Preprint
Review

This version is not peer-reviewed.

β-Lactam/β-Lactamase Inhibitor Combination Antibiotics Under Development

A peer-reviewed article of this preprint also exists.

Submitted:

18 December 2024

Posted:

19 December 2024

You are already at the latest version

Abstract
Antimicrobial resistance remains a public health problem of global concern with a great health and financial burden. Its recognition as a threat by political leadership has boosted the research and development of new antibiotics and particularly novel combinations of β-lactams/β-lactamase inhibitors against multidrug-resistant (MDR) Gram-negative pathogens which remain the major concern in clinical practice. The incorporation of ceftolozane/tazobactam, ceftazidime/avibactam, meropenem/vaborbactam, and imipenem/cilastatin/relebactam has provided new therapeutic options in the treatment of patients with infections due to MDR pathogens. Cefiderocol along with cefepime/enmetazobactam, avibactam/aztreonam, and sulbactam/durlobactam have been recently added to these agents as therapeutic choices, particularly for metallo-β-lactamase producing Gram-negative bacteria. Currently, many combinations are being studied for their in vitro activity against both serine- and metallo-β-lactamases. However, only a few have advanced through phase 1, 2, and 3 clinical trials. Among them, in this article, we focus on the most promising combinations of cefepime/zidebactam, cefepime/taniborbactam, and imipenem/cilastatin/funobactam, which are currently under investigation in phase 3 trials.
Keywords: 
;  ;  ;  ;  ;  ;  ;  

1. Introduction

Antimicrobial resistance (AMR) has been recognized as a global public health problem by the World Health Organization (WHO) [1]. A recent meta-analysis showed that AMR was associated with almost 5 million deaths in 2019; among these, 1.27 million were directly attributed to bacterial resistance [2]. Particularly, multidrug resistance (MDR) in Gram-negative bacteria, which is mostly mediated by β-lactamases, is a major problem in clinical practice. The difficulty in combating MDR Gram-negative bacteria is largely attributed to their distinct cell wall structure compared to Gram-positive bacteria, which impedes antibiotic penetration. Gram-positive pathogens lack the outer membrane found in Gram-negative bacteria, which contains lipopolysaccharides and functions as a barrier to antibiotic penetration [3].
The impact of AMR is not limited to human health [4]. Along with mortality and morbidity, AMR brings a great economic burden according to data from a recent meta-analysis in middle and high-income countries, the healthcare cost associated with drug-resistant infections can vary from $2,371 to $29,289 [4]. In a study of the United States of America's national estimates of healthcare costs associated with AMR in hospitalized patients with bacterial infections, considerably high costs were especially attributable to methicillin-resistant Staphylococcus aureus [$30 998 (95% confidence intervals $25 272-$36 724)] and carbapenem-resistant Acinetobacter baumannii [$74 306 (95% confidence intervals $20 377-$128 235)] infections [5]. In 2017, the World Bank estimated that by 2050, AMR could reduce global gross domestic product by 3.8% each year and push 28 million people into poverty. Losses resulting from the impact of drug resistance on livestock could cost global gross domestic product (GDP) up to $950 billion, while the spread of resistant pathogens from livestock to humans could cost up to $5.2 trillion [1]. The recognition of the problem led the American presidency a decade ago to characterize AMR in general as a “threat to public health and economy”, and the fight against it as “a national security priority” [6]; so, new antibiotic development became one of the US government goals for AMR management [7].
The progress in research has brought to the fore of clinical practice new antibiotics, including β-lactam/β-lactamase inhibitor (BL/BLI) combinations, specifically ceftolozane/tazobactam, ceftazidime/avibactam, meropenem/vaborbactam, imipenem/cilastatin/relebactam [6].. Cefepime/enmetazobactam [8], sulbactam/durlobactam [9], and aztreonam/avibactam are among the recently-approved BL/BLI combinations for use in clinical practice. Drug development has not remained in the realm of BL/BLI combinations, and novel cephalosporins have been incorporated in the pharmaceutical arsenal following U.S. Food and Drug Administration (FDA) approval of cefiderocol in 2019 [6] and of ceftobiprole medocaril sodium earlier this year [10]. Despite the progress and innovation, MDR Gram-negative pathogens remain a significant public health concern and are listed on the WHO Bacterial Priority Pathogens List for 2024 [11].
β-lactam antibiotics are the largest class of antibiotics that is further subdivided into the penicillins, caphalosporins, carbapenems, and monobactams. They bind to and inactivate the transpeptidase domain of penicillin-binding proteins (PBPs) and thus inhibit bacterial cell wall synthesis [12]. The most common mechanism of resistance of Gram-negative bacteria to β-lactams (BL) is through the expression of β-lactamases, which hydrolyze the amide bond within the β-lactam ring leading to antibiotic inactivation. β-lactamases are structurally subdivided into four Ambler classes (Class A, B, C, and D) [12]. Functionally, classes A, C, and D hydrolyze BLs via nucleophilic attack through a conserved serine residue and are thus termed serine-β-lactamases. Class B, on the other hand, requires Zn2+ for BL hydrolysis and are termed metallo-β-lactamases (MBLs) [12]. The most difficult-to-treat Gram-negative pathogens e.g. Pseudomonas aeruginosa [13] and Acinetobacter spp.[14] express extended-spectrum β-lactamases (ESBL) e.g. AmpC-producing Enterobacteriaceae, and/or KPC- or OXA-like carbapenemases e.g. carbapenem-resistant Enterobacteriaceae (CRE).
In this article, we aimed to focus on BL/BLI combination antibiotics under investigation in clinical trials of phases 1, 2, and 3. However, at the time of the writing of this article (11/2024), there were no BL/BLI combination antibiotics at the stage of development of phase 2 clinical trials (with published results) thus, we included relevant agents in the phase 1 and 3 clinical trials.
To provide a comprehensive overview, we compiled the information into three tables. Table 1 presents BL/BLI combination antibiotics currently under investigation in phase 1 trials, while Table 2 includes those in phase 3 trials. Table 3 presents a detailed insight into the antibiotic class, mechanism of action and antimicrobial spectrum of these agents.

