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
|
Popović P, Pirković A, Topalović D, Živković L, Marković M, Spremo-Potparević B. Cytotoxic, Antioxidant, and Anti-Genotoxic Properties of Combretastatin A4 in Human Peripheral Blood Mononuclear Cells: A Comprehensive In Vitro Study. Biomolecules 2024; 14:1535. [PMID: 39766242 PMCID: PMC11673008 DOI: 10.3390/biom14121535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/15/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Despite significant advances in drug discovery and the promising antitumor potential of combretastatin A4 (CA-4), which selectively targets rapidly dividing cancer cells, CA-4's effects on non-dividing human cells, such as peripheral blood mononuclear cells (PBMCs), remain unclear. The aim of this study is to evaluate the in vitro bioactivity of CA-4 in human PBMCs, focusing on its antigenotoxic and antioxidant properties, while comparing its cytotoxic potency against PBMCs, cancer cell lines (JAR and HeLa), and the normal trophoblast cell line HTR-8/SVneo. Cell viability and metabolic activity were evaluated using the MTT assay. ROS production in PBMCs was measured using the H2DCFDA assay, and DNA damage was assessed using the Comet assay. CA-4 showed cytotoxicity in PBMCs and HTR-8/SVneo cells at concentrations above 200 µM, while cancer cells, JAR and HeLa, showed cytotoxicity at 100 µM and 1 µM, respectively. CA-4 also reduced ROS levels in PBMCs under oxidative stress and showed antioxidant effects at concentrations from 1 to 200 µM. In addition, CA-4 showed antigenotoxic effects against H2O2-induced DNA damage in PBMCs at concentrations of up to 1 µM. CA-4 exhibited lower cytotoxicity in human PBMCs compared to cancer cells, inhibited ROS production, and showed antioxidant and antigenotoxic properties, providing insight into its potential therapeutic efficacy and safety.
Collapse
Affiliation(s)
- Petar Popović
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (P.P.); (D.T.); (L.Ž.); (M.M.); (B.S.-P.)
| | - Andrea Pirković
- Department for Biology of Reproduction, Institute for the Application of Nuclear Energy-INEP, University of Belgrade, 11000 Belgrade, Serbia
| | - Dijana Topalović
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (P.P.); (D.T.); (L.Ž.); (M.M.); (B.S.-P.)
| | - Lada Živković
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (P.P.); (D.T.); (L.Ž.); (M.M.); (B.S.-P.)
| | - Milica Marković
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (P.P.); (D.T.); (L.Ž.); (M.M.); (B.S.-P.)
| | - Biljana Spremo-Potparević
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 11000 Belgrade, Serbia; (P.P.); (D.T.); (L.Ž.); (M.M.); (B.S.-P.)
| |
Collapse
|
3
|
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
|
4
|
Fayad E, Altalhi SA, Abualnaja MM, Alrohaimi AH, Elsaid FG, Abu Almaaty AH, Saleem RM, Bazuhair MA, Ahmed Maghrabi AH, Beshay BY, Zaki I. Novel Acrylate-Based Derivatives: Design, Synthesis, Antiproliferative Screening, and Docking Study as Potential Combretastatin Analogues. ACS OMEGA 2023; 8:38394-38405. [PMID: 37867686 PMCID: PMC10586439 DOI: 10.1021/acsomega.3c05051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023]
Abstract
A variety of 3-(4-chlorophenyl) acrylic acids 4a,b and 3-(4-chlorophenyl)acrylate esters 5a-i were synthesized and structurally proven by spectroscopic studies such as IR, 1H NMR, and 13C NMR as well as mass spectrometry. All substances were investigated for their antiproliferative efficacy against the MDA-MB-231 cell line. Among these, acrylic acid compound 4b demonstrated the most potent cytotoxic effect with an IC50 value of 3.24 ± 0.13 μM, as compared to CA-4 (IC50 = 1.27 ± 09 μM). Additionally, acrylic acid molecule 4b displayed an inhibitory effect against β-tubulin polymerization with a percentage inhibition of 80.07%. Furthermore, compound 4b was found to produce considerable cell cycle arrest at the G2/M stage and cellular death, as demonstrated by FACS analysis. In addition, the in vivo antitumor screening of the sodium salt of acrylic acid 4b was carried out, and the results have shown that the tested molecule showed a significant decrease in viable EAC count and EAC volume, accompanied by a considerable increase in the life span prolongation, if compared to the positive control group. Furthermore, molecular modeling studies were performed to understand how the highly efficient chemicals 4b and 5e interact with the colchicine-binding region on tubulin. This work aims to shed light on the reasons behind their exceptional cytotoxicity and their better capacity to inhibit tubulin in comparison to CA-4.
Collapse
Affiliation(s)
- Eman Fayad
- Department
of Biotechnology, College of Sciences, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Sarah Awwadh Altalhi
- Department
of Biotechnology, College of Sciences, Taif
University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Matokah M. Abualnaja
- Department
of Chemistry, Faculty of Applied Science, Umm Al-Qura University, Makkah
Al Mukarrama 24230, Saudi Arabia
| | - Abdulmohsen H. Alrohaimi
- Department
of Pharmacy Practice, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Fahmy G. Elsaid
- Biology
Department, College of Science, King Khalid
University, P.O.Box 960, Asir, Abha 61421, Saudi Arabia
| | - Ali H. Abu Almaaty
- Zoology
Department, Faculty of Science Port Said
University, Port Said 42526, Egypt
| | - Rasha Mohammed Saleem
- Department
of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha 65431, Saudi Arabia
| | - Mohammed A. Bazuhair
- Department
of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ali Hassan Ahmed Maghrabi
- Department
of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah 24381, Saudi Arabia
| | - Botros Y. Beshay
- Pharmaceutical
Sciences (Pharmaceutical Chemistry) Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime
Transport, Alexandria 21913, Egypt
| | - Islam Zaki
- Pharmaceutical
Organic Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| |
Collapse
|
5
|
Li YY, Xiong YM, Zhang SY, Deng JL, Xue Q, Hou XW, Liu WB, Li XH, Qin ZF. Tetrabromobisphenol A-bis(2,3-dibromopropyl ether) impairs Postnatal Testis Development in Mice: The Microtubule Cytoskeleton as a Sensitive Target. ENVIRONMENT & HEALTH (WASHINGTON, D.C.) 2023; 1:168-179. [PMID: 39473615 PMCID: PMC11504584 DOI: 10.1021/envhealth.3c00044] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/08/2023] [Accepted: 06/08/2023] [Indexed: 04/25/2025]
Abstract
Tetrabromobisphenol A-bis(2,3-dibromopropyl ether) (TBBPA-BDBPE), a widely used flame retardant, has been frequently detected in various environmental compartments, but its health hazard remains largely unknown. Here, we investigated the adverse effects of TBBPA-BDBPE (50 and 1000 μg/kg/day) on postnatal testis development in CD-1 mice and the underlying mechanism. Following the first week of maternal exposure, neonatal mice in the high-dose group exhibited reduced seminiferous tubule area, fewer Sertoli cells and germ cells, and damaged microtubules in Sertoli cells; even microtubule damage was also observed in the low-dose group. When exposure extended to adulthood, male offspring in the high-dose group presented more remarkable alterations in reproductive parameters, including reduced sperm count; in the low-dose group, microtubule damage was also observable, along with blood-testis barrier impairment. Further molecular docking analysis and tubulin polymerization assay indicated that TBBPA-BDBPE could interact with tubulin and disrupt its polymerization. Moreover, we observed attenuated microtubules in mouse Sertoli cells in vitro (TM4) following TBBPA-BDBPE treatment, suggesting that TBBPA-BDBPE impaired testis development possibly by interfering with tubulin dynamics. This study not only highlights the male reproductive hazard of TBBPA-BDBPE but also greatly improved the understanding of the molecular mechanism for male reproductive toxicity of chemicals.
