1
|
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
|
2
|
Yang H, Yao Z, Yang K, Wang C, Li M, Zhang Y, Yan J, Lv R, Wang Y, Huang A, Zhang D, Li W, Wu Y, Miao Z. Synthesis and Antibacterial Evaluation of Novel Psoralen Derivatives against Methicillin-Resistant Staphylococcus aureus (MRSA). Chem Biodivers 2024; 21:e202302048. [PMID: 38263380 DOI: 10.1002/cbdv.202302048] [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] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/25/2024]
Abstract
Today, the bacterial infections caused by multidrug-resistant pathogens seriously threaten human health. Thereby, there is an urgent need to discover antibacterial drugs with novel mechanism. Here, novel psoralen derivatives had been designed and synthesized by a scaffold hopping strategy. Among these targeted twenty-five compounds, compound ZM631 showed the best antibacterial activity against methicillin-resistant S. aureus (MRSA) with the low MIC of 1 μg/mL which is 2-fold more active than that of the positive drug gepotidacin. Molecular docking study revealed that compound ZM631 fitted well in the active pockets of bacterial S. aureus DNA gyrase and formed a key hydrogen bond binding with the residue ASP-1083. These findings demonstrated that the psoralen scaffold could serve as an antibacterial lead compound for further drug development against multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Hang Yang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Zheng Yao
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Keli Yang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Chuanhao Wang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei Road, Nanjing, 210094, the People's Republic of China
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, the People's Republic of China
| | - Mochenxuan Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, the People's Republic of China
| | - Yanming Zhang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Jianyu Yan
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Rongxue Lv
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Yongchuang Wang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Anhua Huang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Daozuan Zhang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Wei Li
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, the People's Republic of China
| | - Yuelin Wu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Zhenyuan Miao
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| |
Collapse
|
3
|
Oviatt A, Gibson EG, Huang J, Mattern K, Neuman KC, Chan PF, Osheroff N. Interactions between Gepotidacin and Escherichia coli Gyrase and Topoisomerase IV: Genetic and Biochemical Evidence for Well-Balanced Dual-Targeting. ACS Infect Dis 2024; 10:1137-1151. [PMID: 38606465 PMCID: PMC11015057 DOI: 10.1021/acsinfecdis.3c00346] [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: 07/19/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 04/13/2024]
Abstract
Antimicrobial resistance is a global threat to human health. Therefore, efforts have been made to develop new antibacterial agents that address this critical medical issue. Gepotidacin is a novel, bactericidal, first-in-class triazaacenaphthylene antibacterial in clinical development. Recently, phase III clinical trials for gepotidacin treatment of uncomplicated urinary tract infections caused by uropathogens, including Escherichia coli, were stopped for demonstrated efficacy. Because of the clinical promise of gepotidacin, it is important to understand how the compound interacts with its cellular targets, gyrase and topoisomerase IV, from E. coli. Consequently, we determined how gyrase and topoisomerase IV mutations in amino acid residues that are involved in gepotidacin interactions affect the susceptibility of E. coli cells to the compound and characterized the effects of gepotidacin on the activities of purified wild-type and mutant gyrase and topoisomerase IV. Gepotidacin displayed well-balanced dual-targeting of gyrase and topoisomerase IV in E. coli cells, which was reflected in a similar inhibition of the catalytic activities of these enzymes by the compound. Gepotidacin induced gyrase/topoisomerase IV-mediated single-stranded, but not double-stranded, DNA breaks. Mutations in GyrA and ParC amino acid residues that interact with gepotidacin altered the activity of the compound against the enzymes and, when present in both gyrase and topoisomerase IV, reduced the antibacterial activity of gepotidacin against this mutant strain. Our studies provide insights regarding the well-balanced dual-targeting of gyrase and topoisomerase IV by gepotidacin in E. coli.
Collapse
Affiliation(s)
- Alexandria
A. Oviatt
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Elizabeth G. Gibson
- Department
of Biochemistry, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
- Department
of Pharmacology, Vanderbilt University School
of Medicine, Nashville, Tennessee 37232, United States
| | - Jianzhong Huang
- Infectious
Diseases Research Unit, GlaxoSmithKline, Collegeville, Pennsylvania 19426, United States
| | - Karen Mattern
- Infectious
Diseases Research Unit, GlaxoSmithKline, Collegeville, Pennsylvania 19426, United States
| | - Keir C. Neuman
- Laboratory
of Single Molecule Biophysics, National
Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland 20982, United States
| | - Pan F. Chan
- Infectious
Diseases Research Unit, GlaxoSmithKline, Collegeville, Pennsylvania 19426, 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
- VA
Tennessee
Valley Healthcare System, Nashville, Tennessee 37212, United States
| |
Collapse
|
4
|
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
|
5
|
Cumming JG, Kreis L, Kühne H, Wermuth R, Vercruysse M, Cantrill C, Bissantz C, Qiu H, Kramer C, Andreotti D, Fossati G. Novel Indane-Containing NBTIs with Potent Anti-Gram-Negative Activity and Minimal hERG Inhibition. ACS Med Chem Lett 2023; 14:1791-1799. [PMID: 38116438 PMCID: PMC10726470 DOI: 10.1021/acsmedchemlett.3c00422] [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: 09/19/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023] Open
Abstract
Novel bacterial topoisomerase inhibitors (NBTIs) make up a promising new class of antibiotics with the potential to combat the growing threat of antimicrobial resistance. Two key challenges in the development of NBTIs have been to obtain broad spectrum activity against multidrug-resistant Gram-negative bacteria and to diminish inhibition of the hERG cardiac ion channel. Here we report the optimization of a series of NBTIs bearing a novel indane DNA intercalating moiety. The addition of a basic, polar side chain connected to the indane by an ether or an N-linked secondary amide linkage together with a lipophilicity-lowering modification of the enzyme binding moiety led to compounds such as 2a and 2g which showed excellent broad spectrum potency and minimal hERG inhibition. Compound 2a demonstrated robust bactericidal in vivo activity in a mouse lung infection model with the strain P. aeruginosa ATCC 27853 which is resistant to several clinically relevant antibiotics. Rodent pharmacokinetic studies with 2a revealed an unusual profile characterized by rapid tissue distribution and a prolonged, flat terminal phase. This profile precluded further development of these compounds as potential new antibiotics.
Collapse
Affiliation(s)
- John G. Cumming
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Lukas Kreis
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Holger Kühne
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Roger Wermuth
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Maarten Vercruysse
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Carina Cantrill
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Caterina Bissantz
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Hongxia Qiu
- China
Innovation Center of Roche, Roche R&D
Center (China) Ltd., Shanghai 201203, China
| | - Christian Kramer
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | | | | |
Collapse
|
6
|
Cumming JG, Kreis L, Kühne H, Wermuth R, Vercruysse M, Kramer C, Rudolph MG, Xu Z. Discovery of a Series of Indane-Containing NBTIs with Activity against Multidrug-Resistant Gram-Negative Pathogens. ACS Med Chem Lett 2023; 14:993-998. [PMID: 37465290 PMCID: PMC10350941 DOI: 10.1021/acsmedchemlett.3c00187] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/20/2023] [Indexed: 07/20/2023] Open
Abstract
The rise of multidrug-resistant (MDR) Gram-negative bacteria is a major global health problem necessitating the discovery of new classes of antibiotics. Novel bacterial topoisomerase inhibitors (NBTIs) target the clinically validated bacterial type II topoisomerases with a distinct binding site and mechanism of action to fluoroquinolone antibiotics, thus avoiding cross-resistance to this drug class. Here we report the discovery of a series of NBTIs incorporating a novel indane DNA binding moiety. X-ray cocrystal structures of compounds 2 and 17a bound to Staphylococcus aureus DNA gyrase-DNA were determined, revealing specific interactions with the enzyme binding pocket at the GyrA dimer interface and a long-range electrostatic interaction between the basic amine in the linker and the carboxylate of Asp83. Exploration of the structure-activity relationship within the series led to the identification of lead compound 18c, which showed potent broad-spectrum activity against a panel of MDR Gram-negative bacteria.
