1
|
Dharuman S, Phelps GA, Dunn CM, Wilt LA, Murphy PA, Lee RB, Snoke HE, Selchow P, Haldimann K, Böttger EC, Hobbie SN, Sander P, Lee RE. Synthesis and antibacterial action of 3',6'-disubstituted spectinomycins. J Antibiot (Tokyo) 2024; 77:577-588. [PMID: 38890386 PMCID: PMC11347369 DOI: 10.1038/s41429-024-00750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024]
Abstract
Spectinomycin is an aminocyclitol antibiotic with a unique ribosomal binding site. Prior synthetic modifications of spectinomycin have enhanced potency and antibacterial spectrum through addition at the 6'-position to produce trospectomycin and to the 3'-position to produce spectinamides and aminomethyl spectinomycins. This study focused on the design, synthesis, and evaluation of three 3',6'-disubstituted spectinomycin analogs: trospectinamide, N-benzyl linked aminomethyl, and N-ethylene linked aminomethyl trospectomycins. Computational experiments predicted that these disubstituted analogs would be capable of binding within the SPC ribosomal binding site. The new analogs were synthesized from trospectomycin, adapting the previously established routes for the spectinamide and aminomethyl spectinomycin series. In a cell-free translation assay, the disubstituted analogs showed ribosomal inhibition similar to spectinomycin or trospectomycin. These disubstituted analogs demonstrated inhibitory MIC activity against various bacterial species with the 3'-modification dictating spectrum of activity, leading to improved activity against mycobacterium species. Notably, N-ethylene linked aminomethyl trospectomycins exhibited increased potency against Mycobacterium abscessus and trospectinamide displayed robust activity against M. tuberculosis, aligning with the selective efficacy of spectinamides. The study also found that trospectomycin is susceptible to efflux in M. tuberculosis and M. abscessus. These findings contribute to the understanding of the structure-activity relationship of spectinomycin analogs and can guide the design and synthesis of more effective spectinomycin compounds.
Collapse
Affiliation(s)
- Suresh Dharuman
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
| | - Gregory A Phelps
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38103, USA
| | - Christine M Dunn
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38103, USA
| | - Laura A Wilt
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
| | - Patricia A Murphy
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
| | - Robin B Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
| | - Hannah E Snoke
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38103, USA
| | - Petra Selchow
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, CH-8006, Zurich, Switzerland
| | - Klara Haldimann
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, CH-8006, Zurich, Switzerland
| | - Erik C Böttger
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, CH-8006, Zurich, Switzerland
| | - Sven N Hobbie
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, CH-8006, Zurich, Switzerland
- Division of Clinical Bacteriology and Mycology, University Hospital Basel, Petersgraben 4, CH-4031, Basel, Switzerland
| | - Peter Sander
- Institute of Medical Microbiology, University of Zurich, Gloriastrasse 28/30, CH-8006, Zurich, Switzerland
- National Reference Center for Mycobacteria, Gloriastrasse 28/30, CH-8006, Zurich, Switzerland
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, TN, 38105, USA.
| |
Collapse
|
2
|
Phelps GA, Cheramie MN, Fernando DM, Selchow P, Meyer CJ, Waidyarachchi SL, Dharuman S, Liu J, Meuli M, Molin MD, Killam BY, Murphy PA, Reeve SM, Wilt LA, Anderson SM, Yang L, Lee RB, Temrikar ZH, Lukka PB, Meibohm B, Polikanov YS, Hobbie SN, Böttger EC, Sander P, Lee RE. Development of 2nd generation aminomethyl spectinomycins that overcome native efflux in Mycobacterium abscessus. Proc Natl Acad Sci U S A 2024; 121:e2314101120. [PMID: 38165935 PMCID: PMC10786304 DOI: 10.1073/pnas.2314101120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/11/2023] [Indexed: 01/04/2024] Open
Abstract
Mycobacterium abscessus (Mab), a nontuberculous mycobacterial (NTM) species, is an emerging pathogen with high intrinsic drug resistance. Current standard-of-care therapy results in poor outcomes, demonstrating the urgent need to develop effective antimycobacterial regimens. Through synthetic modification of spectinomycin (SPC), we have identified a distinct structural subclass of N-ethylene linked aminomethyl SPCs (eAmSPCs) that are up to 64-fold more potent against Mab over the parent SPC. Mechanism of action and crystallography studies demonstrate that the eAmSPCs display a mode of ribosomal inhibition consistent with SPC. However, they exert their increased antimicrobial activity through enhanced accumulation, largely by circumventing efflux mechanisms. The N-ethylene linkage within this series plays a critical role in avoiding TetV-mediated efflux, as lead eAmSPC 2593 displays a mere fourfold susceptibility improvement against Mab ΔtetV, in contrast to the 64-fold increase for SPC. Even a minor shortening of the linkage by a single carbon, akin to 1st generation AmSPC 1950, results in a substantial increase in MICs and a 16-fold rise in susceptibility against Mab ΔtetV. These shifts suggest that longer linkages might modify the kinetics of drug expulsion by TetV, ultimately shifting the equilibrium towards heightened intracellular concentrations and enhanced antimicrobial efficacy. Furthermore, lead eAmSPCs were also shown to synergize with various classes of anti-Mab antibiotics and retain activity against clinical isolates and other mycobacterial strains. Encouraging pharmacokinetic profiles coupled with robust efficacy in Mab murine infection models suggest that eAmSPCs hold the potential to be developed into treatments for Mab and other NTM infections.