2. β-Lactam/β-Lactamase Inhibitor Combination Antibiotics in Phase 3 Trials

2.1. Cefepime/Zidebactam

Cefepime/zidebactam is one of the combinations of BL/BLI under development. Cefepime, a fourth-generation cephalosporin [15], has a broad-spectrum activity against Gram-positive and Gram-negative bacteria; it is used for complicated urinary tract infections (cUTI), intra-abdominal infections, respiratory tract infections, and neutropenic fever [16]. Cefepime alone retains activity against AmpC-producing Gram-negative pathogens [17]. Zidebactam belongs to a new β-lactamase inhibitor category (along with avibactam and relebactam) known as diazabicyclooctanes (DBO) [18]. In particular, zidebactam is an ‘enhancer’ that binds with high affinity to penicillin-binding protein 2 (PBP2) and inhibits β-lactamases, thereby preventing hydrolysis of cefepime and enhancing its antimicrobial activity [10,14]. The combination of these two agents has proven in vitro activity against Enterobacteriaceaeand Pseudomonas aeruginosa that produce β-lactamases, including extended-spectrum β-lactamases (ESBL), Klebsiella pneumoniae carbapenemase (KPC), and metallo-β-lactamase (MBL) [20]. However, the in vitro antimicrobial activity of cefepime-zidebactam against Acinetobacter baumannii [21], Stenotrophomonas maltophilia, Proteus species, and Serratia seems limited [18]. Interestingly, zidebactam improved cefepime pharmacodynamics [22] in vivo and the combination effectively reduced carbapenem-resistant Acinetobacter baumannii burden in neutropenic murine lung [23] and thigh [24]infection models.
Cefepime/zidebactam has been reported to be effective in treating patients with extensively drug-resistant Pseudomonas aeruginosa infections under compassionate use as salvage treatment [25,26]. More specifically, its use concerned the case of a young adult suffering from acute T-cell leukemia and disseminated infection from extensively drug-resistant (XDR) Pseudomonas aeruginosa producing New Delhi Metallo-β-Lactamase (NDM) [25]. The isolate was resistant to the combinations of ceftolozane/tazobactam, ceftazidime/avibactam, and carbapenems, yet susceptible to colistin (polymyxin E). The patient was treated with a combination of polymyxin B and meropenem [25]. However, the clinical deterioration with necrotizing ecthyma gangrenosum and lung involvement, along with the polymyxin B-induced neurotoxicity led to the cefepime/zidebactam use as a last-resort treatment; the prolonged antibiotic administration along with surgical source control resulted in gradual clinical improvement [25]. Another female patient with a history of bariatric surgery suffering from multi-organ dysfunction after intra-abdominal infection with XDR Pseudomonas aeruginosa expressing NDM was successfully treated with the new combination after polymyxin failure [26].
Currently, the combination is being investigated in a phase 3, randomized, double-blind clinical trial (NCT04979806) expected to be completed by the end of 2024. It is a multicenter, non-inferiority trial comparing cefepime/zidebactam against meropenem in patients hospitalized for cUTI or acute pyelonephritis. However, pharmacokinetic data in healthy adults showed that plasma and lung concentrations of this drug combination could also support its use for nosocomial pneumonia by susceptible pathogens [27]. Additionally, its use has a reported safety in patients with renal impairment as long as there is a dose adjustment [15].

