Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2018 Dec 21;63(1):e01110-18. doi: 10.1128/AAC.01110-18

New Treatment Options against Carbapenem-Resistant Acinetobacter baumannii Infections

Burcu Isler a, Yohei Doi b, Robert A Bonomo c,d, David L Paterson e,
PMCID: PMC6325237  PMID: 30323035

Carbapenem-resistant Acinetobacter baumannii (CRAB) is a perilous nosocomial pathogen causing substantial morbidity and mortality. Current treatment options for CRAB are limited and suffer from pharmacokinetic limitations, such as high toxicity and low plasma levels.

KEYWORDS: Acinetobacter, cefiderocol

ABSTRACT

Carbapenem-resistant Acinetobacter baumannii (CRAB) is a perilous nosocomial pathogen causing substantial morbidity and mortality. Current treatment options for CRAB are limited and suffer from pharmacokinetic limitations, such as high toxicity and low plasma levels. As a result, CRAB is declared as the top priority pathogen by the World Health Organization for the investment in new drugs. This urgent need for new therapies, in combination with faster FDA approval process, accelerated new drug development and placed several drug candidates in the pipeline. This article reviews available information about the new drugs and other therapeutic options focusing on agents in clinical or late-stage preclinical studies for the treatment of CRAB, and it evaluates their expected benefits and potential shortcomings.

INTRODUCTION

Acinetobacter baumannii has evolved as a significant hospital pathogen by its ability to resist desiccation, disinfectants, and major antimicrobials (1). Today, a substantial proportion of these isolates are carbapenem-resistant A. baumannii (CRAB), i.e., extensively drug-resistant (XDR) or pandrug-resistant (PDR) A. baumannii (2). Carbapenem resistance rates exceed 90% in some parts of the world (3), and mortality rates for the most common CRAB infections, i.e., hospital-acquired pneumonia (HAP) and bloodstream infections (BSI), may approach 60% (4). This is significantly higher than rates for carbapenem-susceptible A. baumannii infections (5). Current antimicrobials for CRAB (i.e., polymyxins, tigecycline, and sometimes aminoglycosides) are far from perfect therapeutic options due to their pharmacokinetic properties and increasing resistance rates.

In these dire circumstances, the need for new therapeutic options for the treatment of CRAB infections is indisputable. Since antimicrobial discovery and resistance development to the new antimicrobial are nearly simultaneous, drugs with novel mechanisms of action that will overcome current resistance mechanisms are sought after. Currently, there are a small number of drug candidates and other therapeutic options that may meet those expectations. This review will focus on such drugs that are in clinical or late preclinical studies (Table 1). In addition, phage therapy and monoclonal antibodies for CRAB will be evaluated. The purpose of this review is to describe the state of anti-CRAB therapies by providing an overall picture of the current pipeline. As all antimicrobials face the eventual fate of being defeated by the bacteria, possible resistance mechanisms against new therapies will also be evaluated. This review is expected to guide the development of new and better drugs and to inspire further novel therapeutic options against resistant microorganisms.

TABLE 1.

New therapeutic options

Drug Preclinical Phase I Phase II Phase III FDA approved
Siderophore cephalosporins
    Cefiderocol -
    Others
        GSK-3342830 -a - - -
        Fimsbactin plus daptomycin - - - -
        GT-1 - - - -
Tetracyclines
    Eravacycline
    TP-6076 - - -
New non-β-lactam–β-lactamase inhibitors paired with existing β-lactam antibiotics
    ETX2514 plus sulbactam - -
    WCK 4234 plus meropenem (WCK-5999) - - - -
    LN-1-255 plus meropenem-imipenem - - - -
    VNRX-5113 (partner β-lactam unspecified) - - -
    WCK 5153 plus sulbactam - - - -
    Zidebactam plus cefepime - - -
New β-lactam antibiotics
    AIC-499 (partner β-lactam inhibitor unspecified) - - -
    FSI-1671 plus sulbactam - - -
Polymyxin B-derived molecules
    SPR741 - - -
    FADDI-287 - - - -
Aminoglycosides
    Apramycin - - - -
Other drug candidates
    LpxC inhibitors - - - -
    RX-P873 - - - -
Other therapeutic options
    Bacteriophage therapy -
    Monoclonal antibodies
        C8 - - - -
        AR-401 - - - -
a

-, terminated and under review.

LIMITATIONS OF CURRENT THERAPEUTIC OPTIONS

Polymyxins, discovered in the 1950s and abandoned in the 1980s due to their toxicity profile and availability of cephalosporins and carbapenems, are currently in use as first-line antimicrobials against CRAB, alone or in combination with other drugs. Polymyxins generally have potent in vitro activity against A. baumannii strains; however, they suffer from a lack of clinically relevant susceptibility breakpoints, very narrow therapeutic spectrum, and serious side effects of nephrotoxicity and neurotoxicity (6). Resistance emerging during therapy due to colistin-heteroresistant strains and difficulty in determining heteroresistance are other important issues that may result in unfavorable clinical outcomes (7). With an undefined optimal dosing, high toxicity, and increasing resistance (79), polymyxin-based therapies are far from being safe and effective for the treatment of CRAB infections (Table 2).

TABLE 2.

Limitations of current therapeutic options

Issue Colistin Tigecycline Minocycline Amikacin Sulbactam
Pharmacokinetic issues
    Narrow therapeutic spectrum
    Low or inconsistent drug levels
        Plasma
        Lung
        Urine
    Toxicity
        Nephrotoxicity
        Neurotoxicity
Resistance
    High resistance rates
    Heteroresistance
    Breakthrough
Only in combination
Increased mortality

Colistin-based combination therapies have been preferred over colistin monotherapy given colistin’s suboptimal pharmacokinetics and pharmacodynamics. However, randomized controlled trials (RCTs) have not yet proven the superiority of combination therapy over colistin monotherapy. A recent international open-label RCT comparing colistin plus meropenem versus monotherapy with colistin against serious carbapenem-resistant Gram-negative infections did not show a statistical difference in mortality rates between two treatment arms (10). BSI and ventilator-associated pneumonia (VAP) constituted 87% of the infections, and CRAB was the primary pathogen (77%). Three previous RCTs which compared colistin with colistin-rifampin or colistin-fosfomycin in CRAB infections, including BSI, did not find evidence for the superiority of combination therapy in mortality (1113). It should be noted that more than 70% of those in the Durante-Mangoni et al. study receiving colistin without rifampin actually received other antibiotics (12). In the study by Aydemir et al. (11), the mortality rate was in fact lower in the colistin-rifampin arm (8/21 [38.1%]) than in the colistin arm (14/22 [63.6%]), but the difference did not reach statistical significance (P = 0.171). Until the results of a large well-designed double-blind randomized trial comparing the colistin-carbapenem combination versus colistin monotherapy (14) are available, it may still be considered that the question of combination therapy is not definitively answered.

Tigecycline, developed for the treatment of multidrug-resistant (MDR) pathogens and having potent in vitro activity against A. baumannii, has been in use for the treatment of CRAB infections since 2006 (15). Though only licensed for complicated intra-abdominal infections (cIAI), skin and soft tissue infections (cSSSI), and community-acquired bacterial pneumonia (CABP), it has been widely used for the treatment of various other infections caused by CRAB (16). Yet, tigecycline suffers from pharmacokinetic issues, such as low plasma levels, limiting its use in BSI. In addition, its use in VAP was hampered when a phase III RCT showed tigecycline to be inferior to the comparator drug and to have higher mortality rates for VAP patients (17). The results from several recent studies and a meta-analysis are in line with these discouraging results (1820), and these combined with increasing rates of tigecycline resistance among CRAB disfavor its use.

Minocycline is an older tetracycline with considerable in vitro activity against CRAB. It is the only agent against CRAB which can be administered by the oral route. High susceptibility rates (72.1% in the United States and 81.4% in Thailand) were shown in some studies (21), although resistant strains are not infrequent (22, 23). Lashinsky et al. recently reviewed seven retrospective studies which evaluated the use of minocycline alone or in combination with other agents against MDR A. baumannii infections and found high clinical and microbiological success rates (78.2% and 50% to 89%, respectively) (21). However, one should be cautious while interpreting the results of this review, since there were only 126 patients, 94 of whom were treated with combination therapy, and most patients (74.6%) suffered from respiratory tract infections. A new intravenous formulation of minocycline was placed in the U.S. market in 2015, and a pharmacokinetic study (Acute Care Unit MINocycline [ACUMIN]) to determine optimal dosing for the critically ill is ongoing (24). Although high susceptibility rates in some series and good safety profile are encouraging, minocycline’s role as an anti-CRAB agent needs further investigation.

Amikacin is another drug used as an anti-CRAB agent because it retains in vitro activity against some strains. However, its nephrotoxicity and high resistance rates (68% to 100%) limit its systemic use (3). A large RCT comparing inhaled amikacin as adjunctive therapy to the standard of care in VAP patients failed to demonstrate its superiority over the standard of care and a placebo (25).

Sulbactam is a β-lactamase inhibitor with intrinsic activity against A. baumannii. The efficacy of sulbactam-containing regimens has been explored in several studies, and it is suggested as an anti-CRAB drug (26); higher doses were found to be effective against CRAB in some small studies (27). However, as with amikacin, high resistance rates among CRAB may limit its potential (28).

Various combinations of these drugs have been used for the treatment of CRAB infections; yet, combination therapies were not found to be superior over monotherapies in preventing resistance and improving clinical outcomes, even though the combination therapies may be better at microbiologic eradication (2932).

None of the recent combinations of β-lactams and β-lactamase inhibitors, i.e., ceftazidime-avibactam, ceftolozane-tazobactam, meropenem-vaborbactam, or imipenem-relebactam, have clinically useful activity against CRAB, and the activity of the new aminoglycoside plazomicin is limited to a minority of strains, which may partly be explained by the increasing frequency of ribosomal methyltransferases in A. baumannii (33).

NEW THERAPEUTIC OPTIONS

Siderophore cephalosporins.

(i) Cefiderocol. Cefiderocol (S-649266) is a recently developed novel cephalosporin conjugated with a catechol siderophore on its side chain. Cefiderocol has a distinctive active uptake mechanism and stability against many β-lactamase classes, which provide enhanced penetration of bacterial cell and activity against highly resistant Gram-negative bacteria, including CRAB (34).

In in vitro studies, cefiderocol was shown to be potent against OXA-23, OXA-40, and OXA-58 as well as NDM- and IMP-producing A. baumannii isolates, with MIC90s ranging from 1 to 8 µg/ml (3540). However, it seems to be less active against some strains of A. baumannii that express OXA-23 and OXA-40 (38, 40, 41).

Characterization of the carbapenemases of CRAB strains from a global surveillance study with a large collection of CRAB isolates showed that the MIC90 was 1 µg/ml for OXA producers and GES producers and 4 µg/ml for NDM producers (41). Among 689 CRAB strains, there were 22 strains with MICs of >4 µg/ml (8, 16, 32, and 64 µg/ml). Twenty-one of these were OXA producers (15 OXA-23 strains and 6 OXA-40 strains), and one strain was a GES producer (Table 3). Another surveillance study, which tested the isolates collected 1 year later, showed that the MIC90 and the highest MIC for cefiderocol were 2 µg/ml and >256 µg/ml, respectively (42). Twenty-two of 558 strains had MICs of >4 µg/ml. The carbapenemase types of these strains have not yet been characterized. In two other studies, CRAB strains with cefiderocol MICs of ≥8 µg/ml similarly were OXA-23 (7 and 2 strains) and OXA-24 (0 and 1 strain) producers (38, 40). Although susceptibility breakpoints for cefiderocol have not yet been established, a recent study defined “resistant” as an MIC of ≥16 µg/ml and identified 6 resistant CRAB strains, two of which were OXA-23 producers (43). The cefiderocol MIC90 was 1 µg/ml in another collection of CRAB strains, and there were 28 strains out of 758 with MICs of >8 µg/ml (44). Interestingly, in a study from Greece, there were not any CRAB strains with MICs of >2 µg/ml (45).

