1
|
Longo BM, Trunfio M, Calcagno A. Dual β-lactams for the treatment of Mycobacterium abscessus: a review of the evidence and a call to act against an antibiotic nightmare. J Antimicrob Chemother 2024; 79:2731-2741. [PMID: 39150384 DOI: 10.1093/jac/dkae288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
Mycobacterium abscessus complex is a group of rapidly growing non-tuberculous mycobacteria (NTM), increasingly emerging as opportunistic pathogens. Current treatment options for these microorganisms are limited and associated with a high rate of treatment failure, toxicity and recurrence. In search of new therapeutic strategies, interest has grown in dual β-lactam (DBL) therapy, as research recently discovered that M. abscessus cell wall synthesis is mainly regulated by two types of enzymes (d,d-transpeptidases and l,d-transpeptidases) differently susceptible to inhibition by distinct β-lactams. In vitro studies testing several DBL combinations have shown synergy in extracellular broth cultures as well as in the intracellular setting: cefoxitin/imipenem, ceftaroline/imipenem, ceftazidime/ceftaroline and ceftazidime/imipenem. The addition of specific β-lactamase inhibitors (BLIs) targeting M. abscessus β-lactamase did not significantly enhance the activity of DBL combinations. However, in vivo data are lacking. We reviewed the literature on DBL/DBL-BLI-based therapies for M. abscessus infections to raise greater attention on this promising yet overlooked treatment option and to guide future preclinical and clinical studies.
Collapse
Affiliation(s)
- Bianca Maria Longo
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
| | - Mattia Trunfio
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, CA 92037, USA
| | - Andrea Calcagno
- Department of Medical Sciences, Unit of Infectious Diseases, Amedeo di Savoia Hospital, University of Turin, 10149 Turin, Italy
| |
Collapse
|
2
|
Nägeli M, Rodriguez S, Manson AL, Earl AM, Brennan-Krohn T. Rapid Emergence of Resistance to Broad-Spectrum Direct Antimicrobial Activity of Avibactam. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615047. [PMID: 39386481 PMCID: PMC11463622 DOI: 10.1101/2024.09.25.615047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Avibactam (AVI) is a diazabicyclooctane (DBO) β-lactamase inhibitor used clinically in combination with ceftazidime. At concentrations higher than those typically achieved in vivo, it also has broad-spectrum direct antibacterial activity against Enterobacterales strains, including metallo-β-lactamase-producing isolates, mediated by inhibition of penicillin-binding protein 2 (PBP2). This activity is mechanistically similar to that of more potent novel DBOs (zidebactam, nacubactam) in late clinical development. We found that resistance to AVI emerged readily, with a mutation frequency of 2×10-6 to 8×10-5. Whole genome sequencing of resistant isolates revealed a heterogeneous mutational target that permitted bacterial survival and replication despite PBP2 inhibition, in line with prior studies of PBP2-targeting drugs. While such mutations are believed to act by upregulating the bacterial stringent response, we found a similarly high mutation frequency in bacteria deficient in components of the stringent response, although we observed a different set of mutations in these strains. Although avibactam-resistant strains had increased lag time, suggesting a fitness cost that might render them less problematic in clinical infections, there was no statistically significant difference in growth rates between susceptible and resistant strains. The finding of rapid emergence of resistance to avibactam as the result of a large mutational target has important implications for novel DBOs with potent direct antibacterial activity, which are being developed with the goal of expanding cell wall-active treatment options for multidrug-resistant gram-negative infections but may be vulnerable to treatment-emergent resistance.
Collapse
Affiliation(s)
- Michelle Nägeli
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Shade Rodriguez
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Abigail L. Manson
- Infectious Disease & Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashlee M. Earl
- Infectious Disease & Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Thea Brennan-Krohn
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Lyons N, Wu W, Jin Y, Lamont IL, Pletzer D. Using host-mimicking conditions and a murine cutaneous abscess model to identify synergistic antibiotic combinations effective against Pseudomonas aeruginosa. Front Cell Infect Microbiol 2024; 14:1352339. [PMID: 38808066 PMCID: PMC11130353 DOI: 10.3389/fcimb.2024.1352339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Antibiotic drug combination therapy is critical for the successful treatment of infections caused by multidrug resistant pathogens. We investigated the efficacy of β-lactam and β-lactam/β-lactamase inhibitor combinations with other antibiotics, against the hypervirulent, ceftazidime/avibactam resistant Pseudomonas aeruginosa Liverpool epidemic strain (LES) B58. Although minimum inhibitory concentrations in vitro differed by up to eighty-fold between standard and host-mimicking media, combinatorial effects only marginally changed between conditions for some combinations. Effective combinations in vitro were further tested in a chronic, high-density murine infection model. Colistin and azithromycin demonstrated combinatorial effects with ceftazidime and ceftazidime/avibactam both in vitro and in vivo. Conversely, while tobramycin and tigecycline exhibited strong synergy in vitro, this effect was not observed in vivo. Our approach of using host-mimicking conditions and a sophisticated animal model to evaluate drug synergy against bacterial pathogens represents a promising approach. This methodology may offer insights into the prediction of combination therapy outcomes and the identification of potential treatment failures.
Collapse
Affiliation(s)
- Nikita Lyons
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Weihui Wu
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yongxin Jin
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Iain L. Lamont
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Daniel Pletzer
- Department of Microbiology and Immunology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
4
|
Nantongo M, Nguyen DC, Bethel CR, Taracila MA, Li Q, Dousa KM, Shin E, Kurz SG, Nguyen L, Kreiswirth BN, Boom WH, Plummer MS, Bonomo RA. Durlobactam, a Diazabicyclooctane β-Lactamase Inhibitor, Inhibits BlaC and Peptidoglycan Transpeptidases of Mycobacterium tuberculosis. ACS Infect Dis 2024; 10:1767-1779. [PMID: 38619138 DOI: 10.1021/acsinfecdis.4c00119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Peptidoglycan synthesis is an underutilized drug target in Mycobacterium tuberculosis (Mtb). Diazabicyclooctanes (DBOs) are a class of broad-spectrum β-lactamase inhibitors that also inhibit certain peptidoglycan transpeptidases that are important in mycobacterial cell wall synthesis. We evaluated the DBO durlobactam as an inhibitor of BlaC, the Mtb β-lactamase, and multiple Mtb peptidoglycan transpeptidases (PonA1, LdtMt1, LdtMt2, LdtMt3, and LdtMt5). Timed electrospray ionization mass spectrometry (ESI-MS) captured acyl-enzyme complexes with BlaC and all transpeptidases except LdtMt5. Inhibition kinetics demonstrated durlobactam was a potent and efficient DBO inhibitor of BlaC (KI app 9.2 ± 0.9 μM, k2/K 5600 ± 560 M-1 s-1) and similar to clavulanate (KI app 3.3 ± 0.6 μM, k2/K 8400 ± 840 M-1 s-1); however, durlobactam had a lower turnover number (tn = kcat/kinact) than clavulanate (1 and 8, respectively). KI app values with durlobactam and clavulanate were similar for peptidoglycan transpeptidases, but ESI-MS captured durlobactam complexes at more time points. Molecular docking and simulation demonstrated several productive interactions of durlobactam in the active sites of BlaC, PonA1, and LdtMt2. Antibiotic susceptibility testing was conducted on 11 Mtb isolates with amoxicillin, ceftriaxone, meropenem, imipenem, clavulanate, and durlobactam. Durlobactam had a minimum inhibitory concentration (MIC) range of 0.5-16 μg/mL, similar to the ranges for meropenem (1-32 μg/mL) and imipenem (0.5-64 μg/mL). In β-lactam + durlobactam combinations (1:1 mass/volume), MICs were lowered 4- to 64-fold for all isolates except one with meropenem-durlobactam. This work supports further exploration of novel β-lactamase inhibitors that target BlaC and Mtb peptidoglycan transpeptidases.
Collapse
Affiliation(s)
- Mary Nantongo
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
| | - David C Nguyen
- Division of Infectious Diseases, Department of Pediatrics and Division of Infectious Diseases, and Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois 60612, United States
| | - Christopher R Bethel
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
| | - Magdalena A Taracila
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Qing Li
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Khalid M Dousa
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Medical Service, Veterans Affairs Northeast Ohio Healthcare System (VANEOHS), Cleveland, Ohio 44106, United States
| | - Eunjeong Shin
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Sebastian G Kurz
- Department of Internal Medicine VIII, Medical Oncology and Pneumology, University of Tübingen, 72076 Tübingen, Germany
| | - Liem Nguyen
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Barry N Kreiswirth
- Center for Discovery and Innovation, Hackensack, New Jersey 07110, United States
| | - W Henry Boom
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
| | - Mark S Plummer
- Biopharmaworks, Groton, Connecticut 06340, United States
| | - Robert A Bonomo
- Department of Molecular Biology and Microbiology, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Department of Medicine, Case Western Reserve University (CWRU), Cleveland, Ohio 44106, United States
- Medical Service, Veterans Affairs Northeast Ohio Healthcare System (VANEOHS), Cleveland, Ohio 44106, United States
- CWRU-Cleveland VAMC Center for Antibiotic Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio 44106, United States
- Departments of Biochemistry, Pharmacology, and Proteomics and Bioinformatics, CWRU, Cleveland, Ohio 44106, United States
- Cleveland Geriatrics Research Education and Clinical Center (GRECC), VANEOHS, Cleveland, Ohio 44106, United States
| |
Collapse
|
5
|
Jacobs LMC, Consol P, Chen Y. Drug Discovery in the Field of β-Lactams: An Academic Perspective. Antibiotics (Basel) 2024; 13:59. [PMID: 38247618 PMCID: PMC10812508 DOI: 10.3390/antibiotics13010059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024] Open
Abstract
β-Lactams are the most widely prescribed class of antibiotics that inhibit penicillin-binding proteins (PBPs), particularly transpeptidases that function in peptidoglycan synthesis. A major mechanism of antibiotic resistance is the production of β-lactamase enzymes, which are capable of hydrolyzing β-lactam antibiotics. There have been many efforts to counter increasing bacterial resistance against β-lactams. These studies have mainly focused on three areas: discovering novel inhibitors against β-lactamases, developing new β-lactams less susceptible to existing resistance mechanisms, and identifying non-β-lactam inhibitors against cell wall transpeptidases. Drug discovery in the β-lactam field has afforded a range of research opportunities for academia. In this review, we summarize the recent new findings on both β-lactamases and cell wall transpeptidases because these two groups of enzymes are evolutionarily and functionally connected. Many efforts to develop new β-lactams have aimed to inhibit both transpeptidases and β-lactamases, while several promising novel β-lactamase inhibitors have shown the potential to be further developed into transpeptidase inhibitors. In addition, the drug discovery progress against each group of enzymes is presented in three aspects: understanding the targets, screening methodology, and new inhibitor chemotypes. This is to offer insights into not only the advancement in this field but also the challenges, opportunities, and resources for future research. In particular, cyclic boronate compounds are now capable of inhibiting all classes of β-lactamases, while the diazabicyclooctane (DBO) series of small molecules has led to not only new β-lactamase inhibitors but potentially a new class of antibiotics by directly targeting PBPs. With the cautiously optimistic successes of a number of new β-lactamase inhibitor chemotypes and many questions remaining to be answered about the structure and function of cell wall transpeptidases, non-β-lactam transpeptidase inhibitors may usher in the next exciting phase of drug discovery in this field.
Collapse
Affiliation(s)
| | | | - Yu Chen
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (L.M.C.J.); (P.C.)
| |
Collapse
|
6
|
Lomovskaya O, Castanheira M, Lindley J, Rubio-Aparicio D, Nelson K, Tsivkovski R, Sun D, Totrov M, Loutit J, Dudley M. 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. Antimicrob Agents Chemother 2023; 67:e0044023. [PMID: 37800963 PMCID: PMC10648875 DOI: 10.1128/aac.00440-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/07/2023] [Indexed: 10/07/2023] Open
Abstract
Recently, several β-lactam (BL)/β-lactamase inhibitor (BLI) combinations have entered clinical testing or have been marketed for use, but limited direct comparative studies of their in vitro activity exist. Xeruborbactam (XER, also known as QPX7728), which is undergoing clinical development, is a cyclic boronate BLI with potent inhibitory activity against serine (serine β-lactamase) and metallo-β-lactamases (MBLs). The objectives of this study were (i) to compare the potency and spectrum of β-lactamase inhibition by various BLIs in biochemical assays using purified β-lactamases and in microbiological assays using the panel of laboratory strains expressing diverse serine and metallo-β-lactamases and (ii) to compare the in vitro potency of XER in combination with multiple β-lactam antibiotics to that of other BL/BLI combinations in head-to-head testing against recent isolates of carbapenem-resistant Enterobacterales (CRE). Minimal inhibitory concentrations (MICs) of XER combinations were tested with XER at fixed 4 or 8 µg/mL, and MIC testing was conducted in a blinded fashion using Clinical and Laboratory Standards Institute reference methods. Xeruborbactam and taniborbactam (TAN) were the only BLIs that inhibited clinically important MBLs. The spectrum of activity of xeruborbactam included several MBLs identified in Enterobacterales, e.g., and various IMP enzymes and NDM-9 that were not inhibited by taniborbactam. Xeruborbactam potency against the majority of purified β-lactamases was the highest in comparison with other BLIs. Meropenem-xeruborbactam (MEM-XER, fixed 8 µg/mL) was the most potent combination against MBL-negative CRE with MIC90 values of 0.125 µg/mL. MEM-XER and cefepime-taniborbactam (FEP-TAN) were the only BL/BLIs with activity against MBL-producing CREs; with MEM-XER (MIC90 of 1 µg/mL) being at least 16-fold more potent than FEP-TAN (MIC90 of 16 µg/mL). MEM-XER MIC values were ≤8 µg/mL for >90% of CRE, including both MBL-negative and MBL-positive isolates, with FEP-TAN MIC of >8 µg/mL. Xeruborbactam also significantly enhanced potency of other β-lactam antibiotics, including cefepime, ceftolozane, ceftriaxone, aztreonam, piperacillin, and ertapenem, against clinical isolates of Enterobacterales that carried various class A, class C, and class D extended-spectrum β-lactamases and carbapenem-resistant Enterobacterales, including metallo-β-lactamase-producing isolates. These results strongly support further clinical development of xeruborbactam combinations.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dongxu Sun
- Qpex Biopharma, San Diego, California, USA
| | | | | | | |
Collapse
|
7
|
Krajewska J, Chyży P, Durka K, Wińska P, Krzyśko KA, Luliński S, Laudy AE. Aromatic Diboronic Acids as Effective KPC/AmpC Inhibitors. Molecules 2023; 28:7362. [PMID: 37959781 PMCID: PMC10648349 DOI: 10.3390/molecules28217362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023] Open
Abstract
Over 30 compounds, including para-, meta-, and ortho-phenylenediboronic acids, ortho-substituted phenylboronic acids, benzenetriboronic acids, di- and triboronated thiophenes, and pyridine derivatives were investigated as potential β-lactamase inhibitors. The highest activity against KPC-type carbapenemases was found for ortho-phenylenediboronic acid 3a, which at the concentration of 8/4 mg/L reduced carbapenems' MICs up to 16/8-fold, respectively. Checkerboard assays revealed strong synergy between carbapenems and 3a with the fractional inhibitory concentrations indices of 0.1-0.32. The nitrocefin hydrolysis test and the whole cell assay with E. coli DH5α transformant carrying blaKPC-3 proved KPC enzyme being its molecular target. para-Phenylenediboronic acids efficiently potentiated carbapenems against KPC-producers and ceftazidime against AmpC-producers, whereas meta-phenylenediboronic acids enhanced only ceftazidime activity against the latter ones. Finally, the statistical analysis confirmed that ortho-phenylenediboronic acids act synergistically with carbapenems significantly stronger than other groups. Since the obtained phenylenediboronic compounds are not toxic to MRC-5 human fibroblasts at the tested concentrations, they can be considered promising scaffolds for the future development of novel KPC/AmpC inhibitors. The complexation of KPC-2 with the most representative isomeric phenylenediboronic acids 1a, 2a, and 3a was modeled by quantum mechanics/molecular mechanics calculations. Compound 3a reached the most effective configuration enabling covalent binding to the catalytic Ser70 residue.
