1
|
Zaware N, Rasal N, Lambate V, Jagtap S. Biological evaluation of newly synthesized α-benzil monoxime thiocarbohydrazide derivatives as an antimicrobial and anticancer agent: In vitro screening and ADMET predictions. Bioorg Med Chem Lett 2025; 118:130079. [PMID: 39710140 DOI: 10.1016/j.bmcl.2024.130079] [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: 06/11/2024] [Revised: 12/01/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The current comprehensive study showcases a meticulous synthesis of novel class of α-benzilmonoxime thiocarbohydrazide (BMOTC) derivatives, and manifesting their multifaceted potential as antibacterial, antifungal, and anticancer agents. The synthesis of target compounds was performed in three phases using literature methods. In the first step, benzilmonoxime is synthesized using benzil and hydroxyl amine hydrochloride, followed by benzilmonoxime imine using thiocarbohydrazide. The final stage involves combining BMOTC imine with various aldehydes and ketones. The antibacterial and antifungal activities of the synthesized derivatives against five bacterial panels, both Gram-positive and Gram-negative, and one fungal pathogen have been screened. Twelve of the twenty-four synthetic derivatives showed noteworthy activity; eight derivatives exhibited growth inhibition (GI) >73 % against Acinetobacter baumannii, two exhibited GI >95 % against Escherichia coli, and two exhibited GI >93 % against Candida albicans at concentration 32 μg/mL. Further assessment revealed that two derivatives 5v and 5w, exhibited negligible cytotoxicity towards human embryonic kidney cells (HK-293) and human red blood cells (RBC), signifying their promising safety profile at concentration 32 μg/mL (GI against Candida albicans - 97.51 % and 93.71 % respectively). The synthesized compounds were subjected to in vitro cytostatic activity, where a rigorous scrutiny against a diverse panel of NCI 60 cancer cell lines representing various malignancies was carried out. A total of eleven compounds emerged as promising candidates, demonstrating significant growth percent (GP) at a concentration of 10 µM. Notably, compounds 5d, 5h, and 5x, turned up as standout performers, exhibiting potent anticancer activity across multiple cancer types, including colon, CNS, melanoma, and breast cancers. Of particular interest, compound 5d displayed notable antiproliferative effects against leukemia cancer cell lines RPMI-8226 & SR, while maintaining non-cytotoxicity against the same. Compound 5h showcased activity against ovarian, non-small cell lung, and prostate cancers, without inducing cytotoxic effects. Compound 5x demonstrated remarkable anticancer activity against leukemia and breast cancer cell lines, further bolstered by its non-cytotoxic nature. A compelling aspect of this study is the comparative analysis with the established drug molecule sunitinib, revealing that compounds 5d, 5h, and 5x exhibit superior potency. These findings not only highlight the therapeutic potential of the BMOTC derivatives but also underscore their viability as promising candidates for future drug development endeavours. This study serves as a pivotal step towards harnessing the untapped therapeutic potential of BMOTC derivatives in combating microbial infections and advancing cancer therapy.
Collapse
Affiliation(s)
- Navnath Zaware
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India
| | - Nishant Rasal
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India
| | - Vinayak Lambate
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India
| | - Sangeeta Jagtap
- Department of Chemistry, PDEA's Baburaoji Gholap College, Sangvi, Pune 27, India.
| |
Collapse
|
2
|
Tailor NK, Deswal G, Guarve K, Grewal AS. Development of Mycobacterium tuberculosis Enoyl Acyl Reductase (InhA) Inhibitors: A Mini-Review. Mini Rev Med Chem 2025; 25:219-233. [PMID: 39301902 DOI: 10.2174/0113895575309785240902102421] [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: 03/12/2024] [Revised: 07/04/2024] [Accepted: 07/27/2024] [Indexed: 09/22/2024]
Abstract
This review article delves into the critical role of Enoyl acyl carrier protein reductase (InhA; ENR), a vital enzyme in the NADH-dependent acyl carrier protein reductase family, emphasizing its significance in fatty acid synthesis and, more specifically, the biosynthesis of mycolic acid. The primary objective of this literature review is to elucidate diverse scaffolds and their developmental progression targeting InhA inhibition, thereby disrupting mycolic acid biosynthesis. Various scaffolds, including thiourea, piperazine, thiadiazole, triazole, quinazoline, benzamide, rhodanine, benzoxazole, and pyridine, have been systematically explored for their potential as InhA inhibitors. Noteworthy findings highlight thiadiazole and triazole derivatives, demonstrating promising IC50 values within the nanomolar concentration range. The review offers comprehensive insights into InhA's structure, structure-activity relationships, and a detailed overview of distinct scaffolds as effective inhibitors of InhA.
Collapse
Affiliation(s)
- Navin Kumar Tailor
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Geeta Deswal
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, 135001, Haryana, India
| | - Kumar Guarve
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, 135001, Haryana, India
| | - Ajmer Singh Grewal
- Department of Pharmacy, Guru Gobind Singh College of Pharmacy, Yamuna Nagar, 135001, Haryana, India
| |
Collapse
|
3
|
AlRashidi E, Ghannay S, Albadri AE, Abid M, Kadri A, Aouadi K. Design, synthesis, biological evaluation, kinetic studies and molecular modeling of imidazo-isoxazole derivatives targeting both α-amylase and α-glucosidase inhibitors. Heliyon 2024; 10:e38376. [PMID: 39640664 PMCID: PMC11619978 DOI: 10.1016/j.heliyon.2024.e38376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 12/07/2024] Open
Abstract
Herein, a novel set of imidazo-isoxazole derivatives containing thiourea and urea scaffolds were synthesized, characterized (1H NMR, 13C NMR, and elemental analysis). These compounds were biologically evaluated for their α-amylase and α-glucosidase inhibitory activity, identifying 5f as the most active (IC50 26.67 ± 1.25 μM and 39.12 ± 1.83 μM against α-amylase α-glucosidase, respectively), better than the standard, acarbose. Enzymatic kinetic results showed that 5f and acarbose complete competitive type inhibitors. The structure-activity relationship (SAR) demonstrated that undergoing substitutions on R1 and R2 groups attached to the thiourea/urea moiety chains controlled the activity. Besides, in-silico ADMET study showed that almost title compounds exhibited satisfactory pharmacokinetic properties. In molecular docking study, the top performing compound (5f) exhibited higher binding energies (-5.501 and -6.414 kcal/mol, respectively) showing crucial interactions and that snuggly fit in their active site. To shed light on their mechanism of action, molecular dynamic (MD) simulations approach executed at 100 ns duration authenticated the high stability of 5f-1B2Y and 5f-3A4A complexes. The results of this investigation disclosed that compound 5f may serve as a potential lead, accomplished with in vivo studies, for the management of diabetes.
