1
|
Omar MH, Emam SH, Mikhail DS, Elmeligie S. Combretastatin A-4 based compounds as potential anticancer agents: A review. Bioorg Chem 2024; 153:107930. [PMID: 39504638 DOI: 10.1016/j.bioorg.2024.107930] [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: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
The current review discusses the importance of combretastatin A-4 (CA-4) as a lead compound of microtubule targeting agents. CA-4 holds a unique place among naturally occurring compounds having cytotoxic activity. In this review an overall picture of design strategies, structure-activity relationship, synthesis, cytotoxic activity, and binding interactions of promising CA-4 analogues, are discussed and arranged chronologically from 2016 to early 2023. Also, this review emphasizes their biological activity as anticancer agents, within an overview of clinical application limitation and suggested strategies to overcome. Dual targeting tubulin inhibitors showed highpotentialto surpass medication resistance and provide synergistic efficacy. Linking platinum (IV), amino acids, and HDAC targeting moieties to active tubulin inhibitorsproduced potent active compounds. Analogues of CA-4 bridged with azetidin-2-one, pyrazole, sulfide, or carrying selenium atom exhibited cytotoxic action against a variety of malignant cell lines through different pathways.
Collapse
Affiliation(s)
- Mai H Omar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Soha H Emam
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Demiana S Mikhail
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Salwa Elmeligie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
2
|
Zhang X, Liu C, Wei W, Zhang Z, Liang T. Iodine-dependent oxidative regioselective aminochalcogenation of indolines. Chem Commun (Camb) 2024; 60:1152-1155. [PMID: 38189976 DOI: 10.1039/d3cc05999e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
A directing-group-free strategy for oxidative regioselective aminochalcogenation of indolines with amines and dichalconides is presented. This strategy combines tandem coupling sequences and oxidative dehydrogenation methods in a multi-component reaction, enabling the fast construction of a series of C2,3- or C2,5-aminochalcogenated indole derivatives. Moreover, the application of this synthetic approach is demonstrated through the late-stage modification of pharmaceuticals and the derivatization of the products, highlighting its potential and significance.
Collapse
Affiliation(s)
- Xiaoxiang Zhang
- Guangxi Key Laboratory of Electrochemical Energy Materials, Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, People's Republic of China.
| | - Chenrui Liu
- Guangxi Key Laboratory of Electrochemical Energy Materials, Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, People's Republic of China.
| | - Wanxing Wei
- Guangxi Key Laboratory of Electrochemical Energy Materials, Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, People's Republic of China.
| | - Zhuan Zhang
- Guangxi Key Laboratory of Electrochemical Energy Materials, Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, People's Republic of China.
| | - Taoyuan Liang
- Guangxi Key Laboratory of Electrochemical Energy Materials, Guangxi Colleges and Universities Key Laboratory of Applied Chemistry Technology and Resource Development, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi 530004, People's Republic of China.
| |
Collapse
|
3
|
Modh DH, Kulkarni VM. Anticancer Drug Discovery By Structure-Based Repositioning Approach. Mini Rev Med Chem 2024; 24:60-91. [PMID: 37165589 DOI: 10.2174/1389557523666230509123036] [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: 12/04/2022] [Revised: 03/07/2023] [Accepted: 03/28/2023] [Indexed: 05/12/2023]
Abstract
Despite the tremendous progress that has occurred in recent years in cell biology and oncology, in chemical, physical and computer sciences, the disease cancer has continued as the major cause of death globally. Research organizations, academic institutions and pharmaceutical companies invest huge amounts of money in the discovery and development of new anticancer drugs. Though much effort is continuing and whatever available approaches are being attempted, the success of bringing one effective drug into the market has been uncertain. To overcome problems associated with drug discovery, several approaches are being attempted. One such approach has been the use of known, approved and marketed drugs to screen these for new indications, which have gained considerable interest. This approach is known in different terms as "drug repositioning or drug repurposing." Drug repositioning refers to the structure modification of the active molecule by synthesis, in vitro/ in vivo screening and in silico computational applications where macromolecular structure-based drug design (SBDD) is employed. In this perspective, we aimed to focus on the application of repositioning or repurposing of essential drug moieties present in drugs that are already used for the treatment of some diseases such as diabetes, human immunodeficiency virus (HIV) infection and inflammation as anticancer agents. This review thus covers the available literature where molecular modeling of drugs/enzyme inhibitors through SBDD is reported for antidiabetics, anti-HIV and inflammatory diseases, which are structurally modified and screened for anticancer activity using respective cell lines.
Collapse
Affiliation(s)
- Dharti H Modh
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Erandwane, Pune, 411038, Maharashtra, India
| | - Vithal M Kulkarni
- Department of Pharmaceutical Chemistry, Poona College of Pharmacy, Bharati Vidyapeeth (Deemed to be) University, Erandwane, Pune, 411038, Maharashtra, India
| |
Collapse
|
4
|
Duan SF, Song L, Guo HY, Deng H, Huang X, Shen QK, Quan ZS, Yin XM. Research status of indole-modified natural products. RSC Med Chem 2023; 14:2535-2563. [PMID: 38107170 PMCID: PMC10718587 DOI: 10.1039/d3md00560g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/16/2023] [Indexed: 12/19/2023] Open
Abstract
Indole is a heterocyclic compound formed by the fusion of a benzene ring and pyrrole ring, which has rich biological activity. Many indole-containing compounds have been sold on the market due to their excellent pharmacological activity. For example, vincristine and reserpine have been widely used in clinical practice. The diverse structures and biological activities of natural products provide abundant resources for the development of new drugs. Therefore, this review classifies natural products by structure, and summarizes the research progress of indole-containing natural product derivatives, their biological activities, structure-activity relationship and research mechanism which has been studied in the past 13 years, so as to provide a basis for the development of new drug development.
Collapse
Affiliation(s)
- Song-Fang Duan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Lei Song
- Yanbian University Hospital, Yanbian University Yanji 133002 People's Republic of China
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Hao Deng
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Xing Huang
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| | - Xiu-Mei Yin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Interdisciplinary Program of Biological Functional Molecules, College of Integration Science, Yanbian University Yanji 133002 China +86 0433 243 6020 +86 0433 243 6019
| |
Collapse
|
5
|
Yu C, E R, Zhang XW, Hu WQ, Bao G, Li Y, Liu Y, He Z, Li J, Ma W, Mou LY, Wang R, Sun W. NaClO-Mediated Cross Installation of Indoles and Azoles Benefits Anticancer Hit Discovery. ChemMedChem 2023; 18:e202200651. [PMID: 36585386 DOI: 10.1002/cmdc.202200651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/26/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023]
Abstract
Innovations in synthetic chemistry have a profound impact on the drug discovery process, and will always be a necessary driver of drug development. As a result, it is of significance to develop novel simple and effective synthetic installation of medicinal modules to promote drug discovery. Herein, we have developed a NaClO-mediated cross installation of indoles and azoles, both of which are frequently encountered in drugs and natural products. This effective toolbox provides a convenient synthetic route to access a library of N-linked 2-(azol-1-yl) indole derivatives, and can be used for late-stage modification of drugs, natural products and peptides. Moreover, biological screening of the library has revealed that several adducts showed promising anticancer activities against A549 and NCI-H1975 cells, which give us a hit for anticancer drug discovery.
Collapse
Affiliation(s)
- Changjun Yu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Ruiyao E
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Xiao-Wei Zhang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Wen-Qian Hu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Guangjun Bao
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Yiping Li
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Yuyang Liu
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Zeyuan He
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Jingyue Li
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Wen Ma
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Ling-Yun Mou
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| | - Rui Wang
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China.,State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, P. R. China
| | - Wangsheng Sun
- School of Life Sciences, Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, 2019RU066, Lanzhou University, 199 West Donggang Road, Lanzhou, 730000, Gansu, P. R. China
| |
Collapse
|
6
|
Yu M, Jin T, Wang X, Li H, Ji D, Yao J, Zeng H, Shi S, Xu K, Zhang L. Regioselective intramolecular cyclization of o-alkynyl arylamines with the in situ formation of ArXCl to construct poly-functionalized 3-selenylindoles. RSC Adv 2023; 13:6210-6216. [PMID: 36825294 PMCID: PMC9941895 DOI: 10.1039/d3ra00030c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
In this article, a practical and metal-free method for the synthesis of poly-functionalized 3-selenyl/sulfenyl/telluriumindoles from o-alkynyl arylamines has been achieved. In this protocol, the in situ formation of selenenyl chloride, sulfenyl chloride or tellurenyl chloride is considered as the key intermediate and the 3-selenyl/sulfenyl/telluriumindoles can be obtained in good to excellent yields. Furthermore, the product 2-phenyl-3-(phenylselanyl)-1-tosyl-1H-indole can be selectively oxidized to compounds 2-phenyl-3-(phenylseleninyl)-1-tosyl-1H-indole and 2-phenyl-3-(phenylselenonyl)-1-tosyl-1H-indole in good yields.
