Abstract
Carbapenem-resistant Enterobacterales (CRE) are a growing threat to human health worldwide. CRE often carry multiple resistance genes that limit treatment options and require longer durations of therapy, are more costly to treat, and necessitate therapies with increased toxicities when compared to carbapenem-susceptible strains. Here, we provide an overview of the mechanisms of resistance in CRE, the epidemiology of CRE infections worldwide, and available treatment options for CRE. We review recentlyapproved agents for the treatment of CRE, including ceftazidime-avibactam, meropenem-vaborbactam, imipenem-relebactam, cefiderocol, and novel aminoglycosides and tetracyclines. We also discuss recent advances in phage therapy and antibiotics that are currently in development targeted to CRE. The potential for the development of resistance to these therapies remains high, and enhanced antimicrobial stewardship is imperative both to reduce the spread of CRE worldwide and to ensure continued access to efficacious treatment options.
Keywords: Carbapenem-resistant, Enterobacterales, CRE, Carbapenemase, Antimicrobial resistance
Introduction
The rise of antimicrobial resistant (AMR) organisms worldwide is considered one of the biggest threats to global health by the World Health Organization (WHO)1. Carbapenem resistant Enterobacterales (CRE) are defined by the United States Centers for Disease Control and Prevention (CDC) as Enterobacterales (formerly Enterobacteriaceae) with in vitro resistance to at least one carbapenem.2 Carbapenems are a potent class of broad-spectrum antibiotics that inhibit penicillin binding proteins, thereby preventing cell wall synthesis3 and were once considered the “last resort” antibiotics in many hospitals. Resistance to carbapenems significantly limits the antibiotic armamentarium available to treat challenging infections. CRE have spread substantially in recent years 4–6 and are now endemic in certain regions of North America, Europe and the Mediterranean, and South Asia7.
CRE are typically healthcare-associated infections, although community spread is becoming more common,8–11 with intestinal colonization and environmental sources as reservoirs of infection12. CRE are of particular concern due to the increased mortality13,14, length of hospital stay, and increased cost when compared to drug-susceptible infections15. An economic prediction model from the United States estimated a societal cost of between $59,692 and $86,940 for each CRE infection16. Additionally, CRE infections are often found in the most vulnerable patients--the elderly, those with underlying comorbidities, and those with indwelling catheters or permanent hardware in place4,17–19.
In October 2020, The Infectious Diseases Society of America (IDSA) released guidance for the treatment of multidrug resistant Gram-negative bacterial infections, including CRE, and offers clinicians preferred and alternative treatment strategies for a variety of clinical scenarios20. The IDSA guidance is divided into infections inside and outside the urinary tract and assumes the organism and susceptibility profile are known. This guidance provides a current overview of treatment options for these challenging infections, albeit with a focus on variants that predominate in North America.
This review will focus on treatment strategies for infections with carbapenem-resistant Enterobacterales, including “traditional” antibiotics that have retained activity against CRE, newly approved antibiotics developed specifically for CRE, phage therapy, and antibiotics that are in development to target multi-drug resistant infections.
Mechanisms of carbapenem resistance in CRE
Resistance to carbapenems can be mediated via alterations to the penicillin binding protein of the bacterial cell wall, an increase in efflux pumps, or a decreases in membrane permeability21,22 as well as through the production of carbapenemase enzymes. Carbapenemases are a diverse family of β-lactamases that have the ability to hydrolyze and inactivate a variety of antibiotics including penicillins, cephalosporins, monobactams, and carbapenems23. These enzymes function by binding to the drug, breaking the amide bond of a four-membered azetidinone ring, and preventing it from binding to the penicillin binding protein of the bacterial cell wall24. Carbapenemases are found in approximately 85% of CRE worldwide, with considerable variation between regions, ranging from 76% in Latin America to 90% in the Middle East and Africa found in a recent global survey25. Other studies have shown lower rates, with the recent CRACKLE-2 study finding carbapenemases in 59% of CRE from the United States26. Using the Ambler classification system, carbapenemases are found within class A, B, and D β-lactamases, with substantial geographic heterogeneity in classes between global regions and with various modes of transmission7,27 (Table 1).
Table 1:
Ambler class | Major enzymes | Active site | Primary geographic distribution | Treatment notes |
---|---|---|---|---|
A | KPC | Serine | United States, Colombia, Greece | Inhibited by clavulanate, tazobactam |
NMC, SME | Rare | |||
B | VIM | Zinc | Spain, Italy, Greece | Do not hydrolyze monobactams* |
IMP | Japan, Taiwan | |||
NDM | India, Pakistan, Romania, Poland | |||
D | OXA-48 | Serine | Turkey, Mediterranean, Morocco | Low-level resistance against cephalosporins* |
Are often co-occurring with ESBL enzymes that confer resistance to these classes
Ambler class A carbapenemases use a serine residue to hydrolyze beta-lactams28 and include the blaKPC, blaNMC/blaIMI, and blaSME29 genes, with blaKPC being the most common carbapenemase of the class30. It was first discovered in 1996 in a Klebsiella pneumoniae isolate from North Carolina, USA31, is plasmid-mediated, and is now endemic in much of the western hemisphere with the highest rates found in Eastern North America25,32,33 and outbreaks reported in South America, including Columbia and Ecuador34,35. Spread from the United States has led to outbreaks outside the hemisphere as well. An outbreak in Israel was traced to a strain from New York36 and KPC enzymes have also been found in a variety of European countries including large outbreaks in Greece37,38, Portugal39, and Poland40 among other countries, where they can significantly impact regional resistance patterns24,25. While found primarily in Klebsiella pneumoniae, KPC enzymes have also been found in a variety of other Enterobacterales including Citrobacter freundii, Enterobacter cloacae, Escherichia coli and Serratia marcescens among others, as well as in Pseudomonas species29.
Class B metallo-β-lactamases (MBLs) are zinc-dependent41 and include the blaVIM, blaIMP, and blaNDM genes41–43, all found on mobile genetic elements and capable of horizontal spread.44 MBLs are able to hydrolyze a wide range of beta-lactams but cannot hydrolyze monobactams such as aztreonam.45 The enzyme IMP was the first enzyme discovered in this class, isolated from an imipenem-resistant Pseudomonas aeruginosa isolate in Japan in 199146 and it now accounts for as much as 15% of the CRE found in Japan, Australia and parts of Southeast Asia25,47,48. VIM, for Verona Integron-encoded Metallo-beta-lactamase, was first isolated in Italy in 199749 and is responsible for approximately 15% of the CRE isolated from Europe25, with the highest rates found in Greece, Italy, Spain, and Hungary50. More recently, the New Delhi Metallo-β-lactamases (NDM), were discovered in 2007 from a Klebsiella pneumoniae isolate of a Swedish patient who had previously been hospitalized in India with a urinary tract infection.51 The highest burden of NDM remains in South Asia as well as the Middle East, where it accounts for up to a third of detected carbapenemases25. NDM is of particular concern given its rapid spread and limited treatment options.45
The Class D carbapenemases include members of the OXA-encoding genes and are largely found in Acinetobacter, however the plasmid-encoded blaOXA-48-like genes are found in Enterobacterales52–5455 and have been implicated in multiple nosocomial CRE outbreaks56–59. OXA-48-like enzymes encompass OXA-48 and related variants, including OXA-181, OXA-162, and OXA-232 among others, with distinct geographic distributions and co-occurring resistance genes52,54. OXA-48-like enzymes are most commonly found in the Middle East and Europe, where over 27% of carbapenem-resistant isolates in each region were recently found to harbor OXA-4825, with endemic levels reported in Turkey, Malta, much of North Africa and the Middle East54. OXA-48 remains uncommon in North America, with only 52 cases reported in the United States between 2010–201560 and only found in 1% of carbapenemase-producing CRE in the CRACKLE-2 study26.
Previously approved antibiotics with CRE activity
Therapeutics for CRE are summarized in Table 2. There are several “traditional” antibiotics that have retained activity against some strains of CRE and are being deployed in new ways or in combination with other drugs for the treatment of severe CRE infections.
Table 2:
Agent | Therapeutic Class | Activity against Class A | Activity against Class B | Activity against Class D | Notes |
---|---|---|---|---|---|
Aztreonam | Monobactam | − | + | + | Not recommended. CRE often have co-occurring ESBL enzymes which render aztreonam ineffective. |
Colistin, Polymyxin B | Polymyxin | +/− | +/− | +/− | Limited efficacy, significant toxicities. |
Fosfomycin | Phosphoenolpyruvate analogue | + | + | + | Primarily used for urinary tract infections. |
Tigecycline | Tetracycline | +/− | +/− | +/− | Typically used as combination therapy. |
Ceftazidime-avibactam | β-lactam-β-lactamase inhibitor | + | − | + | Approved for cUTI, cIAI (with metronidazole), HAP/VAP. Can be used with aztreonam for treatment of NDM-producing infections. |
Meropenem-vaborbactam | β-lactam-β-lactamase inhibitor | + | − | − | Approved for cUTI, cIAI, HAP/VAP. |
Imipenem-relebactam | β-lactam-β-lactamase inhibitor | + | − | − | Approved for cUTI, cIAI by FDA. Approved for HAP/VAP, BSI, resistant GN infections by EMA. |
Plazomicin | Aminoglycoside | + | + | + | NDM-carrying CRE often resistant due to 16s ribosomal methyltransferases. Approved for cUTI by FDA. Not approved by EMA. |
Eravacycline | Tetracycline | + | + | + | Approved for cIAI by FDA and EMA. |
Omadacycline | Tetracycline | + | + | + | Oral and IV formulations. Approved by FDA for ABSSSI and CABP. Not approved by EMA. |
Cefiderocol | Cephalosporin | + | + | + | Approved for cUTI and HAP/VAP by FDA. Approved for resistant GN infections by EMA. CREDIBLE-CR study showed increased all-cause mortality. |
Phage therapy | N/A | + | + | + | Few clinical trials showing efficacy for CRE at this time. Require specificity for infecting organism, often leading to significant lag time to start treatment. |
Zidebactam* | β-lactamase inhibitor | + | +/− | + | Combined with cefepime. Clinical trials pending. |
Taniborbactam* | β-lactamase inhibitor | + | + | + | Combined with cefepime. Currently in phase 3 trials for cUTI. |
LYS228* | Monobactam | + | + | +/− | No clinical trials currently underway |
Nacubactam* | β-lactamase inhibitor | + | +/− | + | Combined with meropenem. Completed phase 1 clinical trials. |
cUTI=complicated urinary tract infection; cIAI=complicated intraabdominal infection; HAP/VAP=hospital acquired pneumonia/ventilator-associated pneumonia; GN=gram negative; ABSSSI=acute bacterial skin and skin structure infection; CABP=community acquired bacterial pneumonia; FDA= United States Food and Drug Administration; EMA= European Medicines Agency.
antibiotic currently in development
Aztreonam
The monobactam antibiotic Aztreonam is effective against bacteria producing Class B and D carbapenemases in isolation, however these bacteria often carry concomitant ESBL genes that hydrolyze aztreonam rendering it ineffective and thus it is often of limited clinical utility as monotherapy61,62. The combination of aztreonam with the novel β-lactam-β-lactamase inhibitor ceftazidime-avibactam is a promising treatment option for MBLs and is discussed in detail below. Notably, aztreonam does not have activity against bacteria producing Class A carbapenemases, including bacteria producing the highly prevalent KPC carbapenemases61.