Collapse
Affiliation(s)
- Yuan-Yuan Li
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Ming Xiong
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Shu-Yan Zhang
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing-Lin Deng
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiao Xue
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Xing-Wang Hou
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Wen-Bin Liu
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
- Hangzhou
Institute for Advanced Study, University
of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Xing-Hong Li
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhan-Fen Qin
- State
Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese
Academy of Sciences, Beijing 100085, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Barzegar-Fallah A, Alimoradi H, Dunlop JL, Torbati E, Baird SK. Serotonin type-3 receptor antagonists selectively kill melanoma cells through classical apoptosis, microtubule depolymerisation, ERK activation, and NF-κB downregulation. Cell Biol Toxicol 2023; 39:1119-1135. [PMID: 34654991 DOI: 10.1007/s10565-021-09667-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/28/2021] [Indexed: 12/26/2022]
Abstract
Malignant melanoma is a highly metastatic tumour, resistant to treatment. Serotonin type-3 (5-HT3) receptor antagonists, such as tropisetron and ondansetron, are well-tolerated antiemetic drugs commonly used to prevent nausea caused by chemotherapy or radiotherapy. We investigated the anticancer effects of these drugs on melanoma cancer cell lines WM-266-4 and B16F10 with or without paclitaxel. We constructed IC50 curves and performed Chou-Talalay analysis, using data obtained with the MTT assay. Flow cytometry and fluorescent microscopy were used to examine characteristics of the cell cycle, cell death and cytoskeleton changes. Protein levels and activation were analysed by western blotting and molecular docking studies carried out. Data were analysed by one way ANOVA and post hoc testing. Ondansetron and tropisetron showed selective concentration-dependent cytotoxicity in melanoma cell lines WM-266-4 and B16F10. The effect in combination with paclitaxel was synergistic. The drugs did not cause cell cycle arrest but did promote characteristics of classical apoptosis, including accumulation of subG1 DNA, cleaved caspase-3, mitochondrial membrane permeability and phosphatidylserine exposure. As well, the cytosolic calcium level in the melanoma cells was enhanced, phosphorylated ERK1/2 induced and NF-κB inhibited. Finally, the formation of microtubules was shown to be impaired in melanoma cells treated with ondansetron or tropisetron. Docking studies were used to predict that these drugs could bind to the colchicine binding site on the tubulin molecule. Antiemetic drugs, already given in combination with chemotherapy, may enhance the cytotoxic effect of chemotherapy, following successful delivery to the tumour site.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Houman Alimoradi
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Jessica L Dunlop
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Elham Torbati
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Sarah K Baird
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
7
|
Zhang J, Aray B, Zhang Y, Bai Y, Yuan T, Ding S, Xue Y, Huang X, Li Z. Synergistic effect of cucurbitacin E and myricetin on Anti-Non-Small cell lung cancer: Molecular mechanism and therapeutic potential. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154619. [PMID: 36706697 DOI: 10.1016/j.phymed.2022.154619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 09/26/2022] [Accepted: 12/19/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is associated with extremely high morbidity and mortality rates worldwide. Citrullus colocynthis (L.) Schrad, widely distributed in Asian and African countries, is used to treat cancers in traditional Uyghur medicine. HYPOTHESIS/PURPOSE The combination of Cucurbitacin E (CuE) and Myricetin (Myr) of C. colocynthis could treat NSCLC by targeting autophagy. STUDY DESIGN The potential anti-cancer components (CuE and Myr) of C. colocynthis were identified using in-silico methods and further in vitro explored the anti-NSCLC properties of the combination of CuE and Myr. METHODS Network pharmacology and molecular docking were used to identify potential therapeutic compounds of C. colocynthis for the treatment of NSCLC. In A549 cells, the anti-cancer activities and synergy of CuE and Myr were studied using CompuSyn, their mechanism behind autophagy regulation was determined by western blotting and immunofluorescence staining. RESULTS CuMy-12 (CuE: 0.5 µM, Myr: 20 µM), a combination of CuE and Myr from C. colocynthis, inhibited A549 cell proliferation and colony formation, and induced apoptosis and cell cycle arrest in the G0/G1 phase, exhibiting a synergistic effect. Furthermore, CuMy-12 inhibited autophagy and activation of the PI3K/AKT/mTOR signaling pathway, which was characterized by a decrease in Beclin 1, AKT, and phospho-AKT proteins. CONCLUSION CuMy-12 can be considered a natural candidate with anticancer activity for autophagy-based regulation, but mechanistic and clinical studies are required to validate its potential.
Collapse
Affiliation(s)
- Jinfang Zhang
- School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Baht Aray
- School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Yan Zhang
- School of Life and Science, Minzu University of China, Beijing 100081, China
| | - Yinglu Bai
- School of Pharmacy, Minzu University of China, Beijing 100081, China
| | - Tao Yuan
- College of Life Sciences, Jiangxi Normal University, Nanchang 330027, China
| | - Shilan Ding
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yanyu Xue
- School of Life and Science, Minzu University of China, Beijing 100081, China
| | - Xiulan Huang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Zhiyong Li
- School of Pharmacy, Minzu University of China, Beijing 100081, China.
| |
Collapse
|
8
|
Dwivedi AR, Rawat SS, Kumar V, Kumar N, Kumar V, Yadav RP, Baranwal S, Prasad A, Kumar V. Benzotriazole Substituted 2-Phenylquinazolines as Anticancer Agents: Synthesis, Screening, Antiproliferative and Tubulin Polymerization Inhibition Activity. Curr Cancer Drug Targets 2023; 23:278-292. [PMID: 36306454 DOI: 10.2174/1568009623666221028121906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022]
Abstract
AIMS Development of anticancer agents targeting tubulin protein. BACKGROUND Tubulin protein is being explored as an important target for anticancer drug development. Ligands binding to the colchicine binding site of the tubulin protein act as tubulin polymerization inhibitors and arrest the cell cycle in the G2/M phase. OBJECTIVE Synthesis and screening of benzotriazole-substituted 2-phenyl quinazolines as potential anticancer agents. METHODS A series of benzotriazole-substituted quinazoline derivatives have been synthesized and evaluated against human MCF-7 (breast), HeLa (cervical) and HT-29 (colon) cancer cell lines using standard MTT assays. RESULTS ARV-2 with IC50 values of 3.16 μM, 5.31 μM, 10.6 μM against MCF-7, HELA and HT29 cell lines, respectively displayed the most potent antiproliferative activities in the series while all the compounds were found non-toxic against HEK293 (normal cells). In the mechanistic studies involving cell cycle analysis, apoptosis assay and JC-1 studies, ARV-2 and ARV-3 were found to induce mitochondria-mediated apoptosis. CONCLUSION The benzotriazole-substituted 2-phenyl quinazolines have the potential to be developed as potent anticancer agents.
Collapse
Affiliation(s)
- Ashish Ranjan Dwivedi
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Suraj Singh Rawat
- School of Basic Sciences, Indian Institute of Technology, Mandi-175005, HP, India
| | - Vijay Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Naveen Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Vinay Kumar
- Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Ravi Prakash Yadav
- Department of Microbiology, School of Biological Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Somesh Baranwal
- Department of Microbiology, School of Biological Sciences, Central University of Punjab, Bathinda-151401, Punjab, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology, Mandi-175005, HP, India
| | - Vinod Kumar
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Bathinda-151401, Punjab, India.,Department of Chemistry, Laboratory of Organic and Medicinal Chemistry, Central University of Punjab, Bathinda-151401, Punjab, India
| |
Collapse
|
9
|
Nolasco-Quintana NY, González-Maya L, Razo-Hernández RS, Alvarez L. Exploring the Gallic and Cinnamic Acids Chimeric Derivatives as Anticancer Agents over HeLa Cell Line: An in silico and in vitro Study. Mol Inform 2023; 42:e2200016. [PMID: 36065495 DOI: 10.1002/minf.202200016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 09/03/2022] [Indexed: 01/12/2023]
Abstract
Cervical cancer is one of the most aggressive and important cancer types in the female population, due to its low survival rate. Actually, the search for new bioactive compounds, like gallic and cinnamic acid, is one of the most employed options to finding a treatment. In the present study, 134 phenolic compounds with cytotoxic activity over HeLa cell line were used to generate a descriptive ( R 2 ${{R}^{2}}$ =0.76) and predictive ( Q 2 ${{Q}^{2}}$ =0.69 and Q e x t 2 ${{Q}_{{\rm e}{\rm x}{\rm t}}^{2}}$ =0.62) QSAR model. Structural, electronic, steric, and hydrophobic features are represented as different molecular descriptors in our QSAR model. From this model, nine gallate-cinnamate ester derivatives (N1-N9) were designed and synthesized. Furthermore, in vitro cytotoxic activity was evaluated against HeLa and non-tumorigenic cells. Derivatives N6, N5, N1, and N9 were the most active molecules with IC50ExpHeLa values from 7.26 to 11.95 μM. Finally, the binding of the synthesized compounds to the colchicine binding site on tubulin was evaluated by molecular docking as a possible action mechanism. N1, N5 and N6 can be considered as templates for the design of new cervical anticancer compounds.
Collapse
Affiliation(s)
- Ninfa Yaret Nolasco-Quintana
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad No. 1001, Col. Chamilpa, C.P. 62210, Cuernavaca, Mor., México.,Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad No. 1001, Col. Chamilpa, C.P. 62210, Cuernavaca, Mor., México
| | - Leticia González-Maya
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, 62209, Morelos, México
| | - Rodrigo Said Razo-Hernández
- Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad No. 1001, Col. Chamilpa, C.P. 62210, Cuernavaca, Mor., México
| | - Laura Alvarez
- Centro de Investigaciones Químicas, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Av. Universidad No. 1001, Col. Chamilpa, C.P. 62210, Cuernavaca, Mor., México
| |
Collapse
|
10
|
Acylhydrazones and Their Biological Activity: A Review. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248719. [PMID: 36557851 PMCID: PMC9783609 DOI: 10.3390/molecules27248719] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
Due to the structure of acylhydrazones both by the pharmacophore -CO-NH-N= group and by the different substituents present in the molecules of compounds of this class, various pharmacological activities were reported, including antitumor, antimicrobial, antiviral, antiparasitic, anti-inflammatory, immunomodulatory, antiedematous, antiglaucomatous, antidiabetic, antioxidant, and actions on the central nervous system and on the cardiovascular system. This fragment is found in the structure of several drugs used in the therapy of some diseases that are at the top of public health problems, like microbial infections and cardiovascular diseases. Moreover, the acylhydrazone moiety is present in the structure of some compounds with possible applications in the treatment of other different pathologies, such as schizophrenia, Parkinson's disease, Alzheimer's disease, and Huntington's disease. Considering these aspects, we consider that a study of the literature data regarding the structural and biological properties of these compounds is useful.
