51
|
Yang HJ, Wang D, Wen X, Weiner DM, Via LE. One Size Fits All? Not in In Vivo Modeling of Tuberculosis Chemotherapeutics. Front Cell Infect Microbiol 2021; 11:613149. [PMID: 33796474 PMCID: PMC8008060 DOI: 10.3389/fcimb.2021.613149] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Tuberculosis (TB) remains a global health problem despite almost universal efforts to provide patients with highly effective chemotherapy, in part, because many infected individuals are not diagnosed and treated, others do not complete treatment, and a small proportion harbor Mycobacterium tuberculosis (Mtb) strains that have become resistant to drugs in the standard regimen. Development and approval of new drugs for TB have accelerated in the last 10 years, but more drugs are needed due to both Mtb's development of resistance and the desire to shorten therapy to 4 months or less. The drug development process needs predictive animal models that recapitulate the complex pathology and bacterial burden distribution of human disease. The human host response to pulmonary infection with Mtb is granulomatous inflammation usually resulting in contained lesions and limited bacterial replication. In those who develop progressive or active disease, regions of necrosis and cavitation can develop leading to lasting lung damage and possible death. This review describes the major vertebrate animal models used in evaluating compound activity against Mtb and the disease presentation that develops. Each of the models, including the zebrafish, various mice, guinea pigs, rabbits, and non-human primates provides data on number of Mtb bacteria and pathology resolution. The models where individual lesions can be dissected from the tissue or sampled can also provide data on lesion-specific bacterial loads and lesion-specific drug concentrations. With the inclusion of medical imaging, a compound's effect on resolution of pathology within individual lesions and animals can also be determined over time. Incorporation of measurement of drug exposure and drug distribution within animals and their tissues is important for choosing the best compounds to push toward the clinic and to the development of better regimens. We review the practical aspects of each model and the advantages and limitations of each in order to promote choosing a rational combination of them for a compound's development.
Collapse
Affiliation(s)
- Hee-Jeong Yang
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Decheng Wang
- Medical College, China Three Gorges University, Yichang, China.,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
| | - Xin Wen
- Medical College, China Three Gorges University, Yichang, China.,Institute of Infection and Inflammation, China Three Gorges University, Yichang, China
| | - Danielle M Weiner
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.,Tuberculosis Imaging Program, DIR, NIAID, NIH, Bethesda, MD, United States
| | - Laura E Via
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research (DIR), National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States.,Tuberculosis Imaging Program, DIR, NIAID, NIH, Bethesda, MD, United States.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
52
|
Kotzé TJ, Duffy S, Avery VM, Jordaan A, Warner DF, Loots L, Smith GS, Chellan P. Synthesis and antimicrobial study of organoiridium amido-sulfadoxine complexes. Inorganica Chim Acta 2021. [DOI: 10.1016/j.ica.2020.120175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
53
|
Mughal H, Wang H, Zimmerman M, Paradis MD, Freundlich JS. Random Forest Model Prediction of Compound Oral Exposure in the Mouse. ACS Pharmacol Transl Sci 2021; 4:338-343. [PMID: 33615183 DOI: 10.1021/acsptsci.0c00197] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Indexed: 11/29/2022]
Abstract
An early hurdle in the optimization of small-molecule chemical probes and drug discovery entities is the attainment of sufficient exposure in the mouse via oral administration of the compound. While computational approaches have attempted to predict molecular properties related to the mouse pharmacokinetic (PK) profile, we present herein a machine learning approach to specifically predict the oral exposure of a compound as measured in the mouse snapshot PK assay. A random forest workflow was found to produce the best cross-validation and external test set statistics after processing of the input data set and optimization of model features. The modeling approach should be useful to the chemical biology and drug discovery communities to predict this key molecular property and afford chemical entities of translational significance.
Collapse
Affiliation(s)
- Haseeb Mughal
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University - New Jersey Medical School, Newark, New Jersey 07103, United States
| | - Han Wang
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, New Jersey 07110, United States
| | - Marc D Paradis
- Holdings & Ventures, Northwell Health, Manhasset, New York 11030, United States
| | - Joel S Freundlich
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers University - New Jersey Medical School, Newark, New Jersey 07103, United States.,Division of Infectious Disease, Department of Medicine and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University - New Jersey Medical School, Newark, New Jersey 07103, United States
| |
Collapse
|
54
|
Jian Y, Forbes HE, Hulpia F, Risseeuw MDP, Caljon G, Munier-Lehmann H, Boshoff HIM, Van Calenbergh S. 2-((3,5-Dinitrobenzyl)thio)quinazolinones: Potent Antimycobacterial Agents Activated by Deazaflavin (F 420)-Dependent Nitroreductase (Ddn). J Med Chem 2021; 64:440-457. [PMID: 33347317 PMCID: PMC10629625 DOI: 10.1021/acs.jmedchem.0c01374] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Swapping the substituents in positions 2 and 4 of the previously synthesized but yet undisclosed 5-cyano-4-(methylthio)-2-arylpyrimidin-6-ones 4, ring closure, and further optimization led to the identification of the potent antitubercular 2-thio-substituted quinazolinone 26. Structure-activity relationship (SAR) studies indicated a crucial role for both meta-nitro substituents for antitubercular activity, while the introduction of polar substituents on the quinazolinone core allowed reduction of bovine serum albumin (BSA) binding (63c, 63d). While most of the tested quinazolinones exhibited no cytotoxicity against MRC-5, the most potent compound 26 was found to be mutagenic via the Ames test. This analogue exhibited moderate inhibitory potency against Mycobacterium tuberculosis thymidylate kinase, the target of the 3-cyanopyridones that lies at the basis of the current analogues, indicating that the whole-cell antimycobacterial activity of the present S-substituted thioquinazolinones is likely due to modulation of alternative or additional targets. Diminished antimycobacterial activity was observed against mutants affected in cofactor F420 biosynthesis (fbiC), cofactor reduction (fgd), or deazaflavin-dependent nitroreductase activity (rv3547), indicating that reductive activation of the 3,5-dinitrobenzyl analogues is key to antimycobacterial activity.
Collapse
Affiliation(s)
- Yanlin Jian
- Laboratory for Medicinal Chemistry (FFW), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - He Eun Forbes
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Fabian Hulpia
- Laboratory for Medicinal Chemistry (FFW), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Martijn D. P. Risseeuw
- Laboratory for Medicinal Chemistry (FFW), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, University of Antwerp, Universiteitsplein 1 (S7), B-2610 Wilrijk, Belgium
| | - Hélène Munier-Lehmann
- Unit of Chemistry and Biocatalysis, Department of Structural Biology and Chemistry, Institut Pasteur, CNRS UMR3523, 28 Rue du Dr. Roux, Cedex 15 75724 Paris, France
| | - Helena I. M. Boshoff
- Tuberculosis Research Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, United States
| | - Serge Van Calenbergh
- Laboratory for Medicinal Chemistry (FFW), Ghent University, Ottergemsesteenweg 460, B-9000 Gent, Belgium
| |
Collapse
|
55
|
Ernest JP, Strydom N, Wang Q, Zhang N, Nuermberger E, Dartois V, Savic RM. Development of New Tuberculosis Drugs: Translation to Regimen Composition for Drug-Sensitive and Multidrug-Resistant Tuberculosis. Annu Rev Pharmacol Toxicol 2021; 61:495-516. [PMID: 32806997 PMCID: PMC7790895 DOI: 10.1146/annurev-pharmtox-030920-011143] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tuberculosis (TB) kills more people than any other infectious disease. Challenges for developing better treatments include the complex pathology due to within-host immune dynamics, interpatient variability in disease severity and drug pharmacokinetics-pharmacodynamics (PK-PD), and the growing emergence of resistance. Model-informed drug development using quantitative and translational pharmacology has become increasingly recognized as a method capable of drug prioritization and regimen optimization to efficiently progress compounds through TB drug development phases. In this review, we examine translational models and tools, including plasma PK scaling, site-of-disease lesion PK, host-immune and bacteria interplay, combination PK-PD models of multidrug regimens, resistance formation, and integration of data across nonclinical and clinical phases.We propose a workflow that integrates these tools with computational platforms to identify drug combinations that have the potential to accelerate sterilization, reduce relapse rates, and limit the emergence of resistance.
Collapse
Affiliation(s)
- Jacqueline P Ernest
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Natasha Strydom
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Qianwen Wang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Nan Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| | - Eric Nuermberger
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine at Seton Hall University, Nutley, New Jersey 07110, USA
| | - Rada M Savic
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94158, USA;
| |
Collapse
|
56
|
Abstract
Mycobacteria are intrinsically resistant to most antimicrobials, which is generally attributed to the impermeability of their cell wall that considerably limits drug uptake. Moreover, like in other pathogenic bacteria, active efflux systems have been widely characterized from diverse mycobacterial species in laboratory conditions, showing that they can promote resistance by extruding noxious compounds prior to their reaching their intended targets. Therefore, the intracellular concentration of a given compound is determined by the balance between permeability, influx, and efflux.Given the urgent need to discover and develop novel antimycobacterial compounds in order to design effective therapeutic strategies, the contributions to drug resistance made by the controlled permeability of the cell wall and the increased activity of efflux pumps must be determined. In this chapter, we will describe a method that allows (1) the measuring of permeability and the quantification of general efflux activity of mycobacteria, by the study of the transport (influx and efflux) of fluorescent compounds, such as ethidium bromide; and (2) the screening of compounds in search of agents that increase the permeability of the cell wall and efflux inhibitors that could restore the effectiveness of antimicrobials that are subject to efflux.
Collapse
|
57
|
Campaniço A, Harjivan SG, Warner DF, Moreira R, Lopes F. Addressing Latent Tuberculosis: New Advances in Mimicking the Disease, Discovering Key Targets, and Designing Hit Compounds. Int J Mol Sci 2020; 21:ijms21228854. [PMID: 33238468 PMCID: PMC7700174 DOI: 10.3390/ijms21228854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Despite being discovered and isolated more than one hundred years ago, tuberculosis (TB) remains a global public health concern arch. Our inability to eradicate this bacillus is strongly related with the growing resistance, low compliance to current drugs, and the capacity of the bacteria to coexist in a state of asymptomatic latency. This last state can be sustained for years or even decades, waiting for a breach in the immune system to become active again. Furthermore, most current therapies are not efficacious against this state, failing to completely clear the infection. Over the years, a series of experimental methods have been developed to mimic the latent state, currently used in drug discovery, both in vitro and in vivo. Most of these methods focus in one specific latency inducing factor, with only a few taking into consideration the complexity of the granuloma and the genomic and proteomic consequences of each physiological factor. A series of targets specifically involved in latency have been studied over the years with promising scaffolds being discovered and explored. Taking in account that solving the latency problem is one of the keys to eradicate the disease, herein we compile current therapies and diagnosis techniques, methods to mimic latency and new targets and compounds in the pipeline of drug discovery.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Shrika G. Harjivan
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa;
- Department of Pathology, SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Welcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
- Correspondence:
| |
Collapse
|
58
|
Chengalroyen MD, Jordaan A, Seldon R, Ioerger T, Franzblau SG, Nasr M, Warner DF, Mizrahi V. Biological Profiling Enables Rapid Mechanistic Classification of Phenotypic Screening Hits and Identification of KatG Activation-Dependent Pyridine Carboxamide Prodrugs With Activity Against Mycobacterium tuberculosis. Front Cell Infect Microbiol 2020; 10:582416. [PMID: 33282750 PMCID: PMC7691319 DOI: 10.3389/fcimb.2020.582416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/20/2020] [Indexed: 01/22/2023] Open
Abstract
Compounds with novel modes of action are urgently needed to develop effective combination therapies for the treatment of tuberculosis. In this study, a series of compounds was evaluated for activity against replicating Mycobacterium tuberculosis and Vero cell line toxicity. Fourteen of the compounds with in vitro activities in the low micrometer range and a favorable selectivity index were classified using reporter strains of M. tuberculosis which showed that six interfered with cell wall metabolism and one disrupted DNA metabolism. Counter-screening against strains carrying mutations in promiscuous drug targets argued against DprE1 and MmpL3 as hits of any of the cell wall actives and eliminated the cytochrome bc1 complex as a target of any of the compounds. Instead, whole-genome sequencing of spontaneous resistant mutants and/or counter-screening against common isoniazid-resistant mutants of M. tuberculosis revealed that four of the six cell wall-active compounds, all pyridine carboxamide analogues, were metabolized by KatG to form InhA inhibitors. Resistance to two of these compounds was associated with mutations in katG that did not confer cross-resistance to isoniazid. Of the remaining seven compounds, low-level resistance to one was associated with an inactivating mutation in Rv0678, the regulator of the MmpS5-MmpL5 system, which has been implicated in non-specific efflux of multiple chemotypes. Another mapped to the mycothiol-dependent reductase, Rv2466c, suggesting a prodrug mechanism of action in that case. The inability to isolate spontaneous resistant mutants to the seven remaining compounds suggests that they act via mechanisms which have yet to be elucidated.