2.2. Cefepime/Taniborbactam

Taniborbactam is a boronic-acid-containing β-lactamase inhibitor of β-lactamases of class A, C, and D, as well as some of class B (including VIM, NDM, SPM-1, GIM-1, but not IMP) [28]. The combination of the fourth-generation cephalosporin with the inhibitor provides an extended in vitro activity against carbapenem-resistant Enterobacteriaceae(CRE) and Pseudomonas aeruginosa (CRPA), either isolates producing carbapenemase or non-producing, as well as against isolates with resistance to the novel combinations (ceftolozane/tazobactam, meropenem/varorbactam, ceftazidime/avibactam) [29]. It also exhibited activity against Pseudomonas aeruginosa resistant to meropenem, ceftazidime-avibactam, ceftolozane/tazobactam, and meropenem/varobactam, as well as MDR and difficult-to-treat resistant (DTR) isolates. DTR refers to isolates resistant to fluoroquinolones and β-lactams, excluding the newer BL/BLI ceftazidime/avibactam, ceftolozane/tazobactam, meropenem/varobactam [30].
The combination has exhibited in vivo activity against Enterobacteriaceae, Pseudomonas aeruginosa, and S. maltophilia in murine models of cUTI [31]. A relevant study also demonstrated the in vivo activity against Enterobacteriaceae and Pseudomonas aeruginosa that were not susceptible to the cephalosporin alone in the pneumonia murine model [32]. Among the novel BL/BLI combinations under investigation, cefepime/taniborbactam is the most studied. The positive results from a phase 3 trial led the pharmaceutical company to apply for drug approval. However, the FDA rejected the company's application in February 2024 [33]. The results from this randomized, non-inferiority trial comparing the combination vs. meropenem for the treatment of cUTI were recently published [34]. Cefepime/taniborbactam was proven to be superior to meropenem in terms of microbiologic and clinical success in patients with Gram-negative pathogens susceptible to both agents of the study (70.6% in the cefepime/taniborbactam group vs. 58% of meropenem-treated, 95% CI, 3.1-22.2; p=0.009) [34]. Adverse events were reported at a similar frequency in the combination-treated patients (35.5% vs. 29%) [34].

2.3. Imipenem/Cilastatin/Funobactam

Funobactam is a serine-β-lactamase inhibitor (in the past known as XNW4107) with a spectrum against β-lactamases of class A, C, and D [35]. Its co-administration with imipenem broadens the activity against Acinetobacter baumannii and Klebsiella pneumoniae resistant to carbapenems; funobactam enhances the activity of imipenem against the above bacteria (previously resistant to imipenem) in vitro and in vivo in mouse models [36]. Currently, two randomized, phase 3 trials are in progress. The first one investigates the role of the intravenous combination of imipenem-cilastatin/funobactam vs. meropenem for cUTI in hospitalized adults (NCT05204368) [37]. The second one evaluates the efficacy of imipenem/cilastatin/funobactam against imipenem/cilastatin/relebactam for the treatment of hospital-acquired pneumonia including ventilator-associated (NCT05204563) [38].

3. B-Lactams/B-Lactamase Inhibitors in Phase 1 Trials

3.1. Meropenem/Nacubactam

Nacubactam is the fourth agent of the bridged DBO β-lactamase inhibitors [39]. When used alone, it has proven effectiveness against Gram-negative bacteria; its activity may be broadened against Enterobacteriaceae producing ESBL, KPC, MBL, AmpC, and OXA-48 when the inhibitor is combined with β-lactams [39]. Consequently, this antimicrobial activity is attributed both to the direct impact on pathogens by targeting PBP2 and concurrently, by enhancing the action of the second β-lactam agent on PBP3 [40]. Additional activity of the combination against Pseudomonas yet not Acinetobacter is supported according to a multi-center study trying to determine the in vitro activity against GNB [28].
When the combination was tested in neutropenic mice, its concentration in the human-simulated epithelial lining fluid was effective against Enterobacteriaceae producing class A serine carbapenemases; the combination was superior to either agent alone in terms of bacterial density decline [41]. Similarly, the meropenem/nacubactam combination was more efficacious than either agent alone in reducing MDR Enterobacteriaceae isolates in neutropenic mice with cUTI [42] supporting potential clinical use in cUTI
The pharmacokinetics of the co-administration of the two drugs for up to 2 weeks has also been studied in a non-randomized trial (NCT03174795) in patients with cUTI [43]; yet the results have not been publicly announced. In 2020, the results from a phase 1 clinical trial showed that nacubactam alone or in combination with meropenem was well tolerated in healthy participants; the adverse reactions were mostly apparent after the intravenous administration of nacubactam [44].

