1
|
Candela A, Fernández-Billón M, Aja-Macaya P, González-Pinto L, Fraile-Ribot PA, Viedma E, Alonso-García I, Blanco-Martín T, Estévez-Alfaya R, Fernández-González A, Beceiro A, López-Causapé C, Oviaño M, Bou G, Oliver A, Arca-Suárez J. Rapid prediction of carbapenemases in Pseudomonas aeruginosa by imipenem/relebactam and MALDI-TOF MS. J Clin Microbiol 2025; 63:e0110524. [PMID: 40130831 PMCID: PMC12077086 DOI: 10.1128/jcm.01105-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 02/19/2025] [Indexed: 03/26/2025] Open
Abstract
Pseudomonas aeruginosa is a major nosocomial pathogen commonly involved in multidrug-resistant (MDR) infections that are very difficult to treat. Imipenem/relebactam is a new carbapenem/β-lactamase inhibitor combination with robust activity against P. aeruginosa. However, resistance is increasingly reported, and rapid detection is, therefore, crucial so that appropriate treatments can be prescribed. We have developed a rapid MALDI TOF-MS-based method that can accurately predict the presence of carbapenemases in P. aeruginosa using imipenem/relebactam. The method was developed using a retrospective and a prospective collection of 419 P. aeruginosa isolates (including recombinant isolates and WGS-characterized clinical strains) encompassing the most important β-lactam resistance mechanisms. The MALDI TOF-MS method is based on the detection of the hydrolysis of imipenem in the presence or absence of relebactam, measuring modifications in the mass spectra of imipenem after incubation with bacteria. The method was evaluated against a retrospective collection and then validated against 250 prospectively collected clinical isolates, showing a 98% (246/250) agreement between the phenotype and the MALDI-TOF MS hydrolysis result and a 100% accordance with the β-lactam resistance genotype. Some errors in detecting GES-producing isolates and in detecting different mutational resistance mechanisms associated with imipenem/relebactam resistance (MICs ranging from 4 to 8 mg/L) were observed. All results were obtained within 1 hour, positioning the MALDI-TOF-based test as a rapid and easy-to-perform method for detection of carbapenemases (except GES enzymes) in P. aeruginosa. Besides, implementation of the method in routine laboratory screening would facilitate the correct use of imipenem/relebactam to treat P. aeruginosa infections.IMPORTANCEWhile several rapid diagnostic methods have been developed for the detection of ESBLs and carbapenemases to improve treatment decision-making in Enterobacterales, there is a lack of approaches to rapidly identify resistance mechanisms and predict β-lactam susceptibility in Pseudomonas aeruginosa. Taking advantage of the mechanism of action and the high efficacy of the newly developed β-lactam/β-lactamase inhibitor combination imipenem/relebactam against P. aeruginosa, we developed a WGS-guided, MALDI-TOF-based algorithm that accurately predicts the presence of carbapenemase enzymes in this bacterium and aids in forecasting the imipenem/relebactam susceptibility profile. The implementation of this method in routine laboratory testing would provide significant support in the rapid decision-making for the use of imipenem/relebactam in severe P. aeruginosa infections.
Collapse
Affiliation(s)
- Ana Candela
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - María Fernández-Billón
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases and Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Lucía González-Pinto
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Pablo Arturo Fraile-Ribot
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases and Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Esther Viedma
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Isaac Alonso-García
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Tania Blanco-Martín
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Roberto Estévez-Alfaya
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Ana Fernández-González
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Alejandro Beceiro
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carla López-Causapé
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases and Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Marina Oviaño
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Germán Bou
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Oliver
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases and Instituto de Investigación Sanitaria Illes Balears, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Arca-Suárez
- Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Uehara T, Chatwin CL, Miller B, Edwards M, Stevenson A, Colombo J, Six DA, Daigle DM, Moeck G, Boyd SA, Pevear DC. Spectrum of cefepime-taniborbactam coverage against 190 β-lactamases defined in engineered isogenic Escherichia coli strains. Antimicrob Agents Chemother 2025; 69:e0169924. [PMID: 40167371 PMCID: PMC12057355 DOI: 10.1128/aac.01699-24] [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: 11/12/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Cefepime-taniborbactam is a β-lactam/β-lactamase inhibitor combination in clinical development for the treatment of Enterobacterales and Pseudomonas infections, including carbapenem-resistant Enterobacterales and multidrug-resistant Pseudomonas aeruginosa. Taniborbactam is a novel cyclic boronate with direct inhibitory activity against clinically relevant Ambler class A, B, C, and D β-lactamases. To further characterize the spectrum of β-lactamase coverage by cefepime-taniborbactam, we constructed 190 isogenic strains of Escherichia coli that constitutively expressed a different β-lactamase. Synthetic codon-optimized genes encoding the mature periplasmic protein linked to the TEM-1 signal sequence were used for optimized expression and periplasmic localization of the β-lactamase. The repertoire of β-lactamases consisted of 50 Ambler class A, 34 class B (metallo), 48 class C, and 58 class D enzymes known to mediate β-lactam resistance in the clinical isolates of Enterobacterales and P. aeruginosa. Overall, in the 190 isogenic strains, the MIC50/MIC90 values were 8/128 µg/mL for cefepime and >128/>128 µg/mL for ceftazidime. Cefepime-taniborbactam (MIC50/MIC90 of 0.25/8 µg/mL) showed greater activity than ceftazidime-avibactam (MIC50/MIC90 of 4/>128 µg/mL) and similar activity to aztreonam-avibactam (MIC50/MIC90 of 0.5/4 µg/mL). Cefepime-taniborbactam inhibited strains overproducing metallo-β-lactamases, including clinically important NDM and VIM enzymes, whereas ceftazidime-avibactam showed no coverage. Among the 129 β-lactamase-overproducing strains with increased cefepime MIC ≥16-fold relative to the control strain, taniborbactam potentiated cefepime MIC by ≥8-fold for 113 strains overexpressing β-lactamases (42 Ambler class A, 24 B, 23 C, and 24 D). Cefepime-taniborbactam demonstrated broader activity relative to ceftazidime-avibactam and comparable activity with aztreonam-avibactam in the overall coverage of both serine- and metallo-β-lactamases from all four Ambler classes.
Collapse
Affiliation(s)
| | | | | | | | | | - Jenna Colombo
- Venatorx Pharmaceuticals Inc, Malvern, Pennsylvania, USA
| | - David A. Six
- Venatorx Pharmaceuticals Inc, Malvern, Pennsylvania, USA
| | | | - Greg Moeck
- Venatorx Pharmaceuticals Inc, Malvern, Pennsylvania, USA
| | - Steven A. Boyd
- Venatorx Pharmaceuticals Inc, Malvern, Pennsylvania, USA
| | | |
Collapse
|
3
|
Boattini M, Gaibani P, Comini S, Costa C, Cavallo R, Broccolo F, Bianco G. In vitro activity and resistance mechanisms of novel antimicrobial agents against metallo-β-lactamase producers. Eur J Clin Microbiol Infect Dis 2025; 44:1041-1068. [PMID: 40064744 PMCID: PMC12062158 DOI: 10.1007/s10096-025-05080-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/18/2025] [Indexed: 05/09/2025]
Abstract
The carbapenemase-producing Gram-negative organisms represent an urgent clinical and public health concern, as they have been associated with increased mortality and high dissemination in healthcare settings. Although overall incidence rates of infections sustained by metallo-β-lactamase (MβL)-producers have remained lower than those sustained by other carbapenemase-producers, albeit with substantial geographic differences, a significant increase in the prevalence of MβL-producers has been observed over the last decade. The recent development of new antimicrobials expanded the armamentarium to counter the challenge of metallo-β-lactamase (MβL)-producers. Cefiderocol and aztreonam/avibactam are already clinically available and recommended by international guidelines. In addition, two new classes of β-lactam/ β-lactamase combinations are under clinical evaluation: (i) combination of β-lactam with novel boronic-derived inhibitors (e.g. taniborbactam and xeruborbactam), (ii) combination of β-lactam with last generation diazabicyclooctane β-lactamase inhibitors (e.g. zidebactam and nacubactam), active on most of serine-β-lactamases but also showing strong intrinsic activity on PBP-2. This review aims to provide up-to-date data on the characteristics, activity and emerging resistance mechanisms of the armamentarium of clinically available or soon-to-be introduced drugs for the treatment of MβL-producing Gram-negative organisms.
Collapse
Affiliation(s)
- Matteo Boattini
- Microbiology and Virology Unit, University Hospital Città Della Salute E Della Scienza Di Torino, Turin, Italy
- Department of Public Health and Paediatrics, University of Torino, Turin, Italy
- Lisbon Academic Medical Centre, Lisbon, Portugal
| | - Paolo Gaibani
- Microbiology and Virology Unit, Department of Pathology, Azienda Ospedaliera Universitaria Integrata Di Verona, Verona, Italy
- Department of Diagnostic and Public Health, Microbiology Section, University of Verona, Verona, Italy
| | - Sara Comini
- Operative Unit of Clinical Pathology, Carlo Urbani Hospital, Ancona, Italy
| | - Cristina Costa
- Microbiology and Virology Unit, University Hospital Città Della Salute E Della Scienza Di Torino, Turin, Italy
- Department of Public Health and Paediatrics, University of Torino, Turin, Italy
| | - Rossana Cavallo
- Microbiology and Virology Unit, University Hospital Città Della Salute E Della Scienza Di Torino, Turin, Italy
- Department of Public Health and Paediatrics, University of Torino, Turin, Italy
| | - Francesco Broccolo
- Department of Experimental Medicine, University of Salento, Lecce, Italy
| | - Gabriele Bianco
- Department of Experimental Medicine, University of Salento, Lecce, Italy.
| |
Collapse
|
4
|
Zins C, Pailhoriès H, Chenouard R, Corvec S, Dahyot S, Woerther PL, Eckert C, Pierrat G, Bourge X, Boyer S, Kempf M. Antimicrobial activity of new anti-Pseudomonas beta-lactam-beta-lactamase inhibitors against Pseudomonas aeruginosa respiratory isolates recovered during the study for Monitoring Antimicrobial Resistance Trends (SMART) program in France (2016-2022). Infect Dis Now 2025; 55:105056. [PMID: 40089153 DOI: 10.1016/j.idnow.2025.105056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/15/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVES To assess the susceptibility of ceftolozane/tazobactam (C/T) and comparators to Pseudomonas aeruginosa isolates recovered from respiratory-tract-infections (RTI) between 2016-2022 in the French SMART study. METHODS Antibiotic susceptibility testing and minimum inhibitory concentrations (MICs) of 717 P.aeruginosa isolates collected in five French hospitals were determined and interpreted according to the EUCAST-2022 guidelines. P. aeruginosa isolates resistant (R) to imipenem and/or C/T were screened by PCR for extended-spectrum-β-lactamases (ESBLs), AmpC and carbapenemase genes. The identified genes were sequenced and the variants determined. RESULTS All in all, 96.5 % of P. aeruginosa isolates were susceptible to C/T, comparable to the susceptibilities of meropenem-vaborbactam (MVB = 96.5 %), imipenem/relebactam (IMI/REL = 96.9 %) and ceftazidime-avibactam (C/A = 97.0 %). MIC50 and MIC90 for C/T were 0.5 and 2 mg/L respectively against the 717 isolates. Among the 242 isolates (33.7 %) resistant to at least one anti-Pseudomonas β-lactam, close to 90 % were susceptible to C/T, C/A, MVB and IMI/REL. Among the 80 isolates resistant to piperacillin-tazobactam, cefepime and ceftazidime, 76.3 % were susceptible to C/T and only IMI/REL and amikacin reached susceptibility exceeding 80 %. Among the 32 isolates resistant to imipenem and meropenem, susceptibility exceeding 60 % was observed only for IMI/REL, C/T, and C/A. For these strains, the MIC50 of C/T was 2 mg/L, while that of C/A was at the resistance threshold (8 mg/L). IMI/REL had the strongest activity (72 %) against the 25 isolates resistant to C/T. Lastly, 53 imipenem and/or C/T-R isolates harbored a class C β-lactam (blaPDC) variant, and one of them also carried the blaPER-1 gene and another, the blaVIM-2 gene. CONCLUSION C/T is a reliable treatment option in RTI caused by P. aeruginosa.