Polymyxins
The polymyxin antibiotics colistin and polymyxin B have long been used for resistant Gram-negative bacteria, including CRE63, however there is emerging resistance developing to these drugs. This is notable, as several studies have shown an association between polymyxin resistance and an increase in mortality64,65, although these studies occurred prior to the development of newer CRE-active agents which are now available. Resistance to polymyxins can occur via chromosomal point mutations leading to changes in the bacterial lipopolysaccharide membrane or an increase in efflux pumps, or it can be plasmid-mediated, via several mcr genes that change lipid A present in the lipopolysaccharide membrane and prevent the target drug from binding. 66. There is also evidence that heteroresistance arising from minor resistant subpopulations in a culture may make colistin resistance difficult to detect in vitro and lead to subsequent treatment failure67,68. Additionally, polymyxins have significant nephrotoxicity, with several studies having shown their inferiority compared to newer drugs against isolates carrying Class A carbapenemases69–71, and as such they are not currently recommended for the treatment of CRE by the IDSA20. Despite this, they are often the only available antibiotic for CRE infections in certain regions despite increasing resistance levels72,73, and thus are considered to be a “highest priority” critically important antimicrobial by the WHO74,75.
Fosfomycin
Fosfomycin, an antibiotic first discovered in 1969, inhibits cell wall synthesis in a variety of Gram-positive and Gram-negative bacteria76, including Enterobacterales, and has retained activity against some CRE isolates77. Resistance to fosfomycin is mediated primarily through the fosA genes which encode fosfomycin hydrolases and are found in many Enterobacterales with the exception of E. coli78,79. Traditionally, fosfomycin has primarily been used as an oral formulation for lower urinary tract infections76,8080, however there is growing interest in intravenous use for MDR organisms, including CRE81–83. Fosfomycin does not have sufficient renal parenchymal penetration, and thus should not be used for upper urinary tract infections84,85.
Tigecycline
The tetracycline antibiotic tigecycline has a broad spectrum of activity against gram positive and gram negative infections and global surveillance data from the TEST study shows that the majority of Enterobacterales isolates collected worldwide between 2014–2016 remain susceptible (≤1.3% resistance in all regions)86. Tigecycline has been used for CRE infections with success, however several recent studies have shown monotherapy to be of limited benefit87 and combination therapy is likely more efficacious88,89. Resistance to tigecycline in Enterobacterales can arise from upregulation of the AcrAB efflux pump90,91 or via the plasmid-mediated tet(X4) gene, which encodes a flavin-dependent monooxygenase that modifies tigecycline92,93. Using “traditional” CRE-active antibiotics in combination with antibiotics with other mechanisms of action or with “repurposed” drugs from other classes has also shown some promise for the treatment of CRE infections94. For example, there are in vitro studies showing synergistic effects of combining colistin with other antibiotics including clarithromycin or rifamycin95 or the HIV drug azidothymidine (AZT)96 for the treatment of CRE that are colistin resistant. Other combinations that have shown in vitro activity against CRE include AZT and tigecycline97, pentamidine in combination with rifampicin, tobramycin, tigecycline or amikacin98 and polymyxin B with citalopram, sertraline, or spironolactone99. Animal studies and clinical trials are needed to determine in vivo efficacy of these combination treatments in true clinical infections and thus the utility of these combination regimens remain theoretical at this time.
β-lactam-β-lactamase inhibitor combinations
Ceftazidime-avibactam
In the last several years, β-lactam-β-lactamase inhibitor combinations have been developed and approved specifically to target multidrug resistant organisms, including CRE. The first of these, avibactam, was developed in 2011 and is a synthetic diazabicyclooctane (DBO) non-β-lactam that covalently and reversibly binds to serine β-lactamases and has activity against class A (KPC)100,101 and class D (OXA-48-like)102,103100,101 carbapenemases, but not MBLs (NDM, VIM, IMP). When compared to polymyxin antibiotics, multiple observational studies have shown ceftazidime/avibactam to be superior for the treatment of CRE infections possessing Class A carbapenemases with fewer side effects and toxicities71,104–106. Ceftazidime-avibactam was approved by the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA) in 2015 for complicated urinary tract infections (cUTI) and for complicated intra-abdominal infections (cIAI) in combination with metronidazole107. Approval was granted following the RECLAIM108 trials, which showed non-inferiority for ceftazidime/avibactam when compared to meropenem for cIAI and the RECAPTURE trial, which showed non-inferiority compared to doripenem for cUTI109. Approval has since been expanded to include hospital-acquired and ventilator-associated pneumonia following the REPROVE trial, a phase-III trial conducted across 23 countries which showed non-inferiority of ceftazidime-avibactam compared to meropenem for nosocomial pneumonia110. It is important to note all three of these studies leading to approval for ceftazidime-avibactam used clinical inclusion criteria and did not select specifically for CRE. Microbiological analysis showed that 13.5% of patients in the RECLAIM trials, 19.6% of patients in the RECAPTURE trial, and 28% in the REPROVE trial had a ceftazidime-resistant organism at baseline. Only the RECLAIM trials reported the rate of MBL infection, at approximately 3%108.
In isolates from hospitalized patients collected worldwide during the INFORM global surveillance survey for AMR resistance, in vitro susceptibility to ceftazidime-avibactam has remained high for CRE; among 816 non-MBL CRE isolates collected between 2012–2014, only 19 (2.3%) were resistant and 97.7% were susceptible to ceftazidime-avibactam111. Subsequent testing of isolates collected between 2015–2017 showed a similarly high rate of 99.8% susceptibility for ceftazidime-avibactam112.
Although overall high rates of susceptibility to ceftazidime-avibactam remain, a number of mutations have been seen clinically that confer resistance, primarily in carriers of KPC-2 and KPC-3 enzymes. The sequence type 258 Klebsiella pneumoniae with KPC-3 has been shown to be resistant to ceftazidime-avibactam due to transposition of KPC-3 onto a second plasmid with subsequent alterations in the porin channels OmpK35 and OmpK36 and upregulation of efflux pumps113–115. Concerningly, mutations in blaKPC-3 conferring resistance to avibactam have been reported in patients while on ceftazidime-avibactam therapy via single amino acid substitutions at D179Y/T243M, D179Y, and V240G leading to alterations in the Ω-loop in KPC-3, however these mutations restore meropenem susceptibility in some isolates116. More recently, a KPC-3 variant named KPC-50 was recovered from a Klebsiella pneumoniae isolate in a Swedish patient and found to contain a three-amino-acid insertion that conferred increased affinity to ceftazidime and decreased activity of avibactam leading to resistance117. KPC-2 variants arising from single amino acid substitutions at the Ω-loop have also been found to confer resistance to ceftazidime-avibactam, likely through increased affinity of the enzyme for ceftazidime, thereby preventing the binding of avibactam118. While most of the above resistance mechanisms have been documented in Klebsiella pneumoniae isolates, point mutations leading to insertion of TIPY in penicillin binding protein 3 of an E. coli isolate containing KPC-3 have been documented, which prevents the binding of ceftazidime and cannot be overcome by avibactam119,120.
Although ceftazidime-avibactam alone does not have activity against MBLs, there is significant in vitro synergy between ceftazidime-avibactam and aztreonam that confers activity against these isolates121. This is of particular importance, given that although aztreonam is active against class B carbapenemases, it is often hydrolyzed by other β-lactamases that co-occur with MBLs122. As a result, only 29.2% of MBLs from a recent global survey were found to retain susceptibility to aztreonam monotherapy123. When tested against the combination of aztreonam-avibactam, all MBL isolates in that study were inhibited by the combination123. A clinical case series evaluating this combination treatment in 10 patients with infections caused by NDM-producing MBLs during an outbreak found 6 of 10 patients had clinical success at 30 days, suggesting the combination of ceftazidime-avibactam plus aztreonam may be a useful clinical option for extensively drug resistant Enterobacterales infections that contain both class B carbapenemases as well as ESBL enzymes124. Additional reports of combined ceftazidime-avibactam plus aztreonam treatment have replicated these early findings125, including in pan-resistant isolates126. Given that the combination of aztreonam plus avibactam alone, without the addition of ceftazidime, appears efficacious, this two-drug combination is currently being tested in a Phase III clinical trial for the treatment of complicated infections caused by MBL-containing gram negative bacteria127. An earlier Phase II pharmacokinetic trial (the REJUVENATE study) showed the combination of aztreonam-avibactam to have similar safety and tolerability to aztreonam monotherapy128. As a result of these findings, the IDSA currently recommends ceftazidime-avibactam alone as the preferred treatment for OXA-48-producing CRE outside the urinary tract and in combination with aztreonam for NDM-producing CRE infections20.
Meropenem-vaborbactam
Meropenem-vaborbactam was approved by the FDA in 2017 for the treatment of complicated urinary tract infections (cUTI)129 and by the EMA in 2018 with an expanded authorization that includes cUTI, cIAI, and hospital acquired or ventilator-associated pneumonia (HAP/VAP)130. Meropenem-vaborbactam was designed to target multidrug-resistant organisms, and specifically the class A KPC carbapenemases131. The drug combines the carbapenem antibiotic meropenem with a novel β-lactamase inhibitor containing a cyclic boronic acid pharmacophore that restores the activity of meropenem against serine carbapenemases132. While it has broad activity against class A carbapenemases (as well as class C β-lactamases conferring cephalosporin resistance), it notably does not have activity against the class B metallo β-lactamases (NDM, VIM, IMP) nor class D (OXA-48-like) carbapenemases133. A survey of meropenem-vaborbactam susceptibility against globally-collected CRE showed the lowest MIC values for isolates from the Americas, consistent with the predominance of KPC-producers in this region134. Given this, meropenem-vaborbactam may be of more limited utility in regions where MBLs and OXA-48-like enzymes predominate, including parts of Asia, the Middle East, and North Africa.
Approval for meropenem-vaborbactam was obtained following the TANGO I trial which showed non-inferiority of meropenem-vaborbactam for cUTI when compared to piperacillin-tazobactam135. TANGO I did not select for patients with CRE organisms and in fact, nearly all baseline uropathogens were susceptible to meropenem. This was later followed by the TANGO II trial to test meropenem-vaborbactam in complicated CRE infections including bloodstream infections (BSI), pyelonephritis, VAP, and cIAI 70. While a descriptive study, TANGO II evaluated 47 patients across 8 countries and found an increase in clinical and microbiologic cure and reduction in death with fewer adverse events compared to best alternative therapy. Vaborbactam enters cells via the membrane porin channels OmpK35 and OmpK36133 and resistance to vaborbactam can develop via downregulation or alteration of these porin channels136–138136,137
Imipenem-relebactam
The most recent drug combination in this class is imipenem-relebactam, a non-β-lactam bicyclic DBO β-lactamase inhibitor that is structurally similar to avibactam, but with the addition of a piperidine ring139. It is believed to reversibly acylate β-lactamases140. Imipenem-relebactam is active against class A carbapenemases but not the metallo-β-lactamases and has little to no activity against the class D OXA-48-like enzymes141. Information from the SMART surveillance study on Enterobacterales isolates collected in Europe showed the addition of relebactam restored imipenem susceptibility in 67% of isolates carrying KPC enzymes, but that nearly all isolates with MBLs or OXA-48-like enzymes remained nonsusceptible, primarily occurring in isolates from countries with endemic levels of these enzymes142. This highlights the importance of determining the underlying mechanism of carbapenem resistance and carbapenemase epidemiology when selecting treatment options.
