1
|
Patel RR, Vidyasagar, Singh SK, Singh M. Recent advances in inhibitor development and metabolic targeting in tuberculosis therapy. Microb Pathog 2025; 203:107515. [PMID: 40154850 DOI: 10.1016/j.micpath.2025.107515] [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: 12/27/2024] [Revised: 03/12/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Despite being a preventable and treatable disease, tuberculosis (TB) remained the second leading infectious cause of death globally in 2022, surpassed only by COVID-19. The death rate from TB is influenced by numerous factors that include antibiotic drug resistance, noncompliance with chemotherapy by patients, concurrent infection with the human immunodeficiency virus, delayed diagnosis, varying effectiveness of the Bacille-Calmette-Guerin vaccine, and other factors. Even with the recent advances in our knowledge of Mycobacterium tuberculosis and the accessibility of advanced genomic tools such as proteomics and microarrays, alongside modern methodologies, the pursuit of next-generation inhibitors targeting distinct or multiple molecular pathways remains essential to combat the increasing antimicrobial resistance. Hence, there is an urgent need to identify and develop new drug targets against TB that have unique mechanisms. Novel therapeutic targets might encompass gene products associated with various aspects of mycobacterial biology, such as transcription, metabolism, cell wall formation, persistence, and pathogenesis. This review focuses on the present state of our knowledge and comprehension regarding various inhibitors targeting key metabolic pathways of M. tuberculosis. The discussion encompasses small molecule, synthetic, peptide, natural product and microbial inhibitors and navigates through promising candidates in different phases of clinical development. Additionally, we explore the crucial enzymes and targets involved in metabolic pathways, highlighting their inhibitors. The metabolic pathways explored include nucleotide synthesis, mycolic acid synthesis, peptidoglycan biosynthesis, and energy metabolism. Furthermore, advancements in genetic approaches like CRISPRi and conditional expression systems are discussed, focusing on their role in elucidating gene essentiality and vulnerability in Mycobacteria.
Collapse
Affiliation(s)
- Ritu Raj Patel
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Vidyasagar
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sudhir Kumar Singh
- Virus Research and Diagnostic Laboratory, Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Meenakshi Singh
- Department of Medicinal Chemistry, Faculty of Ayurveda, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
2
|
Bavishi A, Vala H, Thakrar S, Swami S, Sarkar D, Shukla R, Kamdar J, Shah A. Coumarin hybrids: dual-target candidates for future antimicrobial and antitubercular therapies. Future Med Chem 2025:1-12. [PMID: 40353302 DOI: 10.1080/17568919.2025.2504331] [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: 02/23/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
AIMS This study aimed to synthesize, characterize, and evaluate the antimicrobial and antitubercular activities of two novel series of coumarin-based derivatives (Series 5 and Series 9), focusing on their structure-activity relationship (SAR) and molecular docking interactions with key bacterial enzymes. MATERIALS & METHODS Series 5 (5a-5j) and Series 9 (9a-9t) compounds were synthesized and characterized using spectroscopic techniques. Their antimicrobial and antitubercular activities were evaluated against Mycobacterium tuberculosis, Staphylococcus aureus, Bacillus subtilis, and E. coli. IC₅₀ values were determined, and molecular docking studies were conducted to assess binding interactions with M. tuberculosis enoyl-ACP reductase (InhA) and E. coli DNA gyrase B. RESULTS Series 5 compounds exhibited moderate activity, with 5f, 5 g, 5i, and 5j showing notable inhibition. Series 9 derivatives displayed superior dual-target inhibition, with 9t, 9c, 9a, 9b, and 9p achieving >90% inhibition against S. aureus and B. subtilis. The lowest IC₅₀ against M. tuberculosis was observed for 9c (1.50 µg/mL), followed by 9a (2.84 µg/mL) and 9b (2.73 µg/mL). Molecular docking confirmed strong binding interactions, correlating with observed biological activities. CONCLUSIONS Series 9 compounds, particularly 9t, 9c, and 9a, demonstrate high potential as dual-target antimicrobial drug candidates. Further optimization may enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Abhay Bavishi
- Department of Chemistry, Christ College, Rajkot, India
| | - Hardev Vala
- Department of Chemistry, Saurashtra University, Rajkot, India
| | | | - Sagar Swami