Collapse
Affiliation(s)
- Gregory A. Phelps
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN38103
| | - Martin N. Cheramie
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Dinesh M. Fernando
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Petra Selchow
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
| | - Christopher J. Meyer
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Samanthi L. Waidyarachchi
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Suresh Dharuman
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Michael Meuli
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
- National Reference Center for Mycobacteria, ZurichCH-8006, Switzerland
| | - Michael Dal Molin
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
| | - Benjamin Y. Killam
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
| | - Patricia A. Murphy
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Stephanie M. Reeve
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Laura A. Wilt
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Shelby M. Anderson
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Robin B. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| | - Zaid H. Temrikar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Pradeep B. Lukka
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN38163
| | - Yury S. Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL60607
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL60607
- Center for Biomolecular Sciences, University of Illinois at Chicago, Chicago, IL60607
| | - Sven N. Hobbie
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
| | - Erik C. Böttger
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
- National Reference Center for Mycobacteria, ZurichCH-8006, Switzerland
| | - Peter Sander
- Institute of Medical Microbiology, University of Zurich, ZurichCH-8006, Switzerland
- National Reference Center for Mycobacteria, ZurichCH-8006, Switzerland
| | - Richard E. Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, Memphis, TN38105
| |
Collapse
|
3
|
Dharuman S, Wilt LA, Liu J, Reeve SM, Thompson CW, Elmore JM, Shcherbakov D, Lee RB, Böttger EC, Lee RE. Synthesis, antibacterial action, and ribosome inhibition of deoxyspectinomycins. J Antibiot (Tokyo) 2021; 74:381-396. [PMID: 33504919 PMCID: PMC8154590 DOI: 10.1038/s41429-021-00408-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 12/13/2020] [Accepted: 01/03/2021] [Indexed: 01/30/2023]
Abstract
Spectinomycin, an aminocyclitol antibiotic, is subject to inactivation by aminoglycoside modifying enzymes (AMEs) through adenylylation or phosphorylation of the 6-hydroxy group position. In this study, the effects of deoxygenation of the 2- and 6-hydroxy group positions on the spectinomycin actinamine ring are probed to evaluate their relationship to ribosomal binding and the antimicrobial activities of spectinomycin, semisynthetic aminomethyl spectinomycins (amSPCs), and spectinamides. To generate these analogs, an improved synthesis of 6-deoxyspectinomycin was developed using the Barton deoxygenation reaction. 6-Dehydrospectinamide was also synthesized from spectinamide 4 to evaluate the H-bond acceptor character on the C-6 position. All the synthesized analogs were tested for antibacterial activity against a panel of Gram (+) and Gram (-) pathogens, plus Mycobacterium tuberculosis. The molecular contribution of the 2- and 6-hydroxy group and the aryl functionalities of all analogs were examined by measuring inhibition of ribosomal translation and molecular dynamics experiments with MM/GBSA analysis. The results of this work indicate that the 6-hydroxy group, which is the primary target of AMEs, is a required motif for antimicrobial activity in current analogs. Removal of the 6-hydroxy group could be partially rescued by offsetting ribosomal binding contributions made by the aryl side chains found in the spectinamide and amSPCs. This study builds on the knowledge of the structure-activity relationships of spectinomycin analogs and is being used to aid the design of next-generation spectinomycins.
Collapse
Affiliation(s)
- Suresh Dharuman
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Laura A Wilt
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiuyu Liu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Stephanie M Reeve
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Carl W Thompson
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John M Elmore
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dimitri Shcherbakov
- Institut für Medizinische Mikrobiologie, Universität Zürich, Zürich, Switzerland
| | - Robin B Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Erik C Böttger
- Institut für Medizinische Mikrobiologie, Universität Zürich, Zürich, Switzerland
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
4
|
Smolarczyk K, Mlynarczyk-Bonikowska B, Rudnicka E, Szukiewicz D, Meczekalski B, Smolarczyk R, Pieta W. The Impact of Selected Bacterial Sexually Transmitted Diseases on Pregnancy and Female Fertility. Int J Mol Sci 2021; 22:2170. [PMID: 33671616 PMCID: PMC7926516 DOI: 10.3390/ijms22042170] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Sexually transmitted infections (STIs) caused by Neisseria gonorrhoeae, Chlamydia trachomatis and Mycoplasma genitalium are a common cause of pelvic inflammatory disease (PID) which can lead to tubal factor infertility (TFI). TFI is one of the most common causes of infertility, accounting for 30% of female fertility problems. STIs can also have an impact on pregnancy, leading to adverse pregnancy outcomes. Escalating antibiotic resistance in Neisseria gonorrhoeae and Mycoplasma genitalium represents a significant problem and can be therapeutically challenging. We present a comprehensive review of the current treatment options, as well as the molecular approach to this subject. We have given special attention to molecular epidemiology, molecular diagnostics, current and new treatments, and drug resistance.