Collapse
Affiliation(s)
- Joanna Krajewska
- Department of Pharmaceutical Microbiology and Bioanalysis, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Piotr Chyży
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland;
| | - Krzysztof Durka
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.D.); (P.W.); (S.L.)
| | - Patrycja Wińska
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.D.); (P.W.); (S.L.)
| | | | - Sergiusz Luliński
- Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland; (K.D.); (P.W.); (S.L.)
| | - Agnieszka E. Laudy
- Department of Pharmaceutical Microbiology and Bioanalysis, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
8
|
Nichols WW, Lahiri SD, Bradford PA, Stone GG. The primary pharmacology of ceftazidime/avibactam: resistance in vitro. J Antimicrob Chemother 2023; 78:569-585. [PMID: 36702744 DOI: 10.1093/jac/dkac449] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
This article reviews resistance to ceftazidime/avibactam as an aspect of its primary pharmacology, linked thematically with recent reviews of the basic in vitro and in vivo translational biology of the combination (J Antimicrob Chemother 2022; 77: 2321-40 and 2341-52). In Enterobacterales or Pseudomonas aeruginosa, single-step exposures to 8× MIC of ceftazidime/avibactam yielded frequencies of resistance from <∼0.5 × 10-9 to 2-8 × 10-9, depending on the host strain and the β-lactamase harboured. β-Lactamase structural gene mutations mostly affected the avibactam binding site through changes in the Ω-loop: e.g. Asp179Tyr (D179Y) in KPC-2. Other mutations included ones proposed to reduce the permeability to ceftazidime and/or avibactam through changes in outer membrane structure, up-regulated efflux, or both. The existence, or otherwise, of cross-resistance between ceftazidime/avibactam and other antibacterial agents was also reviewed as a key element of the preclinical primary pharmacology of the new agent. Cross-resistance between ceftazidime/avibactam and other β-lactam-based antibacterial agents was caused by MBLs. Mechanism-based cross-resistance was not observed between ceftazidime/avibactam and fluoroquinolones, aminoglycosides or colistin. A low level of general co-resistance to ceftazidime/avibactam was observed in MDR Enterobacterales and P. aeruginosa. For example, among 2821 MDR Klebsiella spp., 3.4% were resistant to ceftazidime/avibactam, in contrast to 0.07% of 8177 non-MDR isolates. Much of this was caused by possession of MBLs. Among 1151 MDR, XDR and pandrug-resistant isolates of P. aeruginosa from the USA, 11.1% were resistant to ceftazidime/avibactam, in contrast to 3.0% of 7452 unselected isolates. In this case, the decreased proportion susceptible was not due to MBLs.
Collapse
Affiliation(s)
| | - Sushmita D Lahiri
- Infectious Diseases and Vaccines, Johnson & Johnson, Cambridge, MA, USA
| | | | | |
Collapse
|
9
|
Vásquez-Ponce F, Dantas K, Becerra J, Melocco G, Esposito F, Cardoso B, Rodrigues L, Lima K, de Lima AV, Sellera FP, Mattos R, Trevisoli L, Vianello MA, Sincero T, Di Conza J, Vespero E, Gutkind G, Sampaio J, Lincopan N. Detecting KPC-2 and NDM-1 Coexpression in Klebsiella pneumoniae Complex from Human and Animal Hosts in South America. Microbiol Spectr 2022; 10:e0115922. [PMID: 35980188 PMCID: PMC9604071 DOI: 10.1128/spectrum.01159-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 07/12/2022] [Indexed: 12/30/2022] Open
Abstract
Reports of Gram-negative bacteria harboring multiple carbapenemase genes have increased in South America, leading to an urgent need for appropriate microbiological diagnosis. We evaluated phenotypic methods for detecting Klebsiella pneumoniae carbapenemase 2 (KPC-2) and New Delhi metallo-β-lactamase-1 (NDM-1) coexpression in members of the K. pneumoniae complex (i.e., K. pneumoniae, K. quasipneumoniae, and K. variicola) isolated from human and animal hosts, based on inhibition of ceftazidime-avibactam (CZA) and aztreonam (ATM) by dipicolinic acid (DPA), EDTA, or avibactam (AVI). While the presence of blaKPC-2 and blaNDM-1 genes was confirmed by whole-genome sequencing, PCR, and/or GeneXpert, coexpression was successfully detected based on the following: (i) a ≥5-mm increase in the zone diameter of ATM (30 µg) disks plus AVI (4 or 20 µg) and ≥4-mm and ≥10-mm increases in the zone diameters for "CZA 50" (30 µg ceftazidime [CAZ] and 20 µg AVI) and "CZA 14" (10 µg CAZ and 4 µg AVI) disks, respectively, when we added DPA (1 mg/disk) or EDTA (5 mM) in a combined disk test (CDT); (ii) a positive ghost zone (synergism) between ATM (30 µg) and CZA 50 disks and between CZA 50 and DPA (1 mg) disks, using the double-disk synergy test (DDST) at a disk-disk distance of 2.5 cm; (iii) ≥3-fold MIC reductions of ATM and CZA in the presence of AVI (4 µg/mL), DPA (500 µg/mL), or EDTA (320 µg/mL); and (iv) immunochromatography. Although our results demonstrated that inhibition by AVI, DPA, and EDTA may provide simple and inexpensive methods for the presumptive detection of coexpression of KPC-2 and NDM-1 in members of the K. pneumoniae complex, additional studies are necessary to confirm the accuracy of these methodologies by testing other Gram-negative bacterial species and other KPC and NDM variants coexpressed by WHO critical priority pathogens detected worldwide. IMPORTANCE Alerts regarding the emergence and increase of combinations of carbapenemases in Enterobacterales in Latin America and the Caribbean have recently been issued by PAHO and WHO, emphasizing the importance of appropriate microbiological diagnosis and the effective and articulated implementation of infection prevention and control programs. In this study, we evaluated methods based on inhibition of ceftazidime (CAZ), ceftazidime-avibactam (CZA), and aztreonam (ATM) by dipicolinic acid (DPA), EDTA, and avibactam (AVI) inhibitors for the identification of KPC-2- and NDM-1-coexpression in members of the K. pneumoniae complex recovered from human and animal hosts. Our results demonstrate that inhibition by AVI, DPA, and EDTA may provide simple and inexpensive methods for the presumptive detection of coexpression of KPC-2 and NDM-1 in members of the K. pneumoniae complex.
Collapse
Affiliation(s)
- Felipe Vásquez-Ponce
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Karine Dantas
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Johana Becerra
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Gregory Melocco
- Department of Clinical Analysis, School of Pharmacy, Universidade de São Paulo, São Paulo, Brazil
| | - Fernanda Esposito
- Department of Clinical Analysis, School of Pharmacy, Universidade de São Paulo, São Paulo, Brazil
| | - Brenda Cardoso
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Larissa Rodrigues
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Keila Lima
- Department of Clinical Analysis, School of Pharmacy, Universidade de São Paulo, São Paulo, Brazil
| | - Aline V. de Lima
- Department of Clinical Analysis, School of Pharmacy, Universidade de São Paulo, São Paulo, Brazil
| | - Fábio P. Sellera
- Department of Internal Medicine, School of Veterinary Medicine and Animal Science, Universidade de São Paulo, São Paulo, Brazil
- School of Veterinary Medicine, Metropolitan University of Santos, Santos, Brazil
| | | | | | | | - Thais Sincero
- Department of Clinical Analysis, Health Sciences Center, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Jose Di Conza
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriologia y Virología Molecular, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Eliana Vespero
- Department of Pathology, Clinical and Toxicological Analysis, Health Sciences Center, University Hospital of Londrina, Paraná, Brazil
| | - Gabriel Gutkind
- Facultad de Farmacia y Bioquímica, Instituto de Investigaciones en Bacteriologia y Virología Molecular, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Jorge Sampaio
- Department of Clinical Analysis, School of Pharmacy, Universidade de São Paulo, São Paulo, Brazil
- Fleury Medicine and Health, Microbiology Section, São Paulo, Brazil
| | - Nilton Lincopan
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
- Department of Clinical Analysis, School of Pharmacy, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
List KK, Kolpen M, Kragh KN, Charbon G, Radmer S, Hansen F, Løbner-Olesen A, Frimodt-Møller N, Hertz FB. Synergy between Mecillinam and Ceftazidime/Avibactam or Avibactam against Multi-Drug-Resistant Carbapenemase-Producing Escherichia coli and Klebsiella pneumoniae. Antibiotics (Basel) 2022; 11:antibiotics11101280. [PMID: 36289937 PMCID: PMC9599007 DOI: 10.3390/antibiotics11101280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Carbapenemase-producing Klebsiella pneumoniae and Escherichia coli have become a significant global health challenge. This has created an urgent need for new treatment modalities. We evaluated the efficacy of mecillinam in combination with either avibactam or ceftazidime/avibactam against carbapenemase-producing clinical isolates. Materials and methods: Nineteen MDR clinical isolates of K. pneumoniae and E. coli were selected for the presence of blaKPC, blaNDM, blaOXA or blaIMP based on whole-genome sequencing and phenotypic susceptibility testing. We tested the synergy between mecillinam and avibactam or ceftazidime/avibactam. We used time−kill studies in vitro and a mouse peritonitis/sepsis model to confirm the synergistic effect. We investigated avibactam’s impact on mecillinam´s affinity for penicillin-binding proteins with a Bocillin assay, and cell changes with phase-contrast and confocal laser scanning microscopy. Results: Mecillinam combined with ceftazidime/avibactam or avibactam substantially reduced MICs (from up to >256 µg/mL to <0.0016 µg/mL) for 17/18 strains. Significant log-CFU reductions were confirmed in time−kill and in vivo experiments. The Bocillin assay did not reveal changes. Conclusion: Mecillinam in combination with avibactam or ceftazidime/avibactam has a notable effect on most types of CPEs, both in vitro and in vivo. The mecillinam/avibactam combination treatment could be a new efficient antibiotic treatment against multi-drug-resistant carbapenemase-producing Gram-negative pathogens.