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Dhiman Sarkar
- Combi-Chem Bio-Resource Center, Organic Chemistry Division, CSIR-National Chemical Laboratory, Pune, India
| | - Rushit Shukla
- Department of Microbiology, Christ College, Rajkot, India
| | - Jignesh Kamdar
- In Silico Lab, Department of Microbiology, School of Science, RK University, Rajkot, India
| | - Anamik Shah
- Department of Chemistry, Saurashtra University, Rajkot, India
| |
Collapse
|
3
|
Zhou YX, Lun S, Zhang W, Tie FJ, Cai YP, Yang F, Tang J, Bishai WR, Yu LF. Structure-Based Development of 3,4-Fused Tricyclic Benzofuran Derivatives as Polyketide Synthase 13 Inhibitors with Negligible hERG Inhibition. J Med Chem 2025; 68:6312-6327. [PMID: 40045825 DOI: 10.1021/acs.jmedchem.4c02635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Polyketide synthase 13 (Pks13) is vital for synthesizing mycolic acid, which are essential for the survival of Mycobacterium tuberculosis (Mtb). Compounds that target Pks13 hold significant promise for developing new chemical entities for multidrug-resistant TB. The early lead benzofuran-3-carboxamide TAM16, demonstrated robust in vivo efficacy in murine models of tuberculosis infection; however, its further advancement was halted due to the cardiotoxicity associated with hERG inhibition. We implemented a conformational restriction strategy to explore the chemical space of 3,4-fused tricyclic benzofurans and indoles employing a structure-based design approach. Representative compounds were identified as Pks13-TE inhibitors, showing resistance against mutant strains from coumestan-resistant Mtb colonies. Notably, 29 and 30 exhibited potent antitubercular activity against Mtb H37Rv strain (MIC = 0.0156-0.0313 μg/mL), with negligible hERG inhibition (IC50 > 100 μM) suggesting that the 3,4-fused tricyclic benzofurans may present promising scaffold for developing Pks13-TE inhibitors without hERG liability.
Collapse
Affiliation(s)
- Yu-Xin Zhou
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Shichun Lun
- Center for Tuberculosis Research, Department of Medicine, Division of Infectious Disease, Johns Hopkins School of Medicine, Baltimore, Maryland 21231-1044, United States
| | - Wei Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Fa-Jin Tie
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Yan-Peng Cai
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Fan Yang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Jie Tang
- Shanghai Key Laboratory of Green Chemistry and Chemical Process, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - William R Bishai
- Center for Tuberculosis Research, Department of Medicine, Division of Infectious Disease, Johns Hopkins School of Medicine, Baltimore, Maryland 21231-1044, United States
| | - Li-Fang Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| |
Collapse
|
4
|
Liang L, Liu Z, Chen J, Zha Q, Zhou Y, Li J, Hu Y, Chen X, Zhang T, Zhang N. Design and synthesis of Thieno[3, 2-b]pyridinone derivatives exhibiting potent activities against Mycobacterium tuberculosis in vivo by targeting Enoyl-ACP reductase. Eur J Med Chem 2024; 279:116806. [PMID: 39276583 DOI: 10.1016/j.ejmech.2024.116806] [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: 05/30/2024] [Revised: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
In this study, a series of novel thieno [3, 2-b]pyridinone derivatives were designed and synthesized using a scaffold hopping strategy. Six compounds showed potent anti-mycobacterial activity (minimum inhibitory concentration (MIC) ≤ 1 μg/mL) against Mycobacterium tuberculosis (Mtb) UAlRa. Compound 6c displayed good activity against Mtb UAlRv (MIC = 0.5-1 μg/mL). Compounds 6c and 6i also showed activity against Mtb UAlRa in macrophages and exhibited low cytotoxicity against LO-2 cells. The selected compounds displayed a narrow antibacterial spectrum, with no activity against representative Gram-positive, Gram-negative bacteria, as well as fungi. Furthermore, compound 6c demonstrated favorable oral pharmacokinetic properties with a T1/2 value of 47.99 h and exhibited good in vivo activity in an acute mouse model of tuberculosis (TB). The target of compound 6c was identified as a NADH-dependent enoyl-acyl carrier protein reductase (InhA) by genome sequencing of spontaneously compound 6c-resistant Mtb mutants, indicating that compound 6c may not require activation and can directly target InhA. In vitro antimicrobial assays against a recombinant M. smegmatis overexpressing the Mtb-InhA, along with InhA inhibition assays, confirmed that InhA is the target of thieno [3, 2-b]pyridinone derivatives. Overall, this study identified thieno [3, 2-b]pyridinone scaffold as a novel chemotype that is promising for the development of anti-TB agents.