Collapse
Affiliation(s)
- John G. Cumming
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Lukas Kreis
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Holger Kühne
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Roger Wermuth
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Maarten Vercruysse
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Christian Kramer
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Markus G. Rudolph
- Roche
Pharma Research & Early Development, F. Hoffmann-La Roche Ltd., CH-4070 Basel, Switzerland
| | - Zhiheng Xu
- China
Innovation Center of Roche, Roche R&D
Center (China) Ltd., Shanghai 201203, China
| |
Collapse
|
7
|
Bhowmick J, Nag M, Ghosh P, Rajmani RS, Chatterjee R, Karmakar K, Chandra K, Chatterjee J, Chakravortty D, Varadarajan R. A CcdB toxin-derived peptide acts as a broad-spectrum antibacterial therapeutic in infected mice. EMBO Rep 2023; 24:e55338. [PMID: 37166011 PMCID: PMC10328072 DOI: 10.15252/embr.202255338] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 04/16/2023] [Accepted: 04/21/2023] [Indexed: 05/12/2023] Open
Abstract
The bacterial toxin CcdB (Controller of Cell death or division B) targets DNA Gyrase, an essential bacterial topoisomerase, which is also the molecular target for fluoroquinolones. Here, we present a short cell-penetrating 24-mer peptide, CP1-WT, derived from the Gyrase-binding region of CcdB and examine its effect on growth of Escherichia coli, Salmonella Typhimurium, Staphylococcus aureus and a carbapenem- and tigecycline-resistant strain of Acinetobacter baumannii in both axenic cultures and mouse models of infection. The CP1-WT peptide shows significant improvement over ciprofloxacin in terms of its in vivo therapeutic efficacy in treating established infections of S. Typhimurium, S. aureus and A. baumannii. The molecular mechanism likely involves inhibition of Gyrase or Topoisomerase IV, depending on the strain used. The study validates the CcdB binding site on bacterial DNA Gyrase as a viable and alternative target to the fluoroquinolone binding site.
Collapse
Grants
- Department of Biotechnology, Ministry of Science and Technology, India - Indian Institute of Science (DBT-IISc) partnership program
- BT/COE/34/SP15219/2015 Department of Biotechnology, Ministry of Science and Technology, India
- DT.20/11/2015 Department of Biotechnology, Ministry of Science and Technology, India
- Department of Science and Technology, Ministry of Science and Technology, India (DST FIST)
- Ministry of Education, India (MHRD)
- University Grants Commission, Ministry of Education, India (UGC Centre for Advanced Studies)
- Department of Biotechnology, Ministry of Science and Technology, India
- Ministry of Education, India (MHRD)
Collapse
Affiliation(s)
- Jayantika Bhowmick
- Molecular Biophysics Unit (MBU)Indian Institute of ScienceBangaloreIndia
| | - Manish Nag
- Molecular Biophysics Unit (MBU)Indian Institute of ScienceBangaloreIndia
| | - Pritha Ghosh
- Molecular Biophysics Unit (MBU)Indian Institute of ScienceBangaloreIndia
| | - Raju S Rajmani
- Molecular Biophysics Unit (MBU)Indian Institute of ScienceBangaloreIndia
| | - Ritika Chatterjee
- Department of Microbiology and Cell BiologyIndian Institute of ScienceBangaloreIndia
| | - Kapudeep Karmakar
- Department of Microbiology and Cell BiologyIndian Institute of ScienceBangaloreIndia
| | - Kasturi Chandra
- Department of Microbiology and Cell BiologyIndian Institute of ScienceBangaloreIndia
| | - Jayanta Chatterjee
- Molecular Biophysics Unit (MBU)Indian Institute of ScienceBangaloreIndia
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell BiologyIndian Institute of ScienceBangaloreIndia
- School of BiologyIndian Institute of Science Education and Research Thiruvananthapuram (IISER TVM)ThiruvananthapuramIndia
| | | |
Collapse
|
8
|
Chen A, Dellos-Nolan S, Lu Y, West JS, Wozniak DJ, Mitton-Fry MJ. Dioxane-Linked Novel Bacterial Topoisomerase Inhibitors Exhibit Bactericidal Activity against Planktonic and Biofilm Staphylococcus aureus In Vitro. Microbiol Spectr 2022; 10:e0205622. [PMID: 36250857 PMCID: PMC9769912 DOI: 10.1128/spectrum.02056-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/28/2022] [Indexed: 01/06/2023] Open
Abstract
The development of novel treatments for Staphylococcus aureus infections remains a high priority worldwide. We previously reported compounds 0147 and 0186, novel bacterial topoisomerase inhibitors (NBTIs) with potent antibacterial activity against S. aureus, including methicillin-resistant S. aureus. Here, we further investigated the in vitro activity of 0147 and 0186 against S. aureus ATCC 29213. Both compounds demonstrated bactericidal activity against planktonic and biofilm S. aureus, which then translated into significant inhibition of biofilm formation. Combinations of NBTIs and glycopeptides yielded indifferent interactions against planktonic S. aureus, but several had synergistic effects against S. aureus biofilms. This work reinforces the potential of NBTIs as future therapeutics for S. aureus infections. IMPORTANCE The pathogen Staphylococcus aureus contributes substantially to infection-related mortality. Biofilms render bacteria more recalcitrant to antibacterial therapy. The manuscript describes the potent activity of a new class of antibacterial agents against both planktonic and biofilm populations of Staphylococcus aureus.
Collapse
Affiliation(s)
- Anna Chen
- Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sheri Dellos-Nolan
- Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Yanran Lu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jason S. West
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Daniel J. Wozniak
- Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio, USA
- Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Mark J. Mitton-Fry
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
9
|
Why Matter Matters: Fast-Tracking Mycobacterium abscessus Drug Discovery. Molecules 2022; 27:molecules27206948. [PMID: 36296540 PMCID: PMC9608607 DOI: 10.3390/molecules27206948] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Unlike Tuberculosis (TB), Mycobacterium abscessus lung disease is a highly drug-resistant bacterial infection with no reliable treatment options. De novo M. abscessus drug discovery is urgently needed but is hampered by the bacterium's extreme drug resistance profile, leaving the current drug pipeline underpopulated. One proposed strategy to accelerate de novo M. abscessus drug discovery is to prioritize screening of advanced TB-active compounds for anti-M. abscessus activity. This approach would take advantage of the greater chance of homologous drug targets between mycobacterial species, increasing hit rates. Furthermore, the screening of compound series with established structure-activity-relationship, pharmacokinetic, and tolerability properties should fast-track the development of in vitro anti-M. abscessus hits into lead compounds with in vivo efficacy. In this review, we evaluated the effectiveness of this strategy by examining the literature. We found several examples where the screening of advanced TB chemical matter resulted in the identification of anti-M. abscessus compounds with in vivo proof-of-concept, effectively populating the M. abscessus drug pipeline with promising new candidates. These reports validate the screening of advanced TB chemical matter as an effective means of fast-tracking M. abscessus drug discovery.
Collapse
|
10
|
Spiropyrimidinetriones: a Class of DNA Gyrase Inhibitors with Activity against Mycobacterium tuberculosis and without Cross-Resistance to Fluoroquinolones. Antimicrob Agents Chemother 2022; 66:e0219221. [PMID: 35266826 PMCID: PMC9017349 DOI: 10.1128/aac.02192-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Described here is a series of spiropyrimidinetrione (SPT) compounds with activity against Mycobacterium tuberculosis through inhibition of DNA gyrase. The SPT class operates via a novel mode of inhibition, which involves Mg2+-independent stabilization of the DNA cleavage complex with DNA gyrase and is thereby not cross-resistant with other DNA gyrase-inhibiting antibacterials, including fluoroquinolones. Compound 22 from the series was profiled broadly and showed in vitro cidality as well as intracellular activity against M. tuberculosis in macrophages. Evidence for the DNA gyrase mode of action was supported by inhibition of the target in a DNA supercoiling assay and elicitation of an SOS response seen in a recA reporter strain of M. tuberculosis. Pharmacokinetic properties of 22 supported evaluation of efficacy in an acute model of M. tuberculosis infection, where modest reduction in CFU numbers was seen. This work offers promise for deriving a novel drug class of tuberculosis agent without preexisting clinical resistance.
Collapse
|
11
|
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
|
12
|
A Mycobacterium tuberculosis NBTI DNA Gyrase Inhibitor Is Active against Mycobacterium abscessus. Antimicrob Agents Chemother 2021; 65:e0151421. [PMID: 34606340 PMCID: PMC8597734 DOI: 10.1128/aac.01514-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Fluoroquinolones-the only clinically used DNA gyrase inhibitors-are effective against tuberculosis (TB) but are in limited clinical use for nontuberculous mycobacteria (NTM) lung infections due to intrinsic drug resistance. We sought to test alternative DNA gyrase inhibitors for anti-NTM activity. Mycobacterium tuberculosis gyrase inhibitors (MGIs), a subclass of novel bacterial topoisomerase inhibitors (NBTIs), were recently shown to be active against the tubercle bacillus. Here, we show that the MGI EC/11716 not only has potent anti-tubercular activity but is active against M. abscessus and M. avium in vitro. Focusing on M. abscessus, which causes the most difficult to cure NTM disease, we show that EC/11716 is bactericidal, active against drug-tolerant biofilms, and efficacious in a murine model of M. abscessus lung infection. Based on resistant mutant selection experiments, we report a low frequency of resistance to EC/11716 and confirm DNA gyrase as its target. Our findings demonstrate the potential of NBTIs as anti-M. abscessus and possibly broad-spectrum anti-mycobacterial agents.