3.2. Xeruborbactam/β-Lactams

Xeruborbactam (previously known as QPX7728) is a cyclic boronate inhibiting both serine-β-lactamases and metallo-β-lactamases [45]. This BLI alone has a broader activity spectrum compared to the second dual inhibitor - taniborbactam - since it is effective against MBL including IMP [46]. In vitro, xeruborbactam has been proven to be effective against MBL-producing Enterobacteriaceae[45]; when combined with meropenem was more potent than cefepime/taniborbactam against MBL-negative CRE (MIC90=1 μg/ml vs MIC90=16 μg/ml) [45]. The drug safety in combination with meropenem has been tested in a phase 1 trial of which the results were announced in late 2022 [47]. The trial showed the drug safety and tolerance in healthy adults when administered either alone or in combination with meropenem; the authors pointed out that its favorable pharmacokinetics may provide the potential for co-administration with other β-lactams [47].
To date, no other clinical trials have been published regarding this combination. Currently, two phase 1 trials are testing the pharmacokinetics of xeruborbactam in combination with other β-lactams as shown in Table 1. The first one is a double-blind randomized controlled trial concerning its administration with cefiderocol (NCT06547554) [48]. The second one is about the administration of xeruborbactam oral prodrug combined with ceftibuten (NCT06079775) [49]. Finally, another pharmacokinetics trial concerning xeruborbactam oral prodrug with ceftibuten in patients with varying degrees of kidney impairment is expected to start in the coming months (NCT06157242) [50].

3.3. Meropenem/Pralurbactam

Pralurbactam (also known as FL058) is another novel DBO active against β-lactamases of class A, C, and D [51]. The first trial testing the safety of the combination of meropenem/pralurbactam in healthy subjects was recently published, showing the combination safety and tolerability. The most common adverse events concern the gastrointestinal ones, such as nausea and vomiting [51]. A phase 3, randomized, double-blind, multicenter, positive control trial aimed at comparing the efficacy, safety, and pharmacokinetics of meropenem/pralurbactam to that of ceftazidime/avibactam/metronidazole in the treatment of adult complicated intra-abdominal infections is reported to start this year (NCT06633718).
Other BL/BLI being investigated in phase 1 trials, either completed with the results pending to be publicly announced or in progress, are depicted in Table 1.

4. Discussion

Antimicrobial resistance remains a global public health problem as WHO has pointed out (1) with a great impact in terms of health and financial burden. In clinical practice, the combinations of ceftolozane/tazobactam, ceftazidime/avibactam, meropenem/vaborbactam, and imipenem/cilastatin/relebactam have provided and expanded the therapeutic choices against most of the classes of β-lactamases. However, the choices are limited when it comes to class B β-lactamases (MBL), making the development of effective antibiotics still an unmet need. The only options in our pharmaceutical armamentarium for MBL-producing Enterobacteriaceaeare the combination of ceftazidime/avibactam with aztreonam and the newer cefiderocol [52]. Meanwhile, the novel combination of avibactam/aztreonam has been recently approved by the European Medicines Agency for hospital-acquired pneumonia, complicated urinary tract infections, and intra-abdominal infections by Gram-negative pathogens [53] covering additionally MBL-producing bacteria [54]. Combinations such as cefepime/zidebactam, meropenem/nacubactam, and cefepime/taniborbactam may exhibit potential activity against Gram-negative bacteria producing β-lactamases, including MBL [28]. Among these, cefepime/taniborbactam has reached closer to approval after the positive results of a phase 3 trial were published for the treatment of complicated urinary tract infections [31,32].
β-lactamases are enzymes hydrolyzing the β-lactam ring resulting in resistance to β-lactams [55]. The enzymes are further classified into classes A, B, and D which are serine lactamases, and class B which are metallo-β-lactamases (MBL) [55]. The zinc ion is a prerequisite for MBL activity since it opens the β-lactam ring by activating a water molecule [56], inducing particularly a carbapenemase function [57]. The rapid appearance of new variants, the gene transferability of encoding genes, and the different structure from serine-β-lactamases (49) contribute to the difficulty in managing infections by MBLs. Consequently, the development of agents with potential activity against MBL remains of particular interest. The drug evolution has led us to the promising incorporation of the third generation of β-lactamase inhibitors, known as boronates compounds, such as taniborbactam [57]. Interestingly, considering the crucial role of zinc ion in MBL activity, chelating agents such as Aspergillomarasmine A may play a role in MBL management [58].
BL/BLI are still the leading antibiotic class targeting the pathogens that have been prioritized by WHO [11]. However, the research should be intensified to enlighten the exact underlying mechanism of action of the enzymes responsible for antibiotic resistance. In addition, among the questions to be answered are the variable effectiveness of BL/BLI (such as boronates) against the different MBLs, to provide crucial information for further innovation. The in-depth knowledge along with antibiotic stewardship targeting the rational use of antibiotics can be the leading edge to the battle against multidrug-resistant pathogens.