Collapse
Affiliation(s)
- Lihong Liang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Zhiyong Liu
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China; Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Jie Chen
- Jiaxing University Affiliated Hospital, The First Hospital of Jiaxing, Jiaxing, 314001, China
| | - Qin Zha
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Yihuan Zhou
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, 541199, China
| | - Jun Li
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, Shanghai Tech University, Shanghai, 201210, China
| | - Yangbo Hu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Tianyu Zhang
- State Key Laboratory of Respiratory Disease, Guangdong-Hong Kong-Macao Joint Laboratory of Respiratory Infectious Diseases, China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China; Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China; Guangzhou National Laboratory, Guangzhou, 510005, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Niuniu Zhang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, 541199, China.
| |
Collapse
|
5
|
Pan XH, Shi CX, Hou YP, Wang LF, Niu RQ, Guo L. anti-Selective Carboacylation of Alkynes via Photoredox/Nickel Dual Catalysis. Org Lett 2024; 26:9498-9502. [PMID: 39479895 DOI: 10.1021/acs.orglett.4c03439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Here, we report an intermolecular carboacylation of terminal alkynes with tertiary and secondary alkyltrifluoroborates as well as acyl chlorides via photoredox/nickel dual catalysis, affording a varity of stereodefined trisubstituted enones in good to excellent yields and E stereoselectivity, through a radical relay process. This redox-neutral protocol exhibits excellent functional group tolerance, exclusive regio- and stereoselectivity, and broad compatibility with various acyl chlorides and alkyltrifluoroborates.
Collapse
Affiliation(s)
- Xian-Hua Pan
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Chang-Xin Shi
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Ya-Ping Hou
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Li-Fang Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Rui-Qi Niu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Lei Guo
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| |
Collapse
|
6
|
Ieque AL, Palomo CT, Gabriela de Freitas Spanhol V, Fróes da Motta Dacome ML, Júnior do Carmo Pereira J, Candido FC, Caleffi-Ferracioli KR, Dias Siqueira VL, Cardoso RF, Vandresen F, Alves-Olher VG, de Lima Scodro RB. Preclinical tests for salicylhydrazones derivatives to explore their potential for new antituberculosis agents. Tuberculosis (Edinb) 2024; 148:102545. [PMID: 39079220 DOI: 10.1016/j.tube.2024.102545] [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: 03/18/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 09/09/2024]
Abstract
PURPOSE This study target the synthesis of 22 salicylhydrazones derivatives to apply in vitro screening to explore their potential in the search for new anti-TB prototypes drugs. METHODS The minimum inhibitory concentration (MIC) were evaluated against Mycobacterium tuberculosis (Mtb) H37Rv and clinical isolates. Drug combination assay, cytotoxicity assay, ethidium bromide accumulation assay (EtBr) and in silico analysis regarding the absorption, distribution, metabolism, excretion and toxicity (ADMET) and pharmacological properties were also performed. RESULTS Three most promising compounds were selected (10, 11 and 18) to proceed with screening tests. Compound 18 presented the lowest MIC value (0.49 μg/mL) against Mtb H37Rv strain, followed by compounds 11 (3.9 μg/mL) and 10 (7.8 μg/mL). All compounds showed activity against drug susceptible and resistant clinical isolates. Cytotoxicity results were promising for all salicylhydrazones, with SI values up to 4,205 for compound 18. The derivative 10 was the only one that demonstrated a non-promising cytotoxicity scenario for a single cell line. All derivatives showed an additive effect (FICI >0.5 to 4.0) in combination with isoniazid, ethambutol and rifampicin. CONCLUSION All salicylhydrazones showed potential in the screening tests performed in this study and compound 18 stood out due to its activity against susceptible and resistant bacilli at low concentrations and low cytotoxicity.
Collapse
Affiliation(s)
- Andressa Lorena Ieque
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Carolina Trevisolli Palomo
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | | | | | | | | | - Katiany Rizzieri Caleffi-Ferracioli
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Vera Lucia Dias Siqueira
- Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Rosilene Fressatti Cardoso
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Postgraduate Program in Bioscience and Physiopathology, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| | - Fábio Vandresen
- Departament of Chemistry, Federal Technological University of Paraná, Londrina, Paraná, 86036-370, Brazil.
| | | | - Regiane Bertin de Lima Scodro
- Postgraduate Program in Health Sciences, State University of Maringá, Maringá, Paraná, 87020-900, Brazil; Laboratory of Medical Bacteriology, Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, 87020-900, Brazil.
| |
Collapse
|
7
|
Raghu MS, Yogesh Kumar K, Shamala T, Alharti FA, Prashanth MK, Jeon BH. Synthesis, antitubercular profile and molecular docking studies of quinazolinone-based pyridine derivatives against drug-resistant tuberculosis. J Biomol Struct Dyn 2024; 42:3307-3317. [PMID: 37261798 DOI: 10.1080/07391102.2023.2217928] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/03/2023] [Indexed: 06/02/2023]
Abstract
The promising quinazolinone-based pyridine derivatives (4a-j) were synthesized and subsequently tested for their antimycobacterial activities against the various drug-sensitive and drug-resistant Mycobacterium tuberculosis (Mtb) strains to combat infectious diseases and address growing concerns about the devastating effects of tuberculosis (TB). Utilizing 1H NMR, 13C NMR, and mass spectra, the structural and molecular confirmation of the synthesized compounds were deciphered. With minimum inhibitory concentration (MIC) values ranging from 0.31 to 19.13 μM, the results showed that compounds 4e and 4f showed promise anti-TB action against both drug-sensitive and drug-resistant TB strains. To study the cytotoxicity of synthesized molecules, normal Vero and mouse macrophage (RAW264.7) cell lines were utilized. Remarkably, it was revealed that at the highest concentration tested, none of the newly synthesized molecules were toxic to the Vero cell line. The binding patterns of the potent compounds 4b, 4e and 4f in the active site of the mycobacterial membrane protein Large 3 (MmpL3) protein are also revealed by molecular docking studies, which has contributed to the development of a structural rationale for Mtb inhibition. The physicochemical characteristics of the compounds were then predicted using theoretical calculations. Overall, the molecular docking results, physiochemical properties, and observed antimycobacterial activity all point to compound 4e with trifluoromethyl and compound 4f with nitro moiety as potential quinazolinone linked pyridine-based MmpL3 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- M S Raghu
- Department of Chemistry, New Horizon College of Engineering, Bengaluru, India
| | - K Yogesh Kumar
- Department of Chemistry, Faculty of Engineering and Technology, Jain University, Ramanagara, India
| | - T Shamala
- Department of Chemistry, B N M Institute of Technology, Bengaluru, India
| | - Fahad A Alharti
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - M K Prashanth
- Department of Chemistry, B N M Institute of Technology, Bengaluru, India
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
8
|
Singh P, Kumar A, Sharma P, Chugh S, Kumar A, Sharma N, Gupta S, Singh M, Kidwai S, Sankar J, Taneja N, Kumar Y, Dhiman R, Mahajan D, Singh R. Identification and optimization of pyridine carboxamide-based scaffold as a drug lead for Mycobacterium tuberculosis. Antimicrob Agents Chemother 2024; 68:e0076623. [PMID: 38193667 PMCID: PMC10848774 DOI: 10.1128/aac.00766-23] [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: 06/09/2023] [Accepted: 11/10/2023] [Indexed: 01/10/2024] Open
Abstract
New drugs with novel mechanisms of action are urgently needed to tackle the issue of drug-resistant tuberculosis. Here, we have performed phenotypic screening using the Pathogen Box library obtained from the Medicines for Malaria Venture against Mycobacterium tuberculosis in vitro. We have identified a pyridine carboxamide derivative, MMV687254, as a promising hit. This molecule is specifically active against M. tuberculosis and Mycobacterium bovis Bacillus Calmette-Guérin (M. bovis BCG) but inactive against Enterococcus faecalis, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumanii, Pseudomonas aeruginosa, and Escherichia coli pathogens. We demonstrate that MMV687254 inhibits M. tuberculosis growth in liquid cultures in a bacteriostatic manner. Surprisingly, MMV687254 was as active as isoniazid in macrophages and inhibited M. tuberculosis growth in a bactericidal manner. Mechanistic studies revealed that MMV687254 is a prodrug and that its anti-mycobacterial activity requires AmiC-dependent hydrolysis. We further demonstrate that MMV687254 inhibits M. tuberculosis growth in macrophages by inducing autophagy. In the present study, we have also carried out a detailed structure-activity relationship study and identified a promising novel lead candidate. The identified novel series of compounds also showed activity against drug-resistant M. bovis BCG and M. tuberculosis clinical strains. Finally, we demonstrate that in contrast to MMV687254, the lead molecule was able to inhibit M. tuberculosis growth in a chronic mouse model of infection. Taken together, we have identified a novel lead molecule with a dual mechanism of action that can be further optimized to design more potent anti-tubercular agents.
Collapse
Affiliation(s)
- Padam Singh
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Arun Kumar
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Pankaj Sharma
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Saurabh Chugh
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Ashish Kumar
- Department of Life Science, Laboratory of Mycobacterial Immunology, National Institute of Technology, Rourkela, India
| | - Nidhi Sharma
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Sonu Gupta
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Manisha Singh
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Saqib Kidwai
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Jishnu Sankar
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Neha Taneja
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Yashwant Kumar
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Rohan Dhiman
- Department of Life Science, Laboratory of Mycobacterial Immunology, National Institute of Technology, Rourkela, India
| | - Dinesh Mahajan
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Ramandeep Singh
- Translational Health Sciences and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
9
|
Khola S, Kumar S, Bhanwala N, Khatik GL. Polyketide Synthase 13 (Pks13) Inhibition: A Potential Target for New Class of Anti-tubercular Agents. Curr Top Med Chem 2024; 24:2362-2376. [PMID: 39297467 DOI: 10.2174/0115680266322983240906055750] [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: 04/14/2024] [Revised: 07/31/2024] [Accepted: 08/15/2024] [Indexed: 10/29/2024]
Abstract
Tuberculosis is one of the deadly infectious diseases that has resurfaced in multiple/ extensively resistant variants (MDR/XDR), threatening humankind. Today's world has a higher prevalence of tuberculosis (TB) than it has ever had throughout human history. Due to severe adverse effects, the marketed medications are not entirely effective in these forms. So, developing new drugs with a promising target is an immense necessity. Pks13 has emerged as a promising target for the mycobacterium. The concluding step of mycolic acid production involved Pks13, a crucial enzyme that helps form the precursor of mycolic acid via the Claisen-condensation reaction. It has five domains at the active site for targeting the enzyme and is used to test chemical entities for their antitubercular activity. Benzofurans, thiophenes, coumestans, N-phenyl indoles, and β lactones are the ligands that inhibit the Pks13 enzyme, showing potential antitubercular properties.
Collapse
Affiliation(s)
- Sonia Khola
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Sachin Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Neeru Bhanwala
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Gopal L Khatik
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| |
Collapse
|
10
|
Verma A, Naik B, Kumar V, Mishra S, Choudhary M, Khan JM, Gupta AK, Pandey P, Rustagi S, Kakati B, Gupta S. Revolutionizing Tuberculosis Treatment: Uncovering New Drugs and Breakthrough Inhibitors to Combat Drug-Resistant Mycobacterium tuberculosis. ACS Infect Dis 2023; 9:2369-2385. [PMID: 37944023 DOI: 10.1021/acsinfecdis.3c00436] [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] [Indexed: 11/12/2023]
Abstract
Tuberculosis (TB) is a global health threat that causes significant mortality. This review explores chemotherapeutics that target essential processes in Mycobacterium tuberculosis, such as DNA replication, protein synthesis, cell wall formation, energy metabolism, and proteolysis. We emphasize the need for new drugs to treat drug-resistant strains and shorten the treatment duration. Emerging targets and promising inhibitors were identified by examining the intricate biology of TB. This review provides an overview of recent developments in the search for anti-TB drugs with a focus on newly validated targets and inhibitors. We aimed to contribute to efforts to combat TB and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Ankit Verma
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Bindu Naik
- Department of Food Science and Technology, Graphic Era Deemed to be University, Bell Road, Clement Town, Dehradun 248002, Uttarakhand, India
| | - Vijay Kumar
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Sadhna Mishra
- Faculty of Agricultural Sciences, GLA University, Mathura 281406, UP, India
| | - Megha Choudhary
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, Uttarakhand, India
| | - Javed Masood Khan
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, 2460, Riyadh 11451, Saudi Arabia
| | - Arun Kumar Gupta
- Department of Food Science and Technology, Graphic Era Deemed to be University, Bell Road, Clement Town, Dehradun 248002, Uttarakhand, India
| | - Piyush Pandey
- Department of Microbiology, Assam University, Silchur 788011, Assam, India
| | - Sarvesh Rustagi
- Department of Food Technology, UCALS, Uttaranchal University, Dehradun 248007, Uttarakhand, India
| | - Barnali Kakati
- Department of Microbiology, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, U.K., India
| | - Sanjay Gupta
- Himalayan School of Biosciences, Swami Rama Himalayan University, Jolly Grant, Dehradun 248016, Uttarakhand, India
| |
Collapse
|
11
|
Dawood DH, Sayed MM, Tohamy STK, Nossier ES. New Thiophenyl-pyrazolyl-thiazole Hybrids as DHFR Inhibitors: Design, Synthesis, Antimicrobial Evaluation, Molecular Modeling, and Biodistribution Studies. ACS OMEGA 2023; 8:39250-39268. [PMID: 37901585 PMCID: PMC10600881 DOI: 10.1021/acsomega.3c04736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/25/2023] [Indexed: 10/31/2023]
Abstract
The antibiotic resistance problems constitute a considerable threat to human health worldwide; thus, the discovery of new antimicrobial candidates to conquer this issue is an imperative requirement. From this view, new thiophenyl-pyrazolyl-thiazole hybrids 3-10 were synthesized and screened for their antibacterial efficiency versus Gram - and Gram + bacterial strains compared to the reference drug amoxicillin. It was noticed that the new hybrids displayed significant antibacterial efficacy versus Gram - bacteria, especially against Pseudomonas aeruginosa. Also, all the screened candidates demonstrated a noticeable antifungal effect against Candida albicans (MICs = 3.9-125 μg/mL) relative to fluconazole (MIC = 250 μg/mL). Moreover, the new hybrids were investigated for their antituberculosis potency against Mycobacterium tuberculosis (RCMB 010126). Derivatives 4c, 6b, 8b, 9b, and 10b demonstrated prominent antituberculosis efficiency (MICs = 0.12-1.95 μg/mL) compared with the reference drug isoniazid (MIC = 0.12 μg/mL). The latter derivatives were further assessed for their inhibitory potency versus M. tuberculosis DHFR enzyme. The compounds 4c, 6b and 10b presented a remarkable suppression effect with IC50 values of 4.21, 5.70, and 10.59 μM, respectively, compared to that of trimethoprim (IC50 = 6.23 μM). Furthermore, biodistribution profile using radiolabeling way revealed a perceived uptake of 131I-compound 6b into infection induced models. The docking study for the new hybrids 4c, 6b, 8b, 9b and 10b was performed to illustrate the various binding modes with Mtb DHFR enzyme. In silico ADMET studies for the most potent inhibitors 4c, 6b and 10b were also accomplished to predict their pharmacokinetic and physicochemical features.
Collapse
Affiliation(s)
- Dina H. Dawood
- Chemistry
of Natural and Microbial Products Department, Pharmaceutical and Drug
Industries Research Institute, National
Research Centre, 33 El
Bohouth Street, Dokki, Giza 12622, Egypt
| | - Manal M. Sayed
- Labeled
Compounds Department, Hot Labs.center, Egyptian
Atomic Energy Authority (EAEA), P.O.
Box 13759, Cairo, Egypt
| | - Sally T. K. Tohamy
- Department
of Microbiology and Immunology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
| | - Eman S. Nossier
- Department
of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of
Pharmacy (Girls), Al-Azhar University, Cairo 11754, Egypt
- The
National Committee of Drugs, Academy of
Scientific Research and Technology, Cairo 11516, Egypt
| |
Collapse
|
12
|
Arche PDE, Chatterjee S, Talukder MM, Miller JT, Cue JMO, Udamulle Gedara CM, Lord RL, Biewer MC, Cisneros GA, Stefan MC. Regioselective Direct C-H Bond Heteroarylation of Thiazoles Enabled by an Iminopyridine-Based α-Diimine Nickel(II) Complex Evaluated by DFT Studies. J Org Chem 2023; 88:12319-12328. [PMID: 37603582 DOI: 10.1021/acs.joc.3c01021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
Direct C-H bond arylation is a highly effective method for synthesizing arylated heteroaromatics. This method reduces the number of synthetic steps and minimizes the formation of impurities. We report an air- and moisture-stable iminopyridine-based α-diimine nickel(II) complex for direct C5-H bond arylation of thiazole derivatives. Under a low catalyst loading and performing the reactions at lower temperatures (80 °C) under aerobic conditions, we produced mono- and diarylated thiazole units. Competition experiments and density functional theory calculations revealed that the mechanism of C-H activation in 4-methylthiazole involves an electrophilic aromatic substitution.
Collapse
Affiliation(s)
- Phillip Damien E Arche
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Shubham Chatterjee
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Md Muktadir Talukder
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Justin T Miller
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - John Michael O Cue
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Chinthaka M Udamulle Gedara
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Richard L Lord
- Department of Chemistry, Grand Valley State University, Allendale, Michigan 49401, United States
| | - Michael C Biewer
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - G Andrés Cisneros
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
- Department of Physics, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Mihaela C Stefan
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
13
|
Singh P, Rawat S, Agrahari AK, Singh M, Chugh S, Gurcha S, Singh A, Abrahams K, Besra GS, Asthana S, Rawat DS, Singh R. NSC19723, a Thiacetazone-Like Benzaldehyde Thiosemicarbazone Improves the Efficacy of TB Drugs In Vitro and In Vivo. Microbiol Spectr 2022; 10:e0259222. [PMID: 36314972 PMCID: PMC9769743 DOI: 10.1128/spectrum.02592-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/31/2022] [Indexed: 12/24/2022] Open
Abstract
The complexity and duration of tuberculosis (TB) treatment contributes to the emergence of drug resistant tuberculosis (DR-TB) and drug-associated side effects. Alternate chemotherapeutic agents are needed to shorten the time and improve efficacy of current treatment. In this study, we have assessed the antitubercular activity of NSC19723, a benzaldehyde thiosemicarbazone molecule. NSC19723 is structurally similar to thiacetazone (TAC), a second-line anti-TB drug used to treat individuals with DR-TB. NSC19723 displayed better MIC values than TAC against Mycobacterium tuberculosis and Mycobacterium bovis BCG. In our checkerboard experiments, NSC19723 displayed better profiles than TAC in combination with known first-line and recently approved drugs. Mechanistic studies revealed that NSC19723 inhibits mycolic acid biosynthesis by targeting the HadABC complex. Computational studies revealed that the binding pocket of HadAB is similarly occupied by NSC19723 and TAC. NSC19723 also improved the efficacy of isoniazid in macrophages and mouse models of infection. Cumulatively, we have identified a benzaldehyde thiosemicarbazone scaffold that improved the activity of TB drugs in liquid cultures, macrophages, and mice. IMPORTANCE Mycobacterium tuberculosis, the causative agent of TB is among the leading causes of death among infectious diseases in humans. This situation has worsened due to the failure of BCG vaccines and the increased number of cases with HIV-TB coinfections and drug-resistant strains. Another challenge in the field is the lengthy duration of therapy for drug-sensitive and -resistant TB. Here, we have deciphered the mechanism of action of NSC19723, benzaldehyde thiosemicarbazone. We show that NSC19723 targets HadABC complex and inhibits mycolic acid biosynthesis. We also show that NSC19723 enhances the activity of known drugs in liquid cultures, macrophages, and mice. We have also performed molecular docking studies to identify the interacting residues of HadAB with NSC19723. Taken together, we demonstrate that NSC19723, a benzaldehyde thiosemicarbazone, has better antitubercular activity than thiacetazone.
Collapse
Affiliation(s)
- Padam Singh
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Srishti Rawat
- Department of Chemistry, University of Delhi, Delhi, India
| | - Ashish Kumar Agrahari
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Manisha Singh
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Saurabh Chugh
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Sudagar Gurcha
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Albel Singh
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Katherine Abrahams
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Gurdyal S. Besra
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Shailendra Asthana
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| | - Diwan S. Rawat
- Department of Chemistry, University of Delhi, Delhi, India
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
14
|
Recent advancements and developments in search of anti-tuberculosis agents: A quinquennial update and future directions. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.131473] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Mittal L, Tonk RK, Awasthi A, Asthana S. Targeting cryptic-orthosteric site of PD-L1 for inhibitor identification using structure-guided approach. Arch Biochem Biophys 2021; 713:109059. [PMID: 34673001 DOI: 10.1016/j.abb.2021.109059] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/30/2021] [Accepted: 10/10/2021] [Indexed: 12/23/2022]
Abstract
Approved mAbs that block the protein-protein interaction (PPI) interface of the PD-1/PD-L1 immune checkpoint axis have led to significant improvements in cancer treatment. Despite having drawbacks of mAbs only few a compounds are reported till date against this axis. Inhibiting PPIs using small molecules has emerged as a significant therapeutic opportunity, demanding for the identification of drug-like molecules at an accelerated pace under the hit-to-lead campaigns. Due to the PD-L1's cross-talk with PD-1/CD80 and its overexpression on cancer cells, as well as the availability of its crystal structures with small molecules, it is an enticing therapeutic target for structure-assisted small molecule design. Furthermore, the selection of chemical databases enriched with focused designing for PPI interfaces is crucial. Therefore, in this study we have utilized the Asinex signature library for structure-assisted virtual screening to find the potential PD-L1 inhibitors by targeting the cryptic PD-L1 interface, followed by induced fit docking for pose refinements in the pocket. The obtained hits were then subjected to interaction fingerprinting and ligand-based drug-likeness investigations in order to evaluate and analyze their drug-like qualities (ADME). Twelve compounds qualified for molecular dynamics simulations, followed by thermodynamic calculations for evaluation of their stability and energetics inside the pocket. Two novel compounds with different chemical moieties have been identified that are consistent throughout the simulation, mimicking the interactions and binding energies with BMS-1166. These compounds appear as potential therapeutic candidates to be explored experimentally, thereby paving the way for the development of novel leads as immunomodulators.
Collapse
Affiliation(s)
- Lovika Mittal
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India; Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Rajiv K Tonk
- Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Amit Awasthi
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Shailendra Asthana
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India.
| |
Collapse
|