Antibacterial-Resistant Pseudomonas aeruginosa: Clinical Impact and Complex Regulation of Chromosomally Encoded Resistance Mechanisms

Size: px
Start display at page:

Download "Antibacterial-Resistant Pseudomonas aeruginosa: Clinical Impact and Complex Regulation of Chromosomally Encoded Resistance Mechanisms"

Transcription

1 CLINICAL MICROBIOLOGY REVIEWS, Oct. 2009, p Vol. 22, No /09/$ doi: /cmr Copyright 2009, American Society for Microbiology. All Rights Reserved. Antibacterial-Resistant Pseudomonas aeruginosa: Clinical Impact and Complex Regulation of Chromosomally Encoded Resistance Mechanisms Philip D. Lister, 1 * Daniel J. Wolter, 2 and Nancy D. Hanson 1 Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, Nebraska, 1 and Department of Pediatrics, University of Washington, Seattle, Washington 2 INTRODUCTION HISTORICAL AND CLINICAL SIGNIFICANCE OF P. AERUGINOSA RESISTANCE CHALLENGES FOR TREATMENT OF P. AERUGINOSA Antibacterial Resistance Trends Imported Resistance Mechanisms Chromosomally Encoded Resistance Mechanisms AmpC-MEDIATED RESISTANCE AmpC and Resistance to -Lactams Clinical Significance of AmpC Overproduction Pathways for AmpC Overproduction Factors Involved in Regulation of ampc Expression Mechanism of ampc Induction Mechanisms of ampc Derepression AmpD Homologues and Regulation of ampc Expression PBP4 and Regulation of ampc Expression OprD-MEDIATED RESISTANCE OprD and P. aeruginosa Susceptibility to Carbapenems Characterization of oprd Promoter Elements Molecular Mechanisms of OprD-Mediated Resistance Discordance between oprd Expression and Susceptibility to Imipenem EFFLUX-MEDIATED RESISTANCE MexAB-OprM Efflux Pump MexCD-OprJ Efflux Pump MexEF-OprN Efflux Pump MexXY Efflux Pump MexJK Efflux Pump Additional RND Efflux Pumps COREGULATION OF RESISTANCE MECHANISMS MexCD-OprJ Overproduction and Hypersusceptibility to Antibacterials Coregulation of MexEF-OprN and OprD PREVENTING EMERGENCE OF CHROMOSOMALLY ENCODED RESISTANCE CONCLUDING COMMENTS ACKNOWLEDGMENTS REFERENCES INTRODUCTION * Corresponding author. Mailing address: Department of Medical Microbiology and Immunology, Creighton University School of Medicine, 2500 California Plaza, Omaha, NE Phone: (402) Fax: (402) pdlister@creighton.edu. Infectious diseases have been an important cause of morbidity and mortality throughout our history. With the expansion of the antibiotic era during the 20th century, there was a growing confidence that the need for infectious disease specialists would all but disappear. However, no one could have predicted the impact that an increasing immunocompromised population would have on the resurgence of infectious diseases during the last 3 decades. Furthermore, the ability of bacterial pathogens to adapt and to overcome the challenges of antibiotics in their environment has been nothing short of impressive. We are now faced with a growing population of panresistant bacteria that threaten to move us into what some consider the postantibiotic era of infectious diseases. Some of the more problematic drug-resistant pathogens encountered today include methicillin-resistant Staphylococcus aureus, multidrug-resistant Streptococcus pneumoniae, and vancomycin-resistant Enterococcus spp. among the gram-positive bacteria and multidrug-resistant Acinetobacter baumannii, Klebsiella pneumoniae, Escherichia coli, and Pseudomonas aeruginosa among the gram-negative bacteria. This review focuses specifically on the resistance problems associated with P. aeruginosa, with a special emphasis on the complexity by which key chromosomally encoded resistance mechanisms are regulated and coregulated to make P. aeruginosa one of our greatest therapeutic challenges. 582

2 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 583 HISTORICAL AND CLINICAL SIGNIFICANCE OF P. AERUGINOSA The opportunistic bacterial pathogen currently known as P. aeruginosa has received several names throughout its history based on the characteristic blue-green coloration produced during culture. Sédillot in 1850 was first to observe that the discoloration of surgical wound dressings was associated with a transferable agent (196). The pigment responsible for the blue coloration was extracted by Fordos in 1860, and in 1862 Lucke was the first to associate this pigment with rod-shaped organisms (196). P. aeruginosa was not successfully isolated in pure culture until 1882, when Carle Gessard reported in a publication entitled On the Blue and Green Coloration of Bandages the growth of the organism from cutaneous wounds of two patients with bluish-green pus (65). In several additional reports between 1889 and 1894, P. aeruginosa (Bacillus pyocyaneus) was described as the causative agent of blue-green purulence in the wounds of patients (261). A more thorough presentation on the routes of invasion and dissemination of P. aeruginosa leading to acute or chronic infection was provided by Freeman in a 1916 article (56). P. aeruginosa is a ubiquitous organism present in many diverse environmental settings, and it can be isolated from various living sources, including plants, animals, and humans. The ability of P. aeruginosa to survive on minimal nutritional requirements and to tolerate a variety of physical conditions has allowed this organism to persist in both community and hospital settings. In the hospital, P. aeruginosa can be isolated from a variety of sources, including respiratory therapy equipment, antiseptics, soap, sinks, mops, medicines, and physiotherapy and hydrotherapy pools (199). Community reservoirs of this organism include swimming pools, whirlpools, hot tubs, contact lens solution, home humidifiers, soil and rhizosphere, and vegetables (77, 196, 199). P. aeruginosa is seldom a member of the normal microbial flora in humans. Representative colonization rates for specific sites in humans are 0 to 2% for skin, 0 to 3.3% for the nasal mucosa, 0 to 6.6% for the throat, and 2.6 to 24% for fecal samples (164). However, colonization rates may exceed 50% during hospitalization (199), especially among patients who have experienced trauma to or a breach in cutaneous or mucosal barriers by mechanical ventilation, tracheostomy, catheters, surgery, or severe burns (17, 49, 182, 252, 257). Patients with impaired immunity have higher risks for colonization by this organism (164, 199), and disruption in the normal microbial flora as a result of antimicrobial therapy has also been shown to increase colonization by P. aeruginosa (17, 18, 250). Despite the wide distribution of P. aeruginosa in nature and the potential for community-acquired infections, serious infections with P. aeruginosa are predominantly hospital acquired. A review of surveillance data collected by the CDC National Nosocomial Infections Surveillance System from 1986 to 1998 shows that P. aeruginosa was identified as the fifth most frequently isolated nosocomial pathogen, accounting for 9% of all hospital-acquired infections in the United States (48, 171). P. aeruginosa was also the second leading cause of nosocomial pneumonia (14 to 16%), third most common cause of urinary tract infections (7 to 11%), fourth most frequently isolated pathogen in surgical site infections (8%), and seventh leading contributor to bloodstream infections (2 to 6%). Data from more recent studies continue to show P. aeruginosa as the second most common cause of nosocomial pneumonia, health care-associated pneumonia, and ventilator-associated pneumonia (64, 106) and the leading cause of pneumonia among pediatric patients in the intensive care unit (ICU) (214). P. aeruginosa is especially problematic for seriously ill patients in ICUs. From 1992 to 1997, data from the National Nosocomial Infections Surveillance System showed that P. aeruginosa was responsible for 21% of pneumonias, 10% of urinary tract infections, 3% of bloodstream infections, and 13% of eye, ear, nose, and throat infections within ICUs in the United States (213). A similar study conducted in Europe identified P. aeruginosa as the second most frequently isolated organism in reported cases of ICU-acquired infections (242). In this surveillance study, P. aeruginosa was accountable for 30% of pneumonias, 19% of urinary tract infections, and 10% of bloodstream infections. RESISTANCE CHALLENGES FOR TREATMENT OF P. AERUGINOSA P. aeruginosa presents a serious therapeutic challenge for treatment of both community-acquired and nosocomial infections, and selection of the appropriate antibiotic to initiate therapy is essential to optimizing the clinical outcome (15, 156). Unfortunately, selection of the most appropriate antibiotic is complicated by the ability of P. aeruginosa to develop resistance to multiple classes of antibacterial agents, even during the course of treating an infection. Epidemiological outcome studies have shown that infections caused by drug-resistant P. aeruginosa are associated with significant increases in morbidity, mortality, need for surgical intervention, length of hospital stay and chronic care, and overall cost of treating the infection (7, 25, 62). Even more problematic is the development of resistance during the course of therapy, a complication which has been shown to double the length of hospitalization and overall cost of patient care (41). P. aeruginosa can develop resistance to antibacterials either through the acquisition of resistance genes on mobile genetic elements (i.e., plasmids) or through mutational processes that alter the expression and/or function of chromosomally encoded mechanisms. Both strategies for developing drug resistance can severely limit the therapeutic options for treatment of serious infections. Antibacterial Resistance Trends Presented in Table 1 are rates of P. aeruginosa resistance to several antipseudomonal drugs (54, 95, 99, 100, 178, 211, 212). This summary is not meant to be inclusive of all of the published literature, but rather highlights data reported for isolates from several U.S. surveillance studies since January If multiple years were included in a study, the resistance rates for the most recent year are presented in Table 1. P. aeruginosa exhibits the highest rates of resistance for the fluoroquinolones, with resistance to ciprofloxacin and levofloxacin ranging from 20 to 35%. Although not surprising, the highest rates were reported for isolates obtained from patients in ICUs (Table 1). P. aeruginosa isolates from ICU patients also trend toward higher rates of -lactam resistance than

3 584 LISTER ET AL. CLIN. MICROBIOL. REV. Antibiotic TABLE 1. Rates of antibacterial resistance among P. aeruginosa isolates from hospitals and ICUs Hospital study, 2006 (n 606) (211) Hospital study, 2005 (n 589) (212) Hospital study, 2002 (n 9,896) (54) % of strains exhibiting resistance a ICU study, 2002 (n 951) (178) ICU study, (n 7,500) (95) Hospital study, 2001 (n 2,157) (99) ICU study, 2001 (n 543) (99) Hospital study, 2000 (n 882) (100) -Lactams Cefepime Ceftazidime Piperacillin-tazobactam Aztreonam Imipenem Meropenem Fluoroquinolones Ciprofloxacin Levofloxacin Aminoglycosides Amikacin Tobramycin Gentamicin a Based upon CLSI interpretive breakpoints. general trends for hospitalized patients. Based on the data in Table 1, it is difficult to draw any strong conclusions about trends of resistance to various -lactams. Among the aminoglycosides, most studies have focused on gentamicin, with resistance rates ranging from 12 to 22%. Gentamicin was the least active of the aminoglycosides, with lower rates of resistance being reported for tobramycin and amikacin in most studies (Table 1). Although the resistance trends from large national surveillance studies provide important data for consideration, these studies do not address the potential for much higher rates of resistance within individual communities and hospitals. For example, during the years 2001 and 2006, rates of nonsusceptibility among P. aeruginosa isolates in Brooklyn, NY, ranged from 27 to 29% for cefepime, 30 to 31% for imipenem, 23% for meropenem, and 41 to 44% for ciprofloxacin (113). These rates are substantially higher than national trends focusing on all hospital isolates of P. aeruginosa (Table 1). Not only are rates of resistance to individual drugs or drug classes a concern, but the prevalence of multidrug-resistant strains (resistant to three or more drug classes) is an even more serious therapeutic challenge. A national surveillance of 13,999 nonduplicate P. aeruginosa isolates from ICU patients showed that multidrug resistance increased significantly, from 4% in 1993 to 14% in 2002 (Fig. 1A) (178). For comparison, another ICU surveillance study evaluated over 37,000 P. aeruginosa isolates from 1997 to 2002 and reported an increase in prevalence of multidrug-resistant strains from 13% to 21% (Fig. 1B) (132). Finally, Flamm et al. reported rates of multidrug-resistant P. aeruginosa ranging from 23 to 26% among 52,000 P. aeruginosa isolates collected in the United States from 1999 to 2002 (54). The highest prevalence of multidrug-resistant strains was observed among isolates from lower respiratory tract infections, whereas the lowest prevalence was observed among isolates from upper respiratory tract infections. Not surprisingly, multidrug-resistant strains were isolated more frequently from ICU and nursing home patients. A multidrug-resistant phenotype can arise in P. aeruginosa FIG. 1. Increasing prevalence of multidrug resistance among P. aeruginosa isolates from ICU patients in the United States. (A) Data for 13,999 nonduplicate isolates collected from 1993 to 2002 (178); (B) data for 37,390 isolates collected from 1997 to 2000 (132). Data represent the percentage of P. aeruginosa isolates that expressed a phenotype of multidrug resistance (resistance to three or more drug classes) during each year of the studies. (Panel A is adapted from reference 178 with permission; panel B is based on data from reference 132.)

4 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 585 through the acquisition of multiple imported resistance mechanisms on mobile genetic elements, a combination of imported and chromosomally encoded resistance mechanisms, accumulation of multiple chromosomal changes over time, and/or a single mutational event leading to the overexpression of a multidrug resistance mechanism, i.e., an efflux pump. This review focuses primarily on the key chromosomally encoded resistance mechanisms, their clinical significance, and their complex mechanisms of regulation. However, a brief overview of important imported resistance mechanisms is presented first. Imported Resistance Mechanisms In relation to the antipseudomonal drug classes presented in Table 1, imported resistance among P. aeruginosa isolates impacts the -lactams and aminoglycosides but not the fluoroquinolones. As discussed below, fluoroquinolone resistance among P. aeruginosa isolates has been linked only to chromosomal genes, with mutational changes in the fluoroquinolone targets DNA gyrase (gyra and gyrb) and/or topoisomerase IV (parc and pare) and/or overexpression of multidrug efflux pumps (Fig. 2). Although the plasmid-encoded DNA gyrase protection protein Qnr and the fluoroquinolone-modifying enzyme AAC(6 )Ib-cr can contribute to fluoroquinolone resistance among strains of Enterobacteriaceae (89, 186, 215), these two plasmid-encoded mechanisms have not been found in clinical isolates of P. aeruginosa (89, 198). Imported resistance to the -lactams involves the production of inactivating -lactamases, for which several families have been identified among clinical isolates of P. aeruginosa. The variety, prevalence, and clinical significance of imported -lactamases in P. aeruginosa have been addressed in several reviews over the last decade (74, ). The most common imported -lactamases found among P. aeruginosa isolates are penicillinases belonging to the molecular class A serine -lactamases (PSE, CARB, and TEM families). Within this group, enzymes belonging to the PSE family appear to be the most prevalent (14). The therapeutic impact of these penicillinases is relatively limited since they do not impact the clinical efficacy of extended-spectrum cephalosporins, monobactams, or carbapenems. Although less frequent, class A extended-spectrum -lactamases have also been detected in strains of P. aeruginosa and have included enzymes from the TEM, SHV, CTX-M, PER, VEB, GES, and IBC families (27, 36, 155, 175, 192, 197, 268, 284). Extended-spectrum -lactamases from the class D, OXA-type enzymes have also been encountered within P. aeruginosa (170, 191). Similar to the case for the Enterobacteriaceae, extendedspectrum -lactamases alone do not provide P. aeruginosa resistance to the carbapenems. However, the prevalence of different classes of carbapenem-hydrolyzing enzymes has been increasing globally. The first class B metallo- -lactamases in P. aeruginosa were identified in 1991 in Japan (266). Since that initial report, metallo- -lactamases have been reported for P. aeruginosa isolates from nearly all regions of the globe (57, 96, 117, 176, 187, 281), and four major families have been identified (IMP, VIM, SPM, and GIM families) (28, 59, 135, 176, 253). Recently, class A carbapenemases of the KPC family have been identified. The first characterized KPC-producing P. aeruginosa isolate was collected in Colombia and reported in 2007 (262). The most recent report identifies the spread of KPC genes into clonally related and unrelated strains of P. aeruginosa from Puerto Rico (281). The first identification of an imported OXA-type carbapenemase in P. aeruginosa was reported in 2008, and it was shown to be the same OXA-40 carbapenemase previously described for A. baumannii (237). Imported resistance to aminoglycosides most commonly involves enzymatic inactivation of the drug molecule through chemical modification. The history, molecular characterization, prevalence, and clinical significance of aminoglycosideinactivating enzymes in P. aeruginosa were recently covered in an excellent review (200). These enzymes are categorized into the following three families, based upon the chemical modification they mediate: (i) aminoglycoside phosphoryltransferase enzymes phosphorylate the drug molecule, (ii) aminoglycoside acetyltransferase enzymes acetylate the drug molecule, and (iii) aminoglycoside nucleotidyltransferase enzymes adenylate the drug molecule. Although the range of aminoglycosides inactivated by specific enzymes within this family can differ, the ability of P. aeruginosa to carry the genes for multiple aminoglycoside-inactivating enzymes provides individual strains with the potential to develop resistance to all aminoglycosides. In addition to the variety of aminoglycoside-modifying enzymes, high-level resistance to multiple aminoglycosides can be associated with methylation of the 16S rrna. This mechanism was first reported for P. aeruginosa in 1993, and the methylaseencoding gene was designated rmta (287). There are currently five characterized ribosomal methyltransferase enzymes (RmtA, RmtB, RmtC, RmtD, and ArmA) that have been found worldwide among clinical isolates of P. aeruginosa and Enterobacteriaceae (42, 60, 61, 265, 286, 287). Although methyltransferases do not appear to be as common as the aminoglycoside-modifying enzymes, all of the genes encoding these enzymes have been associated with mobile genetic elements (43, 61, 68, 264, 265, 285), raising concern about their widespread dissemination among P. aeruginosa isolates and other gram-negative bacilli. Chromosomally Encoded Resistance Mechanisms Similar to imported resistance mechanisms, there are a variety of resistance mechanisms encoded on the P. aeruginosa chromosome. These mechanisms include several aminoglycoside-inactivating enzymes (200) and a class D oxacillinase, OXA-50 (67). As mentioned above, characterized mechanisms of fluoroquinolone resistance among P. aeruginosa isolates have been restricted to chromosomal genes, including target mutations and active efflux (Fig. 2). Similar to the case for other gram-negative bacteria, DNA gyrase is the primary target for the fluoroquinolones in P. aeruginosa (84). Therefore, the first target-specific mutations are typically observed within the quinolone resistance determining region (QRDR) of gyra (6, 116, 165, 249, 283). The highest levels of resistance are observed in strains that have mutations in the QRDR of both gyra and the topoisomerase IV gene parc (6, 90, 91, 116, 165). Although mutational changes within the other two genes, gyrb and pare, have been described, the prevalence of these mutations appears to be much lower (6). Recent studies involving

5 586 LISTER ET AL. CLIN. MICROBIOL. REV.

6 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 587 screening of the Harvard PA14 library of P. aeruginosa mutants have identified a number of other chromosomal genes that may be involved in antibacterial resistance or in increasing the frequency of mutation to resistance (20, 44, 233, 271). The three most intensely studied chromosomally encoded resistance mechanisms in P. aeruginosa are the AmpC cephalosporinase, the OprD outer membrane porin, and the multidrug efflux pumps (Fig. 2). The remainder of this review focuses specifically on the clinical relevance of these three resistance mechanisms and the complex pathways P. aeruginosa utilizes to regulate their expression. The ability of P. aeruginosa to coregulate different resistance mechanisms makes this pathogen a constantly moving target that continues to challenge therapeutic strategies. AmpC-MEDIATED RESISTANCE AmpC and Resistance to -Lactams P. aeruginosa carries an inducible AmpC cephalosporinase which is similar to the chromosomally encoded AmpC found in several members of the Enterobacteriaceae (23, 34, 76, 222, 227). Wild-type strains of P. aeruginosa produce only low basal levels of AmpC and are susceptible to the antipseudomonal penicillins, penicillin-inhibitor combinations, cephalosporins, and carbapenems (227). However, when AmpC production is significantly increased, P. aeruginosa develops resistance to all -lactams, with the exception of the carbapenems, to be discussed later (34, 227). This is in contrast to some AmpC-overproducing members of the Enterobacteriaceae, which remain susceptible to cefepime and require additional mechanisms to develop cefepime resistance (i.e., downregulation of porin production) (13, 45, 78, 194, 223). Although it is possible that variability in the hydrolytic activity of AmpCs from P. aeruginosa and the Enterobacteriaceae could play a role in these differences, cefepime hydrolysis data obtained with purified AmpC enzymes do not support this hypothesis (208). Rather, the greater impermeability of the P. aeruginosa outer membrane may play an important role in allowing AmpC overproduction to push cefepime MICs above the resistance breakpoint (75). Whereas resistance to most of the -lactams emerges as a result of AmpC overproduction, a definitive relationship between P. aeruginosa AmpC and carbapenem susceptibility remains convoluted. AmpC-deficient strains of P. aeruginosa created through allelic exchange exhibit significant, 4-fold increases in susceptibility to imipenem and panipenem but not to meropenem (148). Additional P. aeruginosa isolates, defined as AmpC deficient yet still producing AmpC, also exhibit increased susceptibility to imipenem and doripenem but not to meropenem (131, 136, 169). These data suggest that AmpC may play a role in the level of intrinsic susceptibility of P. aeruginosa to carbapenems. In contrast, published data have suggested that overproduction of AmpC does not play a discernible role in the development of carbapenem resistance among P. aeruginosa isolates. AmpC overproduction among isogenic mutants selected with -lactams does not significantly decrease P. aeruginosa susceptibility to carbapenems (63, 131, 169, 274). In addition, AmpC overproduction in carbapenemsusceptible clinical isolates of P. aeruginosa has been reported. Data from one study of 47 characterized AmpC-overproducing clinical isolates showed that only 7 (15%) were resistant to imipenem (calculated from the data in Table 2 of reference 225). Gutierrez et al. recently reported that 51% of carbapenem-resistant clinical isolates of P. aeruginosa in their study overproduced AmpC (73). Although statistical analysis suggested that meropenem-resistant strains were more likely to overproduce AmpC than meropenem-susceptible strains, Gutierrez et al. concluded that AmpC hyperproduction is neither sufficient nor necessary for meropenem clinical resistance. The challenge of specifically linking AmpC overproduction to carbapenem resistance is our own limited knowledge of the complex interplay between resistance mechanisms in P. aeruginosa and the multitude of pathways by which P. aeruginosa coregulates resistance mechanisms. How do we specifically link AmpC overproduction to carbapenem resistance among uncontrolled clinical isolates with diverse genetic and environmental backgrounds? Even with isogenic laboratory strains, how do we specifically link AmpC production to a particular phenotype, knowing that altered AmpC production could be accompanied by changes in the expression of other resistance mechanisms through coregulatory pathways? Despite how much we have yet to learn about the resistance potential of P. aeruginosa, the data generated thus far suggest that AmpC overproduction alone does not significantly alter P. aeruginosa s susceptibility to the carbapenems but could certainly contribute to resistance if accompanied by additional resistance mechanisms (e.g., efflux pump overproduction, decreased OprD, and/or production of a class A/class B carbapenemase). Adding even more complexity is the potential for mutational variants of the chromosomally encoded AmpC enzyme (extended-spectrum AmpC) to provide P. aeruginosa with carbapenem resistance (216). Extended-spectrum AmpCs were first identified in Serratia marcescens and Enterobacter spp. (10, 143, 153) and, most recently, in E. coli (141, 142). Amino acid modifications near the active sites of these enzymes can lead to FIG. 2. Mutational resistance to fluoroquinolones and carbapenems involving chromosomally encoded mechanisms expressed by P. aeruginosa. (A) Interactions of fluoroquinolones and carbapenems with wild-type susceptible P. aeruginosa expressing basal levels of AmpC, OprD, and nonmutated fluoroquinolone target genes (gyra, gyrb, parc, and pare). Fluoroquinolone molecules pass through the outer membrane, peptidoglycan, periplasmic space, and cytoplasmic membrane and interact with DNA gyrase and topoisomerase IV (Topo IV) targets in the cytoplasm when these enzymes are complexed with DNA. Carbapenem molecules pass through the outer membrane-specific porin OprD and interact with their target PBPs, located on the outside of the cytoplasmic membrane. (B) Chromosomally encoded mechanisms of resistance to fluoroquinolones and carbapenems. Fluoroquinolone resistance is mediated by (i) overexpression of RND efflux pumps extruding the drug molecules from the periplasmic and cytoplasmic spaces and/or (ii) mutational changes within the target genes. Locations of the QRDRs within target genes are highlighted in yellow. Carbapenem resistance is mediated primarily by (i) decreased production or loss of functional OprD in the outer membrane and/or (ii) overproduction of RND efflux pumps (with the exception of imipenem). Minor changes in susceptibility can be observed due to overexpression of AmpC, adding to the resistance potential.

7 588 LISTER ET AL. CLIN. MICROBIOL. REV.

