Faculty
Wayne State University School of Medicine
Detroit, MI
University of Illinois at Chicago
Colleges of Pharmacy and Medicine
Chicago, IL
Director, Medical ICU
Professor of Medicine
MedStar Health Washington, DC
Activity Goal
The goal of this activity is to advance knowledge in the treatment of metallo-β-lactamase (MBL)-producing, carbapenem-resistant (CR) gram-negative bacterial infections (GNBIs), with specific reference to their epidemiology, resistance challenges, and approved treatment options, with comparative data to support clinical decision making.
Intended Audience
The primary intended audience for the activity includes infectious disease physicians, critical care specialists, pulmonologists, and PharmDs in these specialties. Secondary audiences include other health care professionals who may treat and monitor seriously ill patients with resistant infections, such as nurse practitioners, physician assistants, and microbiologists.
Learning Objectives
After completing this activity, participants should be better able to:
- Describe current knowledge as to the epidemiology of MBL-producing, CR-GNBIs.
- Review approved treatment options for CR-GNBIs.
- Cite comparative in vitro and clinical data to support decision making for patients with CR-GNBIs.
Physician Accreditation Statement
This activity has been planned and implemented in accordance with the accreditation requirements and policies of the Accreditation Council for Continuing Medical Education (ACCME) through the joint providership of Global Education Group (Global) and Applied Clinical Education (ACE). Global is accredited by the ACCME to provide continuing medical education for physicians.
Physician Credit Designation
Global designates this live activity for a maximum of 1.0 AMA PRA Category 1 Credit™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.
Pharmacist Accreditation Statement
Global is accredited by the Accreditation Council for Pharmacy Education (ACPE) as a provider of continuing pharmacy education with Commendation.
Pharmacist Credit Designation
Global designates this continuing education activity for 1.0 contact hour (0.1 CEU) of the ACPE. (Universal Activity Number 0530-9999-24-027-H01-P)
This is an knowledge-based activity.
Global Contact Information
For information about the accreditation of this program, please contact Global at 303-395-1782 or cme@globaleducationgroup.com.
Instructions for Obtaining Credit
To receive credit, participants must participate in the activity, complete and pass the post-test with a minimum score of 70%, and complete the evaluation. CME certificates will be sent via email to those successfully completing the activity.
System Requirements
PC
- 1.4 GHz Intel Pentium 4 or faster processor (or equivalent)
- Windows 10, 8.1 (32-bit/64-bit); Windows 7 (32-bit/64-bit) 512 MB of RAM (1 GB recommended)
- Google Chrome (v70.0 and above), Mozilla Firefox (v65.0 and above), and Edge (v42.0 and above)
Mac
- 1.83 GHz Intel Core Duo or faster processor 512 MB of RAM (1 GB recommended)
- MAC OS X 10.12 or higher
- Google Chrome (v70.0 and above), Apple Safari (v12.0 and above), and Mozilla Firefox (v65.0 and above)
Fee Information and Refund/Cancellation Policy
There is no fee for this educational activity.
Disclosures of Relevant Financial Relationships
Global adheres to the policies and guidelines, including the Standards for Integrity and Independence in Accredited CE, set forth to providers by the Accreditation Council for Continuing Medical Education (ACCME) and all other professional organizations, as applicable, stating those activities where continuing education credits are awarded must be balanced, independent, objective, and scientifically rigorous. All persons in a position to control the content of an accredited continuing education program provided by Global are required to disclose all financial relationships with any ineligible company within the past 24 months to Global. All financial relationships reported are identified as relevant and mitigated by Global in accordance with the Standards for Integrity and Independence in Accredited CE in advance of delivery of the activity to learners. The content of this activity was vetted by Global to assure objectivity and that the activity is free of commercial bias. All relevant financial relationships have been mitigated.
The faculty have the following relevant financial relationships with ineligible companies:
- Consulting fees (eg, advisory boards): Cepheid, Ferring, Melinta, Pfizer, Shionogi
- Speakers’ bureaus: Cepheid, Ferring, Melinta
- Consulting fees (eg, advisory boards): BugWorks Research, Genentech, Micurx Pharmaceuticals, Omnix Medical Ltd, Pulmatrix, Qpex Biopharma, Shionogi, Spero Therapeutics, Venatorx Pharmaceuticals
- Speakers’ bureau: Shionogi
- Other (Independent Data Monitoring Committee): Janssen Pharmaceutica
- Consulting fees (eg, advisory boards): Merck, Pfizer
- Honoraria: Merck, Pfizer
The planners and managers have the following relevant financial relationships with ineligible companies:
The planners and managers at Global Education Group have no relevant financial relationships to disclose.
The planners and managers at ACE have no relevant financial relationships to disclose.
Disclosure of Unlabeled Use
This educational activity may contain discussion of published and/or investigational uses of agents that are not indicated by the FDA. Global and ACE do not recommend the use of any agent outside of the labeled indications. The opinions expressed in the educational activity are those of the faculty and do not necessarily represent the views of any organization associated with this activity. Please refer to the official prescribing information for each product for discussion of approved indications, contraindications, and warnings.
Disclaimer
Participants have an implied responsibility to use the newly acquired information to enhance patient outcomes and their own professional development. The information presented in this activity is not meant to serve as a guideline for patient management. Any procedures, medications, or other courses of diagnosis or treatment discussed in this activity should not be used by clinicians without evaluation of patient conditions and possible contraindications on dangers in use, review of any applicable manufacturer’s product information, and comparison with recommendations of other authorities.
This activity is jointly provided by Global Education Group and Applied Clinical Education.
This activity is supported by an educational grant from Shionogi Inc.
This activity is distributed by Pharmacy Practice News,
Infectious Disease Special Edition, and CMEZone.com.
false
*You must be logged in to see results data.
Acronyms used in this activity
BAT, best available therapy
BLBLI, β-lactam–β-lactamase inhibitor
BSI, bloodstream infection
cIAI, complicated intraabdominal infection
CR, carbapenem-resistant
CRE, carbapenem-resistant Enterobacterales
CLSI, Clinical and Laboratory Standards Institute
cUTI, complicated urinary tract infection
DTR, difficult-to-treat resistance
ESBL, extended-spectrum β-lactamase
EUCAST, European Committee on Antimicrobial Susceptibility Testing
GN, gram-negative
GNB, gram-negative bacteria
GNBI, gram-negative bacterial infection
HAP, hospital-acquired pneumonia
IDSA, Infectious Diseases Society of America
IMP, imipenemase
KPC, Klebsiella pneumoniae carbapenemase
MBL, metallo-β-lactamase
MDR, multidrug-resistant
MEM-NS, meropenem-nonsusceptible Enterobacterales
MIC, minimum inhibitory concentration
NDM, New Delhi metallo-β-lactamase
OR, odds ratio
OXA, oxacillinase
PCR, polymerase chain reaction
SBL, serine β-lactamase
SPM, São Paulo metallo-β-lactamase
VAP, ventilator-associated pneumonia
VIM, Verona integron-encoded metallo-β-lactamase
XDR, extremely drug-resistant
Infections caused by metallo-β-lactamase (MBL)-producing, carbapenem-resistant (CR) gram-negative (GN) bacteria (GNB) pose a significant health threat due to the increasing prevalence of these pathogens and the limited treatment options available.1-9 These pathogens are primarily found among Enterobacterales, Pseudomonas aeruginosa, and Acinetobacter baumannii.9-16 Conventionally, carbapenems represent the gold standard treatment for multidrug-resistant (MDR) GNB infections (GNBIs); however, these agents are rendered ineffective by MBLs given their mechanism of resistance. More specifically, MBLs confer resistance to nearly all β-lactam antibiotics, including carbapenems. This occurs primarily via hydrolysis through zinc-dependent mechanisms.17 The spread of MBLs is facilitated by their presence on mobile genetic elements, which enables rapid dissemination among various bacterial pathogens and across different species of bacteria.17,18 The increase in MBL GNB resistance has generally been driven by the escalating prevalence and complexity of a variety of β-lactamases and carbapenemases, such as those seen in Klebsiella pneumoniae.9-16