Collapse
Affiliation(s)
- Charlie Zins
- Département de Biologie des Agents Infectieux, UF de Bactériologie-Hygiène, Centre Hospitalier Universitaire Angers, France
| | - Hélène Pailhoriès
- Département de Biologie des Agents Infectieux, UF de Bactériologie-Hygiène, Centre Hospitalier Universitaire Angers, France; HIFIH Laboratoire UPRES EA3859, SFR ICAT 4208 Université Angers, Angers, France
| | - Rachel Chenouard
- Département de Biologie des Agents Infectieux, UF de Bactériologie-Hygiène, Centre Hospitalier Universitaire Angers, France
| | - Stéphane Corvec
- CHU Nantes, Service de Bactériologie et des Contrôles Microbiologiques, Université de Nantes, INSERM, INCIT U1302 Nantes, France
| | - Sandrine Dahyot
- Univ Rouen Normandie, Université de Caen Normandie, INSERM, Normandie Univ, DYNAMICURE UMR 1311, CHU Rouen, Service de Bactériologie, F-76000 Rouen, France
| | - Paul-Louis Woerther
- Laboratoire de Bactériologie, département de Microbiologie, Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Paris, France
| | - Catherine Eckert
- Département de Bactériologie, AP-HP, Sorbonne Université (Assistance Publique Hôpitaux de Paris), Groupe Hospitalier Universitaire, Sorbonne Université, Hôpital, Saint-Antoine, Paris, France; U1135, Centre d'Immunologie et Des Maladies Infectieuses (Cimi-Paris), Sorbonne Université, Paris, France
| | - Gautier Pierrat
- Département de Bactériologie, AP-HP, Sorbonne Université (Assistance Publique Hôpitaux de Paris), Groupe Hospitalier Universitaire, Sorbonne Université, Hôpital, Saint-Antoine, Paris, France
| | | | - Sophie Boyer
- Univ Rouen Normandie, Université de Caen Normandie, INSERM, Normandie Univ, DYNAMICURE UMR 1311, CHU Rouen, Service de Bactériologie, F-76000 Rouen, France
| | - Marie Kempf
- Département de Biologie des Agents Infectieux, UF de Bactériologie-Hygiène, Centre Hospitalier Universitaire Angers, France.
| |
Collapse
|
5
|
Le Terrier C, Bouvier M, Kerbol A, Dell'Acqua C, Nordmann P, Poirel L. In-vitro activity of the novel β-lactam/β-lactamase inhibitor combinations and cefiderocol against carbapenem-resistant Pseudomonas spp. clinical isolates collected in Switzerland in 2022. Eur J Clin Microbiol Infect Dis 2025; 44:571-585. [PMID: 39704920 PMCID: PMC11880081 DOI: 10.1007/s10096-024-04994-6] [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: 04/09/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024]
Abstract
To evaluate the in-vitro activity of the novel commercially-available drugs, including meropenem-vaborbactam (MEV), ceftazidime-avibactam (CZA), ceftolozane-tazobactam (C/T), imipenem-relebactam (IPR) as well as cefiderocol (FDC), against carbapenem-resistant Pseudomonas spp. (CRP) isolates. All CRP isolates collected at the Swiss National Reference Laboratory (NARA) over the year 2022 (n = 170) have been included. Most of these isolates (n = 121) were non-carbapenemase producers. Among the 49 carbapenemase producers, 47 isolates produced metallo-β-lactamases (MBL) including NDM-1 (n = 11), VIM-like (n = 28), IMP-like (n = 7), and both NDM-1 and VIM-2 (n = 1) and two isolates produced the class A carbapenemase GES-5. Susceptibility testing was determined by broth microdilution method (BMD), or disk diffusion test, and results interpreted following EUCAST guidelines. The susceptibility rates for MEV, CZA, C/T and IPR were found to be 41%, 45%, 59% and 58%, respectively, for the whole set of isolates tested. Among non-carbapenemase producers, susceptibility rates for these β-lactam/β-lactamase inhibitors (BL/BLI) combinations were higher, determined at 55%, 61%, 83%, and 82%, respectively. The overall susceptibility of carbapenemase-producing Pseudomonas spp. to novel BL/BLI was relatively low, while 80% of these isolates demonstrated susceptibility to FDC, with a similar proportion (79%) observed among MBL producers. A total of 10 MBL-producing isolates (6%), mainly NDM-1, were found to exhibit resistance to all drugs tested, with the exception of colistin. FDC exhibited an excellent in-vitro activity against this collection of CRP recovered from Switzerland in 2022, including MBL producers. The new BL/BLI combinations displayed significant activity against non-carbapenemase CRP, with IPR and C/T showing the highest susceptibility rates.
Collapse
Affiliation(s)
- Christophe Le Terrier
- Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Division of Intensive Care Unit, University Hospitals of Geneva, Geneva, Switzerland
- Emergency Department, Fribourg Hospitals, Fribourg, Switzerland
| | - Maxime Bouvier
- Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Fribourg, Switzerland
| | - Auriane Kerbol
- Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Fribourg, Switzerland
| | - Chloé Dell'Acqua
- Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
| | - Patrice Nordmann
- Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Fribourg, Switzerland
| | - Laurent Poirel
- Medical and Molecular Microbiology Unit, Department of Medicine, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 18, Fribourg, CH-1700, Switzerland.
- Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
6
|
Outeda-García M, Arca-Suárez J, Lence E, Rodriguez-Coello A, Maceiras R, Blanco-Martin T, Guijarro-Sánchez P, Gonzalez-Pinto L, Alonso-Garcia I, García-Pose A, Muras A, Rodriguez-Pallares S, Lasarte-Monterrubio C, Gonzalez-Bello C, Vázquez-Ucha JC, Bou G, Beceiro A. Advancements in the fight against globally distributed OXA-48 carbapenemase: evaluating the new generation of carbapenemase inhibitors. Antimicrob Agents Chemother 2025; 69:e0161424. [PMID: 39791889 PMCID: PMC11823609 DOI: 10.1128/aac.01614-24] [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: 10/29/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025] Open
Abstract
Carbapenemase OXA-48 and its variants pose a serious threat to the development of effective treatments for bacterial infections. OXA-48-producing Enterobacterales are the most prevalent carbapenemase-producing bacteria in large parts of the world. Although these bacteria exhibit low-level carbapenem resistance in vitro, the infections they cause are challenging to treat with conventional therapies, owing to their spread and complex detection in clinical settings. However, numerous β-lactamase inhibitors (BLIs) are currently in the pipeline or late clinical stages. To assess the potential of these compounds, this study compared the efficacy against OXA-48 of novel β-lactamase inhibitors, specifically the 1,6-diazabicyclo[3,2,1]octanes (DBOs) avibactam, relebactam, zidebactam, nacubactam, and durlobactam, along with the cyclic and bicyclic boronates vaborbactam, taniborbactam, and xeruborbactam. The extensive kinetics assays identified xeruborbactam, taniborbactam, and durlobactam, together with the already established avibactam, as BLIs with superior biochemical performance. Susceptibility testing further validated these findings but also demonstrated significantly improved bacterial killing by the DBOs zidebactam, nacubactam, and durlobactam. On the other hand, binding studies demonstrated the superior inhibitory capacity of the BLIs durlobactam and xeruborbactam. Combinations, such as cefepime/zidebactam, meropenem/nacubactam, and sulbactam/durlobactam, show promising activity against OXA-48-producing Enterobacterales, while ceftazidime/avibactam, cefepime/taniborbactam, and meropenem/xeruborbactam combinations also appear highly active, largely due to the excellent kinetics of these new inhibitors. Overall, this comprehensive analysis provides important insights into the effectiveness of new BLIs against OXA-48-producing Enterobacterales, highlighting xeruborbactam, durlobactam, and avibactam as leading candidates. Additionally, BLIs like zidebactam, nacubactam, and taniborbactam also showed potential in addressing the clinical challenges posed by OXA-48-mediated antimicrobial resistance.
Collapse
Affiliation(s)
- Michelle Outeda-García
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Jorge Arca-Suárez
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), A Coruna, Spain
| | - Emilio Lence
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela, Spain
| | - Arianna Rodriguez-Coello
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Romina Maceiras
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Tania Blanco-Martin
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Paula Guijarro-Sánchez
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Lucia Gonzalez-Pinto
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Isaac Alonso-Garcia
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Andrea García-Pose
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Andrea Muras
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Salud Rodriguez-Pallares
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Cristina Lasarte-Monterrubio
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
| | - Concepción Gonzalez-Bello
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, Santiago de Compostela, Spain
| | - Juan Carlos Vázquez-Ucha
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), A Coruna, Spain
| | - German Bou
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), A Coruna, Spain
| | - Alejandro Beceiro
- Microbiology department, A Coruna University Hospital (CHUAC), Institute of Biomedical Research of A Coruna (INIBIC), A Coruna, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), A Coruna, Spain
| |
Collapse
|
7
|
Vidal-Cortés P, Campos-Fernández S, Cuenca-Fito E, del Río-Carbajo L, Fernández-Ugidos P, López-Ciudad VJ, Nieto-del Olmo J, Rodríguez-Vázquez A, Tizón-Varela AI. Difficult-to-Treat Pseudomonas aeruginosa Infections in Critically Ill Patients: A Comprehensive Review and Treatment Proposal. Antibiotics (Basel) 2025; 14:178. [PMID: 40001421 PMCID: PMC11851922 DOI: 10.3390/antibiotics14020178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The management of infections caused by difficult-to-treat Pseudomonas aeruginosa in critically ill patients poses a significant challenge. Optimal antibiotic therapy is crucial for patient prognosis, yet the numerous resistance mechanisms of P. aeruginosa, which may even combine, complicate the selection of an appropriate antibiotic. In this review, we examine the epidemiology, resistance mechanisms, risk factors, and available and future therapeutic options, as well as strategies for treatment optimization. Finally, we propose a treatment algorithm to facilitate decision making based on the resistance patterns specific to each Intensive Care Unit.
Collapse
Affiliation(s)
- Pablo Vidal-Cortés
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Sandra Campos-Fernández
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Elena Cuenca-Fito
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Lorena del Río-Carbajo
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Paula Fernández-Ugidos
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Víctor J. López-Ciudad
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Jorge Nieto-del Olmo
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| | - Ana Rodríguez-Vázquez
- Hospital Pharmacy, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain;
| | - Ana I. Tizón-Varela
- Intensive Care Unit, Complexo Hospitalario Universitario de Ourense, 32003 Ourense, Spain; (S.C.-F.); (E.C.-F.); (L.d.R.-C.); (P.F.-U.); (V.J.L.-C.); (J.N.-d.O.); (A.I.T.-V.)
| |
Collapse
|
8
|
Karlowsky JA, Wise MG, Hackel MA, Six DA, Uehara T, Daigle DM, Pevear DC, Moeck G, Sahm DF. Cefepime-taniborbactam activity against antimicrobial-resistant clinical isolates of Enterobacterales and Pseudomonas aeruginosa: GEARS global surveillance programme 2018-22. J Antimicrob Chemother 2024; 79:3116-3131. [PMID: 39287999 PMCID: PMC11638001 DOI: 10.1093/jac/dkae329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES Taniborbactam is a boronate-based β-lactamase inhibitor in clinical development in combination with cefepime. METHODS Cefepime-taniborbactam and comparator broth microdilution MICs were determined for patient isolates of Enterobacterales (n = 20 725) and Pseudomonas aeruginosa (n = 7919) collected in 59 countries from 2018 to 2022. Taniborbactam was tested at a fixed concentration of 4 mg/L. Isolates with cefepime-taniborbactam MICs ≥ 16 mg/L underwent WGS. β-Lactamase genes were identified in additional meropenem-resistant isolates by PCR/Sanger sequencing. RESULTS Taniborbactam reduced the cefepime MIC90 value for all Enterobacterales from >16 to 0.25 mg/L (>64-fold). At ≤16 mg/L, cefepime-taniborbactam inhibited 99.5% of all Enterobacterales isolates; >95% of isolates with MDR and ceftolozane-tazobactam-resistant phenotypes; ≥ 89% of isolates with meropenem-resistant and difficult-to-treat-resistant (DTR) phenotypes; >80% of isolates with meropenem-vaborbactam-resistant and ceftazidime-avibactam-resistant phenotypes; 100% of KPC-positive, 99% of OXA-48-like-positive, 99% of ESBL-positive, 97% of acquired AmpC-positive, 95% of VIM-positive and 76% of NDM-positive isolates. Against P. aeruginosa, taniborbactam reduced the cefepime MIC90 value from 32 to 8 mg/L (4-fold). At ≤16 mg/L, cefepime-taniborbactam inhibited 96.5% of all P. aeruginosa isolates; 85% of meropenem-resistant phenotype isolates; 80% of isolates with MDR and meropenem-vaborbactam-resistant phenotypes; >70% of isolates with DTR, ceftazidime-avibactam-resistant and ceftolozane-tazobactam-resistant phenotypes; and 82% of VIM-positive isolates. Multiple potential mechanisms of resistance, including carriage of IMP, or alterations in PBP3 (ftsI), porins (decreased permeability) and efflux (up-regulation) were present in most isolates with cefepime-taniborbactam MICs ≥ 16 mg/L. CONCLUSIONS Cefepime-taniborbactam exhibited potent in vitro activity against Enterobacterales and P. aeruginosa, and inhibited most carbapenem-resistant isolates, including those carrying serine carbapenemases or NDM/VIM MBLs.