Collapse
Affiliation(s)
- Melissa D Chengalroyen
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Audrey Jordaan
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Ronnett Seldon
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa.,H3D Drug Discovery and Development Centre, Department of Chemistry, University of Cape Town, Cape Town, South Africa
| | - Thomas Ioerger
- Department of Computer Science and Engineering, Texas A&M University, College Station, TX, United States
| | - Scott G Franzblau
- Institute for Tuberculosis Research, Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, United States
| | - Mohamed Nasr
- Division of AIDS, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| | - Valerie Mizrahi
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Cape Town, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
59
|
Székely R, Rengifo-Gonzalez M, Singh V, Riabova O, Benjak A, Piton J, Cimino M, Kornobis E, Mizrahi V, Johnsson K, Manina G, Makarov V, Cole ST. 6,11-Dioxobenzo[ f]pyrido[1,2- a]indoles Kill Mycobacterium tuberculosis by Targeting Iron-Sulfur Protein Rv0338c (IspQ), A Putative Redox Sensor. ACS Infect Dis 2020; 6:3015-3025. [PMID: 32930569 DOI: 10.1021/acsinfecdis.0c00531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Screening of a diversity-oriented compound library led to the identification of two 6,11-dioxobenzo[f]pyrido[1,2-a]indoles (DBPI) that displayed low micromolar bactericidal activity against the Erdman strain of Mycobacterium tuberculosis in vitro. The activity of these hit compounds was limited to tubercle bacilli, including the nonreplicating form, and to Mycobacterium marinum. On hit expansion and investigation of the structure activity relationship, selected modifications to the dioxo moiety of the DBPI scaffold were either neutral or led to reduction or abolition of antimycobacterial activity. To find the target, DBPI-resistant mutants of M. tuberculosis Erdman were raised and characterized first microbiologically and then by whole genome sequencing. Four different mutations, all affecting highly conserved residues, were uncovered in the essential gene rv0338c (ispQ) that encodes a membrane-bound protein, named IspQ, with 2Fe-2S and 4Fe-4S centers and putative iron-sulfur-binding reductase activity. With the help of a structural model, two of the mutations were localized close to the 2Fe-2S domain in IspQ and another in transmembrane segment 3. The mutant genes were recessive to the wild type in complementation experiments and further confirmation of the hit-target relationship was obtained using a conditional knockdown mutant of rv0338c in M. tuberculosis H37Rv. More mechanistic insight was obtained from transcriptome analysis, following exposure of M. tuberculosis to two different DBPI; this revealed strong upregulation of the redox-sensitive SigK regulon and genes induced by oxidative and thiol-stress. The findings of this investigation pharmacologically validate a novel target in tubercle bacilli and open a new vista for tuberculosis drug discovery.
Collapse
Affiliation(s)
- Rita Székely
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Monica Rengifo-Gonzalez
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Vinayak Singh
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Anzio Road, Observatory 7925, Cape Town 7701, South Africa
| | - Olga Riabova
- FRC Fundamentals of Biotechnology, Russian Academy of Science, 119071 Moscow, Russian Federation
| | - Andrej Benjak
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Jérémie Piton
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Mena Cimino
- Microbial Individuality and Infection, Institut Pasteur, 75015 Paris, France
| | - Etienne Kornobis
- Biomics, C2RT, Institut Pasteur, 75015 Paris, France
- Hub Bioinformatique et Biostatistique, USR 3756 CNRS, Institut Pasteur, 75015 Paris, France
| | - Valerie Mizrahi
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology, University of Cape Town, Anzio Road, Observatory 7925, Cape Town 7701, South Africa
| | - Kai Johnsson
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
| | - Giulia Manina
- Microbial Individuality and Infection, Institut Pasteur, 75015 Paris, France
| | - Vadim Makarov
- FRC Fundamentals of Biotechnology, Russian Academy of Science, 119071 Moscow, Russian Federation
| | - Stewart T. Cole
- Global Health Institute, Ecole Polytechnique Fédérale de Lausanne, CH-1015 Lausanne, Switzerland
- Microbial Individuality and Infection, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
60
|
Pamarthi R, Kumar R, Sankara CS, Lowe GJ, Zuegg J, Singh SK, Ganesh M. α
‐Iodonitroalkenes as Potential Antifungal and Antitubercular Agents. ChemistrySelect 2020. [DOI: 10.1002/slct.202003251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Rambabu Pamarthi
- Department of Chemistry B.M.S. College of Engineering Bull Temple Road Bengaluru Karnataka
- Visveswaraya Technological University Belgaum Karnataka India- 590018
| | - Ram Kumar
- Microbiology Division CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Janakipuram Extension Sitapur Road Lucknow Uttar Pradesh India 226031
| | | | - Gabrielle J. Lowe
- Community for Open antimicrobial Drug Discovery Institute for Molecular Bioscience The University of Queensland 306, Carmody Road St Lucia Qld Australia- 4072
| | - Johannes Zuegg
- Community for Open antimicrobial Drug Discovery Institute for Molecular Bioscience The University of Queensland 306, Carmody Road St Lucia Qld Australia- 4072
| | - Sudheer Kumar Singh
- Microbiology Division CSIR-Central Drug Research Institute, BS-10/1, Sector-10, Janakipuram Extension Sitapur Road Lucknow Uttar Pradesh India 226031
| | - Madhu Ganesh
- Department of Chemistry B.M.S. College of Engineering Bull Temple Road Bengaluru Karnataka
- Department of Pharmaceutical Technology National Institute of Pharmaceutical Education & Research-Hyderabad, Balanagar Telangana India- 500037
| |
Collapse
|
61
|
Dehyab AS, Bakar MFA, AlOmar MK, Sabran SF. A review of medicinal plant of Middle East and North Africa (MENA) region as source in tuberculosis drug discovery. Saudi J Biol Sci 2020; 27:2457-2478. [PMID: 32884430 PMCID: PMC7451596 DOI: 10.1016/j.sjbs.2020.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/05/2020] [Accepted: 07/05/2020] [Indexed: 11/28/2022] Open
Abstract
Tuberculosis (TB) is a disease that affects one-third of the world's population. Although currently available TB drugs have many side effects, such as nausea, headache and gastrointestinal discomfort, no new anti-TB drugs have been produced in the past 30 years. Therefore, the discovery of a new anti-TB agent with minimal or no side effects is urgently needed. Many previous works have reported the effects of medicinal plants against Mycobacterium tuberculosis (MTB). However, none have focused on medicinal plants from the Middle Eastern and North African (MENA) region. This review highlights the effects of medicinal plants from the MENA region on TB. Medicinal plants from the MENA region have been successfully used as traditional medicine and first aid against TB related problems. A total of 184 plants species representing 73 families were studied. Amongst these species, 93 species contained more active compounds with strong anti-MTB activity (crude extracts and/or bioactive compounds with activities of 0-100 µg/ml). The extract of Inula helenium, Khaya senegalensis, Premna odorata and Rosmarinus officinalis presented the strongest anti-MTB activity. In addition, Boswellia papyrifera (Del) Hochst olibanum, Eucalyptus camaldulensis Dehnh leaves (river red gum), Nigella sativa (black cumin) seeds and genus Cymbopogon exhibited anti-TB activity. The most potent bioactive compounds included alantolactone, octyl acetate, 1,8-cineole, thymoquinone, piperitone, α- verbenol, citral b and α-pinene. These compounds affect the permeability of microbial plasma membranes, thus kill the mycobacterium spp. As a conclusion, plant species collected from the MENA region are potential sources of novel drugs against TB.
Collapse
Affiliation(s)
- Ali Sami Dehyab
- Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia (UTHM) – Pagoh Campus, 84600 Muar, Johor, Malaysia
- Department of Medical Laboratory Techniques, Al Maarif University College, Alanbar, Iraq
| | - Mohd Fadzelly Abu Bakar
- Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia (UTHM) – Pagoh Campus, 84600 Muar, Johor, Malaysia
| | | | - Siti Fatimah Sabran
- Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia (UTHM) – Pagoh Campus, 84600 Muar, Johor, Malaysia
| |
Collapse
|
62
|
Baartzes N, Jordaan A, Warner DF, Combrinck J, Taylor D, Chibale K, Smith GS. Antimicrobial evaluation of neutral and cationic iridium(III) and rhodium(III) aminoquinoline-benzimidazole hybrid complexes. Eur J Med Chem 2020; 206:112694. [PMID: 32861176 DOI: 10.1016/j.ejmech.2020.112694] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Abstract
A series of neutral and cationic Ir(III) and Rh(III) aminoquinoline-benzimidazole hybrid complexes were synthesised and their inhibitory activities evaluated against Plasmodium falciparum and Mycobacterium tuberculosis. In general, the hybrid complexes display good activity against the chloroquine-sensitive NF54 strain of P. falciparum. The neutral Ir(III)- and Rh(III)-Cp∗ complexes were the most active (IC50 = 0.488 μM for IrIII), maintaining activity against the multidrug-resistant K1 strain. Low to no cytotoxicity against the Chinese hamster ovarian cell line was observed for the tested complexes. Selected active hybrid complexes demonstrated significant inhibition of β-haematin formation in a cell-free NP-40 assay, suggesting an effect on the host haemoglobin degradation pathway as a potential contributing mechanism of action. When tested against M. tuberculosis H37Rv, most hybrid complexes displayed moderate to good activity. Again, the neutral complexes outperformed the cationic complexes, with the neutral Ir(III)-Cp∗ complexes proving most active (MIC90 = 0.488-1.490 μM).
Collapse
Affiliation(s)
- Nadia Baartzes
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa
| | - Audrey Jordaan
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Clinical Laboratory Sciences, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Clinical Laboratory Sciences, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch, 7701, South Africa
| | - Jill Combrinck
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| | - Dale Taylor
- Drug Discovery and Development Centre (H3D), Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa; South African Medical Research Council, Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
| | - Gregory S Smith
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa.
| |
Collapse
|
63
|
Muliaditan M, Della Pasqua O. Evaluation of pharmacokinetic-pharmacodynamic relationships and selection of drug combinations for tuberculosis. Br J Clin Pharmacol 2020; 87:140-151. [PMID: 32415743 DOI: 10.1111/bcp.14371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 02/07/2020] [Accepted: 04/20/2020] [Indexed: 11/29/2022] Open
Abstract
AIMS Despite evidence of the efficacy of anti-tubercular drug regimens in clinical practice, the rationale underpinning the selection of doses and companion drugs for combination therapy remains empirical. Novel methods are needed to optimise the antibacterial activity in combination therapies. A drug-disease modelling framework for rational selection of dose and drug combinations in tuberculosis is presented here. METHODS A model-based meta-analysis was performed to assess the antibacterial activity of different combinations in infected mice. Data retrieved from the published literature were analysed using a two-state bacterial growth dynamics model, including fast- and slow-growing bacterial populations. The contribution of each drug to the overall antibacterial activity of the combination was parameterised as relative change to the potency of the backbone drug (EC50 -F and/or EC50 -S). Rifampicin and bedaquiline were selected as paradigm drugs to evaluate the predictive performance of the modelling approach. RESULTS Pyrazinamide increased the potency (EC50 -F and EC50 -S) of rifampicin (RZ) and bedaquiline (BZ) by almost two-fold. By contrast, pretomanid and isoniazid were found to worsen the antibacterial activity of BZ and RZ, respectively. Following extrapolation of in vivo pharmacokinetic-pharmacodynamic relationships, the dose of rifampicin showing maximum bactericidal effect in tuberculosis patients was predicted to be 70 mg·kg-1 when given in combination with pyrazinamide. CONCLUSIONS The use of a drug-disease modelling framework may provide a more robust rationale for extrapolation and selection of dose and companion drugs in humans. Our analysis demonstrates that RZ and BZ should be considered as a backbone therapy in prospective novel combination regimens against tuberculosis.