Collapse
|
11
|
Zaki I, Moustafa AMY, Beshay BY, Masoud RE, Elbastawesy MAI, Abourehab MAS, Zakaria MY. Design and synthesis of new trimethoxylphenyl-linked combretastatin analogues loaded on diamond nanoparticles as a panel for ameliorated solubility and antiproliferative activity. J Enzyme Inhib Med Chem 2022; 37:2679-2701. [PMID: 36154552 PMCID: PMC9518609 DOI: 10.1080/14756366.2022.2116016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
A new series of vinyl amide-, imidazolone-, and triazinone-linked combretastatin A-4 analogues have been designed and synthesised. These compounds have been evaluated for their cytotoxic activity against MDA-MB-231 breast cancer cells. The triazinone-linked combretastatin analogues (6 and 12) exhibited the most potent cytotoxic activity, in sub-micromolar concentration compared with combretastatin A-4 as a reference standard. The results of β-tubulin polymerisation inhibition assay appear to correlate well with the ability to inhibit β-tubulin polymerisation. Additionally, these compounds were subjected to biological assays relating to cell cycle aspects and apoptosis induction. In addition, the most potent compound 6 was loaded on PEG-PCL modified diamond nanoparticles (PEG-PCL-NDs) and F4 was picked as the optimum formula. F4 exhibited enhanced solubility and release over the drug suspension. In the comparative cytotoxic activity, PEG-PCL modified F4 was capable of diminishing the IC50 by around 2.89 times for nude F4, while by 3.48 times relative to non-formulated compound 6.
Collapse
Affiliation(s)
- Islam Zaki
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| | - Amal M Y Moustafa
- Chemistry Department, Faculty of Science, Port Said University, Port Said, Egypt
| | - Botros Y Beshay
- Pharmaceutical Sciences (Pharmaceutical Chemistry) Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria, Egypt
| | - Reham E Masoud
- Clinical Pharmacology Department, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Mohammed A I Elbastawesy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.,Department of Pharmaceutics, College of Pharmacy, Minia University, Minia, Egypt
| | - Mohamed Y Zakaria
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Port Said University, Port Said, Egypt
| |
Collapse
|
12
|
Structural insight into SSE15206 in complex with tubulin provides a rational design for pyrazolinethioamides as tubulin polymerization inhibitors. Future Med Chem 2022; 14:785-794. [PMID: 35506429 DOI: 10.4155/fmc-2021-0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background: Tubulin protein is a promising target for antitumor drugs. Some tubulin inhibitors targeting the colchicine binding site are not substrates of the multidrug-resistance efflux pump, which can overcome the mechanism of drug resistance mediated by P-glycoprotein. Methodology/results: SSE15206 is a colchicine binding site inhibitor with antiproliferative activity against different drug-resistant cell lines. Unfortunately, the lack of detailed interaction information about SSE15206 in complex with tubulin impeded the development of potent drugs that possess similar scaffolds. Herein, the authors report the crystal structure of the tubulin-SSE15206 complex at a resolution of 2.8 Å. Conclusion: The complex structure reveals the intermolecular interactions between SSE15206 and tubulin, providing a rationale for the development of pyrazolinethioamides as tubulin polymerization inhibitors and to overcome multidrug resistance.
Collapse
|
13
|
Talimarada D, Sharma A, Holla H. Identification of dual binding mode of Orthodiffenes towards human topoisomerase-I and α-tubulin: exploring the potential role in anti-cancer activity via in silico study. J Biomol Struct Dyn 2022; 41:2789-2803. [PMID: 35174766 DOI: 10.1080/07391102.2022.2039296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The drugs prescribed for targeting the tumour growth comprise of chemotherapy regimen involving combinations to cell-cycle phase specific target receptors. The combination therapy with Topoisomerase-I (Topo-I) & anti-tubulin agents are in the clinical trial stages and have scope for identifying new chemical entities with dual binding and inhibiting potential. The checkpoint proteins present at the interface of cell-cycle phases are considered the link between these two that establish the connectivity across the two phases of cell-cycle. In the present study, this potential cross-link or dual targeting is explored via in silico analysis on the natural molecules, Orthodiffene (OD) A-F which are reported from the medicinal plant, Orthosiphon diffusus. These molecules have been reported to possess significant cytotoxicity against Jurkat and HL-60 cancer cells lines in vitro. A detailed in silico analysis on OD-series molecules to evaluate their plausible anticancer mechanism & potential, as well as their in situ ADMET profile study is reported here. The DFT analysis, molecular modelling and molecular dynamics (MD) collectively establishes Topoisomerase-I & α-Tubulin proteins to be the putative target responsible for the cytotoxic activities of OD-B. Orthodiffene series molecules found to be abiding by Lipinksi's rule of 5 for orally bioavailable drug molecule. The present data & study are useful for further exploration of developing new chemical entities based on the structures of OD-series molecules as dual-target inhibitors of Topo-I & tubulin proteins with better efficacies.
Collapse
Affiliation(s)
| | - Akanksha Sharma
- Department of Chemistry, Central University of Karnataka, Kalaburagi, India
| | - Harish Holla
- Department of Chemistry, Central University of Karnataka, Kalaburagi, India
| |
Collapse
|
14
|
Yao Y, Huang T, Wang Y, Wang L, Feng S, Cheng W, Yang L, Duan Y. Angiogenesis and anti-leukaemia activity of novel indole derivatives as potent colchicine binding site inhibitors. J Enzyme Inhib Med Chem 2022; 37:652-665. [PMID: 35109719 PMCID: PMC8820799 DOI: 10.1080/14756366.2022.2032688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The screened compound DYT-1 from our in-house library was taken as a lead (inhibiting tubulin polymerisation: IC50=25.6 µM, anti-angiogenesis in Zebrafish: IC50=38.4 µM, anti-proliferation against K562 and Jurkat: IC50=6.2 and 7.9 µM, respectively). Further investigation of medicinal chemistry conditions yielded compound 29e (inhibiting tubulin polymerisation: IC50=4.8 µM and anti-angiogenesis in Zebrafish: IC50=3.6 µM) based on tubulin and zebrafish assays, which displayed noteworthily nanomolar potency against a variety of leukaemia cell lines (IC50= 0.09–1.22 µM), especially K562 cells where apoptosis was induced. Molecular docking, molecular dynamics (MD) simulation, radioligand binding assay and cellular microtubule networks disruption results showed that 29e stably binds to the tubulin colchicine site. 29e significantly inhibited HUVEC tube formation, migration and invasion in vitro. Anti-angiogenesis in vivo was confirmed by zebrafish xenograft. 29e also prominently blocked K562 cell proliferation and metastasis in blood vessels and surrounding tissues of the zebrafish xenograft model. Together with promising physicochemical property and metabolic stability, 29e could be considered an effective anti-angiogenesis and -leukaemia drug candidate that binds to the tubulin colchicine site.
Collapse
Affiliation(s)
- Yongfang Yao
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Ministry of Education of China, Key Laboratory of Advanced Drug Preparation Technologies (Zhengzhou University), Zhengzhou, China
| | - Tao Huang
- Medical School, Huanghe Science and Technology University, Zhengzhou, Henan Province, P.R China
| | - Yuyang Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Longfei Wang
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Siqi Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Ministry of Education of China, Key Laboratory of Advanced Drug Preparation Technologies (Zhengzhou University), Zhengzhou, China
| | - Weyland Cheng
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Longhua Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.,Ministry of Education of China, Key Laboratory of Advanced Drug Preparation Technologies (Zhengzhou University), Zhengzhou, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Pediatric Hematology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
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
|
16
|
Sun M, Wang Y, Yuan M, Zhao Q, Zhang Y, Yao Y, Duan Y. Angiogenesis, Anti-Tumor, and Anti-Metastatic Activity of Novel α-Substituted Hetero-Aromatic Chalcone Hybrids as Inhibitors of Microtubule Polymerization. Front Chem 2021; 9:766201. [PMID: 34900935 PMCID: PMC8652888 DOI: 10.3389/fchem.2021.766201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
A library of new heteroaromatic ring-linked chalcone analogs were designed and synthesized of these, compound 7m with α-CH3 substitution and bearing a benzofuran ring, displaying the most potent activity, with IC50 values of 0.07–0.183 µM against three cancer cells. Its low cytotoxicity toward normal human cells and strong potency on drug-resistant cells revealed the possibility for cancer therapy. It also could moderately inhibit in vitro tubulin polymerization with an IC50 value of 12.23 µM, and the disruption of cellular architecture in MCF-7 cells was observed by an immunofluorescence assay. Cellular-based mechanism studies elucidated that 7m arrested the cell cycle at the G2/M phase and induced apoptosis by regulating the expression levels of caspases and PARP protein. Importantly, the compound 7 m was found to inhibit HUVEC tube formation, migration, and invasion in vitro. In vivo assay showed that 7m could effectively destroy angiogenesis of zebrafish embryos. Furthermore, our data suggested that treatment with 7m significantly reduced MCF-7 cell metastasis and proliferation in vitro and in zebrafish xenograft. Collectively, this work showed that chalcone hybrid 7m deserves further investigation as dual potential tubulin polymerization and angiogenesis inhibitor.
