1
|
Dong L, Qi H, Zhu Y, Yang Y, Zhao Y, Zhang S, Su Y, Yue T, Du X, Lei H, Yang Y. Identification of IspD as a novel target for tuberculosis treatment using compound M6. Front Microbiol 2024; 15:1461227. [PMID: 39606116 PMCID: PMC11599207 DOI: 10.3389/fmicb.2024.1461227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
INTRODUCTION Tuberculosis (TB) is a serious infectious disease that endangers human health, and TB becomes more difficult in eradiation due to its multidrug resistance (MDR). The objective of this research was to identify novel targets for treating TB. METHODS A 2-fold serial dilution method was used to determine minimal inhibitory concentrations (MIC) of compound M6 against Mycobacterium smegmatis (M. smegmatis). Compound M6 was subjected to reverse molecular docking with seven Mycobacterium tuberculosis proteins, and the best binding protein with the highest LibDock score was evaluated. The target protein with the highest score was purified through prokaryotic expression. Isolated target proteins were investigated for the enzyme activities and for the kinetic effect of compound M6 by absorbance detection. Subsequently, the CRISPR/Cas9 technology was employed to inhibit target gene expression for detecting MIC changes. Finally, potential targets were evaluated for the effect of the compound M6 in bacteria. RESULTS The MIC values of compound M6 against M. smegmatis were 32 μg/mL. The results from reverse molecular docking show that IspD has the highest LibDock score of 142.50, followed by Rv0674, IspF, and Dxr, with docking scores of 110.762, 71.6955, and 57.7446, respectively. IspD is a key enzyme in the 2-C-methyl-D-erythritol 4-phosphate pathway of MTB. The aKi and Ki values of M6 for the substrate MEP are 609.58 μM and 81.33 μM. For CTP, the aKi and Ki values are 657.89 μM and 40.07 μM. With tetracycline inducing CRISPR/Cas9 to suppress the expression of IspD, the MIC value of M6 against IspD went down significantly from 32 μg/mL to 4 μg/mL. CONCLUSION IspD is a novel target of the compound M6 for treating TB.
Collapse
Affiliation(s)
- Lijun Dong
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen, China
| | - Yue Zhu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yuma Yang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yue Zhao
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Sihan Zhang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yongqiang Su
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Taiyun Yue
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Xiancai Du
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Hetian Lei
- Department of Ophthalmology, The First People’s Hospital of Chenzhou, Chenzhou, China
| | - Yanhui Yang
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, The School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
2
|
Deshpande A, Likhar R, Khan T, Omri A. Decoding drug resistance in Mycobacterium tuberculosis complex: genetic insights and future challenges. Expert Rev Anti Infect Ther 2024; 22:511-527. [PMID: 39219506 DOI: 10.1080/14787210.2024.2400536] [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: 03/25/2024] [Revised: 06/02/2024] [Accepted: 08/31/2024] [Indexed: 09/04/2024]
Abstract
INTRODUCTION Tuberculosis (TB), particularly its drug-resistant forms (MDR-TB and XDR-TB), continues to pose a significant global health challenge. Despite advances in treatment and diagnosis, the evolving nature of drug resistance in Mycobacterium tuberculosis (MTB) complicates TB eradication efforts. This review delves into the complexities of anti-TB drug resistance, its mechanisms, and implications on healthcare strategies globally. AREAS COVERED We explore the genetic underpinnings of resistance to both first-line and second-line anti-TB drugs, highlighting the role of mutations in key genes. The discussion extends to advanced diagnostic techniques, such as Whole-Genome Sequencing (WGS), CRISPR-based diagnostics and their impact on identifying and managing drug-resistant TB. Additionally, we discuss artificial intelligence applications, current treatment strategies, challenges in managing MDR-TB and XDR-TB, and the global disparities in TB treatment and control, translating to different therapeutic outcomes and have the potential to revolutionize our understanding and management of drug-resistant tuberculosis. EXPERT OPINION The current landscape of anti-TB drug resistance demands an integrated approach combining advanced diagnostics, novel therapeutic strategies, and global collaborative efforts. Future research should focus on understanding polygenic resistance and developing personalized medicine approaches. Policymakers must prioritize equitable access to diagnosis and treatment, enhancing TB control strategies, and support ongoing research and augmented government funding to address this critical public health issue effectively.
Collapse
Affiliation(s)
- Amey Deshpande
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Department of Pharmaceutical Chemistry, Bharati Vidyapeeth's College of Pharmacy, Navi Mumbai, India
| | - Rupali Likhar
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Department of Pharmaceutical Chemistry, LSHGCT's Gahlot Institute of Pharmacy, Navi Mumbai, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Abdelwahab Omri
- The Novel Drug & Vaccine Delivery Systems Facility, Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| |
Collapse
|
3
|
Chen JX, Dong HM, Cai YX, Tian LX, Yang ZC. Synthesis of narrow-spectrum anti-mycobacterial molecules without effect on the diversity of gut microbiota in mice based on the structure of rifampicin. Bioorg Chem 2024; 146:107282. [PMID: 38537334 DOI: 10.1016/j.bioorg.2024.107282] [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: 01/19/2024] [Revised: 02/25/2024] [Accepted: 03/10/2024] [Indexed: 04/13/2024]
Abstract
Rifampicin (RIF) is a broad-spectrum antimicrobial agent that is also a first-line drug for treating tuberculosis (TB). Based on the naphthyl ring structure of RIF this study synthesized 16 narrow-spectrum antimicrobial molecules that were specifically anti-Mycobacterium tuberculosis (Mtb). The most potent candidate was 2-((6-hydroxynaphthalen-2-yl) methylene) hydrazine-1-carbothioamide (compound 3c) with minimum inhibitory concentration (MIC) of 1 μg/mL against Mtb. Synergistic anti-Mtb test indicated that none of the combinations of 3c with the major anti-TB drugs are antagonistic. Consistent with RIF, compound 3c induced large amounts of reactive oxygen radicals (ROS) in the cells of Mtb. The killing kinetics of compound 3c and RIF are very similar. Furthermore, molecular docking showed that compound 3c was able to access the RIF binding pocket of the β subunit of Mtb RNA polymerase (RNAP). Experiments in mice showed that compound 3c increased the variety of intestinal flora in mice, while RIF significantly decreased the diversity of intestinal flora in mice. In addition, compound 3c is non-toxic to animal cells with a selection index (SI) much more than 10. The evidence from this study suggests that the further development of 3c could contribute to the development of novel drug for TB treatment.
Collapse
Affiliation(s)
- Jun-Xian Chen
- College of Pharmacy, Guizhou University, Guiyang 550025, China
| | - Hong-Mei Dong
- College of Pharmacy, Guizhou University, Guiyang 550025, China
| | - Yu-Xiang Cai
- College of Pharmacy, Guizhou University, Guiyang 550025, China
| | - Li-Xia Tian
- College of Pharmacy, Guizhou University, Guiyang 550025, China
| | - Zai-Chang Yang
- College of Pharmacy, Guizhou University, Guiyang 550025, China; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China.
| |
Collapse
|
4
|
Kumar G, C A. Natural products and their analogues acting against Mycobacterium tuberculosis: A recent update. Drug Dev Res 2023; 84:779-804. [PMID: 37086027 DOI: 10.1002/ddr.22063] [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: 11/25/2022] [Revised: 02/28/2023] [Accepted: 04/01/2023] [Indexed: 04/23/2023]
Abstract
Tuberculosis (TB) remains one of the deadliest infectious diseases caused by Mycobacterium tuberculosis (M.tb). It is responsible for significant causes of mortality and morbidity worldwide. M.tb possesses robust defense mechanisms against most antibiotic drugs and host responses due to their complex cell membranes with unique lipid molecules. Thus, the efficacy of existing front-line drugs is diminishing, and new and recurring cases of TB arising from multidrug-resistant M.tb are increasing. TB begs the scientific community to explore novel therapeutic avenues. A precise knowledge of the compounds with their mode of action could aid in developing new anti-TB agents that can kill latent and actively multiplying M.tb. This can help in the shortening of the anti-TB regimen and can improve the outcome of treatment strategies. Natural products have contributed several antibiotics for TB treatment. The sources of anti-TB drugs/inhibitors discussed in this work are target-based identification/cell-based and phenotypic screening from natural products. Some of the recently identified natural products derived leads have reached clinical stages of TB drug development, which include rifapentine, CPZEN-45, spectinamide-1599 and 1810. We believe these anti-TB agents could emerge as superior therapeutic compounds to treat TB over known Food and Drug Administration drugs.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Telangana, India
| | - Amrutha C
- Department of Natural Products, Chemical Sciences, National Institute of Pharmaceutical Education and Research-Hyderabad, Hyderabad, Telangana, India
| |
Collapse
|
5
|
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: 70] [Impact Index Per Article: 23.3] [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
|
6
|
Oliveira MEFAG, Silva YJA, Azevedo LA, Linhares LA, Montenegro LML, Alves S, Amorim RVS. Antimycobacterial compound of chitosan and ethambutol: ultrastructural biological evaluation in vitro against Mycobacterium tuberculosis. Appl Microbiol Biotechnol 2021; 105:9167-9179. [PMID: 34841463 DOI: 10.1007/s00253-021-11690-4] [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: 04/23/2021] [Revised: 10/14/2021] [Accepted: 11/08/2021] [Indexed: 11/26/2022]
Abstract
Chitosan (CS) is a promising biopolymer and has been tested as a complement to the action and compensation of toxicity presented by anti-tuberculosis drugs. The present work studied the adjuvant effect of CS with the drug ethambutol (EMB) as a compound (CS-EMB), to explore its antimicrobial and cytotoxic activity, using transmission electron microscopy (TEM), to examine ultracellular changes that represent possible antimycobacterial action of CS on Mycobacterium tuberculosis (Mtb). Antimycobacterial activities were tested against reference strains Mtb ATCC® H37Rv and multidrug resistant (MDR). In vitro cytotoxicity tests were performed on Raw 264.7. For the studied compounds, morphological, ultrastructural, and physical-chemical analyses were performed. Drug-polymer interactions that occur through the H bridges were confirmed by physical-chemical analyses. The CS-EMB compound is stable at pHs of 6.5-7.5, allowing its release at physiological pH. The antibacterial activity (minimum inhibitory concentration) of the CS-EMB compound was 50% greater than that of the EMB in the H37Rv and MDR strains and the ultrastructural changes in the bacilli observed by TEM proved that the CS-EMB compound has a bactericidal action, allowing it to break down the Mtb cell wall. The cytotoxicity of CS-EMB was higher than that of isolated EMB, IC50 279, and 176 μg/mL, respectively. It is concluded that CS-EMB forms a promising composite against strains Mtb H37Rv and multidrug resistant (MDR-TB).Key points• Our study will be the first to observe ultrastructurally the effects of the CS-EMB compound on Mtb cells.• CS-EMB antimicrobial activity in a multidrug-resistant clinical strain.• The CS-EMB compound has promising potential for the development of a new drug to fight tuberculosis.
Collapse
Affiliation(s)
- M E F A G Oliveira
- Programa de Pós-Graduação Em Morfotecnologia, Universidade Federal de Pernambuco (UFPE), Recife, PE, 50670-420, Brazil.
| | - Y J A Silva
- Programa de Pós-Graduação Em Ciência de Materiais, Universidade Federal de Pernambuco (UFPE), Recife, PE, 50740-560, Brazil
| | - L A Azevedo
- Programa de Pós-Graduação Em Ciência de Materiais, Universidade Federal de Pernambuco (UFPE), Recife, PE, 50740-560, Brazil
| | - L A Linhares
- Instituto Aggeu Magalhães/Fundação Oswaldo Cruz (IAM/FIOCRUZ), 50740-465, Recife-PE, Brazil
| | - L M L Montenegro
- Instituto Aggeu Magalhães/Fundação Oswaldo Cruz (IAM/FIOCRUZ), 50740-465, Recife-PE, Brazil
| | - S Alves
- Departamento de Química Fundamental (dQF), Universidade Federal de Pernambuco (UFPE), Recife, PE, 50740-560, Brazil
| | - R V S Amorim
- Departamento de Histologia E Embriologia (DHE-CB), Universidade Federal de Pernambuco (UFPE), Recife, PE, 50670-420, Brazil
| |
Collapse
|
7
|
Advances in the application of 1,2,4-triazole-containing hybrids as anti-tuberculosis agents. Future Med Chem 2021; 13:2107-2124. [PMID: 34698509 DOI: 10.4155/fmc-2020-0295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tuberculosis is a deadly communicable disease caused by the bacillus Mycobacterium tuberculosis (MTB), and pulmonary tuberculosis accounts for over 80% of the total cases. The 1,2,4-triazole is a privileged structure in the discovery of new drugs, and its derivatives act on various targets in MTB. In particular, 1,2,4-triazole hybrids can not only exert dual or multiple antitubercular mechanisms of action but also have the potential to enhance efficacy and reduce side effects. The present work aims to summarize the current status of 1,2,4-triazole hybrids as potential antitubercular agents, covering articles published between 2010 and 2020, to aid the further rational design of novel potential drug candidates endowed with higher efficacy, better compliance and fewer side effects.
Collapse
|
8
|
Walunj Y, Shinde A, Borde K, Abhale Y, Bobade V, Mhaske PC. Synthesis, Anticancer and Antimicrobial Screening of New Naphthalenyl-Thiazole and Quinolinyl-Thiazole. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1991963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Yogesh Walunj
- Post-Graduate Department of Chemistry, S. P. Mandali’s Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University), Pune, India
- Department of Chemistry, Hutatma Rajguru College (Affiliated to Savitribai Phule Pune University), Khed, Pune, India
| | - Abhijit Shinde
- Post-Graduate Department of Chemistry, S. P. Mandali’s Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University), Pune, India
| | - Krishna Borde
- Post-Graduate Department of Chemistry, S. P. Mandali’s Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University), Pune, India
| | - Yogita Abhale
- Department of Chemistry, Government College, Daman, UT–DNH & DD (Affiliated to Veer Narmad South Gujarat University), India
| | - Vivek Bobade
- Post-Graduate Department of Chemistry, H. P. T. Arts and R. Y. K. Science College (Affiliated to Savitribai Phule Pune University), Nashik, India
| | - Pravin C. Mhaske
- Post-Graduate Department of Chemistry, S. P. Mandali’s Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University), Pune, India
| |
Collapse
|
9
|
Thakare P, Shinde A, Dakhane S, Chavan A, Bobade VD, Mhaske PC. Synthesis and biological evaluation of novel 4‐(6‐substituted quinolin‐4‐yl)‐
N
‐aryl thiazol‐2‐amine derivatives as potential antimicrobial agents. J Heterocycl Chem 2021. [DOI: 10.1002/jhet.4317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Prashant Thakare
- Department of Chemistry S. P. Mandali's Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University) Pune India
| | - Abhijit Shinde
- Department of Chemistry S. P. Mandali's Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University) Pune India
| | - Sagar Dakhane
- Department of Chemistry Abasaheb Garware College (Affiliated to Savitribai Phule Pune University) Pune India
| | - Abhijit Chavan
- Department of Chemistry S. P. Mandali's Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University) Pune India
| | - Vivek D. Bobade
- Department of Chemistry H. P. T. Arts and R. Y. K. Science College (Affiliated to Savitribai Phule Pune University) Nashik India
| | - Pravin C. Mhaske
- Department of Chemistry S. P. Mandali's Sir Parashurambhau College (Affiliated to Savitribai Phule Pune University) Pune India
| |
Collapse
|
10
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
11
|
|
12
|
Triazole-containing hybrids with anti- Mycobacterium tuberculosis potential - Part I: 1,2,3-Triazole. Future Med Chem 2021; 13:643-662. [PMID: 33619989 DOI: 10.4155/fmc-2020-0301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Tuberculosis regimens currently applied in clinical practice require months of multidrug therapy, which imposes a major challenge of patient compliance and drug resistance development. Moreover, because of the increasing emergence of hard-to-treat tuberculosis, this disease continues to be a significant threat to the human population. 1,2,3-triazole as a privileged structure has been widely used as an effective template for drug discovery, and 1,2,3-triazole-containing hybrids that can simultaneously act on dual or multiple targets in Mycobacterium tuberculosis have the potential to circumvent drug resistance, enhance efficacy, reduce side effects and improve pharmacokinetic as well as pharmacodynamic profiles. Thus, 1,2,3-triazole-containing hybrids are useful scaffolds for the development of antitubercular agents. This review aims to highlight recent advances of 1,2,3-triazole-containing hybrids with potential activity against various forms of M. tuberculosis, covering articles published between 2015 and 2020. The structure-activity relationship and the mechanism of action are also discussed to facilitate further rational design of more effective drug candidates.
Collapse
|
13
|
Chang M, Mahasenan KV, Hermoso JA, Mobashery S. Unconventional Antibacterials and Adjuvants. Acc Chem Res 2021; 54:917-929. [PMID: 33512995 DOI: 10.1021/acs.accounts.0c00776] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The need for new classes of antibacterials is genuine in light of the dearth of clinical options for the treatment of bacterial infections. The prodigious discoveries of antibiotics during the 1940s to 1970s, a period wistfully referred to as the Golden Age of Antibiotics, have not kept up in the face of emergence of resistant bacteria in the past few decades. There has been a renewed interest in old drugs, the repurposing of the existing antibiotics and pairing of synergistic antibiotics or of an antibiotic with an adjuvant. Notwithstanding, discoveries of novel classes of these life-saving drugs have become increasingly difficult, calling for new paradigms. We describe, herein, three strategies from our laboratories toward discoveries of new antibacterials and adjuvants using computational and multidisciplinary experimental methods. One approach targets penicillin-binding proteins (PBPs), biosynthetic enzymes of cell-wall peptidoglycan, for discoveries of non-β-lactam inhibitors. Oxadiazoles and quinazolinones emerged as two structural classes out of these efforts. Several hundred analogs of these two classes of antibiotics have been synthesized and fully characterized in our laboratories. A second approach ventures into inhibition of allosteric regulation of cell-wall biosynthesis. The mechanistic details of allosteric regulation of PBP2a of Staphylococcus aureus, discovered in our laboratories, is outlined. The allosteric site in this protein is at 60 Å distance to the active site, whereby ligand binding at the former makes access to the latter by the substrate possible. We have documented that both quinazolinones and ceftaroline, a fifth-generation cephalosporin, bind to the allosteric site in manifestation of the antibacterial activity. Attempts at inhibition of the regulatory phosphorylation events identified three classes of antibacterial adjuvants and one class of antibacterials, the picolinamides. The chemical structures for these hits went through diversification by synthesis of hundreds of analogs. These analogs were characterized in various assays for identification of leads with adjuvant and antibacterial activities. Furthermore, we revisited the mechanism of bulgecins, a class of adjuvants discovered and abandoned in the 1980s. These compounds potentiate the activities of β-lactam antibiotics by the formation of bulges at the sites of septum formation during bacterial replication, which are points of structural weakness in the envelope. These bulges experience rupture, which leads to bacterial death. Bulgecin A inhibits the lytic transglycosylase Slt of Pseudomonas aeruginosa as a likely transition-state mimetic for its turnover of the cell-wall peptidoglycan. Once damage to cell wall is inflicted by a β-lactam antibiotic, the function of Slt is to repair the damage. When Slt is inhibited by bulgecin A, the organism cannot cope with it and would undergo rapid lysis. Bulgecin A is an effective adjuvant of β-lactam antibiotics. These discoveries of small-molecule classes of antibacterials or of adjuvants to antibacterials hold promise in strategies for treatment of bacterial infections.
Collapse
Affiliation(s)
- Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, McCourtney Hall, Notre Dame Indiana 46556, United States
| | - Kiran V. Mahasenan
- Department of Chemistry and Biochemistry, University of Notre Dame, McCourtney Hall, Notre Dame Indiana 46556, United States
| | - Juan A. Hermoso
- Departamento de Cristalografía y Biología Estructural, Instituto de Química-Física “Rocasolano”, CSIC, Serrano 119, 28006-Madrid Spain
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, McCourtney Hall, Notre Dame Indiana 46556, United States
| |
Collapse
|
14
|
Ding Y, Yan H, Chen R, Xiao X, Wang Z, Wang L, Ma Y. Expeditious Approach to Indoloquinazolinones via Double Annulations of o-Aminoacetophenones and Isocyanates. J Org Chem 2021; 86:1448-1455. [PMID: 33373228 DOI: 10.1021/acs.joc.0c02155] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A novel procedure for a one-pot cascade reaction of o-aminoacetophenones and aryl/aliphatic isocyanates catalyzed/oxidized by the [Pd]/[Ag] system was developed. The reaction involves two C-N bond and one C-C bond formations during the double annulation process and the desired indoloquinazolinones and derivatives were afforded up to 81% yields from readily available substrates with a tolerance of a broad variety.
Collapse
Affiliation(s)
- Yuxin Ding
- Institute of Advanced Studies and School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou 318000, P R China.,School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, P R China
| | - Huihui Yan
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, P R China
| | - Rener Chen
- Institute of Advanced Studies and School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou 318000, P R China
| | - Xuqiong Xiao
- Key Laboratory of Organosilicon Chemistry and Material Technology of Ministry of Education, Hangzhou Normal University, No. 2318 Yuhangtang Road Hangzhou 311121, P R China
| | - Zhiming Wang
- Institute of Advanced Studies and School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou 318000, P R China
| | - Lei Wang
- Institute of Advanced Studies and School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou 318000, P R China
| | - Yongmin Ma
- Institute of Advanced Studies and School of Pharmaceutical and Chemical Engineering, Taizhou University, Taizhou 318000, P R China.,School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, P R China
| |
Collapse
|