Imipenem-relebactam was approved for use by the US FDA in 2019143 and is available with the carbapenem imipenem/cilastatin for clinical use144. The RESTORE-IMI-1 trial evaluating the safety and efficacy of imipenem-relebactam in a variety of severe imipenem-resistant gram negative infections found higher favorable clinical response rate (71.4% vs 40%), lower 28-day mortality rates (9.5% vs 30%), and lower treatment-associated nephrotoxicity (10.3% vs 56.3%) with imipenem-relebactam compared to imipenem plus colistin69. Notably, most of the isolates in this study were Pseudomonas spp. (77.4%) with the remainder Enterobacterales. The RESTORE-IMI-II trial was a non-inferiority study of imipenem-relebactam compared to piperacillin-tazobactam for HAP/VAP infection and found imipenem-relebactam was non-inferior for both 28-day mortality and favorable clinical response145. When looking specifically at the microbiologic modified intent-to-treat population, mortality rates for intubated patients with HAP/VAP were 12.2% lower for those in the imipenem-relebactam group compared to the piperacillin-tazobactam group. Given the potential for resistance with all of the β-lactam-β-lactamase inhibitor combinations, enhanced antibiotic stewardship will be crucial to ensuring ongoing efficacy of these agents146.
Novel aminoglycosides
Plazomicin
Plazomicin is a novel semisynthetic aminoglycoside that was derived from the antibiotic sisomicin, a naturally occurring aminoglycoside discovered in 1970, and works by binding to the 30s subunit of bacterial ribosomes, inhibiting protein synthesis147. Plazomicin has a broad spectrum of activity against Enterobacterales, including those with ESBL enzymes and multiple classes of CRE, including class A (KPC), class B (VIM, IMP), and class D (OXA-48)148–150. It has shown variable activity against the metallo-beta lactamase NDM-1, largely because NDM-1 often co-produces 16s ribosomal methyltransferases, which modify the 30s ribosomal subunit and prevent aminoglycoside binding148. Given this, it may be of limited clinical utility in regions where NDM-1 are endemic.
Plazomicin was approved by the US FDA in 2018 for cUTI151 following a non-inferiority trial comparing plazomicin to meropenem for cUTI including pyelonephritis caused by Enterobacterales152. This was later followed by the CARE trial, comparing plazomicin to colistin in combination with adjunctive meropenem or tigecycline in patients with CRE-causing BSI or VAP and found a 26% reduction in death or clinically-significant disease-related complications at 28 days in those who received plazomicin, and with fewer adverse events153. The trial was small, however, and the drug was therefore not granted expanded approval for use in BSI154. Plazomicin has not been approved by the European Medicines Agency and the application for approval has since been withdrawn due to financial reasons155, following the parent manufacturer of plazomicin declaring bankruptcy156.
Resistance to aminoglycosides most often occurs via aminoglycoside modifying enzymes (AMEs) that reduce the binding affinity for the ribosomal target157. Plazomicin has several structural modifications that prevent the activity of most AMEs, thereby reducing the risk of AME-mediated resistance158. As noted above, plazomicin cannot overcome modifications caused by 16s ribosomal methyltransferases and bacteria that possess these enzymes are resistant to plazomicin, a concerning finding given that these genes can be transferred horizontally via plasmids159.
Tetracyclines
Eravacycline
Eravacycline is a fully-synthetic tetracycline developed in 2011160 that is structurally similar to tigecycline and inhibits bacterial protein synthesis by binding to the ribosomal 30s subunit resulting in broad gram positive and gram negative activity against both aerobic and anaerobic organisms, with the exception of Pseudomonas161. Eravacycline has activity against CRE including Class A (KPC), class B (VIM, NDM-1) and class D (OXA-48) enzymes 162,163 with consistently lower MICs than for tigecycline162–164. While it has reasonably high oral bioavailability, only IV formulations are available currently.
A pooled analysis of two phase III trials evaluating eravacycline for cIAI showed non-inferiority compared to ertapenem and meropenem, although with higher levels of nausea, vomiting, and diarrhea compared to the carbapenems165. The results of these studies led to approval for the drug in 2018 by both the EMA and the US FDA for use in cIAI166,167. While initially promising as a potential option for urinary tract infections given in vitro activity against biofilms of uropathogenic E. coli168, a phase 3 trial comparing eravacycline to levofloxacin for cUTI failed to show noninferiority and thus it was not approved for this indication166,169.
Resistance to tetracycline antibiotics most often occurs via active drug efflux pumps encoded via tet genes, and ribosomal protection proteins170. Eravacycline evades these resistance mechanisms via a modified D ring side chain that maintains the drug’s efficacy160,171. Notably, the enzyme Tet(X) is a tetracycline destructase that enzymatically inactivates tetracyclines and is active against eravacycline172. This enzyme can be located on mobile genetic elements and has been shown to confer resistance to eravacycline. It has been found in various organisms, including E. coli, and can be found as asymptomatic carriage in human gut flora92,173, indicating the potential for spread of eravacycline resistance.
Omadacycline
Omadacycline is a semisynthetic tetracycline that most closely resembles tigecycline but with an aminomethyl group at the C9 position174. Similar to eravacycline, this substitution results in broad gram positive and gram negative activity and resistance to the activity of the tet efflux pumps and ribosomal protection proteins174,175. Two phase-3 trials showed IV omadacycline to be noninferior to IV linezolid and IV moxifloxacin for acute bacterial skin and skin structure infection (ABSSSI) and community acquired bacterial pneumonia (CABP), respectively176,177. Subsequently, the OASIS-2 trial showed noninferiority of oral omadacycline to oral linezolid for ABSSTI178. Approval was obtained from the FDA in 2018 for both oral and IV formulations for ABSSSI and CABP179. Approval was sought from the EMA for the same, however the agency requested additional studies for an indication for CABP and the manufacturer of omadacycline subsequently withdrew the application for financial reasons180.
As with eravacycline, omadacycline is deactivated by the Tet(X) destructase enzyme172. A recent study of NDM-producing Enterobacterales from the United States found that 59.6% were susceptible to omadacycline, indicating this may be a possible oral treatment option for selected patients infected with CRE181.
Cephalosporins
Cefiderocol
Cefiderocol is a novel siderophore cephalosporin that acts through a “trojan horse” mechanism that uses the bacterial iron transport system to facilitate antibiotic uptake and evade bacterial defense systems182. Once inside the bacterium, cefiderocol has high affinity for several penicillin binding proteins, inhibiting peptidoglycan synthesis and ultimately causing cell death183. Modifications in the C3 and C7 side chains of cefiderocol render it highly stable against a variety of β-lactamases, including carbapenemases184,185. Cefiderocol has a similar safety profile to other cephalosporins, with the most common adverse reactions being gastrointestinal disturbance, rash, and fever186.
In vitro studies show activity of cefiderocol against a variety of CRE, including those harboring Class A (KPC), Class B (NDM, VIM, IMP), and Class D (OXA-48-like) enzymes184,187,188. Cefiderocol was approved by the FDA in 2019189 for cUTI and HAP/VAP following a phase 2 non-inferiority trial comparing cefiderocol to imipenem-cilastatin for treatment of cUTI caused by gram negative uropathogens190 and a phase 3 non-inferiority trial comparing cefiderocol to meropenem for gram negative nosocomial pneumonia191. The EMA authorization is broader, and includes gram negative aerobic infections in patients with limited treatment options192. The CREDIBLE-CR study was subsequently undertaken to evaluate cefiderocol in serious carbapenem-resistant infections193. It found that cefiderocol had comparable clinical and microbiologic effectiveness when compared to the best alternative therapy, however there was an increase in all-cause mortality in the cefiderocol group in those treated for BSI, nosocomial pneumonia, and sepsis193. This increase was not seen for cUTI and appeared to be driven largely by Acinetobacter infections. The clinical efficacy of cefiderocol against CRE remains to be determined in practice and the FDA approval now includes a warning for increased all-cause mortality as a result of the trial189.
There is some evidence of emerging resistance to cefiderocol, however it remains rare194–196. In vitro studies suggest that cefiderocol resistance among Enterobacterales is likely due to the co-production of both serine and metallo-beta lactamases, and may be able to be overcome with the addition of avibactam197.
Phage Therapy
As bacteria become increasingly resistant to chemical antibiotics through mutations and horizontal gene transfer, an area that is gaining increasing attention and promise as a therapeutic option for multidrug resistant organisms is phage therapy. Phage therapy is derived from naturally occurring bacteriophages that use lytic viruses to infect and ultimately lyse bacteria198. Phages attach to receptors on the surface of target bacteria and deliver viral genomic material into the bacterial cell. The bacteria then use that genetic material to produce viral copies and package new viral particles which then escape the bacterium via cell lysis. This kills the infected bacterial cell and releases new phage particles to infect other susceptible bacteria, making the process potentially self-amplifying198, although in clinical practice repeated ongoing dosing is likely required199.
The use of bacteriophages to treat human infections was first pioneered at the turn of the 20th century and used successfully in several human infections including cholera, plague, and conjunctivitis, however their use was limited and phages soon fell out of favor with the advent of chemical antibiotics in the mid-twentieth century200. Phages have several advantages over antibiotics, including specificity for the infecting organism, self-amplification, self-destruction when the bacterial infection is cleared, ability to penetrate biofilms, and preservation of the commensal human microbiota201. However, phages may induce inflammatory immune response202 and antiviral immunity203 in humans. The requirement for strain-specific phages may also limit the timely administration and scaling of phage therapy. As antibiotic resistance has increased at an alarming rate, there has been a renewed interest in phage therapy for treatment of multidrug resistant infections.
While use of phage therapy continued in the twentieth century in Georgia, Poland, and Russia200,204, the first randomized controlled phase I/II trial that met guidelines of good manufacturing practice for phage therapy was the PhagoBurn trial205. It was conducted between 2015 and 2017 and enrolled 27 individuals with burn wounds infected with Pseudomonas aeruginosa to receive topical therapy with a lytic phage cocktail or standard dressings205. The study showed a slower decrease in bacterial burden with phage therapy compared to standard of care, but the study authors note that a low concentration of phage was used. Since then, several case reports have shown efficacy of phages for treating multidrug resistant infections. A case series of 10 patients with highly resistant infections from a single center in the United States showed success with phage therapy in 7 of 10 cases, failure in 2, and uninterpretable results in 1 with few adverse effects206. These infections were primarily MDR Acinetobacter, Pseudomonas, and S. aureus, with one case of a persistent ESBL E. coli infection.
Although clinical trials of phage therapy specifically for CRE treatment are lacking, there is promising data from in vitro studies. Phages have recently been discovered that show in vitro activity against MDR E. coli isolates207, carbapenem-resistant Citrobacter freundii208, and there have been several phages discovered with activity against various strains of carbapenem-resistant Klebsiella isolates209–212. Additional studies in mouse models show success using phages to treat CRE Klebsiella infections213. These provide promising options for future studies targeting infections caused by CRE, where few antibiotic options remain or where toxicities preclude their use198.
As with antibiotics, phages are not immune to the development of bacterial resistance. A variety of resistance mechanisms have been described, including blocking phage attachment and adsorption, cutting phage DNA via the CRISPR system, and mechanisms to block phage transcription, translation, and cell lysis214. Combining phages with traditional antibiotics has proven efficacious in some cases206,215 as a way to overcome these challenges.