Collapse
MESH Headings
- Anti-Bacterial Agents/pharmacology
- Anti-Bacterial Agents/therapeutic use
- Chlamydia Infections/drug therapy
- Chlamydia Infections/etiology
- Chlamydia Infections/microbiology
- Drug Resistance, Bacterial/drug effects
- Fallopian Tubes/microbiology
- Fallopian Tubes/pathology
- Female
- Gonorrhea/drug therapy
- Gonorrhea/etiology
- Humans
- Infertility, Female/microbiology
- Molecular Diagnostic Techniques
- Molecular Epidemiology/methods
- Mycoplasma Infections/drug therapy
- Mycoplasma Infections/etiology
- Mycoplasma genitalium/pathogenicity
- Pregnancy
- Pregnancy Complications, Infectious/diagnosis
- Pregnancy Complications, Infectious/drug therapy
- Pregnancy Complications, Infectious/epidemiology
- Pregnancy Complications, Infectious/etiology
- Sexually Transmitted Diseases, Bacterial/complications
- Sexually Transmitted Diseases, Bacterial/diagnosis
- Sexually Transmitted Diseases, Bacterial/drug therapy
- Sexually Transmitted Diseases, Bacterial/epidemiology
Collapse
Affiliation(s)
- Katarzyna Smolarczyk
- Department of Dermatology and Venereology, Medical University of Warsaw, 02-008 Warsaw, Poland; (K.S.); (B.M.-B.)
| | - Beata Mlynarczyk-Bonikowska
- Department of Dermatology and Venereology, Medical University of Warsaw, 02-008 Warsaw, Poland; (K.S.); (B.M.-B.)
| | - Ewa Rudnicka
- Department of Gynaecological Endocrinology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Dariusz Szukiewicz
- Department of General and Experimental Pathology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Blazej Meczekalski
- Department of Gynaecological Endocrinology, Poznan University of Medical Sciences, 60-535 Poznan, Poland;
| | - Roman Smolarczyk
- Department of Gynaecological Endocrinology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Wojciech Pieta
- Department of Gynaecological Endocrinology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|
5
|
Zhang L, He J, Bai L, Ruan S, Yang T, Luo Y. Ribosome-targeting antibacterial agents: Advances, challenges, and opportunities. Med Res Rev 2021; 41:1855-1889. [PMID: 33501747 DOI: 10.1002/med.21780] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/08/2020] [Accepted: 12/19/2020] [Indexed: 02/05/2023]
Abstract
Ribosomes, which synthesize proteins, are critical organelles for the survival and growth of bacteria. About 60% of approved antibiotics discovered so far combat pathogenic bacteria by targeting ribosomes. However, several issues, such as drug resistance and toxicity, have impeded the clinical use of ribosome-targeting antibiotics. Moreover, the complexity of the bacteria ribosome structure has retarded the discovery of new ribosome-targeting agents that are considered as the key to the drug-resistance and toxicity. To deal with these challenges, efforts such as medicinal chemistry optimization, combination treatment, and new drug delivery system have been developed. But not enough, the development of structural biology and new screening methods bring powerful tools, such as cryo-electron microscopy technology, advanced computer-aided drug design, and cell-free in vitro transcription/translation systems, for the discovery of novel ribosome-targeting antibiotics. Thus, in this paper, we overview the research on different aspects of bacterial ribosomes, especially focus on discussing the challenges in the discovery of ribosome-targeting antibacterial drugs and advances made to address issues such as drug-resistance and selectivity, which, we believe, provide perspectives for the discovery of novel antibiotics.