Collapse
Affiliation(s)
- Minhui Yu
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
- College of Biological, Chemical Sciences and Engineering, Jiaxing University Jiaxing 314001 Zhejiang China
| | - Tao Jin
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| | | | - Haohu Li
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| | - Decai Ji
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| | - Jinzhong Yao
- College of Biological, Chemical Sciences and Engineering, Jiaxing University Jiaxing 314001 Zhejiang China
| | - Heyang Zeng
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| | - Senlei Shi
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| | - Kaimeng Xu
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| | - Lianpeng Zhang
- Yunnan Provincial Key Laboratory of Wood Adhesives and Glued Products, Southwest Forestry University Kunming 650224 Yunnan China
| |
Collapse
|
7
|
Sebastiani J, Puxeddu M, Nalli M, Bai R, Altieri L, Rovella P, Gaudio E, Trisciuoglio D, Spriano F, Lavia P, Fionda C, Masci D, Urbani A, Bigogno C, Dondio G, Hamel E, Bertoni F, Silvestri R, La Regina G. RS6077 induces mitotic arrest and selectively activates cell death in human cancer cell lines and in a lymphoma tumor in vivo. Eur J Med Chem 2023; 246:114997. [PMID: 36502578 DOI: 10.1016/j.ejmech.2022.114997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/20/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022]
Abstract
We synthesized a new inhibitor of tubulin polymerization, the pyrrole (1-(7H-pyrrolo[2,3- d]pyrimidin-4-yl)-1H-pyrrol-3-yl)(3,4,5-trimethoxy-phenyl)methanone 6 (RS6077). Compound 6 inhibited the growth of multiple cancer cell lines, with IC50 values in the nM range, without affecting the growth of non-transformed cells. The novel agent arrested cells in the G2/M phase of the cell cycle in both transformed and non-transformed cell lines, but single cell analysis by time-lapse video recording revealed a remarkable selectivity in cell death induction by compound 6: in RPE-1 non-transformed cells mitotic arrest induced was not necessarily followed by cell death; in contrast, in HeLa transformed and in lymphoid-derived transformed AHH1 cell lines, cell death was effectively induced during mitotic arrest in cells that fail to complete mitosis. Importantly, the agent also inhibited the growth of the lymphoma TMD8 xenograft model. Together these findings suggest that derivative 6 has a selective efficacy in transformed vs non-transformed cells and indicate that the same compound has potential as novel therapeutic agent to treat lymphomas. Compound 6 showed good metabolic stability upon incubation with human liver microsomes.
Collapse
Affiliation(s)
- Jessica Sebastiani
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Roma, Italy
| | - Michela Puxeddu
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Roma, Italy
| | - Marianna Nalli
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Roma, Italy
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, United States
| | - Ludovica Altieri
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy; IBPM Institute of Molecular Biology and Pathology - Consiglio Nazionale Delle Ricerche, Rome, Italy
| | - Paola Rovella
- IBPM Institute of Molecular Biology and Pathology - Consiglio Nazionale Delle Ricerche, Rome, Italy
| | - Eugenio Gaudio
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500, Bellinzona, Switzerland
| | - Daniela Trisciuoglio
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy; IBPM Institute of Molecular Biology and Pathology - Consiglio Nazionale Delle Ricerche, Rome, Italy
| | - Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500, Bellinzona, Switzerland
| | - Patrizia Lavia
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy; IBPM Institute of Molecular Biology and Pathology - Consiglio Nazionale Delle Ricerche, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy
| | - Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Chiara Bigogno
- Aphad SrL, Via Della Resistenza 65, 20090, Buccinasco, Italy
| | - Giulio Dondio
- Aphad SrL, Via Della Resistenza 65, 20090, Buccinasco, Italy
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, United States
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6500, Bellinzona, Switzerland
| | - Romano Silvestri
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Roma, Italy.
| | - Giuseppe La Regina
- Laboratory Affiliated with the Institute Pasteur Italy - Cenci Bolognetti Foundation, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185, Roma, Italy
| |
Collapse
|
8
|
Rios EAM, Gomes CMB, Silvério GL, Luz EQ, Ali S, D'Oca CDRM, Albach B, Campos RB, Rampon DS. Silver-catalyzed direct selanylation of indoles: synthesis and mechanistic insights. RSC Adv 2023; 13:914-925. [PMID: 36686957 PMCID: PMC9811358 DOI: 10.1039/d2ra06813c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/17/2022] [Indexed: 01/05/2023] Open
Abstract
Herein we describe the Ag(i)-catalyzed direct selanylation of indoles with diorganoyl diselenides. The reaction gave 3-selanylindoles with high regioselectivity and also allowed direct access to 2-selanylindoles when the C3 position of the indole ring was blocked via a process similar to Plancher rearrangement. Experimental analyses and density functional theory calculations were carried out in order to picture the reaction mechanism. Among the pathways considered (via concerted metalation-deprotonation, Ag(iii), radical, and electrophilic aromatic substitution), our findings support a classic electrophilic aromatic substitution via Lewis adducts between Ag(i) and diorganoyl diselenides. The results also afforded new insights into the interactions between Ag(i) and diorganoyl diselenides.
Collapse
Affiliation(s)
- Elise Ane Maluf Rios
- Department of Chemistry, Laboratory of Polymers and Catalysis (LaPoCa), Federal University of Paraná - UFPR P. O. Box 19061 Curitiba PR 81531-990 Brazil
| | - Carla M B Gomes
- Department of Chemistry, Laboratory of Polymers and Catalysis (LaPoCa), Federal University of Paraná - UFPR P. O. Box 19061 Curitiba PR 81531-990 Brazil
| | - Gabriel L Silvério
- Department of Chemistry, Laboratory of Polymers and Catalysis (LaPoCa), Federal University of Paraná - UFPR P. O. Box 19061 Curitiba PR 81531-990 Brazil
| | - Eduardo Q Luz
- Department of Chemistry, Laboratory of Polymers and Catalysis (LaPoCa), Federal University of Paraná - UFPR P. O. Box 19061 Curitiba PR 81531-990 Brazil
| | - Sher Ali
- University of São Paulo, Faculty of Animal Science and Food Engineering Pirassununga SP Brazil
| | - Caroline da Ros Montes D'Oca
- Department of Chemistry, Laboratory of Polymers and Catalysis (LaPoCa), Federal University of Paraná - UFPR P. O. Box 19061 Curitiba PR 81531-990 Brazil
| | - Breidi Albach
- Health Department, Unicesumar - The University Center of Maringá Curitiba PR 81070-190 Brazil
| | - Renan B Campos
- Departamento Acadêmico de Química e Biologia, Universidade Tecnológica Federal do Paraná Rua Deputado Heitor de Alencar Furtado, 5000 81280-340 Curitiba Brazil
| | - Daniel S Rampon
- Department of Chemistry, Laboratory of Polymers and Catalysis (LaPoCa), Federal University of Paraná - UFPR P. O. Box 19061 Curitiba PR 81531-990 Brazil
| |
Collapse
|
9
|
Bhuin S, Sharma P, Chakraborty P, Kulkarni OP, Chakravarty M. Solid-state emitting twisted π-conjugate as AIE-active DSE-gen: in vitro anticancer properties against FaDu and 4T1 with biocompatibility and bioimaging. J Mater Chem B 2022; 11:188-203. [PMID: 36477106 DOI: 10.1039/d2tb02078e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Dual-state emissive fluorogens (DSE-gens) are currently defining their importance as a transpiring tool in biological and biomedical applications. This work focuses on designing and synthesizing indole-anthracene-based solid-state emitting twisted π-conjugates using a metal-free protocol to achieve AIE-active DSE-gens, expanding their scope in biological applications. Special effort has been made to introduce proficient and photo/thermostable DSE-gens that inhibit cancer but not normal cells. Here, the lead DSE-gen initially detects cancer and normal cells by bioimaging; however, it could also confirm and distinguish cancer cells from normal cells by its abated fluorescence signal after killing cancer cells. In contrast, the fluorescence signals for a normal cell remain unscathed. Surprisingly, these molecules displayed decent anticancer properties against FaDu and 4T1 but not MCF-7 cell lines. From a series of newly designed indole-based molecules, we report one single 2,3,4-trimethoxybenzene-linked DSE-gen (the lead), exhibiting high ROS generation, less haemolysis, and less cytotoxicity than doxorubicin (DOX) for normal cells, crucial parameters for a biocompatible in vitro anticancer probe. Thus, we present a potentially applicable anticancer drug, offering a bioactive material with bioimaging efficacy and a way to detect dead cancer cells selectively. The primary mechanism behind the identified outcomes is deciphered with the support of experimental (steady-state and time-resolved fluorescence, biological assays, cellular uptake) and molecular docking studies.