8 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 589 FIG. 3. Mechanisms involved in regulation of ampc expression. These figures represent the current knowledge obtained from studies with members of the Enterobacteriaceae and appear to parallel events in P. aeruginosa. (A) Wild-type basal expression of ampc. During normal cell wall recycling, 1,6-anhydromuropeptides are removed from the cell wall and transported into the cytoplasm via the AmpG permease. The 1,6- anhydromuropeptides are cleaved by AmpD to generate free tripeptides, which are later converted into UDP-MurNAc-pentapeptides. UDP- MurNAc-pentapeptide interacts with AmpR bound to the ampr-ampc intergenic region, creating a conformation that represses transcription of ampc. Low basal levels of AmpC are produced, and the enzyme is localized to the periplasmic space. (B) -Lactam induction of ampc expression. Inducing -lactams, such as cefoxitin and imipenem, cross the outer membrane through porins, enter the periplasmic space, and interact with target PBPs. An increase in pools of 1,6-anhydromuropeptides is observed, and AmpD is unable to efficiently process the higher levels of cell wall fragments. The anhydro-murnac-peptides (inducing peptides) replace UDP-MurNAc-pentapeptides (suppressing peptides) bound to AmpR, causing a conformational change in the protein. AmpR is converted into a transcriptional activator, ampc is expressed at higher levels, and levels of AmpC increase in the periplasmic space. When the amount of -lactam decreases below inducing levels, the cytoplasmic pool of anhydro- MurNAc-peptides also decreases, and AmpD is able to efficiently cleave these peptides, restoring wild-type ampc expression, as shown in panel A. (C) AmpD-associated derepression of ampc expression. Mutations leading to the inactivation of AmpD or decreased expression of ampd impair the processing of cell wall recycled products and lead to increased levels of anhydro-murnac-peptides (inducing peptides) in the cytoplasm. As a result, the binding of inducing peptides to AmpR is favored, AmpR is locked in a conformation for transcriptional activation of ampc expression, and high-level constitutive expression of ampc is observed. increased hydrolytic activity against cefepime, ceftazidime, and imipenem. However, increased catalytic activity of these enzymes only reduces susceptibility to cefepime and imipenem. Overproduction of these extended-spectrum AmpCs seems to be a requirement for cefepime (10) and/or carbapenem (216) resistance. Clinical Significance of AmpC Overproduction Similar to the discussion above, the clinical impact of AmpC overproduction by P. aeruginosa is difficult to assess due to the complex interplay of multiple resistance mechanisms. Nevertheless, Tam et al. reported that patients were 67.5 times more likely to be given inappropriate antibiotics when the infections were caused by AmpC-overproducing P. aeruginosa than when they were caused by P. aeruginosa that did not overproduce AmpC (251). In addition, these patients were more likely to have persistent bacteremia (45% versus 6%), underscoring the need to rapidly identify AmpC-overproducing P. aeruginosa isolates in the clinical laboratory and to understand the mechanisms of AmpC overproduction in this pathogen. The ability of resistant P. aeruginosa to emerge during the course of therapy presents an even greater challenge. In this scenario, patients are treated with an appropriate -lactam based on initial susceptibility data, only to fail therapy due to the emergence of AmpC-mediated resistance. This phenomenon has been observed in 14 to 56% of patients treated with antipseudomonal penicillins, penicillin-inhibitor combinations, aztreonam, and extended-spectrum cephalosporins (50, 71, 94, 97, 118, 147, 226, 228, 235), and emergence of resistance/ clinical failure is observed most frequently with infections outside the urinary tract and in patients with underlying cystic fibrosis and neutropenia. Although combining an aminoglyco-

9 590 LISTER ET AL. CLIN. MICROBIOL. REV. side with an antipseudomonal -lactam is one strategy for preventing the emergence of AmpC-mediated resistance, this combination is not always effective in achieving that goal (97, 118, 147, 226, 228, 235). As an initial step toward the future identification of novel strategies for combating AmpC-mediated resistance, it is essential that we elucidate the mechanisms by which the expression of this resistance mechanism is regulated. The following sections review the current understanding of mechanisms involved in regulation of AmpC-mediated resistance among P. aeruginosa isolates and, where appropriate, compare and contrast them with mechanisms of AmpC regulation among Enterobacteriaceae. Pathways for AmpC Overproduction Overproduction of the chromosomally encoded AmpC enzyme in P. aeruginosa and some members of the Enterobacteriaceae can occur either by induction of the ampc gene or through a process of derepression leading to constitutive high-level expression. Overexpression through the induction pathway occurs during exposure to specific -lactams and -lactamase inhibitors (e.g., cefoxitin, imipenem, and clavulanate) (112, 123, 128, 247, 267), but the process is reversible after removal of the inducing agent. In contrast, AmpC derepression occurs when proteins involved in the induction pathway are compromised through chromosomal mutations (83, 97, 108, 111, 114, 124) and the cephalosporinase is constitutively produced at an elevated level, even in the absence of an inducing -lactam (9, 83, 114, 115). As discussed later in this section, phenotypes of AmpC derepression are more diverse among P. aeruginosa isolates than among the Enterobacteriaceae. Strains of P. aeruginosa can transition through a phenotype of partial derepession before achieving full derepression of AmpC. Factors Involved in Regulation of ampc Expression Figure 3 illustrates the key components involved in the regulation of ampc expression. The ampc induction pathway involves the following three major gene products: (i) an inner membrane permease known as AmpG; (ii) a cytosolic amidase, AmpD; and (iii) a transcription factor, AmpR, belonging to the LysR family of regulatory proteins (11, 138, 139). These three proteins are required for induction of the ampc gene in both Enterobacteriaceae and P. aeruginosa, although there is no direct evidence linking AmpG to the ampc induction pathway of P. aeruginosa (11, 81, 88, 109, 110, 127, 138, 139). The P. aeruginosa ampc and ampr genes and their corresponding intergenic region were first described in 1990 (139), and the gene organization is identical to that for members of the Enterobacteriaceae with inducible ampc genes. In both P. aeruginosa and Enterobacteriaceae, the ampr and ampc genes are divergently transcribed, and the binding of AmpR to the intergenic region between ampc and ampr is required for ampc induction (Fig. 3) (12, 16, 86, 126, 138, 139, 217). In the Enterobacteriaceae, AmpR negatively regulates ampc expression as well as the expression of its own gene, ampr (125, 126). In studies with P. aeruginosa, the data have been more conflicting and difficult to compare. Using an ampr knockout of P. aeruginosa PAO1, Kong et al. reported a 12-fold increase in nitrocefin hydrolysis in the absence of AmpR (107). However, interpretation of these data was complicated by the presence of another chromosomal -lactamase, PoxB, which also hydrolyzes nitrocefin and appears to be regulated by AmpR (107). Further experiments with a P ampc -lacz promoter gene construct inserted into the chromosome of P. aeruginosa suggested that AmpR does not negatively regulate ampc (107). More recently, however, Moya et al. concluded that AmpR is a negative regulator of ampc, based upon their more direct analysis of ampc expression in an ampr knockout of P. aeruginosa PAO1 (166). Recent reporter gene studies have also suggested that P. aeruginosa AmpR is a global regulator affecting the expression of multiple genes (lasb, rhlr, poxb, lasa, lasi, and lasr) in addition to ampc (107). Although direct RNA expression experiments are needed to confirm a global regulatory function, these data raise the possibility that regulation by AmpR may be more complex in P. aeruginosa than what has been shown for members of the Enterobacteriaceae (107). Mechanism of ampc Induction Whether ampc is expressed at a low constitutive level or elevated through induction is dependent upon the type of cofactor (i.e., cell wall precursor peptide) that binds to AmpR (Fig. 3). In this respect, the AmpC regulatory pathway is intimately linked to the cell wall recycling pathway via AmpD and AmpR. The general mechanism for regulation of low basal ampc expression in a wild-type P. aeruginosa strain is extrapolated from data obtained from the Enterobacteriaceae and shown in Fig. 3A. During normal growth, cell wall synthesis requires the addition and subtraction of cell wall components, resulting in the release of muropeptides (39, 40, 69, 80, 87). These muropeptides are transported into the cell via the AmpG permease (30, 87). Once inside the cell, these muropeptides are modified by AmpD into free peptide and anhydromuramic acid, with the resulting peptide recycled back into the cell wall synthesis pathway (80, 81, 87, 88). During peptide processing for use in cell wall synthesis, UDP is added to the pentapeptide (80). Excess repressing UDP-pentapeptide binds to AmpR and keeps AmpR in a conformational state that does not allow efficient transcription from the ampc promoter (86). It has been hypothesized that this conformation of AmpR prevents RNA polymerase from interacting efficiently with the ampc promoter, resulting in a low basal level of ampc expression. The process of AmpC induction requires binding of an inducing -lactam or -lactamase inhibitor (e.g., cefoxitin, imipenem, or clavulanate) to penicillin binding proteins (PBPs) (Fig. 3B) (183, 190, 224). Since induction of AmpC does not result from the interactions of all -lactams with PBPs, there clearly is something specific about the interactions of inducing compounds such as cefoxitin, imipenem, and clavulanate. Studies with the Enterobacteriaceae have shown that inducing -lactams have a higher affinity for the low-molecular-weight PBPs (72, 82, 177), and genetic knockout of the genes for these PBPs provides additional support for their role (183, 190, 224). However, these studies were performed in E. coli, which does not possess an intrinsic inducible ampc system, making the data

10 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 591 difficult to interpret (1). Data from a recently published study with P. aeruginosa have demonstrated that loss of the nonessential low-molecular-weight protein PBP4 is associated with increased expression of ampc and partial derepression of ampc (166). Although these experiments clearly demonstrated an association between PBP4 and derepression of ampc (described in more detail later in the review), the authors concluded from their study that PBP4 plays a role in ampc induction as well. However, strains lacking functional PBP4 still demonstrated induction of ampc, suggesting that PBP4 is not essential for the induction pathway (166). The role of other low-molecular-weight PBPs was not addressed in this study and should still be evaluated to determine their selective role in induction of ampc by inducing -lactams. Regardless of the precise mechanism responsible for selective AmpC induction with specific -lactams, the result is an increase in the concentration of inducing muropeptides compared to the amount of repressing UDP-pentapeptide found in the cytoplasm (39, 40, 87). The repressing UDP-pentapeptide bound to AmpR is believed to be replaced by the inducing muropeptide form, changing the conformation of AmpR as it binds to the ampc promoter (76, 86). This conformational change has been suggested to provide a more efficient interaction with RNA polymerase, resulting in a significant increase in ampc expression (86). When the inducing -lactam or -lactamase inhibitor is removed, normal cell wall synthesis and cell wall recycling are restored. The result is a restoration of basal ampc expression through the replacement of the AmpRbound inducing muropeptide with the repressing UDPpentapeptide that is now present at a higher concentration. Mechanisms of ampc Derepression Modification of any protein involved in the induction pathway or modification of the ampc promoter could conceivably lead to derepression of ampc expression. Although studies with the Enterobacteriaceae have identified mutations within ampr (111), the majority of changes observed in clinical isolates have been associated with ampd (46, 83, 108, 231, 246). AmpD-associated derepression has been linked to mutations within the ampd structural gene and mutations leading to decreased ampd expression (231). In these strains, the AmpD amidase that cleaves the muropeptides entering the cell during cell wall synthesis has been modified or decreased, and normal processing of the muropeptides would be compromised (Fig. 3C). As a result, the concentration of inducing muropeptides in the cytoplasm would permanently increase, which favors their binding to AmpR due to the stoichiometric effect described above for the pathway of induction (39, 40, 86, 87). This culminates in a constitutive elevation of ampc expression. Paralleling what is observed for the Enterobacteriaceae, P. aeruginosa AmpD has been characterized as a negative regulator of AmpC (114), and ampd mutations are an important mechanism of ampc derepression (9, 97, 98, 115, 232). Although not as common as ampd mutations, the potential role of mutated AmpR in P. aeruginosa derepression of AmpC has been described and correlated with a similar mutation in Enterobacter cloacae (9, 111, 232). In contrast to the case for the Enterobacteriaceae, full derepression of AmpC in P. aeruginosa is not always a single-step process. Campbell et al. have described the following three phenotypes of ampc expression: (i) low-basal-level expression that is inducible (wild type), (ii) moderate-basal-level expression that is inducible (partial derepression), and (iii) high-basal-level expression that is constitutive (full derepression) (23). A further complexity is that some AmpC-overproducing P. aeruginosa strains do not exhibit mutations in either ampr, ampd, or the ampr-ampc intergenic region and do not exhibit changes in the level of ampd expression (9, 23, 97, 232, 282). In one of these studies, mutants that expressed significant increases in the basal level of ampc were selected from a partially derepressed P. aeruginosa strain that lacked a functional AmpD protein (282). The mechanism of increased ampc expression did not involve additional mutations in ampr, ampd, or the ampr-ampc intergenic region or changes in ampd expression. However, a functional ampd gene was able to completely transcomplement and restore the wild-type phenotype for ampc expression and ceftazidime susceptibility (282). These observations suggest that undiscovered factors or pathways likely contribute to the regulation of ampc and the derepressed phenotype. Additional candidates for regulation of ampc expression include AmpE, homologues of AmpD, and PBP4. In P. aeruginosa and the Enterobacteriaceae, expression of ampd is linked to the ampe gene (83, 114). Although some publications have indicated a potential role for AmpE in the AmpC regulatory pathway (83, 98), a specific role has yet to be established. Other studies suggest that AmpE plays no role in the regulation of AmpC (114, 166). AmpD Homologues and Regulation of ampc Expression The entire genome of P. aeruginosa PAO1 has been sequenced, opening the door for further investigations into the mechanisms involved in the regulation of ampc expression. In 2006, Juan et al. reported the identification of two additional P. aeruginosa AmpD homologues (AmpDh2 and AmpDh3) (98). E. coli possesses an outer membrane lipoprotein designated AmiD that was suggested to be a homologue of AmpDh2 by the same investigators (167), and this was later confirmed by Schmidtke and Hanson (232). There are only two N-acetylanhydro-muramyl-L-alanine amidases in the E. coli genome, so no homologue for AmpDh3 has been determined. Although the level of identity between AmpDh2 or AmpDh3 and the original AmpD protein is only 25 to 27% at the amino acid level, this may be attributed to the fact that the two new homologues possess membrane-spanning tails not observed with AmpD (232). However, AmpDh2 and AmpDh3 retain necessary conserved sequences within the active site compared with AmpD and are able to transcomplement an AmpD-deficient strain of P. aeruginosa PAO1 (98). Using a series of ampd homologue knockout mutants, Juan et al. investigated the relative impact of each homologue on expression of ampc (98). Their observations are summarized in Table 2. The wild-type laboratory strain P. aeruginosa PAO1 served as the parent strain in this study and exhibited the expected low-basal-level inducible phenotype. The loss of ampd from P. aeruginosa PAO1 changed the phenotype to moderate-basal-level inducible expression of ampc (partial derepression). Surprisingly, deletion of ampdh2 or ampdh3 alone or in combination with each other did not alter the

11 592 LISTER ET AL. CLIN. MICROBIOL. REV. TABLE 2. Genetic knockout of ampd homologue genes and ampc expression in P. aeruginosa PAO1 a Strain Avg level of ampc expression SD b Basal low-basal-level inducible phenotype. Although deletion of ampd in combination with deletion of ampdh2 or ampdh3 did not alter the partially derepressed phenotype observed with ampd deletion alone, the double knockout of ampd and ampdh3 did exhibit significant increases in both basal and cefoxitin-induced ampc expression. Finally, full derepression to high-basal-level, constitutive expression of ampc required the combined deletion of all three homologues. From these data, Juan et al. concluded that AmpD exhibits the greatest influence on ampc expression, followed by AmpDh3 and then AmpDh2. The discovery and characterization of the ampd homologues, as well as their coordinated regulation of ampc expression, were important contributions to understanding the complex regulation of ampc expression in this pathogen. Not surprisingly, data from the analysis of clinical isolates suggest the use of additional pathways for regulation of ampc. For example, the same researchers analyzed 10 clinical isolates of P. aeruginosa that overproduced AmpC and found that 4 of the isolates did not exhibit any mutations in ampd (97), suggesting that loss of ampd is not an absolute requirement for ampc derepression in P. aeruginosa. Although potential decreases in ampd homologue expression were not assessed, ampc derepression in these four strains was later shown to be associated with mutations in the nonessential low-molecular-weight PBP4 gene, dacb (see below) (166). Schmidtke and Hanson reported that three clinical isolates of P. aeruginosa exhibiting full derepression of ampc (constitutive high-basal-level expression) exhibited inactivating mutations within ampd only, with no mutations or changes in the expression of ampdh2 or ampdh3 (232). Although the potential role of PBP4 was not evaluated by Schmidtke and Hanson, full derepression of ampc has been observed in a spontaneous dacb (PBP4) mutant of P. aeruginosa PAO1 with subsequent knockout of ampd (166). PBP4 and Regulation of ampc Expression Induced PAO1 1 c 43 9 PA D PA Dh2 1 c PA Dh3 1 c 55 5 PA Dh2Dh PA DDh PA DDh , PA DDh2Dh3 1, , a Modified from reference 91 with permission. b Real-time reverse transcription-pcr was used to measure ampc expression in P. aeruginosa grown in the presence of 50 g/ml of cefoxitin (induced) or the absence of cefoxitin (basal). c Actual ampc expression data were not presented in the original report (91). Based on the methodology described and interpretation of the data presented, basal ampc expression was set to a value of 1 in this table. PBP4 was recently identified as an important component of ampc regulation (166). Genetic knockout of dacb, which encodes PBP4, results in a partially derepressed phenotype in P. aeruginosa PAO1, similar to that of a knockout mutant of ampd (166). Furthermore, PBP4 mutations have also been associated with derepression of ampc in clinical isolates, and the association appears to be at least as common as mutations in ampd (166). PBP4-associated derepression of ampc is also associated with the two-component global regulator CreBC, which is a homologue of the Aeromonas sp. regulator BrlAB (8, 174). Laboratory mutants and clinical isolates that exhibit PBP4- associated derepression of ampc also exhibit a significant increase in the expression of cred (166), a gene regulated by CreBC. However, the protein encoded by cred has not been characterized, and its role in ampc regulation is questionable. Elimination of cred in PBP4-associated derepressed mutants did not significantly alter -lactam susceptibility (166). In contrast, the knockout of crebc significantly increased susceptibility to penicillins, cephalosporins, and aztreonam in ampcderepressed dacb mutants. This link between CreBC and ampc regulation appears to be specific for PBP4, as no correlation was noted for ampd-associated derepressed mutants. Perhaps the most interesting observation is that increased -lactam susceptibility was not associated with any changes in the level of ampc expression, suggesting that CreBC enhances resistance through a pathway other than AmpC. Each of the studies discussed above provides important information on how P. aeruginosa regulates expression of ampc. However, it is clear that we have much to learn about the complex mechanisms by which AmpR, AmpD homologues, and PBP4 interact to control this important resistance mechanism. Furthermore, the pathway that P. aeruginosa uses to selectively induce ampc in the presence of -lactams such as cefoxitin and imipenem remains uncharacterized, despite our advances in understanding the steps to derepression. Of particular interest would be further investigation into other lowmolecular-weight PBPs and their role as initial triggers for ampc induction. A more complete understanding of the factors and complex pathways regulating ampc expression could lead to the identification of potential targets for controlling AmpC-mediated resistance and preserving the antibacterial activity of the -lactam class. OprD-MEDIATED RESISTANCE The outer membrane of gram-negative bacteria constitutes a semipermeable barrier that slows the penetration of antibiotics, and specific to this review, the outer membrane of P. aeruginosa is only 8% as permeable as the outer membrane of Escherichia coli (75). However, in order to survive, P. aeruginosa must allow the passage of nutrients into the cell, and this is accomplished through a collection of water-filled protein channels called porins. Sequence analysis of the P. aeruginosa genome has identified 163 known or predicted outer membrane proteins, with 64 of these outer membrane proteins grouped into three families of porins (75). These porins play an important physiological role in the transport of sugars, amino acids, phosphates, divalent cations, and siderophores (75). Certain hydrophilic antibiotics, such as -lactams, aminoglycosides, tetracyclines, and some fluoroquinolones, have also been shown to transverse the outer membrane through porin channels (173, 289). Not surprisingly, the loss of specific porin channels can decrease the susceptibility of P. aeruginosa to

12 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 593 certain antibacterial agents. This section of the review focuses specifically on the association of the outer membrane porin OprD and the susceptibility of P. aeruginosa to carbapenem antibiotics. OprD and P. aeruginosa Susceptibility to Carbapenems The P. aeruginosa porin OprD is a substrate-specific porin that has been shown to facilitate the diffusion of basic amino acids, small peptides that contain these amino acids, and carbapenems into the cell (254, 255). This aqueous porin shares close homology to the nonspecific porin OmpF in E. coli. Pirnay et al. evaluated oprd genes from 55 strains of P. aeruginosa (environmental and clinical) and found evidence that the genetic sequence of oprd and the amino acid sequence of OprD are diverse across individual strains (195). DNA sequence identities ranged from 91 to 93%, whereas amino acid sequence identities varied from 88 to 93%. There was evidence of intraspecies recombinational events. OprD appears to serve as the preferred portal of entry for the carbapenems, and loss of OprD from the outer membrane significantly decreases the susceptibility of P. aeruginosa to available carbapenems (103, 132, 140, 144, 169, 209, 210, 221, 229, 254). However, data presented later in this section highlight how much we have yet to learn about the dynamic interactions of carbapenems with P. aeruginosa. The impact of OprD-mediated resistance on the carbapenems can be analyzed in two ways. The first consideration is the relative impact on the antibacterial potency of the carbapenems, as measured by increases in MICs. Data from a recent study of isogenic wild-type and OprD-deficient mutant pairs demonstrated that the loss of OprD decreases the susceptibility of P. aeruginosa to meropenem 4- to 32-fold, compared to 4- to 16-fold for imipenem and 8- to 32-fold for doripenem (221). For several OprD-deficient mutants in this study, the impact on meropenem potency was greater than that on the potency of other carbapenems. These data appear to conflict with the conclusions of Perez et al., who suggested that meropenem utilizes alternative pathways for entry across the outer membrane of P. aeruginosa (188). However, Perez et al. did not evaluate isogenic mutant pairs in their analysis of OprD deficiency and meropenem susceptibility. Rather, these investigators focused their study on unrelated clinical isolates that exhibited a phenotype of imipenem resistance and meropenem susceptibility. Although the P. aeruginosa isolates were susceptible to meropenem, MICs ranged from 2 to 4 g/ml, well above expected meropenem MICs against wild-type strains (21, 256). Therefore, it is likely that decreased OprD in the clinical isolates was in fact responsible for elevating meropenem MICs to the susceptible breakpoint. The second aspect of OprD-mediated resistance to consider is the clinical impact on the carbapenems. Although loss of OprD may impact susceptibility to imipenem less than that to meropenem (based on changes in MICs), this resistance mechanism frequently pushes MICs of imipenem above the resistance breakpoint. For example, in a study by Sakyo et al., all 10 OprD-deficient mutants lost susceptibility to imipenem, with MICs of 4 g/ml (221). This is not surprising, since the MIC 50 for imipenem against P. aeruginosa is already 1 g/ml (21, 256), requiring only an eightfold decrease in potency to push the MIC into the intermediate range. In contrast to the case for imipenem, MICs for meropenem remained below 4 g/ml for 4 of 10 mutants, and MICs for doripenem remained below 4 g/ml for 8 of 10 mutants (221). Meropenem and doripenem exhibit an intrinsic potency that is fourfold greater than that of imipenem (21, 256). Therefore, the impact of OprD deficiency on the potency of these carbapenems does not always push the MICs above the susceptible breakpoint, and additional resistance mechanisms (efflux pump and/or carbapenemase) may be required to provide resistance to these two carbapenems. The following sections review the current understanding of how P. aeruginosa regulates the expression of oprd. Published data to this point have identified mechanisms influencing the transcriptional expression of oprd and the translation of a functional porin protein. Characterization of oprd Promoter Elements Although the relationship between OprD and resistance to the carbapenems has been recognized for 2 decades, characterization of the oprd promoter and understanding of the regulation of OprD remain limited compared to our understanding of AmpC- and efflux-mediated resistance mechanisms. Figure 4 depicts the key promoter elements for oprd, based upon 5 rapid amplification of cdna ends of the oprd promoter of P. aeruginosa PAO1 grown in Mueller-Hinton broth (278). In these studies, two start sites for oprd transcription were identified, with transcription initiating at similar frequencies from an adenine 71 bases and a thymine 23 bases upstream of the translational start codon for oprd. Putative 10 and 35 elements are also presented in Fig. 4. The transcriptional start sites and putative promoter elements observed for P. aeruginosa grown in Mueller-Hinton broth are different from those described by Ochs et al. (179). When P. aeruginosa was cultured with arginine, histidine, glutamate, or alanine as the sole source of carbon and with ammonium sulfate as the nitrogen source, the primary start site of transcription was reported to be a thymine located 89 bases upstream of the oprd translational start codon. Furthermore, induction of oprd expression by arginine was shown to be dependent on the arginine-responsive regulatory protein ArgR, which binds to an operator in the promoter region of oprd (179). In contrast, induction with the amino acid glutamate was independent of ArgR activation. Therefore, it appears that P. aeruginosa utilizes multiple transcriptional start sites and mechanisms to regulate expression of oprd, depending upon the growth conditions encountered. Molecular Mechanisms of OprD-Mediated Resistance The pathway to OprD-mediated resistance can involve mechanisms that decrease the transcriptional expression of oprd and/or mutations that disrupt the translational production of a functional porin for the outer membrane. At the level of oprd transcription, characterized mechanisms include (i) disruptions of the oprd promoter, (ii) premature termination of oprd transcription, (iii) coregulation with mechanisms of trace metal resistance, (iv) salicylate-mediated reduction, and (v) decreased transcriptional expression through mechanisms