Collapse
Affiliation(s)
- Laiying Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Jun He
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Lang Bai
- Center of Infectious Diseases, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China
| | - Shihua Ruan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Tao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China.,Laboratory of Human Diseases and Immunotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China.,Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Abstract
Peptide-chain elongation during protein synthesis entails sequential aminoacyl-tRNA selection and translocation reactions that proceed rapidly (2-20 per second) and with a low error rate (around 10-3 to 10-5 at each step) over thousands of cycles1. The cadence and fidelity of ribosome transit through mRNA templates in discrete codon increments is a paradigm for movement in biological systems that must hold for diverse mRNA and tRNA substrates across domains of life. Here we use single-molecule fluorescence methods to guide the capture of structures of early translocation events on the bacterial ribosome. Our findings reveal that the bacterial GTPase elongation factor G specifically engages spontaneously achieved ribosome conformations while in an active, GTP-bound conformation to unlock and initiate peptidyl-tRNA translocation. These findings suggest that processes intrinsic to the pre-translocation ribosome complex can regulate the rate of protein synthesis, and that energy expenditure is used later in the translocation mechanism than previously proposed.
Collapse
|
7
|
Structural Recognition of Spectinomycin by Resistance Enzyme ANT(9) from Enterococcus faecalis. Antimicrob Agents Chemother 2020; 64:AAC.00371-20. [PMID: 32253216 DOI: 10.1128/aac.00371-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/25/2020] [Indexed: 11/20/2022] Open
Abstract
Spectinomycin is a ribosome-binding antibiotic that blocks the translocation step of translation. A prevalent resistance mechanism is modification of the drug by aminoglycoside nucleotidyl transferase (ANT) enzymes of the spectinomycin-specific ANT(9) family or by enzymes of the dual-specificity ANT(3")(9) family, which also acts on streptomycin. We previously reported the structural mechanism of streptomycin modification by the ANT(3")(9) AadA from Salmonella enterica ANT(9) from Enterococcus faecalis adenylates the 9-hydroxyl of spectinomycin. Here, we present the first structures of spectinomycin bound to an ANT enzyme. Structures were solved for ANT(9) in apo form, in complex with ATP, spectinomycin, and magnesium, or in complex with only spectinomycin. ANT(9) shows an overall structure similar to that of AadA, with an N-terminal nucleotidyltransferase domain and a C-terminal α-helical domain. Spectinomycin binds close to the entrance of the interdomain cleft, while ATP is buried at the bottom. Upon drug binding, the C-terminal domain rotates 14 degrees to close the cleft, allowing contacts of both domains with the drug. Comparison with AadA shows that spectinomycin specificity is explained by a straight α5 helix and a shorter α5-α6 loop, which would clash with the larger streptomycin substrate. In the active site, we observed two magnesium ions, one of them in a previously unobserved position that may activate the 9-hydroxyl for deprotonation by the catalytic base Glu-86. The observed binding mode for spectinomycin suggests that spectinamides and aminomethyl spectinomycins, recent spectinomycin analogues with expansions in position 4 of the C ring, are also subjected to modification by ANT(9) and ANT(3")(9) enzymes.
Collapse
|
8
|
Zhou J, Cui S, He Q, Guo Y, Pan X, Zhang P, Huang N, Ge C, Wang G, Gonzalez FJ, Wang H, Hao H. SUMOylation inhibitors synergize with FXR agonists in combating liver fibrosis. Nat Commun 2020; 11:240. [PMID: 31932588 PMCID: PMC6957516 DOI: 10.1038/s41467-019-14138-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023] Open
Abstract
Farnesoid X receptor (FXR) is a promising target for nonalcoholic steatohepatitis (NASH) and fibrosis. Although various FXR agonists have shown anti-fibrotic effects in diverse preclinical animal models, the response rate and efficacies in clinical trials were not optimum. Here we report that prophylactic but not therapeutic administration of obeticholic acid (OCA) prevents hepatic stellate cell (HSC) activation and fibrogenesis. Activated HSCs show limited response to OCA and other FXR agonists due to enhanced FXR SUMOylation. SUMOylation inhibitors rescue FXR signaling and thereby increasing the efficacy of OCA against HSC activation and fibrosis. FXR upregulates Perilipin-1, a direct target gene of FXR, to stabilize lipid droplets and thereby prevent HSC activation. Therapeutic coadministration of OCA and SUMOylation inhibitors drastically impedes liver fibrosis induced by CCl4, bile duct ligation, and more importantly NASH. In conclusion, we propose a promising therapeutic approach by combining SUMOylation inhibitors and FXR agonists for liver fibrosis.
Collapse
Affiliation(s)
- Jiyu Zhou
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Shuang Cui
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Qingxian He
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Yitong Guo
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Xiaojie Pan
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Pengfei Zhang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Ningning Huang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
| | - Chaoliang Ge
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, 230022, Hefei, China
| | - Guangji Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China.