Collapse
Affiliation(s)
- Shouvik Bhuin
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India.
| | - Pravesh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India
| | - Purbali Chakraborty
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India
| | - Onkar Prakash Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India
| | - Manab Chakravarty
- Department of Chemistry, Birla Institute of Technology and Science-Pilani, Hyderabad Campus Jawahar Nagar, Shamirpet, Hyderabad, Telangana, 500078, India.
| |
Collapse
|
10
|
Hawash M. Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy. Biomolecules 2022; 12:biom12121843. [PMID: 36551271 PMCID: PMC9776383 DOI: 10.3390/biom12121843] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer accounts for numerous deaths each year, and it is one of the most common causes of death worldwide, despite many breakthroughs in the discovery of novel anticancer candidates. Each new year the FDA approves the use of new drugs for cancer treatments. In the last years, the biological targets of anticancer agents have started to be clearer and one of these main targets is tubulin protein; this protein plays an essential role in cell division, as well as in intracellular transportation. The inhibition of microtubule formation by targeting tubulin protein induces cell death by apoptosis. In the last years, numerous novel structures were designed and synthesized to target tubulin, and this can be achieved by inhibiting the polymerization or depolymerization of the microtubules. In this review article, recent novel compounds that have antiproliferation activities against a panel of cancer cell lines that target tubulin are explored in detail. This review article emphasizes the recent developments of tubulin inhibitors, with insights into their antiproliferative and anti-tubulin activities. A full literature review shows that tubulin inhibitors are associated with properties in the inhibition of cancer cell line viability, inducing apoptosis, and good binding interaction with the colchicine binding site of tubulin. Furthermore, some drugs, such as cabazitaxel and fosbretabulin, have been approved by FDA in the last three years as tubulin inhibitors. The design and development of efficient tubulin inhibitors is progressively becoming a credible solution in treating many species of cancers.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine
| |
Collapse
|
11
|
Fabitha K, Chandrakanth M, Pramod RN, Arya CG, Li Y, Banothu J. Recent Developments in the Synthesis of Indole‐Pyrazole Hybrids. ChemistrySelect 2022. [DOI: 10.1002/slct.202201064] [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)
- K. Fabitha
- Department of Chemistry National Institute of Technology Calicut Kozhikode 673601 Kerala India
| | - Munugala Chandrakanth
- Department of Chemistry National Institute of Technology Calicut Kozhikode 673601 Kerala India
| | - Rakendu N. Pramod
- Department of Chemistry National Institute of Technology Calicut Kozhikode 673601 Kerala India
| | - C. G. Arya
- Department of Chemistry National Institute of Technology Calicut Kozhikode 673601 Kerala India
| | - Yupeng Li
- Masonic Cancer Center and Department of Medicinal Chemistry University of Minnesota Minneapolis Minnesota 55455 United States
| | - Janardhan Banothu
- Department of Chemistry National Institute of Technology Calicut Kozhikode 673601 Kerala India
| |
Collapse
|
12
|
Perin G, Peglow TJ, Penteado F, Nobre PC, Silva KB, Stach G, Barcellos T, Lenardão EJ, Roehrs JA. UVA Light-promoted Catalyst-free Cyclization of Vinyl Selenides: Green and Efficient Synthesis of C3-Unsubstituted 2-Selanyl Benzochalcogenophenes. Chem Asian J 2022; 17:e202101394. [PMID: 35294794 DOI: 10.1002/asia.202101394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/15/2022] [Indexed: 12/16/2022]
Abstract
A metal- and catalyst-free photo-promoted cyclization of properly substituted vinyl selenides was developed using UVA irradiation. A total of eighteen new C3-unsubstituted 2-selanyl benzochalcogenophenes (benzofurans, benzothiophenes and benzoselenophenes) were prepared in 30-86% yield after irradiation with UVA at room temperature. The usefulness of the title compounds was demonstrated in the easy functionalization of the remaining free C-H bond of the benzochalcogenophenes to form new C-Se and C-Br bonds by simple procedures. Furthermore, the reaction can be performed under natural sunlight irradiation and the solvent is easily reused further in several subsequent runs.
Collapse
Affiliation(s)
- Gelson Perin
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Thiago J Peglow
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Filipe Penteado
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Patrick C Nobre
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Krigor B Silva
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Guilherme Stach
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Thiago Barcellos
- Laboratório de Biotecnologia de Produtos Naturais e Sintéticos, Universidade de Caxias do Sul-UCS, Caxias do Sul, RS, Brazil
| | - Eder J Lenardão
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil
| | - Juliano A Roehrs
- Laboratório de Síntese Orgânica Limpa-LASOL, CCQFA, Universidade Federal de Pelotas-UFPel, P.O. Box 354-96010-900, Pelotas, RS, Brazil.,Instituto Federal de Educação Ciência e Tecnologia Sul-rio-grandense-IFSul, 96015-360, Pelotas, RS, Brazil
| |
Collapse
|
13
|
Hong Y, Zhu YY, He Q, Gu SX. Indole derivatives as tubulin polymerization inhibitors for the development of promising anticancer agents. Bioorg Med Chem 2022; 55:116597. [PMID: 34995858 DOI: 10.1016/j.bmc.2021.116597] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/18/2021] [Accepted: 12/27/2021] [Indexed: 01/01/2023]
Abstract
The α- and β-tubulins are the major polypeptide components of microtubules (MTs), which are attractive targets for anticancer drug development. Indole derivatives display a variety of biological activities including antitumor activity. In recent years, a great number of indole derivatives as tubulin polymerization inhibitors have sprung up, which encourages medicinal chemists to pursue promising inhibitors with improved antitumor activities, excellent physicochemical, pharmacokinetic and pharmacodynamic properties. In this review, the recent progress from 2010 to present in the development of indole derivatives as tubulin polymerization inhibitors was summarized and reviewed, which would provide useful clues and inspirations for further design of outstanding tubulin polymerization inhibitors.
Collapse
Affiliation(s)
- Yu Hong
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yuan-Yuan Zhu
- School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan 430205, China.
| | - Qiuqin He
- Department of Chemistry, Fudan University, Shanghai 200433, China.
| | - Shuang-Xi Gu
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering & Pharmacy, Wuhan Institute of Technology, Wuhan 430205, China.
| |
Collapse
|
14
|
Han Mİ, Küçükgüzel ŞG. Thioethers: An Overview. Curr Drug Targets 2022; 23:170-219. [DOI: 10.2174/1389450122666210614121237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 11/22/2022]
Abstract
:
Spreading rapidly in recent years, cancer has become one of the causes of the highest mor-tality rates after cardiovascular diseases. The reason for cancer development is still not clearly under-stood despite enormous research activities in this area. Scientists are now working on the biology of cancer, especially on the root cause of cancer development. The aim is to treat the cancer disease and thus cure the patients. The continuing efforts for the development of novel molecules as potential anti-cancer agents are essential for this purpose. The main aim of this review was to present a survey on the medicinal chemistry of thioethers and provide practical data on their cytotoxicities against various cancer cell lines. The research articles published between 2001-2020 were consulted to pre-pare this review article; however, patent literature has not been included. The thioether-containing heterocyclic compounds may emerge as a new class of potent and effective anti-cancer agents in the future.
Collapse
Affiliation(s)
- M. İhsan Han
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Erciyes University, Talas, 38050, Kayseri, Turkey
| | - Ş. Güniz Küçükgüzel
- Vocational School of Health Services, Fenerbahçe University, Ataşehir, 34758, İstanbul, Turkey
| |
Collapse
|
15
|
Sinha AK, Equbal D, Rastogi SK, Kumar S, Kumar R. An overview on Indole aryl sulfide/sulfone (IAS) as anti‐HIV non‐nucleoside reverse transcriptase inhibitors (NNRTIs). ASIAN J ORG CHEM 2022. [DOI: 10.1002/ajoc.202100744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Arun Kumar Sinha
- CSIR-CDRI (Central Drug Research Institute) Medicinal and Process Chemistry Sitapur Road 226031 Lucknow INDIA
| | | | - Sumit K. Rastogi
- CSIR-CDRI: Central Drug Research Institute Medicinal and Process Chemistry INDIA
| | - Santosh Kumar
- CSIR-CDRI: Central Drug Research Institute Medicinal and process chemistry INDIA
| | - Ravindra Kumar
- CSIR-CDRI: Central Drug Research Institute Medicinal and process chemistry INDIA
| |
Collapse
|
16
|
Iida H. Recent Development of Aerobic Oxidative Transformations by Flavin Catalysis. J SYN ORG CHEM JPN 2022. [DOI: 10.5059/yukigoseikyokaishi.80.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Hiroki Iida
- Department of Chemistry, Graduate School of Natural Science and Technology, Shimane University
| |
Collapse
|
17
|
Paidakula S, Nerella S, Kankala S, Kankala RK. Recent Trends in Tubulin-Binding Combretastatin A-4 Analogs for Anticancer Drug Development. Curr Med Chem 2021; 29:3748-3773. [PMID: 34856892 DOI: 10.2174/0929867328666211202101641] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/20/2021] [Accepted: 10/20/2021] [Indexed: 11/22/2022]
Abstract
Although significant progress over several decades has been evidenced in cancer therapy, there still remains a need for the development of novel and effective therapeutic strategies to treat several relapsed and intractable cancers. In this regard, tubulin protein has become one of the efficient and major targets for anticancer drug discovery. Considering the antimitotic ability, several tubulin inhibitors have been developed to act against various cancers. Among various tubulin inhibitors available, combretastatin-A4 (CA-4), a naturally occurring lead molecule, offers exceptional cytotoxicity (including the drug-resistant cell lines) and antivascular effects. Although CA-4 offers exceptional therapeutic efficacy, several new advancements have been proposed, such as the structural modification via A and B rings, as well as cis-olefinic bridging, which provide highly efficient analogs with improved tubulin-binding efficiency to meet the anticancer drug development requirements. This review systematically emphasizes the recent trends and latest developments in the anticancer drug design & discovery, using CA-4 analogs as the tubulin inhibiting agents, highlighting their structure-activity relationships (SAR) and resultant pharmacological efficacies.
Collapse
Affiliation(s)
- Suresh Paidakula
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Srinivas Nerella
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | - Shravankumar Kankala
- Department of Chemistry, Kakatiya University, Warangal-506009, Telangana State. India
| | | |
Collapse
|
18
|
Indole derivatives (2010-2020) as versatile tubulin inhibitors: synthesis and structure-activity relationships. Future Med Chem 2021; 13:1795-1828. [PMID: 34468201 DOI: 10.4155/fmc-2020-0385] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Tubulin inhibitors are conjugates that interfere with the dynamic equilibrium of the polymerization and depolymerization of microtubules. Among all the reported conjugates, indole moiety is one of the most significant classes for the development of new drug candidates for cancer therapy. Due to their presence in a wide range of natural as well as synthetic antitubulin agents, indole has become a versatile scaffold in research, and various synthetic and semisynthetic indole-based antitubulin agents have been identified and reported. The present article focuses on the reported indole-based tubulin inhibitors of synthetic origin from last the decade. Synthesis, structure-activity relationships and biological activities of synthetic indole derivatives along with brief updates on their antitubulin activity are presented.