13 594 LISTER ET AL. CLIN. MICROBIOL. REV. FIG. 4. Characterization of oprd promoter elements. Transcription of oprd in P. aeruginosa PAO1 initiates with equal frequencies from two start sites, located 23 bases (SS1) and 71 bases (SS2) upstream of the structural gene (ATG translation start codon is highlighted in orange). The putative 10 and 35 promoter elements for SS1 are highlighted in red, and the putative 10 and 35 promoter elements for SS2 are highlighted in blue. of coregulation with the multidrug efflux pump encoded by mexef-oprn. oprd promoter disruptions have occurred as a result of deletions or insertions within the upstream region of oprd. Yoneyama and Nakae reported the association of a large deletion encompassing the putative promoter and initiation codon that prevented transcription of oprd (288). IS1394 and an ISPa16-like insertion (IS) element have been described upstream of the oprd coding region for imipenem-resistant isolates of P. aeruginosa exhibiting decreased oprd expression (273, 281). El Amin et al. evaluated the transcription of oprd among clinical isolates of P. aeruginosa by using two sets of primers that amplified either the upstream or downstream regions of the structural gene (47). Two strains of P. aeruginosa exhibited normal levels of oprd transcription in experiments with the upstream primers, but oprd transcripts were undetectable with the downstream primers. Four additional strains demonstrated significant differences in the amounts of oprd transcript measured with the two primer sets. El Amin et al. concluded that premature termination of transcription was occurring in these strains, potentially due to mutations within the structural gene sequence. The trace metals zinc and copper have been shown to decrease the expression of oprd in P. aeruginosa, leading to imipenem resistance (22, 189). This negative regulation is mediated through the regulatory proteins CzcR and CopR, which respond to the presence of zinc and copper, respectively, to activate mechanisms of metal resistance. CzcR and CopR both downregulate oprd transcription directly or indirectly, through unidentified factors. The weak aromatic acid salicylate was also shown to repress the transcription of oprd through an uncharacterized mechanism, leading to imipenem resistance among salicylate-exposed P. aeruginosa isolates (180). Perhaps the most complex and intriguing mechanisms impacting the transcription of oprd are those that are linked to the regulation of expression of the mexef-oprn efflux pump (101, 180). These mechanisms of coregulation are discussed in detail later in the review, but they highlight the complexity by which P. aeruginosa is able to regulate expression of resistance mechanisms and why it is sometimes so difficult to definitively link phenotypes to changes in one specific mechanism. Finally, mechanisms of OprD deficiency related to translation of an active porin include (i) mutations, insertions, and/or deletions creating frameshifts and premature stop codons (195) and (ii) disruption of the oprd structural gene by insertion of large IS elements (51, 277). Discordance between oprd Expression and Susceptibility to Imipenem Although the relationship between OprD deficiency and imipenem resistance has been well established in the literature, it should not come as a surprise that P. aeruginosa does not always follow expected rules. The genetic versatility of this pathogen and its ability to coregulate multiple resistance mechanisms make P. aeruginosa a constantly moving target and one of our greatest therapeutic challenges. Studies from our laboratory have identified intriguing strains that exhibit discordance between oprd expression and susceptibility to carbapenems. The first example is an isogenic mutant, P. aeruginosa 410L, that was selected with levofloxacin from P. aeruginosa Tokai#1 (275). As background, strain Tokai#1 is an isogenic mutant of P. aeruginosa PAO1 that lacks susceptibility to both levofloxacin and imipenem (145). The MIC for imipenem against Tokai#1 is 8 g/ml, compared to 1 g/ml against the parental PAO1 strain, and this decreased susceptibility correlates with a fivefold decrease in the level of oprd expression (275). Expression of oprd in mutant strain 410L decreased further, to a level 3-fold below that of Tokai#1 and 17-fold below that of the wild-type parent, PAO1. However, despite the further decrease in oprd expression and the inability to detect OprD in outer membrane preparations, mutant 410L lost its resistance to imipenem and reverted back to a level of susceptibility similar to that of the original PAO1 parent. The second example is an isogenic imipenem-hypersusceptible mutant, P. aeruginosa C, that was selected from an imipenem-resistant clinical isolate, P. aeruginosa strain 244, using ciprofloxacin (274). The mechanism of imipenem resistance for P. aeruginosa 244 (MIC 16 g/ml) was shown to be a base transition creating a premature stop codon and preventing translation of full-length OprD. This mutation was retained

14 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 595 FIG. 5. Structure and function of RND efflux pumps in P. aeruginosa. RND pumps typically exist in a tripartite system consisting of an RND cytoplasmic membrane transporter (RND), an MFP, and an OMF. This complex forms a channel spanning the entire membrane, allowing for the proton-driven transport of lipophilic and amphiphilic drugs from the cytoplasm of the cell across the cytoplasmic membrane, through the periplasmic space, across the peptidoglycan, and across the outer membrane. The RND efflux pumps can also extrude drugs from the periplasmic space before they cross the cytoplasmic membrane. in the isogenic C mutant. Therefore, the eightfold increase in susceptibility of P. aeruginosa C to imipenem (MIC 2 g/ml) occurred despite the absence of an active OprD porin in the outer membrane. The discordance between OprD and susceptibility to imipenem described above highlights how much we have yet to learn about the dynamic interactions of carbapenems with P. aeruginosa. The mechanism(s) responsible for increased susceptibility to imipenem in these strains has yet to be elucidated, but characterization of these mechanisms could provide pathways for development of therapeutic strategies to enhance the efficacy of carbapenems. EFFLUX-MEDIATED RESISTANCE While the loss of porins such as OprD represents an effective barrier for drug entry into the cell, a reduction in drug accumulation can also be achieved through active export by membrane-associated pumps. Efflux pumps have been categorized into five superfamilies, based primarily on amino acid sequence identity, the energy source required to drive export, and substrate specificities of the different pumps (218, 259). The superfamilies include (i) the ATP-binding cassette (ABC) family, (ii) the small multidrug resistance family, (iii) the major facilitator superfamily, (iv) the resistance-nodulation-division (RND) family, and (v) the multidrug and toxic compound extrusion family. Although sequence analysis of the P. aeruginosa genome has revealed the presence of efflux systems from all five superfamilies, the largest number of predicted pumps belong to the RND family, with a total of 12 RND systems (including two divalent metal cation transporters) (248). Unlike the primary active transporters of the ABC superfamily, which utilize ATP hydrolysis for energy, the RND family (as well as the remaining superfamilies) are secondary active transporters (symporters, antiporters, and uniporters) that derive the energy required for compound extrusion by proton motive force. Disruption of the proton gradient through the addition of a proton uncoupler, carbonyl cyanide m-chlorophenylhydrazone, increases the accumulation in these bacteria of substrates that are normally exported (172, 201). RND pumps typically exist as a tripartite system consisting of a periplasmic membrane fusion protein (MFP), an outer membrane factor (OMF), and a cytoplasmic membrane (RND) transporter (Fig. 5). This complex forms a channel spanning the entire membrane, allowing for the transportation of lipophilic and amphiphilic drugs from the periplasmic space and cytoplasm to the extracellular environment. The genes encoding these pumps are organized into operons on the P. aeruginosa chromosome (Fig. 6). Each operon is composed of at least two genes, coding for the MFP and the RND transporter. Six of the 12 operons possess an OMF gene, completing the tripartite system, while the remaining operons are devoid of an OMF gene. Several operons have an adjacent regulatory gene transcribed in the same orientation or divergently from the operon, whose product functions as a repressor or activator of pump expression. Operons may contain additional genes besides those coding for the efflux pump. For example, mexg in the mexghi-opmd operon encodes an integral membrane protein, and PA2528-PA2527-PA2526-opmB possesses a second RND transporter gene, PA2526. The 10 RND pumps in P. aeruginosa (excluding the metal cation transporters) are named MexAB-OprM, MexCD-OprJ, MexEF-OprN, MexXY, MexJK, MexGHI-OpmD, MexVW, MexPQ-OpmE, MexMN, and TriABC (Table 3). Mex is an acronym for multiple efflux, and Tri refers to triclosan efflux. While several of these pumps share common substrates, they are also responsible for unique phenotypes inherent to their expression. Substrates of these pumps include antibiotics, biocides, dyes, detergents, organic solvents, aromatic hydrocarbons, and homoserine lactones (Table 3) (205, 234). Although not discussed in this review, these pumps may also have a physiological role in P. aeruginosa (e.g., cell-to-cell communication and pathogenicity) (193), besides their protective effects against antimicrobials. This section of the review focuses on the complex regulation of these pumps and the antibiotic phenotypes associated with their expression in planktonic cells. While P. aeruginosa can also live as a community encompassed in an exopolysaccharide matrix (i.e., biofilm), recent evidence

15 596 LISTER ET AL. CLIN. MICROBIOL. REV. FIG. 6. RND efflux operons in P. aeruginosa. Genes which encode protein components or characterized pumps are denoted by their gene names, and genes encoding protein components of uncharacterized pumps are designated with the P. aeruginosa (PA) numbers assigned in the annotated P. aeruginosa genome sequence (GenBank). Genes are depicted with the following color scheme: dark red arrow, transcriptional regulator-encoding gene; dark blue arrow, membrane fusion protein-encoding gene; light blue arrow, RND transporter-encoding gene; red arrow, outer membrane protein-encoding gene; and gold arrow, gene encoding a protein with unknown function. (Adapted from reference 234 with permission of the publisher.) has suggested that the RND efflux pumps do not participate in biofilm resistance to typical antipseudomonal agents (38) but may be involved with azithromycin resistance in biofilms (66). MexAB-OprM Efflux Pump During a study examining siderophore-mediated iron transport, the first multidrug efflux pump in P. aeruginosa, MexAB- OprM, was discovered by Poole et al. (204). MexAB-OprM is able to export drugs from several different classes, including fluoroquinolones, tetracyclines, chloramphenicol, -lactams and -lactamase inhibitors, macrolides, novobiocin, trimethoprim, and sulfonamides (102, 120, 121, 243, 244). Of the RND efflux pumps, MexAB-OprM has the broadest substrate profile for the -lactam class, with an ability to export -lactams such as the carboxypenicillins, aztreonam, extended-spectrum cephalosporins (e.g., ceftazidime and cefotaxime), penems (e.g., faropenem), and the carbapenems meropenem and panipenem (but not imipenem and biapenem). MexAB-OprM participates in the intrinsic resistance of P. aeruginosa to the agents listed above through its constitutive production in wildtype cells (205). Knockout studies of mexab-oprm alone or in combination with other resistance mechanisms have confirmed the pump s role in intrinsic resistance, as these mutants become hypersensitive (120, 148, 161). Two additional characteristics are associated with mexaboprm expression. First, growth-phase-dependent expression of mexab-oprm has been demonstrated (53). As the growth cycle progressed and cell density increased, mexab-oprm transcription also increased until maximum expression occurred in late log phase/early stationary phase. The growth-phase-dependent upregulation was suggested to involve a quorum sensing signal. Quorum sensing is a mechanism by which bacteria monitor cell density through cell-to-cell communication, allowing for the coordinated expression of certain genes (e.g., virulence factors) in a cell density-dependent manner (260). Cell-to-cell signaling is mediated by diffusible autoinducers, known as homoserine lactone molecules, which interact with their cognate receptors to activate gene expression. N-Butyryl-L-homoserine lactone (C 4 -HSL) is synthesized as part of the rhl quorum sensing system and was shown to enhance expression of mexab-oprm (146, 230). Second, the OMF gene, oprm, was shown to be expressed independently of mexab (290). A weak promoter was discovered upstream of oprm in the coding region of mexb. This promoter was suggested to be less active than the promoter upstream of mexa, contributing to only a fraction of the total amount of transcript. OprM may serve as an OMF for the MexXY (5, 152, 159), MexJK (33), MexVW (122), and MexMN (158) systems, and possibly other RND pumps. Independent expression of oprm may allow for sufficient levels of OMF to accommodate multiple pumps or to ensure its presence in case expression from the mexab-oprm promoter is compromised. Several regulatory loci influence the expression of the mexab-oprm operon. The mexr gene is located directly upstream of but transcribed divergently from mexab-oprm and encodes a repressor belonging to the MarR family of regulatory proteins (Fig. 6) (206). MexR binds as a stable homodimer to two sites, each consisting of inverted repeat sequences,

16 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 597 TABLE 3. Characteristics of RND efflux pumps in P. aeruginosa Operon Component Function a Regulator e Substrate(s) Primary b Secondary c Antibiotics Additional compounds mexab-oprm MexA MFP MexR NalC Fluoroquinolones, -lactams, Biocides (e.g., triclosan), MexB RND NalD -lactamase inhibitors, detergents, dyes, HSL, f OprM OMF tetracyclines, chloramphenicol, macrolides, novobiocin, trimethoprim, sulfonamides aromatic hydrocarbons mexcd-oprj MexC MFP NfxB Fluoroquinolones, -lactams, Biocides (e.g., triclosan), MexD RND tetracycline, chloramphenicol, detergents, dyes, OprJ OMF macrolides, trimethoprim, novobiocin aromatic hydrocarbons mexef-oprn MexE MFP MexT MexS Fluoroquinolones, Biocides (e.g., triclosan), MexF RND MvaT chloramphenicol, trimethoprim aromatic OprN OMF hydrocarbons mexxy MexX MFP MexZ Fluoroquinolones, -lactams, MexY RND tetracycline, aminoglycosides, OprM/Opm- d OMF macrolides, chloramphenicol mexjk MexJ MFP MexL Tetracycline, erythromycin Biocides (e.g., triclosan) MexK RND OprM/OpmH OMF mexghi-opmd MexG? SoxR Fluoroquinolones Vanadium MexH MFP MexI RND OpmD OMF mexvw MexV MFP Fluoroquinolones, tetracycline, MexW RND chloramphenicol, erythromycin OprM OMF mexpq-opme MexP MFP Fluoroquinolones, tetracycline, MexQ RND chloramphenicol, macrolides OpmE OMF mexmn MexM MFP Chloramphenicol, thiamphenicol MexN RND OprM OMF triabc TriA MFP Triclosan TriB MFP TriC RND OpmH OMF a MFP, membrane fusion protein; RND, resistance-nodulation-division transporter; OMF, outer membrane factor;?, encodes a protein of unknown function. b Regulatory proteins that directly control expression of the efflux operons. c Proteins that indirectly activate operon expression when mutated. d MexXY may utilize OpmB, OpmG, OpmH, and/or OmpI as OMFs. e, not identified. f HSL, homoserine lactones. within the mexr-mexa intergenic region (52). The binding region encompasses overlapping mexr and mexa promoters, and association of MexR dimers with these sites represses transcription of the mexab-oprm operon and negatively autoregulates its own expression (52, 220). A second regulatory factor of the mexab-oprm operon, NalD, was discovered following insertional mutagenesis of a gene, nald, located adjacent to a putative pump gene of the major facilitator superfamily (239). NalD is a repressor of the TetR family that binds to a sequence upstream of mexab-oprm but downstream of the mexr binding sites (160). The NalD operator overlaps a second promoter for mexab-oprm, and binding of NalD restricts expression from this proximal mexa promoter. The C 4 -HSLmediated growth phase regulation of mexab-oprm described above occurs independently of MexR (230). However, the possibility that C 4 -HSL directly or indirectly influences NalDmediated regulation of mexab-oprm to promote growth phase expression has yet to be determined experimentally. Overexpression of mexab-oprm has been detected in nalb-, nalc-, and nald-type multidrug-resistant mutants and selected both in vivo (2, 137, 181, 184, 239, 273) and in vitro (2, 24, 239, 245). In nalb-type strains, mutations within mexr disrupt the translation of full-length protein or compromise the repressor activity of MexR by causing a loss of dimerization, defects in

17 598 LISTER ET AL. CLIN. MICROBIOL. REV. DNA binding, or, possibly, instability of the protein (2, 219). Several mutations in mexr have been described, including nucleotide changes (e.g., base substitutions, deletions, and insertions) and insertion of an IS element (2, 19, 219, 245). The absence of MexR from its operators leads to pump overexpression from the distal mexa promoter (52). nalc-type strains hyperexpress mexab-oprm, but at lower levels than those in nalb-type mutants (245). Transposon insertional mutagenesis identified the site for nalc-type mutations in the PA3721 gene (renamed nalc) (24). nalc encodes a putative repressor of the TetR/AcrR family, whose genes are located upstream of an operon, PA3720-PA3719, that is negatively regulated by NalC (24). Loss of NalC resulted in overexpression of PA3720- PA3719, and subsequent experiments demonstrated that PA3719 (renamed ArmR) upregulates mexab-oprm by interacting with MexR (24, 35). ArmR, a 53-amino-acid antirepressor, allosterically inhibits MexR dimer-dna binding by occupying a hydrophobic binding cavity within the center of the MexR dimer (272). As implied by the name, overexpression of mexab-oprm in nald-type mutants occurs in response to disruption of NalD (239). Complementation of nald-type mutants with a functional nald gene reduced mexab-oprm hyperexpression and drug resistance (239). Mutations within NalD were believed to alleviate repression of the proximal mexa promoter (160), presumably by an inability to bind to its operator. Interestingly, maximum expression from the proximal mexa promoter in NalD-negative strains requires the presence of MexR (160). A mutational event within one of the known regulatory genes may not be the sole mechanism to increase mexab-oprm transcription. Chen et al. recently described the effect of oxidative stress on operon expression (29). Oxidation of two cysteine residues within MexR causes the formation of an intermonomer disulfide bond which alters the conformation of MexR. Oxidized MexR dissociates from the mexr-mexa intergenic region, allowing access of RNA polymerase to the distal mexa promoter. In this situation, the MexR protein serves as a sensor of oxidative stress, and the response culminates in activation of the efflux defense mechanism, possibly to remove the agent responsible for inducing the oxidative stress. MexCD-OprJ Efflux Pump The operon coding for MexCD-OprJ was cloned and sequenced by Poole et al. in 1996 and showed a high degree of homology to MexAB-OprM (203). MexCD-OprJ can extrude a variety of antimicrobial agents, including fluoroquinolones, -lactams, chloramphenicol, tetracycline, novobiocin, trimethoprim, and macrolides (70, 102, 149, 243). Unlike MexAB-OprM, MexCD-OprJ does not have an extensive substrate profile for the -lactams, but rather, it preferentially exports the fourth-generation cephalosporins (e.g., cefepime, cefpirome, and cefozopran) (149, 203). Transcription of mexcd-oprj can be observed in wild-type cells (276), but the levels are most likely not sufficient to produce detectable levels of protein (70, 203). In addition, deletion of mexcdoprj has no impact on wild-type susceptibility, indicating that this pump does not contribute to intrinsic resistance (162, 244). Expression of mexcd-oprj was shown to be inducible in response to benzalkonium chloride, chlorhexidine gluconate, tetraphenylphosphonium chloride, ethidium bromide, rhodamine 6G, and acriflavine but not in response to clinically relevant antibiotics (162). Induction of mexcd-oprj by membrane damaging agents (i.e., chlorhexidine) was dependent upon the stress response sigma factor AlgU (55). Expression of mexcd-oprj is governed by the product of a gene, nfxb, located upstream of mexcd-oprj but transcribed divergently from the operon (Fig. 6) (203). NfxB displays similarity to proteins of the LacI-GalR family who possess a helixloop-helix motif characteristic of DNA binding (203). NfxB negatively regulates expression of mexcd-oprj, as well as its own expression (203, 238), by binding to a site composed of two 39-bp repeats within the nfxb-mexc intergenic region (238). Mutations within nfxb are suggested to alter the repressor activity of NfxB, leading to hyperexpression of mexcd-oprj in so-called nfxb-type mutants (203, 238). Several mutations have been described for nfxb from laboratory strains and clinical isolates, such as base substitutions, deletions, and interruption with an IS element (37, 90, 203). Two types of hyperexpressing mutants, types A and B, have been identified in P. aeruginosa, with differing levels of MexCD-OprJ production and susceptibility (149). Type B mutants produce larger amounts of MexCD-OprJ and have more substantial changes in susceptibility than do type A mutants. Complementation of both types of mutants with wild-type nfxb restored susceptibilities to wild-type levels, suggesting that mutations in NfxB were responsible (149). Regulation of mexcd-oprj expression may not be limited to NfxB. In mexab-oprm and oprm deletion derivatives of wild-type strain PAO1, expression of mexcdoprj markedly increased, indicating that MexAB-OprM may influence the expression of mexcd-oprj (119). The mechanism responsible for mexcd-oprj upregulation in MexAB-OprMdeficient strains has yet to be determined. Whereas the overproduction of MexCD-OprJ in P. aeruginosa provides resistance to several antibacterial drugs, an interesting phenotype of hypersusceptibility ( 4-fold increase in susceptibility) to some -lactams and aminoglycosides is observed in the same strains. The correlation between MexCD- OprJ overproduction and hypersusceptibility and the mechanisms responsible for hypersusceptibility are discussed later in this section. MexEF-OprN Efflux Pump Three open reading frames in an operon named mexefoprn were characterized by Kohler et al., and the products of mexef-oprn exhibit amino acid identity to the components of MexAB-OprM and MexCD-OprJ (104). Substrates recognized by MexEF-OprN include fluoroquinolones, chloramphenicol, and trimethoprim (104), but the pump has no apparent affinity for currently available -lactams. Expression of mexef-oprn was originally reported as quiescent (104), but later studies detected low-level expression in strains with wild-type susceptible phenotypes (119, 276). Disruption of mexef-oprn in wildtype strains did not affect susceptibility, excluding its participation in intrinsic resistance (104). Regulation of mexef-oprn differs from that of mexab-oprm and mexcd-oprj in that operon expression is not suppressed by a negative regulator. Multiple factors are capable of controlling mexef-oprn transcription. mext, located upstream of and

18 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 599 transcribed in the same orientation as mexef-oprn (Fig. 6 and 7A), encodes a protein belonging to the LysR family of transcriptional activators that is capable of positively regulating mexef-oprn expression (Fig. 7A) (101, 180). Because of sequence variations within mext genes of different wild-type P. aeruginosa strains, activation of MexT can occur through different pathways (145). In some P. aeruginosa strains, MexT exists in a dormant form because of suppressing mutations within the coding region, but additional cis-acting mutations or deletions within mext convert inactive MexT into an active form (145). In other strains, suppressing mutations are not present within mext (145), but stimulation of MexT requires potential changes in the levels of cognate effector molecules (101, 145). Mutations in these strains occur within a gene, mexs, located upstream of mext that encodes a putative oxidoreductase/ dehydrogenase homologue (Fig. 7C) (241). Inactivation of MexS is believed to cause a buildup of metabolites that serve as effector molecules for MexT, which, in turn, upregulates mexef-oprn expression to remove the toxic metabolites (241). Therefore, an active MexT protein is required for upregulation of mexef-oprn by this pathway. While mexef-oprn-overexpressing strains have been encountered among nosocomial P. aeruginosa strains (47, 181, 207, 273), a correlation between pump overexpression and genetic changes within mext and/or mexs in clinical isolates has, to the best of our knowledge, been lacking. Expression of mexef-oprn is also controlled by a member of the histone-like nucleoid structuring protein family, MvaT, which serves as a global regulator of genes involved in virulence, housekeeping, and biofilm formation (Fig. 7D) (26, 35, 258, 270). MvaT binds to and oligomerizes across AT-rich regions of DNA with a high affinity and, as a result, silences the expression of certain genes (26). Deletion of mvat caused an increase in mexef-oprn expression that was not dependent on mext or mexs (270). MvaT does not bind upstream of mexefoprn (270). Thus, the loss of this regulatory protein most likely has an indirect effect on regulating mexef-oprn expression. Data from a more recent study suggest that regulation of mexef-oprn is even more complex and that more uncharacterized pathways are likely operative in P. aeruginosa (279). In this study, an isogenic mutant overexpressing mexef-oprn was selected from a clinical isolate of P. aeruginosa by use of levofloxacin. Analysis of gene sequences and transcriptional expression demonstrated that the overexpression of mexef-oprn did not involve changes in mext, mexs, or mvat. The mechanism of mexef-oprn regulation in this isogenic mutant remains to be characterized. Similar to nfxb-type mutants, nfxc-type mutants become hypersusceptible to certain -lactams and aminoglycosides (58, 202). -Lactam hypersusceptibility was associated with repression of the quorum sensing-mediated enhancement of mexaboprm expression by MexT (146). Transcription of the C 4 -HSL autoinducer synthase gene, rhli, was previously shown to be reduced in nfxc-type mutants (105). As mentioned above, growth phase expression of mexab-oprm is dependent on this quorum sensing molecule. Therefore, MexT appears to act as a global regulator controlling expression of mexef-oprn, oprd, and genes regulated by the autoinducer C 4 -HSL, such as mexab-oprm. MexXY Efflux Pump mexxy was cloned from the P. aeruginosa chromosome by Mine et al. in 1999, and the pump displays properties similar to those of the efflux systems described above (159). Unlike the operons described above, mexxy lacks a gene coding for an outer membrane protein (Fig. 6). Instead, MexXY is able to associate with OprM (5, 32, 159), and possibly other outer membrane proteins, such as OpmB, OpmG, OpmH, and OpmI, to form a functional tripartite system (32, 168). Fluoroquinolones, specific -lactams (i.e., cefepime), aminoglycosides, tetracycline, chloramphenicol, and erythromycin are all substrates for MexXY (202, 234). mexxy expression is induced when cells are grown in the presence of tetracycline, erythromycin, and gentamicin (152). Deletion of mexxy in wild-type strains increases their susceptibility to these antibiotics, suggesting that this pump contributes to intrinsic resistance (152, 161). MexXY does not contribute to the intrinsic resistance to fluoroquinolones, a pump substrate, because these agents fail to induce mexxy expression (152). The product of a gene, mexz, located upstream of but transcribed divergently from mexxy, negatively regulates the expression of the operon (Fig. 6) (5, 154). MexZ belongs to the TetR family of transcriptional regulators and contains a characteristic N-terminal helix-turn-helix DNA binding domain. MexZ binds as a homodimer to an inverted repeat region, located in the mexz-mexx intergenic region, which encompasses the putative mexxy promoter (154). Mutations in mexz or the mexz-mexx intergenic region have been associated with hyperexpression of mexxy (85, 263). However, overexpression of mexxy has also been detected in mutants not harboring mutations within mexz or the mexz-mexx intergenic region (137, 240, 269). Llanes et al. proposed the names agrz-type and agrw-type mutants to describe mutants hyperexpressing mexxy with alterations within mexz and outside mexz, respectively (137). Thus, mexxy may be regulated by multiple factors, similar to mexab-oprm, and mutations within as yet unidentified genes are necessary for mexxy overexpression. MexXY hyperexpression has been well documented for agrz-type and agrw-type clinical isolates resistant to fluoroquinolones, cefepime, and aminoglycosides (79, 137, 240, 263, 283). Multiple pathways also participate in the regulation of mexxy induction. Interaction of bacterial ribosomes with protein synthesis inhibitors (e.g., tetracycline, chloramphenicol, and aminoglycosides) is necessary to upregulate mexxy expression, as plasmid-encoded ribosomal protection mechanisms reduce mexxy drug inducibility (93). Induction may not always require the presence of MexZ (93, 163). Inducing antibiotics had no effect on the binding of MexZ to its operator (154) and could further enhance mexxy expression in a mexz-deficient mutant (93), suggesting that mexxy induction was independent of MexZ. A more recent study by Morita et al. implicated the PA5471 gene in mediating mexxy induction (163). Disruption of PA5471 compromised mexxy drug inducibility, while increased expression of PA5471 alone stimulated mexxy expression. PA5471 transcription is also induced by ribosome inhibitors (the same drugs which induce mexxy), and PA5471 induction indirectly or directly alters MexZ activity, but not mexz expression, resulting in mexxy upregulation (163).