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hong Wang
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China.
| | - Haiping Hao
- State Key Laboratory of Natural Medicines, Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
9
|
Aminomethyl spectinomycins: a novel antibacterial chemotype for biothreat pathogens. J Antibiot (Tokyo) 2019; 72:693-701. [PMID: 31164713 PMCID: PMC6684479 DOI: 10.1038/s41429-019-0194-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 04/03/2019] [Accepted: 04/22/2019] [Indexed: 01/06/2023]
Abstract
New antibiotics that are active against multi-drug-resistant strains and difficult-to-treat bacterial infections are needed. Synthetic modification of spectinomycin, a bacterial protein synthesis inhibitor, has been shown to increase antibacterial activity compared with spectinomycin. Aminomethyl spectinomycins are active against Gram-negative and Gram-positive bacterial pathogens. In this study, the ability of aminomethyl spectinomycins to treat biothreat pathogens is examined by MIC profiling, synergy testing, and in vivo efficacy experiments. Compound 1950 exhibited potent antibacterial activity against Gram-negative pathogens Brucella spp., Burkholderia mallei, and Francisella tularensis, but showed little to no growth inhibition against Burkholderia pseudomallei, Bacillus anthracis, and Yersinia pestis. Combination testing in checkerboard assays revealed that aminomethyl spectinomycin-antibiotic combinations had mainly an additive effect against the susceptible biodefense pathogens. The in vivo efficacy of compound 1950 was also demonstrated in mice infected with B. mallei (FMH) or F. tularensis (SchuS4). These results suggest that aminomethyl spectinomycins are promising new candidates for development of therapeutics against biodefense bacterial agents.
Collapse
|
10
|
Iverson A, Meyer CJ, Vogel P, Waidyarachchi S, Das N, Bruhn DF, Poole A, Butler MM, Bowlin TL, Lee RE, Rosch JW. Efficacy of Aminomethyl Spectinomycins against Complex Upper Respiratory Tract Bacterial Infections. Antimicrob Agents Chemother 2019; 63:e02096-18. [PMID: 30858215 PMCID: PMC6496058 DOI: 10.1128/aac.02096-18] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 02/17/2019] [Indexed: 11/20/2022] Open
Abstract
The most frequent ailment for which antibiotics are prescribed is otitis media (ear infections), which is most commonly caused by Haemophilus influenzae, Moraxella catarrhalis, and Streptococcus pneumoniae Treatment of otitis media is complicated by the fact that the bacteria in the middle ear typically form biofilms, which can be recalcitrant to antibiotic treatment. Furthermore, bacterial respiratory infections can be greatly exacerbated by viral coinfection, which is particularly evidenced by the synergy between influenza and S. pneumoniae In this study, we sought to ascertain the in vivo efficacy of aminomethyl spectinomycin lead 1950, an effective antibacterial agent both in vitro and in vivo against Streptococcus pneumoniae in the context of complex respiratory infections and acute otitis media. A single dose of 1950 significantly reduced bacterial burden in the respiratory tract for all three pathogens, even when species were present in a coinfection model. Additionally, a single dose of 1950 effectively reduced pneumococcal acute otitis media from the middle ear. The agent 1950 also proved efficacious in the context of influenza-pneumococcal super infection. These data further support the development of this family of compounds as potential therapeutic agents against the common causes of complex upper respiratory tract infections and acute otitis media.
Collapse
Affiliation(s)
- Amy Iverson
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Christopher J Meyer
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Peter Vogel
- Veterinary Pathology Core, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Samanthi Waidyarachchi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Nisha Das
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - David F Bruhn
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Aaron Poole
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Jason W Rosch
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
11
|
Unemo M, Golparian D, Eyre DW. Antimicrobial Resistance in Neisseria gonorrhoeae and Treatment of Gonorrhea. Methods Mol Biol 2019; 1997:37-58. [PMID: 31119616 DOI: 10.1007/978-1-4939-9496-0_3] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gonorrhea and antimicrobial resistance (AMR) in Neisseria gonorrhoeae are major public health concerns globally. Dual antimicrobial therapy (mainly ceftriaxone 250-500 mg × 1 plus azithromycin 1-2 g × 1) is currently recommended in many countries. These dual therapies have high cure rates, have likely been involved in decreasing the level of cephalosporin resistance internationally, and inhibit the spread of AMR gonococcal strains. However, ceftriaxone-resistant strains are currently spreading internationally, predominately associated with travel to Asia. Furthermore, the first global treatment failure with recommended dual therapy was reported in 2016 and the first isolates with combined ceftriaxone resistance and high-level azithromycin resistance were reported in 2018 in the UK and Australia. New antimicrobials for treatment of gonorrhea are essential and, of the few antimicrobials in clinical development, zoliflodacin particularly appears promising. Holistic actions are imperative. These include an enhanced advocacy; prevention, early diagnosis, contact tracing, treatment, test-of-cure, and additional measures for effective management of anogenital and pharyngeal gonorrhea; antimicrobial stewardship; surveillance of infection, AMR and treatment failures; and intensified research, for example, regarding rapid molecular point-of-care detection of gonococci and AMR, novel AMR determinants, new antimicrobials, and an effective gonococcal vaccine, which is the only sustainable solution for management and control of gonorrhea.