TABLE 3.

Studies evaluating cefiderocol against CRABa

Reference no. β-Lactamase(s) No. of CRAB isolates No. of isolates with MIC >4 or 8 μg/mlb MIC data (μg/ml)
MIC50 MIC90 Range
35 OXA 101 NA 0.25 1 ≤0.03 to >64
36 NA 368 38 0.25 8 0.015 to >256
37 NA 173 (NoA)c 24 0.25 1 ≤0.002 to 8
595 (EU) 0.12 1 0.004 to 64
41 NDM 2 0 NA 4 NA
OXA-23 543 15 0.12 1 ≤0.02 to 16
OXA-40 124 6 0.12 1 0.04 to 64
OXA-58 13 0 0.12 1 0.06 to 1
GES 7 1 NA NA 0.25 to 8
42 NA 558 22 0.5 2 ≤0.002 to >256
136 NA 44 0 0.12 1 0.015 to 4
44 NA 758 28 NA 1 ≤0.004 to 64
39 OXA-23 1 NA NA 0.063 NA
OXA-58 1 NA NA 1 NA
OXA-26, OXA-51-like 1 NA NA 0.5 NA
OXA-51-like 1 NA NA 0.5 NA
OXA-23, OXA-51-like 1 NA NA ≤0.031 NA
OXA-48 1 NA NA ≤0.031 NA
45 NA 107 0 0.06 0.5 0.03 to 2
38 OXA-23/-40/-58/-72 85 7 0.12 4 0.03 to 64
NDM-1, IMP-4 2 NA NA NA NA
137 OXA-23 5 0 NA NA 0.03 to 0.5
40 Random strains 104 0 0.125 2 ≤0.063 to 4
All BL carriers 29 0 0.5 8 0.03 to >32
IMP-1 2 0 NA NA 0.12 to 0.25
OXA-23 12 2 NA NA 0.03 to >32
OXA-24 8 1 NA NA 0.12 to 8
OXA-51/ISAba1 2 0 NA NA 0.5 to 1
OXA-58 5 0 NA NA 0.12 to 4
a

NA, not available; BL, β-lactamase.

b

MIC was >4 µg/ml for studies 35, 36, 37, and 41, >8 µg/ml for study 42, and ≥8 µg/ml for studies 38 and 40.

c

NoA, North America; EU, Europe.

For in vivo studies, the efficacy of humanized exposures of cefiderocol was evaluated in animal infection models. In immunocompetent rats infected with CRAB clinical isolates, a 2-g equivalent of cefiderocol infused every 8 h (3 h infusion) decreased the microbiologic load by >3 log10 after 4 days (46). In another study, immunocompromised mice were infected with CRAB, and a ≥1-log10 reduction in microbiologic load was observed 24 h after 2 g equivalent of cefiderocol was infused every 8 h (3 h infusion) (47). In the second study, the ≥1-log10 reduction was observed only in 2 of 28 strains (19 A. baumannii strains) with MICs of ≥8 µg/ml, whereas the growth of most isolates with MICs of ≤4 µg/ml could be reduced by ≥1 log10, suggesting a susceptibility breakpoint for cefiderocol.

In clinical studies, cefiderocol was found to be superior to imipenem-cilastatin for the treatment of complicated urinary tract infections (cUTI) in a phase III RCT (48). The primary pathogens were Escherichia coli and Klebsiella pneumoniae, and there were not any CRAB strains among other pathogens. Two other phase III trials about cefiderocol are currently recruiting patients. One of them compares cefiderocol with “best available therapy” for severe infections (cUTI, BSI, HAP/VAP, and sepsis), and the other one compares the drug with meropenem for health care-associated pneumonia (HCAP), HAP, and VAP (49, 50). It is expected that both of these studies will enroll considerable numbers of patients with CRAB infection.

(ii) Other siderophores. GSK-3342830 is another siderophore cephalosporin with considerable activity against CRAB (51, 52). A collection of 94 A. baumannii clinical isolates (71.3% carbapenem nonsusceptible) had MIC50/90 values of 0.06/0.5 µg/ml, and the MIC range was ≤0.03 to 4 µg/ml. However, phase I trials of GSK-3342830 have been terminated (53).

Fimsbactin, an A. baumannii-selective siderophore, was coupled to daptomycin by Ghosh et al., with the hypothesis that coupling of a Gram-negative-targeted siderophore to daptomycin could help transfer daptomycin directly into the cytoplasm, bypassing the need for outer membrane penetration. The resulting conjugate had potent in vivo and in vitro activity against 10 reference A. baumannii strains, including MDR strains (54). For in vivo studies, mice infected with A. baumannii ATCC 17961 were treated with ciprofloxacin, daptomycin, or fimsbactin conjugate. Saline treatment was used as a control. All mice in the treatment groups other than fimsbactin conjugate were dead after day 1, whereas 4 out of 5 mice treated with fimsbactin conjugate survived (54). Further development of fimsbactin-daptomycin is unknown at this stage. It is worth considering the inactivation of daptomycin in the lungs, which may diminish the potential utility of this investigational agent for CRAB respiratory tract infections.

GT-1, siderophore cephalosporin developed by Geom Therapeutics, is claimed to have activity against CRAB both in vitro and in vivo in mice. According to the information obtained from the drug developer company’s website, it will enter phase I clinical studies in 2018 under a contract with the National Institute of Allergy and Infectious Diseases (55).

Tetracyclines.

(i) Eravacycline. In in vitro studies, eravacycline (TP-434), a novel fluorocycline of the tetracycline family, shows activity against a broad range of pathogens, including MDR and XDR Gram-negative, Gram-positive, and anaerobic pathogens. Eravacycline MICs were found to be 2- to 8-fold lower than tigecycline MICs against CRAB (28, 5658) (Table 4). The drug is also active against colistin-resistant and ceftazidime-avibactam-resistant strains (28, 59). The eravacycline MIC50/90 values for CRAB were 1 and 2 µg/ml in a large collection of CRAB isolates (60). Similar values were shown (MIC50/90, 0.5/1 µg/ml) in several other studies where isolates produced OXA-23, OXA-40, or OXA-58 or had overexpression of OXA-51 (28, 57, 58). The production of OXA carbapenemases did not change the eravacycline MICs for CRAB, whereas NDM-, OXA-48-, or VIM-producing strains had MIC90s of 4 to 8 µg/ml (61, 62). Increased eravacycline MIC values were also associated with increased expression of the efflux pump AdeABC. Eravacycline MIC and the expression of adeB showed a significant correlation among 38 A. baumannii isolates (26 CRAB isolates) which did not carry any of the OXA genes (MIC range, 0.06 to 4 µg/ml) (58). Eravacycline maintained potency against isolates carrying tetracycline efflux pump genes (59).

TABLE 4.

Studies evaluating eravacycline against CRAB

Reference no. β-Lactamase(s) No. of CRAB isolates No. of isolates with MIC >4 μg/ml MIC data (μg/ml)
MIC50 MIC90 Range
56 163 0.5 1 0.12–8
59 55 0.5 1 0.031–2
28 OXA-23 231 0.5 1
OXA-40 17 0.25 1
OXA-58 27 0.5 0.5
OXA-51a 9 0.125–0.5
Overall 286b 1 0.5 1 ≤0.06–8
57 NDM 5 0 0.13–0.25
OXA-23/-40/-51/-58 39 0 0.5 1 ≤0.06–2
Tigr plus OXA-23c 5 2 4 8 1.0–8.0
Overall 59d 2 0.5 1 ≤0.06–8
58 OXA-23 58 1
OXA-24 2
KPC 1
adeB expression 38e 0.5 2 0.06–4
Overall 158f 0.5 1 ≤0.015–8
61 NDM 62
OXA-48 27
VIM 15
Overall 104 0.5 4
138 OXA 8
NDM 4
Total 14 0.25 1 0.031–2
139 193 1 2 0.12–8
62 NDM, OXA-48, VIM 16 0.125 8
60 707 1 2 0.06–8
140 52 0.5 2 ≤0.016–4
a

Hyperproduced.

b

One isolate with OXA-23 and OXA-58, 1 isolate with NDM-1.

c

Tigr, tigecycline resistant.

d

Forty-nine CRAB isolates and 10 carbapenem-susceptible A. baumannii isolates.

e

Twenty-six CRAB isolates and 12 carbapenem-susceptible A. baumannii isolates.

f

One hundred nine CRAB isolates and 49 carbapenem-susceptible A. baumannii isolates.

The efficacy of eravacycline was shown in several animal studies (63) and in a phase III clinical trial in the setting of cIAI. The drug was noninferior to ertapenem in patients with cIAI, but septic patients and patients with high acute physiology and chronic health evaluation (APACHE) II scores were excluded (64). There were only 3 and 4 CRAB isolates among 220 and 226 isolates in the eravacycline and ertapenem arms, respectively. In a successive RCT, the drug was found to be noninferior to meropenem for the treatment of cIAI (65). Five hundred patients were randomized (39.8% complicated appendicitis and 60.2% other diagnoses, including complicated cholecystitis, intestinal perforation, and stomach/duodenal perforation), and there were 12 Acinetobacter spp. among 400 isolates. The U.S. FDA has approved eravacycline for the treatment of cIAI in adults age 18 years and older. However, eravacycline failed an initial phase III cUTI trial and was found to be inferior to levofloxacin and subsequently in a second phase III cUTI study versus ertapenem (66).

(ii) TP-6076. TP-6076 is another fluorocycline antibiotic being developed for the treatment of MDR pathogens. TP-6076 MICs were very low (MIC range, 0.008 to 0.5 µg/ml) against clinical CRAB isolates producing OXA carbapenemases, including 44 colistin-resistant strains (67) (Table 5). Eravacycline and tigecycline MIC50/90 values were 0.5/1 and 1/2 µg/ml, respectively, for the same isolates (67). The TP-6076 MIC50/90 values were 0.03/0.06 µg/ml when tested against 121 clinical isolates from Greek hospitals between 2015 and 2017 (68). TP-6076 activity was minimally affected by recombinant overexpression of several tetracycline efflux pumps, and it was also potent against clinical CRAB isolates expressing tetA (3 isolates) and tetB (18 isolates) efflux pump genes, showing a maximum MIC90 of 1 µg/ml (69). However, TP-6076 is a substrate of the AdeABC efflux pump (69). TP-6076 was shown to be bactericidal in a mouse CRAB pneumonia model, where a ≥3-log10 reduction in mean lung CFU was observed at 24 h (69). The drug is currently being evaluated in phase I trials (70).

TABLE 5.

Studies evaluating TP6076 against CRAB

Reference no. β-Lactamase(s) No. of CRAB isolates No. of isolates with MIC >4 μg/ml MIC data (μg/ml)
MIC50 MIC90 Range
68 121 0.03 0.06 ≤0.002–0.12
67 Overall 326 0a 0.06 0.125 0.008–0.5
OXA-23 255
OXA-40 23
0XA-58 36
OXA-51b 10
NDM 1
141 41 0 0.008 0.063 002–0.25
a

All isolates had MICs of <0.5 µg/ml.

b

Overexpressed.

New non-β-lactam–β-lactamase inhibitors paired with existing β-lactam antibiotics.

