NDM-Producing E coli Harboring PBP Insert Variants Emerges as New Threat

News
Article
ContagionContagion, Summer 2024 Digital Edition
Volume 09
Issue 02

As with other metallo-β-lactamases, NDM is capable of hydrolyzing nearly all β-lactams, including carbapenems, and the search for the proper antimicrobials is challenging.

bacteria; Image Credit: Adobe Stock

Image Credit: Adobe Stock

New Delhi metallo-β-lactamase (NDM) was first detected in 2009 in Klebsiella pneumoniae.1 Since then, the blaNDM genes, responsible for encoding NDM-type enzymes, have been identified in Acinetobacter, Pseudomonas, and, most commonly, Escherichia coli and other Enterobacterales bacteria. As with other metallo-β-lactamases (MBLs), NDM is capable of hydrolyzing nearly all β-lactams, including carbapenems but notably not aztreonam. Data from the Centers for Disease Control and Prevention from 2017 to 2019 revealed that nearly 9% of all carbapenem-resistant Enterobacterales and approximately 1% of Pseudomonas isolates in the US harbored blaNDM, a figure expected to increase over time.2

The emergence and dissemination of NDM are facilitated by various factors, including increased global travel, widespread antibiotic usage, and inadequate infection control measures in health care settings.3 The Infectious Diseases Society of America recommends β-lactam antibiotics as a treatment option for NDM-producing organisms, specifically ceftazidime/avibactam in combination with aztreonam or cefiderocol as monotherapy.4 Cefiderocol is a novel siderophore-conjugated cephalosporin that harnesses the bacterial iron transport system to access the periplasmic compartment. Cefiderocol is stable to hydrolysis by a broad array of β-lactamases, including MBLs.5 In the CREDIBLE - CR study (NCT02714595), cefiderocol was seen to be effective in the treatment of organisms producing MBLs.6,7 Although aztreonam itself is active against MBL-producing organisms due to stability to hydrolysis by MBLs,8,9 it remains susceptible to hydrolysis by extended-spectrum β-lactamases, AmpC β-lactamases, KPCs, or OXA-48–like carbapenemases, which are commonly coproduced alongside NDMs.9 Partnering aztreonam with an inhibitor of these enzymes, such as avibactam, prevents hydrolysis of aztreonam and allows for activity against MBL-producing organisms. Indeed, aztreonam paired with ceftazidime/avibactam is highly effective in the treatment of infections caused by MBL-producing organisms.10 However, emerging resistance to these frontline treatment options poses a growing challenge in managing infections caused by NDM-producing organisms, complicating treatment strategies.3

β-lactam antibiotics exert bactericidal effects by disrupting the formation of bacterial cell walls through covalent binding to crucial penicillin-binding proteins (PBPs). These enzymes play a vital role in the final stages of peptidoglycan cross-linking. In gram-negative bacteria, crucial PBPs are PBP1a and PBP1b, which participate in cell lysis; PBP2, whose inhibition halts cell division; and PBP3, whose inhibition arrests cell division, causing filamentation. Inhibition of 1 or more of these PBPs can lead to cell death.11 Cephalosporins preferentially target PBP3 and PBP1a, whereas aztreonam binds exclusively to PBP2, PBP3, and PBP4. Resistance to β-lactams in Enterobacterales, including to cefiderocol, is often mediated through β-lactamases, which hydrolyze the functional β-lactam ring and prevent binding to PBPs.12 However, NDM-5–producing E coli bacteria harboring PBP3 variants, specifically YRIN or YRIK insertions after residue 333 (333YRIN/K334), are increasingly prevalent worldwide and account for nearly a quarter of NDM-producing E coli bacteria in India.13-15 Additionally, E coli bacteria that carry PBP3 variants frequently harbor variants of CMY-42, a transferable AmpC enzyme.16 Together, these mechanisms lead to dramatically increased minimum inhibitory concentrations (MICs) to both aztreonam/ avibactam and cefiderocol.13-15,17-19 Concerningly, these enzymes and PBP mutations are now frequently identified in a successful E coli clone, ST167, which has begun to spread globally.20 Sadek et al completed whole-gene sequencing of 19 NDM-producing E coli isolates exhibiting high MICs for aztreonam/avibactam (> 4 μg/mL). All the isolates had either a 333YRIK or YRIN334 insertion, with YRIN being more commonly observed.21

Additionally, each isolate carried a plasmid-borne CMY β-lactamase gene, most commonly CMY-42. The MIC of aztreonam/avibactam was 4-fold higher for the CMY-42–producing isolates compared with CMY-2. Livermore et al specifically assessed the independent contribution of different insert variants and CMY enzymes to the aztreonam/ avibactam MIC.19 Isolates harboring YRIN were typically less resistant than those harboring YRIK, with MICs generally in the range of 0.5 to 2 μg/mL for those harboring YRIN vs at least 8 μg/mL for those harboring YRIK. CMY-42 and other CMY variants led to an additional increase in MIC of 1- or 2-fold relative to isolates with the insertion alone.