Collapse
Affiliation(s)
- Etab AlRashidi
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Siwar Ghannay
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Abuzar E.A.E. Albadri
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Majdi Abid
- Department of Chemistry, College of Science, Jouf University, P.O. Box 2014, Sakaka, Aljouf, Kingdom of Saudi Arabia
| | - Adel Kadri
- Department of Chemistry, Faculty of Science, Al-Baha University, Al-Baha, 65431, Kingdom of Saudi Arabia
- Faculty of Science of Sfax, Department of Chemistry, University of Sfax, B.P. 1171, 3000, Sfax, Tunisia
| | - Kaiss Aouadi
- Department of Chemistry, College of Science, Qassim University, Buraidah, 51452, Saudi Arabia
| |
Collapse
|
4
|
Gavadia R, Rasgania J, Sahu N, Varma-Basil M, Chauhan V, Kumar S, Mor S, Singh D, Jakhar K. Design and Synthesis of Isatin-Tagged Isoniazid Conjugates with Cogent Antituberculosis and Radical Quenching Competence: In-vitro and In-silico Evaluations. Chem Biodivers 2024; 21:e202400765. [PMID: 39024129 DOI: 10.1002/cbdv.202400765] [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: 03/26/2024] [Revised: 06/07/2024] [Accepted: 07/18/2024] [Indexed: 07/20/2024]
Abstract
In pursuit of potential chemotherapeutic alternates to combat severe tuberculosis infections, novel heterocyclic templates derived from clinically approved anti-TB drug isoniazid and isatin have been synthesized that demonstrate potent inhibitory action against Mycobacterium tuberculosis, and compound 4i with nitrophenyl motif exhibited the highest anti-TB efficacy with a MIC value of 2.54 μM/ml. Notably, the same nitro analog 4i shows the best antioxidant efficacy among all the synthesized compounds with an IC50 value of 37.37 μg/ml, suggesting a synergistic influence of antioxidant proficiency on the anti-TB action. The titled compounds exhibit explicit binding affinity with the InhA receptor. The befitting biochemical reactivity and near-appropriate pharmacokinetic proficiency of the isoniazid conjugates is reflected in the density functional theory (DFT) studies and ADMET screening. The remarkable anti-TB action of the isoniazid cognates with marked radical quenching ability may serve as a base for developing multi-target medications to confront drug-resistant TB pathogens.
Collapse
Affiliation(s)
- Renu Gavadia
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Jyoti Rasgania
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Neetu Sahu
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Mandira Varma-Basil
- Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, 110007, India
| | - Varsha Chauhan
- Department of Microbiology, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi, 110007, India
- Department of Microbiology, M. D. University, Rohtak, Haryana, 124001, India
| | - Sanjay Kumar
- Department of Microbiology, M. D. University, Rohtak, Haryana, 124001, India
| | - Satbir Mor
- Department of Chemistry, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, 125001, India
| | - Devender Singh
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| | - Komal Jakhar
- Department of Chemistry, M. D. University, Rohtak, Haryana, 124001, India
| |
Collapse
|
5
|
Kassem AF, Sabt A, Korycka-Machala M, Shaldam MA, Kawka M, Dziadek B, Kuzioła M, Dziadek J, Batran RZ. New coumarin linked thiazole derivatives as antimycobacterial agents: Design, synthesis, enoyl acyl carrier protein reductase (InhA) inhibition and molecular modeling. Bioorg Chem 2024; 150:107511. [PMID: 38870705 DOI: 10.1016/j.bioorg.2024.107511] [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: 03/28/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Tuberculosis is a global serious problem that imposes major health, economic and social challenges worldwide. The search for new antitubercular drugs is extremely important which could be achieved via inhibition of different druggable targets. Mycobacterium tuberculosis enoyl acyl carrier protein reductase (InhA) enzyme is essential for the survival of M. tuberculosis. In this investigation, a series of coumarin based thiazole derivatives was synthesized relying on a molecular hybridization approach and was assessed against thewild typeMtb H37Rv and its mutant strain (ΔkatG) via inhibiting InhA enzyme. Among the synthesized derivatives, compounds 2b, 3i and 3j were the most potent against wild type M. tuberculosis with MIC values ranging from 6 to 8 μg/ mL and displayed low cytotoxicity towards mouse fibroblasts at concentrations 8-13 times higher than the MIC values. The three hybrids could also inhibit the growth of ΔkatGmutant strain which is resistant to isoniazid (INH). Compounds 2b and 3j were able to inhibit the growth of mycobacteria inside human macrophages, indicating their ability to penetrate human professional phagocytes. The two derivatives significantly suppress mycobacterial biofilm formation by 10-15 %. The promising target compounds were also assessed for their inhibitory effect against InhA and showed potent effectiveness with IC50 values of 0.737 and 1.494 µM, respectively. Molecular docking studies revealed that the tested compounds occupied the active site of InhA in contact with the NAD+ molecule. The 4-phenylcoumarin aromatic system showed binding interactions within the hydrophobic pocket of the active site. Furthermore, H-bond formation and π -π stacking interactions were also recorded for the promising derivatives.
Collapse
Affiliation(s)
- Asmaa F Kassem
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Ahmed Sabt
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Małgorzata Korycka-Machala
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Malwina Kawka
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Bożena Dziadek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Magdalena Kuzioła
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, Lodz, Poland
| | - Jarosław Dziadek
- Laboratory of Genetics and Physiology of Mycobacterium, Institute of Medical Biology of the Polish Academy of Sciences, Lodz, Poland.
| | - Rasha Z Batran
- Chemistry of Natural Compounds Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt.
| |
Collapse
|
6
|
Rana P, Parupalli R, Akhir A, Saxena D, Maitra R, Imran M, Malik P, Mahammad Ghouse S, Joshi SV, Srikanth D, Madhavi YV, Dasgupta A, Chopra S, Nanduri S. Synthesis and biological evaluation of new naphthalimide-thiourea derivatives as potent antimicrobial agents active against multidrug-resistant Staphylococcus aureus and Mycobacterium tuberculosis. RSC Med Chem 2024; 15:1381-1391. [PMID: 38665829 PMCID: PMC11042119 DOI: 10.1039/d4md00062e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/21/2024] [Indexed: 04/28/2024] Open
Abstract
The emergence of antibiotic resistance to S. aureus and M. tuberculosis, particularly MRSA, VRSA, and drug-resistant tuberculosis, poses a serious threat to human health. Towards discovering new antibacterial agents, we designed and synthesized a series of new naphthalimide-thiourea derivatives and evaluated them against a panel of bacterial strains consisting of E. coli, S. aureus, K. pneumoniae, P. aeruginosa, A. baumannii and various mycobacterial pathogens. Compounds 4a, 4l, 4m, 4n, 4q, 9f, 9l, 13a, 13d, 13e, 17a, 17b, 17c, 17d, and 17e demonstrated potent antibacterial activity against S. aureus with MIC 0.03-8 μg mL-1. In addition, these compounds have also exhibited potent inhibition against MDR strains of S. aureus, including VRSA with MICs 0.06-4 μg mL-1. Compounds 4h, 4j, 4l, 4m, 4q, 4r, 9a, 9b, 9c, 9d, 9e, 9g, 9h, 9j, 13f and 17e also exhibited good antimycobacterial activity against M. tuberculosis with MIC 2-64 μg mL-1. The cytotoxicity assay using Vero cells revealed that all the compounds were non-toxic and exhibited a favorable selectivity index (SI >40). Time kill kinetics data indicated that compounds exhibited concentration-dependent killing. Furthermore, in silico studies were performed to decipher the possible mechanism of action. Comprehensively, these results highlight the potential of naphthalimide-thiourea derivatives as promising antibacterial agents.