(i) ETX2514. ETX2514 is a broad-spectrum diazabicyclooctanone (DBO) β-lactamase inhibitor similar to avibactam and relebactam. ETX2514 inhibits penicillin-binding protein 2 (PBP2) and enhances β-lactam activity (71). It has been developed by modifying the DBO scaffold to cover a broad range of OXA β-lactamases (72). EXT2514 is being developed in combination with sulbactam. Sulbactam-ETX2514 is a potent combination against CRAB, whereas combinations with imipenem and meropenem did not decrease the MICs to susceptible levels (72). Sulbactam-ETX2514 was found to be active against a large collection of CRAB strains (91% OXA carriers) (MIC50/90, 1/4 µg/ml) and remained active against 56 colistin-resistant strains with an MIC of 2 µg/ml (73). Similar MICs of sulbactam-ETX2514 were observed among 84 CRAB isolates, 54% of which encoded class A in addition to class C and D β-lactamases (60 strains encoded OXA-23, OXA-40, and OXA-58) (72). It should be noted that the number of isolates with MICs of >4 µg/ml were 4 among 731 isolates in those studies (Table 6). The frequency of spontaneous resistance to sulbactam-ETX2514 is low (74).

TABLE 6.

Studies evaluating ETX2514 (4 µg/ml) against CRAB

Reference no. β-Lactamase(s) No. of CRAB isolates No. of isolates with MIC >4 μg/ml BL drug in combination MIC data (μg/ml)
MIC50 MIC90 Range
72 Variousa 195 IPM 0.5 16 ≤0.06 to >64
MEM 1 16 ≤0.06 to >64
CAZ 8 32 ≤0.06 to >64
SUL 1 4 ≤0.06 to 32
ATM 32 >64 ≤0.06 to >64
84 SUL 2 4 0.25 to 16
73 Overall 731 4 SUL 1 4 ≤0.06 to 32
OXA 668 3 1 4 ≤0.06 to 32
MBL 1 1 32
GES 6 0 0.5 to 4
ESBLb 5 0 1.0 to 2.0
bla negative 51 0 2 4 0.25 to 4
142 OXA-23, OXA-58 72c SUL 1 2
a

Of the strains, 52% encoded class A in addition to class C and D β-lactamases, and 60 strains harbored either OXA-23, OXA-40, or OXA-58.

b

ESBL, extended-spectrum β-lactamase.

c

The total number of A. baumannii isolates was 72, and more than half of the isolates were MDR.

In vivo studies of sulbactam-ETX2514 showed a dose-dependent reduction in MDR A. baumannii bacterial counts, and >1-log (72) or >2-log (75) drops in CFU counts were achieved when sulbactam concentrations exceeded the combination MIC of 0.5 µg/ml for 50% of the dosing interval. The strain used in these murine infection models did not harbor any OXAs other than OXA-72 and OXA-66 (72). A phase I study investigating ETX2514 in combination with either sulbactam or imipenem-cilastatin is completed, and the combinations have been found to be generally safe among healthy volunteers (76, 77). Another phase I study conducted to determine plasma and intrapulmonary concentrations of ETX2514 and sulbactam in healthy subjects is completed, but the results are not published yet (78). A further phase I study to evaluate ETX2514-sulbactam in patients with renal impairment and a phase II study to evaluate the drug in hospitalized patients with cUTI are ongoing (79, 80).

(ii) WCK 4234. WCK 4234 is another DBO β-lactamase inhibitor being developed in combination with meropenem as WCK-5999. WCK 4234 is active against several carbapenemases from classes A, C, and D, including OXA-23 and OXA-51 (71, 81, 82).

The WCK 4234 MIC50/90 values were 2/8 µg/ml when combined with meropenem at a dose of 8 µg/ml against a large collection of A. baumannii isolates (83, 84). In this study, 62.8% of the isolates were carbapenem nonsusceptible, but carbapenem resistance genes were not identified. In another study, the MIC50/90 values of the meropenem-WCK 4234 combination were 2/4 µg/ml against OXA-23-producing CRAB strains (82). The same combination was less effective against OXA-40-producing strains (MIC50/90, 4/8 µg/ml). Similarly, both meropenem and imipenem MICs were reduced to 2 µg/ml or below by WCK 4234 in 9 of 10 OXA-23-producing CRAB isolates, whereas OXA-40-producing isolates stayed resistant (81). WCK 4234 reduced imipenem but not meropenem MICs for isolates producing OXA-58 (Table 7). OXA-51 hyperproducers were also suppressed with WCK 4234-potentiated carbapenems, whereas metallo-β-lactamase (MBL) producers remained resistant (81).

TABLE 7.

Studies evaluating WCK 4234 against CRAB

Reference no. β-Lactamase(s) No. of CRAB isolates No. of isolates with MIC >4 μg/ml BL drug in combination MIC data (μg/ml)
MIC50 MIC90 Range
81 OXA-23 10 0 IPM 0.25–4
0 0.5–4
1 MEM 0.5–8
0 0.5–4
OXA-40 5 IPM 1.0–32.0
4 0.5–16
6 MEM 1.0–32.0
4 0.5–16
OXA-51-ISAba1 0 IPM 0.5–4
0 0.25–2
0 MEM 0.25–4
0 0.5–4
OXA-58 0 IPM 1.0–4.0
0 0.5–4
1 MEM 0.5–16
1 0.5–16
NDM 5 5 IPM ≥128
5 ≥128
5 MEM ≥128
5 ≥128
5 IPM ≥128
5 ≥128
5 MEM ≥128
5 ≥128
82 OXA-23 32 3 MEM 2 4
1 2 4
OXA-24 17 16 MEM 8 16
14 4 8
Overall 55a 4 MEM 4 8
83, 84 639b MEM 2 8
a

One strain carried both OXA-23 and OXA-24, and 1 strain carried OXA-23, OXA-24, and OXA-58.

b

Of the isolates, 62.8% were CRAB.

In summary, WCK 4234 in combination with either meropenem or imipenem is effective against OXA-23 producers and OXA-51 hyperproducers. OXA-58 can be inhibited by only imipenem-WCK 4234 combinations, whereas OXA-40 and MBL producers remain resistant to imipenem or meropenem potentiated with WCK 4234. The WCK 4234-meropenem combination showed efficacy against MDR A. baumannii strains producing OXA-23 and OXA-51 in murine peritonitis and neutropenic lung infection models (71). No clinical studies have yet been commenced.

(iii) LN-1-255. LN-1-255 is a non-β-lactam–β-lactamase inhibitor from the penicillanic acid sulfone family. It is active against class D β-lactamases in vitro (85, 86). The inhibition efficiency of LN-1-255 was shown to be superior to those of tazobactam and avibactam in kinetic assays (85). LN-1-255-carbapenem combinations were tested against isogenic CRAB strains and clinical isolates producing various OXA carbapenemases, i.e., OXA-23, OXA-40, OXA-58, and OXA-143 (85). LN-1-255 showed synergy with carbapenems and decreased their MICs by 2- to 32-fold, to ≤ 2 µg/ml in transformant A. baumannii strains. Similar results were obtained with the clinical isolates, although the MICs were slightly higher than those observed in the transformants, mainly in the clinical isolate carrying OXA-40 (85).

Although OXA-48 does not carry the hydrophobic bridge important for the inhibition by LN-1-255, it was inhibited by LN-1-255-carbapenem combinations in both isogenic strains and clinical isolates of Enterobacteriaceae (86).

(iv) VNRX-5113. VNRX-5113 is a β-lactamase inhibitor which is claimed to inhibit MBL activity in MDR Gram-negative bacilli, including A. baumannii. The partner β-lactam is currently unspecified. A phase I study, funded by the NIH, was initiated in 2017. The information about the drug is obtained mainly from the drug developer’s website, and there is not yet further information in the public domain (87).

(v) WCK 5153 and zidebactam. Zidebactam (WCK 5107) and WCK 5153 are DBOs that also inhibit PBP2 and show a potent β-lactam enhancer effect against Gram-negative pathogens, including A. baumannii (71, 88). They are also active against MBL-producing K. pneumoniae strains in vitro (89). WCK 5153 and zidebactam decreased the sulbactam MIC from 16 to 2 µg/ml for MDR A. baumannii (OXA-23-producing ST2 international clone) (88). Combinations with cefepime were not as effective, although the cefepime MIC was lowered from 64 to 16 µg/ml, suggesting some enhancer effect (88). Zidebactam, in combination with cefepime (WCK 5222), is being developed as MDR/XDR therapy mainly against Pseudomonas aeruginosa rather than CRAB. Yet, cefepime-zidebactam was found to be efficacious in murine peritonitis and neutropenic lung infection models against MDR A. baumannii strains that express OXA-23 and OXA-51, among others (71). The drug completed two phase I clinical trials (90, 91), and another phase I trial to investigate the pharmacokinetics of the drug in patients with renal impairment is currently recruiting participants (92).

New β-lactam antibiotics.

(i) AIC-499. AIC-499 is a new β-lactam antibiotic being developed in combination with a β-lactamase inhibitor (currently unspecified). It is claimed to have activity against MDR A. baumannii and MDR P. aeruginosa strains. It is in phase I trials, but information is not currently registered on a government trial registry. According to the information obtained from the drug developer company’s website, a phase I study is conducted in 48 healthy volunteers in a single center at the Medical University of Vienna, Austria. This single-dose study is planned to be followed by a multiple-ascending-dose part in 36 volunteers. Clinical trials are being supported by the Innovative Medicines Initiative (IMI) within the COMBACTE-MAGNET project (93).

(ii) FSI-1671. FSI-1671 is a new class of carbapenems which possesses activity against A. baumannii. The FSI-1671-sulbactam combination was active (MIC50/90, 0.25/1 µg/ml; MIC range, ≤0.008 to 4 µg/ml) against 85 clinical A. baumannii isolates, including OXA producers, though the number of CRAB isolates is not specified (94). The combination also showed in vivo efficacy against CRAB-infected mice (95). Further development of FSI-1671 is unknown at this stage.

Polymyxin B-derived molecules.

(i) SPR741. SPR741 (formerly NAB74) is a polymyxin B (PMB)-derived antibiotic adjuvant that permeabilizes the Gram-negative membrane. It does not exhibit Gram-negative activity itself and is specifically designed to minimize nephrotoxicity. Potentiation of rifampin activity with SPR741 against A. baumannii, including clinical isolates, has been shown in several in vitro studies using checkerboard or time-kill analyses (9698). SPR741 at 8 μg/ml was able to potentiate the activities of rifampin and meropenem, reducing the MICs from 128 to 0.5 µg/ml and from 4 to 0.016 µg/ml, respectively, against 17 CRAB strains (clinical isolates and reference strains) (97). Similarly, the rifampin MIC was at least 4-fold reduced in the presence of SPR741 against 28 reference strains (57% nonsusceptible to carbapenem) (96). A SPR741-rifampin combination achieved significant lung concentrations and 2-log10 reduction in bacterial burden in murine lung infection models (96, 99). SPR741 was found to be generally well tolerated in phase I clinical trials (100, 101).

(ii) FADDI-287. FADDI-287 is a novel polymyxin analogue with an improved safety profile. It has greater potency than PMB against CRAB (102). The MIC50/90 values of FADDI-287 were 0.25/0.5 µg/ml against 210 CRAB isolates (MIC range, 0.25 to 64 µg/ml), whereas the PMB MIC50/90 values were 1/2 µg/ml (MIC range, 0.25 to 64 µg/ml) (102). FADDI-287 and PMB inhibited 91.4% and 11.4% of the isolates at an MIC of ≤0.5 µg/ml, respectively. FADDI-287 showed promising efficacy and reduced renal toxicity in mouse thigh and rat lung infection models against A. baumannii (103).

The aminoglycoside apramycin.