Collapse
Affiliation(s)
- Magnus Unemo
- National Reference Laboratory for STIs, WHO Collaborating Centre for Gonorrhoea and Other STIs, Örebro University Hospital, Örebro, Sweden.
| | - Daniel Golparian
- National Reference Laboratory for STIs, WHO Collaborating Centre for Gonorrhoea and Other STIs, Örebro University Hospital, Örebro, Sweden
| | - David W Eyre
- Big Data Institute, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
12
|
Butler MM, Waidyarachchi SL, Connolly KL, Jerse AE, Chai W, Lee RE, Kohlhoff SA, Shinabarger DL, Bowlin TL. Aminomethyl Spectinomycins as Therapeutics for Drug-Resistant Gonorrhea and Chlamydia Coinfections. Antimicrob Agents Chemother 2018; 62:e00325-18. [PMID: 29483122 PMCID: PMC5923137 DOI: 10.1128/aac.00325-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 02/17/2018] [Indexed: 11/20/2022] Open
Abstract
Bacterial sexually transmitted infections are widespread and common, with Neisseria gonorrhoeae (gonorrhea) and Chlamydia trachomatis (chlamydia) being the two most frequent causes. If left untreated, both infections can cause pelvic inflammatory disease, infertility, ectopic pregnancy, and other sequelae. The recommended treatment for gonorrhea is ceftriaxone plus azithromycin (to empirically treat chlamydial coinfections). Antibiotic resistance to all existing therapies has developed in gonorrheal infections. The need for new antibiotics is great, but the pipeline for new drugs is alarmingly small. The aminomethyl spectinomycins, a new class of semisynthetic analogs of the antibiotic spectinomycin, were developed on the basis of a computational analysis of the spectinomycin binding site of the bacterial 30S ribosome and structure-guided synthesis. The compounds display particular potency against common respiratory tract pathogens as well as the sexually transmitted pathogens that cause gonorrhea and chlamydia. Here, we demonstrate the in vitro potencies of several compounds of this class against both bacterial species; the compounds displayed increased potencies against N. gonorrhoeae compared to that of spectinomycin and, significantly, demonstrated activity against C. trachomatis that is not observed with spectinomycin. Efficacies of the compounds were compared to those of spectinomycin and gentamicin in a murine model of infection caused by ceftriaxone/azithromycin-resistant N. gonorrhoeae; the aminomethyl spectinomycins significantly reduced the colonization load and were as potent as the comparator compounds. In summary, data produced by this study support aminomethyl spectinomycins as a promising replacement for spectinomycin and antibiotics such as ceftriaxone for treating drug-resistant gonorrhea, with the added benefit of treating chlamydial coinfections.
Collapse
Affiliation(s)
| | - Samanthi L Waidyarachchi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | - Ann E Jerse
- Uniformed Services University, Bethesda, Maryland, USA
| | - Weirui Chai
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | |
Collapse
|
13
|
Unemo M, Bradshaw CS, Hocking JS, de Vries HJC, Francis SC, Mabey D, Marrazzo JM, Sonder GJB, Schwebke JR, Hoornenborg E, Peeling RW, Philip SS, Low N, Fairley CK. Sexually transmitted infections: challenges ahead. THE LANCET. INFECTIOUS DISEASES 2017; 17:e235-e279. [PMID: 28701272 DOI: 10.1016/s1473-3099(17)30310-9] [Citation(s) in RCA: 484] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/13/2017] [Accepted: 03/30/2017] [Indexed: 12/30/2022]
Abstract
WHO estimated that nearly 1 million people become infected every day with any of four curable sexually transmitted infections (STIs): chlamydia, gonorrhoea, syphilis, and trichomoniasis. Despite their high global incidence, STIs remain a neglected area of research. In this Commission, we have prioritised five areas that represent particular challenges in STI treatment and control. Chlamydia remains the most commonly diagnosed bacterial STI in high-income countries despite widespread testing recommendations, sensitive and specific non-invasive testing techniques, and cheap effective therapy. We discuss the challenges for chlamydia control and evidence to support a shift from the current focus on infection-based screening to improved management of diagnosed cases and of chlamydial morbidity, such as pelvic inflammatory disease. The emergence and spread of antimicrobial resistance in Neisseria gonorrhoeae is globally recognised. We review current and potential future control and treatment strategies, with a focus on novel antimicrobials. Bacterial vaginosis is the most common vaginal disorder in women, but current treatments are associated with frequent recurrence. Recurrence after treatment might relate to evidence that suggests sexual transmission is integral to the pathogenesis of bacterial vaginosis, which has substantial implications for the development of effective management approaches. STIs disproportionately affect low-income and middle-income countries. We review strategies for case management, focusing on point-of-care tests that hold considerable potential for improving STI control. Lastly, STIs in men who have sex with men have increased since the late 1990s. We discuss the contribution of new biomedical HIV prevention strategies and risk compensation. Overall, this Commission aims to enhance the understanding of some of the key challenges facing the field of STIs, and outlines new approaches to improve the clinical management of STIs and public health.
Collapse
Affiliation(s)
- Magnus Unemo
- WHO Collaborating Centre for Gonorrhoea and Other Sexually Transmitted Infections, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Catriona S Bradshaw
- Central Clinical School, Monash University, Melbourne, VIC, Australia; Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia
| | - Jane S Hocking
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, VIC, Australia
| | - Henry J C de Vries
- STI Outpatient Clinic, Public Health Service of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands; Department of Dermatology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Suzanna C Francis
- MRC Tropical Epidemiology Group, London School of Hygiene & Tropical Medicine, London, UK
| | - David Mabey
- Clinical Research Unit, London School of Hygiene & Tropical Medicine, London, UK
| | - Jeanne M Marrazzo
- Department of Medicine, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Gerard J B Sonder
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, Netherlands; Division of Infectious Diseases, Department of Internal Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jane R Schwebke
- Department of Medicine, University of Alabama School of Medicine, Birmingham, AL, USA
| | - Elske Hoornenborg
- STI Outpatient Clinic, Public Health Service of Amsterdam, Amsterdam, Netherlands
| | - Rosanna W Peeling
- Clinical Research Unit, London School of Hygiene & Tropical Medicine, London, UK
| | - Susan S Philip
- Disease Prevention and Control Population Health Division, San Francisco Department of Public Health, San Francisco, CA, USA
| | - Nicola Low
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Christopher K Fairley
- Central Clinical School, Monash University, Melbourne, VIC, Australia; Melbourne Sexual Health Centre, Alfred Health, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Garza I, Wallace MJ, Fernando D, Singh A, Lee RE, Gerding JS, Franklin C, Yendapally R. Synthesis and Evaluation of Thiazolidine Amide and N-Thiazolyl Amide Fluoroquinolone Derivatives. Arch Pharm (Weinheim) 2017; 350. [PMID: 28429393 DOI: 10.1002/ardp.201700029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 11/11/2022]
Abstract
In an effort to develop new fluoroquinolones, we synthesized eight compounds and tested them against a panel of bacteria. The design of these compounds was guided by the introduction of the isothiazoloquinolone motif. The three most active compounds in this series, 8-10, demonstrated good antibacterial activity against methicillin-sensitive Staphylococcus aureus and healthcare-acquired methicillin-resistant Staphylococcus aureus (MIC 0.62-6.3 µg/mL). Further, when these three active compounds were tested for their inhibitory effects on bacterial enzymes, compound 9 was the most effective agent exhibiting IC50 values of 33.9 and 116.5 μM in the S. aureus deoxyribonucleic acid (DNA) gyrase supercoiling and topoisomerase IV decatenation assays, respectively.
Collapse
Affiliation(s)
- Isaac Garza
- University of the Incarnate Word Feik School of Pharmacy, San Antonio, TX, USA
| | - Miranda J Wallace
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Dinesh Fernando
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Aman Singh
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Richard E Lee
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jason S Gerding
- University of the Incarnate Word Feik School of Pharmacy, San Antonio, TX, USA
| | - Cynthia Franklin
- University of the Incarnate Word Feik School of Pharmacy, San Antonio, TX, USA
| | | |
Collapse
|
15
|
Liu J, Bruhn DF, Lee RB, Zheng Z, Janusic T, Scherbakov D, Scherman MS, Boshoff HI, Das S, Rakesh, Waidyarachchi SL, Brewer TA, Gracia B, Yang L, Bollinger J, Robertson GT, Meibohm B, Lenaerts AJ, Ainsa J, Böttger EC, Lee RE. Structure-Activity Relationships of Spectinamide Antituberculosis Agents: A Dissection of Ribosomal Inhibition and Native Efflux Avoidance Contributions. ACS Infect Dis 2017; 3:72-88. [PMID: 28081607 DOI: 10.1021/acsinfecdis.6b00158] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Spectinamides are a novel class of antitubercular agents with the potential to treat drug-resistant tuberculosis infections. Their antitubercular activity is derived from both ribosomal affinity and their ability to overcome intrinsic efflux mediated by the Mycobacterium tuberculosis Rv1258c efflux pump. This study explores the structure-activity relationships through analysis of 50 targeted spectinamides. Compounds are evaluated for ribosomal translational inhibition, MIC activity in Rv1258c efflux pump deficient and wild type tuberculosis strains, and efficacy in an acute model of tuberculosis infection. The results of this study show a narrow structure-activity relationship, consistent with a tight ribosome-binding pocket and strict structural requirements to overcome native efflux. Rationalization of ribosomal inhibition data using molecular dynamics simulations showed stable complex formation for halogenated spectinamides consistent with the long post antibiotic effects observed. The lead spectinamides identified in this study demonstrated potent MIC activity against MDR and XDR tuberculosis and had desirable antitubercular class specific features including low protein binding, low microsomal metabolism, no cytotoxicity, and significant reductions in bacterial burdens in the lungs of mice infected with M. tuberculosis. The structure-activity relationships detailed here emphasize the need to examine efflux-mediated resistance in the design of antituberculosis drugs and demonstrate that it is possible to overcome intrinsic efflux with synthetic modification. The ability to understand the structure requirements for this class has produced a variety of new substituted spectinamides, which may provide useful alternative candidates and promote the further development of this class.
Collapse
Affiliation(s)
- Jiuyu Liu
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - David F. Bruhn
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Robin B. Lee
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Zhong Zheng
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Tanja Janusic
- Institut
für Medizinische Mikrobiologie, Nationales Zentrum für
Mykobakterien, Universität Zürich, Rämistrasse 71, Gloriastrasse
30/32, CH-8006 Zürich, Switzerland
| | - Dimitri Scherbakov
- Institut
für Medizinische Mikrobiologie, Nationales Zentrum für
Mykobakterien, Universität Zürich, Rämistrasse 71, Gloriastrasse
30/32, CH-8006 Zürich, Switzerland
| | - Michael S. Scherman
- Mycobacterial
Research Laboratories, Department of Microbiology, Colorado State University, 1682 Campus
Delivery, Fort Collins, Colorado 80523, United States
| | - Helena I. Boshoff
- Tuberculosis
Research Section, Laboratory of Clinical Infectious Diseases, National
Institute for Allergy and Infectious Disease, National Institutes of Health, 33 North Drive, Bethesda, Maryland 20814, United States
| | - Sourav Das
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Rakesh
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Samanthi L. Waidyarachchi
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Tiffany A. Brewer
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
- Department
of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Begoña Gracia
- Departamento
de Microbiologı́a, Medicina Preventiva y Salud Pública,
and BIFI, Universidad de Zaragoza and CIBER Enfermedades Respiratorias (CIBERES), 50009 Zaragoza, Spain
| | - Lei Yang
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - John Bollinger
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| | - Gregory T. Robertson
- Mycobacterial
Research Laboratories, Department of Microbiology, Colorado State University, 1682 Campus
Delivery, Fort Collins, Colorado 80523, United States
| | - Bernd Meibohm
- Department
of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, Tennessee 38163, United States
| | - Anne J. Lenaerts
- Mycobacterial
Research Laboratories, Department of Microbiology, Colorado State University, 1682 Campus
Delivery, Fort Collins, Colorado 80523, United States
| | - Jose Ainsa
- Departamento
de Microbiologı́a, Medicina Preventiva y Salud Pública,
and BIFI, Universidad de Zaragoza and CIBER Enfermedades Respiratorias (CIBERES), 50009 Zaragoza, Spain
| | - Erik C. Böttger
- Institut
für Medizinische Mikrobiologie, Nationales Zentrum für
Mykobakterien, Universität Zürich, Rämistrasse 71, Gloriastrasse
30/32, CH-8006 Zürich, Switzerland
| | - Richard E. Lee
- Department
of Chemical Biology and Therapeutics, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, MS#1000, Memphis, Tennessee 38105, United States
| |
Collapse
|
16
|
Silverman SM, Moses JE, Sharpless KB. Reengineering Antibiotics to Combat Bacterial Resistance: Click Chemistry [1,2,3]-Triazole Vancomycin Dimers with Potent Activity against MRSA and VRE. Chemistry 2016; 23:79-83. [PMID: 27747932 DOI: 10.1002/chem.201604765] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Indexed: 01/22/2023]
Abstract
Vancomycin has long been considered a drug of last resort. Its efficiency in treating multiple drug-resistant bacterial infections, particularly methicillin-resistant Staphylococcus aureus (MRSA), has had a profound effect on the treatment of life-threatening infections. However, the emergence of resistance to vancomycin is a cause for significant worldwide concern, prompting the urgent development of new effective treatments for antibiotic resistant bacterial infections. Harnessing the benefits of multivalency and cooperativity against vancomycin-resistant strains, we report a Click Chemistry approach towards reengineered vancomycin derivatives and the synthesis of a number of dimers with increased potency against MRSA and vancomycin resistant Enterococci (VRE; VanB). These semi-synthetic dimeric ligands were linked together with great efficiency using the powerful CuAAC reaction, demonstrating high levels of selectivity and purity.
Collapse
Affiliation(s)
- Steven M Silverman
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - John E Moses
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.,School of Chemistry, University of Nottingham, Nottingham, NG7 2RD, UK
| | - K Barry Sharpless
- Department of Chemistry and The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|