Supplementary Materials

The following supporting information can be downloaded at the website of this paper posted on Preprints.org.

Conflicts of Interest

None for all authors.

Abbreviations List

  • AMR: Antimicrobial resistance
  • BL/BLI: β-lactam/ β-lactamase inhibitors
  • cUTI: Complicated urinary tract infections
  • ESBL: Extended-spectrum β-lactamases
  • GIM-1: German imipenemase-1
  • IMP: Imipenemase metallo-β-lactamase
  • KPC: Klebsiella pneumoniae carbapenemase
  • MBL: Metallo-β-lactamase
  • MDR: multidrug resistance
  • NDM: New Delhi Metallo-β-Lactamase
  • SPM-1: São Paulo metallo-beta-lactamase-1
  • VIM: Verona integron-encoded metallo-β-lactamase
  • WHO: World Health Organization
  • XDR: extensively drug-resistant

References

  1. World Health Organization. Antimicrobial resistance [Internet]. 2023. Available from: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance.
  2. Murray CJL, Ikuta KS, Sharara F, Swetschinski L, Robles Aguilar G, Gray A, et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. The Lancet. 2022 Feb;399(10325):629–55. [CrossRef]
  3. Exner M, Bhattacharya S, Christiansen B, Gebel J, Goroncy-Bermes P, Hartemann P, et al. Antibiotic resistance: What is so special about multidrug-resistant Gram-negative bacteria? GMS Hygiene and Infection Control; 12:Doc05 [Internet]. 2017 Apr 10 [cited 2024 Oct 9]; Available from: http://www.egms.de/en/journals/dgkh/2017-12/dgkh000290.shtml.
  4. Poudel AN, Zhu S, Cooper N, Little P, Tarrant C, Hickman M, et al. The economic burden of antibiotic resistance: A systematic review and meta-analysis. Karunasagar I, editor. PLoS ONE. 2023 May 8;18(5):e0285170. [CrossRef]
  5. Nelson RE, Hatfield KM, Wolford H, Samore MH, Scott RD, Reddy SC, et al. National Estimates of Healthcare Costs Associated With Multidrug-Resistant Bacterial Infections Among Hospitalized Patients in the United States. Clin Infect Dis. 2021 Jan 29;72(Suppl 1):S17–26. [CrossRef]
  6. Barbier F, Hraiech S, Kernéis S, Veluppillai N, Pajot O, Poissy J, et al. Rationale and evidence for the use of new beta-lactam/beta-lactamase inhibitor combinations and cefiderocol in critically ill patients. Ann Intensive Care. 2023 Jul 18;13(1):65. [CrossRef]
  7. Webster P. US tries to stem antimicrobial resistance. CMAJ. 2014 Nov 4;186(16):1207–1207. [CrossRef]
  8. Keam SJ. Cefepime/Enmetazobactam: First Approval. Drugs. 2024 Jun;84(6):737–44. [CrossRef]
  9. Keam SJ. Sulbactam/Durlobactam: First Approval. Drugs. 2023 Sep;83(13):1245–52. [CrossRef]
  10. Beninger P. Ceftobiprole Medocaril Sodium. Clinical Therapeutics. 2024 Aug;46(8):659–60. [CrossRef]
  11. World Health Organization. 2024 WHO Bacterial Priority Pathogens List [Internet]. World Health Organization; 2024. Available from: https://www.who.int/publications/i/item/9789240093461.
  12. Krco S, Davis SJ, Joshi P, Wilson LA, Monteiro Pedroso M, Douw A, et al. Structure, function, and evolution of metallo-β-lactamases from the B3 subgroup—emerging targets to combat antibiotic resistance. Front Chem. 2023 Jun 20;11:1196073.
  13. Glen KA, Lamont IL. β-lactam Resistance in Pseudomonas aeruginosa: Current Status, Future Prospects. Pathogens. 2021 Dec 18;10(12):1638.
  14. Černiauskienė K, Dambrauskienė A, Vitkauskienė A. Associations between β-Lactamase Types of Acinetobacter baumannii and Antimicrobial Resistance. Medicina. 2023 Jul 28;59(8):1386.
  15. Preston RA, Mamikonyan G, DeGraff S, Chiou J, Kemper CJ, Xu A, et al. Single-Center Evaluation of the Pharmacokinetics of WCK 5222 (Cefepime-Zidebactam Combination) in Subjects with Renal Impairment. Antimicrob Agents Chemother. 2019 Jan;63(1):e01484-18. [CrossRef]
  16. Pais GM, Chang J, Barreto EF, Stitt G, Downes KJ, Alshaer MH, et al. Clinical Pharmacokinetics and Pharmacodynamics of Cefepime. Clin Pharmacokinet. 2022 Jul;61(7):929–53. [CrossRef]