Collapse
|
19
|
Natho P, Yang Z, Allen LAT, Rey J, White AJP, Parsons PJ. An entry to 2-(cyclobut-1-en-1-yl)-1 H-indoles through a cyclobutenylation/deprotection cascade. Org Biomol Chem 2021; 19:4048-4053. [PMID: 33885127 DOI: 10.1039/d1ob00430a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A transition-metal-free strategy for the synthesis of 2-(cyclobut-1-en-1-yl)-1H-indoles under mild conditions is described herein. A series of substituted 2-(cyclobut-1-en-1-yl)-1H-indoles are accessed by a one-pot cyclobutenylation/deprotection cascade from N-Boc protected indoles. Preliminary experimental and density functional theory calculations suggest that a Boc-group transfer is involved in the underlying mechanism.
Collapse
Affiliation(s)
- Philipp Natho
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, W12 0BZ London, UK.
| | - Zeyu Yang
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, W12 0BZ London, UK.
| | - Lewis A T Allen
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, W12 0BZ London, UK.
| | - Juliette Rey
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, W12 0BZ London, UK.
| | - Andrew J P White
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, W12 0BZ London, UK.
| | - Philip J Parsons
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, W12 0BZ London, UK.
| |
Collapse
|
20
|
Hawash M, Kahraman DC, Cetin-Atalay R, Baytas SN. Induction of Apoptosis in Hepatocellular Carcinoma Cell Lines by Novel Indolylacrylamide Derivatives: Synthesis and Biological Evaluation. Chem Biodivers 2021; 18:e2001037. [PMID: 33713038 DOI: 10.1002/cbdv.202001037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 03/12/2021] [Indexed: 12/22/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent primary liver cancer and one of the leading causes of cancer associated death worldwide. This is due to the highly resistant nature of this malignancy and the lack of effective treatment options for advanced stage HCC patients. The hyperactivity of PI3K/Akt and Ras/Raf/MEK/ERK signaling pathways contribute to the cancer progression, survival, motility, and resistance mechanisms, and the interaction of these two pathways are responsible for the regulation of cancer cell growth and development. Therefore, it is vital to design and develop novel therapeutic options for HCC treatment targeting these hyperactive pathways. For this purpose, novel series of trans-indole-3-ylacrylamide derivatives originated from the lead compound, 3-(1H-indole-3-yl)-N-(3,4,5-trimethoxyphenyl)acrylamide, have been synthesized and analyzed for their bioactivity on cancer cells along with the lead compound. Based on the initial screening, the most potent compounds were selected to elucidate their effects on cellular signaling activity of HCC cell lines. Cell cycle analysis, immunofluorescence, and Western blot analysis revealed that lead compound and (E)-N-(4-tert-butylphenyl)-3-(1H-indole-3-yl)acrylamide induced cell cycle arrest at the G2/M phase, enhanced chromatin condensation and PARP-cleavage, addressing induction of apoptotic cell death. Additionally, these compounds decreased the activity of ERK signaling pathway, where phosphorylated ERK1/2 and c-Jun protein levels diminished significantly. Relevant to these findings, the lead compound was able to inhibit tubulin polymerization as well. To conclude, the novel trans-indole-3-ylacrylamide derivatives inhibit one of the critical pathways associated with HCC which results in cell cycle arrest and apoptosis in HCC cell lines.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey.,Present address, Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | - Deniz Cansen Kahraman
- Cancer Systems Biology Laboratory, Graduate School of Informatics, METU, 06800, Ankara, Turkey
| | - Rengul Cetin-Atalay
- Cancer Systems Biology Laboratory, Graduate School of Informatics, METU, 06800, Ankara, Turkey
| | - Sultan Nacak Baytas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, 06330, Ankara, Turkey
| |
Collapse
|
21
|
Patel VK, Rajak H. Structural Investigations of Aroylindole Derivatives through 3D-QSAR and Multiple Pharmacophore Modeling for the Search of Novel Colchicines Inhibitor. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999200905092444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background :
The ligand and structure based integrated strategies are being repeatedly
and effectively employed for the precise search and design of novel ligands against various disease
targets. Aroylindole derivative has a similar structural analogy as Combretastatin A-4, and exhibited
potent anticancer activity on several cancer cell lines.
Objective:
To identify structural features of aroylindole derivatives through 3D-QSAR and multiple
pharmacophore modelling for the search of novel colchicines inhibitor via virtual screening.
Method:
The present study utilizes ligand and structure based methodology for the establishment
of structure activity correlation among trimethoxyaroylindole derivatives and the search of novel
colchicines inhibitor via virtual screening. The 3D-QSAR studies were performed using Phase module
and provided details of relationship between structure and biological activity. A single ligand
based pharmacophore model was generated from Phase on compound 3 and compound 29 and three
energetically optimized structure based pharmacophore models were generated from epharmacophore
for co-crystallized ligand, compound 3 and compound 29 with protein PBD ID
1SA0, 5EYP and 5LYJ. These pharmacophoric features containing hit-like compounds were collected
from commercially available ZINC database and screened using virtual screening workflow.
Results and Discussion:
The 3D-QSAR model studies with good PLSs statistics for factor four was
characterized by the best prediction coefficient Q2 (0.8122), regression R2 (0.9405), SD (0.2581), F
(102.7), P (1.56e-015), RMSE (0.402), Stability (0.5411) and Pearson-r (0.9397). The generated epharmacophores
have GH scores over 0.5 and AUAC ≥ 0.7 indicated that all the pharmacophores
were suitable for pharmacophore-based virtual screening. The virtual screened compounds
ZINC12323179, ZINC01642724, and ZINC14238006 have showed similar structural alignment as
co-crystallized ligand and showed the hydrogen bonding of ligand with ASN101, SER178,
THR179, VAL238, CYS241 amino acid of protein.
Conclusion:
The study illustrates that the ligand and structure based pharmacophoric approach is
beneficial for identification of structurally diverse hits, having better binding affinity on colchicines
binding site as novel anticancer agents.
Collapse
Affiliation(s)
- Vijay Kumar Patel
- Medicinal Chemistry Research Laboratory, SLT Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| | - Harish Rajak
- Medicinal Chemistry Research Laboratory, SLT Institute of Pharmaceutical Sciences, Guru Ghasidas University, Bilaspur 495 009, (C.G.), India
| |
Collapse
|
22
|
Tantak MP, Malik M, Klingler L, Olson Z, Kumar A, Sadana R, Kumar D. Indolyl-α-keto-1,3,4-oxadiazoles: Synthesis, anti-cell proliferation activity, and inhibition of tubulin polymerization. Bioorg Med Chem Lett 2021; 37:127842. [PMID: 33556575 DOI: 10.1016/j.bmcl.2021.127842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/14/2021] [Accepted: 01/29/2021] [Indexed: 01/29/2023]
Abstract
A series of novel indolyl-α-keto-1,3,4-oxadiazole derivatives have been synthesized by employing molecular iodine-mediated oxidative cyclization of acylhydrazones. In vitro anti cell proliferation activity of these derivatives against various cancer cells lines such as human lymphoblast (U937), leukemia (Jurkat & SB) and human breast (BT474) was investigated. Among the synthesized indolyl-α-keto-1,3,4-oxadiazoles 19a-p, only one compound (19e) exhibited significant antiproliferative activity against a panel of cell lines. The compound 19e with 3,4,5-trimethoxyphenyl motif, endowed strong cytotoxicity against U937, Jurkat, BT474 and SB cancer cells with IC50 values of 7.1, 3.1, 4.1, and 0.8 µM, respectively. Molecular docking studies suggested a potential binding mode for 19e in the colchicine binding site of tubulin. When tested for in vitro tubulin polymerizaton, 19e inhibited tubulin polymezations (IC50 = 10.66 µM) and induced apoptosis through caspase 3/7 activation. Further, the derivative 19e did not cause necrosis when measured using lactate dehydrogenase assay.
Collapse
Affiliation(s)
- Mukund P Tantak
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India
| | - Monika Malik
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India
| | - Linus Klingler
- Department: Department of Natural Sciences, University of Houston - Downtown, Houston, TX 77002, USA
| | - Zachary Olson
- Department: Department of Natural Sciences, University of Houston - Downtown, Houston, TX 77002, USA
| | - Anil Kumar
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India
| | - Rachna Sadana
- Department: Department of Natural Sciences, University of Houston - Downtown, Houston, TX 77002, USA.
| | - Dalip Kumar
- Department: Department of Chemistry Birla Institute of Technology and Science, Pilani 333 031, India.
| |
Collapse
|
23
|
Tanimoto K, Okai H, Oka M, Ohkado R, Iida H. Aerobic Oxidative C-H Azolation of Indoles and One-Pot Synthesis of Azolyl Thioindoles by Flavin-Iodine-Coupled Organocatalysis. Org Lett 2021; 23:2084-2088. [PMID: 33656903 DOI: 10.1021/acs.orglett.1c00241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The aerobic oxidative cross-coupling of indoles with azoles driven by flavin-iodine-coupled organocatalysis has been developed for the green synthesis of 2-(azol-1-yl)indoles. The coupled organocatalytic system enabled the one-pot three-component synthesis of 2-azolyl-3-thioindoles from indoles, azoles, and thiols in an atom-economical manner by utilizing molecular oxygen as the only sacrificial reagent.
