spectrum oxyimino-cephalosporins (ceftazidime and cefo-
taxime), and the monobactam aztreonam [62, 63].
A more serious therapeutic threat is when the regulation of
ampC is lost through derepression, defined as constitutive high- ESBLs have also been identified in P. aeruginosa. The first
level ampC expression. As a result, P. aeruginosa becomes resis- TEM-derived ESBL reported in P. aeruginosa was TEM-42 from a
tant to virtually all -lactams. The pathway to derepression of clinical isolate in France [64]. Since that first report, only four other
ampC usually involves genetic mutations that alter proteins respon- TEM-derived ESBLs have been described, including TEM-4, TEM-
sible for regulating ampC expression [22], and these mutational 21, TEM-24, and TEM-116 [65-68]. In addition, four SHV-derived
events can lead to the emergence of resistance during therapy. ESBLs (SHV-2, SHV-2a, SHV-5, and SHV-12) have been identi-
Emergence of resistance due to ampC derepression has been re- fied in clinical isolates of P. aeruginosa [69-72].
ported in up to 56% of patients treated with anti-pseudomonal peni- The CTX-M-family of -lactamases represents a group of en-
cillins, penicillin-inhibitor combinations, extended-spectrum cepha- zymes that are not closely related to either the TEM- or SHV-
losporins, and aztreonam [40-48], and most commonly occurs dur- derived ESBLs, but are classified as ESBLs based upon their hydro-
ing treatment of infections outside the urinary tract and in patients lytic profiles [73]. These enzymes share homology with the chro-
with cystic fibrosis and neutropenia. mosomal AmpC of Kluyvera ascorbata [74]. To date, three CTX-
Group 1e Extended-Spectrum AmpC Cephalosporinases. Over- M-family ESBLs have been identified in P. aeruginosa, and include
expression of ampC alone does not always impact susceptibility of CTX-M-1, CTX-M-2, and CTX-M-43 [65, 75, 76].
P. aeruginosa to the carbapenems. However, recently described Additional Group 2be ESBLs that show limited homology with
mutational variants of AmpC (extended-spectrum AmpC) have TEM- or SHV-derived ESBLs include the PER-type, VEB-type,
been characterized that can directly influence carbapenem suscepti- GES-type, BEL-type, and PME-1 -lactamases. PER-1 was first
bility. These AmpC variants were first described in Enterobacte- described in 1993 in a strain of P. aeruginosa from France [77]. A
riaceae [49-53], and more recently among P. aeruginosa [54]. Al- closely related enzyme, PER-2, has also been identified in P. aeru-
though these enzymes can exhibit increased hydrolytic activity ginosa [75]. The first VEB-type ESBL found in P. aeruginosa was
against cephalosporins and imipenem, overproduction of extended- VEB-1, also from a clinical isolate in France [78]. Subsequently,
spectrum AmpCs seems to be a requirement for carbapenem resis- three other VEB-like ESBLs (VEB-1a, VEB-1b, and VEB-2) have
tance [54]. Characterization of 32 carbapenem-intermediate and – been identified in P. aeruginosa and share > 99% amino acid se-
resistant P. aeruginosa isolates from France demonstrated that 21 quence homology with VEB-1 [79, 80]. The GES-1 ESBL was
Antibacterial Resistant Pseudomonas aeruginosa Current Pharmaceutical Design, 2013, Vol. 19, No. 2 213

discovered in a strain of K. pneumoniae from French Guiana [81], enems, they do not efficiently hydrolyze these substrates and their
and then subsequently in P. aeruginosa [82]. Extended-spectrum - clinical significance is difficult to evaluate [95, 99].
lactamases GES-8, GES-9, and GES-13 have also been identified in Group 2f Carbapenemases: The Group 2f carbapenemases are
isolates of P. aeruginosa [83-85]. The extended-spectrum - serine-based -lactamases belonging to molecular class A [24,
lactamase BEL-1 was discovered in 2005 in a clinical isolate of P. 100]. In addition to the carbapenems, these -lactamases are capa-
aeruginosa from Belgium [86], and more recently, BEL-2 was ble of hydrolyzing the penicillins, cephalosporins, and aztreonam,
found in another P. aeruginosa isolate from Belgium [87]. The but they are inhibited by tazobactam and clavulanic acid [24, 99].
most recent ESBL discovered, PME-1 (Pseudomonas aeruginosa Although some Group 2f enzymes are encoded by chromosomal
ESBL 1) was described in a clinical isolate of P. aeruginosa from genes (e.g. SME), the class A carbapenemases of clinical impor-
Pennsylvania and shares 50% amino acid identity with the L2 - tance to P. aeruginosa are carried on plasmids.
lactamase of Stenotrophomonas maltophilia [88].
The first family includes the GES -lactamases, of which GES-
Group 2c Penicillinases: The Group 2c enzymes encompass a 1 was discovered as an ESBL in an isolate of K. pneumonia [81].
relatively small group of serine-based penicillinases from molecular Two variants, GES-2 and GES-5, have been identified among P.
class A [24]. Group 2c enzymes identified in P. aeruginosa include aeruginosa clinical isolates and nosocomial outbreaks from differ-
PSE-1, PSE-3, PSE-4, PSE-5, CARB-3, and CARB-4. Although ent geographic regions [101-105]. Although they were first consid-
these enzymes are inhibited by clavulanate, they hydrolyze ticarcil- ered ESBLs based on their hydrolytic profiles, these enzymes dem-
lin with such efficiency that they can provide P. aeruginosa with onstrate an extension of their hydrolytic activity to include
resistance to ticarcillin-clavulanate [89-91]. In contrast, strains of P. imipenem [99, 100, 104].
aeruginosa that produce PSE-type enzymes are more susceptible to
piperacillin-tazobactam, most likely due to the decreased hydrolysis The second family includes the Klebsiella pneumoniae carbap-
of piperacillin allowing tazobactam to be a more effective inhibitor enemases (KPCs), which were discovered in and most commonly
[90]. found among clinical isolates of K. pneumonia and other members
of the Enterobacteriaceae [99, 100]. Currently, 12 different variants
Group 2d OXA-type Penicillinases: The Group 2d -lactamases of KPCs (KPC-2 through KPC-13) have been identified. The first
are molecular class D enzymes class A enzymes with respect to the KPC-producing P. aeruginosa were reported among clinical iso-
active site serine, but they are only weakly inhibited by clavulanate lates in Colombia, and these isolates produced KPC-2 [106]. Sub-
and tazobactam [24]. The characteristics and clinical importance of sequently, KPC-2 has been identified among isolates of P. aerugi-
these enzymes have been extensively reviewed by Poirel et al. [92]. nosa from Puerto Rico [107], Trinidad and Tobago [108], China
Although these enzymes were first distinguished by the increased [109], and the United States [110], and KPC-5 was characterized in
hydrolytic activity against cloxacillin and oxacillin, thus the desig- a clinical isolate of P. aeruginosa from Puerto Rico [111]. Strains
nation OXA-type -lactamases, this phenotype does not define all of P. aeruginosa that produce KPCs can exhibit resistance to all -
enzymes in the family [92]. The OXA-family of enzymes repre- lactam antibiotics.
sents one of the largest and most diverse groups of -lactamases,
ranking second behind the TEM-family of enzymes [24]. Although Group 3 Metallo--lactamases: The Group 3 metallo--
different OXA-type enzymes have been isolated from a variety of lactamases are molecular class B enzymes that are characterized by
gram-negative bacterial species [92], many of the OXA-type - their requirement for a zinc ion in the active site [24, 112]. As a
lactamase variants can be found in isolates of Acinetobacter result of their broad-spectrum hydrolytic capacity and lack of inhi-
baumannii and P. aeruginosa [92]. Table 1 includes representative bition by tazobactam or clavulanate, metallo--lactamases can pro-
Group 2d OXA-type -lactamases detected in P. aeruginosa. Due vide resistance to the penicillins, inhibitor-penicillin combinations,
to their limited hydrolytic profiles and lack of inhibition by ta- and carbapenems [24, 112]. However, metallo--lactamases do not
zobactam and clavulanate, the Group 2d OXA-type -lactamases efficiently hydrolyze monobactams [24, 112].
primarily provide P. aeruginosa resistance to the anti-pseudomonal The history and clinical significance of metallo--lactamases
penicillins and inhibitor-penicillin combinations [92]. have been the focus of two recent reviews [99, 112]. Similar to the
Group 2de OXA-type Extended-Spectrum -lactamases: The class A carbapenemases described above, metallo--lactamases can
first published extended-spectrum OXA-type -lactamase was be encoded on both the chromosome and mobile genetic elements.
OXA-11 [93]. This enzyme is a variant of the narrow-spectrum The first transferable metallo--lactamase, IMP-1, was identified in
OXA-10 (formerly PSE-2) and was first identified in 1991 in a P. 1988 in a clinical isolate P. aeruginosa from Japan [113]. Since this
aeruginosa isolate from Turkey [93]. Over the past two decades, first report, dozens of transferrable metallo--lactamases have been
several other extended-spectrum OXA-family enzymes have been described globally among isolates of P. aeruginosa, and they be-
identified, primarily among clinical isolates of P. aeruginosa [92, long to four distinct families, namely IMP, VIM, GIM, and SPM
94] (Table 1). Although the hydrolytic profiles can vary substan- [112].
tially, the OXA-type ESBLs are characterized by their increased PORIN-MEDIATED RESISTANCE
hydrolysis of the oxyimino-cephalosporins and aztreonam and can
provide resistance to these drugs [92]. The outer membrane of P. aeruginosa is an effective antibacte-
rial barrier that is only 8% as permeable as the outer membrane of
Group 2df OXA-type Carbapenemases: The carbapenem- Escherichia coli [114]. To aid in the passage of nutrients and other
hydrolyzing OXA-type -lactamases have been identified most desired molecules into the cell, P. aeruginosa utilizes a collection
frequently among isolates of A. baumannii, and most are encoded of porin channels. Based on the completed sequence of the P. aeru-
by chromosomal genes [95]. To date, only a few OXA-type carbap- ginosa genome, 64 known or putative porins have been identified
enemases have been reported in P. aeruginosa (Table 1) [96, 97]. [114]. The following discussion will primarily focus on the well-
The OXA-40 carbapenemase described in P. aeruginosa was shown characterized association between the OprD porin and carbapenem
to be identical to the enzyme found in A. baumannii [97]. Although susceptibility of P. aeruginosa, but also touch upon the potential
the blaOXA-40 gene is found on similar plasmids in both P. aerugi- role of OprF in altering -lactam susceptibility.