Apramycin is an aminoglycoside antibiotic used in veterinary medicine. Its resistance to inactivation by most aminoglycoside-modifying enzymes makes it an attractive therapeutic option against MDR Gram-negative microorganisms. The MIC50/90 values of apramycin were found to be 16/64 µg/ml against carbapenem and aminoglycoside-resistant 594 A. baumannii isolates from Greek hospitals (MIC range, 2 to 256 µg/ml), and 88.6% of the strains were inhibited at a concentration of ≤32 µg/ml, which is the apramycin breakpoint of susceptibility as per the National Antimicrobial Resistance Monitoring System (104). In a previous study, similar values were observed (MIC50/90, 8/32 µg/ml; MIC range, 2 to 256 µg/ml) against 104 A. baumannii isolates (89% CRAB isolates) (105). Apramycin efficacy was also tested in a murine thigh infection model (106). Neutropenic mice were inoculated with three strains of A. baumannii that had apramycin MICs of 2, 16, and 64 µg/ml and were treated with apramycin 2 h postinfection. There was at least a 4-log10 reduction in CFU in all three strains at 24 h postinfection (106). There are no clinical studies as of date.

Other drug candidates.

Molecules that play a role in lipid A biosynthesis have been attractive targets for antibiotic development. The most promising among them is LpxC, a zinc-dependent deacetylase. Inhibition of LpxC increases the antibiotic susceptibility of A. baumannii (107), including XDR clinical isolates (108). The MIC50/90 values of a new LpxC inhibitor were 0.8/3.2 µg/ml (MIC range, 0.5 to ≥64 µg/ml) when tested on 25 clinical A. baumannii isolates, including 19 MDR/XDR strains (7 OXA-23 producers and 1 OXA-40 producer) (109). An LpxC inhibitor, ACHN-975, failed human trials due to inflammation at the infusion site. Nevertheless, this new class of antibiotics may be promising agents for the treatment of CRAB infections if safety and pharmacokinetic properties are optimized and their activity and efficacy against CRAB infections are supported by further studies (110).

Another drug candidate with in vitro activity against CRAB is RX-P873. It binds to a conserved region in the ribosome and inhibits protein synthesis. The MIC50/90 values of RX-P873 were 0.5/1 µg/ml (MIC range, 0.12 to 4 µg/ml) against 202 clinical A. baumannii isolates (52.5% carbapenem resistant) (111). No further studies have been reported on this molecule since 2015, and the latest information from the drug developer company’s website is from 2014.

Other therapeutic options.

(i) Phage therapy. In the era of MDR microorganisms with strictly limited therapeutic options, bacteriophage therapy attracts many clinicians in spite of numerous unanswered questions about its use. Phages were discovered a century ago (112), although the first phages specific to MDR A. baumannii and CRAB were only characterized in 2010 and 2012 (113, 114). CRAB infections have been treated with phages successfully in animals (115121). Data from human infections remain scarce.

Schooley et al. treated a septic patient with phages (122). This was a 68-year-old diabetic man who developed gallstone pancreatitis and a pancreatic pseudocyst, from which PDR A. baumannii strains resistant to meropenem, amikacin, trimethoprim-sulfamethoxazole, tetracycline, ciprofloxacin, and colistin were isolated. Even though the patient was treated with combination therapies according to synergy testing results, MDR A. baumannii strain was repeatedly isolated from multiple sites. The patient eventually deteriorated and required vasopressors. At this time, the phage cocktail was administered, initially into the pseudocyst cavity and intra-abdominally and later intravenously. The patient improved with this therapy and ultimately had a favorable clinical outcome. Another patient infected with PDR A. baumannii was treated with bacteriophages by the same group (123). This was a 77-year-old man who underwent a craniectomy due to assault resulting in subdural and epidural empyema. PDR A. baumannii was isolated from intraoperative cultures. The patient did not improve with combination antibiotic therapies and was put on phage therapy. Phages were administered intravenously for 19 days. Although the craniotomy site and the skin flap healed well, the patient died on day 20. Phage therapy could not be administered intrathecally for this patient, which may be one of the reasons for the failure of phage therapy, another being the severity of his underlying condition.

In another study from Russia, one of the few countries where phage therapy is widely used (124), Bochkareva et al. presented data on 42 intensive care unit (ICU) patients treated with phages. Of those patients, 87.5% had MDR A. baumannii, K. pneumoniae, and P. aeruginosa growth in their endotracheal aspirate, blood, urine, and feces. They had received repeated doses of a phage cocktail either therapeutically or prophylactically. The phage cocktail was defined as “therapeutic and prophylactic product (TPP)” and included two phage strains per each bacterium (A. baumannii, K. pneumoniae, P. aeruginosa, and Staphylococcus aureus). The efficacy of TTP was determined 24 h after the final dose of treatment, and anti-phage immunity was detected after 2 to 3 weeks. It was shown that 54 to 62.5% of patients had “sanitation of the infected loci.” The authors stated that repeated doses of phages do not eradicate infection more efficiently than the single dose and suggest that repeated doses have undesired consequences, such as the development of anti-phage antibodies. A change in the composition of the phage strains is required for future treatments (125). In another study from Russia, 14 patients with MDR A. baumannii, K. pneumoniae, P. aeruginosa, and S. aureus growth in several samples, including endotracheal aspirate, blood, and urine, had a microbiologic response to a 3-day phage therapy (126).

These results, though promising, display the hurdles of the bacteriophage treatment process. Phages are very host specific, and preparation of the phage cocktail in advance is difficult (127). The dosing route and schedule of phages are not known for different body site infections. Schooley et al. administered phages through intracystic, intraabdominal, and intravenous routes, whereas Russian researchers used gastrointestinal (oral and intragastric) and endotracheal routes. There are not clearly defined dosing intervals currently, and unfavorable outcomes of multiple dosing may occur (125). It is necessary to detect anti-phage antibodies for a customized therapy with repeated doses. Another handicap of this therapeutic option is resistance as shown by Schooley et al, where one of the strains developed resistance to the bacteriophage cocktail 8 days after the initiation of therapy (122). This may in part be overcome with the development of new phages against the new resistant strain, which requires an active “on-demand” service for the rapid selection of new phage strains (125).

An RCT comparing phage therapy with antibiotics for cUTI in patients undergoing prostate resection is recruiting patients (128, 129). Finally, phages may have other promising uses in the fight against drug-resistant bacteria. Ho et al. used anti-CRAB phage aerosols to decontaminate patient rooms and showed that phage aerosols reduce CRAB infection rates (130).

(ii) Monoclonal antibodies. Development of monoclonal antibodies (MAbs) for the treatment of bacterial infections is challenging due to several host- and product-specific issues. Currently, there are three FDA-approved antibacterial MAb products (against Bacillus anthracis and Clostridium difficile) and at least nine MAbs (mainly against S. aureus and P. aeruginosa) that are in clinical trials (131). Monoclonal A. baumannii antibodies have been used for protection from sepsis and pneumonia and treatment of wound infections in animal models (132, 133). Nielsen et al. recently developed an MAb active against XDR A. baumannii. The new MAb, called C8, showed antibacterial activity alone and in combination with colistin against XDR A. baumannii in vitro and in mice, and it retained activity after humanization (134). AR-401 is another fully human MAb claimed to be developed against A. baumannii (135).

CONCLUSION

CRAB is arguably the most troublesome Gram-negative microorganism due to its great capacity for resistance development. New agents recently approved for drug-resistant Gram-negative bacilli are effective against KPC producers or carbapenem-resistant P. aeruginosa but not against CRAB.

Cefiderocol is likely to be the first of the new agents active against CRAB to be approved for clinical use. Its in vitro effectiveness against large collections of CRAB strains with a variety of resistance mechanisms provides much hope, as does its superiority over imipenem-cilastatin for the treatment of cUTI. However, there were very few A. baumannii strains in this clinical trial, so its real test will be in phase III trials for patients with serious infections, such as BSI and VAP. Cefiderocol-nonsusceptible strains of CRAB already exist before its approval, and a key question will be the effects of selection pressure once cefiderocol is used clinically.

Eravacycline, recently placed on the market, has better in vitro activity against CRAB than tigecycline. However, existing issues about tigecycline and the failure of eravacycline in phase III trials of cUTI diminish the expectations from this drug. Unfortunately, no trials to evaluate eravacycline efficacy on BSI and VAP are on the horizon.

Among other drug candidates, ETX2514 combined with sulbactam is in phase II studies. TP-6076, VNRX-5113, AIC-499, and SPR741 have commenced or completed phase I studies, though the available information in the public domain is limited for all of these compounds. Other new β-lactamase inhibitors (i.e., WCK 4234, LN-1-255, WCK 5153, and zidebactam) and apramycin are not yet in clinical studies, but their activity against CRAB isolates and efficacy in animal infections are promising. For future clinical trials to be informative and useful, they should include patients with severe infections from areas with high resistance rates. Such patients are the ones for whom the new drugs are needed the most.