Collapse
Affiliation(s)
- Preeti Rana
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| | - Ramulu Parupalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| | - Abdul Akhir
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
| | - Deepanshi Saxena
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
| | - Rahul Maitra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
| | - Mohmmad Imran
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
| | - Pradip Malik
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
| | - Shaik Mahammad Ghouse
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| | - Swanand Vinayak Joshi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| | - Danaboina Srikanth
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| | - Y V Madhavi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| | - Arunava Dasgupta
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Sidharth Chopra
- Division of Molecular Microbiology and Immunology, CSIR-Central Drug Research Institute Sitapur Road, Sector 10, Janakipuram Extension Lucknow-226031 Uttar Pradesh India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR) Ghaziabad 201002 India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER) Hyderabad Telangana-500037 India
| |
Collapse
|
7
|
Shaaban MM, Teleb M, Ragab HM, Singh M, Elwakil BH, A Heikal L, Sriram D, Mahran MA. The first-in-class pyrazole-based dual InhA-VEGFR inhibitors towards integrated antitubercular host-directed therapy. Bioorg Chem 2024; 145:107179. [PMID: 38367430 DOI: 10.1016/j.bioorg.2024.107179] [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: 12/14/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Abstract
Several facets of the host response to tuberculosis have been tapped for clinical investigation, especially targeting angiogenesis mediated by VEGF signaling from infected macrophages. Herein, we rationalized combining the antiangiogenic effects of VEGFR-2 blockade with direct antitubercular InhA inhibition in single hybrid dual inhibitors as advantageous alternatives to the multidrug regimens. Inspired by expanded triclosans, the ether ligation of triclosan was replaced by rationalized linkers to assemble the VEGFR-2 inhibitors thematic scaffold. Accordingly, new series of 3-(p-chlorophenyl)-1-phenylpyrazole derivatives tethered to substituted ureas and their isosteres were synthesized, evaluated against Mycobacterium tuberculosis virulent cell line H37Rv, and assessed for their InhA inhibitory activities. The urea derivatives 8d and 8g exhibited the most promising antitubercular activity (MIC = 6.25 µg/mL) surpassing triclosan (MIC = 20 µg/mL) with potential InhA inhibition, thus identified as the study hits. Interestingly, both compounds inhibited VEGFR-2 at nanomolar IC50 (15.27 and 24.12 nM, respectively). Docking and molecular dynamics simulations presumed that 8d and 8g could bind to their molecular targets InhA and VEGFR-2 posing essential stable interactions shared by the reference inhibitors triclosan and sorafenib. Finally, practical LogP, Lipinski's parameters and in silico ADMET calculations highlighted their drug-likeness as novel leads in the arsenal against TB.
Collapse
Affiliation(s)
- Marwa M Shaaban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt.
| | - Hanan M Ragab
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Monica Singh
- Tuberculosis Drug Discovery Laboratory, Pharmacy Group, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500 0078, India
| | - Bassma H Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences Technology, Pharos University in Alexandria, Alexandria, Egypt
| | - Lamia A Heikal
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - D Sriram
- Tuberculosis Drug Discovery Laboratory, Pharmacy Group, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500 0078, India
| | - Mona A Mahran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
8
|
Dingiş Birgül Sİ, Kumari J, Tamhaev R, Mourey L, Lherbet C, Sriram D, Akdemir A, Küçükgüzel İ. In silico design, synthesis and antitubercular activity of novel 2-acylhydrazono-5-arylmethylene-4-thiazolidinones as enoyl-acyl carrier protein reductase inhibitors. J Biomol Struct Dyn 2024:1-19. [PMID: 38450660 DOI: 10.1080/07391102.2024.2319678] [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: 10/24/2023] [Accepted: 02/12/2024] [Indexed: 03/08/2024]
Abstract
Mycobacteria regulate the synthesis of mycolic acid through the fatty acid synthase system type 1 (FAS I) and the fatty acid synthase system type-2 (FAS-II). Because mammalian cells exclusively utilize the FAS-I enzyme system for fatty acid production, targeting the FAS-II enzyme system could serve as a specific approach for developing selective antimycobacterial drugs. Enoyl-acyl carrier protein reductase enzyme (MtInhA), part of the FAS-II enzyme system, contains the NADH cofactor in its active site and reduces the intermediate. Molecular docking studies were performed on an in-house database (∼2200 compounds). For this study, five different crystal structures of MtInhA (PDB Code: 4TZK, 4BQP, 4D0S, 4BGE, 4BII) were used due to rotamer difference, mutation and the presence of cofactors. Molecular dynamics simulations (250 ns) were performed for the novel 2-acylhydrazono-5-arylmethylene-4-thiazolidinones derivatives selected by molecular docking studies. Twenty-three compounds selected by in silico methods were synthesized. Antitubercular activity and MtInhA enzyme inhibition studies were performed for compounds whose structures were elucidated by IR,1H-NMR,13C-NMR, HSQC, HMBC, MS and elemental analysis.
Collapse
Affiliation(s)
- Serap İpek Dingiş Birgül
- Institute of Health Sciences, Department of Pharmaceutical Chemistry, Marmara University, Istanbul, Türkiye
- Computer-Aided Drug Discovery Laboratory, Department of Pharmacology, Bezmialem Vakif University, Istanbul, Türkiye
| | - Jyothi Kumari
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| | - Rasoul Tamhaev
- Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, Université Toulouse III - Paul Sabatier, Toulouse Cedex 09, France
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Lionel Mourey
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Christian Lherbet
- Laboratoire de Synthèse et Physico-Chimie de Molécules d'Intérêt Biologique, Université Toulouse III - Paul Sabatier, Toulouse Cedex 09, France
| | - Dharmarajan Sriram
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Hyderabad, India
| | - Atilla Akdemir
- Faculty of Pharmacy, Department of Pharmacology, Istinye University, Istanbul, Türkiye
| | - İlkay Küçükgüzel
- Institute of Health Sciences, Department of Pharmaceutical Chemistry, Marmara University, Istanbul, Türkiye
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Fenerbahçe University, Istanbul, Türkiye
| |
Collapse
|
9
|
Bielenica A, Głogowska A, Augustynowicz-Kopeć E, Orzelska-Górka J, Kurpios-Piec D, Struga M. In vitro antimycobacterial activity and interaction profiles of diarylthiourea-copper (II) complexes with antitubercular drugs against Mycobacterium tuberculosis isolates. Tuberculosis (Edinb) 2023; 143:102412. [PMID: 37774599 DOI: 10.1016/j.tube.2023.102412] [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: 07/04/2023] [Revised: 09/19/2023] [Accepted: 09/23/2023] [Indexed: 10/01/2023]
Abstract
The activity of several halogenated copper (II) complexes of 4-chloro-3-nitrophenylthiourea derivatives has been tested against Mycobacterium tuberculosis strains and strains of non-tuberculous mycobacteria. The compounds were 2-16 times more potent than current TB-drugs against multidrug-resistant M. tuberculosis 210. The 3,4-dichlorophenylthiourea complex (5) was equipotent to ethambutol (EMB) towards M. tuberculosis H37Rv and 192 strains. All derivatives acted 2-8 times stronger than isoniazid (INH) against nontuberculous isolates. In the presence of chosen coordinates, the 2-64 times reduction of MIC values of standard drugs was denoted. The synergistic interaction was found between the complex 4 and rifampicin (RMP), and additivity of 1-5, 8 in pairs with EMB and/or streptomycin (SM) against M. tuberculosis 800 was established. All coordination compounds in combination with at least one drug showed additive activity towards both H37Rv and 192 isolates. In 67% incidences of indifference, the individual MIC of a drug decreased 2-16-fold. One can conclude that the novel thiourea chelates described here are potent hits for further developments of new agents against tuberculosis.