Collapse
Affiliation(s)
- Moran Sun
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yuyang Wang
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Minghua Yuan
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Qing Zhao
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yixin Zhang
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yongfang Yao
- School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou, China.,School of Pharmaceutical Sciences and Institute of Drug Discovery and Development, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Pérez-Villanueva J, Matadamas-Martínez F, Yépez-Mulia L, Pérez-Koldenkova V, Leyte-Lugo M, Rodríguez-Villar K, Cortés-Benítez F, Macías-Jiménez AP, González-Sánchez I, Romero-Velásquez A, Palacios-Espinosa JF, Soria-Arteche O. Synthesis and Cytotoxic Activity of Combretastatin A-4 and 2,3-Diphenyl-2 H-indazole Hybrids. Pharmaceuticals (Basel) 2021; 14:ph14080815. [PMID: 34451912 PMCID: PMC8401203 DOI: 10.3390/ph14080815] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/05/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Cancer is the second leading cause of death, after cardiovascular diseases. Different strategies have been developed to treat cancer; however, chemotherapy with cytotoxic agents is still the most widely used treatment approach. Nevertheless, drug resistance to available chemotherapeutic agents is still a serious problem, and the development of new active compounds remains a constant need. Taking advantage of the molecular hybridization approach, in the present work we designed, synthesized, and tested the cytotoxic activity of two hybrid compounds and seven derivatives based on the structure of combretastatin A-4 and 2,3-diphenyl-2H-indazole. Practical modifications of reported synthetic protocols for 2-pheny-2H-indazole and 2,3-dipheny-2H-indazole derivatives under microwave irradiation were implemented. The cytotoxicity assays showed that our designed hybrid compounds possess strong activity, especially compound 5, which resulted even better than the reference drug cisplatin against HeLa and SK-LU-1 cells (IC50 of 0.16 and 6.63 µM, respectively), and it had similar potency to the reference drug imatinib against K562 cells. Additionally, in silico and in vitro studies strongly suggest tubulin as the molecular target for hybrid compound 5.
Collapse
Affiliation(s)
- Jaime Pérez-Villanueva
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (F.M.-M.); (F.C.-B.); (A.P.M.-J.); (J.F.P.-E.); (O.S.-A.)
- Correspondence: (J.P.-V.); (L.Y.-M.); Tel.: +52-5-54-83-72-59 (J.P.-V.); Fax: +52-5-55-94-79-29 (J.P.-V.)
| | - Félix Matadamas-Martínez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (F.M.-M.); (F.C.-B.); (A.P.M.-J.); (J.F.P.-E.); (O.S.-A.)
- Maestría y Doctorado en Ciencias Farmacéuticas, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico
| | - Lilián Yépez-Mulia
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico
- Correspondence: (J.P.-V.); (L.Y.-M.); Tel.: +52-5-54-83-72-59 (J.P.-V.); Fax: +52-5-55-94-79-29 (J.P.-V.)
| | - Vadim Pérez-Koldenkova
- Laboratorio Nacional de Microscopía Avanzada, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México 06720, Mexico;
| | - Martha Leyte-Lugo
- Catedrático CONACYT Comisionado a Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (M.L.-L.); (I.G.-S.)
| | - Karen Rodríguez-Villar
- Doctorado en Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana (UAM), Ciudad de México 04960, Mexico;
| | - Francisco Cortés-Benítez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (F.M.-M.); (F.C.-B.); (A.P.M.-J.); (J.F.P.-E.); (O.S.-A.)
| | - Ana Perla Macías-Jiménez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (F.M.-M.); (F.C.-B.); (A.P.M.-J.); (J.F.P.-E.); (O.S.-A.)
| | - Ignacio González-Sánchez
- Catedrático CONACYT Comisionado a Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (M.L.-L.); (I.G.-S.)
| | - Ariana Romero-Velásquez
- Maestría en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México 04510, Mexico;
| | - Juan Francisco Palacios-Espinosa
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (F.M.-M.); (F.C.-B.); (A.P.M.-J.); (J.F.P.-E.); (O.S.-A.)
| | - Olivia Soria-Arteche
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco (UAM-X), Ciudad de México 04960, Mexico; (F.M.-M.); (F.C.-B.); (A.P.M.-J.); (J.F.P.-E.); (O.S.-A.)
| |
Collapse
|
18
|
Song F, Bian Y, Liu J, Li Z, Zhao L, Fang J, Lai Y, Zhou M. Indole Alkaloids, Synthetic Dimers and Hybrids with Potential In Vivo Anticancer Activity. Curr Top Med Chem 2021; 21:377-403. [PMID: 32901583 DOI: 10.2174/1568026620666200908162311] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022]
Abstract
Indole, a heterocyclic organic compound, is one of the most promising heterocycles found in natural and synthetic sources since its derivatives possess fascinating structural diversity and various therapeutic properties. Indole alkaloids, synthetic dimers and hybrids could act on diverse targets in cancer cells, and consequently, possess potential antiproliferative effects on various cancers both in vitro and in vivo. Vinblastine, midostaurin, and anlotinib as the representative of indole alkaloids, synthetic dimers and hybrids respectively, have already been clinically applied to treat many types of cancers, demonstrating indole alkaloids, synthetic dimers and hybrids are useful scaffolds for the development of novel anticancer agents. Covering articles published between 2010 and 2020, this review emphasizes the recent development of indole alkaloids, synthetic dimers and hybrids with potential in vivo therapeutic application for cancers.
Collapse
Affiliation(s)
- Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Yunqiang Bian
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Jing Liu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Zhenghua Li
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Li Zhao
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Junman Fang
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Yonghong Lai
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Meng Zhou
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| |
Collapse
|
19
|
Sun M, Zhang Y, Qin J, Ba M, Yao Y, Duan Y, Liu H, Yu D. Synthesis and biological evaluation of new 2-methoxyestradiol derivatives: Potent inhibitors of angiogenesis and tubulin polymerization. Bioorg Chem 2021; 113:104988. [PMID: 34034135 DOI: 10.1016/j.bioorg.2021.104988] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 11/17/2022]
Abstract
Here, we report the structural optimization of a hit natural compound, 2-ME2 (2-methoxyestradiol), which exhibited inhibitory activity but low potency on tubulin polymerization, anti- angiogenesis, MCF-7 proliferation and metastasis in vitro and in vivo. A novel series of 3,17-modified and 17-modified analogs of 2-ME2 were synthesized and investigated for their antiproliferative activity against MCF-7 and another five different human cancer cell lines leading to the discovery of 9i. 9i bind to tubulin colchicine site tightly, inhibited tubulin polymerization and disrupted cellular microtubule networks. Cellular mechanism studies revealed that 9i could induce G2/M phase arrest by down-regulated expression of p-Cdc2, P21 and cell apoptosis by regulating apoptosis-related proteins (Parp, Caspase families) in a dose-dependent manner. Importantly, 9i significantly inhibited HUVEC tube formation, proliferation, migration and invasion. The inhibitory effect against angiogenesis in vivo was confirmed by zebrafish xenograft. Furthermore, 9i could effectively inhibit the proliferation and metastasis of MCF-7 cells in vitro and in zebrafish xenograft. The satisfactory physicochemical property and metabolic stability of 9i further indicated that it can act as a promising and potent anti-angiogenesis, inhibiting proliferation and metastasis of breast cancer agent via targeting tubulin colchicine binding site.