REFERENCES

  • 1.Peleg AY, Seifert H, Paterson DL. 2008. Acinetobacter baumannii: emergence of a successful pathogen. Clin Microbiol Rev 21:538–582. doi: 10.1128/CMR.00058-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, Pulcini C, Kahlmeter G, Kluytmans J, Carmeli Y, Ouellette M, Outterson K, Patel J, Cavaleri M, Cox EM, Houchens CR, Grayson ML, Hansen P, Singh N, Theuretzbacher U, Magrini N, WHO Pathogens Priority List Working Group. 2018. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 18:318–327. doi: 10.1016/S1473-3099(17)30753-3. [DOI] [PubMed] [Google Scholar]
  • 3.World Health Organization. 2017. Central Asian and Eastern European surveillance of antimicrobial resistance. Annual report 2017. World Health Organization Regional Office for Europe, Copenhagen, Denmark: http://www.euro.who.int/__data/assets/pdf_file/0005/354434/WHO_CAESAR_AnnualReport_2017.pdf?ua=1. [Google Scholar]
  • 4.Wong D, Nielsen TB, Bonomo RA, Pantapalangkoor P, Luna B, Spellberg B. 2017. Clinical and pathophysiological overview of Acinetobacter infections: a century of challenges. Clin Microbiol Rev 30:409–447. doi: 10.1128/CMR.00058-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Esterly JS, Griffith M, Qi C, Malczynski M, Postelnick MJ, Scheetz MH. 2011. Impact of carbapenem resistance and receipt of active antimicrobial therapy on clinical outcomes of Acinetobacter baumannii bloodstream infections. Antimicrob Agents Chemother 55:4844–4849. doi: 10.1128/AAC.01728-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Landman D, Georgescu C, Martin DA, Quale J. 2008. Polymyxins revisited. Clin Microbiol Rev 21:449–465. doi: 10.1128/CMR.00006-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cai Y, Chai D, Wang R, Liang B, Bai N. 2012. Colistin resistance of Acinetobacter baumannii: clinical reports, mechanisms and antimicrobial strategies. J Antimicrob Chemother 67:1607–1615. doi: 10.1093/jac/dks084. [DOI] [PubMed] [Google Scholar]
  • 8.Giamarellou H. 2016. Epidemiology of infections caused by polymyxin-resistant pathogens. Int J Antimicrob Agents 48:614–621. doi: 10.1016/j.ijantimicag.2016.09.025. [DOI] [PubMed] [Google Scholar]
  • 9.Qureshi ZA, Hittle LE, O'Hara JA, Rivera JI, Syed A, Shields RK, Pasculle AW, Ernst RK, Doi Y. 2015. Colistin-resistant Acinetobacter baumannii: beyond carbapenem resistance. Clin Infect Dis 60:1295–1303. doi: 10.1093/cid/civ048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Paul M, Daikos GL, Durante-Mangoni E, Yahav D, Carmeli Y, Benattar YD, Skiada A, Andini R, Eliakim-Raz N, Nutman A, Zusman O, Antoniadou A, Pafundi PC, Adler A, Dickstein Y, Pavleas I, Zampino R, Daitch V, Bitterman R, Zayyad H, Koppel F, Levi I, Babich T, Friberg LE, Mouton JW, Theuretzbacher U, Leibovici L. 2018. Colistin alone versus colistin plus meropenem for treatment of severe infections caused by carbapenem-resistant Gram-negative bacteria: an open-label, randomised controlled trial. Lancet Infect Dis 18:391–400. doi: 10.1016/S1473-3099(18)30099-9. [DOI] [PubMed] [Google Scholar]
  • 11.Aydemir H, Akduman D, Piskin N, Comert F, Horuz E, Terzi A, Kokturk F, Ornek T, Celebi G. 2013. Colistin vs. the combination of colistin and rifampicin for the treatment of carbapenem-resistant Acinetobacter baumannii ventilator-associated pneumonia. Epidemiol Infect 141:1214–1222. doi: 10.1017/S095026881200194X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Durante-Mangoni E, Signoriello G, Andini R, Mattei A, De Cristoforo M, Murino P, Bassetti M, Malacarne P, Petrosillo N, Galdieri N, Mocavero P, Corcione A, Viscoli C, Zarrilli R, Gallo C, Utili R. 2013. Colistin and rifampicin compared with colistin alone for the treatment of serious infections due to extensively drug-resistant Acinetobacter baumannii: a multicenter, randomized clinical trial. Clin Infect Dis 57:349–358. doi: 10.1093/cid/cit253. [DOI] [PubMed] [Google Scholar]
  • 13.Sirijatuphat R, Thamlikitkul V. 2014. Preliminary study of colistin versus colistin plus fosfomycin for treatment of carbapenem-resistant Acinetobacter baumannii infections. Antimicrob Agents Chemother 58:5598–5601. doi: 10.1128/AAC.02435-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kaye K. 2012. Trial for the treatment of extensively drug-resistant Gram-negative bacilli. Accession no. NCT01597973. ClinicalTrials.gov, NIH; https://clinicaltrials.gov/ct2/show/NCT01597973?term=NCT01597973&rank=1. [Google Scholar]
  • 15.Taccone FS, Rodriguez-Villalobos H, De Backer D, De Moor V, Deviere J, Vincent J-L, Jacobs F. 2006. Successful treatment of septic shock due to pan-resistant Acinetobacter baumannii using combined antimicrobial therapy including tigecycline. Eur J Clin Microbiol Infect Dis 25:257–260. doi: 10.1007/s10096-006-0123-1. [DOI] [PubMed] [Google Scholar]
  • 16.Curcio D. 2009. Off-label use of antibiotics in hospitalized patients: focus on tigecycline. J Antimicrob Chemother 64:1344–1346. doi: 10.1093/jac/dkp342. [DOI] [PubMed] [Google Scholar]
  • 17.Freire AT, Melnyk V, Kim MJ, Datsenko O, Dzyublik O, Glumcher F, Chuang YC, Maroko RT, Dukart G, Cooper CA, Korth-Bradley JM, Dartois N, Gandjini H, 311 Study Group. 2010. Comparison of tigecycline with imipenem/cilastatin for the treatment of hospital-acquired pneumonia. Diagn Microbiol Infect Dis 68:140–151. doi: 10.1016/j.diagmicrobio.2010.05.012. [DOI] [PubMed] [Google Scholar]
  • 18.Liang CA, Lin YC, Lu PL, Chen HC, Chang HL, Sheu CC. 2018. Antibiotic strategies and clinical outcomes in critically ill patients with pneumonia caused by carbapenem-resistant Acinetobacter baumannii. Clin Microbiol Infect 24:908.e1–908.e7. doi: 10.1016/j.cmi.2017.10.033. [DOI] [PubMed] [Google Scholar]
  • 19.Lee YT, Wang YC, Kuo SC, Chen CT, Liu CP, Liu YM, Chen TL, Yang YS. 2017. Multicenter study of clinical features of breakthrough Acinetobacter bacteremia during carbapenem therapy: a multicenter study. Antimicrob Agents Chemother 61:e00661-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ni W, Han Y, Zhao J, Wei C, Cui J, Wang R, Liu Y. 2016. Tigecycline treatment experience against multidrug-resistant Acinetobacter baumannii infections: a systematic review and meta-analysis. Int J Antimicrob Agents 47:107–116. doi: 10.1016/j.ijantimicag.2015.11.011. [DOI] [PubMed] [Google Scholar]
  • 21.Lashinsky JN, Henig O, Pogue JM, Kaye KS. 2017. Minocycline for the treatment of multidrug and extensively drug-resistant A. baumannii: a review. Infect Dis Ther 6:199–211. doi: 10.1007/s40121-017-0153-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Evans SR, Hujer AM, Jiang H, Hill CB, Hujer KM, Mediavilla JR, Manca C, Tran TTT, Domitrovic TN, Higgins PG, Seifert H, Kreiswirth BN, Patel R, Jacobs MR, Chen L, Sampath R, Hall T, Marzan C, Fowler VG, Chambers HF, Bonomo RA. 2017. Informing antibiotic treatment decisions: evaluating rapid molecular diagnostics to identify susceptibility and resistance to carbapenems against Acinetobacter spp. in PRIMERS III. J Clin Microbiol 55:134–144. doi: 10.1128/JCM.01524-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.van Duin D, Perez F, Rudin SD, Cober E, Hanrahan J, Ziegler J, Webber R, Fox J, Mason P, Richter SS, Cline M, Hall GS, Kaye KS, Jacobs MR, Kalayjian RC, Salata RA, Segre JA, Conlan S, Evans S, Fowler VG, Bonomo RA. 2014. Surveillance of carbapenem-resistant Klebsiella pneumoniae: tracking molecular epidemiology and outcomes through a regional network. Antimicrob Agents Chemother 58:4035–4041. doi: 10.1128/AAC.02636-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Doi Y, Bonomo RA, Hooper DC, Kaye KS, Johnson JR, Clancy CJ, Thaden JT, Stryjewski ME, van Duin D. 2017. Gram-negative bacterial infections: research priorities, accomplishments, and future directions of the Antibacterial Resistance Leadership Group. Clin Infect Dis 64:30–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Nektar Therapeutics. 2013. Inhaled amikacin solution BAY41-6551 as adjunctive therapy in the treatment of Gram-negative pneumonia (INHALE 1). Accession no. NCT01799993. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT01799993.
  • 26.Viehman JA, Nguyen MH, Doi Y. 2014. Treatment options for carbapenem-resistant and extensively drug-resistant Acinetobacter baumannii infections. Drugs 74:1315–1333. doi: 10.1007/s40265-014-0267-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Betrosian AP, Frantzeskaki F, Xanthaki A, Georgiadis G. 2007. High-dose ampicillin-sulbactam as an alternative treatment of late-onset VAP from multidrug-resistant Acinetobacter baumannii. Scand J Infect Dis 39:38–43. doi: 10.1080/00365540600951184. [DOI] [PubMed] [Google Scholar]
  • 28.Seifert H, Stefanik D, Sutcliffe JA, Higgins PG. 2018. In-vitro activity of the novel fluorocycline eravacycline against carbapenem non-susceptible Acinetobacter baumannii. Int J Antimicrob Agents 51:62–64. doi: 10.1016/j.ijantimicag.2017.06.022. [DOI] [PubMed] [Google Scholar]
  • 29.Kengkla K, Kongpakwattana K, Saokaew S, Apisarnthanarak A, Chaiyakunapruk N. 2018. Comparative efficacy and safety of treatment options for MDR and XDR Acinetobacter baumannii infections: a systematic review and network meta-analysis. J Antimicrob Chemother 73:22–32. doi: 10.1093/jac/dkx368. [DOI] [PubMed] [Google Scholar]
  • 30.Zusman O, Altunin S, Koppel F, Dishon Benattar Y, Gedik H, Paul M. 2017. Polymyxin monotherapy or in combination against carbapenem-resistant bacteria: systematic review and meta-analysis. J Antimicrob Chemother 72:29–39. doi: 10.1093/jac/dkw377. [DOI] [PubMed] [Google Scholar]
  • 31.Chen Z, Chen Y, Fang Y, Wang X, Chen Y, Qi Q, Huang F, Xiao X. 2015. Meta-analysis of colistin for the treatment of Acinetobacter baumannii infection. Sci Rep 5:17091. doi: 10.1038/srep17091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Paul M, Carmeli Y, Durante-Mangoni E, Mouton JW, Tacconelli E, Theuretzbacher U, Mussini C, Leibovici L. 2014. Combination therapy for carbapenem-resistant Gram-negative bacteria. J Antimicrob Chemother 69:2305–2309. doi: 10.1093/jac/dku168. [DOI] [PubMed] [Google Scholar]
  • 33.Wright MS, Haft DH, Harkins DM, Perez F, Hujer KM, Bajaksouzian S, Benard MF, Jacobs MR, Bonomo RA, Adams MD. 2014. New insights into dissemination and variation of the health care-associated pathogen Acinetobacter baumannii from genomic analysis. mBio 5:e00963-13. doi: 10.1128/mBio.00963-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Paterson DL. 2017. Antibiotics: cephalosporins and related drugs: cefiderocol, p 658–663. In Grayson LM, Cosgrove SE, Crowe S, Hope W, McCarthy JS, Mills J, Mouton JW, Paterson DL (ed), Kucers’ the use of antibiotics: a clinical review of antibacterial, antifungal, antiparasitic, and antiviral Drugs, vol 1, 7th ed CRC Press, Boca Raton, FL. [Google Scholar]