Collapse
Affiliation(s)
- Anna Bielenica
- Department of Biochemistry, Medical University of Warsaw, 02-097, Warsaw, Poland.
| | - Agnieszka Głogowska
- Department of Microbiology, National Tuberculosis and Lung Diseases Research Institute, 01-138, Warsaw, Poland
| | - Ewa Augustynowicz-Kopeć
- Department of Microbiology, National Tuberculosis and Lung Diseases Research Institute, 01-138, Warsaw, Poland
| | - Jolanta Orzelska-Górka
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin, Poland
| | - Dagmara Kurpios-Piec
- Department of Biochemistry, Medical University of Warsaw, 02-097, Warsaw, Poland
| | - Marta Struga
- Department of Biochemistry, Medical University of Warsaw, 02-097, Warsaw, Poland
| |
Collapse
|
10
|
Sahoo SK, Ommi O, Maddipatla S, Singh P, Ahmad MN, Kaul G, Nanduri S, Dasgupta A, Chopra S, Yaddanapudi VM. Isoxazole carboxylic acid methyl ester-based urea and thiourea derivatives as promising antitubercular agents. Mol Divers 2023; 27:2037-2052. [PMID: 36282413 PMCID: PMC9592870 DOI: 10.1007/s11030-022-10543-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/28/2022] [Indexed: 11/01/2022]
Abstract
In our continued efforts to find potential chemotherapeutics active against drug-resistant (DR) Mycobacterium tuberculosis (Mtb), causative agent of Tuberculosis (TB) and to curb the current burdensome treatment regimen, herein we describe the synthesis and biological evaluation of urea and thiourea variants of 5-phenyl-3-isoxazolecarboxylic acid methyl esters as promising anti-TB agent. Majority of the tested compounds displayed potent in vitro activity not only against drug-susceptible (DS) Mtb H37Rv but also against drug-resistant (DR) Mtb. Cell viability test against Vero cells deemed these compounds devoid of significant toxicity. 3,4-Dichlorophenyl derivative (MIC 0.25 µg/mL) and 4-chlorophenyl congener (MIC 1 µg/mL) among urea and thiourea libraries respectively exhibited optimum potency. Lead optimization resulted in the identification of 1,4-linked analogue of 3,4-dichlorophenyl urea derivative demonstrating improved selectivity. Further, in silico study complemented with previously proposed prodrug like attributes of isoxazole esters. Taken together, this molecular hybridization approach presents a new chemotype having potential to be translated into an alternate anti-Mtb agent.
Collapse
Affiliation(s)
- Santosh Kumar Sahoo
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Ojaswitha Ommi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Sarvan Maddipatla
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Priti Singh
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Mohammad Naiyaz Ahmad
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Grace Kaul
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India
| | - Arunava Dasgupta
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Sidharth Chopra
- Division of Microbiology, CSIR-Central Drug Research Institute, Sitapur Road, Sector 10, Jankipuram Extension, Lucknow, Uttar Pradesh, 226031, India.
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, 500037, India.
| |
Collapse
|
11
|
Shalas AF, Winarsih S, Ihsan BRP, Kharismawati A, Firdaus AI, Wiloka E. Molecular docking, synthesis, and antibacterial activity of the analogs of 1-allyl-3-benzoylthiourea. Res Pharm Sci 2023; 18:371-380. [PMID: 37614619 PMCID: PMC10443667 DOI: 10.4103/1735-5362.378084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/15/2022] [Accepted: 05/03/2023] [Indexed: 08/25/2023] Open
Abstract
Background and purpose The incidence of antibiotic resistance rapidly emerges over the globe. In the present study, the synthesis of thiourea derivatives as antibacterial agents and their biological evaluation are reported. Experimental approach Preliminary studies were done by molecular docking of four analogs of 1-allyl-3-benzoylthiourea, clorobiocin, and ciprofloxacin on the DNA gyrase subunit B receptor (PDB: 1KZN). The nucleophilic substitution reaction of benzoyl chloride analogs to the allylthiourea yielded four 1-allyl-3-benzoylthiourea analogs (Cpd 1-4). The reactions were done by a modified Schotten Baumann method. The in vitro antimicrobial activities were determined using the agar dilution method against methicillin-resistant Staphylococcus aureus (MRSA), Salmonella typhi, Escherichia coli, and Pseudomonas aeruginosa. Findings/Results The in-silico study showed that Cpd 1-4 possesses a good interaction on the DNA gyrase subunit B receptor compared to the ciprofloxacin. Cpd 3 had the best binding affinity with a rerank score of - 91.2304. Although the candidate compounds showed unsatisfactory antibacterial activity, they indicated an increasing trend of growth inhibition along with the increment of concentration. Cpd 1 and 4 exhibited in vitro antibacterial activities against MRSA with a minimum inhibitory concentration value of 1000 µg/mL, better compared to the other compounds. Conclusion and implication Despite lacking antibacterial activity, all the synthesized compounds showed an increased trend of growth inhibition along with the increment of concentration. Therefore, additional development should be implemented to the compounds of interest in which optimization of lipophilicity and steric properties are suggested.
Collapse
Affiliation(s)
- Alvan F. Shalas
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Jl. Veteran, Malang, Indonesia
| | - Sri Winarsih
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Jl. Veteran, Malang, Indonesia
| | | | - Aprilia Kharismawati
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Jl. Veteran, Malang, Indonesia
| | - Azatil Ismah Firdaus
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Jl. Veteran, Malang, Indonesia
| | - Era Wiloka
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Jl. Veteran, Malang, Indonesia
| |
Collapse
|
12
|
Alcaraz M, Edwards TE, Kremer L. New therapeutic strategies for Mycobacterium abscessus pulmonary diseases - untapping the mycolic acid pathway. Expert Rev Anti Infect Ther 2023; 21:813-829. [PMID: 37314394 PMCID: PMC10529309 DOI: 10.1080/14787210.2023.2224563] [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: 02/28/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Treatment options against Mycobacterium abscessus infections are very limited. New compounds are needed to cure M. abscessus pulmonary diseases. While the mycolic acid biosynthetic pathway has been largely exploited for the treatment of tuberculosis, this metabolic process has been overlooked in M. abscessus, although it offers many potential drug targets for the treatment of this opportunistic pathogen. AREAS COVERED Herein, the authors review the role of the MmpL3 membrane protein and the enoyl-ACP reductase InhA involved in the transport and synthesis of mycolic acids, respectively. They discuss their importance as two major vulnerable drug targets in M. abscessus and report the activity of MmpL3 and InhA inhibitors. In particular, they focus on NITD-916, a direct InhA inhibitor against M. abscessus, particularly warranted in the context of multidrug resistance. EXPERT OPINION There is an increasing body of evidence validating the mycolic acid pathway as an attractive drug target to be further exploited for M. abscessus lung disease treatments. The NITD-916 studies provide a proof-of-concept that direct inhibitors of InhA are efficient in vitro, in macrophages and in zebrafish. Future work is now required to improve the activity and pharmacological properties of these inhibitors and their evaluation in pre-clinical models.
Collapse
Affiliation(s)
- Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
| | - Thomas E. Edwards
- UCB BioSciences, Bainbridge Island, WA 98109 USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, WA 98109 USA
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, 1919 route de Mende, 34293, Montpellier, France
- INSERM, IRIM, 34293 Montpellier, France
| |
Collapse
|
13
|
Roman R, Pintilie L, Căproiu MT, Dumitrașcu F, Nuță DC, Zarafu I, Ioniță P, Chifiriuc MC, Chiriță C, Moroșan A, Popa M, Bleotu C, Limban C. New N-acyl Thiourea Derivatives: Synthesis, Standardized Quantification Method and In Vitro Evaluation of Potential Biological Activities. Antibiotics (Basel) 2023; 12:antibiotics12050807. [PMID: 37237710 DOI: 10.3390/antibiotics12050807] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 05/28/2023] Open
Abstract
New N-acyl thiourea derivatives with heterocyclic rings have been synthesized by first obtaining isothiocyanate, which further reacted with a heterocyclic amine, characterized by (FT-IR, NMR spectroscopy and FT-ICR) and tested for their in vitro antimicrobial, anti-biofilm and antioxidant activities to obtain a drug candidate in a lead-optimization process. From the tested compounds, those bearing benzothiazole (1b) and 6-methylpyridine (1d) moieties revealed anti-biofilm activity against E. coli ATCC 25922 at MBIC values of 625 µg/mL. Compound 1d exhibited the highest antioxidant capacity (~43%) in the in vitro assay using 1,1-diphenyl-2-picrylhydrazyl (DPPH). Considering the in vitro results, the highest anti-biofilm and antioxidant activities were obtained for compound 1d. Therefore, a reversed-phase high-performance liquid chromatography (RP-HPLC) method has been optimized and validated for the quantitative determination of compound 1d. The detection and quantitation limits were 0.0174 μg/mL and 0.0521 μg/mL, respectively. The R2 correlation coefficient of the LOQ and linearity curves were greater than 0.99, over the concentration range of 0.05 μg/mL-40 μg/mL. The precision and accuracy of the analytical method were within 98-102%, confirming that the method is suitable for the quantitative determination of compound 1d in routine quality control analyses. Evaluating the results, the promising potential of the new N-acyl thiourea derivatives bearing 6-methylpyridine moiety will be further investigated for developing agents with anti-biofilm and antioxidant activities.