Collapse
Affiliation(s)
- Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yixin Zhang
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jinling Qin
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Mengyu Ba
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yongfang Yao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China.
| | - Hongmin Liu
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Dequan Yu
- Chinese Academy of Medical Sciences, Beijing 100021,China
| |
Collapse
|
20
|
Xia LY, Zhang YL, Yang R, Wang ZC, Lu YD, Wang BZ, Zhu HL. Tubulin Inhibitors Binding to Colchicine-Site: A Review from 2015 to 2019. Curr Med Chem 2021; 27:6787-6814. [PMID: 31580244 DOI: 10.2174/0929867326666191003154051] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/25/2019] [Accepted: 08/22/2019] [Indexed: 11/22/2022]
Abstract
Due to the three domains of the colchicine-site which is conducive to the combination with small molecule compounds, colchicine-site on the tubulin has become a common target for antitumor drug development, and accordingly, a large number of tubulin inhibitors binding to the colchicine-site have been reported and evaluated over the past years. In this study, tubulin inhibitors targeting the colchicine-site and their application as antitumor agents were reviewed based on the literature from 2015 to 2019. Tubulin inhibitors were classified into ten categories according to the structural features, including colchicine derivatives, CA-4 analogs, chalcone analogs, coumarin analogs, indole hybrids, quinoline and quinazoline analogs, lignan and podophyllotoxin derivatives, phenothiazine analogs, N-heterocycle hybrids and others. Most of them displayed potent antitumor activity, including antiproliferative effects against Multi-Drug-Resistant (MDR) cell lines and antivascular properties, both in vitro and in vivo. In this review, the design, synthesis and the analysis of the structure-activity relationship of tubulin inhibitors targeting the colchicine-site were described in detail. In addition, multi-target inhibitors, anti-MDR compounds, and inhibitors bearing antitumor activity in vivo are further listed in tables to present a clear picture of potent tubulin inhibitors, which could be beneficial for medicinal chemistry researchers.
Collapse
Affiliation(s)
- Lin-Ying Xia
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Ya-Liang Zhang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Rong Yang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Zhong-Chang Wang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Ya-Dong Lu
- Neonatal Medical Center, Children’s Hospital of Nanjing Medical University, Nanjing 210008, P.R. China
| | - Bao-Zhong Wang
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China,State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210023, P.R. China
| | - Hai-Liang Zhu
- Zhengzhou Children’s Hospital, Zhengzhou 450018, P.R. China
| |
Collapse
|
21
|
Ultrasound assisted synthesis of tetrazole based pyrazolines and isoxazolines as potent anticancer agents via inhibition of tubulin polymerization. Bioorg Med Chem Lett 2020; 30:127592. [PMID: 33010448 DOI: 10.1016/j.bmcl.2020.127592] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/18/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
In search of new active molecules against MCF-7, A549 and HepG2, tetrazole based pyrazoline and isoxazoline derivatives under both conventional and ultrasonic irradiation method were designed and efficiently synthesized. Structures of newly synthesized compounds 5a-h and 6a-h were characterized by 1H NMR, 13C NMR, MS and elemental analysis. Several derivatives were found to be excellent cytotoxic against MCF-7, A549 and HepG2 cell lines characterized by lower IC50 values (0.78-3.12 µg/mL). Compounds 5b and 5c demonstrated an antiproliferative effect comparable to that of CA-4. Western blot analysis revealed that, reported compounds accumulate more tubulin in the soluble fraction. Docking studies suggested that, binding of these compounds mimics at the colchicine site of tubulin. In vitro study revealed that the tetrazole based pyrazolines and isoxazolines may possess ideal structural requirements for further development of novel therapeutic agents.
Collapse
|
22
|
Wang Y, Yao Y, Zhu HL, Duan Y. Butterfly Structure: A Privileged Scaffold Targeting Tubulin-Colchicine Binding Site. Curr Top Med Chem 2020; 20:1505-1508. [PMID: 32543362 DOI: 10.2174/1568026620999200616132924] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022]
Abstract
:
Butterfly-shaped structure, as a novel scaffold with an attractive and certain shape, has been
widely used in new drug discovery. Tubulin, composing of α- and β-tubulin heterodimers, plays a key
role in mitosis and cell division which are regarded as an excellent target for cancer therapy. Currently, a
series of butterfly shape diaryl heterocyclic compounds have been reported with strong potential against
the tubulin-colchicine binding site. It is with one ring buried in the β subunit, another ring interacts with
the α subunit and the main body is located in the flat pocket. Here, we firstly introduce the concept of
butterfly structure for the tubulin inhibitors, focusing on the latest advancements in a variety of molecules
bearing butterfly structure, and then highlight the challenges and future direction of butterfly structure-
based tubulin-colchicine binding site inhibitors.
Collapse
Affiliation(s)
- Yingge Wang
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Yongfang Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | - Hai-Liang Zhu
- Henan Provincial Key Laboratory Of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| | - Yongtao Duan
- Henan Provincial Key Laboratory Of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou 450018, China
| |
Collapse
|
23
|
Karatoprak GŞ, Küpeli Akkol E, Genç Y, Bardakcı H, Yücel Ç, Sobarzo-Sánchez E. Combretastatins: An Overview of Structure, Probable Mechanisms of Action and Potential Applications. Molecules 2020; 25:E2560. [PMID: 32486408 PMCID: PMC7321081 DOI: 10.3390/molecules25112560] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/08/2023] Open
Abstract
Combretastatins are a class of closely related stilbenes (combretastatins A), dihydrostilbenes (combretastatins B), phenanthrenes (combretastatins C) and macrocyclic lactones (combretastatins D) found in the bark of Combretum caffrum (Eckl. & Zeyh.) Kuntze, commonly known as the South African bush willow. Some of the compounds in this series have been shown to be among the most potent antitubulin agents known. Due to their structural simplicity many analogs have also been synthesized. Combretastatin A4 phosphate is the most frequently tested compounds in preclinical and clinical trials. It is a water-soluble prodrug that the body can rapidly metabolize to combretastatin A4, which exhibits anti-tumor properties. In addition, in vitro and in vivo studies on combretastatins have determined that these compounds also have antioxidant, anti-inflammatory and antimicrobial effects. Nano-based formulations of natural or synthetic active agents such as combretastatin A4 phosphate exhibit several clear advantages, including improved low water solubility, prolonged circulation, drug targeting properties, enhanced efficiency, as well as fewer side effects. In this review, a synopsis of the recent literature exploring the combretastatins, their potential effects and nanoformulations as lead compounds in clinical applications is provided.
Collapse
Affiliation(s)
- Gökçe Şeker Karatoprak
- Department of Pharmacognosy, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey;
| | - Esra Küpeli Akkol
- Department of Pharmacognosy Faculty of Pharmacy, Gazi University, 06330 Ankara, Turkey
| | - Yasin Genç
- Department of Pharmacognosy, Faculty of Pharmacy, Hacettepe University, 06100 Sıhhiye, Ankara, Turkey;
| | - Hilal Bardakcı
- Department of Pharmacognosy, Faculty of Pharmacy, Acibadem Mehmet Ali Aydınlar University, 34752 Istanbul, Turkey;
| | - Çiğdem Yücel
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Erciyes University, 38039 Kayseri, Turkey;
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación e Innovación en Salud, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 8330507, Chile;
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
24
|
Song ZL, Zhu Y, Liu JR, Guo SK, Gu YC, Han X, Dong HQ, Sun Q, Zhang WH, Zhang MZ. Diversity-oriented synthesis and antifungal activities of novel pimprinine derivative bearing a 1,3,4-oxadiazole-5-thioether moiety. Mol Divers 2020; 25:205-221. [PMID: 32056130 DOI: 10.1007/s11030-020-10048-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/05/2020] [Indexed: 10/25/2022]
Abstract
Based on the strategy of diversity-oriented synthesis and the structures of natural product pimprinine and streptochlorin, two series of novel pimprinine derivatives containing 1,3,4-oxadiazole-5-thioether moieties were efficiently synthesized under the optimized reaction conditions. Biological assays conducted at Syngenta showed the designed derivatives displayed an altered pattern of biological activity, of which 5h was identified as the most promising compound with strong activity against Pythium dissimile and also a broad antifungal spectrum in primary screening. Further structural optimization of pimprinine and streptochlorin derivatives is well under way, aiming to discover synthetic analogues with improved antifungal activity. Two series of novel pimprinine derivatives containing 1,3,4-oxadiazole-5-thioether moieties were efficiently synthesized through diversity-oriented synthesis strategy under the optimized conditions. Biological assays showed the designed derivatives exhibited potential activity.