Collapse
Affiliation(s)
- James A Karlowsky
- IHMA, Schaumburg, IL, USA
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | | | | | - David A Six
- Venatorx Pharmaceuticals, Inc., Malvern, PA, USA
| | | | | | | | - Greg Moeck
- Venatorx Pharmaceuticals, Inc., Malvern, PA, USA
| | | |
Collapse
|
9
|
He J, Yang Z, Wang M, Jia R, Chen S, Liu M, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Sun D, Tian B, He Y, Wu Z, Cheng A, Zhu D. Integrative and conjugative elements of Pasteurella multocida: Prevalence and signatures in population evolution. Virulence 2024; 15:2359467. [PMID: 38808732 PMCID: PMC11141479 DOI: 10.1080/21505594.2024.2359467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024] Open
Abstract
Pasteurella multocida (P. multocida) is a bacterial pathogen responsible for a range of infections in humans and various animal hosts, causing significant economic losses in farming. Integrative and conjugative elements (ICEs) are important horizontal gene transfer elements, potentially enabling host bacteria to enhance adaptability by acquiring multiple functional genes. However, the understanding of ICEs in P. multocida and their impact on the transmission of this pathogen remains limited. In this study, 42 poultry-sourced P. multocida genomes obtained by high-throughput sequencing together with 393 publicly available P. multocida genomes were used to analyse the horizontal transfer of ICEs. Eighty-two ICEs were identified in P. multocida, including SXT/R391 and Tn916 subtypes, as well as three subtypes of ICEHin1056 family, with the latter being widely prevalent in P. multocida and carrying multiple resistance genes. The correlations between insertion sequences and resistant genes in ICEs were also identified, and some ICEs introduced the carbapenem gene blaOXA-2 and the bleomycin gene bleO to P. multocida. Phylogenetic and collinearity analyses of these bioinformatics found that ICEs in P. multocida were transmitted vertically and horizontally and have evolved with host specialization. These findings provide insight into the transmission and evolution mode of ICEs in P. multocida and highlight the importance of understanding these elements for controlling the spread of antibiotic resistance.
Collapse
Affiliation(s)
- Jiao He
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Zhishuang Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Mingshu Wang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Renyong Jia
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Shun Chen
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Mafeng Liu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Xinxin Zhao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Qiao Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Ying Wu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Shaqiu Zhang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Juan Huang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Xumin Ou
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Di Sun
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Bin Tian
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Yu He
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Zhen Wu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Anchun Cheng
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Institute of Veterinary Medicine and Immunology, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| | - Dekang Zhu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Sicence and Technology Department of Sichuan Province, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- Key Laboratory of Agricultural Bioinformatics, Ministry of Education, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People’s Republic of China, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Hidalgo-Tenorio C, Bou G, Oliver A, Rodríguez-Aguirregabiria M, Salavert M, Martínez-Martínez L. The Challenge of Treating Infections Caused by Metallo-β-Lactamase-Producing Gram-Negative Bacteria: A Narrative Review. Drugs 2024; 84:1519-1539. [PMID: 39467989 PMCID: PMC11652570 DOI: 10.1007/s40265-024-02102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 10/30/2024]
Abstract
Gram-negative multidrug-resistant (MDR) bacteria, including Enterobacterales, Acinetobacter baumannii, and Pseudomonas aeruginosa, pose a significant challenge in clinical practice. Infections caused by metallo-β-lactamase (MBL)-producing Gram-negative organisms, in particular, require careful consideration due to their complexity and varied prevalence, given that the microbiological diagnosis of these pathogens is intricate and compounded by challenges in assessing the efficacy of anti-MBL antimicrobials. We discuss both established and new approaches in the treatment of MBL-producing Gram-negative infections, focusing on 3 strategies: colistin; the recently approved combination of aztreonam with avibactam (or with ceftazidime/avibactam); and cefiderocol. Despite its significant activity against various Gram-negative pathogens, the efficacy of colistin is limited by resistance mechanisms, while nephrotoxicity and acute renal injury call for careful dosing and monitoring in clinical practice. Aztreonam combined with avibactam (or with avibactam/ceftazidime if aztreonam plus avibactam is not available) exhibits potent activity against MBL-producing Gram-negative pathogens. Cefiderocol in monotherapy is effective against a wide range of multidrug-resistant organisms, including MBL producers, and favorable clinical outcomes have been observed in various clinical trials and case series. After examining scientific evidence in the management of infections caused by MBL-producing Gram-negative bacteria, we have developed a comprehensive clinical algorithm to guide therapeutic decision making. We recommend reserving colistin as a last-resort option for MDR Gram-negative infections. Cefiderocol and aztreonam/avibactam represent favorable options against MBL-producing pathogens. In the case of P. aeruginosa with MBL-producing enzymes and with difficult-to-treat resistance, cefiderocol is the preferred option. Further research is needed to optimize treatment strategies and minimize resistance.
Collapse
Affiliation(s)
- Carmen Hidalgo-Tenorio
- Hospital Universitario Virgen de las Nieves de Granada, Instituto de Investigación Biosanitario de Granada (IBS-Granada), Granada, Spain.
- Departamento de Medicina, Universidad de Granada, Granada, Spain.
| | - German Bou
- Servicio de Microbiología, Complejo Hospitalario Universitario A Coruña, A Coruña, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Oliver
- Servicio de Microbiología y Unidad de Investigación, Hospital Son Espases, IdISBa, Palma de Mallorca, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Miguel Salavert
- Infectious Diseases Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Luis Martínez-Martínez
- Microbiology Unit, Hospital Universitario Reina Sofía, Córdoba, Spain
- Department of Agricultural Chemistry, Soil Sciences and Microbiology, Universidad de Córdoba, Córdoba, Spain
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
11
|
Bouvier M, Bachtarzi M, Poirel L, Nordmann P. Rapid detection of imipenem/relebactam susceptibility/resistance in Pseudomonas aeruginosa. Diagn Microbiol Infect Dis 2024; 110:116474. [PMID: 39191152 DOI: 10.1016/j.diagmicrobio.2024.116474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/10/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVES Imipenem-relebactam (IPR) has been reported to exhibit a good activity against non-metallo-ß-lactamase carbapenem-resistant Pseudomonas aeruginosa (CRPA), and the rapid detection of susceptibility/resistance to this new therapeutic alternative may be crucial. Therefore, the Rapid IPR Pseudomonas NP test was developed to quickly identify IPR susceptibility/resistance among multidrug-resistant P. aeruginosa. METHODS The principle of the Rapid IPR Pseudomonas NP test is based on visually detecting glucose metabolization by observing (or not) a color change from yellow to red or orange of the red phenol pH indicator in the presence of imipenem at 2 mg/L and relebactam at 4 mg/L A total of 80 clinical Pseudomonas aeruginosa isolates were analyzed, among which 42 isolates were IPR resistant according to EUCAST guidelines (MICs, susceptible ≤2 mg/L, resistant >2 mg/L). Results obtained with the Rapid IPR Pseudomonas NP test were compared with the reference broth microdilution (BMD). RESULTS The sensitivity, specificity and accuracy of the test were found to be 100 %, 89.5 % and 95 %, respectively, using the BMD reference method as a comparator. Moreover, five out of the IPR-susceptible isolates (n = 38) exhibiting an MIC of IPR close to the breakpoint (MIC = 1 mg/L, n = 2; MIC = 2 mg/L, n = 3) yielded to a major error result, namely a positive result with the rapid IPR Pseudomonas NP test (resistance). By contrast, all IPR-resistant isolates (n = 42) were all correctly categorized. CONCLUSIONS The Rapid IPR Pseudomonas NP test is sensitive, specific, and easy to perform and interpret. Therefore, it is suitable for implementation in routine clinical laboratories.
Collapse
Affiliation(s)
- Maxime Bouvier
- Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Switzerland
| | - Mohamed Bachtarzi
- Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Medical Microbiology laboratory, CHU Mustapha Bacha, Alger, Algeria
| | - Laurent Poirel
- Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Switzerland
| | - Patrice Nordmann
- Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Swiss National Reference Center for Emerging Antibiotic Resistance (NARA), University of Fribourg, Switzerland.
| |
Collapse
|
12
|
Falcone M, Galfo V, Tiseo G. Not all carbapenem-resistant Pseudomonas aeruginosa strains are alike: tailoring antibiotic therapy based on resistance mechanisms. Curr Opin Infect Dis 2024; 37:594-601. [PMID: 39149832 PMCID: PMC11556876 DOI: 10.1097/qco.0000000000001044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
PURPOSE OF REVIEW To correlate the resistance mechanisms and the susceptibility to new antibiotics in Pseudomonas aeruginosa . RECENT FINDINGS Definition of antibiotic resistance in Pseudomonas aeruginosa is still debated. Carbapenem-resistant Pseudomonas aeruginosa (CRPA) and difficult-to-treat resistant Pseudomonas aeruginosa (DTR-PA) are used but which of them better correlate with the risk of mortality remains debated. Mechanisms underlying resistance in Pseudomonas aeruginosa are complex and may be combined, resulting in unpredictable phenotype and cross-resistance. Thus, not all CRPA are alike and tailoring antibiotic therapy on resistance mechanisms is challenging. SUMMARY Current guidelines recommend the use of new antipseudomonal agents for CRPA or DTR-PA infections but they don't provide specific information on how tailoring antibiotic therapy on underlying resistance mechanisms. This review may be useful to understand which mechanisms are involved in CRPA and may have practical implications helping clinicians to select an appropriate antibiotic regimen. Several antibiotics are now available for Pseudomonas aeruginosa but their rational use is important to avoid development of future resistance. The knowledge of local epidemiology and most common resistance mechanisms may guide empirical therapy, but targeted antibiotic therapy should be re-evaluated as soon as susceptibility testing profile is available and selected according to Pseudomonas aeruginosa phenotype.
Collapse
Affiliation(s)
- Marco Falcone
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliero Universitaria Pisana, University of Pisa, Pisa, Italy
| | | | | |
Collapse
|
13
|
González-Pinto L, Blanco-Martín T, Alonso-García I, Rodríguez-Pallares S, Outeda-García M, Gomis-Font MA, Fraile-Ribot PA, Vázquez-Ucha JC, González-Bello C, Beceiro A, Oliver A, Bou G, Arca-Suárez J. Impact of transferable β-lactamases and intrinsic AmpC amino acid substitutions on the activity of cefiderocol against wild-type and iron uptake-deficient mutants of Pseudomonas aeruginosa. J Antimicrob Chemother 2024; 79:3023-3028. [PMID: 39287983 DOI: 10.1093/jac/dkae326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES We aimed to analyse the interplay between impaired iron uptake and β-lactamases on cefiderocol resistance in Pseudomonas aeruginosa. METHODS Thirty-one transferable β-lactamases and 16 intrinsic P. aeruginosa AmpC (PDC) variants were cloned and expressed in wild-type (PAO1) and iron uptake-deficient (PAO ΔpiuC) P. aeruginosa backgrounds. MICs of cefiderocol and antipseudomonal β-lactams were determined by reference broth microdilution. RESULTS Relative to PAO1, deletion of piuC caused a specific 16-fold decrease in cefiderocol activity but negligible effects on the activity of other β-lactams. Among transferable β-lactamases, SHV-12, KPC Ω-loop mutants, NDMs and OXA-15 showed cefiderocol MIC values above the clinical breakpoint (2 mg/L) when expressed in PAO1. When expressed in PAO ΔpiuC, these and the transformants harbouring PER-1, VEB-1, KPC-2, KPC-3, VIM-1, CMY-2, OXA-2 and OXA-14 showed increased MIC values from 16 to >256 mg/L. The PDC variants carrying the Ω-loop changes ΔP215-G222 (PDC-577), E219K (PDC-221 and PDC-558) and the H10 helix change L293P (PDC-219) had the greatest impact on cefiderocol resistance, with MICs of 2-4 mg/L in PAO1 and of up to 32-64 mg/L in PAO ΔpiuC. Widespread enzymes such as GES, CTX-M-9, CTX-M-15, VIM-2-like enzymes, IMPs, DHA-1, FOX-4, OXA-10, OXA-48 and the other PDC variants tested had weaker effects on cefiderocol resistance. CONCLUSION We add evidence about the effect of the interplay between iron uptake and β-lactamases on the acquisition of cefiderocol resistance in P. aeruginosa. These findings may help to anticipate the emergence of resistance and optimize the use of cefiderocol against P. aeruginosa infections.
