Next Article in Journal
Compassion Fatigue in a Cohort of South Italian Nurses and Hospital-Based Clinical Social Workers Following COVID-19: A Cross-Sectional Survey
Previous Article in Journal
Cutting Periprosthetic Infection Rate: Staphylococcus aureus Decolonization as a Mandatory Procedure in Preoperative Knee and Hip Replacement Care—Insights from a Systematic Review and Meta-Analysis of More Than 50,000 Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Delhi Metallo-Beta-Lactamase Inhibitors: A Systematic Scoping Review

1
Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
2
Department of Infectious Diseases, Okayama University Hospital, Okayama 700-8558, Japan
3
Department of Bacteriology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(14), 4199; https://doi.org/10.3390/jcm13144199
Submission received: 5 June 2024 / Revised: 14 July 2024 / Accepted: 16 July 2024 / Published: 18 July 2024
(This article belongs to the Section Infectious Diseases)

Abstract

:
Background/Objectives: Among various carbapenemases, New Delhi metallo-beta-lactamases (NDMs) are recognized as the most powerful type capable of hydrolyzing all beta-lactam antibiotics, often conferring multi-drug resistance to the microorganism. The objective of this review is to synthesize current scientific data on NDM inhibitors to facilitate the development of future therapeutics for challenging-to-treat pathogens. Methods: Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) Extension for Scoping Reviews, we conducted a MEDLINE search for articles with relevant keywords from the beginning of 2009 to December 2022. We employed various generic terms to encompass all the literature ever published on potential NDM inhibitors. Results: Out of the 1760 articles identified through the database search, 91 met the eligibility criteria and were included in our analysis. The fractional inhibitory concentration index was assessed using the checkerboard assay for 47 compounds in 37 articles, which included 8 compounds already approved by the Food and Drug Administration (FDA) of the United States. Time-killing curve assays (14 studies, 25%), kinetic assays (15 studies, 40.5%), molecular investigations (25 studies, 67.6%), in vivo studies (14 studies, 37.8%), and toxicity assays (13 studies, 35.1%) were also conducted to strengthen the laboratory-level evidence of the potential inhibitors. None of them appeared to have been applied to human infections. Conclusions: Ongoing research efforts have identified several potential NDM inhibitors; however, there are currently no clinically applicable drugs. To address this, we must foster interdisciplinary and multifaceted collaborations by broadening our own horizons.

1. Introduction

Antimicrobial resistance (AMR) is a pressing global issue that requires collaborative efforts from nations and foundations worldwide [1]. Clinical and public health challenges posed by emerging AMR pathogens are particularly pronounced in low-resource settings, where enhanced laboratory capabilities and robust data collection systems are needed to fully address this health threat. Until recently, carbapenems served as last-resort treatments for Gram-negative bacterial infections [2]. However, the global emergence and rapid spread of carbapenem-resistant organisms present a significant risk of high mortality across diverse populations due to limited treatment options [3,4]. Carbapenem resistance can develop through various mechanisms, including (i) structural modifications of penicillin-binding proteins, (ii) reductions in outer-membrane porins, (iii) activation of efflux pumps, and (iv) production of β-lactamases (carbapenemases) that degrade or hydrolyze carbapenems [5]. Among these, the producibility of carbapenemases is particularly noteworthy in terms of its impact on infection prevention and treatment.
A wide range of carbapenemases are classified into Ambler Classes based on their hydrolytic profiles and catalytic substrates [6]. Class B enzymes, also known as metallo-β-lactamases (MBLs), employ zinc as a cofactor at the active site of the β-lactam ring. This class mainly includes New Delhi metallo-beta-lactamase (NDM), Verona Integron-encoded metallo-beta-lactamase (VIM), and imipenemase (IMP). Among these, NDM is the most prominent genotype capable of catalyzing a range of β-lactam antibiotics, including carbapenems, and is resistant to various β-lactamase inhibitors [7]. Since the first detection of the NDM-1 gene in Enterobacterales isolated from a patient traveling from India to Sweden in 2008 [8], a total of 41 NDM variants have been identified in clinically significant pathogens such as Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii complex, and Pseudomonas aeruginosa, of which 40 variants have been deposited in the GenBank database [9,10,11]. Due to its high-level and multi-drug resistance nature, only a limited number of treatment options are available for NDM-producing bacterial infections. The endemic regions of these NDM producers have rapidly expanded worldwide, affecting communities, animals, agricultural products, and the environment [12,13], exposing an increasing number of people to untreatable infections. In the era of international travel and medical tourism, this unfavorable situation is accelerating globally [14,15].
In light of these challenges, there is significant value in promoting the development of therapeutic agents against NDM-producing bacteria. However, due to the limited research efforts in this field, progress has been modest. Nonetheless, novel antibiotics with activity against NDM producers, such as ceftazidime/avibactam plus aztreonam, aztreonam/avibactam, cefiderocol, plazomicin, and eravacycline, have recently received approval in American and European countries [16]. However, these new drugs are not yet available globally due to issues related to drug availability and cost. Combination therapy with currently available antibiotics is one approach to combat severe NDM-producing infections [16], though these strategies have not fully addressed the menace. Many studies have focused on combinatory tactics to enhance antibiotic efficacy, utilizing various compounds such as β-lactamase inhibitors, outer-membrane permeabilizers, and efflux pump inhibitors [17]. Among these, experimental and clinical investigations of combination therapy with β-lactam and β-lactamase inhibitors have been particularly explored. As a result, avibactam, relebactam, and vaborbactam have been developed and introduced to the market as serin-β-lactamase inhibitors [18,19]. However, no specific NDM inhibitors have been discovered. A recent literature review on progress in the development of MBL inhibitors summarized the molecular profiles and inhibitory mechanisms of MBLs [20]. Gu et al. have concentrated on NDM-1 inhibitors and reviewed relevant articles published after 2018, indicating chemical complexity and inconsistency [21].
Given this context, a more comprehensive evaluation of published data and a deeper discussion from a clinical applicability perspective, especially focusing on NDM inhibitors, are necessary to prepare for future crises. Therefore, our aim is to conduct a comprehensive research review of existing data on NDM inhibitors to identify promising candidates for further development.

2. Materials and Methods

2.1. Study Design and Strategy

We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) Extension for Scoping Reviews [22,23]. After a pilot search, we conducted a systematic scoping review with the following search phrases to overview MEDLINE for all peer-reviewed publications published between 1 January 2009 and 31 December 2022: “NDM inhibitor” [All Fields] OR “NDM-1 inhibitor” [All Fields] OR “NDM-1 producing bacteria” [All Fields] OR “NDM-1-producing Escherichia coli” [All Fields] OR “beta-lactamase NDM-1” [All Fields] OR “New Delhi Metallo-β-lactamase-producing Enterobacteriaceae” [All Fields] OR “New Delhi Metallo-β-lactamase-1” [All Fields] OR “New Delhi Metallo-β-lactamases” [All Fields] OR “MBL inhibitors” [All Fields] OR “Meropenem resistance” [All Fields] OR “In vitro Meropenem” [All Fields] OR “Overcome antibiotic resistance” [All Fields] OR “Synergistic antibacterial effects” [All Fields]. There were no language or research design filters used.

2.2. Eligibility Criteria

The inclusion criteria were as follows:
Peer-reviewed articles reporting results of in vitro combination tests for potential NDM inhibitors, such as checkerboard (CB) assays, time-killing assays, kinetic assays (enzyme inhibition assays using kinetic parameters such as Ki, Km, Kcat, and Kcat/Km values), molecular studies, in vivo animal studies, and toxicity assays.
The exclusion criteria were as follows:
(1)
Articles published in languages other than English.
(2)
Conference or meeting abstracts, unrelated topics, review articles, guidelines, and commentaries.

2.3. Study Selection, Data Extraction, and Definition

LN and MA collected, analyzed, and assessed the selected full-text articles. Articles that met the criteria for inclusion in this study underwent a comprehensive review. We extracted information regarding the inhibiting compounds, as well as the in vitro and in vivo methods employed to confirm the combination effects and safety data from each study.
In this study, we focused on the results of the fractional inhibitory concentration (FIC) index based on the checkerboard (CB) assay to quantitatively measure the synergistic effects of the inhibitors. Generally, the FIC of an agent is calculated by dividing the minimum inhibitory concentration (MIC) of the agent when used in combination by the MIC of the agent when used alone. The FIC index is the sum of the FICs of the combined drugs. Interactions between the combined drugs were quantified using the FIC index as follows: an FIC index of ≤0.5 was defined as synergistic, and an FIC index of ≥0.5 to ≤4.0 was considered indifferent [24].
The time-killing curve assay is also a fundamental approach to confirm the synergistic efficacy of two or more agents. In this study, we defined a bactericidal effect as a bacterial volume reduction of 3 log10 CFU/mL or more at any time during incubation when the drugs were combined. Conversely, bacteriostatic activity was characterized by a reduction of less than 3 log10 CFU/mL compared to the initial inoculum.

2.4. Data Synthesis and Statistical Analysis

Data processing and aggregation were performed using Microsoft Excel® software version 2021 (Microsoft Corporation, Redmond, WA, USA). We did not perform any statistical analysis since this is a descriptive study.