Collapse
Affiliation(s)
- Kazumasa Tanimoto
- Department of Chemistry, Graduate School of Natural Science and Technology, Shimane University, 1060 Nishikawatsu, Matsue, 690-8504, Japan
| | - Hayaki Okai
- Department of Chemistry, Graduate School of Natural Science and Technology, Shimane University, 1060 Nishikawatsu, Matsue, 690-8504, Japan
| | - Marina Oka
- Department of Chemistry, Graduate School of Natural Science and Technology, Shimane University, 1060 Nishikawatsu, Matsue, 690-8504, Japan
| | - Ryoma Ohkado
- Department of Chemistry, Graduate School of Natural Science and Technology, Shimane University, 1060 Nishikawatsu, Matsue, 690-8504, Japan
| | - Hiroki Iida
- Department of Chemistry, Graduate School of Natural Science and Technology, Shimane University, 1060 Nishikawatsu, Matsue, 690-8504, Japan
| |
Collapse
|
24
|
Ana G, Kelly PM, Malebari AM, Noorani S, Nathwani SM, Twamley B, Fayne D, O’Boyle NM, Zisterer DM, Pimentel EF, Endringer DC, Meegan MJ. Synthesis and Biological Evaluation of 1-(Diarylmethyl)-1 H-1,2,4-triazoles and 1-(Diarylmethyl)-1 H-imidazoles as a Novel Class of Anti-Mitotic Agent for Activity in Breast Cancer. Pharmaceuticals (Basel) 2021; 14:169. [PMID: 33671674 PMCID: PMC7926793 DOI: 10.3390/ph14020169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/21/2022] Open
Abstract
We report the synthesis and biochemical evaluation of compounds that are designed as hybrids of the microtubule targeting benzophenone phenstatin and the aromatase inhibitor letrozole. A preliminary screening in estrogen receptor (ER)-positive MCF-7 breast cancer cells identified 5-((2H-1,2,3-triazol-1-yl)(3,4,5-trimethoxyphenyl)methyl)-2-methoxyphenol 24 as a potent antiproliferative compound with an IC50 value of 52 nM in MCF-7 breast cancer cells (ER+/PR+) and 74 nM in triple-negative MDA-MB-231 breast cancer cells. The compounds demonstrated significant G2/M phase cell cycle arrest and induction of apoptosis in the MCF-7 cell line, inhibited tubulin polymerisation, and were selective for cancer cells when evaluated in non-tumorigenic MCF-10A breast cells. The immunofluorescence staining of MCF-7 cells confirmed that the compounds targeted tubulin and induced multinucleation, which is a recognised sign of mitotic catastrophe. Computational docking studies of compounds 19e, 21l, and 24 in the colchicine binding site of tubulin indicated potential binding conformations for the compounds. Compounds 19e and 21l were also shown to selectively inhibit aromatase. These compounds are promising candidates for development as antiproliferative, aromatase inhibitory, and microtubule-disrupting agents for breast cancer.
Collapse
Affiliation(s)
- Gloria Ana
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Patrick M. Kelly
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Sara Noorani
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Seema M. Nathwani
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, DO2R590 Dublin, Ireland;
| | - Darren Fayne
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Niamh M. O’Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| | - Daniela M. Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (S.M.N.); (D.F.); (D.M.Z.)
| | - Elisangela Flavia Pimentel
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista Vila Velha—Espírito Santo, Vila Velha 29102-920, Brazil; (E.F.P.); (D.C.E.)
| | - Denise Coutinho Endringer
- Department of Pharmaceutical Sciences, University Vila Velha, Av. Comissário José Dantas de Melo, n°21, Boa Vista Vila Velha—Espírito Santo, Vila Velha 29102-920, Brazil; (E.F.P.); (D.C.E.)
| | - Mary J. Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Trinity Biomedical Sciences Institute, 152-160 Pearse Street, Dublin 2, DO2R590 Dublin, Ireland; (G.A.); (P.M.K.); (S.N.); (N.M.O.)
| |
Collapse
|
25
|
Daniele S, La Pietra V, Piccarducci R, Pietrobono D, Cavallini C, D'Amore VM, Cerofolini L, Giuntini S, Russomanno P, Puxeddu M, Nalli M, Pedrini M, Fragai M, Luchinat C, Novellino E, Taliani S, La Regina G, Silvestri R, Martini C, Marinelli L. CXCR4 antagonism sensitizes cancer cells to novel indole-based MDM2/4 inhibitors in glioblastoma multiforme. Eur J Pharmacol 2021; 897:173936. [PMID: 33581134 DOI: 10.1016/j.ejphar.2021.173936] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/28/2021] [Accepted: 02/05/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma Multiforme (GBM) is a highly invasive primary brain tumour characterized by chemo- and radio-resistance and poor overall survival. GBM can present an aberrant functionality of p53, caused by the overexpression of the murine double minute 2 protein (MDM2) and its analogue MDM4, which may influence the response to conventional therapies. Moreover, tumour resistance/invasiveness has been recently attributed to an overexpression of the chemokine receptor CXCR4, identified as a pivotal mediator of glioma neovascularization. Notably, CXCR4 and MDM2-4 cooperate in promoting tumour invasion and progression. Although CXCR4 actively promotes MDM2 activation leading to p53 inactivation, MDM2-4 knockdown induces the downregulation of CXCR4 gene transcription. Our study aimed to assess if the CXCR4 signal blockade could enhance glioma cells' sensitivity to the inhibition of the p53-MDMs axis. Rationally designed inhibitors of MDM2/4 were combined with the CXCR4 antagonist, AMD3100, in human GBM cells and GBM stem-like cells (neurospheres), which are crucial for tumour recurrence and chemotherapy resistance. The dual MDM2/4 inhibitor RS3594 and the CXCR4 antagonist AMD3100 reduced GBM cell invasiveness and migration in single-agent treatment and mainly in combination. AMD3100 sensitized GBM cells to the antiproliferative activity of RS3594. It is noteworthy that these two compounds present synergic effects on cancer stem components: RS3594 inhibited the growth and formation of neurospheres, AMD3100 induced differentiation of neurospheres while enhancing RS3594 effectiveness preventing their proliferation/clonogenicity. These results confirm that blocking CXCR4/MDM2/4 represents a valuable strategy to reduce GBM proliferation and invasiveness, acting on the stem cell component too.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Valeria La Pietra
- Department of Pharmacy, University of Naples "Federico II", 80131, Napoli, Italy
| | | | | | | | | | - Linda Cerofolini
- Magnetic Resonance Center (CERM), University of Florence, And Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (C.I.R.M.M.P), 50019, Sesto Fiorentino (FI), Italy
| | - Stefano Giuntini
- Department of Chemistry "Ugo Schiff″, University of Florence, 50019, Sesto Fiorentino (FI), Italy
| | - Pasquale Russomanno
- Department of Pharmacy, University of Naples "Federico II", 80131, Napoli, Italy
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185, Roma, Italy
| | - Marianna Nalli
- Laboratory Affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185, Roma, Italy
| | - Martina Pedrini
- Department of Chemistry, University of Milan, 20133, Milano, Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence, And Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (C.I.R.M.M.P), 50019, Sesto Fiorentino (FI), Italy; Department of Chemistry "Ugo Schiff″, University of Florence, 50019, Sesto Fiorentino (FI), Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence, And Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (C.I.R.M.M.P), 50019, Sesto Fiorentino (FI), Italy; Department of Chemistry "Ugo Schiff″, University of Florence, 50019, Sesto Fiorentino (FI), Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples "Federico II", 80131, Napoli, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185, Roma, Italy
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia, Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, 00185, Roma, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy.
| | - Luciana Marinelli
- Department of Pharmacy, University of Naples "Federico II", 80131, Napoli, Italy.
| |
Collapse
|
26
|
Li X, Mai S, Li X, Xu J, Xu H, Song Q. Cu-Catalyzed o-Amino Benzofuranthioether Formation from N-Tosylhydrazone-Bearing Thiocarbamates and Arylative Electrophiles. Org Lett 2020; 22:7874-7878. [PMID: 32990445 DOI: 10.1021/acs.orglett.0c02778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An important framework of o-amino benzofuranthioethers was constructed by Cu-catalyzed arylative cyclization of N-tosylhydrazone-bearing thiocarbamates with silylaryl triflates or ArI. This transformation provides a novel strategy for the synthesis of valuable arylative o-amino benzofuranthioethers in moderate yields which could not be obtained from known methods. The reaction features smart design, efficient construction, and mild reaction conditions.