Collapse
Affiliation(s)
| | - Mette Kolpen
- Department of Clinical Microbiology, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Kasper Nørskov Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Godefroid Charbon
- Department of Clinical Microbiology, Rigshospitalet, DK-2100 Copenhagen, Denmark
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen, Denmark
- Correspondence: (G.C.); (F.B.H.)
| | - Stine Radmer
- Department of Clinical Microbiology, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Frank Hansen
- Statens Serum Institut, DK-2300 Copenhagen, Denmark
| | | | - Niels Frimodt-Møller
- Department of Clinical Microbiology, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Frederik Boetius Hertz
- Department of Clinical Microbiology, Rigshospitalet, DK-2100 Copenhagen, Denmark
- Correspondence: (G.C.); (F.B.H.)
| |
Collapse
|
11
|
Intrinsic Antibacterial Activity of Xeruborbactam
In Vitro
: Assessing Spectrum and Mode of Action. Antimicrob Agents Chemother 2022; 66:e0087922. [PMID: 36102663 PMCID: PMC9578396 DOI: 10.1128/aac.00879-22] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Xeruborbactam (formerly QPX7728) is a cyclic boronate inhibitor of numerous serine and metallo-beta-lactamases. At concentrations generally higher than those required for beta-lactamase inhibition, xeruborbactam has direct antibacterial activity against some Gram-negative bacteria, with MIC50/MIC90 values of 16/32 μg/mL and 16/64 μg/mL against carbapenem-resistant Enterobacterales and carbapenem-resistant Acinetobacter baumannii, respectively (the MIC50/MIC90 values against Pseudomonas aeruginosa are >64 μg/mL). In Klebsiella pneumoniae, inactivation of OmpK36 alone or in combination with OmpK35 resulted in 2- to 4-fold increases in the xeruborbactam MIC. In A. baumannii and P. aeruginosa, AdeIJK and MexAB-OprM, respectively, affected xeruborbactam’s antibacterial potency (the MICs were 4- to 16-fold higher in efflux-proficient strains). In Escherichia coli and K. pneumoniae, the 50% inhibitory concentrations (IC50s) of xeruborbactam’s binding to penicillin-binding proteins (PBPs) PBP1a/PBP1b, PBP2, and PBP3 were in the 40 to 70 μM range; in A. baumannii, xeruborbactam bound to PBP1a, PBP2, and PBP3 with IC50s of 1.4 μM, 23 μM, and 140 μM, respectively. Treating K. pneumoniae and P. aeruginosa with xeruborbactam at 1× and 2× MIC resulted in changes of cellular morphology similar to those observed with meropenem; the morphological changes observed after treatment of A. baumannii were consistent with inhibition of multiple PBPs but were unique to xeruborbactam compared to the results for control beta-lactams. No single-step xeruborbactam resistance mutants were obtained after selection at 4× MIC of xeruborbactam using wild-type strains of E. coli, K. pneumoniae, and A. baumannii; mutations selected at 2× MIC in K. pneumoniae did not affect antibiotic potentiation by xeruborbactam through beta-lactamase inhibition. Consistent with inhibition of PBPs, xeruborbactam enhanced the potencies of beta-lactam antibiotics even against strains that lacked beta-lactamase. In a large panel of KPC-producing clinical isolates, the MIC90 values of meropenem tested with xeruborbactam (8 μg/mL) were at least 4-fold lower than those in combination with vaborbactam at 64 μg/mL, the concentration of vaborbactam that is associated with complete inhibition of KPC. The additional enhancement of the potency of beta-lactam antibiotics beyond beta-lactamase inhibition may contribute to the potentiation of beta-lactam antibiotics by xeruborbactam.
Collapse
|
12
|
Shirley JD, Nauta KM, Carlson EE. Live-Cell Profiling of Penicillin-Binding Protein Inhibitors in Escherichia coli MG1655. ACS Infect Dis 2022; 8:1241-1252. [PMID: 35763562 PMCID: PMC10040144 DOI: 10.1021/acsinfecdis.2c00004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Penicillin-binding proteins (PBPs) make up an essential class of bacterial enzymes that carry out the final steps of peptidoglycan synthesis and regulate the recycling of this polymeric structure. PBPs are an excellent drug target and have been the most clinically relevant antibacterial target since the 1940s with the introduction of β-lactams. Despite this, a large gap in knowledge remains regarding the individual function and regulation of each PBP homologue in most bacteria. This can be attributed to a lack of chemical tools and methods that enable the study of individual PBPs in an activity-dependent manner and in their native environment. The development of such methods in Gram-negative bacteria has been particularly challenging due to the presence of an outer membrane and numerous resistance mechanisms. To address this, we have developed an optimized live-cell assay for screening inhibitors of the PBPs in Escherichia coli MG1655. We utilized EDTA to permeabilize Gram-negative cells, enabling increased penetration of our readout probe, Bocillin-FL, and subsequent analysis of PBP-inhibition profiles. To identify scaffolds for future development of PBP-selective activity-based probes, we screened ten β-lactams, one diazabicyclooctane, and one monobactam for their PBP-selectivity profiles in E. coli MG1655. These results demonstrate the utility of our assay for the screening of inhibitors in live, non-hypersusceptible Gram-negative organisms.
Collapse
Affiliation(s)
- Joshua D Shirley
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States
| | - Kelsie M Nauta
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Erin E Carlson
- Department of Medicinal Chemistry, University of Minnesota, 208 Harvard Street SE, Minneapolis, Minnesota 55454, United States.,Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55454, United States.,Department of Pharmacology, University of Minnesota, 321 Church Street SE, Minneapolis, Minnesota 55454, United States
| |
Collapse
|
13
|
Nichols WW, Bradford PA, Lahiri SD, Stone GG. The primary pharmacology of ceftazidime/avibactam: in vitro translational biology. J Antimicrob Chemother 2022; 77:2321-2340. [PMID: 35665807 DOI: 10.1093/jac/dkac171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Previous reviews of ceftazidime/avibactam have focused on in vitro molecular enzymology and microbiology or the clinically associated properties of the combination. Here we take a different approach. We initiate a series of linked reviews that analyse research on the combination that built the primary pharmacology data required to support the clinical and business risk decisions to perform randomized controlled Phase 3 clinical trials, and the additional microbiological research that was added to the above, and the safety and chemical manufacturing and controls data, that constituted successful regulatory licensing applications for ceftazidime/avibactam in multiple countries, including the USA and the EU. The aim of the series is to provide both a source of reference for clinicians and microbiologists to be able to use ceftazidime/avibactam to its best advantage for patients, but also a case study of bringing a novel β-lactamase inhibitor (in combination with an established β-lactam) through the microbiological aspects of clinical development and regulatory applications, updated finally with a review of resistance occurring in patients under treatment. This first article reviews the biochemistry, structural biology and basic microbiology of the combination, showing that avibactam inhibits the great majority of serine-dependent β-lactamases in Enterobacterales and Pseudomonas aeruginosa to restore the in vitro antibacterial activity of ceftazidime. Translation to efficacy against infections in vivo is reviewed in the second co-published article, Nichols et al. (J Antimicrob Chemother 2022; dkac172).
Collapse
|
14
|
Gaibani P, Giani T, Bovo F, Lombardo D, Amadesi S, Lazzarotto T, Coppi M, Rossolini GM, Ambretti S. Resistance to Ceftazidime/Avibactam, Meropenem/Vaborbactam and Imipenem/Relebactam in Gram-Negative MDR Bacilli: Molecular Mechanisms and Susceptibility Testing. Antibiotics (Basel) 2022; 11:antibiotics11050628. [PMID: 35625273 PMCID: PMC9137602 DOI: 10.3390/antibiotics11050628] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/01/2022] [Accepted: 05/03/2022] [Indexed: 01/25/2023] Open
Abstract
Multidrug resistance (MDR) represents a serious global threat due to the rapid global spread and limited antimicrobial options for treatment of difficult-to-treat (DTR) infections sustained by MDR pathogens. Recently, novel β-lactams/β-lactamase inhibitor combinations (βL-βLICs) have been developed for the treatment of DTR infections due to MDR Gram-negative pathogens. Although novel βL-βLICs exhibited promising in vitro and in vivo activities against MDR pathogens, emerging resistances to these novel molecules have recently been reported. Resistance to novel βL-βLICs is due to several mechanisms including porin deficiencies, increasing carbapenemase expression and/or enzyme mutations. In this review, we summarized the main mechanisms related to the resistance to ceftazidime/avibactam, meropenem/vaborbactam and imipenem/relebactam in MDR Gram-negative micro-organisms. We focused on antimicrobial activities and resistance traits with particular regard to molecular mechanisms related to resistance to novel βL-βLICs. Lastly, we described and discussed the main detection methods for antimicrobial susceptibility testing of such molecules. With increasing reports of resistance to novel βL-βLICs, continuous attention should be maintained on the monitoring of the phenotypic traits of MDR pathogens, into the characterization of related mechanisms, and on the emergence of cross-resistance to these novel antimicrobials.
Collapse
Affiliation(s)
- Paolo Gaibani
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
- Correspondence:
| | - Tommaso Giani
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50134 Florence, Italy; (T.G.); (M.C.); (G.M.R.)
- Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy
| | - Federica Bovo
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| | - Donatella Lombardo
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| | - Stefano Amadesi
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| | - Tiziana Lazzarotto
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40100 Bologna, Italy
| | - Marco Coppi
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50134 Florence, Italy; (T.G.); (M.C.); (G.M.R.)
- Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy
| | - Gian Maria Rossolini
- Clinical Microbiology and Virology Unit, Careggi University Hospital, 50134 Florence, Italy; (T.G.); (M.C.); (G.M.R.)
- Department of Experimental and Clinical Medicine, University of Florence, 50100 Florence, Italy
| | - Simone Ambretti
- Division of Microbiology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.B.); (D.L.); (S.A.); (T.L.); (S.A.)
| |
Collapse
|
15
|
Alsenani TA, Viviani SL, Kumar V, Taracila MA, Bethel CR, Barnes MD, Papp-Wallace KM, Shields RK, Nguyen MH, Clancy CJ, Bonomo RA, van den Akker F. Structural Characterization of the D179N and D179Y Variants of KPC-2 β-Lactamase: Ω-Loop Destabilization as a Mechanism of Resistance to Ceftazidime-Avibactam. Antimicrob Agents Chemother 2022; 66:e0241421. [PMID: 35341315 PMCID: PMC9017313 DOI: 10.1128/aac.02414-21] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/23/2022] [Indexed: 11/20/2022] Open
Abstract
Klebsiella pneumoniae carbapenemases (KPC-2 and KPC-3) present a global clinical threat, as these β-lactamases confer resistance to carbapenems and oxyimino-cephalosporins. Recent clinically identified KPC variants with substitutions at Ambler position D179, located in the Ω loop, are resistant to the β-lactam/β-lactamase inhibitor combination ceftazidime-avibactam, but susceptible to meropenem-vaborbactam. To gain insights into ceftazidime-avibactam resistance conferred by D179N/Y variants of KPC-2, crystal structures of these variants were determined. The D179N KPC-2 structure revealed that the change of the carboxyl to an amide moiety at position 179 disrupted the salt bridge with R164 present in wild-type KPC-2. Additional interactions were disrupted in the Ω loop, causing a decrease in the melting temperature. Shifts originating from N179 were also transmitted toward the active site, including ∼1-Å shifts of the deacylation water and interacting residue N170. The structure of the D179Y KPC-2 β-lactamase revealed more drastic changes, as this variant exhibited disorder of the Ω loop, with other flanking regions also being disordered. We postulate that the KPC-2 variants can accommodate ceftazidime because the Ω loop is displaced in D179Y or can be more readily displaced in D179N KPC-2. To understand why the β-lactamase inhibitor vaborbactam is less affected by the D179 variants than avibactam, we determined the crystal structure of D179N KPC-2 in complex with vaborbactam, which revealed wild-type KPC-2-like vaborbactam-active site interactions. Overall, the structural results regarding KPC-2 D179 variants revealed various degrees of destabilization of the Ω loop that contribute to ceftazidime-avibactam resistance, possible substrate-assisted catalysis of ceftazidime, and meropenem and meropenem-vaborbactam susceptibility.
Collapse
Affiliation(s)
- T. A. Alsenani
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - S. L. Viviani
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - V. Kumar
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - M. A. Taracila
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - C. R. Bethel
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - M. D. Barnes
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - K. M. Papp-Wallace
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - R. K. Shields
- University of Pittsburgh, Department of Medicine, Division of Infectious Diseases, Pittsburgh, Pennsylvania, USA
| | - M. H. Nguyen
- University of Pittsburgh, Department of Medicine, Division of Infectious Diseases, Pittsburgh, Pennsylvania, USA
| | - C. J. Clancy
- University of Pittsburgh, Department of Medicine, Division of Infectious Diseases, Pittsburgh, Pennsylvania, USA
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA
| | - R. A. Bonomo
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Senior Clinical Scientist Investigator, CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, USA
| | - F. van den Akker
- Department of Biochemistry, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Perera WPTD, Dissanayake DMRK, Unagolla JM, De Silva RT, Bathige SDNK, Pahalagedara LR. Albumin grafted coaxial electrosparyed polycaprolactone-zinc oxide nanoparticle for sustained release and activity enhanced antibacterial drug delivery. RSC Adv 2022; 12:1718-1727. [PMID: 35425191 PMCID: PMC8978970 DOI: 10.1039/d1ra07847j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/03/2022] [Indexed: 12/01/2022] Open
Abstract
One of the most serious issues faced by the healthcare sector is the development of multidrug resistance among various pathogens. It is such that developing new and more capable drugs takes far too long to counter such resistance. In order to overcome these concerns, this study focused on improving upon the coaxial electrospraying process by producing cloxacillin loaded albumin polycaprolactone (PCL) with a ZnO coating for sustained and activity enhanced drug delivery. Albumin-grafted, polycaprolactone-coated, zinc oxide-loaded cloxacillin (APCL-CLOX-ZnO) nanoparticles with a diameter of 85-110 nm were obtained via a coaxial electrospray technique. The encapsulation efficiency of cloxacillin of ZnO-CLOX was found to be approximately 60%. The loading efficiencies of ZnO-CLOX and APCL-CLOX-ZnO were found to be 40% and 28% respectively. Albumin was employed in order to impart immune evasion properties to the formulation. Drug-loaded ZnO NPs were analyzed using SEM, TEM, FT-IR and TGA. This novel formulation was shown to possess sustained release characteristics owing to the PCL and albumin coatings, relative to uncoated counterparts. ZnO-CLOX and APCL-CLOX-ZnO exhibited 72% and 52% cloxacillin release within 24 h. APCL-CLOX-ZnO exhibited potent antimicrobial activity against S. epidermidis, B. cereus and P. aeruginosa and some activity against E. coli with inhibition zones 32 ± 1.4, 34 ± 0.3, 32 ± 0.6 and 11 ± 0.4 mm, respectively. Cytotoxicity studies against murine preosteoblast cells revealed that the albumin-PCL coating served to drastically reduce initial toxicity against healthy mammalian cells. In vitro lung deposition study showed 70% of APCL-CLOX-ZnO particles can reach up to the alveoli level. Therefore, this novel coaxial nanoformulation may serve as a promising drug delivery platform for the treatment of bacterial infections including respiratory tract complications.