  17. Meini S, Tascini C, Cei M, Sozio E, Rossolini GM. AmpC β-lactamase-producing Enterobacterales: what a clinician should know. Infection. 2019 Jun;47(3):363–75. [CrossRef]
  18. Livermore DM, Mushtaq S, Warner M, Vickers A, Woodford N. In vitro activity of cefepime/zidebactam (WCK 5222) against Gram-negative bacteria. Journal of Antimicrobial Chemotherapy. 2017 May;72(5):1373–85. [CrossRef]
  19. Moya B, Barcelo IM, Bhagwat S, Patel M, Bou G, Papp-Wallace KM, et al. WCK 5107 (Zidebactam) and WCK 5153 Are Novel Inhibitors of PBP2 Showing Potent “β-Lactam Enhancer” Activity against Pseudomonas aeruginosa, Including Multidrug-Resistant Metallo-β-Lactamase-Producing High-Risk Clones. Antimicrob Agents Chemother. 2017 Jun;61(6):e02529-16.
  20. Sader HS, Rhomberg PR, Flamm RK, Jones RN, Castanheira M. WCK 5222 (cefepime/zidebactam) antimicrobial activity tested against Gram-negative organisms producing clinically relevant β-lactamases. Journal of Antimicrobial Chemotherapy. 2017 Jun;72(6):1696–703. [CrossRef]
  21. Yang Y, Guo Y, Yin D, Zheng Y, Wu S, Zhu D, et al. In Vitro Activity of Cefepime-Zidebactam, Ceftazidime-Avibactam, and Other Comparators against Clinical Isolates of Enterobacterales , Pseudomonas aeruginosa, and Acinetobacter baumannii: Results from China Antimicrobial Surveillance Network (CHINET) in 2018. Antimicrob Agents Chemother. 2020 Dec 16;65(1):e01726-20. [CrossRef]
  22. Bhagwat SS, Periasamy H, Takalkar SS, Palwe SR, Khande HN, Patel MV. The Novel β-Lactam Enhancer Zidebactam Augments the In Vivo Pharmacodynamic Activity of Cefepime in a Neutropenic Mouse Lung Acinetobacter baumannii Infection Model. Antimicrob Agents Chemother. 2019 Apr;63(4):e02146-18. [CrossRef]
  23. Avery LM, Abdelraouf K, Nicolau DP. Assessment of the In Vivo Efficacy of WCK 5222 (Cefepime-Zidebactam) against Carbapenem-Resistant Acinetobacter baumannii in the Neutropenic Murine Lung Infection Model. Antimicrob Agents Chemother. 2018 Nov;62(11):e00948-18. [CrossRef]
  24. Almarzoky Abuhussain SS, Avery LM, Abdelraouf K, Nicolau DP. In Vivo Efficacy of Humanized WCK 5222 (Cefepime-Zidebactam) Exposures against Carbapenem-Resistant Acinetobacter baumannii in the Neutropenic Thigh Model. Antimicrob Agents Chemother. 2019 Jan;63(1):e01931-18. [CrossRef]
  25. Tirlangi PK, Wanve BS, Dubbudu RR, Yadav BS, Kumar LS, Gupta A, et al. Successful Use of Cefepime-Zidebactam (WCK 5222) as a Salvage Therapy for the Treatment of Disseminated Extensively Drug-Resistant New Delhi Metallo-β-Lactamase-Producing Pseudomonas aeruginosa Infection in an Adult Patient with Acute T-Cell Leukemia. Antimicrob Agents Chemother. 2023 Aug 17;67(8):e00500-23. [CrossRef]
  26. Dubey D, Roy M, Shah TH, Bano N, Kulshrestha V, Mitra S, et al. Compassionate use of a novel β-lactam enhancer-based investigational antibiotic cefepime/zidebactam (WCK 5222) for the treatment of extensively-drug-resistant NDM-expressing Pseudomonas aeruginosa infection in an intra-abdominal infection-induced sepsis patient: a case report. Ann Clin Microbiol Antimicrob. 2023 Jul 5;22(1):55. [CrossRef]
  27. Rodvold KA, Gotfried MH, Chugh R, Gupta M, Patel A, Chavan R, et al. Plasma and Intrapulmonary Concentrations of Cefepime and Zidebactam following Intravenous Administration of WCK 5222 to Healthy Adult Subjects. Antimicrob Agents Chemother. 2018 Aug;62(8):e00682-18. [CrossRef]
  28. Yahav D, Giske CG, Grāmatniece A, Abodakpi H, Tam VH, Leibovici L. New β-Lactam–β-Lactamase Inhibitor Combinations. Clin Microbiol Rev. 2020 Dec 16;34(1):e00115-20.
  29. Zhanel GG, Mansour C, Mikolayanko S, Lawrence CK, Zelenitsky S, Ramirez D, et al. Cefepime–Taniborbactam: A Novel Cephalosporin/β-Lactamase Inhibitor Combination. Drugs [Internet]. 2024 Aug 30 [cited 2024 Oct 16]; Available from: https://link.springer.com/10.1007/s40265-024-02082-9. [CrossRef]