3. Results

3.1. Search Results and Study Selection

The flowchart depicting the stages of article collection is presented in Figure 1, illustrating the process of identifying relevant reports, screening records, evaluating eligibility, and making final determinations for inclusion or exclusion in accordance with the PRISMA flow diagram. The initial search of MEDLINE databases yielded 1760 articles, which underwent further eligibility screening, resulting in the exclusion of 1628 articles. Subsequently, 132 full-text articles were assessed, and 39 articles lacking experimental data and 2 articles related to triplet agent therapy were excluded. Ultimately, 91 articles (comprising 89 original articles and 2 letter-type articles) were selected for the review.

3.2. Description of the Review Results

The number of articles has significantly increased, especially in the last five years: 12 in 2018, 11 in 2019, 18 in 2020, 15 in 2021, and 22 in 2022 (Figure 2). A summary of 91 articles reporting 154 potential NDM inhibitors is provided in Table 1 [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115]. All 91 studies were found to have conducted CB assays. Time-killing curve assays, kinetic assays, molecular investigations, in vivo (animal- or cell-based) combination studies, and toxicity assays were carried out in 26 (28.6%), 41 (45.1%), 66 (72.5%), 30 (33.0%), and 44 (48.4%) of the studies, respectively. Various strains of NDM-producing bacteria were used in both in vitro and in vivo studies (Supplementary Table S1). The two most common isolates employed were Escherichia coli and Klebsiella pneumoniae, followed by other Enterobacterales species, Pseudomonas aeruginosa, and Acinetobacter baumanii. Clinical, recombinant, standard, reference, and wild strains were used in 57 (62.6%), 27 (29.7%), 25 (27.5%), 3 (3.3%), and 2 (2.2%) of the studies, respectively, including some duplications.
Out of the 154 NDM inhibitors extracted from 91 eligible articles, we specifically identified 47 potential inhibitors in 37 articles, where the FIC index was determined based on the CB assay (Table 2). Among these, eight compounds had already received approval from the United States FDA. Almost all of these compounds exhibited synergistic effects with an FIC index of less than 0.5. However, some cases of indifferent results were identified when various bacterial strains were tested. Out of these, 14 (37.8%) studies included data on the time-killing curve assay. Bacteriostatic effects were reported in 4 studies, while 10 studies (involving 11 inhibitors) demonstrated bactericidal effects.
Additionally, 12 studies (32.4%) conducted kinetic assays, in which kinetic parameters were calculated. Molecular investigations were conducted in 23 (62.2%) studies, with molecular docking and molecular dynamic simulations being commonly employed (15 out of 25 studies, 60%). To validate the efficacy of combination therapy, 10 studies (27%) presented in vivo animal data, all of which used mouse models. To assess the safety of the inhibitory drugs used, 13 (35.1%) studies reported results of toxicity assays using in vivo models. Notably, none of the compounds exhibited apparent toxic effects.

4. Discussion

In this scoping review, we have compiled the presently available data on NDM inhibitors published in MEDLINE. Among the various experimental methods used to evaluate the efficacy of drug combinations, we specifically focused on the FIC index calculated through the CB assay, which serves as a fundamental approach to determine the synergistic effects of two distinct drugs. Since 2014, a total of 47 compounds have been investigated as potential NDM inhibitors, with 8 of them having received approval from the United States FDA. These FDA-approved drugs include various substances such as methimazole, withaferin A, fisetin, isoliquiritin, cefmetazole, pterostilbene, cefoxitin, and tetracycline [34,37,38,74,78,81,83]. In addition to the CB assay, bactericidal effects were observed in 10 compounds through time-killing curve assays, of which 4 substances (methimazole, fisetin, isoliquiritin, and cefmetazole) had already received FDA endorsement [34,38,74,78]. No further investigations had been conducted for methimazole and cefmetazole [34,78], whereas the effectiveness and safety of combining fisetin or isoliquiritin were additionally confirmed through other approaches [38,74]. Regrettably, there were no inhibiting agents that seemed readily available for clinical use, and none of these are within the reach of clinicians.
Kinetic assays and molecular investigations represent more advanced methods for ascertaining combination efficacy. Comparing molecular affinities among compounds of interest using kinetic parameters such as Ki, Km, Kcat, and Kcat/Km can provide insights into inhibitory activity from an enzymatic perspective. Molecular docking simulations of potential inhibitors are well-established computational methods for analyzing molecular binding modes. Among these two elaborated approaches, molecular docking and molecular dynamic simulations were more frequently performed (62.2% vs. 32.4%). Eleven studies did not conduct either of these methods [25,33,34,55,69,74,76,96,97,106,108], while nine studies evaluated both [38,46,48,49,75,80,88,95,113]. Additionally, in vivo animal studies were performed in 10 studies [38,46,48,75,80,81,82,95,96,113], suggesting that the tested compounds, including H2dpa derivatives, sulfamoylfuran-3-carboxylic acid derivatives, peptidomimetic 4, pterostilbene, and aspergillomarasmine A, may hold promise as inhibitors.
For unapproved compounds, ensuring their safety is essential for potential future clinical use. In this sense, toxicity assays provide particularly important data. In our review, 13 out of 37 studies (35.1%) conducted these assays, primarily using a mouse model. Notably, no inhibitors with apparent toxicity were reported. However, it is essential to mention that zinc-chelating agents may not be suitable for therapeutic use due to their well-documented toxicity to human cells [25,44,49,56,82,88].
Our study has a few limitations that should be acknowledged. First, we conducted our search exclusively on MEDLINE due to the unavailability of access to other databases. This could potentially lead to an underestimation of relevant articles. In fact, our search approach failed to include boron-based inhibitors, such as taniborbactam, xeruborbactam, and zidebactam, which have the potential to be available in clinical settings. Possibilities of reporting bias should also be considered. Second, we only included articles in the English language, which may restrict comprehensiveness and affect generalizability. Third, our search period was up to the end of December 2022, which should have been extended to the time of drafting, because an increasing number of relevant articles have been reported in the literature. Due to time constraints, we could not afford to do so. Fourth, the presence of publication bias should be taken into consideration. Data that could be unfavorable for the inhibitors might not have been included in the articles. Fifth, clinical strains may possess various antimicrobial resistance mechanisms, and, therefore, the combination of NDM inhibitors may not necessarily exhibit synergistic effects in clinical settings. Finally, the assessment of the quality of the included studies was not fully performed, although it is a crucial aspect of the review study to ensure the validity and reliability of the conclusion.

5. Conclusions

In summary, there are currently no NDM inhibitors available for therapeutic use. While previous efforts have borne fruit in identifying some potential compounds, there is still a long road ahead to discover clinically applicable and outstanding NDM inhibitors. Just as the development of serine-β-lactamase inhibitors has set an example, it is time for NDM inhibitor research to follow suit. For this purpose, the establishment of a laboratory and clinical research platform under interdisciplinary collaborations is necessary. We believe that our review work will contribute to advancing this challenging journey.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/jcm13144199/s1, Table S1: Species and types of NDM-producing bacteria used in each study [116].