nosa and A. baumannii [97], it was originally described as a chro-
mosomal gene in A. baumannii [98]. P. aeruginosa also encodes for OprD and Susceptibility of P. aeruginosa to Carbapenems
a chromosomal OXA-50 enzyme, of which sequence variants have
The OprD porin of P. aeruginosa is a substrate-specific porin
been described and characterized as carbapenemases [96]. Although
that facilitates the diffusion of basic amino acids, small peptides,
the OXA-type carbapenemases have a high affinity for carbap-
and carbapenems into the cell [115, 116]. Studies suggest that OprD
214 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

is the preferred portal of entry for carbapenems across the outer EFFLUX-MEDIATED RESISTANCE
membrane, as decreases or loss of OprD significantly decrease the -lactams that freely diffuse into the P. aeruginosa cell and
susceptibility of P. aeruginosa to available carbapenems [30, 116- escape inactivation by resident -lactamases may encounter another
119]. However, recent studies have described strains of P. aerugi- formidable obstacle in route to their target, extrusion by efflux
nosa that exhibit a discordance between OprD absence and suscep- pumps. The P. aeruginosa PAO1 genome possesses efflux encod-
tibility to carbapenems [33, 120], suggesting other pathways of ing genes belonging to five different superfamilies [135], but thus
carbapenem penetration. far, only efflux pumps belonging to the resistance-nodulation-
The complex pathways used by P. aeruginosa to regulate levels division (RND) family have been shown to participate in -lactam
of OprD in the outer membrane has been recently reviewed [22], resistance. The RND family represents the highest number of pre-
and include mechanisms that impact oprD expression and mecha- dicted pumps within the P. aeruginosa chromosome, 12 total in-
nisms that influence the production of a functional OprD channel. cluding two divalent metal cation transporters, which far exceeds
Regardless of the pathway, the impact of OprD-mediated resistance the RND pumps present on the E. coli chromosome (4 total) [135].
on the carbapenems can be analyzed by comparing carbapenem Of these, 3 RND efflux pumps are capable of exporting -lactams,
susceptibilities according to clinically-significant breakpoints (es- namely MexAB-OprM, MexCD-OprJ, and MexXY.
tablished by the CLSI) with OprD levels. The P. aeruginosa RND pumps are assembled with three sepa-
Although some investigators have concluded from the analysis rate components, a periplasmic membrane fusion protein (MFP), an
of non-controlled clinical isolates that meropenem is unique in hav- outer membrane factor (OMF), and a cytoplasmic membrane
ing alternative pathways of penetration [121], data from a more (RND) transporter, that form a channel spanning across both the
recent study of controlled isogenic “wild-type” and OprD-deficient inner and outer membranes (Fig. 2) [22]. Although the RND pump
mutant pairs demonstrated that loss of OprD impacted susceptibility creates a passage way from the extracellular milieu to the cyto-
to meropenem to a greater degree than imipenem or doripenem plasm from which substrates may be removed, the architecture of
[122]. When considering the clinical impact of OprD-mediated the pump also permits the recognition and export of compounds
resistance, loss of OprD alone is usually sufficient to push MICs of from the periplasmic space, the site of -lactam activity.
imipenem above the resistance breakpoint. This is not surprising Crystallographic examination of a RND component in E. coli,
since the MIC50 for imipenem against P. aeruginosa is already at 1 AcrB (homologous to MexB in P. aeruginosa) [136, 137], and
μg/ml [123, 124]. By comparison, wild-type P. aeruginosa are
generally  4-fold more susceptible to meropenem and doripenem
than they are to imipenem, and the impact of OprD-mediated resis- H+
tance on potency to these carbapenems does not always push MICs
above the susceptible breakpoint. Additional resistance mecha- O
nisms, e.g. efflux pump overproduction and/or -lactamase produc-
OM
tion, may be required to provide clinical resistance to meropenem
and doripenem. For example, studies using laboratory-derived mu-
tants have shown that a combination of OprD loss with either PG
MexAB-OprM overexpression or AmpC overproduction resulted in
significantly higher MICs to meropenem and doripenem, respec-
tively, compared to porin loss alone [125]. PS
O
Potential Decrease of -lactam Susceptibility Through OprF
Loss CM
The porin, OprF, is a major constituent of the P. aeruginosa
outer membrane and participates in the maintenance of cell shape
[126, 127], supports growth in low osmolarity conditions [128],
potentially serves as a general or nonspecific porin for the passage
of various compounds [128, 129], and has a proposed role in adher-
ence to epithelial cells [130]. Yoon et al. also demonstrated a role
for oprF in biofilm formation, anaerobic growth, and nitrate uptake MFP RND OMP
[131].
Similar to the effect of OprD loss on carbapenem susceptibili-
ties, loss of OprF has been suggested in a few studies to decrease
the susceptibility of P. aeruginosa to certain -lactams. In the first
study, Piddock et al. reported that a multi-drug resistant strain (in-
cluding resistance to several -lactams such as cefotaxime, ceftaz- Transcription
idime, and carbenicillin) of P. aeruginosa that lacked OprF was
isolated from a patient following therapy [132]. Restoration of
mfp rnd omp
OprF production conincided with reversion of -lactam suscepti-
bilities to pre-therapy levels [132]. Secondly, a laboratory-derived
OprF deficient strain of P. aeruginosa had modest increases in Fig. (2). Structure and function of RND efflux pumps in P. aeruginosa.
MICs to several -lactams including carbenicillin and cefotaxime RND pumps typically exist in a tripartite system consisting of an RND
[133]. Loss of OprF may have decreased -lactam susceptibility in cytoplasmic membrane transporter (RND), a membrane fusion protein
these studies by causing either: 1) diminished -lactam uptake di- (MFP), and outer membrane factor (OMF). The genes which encode the
rectly through the OprF channel or 2) altered permeability as the pump components are organized into operons, and following translation of
the polycistronic message, the pump components assemble into a channel
indirect result of defects in membrane architecture since OprF
that spans across the entire membrane. -lactams enter into the RND trans-
serves in anchoring the outer membrane to the peptidoglycan layer porter through openings known as vestibules and are exported through the
[128, 134]. Regardless, OprF may be another mechanism that can channel to the extracellular milieu using proton-motive force. Figure revised
influence -lactam susceptibilities. Additional studies are needed to and reprinted from Lister et al. [22] with permission from the American
address this possibility. Society of Microbiology.
Antibacterial Resistant Pseudomonas aeruginosa Current Pharmaceutical Design, 2013, Vol. 19, No. 2 215

protein modeling of the MexB component in P. aeruginosa [138] moter [155]. However, each promoter is occupied by regulatory
have shown them to exist as a homotrimeric complex with trans- proteins that repress the transcription of the efflux operon. MexR, a
membrane helices lodged into the cytoplasmic membrane (trans- regulatory protein of the MarR family [156], binds to cis-acting
membrane domain) and a periplasmic domain that forms a central sites that encompass the distal mexA promoter, and a trans-acting
cavity. A route from the periplasm to the central cavity exists repressor of the TetR family, NalD, binds to sequences encom-
through openings, referred to as vestibules, between monomers passsing the mexA proximal promoter [157].
along the surface of the cytoplasmic membrane [139]. -lactams in Loss of MexR-mediated regulation of the distal mexA promoter
the periplasm may enter into the RND transporter through these causes mexAB-oprM hyperexpression in mutants termed nalB-type
vestibules. Initial chimeric and mutational studies of E. coli and P. and nalC-type. In nalB-type strains, mutations within mexR can
aeruginosa pumps linked substrate specificity to the cytoplasmic either truncate the production of full length protein, potentially
RND transporter and suggest the location of substrate recognition diminish protein stability, or compromise MexR functionality by
occurs within the central cavity [140, 141]. However, more recent preventing protein dimerization or DNA binding [158, 159]. nalC-
studies have identified a binding pocket within the periplasmic type mutants retain a wild-type mexR, but mutations in another
domain that is connected to the vestibule through an additional gene, nalC (formerly PA3721), indirectly impacts the ability of
channel, and this pocket serves as the substrate recognition site MexR to bind to the distal mexA promoter. NalC, a member of the
[142, 143]. Once inside the binding pocket, RND transporters util- TetR/AcrR family, suppresses the expression of the PA3720-
ize proton-motive force to translocate substrates (e.g. -lactams) PA3719 (renamed ArmR), and inactivation of NalC results in
through the OMF to the extracellular environment. Addition of ArmR overexpression [160, 161]. ArmR is a 53-amino acid antirep-
compounds that interfere with the proton gradient, such as the pro- ressor that binds inside a hydrophobic cavity within the MexR di-
ton uncoupler carbonyl cyanide m-chlorophenylhydrazone (CCCP), mer and allosterically inhibits MexR from docking with the distal
prevents the efflux of substrates from within the cell [144]. mexA promoter [162]. nalC-type mutants overexpress mexAB-oprM
The efflux encoding genes within P. aeruginosa are organized at lower levels in comparison to nalB-type mutants, and as a result,
into operons on the bacterial chromosome (Fig. 2) [22]. mexAB- do not have as great of an impact on -lactam susceptibility than
oprM, mexCD-oprJ, and mexXY each possess a gene that codes for nalB-type mutants [163]. Hyperexpression of mexAB-oprM from
the MFP (mexA, mexC, and mexX) and RND transporter (mexB, the proximal mexA promoter occurs in nalD-type mutants as a re-
mexD, and mexY). The mexAB-oprM and mexCD-oprJ operons also sult of mutations within nalD. These mutations presumably inter-
contain a gene for the OMF (oprM and oprJ) whose product com- fere with the ability of NalD from binding to the operator sequence
pletes the formation of the tripartite efflux systems. However, an associated with the mexA-proximal promoter [157].