Future directions: Antibiotics in the pipeline
The World Health Organization has identified CRE as a critical priority pathogen for prioritizing new drug development216 and there are several drugs currently undergoing clinical trials that are promising candidates for increasing the armamentarium against CRE. Zidebactam (WCK 5222) is a DBO that functions as both a direct antibacterial and a beta lactamase inhibitor that, when combined with cefepime, has activity against KPC, OXA-48, and several class B carbapenemases217,218. Isolates of Enterobacterales, Acinetobacter spp., and Pseudomonas spp. collected worldwide showed high levels of susceptibility to the zidebactam/cefepime combination219, making this a promising drug for clinical trials. Phase 1 pharmacokinetic studies have shown high plasma concentrations as well as good pulmonary penetration of the drug220 and it is well-tolerated in individuals with renal impairment, although it requires dose-adjustment221.
Taniborbactam (VNRX-5133) is a boronic-acid-containing pan-spectrum β-lactamase inhibitor that restores the activity of beta-lactam antibiotics against ESBL and CRE and is considered the first pan-spectrum β-lactamase inhibitor in clinical development222. The boronic acids and esters bind to the active-site serine residue of enzymes, including β-lactamases, thereby inhibiting their function, and bicyclic boronates are able to inactivate serine- and metallo-beta lactamases223. When combined with the β-lactam drug cefepime, taniborbactam restored in vitro activity against all Enterobacterales tested, including CRE with class A, B, and D enzymes, as well as ESBL-Enterobacterales containing class C enzymes224. Studies in animal models showed high in vitro activity of cefepime/taniborbactam against Enterobacterales225 and there is currently a phase 3 trial underway testing cefepime/taniborbactam for cUTI (clinical trials.gov NCT03840148).
LYS228 is a monobactam antibiotic, similar to aztreonam, that retains activity against metallo-β-lactamases but with structural changes that also provide activity against the serine β-lactamases226 by targeting penicillin binding protein 3. In vitro studies have shown potent activity against Class A (KPC) and Class B (NDM) carbapenemases227,228. Pharmacokinetic studies showed good safety and tolerability229. Two phase 2 trials of LYS228 were underway when Novartis, the parent company that developed LYS228, licensed the drug to Boston Pharmaceuticals for further development230. The proposed clinical trials were halted and as of publication there are no additional trials for LYS228/BOS228 yet registered with clinicaltrials.gov.
Nacubactam is a bridged diazabicyclooctane β-lactamase inhibitor that inactivates class A and class C β-lactamases and functions both as an independent antibiotic as well as providing “enhancement” when combined with β-lactam antibiotics with potent activity against Enterobacterales231. When combined with meropenem, nacubactam has shown strong in vitro activity against class A and class D carbapenemases as well as class C ESBL enzymes232 and has shown some activity against the metallo-β-lactamases233. Phase 1 pharmacokinetic trials showed it to be well-tolerated without significant adverse reactions234.
Conclusion
The spread of carbapenem-resistant Enterobacterales is an urgent public health issue and represents a threat to antibiotic efficacy worldwide. There are several treatment classes currently available to clinicians to treat these infections including “traditional” antibiotics that have retained anti-CRE-activity, novel β-lactam-β-lactamase inhibitor combinations that have come on the market in the last decade, and novel aminoglycosides, tetracyclines, and cephalosporins. Local resistance patterns and the regional prevalence of specific carbapenemase enzymes are important to consider when selecting therapy, as not all agents have activity against all classes of enzymes. Phage therapy represents a promising alternative therapy for highly drug-resistant infections, however the applicability of this technology to a broad range of clinical scenarios remains to be seen. With all these treatments, enhanced antimicrobial stewardship will be paramount to ensuring the continued efficacy of these therapies for years to come.
References:
- 1.The World Health Organization. Antibiotic resistance. https://www.who.int/news-room/fact-sheets/detail/antibiotic-resistance.
- 2.United States Centers for Disease Control and Prevention. Clinicians: Information about CRE. https://www.cdc.gov/hai/organisms/cre/cre-clinicians.html (2019).
- 3.Papp-Wallace KM, Endimiani A, Taracila MA & Bonomo RA Carbapenems: past, present, and future. Antimicrob. Agents Chemother. 55, 4943–4960 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.van Duin D & Doi Y The global epidemiology of carbapenemase-producing Enterobacteriaceae. Virulence 8, 460–469 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Nordmann P & Poirel L The difficult-to-control spread of carbapenemase producers among Enterobacteriaceae worldwide. Clin. Microbiol. Infect. 20, 821–830 (2014). [DOI] [PubMed] [Google Scholar]
- 6.Nordmann P, Naas T & Poirel L Global spread of Carbapenemase-producing Enterobacteriaceae. Emerg. Infect. Dis. 17, 1791–1798 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Bonomo RA et al. Carbapenemase-Producing Organisms: A Global Scourge. Clin. Infect. Dis. 66, 1290–1297 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kelly AM, Mathema B & Larson EL Carbapenem-resistant Enterobacteriaceae in the community: a scoping review. Int. J. Antimicrob. Agents 50, 127–134 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Barbadoro P et al. Carriage of Carbapenem-Resistant Enterobacterales in Adult Patients Admitted to a University Hospital in Italy. Antibiotics (Basel) 10, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hu H et al. Clinical and Microbiological Characteristics of Community-Onset Carbapenem-Resistant Enterobacteriaceae Isolates. Infect. Drug Resist. 13, 3131–3143 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.van Duin D & Paterson DL Multidrug-Resistant Bacteria in the Community: An Update. Infect. Dis. Clin. North Am. 34, 709–722 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Taggar G, Attiq Rheman M, Boerlin P & Diarra MS Molecular Epidemiology of Carbapenemases in Enterobacteriales from Humans, Animals, Food and the Environment. Antibiotics (Basel) 9, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Martin A, Fahrbach K, Zhao Q & Lodise T Association Between Carbapenem Resistance and Mortality Among Adult, Hospitalized Patients With Serious Infections Due to Enterobacteriaceae: Results of a Systematic Literature Review and Meta-analysis. Open Forum Infect Dis 5, ofy150 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kohler PP et al. Carbapenem Resistance, Initial Antibiotic Therapy, and Mortality in Klebsiella pneumoniae Bacteremia: A Systematic Review and Meta-Analysis. Infect. Control Hosp. Epidemiol. 38, 1319–1328 (2017). [DOI] [PubMed] [Google Scholar]
- 15.Zilberberg MD, Nathanson BH, Sulham K, Fan W & Shorr AF Carbapenem resistance, inappropriate empiric treatment and outcomes among patients hospitalized with Enterobacteriaceae urinary tract infection, pneumonia and sepsis. BMC Infect. Dis. 17, 279 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bartsch SM et al. Potential economic burden of carbapenem-resistant Enterobacteriaceae (CRE) in the United States. Clin. Microbiol. Infect. 23, 48.e9–48.e16 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Igbinosa O, Dogho P & Osadiaye N Carbapenem-resistant Enterobacteriaceae: A retrospective review of treatment and outcomes in a long-term acute care hospital. Am. J. Infect. Control 48, 7–12 (2020). [DOI] [PubMed] [Google Scholar]
- 18.Adar A et al. Clinical and Demographic Characteristics of Patients With a New Diagnosis of Carriage or Clinical Infection With Carbapenemase-Producing Enterobacterales: A Retrospective Study. Front Public Health 9, 616793 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Voor In ‘t Holt AF, Severin JA, Lesaffre EMEH & Vos MC A systematic review and meta-analyses show that carbapenem use and medical devices are the leading risk factors for carbapenem-resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 58, 2626–2637 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tamma PD et al. Infectious Diseases Society of America Guidance on the Treatment of Extended-Spectrum β-lactamase Producing Enterobacterales (ESBL-E), Carbapenem-Resistant Enterobacterales (CRE), and Pseudomonas aeruginosa with Difficult-to-Treat Resistance (DTR-P. aeruginosa). Clin. Infect. Dis. (2020) doi: 10.1093/cid/ciaa1478. [DOI] [PubMed] [Google Scholar]
- 21.Ruppé É, Woerther P-L & Barbier F Mechanisms of antimicrobial resistance in Gram-negative bacilli. Ann. Intensive Care 5, 61 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nordmann P & Poirel L Epidemiology and Diagnostics of Carbapenem Resistance in Gram-negative Bacteria. Clin. Infect. Dis. 69, S521–S528 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Queenan AM & Bush K Carbapenemases: the versatile beta-lactamases. Clin. Microbiol. Rev. 20, 440–58, table of contents (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bush K & Bradford PA Epidemiology of β-Lactamase-Producing Pathogens. Clin. Microbiol. Rev. 33, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kazmierczak KM, Karlowsky JA, de Jonge BLM, Stone GG & Sahm DF Epidemiology of Carbapenem Resistance Determinants Identified in Meropenem-nonsusceptible Enterobacterales collected as part of a Global Surveillance Program, 2012–2017. Antimicrob. Agents Chemother. (2021) doi: 10.1128/AAC.02000-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.van Duin D et al. Molecular and clinical epidemiology of carbapenem-resistant Enterobacterales in the USA (CRACKLE-2): a prospective cohort study. Lancet Infect. Dis. 20, 731–741 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Nordmann P & Poirel L Emerging carbapenemases in Gram-negative aerobes. Clin. Microbiol. Infect. 8, 321–331 (2002). [DOI] [PubMed] [Google Scholar]
- 28.Palzkill T Structural and Mechanistic Basis for Extended-Spectrum Drug-Resistance Mutations in Altering the Specificity of TEM, CTX-M, and KPC β-lactamases. Front Mol Biosci 5, 16 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Walther-Rasmussen J & Høiby N Class A carbapenemases. J. Antimicrob. Chemother. 60, 470–482 (2007). [DOI] [PubMed] [Google Scholar]
- 30.Hossain A et al. Plasmid-mediated carbapenem-hydrolyzing enzyme KPC-2 in an Enterobacter sp. Antimicrob. Agents Chemother. 48, 4438–4440 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Yigit H et al. Novel carbapenem-hydrolyzing beta-lactamase, KPC-1, from a carbapenem-resistant strain of Klebsiella pneumoniae. Antimicrob. Agents Chemother. 45, 1151–1161 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Deshpande LM, Jones RN, Fritsche TR & Sader HS Occurrence and Characterization of Carbapenemase-Producing Enterobacteriaceae: Report from the SENTRY Antimicrobial Surveillance Program (2000–2004). Microb. Drug Resist. 12, 223–230 (2006). [DOI] [PubMed] [Google Scholar]
- 33.Castanheira M, Farrell SE, Deshpande LM, Mendes RE & Jones RN Prevalence of β-lactamase-encoding genes among Enterobacteriaceae bacteremia isolates collected in 26 U.S. hospitals: report from the SENTRY Antimicrobial Surveillance Program (2010). Antimicrob. Agents Chemother. 57, 3012–3020 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Villegas MV et al. First detection of the plasmid-mediated class A carbapenemase KPC-2 in clinical isolates of Klebsiella pneumoniae from South America. Antimicrob. Agents Chemother. 50, 2880–2882 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Rada AM et al. Dynamics of blaKPC-2 Dissemination from Non-CG258 Klebsiella pneumoniae to Other Enterobacterales via IncN Plasmids in an Area of High Endemicity. Antimicrob. Agents Chemother. 64, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Navon-Venezia S et al. First report on a hyperepidemic clone of KPC-3-producing Klebsiella pneumoniae in Israel genetically related to a strain causing outbreaks in the United States. Antimicrob. Agents Chemother. 53, 818–820 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Karampatakis T, Antachopoulos C, Iosifidis E, Tsakris A & Roilides E Molecular epidemiology of carbapenem-resistant Klebsiella pneumoniae in Greece. Future Microbiol. 11, 809–823 (2016). [DOI] [PubMed] [Google Scholar]
- 38.Gartzonika K et al. Identification of a KPC-9-producing Klebsiella pneumoniae ST258 cluster among KPC-2-producing isolates of an ongoing outbreak in Northwestern Greece: a retrospective study. Clin. Microbiol. Infect. 24, 558–560 (2018). [DOI] [PubMed] [Google Scholar]
- 39.Vubil D et al. Outbreak of KPC-3-producing ST15 and ST348 Klebsiella pneumoniae in a Portuguese hospital. Epidemiol. Infect. 145, 595–599 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Baraniak A et al. Multiregional dissemination of KPC-producing Klebsiella pneumoniae ST258/ST512 genotypes in Poland, 2010–14. J. Antimicrob. Chemother. 72, 1610–1616 (2017). [DOI] [PubMed] [Google Scholar]
- 41.Tooke CL et al. β-Lactamases and β-Lactamase Inhibitors in the 21st Century. J. Mol. Biol. 431, 3472–3500 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Potter RF, D’Souza AW & Dantas G The rapid spread of carbapenem-resistant Enterobacteriaceae. Drug Resist. Updat. 29, 30–46 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Walsh TR, Toleman MA, Poirel L & Nordmann P Metallo-beta-lactamases: the quiet before the storm? Clin. Microbiol. Rev. 18, 306–325 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Walsh TR The emergence and implications of metallo-β-lactamases in Gram-negative bacteria. Clin. Microbiol. Infect. 11, 2–9 (2005). [DOI] [PubMed] [Google Scholar]
- 45.Wu W et al. NDM Metallo-β-Lactamases and Their Bacterial Producers in Health Care Settings. Clin. Microbiol. Rev. 32, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Watanabe M, Iyobe S, Inoue M & Mitsuhashi S Transferable imipenem resistance in Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 35, 147–151 (1991). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Malchione MD, Torres LM, Hartley DM, Koch M & Goodman JL Carbapenem and colistin resistance in Enterobacteriaceae in Southeast Asia: Review and mapping of emerging and overlapping challenges. Int. J. Antimicrob. Agents 54, 381–399 (2019). [DOI] [PubMed] [Google Scholar]
- 48.Matsumura Y et al. Global Molecular Epidemiology of IMP-Producing Enterobacteriaceae. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lauretti L et al. Cloning and characterization of blaVIM, a new integron-borne metallo-beta-lactamase gene from a Pseudomonas aeruginosa clinical isolate. Antimicrob. Agents Chemother. 43, 1584–1590 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Albiger B et al. Carbapenemase-producing Enterobacteriaceae in Europe: assessment by national experts from 38 countries, May 2015. Euro Surveill. 20, (2015). [DOI] [PubMed] [Google Scholar]
- 51.Yong D et al. Characterization of a new metallo-beta-lactamase gene, bla(NDM-1), and a novel erythromycin esterase gene carried on a unique genetic structure in Klebsiella pneumoniae sequence type 14 from India. Antimicrob. Agents Chemother. 53, 5046–5054 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Mairi A, Pantel A, Sotto A, Lavigne J-P & Touati A OXA-48-like carbapenemases producing Enterobacteriaceae in different niches. Eur. J. Clin. Microbiol. Infect. Dis. 37, 587–604 (2018). [DOI] [PubMed] [Google Scholar]
- 53.Poirel L, Potron A & Nordmann P OXA-48-like carbapenemases: the phantom menace. J. Antimicrob. Chemother. 67, 1597–1606 (2012). [DOI] [PubMed] [Google Scholar]
- 54.Pitout JDD, Peirano G, Kock MM, Strydom K-A & Matsumura Y The Global Ascendency of OXA-48-Type Carbapenemases. Clin. Microbiol. Rev. 33, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Walther-Rasmussen J & Høiby N OXA-type carbapenemases. J. Antimicrob. Chemother. 57, 373–383 (2006). [DOI] [PubMed] [Google Scholar]
- 56.Nordmann P, Dortet L & Poirel L Carbapenem resistance in Enterobacteriaceae: here is the storm! Trends Mol. Med. 18, 263–272 (2012). [DOI] [PubMed] [Google Scholar]
- 57.Guzmán-Puche J et al. Characterization of OXA-48-producing Klebsiella oxytoca isolates from a hospital outbreak in Tunisia. J Glob Antimicrob Resist 24, 306–310 (2021). [DOI] [PubMed] [Google Scholar]
- 58.Heireman L et al. Toilet drain water as a potential source of hospital room-to-room transmission of carbapenemase-producing Klebsiella pneumoniae. J. Hosp. Infect. 106, 232–239 (2020). [DOI] [PubMed] [Google Scholar]
- 59.Shaidullina E et al. Antimicrobial Resistance and Genomic Characterization of OXA-48- and CTX-M-15-Co-Producing Hypervirulent Klebsiella pneumoniae ST23 Recovered from Nosocomial Outbreak. Antibiotics (Basel) 9, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lyman M et al. Notes from the Field: Carbapenem-resistant Enterobacteriaceae Producing OXA-48-like Carbapenemases--United States, 2010–2015. MMWR Morb. Mortal. Wkly. Rep. 64, 1315–1316 (2015). [DOI] [PubMed] [Google Scholar]
- 61.Jean S-S et al. Carbapenemase-producing Gram-negative bacteria: current epidemics, antimicrobial susceptibility and treatment options. Future Microbiol. 10, 407–425 (2015). [DOI] [PubMed] [Google Scholar]
- 62.Ract P et al. Synergistic in vitro activity between aztreonam and amoxicillin-clavulanate against Enterobacteriaceae-producing class B and/or class D carbapenemases with or without extended-spectrum β-lactamases. J. Med. Microbiol. 68, 1292–1298 (2019). [DOI] [PubMed] [Google Scholar]
- 63.Biswas S, Brunel J-M, Dubus J-C, Reynaud-Gaubert M & Rolain J-M Colistin: an update on the antibiotic of the 21st century. Expert Rev. Anti. Infect. Ther. 10, 917–934 (2012). [DOI] [PubMed] [Google Scholar]
- 64.Capone A et al. High rate of colistin resistance among patients with carbapenem-resistant Klebsiella pneumoniae infection accounts for an excess of mortality. Clin. Microbiol. Infect. 19, E23–E30 (2013). [DOI] [PubMed] [Google Scholar]
- 65.Giacobbe DR et al. Risk factors for bloodstream infections due to colistin-resistant KPC-producing Klebsiella pneumoniae: results from a multicenter case-control-control study. Clin. Microbiol. Infect. 21, 1106.e1–8 (2015). [DOI] [PubMed] [Google Scholar]
- 66.Kai J & Wang S Recent progress on elucidating the molecular mechanism of plasmid-mediated colistin resistance and drug design. Int. Microbiol. 23, 355–366 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Band VI et al. Colistin Heteroresistance Is Largely Undetected among Carbapenem-Resistant Enterobacterales in the United States. MBio 12, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Seo J, Wi YM, Kim JM, Kim Y-J & Ko KS Detection of colistin-resistant populations prior to antibiotic exposure in KPC-2-producing Klebsiella pneumoniae clinical isolates. J. Microbiol. 59, 590–597 (2021). [DOI] [PubMed] [Google Scholar]
- 69.Motsch J et al. RESTORE-IMI 1: A Multicenter, Randomized, Double-blind Trial Comparing Efficacy and Safety of Imipenem/Relebactam vs Colistin Plus Imipenem in Patients With Imipenem-nonsusceptible Bacterial Infections. Clin. Infect. Dis. 70, 1799–1808 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wunderink RG et al. Effect and Safety of Meropenem-Vaborbactam versus Best-Available Therapy in Patients with Carbapenem-Resistant Enterobacteriaceae Infections: The TANGO II Randomized Clinical Trial. Infect Dis Ther 7, 439–455 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.van Duin D et al. Colistin Versus Ceftazidime-Avibactam in the Treatment of Infections Due to Carbapenem-Resistant Enterobacteriaceae. Clin. Infect. Dis. 66, 163–171 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Olowo-Okere A & Yacouba A Molecular mechanisms of colistin resistance in Africa: A systematic review of literature. Germs 10, 367–379 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Osei Sekyere J & Reta MA Genomic and Resistance Epidemiology of Gram-Negative Bacteria in Africa: a Systematic Review and Phylogenomic Analyses from a One Health Perspective. mSystems 5, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.World Health Organization. The detection and reporting of colistin resistance. https://apps.who.int/iris/bitstream/handle/10665/277175/WHO-WSI-AMR-2018.4-eng.pdf (2018).
- 75.WHO | Highest Priority Critically Important Antimicrobials. (2019).
- 76.Silver LL Fosfomycin: Mechanism and Resistance. Cold Spring Harb. Perspect. Med. 7, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Falagas ME, Vouloumanou EK, Samonis G & Vardakas KZ Fosfomycin. Clin. Microbiol. Rev. 29, 321–347 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ito R et al. Widespread Fosfomycin Resistance in Gram-Negative Bacteria Attributable to the Chromosomal fosA Gene. MBio 8, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Huang L et al. Prevalence and mechanisms of fosfomycin resistance among KPC-producing Klebsiella pneumoniae clinical isolates in China. Int. J. Antimicrob. Agents 57, 106226 (2021). [DOI] [PubMed] [Google Scholar]
- 80.Michalopoulos AS, Livaditis IG & Gougoutas V The revival of fosfomycin. Int. J. Infect. Dis. 15, e732–9 (2011). [DOI] [PubMed] [Google Scholar]
- 81.Falagas ME, Giannopoulou KP, Kokolakis GN & Rafailidis PI Fosfomycin: use beyond urinary tract and gastrointestinal infections. Clin. Infect. Dis. 46, 1069–1077 (2008). [DOI] [PubMed] [Google Scholar]
- 82.Michalopoulos A et al. Intravenous fosfomycin for the treatment of nosocomial infections caused by carbapenem-resistant Klebsiella pneumoniae in critically ill patients: a prospective evaluation. Clin. Microbiol. Infect. 16, 184–186 (2010). [DOI] [PubMed] [Google Scholar]
- 83.Pontikis K et al. Outcomes of critically ill intensive care unit patients treated with fosfomycin for infections due to pandrug-resistant and extensively drug-resistant carbapenemase-producing Gram-negative bacteria. Int. J. Antimicrob. Agents 43, 52–59 (2014). [DOI] [PubMed] [Google Scholar]
- 84.US Food and Drug Administration. MONUROL. US Food and Drug Administration; https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/050717s007lbl.pdf. [Google Scholar]
- 85.Dimitrova EK Fosfomycin-containing medicinal products. https://www.ema.europa.eu/en/medicines/human/referrals/fosfomycin-containing-medicinal-products (2018).
- 86.Seifert H, Blondeau J & Dowzicky MJ In vitro activity of tigecycline and comparators (2014–2016) among key WHO ‘priority pathogens’ and longitudinal assessment (2004–2016) of antimicrobial resistance: a report from the T.E.S.T. study. Int. J. Antimicrob. Agents 52, 474–484 (2018). [DOI] [PubMed] [Google Scholar]
- 87.Ni W et al. Tigecycline Treatment for Carbapenem-Resistant Enterobacteriaceae Infections: A Systematic Review and Meta-Analysis. Medicine 95, e3126 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Sbrana F et al. Carbapenem-sparing antibiotic regimens for infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae in intensive care unit. Clin. Infect. Dis. 56, 697–700 (2013). [DOI] [PubMed] [Google Scholar]
- 89.Tumbarello M et al. Predictors of mortality in bloodstream infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae: importance of combination therapy. Clin. Infect. Dis. 55, 943–950 (2012). [DOI] [PubMed] [Google Scholar]
- 90.Pournaras S, Koumaki V, Spanakis N, Gennimata V & Tsakris A Current perspectives on tigecycline resistance in Enterobacteriaceae: susceptibility testing issues and mechanisms of resistance. Int. J. Antimicrob. Agents 48, 11–18 (2016). [DOI] [PubMed] [Google Scholar]
- 91.Yoon EJ, Oh Y & Jeong SH Development of Tigecycline Resistance in Carbapenemase-Producing Klebsiella pneumoniae Sequence Type 147 via AcrAB Overproduction Mediated by Replacement of the ramA Promoter. Ann. Lab. Med. 40, 15–20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Sun J et al. Plasmid-encoded tet(X) genes that confer high-level tigecycline resistance in Escherichia coli. Nat Microbiol 4, 1457–1464 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.He T et al. Emergence of plasmid-mediated high-level tigecycline resistance genes in animals and humans. Nat Microbiol 4, 1450–1456 (2019). [DOI] [PubMed] [Google Scholar]
- 94.Peyclit L, Baron SA & Rolain J-M Drug Repurposing to Fight Colistin and Carbapenem-Resistant Bacteria. Front. Cell. Infect. Microbiol. 9, 193 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.MacNair CR et al. Overcoming mcr-1 mediated colistin resistance with colistin in combination with other antibiotics. Nat. Commun. 9, 458 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Hu Y, Liu Y & Coates A Azidothymidine Produces Synergistic Activity in Combination with Colistin against Antibiotic-Resistant Enterobacteriaceae. Antimicrob. Agents Chemother. 63, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Ng SMS et al. Repurposing Zidovudine in combination with Tigecycline for treating carbapenem-resistant Enterobacteriaceae infections. Eur. J. Clin. Microbiol. Infect. Dis. 37, 141–148 (2018). [DOI] [PubMed] [Google Scholar]
- 98.Cebrero-Cangueiro T et al. In vitro Activity of Pentamidine Alone and in Combination With Aminoglycosides, Tigecycline, Rifampicin, and Doripenem Against Clinical Strains of Carbapenemase-Producing and/or Colistin-Resistant Enterobacteriaceae. Front. Cell. Infect. Microbiol. 8, 363 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Otto RG et al. An alternative strategy for combination therapy: Interactions between polymyxin B and non-antibiotics. Int. J. Antimicrob. Agents 53, 34–39 (2019). [DOI] [PubMed] [Google Scholar]
- 100.Zhanel GG et al. Ceftazidime-Avibactam: a Novel Cephalosporin/b-lactamase Inhibitor Combination. Drugs; Auckland 73, 159–177 (2013). [DOI] [PubMed] [Google Scholar]
- 101.Ehmann DE et al. Avibactam is a covalent, reversible, non-β-lactam β-lactamase inhibitor. Proc. Natl. Acad. Sci. U. S. A. 109, 11663–11668 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Sousa A et al. Effectiveness of ceftazidime/avibactam as salvage therapy for treatment of infections due to OXA-48 carbapenemase-producing Enterobacteriaceae. J. Antimicrob. Chemother. 73, 3170–3175 (2018). [DOI] [PubMed] [Google Scholar]
- 103.De la Calle C et al. Clinical characteristics and prognosis of infections caused by OXA-48 carbapenemase-producing Enterobacteriaceae in patients treated with ceftazidime-avibactam. Int. J. Antimicrob. Agents 53, 520–524 (2019). [DOI] [PubMed] [Google Scholar]
- 104.Shields RK et al. Ceftazidime-Avibactam Is Superior to Other Treatment Regimens against Carbapenem-Resistant Klebsiella pneumoniae Bacteremia. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tumbarello M et al. Ceftazidime-avibactam use for KPC-Kp infections: a retrospective observational multicenter study. Clin. Infect. Dis. (2021) doi: 10.1093/cid/ciab176. [DOI] [PubMed] [Google Scholar]
- 106.Wilson GM et al. Meta-analysis of Clinical Outcomes Using Ceftazidime/Avibactam, Ceftolozane/Tazobactam, and Meropenem/Vaborbactam for the Treatment of Multidrug-Resistant Gram-Negative Infections. Open Forum Infect Dis 8, ofaa651 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.United States Food and Drug Administration. AVYCAZ safely and effectively. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/206494s005,s006lbl.pdf.
- 108.Mazuski JE et al. Efficacy and Safety of Ceftazidime-Avibactam Plus Metronidazole Versus Meropenem in the Treatment of Complicated Intra-abdominal Infection: Results From a Randomized, Controlled, Double-Blind, Phase 3 Program. Clin. Infect. Dis. 62, 1380–1389 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Wagenlehner FM et al. Ceftazidime-avibactam Versus Doripenem for the Treatment of Complicated Urinary Tract Infections, Including Acute Pyelonephritis: RECAPTURE, a Phase 3 Randomized Trial Program. Clin. Infect. Dis. 63, 754–762 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Torres A et al. Ceftazidime-avibactam versus meropenem in nosocomial pneumonia, including ventilator-associated pneumonia (REPROVE): a randomised, double-blind, phase 3 non-inferiority trial. Lancet Infect. Dis. 18, 285–295 (2018). [DOI] [PubMed] [Google Scholar]
- 111.de Jonge BLM et al. In Vitro Susceptibility to Ceftazidime-Avibactam of Carbapenem-Nonsusceptible Enterobacteriaceae Isolates Collected during the INFORM Global Surveillance Study (2012 to 2014). Antimicrob. Agents Chemother. 60, 3163–3169 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Spiliopoulou I, Kazmierczak K & Stone GG In vitro activity of ceftazidime/avibactam against isolates of carbapenem-non-susceptible Enterobacteriaceae collected during the INFORM global surveillance programme (2015–17). J. Antimicrob. Chemother. 75, 384–391 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Nelson K et al. Resistance to Ceftazidime-Avibactam Is Due to Transposition of KPC in a Porin-Deficient Strain of Klebsiella pneumoniae with Increased Efflux Activity. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Humphries RM & Hemarajata P Resistance to Ceftazidime-Avibactam in Klebsiella pneumoniae Due to Porin Mutations and the Increased Expression of KPC-3. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Humphries RM et al. First Report of Ceftazidime-Avibactam Resistance in a KPC-3-Expressing Klebsiella pneumoniae Isolate. Antimicrob. Agents Chemother. 59, 6605–6607 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Shields RK et al. Emergence of Ceftazidime-Avibactam Resistance Due to Plasmid-Borne blaKPC-3 Mutations during Treatment of Carbapenem-Resistant Klebsiella pneumoniae Infections. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Poirel L et al. KPC-50 Confers Resistance to Ceftazidime-Avibactam Associated with Reduced Carbapenemase Activity. Antimicrob. Agents Chemother. 64, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Winkler ML, Papp-Wallace KM & Bonomo RA Activity of ceftazidime/avibactam against isogenic strains of Escherichia coli containing KPC and SHV β-lactamases with single amino acid substitutions in the Ω-loop. J. Antimicrob. Chemother. 70, 2279–2286 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Shields RK et al. Clinical Outcomes, Drug Toxicity, and Emergence of Ceftazidime-Avibactam Resistance Among Patients Treated for Carbapenem-Resistant Enterobacteriaceae Infections. Clin. Infect. Dis. 63, 1615–1618 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Zhang Y, Kashikar A, Brown CA, Denys G & Bush K Unusual Escherichia coli PBP 3 Insertion Sequence Identified from a Collection of Carbapenem-Resistant Enterobacteriaceae Tested In Vitro with a Combination of Ceftazidime-, Ceftaroline-, or Aztreonam-Avibactam. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Maraki S et al. Ceftazidime-avibactam, meropenen-vaborbactam, and imipenem-relebactam in combination with aztreonam against multidrug-resistant, metallo-β-lactamase-producing Klebsiella pneumoniae. Eur. J. Clin. Microbiol. Infect. Dis. (2021) doi: 10.1007/s10096-021-04197-3. [DOI] [PubMed] [Google Scholar]
- 122.Shields RK & Doi Y Aztreonam Combination Therapy: An Answer to Metallo-β-Lactamase-Producing Gram-Negative Bacteria? Clin. Infect. Dis. 71, 1099–1101 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Karlowsky JA et al. In Vitro Activity of Aztreonam-Avibactam against Enterobacteriaceae and Pseudomonas aeruginosa Isolated by Clinical Laboratories in 40 Countries from 2012 to 2015. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Shaw E et al. Clinical outcomes after combination treatment with ceftazidime/avibactam and aztreonam for NDM-1/OXA-48/CTX-M-15-producing Klebsiella pneumoniae infection. J. Antimicrob. Chemother. 73, 1104–1106 (2018). [DOI] [PubMed] [Google Scholar]
- 125.Falcone M et al. Efficacy of ceftazidime-avibactam plus aztreonam in patients with bloodstream infections caused by MBL- producing Enterobacterales. Clin. Infect. Dis. (2020) doi: 10.1093/cid/ciaa586. [DOI] [PubMed] [Google Scholar]
- 126.Alghoribi MF et al. Successful treatment of infective endocarditis due to pandrug-resistant Klebsiella pneumoniae with ceftazidime-avibactam and aztreonam. Sci. Rep. 11, 9684 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Efficacy, Safety, and Tolerability of ATM-AVI in the Treatment of Serious Infection Due to MBL-producing Gram-negative Bacteria. https://clinicaltrials.gov/ct2/show/NCT03580044.
- 128.Cornely OA et al. Pharmacokinetics and safety of aztreonam/avibactam for the treatment of complicated intra-abdominal infections in hospitalized adults: results from the REJUVENATE study. J. Antimicrob. Chemother. 75, 618–627 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.United States Food and Drug Administration. VABOMERE (meropenem and vaborbactam) for injection. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/209776lbl.pdf.
- 130.European Medicines Agency. Vaborem. https://www.ema.europa.eu/en/medicines/human/EPAR/vaborem (2018).