Author Contributions

Study concept: H.H.; data collection and reviewing: L.N. and M.A.; drafting: L.N. and H.H.; revising: K.G. and F.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Available with a valid reason from a corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Murray, C.J.L.; Ikuta, K.S.; Sharara, F.; Swetschinski, L.; Aguilar, G.R.; Gray, A.; Han, C.; Bisignano, C.; Rao, P.; Wool, E.; et al. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  2. Tamma, P.D.; Aitken, S.L.; Bonomo, R.A.; Mathers, A.J.; van Duin, D.; Clancy, C.J. Infectious Diseases Society of America 2023 Guidance on the Treatment of Antimicrobial Resistant Gram-Negative Infections. Clin. Infect. Dis. 2023, ciad428. [Google Scholar] [CrossRef]
  3. Centers for Disease Control and Prevention. CDC’s Antibiotic Resistance Threats in the United States; Centers for Disease Control and Prevention: Atlanta, GA, USA, 2019. Available online: https://www.cdc.gov/antimicrobial-resistance/media/pdfs/2019-ar-threats-report-508.pdf?CDC_AAref_Val (accessed on 6 September 2022).
  4. Davies, O.L.; Bennett, S. WHO Publishes List of Bacteria for Which New Antibiotics Are Urgently Needed. WHO Newsletters. 2017. Available online: https://www.who.int/news/item/27-02-2017-who-publishes-list-of-bacteria-for-which-new-antibiotics-are-urgently-needed (accessed on 6 September 2022).
  5. Papp-Wallace, K.M.; Endimiani, A.; Taracila, M.A.; Bonomo, R.A. Carbapenems: Past, present, and future. Antimicrob. Agents Chemother. 2011, 55, 4943–4960. [Google Scholar] [CrossRef]
  6. Codjoe, F.S.; Donkor, E.S. Carbapenem Resistance: A Review. Med. Sci. 2017, 6, 1. [Google Scholar] [CrossRef] [PubMed]
  7. Li, T.; Wang, Q.; Chen, F.; Li, X.; Luo, S.; Fang, H.; Wang, D.; Li, Z.; Hou, X.; Wang, H. Biochemical Characteristics of New Delhi Metallo-β-Lactamase-1 Show Unexpected Difference to Other MBLs. PLoS ONE 2013, 8, e61914. [Google Scholar] [CrossRef]
  8. Kumarasamy, K.K.; Toleman, M.A.; Walsh, T.R.; Bagaria, J.; Butt, F.; Balakrishnan, R.; Chaudhary, U.; Doumith, M.; Giske, C.G.; Irfan, S.; et al. Emergence of a new antibiotic resistance mechanism in India, Pakistan, and the UK: A molecular, biological, and epidemiological study. Lancet Infect. Dis. 2010, 10, 597–602. [Google Scholar] [CrossRef] [PubMed]
  9. Ma, W.; Zhu, B.; Wang, W.; Wang, Q.; Cui, X.; Wang, Y.; Dong, X.; Li, X.; Ma, J.; Cheng, F.; et al. Genetic and enzymatic characterization of two novel blaNDM-36, -37 variants in Escherichia coli strains. Eur. J. Clin. Microbiol. Infect. Dis. 2023, 42, 471–480. [Google Scholar] [CrossRef]
  10. Boyd, S.E.; Livermore, D.M.; Hooper, D.C.; Hope, W.W. Metallo-β-lactamases: Structure, function, epidemiology, treatment options, and the development pipeline. Antimicrob. Agents Chemother. 2020, 64, e00397-20. [Google Scholar] [CrossRef] [PubMed]
  11. Farooq, S.; Khan, A.U. Current Update on New Delhi Metallo-β-lactamase (NDM) Variants: New Challenges in the Journey of Evolution. Curr. Protein Pept. Sci. 2023, 24, 655–665. [Google Scholar] [CrossRef]
  12. Mills, M.C.; Lee, J. The threat of carbapenem-resistant bacteria in the environment: Evidence of widespread contamination of reservoirs at a global scale. Environ. Pollut. 2019, 255, 113143. [Google Scholar] [CrossRef]
  13. Taggar, G.; Attiq Rheman, M.; Boerlin, P.; Diarra, M.S. Molecular epidemiology of carbapenemases in enterobacteriales from humans, animals, food and the environment. Antibiotics 2020, 9, 693. [Google Scholar] [CrossRef]
  14. Mellon, G.; Turbett, S.E.; Worby, C.; Oliver, E.; Walker, A.T.; Walters, M.; Kelly, P.; Leung, D.T.; Knouse, M.; Hagmann, S.; et al. Acquisition of antibiotic-resistant bacteria by US international travelers. N. Engl. J. Med. 2020, 382, 1372–1374. [Google Scholar] [CrossRef]
  15. Theriault, N.; Tillotson, G.; Sandrock, C.E. Global travel and Gram-negative bacterial resistance; implications on clinical management. Expert Rev. Anti-Infect. Ther. 2020, 19, 181–196. [Google Scholar] [CrossRef] [PubMed]
  16. Corona, A.; De Santis, V.; Agarossi, A.; Prete, A.; Cattaneo, D.; Tomasini, G.; Bonetti, G.; Patroni, A.; Latronico, N. Antibiotic Therapy Strategies for Treating Gram-Negative Severe Infections in the Critically Ill: A Narrative Review. Antibiotics 2023, 12, 1262. [Google Scholar] [CrossRef] [PubMed]
  17. González-Bello, C.; Rodríguez, D.; Pernas, M.; Rodríguez, A.; Colchón, E. β-Lactamase Inhibitors to Restore the Efficacy of Antibiotics against Superbugs. J. Med. Chem. 2020, 63, 1859–1881. [Google Scholar] [CrossRef]
  18. Yahav, D.; Giske, C.G.; Grāmatniece, A.; Abodakpi, H.; Tam, V.H.; Leibovici, L. New β-Lactam–β-Lactamase Inhibitor Combinations. Clin. Microbiol. Rev. 2021, 34, 2021. [Google Scholar] [CrossRef]
  19. Olney, K.B.; Thomas, J.K.; Johnson, W.M. Review of novel β-lactams and β-lactam/β-lactamase inhibitor combinations with implications for pediatric use. Pharmacotherapy 2023, 43, 713–731. [Google Scholar] [CrossRef] [PubMed]
  20. Yang, Y.; Yan, Y.-H.; Schofield, C.J.; McNally, A.; Zong, Z.; Li, G.-B. Metallo-β-lactamase-mediated antimicrobial resistance and progress in inhibitor discovery. Trends Microbiol. 2023, 31, 735–748. [Google Scholar] [CrossRef]
  21. Gu, X.; Zheng, M.; Chen, L.; Li, H. The development of New Delhi metallo-β-lactamase-1 inhibitors since 2018. Microbiol. Res. 2022, 261, 127079. [Google Scholar] [CrossRef]
  22. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Horsley, T.; Weeks, L.; Hempel, S.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef]
  23. McGowan, J.; Straus, S.; Moher, D.; Langlois, E.V.; O’Brien, K.K.; Horsley, T.; Aldcroft, A.; Zarin, W.; Garitty, C.M.; Hempel, S.; et al. Reporting scoping reviews—PRISMA ScR extension. J. Clin. Epidemiol. 2020, 123, 177–179. [Google Scholar] [CrossRef] [PubMed]
  24. Hollander, J.G.D.; Mouton, J.W.; Verbrugh, H.A. Use of pharmacodynamic parameters to predict efficacy of combination therapy by using fractional inhibitory concentration kinetics. Antimicrob. Agents Chemother. 1998, 42, 744–748. [Google Scholar] [CrossRef] [PubMed]
  25. Rudresh, S.M.; Ravi, G.S.; Raksha, Y. In Vitro Efficacy of Biocompatible Zinc Ion Chelating Molecules as Metallo-β-Lactamase Inhibitor among NDM Producing Escherichia coli. J. Lab. Physicians 2022, 15, 62–68. [Google Scholar] [CrossRef] [PubMed]
  26. Legru, A.; Verdirosa, F.; Vo-Hoang, Y.; Tassone, G.; Vascon, F.; Thomas, C.A.; Sannio, F.; Corsica, G.; Benvenuti, M.; Feller, G.; et al. Optimization of 1,2,4-Triazole-3-thiones toward Broad-Spectrum Metallo-β-lactamase Inhibitors Showing Potent Synergistic Activity on VIM- and NDM-1-Producing Clinical Isolates. J. Med. Chem. 2022, 65, 16392–16419. [Google Scholar] [CrossRef] [PubMed]
  27. Yue, K.; Xu, C.; Wang, Z.; Liu, W.; Liu, C.; Xu, X.; Xing, Y.; Chen, S.; Li, X.; Wan, S. 1,2-Isoselenazol-3(2H)-one derivatives as NDM-1 inhibitors displaying synergistic antimicrobial effects with meropenem on NDM-1 producing clinical isolates. Bioorganic Chem. 2022, 129, 106153. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, C.; Xiang, Y.; Yang, K.-W.; Wang, D.; Dan, H.; Wang, N.-N. Discovery of environment-sensitive fluorescent probes for detecting and inhibiting metallo-β-lactamase. Bioorganic Chem. 2022, 128, 106048. [Google Scholar] [CrossRef] [PubMed]