Collapse
Affiliation(s)
- Lucía González-Pinto
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Tania Blanco-Martín
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Isaac Alonso-García
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Salud Rodríguez-Pallares
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Michelle Outeda-García
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - María Antonia Gomis-Font
- Servicio de Microbiología, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Pablo Arturo Fraile-Ribot
- Servicio de Microbiología, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Carlos Vázquez-Ucha
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Concepción González-Bello
- Departamento de Química Orgánica, Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Universidade de Santiago de Compostela, Jenaro de la Fuente s/n, 15782 Santiago de Compostela, Spain
| | - Alejandro Beceiro
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Oliver
- Servicio de Microbiología, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Germán Bou
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Arca-Suárez
- Servicio de Microbiología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
14
|
Zhanel GG, Mansour C, Mikolayanko S, Lawrence CK, Zelenitsky S, Ramirez D, Schweizer F, Bay D, Adam H, Lagacé-Wiens P, Walkty A, Irfan N, Clark N, Nicolau D, Tascini C, Karlowsky JA. Cefepime-Taniborbactam: A Novel Cephalosporin/β-Lactamase Inhibitor Combination. Drugs 2024; 84:1219-1250. [PMID: 39214942 DOI: 10.1007/s40265-024-02082-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Taniborbactam (formerly known as VNRX-5133) is a novel bicyclic boronate β-lactamase inhibitor of serine β-lactamases (SBLs) [Ambler classes A, C, and D] and metallo-β-lactamases (MBLs) [Ambler class B], including NDM and VIM, but not IMP. Cefepime-taniborbactam is active in vitro against most isolates of carbapenem-resistant Enterobacterales (CRE) and carbapenem-resistant Pseudomonas aeruginosa (CRPA), including both carbapenemase-producing and carbapenemase-non-producing CRE and CRPA, as well as against multidrug-resistant (MDR), ceftazidime-avibactam-resistant, meropenem-vaborbactam-resistant, and ceftolozane-tazobactam-resistant Enterobacterales and P. aeruginosa. The addition of taniborbactam to cefepime resulted in a > 64-fold reduction in MIC90 compared with cefepime alone for a 2018-2021 global collection of > 13,000 clinical isolates of Enterobacterales. In the same study, against > 4600 P. aeruginosa, a fourfold MIC reduction was observed with cefepime-taniborbactam, compared with cefepime alone. Whole genome sequencing studies have shown that resistance towards cefepime-taniborbactam in Enterobacterales arises due to the presence of multiple resistance mechanisms, often in concert, including production of IMP, PBP3 alterations, permeability (porin) defects, and upregulation of efflux pumps. In P. aeruginosa, elevated cefepime-taniborbactam MICs are also associated with the presence of multiple, concurrent mechanisms, most frequently IMP, PBP3 mutations, and upregulation of efflux pumps, as well as AmpC (PDC) overexpression. The pharmacokinetics of taniborbactam are dose proportional, follow a linear model, and do not appear to be affected when combined with cefepime. Taniborbactam's approximate volume of distribution (Vd) at steady state is 20 L and the approximate elimination half-life (t½) is 2.3 h, which are similar to cefepime. Furthermore, like cefepime, taniborbactam is primarily cleared renally, and clearance corresponds with renal function. Pharmacodynamic studies (in vitro and in vivo) have reported that cefepime-taniborbactam has bactericidal activity against various β-lactamase-producing Gram-negative bacilli that are not susceptible to cefepime alone. It has been reported that antimicrobial activity best correlated with taniborbactam exposure (area under the curve). A phase III clinical trial showed that cefepime-taniborbactam (2 g/0.5 g administered as an intravenous infusion over 2 h) was superior to meropenem for the treatment of complicated urinary tract infection (cUTI), including acute pyelonephritis, caused by Enterobacterales species and P. aeruginosa while demonstrating similar safety compared with meropenem. The safety and tolerability of taniborbactam and cefepime-taniborbactam has been reported in one pharmacokinetic trial, and in two pharmacokinetic trials and one phase III clinical trial, respectively. Cefepime-taniborbactam appears to be well tolerated in both healthy subjects and patients. Headache and gastrointestinal upset are the most common drug-related adverse effects associated with cefepime-taniborbactam use. Cefepime-taniborbactam will likely have a role in the treatment of infections proven or suspected to be caused by MDR Gram-negative bacteria, including Enterobacterales and P. aeruginosa. In particular, it may be useful in the treatment of infections caused by isolates that harbor an MBL (NDM, VIM) enzyme, although further clinical data are needed. Additional safety and efficacy studies may support indications for cefepime-taniborbactam beyond cUTI.
Collapse
Affiliation(s)
- George G Zhanel
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| | - Celine Mansour
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Stacey Mikolayanko
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Courtney K Lawrence
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Sheryl Zelenitsky
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Danyel Ramirez
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, Canada
| | - Frank Schweizer
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Department of Chemistry, Faculty of Science, University of Manitoba, Winnipeg, MB, Canada
| | - Denice Bay
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Heather Adam
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Clinical Microbiology, Diagnostic Services, Shared Health, Winnipeg, MB, Canada
| | - Philippe Lagacé-Wiens
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Clinical Microbiology, Diagnostic Services, Shared Health, Winnipeg, MB, Canada
| | - Andrew Walkty
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Clinical Microbiology, Diagnostic Services, Shared Health, Winnipeg, MB, Canada
| | - Neal Irfan
- Department of Pharmacy, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Nina Clark
- Division of Infectious Diseases, Department of Medicine, Loyola University Medical Center, Maywood, IL, USA
| | - David Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, Hartford, CT, USA
| | - Carlo Tascini
- First Division of Infectious Diseases, Cotugno Hospital, Naples, Italy
| | - James A Karlowsky
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 502 Basic Medical Sciences Building, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
- Clinical Microbiology, Diagnostic Services, Shared Health, Winnipeg, MB, Canada
| |
Collapse
|
15
|
González-Pinto L, Alonso-García I, Blanco-Martín T, Camacho-Zamora P, Fraile-Ribot PA, Outeda-García M, Lasarte-Monterrubio C, Guijarro-Sánchez P, Maceiras R, Moya B, Juan C, Vázquez-Ucha JC, Beceiro A, Oliver A, Bou G, Arca-Suárez J. Impact of chromosomally encoded resistance mechanisms and transferable β-lactamases on the activity of cefiderocol and innovative β-lactam/β-lactamase inhibitor combinations against Pseudomonas aeruginosa. J Antimicrob Chemother 2024; 79:2591-2597. [PMID: 39073766 PMCID: PMC11441999 DOI: 10.1093/jac/dkae263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVES We aimed to compare the stability of the newly developed β-lactams (cefiderocol) and β-lactam/β-lactamase inhibitor combinations (ceftazidime/avibactam, ceftolozane/tazobactam, aztreonam/avibactam, cefepime/taniborbactam, cefepime/zidebactam, imipenem/relebactam, meropenem/vaborbactam, meropenem/nacubactam and meropenem/xeruborbactam) against the most clinically relevant mechanisms of mutational and transferable β-lactam resistance in Pseudomonas aeruginosa. METHODS We screened a collection of 61 P. aeruginosa PAO1 derivatives. Eighteen isolates displayed the most relevant mechanisms of mutational resistance to β-lactams. The other 43 constructs expressed transferable β-lactamases from genes cloned in pUCP-24. MICs were determined by reference broth microdilution. RESULTS Cefiderocol and imipenem/relebactam exhibited excellent in vitro activity against all of the mutational resistance mechanisms studied. Aztreonam/avibactam, cefepime/taniborbactam, cefepime/zidebactam, meropenem/vaborbactam, meropenem/nacubactam and meropenem/xeruborbactam proved to be more vulnerable to mutational events, especially to overexpression of efflux operons. The agents exhibiting the widest spectrum of activity against transferable β-lactamases were aztreonam/avibactam and cefepime/zidebactam, followed by cefepime/taniborbactam, cefiderocol, meropenem/xeruborbactam and meropenem/nacubactam. However, some MBLs, particularly NDM enzymes, may affect their activity. Combined production of certain enzymes (e.g. NDM-1) with increased MexAB-OprM-mediated efflux and OprD deficiency results in resistance to almost all agents tested, including last options such as aztreonam/avibactam and cefiderocol. CONCLUSIONS Cefiderocol and new β-lactam/β-lactamase inhibitor combinations show promising and complementary in vitro activity against mutational and transferable P. aeruginosa β-lactam resistance. However, the combined effects of efflux pumps, OprD deficiency and efficient β-lactamases could still result in the loss of all therapeutic options. Resistance surveillance, judicious use of new agents and continued drug development efforts are encouraged.
Collapse
Affiliation(s)
- Lucía González-Pinto
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Isaac Alonso-García
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Tania Blanco-Martín
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Pablo Camacho-Zamora
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Pablo Arturo Fraile-Ribot
- Servicio de Microbiología & Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Michelle Outeda-García
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Cristina Lasarte-Monterrubio
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Paula Guijarro-Sánchez
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Romina Maceiras
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Bartolome Moya
- Servicio de Microbiología & Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Juan
- Servicio de Microbiología & Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Carlos Vázquez-Ucha
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandro Beceiro
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Oliver
- Servicio de Microbiología & Instituto de Investigación Sanitaria Illes Balears (IdISBa), Hospital Universitario Son Espases, Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Germán Bou
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Arca-Suárez
- Servicio de Microbiología & Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
16
|
Smith HG, Basak S, Aniebok V, Beech MJ, Alshref FM, Allen MD, Farley AJM, Schofield CJ. Structural basis of Pseudomonas aeruginosa penicillin binding protein 3 inhibition by the siderophore-antibiotic cefiderocol. Chem Sci 2024:d4sc04937c. [PMID: 39328188 PMCID: PMC11423509 DOI: 10.1039/d4sc04937c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
The breakthrough cephalosporin cefiderocol, approved for clinical use in 2019, has activity against many Gram-negative bacteria. The catechol group of cefiderocol enables it to efficiently enter bacterial cells via the iron/siderophore transport system thereby reducing resistance due to porin channel mutations and efflux pump upregulation. Limited information is reported regarding the binding of cefiderocol to its key proposed target, the transpeptidase penicillin binding protein 3 (PBP3). We report studies on the reaction of cefiderocol and the related cephalosporins ceftazidime and cefepime with Pseudomonas aeruginosa PBP3, including inhibition measurements, protein observed mass spectrometry, and X-ray crystallography. The three cephalosporins form analogous 3-exomethylene products with P. aeruginosa PBP3 following elimination of the C3' side chain. pIC50 and k inact/K i measurements with isolated PBP3 imply ceftazidime and cefiderocol react less efficiently than cefepime and, in particular, meropenem with P. aeruginosa PBP3. Crystal structures inform on conserved and different interactions involved in binding of the three cephalosporins and meropenem to P. aeruginosa PBP3. The results will aid development of cephalosporins with improved PBP3 inhibition properties.
Collapse
Affiliation(s)
- Helen G Smith
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Shyam Basak
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Victor Aniebok
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Matthew J Beech
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Faisal M Alshref
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Department of Biochemistry, Faculty of Science, King AbdulAziz University Jeddah Saudi Arabia
| | - Mark D Allen
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Alistair J M Farley
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| | - Christopher J Schofield
- Department of Chemistry, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
- Ineos Oxford Institute for Antimicrobial Research, University of Oxford 12 Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
17
|
Maruri-Aransolo A, López-Causapé C, Hernández-García M, García-Castillo M, Caballero-Pérez JDD, Oliver A, Cantón R. In vitro activity of cefiderocol in Pseudomonas aeruginosa isolates from people with cystic fibrosis recovered during three multicentre studies in Spain. J Antimicrob Chemother 2024; 79:1432-1440. [PMID: 38708553 DOI: 10.1093/jac/dkae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
OBJECTIVES Despite the introduction of cystic fibrosis transmembrane conductance regulator (CFTR) modulators, Pseudomonas aeruginosa is still a major pathogen in people with cystic fibrosis (pwCF). We determine the activity of cefiderocol and comparators in a collection of 154 P. aeruginosa isolates recovered from pwCF during three multicentre studies performed in 17 Spanish hospitals in 2013, 2017 and 2021. METHODS ISO broth microdilution was performed and MICs were interpreted with CLSI and EUCAST criteria. Mutation frequency and WGS were also performed. RESULTS Overall, 21.4% were MDR, 20.8% XDR and 1.3% pandrug-resistant (PDR). Up to 17% of the isolates showed a hypermutator phenotype. Cefiderocol demonstrated excellent activity; only 13 isolates (8.4%) were cefiderocol resistant by EUCAST (none using CLSI). A high proportion of the isolates resistant to ceftolozane/tazobactam (71.4%), meropenem/vaborbactam (70.0%), imipenem/relebactam (68.0%) and ceftazidime/avibactam (55.6%) were susceptible to cefiderocol. Nine out of 13 cefiderocol-resistant isolates were hypermutators (P < 0.001). Eighty-three STs were detected, with ST98 being the most frequent. Only one isolate belonging to the ST175 high-risk clone carried blaVIM-2. Exclusive mutations affecting genes involved in membrane permeability, AmpC overexpression (L320P-AmpC) and efflux pump up-regulation were found in cefiderocol-resistant isolates (MIC = 4-8 mg/L). Cefiderocol resistance could also be associated with mutations in genes related to iron uptake (tonB-dependent receptors and pyochelin/pyoverdine biosynthesis). CONCLUSIONS Our results position cefiderocol as a therapeutic option in pwCF infected with P. aeruginosa resistant to most recent β-lactam/β-lactamase inhibitor combinations.
Collapse
Affiliation(s)
- Ainhize Maruri-Aransolo
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carla López-Causapé
- Servicio de Microbiología, Hospital Universitario de Son Espases and (IdISBa), Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Hernández-García
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - María García-Castillo
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Juan de Dios Caballero-Pérez
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Oliver
- Servicio de Microbiología, Hospital Universitario de Son Espases and (IdISBa), Palma de Mallorca, Spain
- CIBER de Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
Meschiari M, Faltoni M, Kaleci S, Tassoni G, Orlando G, Franceschini E, Burastero G, Bedini A, Serio L, Biagioni E, Melegari G, Venturelli C, Sarti M, Bertellini E, Girardis M, Mussini C. Intravenous fosfomycin in combination regimens as a treatment option for difficult-to-treat infections due to multi-drug-resistant Gram-negative organisms: A real-life experience. Int J Antimicrob Agents 2024; 63:107134. [PMID: 38453094 DOI: 10.1016/j.ijantimicag.2024.107134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/09/2024]
Abstract
AIM To investigate the efficacy of intravenous (IV) fosfomycin as combination therapy for treatment of difficult-to-treat (DTT) acute and subacute infections with multi-drug-resistant (MDR) Gram-negative bacteria (GNB), and risk factors associated with 90-day mortality. METHODS A retrospective, observational, monocentric study enrolled patients treated with IV fosfomycin in combination regimens (≥72 h) for proven DTT-MDR-GNB infection. Multi-variate regression analysis identified independent risk factors for 90-day mortality. A propensity score for receiving fosfomycin was performed to control for confounding factors. RESULTS In total, 70 patients were included in this study: 54.3% had carbapenem-resistant isolates, 31.4% had ceftazidime/avibactam-resistant isolates and 28.6% had ceftolozane/tazobactam-resistant isolates. The main pathogens were Pseudomonas aeruginosa (57.1%) and Klebsiella pneumoniae (22.9%). The most prevalent infections were nosocomial pneumonia (42.9%), osteomyelitis (17.1%) and intra-abdominal infections. All-cause 30- and 90-day mortality were 15.7% and 31.4%, respectively (18.9% and 50% considering acute DTT-MDR-GNB infections alone). Relapse at 30 days occurred in 22.9% of cases (29% with emergence of fosfomycin resistance). Mortality at 90 days was independently associated with septic shock and ceftolozane/tazobactam resistance. The relationship between resistance to ceftolozane/tazobactam and 90-day mortality was confirmed to be significant after adjustment by propensity score analysis (hazard ratio 5.84, 95% confidence interval 1.65-20.68; P=0.006). CONCLUSIONS Fosfomycin seems to be a promising salvage, combination treatment in DTT-MDR-GNB infections. Resistance to ceftolozane/tazobactam seems to be independently associated with treatment failure. Randomized clinical trials focusing on pathogen and infection sites are needed urgently to demonstrate the superiority of fosfomycin in combination with other agents for the resolution of DTT-MDR-GNB infections.