Collapse
Affiliation(s)
- Xue Li
- Institute of Next Generation Matter Transformation, College of Material Sciences Engineering, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Shaoyu Mai
- Institute of Next Generation Matter Transformation, College of Material Sciences Engineering, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Xin Li
- Institute of Next Generation Matter Transformation, College of Material Sciences Engineering, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Jian Xu
- Institute of Next Generation Matter Transformation, College of Material Sciences Engineering, Huaqiao University, Xiamen, Fujian 361021, P.R. China
| | - Hetao Xu
- Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry at Fuzhou University, Fuzhou, Fujian 350108, P.R. China
| | - Qiuling Song
- Institute of Next Generation Matter Transformation, College of Material Sciences Engineering, Huaqiao University, Xiamen, Fujian 361021, P.R. China.,Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry at Fuzhou University, Fuzhou, Fujian 350108, P.R. China
| |
Collapse
|
27
|
Hazarika S, Barman P. Visible‐Light Cercosporin Catalyzed Sulfenylation of Electron‐Rich Compounds with Thiols under Transition‐Metal‐Free Conditions. ChemistrySelect 2020. [DOI: 10.1002/slct.202002512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Sukanya Hazarika
- Department of Chemistry National Institute of Technology Silchar Assam 788010 India
| | - Pranjit Barman
- Department of Chemistry National Institute of Technology Silchar Assam 788010 India
| |
Collapse
|
28
|
Hartwig D, Nascimento JE, Bettanin L, Aquino TFB, Jacob RG, Lenardão EJ. Deep Eutectic Solvents: An Alternative Medium for the Preparation of Organosulfur Compounds. CURRENT GREEN CHEMISTRY 2020. [DOI: 10.2174/2213346107999200616110434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Deep Eutectic Solvent (DES) as a “green solvent” has been used as an alternative to replace
Volatile Organic Compounds (VOCs) and traditional Ionic Liquids (ILs). In recent years, DES has
gained much attention due to its excellent properties such as low cost, easy preparation, high viscosity,
low vapor pressure, low volatility, high thermal stability, biodegradability and non-toxicity, among
others. Other classes of compounds with increased interest are organosulfur compounds due to their
applicability as synthetic intermediates in organic reactions and their high importance in pharmaceutical
and agrochemical industries. This review describes the recent advances in the preparation of organosulfur
compounds using DES as an alternative solvent, focusing on several types of organic reactions,
including aromatic substitution reactions (SNAr), condensation, cyclocondensation, cyclization,
ring-opening, thia-Michael addition, one-pot reactions and heterocyclodehydrations.
Collapse
Affiliation(s)
- Daniela Hartwig
- Laboratorio de Síntese Organica Limpa – LASOL, CCQFA, Universidade Federal de Pelotas – UFPel, P.O. Box 354, 96010-900, Pelotas, RS, Brazil
| | - José E.R. Nascimento
- Laboratorio de Síntese Organica Limpa – LASOL, CCQFA, Universidade Federal de Pelotas – UFPel, P.O. Box 354, 96010-900, Pelotas, RS, Brazil
| | - Luana Bettanin
- Laboratorio de Síntese Organica Limpa – LASOL, CCQFA, Universidade Federal de Pelotas – UFPel, P.O. Box 354, 96010-900, Pelotas, RS, Brazil
| | - Thalita F. B. Aquino
- Laboratorio de Síntese Organica Limpa – LASOL, CCQFA, Universidade Federal de Pelotas – UFPel, P.O. Box 354, 96010-900, Pelotas, RS, Brazil
| | - Raquel G. Jacob
- Laboratorio de Síntese Organica Limpa – LASOL, CCQFA, Universidade Federal de Pelotas – UFPel, P.O. Box 354, 96010-900, Pelotas, RS, Brazil
| | - Eder J. Lenardão
- Laboratorio de Síntese Organica Limpa – LASOL, CCQFA, Universidade Federal de Pelotas – UFPel, P.O. Box 354, 96010-900, Pelotas, RS, Brazil
| |
Collapse
|
29
|
Penteado F, Bettanin L, Machado K, Perin G, Alves D, Lenardão EJ. Sonochemistry and Copper Catalysis: An Efficient Duo in the Synthesis of Chalcogenylindolizines. ASIAN J ORG CHEM 2020. [DOI: 10.1002/ajoc.202000318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Filipe Penteado
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Luana Bettanin
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Kethelyn Machado
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Gelson Perin
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Diego Alves
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Eder J. Lenardão
- LASOL – CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| |
Collapse
|
30
|
Barce Ferro CT, dos Santos BF, da Silva CDG, Brand G, da Silva BAL, de Campos Domingues NL. Review of the Syntheses and Activities of Some Sulfur-Containing Drugs. Curr Org Synth 2020; 17:192-210. [DOI: 10.2174/1570179417666200212113412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/06/2019] [Accepted: 12/14/2019] [Indexed: 11/22/2022]
Abstract
Background:
Sulfur-containing compounds represent an important class of chemical compounds due
to their wide range of biological and pharmaceutical properties. Moreover, sulfur-containing compounds may be
applied in other fields, such as biological, organic, and materials chemistry. Several studies on the activities of
sulfur compounds have already proven their anti-inflammatory properties and use to treat diseases, such as
Alzheimer’s, Parkinson’s, and HIV. Moreover, examples of sulfur-containing compounds include dapsone,
quetiapine, penicillin, probucol, and nelfinavir, which are important drugs with known activities.
Objective:
This review will focus on the synthesis and application of some sulfur-containing compounds used to
treat several diseases, as well as promising new drug candidates.
Results:
Due to the variety of compounds containing C-S bonds, we have reviewed the different synthetic
routes used toward the synthesis of sulfur-containing drugs and other compounds.
Collapse
Affiliation(s)
- Criscieli Taynara Barce Ferro
- Faculty of Exact Sciences and Technology, Organic Catalysis and Biocatalysis Laboratory – (LACOB), Federal University of Grande Dourados – UFGD, Dourados/MS, Brazil
| | - Beatriz Fuzinato dos Santos
- Faculty of Exact Sciences and Technology, Organic Catalysis and Biocatalysis Laboratory – (LACOB), Federal University of Grande Dourados – UFGD, Dourados/MS, Brazil
| | - Caren Daniele Galeano da Silva
- Faculty of Exact Sciences and Technology, Organic Catalysis and Biocatalysis Laboratory – (LACOB), Federal University of Grande Dourados – UFGD, Dourados/MS, Brazil
| | - George Brand
- Faculty of Exact Sciences and Technology, Organic Catalysis and Biocatalysis Laboratory – (LACOB), Federal University of Grande Dourados – UFGD, Dourados/MS, Brazil
| | - Beatriz Amaral Lopes da Silva
- Faculty of Exact Sciences and Technology, Organic Catalysis and Biocatalysis Laboratory – (LACOB), Federal University of Grande Dourados – UFGD, Dourados/MS, Brazil
| | - Nelson Luís de Campos Domingues
- Faculty of Exact Sciences and Technology, Organic Catalysis and Biocatalysis Laboratory – (LACOB), Federal University of Grande Dourados – UFGD, Dourados/MS, Brazil
| |
Collapse
|
31
|
Jiang X, Zhao Z, Shen Z, Chen K, Fang L, Yu C. Flavin/I2
-Catalyzed Aerobic Oxidative C-H Sulfenylation of Aryl-Fused Cyclic Amines. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000508] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Xinpeng Jiang
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P.R. China
| | - Zongchen Zhao
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P.R. China
| | - Zhifeng Shen
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P.R. China
| | - Keda Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals; Zhejiang University of Technology; Hangzhou P.R. China
| | - Liyun Fang
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P.R. China
| | - Chuanming Yu
- College of Pharmaceutical Sciences; Zhejiang University of Technology; Hangzhou P.R. China
| |
Collapse
|
32
|
Santos MS, Betim HLI, Kisukuri CM, Campos Delgado JA, Corrêa AG, Paixão MW. Photoredox Catalysis toward 2-Sulfenylindole Synthesis through a Radical Cascade Process. Org Lett 2020; 22:4266-4271. [DOI: 10.1021/acs.orglett.0c01297] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Marilia S. Santos
- Center of Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos, Rodovia Washington Luís, km 235, São Carlos, SP 13565-905, Brazil
| | - Hugo L. I. Betim
- Center of Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos, Rodovia Washington Luís, km 235, São Carlos, SP 13565-905, Brazil
| | - Camila M. Kisukuri
- Center of Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos, Rodovia Washington Luís, km 235, São Carlos, SP 13565-905, Brazil
| | - Jose Antonio Campos Delgado
- Center of Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos, Rodovia Washington Luís, km 235, São Carlos, SP 13565-905, Brazil
| | - Arlene G. Corrêa
- Center of Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos, Rodovia Washington Luís, km 235, São Carlos, SP 13565-905, Brazil
| | - Márcio W. Paixão
- Center of Excellence for Research in Sustainable Chemistry (CERSusChem), Department of Chemistry, Federal University of São Carlos, Rodovia Washington Luís, km 235, São Carlos, SP 13565-905, Brazil
| |
Collapse
|
33
|
Jia Y, Wen X, Gong Y, Wang X. Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur J Med Chem 2020; 200:112359. [PMID: 32531682 DOI: 10.1016/j.ejmech.2020.112359] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 02/07/2023]
Abstract
Cancer chemotherapy is frequently hampered by drug resistance, so the resistance to anticancer agents represents one of the major obstacles for the effective cancer treatment. Indole derivatives have the potential to act on diverse targets in cancer cells and exhibit promising activity against drug-resistant cancers. Moreover, some indole-containing compounds such as Semaxanib, Sunitinib, Vinorelbine, and Vinblastine have already been applied in clinics for various kinds of cancer even drug-resistant cancer therapy. Thus, indole derivatives are one of significant resources for the development of novel anti-drug-resistant cancer agents. This review focuses on the recent development of indole derivatives with potential therapeutic application for drug-resistant cancers, and the mechanisms of action, the critical aspects of design as well as structure-activity relationships, covering articles published from 2010 to 2020.