Collapse
Affiliation(s)
- W Pamoda Thavish D Perera
- Academy of the Sri Lanka Institute of Nanotechnology Nanotechnology and Science Park, Mahenwatte, Pitipana Homagama 10206 Sri Lanka
- Sri Lanka Institute of Nanotechnology Nanotechnology and Science Park, Mahenwatte, Pitipana Homagama 10206 Sri Lanka
| | - D M Ranga K Dissanayake
- Sri Lanka Institute of Nanotechnology Nanotechnology and Science Park, Mahenwatte, Pitipana Homagama 10206 Sri Lanka
- Department of Pharmacy and Pharmaceutical Sciences, University of Sri Jayewardenepura Gangodawila Nugegoda 10250 Sri Lanka
| | - Janitha M Unagolla
- Department of Bioengineering, College of Engineering, University of Toledo Toledo OH 43607 USA
| | - Rangika T De Silva
- Sri Lanka Institute of Nanotechnology Nanotechnology and Science Park, Mahenwatte, Pitipana Homagama 10206 Sri Lanka
| | - Sanjaya D N K Bathige
- Sri Lanka Institute of Nanotechnology Nanotechnology and Science Park, Mahenwatte, Pitipana Homagama 10206 Sri Lanka
| | - Lakshitha R Pahalagedara
- Sri Lanka Institute of Nanotechnology Nanotechnology and Science Park, Mahenwatte, Pitipana Homagama 10206 Sri Lanka
| |
Collapse
|
17
|
Kumar V, Viviani SL, Ismail J, Agarwal S, Bonomo RA, van den Akker F. Structural analysis of the boronic acid β-lactamase inhibitor vaborbactam binding to Pseudomonas aeruginosa penicillin-binding protein 3. PLoS One 2021; 16:e0258359. [PMID: 34653211 PMCID: PMC8519428 DOI: 10.1371/journal.pone.0258359] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 09/24/2021] [Indexed: 11/18/2022] Open
Abstract
Antimicrobial resistance (AMR) mediated by β-lactamases is the major and leading cause of resistance to penicillins and cephalosporins among Gram-negative bacteria. β-Lactamases, periplasmic enzymes that are widely distributed in the bacterial world, protect penicillin-binding proteins (PBPs), the major cell wall synthesizing enzymes, from inactivation by β-lactam antibiotics. Developing novel PBP inhibitors with a non-β-lactam scaffold could potentially evade this resistance mechanism. Based on the structural similarities between the evolutionary related serine β-lactamases and PBPs, we investigated whether the potent β-lactamase inhibitor, vaborbactam, could also form an acyl-enzyme complex with Pseudomonas aeruginosa PBP3. We found that this cyclic boronate, vaborbactam, inhibited PBP3 (IC50 of 262 μM), and its binding to PBP3 increased the protein thermal stability by about 2°C. Crystallographic analysis of the PBP3:vaborbactam complex reveals that vaborbactam forms a covalent bond with the catalytic S294. The amide moiety of vaborbactam hydrogen bonds with N351 and the backbone oxygen of T487. The carboxyl group of vaborbactam hydrogen bonds with T487, S485, and S349. The thiophene ring and cyclic boronate ring of vaborbactam form hydrophobic interactions, including with V333 and Y503. The active site of the vaborbactam-bound PBP3 harbors the often observed ligand-induced formation of the aromatic wall and hydrophobic bridge, yet the residues involved in this wall and bridge display much higher temperature factors compared to PBP3 structures bound to high-affinity β-lactams. These insights could form the basis for developing more potent novel cyclic boronate-based PBP inhibitors to inhibit these targets and overcome β-lactamases-mediated resistance mechanisms.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Samantha L. Viviani
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Jeeda Ismail
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Shreya Agarwal
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Robert A. Bonomo
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
- Louis Stokes Cleveland Veteran’s Affairs Medical Center Research Service, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Molecular Biology and Microbiology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Pharmacology, Case Western Reserve University, Cleveland, Ohio, United States of America
- Department of Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio, United States of America
- VA Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Focco van den Akker
- Department of Biochemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
18
|
Guo Y, Liu N, Lin Z, Ba X, Zhuo C, Li F, Wang J, Li Y, Yao L, Liu B, Xiao S, Jiang Y, Zhuo C. Mutations in porin LamB contribute to ceftazidime-avibactam resistance in KPC-producing Klebsiella pneumoniae. Emerg Microbes Infect 2021; 10:2042-2051. [PMID: 34551677 PMCID: PMC8567916 DOI: 10.1080/22221751.2021.1984182] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Ceftazidime-avibactam (CAZ-AVI) shows promising activity against carbapenem-resistant Klebsiella pneumoniae (CRKP), however, CAZ-AVI resistance have emerged recently. Mutations in KPCs, porins OmpK35 and/or OmpK36, and PBPs are known to contribute to the resistance to CAZ-AVI in CRKP. To identify novel CAZ-AVI resistance mechanism, we generated 10 CAZ-AVI-resistant strains from 14 CAZ-AVI susceptible KPC-producing K. pneumoniae (KPC-Kp) strains through in vitro multipassage resistance selection using low concentrations of CAZ-AVI. Comparative genomic analysis for the original and derived mutants identified CAZ-AVI resistance-associated mutations in KPCs, PBP3 (encoded by ftsI), and LamB, an outer membrane maltoporin. CAZ-AVI susceptible KPC-Kp strains became resistant when complemented with mutated blaKPC genes. Complementation experiments also showed that a plasmid borne copy of wild-type lamB or ftsI gene reduced the MIC value of CAZ-AVI in the induced resistant strains. In addition, blaKPC expression level increased in four of the six CAZ-AVI-resistant strains without KPC mutations, indicating a probable association between increased blaKPC expression and increased resistance in these strains. In conclusion, we here identified a novel mechanism of CAZ-AVI resistance associated with mutations in porin LamB in KPC-Kp.
Collapse
Affiliation(s)
- Yingyi Guo
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ningjing Liu
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhiwei Lin
- Laboratory of Respiratory Disease, People's Hospital of Yangjiang, Guangdong, People's Republic of China
| | - Xiaoliang Ba
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Chuyue Zhuo
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Feifeng Li
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jiong Wang
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yitan Li
- Laboratory of Respiratory Disease, People's Hospital of Yangjiang, Guangdong, People's Republic of China
| | - Likang Yao
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Baomo Liu
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Shunian Xiao
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ying Jiang
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Chao Zhuo
- Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
19
|
Time-Kill Evaluation of Antibiotic Combinations Containing Ceftazidime-Avibactam against Extensively Drug-Resistant Pseudomonas aeruginosa and Their Potential Role against Ceftazidime-Avibactam-Resistant Isolates. Microbiol Spectr 2021; 9:e0058521. [PMID: 34319141 PMCID: PMC8552783 DOI: 10.1128/spectrum.00585-21] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Ceftazidime-avibactam (CZA) has emerged as a promising solution to the lack of new antibiotics against Pseudomonas aeruginosa infections. Data from in vitro assays of CZA combinations, however, are scarce. The objective of our study was to perform a time-kill analysis of the effectiveness of CZA alone and in combination with other antibiotics against a collection of extensively drug-resistant (XDR) P. aeruginosa isolates. Twenty-one previously characterized representative XDR P. aeruginosa isolates were selected. Antibiotic susceptibility was tested by broth microdilution, and results were interpreted using CLSI criteria. The time-kill experiments were performed in duplicate for each isolate. Antibiotics were tested at clinically achievable free-drug concentrations. Different treatment options, including CZA alone and combined with amikacin, aztreonam, meropenem, and colistin, were evaluated to identify the most effective combinations. Seven isolates were resistant to CZA (MIC ≥ 16/4 mg/liter), including four metallo-β-lactamase (MBL)-carrying isolates and two class A carbapenemases. Five of them were resistant or intermediate to aztreonam (MIC ≥ 16 mg/liter). Three isolates were resistant to amikacin (MIC ≥ 64 mg/liter) and one to colistin (MIC ≥ 4 mg/liter). CZA monotherapy had a bactericidal effect in 100% (14/14) of the CZA-susceptible isolates. Combination therapies achieved a greater overall reduction in bacterial load than monotherapy for the CZA-resistant isolates. CZA plus colistin was additive or synergistic in 100% (7/7) of the CZA-resistant isolates, while CZA plus amikacin and CZA plus aztreonam were additive or synergistic in 85%. CZA combined with colistin, amikacin, or aztreonam was more effective than monotherapy against XDR P. aeruginosa isolates. A CZA combination could be useful for treating XDR P. aeruginosa infections, including those caused by CZA-resistant isolates. IMPORTANCE The emergence of resistance to antibiotics is a serious public health problem worldwide and can be a cause of mortality. For this reason, antibiotic treatment is compromised, and we have few therapeutic options to treat infections. The main goal of our study is to search for new treatment options for infections caused by difficult-to-treat resistant germs. Pseudomonas aeruginosa is a Gram-negative bacterium distributed throughout the world with the ability to become resistant to most available antibiotics. Ceftazidime-avibactam (CZA) emerged as a promising solution to the lack of new antibiotics against infections caused by P. aeruginosa strains. This study intended to analyze the effect of CZA alone or in combination with other available antibiotics against P. aeruginosa strains. The combination of CZA with other antibiotics could be more effective than monotherapy against extensively drug-resistant P. aeruginosa strains.
Collapse
|
20
|
Bhagwat SS, Hariharan P, Joshi PR, Palwe SR, Shrivastava R, Patel MV, Devanga Ragupathi NK, Bakthavatchalam YD, Ramesh MS, Soman R, Veeraraghavan B. 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 2021; 75:3563-3567. [PMID: 32772098 DOI: 10.1093/jac/dkaa353] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Recent reports reveal the emergence of Escherichia coli isolates harbouring a novel resistance mechanism based on four-amino-acid inserts in PBP3. These organisms concomitantly expressed ESBLs or/and serine-/metallo-carbapenemases and were phenotypically detected by elevated aztreonam/avibactam MICs. OBJECTIVES The in vitro activities of the investigational antibiotic cefepime/zidebactam and approved antibiotics (ceftazidime/avibactam, ceftolozane/tazobactam, imipenem/relebactam and others) were determined against E. coli isolates harbouring four-amino-acid inserts in PBP3. METHODS Whole-genome sequenced E. coli isolates (n = 89) collected from a large tertiary care hospital in Southern India (n = 64) and from 12 tertiary care hospitals located across India (n = 25) during 2016-18, showing aztreonam/avibactam MICs ≥1 mg/L (≥4 times the aztreonam epidemiological cut-off) were included in this study. The MICs of antibiotics were determined using the reference broth microdilution method. RESULTS Four-amino-acid inserts [YRIK (n = 30) and YRIN (n = 53)] were found in 83/89 isolates. Among 83 isolates, 65 carried carbapenemase genes [blaNDM (n = 39), blaOXA-48-like (n = 11) and blaNDM + blaOXA-48-like (n = 15)] and 18 isolates produced ESBLs/class C β-lactamases only. At least 16 unique STs were noted. Cefepime/zidebactam demonstrated potent activity, with all isolates inhibited at ≤1 mg/L. Comparator antibiotics including ceftazidime/avibactam and imipenem/relebactam showed limited activities. CONCLUSIONS E. coli isolates concurrently harbouring four-amino-acid inserts in PBP3 and NDM are an emerging therapeutic challenge. Assisted by the PBP2-binding action of zidebactam, the cefepime/zidebactam combination overcomes both target modification (PBP3 insert)- and carbapenemase (NDM)-mediated resistance mechanisms in E. coli.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Mayur S Ramesh
- Department of Infectious Disease, Henry Ford Hospital, Detroit, MI, USA
| | - Rajeev Soman
- Department of Infectious Diseases, Jupiter Hospital, Pune, India
| | | |
Collapse
|
21
|
Nguyen DC, Dousa KM, Kurz SG, Brown ST, Drusano G, Holland SM, Kreiswirth BN, Boom WH, Daley CL, Bonomo RA. "One-two Punch": Synergistic β-lactam Combinations for Mycobacterium abscessus and Target Redundancy in the Inhibition of Peptidoglycan Synthesis Enzymes. Clin Infect Dis 2021; 73:1532-1536. [PMID: 34113990 DOI: 10.1093/cid/ciab535] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium abscessus subsp. abscessus is one of the most difficult pathogens to treat and its incidence in disease is increasing. Dual β-lactam combinations act synergistically in vitro, but are not widely employed in practice. A recent study shows that a combination of imipenem and ceftaroline significantly lowers the minimum inhibitory concentration (MIC) of clinical isolates despite both drugs targeting the same peptidoglycan synthesis enzymes. The underlying mechanism of this effect provides a basis for further investigations of dual β-lactam combinations in the treatment of M. abscessus subsp. abscessus eventually leading to a clinical trial. Furthermore, dual β-lactam strategies may be explored for other difficult mycobacterial infections.
Collapse
Affiliation(s)
- David C Nguyen
- Division of Infectious Diseases & HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.,Division of Pediatric Infectious Diseases, Rainbow Babies and Children's Hospital, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Khalid M Dousa
- Department of Internal Medicine and Infectious Diseases, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA
| | - Sebastian G Kurz
- Mount Sinai National Jewish Respiratory Institute, New York City, NY, USA
| | - Sheldon T Brown
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Medicine, James J. Peters VA Medical Center, Bronx, NY, USA
| | - George Drusano
- Institute for Therapeutic Innovation, University of Florida, Orlando, FL, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barry N Kreiswirth
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, USA
| | - W Henry Boom
- Division of Infectious Diseases & HIV Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.,Tuberculosis Research Unit, Case Western Reserve University, Cleveland, OH, USA
| | - Charles L Daley
- Division of Mycobacterial and Respiratory Infections, National Jewish Health, Denver, CO, USA
| | - Robert A Bonomo
- Department of Medicine, Pharmacology, Molecular Biology and Microbiology, Biochemistry, Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Medical Service, Research Service, and GRECC, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, OH, USA.,CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology, Cleveland, OH, USA
| |
Collapse
|
22
|
Perrotta F, Perrini MP. Successful Treatment of Klebsiella pneumoniae NDM Sepsis and Intestinal Decolonization with Ceftazidime/Avibactam Plus Aztreonam Combination in a Patient with TTP Complicated by SARSCoV-2 Nosocomial Infection. ACTA ACUST UNITED AC 2021; 57:medicina57050424. [PMID: 33924769 PMCID: PMC8145860 DOI: 10.3390/medicina57050424] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022]
Abstract
Carbapenem-resistant Enterobacteriaceae (CRE) are a serious public health threat. Infections due to these organisms are associated with significant morbidity and mortality. Among them, metallo-β-lactamases (MBLs)-producing Klebsiella pneumoniae are of global concern today. The ceftazidime/avibactam combination and the ceftazidime/avibactam + aztreonam combination currently represent the most promising antibiotic strategies to stave off these kinds of infections. We describe the case of a patient affected by thrombotic thrombocytopenic purpura (TTP) admitted in our ICU after developing a hospital-acquired SARS-CoV-2 interstitial pneumonia during his stay in the hematology department. His medical conditions during his ICU stay were further complicated by a K. Pneumoniae NDM sepsis. To our knowledge, the patient had no risk factors for multidrug-resistant bacteria exposure or contamination during his stay in the hematology department. During his stay in the ICU, we treated the sepsis with a combination therapy of ceftazidime/avibactam + aztreonam. The therapy solved his septic state, allowing for a progressive improvement in his general condition. Moreover, we noticed that the negativization of the hemocultures was also associated to a decontamination of his known rectal colonization. The ceftazidime/avibactam + aztreonam treatment could not only be a valid therapeutic option for these kinds of infections, but it could also be considered as a useful tool in selected patients’ intestinal decolonizations.
Collapse
Affiliation(s)
- Francesco Perrotta
- Department of Anesthesia and Intensive Care Unit, IRCCS Casa Sollievo della Sofferenza, Viale Cappuccini, 1, 71013 San Giovanni Rotondo, FG, Italy;
| | - Marco Paolo Perrini
- Department of Anesthesia and Intensive Care Unit, Università degli Studi di Foggia, Azienda Ospedaliero Universitaria Ospedali Riuniti di Foggia, 1, 71122 Viale Pinto, FG, Italy
- Correspondence:
| |
Collapse
|
23
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
24
|
Molecular Basis of AmpC β-Lactamase Induction by Avibactam in Pseudomonas aeruginosa: PBP Occupancy, Live Cell Binding Dynamics and Impact on Resistant Clinical Isolates Harboring PDC-X Variants. Int J Mol Sci 2021; 22:ijms22063051. [PMID: 33802668 PMCID: PMC8002452 DOI: 10.3390/ijms22063051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/10/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Avibactam belongs to the new class of diazabicyclooctane β-lactamase inhibitors. Its inhibitory spectrum includes class A, C and D enzymes, including P. aeruginosa AmpC. Nonetheless, recent reports have revealed strain-dependent avibactam AmpC induction. In the present work, we wanted to assess the mechanistic basis underlying AmpC induction and determine if derepressed PDC-X mutated enzymes from ceftazidime/avibactam-resistant clinical isolates were further inducible. We determined avibactam concentrations that half-maximally inhibited (IC50) bocillin FL binding. Inducer β-lactams were also studied as comparators. Live cells’ time-course penicillin-binding proteins (PBPs) occupancy of avibactam was studied. To assess the ampC induction capacity of avibactam and comparators, qRT-PCR was performed in wild-type PAO1, PBP4, triple PBP4, 5/6 and 7 knockout derivatives and two ceftazidime/avibactam-susceptible/resistant XDR clinical isolates belonging to the epidemic high-risk clone ST175. PBP4 inhibition was observed for avibactam and β-lactam comparators. Induction capacity was consistently correlated with PBP4 binding affinity. Outer membrane permeability-limited PBP4 binding was observed in the live cells’ assay. As expected, imipenem and cefoxitin showed strong induction in PAO1, especially for carbapenem; avibactam induction was conversely weaker. Overall, the inducer effect was less remarkable in ampC-derepressed mutants and nonetheless absent upon avibactam exposure in the clinical isolates harboring mutated AmpC variants and their parental strains.
Collapse
|
25
|
Structural Characterization of Diazabicyclooctane β-Lactam "Enhancers" in Complex with Penicillin-Binding Proteins PBP2 and PBP3 of Pseudomonas aeruginosa. mBio 2021; 12:mBio.03058-20. [PMID: 33593978 PMCID: PMC8545096 DOI: 10.1128/mbio.03058-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Multidrug-resistant (MDR) pathogens pose a significant public health threat. A major mechanism of resistance expressed by MDR pathogens is β-lactamase-mediated degradation of β-lactam antibiotics. The diazabicyclooctane (DBO) compounds zidebactam and WCK 5153, recognized as β-lactam “enhancers” due to inhibition of Pseudomonas aeruginosa penicillin-binding protein 2 (PBP2), are also class A and C β-lactamase inhibitors. To structurally probe their mode of PBP2 inhibition as well as investigate why P. aeruginosa PBP2 is less susceptible to inhibition by β-lactam antibiotics compared to the Escherichia coli PBP2, we determined the crystal structure of P. aeruginosa PBP2 in complex with WCK 5153. WCK 5153 forms an inhibitory covalent bond with the catalytic S327 of PBP2. The structure suggests a significant role for the diacylhydrazide moiety of WCK 5153 in interacting with the aspartate in the S-X-N/D PBP motif. Modeling of zidebactam in the active site of PBP2 reveals a similar binding mode. Both DBOs increase the melting temperature of PBP2, affirming their stabilizing interactions. To aid in the design of DBOs that can inhibit multiple PBPs, the ability of three DBOs to interact with P. aeruginosa PBP3 was explored crystallographically. Even though the DBOs show covalent binding to PBP3, they destabilized PBP3. Overall, the studies provide insights into zidebactam and WCK 5153 inhibition of PBP2 compared to their inhibition of PBP3 and the evolutionarily related KPC-2 β-lactamase. These molecular insights into the dual-target DBOs advance our knowledge regarding further DBO optimization efforts to develop novel potent β-lactamase-resistant, non-β-lactam PBP inhibitors.
Collapse
|
26
|
Structural Investigations of the Inhibition of Escherichia coli AmpC β-Lactamase by Diazabicyclooctanes. Antimicrob Agents Chemother 2021; 65:AAC.02073-20. [PMID: 33199391 PMCID: PMC7849013 DOI: 10.1128/aac.02073-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/12/2020] [Indexed: 12/24/2022] Open
Abstract
β-Lactam antibiotics are presently the most important treatments for infections by pathogenic Escherichia coli, but their use is increasingly compromised by β-lactamases, including the chromosomally encoded class C AmpC serine-β-lactamases (SBLs). The diazabicyclooctane (DBO) avibactam is a potent AmpC inhibitor; the clinical success of avibactam combined with ceftazidime has stimulated efforts to optimize the DBO core. We report kinetic and structural studies, including four high-resolution crystal structures, concerning inhibition of the AmpC serine-β-lactamase from E. coli (AmpC EC ) by clinically relevant DBO-based inhibitors: avibactam, relebactam, nacubactam, and zidebactam. Kinetic analyses and mass spectrometry-based assays were used to study their mechanisms of AmpC EC inhibition. The results reveal that, under our assay conditions, zidebactam manifests increased potency (apparent inhibition constant [K iapp], 0.69 μM) against AmpC EC compared to that of the other DBOs (K iapp = 5.0 to 7.4 μM) due to an ∼10-fold accelerated carbamoylation rate. However, zidebactam also has an accelerated off-rate, and with sufficient preincubation time, all the DBOs manifest similar potencies. Crystallographic analyses indicate a greater conformational freedom of the AmpC EC -zidebactam carbamoyl complex compared to those for the other DBOs. The results suggest the carbamoyl complex lifetime should be a consideration in development of DBO-based SBL inhibitors for the clinically important class C SBLs.
Collapse
|
27
|
de Sousa Coelho F, Mainardi JL. The multiple benefits of second-generation β-lactamase inhibitors in treatment of multidrug-resistant bacteria. Infect Dis Now 2021; 51:510-517. [PMID: 33870896 DOI: 10.1016/j.idnow.2020.11.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 12/26/2022]
Abstract
The World Health Organisation (WHO) has designated antibiotic resistance as one of the most challenging public health threats of the 21st century. Production of β-lactamase enzymes by Gram-negative bacteria is the main mechanism of resistance to β-lactam (BL), the most widely used antibiotic in clinics. In an attempt to neutralise the hydrolytic activity of these enzymes, β-lactamase inhibitors (BLIs) have been developed. First-generation BLIs include clavulanic acid, sulbactam and tazobactam. However, none of them cover all β-lactamase classes, and an increasingly wide panel of inhibitor-resistant bacterial strains has developed. Second-generation BLIs function via different mechanisms and were developed by novel scaffolds from which diazabicyclooctane (DBOs) and boronic acids have emerged. In this paper, we provide descriptions of promisor second-generation β-lactamase inhibitors, such as avibactam, vaborbactam and boronic acids, as well as several BL-BLI combinations that have been designed. While some combinations are now being used in clinical practice, most are presently limited to clinical trials or pre-clinical studies. In this paper, we emphasise the continuous need to develop novel and different BLIs to keep up with the multidrug-resistant bacteria that arise. At this time, however, second-generation BLIs constitute a promising and effective approach.
Collapse
Affiliation(s)
- F de Sousa Coelho
- Faculdade de Engenharia da Universidade do Porto, Instituto de Ciências Biomédicas Abel Salazar, Porto, Portugal.
| | - J-L Mainardi
- Centre de recherche des Cordeliers, INSERM UMRS 1138, Sorbonne université, université de Paris, Paris, France; Service de microbiologie, hôpital européen Georges-Pompidou, Assistance publique-Hôpitaux de Paris, centre-université de Paris, Paris, France.
| |
Collapse
|
28
|
New Carbapenemase Inhibitors: Clearing the Way for the β-Lactams. Int J Mol Sci 2020; 21:ijms21239308. [PMID: 33291334 PMCID: PMC7731173 DOI: 10.3390/ijms21239308] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 01/13/2023] Open
Abstract
Carbapenem resistance is a major global health problem that seriously compromises the treatment of infections caused by nosocomial pathogens. Resistance to carbapenems mainly occurs via the production of carbapenemases, such as VIM, IMP, NDM, KPC and OXA, among others. Preclinical and clinical trials are currently underway to test a new generation of promising inhibitors, together with the recently approved avibactam, relebactam and vaborbactam. This review summarizes the main, most promising carbapenemase inhibitors synthesized to date, as well as their spectrum of activity and current stage of development. We particularly focus on β-lactam/β-lactamase inhibitor combinations that could potentially be used to treat infections caused by carbapenemase-producer pathogens of critical priority. The emergence of these new combinations represents a step forward in the fight against antimicrobial resistance, especially in regard to metallo-β-lactamases and carbapenem-hydrolysing class D β-lactamases, not currently inhibited by any clinically approved inhibitor.
Collapse
|
29
|
Wang Y, Wang J, Wang R, Cai Y. Resistance to ceftazidime–avibactam and underlying mechanisms. J Glob Antimicrob Resist 2020; 22:18-27. [DOI: 10.1016/j.jgar.2019.12.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 01/08/2023] Open
|
30
|
Global Trends in Proteome Remodeling of the Outer Membrane Modulate Antimicrobial Permeability in Klebsiella pneumoniae. mBio 2020; 11:mBio.00603-20. [PMID: 32291303 PMCID: PMC7157821 DOI: 10.1128/mbio.00603-20] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Klebsiella pneumoniae is a pathogen of humans with high rates of mortality and a recognized global rise in incidence of carbapenem-resistant K. pneumoniae (CRKP). The outer membrane of K. pneumoniae forms a permeability barrier that modulates the ability of antibiotics to reach their intracellular target. OmpK35, OmpK36, OmpK37, OmpK38, PhoE, and OmpK26 are porins in the outer membrane of K. pneumoniae, demonstrated here to have a causative relationship to drug resistance phenotypes in a physiological context. The data highlight that currently trialed combination treatments with a carbapenem and β-lactamase inhibitors could be effective on porin-deficient K. pneumoniae. Together with structural data, the results reveal the role of outer membrane proteome remodeling in antimicrobial resistance of K. pneumoniae and point to the role of extracellular loops, not channel parameters, in drug permeation. This significant finding warrants care in the development of phage therapies for K. pneumoniae infections, given the way porin expression will be modulated to confer phage-resistant—and collateral drug-resistant—phenotypes in K. pneumoniae. In Gram-negative bacteria, the permeability of the outer membrane governs rates of antibiotic uptake and thus the efficacy of antimicrobial treatment. Hydrophilic drugs like β-lactam antibiotics depend on diffusion through pore-forming outer membrane proteins to reach their intracellular targets. In this study, we investigated the distribution of porin genes in more than 2,700 Klebsiella isolates and found a widespread loss of OmpK35 functionality, particularly in those strains isolated from clinical environments. Using a defined set of outer-membrane-remodeled mutants, the major porin OmpK35 was shown to be largely responsible for β-lactam permeation. Sequence similarity network analysis characterized the porin protein subfamilies and led to discovery of a new porin family member, OmpK38. Structure-based comparisons of OmpK35, OmpK36, OmpK37, OmpK38, and PhoE showed near-identical pore frameworks but defining differences in the sequence characteristics of the extracellular loops. Antibiotic sensitivity profiles of isogenic Klebsiella pneumoniae strains, each expressing a different porin as its dominant pore, revealed striking differences in the antibiotic permeability characteristics of each channel in a physiological context. Since K. pneumoniae is a nosocomial pathogen with high rates of antimicrobial resistance and concurrent mortality, these experiments elucidate the role of porins in conferring specific drug-resistant phenotypes in a global context, informing future research to combat antimicrobial resistance in K. pneumoniae.
Collapse
|
31
|
Ulloa ER, Dillon N, Tsunemoto H, Pogliano J, Sakoulas G, Nizet V. Avibactam Sensitizes Carbapenem-Resistant NDM-1-Producing Klebsiella pneumoniae to Innate Immune Clearance. J Infect Dis 2020; 220:484-493. [PMID: 30923801 PMCID: PMC6603980 DOI: 10.1093/infdis/jiz128] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/05/2019] [Indexed: 11/16/2022] Open
Abstract
Infections caused by New Delhi metallo-β-lactamase (NDM)–producing strains of multidrug-resistant Klebsiella pneumoniae are a global public health threat lacking reliable therapies. NDM is impervious to all existing β-lactamase inhibitor (BLI) drugs, including the non–β-lactam BLI avibactam (AVI). Though lacking direct activity against NDMs, AVI can interact with penicillin-binding protein 2 in a manner that may influence cell wall dynamics. We found that exposure of NDM-1–producing K. pneumoniae to AVI led to striking bactericidal interactions with human cathelicidin antimicrobial peptide LL-37, a frontline component of host innate immunity. Moreover, AVI markedly sensitized NDM-1–producing K. pneumoniae to killing by freshly isolated human neutrophils, platelets, and serum when complement was active. Finally, AVI monotherapy reduced lung counts of NDM-1–producing K. pneumoniae in a murine pulmonary challenge model. AVI sensitizes NDM-1–producing K. pneumoniae to innate immune clearance in ways that are not appreciated by standard antibiotic testing and that merit further study.
Collapse
Affiliation(s)
- Erlinda R Ulloa
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, La Jolla.,Division of Infectious Disease, Department of Pediatrics, Children's Hospital of Philadelphia, Pennsylvania
| | - Nicholas Dillon
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, La Jolla
| | - Hannah Tsunemoto
- Division of Biological Sciences, University of California-San Diego, La Jolla
| | - Joe Pogliano
- Division of Biological Sciences, University of California-San Diego, La Jolla
| | - George Sakoulas
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, La Jolla.,Sharp Healthcare System, San Diego, California
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, La Jolla.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla
| |
Collapse
|
32
|
Côté-Gravel J, Brouillette E, Malouin F. Vaccination with a live-attenuated small-colony variant improves the humoral and cell-mediated responses against Staphylococcus aureus. PLoS One 2019; 14:e0227109. [PMID: 31881064 PMCID: PMC6934294 DOI: 10.1371/journal.pone.0227109] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/12/2019] [Indexed: 02/07/2023] Open
Abstract
Staphylococcus aureus is known to produce persistent and chronic infections in both humans and animals. It is recognized that small-colony variants (SCVs), which produce higher levels of biofilm and that are capable of intracellular persistence, contribute to the chronicity or recurrence of infections and that this phenotype is inherent to the pathogenesis process. Prevention of S. aureus infections through vaccination has not yet met with considerable success. Some of the current vaccine formulations for S. aureus bovine mastitis consist of inactivated S. aureus bacteria, sometimes combined to E. coli J5. As such, the stimulation of cell-mediated immunity by these vaccines might not be optimal. With this in mind, we recently engineered a genetically stable double mutant SCV (ΔvraGΔhemB), which was highly attenuated in a mastitis model of infection. The present work describes the immune responses elicited in mice by various experimental vaccine compositions including the live-attenuated SCV double mutant and its inactivated form, combined or not with inactivated E. coli J5. The live-attenuated SCV was found to provoke a strong and balanced humoral response in immunized mice, as well as strong proliferation of ex-vivo stimulated splenocytes isolated from these animals. These splenocytes were also found to release high concentration of IL-17 and IFN-γ when compared to every other vaccination formulation. Inversely, the inactivated whole-cell vaccine, alone or in combination with the E. coli J5 bacterin, elicited lower antibody titers and failed to induce Th1 or Th17 cell-mediated responses in the splenocyte proliferation assay. Our results suggest that live-attenuated SCVs can trigger host immunity differently than inactivated bacteria and could represent a suitable vector for inducing strong humoral and cell-mediated immune responses, which are crucial for protection. This could represent an important improvement over existing vaccine formulations for preventing S. aureus bovine mastitis and other infections caused by this pathogen.
Collapse
Affiliation(s)
- Julie Côté-Gravel
- Centre d’étude et de Valorisation de la Diversité Microbienne (CEVDM), Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - Eric Brouillette
- Centre d’étude et de Valorisation de la Diversité Microbienne (CEVDM), Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Canada
| | - François Malouin
- Centre d’étude et de Valorisation de la Diversité Microbienne (CEVDM), Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke, Canada
- * E-mail:
| |
Collapse
|
33
|
Chauzy A, Gaelzer Silva Torres B, Buyck J, de Jonge B, Adier C, Marchand S, Couet W, Grégoire N. Semimechanistic Pharmacodynamic Modeling of Aztreonam-Avibactam Combination to Understand Its Antimicrobial Activity Against Multidrug-Resistant Gram-Negative Bacteria. CPT Pharmacometrics Syst Pharmacol 2019; 8:815-824. [PMID: 31420947 PMCID: PMC6876579 DOI: 10.1002/psp4.12452] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/17/2019] [Indexed: 11/24/2022] Open
Abstract
Aztreonam-avibactam (ATM-AVI) is a promising combination to treat serious infections caused by multidrug-resistant (MDR) pathogens. Three distinct mechanisms of action have been previously characterized for AVI: inhibition of ATM degradation by β-lactamases, proper bactericidal effect, and enhancement of ATM bactericidal activity. The aim of this study was to quantify the individual contribution of each of the three AVI effects. In vitro static time-kill studies were performed on four MDR Enterobacteriaceae with different β-lactamase profiles. β-Lactamase activity was characterized by measuring ATM concentrations over 27 hours. Data were analyzed by a semimechanistic pharmacodynamics modeling approach. Surprisingly, even though AVI prevented ATM degradation, the combined bactericidal activity was mostly explained by the enhancement of ATM effect within clinical range of ATM (5-125 mg/L) and AVI concentrations (0.9-22.5 mg/L). Therefore, when selecting a β-lactamase inhibitor for combination with a β-lactam, its capability to enhance the β-lactam activity should be considered in addition to the spectrum of β-lactamases inhibited.
Collapse
Affiliation(s)
- Alexia Chauzy
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Université de PoitiersUFR de Médecine PharmaciePoitiersFrance
| | - Bruna Gaelzer Silva Torres
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Université de PoitiersUFR de Médecine PharmaciePoitiersFrance
| | - Julien Buyck
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Université de PoitiersUFR de Médecine PharmaciePoitiersFrance
| | | | - Christophe Adier
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Laboratoire de Toxicologie‐PharmacocinétiqueCHU de PoitiersPoitiersFrance
| | - Sandrine Marchand
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Université de PoitiersUFR de Médecine PharmaciePoitiersFrance
- Laboratoire de Toxicologie‐PharmacocinétiqueCHU de PoitiersPoitiersFrance
| | - William Couet
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Université de PoitiersUFR de Médecine PharmaciePoitiersFrance
- Laboratoire de Toxicologie‐PharmacocinétiqueCHU de PoitiersPoitiersFrance
| | - Nicolas Grégoire
- INSERMU1070Pôle Biologie SantéPoitiers Cedex 9France
- Université de PoitiersUFR de Médecine PharmaciePoitiersFrance
| |
Collapse
|
34
|
Reck F, Bermingham A, Blais J, Casarez A, Colvin R, Dean CR, Furegati M, Gamboa L, Growcott E, Li C, Lopez S, Metzger L, Nocito S, Ossola F, Phizackerley K, Rasper D, Shaul J, Shen X, Simmons RL, Tang D, Tashiro K, Yue Q. IID572: A New Potentially Best-In-Class β-Lactamase Inhibitor. ACS Infect Dis 2019; 5:1045-1051. [PMID: 30861342 DOI: 10.1021/acsinfecdis.9b00031] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Resistance in Gram-negative bacteria to β-lactam drugs is mediated primarily by the expression of β-lactamases, and co-dosing of β-lactams with a β-lactamase inhibitor (BLI) is a clinically proven strategy to address resistance. New β-lactamases that are not impacted by existing BLIs are spreading and creating the need for development of novel broader spectrum BLIs. IID572 is a novel broad spectrum BLI of the diazabicyclooctane (DBO) class that is able to restore the antibacterial activity of piperacillin against piperacillin/tazobactam-resistant clinical isolates. IID572 is differentiated from other DBOs by its broad inhibition of β-lactamases and the lack of intrinsic antibacterial activity.
Collapse
Affiliation(s)
- Folkert Reck
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Alun Bermingham
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Johanne Blais
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Anthony Casarez
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Richard Colvin
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Charles R. Dean
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Markus Furegati
- Synthesis and Technologies Group, Novartis Institutes for BioMedical Research, Klybeckstrasse 141, Basel 4057, Switzerland
| | - Luis Gamboa
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Ellena Growcott
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Cindy Li
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Sara Lopez
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Louis Metzger
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Sandro Nocito
- Synthesis and Technologies Group, Novartis Institutes for BioMedical Research, Klybeckstrasse 141, Basel 4057, Switzerland
| | - Flavio Ossola
- Synthesis and Technologies Group, Novartis Institutes for BioMedical Research, Klybeckstrasse 141, Basel 4057, Switzerland
| | - Kaci Phizackerley
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Dita Rasper
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Jacob Shaul
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Xiaoyu Shen
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Robert L. Simmons
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Dazhi Tang
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Kyuto Tashiro
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Qin Yue
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| |
Collapse
|
35
|
Godinez WJ, Chan H, Hossain I, Li C, Ranjitkar S, Rasper D, Simmons RL, Zhang X, Feng BY. Morphological Deconvolution of Beta-Lactam Polyspecificity in E. coli. ACS Chem Biol 2019; 14:1217-1226. [PMID: 31184469 DOI: 10.1021/acschembio.9b00141] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Beta-lactams comprise one of the earliest classes of antibiotic therapies. These molecules covalently inhibit enzymes from the family of penicillin-binding proteins (PBPs), which are essential in construction of the bacterial cell wall. As a result, beta-lactams cause striking changes to cellular morphology, the nature of which varies by the range of PBPs simultaneously engaged in the cell. The traditional method of exploring beta-lactam polyspecificity is a gel-based binding assay which is low-throughput and typically is run ex situ in cell extracts. Here, we describe a medium-throughput, image-based assay combined with machine learning methods to automatically profile the activity of beta-lactams in E. coli cells. By testing for morphological change across a panel of strains with perturbations to individual PBP enzymes, our approach automatically and quantifiably relates different beta-lactam antibiotics according to their preferences for individual PBPs in cells. We show the potential of our approach for guiding the design of novel inhibitors toward different PBP-binding profiles by predicting the mechanisms of two recently reported PBP inhibitors.
Collapse
Affiliation(s)
- William J. Godinez
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| | - Helen Chan
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| | - Imtiaz Hossain
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Cindy Li
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| | - Srijan Ranjitkar
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| | - Dita Rasper
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| | - Robert L. Simmons
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| | - Xian Zhang
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Brian Y. Feng
- Infectious Diseases, Novartis Institutes for BioMedical Research, Emeryville, California, United States
| |
Collapse
|
36
|
Viaggi V, Pini B, Tonolo S, Luzzaro F, Principe L. In vitro activity of ceftazidime/avibactam against clinical isolates of ESBL-producing Enterobacteriaceae in Italy. J Chemother 2019; 31:195-201. [PMID: 31130090 DOI: 10.1080/1120009x.2019.1620406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Resistance to carbapenems in Enterobacteriaceae is a serious concern for public health. Alternative treatment options involving carbapenem-sparing regimen for patients with serious infections caused by multidrug-resistant Enterobacteriaceae are urgently needed. Ceftazidime/avibactam (CZA) is a new combination of a third generation cephalosporin and a non-β-lactam β-lactamase inhibitor, in which avibactam is capable to expand the ceftazidime activity also against extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae. To date, no data exist regarding the activity of CZA against strains isolated in the Italian context, which is known as endemic for ESBL producers. The aim of this study was to evaluate the in vitro activity of CZA, in comparison to ceftazidime (CAZ), against 90 ESBL-producing Escherichia coli and Klebsiella pneumoniae isolates, collected from blood and urine samples at our Institute. Thus, avibactam has been able to restore the activity of CAZ in all cases, suggesting the potential use of CZA as a carbapenem-sparing model, especially when limited therapeutic options exist.
Collapse
Affiliation(s)
- Valentina Viaggi
- a Clinical Microbiology and Virology Unit , A. Manzoni Hospital , Lecco , Italy
| | - Beatrice Pini
- a Clinical Microbiology and Virology Unit , A. Manzoni Hospital , Lecco , Italy
| | - Silvia Tonolo
- a Clinical Microbiology and Virology Unit , A. Manzoni Hospital , Lecco , Italy
| | - Francesco Luzzaro
- a Clinical Microbiology and Virology Unit , A. Manzoni Hospital , Lecco , Italy
| | - Luigi Principe
- a Clinical Microbiology and Virology Unit , A. Manzoni Hospital , Lecco , Italy
| |
Collapse
|
37
|
El Hafi B, Rasheed SS, Abou Fayad AG, Araj GF, Matar GM. Evaluating the Efficacies of Carbapenem/β-Lactamase Inhibitors Against Carbapenem-Resistant Gram-Negative Bacteria in vitro and in vivo. Front Microbiol 2019; 10:933. [PMID: 31114565 PMCID: PMC6503214 DOI: 10.3389/fmicb.2019.00933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 04/12/2019] [Indexed: 11/13/2022] Open
Abstract
Background Carbapenem-resistant Gram-negative bacteria are a major clinical concern as they cause virtually untreatable infections since carbapenems are among the last-resort antimicrobial agents. β-Lactamases implicated in carbapenem resistance include KPC, NDM, and OXA-type carbapenemases. Antimicrobial combination therapy is the current treatment approach against carbapenem resistance in order to limit the excessive use of colistin; however, its advantages over monotherapy remain debatable. An alternative treatment strategy would be the use of carbapenem/β-lactamase inhibitor (βLI) combinations. In this study, we assessed the in vitro and in vivo phenotypic and molecular efficacies of three βLIs when combined with different carbapenems against carbapenem-resistant Gram-negative clinical isolates. The chosen βLIs were (1) Avibactam, against OXA-type carbapenemases, (2) calcium-EDTA, against NDM-1, and (3) Relebactam, against KPC-2. Methods Six Acinetobacter baumannii clinical isolates were screened for bla OXA-23-like, bla OXA-24/40, bla OXA-51-like, bla OXA-58, and bla OXA-143-like, and eight Enterobacteriaceae clinical isolates were screened for bla OXA-48, bla NDM-1, and bla KPC-2. The minimal inhibitory concentrations of Imipenem (IPM), Ertapenem (ETP), and Meropenem (MEM) with corresponding βLIs for each isolate were determined. The efficacy of the most suitable in vitro treatment option against each of bla OXA-48, bla NDM-1, and bla KPC-2 was assessed via survival studies in a BALB/c murine infection model. Finally, RT-qPCR was performed to assess the molecular response of the genes of resistance to the carbapenem/βLI combinations used under both in vitro and in vivo settings. Results Combining MEM, IPM, and ETP with the corresponding βLIs restored the isolates' susceptibilities to those antimicrobial agents in 66.7%, 57.1%, and 30.8% of the samples, respectively. Survival studies in mice revealed 100% survival rates when MEM was combined with either Avibactam or Relebactam against bla OXA-48 and bla KPC-2, respectively. RT-qPCR demonstrated the consistent overexpression of bla OXA-48 upon treatment, without hindering Avibactam's activity, while bla NDM-1 and bla KPC-2 experienced variable expression levels upon treatment under in vitro and in vivo settings despite their effective phenotypic results. Conclusion New carbapenem/βLI combinations may be viable alternatives to antimicrobial combination therapy as they displayed high efficacy in vitro and in vivo. Meropenem/Avibactam and Meropenem/Relebactam should be tested on larger sample sizes with different carbapenemases before progressing further in its preclinical development.
Collapse
Affiliation(s)
- Bassam El Hafi
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Sari S Rasheed
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Antoine G Abou Fayad
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - George F Araj
- Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.,Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
38
|
Levy N, Bruneau JM, Le Rouzic E, Bonnard D, Le Strat F, Caravano A, Chevreuil F, Barbion J, Chasset S, Ledoussal B, Moreau F, Ruff M. Structural Basis for E. coli Penicillin Binding Protein (PBP) 2 Inhibition, a Platform for Drug Design. J Med Chem 2019; 62:4742-4754. [PMID: 30995398 DOI: 10.1021/acs.jmedchem.9b00338] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Penicillin-binding proteins (PBPs) are the targets of the β-lactams, the most successful class of antibiotics ever developed against bacterial infections. Unfortunately, the worldwide and rapid spread of large spectrum β-lactam resistance genes such as carbapenemases is detrimental to the use of antibiotics in this class. New potent PBP inhibitors are needed, especially compounds that resist β-lactamase hydrolysis. Here we describe the structure of the E. coli PBP2 in its Apo form and upon its reaction with 2 diazabicyclo derivatives, avibactam and CPD4, a new potent PBP2 inhibitor. Examination of these structures shows that unlike avibactam, CPD4 can perform a hydrophobic stacking on Trp370 in the active site of E. coli PBP2. This result, together with sequence analysis, homology modeling, and SAR, allows us to propose CPD4 as potential starting scaffold to develop molecules active against a broad range of bacterial species at the top of the WHO priority list.
Collapse
Affiliation(s)
- Nicolas Levy
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France.,IGBMC , 1 Rue Laurent Fries , 67404 Illkirch , France
| | | | - Erwann Le Rouzic
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | - Damien Bonnard
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | | | - Audrey Caravano
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | | | - Julien Barbion
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | - Sophie Chasset
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | - Benoît Ledoussal
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | - François Moreau
- Mutabilis , 102 Avenue Gaston Roussel , 93230 Romainville , France
| | - Marc Ruff
- IGBMC , 1 Rue Laurent Fries , 67404 Illkirch , France
| |
Collapse
|
39
|
In Vitro Activity of Tebipenem (SPR859) against Penicillin-Binding Proteins of Gram-Negative and Gram-Positive Bacteria. Antimicrob Agents Chemother 2019; 63:AAC.02181-18. [PMID: 30718255 DOI: 10.1128/aac.02181-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/29/2019] [Indexed: 12/25/2022] Open
Abstract
Tebipenem (SPR859) is the microbiologically active form of SPR994 (tebipenem-pivoxil), an orally available carbapenem with activity against extended-spectrum β-lactamase (ESBL)-producing Enterobacteriaceae Measurement of the relative binding of SPR859 to the bacterial cell targets revealed that it is a potent inhibitor of multiple penicillin-binding proteins (PBPs) but primarily a Gram-negative PBP 2 inhibitor, similar to other compounds in this class. These data support further clinical development of SPR994.
Collapse
|
40
|
Dean CR, Barkan DT, Bermingham A, Blais J, Casey F, Casarez A, Colvin R, Fuller J, Jones AK, Li C, Lopez S, Metzger LE, Mostafavi M, Prathapam R, Rasper D, Reck F, Ruzin A, Shaul J, Shen X, Simmons RL, Skewes-Cox P, Takeoka KT, Tamrakar P, Uehara T, Wei JR. Mode of Action of the Monobactam LYS228 and Mechanisms Decreasing In Vitro Susceptibility in Escherichia coli and Klebsiella pneumoniae. Antimicrob Agents Chemother 2018; 62:e01200-18. [PMID: 30061293 PMCID: PMC6153799 DOI: 10.1128/aac.01200-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/25/2018] [Indexed: 12/14/2022] Open
Abstract
The monobactam scaffold is attractive for the development of new agents to treat infections caused by drug-resistant Gram-negative bacteria because it is stable to metallo-β-lactamases (MBLs). However, the clinically used monobactam aztreonam lacks stability to serine β-lactamases (SBLs) that are often coexpressed with MBLs. LYS228 is stable to MBLs and most SBLs. LYS228 bound purified Escherichia coli penicillin binding protein 3 (PBP3) similarly to aztreonam (derived acylation rate/equilibrium dissociation constant [k2/Kd ] of 367,504 s-1 M-1 and 409,229 s-1 M-1, respectively) according to stopped-flow fluorimetry. A gel-based assay showed that LYS228 bound mainly to E. coli PBP3, with weaker binding to PBP1a and PBP1b. Exposing E. coli cells to LYS228 caused filamentation consistent with impaired cell division. No single-step mutants were selected from 12 Enterobacteriaceae strains expressing different classes of β-lactamases at 8× the MIC of LYS228 (frequency, <2.5 × 10-9). At 4× the MIC, mutants were selected from 2 of 12 strains at frequencies of 1.8 × 10-7 and 4.2 × 10-9 LYS228 MICs were ≤2 μg/ml against all mutants. These frequencies compared favorably to those for meropenem and tigecycline. Mutations decreasing LYS228 susceptibility occurred in ramR and cpxA (Klebsiella pneumoniae) and baeS (E. coli and K. pneumoniae). Susceptibility of E. coli ATCC 25922 to LYS228 decreased 256-fold (MIC, 0.125 to 32 μg/ml) after 20 serial passages. Mutants accumulated mutations in ftsI (encoding the target, PBP3), baeR, acrD, envZ, sucB, and rfaI These results support the continued development of LYS228, which is currently undergoing phase II clinical trials for complicated intraabdominal infection and complicated urinary tract infection (registered at ClinicalTrials.gov under identifiers NCT03377426 and NCT03354754).
Collapse
Affiliation(s)
- Charles R Dean
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - David T Barkan
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Alun Bermingham
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Johanne Blais
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Fergal Casey
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Anthony Casarez
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Richard Colvin
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - John Fuller
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Adriana K Jones
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Cindy Li
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Sara Lopez
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Louis E Metzger
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Mina Mostafavi
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Ramadevi Prathapam
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Dita Rasper
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Folkert Reck
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Alexey Ruzin
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Jacob Shaul
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Xiaoyu Shen
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Robert L Simmons
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Peter Skewes-Cox
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Kenneth T Takeoka
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Pramila Tamrakar
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Tsuyoshi Uehara
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| | - Jun-Rong Wei
- Novartis Institutes for BioMedical Research, Emeryville, California, USA
| |
Collapse
|
41
|
Decuyper L, Deketelaere S, Vanparys L, Jukič M, Sosič I, Sauvage E, Amoroso AM, Verlaine O, Joris B, Gobec S, D'hooghe M. In Silico Design and Enantioselective Synthesis of Functionalized Monocyclic 3-Amino-1-carboxymethyl-β-lactams as Inhibitors of Penicillin-Binding Proteins of Resistant Bacteria. Chemistry 2018; 24:15254-15266. [PMID: 29882610 DOI: 10.1002/chem.201801868] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 06/07/2018] [Indexed: 01/20/2023]
Abstract
As a complement to the renowned bicyclic β-lactam antibiotics, monocyclic analogues provide a breath of fresh air in the battle against resistant bacteria. In that framework, the present study discloses the in silico design and unprecedented ten-step synthesis of eleven nocardicin-like enantiomerically pure 2-{3-[2-(2-aminothiazol-4-yl)-2-(methoxyimino)acetamido]-2-oxoazetidin-1-yl}acetic acids starting from serine as a readily accessible precursor. The capability of this novel class of monocyclic 3-amino-β-lactams to inhibit penicillin-binding proteins (PBPs) of various (resistant) bacteria was assessed, revealing the potential of α-benzylidenecarboxylates as interesting leads in the pursuit of novel PBP inhibitors. No deactivation by representative enzymes belonging to the four β-lactamase classes was observed, while weak inhibition of class C β-lactamase P99 was demonstrated.
Collapse
Affiliation(s)
- Lena Decuyper
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Sari Deketelaere
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Lore Vanparys
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Marko Jukič
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Izidor Sosič
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Eric Sauvage
- Center for Protein Engineering, Faculty of Sciences, University of Liège, Quartier Agora, Allée du 6 Août 13, Bât B6a, 4000, Liège-Sart Tilman, Belgium
| | - Ana Maria Amoroso
- Center for Protein Engineering, Faculty of Sciences, University of Liège, Quartier Agora, Allée du 6 Août 13, Bât B6a, 4000, Liège-Sart Tilman, Belgium
| | - Olivier Verlaine
- Center for Protein Engineering, Faculty of Sciences, University of Liège, Quartier Agora, Allée du 6 Août 13, Bât B6a, 4000, Liège-Sart Tilman, Belgium
| | - Bernard Joris
- Center for Protein Engineering, Faculty of Sciences, University of Liège, Quartier Agora, Allée du 6 Août 13, Bât B6a, 4000, Liège-Sart Tilman, Belgium
| | - Stanislav Gobec
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000, Ljubljana, Slovenia
| | - Matthias D'hooghe
- SynBioC Research Group, Department of Green Chemistry and Technology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| |
Collapse
|
42
|
A Systematic Approach to the Selection of the Appropriate Avibactam Concentration for Use with Ceftazidime in Broth Microdilution Susceptibility Testing. Antimicrob Agents Chemother 2018; 62:AAC.00223-18. [PMID: 29735568 DOI: 10.1128/aac.00223-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/03/2018] [Indexed: 11/20/2022] Open
Abstract
Selection of the avibactam concentration to combine with ceftazidime in susceptibility testing was determined using Gram-negative isolates with characterized β-lactamases predefined as susceptible or resistant, based on the known inhibition spectrum of avibactam. MIC values were determined by broth microdilution of ceftazidime with fixed concentrations and ratios of avibactam. A constant concentration of 4 μg/ml of avibactam was selected for susceptibility testing with ceftazidime because of its ability to correctly categorize susceptible and resistant isolates while minimizing categorical errors.
Collapse
|
43
|
Edoo Z, Iannazzo L, Compain F, Li de la Sierra Gallay I, van Tilbeurgh H, Fonvielle M, Bouchet F, Le Run E, Mainardi JL, Arthur M, Ethève-Quelquejeu M, Hugonnet JE. Synthesis of Avibactam Derivatives and Activity on β-Lactamases and Peptidoglycan Biosynthesis Enzymes of Mycobacteria. Chemistry 2018; 24:8081-8086. [DOI: 10.1002/chem.201800923] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Zainab Edoo
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
| | - Laura Iannazzo
- Laboratoire de Chimie et de Biochimie, Pharmacologiques et Toxicologiques; Université Paris Descartes, UMR 8601; 75005 Paris France
- CNRS UMR 8601; 75006 Paris France
| | - Fabrice Compain
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
- Service de Microbiologie; Assistance Publique-Hôpitaux de Paris; Hôpital Européen Georges Pompidou; Paris France
| | - Inès Li de la Sierra Gallay
- Fonction et Architecture des Assemblages Macromoléculaires; Institute for Integrative Biology of the Cell (I2BC); CEA; CNRS; Univ Paris-Sud; Université Paris-Saclay; 91198 Gif-sur-Yvette cedex France
| | - Herman van Tilbeurgh
- Fonction et Architecture des Assemblages Macromoléculaires; Institute for Integrative Biology of the Cell (I2BC); CEA; CNRS; Univ Paris-Sud; Université Paris-Saclay; 91198 Gif-sur-Yvette cedex France
| | - Matthieu Fonvielle
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
| | - Flavie Bouchet
- Laboratoire de Chimie et de Biochimie, Pharmacologiques et Toxicologiques; Université Paris Descartes, UMR 8601; 75005 Paris France
| | - Eva Le Run
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
| | - Jean-Luc Mainardi
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
- Service de Microbiologie; Assistance Publique-Hôpitaux de Paris; Hôpital Européen Georges Pompidou; Paris France
| | - Michel Arthur
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
| | - Mélanie Ethève-Quelquejeu
- Laboratoire de Chimie et de Biochimie, Pharmacologiques et Toxicologiques; Université Paris Descartes, UMR 8601; 75005 Paris France
- CNRS UMR 8601; 75006 Paris France
| | - Jean-Emmanuel Hugonnet
- Sorbonne Université; Sorbonne Paris Cité; Université Paris Descartes; Université Paris Diderot; INSERM; Centre de Recherche des Cordeliers; CRC; 75006 Paris France
| |
Collapse
|
44
|
Khan A, Miller WR, Arias CA. Mechanisms of antimicrobial resistance among hospital-associated pathogens. Expert Rev Anti Infect Ther 2018; 16:269-287. [PMID: 29617188 DOI: 10.1080/14787210.2018.1456919] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The introduction of antibiotics revolutionized medicine in the 20th-century permitting the treatment of once incurable infections. Widespread use of antibiotics, however, has led to the development of resistant organisms, particularly in the healthcare setting. Today, the clinician is often faced with pathogens carrying a cadre of resistance determinants that severely limit therapeutic options. The genetic plasticity of microbes allows them to adapt to stressors via genetic mutations, acquisition or sharing of genetic material and modulation of genetic expression leading to resistance to virtually any antimicrobial used in clinical practice. Areas covered: This is a comprehensive review that outlines major mechanisms of resistance in the most common hospital-associated pathogens including bacteria and fungi. Expert commentary: Understanding the genetic and biochemical mechanisms of such antimicrobial adaptation is crucial to tackling the rapid spread of resistance, can expose unconventional therapeutic targets to combat multidrug resistant pathogens and lead to more accurate prediction of antimicrobial susceptibility using rapid molecular diagnostics. Clinicians making treatment decisions based on the molecular basis of resistance may design therapeutic strategies that include de-escalation of broad spectrum antimicrobial usage, more focused therapies or combination therapies. These strategies are likely to improve patient outcomes and decrease the risk of resistance in hospital settings.
Collapse
Affiliation(s)
- Ayesha Khan
- a Department of Microbiology and Molecular Genetics , University of Texas McGovern Medical School , Houston , Texas , USA.,b Center for Antimicrobial Resistance and Microbial Genomics , University of Texas Health Science Center , Houston , TX , USA
| | - William R Miller
- b Center for Antimicrobial Resistance and Microbial Genomics , University of Texas Health Science Center , Houston , TX , USA.,c Department of Internal Medicine, Division of Infectious Diseases , McGovern Medical School
| | - Cesar A Arias
- a Department of Microbiology and Molecular Genetics , University of Texas McGovern Medical School , Houston , Texas , USA.,b Center for Antimicrobial Resistance and Microbial Genomics , University of Texas Health Science Center , Houston , TX , USA.,c Department of Internal Medicine, Division of Infectious Diseases , McGovern Medical School.,d Molecular Genetics and Antimicrobial Resistance Unit and International Center for Microbial Genomics , Universidad El Bosque , Bogota , Colombia.,e School of Public Health , UTHealth Center for Infectious Diseases , Houston , TX , USA
| |
Collapse
|
45
|
Mutations in Genes Encoding Penicillin-Binding Proteins and Efflux Pumps Play a Role in β-Lactam Resistance in Helicobacter cinaedi. Antimicrob Agents Chemother 2018; 62:AAC.02036-17. [PMID: 29203490 DOI: 10.1128/aac.02036-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/21/2017] [Indexed: 01/06/2023] Open
Abstract
β-Lactams are often used to treat Helicobacter cinaedi infections; however, the mechanism underlying β-lactam resistance is unknown. In this study, we investigated β-lactam resistance in an H. cinaedi strain, MRY12-0051 (MICs of amoxicillin [AMX] and ceftriaxone [CRO], 32 and 128 μg/ml; obtained from human feces). Based on a comparative whole-genome analysis of MRY12-0051 and the CRO-susceptible H. cinaedi strain MRY08-1234 (MICs of AMX and CRO, 1 and 4 μg/ml; obtained from human blood), we identified five mutations in genes encoding penicillin-binding proteins (PBPs), including two in pbpA, one in pbp2, and two in ftsI Transformation and penicillin binding assays indicated that CRO resistance was mainly associated with mutations in pbpA; mutations in ftsI also led to increased resistance to AMX. Knocking out cmeB and cmeD, which encode resistance-nodulation-division-type efflux pump components, in H. cinaedi type strain CCUG18818 (AMX MIC, 4 to 8 μg/ml) resulted in 8- and 64-fold decreases, respectively, in the AMX MIC. Hence, MICs of AMX in H. cinaedi become similar to those of Helicobacter pylori isolates in the absence of cmeD In conclusion, the difference in susceptibility to β-lactams between H. pylori and H. cinaedi is explained by differences in efflux pump components. Mutations in pbpA are the primary determinant of high resistance to β-lactams in H. cinaedi.
Collapse
|
46
|
Cherkaoui A, Diene SM, Fischer A, Leo S, François P, Schrenzel J. Transcriptional Modulation of Penicillin-Binding Protein 1b, Outer Membrane Protein P2 and Efflux Pump (AcrAB-TolC) during Heat Stress Is Correlated to Enhanced Bactericidal Action of Imipenem on Non-typeable Haemophilus influenzae. Front Microbiol 2018; 8:2676. [PMID: 29375536 PMCID: PMC5770572 DOI: 10.3389/fmicb.2017.02676] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/21/2017] [Indexed: 11/13/2022] Open
Abstract
Objective: The purpose of the present study was to investigate the penicillin binding proteins (PBPs), drug influx and efflux modulations during heat stress and their effects on the bactericidal action of imipenem on non-typeable Haemophilus influenzae (NTHi). Methods: The two NTHi clinical isolates (GE47 and GE88, imipenem MICs by E-test > 32 μg/mL) examined in this study were collected at Geneva University Hospitals. The imipenem killing activity was assessed after incubation of the NTHi strains at either 37 or 42°C for 3 h with increasing concentrations of imipenem. The detection of PBPs was carried out by Bocillin-FL. Global transcriptional changes were monitored by RNA-seq after pre-incubation of bacterial cells at either 37 or 42°C, and the expression levels of relevant target genes were confirmed by qRT-PCR. Results: Quantitation of NTHi viable cells after incubation with 0.25 μg/mL of imipenem for 3 h revealed more than a twofold decrease in GE47 and GE88 viable cells at 42°C as compared to 37°C. Transcriptome analysis showed that under heat stress conditions, there were 141 differentially expressed genes with a | log2(fold change)| > 1, including 67 up-regulated and 74 down-regulated genes. The expression levels of ponB (encoding PBP1b) and acrR (regulator of AcrAB-TolC efflux pump) were significantly increased at 42°C. In contrast, the transcript levels of ompP2 (encoding the outer membrane protein P2) and acrB gene (encoding AcrB) were significantly lower under heat stress condition. Conclusion: This study shows that the transcriptional modulation of ponB, ompP2, acrR, and acrB in the heat stress response is correlated to enhanced antimicrobial effects of imipenem on non-typeable H. influenzae.
Collapse
Affiliation(s)
- Abdessalam Cherkaoui
- Bacteriology Laboratory, Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Seydina M Diene
- Genomic Research Laboratory, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Adrien Fischer
- Bacteriology Laboratory, Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Stefano Leo
- Genomic Research Laboratory, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Patrice François
- Genomic Research Laboratory, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Schrenzel
- Bacteriology Laboratory, Division of Laboratory Medicine, Department of Genetics and Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland.,Genomic Research Laboratory, Division of Infectious Diseases, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
47
|
Kozlov RS, Stetsiouk OU, Andreeva IV. Ceftazidime-avibactam: new rules for the game against multidrug-resistant gram-negative bacteria. CLINICAL MICROBIOLOGY AND ANTIMICROBIAL CHEMOTHERAPY 2018. [DOI: 10.36488/cmac.2018.1.24-34] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The rapid spread of multidrug-resistant Gram-negative bacteria in hospital settings all over the world makes a demand for the new options to overcome antimicrobial resistance. Ceftazidime-avibactam is the first approved antibiotic that contains a new beta-lactamase inhibitor with unique properties. This review provides insight into the spectrum of activity, pharmacological characteristics, data on efficacy and safety of ceftazidime-avibactam obtained from the clinical trials and real clinical practice, as well as prospects for further studies and clinical application of this new antimicrobial agent.
Collapse
Affiliation(s)
- Roman S. Kozlov
- Institute of Antimicrobial Chemotherapy, Smolensk State Medical University (Smolensk, Russia)
| | - Olga U. Stetsiouk
- Institute of Antimicrobial Chemotherapy, Smolensk State Medical University (Smolensk, Russia)
| | - Irina V. Andreeva
- Institute of Antimicrobial Chemotherapy, Smolensk State Medical University (Smolensk, Russia)
| |
Collapse
|
48
|
Tuon FF, Rocha JL, Formigoni-Pinto MR. Pharmacological aspects and spectrum of action of ceftazidime-avibactam: a systematic review. Infection 2017; 46:165-181. [PMID: 29110143 DOI: 10.1007/s15010-017-1096-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/27/2017] [Indexed: 12/11/2022]
Abstract
PURPOSE Ceftazidime-avibactam is an antimicrobial association active against several Enterobacteriaceae species, including those resistant to carbapenem. Considering the importance of this drug in the current panorama of multidrug-resistant bacteria, we performed a systematic review about ceftazidime-avibactam with emphasis on clinical and pharmacological published data. METHODS A systematic search of the medical literature was performed. The databases searched included MEDLINE, EMBASE and Web of Science (until September 2017). The search terms used were 'avibactam', 'NXL104' and 'AVE1330A'. Bibliographies from those studies were also reviewed. Ceftazidime was not included as a search term, once relevant studies about avibactam in association with other drugs could be excluded. Only articles in English were selected. No statistical analysis or quality validation was included in this review. RESULTS A total of 151 manuscripts were included. Ceftazidime-avibactam has limited action against anaerobic bacteria. Avibactam is a potent inhibitor of class A, class C, and some class D enzymes, which includes KPC-2. The best pharmacodynamic profile of ceftazidime-avibactam is ƒT > MIC, validated in an animal model of soft tissue infection. Three clinical trials showed the efficacy of ceftazidime-avibactam in patients with intra-abdominal and urinary infections. Ceftazidime-avibactam has been evaluated versus meropenem/doripenem in hospitalized adults with nosocomial pneumonia, neutropenic patients and pediatric patients. CONCLUSION Ceftazidime-avibactam has a favorable pharmacokinetic profile for severe infections and highly active against carbapenemases of KPC-2 type.
Collapse
Affiliation(s)
- Felipe Francisco Tuon
- Department of Medicine, School of Health and Biosciences, Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil. .,Hospital de Clínicas-Serviço de Infectologia, 3º. andar, Rua General Carneiro, 180-Alto da Glória, Curitiba, PR, 80060-900, Brazil.
| | - Jaime L Rocha
- Department of Medicine, School of Health and Biosciences, Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil
| | | |
Collapse
|
49
|
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:e00389-17. [PMID: 28559260 PMCID: PMC5527577 DOI: 10.1128/aac.00389-17] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 05/13/2017] [Indexed: 01/05/2023] Open
Abstract
Carbapenemase-producing Enterobacteriaceae isolates (n = 110) from health care centers in central Indiana (from 2010 to 2013) were tested for susceptibility to combinations of avibactam (4 μg/ml) with ceftazidime, ceftaroline, or aztreonam. MIC50/MIC90 values were 1/2 μg/ml (ceftazidime-avibactam), 0.5/2 μg/ml (ceftaroline-avibactam), and 0.25/0.5 μg/ml (aztreonam-avibactam.) A β-lactam MIC of 8 μg/ml was reported for the three combinations against one Escherichia coli isolate with an unusual TIPY insertion following Tyr344 in penicillin-binding protein 3 (PBP 3) as the result of gene duplication.
Collapse
Affiliation(s)
| | | | | | - Gerald Denys
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Karen Bush
- Indiana University, Bloomington, Indiana, USA
| |
Collapse
|
50
|
Schillaci D, Spanò V, Parrino B, Carbone A, Montalbano A, Barraja P, Diana P, Cirrincione G, Cascioferro S. Pharmaceutical Approaches to Target Antibiotic Resistance Mechanisms. J Med Chem 2017; 60:8268-8297. [PMID: 28594170 DOI: 10.1021/acs.jmedchem.7b00215] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
There is urgent need for new therapeutic strategies to fight the global threat of antibiotic resistance. The focus of this Perspective is on chemical agents that target the most common mechanisms of antibiotic resistance such as enzymatic inactivation of antibiotics, changes in cell permeability, and induction/activation of efflux pumps. Here we assess the current landscape and challenges in the treatment of antibiotic resistance mechanisms at both bacterial cell and community levels. We also discuss the potential clinical application of chemical inhibitors of antibiotic resistance mechanisms as add-on treatments for serious drug-resistant infections. Enzymatic inhibitors, such as the derivatives of the β-lactamase inhibitor avibactam, are closer to the clinic than other molecules. For example, MK-7655, in combination with imipenem, is in clinical development for the treatment of infections caused by carbapenem-resistant Enterobacteriaceae and Pseudomonas aeruginosa, which are difficult to treat. In addition, other molecules targeting multidrug-resistance mechanisms, such as efflux pumps, are under development and hold promise for the treatment of multidrug resistant infections.
Collapse
Affiliation(s)
- Domenico Schillaci
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Carbone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| | - Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Sezione di Chimica e Tecnologie Farmaceutiche, Università degli Studi di Palermo , Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|