19 FIG. 7. Coregulation of mexef-oprn and oprd in P. aeruginosa. The models represent the proposed mechanisms of coregulation of mexef-oprn and oprd in P. aeruginosa. Each panel represents the chromosome of P. aeruginosa, highlighting the mexe, mexf, oprn, and oprd structural genes and the proposed genes involved in coregulation, mext, mexs, and mvat. (A) Basal expression of mexef-oprn and oprd in wild-type P. aeruginosa. In wild-type P. aeruginosa, MexT is functionally inactive due to either the presence of suppressing mutations or the lack of a secondary effector molecule. As a result, expression of mexef-oprn occurs at a low basal level, and expression of oprd occurs at a level sufficient to provide quantities of OprD in the outer membrane sufficient for normal cellular function. (B) MexT-associated coregulation of mexef-oprn and oprd. In nfxc-type mutants, MexT becomes active through a mutation within the structural gene. The activated MexT protein positively regulates (green arrow) transcription of mexef-oprn, leading to overexpression of the efflux operon and overproduction of the MexEF-OprN efflux pump. Simultaneously, MexT negatively regulates (red arrow) oprd at the transcriptional and posttranscriptional levels, leading to decreased production of OprD. (C) MexS-associated coregulation of mexef-oprn and oprd. Loss of MexS, a putative oxidoreductase/dehydrogenase, has been suggested to cause a buildup of secondary metabolites which may serve as effector molecules for MexT. These effector molecules could bind to MexT, alter the conformational state of the regulatory protein, and transform MexT into an activating transcriptional regulator. As a result, MexT can positively regulate (green arrow) the expression of mexef-oprn and negatively regulate (red arrow) the expression of oprd, similar to what is described for panel B. (D) MvaT-associated coregulation of mexef-oprn and oprd. Loss of the global regulatory protein MvaT is also associated with the upregulation of the mexef-oprn operon. The mechanism of MvaT-associated regulation has not been elucidated, but it functions independent of MexT and MexS. In contrast to the case for the MexT- and MexS-associated regulatory pathways, loss of MvaT causes an upregulation of both mexef-oprn and oprd expression. 600

20 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 601 MexJK Efflux Pump The MexJK efflux pump was discovered in a mexab-oprm mexcd-oprj background strain following selection with the broad-spectrum biocide triclosan (33). An OMF-encoding gene is also absent from the mexjk operon (Fig. 6), but a three-component system is formed with the addition of either OprM (32, 33) or OpmH (32). MexJK-OprM exports erythromycin and tetracycline from the cell, while MexJK-OpmH removes triclosan (32). mexjk was reported not to be expressed in wild-type cells (33). A member of the TetR regulatory family, mexl, is present upstream of mexjk but transcribed in the opposite orientation (Fig. 6) (33). MexL binds as a multimer to inverted repeats within the mexl-mexj intergenic region encompassing overlapping mexjk and mexl promoters (31). MexL binding to this site represses the transcription of both mexjk and mexl, thereby negatively autoregulating its own expression. A mutation within the helix-turn-helix motif of MexL prevented binding to its operator site and led to mexjk overexpression in a laboratory mutant (31). Overexpression of mexjk has also been detected in a few clinical isolates, but the pump s contribution to resistance and potential mutations in mexl were not evaluated (79). Additional RND Efflux Pumps The contributions of the remaining RND efflux pumps to resistance and the factor(s) governing their expression have just begun to be elucidated. Their up- or downregulation in clinical isolates with various phenotypes has yet to be reported. mexghi-opmd expression is detectable in wild-type cells and is under the control of a redox-active protein, SoxR, belonging to the MerR family of transcriptional regulators (4, 185). Norfloxacin was the only antibiotic reported to be a substrate of MexGHI-OpmD (236), but this pump was also implicated in the export of other nonantibiotic compounds, including vanadium (4). Interestingly, a mutant incapable of expressing mexghi-opmd showed decreased susceptibility to tetracycline and ticarcillin-clavulanate (4). mexi and opmd knockout mutants in a subsequent study also exhibited decreased susceptibility to kanamycin, spectinomycin, carbenicillin, nalidixic acid, tetracycline, and chloramphenicol (3). Perhaps the loss of mexghi-opmd triggered the overexpression of another RND efflux pump, similar to the overexpression of mexcd-oprj and mexef-oprn observed in P. aeruginosa knockout mutants of mexab-oprm (119). The substrate profiles of MexVW, MexPQ-OpmE, and MexMN were all examined in an efflux-deficient strain ( mexab-oprm mexcd-oprj mexef-oprn mexxy) harboring a plasmid coding for each system individually (122, 158). MexVW used OprM and other, as yet unidentified OMF proteins for the export of fluoroquinolones, tetracycline, chloramphenicol, and erythromycin (122). MexPQ-OpmE elevated the MICs of fluoroquinolones, tetracycline, chloramphenicol, and several macrolides in the recipient strain (158). Susceptibility to chloramphenicol and thiamphenicol was decreased in the recipient containing MexMN in combination with OprM (158). TriABC was the last RND efflux pump to be characterized for P. aeruginosa. triabc differs from the other RND operons in that tria and trib each code for MFPs, both of which are required for substrate export (157). The OMF OpmH associates with TriABC to assemble a functional pump capable of exporting triclosan (157). No antibiotics were reported as substrates for this pump. COREGULATION OF RESISTANCE MECHANISMS MexCD-OprJ Overproduction and Hypersusceptibility to Antibacterials While the overexpression of mexcd-oprj provides resistance to several classes of antibacterial drugs, hypersusceptibility to aminoglycosides and other -lactams is also observed (149, 203). Hypersusceptibility is defined as a fourfold or greater increase in susceptibility, and the level of hypersusceptibility is related to the level of MexCD-OprJ production. Masuda et al. demonstrated that type B mutants (most production of MexCD-OprJ) exhibit larger increases in susceptibility than do type A mutants (moderate production of MexCD-OprJ) (149). However, the mechanism(s) responsible for hypersusceptibility is much more complex than simply being associated with production of MexCD-OprJ. Phenotypic reversion studies with mexcd-oprj-overexpressing mutants have demonstrated that hypersusceptibility to some antibiotics is not always associated with the level of mexcd-oprj overexpression (280). Furthermore, as discussed below, a single mechanism does not account for hypersusceptibility to all affected antibiotics, and the mechanism(s) responsible for hypersusceptibility to imipenem remains uncharacterized. Hypersusceptibility of laboratory-generated nfxb-type mutants to carbenicillin and aztreonam (MexAB-OprM substrates) has previously been linked to a concurrent downregulation of MexAB-OprM production (70). However, during the analysis of nfxb-type clinical isolates, mexb transcript and MexB protein levels were not downregulated, despite hypersusceptibility to these drugs (92). Instead, a decrease in MexAB-OprM pump activity in these strains was proposed as the mechanism (92). These results do not necessarily exclude a possible coregulation of mexcd-oprj and oprm expression, since transcription of oprm can occur independently of that of mexab (290). MexCD-OprJ-overproducing mutants become hypersusceptible to additional -lactams, including sulbenicillin, cefpodoxime, ceftriaxone, imipenem, and biapenem (151), but none of the aforementioned drugs are exported by MexAB-OprM, implying an additional mechanism(s). A disruption in the induction pathway of AmpC was suggested to cause hypersusceptibility to these -lactams (151). Although data from our own studies also showed decreased induction of ampc expression associated with overexpression of mexcd-oprj (274), this association was observed only in isogenic mutants from a clinical isolate with wild-type ampc expression. When the parent strains were derepressed for ampc expression, overexpression of mexc was not associated with any change in ampc expression among the isogenic mutants selected (274). The mechanism(s) involved in hypersusceptibility to imipenem remains uncharacterized. Although other investigators have suggested that imipenem hypersusceptibility is a result of disrupted ampc induction (151), the observation

21 602 LISTER ET AL. CLIN. MICROBIOL. REV. that AmpC overproduction does not significantly increase imipenem MICs (63, 131, 169, 274) argues against this hypothesis. Furthermore, our own studies have shown that imipenem-hypersusceptible mutants can be selected from P. aeruginosa parent strains that are partially and fully derepressed for ampc expression, without any changes in either the expression of ampc or AmpC hydrolysis activity (274). These studies also demonstrated that imipenem hypersusceptibility is unrelated to the level of OprD, as hypersusceptible mutants failed to exhibit any changes in oprd expression or OprD protein in the outer membrane (274). Furthermore, imipenem-hypersusceptible mutants were selected from a P. aeruginosa strain that lacked a functional OprD protein (274). Characterization of the mechanism(s) responsible for imipenem hypersusceptibility among mexcd-oprj-overexpressing P. aeruginosa strains may uncover potential drug targets to enhance the activity of carbapenems. The mechanism responsible for hypersusceptibility to aminoglycosides remains unknown. As previously discussed, aminoglycosides are substrates for the MexXY efflux pump, and suppression of MexXY in strains overproducing MexCD-OprJ has been suggested (205), but this inverse relationship has not been observed in clinical isolates (92). Coregulation of MexEF-OprN and OprD One of the most intriguing and complex coregulatory pathways involves the concurrent overproduction of the MexEF- OprN efflux pump and downregulation/upregulation of the OprD porin. Clinically, this coregulatory process allows a single mutational process to impact the potencies of both the fluoroquinolones and carbapenems. To date, three proteins, MexT, MexS, and MvaT, have been linked directly or indirectly to the coregulation of MexEF-OprN and OprD in P. aeruginosa (Fig. 7). P. aeruginosa strains that overexpress mexef-oprn through MexT become resistant to antibiotics which are substrates for the pump (listed above) but also lose susceptibility to imipenem, which is not extruded by MexEF-OprN. Rather, the loss of susceptibility to imipenem is associated with a concurrent decrease of oprd expression and OprD (104, 150, 180). The link between pump hyperexpression and porin loss involves coregulation by MexT (Fig. 7B). Studies have shown that MexT alone is capable of downregulating oprd at the transcriptional and posttranscriptional levels, causing a significant reduction in the amount of OprD (101, 180). Further study into this mechanism of coregulation has shown that neither premature termination of transcription nor changes in transcript stability are responsible for the reduced levels of oprd transcript (278). Instead, the MexT-associated decrease in oprd expression involves decreased transcription initiation from the start-site-proximal oprd promoter (Fig. 8) (278). It is possible that MexT or another regulatory factor(s) and/or cofactors bind to the region between SS1 and SS2 and inhibit transcription initiation from SS1. Mutations within mexs, encoding a putative oxidoreductase/dehydrogenase, are also associated with concurrent FIG. 8. MexT-associated downregulation of oprd expression. Transcription of oprd in wild-type P. aeruginosa PAO1 initiates from two start sites, SS1 and SS2. MexT-associated downregulation of oprd expression is associated with a selective inhibition of transcription from SS1 (278). The model proposed in this figure involves the binding of a regulatory protein, potentially MexT, with or without cofactors, within the promoter region between SS1 and SS2, blocking efficient initiation of transcription from SS1. overexpression of mexef-oprn and downregulation of oprd expression (Fig. 7C) (241). As discussed above for regulation of MexEF-OprN, inactivation of MexS is believed to cause a buildup of metabolites that serve as effector molecules for MexT (241), which mediates the downregulation of oprd expression following activation. In contrast to MexTand MexS-associated downregulation of oprd expression, inactivation of the global regulator MvaT causes an upregulation in both mexef-oprn and oprd transcription (Fig. 7D) (270). However, the mechanistic pathway responsible for oprd upregulation has not been characterized. Regulation of mexef-oprn expression by MexT (with or without mexs inactivation) and MvaT can impact susceptibility to imipenem through oprd coregulation. However, coregulation of mexef-oprn and oprd is not always observed, further highlighting the complexity of resistance mechanism regulation in P. aeruginosa. For example, in a recent study, an isogenic mutant overexpressing mexefoprn showed no phenotypic change in imipenem susceptibility, oprd transcript levels, or levels of OprD in the outer membrane (279). Sequence and expression analysis of the regulatory genes mext, mexs, and mvat failed to show any genetic changes in the mutant, suggesting an alternative pathway of mexef-oprn regulation without concurrent oprd regulation (279). Reminiscent of increased mexcd-oprj expression in mexab-oprm and oprm knockout mutants, mexef-oprn was also hyperexpressed in mexab-oprm-inactivated mutants, but a change in imipenem susceptibility was not observed in the deletion mutants (119).

22 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 603 PREVENTING EMERGENCE OF CHROMOSOMALLY ENCODED RESISTANCE This review has highlighted the impressive ability of P. aeruginosa to develop antibacterial resistance through mutational changes in the function and/or production of chromosomally encoded resistance mechanisms. Furthermore, the most difficult challenge with this pathogen is the ability of P. aeruginosa to become resistant during treatment of an infection. Unfortunately, the prospect for bringing new, and specifically novel, antipseudomonal drugs to clinical use in the near future is not promising. Therefore, the challenge facing us today is to slow the emergence of resistance through optimizing therapy with currently available drugs. The two most common strategies considered to address this need are (i) optimizing therapy through our understanding of basic antibacterial pharmacodynamic principles and (ii) treating P. aeruginosa with a combination of antibacterial drugs. Although our understanding of antibacterial pharmacodynamics can help in the selection of the best antibiotic and/or dosing strategy to optimize therapy, preventing the emergence of resistance requires the inclusion of potential resistant subpopulations into the equation. Unfortunately, this is not always straightforward, since the shifts in susceptibility associated with mutations can vary widely depending upon the resistance mechanism involved, the differential impact of resistance mechanisms on specific antibiotics, and the potential cooperation of multiple resistance mechanisms. Therefore, pharmacodynamic optimization based upon the susceptibility of the original clinical isolate does not always address the risk of resistance emerging during therapy. Some investigators have promoted replacing the MIC with the mutation prevention concentration, but this approach has not been adopted by clinical laboratories. A more accepted approach is to treat serious P. aeruginosa infections with a combination of antibacterial agents. Although synergistic interactions are an important aspect for some drug combinations (e.g., trimethoprim-sulfamethoxazole), the primary focus of combination therapy against P. aeruginosa is preventing the emergence of resistance. The combination of an antipseudomonal -lactam with an aminoglycoside has often been the treatment of choice for this pathogen. However, this combination does not always prevent the emergence of AmpCmediated resistance to the -lactams, and clinical failures are still a risk (97, 118, 147, 226, 228, 235). Therefore, the search for more effective combinations must be a priority. The combination of levofloxacin-imipenem has been shown to prevent the emergence of resistance during therapy of P. aeruginosa in a two-compartment pharmacodynamic model (129, 130). In these in vitro studies, the levofloxacin-imipenem combination prevented emergence of resistance in susceptible clinical isolates, as well as in strains with characterized mechanisms that provided resistance to one or both drugs in the combination. Although these two studies focused specifically on levofloxacin-imipenem, it is expected that ciprofloxacin, meropenem, and doripenem could be used as alternatives in this combination without compromising efficacy. At this time, clinical evaluation of the combination is needed. CONCLUDING COMMENTS Treatment of infectious diseases becomes more challenging with each passing year. Continued increases in immunosuppressed/compromised patient populations and the evolutionary advantage of bacteria to rapidly mutate and adapt to antibacterial/biocide threats in their environment make the treatment of infectious diseases a serious challenge. This is especially true for the opportunistic pathogen P. aeruginosa and its ability to develop a multidrug-resistant phenotype. Although the potential import of resistance mechanisms on mobile genetic elements is a continuing threat, perhaps the most difficult challenge we face with P. aeruginosa is its ability to rapidly develop resistance to multiple classes of antibiotics during the course of treating a patient. The chromosomal AmpC cephalosporinase, the outer membrane porin OprD, and the multitude of efflux pumps are particularly relevant to this therapeutic challenge, and the discussion presented in this review highlights the complex mechanisms and pathways by which P. aeruginosa regulates and/or coregulates their expression. As the pipeline of new drugs achieving FDA approval continues to diminish, it is critical that we look for novel strategies to combat the threat of antibacterial resistance. One potential strategy is to target the regulation of bacterial resistance mechanisms as a pathway to enhance the potency of available drugs and, perhaps, restore the efficacy of available drugs. In addition to the development of direct inhibitors of resistance mechanisms, i.e., -lactamase inhibitors, another strategy is to target the regulation of gene expression. Although a great deal of knowledge has been gained toward understanding the mechanisms by which P. aeruginosa regulates AmpC, OprD, and efflux pumps, it is clear that we have a long road ahead and have much to learn about how this resourceful pathogen coregulates different resistance mechanisms to meet the antibacterial challenges it faces. ACKNOWLEDGMENTS Daniel Wolter created Fig. 2 through 8. We acknowledge Patrick Lane for his enhancement of the figures. Philip Lister received research grant support from the National Institutes of Health (5R21AI ), Ortho-McNeil, Merck, Wyeth-Ayerst, and Astra-Zeneca. Nancy Hanson received research grant support from the National Institutes of Health (5R21AI ), Merck, Astra-Zeneca, Johnson & Johnson, and Becton-Dickinson. REFERENCES 1. Abdalhamid, B., P. A. Wickman, and N. D. Hanson Correlation of ampc induction with PBP binding in Enterobacter cloacae, abstr. C Abstr. 45th Intersci. Conf. Antimicrob. Agents Chemother., Washington, DC. 2. Adewoye, L., A. Sutherland, R. Srikumar, and K. Poole The mexr repressor of the mexab-oprm multidrug efflux operon in Pseudomonas aeruginosa: characterization of mutations compromising activity. J. Bacteriol. 184: Aendekerk, S., S. P. Diggle, Z. Song, N. Hoiby, P. Cornelis, P. Williams, and M. Camara The MexGHI-OpmD multidrug efflux pump controls growth, antibiotic susceptibility and virulence in Pseudomonas aeruginosa via 4-quinolone-dependent cell-to-cell communication. Microbiology 151: Aendekerk, S., B. Ghysels, P. Cornelis, and C. Baysse Characterization of a new efflux pump, MexGHI-OpmD from Pseudomonas aeruginosa that confers resistance to vanadium. Microbiology 148: Aires, J. R., T. Kohler, H. Nikaido, and P. Plesiat Involvement of an active efflux system in the natural resistance of Pseudomonas aeruginosa to aminoglycosides. Antimicrob. Agents Chemother. 43: Akasaka, J. R., M. Tanaka, A. Yamaguchi, and K. Sato Type II