Collapse
Affiliation(s)
- Zi-Long Song
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yun Zhu
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jing-Rui Liu
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shu-Ke Guo
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yu-Cheng Gu
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, UK
| | - Xinya Han
- School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243002, China.
| | - Hong-Qiang Dong
- College of Plant Science, Tarim University, Alaer, 843300, Xinjiang, China
| | - Qi Sun
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemistry and Environmental Engineering, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Wei-Hua Zhang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Ming-Zhi Zhang
- Jiangsu Key Laboratory of Pesticide Science, College of Sciences, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
25
|
O'Boyle NM, Ana G, Kelly PM, Nathwani SM, Noorani S, Fayne D, Bright SA, Twamley B, Zisterer DM, Meegan MJ. Synthesis and evaluation of antiproliferative microtubule-destabilising combretastatin A-4 piperazine conjugates. Org Biomol Chem 2020; 17:6184-6200. [PMID: 31173031 DOI: 10.1039/c9ob00558g] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microtubules are a validated clinical target for the treatment of many cancers. We describe the design, synthesis, biochemical evaluation, and molecular modelling studies of a series of analogues of the microtubule-destabilising agent, combretastatin A-4 (CA-4). Our series of 33 novel compounds contain the CA-4 core structure with modifications to the stilbene linking group, and are predominantly piperazine derivatives. Synthesis was achieved in a two-step process by firstly obtaining the acrylic acid via a Perkin reaction using microwave enhanced synthesis, followed by coupling using either DCC or Mukaiyama's reagent. All target compounds were screened for antiproliferative activity in MCF-7 breast cancer cells. Hydroxyl derivative (E)-3-(4-hydroxy-3-methoxyphenyl)-1-(4-phenylpiperazin-1-yl)-2-(3,4,5-trimethoxyphenyl) propenone (4m) displayed potent antiproliferative activity (IC50 = 190 nM). Two amino-containing derivatives, (E)-3-(3-amino-4-methoxyphenyl)-1-(4-phenylpiperazin-1-yl)-2-(3,4,5-trimethoxyphenyl)prop-2-en-1-one (4q) and (E)-3-(3-amino-4-methoxyphenyl)-1-(4-(p-tolyl)piperazin-1-yl)-2-(3,4,5-trimethoxyphenyl)prop-2-en-1-one (4x), were the most potent with IC50 values of 130 nM and 83 nM respectively. Representative compounds were shown to depolymerise tubulin, induce G2/M arrest and apoptosis in MCF-7 cells but not peripheral blood mononuclear cells, and induce cleavage of the DNA repair enzyme poly ADP ribose polymerase (PARP) in MCF-7 cells. Modelling studies predict that the compounds bind to tubulin within the colchicine-binding site. These compounds are a valuable addition to the library of CA-4 analogues and 4m, 4q and 4x will be developed further as novel, water-soluble molecules targeting microtubules.
Collapse
Affiliation(s)
- Niamh M O'Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2 D02 R590, Ireland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The stilbenoid combretastatin and its derivatives are potent inhibitors of angiogenesis and cell proliferation and induce apoptosis. They disrupt cytoskeletal dynamics and modulate cell morphology, motility, and invasion. Hence they have been viewed as potential as anticancer agents. The impediments of poor solubility and bioavailability and the spontaneous geometric isomerisation of combretastatin into an inactive form have led to intensive efforts towards evolving novel analogues to provide more efficacious biological outcome. Importantly, isomerically stable and biologically active cis-restricted analogues have been synthesised and tested. However, very few analogues have been tested in preclinical models to assess their effects on processes relevant to cancer development and progression. Hence the accent here is on the signalling systems operated by the new derivatives and their biological effects with reference to cancer progression. Combretastatins modulate an extensive network of signalling emphasising their varied versatility. Harnessing these systems and accentuating or counteracting aberrant signalling could open potential avenues of approach to the designing of novel derivatives with enhanced performance. The import of mammalian target of rapamycin pathway, which co-ordinates growth factor receptor signalling, epithelial-mesenchymal transition activation and angiogenic signalling, is emphasised. It may be viewed as a prime target for allosteric inhibition in combination with combretastatin analogues to ascertain their potential in cancer control.
Collapse
Affiliation(s)
- Gajanan V Sherbet
- School of Engineering, University of Newcastle Upon Tyne, Newcastle Upon Tyne, UK.,The Institute for Molecular Medicine, Huntington Beach, California
| |
Collapse
|
27
|
Duan Y, Liu W, Tian L, Mao Y, Song C. Targeting Tubulin-colchicine Site for Cancer Therapy: Inhibitors, Antibody- Drug Conjugates and Degradation Agents. Curr Top Med Chem 2019; 19:1289-1304. [PMID: 31210108 DOI: 10.2174/1568026619666190618130008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/22/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022]
Abstract
Microtubules are essential for the mitotic division of cells and have been an attractive target
for antitumour drugs due to the increased incidence of cancer and significant mitosis rate of tumour cells.
In the past few years, tubulin-colchicine binding site, as one of the three binding pockets including taxol-,
vinblastine- and colchicine-binding sites, has been focused on to design tubulin-destabilizing agents including
inhibitors, antibody-drug conjugates and degradation agents. The present review is the first to
cover a systemic and recent synopsis of tubulin-colchicine binding site agents. We believe that it would
provide an increase in our understanding of receptor-ligand interaction pattern and consciousness of a
series of challenges about tubulin target druggability.
Collapse
Affiliation(s)
- Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Liang Tian
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yanna Mao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Chuanjun Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
28
|
Xia LY, Yang R, Zhang YL, Chu YC, Qi YL, Man RJ, Wang ZC, Wang BZ, Zhu HL. Design, synthesis, and biological evaluation of 2,3-diphenyl-cycloalkyl pyrazole derivatives as potential tubulin polymerization inhibitors. Chem Biol Drug Des 2019; 94:1894-1904. [PMID: 31106514 DOI: 10.1111/cbdd.13565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/06/2019] [Accepted: 05/13/2019] [Indexed: 01/06/2023]
Abstract
Several novel cycloalkyl-fused 2,3-diaryl pyrazole derivatives were designed, synthesized, and evaluated as potential anti-tubulin agents. Compound A10 exhibited the most potent antiproliferative activity against a panel of cancer lines (IC50 = 0.78-2.42 μM) and low cytotoxicity against 293T & L02 (CC50 values of 131.74 and 174.89 μM, respectively). Moreover, A10 displayed inhibition of tubulin polymerization in vitro, arrested the G2/M phase of the cell cycle, changed morphology of tubulin, increased intracellular reactive oxygen species, and induced apoptosis of HeLa cells. Docking simulation and 3D-QSAR models were performed to elaborate on the anti-tubulin mechanism of the derivatives. The inhibition of monoclonal colony formation provided more intuitional data to verify the possibility of A10 as a novel tubulin assembling inhibitor.
Collapse
Affiliation(s)
- Lin-Ying Xia
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Rong Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ya-Liang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yi-Chun Chu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ya-Lin Qi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ruo-Jun Man
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Guangxi Biological Polysaccharide Separation, Purification and Modification Research Platform, Guangxi University for Nationalities, Nanning, China
| | - Zhong-Chang Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Bao-Zhong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
29
|
Duan YT, Sangani CB, Liu W, Soni KV, Yao Y. New Promises to Cure Cancer and Other Genetic Diseases/Disorders: Epi-drugs Through Epigenetics. Curr Top Med Chem 2019; 19:972-994. [DOI: 10.2174/1568026619666190603094439] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/05/2019] [Accepted: 05/27/2019] [Indexed: 12/13/2022]
Abstract
All the heritable alterations in gene expression and chromatin structure due to chemical modifications that do not involve changes in the primary gene nucleotide sequence are referred to as epigenetics. DNA methylation, histone modifications, and non-coding RNAs are distinct types of epigenetic inheritance. Epigenetic patterns have been linked to the developmental stages, environmental exposure, and diet. Therapeutic strategies are now being developed to target human diseases such as cancer with mutations in epigenetic regulatory genes using specific inhibitors. Within the past two decades, seven epigenetic drugs have received regulatory approval and many others show their candidature in clinical trials. The current article represents a review of epigenetic heritance, diseases connected with epigenetic alterations and regulatory approved epigenetic drugs as future medicines.
Collapse
Affiliation(s)
- Yong-Tao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Chetan B. Sangani
- Shri Maneklal M. Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya University, Gandhinagar, Gujarat, 362024, India
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Zhengzhou Children's Hospital, Zhengzhou University, Zhengzhou 450018, China
| | - Kunjal V. Soni
- Shri Maneklal M. Patel Institute of Sciences and Research, Kadi Sarva Vishwavidyalaya University, Gandhinagar, Gujarat, 362024, India
| | - Yongfang Yao
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
30
|
Synthesis and in vitro evaluation of substituted tetrahydroquinoline-isoxazole hybrids as anticancer agents. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02363-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
31
|
Abdelbaset MS, Abdel-Aziz M, Ramadan M, Abdelrahman MH, Abbas Bukhari SN, Ali TF, Abuo-Rahma GEDA. Discovery of novel thienoquinoline-2-carboxamide chalcone derivatives as antiproliferative EGFR tyrosine kinase inhibitors. Bioorg Med Chem 2019; 27:1076-1086. [DOI: 10.1016/j.bmc.2019.02.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
|
32
|
Indole derivatives as multifunctional drugs: Synthesis and evaluation of antioxidant, photoprotective and antiproliferative activity of indole hydrazones. Bioorg Chem 2019; 85:568-576. [PMID: 30825715 DOI: 10.1016/j.bioorg.2019.02.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/28/2019] [Accepted: 02/03/2019] [Indexed: 01/09/2023]
Abstract
Two series of indole derivatives 4-17, 20-22 were easily prepared and assayed for their radical-scavenging ability. Arylidene-1H-indole-2-carbohydrazones showed different extent antioxidant activity in DPPH, FRAP and ORAC assays. Good antioxidant activity is related to the number and position of hydroxyl groups on the arylidene moiety as well as to the presence of methoxy or 4-(diethylamino) group. On the contrary low antioxidant activity is showed by the isomeric 1H-indol-2-yl(methylene)-benzohydrazides. Furthermore, hydrazones 4-17 showed photoprotective capacities with satisfactory in vitro SPF as compared to the commercial PBSA sunscreen filter. The indole 16 and 17, showing the best antioxidant and photoprotective profile, were included in different formulation and their topical release was evaluated. Varying the formulation composition, it was possible to optimize skin adsorption and solubility of the active indole in the formulation. The antiproliferative effect of the hydrazones 4-17 was tested on human erythroleukemia K562 and melanoma Colo-38 cells. Hydrazones 11, 16 and 17 showed growth inhibition at sub micromolar concentrations on both cell lines. These results indicate indole hydrazones as potential multifunctional molecules especially in the treatment of neoplastic diseases being the good antioxidant properties of 16 and 17 correlated to their high antiproliferative activity.