Collapse
Affiliation(s)
- Marianna Meschiari
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy.
| | - Matteo Faltoni
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Shaniko Kaleci
- Clinical and Experimental Medicine Department of Surgical, Medical , Dental and Morphological Sciences With Interest in Transplant Oncology and Regenerative Medicine University of Modena and Reggio Emilia, Modena, Italy
| | - Giovanni Tassoni
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Gabriella Orlando
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Erica Franceschini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Giulia Burastero
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Andrea Bedini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Lucia Serio
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Emanuela Biagioni
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Gabriele Melegari
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Claudia Venturelli
- Clinical Microbiology Laboratory, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Mario Sarti
- Clinical Microbiology Laboratory, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Elisabetta Bertellini
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Massimo Girardis
- Department of Anaesthesia and Intensive Care, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| | - Cristina Mussini
- Department of Infectious Diseases, Azienda Ospedaliero-Universitaria of Modena, Modena, Italy
| |
Collapse
|
19
|
Santerre Henriksen A, Jeannot K, Oliver A, Perry JD, Pletz MW, Stefani S, Morrissey I, Longshaw C. In vitro activity of cefiderocol against European Pseudomonas aeruginosa and Acinetobacter spp., including isolates resistant to meropenem and recent β-lactam/β-lactamase inhibitor combinations. Microbiol Spectr 2024; 12:e0383623. [PMID: 38483164 PMCID: PMC10986614 DOI: 10.1128/spectrum.03836-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/05/2024] [Indexed: 04/06/2024] Open
Abstract
Carbapenem-resistant Pseudomonas aeruginosa and Acinetobacter spp. represent major threats and have few approved therapeutic options. Non-fermenting Gram-negative isolates were collected from hospitalized inpatients from 49 sites in 6 European countries between 01 January 2020 and 31 December 2020 and underwent susceptibility testing against cefiderocol and β-lactam/β-lactamase inhibitor combinations. Meropenem-resistant (MIC >8 mg/L), cefiderocol-susceptible isolates were analyzed by PCR, and cefiderocol-resistant isolates were analyzed by whole-genome sequencing to identify resistance mechanisms. Overall, 1,451 (950 P. aeruginosa; 501 Acinetobacter spp.) isolates were collected, commonly from the respiratory tract (42.0% and 39.3%, respectively). Cefiderocol susceptibility was higher than β-lactam/β-lactamase inhibitor combinations against P. aeruginosa (98.9% vs 83.3%-91.4%), and P. aeruginosa resistant to meropenem (n = 139; 97.8% vs 12.2%-59.7%), β-lactam/β-lactamase inhibitor combinations (93.6%-98.1% vs 10.7%-71.8%), and both meropenem and ceftazidime-avibactam (96.7% vs 5.0%-45.0%) or ceftolozane-tazobactam (98.4% vs 8.1%-54.8%), respectively. Cefiderocol and sulbactam-durlobactam susceptibilities were high against Acinetobacter spp. (92.4% and 97.0%) and meropenem-resistant Acinetobacter spp. (n = 227; 85.0% and 93.8%) but lower against sulbactam-durlobactam- (n = 15; 13.3%) and cefiderocol- (n = 38; 65.8%) resistant isolates, respectively. Among meropenem-resistant P. aeruginosa and Acinetobacter spp., the most common β-lactamase genes were metallo-β-lactamases [30/139; blaVIM-2 (15/139)] and oxacillinases [215/227; blaOXA-23 (194/227)], respectively. Acquired β-lactamase genes were identified in 1/10 and 32/38 of cefiderocol-resistant P. aeruginosa and Acinetobacter spp., and pirA-like or piuA mutations in 10/10 and 37/38, respectively. Conclusion: cefiderocol susceptibility was high against P. aeruginosa and Acinetobacter spp., including meropenem-resistant isolates and those resistant to recent β-lactam/β-lactamase inhibitor combinations common in first-line treatment of European non-fermenters. IMPORTANCE This was the first study in which the in vitro activity of cefiderocol and non-licensed β-lactam/β-lactamase inhibitor combinations were directly compared against Pseudomonas aeruginosa and Acinetobacter spp., including meropenem- and β-lactam/β-lactamase inhibitor combination-resistant isolates. A notably large number of European isolates were collected. Meropenem resistance was defined according to the MIC breakpoint for high-dose meropenem, ensuring that data reflect antibiotic activity against isolates that would remain meropenem resistant in the clinic. Cefiderocol susceptibility was high against non-fermenters, and there was no apparent cross resistance between cefiderocol and β-lactam/β-lactamase inhibitor combinations, with the exception of sulbactam-durlobactam. These results provide insights into therapeutic options for infections due to resistant P. aeruginosa and Acinetobacter spp. and indicate how early susceptibility testing of cefiderocol in parallel with β-lactam/β-lactamase inhibitor combinations will allow clinicians to choose the effective treatment(s) from all available options. This is particularly important as current treatment options against non-fermenters are limited.
Collapse
Affiliation(s)
| | - Katy Jeannot
- Laboratory of Bacteriology, University Hospital of Besançon, University of Franche-Comté, Besançon, France
| | - Antonio Oliver
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Palma de Mallorca, Spain
| | - John D. Perry
- Microbiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Mathias W. Pletz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Ian Morrissey
- Antimicrobial Focus Ltd., Sawbridgeworth, United Kingdom
| | | | - ARTEMIS Study InvestigatorsWillingerBirgitLeysseneDavidCattoenChristianAlauzetCorentineBoyerPierreDuboisVéroniqueJeannotKatyCorvecStephaneLavigneJean-PhilippeGuillardThomasGontierAudrey MerensNaasThierryRohdeHolgerZiesingStefanImirzaliogluCanHunfeldKlaus-PeterJungJetteGatermannSörenPletzMathiasBiancoGabrieleGiammancoAnnaCarcioneDavideRaponiGiammarcoMatinatoCaterinaDomenicoEnea Gino DiGaibaniPaoloMarcheseAnnaArenaFabioNiccolaiClaudiaStefaniStefaniaPitartCristinaBarriosJose LuisCercenadoEmiliaBouGermanLopezAlicia BetetaCantonRafaelHontangasJose LopezGracia-AhufingerIreneOliverAntonioLopez-CereroLorenaLarrosaNievesWarehamDavidPerryJohnCaseyAnnaNahlJasvirHughesDanielCoyneMichaelListerMichelleAttwoodMarie
- Medical Affairs, Shionogi B.V., London, United Kingdom
- Laboratory of Bacteriology, University Hospital of Besançon, University of Franche-Comté, Besançon, France
- Servicio de Microbiología and Unidad de Investigación, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Palma de Mallorca, Spain
- Microbiology Department, Freeman Hospital, Newcastle upon Tyne, United Kingdom
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Jena, Germany
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
- Antimicrobial Focus Ltd., Sawbridgeworth, United Kingdom
| |
Collapse
|
20
|
Oliver A, Rojo-Molinero E, Arca-Suarez J, Beşli Y, Bogaerts P, Cantón R, Cimen C, Croughs PD, Denis O, Giske CG, Graells T, Daniel Huang TD, Iorga BI, Karatuna O, Kocsis B, Kronenberg A, López-Causapé C, Malhotra-Kumar S, Martínez LM, Mazzariol A, Meyer S, Naas T, Notermans DW, Oteo-Iglesias J, Pedersen T, Pirš M, Poeta P, Poirel L, Pournaras S, Sundsfjord A, Szabó D, Tambić-Andrašević A, Vatcheva-Dobrevska R, Vitkauskienė A, Jeannot K. Pseudomonasaeruginosa antimicrobial susceptibility profiles, resistance mechanisms and international clonal lineages: update from ESGARS-ESCMID/ISARPAE Group. Clin Microbiol Infect 2024; 30:469-480. [PMID: 38160753 DOI: 10.1016/j.cmi.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/18/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
SCOPE Pseudomonas aeruginosa, a ubiquitous opportunistic pathogen considered one of the paradigms of antimicrobial resistance, is among the main causes of hospital-acquired and chronic infections associated with significant morbidity and mortality. This growing threat results from the extraordinary capacity of P. aeruginosa to develop antimicrobial resistance through chromosomal mutations, the increasing prevalence of transferable resistance determinants (such as the carbapenemases and the extended-spectrum β-lactamases), and the global expansion of epidemic lineages. The general objective of this initiative is to provide a comprehensive update of P. aeruginosa resistance mechanisms, especially for the extensively drug-resistant (XDR)/difficult-to-treat resistance (DTR) international high-risk epidemic lineages, and how the recently approved β-lactams and β-lactam/β-lactamase inhibitor combinations may affect resistance mechanisms and the definition of susceptibility profiles. METHODS To address this challenge, the European Study Group for Antimicrobial Resistance Surveillance (ESGARS) from the European Society of Clinical Microbiology and Infectious Diseases launched the 'Improving Surveillance of Antibiotic-Resistant Pseudomonas aeruginosa in Europe (ISARPAE)' initiative in 2022, supported by the Joint programming initiative on antimicrobial resistance network call and included a panel of over 40 researchers from 18 European Countries. Thus, a ESGARS-ISARPAE position paper was designed and the final version agreed after four rounds of revision and discussion by all panel members. QUESTIONS ADDRESSED IN THE POSITION PAPER To provide an update on (a) the emerging resistance mechanisms to classical and novel anti-pseudomonal agents, with a particular focus on β-lactams, (b) the susceptibility profiles associated with the most relevant β-lactam resistance mechanisms, (c) the impact of the novel agents and resistance mechanisms on the definitions of resistance profiles, and (d) the globally expanding XDR/DTR high-risk lineages and their association with transferable resistance mechanisms. IMPLICATION The evidence presented herein can be used for coordinated epidemiological surveillance and decision making at the European and global level.
Collapse
Affiliation(s)
- Antonio Oliver
- Servicio de Microbiología, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
| | - Estrella Rojo-Molinero
- Servicio de Microbiología, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jorge Arca-Suarez
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Microbiología and Instituto de Investigación Biomédica A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - Yeşim Beşli
- Department of Medical Microbiology, Amerikan Hastanesi, Istanbul, Turkey
| | - Pierre Bogaerts
- National Center for Antimicrobial Resistance in Gram, CHU UCL Namur, Yvoir, Belgium
| | - Rafael Cantón
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Microbiología, Hospital Universitario Ramón y Cajal-IRYCIS, Madrid, Spain
| | - Cansu Cimen
- Institute for Medical Microbiology and Virology, University of Oldenburg, Oldenburg, Germany; Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter D Croughs
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Olivier Denis
- Department of Microbiology, CHU Namur Site-Godinne, Université Catholique de Louvain, Yvoir, Belgium; Ecole de Santé Publique, Université Libre de Bruxelles, Brussels, Belgium
| | - Christian G Giske
- Department of Clinical Microbiology, Karolinska University Hospital, Stockholm, Sweden; Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Tíscar Graells
- Department of Neurobiology, Care Sciences and Society (NVS), Division of Family Medicine and Primary Care, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Te-Din Daniel Huang
- National Center for Antimicrobial Resistance in Gram, CHU UCL Namur, Yvoir, Belgium
| | - Bogdan I Iorga
- CNRS, Institut de Chimie des Substances Naturelles, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Onur Karatuna
- EUCAST Development Laboratory, Clinical Microbiology, Central Hospital, Växjö, Sweden
| | - Béla Kocsis
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary
| | - Andreas Kronenberg
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Carla López-Causapé
- Servicio de Microbiología, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), Palma de Mallorca, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine & Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Luis Martínez Martínez
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Unidad de Microbiología, Hospital Universitario Reina Sofía, Departamento de Química Agrícola, Edafología y Microbiología, Universidad de Córdoba, e Instituto Maimonides de Investigación Biomédica de Córdoba (IMIBIC), Spain
| | - Annarita Mazzariol
- Microbiology and Virology Section, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Sylvain Meyer
- INSERM UMR 1092 and Université of Limoges, Limoges, France
| | - Thierry Naas
- Laboratoire Associé du Centre National de Référence de la Résistance aux Antibiotiques: Entérobactéries Résistantes aux Carbapénèmes, Le Kremlin-Bicêtre, France; Équipe INSERM ReSIST, Faculté de Médecine, Université Paris-Saclay, Paris, France
| | - Daan W Notermans
- Centre for Infectious Disease Control. National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Jesús Oteo-Iglesias
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain; Reference and Research Laboratory in Resistance to Antibiotics and Infections Related to Healthcare, National Centre for Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Torunn Pedersen
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway
| | - Mateja Pirš
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Patricia Poeta
- MicroART-Microbiology and Antibiotic Resistance Team, Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; Associated Laboratory for Green Chemistry (LAQV-REQUIMTE), University NOVA of Lisboa, Lisboa, Portugal; Veterinary and Animal Research Centre (CECAV), University of Trás-os-Montes and Alto Douro (UTAD), Vila Real, Portugal; University of Trás-os-Montes and Alto Douro, Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Vila Real, Portugal
| | - Laurent Poirel
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Department of Medicine, University of Fribourg, Fribourg, Switzerland; University of Fribourg, Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland
| | - Spyros Pournaras
- Laboratory of Clinical Microbiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Arnfinn Sundsfjord
- Norwegian National Advisory Unit on Detection of Antimicrobial Resistance, Department of Microbiology and Infection Control, University Hospital of North Norway, Tromsø, Norway; Research Group on Host-Microbe Interactions, Department of Medical Biology, UiT The Arctic University of Norway, Tromsø, Norway
| | - Dora Szabó
- Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary; Human Microbiota Study Group, Semmelweis University-Eötvös Lóránd Research Network, Budapest, Hungary
| | - Arjana Tambić-Andrašević
- Department of Clinical Microbiology, University Hospital for Infectious Diseases, Zagreb, Croatia; School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | | | - Astra Vitkauskienė
- Department of Laboratory Medicine, Faculty of Medicine, Medical Academy, Lithuanian University of Health Science, Kaunas, Lithuania
| | - Katy Jeannot
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire de Besançon, Besançon, France; Laboratoire associé du Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon, Besançon, France; Chrono-environnement UMR 6249, CNRS, Université Franche-Comté, Besançon, France
| |
Collapse
|
21
|
Wagenlehner FM, Gasink LB, McGovern PC, Moeck G, McLeroth P, Dorr M, Dane A, Henkel T. Cefepime-Taniborbactam in Complicated Urinary Tract Infection. N Engl J Med 2024; 390:611-622. [PMID: 38354140 DOI: 10.1056/nejmoa2304748] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
BACKGROUND Carbapenem-resistant Enterobacterales species and multidrug-resistant Pseudomonas aeruginosa are global health threats. Cefepime-taniborbactam is an investigational β-lactam and β-lactamase inhibitor combination with activity against Enterobacterales species and P. aeruginosa expressing serine and metallo-β-lactamases. METHODS In this phase 3, double-blind, randomized trial, we assigned hospitalized adults with complicated urinary tract infection (UTI), including acute pyelonephritis, in a 2:1 ratio to receive intravenous cefepime-taniborbactam (2.5 g) or meropenem (1 g) every 8 hours for 7 days; this duration could be extended up to 14 days in case of bacteremia. The primary outcome was both microbiologic and clinical success (composite success) on trial days 19 to 23 in the microbiologic intention-to-treat (microITT) population (patients who had a qualifying gram-negative pathogen against which both study drugs were active). A prespecified superiority analysis of the primary outcome was performed after confirmation of noninferiority. RESULTS Of the 661 patients who underwent randomization, 436 (66.0%) were included in the microITT population. The mean age of the patients was 56.2 years, and 38.1% were 65 years of age or older. In the microITT population, 57.8% of the patients had complicated UTI, 42.2% had acute pyelonephritis, and 13.1% had bacteremia. Composite success occurred in 207 of 293 patients (70.6%) in the cefepime-taniborbactam group and in 83 of 143 patients (58.0%) in the meropenem group. Cefepime-taniborbactam was superior to meropenem regarding the primary outcome (treatment difference, 12.6 percentage points; 95% confidence interval, 3.1 to 22.2; P = 0.009). Differences in treatment response were sustained at late follow-up (trial days 28 to 35), when cefepime-taniborbactam had higher composite success and clinical success. Adverse events occurred in 35.5% and 29.0% of patients in the cefepime-taniborbactam group and the meropenem group, respectively, with headache, diarrhea, constipation, hypertension, and nausea the most frequently reported; the frequency of serious adverse events was similar in the two groups. CONCLUSIONS Cefepime-taniborbactam was superior to meropenem for the treatment of complicated UTI that included acute pyelonephritis, with a safety profile similar to that of meropenem. (Funded by Venatorx Pharmaceuticals and others; CERTAIN-1 ClinicalTrials.gov number, NCT03840148.).
Collapse
Affiliation(s)
- Florian M Wagenlehner
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - Leanne B Gasink
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - Paul C McGovern
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - Greg Moeck
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - Patrick McLeroth
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - MaryBeth Dorr
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - Aaron Dane
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| | - Tim Henkel
- From Justus Liebig University, Giessen, Germany (F.M.W.); LBG Consulting, Philadelphia (L.B.G.), and Venatorx Pharmaceuticals, Malvern (P.C.M., G.M., M.D., T.H.) - both in Pennsylvania; Fortrea, Burlington, NC (P.M.); and DaneStat Consulting, Macclesfield, United Kingdom (A.D.)
| |
Collapse
|
22
|
Ono D, Mojica MF, Bethel CR, Ishii Y, Drusin SI, Moreno DM, Vila AJ, Bonomo RA. Structural role of K224 in taniborbactam inhibition of NDM-1. Antimicrob Agents Chemother 2024; 68:e0133223. [PMID: 38174924 PMCID: PMC10848753 DOI: 10.1128/aac.01332-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
Taniborbactam (TAN; VNRX-5133) is a novel bicyclic boronic acid β-lactamase inhibitor (BLI) being developed in combination with cefepime (FEP). TAN inhibits both serine and some metallo-β-lactamases. Previously, the substitution R228L in VIM-24 was shown to increase activity against oxyimino-cephalosporins like FEP and ceftazidime (CAZ). We hypothesized that substitutions at K224, the homologous position in NDM-1, could impact FEP/TAN resistance. To evaluate this, a library of codon-optimized NDM K224X clones for minimum inhibitory concentration (MIC) measurements was constructed; steady-state kinetics and molecular docking simulations were next performed. Surprisingly, our investigation revealed that the addition of TAN restored FEP susceptibility only for NDM-1, as the MICs for the other 19 K224X variants remained comparable to those of FEP alone. Moreover, compared to NDM-1, all K224X variants displayed significantly lower MICs for imipenem, tebipenem, and cefiderocol (32-, 133-, and 33-fold lower, respectively). In contrast, susceptibility to CAZ was mostly unaffected. Kinetic assays with the K224I variant, the only variant with hydrolytic activity to FEP comparable to NDM-1, confirmed that the inhibitory capacity of TAN was modestly compromised (IC50 0.01 µM vs 0.14 µM for NDM-1). Lastly, structural modeling and docking simulations of TAN in NDM-1 and in the K224I variant revealed that the hydrogen bond between TAN's carboxylate with K224 is essential for the productive binding of TAN to the NDM-1 active site. In addition to the report of NDM-9 (E149K) as FEP/TAN resistant, this study demonstrates the fundamental role of single amino acid substitutions in the inhibition of NDM-1 by TAN.
Collapse
Affiliation(s)
- Daisuke Ono
- Department of Medicine, Division of Infectious Diseases, 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
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Maria F. Mojica
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, USA
| | - Christopher R. Bethel
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Yoshikazu Ishii
- Department of Microbiology and Infectious Disease, Toho University School of Medicine, Tokyo, Japan
| | - Salvador I. Drusin
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
- Instituto de Química Rosario (IQUIR), CONICET, Rosario, Argentina
| | - Diego M. Moreno
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
- Instituto de Química Rosario (IQUIR), CONICET, Rosario, Argentina
| | - Alejandro J. Vila
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, Rosario, Argentina
- Instituto de Biología Molecular y Celular de Rosario (IBR), CONICET, Universidad Nacional de Rosario, Rosario, Argentina
| | - Robert A. Bonomo
- Department of Medicine, Division of Infectious Diseases, 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
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, USA
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Biochemistry, 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
- Clinician Scientist Investigator, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio, USA
| |
Collapse
|
23
|
Karakonstantis S, Rousaki M, Vassilopoulou L, Kritsotakis EI. Global prevalence of cefiderocol non-susceptibility in Enterobacterales, Pseudomonas aeruginosa, Acinetobacter baumannii, and Stenotrophomonas maltophilia: a systematic review and meta-analysis. Clin Microbiol Infect 2024; 30:178-188. [PMID: 37666449 DOI: 10.1016/j.cmi.2023.08.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Cefiderocol is a last resort option for carbapenem-resistant (CR) Gram-negative bacteria, especially metallo-β-lactamase-producing Pseudomonas aeruginosa and CR Acinetobacter baumannii. Monitoring global levels of cefiderocol non-susceptibility (CFDC-NS) is important. OBJECTIVES To systematically collate and examine studies investigating in vitro CFDC-NS and estimate the global prevalence of CFDC-NS against major Gram-negative pathogens. DATA SOURCES PubMed and Scopus, up to May 2023. STUDY ELIGIBILITY CRITERIA Eligible were studies reporting CFDC-NS in Enterobacterales, P. aeruginosa, A. baumannii, or Stenotrophomonas maltophilia clinical isolates. RISK-OF-BIAS ASSESSMENT Two independent reviewers extracted study data and assessed the risk of bias on the population, setting, and measurement (susceptibility testing) domains. DATA SYNTHESIS Binomial-Normal mixed-effects models were applied to estimate CFDC-NS prevalence by species, coresistance phenotype, and breakpoint definition (EUCAST, CLSI, and FDA). Sources of heterogeneity were investigated by subgroup and meta-regression analyses. RESULTS In all, 78 studies reporting 82 035 clinical isolates were analysed (87% published between 2020 and 2023). CFDC-NS prevalence (EUCAST breakpoints) was low overall but varied by species (S. maltophilia 0.4% [95% CI 0.2-0.7%], Enterobacterales 3.0% [95% CI 1.5-6.0%], P. aeruginosa 1.4% [95% CI 0.5-4.0%]) and was highest for A. baumannii (8.8%, 95% CI 4.9-15.2%). CFDC-NS was much higher in CR Enterobacterales (12.4%, 95% CI 7.3-20.0%) and CR A. baumannii (13.2%, 95% CI 7.8-21.5%), but relatively low for CR P. aeruginosa (3.5%, 95% CI 1.6-7.8%). CFDC-NS was exceedingly high in New Delhi metallo-β-lactamase-producing Enterobacterales (38.8%, 95% CI 22.6-58.0%), New Delhi metallo-β-lactamase-producing A. baumannii (44.7%, 95% CI 34.5-55.4%), and ceftazidime/avibactam-resistant Enterobacterales (36.6%, 95% CI 22.7-53.1%). CFDC-NS varied considerably with breakpoint definition, predominantly among CR bacteria. Additional sources of heterogeneity were single-centre investigations and geographical regions. CONCLUSIONS CFDC-NS prevalence is low overall, but alarmingly high for specific CR phenotypes circulating in some institutions or regions. Continuous surveillance and updating of global CFDC-NS estimates are imperative while cefiderocol is increasingly introduced into clinical practice. The need to harmonize EUCAST and CLSI breakpoints was evident.
Collapse
Affiliation(s)
- Stamatis Karakonstantis
- Internal Medicine Department, Infectious Diseases Division, University Hospital of Heraklion, Crete, Greece
| | - Maria Rousaki
- Master of Public Health Program, Department of Social Medicine, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Loukia Vassilopoulou
- 2nd Department of Internal Medicine, Venizeleio-Pananeio General Hospital, Heraklion, Crete, Greece
| | - Evangelos I Kritsotakis
- Laboratory of Biostatistics, Department of Social Medicine, School of Medicine, University of Crete, Heraklion, Crete, Greece; School of Health and Related Research, Faculty of Medicine, Dentistry and Health, University of Sheffield, Sheffield, UK.
| |
Collapse
|
24
|
Hernández-García M, García-Castillo M, Nieto-Torres M, Bou G, Ocampo-Sosa A, Pitart C, Gracia-Ahufinger I, Mulet X, Pascual Á, Tormo N, Oliver A, Ruiz-Garbajosa P, Cantón R. Deciphering mechanisms affecting cefepime-taniborbactam in vitro activity in carbapenemase-producing Enterobacterales and carbapenem-resistant Pseudomonas spp. isolates recovered during a surveillance study in Spain. Eur J Clin Microbiol Infect Dis 2024; 43:279-296. [PMID: 38041722 DOI: 10.1007/s10096-023-04697-4] [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: 08/17/2023] [Accepted: 10/27/2023] [Indexed: 12/03/2023]
Abstract
PURPOSE To characterize the resistance mechanisms affecting the cefepime-taniborbactam combination in a collection of carbapenemase-producing Enterobacterales (CPE) and carbapenem-resistant Pseudomonas spp. (predominantly P. aeruginosa; CRPA) clinical isolates. METHODS CPE (n = 247) and CRPA (n = 170) isolates were prospectively collected from patients admitted to 8 Spanish hospitals. Susceptibility to cefepime-taniborbactam and comparators was determined by broth microdilution. Cefepime-taniborbactam was the most active agent, inhibiting 97.6% of CPE and 67.1% of CRPA (MICs ≤ 8/4 mg/L). All isolates with cefepime-taniborbactam MIC > 8/4 mg/L (5 CPE and 52 CRPA) and a subset with MIC ≤ 8/4 mg/L (23 CPE and 24 CRPA) were characterized by whole genome sequencing. RESULTS A reduced cefepime-taniborbactam activity was found in two KPC-ST307-Klebsiella pneumoniae isolates with altered porins [KPC-62-K. pneumoniae (OmpA, OmpR/EnvZ), KPC-150-K. pneumoniae (OmpK35, OmpK36)] and one each ST133-VIM-1-Enterobacter hormaechei with altered OmpD, OmpR, and OmpC; IMP-8-ST24-Enterobacter asburiae; and NDM-5-Escherichia coli with an YRIN-inserted PBP3 and a mutated PBP2. Among the P. aeruginosa (68/76), elevated cefepime-taniborbactam MICs were mostly associated with GES-5-ST235, OXA-2+VIM-2-ST235, and OXA-2+VIM-20-ST175 isolates also carrying mutations in PBP3, efflux pump (mexR, mexZ) and AmpC (mpl) regulators, and non-carbapenemase-ST175 isolates with AmpD-T139M and PBP3-R504C mutations. Overall, accumulation of these mutations was frequently detected among non-carbapenemase producers. CONCLUSIONS The reduced cefepime-taniborbactam activity among the minority of isolates with elevated cefepime-taniborbactam MICs is not only due to IMP carbapenemases but also to the accumulation of multiple resistance mechanisms, including PBP and porin mutations in CPE and chromosomal mutations leading to efflux pumps up-regulation, AmpC overexpression, and PBP modifications in P. aeruginosa.
Collapse
Affiliation(s)
- Marta Hernández-García
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| | - María García-Castillo
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Nieto-Torres
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Germán Bou
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Microbiología, Hospital Universitario A Coruña and Instituto de Investigación Biomédica A Coruña (INIBIC), A Coruña, Spain
| | - Alain Ocampo-Sosa
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Microbiología, Hospital Universitario Marqués de Valdecilla and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), Santander, Spain
| | - Cristina Pitart
- Laboratorio de Microbiología, Hospital Clínic i Provincial, Barcelona, Spain
| | - Irene Gracia-Ahufinger
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- UGC de Microbiología, Hospital Universitario Reina Sofía-Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain
| | - Xavier Mulet
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Microbiología, Hospital Universitario Son Espases and Instituto de investigación Sanitaria Illes Balears (idISBa), Palma de Mallorca, Spain
| | - Álvaro Pascual
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Enfermedades Infecciosas y Microbiología Clínica, Hospital Universitario Virgen Macarena, Departamento de Microbiología, Universidad de Sevilla and Instituto de Biomedicina de Sevilla (IBiS) (Hospital Universitario Virgen Macarena/CSIC/Universidad de Sevilla), Sevilla, Spain
| | - Nuria Tormo
- Servicio de Microbiología, Consorcio Hospital General Universitario de Valencia, Valencia, Spain
| | - Antonio Oliver
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Microbiología, Hospital Universitario Son Espases and Instituto de investigación Sanitaria Illes Balears (idISBa), Palma de Mallorca, Spain
| | - Patricia Ruiz-Garbajosa
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
25
|
Le Terrier C, Freire S, Nordmann P, Poirel L. Multidrug-resistant Gram-negative clinical isolates with reduced susceptibility/resistance to cefiderocol: which are the best present and future therapeutic alternatives? Eur J Clin Microbiol Infect Dis 2024; 43:339-354. [PMID: 38095831 PMCID: PMC10821827 DOI: 10.1007/s10096-023-04732-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 11/29/2023] [Indexed: 01/28/2024]
Abstract
PURPOSE To evaluate the different present and future therapeutic β-lactam/β-lactamase inhibitor (BL/BLI) alternatives, namely aztreonam-avibactam, imipenem-relebactam, meropenem-vaborbactam, cefepime-zidebactam, cefepime-taniborbactam, meropenem-nacubactam, and sulbactam-durlobactam against clinical isolates showing reduced susceptibility or resistance to cefiderocol in Enterobacterales, Acinetobacter baumannii, and Pseudomonas aeruginosa. METHODS MIC values of aztreonam, aztreonam-avibactam, cefepime, cefepime-taniborbactam, cefepime-zidebactam, imipenem, imipenem-relebactam, meropenem, meropenem-vaborbactam, meropenem-nacubactam, sulbactam-durlobactam, and cefiderocol combined with a BLI were determined for 67, 9, and 11 clinical Enterobacterales, P. aeruginosa or A. baumannii isolates, respectively, showing MIC values of cefiderocol being ≥1 mg/L. If unavailable, the respective β-lactam breakpoints according to EUCAST were used for BL/BLI combinations. RESULTS For Enterobacterales, the susceptibility rates for aztreonam, cefepime, imipenem, and meropenem were 7.5%, 0%, 10.4%, and 10.4%, respectively, while they were much higher for cefepime-zidebactam (91%), cefiderocol-zidebactam (91%), meropenem-nacubactam (71.6%), cefiderocol-nacubactam (74.6%), and cefiderocol-taniborbactam (76.1%), as expected. For P. aeruginosa isolates, the higher susceptibility rates were observed for imipenem-relebactam, cefiderocol-zidebactam, and meropenem-vaborbactam (56% for all combinations). For A. baumannii isolates, lower susceptibility rates were observed with commercially or under development BL/BLI combos; however, a high susceptibility rate (70%) was found for sulbactam-durlobactam and when cefiderocol was associated to some BLIs. CONCLUSIONS Zidebactam- and nacubactam-containing combinations showed a significant in vitro activity against multidrug-resistant Enterobacterales clinical isolates with reduced susceptibility to cefiderocol. On the other hand, imipenem-relebactam and meropenem-vaborbactam showed the highest susceptibility rates against P. aeruginosa isolates. Finally, sulbactam-durlobactam and cefiderocol combined with a BLI were the only effective options against A. baumannii tested isolates.
Collapse
Affiliation(s)
- Christophe Le Terrier
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Department of Medicine, University of Fribourg, Chemin du Musée 18, CH-1700, Fribourg, Switzerland
- Division of Intensive Care Unit, University hospitals of Geneva, Geneva, Switzerland
| | - Samanta Freire
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Department of Medicine, University of Fribourg, Chemin du Musée 18, CH-1700, Fribourg, Switzerland
| | - Patrice Nordmann
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Department of Medicine, University of Fribourg, Chemin du Musée 18, CH-1700, Fribourg, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland
| | - Laurent Poirel
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Department of Medicine, University of Fribourg, Chemin du Musée 18, CH-1700, Fribourg, Switzerland.
- Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland.
| |
Collapse
|
26
|
Hujer AM, Marshall SH, Mack AR, Hujer KM, Bakthavatchalam YD, Umarkar K, Palwe SR, Takalkar S, Joshi PR, Shrivastava R, Periasamy H, Bhagwat SS, Patel MV, Veeraraghavan B, Bonomo RA. Transcending the challenge of evolving resistance mechanisms in Pseudomonas aeruginosa through β-lactam-enhancer-mechanism-based cefepime/zidebactam. mBio 2023; 14:e0111823. [PMID: 37889005 PMCID: PMC10746216 DOI: 10.1128/mbio.01118-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 09/14/2023] [Indexed: 10/28/2023] Open
Abstract
Compared to other genera of Gram-negative pathogens, Pseudomonas is adept in acquiring complex non-enzymatic and enzymatic resistance mechanisms thus remaining a challenge to even novel antibiotics including recently developed β-lactam and β-lactamase inhibitor combinations. This study shows that the novel β-lactam enhancer approach enables cefepime/zidebactam to overcome both non-enzymatic and enzymatic resistance mechanisms associated with a challenging panel of P. aeruginosa. This study highlights that the β-lactam enhancer mechanism is a promising alternative to the conventional β-lactam/β-lactamase inhibitor approach in combating ever-evolving MDR P. aeruginosa.
Collapse
Affiliation(s)
- Andrea M. Hujer
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Steven H. Marshall
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
| | - Andrew R. Mack
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Kristine M. Hujer
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, Cleveland, Ohio, USA
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Kushal Umarkar
- Wockhardt Research Centre, Aurangabad, Maharashtra, India
| | | | | | | | | | | | | | | | - Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Robert A. Bonomo
- Research Service, Louis Stokes Cleveland Department of Veterans Affairs, 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
- Departments of Pharmacology, Biochemistry, and Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, and the CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, USA
| |
Collapse
|
27
|
Russo C, Humphries R. Approaches to Testing Novel β-Lactam and β-Lactam Combination Agents in the Clinical Laboratory. Antibiotics (Basel) 2023; 12:1700. [PMID: 38136734 PMCID: PMC10740869 DOI: 10.3390/antibiotics12121700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/25/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
The rapid emergence of multi-drug resistant Gram-negative pathogens has driven the introduction of novel β-lactam combination agents (BLCs) to the antibiotic market: ceftolozane-tazobactam, ceftazidime-avibactam, meropenem-vaborbactam, imipenem-relebactam, cefiderocol, and sulbactam-durlobactam. These agents are equipped with innovative mechanisms that confer broad Gram-negative activity, notably against certain challenging carbapenemases. While their introduction offers a beacon of hope, clinical microbiology laboratories must navigate the complexities of susceptibility testing for these agents due to their diverse activity profiles against specific β-lactamases and the possibility of acquired resistance mechanisms in some bacterial isolates. This review explores the complexities of these novel antimicrobial agents detailing the intricacies of their application, providing guidance on the nuances of susceptibility testing, interpretation, and result reporting in clinical microbiology laboratories.
Collapse
Affiliation(s)
| | - Romney Humphries
- Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| |
Collapse
|
28
|
Cosentino F, Viale P, Giannella M. MDR/XDR/PDR or DTR? Which definition best fits the resistance profile of Pseudomonas aeruginosa? Curr Opin Infect Dis 2023; 36:564-571. [PMID: 37930070 PMCID: PMC10836784 DOI: 10.1097/qco.0000000000000966] [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: 11/07/2023]
Abstract
PURPOSE OF REVIEW The aim of this narrative review is to compare the prognostic utility of the new definition of difficult-to-treat resistance (DTR) vs. established definitions in patients with Pseudomonas aeruginosa infection to understand the therapeutic implications of resistance classification and its impact on clinical outcome. RECENT FINDINGS Among Gram-negative bacteria (GNB), P. aeruginosa (PA) is associated with high rates of morbidity and mortality, mostly related to its intrinsic capacity of developing antibiotic resistance. Several classifications of antibiotic resistance have been proposed in the last 15 years. The most common used is that from Magiorakos et al. including multidrug resistance (MDR), extensively drug-resistant (XDR) and pan drug resistance (PDR) according to the number of antibiotic classes showing in vitro activity. A further classification based on the resistance to specific antibiotic classes (i.e. fluoroquinolones, cephalosporins, carbapenem resistance) was also proposed. However, both of them have been criticized because of limited usefulness in clinical practice and for poor correlation with patient outcome, mainly in infections due to PA. More recently the new definition of difficult-to-treat resistance (DTR) has been proposed referring to nonsusceptibility to all first-line agents showing high-efficacy and low-toxicity (i.e. carbapenems, β-lactam-β-lactamase inhibitor combinations, and fluoroquinolones). Studies including large cohorts of patients with GNB bloodstream infections have confirmed the prognostic value of DTR classification and its clinical usefulness mainly in infections due to PA. Indeed, in the recent documents from the Infectious Diseases Society of America (IDSA) on the management of antibiotic resistant GNB infections, the DTR classification was applied to PA. SUMMARY DTR definition seems to identify better than MDR/XDR/PDR and single class resistant categories the cases of PA with limited treatment options. It requires periodic revision in order to remain up-to-date with the introduction of new antibiotics and the evolving pattern of resistance.
Collapse
Affiliation(s)
- Federica Cosentino
- Department of Medical and Surgical Sciences, University of Bologna
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierluigi Viale
- Department of Medical and Surgical Sciences, University of Bologna
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maddalena Giannella
- Department of Medical and Surgical Sciences, University of Bologna
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
29
|
Giovagnorio F, De Vito A, Madeddu G, Parisi SG, Geremia N. Resistance in Pseudomonas aeruginosa: A Narrative Review of Antibiogram Interpretation and Emerging Treatments. Antibiotics (Basel) 2023; 12:1621. [PMID: 37998823 PMCID: PMC10669487 DOI: 10.3390/antibiotics12111621] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Pseudomonas aeruginosa is a ubiquitous Gram-negative bacterium renowned for its resilience and adaptability across diverse environments, including clinical settings, where it emerges as a formidable pathogen. Notorious for causing nosocomial infections, P. aeruginosa presents a significant challenge due to its intrinsic and acquired resistance mechanisms. This comprehensive review aims to delve into the intricate resistance mechanisms employed by P. aeruginosa and to discern how these mechanisms can be inferred by analyzing sensitivity patterns displayed in antibiograms, emphasizing the complexities encountered in clinical management. Traditional monotherapies are increasingly overshadowed by the emergence of multidrug-resistant strains, necessitating a paradigm shift towards innovative combination therapies and the exploration of novel antibiotics. The review accentuates the critical role of accurate antibiogram interpretation in guiding judicious antibiotic use, optimizing therapeutic outcomes, and mitigating the propagation of antibiotic resistance. Misinterpretations, it cautions, can inadvertently foster resistance, jeopardizing patient health and amplifying global antibiotic resistance challenges. This paper advocates for enhanced clinician proficiency in interpreting antibiograms, facilitating informed and strategic antibiotic deployment, thereby improving patient prognosis and contributing to global antibiotic stewardship efforts.