  • 35.Jacobs M, Bajaksouzian S, Good C, Hujer K, Hujer A, Bonomo RA. 2018. In vitro activity of cefiderocol (S-649266), a siderophore cephalosporin, against carbapenem-susceptible and resistant non-fermenting Gram-negative bacteria, abstr P-0187. Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 36.Hackel MA, Tsuji M, Yamano Y, Echols R, Karlowsky JA, Sahm DF. 2017. In vitro activity of the siderophore cephalosporin, cefiderocol, against carbapenem-non-susceptible and multidrug-resistant isolates of Gram-negative bacilli collected worldwide in 2014–2016. Antimicrob Agents Chemother 20:e01968-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hackel MA, Tsuji M, Yamano Y, Echols R, Karlowsky JA, Sahm DF. 2017. In vitro activity of the siderophore cephalosporin, cefiderocol, against a recent collection of clinically relevant Gram-negative bacilli from North America and Europe, including carbapenem non-susceptible isolates: the SIDERO-WT-2014 study. Antimicrob Agents Chemother 9:e00093-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dobias J, Dénervaud-Tendon V, Poirel L, Nordmann P. 2017. Activity of the novel siderophore cephalosporin cefiderocol against multidrug-resistant Gram-negative pathogens. Eur J Clin Microbiol Infect Dis 26:2319–2327. [DOI] [PubMed] [Google Scholar]
  • 39.Ito A, Sato T, Ota M, Takemura M, Nishikawa T, Toba S, Kohira N, Miyagawa S, Ishibashi N, Matsumoto S, Nakamura R. 2017. In vitro antibacterial properties of cefiderocol, a novel siderophore cephalosporin, against Gram-negative bacteria. Antimicrob Agents Chemother 23:e01454-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ito A, Kohira N, Bouchillon SK, West J, Rittenhouse S, Sader HS, Rhomberg PR, Jones RN, Yoshizawa H, Nakamura R, Tsuji M, Yamano Y. 2016. In vitro antimicrobial activity of S-649266, a catechol-substituted siderophore cephalosporin, when tested against non-fermenting Gram-negative bacteria. J Antimicrob Chemother 71:670–677. doi: 10.1093/jac/dkv402. [DOI] [PubMed] [Google Scholar]
  • 41.Tsuji M, Kazmierczak KM, Hackel M, Echols R, Yamano Y, Sahm DF. 2017. Cefiderocol (S-649266) susceptibility against globally isolated meropenem non-susceptible Gram-negative bacteria containing serine- and metallo-carbapenemase genes, abstr SUNDAY-25. ASM Microbe, 1 to 5 June 2017, New Orleans, LA. [Google Scholar]
  • 42.Tsuji M, Hackel M, Echols R, Yamano Y, Sahm DF. 2017. In vitro antibacterial activity of cefiderocol (S-649266) against Gram-negative clinical strains collected in North America and Europe: SIDERO-WT-2015, abstr SUNDAY-11. ASM Microbe, 1 to 5 June 2017, New Orleans, LA. [Google Scholar]
  • 43.Boyd S, Anderson K, Albrecht V, Campbell D, Karlsson MS, Rasheed KJ. 2017. In vitro activity of cefiderocol against multi-drug resistant carbapenemase-producing Gram-negative pathogens, abstr 1230. IDWeek 2017, 4 to 8 October 2017, San Diego, CA. [Google Scholar]
  • 44.Ito A, Miyagawa S, Ishibashi N, Nishikawa T, Kohira N, Sato T, Tsuji M, Yamano Y. 2017. Anti-Acinetobacter activity of cefiderocol (S-649266), a novel siderophore cephalosporin: bactericidal activity due to penicillin-binding proteins inhibition and antibacterial activity against globally collected clinical isolates, abstr SATURDAY-115 ASM Microbe, 1 to 5 June 2017, New Orleans, LA. [Google Scholar]
  • 45.Falagas ME, Skalidis T, Vardakas KZ, Legakis NJ, Hellenic Cefiderocol Study Group. 2017. Activity of cefiderocol (S-649266) against carbapenem-resistant Gram-negative bacteria collected from inpatients in Greek hospitals. J Antimicrob Chemother 72:1704–1708. doi: 10.1093/jac/dkx049. [DOI] [PubMed] [Google Scholar]
  • 46.Matsumoto S, Singley CM, Hoover J, Nakamura R, Echols R, Rittenhouse S, Tsuji M, Yamano Y. 2017. Efficacy of cefiderocol against carbapenem-resistant Gram-negative bacilli in immunocompetent-rat respiratory tract infection models recreating human plasma pharmacokinetics. Antimicrob Agents Chemother 61:e00700-17. doi: 10.1128/AAC.00700-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Monogue ML, Tsuji M, Yamano Y, Echols R, Nicolau DP. 2017. Efficacy of humanized exposures of cefiderocol (S-649266) against a diverse population of Gram-negative bacteria in a murine thigh infection model. Antimicrob Agents Chemother 61:e01022-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Portsmouth S, Van Veenhuyzen D, Echols R, Machida M, Ferreira JC, Ariyasu M, Nagata TD. 2017. Clinical response of cefiderocol compared with imipenem/cilastatin in the treatment of adults with complicated urinary tract infections with or without pyelonephritis or acute uncomplicated pyelonephritis: results from a multicenter, double-blind, randomized study (APEKS-cUTI). Open Forum Infect Dis 4:S537–S538. doi: 10.1093/ofid/ofx163.1399. [DOI] [Google Scholar]
  • 49.Shionogi, Inc. 2016. Study of S-649266 or best available therapy for the treatment of severe infections caused by carbapenem-resistant Gram-negative pathogens (CREDIBLE-CR). Accession no. NCT02714595. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT02714595.
  • 50.Shionogi, Inc. 2017. Clinical study of S-649266 for the treatment of nosocomial pneumonia caused by Gram-negative pathogens (APEKS-NP). Accession no. NCT03032380. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT03032380.
  • 51.Hackel M, Butler D, Miller L, Bouchillon S, Sahm D. 2017. In vitro antibacterial activity of GSK3342830 against a global collection of clinically relevant Gram-negative bacteria, abstr SATURDAY-288. ASM Microbe, 1 to 5 June 2017, New Orleans, LA. [Google Scholar]
  • 52.Rhomberg PR, Shortridge D, Huband MD, Butler D, West J, Flamm RK. 2017. Multilaboratory broth microdilution MIC reproducibility study for GSK3342830, a novel catechol-cephem, abstr SATURDAY-287. ASM Microbe, 1 to 5 June 2017, New Orleans, LA. [Google Scholar]
  • 53.GlaxoSmithKline. 2016. A study to evaluate safety, tolerability and pharmacokinetics of ascending intravenous single dose and repeat dose of GSK3342830. Accession no. NCT02751424. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT02751424?cond=GSK3342830&rank=1.
  • 54.Ghosh M, Miller PA, Möllmann U, Claypool WD, Schroeder VA, Wolter WR, Suckow M, Yu H, Li S, Huang W, Zajicek J, Miller MJ. 2017. Targeted antibiotic delivery: selective siderophore conjugation with daptomycin confers potent activity against multidrug resistant Acinetobacter baumannii both in vitro and in vivo. J Med Chem 60:4577–4583. doi: 10.1021/acs.jmedchem.7b00102. [DOI] [PubMed] [Google Scholar]
  • 55.Geom Therapeutics. 2017. Geom Therapeutics announces agreement with National Institute of Allergy and Infectious Diseases (NIAID) for clinical Advancement of GT-1. Geom Therapeutics, San Francisco, CA. [Google Scholar]
  • 56.Olesky M, Morrissey I, Hawser S, Magnet S, Guemmaz A, Monti F, Fyfe C, Bassetti M. 2018. In vitro activity of eravacycline and comparators against resistant Gram negative isolates collected in 2016 from patients in Europe, abstr P-0099. Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 57.Livermore DM, Mushtaq S, Warner M, Woodford N. 2016. In-vitro activity of eravacycline against carbapenem-resistant Enterobacteriaceae and Acinetobacter baumannii. Antimicrob Agents Chemother 4:3840–3844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Abdallah M, Olafisoye O, Cortes C, Urban C, Landman D, Quale J. 2015. Activity of eravacycline against Enterobacteriaceae and Acinetobacter baumannii, including multidrug-resistant isolates, from New York City. Antimicrob Agents Chemother 59:1802–1805. doi: 10.1128/AAC.04809-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fyfe C, Barry R, Close B, Kerstein K, Nordmann P, Newman J. 2018. Eravacycline is active against bacterial isolates carrying emergent resistance types, abstr P-1156. Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 60.Bassetti M, Corey R, Doi Y, Morrissey I, Grossman T, Olesky M, Sutcliffe J. 2016. In vitro global surveillance of eravacycline and comparators against Enterobacteriaceae, Acinetobacter baumannii, Stenotrophomonas maltophilia, including multidrug-resistant (MDR) isolates, over a three-year period (2013-15), abstr 1825. IDWeek 2016, 26 to 30 October 2017, New Orleans, LA. [Google Scholar]
  • 61.El-Bouseary M, Tyrrell J, Walsh TR, Olesky M. 2017. Eravacycline, a novel fluorocycline, has antibacterial activity against carbapenemresistant Enterobacteriaceae (CRE) and Acinetobacter spp. (CRA), abstr P-1259. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 62.El-Bouseary M, Tyrrell J, Walsh TR, Olesky M. 2017. Antibacterial activity of eravacycline, a novel fluorocycline, compared to established antimicrobials, against contemporary clinical isolates from Tanta, Egypt, abstr P-1261. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 63.Zhanel GG, Cheung D, Adam H, Zelenitsky S, Golden A, Schweizer F, Gorityala B, Lagacé-Wiens PRS, Walkty A, Gin AS, Hoban DJ, Karlowsky JA. 2016. Review of eravacycline, a novel fluorocycline antibacterial agent. Drugs 76:567–588. doi: 10.1007/s40265-016-0545-8. [DOI] [PubMed] [Google Scholar]
  • 64.Solomkin J, Evans D, Slepavicius A, Lee P, Marsh A, Tsai L, Sutcliffe JA, Horn P. 2017. Assessing the efficacy and safety of eravacycline vs ertapenem in complicated intra-abdominal infections in the Investigating Gram-Negative Infections Treated with Eravacycline (IGNITE 1) trial: a randomized clinical trial. JAMA Surg 152:224–232. doi: 10.1001/jamasurg.2016.4237. [DOI] [PubMed] [Google Scholar]
  • 65.Horn P, Tsai L, Solomkin J, Evans D, Gardovskis J. 2018. Results of IGNITE4: a phase-3 study to evaluate the efficacy and safety of eravacycline versus meropenem in complicated intra-abdominal infections. abtr O0421. Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 66.Tetraphase Pharmaceuticals. 2015. Tetraphase announces top-line results from IGNITE2 phase 3 clinical trial of eravacycline in cUTI. Tetraphase Pharmaceuticals, Watertown, MA. [Google Scholar]
  • 67.Seifert H, Stefanik D, Sutcliffe J, Higgins PG. 2017. In-vitro activity of the novel fluorocycline TP-6076 against carbapenem non-susceptible Acinetobacter baumannii, abstr P-1364. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 68.Falagas ME, Skalidis T, Vardakas KZ, Voulgaris GL, Papanikolaou G, Legakis N. 2018. Activity of TP-6076 against carbapenem-resistant Acinetobacter baumannii isolates collected from inpatients in Greek hospitals. Int J Antimicrob Agents 52:269–271. doi: 10.1016/j.ijantimicag.2018.03.009. [DOI] [PubMed] [Google Scholar]
  • 69.Grossman T, Fyfe C, Kerstein K, Xiao XY, Sun C, Newman J, Nguyen P, Pulse M, Weiss W, Dumas J, Sutcliffe J. 2017. TP-6076 is efficacious in a mouse pneumonia model with carbapenem-resistant Acinetobacter baumannii (CRAB) and retains potency against common tetracycline-resistance mechanisms, abstr P-1310. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 70.Tetraphase Pharmaceuticals. 2017. Tetraphase Pharmaceuticals’ TP-6076 selected by CARB-X to receive $4 million in research funding. Tetraphase Pharmaceuticals, Watertown, MA. [Google Scholar]
  • 71.Papp-Wallace KM, Nguyen NQ, Jacobs MR, Bethel CR, Barnes MD, Kumar V, Bajaksouzian S, Rudin SD, Rather PN, Bhavsar S, Ravikumar T, Deshpande PK, Patil V, Yeole R, Bhagwat SS, Patel MV, van den Akker F, Bonomo RA. 2018. Strategic approaches to overcome resistance against Gram negative pathogens using β-lactamase inhibitors and β-lactam enhancers: The activity of three novel diazabicyclooctanes, WCK 5153, zidebactam (WCK 5107), and WCK 4234. J Med Chem 61:4067–4086. doi: 10.1021/acs.jmedchem.8b00091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Durand-Réville TF, Guler S, Comita-Prevoir J, Chen B, Bifulco N, Huynh H, Lahiri S, Shapiro AB, McLeod SM, Carter NM, Moussa SH, Velez-Vega C, Olivier NB, McLaughlin R, Gao N, Thresher J, Palmer T, Andrews B, Giacobbe RA, Newman JV, Ehmann DE, de Jonge B, O’Donnell J, Mueller JP, Tommasi RA, Miller AA. 2017. ETX2514 is a broad-spectrum β-lactamase inhibitor for the treatment of drug-resistant Gram-negative bacteria including Acinetobacter baumannii. Nat Microbiol 2:17104. doi: 10.1038/nmicrobiol.2017.104. [DOI] [PubMed] [Google Scholar]
  • 73.Hackel M, Bouchillon S, deJonge B, Lawrence K, Mueller J, Tommasi R, Miller A. 2016. Global surveillance of the activity of sulbactam combined with the novel β-lactamase inhibitor ETX2514 against clinical isolates of Acinetobacter baumannii from 2014, abstr 2243. IDWeek 2016, 26 to 30 October 2017, New Orleans, LA. [Google Scholar]
  • 74.McLeod SM, Shapiro AB, Moussa SH, Johnstone M, McLaughlin RE, de Jonge BL, Miller AA. 2017. The frequency and mechanism of spontaneous resistance to sulbactam combined with the novel β-lactamase inhibitor ETX2514 in clinical isolates of Acinetobacter baumannii. Antimicrob Agents Chemother 62:e01576-17. doi: 10.1128/AAC.01576-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tommasi R, Tanudra A, Chen A, Mueller J, O’Donnell J. 2017. ETX2514: responding to the global threat of nosocomial multidrug and extremely drug-resistant Gram-negative pathogens, abstr OS-0559. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 76.Entasis Therapeutics. 2016. Evaluation of the safety, tolerability and pharmacokinetics of intravenous ETX2514 administered in healthy subjects. Accession no. NCT02971423. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT02971423.
  • 77.Lickliter J, Lawrence K, O’Donnell J, Isaacs R. 2017. Safety and pharmacokinetics (PK) in humans of intravenous ETX2514, a β-lactamase inhibitor (BLI) which broadly inhibits Ambler class A, C, and D β-lactamases, abstr 1836. IDWeek 2017, 4 to 8 October 2017, San Diego, CA. [Google Scholar]
  • 78.Entasis Therapeutics. 2017. Study to determine and compare plasma and intrapulmonary concentrations of ETX2514 and sulbactam in healthy subjects. Accession no. NCT03303924. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT03303924?term=NCT03303924&rank=1.
  • 79.Entasis Therapeutics. 2017. Evaluation of the pharmacokinetics, safety, and tolerability of intravenous ETX2514 and sulbactam administered concurrently to subjects with various degrees of renal impairment and healthy matched control subjects. Accession no. NCT03310463. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT03310463?term=NCT03310463&rank=1.
  • 80.Entasis Therapeutics. 2018. Evaluation of safety and efficacy of intravenous sulbactam-ETX2514 in the treatment of hospitalized adults with complicated urinary tract infections. Accession no. NCT03445195. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT03445195?term=NCT03445195&rank=1.
  • 81.Mushtaq S, Vickers A, Woodford N, Livermore DM. 2017. WCK 4234, a novel diazabicyclooctane potentiating carbapenems against Enterobacteriaceae, Pseudomonas and Acinetobacter with class A, C and D β-lactamases. J Antimicrob Chemother 72:1688–1695. doi: 10.1093/jac/dkx035. [DOI] [PubMed] [Google Scholar]
  • 82.Castanheira M, Rhomberg PR, Lindley JM, Jones RN, Sader HS. 2016. Activity of the new carbapenem/β-lactamase inhibitor combination WCK 5999 against Gram-negative isolates producing oxacillinases (OXAs), abstr MONDAY-422. ASM Microbe, 16 to 20 June 2016, Boston, MA. [Google Scholar]
  • 83.Sader HS, Flamm RK, Huband MD, Rhomberg PR, Castanheira M. 2017. Antimicrobial activity of meropenem-WCK 4234 (WCK 5999) against clinical isolates of Acinetobacter spp. collected worldwide and stratified by infection type, abstr SATURDAY-97. ASM Microbe, 1 to 5 June 2017, New Orleans, LA. [Google Scholar]
  • 84.Huband MD, Farrell DJ, Flamm RK, Jones RN, Sader HS. 2016. In vitro activity of WCK 5999 against Acinetobacter baumannii and Pseudomonas aeruginosa isolates from a worldwide surveillance program (2015), abstr FRIDAY-476. ASM Microbe, 16 to 20 June 2016, Boston, MA. [Google Scholar]
  • 85.Vázquez-Ucha JC, Maneiro M, Martínez-Guitián M, Buynak J, Bethel CR, Bonomo RA, Bou G, Poza M, González-Bello C, Beceiro A. 2017. Activity of the β-lactamase inhibitor LN-1-255 against carbapenem-hydrolyzing class D β-lactamases from Acinetobacter baumannii. Antimicrob Agents Chemother 61:e01172-17. doi: 10.1128/AAC.01172-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Vallejo JA, Martínez-Guitián M, Vázquez-Ucha JC, González-Bello C, Poza M, Buynak JD, Bethel CR, Bonomo RA, Bou G, Beceiro A. 2016. LN-1-255, a penicillanic acid sulfone able to inhibit the class D carbapenemase OXA-48. J Antimicrob Chemother 71:2171–2180. doi: 10.1093/jac/dkw105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Xerri L. 2015. Phase II SBIR: responding to NDM-1–advancement of a new MBL inhibitor to IND. VenatoRx Pharmaceuticals, Inc, Malvern, PA. [Google Scholar]
  • 88.Moya B, Barcelo IM, Bhagwat S, Patel M, Bou G, Papp-Wallace KM, Bonomo RA, Oliver A. 2017. Potent β-lactam enhancer activity of zidebactam and WCK 5153 against Acinetobacter baumannii, including carbapenemase-producing clinical isolates. Antimicrob Agents Chemother 61:e01238-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Moya B, Barcelo IM, Bhagwat S, Patel M, Bou G, Oliver A. 2017. Zidebactam (WCK 5107) and WCK 5153: bicyclo-acyl hydrazide penicillin-bindingprotein (PBP) inhibitors showing potent beta-lactam enhancer activity against multidrug-resistant (MDR) metallo-beta-lactamase (MBL)-producing Klebsiella pneumoniae, abstr P-1300. Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 90.Wockhardt. 2016. MED study to evaluate the safety, tolerability and pharmacokinetics of intravenous WCK 5222 (zidebactam and cefepime) in healthy volunteers. Accession no. NCT02707107. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT02707107.
  • 91.Wockhardt. 2015. Study to evaluate the safety, tolerability, and pharmacokinetics of WCK 5107 alone and in combination with cefepime. Accession no. NCT02532140. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT02532140.
  • 92.Wockhardt. 2016. To investigate the pharmacokinetics of intravenous WCK 5222 (FEP-ZID) in patients with renal impairment. Accession no. NCT02942810. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT02942810.
  • 93.AiCuris Pharmaceuticals. 2017. AiCuris initiates clinical development of AIC499, a novel resistance-breaking antibiotic against a broad range of MDR Gram-negative bacteria. AiCuris Pharmaceuticals, Wuppertal, Germany. [Google Scholar]
  • 94.Joo HY, Kim DI, Kowalik E, Li Y, Mao S, Liu S, Hager MW, Choi WB. 2013. FSI-1671, a novel anti-Acinetobacter carbapenem; in vitro activities of FSI-1671 and FSI-1671/sulbactam against MDR-A. baumannii, abstr F-1202. Abstr 53r Intersci Conf Antimicrob Agents Chemother, 10 to 13 September 2013, Denver, CO. [Google Scholar]
  • 95.Joo HY, Kim DI, Kowalik E, Li Y, Mao S, Liu S, Hager MW, Choi WB. 2013. FSI-1671, a novel anti-Acinetobacter carbapenem; in vivo efficacy against carbapenem-resistance Gram-negative bacterial infection, abstr F-1201 Abstr 53r Intersci Conf Antimicrob Agents Chemother, 10 to 13 September 2013, Denver, CO. [Google Scholar]
  • 96.Zurawski DV, Reinhart AA, Alamneh YA, Pucci MJ, Si Y, Abu-Taleb R, Shearer JP, Demons ST, Tyner SD, Lister T. 2017. SPR741, an antibiotic adjuvant, potentiates the in vitro and in vivo activity of rifampin against clinically relevant extensively drug-resistant Acinetobacter baumannii. Antimicrob Agents Chemother 61:e01239-17. doi: 10.1128/AAC.01239-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Corbett D, Wise A, Langley T, Skinner K, Trimby E, Birchall S, Dorali A, Sandiford S, Williams J, Warn P, Vaara M. 2017. Potentiation of antibiotic activity by a novel cationic peptide: potency and spectrum of activity of SPR741. Antimicrob Agents Chemother 61:e0020-17. doi: 10.1128/AAC.00200-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mendes RE, Rhomberg PR, Becker HK, Davis AP, Lister T, Parr TR, Vaara M, Flamm RK. 2016. Synergistic effect of Gram-positive agents tested in combination with a new polymyxin derivative (SPR741) against multidrug-resistant Gram-negative pathogens, abstr SATURDAY-490 ASM Microbe, 16 to 20 June 2016, Boston, MA. [Google Scholar]
  • 99.Warn P, Teague J, Corbett D, Payne L, Burgess E. 2017. In-vivo efficacy of combinations of novel antimicrobial peptide SPR741 and rifampicin in neutropenic murine thigh infection models of Gram-negative bacterial infection, abstr OS-0563 Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 100.Utley L, Lister T, Coleman S, Eckburg P. 2018. Determination of the pharmacokinetics of single (SAD) and multiple ascending doses (MAD) of SPR741 in healthy volunteers, abstr P-2233 Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 101.Keutzer T, Eckburg P, Farinola N, Walpole S, Utley L, Dwyer J, Kopp E, Coleman S, Tomayko J. 2018. Safety of SPR741, a novel polymyxin potentiator, in healthy adults receiving single- and multiple-dose intravenous administrations, abstr P-2206 Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 102.Rubio-Aparicio D, Nelson K, Roberts KD, Thompson PE, Nation RL, Velkov T, Li J, Hecker SJ, Griffith DC, Dudley MN, Lomovskaya O. 2016. In vitro activity of FADDI-287, a representative of a novel series of polymyxins (PM) with reduced nephrotoxic potential, abstr SATURDAY-495 ASM Microbe, 16 to 20 June 2016, Boston, MA. [Google Scholar]
  • 103.Sabet M, Tarazi Z, Nolan T, Parkinson J, Rubio-Aparicio D, Roberts KD, Thompson PE, Nation RL, Velkov T, Li J, Hecker SJ, Lomovskaya O, Dudley MN, Griffith DC. 2016. Pharmacology of the novel polymyxin FADDI-287 in preclinical models, abstr SATURDAY-499 ASM Microbe, 16 to 20 June 2016, Boston, MA. [Google Scholar]
  • 104.Galani I, Nafplioti K, Chatzikonstantinou M, Giamarellou H, Souli M. 2018. Evaluation of apramycin activity against carbapenem-resistant Enterobacteriaceae and Acinetobacter baumannii, abstr P-0096 Abstr 28th Eur Cong Clin Microbiol Infect Dis, 21 to 24 April 2018, Madrid, Spain. [Google Scholar]
  • 105.Kang AD, Smith KP, Eliopoulos GM, Berg AH, McCoy C, Kirby JE. 2017. In vitro apramycin activity against multidrug-resistant Acinetobacter baumannii and Pseudomonas aeruginosa. Diagn Microbiol Infect Dis 88:188–191. doi: 10.1016/j.diagmicrobio.2017.03.006. [DOI] [PubMed] [Google Scholar]