The incidence of CR Enterobacterales (CRE) infections has risen in parallel and more than doubled between 2013 and 2020, with MBL-producing strains being major contributors.19 Infections caused by MBL-producing P. aeruginosa are also increasing and are particularly challenging due to their resistance to most β-lactams, including β-lactam–β-lactamase inhibitor (BLBI) combinations.20 Co-production of other β-lactamases that hydrolyze aztreonam further limits treatment options.9,11,12,15,21,22 MBL GNBIs are also associated with high mortality rates, ranging from 30% to 50%, highlighting the urgent need for effective therapies.23
The clinical presentation and severity of infections caused by MBL-producing CR-GNB varies widely, depending on the site of infection and the patient’s underlying health condition. Common examples of these infections include bloodstream infections (BSIs), hospital-acquired pneumonia (HAP), ventilator-associated pneumonia (VAP), complicated urinary tract infections (cUTIs), and complicated intraabdominal infections (cIAIs).24 These infections are often associated with prolonged hospital stays, increased health care costs, and high mortality rates, particularly in critically ill patients.24 The most clinically relevant MBLs include the New Delhi metallo-β-lactamase (NDM), Verona integron-encoded metallo-β-lactamase (VIM), and imipenemase (IMP) types.24,25 Among these, NDM-producing Enterobacterales pose particular difficulties due to their global dissemination and poor clinical outcomes.24
The diagnosis of infections with MBL-producing organisms represents a major challenge to their effective control and treatment. This is a particularly acute issue for clinical microbiology laboratories. Rapid detection of MBL-producing GNB is crucial for guiding early and appropriate treatment, as the mechanism of carbapenem resistance dictates the preferred therapeutic approach.23 Various FDA-approved molecular platforms can identify clinically relevant MBLs and other important β-lactamases. Phenotypic testing methods are widely used, but they lack sensitivity and specificity; molecular methods, including polymerase chain reaction (PCR) and whole-genome sequencing, offer more accurate detection, but they are not universally available in all clinical settings.23,24 Clinical guidelines recommend a combination approach, particularly for severe infections, until antibiotic-susceptibility testing results are available.23
Treatment for MBL-producing GNBIs is as challenging as the identification of these pathogens. Cefiderocol, a siderophore cephalosporin, is the only recently approved antibiotic in the United States with demonstrated in vitro activity against these pathogens.8 Other antibiotics, such as polymyxins, fosfomycin, and tetracyclines, exhibit in vitro activity against MBL-producing GNB but may have suboptimal clinical efficacy. The combination of ceftazidime-avibactam with aztreonam is considered a “bridge” treatment strategy.14,15,21,22 In this combination, ceftazidime-avibactam inhibits extended-spectrum β-lactamases (ESBLs) and K. pneumoniae carbapenemases (KPCs) often co-produced by MBL-producing GNB, allowing aztreonam to bind effectively to bacterial penicillin-binding proteins. Limited data suggest that the ceftazidime-avibactam-aztreonam combination is a potential treatment for patients with MBL-producing GNBIs.14,15,21,26-28 Several other therapeutics are also in clinical development.8,23 In addition to antibiotic treatment, infection prevention and control measures, including active surveillance and antimicrobial stewardship programs, play vital roles in limiting the spread of highly resistant MBL-producing GNBIs.
This activity provides an overview of β-lactamases, focusing on the MBL variants, rapid diagnostic methods for detecting MBL-producing GNBIs, available treatment options for MBL-producing Enterobacterales and P. aeruginosa, and clinical treatment guideline recommendations. It also reviews supporting data for new agents in development with activity against MBL-producing Enterobacterales and P. aeruginosa.
β-lactam antibiotics are widely used to treat bacterial infections and account for 65% of all US prescriptions for injectable antibiotics.29 Although these agents are well tolerated and efficacious, antimicrobial resistance has become a major public health concern. Resistance to penicillin was first detected in 1940, and resistance has emerged with each new generation of antibiotics, exacerbated by the advent of MDR pathogens.23 Various mechanisms may cause β-lactam resistance, including efflux pump expression, mutations in penicillin-binding proteins, alterations to membrane permeability, and the production of β-lactamase enzymes.23 Understanding β-lactamases is essential for clinicians to make informed decisions about antibiotic therapy and infection control.
Bacterial β-lactamases hydrolyze the β-lactam ring of antibiotics, rendering these agents ineffective. These enzymes are now widespread in many GNBs, particularly enteric and nonlactose fermenters, where they collectively confer resistance to β-lactam–containing antibiotics.2 The presence of β-lactamases in GNB includes chromosomally encoded and transferable enzymes that migrate on mobile elements across different species.29 Furthermore, their prevalence and complexity are continually increasing.
The 2 primary classes of bacterial β-lactamases differ structurally and mechanistically but serve the same purpose1,2:
- MBLs use a zinc ion to activate the β-lactam toward nucleophilic attack by a hydroxide anion held between 2 zinc ions.
- Active-site serine β-lactamases (SBLs) use an active serine group as a nucleophile to attack the β-lactamase ring, forming a covalent intermediate that is subsequently hydrolyzed.
Two main classification systems are used for β-lactamases: molecular (Ambler) and functional (Bush-Jacoby-Medeiros; Figure 1).30 In the more commonly used Ambler system, classification is based on the enzymes’ genetic amino acid sequences and mechanisms. β-lactamases in classes A, C, and D contain serine as the enzyme active site, and those in class B contain zinc. In the Bush-Jacoby-Medeiros classification, β-lactamases are categorized into 3 classes according to the β-lactam substrate degradation pattern and inhibitor effects.30 Most MBLs identified in clinical isolates belong to subclass B1 and are named based primarily on where they were first described, with the 3 most common being NDM, VIM, and IMP.1 The B1 subclass is the most clinically relevant, as the β-lactamase genes encoding subclass B1 MBLs are primarily plasmid-borne.1
Carbapenemases are categorized as MBL Ambler class B or SBL functional A or D subgroups. They confer resistance to nearly all β-lactams, including penicillins, cephalosporins, and carbapenems; and are not inhibited by clavulanic acid, sulbactam, tazobactam, avibactam, penicillanic acid sulfones, or diazabicyclooctanes.1,23 Enterobacterales and P. aeruginosa MBLs are associated with extremely drug-resistant (XDR) phenotypes, often including resistance to multiple aminoglycosides, fluoroquinolones, other agents, and β-lactams.1
MBLs can also be broadly viewed as those that are chromosomal in nature and those that are due to plasmid-associated enzymes. Chromosomally encoded MBL genes are typically derived from environmental organisms, whereas acquired MBLs are carried on mobile genetic elements that facilitate their dissemination (Table 1).2,9,32-39 Acquired MBLs in GNB include cfiA, IMP, VIM, São Paulo MBL (SPM), and German IMP. The IMP group includes more than 85 variants, and the VIM-type MBLs include more than 69.1 Co-expression of MBL and ESBL genes, such as blaTEM, blaSHV, and blaOXA, has been reported, complicating the resistance profiles of these bacteria.40
Table 1. Acquired MBLs2,32-3 | |
IMP carbapenemases2 |
|
VIM carbapenemases2 |
|
NDM carbapenemases |
|
ATM, aztreonam; AVI, avibactam; CR, carbapenem-resistant; IMP, imipenemase; MBL, metallo-β-lactamase; NDM, New Delhi metallo-β-lactamase; MDR, multidrug-resistant; VIM, Verona integron-encoded metallo-β-lactamase. |
MBL enzymes are encountered worldwide, with considerable regional variance. Initially confined to health care settings, these pathogens have increasingly been identified in community-acquired infections, raising concerns about their potential for widespread dissemination.
The greatest burden lies in South and Southeast Asia, where NDM types are most prevalent (Figure 2).1 Bacteria with IMP, VIM, and NDM enzymes have been identified in various community, health care, and environmental settings in these regions. Indeed, retrospective analyses by the SENTRY Antimicrobial Surveillance Program suggest that blaNDM-1 was circulating in India as early as 2006 and that this enzyme has since become endemic and co-expressed with other carbapenemases.1,41-43 Transmission of β-lactamase-producing bacterial pathogens depends on multiple factors, including geographic location, population density, hygiene, and antibiotic use.2
In North America, MBLs have remained uncommon, although cases involving VIM, IMP, and NDM have been reported with increasing prevalence since 2010.1 The Antibiotic Resistance Laboratory Ne2rk, initiated by the Centers for Disease Control and Prevention characterizes clinical isolates of CR Enterobacterales, CR P. aeruginosa and CR A. baumannii and conducts colonization screens to detect the presence of mobile carbapenemase genes in North America.44 What the future holds for region regarding the spread of MBLs is unclear. Hence vigilance is required.
In Europe, KPC predominates in Mediterranean countries, whereas NDM producers are most common in Romania, Poland, and Denmark; this is assumed to be caused by inter-regional spread (epidemiologic stage 4).43 VIM is the predominant MBL in Spain, Italy, and Hungary, also associated with inter-regional spread. Data published by the European Centre for Disease Prevention and Control’s European Survey of Carbapenemase-Producing Enterobacteriaceae project in 2017 showed that SBLs were more prevalent than MBLs in most countries.1
MBLs in Meropenem-Resistant Enterobacterales Isolates
The global ATLAS (Antimicrobial Testing Leadership and Surveillance) program estimated the frequency of resistance determinants in Enterobacterales isolates (N=39,368) collected in 2018 and 2019.45 Its report showed that 5.7% of all isolates were meropenem-nonsusceptible (MEM-NS) Enterobacterales, 36.7% of the 2228 identified carbapenem resistance mechanisms were MBLs, and the proportions of KPC or oxacillinase (OXA)-48–like β-lactamases were both approximately 25% (Figure 3).45 Most of the identified MBL genes encoded NDM variants (88.4%), whereas 11.1% and 0.5% encoded VIM and IMP variants, respectively. However, considerable variation among regions was observed, with the proportion of MEM-NS isolates ranging from 1.9% (North America) to 8.4% (Asia/Pacific), and the majority were KPC (71.5%).45
Rapid Diagnostics and Antimicrobial Stewardship
Optimal treatment for CRE infections generally and for MBLs varies considerably, depending on the precise resistance mechanism. Importantly, conventional phenotypic susceptibility testing cannot determine underlying resistance mechanisms, underscoring the need to rapidly detect MBL production in the causative pathogen.23 Several molecular platforms are commercially available to identify MBLs and other important SBLs (Table 2).23 Put simply, one cannot look at a general susceptibility report from the microbiology lab and determine what mechanisms of resistance may be presented in a CR organism.
Table 2. Rapid diagnostic tests relevant to MBL-producing Enterobacterales2 | ||||
Test | Technology | Enterobacterales detected? | Resistance-determinant genes detected | Specimen type |
---|---|---|---|---|
Molecular assays | ||||
Verigene BC-GN | Multiplex PCR and hybridization | Yes | blaNDM, blaVIM, blaIMP, blaCTX-M, blaKPC, blaOXA-48, blaOXA-23, blaOXA-40, blaOXA-58 | Positive blood culture |
Biofire BCID2 Panel | Multiplex PCR | Yes | blaNDM, blaVIM, blaIMP, blaKPC, blaOXA-48-like | Positive blood culture |
GenMark Diagnostics ePlex BCID-GN | Multiplex PCR | Yes | blaNDM, blaVIM, blaIMP, blaCTX-M, blaKPC, blaOXA-23, blaOXA-48 | Positive blood culture |
Biofire FilmArray Pneumonia Panel | Multiplex PCR | Yes | blaNDM, blaVIM, blaIMP, blaKPC, blaOXA-48-like | BAL, sputum |
Unyvero LRT Application | Multiplex PCR | Yes | blaNDM, blaVIM, blaIMP, blaCTX-M, blaKPC, blaTEM, blaOXA-48, blaOXA-23, blaOXA-24, blaOXA-58 | Endotracheal aspirate, BAL |
Cepheid Xpert Carba-R | Qualitative PCR | No | blaNDM, blaVIM, blaIMP, blaKPC, blaOXA-48 | Rectal swabs, pure colony |
BD MAX Check-Points CPO | Qualitative PCR | No | blaNDM, blaVIM/blaIMP, blaKPC, blaOXA-48 | Rectal swabs |
GenePOC Carba | Qualitative PCR | No | blaNDM, blaVIM, blaIMP, blaKPC, blaOXA-48-like | Pure colony |
Biochemical assays | ||||
NG-Test CARBA 5 | Qualitative multiplex immuno-chroma-tographic assay | No | blaNDM, blaVIM, blaIMP, blaKPC, blaOXA-48-like | Pure colony |
Rapidec Carba NP | Colorimetric test | No | None, detects hydrolysis of imipenem | Pure colony |
Other | ||||
MBT STAR-Carba IVD Kit | MALDI-TOF-MS | No | None, detects carbapenem hydrolysis product | Pure colony |
Accelerate PhenoTest BC Kit | Morphokinetic cellular analysis | Yes | None, tests susceptibilities to meropenem (among other antibiotics) | Positive blood culture |
Note: MBL genes bolded. BAL, bronchoalveolar lavage; BC, blood culture; BCID, blood culture identification; CPO, carbapenemase-producing organisms; GN, gram-negative; IVD, in vitro diagnostic; LRT, lower respiratory tract; MALDI-TOF-MS, matrix-assisted laser desorption ionization-time of flight mass spectrometry; MBL, metallo-β-lactamase; PCR, polymerase chain reaction; POC, point of care.
Infection and Drug Resistance. 2021;14:125-142. Originally published by, adapted, and used with permission from Dove Medical Press Ltd. |
Early detection of MBL-producing bacteria allows for the timely initiation of appropriate antimicrobial therapy, avoiding the use of ineffective β-lactam antibiotics.23 This targeted approach can significantly improve outcomes, particularly in critically ill patients. Indeed, rapid diagnostics involving organism identification and genotypic resistance mechanism detection have been shown to decrease mortality in GN BSIs, as have the coordinated efforts of antimicrobial stewardship teams.23,46 A systematic review and meta-analysis demonstrated that mortality rates fell with molecular rapid diagnostic testing vs conventional BSI testing (odds ratio [OR], 0.66).46 Stratified analysis revealed a significantly lower mortality risk when rapid testing was used with antimicrobial stewardship programs (OR, 0.64) but not without such support (OR, 0.72). In addition, time to effective therapy decreased by a weighted mean difference of 5.03 hours with molecular rapid diagnostic testing, and hospital length of stay decreased by 2.48 days.
Timely identification of MBL-producing pathogens enables health care facilities to implement timely infection control, antimicrobial stewardship, and epidemiological surveillance measures to prevent the spread of these highly resistant bacteria within the hospital environment.23,47,48 Awareness of the presence of MBL enzymes helps clinicians make informed decisions about antibiotic use, support antimicrobial stewardship efforts by reducing unnecessary broad-spectrum antibiotic use, and preserve the effectiveness of last-line antibiotics.46-48 Timely MBL detection also aids in tracking the prevalence and spread of MBL-producing organisms, which is essential for public health interventions and policymaking.48
Molecular and Biochemical Assays
Available methods for detecting MBLs can be categorized into molecular and biochemical assays. Several molecular platforms can identify multiple bacterial species and corresponding resistance mechanisms.23 These assays allow for direct testing of blood cultures or respiratory samples and can detect polymicrobial infections and resistance markers. Biochemical assays can detect the presence of carbapenemases, but these are intended for infection control purposes, not treatment guidance.23
Phenotypic Carbapenemase Detection Assays
Rapid confirmation and differentiation of carbapenemase production are crucial for initiating early effective therapy for CR-GNBIs. Recently, an algorithm was developed to guide laboratory-based screening of CR P. aeruginosa, which can guide antimicrobial stewardship efforts and improve the selection of appropriate therapy (Figure 4).47
A recent study evaluated the accuracy of 11 phenotypic assays for detecting carbapenemase-producing glucose-nonfermenting GN bacilli.49 Among 96 CR isolates, 29% produced carbapenemases, with 86% of carbapenemase-producing P. aeruginosa isolates producing class B MBLs, primarily VIM and IMP carbapenemases. Several assays, including all rapid chromogenic assays and the modified carbapenem-inactivation method, had 90% sensitivity for detecting carbapenemase-producing P. aeruginosa, and most tests had 90% specificity (except the Manual Blue Carba assay, Modified Carba NP assay, boronic acid synergy test, and metallo-β-lactamase Etest). Conversely, most tests performed poorly for A. baumannii isolates; although several achieved 90% specificity, none achieved a sensitivity of greater than 90%. Furthermore, some assays were inefficient at identifying MBLs (eg, the Manual Carba NP CLSI; MBL Etest; modified Hodge test; and carbapenem-inactivation method).49
A multicenter study evaluated a qualitative PCR assay (Cepheid Xpert Carba-R) designed to rapidly detect carbapenem resistance genes, including blaKPC, blaVIM, blaNDM, and blaIMP.50 Compared with reference methods, the overall sensitivity was 100% for isolates grown on both blood and MacConkey agars, and overall specificity was 97.1% to 98.1%.50
Immunochromatographic assays can detect MBL enzymes rapidly, and they are simple to perform and interpret, making them suitable for point-of-care use. A study evaluating recombinase-aided amplification directly on clinical samples demonstrated rapid detection of the NDM gene in a single one-step reaction that was comparable to standard PCR.48
Clinical Guideline Recommendations for MBL Detection
Clinical guideline recommendations for the diagnosis and management of infections caused by MDR GN bacilli have been issued by the European Society of Clinical Microbiology and Infectious Diseases (ESCMID; endorsed by the European Society of Intensive Care Medicine) and the Infectious Diseases Society of America (IDSA).51,52 The IDSA guidelines on antimicrobial-resistant infections do not explicitly outline methods for MBL detection; however, they emphasize the importance of accurate and timely identification of resistance mechanisms, including MBLs, to guide appropriate antibiotic therapy.52 The ESCMID guidelines provide more detailed recommendations on the laboratory detection of carbapenemases, including MBLs.51 ESCMID recommends using phenotypic methods, such as combination disk tests, for initial screening of MBL production and molecular methods, such as PCR-based techniques or whole-genome sequencing, to confirm MBL genes. The guidelines also recommend using rapid molecular or immunochromatographic tests to detect carbapenemases, including MBLs, in clinical settings where quick results are needed.51