- 131.Cho JC, Zmarlicka MT, Shaeer KM & Pardo J Meropenem/Vaborbactam, the First Carbapenem/β-Lactamase Inhibitor Combination. Ann. Pharmacother. 52, 769–779 (2018). [DOI] [PubMed] [Google Scholar]
- 132.Hecker SJ et al. Discovery of a Cyclic Boronic Acid β-Lactamase Inhibitor (RPX7009) with Utility vs Class A Serine Carbapenemases. J. Med. Chem. 58, 3682–3692 (2015). [DOI] [PubMed] [Google Scholar]
- 133.Lomovskaya O et al. Vaborbactam: Spectrum of Beta-Lactamase Inhibition and Impact of Resistance Mechanisms on Activity in Enterobacteriaceae. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Castanheira M, Huband MD, Mendes RE & Flamm RK Meropenem-Vaborbactam Tested against Contemporary Gram-Negative Isolates Collected Worldwide during 2014, Including Carbapenem-Resistant, KPC-Producing, Multidrug-Resistant, and Extensively Drug-Resistant Enterobacteriaceae. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kaye KS et al. Effect of Meropenem-Vaborbactam vs Piperacillin-Tazobactam on Clinical Cure or Improvement and Microbial Eradication in Complicated Urinary Tract Infection: The TANGO I Randomized Clinical Trial. JAMA 319, 788–799 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Sun D, Rubio-Aparicio D, Nelson K, Dudley MN & Lomovskaya O Meropenem-Vaborbactam Resistance Selection, Resistance Prevention, and Molecular Mechanisms in Mutants of KPC-Producing Klebsiella pneumoniae. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Wilson WR et al. Effects of KPC Variant and Porin Genotype on the In Vitro Activity of Meropenem-Vaborbactam against Carbapenem-Resistant Enterobacteriaceae. Antimicrob. Agents Chemother. 63, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Dulyayangkul P, Wan Nur Ismah WAK, Douglas EJA & Avison MB Mutation of kvrA Causes OmpK35 and OmpK36 Porin Downregulation and Reduced Meropenem-Vaborbactam Susceptibility in KPC-Producing Klebsiella pneumoniae. Antimicrob. Agents Chemother. 64, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Olsen I New promising β-lactamase inhibitors for clinical use. Eur. J. Clin. Microbiol. Infect. Dis. 34, 1303–1308 (2015). [DOI] [PubMed] [Google Scholar]
- 140.Zhanel GG et al. Imipenem–Relebactam and Meropenem–Vaborbactam: Two Novel Carbapenem-ß-Lactamase Inhibitor Combinations. Drugs; Auckland 78, 65–98 (2018). [DOI] [PubMed] [Google Scholar]
- 141.Campanella TA & Gallagher JC A Clinical Review and Critical Evaluation of Imipenem-Relebactam: Evidence to Date. Infect. Drug Resist. 13, 4297–4308 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Lob SH et al. In vitro activity of imipenem-relebactam against resistant phenotypes of Enterobacteriaceae and Pseudomonas aeruginosa isolated from intraabdominal and urinary tract infection samples - SMART Surveillance Europe 2015–2017. J. Med. Microbiol. 69, 207–217 (2020). [DOI] [PubMed] [Google Scholar]
- 143.United States Food and Drug Administration. RECARBRIO. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/212819s000lbl.pdf.
- 144.Smith JR, Rybak JM & Claeys KC Imipenem-Cilastatin-Relebactam: A Novel β-Lactam-β-Lactamase Inhibitor Combination for the Treatment of Multidrug-Resistant Gram-Negative Infections. Pharmacotherapy 40, 343–356 (2020). [DOI] [PubMed] [Google Scholar]
- 145.Titov I et al. A Randomized, Double-blind, Multicenter Trial Comparing Efficacy and Safety of Imipenem/Cilastatin/Relebactam Versus Piperacillin/Tazobactam in Adults With Hospital-acquired or Ventilator-associated Bacterial Pneumonia (RESTORE-IMI 2 Study). Clin. Infect. Dis. (2020) doi: 10.1093/cid/ciaa803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.van Duin D & Bonomo RA Ceftazidime/Avibactam and Ceftolozane/Tazobactam: Second-generation β-Lactam/β-Lactamase Inhibitor Combinations. Clin. Infect. Dis. 63, 234–241 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Armstrong ES & Miller GH Combating evolution with intelligent design: the neoglycoside ACHN-490. Curr. Opin. Microbiol. 13, 565–573 (2010). [DOI] [PubMed] [Google Scholar]
- 148.Livermore DM et al. Activity of aminoglycosides, including ACHN-490, against carbapenem-resistant Enterobacteriaceae isolates. J. Antimicrob. Chemother. 66, 48–53 (2011). [DOI] [PubMed] [Google Scholar]
- 149.Eljaaly K, Alharbi A, Alshehri S, Ortwine JK & Pogue JM Plazomicin: A Novel Aminoglycoside for the Treatment of Resistant Gram-Negative Bacterial Infections. Drugs 79, 243–269 (2019). [DOI] [PubMed] [Google Scholar]
- 150.Walkty A et al. In vitro activity of plazomicin against 5,015 gram-negative and gram-positive clinical isolates obtained from patients in canadian hospitals as part of the CANWARD study, 2011–2012. Antimicrob. Agents Chemother. 58, 2554–2563 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.United States Food and Drug Administration. Zemdri. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/210303orig1s000lbl.pdf. [Google Scholar]
- 152.Wagenlehner FME et al. Once-Daily Plazomicin for Complicated Urinary Tract Infections. N. Engl. J. Med. 380, 729–740 (2019). [DOI] [PubMed] [Google Scholar]
- 153.McKinnell JA et al. Plazomicin for Infections Caused by Carbapenem-Resistant Enterobacteriaceae. N. Engl. J. Med. 380, 791–793 (2019). [DOI] [PubMed] [Google Scholar]
- 154.Theuretzbacher U & Paul M Developing a new antibiotic for extensively drug-resistant pathogens: the case of plazomicin. Clin. Microbiol. Infect. 24, 1231–1233 (2018). [DOI] [PubMed] [Google Scholar]
- 155.Dimitrova EK Zemdri: Withdrawn application - European Medicines Agency. https://www.ema.europa.eu/en/medicines/human/withdrawn-applications/zemdri (2020).
- 156.Mullard A Achaogen bankruptcy highlights antibacterial development woes. Nat. Rev. Drug Discov. 18, 411 (2019). [DOI] [PubMed] [Google Scholar]
- 157.Shaeer KM, Zmarlicka MT, Chahine EB, Piccicacco N & Cho JC Plazomicin: A Next-Generation Aminoglycoside. Pharmacotherapy 39, 77–93 (2019). [DOI] [PubMed] [Google Scholar]
- 158.Zhanel GG et al. Comparison of the next-generation aminoglycoside plazomicin to gentamicin, tobramycin and amikacin. Expert Rev. Anti. Infect. Ther. 10, 459–473 (2012). [DOI] [PubMed] [Google Scholar]
- 159.Roch M et al. Vertical and horizontal dissemination of an IncC plasmid harbouring rmtB 16S rRNA methylase gene, conferring resistance to plazomicin, among invasive ST258 and ST16 KPC-producing Klebsiella pneumoniae. J Glob Antimicrob Resist 24, 183–189 (2020). [DOI] [PubMed] [Google Scholar]
- 160.Xiao X-Y et al. Fluorocyclines. 1. 7-fluoro-9-pyrrolidinoacetamido-6-demethyl-6-deoxytetracycline: a potent, broad spectrum antibacterial agent. J. Med. Chem. 55, 597–605 (2012). [DOI] [PubMed] [Google Scholar]
- 161.Zhanel GG et al. Review of Eravacycline, a Novel Fluorocycline Antibacterial Agent. Drugs 76, 567–588 (2016). [DOI] [PubMed] [Google Scholar]
- 162.Zhang Y, Lin X & Bush K In vitro susceptibility of β-lactamase-producing carbapenem-resistant Enterobacteriaceae (CRE) to eravacycline. J. Antibiot. 69, 600–604 (2016). [DOI] [PubMed] [Google Scholar]
- 163.Livermore DM, Mushtaq S, Warner M & Woodford N In Vitro Activity of Eravacycline against Carbapenem-Resistant Enterobacteriaceae and Acinetobacter baumannii. Antimicrob. Agents Chemother. 60, 3840–3844 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Clark JA, Kulengowski B & Burgess DS In vitro activity of eravacycline compared with tigecycline against carbapenem-resistant Enterobacteriaceae. Int. J. Antimicrob. Agents 56, 106178 (2020). [DOI] [PubMed] [Google Scholar]
- 165.Solomkin JS et al. Eravacycline: a new treatment option for complicated intra-abdominal infections in the age of multidrug resistance. Future Microbiol. 14, 1293–1308 (2019). [DOI] [PubMed] [Google Scholar]
- 166.United States Food and Drug Administration. XERAVA (eravacycline) for injection. https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/211109lbl.pdf.
- 167.European Medicines Agency. Xerava, INN-eravacycline. https://www.ema.europa.eu/en/documents/product-information/xerava-epar-product-information_en.pdf.
- 168.Grossman TH, O’Brien W, Kerstein KO & Sutcliffe JA Eravacycline (TP-434) is active in vitro against biofilms formed by uropathogenic Escherichia coli. Antimicrob. Agents Chemother. 59, 2446–2449 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Efficacy and Safety Study of Eravacycline Compared With Levofloxacin in Complicated Urinary Tract Infections. https://clinicaltrials.gov/ct2/show/NCT01978938.
- 170.Heaney M, Mahoney MV & Gallagher JC Eravacycline: The Tetracyclines Strike Back. Ann. Pharmacother. 53, 1124–1135 (2019). [DOI] [PubMed] [Google Scholar]
- 171.Grossman TH et al. Target- and resistance-based mechanistic studies with TP-434, a novel fluorocycline antibiotic. Antimicrob. Agents Chemother. 56, 2559–2564 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Park J et al. Plasticity, dynamics, and inhibition of emerging tetracycline resistance enzymes. Nat. Chem. Biol. 13, 730–736 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ding Y et al. Emergence of tigecycline- and eravacycline-resistant Tet(X4)-producing Enterobacteriaceae in the gut microbiota of healthy Singaporeans. J. Antimicrob. Chemother. 75, 3480–3484 (2020). [DOI] [PubMed] [Google Scholar]
- 174.Honeyman L et al. Structure-activity relationship of the aminomethylcyclines and the discovery of omadacycline. Antimicrob. Agents Chemother. 59, 7044–7053 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Zhanel GG et al. Omadacycline: A Novel Oral and Intravenous Aminomethylcycline Antibiotic Agent. Drugs 80, 285–313 (2020). [DOI] [PubMed] [Google Scholar]
- 176.O’Riordan W et al. Omadacycline for Acute Bacterial Skin and Skin-Structure Infections. N. Engl. J. Med. 380, 528–538 (2019). [DOI] [PubMed] [Google Scholar]
- 177.Stets R et al. Omadacycline for Community-Acquired Bacterial Pneumonia. N. Engl. J. Med. 380, 517–527 (2019). [DOI] [PubMed] [Google Scholar]
- 178.O’Riordan W et al. Once-daily oral omadacycline versus twice-daily oral linezolid for acute bacterial skin and skin structure infections (OASIS-2): a phase 3, double-blind, multicentre, randomised, controlled, non-inferiority trial. Lancet Infect. Dis. 19, 1080–1090 (2019). [DOI] [PubMed] [Google Scholar]
- 179.United States Food and Drug Administration. NUZYRA (omadacycline). https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/209816_209817lbl.pdf.
- 180.Francisco EM Nuzyra: Withdrawn application - European Medicines Agency. https://www.ema.europa.eu/en/medicines/human/withdrawn-applications/nuzyra (2019).