Collapse
Affiliation(s)
- Roxana Roman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956 Bucharest, Romania
| | - Lucia Pintilie
- National Institute for Chemical-Pharmaceutical Research & Development, 112 Vitan Av., 031299 Bucharest, Romania
| | - Miron Teodor Căproiu
- "C. D. Nenitzescu" Institute of Organic and Supramolecular Chemistry, 202B Splaiul Independenței, 060023 Bucharest, Romania
| | - Florea Dumitrașcu
- "C. D. Nenitzescu" Institute of Organic and Supramolecular Chemistry, 202B Splaiul Independenței, 060023 Bucharest, Romania
| | - Diana Camelia Nuță
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956 Bucharest, Romania
| | - Irina Zarafu
- Department of Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, 4-12 Regina Elisabeta, 030018 Bucharest, Romania
| | - Petre Ioniță
- Department of Organic Chemistry, Biochemistry and Catalysis, Faculty of Chemistry, University of Bucharest, 4-12 Regina Elisabeta, 030018 Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Department of Microbiology, Faculty of Biology & Research Institute of the University of Bucharest (ICUB), University of Bucharest, 060101 Bucharest, Romania
- Romanian Academy, 010071 Bucharest, Romania
| | - Cornel Chiriță
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956 Bucharest, Romania
| | - Alina Moroșan
- Department of Organic Chemistry "Costin Nenitescu", Faculty of Applied Chemistry and Materials Science, University POLITEHNICA of Bucharest, 011061 Bucharest, Romania
| | - Marcela Popa
- Department of Microbiology, Faculty of Biology & Research Institute of the University of Bucharest (ICUB), University of Bucharest, 060101 Bucharest, Romania
| | - Coralia Bleotu
- Department of Microbiology, Faculty of Biology & Research Institute of the University of Bucharest (ICUB), University of Bucharest, 060101 Bucharest, Romania
- Department of Celular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 285 Mihai Bravu Ave., 030304 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov No. 3, 050044 Bucharest, Romania
| | - Carmen Limban
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, "Carol Davila" University of Medicine and Pharmacy, 6 Traian Vuia, 020956 Bucharest, Romania
| |
Collapse
|
14
|
Roquet-Banères F, Alcaraz M, Hamela C, Abendroth J, Edwards TE, Kremer L. In Vitro and In Vivo Efficacy of NITD-916 against Mycobacterium fortuitum. Antimicrob Agents Chemother 2023; 67:e0160722. [PMID: 36920188 PMCID: PMC10112203 DOI: 10.1128/aac.01607-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/16/2023] [Indexed: 03/16/2023] Open
Abstract
Mycobacterium fortuitum represents one of the most clinically relevant rapid-growing mycobacterial species. Treatments are complex due to antibiotic resistance and to severe side effects of effective drugs, prolonged time of treatment, and co-infection with other pathogens. Herein, we explored the activity of NITD-916, a direct inhibitor of the enoyl-ACP reductase InhA of the type II fatty acid synthase in Mycobacterium tuberculosis. We found that this compound displayed very low MIC values against a panel of M. fortuitum clinical strains and exerted potent antimicrobial activity against M. fortuitum in macrophages. Remarkably, the compound was also highly efficacious in a zebrafish model of infection. Short duration treatments were sufficient to significantly protect the infected larvae from M. fortuitum-induced killing, which correlated with reduced bacterial burdens and abscesses. Biochemical analyses demonstrated an inhibition of de novo synthesis of mycolic acids. Resolving the crystal structure of the InhAMFO in complex with NAD and NITD-916 confirmed that NITD-916 is a direct inhibitor of InhAMFO. Importantly, single nucleotide polymorphism leading to a G96S substitution in InhAMFO conferred high resistance levels to NITD-916, thus resolving its target in M. fortuitum. Overall, these findings indicate that NITD-916 is highly active against M. fortuitum both in vitro and in vivo and should be considered in future preclinical evaluations for the treatment of M. fortuitum pulmonary diseases.
Collapse
Affiliation(s)
- Françoise Roquet-Banères
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Matthéo Alcaraz
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Claire Hamela
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
| | - Jan Abendroth
- UCB BioSciences, Bainbridge Island, Washington, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Thomas E. Edwards
- UCB BioSciences, Bainbridge Island, Washington, USA
- Seattle Structural Genomics Center for Infectious Disease (SSGCID), Seattle, Washington, USA
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier, France
- INSERM, IRIM, Montpellier, France
| |
Collapse
|
15
|
Teneva Y, Simeonova R, Valcheva V, Angelova VT. Recent Advances in Anti-Tuberculosis Drug Discovery Based on Hydrazide-Hydrazone and Thiadiazole Derivatives Targeting InhA. Pharmaceuticals (Basel) 2023; 16:ph16040484. [PMID: 37111241 PMCID: PMC10140854 DOI: 10.3390/ph16040484] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Tuberculosis is an extremely serious problem of global public health. Its incidence is worsened by the presence of multidrug-resistant (MDR) strains of Mycobacterium tuberculosis. More serious forms of drug resistance have been observed in recent years. Therefore, the discovery and/or synthesis of new potent and less toxic anti-tubercular compounds is very critical, especially having in mind the consequences and the delays in treatment caused by the COVID-19 pandemic. Enoyl-acyl carrier protein reductase (InhA) is an important enzyme involved in the biosynthesis of mycolic acid, a major component of the M. tuberculosis cell wall. At the same time, it is a key enzyme in the development of drug resistance, making it an important target for the discovery of new antimycobacterial agents. Many different chemical scaffolds, including hydrazide hydrazones and thiadiazoles, have been evaluated for their InhA inhibitory activity. The aim of this review is to evaluate recently described hydrazide-hydrazone- and thiadiazole-containing derivatives that inhibit InhA activity, resulting in antimycobacterial effects. In addition, a brief review of the mechanisms of action of currently available anti-tuberculosis drugs is provided, including recently approved agents and molecules in clinical trials.