Conventional treatment options for CR-GNB are limited to antibiotics, such as colistin and tigecycline, often used in combination with other agents. However, these agents have many limitations ranging from tolerability and toxicity issues to the promotion of future resistance. Historically, colistin, a polymyxin antibiotic, was the gold standard for CR-GNB treatment, but this agent is associated with significant nephrotoxicity and neurotoxicity. Tigecycline, conventionally used for various MDR infections, has limitations in treating BSIs due to its low serum concentration and poor lung penetration.53
The following section reviews current treatment options for MBL-producing CR-GNBIs, focusing on newer monotherapies and key clinical trial data. Table 3 summarizes key antimicrobial susceptibility data of different CR Enterobacterales for current CR-GNBI treatment options.54
Table 3. Antimicrobial susceptibility of different carbapenem-resistant Enterobacterales5 | |||||||||
Antibiotic | KPC (n=569) | NDM (n=351) | IMP (n=49) | ||||||
---|---|---|---|---|---|---|---|---|---|
% S | MIC50 | MIC90 | % S | MIC50 | MIC90 | % S | MIC50 | MIC90 | |
Amikacin | 39.7 | 256 | 256 | 79.8 | 4 | 256 | 87.8 | 4 | 256 |
Aztreonam | 1.4 | >256 | >256 | 24.8 | 64 | >256 | 36.7 | 32 | >256 |
Aztreonam-avibactam | 99.5 | 0.5 | 1 | 97.2 | 0.125 | 1 | 98 | 0.125 | 2 |
Cefepime | 0.9 | 128 | 256 | 0.9 | 128 | >256 | 6.1 | 64 | 256 |
Cefiderocol | 99.6 | 2 | 2 | 92 | 2 | 4 | 100 | 1 | 2 |
Cefoperazone-sulbactam | 0.7 | >256 | >256 | 0.9 | >256 | >256 | 8.2 | 256 | >256 |
Cefotaxime | 0.9 | 256 | >256 | 0.3 | >256 | >256 | 6.1 | 256 | >256 |
Cefoxitin | 2.8 | 256 | >256 | 0.9 | >256 | >256 | 8.2 | >256 | >256 |
Ceftazidime | 1.8 | >256 | >256 | 1.1 | >256 | >256 | 6.1 | >256 | >256 |
Ceftazidime-avibactam | 97.4 | 2 | 4 | 0 | >256 | >256 | 0 | >256 | >256 |
Chloramphenicol | 19.3 | 32 | >256 | 51.6 | 8 | >256 | 59.2 | 8 | 256 |
Ciprofloxacin | 2.8 | 64 | 128 | 21.9 | 16 | 128 | 36.7 | 4 | 128 |
Colistin | 96 | 0.125 | 0.5 | 97.7 | 0.125 | 0.5 | 100 | 0.125 | 0.5 |
Ertapenem | 1.1 | 256 | >256 | 0.6 | 32 | 64 | 14.3 | 8 | 256 |
Fosfomycin | 24.8 | 128 | >256 | 83.5 | 16 | 256 | 67.3 | 32 | >256 |
Imipenem | 7 | 16 | 32 | 7.1 | 4 | 16 | 44.9 | 2 | 32 |
Levofloxacin | 3.7 | 32 | 128 | 27.4 | 8 | 64 | 36.7 | 8 | 64 |
Meropenem | 6.9 | 64 | 256 | 6.8 | 8 | 32 | 30.6 | 4 | 64 |
Minocycline | 59.6 | 4 | 32 | 56.1 | 4 | 32 | 59.2 | 2 | 16 |
Piperacillin-tazobactam | 0.9 | >256 | >256 | 2 | >256 | >256 | 36.7 | 256 | >256 |
Tigecycline | 92.3 | 1 | 2 | 93.4 | 0.5 | 2 | 89.8 | 0.5 | 4 |
IMP, imipenemase; KPC, Klebsiella pneumoniae carbapenemase; MIC50/90, minimum inhibitory concentration where 50% or 90% of isolates are inhibited; NDM, New Delhi metallo-β-lactamase; S, susceptible. Reproduced with permission. Open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license. http://creativecommons.org/licenses/by/4.0/ |
Cefiderocol
Cefiderocol is the first siderophore-conjugated cephalosporin approved for adults with cUTIs (including pyelonephritis), HAP, and VAP caused by susceptible GNB.55 It exhibits activity against various GNB, including CR isolates, and is stable against certain carbapenemases, including MBLs such as IMP, VIM, and NDM.56-58 Cefiderocol binds to extracellular free iron and uses bacterial active iron transport channels to penetrate outer cell membranes, allowing it to overcome many GNB resistance mechanisms (Figure 5).59,60 Furthermore, its side-chain properties confer stability against hydrolysis by various β-lactamases, including SBLs and MBLs.
Cefiderocol antibacterial activity was recently evaluated in vitro in 452 MBL-producing strains, including Enterobacterales, P. aeruginosa, and A. baumannii.57 Among MBL-producing Enterobacterales, 91.5% had cefiderocol MIC values ≤4 mg/L according to the Clinical and Laboratory Standards Institute (CLSI) susceptibility breakpoint, and 67.5% had MIC values ≤2 mg/L according to the European Committee on Antimicrobial Susceptibility Testing (EUCAST) susceptibility breakpoint. Among MBL-producing P. aeruginosa, 100% had cefiderocol MIC values ≤4 mg/L (CLSI susceptibility breakpoint), and 97.4% had cefiderocol MIC values ≤2 mg/L (EUCAST susceptibility breakpoint). For A. baumannii complex, MIC values were ≤4 mg/L for 60% of MBL-producing strains and ≤2 mg/L for 44%. For all MBL-producing strains, MIC distribution curves for cefiderocol were in the lowest numerical values vs other β-lactams, BLBLI combinations, and ciprofloxacin.57
Another study evaluated cefiderocol activity and synergy of BLBI-based combinations against Enterobacterales and MEM-NS P. aeruginosa, including MBL-producers.61 It showed that among MBL-producers, cefiderocol in combination with ceftazidime-avibactam showed a synergy rate of 20%, and VIM-producing isolates were almost all susceptible to cefiderocol. Other preclinical data have demonstrated the in vitro activity of cefiderocol against A. baumannii complex and Stenotrophomonas maltophilia, with 96% to 99% susceptibility against clinical isolates in vitro.62
Clinical Data for Cefiderocol
Approval of cefiderocol for cUTIs was based on a study demonstrating superior efficacy in clinical response (73%) compared with imipenem-cilastatin (55%) and microbiological eradication in hospitalized adults.55,63 Approval in HAP/VAP was based on the APEKS-NP trial, which showed noninferiority in all-cause mortality at day 14 vs meropenem in hospitalized adults (12.4% vs 11.6% for meropenem).55,64 Other recent data emerged from the CREDIBLE-CR study, in which cefiderocol performed similarly to BAT in a heterogeneous patient cohort with infections caused by CR-GNB—most frequently A. baumannii, K. pneumoniae, and Pseudomonas spp.65
Together, APEKS-NP and CREDIBLE-CR included 34 patients with infections caused by MBLs (11 and 23, respectively).25 Overall, in this subpopulation vs the comparators, cefiderocol was associated with higher rates of clinical cure and microbiological eradication at the end of therapy, as well as lower day-28 mortality (Figure 6; 70.8% [n=17/24] vs 40% [n=4/10] for a clinical cure; 58.3% [n=14/24] vs 30% [n=3/10] for eradication; and 12.5% [n=3/24] vs 50% [n=5/10] for mortality).19,25 Clinical cure rates were higher for non-NDM vs NDM infections (100% [n=8/8] vs 56.2% [n=9/16]).19 Furthermore, cefiderocol demonstrated superiority in clinical cure and microbiological eradication for isolates containing OXA-48 carbapenemases (n=10).66 Despite the small number of patients, the authors concluded that “the clinical and microbiological outcomes were consistent with the studies’ overall findings of efficacy against Enterobacterales.”66
Real-World Data for Cefiderocol
Several real-world studies corroborate the efficacy of cefiderocol in a range of organisms. PROVE, an ongoing retrospective study reported clinical cure rates in several of the most difficult-to-treat populations treated with cefiderocol.67 Interim results showed cefiderocol achieved clinical cure in 65% of critically ill patients with P. aeruginosa infections, with 81% survival at 30 days. In patients with A. baumannii infections, cefiderocol treatment achieved clinical cure in 60% of cases, with 76% survival at 30 days.67
A cohort analysis in intensive care unit (ICU) patients with P. aeruginosa (55.7%) and A. baumannii (25.7%) respiratory tract infections reported similar outcomes for cefiderocol monotherapy and combination regimens, with 30-day mortality rates of 20% and 28.6%, respectively.68 A prospective study of cefiderocol treatment in 10 patients, mostly with VAP and bacteremia caused by MDR or XDR P. aeruginosa, demonstrated clinical and microbiological cures in 90% and 80% of patients, respectively, with a 90-day mortality rate of 50%.69
The SENTRY program monitors predominant bacterial pathogens and antimicrobial resistance patterns at more than 200 medical centers in 45 countries. A recent report highlights a continuous rise in MDR-GNBIs, including CR isolates, emphasizing the need for effective treatments.70 Importantly, a study analyzing cefiderocol efficacy against GN clinical isolates as part of the SENTRY program indicated cefiderocol potential against serious infections by GN organisms, demonstrating high susceptibility rates among Enterobacterales, P. aeruginosa, Acinetobacter spp, and S. Maltophilia, including CRE, XDR, and BLBI-resistant isolates.71
Plazomicin
Plazomicin, an aminoglycoside, was approved in 2018 for adults with cUTIs, including pyelonephritis, caused by Escherichia coli, K. pneumoniae, E. cloacae, and Proteus mirabilis.72,73 It is recommended for patients with limited or no alternative treatment options due to its activity against aminoglycoside-modifying enzymes and significant in vitro activity against MDR Enterobacteriaceae, including aminoglycoside-resistant isolates.73
A phase 3 open-label study (CARE) evaluated the efficacy and safety of plazomicin vs colistin in combination therapy regimens for serious CR-GNBIs.74 The primary end point, a composite of death from any cause at 28 days or clinically significant disease-related complications, occurred in 24% and 50% of patients treated with plazomicin and colistin, respectively. In a phase 3 study (EPIC) comparing plazomicin vs meropenem in patients with CR cUTIs, plazomicin demonstrated a composite cure rate of 81.7% vs 70.1% with meropenem at 15 to 19 days.75 However, none of the patients in this study had an MBL infection.
That said, plazomicin effectiveness against MBL-producing Enterobacterales is compromised due to 16S rRNA methylation by methylating enzymes. These enzymes, frequently located on transferable plasmids, have been increasingly reported among aminoglycoside-resistant clinical isolates, particularly Enterobacterales-producing NDM and A. baumannii. Isolates with 16S-RMTases show high minimal inhibitory concentration (MIC) values to all legacy aminoglycosides and plazomicin.76
The CR-GNBI treatment landscape continuously evolves with emerging therapies and combinations. Several of these are described below and in Table 4.5,26,77-94
Table 4. Emerging combinations for MBL-producing pathogens5,26,77-9 | |
CZA-AVI |
|
CZA-AVI-ATM |
|
ATM-AVI |
|
CEF-TAN |
|
CEF-ZID |
|
Diazabicyclo- octanes |
|
MER-ANT2681 |
|
MER-QPX7728 |
|
KBP-7072 |
|
LYS228 |
|
ATM, aztreonam; AVI, avibactam; CAP, community-acquired pneumonia; CEF, cefepime; cIAI, complicated intraabdominal infection; CRE, carbapenem-resistant Enterobacteriaceae; cURI, complicated urinary tract infection; CZA, ceftazidime; GNB, gram-negative bacteria; MBL, metallo-β-lactamase; MER, meropenem; NDA, New Drug Application; NDM, New Delhi metallo-β-lactamase; TAN, taniborbactam; ZID, zidebactam. |
Ceftazidime-Avibactam
Ceftazidime-avibactam combines an extended-spectrum anti-pseudomonal cephalosporin with a BLBI. It is effective against a broad range of MBL-producing Enterobacteriaceae, including KPC-producing isolates. A phase 3 study (REPRISE) compared ceftazidime-avibactam to BAT in patients with ceftazidime-resistant Enterobacteriaceae and P. aeruginosa infections.77 It showed similar overall clinical cure rates for ceftazidime-avibactam and BAT (90.9% and 91.2%, respectively). In a prospective observational study (CRACKLE) involving patients with CRE infections, ceftazidime-avibactam treatment was associated with improved survival vs colistin, with 30-day mortality rates of 9% and 32%, respectively.78 Of note, however, this combination has no intrinsic activity against MBLs.
Ceftazidime-Avibactam-Aztreonam
Because many MBL-producing GNs co-express ESBLs, ceftazidime is also given in combination with avibactam and aztreonam. In a prospective study in patients with BSIs mostly due to MBL-producing Enterobacterales, combination treatment with ceftazidime-avibactam-aztreonam was associated with lower 30-day mortality (19.2% vs 44%), lower rates of clinical failure at day 14, and shorter length of stay than other active antibiotics.26 A retrospective study of this combination in 24 patients with P. aeruginosa and MBL VIM-type-producing Enterobacterales infections reported a 30-day mortality of 17% and a 14-day clinical failure rate of 8% (18% and 9% in patients with bacteremia).79 A meta-analysis evaluating ceftazidime-avibactam-aztreonam vs polymyxins in patients with BSIs caused by MBL-CRE found a pooled risk ratio for 30-day mortality of 0.51 (P<0.001).95
A real-world cohort study in 27 hospitalized patients with severe infections caused by GNB with difficult-to-treat resistance (DTR), ceftazidime-avibactam-aztreonam was primarily used for NDM-producing Enterobacterales, with a 33% probability of infection-free survival.78 In comparison, cefiderocol and imipenem-cilastatin-relebactam regimens were primarily used for DTR–nonfermenting infections and were associated with 48% and 67% probabilities of infection-free survival, respectively. In another real-world study, the triple combination achieved clinical success in 6 of 10 patients with K. pneumoniae HUB-ST147 infections caused by an XDR strain producing NDM-1/OXA-48/CTX-M-15, although 2 patients had recurrence within 90days.79 Case reports have also described successful treatment of MBL-producing infections with this combination.15,27
Aztreonam-Avibactam
A single-product formulation of aztreonam-avibactam is in clinical development to streamline the ceftazidime-avibactam-aztreonam combination.23 The European Medicines Agency recently approved aztreonam-avibactam treatment for various indications, including cIAIs, cUTIs, HAP, and infections caused by aerobic GN bacteria, for which treatment options are limited.96
Two phase 3 clinical trials (REVISIT and ASSEMBLE) evaluated aztreonam-avibactam combination treatment for treating serious bacterial infections caused by GNB, including MBLs.97,82,83 In REVISIT, aztreonam-avibactam ± metronidazole demonstrated cure rates of 76.4% for cIAIs and 45.9% for HAP and VAP, which were noninferior to meropenem-colistin (74% and 41.7% for cIAIs and HAP/VAP, respectively).82 In ASSEMBLE, which compared the aztreonam-avibactam combination with BAT in hospitalized adults with infections due to MBLs, reported cure rates were 42% and 0%, respectively.83 However, this trial was terminated due to recruitment challenges.
Cefepime-Taniborbactam
Cefepime-taniborbactam is an investigational combination inhibiting β-lactamase with activity against Enterobacterales spp and P. aeruginosa expressing SBLs and MBLs. In vitro studies demonstrate potent activity against a wide range of resistant phenotypes, including MBL-positive isolates.84 A phase 3 trial in patients with cUTIs, including acute pyelonephritis, demonstrated the superiority of cefepime-taniborbactam to meropenem, with similar safety profiles.85 However, the FDA issued a Complete Response Letter regarding the New Drug Application for cefepime-taniborbactam in February 2024, requesting additional data on chemistry, manufacturing, controls, testing methods, and manufacturing process.86
Cefepime-Zidebactam (WCK 5222)
Zidebactam is a novel β-lactamase inhibitor currently under clinical investigation in combination with cefepime for treating CR and MDR GN infections. Pharmacokinetic/pharmacodynamic studies have shown potent activity against CR Enterobacterales and MDR-producing P. aeruginosa, including MBL-positive isolates.5 Cefepime-zidebactam is currently in phase 3 clinical investigation for treating cUTIs or acute pyelonephritis.