Collapse
|
33
|
Abdelbaset MS, Abuo-Rahma GEDA, Abdelrahman MH, Ramadan M, Youssif BG, Bukhari SNA, Mohamed MF, Abdel-Aziz M. Novel pyrrol-2(3H)-ones and pyridazin-3(2H)-ones carrying quinoline scaffold as anti-proliferative tubulin polymerization inhibitors. Bioorg Chem 2018; 80:151-163. [DOI: 10.1016/j.bioorg.2018.06.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 10/14/2022]
|
34
|
Fan A, Zhang Y, Zhang Q, Wei J, Lu X, Ren G, Zhao D, Li N, Zhu H, Chen X. Evaluation of the pharmacokinetics, tissue distribution and excretion studies of YMR-65, a tubulin polymerization inhibitor with potential anticancer activity, in rats using UPLC-MS/MS. Xenobiotica 2017; 48:920-926. [PMID: 29050520 DOI: 10.1080/00498254.2017.1380865] [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] [Indexed: 12/20/2022]
Abstract
1. YMR-65, 5-(5-bromo-1-methyl-1H-indol-3-yl)-3-(3-methoxyphenyl)-4, 5-dihydro-1H-pyrazole-1-carboxamide, is a new tubulin polymerization inhibitor with encouraging anticancer activity. 2. The validated ultra-performance liquid chromatography-tandem mass spectrometer (UPLC-MS/MS) method was successfully applied to the pharmacokinetics, tissue distribution and excretion study of YMR-65 after oral and intravenous administration. The area under concentration-time curve (AUC0-∞) for YMR-65 were 151.67 ± 54.48 and 459.45 ± 49.23 ng/ml*h for oral and intravenous administration at the dosage of 1.5 mg/kg, respectively and the oral bioavailability was about 33.01%. Moreover, YMR-65 was extensively distributed in heart, liver, spleen, lung, kidney, stomach, intestine and testis and the highest were detected in heart, followed by stomach, intestine and liver. The majority of YMR-65 was excreted via feces and its accumulative excretion ratio during the period of 96 h was 19.83 ± 3.01%, but only 1.54 ± 0.37 and 0.215 ± 0.026% for urine within 96 h and bile within 10 h after intravenous administration, respectively, though the fecal and urine excretion were incomplete within 96 h. 3. In summary, this study defined the pharmacokinetic characteristics of YMR-65 in vivo and the important data can be a useful resource for further research and development.
Collapse
Affiliation(s)
- Ali Fan
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Yaliang Zhang
- b State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University , Nanjing , P.R. China
| | - Qing Zhang
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Jiali Wei
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Xiaoyu Lu
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Guanghui Ren
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Di Zhao
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Ning Li
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| | - Hailiang Zhu
- b State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University , Nanjing , P.R. China
| | - Xijing Chen
- a Clinical Pharmacokinetics Laboratory, China Pharmaceutical University , Nanjing , P.R. China and
| |
Collapse
|
35
|
Structure-Activity Relationship Studies of β-Lactam-azide Analogues as Orally Active Antitumor Agents Targeting the Tubulin Colchicine Site. Sci Rep 2017; 7:12788. [PMID: 28986548 PMCID: PMC5630639 DOI: 10.1038/s41598-017-12912-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 09/12/2017] [Indexed: 11/29/2022] Open
Abstract
We have synthesized a series of new β-lactam-azide derivatives as orally active anti-tumor agents by targeting tubulin colchicine binding site and examined their structure activity relationship (SAR). Among them, compound 28 exhibited the most potent antiproliferative activity against MGC-803 cells with an IC50 value of 0.106 μM by induction of G2/M arrest and apoptosis and inhibition of the epithelial to mesenchymal transition. 28 acted as a novel inhibitor of tubulin polymerization by its binding to the colchicine site. SAR analysis revealed that a hydrogen atom at the C-3 position of the β-lactam was required for the potent antiproliferative activity of β-lactam-azide derivatives. Oral administration of compound 28 also effectively inhibited MGC-803 xenograft tumor growth in vivo in nude mice without causing significant loss of body weight. These results suggested that compound 28 is a promising orally active anticancer agent with potential for development of further clinical applications.
Collapse
|
36
|
Xu Q, Bao K, Sun M, Xu J, Wang Y, Tian H, Zuo D, Guan Q, Wu Y, Zhang W. Design, synthesis and structure-activity relationship of 3,6-diaryl-7H-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazines as novel tubulin inhibitors. Sci Rep 2017; 7:11997. [PMID: 28931885 PMCID: PMC5607265 DOI: 10.1038/s41598-017-10860-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/11/2017] [Indexed: 12/18/2022] Open
Abstract
A novel series of 3,6-diaryl-7H-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazines were designed, synthesized and biologically evaluated as vinylogous CA-4 analogues, which involved a rigid [1,2,4]triazolo[3,4-b][1,3,4]thiadiazine scaffold to fix the configuration of (Z,E)-butadiene linker of A-ring and B-ring. Among these rigidly vinylogous CA-4 analogues, compounds 4d, 5b, 5i, 6c, 6e, 6g, 6i and 6k showed excellent antiproliferative activities against SGC-7901, A549 and HT-1080 cell lines with IC50 values at the nanomolar level. Compound 6i showed the most highly active antiproliferative activity against the three human cancer cell lines with an IC50 values of 0.011-0.015 µM, which are comparable to those of CA-4 (IC50 = 0.009-0.013 µM). Interestingly, SAR studies revealed that 3,4-methylenedioxyphenyl, 3,4-dimethoxyphenyl, 3-methoxyphenyl and 4-methoxyphenyl could replace the classic 3,4,5-trimethoxyphenyl in CA-4 structure and keep antiproliferative activity in this series of designed compounds. Tubulin polymerization experiments showed that 6i could effectively inhibit tubulin polymerization, which was corresponded with CA-4, and immunostaining experiments suggested that 6i significantly disrupted microtubule/tubulin dynamics. Furthermore, 6i potently induced cell cycle arrest at G2/M phase in SGC-7901 cells. Competitive binding assays and docking studies suggested that compound 6i binds to the tubulin perfectly at the colchicine binding site. Taken together, these results revealed that 6i may become a promising lead compound for new anticancer drugs discovery.
Collapse
Affiliation(s)
- Qile Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Kai Bao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Maolin Sun
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Jingwen Xu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yueting Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Haiqiu Tian
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
37
|
Seddigi ZS, Malik MS, Saraswati AP, Ahmed SA, Babalghith AO, Lamfon HA, Kamal A. Recent advances in combretastatin based derivatives and prodrugs as antimitotic agents. MEDCHEMCOMM 2017; 8:1592-1603. [PMID: 30108870 DOI: 10.1039/c7md00227k] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/03/2017] [Indexed: 01/01/2023]
Abstract
The dynamic and crucial role of tubulin in different cellular functions rendered it a promising target in anticancer drug development. Combretastatin A-4 (CA-4), an inhibitor of tubulin polymerization isolated from natural sources, is a lead molecule with significant cytotoxicity against tumour cells. Owing to its non polar nature it exhibits low solubility in natural biological fluids, thereby prompting the development of new CA-4 based derivatives. The modification of this lead molecule was mostly carried out by keeping the crucial cis-orientation of the double bond intact, along with a trimethoxyphenyl aromatic ring, by employing different approaches. The issue of solubility was also addressed by the development of water soluble prodrugs of CA-4. The present review highlights the investigations into the parallel development of both new CA-4 based derivatives and prodrugs in the past few years.