Collapse
|
13
|
Lu Y, Vibhute S, Li L, Okumu A, Ratigan SC, Nolan S, Papa JL, Mann CA, English A, Chen A, Seffernick JT, Koci B, Duncan LR, Roth B, Cummings JE, Slayden RA, Lindert S, McElroy CA, Wozniak DJ, Yalowich J, Mitton-Fry MJ. Optimization of TopoIV Potency, ADMET Properties, and hERG Inhibition of 5-Amino-1,3-dioxane-Linked Novel Bacterial Topoisomerase Inhibitors: Identification of a Lead with In Vivo Efficacy against MRSA. J Med Chem 2021; 64:15214-15249. [PMID: 34614347 DOI: 10.1021/acs.jmedchem.1c01250] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel bacterial topoisomerase inhibitors (NBTIs) are among the most promising new antibiotics in preclinical/clinical development. We previously reported dioxane-linked NBTIs with potent antistaphylococcal activity and reduced hERG inhibition, a key safety liability. Herein, polarity-focused optimization enabled the delineation of clear structure-property relationships for both microsomal metabolic stability and hERG inhibition, resulting in the identification of lead compound 79. This molecule demonstrates potent antibacterial activity against diverse Gram-positive pathogens, inhibition of both DNA gyrase and topoisomerase IV, a low frequency of resistance, a favorable in vitro cardiovascular safety profile, and in vivo efficacy in a murine model of methicillin-resistant Staphylococcus aureus infection.
Collapse
Affiliation(s)
- Yanran Lu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Sandip Vibhute
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Linsen Li
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Antony Okumu
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Steven C Ratigan
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Sheri Nolan
- Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jonathan L Papa
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chelsea A Mann
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Anthony English
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Anna Chen
- Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
| | - Justin T Seffernick
- Department of Chemistry and Biochemistry, College of Arts and Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Bryan Koci
- Eurofins Panlabs, St. Charles, Missouri 63304, United States
| | | | - Brieanna Roth
- JMI Laboratories, North Liberty, Iowa 52317, United States
| | - Jason E Cummings
- Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Richard A Slayden
- Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, College of Arts and Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Craig A McElroy
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Daniel J Wozniak
- Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States.,Department of Microbiology, College of Arts and Sciences, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jack Yalowich
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Mark J Mitton-Fry
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
14
|
Am Mucke H. Patent highlights August-September 2020. Pharm Pat Anal 2021; 10:1-7. [PMID: 33441018 DOI: 10.4155/ppa-2020-0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/18/2020] [Indexed: 11/17/2022]
Abstract
A snapshot of noteworthy recent developments in the patent literature of relevance to pharmaceutical and medical research and development.
Collapse
|
15
|
Rubab M, Chelliah R, Saravanakumar K, Barathikannan K, Wei S, Kim JR, Yoo D, Wang MH, Oh DH. Bioactive Potential of 2-Methoxy-4-vinylphenol and Benzofuran from Brassica oleracea L. var. capitate f, rubra (Red Cabbage) on Oxidative and Microbiological Stability of Beef Meat. Foods 2020; 9:E568. [PMID: 32375308 PMCID: PMC7278745 DOI: 10.3390/foods9050568] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/13/2020] [Accepted: 04/15/2020] [Indexed: 02/02/2023] Open
Abstract
In the future, plant based phytochemicals will be considered as efficient replacement sources of chemical preservatives, to act as potential bio-preservatives. We investigated the antibacterial and antioxidant activity of red cabbage (RC) extracts using different solvents. Among all extracts, chloroform extract exhibited strong antimicrobial and antioxidant activities. Hence, the phytochemical constitutions of the RC chloroform extract was examined by GC-MS analysis, and further, based on molecular docking analysis, revealed 2-Methoxy-4-vinylphenol and benzofuran as two major compounds found to be possessing higher degrees of interaction with DNA gyrase (4PLB; -8.63 Kcal.mol-1) and lipoprotein (LpxC-8.229 Kcal.mol-1), respectively, of the bacterial cell wall, which leads to higher antimicrobial efficacy. Further, it was confirmed with that the in vivo Caenorhabditis elegans model (but no cytotoxic effect) was exhibited in the MCF-7 cell line. Thus, we investigated the influence of this extract on the shelf life of meat under refrigeration storage. The physicochemical properties were observed periodically, and microbial analysis was conducted. The shelf life of the beef was enhanced (up to eight days) in terms of microbial and physiochemical properties, at 4 ± 2 °C when compared to control. We concluded that chloroform extract of RC has potential as a natural preservative in the meat processing industry.
Collapse
Affiliation(s)
- Momna Rubab
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 200-701, Korea; (M.R.); (R.C.); (K.B.); (J.-R.K.); (D.Y.)
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 200-701, Korea; (M.R.); (R.C.); (K.B.); (J.-R.K.); (D.Y.)
| | - Kandasamy Saravanakumar
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (M.-H.W.)
| | - Kaliyan Barathikannan
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 200-701, Korea; (M.R.); (R.C.); (K.B.); (J.-R.K.); (D.Y.)
| | - Shuai Wei
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China;
| | - Jong-Rae Kim
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 200-701, Korea; (M.R.); (R.C.); (K.B.); (J.-R.K.); (D.Y.)
- Hanmi Natural Nutrition Co., LTD 44-20, Tongil-ro 1888 beon-gil, Munsan, Paju, Gyeonggi 10808, Korea
| | - Daesang Yoo
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 200-701, Korea; (M.R.); (R.C.); (K.B.); (J.-R.K.); (D.Y.)
- H-FOOD, 108-66, 390 gil, Jingun Oh Nam-Ro, Nam Yang, Ju-Shi, Gyung Gi-Do 12041, Korea
| | - Myeong-Hyeon Wang
- Department of Medical Biotechnology, College of Biomedical Sciences, Kangwon National University, Chuncheon 200-701, Korea; (K.S.); (M.-H.W.)
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kangwon National University, Chuncheon 200-701, Korea; (M.R.); (R.C.); (K.B.); (J.-R.K.); (D.Y.)
| |
Collapse
|
16
|
Antibacterial activity of novel dual bacterial DNA type II topoisomerase inhibitors. PLoS One 2020; 15:e0228509. [PMID: 32074119 PMCID: PMC7029851 DOI: 10.1371/journal.pone.0228509] [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] [Received: 08/01/2019] [Accepted: 01/16/2020] [Indexed: 11/24/2022] Open
Abstract
In this study, a drug discovery programme that sought to identify novel dual bacterial topoisomerase II inhibitors (NBTIs) led to the selection of six optimized compounds. In enzymatic assays, the molecules showed equivalent dual-targeting activity against the DNA gyrase and topoisomerase IV enzymes of Staphylococcus aureus and Escherichia coli. Consistently, the compounds demonstrated potent activity in susceptibility tests against various Gram-positive and Gram-negative reference species, including ciprofloxacin-resistant strains. The activity of the compounds against clinical multidrug-resistant isolates of S. aureus, Clostridium difficile, Acinetobacter baumannii, Neisseria gonorrhoeae, E. coli and vancomycin-resistant Enterococcus spp. was also confirmed. Two compounds (1 and 2) were tested in time-kill and post-antibiotic effect (PAE) assays. Compound 1 was bactericidal against all tested reference strains and showed higher activity than ciprofloxacin, and compound 2 showed a prolonged PAE, even against the ciprofloxacin-resistant S. aureus BAA-1720 strain. Spontaneous development of resistance to both compounds was selected for in S. aureus at frequencies comparable to those obtained for quinolones and other NBTIs. S. aureus BAA-1720 mutants resistant to compounds 1 and 2 had single point mutations in gyrA or gyrB outside of the quinolone resistance-determining region (QRDR), confirming the distinct site of action of these NBTIs compared to that of quinolones. Overall, the very good antibacterial activity of the compounds and their optimizable in vitro safety and physicochemical profile may have relevant implications for the development of new broad-spectrum antibiotics.
Collapse
|
17
|
Gibson EG, Oviatt AA, Cacho M, Neuman KC, Chan PF, Osheroff N. Bimodal Actions of a Naphthyridone/Aminopiperidine-Based Antibacterial That Targets Gyrase and Topoisomerase IV. Biochemistry 2019; 58:4447-4455. [PMID: 31617352 PMCID: PMC7450530 DOI: 10.1021/acs.biochem.9b00805] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Gyrase and topoisomerase IV are the targets of fluoroquinolone antibacterials. However, the rise in antimicrobial resistance has undermined the clinical use of this important drug class. Therefore, it is critical to identify new agents that maintain activity against fluoroquinolone-resistant strains. One approach is to develop non-fluoroquinolone drugs that also target gyrase and topoisomerase IV but interact differently with the enzymes. This has led to the development of the "novel bacterial topoisomerase inhibitor" (NBTI) class of antibacterials. Despite the clinical potential of NBTIs, there is a relative paucity of data describing their mechanism of action against bacterial type II topoisomerases. Consequently, we characterized the activity of GSK126, a naphthyridone/aminopiperidine-based NBTI, against a variety of Gram-positive and Gram-negative bacterial type II topoisomerases, including gyrase from Mycobacterium tuberculosis and gyrase and topoisomerase IV from Bacillus anthracis and Escherichia coli. GSK126 enhanced single-stranded DNA cleavage and suppressed double-stranded cleavage mediated by these enzymes. It was also a potent inhibitor of gyrase-catalyzed DNA supercoiling and topoisomerase IV-catalyzed decatenation. Thus, GSK126 displays a similar bimodal mechanism of action across a variety of species. In contrast, GSK126 displayed a variable ability to overcome fluoroquinolone resistance mutations across these same species. Our results suggest that NBTIs elicit their antibacterial effects by two different mechanisms: inhibition of gyrase/topoisomerase IV catalytic activity or enhancement of enzyme-mediated DNA cleavage. Furthermore, the relative importance of these two mechanisms appears to differ from species to species. Therefore, we propose that the mechanistic basis for the antibacterial properties of NBTIs is bimodal in nature.
Collapse
Affiliation(s)
- Elizabeth G. Gibson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Alexandria A. Oviatt
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
| | - Monica Cacho
- Department of Diseases of the Developing World, GlaxoSmithKline, Parque Tecnológico de Madrid, Calle de Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain
| | - Keir C. Neuman
- Laboratory of Single Molecule Biophysics, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20982, United States
| | - Pan F. Chan
- Infectious Diseases Discovery, Medicines Opportunities Research Unit, GlaxoSmithKline, Collegeville, PA 19426, United States
| | - Neil Osheroff
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States
- Department of Medicine (Hematology/Oncology), Vanderbilt University School of Medicine, Nashville, TN 37232, United States
- VA Tennessee Valley Healthcare System, Nashville, TN 37212, United States
| |
Collapse
|
18
|
Rapid Evolution of Reduced Susceptibility against a Balanced Dual-Targeting Antibiotic through Stepping-Stone Mutations. Antimicrob Agents Chemother 2019; 63:AAC.00207-19. [PMID: 31235632 DOI: 10.1128/aac.00207-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/14/2019] [Indexed: 11/20/2022] Open
Abstract
Multitargeting antibiotics, i.e., single compounds capable of inhibiting two or more bacterial targets, are generally considered to be a promising therapeutic strategy against resistance evolution. The rationale for this theory is that multitargeting antibiotics demand the simultaneous acquisition of multiple mutations at their respective target genes to achieve significant resistance. The theory presumes that individual mutations provide little or no benefit to the bacterial host. Here, we propose that such individual stepping-stone mutations can be prevalent in clinical bacterial isolates, as they provide significant resistance to other antimicrobial agents. To test this possibility, we focused on gepotidacin, an antibiotic candidate that selectively inhibits both bacterial DNA gyrase and topoisomerase IV. In a susceptible organism, Klebsiella pneumoniae, a combination of two specific mutations in these target proteins provide an >2,000-fold reduction in susceptibility, while individually, none of these mutations affect resistance significantly. Alarmingly, strains with decreased susceptibility against gepotidacin are found to be as virulent as the wild-type Klebsiella pneumoniae strain in a murine model. Moreover, numerous pathogenic isolates carry mutations which could promote the evolution of clinically significant reduction of susceptibility against gepotidacin in the future. As might be expected, prolonged exposure to ciprofloxacin, a clinically widely employed gyrase inhibitor, coselected for reduced susceptibility against gepotidacin. We conclude that extensive antibiotic usage could select for mutations that serve as stepping-stones toward resistance against antimicrobial compounds still under development. Our research indicates that even balanced multitargeting antibiotics are prone to resistance evolution.
Collapse
|
19
|
Magarò G, Prati F, Garofalo B, Corso G, Furlotti G, Apicella C, Mangano G, D'Atanasio N, Robinson D, Di Giorgio FP, Ombrato R. Virtual Screening Approach and Investigation of Structure-Activity Relationships To Discover Novel Bacterial Topoisomerase Inhibitors Targeting Gram-Positive and Gram-Negative Pathogens. J Med Chem 2019; 62:7445-7472. [PMID: 31276392 DOI: 10.1021/acs.jmedchem.9b00394] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bacterial resistance is increasing rapidly, requiring urgent identification of new antibacterial drugs that are effective against multidrug-resistant pathogens. Novel bacterial topoisomerase inhibitors (NBTIs) provide a new strategy for investigating the well-validated DNA gyrase and topoisomerase IV targets while preventing cross-resistance issues. On this basis, starting from a virtual screening campaign and subsequent structure-based hit optimization guided by X-ray studies, a novel class of piperazine-like NBTIs with outstanding enzymatic activity against Staphylococcus aureus and Escherichia coli DNA gyrase and topoisomerase IV was identified. Notably, compounds (±)-33, (±)-35, and (±)-36 with potent and balanced multitarget enzymatic profiles exhibited excellent efficacy against selected Gram-positive and Gram-negative pathogens, as well as clinically relevant resistant strains. Overall, the new NBTI chemotype described herein, owing to the broad-spectrum antibacterial activity and favorable in vitro safety profile, might serve as a basis for the development of novel treatments against serious infections.
Collapse
Affiliation(s)
- Gabriele Magarò
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Federica Prati
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Barbara Garofalo
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Gaia Corso
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Guido Furlotti
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Claudia Apicella
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Giorgina Mangano
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Noemi D'Atanasio
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Daniel Robinson
- Schrodinger , 120 West 45th Street , New York , New York 10036 , United States
| | - Francesco Paolo Di Giorgio
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| | - Rosella Ombrato
- Angelini RR&D (Research, Regulatory & Development) , Angelini S.p.A. , Piazzale della Stazione SNC, S. Palomba-Pomezia , Rome 00071 , Italy
| |
Collapse
|
20
|
Yi L, Lü X. New Strategy on Antimicrobial-resistance: Inhibitors of DNA Replication Enzymes. Curr Med Chem 2019; 26:1761-1787. [PMID: 29110590 DOI: 10.2174/0929867324666171106160326] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/31/2017] [Accepted: 10/30/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Antimicrobial resistance is found in all microorganisms and has become one of the biggest threats to global health. New antimicrobials with different action mechanisms are effective weapons to fight against antibiotic-resistance. OBJECTIVE This review aims to find potential drugs which can be further developed into clinic practice and provide clues for developing more effective antimicrobials. METHODS DNA replication universally exists in all living organisms and is a complicated process in which multiple enzymes are involved in. Enzymes in bacterial DNA replication of initiation and elongation phases bring abundant targets for antimicrobial development as they are conserved and indispensable. In this review, enzyme inhibitors of DNA helicase, DNA primase, topoisomerases, DNA polymerase and DNA ligase were discussed. Special attentions were paid to structures, activities and action modes of these enzyme inhibitors. RESULTS Among these enzymes, type II topoisomerase is the most validated target with abundant inhibitors. For type II topoisomerase inhibitors (excluding quinolones), NBTIs and benzimidazole urea derivatives are the most promising inhibitors because of their good antimicrobial activity and physicochemical properties. Simultaneously, DNA gyrase targeted drugs are particularly attractive in the treatment of tuberculosis as DNA gyrase is the sole type II topoisomerase in Mycobacterium tuberculosis. Relatively, exploitation of antimicrobial inhibitors of the other DNA replication enzymes are primeval, in which inhibitors of topo III are even blank so far. CONCLUSION This review demonstrates that inhibitors of DNA replication enzymes are abundant, diverse and promising, many of which can be developed into antimicrobials to deal with antibioticresistance.
Collapse
Affiliation(s)
- Lanhua Yi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi Province 712100, China
| |
Collapse
|
21
|
Gibson EG, Bax B, Chan PF, Osheroff N. Mechanistic and Structural Basis for the Actions of the Antibacterial Gepotidacin against Staphylococcus aureus Gyrase. ACS Infect Dis 2019; 5:570-581. [PMID: 30757898 DOI: 10.1021/acsinfecdis.8b00315] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Gepotidacin is a first-in-class triazaacenaphthylene novel bacterial topoisomerase inhibitor (NBTI). The compound has successfully completed phase II trials for the treatment of acute bacterial skin/skin structure infections and for the treatment of uncomplicated urogenital gonorrhea. It also displays robust in vitro activity against a range of wild-type and fluoroquinolone-resistant bacteria. Due to the clinical promise of gepotidacin, a detailed understanding of its interactions with its antibacterial targets is essential. Thus, we characterized the mechanism of action of gepotidacin against Staphylococcus aureus gyrase. Gepotidacin was a potent inhibitor of gyrase-catalyzed DNA supercoiling (IC50 ≈ 0.047 μM) and relaxation of positively supercoiled substrates (IC50 ≈ 0.6 μM). Unlike fluoroquinolones, which induce primarily double-stranded DNA breaks, gepotidacin induced high levels of gyrase-mediated single-stranded breaks. No double-stranded breaks were observed even at high gepotidacin concentration, long cleavage times, or in the presence of ATP. Moreover, gepotidacin suppressed the formation of double-stranded breaks. Gepotidacin formed gyrase-DNA cleavage complexes that were stable for >4 h. In vitro competition suggests that gyrase binding by gepotidacin and fluoroquinolones are mutually exclusive. Finally, we determined crystal structures of gepotidacin with the S. aureus gyrase core fusion truncate with nicked (2.31 Å resolution) or intact (uncleaved) DNA (2.37 Å resolution). In both cases, a single gepotidacin molecule was bound midway between the two scissile DNA bonds and in a pocket between the two GyrA subunits. A comparison of the two structures demonstrates conformational flexibility within the central linker of gepotidacin, which may contribute to the activity of the compound.
Collapse
Affiliation(s)
| | - Ben Bax
- Medicines Discovery Institute, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, United Kingdom
| | - Pan F. Chan
- Infectious Diseases Discovery, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Neil Osheroff
- VA Tennessee Valley Healthcare System, 1310 24th Avenue S., Nashville, Tennessee 37212, United States
| |
Collapse
|
22
|
The Role of DNA Gyrase (gyrA) in Ciprofloxacin-Resistant Locally Isolates Pseudomonas aeruginosa in Al-Khadhmiya Teaching Hospital Baghdad, Iraq. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2019. [DOI: 10.22207/jpam.13.1.56] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
23
|
Gibson EG, Blower TR, Cacho M, Bax B, Berger JM, Osheroff N. Mechanism of Action of Mycobacterium tuberculosis Gyrase Inhibitors: A Novel Class of Gyrase Poisons. ACS Infect Dis 2018; 4:1211-1222. [PMID: 29746087 DOI: 10.1021/acsinfecdis.8b00035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tuberculosis is one of the leading causes of morbidity worldwide, and the incidences of drug resistance and intolerance are prevalent. Thus, there is a desperate need for the development of new antitubercular drugs. Mycobacterium tuberculosis gyrase inhibitors (MGIs) are napthyridone/aminopiperidine-based drugs that display activity against M. tuberculosis cells and tuberculosis in mouse models [Blanco, D., et al. (2015) Antimicrob. Agents Chemother. 59, 1868-1875]. Genetic and mutagenesis studies suggest that gyrase, which is the target for fluoroquinolone antibacterials, is also the target for MGIs. However, little is known regarding the interaction of these drugs with the bacterial type II enzyme. Therefore, we examined the effects of two MGIs, GSK000 and GSK325, on M. tuberculosis gyrase. MGIs greatly enhanced DNA cleavage mediated by the bacterial enzyme. In contrast to fluoroquinolones (which induce primarily double-stranded breaks), MGIs induced only single-stranded DNA breaks under a variety of conditions. MGIs work by stabilizing covalent gyrase-cleaved DNA complexes and appear to suppress the ability of the enzyme to induce double-stranded breaks. The drugs displayed little activity against type II topoisomerases from several other bacterial species, suggesting that these drugs display specificity for M. tuberculosis gyrase. Furthermore, MGIs maintained activity against M. tuberuclosis gyrase enzymes that contained the three most common fluoroquinolone resistance mutations seen in the clinic and displayed no activity against human topoisomerase IIα. These findings suggest that MGIs have potential as antitubercular drugs, especially in the case of fluoroquinolone-resistant disease.
Collapse
Affiliation(s)
| | - Tim R. Blower
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2185, United States
| | - Monica Cacho
- Department of Diseases of the Developing World, GlaxoSmithKline, Parque Tecnológico de Madrid, Calle de Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain
| | - Ben Bax
- Medicines Discovery Institute, Cardiff University, Cardiff CF10 3AT, United Kingdom
| | - James M. Berger
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205-2185, United States
| | - Neil Osheroff
- VA Tennessee Valley Healthcare System, Nashville, Tennessee 37212, United States
| |
Collapse
|
24
|
Kaguni JM. The Macromolecular Machines that Duplicate the Escherichia coli Chromosome as Targets for Drug Discovery. Antibiotics (Basel) 2018. [PMID: 29538288 PMCID: PMC5872134 DOI: 10.3390/antibiotics7010023] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
DNA replication is an essential process. Although the fundamental strategies to duplicate chromosomes are similar in all free-living organisms, the enzymes of the three domains of life that perform similar functions in DNA replication differ in amino acid sequence and their three-dimensional structures. Moreover, the respective proteins generally utilize different enzymatic mechanisms. Hence, the replication proteins that are highly conserved among bacterial species are attractive targets to develop novel antibiotics as the compounds are unlikely to demonstrate off-target effects. For those proteins that differ among bacteria, compounds that are species-specific may be found. Escherichia coli has been developed as a model system to study DNA replication, serving as a benchmark for comparison. This review summarizes the functions of individual E. coli proteins, and the compounds that inhibit them.
Collapse
Affiliation(s)
- Jon M Kaguni
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824-1319, USA.
| |
Collapse
|
25
|
van Eijk E, Wittekoek B, Kuijper EJ, Smits WK. DNA replication proteins as potential targets for antimicrobials in drug-resistant bacterial pathogens. J Antimicrob Chemother 2018; 72:1275-1284. [PMID: 28073967 PMCID: PMC5400081 DOI: 10.1093/jac/dkw548] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
With the impending crisis of antimicrobial resistance, there is an urgent need to develop novel antimicrobials to combat difficult infections and MDR pathogenic microorganisms. DNA replication is essential for cell viability and is therefore an attractive target for antimicrobials. Although several antimicrobials targeting DNA replication proteins have been developed to date, gyrase/topoisomerase inhibitors are the only class widely used in the clinic. Given the numerous essential proteins in the bacterial replisome that may serve as a potential target for inhibitors and the relative paucity of suitable compounds, it is evident that antimicrobials targeting the replisome are underdeveloped so far. In this review, we report on the diversity of antimicrobial compounds targeting DNA replication and highlight some of the challenges in developing new drugs that target this process.
Collapse
|
26
|
In Vivo Bioluminescent Monitoring of Therapeutic Efficacy and Pharmacodynamic Target Assessment of Antofloxacin against Escherichia coli in a Neutropenic Murine Thigh Infection Model. Antimicrob Agents Chemother 2017; 62:AAC.01281-17. [PMID: 29038275 DOI: 10.1128/aac.01281-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/16/2017] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial resistance among uropathogens has increased the rates of infection-related morbidity and mortality. Antofloxacin is a novel fluoroquinolone with broad-spectrum antibacterial activity against urinary Gram-negative bacilli, such as Escherichia coli This study monitored the in vivo efficacy of antofloxacin using bioluminescent imaging and determined pharmacokinetic (PK)/pharmacodynamic (PD) targets against E. coli isolates in a neutropenic murine thigh infection model. The PK properties were determined after subcutaneous administration of antofloxacin at 2.5, 10, 40, and 160 mg/kg of body weight. Following thigh infection, the mice were treated with 2-fold-increasing doses of antofloxacin from 2.5 to 80 mg/kg administered every 12 h. Efficacy was assessed by quantitative determination of the bacterial burdens in thigh homogenates and was compared with the bioluminescent density. Antofloxacin demonstrated both static and killing endpoints in relation to the initial burden against all study strains. The PK/PD index area under the concentration-time curve (AUC)/MIC correlated well with efficacy (R2 = 0.92), and the dose-response relationship was relatively steep, as observed with escalating doses of antofloxacin. The mean free drug AUC/MIC targets necessary to produce net bacterial stasis and 1-log10 and 2-log10 kill for each isolate were 38.7, 66.1, and 147.0 h, respectively. In vivo bioluminescent imaging showed a rapid decrease in the bioluminescent density at free drug AUC/MIC exposures that exceeded the stasis targets. The integration of these PD targets combined with the results of PK studies with humans will be useful in setting optimal dosing regimens for the treatment of urinary tract infections due to E. coli.
Collapse
|
27
|
Gepotidacin (GSK2140944) In Vitro Activity against Gram-Positive and Gram-Negative Bacteria. Antimicrob Agents Chemother 2017; 61:AAC.00468-17. [PMID: 28483959 DOI: 10.1128/aac.00468-17] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/04/2017] [Indexed: 01/31/2023] Open
Abstract
Gepotidacin is a first-in-class, novel triazaacenaphthylene antibiotic that inhibits bacterial DNA replication and has in vitro activity against susceptible and drug-resistant pathogens. Reference in vitro methods were used to investigate the MICs and minimum bactericidal concentrations (MBCs) of gepotidacin and comparator agents for Staphylococcus aureus, Streptococcus pneumoniae, and Escherichia coli Gepotidacin in vitro activity was also evaluated by using time-kill kinetics and broth microdilution checkerboard methods for synergy testing and for postantibiotic and subinhibitory effects. The MIC90 of gepotidacin for 50 S. aureus (including methicillin-resistant S. aureus [MRSA]) and 50 S. pneumoniae (including penicillin-nonsusceptible) isolates was 0.5 μg/ml, and for E. coli (n = 25 isolates), it was 4 μg/ml. Gepotidacin was bactericidal against S. aureus, S. pneumoniae, and E. coli, with MBC/MIC ratios of ≤4 against 98, 98, and 88% of the isolates tested, respectively. Time-kill curves indicated that the bactericidal activity of gepotidacin was observed at 4× or 10× MIC at 24 h for all of the isolates. S. aureus regrowth was observed in the presence of gepotidacin, and the resulting gepotidacin MICs were 2- to 128-fold higher than the baseline gepotidacin MICs. Checkerboard analysis of gepotidacin combined with other antimicrobials demonstrated no occurrences of antagonism with agents from multiple antimicrobial classes. The most common interaction when testing gepotidacin was indifference (fractional inhibitory concentration index of >0.5 to ≤4; 82.7% for Gram-positive isolates and 82.6% for Gram-negative isolates). The postantibiotic effect (PAE) of gepotidacin was short when it was tested against S. aureus (≤0.6 h against MRSA and MSSA), and the PAE-sub-MIC effect (SME) was extended (>8 h; three isolates at 0.5× MIC). The PAE of levofloxacin was modest (0.0 to 2.4 h), and the PAE-SME observed varied from 1.2 to >9 h at 0.5× MIC. These in vitro data indicate that gepotidacin is a bactericidal agent that exhibits a modest PAE and an extended PAE-SME against Gram-positive and -negative bacteria and merits further study for potential use in treating infections caused by these pathogens.
Collapse
|
28
|
Charrier C, Salisbury AM, Savage VJ, Duffy T, Moyo E, Chaffer-Malam N, Ooi N, Newman R, Cheung J, Metzger R, McGarry D, Pichowicz M, Sigerson R, Cooper IR, Nelson G, Butler HS, Craighead M, Ratcliffe AJ, Best SA, Stokes NR. Novel Bacterial Topoisomerase Inhibitors with Potent Broad-Spectrum Activity against Drug-Resistant Bacteria. Antimicrob Agents Chemother 2017; 61:e02100-16. [PMID: 28223393 PMCID: PMC5404544 DOI: 10.1128/aac.02100-16] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/27/2017] [Indexed: 12/30/2022] Open
Abstract
The novel bacterial topoisomerase inhibitor class is an investigational type of antibacterial inhibitor of DNA gyrase and topoisomerase IV that does not have cross-resistance with the quinolones. Here, we report the evaluation of the in vitro properties of a new series of this type of small molecule. Exemplar compounds selectively and potently inhibited the catalytic activities of Escherichia coli DNA gyrase and topoisomerase IV but did not block the DNA breakage-reunion step. Compounds showed broad-spectrum inhibitory activity against a wide range of Gram-positive and Gram-negative pathogens, including biodefence microorganisms and Mycobacterium tuberculosis No cross-resistance with fluoroquinolone-resistant Staphylococcus aureus and E. coli isolates was observed. Measured MIC90 values were 4 and 8 μg/ml against a panel of contemporary multidrug-resistant isolates of Acinetobacter baumannii and E. coli, respectively. In addition, representative compounds exhibited greater antibacterial potency than the quinolones against obligate anaerobic species. Spontaneous mutation rates were low, with frequencies of resistance typically <10-8 against E. coli and A. baumannii at concentrations equivalent to 4-fold the MIC. Compound-resistant E. coli mutants that were isolated following serial passage were characterized by whole-genome sequencing and carried a single Arg38Leu amino acid substitution in the GyrA subunit of DNA gyrase. Preliminary in vitro safety data indicate that the series shows a promising therapeutic index and potential for low human ether-a-go-go-related gene (hERG) inhibition (50% inhibitory concentration [IC50], >100 μM). In summary, the compounds' distinct mechanism of action relative to the fluoroquinolones, whole-cell potency, low potential for resistance development, and favorable in vitro safety profile warrant their continued investigation as potential broad-spectrum antibacterial agents.
Collapse
Affiliation(s)
| | | | | | - Thomas Duffy
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | - Emmanuel Moyo
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | | | - Nicola Ooi
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | | | | | | | - David McGarry
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | | | | | - Ian R Cooper
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | - Gary Nelson
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | | | | | | | - Stuart A Best
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| | - Neil R Stokes
- Redx Pharma, Alderley Park, Cheshire, United Kingdom
| |
Collapse
|
29
|
In Vitro Activity of Gepotidacin (GSK2140944) against Neisseria gonorrhoeae. Antimicrob Agents Chemother 2017; 61:AAC.02047-16. [PMID: 28069643 DOI: 10.1128/aac.02047-16] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/22/2016] [Indexed: 11/20/2022] Open
Abstract
Gepotidacin (formerly GSK2140944) is a novel, first-in-class, triazaacenaphthylene antibacterial that inhibits bacterial DNA gyrase and topoisomerase IV via a unique mechanism and has demonstrated in vitro activity against Neisseria gonorrhoeae, including drug-resistant strains, and also targets pathogens associated with other conventional and biothreat infections. Broth microdilution was used to evaluate the MIC and minimum bactericidal concentration (MBC) activity of gepotidacin and comparators against 25 N. gonorrhoeae strains (including five ciprofloxacin-nonsusceptible strains). Gepotidacin activity was also evaluated against three N. gonorrhoeae strains (including a ciprofloxacin-nonsusceptible strain) for resistance development, against three N. gonorrhoeae strains (including two tetracycline- and azithromycin-nonsusceptible strains) using time-kill kinetics and checkerboard methods, and against two N. gonorrhoeae strains for the investigation of postantibiotic (PAE) and subinhibitory (PAE-SME) effects. The MIC50 and MIC90 for gepotidacin against the 25 N. gonorrhoeae isolates tested were 0.12 and 0.25 μg/ml, respectively. The MBC50 and MBC90 for gepotidacin were 0.25 and 0.5 μg/ml, respectively. Gepotidacin was bactericidal, and single-step resistance selection studies did not recover any mutants, indicating a low rate of spontaneous single-step resistance. For combinations of gepotidacin and comparators tested using checkerboard methods, there were no instances where antagonism occurred and only one instance of synergy (with moxifloxacin; fractional inhibitory concentration, 0.375). This was not confirmed by in vitro time-kill studies. The PAE for gepotidacin against the wild-type strain ranged from 0.5 to >2.5 h, and the PAE-SME was >2.5 h. These in vitro data indicate that further study of gepotidacin is warranted for potential use in treating infections caused by N. gonorrhoeae.
Collapse
|
30
|
Bansal S, Bajaj P, Pandey S, Tandon V. Topoisomerases: Resistance versus Sensitivity, How Far We Can Go? Med Res Rev 2016; 37:404-438. [PMID: 27687257 DOI: 10.1002/med.21417] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/04/2016] [Accepted: 08/29/2016] [Indexed: 12/15/2022]
Abstract
DNA topoisomerases are ubiquitously present remarkable molecular machines that help in altering topology of DNA in living cells. The crucial role played by these nucleases during DNA replication, transcription, and recombination vis-à-vis less sequence similarity among different species makes topoisomerases unique and attractive targets for different anticancer and antibacterial drugs. However, druggability of topoisomerases by the existing class of molecules is increasingly becoming questationable due to resistance development predominated by mutations in the corresponding genes. The current scenario facing a decline in the development of new molecules further comprises an important factor that may challenge topoisomerase-targeting therapy. Thus, it is imperative to wisely use the existing inhibitors lest with this rapid rate of losing grip over the target we may not go too far. Furthermore, it is important not only to design new molecules but also to develop new approaches that may avoid obstacles in therapies due to multiple resistance mechanisms. This review provides a succinct account of different classes of topoisomerase inhibitors, focuses on resistance acquired by mutations in topoisomerases, and discusses the various approaches to increase the efficacy of topoisomerase inhibitors. In a later section, we also suggest the possibility of using bisbenzimidazoles along with efflux pump inhibitors for synergistic bactericidal effects.
Collapse
Affiliation(s)
- Sandhya Bansal
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, New Delhi, India
| | - Priyanka Bajaj
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India
| | - Stuti Pandey
- Department of Chemistry, University of Delhi, New Delhi, India
| | - Vibha Tandon
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.,Department of Chemistry, University of Delhi, New Delhi, India
| |
Collapse
|
31
|
Abstract
DNA gyrase and topoisomerase IV are type IIA bacterial topoisomerases that are targeted by highly effective antibiotics. However, resistance via multiple mechanisms arises to limit the efficacies of these drugs. Continued research on type IIA bacterial topoisomerases has provided novel approaches to counter the most common resistance mechanism for utilization of these proven targets in antibacterial therapy. Bacterial topoisomerase I is being explored as an alternative target that is not expected to show cross-resistance. Dual targeting or combination therapy could be strategies for circumventing the development of resistance to topoisomerase-targeting antibiotics. Bacterial topoisomerases are high-value bactericidal targets that could continue to be exploited for antibacterial therapy, if new tactics to counter resistance can be adopted.
Collapse
|
32
|
In Vivo Pharmacodynamic Target Investigation of Two Bacterial Topoisomerase Inhibitors, ACT-387042 and ACT-292706, in the Neutropenic Murine Thigh Model against Streptococcus pneumoniae and Staphylococcus aureus. Antimicrob Agents Chemother 2016; 60:3626-32. [PMID: 27044547 DOI: 10.1128/aac.00363-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 03/26/2016] [Indexed: 01/07/2023] Open
Abstract
ACT-387042 and ACT-292706 are two novel bacterial topoisomerase inhibitors with broad-spectrum activity against Gram-positive and -negative bacteria, including methicillin-resistant Staphylococcus aureus and penicillin- and fluoroquinolone-resistant Streptococcus pneumoniae We used the neutropenic murine thigh infection model to characterize the pharmacokinetics (PK)/pharmacodynamics (PD) of these investigational compounds against a group of 10 S. aureus and S. pneumoniae isolates with phenotypic resistance to beta-lactams and fluoroquinolones. The in vitro activities of the two compounds were very similar (MIC range, 0.03 to 0.125 mg/liter). Plasma pharmacokinetics were determined for each compound by using four escalating doses administered by the subcutaneous route. In treatment studies, mice had 10(7.4) to 10(8) CFU/thigh at the start of therapy with ACT-387042 and 10(6.7) to 10(8.3) CFU/thigh at the start of therapy with ACT-292706. A dose-response relationship was observed with all isolates over the dose range. Maximal kill approached 3 to 4 log10 CFU/thigh compared to the burden at the start of therapy for the highest doses examined. There was a strong relationship between the PK/PD index AUC/MIC ratio (area under the concentration-time curve over 24 h in the steady state divided by the MIC) and therapeutic efficacy in the model (R(2), 0.63 to 0.82). The 24-h free-drug AUC/MIC ratios associated with net stasis for ACT-387042 against S. aureus and S. pneumoniae were 43 and 10, respectively. The 24-h free-drug AUC/MIC ratios associated with net stasis for ACT-292706 against S. aureus and S. pneumoniae were 69 and 25, respectively. The stasis PD targets were significantly lower for S. pneumoniae (P < 0.05) for both compounds. The 1-log-kill AUC/MIC ratio targets were ∼2- to 4-fold higher than stasis targets. Methicillin, penicillin, or ciprofloxacin resistance did not alter the magnitude of the AUC/MIC ratio required for efficacy. These results should be helpful in the design of clinical trials for topoisomerase inhibitors.
Collapse
|
33
|
Comparative Study of Activities of a Diverse Set of Antimycobacterial Agents against Mycobacterium tuberculosis and Mycobacterium ulcerans. Antimicrob Agents Chemother 2016; 60:3132-7. [PMID: 26883701 DOI: 10.1128/aac.02658-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 02/08/2016] [Indexed: 11/20/2022] Open
Abstract
A library of compounds covering a broad chemical space was selected from a tuberculosis drug development program and was screened in a whole-cell assay against Mycobacterium ulcerans, the causative agent of the necrotizing skin disease Buruli ulcer. While a number of potent antitubercular agents were only weakly active or inactive against M. ulcerans, five compounds showed high activity (90% inhibitory concentration [IC90], ≤1 μM), making screening of focused antitubercular libraries a good starting point for lead generation against M. ulcerans.
Collapse
|
34
|
Miles TJ, Hennessy AJ, Bax B, Brooks G, Brown BS, Brown P, Cailleau N, Chen D, Dabbs S, Davies DT, Esken JM, Giordano I, Hoover JL, Jones GE, Kusalakumari Sukmar SK, Markwell RE, Minthorn EA, Rittenhouse S, Gwynn MN, Pearson ND. Novel tricyclics (e.g., GSK945237) as potent inhibitors of bacterial type IIA topoisomerases. Bioorg Med Chem Lett 2016; 26:2464-2469. [PMID: 27055939 DOI: 10.1016/j.bmcl.2016.03.106] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 03/28/2016] [Accepted: 03/29/2016] [Indexed: 01/06/2023]
Abstract
During the course of our research on the lead optimisation of the NBTI (Novel Bacterial Type II Topoisomerase Inhibitors) class of antibacterials, we discovered a series of tricyclic compounds that showed good Gram-positive and Gram-negative potency. Herein we will discuss the various subunits that were investigated in this series and report advanced studies on compound 1 (GSK945237) which demonstrates good PK and in vivo efficacy properties.
Collapse
Affiliation(s)
- Timothy J Miles
- Diseases of the Developing World CEDD, GlaxoSmithKline, Calle Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain.
| | - Alan J Hennessy
- Infectious Diseases CEDD, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - Ben Bax
- Platform Technology & Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - Gerald Brooks
- Infectious Diseases CEDD, GlaxoSmithKline, Third Avenue, Harlow CM19 5AW, UK
| | - Barry S Brown
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Pamela Brown
- Infectious Diseases CEDD, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - Nathalie Cailleau
- Infectious Diseases CEDD, GlaxoSmithKline, Third Avenue, Harlow CM19 5AW, UK
| | - Dongzhao Chen
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Steven Dabbs
- Infectious Diseases CEDD, GlaxoSmithKline, Gunnels Wood Road, Stevenage SG1 2NY, UK
| | - David T Davies
- Infectious Diseases CEDD, GlaxoSmithKline, Third Avenue, Harlow CM19 5AW, UK
| | - Joel M Esken
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Ilaria Giordano
- Diseases of the Developing World CEDD, GlaxoSmithKline, Calle Severo Ochoa, 2, 28760 Tres Cantos, Madrid, Spain
| | - Jennifer L Hoover
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Graham E Jones
- Infectious Diseases CEDD, GlaxoSmithKline, Third Avenue, Harlow CM19 5AW, UK
| | | | - Roger E Markwell
- Infectious Diseases CEDD, GlaxoSmithKline, Third Avenue, Harlow CM19 5AW, UK
| | - Elisabeth A Minthorn
- Oncology TA, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Steve Rittenhouse
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Michael N Gwynn
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| | - Neil D Pearson
- Infectious Diseases CEDD, GlaxoSmithKline, 1250 South Collegeville Road, Collegeville, PA 19426, USA
| |
Collapse
|
35
|
Wagner S, Sommer R, Hinsberger S, Lu C, Hartmann RW, Empting M, Titz A. Novel Strategies for the Treatment of Pseudomonas aeruginosa Infections. J Med Chem 2016; 59:5929-69. [DOI: 10.1021/acs.jmedchem.5b01698] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Stefanie Wagner
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| | - Roman Sommer
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| | - Stefan Hinsberger
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Cenbin Lu
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Rolf W. Hartmann
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Martin Empting
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
- Drug
Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
| | - Alexander Titz
- Chemical
Biology of Carbohydrates, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), D-66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 30625 Standort Hannover-Braunschweig, Germany
| |
Collapse
|
36
|
Klahn P, Brönstrup M. New Structural Templates for Clinically Validated and Novel Targets in Antimicrobial Drug Research and Development. Curr Top Microbiol Immunol 2016; 398:365-417. [PMID: 27704270 DOI: 10.1007/82_2016_501] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of bacterial resistance against current antibiotic drugs necessitates a continuous renewal of the arsenal of efficacious drugs. This imperative has not been met by the output of antibiotic research and development of the past decades for various reasons, including the declining efforts of large pharma companies in this area. Moreover, the majority of novel antibiotics are chemical derivatives of existing structures that represent mostly step innovations, implying that the available chemical space may be exhausted. This review negates this impression by showcasing recent achievements in lead finding and optimization of antibiotics that have novel or unexplored chemical structures. Not surprisingly, many of the novel structural templates like teixobactins, lysocin, griselimycin, or the albicidin/cystobactamid pair were discovered from natural sources. Additional compounds were obtained from the screening of synthetic libraries and chemical synthesis, including the gyrase-inhibiting NTBI's and spiropyrimidinetrione, the tarocin and targocil inhibitors of wall teichoic acid synthesis, or the boronates and diazabicyclo[3.2.1]octane as novel β-lactamase inhibitors. A motif that is common to most clinically validated antibiotics is that they address hotspots in complex biosynthetic machineries, whose functioning is essential for the bacterial cell. Therefore, an introduction to the biological targets-cell wall synthesis, topoisomerases, the DNA sliding clamp, and membrane-bound electron transport-is given for each of the leads presented here.
Collapse
Affiliation(s)
- Philipp Klahn
- 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.
| |
Collapse
|
37
|
Mutant Alleles of lptD Increase the Permeability of Pseudomonas aeruginosa and Define Determinants of Intrinsic Resistance to Antibiotics. Antimicrob Agents Chemother 2015; 60:845-54. [PMID: 26596941 DOI: 10.1128/aac.01747-15] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 11/15/2015] [Indexed: 01/11/2023] Open
Abstract
Gram-negative bacteria provide a particular challenge to antibacterial drug discovery due to their cell envelope structure. Compound entry is impeded by the lipopolysaccharide (LPS) of the outer membrane (OM), and those molecules that overcome this barrier are often expelled by multidrug efflux pumps. Understanding how efflux and permeability affect the ability of a compound to reach its target is paramount to translating in vitro biochemical potency to cellular bioactivity. Herein, a suite of Pseudomonas aeruginosa strains were constructed in either a wild-type or efflux-null background in which mutations were engineered in LptD, the final protein involved in LPS transport to the OM. These mutants were demonstrated to be defective in LPS transport, resulting in compromised barrier function. Using isogenic strain sets harboring these newly created alleles, we were able to define the contributions of permeability and efflux to the intrinsic resistance of P. aeruginosa to a variety of antibiotics. These strains will be useful in the design and optimization of future antibiotics against Gram-negative pathogens.
Collapse
|
38
|
Insights into the mechanism of inhibition of novel bacterial topoisomerase inhibitors from characterization of resistant mutants of Staphylococcus aureus. Antimicrob Agents Chemother 2015; 59:5278-87. [PMID: 26077256 DOI: 10.1128/aac.00571-15] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 06/09/2015] [Indexed: 11/20/2022] Open
Abstract
The type II topoisomerases DNA gyrase and topoisomerase IV are clinically validated bacterial targets that catalyze the modulation of DNA topology that is vital to DNA replication, repair, and decatenation. Increasing resistance to fluoroquinolones, which trap the topoisomerase-DNA complex, has led to significant efforts in the discovery of novel inhibitors of these targets. AZ6142 is a member of the class of novel bacterial topoisomerase inhibitors (NBTIs) that utilizes a distinct mechanism to trap the protein-DNA complex. AZ6142 has very potent activity against Gram-positive organisms, including Staphylococcus aureus, Streptococcus pneumoniae, and Streptococcus pyogenes. In this study, we determined the frequencies of resistance to AZ6142 and other representative NBTI compounds in S. aureus and S. pneumoniae. The frequencies of selection of resistant mutants at 4× the MIC were 1.7 × 10(-8) for S. aureus and <5.5 × 10(-10) for S. pneumoniae. To improve our understanding of the NBTI mechanism of inhibition, the resistant S. aureus mutants were characterized and 20 unique substitutions in the topoisomerase subunits were identified. Many of these substitutions were located outside the NBTI binding pocket and impact the susceptibility of AZ6142, resulting in a 4- to 32-fold elevation in the MIC over the wild-type parent strain. Data on cross-resistance with other NBTIs and fluoroquinolones enabled the differentiation of scaffold-specific changes from compound-specific variations. Our results suggest that AZ6142 inhibits both type II topoisomerases in S. aureus but that DNA gyrase is the primary target. Further, the genotype of the resistant mutants suggests that domain conformations and DNA interactions may uniquely impact NBTIs compared to fluoroquinolones.
Collapse
|
39
|
Target-based resistance in Pseudomonas aeruginosa and Escherichia coli to NBTI 5463, a novel bacterial type II topoisomerase inhibitor. Antimicrob Agents Chemother 2014; 59:331-7. [PMID: 25348539 DOI: 10.1128/aac.04077-14] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
In a previous report (T. J. Dougherty, A. Nayar, J. V. Newman, S. Hopkins, G. G. Stone, M. Johnstone, A. B. Shapiro, M. Cronin, F. Reck, and D. E. Ehmann, Antimicrob Agents Chemother 58:2657-2664, 2014), a novel bacterial type II topoisomerase inhibitor, NBTI 5463, with activity against Gram-negative pathogens was described. First-step resistance mutations in Pseudomonas aeruginosa arose exclusively in the nfxB gene, a regulator of the MexCD-OprJ efflux pump system. The present report describes further resistance studies with NBTI 5463 in both Pseudomonas aeruginosa and Escherichia coli. Second-step mutations in P. aeruginosa arose at aspartate 82 of the gyrase A subunit and led to 4- to 8-fold increases in the MIC over those seen in the parental strain with a first-step nfxB efflux mutation. A third-step mutant showed additional GyrA changes, with no changes in topoisomerase IV. Despite repeated efforts, resistance mutations could not be selected in E. coli. Genetic introduction of the Asp82 mutations observed in P. aeruginosa did not significantly increase the NBTI MIC in E. coli. However, with the aspartate 82 mutation present, it was possible to select second-step mutations in topoisomerase IV that did lead to MIC increases of 16- and 128-fold. As with the gyrase aspartate 82 mutation, the mutations in topoisomerase IV did not by themselves raise the NBTI MIC in E. coli. Only the presence of mutations in both targets of E. coli led to an increase in NBTI MIC values. This represents a demonstration of the value of balanced dual-target activity in mitigating resistance development.
Collapse
|
40
|
Ehmann DE, Lahiri SD. Novel compounds targeting bacterial DNA topoisomerase/DNA gyrase. Curr Opin Pharmacol 2014; 18:76-83. [PMID: 25271174 DOI: 10.1016/j.coph.2014.09.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 09/12/2014] [Indexed: 02/08/2023]
Abstract
Among the targets for the development of new antibacterial agents, bacterial topoisomerases remain a vibrant area of discovery. A structurally diverse set of inhibitors that bind to the adenosine 5'-triphosphate (ATP) site of type II topoisomerases have been disclosed recently. Seven compounds with this mechanism are highlighted, focusing on antibacterial potency and spectrum, as well as examples of in vivo efficacy against pathogens including Staphylococcus aureus and Mycobacterium tuberculosis. Five compounds from two structural classes are exemplified that are inhibitors that bind to the catalytic site of DNA gyrase and topoisomerase IV. The pharmacokinetic and pharmacodynamic properties of these molecules, derived from in vivo efficacy against Gram-positive and Gram-negative pathogens, define the potential for these agents with broad-spectrum and targeted-spectrum clinical utilities.
Collapse
Affiliation(s)
- David E Ehmann
- Infection Innovative Medicines Unit, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, MA 02451, USA.
| | - Sushmita D Lahiri
- Infection Innovative Medicines Unit, AstraZeneca R&D Boston, 35 Gatehouse Drive, Waltham, MA 02451, USA
| |
Collapse
|