Clinical guideline recommendations for the treatment of infections caused by MDR GN bacilli, as issued by ESCMID and IDSA, are summarized in Table 5.51,52 Both guidelines recommend cefiderocol as the preferred treatment for MBL infections or ceftazidime-avibactam-aztreonam for some MBL-producing organisms.51,52 The main difference between the recommendations is that IDSA offers guidance on P. aeruginosa isolates that are MBL-producing and includes when treatment for MBLs should be considered before confirmation testing.
Table 5. ESCMID and IDSA guidance for treatment of MBLs51,52 | |
ESCMID51 |
|
IDSA52 |
|
AST, antimicrobial susceptibility testing; ATM, aztreonam; AVI, avibactam; CRE, carbapenem-resistant Enterobacterales; CZA, ceftazidime; DTR, difficult-to-treat resistance; ESCMID, European Society of Clinical Microbiology and Infectious Diseases; IDSA, Infectious Disease Society of America; MBL, metallo-β-lactamase; NDM, New Delhi metallo-β-lactamase; UTI, urinary tract infections; VAB, vaborbactam. |
MBL-producing Enterobacterales represent an urgent public health threat that remains a challenge worldwide due to high rates of morbidity and mortality. Rapid diagnostic tests are important to detect MBLs and guide timely and optimal treatment initiation, and numerous FDA-approved platforms are now available. Cefiderocol, a siderophore cephalosporin, is currently the only FDA-approved agent for treating infections caused by MBL-producing CR pathogens. Cefiderocol is stable against hydrolysis by IMP, VIM, and NDM MBLs. Guideline recommendations and clinical data support cefiderocol as a preferred treatment for MBL infections or ceftazidime-avibactam in combination with aztreonam for some MBL-producing organisms. In addition, emerging therapeutics are in clinical development to expand treatment options for MBL-producing Enterobacterales to improve patient outcomes. Antimicrobial stewardship remains essential in preserving the effectiveness of current and emerging therapies and mitigating further resistance development.
Case 1
Roger is a critically ill 62-year-old man in the ICU with VAP and sepsis. He was initially treated with broad-spectrum agents, including ceftolozane-tazobactam. He failed to respond, and subsequent cultures from a lower airway sample revealed XDR/DTR P. aeruginosa. Because of the resistance pattern seen from cultures, cefiderocol was administered in a 3-hour IV infusion at a standard dose of 2 g every 8 hours diluted in 100 mL of normal saline solution. The patient clinically improved and was eventually liberated from the ventilator.
Case 2
Felicia is a 50-year-old woman who developed a catheter-related infection while in the ICU. Despite removal of the catheter, the infection was difficult to eradicate, as it was caused by K. pneumoniae. The isolate was found to produce Oxa48 and NDM-1 carbapenemases. It was susceptible only to amikacin and colistin. Previous treatment regimens included meropenem and amikacin. After the bacteremia failed to clear, and considering the microbiology findings, the patient was switched to a combination of ceftazidime-avibactam (2–0.5 g 3 times daily) and aztreonam (2 g 3 times daily) for 10 days, administered in that order. Blood cultures eventually cleared.
- Boyd SE, Livermore DM, Hooper DC, Hope WW. Metallo-β-lactamases: structure, function, epidemiology, treatment options, and the development pipeline. Antimicrob Agents Chemother. 2020;64(10):e00397-20.
- Bush K, Bradford PA. Epidemiology of β-lactamase-producing pathogens. Clin Microbiol Rev. 2020;33(2):e00047-19.
- Centers for Disease Control and Prevention. Antibiotic resistant threats in the United States 2019. Revised December 2019. Accessed May 31, 2024. www.cdc.gov/antimicrobial-resistance/media/pdfs/2019-ar-threats-report-508.pdf
- World Health Organization. Antibicrobial resistance. November 21, 2023. Accessed May 31, 2024. www.who.int/en/news-room/fact-sheets/detail/antimicrobial-resistance
- Karlowsky JA, Hackel MA, Bouchillon SK, Sahm DF. In vitro activity of WCK 5222 (cefepime-zidebactam) against worldwide collected gram-negative bacilli not susceptible to carbapenems. Antimicrob Agents Chemother. 2020;64(12):e01432-20.
- Lodise TP, Bassetti M, Ferrer R, et al. All-cause mortality rates in adults with carbapenem-resistant gram-negative bacterial infections: a comprehensive review of pathogen-focused, prospective, randomized, interventional clinical studies. Expert Rev Anti Infect Ther. 2022;20(5):707-719.
- Weiner LM, Webb AK, Limbago B, et al. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2011-2014. Infect Control Hosp Epidemiol. 2016;37(11):1288-1301.
- Wright H, Bonomo RA, Paterson DL. New agents for the treatment of infections with gram-negative bacteria: restoring the miracle or false dawn? Clin Microbiol Infect. 2017;23(10):704-712.
- Walsh TR. The emergence and implications of metallo-beta-lactamases in gram-negative bacteria. Clin Microbiol Infect. 2005;11(suppl 6):2-9.
- Jacoby GA, Munoz-Price LS. The new beta-lactamases. N Engl J Med. 2005;352(4):380-391.
- Karlowsky JA, Kazmierczak KM, de Jonge BLM, 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. 2017;61(9):e00472-17.
- Kazmierczak KM, Rabine S, Hackel M, et al. Multiyear, multinational survey of the incidence and global distribution of metallo-beta-lactamase-producing Enterobacteriaceae and Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2015;60:1067-1078.
- Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo-beta-lactamases: the quiet before the storm? Clin Microbiol Rev. 2005;18(2):306-325.
- Wenzler E, Deraedt MF, Harrington AT, Danizger LH. Synergistic activity of ceftazidime-avibactam and aztreonam against serine and metallo-β-lactamase-producing gram-negative pathogens. Diagn Microbiol Infect Dis. 2017;88(4):352-354.
- Davido B, Fellous L, Lawrence C, et al. Ceftazidime-avibactam and aztreonam, an interesting strategy to overcome β-lactam resistance conferred by metallo-β-lactamases in Enterobacteriaceae and Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2017;61(9):e01008-17.
- Singh R, Kim A, Tanudra MA, et al. Pharmacokinetics/pharmacodynamics of a beta-lactam and beta-lactamase inhibitor combination: a novel approach for aztreonam/avibactam. J Antimicrob Chemother. 2015;70(9):2618-2626.
- Berglund F, Johnning A, Larsson DGJ, Kristiansson E. An updated phylogeny of the metallo-β-lactamases. J Antimicrob Chemother. 2021;76(1):117-123.
- Zakhour J, El Ayoubi LW, Kanj SS. Metallo-beta-lactamases: mechanisms, treatment challenges, and future prospects. Expert Rev Anti Infect Ther. 2024;22(4):189-201.
- Timsit JF, Wicky PH, de Montmollin E. Treatment of severe infections due to metallo-betalactamases Enterobacterales in critically ill patients. Antibiotics (Basel). 2022;11(2):144.
- Findlay J, Raro OHF, Poirel L, et al. Molecular analysis of metallo-beta-lactamase-producing Pseudomonas aeruginosa in Switzerland 2022-2023. Eur J Clin Microbiol Infect Dis. 2024;43:551-557.
- Marshall S, Hujer AM, Rojas LJ, et al. Can ceftazidime-avibactam and aztreonam overcome beta-lactam resistance conferred by metallo-beta-lactamases in Enterobacteriaceae? Antimicrob Chemother. 2017;61(4) e02243-16.
- Monogue ML, Abbo LM, Rosa R, et al. In vitro discordance with in vivo activity: humanized exposures of ceftazidime-avibactam, aztreonam, and tigecycline alone and in combination against New Delhi metallo-beta-lactamase-producing Klebsiella pneumoniae in a murine lung infection model. Antimicrob Agents Chemother. 2017;61(7):e00486-17.
- Tan X, Kim HS, Baugh K, et al. Therapeutic options for metallo-β-lactamase-producing Enterobacterales. Infect Drug Resist. 2021;14:125-142.
- Falcone M, Giordano C, Leonildi A, et al. Clinical features and outcomes of infections caused by metallo-β-lactamase-producing Enterobacterales: a 3-year prospective study from an endemic area. Clin Infect Dis. 2024;78(5):1111-1119.
- Timsit JF, Paul M, Shields RK, et al. Cefiderocol for the treatment of infections due to metallo-β-lactamase-producing pathogens in the CREDIBLE-CR and APEKS-NP phase 3 randomized studies. Clin Infect Dis. 2022;75(6):1081-1084.
- Falcone M, Daikos GL, Tiseo G, et al. Efficacy of ceftazidime-avibactam plus aztreonam in patients with bloodstream infections caused by metallo-β-lactamase-producing Enterobacterales. Clin Infect Dis. 2021;72(11):1871-1878.