Collapse
Affiliation(s)
- Morris Muliaditan
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK.,Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Uxbridge, UK
| | - Oscar Della Pasqua
- Clinical Pharmacology & Therapeutics Group, University College London, London, UK.,Clinical Pharmacology Modelling and Simulation, GlaxoSmithKline, Uxbridge, UK
| |
Collapse
|
64
|
Yang D, Taylor ZE, Handy S, Li S, Liu J, Stabenow J, Zalduondo L, Jonsson CB, Altman E, Kong Y. Identification of Anti-tuberculosis Compounds From Aurone Analogs. Front Microbiol 2020; 11:1004. [PMID: 32508798 PMCID: PMC7251074 DOI: 10.3389/fmicb.2020.01004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/24/2020] [Indexed: 11/25/2022] Open
Abstract
The emergence of multidrug-resistant Mycobacterium tuberculosis (Mtb) strains has made tuberculosis (TB) control more difficult. Aurone derivatives have demonstrated promising anti-bacterial activities, but their effects against Mtb have not been thoroughly determined. In this study, we aimed to develop anti-TB compounds from aurone analogs. We used a fluorescent protein tdTomato labeled Mtb CDC1551 strain to screen 146 synthesized aurone derivatives for effective anti-TB compounds. The 9504, 9505, 9501, 9510, AA2A, and AA8 aurones inhibited the growth of Mtb with minimal inhibitory concentrations of 6.25, 12.5, 25, 25, 25, and 50 μM, respectively. We also examined cytotoxicities of the six leads against the human liver cell line HepG2, the primate kidney cell line Vero and human monocyte THP-1 derived macrophages. Three of the aurone leads (9504, 9501, and 9510) showed low cytotoxic effects on all three cell lines and high Mtb inhibitory efficacy (selectivity index > 10). Aurone 9504, 9501, AA2A, or AA8 significantly reduced the Mtb load in the lungs of infected mice after a 12-days treatment. We determined that the aurone leads inhibit Mtb chorismate synthase, an essential enzyme for aromatic acid synthesis. Our studies demonstrate the promise of synthetic aurones as novel anti-TB therapeutics.
Collapse
Affiliation(s)
- Dong Yang
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Zachary E Taylor
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN, United States.,Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN, United States
| | - Scott Handy
- Department of Chemistry, Middle Tennessee State University, Murfreesboro, TN, United States.,Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN, United States
| | - Shaoji Li
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jiawang Liu
- Medicinal Chemistry Core, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jennifer Stabenow
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Lillian Zalduondo
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Colleen B Jonsson
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States.,Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Elliot Altman
- Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN, United States.,Department of Biology, Middle Tennessee State University, Murfreesboro, TN, United States
| | - Ying Kong
- Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
65
|
Horn CM, Aucamp J, Smit FJ, Seldon R, Jordaan A, Warner DF, N’Da DD. Synthesis and in vitro antimycobacterial and antileishmanial activities of hydroquinone-triazole hybrids. Med Chem Res 2020. [DOI: 10.1007/s00044-020-02553-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
66
|
Asay BC, Edwards BB, Andrews J, Ramey ME, Richard JD, Podell BK, Gutiérrez JFM, Frank CB, Magunda F, Robertson GT, Lyons M, Ben-Hur A, Lenaerts AJ. Digital Image Analysis of Heterogeneous Tuberculosis Pulmonary Pathology in Non-Clinical Animal Models using Deep Convolutional Neural Networks. Sci Rep 2020; 10:6047. [PMID: 32269234 PMCID: PMC7142129 DOI: 10.1038/s41598-020-62960-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 03/18/2020] [Indexed: 01/28/2023] Open
Abstract
Efforts to develop effective and safe drugs for treatment of tuberculosis require preclinical evaluation in animal models. Alongside efficacy testing of novel therapies, effects on pulmonary pathology and disease progression are monitored by using histopathology images from these infected animals. To compare the severity of disease across treatment cohorts, pathologists have historically assigned a semi-quantitative histopathology score that may be subjective in terms of their training, experience, and personal bias. Manual histopathology therefore has limitations regarding reproducibility between studies and pathologists, potentially masking successful treatments. This report describes a pathologist-assistive software tool that reduces these user limitations, while providing a rapid, quantitative scoring system for digital histopathology image analysis. The software, called 'Lesion Image Recognition and Analysis' (LIRA), employs convolutional neural networks to classify seven different pathology features, including three different lesion types from pulmonary tissues of the C3HeB/FeJ tuberculosis mouse model. LIRA was developed to improve the efficiency of histopathology analysis for mouse tuberculosis infection models, this approach has also broader applications to other disease models and tissues. The full source code and documentation is available from https://Github.com/TB-imaging/LIRA.
Collapse
Affiliation(s)
- Bryce C Asay
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Blake Blue Edwards
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- Department of Computer Science, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jenna Andrews
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Michelle E Ramey
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jameson D Richard
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Brendan K Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Juan F Muñoz Gutiérrez
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Chad B Frank
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Forgivemore Magunda
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Gregory T Robertson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Michael Lyons
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Asa Ben-Hur
- Department of Computer Science, Colorado State University, Fort Collins, Colorado, United States of America
| | - Anne J Lenaerts
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America.
| |
Collapse
|
67
|
Preclinical models to optimize treatment of tuberculous meningitis - A systematic review. Tuberculosis (Edinb) 2020; 122:101924. [PMID: 32501258 DOI: 10.1016/j.tube.2020.101924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/19/2020] [Accepted: 03/20/2020] [Indexed: 01/04/2023]
Abstract
Tuberculous meningitis (TBM) is the most devastating form of TB, resulting in death or neurological disability in up to 50% of patients affected. Treatment is similar to that of pulmonary TB, despite poor cerebrospinal fluid (CSF) penetration of the cornerstone anti-TB drug rifampicin. Considering TBM pathology, it is critical that optimal drug concentrations are reached in the meninges, brain and/or the surrounding CSF. These type of data are difficult to collect in TBM patients. This review aims to identify and describe a preclinical model representative for human TBM which can provide the indispensable data needed for future pharmacological characterization and prioritization of new TBM regimens in the clinical setting. We reviewed existing literature on treatment of TBM in preclinical models: only eight articles, all animal studies, could be identified. None of the animal models completely recapitulated human disease and in most of the animal studies key pharmacokinetic data were missing, making the comparison with human exposure and CNS distribution, and the study of pharmacokinetic-pharmacodynamic relationships impossible. Another 18 articles were identified using other bacteria to induce meningitis with treatment including anti-TB drugs (predominantly rifampicin, moxifloxacin and levofloxacin). Of these articles the pharmacokinetics, i.e. plasma exposure and CSF:plasma ratios, of TB drugs in meningitis could be evaluated. Exposures (except for levofloxacin) agreed with human exposures and also most CSF:plasma ratios agreed with ratios in humans. Considering the lack of an ideal preclinical pharmacological TBM model, we suggest a combination of 1. basic physicochemical drug data combined with 2. in vitro pharmacokinetic and efficacy data, 3. an animal model with adequate pharmacokinetic sampling, microdialysis or imaging of drug distribution, all as a base for 4. physiologically based pharmacokinetic (PBPK) modelling to predict response to TB drugs in treatment of TBM.
Collapse
|
68
|
Model-Informed Drug Discovery and Development Strategy for the Rapid Development of Anti-Tuberculosis Drug Combinations. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10072376] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The increasing emergence of drug-resistant tuberculosis requires new effective and safe drug regimens. However, drug discovery and development are challenging, lengthy and costly. The framework of model-informed drug discovery and development (MID3) is proposed to be applied throughout the preclinical to clinical phases to provide an informative prediction of drug exposure and efficacy in humans in order to select novel anti-tuberculosis drug combinations. The MID3 includes pharmacokinetic-pharmacodynamic and quantitative systems pharmacology models, machine learning and artificial intelligence, which integrates all the available knowledge related to disease and the compounds. A translational in vitro-in vivo link throughout modeling and simulation is crucial to optimize the selection of regimens with the highest probability of receiving approval from regulatory authorities. In vitro-in vivo correlation (IVIVC) and physiologically-based pharmacokinetic modeling provide powerful tools to predict pharmacokinetic drug-drug interactions based on preclinical information. Mechanistic or semi-mechanistic pharmacokinetic-pharmacodynamic models have been successfully applied to predict the clinical exposure-response profile for anti-tuberculosis drugs using preclinical data. Potential pharmacodynamic drug-drug interactions can be predicted from in vitro data through IVIVC and pharmacokinetic-pharmacodynamic modeling accounting for translational factors. It is essential for academic and industrial drug developers to collaborate across disciplines to realize the huge potential of MID3.
Collapse
|
69
|
Zhou B, Shetye G, Yu Y, Santarsiero BD, Klein LL, Abad-Zapatero C, Wolf NM, Cheng J, Jin Y, Lee H, Suh JW, Lee H, Bisson J, McAlpine JB, Chen SN, Cho SH, Franzblau SG, Pauli GF. Antimycobacterial Rufomycin Analogues from Streptomyces atratus Strain MJM3502. JOURNAL OF NATURAL PRODUCTS 2020; 83:657-667. [PMID: 32031795 PMCID: PMC7384767 DOI: 10.1021/acs.jnatprod.9b01095] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study represents a systematic chemical and biological study of the rufomycin (RUF) class of cyclic heptapeptides, which our anti-TB drug discovery efforts have identified as potentially promising anti-TB agents that newly target the caseinolytic protein C1, ClpC1. Eight new RUF analogues, rufomycins NBZ1-NBZ8 (1-8), as well as five known peptides (9-13) were isolated and characterized from the Streptomyces atratus strain MJM3502. Advanced Marfey's and X-ray crystallographic analysis led to the assignment of the absolute configuration of the RUFs. Several isolates exhibited potent activity against both pathogens M. tuberculosis H37Rv and M. abscessus, paired with favorable selectivity (selectivity index >60), which collectively underscores the promise of the rufomycins as potential anti-TB drug leads.
Collapse
Affiliation(s)
- Bin Zhou
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Gauri Shetye
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Yang Yu
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Bernard D. Santarsiero
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Street, Chicago, Illinois 60612, United States
| | - Larry L. Klein
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Cele Abad-Zapatero
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Street, Chicago, Illinois 60612, United States
| | - Nina M. Wolf
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Jinhua Cheng
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do, Republic of Korea
- Division of Bioscience and Bioinformatics, College of Natural Science, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Yingyu Jin
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Hanki Lee
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Joo-Won Suh
- Center for Nutraceutical and Pharmaceutical Materials, Myongji University, Cheoin-gu, Yongin-si, Gyeonggi-do, Republic of Korea
| | - Hyun Lee
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Center for Biomolecular Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Street, Chicago, Illinois 60612, United States
| | - Jonathan Bisson
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - James B. McAlpine
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Shao-Nong Chen
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Sang-Hyun Cho
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Scott G. Franzblau
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| | - Guido F. Pauli
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, Illinois 60612, United States
| |
Collapse
|
70
|
Yam YK, Alvarez N, Go ML, Dick T. Extreme Drug Tolerance of Mycobacterium abscessus "Persisters". Front Microbiol 2020; 11:359. [PMID: 32194537 PMCID: PMC7064438 DOI: 10.3389/fmicb.2020.00359] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/18/2020] [Indexed: 01/25/2023] Open
Abstract
Persistence of infection despite extensive chemotherapy with antibiotics displaying low MICs is a hallmark of lung disease caused by Mycobacterium abscessus (Mab). Thus, the classical MIC assay is a poor predictor of clinical outcome. Discovery of more efficacious antibiotics requires more predictive in vitro potency assays. As a mycobacterium, Mab is an obligate aerobe and a chemo-organo-heterotroph – it requires oxygen and organic carbon sources for growth. However, bacteria growing in patients can encounter micro-environmental conditions that are different from aerated nutrient-rich broth used to grow planktonic cultures for MIC assays. These in vivo conditions may include oxygen and nutrient limitation which should arrest growth. Furthermore, Mab was shown to grow as biofilms in vivo. Here, we show Mab Bamboo, a clinical isolate we use for Mab drug discovery, can survive oxygen deprivation and nutrient starvation for extended periods of time in non-replicating states and developed an in vitro model where the bacterium grows as biofilm. Using these culture models, we show that non-replicating or biofilm-growing bacteria display tolerance to clinically used anti-Mab antibiotics, consistent with the observed persistence of infection in patients. To demonstrate the utility of the developed “persister” assays for drug discovery, we determined the effect of novel agents targeting membrane functions against these physiological forms of the bacterium and find that these compounds show “anti-persister” activity. In conclusion, we developed in vitro “persister” assays to fill an assay gap in Mab drug discovery compound progression and to enable identification of novel lead compounds showing “anti-persister” activity.
Collapse
Affiliation(s)
- Yee-Kuen Yam
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Public Health Research Institute, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Nadine Alvarez
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Mei-Lin Go
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States.,Department of Medical Sciences, Hackensack Meridian School of Medicine at Seton Hall University, Nutley, NJ, United States
| |
Collapse
|
71
|
Zhang N, Strydom N, Tyagi S, Soni H, Tasneen R, Nuermberger EL, Savic RM. Mechanistic Modeling of Mycobacterium tuberculosis Infection in Murine Models for Drug and Vaccine Efficacy Studies. Antimicrob Agents Chemother 2020; 64:e01727-19. [PMID: 31907182 PMCID: PMC7038312 DOI: 10.1128/aac.01727-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB) drug, regimen, and vaccine development rely heavily on preclinical animal experiments, and quantification of bacterial and immune response dynamics is essential for understanding drug and vaccine efficacy. A mechanism-based model was built to describe Mycobacterium tuberculosis H37Rv infection over time in BALB/c and athymic nude mice, which consisted of bacterial replication, bacterial death, and adaptive immune effects. The adaptive immune effect was best described by a sigmoidal function on both bacterial load and incubation time. Applications to demonstrate the utility of this baseline model showed (i) the important influence of the adaptive immune response on pyrazinamide (PZA) drug efficacy, (ii) a persistent adaptive immune effect in mice relapsing after chemotherapy cessation, and (iii) the protective effect of vaccines after M. tuberculosis challenge. These findings demonstrate the utility of our model for describing M. tuberculosis infection and corresponding adaptive immune dynamics for evaluating the efficacy of TB drugs, regimens, and vaccines.
Collapse
Affiliation(s)
- Nan Zhang
- University of California San Francisco, San Francisco, California, USA
| | - Natasha Strydom
- University of California San Francisco, San Francisco, California, USA
| | - Sandeep Tyagi
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Heena Soni
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rokeya Tasneen
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eric L Nuermberger
- Center for Tuberculosis Research, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Rada M Savic
- University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
72
|
Braissant O, Theron G, Friedrich SO, Diacon AH, Bonkat G. Comparison of isothermal microcalorimetry and BACTEC MGIT960 for the detection of the metabolic activity of Mycobacterium tuberculosis in sputum samples. J Appl Microbiol 2019; 128:1497-1502. [PMID: 31834654 DOI: 10.1111/jam.14549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 11/19/2019] [Accepted: 12/08/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION This study explores the uses of microcalorimetry to detect Mycobacterium tuberculosis (TB) in sputum. Microcalorimetry measures metabolic heat evolution during cellular proliferation of tuberculosis (TB) and is considered as a possible alternative to conventional diagnostic tools. OBJECTIVES To compare the time to detection (TTD) from the BACTEC™ MGIT™ 960 and the calScreener™ calorimetric system. METHODS Sixty-four sputa samples were selected from patients with confirmed pulmonary tuberculosis. Those sample were then decontaminated and analysed using calorimetry and BACTEC MGIT 960 system. RESULTS The incubation period until detection of M. tuberculosis in the sample was 8·5 ± 3·7 days for the MGIT system and 10·1 ± 4·1 days (mean ± SD) for calorimetry. CONCLUSIONS The microincubations in the 48-well format calScreener offers potential for rapid and accurate diagnostic of TB in different samples. Although TTD from calorimetry is still longer than with the MGIT, our findings suggest that several improvements are possible. Still, the instrument is ideal for continuous, real-time analysis of net metabolic heat release of limited sample numbers. SIGNIFICANCE AND IMPACT OF THE STUDY Our result emphasizes that with further optimization, calorimetry can become an alternative detection method for tuberculosis.
Collapse
Affiliation(s)
- O Braissant
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland
| | - G Theron
- TASK Applied Science, Bellville, Cape Town, South Africa
| | - S O Friedrich
- TASK Applied Science, Bellville, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, MRC Centre for Tuberculosis Research, Stellenbosch University, Tygerberg, South Africa
| | - A H Diacon
- TASK Applied Science, Bellville, Cape Town, South Africa.,Division of Medical Physiology, Faculty of Medicine and Health Sciences, MRC Centre for Tuberculosis Research, Stellenbosch University, Tygerberg, South Africa
| | - G Bonkat
- Department of Biomedical Engineering, University of Basel, Allschwil, Switzerland.,Alta-Uro AG, Basel, Switzerland
| |
Collapse
|
73
|
Ma S, Jaipalli S, Larkins-Ford J, Lohmiller J, Aldridge BB, Sherman DR, Chandrasekaran S. Transcriptomic Signatures Predict Regulators of Drug Synergy and Clinical Regimen Efficacy against Tuberculosis. mBio 2019; 10:e02627-19. [PMID: 31719182 PMCID: PMC6851285 DOI: 10.1128/mbio.02627-19] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 01/31/2023] Open
Abstract
The rapid spread of multidrug-resistant strains has created a pressing need for new drug regimens to treat tuberculosis (TB), which kills 1.8 million people each year. Identifying new regimens has been challenging due to the slow growth of the pathogen Mycobacterium tuberculosis (MTB), coupled with the large number of possible drug combinations. Here we present a computational model (INDIGO-MTB) that identified synergistic regimens featuring existing and emerging anti-TB drugs after screening in silico more than 1 million potential drug combinations using MTB drug transcriptomic profiles. INDIGO-MTB further predicted the gene Rv1353c as a key transcriptional regulator of multiple drug interactions, and we confirmed experimentally that Rv1353c upregulation reduces the antagonism of the bedaquiline-streptomycin combination. A retrospective analysis of 57 clinical trials of TB regimens using INDIGO-MTB revealed that synergistic combinations were significantly more efficacious than antagonistic combinations (P value = 1 × 10-4) based on the percentage of patients with negative sputum cultures after 8 weeks of treatment. Our study establishes a framework for rapid assessment of TB drug combinations and is also applicable to other bacterial pathogens.IMPORTANCE Multidrug combination therapy is an important strategy for treating tuberculosis, the world's deadliest bacterial infection. Long treatment durations and growing rates of drug resistance have created an urgent need for new approaches to prioritize effective drug regimens. Hence, we developed a computational model called INDIGO-MTB that identifies synergistic drug regimens from an immense set of possible drug combinations using the pathogen response transcriptome elicited by individual drugs. Although the underlying input data for INDIGO-MTB was generated under in vitro broth culture conditions, the predictions from INDIGO-MTB correlated significantly with in vivo drug regimen efficacy from clinical trials. INDIGO-MTB also identified the transcription factor Rv1353c as a regulator of multiple drug interaction outcomes, which could be targeted for rationally enhancing drug synergy.
Collapse
Affiliation(s)
- Shuyi Ma
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Pathobiology Program, Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Suraj Jaipalli
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jonah Larkins-Ford
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Jenny Lohmiller
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Bree B Aldridge
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Biomedical Engineering, Tufts University School of Engineering, Medford, Massachusetts, USA
- Tufts Center for Integrated Management of Antimicrobial Resistance, Tufts University, Boston, Massachusetts, USA
| | - David R Sherman
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Pathobiology Program, Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Sriram Chandrasekaran
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
74
|
Baruah D, Yadav RNS, Yadav A, Das AM. Alpinia nigra fruits mediated synthesis of silver nanoparticles and their antimicrobial and photocatalytic activities. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 201:111649. [PMID: 31710925 DOI: 10.1016/j.jphotobiol.2019.111649] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/11/2019] [Accepted: 10/09/2019] [Indexed: 02/05/2023]
Abstract
In the present systematic study, silver nanoparticles have been synthesized using the fruits of Alpinia nigra. Apart from the presence of saponins, glycosides, alkaloids, steroids, the extract of A. nigra fruits are rich in polyphenols. The Total Flavonoid and Phenol Content of A. nigra fruits extract is 718 mgRE/g extract and 74.9 mgGAE/g extract respectively. The formation of the nanoparticles was validated through characterization techniques like UV-Vis spectroscopy, X- ray diffraction (XRD), X-ray photoelectron spectroscopy (XPS) and Energy dispersive X-ray spectroscopy (EDX). The spherical shape of silver nanoparticles is observed in Transmission Electron Microscopy (TEM) images. The average particle size of the silver nanoparticles is 6 nm. The biomolecules of the fruit extract played the dual role of reducing and capping agents which is evident from Fourier Transform Infrared (FTIR) spectrometer and Scanning Electron Microscopy (SEM) image analysis. The A. nigra capped silver nanoparticles exhibited promising antimicrobial activity against gram negative bacteria Klebsiella pneumoniae, gram positive bacteria Staphylococcus aureus and the pathogenic fungus, Candida albicans. Amongst the three pathogens, Klebsiella pneumoniae is the most susceptible to silver nanoparticles. Furthermore, the nanoparticles efficiently catalysed the degradation of the anthropogenic dyes Methyl orange, Rhodamine B and Orange G in the presence of sunlight. The photocatalytic degradation process follows the pseudo-first order kinetics. These results confirm that the silver nanoparticles can be efficiently synthesized via a green route using A. nigra fruits with applications as antimicrobial and catalytic agents.
Collapse
Affiliation(s)
- Debjani Baruah
- Natural Products Chemistry Group, Chemical Science and Technology Division, CSIR- North East Institute of Science and Technology, Jorhat, Assam 785006, India; Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam 786004, India
| | | | - Archana Yadav
- Biotechnology Group, Biological Science and Technology Division, CSIR- North East Institute of Science and Technology, Jorhat, Assam 785006, India
| | - Archana Moni Das
- Natural Products Chemistry Group, Chemical Science and Technology Division, CSIR- North East Institute of Science and Technology, Jorhat, Assam 785006, India.
| |
Collapse
|
75
|
|
76
|
Safi H, Gopal P, Lingaraju S, Ma S, Levine C, Dartois V, Yee M, Li L, Blanc L, Ho Liang HP, Husain S, Hoque M, Soteropoulos P, Rustad T, Sherman DR, Dick T, Alland D. Phase variation in Mycobacterium tuberculosis glpK produces transiently heritable drug tolerance. Proc Natl Acad Sci U S A 2019; 116:19665-19674. [PMID: 31488707 PMCID: PMC6765255 DOI: 10.1073/pnas.1907631116] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The length and complexity of tuberculosis (TB) therapy, as well as the propensity of Mycobacterium tuberculosis to develop drug resistance, are major barriers to global TB control efforts. M. tuberculosis is known to have the ability to enter into a drug-tolerant state, which may explain many of these impediments to TB treatment. We have identified a mechanism of genetically encoded but rapidly reversible drug tolerance in M. tuberculosis caused by transient frameshift mutations in a homopolymeric tract (HT) of 7 cytosines (7C) in the glpK gene. Inactivating frameshift mutations associated with the 7C HT in glpK produce small colonies that exhibit heritable multidrug increases in minimal inhibitory concentrations and decreases in drug-dependent killing; however, reversion back to a fully drug-susceptible large-colony phenotype occurs rapidly through the introduction of additional insertions or deletions in the same glpK HT region. These reversible frameshift mutations in the 7C HT of M. tuberculosis glpK occur in clinical isolates, accumulate in M. tuberculosis-infected mice with further accumulation during drug treatment, and exhibit a reversible transcriptional profile including induction of dosR and sigH and repression of kstR regulons, similar to that observed in other in vitro models of M. tuberculosis tolerance. These results suggest that GlpK phase variation may contribute to drug tolerance, treatment failure, and relapse in human TB. Drugs effective against phase-variant M. tuberculosis may hasten TB treatment and improve cure rates.
Collapse
Affiliation(s)
- Hassan Safi
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103;
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Pooja Gopal
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Republic of Singapore
| | - Subramanya Lingaraju
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Shuyi Ma
- Center for Infectious Disease, Seattle Children's Hospital, Seattle, WA 98105
| | - Carly Levine
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Veronique Dartois
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Michelle Yee
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore, Republic of Singapore
| | - Liping Li
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Landry Blanc
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Hsin-Pin Ho Liang
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ 07103
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - Seema Husain
- Genomics Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | - Mainul Hoque
- Genomics Center, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| | | | - Tige Rustad
- Center for Infectious Disease, Seattle Children's Hospital, Seattle, WA 98105
| | - David R Sherman
- Center for Infectious Disease, Seattle Children's Hospital, Seattle, WA 98105
| | - Thomas Dick
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110
| | - David Alland
- Center for Emerging Pathogens, New Jersey Medical School, Rutgers University, Newark, NJ 07103;
- Department of Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103
| |
Collapse
|
77
|
Njoroge M, Kaur G, Espinoza-Moraga M, Wasuna A, Dziwornu GA, Seldon R, Taylor D, Okombo J, Warner DF, 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; 5:1634-1644. [PMID: 31309823 DOI: 10.1021/acsinfecdis.9b00208] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
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 metabolism of FA and semisynthesized ester derivatives in rat liver microsomes, rat plasma, and mycobacterial cell culture. FA and derivative molecules with a free C-3 OH underwent species-specific metabolism to the corresponding 3-OH epimer, 3-epifusidic acid (3-epiFA). FA was also metabolized in rat plasma to form FA lactone. These additional routes of metabolism may contribute to the more rapid clearance of FA observed 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 compounds were stable in microsomes and plasma, identifying them as potential candidates for evaluation in a rodent model of tuberculosis.
Collapse
Affiliation(s)
- Mathew Njoroge
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Gurminder Kaur
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Antonina Wasuna
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | | | - Ronnett Seldon
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - Dale Taylor
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
| | - John Okombo
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Rondebosch 7701, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch 7701, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch 7701, South Africa
| |
Collapse
|
78
|
First-Time-in-Human Study and Prediction of Early Bactericidal Activity for GSK3036656, a Potent Leucyl-tRNA Synthetase Inhibitor for Tuberculosis Treatment. Antimicrob Agents Chemother 2019; 63:AAC.00240-19. [PMID: 31182528 PMCID: PMC6658769 DOI: 10.1128/aac.00240-19] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/19/2019] [Indexed: 02/02/2023] Open
Abstract
This first-time-in-human (FTIH) study evaluated the safety, tolerability, pharmacokinetics, and food effect of single and repeat oral doses of GSK3036656, a leucyl-tRNA synthetase inhibitor. In part A, GSK3036656 single doses of 5 mg (fed and fasted), 15 mg, and 25 mg and placebo were administered. In part B, repeat doses of 5 and 15 mg and placebo were administered for 14 days once daily. This first-time-in-human (FTIH) study evaluated the safety, tolerability, pharmacokinetics, and food effect of single and repeat oral doses of GSK3036656, a leucyl-tRNA synthetase inhibitor. In part A, GSK3036656 single doses of 5 mg (fed and fasted), 15 mg, and 25 mg and placebo were administered. In part B, repeat doses of 5 and 15 mg and placebo were administered for 14 days once daily. GSK3036656 showed dose-proportional increase following single-dose administration and after dosing for 14 days. The maximum concentration of drug in serum (Cmax) and area under the concentration-time curve from 0 h to the end of the dosing period (AUC0–τ) showed accumulation with repeated administration of approximately 2- to 3-fold. Pharmacokinetic parameters were not altered in the presence of food. Unchanged GSK3036656 was the only drug-related component detected in plasma and accounted for approximately 90% of drug-related material in urine. Based on total drug-related material detected in urine, the minimum absorbed doses after single (25 mg) and repeat (15 mg) dosing were 50 and 78%, respectively. Unchanged GSK3036656 represented at least 44% and 71% of the 25- and 15-mg doses, respectively. Clinical trial simulations were performed to guide dose escalation during the FTIH study and to predict the GSK3036656 dose range that produces the highest possible early bactericidal activity (EBA0–14) in the prospective phase II trial, with consideration of the predefined exposure limit. GSK3036656 was well tolerated after single and multiple doses, with no reports of serious adverse events. (This study has been registered at ClinicalTrials.gov under identifier NCT03075410.)
Collapse
|
79
|
Campaniço A, Carrasco MP, Njoroge M, Seldon R, Chibale K, Perdigão J, Portugal I, Warner DF, Moreira R, Lopes F. Azaaurones as Potent Antimycobacterial Agents Active against MDR- and XDR-TB. ChemMedChem 2019; 14:1537-1546. [PMID: 31294529 DOI: 10.1002/cmdc.201900289] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 06/19/2019] [Indexed: 12/31/2022]
Abstract
Herein we report the screening of a small library of aurones and their isosteric counterparts, azaaurones and N-acetylazaaurones, against Mycobacterium tuberculosis. Aurones were found to be inactive at 20 μm, whereas azaaurones and N-acetylazaaurones emerged as the most potent compounds, with nine derivatives displaying MIC99 values ranging from 0.4 to 2.0 μm. In addition, several N-acetylazaaurones were found to be active against multidrug-resistant (MDR) and extensively drug-resistant (XDR) clinical M. tuberculosis isolates. The antimycobacterial mechanism of action of these compounds remains to be determined; however, a preliminary mechanistic study confirmed that they do not inhibit the mycobacterial cytochrome bc1 complex. Additionally, microsomal metabolic stability and metabolite identification studies revealed that N-acetylazaaurones are deacetylated to their azaaurone counterparts. Overall, these results demonstrate that azaaurones and their N-acetyl counterparts represent a new entry in the toolbox of chemotypes capable of inhibiting M. tuberculosis growth.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Marta P Carrasco
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Mathew Njoroge
- Division of Clinical Pharmacology, Department of Medicine, Drug Discovery and Development Centre (H3D), University of Cape Town, Observatory, 7925, South Africa
| | - Ronnett Seldon
- Department of Chemistry, South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, 7701, South Africa
| | - Kelly Chibale
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, South Africa.,Department of Chemistry, South African Medical Research Council Drug Discovery and Development Research Unit, University of Cape Town, Rondebosch, 7701, South Africa.,Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
| | - João Perdigão
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Isabel Portugal
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Digby F Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa.,Department of Pathology, SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, University of Cape Town, Rondebosch, 7701, South Africa.,Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch, 7701, South Africa
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| |
Collapse
|
80
|
Mashabela GT, de Wet TJ, Warner DF. Mycobacterium tuberculosis Metabolism. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0067-2019. [PMID: 31350832 PMCID: PMC10957194 DOI: 10.1128/microbiolspec.gpp3-0067-2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Indexed: 02/06/2023] Open
Abstract
Mycobacterium tuberculosis is the cause of tuberculosis (TB), a disease which continues to overwhelm health systems in endemic regions despite the existence of effective combination chemotherapy and the widespread use of a neonatal anti-TB vaccine. For a professional pathogen, M. tuberculosis retains a surprisingly large proportion of the metabolic repertoire found in nonpathogenic mycobacteria with very different lifestyles. Moreover, evidence that additional functions were acquired during the early evolution of the M. tuberculosis complex suggests the organism has adapted (and augmented) the metabolic pathways of its environmental ancestor to persistence and propagation within its obligate human host. A better understanding of M. tuberculosis pathogenicity, however, requires the elucidation of metabolic functions under disease-relevant conditions, a challenge complicated by limited knowledge of the microenvironments occupied and nutrients accessed by bacilli during host infection, as well as the reliance in experimental mycobacteriology on a restricted number of experimental models with variable relevance to clinical disease. Here, we consider M. tuberculosis metabolism within the framework of an intimate host-pathogen coevolution. Focusing on recent advances in our understanding of mycobacterial metabolic function, we highlight unusual adaptations or departures from the better-characterized model intracellular pathogens. We also discuss the impact of these mycobacterial "innovations" on the susceptibility of M. tuberculosis to existing and experimental anti-TB drugs, as well as strategies for targeting metabolic pathways. Finally, we offer some perspectives on the key gaps in the current knowledge of fundamental mycobacterial metabolism and the lessons which might be learned from other systems.
Collapse
Affiliation(s)
- Gabriel T Mashabela
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Current address: Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, University of Stellenbosch, South Africa
| | - Timothy J de Wet
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Department of Integrative Biomedical Sciences, University of Cape Town, South Africa
| | - Digby F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
- Wellcome Centre for Infectious Disease Research in Africa, University of Cape Town, South Africa
| |
Collapse
|
81
|
Baartzes N, Stringer T, Seldon R, Warner DF, Taylor D, Wittlin S, Chibale K, Smith GS. Bioisosteric ferrocenyl aminoquinoline-benzimidazole hybrids: Antimicrobial evaluation and mechanistic insights. Eur J Med Chem 2019; 180:121-133. [PMID: 31301563 DOI: 10.1016/j.ejmech.2019.06.069] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/08/2019] [Accepted: 06/25/2019] [Indexed: 01/29/2023]
Abstract
Phenyl- and bioisosteric ferrocenyl-derived aminoquinoline-benzimidazole hybrid compounds were synthesised and evaluated for their in vitro antiplasmodial activity against the chloroquine-sensitive NF54 and multi-drug resistant K1 strains of the human malaria parasite, Plasmodium falciparum. All compounds were active against the two strains, generally showing enhanced activity in the K1 strain, with resistance indices less than 1. Cytotoxicity studies using Chinese hamster ovarian cells revealed that the hybrids were relatively non-cytotoxic and demonstrated selective killing of the parasite. Based on favourable in vitro antiplasmodial and cytotoxicity data, the most active phenyl (4c) and ferrocenyl (5b) hybrids were tested in vivo against the rodent Plasmodium berghei mouse model. Both compounds caused a reduction in parasitemia relative to the control, with 5c displaying superior activity (92% reduction in parasitemia at 4 × 50 mg/kg oral doses). The most active phenyl and ferrocenyl derivatives showed inhibition of β-haematin formation in a NP-40 detergent-mediated assay, indicating a possible contributing mechanism of antiplasmodial action. The most active ferrocenyl hybrid did not display appreciable reactive oxygen species (ROS) generation in a ROS-induced DNA cleavage gel electrophoresis study. The compounds were also screened for their in vitro activity against Mycobacterium tuberculosis. The hybrids containing a more hydrophobic substituent had enhanced activity (<32.7 μM) compared to those with a less hydrophobic substituent (>62.5 μM).
Collapse
Affiliation(s)
- N Baartzes
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa
| | - T Stringer
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa
| | - R Seldon
- Drug Discovery and Development Centre (H3D), Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa
| | - D F Warner
- SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, DST/NRF Centre of Excellence for Biomedical TB Research, Department of Clinical Laboratory Sciences, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa; Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch, 7701, South Africa
| | - D Taylor
- H3D, Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| | - S Wittlin
- Swiss Tropical and Public Health Institute, Socinstrasse 57, 4002, Basel, Switzerland; University of Basel, 4003, Basel, Switzerland
| | - K Chibale
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa; South African Medical Research Council, Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701, South Africa
| | - G S Smith
- Department of Chemistry, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa.
| |
Collapse
|
82
|
Akhova AV, Tkachenko AG. HPLC–UV method for simultaneous determination of adenosine triphosphate and its metabolites inMycobacterium smegmatis. ACTA CHROMATOGR 2019. [DOI: 10.1556/1326.2017.00344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Anna V. Akhova
- Institute of Ecology and Genetics of Microorganisms, Russian Academy of Sciences, 13, Golev str., Perm 614081, Russia
- Perm National Research Polytechnic University, 29, Komsomolsky Ave., Perm 614990, Russia
| | - Alexander G. Tkachenko
- Institute of Ecology and Genetics of Microorganisms, Russian Academy of Sciences, 13, Golev str., Perm 614081, Russia
- Perm State National Research University, 15, Bukirev str., Perm 614068, Russia
| |
Collapse
|
83
|
Rufomycin Targets ClpC1 Proteolysis in Mycobacterium tuberculosis and M. abscessus. Antimicrob Agents Chemother 2019; 63:AAC.02204-18. [PMID: 30602512 PMCID: PMC6395927 DOI: 10.1128/aac.02204-18] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/14/2018] [Indexed: 11/29/2022] Open
Abstract
ClpC1 is an emerging new target for the treatment of Mycobacterium tuberculosis infections, and several cyclic peptides (ecumicin, cyclomarin A, and lassomycin) are known to act on this target. This study identified another group of peptides, the rufomycins (RUFs), as bactericidal to M. tuberculosis through the inhibition of ClpC1 and subsequent modulation of protein degradation of intracellular proteins. ClpC1 is an emerging new target for the treatment of Mycobacterium tuberculosis infections, and several cyclic peptides (ecumicin, cyclomarin A, and lassomycin) are known to act on this target. This study identified another group of peptides, the rufomycins (RUFs), as bactericidal to M. tuberculosis through the inhibition of ClpC1 and subsequent modulation of protein degradation of intracellular proteins. Rufomycin I (RUFI) was found to be a potent and selective lead compound for both M. tuberculosis (MIC, 0.02 μM) and Mycobacterium abscessus (MIC, 0.4 μM). Spontaneously generated mutants resistant to RUFI involved seven unique single nucleotide polymorphism (SNP) mutations at three distinct codons within the N-terminal domain of clpC1 (V13, H77, and F80). RUFI also significantly decreased the proteolytic capabilities of the ClpC1/P1/P2 complex to degrade casein, while having no significant effect on the ATPase activity of ClpC1. This represents a marked difference from ecumicin, which inhibits ClpC1 proteolysis but stimulates the ATPase activity, thereby providing evidence that although these peptides share ClpC1 as a macromolecular target, their downstream effects are distinct, likely due to differences in binding.
Collapse
|
84
|
Structure-Activity Relationships of Wollamide Cyclic Hexapeptides with Activity against Drug-Resistant and Intracellular Mycobacterium tuberculosis. Antimicrob Agents Chemother 2019; 63:AAC.01773-18. [PMID: 30602509 DOI: 10.1128/aac.01773-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/14/2018] [Indexed: 12/15/2022] Open
Abstract
Wollamides are cyclic hexapeptides, recently isolated from an Australian soil Streptomyces isolate, that exhibit promising in vitro antimycobacterial activity against Mycobacterium bovis Bacille Calmette Guérin without displaying cytotoxicity against a panel of mammalian cells. Here, we report the synthesis and antimycobacterial activity of 36 new synthetic wollamides, collated with all known synthetic and natural wollamides, to reveal structure characteristics responsible for in vitro growth-inhibitory activity against Mycobacterium tuberculosis (H37Rv, H37Ra, CDC1551, HN878, and HN353). The most potent antimycobacterial wollamides were those where residue VI d-Orn (wollamide B) was replaced by d-Arg (wollamide B1) or d-Lys (wollamide B2), with all activity being lost when residue VI was replaced by Gly, l-Arg, or l-Lys (wollamide B3). Substitution of other amino acid residues mainly reduced or ablated antimycobacterial activity. Significantly, whereas wollamide B2 was the most potent in restricting M. tuberculosis in vitro, wollamide B1 restricted M. tuberculosis intracellular burden in infected macrophages. Wollamide B1 synergized with pretomanid (PA-824) in inhibiting M. tuberculosis in vitro growth but did not antagonize prominent first- and second-line tuberculosis antibiotics. Furthermore, wollamide B1 exerted bactericidal activity against nonreplicating M. tuberculosis and impaired growth of multidrug- and extensively drug-resistant clinical isolates. In vivo pharmacokinetic profiles for wollamide B1 in rats and mice encourage further optimization of the wollamide pharmacophore for in vivo bioavailability. Collectively, these observations highlight the potential of the wollamide antimycobacterial pharmacophore.
Collapse
|
85
|
Tanner L, Haynes RK, Wiesner L. An in vitro ADME and in vivo Pharmacokinetic Study of Novel TB-Active Decoquinate Derivatives. Front Pharmacol 2019; 10:120. [PMID: 30833898 PMCID: PMC6387968 DOI: 10.3389/fphar.2019.00120] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/31/2019] [Indexed: 02/03/2023] Open
Abstract
Tuberculosis (TB) is currently the leading cause of mortality due to an infectious disease, despite the existence of multiple effective first-line and second-line drugs. The current anti-TB regimen requires a prolonged treatment period of around 6 months and is only efficacious against drug-sensitive strains of Mycobacterium tuberculosis (Mtb). With a rise in cases of multi-drug resistant and extensively drug resistant strains of Mtb, newer treatments comprising compounds with novel mechanisms of action are required. Although decoquinate (DQ) is inactive against Mtb, its derivatives are of interest to anti-TB drug discovery because of their potential to permeate the mycobacterial cell wall, Mtb-infected macrophages, and granulomatous lesions by passive diffusion. The compounds also display mechanisms of action which are unlike those of currently used quinolones, potentially displaying activity against new targets. Three such derivatives bearing an alkyl group at N-1 and an amide group at C-3 (RMB 041, -043, and -073) displayed potent in vitro activities against Mtb H37Rv (90% minimum inhibitory concentrations, MIC90 = 1.61, 4.18, and 1.88 μM, respectively) and high selectivity indices (10-25). In this study, we evaluated the drug-like properties (in vitro microsomal stability, microsomal/plasma protein binding, kinetic solubility, lipophilicity, and passive permeability) and pharmacokinetic (PK) parameters of these compounds after intravenous and oral administration to male C57BL/6 mice. The compounds showed markedly improved kinetic solubilities compared to that of the parental DQ and were metabolically stable in vitro. The maximum concentrations reached after oral administration were 5.4 ± 0.40, 5.6 ± 1.40, and 2.0 ± 0.03 μM; elimination half-lives were 23.4 ± 2.50, 6.2 ± 0.80, and 11.6 ± 1.30 h; and bioavailabilities were 21.4 ± 1.0, 22.1 ± 2.2, and 5.9 ± 1.3 for RMB041, -043, and -073, respectively. These compounds therefore display promising drug-like properties, and their PK/toxicity profiles (including long half-lives both in vitro and in vivo) support their potential as candidates for further investigation in animal models of Mtb infection.
Collapse
Affiliation(s)
- Lloyd Tanner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Richard K. Haynes
- Centre of Excellence for Pharmaceutical Sciences, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Lubbe Wiesner
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
86
|
Abstract
Tuberculosis (TB) remains a leading cause of death globally among infectious diseases that has killed more numbers of people than any other infectious diseases. Animal models have become the lynchpin for mimicking human infectious diseases. Research on TB could be facilitated by animal challenge models such as the guinea pig, mice, rabbit and non-human primates. No single model presents all aspects of disease pathogenesis due to considerable differences in disease resistance/susceptibility between these models. Availability of a wide range of animal strains, Mycobacterium tuberculosis strains, route of infection and doses affect the disease progression and intervention outcome. Different animal models have contributed significantly to the drug and vaccine development, identification of biomarkers, understanding of TB immunopathogenesis and host genetic influence on infection. In this review, the commonly used animal models in TB research are discussed along with their advantages and limitations.
Collapse
Affiliation(s)
- Amit Kumar Singh
- ICMR-National JALMA Institute of Leprosy & Other Mycobacterial Diseases, Agra, India
| | - Umesh D Gupta
- ICMR-National JALMA Institute of Leprosy & Other Mycobacterial Diseases, Agra, India
| |
Collapse
|
87
|
Graham J, Wong CE, Day J, McFaddin E, Ochsner U, Hoang T, Young CL, Ribble W, DeGroote MA, Jarvis T, Sun X. Discovery of benzothiazole amides as potent antimycobacterial agents. Bioorg Med Chem Lett 2018; 28:3177-3181. [PMID: 30172617 PMCID: PMC6263154 DOI: 10.1016/j.bmcl.2018.08.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 11/17/2022]
Abstract
From a high throughput screening of commercially available libraries against nontuberculous mycobacteria and Mycobacterium tuberculosis, numerous hits were identified with moderate activity. Extensive medicinal chemistry optimization has led to a series of potent benzothiazole amide antimycobacterial agents. Replacement of the adamantyl group with cyclohexyl derivatives and further development of this series resulted in an advanced lead compound, CRS400393, which demonstrated excellent potency and a mycobacteria-specific spectrum of activity. MIC values ranged from 0.03 to 0.12 μg/mL against Mycobacterium abscessus and other rapid-grower NTM, and 1-2 μg/mL against Mycobacterium avium complex. The preliminary mechanism of action studies suggested these agents may target MmpL3, a mycobacterial mycolic acid transporter. The series has demonstrated in vivo efficacy in a proof of concept mouse model of M. abscessus infection.
Collapse
Affiliation(s)
- James Graham
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Christina E Wong
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Joshua Day
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | | | - Urs Ochsner
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Teresa Hoang
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Casey L Young
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Wendy Ribble
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Mary A DeGroote
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Thale Jarvis
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA
| | - Xicheng Sun
- Crestone, Inc, 6075 Longbow Dr. Suite 130, Boulder, CO 80301, USA.
| |
Collapse
|
88
|
Hussain A, Rather MA, Bhat ZS, Majeed A, Maqbool M, Shah AM, Aga MA, Shah A, Mushtaq S, Sangwan PL, Hassan QP, Ahmad Z. In vitro evaluation of dinactin, a potent microbial metabolite against Mycobacterium tuberculosis. Int J Antimicrob Agents 2018; 53:49-53. [PMID: 30267759 DOI: 10.1016/j.ijantimicag.2018.09.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/17/2018] [Accepted: 09/22/2018] [Indexed: 12/24/2022]
Abstract
Current long duration treatment options and the emergence of drug resistance in tuberculosis (TB) have led to renewed interest in discovery of novel anti-tubercular agents or the scaffolds exhibiting enhanced efficacy with current anti-TB drugs. Herein, dinactin, a potent bioactive macrotetrolide isolated from Streptomyces puniceus AS13, was evaluated against Mycobacterium tuberculosis H37Rv and other susceptible and drug-resistant clinical isolates of M. tuberculosis. In vitro pharmacological assays showed that dinactin is bactericidal against laboratory standard strain M. tuberculosis H37Rv (minimum inhibitory concentration [MIC] 1 µg/mL and minimum bactericidal concentration [MBC] 4 µg/mL). Dinactin also retained its activity against various clinical isolates, including multidrug-resistant strains of M. tuberculosis. Whole cell interaction assays with standard first- and second-line anti-TB drugs showed the synergistic interaction of dinactin with rifampicin or amikacin, reflecting its suitability for use in combination regimens. The killing kinetics studies of dinactin against M. tuberculosis H37Rv revealed that it has strong concentration-dependent anti-TB activity that is also dependent on time. The kill curve also showed dynamic killing capacity of dinactin as it exhibited bactericidal potential at all concentrations tested. Kill curve data demonstrated that dinactin, like isoniazid, exerts its strong tuberculocidal activity within the first two days of exposure. This evidence strongly supports further evaluation of dinactin as a new option in the treatment of TB.
Collapse
Affiliation(s)
- Aehtesham Hussain
- Microbial Biotechnology Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India; Academy of Scientific and Innovative Research, CSIR - Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India
| | - Muzafar Ahmad Rather
- Clinical Microbiology and PK/PD Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Zubair Shanib Bhat
- Academy of Scientific and Innovative Research, CSIR - Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India; Clinical Microbiology and PK/PD Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Aasif Majeed
- Microbial Biotechnology Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Mubashir Maqbool
- Clinical Microbiology and PK/PD Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Aabid Manzoor Shah
- Microbial Biotechnology Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Mushtaq A Aga
- Bio-organic chemistry division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Aiyatullah Shah
- Microbial Biotechnology Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Saleem Mushtaq
- Microbial Biotechnology Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India
| | - Payare L Sangwan
- Bio-organic chemistry division, CSIR - Indian Institute of Integrative Medicine, Jammu Tawi, 180001, India
| | - Qazi Parvaiz Hassan
- Microbial Biotechnology Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India; Academy of Scientific and Innovative Research, CSIR - Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India.
| | - Zahoor Ahmad
- Academy of Scientific and Innovative Research, CSIR - Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India; Clinical Microbiology and PK/PD Division, CSIR - Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, Jammu & Kashmir 190005, India.
| |
Collapse
|
89
|
De Groote MA, Jarvis TC, Wong C, Graham J, Hoang T, Young CL, Ribble W, Day J, Li W, Jackson M, Gonzalez-Juarrero M, Sun X, Ochsner UA. Optimization and Lead Selection of Benzothiazole Amide Analogs Toward a Novel Antimycobacterial Agent. Front Microbiol 2018; 9:2231. [PMID: 30294313 PMCID: PMC6158578 DOI: 10.3389/fmicb.2018.02231] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
Mycobacteria remain an important problem worldwide, especially drug resistant human pathogens. Novel therapeutics are urgently needed to tackle both drug-resistant tuberculosis (TB) and difficult-to-treat infections with nontuberculous mycobacteria (NTM). Benzothiazole adamantyl amide had previously emerged as a high throughput screening hit against M. tuberculosis (Mtb) and was subsequently found to be active against NTM as well. For lead optimization, we applied an iterative process of design, synthesis and screening of several 100 analogs to improve antibacterial potency as well as physicochemical and pharmacological properties to ultimately achieve efficacy. Replacement of the adamantyl group with cyclohexyl derivatives, including bicyclic moieties, resulted in advanced lead compounds that showed excellent potency and a mycobacteria-specific spectrum of activity. MIC values ranged from 0.03 to 0.12 μg/mL against M. abscessus (Mabs) and other rapid- growing NTM, 1–2 μg/mL against M. avium complex (MAC), and 0.12–0.5 μg/mL against Mtb. No pre-existing resistance was found in a collection of n = 54 clinical isolates of rapid-growing NTM. Unlike many antibacterial agents commonly used to treat mycobacterial infections, benzothiazole amides demonstrated bactericidal effects against both Mtb and Mabs. Metabolic labeling provided evidence that the compounds affect the transfer of mycolic acids to their cell envelope acceptors in mycobacteria. Mapping of resistance mutations pointed to the trehalose monomycolate transporter (MmpL3) as the most likely target. In vivo efficacy and tolerability of a benzothiazole amide was demonstrated in a mouse model of chronic NTM lung infection with Mabs. Once daily dosing over 4 weeks by intrapulmonary microspray administration as 5% corn oil/saline emulsion achieved statistically significant CFU reductions compared to vehicle control and non-inferiority compared to azithromycin. The benzothiazole amides hold promise for development of a novel therapeutic agent with broad antimycobacterial activity, though further work is needed to develop drug formulations for direct intrapulmonary delivery via aerosol.
Collapse
Affiliation(s)
- Mary A De Groote
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | | | | | | | | | | | | | - Joshua Day
- Crestone, Inc., Boulder, CO, United States
| | - Wei Li
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | | | | |
Collapse
|
90
|
Santos K, Lukka PB, Grzegorzewicz A, Jackson M, Trivedi A, Pavan F, Chorilli M, Braunstein M, Hickey A, Meibohm B, Gonzalez-Juarrero M. Primary Lung Dendritic Cell Cultures to Assess Efficacy of Spectinamide-1599 Against Intracellular Mycobacterium tuberculosis. Front Microbiol 2018; 9:1895. [PMID: 30186246 PMCID: PMC6110900 DOI: 10.3389/fmicb.2018.01895] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/27/2018] [Indexed: 01/12/2023] Open
Abstract
There is an urgent need to treat tuberculosis (TB) quickly, effectively and without side effects. Mycobacterium tuberculosis (Mtb), the causative organism of TB, can survive for long periods of time within macrophages and dendritic cells and these intracellular bacilli are difficult to eliminate with current drug regimens. It is well established that Mtb responds differentially to drug treatment depending on its extracellular and intracellular location and replicative state. In this study, we isolated and cultured lung derived dendritic cells to be used as a screening system for drug efficacy against intracellular mycobacteria. Using mono- or combination drug treatments, we studied the action of spectinamide-1599 and pyrazinamide (antibiotics targeting slow-growing bacilli) in killing bacilli located within lung derived dendritic cells. Furthermore, because IFN-γ is an essential cytokine produced in response to Mtb infection and present during TB chemotherapy, we also assessed the efficacy of these drugs in the presence and absence of IFN-γ. Our results demonstrated that monotherapy with either spectinamide-1599 or pyrazinamide can reduce the intracellular bacterial burden by more than 99.9%. Even more impressive is that when TB infected lung derived dendritic cells are treated with spectinamide-1599 and pyrazinamide in combination with IFN-γ a strong synergistic effect was observed, which reduced the intracellular burden below the limit of detection. We concluded that IFN-γ activation of lung derived dendritic cells is essential for synergy between spectinamide-1599 and pyrazinamide.
Collapse
Affiliation(s)
- Karen Santos
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO, United States.,School of Pharmaceutical Sciences, São Paulo State University, São Paulo, Brazil
| | - Pradeep B Lukka
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Anne Grzegorzewicz
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO, United States
| | - Mary Jackson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO, United States
| | - Ashit Trivedi
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Fernando Pavan
- School of Pharmaceutical Sciences, São Paulo State University, São Paulo, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University, São Paulo, Brazil
| | - Miriam Braunstein
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Anthony Hickey
- Discovery Science and Technology, RTI International, Durham, NC, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology & Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
91
|
Gumbo M, Beteck RM, Mandizvo T, Seldon R, Warner DF, Hoppe HC, Isaacs M, Laming D, Tam CC, Cheng LW, Liu N, Land KM, Khanye SD. Cinnamoyl-Oxaborole Amides: Synthesis and Their in Vitro Biological Activity. Molecules 2018; 23:E2038. [PMID: 30111695 PMCID: PMC6222898 DOI: 10.3390/molecules23082038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 08/06/2018] [Accepted: 08/07/2018] [Indexed: 11/19/2022] Open
Abstract
Due to the increased interest in their application in the treatment of infectious diseases, boron-containing compounds have received a significant coverage in the literature. Herein, a small set of novel cinnamoly-oxaborole amides were synthesized and screened against nagana Trypanosoma brucei brucei for antitrypanosomal activity. Compound 5g emerged as a new hit with an in vitro IC50 value of 0.086 μM against T. b. brucei without obvious inhibitory activity against HeLa cell lines. The same series was also screened against other human pathogens, including Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), for which moderate to weak activity (10 to >125 μM) was observed. Similarly, these compounds exhibited moderate activity against the human protozoal pathogen Trichomonas vaginalis with no observed effect on common microbiome bacterial species. The cross-species inhibitory activity presents the possibility of these compounds serving as broad-spectrum antibiotics for these prevalent three human pathogens.
Collapse
Affiliation(s)
- Maureen Gumbo
- Faculty of Science, Department of Chemistry, Rhodes University, Grahamstown 6140, South Africa.
| | - Richard M Beteck
- Faculty of Science, Department of Chemistry, Rhodes University, Grahamstown 6140, South Africa.
| | - Tawanda Mandizvo
- Faculty of Science, Department of Chemistry, Rhodes University, Grahamstown 6140, South Africa.
| | - Ronnett Seldon
- Drug Discovery and Development Centre (H3-D), Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa.
| | - Digby F Warner
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit, Department of Pathology, University of Cape Town, Rondebosch 7701, South Africa.
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa.
| | - Heinrich C Hoppe
- Faculty of Science, Department of Biochemistry and Microbiology, Rhodes University, Grahamstown 6140, South Africa.
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| | - Michelle Isaacs
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| | - Dustin Laming
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| | - Christina C Tam
- Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA.
| | - Luisa W Cheng
- Foodborne Toxin Detection and Prevention Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, CA 94710, USA.
| | - Nicole Liu
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA.
| | - Kirkwood M Land
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA.
| | - Setshaba D Khanye
- Faculty of Science, Department of Chemistry, Rhodes University, Grahamstown 6140, South Africa.
- Centre for Chemico- and Biomedicinal Research, Rhodes University, Grahamstown 6140, South Africa.
| |
Collapse
|
92
|
Perryman AL, Patel JS, Russo R, Singleton E, Connell N, Ekins S, Freundlich JS. Naïve Bayesian Models for Vero Cell Cytotoxicity. Pharm Res 2018; 35:170. [PMID: 29959603 DOI: 10.1007/s11095-018-2439-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 06/05/2018] [Indexed: 11/30/2022]
Abstract
PURPOSE To advance translational research of potential therapeutic small molecules against infectious microbes, the compounds must display a relative lack of mammalian cell cytotoxicity. Vero cell cytotoxicity (CC50) is a common initial assay for this metric. We explored the development of naïve Bayesian models that can enhance the probability of identifying non-cytotoxic compounds. METHODS Vero cell cytotoxicity assays were identified in PubChem, reformatted, and curated to create a training set with 8741 unique small molecules. These data were used to develop Bayesian classifiers, which were assessed with internal cross-validation, external tests with a set of 193 compounds from our laboratory, and independent validation with an additional diverse set of 1609 unique compounds from PubChem. RESULTS Evaluation with independent, external test and validation sets indicated that cytotoxicity Bayesian models constructed with the ECFP_6 descriptor were more accurate than those that used FCFP_6 fingerprints. The best cytotoxicity Bayesian model displayed predictive power in external evaluations, according to conventional and chance-corrected statistics, as well as enrichment factors. CONCLUSIONS The results from external tests demonstrate that our novel cytotoxicity Bayesian model displays sufficient predictive power to help guide translational research. To assist the chemical tool and drug discovery communities, our curated training set is being distributed as part of the Supplementary Material. Graphical Abstract Naive Bayesian models have been trained with publically available data and offer a useful tool for chemical biology and drug discovery to select for small molecules with a high probability of exhibiting acceptably low Vero cell cytotoxicity.
Collapse
Affiliation(s)
- Alexander L Perryman
- Department of Pharmacology, Physiology and Neuroscience, and Medicine, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Jimmy S Patel
- Department of Pharmacology, Physiology and Neuroscience, and Medicine, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Riccardo Russo
- Division of Infectious Diseases, Department of Medicine, and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Eric Singleton
- Division of Infectious Diseases, Department of Medicine, and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Nancy Connell
- Division of Infectious Diseases, Department of Medicine, and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., Main Campus Drive Lab 3510, Raleigh, North Carolina,, 27606, USA
| | - Joel S Freundlich
- Department of Pharmacology, Physiology and Neuroscience, and Medicine, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA. .,Division of Infectious Diseases, Department of Medicine, and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave, Newark, NJ, 07103, USA.
| |
Collapse
|
93
|
Antonova AV, Gryadunov DA, Zimenkov DV. Molecular Mechanisms of Drug Tolerance in Mycobacterium tuberculosis. Mol Biol 2018. [DOI: 10.1134/s0026893318030020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
94
|
Sng LH, Peh JWL, Kee MTL, Ya'akob NBM, Ong RTH, Wong CW, Chee CBE, Wang YT. Clofazimine drug susceptibility testing for Mycobacterium tuberculosis: the case of using the right diluent. Pathology 2018; 50:549-553. [PMID: 29891191 DOI: 10.1016/j.pathol.2018.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 01/05/2018] [Accepted: 01/22/2018] [Indexed: 11/19/2022]
Abstract
Accurate and reliable drug susceptibility testing (DST) is essential for the effective treatment and control of tuberculosis. With the increase in drug-resistant organisms, newer and less conventional antimicrobial agents are used for treatment. Recently, we found an unprecedented rise in the number of clofazimine-resistant Mycobacterium tuberculosis isolates in our laboratory. An investigation found that this phenomenon was due to a change in the method of drug preparation. We performed studies to assess the impact of water and dimethyl sulfoxide (DMSO) as a final diluent for clofazimine drug testing. Based on our findings, the use of DMSO as a solvent for M. tuberculosis DST was optimised using the BACTEC MGIT 960 platform.
Collapse
Affiliation(s)
- Li-Hwei Sng
- Central Tuberculosis Laboratory, Singapore General Hospital, Singapore.
| | | | | | | | - Rick Twee-Hee Ong
- Saw Swee Hock School of Public Health, National University Health Singapore, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
95
|
Chacko S, Boshoff HIM, Singh V, Ferraris DM, Gollapalli DR, Zhang M, Lawson AP, Pepi MJ, Joachimiak A, Rizzi M, Mizrahi V, Cuny GD, Hedstrom L. Expanding Benzoxazole-Based Inosine 5'-Monophosphate Dehydrogenase (IMPDH) Inhibitor Structure-Activity As Potential Antituberculosis Agents. J Med Chem 2018; 61:4739-4756. [PMID: 29746130 DOI: 10.1021/acs.jmedchem.7b01839] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
New drugs and molecular targets are urgently needed to address the emergence and spread of drug-resistant tuberculosis. Mycobacterium tuberculosis ( Mtb) inosine 5'-monophosphate dehydrogenase 2 ( MtbIMPDH2) is a promising yet controversial potential target. The inhibition of MtbIMPDH2 blocks the biosynthesis of guanine nucleotides, but high concentrations of guanine can potentially rescue the bacteria. Herein we describe an expansion of the structure-activity relationship (SAR) for the benzoxazole series of MtbIMPDH2 inhibitors and demonstrate that minimum inhibitory concentrations (MIC) of ≤1 μM can be achieved. The antibacterial activity of the most promising compound, 17b (Q151), is derived from the inhibition of MtbIMPDH2 as demonstrated by conditional knockdown and resistant strains. Importantly, guanine does not change the MIC of 17b, alleviating the concern that guanine salvage can protect Mtb in vivo. These findings suggest that MtbIMPDH2 is a vulnerable target for tuberculosis.
Collapse
Affiliation(s)
| | - Helena I M Boshoff
- Tuberculosis Research Section , National Institute of Allergy and Infectious Diseases , Bethesda , Maryland 20892 , United States
| | - Vinayak Singh
- Department of Drug Discovery and Development & Institute of Infectious Disease and Molecular Medicine , H3D Drug Discovery and Development Centre, University of Cape Town , Rondebosch , Cape Town 7701 , South Africa
| | - Davide M Ferraris
- Dipartimento di Scienze del Farmaco , Universitá del Piemonte Orientale , Via Bovio 6 , 28100 Novara , Italy
| | | | | | | | | | - Andrzej Joachimiak
- Center for Structural Genomics of Infectious Diseases and Department of Biochemistry and Molecular Biology , University of Chicago , Chicago , Illinois 60557 , United States.,Structural Biology Center, Biosciences , Argonne National Laboratory , 9700 S. Cass Avenue, Argonne , Illinois 60439 , United States
| | - Menico Rizzi
- Dipartimento di Scienze del Farmaco , Universitá del Piemonte Orientale , Via Bovio 6 , 28100 Novara , Italy
| | - Valerie Mizrahi
- MRC/NHLS/UCT Molecular Mycobacteriology Research Unit & DST/NRF Centre of Excellence for Biomedical TB Research, Institute of Infectious Disease and Molecular Medicine & Department of Pathology , University of Cape Town , Anzio Road , Observatory 7925 , South Africa
| | - Gregory D Cuny
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy , University of Houston , Health Building 2, 4849 Calhoun Road , Houston , Texas 77204 , United States
| | | |
Collapse
|
96
|
Diop EA, Queiroz EF, Kicka S, Rudaz S, Diop T, Soldati T, Wolfender JL. Survey on medicinal plants traditionally used in Senegal for the treatment of tuberculosis (TB) and assessment of their antimycobacterial activity. JOURNAL OF ETHNOPHARMACOLOGY 2018; 216:71-78. [PMID: 29289797 DOI: 10.1016/j.jep.2017.12.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 12/21/2017] [Accepted: 12/23/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In West Africa, populations are used to taking traditional medicine as a first aid against common health problems. In this aspect, many plants are claimed to be effective in the treatment of Tuberculosis (TB), which according to the World Health Organization (WHO) remains one of the world's deadliest communicable diseases. AIM OF THE STUDY The main aim of this study was to identify plants used to treat TB-symptoms by the population of Senegal and to evaluate their possible concomitant use with clinically approved TB-drugs. This approach allowed the selection of plants effectively used in traditional medicine. In order to verify if the usage of some of these plants can be rationalized, the activity of their traditional preparations was assessed with both an intracellular and extracellular antimycobacterial host-pathogen assays. MATERIALS AND METHODS An ethnopharmacological survey conducted on 117 TB-patients and 30 healers in Senegal from March to May 2014. The questionnaires were focused on the use of medicinal plants to treat common TB -symptoms (cough longer than 2 weeks, fever, night sweats, weight loss and bloody sputum). Local plant names, utilized organs (herbal drugs) and traditional formulations of the plants were recorded. Extracts were prepared by mimicking the traditional decoction in boiling water and screened for their antimycobacterial activity using Mycobacterium marinum, as a validated TB surrogate, and an Acanthamoeba castellanii - M. marinum whole-cell based host-pathogen assay, to detect anti-infective activities. RESULTS By the end of the survey, nearly 30 plants were cited and the 12 most cited herbal drugs were collected and their usage documented by extensive literature search. Extracts of the chosen herbs were screened with the described assays; with a main focus on traditional formulas (mainly herbal decoctions). Two of the water extracts from Combretum aculeatum and Guiera senegalensis showed significant antimycobacterial activities when compared to the positive control drug (rifampin). These extracts showed no observable toxicity against amoeba host cells (Acanthamoeba castellanii). CONCLUSIONS This study demonstrates that most of the patients do not concomitantly use plants and TB drugs (~90% of informants) but, instead, most are treated with medicinal plants before they are admitted to a hospital (41%). Interestingly, among the aqueous extracts assayed, two extracts (Combretum aculeatum (Combretaceae) and Guiera senegalensis (Combretaceae)) collected within this survey demonstrate antimycobacterial activities on the validated whole-cell based host-pathogen assay. Both extracts showed significant activities against intracellular and extracellular - M. marinum growth presenting IC50 lower than 0.5mg/ml compared to the reference drug Rifampin (IC50 of 0.4 and 7µg/ml). No toxicity was observed for amoebae cells at concentration until 0.8mg/ml.
Collapse
Affiliation(s)
- ElHadji Assane Diop
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland; Biology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland
| | - Sébastien Kicka
- Department of Biochemistry, Faculty of Science, University of Geneva, Quai Ansermet 30, Geneva 4, Switzerland
| | - Serge Rudaz
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland
| | - Tahir Diop
- Biology Department, University Cheikh Anta Diop, Dakar, Senegal
| | - Thierry Soldati
- Department of Biochemistry, Faculty of Science, University of Geneva, Quai Ansermet 30, Geneva 4, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU - Rue Michel Servet 1, 1211 Geneva 11, Switzerland.
| |
Collapse
|
97
|
Lupoli TJ, Vaubourgeix J, Burns-Huang K, Gold B. Targeting the Proteostasis Network for Mycobacterial Drug Discovery. ACS Infect Dis 2018; 4:478-498. [PMID: 29465983 PMCID: PMC5902792 DOI: 10.1021/acsinfecdis.7b00231] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the world's deadliest infectious diseases and urgently requires new antibiotics to treat drug-resistant strains and to decrease the duration of therapy. During infection, Mtb encounters numerous stresses associated with host immunity, including hypoxia, reactive oxygen and nitrogen species, mild acidity, nutrient starvation, and metal sequestration and intoxication. The Mtb proteostasis network, composed of chaperones, proteases, and a eukaryotic-like proteasome, provides protection from stresses and chemistries of host immunity by maintaining the integrity of the mycobacterial proteome. In this Review, we explore the proteostasis network as a noncanonical target for antibacterial drug discovery.
Collapse
Affiliation(s)
- Tania J. Lupoli
- Department of Microbiology and Immunology, Weill Cornell Medicine, 413 East 69th Street, New York, New York 10021, United States
| | - Julien Vaubourgeix
- Department of Microbiology and Immunology, Weill Cornell Medicine, 413 East 69th Street, New York, New York 10021, United States
| | - Kristin Burns-Huang
- Department of Microbiology and Immunology, Weill Cornell Medicine, 413 East 69th Street, New York, New York 10021, United States
| | - Ben Gold
- Department of Microbiology and Immunology, Weill Cornell Medicine, 413 East 69th Street, New York, New York 10021, United States
| |
Collapse
|
98
|
Malm S, Maaß S, Schaible UE, Ehlers S, Niemann S. In vivo virulence of Mycobacterium tuberculosis depends on a single homologue of the LytR-CpsA-Psr proteins. Sci Rep 2018; 8:3936. [PMID: 29500450 PMCID: PMC5834633 DOI: 10.1038/s41598-018-22012-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 02/12/2018] [Indexed: 12/27/2022] Open
Abstract
LytR-cpsA-Psr (LCP) domain containing proteins fulfil important functions in bacterial cell wall synthesis. In Mycobacterium tuberculosis complex (Mtbc) strains, the causative agents of tuberculosis (TB), the genes Rv3484 and Rv3267 encode for LCP proteins which are putatively involved in arabinogalactan transfer to peptidoglycan. To evaluate the significance of Rv3484 for Mtbc virulence, we generated a deletion mutant in the Mtbc strain H37Rv and studied its survival in mice upon aerosol infection. The deletion mutant failed to establish infection demonstrating that Rv3484 is essential for growth in mice. Following an initial phase of marginal replication in the lungs until day 21, the Rv3484 deletion mutant was almost eliminated by day 180 post-infectionem. Interestingly, the mutant also showed higher levels of resistance to meropenem/clavulanate and lysozyme, both targeting peptidoglycan structure. We conclude that Rv3484 is essential for Mtbc virulence in vivo where its loss of function cannot be compensated by Rv3267.
Collapse
Affiliation(s)
- S Malm
- Molecular and Experimental Mycobacteriology, Priority Area Infections, Research Center Borstel - Leibniz Center for Medicine and Biosciences, 23845, Borstel, Germany.
| | - S Maaß
- Molecular and Experimental Mycobacteriology, Priority Area Infections, Research Center Borstel - Leibniz Center for Medicine and Biosciences, 23845, Borstel, Germany
| | - U E Schaible
- Cellular Microbiology, Priority Area Infections, Research Center Borstel - Leibniz Center for Medicine and Biosciences, 23845, Borstel, Germany
| | - S Ehlers
- Molecular Inflammation Medicine, Priority Area Infections, Research Center Borstel - Leibniz Center for Medicine and Biosciences, 23845, Borstel, Germany
| | - S Niemann
- Molecular and Experimental Mycobacteriology, Priority Area Infections, Research Center Borstel - Leibniz Center for Medicine and Biosciences, 23845, Borstel, Germany
- German Center for Infection Research, Borstel Site, Borstel, Germany
| |
Collapse
|
99
|
Novel Pyrimidines as Antitubercular Agents. Antimicrob Agents Chemother 2018; 62:AAC.02063-17. [PMID: 29311070 DOI: 10.1128/aac.02063-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/02/2017] [Indexed: 01/25/2023] Open
Abstract
Mycobacterium tuberculosis infection is responsible for a global pandemic. New drugs are needed that do not show cross-resistance with the existing front-line therapeutics. A triazine antitubercular hit led to the design of a related pyrimidine family. The synthesis of a focused series of these analogs facilitated exploration of their in vitro activity, in vitro cytotoxicity, and physiochemical and absorption-distribution-metabolism-excretion properties. Select pyrimidines were then evaluated for their pharmacokinetic profiles in mice. The findings suggest a rationale for the further evolution of this promising series of antitubercular small molecules, which appear to share some similarities with the clinical compound PA-824 in terms of activation, while highlighting more general guidelines for the optimization of small-molecule antitubercular agents.
Collapse
|
100
|
Reinheimer C, Büttner D, Proschak E, Bode HB, Kempf VAJ, Wichelhaus TA. Anti-tubercular activity of a natural stilbene and its synthetic derivatives. GMS INFECTIOUS DISEASES 2018; 6:Doc01. [PMID: 30671332 PMCID: PMC6301740 DOI: 10.3205/id000036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objectives: Tuberculosis (TB) and multidrug- and extensively drug-resistant TB in particular are remaining a major global health challenge and efficient new drugs against TB are needed. This study evaluated the anti-tubercular activity of a natural stilbene and its synthetic derivatives against M. tuberculosis. Methods: Isopropylstilbene and its synthetic derivatives were analyzed for their anti-tubercular activity against M. tuberculosis ATCC 27294 as well as multidrug- and extensively drug-resistant M. tuberculosis clinical isolates by using MGIT 960 instrumentation and EpiCenter software equipped with TB eXiST module. Cytotoxic effects of drug candidates were determined by a MTT dye reduction assay using A549 adenocarcinomic human alveolar basal epithelial cells. Results: Growth of M. tuberculosis ATCC 27294 was suppressed by the natural isopropylstilbene HB64 as well as synthetic derivatives DB56 and DB55 at 25 µg/ml. Growth of clinical isolates MDR and XDR M. tuberculosis was suppressed by HB64 at 100 µg/ml as well as by synthetic derivatives DB56 and DB55 at 50 µg/ml and 25 µg/ml, respectively. No anti-tubercular activity was demonstrated for synthetic derivatives DB53, EB251, and RB57 at 100 µg/ml. Toxicity in terms of IC50 values of HB64, DB55 and DB56 were 7.92 µg/ml, 12.15 µg/ml and 16.01 µg/ml, respectively. Conclusions: Synthetical derivatives of stilbene might be effective candidates as anti-tubercular drugs. However, toxicity of these substances as determined by IC50 values might limit therapeutic success in vivo. Further investigations should address lowering the toxicity for parenteral administration by remodeling stilbene derivatives.
Collapse
Affiliation(s)
- Claudia Reinheimer
- Institute of Medical Microbiology and Infection Control, Hospital of Goethe-University, Frankfurt, Germany
| | - Dominik Büttner
- Institute of Pharmaceutical Chemistry, Goethe-University, Frankfurt, Germany
| | - Eugen Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University, Frankfurt, Germany
| | - Helge B Bode
- Merck endowed chair for Molecular Biotechnology, Department of Biosciences and Buchmann Institute for Molecular Life Sciences (BMLS), Goethe-University, Frankfurt, Germany
| | - Volkhard A J Kempf
- Institute of Medical Microbiology and Infection Control, Hospital of Goethe-University, Frankfurt, Germany
| | - Thomas A Wichelhaus
- Institute of Medical Microbiology and Infection Control, Hospital of Goethe-University, Frankfurt, Germany
| |
Collapse
|