Collapse
Affiliation(s)
- Yoanna Teneva
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Rumyana Simeonova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria
| | - Violeta Valcheva
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | | |
Collapse
|
16
|
Paz JD, Denise de Moura Sperotto N, Ramos AS, Pissinate K, da Silva Rodrigues Junior V, Abbadi BL, Borsoi AF, Rambo RS, Corso Minotto AC, da Silva Dadda A, Galina L, Macchi Hopf FS, Muniz MN, Borges Martinelli LK, Roth CD, Madeira Silva RB, Perelló MA, de Matos Czeczot A, Neves CE, Duarte LS, Leyser M, Dias de Oliveira S, Bizarro CV, Machado P, Basso LA. Novel 4-aminoquinolines: Synthesis, inhibition of the Mycobacterium tuberculosis enoyl-acyl carrier protein reductase, antitubercular activity, SAR, and preclinical evaluation. Eur J Med Chem 2022; 245:114908. [DOI: 10.1016/j.ejmech.2022.114908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022]
|
17
|
Mumtaz S, Iqbal S, Shah M, Hussain R, Rahim F, Rehman W, Khan S, Abid OUR, Rasheed L, Dera AA, Al-ghulikah HA, Kehili S, Elkaeed EB, Alrbyawi H, Alahmdi MI. New Triazinoindole Bearing Benzimidazole/Benzoxazole Hybrids Analogs as Potent Inhibitors of Urease: Synthesis, In Vitro Analysis and Molecular Docking Studies. Molecules 2022; 27:6580. [PMID: 36235116 PMCID: PMC9571547 DOI: 10.3390/molecules27196580] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Twenty-four analogs based on triazinoindole bearing benzimidazole/benzoxazole moieties (1-25) were synthesized. Utilizing a variety of spectroscopic methods, including 1H-, 13C-NMR, and HREI-MS, the newly afforded compounds (1-25) were analyzed. The synthesized analogs were tested against urease enzyme (in vitro) as compared to the standard thiourea drug. All triazinoindole-based benzimidazole/benzoxazole analogs (1-25) exhibited moderate to excellent inhibition profiles, having IC50 values of 0.20 ± 0.01 to 36.20 ± 0.70 μM when evaluated under the positive control of thiourea as a standard drug. To better understand the structure-activity relationship, the synthesized compounds were split into two groups, "A" and "B." Among category "A" analogs, analogs 8 (bearing tri-hydroxy substitutions at the 2,4,6-position of aryl ring C) and 5 (bearing di-hydroxy substitutions at the 3,4-position of aryl ring C) emerged as the most potent inhibitors of urease enzyme and displayed many times more potency than a standard thiourea drug. Besides that, analog 22 (which holds di-hydroxy substitutions at the 2,3-position of the aryl ring) and analog 23 (bearing ortho-fluoro substitution) showed ten-fold-enhanced inhibitory potential compared to standard thiourea among category "B" analogs. Molecular docking studies on the active analogs of each category were performed; the results obtained revealed that the presence of hydroxy and fluoro-substitutions on different positions of aryl ring C play a pivotal role in binding interactions with the active site of the targeted urease enzyme.
Collapse
Affiliation(s)
- Sundas Mumtaz
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Shahid Iqbal
- Department of Chemistry, School of Natural Sciences (SNS), National University of Sciences and Technology (NUST), H-12, Islamabad 46000, Pakistan
| | - Mazloom Shah
- Department of Chemistry, Abbottabad University of Science and Technology (AUST), Abbottabad 22010, Pakistan
| | - Rafaqat Hussain
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Fazal Rahim
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Wajid Rehman
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Shoaib Khan
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | | | - Liaqat Rasheed
- Department of Chemistry, Hazara University, Mansehra 21300, Pakistan
| | - Ayed A. Dera
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61413, Saudi Arabia
| | - Hanan A. Al-ghulikah
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Sana Kehili
- Adham University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Riyadh 13713, Saudi Arabia
| | - Hamad Alrbyawi
- Pharmaceutics and Pharmaceutical Technology Department, College of Pharmacy, Taibah University, Medina 42353, Saudi Arabia
| | - Mohammed Issa Alahmdi
- Department of Chemistry, Faculty of Science, University of Tabuk, Tabuk 71491, Saudi Arabia
| |
Collapse
|
18
|
Biasi‐Garbin RP, Fabris M, Morguette AEB, Andriani GM, Cabral WRC, Pereira PML, Brito TO, Macedo F, Da Silva Lima CH, Lancheros CAC, Nakamura CV, Pinge‐Filho P, Tavares ER, Yamauchi LM, Bispo MLF, Yamada‐Ogatta SF. In Vitro Antimicrobial Screening of Benzoylthioureas: Synthesis, Antibacterial Activity toward
Streptococcus agalactiae
and Molecular Docking Study. ChemistrySelect 2022. [DOI: 10.1002/slct.202202117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Renata P. Biasi‐Garbin
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Marciéli Fabris
- Departamento de Química Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Ana Elisa B. Morguette
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Gabriella M. Andriani
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Weslei R. C. Cabral
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Patrícia M. L. Pereira
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Tiago O. Brito
- Departamento de Química Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Fernando Macedo
- Departamento de Química Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Camilo H. Da Silva Lima
- Departamento de Química Orgânica Universidade Federal do Rio de Janeiro Avenida Athos da Silveira Ramos, n° 149, Bloco A, Centro de Tecnologia, Cidade Universitária 21941-909 Rio de Janeiro Rio de Janeiro - RJ Brazil
| | | | - Celso V. Nakamura
- Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Phileno Pinge‐Filho
- Departamento de Ciências Patológicas Universidade Estadual de Londrina Departamento de Ciências Básicas da Saúde Universidade Estadual de Maringá Avenida Colombo, 5790, Campus Universitário 87020-900 Maringá Paraná Brazil
| | - Eliandro R. Tavares
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Lucy M. Yamauchi
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Marcelle L. F. Bispo
- Departamento de Química Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| | - Sueli F. Yamada‐Ogatta
- Programa de Pós-graduação em Microbiologia Departamento de Microbiologia Universidade Estadual de Londrina Rodovia Celso Garcia Cid s/n, Campus Universitário 86057-970 Londrina Paraná Brazil
| |
Collapse
|
19
|
Chemical Exploration of a Highly Selective Scaffold with Activity against Intracellular Mycobacterium tuberculosis. Microbiol Spectr 2022; 10:e0116122. [PMID: 35612308 PMCID: PMC9241686 DOI: 10.1128/spectrum.01161-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
We previously identified a phenylthiourea series with activity against intracellular Mycobacterium tuberculosis using a high-throughput, high-content assay. We conducted a catalog structure-activity relationship study with a collection of 35 analogs. We identified several thiourea derivatives with excellent potency against intracellular bacteria and good selectivity over eukaryotic cells. Compounds had much lower activity against extracellular bacteria, which was not increased by using cholesterol as the sole carbon source. Compounds were equally active against strains with mutations in QcrB or MmpL3, thereby excluding common, promiscuous targets as the mode of action. The phenylthiourea series represents a good starting point for further exploration to develop novel antitubercular agents. IMPORTANCEMycobacterium tuberculosis is responsible for the highest number of deaths from a bacterial pathogen, with >1.5 million in 2020. M. tuberculosis is a sophisticated pathogen that can replicate inside immune cells. There is an urgent need for new drugs to combat M. tuberculosis and to shorten therapy from 6 to 24 months. We have identified a series of molecules that inhibit the growth of M. tuberculosis inside macrophages; we tested a number of derivatives to link structural features to biological activity. The compounds are likely to have novel mechanism of action and so could be developed as new agents for drug-resistant tuberculosis.
Collapse
|
20
|
Angelova VT, Pencheva T, Vassilev N, K-Yovkova E, Mihaylova R, Petrov B, Valcheva V. Development of New Antimycobacterial Sulfonyl Hydrazones and 4-Methyl-1,2,3-thiadiazole-Based Hydrazone Derivatives. Antibiotics (Basel) 2022; 11:antibiotics11050562. [PMID: 35625207 PMCID: PMC9137698 DOI: 10.3390/antibiotics11050562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 12/03/2022] Open
Abstract
Fifteen 4-methyl-1,2,3-thiadiazole-based hydrazone derivatives 3a–d and sulfonyl hydrazones 5a–k were synthesized. They were characterized by 1H-NMR, 13C NMR, and HRMS. Mycobacterium tuberculosis strain H37Rv was used to assess their antimycobacterial activity. All compounds demonstrated significant minimum inhibitory concentrations (MIC) from 0.07 to 0.32 µM, comparable to those of isoniazid. The cytotoxicity was evaluated using the standard MTT-dye reduction test against human embryonic kidney cells HEK-293T and mouse fibroblast cell line CCL-1. 4-Hydroxy-3-methoxyphenyl substituted 1,2,3-thiadiazole-based hydrazone derivative 3d demonstrated the highest antimycobacterial activity (MIC = 0.0730 µM) and minimal associated cytotoxicity against two normal cell lines (selectivity index SI = 3516, HEK-293, and SI = 2979, CCL-1). The next in order were sulfonyl hydrazones 5g and 5k with MIC 0.0763 and 0.0716 µM, respectively, which demonstrated comparable minimal cytotoxicity. All compounds were subjected to ADME/Tox computational predictions, which showed that all compounds corresponded to Lipinski’s Ro5, and none were at risk of toxicity. The suitable scores of molecular docking performed on two crystallographic structures of enoyl-ACP reductase (InhA) provide promising insight into possible interaction with the InhA receptor. The 4-methyl-1,2,3-thiadiazole-based hydrazone derivatives and sulfonyl hydrazones proved to be new classes of lead compounds having the potential of novel candidate antituberculosis drugs.
Collapse
Affiliation(s)
- Violina T. Angelova
- Department of Chemistry, Faculty of Pharmacy, Medical University, 1431 Sofia, Bulgaria;
- Correspondence: or (V.T.A.); (V.V.)
| | - Tania Pencheva
- Department of QSAR and Molecular Modeling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Nikolay Vassilev
- Laboratory “Nuclear Magnetic Resonance”, Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Elena K-Yovkova
- Faculty of Computer Systems and Technologies, Technical University, 1756 Sofia, Bulgaria;
| | - Rositsa Mihaylova
- Laboratory “Drug Metabolism and Drug Toxicity”, Department “Pharmacology, Pharmacotherapy and Toxicology”, Faculty of Pharmacy, Medical University, 1431 Sofia, Bulgaria;
| | - Boris Petrov
- Department of Chemistry, Faculty of Pharmacy, Medical University, 1431 Sofia, Bulgaria;
| | - Violeta Valcheva
- Laboratory of Molecular Biology of Mycobacteria, Department of Infectious Microbiology, The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
- Correspondence: or (V.T.A.); (V.V.)
| |
Collapse
|
21
|
Yang R, Cao W, Liu S, Li Q, Sun Y, Liang C, Ren W, Liu Y, Meng J, Li C. Evaluation of a novel inhibitor of aspartate semialdehyde dehydrogenase as a potent antitubercular agent against Mycobacterium tuberculosis. J Antibiot (Tokyo) 2022; 75:333-340. [DOI: 10.1038/s41429-022-00520-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/09/2022]
|
22
|
Kuang W, Zhang H, Wang X, Yang P. Overcoming Mycobacterium tuberculosis through small molecule inhibitors to break down cell wall synthesis. Acta Pharm Sin B 2022; 12:3201-3214. [PMID: 35967276 PMCID: PMC9366312 DOI: 10.1016/j.apsb.2022.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/08/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022] Open
Abstract
Mycobacterium tuberculosis (MTB) utilizes multiple mechanisms to obtain antibiotic resistance during the treatment of infections. In addition, the biofilms, secreted by MTB, can further protect the latter from the contact with drug molecules and immune cells. These self-defending mechanisms lay a formidable challenge to develop effective therapeutic agents against chronic and recurring antibiotic-tolerant MTB infections. Although several inexpensive and effective drugs (isoniazid, rifampicin, pyrazinamide and ethambutol) have been discovered for the treatment regimen, MTB continues to cause considerable morbidity and mortality worldwide. Antibiotic resistance and tolerance remain major global issues, and innovative therapeutic strategies are urgently needed to address the challenges associated with pathogenic bacteria. Gratifyingly, the cell wall synthesis of tubercle bacilli requires the participation of many enzymes which exclusively exist in prokaryotic organisms. These enzymes, absent in human hepatocytes, are recognized as promising targets to develop anti-tuberculosis drug. In this paper, we discussed the critical roles of potential drug targets in regulating cell wall synthesis of MTB. And also, we systematically reviewed the advanced development of novel bioactive compounds or drug leads for inhibition of cell wall synthesis, including their discovery, chemical modification, in vitro and in vivo evaluation.
Collapse
Affiliation(s)
- Wenbin Kuang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Haolin Zhang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiang Su Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Corresponding author.
| |
Collapse
|
23
|
Isoquinolinedione-urea hybrids: Synthesis, antibacterial evaluation, drug-likeness, molecular docking and DFT studies. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2021.132007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
24
|
Ahmed S, Manning A, Flint L, Awasthi D, Ovechkina Y, Parish T. Identification of Novel Chemical Scaffolds that Inhibit the Growth of Mycobacterium tuberculosis in Macrophages. Front Pharmacol 2022; 12:790583. [PMID: 35046812 PMCID: PMC8762250 DOI: 10.3389/fphar.2021.790583] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/22/2021] [Indexed: 11/13/2022] Open
Abstract
Mycobacterium tuberculosis is an important global pathogen for which new drugs are urgently required. The ability of the organism to survive and multiply within macrophages may contribute to the lengthy treatment regimen with multiple drugs that are required to cure the infection. We screened the MyriaScreen II diversity library of 10,000 compounds to identify novel inhibitors of M. tuberculosis growth within macrophage-like cells using high content analysis. Hits were selected which inhibited the intramacrophage growth of M. tuberculosis without significant cytotoxicity to infected macrophages. We selected and prioritized compound series based on their biological and physicochemical properties and the novelty of the chemotypes. We identified five chemical classes of interest and conducted limited catalog structure-activity relationship studies to determine their tractability. We tested activity against intracellular and extracellular M. tuberculosis, as well as cytoxicity against murine RAW264.7 and human HepG2 cells. Benzene amide ethers, thiophene carboxamides and thienopyridines were only active against intracellular bacteria, whereas the phenylthiourea series was also active against extracellular bacteria. One member of a phenyl pyrazole series was moderately active against extracellular bacteria. We identified the benzene amide ethers as an interesting series for further work. These new compound classes serve as starting points for the development of novel drugs to target intracellular M. tuberculosis.
Collapse
Affiliation(s)
- Sara Ahmed
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States
| | - Alyssa Manning
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States
| | - Lindsay Flint
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States
| | - Divya Awasthi
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States
| | - Yulia Ovechkina
- TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States
| | - Tanya Parish
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States.,TB Discovery Research, Infectious Disease Research Institute, Seattle, WA, United States
| |
Collapse
|
25
|
Strzyga-Łach P, Chrzanowska A, Podsadni K, Bielenica A. Investigation of the Mechanisms of Cytotoxic Activity of 1,3-Disubstituted Thiourea Derivatives. Pharmaceuticals (Basel) 2021; 14:ph14111097. [PMID: 34832881 PMCID: PMC8623398 DOI: 10.3390/ph14111097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Substituted thiourea derivatives possess confirmed cytotoxic activity towards cancer but also normal cells. To develop new selective antitumor agents, a series of 3-(trifluoromethyl)phenylthiourea analogs were synthesized, and their cytotoxicity was evaluated in vitro against the cell line panel. Compounds 1-5, 8, and 9 were highly cytotoxic against human colon (SW480, SW620) and prostate (PC3) cancer cells, and leukemia K-562 cell lines (IC50 ≤ 10 µM), with favorable selectivity over normal HaCaT cells. The derivatives exerted better growth inhibitory profiles towards selected tumor cells than the reference cisplatin. Compounds incorporating 3,4-dichloro- (2) and 4-CF3-phenyl (8) substituents displayed the highest activity (IC50 from 1.5 to 8.9 µM). The mechanisms of cytotoxic action of the most effective thioureas 1-3, 8, and 9 were studied, including the trypan blue exclusion test of cell viability, interleukin-6, and apoptosis assessments. Compounds reduced all cancerous cell numbers (especially SW480 and SW620) by 20-93%. Derivatives 2 and 8 diminished the viability of SW620 cells by 45-58%. Thioureas 1, 2, and 8 exerted strong pro-apoptotic activity. Compound 2 induced late apoptosis in both colon cancer cell lines (95-99%) and in K-562 cells (73%). All derivatives acted as inhibitors of IL-6 levels in both SW480 and SW620 cells, decreasing its secretion by 23-63%.
Collapse
Affiliation(s)
| | | | | | - Anna Bielenica
- Correspondence: ; Tel.: +(48)-022-572-06-93; Fax: +(48)-022-572-06-79
| |
Collapse
|
26
|
Parmar DR, Soni JY, Guduru R, Rayani RH, Kusurkar RV, Vala AG, Talukdar SN, Eissa IH, Metwaly AM, Khalil A, Zunjar V, Battula S. Discovery of new anticancer thiourea-azetidine hybrids: design, synthesis, in vitro antiproliferative, SAR, in silico molecular docking against VEGFR-2, ADMET, toxicity, and DFT studies. Bioorg Chem 2021; 115:105206. [PMID: 34339975 DOI: 10.1016/j.bioorg.2021.105206] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/29/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022]
Abstract
With the aim to discover potent and novel antitumor agents, a series of thiourea compounds bearing 3-(4-methoxyphenyl)azetidine moiety were designed according to the essential pharmacophoric features of the reported VEGFR-2 inhibitors and synthesized. All the synthesized compounds were evaluated for their in vitro anticancer activity against various human cancer cell lines (lung (A549), prostate (PC3), breast (MCF-7), liver (HepG2), colon (HCT-116), ovarian (SKOV-3), skin (A431), brain (U251) and kidney (786-O)). 3-(4-Methoxy-3-(2-methoxypyridin-4-yl)phenyl)-N-(4-methoxyphenyl)azetidine-1-carbothioamide (3B) was found to be most potent member against PC3, U251, A431, and 786-O cancer cell lines with EC50 values 0.25, 0.6, 0.03, and 0.03 µM, respectively and showed more potency than Doxorubicin in PC3, A431, and 786-O cell lines. Compounds 1B to 7B showed EC50 values ranging from 0.03 to 12.55 µM in A431 cell line. Compound 3-(4-methoxy-3-(pyridin-4-yl)phenyl)-N-(4-methoxyphenyl)azetidine-1-carbothioamide (1B) was found to be highly efficient in A431 and 786-O cell line with EC50 values of 0.77 and 0.73 µM respectively. All the compounds exhibited good to moderate cytotoxic activity. The pharmacophoric features and molecular docking studies confirmed the potentialities of compounds 1B, 2B, 3B and 5B to be VEGFR-2 inhibitors. Moreover, in silico ADMET prediction indicated that most of the synthesized compounds have drug-like properties, possess low adverse effects and toxicity. In addition, the DFT studies for the most active compounds (1B and 3B) were carried out. In the end, our studies revealed that the compounds 1B and 3B represent promising anticancer potentialities through their VEGFR-2 inhibition.
Collapse
Affiliation(s)
- Deepa R Parmar
- Department of Chemistry, Faculty of Basic and Applied Sciences, Madhav University, Abu Road, Sirohi, Rajasthan, India; Piramal Pharma Limited - Discovery Solutions, Plot no. 18, Pharmaceutical Special Economic Zone, Village Matoda, Ahmedabad, Gujarat, India
| | - Jigar Y Soni
- Department of Chemistry, Faculty of Basic and Applied Sciences, Madhav University, Abu Road, Sirohi, Rajasthan, India.
| | - Ramakrishna Guduru
- Piramal Pharma Limited - Discovery Solutions, Plot no. 18, Pharmaceutical Special Economic Zone, Village Matoda, Ahmedabad, Gujarat, India
| | - Rahul H Rayani
- Department of Chemistry, Faculty of Basic and Applied Sciences, Madhav University, Abu Road, Sirohi, Rajasthan, India; Piramal Pharma Limited - Discovery Solutions, Plot no. 18, Pharmaceutical Special Economic Zone, Village Matoda, Ahmedabad, Gujarat, India
| | - Rakesh V Kusurkar
- Department of Chemistry, Faculty of Basic and Applied Sciences, Madhav University, Abu Road, Sirohi, Rajasthan, India; Piramal Pharma Limited - Discovery Solutions, Plot no. 18, Pharmaceutical Special Economic Zone, Village Matoda, Ahmedabad, Gujarat, India
| | - Anand G Vala
- Department of Chemistry, Faculty of Basic and Applied Sciences, Madhav University, Abu Road, Sirohi, Rajasthan, India; Piramal Pharma Limited - Discovery Solutions, Plot no. 18, Pharmaceutical Special Economic Zone, Village Matoda, Ahmedabad, Gujarat, India
| | - Sahista N Talukdar
- In vitro Department, Piramal Pharma Limited - Discovery Solutions, Plot no. 18, Pharmaceutical Special Economic Zone, Village Matoda, Ahmedabad, Gujarat, India; Pharmacokinetics Drug Metabolism Department, Syngene Amgen Research Centre, Plot no. 1,2,3,4, and 5, Bommasandra Jigani Link Road, Bommasandra Industrial Area, Bengaluru, Karnataka, India
| | - Ibrahim H Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo11884, Egypt
| | - Ahmed M Metwaly
- Pharmacognosy and Medicinal Plants Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed Khalil
- Department of Chemistry, College of Science, King Faisal University, Al-Hofuf, Al-Ahsa 31982, Saudi Arabia; Chemistry Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
| | - Vishwanath Zunjar
- School of Engineering and Technology, Navrachana University, Vadodara, Gujarat, India
| | - Satyanarayana Battula
- Department of Chemistry, Uka Tarsadia University, Maliba campus, Bardoli, Gujarat, India
| |
Collapse
|
27
|
Campaniço A, Harjivan SG, Warner DF, Moreira R, Lopes F. Addressing Latent Tuberculosis: New Advances in Mimicking the Disease, Discovering Key Targets, and Designing Hit Compounds. Int J Mol Sci 2020; 21:ijms21228854. [PMID: 33238468 PMCID: PMC7700174 DOI: 10.3390/ijms21228854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Despite being discovered and isolated more than one hundred years ago, tuberculosis (TB) remains a global public health concern arch. Our inability to eradicate this bacillus is strongly related with the growing resistance, low compliance to current drugs, and the capacity of the bacteria to coexist in a state of asymptomatic latency. This last state can be sustained for years or even decades, waiting for a breach in the immune system to become active again. Furthermore, most current therapies are not efficacious against this state, failing to completely clear the infection. Over the years, a series of experimental methods have been developed to mimic the latent state, currently used in drug discovery, both in vitro and in vivo. Most of these methods focus in one specific latency inducing factor, with only a few taking into consideration the complexity of the granuloma and the genomic and proteomic consequences of each physiological factor. A series of targets specifically involved in latency have been studied over the years with promising scaffolds being discovered and explored. Taking in account that solving the latency problem is one of the keys to eradicate the disease, herein we compile current therapies and diagnosis techniques, methods to mimic latency and new targets and compounds in the pipeline of drug discovery.
Collapse
Affiliation(s)
- André Campaniço
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Shrika G. Harjivan
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Digby F. Warner
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa;
- Department of Pathology, SAMRC/NHLS/UCT Molecular Mycobacteriology Research Unit, University of Cape Town, Rondebosch 7701, South Africa
- Welcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Rondebosch 7701, South Africa
| | - Rui Moreira
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
| | - Francisca Lopes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; (A.C.); (S.G.H.); (R.M.)
- Correspondence:
| |
Collapse
|