Mutations in PBPs pose a relatively unique challenge in antimicrobial drug development. In contrast to β-lactamase– mediated resistance, where addition of a β-lactamase inhibitor allows for restoration of activity for the parent compound, this approach does not work for PBP mutations. The addition of a β-lactamase inhibitor alone does not restore activity of the partner β-lactam compound due to the presence of non–β-lactamase–mediated resistance. For example, cefepime/ taniborbactam is a broad-spectrum cephalosporin/ boronic acid–based β-lactamase inhibitor in late-stage development specifically for the treatment of MBL-producing organisms.22 Although taniborbactam usually readily inhibits most NDM variants, inhibition of NDM-5 is insufficient to restore activity of cefepime/taniborbactam against PBP3 mutations due to inability of cefepime to bind to PBP3.23,24 This is in contrast to organisms harboring NDM-9 or NDM-30, which are NDM variants resistant to inhibition by taniborbactam. In these isolates, cefepime is hydrolyzed before it has the opportunity to interact with PBP3. Therefore, the activity of any MBL-active β-lactamase inhibitor combination will depend not only on the spectrum of the inhibitor but also on the PBP-binding spectrum of the parent β-lactam compound and specifically inhibition of PBPs other than PBP3.

Several such compounds are in development or commercially available. Among these, the compound with the longest track record of use is mecillinam or the oral prodrug pivmecillinam. Mecillinam is a β-lactam that inhibits PBP2 and is widely used in Scandinavia for treatment of urinary tract infection. Pivmecillinam was recently approved by the US Food and Drug Administration in the US for treatment of uncomplicated urinary tract infection.25 Mecillinam is generally active against a broad array of β-lactamase– producing Enterobacterales bacteria, including those producing NDM enzymes.26 Zidebactam, a novel diazabicycloooctane (DBO) β-lactamase inhibitor, serves a dual role as both a broad-spectrum inhibitor of serine β-lactamases and β-lactam enhancer due to its complementary inhibition of PBP2.27 Cefepime/ zidebactam is highly active in vitro against NDM-producing E coli bacteria harboring PBP insert variants.28,29 Durlobactam, another broad-spectrum DBO inhibitor, is similarly active against NDM-producing E coli bacteria with PBP inserts, although whether it has an enhancer effect similar to zidebactam is presently unknown.30,31 Historically, rapid development of resistance to PBP2 inhibitors has been a significant concern, and thus, any successful use of these compounds is likely dependent on partnering with a drug with some residual inhibition of PBP3 or other PBPs, such as carbapenems or aztreonam.32

In conclusion, the emergence and spread of NDM-producing organisms harboring PBP insert mutations pose significant threats to existing therapeutics. Efforts to address PBP-mediated resistance require a multifaceted approach, encompassing enhanced surveillance, stringent infection control measures, and the development of novel therapeutics targeting resistance mechanisms. Furthermore, continued research into the molecular basis of resistance and the identification of new drug targets are essential to stay ahead of antimicrobial resistance.