OMF encoding gene is absent from the mexXY operon. Therefore,
MexXY must associate with other outer membrane protein compo- MexCD-OprJ
nents to create a functional pump. OprM was shown to serve as the In comparison to MexAB-OprM, the MexCD-OprJ efflux
OMF for MexXY [145-147] and several other candidate OMFs pump has a more restricted substrate profile for the -lactams.
have been proposed including OpmB, OpmG, OpmH, and possibly MexCD-OprJ is capable of exporting the fourth generation cepha-
OpmI [146, 148]. The regulatory genes mexR, nfxB, and mexZ re- losporins (i.e. cefepime, cefpirome, and cefozopran) [164, 165].
side directly upstream of mexAB-oprM, mexCD-oprJ, and mexXY, Studies have suggested that additional -lactams, such as the car-
respectively, and are transcribed divergently from the operon [22]. bapenems meropenem and doripenem, may also be exported by
The products of these regulatory genes bind to operator sites within MexCD-OprJ [166, 167]. However, substrate specificities in these
the intergenic regions to control expression of the efflux operons studies were determined using isogenic mutants containing dele-
(discussed below). tions of other efflux systems. The possibility that these deletions
may have triggered changes in other mechanisms that could impact
MexAB-OprM drug susceptibility can not be ruled out. Nonetheless, overproduc-
MexAB-OprM, the first discovered P. aeruginosa RND pump, tion of MexCD-OprJ does not decrease the susceptibility of P.
contributes to both the intrinsic and adaptive resistance of P. aeru- aeruginosa mutants to these additional -lactams [164], but rather,
ginosa to the -lactam class of antibiotics. MexAB-OprM is able to only impacts the fourth generation cephalosporins. MexCD-OprJ
export the widest range of -lactams among the RND pumps such also differs from MexAB-OprM in that the pump is not detectable
as carbenicillin and ticarcillin (carboxypenicillins), piperacillin in wild-type cells [165, 168]. As such, MexCD-OprJ does not
(ureidopenicillin), aztreonam (monobactam), ceftazidime and cefo- contribute to intrinisic -lactam resistance [169, 170].
taxime (3rd generation cephalosporins), faropenem (penem), and Expression of the mexCD-oprJ operon is tightly controlled by
the carbapenems meropenem and panipenem (excluding imipenem the regulatory factor NfxB, a protein that displays similarity to the
and biapenem). A recent study showed that overproduction of LacI-GalR family [165]. NfxB binds to a cis-acting site within the
MexAB-OprM in a defined laboratory mutant led to slightly ele- nfxB-mexC intergenic region [171] and represses expression of the
vated MICs for doripenem compared to the parental P. aeruginosa efflux operon [165, 171]. Mutations within nfxB (e.g. base substitu-
strain suggesting the recognition and export of this drug at a low tions, deletions, IS element disruption) have been suggested to alle-
level [125]. -lactam inhibitors (e.g. clavulanate and cloxacillin) are viate repression of mexCD-oprJ leading to its overexpression in
also substrates for MexAB-OprM [149]. Intrinsic resistance to these mutants termed nfxB-type [165, 171-173]. Overproduction of
-lactams is attributed to the constitutive expression of mexAB- MexCD-OprJ has been shown to occur at two different levels in
oprM in wild-type cells [150]. Deletion of any one component mutants defined as type A and type B. Type A mutants produce
(mexA, mexB, or oprM) through genetic knockout renders P. aeru- lower amounts of MexCD-OprJ than type B mutants, and as a re-
ginosa hypersusceptible to the -lactams listed above [151, 152]. sult, have a smaller decrease in susceptibility to antibiotics exported
Expression of mexAB-oprM is growth-phase dependent in wild-type by this pump in comparison to type B mutants [164]. Regardless,
cells and is regulated by the N-Butyryl-L-homoserine lactone (C4- overproduction of MexCD-OprJ in both types of mutants is de-
HSL) quorum sensing autoinducer [153, 154]. pendent on alterations within NfxB [164].
Although expressed at sufficient levels to influence -lactam
susceptibility, transcription of the mexAB-oprM operon in wild-type MexXY
P. aeruginosa is not fully derepressed. Expression of mexAB-oprM The third RND efflux system responsible for -lactam resis-
is initiated from two separate promoters, a distal mexA promoter tance in P. aeruginosa is the MexXY pump. As mentioned above,
that overlaps with the mexR promoter and a proximal mexA pro- an OMF encoding gene is absent from the mexXY operon, but in-
216 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

stead, MexXY associates with OprM [145-147] and possibly other P. aeruginosa and have been analyzed quite extensively as a result
candidate OMFs [146, 148] to form a tripartite pump. MexXY has a of their impact on drug susceptibility. However, recent studies us-
similar substrate profile to the MexCD-OprJ pump with its ability ing more global screening methods (e.g. analysis of transposon
to export fourth generation cephalosporins [166, 174] but differs mutant libraries and proteomics) have indicated that additional
from MexCD-OprJ in that MexXY accommodates the removal of genetic determinants may participate in decreasing -lactam suscep-
the "fifth generation" cephalosporin ceftobiprole [175, 176]. mexXY tibility [210-212]. One such library, the Harvard PA14 nonredun-
expression in P. aeruginosa is inducible when cells are exposed to dant library, represents a collection of mutants in which nonessen-
ribosomal inhibitors (e.g. tetracycline, erythromycin, and gentami- tial genes have been inactivated by a single transposon insertion
cin) [177], but expression does not increase when challenged with [213]. This library has been used by two independent research
sub-inhibitory concentrations of cefepime [178]. Thus, MexXY groups to identify genes that directly cause -lactam resistance
most likely does not contribute to intrinsic -lactam resistance in when inactivated or indirectly by increasing the mutational fre-
wild-type cells. quency [210, 211].
Expression of mexXY is negatively regulated by MexZ whose Susceptibility testing of mutants in the PA14 library to various
gene is located directly upstream of the efflux operon. MexZ, a -lactams revealed novel genes responsible for altering -lactam
member of the TetR regulatory family, binds to an inverted repeat susceptibility. Many of these genes participate in the synthesis of
sequence within the mexZ-mexX intergenic region and blocks ac- the bacterial cell wall or lipopolysaccharide (LPS) including: galU
cess of the RNA polymerase to the putative mexXY promoter [179]. (PAO1 ortholog PA2023), a UDP-glucose pyrophosphorylase re-
Overexpression of mexXY has been associated with mutations in sponsible for synthesis of a complete LPS core, and wbpM (PAO1
mexZ or the mexZ-mexX intergenic region in mutants termed agrZ- ortholog PA3141), a UDP-N-acetylglucosamine 4,6-dehydratase
type [180]. However, P. aeruginosa strains hyperexpressing mexXY essential for the biosynthesis of B-band LPS. Inactivation of genes
but lacking mutations within these sites, called agrW-type mutants, within a large 17 ORF gene cluster (PA4996–PA5012) that are
have also been described [180]. mexXY overexpression in agrW- involved in the assembly of the core oligosaccharide component of
type mutants has recently been linked to a two-component regula- LPS were also identified by both research groups as causing de-
tory system, ParRS [181]. creased -lactam susceptibility [210, 211]. Alterations in cell wall
and LPS architecture are believed to decrease the penetration of -
Contribution of Efflux to -lactam Resistance in Clinical Iso- lactams into the periplasmic space. Various modifications in LPS
lates composition were previously shown to either decrease [214, 215] or
Numerous studies have detected the overexpression of an RND increase [216] -lactam susceptibility in P. aeruginosa supporting
efflux pump in -lactam resistant P. aeruginosa clinical isolates the role of LPS in antibiotic uptake.
suggesting their participation in the resistant phenotype. MexAB- Besides genes involved with cell wall and LPS synthesis,
OprM hyperexpressing clinical isolates have been observed for analysis of the PA14 library has identified additional genes with
each of the nal (B, C, and D)-type mutants and have often been diverse functions that alter -lactam susceptibility when deleted.
identified in studies examining the mechanism(s) associated with Inactivation of genes that participate in chemotaxis and biofilm
carbapenem resistance in P. aeruginosa [182-186]. mexXY overex- formation (wspE, a histidine kinase-response regulator and wspR, a
pressing agrZ- and agrW-type mutants from the clinical setting diguanylate cyclase), amino acid metabolism (aroB, a 3-
have also been encountered frequently [187-191]. MexCD-OprJ dehydroquinate synthetase), carbon compound catabolism and en-
overproducing clinical isolates have been described in the literature ergy metabolism (PA5192, a phosphoenolpyruvate carboxykinase),
[192-194] but are not as common as the other pump mutants, possi- and transcriptional regulation (PA0479, regulator of the LysR fam-
bly suggesting a disadvantage for P. aeruginosa that utilize this ily) all decreased susceptibility of P. aeruginosa to several -
resistance mechanism. Indeed, studies have shown that nfxB-type lactams [210, 211]. It is unknown at this time how the loss of these
mutants have diminished fitness and virulence traits [195-197]. genes influence -lactam susceptibility. However, these results
Overexpression of the RND efflux pumps typically reduces the illustrate the vast array of mechanisms capable of altering suscepti-
susceptibility (2-8-fold decrease in MIC) of P. aeruginosa to - bility of P. aeruginosa to -lactams.
lactams that are substrates for a particular pump, but clinical resis-
tance (according to CLSI breakpoints) often requires the involve- CONFLICT OF INTEREST
ment of an additional resistance mechanism. For example, nalB- The authors confirm that this article content has no conflicts of
type mutants have decreased susceptibility to meropenem, but full interest.
resistance occurs with a concomittant loss of OprD [117]. The
combination of OprD loss and MexAB-OprM overepxression has ACKNOWLEDGEMENT
been detected in several meropenem resistant clinical isolates [198- Declared none.
200]. Additional combinations of -lactam resistance mechanisms
have been reported including the simultaneous overexpression of REFERENCES
two efflux pumps. Several groups have detected the co- [1] Pollack M. Pseudomonas aeruginosa. In: Mandell GL, Bennett JE,
hyperexpression of MexAB-OprM and MexXY in resistant P. Dolin R, ed.^eds., Principles and Practice of Infectious Diseases.
aeruginosa [180, 201, 202]. Efflux pump overexpression has been Churchill Livingstone: New York 1995; pp 1980-2003.
discovered in isolates with concurrent chromosomal AmpC derep- [2] Pitt TL. Pseudomonas, Burkholderia, and Related Genera. In:
ression [203-206] or when harboring an acquired -lactamase (e.g. Duerden BI, ed.^eds., Microbiology and Microbial Infections.
OXA, GES) [174, 207]. Isolates have even been encountered con- Oxford University Press Inc: New York City 1998; pp. 1109-38.
taining at least three different -lactam resistance mechanisms [3] Pollack M. Pseudomonas aeruginosa. In: Mandell GL, Dolan R,
(OprD loss, pump hyperexpression, and -lactamase production) Bennett JE, ed.^eds., Principles and Practices of Infectious
Diseases. Churchill Livingstone: New York 1995; pp. 1820-2003.
[107, 208, 209]. The repeated identification of efflux overexpress- [4] Harris AA, Goodman L, Levin S. community-acquired
ing P. aeruginosa clinical isolates, either alone or in combination Pseudomonas aeruginosa pneumonia associated with the use of a
with other mechanisms, highlights their importance in -lactam home humidifier. West J Med 1984; 141: 521-3.
resistance. [5] Morrison AJ, Wenzel RP. Epidemiology of infections due to
Pseudomonas aeruginosa. Rev. Infect. Dis 1984; 6 (Suppl 3):
Pseudomonas aeruginosa -LACTAM RESISTOME S627-42.
The mechanisms discussed so far in this review are typically [6] Blanc DS, Petignat C, Janin B, Bille J, Fancioli P. Frequency and
considered as the predominant contributors to -lactam resistance in molecular diversity of Pseudomonas aeruginosa upon admission
Antibacterial Resistant Pseudomonas aeruginosa Current Pharmaceutical Design, 2013, Vol. 19, No. 2 217

and during hospitalization: a prospective epidemiologic study. Clin [29] Masuda N, Gotoh N, Ishii C, Sakagawa E, Ohya S, Nishino T.
Microbiol Infect 1998; 4: 242-7. Interplay between chromosomal -lactamase and the MexAB-
[7] Thuong M, Arvaniti K, Ruimy R, et al. Epidemiology of OprM efflux system in intrinsic resistance to -lactams in
Pseudomonas aeruginosa and risk factors for carriage acquisition Pseudomonas aeruginosa. Antimicrob Agents Chemother 1999;
in an intensive care unit. J Hosp Infect 2003; 53: 274-82. 43: 400-2.
[8] Valles J, Mariscal D, Cortes P, et al. Patterns of colonization by [30] Mushtaq S, Ge Y, Livermore DM. Doripenem versus Pseudomonas
Pseudomonas aeruginosa in intubated patients: a 3-year aeruginosa in vitro: activity against characterized isolates, mutants,
prospective study of 1607 isolates using pulsed-field gel and transconjugants and resistance selection potential.
electrophoresis with implications for prevention of ventilator- Antimicrobial Agents Chemotherap 2004; 48: 3086-92.
associated pneumonia. Intensive Care Med. 2004; 30: 1768-75. [31] Gates ML, Sanders CC, Goering RV, Sanders WE, Jr. Evidence for
[9] Ohara T, Itoh K. Significance of Pseudomonas aeruginosa multiple forms of type I chromosomal beta-lactamase in
colonization of the gastrointestinal tract. Intern. Med. 2003; 42: Pseudomonas aeruginosa. Antimicrob Agents Chemother 1986;
1072-6. 30: 453-7.
[10] Erol S, Altoparlak U, Akcay MN, Celebi F, Parlak M. Changes of [32] Sanders CC, Gates ML, Sanders WE, Jr. Heterogeneity of class I -
microbial flora and wound colonization in burned patients. Burns lactamase expression in clinical isolates of Pseudomonas
2004; 30: 357-61. aeruginosa. Antimicrobial Agents Chemotherap 1988; 32: 1893-5.
[11] Richards MJ, Edwards JR, Culver DH, Gaynes RP. Nosocomial [33] Wolter DJ, Hanson ND, Lister PD. AmpC and OprD are not
infections in medical intensive care units in the United States. involved in the mechanism of imipenem hypersusceptibility among
National Nosocomial Infections Surveillance System. Crit Care Pseudomonas aeruginosa isolates overexpressing the mexCD-oprJ
Med 1999; 27: 887-92. efflux pump. Antimicrob Agents Chemother 2005; 49: 4763-6.
[12] Spencer RC. Predominant pathogens found in the European [34] Labia R, Morand A, Peduzzi J. Timentin and beta-lactamases. J
Prevalence of Infection in Intensive Care Study. Eur J Clin antimicrobial chemotherapy 1986; 17 Suppl C: 17-26.
Microbiol Infect Dis 1996; 15: 281-5. [35] Lindberg F, Lindquist S, Normark S. Genetic basis of induction
[13] Bonten MJ, Bergmans DC, Speijer H, Stobberingh EE. and overproduction of chromosomal class I beta-lactamase in
Characteristics of polyclonal endemicity of Pseudomonas nonfastidious gram-negative bacilli. Reviews of infectious diseases
aeruginosa colonization in intensive care units. Implications for 1988; 10: 782-5.
infection control. Am. J. Crit. Care Med 1999; 160: 1212-9. [36] Lister PD, Gardner VM, Sanders CC. Clavulanate induces
[14] Takesue Y, Yokoyama T, Akagi S, et al. Changes in the intestinal expression of the Pseudomonas aeruginosa AmpC
flora after the administration of prophylactic antibiotics to patients cephalosporinase at physiologically relevant concentrations and
undergoing a gastrectomy. Surg. Today 2002; 32: 581-6. antagonizes the antibacterial activity of ticarcillin. Antimicrob
[15] . National Nosocomial Infections Surveillance (NNIS) System Agents Chemother 1999; 43: 882-9.
report, data summary from October 1986-April 1998, issued June [37] Stobberingh EE. Induction of chromosomal beta-lactamases by
1998. American J infection control 1998; 26: 522-33. different concentrations of clavulanic acid in combination with
[16] Emori TG, Gaynes RP. An overview of nosocomial infections, ticarcillin. J antimicrobial chemotherapy 1988; 21: 9-16.
including the role of the microbiology laboratory. Clin Microbiol [38] Weber DA, Sanders CC. Diverse potential of beta-lactamase
Rev 1993; 6: 428-42. inhibitors to induce class I enzymes. Antimicrobial Agents
[17] Kolleff MH, Shorr A, Tabak YP, Gupta V, Liu LZ, Johannes RS. Chemotherap 1990; 34: 156-8.
Epidemiology and outcomes of health-care-associated pneumonia: [39] Akova M, Yang Y, Livermore DM. Interactions of tazobactam and
results from a large US database of culture-positive pneumonia. clavulanate with inducibly- and constitutively-expressed Class I -
Chest 2005; 128: 3854-62. lactamases. J antimicrobial chemotherapy 1990; 25: 199-208.
[18] Gaynes R, Edwards JR. Overview of nosocomial infections caused [40] Eron LJ, Goldenberg RI, Poretz DM, Park CH. Piperacillin therapy
by gram-negative bacilli. Clin. Infect. Dis. 2005; 41: 848-54. for Pseudomonas infections. Southern medical journal 1983; 76:
[19] Micek ST, Lloyd AE, Ritchie DJ, Reichley RM, Fraser VJ, Kollef 859-62.
MH. Pseudomonas aeruginosa bloodstream infection: importance [41] Gribble MJ, Chow AW, Naiman SC, et al. Prospective randomized
of appropriate initial antimicrobial treatment. Antimicrob. Agents trial of piperacillin monotherapy versus carboxypenicillin-
Chemother. 2005; 49: 1306-11. aminoglycoside combination regimens in the empirical treatment of
[20] Bisbe J, Gatell JM, Puig J. Pseudomonas aeruginosa bacteremia: serious bacterial infections. Antimicrobial Agents Chemotherap
univariate and multivariate analyses of factors influencing the 1983; 24: 388-93.
prognosis in 133 episodes. Rev. Infect. Dis 1988; 10: 629-35. [42] Jimenez-Lucho VE, Saravolatz LD, Medeiros AA, Pohlod D.
[21] Hancock REW, Brinkman FS. Function of pseudomonas porins in Failure of therapy in pseudomonas endocarditis: selection of
uptake and efflux. Annu. Rev. Microbiol. 2002; 56: 17-38. resistant mutants. J infectious diseases 1986; 154: 64-8.
[22] Lister PD, Wolter DJ, Hanson ND. Antibacterial-resistant [43] Juan C, Macia MD, Gutierrez O, Vidal C, Perez JL, Oliver A.
Pseudomonas aeruginosa: clinical impact and complex regulation Molecular mechanisms of beta-lactam resistance mediated by
of chromosomally encoded resistance mechanisms. Clinical AmpC hyperproduction in Pseudomonas aeruginosa clinical
microbiology reviews 2009; 22: 582-610. strains. Antimicrobial Agents Chemotherap 2005; 49: 4733-8.
[23] Dimatatac EL, Alejandria MM, Montalban C, Pineda C, Ang C, [44] Letendre ED, Mantha R, Turgeon PL. Selection of resistance by
Delino R. Clinical outcomes and costs of care of antibiotic resistant piperacillin during Pseudomonas aeruginosa endocarditis. J
Pseudomonas aeruginosa infections. Philipp. J.Microbiol Infect. antimicrobial chemotherapy 1988; 22: 557-62.
Dis. 2003; 32: 159-67. [45] Masterton RG, Garner PJ, Harrison NA, Rainford DJ. Timentin
[24] Bush K, Jacoby GA. Updated functional classification of - resistance. Lancet 1987; 2: 975-6.
lactamases. Antimicrobial Agents Chemotherap 2010; 54: 969-76. [46] Scully BE, Ores CN, Prince AS, Neu HC. Treatment of lower
[25] Sanders CC, Sanders WE. Type I beta-lactamases of gram-negative respiratory tract infections due to Pseudomonas aeruginosa in
bacteria: interactions with beta-lactam antibiotics. J. Infect. Dis patients with cystic fibrosis. Reviews of infectious diseases 1985; 7
1986; 154: 792-800. Suppl 4: S669-74.
[26] Colom K, Fdz-Aranguiz A, Suinaga E, Cisterna R. Emergence of [47] Sanders CC, Sanders WE, Jr. Emergence of resistance during
resistance to -lactam agents in Pseudomonas aeruginosa with therapy with the newer beta-lactam antibiotics: role of inducible
group I -lactamases in Spain. Eur J Clin Microbiol Infect Dis beta-lactamases and implications for the future. Reviews of
1995; 14: 964-71. infectious diseases 1983; 5: 639-48.
[27] Livermore DM. Interplay of impermeability and chromosomal [48] Sanders WE, Jr., Sanders CC. Inducible beta-lactamases: clinical
beta-lactamase activity in imipenem-resistant Pseudomonas and epidemiologic implications for use of newer cephalosporins.
aeruginosa. Antimicrob Agents Chemother 1992; 36: 2046-8. Reviews of infectious diseases 1988; 10: 830-8.
[28] Livermore DM, Yang YJ. Beta-lactamase lability and inducer [49] Barnaud G, Labia R, Raskine L, Sanson-Le Pors MJ, Philippon A,
power of newer beta-lactam antibiotics in relation to their activity Arlet G. Extension of resistance to cefepime and cefpirome
against beta-lactamase-inducibility mutants of Pseudomonas associated to a six amino acid deletion in the H-10 helix of the
aeruginosa. J Infect Dis 1987; 155: 775-82. cephalosporinase of an Enterobacter cloacae clinical isolate. FEMS
microbiology letters 2001; 195: 185-90.
218 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

[50] Mammeri H, Galleni M, Nordmann P. Role of the Ser-287-Asn [70] Naas T, Philippon L, Poirel L, Ronco E, Nordmann P. An SHV-
replacement in the hydrolysis spectrum extension of AmpC beta- derived extended-spectrum -lactamase in Pseudomonas
lactamases in Escherichia coli. Antimicrobial Agents Chemotherap aeruginosa. Antimicrobial Agents Chemotherap 1999; 43: 1281-4.
2009; 53: 323-6. [71] al Naiemi NA, Duim B, Savelkoul PH, et al. Widespread transfer
[51] Mammeri H, Nordmann P, Berkani A, Eb F. Contribution of of resistance genes between bacterial species in an intensive care
extended-spectrum AmpC (ESAC) beta-lactamases to carbapenem unit: implications for hospital epidemiology. Journal of clinical
resistance in Escherichia coli. FEMS microbiology letters 2008; microbiology 2005; 43: 4862-4.