Collapse
Affiliation(s)
- Zaki S Seddigi
- Department of Environmental Health , College of Public Health and Health Informatics , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - M Shaheer Malik
- Science and Technology Unit , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - A Prasanth Saraswati
- Department of Medicinal Chemistry and Pharmacology , CSIR - Indian Institute of Chemical Technology , Hyderabad 500 007 , India . ; ; Tel: +91 40 27193157
| | - Saleh A Ahmed
- Department of Chemistry , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - Ahmed O Babalghith
- Department of Medical Genetics, Faculty of Medicine , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - Hawazen A Lamfon
- Department of Biology , Faculty of Applied Sciences , Umm Al-Qura University , 21955 Makkah , Saudi Arabia
| | - Ahmed Kamal
- Department of Medicinal Chemistry and Pharmacology , CSIR - Indian Institute of Chemical Technology , Hyderabad 500 007 , India . ; ; Tel: +91 40 27193157
| |
Collapse
|
38
|
Zuo D, Pang L, Shen J, Guan Q, Bai Z, Zhang H, Li Y, Lu G, Zhang W, Wu Y. 5-(Furan-2-yl)-4-(3,4,5-trimethoxyphenyl)-3H-1,2-dithiol-3-one oxime (6f), a new synthetic compound, causes human fibrosarcoma HT-1080 cell apoptosis by disrupting tubulin polymerisation and inducing G2/M arrest. Int J Oncol 2017; 50:2069-2078. [PMID: 28440465 DOI: 10.3892/ijo.2017.3963] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/13/2017] [Indexed: 11/06/2022] Open
Abstract
In the current study, we synthesized a series of new compounds targeting tubulin and tested their anti-proliferative activities. Among these new synthetic com-pounds, 5-(furan-2-yl)-4-(3,4,5-trimethoxyphenyl)-3H-1,2-dithiol-3-one oxime (6f) exhibited significant anti-proliferative activity against different human cancer cell lines including human gastric adenocarcinoma SGC-7901, human non-small cell lung cancer A549, and human fibrosarcoma HT-1080. As a result, 6f was selected to further test the sensitivity to different cancer cell lines including human cervical cancer cell line HeLa, human breast cancer cell line MCF-7, non-small cell lung cancer cell line A549, human liver carcinoma cell line HepG-2, human oral squamous cell carcinoma cell lines KB, SGC-7901 and HT-1080. Among these cell lines, HT-1080 and HeLa are the most sensitive. Therefore, HT-1080 was selected to further explore the properties of anti-proliferative activity and the underlying mechanisms. Our data proved that 6f exhibited strong anti-proliferative effects against HT-1080 cells in a time- and dose-dependent manner. We showed that the growth inhibitory effect of 6f in HT-1080 cells was related with microtubule depolymerisation. Molecular docking studies revealed that 6f interacted and bound efficiently with the colchicine-binding site of tubulin. In addition, 6f treatment induced G2/M cell cycle arrest dose-dependently and subsequently induced cell apoptosis. Western blot study indicated that upregulation of cyclin B1 and p-cdc2 was related with G2/M arrest. 6f-induced cell apoptosis was associated with both mitochondrial and death receptor pathway. In conclusion, our data showed that 6f, among the newly synthetic compounds, exhibited highest anti-proliferative activity by disrupting the microtubule polymerisation, causing G2/M arrest and subsequently inducing cell apoptosis in HT-1080 cells. Hence, 6f is a promising microtubule depolymerising agent for the treatment of various cancers especially human fibrosarcoma.
Collapse
Affiliation(s)
- Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Lili Pang
- Department of Clinical Pharmacy, The Sixth People's Hospital of Chengdu, Chengdu, Sichuan 610051, P.R. China
| | - Jiwei Shen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Zhaoshi Bai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Huijuan Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yao Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Guodong Lu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
39
|
Bukhari SNA, Kumar GB, Revankar HM, Qin HL. Development of combretastatins as potent tubulin polymerization inhibitors. Bioorg Chem 2017; 72:130-147. [PMID: 28460355 DOI: 10.1016/j.bioorg.2017.04.007] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 03/22/2017] [Accepted: 04/13/2017] [Indexed: 11/18/2022]
Abstract
The combretastatins are isolated from South African tree combretum caffrum kuntze. The lead compound combretastatin A-4 has displayed remarkable cytotoxic effect in a wide variety of preclinical tumor models and inhibits tubulin polymerization by interacting at colchicine binding site of microtubule. However, the structural simplicity of C A-4 is favorable for synthesis of various derivatives projected to induce rapid and selective vascular shutdown in tumors. Majority of the molecules have shown excellent antiproliferative activity and are able to inhibit tubulin polymerization as well as possible mechanisms of action have been investigated. In this review article, the synthesis and structure-activity relationships of C A-4 and immense number of its synthetic derivatives with various modifications on the A, B-rings, bridge carbons and their anti mitotic activities are discussed.
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China; Department of Pharmaceutical Chemistry, College of Pharmacy, Aljouf University, Aljouf, Sakaka 2014, Saudi Arabia.
| | - Gajjela Bharath Kumar
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| | - Hrishikesh Mohan Revankar
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China
| | - Hua-Li Qin
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan 430070, China.
| |
Collapse
|
40
|
Sakchaisri K, Kim SO, Hwang J, Soung NK, Lee KH, Choi TW, Lee Y, Park CM, Thimmegowda NR, Lee PY, Shwetha B, Srinivasrao G, Pham TTH, Jang JH, Yum HW, Surh YJ, Lee KS, Park H, Kim SJ, Kwon YT, Ahn JS, Kim BY. Anticancer activity of a novel small molecule tubulin inhibitor STK899704. PLoS One 2017; 12:e0173311. [PMID: 28296906 PMCID: PMC5351965 DOI: 10.1371/journal.pone.0173311] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/17/2017] [Indexed: 12/15/2022] Open
Abstract
We have identified the small molecule STK899704 as a structurally novel tubulin inhibitor. STK899704 suppressed the proliferation of cancer cell lines from various origins with IC50 values ranging from 0.2 to 1.0 μM. STK899704 prevented the polymerization of purified tubulin in vitro and also depolymerized microtubule in cultured cells leading to mitotic arrest, associated with increased Cdc25C phosphorylation and the accumulation of both cyclin B1 and polo-like kinase 1 (Plk1), and apoptosis. Unlike many anticancer drugs such as Taxol and doxorubicin, STK899704 effectively displayed antiproliferative activity against multidrug-resistant cancer cell lines. The proposed binding mode of STK899704 is at the interface between αβ-tubulin heterodimer overlapping with the colchicine-binding site. Our in vivo carcinogenesis model further showed that STK 899704 is potent in both the prevention and regression of tumors, remarkably reducing the number and volume of skin tumor by STK899704 treatment. Moreover, it was significant to note that the efficacy of STK899704 was surprisingly comparable to 5-fluorouracil, a widely used anticancer therapeutic. Thus, our results demonstrate the potential of STK899704 to be developed as an anticancer chemotherapeutic and an alternative candidate for existing therapies.
Collapse
Affiliation(s)
- Krisada Sakchaisri
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok, Thailand
| | - Sun-Ok Kim
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Joonsung Hwang
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Nak Kyun Soung
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Kyung Ho Lee
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Tae Woong Choi
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Yongjun Lee
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Chan-Mi Park
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Naraganahalli R. Thimmegowda
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Phil Young Lee
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Bettaswamigowda Shwetha
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Ganipisetti Srinivasrao
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Thi Thu Huong Pham
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
- The Key Laboratory of Enzyme & Protein Technology (KLEPT), VNU University of Science, Vietnam National University, Hanoi, Vietnam
| | - Jae-Hyuk Jang
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Hye-Won Yum
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul, Korea
| | - Kyung S. Lee
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Hwangseo Park
- Department of Bioscience and Biotechnology, Sejong University, Seoul, Korea
| | - Seung Jun Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Yong Tae Kwon
- Department of Biomedical Sciences and Protein Metabolism Medical Research Center, College of Medicine, Seoul National University, Seoul, Korea
| | - Jong Seog Ahn
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| | - Bo Yeon Kim
- Anticancer Agents Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang, Cheongwon, Korea
| |
Collapse
|
41
|
Mutai P, Breuzard G, Pagano A, Allegro D, Peyrot V, Chibale K. Synthesis and biological evaluation of 4 arylcoumarin analogues as tubulin-targeting antitumor agents. Bioorg Med Chem 2017; 25:1652-1665. [DOI: 10.1016/j.bmc.2017.01.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 10/20/2022]
|
42
|
Novel Natural Product- and Privileged Scaffold-Based Tubulin Inhibitors Targeting the Colchicine Binding Site. Molecules 2016; 21:molecules21101375. [PMID: 27754459 PMCID: PMC6273505 DOI: 10.3390/molecules21101375] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 10/10/2016] [Accepted: 10/11/2016] [Indexed: 12/20/2022] Open
Abstract
Tubulin inhibitors are effective anticancer agents, however, there are many limitations to the use of available tubulin inhibitors in the clinic, such as multidrug resistance, severe side-effects, and generally poor bioavailability. Thus, there is a constant need to search for novel tubulin inhibitors that can overcome these limitations. Natural product and privileged structures targeting tubulin have promoted the discovery and optimization of tubulin inhibitors. This review will focus on novel tubulin inhibitors derived from natural products and privileged structures targeting the colchicine binding site on tubulin.
Collapse
|