  30. Karlowsky JA, Hackel MA, Wise MG, Six DA, Uehara T, Daigle DM, 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 Jan 24;67(1):e01281-22. [CrossRef]
  31. Lasko MJ, Nicolau DP, Asempa TE. Clinical exposure–response relationship of cefepime/taniborbactam against Gram-negative organisms in the murine complicated urinary tract infection model. Journal of Antimicrobial Chemotherapy. 2022 Feb 2;77(2):443–7. [CrossRef]
  32. Abdelraouf K, Nicolau DP. In vivo pharmacokinetic/pharmacodynamic evaluation of cefepime/taniborbactam combination against cefepime-non-susceptible Enterobacterales and Pseudomonas aeruginosa in a murine pneumonia model. Journal of Antimicrobial Chemotherapy. 2023 Mar 2;78(3):692–702.
  33. Chris Dall. FDA rejects new drug application for cefepime-taniborbactam [Internet]. 2024. Available from: https://www.cidrap.umn.edu/antimicrobial-stewardship/fda-rejects-new-drug-application-cefepime-taniborbactam.
  34. Wagenlehner FM, Gasink LB, McGovern PC, Moeck G, McLeroth P, Dorr M, et al. Cefepime–Taniborbactam in Complicated Urinary Tract Infection. N Engl J Med. 2024 Feb 15;390(7):611–22. [CrossRef]
  35. Zhang S, Liao X, Ding T, Ahn J. Role of β-Lactamase Inhibitors as Potentiators in Antimicrobial Chemotherapy Targeting Gram-Negative Bacteria. Antibiotics. 2024 Mar 15;13(3):260. [CrossRef]
  36. Li Y, Yan M, Xue F, Zhong W, Liu X, Chen X, et al. In vitro and in vivo activities of a novel β-lactamase inhibitor combination imipenem/XNW4107 against recent clinical Gram-negative bacilli from China. Journal of Global Antimicrobial Resistance. 2022 Dec;31:1–9. [CrossRef]
  37. Evaluation of the Efficacy and Safety of Intravenous Imipenem/​Cilastatin/​XNW4107 in Comparison With Meropenem in Hospitalized Adults With cUTI Including AP (EudraCT no. 2022-000061-40) [Internet]. ClinicalTrials.gov; Available from: https://clinicaltrials.gov/study/NCT05204368.
  38. Imipenem/​Cilastatin-XNW4107 Versus Imipenem/​Cilastatin/​Relebactam for Treatment of Participants With Bacterial Pneumonia (XNW4107-302, REITAB-2) (REITAB-2) [Internet]. ClinicalTrials.gov; Available from: https://clinicaltrials.gov/study/NCT05204563.
  39. Barnes MD, Taracila MA, Good CE, Bajaksouzian S, Rojas LJ, Van Duin D, et al. Nacubactam Enhances Meropenem Activity against Carbapenem-Resistant Klebsiella pneumoniae Producing KPC. Antimicrob Agents Chemother. 2019 Aug;63(8):e00432-19. [CrossRef]
  40. Mushtaq S, Vickers A, Woodford N, Haldimann A, Livermore DM. Activity of nacubactam (RG6080/OP0595) combinations against MBL-producing Enterobacteriaceae. Journal of Antimicrobial Chemotherapy. 2019 Apr 1;74(4):953–60.
  41. Asempa TE, Motos A, Abdelraouf K, Bissantz C, Zampaloni C, Nicolau DP. Efficacy of Human-Simulated Epithelial Lining Fluid Exposure of Meropenem-Nacubactam Combination against Class A Serine β-Lactamase-Producing Enterobacteriaceae in the Neutropenic Murine Lung Infection Model. Antimicrob Agents Chemother. 2019 Apr;63(4):e02382-18. [CrossRef]
  42. Monogue ML, Giovagnoli S, Bissantz C, Zampaloni C, Nicolau DP. In Vivo Efficacy of Meropenem with a Novel Non-β-Lactam–β-Lactamase Inhibitor, Nacubactam, against Gram-Negative Organisms Exhibiting Various Resistance Mechanisms in a Murine Complicated Urinary Tract Infection Model. Antimicrob Agents Chemother. 2018 Sep;62(9):e02596-17. [CrossRef]
  43. Clinicaltrials.gov. A Study to Investigate the Pharmacokinetics of RO7079901 and Meropenem in Participants With a Complicated Urinary Tract Infection [Internet]. 2018. Available from: https://clinicaltrials.gov/study/NCT03174795#study-overview.
  44. Mallalieu NL, Winter E, Fettner S, Patel K, Zwanziger E, Attley G, 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. 2020 Apr 21;64(5):e02229-19.