Collapse
Affiliation(s)
- Yanshu Jia
- Chongqing Institute of Engineering, Chongqing, 400056, China
| | - Xiaoyue Wen
- The Institute of Infection and Inflammation, China Three Gorges University, Yichang, Hubei, 443000, China
| | - Yufeng Gong
- The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157000, China
| | - Xuefeng Wang
- Department of Surgery, Zhuji Affiliated Hospital of Shaoxing University, Zhejiang Province, 311800, China.
| |
Collapse
|
34
|
Penteado F, Gomes CS, Monzon LI, Perin G, Silveira CC, Lenardão EJ. Photocatalytic Synthesis of 3-Sulfanyl- and 1,3-Bis(sulfanyl)indolizines Mediated by Visible Light. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000162] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Filipe Penteado
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Caroline S. Gomes
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Loana I. Monzon
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Gelson Perin
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| | - Claudio C. Silveira
- Departamento de Química; Universidade Federal de Santa Maria - UFSM; CEP: 97105-900 Santa Maria RS Brazil
| | - Eder J. Lenardão
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos; Universidade Federal de Pelotas - UFPel; P. O. box 354 CEP: 96010-900 Pelotas RS Brazil
| |
Collapse
|
35
|
Hałuszczuk A, Babul N, Nierzwicki Ł, Przychodzeń W. General, Mild, and Metal-Free Functionalization of Indole and Its Derivatives Through Direct C3-Selenylation. European J Org Chem 2019. [DOI: 10.1002/ejoc.201900632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Adam Hałuszczuk
- Department of Organic Chemistry; Gdansk University of Technology; Narutowicza St. 11/12 80-233 Gdansk Poland
| | - Natalia Babul
- Department of Organic Chemistry; Gdansk University of Technology; Narutowicza St. 11/12 80-233 Gdansk Poland
| | - Łukasz Nierzwicki
- Department of Physical Chemistry; Gdansk University of Technology; Narutowicza St. 11/12 80-233 Gdansk Poland
| | - Witold Przychodzeń
- Department of Organic Chemistry; Gdansk University of Technology; Narutowicza St. 11/12 80-233 Gdansk Poland
| |
Collapse
|
36
|
Hazarika S, Barman P. Ultrasound Assisted Solvent/Metal Free Synthesis of 3‐Sulfenylindoles Employing TBATB‐Grafted MCM‐48 as a suitable Heterogeneous Catalyst. ChemistrySelect 2019. [DOI: 10.1002/slct.201901455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Sukanya Hazarika
- Department of ChemistryNational Institute of Technology Silchar 788010, Assam India
| | - Pranjit Barman
- Department of ChemistryNational Institute of Technology Silchar 788010, Assam India
| |
Collapse
|
37
|
Liu J, Xue D, Zhu X, Yu L, Mao M, Liu Y. Anticancer evaluation of a novel dithiocarbamate hybrid as the tubulin polymerization inhibitor. Invest New Drugs 2019; 38:525-532. [PMID: 31183632 DOI: 10.1007/s10637-019-00799-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 05/22/2019] [Indexed: 01/30/2023]
Abstract
Novel quinoline-dithiocarbamate hybrids were synthesized and designed by the molecular hybridization strategy. All these derivatives were evaluated for their antiproliferative activity against three selected cancer cell lines (MGC-803, HepG-2 and PC-3). Among them, compound 10c displayed the best antiproliferative activity against PC-3 cells with an IC50 value of 0.43 μM. Celluar mechanisms investigated that compound 10c could inhibit the migration against PC-3 cells by regulation the expression levels of E-cadherin and N-cadherin. Compound 10c induced morphological changes of PC-3 cells and regulated apoptosis-related proteins (Bcl-2, Bax and Cleaved-Parp). In addition, compound 10c inhibited tubulin polymerization in vitro with an IC50 value of 4.02 μM. Importantly, compound 10c inhibited the growth of PC-3 cells in vivo with the low toxicity toward mice. These results suggested that compound 10c might be an antitumor agent with potential for treating prostate cancer.
Collapse
Affiliation(s)
- Jia Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Dongwei Xue
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Xingwang Zhu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Liu Yu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Minghuan Mao
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yili Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| |
Collapse
|
38
|
|
39
|
Equbal D, Singh R, Saima, Lavekar AG, Sinha AK. Synergistic Dual Role of [hmim]Br-ArSO2Cl in Cascade Sulfenylation–Halogenation of Indole: Mechanistic Insight into Regioselective C–S and C–S/C–X (X = Cl and Br) Bond Formation in One Pot. J Org Chem 2019; 84:2660-2675. [DOI: 10.1021/acs.joc.8b03097] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Danish Equbal
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031 India
| | - Richa Singh
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031 India
| | - Saima
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001 India
| | - Aditya G. Lavekar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001 India
| | - Arun K. Sinha
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow 226031 India
- Academy of Scientific and Innovative Research (AcSIR), New Delhi 110001 India
| |
Collapse
|
40
|
Synthesis and evaluation of clioquinol-rolipram/roflumilast hybrids as multitarget-directed ligands for the treatment of Alzheimer's disease. Eur J Med Chem 2019; 163:512-526. [DOI: 10.1016/j.ejmech.2018.12.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 02/07/2023]
|
41
|
La Regina G, Coluccia A, Naccarato V, Silvestri R. Towards modern anticancer agents that interact with tubulin. Eur J Pharm Sci 2019; 131:58-68. [PMID: 30690185 DOI: 10.1016/j.ejps.2019.01.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/19/2018] [Accepted: 01/22/2019] [Indexed: 11/25/2022]
Abstract
Tubulin is the primary target of an ever growing number of natural, semisynthetic and synthetic products as potential anticancer agents. The mechanisms of interaction of these molecules with tubulin are varied. These drug classes have shown to inhibit effectively several cancer types with IC50 from midmicromolar to low nanomolar concentrations. However, some limiting obstacles still remain, such as the development of multidrug resistance and cytotoxicity. We have reviewed recent advances in different classes of tubulin binding agents, including colchicine site agents, Vinca alkaloids, tryprostatins, moroidin, hemiasterlin, diazonamide, taxanes, epothilones and laulimalide.
Collapse
Affiliation(s)
- Giuseppe La Regina
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Piazzale Aldo Moro 5, I-00185 Roma, Italy
| | - Antonio Coluccia
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Piazzale Aldo Moro 5, I-00185 Roma, Italy
| | - Valentina Naccarato
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Piazzale Aldo Moro 5, I-00185 Roma, Italy
| | - Romano Silvestri
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Piazzale Aldo Moro 5, I-00185 Roma, Italy.
| |
Collapse
|
42
|
Hu J, Huang YD, Pan T, Zhang T, Su T, Li X, Luo HB, Huang L. Design, Synthesis, and Biological Evaluation of Dual-Target Inhibitors of Acetylcholinesterase (AChE) and Phosphodiesterase 9A (PDE9A) for the Treatment of Alzheimer's Disease. ACS Chem Neurosci 2019; 10:537-551. [PMID: 30252439 DOI: 10.1021/acschemneuro.8b00376] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
A series of dual-target AChE/PDE9A inhibitor compounds were designed, synthesized, and evaluated as anti-Alzheimer's Disease (AD) agents. Among these target compounds, 11a (AChE: IC50 = 0.048 μM; PDE9A: IC50 = 0.530 μM) and 11b (AChE: IC50 = 0.223 μM; PDE9A: IC50 = 0.285 μM) exhibited excellent and balanced dual-target AChE/PDE9A inhibitory activities. Meanwhile, those two compounds possess good blood-brain barrier (BBB) penetrability and low neurotoxicity. Especially, 11a and 11b could ameliorate learning deficits induced by scopolamine (Scop). Moreover, 11a could also improve cognitive and spatial memory in Aβ25-35-induced cognitive deficit mice in the Morris water-maze test. In summary, our research developed a series of potential dual-target AChE/PDE9A inhibitors, and the data indicated that 11a was a promising candidate drug for the treatment of AD.
Collapse
Affiliation(s)
- Jinhui Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ya-Dan Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tingting Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tianhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tao Su
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xingshu Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Ling Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
43
|
Quatrin PM, Dalla Lana DF, Bazana LCG, de Oliveira LFS, Lettieri Teixeira M, Silva EE, Lopes W, Canto RFS, Silveira GP, Fuentefria AM. 3-Selenocyanate-indoles as new agents for the treatment of superficial and mucocutaneous infections. NEW J CHEM 2019. [DOI: 10.1039/c8nj04935a] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The development of resistance to the current antifungal agents is an alarming problem.
