An Iterative Approach Guides Discovery of the FabI Inhibitor Fabimycin, a Late-Stage Antibiotic Candidate with In Vivo Efficacy against Drug-Resistant Gram-Negative InfectionsClick to copy article linkArticle link copied!
- Erica N. ParkerErica N. ParkerDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Erica N. Parker
- Brett N. CainBrett N. CainDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Brett N. Cain
- Behnoush HajianBehnoush HajianBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Behnoush Hajian
- Rebecca J. UlrichRebecca J. UlrichDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Rebecca J. Ulrich
- Emily J. GeddesEmily J. GeddesDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Emily J. Geddes
- Sulyman BarkhoSulyman BarkhoBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Sulyman Barkho
- Hyang Yeon LeeHyang Yeon LeeDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Hyang Yeon Lee
- John D. WilliamsJohn D. WilliamsWalter Reed Army Institute of Research, Silver Spring, Maryland 20910 United StatesMore by John D. Williams
- Malik RaynorMalik RaynorWalter Reed Army Institute of Research, Silver Spring, Maryland 20910 United StatesMore by Malik Raynor
- Diana CaridhaDiana CaridhaWalter Reed Army Institute of Research, Silver Spring, Maryland 20910 United StatesMore by Diana Caridha
- Angela ZainoAngela ZainoBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Angela Zaino
- Mrinal ShekharMrinal ShekharBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Mrinal Shekhar
- Kristen A. MuñozKristen A. MuñozDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Kristen A. Muñoz
- Kara M. RzasaKara M. RzasaBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Kara M. Rzasa
- Emily R. TempleEmily R. TempleBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Emily R. Temple
- Diana HuntDiana HuntBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesDepartment of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United StatesMore by Diana Hunt
- Xiannu JinXiannu JinWalter Reed Army Institute of Research, Silver Spring, Maryland 20910 United StatesMore by Xiannu Jin
- Chau VuongChau VuongWalter Reed Army Institute of Research, Silver Spring, Maryland 20910 United StatesMore by Chau Vuong
- Kristina PannoneKristina PannoneWalter Reed Army Institute of Research, Silver Spring, Maryland 20910 United StatesMore by Kristina Pannone
- Aya M. KellyAya M. KellyDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Aya M. Kelly
- Michael P. MulliganMichael P. MulliganDepartment of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Michael P. Mulligan
- Katie K. LeeKatie K. LeeBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesMore by Katie K. Lee
- Gee W. LauGee W. LauDepartment of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Gee W. Lau
- Deborah T. HungDeborah T. HungBroad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United StatesDepartment of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02115, United StatesMore by Deborah T. Hung
- Paul J. Hergenrother*Paul J. Hergenrother*E-mail: [email protected]Department of Chemistry and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana−Champaign, Urbana, Illinois 61801, United StatesMore by Paul J. Hergenrother
Abstract
Genomic studies and experiments with permeability-deficient strains have revealed a variety of biological targets that can be engaged to kill Gram-negative bacteria. However, the formidable outer membrane and promiscuous efflux pumps of these pathogens prevent many candidate antibiotics from reaching these targets. One such promising target is the enzyme FabI, which catalyzes the rate-determining step in bacterial fatty acid biosynthesis. Notably, FabI inhibitors have advanced to clinical trials for Staphylococcus aureus infections but not for infections caused by Gram-negative bacteria. Here, we synthesize a suite of FabI inhibitors whose structures fit permeation rules for Gram-negative bacteria and leverage activity against a challenging panel of Gram-negative clinical isolates as a filter for advancement. The compound to emerge, called fabimycin, has impressive activity against >200 clinical isolates of Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii, and does not kill commensal bacteria. X-ray structures of fabimycin in complex with FabI provide molecular insights into the inhibition. Fabimycin demonstrates activity in multiple mouse models of infection caused by Gram-negative bacteria, including a challenging urinary tract infection model. Fabimycin has translational promise, and its discovery provides additional evidence that antibiotics can be systematically modified to accumulate in Gram-negative bacteria and kill these problematic pathogens.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Synopsis
Fabimycin is a bacterial FabI inhibitor that possesses potent antibiotic activity against multiple ESKAPE pathogens and efficacy in several mouse infection models, while largely sparing human commensal bacteria.
Introduction
Results
Identification of Fabimycin and Its Activity against Drug-Resistant Gram-Negative Pathogens
MIC experiments with commensal bacteria and pathogenic bacteria possessing alternate enoyl-acyl carrier protein reductases (29,31,36) indicate these strains are not susceptible to fabimycin.
Mode of Action
Crystallography, Molecular Dynamics, and Biophysics
In Vivo Experiments
Discussion
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.2c00598.
Supporting figures and tables, materials and methods, experimental procedures, and characterization of chemical products including LCMS traces and NMR (1H, 13C) spectra (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
This work was supported by the University of Illinois and the NIH (AI136773 to P.J.H. and G.W.L.). K.A.M. and M.P.M. are members of the NIH Chemistry–Biology Interface Training Grant (T32-GM136629). R.J.U. is supported by an NIH Ruth Kirschstein Award (F31AI161953) and was an NSF predoctoral fellow. This work utilized NIAID’s suite of preclinical services for in vitro and in vivo assessment (Contract Numbers HHSN272201700020I_75N93021F00001, HHSN272201700020I_75N93021F00002, and 75N93019D00022_75N93020F00001). The Collaborative Hub for Early Antibiotic Discovery (CHEAD) is grateful for financial support from the Combating Antibiotic-Resistant Bacteria Biopharmaceutical Accelerator (CARB-X). This work was supported by a gift from Anita and Josh Bekenstein and by funding provided under Cooperative Agreement #IDSEP160030-01-00 from Biomedical Advanced Research and Development Authority (BARDA). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of The Assistant Secretary for Preparedness and Response. This project has been funded in part by federal funds from the Military Infectious Disease Research Program Proposals MI210200 and MI220020. We thank L. Li (Metabolomics Center, Roy J. Carver Biotechnology Center, UIUC) for LC-MS/MS analysis. We thank Dean Olson, Lingyang Zhu, and N. Duay at the School of Chemical Sciences NMR Laboratory at UIUC for NMR services. The Bruker 500 MHz NMR spectrometer was obtained with the financial support of the Roy J. Carver Charitable Trust, Muscatine, Iowa.
References
This article references 53 other publications.
- 1Konychev, A.; Heep, M.; Moritz, R. K.; Kreuter, A.; Shulutko, A.; Fierlbeck, G.; Bouylout, K.; Pathan, R.; Trostmann, U.; Chaves, R. L. Safety and efficacy of daptomycin as first-line treatment for complicated skin and soft tissue infections in elderly patients: an open-label, multicentre, randomized phase IIIb trial. Drugs Aging 2013, 30 (10), 829– 836, DOI: 10.1007/s40266-013-0114-8Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVWmt7bE&md5=51940b8a92bebe81aef5707dce85bf5dSafety and Efficacy of Daptomycin as First-Line Treatment for Complicated Skin and Soft Tissue Infections in Elderly Patients: An Open-Label, Multicentre, Randomized Phase IIIb TrialKonychev, Alexander; Heep, Markus; Moritz, Rose K. C.; Kreuter, Alexander; Shulutko, Alexander; Fierlbeck, Gerhard; Bouylout, Kamel; Pathan, Rashidkhan; Trostmann, Uwe; Chaves, Ricardo L.Drugs & Aging (2013), 30 (10), 829-836CODEN: DRAGE6; ISSN:1170-229X. (Springer International Publishing AG)Background: Daptomycin has proven efficacy in patients with Gram-pos. complicated skin and soft tissue infections (cSSTIs), including those caused by Staphylococcus aureus, regardless of methicillin susceptibility. Objective: This study was undertaken to evaluate the efficacy and safety of daptomycin in elderly patients. Study Design: This was an open-label, multicentre, randomized phase IIIb study conducted in hospitalized patients Patients Patients aged ≥65 years with a diagnosis of Gram-pos. cSSTIs with or without bacteremia were included. In addn., infections were required to be of sufficient severity to require inpatient hospitalization and treatment with parenteral antibiotics for at least 96 h. The main exclusion criterion was the presence of a non-complicated SSTI that could heal by itself or be cured by surgical removal of the site of infection. Intervention: Patients were randomized 2:1 to i.v. daptomycin or pooled i.v. std. therapies semi-synthetic penicillin or vancomycin, referred to as the comparator. Duration of treatment was between 5 and 14 days for cSSTIs without bacteremia and between 10 and 28 days for cSSTIs with bacteremia. Main Outcome Measure: The primary objective was descriptive comparison of clin. success in clin. evaluable patients at test of cure, 7-14 days post treatment. Secondary objectives were microbiol. outcome, duration of treatment and safety. Results: In total, 120 patients were randomized (81 to daptomycin; 39 to the comparator) and 102 patients completed the study. Baseline characteristics were similar between the two groups. Common infections included cellulitis, ulcers and abscesses; six patients had bacteremia [five documented (daptomycin, n = 3; comparator, n = 2); and one suspected (daptomycin, n = 1)]. Test-of-cure clin. success rates were numerically higher for daptomycin than for the comparator [89.0 % (65/73) vs. 83.3 % (25/30); odds ratio 1.65 (95 % confidence interval 0.49-5.54)]. For patients with S. aureus infections, cure rates were 89.7 % (35/39) vs. 69.2 % (9/13), resp.; percentage points difference, 20.5 (95 % confidence interval -12.2 to 50.9). Rates of adverse events (AEs) and serious AEs were similar in both treatment arms; however, discontinuation rates for AEs/serious AEs were lower for daptomycin than for the comparator (3.8 % vs. 10.0 %). Three serious AEs were considered to be related to the study drug: one case each of pancytopenia (semi-synthetic penicillin), renal failure (vancomycin) and asymptomatic increase in creatine phosphokinase concns. (daptomycin). Conclusion: In elderly patients, for whom data were previously limited, the efficacy and safety of daptomycin have been confirmed, including for infections caused by S. aureus, regardless of methicillin susceptibility.
- 2Moran, G. J.; Fang, E.; Corey, G. R.; Das, A. F.; De Anda, C.; Prokocimer, P. Tedizolid for 6 days versus linezolid for 10 days for acute bacterial skin and skin-structure infections (ESTABLISH-2): a randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect Dis 2014, 14 (8), 696– 705, DOI: 10.1016/S1473-3099(14)70737-6Google Scholar2https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVWrs7bK&md5=70ead6ed25db678eeec65a6d7e00f151Tedizolid for 6 days versus linezolid for 10 days for acute bacterial skin and skin-structure infections (ESTABLISH-2): a randomised, double-blind, phase 3, non-inferiority trialMoran, Gregory J.; Fang, Edward; Corey, G. Ralph; Das, Anita F.; De Anda, Carisa; Prokocimer, PhilippeLancet Infectious Diseases (2014), 14 (8), 696-705CODEN: LIDABP; ISSN:1473-3099. (Elsevier Ltd.)New antibiotics are needed to treat infections caused by drug-resistant bacteria. Tedizolid is a novel oxazolidinone antibacterial drug designed to provide enhanced activity against Gram-pos. pathogens. We aimed to assess the efficacy and safety of i.v. to oral tedizolid for treatment of patients with acute bacterial skin and skin-structure infections. ESTABLISH-2 was a randomised, double-blind, phase 3, non-inferiority trial done between Sept 28, 2011, and Jan 10, 2013, at 58 centers in nine countries. Patients (aged ≥12 years) with acute bacterial skin and skin-structure infections (cellulitis or erysipelas, major cutaneous abscess, or wound infection) that had a min. lesion area of 75 cm2 and were suspected or documented to be assocd. with a Gram-pos. pathogen, were randomly assigned (1:1), via an interactive voice-response system with block randomisation, to receive i.v. once-daily tedizolid (200 mg for 6 days) or twice-daily linezolid (600 mg for 10 days), with optional oral step-down. Randomisation was stratified by geog. region and type of acute bacterial skin and skin-structure infection. The primary endpoint was early clin. response (≥20% redn. in lesion area at 48-72 h compared with baseline), with a non-inferiority margin of -10%. Anal. was by intention to treat. This study is registered with ClinicalTrials.gov, no. NCT01421511.666 patients were randomly assigned to receive tedizolid (n=332) or linezolid (n=334). 283 (85%) patients in the tedizolid group and 276 (83%) in the linezolid group achieved early clin. response (difference 2·6%, 95% CI -3·0 to 8·2), meeting the prespecified non-inferiority margin. Gastrointestinal adverse events were less frequent with tedizolid than linezolid, taking place in 52 (16%) of 331 patients and 67 (20%) of 327 patients in the safety population. Treatment-emergent adverse events leading to discontinuation of study drug were reported by one (<1%) patient in the tedizolid group and four (1%) patients in the linezolid group. I.v. to oral once-daily tedizolid 200 mg for 6 days was non-inferior to twice-daily linezolid 600 mg for 10 days for treatment of patients with acute bacterial skin and skin-structure infections. Tedizolid could become a useful option for the treatment of acute bacterial skin and skin-structure infections in the hospital and outpatient settings.
- 3File, T. M.; Goldberg, L.; Das, A.; Sweeney, C.; Saviski, J.; Gelone, S. P.; Seltzer, E.; Paukner, S.; Wicha, W. W.; Talbot, G. H.; Gasink, L. B. Efficacy and Safety of Intravenous-to-oral Lefamulin, a Pleuromutilin Antibiotic, for the Treatment of Community-acquired Bacterial Pneumonia: The Phase III Lefamulin Evaluation Against Pneumonia (LEAP 1) Trial. Clin Infect Dis 2019, 69 (11), 1856– 1867, DOI: 10.1093/cid/ciz090Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsVKlurY%253D&md5=8f51288366ed79a4a793c07e66076187Efficacy and safety of intravenous-to-oral lefamulin, a pleuromutilin antibiotic, for the treatment of community-acquired bacterial pneumonia: the phase III lefamulin evaluation against pneumonia (LEAP 1) trialFile, Thomas M., Jr.; Goldberg, Lisa; Das, Anita; Sweeney, Carolyn; Saviski, John; Gelone, Steven P.; Seltzer, Elyse; Paukner, Susanne; Wicha, Wolfgang W.; Talbot, George H.; Gasink, Leanne B.Clinical Infectious Diseases (2019), 69 (11), 1856-1867CODEN: CIDIEL; ISSN:1537-6591. (Oxford University Press)Lefamulin, a pleuromutilin antibiotic, is active against pathogens commonly causing community-acquired bacterial pneumonia (CABP). The Lefamulin Evaluation Against Pneumonia (LEAP 1) study was a global noninferiority trial to evaluate the efficacy and safety of lefamulin for the treatment of CABP. In this double-blind study, adults with CABP of Pneumonia Outcomes Research Team risk class ≥III were randomized 1:1 to receive lefamulin at 150 mg i.v. (IV) every 12 h or moxifloxacin at 400 mg IV every 24 h. After 6 doses, patients could be switched to an oral study drug if prespecified improvement criteria were met. If methicillin-resistant Staphylococcus aureus was suspected, either linezolid or placebo was added to moxifloxacin or lefamulin, resp. The US Food and Drug Administration primary endpoint was an early clin. response (ECR) 96 ± 24 h after the first dose of the study drug in the intent-to-treat (ITT) population (noninferiority margin, 12.5%). The European Medicines Agency co-primary endpoints were an investigator assessment of clin. response (IACR) 5-10 days after the last dose of the study drug in the modified ITT (mITT) and clin. evaluable (CE) populations (noninferiority margin, 10%). There were 551 patients randomized (n = 276 lefamulin; n = 275 moxifloxacin). Lefamulin was noninferior to moxifloxacin for ECR (87.3% vs 90.2%, resp.; difference -2.9%, 95% confidence interval [CI] g -8.5 to 2.8) and IACR (mITT, 81.7% vs 84.2%, resp.; difference -2.6%, 95% CI -8.9 to 3.9; CE, 86.9% vs 89.4%, resp.; difference -2.5%, 95% CI -8.4 to 3.4). Rates of study drug discontinuation due to treatment-emergent adverse events were 2.9% for lefamulin and 4.4% for moxifloxacin. Lefamulin was noninferior to moxifloxacin for the primary efficacy endpoints and was generally safe and well tolerated.
- 4WHO Antibacterial products in clinical development for priority pathogens. https://www.who.int/observatories/global-observatory-on-health-research-and-development/monitoring/antibacterial-products-in-clinical-development-for-priority-pathogens (accessed June 13, 2022).Google ScholarThere is no corresponding record for this reference.
- 5Rice, L. B. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J. Infect Dis 2008, 197 (8), 1079– 81, DOI: 10.1086/533452Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c3lvFChug%253D%253D&md5=c2c5ec8ad941f702f588f89741d9410eFederal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPERice Louis BThe Journal of infectious diseases (2008), 197 (8), 1079-81 ISSN:0022-1899.There is no expanded citation for this reference.
- 6Boucher, H. W.; Talbot, G. H.; Bradley, J. S.; Edwards, J. E.; Gilbert, D.; Rice, L. B.; Scheld, M.; Spellberg, B.; Bartlett, J. Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis 2009, 48 (1), 1– 12, DOI: 10.1086/595011Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M%252Fit1Krsg%253D%253D&md5=a3f71406b891f90f4f0f77b5f1ebc4f8Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of AmericaBoucher Helen W; Talbot George H; Bradley John S; Edwards John E; Gilbert David; Rice Louis B; Scheld Michael; Spellberg Brad; Bartlett JohnClinical infectious diseases : an official publication of the Infectious Diseases Society of America (2009), 48 (1), 1-12 ISSN:.The Infectious Diseases Society of America (IDSA) continues to view with concern the lean pipeline for novel therapeutics to treat drug-resistant infections, especially those caused by gram-negative pathogens. Infections now occur that are resistant to all current antibacterial options. Although the IDSA is encouraged by the prospect of success for some agents currently in preclinical development, there is an urgent, immediate need for new agents with activity against these panresistant organisms. There is no evidence that this need will be met in the foreseeable future. Furthermore, we remain concerned that the infrastructure for discovering and developing new antibacterials continues to stagnate, thereby risking the future pipeline of antibacterial drugs. The IDSA proposed solutions in its 2004 policy report, "Bad Bugs, No Drugs: As Antibiotic R&D Stagnates, a Public Health Crisis Brews," and recently issued a "Call to Action" to provide an update on the scope of the problem and the proposed solutions. A primary objective of these periodic reports is to encourage a community and legislative response to establish greater financial parity between the antimicrobial development and the development of other drugs. Although recent actions of the Food and Drug Administration and the 110th US Congress present a glimmer of hope, significant uncertainly remains. Now, more than ever, it is essential to create a robust and sustainable antibacterial research and development infrastructure--one that can respond to current antibacterial resistance now and anticipate evolving resistance. This challenge requires that industry, academia, the National Institutes of Health, the Food and Drug Administration, the Centers for Disease Control and Prevention, the US Department of Defense, and the new Biomedical Advanced Research and Development Authority at the Department of Health and Human Services work productively together. This report provides an update on potentially effective antibacterial drugs in the late-stage development pipeline, in the hope of encouraging such collaborative action.
- 7Cassini, A.; Hogberg, L. D.; Plachouras, D.; Quattrocchi, A.; Hoxha, A.; Simonsen, G. S.; Colomb-Cotinat, M.; Kretzschmar, M. E.; Devleesschauwer, B.; Cecchini, M.; Ouakrim, D. A.; Oliveira, T. C.; Struelens, M. J.; Suetens, C.; Monnet, D. L.; the Burden of AMR Collective Group Attributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Economic Area in 2015: a population-level modelling analysis. Lancet Infect. Dis. 2019, 19 (1), 56– 66, DOI: 10.1016/S1473-3099(18)30605-4Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cvotFGluw%253D%253D&md5=151619810730b07e3d84692f1d966b9aAttributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Economic Area in 2015: a population-level modelling analysisCassini Alessandro; Hogberg Liselotte Diaz; Plachouras Diamantis; Quattrocchi Annalisa; Hoxha Ana; Struelens Marc J; Suetens Carl; Monnet Dominique L; Simonsen Gunnar Skov; Colomb-Cotinat Melanie; Kretzschmar Mirjam E; Devleesschauwer Brecht; Cecchini Michele; Ouakrim Driss Ait; Oliveira Tiago CravoThe Lancet. Infectious diseases (2019), 19 (1), 56-66 ISSN:.BACKGROUND: Infections due to antibiotic-resistant bacteria are threatening modern health care. However, estimating their incidence, complications, and attributable mortality is challenging. We aimed to estimate the burden of infections caused by antibiotic-resistant bacteria of public health concern in countries of the EU and European Economic Area (EEA) in 2015, measured in number of cases, attributable deaths, and disability-adjusted life-years (DALYs). METHODS: We estimated the incidence of infections with 16 antibiotic resistance-bacterium combinations from European Antimicrobial Resistance Surveillance Network (EARS-Net) 2015 data that was country-corrected for population coverage. We multiplied the number of bloodstream infections (BSIs) by a conversion factor derived from the European Centre for Disease Prevention and Control point prevalence survey of health-care-associated infections in European acute care hospitals in 2011-12 to estimate the number of non-BSIs. We developed disease outcome models for five types of infection on the basis of systematic reviews of the literature. FINDINGS: From EARS-Net data collected between Jan 1, 2015, and Dec 31, 2015, we estimated 671 689 (95% uncertainty interval [UI] 583 148-763 966) infections with antibiotic-resistant bacteria, of which 63·5% (426 277 of 671 689) were associated with health care. These infections accounted for an estimated 33 110 (28 480-38 430) attributable deaths and 874 541 (768 837-989 068) DALYs. The burden for the EU and EEA was highest in infants (aged <1 year) and people aged 65 years or older, had increased since 2007, and was highest in Italy and Greece. INTERPRETATION: Our results present the health burden of five types of infection with antibiotic-resistant bacteria expressed, for the first time, in DALYs. The estimated burden of infections with antibiotic-resistant bacteria in the EU and EEA is substantial compared with that of other infectious diseases, and has increased since 2007. Our burden estimates provide useful information for public health decision-makers prioritising interventions for infectious diseases. FUNDING: European Centre for Disease Prevention and Control.
- 8Antimicrobial Resistance, C. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 2022, 399 (10325), 629– 655, DOI: 10.1016/S0140-6736(21)02724-0Google ScholarThere is no corresponding record for this reference.
- 9Richter, M. F.; Drown, B. S.; Riley, A. P.; Garcia, A.; Shirai, T.; Svec, R. L.; Hergenrother, P. J. Predictive compound accumulation rules yield a broad-spectrum antibiotic. Nature 2017, 545 (7654), 299– 304, DOI: 10.1038/nature22308Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXnsVeis7Y%253D&md5=ae869cc99ef0e6805b96b6595a222cdcPredictive compound accumulation rules yield a broad-spectrum antibioticRichter, Michelle F.; Drown, Bryon S.; Riley, Andrew P.; Garcia, Alfredo; Shirai, Tomohiro; Svec, Riley L.; Hergenrother, Paul J.Nature (London, United Kingdom) (2017), 545 (7654), 299-304CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Most small mols. are unable to rapidly traverse the outer membrane of Gram-neg. bacteria and accumulate inside these cells, making the discovery of much-needed drugs against these pathogens challenging. Current understanding of the physicochem. properties that dictate small-mol. accumulation in Gram-neg. bacteria is largely based on retrospective analyses of antibacterial agents, which suggest that polarity and mol. wt. are key factors. Here the authors assess the ability of over 180 diverse compds. to accumulate in Escherichia coli. Computational anal. of the results reveals major differences from the retrospective studies, namely that the small mols. that are most likely to accumulate contain an amine, are amphiphilic and rigid, and have low globularity. These guidelines were then applied to convert deoxynybomycin, a natural product that is active only against Gram-pos. organisms, into an antibiotic with activity against a diverse panel of multi-drug-resistant Gram-neg. pathogens. The authors anticipate that these findings will aid in the discovery and development of antibiotics against Gram-neg. bacteria.
- 10Zhao, S.; Adamiak, J. W.; Bonifay, V.; Mehla, J.; Zgurskaya, H. I.; Tan, D. S. Defining new chemical space for drug penetration into Gram-negative bacteria. Nat. Chem. Biol. 2020, 16 (12), 1293– 1302, DOI: 10.1038/s41589-020-00674-6Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlGjsLfM&md5=43f7ee77d0e360e4666c569eb5ef728fDefining new chemical space for drug penetration into Gram-negative bacteriaZhao, Shibin; Adamiak, Justyna W.; Bonifay, Vincent; Mehla, Jitender; Zgurskaya, Helen I.; Tan, Derek S.Nature Chemical Biology (2020), 16 (12), 1293-1302CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Abstr.: We live in the era of antibiotic resistance, and this problem will progressively worsen if no new solns. emerge. In particular, Gram-neg. pathogens present both biol. and chem. challenges that hinder the discovery of new antibacterial drugs. First, these bacteria are protected from a variety of structurally diverse drugs by a low-permeability barrier composed of two membranes with distinct permeability properties, in addn. to active drug efflux, making this cell envelope impermeable to most compds. Second, chem. libraries currently used in drug discovery contain few compds. that can penetrate Gram-neg. bacteria. As a result of these challenges, intensive screening campaigns have led to few successes, highlighting the need for new approaches to identify regions of chem. space that are specifically relevant to antibacterial drug discovery. Herein we provide an overview of emerging insights into this problem and outline a general approach to addressing it using prospective anal. of chem. libraries for the ability of compds. to accumulate in Gram-neg. bacteria. The overall goal is to develop robust cheminformatic tools to predict Gram-neg. permeation and efflux, which can then be used to guide medicinal chem. campaigns and the design of antibacterial discovery libraries.
- 11Munoz, K. A.; Hergenrother, P. J. Facilitating Compound Entry as a Means to Discover Antibiotics for Gram-Negative Bacteria. Acc. Chem. Res. 2021, 54 (6), 1322– 1333, DOI: 10.1021/acs.accounts.0c00895Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkvFyisb8%253D&md5=bd3fe94a1db2612554c9d3557ee7205eFacilitating Compound Entry as a Means to Discover Antibiotics for Gram-Negative BacteriaMunoz, Kristen A.; Hergenrother, Paul J.Accounts of Chemical Research (2021), 54 (6), 1322-1333CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. It has been over half a century since the last class of antibiotics active against the most problematic Gram-neg. bacteria was approved by the Food and Drug Administration (FDA). The major challenge with developing antibiotics to treat these infections is not drug-target engagement but rather the inability of most small mols. to traverse the Gram-neg. membranes, be retained, and accumulate within the cell. Despite an abundance of lead compds., limited understanding of the physicochem. properties needed for compd. accumulation (or avoidance of efflux) in Gram-neg. bacteria has precluded a generalizable approach for developing Gram-neg. antibiotics. Indeed, in many instances, despite years of intensive derivatization efforts and the synthesis of hundreds of compds. aimed at building in Gram-neg. activity, little or no progress has been made in expanding the spectrum of activity for many Gram-pos.-only antibiotics. We describe the discovery and successful applications of a promising strategy for enhancing the accumulation of Gram-pos.-only antibiotics as a means of imbuing compds. with broad-spectrum activity. Utilizing a prospective approach examg. the accumulation in Escherichia coli for >180 diverse compds., we found that small mols. have an increased likelihood to accumulate in E. coli when they contain an ionizable nitrogen, have low 3-dimensionality, and are rigid. Implementing these guidelines, codified as the eNTRy rules and assisted by web application www.entry-way.org, we have facilitated compd. entry and systematically built Gram-neg. activity into Gram-pos.-only antibiotics. Though each antibiotic will have case-specific considerations, we describe a set of important criteria to consider when selecting candidate Gram-pos.-only antibiotics for conversion to Gram-neg.-active versions via the eNTRy rules. As detailed herein, using this blueprint the spectrum of activity was expanded for 3 antibiotic classes that engage 3 different biol. targets: DNA gyrase inhibitor 6DNM, FabI inhibitor Debio-1452, and FMN riboswitch inhibitor Ribocil C. In each scenario, the eNTRy rules guided the synthesis of key analogs predisposed to accumulate in Gram-neg. bacteria leading to compds. that display antibiotic activity (min. inhibitory concns. (MIC) ≤8μg/mL) against E. coli and other Gram-neg. ESKAPE pathogens. While the eNTRy rules will continue to be refined and enhanced as more accumulation data is gathered, on the basis of these collective results and on other examples not covered herein it is clear that the eNTRy rules are actionable for the development of novel broad-spectrum antibiotics from Gram-pos.-only compds. By enabling the prediction of compd. accumulation, the eNTRy rules should facilitate the process of discovering and developing novel antibiotics active against Gram-neg. bacteria.
- 12Payne, D. J.; Gwynn, M. N.; Holmes, D. J.; Pompliano, D. L. Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat. Rev. Drug Discov 2007, 6 (1), 29– 40, DOI: 10.1038/nrd2201Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtlGktbfM&md5=18100253f9868af578ea621ab98a5c86Drugs for bad bugs: confronting the challenges of antibacterial discoveryPayne, David J.; Gwynn, Michael N.; Holmes, David J.; Pompliano, David L.Nature Reviews Drug Discovery (2007), 6 (1), 29-40CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The sequencing of the first complete bacterial genome in 1995 heralded a new era of hope for antibacterial drug discoverers, who now had the tools to search entire genomes for new antibacterial targets. Several companies, including GlaxoSmithKline, moved back into the antibacterials area and embraced a genomics-derived, target-based approach to screen for new classes of drugs with novel modes of action. Here, we share our experience of evaluating more than 300 genes and 70 high-throughput screening campaigns over a period of 7 years, and look at what we learned and how that has influenced GlaxoSmithKline's antibacterials strategy going forward.
- 13Tommasi, R.; Brown, D. G.; Walkup, G. K.; Manchester, J. I.; Miller, A. A. ESKAPEing the labyrinth of antibacterial discovery. Nat. Rev. Drug Discov 2015, 14 (8), 529– 42, DOI: 10.1038/nrd4572Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFyks77M&md5=0d3159250e27926ab280c305ea5fb9efESKAPEing the labyrinth of antibacterial discoveryTommasi, Ruben; Brown, Dean G.; Walkup, Grant K.; Manchester, John I.; Miller, Alita A.Nature Reviews Drug Discovery (2015), 14 (8), 529-542CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Antimicrobial drug resistance is a growing threat to global public health. Multidrug resistance among the 'ESKAPE' organisms - encompassing Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. - is of particular concern because they are responsible for many serious infections in hospitals. Although some promising agents are in the pipeline, there is an urgent need for new antibiotic scaffolds. However, antibacterial researchers have struggled to identify new small mols. with meaningful cellular activity, esp. those effective against multidrug-resistant Gram-neg. pathogens. This difficulty ultimately stems from an incomplete understanding of efflux systems and compd. permeation through bacterial membranes. This Opinion article describes findings from target-based and phenotypic screening efforts carried out at AstraZeneca over the past decade, discusses some of the subsequent chem. challenges and concludes with a description of new approaches comprising a combination of computational modeling and advanced biol. tools which may pave the way towards the discovery of new antibacterial agents.
- 14Yao, J.; Rock, C. O. Resistance Mechanisms and the Future of Bacterial Enoyl-Acyl Carrier Protein Reductase (FabI) Antibiotics. Cold Spring Harb Perspect Med. 2016, 6 (3), a027045, DOI: 10.1101/cshperspect.a027045Google ScholarThere is no corresponding record for this reference.
- 15Seefeld, M. A.; Miller, W. H.; Newlander, K. A.; Burgess, W. J.; Payne, D. J.; Rittenhouse, S. F.; Moore, T. D.; DeWolf, W. E., Jr.; Keller, P. M.; Qiu, X.; Janson, C. A.; Vaidya, K.; Fosberry, A. P.; Smyth, M. G.; Jaworski, D. D.; Slater-Radosti, C.; Huffman, W. F. Inhibitors of bacterial enoyl acyl carrier protein reductase (FabI): 2,9-disubstituted 1,2,3,4-tetrahydropyrido[3,4-b]indoles as potential antibacterial agents. Bioorg. Med. Chem. Lett. 2001, 11 (17), 2241– 4, DOI: 10.1016/S0960-894X(01)00405-XGoogle Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXmt1yqtbY%253D&md5=6904b75740db275d6defbbc08009a6ebInhibitors of bacterial enoyl acyl carrier protein reductase (FabI): 2,9-disubstituted 1,2,3,4-tetrahydropyrido[3,4-b]indoles as potential antibacterial agentsSeefeld, M. A.; Miller, W. H.; Newlander, K. A.; Burgess, W. J.; Payne, D. J.; Rittenhouse, S. F.; Moore, T. D.; DeWolf, W. E.; Keller, P. M.; Qiu, X.; Janson, C. A.; Vaidya, K.; Fosberry, A. P.; Smyth, M. G.; Jaworski, D. D.; Slater-Radosti, C.; Huffman, W. F.Bioorganic & Medicinal Chemistry Letters (2001), 11 (17), 2241-2244CODEN: BMCLE8; ISSN:0960-894X. (Elsevier Science Ltd.)An SAR study of a screening lead has led to the identification of 2,9-disubstituted 1,2,3,4-tetrahydropyrido[3,4-b]indoles as inhibitors of Staphylococcus aureus enoyl acyl carrier protein reductase (FabI).
- 16Payne, D. J.; Miller, W. H.; Berry, V.; Brosky, J.; Burgess, W. J.; Chen, E.; DeWolf Jr, W. E., Jr.; Fosberry, A. P.; Greenwood, R.; Head, M. S.; Heerding, D. A.; Janson, C. A.; Jaworski, D. D.; Keller, P. M.; Manley, P. J.; Moore, T. D.; Newlander, K. A.; Pearson, S.; Polizzi, B. J.; Qiu, X.; Rittenhouse, S. F.; Slater-Radosti, C.; Salyers, K. L.; Seefeld, M. A.; Smyth, M. G.; Takata, D. T.; Uzinskas, I. N.; Vaidya, K.; Wallis, N. G.; Winram, S. B.; Yuan, C. C.; Huffman, W. F. Discovery of a novel and potent class of FabI-directed antibacterial agents. Antimicrob. Agents Chemother. 2002, 46 (10), 3118– 24, DOI: 10.1128/AAC.46.10.3118-3124.2002Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XntlOgtbk%253D&md5=b56fe92bf2328df5aacd97fe89f44226Discovery of a novel and potent class of FabI-directed antibacterial agentsPayne, David J.; Miller, William H.; Berry, Valerie; Brosky, John; Burgess, Walter J.; Chen, Emile; DeWolf, Walter E., Jr.; Fosberry, Andrew P.; Greenwood, Rebecca; Head, Martha S.; Heerding, Dirk A.; Janson, Cheryl A.; Jaworski, Deborah D.; Keller, Paul M.; Manley, Peter J.; Moore, Terrance D.; Newlander, Kenneth A.; Pearson, Stewart; Polizzi, Brian J.; Qiu, Xiayang; Rittenhouse, Stephen F.; Slater-Radosti, Courtney; Salyers, Kevin L.; Seefeld, Mark A.; Smyth, Martin G.; Takata, Dennis T.; Uzinskas, Irene N.; Vaidya, Kalindi; Wallis, Nicola G.; Winram, Scott B.; Yuan, Catherine C. K.; Huffman, William F.Antimicrobial Agents and Chemotherapy (2002), 46 (10), 3118-3124CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Bacterial enoyl-acyl carrier protein (ACP) reductase (FabI) catalyzes the final step in each elongation cycle of bacterial fatty acid biosynthesis and is an attractive target for the development of new antibacterial agents. High-throughput screening of the Staphylococcus aureus FabI enzyme identified a novel, weak inhibitor with no detectable antibacterial activity against S. aureus. Iterative medicinal chem. and x-ray crystal structure-based design led to the identification of compd. 4 [(E)-N-methyl-N-(2-methyl-1H-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide], which is 350-fold more potent than the original lead compd. obtained by high-throughput screening in the FabI inhibition assay. Compd. 4 has exquisite antistaphylococci activity, achieving MICs at which 90% of isolates are inhibited more than 500 times lower than those of nine currently available antibiotics against a panel of multidrug-resistant strains of S. aureus and Staphylococcus epidermidis. Furthermore, compd. 4 exhibits excellent in vivo efficacy in an S. aureus infection model in rats. Biochem. and genetic approaches have confirmed that the mode of antibacterial action of compd. 4 and related compd. is via inhibition of FabI. Compd. 4 also exhibits weak FabK inhibitory activity, which may explain its antibacterial activity against Streptococcus pneumoniae and Enterococcus faecalis, which depend on FabK and both FabK and FabI, resp., for their enoyl-ACP reductase function. These results show that compd. 4 is representative of a new, totally synthetic series of antibacterial agents that has the potential to provide novel alternatives for the treatment of S. aureus infections that are resistant to our present armory of antibiotics.
- 17Wittke, F.; Vincent, C.; Chen, J.; Heller, B.; Kabler, H.; Overcash, J. S.; Leylavergne, F.; Dieppois, G. Afabicin, a First-in-Class Antistaphylococcal Antibiotic, in the Treatment of Acute Bacterial Skin and Skin Structure Infections: Clinical Noninferiority to Vancomycin/Linezolid. Antimicrob. Agents Chemother. 2020, 64 (10), e00250-20, DOI: 10.1128/AAC.00250-20Google ScholarThere is no corresponding record for this reference.
- 18Karlowsky, J. A.; Kaplan, N.; Hafkin, B.; Hoban, D. J.; Zhanel, G. G. AFN-1252, a FabI inhibitor, demonstrates a Staphylococcus-specific spectrum of activity. Antimicrob. Agents Chemother. 2009, 53 (8), 3544– 8, DOI: 10.1128/AAC.00400-09Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXps1ygsL0%253D&md5=48086b24a4c38d14373320c89c91f1bfAFN-1252, a FabI inhibitor, demonstrates a staphylococcus-specific spectrum of activityKarlowsky, James A.; Kaplan, Nachum; Hafkin, Barry; Hoban, Daryl J.; Zhanel, George G.Antimicrobial Agents and Chemotherapy (2009), 53 (8), 3544-3548CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)AFN-1252, a potent inhibitor of enoyl-acyl carrier protein reductase (FabI), inhibited all clin. isolates of Staphylococcus aureus (n = 502) and Staphylococcus epidermidis (n = 51) tested, including methicillin (methicillin)-resistant isolates, at concns. of ≤0.12 μg/mL. In contrast, AFN-1252 was inactive (MIC90, >4 μg/mL) against clin. isolates of Streptococcus pneumoniae, beta-hemolytic streptococci, Enterococcus spp., Enterobacteriaceae, nonfermentative gram-neg. bacilli, and Moraxella catarrhalis. These data support the continued development of AFN-1252 for the treatment of patients with resistant staphylococcal infections.
- 19Parker, E. N.; Drown, B. S.; Geddes, E. J.; Lee, H. Y.; Ismail, N.; Lau, G. W.; Hergenrother, P. J. Implementation of permeation rules leads to a FabI inhibitor with activity against Gram-negative pathogens. Nat. Microbiol 2020, 5 (1), 67– 75, DOI: 10.1038/s41564-019-0604-5Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFOru7jM&md5=39c84cdc028ef0c5568f2cecd60cd209Implementation of permeation rules leads to a FabI inhibitor with activity against Gram-negative pathogensParker, Erica N.; Drown, Bryon S.; Geddes, Emily J.; Lee, Hyang Yeon; Ismail, Nahed; Lau, Gee W.; Hergenrother, Paul J.Nature Microbiology (2020), 5 (1), 67-75CODEN: NMAICH; ISSN:2058-5276. (Nature Research)Gram-neg. bacterial infections are a significant public health concern, and the lack of new drug classes for these pathogens is linked to the inability of most drug leads to accumulate inside Gram-neg. bacteria1-7. Here, we report the development of a web application-eNTRyway-that predicts compd. accumulation (in Escherichia coli) from its structure. In conjunction with structure-activity relationships and X-ray data, eNTRyway was utilized to re-design Debio-1452-a Gram-pos.-only antibiotic8-into versions that accumulate in E. coli and possess antibacterial activity against high-priority Gram-neg. pathogens. The lead compd. Debio-1452-NH3 operates as an antibiotic via the same mechanism as Debio-1452, namely potent inhibition of the enoyl-acyl carrier protein reductase FabI, as validated by in vitro enzyme assays and the generation of bacterial isolates with spontaneous target mutations. Debio-1452-NH3 is well tolerated in vivo, reduces bacterial burden in mice and rescues mice from lethal infections with clin. isolates of Acinetobacter baumannii, Klebsiella pneumoniae and E. coli. This work provides tools for the facile discovery and development of high-accumulating compds. in E. coli, and a general blueprint for the conversion of Gram-pos.-only compds. into broad-spectrum antibiotics.
- 20Richter, M. F.; Hergenrother, P. J. The challenge of converting Gram-positive-only compounds into broad-spectrum antibiotics. Ann. N.Y. Acad. Sci. 2019, 1435 (1), 18– 38, DOI: 10.1111/nyas.13598Google Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlyktw%253D%253D&md5=12fffe0d10e7d3e5c134ac28ce929ab3The challenge of converting Gram-positive-only compounds into broad-spectrum antibioticsRichter, Michelle F.; Hergenrother, Paul J.Annals of the New York Academy of Sciences (2019), 1435 (1, Antimicrobial Therapeutics Reviews), 18-38CODEN: ANYAA9; ISSN:0077-8923. (John Wiley & Sons, Inc.)A review. Multidrug resistant Gram-neg. bacterial infections are on the rise, and there is a lack of new classes of drugs to treat these pathogens. This drug shortage is largely due to the challenge of finding antibiotics that can permeate and persist inside Gram-neg. species. Efforts to understand the mol. properties that enable certain compds. to accumulate in Gram-neg. bacteria based on retrospective studies of known antibiotics have not been generally actionable in the development of new antibiotics. A recent assessment of the ability of >180 diverse small mols. to accumulate in Escherichia coli led to predictive guidelines for compd. accumulation in E. coli. These "eNTRy rules" state that compds. are most likely to accumulate if they contain a nonsterically encumbered ionizable Nitrogen (primary amines are the best), have low Three-dimensionality (globularity ≤ 0.25), and are relatively Rigid (rotatable bonds ≤ 5). In this review, we look back through 50+ years of antibacterial research and 1000s of derivs. and assess this historical data set through the lens of these predictive guidelines. The results are consistent with the eNTRy rules, suggesting that the eNTRy rules may provide an actionable and general roadmap for the conversion of Gram-pos.-only compds. into broad-spectrum antibiotics.
- 21Kaplan, N.; Albert, M.; Awrey, D.; Bardouniotis, E.; Berman, J.; Clarke, T.; Dorsey, M.; Hafkin, B.; Ramnauth, J.; Romanov, V.; Schmid, M. B.; Thalakada, R.; Yethon, J.; Pauls, H. W. Mode of action, in vitro activity, and in vivo efficacy of AFN-1252, a selective antistaphylococcal FabI inhibitor. Antimicrob. Agents Chemother. 2012, 56 (11), 5865– 5874, DOI: 10.1128/AAC.01411-12Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsF2iurfK&md5=67bc12e665ce45b808d799db4fb09c92Mode of action, in vitro activity, and in vivo efficacy of AFN-1252, a selective antistaphylococcal FabI inhibitorKaplan, Nachum; Albert, Monique; Awrey, Donald; Bardouniotis, Elias; Berman, Judd; Clarke, Teresa; Dorsey, Mandy; Hafkin, Barry; Ramnauth, Jaillal; Romanov, Vladimir; Schmid, Molly B.; Thalakada, Rosanne; Yethon, Jeremy; Pauls, Henry W.Antimicrobial Agents and Chemotherapy (2012), 56 (11), 5865-5874CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)The mechanism of action of AFN-1252, a selective inhibitor of Staphylococcus aureus enoyl-acyl carrier protein reductase (FabI), which is involved in fatty acid biosynthesis, was confirmed by using biochem., macromol. synthesis, genetics, and co-crystn. of an AFN-1252-FabI complex. AFN-1252 demonstrated a low propensity for spontaneous resistance development and a time-dependent redn. of the viability of both methicillin-susceptible and methicillin-resistant S. aureus, achieving a ≥2-log10 redn. in S. aureus counts over 24 h, and was extremely potent against clin. isolates of S. aureus (MIC90, 0.015 μg/mL) and coagulase-neg. staphylococci (MIC90, 0.12 μg/mL), regardless of their drug resistance, hospital- or community-assocd. origin, or other clin. subgroup. AFN-1252 was orally available in mouse pharmacokinetic studies, and a single oral dose of 1 mg/kg AFN-1252 was efficacious in a mouse model of septicemia, providing 100% protection from an otherwise lethal peritoneal infection of S. aureus Smith. A median ED of 0.15 mg/kg indicated that AFN-1252 was 12 to 24 times more potent than linezolid in the model. These studies, demonstrating a selective mode of action, potent in vitro activity, and in vivo efficacy, support the continued investigation of AFN-1252 as a targeted therapeutic for staphylococcal infections.
- 22Seefeld, M. A.; Miller, W. H.; Newlander, K. A.; Burgess, W. J.; DeWolf, W. E., Jr.; Elkins, P. A.; Head, M. S.; Jakas, D. R.; Janson, C. A.; Keller, P. M.; Manley, P. J.; Moore, T. D.; Payne, D. J.; Pearson, S.; Polizzi, B. J.; Qiu, X.; Rittenhouse, S. F.; Uzinskas, I. N.; Wallis, N. G.; Huffman, W. F. Indole naphthyridinones as inhibitors of bacterial enoyl-ACP reductases FabI and FabK. J. Med. Chem. 2003, 46 (9), 1627– 35, DOI: 10.1021/jm0204035Google Scholar22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXitlGlur8%253D&md5=0626ec2f4185fa86fadbba7867dc1dd8Indole Naphthyridinones as Inhibitors of Bacterial Enoyl-ACP Reductases FabI and FabKSeefeld, Mark A.; Miller, William H.; Newlander, Kenneth A.; Burgess, Walter J.; DeWolf, Walter E., Jr.; Elkins, Patricia A.; Head, Martha S.; Jakas, Dalia R.; Janson, Cheryl A.; Keller, Paul M.; Manley, Peter J.; Moore, Terrance D.; Payne, David J.; Pearson, Stewart; Polizzi, Brian J.; Qiu, Xiayang; Rittenhouse, Stephen F.; Uzinskas, Irene N.; Wallis, Nicola G.; Huffman, William F.Journal of Medicinal Chemistry (2003), 46 (9), 1627-1635CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bacterial enoyl-ACP reductase (FabI) is responsible for catalyzing the final step of bacterial fatty acid biosynthesis and is an attractive target for the development of novel antibacterial agents. Previously we reported the development of FabI inhibitor 4 with narrow spectrum antimicrobial activity and in vivo efficacy against Staphylococcus aureus via i.p. administration. Through iterative medicinal chem. aided by x-ray crystal structure anal., a new series of inhibitors has been developed with greatly increased potency against FabI-contg. organisms. Several of these new inhibitors have potent antibacterial activity against multidrug resistant strains of S. aureus, and compd. 30 demonstrates exceptional oral (po) in vivo efficacy in a S. aureus infection model in rats. While optimizing FabI inhibitory activity, compds. 29 and 30 were identified as having low micromolar FabK inhibitory activity, thereby increasing the antimicrobial spectrum of these compds. to include the FabK-contg. pathogens Streptococcus pneumoniae and Enterococcus faecalis. The results described herein support the hypothesis that bacterial enoyl-ACP reductases are valid targets for antibacterial agents.
- 23Ramnauth, J.; Surman, M. D.; Sampson, P. B.; Forrest, B.; Wilson, J.; Freeman, E.; Manning, D. D.; Martin, F.; Toro, A.; Domagala, M.; Awrey, D. E.; Bardouniotis, E.; Kaplan, N.; Berman, J.; Pauls, H. W. 2,3,4,5-Tetrahydro-1H-pyrido[2,3-b and e][1,4]diazepines as inhibitors of the bacterial enoyl ACP reductase, FabI. Bioorg. Med. Chem. Lett. 2009, 19 (18), 5359– 5362, DOI: 10.1016/j.bmcl.2009.07.094Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVeqsL3K&md5=b9194f82fe56e322474adcc88e8c0f5e2,3,4,5-Tetrahydro-1H-pyrido[2,3-b and e][1,4]diazepines as inhibitors of the bacterial enoyl ACP reductase, FabIRamnauth, Jailall; Surman, Mathew D.; Sampson, Peter B.; Forrest, Bryan; Wilson, Jeff; Freeman, Emily; Manning, David D.; Martin, Fernando; Toro, Andras; Domagala, Megan; Awrey, Donald E.; Bardouniotis, Elias; Kaplan, Nachum; Berman, Judd; Pauls, Henry W.Bioorganic & Medicinal Chemistry Letters (2009), 19 (18), 5359-5362CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)In the search for new antibacterial agents, the enzyme FabI has been identified as an attractive target. Employing a structure guided approach, the previously reported ene-amide series of FabI inhibitors were expanded to include 2,3,4,5-tetrahydro-1H-pyrido[2,3-b and e][1,4]diazepines, e.g. I (R = 1-methyl-2-indolyl, 3-methyl-2-benzofuranyl, etc.; X = H2, O) and II. These novel series incorporate addnl. H-bonding functions and can be more water sol. than their naphthyridinone progenitors; diazepine II (R = 1-methyl-2-indolyl; X = O) is shown to be efficacious in a mouse infection model.
- 24Takhi, M.; Sreenivas, K.; Reddy, C. K.; Munikumar, M.; Praveena, K.; Sudheer, P.; Rao, B. N.; Ramakanth, G.; Sivaranjani, J.; Mulik, S.; Reddy, Y. R.; Narasimha Rao, K.; Pallavi, R.; Lakshminarasimhan, A.; Panigrahi, S. K.; Antony, T.; Abdullah, I.; Lee, Y. K.; Ramachandra, M.; Yusof, R.; Rahman, N. A.; Subramanya, H. Discovery of azetidine based ene-amides as potent bacterial enoyl ACP reductase (FabI) inhibitors. Eur. J. Med. Chem. 2014, 84, 382– 394, DOI: 10.1016/j.ejmech.2014.07.036Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1Grs7bE&md5=836c98d7e9a806615200c312836adae7Discovery of azetidine based ene-amides as potent bacterial enoyl ACP reductase (FabI) inhibitorsTakhi, Mohamed; Sreenivas, Kandepu; Reddy, Chandrashekar K.; Munikumar, Mahadari; Praveena, Kolakota; Sudheer, Pabolu; Rao, Bandaru N. V. M.; Ramakanth, Gollamudi; Sivaranjani, Jampala; Mulik, Shardaprasad; Reddy, Yeruva R.; Narasimha Rao, Krishnamurthy; Pallavi, Rentala; Lakshminarasimhan, Anirudha; Panigrahi, Sunil K.; Antony, Thomas; Abdullah, Iskandar; Lee, Yean K.; Ramachandra, Murali; Yusof, Rohana; Rahman, Noorsaadah A.; Subramanya, HosahalliEuropean Journal of Medicinal Chemistry (2014), 84 (), 382-394CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A novel and potent series of ene-amides featuring azetidines has been developed as FabI inhibitors active against drug resistant Gram-pos. pathogens particularly staphylococcal organisms. Most of the compds. from the series possessed excellent biochem. inhibition of Staphylococcus aureus FabI enzyme and whole cell activity against clin. relevant MRSA, MSSA and MRSE organisms which are responsible for significant morbidity and mortality in community as well as hospital settings. The binding mode of one of the leads, I, in Escherichia coli FabI enzyme was detd. unambiguously using x-ray crystallog. The lead compds. displayed good metabolic stability in mice liver microsomes and pharmacokinetic profile in mice. The in vivo efficacy of lead AEA16 has been demonstrated in a lethal murine systemic infection model.
- 25Hergenrother, P. J.; Geddes, E. J.; Drown, B. S.; Motika, S. E.; Parker, S. E. Antibiotics Effective for Gram-Negative Pathogens. WO2019/177975A1, 2019.Google ScholarThere is no corresponding record for this reference.
- 26Gerusz, V. B.; Bravo, J.; Pauls, H.; Berman, J.; Finn, T. Novel Compounds and their Use. WO2021/123372A1, 2021.Google ScholarThere is no corresponding record for this reference.
- 27Hergenrother, P. J.; Parker, E. N.; Hung, D.; Serrano-Wu, M.; Lee, K. K. FabI inhibitors for Gram-negative pathogens. US Provisional Patent Application No. 63/156,145. 2021.Google ScholarThere is no corresponding record for this reference.
- 28Geddes, E. J.; Li, Z.; Hergenrother, P. J. An LC-MS/MS assay and complementary web-based tool to quantify and predict compound accumulation in E. coli. Nat. Protoc 2021, 16 (10), 4833– 4854, DOI: 10.1038/s41596-021-00598-yGoogle Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFGltrvP&md5=7a4d4b07b133a2e525f8f88c1db3ba11An LC-MS/MS assay and complementary web-based tool to quantify and predict compound accumulation in E. coliGeddes, Emily J.; Li, Zhong; Hergenrother, Paul J.Nature Protocols (2021), 16 (10), 4833-4854CODEN: NPARDW; ISSN:1750-2799. (Nature Portfolio)A review. Novel classes of broad-spectrum antibiotics have been extremely difficult to discover, largely due to the impermeability of the Gram-neg. membranes coupled with a poor understanding of the physicochem. properties a compd. should possess to promote its accumulation inside the cell. To address this challenge, numerous methodologies for assessing intracellular compd. accumulation in Gram-neg. bacteria have been established, including classic radiometric and fluorescence-based methods. The recent development of accumulation assays that utilize liq. chromatog.-tandem mass spectrometry (LC-MS/MS) have circumvented the requirement for labeled compds., enabling assessment of a substantially broader range of small mols. Our unbiased study of accumulation trends in Escherichia coli using an LC-MS/MS-based assay led to the development of the eNTRy rules, which stipulate that a compd. is most likely to accumulate in E. coli if it has an ionizable Nitrogen, has low Three-dimensionality and is relatively Rigid. To aid in the implementation of the eNTRy rules, we developed a complementary web tool, eNTRyway, which calcs. relevant properties and predicts compd. accumulation. Here we provide a comprehensive protocol for anal. and prediction of intracellular accumulation of small mols. in E. coli using an LC-MS/MS-based assay (which takes ∼2 d) and eNTRyway, a workflow that is readily adoptable by any microbiol., biochem. or chem. biol. lab.
- 29Zhu, L.; Lin, J.; Ma, J.; Cronan, J. E.; Wang, H. Triclosan resistance of Pseudomonas aeruginosa PAO1 is due to FabV, a triclosan-resistant enoyl-acyl carrier protein reductase. Antimicrob. Agents Chemother. 2010, 54 (2), 689– 698, DOI: 10.1128/AAC.01152-09Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhvFSiur4%253D&md5=626edbf70ba35c6ba993ec09a0380eafTriclosan resistance of Pseudomonas aeruginosa PAO1 is due to FabV, a triclosan-resistant enoyl-acyl carrier protein reductaseZhu, Lei; Lin, Jinshui; Ma, Jincheng; Cronan, John E.; Wang, HaihongAntimicrobial Agents and Chemotherapy (2010), 54 (2), 689-698CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Triclosan, a very widely used biocide, specifically inhibits fatty acid synthesis by inhibition of enoyl-acyl carrier protein (ACP) reductase. Escherichia coli FabI is the prototypical triclosan-sensitive enoyl-ACP reductase, and E. coli is extremely sensitive to the biocide. However, other bacteria are resistant to triclosan, because they encode triclosan-resistant enoyl-ACP reductase isoenzymes. In contrast, the triclosan resistance of Pseudomonas aeruginosa PAO1 has been attributed to active efflux of the compd. We report that P. aeruginosa contains two enoyl-ACP reductase isoenzymes, the previously characterized FabI homolog plus a homolog of FabV, a triclosan-resistant enoyl-ACP reductase recently demonstrated in Vibrio cholerae. By deletion of the genes encoding P. aeruginosa FabI and FabV, we demonstrated that FabV confers triclosan resistance on P. aeruginosa. Upon deletion of the fabV gene, the mutant strain became extremely sensitive to triclosan (>2,000-fold more sensitive than the wild-type strain), whereas the mutant strain lacking FabI remained completely resistant to the biocide.
- 30Parsons, J. B.; Frank, M. W.; Subramanian, C.; Saenkham, P.; Rock, C. O. Metabolic basis for the differential susceptibility of Gram-positive pathogens to fatty acid synthesis inhibitors. Proc. Natl. Acad. Sci. U. S. A. 2011, 108 (37), 15378– 15383, DOI: 10.1073/pnas.1109208108Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1KisLrJ&md5=c5f9058eee8fb610fce3934e655b6dadMetabolic basis for the differential susceptibility of gram-positive pathogens to fatty acid synthesis inhibitorsParsons, Joshua B.; Frank, Matthew W.; Subramanian, Chitra; Saenkham, Panatda; Rock, Charles O.Proceedings of the National Academy of Sciences of the United States of America (2011), 108 (37), 15378-15383, S15378/1-S15378/10CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The rationale for the pursuit of bacterial type 2 fatty acid synthesis (FASII) as a target for antibacterial drug discovery in Gram-pos. organisms is being debated vigorously based on their ability to incorporate extracellular fatty acids. The regulation of FASII by extracellular fatty acids was examd. in Staphylococcus aureus and Streptococcus pneumoniae, representing two important groups of pathogens. Both bacteria use the same enzymic tool kit for the conversion of extracellular fatty acids to acyl-acyl carrier protein, elongation, and incorporation into phospholipids. Exogenous fatty acids completely replace the endogenous fatty acids in S. pneumoniae but support only 50% of phospholipid synthesis in S. aureus. Fatty acids overcame FASII inhibition in S. pneumoniae but not in S. aureus. Extracellular fatty acids strongly suppress malonyl-CoA levels in S. pneumoniae but not in S. aureus, showing a feedback regulatory system in S. pneumoniae that is absent in S. aureus. Fatty acids overcame either a biochem. or a genetic block at acetyl-CoA carboxylase (ACC) in S. aureus, confirming that regulation at the ACC step is the key difference between these two species. Bacteria that possess a stringent biochem. feedback inhibition of ACC and malonyl-CoA formation triggered by environmental fatty acids are able to circumvent FASII inhibition. However, if exogenous fatty acids do not suppress malonyl-CoA formation, FASII inhibitors remain effective in the presence of fatty acid supplements.
- 31Zhu, L.; Bi, H.; Ma, J.; Hu, Z.; Zhang, W.; Cronan, J. E.; Wang, H. The two functional enoyl-acyl carrier protein reductases of Enterococcus faecalis do not mediate triclosan resistance. mBio 2013, 4 (5), e00613– 13, DOI: 10.1128/mBio.00613-13Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXntVart7g%253D&md5=4dd4f9fb0167083d7a096ef36e9f0c64The two functional enoyl-acyl carrier protein reductases of Enterococcus faecalis do not mediate triclosan resistanceZhu, Lei; Bi, Hongkai; Ma, Jincheng; Hu, Zhe; Zhang, Wenbin; Cronan, John E.; Wang, HaihongmBio (2013), 4 (5), e00613/1-e00613/11CODEN: MBIOCL; ISSN:2150-7511. (American Society for Microbiology)Enoyl-acyl carrier protein (enoyl-ACP) reductase catalyzes the last step of the elongation cycle in the synthesis of bacterial fatty acids. The Enterococcus faecalis genome contains two genes annotated as enoyl-ACP reductases, a FabI-type enoyl- ACP reductase and a FabK-type enoyl-ACP reductase. The authors report that expression of either of the two proteins restores growth of an Escherichia coli fabI temp.-sensitive mutant strain under nonpermissive conditions. In vitro assays demonstrated that both proteins support fatty acid synthesis and are active with substrates of all fatty acid chain lengths. Although expression of E. faecalis fabK confers to E. coli high levels of resistance to the antimicrobial triclosan, deletion of fabK from the E. faecalis genome showed that FabK does not play a detectable role in the inherent triclosan resistance of E. faecalis. Indeed, FabK seems to play only a minor role in modulating fatty acid compn. Strains carrying a deletion of fabK grow normally without fatty acid supplementation, whereas fabI deletion mutants make only traces of fatty acids and are unsatd. fatty acid auxotrophs.
- 32Yao, J.; Carter, R. A.; Vuagniaux, G.; Barbier, M.; Rosch, J. W.; Rock, C. O. A Pathogen-Selective Antibiotic Minimizes Disturbance to the Microbiome. Antimicrob. Agents Chemother. 2016, 60 (7), 4264– 73, DOI: 10.1128/AAC.00535-16Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVyksLk%253D&md5=0bc461975d3a19a2696bb7cc66f4744fA pathogen-selective antibiotic minimizes disturbance to the microbiomeYao, Jiangwei; Carter, Robert A.; Vuagniaux, Gregoire; Barbier, Maryse; Rosch, Jason W.; Rock, Charles O.Antimicrobial Agents and Chemotherapy (2016), 60 (7), 4264-4273CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)Broad-spectrum antibiotic therapy decimates the gut microbiome, resulting in a variety of neg. health consequences. Debio 1452 is a staphylococcus-selective enoyl-acyl carrier protein reductase (FabI) inhibitor under clin. development and was used to det. whether treatment with pathogen-selective antibiotics would minimize disturbance to the microbiome. The effect of oral Debio 1452 on the microbiota of mice was compared to the effects of four commonly used broad-spectrum oral antibiotics. During the 10 days of oral Debio 1452 treatment, there was minimal disturbance to the gut bacterial abundance and compn., with only the unclassified S24-7 taxon reduced at days 6 and 10. In comparison, broad-spectrum oral antibiotics caused ∼100- to 4000-fold decreases in gut bacterial abundance and severely altered the microbial compn. The gut bacterial abundance and compn. of Debio 1452-treated mice were indistinguishable from those of untreated mice 2 days after the antibiotic treatment was stopped. In contrast, the bacterial abundance in broad-spectrum-antibiotic-treated mice took up to 7 days to recover, and the gut compn. of the broad-spectrum-antibiotic-treated mice remained different from that of the control group 20 days after the cessation of antibiotic treatment. These results illustrate that a pathogen-selective approach to antibiotic development will minimize disturbance to the gut microbiome.
- 33Imai, Y.; Meyer, K. J.; Iinishi, A.; Favre-Godal, Q.; Green, R.; Manuse, S.; Caboni, M.; Mori, M.; Niles, S.; Ghiglieri, M.; Honrao, C.; Ma, X.; Guo, J. J.; Makriyannis, A.; Linares-Otoya, L.; Bohringer, N.; Wuisan, Z. G.; Kaur, H.; Wu, R.; Mateus, A.; Typas, A.; Savitski, M. M.; Espinoza, J. L.; O’Rourke, A.; Nelson, K. E.; Hiller, S.; Noinaj, N.; Schaberle, T. F.; D’Onofrio, A.; Lewis, K. A new antibiotic selectively kills Gram-negative pathogens. Nature 2019, 576 (7787), 459– 464, DOI: 10.1038/s41586-019-1791-1Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlOmurfP&md5=3b7105cfeff0ae8970065bd94e6b12eeA new antibiotic selectively kills Gram-negative pathogensImai, Yu; Meyer, Kirsten J.; Iinishi, Akira; Favre-Godal, Quentin; Green, Robert; Manuse, Sylvie; Caboni, Mariaelena; Mori, Miho; Niles, Samantha; Ghiglieri, Meghan; Honrao, Chandrashekhar; Ma, Xiaoyu; Guo, Jason J.; Makriyannis, Alexandros; Linares-Otoya, Luis; Bohringer, Nils; Wuisan, Zerlina G.; Kaur, Hundeep; Wu, Runrun; Mateus, Andre; Typas, Athanasios; Savitski, Mikhail M.; Espinoza, Josh L.; O'Rourke, Aubrie; Nelson, Karen E.; Hiller, Sebastian; Noinaj, Nicholas; Schaberle, Till F.; D'Onofrio, Anthony; Lewis, KimNature (London, United Kingdom) (2019), 576 (7787), 459-464CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The current need for novel antibiotics is esp. acute for drug-resistant Gram-neg. pathogens1,2. These microorganisms have a highly restrictive permeability barrier, which limits the penetration of most compds.3,4. As a result, the last class of antibiotics that acted against Gram-neg. bacteria was developed in the 1960s2. We reason that useful compds. can be found in bacteria that share similar requirements for antibiotics with humans, and focus on Photorhabdus symbionts of entomopathogenic nematode microbiomes. Here we report a new antibiotic that we name darobactin, which was obtained using a screen of Photorhabdus isolates. Darobactin is coded by a silent operon with little prodn. under lab. conditions, and is ribosomally synthesized. Darobactin has an unusual structure with two fused rings that form post-translationally. The compd. is active against important Gram-neg. pathogens both in vitro and in animal models of infection. Mutants that are resistant to darobactin map to BamA, an essential chaperone and translocator that folds outer membrane proteins. Our study suggests that bacterial symbionts of animals contain antibiotics that are particularly suitable for development into therapeutics.
- 34Leimer, N.; Wu, X.; Imai, Y.; Morrissette, M.; Pitt, N.; Favre-Godal, Q.; Iinishi, A.; Jain, S.; Caboni, M.; Leus, I. V.; Bonifay, V.; Niles, S.; Bargabos, R.; Ghiglieri, M.; Corsetti, R.; Krumpoch, M.; Fox, G.; Son, S.; Klepacki, D.; Polikanov, Y. S.; Freliech, C. A.; McCarthy, J. E.; Edmondson, D. G.; Norris, S. J.; D’Onofrio, A.; Hu, L. T.; Zgurskaya, H. I.; Lewis, K. A selective antibiotic for Lyme disease. Cell 2021, 184 (21), 5405– 5418, DOI: 10.1016/j.cell.2021.09.011Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitF2ntrnO&md5=3eb28b19cf0bfd3e00e4dd374cfe70bbA selective antibiotic for Lyme diseaseLeimer, Nadja; Wu, Xiaoqian; Imai, Yu; Morrissette, Madeleine; Pitt, Norman; Favre-Godal, Quentin; Iinishi, Akira; Jain, Samta; Caboni, Mariaelena; Leus, Inga V.; Bonifay, Vincent; Niles, Samantha; Bargabos, Rachel; Ghiglieri, Meghan; Corsetti, Rachel; Krumpoch, Megan; Fox, Gabriel; Son, Sangkeun; Klepacki, Dorota; Polikanov, Yury S.; Freliech, Cecily A.; McCarthy, Julie E.; Edmondson, Diane G.; Norris, Steven J.; D'Onofrio, Anthony; Hu, Linden T.; Zgurskaya, Helen I.; Lewis, KimCell (Cambridge, MA, United States) (2021), 184 (21), 5405-5418.e16CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Lyme disease is on the rise. Caused by a spirochete Borreliella burgdorferi, it affects an estd. 500,000 people in the United States alone. The antibiotics currently used to treat Lyme disease are broad spectrum, damage the microbiome, and select for resistance in non-target bacteria. We therefore sought to identify a compd. acting selectively against B. burgdorferi. A screen of soil micro-organisms revealed a compd. highly selective against spirochetes, including B. burgdorferi. Unexpectedly, this compd. was detd. to be hygromycin A, a known antimicrobial produced by Streptomyces hygroscopicus. Hygromycin A targets the ribosomes and is taken up by B. burgdorferi, explaining its selectivity. Hygromycin A cleared the B. burgdorferi infection in mice, including animals that ingested the compd. in a bait, and was less disruptive to the fecal microbiome than clin. relevant antibiotics. This selective antibiotic holds the promise of providing a better therapeutic for Lyme disease and eradicating it in the environment.
- 35Poyet, M.; Groussin, M.; Gibbons, S. M.; Avila-Pacheco, J.; Jiang, X.; Kearney, S. M.; Perrotta, A. R.; Berdy, B.; Zhao, S.; Lieberman, T. D.; Swanson, P. K.; Smith, M.; Roesemann, S.; Alexander, J. E.; Rich, S. A.; Livny, J.; Vlamakis, H.; Clish, C.; Bullock, K.; Deik, A.; Scott, J.; Pierce, K. A.; Xavier, R. J.; Alm, E. J. A library of human gut bacterial isolates paired with longitudinal multiomics data enables mechanistic microbiome research. Nat. Med. 2019, 25 (9), 1442– 1452, DOI: 10.1038/s41591-019-0559-3Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12gurrP&md5=016ca835384cb8d48bbc80f21c5284beA library of human gut bacterial isolates paired with longitudinal multiomics data enables mechanistic microbiome researchPoyet, M.; Groussin, M.; Gibbons, S. M.; Avila-Pacheco, J.; Jiang, X.; Kearney, S. M.; Perrotta, A. R.; Berdy, B.; Zhao, S.; Lieberman, T. D.; Swanson, P. K.; Smith, M.; Roesemann, S.; Alexander, J. E.; Rich, S. A.; Livny, J.; Vlamakis, H.; Clish, C.; Bullock, K.; Deik, A.; Scott, J.; Pierce, K. A.; Xavier, R. J.; Alm, E. J.Nature Medicine (New York, NY, United States) (2019), 25 (9), 1442-1452CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)Our understanding of how the gut microbiome interacts with its human host has been restrained by limited access to longitudinal datasets to examine stability and dynamics, and by having only a few isolates to test mechanistic hypotheses. Here, we present the Broad Institute-OpenBiome Microbiome Library (BIO-ML), a comprehensive collection of 7,758 gut bacterial isolates paired with 3,632 genome sequences and longitudinal multi-omics data. We show that microbial species maintain stable population sizes within and across humans and that commonly used 'omics' survey methods are more reliable when using avs. over multiple days of sampling. Variation of gut metabolites within people over time is assocd. with amino acid levels, and differences across people are assocd. with differences in bile acids. Finally, we show that genomic diversification can be used to infer eco-evolutionary dynamics and in vivo selection pressures for strains within individuals. The BIO-ML is a unique resource designed to enable hypothesis-driven microbiome research.
- 36Marrakchi, H.; Dewolf, W. E., Jr.; Quinn, C.; West, J.; Polizzi, B. J.; So, C. Y.; Holmes, D. J.; Reed, S. L.; Heath, R. J.; Payne, D. J.; Rock, C. O.; Wallis, N. G. Characterization of Streptococcus pneumoniae enoyl-(acyl-carrier protein) reductase (FabK). Biochem. J. 2003, 370 (3), 1055– 1062, DOI: 10.1042/bj20021699Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXhvFKhsLc%253D&md5=abef169e126ec1ea70ea8c291db1a3a1Characterization of Streptococcus pneumoniae enoyl-(acyl-carrier protein) reductase (FabK)Marrakchi, Hedia; Dewolf, Walter E., Jr.; Quinn, Chad; West, Joshua; Polizzi, Brian J.; So, Chi Y.; Holmes, David J.; Reed, Shannon L.; Heath, Richard J.; Payne, David J.; Rock, Charles O.; Wallis, Nicola G.Biochemical Journal (2003), 370 (3), 1055-1062CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)The enoyl-(acyl-carrier protein) (ACP) reductase catalyzes the last step in each cycle of fatty acid elongation in the type II fatty acid synthase systems. An extensively characterized NADH-dependent reductase, FabI, is widely distributed in bacteria and plants, whereas the enoyl-ACP reductase, FabK, is a distinctly different member of this enzyme group discovered in Streptococcus pneumoniae . We were unable to delete the fabK gene from Strep. pneumoniae , suggesting that this is the only enoyl-ACP reductase in this organism. The FabK enzyme was purified and the biochem. properties of the reductase were examd. The visible absorption spectrum of the purified protein indicated the presence of a flavin cofactor that was identified as FMN by MS, and was present in a 1:1 molar ratio with protein. FabK specifically required NADH and the protein activity was stimulated by ammonium ions. FabK also exhibited NADH oxidase activity in the absence of substrate. Strep. pneumoniae belongs to the Bacillus/Lactobacillus/Streptococcus group that includes Staphylococcus aureus and Bacillus subtilis . These two organisms also contain FabK-related genes, suggesting that they may also express a FabK-like enoyl-ACP reductase. However, the genes did not complement a fabI (Ts) mutant and the purified flavoproteins were unable to reduce enoyl-ACP in vitro and did not exhibit NAD(P)H oxidase activity, indicating they were not enoyl-ACP reductases. The restricted occurrence of the FabK enoyl-ACP reductase may be related to the role of substrate-independent NADH oxidn. in oxygen-dependent anaerobic energy metab.
- 37Armstrong, J. D.; Eng, K. K.; Keller, J. L.; Purick, R. M.; Hartner, F. W.; Choi, W.-B.; Askin, D.; Volante, R.P. An Efficient Asymmetric Synthesis of (R)-3-Amino-2,3,4,5-tetrahydro-1H-[1]benzazepin-2-one. Tetrahedron Lett. 1994, 35 (20), 3239– 3242, DOI: 10.1016/S0040-4039(00)76874-2Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXkvVejsb4%253D&md5=62d7610af0359f04618b99e65337b5acAn efficient asymmetric synthesis of (R)-3-amino-2,3,4,5-tetrahydro-1H-[1]benzazepin-2-oneArmstrong, Joseph D., III; Eng, Kan K.; Keller, Jennifer L.; Purick, Robert M.; Hartner, Frederick W., Jr.; Choi, Woo-Baeg; Askin, David; Volante, R. P.Tetrahedron Letters (1994), 35 (20), 3239-42CODEN: TELEAY; ISSN:0040-4039.Two approaches for the asym. prepn. of (-)- or (+)-α-aminobenzlactam I are described. One route is based on the asym. hydrogenation of enamide II and the other on the racemization/resoln. of (±)-I.
- 38Galac, M. R.; Snesrud, E.; Lebreton, F.; Stam, J.; Julius, M.; Ong, A. C.; Maybank, R.; Jones, A. R.; Kwak, Y. I.; Hinkle, K.; Waterman, P. E.; Lesho, E. P.; Bennett, J. W.; Mc Gann, P. A Diverse Panel of Clinical Acinetobacter baumannii for Research and Development. Antimicrob. Agents Chemother. 2020, 64 (10), e00840-20, DOI: 10.1128/AAC.00840-20Google ScholarThere is no corresponding record for this reference.
- 39Rao, N. K.; Nataraj, V.; Ravi, M.; Panchariya, L.; Palai, K.; Talapati, S. R.; Lakshminarasimhan, A.; Ramachandra, M.; Antony, T. Ternary complex formation of AFN-1252 with Acinetobacter baumannii FabI and NADH: Crystallographic and biochemical studies. Chem. Biol. Drug Des 2020, 96 (2), 704– 713, DOI: 10.1111/cbdd.13686Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXns1Cnsrs%253D&md5=620921197d60d1f4c194ab5eef70dde8Ternary complex formation of AFN-1252 with Acinetobacter baumannii FabI and NADH: Crystallographic and biochemical studiesRao, Narasimha K.; Nataraj, Vijayashankar; Ravi, Mohan; Panchariya, Love; Palai, Kirttija; Talapati, Sumalatha R.; Lakshminarasimhan, Anirudha; Ramachandra, Murali; Antony, ThomasChemical Biology & Drug Design (2020), 96 (2), 704-713CODEN: CBDDAL; ISSN:1747-0277. (Wiley-Blackwell)Acinetobacter baumannii is an opportunistic Gram-neg. bacterial pathogen, assocd. mostly with hospital-acquired infections. The emergence of drug resistance strains made it necessary to explore new pathways for the development of more effective antibiotics. Enoyl CoA reductase (FabI), a key enzyme in the fatty acid biosynthesis (FAS) pathway, has emerged as a potential target for antibacterial drug development. Earlier reports show that the lead SaFabI inhibitor AFN-1252 can inhibit FabI from other organisms including Escherichia coli and Burkholderia pseudomallei, but with differential potency. In the present work, we show that AFN-1252 is a moderate inhibitor of AbFabI with an IC50 of 216 nM. AFN-1252 stabilized AbFabI with a 4.2°C increase in the melting temp. (Tm) and, interestingly, the stabilization effect was significantly increased in presence of the cofactor NADH (ΔTm = 17°C), suggesting the formation of a ternary complex AbFabI: AFN-1252: NADH. X-ray crystallog. studies of AbFabI co-crystd. with AFN-1252 and NADH confirmed the ternary complex formation. The crit. interactions of AFN-1252 with AbFabI and NADH identified from the co-crystal structure may facilitate the design and development of new drugs against A. baumannii infections by targeting the FAS pathway.
- 40Sihra, N.; Goodman, A.; Zakri, R.; Sahai, A.; Malde, S. Nonantibiotic prevention and management of recurrent urinary tract infection. Nat. Rev. Urol 2018, 15 (12), 750– 776, DOI: 10.1038/s41585-018-0106-xGoogle Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cvjtlGhsg%253D%253D&md5=7bb6b1d6bb8b83bf93269ab7105b964aNonantibiotic prevention and management of recurrent urinary tract infectionSihra Neha; Zakri Rhana; Sahai Arun; Malde Sachin; Goodman AnnaNature reviews. Urology (2018), 15 (12), 750-776 ISSN:.Urinary tract infections (UTIs) are highly prevalent, lead to considerable patient morbidity, incur large financial costs to health-care systems and are one of the most common reasons for antibiotic use worldwide. The growing problem of antimicrobial resistance means that the search for nonantibiotic alternatives for the treatment and prevention of UTI is of critical importance. Potential nonantibiotic measures and treatments for UTIs include behavioural changes, dietary supplementation (such as Chinese herbal medicines and cranberry products), NSAIDs, probiotics, D-mannose, methenamine hippurate, estrogens, intravesical glycosaminoglycans, immunostimulants, vaccines and inoculation with less-pathogenic bacteria. Some of the results of trials of these approaches are promising; however, high-level evidence is required before firm recommendations for their use can be made. A combination of these agents might provide the optimal treatment to reduce recurrent UTI, and trials in specific population groups are required.
- 41Flores-Mireles, A. L.; Walker, J. N.; Caparon, M.; Hultgren, S. J. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat. Rev. Microbiol 2015, 13 (5), 269– 284, DOI: 10.1038/nrmicro3432Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmtVaqs78%253D&md5=b81a25f1f4ab04770982ee01b41b2427Urinary tract infections: epidemiology, mechanisms of infection and treatment optionsFlores-Mireles, Ana L.; Walker, Jennifer N.; Caparon, Michael; Hultgren, Scott J.Nature Reviews Microbiology (2015), 13 (5), 269-284CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)Urinary tract infections (UTIs) are a severe public health problem and are caused by a range of pathogens, but most commonly by Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, Enterococcus faecalis and Staphylococcus saprophyticus. High recurrence rates and increasing antimicrobial resistance among uropathogens threaten to greatly increase the economic burden of these infections. In this Review, we discuss how basic science studies are elucidating the mol. details of the crosstalk that occurs at the host-pathogen interface, as well as the consequences of these interactions for the pathophysiol. of UTIs. We also describe current efforts to translate this knowledge into new clin. treatments for UTIs.
- 42Goebel, M. C.; Trautner, B. W.; Grigoryan, L. The Five Ds of Outpatient Antibiotic Stewardship for Urinary Tract Infections. Clin Microbiol Rev. 2021, 34 (4), e0000320, DOI: 10.1128/CMR.00003-20Google ScholarThere is no corresponding record for this reference.
- 43Nielubowicz, G. R.; Mobley, H. L. Host-pathogen interactions in urinary tract infection. Nat. Rev. Urol 2010, 7 (8), 430– 441, DOI: 10.1038/nrurol.2010.101Google Scholar42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVSmsbzF&md5=84927025a6555c9a93860619e82571ccHost-pathogen interactions in urinary tract infectionNielubowicz, Greta R.; Mobley, Harry L. T.Nature Reviews Urology (2010), 7 (8), 430-441CODEN: NRNADQ; ISSN:1759-4812. (Nature Publishing Group)A review. Urinary tract infections are experienced by a large section of the population and cause significant morbidity. Here, Nielubowicz and Mobley explain the pathogenic mechanisms utilized by Escherichia coli and Proteus mirabilis during complicated and uncomplicated infection, resp. They outline the strategies currently underway to exploit these processes for the identification of new treatment options. The urinary tract is a common site of bacterial infections; nearly half of all women experience at least one urinary tract infection (UTI) during their lifetime. These infections are classified based on the condition of the host. Uncomplicated infections affect otherwise healthy individuals and are most commonly caused by uropathogenic Escherichia coli, whereas complicated infections affect patients with underlying difficulties, such as a urinary tract abnormality or catheterization, and are commonly caused by species such as Proteus mirabilis. Virulence and fitness factors produced by both pathogens include fimbriae, toxins, flagella, iron acquisition systems, and proteins that function in immune evasion. Addnl. factors that contribute to infection include the formation of intracellular bacterial communities by E. coli and the prodn. of urease by P. mirabilis, which can result in urinary stone formation. Innate immune responses are induced or mediated by pattern recognition receptors, antimicrobial peptides, and neutrophils. The adaptive immune response to UTI is less well understood. Host factors TLR4 and CXCR1 are implicated in disease outcome and susceptibility, resp. Low levels of TLR4 are assocd. with asymptomatic bacteriuria while low levels of CXCR1 are assocd. with increased incidence of acute pyelonephritis. Current research is focused on the identification of addnl. virulence factors and therapeutic or prophylactic targets that might be used in the generation of vaccines against both uropathogens.
- 44Flamm, R. K.; Rhomberg, P. R.; Kaplan, N.; Jones, R. N.; Farrell, D. J. Activity of Debio1452, a FabI inhibitor with potent activity against Staphylococcus aureus and coagulase-negative Staphylococcus spp., including multidrug-resistant strains. Antimicrob. Agents Chemother. 2015, 59 (5), 2583– 2587, DOI: 10.1128/AAC.05119-14Google Scholar43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntVGjsbw%253D&md5=9b0e6360938e855fe2513d48d8016cacActivity of Debio1452, a FabI inhibitor with potent activity against Staphylococcus aureus and coagulase-negative Staphylococcus spp., including multidrug-resistant strainsFlamm, Robert K.; Rhomberg, Paul R.; Kaplan, Nachum; Jones, Ronald N.; Farrell, David J.Antimicrobial Agents and Chemotherapy (2015), 59 (5), 2583-2587CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)Staphylococcus aureus and coagulase-neg. staphylococci (CoNS) are responsible for a wide variety of human infections. The investigational antibacterial Debio1450 (previously AFN-1720), a prodrug of Debio1452 (previously AFN-1252), specifically targets staphylococci without significant activity against other Gram-pos. or Gram-neg. species. Debio1452 inhibits FabI, an enzyme crit. to fatty acid biosynthesis in staphylococci. The activity of Debio1452 against CoNS, methicillin-susceptible S. aureus (MSSA), and methicillin-resistant S. aureus (MRSA), including significant clones, was detd. A globally diverse collection of 574 patient isolates from 35 countries was tested that included CoNS (6 species, 103 strains), MSSA (154 strains), MRSA (163 strains), and molecularly characterized strains (including spa-typed MRSA clones; 154 strains). The isolates were tested for susceptibility by CLSI broth microdilution methods against Debio1452 and 10 comparators. The susceptibility rates for the comparators were detd. using CLSI and EUCAST breakpoint criteria. All S. aureus and CoNS strains were inhibited by Debio1452 concns. of ≤0.12 and ≤0.5 μg/mL, resp. The MIC50s for MSSA, MRSA, and molecularly characterized MRSA strains were 0.004 μg/mL, and the MIC90s ranged from 0.008 to 0.03 μg/mL. The MICs were higher for the CoNS isolates (MIC50/90, 0.015/0.12 μg/mL). Among S. aureus strains, resistance was common for erythromycin (61.6%), levofloxacin (49.0%), clindamycin (27.6%), tetracycline (15.7%), and trimethoprim-sulfamethoxazole (7.0%). Debio1452 demonstrated potent activity against MSSA, MRSA, and CoNS. Debio1452 showed significantly greater activity overall (MIC50, 0.004 μg/mL) than the other agents tested against these staphylococcal species, which included dominant MRSA clones and strains resistant to currently used antimicrobial agents.
- 45Hafkin, B.; Kaplan, N.; Murphy, B. Efficacy and Safety of AFN-1252, the First Staphylococcus-Specific Antibacterial Agent, in the Treatment of Acute Bacterial Skin and Skin Structure Infections, Including Those in Patients with Significant Comorbidities. Antimicrob. Agents Chemother. 2016, 60 (3), 1695– 701, DOI: 10.1128/AAC.01741-15Google Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsF2ntbnF&md5=a108d5ddd834788bbdb10d57dd07af4eEfficacy and safety of AFN-1252, the first Staphylococcus-specific antibacterial agent, in the treatment of acute bacterial skin and skin structure infections, including those in patients with significant comorbiditiesHafkin, B.; Kaplan, N.; Murphy, B.Antimicrobial Agents and Chemotherapy (2016), 60 (3), 1695-1701CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)This open-label noncontrolled, phase II multicenter trial was designed to evaluate the safety, tolerability, and efficacy of 200 mg of AFN-1252, a selective inhibitor of Staphylococcus aureus enoyl-acyl carrier protein reductase (FabI), given by mouth twice daily in the treatment of acute bacterial skin and skin structure infections (ABSSSI) due to staphylococci. Important aspects of the current study included a comparison of early response efficacy endpoints with end-of-treatment and follow-up endpoints. Many patients in the intent-to-treat population (n = 103) had significant comorbidities. The overall early response rate at day 3 was 97.3% (wound, 100%; abscess, 96.6%; cellulitis, 94.4%) in the microbiol. evaluable (ME) population. Within the ME population, 82.9% of patients had a ≥ 20% decrease in the area of erythema, and 77.9% of patients had a ≥ 20% decrease in the area of induration, on day 3. S. aureus was detected in 97.7% of patients (n = 37 patients with methicillin-resistant S. aureus [MRSA], and n = 39 with methicillin-sensitive S. aureus [MSSA]). No isolates had increased AFN-1252 MICs posttreatment. Microbiol. eradication rates for S. aureus were 93.2% at short-term follow-up (STFU) and 91.9% at long-term follow-up (LTFU) in the ME population. Eradication rates for MRSA and MSSA were 91.9% and 92.3%, resp., at STFU and 91.9% and 89.7%, resp., at LTFU. The most frequently reported drug-related adverse events, which were mostly mild or moderate, were headache (26.2%) and nausea (21.4%). These studies demonstrate that AFN-1252 is generally well tolerated and effective in the treatment of ABSSSI due to S. aureus, including MRSA. This study has been registered at ClinicalTrials.gov under registration no.NCT01519492.
- 46Radka, C. D.; Rock, C. O. Mining Fatty Acid Biosynthesis for New Antimicrobials. Annu. Rev. Microbiol. 2022, 76, DOI: 10.1146/annurev-micro-041320-110408Google ScholarThere is no corresponding record for this reference.
- 47Payne, D. J.; Warren, P. V.; Holmes, D. J.; Ji, Y.; Lonsdale, J. T. Bacterial fatty-acid biosynthesis: a genomics-driven target for antibacterial drug discovery. Drug Discov Today 2001, 6 (10), 537– 544, DOI: 10.1016/S1359-6446(01)01774-3Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXjtlCjsbo%253D&md5=50cbe09ba8e48f9aca6a5b847477f24cBacterial fatty-acid biosynthesis: a genomics-driven target for antibacterial drug discoveryPayne, D. J.; Warren, P. V.; Holmes, D. J.; Ji, Y.; Lonsdale, J. T.Drug Discovery Today (2001), 6 (10), 537-544CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Science Ltd.)A review with 46 refs. demonstrating how the interplay of genomics, bioinformatics and genomic technologies has enabled an in-depth anal. of the component enzymes of the bacterial fatty-acid biosynthesis pathway as a source of novel antibacterial targets. This evaluation has revealed that many of the enzymes are potentially selective, broad-spectrum antibacterial targets. We also illustrate the suitability of some of these targets for HTS. Furthermore, we discuss how the availability of a robust selectivity assay, mode-of-action assays and numerous crystal structures provide an excellent set of tools with which to initiate integrated programs of research to identify novel antibiotics targeted at these enzymes.
- 48Yao, J.; Rock, C. O. Bacterial fatty acid metabolism in modern antibiotic discovery. Biochim Biophys Acta Mol. Cell Biol. Lipids 2017, 1862 (11), 1300– 1309, DOI: 10.1016/j.bbalip.2016.09.014Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFKmur3E&md5=c6b0c7f2c7997bed206046b62041e965Bacterial fatty acid metabolism in modern antibiotic discoveryYao, Jiangwei; Rock, Charles O.Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2017), 1862 (11), 1300-1309CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B.V.)Bacterial fatty acid synthesis is essential for many pathogens and different from the mammalian counterpart. These features make bacterial fatty acid synthesis a desirable target for antibiotic discovery. The structural divergence of the conserved enzymes and the presence of different isoenzymes catalyzing the same reactions in the pathway make bacterial fatty acid synthesis a narrow spectrum target rather than the traditional broad spectrum target. Furthermore, bacterial fatty acid synthesis inhibitors are single-targeting, rather than multi-targeting like traditional monotherapeutic, broad-spectrum antibiotics. The single-targeting nature of bacterial fatty acid synthesis inhibitors makes overcoming fast-developing, target-based resistance a necessary consideration for antibiotic development. Target-based resistance can be overcome through multi-targeting inhibitors, a cocktail of single-targeting inhibitors, or by making the single targeting inhibitor sufficiently high affinity through a pathogen selective approach such that target-based mutants are still susceptible to therapeutic concns. of drug. Many of the pathogens requiring new antibiotic treatment options encode for essential bacterial fatty acid synthesis enzymes. This review will evaluate the most promising targets in bacterial fatty acid metab. for antibiotic therapeutics development and review the potential and challenges in advancing each of these targets to the clinic and circumventing target-based resistance. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
- 49Staley, C.; Vaughn, B. P.; Graiziger, C. T.; Sadowsky, M. J.; Khoruts, A. Gut-sparing treatment of urinary tract infection in patients at high risk of Clostridium difficile infection. J. Antimicrob. Chemother. 2017, 72 (2), 522– 528, DOI: 10.1093/jac/dkw499Google Scholar48https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFKju7nK&md5=1d8a5c68e27d005d9c0a022569efb66eGut-sparing treatment of urinary tract infection in patients at high risk of Clostridium difficile infectionStaley, Christopher; Vaughn, Byron P.; Graiziger, Carolyn T.; Sadowsky, Michael J.; Khoruts, AlexanderJournal of Antimicrobial Chemotherapy (2017), 72 (2), 522-528CODEN: JACHDX; ISSN:1460-2091. (Oxford University Press)Recipients of fecal microbiota transplantation (FMT) in treatment of recurrent Clostridium difficile infection (RCDI) remain at markedly increased risk of re-infection with C. difficile with new antibiotic provocations. Urinary tract infections (UTIs) are common indications for antibiotics in these patients, often resulting in C. difficile re-infection. We present a case series of 19 patients treated with parenteral aminoglycosides for UTI following FMT for RCDI. A 3 day outpatient regimen of once-daily i.m. administration of gentamicin was used to treat 18 consecutive FMT recipients with uncomplicated UTI. One other patient was treated for a complicated UTI with i.v. amikacin. Profiling of 16S rRNA genes was used to track changes in fecal microbial community structure during this regimen in three patients. The protocol was highly effective in treating UTI symptoms. None of the patients suffered a re-infection with C. difficile. The fecal microbial communities remained undisturbed by treatment with i.m. administration of gentamicin. Despite falling out of favor in recent years, aminoglycoside antibiotics given parenterally have the advantage of minimal penetration into the gut lumen. A brief (3 day) course of parenteral gentamicin was safe and effective in curing UTI in patients at high risk of C. difficile infection without perturbing their gut microbiota.
- 50Worby, C. J.; Schreiber, H. L. t.; Straub, T. J.; van Dijk, L. R.; Bronson, R. A.; Olson, B. S.; Pinkner, J. S.; Obernuefemann, C. L. P.; Munoz, V. L.; Paharik, A. E.; Azimzadeh, P. N.; Walker, B. J.; Desjardins, C. A.; Chou, W. C.; Bergeron, K.; Chapman, S. B.; Klim, A.; Manson, A. L.; Hannan, T. J.; Hooton, T. M.; Kau, A. L.; Lai, H. H.; Dodson, K. W.; Hultgren, S. J.; Earl, A. M. Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in women. Nat. Microbiol 2022, 7 (5), 630– 639, DOI: 10.1038/s41564-022-01107-xGoogle Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFyqtrzF&md5=a5458d92f0b40871578f476e448e9ae2Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in womenWorby, Colin J.; Schreiber IV, Henry L.; Straub, Timothy J.; van Dijk, Lucas R.; Bronson, Ryan A.; Olson, Benjamin S.; Pinkner, Jerome S.; Obernuefemann, Chloe L. P.; Munoz, Vanessa L.; Paharik, Alexandra E.; Azimzadeh, Philippe N.; Walker, Bruce J.; Desjardins, Christopher A.; Chou, Wen-Chi; Bergeron, Karla; Chapman, Sinead B.; Klim, Aleksandra; Manson, Abigail L.; Hannan, Thomas J.; Hooton, Thomas M.; Kau, Andrew L.; Lai, H. Henry; Dodson, Karen W.; Hultgren, Scott J.; Earl, Ashlee M.Nature Microbiology (2022), 7 (5), 630-639CODEN: NMAICH; ISSN:2058-5276. (Nature Portfolio)Abstr.: Recurrent urinary tract infections (rUTIs) are a major health burden worldwide, with history of infection being a significant risk factor. While the gut is a known reservoir for uropathogenic bacteria, the role of the microbiota in rUTI remains unclear. We conducted a year-long study of women with (n = 15) and without (n = 16) history of rUTI, from whom we collected urine, blood and monthly faecal samples for metagenomic and transcriptomic interrogation. During the study 24 UTIs were reported, with addnl. samples collected during and after infection. The gut microbiome of individuals with a history of rUTI was significantly depleted in microbial richness and butyrate-producing bacteria compared with controls, reminiscent of other inflammatory conditions. However, Escherichia coli gut and bladder populations were comparable between cohorts in both relative abundance and phylogroup. Transcriptional anal. of peripheral blood mononuclear cells revealed expression profiles indicative of differential systemic immunity between cohorts. Altogether, these results suggest that rUTI susceptibility is in part mediated through the gut-bladder axis, comprising gut dysbiosis and differential immune response to bacterial bladder colonization, manifesting in symptoms.
- 51Schembri, M. A.; Nhu, N. T. K.; Phan, M. D. Gut-bladder axis in recurrent UTI. Nat. Microbiol 2022, 7 (5), 601– 602, DOI: 10.1038/s41564-022-01113-zGoogle Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFyqtr3M&md5=7ff22d3120b3ec5116cfdfc5ad91458bGut-bladder axis in recurrent UTISchembri, Mark A.; Nhu, Nguyen Thi Khanh; Phan, Minh-DuyNature Microbiology (2022), 7 (5), 601-602CODEN: NMAICH; ISSN:2058-5276. (Nature Portfolio)Dysbiosis of the gut microbiome is assocd. with increased susceptibility to recurrent urinary tract infection, defining a gut-bladder axis.
- 52Njoroge, M.; Kaur, G.; Espinoza-Moraga, M.; Wasuna, A.; Dziwornu, G. A.; Seldon, R.; Taylor, D.; Okombo, J.; Warner, D. F.; Chibale, K. Semisynthetic Antimycobacterial C-3 Silicate and C-3/C-21 Ester Derivatives of Fusidic Acid: Pharmacological Evaluation and Stability Studies in Liver Microsomes, Rat Plasma, and Mycobacterium tuberculosis culture. ACS Infect Dis 2019, 5 (9), 1634– 1644, DOI: 10.1021/acsinfecdis.9b00208Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlOmsbfP&md5=e1362a952f1eecb77341cb9386a03872Semisynthetic Antimycobacterial C-3 Silicate and C-3/C-21 Ester Derivatives of Fusidic Acid: Pharmacological Evaluation and Stability Studies in Liver Microsomes, Rat Plasma, and Mycobacterium tuberculosis cultureNjoroge, Mathew; Kaur, Gurminder; Espinoza-Moraga, Marlene; Wasuna, Antonina; Dziwornu, Godwin Akpeko; Seldon, Ronnett; Taylor, Dale; Okombo, John; Warner, Digby F.; Chibale, KellyACS Infectious Diseases (2019), 5 (9), 1634-1644CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Fusidic acid (FA), a natural product fusidane triterpene-based antibiotic with unique structural features, is active in vitro against Mycobacterium tuberculosis, the causative agent of tuberculosis (TB). While possessing good pharmacokinetics in man, FA is rapidly metabolized in rodents, thus complicating proof-of-concept studies in this model. Toward the repositioning of FA as an anti-TB agent, we herein describe the synthesis, activity, and metab. of FA and semisynthesized ester derivs. in rat liver microsomes, rat plasma, and mycobacterial cell culture. FA and deriv. mols. with a free C-3 OH underwent species-specific metab. to the corresponding 3-OH epimer, 3-epifusidic acid (3-epiFA). FA was also metabolized in rat plasma to form FA lactone. These addnl. routes of metab. may contribute to the more rapid clearance of FA obsd. in rodents. C-3 alkyl and aryl esters functioned as classic prodrugs of FA, being hydrolyzed to FA in microsomes, plasma, and Mycobacterium tuberculosis culture. In contrast, C-3 silicate esters and C-21 esters were inert to hydrolysis and so did not act as prodrugs. The antimycobacterial activity of the C-3 silicate esters was comparable to that of FA, and these compds. were stable in microsomes and plasma, identifying them as potential candidates for evaluation in a rodent model of tuberculosis.
- 53Smith, P. W.; Zuccotto, F.; Bates, R. H.; Martinez-Martinez, M. S.; Read, K. D.; Peet, C.; Epemolu, O. Pharmacokinetics of beta-Lactam Antibiotics: Clues from the Past To Help Discover Long-Acting Oral Drugs in the Future. ACS Infect Dis 2018, 4 (10), 1439– 1447, DOI: 10.1021/acsinfecdis.8b00160Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFOrtbfF&md5=ca8591280316b2c1733e911feb5f0150Pharmacokinetics of β-Lactam Antibiotics: Clues from the Past To Help Discover Long-Acting Oral Drugs in the FutureSmith, Paul W.; Zuccotto, Fabio; Bates, Robert H.; Martinez-Martinez, Maria Santos; Read, Kevin D.; Peet, Caroline; Epemolu, OlaACS Infectious Diseases (2018), 4 (10), 1439-1447CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A review. β-Lactams represent perhaps the most important class of antibiotics yet discovered. However, despite many years of active research, none of the currently approved drugs in this class combine oral activity with long duration of action. Recent developments suggest that new β-lactam antibiotics with such a profile would have utility in the treatment of tuberculosis. Consequently, the historical β-lactam pharmacokinetic data have been compiled and analyzed to identify possible directions and drug discovery strategies aimed toward new β-lactam antibiotics with this profile.
Cited By
This article is cited by 40 publications.
- Mintesinot Kassu, Katelyn E. Zulauf, Jessica N. Ross, James E. Kirby, Roman Manetsch. Development of CGS-15943 Adjunctives for the Disruption of Plasmid Maintenance in Multidrug Resistant E. coli. ACS Infectious Diseases 2025, 11
(1)
, 80-87. https://doi.org/10.1021/acsinfecdis.4c00587
- Irena Audzeyenka, Agnieszka Piwkowska, Dorota Rogacka, Mariusz Makowski, Mateusz Kowalik. Biological Evaluation of a Rhodium(III) Bipyridylsulfonamide Complex: Effects on Mitochondrial Dynamics and Cytoskeletal Remodeling in Breast Cancer Cells. Journal of Medicinal Chemistry 2024, 67
(23)
, 21364-21379. https://doi.org/10.1021/acs.jmedchem.4c02284
- Abhishek Acharya, Pratik Kumar Behera, Ulrich Kleinekathöfer. Molecular Mechanism of Ciprofloxacin Translocation Through the Major Diffusion Channels of the ESKAPE Pathogens Klebsiella pneumoniae and Enterobacter cloacae. The Journal of Physical Chemistry B 2024, 128
(35)
, 8376-8387. https://doi.org/10.1021/acs.jpcb.4c03327
- Mateusz Rzycki, Dominik Drabik. Multifaceted Activity of Fabimycin: Insights from Molecular Dynamics Studies on Bacterial Membrane Models. Journal of Chemical Information and Modeling 2024, 64
(10)
, 4204-4217. https://doi.org/10.1021/acs.jcim.4c00228
- Rachel M. Johnson, Kelin Li, Xiaoyan Chen, Gina L. Morgan, Jeffrey Aubé, Bo Li. The Hybrid Antibiotic Thiomarinol A Overcomes Intrinsic Resistance in Escherichia coli Using a Privileged Dithiolopyrrolone Moiety. ACS Infectious Diseases 2024, 10
(2)
, 582-593. https://doi.org/10.1021/acsinfecdis.3c00504
- Abhishek Acharya, Kalyanashis Jana, Ulrich Kleinekathöfer. Antibiotic Charge Profile Determines the Extent of L3 Dynamics in OmpF: An Expedited Passage for Molecules with a Positive Charge. The Journal of Physical Chemistry B 2023, 127
(50)
, 10766-10777. https://doi.org/10.1021/acs.jpcb.3c04557
- Harsha Valluri, Amritansh Bhanot, Shreeya Shah, Navya Bhandaru, Sandeep Sundriyal. Basic Nitrogen (BaN) Is a Key Property of Antimalarial Chemical Space. Journal of Medicinal Chemistry 2023, 66
(13)
, 8382-8406. https://doi.org/10.1021/acs.jmedchem.3c00206
- Vinicius Gonçalves Maltarollo, Ekaterina Shevchenko, Igor Daniel de Miranda Lima, Elio A. Cino, Glaucio Monteiro Ferreira, Antti Poso, Thales Kronenberger. Do Go Chasing Waterfalls: Enoyl Reductase (FabI) in Complex with Inhibitors Stabilizes the Tetrameric Structure and Opens Water Channels. Journal of Chemical Information and Modeling 2022, 62
(22)
, 5746-5761. https://doi.org/10.1021/acs.jcim.2c01178
- Ziyang Zhang. Hacking the Permeability Barrier of Gram-Negative Bacteria. ACS Central Science 2022, 8
(8)
, 1043-1046. https://doi.org/10.1021/acscentsci.2c00750
- Zhenlong Zhou, Ruoxin Chen, Pengzhen Li, Penghui Fan, Lin Ma, Xinyu Cai, Yuchao Hou, Binbin Li, Jianyu Su. Natural borneol improves cellular uptake of curcumin to enhance its photodynamic bactericidal activity against Escherichia coli ATCC 8739. Food Microbiology 2025, 127 , 104686. https://doi.org/10.1016/j.fm.2024.104686
- Shubhi Singh, Sahithya Selvakumar, Priya Swaminathan. A comprehensive computer-based assessment of Deacetylnomilin as an inhibitor for antibiotic-resistant genes identified from the whole genome sequence of the multidrug-resistant Enterobacter cloacae isolate 1382. Molecular Diversity 2024, 399 https://doi.org/10.1007/s11030-024-11077-3
- Jiekai Sun, Xu Wang, Ye Gao, Shuangyu Li, Ziwei Hu, Yan Huang, Baoqiang Fan, Xia Wang, Miao Liu, Chunhua Qiao, Wei Zhang, Yipeng Wang, Xingyue Ji. H2S scavenger as a broad-spectrum strategy to deplete bacteria-derived H2S for antibacterial sensitization. Nature Communications 2024, 15
(1)
https://doi.org/10.1038/s41467-024-53764-7
- Norhan H. Mahdally, Abdalla E. M. Salih, Riham A. El-Shiekh, Ahmed M. Sayed, Noha M. Elhosseiny, Mona T. Kashef, Mohammed Yaseen, William Mackay, Ali M. El Halawany, Mostafa E. Rateb, Ahmed S. Attia. Exploring the antimicrobial activity of Origanum majorana L. against the highly virulent multidrug-resistant Acinetobacter baumannii AB5075: UPLC-HRMS profiling with in vitro and in silico studies. Future Journal of Pharmaceutical Sciences 2024, 10
(1)
https://doi.org/10.1186/s43094-024-00641-1
- Eduardo Hernández-Vázquez, Ángel Ramírez-Trinidad, César E. Tovar-Román, José A. Rivera Chávez, Elizabeth Huerta-Salazar. N-acyl-4-arylaminopiperidines: Design and synthesis of a potential antimicrobial scaffold. Bioorganic & Medicinal Chemistry Letters 2024, 112 , 129936. https://doi.org/10.1016/j.bmcl.2024.129936
- S. Raghunadh Acharyulu, N. Srinivasu, Sivaranjani Jampala, Arundhuthi M. An Exquisite Design Process and Impacting Synthesis of 2-methyl Nicotinamide Derivatives and their Anti-bacterial Activity Closet to Fab I Inhibitors. Oriental Journal Of Chemistry 2024, 40
(5)
, 1250-1260. https://doi.org/10.13005/ojc/400506
- Okan Yildirim, Dipti Barman, Mia Chung, Samantha Stone, Raphael Geißen, Melissa L. Boby, Bradley S. Sherborne, Derek S. Tan. Design and synthesis of a library of C8-substituted sulfamidoadenosines to probe bacterial permeability. Bioorganic & Medicinal Chemistry Letters 2024, 110 , 129844. https://doi.org/10.1016/j.bmcl.2024.129844
- Francesca Palma, Marina Acunzo, Roberta Della Marca, Federica Dell’Annunziata, Veronica Folliero, Annalisa Chianese, Carla Zannella, Gianluigi Franci, Anna De Filippis, Massimiliano Galdiero. Evaluation of antifungal spectrum of Cupferron against Candida albicans. Microbial Pathogenesis 2024, 194 , 106835. https://doi.org/10.1016/j.micpath.2024.106835
- Fatma M. Abdel Bar, Engy Elekhnawy, Taif H. Aldawsari, Shatha F. Alkanhal, Raghad M. Alanazi, Ghida A. Al-Akeel, Mai H. ElNaggar. Unveiling the role of macranthoin G in the traditional anti-infective properties of Launaea nudicaulis. South African Journal of Botany 2024, 172 , 302-311. https://doi.org/10.1016/j.sajb.2024.07.043
- Kellen Christina Malheiros Borges, André Kipnis, Bruno Junior Neves, Ana Paula Junqueira-Kipnis. Promising New Targets for the Treatment of Infections Caused by Acinetobacter baumannii: A Review. Current Drug Targets 2024, 25
(14)
, 971-986. https://doi.org/10.2174/0113894501319269240819060245
- Joanna Krajewska, Stefan Tyski, Agnieszka E. Laudy. In Vitro Resistance-Predicting Studies and In Vitro Resistance-Related Parameters—A Hit-to-Lead Perspective. Pharmaceuticals 2024, 17
(8)
, 1068. https://doi.org/10.3390/ph17081068
- Susannah L. Parkhill, Eachan O. Johnson. Integrating bacterial molecular genetics with chemical biology for renewed antibacterial drug discovery. Biochemical Journal 2024, 481
(13)
, 839-864. https://doi.org/10.1042/BCJ20220062
- Kristen A. Muñoz, Rebecca J. Ulrich, Archit K. Vasan, Matt Sinclair, Po-Chao Wen, Jessica R. Holmes, Hyang Yeon Lee, Chien-Che Hung, Christopher J. Fields, Emad Tajkhorshid, Gee W. Lau, Paul J. Hergenrother. A Gram-negative-selective antibiotic that spares the gut microbiome. Nature 2024, 630
(8016)
, 429-436. https://doi.org/10.1038/s41586-024-07502-0
- Paprapach Wongdontree, Aaron Millan-Oropeza, Jennifer Upfold, Jean-Pierre Lavergne, David Halpern, Clara Lambert, Adeline Page, Gérald Kénanian, Christophe Grangeasse, Céline Henry, Agnès Fouet, Karine Gloux, Jamila Anba-Mondoloni, Alexandra Gruss. Oxidative stress is intrinsic to staphylococcal adaptation to fatty acid synthesis antibiotics. iScience 2024, 27
(4)
, 109505. https://doi.org/10.1016/j.isci.2024.109505
- Brett N. Cain, Paul J. Hergenrother. Using permeation guidelines to design new antibiotics—A PASsagE into
Pseudomonas aeruginosa. Clinical and Translational Medicine 2024, 14
(3)
https://doi.org/10.1002/ctm2.1600
- Soumya Biswas, Anupam Patra, Prajita Paul, Namrata Misra, Gajraj Singh Kushwaha, Mrutyunjay Suar. Structural and Biochemical Studies on Klebsiella Pneumoniae Enoyl-ACP Reductase (FabI) Suggest Flexible Substrate Binding Site. The Protein Journal 2024, 43
(1)
, 84-95. https://doi.org/10.1007/s10930-023-10176-8
- Shibin Zhao, Julian Maceren, Mia Chung, Samantha Stone, Raphael Geißen, Melissa L. Boby, Bradley S. Sherborne, Derek S. Tan. Design and synthesis of a library of C2-substituted sulfamidoadenosines to probe bacterial permeability. Bioorganic & Medicinal Chemistry Letters 2024, 97 , 129486. https://doi.org/10.1016/j.bmcl.2023.129486
- Eduardo Hernández-Vázquez, Angel Ramírez Trinidad, Cesar E. Tovar-Roman, José Rivera-Chávez, Elizabeth Huerta-Salazar. Design and Synthesis of N-Acyl-4-Arylaminopiperidines as Potential Compounds Against Multiresistant Bacteria. 2024https://doi.org/10.2139/ssrn.4828092
- Emily J. Geddes, Morgan K. Gugger, Alfredo Garcia, Martin Garcia Chavez, Myung Ryul Lee, Sarah J. Perlmutter, Christoph Bieniossek, Laura Guasch, Paul J. Hergenrother. Porin-independent accumulation in Pseudomonas enables antibiotic discovery. Nature 2023, 624
(7990)
, 145-153. https://doi.org/10.1038/s41586-023-06760-8
- Ursula Theuretzbacher, Benjamin Blasco, Maëlle Duffey, Laura J. V. Piddock. Unrealized targets in the discovery of antibiotics for Gram-negative bacterial infections. Nature Reviews Drug Discovery 2023, 22
(12)
, 957-975. https://doi.org/10.1038/s41573-023-00791-6
- Harpreet Kaur, Manmohit Kalia, Vikram Singh, Neelam Taneja. Identification of novel inhibitors against
Escherichia coli
utilizing HisC as a target from histidine biosynthesis pathway. Journal of Biomolecular Structure and Dynamics 2023, 41
(19)
, 9907-9914. https://doi.org/10.1080/07391102.2022.2148319
- Francesca Palma, Federica Dell'Annunziata, Veronica Folliero, Francesco Foglia, Roberta Della Marca, Carla Zannella, Anna De Filippis, Gianluigi Franci, Massimiliano Galdiero. Cupferron impairs the growth and virulence of
Escherichia coli
clinical isolates. Journal of Applied Microbiology 2023, 134
(10)
https://doi.org/10.1093/jambio/lxad222
- Mahdi Redha, Noura Al Sweih, M. Albert. Multidrug-Resistant and Extensively Drug-Resistant Escherichia coli in Sewage in Kuwait: Their Implications. Microorganisms 2023, 11
(10)
, 2610. https://doi.org/10.3390/microorganisms11102610
- Francesca Ruggieri, Nina Compagne, Kevin Antraygues, Maxime Eveque, Marion Flipo, Nicolas Willand. Antibiotics with novel mode of action as new weapons to fight antimicrobial resistance. European Journal of Medicinal Chemistry 2023, 256 , 115413. https://doi.org/10.1016/j.ejmech.2023.115413
- Abhishek Acharya, Kalyanashis Jana, Dominik Gurvic, Ulrich Zachariae, Ulrich Kleinekathöfer. Fast prediction of antibiotic permeability through membrane channels using Brownian dynamics. Biophysical Journal 2023, 122
(14)
, 2996-3007. https://doi.org/10.1016/j.bpj.2023.03.035
- Lindsey A. Carfrae, Kenneth Rachwalski, Shawn French, Rodion Gordzevich, Laura Seidel, Caressa N. Tsai, Megan M. Tu, Craig R. MacNair, Olga G. Ovchinnikova, Bradley R. Clarke, Chris Whitfield, Eric D. Brown. Inhibiting fatty acid synthesis overcomes colistin resistance. Nature Microbiology 2023, 8
(6)
, 1026-1038. https://doi.org/10.1038/s41564-023-01369-z
- S. N. Gavrilov, A. S. Barashkova, T. A. Cherdyntseva, M. I. Prokofeva, O. V. Tresvyatskii, D. A. Lukianov, A. A. Nikandrova, T. Haertlé, A. Yu. Merkel, E. A. Bonch-Osmolovskaya, E. A. Rogozhin. Search for Novel Halophilic and Halotolerant Producers of Antimicrobial Compounds in Various Extreme Ecosystems. Microbiology 2023, 92
(3)
, 342-357. https://doi.org/10.1134/S0026261723600313
- Margot Draveny, Clémence Rose, Alexis Pinet, Laurent Ferrié, Bruno Figadère, Jean-Michel Brunel, Muriel Masi. Scope and Limitations of Exploiting the Ability of the Chemosensitizer NV716 to Enhance the Activity of Tetracycline Derivatives against Pseudomonas aeruginosa. Molecules 2023, 28
(11)
, 4262. https://doi.org/10.3390/molecules28114262
- S. N. Gavrilov, A. S. Barashkova, T. A. Cherdyntseva, M. I. Prokofeva, O. V. Tresvyatskii, D. A. Lukianov, A. A. Nikandrova, T. Haertle, A. Yu. Merkel, E. A. Bonch-Osmolovskaya, E. A. Rogozhin. Search for Novel Halophilic and Halotolerant Producers of Antimicrobial Compounds in Various Extreme Ecosystems. Микробиология 2023, 92
(3)
, 261-278. https://doi.org/10.31857/S0026365622600845
- Laurie Bibens, Jean-Paul Becker, Alexandra Dassonville-Klimpt, Pascal Sonnet. A Review of Fatty Acid Biosynthesis Enzyme Inhibitors as Promising Antimicrobial Drugs. Pharmaceuticals 2023, 16
(3)
, 425. https://doi.org/10.3390/ph16030425
- Yuzhen Zhang, Yumin Cai, Tianmin Wang, Qile Wu. A common tolerance mechanism of bacterial biofilms to antibiotics. 2023https://doi.org/10.1101/2023.01.30.526163
- . The Fabulous Fabimycin. Synfacts 2022, 1367. https://doi.org/10.1055/s-0042-1753108
- Gustavo Schottlender, Juan Manuel Prieto, Miranda Clara Palumbo, Florencia A. Castello, Federico Serral, Ezequiel J. Sosa, Adrián G. Turjanski, Marcelo A. Martì, Darío Fernández Do Porto. From drugs to targets: Reverse engineering the virtual screening process on a proteomic scale. Frontiers in Drug Discovery 2022, 2 https://doi.org/10.3389/fddsv.2022.969983
- Paprapach Wongdontree, Aaron Millan-Oropeza, Jennifer Upfold, Jean-Pierre Lavergne, David Halpern, Clara Lambert, Adeline Page, Gérald Kénanian, Christophe Grangeasse, Céline Henry, Agnès Fouet, Karine Gloux, Jamila Anba-Mondoloni, Alexandra Gruss. Oxidative Stress is Intrinsic to Staphylococcal Adaptation to Fatty Acid Synthesis Antibiotics. 2022https://doi.org/10.1101/2022.09.09.506511
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 53 other publications.
- 1Konychev, A.; Heep, M.; Moritz, R. K.; Kreuter, A.; Shulutko, A.; Fierlbeck, G.; Bouylout, K.; Pathan, R.; Trostmann, U.; Chaves, R. L. Safety and efficacy of daptomycin as first-line treatment for complicated skin and soft tissue infections in elderly patients: an open-label, multicentre, randomized phase IIIb trial. Drugs Aging 2013, 30 (10), 829– 836, DOI: 10.1007/s40266-013-0114-81https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVWmt7bE&md5=51940b8a92bebe81aef5707dce85bf5dSafety and Efficacy of Daptomycin as First-Line Treatment for Complicated Skin and Soft Tissue Infections in Elderly Patients: An Open-Label, Multicentre, Randomized Phase IIIb TrialKonychev, Alexander; Heep, Markus; Moritz, Rose K. C.; Kreuter, Alexander; Shulutko, Alexander; Fierlbeck, Gerhard; Bouylout, Kamel; Pathan, Rashidkhan; Trostmann, Uwe; Chaves, Ricardo L.Drugs & Aging (2013), 30 (10), 829-836CODEN: DRAGE6; ISSN:1170-229X. (Springer International Publishing AG)Background: Daptomycin has proven efficacy in patients with Gram-pos. complicated skin and soft tissue infections (cSSTIs), including those caused by Staphylococcus aureus, regardless of methicillin susceptibility. Objective: This study was undertaken to evaluate the efficacy and safety of daptomycin in elderly patients. Study Design: This was an open-label, multicentre, randomized phase IIIb study conducted in hospitalized patients Patients Patients aged ≥65 years with a diagnosis of Gram-pos. cSSTIs with or without bacteremia were included. In addn., infections were required to be of sufficient severity to require inpatient hospitalization and treatment with parenteral antibiotics for at least 96 h. The main exclusion criterion was the presence of a non-complicated SSTI that could heal by itself or be cured by surgical removal of the site of infection. Intervention: Patients were randomized 2:1 to i.v. daptomycin or pooled i.v. std. therapies semi-synthetic penicillin or vancomycin, referred to as the comparator. Duration of treatment was between 5 and 14 days for cSSTIs without bacteremia and between 10 and 28 days for cSSTIs with bacteremia. Main Outcome Measure: The primary objective was descriptive comparison of clin. success in clin. evaluable patients at test of cure, 7-14 days post treatment. Secondary objectives were microbiol. outcome, duration of treatment and safety. Results: In total, 120 patients were randomized (81 to daptomycin; 39 to the comparator) and 102 patients completed the study. Baseline characteristics were similar between the two groups. Common infections included cellulitis, ulcers and abscesses; six patients had bacteremia [five documented (daptomycin, n = 3; comparator, n = 2); and one suspected (daptomycin, n = 1)]. Test-of-cure clin. success rates were numerically higher for daptomycin than for the comparator [89.0 % (65/73) vs. 83.3 % (25/30); odds ratio 1.65 (95 % confidence interval 0.49-5.54)]. For patients with S. aureus infections, cure rates were 89.7 % (35/39) vs. 69.2 % (9/13), resp.; percentage points difference, 20.5 (95 % confidence interval -12.2 to 50.9). Rates of adverse events (AEs) and serious AEs were similar in both treatment arms; however, discontinuation rates for AEs/serious AEs were lower for daptomycin than for the comparator (3.8 % vs. 10.0 %). Three serious AEs were considered to be related to the study drug: one case each of pancytopenia (semi-synthetic penicillin), renal failure (vancomycin) and asymptomatic increase in creatine phosphokinase concns. (daptomycin). Conclusion: In elderly patients, for whom data were previously limited, the efficacy and safety of daptomycin have been confirmed, including for infections caused by S. aureus, regardless of methicillin susceptibility.
- 2Moran, G. J.; Fang, E.; Corey, G. R.; Das, A. F.; De Anda, C.; Prokocimer, P. Tedizolid for 6 days versus linezolid for 10 days for acute bacterial skin and skin-structure infections (ESTABLISH-2): a randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect Dis 2014, 14 (8), 696– 705, DOI: 10.1016/S1473-3099(14)70737-62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhtVWrs7bK&md5=70ead6ed25db678eeec65a6d7e00f151Tedizolid for 6 days versus linezolid for 10 days for acute bacterial skin and skin-structure infections (ESTABLISH-2): a randomised, double-blind, phase 3, non-inferiority trialMoran, Gregory J.; Fang, Edward; Corey, G. Ralph; Das, Anita F.; De Anda, Carisa; Prokocimer, PhilippeLancet Infectious Diseases (2014), 14 (8), 696-705CODEN: LIDABP; ISSN:1473-3099. (Elsevier Ltd.)New antibiotics are needed to treat infections caused by drug-resistant bacteria. Tedizolid is a novel oxazolidinone antibacterial drug designed to provide enhanced activity against Gram-pos. pathogens. We aimed to assess the efficacy and safety of i.v. to oral tedizolid for treatment of patients with acute bacterial skin and skin-structure infections. ESTABLISH-2 was a randomised, double-blind, phase 3, non-inferiority trial done between Sept 28, 2011, and Jan 10, 2013, at 58 centers in nine countries. Patients (aged ≥12 years) with acute bacterial skin and skin-structure infections (cellulitis or erysipelas, major cutaneous abscess, or wound infection) that had a min. lesion area of 75 cm2 and were suspected or documented to be assocd. with a Gram-pos. pathogen, were randomly assigned (1:1), via an interactive voice-response system with block randomisation, to receive i.v. once-daily tedizolid (200 mg for 6 days) or twice-daily linezolid (600 mg for 10 days), with optional oral step-down. Randomisation was stratified by geog. region and type of acute bacterial skin and skin-structure infection. The primary endpoint was early clin. response (≥20% redn. in lesion area at 48-72 h compared with baseline), with a non-inferiority margin of -10%. Anal. was by intention to treat. This study is registered with ClinicalTrials.gov, no. NCT01421511.666 patients were randomly assigned to receive tedizolid (n=332) or linezolid (n=334). 283 (85%) patients in the tedizolid group and 276 (83%) in the linezolid group achieved early clin. response (difference 2·6%, 95% CI -3·0 to 8·2), meeting the prespecified non-inferiority margin. Gastrointestinal adverse events were less frequent with tedizolid than linezolid, taking place in 52 (16%) of 331 patients and 67 (20%) of 327 patients in the safety population. Treatment-emergent adverse events leading to discontinuation of study drug were reported by one (<1%) patient in the tedizolid group and four (1%) patients in the linezolid group. I.v. to oral once-daily tedizolid 200 mg for 6 days was non-inferior to twice-daily linezolid 600 mg for 10 days for treatment of patients with acute bacterial skin and skin-structure infections. Tedizolid could become a useful option for the treatment of acute bacterial skin and skin-structure infections in the hospital and outpatient settings.
- 3File, T. M.; Goldberg, L.; Das, A.; Sweeney, C.; Saviski, J.; Gelone, S. P.; Seltzer, E.; Paukner, S.; Wicha, W. W.; Talbot, G. H.; Gasink, L. B. Efficacy and Safety of Intravenous-to-oral Lefamulin, a Pleuromutilin Antibiotic, for the Treatment of Community-acquired Bacterial Pneumonia: The Phase III Lefamulin Evaluation Against Pneumonia (LEAP 1) Trial. Clin Infect Dis 2019, 69 (11), 1856– 1867, DOI: 10.1093/cid/ciz0903https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXnsVKlurY%253D&md5=8f51288366ed79a4a793c07e66076187Efficacy and safety of intravenous-to-oral lefamulin, a pleuromutilin antibiotic, for the treatment of community-acquired bacterial pneumonia: the phase III lefamulin evaluation against pneumonia (LEAP 1) trialFile, Thomas M., Jr.; Goldberg, Lisa; Das, Anita; Sweeney, Carolyn; Saviski, John; Gelone, Steven P.; Seltzer, Elyse; Paukner, Susanne; Wicha, Wolfgang W.; Talbot, George H.; Gasink, Leanne B.Clinical Infectious Diseases (2019), 69 (11), 1856-1867CODEN: CIDIEL; ISSN:1537-6591. (Oxford University Press)Lefamulin, a pleuromutilin antibiotic, is active against pathogens commonly causing community-acquired bacterial pneumonia (CABP). The Lefamulin Evaluation Against Pneumonia (LEAP 1) study was a global noninferiority trial to evaluate the efficacy and safety of lefamulin for the treatment of CABP. In this double-blind study, adults with CABP of Pneumonia Outcomes Research Team risk class ≥III were randomized 1:1 to receive lefamulin at 150 mg i.v. (IV) every 12 h or moxifloxacin at 400 mg IV every 24 h. After 6 doses, patients could be switched to an oral study drug if prespecified improvement criteria were met. If methicillin-resistant Staphylococcus aureus was suspected, either linezolid or placebo was added to moxifloxacin or lefamulin, resp. The US Food and Drug Administration primary endpoint was an early clin. response (ECR) 96 ± 24 h after the first dose of the study drug in the intent-to-treat (ITT) population (noninferiority margin, 12.5%). The European Medicines Agency co-primary endpoints were an investigator assessment of clin. response (IACR) 5-10 days after the last dose of the study drug in the modified ITT (mITT) and clin. evaluable (CE) populations (noninferiority margin, 10%). There were 551 patients randomized (n = 276 lefamulin; n = 275 moxifloxacin). Lefamulin was noninferior to moxifloxacin for ECR (87.3% vs 90.2%, resp.; difference -2.9%, 95% confidence interval [CI] g -8.5 to 2.8) and IACR (mITT, 81.7% vs 84.2%, resp.; difference -2.6%, 95% CI -8.9 to 3.9; CE, 86.9% vs 89.4%, resp.; difference -2.5%, 95% CI -8.4 to 3.4). Rates of study drug discontinuation due to treatment-emergent adverse events were 2.9% for lefamulin and 4.4% for moxifloxacin. Lefamulin was noninferior to moxifloxacin for the primary efficacy endpoints and was generally safe and well tolerated.
- 4WHO Antibacterial products in clinical development for priority pathogens. https://www.who.int/observatories/global-observatory-on-health-research-and-development/monitoring/antibacterial-products-in-clinical-development-for-priority-pathogens (accessed June 13, 2022).There is no corresponding record for this reference.
- 5Rice, L. B. Federal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPE. J. Infect Dis 2008, 197 (8), 1079– 81, DOI: 10.1086/5334525https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1c3lvFChug%253D%253D&md5=c2c5ec8ad941f702f588f89741d9410eFederal funding for the study of antimicrobial resistance in nosocomial pathogens: no ESKAPERice Louis BThe Journal of infectious diseases (2008), 197 (8), 1079-81 ISSN:0022-1899.There is no expanded citation for this reference.
- 6Boucher, H. W.; Talbot, G. H.; Bradley, J. S.; Edwards, J. E.; Gilbert, D.; Rice, L. B.; Scheld, M.; Spellberg, B.; Bartlett, J. Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis 2009, 48 (1), 1– 12, DOI: 10.1086/5950116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1M%252Fit1Krsg%253D%253D&md5=a3f71406b891f90f4f0f77b5f1ebc4f8Bad bugs, no drugs: no ESKAPE! An update from the Infectious Diseases Society of AmericaBoucher Helen W; Talbot George H; Bradley John S; Edwards John E; Gilbert David; Rice Louis B; Scheld Michael; Spellberg Brad; Bartlett JohnClinical infectious diseases : an official publication of the Infectious Diseases Society of America (2009), 48 (1), 1-12 ISSN:.The Infectious Diseases Society of America (IDSA) continues to view with concern the lean pipeline for novel therapeutics to treat drug-resistant infections, especially those caused by gram-negative pathogens. Infections now occur that are resistant to all current antibacterial options. Although the IDSA is encouraged by the prospect of success for some agents currently in preclinical development, there is an urgent, immediate need for new agents with activity against these panresistant organisms. There is no evidence that this need will be met in the foreseeable future. Furthermore, we remain concerned that the infrastructure for discovering and developing new antibacterials continues to stagnate, thereby risking the future pipeline of antibacterial drugs. The IDSA proposed solutions in its 2004 policy report, "Bad Bugs, No Drugs: As Antibiotic R&D Stagnates, a Public Health Crisis Brews," and recently issued a "Call to Action" to provide an update on the scope of the problem and the proposed solutions. A primary objective of these periodic reports is to encourage a community and legislative response to establish greater financial parity between the antimicrobial development and the development of other drugs. Although recent actions of the Food and Drug Administration and the 110th US Congress present a glimmer of hope, significant uncertainly remains. Now, more than ever, it is essential to create a robust and sustainable antibacterial research and development infrastructure--one that can respond to current antibacterial resistance now and anticipate evolving resistance. This challenge requires that industry, academia, the National Institutes of Health, the Food and Drug Administration, the Centers for Disease Control and Prevention, the US Department of Defense, and the new Biomedical Advanced Research and Development Authority at the Department of Health and Human Services work productively together. This report provides an update on potentially effective antibacterial drugs in the late-stage development pipeline, in the hope of encouraging such collaborative action.
- 7Cassini, A.; Hogberg, L. D.; Plachouras, D.; Quattrocchi, A.; Hoxha, A.; Simonsen, G. S.; Colomb-Cotinat, M.; Kretzschmar, M. E.; Devleesschauwer, B.; Cecchini, M.; Ouakrim, D. A.; Oliveira, T. C.; Struelens, M. J.; Suetens, C.; Monnet, D. L.; the Burden of AMR Collective Group Attributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Economic Area in 2015: a population-level modelling analysis. Lancet Infect. Dis. 2019, 19 (1), 56– 66, DOI: 10.1016/S1473-3099(18)30605-47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cvotFGluw%253D%253D&md5=151619810730b07e3d84692f1d966b9aAttributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the European Economic Area in 2015: a population-level modelling analysisCassini Alessandro; Hogberg Liselotte Diaz; Plachouras Diamantis; Quattrocchi Annalisa; Hoxha Ana; Struelens Marc J; Suetens Carl; Monnet Dominique L; Simonsen Gunnar Skov; Colomb-Cotinat Melanie; Kretzschmar Mirjam E; Devleesschauwer Brecht; Cecchini Michele; Ouakrim Driss Ait; Oliveira Tiago CravoThe Lancet. Infectious diseases (2019), 19 (1), 56-66 ISSN:.BACKGROUND: Infections due to antibiotic-resistant bacteria are threatening modern health care. However, estimating their incidence, complications, and attributable mortality is challenging. We aimed to estimate the burden of infections caused by antibiotic-resistant bacteria of public health concern in countries of the EU and European Economic Area (EEA) in 2015, measured in number of cases, attributable deaths, and disability-adjusted life-years (DALYs). METHODS: We estimated the incidence of infections with 16 antibiotic resistance-bacterium combinations from European Antimicrobial Resistance Surveillance Network (EARS-Net) 2015 data that was country-corrected for population coverage. We multiplied the number of bloodstream infections (BSIs) by a conversion factor derived from the European Centre for Disease Prevention and Control point prevalence survey of health-care-associated infections in European acute care hospitals in 2011-12 to estimate the number of non-BSIs. We developed disease outcome models for five types of infection on the basis of systematic reviews of the literature. FINDINGS: From EARS-Net data collected between Jan 1, 2015, and Dec 31, 2015, we estimated 671 689 (95% uncertainty interval [UI] 583 148-763 966) infections with antibiotic-resistant bacteria, of which 63·5% (426 277 of 671 689) were associated with health care. These infections accounted for an estimated 33 110 (28 480-38 430) attributable deaths and 874 541 (768 837-989 068) DALYs. The burden for the EU and EEA was highest in infants (aged <1 year) and people aged 65 years or older, had increased since 2007, and was highest in Italy and Greece. INTERPRETATION: Our results present the health burden of five types of infection with antibiotic-resistant bacteria expressed, for the first time, in DALYs. The estimated burden of infections with antibiotic-resistant bacteria in the EU and EEA is substantial compared with that of other infectious diseases, and has increased since 2007. Our burden estimates provide useful information for public health decision-makers prioritising interventions for infectious diseases. FUNDING: European Centre for Disease Prevention and Control.
- 8Antimicrobial Resistance, C. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet 2022, 399 (10325), 629– 655, DOI: 10.1016/S0140-6736(21)02724-0There is no corresponding record for this reference.
- 9Richter, M. F.; Drown, B. S.; Riley, A. P.; Garcia, A.; Shirai, T.; Svec, R. L.; Hergenrother, P. J. Predictive compound accumulation rules yield a broad-spectrum antibiotic. Nature 2017, 545 (7654), 299– 304, DOI: 10.1038/nature223089https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXnsVeis7Y%253D&md5=ae869cc99ef0e6805b96b6595a222cdcPredictive compound accumulation rules yield a broad-spectrum antibioticRichter, Michelle F.; Drown, Bryon S.; Riley, Andrew P.; Garcia, Alfredo; Shirai, Tomohiro; Svec, Riley L.; Hergenrother, Paul J.Nature (London, United Kingdom) (2017), 545 (7654), 299-304CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Most small mols. are unable to rapidly traverse the outer membrane of Gram-neg. bacteria and accumulate inside these cells, making the discovery of much-needed drugs against these pathogens challenging. Current understanding of the physicochem. properties that dictate small-mol. accumulation in Gram-neg. bacteria is largely based on retrospective analyses of antibacterial agents, which suggest that polarity and mol. wt. are key factors. Here the authors assess the ability of over 180 diverse compds. to accumulate in Escherichia coli. Computational anal. of the results reveals major differences from the retrospective studies, namely that the small mols. that are most likely to accumulate contain an amine, are amphiphilic and rigid, and have low globularity. These guidelines were then applied to convert deoxynybomycin, a natural product that is active only against Gram-pos. organisms, into an antibiotic with activity against a diverse panel of multi-drug-resistant Gram-neg. pathogens. The authors anticipate that these findings will aid in the discovery and development of antibiotics against Gram-neg. bacteria.
- 10Zhao, S.; Adamiak, J. W.; Bonifay, V.; Mehla, J.; Zgurskaya, H. I.; Tan, D. S. Defining new chemical space for drug penetration into Gram-negative bacteria. Nat. Chem. Biol. 2020, 16 (12), 1293– 1302, DOI: 10.1038/s41589-020-00674-610https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitlGjsLfM&md5=43f7ee77d0e360e4666c569eb5ef728fDefining new chemical space for drug penetration into Gram-negative bacteriaZhao, Shibin; Adamiak, Justyna W.; Bonifay, Vincent; Mehla, Jitender; Zgurskaya, Helen I.; Tan, Derek S.Nature Chemical Biology (2020), 16 (12), 1293-1302CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Abstr.: We live in the era of antibiotic resistance, and this problem will progressively worsen if no new solns. emerge. In particular, Gram-neg. pathogens present both biol. and chem. challenges that hinder the discovery of new antibacterial drugs. First, these bacteria are protected from a variety of structurally diverse drugs by a low-permeability barrier composed of two membranes with distinct permeability properties, in addn. to active drug efflux, making this cell envelope impermeable to most compds. Second, chem. libraries currently used in drug discovery contain few compds. that can penetrate Gram-neg. bacteria. As a result of these challenges, intensive screening campaigns have led to few successes, highlighting the need for new approaches to identify regions of chem. space that are specifically relevant to antibacterial drug discovery. Herein we provide an overview of emerging insights into this problem and outline a general approach to addressing it using prospective anal. of chem. libraries for the ability of compds. to accumulate in Gram-neg. bacteria. The overall goal is to develop robust cheminformatic tools to predict Gram-neg. permeation and efflux, which can then be used to guide medicinal chem. campaigns and the design of antibacterial discovery libraries.
- 11Munoz, K. A.; Hergenrother, P. J. Facilitating Compound Entry as a Means to Discover Antibiotics for Gram-Negative Bacteria. Acc. Chem. Res. 2021, 54 (6), 1322– 1333, DOI: 10.1021/acs.accounts.0c0089511https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXkvFyisb8%253D&md5=bd3fe94a1db2612554c9d3557ee7205eFacilitating Compound Entry as a Means to Discover Antibiotics for Gram-Negative BacteriaMunoz, Kristen A.; Hergenrother, Paul J.Accounts of Chemical Research (2021), 54 (6), 1322-1333CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. It has been over half a century since the last class of antibiotics active against the most problematic Gram-neg. bacteria was approved by the Food and Drug Administration (FDA). The major challenge with developing antibiotics to treat these infections is not drug-target engagement but rather the inability of most small mols. to traverse the Gram-neg. membranes, be retained, and accumulate within the cell. Despite an abundance of lead compds., limited understanding of the physicochem. properties needed for compd. accumulation (or avoidance of efflux) in Gram-neg. bacteria has precluded a generalizable approach for developing Gram-neg. antibiotics. Indeed, in many instances, despite years of intensive derivatization efforts and the synthesis of hundreds of compds. aimed at building in Gram-neg. activity, little or no progress has been made in expanding the spectrum of activity for many Gram-pos.-only antibiotics. We describe the discovery and successful applications of a promising strategy for enhancing the accumulation of Gram-pos.-only antibiotics as a means of imbuing compds. with broad-spectrum activity. Utilizing a prospective approach examg. the accumulation in Escherichia coli for >180 diverse compds., we found that small mols. have an increased likelihood to accumulate in E. coli when they contain an ionizable nitrogen, have low 3-dimensionality, and are rigid. Implementing these guidelines, codified as the eNTRy rules and assisted by web application www.entry-way.org, we have facilitated compd. entry and systematically built Gram-neg. activity into Gram-pos.-only antibiotics. Though each antibiotic will have case-specific considerations, we describe a set of important criteria to consider when selecting candidate Gram-pos.-only antibiotics for conversion to Gram-neg.-active versions via the eNTRy rules. As detailed herein, using this blueprint the spectrum of activity was expanded for 3 antibiotic classes that engage 3 different biol. targets: DNA gyrase inhibitor 6DNM, FabI inhibitor Debio-1452, and FMN riboswitch inhibitor Ribocil C. In each scenario, the eNTRy rules guided the synthesis of key analogs predisposed to accumulate in Gram-neg. bacteria leading to compds. that display antibiotic activity (min. inhibitory concns. (MIC) ≤8μg/mL) against E. coli and other Gram-neg. ESKAPE pathogens. While the eNTRy rules will continue to be refined and enhanced as more accumulation data is gathered, on the basis of these collective results and on other examples not covered herein it is clear that the eNTRy rules are actionable for the development of novel broad-spectrum antibiotics from Gram-pos.-only compds. By enabling the prediction of compd. accumulation, the eNTRy rules should facilitate the process of discovering and developing novel antibiotics active against Gram-neg. bacteria.
- 12Payne, D. J.; Gwynn, M. N.; Holmes, D. J.; Pompliano, D. L. Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat. Rev. Drug Discov 2007, 6 (1), 29– 40, DOI: 10.1038/nrd220112https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtlGktbfM&md5=18100253f9868af578ea621ab98a5c86Drugs for bad bugs: confronting the challenges of antibacterial discoveryPayne, David J.; Gwynn, Michael N.; Holmes, David J.; Pompliano, David L.Nature Reviews Drug Discovery (2007), 6 (1), 29-40CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. The sequencing of the first complete bacterial genome in 1995 heralded a new era of hope for antibacterial drug discoverers, who now had the tools to search entire genomes for new antibacterial targets. Several companies, including GlaxoSmithKline, moved back into the antibacterials area and embraced a genomics-derived, target-based approach to screen for new classes of drugs with novel modes of action. Here, we share our experience of evaluating more than 300 genes and 70 high-throughput screening campaigns over a period of 7 years, and look at what we learned and how that has influenced GlaxoSmithKline's antibacterials strategy going forward.
- 13Tommasi, R.; Brown, D. G.; Walkup, G. K.; Manchester, J. I.; Miller, A. A. ESKAPEing the labyrinth of antibacterial discovery. Nat. Rev. Drug Discov 2015, 14 (8), 529– 42, DOI: 10.1038/nrd457213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFyks77M&md5=0d3159250e27926ab280c305ea5fb9efESKAPEing the labyrinth of antibacterial discoveryTommasi, Ruben; Brown, Dean G.; Walkup, Grant K.; Manchester, John I.; Miller, Alita A.Nature Reviews Drug Discovery (2015), 14 (8), 529-542CODEN: NRDDAG; ISSN:1474-1776. (Nature Publishing Group)A review. Antimicrobial drug resistance is a growing threat to global public health. Multidrug resistance among the 'ESKAPE' organisms - encompassing Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp. - is of particular concern because they are responsible for many serious infections in hospitals. Although some promising agents are in the pipeline, there is an urgent need for new antibiotic scaffolds. However, antibacterial researchers have struggled to identify new small mols. with meaningful cellular activity, esp. those effective against multidrug-resistant Gram-neg. pathogens. This difficulty ultimately stems from an incomplete understanding of efflux systems and compd. permeation through bacterial membranes. This Opinion article describes findings from target-based and phenotypic screening efforts carried out at AstraZeneca over the past decade, discusses some of the subsequent chem. challenges and concludes with a description of new approaches comprising a combination of computational modeling and advanced biol. tools which may pave the way towards the discovery of new antibacterial agents.
- 14Yao, J.; Rock, C. O. Resistance Mechanisms and the Future of Bacterial Enoyl-Acyl Carrier Protein Reductase (FabI) Antibiotics. Cold Spring Harb Perspect Med. 2016, 6 (3), a027045, DOI: 10.1101/cshperspect.a027045There is no corresponding record for this reference.
- 15Seefeld, M. A.; Miller, W. H.; Newlander, K. A.; Burgess, W. J.; Payne, D. J.; Rittenhouse, S. F.; Moore, T. D.; DeWolf, W. E., Jr.; Keller, P. M.; Qiu, X.; Janson, C. A.; Vaidya, K.; Fosberry, A. P.; Smyth, M. G.; Jaworski, D. D.; Slater-Radosti, C.; Huffman, W. F. Inhibitors of bacterial enoyl acyl carrier protein reductase (FabI): 2,9-disubstituted 1,2,3,4-tetrahydropyrido[3,4-b]indoles as potential antibacterial agents. Bioorg. Med. Chem. Lett. 2001, 11 (17), 2241– 4, DOI: 10.1016/S0960-894X(01)00405-X15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXmt1yqtbY%253D&md5=6904b75740db275d6defbbc08009a6ebInhibitors of bacterial enoyl acyl carrier protein reductase (FabI): 2,9-disubstituted 1,2,3,4-tetrahydropyrido[3,4-b]indoles as potential antibacterial agentsSeefeld, M. A.; Miller, W. H.; Newlander, K. A.; Burgess, W. J.; Payne, D. J.; Rittenhouse, S. F.; Moore, T. D.; DeWolf, W. E.; Keller, P. M.; Qiu, X.; Janson, C. A.; Vaidya, K.; Fosberry, A. P.; Smyth, M. G.; Jaworski, D. D.; Slater-Radosti, C.; Huffman, W. F.Bioorganic & Medicinal Chemistry Letters (2001), 11 (17), 2241-2244CODEN: BMCLE8; ISSN:0960-894X. (Elsevier Science Ltd.)An SAR study of a screening lead has led to the identification of 2,9-disubstituted 1,2,3,4-tetrahydropyrido[3,4-b]indoles as inhibitors of Staphylococcus aureus enoyl acyl carrier protein reductase (FabI).
- 16Payne, D. J.; Miller, W. H.; Berry, V.; Brosky, J.; Burgess, W. J.; Chen, E.; DeWolf Jr, W. E., Jr.; Fosberry, A. P.; Greenwood, R.; Head, M. S.; Heerding, D. A.; Janson, C. A.; Jaworski, D. D.; Keller, P. M.; Manley, P. J.; Moore, T. D.; Newlander, K. A.; Pearson, S.; Polizzi, B. J.; Qiu, X.; Rittenhouse, S. F.; Slater-Radosti, C.; Salyers, K. L.; Seefeld, M. A.; Smyth, M. G.; Takata, D. T.; Uzinskas, I. N.; Vaidya, K.; Wallis, N. G.; Winram, S. B.; Yuan, C. C.; Huffman, W. F. Discovery of a novel and potent class of FabI-directed antibacterial agents. Antimicrob. Agents Chemother. 2002, 46 (10), 3118– 24, DOI: 10.1128/AAC.46.10.3118-3124.200216https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XntlOgtbk%253D&md5=b56fe92bf2328df5aacd97fe89f44226Discovery of a novel and potent class of FabI-directed antibacterial agentsPayne, David J.; Miller, William H.; Berry, Valerie; Brosky, John; Burgess, Walter J.; Chen, Emile; DeWolf, Walter E., Jr.; Fosberry, Andrew P.; Greenwood, Rebecca; Head, Martha S.; Heerding, Dirk A.; Janson, Cheryl A.; Jaworski, Deborah D.; Keller, Paul M.; Manley, Peter J.; Moore, Terrance D.; Newlander, Kenneth A.; Pearson, Stewart; Polizzi, Brian J.; Qiu, Xiayang; Rittenhouse, Stephen F.; Slater-Radosti, Courtney; Salyers, Kevin L.; Seefeld, Mark A.; Smyth, Martin G.; Takata, Dennis T.; Uzinskas, Irene N.; Vaidya, Kalindi; Wallis, Nicola G.; Winram, Scott B.; Yuan, Catherine C. K.; Huffman, William F.Antimicrobial Agents and Chemotherapy (2002), 46 (10), 3118-3124CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Bacterial enoyl-acyl carrier protein (ACP) reductase (FabI) catalyzes the final step in each elongation cycle of bacterial fatty acid biosynthesis and is an attractive target for the development of new antibacterial agents. High-throughput screening of the Staphylococcus aureus FabI enzyme identified a novel, weak inhibitor with no detectable antibacterial activity against S. aureus. Iterative medicinal chem. and x-ray crystal structure-based design led to the identification of compd. 4 [(E)-N-methyl-N-(2-methyl-1H-indol-3-ylmethyl)-3-(7-oxo-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)acrylamide], which is 350-fold more potent than the original lead compd. obtained by high-throughput screening in the FabI inhibition assay. Compd. 4 has exquisite antistaphylococci activity, achieving MICs at which 90% of isolates are inhibited more than 500 times lower than those of nine currently available antibiotics against a panel of multidrug-resistant strains of S. aureus and Staphylococcus epidermidis. Furthermore, compd. 4 exhibits excellent in vivo efficacy in an S. aureus infection model in rats. Biochem. and genetic approaches have confirmed that the mode of antibacterial action of compd. 4 and related compd. is via inhibition of FabI. Compd. 4 also exhibits weak FabK inhibitory activity, which may explain its antibacterial activity against Streptococcus pneumoniae and Enterococcus faecalis, which depend on FabK and both FabK and FabI, resp., for their enoyl-ACP reductase function. These results show that compd. 4 is representative of a new, totally synthetic series of antibacterial agents that has the potential to provide novel alternatives for the treatment of S. aureus infections that are resistant to our present armory of antibiotics.
- 17Wittke, F.; Vincent, C.; Chen, J.; Heller, B.; Kabler, H.; Overcash, J. S.; Leylavergne, F.; Dieppois, G. Afabicin, a First-in-Class Antistaphylococcal Antibiotic, in the Treatment of Acute Bacterial Skin and Skin Structure Infections: Clinical Noninferiority to Vancomycin/Linezolid. Antimicrob. Agents Chemother. 2020, 64 (10), e00250-20, DOI: 10.1128/AAC.00250-20There is no corresponding record for this reference.
- 18Karlowsky, J. A.; Kaplan, N.; Hafkin, B.; Hoban, D. J.; Zhanel, G. G. AFN-1252, a FabI inhibitor, demonstrates a Staphylococcus-specific spectrum of activity. Antimicrob. Agents Chemother. 2009, 53 (8), 3544– 8, DOI: 10.1128/AAC.00400-0918https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXps1ygsL0%253D&md5=48086b24a4c38d14373320c89c91f1bfAFN-1252, a FabI inhibitor, demonstrates a staphylococcus-specific spectrum of activityKarlowsky, James A.; Kaplan, Nachum; Hafkin, Barry; Hoban, Daryl J.; Zhanel, George G.Antimicrobial Agents and Chemotherapy (2009), 53 (8), 3544-3548CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)AFN-1252, a potent inhibitor of enoyl-acyl carrier protein reductase (FabI), inhibited all clin. isolates of Staphylococcus aureus (n = 502) and Staphylococcus epidermidis (n = 51) tested, including methicillin (methicillin)-resistant isolates, at concns. of ≤0.12 μg/mL. In contrast, AFN-1252 was inactive (MIC90, >4 μg/mL) against clin. isolates of Streptococcus pneumoniae, beta-hemolytic streptococci, Enterococcus spp., Enterobacteriaceae, nonfermentative gram-neg. bacilli, and Moraxella catarrhalis. These data support the continued development of AFN-1252 for the treatment of patients with resistant staphylococcal infections.
- 19Parker, E. N.; Drown, B. S.; Geddes, E. J.; Lee, H. Y.; Ismail, N.; Lau, G. W.; Hergenrother, P. J. Implementation of permeation rules leads to a FabI inhibitor with activity against Gram-negative pathogens. Nat. Microbiol 2020, 5 (1), 67– 75, DOI: 10.1038/s41564-019-0604-519https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitFOru7jM&md5=39c84cdc028ef0c5568f2cecd60cd209Implementation of permeation rules leads to a FabI inhibitor with activity against Gram-negative pathogensParker, Erica N.; Drown, Bryon S.; Geddes, Emily J.; Lee, Hyang Yeon; Ismail, Nahed; Lau, Gee W.; Hergenrother, Paul J.Nature Microbiology (2020), 5 (1), 67-75CODEN: NMAICH; ISSN:2058-5276. (Nature Research)Gram-neg. bacterial infections are a significant public health concern, and the lack of new drug classes for these pathogens is linked to the inability of most drug leads to accumulate inside Gram-neg. bacteria1-7. Here, we report the development of a web application-eNTRyway-that predicts compd. accumulation (in Escherichia coli) from its structure. In conjunction with structure-activity relationships and X-ray data, eNTRyway was utilized to re-design Debio-1452-a Gram-pos.-only antibiotic8-into versions that accumulate in E. coli and possess antibacterial activity against high-priority Gram-neg. pathogens. The lead compd. Debio-1452-NH3 operates as an antibiotic via the same mechanism as Debio-1452, namely potent inhibition of the enoyl-acyl carrier protein reductase FabI, as validated by in vitro enzyme assays and the generation of bacterial isolates with spontaneous target mutations. Debio-1452-NH3 is well tolerated in vivo, reduces bacterial burden in mice and rescues mice from lethal infections with clin. isolates of Acinetobacter baumannii, Klebsiella pneumoniae and E. coli. This work provides tools for the facile discovery and development of high-accumulating compds. in E. coli, and a general blueprint for the conversion of Gram-pos.-only compds. into broad-spectrum antibiotics.
- 20Richter, M. F.; Hergenrother, P. J. The challenge of converting Gram-positive-only compounds into broad-spectrum antibiotics. Ann. N.Y. Acad. Sci. 2019, 1435 (1), 18– 38, DOI: 10.1111/nyas.1359820https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlyktw%253D%253D&md5=12fffe0d10e7d3e5c134ac28ce929ab3The challenge of converting Gram-positive-only compounds into broad-spectrum antibioticsRichter, Michelle F.; Hergenrother, Paul J.Annals of the New York Academy of Sciences (2019), 1435 (1, Antimicrobial Therapeutics Reviews), 18-38CODEN: ANYAA9; ISSN:0077-8923. (John Wiley & Sons, Inc.)A review. Multidrug resistant Gram-neg. bacterial infections are on the rise, and there is a lack of new classes of drugs to treat these pathogens. This drug shortage is largely due to the challenge of finding antibiotics that can permeate and persist inside Gram-neg. species. Efforts to understand the mol. properties that enable certain compds. to accumulate in Gram-neg. bacteria based on retrospective studies of known antibiotics have not been generally actionable in the development of new antibiotics. A recent assessment of the ability of >180 diverse small mols. to accumulate in Escherichia coli led to predictive guidelines for compd. accumulation in E. coli. These "eNTRy rules" state that compds. are most likely to accumulate if they contain a nonsterically encumbered ionizable Nitrogen (primary amines are the best), have low Three-dimensionality (globularity ≤ 0.25), and are relatively Rigid (rotatable bonds ≤ 5). In this review, we look back through 50+ years of antibacterial research and 1000s of derivs. and assess this historical data set through the lens of these predictive guidelines. The results are consistent with the eNTRy rules, suggesting that the eNTRy rules may provide an actionable and general roadmap for the conversion of Gram-pos.-only compds. into broad-spectrum antibiotics.
- 21Kaplan, N.; Albert, M.; Awrey, D.; Bardouniotis, E.; Berman, J.; Clarke, T.; Dorsey, M.; Hafkin, B.; Ramnauth, J.; Romanov, V.; Schmid, M. B.; Thalakada, R.; Yethon, J.; Pauls, H. W. Mode of action, in vitro activity, and in vivo efficacy of AFN-1252, a selective antistaphylococcal FabI inhibitor. Antimicrob. Agents Chemother. 2012, 56 (11), 5865– 5874, DOI: 10.1128/AAC.01411-1221https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhsF2iurfK&md5=67bc12e665ce45b808d799db4fb09c92Mode of action, in vitro activity, and in vivo efficacy of AFN-1252, a selective antistaphylococcal FabI inhibitorKaplan, Nachum; Albert, Monique; Awrey, Donald; Bardouniotis, Elias; Berman, Judd; Clarke, Teresa; Dorsey, Mandy; Hafkin, Barry; Ramnauth, Jaillal; Romanov, Vladimir; Schmid, Molly B.; Thalakada, Rosanne; Yethon, Jeremy; Pauls, Henry W.Antimicrobial Agents and Chemotherapy (2012), 56 (11), 5865-5874CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)The mechanism of action of AFN-1252, a selective inhibitor of Staphylococcus aureus enoyl-acyl carrier protein reductase (FabI), which is involved in fatty acid biosynthesis, was confirmed by using biochem., macromol. synthesis, genetics, and co-crystn. of an AFN-1252-FabI complex. AFN-1252 demonstrated a low propensity for spontaneous resistance development and a time-dependent redn. of the viability of both methicillin-susceptible and methicillin-resistant S. aureus, achieving a ≥2-log10 redn. in S. aureus counts over 24 h, and was extremely potent against clin. isolates of S. aureus (MIC90, 0.015 μg/mL) and coagulase-neg. staphylococci (MIC90, 0.12 μg/mL), regardless of their drug resistance, hospital- or community-assocd. origin, or other clin. subgroup. AFN-1252 was orally available in mouse pharmacokinetic studies, and a single oral dose of 1 mg/kg AFN-1252 was efficacious in a mouse model of septicemia, providing 100% protection from an otherwise lethal peritoneal infection of S. aureus Smith. A median ED of 0.15 mg/kg indicated that AFN-1252 was 12 to 24 times more potent than linezolid in the model. These studies, demonstrating a selective mode of action, potent in vitro activity, and in vivo efficacy, support the continued investigation of AFN-1252 as a targeted therapeutic for staphylococcal infections.
- 22Seefeld, M. A.; Miller, W. H.; Newlander, K. A.; Burgess, W. J.; DeWolf, W. E., Jr.; Elkins, P. A.; Head, M. S.; Jakas, D. R.; Janson, C. A.; Keller, P. M.; Manley, P. J.; Moore, T. D.; Payne, D. J.; Pearson, S.; Polizzi, B. J.; Qiu, X.; Rittenhouse, S. F.; Uzinskas, I. N.; Wallis, N. G.; Huffman, W. F. Indole naphthyridinones as inhibitors of bacterial enoyl-ACP reductases FabI and FabK. J. Med. Chem. 2003, 46 (9), 1627– 35, DOI: 10.1021/jm020403522https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXitlGlur8%253D&md5=0626ec2f4185fa86fadbba7867dc1dd8Indole Naphthyridinones as Inhibitors of Bacterial Enoyl-ACP Reductases FabI and FabKSeefeld, Mark A.; Miller, William H.; Newlander, Kenneth A.; Burgess, Walter J.; DeWolf, Walter E., Jr.; Elkins, Patricia A.; Head, Martha S.; Jakas, Dalia R.; Janson, Cheryl A.; Keller, Paul M.; Manley, Peter J.; Moore, Terrance D.; Payne, David J.; Pearson, Stewart; Polizzi, Brian J.; Qiu, Xiayang; Rittenhouse, Stephen F.; Uzinskas, Irene N.; Wallis, Nicola G.; Huffman, William F.Journal of Medicinal Chemistry (2003), 46 (9), 1627-1635CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Bacterial enoyl-ACP reductase (FabI) is responsible for catalyzing the final step of bacterial fatty acid biosynthesis and is an attractive target for the development of novel antibacterial agents. Previously we reported the development of FabI inhibitor 4 with narrow spectrum antimicrobial activity and in vivo efficacy against Staphylococcus aureus via i.p. administration. Through iterative medicinal chem. aided by x-ray crystal structure anal., a new series of inhibitors has been developed with greatly increased potency against FabI-contg. organisms. Several of these new inhibitors have potent antibacterial activity against multidrug resistant strains of S. aureus, and compd. 30 demonstrates exceptional oral (po) in vivo efficacy in a S. aureus infection model in rats. While optimizing FabI inhibitory activity, compds. 29 and 30 were identified as having low micromolar FabK inhibitory activity, thereby increasing the antimicrobial spectrum of these compds. to include the FabK-contg. pathogens Streptococcus pneumoniae and Enterococcus faecalis. The results described herein support the hypothesis that bacterial enoyl-ACP reductases are valid targets for antibacterial agents.
- 23Ramnauth, J.; Surman, M. D.; Sampson, P. B.; Forrest, B.; Wilson, J.; Freeman, E.; Manning, D. D.; Martin, F.; Toro, A.; Domagala, M.; Awrey, D. E.; Bardouniotis, E.; Kaplan, N.; Berman, J.; Pauls, H. W. 2,3,4,5-Tetrahydro-1H-pyrido[2,3-b and e][1,4]diazepines as inhibitors of the bacterial enoyl ACP reductase, FabI. Bioorg. Med. Chem. Lett. 2009, 19 (18), 5359– 5362, DOI: 10.1016/j.bmcl.2009.07.09423https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtVeqsL3K&md5=b9194f82fe56e322474adcc88e8c0f5e2,3,4,5-Tetrahydro-1H-pyrido[2,3-b and e][1,4]diazepines as inhibitors of the bacterial enoyl ACP reductase, FabIRamnauth, Jailall; Surman, Mathew D.; Sampson, Peter B.; Forrest, Bryan; Wilson, Jeff; Freeman, Emily; Manning, David D.; Martin, Fernando; Toro, Andras; Domagala, Megan; Awrey, Donald E.; Bardouniotis, Elias; Kaplan, Nachum; Berman, Judd; Pauls, Henry W.Bioorganic & Medicinal Chemistry Letters (2009), 19 (18), 5359-5362CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)In the search for new antibacterial agents, the enzyme FabI has been identified as an attractive target. Employing a structure guided approach, the previously reported ene-amide series of FabI inhibitors were expanded to include 2,3,4,5-tetrahydro-1H-pyrido[2,3-b and e][1,4]diazepines, e.g. I (R = 1-methyl-2-indolyl, 3-methyl-2-benzofuranyl, etc.; X = H2, O) and II. These novel series incorporate addnl. H-bonding functions and can be more water sol. than their naphthyridinone progenitors; diazepine II (R = 1-methyl-2-indolyl; X = O) is shown to be efficacious in a mouse infection model.
- 24Takhi, M.; Sreenivas, K.; Reddy, C. K.; Munikumar, M.; Praveena, K.; Sudheer, P.; Rao, B. N.; Ramakanth, G.; Sivaranjani, J.; Mulik, S.; Reddy, Y. R.; Narasimha Rao, K.; Pallavi, R.; Lakshminarasimhan, A.; Panigrahi, S. K.; Antony, T.; Abdullah, I.; Lee, Y. K.; Ramachandra, M.; Yusof, R.; Rahman, N. A.; Subramanya, H. Discovery of azetidine based ene-amides as potent bacterial enoyl ACP reductase (FabI) inhibitors. Eur. J. Med. Chem. 2014, 84, 382– 394, DOI: 10.1016/j.ejmech.2014.07.03624https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXht1Grs7bE&md5=836c98d7e9a806615200c312836adae7Discovery of azetidine based ene-amides as potent bacterial enoyl ACP reductase (FabI) inhibitorsTakhi, Mohamed; Sreenivas, Kandepu; Reddy, Chandrashekar K.; Munikumar, Mahadari; Praveena, Kolakota; Sudheer, Pabolu; Rao, Bandaru N. V. M.; Ramakanth, Gollamudi; Sivaranjani, Jampala; Mulik, Shardaprasad; Reddy, Yeruva R.; Narasimha Rao, Krishnamurthy; Pallavi, Rentala; Lakshminarasimhan, Anirudha; Panigrahi, Sunil K.; Antony, Thomas; Abdullah, Iskandar; Lee, Yean K.; Ramachandra, Murali; Yusof, Rohana; Rahman, Noorsaadah A.; Subramanya, HosahalliEuropean Journal of Medicinal Chemistry (2014), 84 (), 382-394CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A novel and potent series of ene-amides featuring azetidines has been developed as FabI inhibitors active against drug resistant Gram-pos. pathogens particularly staphylococcal organisms. Most of the compds. from the series possessed excellent biochem. inhibition of Staphylococcus aureus FabI enzyme and whole cell activity against clin. relevant MRSA, MSSA and MRSE organisms which are responsible for significant morbidity and mortality in community as well as hospital settings. The binding mode of one of the leads, I, in Escherichia coli FabI enzyme was detd. unambiguously using x-ray crystallog. The lead compds. displayed good metabolic stability in mice liver microsomes and pharmacokinetic profile in mice. The in vivo efficacy of lead AEA16 has been demonstrated in a lethal murine systemic infection model.
- 25Hergenrother, P. J.; Geddes, E. J.; Drown, B. S.; Motika, S. E.; Parker, S. E. Antibiotics Effective for Gram-Negative Pathogens. WO2019/177975A1, 2019.There is no corresponding record for this reference.
- 26Gerusz, V. B.; Bravo, J.; Pauls, H.; Berman, J.; Finn, T. Novel Compounds and their Use. WO2021/123372A1, 2021.There is no corresponding record for this reference.
- 27Hergenrother, P. J.; Parker, E. N.; Hung, D.; Serrano-Wu, M.; Lee, K. K. FabI inhibitors for Gram-negative pathogens. US Provisional Patent Application No. 63/156,145. 2021.There is no corresponding record for this reference.
- 28Geddes, E. J.; Li, Z.; Hergenrother, P. J. An LC-MS/MS assay and complementary web-based tool to quantify and predict compound accumulation in E. coli. Nat. Protoc 2021, 16 (10), 4833– 4854, DOI: 10.1038/s41596-021-00598-y27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFGltrvP&md5=7a4d4b07b133a2e525f8f88c1db3ba11An LC-MS/MS assay and complementary web-based tool to quantify and predict compound accumulation in E. coliGeddes, Emily J.; Li, Zhong; Hergenrother, Paul J.Nature Protocols (2021), 16 (10), 4833-4854CODEN: NPARDW; ISSN:1750-2799. (Nature Portfolio)A review. Novel classes of broad-spectrum antibiotics have been extremely difficult to discover, largely due to the impermeability of the Gram-neg. membranes coupled with a poor understanding of the physicochem. properties a compd. should possess to promote its accumulation inside the cell. To address this challenge, numerous methodologies for assessing intracellular compd. accumulation in Gram-neg. bacteria have been established, including classic radiometric and fluorescence-based methods. The recent development of accumulation assays that utilize liq. chromatog.-tandem mass spectrometry (LC-MS/MS) have circumvented the requirement for labeled compds., enabling assessment of a substantially broader range of small mols. Our unbiased study of accumulation trends in Escherichia coli using an LC-MS/MS-based assay led to the development of the eNTRy rules, which stipulate that a compd. is most likely to accumulate in E. coli if it has an ionizable Nitrogen, has low Three-dimensionality and is relatively Rigid. To aid in the implementation of the eNTRy rules, we developed a complementary web tool, eNTRyway, which calcs. relevant properties and predicts compd. accumulation. Here we provide a comprehensive protocol for anal. and prediction of intracellular accumulation of small mols. in E. coli using an LC-MS/MS-based assay (which takes ∼2 d) and eNTRyway, a workflow that is readily adoptable by any microbiol., biochem. or chem. biol. lab.
- 29Zhu, L.; Lin, J.; Ma, J.; Cronan, J. E.; Wang, H. Triclosan resistance of Pseudomonas aeruginosa PAO1 is due to FabV, a triclosan-resistant enoyl-acyl carrier protein reductase. Antimicrob. Agents Chemother. 2010, 54 (2), 689– 698, DOI: 10.1128/AAC.01152-0928https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhvFSiur4%253D&md5=626edbf70ba35c6ba993ec09a0380eafTriclosan resistance of Pseudomonas aeruginosa PAO1 is due to FabV, a triclosan-resistant enoyl-acyl carrier protein reductaseZhu, Lei; Lin, Jinshui; Ma, Jincheng; Cronan, John E.; Wang, HaihongAntimicrobial Agents and Chemotherapy (2010), 54 (2), 689-698CODEN: AMACCQ; ISSN:0066-4804. (American Society for Microbiology)Triclosan, a very widely used biocide, specifically inhibits fatty acid synthesis by inhibition of enoyl-acyl carrier protein (ACP) reductase. Escherichia coli FabI is the prototypical triclosan-sensitive enoyl-ACP reductase, and E. coli is extremely sensitive to the biocide. However, other bacteria are resistant to triclosan, because they encode triclosan-resistant enoyl-ACP reductase isoenzymes. In contrast, the triclosan resistance of Pseudomonas aeruginosa PAO1 has been attributed to active efflux of the compd. We report that P. aeruginosa contains two enoyl-ACP reductase isoenzymes, the previously characterized FabI homolog plus a homolog of FabV, a triclosan-resistant enoyl-ACP reductase recently demonstrated in Vibrio cholerae. By deletion of the genes encoding P. aeruginosa FabI and FabV, we demonstrated that FabV confers triclosan resistance on P. aeruginosa. Upon deletion of the fabV gene, the mutant strain became extremely sensitive to triclosan (>2,000-fold more sensitive than the wild-type strain), whereas the mutant strain lacking FabI remained completely resistant to the biocide.
- 30Parsons, J. B.; Frank, M. W.; Subramanian, C.; Saenkham, P.; Rock, C. O. Metabolic basis for the differential susceptibility of Gram-positive pathogens to fatty acid synthesis inhibitors. Proc. Natl. Acad. Sci. U. S. A. 2011, 108 (37), 15378– 15383, DOI: 10.1073/pnas.110920810829https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht1KisLrJ&md5=c5f9058eee8fb610fce3934e655b6dadMetabolic basis for the differential susceptibility of gram-positive pathogens to fatty acid synthesis inhibitorsParsons, Joshua B.; Frank, Matthew W.; Subramanian, Chitra; Saenkham, Panatda; Rock, Charles O.Proceedings of the National Academy of Sciences of the United States of America (2011), 108 (37), 15378-15383, S15378/1-S15378/10CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The rationale for the pursuit of bacterial type 2 fatty acid synthesis (FASII) as a target for antibacterial drug discovery in Gram-pos. organisms is being debated vigorously based on their ability to incorporate extracellular fatty acids. The regulation of FASII by extracellular fatty acids was examd. in Staphylococcus aureus and Streptococcus pneumoniae, representing two important groups of pathogens. Both bacteria use the same enzymic tool kit for the conversion of extracellular fatty acids to acyl-acyl carrier protein, elongation, and incorporation into phospholipids. Exogenous fatty acids completely replace the endogenous fatty acids in S. pneumoniae but support only 50% of phospholipid synthesis in S. aureus. Fatty acids overcame FASII inhibition in S. pneumoniae but not in S. aureus. Extracellular fatty acids strongly suppress malonyl-CoA levels in S. pneumoniae but not in S. aureus, showing a feedback regulatory system in S. pneumoniae that is absent in S. aureus. Fatty acids overcame either a biochem. or a genetic block at acetyl-CoA carboxylase (ACC) in S. aureus, confirming that regulation at the ACC step is the key difference between these two species. Bacteria that possess a stringent biochem. feedback inhibition of ACC and malonyl-CoA formation triggered by environmental fatty acids are able to circumvent FASII inhibition. However, if exogenous fatty acids do not suppress malonyl-CoA formation, FASII inhibitors remain effective in the presence of fatty acid supplements.
- 31Zhu, L.; Bi, H.; Ma, J.; Hu, Z.; Zhang, W.; Cronan, J. E.; Wang, H. The two functional enoyl-acyl carrier protein reductases of Enterococcus faecalis do not mediate triclosan resistance. mBio 2013, 4 (5), e00613– 13, DOI: 10.1128/mBio.00613-1330https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXntVart7g%253D&md5=4dd4f9fb0167083d7a096ef36e9f0c64The two functional enoyl-acyl carrier protein reductases of Enterococcus faecalis do not mediate triclosan resistanceZhu, Lei; Bi, Hongkai; Ma, Jincheng; Hu, Zhe; Zhang, Wenbin; Cronan, John E.; Wang, HaihongmBio (2013), 4 (5), e00613/1-e00613/11CODEN: MBIOCL; ISSN:2150-7511. (American Society for Microbiology)Enoyl-acyl carrier protein (enoyl-ACP) reductase catalyzes the last step of the elongation cycle in the synthesis of bacterial fatty acids. The Enterococcus faecalis genome contains two genes annotated as enoyl-ACP reductases, a FabI-type enoyl- ACP reductase and a FabK-type enoyl-ACP reductase. The authors report that expression of either of the two proteins restores growth of an Escherichia coli fabI temp.-sensitive mutant strain under nonpermissive conditions. In vitro assays demonstrated that both proteins support fatty acid synthesis and are active with substrates of all fatty acid chain lengths. Although expression of E. faecalis fabK confers to E. coli high levels of resistance to the antimicrobial triclosan, deletion of fabK from the E. faecalis genome showed that FabK does not play a detectable role in the inherent triclosan resistance of E. faecalis. Indeed, FabK seems to play only a minor role in modulating fatty acid compn. Strains carrying a deletion of fabK grow normally without fatty acid supplementation, whereas fabI deletion mutants make only traces of fatty acids and are unsatd. fatty acid auxotrophs.
- 32Yao, J.; Carter, R. A.; Vuagniaux, G.; Barbier, M.; Rosch, J. W.; Rock, C. O. A Pathogen-Selective Antibiotic Minimizes Disturbance to the Microbiome. Antimicrob. Agents Chemother. 2016, 60 (7), 4264– 73, DOI: 10.1128/AAC.00535-1631https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtVyksLk%253D&md5=0bc461975d3a19a2696bb7cc66f4744fA pathogen-selective antibiotic minimizes disturbance to the microbiomeYao, Jiangwei; Carter, Robert A.; Vuagniaux, Gregoire; Barbier, Maryse; Rosch, Jason W.; Rock, Charles O.Antimicrobial Agents and Chemotherapy (2016), 60 (7), 4264-4273CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)Broad-spectrum antibiotic therapy decimates the gut microbiome, resulting in a variety of neg. health consequences. Debio 1452 is a staphylococcus-selective enoyl-acyl carrier protein reductase (FabI) inhibitor under clin. development and was used to det. whether treatment with pathogen-selective antibiotics would minimize disturbance to the microbiome. The effect of oral Debio 1452 on the microbiota of mice was compared to the effects of four commonly used broad-spectrum oral antibiotics. During the 10 days of oral Debio 1452 treatment, there was minimal disturbance to the gut bacterial abundance and compn., with only the unclassified S24-7 taxon reduced at days 6 and 10. In comparison, broad-spectrum oral antibiotics caused ∼100- to 4000-fold decreases in gut bacterial abundance and severely altered the microbial compn. The gut bacterial abundance and compn. of Debio 1452-treated mice were indistinguishable from those of untreated mice 2 days after the antibiotic treatment was stopped. In contrast, the bacterial abundance in broad-spectrum-antibiotic-treated mice took up to 7 days to recover, and the gut compn. of the broad-spectrum-antibiotic-treated mice remained different from that of the control group 20 days after the cessation of antibiotic treatment. These results illustrate that a pathogen-selective approach to antibiotic development will minimize disturbance to the gut microbiome.
- 33Imai, Y.; Meyer, K. J.; Iinishi, A.; Favre-Godal, Q.; Green, R.; Manuse, S.; Caboni, M.; Mori, M.; Niles, S.; Ghiglieri, M.; Honrao, C.; Ma, X.; Guo, J. J.; Makriyannis, A.; Linares-Otoya, L.; Bohringer, N.; Wuisan, Z. G.; Kaur, H.; Wu, R.; Mateus, A.; Typas, A.; Savitski, M. M.; Espinoza, J. L.; O’Rourke, A.; Nelson, K. E.; Hiller, S.; Noinaj, N.; Schaberle, T. F.; D’Onofrio, A.; Lewis, K. A new antibiotic selectively kills Gram-negative pathogens. Nature 2019, 576 (7787), 459– 464, DOI: 10.1038/s41586-019-1791-132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlOmurfP&md5=3b7105cfeff0ae8970065bd94e6b12eeA new antibiotic selectively kills Gram-negative pathogensImai, Yu; Meyer, Kirsten J.; Iinishi, Akira; Favre-Godal, Quentin; Green, Robert; Manuse, Sylvie; Caboni, Mariaelena; Mori, Miho; Niles, Samantha; Ghiglieri, Meghan; Honrao, Chandrashekhar; Ma, Xiaoyu; Guo, Jason J.; Makriyannis, Alexandros; Linares-Otoya, Luis; Bohringer, Nils; Wuisan, Zerlina G.; Kaur, Hundeep; Wu, Runrun; Mateus, Andre; Typas, Athanasios; Savitski, Mikhail M.; Espinoza, Josh L.; O'Rourke, Aubrie; Nelson, Karen E.; Hiller, Sebastian; Noinaj, Nicholas; Schaberle, Till F.; D'Onofrio, Anthony; Lewis, KimNature (London, United Kingdom) (2019), 576 (7787), 459-464CODEN: NATUAS; ISSN:0028-0836. (Nature Research)The current need for novel antibiotics is esp. acute for drug-resistant Gram-neg. pathogens1,2. These microorganisms have a highly restrictive permeability barrier, which limits the penetration of most compds.3,4. As a result, the last class of antibiotics that acted against Gram-neg. bacteria was developed in the 1960s2. We reason that useful compds. can be found in bacteria that share similar requirements for antibiotics with humans, and focus on Photorhabdus symbionts of entomopathogenic nematode microbiomes. Here we report a new antibiotic that we name darobactin, which was obtained using a screen of Photorhabdus isolates. Darobactin is coded by a silent operon with little prodn. under lab. conditions, and is ribosomally synthesized. Darobactin has an unusual structure with two fused rings that form post-translationally. The compd. is active against important Gram-neg. pathogens both in vitro and in animal models of infection. Mutants that are resistant to darobactin map to BamA, an essential chaperone and translocator that folds outer membrane proteins. Our study suggests that bacterial symbionts of animals contain antibiotics that are particularly suitable for development into therapeutics.
- 34Leimer, N.; Wu, X.; Imai, Y.; Morrissette, M.; Pitt, N.; Favre-Godal, Q.; Iinishi, A.; Jain, S.; Caboni, M.; Leus, I. V.; Bonifay, V.; Niles, S.; Bargabos, R.; Ghiglieri, M.; Corsetti, R.; Krumpoch, M.; Fox, G.; Son, S.; Klepacki, D.; Polikanov, Y. S.; Freliech, C. A.; McCarthy, J. E.; Edmondson, D. G.; Norris, S. J.; D’Onofrio, A.; Hu, L. T.; Zgurskaya, H. I.; Lewis, K. A selective antibiotic for Lyme disease. Cell 2021, 184 (21), 5405– 5418, DOI: 10.1016/j.cell.2021.09.01133https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitF2ntrnO&md5=3eb28b19cf0bfd3e00e4dd374cfe70bbA selective antibiotic for Lyme diseaseLeimer, Nadja; Wu, Xiaoqian; Imai, Yu; Morrissette, Madeleine; Pitt, Norman; Favre-Godal, Quentin; Iinishi, Akira; Jain, Samta; Caboni, Mariaelena; Leus, Inga V.; Bonifay, Vincent; Niles, Samantha; Bargabos, Rachel; Ghiglieri, Meghan; Corsetti, Rachel; Krumpoch, Megan; Fox, Gabriel; Son, Sangkeun; Klepacki, Dorota; Polikanov, Yury S.; Freliech, Cecily A.; McCarthy, Julie E.; Edmondson, Diane G.; Norris, Steven J.; D'Onofrio, Anthony; Hu, Linden T.; Zgurskaya, Helen I.; Lewis, KimCell (Cambridge, MA, United States) (2021), 184 (21), 5405-5418.e16CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Lyme disease is on the rise. Caused by a spirochete Borreliella burgdorferi, it affects an estd. 500,000 people in the United States alone. The antibiotics currently used to treat Lyme disease are broad spectrum, damage the microbiome, and select for resistance in non-target bacteria. We therefore sought to identify a compd. acting selectively against B. burgdorferi. A screen of soil micro-organisms revealed a compd. highly selective against spirochetes, including B. burgdorferi. Unexpectedly, this compd. was detd. to be hygromycin A, a known antimicrobial produced by Streptomyces hygroscopicus. Hygromycin A targets the ribosomes and is taken up by B. burgdorferi, explaining its selectivity. Hygromycin A cleared the B. burgdorferi infection in mice, including animals that ingested the compd. in a bait, and was less disruptive to the fecal microbiome than clin. relevant antibiotics. This selective antibiotic holds the promise of providing a better therapeutic for Lyme disease and eradicating it in the environment.
- 35Poyet, M.; Groussin, M.; Gibbons, S. M.; Avila-Pacheco, J.; Jiang, X.; Kearney, S. M.; Perrotta, A. R.; Berdy, B.; Zhao, S.; Lieberman, T. D.; Swanson, P. K.; Smith, M.; Roesemann, S.; Alexander, J. E.; Rich, S. A.; Livny, J.; Vlamakis, H.; Clish, C.; Bullock, K.; Deik, A.; Scott, J.; Pierce, K. A.; Xavier, R. J.; Alm, E. J. A library of human gut bacterial isolates paired with longitudinal multiomics data enables mechanistic microbiome research. Nat. Med. 2019, 25 (9), 1442– 1452, DOI: 10.1038/s41591-019-0559-334https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhs12gurrP&md5=016ca835384cb8d48bbc80f21c5284beA library of human gut bacterial isolates paired with longitudinal multiomics data enables mechanistic microbiome researchPoyet, M.; Groussin, M.; Gibbons, S. M.; Avila-Pacheco, J.; Jiang, X.; Kearney, S. M.; Perrotta, A. R.; Berdy, B.; Zhao, S.; Lieberman, T. D.; Swanson, P. K.; Smith, M.; Roesemann, S.; Alexander, J. E.; Rich, S. A.; Livny, J.; Vlamakis, H.; Clish, C.; Bullock, K.; Deik, A.; Scott, J.; Pierce, K. A.; Xavier, R. J.; Alm, E. J.Nature Medicine (New York, NY, United States) (2019), 25 (9), 1442-1452CODEN: NAMEFI; ISSN:1078-8956. (Nature Research)Our understanding of how the gut microbiome interacts with its human host has been restrained by limited access to longitudinal datasets to examine stability and dynamics, and by having only a few isolates to test mechanistic hypotheses. Here, we present the Broad Institute-OpenBiome Microbiome Library (BIO-ML), a comprehensive collection of 7,758 gut bacterial isolates paired with 3,632 genome sequences and longitudinal multi-omics data. We show that microbial species maintain stable population sizes within and across humans and that commonly used 'omics' survey methods are more reliable when using avs. over multiple days of sampling. Variation of gut metabolites within people over time is assocd. with amino acid levels, and differences across people are assocd. with differences in bile acids. Finally, we show that genomic diversification can be used to infer eco-evolutionary dynamics and in vivo selection pressures for strains within individuals. The BIO-ML is a unique resource designed to enable hypothesis-driven microbiome research.
- 36Marrakchi, H.; Dewolf, W. E., Jr.; Quinn, C.; West, J.; Polizzi, B. J.; So, C. Y.; Holmes, D. J.; Reed, S. L.; Heath, R. J.; Payne, D. J.; Rock, C. O.; Wallis, N. G. Characterization of Streptococcus pneumoniae enoyl-(acyl-carrier protein) reductase (FabK). Biochem. J. 2003, 370 (3), 1055– 1062, DOI: 10.1042/bj2002169935https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXhvFKhsLc%253D&md5=abef169e126ec1ea70ea8c291db1a3a1Characterization of Streptococcus pneumoniae enoyl-(acyl-carrier protein) reductase (FabK)Marrakchi, Hedia; Dewolf, Walter E., Jr.; Quinn, Chad; West, Joshua; Polizzi, Brian J.; So, Chi Y.; Holmes, David J.; Reed, Shannon L.; Heath, Richard J.; Payne, David J.; Rock, Charles O.; Wallis, Nicola G.Biochemical Journal (2003), 370 (3), 1055-1062CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)The enoyl-(acyl-carrier protein) (ACP) reductase catalyzes the last step in each cycle of fatty acid elongation in the type II fatty acid synthase systems. An extensively characterized NADH-dependent reductase, FabI, is widely distributed in bacteria and plants, whereas the enoyl-ACP reductase, FabK, is a distinctly different member of this enzyme group discovered in Streptococcus pneumoniae . We were unable to delete the fabK gene from Strep. pneumoniae , suggesting that this is the only enoyl-ACP reductase in this organism. The FabK enzyme was purified and the biochem. properties of the reductase were examd. The visible absorption spectrum of the purified protein indicated the presence of a flavin cofactor that was identified as FMN by MS, and was present in a 1:1 molar ratio with protein. FabK specifically required NADH and the protein activity was stimulated by ammonium ions. FabK also exhibited NADH oxidase activity in the absence of substrate. Strep. pneumoniae belongs to the Bacillus/Lactobacillus/Streptococcus group that includes Staphylococcus aureus and Bacillus subtilis . These two organisms also contain FabK-related genes, suggesting that they may also express a FabK-like enoyl-ACP reductase. However, the genes did not complement a fabI (Ts) mutant and the purified flavoproteins were unable to reduce enoyl-ACP in vitro and did not exhibit NAD(P)H oxidase activity, indicating they were not enoyl-ACP reductases. The restricted occurrence of the FabK enoyl-ACP reductase may be related to the role of substrate-independent NADH oxidn. in oxygen-dependent anaerobic energy metab.
- 37Armstrong, J. D.; Eng, K. K.; Keller, J. L.; Purick, R. M.; Hartner, F. W.; Choi, W.-B.; Askin, D.; Volante, R.P. An Efficient Asymmetric Synthesis of (R)-3-Amino-2,3,4,5-tetrahydro-1H-[1]benzazepin-2-one. Tetrahedron Lett. 1994, 35 (20), 3239– 3242, DOI: 10.1016/S0040-4039(00)76874-236https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2cXkvVejsb4%253D&md5=62d7610af0359f04618b99e65337b5acAn efficient asymmetric synthesis of (R)-3-amino-2,3,4,5-tetrahydro-1H-[1]benzazepin-2-oneArmstrong, Joseph D., III; Eng, Kan K.; Keller, Jennifer L.; Purick, Robert M.; Hartner, Frederick W., Jr.; Choi, Woo-Baeg; Askin, David; Volante, R. P.Tetrahedron Letters (1994), 35 (20), 3239-42CODEN: TELEAY; ISSN:0040-4039.Two approaches for the asym. prepn. of (-)- or (+)-α-aminobenzlactam I are described. One route is based on the asym. hydrogenation of enamide II and the other on the racemization/resoln. of (±)-I.
- 38Galac, M. R.; Snesrud, E.; Lebreton, F.; Stam, J.; Julius, M.; Ong, A. C.; Maybank, R.; Jones, A. R.; Kwak, Y. I.; Hinkle, K.; Waterman, P. E.; Lesho, E. P.; Bennett, J. W.; Mc Gann, P. A Diverse Panel of Clinical Acinetobacter baumannii for Research and Development. Antimicrob. Agents Chemother. 2020, 64 (10), e00840-20, DOI: 10.1128/AAC.00840-20There is no corresponding record for this reference.
- 39Rao, N. K.; Nataraj, V.; Ravi, M.; Panchariya, L.; Palai, K.; Talapati, S. R.; Lakshminarasimhan, A.; Ramachandra, M.; Antony, T. Ternary complex formation of AFN-1252 with Acinetobacter baumannii FabI and NADH: Crystallographic and biochemical studies. Chem. Biol. Drug Des 2020, 96 (2), 704– 713, DOI: 10.1111/cbdd.1368638https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXns1Cnsrs%253D&md5=620921197d60d1f4c194ab5eef70dde8Ternary complex formation of AFN-1252 with Acinetobacter baumannii FabI and NADH: Crystallographic and biochemical studiesRao, Narasimha K.; Nataraj, Vijayashankar; Ravi, Mohan; Panchariya, Love; Palai, Kirttija; Talapati, Sumalatha R.; Lakshminarasimhan, Anirudha; Ramachandra, Murali; Antony, ThomasChemical Biology & Drug Design (2020), 96 (2), 704-713CODEN: CBDDAL; ISSN:1747-0277. (Wiley-Blackwell)Acinetobacter baumannii is an opportunistic Gram-neg. bacterial pathogen, assocd. mostly with hospital-acquired infections. The emergence of drug resistance strains made it necessary to explore new pathways for the development of more effective antibiotics. Enoyl CoA reductase (FabI), a key enzyme in the fatty acid biosynthesis (FAS) pathway, has emerged as a potential target for antibacterial drug development. Earlier reports show that the lead SaFabI inhibitor AFN-1252 can inhibit FabI from other organisms including Escherichia coli and Burkholderia pseudomallei, but with differential potency. In the present work, we show that AFN-1252 is a moderate inhibitor of AbFabI with an IC50 of 216 nM. AFN-1252 stabilized AbFabI with a 4.2°C increase in the melting temp. (Tm) and, interestingly, the stabilization effect was significantly increased in presence of the cofactor NADH (ΔTm = 17°C), suggesting the formation of a ternary complex AbFabI: AFN-1252: NADH. X-ray crystallog. studies of AbFabI co-crystd. with AFN-1252 and NADH confirmed the ternary complex formation. The crit. interactions of AFN-1252 with AbFabI and NADH identified from the co-crystal structure may facilitate the design and development of new drugs against A. baumannii infections by targeting the FAS pathway.
- 40Sihra, N.; Goodman, A.; Zakri, R.; Sahai, A.; Malde, S. Nonantibiotic prevention and management of recurrent urinary tract infection. Nat. Rev. Urol 2018, 15 (12), 750– 776, DOI: 10.1038/s41585-018-0106-x39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3cvjtlGhsg%253D%253D&md5=7bb6b1d6bb8b83bf93269ab7105b964aNonantibiotic prevention and management of recurrent urinary tract infectionSihra Neha; Zakri Rhana; Sahai Arun; Malde Sachin; Goodman AnnaNature reviews. Urology (2018), 15 (12), 750-776 ISSN:.Urinary tract infections (UTIs) are highly prevalent, lead to considerable patient morbidity, incur large financial costs to health-care systems and are one of the most common reasons for antibiotic use worldwide. The growing problem of antimicrobial resistance means that the search for nonantibiotic alternatives for the treatment and prevention of UTI is of critical importance. Potential nonantibiotic measures and treatments for UTIs include behavioural changes, dietary supplementation (such as Chinese herbal medicines and cranberry products), NSAIDs, probiotics, D-mannose, methenamine hippurate, estrogens, intravesical glycosaminoglycans, immunostimulants, vaccines and inoculation with less-pathogenic bacteria. Some of the results of trials of these approaches are promising; however, high-level evidence is required before firm recommendations for their use can be made. A combination of these agents might provide the optimal treatment to reduce recurrent UTI, and trials in specific population groups are required.
- 41Flores-Mireles, A. L.; Walker, J. N.; Caparon, M.; Hultgren, S. J. Urinary tract infections: epidemiology, mechanisms of infection and treatment options. Nat. Rev. Microbiol 2015, 13 (5), 269– 284, DOI: 10.1038/nrmicro343240https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXmtVaqs78%253D&md5=b81a25f1f4ab04770982ee01b41b2427Urinary tract infections: epidemiology, mechanisms of infection and treatment optionsFlores-Mireles, Ana L.; Walker, Jennifer N.; Caparon, Michael; Hultgren, Scott J.Nature Reviews Microbiology (2015), 13 (5), 269-284CODEN: NRMACK; ISSN:1740-1526. (Nature Publishing Group)Urinary tract infections (UTIs) are a severe public health problem and are caused by a range of pathogens, but most commonly by Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, Enterococcus faecalis and Staphylococcus saprophyticus. High recurrence rates and increasing antimicrobial resistance among uropathogens threaten to greatly increase the economic burden of these infections. In this Review, we discuss how basic science studies are elucidating the mol. details of the crosstalk that occurs at the host-pathogen interface, as well as the consequences of these interactions for the pathophysiol. of UTIs. We also describe current efforts to translate this knowledge into new clin. treatments for UTIs.
- 42Goebel, M. C.; Trautner, B. W.; Grigoryan, L. The Five Ds of Outpatient Antibiotic Stewardship for Urinary Tract Infections. Clin Microbiol Rev. 2021, 34 (4), e0000320, DOI: 10.1128/CMR.00003-20There is no corresponding record for this reference.
- 43Nielubowicz, G. R.; Mobley, H. L. Host-pathogen interactions in urinary tract infection. Nat. Rev. Urol 2010, 7 (8), 430– 441, DOI: 10.1038/nrurol.2010.10142https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVSmsbzF&md5=84927025a6555c9a93860619e82571ccHost-pathogen interactions in urinary tract infectionNielubowicz, Greta R.; Mobley, Harry L. T.Nature Reviews Urology (2010), 7 (8), 430-441CODEN: NRNADQ; ISSN:1759-4812. (Nature Publishing Group)A review. Urinary tract infections are experienced by a large section of the population and cause significant morbidity. Here, Nielubowicz and Mobley explain the pathogenic mechanisms utilized by Escherichia coli and Proteus mirabilis during complicated and uncomplicated infection, resp. They outline the strategies currently underway to exploit these processes for the identification of new treatment options. The urinary tract is a common site of bacterial infections; nearly half of all women experience at least one urinary tract infection (UTI) during their lifetime. These infections are classified based on the condition of the host. Uncomplicated infections affect otherwise healthy individuals and are most commonly caused by uropathogenic Escherichia coli, whereas complicated infections affect patients with underlying difficulties, such as a urinary tract abnormality or catheterization, and are commonly caused by species such as Proteus mirabilis. Virulence and fitness factors produced by both pathogens include fimbriae, toxins, flagella, iron acquisition systems, and proteins that function in immune evasion. Addnl. factors that contribute to infection include the formation of intracellular bacterial communities by E. coli and the prodn. of urease by P. mirabilis, which can result in urinary stone formation. Innate immune responses are induced or mediated by pattern recognition receptors, antimicrobial peptides, and neutrophils. The adaptive immune response to UTI is less well understood. Host factors TLR4 and CXCR1 are implicated in disease outcome and susceptibility, resp. Low levels of TLR4 are assocd. with asymptomatic bacteriuria while low levels of CXCR1 are assocd. with increased incidence of acute pyelonephritis. Current research is focused on the identification of addnl. virulence factors and therapeutic or prophylactic targets that might be used in the generation of vaccines against both uropathogens.
- 44Flamm, R. K.; Rhomberg, P. R.; Kaplan, N.; Jones, R. N.; Farrell, D. J. Activity of Debio1452, a FabI inhibitor with potent activity against Staphylococcus aureus and coagulase-negative Staphylococcus spp., including multidrug-resistant strains. Antimicrob. Agents Chemother. 2015, 59 (5), 2583– 2587, DOI: 10.1128/AAC.05119-1443https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXntVGjsbw%253D&md5=9b0e6360938e855fe2513d48d8016cacActivity of Debio1452, a FabI inhibitor with potent activity against Staphylococcus aureus and coagulase-negative Staphylococcus spp., including multidrug-resistant strainsFlamm, Robert K.; Rhomberg, Paul R.; Kaplan, Nachum; Jones, Ronald N.; Farrell, David J.Antimicrobial Agents and Chemotherapy (2015), 59 (5), 2583-2587CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)Staphylococcus aureus and coagulase-neg. staphylococci (CoNS) are responsible for a wide variety of human infections. The investigational antibacterial Debio1450 (previously AFN-1720), a prodrug of Debio1452 (previously AFN-1252), specifically targets staphylococci without significant activity against other Gram-pos. or Gram-neg. species. Debio1452 inhibits FabI, an enzyme crit. to fatty acid biosynthesis in staphylococci. The activity of Debio1452 against CoNS, methicillin-susceptible S. aureus (MSSA), and methicillin-resistant S. aureus (MRSA), including significant clones, was detd. A globally diverse collection of 574 patient isolates from 35 countries was tested that included CoNS (6 species, 103 strains), MSSA (154 strains), MRSA (163 strains), and molecularly characterized strains (including spa-typed MRSA clones; 154 strains). The isolates were tested for susceptibility by CLSI broth microdilution methods against Debio1452 and 10 comparators. The susceptibility rates for the comparators were detd. using CLSI and EUCAST breakpoint criteria. All S. aureus and CoNS strains were inhibited by Debio1452 concns. of ≤0.12 and ≤0.5 μg/mL, resp. The MIC50s for MSSA, MRSA, and molecularly characterized MRSA strains were 0.004 μg/mL, and the MIC90s ranged from 0.008 to 0.03 μg/mL. The MICs were higher for the CoNS isolates (MIC50/90, 0.015/0.12 μg/mL). Among S. aureus strains, resistance was common for erythromycin (61.6%), levofloxacin (49.0%), clindamycin (27.6%), tetracycline (15.7%), and trimethoprim-sulfamethoxazole (7.0%). Debio1452 demonstrated potent activity against MSSA, MRSA, and CoNS. Debio1452 showed significantly greater activity overall (MIC50, 0.004 μg/mL) than the other agents tested against these staphylococcal species, which included dominant MRSA clones and strains resistant to currently used antimicrobial agents.
- 45Hafkin, B.; Kaplan, N.; Murphy, B. Efficacy and Safety of AFN-1252, the First Staphylococcus-Specific Antibacterial Agent, in the Treatment of Acute Bacterial Skin and Skin Structure Infections, Including Those in Patients with Significant Comorbidities. Antimicrob. Agents Chemother. 2016, 60 (3), 1695– 701, DOI: 10.1128/AAC.01741-1544https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsF2ntbnF&md5=a108d5ddd834788bbdb10d57dd07af4eEfficacy and safety of AFN-1252, the first Staphylococcus-specific antibacterial agent, in the treatment of acute bacterial skin and skin structure infections, including those in patients with significant comorbiditiesHafkin, B.; Kaplan, N.; Murphy, B.Antimicrobial Agents and Chemotherapy (2016), 60 (3), 1695-1701CODEN: AMACCQ; ISSN:1098-6596. (American Society for Microbiology)This open-label noncontrolled, phase II multicenter trial was designed to evaluate the safety, tolerability, and efficacy of 200 mg of AFN-1252, a selective inhibitor of Staphylococcus aureus enoyl-acyl carrier protein reductase (FabI), given by mouth twice daily in the treatment of acute bacterial skin and skin structure infections (ABSSSI) due to staphylococci. Important aspects of the current study included a comparison of early response efficacy endpoints with end-of-treatment and follow-up endpoints. Many patients in the intent-to-treat population (n = 103) had significant comorbidities. The overall early response rate at day 3 was 97.3% (wound, 100%; abscess, 96.6%; cellulitis, 94.4%) in the microbiol. evaluable (ME) population. Within the ME population, 82.9% of patients had a ≥ 20% decrease in the area of erythema, and 77.9% of patients had a ≥ 20% decrease in the area of induration, on day 3. S. aureus was detected in 97.7% of patients (n = 37 patients with methicillin-resistant S. aureus [MRSA], and n = 39 with methicillin-sensitive S. aureus [MSSA]). No isolates had increased AFN-1252 MICs posttreatment. Microbiol. eradication rates for S. aureus were 93.2% at short-term follow-up (STFU) and 91.9% at long-term follow-up (LTFU) in the ME population. Eradication rates for MRSA and MSSA were 91.9% and 92.3%, resp., at STFU and 91.9% and 89.7%, resp., at LTFU. The most frequently reported drug-related adverse events, which were mostly mild or moderate, were headache (26.2%) and nausea (21.4%). These studies demonstrate that AFN-1252 is generally well tolerated and effective in the treatment of ABSSSI due to S. aureus, including MRSA. This study has been registered at ClinicalTrials.gov under registration no.NCT01519492.
- 46Radka, C. D.; Rock, C. O. Mining Fatty Acid Biosynthesis for New Antimicrobials. Annu. Rev. Microbiol. 2022, 76, DOI: 10.1146/annurev-micro-041320-110408There is no corresponding record for this reference.
- 47Payne, D. J.; Warren, P. V.; Holmes, D. J.; Ji, Y.; Lonsdale, J. T. Bacterial fatty-acid biosynthesis: a genomics-driven target for antibacterial drug discovery. Drug Discov Today 2001, 6 (10), 537– 544, DOI: 10.1016/S1359-6446(01)01774-346https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXjtlCjsbo%253D&md5=50cbe09ba8e48f9aca6a5b847477f24cBacterial fatty-acid biosynthesis: a genomics-driven target for antibacterial drug discoveryPayne, D. J.; Warren, P. V.; Holmes, D. J.; Ji, Y.; Lonsdale, J. T.Drug Discovery Today (2001), 6 (10), 537-544CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Science Ltd.)A review with 46 refs. demonstrating how the interplay of genomics, bioinformatics and genomic technologies has enabled an in-depth anal. of the component enzymes of the bacterial fatty-acid biosynthesis pathway as a source of novel antibacterial targets. This evaluation has revealed that many of the enzymes are potentially selective, broad-spectrum antibacterial targets. We also illustrate the suitability of some of these targets for HTS. Furthermore, we discuss how the availability of a robust selectivity assay, mode-of-action assays and numerous crystal structures provide an excellent set of tools with which to initiate integrated programs of research to identify novel antibiotics targeted at these enzymes.
- 48Yao, J.; Rock, C. O. Bacterial fatty acid metabolism in modern antibiotic discovery. Biochim Biophys Acta Mol. Cell Biol. Lipids 2017, 1862 (11), 1300– 1309, DOI: 10.1016/j.bbalip.2016.09.01447https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFKmur3E&md5=c6b0c7f2c7997bed206046b62041e965Bacterial fatty acid metabolism in modern antibiotic discoveryYao, Jiangwei; Rock, Charles O.Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids (2017), 1862 (11), 1300-1309CODEN: BBMLFG; ISSN:1388-1981. (Elsevier B.V.)Bacterial fatty acid synthesis is essential for many pathogens and different from the mammalian counterpart. These features make bacterial fatty acid synthesis a desirable target for antibiotic discovery. The structural divergence of the conserved enzymes and the presence of different isoenzymes catalyzing the same reactions in the pathway make bacterial fatty acid synthesis a narrow spectrum target rather than the traditional broad spectrum target. Furthermore, bacterial fatty acid synthesis inhibitors are single-targeting, rather than multi-targeting like traditional monotherapeutic, broad-spectrum antibiotics. The single-targeting nature of bacterial fatty acid synthesis inhibitors makes overcoming fast-developing, target-based resistance a necessary consideration for antibiotic development. Target-based resistance can be overcome through multi-targeting inhibitors, a cocktail of single-targeting inhibitors, or by making the single targeting inhibitor sufficiently high affinity through a pathogen selective approach such that target-based mutants are still susceptible to therapeutic concns. of drug. Many of the pathogens requiring new antibiotic treatment options encode for essential bacterial fatty acid synthesis enzymes. This review will evaluate the most promising targets in bacterial fatty acid metab. for antibiotic therapeutics development and review the potential and challenges in advancing each of these targets to the clinic and circumventing target-based resistance. This article is part of a Special Issue entitled: Bacterial Lipids edited by Russell E. Bishop.
- 49Staley, C.; Vaughn, B. P.; Graiziger, C. T.; Sadowsky, M. J.; Khoruts, A. Gut-sparing treatment of urinary tract infection in patients at high risk of Clostridium difficile infection. J. Antimicrob. Chemother. 2017, 72 (2), 522– 528, DOI: 10.1093/jac/dkw49948https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFKju7nK&md5=1d8a5c68e27d005d9c0a022569efb66eGut-sparing treatment of urinary tract infection in patients at high risk of Clostridium difficile infectionStaley, Christopher; Vaughn, Byron P.; Graiziger, Carolyn T.; Sadowsky, Michael J.; Khoruts, AlexanderJournal of Antimicrobial Chemotherapy (2017), 72 (2), 522-528CODEN: JACHDX; ISSN:1460-2091. (Oxford University Press)Recipients of fecal microbiota transplantation (FMT) in treatment of recurrent Clostridium difficile infection (RCDI) remain at markedly increased risk of re-infection with C. difficile with new antibiotic provocations. Urinary tract infections (UTIs) are common indications for antibiotics in these patients, often resulting in C. difficile re-infection. We present a case series of 19 patients treated with parenteral aminoglycosides for UTI following FMT for RCDI. A 3 day outpatient regimen of once-daily i.m. administration of gentamicin was used to treat 18 consecutive FMT recipients with uncomplicated UTI. One other patient was treated for a complicated UTI with i.v. amikacin. Profiling of 16S rRNA genes was used to track changes in fecal microbial community structure during this regimen in three patients. The protocol was highly effective in treating UTI symptoms. None of the patients suffered a re-infection with C. difficile. The fecal microbial communities remained undisturbed by treatment with i.m. administration of gentamicin. Despite falling out of favor in recent years, aminoglycoside antibiotics given parenterally have the advantage of minimal penetration into the gut lumen. A brief (3 day) course of parenteral gentamicin was safe and effective in curing UTI in patients at high risk of C. difficile infection without perturbing their gut microbiota.
- 50Worby, C. J.; Schreiber, H. L. t.; Straub, T. J.; van Dijk, L. R.; Bronson, R. A.; Olson, B. S.; Pinkner, J. S.; Obernuefemann, C. L. P.; Munoz, V. L.; Paharik, A. E.; Azimzadeh, P. N.; Walker, B. J.; Desjardins, C. A.; Chou, W. C.; Bergeron, K.; Chapman, S. B.; Klim, A.; Manson, A. L.; Hannan, T. J.; Hooton, T. M.; Kau, A. L.; Lai, H. H.; Dodson, K. W.; Hultgren, S. J.; Earl, A. M. Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in women. Nat. Microbiol 2022, 7 (5), 630– 639, DOI: 10.1038/s41564-022-01107-x49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFyqtrzF&md5=a5458d92f0b40871578f476e448e9ae2Longitudinal multi-omics analyses link gut microbiome dysbiosis with recurrent urinary tract infections in womenWorby, Colin J.; Schreiber IV, Henry L.; Straub, Timothy J.; van Dijk, Lucas R.; Bronson, Ryan A.; Olson, Benjamin S.; Pinkner, Jerome S.; Obernuefemann, Chloe L. P.; Munoz, Vanessa L.; Paharik, Alexandra E.; Azimzadeh, Philippe N.; Walker, Bruce J.; Desjardins, Christopher A.; Chou, Wen-Chi; Bergeron, Karla; Chapman, Sinead B.; Klim, Aleksandra; Manson, Abigail L.; Hannan, Thomas J.; Hooton, Thomas M.; Kau, Andrew L.; Lai, H. Henry; Dodson, Karen W.; Hultgren, Scott J.; Earl, Ashlee M.Nature Microbiology (2022), 7 (5), 630-639CODEN: NMAICH; ISSN:2058-5276. (Nature Portfolio)Abstr.: Recurrent urinary tract infections (rUTIs) are a major health burden worldwide, with history of infection being a significant risk factor. While the gut is a known reservoir for uropathogenic bacteria, the role of the microbiota in rUTI remains unclear. We conducted a year-long study of women with (n = 15) and without (n = 16) history of rUTI, from whom we collected urine, blood and monthly faecal samples for metagenomic and transcriptomic interrogation. During the study 24 UTIs were reported, with addnl. samples collected during and after infection. The gut microbiome of individuals with a history of rUTI was significantly depleted in microbial richness and butyrate-producing bacteria compared with controls, reminiscent of other inflammatory conditions. However, Escherichia coli gut and bladder populations were comparable between cohorts in both relative abundance and phylogroup. Transcriptional anal. of peripheral blood mononuclear cells revealed expression profiles indicative of differential systemic immunity between cohorts. Altogether, these results suggest that rUTI susceptibility is in part mediated through the gut-bladder axis, comprising gut dysbiosis and differential immune response to bacterial bladder colonization, manifesting in symptoms.
- 51Schembri, M. A.; Nhu, N. T. K.; Phan, M. D. Gut-bladder axis in recurrent UTI. Nat. Microbiol 2022, 7 (5), 601– 602, DOI: 10.1038/s41564-022-01113-z50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFyqtr3M&md5=7ff22d3120b3ec5116cfdfc5ad91458bGut-bladder axis in recurrent UTISchembri, Mark A.; Nhu, Nguyen Thi Khanh; Phan, Minh-DuyNature Microbiology (2022), 7 (5), 601-602CODEN: NMAICH; ISSN:2058-5276. (Nature Portfolio)Dysbiosis of the gut microbiome is assocd. with increased susceptibility to recurrent urinary tract infection, defining a gut-bladder axis.
- 52Njoroge, M.; Kaur, G.; Espinoza-Moraga, M.; Wasuna, A.; Dziwornu, G. A.; Seldon, R.; Taylor, D.; Okombo, J.; Warner, D. F.; Chibale, K. Semisynthetic Antimycobacterial C-3 Silicate and C-3/C-21 Ester Derivatives of Fusidic Acid: Pharmacological Evaluation and Stability Studies in Liver Microsomes, Rat Plasma, and Mycobacterium tuberculosis culture. ACS Infect Dis 2019, 5 (9), 1634– 1644, DOI: 10.1021/acsinfecdis.9b0020851https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtlOmsbfP&md5=e1362a952f1eecb77341cb9386a03872Semisynthetic Antimycobacterial C-3 Silicate and C-3/C-21 Ester Derivatives of Fusidic Acid: Pharmacological Evaluation and Stability Studies in Liver Microsomes, Rat Plasma, and Mycobacterium tuberculosis cultureNjoroge, Mathew; Kaur, Gurminder; Espinoza-Moraga, Marlene; Wasuna, Antonina; Dziwornu, Godwin Akpeko; Seldon, Ronnett; Taylor, Dale; Okombo, John; Warner, Digby F.; Chibale, KellyACS Infectious Diseases (2019), 5 (9), 1634-1644CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)Fusidic acid (FA), a natural product fusidane triterpene-based antibiotic with unique structural features, is active in vitro against Mycobacterium tuberculosis, the causative agent of tuberculosis (TB). While possessing good pharmacokinetics in man, FA is rapidly metabolized in rodents, thus complicating proof-of-concept studies in this model. Toward the repositioning of FA as an anti-TB agent, we herein describe the synthesis, activity, and metab. of FA and semisynthesized ester derivs. in rat liver microsomes, rat plasma, and mycobacterial cell culture. FA and deriv. mols. with a free C-3 OH underwent species-specific metab. to the corresponding 3-OH epimer, 3-epifusidic acid (3-epiFA). FA was also metabolized in rat plasma to form FA lactone. These addnl. routes of metab. may contribute to the more rapid clearance of FA obsd. in rodents. C-3 alkyl and aryl esters functioned as classic prodrugs of FA, being hydrolyzed to FA in microsomes, plasma, and Mycobacterium tuberculosis culture. In contrast, C-3 silicate esters and C-21 esters were inert to hydrolysis and so did not act as prodrugs. The antimycobacterial activity of the C-3 silicate esters was comparable to that of FA, and these compds. were stable in microsomes and plasma, identifying them as potential candidates for evaluation in a rodent model of tuberculosis.
- 53Smith, P. W.; Zuccotto, F.; Bates, R. H.; Martinez-Martinez, M. S.; Read, K. D.; Peet, C.; Epemolu, O. Pharmacokinetics of beta-Lactam Antibiotics: Clues from the Past To Help Discover Long-Acting Oral Drugs in the Future. ACS Infect Dis 2018, 4 (10), 1439– 1447, DOI: 10.1021/acsinfecdis.8b0016052https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhsFOrtbfF&md5=ca8591280316b2c1733e911feb5f0150Pharmacokinetics of β-Lactam Antibiotics: Clues from the Past To Help Discover Long-Acting Oral Drugs in the FutureSmith, Paul W.; Zuccotto, Fabio; Bates, Robert H.; Martinez-Martinez, Maria Santos; Read, Kevin D.; Peet, Caroline; Epemolu, OlaACS Infectious Diseases (2018), 4 (10), 1439-1447CODEN: AIDCBC; ISSN:2373-8227. (American Chemical Society)A review. β-Lactams represent perhaps the most important class of antibiotics yet discovered. However, despite many years of active research, none of the currently approved drugs in this class combine oral activity with long duration of action. Recent developments suggest that new β-lactam antibiotics with such a profile would have utility in the treatment of tuberculosis. Consequently, the historical β-lactam pharmacokinetic data have been compiled and analyzed to identify possible directions and drug discovery strategies aimed toward new β-lactam antibiotics with this profile.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acscentsci.2c00598.
Supporting figures and tables, materials and methods, experimental procedures, and characterization of chemical products including LCMS traces and NMR (1H, 13C) spectra (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.