- 181.Lutgring JD et al. Antibiotic Susceptibility of NDM-Producing Enterobacterales Collected in the United States in 2017 and 2018. Antimicrob. Agents Chemother. 64, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Möllmann U, Heinisch L, Bauernfeind A, Köhler T & Ankel-Fuchs D Siderophores as drug delivery agents: application of the ‘Trojan Horse’ strategy. Biometals 22, 615–624 (2009). [DOI] [PubMed] [Google Scholar]
- 183.El-Lababidi RM & Rizk JG Cefiderocol: A Siderophore Cephalosporin. Ann. Pharmacother. 54, 1215–1231 (2020). [DOI] [PubMed] [Google Scholar]
- 184.Ito-Horiyama T et al. Stability of Novel Siderophore Cephalosporin S-649266 against Clinically Relevant Carbapenemases. Antimicrob. Agents Chemother. 60, 4384–4386 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Poirel L, Kieffer N & Nordmann P Stability of cefiderocol against clinically significant broad-spectrum oxacillinases. Int. J. Antimicrob. Agents 52, 866–867 (2018). [DOI] [PubMed] [Google Scholar]
- 186.Katsube T, Echols R & Wajima T Pharmacokinetic and Pharmacodynamic Profiles of Cefiderocol, a Novel Siderophore Cephalosporin. Clin. Infect. Dis. 69, S552–S558 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Kohira N et al. In Vitro Antimicrobial Activity of a Siderophore Cephalosporin, S-649266, against Enterobacteriaceae Clinical Isolates, Including Carbapenem-Resistant Strains. Antimicrob. Agents Chemother. 60, 729–734 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zhanel GG et al. Cefiderocol: A Siderophore Cephalosporin with Activity Against Carbapenem-Resistant and Multidrug-Resistant Gram-Negative Bacilli. Drugs 79, 271–289 (2019). [DOI] [PubMed] [Google Scholar]
- 189.United States Food and Drug Administration. FETROJA (cefiderocol) for injection, for intravenous use. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/209445s002lbl.pdf.
- 190.Portsmouth S et al. Cefiderocol versus imipenem-cilastatin for the treatment of complicated urinary tract infections caused by Gram-negative uropathogens: a phase 2, randomised, double-blind, non-inferiority trial. Lancet Infect. Dis. 18, 1319–1328 (2018). [DOI] [PubMed] [Google Scholar]
- 191.Wunderink RG et al. Cefiderocol versus high-dose, extended-infusion meropenem for the treatment of Gram-negative nosocomial pneumonia (APEKS-NP): a randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect. Dis. 21, 213–225 (2021). [DOI] [PubMed] [Google Scholar]
- 192.Dimitrova EK Fetcroja. https://www.ema.europa.eu/en/medicines/human/EPAR/fetcroja (2020).
- 193.Bassetti M et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant Gram-negative bacteria (CREDIBLE-CR): a randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect. Dis. 21, 226–240 (2021). [DOI] [PubMed] [Google Scholar]
- 194.Yamano Y In Vitro Activity of Cefiderocol Against a Broad Range of Clinically Important Gram-negative Bacteria. Clin. Infect. Dis. 69, S544–S551 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Hackel MA et al. 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-Nonsusceptible Isolates (SIDERO-WT-2014 Study). Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Hackel MA et al. In Vitro Activity of the Siderophore Cephalosporin, Cefiderocol, against Carbapenem-Nonsusceptible and Multidrug-Resistant Isolates of Gram-Negative Bacilli Collected Worldwide in 2014 to 2016. Antimicrob. Agents Chemother. 62, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Kohira N et al. Reduced susceptibility mechanism to cefiderocol, a siderophore cephalosporin, among clinical isolates from a global surveillance programme (SIDERO-WT-2014). J Glob Antimicrob Resist 22, 738–741 (2020). [DOI] [PubMed] [Google Scholar]
- 198.Kortright KE, Chan BK, Koff JL & Turner PE Phage Therapy: A Renewed Approach to Combat Antibiotic-Resistant Bacteria. Cell Host Microbe 25, 219–232 (2019). [DOI] [PubMed] [Google Scholar]
- 199.Dedrick RM et al. Engineered bacteriophages for treatment of a patient with a disseminated drug-resistant Mycobacterium abscessus. Nat. Med. 25, 730–733 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Gordillo Altamirano FL & Barr JJ Phage Therapy in the Postantibiotic Era. Clin. Microbiol. Rev. 32, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Rehman S, Ali Z, Khan M, Bostan N & Naseem S The dawn of phage therapy. Rev. Med. Virol. 29, e2041 (2019). [DOI] [PubMed] [Google Scholar]
- 202.Reindel R & Fiore CR Phage Therapy: Considerations and Challenges for Development. Clinical infectious diseases: an official publication of the Infectious Diseases Society of America vol. 64 1589–1590 (2017). [DOI] [PubMed] [Google Scholar]
- 203.Sweere JM et al. Bacteriophage trigger antiviral immunity and prevent clearance of bacterial infection. Science 363, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Chanishvili N Phage therapy--history from Twort and d’Herelle through Soviet experience to current approaches. Adv. Virus Res. 83, 3–40 (2012). [DOI] [PubMed] [Google Scholar]
- 205.Jault P et al. Efficacy and tolerability of a cocktail of bacteriophages to treat burn wounds infected by Pseudomonas aeruginosa (PhagoBurn): a randomised, controlled, double-blind phase 1/2 trial. Lancet Infect. Dis. 19, 35–45 (2019). [DOI] [PubMed] [Google Scholar]
- 206.Aslam S et al. Lessons Learned From the First 10 Consecutive Cases of Intravenous Bacteriophage Therapy to Treat Multidrug-Resistant Bacterial Infections at a Single Center in the United States. Open Forum Infect Dis 7, ofaa389 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Amarillas L et al. Isolation and Characterization of phiLLS, a Novel Phage with Potential Biocontrol Agent against Multidrug-Resistant Escherichia coli. Front. Microbiol. 8, 1355 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Oliveira H et al. Characterization and genome sequencing of a Citrobacter freundii phage CfP1 harboring a lysin active against multidrug-resistant isolates. Appl. Microbiol. Biotechnol. 100, 10543–10553 (2016). [DOI] [PubMed] [Google Scholar]
- 209.Li M et al. Characterization and genome analysis of Klebsiella phage P509, with lytic activity against clinical carbapenem-resistant Klebsiella pneumoniae of the KL64 capsular type. Arch. Virol. 165, 2799–2806 (2020). [DOI] [PubMed] [Google Scholar]
- 210.Li M et al. Isolation and Characterization of Novel Lytic Bacteriophages Infecting Epidemic Carbapenem-Resistant Klebsiella pneumoniae Strains. Front. Microbiol. 11, 1554 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Horváth M et al. Identification of a newly isolated lytic bacteriophage against K24 capsular type, carbapenem resistant Klebsiella pneumoniae isolates. Sci. Rep. 10, 5891 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Ciacci N et al. Characterization of vB_Kpn_F48, a Newly Discovered Lytic Bacteriophage for Klebsiella pneumoniae of Sequence Type 101. Viruses 10, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Anand T et al. Phage therapy for treatment of virulent Klebsiella pneumoniae infection in a mouse model. J Glob Antimicrob Resist 21, 34–41 (2020). [DOI] [PubMed] [Google Scholar]
- 214.Labrie SJ, Samson JE & Moineau S Bacteriophage resistance mechanisms. Nat. Rev. Microbiol. 8, 317–327 (2010). [DOI] [PubMed] [Google Scholar]
- 215.Guo D et al. Genetic and Chemical Engineering of Phages for Controlling Multidrug-Resistant Bacteria. Antibiotics (Basel) 10, (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.World Health Organization. ANTIBACTERIAL AGENTS IN CLINICAL DEVELOPMENT An analysis of the antibacterial clinical development pipeline, including tuberculosis. (2017).
- 217.Livermore DM, Mushtaq S, Warner M, Vickers A & Woodford N In vitro activity of cefepime/zidebactam (WCK 5222) against Gram-negative bacteria. J. Antimicrob. Chemother. 72, 1373–1385 (2017). [DOI] [PubMed] [Google Scholar]
- 218.Thomson KS, AbdelGhani S, Snyder JW & Thomson GK Activity of Cefepime-Zidebactam against Multidrug-Resistant (MDR) Gram-Negative Pathogens. Antibiotics (Basel) 8, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Sader HS, Castanheira M, Huband M, Jones RN & Flamm RK WCK 5222 (Cefepime-Zidebactam) Antimicrobial Activity against Clinical Isolates of Gram-Negative Bacteria Collected Worldwide in 2015. Antimicrob. Agents Chemother. 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Rodvold KA et al. Plasma and Intrapulmonary Concentrations of Cefepime and Zidebactam following Intravenous Administration of WCK 5222 to Healthy Adult Subjects. Antimicrob. Agents Chemother. 62, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Preston RA et al. Single-Center Evaluation of the Pharmacokinetics of WCK 5222 (Cefepime-Zidebactam Combination) in Subjects with Renal Impairment. Antimicrob. Agents Chemother. 63, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Liu B et al. Discovery of Taniborbactam (VNRX-5133): A Broad-Spectrum Serine- and Metallo-β-lactamase Inhibitor for Carbapenem-Resistant Bacterial Infections. J. Med. Chem. 63, 2789–2801 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Krajnc A et al. Bicyclic Boronate VNRX-5133 Inhibits Metallo- and Serine-β-Lactamases. J. Med. Chem. 62, 8544–8556 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Hamrick JC et al. VNRX-5133 (Taniborbactam), a Broad-Spectrum Inhibitor of Serine- and Metallo-β-Lactamases, Restores Activity of Cefepime in Enterobacterales and Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 64, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Abdelraouf K, Almarzoky Abuhussain S & Nicolau DP In vivo pharmacodynamics of new-generation β-lactamase inhibitor taniborbactam (formerly VNRX-5133) in combination with cefepime against serine-β-lactamase-producing Gram-negative bacteria. J. Antimicrob. Chemother. 75, 3601–3610 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Reck F et al. Optimization of novel monobactams with activity against carbapenem-resistant Enterobacteriaceae - Identification of LYS228. Bioorg. Med. Chem. Lett. 28, 748–755 (2018). [DOI] [PubMed] [Google Scholar]
- 227.Blais J et al. In Vitro Activity of LYS228, a Novel Monobactam Antibiotic, against Multidrug-Resistant Enterobacteriaceae. Antimicrob. Agents Chemother. 62, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Dean CR et al. Mode of Action of the Monobactam LYS228 and Mechanisms Decreasing In Vitro Susceptibility in Escherichia coli and Klebsiella pneumoniae. Antimicrob. Agents Chemother. 62, (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Osborn M et al. A First-in-Human Study To Assess the Safety and Pharmacokinetics of LYS228, a Novel Intravenous Monobactam Antibiotic in Healthy Volunteers. Antimicrob. Agents Chemother. 63, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Novartis licenses three novel anti-infective programs to Boston Pharmaceuticals. https://www.novartis.com/news/media-releases/novartis-licenses-three-novel-anti-infective-programs-boston-pharmaceuticals.
- 231.Barnes MD et al. Nacubactam Enhances Meropenem Activity against Carbapenem-Resistant Klebsiella pneumoniae Producing KPC. Antimicrob. Agents Chemother. 63, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Davies DT et al. Discovery of ANT3310, a Novel Broad-Spectrum Serine β-Lactamase Inhibitor of the Diazabicyclooctane Class, Which Strongly Potentiates Meropenem Activity against Carbapenem-Resistant Enterobacterales and Acinetobacter baumannii. J. Med. Chem. 63, 15802–15820 (2020). [DOI] [PubMed] [Google Scholar]
- 233.Mushtaq S, Vickers A, Woodford N, Haldimann A & Livermore DM Activity of nacubactam (RG6080/OP0595) combinations against MBL-producing Enterobacteriaceae. J. Antimicrob. Chemother. 74, 953–960 (2019). [DOI] [PubMed] [Google Scholar]
- 234.Mallalieu NL et al. Safety and Pharmacokinetic Characterization of Nacubactam, a Novel β-Lactamase Inhibitor, Alone and in Combination with Meropenem, in Healthy Volunteers. Antimicrob. Agents Chemother. 64, (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]