Collapse
Affiliation(s)
- Priscilla Maciel Quatrin
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
| | - Daiane Flores Dalla Lana
- Programa de Pós-Graduação em Ciências Farmacêuticas
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
| | - Luana Candice Genz Bazana
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
| | | | | | - Edilma Elaine Silva
- Departamento de Química Orgânica
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
| | - William Lopes
- Department of Molecular Biology and Biotechnology
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
| | - Rômulo Faria Santos Canto
- Programa de Pós-Graduação em Ciências da Saúde
- Universidade Federal de Ciências da Saúde de Porto Alegre
- Porto Alegre
- Brazil
| | - Gustavo Pozza Silveira
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
- Departamento de Química Orgânica
| | - Alexandre Meneghello Fuentefria
- Programa de Pós-Graduação em Microbiologia Agrícola e do Ambiente
- Universidade Federal do Rio Grande do Sul
- Porto Alegre
- Brazil
- Programa de Pós-Graduação em Ciências Farmacêuticas
| |
Collapse
|
44
|
Wang YT, Shi TQ, Zhu HL, Liu CH. Synthesis, biological evaluation and molecular docking of benzimidazole grafted benzsulfamide-containing pyrazole ring derivatives as novel tubulin polymerization inhibitors. Bioorg Med Chem 2018; 27:502-515. [PMID: 30606674 DOI: 10.1016/j.bmc.2018.12.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/14/2018] [Accepted: 12/21/2018] [Indexed: 12/29/2022]
Abstract
Tubulin-targeting drugs have increasingly become the focus of anticancer drugs research. Twenty-five novel benzimidazole grafted benzsulfamide-containing pyrazole ring derivatives were synthesized and evaluated for bioactivity as potential tubulin polymerization inhibitors. Among them, compound 30 showed the most excellent inhibition against tubulin assembly (IC50 = 1.52 μM) and in vitro growth inhibitory activity against a panel of four human cancer cell lines (IC50 = 0.15, 0.21, 0.33 and 0.17 μM, respectively for A549, Hela, HepG2 and MCF-7). It could also validly induce A549 cell apoptosis, cause cell cycle arrest in G2/M phase and disrupt the cellular microtubule network. These results, along with molecular docking data, provided an important basis for further optimization of compound 30 as a potential anticancer agent.
Collapse
Affiliation(s)
- Yan-Ting Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China; Department of Environmental Science & Engineering, Fudan University, Shanghai 200433, PR China
| | - Tian-Qi Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China.
| | - Chang-Hong Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, PR China.
| |
Collapse
|
45
|
Bzeih T, Zhang K, Khalaf A, Hachem A, Alami M, Hamze A. One-Pot Reaction between N-Tosylhydrazones and 2-Nitrobenzyl Bromide: Route to NH-Free C2-Arylindoles. J Org Chem 2018; 84:228-238. [DOI: 10.1021/acs.joc.8b02623] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Tourin Bzeih
- LabEx LERMIT, Laboratoire de Chimie Thérapeutique, Faculté de Pharmacie, Université Paris-Sud, CNRS, BioCIS−UMR 8076, rue J.-B. Clément, Châtenay-Malabry F−92296, France
- Laboratory for Medicinal Chemistry and Natural Products, Faculty of Sciences (1), and PRASE-EDST, Lebanese University, Hadath, Lebanon
| | - Kena Zhang
- LabEx LERMIT, Laboratoire de Chimie Thérapeutique, Faculté de Pharmacie, Université Paris-Sud, CNRS, BioCIS−UMR 8076, rue J.-B. Clément, Châtenay-Malabry F−92296, France
| | - Ali Khalaf
- Laboratory for Medicinal Chemistry and Natural Products, Faculty of Sciences (1), and PRASE-EDST, Lebanese University, Hadath, Lebanon
| | - Ali Hachem
- Laboratory for Medicinal Chemistry and Natural Products, Faculty of Sciences (1), and PRASE-EDST, Lebanese University, Hadath, Lebanon
| | - Mouad Alami
- LabEx LERMIT, Laboratoire de Chimie Thérapeutique, Faculté de Pharmacie, Université Paris-Sud, CNRS, BioCIS−UMR 8076, rue J.-B. Clément, Châtenay-Malabry F−92296, France
| | - Abdallah Hamze
- LabEx LERMIT, Laboratoire de Chimie Thérapeutique, Faculté de Pharmacie, Université Paris-Sud, CNRS, BioCIS−UMR 8076, rue J.-B. Clément, Châtenay-Malabry F−92296, France
| |
Collapse
|
46
|
Chauhan J, Dasgupta M, Luthra T, Awasthi A, Tripathy S, Banerjee A, Paul S, Nag D, Chakrabarti S, Chakrabarti G, Sen S. Design, synthesis and biological evaluation of a novel library of antimitotic C2-aroyl/arylimino tryptamine derivatives that are also potent inhibitors of indoleamine-2, 3-dioxygenase (IDO). Eur J Pharm Sci 2018; 124:249-265. [DOI: 10.1016/j.ejps.2018.08.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 08/16/2018] [Accepted: 08/23/2018] [Indexed: 01/26/2023]
|
47
|
La Regina G, Bai R, Coluccia A, Naccarato V, Famiglini V, Nalli M, Masci D, Verrico A, Rovella P, Mazzoccoli C, Da Pozzo E, Cavallini C, Martini C, Vultaggio S, Dondio G, Varasi M, Mercurio C, Hamel E, Lavia P, Silvestri R. New 6- and 7-heterocyclyl-1H-indole derivatives as potent tubulin assembly and cancer cell growth inhibitors. Eur J Med Chem 2018; 152:283-297. [DOI: 10.1016/j.ejmech.2018.04.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 01/19/2023]
|
48
|
Gazvoda M, Krivec M, Časar Z, Košmrlj J. En Route to 2-(Cyclobuten-1-yl)-3-(trifluoromethyl)-1H-indole. J Org Chem 2018; 83:2486-2493. [PMID: 29378397 DOI: 10.1021/acs.joc.8b00100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
A six-step synthetic route from 4-chloro-2-methylaniline to 5-chloro-2-(cyclobut-1-en-1-yl)-3-(trifluoromethyl)-1H-indole (1) has been reported. Compound 1a is a key impurity of reverse transcriptase inhibitor efavirenz, an important anti-HIV/AIDS drug. Synthetic challenges, dead ends, and detours are discussed.
Collapse
Affiliation(s)
- Martin Gazvoda
- Faculty of Chemistry and Chemical Technology, University of Ljubljana , Vec̆na pot 113, SI-1000 Ljubljana, Slovenia
| | - Marko Krivec
- Faculty of Chemistry and Chemical Technology, University of Ljubljana , Vec̆na pot 113, SI-1000 Ljubljana, Slovenia
| | - Zdenko Časar
- Lek Pharmaceuticals, d.d., Sandoz Development Center Slovenia , Verovškova ulica 57, SI-1526 Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana , Aškerčeva cesta 7, SI-1000 Ljubljana, Slovenia
| | - Janez Košmrlj
- Faculty of Chemistry and Chemical Technology, University of Ljubljana , Vec̆na pot 113, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
49
|
Barrows RD, Blacklock KM, Rablen PR, Khare SD, Knapp S. Computational assessment of thioether isosteres. J Mol Graph Model 2018; 80:282-292. [PMID: 29414047 DOI: 10.1016/j.jmgm.2018.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/25/2018] [Accepted: 01/29/2018] [Indexed: 11/16/2022]
Abstract
Replacement of the sulfur atom in biologically active diaryl and heteroaryl thioethers (Ar-S-Ar', HAr-S-Ar, and HAr-S-HAr') with any of several one-atom or two-atom linkers can be expected to reduce the susceptibility of the analogue to metabolic oxidation, a well-documented problem for thioethers intended for medicinal chemistry applications. Ab initio calculations indicate how well various proposed thioether isosteric groups, including some new and unusual ones, may perform structurally and electronically in replacing the bridging sulfur atom. Four of these are calculationally evaluated as proposed substructures in Axitinib analogues. The predicted binding behavior of the latter within two different previously crystallographically characterized protein-Axitinib binding sites (VEGFR2 kinase and ABL1 T315I gatekeeper mutant kinase), and an assessment of their suitability and anticipated shortcomings, are presented.
Collapse
Affiliation(s)
- Robert D Barrows
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA
| | - Kristin M Blacklock
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA
| | - Paul R Rablen
- Department of Chemistry & Biochemistry, Swarthmore College, 500 College Ave., Swarthmore, PA 19081 USA
| | - Sagar D Khare
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA
| | - Spencer Knapp
- Department of Chemistry & Chemical Biology, Rutgers The State University of New Jersey, 610 Taylor Rd., Piscataway, NJ 08854 USA.
| |
Collapse
|
50
|
Thurow S, Penteado F, Perin G, Alves D, Santi C, Monti B, Schiesser CH, Barcellos T, Lenardão EJ. Selenium dioxide-promoted selective synthesis of mono- and bis-sulfenylindoles. Org Chem Front 2018. [DOI: 10.1039/c8qo00360b] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Easy access to mono- and bis-sulfenylindoles using the SeO2/I2 system.
Collapse
Affiliation(s)
- Samuel Thurow
- Centro de Ciências Químicas
- Farmacêuticas e de Alimentos Universidade Federal de Pelotas – UFPel
- Pelotas
- Brazil
| | - Filipe Penteado
- Centro de Ciências Químicas
- Farmacêuticas e de Alimentos Universidade Federal de Pelotas – UFPel
- Pelotas
- Brazil
| | - Gelson Perin
- Centro de Ciências Químicas
- Farmacêuticas e de Alimentos Universidade Federal de Pelotas – UFPel
- Pelotas
- Brazil
| | - Diego Alves
- Centro de Ciências Químicas
- Farmacêuticas e de Alimentos Universidade Federal de Pelotas – UFPel
- Pelotas
- Brazil
| | - Claudio Santi
- Department of Pharmaceutical Sciences
- University of Perugia
- Perugia
- Italy
| | - Bonifacio Monti
- Department of Pharmaceutical Sciences
- University of Perugia
- Perugia
- Italy
| | | | - Thiago Barcellos
- Laboratory of Biotechnology of Natural and Synthetic Products
- Universidade de Caxias do Sul – UCS
- Caxias do Sul
- Brazil
| | - Eder J. Lenardão
- Centro de Ciências Químicas
- Farmacêuticas e de Alimentos Universidade Federal de Pelotas – UFPel
- Pelotas
- Brazil
| |
Collapse
|