  29. Tian, H.; Wang, Y.; Dai, Y.; Mao, A.; Zhou, W.; Cao, X.; Deng, H.; Lu, H.; Ding, L.; Wang, X.; et al. A Cephalosporin-Tripodalamine Conjugate Inhibits Metallo-β-Lactamase with High Efficacy and Low Toxicity. Antimicrob. Agents Chemother. 2022, 66, e0035222. [Google Scholar] [CrossRef]
  30. Caburet, J.; Boucherle, B.; Bourdillon, S.; Simoncelli, G.; Verdirosa, F.; Docquier, J.-D.; Moreau, Y.; Krimm, I.; Crouzy, S.; Peuchmaur, M. A fragment-based drug discovery strategy applied to the identification of NDM-1 β-lactamase inhibitors. Eur. J. Med. Chem. 2022, 240, 114599. [Google Scholar] [CrossRef] [PubMed]
  31. Fasim, A.; More, S.S. Identification of a potential inhibitor for New Delhi metallo-β-lactamase 1 (NDM-1) from FDA approved chemical library- a drug repurposing approach to combat carbapenem resistance. J. Biomol. Struct. Dyn. 2022, 41, 7700–7711. [Google Scholar] [CrossRef]
  32. Zhai, L.; Jiang, Y.; Shi, Y.; Lv, M.; Pu, Y.-L.; Cheng, H.-L.; Zhu, J.-Y.; Yang, K.-W. Aromatic Schiff bases confer inhibitory efficacy against New Delhi metallo-β-lactamase-1 (NDM-1). Bioorganic Chem. 2022, 126, 105910. [Google Scholar] [CrossRef]
  33. Scaccaglia, M.; Rega, M.; Bacci, C.; Giovanardi, D.; Pinelli, S.; Pelosi, G.; Bisceglie, F. Bismuth complex of quinoline thiosemicarbazone restores carbapenem sensitivity in NDM-1-positive Klebsiella pneumoniae. J. Inorg. Biochem. 2022, 234, 111887. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, B.; Yang, Y.; Yuan, J.; Chen, L.; Tong, H.; Huang, T.; Shi, L.; Jiang, Z. Methimazole and α-lipoic acid as metallo-β-lactamases inhibitors. J. Antibiot. 2022, 75, 282–286. [Google Scholar] [CrossRef] [PubMed]
  35. Thomas, P.W.; Cho, E.J.; Bethel, C.R.; Smisek, T.; Ahn, Y.-C.; Schroeder, J.M.; Thomas, C.A.; Dalby, K.N.; Beckham, J.T.; Crowder, M.W.; et al. Discovery of an Effective Small-Molecule Allosteric Inhibitor of New Delhi Metallo-β-lactamase (NDM). ACS Infect. Dis. 2022, 8, 811–824. [Google Scholar] [CrossRef] [PubMed]
  36. Hu, L.; Yang, H.; Yu, T.; Chen, F.; Liu, R.; Xue, S.; Zhang, S.; Mao, W.; Ji, C.; Wang, H.; et al. Stereochemically altered cephalosporins as potent inhibitors of New Delhi metallo-β-lactamases. Eur. J. Med. Chem. 2022, 232, 114174. [Google Scholar] [CrossRef] [PubMed]
  37. Vasudevan, A.; Kesavan, D.K.; Wu, L.; Su, Z.; Wang, S.; Ramasamy, M.K.; Hopper, W.; Xu, H. In Silico and In Vitro Screening of Natural Compounds as Broad-Spectrum β-Lactamase Inhibitors against Acinetobacter baumannii New Delhi Metallo-β-lactamase-1 (NDM-1). BioMed Res. Int. 2022, 2022, 4230788. [Google Scholar] [CrossRef] [PubMed]
  38. Guo, Y.; Yang, Y.; Xu, X.; Li, L.; Zhou, Y.; Jia, G.; Wei, L.; Yu, Q.; Wang, J. Metallo-β-lactamases inhibitor fisetin attenuates meropenem resistance in NDM-1-producing Escherichia coli. Eur. J. Med. Chem. 2022, 231, 114108. [Google Scholar] [CrossRef] [PubMed]
  39. Chigan, J.-Z.; Hu, Z.; Liu, L.; Xu, Y.-S.; Ding, H.-H.; Yang, K.-W. Quinolinyl sulfonamides and sulphonyl esters exhibit inhibitory efficacy against New Delhi metallo-β-lactamase-1 (NDM-1). Bioorganic Chem. 2022, 120, 105654. [Google Scholar] [CrossRef] [PubMed]
  40. He, Y.; Zhou, S.; Sun, W.; Li, Q.; Wang, J.; Zhang, J. Emerione A, a novel fungal metabolite as an inhibitor of New Delhi metallo-β-lactamase 1, restores carbapenem susceptibility in carbapenem-resistant isolates. J. Glob. Antimicrob. Resist. 2022, 28, 216–222. [Google Scholar] [CrossRef]
  41. Muteeb, G.; Alsultan, A.; Farhan, M.; Aatif, M. Risedronate and Methotrexate Are High-Affinity Inhibitors of New Delhi Metallo-β-Lactamase-1 (NDM-1): A Drug Repurposing Approach. Molecules 2022, 27, 1283. [Google Scholar] [CrossRef]
  42. Koteva, K.; Sychantha, D.; Rotondo, C.M.; Hobson, C.; Britten, J.F.; Wright, G.D. Three-Dimensional Structure and Optimization of the Metallo-β-Lactamase Inhibitor Aspergillomarasmine A. ACS Omega 2022, 7, 4170–4184. [Google Scholar] [CrossRef]
  43. Grigorenko, V.G.; Khrenova, M.G.; Andreeva, I.P.; Rubtsova, M.Y.; Lev, A.I.; Novikova, T.S.; Detusheva, E.V.; Fursova, N.K.; Dyatlov, I.A.; Egorov, A.M. Drug Repurposing of the Unithiol: Inhibition of Metallo-β-Lactamases for the Treatment of Carbapenem-Resistant Gram-Negative Bacterial Infections. Int. J. Mol. Sci. 2022, 23, 1834. [Google Scholar] [CrossRef]
  44. Proschak, A.; Martinelli, G.; Frank, D.; Rotter, M.J.; Brunst, S.; Weizel, L.; Burgers, L.D.; Fürst, R.; Proschak, E.; Sosič, I.; et al. Nitroxoline and its derivatives are potent inhibitors of metallo-β-lactamases. Eur. J. Med. Chem. 2022, 228, 113975. [Google Scholar] [CrossRef]
  45. Brem, J.; Panduwawala, T.; Hansen, J.U.; Hewitt, J.; Liepins, E.; Donets, P.; Espina, L.; Farley, A.J.M.; Shubin, K.; Campillos, G.G.; et al. Imitation of β-lactam binding enables broad-spectrum metallo-β-lactamase inhibitors. Nat. Chem. 2022, 14, 15–24. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, C.; Yang, K.-W.; Zhai, L.; Ding, H.-H.; Chigan, J.-Z. Dithiocarbamates combined with copper for revitalizing meropenem efficacy against NDM-1-producing Carbapenem-resistant Enterobacteriaceae. Bioorganic Chem. 2022, 118, 105474. [Google Scholar] [CrossRef]
  47. Krasavin, M.; Zhukovsky, D.; Solovyev, I.; Barkhatova, D.; Dar’in, D.; Frank, D.; Martinelli, G.; Weizel, L.; Proschak, A.; Proschak, E.; et al. RhII-Catalyzed De-symmetrization of Ethane-1,2-dithiol and Propane-1,3-dithiol Yields Metallo-β-lactamase Inhibitors. ChemMedChem 2021, 16, 3410–3417. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, F.; Bai, M.; Liu, W.; Kong, H.; Zhang, T.; Yao, H.; Zhang, E.; Du, J.; Qin, S. H2dpa derivatives containing pentadentate ligands: An acyclic adjuvant potentiates meropenem activity in vitro and in vivo against metallo-β-lactamase-producing Enterobacterales. Eur. J. Med. Chem. 2021, 224, 113702. [Google Scholar] [CrossRef] [PubMed]
  49. Moreira, J.S.; Galvão, D.S.; Xavier, C.F.C.; Cunha, S.; Pita, S.S.d.R.; Reis, J.N.; de Freitas, H.F. Phenotypic and in silico studies for a series of synthetic thiosemicarbazones as New Delhi metallo-beta-lactamase carbapenemase inhibitors. J. Biomol. Struct. Dyn. 2021, 40, 14223–14235. [Google Scholar] [CrossRef]
  50. Ge, Y.; Kang, P.-W.; Li, J.-Q.; Gao, H.; Zhai, L.; Sun, L.-Y.; Chen, C.; Yang, K.-W. Thiosemicarbazones exhibit inhibitory efficacy against New Delhi metallo-β-lactamase-1 (NDM-1). J. Antibiot. 2021, 74, 574–579. [Google Scholar] [CrossRef]
  51. Gao, H.; Li, J.Q.; Kang, P.W.; Chigan, J.Z.; Wang, H.; Liu, L.; Xu, Y.; Zhai, L.; Yang, K.W. N-acylhydrazones confer inhibitory efficacy against New Delhi metallo-β-lactamase-1. Bioorganic Chem. 2021, 114, 105138. [Google Scholar] [CrossRef]
  52. Romero, E.; Oueslati, S.; Benchekroun, M.; D’hollander, A.C.; Ventre, S.; Vijayakumar, K.; Minard, C.; Exilie, C.; Tlili, L.; Cariou, K.; et al. Azetidinimines as a novel series of non-covalent broad-spectrum inhibitors of β-lactamases with submicromolar activities against carbapenemases KPC-2 (class A), NDM-1 (class B) and OXA-48 (class D). Eur. J. Med. Chem. 2021, 219, 113418. [Google Scholar] [CrossRef]
  53. Farley, A.J.; Ermolovich, Y.; Calvopiña, K.; Rabe, P.; Panduwawala, T.; Brem, J.; Björkling, F.; Schofield, C.J. Structural Basis of Metallo-β-lactamase Inhibition by N-Sulfamoylpyrrole-2-carboxylates. ACS Infect. Dis. 2021, 7, 1809–1817. [Google Scholar] [CrossRef]
  54. Wade, N.; Tehrani, K.H.M.E.; Brüchle, N.C.; van Haren, M.J.; Mashayekhi, V.; Martin, N.I. Mechanistic Investigations of Metallo-β-lactamase Inhibitors: Strong Zinc Binding Is Not Required for Potent Enzyme Inhibition**. ChemMedChem 2021, 16, 1651–1659. [Google Scholar] [CrossRef]
  55. van Haren, M.J.; Tehrani, K.H.; Kotsogianni, I.; Wade, N.; Brüchle, N.C.; Mashayekhi, V.; Martin, N.I. Cephalosporin Prodrug Inhibitors Overcome Metallo-β-Lactamase Driven Antibiotic Resistance. Chem. A Eur. J. 2021, 27, 3806–3811. [Google Scholar] [CrossRef]
  56. Jackson, A.C.; Pinter, T.B.; Talley, D.C.; Baker-Agha, A.; Patel, D.; Smith, P.J.; Franz, K.J. Benzimidazole and Benzoxazole Zinc Chelators as Inhibitors of Metallo-β-Lactamase NDM-1. ChemMedChem 2021, 16, 654–661. [Google Scholar] [CrossRef]
  57. Li, J.-Q.; Sun, L.-Y.; Jiang, Z.; Chen, C.; Gao, H.; Chigan, J.-Z.; Ding, H.-H.; Yang, K.-W. Diaryl-substituted thiosemicarbazone: A potent scaffold for the development of New Delhi metallo-β-lactamase-1 inhibitors. Bioorganic Chem. 2021, 107, 104576. [Google Scholar] [CrossRef]
  58. Ooi, N.; Lee, V.E.; Chalam-Judge, N.; Newman, R.; Wilkinson, A.J.; Cooper, I.R.; Orr, D.; Lee, S.; Savage, V.J. Restoring carbapenem efficacy: A novel carbapenem companion targeting metallo-β-lactamases in carbapenem-resistant Enterobacterales. J. Antimicrob. Chemother. 2021, 76, 460–466. [Google Scholar] [CrossRef]
  59. Rossi, M.-A.; Martinez, V.; Hinchliffe, P.; Mojica, M.F.; Castillo, V.; Moreno, D.M.; Smith, R.; Spellberg, B.; Drusano, G.L.; Banchio, C.; et al. 2-Mercaptomethyl-thiazolidines use conserved aromatic–S interactions to achieve broad-range inhibition of metallo-β-lactamases. Chem. Sci. 2021, 12, 2898–2908. [Google Scholar] [CrossRef]
  60. Zhao, B.; Zhang, X.; Yu, T.; Liu, Y.; Zhang, X.; Yao, Y.; Feng, X.; Liu, H.; Yu, D.; Qin, S.; et al. Discovery of thiosemicarbazone derivatives as effective New Delhi metallo-β-lactamase-1 (NDM-1) inhibitors against NDM-1 producing clinical isolates. Acta Pharm. Sin. B 2021, 11, 203–221. [Google Scholar] [CrossRef]
  61. Ma, G.; Wang, S.; Wu, K.; Zhang, W.; Ahmad, A.; Hao, Q.; Lei, X.; Zhang, H. Structure-guided optimization of D-captopril for discovery of potent NDM-1 inhibitors. Bioorganic Med. Chem. 2021, 29, 115902. [Google Scholar] [CrossRef] [PubMed]
  62. Gavara, L.; Sevaille, L.; De Luca, F.; Mercuri, P.; Bebrone, C.; Feller, G.; Legru, A.; Cerboni, G.; Tanfoni, S.; Hernandez, J.F.; et al. 4-Amino-1,2,4-triazole-3-thione-derived Schiff bases as metallo-β-lactamase inhibitors. Eur. J. Med. Chem. 2020, 208, 112720. [Google Scholar] [CrossRef] [PubMed]
  63. Yang, Y.; Guo, Y.; Zhou, Y.; Gao, Y.; Wang, X.; Wang, J.; Niu, X. Discovery of a Novel Natural Allosteric Inhibitor That Targets NDM-1 Against Escherichia coli. Front. Pharmacol. 2020, 11, 581001. [Google Scholar] [CrossRef] [PubMed]
  64. Kazi, M.I.; Perry, B.W.; Card, D.C.; Schargel, R.D.; Ali, H.B.; Obuekwe, V.C.; Sapkota, M.; Kang, K.N.; Pellegrino, M.W.; Boll, J.M.; et al. Discovery and characterization of New Delhi metallo-b-lactamase-1 inhibitor peptides that potentiate meropenem-dependent killing of carbapenemase-producing Enterobacteriaceae. J. Antimicrob. Chemother. 2020, 75, 2843–2851. [Google Scholar] [CrossRef] [PubMed]
  65. Principe, L.; Vecchio, G.; Sheehan, G.; Kavanagh, K.; Morroni, G.; Viaggi, V.; di Masi, A.; Giacobbe, D.R.; Luzzaro, F.; Luzzati, R.; et al. Zinc Chelators as Carbapenem Adjuvants for Metallo-β-Lactamase-Producing Bacteria: In Vitro and In Vivo Evaluation. Microb. Drug Resist. 2020, 26, 1133–1143. [Google Scholar] [CrossRef] [PubMed]
  66. Davies, D.T.; Leiris, S.; Sprynski, N.; Castandet, J.; Lozano, C.; Bousquet, J.; Zalacain, M.; Vasa, S.; Dasari, P.K.; Pattipati, R.; et al. ANT2681: SAR Studies Leading to the Identification of a Metallo-β-lactamase Inhibitor with Potential for Clinical Use in Combination with Meropenem for the Treatment of Infections Caused by NDM-Producing Enterobacteriaceae. ACS Infect. Dis. 2020, 6, 2419–2430. [Google Scholar] [CrossRef] [PubMed]
  67. Cui, D.-Y.; Yang, Y.; Bai, M.-M.; Han, J.-X.; Wang, C.-C.; Kong, H.-T.; Shen, B.-Y.; Yan, D.-C.; Xiao, C.-L.; Liu, Y.-S.; et al. Systematic research of H2dedpa derivatives as potent inhibitors of New Delhi Metallo-β-lactamase-1. Bioorganic Chem. 2020, 101, 103965. [Google Scholar] [CrossRef]
  68. Jin, W.B.; Xu, C.; Cheung, Q.; Gao, W.; Zeng, P.; Liu, J.; Chan, E.; Leung, Y.; Chan, T.H.; Chan, K.F.; et al. Bioisosteric investigation of ebselen: Synthesis and in vitro characterization of 1,2-benzisothiazol-3(2H)-one derivatives as potent New Delhi metallo-β-lactamase inhibitors. Bioorganic Chem. 2020, 100, 103873. [Google Scholar] [CrossRef]
  69. Tehrani, K.H.M.E.; Brüchle, N.C.; Wade, N.; Mashayekhi, V.; Pesce, D.; van Haren, M.J.; Martin, N.I. Small Molecule Carboxylates Inhibit Metallo-β-lactamases and Resensitize Carbapenem-Resistant Bacteria to Meropenem. ACS Infect. Dis. 2020, 6, 1366–1371. [Google Scholar] [CrossRef]
  70. Thomas, C.S.; Braun, D.R.; Olmos, J.L., Jr.; Rajski, S.R.; Phillips, G.N., Jr.; Andes, D.; Bugni, T.S. Pyridine-2,6-Dithiocarboxylic Acid and Its Metal Complexes: New Inhibitors of New Delhi Metallo β-Lactamase-1. Mar. Drugs 2020, 18, 295. [Google Scholar] [CrossRef]
  71. Wang, X.; Yang, Y.; Gao, Y.; Niu, X. Discovery of the novel inhibitor against New Delhi metallo-β-lactamase based on virtual screening and molecular modelling. Int. J. Mol. Sci. 2020, 21, 3567. [Google Scholar] [CrossRef]
  72. Jackson, A.C.; Zaengle-Barone, J.M.; Puccio, E.A.; Franz, K.J. A Cephalosporin Prochelator Inhibits New Delhi Metallo-β-lactamase 1 without Removing Zinc. ACS Infect. Dis. 2020, 6, 1264–1272. [Google Scholar] [CrossRef]
  73. Guo, H.; Cheng, K.; Gao, Y.; Bai, W.; Wu, C.; He, W.; Li, C.; Li, Z. A novel potent metal-binding NDM-1 inhibitor was identified by fragment virtual, SPR and NMR screening. Bioorganic Med. Chem. 2020, 28, 115437. [Google Scholar] [CrossRef]
  74. Wang, Y.; Sun, X.; Kong, F.; Xia, L.; Deng, X.; Wang, D.; Wang, J. Specific NDM-1 inhibitor of isoliquiritin enhances the activity of meropenem against NDM-1-positive Enterobacteriaceae in vitro. Int. J. Environ. Res. Public Health 2020, 17, 2162. [Google Scholar] [CrossRef]
  75. Wachino, J.-I.; Jin, W.; Kimura, K.; Kurosaki, H.; Sato, A.; Arakawa, Y. Sulfamoyl Heteroarylcarboxylic Acids as Promising Metallo-β-Lactamase Inhibitors for Controlling Bacterial Carbapenem Resistance. mBio 2020, 11, e03144-19. [Google Scholar] [CrossRef]
  76. Tehrani, K.H.; Fu, H.; Brüchle, N.C.; Mashayekhi, V.; Luján, A.P.; van Haren, M.J.; Poelarends, G.J.; Martin, N.I. Aminocarboxylic acids related to aspergillomarasmine A (AMA) and ethylenediamine-N,N′-disuccinic acid (EDDS) are strong zinc-binders and inhibitors of the metallo-beta-lactamase NDM-1. Chem. Commun. 2020, 56, 3047–3049. [Google Scholar] [CrossRef]
  77. Chen, C.; Yang, K.-W.; Wu, L.-Y.; Li, J.-Q.; Sun, L.-Y. Disulfiram as a potent metallo-β-lactamase inhibitor with dual functional mechanisms. Chem. Commun. 2020, 56, 2755–2758. [Google Scholar] [CrossRef]
  78. Hagiya, H.; Sugawara, Y.; Tsutsumi, Y.; Akeda, Y.; Yamamoto, N.; Sakamoto, N.; Shanmugakani, R.K.; Abe, R.; Takeuchi, D.; Nishi, I.; et al. In Vitro Efficacy of Meropenem-Cefmetazole Combination Therapy against New Delhi Metallo-β-lactamase-producing Enterobacteriaceae. Int. J. Antimicrob. Agents 2020, 55, 105905. [Google Scholar] [CrossRef]
  79. Kang, P.-W.; Su, J.-P.; Sun, L.-Y.; Gao, H.; Yang, K.-W. 3-Bromopyruvate as a potent covalently reversible inhibitor of New Delhi metallo-β-lactamase-1 (NDM-1). Eur. J. Pharm. Sci. 2020, 142, 105161. [Google Scholar] [CrossRef]
  80. Liu, Y.; Yang, K.; Jia, Y.; Wang, Z. Repurposing Peptidomimetic as Potential Inhibitor of New Delhi Metallo-β-lactamases in Gram-Negative Bacteria. ACS Infect. Dis. 2019, 5, 2061–2066. [Google Scholar] [CrossRef]
  81. Liu, S.; Zhang, J.; Zhou, Y.; Hu, N.; Li, J.; Wang, Y.; Niu, X.; Deng, X.; Wang, J. Pterostilbene restores carbapenem susceptibility in New Delhi metallo-β-lactamase-producing isolates by inhibiting the activity of New Delhi metallo-β-lactamases. Br. J. Pharmacol. 2019, 176, 4548–4557. [Google Scholar] [CrossRef] [PubMed]
  82. Meng, Z.; Tang, M.-L.; Yu, L.; Liang, Y.; Han, J.; Zhang, C.; Hu, F.; Yu, J.-M.; Sun, X. Novel Mercapto Propionamide Derivatives with Potent New Delhi Metallo-β-Lactamase-1 Inhibitory Activity and Low Toxicity. ACS Infect. Dis. 2019, 5, 903–916. [Google Scholar] [CrossRef] [PubMed]
  83. Maryam, L.; Khalid, S.; Ali, A.; Khan, A.U. Synergistic effect of doripenem in combination with cefoxitin and tetracycline in inhibiting NDM-1 producing bacteria. Futur. Microbiol. 2019, 14, 671–689. [Google Scholar] [CrossRef] [PubMed]
  84. Kumar, N.G.; Kumar, G.; Mallick, S.; Ghosh, S.K.; Pramanick, P.; Ghosh, A.S. Bio-surfactin stabilised silver nanoparticles exert inhibitory effect over New-Delhi metallo-beta-lactamases (NDMs) and the cells harbouring NDMs. FEMS Microbiol. Lett. 2019, 366, fnz118. [Google Scholar] [CrossRef] [PubMed]
  85. Shi, X.-F.; Wang, M.-M.; Huang, S.-C.; Han, J.-X.; Chu, W.-C.; Xiao, C.; Zhang, E.; Qin, S. H2depda: An acyclic adjuvant potentiates meropenem activity in vitro against metallo-β-lactamase-producing enterobacterales. Eur. J. Med. Chem. 2019, 167, 367–376. [Google Scholar] [CrossRef] [PubMed]
  86. Sosibo, S.C.; Somboro, A.M.; Amoako, D.G.; Sekyere, J.O.; Bester, L.A.; Ngila, J.C.; Sun, D.D.; Kumalo, H.M. Impact of Pyridyl Moieties on the Inhibitory Properties of Prominent Acyclic Metal Chelators Against Metallo-β-Lactamase-Producing Enterobacteriaceae: Investigating the Molecular Basis of Acyclic Metal Chelators’ Activity. Microb. Drug Resist. 2019, 25, 439–449. [Google Scholar] [CrossRef] [PubMed]
  87. Su, J.; Liu, J.; Chen, C.; Zhang, Y.; Yang, K. Ebsulfur as a potent scaffold for inhibition and labelling of New Delhi metallo-β-lactamase-1 in vitro and in vivo. Bioorganic Chem. 2019, 84, 192–201. [Google Scholar] [CrossRef] [PubMed]
  88. Somboro, A.M.; Amoako, D.G.; Sekyere, J.O.; Kumalo, H.M.; Khan, R.; Bester, L.A.; Essack, S.Y. 1,4,7-Triazacyclononane Restores the Activity of β-Lactam Antibiotics against Metallo-β-Lactamase-Producing Enterobacteriaceae: Exploration of Potential Metallo-β-Lactamase Inhibitors. Appl. Environ. Microbiol. 2019, 85, e02077-18. [Google Scholar] [CrossRef] [PubMed]
  89. Liu, X.-L.; Xiang, Y.; Chen, C.; Yang, K.-W. Azolylthioacetamides as potential inhibitors of New Delhi metallo-β-lactamase-1 (NDM-1). J. Antibiot. 2019, 72, 118–121. [Google Scholar] [CrossRef] [PubMed]
  90. Somboro, A.M.; Sekyere, J.O.; Amoako, D.G.; Kumalo, H.M.; Khan, R.; Bester, L.A.; Essack, S.Y. In vitro potentiation of carbapenems with tannic acid against carbapenemase-producing enterobacteriaceae: Exploring natural products as potential carbapenemase inhibitors. J. Appl. Microbiol. 2019, 126, 452–467. [Google Scholar] [CrossRef] [PubMed]
  91. Chen, A.Y.; Thomas, P.W.; Stewart, A.C.; Bergstrom, A.; Cheng, Z.; Miller, C.; Bethel, C.R.; Marshall, S.H.; Page, R.C.; Cohen, S.M.; et al. Correction to: Dipicolinic acid derivatives as inhibitors of new delhi metallo-β-lactamase-1. J. Med. Chem. 2018, 61, 6400. [Google Scholar] [CrossRef]
  92. Liu, S.; Zhou, Y.; Niu, X.; Wang, T.; Li, J.; Liu, Z.; Wang, J.; Tang, S.; Wang, Y.; Deng, X. Magnolol restores the activity of meropenem against NDM-1-producing Escherichia coli by inhibiting the activity of metallo-beta-lactamase. Cell Death Discov. 2018, 4, 28. [Google Scholar] [CrossRef]
  93. Wang, M.-M.; Chu, W.-C.; Yang, Y.; Yang, Q.-Q.; Qin, S.-S.; Zhang, E. Dithiocarbamates: Efficient metallo-β-lactamase inhibitors with good antibacterial activity when combined with meropenem. Bioorganic Med. Chem. Lett. 2018, 28, 3436–3440. [Google Scholar] [CrossRef]
  94. Schnaars, C.; Kildahl-Andersen, G.; Prandina, A.; Popal, R.; Radix, S.L.; Le Borgne, M.; Gjøen, T.; Andresen, A.M.S.; Heikal, A.; Økstad, O.A.; et al. Synthesis and Preclinical Evaluation of TPA-Based Zinc Chelators as Metallo-β-lactamase Inhibitors. ACS Infect. Dis. 2018, 4, 1407–1422. [Google Scholar] [CrossRef]
  95. Jin, W.B.; Xu, C.; Cheng, Q.; Qi, X.L.; Gao, W.; Zheng, Z.; Chan, E.; Leung, Y.; Chan, T.; Chan, K.F.; et al. Investigation of synergistic antimicrobial effects of the drug combinations of meropenem and 1,2-benzisoselenazol-3(2H)-one derivatives on carbapenem-resistant Enterobacteriaceae producing NDM-1. Eur. J. Med. Chem. 2018, 155, 285–302. [Google Scholar] [CrossRef]
  96. Yarlagadda, V.; Sarkar, P.; Samaddar, S.; Manjunath, G.B.; Das Mitra, S.; Paramanandham, K.; Shome, B.R.; Haldar, J. Vancomycin Analogue Restores Meropenem Activity against NDM-1 Gram-Negative Pathogens. ACS Infect. Dis. 2018, 4, 1093–1101. [Google Scholar] [CrossRef]
  97. Horie, H.; Chiba, A.; Wada, S. Inhibitory effect of soy saponins on the activity of β-lactamases, including New Delhi metallo-β-lactamase 1. J. Food Sci. Technol. 2018, 55, 1948–1952. [Google Scholar] [CrossRef] [PubMed]
  98. Wang, Q.; He, Y.; Lu, R.; Wang, W.-M.; Yang, K.-W.; Fan, H.M.; Jin, Y.; Blackburn, G.M. Thermokinetic profile of NDM-1 and its inhibition by small carboxylic acids. Biosci. Rep. 2018, 38, BSR20180244. [Google Scholar] [CrossRef]
  99. Büttner, D.; Kramer, J.S.; Klingler, F.-M.; Wittmann, S.K.; Hartmann, M.R.; Kurz, C.G.; Kohnhäuser, D.; Weizel, L.; Brüggerhoff, A.; Frank, D.; et al. Challenges in the Development of a Thiol-Based Broad-Spectrum Inhibitor for Metallo-β-Lactamases. ACS Infect. Dis. 2018, 4, 360–372. [Google Scholar] [CrossRef]
  100. Cain, R.; Brem, J.; Zollman, D.; McDonough, M.A.; Johnson, R.M.; Spencer, J.; Makena, A.; Abboud, M.I.; Cahill, S.; Lee, S.Y.; et al. In Silico Fragment-Based Design Identifies Subfamily B1 Metallo-β-lactamase Inhibitors. J. Med. Chem. 2018, 61, 1255–1260. [Google Scholar] [CrossRef] [PubMed]
  101. Ning, N.-Z.; Liu, X.; Chen, F.; Zhou, P.; Hu, L.; Huang, J.; Li, Z.; Huang, J.; Li, T.; Wang, H. Embelin restores carbapenem efficacy against NDM-1-positive pathogens. Front. Microbiol. 2018, 9, 71. [Google Scholar] [CrossRef] [PubMed]
  102. Zhang, E.; Wang, M.-M.; Huang, S.-C.; Xu, S.-M.; Cui, D.-Y.; Bo, Y.-L.; Bai, P.-Y.; Hua, Y.-G.; Xiao, C.-L.; Qin, S. NOTA analogue: A first dithiocarbamate inhibitor of metallo-β-lactamases. Bioorganic Med. Chem. Lett. 2018, 28, 214–221. [Google Scholar] [CrossRef]
  103. Spyrakis, F.; Celenza, G.; Marcoccia, F.; Santucci, M.; Cross, S.; Bellio, P.; Cendron, L.; Perilli, M.; Tondi, D. Structure-based virtual screening for the discovery of novel inhibitors of New Delhi metallo-β-lactamase-1. ACS Med. Chem. Lett. 2017, 9, 45–50. [Google Scholar] [CrossRef]
  104. Sully, E.K.; Geller, B.L.; Li, L.; Moody, C.M.; Bailey, S.M.; Moore, A.L.; WONG, M.; Nordmann, P.; Daly, S.M.; Greenberg, D.E.; et al. Peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO) restores carbapenem susceptibility to NDM-1-positive pathogens in vitro and in vivo. J. Antimicrob. Chemother. 2017, 72, 782–790. [Google Scholar] [PubMed]
  105. Tehrani, K.H.M.E.; Martin, N.I. Thiol-Containing Metallo-β-Lactamase Inhibitors Resensitize Resistant Gram-Negative Bacteria to Meropenem. ACS Infect. Dis. 2017, 3, 711–717. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, J.; Wang, S.; Wei, Q.; Guo, Q.; Bai, Y.; Yang, S.; Song, F.; Zhang, L.; Lei, X. Synthesis and biological evaluation of Aspergillomarasmine A derivatives as novel NDM-1 inhibitor to overcome antibiotics resistance. Bioorganic Med. Chem. 2017, 25, 5133–5141. [Google Scholar] [CrossRef]
  107. Khan, A.U.; Ali, A.; Danishuddin; Srivastava, G.; Sharma, A. Potential inhibitors designed against NDM-1 type metallo-β-lactamases: An attempt to enhance efficacies of antibiotics against multi-drug-resistant bacteria. Sci. Rep. 2017, 7, 9207. [Google Scholar] [CrossRef] [PubMed]
  108. Chandar, B.; Poovitha, S.; Ilango, K.; MohanKumar, R.; Parani, M. Inhibition of New Delhi metallo-β-lactamase 1 (NDM-1) producing Escherichia coli IR-6 by selected plant extracts and their synergistic actions with antibiotics. Front. Microbiol. 2017, 8, 1580. [Google Scholar] [CrossRef]
  109. Brem, J.; van Berkel, S.S.; Zollman, D.; Lee, S.Y.; Gileadi, O.; McHugh, P.J.; Wash, T.R.; McDonough, M.A.; Schofield, C.J. Structural Basis of Metallo-β-Lactamase Inhibition by Captopril Stereoisomers. Antimicrob. Agents Chemother. 2016, 60, 142–150. [Google Scholar] [CrossRef]
  110. Azumah, R.; Dutta, J.; Somboro, A.; Ramtahal, M.; Chonco, L.; Parboosing, R.; Bester, L.; Kruger, H.; Naicker, T.; Essack, S.; et al. In vitro evaluation of metal chelators as potential metallo- β -lactamase inhibitors. J. Appl. Microbiol. 2016, 120, 860–867. [Google Scholar] [CrossRef]
  111. González, M.M.; Kosmopoulou, M.; Mojica, M.F.; Castillo, V.; Hinchliffe, P.; Pettinati, I.; Brem, J.; Schofield, C.J.; Mahler, G.; Bonomo, R.A.; et al. Bisthiazolidines: A Substrate-Mimicking Scaffold as an Inhibitor of the NDM-1 Carbapenemase. ACS Infect. Dis. 2016, 1, 544–554. [Google Scholar] [CrossRef]
  112. Chiou, J.; Wan, S.; Chan, K.-F.; So, P.-K.; He, D.; Chan, E.W.-C.; Chan, T.-H.; Wong, K.-Y.; Tao, J.; Chen, S. Ebselen as a potent covalent inhibitor of New Delhi metallo-β-lactamase (NDM-1). Chem. Commun. 2015, 51, 9543–9546. [Google Scholar] [CrossRef]
  113. King, A.M.; Reid-Yu, S.A.; Wang, W.; King, D.T.; De Pascale, G.; Strynadka, N.C.; Walsh, T.R.; Coombes, B.K.; Wright, G.D. Aspergillomarasmine A overcomes metallo-β-lactamase antibiotic resistance. Nature 2014, 510, 503–506. [Google Scholar] [CrossRef] [PubMed]
  114. Gan, M.; Liu, Y.; Bai, Y.; Guan, Y.; Li, L.; Gao, R.; He, W.; You, X.; Li, Y.; Yu, L.; et al. Polyketides with New Delhi metallo-β-lactamase 1 inhibitory activity from Penicillium sp. J. Nat. Prod. 2013, 76, 1535–1540. [Google Scholar] [CrossRef] [PubMed]
  115. Shen, B.; Yu, Y.; Chen, H.; Cao, X.; Lao, X.; Fang, Y.; Shi, Y.; Chen, J.; Zheng, H. Inhibitor Discovery of Full-Length New Delhi Metallo-β-Lactamase-1 (NDM-1). PLoS ONE 2013, 8, e62955. [Google Scholar] [CrossRef] [PubMed]
  116. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flowchart of the study process.
Figure 1. Flowchart of the study process.
Jcm 13 04199 g001
Figure 2. Annual numbers of eligible articles, by publication year.
Figure 2. Annual numbers of eligible articles, by publication year.
Jcm 13 04199 g002
Table 1. A summary of 91 articles reporting the 154 potential NDM inhibitors.
Table 1. A summary of 91 articles reporting the 154 potential NDM inhibitors.
No.YearNDM InhibitorsCB AssayTKC AssayKinetic AssayMolecular MethodsIn Vivo StudyToxicity AssayRef.
12022EDTA, Captopril, Ciprofloxacin×××××[25]
220221,2,4-triazole-3 thiones derivative××[26]
320221,2-Isoselenazol-3(2H) derivatives×××[27]
42022Ebselen scaffold×××[28]
52022Cephalosporin-Tripodalamin conjugate×[29]
62022Fragment-based compounds×××[30]
72022Adapelen×××[31]
82022Aromatic Schiff bases×××[32]
92022Bismuth dichloride ××××[33]
102022Alpha Lipoic acid, methimazole××××[34]
112022QDP-1 (Phenyl ring)×××[35]
122022Trans-cephalosporin××[36]
132022Withaferin A××××[37]
142022Fisetin×[38]
152022Quinolinyl-Sulphonamides sulphonyl esters×[39]
162022Emerione A, Asperfunolone A×××[40]
172022Risedronate, Methotrexate×××[41]
182022Aspergillomarasmine A analogue×××[42]
192022Unithiole derivative××[43]
202022Nitroxoline derivative××[44]
212022Indole-2-carboxylates derivative××[45]
222022Di-thiocarbamates-copper[46]
232021Alkylthio-substituted thiols derivatives××××[47]
242021H2dpa derivatives[48]
252021Thiosemicarbazone derivative×[49]
262021Thiosemicarbazones derivative×××[50]
272021N-acylhydrazones derivative×[51]
282021Azetidinimines derivatives×[52]
292021N-Sulfamoylpyrrole-2-carboxylates derivatives××××[53]
302021Indole-carboxylate derivative××××[54]
312021Cephalosporin-prodrug ×××××[55]
322021Benzimidazole and benzoxazole zinc chelator××××[56]
332021Diaryl-substituted thiosemicarbazone derivative×[57]
342021Fragment-based compound××[58]
3520212-Mercaptomethyl-thiazolidines derivative×××[59]
362021Thiosemicarbazone derivatives[60]
372021D-captopril’s derivatives××××[61]
3820204-Amino-1,2,4-triazole-3-thione-derived Schiff bases ×[62]
392020Carnosic acid×××[63]
402020Chemical peptide sequences×××[64]
412020Disulfiram, nitroxoline, 5-amino-8-hydroxyquinoline, DOTA, cyclam, TPEN ×[65]
422020ANT2681 (thiazolyl acid derivatives)×[66]
432020H2dedpa derivatives××[67]
4420201,2-benzisothiazol-3(2H) derivative×××[68]
452020Carboxylates small molecules×××××[69]
462020Metal complex scaffold (PDTC2-Fe)×××[70]
472020ZINC05683641××××[71]
482020PcephPT (cephalosporin prochelator)×××[72]
492020α-hydrazono carboxylic acid fragments××××[73]
502020Isoliquiritin××××[74]
512020Sulfamoyl hetero-arylcarboxylic acids derivatives×[75]
522020Amino-carboxylic acid analogues×××××[76]
532020Disulfiram××[77]
542020Cefmetazole×××[78]
5520203-bromopyruvate×[79]
562019Peptidomimetic 4 (PEP4)[80]
572019Pterostilbene××[81]
582019Mercapto propionamide derivatives××[82]
592019Cefoxitin, tetracycline××××[83]
602019Silver nanoparticles (AgNPs)××××[84]
612019H2-dedpa derivative×[85]
622019Tris-(2-picolyl) amine×××[86]
632019Ebsulfur scaffolds××[87]
6420191,4,7-Triazacyclononane×[88]
652019Azolyl-thio acetamides derivatives×××[89]
662019Tannic acid×××[90]
672018Dipicolinic acid derivative××[91]
682018Magnolol×××[92]
692018Di-thiocarbamate derivatives××××[93]
702018Tris-picolylamine-based zinc chelators××[94]
7120181,2-benzisoselenazol-3(2H) derivatives×[95]
722018Dipicolyl-vancomycin conjugate×××[96]
732018Crude soy saponins×××××[97]
742018Small carboxylic acid derivatives×××[98]
752018Thiol based inhibitors×××[99]
762018Fragment-based derivative××××[100]
772018Embelin××××[101]
782018Dithiocarbamate derivatives××[102]
792017Triazol-thiol derivatives××××[103]
802017Peptide-conjugated phosphorodiamidate morpholino oligomer (PPMO)××××[104]
8120172-mercapto-3-phenylpropionic acid derivative××××[105]
822017Aspergillomarasmine A derivative××××[106]
832017AW01120, BTB02323××[107]
842017Hibiscus cannabinus, Tamarindus indica, Combretum albidum, Hibiscus acetosella, Hibiscus furcatus, Punica granatum×××××[108]
852016Captopril Stereoisomers××××[109]
862016Metal chelators (1) DPA, (2) TPEN××××[110]
872016Bisthiazolidines (compound-f L-CS319)×[111]
882015Ebselen×××[112]
892014Aspergillomarasmine A××[113]
902013Polyketide compounds××××[114]
912013Thiophene-carboxylic acid derivatives×××[115]
CB, checkerboard; TKC, time-killing curve; ○ indicates a conducted assay, while × indicates that assay was not performed. Various assays were adopted for each compound.
Table 2. Detailed summary of 37 articles reporting the 47 NDM inhibitors with data for the fractional inhibitory concentration (FIC) index.
Table 2. Detailed summary of 37 articles reporting the 47 NDM inhibitors with data for the fractional inhibitory concentration (FIC) index.
No.YearTested Compounds
[Combined Drugs] (1)
** FIC Index by CB AssayTKC AssayKinetic AssayMolecular Investigation (2)In Vivo Study
(Animal)
*** Toxicity Assay (Model)Ref.
12022(1) EDTA
(2) Captopril
(3) Ciprofloxacin
[MEPM, IPM]
(1) Synergistic
(2) Synergistic and indifferent
(3) Synergistic and indifferent
-----[25]
220221, 2-Isoselenazol-3(2H) derivatives
[MEPM]
Synergistic--MDS-Not toxic (mammalian cell)[27]
32022Adapelen
[MEPM]
Synergistic and indifferentBacteriostatic-MDS--[31]
42022Bismuth dichloride (C4)
[MEPM]
Synergistic----Toxic (human embryonic kidney cell)[33]
52022(1) Alpha Lipoic acid
(2) Methimazole *
[MEPM]
All synergisticBactericidal----[34]
62022Withaferin A *
[IPM]
Synergistic--MDS--[37]
72022Fisetin *
[MEPM]
Synergistic and indifferentBactericidalPerformedMDSMouse-[38]
82022(1) Emerione A,
(2) Asperfunolone A
[MEPM, IPM, CTRX, ABPC]
---MDS--[40]
92022Nitroxoline derivative
[IPM]
SynergisticBactericidal-SAR-Non-specific (3)
(endothelial cell)
[44]
102022Di-thiocarbamates-copper (SA09-Cu)
[MEPM]
SynergisticBacteriostaticPerformedSAR MouseLess toxic (mouse)[46]
112021H2dpa derivativesAll Synergistic BactericidalPerformedMDSMouseLess toxic (mouse)[48]
122021Thiosemicarbazone derivative [MEPM]Synergistic BacteriostaticPerformedMDS--[49]
132021Indole-carboxylate derivative
[MEPM]
Synergistic--ITC --[54]
142021Cephalosporin-prodrug
[MEPM]
Synergistic -----[55]
1520201,2-benzisothiazol-3(2H) derivative [MEPM]Synergistic --MDS, ESI-MS-Acceptable toxicity
(human embryonic kidney cell)
[68]
162020Carboxylates small molecules
[MEPM]
Synergistic and indifferent-----[69]
172020ZINC05683641
[MEPM]
Synergistic--MDS--[71]
182020Isoliquiritin *
[MEPM]
Synergistic and indifferentBactericidal----[74]
192020Sulfamoyl hetero-arylcarboxylic acid derivatives
[MEPM]
All synergistic-PerformedProtein CrystallizationMouseLess toxic (mouse)[75]
202020Aminocarboxylic acid analogues
[MEPM]
All synergistic-----[76]
212020Cefmetazole *
[MEPM]
SynergisticBactericidalPerformed---[78]
222019Peptidomimetic 4 (PEP4)
[MEPM]
Synergistic and indifferentBactericidalPerformedMDSMouseNon-specific (3)
(mammalian cell)
[80]
232019Pterostilbene *
[MEPM]
Synergistic and indifferentBacteriostatic-MDSMouse-[81]
242019Mercapto propionamide derivative
[MEPM]
All synergistic--X-ray crystallography MouseNon-specific (3)
(mouse)
[82]
252019(1) Cefoxitin *
(2) Tetracycline *
[DRPM]
All Synergistic-Performed --[83]
262019Tris-(2-picolyl) amine (TPA)
[MEPM]
SynergisticBactericidal-MDS--[86]
2720191,4,7-Triazacyclononane
[MEPM]
SynergisticBactericidalPerformedMDS -Non-specific (3)
(immortalized liver carcinoma cells)
[88]
282018Magnolol
[MEPM]
SynergisticBactericidal-MDS--[92]
2920181,2-benzisoselenazol-3(2H) derivatives [MEPM]Synergistic and indifferent-PerformedESI-MSMouseLess toxic (larvae)[95]
302018Vancomycin analogue (dipicolyl-vancomycin conjugate)
[MEPM]
Synergistic---MouseNon-specific (3)
(mouse model, mammalian cell)
[96]
312018Crude soy saponins
[PIPC, ABPC, MPIPC, PCG]
Synergistic-----[97]
322018Embelin
[IPM]
Synergistic--MDS --[101]
332017Triazol-thiol derivatives
[CTX, MEPM]
All synergistic-Performed---[103]
3420172- mercapto-3-phenylpropionic acid derivative
[MEPM]
Synergistic--ITC--[105]
352017Aspergillomarasmine A derivatives
[MEPM]
All synergistic-----[106]
362017(1) Hibiscus cannabinus
(2) Tamarindus indica
(3) Combretum albidum
(4) Hibiscus acetosella
(5) Hibiscus furcatus
(6) Punica granatum
[MEPM]
All synergistic-----[108]
372014Aspergillomarasmine A
[MEPM]
Synergistic-PerformedICP-MSMouse-[113]
CB, checkerboard; TKC, time-killing curve. (1) Abbreviations of combined drugs: MEPM, meropenem; IPM, imipenem; CTRX, ceftriaxone; ABPC, ampicillin; DRPM, doripenem; PIPC, piperacillin; MPIPC, oxacillin; PCG, benzylpenicillin; CTX, cefotaxime. (2) Abbreviations of methods: MDS, molecular docking and molecular dynamic simulation; SAR, structural activity relationship analysis; ESI-MS, electrospray ionization mass spectrometry; ITC, isothermal titration assay; ICP-MS, inductively coupled mass spectrometry. (3) Non-lethal doses were used. * FDA-approved drug. ** Synergistic effect was determined as that with an FIC index of ≤0.5. *** “Not toxic” was defined as those without any side effects shown in the experimental model. “Less toxic” was defined as when any signs of drug-associated adverse effects were observed.
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.

Share and Cite

MDPI and ACS Style

Nahar, L.; Hagiya, H.; Gotoh, K.; Asaduzzaman, M.; Otsuka, F. New Delhi Metallo-Beta-Lactamase Inhibitors: A Systematic Scoping Review. J. Clin. Med. 2024, 13, 4199. https://doi.org/10.3390/jcm13144199

AMA Style

Nahar L, Hagiya H, Gotoh K, Asaduzzaman M, Otsuka F. New Delhi Metallo-Beta-Lactamase Inhibitors: A Systematic Scoping Review. Journal of Clinical Medicine. 2024; 13(14):4199. https://doi.org/10.3390/jcm13144199

Chicago/Turabian Style

Nahar, Lutfun, Hideharu Hagiya, Kazuyoshi Gotoh, Md Asaduzzaman, and Fumio Otsuka. 2024. "New Delhi Metallo-Beta-Lactamase Inhibitors: A Systematic Scoping Review" Journal of Clinical Medicine 13, no. 14: 4199. https://doi.org/10.3390/jcm13144199

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop