1
|
Li F, Marwitz F, Rudolph D, Gauda W, Cohrs M, Neumann PR, Lucas H, Kollan J, Tahir A, Schwudke D, Feldmann C, Hädrich G, Dailey LA. A Comparative Pharmacokinetics Study of Orally and Intranasally Administered 8-Nitro-1,3-benzothiazin-4-one (BTZ043) Amorphous Drug Nanoparticles. ACS Pharmacol Transl Sci 2024; 7:4123-4134. [PMID: 39698258 PMCID: PMC11651166 DOI: 10.1021/acsptsci.4c00558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 12/20/2024]
Abstract
BTZ043 is an 8-nitro-1,3-benzothiazin-4-one with potency against multidrug-resistant Mycobacterium tuberculosis. Low solubility and hepatic metabolism are linked to poor oral bioavailability. Amorphous drug nanoparticles (ADN) were formulated to improve the bioavailability. Comparative pharmacokinetics of BTZ043 ADN following intranasal (2.5 mg kg-1) and oral administration (25 mg kg-1) in Balb/c mice was investigated using oral BTZ043 drug suspensions (neat; 25 mg kg-1) as a standard-of-care reference. Plasma exposure following oral ADN administration was 8-fold higher than for oral neat BTZ043. Intranasal ADN increased plasma exposure 18-fold compared to oral neat BTZ043 after dose normalization. BTZ043 was detectable in lung lining fluid following ADN administration, but not after oral neat BTZ043 dosing. BTZ043 was cleared faster from the lung and plasma following intranasal administration with a shorter time above the minimum inhibitory concentration (MIC) compared to oral ADN. Since time > MIC is reported to drive activity, oral ADN may represent a promising delivery strategy for BTZ043.
Collapse
Affiliation(s)
- Feng Li
- Department
of Pharmaceutical Sciences, Division of Pharmaceutical Technology
and Biopharmaceutics, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
- Department
of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
- Vienna
Doctoral School of Pharmaceutical, Nutritional and Sport Sciences
(PhaNuSpo), University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Franziska Marwitz
- Division
of Bioanalytical Chemistry, Research Center
Borstel, Leibniz Lung Center, Borstel 23845, Germany
- Thematic
Translational Unit Tuberculosis, German
Center for Infection Research (DZIF), Borstel 23845, Germany
| | - David Rudolph
- Institute
of Inorganic Chemistry, Karlsruhe Institute of Technology (KIT), Engesserstraße 15, Karlsruhe 76131, Germany
| | - Wiebke Gauda
- Division
of Bioanalytical Chemistry, Research Center
Borstel, Leibniz Lung Center, Borstel 23845, Germany
| | - Michaela Cohrs
- Laboratory
for General Biochemistry and Physical Pharmacy, Ghent University, 9000 Gent, Belgium
| | - Paul Robert Neumann
- Department
of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Henrike Lucas
- Department
of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Julia Kollan
- Department
of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Ammar Tahir
- Department
of Pharmaceutical Sciences, Division of Pharmacognosy, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Dominik Schwudke
- Division
of Bioanalytical Chemistry, Research Center
Borstel, Leibniz Lung Center, Borstel 23845, Germany
- Thematic
Translational Unit Tuberculosis, German
Center for Infection Research (DZIF), Borstel 23845, Germany
- German
Center for Lung Research (DZL), Airway Research
Center North (ARCN), Research Center Borstel, Leibniz Lung Center, Borstel 23845, Germany
- Kiel
Nano, Surface and Interface Sciences (KiNSIS), Kiel University, Kiel 24118, Germany
| | - Claus Feldmann
- Institute
of Inorganic Chemistry, Karlsruhe Institute of Technology (KIT), Engesserstraße 15, Karlsruhe 76131, Germany
| | - Gabriela Hädrich
- Department
of Pharmaceutical Sciences, Division of Pharmaceutical Technology
and Biopharmaceutics, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
- Department
of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| | - Lea Ann Dailey
- Department
of Pharmaceutical Sciences, Division of Pharmaceutical Technology
and Biopharmaceutics, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
- Department
of Pharmaceutical Technology and Biopharmaceutics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale) 06120, Germany
| |
Collapse
|
2
|
Sonawane K, Said R, Lele M, Chaudhari H, Hatvate N. Recent Advancements in Benzothiazinones (BTZ) Analogs as DprE1 Inhibitor for Potent Antitubercular Therapeutics. ChemistrySelect 2024; 9. [DOI: 10.1002/slct.202404094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/30/2024] [Indexed: 01/05/2025]
Abstract
AbstractBenzothiazinone analogs have emerged as a promising class of compounds having potent antimycobacterial activity, particularly against Mycobacterium tuberculosis, the pathogen responsible for tuberculosis. This review highlights the development of benzothiazinone analogs as potential antitubercular agents from the beginning to the recent advancement in the past decade. These compounds have shown potent activity, including drug‐resistant strains of Mycobacterium tuberculosis. Structure–activity relationship studies and modifications have improved their efficacy. Benzothiazinone analogs have favorable pharmacokinetic profiles and show promise in preclinical studies. Challenges include addressing resistance mechanisms and ensuring safety. Their unique mode of action and promising properties make them attractive candidates for the battle against drug‐resistant tuberculosis.
Collapse
Affiliation(s)
- Kalyani Sonawane
- Department of Pharmaceutical Technology Institute of Chemical Technology Mumbai Marathwada Campus Jalna Maharashtra 431203 India
| | - Rushikesh Said
- Department of Pharmaceutical Technology Institute of Chemical Technology Mumbai Marathwada Campus Jalna Maharashtra 431203 India
| | - Mukta Lele
- Department of Pharmaceutical Technology Institute of Chemical Technology Mumbai Marathwada Campus Jalna Maharashtra 431203 India
| | - Hemchandra Chaudhari
- Department of Pharmaceutical Sciences and Technology Institute of Chemical Technology Mumbai Maharashtra 400019 India
| | - Navnath Hatvate
- Department of Pharmaceutical Technology Institute of Chemical Technology Mumbai Marathwada Campus Jalna Maharashtra 431203 India
| |
Collapse
|
3
|
Shaikh J, Patel S, Nagani A, Shah M, Ugharatdar S, Patel A, Shah D, Patel D. Pharmacophore mapping, 3D QSAR, molecular docking, and ADME prediction studies of novel Benzothiazinone derivatives. In Silico Pharmacol 2024; 12:79. [PMID: 39220602 PMCID: PMC11362452 DOI: 10.1007/s40203-024-00255-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
In the quest to combat tuberculosis, DprE1, a challenging target for novel anti-tubercular agents due to its small size and membrane location, has been a focus of research. DprE1 catalyzes the transformation of DPR into Ketoribose DPX, with Benzothiazinone emerging as a potent pharmacophore for inhibiting DprE1. Clinical trial drugs such as BTZ043, BTZ038, PBTZ169, and TMC-207 have shown promising results as DprE1 inhibitors. This study employed pharmacophore mapping of Pyrazolopyridine, Dinitrobenzamide, and Benzothiazinone derivatives to identify crucial features for eliciting a biological response. Benzothiazinone (Ligand code: 73) emerged as a reference ligand with a fitness score of 3.000. ROC analysis validated the pharmacophore with an excellent score of 0.71. To build a 3D QSAR model, a series of Benzothiazinone congeneric derivatives were explored. The model exhibited strong performance, with a standard deviation of 0.1531, a correlation coefficient for the training set (R2) value of 0.9754, and a correlation coefficient for test set Q2 value of 0.7632, indicating robust predictive capabilities. Contour maps guided the design of novel benzothiazinone derivatives, emphasizing steric, electrostatic, hydrophobic, H-bond acceptor, and H-bond donor groups for structure-activity relationships. Docking studies against PDB ID: 4NCR demonstrated favorable scores, with interactions aligning well with the in-built ligand 26 J. Docking validation via RMSD values supported the reliability of the docking results. This comprehensive approach aids in the design of novel benzothiazinone derivatives with potential anti-tubercular properties, contributing to the development of novel anti-tubercular agents which can be pivotal in the eradication of tuberculosis.
Collapse
Affiliation(s)
- Jahaan Shaikh
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat India
| | - Salman Patel
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat India
| | - Afzal Nagani
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat India
- Research and Development Cell, Parul University, Vadodara, Gujarat India
| | - Moksh Shah
- Department of Pharmaceutical Chemistry, Parul Institute of Pharmacy, Parul University, Vadodara, Gujarat India
| | - Siddik Ugharatdar
- Department of Pharmaceutical Chemistry, Laxminarayandev College of Pharmacy, Bholav, Bharuch, Gujarat India
| | - Ashish Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Anand, Gujarat India
| | - Drashti Shah
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Anand, Gujarat India
| | - Dharti Patel
- Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology, CHARUSAT Campus, Changa, Anand, Gujarat India
| |
Collapse
|
4
|
Ramey ME, Kaya F, Bauman AA, Massoudi LM, Sarathy JP, Zimmerman MD, Scott DWL, Job AM, Miller-Dawson JA, Podell BK, Lyons MA, Dartois V, Lenaerts AJ, Robertson GT. Drug distribution and efficacy of the DprE1 inhibitor BTZ-043 in the C3HeB/FeJ mouse tuberculosis model. Antimicrob Agents Chemother 2023; 67:e0059723. [PMID: 37791784 PMCID: PMC10648937 DOI: 10.1128/aac.00597-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/04/2023] [Indexed: 10/05/2023] Open
Abstract
BTZ-043, a suicide inhibitor of the Mycobacterium tuberculosis cell wall synthesis decaprenylphosphoryl-beta-D-ribose 2' epimerase, is under clinical development as a potential new anti-tuberculosis agent. BTZ-043 is potent and bactericidal in vitro but has limited activity against non-growing bacilli in rabbit caseum. To better understand its behavior in vivo, BTZ-043 was evaluated for efficacy and spatial drug distribution as a single agent in the C3HeB/FeJ mouse model presenting with caseous necrotic pulmonary lesions upon Mycobacterium tuberculosis infection. BTZ-043 promoted significant reductions in lung and spleen bacterial burdens in the C3HeB/FeJ mouse model after 2 months of therapy. BTZ-043 penetrates cellular and necrotic lesions and was retained at levels above the serum-shifted minimal inhibitory concentration in caseum. The calculated rate of kill was found to be highest and dose-dependent during the second month of treatment. BTZ-043 treatment was associated with improved histology scores of pulmonary lesions, especially compared to control mice, which experienced advanced fulminant neutrophilic alveolitis in the absence of treatment. These positive treatment responses to BTZ-043 monotherapy in a mouse model of advanced pulmonary disease can be attributed to favorable distribution in tissues and lesions, retention in the caseum, and its high potency and bactericidal nature at drug concentrations achieved in necrotic lesions.
Collapse
Affiliation(s)
- Michelle E. Ramey
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Firat Kaya
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Allison A. Bauman
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Lisa M. Massoudi
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Jansy P. Sarathy
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Matthew D. Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Dashick W. L. Scott
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Alyx M. Job
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Jake A. Miller-Dawson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Brendan K. Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Michael A. Lyons
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian School of Medicine, Nutley, New Jersey, USA
| | - Anne J. Lenaerts
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - Gregory T. Robertson
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
5
|
Yadav S, Soni A, Tanwar O, Bhadane R, Besra GS, Kawathekar N. DprE1 Inhibitors: Enduring Aspirations for Future Antituberculosis Drug Discovery. ChemMedChem 2023; 18:e202300099. [PMID: 37246503 DOI: 10.1002/cmdc.202300099] [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: 02/19/2023] [Revised: 05/22/2023] [Accepted: 05/24/2023] [Indexed: 05/30/2023]
Abstract
DprE1 is a crucial enzyme involved in the cell wall synthesis of Mycobacterium tuberculosis and a promising target for antituberculosis drug development. However, its unique structural characteristics for ligand binding and association with DprE2 make developing new clinical compounds challenging. This review provides an in-depth analysis of the structural requirements for both covalent and non-covalent inhibitors, their 2D and 3D binding patterns, as well as their biological activity data in vitro and in vivo, including pharmacokinetic information. We also introduce a protein quality score (PQS) and an active-site map of the DprE1 enzyme to help medicinal chemists better understand DprE1 inhibition and develop new and effective anti-TB drugs. Furthermore, we examine the resistance mechanisms associated with DprE1 inhibitors to understand future developments due to resistance emergence. This comprehensive review offers insight into the DprE1 active site, including protein-binding maps, PQS, and graphical representations of known inhibitors, making it a valuable resource for medicinal chemists working on future antitubercular compounds.
Collapse
Affiliation(s)
- Saloni Yadav
- Department of Pharmacy, Shri Govindram Seksaria Institute of Technology and Science, 23-Park Road, Indore, Madhya Pradesh, India
| | - Aastha Soni
- Department of Pharmacy, Shri Govindram Seksaria Institute of Technology and Science, 23-Park Road, Indore, Madhya Pradesh, India
| | - Omprakash Tanwar
- Department of Pharmacy, Shri Govindram Seksaria Institute of Technology and Science, 23-Park Road, Indore, Madhya Pradesh, India
| | - Rajendra Bhadane
- Turku Cellular Microbiology Laboratory (TCML), Åbo Akademi University, 20014, Turku, Finland
- Institute of Biomedicine, University of Turku, 20520, Turku, Finland
| | - Gurdyal S Besra
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Neha Kawathekar
- Department of Pharmacy, Shri Govindram Seksaria Institute of Technology and Science, 23-Park Road, Indore, Madhya Pradesh, India
| |
Collapse
|
6
|
Eckhardt E, Li Y, Mamerow S, Schinköthe J, Sehl-Ewert J, Dreisbach J, Corleis B, Dorhoi A, Teifke J, Menge C, Kloss F, Bastian M. Pharmacokinetics and Efficacy of the Benzothiazinone BTZ-043 against Tuberculous Mycobacteria inside Granulomas in the Guinea Pig Model. Antimicrob Agents Chemother 2023; 67:e0143822. [PMID: 36975792 PMCID: PMC10112198 DOI: 10.1128/aac.01438-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 02/16/2023] [Indexed: 03/29/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, is the world's leading cause of mortality from a single bacterial pathogen. With increasing frequency, emergence of drug-resistant mycobacteria leads to failures of standard TB treatment regimens. Therefore, new anti-TB drugs are urgently required. BTZ-043 belongs to a novel class of nitrobenzothiazinones, which inhibit mycobacterial cell wall formation by covalent binding of an essential cysteine in the catalytic pocket of decaprenylphosphoryl-β-d-ribose oxidase (DprE1). Thus, the compound blocks the formation of decaprenylphosphoryl-β-d-arabinose, a precursor for the synthesis of arabinans. An excellent in vitro efficacy against M. tuberculosis has been demonstrated. Guinea pigs are an important small-animal model to study anti-TB drugs, as they are naturally susceptible to M. tuberculosis and develop human-like granulomas after infection. In the current study, dose-finding experiments were conducted to establish the appropriate oral dose of BTZ-043 for the guinea pig. Subsequently, it could be shown that the active compound was present at high concentrations in Mycobacterium bovis BCG-induced granulomas. To evaluate its therapeutic effect, guinea pigs were subcutaneously infected with virulent M. tuberculosis and treated with BTZ-043 for 4 weeks. BTZ-043-treated guinea pigs had reduced and less necrotic granulomas than vehicle-treated controls. In comparison to the vehicle controls a highly significant reduction of the bacterial burden was observed after BTZ-043 treatment at the site of infection and in the draining lymph node and spleen. Together, these findings indicate that BTZ-043 holds great promise as a new antimycobacterial drug.
Collapse
Affiliation(s)
- Emmelie Eckhardt
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Yan Li
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology, Leibniz-HKI, Jena, Germany
| | - Svenja Mamerow
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Jena, Germany
| | - Jan Schinköthe
- Institute of Veterinary Pathology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Julia Sehl-Ewert
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Julia Dreisbach
- Division of Infectious Diseases and Tropical Medicine, University Hospital of the University of Munich (LMU), Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Björn Corleis
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Anca Dorhoi
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Jens Teifke
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| | - Christian Menge
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Jena, Germany
| | - Florian Kloss
- Transfer Group Anti-infectives, Leibniz Institute for Natural Product Research and Infection Biology, Leibniz-HKI, Jena, Germany
| | - Max Bastian
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald, Germany
| |
Collapse
|
7
|
Amado PM, Woodley C, Cristiano MLS, O’Neill PM. Recent Advances of DprE1 Inhibitors against Mycobacterium tuberculosis: Computational Analysis of Physicochemical and ADMET Properties. ACS OMEGA 2022; 7:40659-40681. [PMID: 36406587 PMCID: PMC9670723 DOI: 10.1021/acsomega.2c05307] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/21/2022] [Indexed: 05/14/2023]
Abstract
Decaprenylphosphoryl-β-d-ribose 2'-epimerase (DprE1) is a critical flavoenzyme in Mycobacterium tuberculosis, catalyzing a vital step in the production of lipoarabinomannan and arabinogalactan, both of which are essential for cell wall biosynthesis. Due to its periplasmic localization, DprE1 is a susceptible target, and several compounds with diverse scaffolds have been discovered that inhibit this enzyme, covalently or noncovalently. We evaluated a total of ∼1519 DprE1 inhibitors disclosed in the literature from 2009 to April 2022 by performing an in-depth analysis of physicochemical descriptors and absorption, distribution, metabolism, excretion, and toxicity (ADMET), to gain new insights into these properties in DprE1 inhibitors. Several molecular properties that should facilitate the design and optimization of future DprE1 inhibitors are described, allowing for the development of improved analogues targeting M. tuberculosis.
Collapse
Affiliation(s)
- Patrícia
S. M. Amado
- Center
of Marine Sciences - CCMAR, University of
Algarve, P-8005-039 Faro, Portugal
- Department
of Chemistry and Pharmacy, FCT, University
of Algarve, P-8005-039 Faro, Portugal
- Department
of Chemistry, University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Christopher Woodley
- Department
of Chemistry, University of Liverpool, Liverpool L69 7ZD, United Kingdom
| | - Maria L. S. Cristiano
- Center
of Marine Sciences - CCMAR, University of
Algarve, P-8005-039 Faro, Portugal
- Department
of Chemistry and Pharmacy, FCT, University
of Algarve, P-8005-039 Faro, Portugal
- Email
for M.L.S.C.:
| | - Paul M. O’Neill
- Department
of Chemistry, University of Liverpool, Liverpool L69 7ZD, United Kingdom
- Email for P.M.O.:
| |
Collapse
|
8
|
Fernandes GFS, Thompson AM, Castagnolo D, Denny WA, Dos Santos JL. Tuberculosis Drug Discovery: Challenges and New Horizons. J Med Chem 2022; 65:7489-7531. [PMID: 35612311 DOI: 10.1021/acs.jmedchem.2c00227] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Over the past 2000 years, tuberculosis (TB) has claimed more lives than any other infectious disease. In 2020 alone, TB was responsible for 1.5 million deaths worldwide, comparable to the 1.8 million deaths caused by COVID-19. The World Health Organization has stated that new TB drugs must be developed to end this pandemic. After decades of neglect in this field, a renaissance era of TB drug discovery has arrived, in which many novel candidates have entered clinical trials. However, while hundreds of molecules are reported annually as promising anti-TB agents, very few successfully progress to clinical development. In this Perspective, we critically review those anti-TB compounds published in the last 6 years that demonstrate good in vivo efficacy against Mycobacterium tuberculosis. Additionally, we highlight the main challenges and strategies for developing new TB drugs and the current global pipeline of drug candidates in clinical studies to foment fresh research perspectives.
Collapse
Affiliation(s)
- Guilherme F S Fernandes
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - Andrew M Thompson
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Daniele Castagnolo
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, United Kingdom
| | - William A Denny
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Jean L Dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800903, Brazil
| |
Collapse
|
9
|
Bharti H, Singal A, Saini M, Cheema PS, Raza M, Kundu S, Nag A. Repurposing the Pathogen Box compounds for identification of potent anti-malarials against blood stages of Plasmodium falciparum with PfUCHL3 inhibitory activity. Sci Rep 2022; 12:918. [PMID: 35042884 PMCID: PMC8766476 DOI: 10.1038/s41598-021-04619-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022] Open
Abstract
Malaria has endured as a global epidemic since ages and its eradication poses an immense challenge due to the complex life cycle of the causative pathogen and its tolerance to a myriad of therapeutics. PfUCHL3, a member of the ubiquitin C-terminal hydrolase (UCH) family of deubiquitinases (DUBs) is cardinal for parasite survival and emerges as a promising therapeutic target. In this quest, we employed a combination of computational and experimental approaches to identify PfUCHL3 inhibitors as novel anti-malarials. The Pathogen Box library was screened against the crystal structure of PfUCHL3 (PDB ID: 2WE6) and its human ortholog (PDB ID: 1XD3). Fifty molecules with better comparative score, bioavailability and druglikeliness were subjected to in-vitro enzyme inhibition assay and among them only two compounds effectively inhibited PfUCHL3 activity at micro molar concentrations. Both MMV676603 and MMV688704 exhibited anti-plasmodial activity by altering the parasite phenotype at late stages of the asexual life cycle and inducing the accumulation of polyubiquitinated substrates. In addition, both the compounds were non-toxic and portrayed high selectivity window for the parasite over mammalian cells. This is the first comprehensive study to demonstrate the anti-malarial efficacy of PfUCHL3 inhibitors and opens new avenues to exploit UCH family of DUBs as a promising target for the development of next generation anti-malaria therapy.
Collapse
Affiliation(s)
- Hina Bharti
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Aakriti Singal
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Manisha Saini
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Pradeep Singh Cheema
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Mohsin Raza
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Suman Kundu
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India
| | - Alo Nag
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, New Delhi, 110021, India.
| |
Collapse
|
10
|
In silico analyses of predicted substitutions in fibrinolytic protein ‘Lumbrokinase-6’ suggest enhanced activity. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Chemical Classes Presenting Novel Antituberculosis Agents Currently in Different Phases of Drug Development: A 2010-2020 Review. PHARMACEUTICALS (BASEL, SWITZERLAND) 2021; 14:ph14050461. [PMID: 34068171 PMCID: PMC8152995 DOI: 10.3390/ph14050461] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 01/18/2023]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a curable airborne disease currently treated using a drug regimen consisting of four drugs. Global TB control has been a persistent challenge for many decades due to the emergence of drug-resistant Mtb strains. The duration and complexity of TB treatment are the main issues leading to treatment failures. Other challenges faced by currently deployed TB regimens include drug-drug interactions, miss-matched pharmacokinetics parameters of drugs in a regimen, and lack of activity against slow replicating sub-population. These challenges underpin the continuous search for novel TB drugs and treatment regimens. This review summarizes new TB drugs/drug candidates under development with emphasis on their chemical classes, biological targets, mode of resistance generation, and pharmacokinetic properties. As effective TB treatment requires a combination of drugs, the issue of drug-drug interaction is, therefore, of great concern; herein, we have compiled drug-drug interaction reports, as well as efficacy reports for drug combinations studies involving antitubercular agents in clinical development.
Collapse
|
12
|
Stephanie F, Saragih M, Tambunan USF. Recent Progress and Challenges for Drug-Resistant Tuberculosis Treatment. Pharmaceutics 2021; 13:pharmaceutics13050592. [PMID: 33919204 PMCID: PMC8143172 DOI: 10.3390/pharmaceutics13050592] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 11/23/2022] Open
Abstract
Control of Mycobacterium tuberculosis infection continues to be an issue, particularly in countries with a high tuberculosis (TB) burden in the tropical and sub-tropical regions. The effort to reduce the catastrophic cost of TB with the WHO’s End TB Strategy in 2035 is still obstructed by the emergence of drug-resistant TB (DR-TB) cases as result of various mutations of the MTB strain. In the approach to combat DR-TB, several potential antitubercular agents were discovered as inhibitors for various existing and novel targets. Host-directed therapy and immunotherapy also gained attention as the drug-susceptibility level of the pathogen can be reduced due to the pathogen’s evolutionary dynamics. This review is focused on the current progress and challenges in DR-TB treatment. We briefly summarized antitubercular compounds that are under development and trials for both DR-TB drug candidates and host-directed therapy. We also highlighted several problems in DR-TB diagnosis, the treatment regimen, and drug discovery that have an impact on treatment adherence and treatment failure.
Collapse
|
13
|
Zhang G, Sheng L, Hegde P, Li Y, Aldrich CC. 8-cyanobenzothiazinone analogs with potent antitubercular activity. Med Chem Res 2021; 30:449-458. [PMID: 33462533 PMCID: PMC7805566 DOI: 10.1007/s00044-020-02676-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/01/2020] [Indexed: 01/27/2023]
Abstract
8-Nitrobenzothiazinones (BTZs) exemplified by macozinone are a new class of antitubercular agents with exceptionally potent activity. The aryl nitro group has been considered indispensable for activity since this is bioactivated within mycobacteria by the flavoenzyme DprE1 to a reactive nitroso metabolite that covalently labels Cys387. However, the aryl nitro group is a potential liability with regards to safety, stability, and resistance. In this paper, we introduced a nitrile as a bioisosteric replacement of the nitro group, which we hypothesize can maintain a similar covalent mechanism of inhibition, but mitigate against the aforementioned concerns. 8-cyanobenzothiazinone 1d displayed potent antitubercular activity with an MIC of 130 nM and had an improved volume of distribution in mice that increased the intrinsic half-life by twofold compared to macozinone. Analysis of the C-2 substituent of 1d revealed similar structure-activity relationships as observed for macozinone. Overall, the results confirm the 8-nitro group of benzothiazinones can be successfully replaced with a nitrile to retain useful activity and favorable pharmacokinetic properties.
Collapse
Affiliation(s)
- Gang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050 China
| | - Li Sheng
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050 China
| | - Pooja Hegde
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| | - Yan Li
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050 China
| | - Courtney C. Aldrich
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100050 China
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455 USA
| |
Collapse
|
14
|
Zhang G, Chen D, Wang S, Chen H, Wei N, Chen G. Molecular Insight into the Discrepancy of Antitubercular Activity between 8‐Nitro and 8‐Cyano Benzothiazinones. ChemistrySelect 2020. [DOI: 10.1002/slct.202004156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Gang Zhang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences Beijing 100050 China
| | - Daoxing Chen
- School of Pharmaceutical Sciences Wenzhou Medical University, Wenzhou Zhejiang 325035 China
| | - Sufang Wang
- The Environment Analysis and Testing Laboratory Chinese Research Academy of Environmental Sciences Beijing 100012 China
| | - Hualong Chen
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology Planning and Research Department, Beijing Science Center Collaborative Innovation Center for Brain Disorders, Capital Medical University Beijing 100029 China
| | - Ning Wei
- Division of Hematology-Oncology Cancer Therapeutics Program, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh Department of Medicine, University of Pittsburgh School of Medicine Pittsburgh PA 15261 USA
| | - Guirong Chen
- Liaoning University of Traditional Chinese Medicine, Shenyang Liaoning 110847 China
| |
Collapse
|
15
|
Shetye GS, Franzblau SG, Cho S. New tuberculosis drug targets, their inhibitors, and potential therapeutic impact. Transl Res 2020; 220:68-97. [PMID: 32275897 DOI: 10.1016/j.trsl.2020.03.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/28/2020] [Accepted: 03/09/2020] [Indexed: 11/18/2022]
Abstract
The current tuberculosis (TB) predicament poses numerous challenges and therefore every incremental scientific work and all positive socio-political engagements, are steps taken in the right direction to eradicate TB. Progression of the late stage TB-drug pipeline into the clinics is an immediate deliverable of this global effort. At the same time, fueling basic research and pursuing early discovery work must be sustained to maintain a healthy TB-drug pipeline. This review encompasses a broad analysis of chemotherapeutic strategies that target the DNA replication, protein synthesis, cell wall biosynthesis, energy metabolism and proteolysis of Mycobacterium tuberculosis (Mtb). It includes a status check of the current TB-drug pipeline with a focus on the associated biology, emerging targets, and their promising chemical inhibitors. Potential synergies and/or gaps within or across different chemotherapeutic strategies are systematically reviewed as well.
Collapse
Affiliation(s)
- Gauri S Shetye
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Scott G Franzblau
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois
| | - Sanghyun Cho
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
16
|
Anti-tuberculosis activity of bio-active compounds from Lantana camara L., Euphorbia hirta L., Mukia maderaspatana (L.) M. Roem, and Abutilon indicum (L.). Eur J Integr Med 2020. [DOI: 10.1016/j.eujim.2020.101105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Mycobacterial Cell Wall: A Source of Successful Targets for Old and New Drugs. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10072278] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Eighty years after the introduction of the first antituberculosis (TB) drug, the treatment of drug-susceptible TB remains very cumbersome, requiring the use of four drugs (isoniazid, rifampicin, ethambutol and pyrazinamide) for two months followed by four months on isoniazid and rifampicin. Two of the drugs used in this “short”-course, six-month chemotherapy, isoniazid and ethambutol, target the mycobacterial cell wall. Disruption of the cell wall structure can enhance the entry of other TB drugs, resulting in a more potent chemotherapy. More importantly, inhibition of cell wall components can lead to mycobacterial cell death. The complexity of the mycobacterial cell wall offers numerous opportunities to develop drugs to eradicate Mycobacterium tuberculosis, the causative agent of TB. In the past 20 years, researchers from industrial and academic laboratories have tested new molecules to find the best candidates that will change the face of TB treatment: drugs that will shorten TB treatment and be efficacious against active and latent, as well as drug-resistant TB. Two of these new TB drugs block components of the mycobacterial cell wall and have reached phase 3 clinical trial. This article reviews TB drugs targeting the mycobacterial cell wall in use clinically and those in clinical development.
Collapse
|
18
|
Zhao W, Wang B, Liu Y, Fu L, Sheng L, Zhao H, Lu Y, Zhang D. Design, synthesis, and biological evaluation of novel 4H-chromen-4-one derivatives as antituberculosis agents against multidrug-resistant tuberculosis. Eur J Med Chem 2020; 189:112075. [DOI: 10.1016/j.ejmech.2020.112075] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 11/28/2022]
|
19
|
Khaligh NG, Abbo H, Titinchi SJ, Johan MR. An Overview of Recent Advances in Biological and Pharmaceutical Developments of Fluoro-containing Drugs. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824666191213123930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
:
This review article provides a brief assessment of the biological and pharmaceutical
developments of fluorinated drugs. It also discusses possible impacts on the further
development of new fluoro-containing pharmaceuticals. Structural aspects of new
drug-candidates currently under development and their biological properties, therapeutic
potential and syntheses are critically evaluated
Collapse
Affiliation(s)
- Nader G. Khaligh
- Nanotechnology and Catalysis Research Center, Institute of Postgraduate Studies, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Hanna Abbo
- Department of Chemistry, College of Science, University of Basrah, Basrah, Iraq
| | - Salam J.J. Titinchi
- Department of Chemistry, University of the Western Cape, Cape Town, South Africa
| | - Mohd R. Johan
- Nanotechnology and Catalysis Research Center, Institute of Postgraduate Studies, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Verma H, Choudhary S, Singh PK, Kashyap A, Silakari O. Decoding the signature of molecular mechanism involved in mutation associated resistance to 1, 3-benzothiazin-4-ones (Btzs) based DprE1 inhibitors using BTZ043 as a reference drug. MOLECULAR SIMULATION 2019. [DOI: 10.1080/08927022.2019.1659507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Himanshu Verma
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Shalki Choudhary
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Pankaj Kumar Singh
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Aanchal Kashyap
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Om Silakari
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| |
Collapse
|
21
|
Al-Ghafli H, Al-Hajoj S. Clinical Management of Drug-resistant Mycobacterium tuberculosis Strains: Pathogen-targeted Versus Host-directed Treatment Approaches. Curr Pharm Biotechnol 2019; 20:272-284. [DOI: 10.2174/1389201019666180731120544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/19/2018] [Accepted: 07/24/2018] [Indexed: 12/13/2022]
Abstract
Background:
Despite exerted efforts to control and treat Mycobacterium tuberculosis (MTB)
strains, Tuberculosis (TB) remains a public health menace. The emergence of complex drug-resistant profiles,
such as multi-drug resistant and extensively drug-resistant MTB strains, emphasizes the need for
early diagnosis of resistant cases, shorter treatment options, and effective medical interventions.
Objective:
Solutions for better clinical management of drug-resistant cases are either pathogencentered
(novel chemotherapy agents) or host-directed approaches (modulating host immune response
to prevent MTB invasion and pathogenesis).
Results:
Despite the overall potentiality of several chemotherapy agents, it is feared that their effectiveness
could be challenged by sequential pathogen adaptation tactics. On the contrary, host-directed
therapy options might offer a long-term conceivable solution.
Conclusion:
This review discusses the main suggestions proposed so far to resolve the clinical challenges
associated with drug resistance, in the context of TB. These suggestions include novel drug delivery approaches
that could optimize treatment outcome and increase patients’ compliance to the treatment.
Collapse
Affiliation(s)
- Hawra Al-Ghafli
- Department of Infections and Immunity, King Faisal Specialist Hospital and Research Center, P.O. Box. 3354 Riyadh 11211 MBC:03, Riyadh, Saudi Arabia
| | - Sahal Al-Hajoj
- Department of Infections and Immunity, King Faisal Specialist Hospital and Research Center, P.O. Box. 3354 Riyadh 11211 MBC:03, Riyadh, Saudi Arabia
| |
Collapse
|
22
|
Novel 1,3,4-oxadiazoles as antitubercular agents with limited activity against drug-resistant tuberculosis. Future Med Chem 2019; 11:499-510. [PMID: 30892944 DOI: 10.4155/fmc-2018-0378] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
AIM In recent times, heterocyclic chemotypes are being explored for the development of new antimycobacterials that target the drug-resistant tuberculosis. Here, we are disclosing the 5-substitued 2-mercapto-1,3,4-oxadiazoles as potent antitubercular agents. METHODOLOGY A small library of 2-mercapto-1,3,4-oxadiazoles was synthesized using various acids. The compounds were evaluated for antituberculosis activity against M. tuberculosis H37Rv. RESULTS Compound 8j was identified as antitubercular lead with MIC of 0.6 μg/ml against M. tuberculosis H37Rv. This compound was nontoxic to CHO-K1 cells and showed selectivity index of 39. Of note, 8j showed antitubercular activity against pre-extensively drug-resistant clinical isolate of Mycobacterium with MIC of 2 μg/ml. CONCLUSION This study provides potent antitubercular agent which can be further optimized to discover novel antibiotics.
Collapse
|
23
|
Veale CGL. Unpacking the Pathogen Box-An Open Source Tool for Fighting Neglected Tropical Disease. ChemMedChem 2019; 14:386-453. [PMID: 30614200 DOI: 10.1002/cmdc.201800755] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Indexed: 12/13/2022]
Abstract
The Pathogen Box is a 400-strong collection of drug-like compounds, selected for their potential against several of the world's most important neglected tropical diseases, including trypanosomiasis, leishmaniasis, cryptosporidiosis, toxoplasmosis, filariasis, schistosomiasis, dengue virus and trichuriasis, in addition to malaria and tuberculosis. This library represents an ensemble of numerous successful drug discovery programmes from around the globe, aimed at providing a powerful resource to stimulate open source drug discovery for diseases threatening the most vulnerable communities in the world. This review seeks to provide an in-depth analysis of the literature pertaining to the compounds in the Pathogen Box, including structure-activity relationship highlights, mechanisms of action, related compounds with reported activity against different diseases, and, where appropriate, discussion on the known and putative targets of compounds, thereby providing context and increasing the accessibility of the Pathogen Box to the drug discovery community.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Scottsville, 3209, South Africa
| |
Collapse
|
24
|
Li P, Wang B, Zhang X, Batt SM, Besra GS, Zhang T, Ma C, Zhang D, Lin Z, Li G, Huang H, Lu Y. Identification of novel benzothiopyranone compounds against Mycobacterium tuberculosis through scaffold morphing from benzothiazinones. Eur J Med Chem 2018; 160:157-170. [DOI: 10.1016/j.ejmech.2018.09.042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 08/18/2018] [Accepted: 09/14/2018] [Indexed: 12/20/2022]
|
25
|
Krutikov M, Bruchfeld J, Migliori GB, Borisov S, Tiberi S. New and repurposed drugs. Tuberculosis (Edinb) 2018. [DOI: 10.1183/2312508x.10021517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
26
|
Metabolism of SKLB-TB1001, a Potent Antituberculosis Agent, in Animals. Antimicrob Agents Chemother 2018; 62:AAC.02375-17. [PMID: 29686156 DOI: 10.1128/aac.02375-17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 03/27/2018] [Indexed: 02/05/2023] Open
Abstract
Tuberculosis is a major global health problem, and the emergence of multidrug-resistant and extensively drug-resistant strains has increased the difficulty of treating this disease. Among the novel antituberculosis drugs in the pipeline, decaprenylphosphoryl-beta-d-ribose-2-epimerase (DprE1) inhibitors such as BTZ043 and pBTZ169 exhibited extraordinary antituberculosis potency. Here, the metabolites of the new DprE1 inhibitor SKLB-TB1001 in vivo and its inhibition of cytochrome P450 isoforms and plasma protein binding (PPB) in vitro were studied. The results showed that rapid transformation and high PPB resulted in inadequate exposure in vivo and thus led to the moderate potency of SKLB-TB1001 in vivo This study provided explanations for the discrepant potency of this scaffold in vivo and in vitro Meanwhile, it also provides a rationale for lead optimization of this very promising scaffold of antituberculosis agents to prevent them from being metabolized, thus improving their exposure in vivo.
Collapse
|
27
|
Campaniço A, Moreira R, Lopes F. Drug discovery in tuberculosis. New drug targets and antimycobacterial agents. Eur J Med Chem 2018; 150:525-545. [DOI: 10.1016/j.ejmech.2018.03.020] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 01/24/2023]
|
28
|
Fernandes GFDS, Man Chin C, Dos Santos JL. Advances in Drug Discovery of New Antitubercular Multidrug-Resistant Compounds. Pharmaceuticals (Basel) 2017; 10:ph10020051. [PMID: 28587160 PMCID: PMC5490408 DOI: 10.3390/ph10020051] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 05/09/2017] [Accepted: 05/29/2017] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis (TB), a disease caused mainly by the Mycobacterium tuberculosis (Mtb), is according to the World Health Organization (WHO) the infectious disease responsible for the highest number of deaths worldwide. The increased number of multidrug-resistant (MDR-TB) and extensively drug-resistant (XDR-TB) strains, and the ineffectiveness of the current treatment against latent tuberculosis are challenges to be overcome in the coming years. The scenario of drug discovery becomes alarming when it is considered that the number of new drugs does not increase proportionally to the emergence of drug resistance. In this review, we will demonstrate the current advances in antitubercular drug discovery, focusing on the research of compounds with potent antituberculosis activity against MDR-TB strains. Herein, active compounds against MDR-TB with minimum inhibitory concentrations (MICs) less than 11 µM and low toxicity published in the last 4 years in the databases PubMed, Web of Science and Scopus will be presented and discussed.
Collapse
Affiliation(s)
- Guilherme Felipe Dos Santos Fernandes
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800060, Brazil.
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800903, Brazil.
| | - Chung Man Chin
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800903, Brazil.
| | - Jean Leandro Dos Santos
- Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800060, Brazil.
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara 14800903, Brazil.
| |
Collapse
|