282: 238-40. [72] Poirel L, Lebessi E, Castro M, Fevre C, Foustoukou M, Nordmann
[52] Mammeri H, Poirel L, Bemer P, Drugeon H, Nordmann P. P. Nosocomial outbreak of extended-spectrum -lactamase SHV-5-
Resistance to cefepime and cefpirome due to a 4-amino-acid producing isolates of Pseudomonas aeruginosa in Athens, Greece.
deletion in the chromosome-encoded AmpC beta-lactamase of a Antimicrobial Agents Chemotherap 2004; 48: 2277-9.
Serratia marcescens clinical isolate. Antimicrobial Agents [73] Bonnet R. Growing group of extended-spectrum -lactamases: the
Chemotherap 2004; 48: 716-20. CTX-M enzymes. Antimicrobial Agents Chemotherap 2004; 48: 1-
[53] Matsumura N, Minami S, Mitsuhashi S. Sequences of homologous 14.
beta-lactamases from clinical isolates of Serratia marcescens with [74] Humeniuk C, Arlet G, Gautier V, Grimont P, Labia R, Philippon A.
different substrate specificities. Antimicrobial Agents Chemotherap -lactamases of Kluyvera ascorbata, probable progenitors of some
1998; 42: 176-9. plasmid-encoded CTX-M types. Antimicrobial Agents
[54] Rodriguez-Martinez JM, Poirel L, Nordmann P. Extended- Chemotherap 2002; 46: 3045-9.
spectrum cephalosporinases in Pseudomonas aeruginosa. [75] Celenza G, Pellegrini C, Caccamo M, Segatore B, Amicosante G,
Antimicrobial Agents Chemotherap 2009; 53: 1766-71. Perilli M. Spread of bla(CTX-M-type) and bla(PER-2) -lactamase
[55] Rodriguez-Martinez JM, Poirel L, Nordmann P. Molecular genes in clinical isolates from Bolivian hospitals. J antimicrobial
epidemiology and mechanisms of carbapenem resistance in chemotherapy 2006; 57: 975-8.
Pseudomonas aeruginosa. Antimicrobial Agents Chemotherap [76] Picao RC, Poirel L, Gales AC, Nordmann P. Further identification
2009; 53: 4783-8. of CTX-M-2 extended-spectrum beta-lactamase in Pseudomonas
[56] Datta N, Richmond MH. The purification and properties of a aeruginosa. Antimicrobial Agents Chemotherap 2009; 53: 2225-6.
penicillinase whose synthesis is mediated by an R-factor in [77] Nordmann P, Ronco E, Naas T, Duport C, Michel-Briand Y, Labia
Escherichia coli. The Biochemical journal 1966; 98: 204-9. R. Characterization of a novel extended-spectrum -lactamase from
[57] Matthew M. Plasmid-mediated -lactamases of Gram-negative Pseudomonas aeruginosa. Antimicrobial Agents Chemotherap
bacteria: properties and distribution. J antimicrobial chemotherapy 1993; 37: 962-9.
1979; 5: 349-58. [78] Naas T, Poirel L, Karim A, Nordmann P. Molecular
[58] Jacoby GA, Matthew M. The distribution of -lactamase genes on characterization of In50, a class 1 integron encoding the gene for
plasmids found in Pseudomonas. Plasmid 1979; 2: 41-7. the extended-spectrum -lactamase VEB-1 in Pseudomonas
[59] Brun-Buisson C, Legrand P, Rauss A, et al. Intestinal aeruginosa. FEMS microbiology letters 1999; 176: 411-9.
decontamination for control of nosocomial multiresistant gram- [79] Girlich D, Naas T, Leelaporn A, Poirel L, Fennewald M,
negative bacilli. Study of an outbreak in an intensive care unit. Nordmann P. Nosocomial spread of the integron-located VEB-1-
Annals of internal medicine 1989; 110: 873-81. like cassette encoding an extended-pectrum -lactamase in
[60] Kliebe C, Nies BA, Meyer JF, Tolxdorff-Neutzling RM, Pseudomonas aeruginosa in Thailand. Clinical infectious diseases :
Wiedemann B. Evolution of plasmid-coded resistance to broad- an official publication of the Infectious Diseases Society of
spectrum cephalosporins. Antimicrobial Agents Chemotherap America 2002; 34: 603-11.
1985; 28: 302-7. [80] Poirel L, Rotimi VO, Mokaddas EM, Karim A, Nordmann P. VEB-
[61] Knothe H, Shah P, Krcmery V, Antal M, Mitsuhashi S. 1-like extended-spectrum -lactamases in Pseudomonas
Transferable Resistance to Cefotaxime, Cefoxitin, Cefamandole aeruginosa, Kuwait. Emerging infectious diseases 2001; 7: 468-70.
and Cefuroxime in Clinical Isolates of Klebsiella pneumoniae and [81] Poirel L, Le Thomas I, Naas T, Karim A, Nordmann P.
Serratia marcescens. Infection 1983; 11: 315-7. Biochemical sequence analyses of GES-1, a novel class A
[62] Bradford PA. Extended-spectrum beta-lactamases in the 21st extended-spectrum -lactamase, and the class 1 integron In52 from
century: characterization, epidemiology, and detection of this Klebsiella pneumoniae. Antimicrobial Agents Chemotherap 2000;
important resistance threat. Clin Microbiol Rev 2001; 14: 933-51, 44: 622-32.
table of contents. [82] Dubois V, Poirel L, Marie C, Arpin C, Nordmann P, Quentin C.
[63] Paterson DL, Bonomo RA. Extended-spectrum -lactamases: a Molecular characterization of a novel class 1 integron containing
clinical update. Clinical microbiology reviews 2005; 18: 657-86. bla(GES-1) and a fused product of aac3-Ib/aac6'-Ib' gene cassettes
[64] Mugnier P, Dubrous P, Casin I, Arlet G, Collatz E. A TEM-derived in Pseudomonas aeruginosa. Antimicrobial Agents Chemotherap
extended-spectrum -lactamase in Pseudomonas aeruginosa. 2002; 46: 638-45.
Antimicrobial Agents Chemotherap 1996; 40: 2488-93. [83] Kotsakis SD, Papagiannitsis CC, Tzelepi E, Legakis NJ, Miriagou
[65] al Naiemi N, Duim B, Bart A. A CTX-M extended-spectrum - V, Tzouvelekis LS. GES-13, a -lactamase variant possessing Lys-
lactamase in Pseudomonas aeruginosa and Stenotrophomonas 104 and Asn-170 in Pseudomonas aeruginosa. Antimicrobial
maltophilia. Journal of medical microbiology 2006; 55: 1607-8. Agents Chemotherap 2010; 54: 1331-3.
[66] Dubois V, Arpin C, Noury P, Quentin C. Clinical strain of [84] Mavroidi A, Tzelepi E, Tsakris A, Miriagou V, Sofianou D,
Pseudomonas aeruginosa carrying a bla(TEM-21) gene located on Tzouvelekis LS. An integron-associated -lactamase (IBC-2) from
a chromosomal interrupted TnA type transposon. Antimicrobial Pseudomonas aeruginosa is a variant of the extended-spectrum
Agents Chemotherap 2002; 46: 3624-6. beta-lactamase IBC-1. J antimicrobial chemotherapy 2001; 48:
[67] Marchandin H, Jean-Pierre H, De Champs C, et al. Production of a 627-30.
TEM-24 plasmid-mediated extended-spectrum -lactamase by a [85] Poirel L, Brinas L, Fortineau N, Nordmann P. Integron-encoded
clinical isolate of Pseudomonas aeruginosa. Antimicrobial Agents GES-type extended-spectrum -lactamase with increased activity
Chemotherap 2000; 44: 213-6. toward aztreonam in Pseudomonas aeruginosa. Antimicrobial
[68] Poirel L, Ronco E, Naas T, Nordmann P. Extended-spectrum - Agents Chemotherap 2005; 49: 3593-7.
lactamase TEM-4 in Pseudomonas aeruginosa. Clinical [86] Poirel L, Brinas L, Verlinde A, Ide L, Nordmann P. BEL-1, a novel
microbiology and infection : the official publication of the clavulanic acid-inhibited extended-spectrum -lactamase, and the
European Society of Clinical Microbiology and Infectious Diseases class 1 integron In120 in Pseudomonas aeruginosa. Antimicrobial
1999; 5: 651-2. Agents Chemotherap 2005; 49: 3743-8.
[69] Chanawong A, M'Zali FH, Heritage J, Lulitanond A, Hawkey PM. [87] Poirel L, Docquier JD, De Luca F, et al. BEL-2, an extended-
SHV-12, SHV-5, SHV-2a and VEB-1 extended-spectrum - spectrum -lactamase with increased activity toward expanded-
lactamases in Gram-negative bacteria isolated in a university spectrum cephalosporins in Pseudomonas aeruginosa.
hospital in Thailand. J antimicrobial chemotherapy 2001; 48: 839- Antimicrobial Agents Chemotherap 2010; 54: 533-5.
52. [88] Tian GB, Adams-Haduch JM, Bogdanovich T, Wang HN, Doi Y.
PME-1, an extended-spectrum -lactamase identified in
Antibacterial Resistant Pseudomonas aeruginosa Current Pharmaceutical Design, 2013, Vol. 19, No. 2 219

Pseudomonas aeruginosa. Antimicrobial Agents Chemotherap [109] Ge C, Wei Z, Jiang Y, Shen P, Yu Y, Li L. Identification of KPC-
2011; 55: 2710-3. 2-producing Pseudomonas aeruginosa isolates in China. J
[89] Paisley JW, Washington JA, 2nd. Combined activitiy of clavulanic antimicrobial chemotherap 2011; 66: 1184-6.
acid and ticarcillin against ticarcillin-resistant, gram-negative [110] Poirel L, Nordmann P, Lagrutta E, Cleary T, Munoz-Price LS.
bacilli. Antimicrobial Agents Chemotherap 1978; 14: 224-7. Emergence of KPC-producing Pseudomonas aeruginosa in the
[90] Thomson KS, Weber DA, Sanders CC, Sanders WE, Jr. - United States. Antimicrobial Agents Chemotherap 2010; 54: 3072.
lactamase production in members of the family Enterobacteriaceae [111] Wolter DJ, Kurpiel PM, Woodford N, Palepou MF, Goering RV,
and resistance to -lactam-enzyme inhibitor combinations. Hanson ND. Phenotypic and enzymatic comparative analysis of the
Antimicrobial Agents Chemotherap 1990; 34: 622-7. novel KPC variant KPC-5 and its evolutionary variants, KPC-2 and
[91] Sanders CC, Iaconis JP, Bodey GP, Samonis G. Resistance to KPC-4. Antimicrob Agents Chemother 2009; 53: 557-62.
ticarcillin-potassium clavulanate among clinical isolates of the [112] Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo-beta-
family Enterobacteriaceae: role of PSE-1 -lactamase and high lactamases: the quiet before the storm? Clinical microbiology
levels of TEM-1 and SHV-1 and problems with false susceptibility reviews 2005; 18: 306-25.
in disk diffusion tests. Antimicrobial Agents Chemotherap 1988; [113] Watanabe M, Iyobe S, Inoue M, Mitsuhashi S. Transferable
32: 1365-9. imipenem resistance in Pseudomonas aeruginosa. Antimicrob
[92] Poirel L, Naas T, Nordmann P. Diversity, epidemiology, and Agents Chemother 1991; 35: 147-51.
genetics of class D -lactamases. Antimicrobial Agents [114] Hancock RE, Brinkman FS. Function of pseudomonas porins in
Chemotherap 2010; 54: 24-38. uptake and efflux. Annu Rev Microbiol 2002; 56: 17-38.
[93] Hall LM, Livermore DM, Gur D, Akova M, Akalin HE. OXA-11, [115] Trias J, Nikaido H. Protein D2 channel of the Pseudomonas
an extended-spectrum variant of OXA-10 (PSE-2) -lactamase aeruginosa outer membrane has a binding site for basic amino
from Pseudomonas aeruginosa. Antimicrobial Agents acids and peptides. J Biol Chem 1990; 265: 15680-4.
Chemotherap 1993; 37: 1637-44. [116] Trias J, Nikaido H. Outer membrane protein D2 catalyzes
[94] Juan C, Mulet X, Zamorano L, Alberti S, Perez JL, Oliver A. facilitated diffusion of carbapenems and penems through the outer
Detection of the novel extended-spectrum -lactamase OXA-161 membrane of Pseudomonas aeruginosa. Antimicrob Agents
from a plasmid-located integron in Pseudomonas aeruginosa Chemother 1990; 34: 52-7.
clinical isolates from Spain. Antimicrobial Agents Chemotherap [117] Kohler T, Michea-Hamzehpour M, Epp SF, Pechere JC.
2009; 53: 5288-90. Carbapenem activities against Pseudomonas aeruginosa: respective
[95] Walther-Rasmussen J, Hoiby N. OXA-type carbapenemases. J contributions of OprD and efflux systems. Antimicrob Agents
antimicrobial chemotherapy 2006; 57: 373-83. Chemother 1999; 43: 424-7.
[96] Girlich D, Naas T, Nordmann P. Biochemical characterization of [118] Livermore DM. Multiple mechanisms of antimicrobial resistance in
the naturally occurring oxacillinase OXA-50 of Pseudomonas Pseudomonas aeruginosa: our worst nightmare? Clin Infect Dis
aeruginosa. Antimicrob Agents Chemother 2004; 48: 2043-8. 2002; 34: 634-40.
[97] Sevillano E, Gallego L, Garcia-Lobo JM. First detection of the [119] Quinn JP, Studemeister AE, DiVincenzo CA, Lerner SA.
OXA-40 carbapenemase in P. aeruginosa isolates, located on a Resistance to imipenem in Pseudomonas aeruginosa: clinical
plasmid also found in A. baumannii. Pathologie-biologie 2009; 57: experience and biochemical mechanisms. Rev Infect Dis 1988; 10:
493-5. 892-8.
[98] Bou G, Oliver A, Martinez-Beltran J. OXA-24, a novel class D - [120] Wolter DJ, Hanson ND, Lister PD. Discordance Between
lactamase with carbapenemase activity in an Acinetobacter Imipenem (IPM) Resistance and oprD Expression in a
baumannii clinical strain. Antimicrobial Agents Chemotherap Pseudomonas aeruginosa (PA) Overexpressing mexEF-oprN
2000; 44: 1556-61. (EFN), abstr. C1-1061. In Program and abstracts of the 48rd
[99] Queenan AM, Bush K. Carbapenemases: the versatile - Interscience Conference on Antimicrobial Agents Chemotherap.
lactamases. Clin Microbiol Rev 2007; 20: 440-58, table of contents. American Society for Microbiology, Washington, D.C. 2008.
[100] Walther-Rasmussen J, Hoiby N. Class A carbapenemases. J [121] Perez FJ, Gimeno C, Navarro D, Garcia-de-Lomas J. Meropenem
antimicrobial chemotherapy 2007; 60: 470-82. permeation through the outer membrane of Pseudomonas
[101] Labuschagne Cde J, Weldhagen GF, Ehlers MM, Dove MG. aeruginosa can involve pathways other than the OprD porin
Emergence of class 1 integron-associated GES-5 and GES-5-like channel. Chemotherapy 1996; 42: 210-14.
extended-spectrum beta-lactamases in clinical isolates of [122] Sakyo S, Tomita H, Tanimoto K, Fujimoto S, Ike Y. Potency of
Pseudomonas aeruginosa in South Africa. Int J Antimicrobial carbapenems for the prevention of carbapenem-resistant mutants of
Agents 2008; 31: 527-30. Pseudomonas aeruginosa: the high potency of a new carbapenem
[102] Picao RC, Poirel L, Gales AC, Nordmann P. Diversity of - doripenem. Journal of antibiotics 2006; 59: 220-8.
lactamases produced by ceftazidime-resistant Pseudomonas [123] Brown SD, Traczewski MM. Comparative in vitro antimicrobial
aeruginosa isolates causing bloodstream infections in Brazil. activity of a new carbapenem, doripenem: tentative disc diffusion
Antimicrobial Agents Chemotherap 2009; 53: 3908-13. criteria and quality control. Journal of antimicrobial chemotherapy
[103] Poirel L, Weldhagen GF, De Champs C, Nordmann P. A 2005; 55: 944-9.
nosocomial outbreak of Pseudomonas aeruginosa isolates [124] Tsuji M, Ishii Y, Ohno A, Miyazaki S, Yamaguchi K. In vitro and
expressing the extended-spectrum -lactamase GES-2 in South in vivo antibacterial activities of S-4661, a new carbapenem.
Africa. J antimicrobial chemotherapy 2002; 49: 561-5. Antimicrobial Agents Chemotherap 1998; 42: 94-9.
[104] Poirel L, Weldhagen GF, Naas T, De Champs C, Dove MG, [125] Riera E, Cabot G, Mulet X, et al. Pseudomonas aeruginosa
Nordmann P. GES-2, a class A -lactamase from Pseudomonas carbapenem resistance mechanisms in Spain: impact on the activity
aeruginosa with increased hydrolysis of imipenem. Antimicrobial of imipenem, meropenem and doripenem. J Antimicrobial
Agents Chemotherap 2001; 45: 2598-603. Chemotherapy 2011.
[105] Wang C, Cai P, Chang D, Mi Z. A Pseudomonas aeruginosa [126] Gotoh N, Wakebe H, Nishino T. Ultrastructural aspects of fragility
isolate producing the GES-5 extended-spectrum -lactamase. J of Pseudomonas aeruginosa outer membrane devoid of protein F.
antimicrobial chemotherapy 2006; 57: 1261-2. FEMS microbiology letters 1989; 50: 51-3.
[106] Villegas MV, Lolans K, Correa A, Kattan JN, Lopez JA, Quinn JP. [127] Gotoh N, Wakebe H, Yoshihara E, Nakae T, Nishino T. Role of
First identification of Pseudomonas aeruginosa isolates producing protein F in maintaining structural integrity of the Pseudomonas
a KPC-type carbapenem-hydrolyzing -lactamase. Antimicrob aeruginosa outer membrane. Journal of bacteriology 1989; 171:
Agents Chemother 2007; 51: 1553-5. 983-90.
[107] Wolter DJ, Khalaf N, Robledo IE, et al. Surveillance of [128] Rawling EG, Brinkman FS, Hancock RE. Roles of the carboxy-
Carbapenem Resistant Pseudomonas aeruginosa from Puerto Rico terminal half of Pseudomonas aeruginosa major outer membrane
Medical Center Hospitals: Dissemination of KPC and IMP-18 - protein OprF in cell shape, growth in low-osmolarity medium, and
lactamases. Antimicrob Agents Chemother 2009. peptidoglycan association. J Bacteriol 1998; 180: 3556-62.
[108] Akpaka PE, Swanston WH, Ihemere HN, et al. Emergence of KPC- [129] Nestorovich EM, Sugawara E, Nikaido H, Bezrukov SM.
producing Pseudomonas aeruginosa in Trinidad and Tobago. Pseudomonas aeruginosa porin OprF: properties of the channel. J
Journal of clinical microbiology 2009; 47: 2670-1. biological chemistry 2006; 281: 16230-7.
220 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

[130] Azghani AO, Idell S, Bains M, Hancock RE. Pseudomonas [152] Morita Y, Kimura N, Mima T, Mizushima T, Tsuchiya T. Roles of
aeruginosa outer membrane protein F is an adhesin in bacterial MexXY- and MexAB-multidrug efflux pumps in intrinsic
binding to lung epithelial cells in culture. Microb Pathog 2002; 33: multidrug resistance of Pseudomonas aeruginosa PAO1. J Gen
109-14. Appl Microbiol 2001; 47: 27-32.
[131] Yoon SS, Hennigan RF, Hilliard GM, et al. Pseudomonas [153] Maseda H, Sawada I, Saito K, Uchiyama H, Nakae T, Nomura N.
aeruginosa anaerobic respiration in biofilms: relationships to cystic Enhancement of the mexAB-oprM efflux pump expression by a
fibrosis pathogenesis. Dev Cell 2002; 3: 593-603. quorum-sensing autoinducer and its cancellation by a regulator,
[132] Piddock LJ, Hall MC, Bellido F, Bains M, Hancock RE. A MexT, of the mexEF-oprN efflux pump operon in Pseudomonas
pleiotropic, posttherapy, enoxacin-resistant mutant of aeruginosa. Antimicrob Agents Chemother 2004; 48: 1320-8.
Pseudomonas aeruginosa. Antimicrob Agents Chemother 1992; [154] Sawada I, Maseda H, Nakae T, Uchiyama H, Nomura N. A
36: 1057-61. quorum-sensing autoinducer enhances the mexAB-oprM efflux-
[133] Woodruff WA, Hancock RE. Construction and characterization of pump expression without the MexR-mediated regulation in
Pseudomonas aeruginosa protein F-deficient mutants after in vitro Pseudomonas aeruginosa. Microbiol Immunol 2004; 48: 435-9.
and in vivo insertion mutagenesis of the cloned gene. J Bacteriol [155] Evans K, Adewoye L, Poole K. MexR repressor of the mexAB-
1988; 170: 2592-8. oprM multidrug efflux operon of Pseudomonas aeruginosa:
[134] Woodruff WA, Hancock RE. Pseudomonas aeruginosa outer identification of MexR binding sites in the mexA-mexR intergenic
membrane protein F: structural role and relationship to the region. J Bacteriol 2001; 183: 807-12.
Escherichia coli OmpA protein. J Bacteriol 1989; 171: 3304-9. [156] Poole K, Tetro K, Zhao Q, Neshat S, Heinrichs DE, Bianco N.
[135] Stover CK, Pham XQ, Erwin AL, et al. Complete genome Expression of the multidrug resistance operon mexA-mexB-oprM in
sequence of Pseudomonas aeruginosa PA01, an opportunistic Pseudomonas aeruginosa: mexR encodes a regulator of operon
pathogen. Nature 2000; 406: 959-64. expression. Antimicrob Agents Chemother 1996; 40: 2021-8.
[136] Pos KM, Schiefner A, Seeger MA, Diederichs K. Crystallographic [157] Morita Y, Cao L, Gould VC, Avison MB, Poole K. nalD encodes a
analysis of AcrB. FEBS letters 2004; 564: 333-9. second repressor of the mexAB-oprM multidrug efflux operon of
[137] Yu EW, McDermott G, Zgurskaya HI, Nikaido H, Koshland DE, Pseudomonas aeruginosa. J Bacteriol 2006; 188: 8649-54.
Jr. Structural basis of multiple drug-binding capacity of the AcrB [158] Adewoye L, Sutherland A, Srikumar R, Poole K. The mexR
multidrug efflux pump. Science 2003; 300: 976-80. repressor of the mexAB-oprM multidrug efflux operon in
[138] Middlemiss JK, Poole K. Differential impact of MexB mutations Pseudomonas aeruginosa: characterization of mutations
on substrate selectivity of the MexAB-OprM multidrug efflux compromising activity. J Bacteriol 2002; 184: 4308-12.
pump of Pseudomonas aeruginosa. J Bacteriol 2004; 186: 1258-69. [159] Saito K, Akama H, Yoshihara E, Nakae T. Mutations affecting
[139] Yu EW, Aires JR, Nikaido H. AcrB multidrug efflux pump of DNA-binding activity of the MexR repressor of mexR-mexA-
Escherichia coli: composite substrate-binding cavity of exceptional mexB-oprM operon expression. J Bacteriol 2003; 185: 6195-8.
flexibility generates its extremely wide substrate specificity. J [160] Cao L, Srikumar R, Poole K. MexAB-OprM hyperexpression in
Bacteriol 2003; 185: 5657-64. NalC-type multidrug-resistant Pseudomonas aeruginosa:
[140] Mao W, Warren MS, Black DS, et al. On the mechanism of identification and characterization of the nalC gene encoding a
substrate specificity by resistance nodulation division (RND)-type repressor of PA3720-PA3719. Mol Microbiol 2004; 53: 1423-36.
multidrug resistance pumps: the large periplasmic loops of MexD [161] Daigle DM, Cao L, Fraud S, et al. Protein modulator of multidrug
from Pseudomonas aeruginosa are involved in substrate efflux gene expression in Pseudomonas aeruginosa. J Bacteriol
recognition. Mol Microbiol 2002; 46: 889-901. 2007; 189: 5441-51.
[141] Yu EW, Aires JR, McDermott G, Nikaido H. A periplasmic drug- [162] Wilke MS, Heller M, Creagh AL, et al. The crystal structure of
binding site of the AcrB multidrug efflux pump: a crystallographic MexR from Pseudomonas aeruginosa in complex with its
and site-directed mutagenesis study. Journal of bacteriology 2005; antirepressor ArmR. Proc Natl Acad Sci U S A 2008; 105: 14832-
187: 6804-15. 7.
[142] Bohnert JA, Schuster S, Seeger MA, Fahnrich E, Pos KM, Kern [163] Srikumar R, Paul CJ, Poole K. Influence of mutations in the mexR
WV. Site-directed mutagenesis reveals putative substrate binding repressor gene on expression of the MexA-MexB-oprM multidrug
residues in the Escherichia coli RND efflux pump AcrB. Journal of efflux system of Pseudomonas aeruginosa. J Bacteriol 2000; 182:
bacteriology 2008; 190: 8225-9. 1410-4.
[143] Murakami S, Nakashima R, Yamashita E, Matsumoto T, [164] Masuda N, Gotoh N, Ohya S, Nishino T. Quantitative correlation
Yamaguchi A. Crystal structures of a multidrug transporter reveal a between susceptibility and OprJ production in NfxB mutants of
functionally rotating mechanism. Nature 2006; 443: 173-9. Pseudomonas aeruginosa. Antimicrob Agents Chemother 1996;
[144] Pages JM, Masi M, Barbe J. Inhibitors of efflux pumps in Gram- 40: 909-13.
negative bacteria. Trends in molecular medicine 2005; 11: 382-9. [165] Poole K, Gotoh N, Tsujimoto H, et al. Overexpression of the
[145] Aires JR, Kohler T, Nikaido H, Plesiat P. Involvement of an active mexC-mexD-oprJ efflux operon in nfxB-type multidrug-resistant
efflux system in the natural resistance of Pseudomonas aeruginosa strains of Pseudomonas aeruginosa. Mol Microbiol 1996; 21: 713-
to aminoglycosides. Antimicrob Agents Chemother 1999; 43: 24.
2624-8. [166] Masuda N, Sakagawa E, Ohya S, Gotoh N, Tsujimoto H, Nishino
[146] Chuanchuen R, Murata T, Gotoh N, Schweizer HP. Substrate- T. Substrate specificities of MexAB-OprM, MexCD-OprJ, and
dependent utilization of OprM or OpmH by the Pseudomonas MexXY-oprM efflux pumps in Pseudomonas aeruginosa.
aeruginosa MexJK efflux pump. Antimicrob Agents Chemother Antimicrob Agents Chemother 2000; 44: 3322-7.
2005; 49: 2133-6. [167] Okamoto K, Gotoh N, Nishino T. Alterations of susceptibility of
[147] Mine T, Morita Y, Kataoka A, Mizushima T, Tsuchiya T. Pseudomonas aeruginosa by overproduction of multidrug efflux
Expression in Escherichia coli of a new multidrug efflux pump, systems, MexAB-OprM, MexCD-OprJ, and MexXY/OprM to
MexXY, from Pseudomonas aeruginosa. Antimicrob Agents carbapenems: substrate specificities of the efflux systems. Journal
Chemother 1999; 43: 415-7. of infection and chemotherapy : official journal of the Japan
[148] Murata T, Gotoh N, Nishino T. Characterization of outer Society of Chemotherapy 2002; 8: 371-3.
membrane efflux proteins OpmE, OpmD and OpmB of [168] Gotoh N, Tsujimoto H, Tsuda M, et al. Characterization of the
Pseudomonas aeruginosa: molecular cloning and development of MexC-MexD-OprJ multidrug efflux system in mexA-mexB-oprM
specific antisera. FEMS Microbiol Lett 2002; 217: 57-63. mutants of Pseudomonas aeruginosa. Antimicrob Agents
[149] Li XZ, Zhang L, Srikumar R, Poole K. -lactamase inhibitors are Chemother 1998; 42: 1938-43.
substrates for the multidrug efflux pumps of Pseudomonas [169] Morita Y, Komori Y, Mima T, Kuroda T, Mizushima T, Tsuchiya
aeruginosa. Antimicrob Agents Chemother 1998; 42: 399-403. T. Construction of a series of mutants lacking all of the four major
[150] Poole K, Srikumar R. Multidrug efflux in Pseudomonas mex operons for multidrug efflux pumps or possessing each one of
aeruginosa: components, mechanisms and clinical significance. the operons from Pseudomonas aeruginosa PAO1: MexCD-OprJ is
Curr Top Med Chem 2001; 1: 59-71. an inducible pump. FEMS Microbiol Lett 2001; 202: 139-43.
[151] Li XZ, Nikaido H, Poole K. Role of mexA-mexB-oprM in antibiotic [170] Srikumar R, Li XZ, Poole K. Inner membrane efflux components
efflux in Pseudomonas aeruginosa. Antimicrob Agents Chemother are responsible for -lactam specificity of multidrug efflux pumps
1995; 39: 1948-53. in Pseudomonas aeruginosa. J Bacteriol 1997; 179: 7875-81.
Antibacterial Resistant Pseudomonas aeruginosa Current Pharmaceutical Design, 2013, Vol. 19, No. 2 221

[171] Shiba T, Ishiguro K, Takemoto N, Koibuchi H, Sugimoto K. resistance to aminoglycosides among Pseudomonas aeruginosa
Purification and characterization of the Pseudomonas aeruginosa isolates from patients with cystic fibrosis. Antimicrobial Agents
NfxB protein, the negative regulator of the nfxB gene. J Bacteriol Chemotherap 2004; 48: 1676-1680.
1995; 177: 5872-7. [190] Wolter DJ, Smith-Moland E, Goering RV, Hanson ND, Lister PD.
[172] Dean CR, Visalli MA, Projan SJ, Sum PE, Bradford PA. Efflux- Multidrug resistance associated with mexXY expression in clinical
mediated resistance to tigecycline (GAR-936) in Pseudomonas isolates of Pseudomonas aeruginosa from a Texas hospital.
aeruginosa PAO1. Antimicrob Agents Chemother 2003; 47: 972-8. Diagnostic Microbiology and Infectious Disease 2004; 50: 43-50.
[173] Jalal S, Ciofu O, Hoiby N, Gotoh N, Wretlind B. Molecular [191] Laohavaleeson S, Lolans K, Quinn JP, Kuti JL, Nicolau DP.
mechanisms of fluoroquinolone resistance in Pseudomonas Expression of the MexXY-OprM efflux system in Pseudomonas
aeruginosa isolates from cystic fibrosis patients. Antimicrob aeruginosa with discordant cefepime/ceftazidime susceptibility
Agents Chemother 2000; 44: 710-2. profiles. Infection and drug resistance 2008; 1: 51-5.
[174] Hocquet D, Nordmann P, El Garch F, Cabanne L, Plesiat P. [192] Jeannot K, Elsen S, Kohler T, Attree I, van Delden C, Plesiat P.
Involvement of the MexXY-OprM efflux system in emergence of Resistance and virulence of Pseudomonas aeruginosa clinical
cefepime resistance in clinical strains of Pseudomonas aeruginosa. strains overproducing the MexCD-OprJ efflux pump. Antimicrob
Antimicrob Agents Chemother 2006; 50: 1347-51. Agents Chemother 2008; 52: 2455-62.
[175] Baum EZ, Crespo-Carbone SM, Morrow BJ, et al. Effect of [193] Reinhardt A, Kohler T, Wood P, et al. Development and
MexXY overexpression on ceftobiprole susceptibility in persistence of antimicrobial resistance in Pseudomonas aeruginosa:
Pseudomonas aeruginosa. Antimicrobial Agents Chemotherap a longitudinal observation in mechanically ventilated patients.
2009; 53: 2785-90. Antimicrobial Agents Chemotherap 2007; 51: 1341-50.
[176] Queenan AM, Shang W, Bush K, Flamm RK. Differential selection [194] Campo Esquisabel AB, Rodriguez MC, et al. Mechanisms of
of single-step AmpC or efflux mutants of Pseudomonas aeruginosa resistance in clinical isolates of Pseudomonas aeruginosa less
by using cefepime, ceftazidime, or ceftobiprole. Antimicrobial susceptible to cefepime than to ceftazidime. Clinical microbiology
Agents Chemotherap 2010; 54: 4092-7. and infection : the official publication of the European Society of
[177] Masuda N, Sakagawa E, Ohya S, Gotoh N, Tsujimoto H, Nishino Clinical Microbiology and Infectious Diseases 2011.
T. Contribution of the MexX-MexY-oprM efflux system to [195] Linares JF, Lopez JA, Camafeita E, Albar JP, Rojo F, Martinez JL.
intrinsic resistance in Pseudomonas aeruginosa. Antimicrob Overexpression of the multidrug efflux pumps MexCD-OprJ and
Agents Chemother 2000; 44: 2242-6. MexEF-OprN is associated with a reduction of type III secretion in
[178] Jeannot K, Sobel ML, El Garch F, Poole K, Plesiat P. Induction of Pseudomonas aeruginosa. Journal of bacteriology 2005; 187: 1384-
the MexXY efflux pump in Pseudomonas aeruginosa is dependent 91.
on drug-ribosome interaction. J Bacteriol 2005; 187: 5341-6. [196] Sanchez P, Linares JF, Ruiz-Diez B, et al. Fitness of in vitro
[179] Matsuo Y, Eda S, Gotoh N, Yoshihara E, Nakae T. MexZ-mediated selected Pseudomonas aeruginosa nalB and nfxB multidrug
regulation of mexXY multidrug efflux pump expression in resistant mutants. J antimicrobial chemotherapy 2002; 50: 657-64.
Pseudomonas aeruginosa by binding on the mexZ-mexX intergenic [197] Stickland HG, Davenport PW, Lilley KS, Griffin JL, Welch M.
DNA. FEMS Microbiol Lett 2004; 238: 23-8. Mutation of nfxB causes global changes in the physiology and
[180] Llanes C, Hocquet D, Vogne C, Benali-Baitich D, Neuwirth C, metabolism of Pseudomonas aeruginosa. Journal of proteome
Plesiat P. Clinical strains of Pseudomonas aeruginosa research 2010; 9: 2957-67.
overproducing MexAB-OprM and MexXY efflux pumps [198] Pai H, Kim J, Lee JH, Choe KW, Gotoh N. Carbapenem resistance
simultaneously. Antimicrob Agents Chemother 2004; 48: 1797- mechanisms in Pseudomonas aeruginosa clinical isolates.
802. Antimicrob Agents Chemother 2001; 45: 480-4.
[181] Muller C, Plesiat P, Jeannot K. A two-component regulatory [199] Tomas M, Doumith M, Warner M, et al. Efflux pumps, OprD
system interconnects resistance to polymyxins, aminoglycosides, porin, AmpC beta-lactamase, and multiresistance in Pseudomonas
fluoroquinolones, and beta-lactams in Pseudomonas aeruginosa. aeruginosa isolates from cystic fibrosis patients. Antimicrobial
Antimicrobial Agents Chemotherap 2011; 55: 1211-21. Agents Chemotherap 2010; 54: 2219-24.
[182] Adewoye L, Sutherland A, Srikumar R, Poole K. The mexR [200] Wolter DJ, Acquazzino D, Goering RV, Sammut P, Khalaf N,
repressor of the mexAB-oprM multidrug efflux operon in Hanson ND. Emergence of carbapenem resistance in Pseudomonas
Pseudomonas aeruginosa: characterization of mutations aeruginosa isolates from a patient with cystic fibrosis in the
compromising activity. Journal of Bacteriology 2002; 184: 4308- absence of carbapenem therapy. Clin Infect Dis 2008; 46: e137-41.
4312. [201] Pumbwe L, Piddock LJ. Two efflux systems expressed
[183] Llanes C, Hocquet D, Vogne C, Benali-Baitich D, Neuwirth C, simultaneously in multidrug-resistant Pseudomonas aeruginosa.
Plesiat P. Clinical strains of Pseudomonas aeruginosa Antimicrob Agents Chemother 2000; 44: 2861-4.
overproducing MexAB-OprM and MexXY efflux pumps [202] Hocquet D, Roussel-Delvallez M, Cavallo JD, Plesiat P. MexAB-
simultaneously. Antimicrobial Agents Chemotherap 2004; 48: OprM- and MexXY-overproducing mutants are very prevalent
1797-1802. among clinical strains of Pseudomonas aeruginosa with reduced
[184] Pai H, Kim J, Lee JH, Choe KW, Gotoh N. Carbapenem resistance susceptibility to ticarcillin. Antimicrobial Agents Chemotherap
mechanims in Pseudomonas aeruginosa clinical isolates. 2007; 51: 1582-3.
Antimicrobial Agents Chemotherap 2001; 45: 480-484. [203] Cabot G, Ocampo-Sosa AA, Tubau F, et al. Overexpression of
[185] Sobel ML, Hocquet D, Cao L, Plesiat P, Poole K. Mutations in AmpC and efflux pumps in Pseudomonas aeruginosa isolates from
PA3574 (nalD) lead to increased MexAB-OprM expression and bloodstream infections: prevalence and impact on resistance in a
multidrug resistance in laboratory and clinical isolates of Spanish multicenter study. Antimicrobial Agents Chemotherap
Pseudomonas aeruginosa. Antimicrobial Agents Chemotherap 2011; 55: 1906-11.
2005; 49: 1782-1786. [204] Quale J, Bratu S, Gupta J, Landman D. Interplay of efflux system,
[186] Wolter DJ, Acquazzino D, Goering RV, Sammut P, Khalaf N, ampC, and oprD expression in carbapenem resistance of
Hanson ND. Emergence of carbapenem resistance in Pseudomonas Pseudomonas aeruginosa clinical isolates. Antimicrobial Agents
aeruginosa isolates from a patient with cystic fibrosis in the Chemotherap 2006; 50: 1633-41.
absence of carbapenem therapy. Clinical Infectious Diseases 2008; [205] Hocquet D, Berthelot P, Roussel-Delvallez M, et al. Pseudomonas
46:e137-141. aeruginosa may accumulate drug resistance mechanisms without
[187] Hocquet D, Nordmann P, El Garch F, Cabanne L, Plesiat P. losing its ability to cause bloodstream infections. Antimicrobial
Involvement of the MexXY-OprM efflux system in emergence of Agents Chemotherap 2007; 51: 3531-6.
cefepime resistance in clinical strains of Pseudomonas aeruginosa. [206] Wolter DJ, Smith-Moland E, Goering RV, Hanson ND, Lister PD.
Antimicrobial Agents Chemotherap 2006; 50: 1347-1351. Multidrug resistance associated with mexXY expression in clinical
[188] Sobel ML, McKay GA, Poole K. Contribution of the MexXY isolates of Pseudomonas aeruginosa from a Texas hospital. Diagn
multidrug transporter to aminoglycoside resistance in Pseudomonas Microbiol Infect Dis 2004; 50: 43-50.
aeruginosa clinical isolates. Antimicrobial Agents Chemotherap [207] Xavier DE, Picao RC, Girardello R, Fehlberg LC, Gales AC.
2003; 48: 3202-3207. Efflux pumps expression and its association with porin down-
[189] Vogne C, Aires JR, Bailly C, Hocquet D, Plesiat P. Role of the regulation and -lactamase production among Pseudomonas
multidrug efflux system MexXY in the emergence of moderate
222 Current Pharmaceutical Design, 2013, Vol. 19, No. 2 Wolter and Lister

aeruginosa causing bloodstream infections in Brazil. BMC [212] Peng X, Xu C, Ren H, Lin X, Wu L, Wang S. Proteomic analysis
microbiology 2010; 10: 217. of the sarcosine-insoluble outer membrane fraction of
[208] Maniati M, Ikonomidis A, Mantzana P, Daponte A, Maniatis AN, Pseudomonas aeruginosa responding to ampicilin, kanamycin, and
Pournaras S. A highly carbapenem-resistant Pseudomonas tetracycline resistance. Journal of proteome research 2005; 4: 2257-
aeruginosa isolate with a novel blaVIM-4/blaP1b integron 65.
overexpresses two efflux pumps and lacks OprD. J antimicrobial [213] Liberati NT, Urbach JM, Miyata S, et al. An ordered, nonredundant
chemotherapy 2007; 60: 132-5. library of Pseudomonas aeruginosa strain PA14 transposon
[209] Vettoretti L, Floret N, Hocquet D, et al. Emergence of extensive- insertion mutants. Proceedings of the National Academy of
drug-resistant Pseudomonas aeruginosa in a French university Sciences of the USA 2006; 103: 2833-8.
hospital. European journal of clinical microbiology & infectious [214] Godfrey AJ, Hatlelid L, Bryan LE. Correlation between
diseases : official publication of the European Society of Clinical lipopolysaccharide structure and permeability resistance in -
Microbiology 2009; 28: 1217-22. lactam-resistant Pseudomonas aeruginosa. Antimicrobial Agents
[210] Alvarez-Ortega C, Wiegand I, Olivares J, Hancock RE, Martinez Chemotherap 1984; 26: 181-6.
JL. Genetic determinants involved in the susceptibility of [215] Leying HJ, Buscher KH, Cullmann W, Then RL.
Pseudomonas aeruginosa to -lactam antibiotics. Antimicrobial Lipopolysaccharide alterations responsible for combined quinolone
Agents Chemotherap 2010; 54: 4159-67. and -lactam resistance in Pseudomonas aeruginosa. Chemotherapy
[211] Dotsch A, Becker T, Pommerenke C, Magnowska Z, Jansch L, 1992; 38: 82-91.
Haussler S. Genomewide identification of genetic determinants of [216] Kropinski AM, Kuzio J, Angus BL, Hancock RE. Chemical and
antimicrobial drug resistance in Pseudomonas aeruginosa. chromatographic analysis of lipopolysaccharide from an antibiotic-
Antimicrob Agents Chemother 2009; 53: 2522-31. supersusceptible mutant of Pseudomonas aeruginosa. Antimicrob
Agents Chemother 1982; 21: 310-9.

Received: June 13, 2012 Accepted: August 2, 2012

You might also like