- Mojica MF, Ouellette CP, Leber A, et al. Successful treatment of bloodstream infection due to metallo-β-lactamase-producing Stenotrophomonas maltophilia in a renal transplant patient. Antimicrob Agents Chemother. 2016;60(9):5130-5134.
- Lodise TP, Smith NM, O’Donnell N, et al. Determining the optimal dosing of a novel combination regimen of ceftazidime/avibactam with aztreonam against NDM-1-producing Enterobacteriaceae using a hollow-fibre infection model. J Antimicrob Chemother. 2020;75(9):2622-2632.
- Bush K, Bradford PA. β-lactams and β-lactamase inhibitors: an overview. Cold Spring Harb Perspect Med. 2016;6(8):a025247.
- Sawa T, Kooguchi K, Moriyama K. Molecular diversity of extended-spectrum β-lactamases and carbapenemases, and antimicrobial resistance. J Intensive Care. 2020;8:13.
- Tooke CL, Hinchliffe P, Bragginton EC, et al. β-lactamases and β-lactamase inhibitors in the 21st century. J Mol Biol. 2019;431(18):3472-3500.
- Findlay J, Poirel L, Kessler J, et al. New Delhi metallo-β-lactamase–producing Enterobacterales bacteria, Switzerland, 2019-2020. Emerg Infect Dis. 2021;27(10):2628-2637.
- Melgarejo JL, Cardoso MH, Pinto IB, et al. Identification, molecular characterization, and structural analysis of the blaNDM-1 gene/enzyme from NDM-1-producing Klebsiella pneumoniae isolates. J Antibiot. 2019;72:155-163.
- Nordmann P, Boulanger AE, Poirel L. NDM-4 metallo-β-lactamase with increased carbapenemase activity from Escherichia coli. Antimicrob Agents Chemother. 2012;56(4):2184-2186.
- Hornsey M, Phee L, Wareham DW. A novel variant, NDM-5, of the New Delhi metallo-β-lactamase in a multidrug-resistant Escherichia coli ST648 isolate recovered from a patient in the United Kingdom. Antimicrob Agents Chemother. 2011;55:5952-5954.
- Aitken SL, Pierce VM, Pogue JM, et al. The growing threat of NDM-producing E. coli with penicillin-binding protein 3 mutations in the United States - is there a potential role for durlobactam? Clin Infect Dis. 2024 Apr 25:ciae229.
- Cuzon G, Bonnin RA, Nordmann P. First identification of novel NDM carbapenemase, NDM-7, in Escherichia coli in France. PLoS One. 2013;8(4):e61322.
- Shao C, Hao Y, Wang Y, et al. Genotypic and phenotypic characterization of blaNDM-7-harboring IncX3 plasmid in a ST11 Klebsiella pneumoniae isolated from a pediatric patient in China. Front Microbiol. 2020;11:576823.
- Tada T, Shrestha B, Miyoshi-Akiyama T, et al. NDM-12, a novel New Delhi metallo-β-lactamase variant from a carbapenem-resistant Escherichia coli clinical isolate in Nepal. Antimicrob Agents Chemother. 2014;58(10):6302-6305.
- Ain NU, Iftikhar A, Bukhari SS, et al. High frequency and molecular epidemiology of metallo-β-lactamase-producing gram-negative bacilli in a tertiary care hospital in Lahore, Pakistan. Antimicrob Resist Infect Control. 2018;7:128.
- Castanheira M, Deshpande LM, Mathai D, et al. Early dissemination of NDM-1- and OXA-181-producing Enterobacteriaceae in Indian hospitals: report from the SENTRY Antimicrobial Surveillance Program (2006-2007). Antimicrob Agents Chemother. 2011;55:1274-1278.
- Johnson AP Woodford N. Global spread of antibiotic resistance: the example of New Delhi metallo-β-lactamase (NDM)-mediated carbapenem resistance. J Med Microbiol. 2013;62:499-513.
- van Duin D, Doi Y. The global epidemiology of carbapenemase-producing Enterobacteriaceae. Virulence. 2017;8(4):460-469.
- Sabour S, Huang JY, Bhatnagar A, et al. Detection and characterization of targeted carbapenem-resistant health care-associated threats: findings from the Antibiotic Resistance Laboratory Network, 2017 to 2019. Antimicrob Agents Chemother. 2021;65(12):e0110521.
- Estabrook M, Muyldermans A, Sahm D, et al. Epidemiology of resistance determinants identified in meropenem-nonsusceptible Enterobacterales collected as part of a global surveillance study, 2018 to 2019. Antimicrob Agents Chemother. 2023;67(5):e0140622.
- Timbrook TT, Morton JB, McConeghy KW, et al. The effect of molecular rapid diagnostic testing on clinical outcomes in bloodstream infections: a systematic review and meta-analysis. Clin Infect Dis. 2017;64(1):15-23.
- Tenover FC, Nicolau DP, Gill CM. Carbapenemase-producing Pseudomonas aeruginosa -an emerging challenge. Emerg Microbes Infect. 2022;11(1):811-814.
- Feng Y, Xue G, Feng J, et al. Rapid detection of New Delhi metallo-β-lactamase gene using recombinase-aided amplification directly on clinical samples from children. Front Microbiol. 2021;12:691289.
- Simner PJ, Opene BNA, Chambers KK, et al. Carbapenemase detection among carbapenem-resistant glucose-nonfermenting gram-negative bacilli. J Clin Microbiol. 2017;55(9):2858-2864.
- Traczewski MM, Carretto E, Canton R, et al. Multicenter evaluation of the Xpert Carba-R assay for detection of carbapenemase genes in gram-negative isolates. J Clin Microbiol. 2018;56(8):e00272-18.
- Paul M, Carrara E, Retamar P, et al. European Society of Clinical Microbiology and Infectious Diseases (ESCMID) guidelines for the treatment of infections caused by multidrug-resistant gram-negative bacilli (endorsed by European Society of Intensive Care Medicine). Clin Microbiol Infect. 2022;28(4):521-547.
- Tamma PD, Aitken SL, Bonomo RA, et al. Infectious Diseases Society of America 2023 guidance on the treatment of antimicrobial resistant gram-negative infections. Clin Infect Dis. 2023 Jul 18:ciad428.
- Doi Y. Treatment options for carbapenem-resistant gram-negative bacterial infections. Clin Infect Dis. 2019;69(suppl 7):S565-S575.
- Wang Q, Jin L, Sun S, et al. Occurrence of high levels of cefiderocol resistance in carbapenem-resistant Escherichia coli before its approval in China: a report from China CRE-Network. Microbiol Spectr. 2022;10(3):e0267021.
- Fetroja (cefiderocol) prescribing information. Florham Park, NJ: Shionogi Inc. November 2021.
- Ito-Horiyama T, Ishii Y, Ito A, et al. Stability of novel siderophore cephalosporin S-649266 against clinically relevant carbapenemases. Antimicrob Agents Chemother. 2016;60:4384-4386.
- Takemura M, Wise MG, Hackel MA, et al. In vitro activity of cefiderocol against MBL-producing gram-negative bacteria collected in North America and Europe in five consecutive annual multinational SIDERO-WT surveillance studies (2014-2019). J Antimicrob Chemother. 2023;78(8):2019-2027.
- Zhanel GG, Golden AR, Zelenitsky S, et al. Cefiderocol: a siderophore cephalosporin with activity against carbapenem-resistant and multidrug-resistant gram-negative bacilli. Drugs. 2019;79:271-289.
- Silva JT, López-Medrano F. Cefiderocol, a new antibiotic against multidrug-resistant gram-negative bacteria. Rev Esp Quimioter. 2021;34(suppl 1):41-43.
- Domingues S, Lima T, Saavedra MJ, Da Silva GJ. An overview of cefiderocol’s therapeutic potential and underlying resistance mechanisms. Life (Basel). 2023;13(7):1427.
- Boattini M, Comini S, Bianco G, et al. Activity of cefiderocol and synergy of novel β-lactam-β-lactamase inhibitor-based combinations against metallo-β-lactamase-producing gram-negative bacilli: insights from a 2-year study (2019-2020). J Chemother. 2023;35(3):198-204.
- Karlowsky JA, Hackel MA, Takemura M, et al. In vitro susceptibility of gram-negative pathogens to cefiderocol in five consecutive annual multinational SIDEROWT surveillance studies, 2014 to 2019. Antimicrob Agents Chemother. 2022;66(2):e0199021.
- Portsmouth S, van Veenhuyzen D, Echols R, 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. 2018;18(12):1319-1328.
- Wunderink RG, Matsunaga Y, Ariyasu M, 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. 2021;21(2):213-225.
- Bassetti M, Echols R, Matsunaga Y, 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. 2021;21:226-240.
- Longshaw C, Roger E, Henriksen AS, et al. Evidence for efficacy of cefiderocol against OXA-48-containing isolates from the APEKS-NP and CREDIBLE-CR trials. Antimicrob Agents Chemother. 2022;e0110022.
- Shionogi & Co, Ltd. ECCMID 2023: Shionogi announces real-world evidence demonstrating the efficacy of Fetcroja® /Fetroja® (cefiderocol) against some of the most difficult-to-treat gram-negative bacterial pathogens. April 17, 2023. Accessed June 12, 2024. www.shionogi.com/us/en/news/2023/04/eccmid-2023-shionogi-announces-real-world-evidence-demonstrating-the-efficacy-of-fetcroja-fetroja-cefiderocol-against-some-of-the-most-difficult-to-treat-gram-negative-bacterial-pathogens.
- El Ghali AE, Kunz-Coyne A, Lucas K, et al. 2118. Comparative analysis of cefiderocol mono- and combination therapy for multi-drug resistant gram-negative infections. Open Forum Infect Dis. 2023;10(suppl 2):ofad500.1741.
- Fendian áM, Albanell-Fernández M, Tuset M, et al. Real-life data on the effectiveness and safety of cefiderocol in severely infected patients: a case series. Infect Dis Ther. 2023;12(4):1205-1216.
- Diekema DJ, Hsueh PR, Mendes RE, et al. The microbiology of bloodstream infection: 20-year trends from the SENTRY Antimicrobial Surveillance Program. Antimicrob Agents Chemother. 2019;63(7):e00355-19.
- Shortridge D, Streit JM, Mendes R et al. In vitro activity of cefiderocol against U.S. and European gram-negative clinical isolates. Collected in 2020 as part of the SENTRY Antimicrobial Surveillance Program. Microbiol Spectr. 2022;10(2):e0271221.
- Zemdri (plazomicin) prescribing information. Warren, NJ: Cipla USA, Inc. Feb 2023.
- Serio AW, Keepers T, Krause KM. Plazomicin is active against metallo-β-lactamase-producing Enterobacteriaceae. Open Forum Infect Dis. 2019;6(4):ofz123.
- McKinnell JA, Dwyer JP, Talbot GH, et al. Plazomicin for infections caused by carbapenem-resistant Enterobacteriaceae. New Engl J Med. 2019;380:791-793.
- Wagenlehner FME, Cloutier DJ, Komirenko AS, et al. Once-daily plazomicin for complicated urinary tract infections. N Engl J Med. 2019;380(8):729-740.
- Costello SE, Deshpande LM, Davis AP, et al. Aminoglycoside-modifying enzyme and 16S ribosomal RNA methyltransferase genes among a global collection of gram-negative isolates. J Glob Antimicrob Resist. 2019;16:278-285.
- Carmeli Y, Armstrong J, Laud PJ, et al. Ceftazidime-avibactam or best available therapy in patients with ceftazidime-resistant Enterobacteriaceae and Pseudomonas aeruginosa complicated urinary tract infections or complicated intra-abdominal infections (REPRISE): a randomised, pathogen-directed, phase 3 study. Lancet Infect Dis. 2016;16(6):661-673.
- van Duin D, Lok JJ, Earley M, et al. Colistin versus ceftazidime-avibactam in the treatment of infections due to carbapenem-resistant Enterobacteriaceae. Clin Infect Dis. 2018;66(2):163-171.
- Sempere A, Viñado B, Los-Arcos I, et al. Ceftazidime- avibactam plus aztreonam for the treatment of infections by VIM-type-producing gram-negative bacteria. Antimicrob Agents Chemother. 2022;66(10):e0075122.
- Larcher R, Laffont-Lozes P, Roger C, et al. Last resort beta-lactam antibiotics for treatment of New-Delhi metallo-beta-lactamase producing Enterobacterales and other difficult-to-treat resistance in gram-negative bacteria: a real-life study. Front Cell Infect Microbiol. 2022;12:1048633.
- Shaw E, Rombauts A, Tubau F, 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. 2018;73(4):1104-1106.
- REVISIT trial. A study to determine the efficacy, safety and tolerability of aztreonam-avibactam (ATM-AVI) ± metronidazole (MTZ) versus meropenem (MER) ± colistin (COL) for the treatment of serious infections due to gram negative bacteria (REVISIT). ClinicalTrials.gov. Updated May 7, 2024. Accessed May 30, 2024. https://classic.clinicaltrials.gov/ct2/show/results/NCT03329092
- ASSEMBLE trial. Efficacy, safety, and tolerability of ATM-AVI in the treatment of serious infection due to MBL-producing gram-negative bacteria (ASSEMBLE). ClinicalTrials.gov. Updated February 2, 2024. Accessed May 31, 2024. https://classic.clinicaltrials.gov/ct2/show/results/NCT03580044
- Karlowsky JA, Hackel MA, Wise MG, et al. In vitro activity of cefepime-taniborbactam and comparators against clinical isolates of gram-negative bacilli from 2018 to 2020: results from the Global Evaluation of Antimicrobial Resistance via Surveillance (GEARS) program. Antimicrob Agents Chemother. 2023;67(1):e0128122.
- Wagenlehner FM, Gasink LB, McGovern PC, et al. Cefepime-taniborbactam in complicated urinary tract infection. N Engl J Med. 2024;390(7):611-622.
- Dall C. FDA rejects new drug application for cefepime-taniborbactam. University of Minnesota Center for Infectious Disease Research & Policy. February 27, 2024. Accessed May 5, 2024. www.cidrap.umn.edu/antimicrobial-stewardship/fda-rejects-new-drug-application-cefepime-taniborbactam
- Livermore DM, Mushtaq S, Warner M, et al. In vitro activity of cefepime/zidebactam (WCK 5222) against gram-negative bacteria. J Antimicrob Chemother. 2017;72;1373-1385.
- Li X, Zhao J, Zhang B, et al. Drug development concerning metallo-β-lactamases in gram-negative bacteria. Front Microbiol. 2022;13:959107.
- Mushtaq S, Vickers A, Woodford N, et al. Activity of nacubactam (RG6080/OP0595) combinations against MBL-producing Enterobacteriaceae. J Antimicrob Chemother. 2019:74;953-960.
- Zalacain M, Lozano C, Llanos A, et al. Novel specific metallo-β-lactamase inhibitor ANT2681 restores meropenem activity to clinically effective levels against NDM-positive Enterobacterales. Antimicrob Agents Chemother. 2021;65(6):e00203-21.
- Lomovskaya O, Rubio-Aparicio D, Nelson K, et al. In vitro activity of the ultrabroad-spectrum beta-lactamase inhibitor QPX7728 in combination with multiple beta-lactam antibiotics against Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2021;65(6):e00210-21.
- Lepak AJ, Zhao M, Liu Q, et al. Pharmacokinetic/pharmacodynamic evaluation of a novel aminomethylcycline antibiotic, KBP-7072, in the neutropenic murine pneumonia model against Staphylococcus aureus and Streptococcus pneumoniae. Antimicrob Agents Chemother. 2019;63(3):e02404-18.
- Huband MD, Mendes RE, Pfaller MA, et al. In vitro activity of KBP-7072, a novel third-generation tetracycline, against 531 recent geographically diverse and molecularly characterized Acinetobacter baumannii species complex isolates. Antimicrob Agents Chemother. 2020;64(5):e02375
- Blais J, Lopez S, Li C, et al. In vitro activity of LYS228, a novel monobactam antibiotic, against multidrug-resistant enterobacteriaceae. Antimicrob Agents Chemother. 2018;62(10):e00552-18.
- Gupta N, Boodman C, Prayag P, et al. Ceftazidime-avibactam and aztreonam combination for carbapenem-resistant Enterobacterales bloodstream infections with presumed metallo-β-lactamase production: a systematic review and meta-analysis. Expert Rev Anti Infect Ther. 2024;22(4):203-209.
- European Medicines Agency. New antibiotic to fight infections caused by multidrug-resistant bacteria (press release). March 22, 2024. Accessed May 30, 2024. www.ema.europa.eu/en/news/new-antibiotic-fight-infections-caused-multidrug-resistant-bacteria
- Pfizer. Phase 3 studies of Pfizer’s novel antibiotic combination offer new treatment hope for patients with multidrug-resistant infections and limited treatment options (press release). June 1, 2023. Accessed May 31, 2024. www.pfizer.com/news/press-release/press-release-detail/phase-3-studies-pfizers-novel-antibiotic-combination-offer
false
*You must be logged in to see results data.
false