References
1. Yong D, Toleman MA, Giske CG, et al. Characterization of a new metallo-beta-lactamase gene, bla(NDM-1), and a novel erythromycin esterase gene carried on a unique genetic structure in Klebsiella pneumoniae sequence type 14 from India. Antimicrob Agents Chemother. 2009;53(12):5046-5054. doi:10.1128/aac.00774-09
2. Sabour S, Huang JY, Bhatnagar A, et al. Detection and characterization of targeted carbapenem-resistant health care-associated threats: findings from the Antibiotic Resistance Laboratory Network, 2017 to 2019. Antimicrob Agents Chemother. 2021;65(12):e0110521. doi:10.1128/AAC.01105-21
3.Khan AU, Maryam L, Zarrilli R. Structure, genetics and worldwide spread of New Delhi metallo-β-lactamase (NDM): a threat to public health. BMC Microbiol. 2017;17(1):101. doi:10.1186/s12866-017-1012-8
4. Tamma PD, Aitken SL, Bonomo RA, Mathers AJ, van Duin D, Clancy CJ. Infectious Diseases Society of America 2023 guidance on the treatment of antimicrobial resistant gram-negative infections. Clin Infect Dis. Published online July 18, 2023. doi:10.1093/cid/ciad428
5. Sato T, Yamawaki K. Cefiderocol: discovery, chemistry, and in vivo profiles of a novel siderophore cephalosporin. Clin Infect Dis. 2019;69(suppl 7):S538-S543. doi:10.1093/cid/ciz826
6. Timsit JF, Paul M, Shields RK, et al. Cefiderocol for the treatment of infections due to metallo-B-lactamase-producing pathogens in the CREDIBLE-CR and APEKS-NP phase 3 randomized studies. Clin Infect Dis. 2022;75(6):1081-1084. doi:10.1093/cid/ciac078
7. Bassetti M, Echols R, Matsunaga Y, et al. Efficacy and safety of cefiderocol or best available therapy for the treatment of serious infections caused by carbapenem-resistant gram-negative bacteria (CREDIBLE-CR): a randomised, open-label, multicentre, pathogen-focused, descriptive, phase 3 trial. Lancet Infect Dis. 2021;21(2):226-240. doi:10.1016/S1473-3099(20)30796-9
8. Poeylaut-Palena AA, Tomatis PE, Karsisiotis AI, Damblon C, Mata EG, Vila AJ. A minimalistic approach to identify substrate binding features in B1 metallo-beta-lactamases. Bioorg Med Chem Lett. 2007;17(18):5171-5174. doi:10.1016/j.bmcl.2007.06.089
9. Drawz SM, Papp-Wallace KM, Bonomo RA. New β-lactamase inhibitors: a therapeutic renaissance in an MDR world. Antimicrob Agents Chemother. 2014;58(4):1835-1846. doi:10.1128/AAC.00826-13
10. Falcone M, Daikos GL, Tiseo G, et al. Efficacy of ceftazidime-avibactam plus aztreonam in patients with bloodstream infections caused by metallo-β-lactamase-producing Enterobacterales. Clin Infect Dis. 2021;72(11):1871-1878. doi:10.1093/cid/ciaa586
11. Zapun A, Contreras-Martel C, Vernet T. Penicillin-binding proteins and beta-lactam resistance. FEMS Microbiol Rev. 2008;32(2):361-385. doi:10.1111/j.1574-6976.2007.00095.x
12. Simner PJ, Mostafa HH, Bergman Y, et al. Progressive development of cefiderocol resistance in Escherichia coli during therapy is associated with an increase in blaNDM-5 copy number and gene expression. Clin Infect Dis. 2022;75(1):47-54. doi:10.1093/cid/ciab888
13. Alm RA, Johnstone MR, Lahiri SD. Characterization of Escherichia coli NDM isolates with decreased susceptibility to aztreonam/avibactam: role of a novel insertion in PBP3. J Antimicrob Chemother. 2015;70(5):1420-1428. doi:10.1093/jac/dku568
14. Periasamy H, Joshi P, Palwe S, Shrivastava R, Bhagwat S, Patel M. High prevalence of Escherichia coli clinical isolates in India harbouring four amino acid inserts in PBP3 adversely impacting activity of aztreonam/avibactam. J Antimicrob Chemother. 2020;75(6):1650-1651. doi:10.1093/jac/dkaa021
15. Zhang Y, Kashikar A, Brown CA, Denys G, Bush K. Unusual Escherichia coli PBP 3 insertion sequence identified from a collection of carbapenem-resistant Enterobacteriaceae tested in vitro with a combination of ceftazidime-, ceftaroline-, or aztreonam-avibactam. Antimicrob Agents Chemother. 2017;61(8):e00389-17. doi:10.1128/AAC.00389-17
16. Sadek M, Ruppe E, Habib A, Zahra R, Poirel L, Nordmann P. International circulation of aztreonam/avibactam-resistant NDM-5-producing Escherichia coli isolates: successful epidemic clones. J Glob Antimicrob Resist. 2021;27:326-328. doi:10.1016/j.jgar.2021.09.016
17. Tamma PD, Munita JM. The metallo-β-lactamases strike back: emergence of taniborbactam escape variants. Antimicrob Agents Chemother. 2024;68(2):e0151023. doi:10.1128/aac.01510-23
18. Nordmann P, Yao Y, Falgenhauer L, Sadek M, Imirzalioglu C, Chakraborty T. Recent emergence of aztreonam-avibactam resistance in NDM and OXA-48 carbapenemase-producing Escherichia coli in Germany. Antimicrob Agents Chemother. 2021;65(11):e0109021. doi:10.1128/AAC.01090-21
19. Livermore DM, Mushtaq S, Vickers A, Woodford N. Activity of aztreonam/avibactam against metallo-β-lactamase-producing Enterobacterales from the UK: impact of penicillin-binding protein-3 inserts and CMY-42 β-lactamase in Escherichia coli. Int J Antimicrob Agents. 2023;61(5):106776. doi:10.1016/j.ijantimicag.2023.106776
20. Garcia-Fernandez A, Villa L, Bibbolino G, et al. Novel insights and features of the NDM-5-producing Escherichia coli sequence type 167 high-risk clone. mSphere. 2020;5(2):e00269-20. doi:10.1128/mSphere.00269-20
21. Sadek M, Juhas M, Poirel L, Nordmann P. Genetic features leading to reduced susceptibility to aztreonam-avibactam among metallo-β-lactamase-producing Escherichia coli isolates. Antimicrob Agents Chemother. 2020;64(12):e01659-20. doi:10.1128/AAC.01659-20
22. Wagenlehner FM, Gasink LB, McGovern PC, et al. Cefepime-taniborbactam in complicated urinary tract infection. N Engl J Med. 2024;390(7):611-622. doi:10.1056/NEJMoa2304748
23. Le Terrier C, Nordmann P, Buchs C, Poirel L. Effect of modification of penicillin-binding protein 3 on susceptibility to ceftazidime-avibactam, imipenem-relebactam, meropenem-vaborbactam, aztreonam-avibactam, cefepime-taniborbactam, and cefiderocol of Escherichia coli strains producing broad-spectrum β-lactamases. Antimicrob Agents Chemother. 2024;68(4):e0154823. doi:10.1128/aac.01548-23
24. Le Terrier C, Viguier C, Nordmann P, Vila AJ, Poirel L. Relative inhibitory activities of the broad-spectrum β-lactamase inhibitor taniborbactam against metallo-β-lactamases. Antimicrob Agents Chemother. 2024;68(2):e0099123. doi:10.1128/aac.00991-23
25. Jansåker F, Frimodt-Møller N, Benfield TL, Knudsen JD. Mecillinam for the treatment of acute pyelonephritis and bacteremia caused by Enterobacteriaceae: a literature review. Infect Drug Resist. 2018;11:761-771. doi:10.2147/IDR.S163280
26. Emeraud C, Godmer A, Girlich D, et al. Activity of mecillinam against carbapenem-resistant Enterobacterales. J Antimicrob Chemother. 2022;77(10):2835-2839. doi:10.1093/jac/dkac226
27. Lepak AJ, Zhao M, Andes DR. WCK 5222 (cefepime/zidebactam) pharmacodynamic target analysis against metallo-β-lactamase producing Enterobacteriaceae in the neutropenic mouse pneumonia model. Antimicrob Agents Chemother. 2019;63(12):e01648-19. doi:10.1128/AAC.01648-19
28. Le Terrier C, Nordmann P, Sadek M, Poirel L. In vitro activity of cefepime/zidebactam and cefepime/taniborbactam against aztreonam/avibactam-resistant NDM-like-producing Escherichia coli clinical isolates. J Antimicrob Chemother. 2023;78(5):1191-1194. doi:10.1093/jac/dkad061
29. Bhagwat SS, Hariharan P, Joshi PR, et al. Activity of cefepime/zidebactam against MDR Escherichia coli isolates harbouring a novel mechanism of resistance based on four-amino-acid inserts in PBP3. J Antimicrob Chemother. 2020;75(12):3563-3567. doi:10.1093/jac/dkaa353
30. Aitken SL, Pierce VM, Pogue JM, Kline EG, Tverdek FP, Shields RK. The growing threat of NDM-producing E. coli with penicillin-binding protein 3 mutations in the United States - is there a potential role for durlobactam? Clin Infect Dis. Published online April 25, 2024. doi:10.1093/cid/ciae229
31. Fouad A, Nicolau DP, Gill CM. In vitro synergy of the combination of sulbactam-durlobactam and cefepime at clinically relevant concentrations against A. baumannii, P. aeruginosa and Enterobacterales. J Antimicrob Chemother. 2023;78(12):2801-2809. doi:10.1093/jac/dkad244
32. Kresken M, Pfeifer Y, Wagenlehner F, Werner G, Wohlfarth E; Study Group ‘Antimicrobial Resistance’ of the Paul Ehrlich Society for Infection Therapy. Resistance to mecillinam and nine other antibiotics for oral use in Escherichia coli isolated from urine specimens of primary care patients in Germany, 2019/20. Antibiotics (Basel). 2022;11(6):751. doi:10.3390/antibiotics11060751
Recent Videos
© 2024 MJH Life Sciences

All rights reserved.