Collapse
Affiliation(s)
- Federico Giovagnorio
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy; (F.G.); (S.G.P.)
| | - Andrea De Vito
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - Giordano Madeddu
- Unit of Infectious Diseases, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | | | - Nicholas Geremia
- Unit of Infectious Diseases, Department of Clinical Medicine, Ospedale “dell’Angelo”, 30174 Venice, Italy
- Unit of Infectious Diseases, Department of Clinical Medicine, Ospedale Civile “S.S. Giovanni e Paolo”, 30122 Venice, Italy
| |
Collapse
|
30
|
Jordana-Lluch E, Barceló IM, Escobar-Salom M, Estévez MA, Zamorano L, Gómez-Zorrilla S, Sendra E, Oliver A, Juan C. The balance between antibiotic resistance and fitness/virulence in Pseudomonas aeruginosa: an update on basic knowledge and fundamental research. Front Microbiol 2023; 14:1270999. [PMID: 37840717 PMCID: PMC10569695 DOI: 10.3389/fmicb.2023.1270999] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The interplay between antibiotic resistance and bacterial fitness/virulence has attracted the interest of researchers for decades because of its therapeutic implications, since it is classically assumed that resistance usually entails certain biological costs. Reviews on this topic revise the published data from a general point of view, including studies based on clinical strains or in vitro-evolved mutants in which the resistance phenotype is seen as a final outcome, i.e., a combination of mechanisms. However, a review analyzing the resistance/fitness balance from the basic research perspective, compiling studies in which the different resistance pathways and respective biological costs are individually approached, was missing. Here we cover this gap, specifically focusing on Pseudomonas aeruginosa, a pathogen that stands out because of its extraordinary capacity for resistance development and for which a considerable number of recent and particular data on the interplay with fitness/virulence have been released. The revised information, split into horizontally-acquired vs. mutation-driven resistance, suggests a great complexity and even controversy in the resistance-fitness/virulence balance in the acute infection context, with results ranging from high costs linked to certain pathways to others that are seemingly cost-free or even cases of resistance mechanisms contributing to increased pathogenic capacities. The elusive mechanistic basis for some enigmatic data, knowledge gaps, and possibilities for therapeutic exploitation are discussed. The information gathered suggests that resistance-fitness/virulence interplay may be a source of potential antipseudomonal targets and thus, this review poses the elementary first step for the future development of these strategies harnessing certain resistance-associated biological burdens.
Collapse
Affiliation(s)
- Elena Jordana-Lluch
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Isabel Mª Barceló
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - María Escobar-Salom
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Miguel A. Estévez
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
| | - Laura Zamorano
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Silvia Gómez-Zorrilla
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Infectious Diseases Service, Hospital del Mar, Hospital del Mar Research Institute, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelóna (UAB), Barcelona, Spain
| | - Elena Sendra
- Infectious Diseases Service, Hospital del Mar, Hospital del Mar Research Institute, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Universitat Autònoma de Barcelóna (UAB), Barcelona, Spain
| | - Antonio Oliver
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Carlos Juan
- Research Unit, University Hospital Son Espases-Health Research Institute of the Balearic Islands (IdISBa), Palma, Spain
- Department of Microbiology, University Hospital Son Espases, Palma, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
31
|
Vuillemin X, Da Silva M, Bour M, Landon C, Plésiat P, Jeannot K. Cefiderocol activity is compromised by acquired extended-spectrum oxacillinases in Pseudomonas aeruginosa. Int J Antimicrob Agents 2023; 62:106917. [PMID: 37429451 DOI: 10.1016/j.ijantimicag.2023.106917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/16/2023] [Accepted: 07/04/2023] [Indexed: 07/12/2023]
Abstract
OBJECTIVES Cefiderocol has an excellent in vitro activity on clinical strains of Pseudomonas aeruginosa (P. aeruginosa). However, the resistance of some isolates has been associated with the production of some β-lactamases. Whether some acquired extended-spectrum oxacillinases (ES-OXA) common in this species may compromise the susceptibility of P. aeruginosa to cefiderocol has not been evaluated so far. METHODS Eighteen genes encoding OXA belonging to the major subgroups identified in P. aeruginosa OXA-1 (n = 3); - 2 (n = 5); - 10 (n = 8), and - 46 (n = 2) were cloned into pUCP24 shuttle vector and transferred into reference strain PAO1. RESULTS Although production of the OXA-1 subgroup enzymes did not alter cefiderocol MICs, the β-lactamases of OXA-2, OXA-46, and four variants of the OXA-10 subgroup resulted in an 8-fold to 32-fold decrease in susceptibility in the PAO1 background. Interestingly, point mutations Ala149Pro and Asp150Gly in OXA-2 subgroup, Trp154Cys and Gly157Asp in OXA-10 subgroup (all located in the Ω loop), and the duplication of a Thr206 and a Gly207 in the β5-β6 loop of OXA-10 subgroup were related to decreased susceptibility to cefiderocol. We also showed that some ES-OXA, including the most frequent ES-OXA in P. aeruginosa strains, OXA-19 (derived from OXA-10 subgroup), significantly compromised activity of cefiderocol in addition to ceftazidime, ceftolozane/tazobactam, and ceftazidime/avibactam in clinical strains. CONCLUSION This work shows that several ES-OXA have a significant effect on cefiderocol susceptibility. Of concern are the Trp154Cys and Gly157Asp mutations that occur in some of these β-lactamases, as they are associated with a decreased activity of the most recent cephalosporins introduced to combat P. aeruginosa infections.
Collapse
Affiliation(s)
- Xavier Vuillemin
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire de Besançon, Besançon, France; Laboratoire Associé au Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon, Besançon, France; Chrono-environnement, Université Bourgogne-Franche Comté, Besançon, France
| | - Maëlle Da Silva
- Laboratoire Associé au Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | - Maxime Bour
- Laboratoire Associé au Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon, Besançon, France
| | | | - Patrick Plésiat
- Chrono-environnement, Université Bourgogne-Franche Comté, Besançon, France
| | - Katy Jeannot
- Laboratoire de Bactériologie, Centre Hospitalier Universitaire de Besançon, Besançon, France; Laboratoire Associé au Centre National de Référence de la Résistance aux Antibiotiques, Centre Hospitalier Universitaire de Besançon, Besançon, France; Chrono-environnement, Université Bourgogne-Franche Comté, Besançon, France.
| |
Collapse
|
32
|
Egge SL, Lewis JS, Hakki M. Case Commentary: Successful Use of Cefepime/Zidebactam (WCK 5222) as a Salvage Therapy for the Treatment of Disseminated Extensively Drug-Resistant New Delhi Metallo-β-Lactamase-Producing Pseudomonas aeruginosa Infection in an Adult Patient with Acute T-Cell Leukemia. Antimicrob Agents Chemother 2023; 67:e0066323. [PMID: 37395652 PMCID: PMC10433852 DOI: 10.1128/aac.00663-23] [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: 07/04/2023] Open
Abstract
Multidrug-resistant/extensively drug-resistant (MDR/XDR) Pseudomonas aeruginosa (PA) are critical antimicrobial resistance threats. Despite their increasing prevalence, treatment options for metallo-β-lactamase (MBL)-producing PA are limited, especially for New Delhi metallo-β-lactamase (NDM) producers. Pending further clinical studies, this case provides support for limited-scope use of cefepime-zidebactam for treating disseminated infections secondary to NDM-producing XDR PA. Susceptibilities should be tested and/or alternative regimens considered when treating isolates with alternative MBLs or increased efflux pump expression because some in vitro data suggest associated loss of cefepime-zidebactam susceptibility.
Collapse
Affiliation(s)
- Stephanie L. Egge
- Division of Infectious Diseases, Department of Internal Medicine, Houston Methodist Hospital, Houston, Texas, USA
| | - James S. Lewis
- Division of Infectious Diseases, Department of Internal Medicine, Oregon Health and Science University, Portland, Oregon, USA
- Department of Pharmacy, Oregon Health and Science University, Portland, Oregon, USA
| | - Morgan Hakki
- Division of Infectious Diseases, Department of Internal Medicine, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
33
|
Le Terrier C, Nordmann P, Freret C, Seigneur M, POIREL L. Impact of Acquired Broad Spectrum β-Lactamases on Susceptibility to Novel Combinations Made of β-Lactams (Aztreonam, Cefepime, Meropenem, and Imipenem) and Novel β-Lactamase Inhibitors in Escherichia coli and Pseudomonas aeruginosa. Antimicrob Agents Chemother 2023; 67:e0033923. [PMID: 37255469 PMCID: PMC10353362 DOI: 10.1128/aac.00339-23] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/15/2023] [Indexed: 06/01/2023] Open
Abstract
The impact of broad-spectrum β-lactamases on the susceptibility to novel β-lactamase/β-lactamase inhibitor combinations was evaluated both in Pseudomonas aeruginosa and Escherichia coli using isogenic backgrounds. Cefepime-zidebactam displayed low MICs, mainly due to the significant intrinsic antibacterial activity of zidebactam. Cefepime-taniborbactam showed excellent activity against recombinant E. coli strains, including metallo-β-lactamase producers, whereas aztreonam-avibactam remained the best therapeutic option against class B β-lactamase-producing P. aeruginosa.
Collapse
Affiliation(s)
- Christophe Le Terrier
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Division of Intensive care unit, University hospitals of Geneva, Geneva, Switzerland
| | - Patrice Nordmann
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland
- University of Lausanne and University hospital Center, Lausanne, Switzerland
| | - Charlotte Freret
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Marion Seigneur
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Laurent POIREL
- Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Swiss National Reference Center for Emerging Antibiotic Resistance, Fribourg, Switzerland
| |
Collapse
|
34
|
Karvouniaris M, Almyroudi MP, Abdul-Aziz MH, Blot S, Paramythiotou E, Tsigou E, Koulenti D. Novel Antimicrobial Agents for Gram-Negative Pathogens. Antibiotics (Basel) 2023; 12:761. [PMID: 37107124 PMCID: PMC10135111 DOI: 10.3390/antibiotics12040761] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Gram-negative bacterial resistance to antimicrobials has had an exponential increase at a global level during the last decades and represent an everyday challenge, especially for the hospital practice of our era. Concerted efforts from the researchers and the industry have recently provided several novel promising antimicrobials, resilient to various bacterial resistance mechanisms. There are new antimicrobials that became commercially available during the last five years, namely, cefiderocol, imipenem-cilastatin-relebactam, eravacycline, omadacycline, and plazomicin. Furthermore, other agents are in advanced development, having reached phase 3 clinical trials, namely, aztreonam-avibactam, cefepime-enmetazobactam, cefepime-taniborbactam, cefepime-zidebactam, sulopenem, tebipenem, and benapenem. In this present review, we critically discuss the characteristics of the above-mentioned antimicrobials, their pharmacokinetic/pharmacodynamic properties and the current clinical data.
Collapse
Affiliation(s)
- Marios Karvouniaris
- Intensive Care Unit, AHEPA University Hospital, 546 36 Thessaloniki, Greece;
| | | | - Mohd Hafiz Abdul-Aziz
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QL 4029, Australia; (M.H.A.-A.); (S.B.)
| | - Stijn Blot
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QL 4029, Australia; (M.H.A.-A.); (S.B.)
- Department of Internal Medicine and Pediatrics, Ghent University, 9000 Ghent, Belgium
| | | | - Evdoxia Tsigou
- Intensive Care Department, ‘Aghioi Anargyroi’ Hospital of Kifissia, 145 64 Athens, Greece;
| | - Despoina Koulenti
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, QL 4029, Australia; (M.H.A.-A.); (S.B.)
- Second Critical Care Department, Attikon University Hospital, 124 62 Athens, Greece;
| |
Collapse
|
35
|
Antimicrobial Treatment of Pseudomonas aeruginosa Severe Sepsis. Antibiotics (Basel) 2022; 11:antibiotics11101432. [PMID: 36290092 PMCID: PMC9598900 DOI: 10.3390/antibiotics11101432] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
Pseudomonas aeruginosa is a pathogen often encountered in a healthcare setting. It has consistently ranked among the most frequent pathogens seen in nosocomial infections, particularly bloodstream and respiratory tract infections. Aside from having intrinsic resistance to many antibiotics, it rapidly acquires resistance to novel agents. Given the high mortality of pseudomonal infections generally, and pseudomonal sepsis particularly, and with the rise of resistant strains, treatment can be very challenging for the clinician. In this paper, we will review the latest evidence for the optimal treatment of P. aeruginosa sepsis caused by susceptible as well as multidrug-resistant strains including the difficult to treat pathogens. We will also discuss the mode of drug infusion, indications for combination therapy, along with the proper dosing and duration of treatment for various conditions with a brief discussion of the use of non-antimicrobial agents.
Collapse
|