23 604 LISTER ET AL. CLIN. MICROBIOL. REV. topoisomerase mutations in fluoroquinolone-resistant clinical strains of Pseudomonas aeruginosa isolated in 1998 and 1999: role of target enzyme in mechanism of fluoroquinolone resistance. Antimicrob. Agents Chemother. 45: Aloush, V., S. Navon-Venezia, Y. Seigman-Igra, S. Cabili, and Y. Carmeli Multidrug-resistant Pseudomonas aeruginosa: risk factors and clinical impact. Antimicrob. Agents Chemother. 50: Avison, M. B., P. Niumpsup, K. Nurmahomed, T. R. Walsh, and P. M. Bennett Role of the cre/blr-tag DNA sequence in regulation of gene expression by the Aeromonas hydrophila beta-lactamase regulator, BlrA. J. Antimicrob. Chemother. 53: Bagge, N., O. Ciofu, M. Hentzer, J. I. Cambell, M. Givskov, and N. Hoiby Constitutive high expression of chromosomal beta-lactamase in Pseudomonas aeruginosa caused by a new insertion sequence (IS1669) located in ampd. Antimicrob. Agents Chemother. 46: Barnaud, G., R. Labia, L. Raskine, M. J. Sanon-Le Pors, A. Philippon, and G. Arlet Extension of resistance to cefepime and cefpirome associated to a six amino acid deletion in the H-10 helix of the cephalosporinase of an Enterobacter cloacae clinical isolate. FEMS Microbiol. Lett. 195: Bartowsky, E., and S. Normark Interactions of wild-type and mutant AmpR of Citrobacter freundii with target DNA. Mol. Microbiol. 10: Bartowsky, E., and S. Normark Purification and mutant analysis of Citrobacter freundii AmpR, the regulator for chromosomal AmpC betalactamase. Mol. Microbiol. 5: Bellido, F., J. C. Pechere, and R. E. W. Hancock Reevaluation of the factors involved in the efficacy of new beta-lactams against Enterobacter cloacae. Antimicrob. Agents Chemother. 35: Bert, F., C. Branger, and N. Lambert-Zechovsky Identification of PSE and OXA beta-lactamase genes in Pseudomonas aeruginosa using PCR-restriction fragment length polymorphism. J. Antimicrob. Chemother. 50: Bisbe, J., J. M. Gatell, and J. Puig Pseudomonas aeruginosa bacteremia: univariate and multivariate analyses of factors influencing the prognosis in 133 episodes. Rev. Infect. Dis. 10: Bishop, R. E., and J. H. Weiner Overproduction, solubilization, purification, and DNA-binding properties of AmpR from Citrobacter freundii. Eur. J. Biochem. 213: Blanc, D. S., C. Petignat, B. Janin, J. Bille, and P. Fancioli Frequency and molecular diversity of Pseudomonas aeruginosa upon admission and during hospitalization: a prospective epidemiologic study. Clin. Microbiol. Infect. 4: Bonten, M. J., D. C. Bergmans, H. Speijer, and E. E. Stobberingh Characteristics of polyclonal endemicity of Pseudomonas aeruginosa colonization in intensive care units. Implications for infection control. Am. J. Crit. Care Med. 160: Boutoille, D., S. Corvec, N. Caroff, C. Giraudeau, E. Espaze, J. Caillon, P. Plesiat, and A. Reynaud Detection of an IS21 insertion sequence in the mexr gene of Pseudomonas aeruginosa increasing beta-lactam resistance. FEMS Microbiol. Lett. 230: Breidenstein, E. B. M., B. K. Khaira, I. Wiegand, J. Overhage, and R. E. Hancock Complex ciprofloxacin resistome revealed by screening a Pseudomonas aeruginosa mutant library for altered susceptibility. Antimicrob. Agents Chemother. 52: Brown, S. D., and M. M. Traczewski Comparative in vitro antimicrobial activity of a new carbapenem, doripenem: tentative disc diffusion criteria and quality control. J. Antimicrob. Chemother. 55: Caille, O., C. Rossier, and K. Perron A copper-activated two-component system interacts with zinc and imipenem resistance in Pseudomonas aeruginosa. J. Bacteriol. 189: Campbell, J. I. A., O. Cioufu, and N. Hoiby Pseudomonas aeruginosa isolates from patients with cystic fibrosis have different beta-lactamase expression phenotypes but are homogenous in the ampc-ampr genetic region. Antimicrob. Agents Chemother. 41: Cao, L., R. Srikumar, and K. Poole MexAB-OprM hyperexpression in NalC-type multidrug-resistant Pseudomonas aeruginosa: identification and characterization of nalc gene encoding a repressor of PA3720-PA3719. Mol. Microbiol. 53: Carmeli, Y., N. Troillet, A. W. Karchmer, and M. H. Samore Health and economic outcomes of antibiotic resistant Pseudomonas aeruginosa. Arch. Intern. Med. 159: Castang, S., H. R. McManus, K. H. Turner, and S. L. Dove H-NS family members function coordinately in an opportunistic pathogen. Proc. Natl. Acad. Sci. USA 105: Castanheira, M., R. E. Mendes, T. R. Walsh, A. C. Gales, and R. N. Jones Emergence of the extended-spectrum beta-lactamase GES-1 in a Pseudomonas aeruginosa strain from Brazil: report from the Sentry antimicrobial surveillance program. Antimicrob. Agents Chemother. 48: Castanheira, M., M. A. Toleman, R. N. Jones, F. J. Schmidt, and T. R. Walsh Molecular characterization of a beta-lactamase gene, bla GIM-1, encoding a new subclass of metallo-beta-lactamase. Antimicrob. Agents Chemother. 48: Chen, H., J. Hu, P. R. Chen, L. Lan, Z. Li, L. M. Hicks, A. R. Dinner, and C. He The Pseudomonas aeruginosa multidrug efflux regulator MexR uses an oxidation-sensing mechanism. Proc. Natl. Acad. Sci. USA 105: Cheng, Q., and J. T. Park Substrate specificity of the AmpG permease required for recycling of cell wall anhydro-muropeptides. J. Bacteriol. 184: Chuanchuen, R., J. B. Gaynor, R. Karkhoff-Schweizer, and H. P. Schweizer Molecular characterization of MexL, the transcriptional repressor of the mexjk multidrug efflux operon in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 49: Chuanchuen, R., T. Murata, N. Gotoh, and H. P. Schweizer Substratedependent utilization of OprM and OpmH by the Pseudomonas aeruginosa MexJK efflux pump. Antimicrob. Agents Chemother. 49: Chuanchuen, R., C. T. Narasaki, and H. P. Schweizer The MexJK efflux pump of Pseudomonas aeruginosa requires OprM for antibiotic efflux but not for efflux of triclosan. J. Bacteriol. 184: Colom, K., A. Fdz-Aranguiz, E. Suinaga, and R. Cisterna Emergence of resistance to beta-lactam agents in Pseudomonas aeruginosa with group 1 beta-lactamases in Spain. Eur. J. Clin. Microbiol. Infect. Dis. 14: Daigle, D. M., L. Cao, S. Fraud, M. S. Wilke, A. Pacey, R. Klinoski, N. C. Strynadka, C. R. Dean, and K. Poole Protein modulator of multidrug efflux gene expression in Pseudomonas aeruginosa. J. Bacteriol. 189: Danel, F., L. M. Hall, D. Gur, H. E. Akalin, and D. M. Livermore Transferable production of PER-1 beta-lactamase in Pseudomonas aeruginosa. J. Antimicrob. Chemother. 35: Dean, C. R., M. A. Visalli, S. J. Projan, P. E. Sum, and P. A. Bradford Efflux-mediated resistance to tigecycline (GAR-936) in Pseudomonas aeruginosa PAO1. Antimicrob. Agents Chemother. 47: De Kievit, T. R., M. D. Parkins, R. J. Gillis, R. Srikumar, H. Ceri, K. Poole, B. H. Iglewski, and D. G. Storey Multidrug efflux pumps: expression patterns and contribution to antibiotic resistance in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 45: Dietz, H., D. Pfeifle, and B. Wiedemann Location of N-acetylmuramyl-L-alanyl-D-glutamylmesodiaminopimelic acid, presumed signal molecule for beta-lactamase induction, in the bacterial cell. Antimicrob. Agents Chemother. 40: Dietz, H., D. Pfeifle, and B. Wiedemann The signal molecule for beta-lactamase induction in Enterobacter cloacae is the anhydromuramylpentapeptide. Antimicrob. Agents Chemother. 41: Dimatatac, E. L., M. M. Alejandria, C. Montalban, C. Pineda, C. Ang, and R. Delino Clinical outcomes and costs of care of antibiotic resistant Pseudomonas aeruginosa infections. Philipp. J. Microbiol. Infect. Dis. 32: Doi, Y., D. de Oliveira Garcia, J. Adams, and D. Paterson Coproduction of novel 16S rrna methylase RmtD and metallo-beta-lactamase SPM-1 in a panresistant Pseudomonas aeruginosa isolate from Brazil. Antimicrob. Agents Chemother. 51: Doi, Y., K. Yokoyama, K. Yamane, J. Wachino, N. Shibata, T. Yagi, K. Shibayama, H. Kato, and Y. Arakawa Plasmid-mediated 16S rrna methylase in Serratia marcescens conferring high-level resistance to aminoglycosides. Antimicrob. Agents Chemother. 48: Dotsch, A., T. Becker, C. Pommerenke, Z. Magnowska, L. Jansch, and S. Haussler Genomewide identification of genetic determinants of antimicrobial drug resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 53: Ehrhardt, A. F., and C. C. Sanders Beta-lactam resistance amongst Enterobacter species. J. Antimicrob. Chemother. 32(Suppl. B): Ehrhardt, A. F., C. C. Sanders, J. R. Romero, and J. S. Leser Sequencing and analysis of four new Enterobacter ampd alleles. Antimicrob. Agents Chemother. 40: El Amin, N., C. G. Giske, S. Jalal, B. Keijser, G. Kronvall, and B. Wretlind Carbapenem resistance mechanisms in Pseudomonas aeruginosa: alterations of porin OprD and efflux proteins do not fully explain resistance patterns observe in clinical isolates. APMIS 113: Emori, T. G., and R. P. Gaynes An overview of nosocomial infections, including the role of the microbiology laboratory. Clin. Microbiol. Rev. 6: Erol, S., U. Altoparlak, M. N. Akcay, F. Celebi, and M. Parlak Changes of microbial flora and wound colonization in burned patients. Burns 30: Eron, L. J., R. I. Goldenberg, D. M. Poretz, and C. H. Park Piperacillin therapy for Pseudomonas infections. South. Med. J. 76: Evans, J. C., and H. Segal A novel insertion sequence, ISPA26, in oprd of Pseudomonas aeruginosa is associated with carbapenem resistance. Antimicrob. Agents Chemother. 51: Evans, K., L. Adewoye, and K. Poole MexR repressor of the mexaboprm multidrug efflux operon of Pseudomonas aeruginosa: identification of MexR binding sites in the mexa-mexr intergenic region. J. Bacteriol. 183: Evans, K., and K. Poole The MexA-MexB-OprM multidrug efflux system of Pseudomonas aeruginosa is growth-phase regulated. FEMS Microbiol. Lett. 173: Flamm, R. K., M. K. Weaver, C. Thornsberry, M. E. Jones, J. A. Karlowsky, and D. F. Sahm Factors associated with relative rates of antibiotic

24 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 605 resistance in Pseudomonas aeruginosa isolates tested in clinical laboratories in the United States from 1999 to Antimicrob. Agents Chemother. 48: Fraud, S., A. J. Campigotto, Z. Chen, and K. Poole MexCD-OprJ multidrug efflux system of Pseudomonas aeruginosa: involvement in chlorhexidine resistance and induction by membrane-damaging agents dependent upon the AlgU stress response sigma factor. Antimicrob. Agents Chemother. 52: Freeman, L Chronic general infection with the Bacillus pyocyaneus. Ann. Surg. 64: Fritsche, T. R., H. S. Sader, M. A. Toleman, T. R. Walsh, and R. N. Jones Emerging metallo-beta-lactamase-mediated resistances: a summary report from the worldwide SENTRY antimicrobial surveillance program. Clin. Infect. Dis. 41(Suppl. 4):S276 S Fukuda, H., M. Hosaka, S. Iyobe, N. Gotoh, T. Nishino, and K. Hirai nfxc-type quinolone resistance in a clinical isolate of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 39: Gales, A. C., L. C. Menezes, S. Silbert, and H. S. Sader Dissemination in distinct Brazilian regions of an epidemic carbapenem-resistant Pseudomonas aeruginosa producing SPM metallo-beta-lactamase. J. Antimicrob. Chemother. 52: Galimand, M., P. Courvalin, and T. Lambert Plasmid-mediated high-level resistance to aminoglycosides in Enterobacteriaceae due to 16S rrna methylation. Antimicrob. Agents Chemother. 47: Galimand, M., P. Sabtcheva, P. Courvalin, and T. Lambert Worldwide disseminated arma aminoglycoside resistance methylase gene borne by composite transposon Tn1548. Antimicrob. Agents Chemother. 49: Gasink, L. B., N. O. Fishman, M. G. Weiner, I. Nachamkin, W. B. Bilker, and E. Lautenbach Fluoroquinolone-resistant Pseudomonas aeruginosa: assessment of risk factors and clinical impact. Am. J. Med. 119: 526e19 526e Gates, M. L., C. C. Sanders, R. V. Goering, and W. E. Sanders Evidence for multiple forms of type I chromosomal beta-lactamase in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 30: Gaynes, R., and J. R. Edwards Overview of nosocomial infections caused by gram-negative bacilli. Clin. Infect. Dis. 41: Gessard, C Classics in infectious diseases. On the blue and green coloration that appears on bandages. Rev. Infect. Dis. 6(Suppl. 3):S775 S Gillis, R. J., K. G. White, K. H. Choi, V. E. Wagner, H. P. Schweizer, and B. H. Iglewski Molecular basis of azithromycin-resistant Pseudomonas aeruginosa biofilms. Antimicrob. Agents Chemother. 49: Girlich, D., T. Naas, and P. Nordmann Biochemical characterization of the naturally occurring oxacillinase OXA-50 of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 48: Gonzalez-Zorn, B., A. Catalan, J. A. Escudero, L. Dominguez, T. Teshager, C. Porrero, and M. A. Moreno Genetic basis for dissemination of arma. J. Antimicrob. Chemother. 56: Goodell, E. W Recycling of murein by Escherichia coli. J. Bacteriol. 163: Gotoh, N., H. Tsujimoto, M. Tsuda, K. Okamoto, A. Nomura, T. Wada, M. Nakahashi, and T. Nishino Characterization of the MexC-MexD- OprJ multidrug efflux system in delta-mexa-mexb-oprm mutants of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 42: Gribble, M. J., A. W. Chow, S. C. Naiman, J. A. Smith, W. R. Bowie, S. L. Sacks, L. Grossman, N. Buskard, G. H. Growe, and L. H. Plenderleith Prospective randomized trial of piperacillin monotherapy versus carboxypenicillin-aminoglycoside combination regimens in the empirical treatment of serious bacterial infections. Antimicrob. Agents Chemother. 24: Grundstrom, T., and S. Normark Initiation of translation makes attenuation of ampc in E. coli dependent on growth rate. Mol. Gen. Genet. 198: Gutierrez, O., C. Juan, E. Cercenado, F. Navarro, E. Bouza, P. Coll, J. L. Perez, and A. Oliver Molecular epidemiology and mechanisms of carbapenem resistance in Pseudomonas aeruginosa isolates from Spanish hospitals. Antimicrob. Agents Chemother. 51: Hancock, R. E. W Resistance mechanisms in Pseudomonas aeruginosa and other nonfermentative gram-negative bacteria. Clin. Infect. Dis. 27(Suppl. 1):S93 S Hancock, R. E. W., and F. S. Brinkman Function of pseudomonas porins in uptake and efflux. Annu. Rev. Microbiol. 56: Hanson, N. D., and C. C. Sanders Regulation of inducible AmpC beta-lactamase expression among Enterobacteriaceae. Curr. Pharm. Des. 5: Harris, A. A., L. Goodman, and S. Levin Community-acquired Pseudomonas aeruginosa pneumonia associated with the use of a home humidifier. West. J. Med. 141: Hiraoka, M., S. Masuyoshi, S. Mitsuhashi, K. Tomatsu, and M. Inoue Cephalosporinase interactions and antimicrobial activity of BMY , ceftazidime, and cefotaxime. J. Antibiot. 41: Hocquet, D., P. Nordmann, F. El Garch, L. Cabanne, and P. Plesiat Involvement of the MexXY-OprM efflux system in emergence of cefepime resistance in clinical strains of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 50: Holtje, J. V Growth of the stress-bearing and shape-maintaining murein sacculus of Escherichia coli. Microbiol. Mol. Biol. Rev. 62: Holtje, J. V., U. Kopp, A. Ursinus, and B. Wiedemann The negative regulator of beta-lactamase induction AmpD is a N-acetyl-anhydromuramyl-L-alanine amidase. FEMS Microbiol. Lett. 122: Honore, N., M. H. Nicolas, and S. T. Cole Inducible cephalosporinase production in clinical isolates of Enterobacter cloacae is controlled by a regulatory gene that has been deleted from Escherichia coli. EMBO J. 20: Honore, N., M. H. Nicolas, and S. T. Cole Regulation of enterobacterial cephalosporinase production: the role of a membrane-bound sensory transducer. Mol. Microbiol. 3: Hooper, D. C Mechanisms of action and resistance of older and newer fluoroquinolones. Clin. Infect. Dis. 31:S24 S Islam, S., S. Jalal, and B. Wretlind Expression of MexXY efflux pump in amikacin-resistant isolates of Pseudomonas aeruginosa. Clin. Microbiol. Infect. 10: Jacobs, C., J. M. Frere, and S. Normark Cytosolic intermediates for cell wall biosynthesis and degradation control inducible beta-lactam resistance in gram-negative bacteria. Cell 88: Jacobs, C., L. J. Huang, E. Bartowsky, S. Normark, and J. T. Park Bacterial cell wall recycling provides cytosolic muropeptides as effectors for beta-lactamase induction. EMBO J. 13: Jacobs, C., B. Joris, M. Jamin, K. Klarsov, J. Van Beeumen, D. Mengin- Lecreuix, J. van Heijenoort, J. T. Park, S. Normark, and J.-M. Frere AmpD, essential for both beta-lactamase regulation and cell wall recycling, is a novel cytosolic N-acetylmuramyl-L-alanine amidase. Mol. Microbiol. 15: Jacoby, G. A., N. Chow, and K. B. Waites Prevalence of plasmidmediated quinolone resistance. Antimicrob. Agents Chemother. 47: Jalal, S., O. Ciofu, N. Hoiby, N. Gotoh, and B. Wretlind Molecular mechanisms of fluoroquinolone resistance in Pseudomonas aeruginosa isolates from cystic fibrosis patients. Antimicrob. Agents Chemother. 44: Jalal, S., and B. Wretlind Mechanisms of quinolone resistance in clinical strains of Pseudomonas aeruginosa. Microb. Drug Resist. 4: Jeannot, K., S. Elsen, T. Kohler, I. Attree, C. van Delden, and P. Plesiat Resistance and virulence of Pseudomonas aeruginosa clinical strains overproducing the MexCD-OprJ efflux pump. Antimicrob. Agents Chemother. 52: Jeannot, K., M. L. Sobel, F. El Garch, K. Poole, and P. Plesiat Induction of the MexXY efflux pump in Pseudomonas aeruginosa is dependent on drug-ribosome interaction. J. Bacteriol. 187: Jimenez-Lucho, V. E., L. D. Saravolatz, A. A. Meideros, and D. Pohlod Failure of therapy in Pseudomonas endocarditis: selection of resistant mutant. J. Infect. Dis. 154: Jones, M. E., D. C. Draghi, C. Thornsberry, J. A. Karlowsky, D. F. Sahm, and R. P. Wenzel Emerging resistance among bacterial pathogens in the intensive care unit a European and North American surveillance study ( ). Ann. Clin. Microbiol. Antimicrob. 3: Jones, R. N., D. J. Biedenbach, H. S. Sader, T. R. Fritsche, M. A. Toleman, and T. R. Walsh Emerging epidemic of metallo-beta-lactamasemediated resistances. Diagn. Microbiol. Infect. Dis. 51: Juan, C., M. D. Macia, O. Gutierrez, C. Vidal, J. L. Perez, and A. Oliver Molecular mechanisms of beta-lactam resistance mediated by AmpC hyperproduction in Pseudomonas aeruginosa clinical strains. Antimicrob. Agents Chemother. 49: Juan, C., B. Moya, J. L. Perez, and A. Oliver Stepwise upregulation of the Pseudomonas aeruginosa chromosomal cephalosporinase conferring high-level beta-lactam resistance involves three AmpD homologues. Antimicrob. Agents Chemother. 50: Karlowsky, J. A., D. C. Draghi, M. E. Jones, C. Thornsberry, I. R. Friedland, and D. F. Sahm Surveillance for antimicrobial susceptibility among clinical isolates of Pseudomonas aeruginosa and Acinetobacter baumannii from hospitalized patients in the United States, Antimicrob. Agents Chemother. 47: Karlowsky, J. A., L. J. Kelly, C. Thornsberry, M. E. Jones, A. T. Evangelista, I. A. Critchley, and D. F. Sahm Susceptibility to fluoroquinolones among commonly isolated gram-negative bacilli in 2000: TRUST and TSN data for the United States. Int. J. Antimicrob. Agents 19: Kohler, T., S. F. Epp, L. K. Curty, and J. C. Pechere Characterization of MexT, the regulator of the MexE-MexF-OprN multidrug efflux system of Pseudomonas aeruginosa. J. Bacteriol. 181: Kohler, T., M. Kok, M. Michea-Hamzehpour, P. Plesiat, T. Gotoh, T. Nishino, L. K. Curty, and J. C. Pechere Multidrug efflux in intrinsic resistance to trimethoprim and sulfamethoxazole in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 40: Kohler, T., M. Michea-Hamzehpour, S. F. Epp, and J. C. Pechere Carbapenem activities against Pseudomonas aeruginosa: respective contributions of OprD and efflux systems. Antimicrob. Agents Chemother. 43: Kohler, T., M. Michea-Hamzehpour, U. Henz, N. Gotoh, L. K. Curty, and

25 606 LISTER ET AL. CLIN. MICROBIOL. REV. J. C. Pechere Characterization of MexE-MexF-OprN, a positively regulated multidrug efflux system of Pseudomonas aeruginosa. Mol. Microbiol. 23: Kohler, T., C. van Delden, L. K. Curty, M. M. Hamzehpour, and J. C. Pechere Overexpression of the MexEF-OprN multidrug efflux system affects cell-to-cell signaling in Pseudomonas aeruginosa. J. Bacteriol. 183: Kolleff, M. H., A. Shorr, Y. P. Tabak, V. Gupta, L. Z. Liu, and R. S. Johannes Epidemiology and outcomes of health-care-associated pneumonia: results from a large US database of culture-positive pneumonia. Chest 128: Kong, K.-F., S. R. Jayawardena, S. D. Indulkar, A. del Puerto, C.-L. Koh, N. Hoiby, and K. Mathee Pseudomonas aeruginosa AmpR is a global transcriptional factor that regulates expression of AmpC and PoxB betalactamases, proteases, quorum sensing, and other virulence factors. Antimicrob. Agents Chemother. 49: Kopp, U., B. Wiedemann, S. Linquist, and S. Normark Sequences of wild-type and mutant ampd genes of Citrobacter freundii and Enterobacter cloacae. Antimicrob. Agents Chemother. 37: Korfmann, G., and C. C. Sanders ampg is essential for high-level expression of AmpC beta-lactamase activity in Enterobacter cloacae. Antimicrob. Agents Chemother. 33: Korfmann, G., C. C. Sanders, and E. S. Moland Altered phenotypes associated with ampd mutations in Enterobacter cloacae. Antimicrob. Agents Chemother. 35: Kuga, A., R. Okamoto, and M. Inoue ampr gene mutations that greatly increase class C beta-lactamase activity in Enterobacter cloacae. Antimicrob. Agents Chemother. 44: Labia, R., A. Morand, and J. Peduzzi Timentin and beta-lactamases. Antimicrob. Agents Chemother. 17(Suppl. C): Landman, D., S. Bratu, S. Kochar, M. Panwar, M. Trehan, M. Doymaz, and J. Quale Evolution of antimicrobial resistance among Pseudomonas aeruginosa, Acinetobacter baumannii, and Klebsiella pneumoniae in Brooklyn, N.Y. J. Antimicrob. Chemother. 60: Langaee, T. Y., M. Dargis, and A. Huletsky An ampd gene in Pseudomonas aeruginosa encodes a negative regulator of AmpC beta-lactamase expression. Antimicrob. Agents Chemother. 42: Langaee, T. Y., L. Gagnon, and A. Huletsky Inactivation of the ampd gene in Pseudomonas aeruginosa leads to moderate-basal-level and hyperinducible AmpC beta-lactamase expression. Antimicrob. Agents Chemother. 44: Lee, J. K., Y. S. Lee, Y. K. Park, and B. S. Kim Alterations in the GyrA and GyrB subunits of topoisomerase II and the ParC and ParE subunits of topoisomerase IV in ciprofloxacin-resistant clinical isolates of Pseudomonas aeruginosa. Int. J. Antimicrob. Agents 25: Lee, K., G. Y. Ha, B. M. Shin, J. J. Kim, J. O. Kang, S. J. Jang, D. Yong, and Y. Chong Metallo-beta-lactamase-producing gram-negative bacilli in Korean Nationwide Surveillance of Antimicrobial Resistance group hospitals 2003: continued prevalence of VIM-producing Pseudomonas spp. and increase of IMP-producing Acinetobacter spp. Diagn. Microbiol. Infect. Dis. 50: Letendre, E. D., R. Mantha, and P. L. Turgeon Selection of resistance by piperacillin during Pseudomonas aeruginosa endocarditis. J. Antimicrob. Chemother. 22: Li, X. Z., N. Barre, and K. Poole Influence of MexA-MexB-OprM multidrug efflux system on expression of the MexC-MexD-OprJ and MexE- MexF-OprN multidrug efflux systems in Pseudomonas aeruginosa. J. Antimicrob. Chemother. 46: Li, X. Z., H. Nikaido, and K. Poole Role of mexa-mexb-oprm in antibiotic efflux in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 39: Li, X. Z., L. Zhang, R. Srikumar, and K. Poole Beta-lactamase inhibitors are substrates for the multidrug efflux pumps of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 42: Li, Y., T. Mima, Y. Komori, Y. Morita, T. Kuroda, T. Mizushima, and T. Tsuchiya A new member of the tripartite multidrug efflux pumps, MexVW-OprM, in Pseudomonas aeruginosa. J. Antimicrob. Chemother. 52: Lindberg, F., S. Lindquist, and S. Normark Genetic basis of induction and overproduction of chromosomal class I beta-lactamase in nonfastidious gram-negative bacilli. Rev. Infect. Dis. 10: Lindberg, F., S. Lindquist, and S. Normark Inactivation of the ampd gene causes semiconstitutive overproduction of the inducible Citrobacter freundii beta-lactamase. J. Bacteriol. 169: Lindberg, F., L. Westman, and S. Normark Regulatory components in Citrobacter freundii ampc beta-lactamase induction. Proc. Natl. Acad. Sci. USA 82: Lindquist, S., F. Lindberg, and S. Normark Binding of the Citrobacter freundii AmpR regulator to a single DNA site provides both autoregulation and activation of the inducible AmpC beta-lactamase. J. Bacteriol. 171: Lindquist, S., K. Weston-Hafer, H. Schmidt, C. Pul, G. Korfmann, J. Erickson, C. C. Sanders, H. H. Martin, and S. Normark AmpG, a signal transducer in chromosomal beta-lactamase induction. Mol. Microbiol. 9: Lister, P. D., V. M. Gardner, and C. C. Sanders Clavulanate induces expression of the Pseudomonas aeruginosa AmpC cephalosporinase at physiologically relevant concentrations and antagonizes the antibacterial activity of ticarcillin. Antimicrob. Agents Chemother. 43: Lister, P. D., and D. J. Wolter Levofloxacin-imipenem combination prevents the emergence of resistance among clinical isolates of Pseudomonas aeruginosa. Clin. Infect. Dis. 40:S105 S Lister, P. D., D. J. Wolter, P. A. Wickman, and M. D. Reisbig Levofloxacin/imipenem prevents the emergence of high-level resistance among Pseudomonas aeruginosa strains already lacking susceptibility to one or both drugs. J. Antimicrob. Chemother. 57: Livermore, D. M Interplay of impermeability and chromosomal betalactamase activity in imipenem-resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 36: Livermore, D. M Multiple mechanisms of antimicrobial resistance in Pseudomonas aeruginosa: our worst nightmare. Clin. Infect. Dis. 34: Livermore, D. M The threat from the pink corner. Ann. Med. 35: Livermore, D. M., and N. Woodford The beta-lactamase threat in Enterobacteriaceae, Pseudomonas, and Acinetobacter. Trends Microbiol. 14: Livermore, D. M., and N. Woodford Carbapenemases: a problem in waiting? Curr. Opin. Microbiol. 3: Livermore, D. M., and Y.-J. Yang Beta-lactamase lability and inducer power of newer beta-lactam antibiotics in relation to their activity against beta-lactamase-inducibility mutants of Pseudomonas aeruginosa. J. Infect. Dis. 4: Llanes, C., D. Hocquet, C. Vogne, D. Benali-Baitich, C. Neuwirth, and P. Plesiat Clinical strains of Pseudomonas aeruginosa overproducing MexAB-OprM and MexXY efflux pumps simultaneously. Antimicrob. Agents Chemother. 48: Lodge, J. M., S. J. W. Busby, and L. J. Piddock Investigation of the Pseudomonas aeruginosa ampr gene and its role at the chromosomal ampc beta-lactamase promoter. FEMS Microbiol. Lett. 111: Lodge, J. M., S. D. Minchin, L. J. Piddock, and S. J. W. Busby Cloning, sequencing and analysis of the structural gene and regulatory region of the Pseudomonas aeruginosa chromosomal ampc beta-lactamase. Biochem. J. 272: Lynch, M. J., G. L. Drusano, and H. L. Mobley Emergence of resistance to imipenem in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 31: Mammeri, H., M. Galleni, and P. Nordmann Role of the Ser-287- Asn replacement in the hydrolysis spectrum extension of AmpC betalactamases in Escherichia coli. Antimicrob. Agents Chemother. 53: Mammeri, H., P. Nordmann, A. Berkani, and F. Eb Contribution of extended-spectrum AmpC (ESAC) beta-lactamases to carbapenem resistance in Escherichia coli. FEMS Microbiol. Lett. 282: Mammeri, H., L. Poirel, P. Bemer, H. Drugeon, and P. Nordmann Resistance to cefepime and cefpirome due to a 4-amino-acid deletion in the chromosome-encoded AmpC beta-lactamase of a Serratia marcescens clinical isolate. Antimicrob. Agents Chemother. 48: Margaret, B. S., G. L. Drusano, and H. C. Standiford Emergence of resistance to carbapenem antibiotics in Pseudomonas aeruginosa. J. Antimicrob. Chemother. 24(Suppl. A): Maseda, H., K. Saito, A. Nakajima, and T. Nakae Variation of the mext gene, a regulator of the MexEF-OprN efflux pump expression in wild-type strains of Pseudomonas aeruginosa. FEMS Microbiol. Lett. 192: Maseda, H., I. Sawada, K. Saito, H. Uchiyama, T. Nakae, and N. Nomura Enhancement of the mexab-oprm efflux pump expression by a quorum-sensing autoinducer and its cancellation by a regulator, MexT, of the mexef-oprn efflux pump operon in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 48: Masterton, R. G., P. J. Garner, N. A. Harrison, and D. J. Rainford Timentin resistance. Lancet ii: Masuda, N., N. Gotoh, C. Ishii, E. Sakagawa, S. Ohya, and T. Nishino Interplay between chromosomal beta-lactamase and the MexAB- OprM efflux system in intrinsic resistance to beta-lactams in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 43: Masuda, N., N. Gotoh, S. Ohya, and T. Nishino Quantitative correlation between susceptibility and OprJ production in NfxB mutants of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 40: Masuda, N., E. Sakagawa, and S. Ohya Outer membrane proteins responsible for multiple drug resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 39: Masuda, N., E. Sakagawa, S. Ohya, N. Gotoh, and T. Nishino Hypersusceptibility of the Pseudomonas aeruginosa nfxb mutant to beta-lactams due to reduced expression of the AmpC beta-lactamase. Antimicrob. Agents Chemother. 45:

26 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA Masuda, N., E. Sakagawa, S. Ohya, N. Gotoh, H. Tsujimoto, and T. Nishino Contribution of the MexX-MexY-OprM efflux system to intrinsic resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 44: Matsumura, N., S. Minami, and S. Mitsuhashi Sequences of homologous beta-lactamases from clinical isolates of Serratia marcescens with different substrate specificities. Antimicrob. Agents Chemother. 42: Matsuo, Y., S. Eda, N. Gotoh, E. Yoshihara, and T. Nakae MexZmediated regulation of mexxy multidrug efflux pump expression in Pseudomonas aeruginosa by binding on the mexz-mexx intergenic DNA. FEMS Microbiol. Lett. 238: Mavroidi, A., E. Tzelepi, A. Tsakris, V. Miriagou, D. Sofianou, and L. S. Tzouvelekis An integron associated beta-lactamase (IBC-2) from Pseudomonas aeruginosa is a variant of the extended-spectrum beta-lactamase IBC-1. J. Antimicrob. Chemother. 48: Micek, S. T., A. E. Lloyd, D. J. Ritchie, R. M. Reichley, V. J. Fraser, and M. H. Kollef Pseudomonas aeruginosa bloodstream infection: importance of appropriate initial antimicrobial treatment. Antimicrob. Agents Chemother. 49: Mima, T., S. Joshi, M. Gomez-Escalada, and H. P. Schweizer Identification and characterization of TriABC-OpmH, a triclosan efflux pump of Pseudomonas aeruginosa requiring two membrane fusion proteins. J. Bacteriol. 189: Mima, T., H. Sekiya, T. Mizushima, T. Kuroda, and T. Tsuchiya Gene cloning and properties of the RND-type multidrug efflux pump, MexPQ-OpmE and MexMN-OprM from Pseudomonas aeruginosa. Microbiol. Immunol. 49: Mine, T., Y. Morita, T. Kataoka, T. Mizushima, and T. Tsuchiya Expression in Escherichia coli of a new multidrug efflux pump, MexXY, from Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 43: Morita, Y., L. Cao, V. C. Gould, M. B. Avison, and K. Poole nald encodes a second repressor of the mexab-oprm multidrug efflux operon of Pseudomonas aeruginosa. J. Bacteriol. 188: Morita, Y., N. Kimura, T. Mima, T. Mizushima, and T. Tsuchiya Roles of MexXY- and MexAB-multidrug efflux pumps in intrinsic multidrug resistance of Pseudomonas aeruginosa. J. Gen. Appl. Microbiol. 47: Morita, Y., Y. Komori, T. Mima, T. Kuroda, T. Mizushima, and T. Tsuchiya Construction of a series of mutants lacking all of the four major mex operons for multidrug efflux pumps or possessing each one of the operons from Pseudomonas aeruginosa PAO1: MexCD-OprJ is an inducible pump. FEMS Microbiol. Lett. 202: Morita, Y., M. L. Sobel, and K. Poole Antibiotic inducibility of the MexXY multidrug efflux system of Pseudomonas aeruginosa: involvement of the antibiotic-inducible PA5471 gene product. J. Bacteriol. 188: Morrison, A. J., and R. P. Wenzel Epidemiology of infections due to Pseudomonas aeruginosa. Rev. Infect. Dis. 6(Suppl. 3):S627 S Mouneimne, H., J. Robert, V. Jarlier, and E. Cambau Type II topoisomerase mutations in ciprofloxacin-resistant strains of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 43: Moya, B., A. Dotsch, C. Juan, J. Blazquez, L. Zamorano, S. Haussler, and A. Oliver Beta-lactam resistance response triggered by inactivation of a nonessential penicillin-binding protein. PLoS Pathog. 5:e Moya, B., C. Juan, S. Alberti, J. L. Perez, and A. Oliver Benefit of having multiple ampd genes for acquiring beta-lactam resistance without losing fitness and virulence in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 52: Murata, T., N. Gotoh, and T. Nishino Characterization of outer membrane efflux proteins OpmE, OpmD and OpmB of Pseudomonas aeruginosa: molecular cloning and development of specific antisera. FEMS Microbiol. Lett. 217: Mushtaq, S., Y. Ge, and D. M. Livermore Doripenem versus Pseudomonas aeruginosa in vitro: activity against characterized isolates, mutants, and transconjugants and resistance selection potential. Antimicrob. Agents Chemother. 48: Naas, T., and P. Nordmann OXA-type beta-lactamases. Curr. Pharm. Des. 5: National Nosocomial Infections Surveillance System National Nosocomial Infections Surveillance (NNIS) System report, data summary from October 1986 to April 1998, issued June Am. J. Infect. Control 26: Nikaido, H Multidrug efflux pumps of gram-negative bacteria. J. Bacteriol. 178: Nikaido, H Outer membrane barrier as a mechanism of antimicrobial resistance. Antimicrob. Agents Chemother. 33: Niumsup, P., A. M. Simm, K. Nurmahomed, T. R. Walsh, P. M. Bennett, and M. B. Avison Genetic linkage of the penicillinase gene, amp, and blrab, encoding the regulator of beta-lactamase expression in Aeromonas spp. J. Antimicrob. Chemother. 51: Nordmann, P., and T. Nass Sequence analysis of PER-1 extendedspectrum beta-lactamase from Pseudomonas aeruginosa and comparison with class A beta-lactamases. Antimicrob. Agents Chemother. 38: Nordmann, P., and L. Poirel Emerging carbapenemases in gramnegative aerobes. Clin. Microbiol. Infect. 8: Normark, S., T. Edlund, T. Grundstrom, S. Bergstrom, and H. Wolf-Watz Escherichia coli K-12 mutants hyperproducing chromosomal betalactamase by gene repetitions. J. Bacteriol. 132: Obritsch, M. D., D. N. Fish, R. MacLaren, and R. Jung National surveillance of antimicrobial resistance in Pseudomonas aeruginosa isolates obtained from intensive care unit patients from 1993 to Antimicrob. Agents Chemother. 48: Ochs, M. M., C.-D. Lu, R. E. W. Hancock, and A. T. Abdelal Amino acid-mediated induction of the basic amino acid-specific outer membrane porin OprD from Pseudomonas aeruginosa. J. Bacteriol. 181: Ochs, M. M., M. P. McCusker, M. Bains, and R. E. Hancock Negative regulation of the Pseudomonas aeruginosa outer membrane porin OprD selective for imipenem and basic amino acids. Antimicrob. Agents Chemother. 43: Oh, H., J. Stenhoff, S. Jalal, and B. Wretlind Role of efflux pumps and mutations in genes for topoisomerases II and IV in fluoroquinolone-resistant Pseudomonas aeruginosa strains. Microb. Drug Resist. 9: Ohara, T., and K. Itoh Significance of Pseudomonas aeruginosa colonization of the gastrointestinal tract. Intern. Med. 42: Oliva, B., P. M. Bennett, and I. Chopra Penicillin-binding protein 2 is required for induction of the Citrobacter freundii class I chromosomal beta-lactamase in Escherichia coli. Antimicrob. Agents Chemother. 33: Pai, H., J. Kim, J. H. Lee, K. W. Choe, and N. Gotoh Carbapenem resistance mechanisms in Pseudomonas aeruginosa clinical isolates. Antimicrob. Agents Chemother. 45: Palma, M., J. Zurita, J. A. Ferraras, S. Worgall, D. H. Larone, L. Shi, F. Campagne, and L. E. Quadri Pseudomonas aeruginosa SoxR does not conform to the archetypal paradigm for SoxR-dependent regulation of the bacterial oxidative stress adaptive response. Infect. Immun. 73: Park, C. H., A. Robicsek, G. A. Jacoby, D. Sahm, and D. C. Hooper Prevalence in the United States of aac(6 )-Ib-cr, encoding a ciprofloxacinmodifying enzyme. Antimicrob. Agents Chemother. 50: Peleg, A. Y., C. Franklin, J. Bell, and D. W. Spelman Emergence of IMP-4 metallo-beta-lactamase in a clinical isolate from Australia. J. Antimicrob. Chemother. 54: Perez, F. J., C. Gimeno, D. Navarro, and J. Garcia-de-Lomas Meropenem permeation through the outer membrane of Pseudomonas aeruginosa can involve pathways other than OprD porin channel. Chemotherapy 42: Perron, K., O. Caille, C. Rossier, C. Van Delden, J. L. Dumas, and T. Kohler CzcR-CzcS, a two-component system involved in heavy metal and carbapenem resistance in Pseudomonas aeruginosa. J. Biol. Chem. 279: Pfeifle, D., E. Janas, and B. Wiedemann Role of penicillin-binding proteins in the initiation of AmpC beta-lactamase expression in Enterobacter cloacae. Antimicrob. Agents Chemother. 44: Philippon, L. N., T. Naas, A.-T. Bouthors, V. Barakett, and P. Nordmann OXA-18, a class D clavulanic acid-inhibited extended-spectrum betalactamase from Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 41: Picao, R. C., L. Poirel, A. C. Gales, and P. Nordmann Further identification of CTX-M-2 extended-spectrum beta-lactamase in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 53: Piddock, L. J Multidrug-resistance efflux pumps not just for resistance. Nat. Rev. Microbiol. 4: Piddock, L. J., and D. J. Griggs Selection and characterization of cefepime-resistant gram-negative bacteria. J. Antimicrob. Chemother. 28: Pirnay, J.-P., D. De Vos, D. Mossialos, A. Vanderkelen, P. Cornelis, and M. Zizi Analysis of the Pseudomonas aeruginosa oprd gene from clinical and environmental isolates. Environ. Microbiol. 4: Pitt, T. L Pseudomonas, Burkholderia, and related genera, p In B. I. Duerden (ed.), Microbiology and microbial infections, vol. 2. Oxford University Press Inc., New York, NY Poirel, L., V. O. Rotimi, and E. M. Mokaddas VEB-1-like extended spectrum beta-lactamases in Pseudomonas aeruginosa, Kuwait. Emerg. Infect. Dis. 17: Poirel, L., M. Van De Loo, H. Mammeri, and P. Nordmann Association of plasmid-mediated quinolone resistance with extended-spectrum beta-lactamase VEB-1. Antimicrob. Agents Chemother. 49: Pollack, M Pseudomonas aeruginosa, p In G. L. Mandell, R. Dolan, and J. E. Bennett (ed.), Principles and practices of infectious diseases. Churchill Livingstone, New York, NY Poole, K Aminoglycoside resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 49: Poole, K Efflux-mediated resistance to fluoroquinolones in gramnegative bacteria. Antimicrob. Agents Chemother. 44:

27 608 LISTER ET AL. CLIN. MICROBIOL. REV Poole, K Outer membranes and efflux: the path to multidrug resistance in gram-negative bacteria. Curr. Pharm. Biotechnol. 3: Poole, K., N. Gotoh, H. Tsujimoto, Q. Zhao, A. Wada, T. Yamasaki, S. Neshat, J. Yamagishi, X. Z. Li, and T. Nishino Overexpression of the mexc-mexd-oprj efflux operon in nfxb-type multidrug resistant strains of Pseudomonas aeruginosa. Mol. Microbiol. 21: Poole, K., D. E. Heinrichs, and S. Neshat Cloning and sequence analysis of an EnvCD homologue in Pseudomonas aeruginosa: regulation by iron and possible involvement in the secretion of the siderophore pyoverdine. Mol. Microbiol. 10: Poole, K., and R. Srikumar Multidrug efflux in Pseudomonas aeruginosa: components, mechanisms, and clinical significance. Curr. Top. Med. Chem. 1: Poole, K., K. Tetro, Q. Zhao, S. Neshat, D. E. Heinrichs, and N. Bianco Expression of the multidrug resistance operon mexa-mexb-oprm in Pseudomonas aeruginosa: mexr encodes a regulator of operon expression. Antimicrob. Agents Chemother. 40: Pumbwe, L., and L. J. V. Piddock Two efflux systems expressed simultaneously in multidrug-resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 44: Queenan, A. M., W. Shang, M. Kania, M. G. P. Page, and K. Bush Interactions of ceftobiprole with beta-lactamases from molecular classes A to D. Antimicrob. Agents Chemother. 51: Quinn, J. P., E. J. Dudek, C. A. DiVincenzo, D. A. Lucks, and S. A. Lerner Emergence of resistance to imipenem during therapy for Pseudomonas aeruginosa infections. J. Infect. Dis. 154: Quinn, J. P., A. E. Studemeister, C. A. DiVincenzo, and S. A. Lerner Resistance to imipenem in Pseudomonas aeruginosa: clinical experience and biochemical mechanisms. Rev. Infect. Dis. 10: Rhomberg, P. R., L. M. Deshpande, J. T. Kirby, and R. N. Jones Activity of meropenem as serine carbapenemases evolve in US medical centers: monitoring report from the MYSTIC program (2006). Diagn. Microbiol. Infect. Dis. 59: Rhomberg, P. R., and R. N. Jones Contemporary activity of meropenem and comparator broad-spectrum agents: MYSTIC program report from the United States component (2005). Diagn. Microbiol. Infect. Dis. 57: Richards, M. J., J. R. Edwards, D. H. Culver, R. P. Gaynes, et al Nosocomial infections in medical intensive care units in the United States. Crit. Care Med. 5: Richards, M. J., J. R. Edwards, D. H. Culver, R. P. Gaynes, et al Nosocomial infections in pediatric intensive care units in the United States. Pediatrics 103:e Robicsek, A., J. Strahilevitz, D. Sahm, G. A. Jacoby, and D. C. Hooper qnr prevalence in ceftazidime-resistant Enterobacteriaceae isolates from the United States. Antimicrob. Agents Chemother. 50: Rodriquez-Martinez, J.-M., L. Poirel, and P. Nordmann Extendedspectrum cephalosporinases in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 53: Roh, I. K., I. J. Kim, J. H. Chung, and S. M. Byun Affinity purification and binding characteristics of Citrobacter freundii AmpR, the transcriptional regulator of the ampc beta-lactamase gene. Biotechnol. Appl. Biochem. 23: Saier, M. H., I. T. Paulsen, M. K. Sliwinski, S. S. Pao, R. A. Skurray, and H. Nikaido Evolutionary origins of multidrug and drug-specific efflux pumps in bacteria. FASEB J. 12: Saito, K., H. Akama, E. Yoshihara, and T. Nakae Mutations affecting DNA-binding activity of the MexR repressor of mexr-mexa-mexb-oprm operon expression. J. Bacteriol. 185: Saito, K., S. Eda, H. Maseda, and T. Nakae Molecular mechanism of MexR-mediated regulation of MexAB-OprM efflux pump expression in Pseudomonas aeruginosa. FEMS Microbiol. Lett. 195: Sakyo, S., H. Tomita, K. Tanimoto, S. Fujimoto, and Y. Ike Potency of carbapenems for the prevention of carbapenem-resistant mutants of Pseudomonas aeruginosa. J. Antibiot. 59: Sanders, C. C Beta-lactamases of gram-negative bacteria: new challenges for new drugs. Clin. Infect. Dis. 14: Sanders, C. C Cefepime: the next generation? Clin. Infect. Dis. 17: Sanders, C. C., P. A. Bradford, A. F. Ehrhardt, K. Bush, K. D. Young, T. A. Henderson, and W. E. Sanders Penicillin-binding proteins and induction of AmpC beta-lactamase. Antimicrob. Agents Chemother. 41: Sanders, C. C., M. L. Gates, and W. E. Sanders Heterogeneity of class I beta-lactamase expression in clinical isolates of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 32: Sanders, C. C., and E. C. Sanders Emergence of resistance during therapy with newer beta-lactam antibiotics: role of inducible beta-lactamases and implications for the future. Rev. Infect. Dis. 5: Sanders, C. C., and W. E. Sanders Type I beta-lactamases of gramnegative bacteria: interactions with beta-lactam antibiotics. J. Infect. Dis. 154: Sanders, W. E., and C. C. Sanders Inducible beta-lactamases: clinical and epidemiological implications for use of newer cephalosporins. Rev. Infect. Dis. 10: Satake, S., H. Yoneyama, and T. Nakae Role of OmpD2 and chromosomal beta-lactamase in carbapenem resistance in clinical isolates of Pseudomonas aeruginosa. J. Antimicrob. Chemother. 28: Sawada, I., H. Maseda, T. Nakae, H. Uchiyama, and N. Nomura A quorum-sensing autoinducer enhances the mexab-oprm efflux pump expression without the MexR-mediated regulation of Pseudomonas aeruginosa. Microbiol. Immunol. 48: Schmidtke, A. J., and N. D. Hanson Model system to evaluate the effect of ampd mutations on AmpC-mediated beta-lactam resistance. Antimicrob. Agents Chemother. 50: Schmidtke, A. J., and N. D. Hanson Role of ampd homologs in overproduction of AmpC in clinical isolates of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 52: Schurek, K. N., A. K. Marr, P. K. Taylor, I. Wiegand, L. Semenec, B. K. Khaira, and R. E. W. Hancock Novel genetic determinants of lowlevel aminoglycoside resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 52: Schweizer, H. P Efflux as a mechanism of resistance to antimicrobials in Pseudomonas aeruginosa and related bacteria: unanswered questions. Genet. Mol. Res. 2: Scully, B. E., C. N. Ores, A. S. Prince, and H. C. Neu Treatment of lower respiratory tract infections due to Pseudomonas aeruginosa in patients with cystic fibrosis. Rev. Infect. Dis. 7(Suppl.):S669 S Sekiya, H., T. Mima, Y. Morita, T. Kuroda, T. Mizushima, and T. Tsuchiya Functional cloning and characterization of a multidrug efflux pump, mexhi-opmd, from a Pseudomonas aeruginosa mutant. Antimicrob. Agents Chemother. 47: Sevillano, E., L. Gallego, and J. M. Garcia-Lobo. 26 June 2008, posting date. First detection of the OXA-40 carbapenemase in P. aeruginosa isolates, located on a plasmid also found in A. baumannii. Pathol. Biol. (Paris) [Epub ahead of print.] doi: /j.patbio Shiba, T., K. Ishiguro, N. Takemoto, H. Koibuchi, and K. Sugimoto Purification and characterization of the Pseudomonas aeruginosa NfxB protein, the negative regulator of the nfxb gene. J. Bacteriol. 177: Sobel, M. L., D. Hocquet, L. Cao, P. Plesiat, and K. Poole Mutations in PA3574 (nald) lead to increased MexAB-OprM expression and multidrug resistance in laboratory and clinical isolates of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 49: Sobel, M. L., G. A. McKay, and K. Poole Contribution of the MexXY multidrug transporter to aminoglycoside resistance in Pseudomonas aeruginosa clinical isolates. Antimicrob. Agents Chemother. 48: Sobel, M. L., S. Neshat, and K. Poole Mutations in PA2491 (mexs) promote MexT-dependent mexef-oprn expression and multidrug resistance in a clinical strain of Pseudomonas aeruginosa. J. Bacteriol. 187: Spencer, R. C Predominant pathogens found in the European prevalence of infection in intensive care study. Eur. J. Clin. Microbiol. Infect. Dis. 15: Srikumar, R., T. Kon, N. Gotoh, and K. Poole Expression of Pseudomonas aeruginosa multidrug efflux pumps MexA-MexB-OprM and MexC- MexD-OprJ in a multidrug-sensitive E. coli strain. Antimicrob. Agents Chemother. 42: Srikumar, R., X. Z. Li, and K. Poole Inner membrane efflux components are responsible for beta-lactam specificity of multidrug efflux pumps in Pseudomonas aeruginosa. J. Bacteriol. 179: Srikumar, R., C. J. Paul, and K. Poole Influence of mutations in the mexr repressor gene on expression of the MexA-MexB-OprM efflux system of Pseudomonas aeruginosa. J. Bacteriol. 182: Stapleton, P., K. Shannon, and I. Phillips DNA sequence differences of ampd mutants of Citrobacter freundii. Antimicrob. Agents Chemother. 39: Stobberingh, E. E Induction of chromosomal beta-lactamase by different concentrations of clavulanic acid in combination with ticarcillin. Antimicrob. Agents Chemother. 21: Stover, C. K., X. Q. Pham, A. L. Erwin, S. D. Mizoguchi, P. Warrener, M. J. Hickey, F. S. Brinkman, W. O. Hufnagle, D. J. Kowalik, M. Lagrou, R. L. Garber, L. Goltry, E. Tolentino, S. Westbrock-Wadman, and Y. Yuan Complete genome sequence of Pseudomonas aeruginosa PAO1, an opportunistic pathogen. Nature 406: Takenouchi, T., E. Sakagawa, and M. Sugawara Detection of gyra mutations among 335 Pseudomonas aeruginosa strains isolated in Japan and their susceptibilities to fluoroquinolones. Antimicrob. Agents Chemother. 43: Takesue, Y., T. Yokoyama, S. Akagi, H. Ohge, Y. Imamura, Y. Murakami, and T. Sueda Changes in the intestinal flora after the administration of prophylactic antibiotics to patients undergoing a gastrectomy. Surg. Today 32: Tam, V. M., K. T. Chang, A. N. Schilling, M. T. LaRocco, L. O. Gentry, and K. W. Garey Impact of AmpC overexpression on outcomes of pa-

28 VOL. 22, 2009 ANTIBACTERIAL-RESISTANT PSEUDOMONAS AERUGINOSA 609 tients with Pseudomonas aeruginosa bacteremia. Diagn. Microbiol. Infect. Dis. 63: Thuong, M., K. Arvaniti, R. Ruimy, P. de la Salmoniere, A. Scanvic-Hameg, J. C. Lucet, and B. Regnier Epidemiology of Pseudomonas aeruginosa and risk factors for carriage acquisition in an intensive care unit. J. Hosp. Infect. 53: Toleman, M. A., A. M. Simm, T. A. Murphy, A. C. Gales, D. J. Biedenbach, M. E. Jones, and T. R. Walsh Molecular characterization of SPM-1, a novel metallo-beta-lactamase isolated in Latin America: report from the SENTRY antimicrobial surveillance program. J. Antimicrob. Chemother. 50: Trias, J., and H. Nikaido Outer membrane protein D2 catalyzes facilitated diffusion of carbapenems and penems through the outer membrane of Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 34: Trias, J., and H. Nikaido Protein D2 channel of the Pseudomonas aeruginosa outer membrane has a binding site for basic amino acids and peptides. J. Biol. Chem. 265: Tsuji, M., Y. Ishii, A. Ohno, S. Miyazaki, and K. Yamaguchi In vitro and in vivo antibacterial activities of S-4661, a new carbapenem. Antimicrob. Agents Chemother. 42: Valles, J., D. Mariscal, P. Cortes, P. Coll, A. Villagra, E. Diaz, A. Artigas, and J. Rello Patterns of colonization by Pseudomonas aeruginosa in intubated patients: a 3-year prospective study of 1607 isolates using pulsedfield gel electrophoresis with implications for prevention of ventilatorassociated pneumonia. Intensive Care Med. 30: Vallet, I., S. P. Diggle, R. E. Stacey, M. Camara, I. Ventre, S. Lory, A. Lazdunski, P. Williams, and A. Filloux Biofilm formation in Pseudomonas aeruginosa: fimbrial cup gene clusters are controlled by the transcriptional regulator MvaT. J. Bacteriol. 186: Van Bambeke, R., E. Balzi, and P. M. Tulkens Antibiotic efflux pumps. Biochem. Pharmacol. 60: Van Delden, C., and B. H. Iglewski Cell-to-cell signaling and Pseudomonas aeruginosa infections. Emerg. Infect. Dis. 4: Villavicencio, R. T The history of blue pus. J. Am. Coll. Surg. 187: Villegas, M. V., K. Lolans, A. Correa, J. N. Kattan, J. A. Lopez, J. P. Quinn, and the Colombian Nosocomial Resistance Study Group First identification of Pseudomonas aeruginosa isolates producing a KPC-type carbapenem-hydrolyzing beta-lactamase. Antimicrob. Agents Chemother. 51: Vogne, C., J. R. Aires, C. Bailly, D. Hocquet, and P. Plesiat Role of the multidrug efflux system MexXY in the emergence of moderate resistance to aminoglycosides among Pseudomonas aeruginosa isolates from patients with cystic fibrosis. Antimicrob. Agents Chemother. 48: Wachino, J., K. Yamane, K. Kimura, N. Shibata, S. Suzuki, Y. Ike, and Y. Arakawa Mode of transposition and expression of 16S rrna methyltransferase gene rmtc accompanied by ISEcp1. Antimicrob. Agents Chemother. 50: Wachino, J., K. Yamane, K. Shibayama, H. Kurokawa, N. Shibata, S. Suzuki, Y. Doi, N. Kimura, Y. Ike, and Y. Arakawa Novel plasmidmediated 16S rrna methylase, RmtC, found in a Proteus mirabilis isolate demonstrating extraordinary high-level resistance against various aminoglycosides. Antimicrob. Agents Chemother. 50: Watanabe, M., S. Iyobe, M. Inoue, and S. Mitsuhashi Transferable imipenem resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 35: Weber, D. A., and C. C. Sanders Diverse potential of beta-lactamase inhibitors to induce class I enzymes. Antimicrob. Agents Chemother. 34: Weldhagen, G. F., L. Poirel, and P. Nordmann Ambler class A extendedspectrum beta-lactamases in Pseudomonas aeruginosa: novel developments and clinical impact. Antimicrob. Agents Chemother. 47: Westbrock-Wadman, S., D. R. Sherman, M. J. Hickey, S. N. Coulter, Y. Q. Zhu, P. Warrener, L. Y. Nguyen, R. M. Shawar, K. R. Folger, and C. K. Stover Characterization of a Pseudomonas aeruginosa efflux pump contributing to aminoglycoside impermeability. Antimicrob. Agents Chemother. 43: Westfall, L. W., N. L. Carty, N. Layland, P. Kuan, J. A. Colmer-Hamood, and A. N. Hamood mvat mutation modifies the expression of the Pseudomonas aeruginosa multidrug efflux operon mexef-oprn. FEMS Microbiol. Lett. 255: Wiegand, I., A. K. Marr, E. B. M. Breidenstein, K. N. Schurek, P. Taylor, and R. E. W. Hancock Mutator genes giving rise to decreased antibiotic susceptibility in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 52: Wilke, M. S., M. Heller, A. L. Creagh, C. A. Haynes, L. P. McIntosh, K. Poole, and N. C. Strynadka The crystal structure of MexR from Pseudomonas aeruginosa in complex with its antirepressor ArmR. Proc. Natl. Acad. Sci. USA 105: Wolter, D. J., D. Acquazzino, R. V. Goering, P. Sammut, N. Khalaf, and N. D. Hanson Emergence of carbapenem resistance in Pseudomonas aeruginosa isolates from a patient with cystic fibrosis in the absence of carbapenem therapy. Clin. Infect. Dis. 46: Wolter, D. J., N. D. Hanson, and P. D. Lister AmpC and OprD are not involved in the mechanism of imipenem hypersusceptibility among Pseudomonas aeruginosa isolates overexpressing the mexcd-oprj efflux pump. Antimicrob. Agents Chemother. 49: Wolter, D. J., N. D. Hanson, and P. D. Lister Discordance between imipenem resistance and oprd expression in a Pseudomonas aeruginosa overexpressing mexef-oprn, abstr. C Abstr. 48th Intersci. Conf. Antimicrob. Agents Chemother., Washington, DC Wolter, D. J., N. D. Hanson, and P. D. Lister Efflux-mediated resistance among Pseudomonas aeruginosa: complex regulation of a complex resistance mechanism, abstr. A-132. Abstr. 102nd Gen. Meet. Am. Soc. Microbiol., Salt Lake City, UT Wolter, D. J., N. D. Hanson, and P. D. Lister Insertional inactivation of oprd in clinical isolates of Pseudomonas aeruginosa leading to carbapenem resistance. FEMS Microbiol. Lett. 236: Wolter, D. J., N. D. Hanson, and P. D. Lister MexT-associated downregulation of oprd in Pseudomonas aeruginosa involves inhibition of transcription initiation, abstr. C Abstr. 48th Intersci. Conf. Antimicrob. Agents Chemother., Washington, DC Wolter, D. J., N. D. Hanson, and P. D. Lister Novel mechanism of mexef-oprn efflux pump overexpression in Pseudomonas aeruginosa without coregulation of oprd expression, abstr. C Abstr. 48th Intersci. Conf. Antimicrob. Agents Chemother., Washington, DC Wolter, D. J., N. D. Hanson, and P. D. Lister Phenotypic reversion of imipenem hypersusceptibility among mexcd-oprj-overexpressing P. aeruginosa: a complex system becomes even more complex, abstr. C Abstr. 45th Intersci. Conf. Antimicrob. Agents Chemother., Washington, DC Wolter, D. J., N. Khalaf, I. E. Robledo, G. J. Vazquez, M. I. Sante, E. E. Aquino, R. V. Goering, and N. D. Hanson Surveillance of carbapenem-resistant Pseudomonas aeruginosa isolates from Puerto Rican medical center hospitals: dissemination of KPC and IMP-18 beta-lactamases. Antimicrob. Agents Chemother. 53: Wolter, D. J., A. J. Schmidtke, N. D. Hanson, and P. D. Lister Increased expression of ampc in Pseudomonas aeruginosa mutants selected with ciprofloxacin. Antimicrob. Agents Chemother. 51: Wolter, D. J., E. Smith-Moland, R. V. Goering, N. D. Hanson, and P. D. Lister Multidrug resistance associated with mexxy expression in clinical isolates of Pseudomonas aeruginosa from a Texas hospital. Diagn. Microbiol. Infect. Dis. 50: Woodford, N., J. Zhang, M. E. Kaufmann, S. Yarde, M. D. M. Tomas, C. Faris, M. S. Vardhan, S. Dawson, S. L. Cotterill, and D. M. Livermore Detection of Pseudomonas aeruginosa isolates producing VEB-type extended-spectrum beta-lactamases in the United Kingdom. J. Antimicrob. Chemother. 62: Yamane, K., Y. Doi, K. Yokoyama, T. Yagi, H. Kurokawa, N. Shibata, K. Shibayama, H. Kato, and Y. Arakawa Genetic environments of the rmta gene in Pseudomonas aeruginosa clinical isolates. Antimicrob. Agents Chemother. 48: Yamane, K., J. Wachino, Y. Doi, H. Kurokawa, and Y. Arakawa Global spread of multiple aminoglycoside resistance genes. Emerg. Infect. Dis. 11: Yokoyama, K., Y. Doi, K. Yamane, H. Kurokawa, N. Shibata, K. Shibayama, T. Yagi, H. Kato, and Y. Arakawa Acquisition of 16S rrna methylase gene in Pseudomonas aeruginosa. Lancet 362: Yoneyama, H., and T. Nakae Mechanism of efficient elimination of protein D2 in outer membrane of imipenem-resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 37: Yoshimura, F., and H. Nikaido Diffusion of beta-lactam antibiotics through the porin channels of Escherichia coli K-12. Antimicrob. Agents Chemother. 27: Zhao, Q., X. Z. Li, R. Srikumar, and K. Poole Contribution of outer membrane efflux protein OprM to antibiotic resistance in Pseudomonas aeruginosa independent of MexAB. Antimicrob. Agents Chemother. 42: Continued next page

29 610 LISTER ET AL. CLIN. MICROBIOL. REV. Philip Lister is a Professor of Medical Microbiology and Immunology and Associate Director of the Center for Research in Anti- Infectives and Biotechnology at Creighton University. Dr. Lister received his B.S. in microbiology from Kansas State University (1986) and his Ph.D. in medical microbiology from Creighton University (1992). After a postdoctoral fellowship, Dr. Lister joined the faculty at Creighton University in 1994 and was promoted to Professor of Medical Microbiology and Immunology in 2007, with a secondary appointment for the School of Pharmacy Science. Dr. Lister has been involved in antibacterial pharmacodynamics and resistance research for 15 years and currently serves as an Editor for the Journal of Antimicrobial Chemotherapy and on the editorial boards of other journals. Dr. Lister s research has focused on clinically important gram-positive and gram-negative pathogens, but it has been the therapeutic challenge of P. aeruginosa that has been a primary focus. Nancy Hanson is a Professor of Medical Microbiology and Immunology and Director of Molecular Biology for the Center for Research in Anti-Infectives and Biotechnology at Creighton University. Dr. Hanson received her Ph.D. in medical microbiology from the University of Nebraska Medical Center in 1991 and joined the faculty of Creighton University in Her area of expertise involves the study of molecular mechanisms of antibiotic resistance in gram-negative organisms, such as E. coli, K. pneumoniae, Salmonella spp., and Pseudomonas aeruginosa. Her research explores the following two aspects of antibiotic resistance mechanisms: (i) the regulation of the genes involved in resistance and (ii) the development of PCR-based diagnostic tests that can be used by clinical laboratories to detect resistance genes in clinical isolates. In 2007, Dr. Hanson was awarded researcher of the year by the Nebraska Chapter of the Cystic Fibrosis Foundation for her work on P. aeruginosa. Daniel Wolter received his B.S. in biology from Gonzaga University in 1997 and his Ph.D. in medical microbiology from Creighton University in Dr. Wolter continued his training as a postdoctoral fellow in medical microbiology at Creighton University until 2008 and has since joined the Department of Pediatrics at the University of Washington as a Senior Fellow. A majority of his research has focused on the antimicrobial resistance mechanisms of Pseudomonas aeruginosa, with particular emphasis on efflux-mediated drug resistance. His current research at the University of Washington explores the genotypic and phenotypic adaptations of P. aeruginosa isolated from patients with cystic fibrosis (CF) and the polymicrobial interactions of this organism with other CF lung microbiota, such as Staphylococcus aureus.

Translocation Studies Mid-Term Review (MTR) Meeting Marseille, France

Translocation Studies Mid-Term Review (MTR) Meeting Marseille, France Marie Curie Actions Research Training Networks (RTN) Translocation Studies Mid-Term Review (MTR) Meeting Marseille, France F. Vidal-Aroca, M.G.P. Page and J. Dreier Background Deteriorating situation regarding

More information

Discussion points CLSI M100 S19 Update. #1 format of tables has changed. #2 non susceptible category

Discussion points CLSI M100 S19 Update. #1 format of tables has changed. #2 non susceptible category Discussion points 2009 CLSI M100 S19 Update Nebraska Public Health Laboratory Changes most important to routine antimicrobial susceptibility testing. Documents available Janet Hindler discussion slide

More information

NONFERMENTING GRAM NEGATIVE RODS. April Abbott Deaconess Health System Evansville, IN

NONFERMENTING GRAM NEGATIVE RODS. April Abbott Deaconess Health System Evansville, IN NONFERMENTING GRAM NEGATIVE RODS April Abbott Deaconess Health System Evansville, IN OBJECTIVES Discuss basic limitations to assessing carbapenem resistance in nonfermenting GNRs Discuss antimicrobial

More information

Affinity of Doripenem and Comparators to Penicillin-Binding Proteins in Escherichia coli and ACCEPTED

Affinity of Doripenem and Comparators to Penicillin-Binding Proteins in Escherichia coli and ACCEPTED AAC Accepts, published online ahead of print on February 00 Antimicrob. Agents Chemother. doi:./aac.01-0 Copyright 00, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights

More information

Development of C sporins. Beta-lactam antibiotics - Cephalosporins. Second generation C sporins. Targets - PBP s

Development of C sporins. Beta-lactam antibiotics - Cephalosporins. Second generation C sporins. Targets - PBP s Beta-lactam antibiotics - Cephalosporins Development of C sporins Targets - PBP s Activity - Cidal - growing organisms (like the penicillins) Principles of action - Affinity for PBP s Permeability properties

More information

Clinical Management of Infections Caused by Enterobacteriaceae that Express Extended- Spectrum β-lactamase and AmpC Enzymes

Clinical Management of Infections Caused by Enterobacteriaceae that Express Extended- Spectrum β-lactamase and AmpC Enzymes 56 Clinical Management of Infections Caused by Enterobacteriaceae that Express Extended- Spectrum β-lactamase and AmpC Enzymes Patrick N. A. Harris, BSc, MBBS, MRCP, DTM&H, FRACP, FRCPA 1 1 Infection and

More information

ALERT. Clinical microbiology considerations related to the emergence of. New Delhi metallo beta lactamases (NDM 1) and Klebsiella

ALERT. Clinical microbiology considerations related to the emergence of. New Delhi metallo beta lactamases (NDM 1) and Klebsiella ALERT Clinical microbiology considerations related to the emergence of New Delhi metallo beta lactamases (NDM 1) and Klebsiella pneumoniae carbapenemases (KPC) amongst hospitalized patients in South Africa

More information

β-lactamase inhibitors

β-lactamase inhibitors β-lactamase inhibitors Properties, microbiology & enzymology DAVID M LIVERMORE Professor of Medical Microbiology, UEA Lead on Antibiotic Resistance, Public Health England β-lactamase classes A B C D Serine

More information

Rapid identification and resistance assessment: The future is mass spectrometry

Rapid identification and resistance assessment: The future is mass spectrometry Rapid identification and resistance assessment: The future is mass spectrometry Dr Sanmarié Schlebusch Director of Microbiology Mater Pathology Brisbane Outline Introduction Plug and play Pre-prep and

More information

Public Health Surveillance for Multi Drug Resistant Organisms in Orange County

Public Health Surveillance for Multi Drug Resistant Organisms in Orange County Public Health Surveillance for Multi Drug Resistant Organisms in Orange County Matt Zahn, MD Medical Director Epidemiology and Assessment Orange County Public Health Antimicrobial Mechanisms of Action

More information

Pseudomonas aeruginosa: from environment to humans

Pseudomonas aeruginosa: from environment to humans Pseudomonas aeruginosa: from environment to humans Finovi Thursday 9 th december 2010, Lyon Jean Freney UMR 5557 CNRS Ecologie Microbienne University of Lyon 1 Pseudomonas aeruginosa Carle Gessard (1850-1925)

More information

MHSAL Guidelines for the Prevention and Control of Antimicrobial Resistant Organisms (AROs) - Response to Questions

MHSAL Guidelines for the Prevention and Control of Antimicrobial Resistant Organisms (AROs) - Response to Questions MHSAL Guidelines for the Prevention and Control of Antimicrobial Resistant Organisms (AROs) - Response to Questions Dr. Andrew Walkty Medical Microbiologist, Diagnostic Services Manitoba (DSM) June. 17,

More information

Laboratory CLSI M100-S18 update. Paul D. Fey, Ph.D. Associate Professor/Associate Director Josh Rowland, M.T. (ASCP) State Training Coordinator

Laboratory CLSI M100-S18 update. Paul D. Fey, Ph.D. Associate Professor/Associate Director Josh Rowland, M.T. (ASCP) State Training Coordinator Nebraska Public Health Laboratory 2008 CLSI M100-S18 update Paul D. Fey, Ph.D. Associate Professor/Associate Director Josh Rowland, M.T. (ASCP) State Training Coordinator Agenda Discuss 2008 M100- S18

More information

Macrolides, Clindamycin & Ketolides Polymyxins

Macrolides, Clindamycin & Ketolides Polymyxins Macrolides, Clindamycin & Ketolides Polymyxins Kwan Soo Ko Macrolides - Erythromycin - Azithromycin - Clarithromycin Lincosamides - Lincomycin - Clindamycin Unrelated chemically But, many similar biological

More information

Overcoming the PosESBLities of Enterobacteriaceae Resistance

Overcoming the PosESBLities of Enterobacteriaceae Resistance Overcoming the PosESBLities of Enterobacteriaceae Resistance Review of current treatment options Jamie Reed, PharmD Pharmacy Grand Rounds August 28, 2018 Rochester, MN 2018 MFMER slide-1 Disclosure No

More information

Infection Control Strategies to Avoid Carbapenam Resistance in Hospitals. Victor Lim International Medical University Malaysia

Infection Control Strategies to Avoid Carbapenam Resistance in Hospitals. Victor Lim International Medical University Malaysia Infection Control Strategies to Avoid Carbapenam Resistance in Hospitals Victor Lim International Medical University Malaysia Outline of Lecture 1. Carbapenam resistance 2. Epidemiology of carbapenam resistance

More information

Determining the Optimal Carbapenem MIC that Distinguishes Carbapenemase-Producing

Determining the Optimal Carbapenem MIC that Distinguishes Carbapenemase-Producing AAC Accepted Manuscript Posted Online 8 August 2016 Antimicrob. Agents Chemother. doi:10.1128/aac.00838-16 Copyright 2016, American Society for Microbiology. All Rights Reserved. 1 1 2 Determining the

More information

Emerging broad-spectrum resistance in Pseudomonas aeruginosa and Acinetobacter baumannii: Mechanisms and epidemiology

Emerging broad-spectrum resistance in Pseudomonas aeruginosa and Acinetobacter baumannii: Mechanisms and epidemiology Published in which should be cited to refer to this work. Emerging broad-spectrum resistance in and : Mechanisms and epidemiology Anaïs Potron a, Laurent Poirel b,, Patrice Nordmann b,c a Laboratoire de

More information

Annual Surveillance Summary: Pseudomonas aeruginosa Infections in the Military Health System (MHS), 2016

Annual Surveillance Summary: Pseudomonas aeruginosa Infections in the Military Health System (MHS), 2016 Annual Surveillance Summary: Pseudomonas aeruginosa Infections in the Military Health System (MHS), 2016 Sarah Gierhart and Uzo Chukwuma Approved for public release. Distribution is unlimited. The views

More information

Update on CLSI and EUCAST

Update on CLSI and EUCAST Update on CLSI and EUCAST 1 Completed work» Cephalosporin breakpoints for Enterobacteriaceae ESBL screens MIC versus resistance mechanism» Carbapenem breakpoints for Enterobacteriaceae Modified Hodge Test»

More information

Frequency of Occurrence and Antimicrobial Susceptibility of Bacteria from ICU Patients with Pneumonia

Frequency of Occurrence and Antimicrobial Susceptibility of Bacteria from ICU Patients with Pneumonia Frequency of Occurrence and Antimicrobial Susceptibility of Bacteria from ICU Patients with Pneumonia Helio S. Sader, M.D.* Mariana Castanheira, Ph.D. Rodrigo E. Mendes, Ph.D. Robert K. Flamm, Ph.D. JMI

More information

Prevalence of Extended Spectrum -Lactamases In E.coli and Klebsiella spp. in a Tertiary Care Hospital

Prevalence of Extended Spectrum -Lactamases In E.coli and Klebsiella spp. in a Tertiary Care Hospital ISSN: 2319-7706 Volume 3 Number 10 (2014) pp. 474-478 http://www.ijcmas.com Original Research Article Prevalence of Extended Spectrum -Lactamases In E.coli and Klebsiella spp. in a Tertiary Care Hospital

More information

Resistance to Polymyxins in France

Resistance to Polymyxins in France Resistance to Polymyxins in France Paris Prof. Patrice Nordmann NDM producers in Enterobacteriaceae The polymyxins; colistin and polymyxin B Colistin - Synthesis by Bacillus polymyxa spp colistinus -

More information

Carbapenemases: the Versatile -Lactamases

Carbapenemases: the Versatile -Lactamases CLINICAL MICROBIOLOGY REVIEWS, July 2007, p. 440 458 Vol. 20, No. 3 0893-8512/07/$08.00 0 doi:10.1128/cmr.00001-07 Copyright 2007, American Society for Microbiology. All Rights Reserved. Carbapenemases:

More information

Guidance on screening and confirmation of carbapenem resistant Enterobacteriacae (CRE) December 12, 2011

Guidance on screening and confirmation of carbapenem resistant Enterobacteriacae (CRE) December 12, 2011 Guidance on screening and confirmation of carbapenem resistant Enterobacteriacae (CRE) December 12, 2011 Objectives: To discuss the guidelines for detection of CRE in the laboratory setting. To review

More information

Navigating Through Current and Emerging Issues in Outbreaks

Navigating Through Current and Emerging Issues in Outbreaks Navigating Through Current and Emerging Issues in Outbreaks 7th GCC Conference on Infection Prevention and Control December 1-3, 2013 Kuwait City, Kuwait William R. Jarvis, M.D. Jason and Jarvis Associates,

More information

Regional Emergence of VIM producing carbapenem resistant Pseudomonas aeruginosa (VIM CRPA)

Regional Emergence of VIM producing carbapenem resistant Pseudomonas aeruginosa (VIM CRPA) National Center for Emerging and Zoonotic Infectious Diseases Regional Emergence of VIM producing carbapenem resistant Pseudomonas aeruginosa (VIM CRPA) Chris Prestel, MD Epidemic Intelligence Service

More information

Shirin Abadi, B.Sc.(Pharm.), ACPR, Pharm.D. Clinical Pharmacy Specialist & Pharmacy Education Coordinator, BC Cancer Agency Clinical Associate

Shirin Abadi, B.Sc.(Pharm.), ACPR, Pharm.D. Clinical Pharmacy Specialist & Pharmacy Education Coordinator, BC Cancer Agency Clinical Associate Shirin Abadi, B.Sc.(Pharm.), ACPR, Pharm.D. Clinical Pharmacy Specialist & Pharmacy Education Coordinator, BC Cancer Agency Clinical Associate Professor of Pharmacy & Associate Member of Medicine, UBC

More information

Carbapenemases in Enterobacteriaceae: Prof P. Nordmann Bicêtre hospital, South-Paris Med School

Carbapenemases in Enterobacteriaceae: Prof P. Nordmann Bicêtre hospital, South-Paris Med School Carbapenemases in Enterobacteriaceae: 2012 Prof P. Nordmann Bicêtre hospital, South-Paris Med School March 21, 2012 Trends in Molecular Medecine NDM IMP OXA-48 KPC VIM ALERT VI M KPC KPC NDM I MP OXA-

More information

Detection of Carbapenem Resistant Enterobacteriacae from Clinical Isolates

Detection of Carbapenem Resistant Enterobacteriacae from Clinical Isolates International Journal of Current Microbiology and Applied Sciences ISSN: 2319-7706 Volume 5 Number 5 (2016) pp. 864-869 Journal homepage: http://www.ijcmas.com Original Research Article http://dx.doi.org/10.20546/ijcmas.2016.505.089

More information

Adenium Biotech. Management: - Peter Nordkild, MD, CEO, ex Novo Nordisk, Ferring, Egalet - Søren Neve, PhD, project director, ex Lundbeck, Novozymes

Adenium Biotech. Management: - Peter Nordkild, MD, CEO, ex Novo Nordisk, Ferring, Egalet - Søren Neve, PhD, project director, ex Lundbeck, Novozymes Adenium Biotech Management: - Peter Nordkild, MD, CEO, ex Novo Nordisk, Ferring, Egalet - Søren Neve, PhD, project director, ex Lundbeck, Novozymes Board of Directors: - Stephan Christgau, PhD, chairman,

More information

Carbapenems and Enterobacteriaceae

Carbapenems and Enterobacteriaceae Title Carbapenems and Enterobacteriaceae Presenter s details NHLS Dr Khine Swe Swe/Han FC Path ( Micro), SA MMed( micro), SA DTMH(Wits univ),sa PDIC(Stellen univ)sa MB,BS(Yangon),Myanmar Pathologist,Consultant/Lecturer,

More information

Detecting CRE. what does one need to do?

Detecting CRE. what does one need to do? 5 th ICAN Conference, Harare 4 th November 2014 Room 2: 10:30-12:00 Detecting CRE (Carbapenem-resistant Enterobacteriaceae) what does one need to do? Dr Nizam Damani Associate Medical Director Infection

More information

Mechanisms of -lactam Resistance Among Pseudomonas aeruginosa

Mechanisms of -lactam Resistance Among Pseudomonas aeruginosa Send rders of Reprints at reprints@benthamscience.org Current Pharmaceutical Design, 2013, 19, 209-222 209 Mechanisms of -lactam Resistance Among Pseudomonas aeruginosa Daniel J. Wolter 1 and Philip D.

More information

ST11 KPC-2 Klebsiella pneumoniae detected in Taiwan

ST11 KPC-2 Klebsiella pneumoniae detected in Taiwan AAC Accepts, published online ahead of print on 30 January 2012 Antimicrob. Agents Chemother. doi:10.1128/aac.05576-11 Copyright 2012, American Society for Microbiology. All Rights Reserved. 1 2 3 4 5

More information

AAC Accepts, published online ahead of print on 13 October 2008 Antimicrob. Agents Chemother. doi: /aac

AAC Accepts, published online ahead of print on 13 October 2008 Antimicrob. Agents Chemother. doi: /aac AAC Accepts, published online ahead of print on 13 October 2008 Antimicrob. Agents Chemother. doi:10.1128/aac.00931-08 Copyright 2008, American Society for Microbiology and/or the Listed Authors/Institutions.

More information

Rapid identification of emerging resistance in Gram negatives. Prof. Patrice Nordmann

Rapid identification of emerging resistance in Gram negatives. Prof. Patrice Nordmann Rapid identification of emerging resistance in Gram negatives Prof. Patrice Nordmann Emerging Resistance threats, CDC USA-2013 Enterobacteriaceae producing extendedspectrum β-lactamases (ESBL) Multi-resistant

More information

Annual Surveillance Summary: Pseudomonas aeruginosa Infections in the Military Health System (MHS), 2017

Annual Surveillance Summary: Pseudomonas aeruginosa Infections in the Military Health System (MHS), 2017 i Annual Surveillance Summary: Pseudomonas aeruginosa Infections in the Military Health System (MHS), 2017 Jessica R. Spencer and Uzo Chukwuma Approved for public release. Distribution is unlimited. The

More information

Expert rules in antimicrobial susceptibility testing: State of the art

Expert rules in antimicrobial susceptibility testing: State of the art Expert rules in antimicrobial susceptibility testing: State of the art ESCMID Postgraduate Education Course Antimicrobial Susceptibility Testing and Surveillance: from Laboratory to Clinic Hospital Universitario

More information

Expert rules. for Gram-negatives

Expert rules. for Gram-negatives Academic Perspective in Expert rules Emerging Issues of Resistance in Gram-ve Bacteria for Gram-negatives Trevor Winstanley Sheffield Teaching Hospitals Presented on behalf of David Livermore University

More information

Other β-lactam. A. Carbapenems:

Other β-lactam. A. Carbapenems: A. Carbapenems: Other β-lactam Carbapenems are synthetic β-lactam antibiotics Differ in structure from the penicillins in that the sulfur atom of the thiazolidine ring. Imipenem, meropenem, doripenem,

More information

Enterobacteriaceae with acquired carbapenemases, 2016

Enterobacteriaceae with acquired carbapenemases, 2016 Enterobacteriaceae with acquired carbapenemases, 2016 Background The acquired or transferable (as opposed to chromosomally encoded) carbapenemases found in Enterobacteriaceae belong to three of the four

More information

In Vitro Activity of Ceftazidime-Avibactam Against Isolates. in a Phase 3 Open-label Clinical Trial for Complicated

In Vitro Activity of Ceftazidime-Avibactam Against Isolates. in a Phase 3 Open-label Clinical Trial for Complicated AAC Accepted Manuscript Posted Online 21 November 2016 Antimicrob. Agents Chemother. doi:10.1128/aac.01820-16 Copyright 2016, American Society for Microbiology. All Rights Reserved. 1 2 3 4 5 6 7 8 9 10

More information

(multidrug-resistant Pseudomonas aeruginosa; MDRP)

(multidrug-resistant Pseudomonas aeruginosa; MDRP) 220 2009 (multidrug-resistant Pseudomonas aeruginosa; MDRP) 21 4 1 21 10 4 amikacin (AMK), imipenem/cilastatin (IPM), ciprofloxacin (CPFX) multidrug-resistant Pseudomonas aeruginosa (MDRP) CHROMagar TM

More information

Disclosure. Objectives. Evolution of β Lactamases. Extended Spectrum β Lactamases: The New Normal. Prevalence of ESBL Mystic Program

Disclosure. Objectives. Evolution of β Lactamases. Extended Spectrum β Lactamases: The New Normal. Prevalence of ESBL Mystic Program 47 th Annual Meeting August 2-4, 2013 Orlando, FL Extended Spectrum β Lactamases: The New Normal Disclosure I do have a vested interest in or affiliation with the following companies or organizations Triax

More information

Carbapenem-resistant Escherichia coli and Klebsiella pneumoniae in Taiwan

Carbapenem-resistant Escherichia coli and Klebsiella pneumoniae in Taiwan Carbapenem-resistant Escherichia coli and Klebsiella pneumoniae in Taiwan An Infection Control Emergency Speaker: L Kristopher Siu Principal Investigator Division of Infectious Diseases National Institute

More information

Received 31 January 2011/Returned for modification 2 March 2011/Accepted 15 March 2011

Received 31 January 2011/Returned for modification 2 March 2011/Accepted 15 March 2011 JOURNAL OF CLINICAL MICROBIOLOGY, May 2011, p. 1965 1969 Vol. 49, No. 5 0095-1137/11/$12.00 doi:10.1128/jcm.00203-11 Copyright 2011, American Society for Microbiology. All Rights Reserved. Comparative

More information

Development of Resistance mechanisms in Pseudomonas aeruginosa

Development of Resistance mechanisms in Pseudomonas aeruginosa Development of Resistance mechanisms in Pseudomonas aeruginosa Luis Martínez Martínez ervice of Microbiology University Hospital Marqués de Valdecilla antander, pain 5th ECMID chool, antander June 2006

More information

The b-lactamase threat in Enterobacteriaceae, Pseudomonas and Acinetobacter

The b-lactamase threat in Enterobacteriaceae, Pseudomonas and Acinetobacter Review TRENDS in Microbiology Vol.14 No.9 The b-lactamase threat in Enterobacteriaceae, Pseudomonas and Acinetobacter David M. Livermore and Neil Woodford Antibiotic Resistance Monitoring and Reference

More information

Phenotypic detection of ESBLs and carbapenemases

Phenotypic detection of ESBLs and carbapenemases Phenotypic detection of ESBLs and carbapenemases Standardized susceptibility testing residential workshop 2016 Katie Hopkins PhD Clinical Scientist Antimicrobial Resistance and Healthcare Associated Infections

More information

Global Epidemiology of Carbapenem- Resistant Enterobacteriaceae (CRE)

Global Epidemiology of Carbapenem- Resistant Enterobacteriaceae (CRE) Global Epidemiology of Carbapenem- Resistant Enterobacteriaceae (CRE) Mitchell J. Schwaber, MD MSc Director, National Center for Infection Control Ministry of Health State of Israel November 27, 2012 1

More information

National Center for Emerging and Zoonotic Infectious Diseases The Biggest Antibiotic Resistance Threats

National Center for Emerging and Zoonotic Infectious Diseases The Biggest Antibiotic Resistance Threats National Center for Emerging and Zoonotic Infectious Diseases The Biggest Antibiotic Resistance Threats Jean B. Patel, PhD, D(ABMM) Science Lead, Antibiotic Resistance and Coordination Unit Centers for

More information

Antibiotic Treatment of GNR MDR Infections. Stan Deresinski

Antibiotic Treatment of GNR MDR Infections. Stan Deresinski Antibiotic Treatment of GNR MDR Infections Stan Deresinski Kucers: The Use of Antibiotics 1st Edition 1972 392 pages Kucers: The Use of Antibiotics 7 th Edition 2017 5338 pages Carbapenem Susceptibility

More information

AMR prediction based on WGS data

AMR prediction based on WGS data AMR prediction based on WGS data Valeria Bortolaia, DVM, PhD Research Group for Genomic Epidemiology National Food Institute Technical University of Denmark This lecture How to use WGS for AMR surveillance?

More information

Screening and detection of carbapenemases

Screening and detection of carbapenemases Screening and detection of carbapenemases For many isolates with carbapenemases the MICs of carbapenems are around the susceptible breakpoint making resistance difficult to detect - particularly with automated

More information

Treatment of febrile neutropenia in patients with neoplasia

Treatment of febrile neutropenia in patients with neoplasia Treatment of febrile neutropenia in patients with neoplasia George Samonis MD, PhD Medical Oncologist Infectious Diseases Specialist Professor of Medicine The University of Crete, Heraklion,, Crete, Greece

More information

A Snapshot of Colistin Use in South-East Europe and Particularly in Greece

A Snapshot of Colistin Use in South-East Europe and Particularly in Greece A Snapshot of Colistin Use in South-East Europe and Particularly in Greece Helen Giamarellou 02.05.2013 When Greek Physicians Prescribe Colistin? It is mainly prescribed in the ICU for VAP, bacteremia

More information

Revised AAC Version 2» New-Data Letter to the Editor ACCEPTED. Plasmid-Mediated Carbapenem-Hydrolyzing β-lactamase KPC-2 in

Revised AAC Version 2» New-Data Letter to the Editor ACCEPTED. Plasmid-Mediated Carbapenem-Hydrolyzing β-lactamase KPC-2 in AAC Accepts, published online ahead of print on 3 December 2007 Antimicrob. Agents Chemother. doi:10.1128/aac.01180-07 Copyright 2007, American Society for Microbiology and/or the Listed Authors/Institutions.

More information

Activity of Ceftolozane/Tazobactam Against a Broad Spectrum of Recent Clinical Anaerobic Isolates

Activity of Ceftolozane/Tazobactam Against a Broad Spectrum of Recent Clinical Anaerobic Isolates AAC Accepts, published online ahead of print on 25 November 2013 Antimicrob. Agents Chemother. doi:10.1128/aac.02253-13 Copyright 2013, American Society for Microbiology. All Rights Reserved. Activity

More information

ANTIPSEUDOMONAL ANTIBIOTICS

ANTIPSEUDOMONAL ANTIBIOTICS Use of this content is subject to the Terms and Conditions ANTIPSEUDOMONAL ANTIBIOTICS Medical Clinics of North America - Volume 85, Issue 1 (January 2001) - Copyright 2001 W. B. Saunders Company DOI:

More information

Use of imipenem. with the support of Wallonie-Bruxelles International. Magali Dodémont Microbiology Hospital Erasme Université Libre de Bruxelles

Use of imipenem. with the support of Wallonie-Bruxelles International. Magali Dodémont Microbiology Hospital Erasme Université Libre de Bruxelles with the support of Wallonie-Bruxelles International Use of imipenem Magali Dodémont Microbiology Hospital Erasme Université Libre de Bruxelles 1 Β-lactams classification 2 Β-lactams: mode of action Inhibition

More information

Detecting carbapenemases in Enterobacteriaceae

Detecting carbapenemases in Enterobacteriaceae Detecting carbapenemases in Enterobacteriaceae David Livermore Health Protection Agency, Colindale, London 12 August 2003 Mechanisms of carbapenem R in Enterobacteria Impermeability + AmpC or ESBL Metallo

More information

Emergence of non-kpc carbapenemases: NDM and more

Emergence of non-kpc carbapenemases: NDM and more Emergence of non-kpc carbapenemases: NDM and more --- David Livermore Health Protection Agency, UK The first acquired carbapenemase to be recognised in gram-negative bacteria was IMP-1, a metallo-type,

More information

Annual Surveillance Summary: Klebsiella species Infections in the Military Health System (MHS), 2017

Annual Surveillance Summary: Klebsiella species Infections in the Military Health System (MHS), 2017 i Annual Surveillance Summary: Klebsiella species Infections in the Military Health System (MHS), 2017 Tej Mishra and Uzo Chukwuma Approved for public release. Distribution is unlimited. The views expressed

More information

Mechanisms of resistance to ceftolozane tazobactam Antonio Oliver Servicio de Microbiología, Hospital Son Espases.

Mechanisms of resistance to ceftolozane tazobactam Antonio Oliver Servicio de Microbiología, Hospital Son Espases. Mechanisms of resistance to ceftolozane tazobactam Antonio Oliver Servicio de Microbiología, Hospital Son Espases. Palma de Mallorca SPAIN ECCMID 2018. Madrid, April 22 nd Disclosures: Awarded research

More information

Emergence of Klebsiella pneumoniae ST258 with KPC-2 in Hong Kong. Title. Ho, PL; Tse, CWS; Lai, EL; Lo, WU; Chow, KH

Emergence of Klebsiella pneumoniae ST258 with KPC-2 in Hong Kong. Title. Ho, PL; Tse, CWS; Lai, EL; Lo, WU; Chow, KH Title Emergence of Klebsiella pneumoniae ST258 with KPC-2 in Hong Kong Author(s) Ho, PL; Tse, CWS; Lai, EL; Lo, WU; Chow, KH Citation International Journal Of Antimicrobial Agents, 2011, v. 37 n. 4, p.

More information

Molecular mechanisms of antibiotic resistance in Neisseria gonorrhoeae

Molecular mechanisms of antibiotic resistance in Neisseria gonorrhoeae medicaldaily.com http://en.vircell.com/diseases ppcorn.com Molecular mechanisms of antibiotic resistance in Neisseria gonorrhoeae Robert Nicholas University of North Carolina at Chapel Hill cdc.gov Timeline

More information

ESCMID Online Lecture Library. by author

ESCMID Online Lecture Library. by author www.eucast.org EXPERT RULES IN ANTIMICROBIAL SUSCEPTIBILITY TESTING Dr. Rafael Cantón Hospital Universitario Ramón y Cajal SERVICIO DE MICROBIOLOGÍA Y PARASITOLOGÍA Departamento de Microbiología II Universidad

More information

Carbapenemase-producing Enterobacteriaceae

Carbapenemase-producing Enterobacteriaceae Carbapenemase-producing Enterobacteriaceae 2012 CNR Associé Résistance aux Antibiotiques Prof. P. Nordmann Carbapenemases in Enterobacteriaceae May, 2012 Penicillins Cephalosporins Carbapenems Extended-spectrum

More information

Aminoglycoside Resistance in Gram-negative Bacilli

Aminoglycoside Resistance in Gram-negative Bacilli Korean J Clin Microbiol Vol. 12, No. 2, June, 2009 Aminoglycoside Resistance in Gram-negative Bacilli Yeon-Joon Park Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea,

More information

Aminoglycosides. Not orally absorbed. Interact with negatively charged lipopolysaccharide on Gram- cell wall. Aminoglycoside properties

Aminoglycosides. Not orally absorbed. Interact with negatively charged lipopolysaccharide on Gram- cell wall. Aminoglycoside properties Aminoglycosides Tobramycin Kanamycin H2N Gentamicin Amikacin NHCOCHCH2CH2NH2 OH Aminoglycosides made of linked sugars. Decorated with many OH and NH2 groups, which render these compounds positively charged

More information

CARBAPENEMASE PRODUCING ENTEROBACTERIACEAE

CARBAPENEMASE PRODUCING ENTEROBACTERIACEAE CARBAPENEMASE PRODUCING ENTEROBACTERIACEAE Veroniek Saegeman Veroniek Saegeman UZLeuven Carbapenemase producing Enterobacteriaceae (CPE) Introduction on antibiotic resistance Classification of ß-lactamases

More information

(Plasmid mediated) Carbapenemases. Timothy R. Walsh, Cardiff University, Wales

(Plasmid mediated) Carbapenemases. Timothy R. Walsh, Cardiff University, Wales (Plasmid mediated) Carbapenemases Timothy R. Walsh, Cardiff University, Wales What is a carbapenemase? How much carbapenem do they need to breakdown before they are called a carbapenemase? ESBL-enzymes

More information

Rapid detection of carbapenemase-producing Pseudomonas spp.

Rapid detection of carbapenemase-producing Pseudomonas spp. JCM Accepts, published online ahead of print on 12 September 2012 J. Clin. Microbiol. doi:10.1128/jcm.01597-12 Copyright 2012, American Society for Microbiology. All Rights Reserved. 1 Rapid detection

More information

Cefotaxime Rationale for the EUCAST clinical breakpoints, version th September 2010

Cefotaxime Rationale for the EUCAST clinical breakpoints, version th September 2010 Cefotaxime Rationale for the EUCAST clinical breakpoints, version 1.0 26 th September 2010 Foreword EUCAST The European Committee on Antimicrobial Susceptibility Testing (EUCAST) is organised by the European

More information

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors

We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists. International authors and editors We are IntechOpen, the world s leading publisher of Open Access books Built by scientists, for scientists 3,900 116,000 120M Open access books available International authors and editors Downloads Our

More information

Academic Perspective in. David Livermore Prof of Medical Microbiology, UEA Lead on Antibiotic resistance PHE

Academic Perspective in. David Livermore Prof of Medical Microbiology, UEA Lead on Antibiotic resistance PHE Academic Perspective in Emerging No, we can t Issues treat of carbapenemase Resistance and ESBL in Gram-ve producers Bacteria based on MIC David Livermore Prof of Medical Microbiology, UEA Lead on Antibiotic

More information

Healthcare-associated infections acquired in intensive care units

Healthcare-associated infections acquired in intensive care units SURVEILLANCE REPORT Annual Epidemiological Report for 2015 Healthcare-associated infections acquired in intensive care units Key facts In 2015, 11 788 (8.3%) of patients staying in an intensive care unit

More information

Below is an overview of the oral and poster presentations featuring cefiderocol and S at IDWeek 2017:

Below is an overview of the oral and poster presentations featuring cefiderocol and S at IDWeek 2017: SHIONOGI TO HIGHLIGHT RESEARCH ON CEFIDEROCOL (S-649266), A SIDEROPHORE CEPHALOSPORIN, AND S-033188, A CAP-DEPENDENT ENDONUCLEASE INHIBITOR FOR TREATMENT OF INFLUENZA, AT IDWEEK 2017 OSAKA, Japan and FLORHAM

More information

High Incidence of Multidrug Resistant Pseudomonas aeruginosa Isolated from Infected Burn Wounds in a Tertiary Hospital

High Incidence of Multidrug Resistant Pseudomonas aeruginosa Isolated from Infected Burn Wounds in a Tertiary Hospital International Journal of Current Microbiology and Applied Sciences ISSN: 2319-7706 Volume 6 Number 2 (2017) pp. 1134-1139 Journal homepage: http://www.ijcmas.com Original Research Article http://dx.doi.org/10.20546/ijcmas.2017.602.128

More information

GUIDE TO INFECTION CONTROL IN THE HOSPITAL. Carbapenem-resistant Enterobacteriaceae

GUIDE TO INFECTION CONTROL IN THE HOSPITAL. Carbapenem-resistant Enterobacteriaceae GUIDE TO INFECTION CONTROL IN THE HOSPITAL CHAPTER 47: Carbapenem-resistant Enterobacteriaceae Authors E-B Kruse, MD H. Wisplinghoff, MD Chapter Editor Michelle Doll, MD, MPH) Topic Outline Key Issue Known

More information

In-House Standardization of Carba NP Test for Carbapenemase Detection in Gram Negative Bacteria

In-House Standardization of Carba NP Test for Carbapenemase Detection in Gram Negative Bacteria International Journal of Current Microbiology and Applied Sciences ISSN: 2319-7706 Volume 7 Number 01 (2018) Journal homepage: http://www.ijcmas.com Original Research Article https://doi.org/10.20546/ijcmas.2018.701.342

More information

Differentiation of Carbapenemase producing Enterobacteriaceae by Triple disc Test

Differentiation of Carbapenemase producing Enterobacteriaceae by Triple disc Test Original article: Differentiation of Carbapenemase producing Enterobacteriaceae by Triple disc Test Manish Bansal 1, Nitya Vyas 2, Babita Sharma 3, R.K.Maheshwari 4 1PG Resident, 2 Professor, 3 Assistant

More information

Report on susceptibility of Salmonella serotypes in Belgium Vicky Jasson

Report on susceptibility of Salmonella serotypes in Belgium Vicky Jasson CODA-CERVA Report on susceptibility of Salmonella serotypes in Belgium 2014. Vicky Jasson Veterinary and Agrochemical Research Centre 1 Introduction Salmonella is one of the most important bacterial zoonotic

More information

Surveillance of antimicrobial susceptibility of Enterobacteriaceae pathogens isolated from intensive care units and surgical units in Russia

Surveillance of antimicrobial susceptibility of Enterobacteriaceae pathogens isolated from intensive care units and surgical units in Russia Feb. 2016 THE JAPANESE JOURNAL OF ANTIBIOTICS 69 1 41 41 Surveillance of antimicrobial susceptibility of Enterobacteriaceae pathogens isolated from intensive care units and surgical units in Russia IRINA

More information

Chalhoub et al. Avibactam and resistance mechanisms in P. aeruginosa - Page 1 of 17

Chalhoub et al. Avibactam and resistance mechanisms in P. aeruginosa - Page 1 of 17 Chalhoub et al. Avibactam and resistance mechanisms in P. aeruginosa - Page 1 of 17 International Journal of Antimicrobial Agents (IJAA-D-17-00824) Short communication (In press) Loss of activity of ceftazidime-avibactam

More information

HOSPITAL EPIDEMOLOGY AND INFECTION CONTROL: STANDARD AND TRANSMISSION-BASED ISOLATION

HOSPITAL EPIDEMOLOGY AND INFECTION CONTROL: STANDARD AND TRANSMISSION-BASED ISOLATION Appendix 1: Carbapenem-Resistant Enterobacteriacaea (CRE) I. Definition: 2015 CDC definition of CRE are Enterobacteriaceae 1 that are: A. Resistant to any carbapenem antimicrobial (i.e., minimum inhibitory

More information

Sepsis Treatment: Early Identification Remains the Key Issue

Sepsis Treatment: Early Identification Remains the Key Issue Sepsis Treatment: Early Identification Remains the Key Issue Marin H. Kollef, MD Professor of Medicine Washington University School of Medicine Director, Medical Critical Care Director, Respiratory Care

More information

STRUCTURE OF COMMONLY USED PENICILLINS

STRUCTURE OF COMMONLY USED PENICILLINS PENICILLINS Alice Prince I. CHEMISTRY A basic structure of penicillins consists of a nucleus with three components: a thiazolidine ring, a β-lactam ring and a side chain. The side chain determines, in

More information

Helen Heffernan and Rosemary Woodhouse Antibiotic Reference Laboratory, Institute of Environmental Science and Research Limited (ESR); July 2014.

Helen Heffernan and Rosemary Woodhouse Antibiotic Reference Laboratory, Institute of Environmental Science and Research Limited (ESR); July 2014. Annual survey of extended-spectrum -lactamase (ESBL)-producing Enterobacteriaceae, 2013 Helen Heffernan and Rosemary Woodhouse Antibiotic Reference Laboratory, Institute of Environmental Science and Research

More information

University of Alberta Hospital Antibiogram for 2007 and 2008 Division of Medical Microbiology Department of Laboratory Medicine and Pathology

University of Alberta Hospital Antibiogram for 2007 and 2008 Division of Medical Microbiology Department of Laboratory Medicine and Pathology University of Alberta Hospital Antibiogram for 2007 and 2008 Division of Medical Microbiology Department of Laboratory Medicine and Pathology This material is supported in part by unrestricted educational

More information

Evaluation of the in vitro activity of ceftazidime-avibactam and ceftolozane-tazobactam

Evaluation of the in vitro activity of ceftazidime-avibactam and ceftolozane-tazobactam AAC Accepted Manuscript Posted Online 14 March 2016 Antimicrob. Agents Chemother. doi:10.1128/aac.02969-15 Copyright 2016, American Society for Microbiology. All Rights Reserved. 1 2 Evaluation of the

More information

Activity of the new cephalosporin CXA-101 (FR264205) against Pseudomonas aeruginosa isolates from chronically-infected cystic fibrosis patients

Activity of the new cephalosporin CXA-101 (FR264205) against Pseudomonas aeruginosa isolates from chronically-infected cystic fibrosis patients ORIGINAL ARTICLE BACTERIOLOGY Activity of the new cephalosporin CXA-101 (FR264205) against Pseudomonas aeruginosa isolates from chronically-infected cystic fibrosis patients L. Zamorano 1, C. Juan 1, A.

More information

Enterobacteriaceae with acquired carbapenemases, 2015

Enterobacteriaceae with acquired carbapenemases, 2015 Enterobacteriaceae with acquired carbapenemases, 2015 Background The acquired or transferable (as opposed to chromosomally encoded) carbapenemases found in Enterobacteriaceae belong to three of the four

More information

Pseudomonas aeruginosa may accumulate drug resistance mechanisms without losing its ability to cause bloodstream infections ACCEPTED

Pseudomonas aeruginosa may accumulate drug resistance mechanisms without losing its ability to cause bloodstream infections ACCEPTED AAC Accepts, published online ahead of print on August 00 Antimicrob. Agents Chemother. doi:./aac.000-0 Copyright 00, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights

More information

Clinical Microbiology Newsletter

Clinical Microbiology Newsletter Clinical Microbiology Newsletter $88 Vol. 30, No. 10 www.cmnewsletter.com May 15, 2008 Newer β-lactamases: Clinical and Laboratory Implications, Part I * Ellen Smith Moland, B.S.M.T., Soo-Young Kim, M.D.,

More information

β CARBA Test Rapid detection of carbapenemase-producing Enterobacteriaceae strains Contents 1. INTENDED USE

β CARBA Test Rapid detection of carbapenemase-producing Enterobacteriaceae strains Contents 1. INTENDED USE β CARBA Test 25 68260 Rapid detection of carbapenemase-producing Enterobacteriaceae strains 881159 2015/05 Contents 1. INTENDED USE 2. SUMMARY AND EXPLANATION OF THE TEST 3. PRINCIPLE OF THE PROCEDURE

More information

10/4/16. mcr-1. Emerging Resistance Updates. Objectives. National Center for Emerging and Zoonotic Infectious Diseases. Alex Kallen, MD, MPH, FACP

10/4/16. mcr-1. Emerging Resistance Updates. Objectives. National Center for Emerging and Zoonotic Infectious Diseases. Alex Kallen, MD, MPH, FACP National Center for Emerging and Zoonotic Infectious Diseases Emerging Resistance Updates Alex Kallen, MD, MPH, FACP Lead Antimicrobial Resistance and Emerging Pathogens Team Prevention and Response Branch

More information

Antibiotic Resistance Pattern of Blood and CSF Culture Isolates At NHLS Academic Laboratories (2005)

Antibiotic Resistance Pattern of Blood and CSF Culture Isolates At NHLS Academic Laboratories (2005) Antibiotic Resistance Pattern of Blood and CSF Culture Isolates At NHLS Academic Laboratories (2005) Streptococcus pneumoniae (SP) Blood Culture Isolates Penicillin intermediate Penicillin Cefotaxime 336

More information