  • 106.Kang AD, Smith KP, Berg AH, Truelson KA, Eliopoulos GM, McCoy C, Kirby JE. 2018. Efficacy of apramycin against multidrug-resistant Acinetobacter baumannii in the murine neutropenic thigh model. Antimicrob Agents Chemother 16:e02585-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.García-Quintanilla M, Caro-Vega JM, Pulido MR, Moreno-Martínez P, Pachón J, McConnell MJ. 2016. Inhibition of LpxC increases antibiotic susceptibility in Acinetobacter baumannii. Antimicrob Agents Chemother 60:5076–5079. doi: 10.1128/AAC.00407-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Titecat M, Liang X, Lee C-J, Charlet A, Hocquet D, Lambert T, Pagès J-M, Courcol R, Sebbane F, Toone EJ, Zhou P, Lemaitre N. 2016. High susceptibility of MDR and XDR Gram-negative pathogens to biphenyl-diacetylene-based difluoromethyl-allo-threonyl-hydroxamate LpxC inhibitors. J Antimicrob Chemother 71:2874–2882. doi: 10.1093/jac/dkw210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lemaître N, Liang X, Najeeb J, Lee CJ, Titecat M, Leteurtre E, Simonet M, Toone EJ, Zhou P, Sebbane F. 2017. Curative treatment of severe Gram-negative bacterial infections by a new class of antibiotics targeting LpxC. mBio 8:e00674-17. doi: 10.1128/mBio.00674-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Kalinin DV, Holl R. 2017. LpxC inhibitors: a patent review (2010–2016). Expert Opin Ther Pat 27:1227–1250. doi: 10.1080/13543776.2017.1360282. [DOI] [PubMed] [Google Scholar]
  • 111.Flamm RK, Rhomberg PR, Jones RN, Farrell DJ. 2015. In vitro activity of RX-P873 against Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii. Antimicrob Agents Chemother 59:2280–2285. doi: 10.1128/AAC.04840-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cisek AA, Dąbrowska I, Gregorczyk KP, Wyżewski Z. 2017. Phage therapy in bacterial infections treatment: one hundred years after the discovery of bacteriophages. Curr Microbiol 74:277–283. doi: 10.1007/s00284-016-1166-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lin NT, Chiou PY, Chang KC, Chen LK, Lai MJ. 2010. Isolation and characterization of ϕAB2: a novel bacteriophage of Acinetobacter baumannii. Res Microbiol 161:308–314. doi: 10.1016/j.resmic.2010.03.007. [DOI] [PubMed] [Google Scholar]
  • 114.Shen GH, Wang JL, Wen FS, Chang KM, Kuo CF, Lin CH, Luo HR, Hung CH. 2012. Isolation and characterization of φkm18p, a novel lytic phage with therapeutic potential against extensively drug resistant Acinetobacter baumannii. PLoS One 7:e46537. doi: 10.1371/annotation/86d9f6df-7175-467c-a9ff-94eac53af128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hua Y, Luo T, Yang Y, Dong D, Wang R, Wang Y, Xu M, Guo X, Hu F, Ping H. 2018. Phage therapy as a promising new treatment for lung infection caused by carbapenem-resistant Acinetobacter baumannii in mice. Front Microbiol 8:2659. doi: 10.3389/fmicb.2017.02659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Yin S, Huang G, Zhang Y, Jiang B, Yang Z, Dong Z, You B, Yuan Z, Hu F, Zhao Y, Peng Y. 2017. Phage Abp1 rescues human cells and mice from infection by pan-drug resistant Acinetobacter baumannii. Cell Physiol Biochem 44:2337–2345. doi: 10.1159/000486117. [DOI] [PubMed] [Google Scholar]
  • 117.Deng LY, Yang ZC, Gong YL, Huang GT, Yin SP, Jiang B, Peng YZ. 2016. Therapeutic effect of phages on extensively drug-resistant Acinetobacter baumannii-induced sepsis in mice. Zhonghua Shao Shang Za Zhi 32:523–528. (In Chinese.) [DOI] [PubMed] [Google Scholar]
  • 118.Kusradze I, Karumidze N, Rigvava S, Dvalidze T, Katsitadze M, Amiranashvili I, Goderdzishvili M. 2016. Characterization and testing the efficiency of Acinetobacter baumannii phage vB-GEC_Ab-M-G7 as an antibacterial agent. Front Microbiol 7:1590. doi: 10.3389/fmicb.2016.01590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Regeimbal JM, Jacobs AC, Corey BW, Henry MS, Thompson MG, Pavlicek RL, Quinones J, Hannah RM, Ghebremedhin M, Crane NJ, Zurawski DV, Teneza-Mora NC, Biswas B, Hall ER. 2016. Personalized therapeutic cocktail of wild environmental phages rescues mice from Acinetobacter baumannii wound infections. Antimicrob Agents Chemother 60:5806–5816. doi: 10.1128/AAC.02877-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lood R, Winer BY, Pelzek AJ, Diez-Martinez R, Thandar M, Euler CW, Schuch R, Fischetti VA. 2015. Novel phage lysin capable of killing the multidrug-resistant gram-negative bacterium Acinetobacter baumannii in a mouse bacteremia model. Antimicrob Agents Chemother 59:1983–1991. doi: 10.1128/AAC.04641-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Shivaswamy VC, Kalasuramath SB, Sadanand CK, Basavaraju AK, Ginnavaram V, Bille S, Ukken SS, Pushparaj UN. 2015. Ability of bacteriophage in resolving wound infection caused by multidrug-resistant Acinetobacter baumannii in uncontrolled diabetic rats. Microb Drug Resist 21:171–177. doi: 10.1089/mdr.2014.0120. [DOI] [PubMed] [Google Scholar]
  • 122.Schooley RT, Biswas B, Gill JJ, Hernandez-Morales A, Lancaster J, Lessor L, Barr JJ, Reed SL, Rohwer F, Benler S, Segall AM. 2017. Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii infection. Antimicrob Agents Chemother 61:e00954-17. doi: 10.1128/AAC.00954-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.LaVergne S, Hamilton T, Biswas B, Kumaraswamy M, Schooley RT, Wooten D. 2018. Phage therapy for a multi drug resistant Acinetobacter baumannii craniectomy site infection. Open Forum Infect Dis 5:ofy064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Abedon ST, Kuhl SJ, Blasdel BG, Kutter EM. 2011. Phage treatment of human infections. Bacteriophage 1:66–85. doi: 10.4161/bact.1.2.15845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Bochkareva SS, Aleshkin AV, Ershova OG, Novikova LI, Karaulov AV, Kiseleva IA, Zul’karneev ED, Rubal’skiy EO, Zeigarnik MV. 2017. Anti-phage аntibody response in phage therapy against healthcare-associated infections (HAIs). Infekc Bolezni 15:35–40. doi: 10.20953/1729-9225-2017-1-35-40. [DOI] [Google Scholar]
  • 126.Aleshkin AV, Ershova ON, Volozhantsev NV, Svetoch EA, Popova AV, Rubalskii EO, Borzilov AI, Aleshkin VA, Afanas' Ev SS, Karaulov AV, Galimzyanov KM. 2016. Phagebiotics in treatment and prophylaxis of healthcare-associated infections. Bacteriophage 6:40–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Mattila S, Ruotsalainen P, Jalasvuori M. 2015. On-demand isolation of bacteriophages against drug-resistant bacteria for personalized phage therapy. Front Microbiol 6:1271. doi: 10.3389/fmicb.2015.01271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Leitner L, Sybesma W, Chanishvili N, Goderdzishvili M, Chkhotua A, Ujmajuridze A, Schneider MP, Sartori A, Mehnert U, Bachmann LM, Kessler TM. 2017. Bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: a randomized, placebo-controlled, double-blind clinical trial. BMC Urol 17:90. doi: 10.1186/s12894-017-0283-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kessler T. 2017. Bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate. Accession no. NCT03140085. ClinicalTrials.gov, NIH. https://clinicaltrials.gov/ct2/show/NCT03140085.
  • 130.Ho YH, Tseng CC, Wang LS, Chen YT, Ho GJ, Lin TY, Wang LY, Chen LK. 2016. Application of bacteriophage-containing aerosol against nosocomial transmission of carbapenem-resistant Acinetobacter baumannii in an intensive care unit. PLoS One 11:e0168380. doi: 10.1371/journal.pone.0168380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Wang-Lin SX, Balthasar JP. 2018. Pharmacokinetic and pharmacodynamic considerations for the use of monoclonal antibodies in the treatment of bacterial infections. Antibodies 7:5. doi: 10.3390/antib7010005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.McConnell MJ, Pachón J. 2010. Active and passive immunization against Acinetobacter baumannii using an inactivated whole cell vaccine. Vaccine 29:1–5. doi: 10.1016/j.vaccine.2010.10.052. [DOI] [PubMed] [Google Scholar]
  • 133.McConnell MJ, Rumbo C, Bou G, Pachón J. 2011. Outer membrane vesicles as an acellular vaccine against Acinetobacter baumannii. Vaccine 29:5705–5710. doi: 10.1016/j.vaccine.2011.06.001. [DOI] [PubMed] [Google Scholar]
  • 134.Nielsen TB, Pantapalangkoor P, Luna BM, Bruhn KW, Yan J, Dekitani K, Hsieh S, Yeshoua B, Pascual B, Vinogradov E, Hujer KM, Domitrovic TN, Bonomo RA, Russo TA, Lesczcyniecka M, Schneider T, Spellberg B. 2017. Monoclonal antibody protects against Acinetobacter baumannii infection by enhancing bacterial clearance and evading sepsis. J Infect Dis 216:489–501. doi: 10.1093/infdis/jix315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Aridis Pharmaceuticals. 2018. AR-401: fully human monoclonal antibody (mAb) against Acinetobacter baumannii. Aridis Pharmaceuticals, San Jose, CA. [Google Scholar]
  • 136.Tsuji M, Hackel M, Echols R, Yamano Y, Sahm DF. 2017. In vitro activity of cefiderocol against globally collected carbapenem-resistant Gram-negative bacteria isolated from urinary tract source: SIDERO-CR-2014/2016, abstr 1199. IDWeek 2017, 4 to 8 October 2017, San Diego, CA. [Google Scholar]
  • 137.Ito-Horiyama T, Ishii Y, Ito A, Sato T, Nakamura R, Fukuhara N, Tsuji M, Yamano Y, Yamaguchi K, Tateda K. 2016. Stability of novel siderophore cephalosporin S-649266 against clinically relevant carbapenemases. Antimicrob Agents Chemother 60:4384–4386. doi: 10.1128/AAC.03098-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Fyfe C, Leblanc G, Sutcliffe J, Grossman T. 2016. Eravacycline is active against carbapenem-resistant Enterobacteriaceae and Acinetobacter baumannii isolates in the FDA-CDC antimicrobial resistance isolate bank panels, abstr 1844 IDWeek 2016, 26 to 30 October 2017, New Orleans, LA. [Google Scholar]
  • 139.Morrissey I, Bassetti M, Magnet S, Hawser S, Olesky M, Fyfe C. 2017. In vitro activity of eravacycline and comparators against Acinetobacter baumannii, Stenotrophomonas maltophilia and Enterobacteriaceae, including carbapenem- resistant and ESBL phenotype subgroups, collected from European hospitals in 2015, abstr P-1260. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 140.Sutcliffe JA, O'Brien W, Fyfe C, Grossman TH. 2013. Antibacterial activity of eravacycline (TP-434), a novel fluorocycline, against hospital and community pathogens. Antimicrob Agents Chemother 57:5548–5558. doi: 10.1128/AAC.01288-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Fyfe C, Close B, Leblanc G, Newman J. 2017. TP-6076 is active against carbapenem- and polymyxin-resistant Enterobacteriaceae and Acinetobacter baumannii isolates in the FDA-CDC antimicrobial isolate bank panels, abstr P-1365. Abstr 27th Eur Cong Clin Microbiol Infect Dis, 22 to 25 April 2017, Vienna, Austria. [Google Scholar]
  • 142.Barnes MD, Christopher BR, Joseph RD, Van Den Akker F, Bonomo RA. 2017. The novel β-lactamase inhibitor, ETX-2514, in combination with sulbactam effectively inhibits Acinetobacter baumannii, abstr 1204. IDWeek 2017, 4 to 8 October 2017, San Diego, CA. [Google Scholar]

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES