1
|
Süssmuth RD, Kulike‐Koczula M, Gao P, Kosol S. Fighting Antimicrobial Resistance: Innovative Drugs in Antibacterial Research. Angew Chem Int Ed Engl 2025; 64:e202414325. [PMID: 39611429 PMCID: PMC11878372 DOI: 10.1002/anie.202414325] [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: 07/29/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/30/2024]
Abstract
In the fight against bacterial infections, particularly those caused by multi-resistant pathogens known as "superbugs", the need for new antibacterials is undoubted in scientific communities and is by now also widely perceived by the general population. However, the antibacterial research landscape has changed considerably over the past years. With few exceptions, the majority of big pharma companies has left the field and thus, the decline in R&D on antibacterials severely impacts the drug pipeline. In recent years, antibacterial research has increasingly relied on smaller companies or academic research institutions, which mostly have only limited financial resources, to carry a drug discovery and development process from the beginning and through to the beginning of clinical phases. This review formulates the requirements for an antibacterial in regard of targeted pathogens, resistance mechanisms and drug discovery. Strategies are shown for the discovery of new antibacterial structures originating from natural sources, by chemical synthesis and more recently from artificial intelligence approaches. This is complemented by principles for the computer-aided design of antibacterials and the refinement of a lead structure. The second part of the article comprises a compilation of antibacterial molecules classified according to bacterial target structures, e.g. cell wall synthesis, protein synthesis, as well as more recently emerging target classes, e.g. fatty acid synthesis, proteases and membrane proteins. Aspects of the origin, the antibacterial spectrum, resistance and the current development status of the presented drug molecules are highlighted.
Collapse
Affiliation(s)
- Roderich D. Süssmuth
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Marcel Kulike‐Koczula
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Peng Gao
- Institut für ChemieTechnische Universität BerlinStrasse des 17. Juni 124, TC210629BerlinGermany
| | - Simone Kosol
- Medical School BerlinDepartment Human MedicineRüdesheimer Strasse 5014195BerlinGermany
| |
Collapse
|
2
|
Heimann D, Kohnhäuser D, Kohnhäuser AJ, Brönstrup M. Antibacterials with Novel Chemical Scaffolds in Clinical Development. Drugs 2025; 85:293-323. [PMID: 39847315 PMCID: PMC11891108 DOI: 10.1007/s40265-024-02137-x] [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: 12/02/2024] [Indexed: 01/24/2025]
Abstract
The rise of antimicrobial resistance represents a significant global health threat, driven by the diminishing efficacy of existing antibiotics, a lack of novel antibacterials entering the market, and an over- or misuse of existing antibiotics, which accelerates the evolution of resistant bacterial strains. This review focuses on innovative therapies by highlighting 19 novel antibacterials in clinical development as of June 2024. These selected compounds are characterized by new chemical scaffolds, novel molecular targets, and/or unique mechanisms of action, which render their potential to break antimicrobial resistance particularly high. A detailed analysis of the scientific foundations behind each of these compounds is provided, including their pharmacodynamic profiles, current development state, and potential for overcoming existing limitations in antibiotic therapy. By presenting this subset of chemically novel antibacterials, the review highlights the ability to innovate in antibiotic drug development to counteract bacterial resistance and improve treatment outcomes.
Collapse
Affiliation(s)
- Dominik Heimann
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | - Daniel Kohnhäuser
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany
| | | | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Braunschweig, Germany.
- Institute of Organic Chemistry and Biomolecular Drug Research Centre (BMWZ), Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany.
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Inhoffenstraße 7, 38124, Braunschweig, Germany.
| |
Collapse
|
3
|
Gedeon A, Yab E, Dinut A, Sadowski E, Capton E, Dreneau A, Petit J, Gioia B, Piveteau C, Djaout K, Lecat E, Wehenkel AM, Gubellini F, Mechaly A, Alzari PM, Deprez B, Baulard A, Aubry A, Willand N, Petrella S. Molecular mechanism of a triazole-containing inhibitor of Mycobacterium tuberculosis DNA gyrase. iScience 2024; 27:110967. [PMID: 39429773 PMCID: PMC11489056 DOI: 10.1016/j.isci.2024.110967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 10/22/2024] Open
Abstract
Antimicrobial resistance remains a persistent and pressing public health concern. Here, we describe the synthesis of original triazole-containing inhibitors targeting the DNA gyrase, a well-validated drug target for developing new antibiotics. Our compounds demonstrate potent antibacterial activity against various pathogenic bacteria, with notable potency against Mycobacterium tuberculosis (Mtb). Moreover, one hit, compound 10a, named BDM71403, was shown to be more potent in Mtb than the NBTI of reference, gepotidacin. Mechanistic enzymology assays reveal a competitive interaction of BDM71403 with fluoroquinolones within the Mtb gyrase cleavage core. High-resolution cryo-electron microscopy structural analysis provides detailed insights into the ternary complex formed by the Mtb gyrase, double-stranded DNA, and either BDM71403 or gepotidacin, providing a rational framework to understand the superior in vitro efficacy on Mtb. This study highlights the potential of triazole-based scaffolds as promising gyrase inhibitors, offering new avenues for drug development in the fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Antoine Gedeon
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Emilie Yab
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Aurelia Dinut
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Elodie Sadowski
- Cimi-Paris, INSERM U1135, Sorbonne Université, AP-HP. Sorbonne Université, Laboratoire de Bactériologie-Hygiène, CNR des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, 75005 Paris, France
| | - Estelle Capton
- Cimi-Paris, INSERM U1135, Sorbonne Université, AP-HP. Sorbonne Université, Laboratoire de Bactériologie-Hygiène, CNR des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, 75005 Paris, France
| | - Aurore Dreneau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Julienne Petit
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Bruna Gioia
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Catherine Piveteau
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Kamel Djaout
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Estelle Lecat
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Anne Marie Wehenkel
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Bacterial Cell Cycle Mechanisms Unit, 75015 Paris, France
| | - Francesca Gubellini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Ariel Mechaly
- Institut Pasteur, Plate-Forme de Cristallographie, CNRS UMR 3528, 75015 Paris, France
| | - Pedro M. Alzari
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
| | - Benoît Deprez
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Alain Baulard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Center for Infection and Immunity of Lille, 59000 Lille, France
| | - Alexandra Aubry
- Cimi-Paris, INSERM U1135, Sorbonne Université, AP-HP. Sorbonne Université, Laboratoire de Bactériologie-Hygiène, CNR des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, 75005 Paris, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177 - Drugs and Molecules for living Systems, 59000 Lille, France
| | - Stéphanie Petrella
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Unité de Microbiologie Structurale, 75015 Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 3528, Bacterial Cell Cycle Mechanisms Unit, 75015 Paris, France
| |
Collapse
|
4
|
Butler MS, Vollmer W, Goodall ECA, Capon RJ, Henderson IR, Blaskovich MAT. A Review of Antibacterial Candidates with New Modes of Action. ACS Infect Dis 2024; 10:3440-3474. [PMID: 39018341 PMCID: PMC11474978 DOI: 10.1021/acsinfecdis.4c00218] [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: 03/17/2024] [Revised: 06/15/2024] [Accepted: 06/17/2024] [Indexed: 07/19/2024]
Abstract
There is a lack of new antibiotics to combat drug-resistant bacterial infections that increasingly threaten global health. The current pipeline of clinical-stage antimicrobials is primarily populated by "new and improved" versions of existing antibiotic classes, supplemented by several novel chemical scaffolds that act on traditional targets. The lack of fresh chemotypes acting on previously unexploited targets (the "holy grail" for new antimicrobials due to their scarcity) is particularly unfortunate as these offer the greatest opportunity for innovative breakthroughs to overcome existing resistance. In recognition of their potential, this review focuses on this subset of high value antibiotics, providing chemical structures where available. This review focuses on candidates that have progressed to clinical trials, as well as selected examples of promising pioneering approaches in advanced stages of development, in order to stimulate additional research aimed at combating drug-resistant infections.
Collapse
Affiliation(s)
- Mark S. Butler
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Waldemar Vollmer
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Emily C. A. Goodall
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Robert J. Capon
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Ian R. Henderson
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| | - Mark A. T. Blaskovich
- Centre
for Superbug Solutions and ARC Training Centre for Environmental and
Agricultural Solutions to Antimicrobial Resistance, Institute for
Molecular Bioscience, The University of
Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
5
|
Hameed P S, Kotakonda H, Sharma S, Nandishaiah R, Katagihallimath N, Rao R, Sadler C, Slater I, Morton M, Chandrasekaran A, Griffen E, Pillai D, Reddy S, Bharatham N, Venkatesan S, Jonnalagadda V, Jayaraman R, Nanjundappa M, Sharma M, Raveendran S, Rajagopal S, Tumma H, Watters A, Becker H, Lindley J, Flamm R, Huband M, Sahm D, Hackel M, Mathur T, Kolamunnage-Dona R, Unsworth J, Mcentee L, Farrington N, Manickam D, Chandrashekara N, Jayachandiran S, Reddy H, Shanker S, Richard V, Thomas T, Nagaraj S, Datta S, Sambandamurthy V, Ramachandran V, Clay R, Tomayko J, Das S, V B. BWC0977, a broad-spectrum antibacterial clinical candidate to treat multidrug resistant infections. Nat Commun 2024; 15:8202. [PMID: 39294149 PMCID: PMC11410943 DOI: 10.1038/s41467-024-52557-2] [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: 12/18/2023] [Accepted: 09/12/2024] [Indexed: 09/20/2024] Open
Abstract
The global crisis of antimicrobial resistance (AMR) necessitates the development of broad-spectrum antibacterial drugs effective against multi-drug resistant (MDR) pathogens. BWC0977, a Novel Bacterial Topoisomerase Inhibitor (NBTI) selectively inhibits bacterial DNA replication via inhibition of DNA gyrase and topoisomerase IV. BWC0977 exhibited a minimum inhibitory concentration (MIC90) of 0.03-2 µg/mL against a global panel of MDR Gram-negative bacteria including Enterobacterales and non-fermenters, Gram-positive bacteria, anaerobes and biothreat pathogens. BWC0977 retains activity against isolates resistant to fluoroquinolones (FQs), carbapenems and colistin and demonstrates efficacy against multiple pathogens in two rodent species with significantly higher drug levels in the epithelial lining fluid of infected lungs. In healthy volunteers, single-ascending doses of BWC0977 administered intravenously ( https://clinicaltrials.gov/study/NCT05088421 ) was found to be safe, well tolerated (primary endpoint) and achieved dose-proportional exposures (secondary endpoint) consistent with modelled data from preclinical studies. Here, we show that BWC0977 has the potential to treat a range of critical-care infections including MDR bacterial pneumonias.
Collapse
Affiliation(s)
- Shahul Hameed P
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Harish Kotakonda
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Sreevalli Sharma
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Radha Nandishaiah
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Nainesh Katagihallimath
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Ranga Rao
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Claire Sadler
- Apconix Ltd. Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | - Ian Slater
- Apconix Ltd. Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | - Michael Morton
- Apconix Ltd. Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | | | - Ed Griffen
- Medchemica Ltd., No. 8162245, Ebenezer House, Newcastle-under-Lyme, Staffordshire, ST5 2BE, England
| | - Dhanashree Pillai
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Sambasiva Reddy
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Nagakumar Bharatham
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Suryanarayanan Venkatesan
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Venugopal Jonnalagadda
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Ramesh Jayaraman
- TheraIndx Lifesciences Pvt. Ltd., Sy No. 27, Deganahalli, Bangalore, 562123, India
| | - Mahesh Nanjundappa
- TheraIndx Lifesciences Pvt. Ltd., Sy No. 27, Deganahalli, Bangalore, 562123, India
| | - Maitrayee Sharma
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Savitha Raveendran
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Sreenath Rajagopal
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Harikrishna Tumma
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Amy Watters
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Holly Becker
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Jill Lindley
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Robert Flamm
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Michael Huband
- JMI Laboratories, 345 Beaver Kreek Center, North Liberty, IA, 52317, USA
| | - Dan Sahm
- IHMA USA, 2122 Palmer Drive, Schaumburg, IL, 60173-3817, USA
| | - Meredith Hackel
- IHMA USA, 2122 Palmer Drive, Schaumburg, IL, 60173-3817, USA
| | | | - Ruwanthi Kolamunnage-Dona
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Jennifer Unsworth
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Laura Mcentee
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Nikki Farrington
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Dhanasekaran Manickam
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Narayana Chandrashekara
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Sivakandan Jayachandiran
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Hrushikesava Reddy
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Sathya Shanker
- Syngene International Ltd., Biocon Park, Plot No. 2 & 3, Bommasandra Jigani Link Road, Bangalore, 560 099, India
| | - Vijay Richard
- Narayana Health, Mazumdar Shaw Medical Center, 258/A, Bommasandra Industrial Area, Hosur Road, Bangalore, 560 099, India
| | - Teby Thomas
- Microbiology laboratory, St. John's Hospital, Sarjapur Road, Bangalore, 560 034, India
| | - Savitha Nagaraj
- Microbiology laboratory, St. John's Hospital, Sarjapur Road, Bangalore, 560 034, India
| | - Santanu Datta
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Vasan Sambandamurthy
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Vasanthi Ramachandran
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Robert Clay
- Highbury Regulatory Science Limited, SK10 4TG, Nether Alderley, Cheshire, SK10 4TG, UK
| | - John Tomayko
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India
| | - Shampa Das
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, L69 7BE, UK
| | - Balasubramanian V
- Bugworks Research India Pvt. Ltd. Center for Cellular & Molecular Platforms, National Center for Biological Sciences, GKVK Campus, Bellary Road, Bangalore, 560 065, India.
| |
Collapse
|
6
|
Boonyalai N, Peerapongpaisarn D, Thamnurak C, Oransathid W, Wongpatcharamongkol N, Oransathid W, Lurchachaiwong W, Griesenbeck JS, Waters NC, Demons ST, Ruamsap N, Vesely BA. Screening of the Pandemic Response Box library identified promising compound candidate drug combinations against extensively drug-resistant Acinetobacter baumannii. Sci Rep 2024; 14:21709. [PMID: 39289446 PMCID: PMC11408719 DOI: 10.1038/s41598-024-72603-9] [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] [Received: 11/27/2023] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
Infections caused by antimicrobial-resistant Acinetobacter baumannii pose a significant threat to human health, particularly in the context of hospital-acquired infections. As existing antibiotics lose efficacy against Acinetobacter isolates, there is an urgent need for the development of novel antimicrobial agents. In this study, we assessed 400 structurally diverse compounds from the Medicines for Malaria Pandemic Response Box for their activity against two clinical isolates of A. baumannii: A. baumannii 5075, known for its extensive drug resistance, and A. baumannii QS17-1084, obtained from an infected wound in a Thai patient. Among the compounds tested, seven from the Pathogen box exhibited inhibitory effects on the in vitro growth of A. baumannii isolates, with IC50s ≤ 48 µM for A. baumannii QS17-1084 and IC50s ≤ 17 µM for A. baumannii 5075. Notably, two of these compounds, MUT056399 and MMV1580854, shared chemical scaffolds resembling triclosan. Further investigations involving drug combinations identified five synergistic drug combinations, suggesting potential avenues for therapeutic development. The combination of MUT056399 and brilacidin against A. baumannii QS17-1084 and that of MUT056399 and eravacycline against A. baumannii 5075 showed bactericidal activity. These combinations significantly inhibited biofilm formation produced by both A. baumannii strains. Our findings highlight the drug combinations as promising candidates for further evaluation in murine wound infection models against multidrug-resistant A. baumannii. These compounds hold potential for addressing the critical need for effective antibiotics in the face of rising antimicrobial resistance.
Collapse
Affiliation(s)
- Nonlawat Boonyalai
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
- Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Dutsadee Peerapongpaisarn
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Chatchadaporn Thamnurak
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Wilawan Oransathid
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Nantanat Wongpatcharamongkol
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Wirote Oransathid
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Woradee Lurchachaiwong
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
- Division of Global Health Protection, Thailand MoPH-US CDC Collaboration, Nonthaburi, Thailand
| | - John S Griesenbeck
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Norman C Waters
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Samandra T Demons
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Nattaya Ruamsap
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| | - Brian A Vesely
- Department of Bacterial and Parasitic Diseases, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand.
| |
Collapse
|
7
|
Collins J, Basarab GS, Chibale K, Osheroff N. Interactions between Zoliflodacin and Neisseria gonorrhoeae Gyrase and Topoisomerase IV: Enzymological Basis for Cellular Targeting. ACS Infect Dis 2024; 10:3071-3082. [PMID: 39082980 PMCID: PMC11320581 DOI: 10.1021/acsinfecdis.4c00438] [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: 05/28/2024] [Revised: 07/09/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Gyrase and topoisomerase IV are the cellular targets for fluoroquinolones, a critically important class of antibacterial agents used to treat a broad spectrum of human infections. Unfortunately, the clinical efficacy of the fluoroquinolones has been curtailed by the emergence of target-mediated resistance. This is especially true for Neisseria gonorrhoeae, the causative pathogen of the sexually transmitted infection gonorrhea. Spiropyrimidinetriones (SPTs), a new class of antibacterials, were developed to combat the growing antibacterial resistance crisis. Zoliflodacin is the most clinically advanced SPT and displays efficacy against uncomplicated urogenital gonorrhea in human trials. Like fluoroquinolones, the primary target of zoliflodacin in N. gonorrhoeae is gyrase, and topoisomerase IV is a secondary target. Because unbalanced gyrase/topoisomerase IV targeting has facilitated the evolution of fluoroquinolone-resistant bacteria, it is important to understand the underlying basis for the differential targeting of zoliflodacin in N. gonorrhoeae. Therefore, we assessed the effects of this SPT on the catalytic and DNA cleavage activities of N. gonorrhoeae gyrase and topoisomerase IV. In all reactions examined, zoliflodacin displayed higher potency against gyrase than topoisomerase IV. Moreover, zoliflodacin generated more DNA cleavage and formed more stable enzyme-cleaved DNA-SPT complexes with gyrase. The SPT also maintained higher activity against fluoroquinolone-resistant gyrase than topoisomerase IV. Finally, when compared to zoliflodacin, the novel SPT H3D-005722 induced more balanced double-stranded DNA cleavage with gyrase and topoisomerase IV from N. gonorrhoeae, Escherichia coli, and Bacillus anthracis. This finding suggests that further development of the SPT class could yield compounds with a more balanced targeting against clinically important bacterial infections.
Collapse
Affiliation(s)
- Jessica
A. Collins
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Gregory S. Basarab
- Holistic
Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Holistic
Drug Discovery and Development (H3D) Centre, and South African Medical
Research Council Drug Discovery and Development Research Unit, Department
of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Neil Osheroff
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Medicine (Hematology/Oncology), Vanderbilt
University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
8
|
Brown-Elliott BA, Bush G, Hughes MD, Rodriguez E, Weikel CA, Min SB, Wallace RJ. In vitro activity of gepotidacin and comparator antimicrobials against isolates of nontuberculous mycobacteria (NTM). Antimicrob Agents Chemother 2024; 68:e0168423. [PMID: 38656138 PMCID: PMC11620510 DOI: 10.1128/aac.01684-23] [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] [Received: 12/19/2023] [Accepted: 03/31/2024] [Indexed: 04/26/2024] Open
Abstract
Novel antimicrobials are needed to treat rising nontuberculous mycobacteria (NTM) infections. Using standard broth microdilution methods, 68 NTM isolates were tested against gepotidacin, a new, first-in-class, oral triazaacenaphthylene bacterial topoisomerase inhibitor. MICs varied (0.25 to >64 µg/mL) with the lowest being M. fortuitum complex (0.25-8 µg/mL), M. mucogenicum complex (1-2 µg/mL), M. kansasii (0.25-8 µg/mL), and M. marinum (4-16 µg/mL). Testing greater numbers of some species is suggested to better understand gepotidacin activity against NTM.
Collapse
Affiliation(s)
- Barbara A. Brown-Elliott
- The University of Texas Health Science Center at Tyler, Mycobacteria/Nocardia Laboratory, The University of Texas at Tyler School of Medicine, Tyler, Texas, USA
| | - Georgie Bush
- The University of Texas Health Science Center at Tyler, Mycobacteria/Nocardia Laboratory, The University of Texas at Tyler School of Medicine, Tyler, Texas, USA
| | - M. Dolores Hughes
- The University of Texas Health Science Center at Tyler, Mycobacteria/Nocardia Laboratory, The University of Texas at Tyler School of Medicine, Tyler, Texas, USA
| | - Eliana Rodriguez
- The University of Texas Health Science Center at Tyler, Mycobacteria/Nocardia Laboratory, The University of Texas at Tyler School of Medicine, Tyler, Texas, USA
| | - Chase A. Weikel
- Department of Infectious Diseases, GSK, Collegeville, Pennsylvania, USA
| | - Sharon B. Min
- Department of Infectious Diseases, GSK, Collegeville, Pennsylvania, USA
| | - Richard J. Wallace
- The University of Texas Health Science Center at Tyler, Mycobacteria/Nocardia Laboratory, The University of Texas at Tyler School of Medicine, Tyler, Texas, USA
| |
Collapse
|
9
|
Collins J, Osheroff N. Gyrase and Topoisomerase IV: Recycling Old Targets for New Antibacterials to Combat Fluoroquinolone Resistance. ACS Infect Dis 2024; 10:1097-1115. [PMID: 38564341 PMCID: PMC11019561 DOI: 10.1021/acsinfecdis.4c00128] [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: 02/16/2024] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/04/2024]
Abstract
Beyond their requisite functions in many critical DNA processes, the bacterial type II topoisomerases, gyrase and topoisomerase IV, are the targets of fluoroquinolone antibacterials. These drugs act by stabilizing gyrase/topoisomerase IV-generated DNA strand breaks and by robbing the cell of the catalytic activities of these essential enzymes. Since their clinical approval in the mid-1980s, fluoroquinolones have been used to treat a broad spectrum of infectious diseases and are listed among the five "highest priority" critically important antimicrobial classes by the World Health Organization. Unfortunately, the widespread use of fluoroquinolones has been accompanied by a rise in target-mediated resistance caused by specific mutations in gyrase and topoisomerase IV, which has curtailed the medical efficacy of this drug class. As a result, efforts are underway to identify novel antibacterials that target the bacterial type II topoisomerases. Several new classes of gyrase/topoisomerase IV-targeted antibacterials have emerged, including novel bacterial topoisomerase inhibitors, Mycobacterium tuberculosis gyrase inhibitors, triazaacenaphthylenes, spiropyrimidinetriones, and thiophenes. Phase III clinical trials that utilized two members of these classes, gepotidacin (triazaacenaphthylene) and zoliflodacin (spiropyrimidinetrione), have been completed with positive outcomes, underscoring the potential of these compounds to become the first new classes of antibacterials introduced into the clinic in decades. Because gyrase and topoisomerase IV are validated targets for established and emerging antibacterials, this review will describe the catalytic mechanism and cellular activities of the bacterial type II topoisomerases, their interactions with fluoroquinolones, the mechanism of target-mediated fluoroquinolone resistance, and the actions of novel antibacterials against wild-type and fluoroquinolone-resistant gyrase and topoisomerase IV.
Collapse
Affiliation(s)
- Jessica
A. Collins
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Neil Osheroff
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Medicine (Hematology/Oncology), Vanderbilt
University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
10
|
Hilliard JJ, Jakielaszek C, Mannino F, Hossain M, Qian L, Fishman C, Demons S, Hershfield J, Soffler C, Russo R, Henning L, Novak J, O'Dwyer K. Efficacy of therapeutically administered gepotidacin in a rabbit model of inhalational anthrax. Antimicrob Agents Chemother 2024; 68:e0149723. [PMID: 38358266 PMCID: PMC10916377 DOI: 10.1128/aac.01497-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/17/2024] [Indexed: 02/16/2024] Open
Abstract
Bacillus anthracis is a Gram-positive Centers for Disease Control and Prevention category "A" biothreat pathogen. Without early treatment, inhalation of anthrax spores with progression to inhalational anthrax disease is associated with high fatality rates. Gepotidacin is a novel first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action and is being evaluated for use against biothreat and conventional pathogens. Gepotidacin selectively inhibits bacterial DNA replication via a unique binding mode and has in vitro activity against a collection of B. anthracis isolates including antibacterial-resistant strains, with the MIC90 ranging from 0.5 to 1 µg/mL. In vivo activity of gepotidacin was also evaluated in the New Zealand White rabbit model of inhalational anthrax. The primary endpoint was survival, with survival duration and bacterial clearance as secondary endpoints. The trigger for treatment was the presence of anthrax protective antigen in serum. New Zealand White rabbits were dosed intravenously for 5 days with saline or gepotidacin at 114 mg/kg/d to simulate a dosing regimen of 1,000 mg intravenous (i.v.) three times a day (TID) in humans. Gepotidacin provided a survival benefit compared to saline control, with 91% survival (P-value: 0.0001). All control animals succumbed to anthrax and were found to be blood- and organ culture-positive for B. anthracis. The novel mode of action, in vitro microbiology, preclinical safety, and animal model efficacy data, which were generated in line with Food and Drug Administration Animal Rule, support gepotidacin as a potential treatment for anthrax in an emergency biothreat situation.
Collapse
Affiliation(s)
| | | | | | | | - Lian Qian
- GSK, Collegeville, Pennsylvania, USA
| | | | - Samandra Demons
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Jeremy Hershfield
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Carl Soffler
- US Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Riccardo Russo
- Rutgers University School of Medicine, Newark, New Jersey, USA
| | - Lisa Henning
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | - Joseph Novak
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | | |
Collapse
|
11
|
Arends SJR, Butler D, Scangarella-Oman N, Castanheira M, Mendes R. Intermethod comparability analyses of gepotidacin antimicrobial susceptibility tests using a large collection of globally collected Escherichia coli and Staphylococcus saprophyticus clinical isolates. Diagn Microbiol Infect Dis 2024; 108:116181. [PMID: 38215519 DOI: 10.1016/j.diagmicrobio.2024.116181] [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: 12/09/2022] [Revised: 08/22/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024]
Abstract
Gepotidacin (GSK2140944) is a novel, bactericidal, first in class triazaacenaphthylene bacterial type II topoisomerase inhibitor in development for the treatment of uncomplicated urinary tract infections and gonorrhea. The performance of several antimicrobial susceptibility methods (broth microdilution, gradient diffusion, and disk diffusion) for gepotidacin were evaluated using over 5800 recent Escherichia coli and Staphylococcus saprophyticus clinical isolates. Reference broth microdilution gepotidacin MICs showed an essential agreement of 95.9 % and 98.1 % with MICs by gradient diffusion for E. coli and S. saprophyticus isolates, respectively. Gepotidacin susceptibility using disks produced by 2 manufacturers had good agreement with an R2 values of 0.95 and 99.2 % of overall zone diameters agreeing within 3 mm. A correlation with an overall R2 value of 0.72 between MICs by broth microdilution and zone diameters by disk diffusion was observed. This data should assist in the clinical development of gepotidacin and provide reliable susceptibility methods to evaluate its activity.
Collapse
Affiliation(s)
- S J Ryan Arends
- JMI Laboratories, 345 Beaver Kreek Centre, Suite A, North Liberty, IA 52317, United States.
| | | | | | - Mariana Castanheira
- JMI Laboratories, 345 Beaver Kreek Centre, Suite A, North Liberty, IA 52317, United States
| | - Rodrigo Mendes
- JMI Laboratories, 345 Beaver Kreek Centre, Suite A, North Liberty, IA 52317, United States
| |
Collapse
|
12
|
Wagenlehner F, Perry CR, Hooton TM, Scangarella-Oman NE, Millns H, Powell M, Jarvis E, Dennison J, Sheets A, Butler D, Breton J, Janmohamed S. Oral gepotidacin versus nitrofurantoin in patients with uncomplicated urinary tract infection (EAGLE-2 and EAGLE-3): two randomised, controlled, double-blind, double-dummy, phase 3, non-inferiority trials. Lancet 2024; 403:741-755. [PMID: 38342126 DOI: 10.1016/s0140-6736(23)02196-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/14/2023] [Accepted: 09/27/2023] [Indexed: 02/13/2024]
Abstract
BACKGROUND Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action and a unique binding site, providing well balanced inhibition of two type II topoisomerase enzymes. Oral gepotidacin is under investigation to treat uncomplicated urinary tract infections. We aimed to compare the efficacy and safety of oral gepotidacin with that of nitrofurantoin in adolescent and adult female individuals with uncomplicated urinary tract infections. METHODS EAGLE-2 and EAGLE-3 were phase 3, randomised, multicentre, double-blind, double-dummy, non-inferiority (10% margin) trials, in which patients were enrolled at 219 centres worldwide. Patients assigned female at birth, non-pregnant, aged 12 years or older, weighing 40 kg or more, with two or more symptoms of dysuria, frequency, urgency, or lower abdominal pain, and with evidence of urinary nitrite, pyuria, or both were eligible for inclusion. Patients were randomly assigned (1:1) centrally by interactive response technology to receive oral gepotidacin (1500 mg twice daily for 5 days) or oral nitrofurantoin (100 mg twice daily for 5 days), with randomisation stratified by age category and history of recurrent uncomplicated urinary tract infections. Patients, investigators, and the sponsor study team were masked to treatment assignment. The primary endpoint, therapeutic response (success or failure) at test-of-cure (ie, day 10-13), was evaluated in randomly assigned patients with nitrofurantoin-susceptible qualifying uropathogens (≥105 colony-forming units [CFU] per mL) and who received at least one dose of study treatment. Conforming to regulatory guidance, therapeutic success was defined as combined clinical success (ie, complete symptom resolution) and microbiological success (ie, reduction of qualifying uropathogens to <103 CFU/mL) without other systemic antimicrobial use. Safety analyses included patients who were randomly assigned and who received at least one dose of study treatment. The trials are registered with ClinicalTrials.gov, NCT04020341 (EAGLE-2) and NCT04187144 (EAGLE-3), and are completed. FINDINGS Studies were undertaken from Oct 17, 2019, to Nov 30, 2022 (EAGLE-2), and from April 23, 2020, to Dec 1, 2022 (EAGLE-3). 1680 patients in EAGLE-2 and 1731 patients in EAGLE-3 were screened for eligibility, of whom 1531 and 1605 were randomly assigned, respectively (767 in the gepotidacin group and 764 in the nitrofurantoin group in EAGLE-2, and 805 in the gepotidacin group and 800 in the nitrofurantoin group in EAGLE-3). After an interim analysis, which was prospectively agreed as a protocol amendment, both studies were stopped for efficacy. Thus, the primary analysis population included only patients who, at the time of the interim analysis data cutoff, had the opportunity to reach the test-of-cure visit or were known to not have attained therapeutic success before the test-of-cure visit. In EAGLE-2, 162 (50·6%) of 320 patients assigned gepotidacin and 135 (47·0%) of 287 patients assigned nitrofurantoin had therapeutic success (adjusted difference 4·3%, 95% CI -3·6 to 12·1). In EAGLE-3, 162 (58·5%) of 277 patients assigned gepotidacin and 115 (43·6%) of 264 patients assigned nitrofurantoin had therapeutic success (adjusted difference 14·6%, 95% CI 6·4 to 22·8). Gepotidacin was non-inferior to nitrofurantoin in both studies and superior to nitrofurantoin in EAGLE-3. The most common adverse event with gepotidacin was diarrhoea (observed in 111 [14%] of 766 patients in EAGLE-2 and in 147 [18%] of 804 patients in EAGLE-3), whereas the most common adverse event with nitrofurantoin was nausea (in 29 [4%] of 760 patients in EAGLE-2 and in 35 [4%] of 798 patients in EAGLE-3). Cases were mostly mild or moderate. No life-threatening or fatal events occurred. INTERPRETATION Gepotidacin is an efficacious oral antibiotic with acceptable safety and tolerability profiles. As a first-in-class investigational oral antibiotic with activity against common uropathogens, including clinically important drug-resistant phenotypes, gepotidacin has the potential to offer substantial benefit to patients. FUNDING GSK and the US Office of the Assistant Secretary for Preparedness and Response, Biomedical Advanced Research and Development Authority.
Collapse
Affiliation(s)
- Florian Wagenlehner
- Clinic for Urology, Pediatric Urology and Andrology, Justus-Liebig-University, Giessen, Germany.
| | | | - Thomas M Hooton
- Miller School of Medicine, Medical Campus, University of Miami, Miami, FL, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Lim JS, Chai YY, Ser WX, Haeren AV, Lim YH, Raja T, Foo JB, Hamzah S, Sellappans R, Yow HY. Novel drug candidates against antibiotic-resistant microorganisms: A review. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:134-150. [PMID: 38234674 PMCID: PMC10790292 DOI: 10.22038/ijbms.2023.71672.15593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/12/2023] [Indexed: 01/19/2024]
Abstract
Antibiotic resistance is fast spreading globally, leading to treatment failures and adverse clinical outcomes. This review focuses on the resistance mechanisms of the top five threatening pathogens identified by the World Health Organization's global priority pathogens list: carbapenem-resistant Acinetobacter baumannii, carbapenem-resistant Pseudomonas aeruginosa, carbapenem-resistant, extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae, vancomycin-resistant Enterococcus faecium and methicillin, vancomycin-resistant Staphylococcus aureus. Several novel drug candidates have shown promising results from in vitro and in vivo studies, as well as clinical trials. The novel drugs against carbapenem-resistant bacteria include LCB10-0200, apramycin, and eravacycline, while for Enterobacteriaceae, the drug candidates are LysSAP-26, DDS-04, SPR-206, nitroxoline, cefiderocol, and plazomicin. TNP-209, KBP-7072, and CRS3123 are agents against E. faecium, while Debio 1450, gepotidacin, delafloxacin, and dalbavancin are drugs against antibiotic-resistant S. aureus. In addition to these identified drug candidates, continued in vitro and in vivo studies are required to investigate small molecules with potential antibacterial effects screened by computational receptor docking. As drug discovery progresses, preclinical and clinical studies should also be extensively conducted on the currently available therapeutic agents to unravel their potential antibacterial effect and spectrum of activity, as well as safety and efficacy profiles.
Collapse
Affiliation(s)
- Jing-Sheng Lim
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Yoke-Yen Chai
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Wei-Xin Ser
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Aniqah Van Haeren
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Yan-Hong Lim
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Tarshiiny Raja
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Jhi-Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
- Medical Advancement for Better Quality of Life Impact Lab, Taylor’s University, 47500 Selangor, Malaysia
| | - Sharina Hamzah
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
- Medical Advancement for Better Quality of Life Impact Lab, Taylor’s University, 47500 Selangor, Malaysia
| | - Renukha Sellappans
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
| | - Hui Yin Yow
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya, Malaysia
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
14
|
Perry CR, Scangarella-Oman NE, Millns H, Flight W, Gatsi S, Jakielaszek C, Janmohamed S, Lewis DA. Efficacy and Safety of Gepotidacin as Treatment of Uncomplicated Urogenital Gonorrhea (EAGLE-1): Design of a Randomized, Comparator-Controlled, Phase 3 Study. Infect Dis Ther 2023; 12:2307-2320. [PMID: 37751016 PMCID: PMC10581980 DOI: 10.1007/s40121-023-00862-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/18/2023] [Indexed: 09/27/2023] Open
Abstract
INTRODUCTION Gonorrhea, caused by Neisseria gonorrhoeae (NG), is the second most common bacterial sexually transmitted infection (STI). Rates of antimicrobial resistance to standard care are increasing worldwide, with many antibiotic classes now ineffective against NG. Gepotidacin is a first-in-class, bactericidal, triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by inhibition of two enzymes, where a single target-specific mutation does not significantly impact susceptibility. Gepotidacin confers activity against NG, including most strains resistant to marketed antibiotics. Here, we describe the design of a phase 3 clinical trial (EAGLE-1; NCT04010539) evaluating gepotidacin for the treatment of uncomplicated urogenital gonorrhea. METHODS This phase 3, randomized, multicenter, sponsor-blinded, noninferiority study across six countries is comparing the efficacy of gepotidacin with ceftriaxone plus azithromycin in 400 patients with uncomplicated urogenital gonorrhea (microbiological intent-to-treat population) and assessing the safety of gepotidacin in approximately 600 patients (intent-to-treat population). Eligible participants 12 years of age or older with clinical suspicion of urogenital gonococcal infection and a NG-positive urogenital sample and/or purulent discharge are randomized 1:1 to receive oral gepotidacin (2 × 3000 mg 10-12 h apart) or ceftriaxone (500 mg, intramuscular) plus azithromycin (1 g, oral). The primary endpoint is culture-confirmed bacterial eradication of NG from the urogenital site at the test-of-cure (days 4-8) visit. PLANNED OUTCOMES This trial was designed in accordance with US Food and Drug Administration (2015) and European Medicines Agency (2011) guidance, particularly the primary endpoint and microbiological evaluability requirements. This study will help characterize the risk-benefit profile of gepotidacin for treating uncomplicated urogenital gonorrhea. Gepotidacin is an important potential treatment for gonorrhea to help address the urgent unmet need of multidrug resistance and the increasingly limited number of oral treatment options. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT04010539.
Collapse
Affiliation(s)
- Caroline R Perry
- GSK, 1250 S. Collegeville Road, Collegeville, PA, 19486-0989, USA.
| | | | | | | | - Sally Gatsi
- GSK, 1250 S. Collegeville Road, Collegeville, PA, 19486-0989, USA
| | | | | | - David A Lewis
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Western Sydney Sexual Health Centre, Parramatta, NSW, Australia
| |
Collapse
|
15
|
Butler MS, Henderson IR, Capon RJ, Blaskovich MAT. Antibiotics in the clinical pipeline as of December 2022. J Antibiot (Tokyo) 2023; 76:431-473. [PMID: 37291465 PMCID: PMC10248350 DOI: 10.1038/s41429-023-00629-8] [Citation(s) in RCA: 101] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 06/10/2023]
Abstract
The need for new antibacterial drugs to treat the increasing global prevalence of drug-resistant bacterial infections has clearly attracted global attention, with a range of existing and upcoming funding, policy, and legislative initiatives designed to revive antibacterial R&D. It is essential to assess whether these programs are having any real-world impact and this review continues our systematic analyses that began in 2011. Direct-acting antibacterials (47), non-traditional small molecule antibacterials (5), and β-lactam/β-lactamase inhibitor combinations (10) under clinical development as of December 2022 are described, as are the three antibacterial drugs launched since 2020. Encouragingly, the increased number of early-stage clinical candidates observed in the 2019 review increased in 2022, although the number of first-time drug approvals from 2020 to 2022 was disappointingly low. It will be critical to monitor how many Phase-I and -II candidates move into Phase-III and beyond in the next few years. There was also an enhanced presence of novel antibacterial pharmacophores in early-stage trials, and at least 18 of the 26 phase-I candidates were targeted to treat Gram-negative bacteria infections. Despite the promising early-stage antibacterial pipeline, it is essential to maintain funding for antibacterial R&D and to ensure that plans to address late-stage pipeline issues succeed.
Collapse
Affiliation(s)
- Mark S Butler
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia.
| | - Ian R Henderson
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia
| | - Robert J Capon
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, 4072, Australia.
| |
Collapse
|
16
|
Whelan AO, Cooper I, Ooi N, Orr D, Blades K, Kirkham J, Lyons A, Barnes KB, Richards MI, Salisbury AM, Craighead M, Harding SV. In Vitro Activity of Novel Topoisomerase Inhibitors against Francisella tularensis and Burkholderia pseudomallei. Antibiotics (Basel) 2023; 12:983. [PMID: 37370302 DOI: 10.3390/antibiotics12060983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/03/2023] [Accepted: 05/25/2023] [Indexed: 06/29/2023] Open
Abstract
Antimicrobial resistance is a global issue, and the investigation of alternative therapies that are not traditional antibiotics are warranted. Novel bacterial type II topoisomerase inhibitors (NBTIs) have recently emerged as a novel class of antibiotics with reduced potential for cross-resistance to fluoroquinolones due to their novel mechanism of action. This study investigated the in vitro activity of a series of cyclohexyl-oxazolidinone bacterial topoisomerase inhibitors against type strains of Francisella tularensis and Burkholderia pseudomallei. Broth microdilution, time-kill, and cell infection assays were performed to determine activity against these biothreat pathogens. Two candidates were identified that demonstrated in vitro activity in multiple assays that in some instances was equivalent to ciprofloxacin and doxycycline. These data warrant the further evaluation of these novel NBTIs and future iterations in vitro and in vivo.
Collapse
Affiliation(s)
- Adam O Whelan
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
| | - Ian Cooper
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - Nicola Ooi
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - David Orr
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - Kevin Blades
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - James Kirkham
- Infex Therapeutics Ltd., Mereside, Alderley Park, Macclesfield SK10 4TG, UK
| | - Amanda Lyons
- Redx Anti-Infectives Ltd., Alderley Park, Macclesfield SK10 4TG, UK
| | - Kay B Barnes
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
| | - Mark I Richards
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
| | | | - Mark Craighead
- Redx Anti-Infectives Ltd., Alderley Park, Macclesfield SK10 4TG, UK
| | - Sarah V Harding
- Defence Science and Technology Laboratory, Porton Down, Salisbury SP4 0JQ, UK
- School of Respiratory Sciences, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
17
|
Jakielaszek C, Hilliard JJ, Mannino F, Hossain M, Qian L, Fishman C, Chou YL, Henning L, Novak J, Demons S, Hershfield J, O’Dwyer K. Efficacy of Intravenously Administered Gepotidacin in Cynomolgus Macaques following a Francisella tularensis Inhalational Challenge. Antimicrob Agents Chemother 2023; 67:e0138122. [PMID: 37097147 PMCID: PMC10190672 DOI: 10.1128/aac.01381-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/18/2023] [Indexed: 04/26/2023] Open
Abstract
Francisella tularensis (F. tularensis) is a Centers for Disease Control (CDC) category "A" Gram-negative biothreat pathogen. Inhalation of F. tularensis can cause pneumonia and respiratory failure and is associated with high mortality rates without early treatment. Gepotidacin is a novel, first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action. Gepotidacin selectively inhibits bacterial DNA replication via a unique binding mode, has activity against multidrug-resistant target pathogens, and has demonstrated in vitro activity against diverse collections of F. tularensis isolates (MIC90 of 0.5 to 1 μg/mL). Gepotidacin was evaluated in the cynomolgus macaque model of inhalational tularemia, using the SCHU S4 strain, with treatment initiated after exposure and sustained fever. Macaques were dosed via intravenous (i.v.) infusion with saline or gepotidacin at 72 mg/kg/day to support a human i.v. infusion dosing regimen of 1,000 mg three times daily. The primary study endpoint was survival, with survival duration and bacterial clearance as secondary endpoints. Gepotidacin treatment resulted in 100% survival compared to 12.5% in the saline-treated control group (P < 0.0001) at Day 43 postinhalational challenge. All gepotidacin-treated animals were blood and organ culture negative for F. tularensis at the end of the study. In contrast, none of the saline control animals were blood and organ culture negative. Gepotoidacin's novel mechanism of action and the efficacy data reported here (aligned with the Food and Drug Administration Animal Rule) support gepotidacin as a potential treatment for pneumonic tularemia in an emergency biothreat situation.
Collapse
Affiliation(s)
| | | | - Frank Mannino
- GSK Pharmaceuticals, Collegeville, Pennsylvania, USA
| | | | - Lian Qian
- GSK Pharmaceuticals, Collegeville, Pennsylvania, USA
| | - Cindy Fishman
- GSK Pharmaceuticals, Collegeville, Pennsylvania, USA
| | - Ying-Liang Chou
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | - Lisa Henning
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | - Joseph Novak
- Battelle Biomedical Research Center (BBRC), Columbus, Ohio, USA
| | - Samandra Demons
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Jeremy Hershfield
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA
| | - Karen O’Dwyer
- GSK Pharmaceuticals, Collegeville, Pennsylvania, USA
| |
Collapse
|
18
|
Ruggieri F, Compagne N, Antraygues K, Eveque M, Flipo M, Willand N. Antibiotics with novel mode of action as new weapons to fight antimicrobial resistance. Eur J Med Chem 2023; 256:115413. [PMID: 37150058 DOI: 10.1016/j.ejmech.2023.115413] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/09/2023] [Accepted: 04/22/2023] [Indexed: 05/09/2023]
Abstract
Antimicrobial resistance (AMR) is a major public health issue, causing 5 million deaths per year. Without any action plan, AMR will be in a near future the leading cause of death ahead of cancer. AMR comes from the ability of bacteria to rapidly develop and share resistance mechanisms towards current antibiotics, rendering them less effective. To circumvent this issue and avoid the phenomenon of cross-resistance, new antibiotics acting on novel targets or with new modes of action are required. Today, the pipeline of potential new treatments with these characteristics includes promising compounds such as gepotidacin, zoliflodacin, ibezapolstat, MGB-BP-3, CRS-3123, afabicin and TXA-709, which are currently in clinical trials, and lefamulin, which has been recently approved by FDA and EMA. In this review, we report the chemical synthesis, mode of action, structure-activity relationships, in vitro and in vivo activities as well as clinical data of these eight small molecules listed above.
Collapse
Affiliation(s)
- Francesca Ruggieri
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Nina Compagne
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Kevin Antraygues
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Maxime Eveque
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Marion Flipo
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France
| | - Nicolas Willand
- Univ. Lille, Inserm, Institut Pasteur de Lille, U1177-Drugs and Molecules for Living Systems, F-59000, Lille, France.
| |
Collapse
|
19
|
Arends SJR, Butler D, Scangarella-Oman N, Castanheira M, Mendes RE. Antimicrobial Activity of Gepotidacin Tested against Escherichia coli and Staphylococcus saprophyticus Isolates Causing Urinary Tract Infections in Medical Centers Worldwide (2019 to 2020). Antimicrob Agents Chemother 2023; 67:e0152522. [PMID: 36877017 PMCID: PMC10112209 DOI: 10.1128/aac.01525-22] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 02/02/2023] [Indexed: 03/07/2023] Open
Abstract
The in vitro activities of gepotidacin and comparator agents against 3,560 Escherichia coli and 344 Staphylococcus saprophyticus collected from female (81.1%) and male (18.9%) patients with urinary tract infections (UTIs) in a global prospective surveillance program in 2019 to 2020 were determined. Isolates collected from 92 medical centers in 25 countries, including the United States, Europe, Latin America, and Japan, were tested for susceptibility by reference methods in a central monitoring laboratory. Gepotidacin inhibited 98.0% (3,488/3,560 isolates) of E. coli and 100% (344/344 isolates) of S. saprophyticus at gepotidacin concentrations of ≤4 μg/mL and ≤0.25 μg/mL, respectively. This activity was largely unaffected with isolates that demonstrated resistance phenotypes to other oral standard-of-care antibiotics, including amoxicillin-clavulanic acid, cephalosporins, fluoroquinolones, fosfomycin, nitrofurantoin, and trimethoprim-sulfamethoxazole. Gepotidacin also inhibited 94.3% (581/616 isolates) of E. coli isolates with an extended-spectrum β-lactamase-producing phenotype, 97.2% (1,085/1,129 isolates) of E. coli isolates resistant to ciprofloxacin, 96.1% (874/899) of E. coli isolates resistant to trimethoprim-sulfamethoxazole, and 96.3% (235/244 isolates) of multidrug-resistant E. coli isolates at gepotidacin concentrations of ≤4 μg/mL. In summary, gepotidacin demonstrated potent activity against a large collection of contemporary UTI E. coli and S. saprophyticus strains collected from patients worldwide. These data support the further clinical development of gepotidacin as a potential treatment option for patients with uncomplicated UTIs.
Collapse
|
20
|
Khalid K, Rox K. All Roads Lead to Rome: Enhancing the Probability of Target Attainment with Different Pharmacokinetic/Pharmacodynamic Modelling Approaches. Antibiotics (Basel) 2023; 12:antibiotics12040690. [PMID: 37107052 PMCID: PMC10135278 DOI: 10.3390/antibiotics12040690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
In light of rising antimicrobial resistance and a decreasing number of antibiotics with novel modes of action, it is of utmost importance to accelerate development of novel treatment options. One aspect of acceleration is to understand pharmacokinetics (PK) and pharmacodynamics (PD) of drugs and to assess the probability of target attainment (PTA). Several in vitro and in vivo methods are deployed to determine these parameters, such as time-kill-curves, hollow-fiber infection models or animal models. However, to date the use of in silico methods to predict PK/PD and PTA is increasing. Since there is not just one way to perform the in silico analysis, we embarked on reviewing for which indications and how PK and PK/PD models as well as PTA analysis has been used to contribute to the understanding of the PK and PD of a drug. Therefore, we examined four recent examples in more detail, namely ceftazidime-avibactam, omadacycline, gepotidacin and zoliflodacin as well as cefiderocol. Whereas the first two compound classes mainly relied on the ‘classical’ development path and PK/PD was only deployed after approval, cefiderocol highly profited from in silico techniques that led to its approval. Finally, this review shall highlight current developments and possibilities to accelerate drug development, especially for anti-infectives.
Collapse
Affiliation(s)
- Kashaf Khalid
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Katharina Rox
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Inhoffenstraße 7, 38124 Braunschweig, Germany
- German Center for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany
| |
Collapse
|
21
|
Koeth LM, DiFranco-Fisher JM, Scangarella-Oman NE. Analysis of the effect of urine on the in vitro activity of gepotidacin and levofloxacin against Escherichia coli, Staphylococcus epidermidis and Staphylococcus saprophyticus. Diagn Microbiol Infect Dis 2023; 106:115946. [DOI: 10.1016/j.diagmicrobio.2023.115946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 11/05/2022] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
|
22
|
Bhattacharjee R, Negi A, Bhattacharya B, Dey T, Mitra P, Preetam S, Kumar L, Kar S, Das SS, Iqbal D, Kamal M, Alghofaili F, Malik S, Dey A, Jha SK, Ojha S, Paiva-Santos AC, Kesari KK, Jha NK. Nanotheranostics to Target Antibiotic-resistant Bacteria: Strategies and Applications. OPENNANO 2023. [DOI: 10.1016/j.onano.2023.100138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
|
23
|
Barth A, Perry CR, Shabbir S, Zamek-Gliszczynski MJ, Thomas S, Dumont EF, Brimhall DB, Nguyen D, Srinivasan M, Swift B. Clinical assessment of gepotidacin (GSK2140944) as a victim and perpetrator of drug-drug interactions via CYP3A metabolism and transporters. Clin Transl Sci 2023; 16:647-661. [PMID: 36642822 PMCID: PMC10087077 DOI: 10.1111/cts.13477] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/14/2022] [Accepted: 12/16/2022] [Indexed: 01/17/2023] Open
Abstract
Gepotidacin is a novel triazaacenaphthylene antibiotic in phase III development. Based on nonclinical in vitro characterization of gepotidacin metabolism, two phase I studies were conducted in healthy participants to investigate clinical drug-drug interactions (DDIs). We assessed gepotidacin as a DDI victim with a potent cytochrome P450 (CYP) 3A4/P-glycoprotein (P-gp) inhibitor (itraconazole), potent CYP3A4 inducer (rifampicin), and nonspecific organic cation transporter (OCT)/multidrug and toxic extrusion transporter (MATE) renal transport inhibitor (cimetidine) via single doses of gepotidacin before and after co-administration with multiple doses of the modulator drugs. Gepotidacin DDI perpetrator potential for P-gp inhibition (digoxin) and CYP3A4 inhibition (midazolam) was evaluated via single doses of the two-drug cocktail without and with gepotidacin. The DDI magnitudes were interpreted based on area under the concentration-time curve (AUC). A weak DDI (AUC increase 48%-50%) was observed for gepotidacin co-administered with itraconazole. A clinically significant decrease in gepotidacin plasma AUC (52%) was observed with rifampicin coadministration, indicating a moderate DDI. There was no DDI for gepotidacin with cimetidine; a unique biomarker approach showed increased serum creatinine (24%), decreased renal clearance of creatinine (21%), and N1-methylnicotinamide (39%), which confirmed extensive MATE inhibition and partial OCT2 inhibition. Gepotidacin was not a P-gp DDI perpetrator, although the maximum plasma concentration of digoxin increased (53%) and is potentially clinically relevant given its narrow therapeutic index. Gepotidacin demonstrated weak CYP3A4 inhibition with midazolam (<2-fold AUC increase). There were no new safety-risk profile findings. These results will inform the safe and efficacious clinical use of gepotidacin when co-administered with other drugs.
Collapse
Affiliation(s)
- Aline Barth
- Global Blood Therapeutics, South San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Barth A, Hossain M, Perry CR, Gross AS, Ogura H, Shabbir S, Thomas S, Dumont EF, Brimhall DB, Srinivasan M, Swift B. Pharmacokinetic, Safety, and Tolerability Evaluations of Gepotidacin (GSK2140944) in Healthy Japanese Participants. Clin Pharmacol Drug Dev 2023; 12:38-56. [PMID: 36468634 PMCID: PMC10107257 DOI: 10.1002/cpdd.1192] [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: 05/20/2022] [Accepted: 10/02/2022] [Indexed: 12/12/2022]
Abstract
Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene antibiotic in late-phase development for uncomplicated urinary tract infection and uncomplicated urogenital gonorrhea. Two clinical studies were conducted to assess the pharmacokinetics (PK) and interethnic comparisons of oral gepotidacin (free-base and to-be-marketed mesylate formulations) administered as single doses ranging from 1500 to 3000 mg in fed and fasted states, and as 2 × 3000-mg doses given 12 hours apart under fed conditions in healthy participants of Japanese ancestry. Dose proportionality was observed in plasma exposures, and comparable area under the concentration-time curve (AUC) and maximum concentration were observed in fed and fasted states. Interethnic comparisons for Japanese versus non-Japanese participant data showed slightly higher plasma maximum concentration (7%-30%) yet similar plasma AUCs; slightly lower urine AUCs (11%-18%) were observed. The slightly higher plasma exposures in healthy Japanese versus White participants in the same study were attributed to lower mean body weights (64 kg versus ≈80 kg). Adverse events were primarily gastrointestinal, and when administered with food, gastrointestinal tolerability was improved. Overall, the gepotidacin PK and safety-risk profiles in healthy Japanese support potential evaluation of the global clinical doses in future studies.
Collapse
Affiliation(s)
- Aline Barth
- GSK, Collegeville, Pennsylvania, USA.,Present affiliation: Global Blood Therapeutics, San Francisco, California, USA
| | - Mohammad Hossain
- GSK, Collegeville, Pennsylvania, USA.,Present affiliation: Servier Pharmaceuticals, Boston, Massachusetts, USA
| | | | | | | | | | | | - Etienne F Dumont
- GSK, Collegeville, Pennsylvania, USA.,Present affiliation: Boston Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | | | | |
Collapse
|
25
|
In Vitro and In Vivo Activity of Gepotidacin against Drug-Resistant Mycobacterial Infections. Antimicrob Agents Chemother 2022; 66:e0056422. [PMID: 36445129 PMCID: PMC9765166 DOI: 10.1128/aac.00564-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Mycobacterial pathogens, including nontuberculous mycobacteria (NTM) and Mycobacterium tuberculosis, are pathogens of significant worldwide interest owing to their inherent drug resistance to a wide variety of FDA-approved drugs as well as causing a broad range of serious infections. Identifying new antibiotics active against mycobacterial pathogens is an urgent unmet need, especially those antibiotics that can bypass existing resistance mechanisms. In this study, we demonstrate that gepotidacin, a first-in-class triazaacenapthylene topoisomerase inhibitor, demonstrates potent activity against M. tuberculosis and M. fortuitum, as well as against other clinically relevant NTM species, including fluoroquinolone-resistant M. abscessus. Furthermore, gepotidacin exhibits concentration-dependent bactericidal activity against various mycobacterial pathogens, synergizes with several drugs utilized for their treatment, and reduces bacterial load in macrophages in intracellular killing assays comparably to amikacin. Additionally, M. fortuitum ATCC 6841 was unable to generate resistance to gepotidacin in vitro. When tested in a murine neutropenic M. fortuitum infection model, gepotidacin caused a significant reduction in bacterial load in various organs at a 10-fold lower concentration than amikacin. Taken together, these findings show that gepotidacin possesses a potentially new mechanism of action that enables it to escape existing resistance mechanisms. Thus, it can be projected as a potent novel lead for the treatment of mycobacterial infections, particularly for NTM, where present therapeutic interventions are extremely limited.
Collapse
|
26
|
Kresken M, Wohlfarth E, Weikel C, Butler D, Pfeifer Y, Werner G. In vitro activity of gepotidacin against urine isolates of Escherichia coli from outpatient departments in Germany. J Antimicrob Chemother 2022; 78:dkac406. [PMID: 36474310 DOI: 10.1093/jac/dkac406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/08/2022] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Escherichia coli is the leading pathogen of community-acquired urinary tract infections. Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene oral antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action that confers activity against most strains of target pathogens, such as E. coli, Staphylococcus saprophyticus and Neisseria gonorrhoeae, including those resistant to other antibiotics. OBJECTIVES This study assessed the in vitro activity of gepotidacin in comparison with ciprofloxacin and other oral standard-of-care antibiotics using a large collection of urine isolates of E. coli obtained from outpatients in Germany. METHODS Four hundred and sixty E. coli collected from 23 laboratories during a surveillance study in 2019/2020 were tested. Forty-six isolates (10.0%) produced an ESBL of the CTX-M family, half of which belonged to MDR clonal subgroups of E. coli ST131. Antibiotic susceptibilities were tested at a reference laboratory by broth microdilution according to the standard ISO 20776-1. RESULTS Fifty-three (11.5%) isolates were ciprofloxacin resistant, 25 (47.2%) of which also produced an ESBL. Overall, MIC50/90 values for gepotidacin were 2/4 mg/L (MIC range 0.125-16 mg/L), with no differences in activity between ciprofloxacin-susceptible and ciprofloxacin-resistant isolates, ESBL-producing and non-ESBL isolates, O25b-ST131 isolates, and isolates susceptible or resistant to fosfomycin, mecillinam or nitrofurantoin. CONCLUSIONS Gepotidacin showed promising in vitro activity against urine isolates of E. coli, including ciprofloxacin-resistant isolates, ESBL-producing isolates and isolates resistant to oral standard-of-care antibiotics.
Collapse
Affiliation(s)
- Michael Kresken
- Antiinfectives Intelligence GmbH, Cologne, Germany
- Rheinische Fachhochschule Köln gGmbH, Cologne, Germany
| | | | - Chase Weikel
- Infectious Diseases Research Unit, GSK, Collegeville, PA, USA
| | - Deborah Butler
- Infectious Diseases Research Unit, GSK, Collegeville, PA, USA
| | - Yvonne Pfeifer
- Department of Infectious Diseases, Division Nosocomial Pathogens and Antibiotic Resistances, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| | - Guido Werner
- Department of Infectious Diseases, Division Nosocomial Pathogens and Antibiotic Resistances, Robert Koch Institute, Wernigerode Branch, Wernigerode, Germany
| |
Collapse
|
27
|
Novel Bacterial Topoisomerase Inhibitor Gepotidacin Demonstrates Absence of Fluoroquinolone-Like Arthropathy in Juvenile Rats. Antimicrob Agents Chemother 2022; 66:e0048322. [PMID: 36255258 DOI: 10.1128/aac.00483-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Fluoroquinolone use in children is limited due to its potential toxicity and negative effects on skeletal development, but the actual effects/risks of fluoroquinolones on bone growth and the mechanisms behind fluoroquinolone-driven arthropathy remain unknown. Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene antibiotic with a unique mechanism of action that is not anticipated to have the same risks to bone growth as those of fluoroquinolones. Gepotidacin is in phase III clinical development for uncomplicated urinary tract infections (ClinicalTrials.gov identifiers NCT04020341 and NCT04187144) and urogenital gonorrhea (ClinicalTrials.gov identifier NCT04010539) in adults and adolescents ≥12 years of age. To inform arthropathy and other potential toxicity risks of gepotidacin in pediatric studies, this nonclinical study assessed oral gepotidacin toxicity in juvenile rats from postnatal day (PND) 4 to PND 32/35 (approximately equivalent to human ages from newborn to 11 years), using both in-life assessments (tolerability, toxicity, and toxicokinetics) and terminal assessments (necropsy with macroscopic and microscopic skeletal femoral head and/or stifle joint examinations). Gepotidacin doses of ≤300 mg/kg of body weight/day were well tolerated from PND 4 to PND 21, and higher doses of ≤1,250 mg/kg/day were well tolerated from PND 22 when the dose levels were escalated to maintain systemic exposure levels up to PND 35, with no observed treatment-related clinical signs, effects on mean body weight gain, or macroscopic findings on articular surfaces. A dose of 1,000 mg/kg/day was not tolerated during the dosing period from PND 4 to 21, with effects on body weight gain, fecal consistency, and body condition. Microscopic effects on articular surfaces were evaluated after 32 days of gepotidacin treatment at the highest tolerated dose. After 32 days of treatment with the highest tolerated gepotidacin dose of 300/1,250 mg/kg/day (systemic concentrations [area under the curve {AUC} values] of 93.7 μg · h/mL [males] and 121 μg · h/mL [females]), no skeletal effects on articular surfaces of the femoral head or stifle joint were observed. The absence of treatment-related clinical signs and arthropathy in juvenile rats provides evidence to support the potential future use of gepotidacin in children.
Collapse
|
28
|
Tiffany C, Dumont EF, Hossain M, Srinivasan M, Swift B. Pharmacokinetics, safety, and tolerability of gepotidacin administered as single or repeat ascending doses, in healthy adults and elderly subjects. Clin Transl Sci 2022; 15:2251-2264. [PMID: 35769034 PMCID: PMC9468557 DOI: 10.1111/cts.13359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/06/2022] [Accepted: 06/03/2022] [Indexed: 01/25/2023] Open
Abstract
Gepotidacin, a novel, first-in-class triazaacenaphthylene antibiotic, inhibits bacterial DNA replication by a distinct mechanism of action. We report the pharmacokinetics (PKs), safety, and tolerability of gepotidacin following single or multiple ascending doses. Studies 1 and 2 were randomized, single-blind, placebo-controlled trials in healthy adults aged 18-60 years, who received single (study 1 [NCT02202187]; 100-3000 mg) or repeat (study 2 [NCT01706315]; 400 mg twice daily to 2000 mg thrice daily) ascending doses of gepotidacin. Study 3 (NCT02045849) was an open-label, three-part, study in healthy adults; here, we report on part 3, a two-period, repeat-dose, crossover study. Healthy elderly participants received repeat 1500 mg gepotidacin twice daily with or without a moderate-fat meal. Primary end points were PKs (studies 1 and 2) and safety (studies 1 and 3 part 3). Gepotidacin PK parameters were comparable across all ages and were dose proportional. In all studies, gepotidacin was readily absorbed with median time to maximum concentration observed ranging from 1.0 to 4.0 h across all doses. Median apparent terminal phase half-life was consistent across studies and doses (range: 5.97-19.2 h). Steady-state was achieved following repeated dosing for 3-5 days; gepotidacin PK parameters were time invariant after repeated oral dosing. A moderate-fat meal did not affect gepotidacin PK parameters. Gepotidacin was generally well-tolerated, with no drug-related serious adverse events reported. Collectively, these PK and safety data across a wide range of doses in healthy participants aged greater than or equal to 18 years support the development of gepotidacin in further clinical studies.
Collapse
|
29
|
A Novel Oral GyrB/ParE Dual Binding Inhibitor Effective against Multidrug-Resistant Neisseria gonorrhoeae and Other High-Threat Pathogens. Antimicrob Agents Chemother 2022; 66:e0041422. [PMID: 35972242 PMCID: PMC9487510 DOI: 10.1128/aac.00414-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Drug-resistant Neisseria gonorrhoeae is a serious global health concern. New drugs are needed that can overcome existing drug resistance and limit the development of new resistances. Here, we describe the small molecule tricyclic pyrimidoindole JSF-2414 [8-(6-fluoro-8-(methylamino)-2-((2-methylpyrimidin-5-yl)oxy)-9H-pyrimido[4,5-b]indol-4-yl)-2-oxa-8-azaspiro[4.5]decan-3-yl)methanol], which was developed to target both ATP-binding regions of DNA gyrase (GyrB) and topoisomerase (ParE). JSF-2414 displays potent activity against N. gonorrhoeae, including drug-resistant strains. A phosphate pro-drug, JSF-2659, was developed to facilitate oral dosing. In two different animal models of Neisseria gonorrhoeae vaginal infection, JSF-2659 was highly efficacious in reducing microbial burdens to the limit of detection. The parent molecule also showed potent in vitro activity against high-threat Gram-positive organisms, and JSF-2659 was shown in a deep tissue model of vancomycin-resistant Staphylococcus aureus (VRSA) and a model of Clostridioides difficile-induced colitis to be highly efficacious and protective. JSF-2659 is a novel preclinical drug candidate against high-threat multidrug resistant organisms with low potential to develop new resistance.
Collapse
|
30
|
Jakielaszek C, Hossain M, Qian L, Fishman C, Widdowson K, Hilliard JJ, Mannino F, Raychaudhuri A, Carniel E, Demons S, Heine HS, Hershfield J, Russo R, Mega WM, Revelli D, O'Dwyer K. Gepotidacin is efficacious in a nonhuman primate model of pneumonic plague. Sci Transl Med 2022; 14:eabg1787. [PMID: 35648812 DOI: 10.1126/scitranslmed.abg1787] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gepotidacin is a first-in-class triazaacenaphthylene antibacterial agent that selectively inhibits bacterial DNA gyrase and topoisomerase IV through a unique binding mode and has the potential to treat a number of bacterial diseases. Development of this new agent to treat pneumonic plague caused by Yersinia pestis depends on the U.S. Food and Drug Administration Animal Rule testing pathway, as testing in humans is not feasible. Here, preclinical studies were conducted in the African green monkey (AGM) inhalational model of pneumonic plague to test the efficacy of gepotidacin. AGMs infected with Y. pestis were dosed intravenously with gepotidacin (48, 36, or 28 milligrams/kilogram per day) for 10 days to provide a plasma concentration that would support a rationale for a 1000 mg twice or thrice daily intravenous dose in humans or saline as a control. The primary end point was AGM survival with predefined euthanasia criteria. Secondary end points included survival duration and bacterial clearance. Gepotidacin showed activity in vitro against diverse Y. pestis isolates including antibiotic-resistant strains. All control animals in the inhalational plague studies succumbed to plague and were blood culture and organ culture positive for Y. pestis. Gepotidacin provided a 75 to 100% survival benefit with all dose regimens. All surviving animals were blood culture and organ culture negative for Y. pestis. Our randomized, controlled efficacy trials in the AGM pneumonic plague nonhuman primate model together with the in vitro Y. pestis susceptibility data support the use of gepotidacin as a treatment for pneumonic plague caused by Y. pestis.
Collapse
Affiliation(s)
| | | | - Lian Qian
- GlaxoSmithKline Pharmaceuticals, Collegeville, PA, USA
| | - Cindy Fishman
- GlaxoSmithKline Pharmaceuticals, Collegeville, PA, USA
| | | | | | - Frank Mannino
- GlaxoSmithKline Pharmaceuticals, Collegeville, PA, USA
| | | | | | - Samandra Demons
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Henry S Heine
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Jeremy Hershfield
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | | | - William M Mega
- Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, USA
| | - David Revelli
- Lovelace Biomedical and Environmental Research Institute, Albuquerque, NM, USA
| | - Karen O'Dwyer
- GlaxoSmithKline Pharmaceuticals, Collegeville, PA, USA
| |
Collapse
|
31
|
Lyons A, Kirkham J, Blades K, Orr D, Dauncey E, Smith O, Dick E, Walker R, Matthews T, Bunt A, Finlayson J, Morrison I, Savage VJ, Moyo E, Butler HS, Newman R, Ooi N, Smith A, Charrier C, Ratcliffe AJ, Stokes NR, Best S, Salisbury AM, Craighead M, Cooper IR. Discovery and structure-activity relationships of a novel oxazolidinone class of bacterial type II topoisomerase inhibitors. Bioorg Med Chem Lett 2022; 65:128648. [PMID: 35231579 DOI: 10.1016/j.bmcl.2022.128648] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 11/02/2022]
Abstract
There is an increasingly urgent and unmet medical need for novel antibiotic drugs that tackle infections caused by multidrug-resistant (MDR) pathogens. Novel bacterial type II topoisomerase inhibitors (NBTIs) are of high interest due to limited cross-resistance with fluoroquinolones, however analogues with Gram-negative activity often suffer from hERG channel inhibition. A novel series of bicyclic-oxazolidinone inhibitors of bacterial type II topoisomerase were identified which display potent broad-spectrum anti-bacterial activity, including against MDR strains, along with an encouraging in vitro safety profile. In vivo proof of concept was achieved in a A. baumannii mouse thigh infection model.
Collapse
Affiliation(s)
- Amanda Lyons
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - James Kirkham
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Kevin Blades
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - David Orr
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | | | - Oliver Smith
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Emma Dick
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Rolf Walker
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Teresa Matthews
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Adam Bunt
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | | | - Ian Morrison
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Victoria J Savage
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Emmanuel Moyo
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Hayley S Butler
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Rebecca Newman
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Nicola Ooi
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK
| | - Andrew Smith
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Cédric Charrier
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | | | - Neil R Stokes
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Stuart Best
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | | | - Mark Craighead
- Redx Anti-Infectives Ltd, Alderley Park, Cheshire SK10 4TG, UK
| | - Ian R Cooper
- Infex Therapeutics Ltd, Mereside, Alderley Park, Macclesfield SK10 4TG,UK.
| |
Collapse
|
32
|
Giacobbe DR, Dettori S, Corcione S, Vena A, Sepulcri C, Maraolo AE, De Rosa FG, Bassetti M. Emerging Treatment Options for Acute Bacterial Skin and Skin Structure Infections and Bloodstream Infections Caused by Staphylococcus aureus: A Comprehensive Review of the Evidence. Infect Drug Resist 2022; 15:2137-2157. [PMID: 35498629 PMCID: PMC9041368 DOI: 10.2147/idr.s318322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Daniele Roberto Giacobbe
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
- Correspondence: Daniele Roberto Giacobbe, Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, L.go R. Benzi 10, Genoa, 16132, Italy, Tel +390105554658, Email
| | - Silvia Dettori
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Silvia Corcione
- Department of Medical Sciences, Division of Infectious Diseases, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Antonio Vena
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Chiara Sepulcri
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | | | - Francesco Giuseppe De Rosa
- Department of Medical Sciences, Division of Infectious Diseases, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Matteo Bassetti
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
33
|
Scangarella-Oman NE, Hossain M, Hoover JL, Perry CR, Tiffany C, Barth A, Dumont EF. Dose Selection for Phase III Clinical Evaluation of Gepotidacin (GSK2140944) in the Treatment of Uncomplicated Urinary Tract Infections. Antimicrob Agents Chemother 2022; 66:e0149221. [PMID: 34978887 PMCID: PMC8923173 DOI: 10.1128/aac.01492-21] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Antibiotics are the current standard-of-care treatment for uncomplicated urinary tract infections (uUTIs). However, increasing rates of bacterial antibiotic resistance necessitate novel therapeutic options. Gepotidacin is a first-in-class triazaacenaphthylene antibiotic that selectively inhibits bacterial DNA replication by interaction with the bacterial subunits of DNA gyrase (GyrA) and topoisomerase IV (ParC). Gepotidacin is currently in clinical development for the treatment of uUTIs and other infections. In this article, we review data for gepotidacin from nonclinical studies, including in vitro activity, in vivo animal efficacy, and pharmacokinetic (PK) and pharmacokinetic/pharmacodynamic (PK/PD) models that informed dose selection for phase III clinical evaluation of gepotidacin. Based on this translational package of data, a gepotidacin 1,500-mg oral dose twice daily for 5 days was selected for two ongoing, randomized, multicenter, parallel-group, double-blind, double-dummy, active-comparator phase III clinical studies evaluating the safety and efficacy of gepotidacin in adolescent and adult female participants with uUTIs (ClinicalTrials.gov identifiers NCT04020341 and NCT04187144).
Collapse
Affiliation(s)
| | - Mohammad Hossain
- Research and Development, GlaxoSmithKline plc, Upper Providence, Pennsylvania, USA
| | - Jennifer L. Hoover
- Research and Development, GlaxoSmithKline plc, Upper Providence, Pennsylvania, USA
| | - Caroline R. Perry
- Research and Development, GlaxoSmithKline plc, Upper Providence, Pennsylvania, USA
| | - Courtney Tiffany
- Research and Development, GlaxoSmithKline plc, Upper Providence, Pennsylvania, USA
| | - Aline Barth
- Research and Development, GlaxoSmithKline plc, Upper Providence, Pennsylvania, USA
| | - Etienne F. Dumont
- Research and Development, GlaxoSmithKline plc, Upper Providence, Pennsylvania, USA
| |
Collapse
|
34
|
Hackel MA, Karlowsky JA, Canino MA, Sahm DF, Scangarella-Oman NE. In Vitro Activity of Gepotidacin against Gram-Negative and Gram-Positive Anaerobes. Antimicrob Agents Chemother 2022; 66:e0216521. [PMID: 34930028 PMCID: PMC8846401 DOI: 10.1128/aac.02165-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 11/21/2022] Open
Abstract
Gepotidacin (formerly GSK2140944) is a first-in-class triazaacenaphthylene antibacterial currently in phase III clinical trials. When tested against Gram-negative (n = 333) and Gram-positive (n = 225) anaerobes by agar dilution, gepotidacin inhibited 90% of isolates at concentrations of 4 and 2 μg/mL, respectively. Given gepotidacin's in vitro activity against the anaerobic isolates tested, further study is warranted to better understand the utility of gepotidacin in the treatment of infections caused by clinically relevant anaerobic organisms.
Collapse
Affiliation(s)
| | - James A. Karlowsky
- IHMA, Schaumburg, Illinois, USA
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | | | | |
Collapse
|
35
|
Sanders MI, Ali E, Buer J, Steinmann J, Rath PM, Verhasselt HL, Kirchhoff L. Antibacterial Activity of the Novel Drug Gepotidacin against Stenotrophomonas maltophilia—An In Vitro and In Vivo Study. Antibiotics (Basel) 2022; 11:antibiotics11020192. [PMID: 35203795 PMCID: PMC8868531 DOI: 10.3390/antibiotics11020192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 02/04/2023] Open
Abstract
Stenotrophomonas maltophilia is increasingly recognized as a nosocomial bacterial pathogen with a multi-drug resistance profile. In this study, the novel drug gepotidacin, the first compound of the novel triazaacenaphthylene topoisomerase inhibitor antibiotics class, was evaluated on its activity against clinical S. maltophilia isolates. Ninety-nine S. maltophilia isolates plus reference strain K279a (N = 100) were tested on their susceptibility towards gepotidacin in a broth microdilution. Additional susceptibility testing was performed towards the commonly applied combination trimethoprim/sulfamethoxazole (TMP/SXT), moxifloxacin, and levofloxacin. The time–kill kinetic of gepotidacin was observed in a time–kill assay. The greater wax moth Galleria mellonella was used to determine the activity of gepotidacin against S. maltophilia in vivo. Gepotidacin showed minimum inhibitory concentrations (MICs) between 0.25 and 16 mg/L (MIC50: 2 mg/L; MIC90: 8 mg/L), independently of its susceptibility towards TMP/SXT. The five TMP/SXT resistant strains exhibited gepotidacin MICs from 1 to 4 mg/L. The S. maltophilia strains resistant to the assessed fluoroquinolones showed in parts high MICs of gepotidacin. The time–kill assay revealed a time- and strain-dependent killing effect of gepotidacin. In vivo, injection of gepotidacin increased the survival rate of the larvae from 61 % to 90 % after 2 days. This study showed antimicrobial effects of gepotidacin towards S. maltophilia.
Collapse
Affiliation(s)
- Maike Isabell Sanders
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
| | - Eyhab Ali
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
| | - Joerg Steinmann
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
- Institute of Clinical Hygiene, Medical Microbiology and Infectiology, Klinikum Nürnberg, Paracelsus Medical University, 90419 Nuremberg, Germany
| | - Peter-Michael Rath
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
| | - Hedda Luise Verhasselt
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
| | - Lisa Kirchhoff
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany; (M.I.S.); (E.A.); (J.B.); (J.S.); (P.-M.R.); (H.L.V.)
- Correspondence: ; Tel.: +49-201-723-3505
| |
Collapse
|
36
|
Gepotidacin Pharmacokinetics-Pharmacodynamics against Escherichia coli in the One-Compartment and Hollow-Fiber In Vitro Infection Model Systems. Antimicrob Agents Chemother 2021; 65:e0012221. [PMID: 34543096 PMCID: PMC8597728 DOI: 10.1128/aac.00122-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Gepotidacin is a novel, first-in-class triazaacenaphthylene antibiotic that inhibits bacterial DNA replication by a distinct mechanism of action with an in vitro spectrum of activity that includes Escherichia coli. Our objectives herein were the following: (i) to identify the pharmacokinetic-pharmacodynamic (PK-PD) index associated with the efficacy of gepotidacin against E. coli; (ii) to determine the magnitude of the above-described PK-PD index associated with various bacterial reduction endpoints for E. coli; and (iii) to characterize the relationship between gepotidacin exposure and on-therapy E. coli resistance amplification. A 24-h one-compartment in vitro infection model was used to investigate the first two study objectives, and a 10-day hollow-fiber in vitro infection model was used to evaluate the third objective. For the dose-fractionation studies (objective i) in which E. coli NCTC 13441 (gepotidacin MIC, 2 mg/liter) was evaluated, gepotidacin free-drug area under the concentration-time curve (AUC) from 0 to 24 h to the MIC (AUC/MIC ratio) was identified as the PK-PD index most closely associated with change in bacterial burden (r2 = 0.925). For the dose-ranging studies (objective ii), in which four E. coli isolates (gepotidacin MIC range, 1 to 4 mg/liter) were studied, the magnitude of the median gepotidacin free-drug AUC/MIC ratio associated with net bacterial stasis and 1- and 2-log10 CFU reductions for the pooled data set was 33.9, 43.7, and 60.7, respectively. For the hollow-fiber in vitro infection model studies (objective iii), in which one isolate (E. coli NCTC 13441; gepotidacin MIC, 2 mg/liter) was evaluated, gepotidacin free-drug AUC/MIC ratios of 275 and greater were sufficient to suppress on-therapy resistance amplification. Together, the data generated from these studies will be useful to support discrimination among candidate dosing regimens for future clinical study.
Collapse
|
37
|
Pharmacokinetics of Oral Formulations of Gepotidacin (GSK2140944), a Triazaacenaphthylene Bacterial Type II Topoisomerase Inhibitor, in Healthy Adult and Adolescent Participants. Antimicrob Agents Chemother 2021; 66:e0126321. [PMID: 34633853 PMCID: PMC8765319 DOI: 10.1128/aac.01263-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Gepotidacin is a novel, first-in-class triazaacenaphthylene antibiotic that may provide a new treatment option for antibiotic-resistant pathogens. Two pharmacokinetic evaluations of oral gepotidacin are presented; a relative bioavailability study that guided formulation development, followed by an adult and adolescent study of the final formulation. In the relative bioavailability study, after gepotidacin administration to 26 healthy adults as free base roller compacted (RC) tablets, free base high shear wet granulation (HSWG) tablets, and mesylate salt reference capsules, the RC tablet exposure ratios and 90% confidence intervals (CIs) were within the 0.80 to 1.25 confidence bounds; however, the HSWG tablet maximum observed concentration (Cmax) was higher compared to the reference (ratio: 1.15; 90% CIs: 1.0113, 1.3047). In the healthy adult (n=16) and adolescent (n=17) study, a gepotidacin mesylate salt tablet was evaluated as a 1,500-mg single dose, 2 doses administered 6 or 12 h apart (6,000 mg total), or placebo. Single-dose mean Cmax was ∼27% higher in adolescents versus adults and area under the concentration-time curve (AUC) was comparable in both populations. After 2 doses were administered, mean Cmax was similar for both ages and mean AUC was ∼35% higher in adolescents versus adults. Concentrations increased proportionally with dose. Safety-risk profiles were similar in both ages. Across studies, the most common adverse events were gastrointestinal. Overall, the pharmacokinetics of the final gepotidacin mesylate salt tablet have been well-characterized, enrollment of adolescents into the pivotal trials is supported, and dosing intervals were determined that should provide adequate exposures for microbiological efficacy.
Collapse
|
38
|
Jaradat ZW, Khwaileh M, Al Mousa W, Ababneh QO, Al Nabulsi A. Occurrence, distribution and pattern analysis of methicillin resistant (MRSA) and methicillin sensitive (MSSA) Staphylococcus aureus on fomites in public facilities. Pathog Glob Health 2021; 115:377-391. [PMID: 34338618 PMCID: PMC8592605 DOI: 10.1080/20477724.2021.1906563] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a human pathogen incriminated as a causative agent of hospital nosocomial infections as well as a wide range of diseases in communities. This study was conducted to assess the occurrence and distribution of MRSA and methicillin-sensitive (MSSA) on different fomites in public facilities in northern Jordan and to determine their antibiograms, toxin genes profiles, as well as identify their genetic relatedness. A total of 2600 swabs were collected from 14 fomite surfaces in a variety of public facilities including hospitals, universities, schools, transportation sites, and market places. The identity of the 380 S. aureus isolates was confirmed. Among them, 158 (41.6%) were MRSA while the rest of the isolates, 222 (58.4%) were MSSA. MRSA isolates were recovered from all fomites sites. However, among the total collected samples, the percentages of MRSA in public facilities were significantly higher in hospitals and transportation fomites, while percentages of MRSA among fomites sites were higher in public reception sites, chairs, and toilet seats. Antibiotic resistance profiles indicated that 24.5% of the isolates were resistant to cefoxitin, oxacillin, and oxytetracycline. In contrast, only 3.95% were resistant to trimethoprim-sulfamethoxazole, and 15.3% were resistant to ciprofloxacin. Multidrug-resistant patterns were higher in MRSA than in MSSA isolates. There was no apparent difference in toxin gene profiles between MRSA and MSSA. Molecular analysis revealed 85 patterns and 16 clusters at a 9% mean similarity level. In conclusion, this study provides evidence for the potential of MRSA transmission via inanimate surfaces.
Collapse
Affiliation(s)
- Ziad W Jaradat
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Maysoon Khwaileh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Waseem Al Mousa
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Qutaiba O Ababneh
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, Jordan
| | - Anas Al Nabulsi
- Department of Nutrition and Food Technology, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
39
|
Analysis of antimicrobial susceptibility testing methods and variables and in vitro activity of gepotidacin against urogenital Neisseria gonorrhoeae in men. Diagn Microbiol Infect Dis 2021; 101:115484. [PMID: 34419740 DOI: 10.1016/j.diagmicrobio.2021.115484] [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: 04/09/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 11/21/2022]
Abstract
Gepotidacin is a triazaacenaphthylene antibiotic with activity against Neisseria gonorrhoeae including strains resistant to current agents. We tested 145 N. gonorrhoeae isolates by agar dilution according to Gonococcal Isolate Surveillance Program and Clinical and Laboratory Standards Institute methodologies. Gepotidacin demonstrated a minimum inhibitory concentration (MIC)50 of 0.25 μg/mL and a MIC90 of 0.5 μg/mL (highest gepotidacin MIC was 1 μg/mL) against the 145 N. gonorrhoeae isolates tested. We also assessed the impact of test variables on antimicrobial susceptibility test results for gepotidacin, ciprofloxacin, and ceftriaxone against 10 N. gonorrhoeae isolates. Media type had the biggest effect but wasn't specific to gepotidacin. Gepotidacin MICs were also affected by inoculum, pH, and 10% CO2. These in vitro data indicate that further study of gepotidacin is warranted for potential use in treating gonorrhea infections and highlight the importance of controlling for media type, inoculum, CO2, and pH when performing MIC testing with gepotidacin.
Collapse
|
40
|
Potential role of new-generation antibiotics in acute bacterial skin and skin structure infections. Curr Opin Infect Dis 2021; 34:109-117. [PMID: 33395093 DOI: 10.1097/qco.0000000000000708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To summarize the available results of primary analyses from high-quality randomized studies of either recently approved or possible future agents for the treatment of acute bacterial skin and skin structure infections (ABSSSI). RECENT FINDINGS In the last 2 decades, several novel agents have been approved for the treatment of ABSSSI, that are also active against methicillin-resistant Staphylococcus aureus (MRSA). In addition to already available agents, further molecules are in clinical development that could become available for treating ABSSSI in the forthcoming future. SUMMARY The current and future availability of several new-generation antibiotics will allow to modulate therapeutic choices not only on efficacy but also on other relevant factors such as the combination of the drug safety profile and the comorbidities of any given patient, the expected adherence to outpatient therapy, and the possibilities of early discharge or avoiding hospitalization by means of oral formulations, early switch from intravenous to oral therapy, or single-dose administration of long-acting intravenous agents. With the advent of new-generation antibiotics, all these factors are becoming increasingly essential for tailoring treatment to individual patients in line with the principles of personalized medicine, and for optimizing the use of healthcare resources.
Collapse
|
41
|
Nguyen D, Shaik JS, Tai G, Tiffany C, Perry C, Dumont E, Gardiner D, Barth A, Singh R, Hossain M. Comparison between physiologically based pharmacokinetic and population pharmacokinetic modelling to select paediatric doses of gepotidacin in plague. Br J Clin Pharmacol 2021; 88:416-428. [PMID: 34289143 PMCID: PMC9293063 DOI: 10.1111/bcp.14996] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 05/03/2021] [Accepted: 05/09/2021] [Indexed: 12/31/2022] Open
Abstract
Aims To develop physiologically based pharmacokinetic (PBPK) and population pharmacokinetic (PopPK) models to predict effective doses of gepotidacin in paediatrics for the treatment of pneumonic plague (Yersinia pestis). Methods A gepotidacin PBPK model was constructed using a population‐based absorption, distribution, metabolism and excretion simulator, Simcyp®, with physicochemical and in vitro data, optimized with clinical data from a dose‐escalation intravenous (IV) study and a human mass balance study. A PopPK model was developed with pooled PK data from phase 1 studies with IV gepotidacin in healthy adults. Results For both the PopPK and PBPK models, body weight was found to be a key covariate affecting gepotidacin clearance. With PBPK, ~90% of the predicted PK for paediatrics fell between the 5th and 95th percentiles of adult values except for subjects weighing ≤5 kg. PopPK‐simulated paediatric means for Cmax and AUC(0‐τ) were similar to adult exposures across various weight brackets. The proposed dosing regimens were weight‐based for subjects ≤40 kg and fixed‐dose for subjects >40 kg. Comparison of observed and predicted exposures in adults indicated that both PBPK and PopPK models achieved similar AUC and Cmax for a given dose, but the Cmax predictions with PopPK were slightly higher than with PBPK. The two models differed on dose predictions in children <3 months old. The PopPK model may be suboptimal for low age groups due to the absence of maturation characterization of drug‐metabolizing enzymes involved with clearance in adults. Conclusions Both PBPK and PopPK approaches can reasonably predict gepotidacin exposures in children.
Collapse
Affiliation(s)
- Dung Nguyen
- GlaxoSmithKline, Collegeville, PA, United States
| | | | - Guoying Tai
- GlaxoSmithKline, Collegeville, PA, United States
| | | | | | | | | | - Aline Barth
- GlaxoSmithKline, Collegeville, PA, United States
| | | | | |
Collapse
|
42
|
Nuzzo A, Van Horn S, Traini C, Perry CR, Dumont EF, Scangarella-Oman NE, Gardiner DF, Brown JR. Microbiome recovery in adult females with uncomplicated urinary tract infections in a randomised phase 2A trial of the novel antibiotic gepotidacin (GSK140944). BMC Microbiol 2021; 21:181. [PMID: 34130619 PMCID: PMC8207760 DOI: 10.1186/s12866-021-02245-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 06/02/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND With increasing concerns about the impact of frequent antibiotic usage on the human microbiome, it is important to characterize the potential for such effects in early antibiotic drug development clinical trials. In a randomised Phase 2a clinical trial study that evaluated the pharmacokinetics of repeated oral doses of gepotidacin, a first-in-chemical-class triazaacenaphthylene antibiotic with a distinct mechanism of action, in adult females with uncomplicated urinary tract infections for gepotidacin (GSK2140944) we evaluated the potential changes in microbiome composition across multiple time points and body-sites ( ClinicalTrials.gov : NCT03568942). RESULTS Samples of gastrointestinal tract (GIT), pharyngeal cavity and vaginal microbiota were collected with consent from 22 patients at three time points relative to the gepotidacin dosing regimen; Day 1 (pre-dose), Day 5 (end of dosing) and Follow-up (Day 28 ± 3 days). Microbiota composition was determined by DNA sequencing of 16S rRNA gene variable region 4 amplicons. By Day 5, significant changes were observed in the microbiome diversity relative to pre-dose across the tested body-sites. However, by the Follow-up visit, microbiome diversity changes were reverted to compositions comparable to Day 1. The greatest range of microbiome changes by body-site were GIT followed by the pharyngeal cavity then vagina. In Follow-up visit samples we found no statistically significant occurrences of pathogenic taxa. CONCLUSION Our findings suggest that gepotidacin alteration of the human microbiome after 5 days of dosing is temporary and rebound to pre-dosing states is evident within the first month post-treatment. We recommend that future antibiotic drug trials include similar exploratory investigations into the duration and context of microbiome modification and recovery. TRIAL REGISTRATION NCT03568942 . Registered 26 June 2018.
Collapse
Affiliation(s)
- Andrea Nuzzo
- Human Genetics, GlaxoSmithKline R&D, Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY UK
| | | | | | | | | | | | | | - James R. Brown
- Human Genetics, GlaxoSmithKline R&D, Collegeville, PA USA
- Present Address: Kaleido Biosciences, 65 Hayden Avenue, Lexington, MA 02421 USA
| |
Collapse
|
43
|
Towards Conformation-Sensitive Inhibition of Gyrase: Implications of Mechanistic Insight for the Identification and Improvement of Inhibitors. Molecules 2021; 26:molecules26051234. [PMID: 33669078 PMCID: PMC7956263 DOI: 10.3390/molecules26051234] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 12/17/2022] Open
Abstract
Gyrase is a bacterial type IIA topoisomerase that catalyzes negative supercoiling of DNA. The enzyme is essential in bacteria and is a validated drug target in the treatment of bacterial infections. Inhibition of gyrase activity is achieved by competitive inhibitors that interfere with ATP- or DNA-binding, or by gyrase poisons that stabilize cleavage complexes of gyrase covalently bound to the DNA, leading to double-strand breaks and cell death. Many of the current inhibitors suffer from severe side effects, while others rapidly lose their antibiotic activity due to resistance mutations, generating an unmet medical need for novel, improved gyrase inhibitors. DNA supercoiling by gyrase is associated with a series of nucleotide- and DNA-induced conformational changes, yet the full potential of interfering with these conformational changes as a strategy to identify novel, improved gyrase inhibitors has not been explored so far. This review highlights recent insights into the mechanism of DNA supercoiling by gyrase and illustrates the implications for the identification and development of conformation-sensitive and allosteric inhibitors.
Collapse
|
44
|
New Topoisomerase Inhibitors: Evaluating the Potency of Gepotidacin and Zoliflodacin in Fluoroquinolone-Resistant Escherichia coli upon tolC Inactivation and Differentiating Their Efflux Pump Substrate Nature. Antimicrob Agents Chemother 2021; 65:AAC.01803-20. [PMID: 33199388 DOI: 10.1128/aac.01803-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Inactivating tolC in multidrug-resistant Escherichia coli with differing sequence types and quinolone resistance-determining mutations reveals remarkably potentiated activity of the first-in-class topoisomerase inhibitors gepotidacin and zoliflodacin. Differences between both structurally unrelated compounds in comparison to fluoroquinolones regarding the selectivity of E. coli RND (resistance-nodulation-cell division)-type transporters, efflux inhibitors, and AcrB porter domain mutations were demonstrated. The findings should reinforce efforts to develop efflux-bypassing drugs and provide AcrB targets with critical relevance for this purpose.
Collapse
|
45
|
Kong Q, Yang Y. Recent advances in antibacterial agents. Bioorg Med Chem Lett 2021; 35:127799. [PMID: 33476772 DOI: 10.1016/j.bmcl.2021.127799] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/10/2021] [Accepted: 01/11/2021] [Indexed: 12/12/2022]
Abstract
Antimicrobial resistance is a global challenge and the effectiveness of old antibiotics is decreasing. Discovery and development of antibacterial agents have been accelerated to replenish the arsenal of antibiotics which is limited and shrinking. In recent years, significant advances have achieved in the antibacterial area, including new compounds of known classes and new compounds with new mechanisms. This review summarizes these advances and provides perspective on future directions of antibacterial agents.
Collapse
Affiliation(s)
- Qidi Kong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, China
| | - Yushe Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No.19(A) Yuquan Road, Beijing 100049, China.
| |
Collapse
|
46
|
Hossain M, Tiffany C, Tao Y, Barth A, Marbury TC, Preston RA, Dumont E. Pharmacokinetics of Gepotidacin in Subjects With Normal Hepatic Function and Hepatic Impairment. Clin Pharmacol Drug Dev 2021; 10:588-597. [PMID: 33450142 PMCID: PMC8248074 DOI: 10.1002/cpdd.913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 11/18/2022]
Abstract
Gepotidacin is a novel triazaacenaphthylene bacterial topoisomerase inhibitor. This phase 1 nonrandomized, open‐label, multicenter, 2‐part study evaluated the pharmacokinetics, safety, and tolerability of oral gepotidacin 1500 mg in 3 different hepatic settings (normal, moderate impairment, and severe impairment). Gepotidacin was safe and generally tolerated in all subjects. Compared to subjects with normal hepatic function, gepotidacin plasma area under the plasma concentration–time curve from time 0 to infinity (AUC0–∞) and maximum concentration significantly increased by 1.7‐ and 1.9‐fold, respectively, in severe hepatic impairment; increases in moderate impairment were not statistically significant. No significant effect was observed for gepotidacin plasma elimination half‐life (geometric mean range, 8.2–9.1 hours) across hepatic groups. Renal clearance increased in moderate (16%) and severe (52%) hepatic impairment vs normal. The mean fraction of gepotidacin dose excreted in urine increased with increasing hepatic impairment (normal, 7.5%; moderate, 11.2%; and severe, 19.9%). Urine gepotidacin concentrations remained high for 12 hours in all hepatic groups after dosing. Saliva gepotidacin concentrations displayed a linear relationship with plasma concentrations (R2 = 0.76). The ratio of saliva AUC to unbound plasma AUC and elimination half‐life were not affected by hepatic impairment. These data indicate that gepotidacin dose adjustment is not required in mild to moderate hepatic impairment; severe hepatic impairment may require increases in dosing interval or dose reduction.
Collapse
Affiliation(s)
| | | | - Yu Tao
- GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Aline Barth
- GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | | | - Richard A Preston
- Clinical Pharmacology Research Unit, Division of Clinical Pharmacology University of Miami, Department of Medicine, Miller School of Medicine, University of Miami Clinical and Translational Science Institutes (CTSI) and The Peggy and Harold Katz Family Drug Discovery Center, Miami, Florida, USA
| | | |
Collapse
|
47
|
Cools F, Delputte P, Cos P. The search for novel treatment strategies for Streptococcus pneumoniae infections. FEMS Microbiol Rev 2021; 45:6064299. [PMID: 33399826 PMCID: PMC8371276 DOI: 10.1093/femsre/fuaa072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 01/01/2021] [Indexed: 12/13/2022] Open
Abstract
This review provides an overview of the most important novel treatment strategies against Streptococcus pneumoniae infections published over the past 10 years. The pneumococcus causes the majority of community-acquired bacterial pneumonia cases, and it is one of the prime pathogens in bacterial meningitis. Over the last 10 years, extensive research has been conducted to prevent severe pneumococcal infections, with a major focus on (i) boosting the host immune system and (ii) discovering novel antibacterials. Boosting the immune system can be done in two ways, either by actively modulating host immunity, mostly through administration of selective antibodies, or by interfering with pneumococcal virulence factors, thereby supporting the host immune system to effectively overcome an infection. While several of such experimental therapies are promising, few have evolved to clinical trials. The discovery of novel antibacterials is hampered by the high research and development costs versus the relatively low revenues for the pharmaceutical industry. Nevertheless, novel enzymatic assays and target-based drug design, allow the identification of targets and the development of novel molecules to effectively treat this life-threatening pathogen.
Collapse
Affiliation(s)
- F Cools
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Delputte
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - P Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
48
|
Lu Y, Papa JL, Nolan S, English A, Seffernick JT, Shkolnikov N, Powell J, Lindert S, Wozniak DJ, Yalowich J, Mitton-Fry MJ. Dioxane-Linked Amide Derivatives as Novel Bacterial Topoisomerase Inhibitors against Gram-Positive Staphylococcus aureus. ACS Med Chem Lett 2020; 11:2446-2454. [PMID: 33335666 DOI: 10.1021/acsmedchemlett.0c00428] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
In recent years, novel bacterial topoisomerase inhibitors (NBTIs) have been developed as future antibacterials for treating multidrug-resistant bacterial infections. A series of dioxane-linked NBTIs with an amide moiety has been synthesized and evaluated. Compound 3 inhibits DNA gyrase, induces the formation of single strand breaks to bacterial DNA, and achieves potent antibacterial activity against a variety of Gram-positive pathogens, including methicillin-resistant Staphylococcus aureus (MRSA). Optimization of this series of analogues led to the discovery of a subseries of compounds (22-25) with more potent anti-MRSA activity, dual inhibition of DNA gyrase and topoisomerase IV, and the ability to induce double strand breaks through inhibition of S. aureus DNA gyrase.
Collapse
|
49
|
Nyerges A, Tomašič T, Durcik M, Revesz T, Szili P, Draskovits G, Bogar F, Skok Ž, Zidar N, Ilaš J, Zega A, Kikelj D, Daruka L, Kintses B, Vasarhelyi B, Foldesi I, Kata D, Welin M, Kimbung R, Focht D, Mašič LP, Pal C. Rational design of balanced dual-targeting antibiotics with limited resistance. PLoS Biol 2020; 18:e3000819. [PMID: 33017402 PMCID: PMC7561186 DOI: 10.1371/journal.pbio.3000819] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 10/15/2020] [Accepted: 08/26/2020] [Indexed: 12/02/2022] Open
Abstract
Antibiotics that inhibit multiple bacterial targets offer a promising therapeutic strategy against resistance evolution, but developing such antibiotics is challenging. Here we demonstrate that a rational design of balanced multitargeting antibiotics is feasible by using a medicinal chemistry workflow. The resultant lead compounds, ULD1 and ULD2, belonging to a novel chemical class, almost equipotently inhibit bacterial DNA gyrase and topoisomerase IV complexes and interact with multiple evolutionary conserved amino acids in the ATP-binding pockets of their target proteins. ULD1 and ULD2 are excellently potent against a broad range of gram-positive bacteria. Notably, the efficacy of these compounds was tested against a broad panel of multidrug-resistant Staphylococcus aureus clinical strains. Antibiotics with clinical relevance against staphylococcal infections fail to inhibit a significant fraction of these isolates, whereas both ULD1 and ULD2 inhibit all of them (minimum inhibitory concentration [MIC] ≤1 μg/mL). Resistance mutations against these compounds are rare, have limited impact on compound susceptibility, and substantially reduce bacterial growth. Based on their efficacy and lack of toxicity demonstrated in murine infection models, these compounds could translate into new therapies against multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Akos Nyerges
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Martina Durcik
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Tamas Revesz
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
- Doctoral School of Theoretical Medicine, University of Szeged, Szeged, Hungary
| | - Petra Szili
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, Hungary
| | - Gabor Draskovits
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
| | - Ferenc Bogar
- MTA-SZTE Biomimetic Systems Research Group, Department of Medical Chemistry, University of Szeged, Hungary
| | - Žiga Skok
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Janez Ilaš
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Anamarija Zega
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Danijel Kikelj
- University of Ljubljana, Faculty of Pharmacy, Ljubljana, Slovenia
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
- Doctoral School of Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Balint Kintses
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
- HCEMM-BRC Translational Microbiology Lab, Szeged, Hungary
| | - Balint Vasarhelyi
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
| | - Imre Foldesi
- Department of Laboratory Medicine, University of Szeged, Szeged, Hungary
| | - Diána Kata
- Department of Laboratory Medicine, University of Szeged, Szeged, Hungary
| | - Martin Welin
- SARomics Biostructures, Medicon Village, Lund, Sweden
| | | | - Dorota Focht
- SARomics Biostructures, Medicon Village, Lund, Sweden
| | | | - Csaba Pal
- Synthetic and Systems Biology Unit, Biological Research Center, Szeged, Hungary
| |
Collapse
|
50
|
Lewis DA. New treatment options for Neisseria gonorrhoeae in the era of emerging antimicrobial resistance. Sex Health 2020; 16:449-456. [PMID: 31292063 DOI: 10.1071/sh19034] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/16/2019] [Indexed: 12/27/2022]
Abstract
Neisseria gonorrhoeae, the causative agent of gonorrhoea, has rapidly evolved from an exquisitely susceptible pathogen into a 'superbug' with the capacity to exhibit an extensively drug resistant (XDR) phenotype. The threat of untreatable gonorrhoea now looms on the horizon while the arsenal of effective antimicrobial agents diminishes with time. Ceftriaxone remains the mainstay of first-line therapy as a single agent or as the backbone of a dual therapy regimen. The implementation of new assays to facilitate 'precision' treatment, based on the prediction of N. gonorrhoeae susceptibility to old anti-gonococcal drugs, may enable sparing use of ceftriaxone in those countries that can afford this technology. A few existing drugs, such as ertapenem, can be repositioned to help manage multi-drug resistant and XDR gonorrhoea. Recent clinical trials involving solithromycin and delafloxacin have generated disappointing results in that both agents failed to show non-inferiority to conventional ceftriaxone-based regimens. At present, zoliflodacin and gepotidacin appear to be the most promising antimicrobial agents in clinical development. Both drugs performed well in eradicating urogenital gonorrhoea in recent Phase 2 trials; however, treatment failures were reported at the oropharyngeal site, which is an important site of infection in men who have sex with men and sex workers. Given this observation, it is unlikely that either of these new agents could be promoted for monotherapy of gonorrhoea. The pre-clinical pipeline remains relatively empty of agents likely to progress to clinical development for gonorrhoea treatment and increased investment into gonorrhoea-specific drug discovery is recommended.
Collapse
Affiliation(s)
- David A Lewis
- Western Sydney Sexual Health Centre, Western Sydney Local Health District, 162 Marsden Street, Parramatta, NSW 2150, Australia
| |
Collapse
|