  45. Lomovskaya O, Castanheira M, Lindley J, Rubio-Aparicio D, Nelson K, Tsivkovski R, et al. In vitro potency of xeruborbactam in combination with multiple β-lactam antibiotics in comparison with other β-lactam/β-lactamase inhibitor (BLI) combinations against carbapenem-resistant and extended-spectrum β-lactamase-producing Enterobacterales. Poirel L, editor. Antimicrob Agents Chemother. 2023 Nov 15;67(11):e00440-23. [CrossRef]
  46. Lomovskaya O, Tsivkovski R, Totrov M, Dressel D, Castanheira M, Dudley M. New boronate drugs and evolving NDM-mediated beta-lactam resistance. Poirel L, editor. Antimicrob Agents Chemother. 2023 Sep 19;67(9):e00579-23. [CrossRef]
  47. Griffith D, Roberts J, Wallis S, Hernandez-Mitre MP, Morgan E, Gehrke S, et al. 216. A Phase 1 Study of the Safety, Tolerability, and Pharmacokinetics of Multiple Doses of the Beta-lactamase inhibitor Xeruborbactam Alone and in Combination Meropenem in Healthy Adult Subjects. Open Forum Infectious Diseases. 2022 Dec 15;9(Supplement_2):ofac492.294. [CrossRef]
  48. A DDI Study to Investigate PK and Safety of Cefiderocol in Combination With Xeruborbactam in Healthy Adult Participants [Internet]. ClinicalTrials.gov; Available from: https://clinicaltrials.gov/study/NCT06547554?intr=xeruborbactam&rank=1#study-plan.
  49. P1, DDI & MAD PK and Safety Study of Xeruborbactam Oral Prodrug in Combo With Ceftibuten in Healthy Participants [Internet]. ClinicalTrials.gov; Available from: https://clinicaltrials.gov/study/NCT06079775?intr=xeruborbactam&rank=3.
  50. PK & Safety Study of Xeruborbactam Oral Prodrug Combined With Ceftibuten in Participants With Renal Impairment [Internet]. ClinicalTrials.gov; Available from: https://clinicaltrials.gov/study/NCT06157242?intr=xeruborbactam&rank=2.
  51. Huang Z, Yang X, Jin Y, Yu J, Cao G, Wang J, et al. First-in-human study to evaluate the safety, tolerability, and population pharmacokinetic/pharmacodynamic target attainment analysis of FL058 alone and in combination with meropenem in healthy subjects. Leggett JE, editor. Antimicrob Agents Chemother. 2024 Jan 10;68(1):e01330-23. [CrossRef]
  52. Tamma PD, Heil EL, Justo JA, Mathers AJ, Satlin MJ, Bonomo RA. Infectious Diseases Society of America 2024 Guidance on the Treatment of Antimicrobial-Resistant Gram-Negative Infections. Clinical Infectious Diseases. 2024 Aug 7;ciae403. [CrossRef]
  53. EMA. New antibiotic to fight infections caused by multidrug-resistant bacteria [Internet]. 2024. Available from: https://www.ema.europa.eu/en/news/new-antibiotic-fight-infections-caused-multidrug-resistant-bacteria.
  54. Carmeli Y, Cisneros JM, Paul M, Daikos GL, Wang M, Torre-Cisneros J, et al. Aztreonam–avibactam versus meropenem for the treatment of serious infections caused by Gram-negative bacteria (REVISIT): a descriptive, multinational, open-label, phase 3, randomised trial. The Lancet Infectious Diseases. 2024 Oct;S1473309924004997.
  55. Carcione D, Siracusa C, Sulejmani A, Leoni V, Intra J. Old and New Beta-Lactamase Inhibitors: Molecular Structure, Mechanism of Action, and Clinical Use. Antibiotics. 2021 Aug 17;10(8):995. [CrossRef]
  56. Boyd SE, Livermore DM, Hooper DC, Hope WW. Metallo-β-Lactamases: Structure, Function, Epidemiology, Treatment Options, and the Development Pipeline. Antimicrob Agents Chemother. 2020 Sep 21;64(10):e00397-20.
  57. Mojica MF, Rossi MA, Vila AJ, Bonomo RA. The urgent need for metallo-β-lactamase inhibitors: an unattended global threat. The Lancet Infectious Diseases. 2022 Jan;22(1):e28–34. [CrossRef]
  58. King AM, Reid-Yu SA, Wang W, King DT, De Pascale G, Strynadka NC, et al. Aspergillomarasmine A overcomes metallo-β-lactamase antibiotic resistance. Nature. 2014 Jun;510(7506):503–6. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated