1
|
Jiang B, Zheng Y, Xue T, Wu J, Song H, Zhou S, Li Y, Gong J, Wei M, Ji X, Wei M, Wang L, Gong J, Liu M, Wang A, Zhang K, Lv K, Zheng Y. Identification of selenium-containing benzamides as potent microtubule-targeting antitumor agents. Bioorg Chem 2025; 159:108355. [PMID: 40090150 DOI: 10.1016/j.bioorg.2025.108355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
IMB5046, a microtubule inhibitor discovered by our team, served as the lead compound for designing a series of selenium-containing benzoates and benzamides. Among these, compound 2g emerged as a lead candidate, demonstrating potent antiproliferative activity. Mechanistic studies revealed that 2g bound to the colchicine site of tubulin, caused G2/M cell cycle arrest, and generated ROS. Notably, 2g exhibited exceptional efficacy in P-gp overexpressing MCF7/ADR and KBV200 cell lines, with drug-resistance indices (DRI) of 0.83 and 0.58, respectively, significantly outperforming colchicine (DRIs: 25.4 and 8.03) and paclitaxel (DRIs: 41.0 and 4.96). In an MCF-7 xenograft model, 2g (25 mg/kg, IP) achieved a tumor growth inhibition rate of 57.2 %, surpassing IMB5046 (47.6 %). To enhance solubility and pharmacokinetics, prodrug 2g-P was developed, showing 69 % bioavailability but reduced in vivo efficacy. Further investigation is warranted to elucidate the factors underlying the discrepancy, such as the efficiency of prodrug-to-drug conversion and intracellular accumulation of active 2g. In summary, our study not only identified a novel selenium-containing lead compound, but also provided important insights into prodrug design. These findings lay a solid foundation for the development of next-generation microtubule-targeting agents capable of overcoming drug resistance.
Collapse
Affiliation(s)
- Bin Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Yijia Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Tiezheng Xue
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Jizhou Wu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Huijuan Song
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Sheng Zhou
- Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China
| | - Yujing Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jiaqi Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meng Wei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xiaorui Ji
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Meijiao Wei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Lujun Wang
- Department of Pharmacy, Medical Supplies Center of PLA General Hospital, China
| | - Jianhua Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingliang Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Apeng Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Kai Zhang
- Department of Pharmaceutical Chemistry, School of Pharmacy, Hebei Medical University, Shijiazhuang 050017, China.
| | - Kai Lv
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanbo Zheng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
2
|
Qu X, Wang Y, Xu Y, Xu L, Ye X, Cai H, Bu L, Zeng Z, Zhou H. Aromatic nitroolefin with inhibition efficacy in triple-negative breast cancer cells by dual targeting RXRα and tubulins. Eur J Med Chem 2025; 289:117486. [PMID: 40090298 DOI: 10.1016/j.ejmech.2025.117486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/18/2025]
Abstract
We previously identified that 1-(2-Nitrovinyl)naphthalene (Z-10) is a ligand of retinoid x receptor α (RXRα) with a potent anti-breast cancer efficacy and revealed that nitro group is an essential pharmacophore in Z-10. In this study, we defined that the double bond of the nitrovinyl group is also vital for Z-10 to bind and activate RXRα. Mechanistically, the double bond has a chemical ability to mediate Z-10's covalent binding of RXRα via the Michael addition reaction with Cys432. By retaining the nitrovinyl group, a series of Z-10 analogues with different aromatic groups and different aromatic ring-positions of nitrovinyl group and alkoxy groups were designed and synthesized. We found that some analogues including compound 30 show stronger ability than Z-10 in inhibiting TNFα survival signal in MDA-MB-231 breast cancer cells. Interestingly, these RXRα ligands also bind to tubulins likely through the similar covalent interaction and induce the degradation of tubulins and cell cycle arrest in MDA-MB-231 cells, of which 30 displays the strongest efficacy. Importantly, these analogues and TNFα exhibit synergistic effects in inducing breast cancer cell apoptosis, of which 30 shows greater efficacy than Z-10. Together, our study provides a theoretical basis for the RXRα and tubulin dual-targeting drug design for breast cancer treatment.
Collapse
Affiliation(s)
- Xiaofang Qu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yanxia Wang
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunqing Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lin Xu
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiaohong Ye
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hongchen Cai
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China
| | - Liang Bu
- Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
| | - Zhiping Zeng
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen, Fujian, 361102, China; High Throughput Drug Screening Platform, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
3
|
Huang X, Zhou J, Li Z, Ye M, Hou C, Zhang Q, Chen Y, Li Q, Li F, Zhu X, Jiang J. EM-12, a natural sesquiterpene lactone extracted from Elephantopus mollis, promotes cancer cell apoptosis by activating ER stress. Med Oncol 2025; 42:115. [PMID: 40100452 DOI: 10.1007/s12032-025-02654-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
The Elephantopus mollis H.B.K. contains various sesquiterpene lactones that have shown anti-proliferative and proapoptotic effects in various cancers, although the underlying mechanisms are partially understood. Inducing of excessive ER stress is a potential cancer therapeutic strategy. However, ER stress activator remain limited in current clinical applications. In this study, we identified that EM-12, an uncovered sesquiterpene lactone isolated from Elephantopus mollis H.B.K., as a BiP ATPase activity inhibitor through BiP ATPase activity assay in vitro. This molecule also exhibits significantly greater cytotoxicity in numerous ovarian cancer cell lines, including paclitaxel-resistance ovarian cancer cell line, compared to transformed ovarian epithelial cell lines. In addition, EM-12 exerts broad-spectrum cytotoxicity against various human cancer cell lines, including liver, nasopharyngeal, and breast cancer cell lines. Mechanically, EM-12 promotes ER stress and ER-stress-related apoptosis to against cancer cells through inhibiting BiP ATPase activity.
Collapse
Affiliation(s)
- Xiang Huang
- Medical College of Jiaying University, Meizhou, 514031, China
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Junzhen Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zhenhua Li
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, China
| | - Meijun Ye
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Changyan Hou
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, China
| | - Qing Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yuanhong Chen
- Department of Gynecology, Dongguan Eastern Central Hospital, The Sixth Affiliated Hospital of Jinan University, Dongguan, 523560, China
| | - Qiang Li
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Fengying Li
- Department of Gynecology, The Affiliated Shunde Hospital of Jinan University, Foshan, 528000, China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Jianwei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
4
|
Wang W, Jin Y, Liu MK, Zhang SY, Chen H, Song J. Current Progress of Hederagenin and Its Derivatives for Disease Therapy (2017-Present). Molecules 2025; 30:1275. [PMID: 40142049 PMCID: PMC11944430 DOI: 10.3390/molecules30061275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Natural products have emerged as crucial sources of biologically active compounds, holding promise for applications in drug development. Among the extensively researched pentacyclic triterpenes, hederagenin (HG) stands out for its diverse biological activities and serves as a valuable scaffold for synthesizing novel derivatives. These derivatives hold significant promise for the development of novel therapeutic agents aimed at treating a wide range of diseases. Over the past years, a multitude of HG derivatives with varied bioactivities have been synthesized through chemical modifications. This review article consolidates the most recent findings (since 2017) on HG derivatives, emphasizing their biological effects and mechanisms of action in both in vitro and in vivo models. The objective of this compilation is to offer insights and direct future research endeavors in the realm of HG.
Collapse
Affiliation(s)
- Wang Wang
- Luoyang Key Laboratory of Organic Functional Molecules, School of Food and Drug, Luoyang Normal University, Luoyang 471934, China; (W.W.); (Y.J.); (M.-K.L.)
| | - Yan Jin
- Luoyang Key Laboratory of Organic Functional Molecules, School of Food and Drug, Luoyang Normal University, Luoyang 471934, China; (W.W.); (Y.J.); (M.-K.L.)
| | - Meng-Ke Liu
- Luoyang Key Laboratory of Organic Functional Molecules, School of Food and Drug, Luoyang Normal University, Luoyang 471934, China; (W.W.); (Y.J.); (M.-K.L.)
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China;
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| | - Hong Chen
- Luoyang Key Laboratory of Organic Functional Molecules, School of Food and Drug, Luoyang Normal University, Luoyang 471934, China; (W.W.); (Y.J.); (M.-K.L.)
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China;
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou 450001, China
| |
Collapse
|
5
|
Sun G, Wang Z, Li Y, Wang J, Liu F, Yu J, Yuan M, Wang N, Liu Z, Xiang C, Zhang Y, Oumata N, Yu P, Teng Y. Design and synthesis of isatin derivative payloaded peptide-drug conjugate as tubulin inhibitor against colorectal cancer. Eur J Med Chem 2025; 285:117276. [PMID: 39818012 DOI: 10.1016/j.ejmech.2025.117276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/18/2025]
Abstract
A series of isatin derivatives which could inhibit colorectal cancer (CRC) were synthesized. Among those compounds, 5B exhibited good inhibitory activity of CRC through the inhibition of tubulin expression, inducing apoptosis, and causing G2/M phase cell cycle arrest pathway, which suggested that 5B could be a potential tubulin inhibitor. Based on that, a novel peptide-drug conjugate (PDC), which employed the CRC cells related receptor CD44 ligand peptide A6 coupling to 5B to accomplish A6-5B. The in vitro and in vivo studies showed that A6-5B could significantly inhibit the tumor growth and metastasis in CRC cells. Mechanistic studies revealed that both 5B and A6-5B exert their antitumor effects by inhibiting tubulin, demonstrating that 5B might play a payload role and A6 could act as a targeting moiety for selective drug delivery to tumor cells.
Collapse
Affiliation(s)
- Guoyang Sun
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Zhaoyang Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yanping Li
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jinjin Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Futao Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Jiajia Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Mengzhen Yuan
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Ning Wang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Zhen Liu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Cen Xiang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China
| | - Yongmin Zhang
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China; Sorbonne Université, Institut Parisien de Chimie Moléculaire, UMR8232 CNRS, 4 Place Jussieu, 75005, Paris, France
| | - Nassima Oumata
- Université Paris Cité, 4, avenue de l'Observatoire, 75006, Paris, France
| | - Peng Yu
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China.
| | - Yuou Teng
- China International Science and Technology Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science and Technology, Tianjin, 300457, China.
| |
Collapse
|
6
|
Astiawati T, Rohman MS, Wihastuti T, Sujuti H, Endharti A, Sargowo D, Oceandy D, Lestari B, Triastuti E, Nugraha RA. The Emerging Role of Colchicine to Inhibit NOD-like Receptor Family, Pyrin Domain Containing 3 Inflammasome and Interleukin-1β Expression in In Vitro Models. Biomolecules 2025; 15:367. [PMID: 40149903 PMCID: PMC11940210 DOI: 10.3390/biom15030367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/22/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
While the beneficial effects of colchicine on inflammation and infarcted myocardium have been documented, its impact on cardiac fibroblast activation in the context of myocardial infarction (MI) remains unknown. This study aimed to investigate the effect of colchicine on the regulation of NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation and Interleukin-1β (IL-1β) expression in fibroblasts. 3T3 fibroblasts were exposed to 600 μM CoCl2 for 24 h to simulate hypoxia, with normoxic cells as controls. Colchicine (1 μM) was administered for 24 h. ASC-NLRP3 colocalization and IL-1β expression were evaluated using immunofluorescence and flow cytometry, respectively. Data were analyzed using t-tests and one-way ANOVA with post hoc tests. Hypoxia treatment significantly induced apoptosis-associated speck-like protein containing a CARD (ASC)-NLRP3 colocalization (p < 0.05). Colchicine treatment of hypoxic 3T3 cells reduced ASC-NLRP3 colocalization, although this reduction was not statistically significant. Additionally, IL-1β expression was significantly inhibited in colchicine-treated hypoxic 3T3 cells compared to those treated with placebo (p < 0.05). The findings of this study indicate that colchicine treatment inhibits the activation of the NLRP3 inflammasome by disrupting the colocalization of ASC and NLRP3, thereby reducing IL-1β expression in CoCl2-treated 3T3 cells.
Collapse
Affiliation(s)
- Tri Astiawati
- Doctoral Program of Medical Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
- Department of Cardiology, Doctor Iskak General Hospital, Tulungagung 62233, Indonesia
| | - Mohammad Saifur Rohman
- Department of Cardiology and Cardiovascular Medicine, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
- Cardiovascular Research Centre, Brawijaya University, Malang 65145, Indonesia
| | - Titin Wihastuti
- Department of Biomedical, Nursing Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Hidayat Sujuti
- Department of Biochemistry, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Agustina Endharti
- Doctoral Program of Medical Science, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Delvac Oceandy
- Division of Cardiovascular Science, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Bayu Lestari
- Department of Pharmacology, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PT, UK;
| | - Efta Triastuti
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang 65145, Indonesia;
| | - Ricardo Adrian Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Universitas Airlangga—Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia;
| |
Collapse
|
7
|
Rubicondo M, Ciardelli G, Mattu C, Tuszynski JA. Recent advancements in colchicine derivatives: Exploring synthesis, activities, and nanoformulations for enhanced therapeutic efficacy. Drug Discov Today 2025; 30:104312. [PMID: 39947582 DOI: 10.1016/j.drudis.2025.104312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
The multifaceted anti-cancer properties of colchicine make it a promising candidate for tumor treatment. However, its application has been limited by poor solubility, low bioavailability, and systemic toxicity. Considerable efforts have been directed toward the development of colchicine derivatives and nanoformulations to overcome these challenges. In this review, we provide a comprehensive overview of recent advances in colchicine derivatives and nanoformulations for cancer treatment. Synthesis methods and in vitro antiproliferative assays for the reviewed derivatives and formulations are explored. Challenges, such as drug resistance and formulation optimization, are also addressed, along with future perspectives for leveraging the full potential of colchicine derivatives and their nanoformulations as innovative anti-cancer strategies, toward successful clinical applications.
Collapse
Affiliation(s)
- Marialucia Rubicondo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Polito BIOMed Lab, Politecnico di Torino, Turin, Italy
| | - Jack A Tuszynski
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy; Department of Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada.
| |
Collapse
|
8
|
Lai Q, Wang Z, Wu C, Zhang R, Li L, Tao Y, Mo D, Zhang J, Gou L, Wang Y. Design, synthesis, and antitumor evaluation of quinazoline-4-tetrahydroquinoline chemotypes as novel tubulin polymerization inhibitors targeting the colchicine site. Eur J Med Chem 2025; 283:117139. [PMID: 39662284 DOI: 10.1016/j.ejmech.2024.117139] [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: 10/16/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024]
Abstract
We designed, synthesized, and evaluated the antitumor activity of a series of novel quinazoline-4-(6-methoxytetrahydroquinoline) analogues. Among the tested compounds, 4a4 exhibited the most potent antiproliferative activities across four human cancer cell lines with half-maximal inhibitory concentration (IC50) values ranging from 0.4 to 2.7 nM, more potent than the lead compound. The 2.71 Å resolution co-crystal structure of 4a4 with tubulin (PDB code: 8YER) confirmed its critical binding at the colchicine site. Moreover, 4a4 inhibited the polymerization of tubulin, colony formation, and tumor cell migration, while inducing G2/M phase arrest and apoptosis. In vivo, 4a4 significantly delayed primary tumor growth in the SKOV3 xenograft model without obvious side effect. Our research enhances the structure-activity relationships (SARs) understanding of the quinazoline-4-tetrahydroquinoline scaffold and provides new insights for potential structural optimization and the development of novel colchicine binding site inhibitors (CBSIs).
Collapse
Affiliation(s)
- Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhijia Wang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruofei Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Leyan Li
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China; Course of Biological Sciences, Department of Life Science, Imperial College London, United Kingdom
| | - Yiran Tao
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Mo
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
9
|
Zhao YC, Yan LQ, Xu Y. Recent advances of selenized tubulin inhibitors in cancer therapy. Bioorg Med Chem Lett 2025; 116:130037. [PMID: 39581555 DOI: 10.1016/j.bmcl.2024.130037] [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: 08/24/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024]
Abstract
Cancer treatment always a huge challenge amidst the resistance and relapse caused by the various treatments. Inhibitors targeting mitosis have been considered as promising therapeutic drugs in clinic, of which tubulins play an important role. Selenium (Se) as an essential microelement in humans and animals, playing a crucial role in the formation of anti-oxidase (glutathione peroxidase) and selenoprotein, also attracted broad attention in cancer therapy. Because the introduction of Se atom could change the length and angle of chemical bond and alter their functional properties, regulating selenized chemotherapeutics has become one of the hot spots. However, little attention has been paid to studying the combination of Se and tubulin inhibitors. Herein, we review the latest research results of selenized tubulin inhibitors in cancer therapy, including its mechanisms, categories and biological activities, providing a theoretical basis for different selenized microtubules inhibitors therapies.
Collapse
Affiliation(s)
- Yong-Chang Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China
| | - Liang-Qing Yan
- Department of Radiology, The People's Hospital of Yuhuan, Taizhou 317600, China
| | - Yuan Xu
- Department of Pharmacy, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu 322000, China.
| |
Collapse
|
10
|
Dwivedi AR, Kumar V, Prashar V, Jangid K, Kumar N, Devi B, Parkash J, Kumar V. Synthesis and screening of novel 2,4-bis substituted quinazolines as tubulin polymerization promoters and antiproliferative agents. RSC Med Chem 2025:d4md00755g. [PMID: 39897391 PMCID: PMC11781317 DOI: 10.1039/d4md00755g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/07/2025] [Indexed: 02/04/2025] Open
Abstract
Twelve 2,4-bis-substituted quinazoline-based compounds were synthesized and screened for antiproliferative and tubulin polymerization enhancing potential. In the series, compound A4V-3 substituted with an imidazole ring displayed IC50 values of 4.25 μM, 2.65 μM, and 9.95 μM, and A4V-5 with a benzotriazole substitution displayed IC50 values of 3.45 μM, 7.25 μM, and 8.14 μM against MCF-7, HCT-116 and SHSY-5Y cancer cells, respectively. In the mechanistic studies involving cell cycle analysis, apoptosis assay and JC-1 studies, compound A4V-3 was found to arrest the cells in the G2/M phase of the cell cycle and induce mitochondria-mediated apoptosis. In addition, compound A4V-3 displayed significant tubulin polymerization-enhancing potential. 2,4-Bis-substituted quinazoline-based compounds showed appreciable drug-like characteristics and can be developed as potent anticancer agents.
Collapse
Affiliation(s)
- Ashish Ranjan Dwivedi
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Bathinda Punjab 151401 India
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab Bathinda Punjab 151401 India +911642864269
- Gitam School of Pharmacy Hyderabad Telangana 502329 India
| | - Vijay Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab Bathinda Punjab 151401 India +911642864269
| | - Vikash Prashar
- Department of Zoology, School of Biological Sciences, Central University of Punjab Bathinda Punjab 151401 India
| | - Kailash Jangid
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab Bathinda Punjab 151401 India
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab Bathinda Punjab 151401 India +911642864269
| | - Naveen Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab Bathinda Punjab 151401 India +911642864269
| | - Bharti Devi
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab Bathinda Punjab 151401 India +911642864269
| | - Jyoti Parkash
- Department of Zoology, School of Biological Sciences, Central University of Punjab Bathinda Punjab 151401 India
| | - Vinod Kumar
- Laboratory of Organic and Medicinal Chemistry, Department of Chemistry, Central University of Punjab Bathinda Punjab 151401 India +911642864269
| |
Collapse
|
11
|
Zheng M, Liu G, Han Y, Qian P, Wu M, Xiang M, Zhou Y. Synthesis, biological evaluation and mechanism study of a novel indole-pyridine chalcone derivative as antiproliferative agent against tumor cells through dual targeting tubulin and HK2. Eur J Med Chem 2025; 282:117058. [PMID: 39571460 DOI: 10.1016/j.ejmech.2024.117058] [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: 08/28/2024] [Revised: 11/03/2024] [Accepted: 11/12/2024] [Indexed: 12/10/2024]
Abstract
Chalcones have the characteristics of simple structure, easy synthesis and potent anti-tumor activity. Herein, a small library of fifty-five novel indole-chalcone derivatives were rationally designed and facilely synthesized. Consequently, their antiproliferative activity was systematically evaluated. Among which, compound 26 exhibited the most potent antiproliferative activity, with IC50 value of 0.764 μM against MD-MBA-231 cells. Moreover, it displayed a 5-fold selectivity compared with normal human cells. Further investigation revealed that compound 26 bound at the colchicine binding site of tubulin, disrupted their fibrous structure, thereby blocking the progression of the cell cycle and inducing apoptosis. Molecular docking and cellular thermal shift assay (CETSA) experiments further demonstrated that compound 26 could specifically bind to hexokinase 2 (HK2) and inhibit its activity, leading to impaired mitochondrial function and hindered mitochondrial respiration. Based on the quantitative structure-activity relationship study, further structure modifications were performed. Employing biotin probe pull-down assays, we demonstrated that compound 26 exerted its antiproliferative activity through a dual targeting mechanism, which simultaneously disrupted microtubule function and inhibited HK2 activity. Taken together, these results highlighted that compound 26 might be a promising antiproliferative agent for human cancer therapy.
Collapse
Affiliation(s)
- Mengzhu Zheng
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Guangyuan Liu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Yawei Han
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Pengyu Qian
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Mingze Wu
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China
| | - Ming Xiang
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China.
| | - Yirong Zhou
- Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, China.
| |
Collapse
|
12
|
Adekunle YA, Samuel BB, Ezeude CM, Nahar L, Fatokun AA, Sarker SD. Isolation, cytotoxicity evaluation, and molecular docking of 3,4,3'-tri- O-methylflavellagic acid from Anogeissus leiocarpus (DC.) Guill. & Perr. root. Nat Prod Res 2025:1-8. [PMID: 39798146 DOI: 10.1080/14786419.2025.2451218] [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: 05/24/2024] [Revised: 12/11/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
Cancer kills about 10 million people every year. Medicinal plants remain a major source in the global search for anticancer drugs. In this study, 3,4,3'-tri-O-methylflavellagic acid (MFA) was isolated from the methanol root extract of Anogeissus leiocarpus. The structure was determined by 1D- and 2D-NMR data. The cytotoxic effects of MFA were evaluated against human breast (MCF-7), colorectal (Caco-2), and cervical (HeLa) cancer cell lines using the 3-[4,5-dimethylthiazole-2-yl] 3,5-diphenyltetrazolium bromide assay. A multi-protein target screening via molecular docking was conducted against ten cancer-related proteins, and ADMET properties were evaluated. MFA exhibited the most potent activity against Caco-2 (IC50: 46.75 ± 13.00 µM). Molecular docking analysis showed that MFA had a strong binding affinity for the colchicine-binding site of αβ-tubulin and polo-like kinase-1 (binding energies: -8.5 and -8.4 kcal/mol, respectively). MFA also satisfied the Lipinski's Rule of Five. MFA could, therefore, potentially serve as a scaffold for developing new anticancer molecules.
Collapse
Affiliation(s)
- Yemi A Adekunle
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
- Department of Pharmaceutical Chemistry, Dora Akunyili College of Pharmacy, Igbinedion University, Benin City, Edo State, Nigeria
| | - Babatunde B Samuel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Chinemenma M Ezeude
- Department of Pharmaceutical Chemistry, Dora Akunyili College of Pharmacy, Igbinedion University, Benin City, Edo State, Nigeria
| | - Lutfun Nahar
- Laboratory of Growth Regulators, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czech Republic
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| | - Satyajit D Sarker
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
13
|
Gareev I, Jiang J, Beylerli O, Beilerli A, Ilyasova T, Shumadalova A, Bai Y, Du W, Yang B. Adjuvant Anti-tumor Therapy with Polyphenolic Compounds: A Review. Curr Med Chem 2025; 32:1934-1967. [PMID: 40351076 DOI: 10.2174/0109298673284605240301035057] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/25/2023] [Accepted: 01/03/2024] [Indexed: 05/14/2025]
Abstract
The search for effective methods of treatment and prevention of oncological diseases, despite the successes achieved in recent decades, remains one of the most urgent issues in modern medicine. It is known that chemotherapy and radiation therapy are based on the induction of cell death by increasing the intracellular concentration of reactive oxygen species (ROS). To increase the effectiveness of chemo- and radiotherapy, inducing and increasing oxidative stress in tumor cells has been proposed. A new class of promising adjuvants in combination with anticancer therapy, which has already been shown to be effective in preclinical and clinical studies, includes natural and synthetic polyphenols. Polyphenolic compounds not only exhibit antitumor activity but also significantly reduce the resistance of tumor cells to chemo- and radiotherapy. However, almost all chemotherapeutic drugs and regimens of radiation treatment have a damaging toxic effect on normal tissues, which significantly affects the quality of life of patients, and treatment options for managing these side effects are limited. In this regard, some of the most promising agents for the management of toxic side effects are natural polyphenols. This study discusses the possible molecular mechanisms and prospects for the clinical use of natural and synthetic polyphenolic compounds in chemo- and radiotherapy. In addition, the protective role/effect of polyphenols on the effects of chemoand radiotherapy in tumor patients is discussed.
Collapse
Affiliation(s)
- Ilgiz Gareev
- Central Research Laboratory, Department of Pharmacology, Bashkir State Medical University, Republic of Bashkortostan, 3 Lenin Street, Ufa, 450008, Russia
| | - Jianhao Jiang
- The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, P.R. China
| | - Ozal Beylerli
- Central Research Laboratory, Department of Pharmacology, Bashkir State Medical University, Republic of Bashkortostan, 3 Lenin Street, Ufa, 450008, Russia
| | - Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 3 Lenin Street, Ufa, 450008, Russia
| | - Alina Shumadalova
- Department of General Chemistry, Bashkir State Medical University, Republic of Bashkortostan, 3 Lenin Street, Ufa, 450008, Russia
| | - Yunlong Bai
- The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, P.R. China
| | - Weijie Du
- The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, P.R. China
| | - Baofeng Yang
- The State-Province Key Laboratories of Biomedicine Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, 150067, China
- Translational Medicine Research and Cooperation Center of Northern China, Heilongjiang Academy of Medical Sciences, Harbin, 150081, P.R. China
| |
Collapse
|
14
|
Orleanska J, Bik E, Baranska M, Majzner K. Mechanisms of mitotic inhibition in human aorta endothelial cells: Molecular and morphological in vitro spectroscopic studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 322:124623. [PMID: 39002470 DOI: 10.1016/j.saa.2024.124623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 07/15/2024]
Abstract
Mitotic inhibitors are drugs commonly used in chemotherapy, but their nonspecific and indiscriminate distribution throughout the body after intravenous administration can lead to serious side effects, particularly on the cardiovascular system. In this context, our investigation into the mechanism of the cytotoxic effects on endothelial cells of mitotic inhibitors widely used in cancer treatment, such as paclitaxel (also known as Taxol) and Vinca alkaloids, holds significant practical implications. Understanding these mechanisms can lead to more targeted and less harmful cancer treatments. Human aorta endothelial cells (HAECs) were incubated with selected mitotic inhibitors in a wide range of concentrations close to those in human plasma during anticancer therapy. The analysis of single cells imaged by Raman spectroscopy allowed for visualization of the nuclear, cytoplasmic, and perinuclear areas to assess biochemical changes induced by the drug's action. The results showed significant changes in the morphology and molecular composition of the nucleus. Moreover, an effect of a given drug on the cytoplasm was observed, which can be related to its mechanism of action (MoA). Raman data supported by fluorescence microscopy measurements identified unique changes in DNA form and proteins and revealed drug-induced inflammation of endothelial cells. The primary goal of mitotic inhibitors is based on the impairment of tubulin formation and the inhibition of the mitosis process. While all three drugs affect microtubules and disrupt cell division, they do so through different MoA, i.e., Vinca alkaloids inhibit microtubule formation, whereas paclitaxel stabilizes microtubules. To sum up, the work shows how a specific drug can interact with endothelial cells.
Collapse
Affiliation(s)
- Jagoda Orleanska
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland; Jagiellonian University, Doctoral School of Exact and Natural Sciences, Lojasiewicza 11, 30-348 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Ewelina Bik
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland; Academic Centre for Materials and Nanotechnology, AGH University of Krakow, Mickiewicza Av. 30, 30-059 Krakow, Poland
| | - Malgorzata Baranska
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348 Krakow, Poland
| | - Katarzyna Majzner
- Jagiellonian University, Faculty of Chemistry, Gronostajowa 2, 30-387 Krakow, Poland.
| |
Collapse
|
15
|
Komal, Nanda BP, Singh L, Bhatia R, Singh A. Paclitaxel in colon cancer management: from conventional chemotherapy to advanced nanocarrier delivery systems. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9449-9474. [PMID: 38990305 DOI: 10.1007/s00210-024-03256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 06/22/2024] [Indexed: 07/12/2024]
Abstract
Paclitaxel, a potent chemotherapeutic agent derived from the bark of the Pacific yew tree, has demonstrated significant efficacy in the treatment of various cancers, including colon cancer. This comprehensive review delves into the conventional treatments for colon cancer, emphasizing the crucial role of paclitaxel in contemporary management strategies. It explores the intricate process of sourcing and synthesizing paclitaxel, highlighting the importance of its structural properties in its anticancer activity. The review further elucidates the mechanism of action of paclitaxel, its pharmacological effects, and its integration into chemotherapy regimens for colon cancer. Additionally, novel drug delivery systems, such as nanocarriers, liposomes, nanoparticles, microspheres, micelles, microemulsions, and niosomes, are examined for their potential to enhance the therapeutic efficacy of paclitaxel. The discussion extends to recent clinical trials and patents, showcasing advancements in paclitaxel formulations aimed at improving treatment outcomes. The review concludes with prospects in the field underscoring the ongoing innovation and potential breakthroughs in colon cancer therapy.
Collapse
Affiliation(s)
- Komal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India
| | - Bibhu Prasad Nanda
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Lovekesh Singh
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Rohit Bhatia
- Department of Pharmaceutical Chemistry, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Amandeep Singh
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, Punjab, 142001, India.
| |
Collapse
|
16
|
Chowdhary S, Preeti, Shekhar, Gupta N, Kumar R, Kumar V. Advances in chalcone-based anticancer therapy: mechanisms, preclinical advances, and future perspectives. Expert Opin Drug Discov 2024; 19:1417-1437. [PMID: 39621431 DOI: 10.1080/17460441.2024.2436908] [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: 06/17/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
INTRODUCTION Cancer remains a leading cause of death worldwide with traditional treatments like chemotherapy, and radiotherapy becoming less effective due to multidrug resistance (MDR). This highlights the necessity for novel chemotherapeutics like chalcone-based compounds, which demonstrate broad anti-cancer properties and target multiple pathways. These compounds hold promise for improving cancer treatment outcomes compared to existing therapies. AREAS COVERED This review provides a comprehensive synopsis of the recent literature (2018-2024) for anti-proliferative/anti-cancer activity of chalcones. It includes the identification of potential targets, their mechanisms of action, and possible modes of binding. Additionally, chalcone derivatives in preclinical trials are also discussed. EXPERT OPINION Chalcones mark a significant stride in anticancer therapies due to their multifaceted approach in targeting various cellular pathways. Their ability to simultaneously target multiple pathways enables them to overcome drug resistance as compared to traditional therapies. With well-defined mechanisms of action, these compounds can serve as lead molecules for designing new, more promising treatments. Continued progress in synthesis and structural optimization, along with promising results from preclinical trials, offers hope for the development of more potent molecules, heralding a new era in cancer therapeutics.
Collapse
Affiliation(s)
| | - Preeti
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Shekhar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Nikita Gupta
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| | - Rajesh Kumar
- Department of Physics, Lovely Professional University, Phagwara, India
| | - Vipan Kumar
- Department of Chemistry, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
17
|
Nemati F, Ata Bahmani Asl A, Salehi P. Synthesis and modification of noscapine derivatives as promising future anticancer agents. Bioorg Chem 2024; 153:107831. [PMID: 39321713 DOI: 10.1016/j.bioorg.2024.107831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024]
Abstract
Noscapine, a tetrahydroisoquinoline alkaloid, was first isolated from Papaver somniferum and identified by Rabiquet in 1817. It has been used as an anti-tussive agent since the mid-1950 s. After the discovery of its anti-mitotic potential, it was into the limelight once again. Due to its low toxicity, high bioactivity and oral administration, It was regarded as a formidable framework for subsequent modification and advancement in the pursuit of innovative chemotherapeutic agents. Up to now, the rational derivatives of the noscapine have been designed and the biological activities of these analogues have been extensively investigated. This review provides a comprehensive examination of the chemical characteristics of noscapine and its semi-synthetic derivatives up to the present, encompassing a concise investigation into the biological properties of these compounds and additionally a discussion about biosynthesis and total synthesis of noscapine is also provided. In summary, our aim is to contribute to a more thorough comprehension of this structure. It can be asserted that a promising future lies ahead for noscapine and its engineered derivatives as noteworthy candidates for pharmaceutical drugs.
Collapse
Affiliation(s)
- Faezeh Nemati
- Department of Synthesis of Medicinal Organic Compounds, Institute of Medicinal Chemistry, Iranian Research Organization for Science and Technology (IROST), P.O. Box 33535111, Tehran, Iran
| | - Amir Ata Bahmani Asl
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin, 1983963113 Tehran, Iran
| | - Peyman Salehi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin, 1983963113 Tehran, Iran.
| |
Collapse
|
18
|
Lv X, Cheng WH, Li XX, Shang H, Zhang JY, Hong HY, Zheng YJ, Dong YQ, Gong JH, Zheng YB, Zou ZM. Dual inhibition of topoisomerase II and microtubule of podophyllotoxin derivative 5p overcomes cancer multidrug resistance. Eur J Pharmacol 2024; 983:176968. [PMID: 39233039 DOI: 10.1016/j.ejphar.2024.176968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
Compound 5p is a 4β-N-substituted podophyllotoxin derivative, which exhibited potent activity toward drug-resistant K562/A02 cells and decreased MDR-1 mRNA expression. Here, we further investigated its detail mechanism and tested its antitumor activity. 5p exerted catalytic inhibition of topoisomerase IIα, and didn't show the inhibitor of topoisomerase I. 5p exhibited the inhibitory effect on microtubule polymerization. 5p showed potent anti-proliferation against breast cancer, oral squamous carcinoma, and their drug-resistant cell lines, with resistance index of 0.61 and 0.86, respectively. 5p downregulated the expression levels of P-gp in KBV200 cells and BCRP in MCF7/ADR cells in dose-dependent manner. Moreover, 5p induced KB and KBV200 cells arrest at G2/M phase by up-regulating the expression of γ-H2AX, p-Histone H3 and cyclin B1. 5p induced apoptosis and pyroptosis by increased the expression levels of cleaved-PARP, cleaved-caspase3, N-GSDME as well as LDH release in KB and KBV200 cells. In addition, 5p efficiently impaired tumor growth in KB and KBV200 xenograft mice. Conclusively, this work elucidated the dual inhibitor of topoisomerase II and microtubule of 5p and its mechanism of overcoming the multidrug resistance, indicating that 5p exerts the antitumor potentiality.
Collapse
Affiliation(s)
- Xing Lv
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China
| | - Wei-Hua Cheng
- HTA Co., Ltd., CAEA Center of Excellence on Nuclear Technology Applications for Engineering and Industrialization of Radiopharmaceuticals, CNNC Engineering Research Center of Radiopharmaceuticals, 102413, Beijing, China
| | - Xiao-Xue Li
- The State Key Laboratory of Basis and New Drug Development of Natural and Nuclear Drugs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 100193, Beijing, China
| | - Hai Shang
- The State Key Laboratory of Basis and New Drug Development of Natural and Nuclear Drugs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 100193, Beijing, China
| | - Jun-Yi Zhang
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China
| | - Han-Yu Hong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China
| | - Yi-Jia Zheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China
| | - Yan-Qun Dong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China
| | - Jian-Hua Gong
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China.
| | - Yan-Bo Zheng
- Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Tiantan Xili, 100050, Beijing, China.
| | - Zhong-Mei Zou
- The State Key Laboratory of Basis and New Drug Development of Natural and Nuclear Drugs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, 100193, Beijing, China.
| |
Collapse
|
19
|
Zhao W, Shen R, Li HM, Zhong JJ, Tang YJ. Podophyllotoxin derivatives-tubulin complex reveals a potential binding site of tubulin polymerization inhibitors in α-tubulin adjacent to colchicine site. Int J Biol Macromol 2024; 276:133678. [PMID: 38971286 DOI: 10.1016/j.ijbiomac.2024.133678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 06/12/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The colchicine site of β-tubulin has been proven to be essential binding sites of microtubule polymerization inhibitors. Recent studies implied that GTP pocket of α-tubulin adjacent to colchicine sites is a potential binding site for developing tubulin polymerization inhibitors. However, the structural basis for which type of structural fragments was more beneficial for enhancing the affinity of α-tubulin is still unclear. Here, podophyllotoxin derivatives-tubulin complex crystals indicated that heterocyclic with the highly electronegative and small steric hindrance was conducive to change configuration and enhance the affinity of the residues in GTP pocket of α-tubulin. Triazole with lone-pairs electrons and small steric hindrance exhibited the strongest affinity for enhancing affinity of podophyllotoxin derivatives by forming two hydrogen bonds with αT5 Ser178. Pyrimidine with the secondary strong affinity could bind Asn101 to make the αH7 configuration deflection, which reduces the stability of tubulin result in its depolymerization. Conversely, 4β-quinoline-podophyllotoxin with the weakest affinity did not interact with α-tubulin. The molecular dynamics simulation and protein thermal shift results showed that 4β-triazole-podophyllotoxin-tubulin was the most stable mainly due to two hydrogen bonds and the higher van der Waals force. This work provided a structural basis of the potential binding sites for extending the α/β-tubulin dual-binding sites inhibitors design strategy.
Collapse
Affiliation(s)
- Wei Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Rong Shen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Hong-Mei Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China.
| |
Collapse
|
20
|
Pospíšilová J, Heger T, Kurka O, Kvasnicová M, Chládková A, Nemec I, Rárová L, Cankař P. Atropisomeric 1-phenylbenzimidazoles affecting microtubule organization: influence of axial chirality. Org Biomol Chem 2024; 22:6966-6980. [PMID: 38988246 DOI: 10.1039/d4ob00863d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Benzimidazoles are frequently used in medicinal chemistry. Their anticancer effect is among the most prominent biological activities exhibited by this scaffold. Although numerous benzimidazole derivatives have been synthesized, possible atropisomerism of ortho-substituted 1-phenylbenzimidazoles has been largely overlooked. The aim of this research was to synthesize a small library of novel atropisomeric benzimidazole derivatives and explore their biological activity in various cancer and normal human cell lines. The new unique structural motif provides an interesting 3D architecture with axial chirality, which further contributes to molecular complexity and specificity. Racemates and their separated atropisomers arrested the cell cycle, caused apoptosis, and affected microtubule organization in cancer cells in vitro at different intensities. Moreover, this phenomenon was also verified by the inhibition of endothelial cell migration. These results showed that (+)-atropisomers, especially 5n, exhibit a stronger effect and show promise as agents for cancer therapy.
Collapse
Affiliation(s)
- Jana Pospíšilová
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| | - Tomáš Heger
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
| | - Ondřej Kurka
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences and Faculty of Science, Palacký University, Slechtitelu 27, Olomouc CZ-77900, Czech Republic
| | - Marie Kvasnicová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Science, Palacký University, Slechtitelu 27, 77900 Olomouc, Czech Republic
| | - Anna Chládková
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| | - Ivan Nemec
- Department of Inorganic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic
| | - Lucie Rárová
- Department of Experimental Biology, Faculty of Science, Palacký University Olomouc, Slechtitelu 27, 77900 Olomouc, Czech Republic.
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Science, Palacký University, Slechtitelu 27, 77900 Olomouc, Czech Republic
| | - Petr Cankař
- Department of Organic Chemistry, Faculty of Science, Palacký University Olomouc, 17. listopadu 1192/12, 77900 Olomouc, Czech Republic.
| |
Collapse
|
21
|
Lin HY, Tsai TN, Hsu KC, Hsu YM, Chiang LC, El-Shazly M, Chang KM, Lin YH, Tu SY, Lin TE, Du YC, Liu YC, Lu MC. From Sea to Science: Coral Aquaculture for Sustainable Anticancer Drug Development. Mar Drugs 2024; 22:323. [PMID: 39057432 PMCID: PMC11277741 DOI: 10.3390/md22070323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Marine natural products offer immense potential for drug development, but the limited supply of marine organisms poses a significant challenge. Establishing aquaculture presents a sustainable solution for this challenge by facilitating the mass production of active ingredients while reducing our reliance on wild populations and harm to local environments. To fully utilize aquaculture as a source of biologically active products, a cell-free system was established to target molecular components with protein-modulating activity, including topoisomerase II, HDAC, and tubulin polymerization, using extracts from aquaculture corals. Subsequent in vitro studies were performed, including MTT assays, flow cytometry, confocal microscopy, and Western blotting, along with in vivo xenograft models, to verify the efficacy of the active extracts and further elucidate their cytotoxic mechanisms. Regulatory proteins were clarified using NGS and gene modification techniques. Molecular docking and SwissADME assays were performed to evaluate the drug-likeness and pharmacokinetic and medicinal chemistry-related properties of the small molecules. The extract from Lobophytum crassum (LCE) demonstrated potent broad-spectrum activity, exhibiting significant inhibition of tubulin polymerization, and showed low IC50 values against prostate cancer cells. Flow cytometry and Western blotting assays revealed that LCE induced apoptosis, as evidenced by the increased expression of apoptotic protein-cleaved caspase-3 and the populations of early and late apoptotic cells. In the xenograft tumor experiments, LCE significantly suppressed tumor growth and reduced the tumor volume (PC3: 43.9%; Du145: 49.2%) and weight (PC3: 48.8%; Du145: 7.8%). Additionally, LCE inhibited prostate cancer cell migration, and invasion upregulated the epithelial marker E-cadherin and suppressed EMT-related proteins. Furthermore, LCE effectively attenuated TGF-β-induced EMT in PC3 and Du145 cells. Bioactivity-guided fractionation and SwissADME validation confirmed that LCE's main component, 13-acetoxysarcocrassolide (13-AC), holds greater potential for the development of anticancer drugs.
Collapse
Affiliation(s)
- Hung-Yu Lin
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung 824, Taiwan
- Division of Urology, Department of Surgery, E-Da Cancer Hospital, I-Shou University, Kaohsiung 824, Taiwan
| | - Tsen-Ni Tsai
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
| | - Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Ming Hsu
- Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Lin-Chien Chiang
- Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Organization of African Unity Street, Abassia, Cairo 11566, Egypt
| | - Ken-Ming Chang
- Department of Pharmacy and Master Program, Tajen University, Pingtung 907, Taiwan
| | - Yu-Hsuan Lin
- Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
| | - Shang-Yi Tu
- Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Ying-Chi Du
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Yi-Chang Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Mei-Chin Lu
- Graduate Institute of Marine Biology, National Dong Hwa University, Pingtung 944, Taiwan
- National Museum of Marine Biology and Aquarium, Pingtung 944, Taiwan
| |
Collapse
|
22
|
Liu Z, Yang Z, Ablise M. Design and synthesis of novel imidazole-chalcone derivatives as microtubule protein polymerization inhibitors to treat cervical cancer and reverse cisplatin resistance. Bioorg Chem 2024; 147:107310. [PMID: 38583249 DOI: 10.1016/j.bioorg.2024.107310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/09/2024]
Abstract
Using the licochalcone moiety as a lead compound scaffold, 16 novel imidazole-chalcone derivatives were designed and synthesized as microtubule protein polymerization inhibitors. The proliferation inhibitory activities of the derivatives against SiHa (human cervical squamous cell carcinoma), C-33A (human cervical cancer), HeLa (human cervical cancer), HeLa/DDP (cisplatin-resistant human cervical cancer), and H8 (human cervical epithelial immortalized) cells were evaluated. Compound 5a exhibited significant anticancer activity with IC50 values ranging from 2.28 to 7.77 μM and a resistance index (RI) of 1.63, while showing minimal toxicity to normal H8 cells. When compound 5a was coadministered with cisplatin, the RI of cisplatin to HeLa/DDP cells decreased from 6.04 to 2.01, while compound 5a enhanced the fluorescence intensity of rhodamine 123 in HeLa/DDP cells. Further studies demonstrated that compound 5a arrested cells at the G2/M phase, induced apoptosis, reduced colony formation, inhibited cell migration, and inhibited cell invasion. Preliminary mechanistic studies revealed that compound 5a decreased the immunofluorescence intensity of α-/β-tubulin in cancer cells, reduced the expression of polymerized α-/β-tubulin, and increased the expression of depolymerized α-/β-tubulin. Additionally, the molecular docking results demonstrate that compound 5a can interact with the tubulin colchicine binding site and generate multiple types of interactions. These results suggested that compound 5a has anticancer effects and significantly reverses cervical cancer resistance to cisplatin, which may be related to its inhibition of microtubule and P-glycoprotein (P-gp) activity.
Collapse
Affiliation(s)
- Zhengye Liu
- The Xinjiang Key Laboratory of Natural Medicine Active Components and Drug Release Technology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Zheng Yang
- The Xinjiang Key Laboratory of Natural Medicine Active Components and Drug Release Technology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China
| | - Mourboul Ablise
- The Xinjiang Key Laboratory of Natural Medicine Active Components and Drug Release Technology, College of Pharmacy, Xinjiang Medical University, Urumqi 830011, China.
| |
Collapse
|
23
|
Gach-Janczak K, Drogosz-Stachowicz J, Janecka A, Wtorek K, Mirowski M. Historical Perspective and Current Trends in Anticancer Drug Development. Cancers (Basel) 2024; 16:1878. [PMID: 38791957 PMCID: PMC11120596 DOI: 10.3390/cancers16101878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Cancer is considered one of the leading causes of death in the 21st century. The intensive search for new anticancer drugs has been actively pursued by chemists and pharmacologists for decades, focusing either on the isolation of compounds with cytotoxic properties from plants or on screening thousands of synthetic molecules. Compounds that could potentially become candidates for new anticancer drugs must have the ability to inhibit proliferation and/or induce apoptosis in cancer cells without causing too much damage to normal cells. Some anticancer compounds were discovered by accident, others as a result of long-term research. In this review, we have presented a brief history of the development of the most important groups of anticancer drugs, pointing to the fact that they all have many side effects.
Collapse
Affiliation(s)
- Katarzyna Gach-Janczak
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.J.); (K.W.)
| | | | - Anna Janecka
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.J.); (K.W.)
| | - Karol Wtorek
- Department of Biomolecular Chemistry, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland; (K.G.-J.); (A.J.); (K.W.)
| | - Marek Mirowski
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
24
|
Feng Y, Zhang T, Liu H. circPDK1 competitively binds miR-4731-5p to mediate GIGYF1 expression and increase paclitaxel sensitivity in non-small cell lung cancer. Discov Oncol 2024; 15:157. [PMID: 38733530 PMCID: PMC11088590 DOI: 10.1007/s12672-024-01003-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
OBJECTIVE To investigate the action of circPDK1 in paclitaxel (PTX) resistance in non-small cell lung cancer (NSCLC). METHODS circPDK1, miR-4731-5p, and GIGYF1 levels were determined by RT-qPCR and Western blot. Cell proliferation was detected by CCK-8 and colony formation assay, apoptosis by flow cytometry, invasion by Transwell assay. The targeting relationship between miR-4731-5p and circPDK1 or GIGYF1 was confirmed by dual luciferase reporter gene and RIP assay. A xenograft tumor model was established to determine the role of circPDK1 in PTX resistance. RESULTS circPDK1 was overexpressed in PTX-resistant NSCLC, and depleting circPDK1 hampered proliferation and invasion of PTX-resistant cells, activated apoptosis, and improved PTX sensitivity. circPDK1 bound to miR-4731-5p, and increasing miR-4731-5p expression salvaged the effect of circPDK1 depletion on PTX resistance. miR-4731-5p directly targeted GIGYF1, and upregulating GIGYF1 offset the promoting effect of circPDK1 knockdown on PTX sensitivity. NSCLC tumor growth was inhibited and PTX sensitivity improved when circPDK1 was suppressed. CONCLUSION Depleting circPDK1 promotes PTX sensitivity of NSCLC cells via miR-4731-5p/GIGYF1 axis, thereby inhibiting NSCLC pregnancy.
Collapse
Affiliation(s)
- YunYin Feng
- Department of Respiratory, Kaihua County Traditional Chinese Medicine Hospital, No.10 Zhongshan Road, Qinyang Office, Quzhou City, 324000, Zhejiang Province, China.
| | - TaoLong Zhang
- Department of Gastroenterology, Kaihua County Traditional Chinese Medicine Hospital, Quzhou City, 324300, Zhejiang Province, China
| | - Hong Liu
- Department of Respiratory, Kaihua County Traditional Chinese Medicine Hospital, No.10 Zhongshan Road, Qinyang Office, Quzhou City, 324000, Zhejiang Province, China
| |
Collapse
|
25
|
Tjeerdema E, Lee Y, Metry R, Hamdoun A. Semi-automated, high-content imaging of drug transporter knockout sea urchin (Lytechinus pictus) embryos. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART B, MOLECULAR AND DEVELOPMENTAL EVOLUTION 2024; 342:313-329. [PMID: 38087422 PMCID: PMC12010930 DOI: 10.1002/jez.b.23231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 11/08/2023] [Accepted: 11/19/2023] [Indexed: 05/01/2024]
Abstract
A defining feature of sea urchins is their extreme fecundity. Urchins produce millions of transparent, synchronously developing embryos, ideal for spatial and temporal analysis of development. This biological feature has been effectively utilized for ensemble measurement of biochemical changes. However, it has been underutilized in imaging studies, where single embryo measurements are used. Here we present an example of how stable genetics and high content imaging, along with machine learning-based image analysis, can be used to exploit the fecundity and synchrony of sea urchins in imaging-based drug screens. Building upon our recently created sea urchin ABCB1 knockout line, we developed a high-throughput assay to probe the role of this drug transporter in embryos. We used high content imaging to compare accumulation and toxicity of canonical substrates and inhibitors of the transporter, including fluorescent molecules and antimitotic cancer drugs, in homozygous knockout and wildtype embryos. To measure responses from the resulting image data, we used a nested convolutional neural network, which rapidly classified embryos according to fluorescence or cell division. This approach identified sea urchin embryos with 99.8% accuracy and determined two-cell and aberrant embryos with 96.3% and 89.1% accuracy, respectively. The results revealed that ABCB1 knockout embryos accumulated the transporter substrate calcein 3.09 times faster than wildtypes. Similarly, knockouts were 4.71 and 3.07 times more sensitive to the mitotic poisons vinblastine and taxol. This study paves the way for large scale pharmacological screens in the sea urchin embryo.
Collapse
Affiliation(s)
- Evan Tjeerdema
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Yoon Lee
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Rachel Metry
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Amro Hamdoun
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
26
|
Ghani U, Syed SA, Aljunidel S, Khan AA, Nur-E-Alam M, AlNoshan A, Al-Rehaily AJ, AlObaid A, Bari A. Synthesis of Competitive and Noncompetitive Inhibitors of Alpha-Glucosidase and Anticancer Agents. Chem Biodivers 2024; 21:e202301399. [PMID: 38393939 DOI: 10.1002/cbdv.202301399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/05/2024] [Accepted: 02/21/2024] [Indexed: 02/25/2024]
Abstract
Imidazoles and phenylthiazoles are an important class of heterocycles that demonstrate a wide range of biological activities against various types of cancers, diabetes mellitus and pathogenic microorganisms. The heterocyclic structure having oxothiazolidine moiety is an important scaffold present in various drugs, with potential for enzyme inhibition. In an effort to discover new heterocyclic compounds, we synthesized 26 new 4,5-diphenyl-1H-imidazole, phenylthiazole, and oxothiazolidine heterocyclic analogues that demonstrated potent α-glucosidase inhibition and anticancer activities. Majority of the compounds noncompetitively inhibited α-glucosidase except for two that exhibited competitive inhibition of the enzyme. Docking results suggested that the noncompetitive inhibitors bind to an apparent allosteric site on the enzyme located in the vicinity of the active site. Additionally, the analogues also exhibited significant activity against various types of cancers including non-small lung cancer. Since tubulin protein plays an important role in the pathogenesis of non-small lung cancer, molecular docking with one of the target compounds provided important clues to its binding mode. The current work on imidazoles and phenylthiazole derivatives bears importance for designing of new antidiabetic and anticancer drugs.
Collapse
Affiliation(s)
- Usman Ghani
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh, 12372, Saudi Arabia
| | - Saeed Ali Syed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 12372, Saudi Arabia
| | - Sarah Aljunidel
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh, 12372, Saudi Arabia
| | | | - Mohammad Nur-E-Alam
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 12372, Saudi Arabia
| | - Abdulrahman AlNoshan
- Clinical Biochemistry Unit, Department of Pathology, College of Medicine, King Saud University, Riyadh, 12372, Saudi Arabia
| | - Adnan J Al-Rehaily
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 12372, Saudi Arabia
| | - Abdulrahman AlObaid
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 12372, Saudi Arabia
| | - Ahmed Bari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 12372, Saudi Arabia
| |
Collapse
|
27
|
Bózsity N, Nagy V, Szabó J, Pálházi B, Kele Z, Resch V, Paragi G, Zupkó I, Minorics R, Mernyák E. Synthesis of Estrone Heterodimers and Evaluation of Their In Vitro Antiproliferative Activity. Int J Mol Sci 2024; 25:4274. [PMID: 38673860 PMCID: PMC11050183 DOI: 10.3390/ijms25084274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 04/04/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Directed structural modifications of natural products offer excellent opportunities to develop selectively acting drug candidates. Natural product hybrids represent a particular compound group. The components of hybrids constructed from different molecular entities may result in synergic action with diminished side effects. Steroidal homo- or heterodimers deserve special attention owing to their potentially high anticancer effect. Inspired by our recently described antiproliferative core-modified estrone derivatives, here, we combined them into heterodimers via Cu(I)-catalyzed azide-alkyne cycloaddition reactions. The two trans-16-azido-3-(O-benzyl)-17-hydroxy-13α-estrone derivatives were reacted with 3-O-propargyl-D-secoestrone alcohol or oxime. The antiproliferative activities of the four newly synthesized dimers were evaluated against a panel of human adherent gynecological cancer cell lines (cervical: Hela, SiHa, C33A; breast: MCF-7, T47D, MDA-MB-231, MDA-MB-361; ovarian: A2780). One heterodimer (12) exerted substantial antiproliferative activity against all investigated cell lines in the submicromolar or low micromolar range. A pronounced proapoptotic effect was observed by fluorescent double staining and flow cytometry on three cervical cell lines. Additionally, cell cycle blockade in the G2/M phase was detected, which might be a consequence of the effect of the dimer on tubulin polymerization. Computational calculations on the taxoid binding site of tubulin revealed potential binding of both steroidal building blocks, mainly with hydrophobic interactions and water bridges.
Collapse
Affiliation(s)
- Noémi Bózsity
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (N.B.); (V.N.)
| | - Viktória Nagy
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (N.B.); (V.N.)
| | - Johanna Szabó
- Department of Analytical and Molecular Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary; (J.S.); (B.P.)
| | - Balázs Pálházi
- Department of Analytical and Molecular Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary; (J.S.); (B.P.)
| | - Zoltán Kele
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary; (Z.K.); (V.R.); (G.P.)
| | - Vivien Resch
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary; (Z.K.); (V.R.); (G.P.)
| | - Gábor Paragi
- Department of Medicinal Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary; (Z.K.); (V.R.); (G.P.)
- Institute of Physics, University of Pécs, Ifjúság útja 6, H-7625 Pécs, Hungary
- Department of Theoretical Physics, University of Szeged, Tisza Lajos krt. 84-86, H-6720 Szeged, Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (N.B.); (V.N.)
| | - Renáta Minorics
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary; (N.B.); (V.N.)
| | - Erzsébet Mernyák
- Department of Analytical and Molecular Chemistry, University of Szeged, Dóm tér 8, H-6720 Szeged, Hungary; (J.S.); (B.P.)
- Institute of Pharmacognosy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| |
Collapse
|
28
|
Huang M, Han H, Liu H, Liu R, Li J, Li M, Guan Q, Zhang W, Wang D. Structure-based approaches for the design of 6-aryl-1-(3,4,5-trimethoxyphenyl)-1H-benzo[d][1,2,3]triazoles as tubulin polymerization inhibitors. Eur J Med Chem 2024; 269:116309. [PMID: 38471357 DOI: 10.1016/j.ejmech.2024.116309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
The colchicine binding site on tubulin has been widely acknowledged as an attractive target for anticancer drug exploitation. Here, we reported the structural optimization of the lead compound 4, which was proved in our previous work as a colchicine binding site inhibitor (CBSI). Based on docking researches for the active binding conformation of compound 4, a series of novel 6-aryl-1-(3,4,5-trimethoxyphenyl)-1H-benzo[d][1,2,3]triazole derivatives (9a-9x) were developed by replacing a CH group in the 1H-benzo[d]imidazole skeleton of compound 4 with a nitrogen atom as a hydrogen bond acceptor. Among them, compound 9a showed the strongest antiproliferative activity with IC50 values ranging from 14 to 45 nM against three human cancer cell lines (MCF-7, SGC-7901 and A549), lower than that of compound 4. Mechanistic studies indicated that compound 9a could inhibit tubulin polymerization, destroy the microtubule skeleton, block the cell cycle in G2/M phase, induce cancer cell apoptosis, prevent cancer cell migration and colony formation. Moreover, compound 9a significantly inhibited tumor growth in vivo without observable toxicity in the mice 4T1 xenograft tumor model. In conclusion, this report shows a successful case of the structure-based design approach of a potent tubulin polymerization inhibitor for cancer treatment.
Collapse
Affiliation(s)
- Mingxin Huang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Hongyao Han
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Haoyuan Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Runlai Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Jiwei Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mi Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qi Guan
- School of Pharmaceutical Engineering, 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.
| | - Dun Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
29
|
Han H, Yang M, Wen Z, Wang X, Lai X, Zhang Y, Fang R, Yin T, Yang X, Wang X, Zhao Q, Qi J, Chen H, Lin H, Yang Y. A modified natural small molecule inhibits triple-negative breast cancer growth by interacting with Tubb3. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 126:154894. [PMID: 38377719 DOI: 10.1016/j.phymed.2023.154894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/28/2023] [Accepted: 05/17/2023] [Indexed: 02/22/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a malignant tumor without specific therapeutic targets and a poor prognosis. Chemotherapy is currently the first-line therapeutic option for TNBC. However, due to the heterogeneity of TNBC, not all of TNBC patients are responsive to chemotherapeutic agents. Therefore, the demand for new targeted agents is critical. β-tubulin isotype III (Tubb3) is a prognostic factor associated with cancer progression, including breast cancer, and targeting Tubb3 may lead to improve TNBC disease control. Shikonin, the active compound in the roots of Lithospermun erythrorhizon suppresses the growth of various types of tumors, and its efficacy can be improved by altering its chemical structure. PURPOSE In this work, the anti-TNBC effect of a shikonin derivative (PMMB276) was investigated, and its mechanism was also investigated. STUDY DESIGN/METHODS This study combines flow cytometry, immunofluorescence staining, immunoblotting, immunoprecipitation, siRNA silencing, and the iTRAQ proteomics assay to analyze the inhibition potential of PMMB276 on TNBC. In vivo study was performed, Balb/c female murine models with or without the small molecule treatments. RESULTS Herein, we screened 300 in-house synthesized analogs of shikonin against TNBC and identified a novel small molecule, PMMB276; it suppressed cell proliferation, induced apoptosis, and arrested the cell cycle at the G2/M phase, suggesting that it could have a tumor suppressive role in TNBC. Tubb3 was identified as the target of PMMB276 using proteomic and biological activity analyses. Meanwhile, PMMB276 regulated microtubule dynamics in vitro by inducing microtubule depolymerization and it could act as a tubulin stabilizer by a different process than that of paclitaxel. Moreover, suppressing or inhibiting Tubb3 with PMMB276 reduced the growth of breast cancer in an experimental mouse model, indicating that Tubb3 plays a significant role in TNBC progression. CONCLUSION The findings support the therapeutic potential of PMMB276, a Tubb3 inhibitor, as a treatment for TNBC. Our findings might serve as a foundation for the utilization of shikonin and its derivatives in the development of anti-TNBC.
Collapse
Affiliation(s)
- Hongwei Han
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China
| | - Minkai Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China
| | - Zhongling Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China
| | - Xuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China
| | - Xiaohui Lai
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; School of Biology and Geography Science, Yili Normal University, Yining, 835000, China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Yahan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Rongjun Fang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Tongming Yin
- Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China
| | - Xiaorong Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; School of Biology and Geography Science, Yili Normal University, Yining, 835000, China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Xiaoming Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Quan Zhao
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China
| | - Hongyuan Chen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China
| | - Hongyan Lin
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China.
| | - Yonghua Yang
- State Key Laboratory of Pharmaceutical Biotechnology, Institute of Plant Molecular Biology, School of Life Sciences, Nanjing University, Nanjing, 210023, China; Co-Innovation Center for Sustainable Forestry in Southern China, MOE Key Laboratory of Forest Genetics and Biotechnology, Nanjing Forestry University, Nanjing, 210037, China; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, China.
| |
Collapse
|
30
|
Ezelarab HAA, Ali TFS, Abbas SH, Sayed AM, Beshr EAM, Hassan HA. New antiproliferative 3-substituted oxindoles inhibiting EGFR/VEGFR-2 and tubulin polymerization. Mol Divers 2024; 28:563-580. [PMID: 36790582 PMCID: PMC11070402 DOI: 10.1007/s11030-023-10603-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 01/06/2023] [Indexed: 02/16/2023]
Abstract
New 3-substituted oxindole derivatives were designed and synthesized as antiproliferative agents. The antiproliferative activity of compounds 6a-j was evaluated against 60 NCI cell lines. Among these tested compounds, compounds 6f and 6g showed remarkable antiproliferative activity, specifically against leukemia and breast cancer cell lines. Compound 6f was the most promising antiproliferative agent against MCF-7 (human breast cancer) with an IC50 value of 14.77 µM compared to 5-fluorouracil (5FU) (IC50 = 2.02 µM). Notably, compound 6f hampered receptor tyrosine EGFR fundamentally with an IC50 value of 1.38 µM, compared to the reference sunitinib with an IC50 value of 0.08 µM. Moreover, compound 6f afforded anti-tubulin polymerization activity with an IC50 value of 7.99 µM as an outstanding observable activity compared with the reference combretastatin A4 with an IC50 value of 2.64 µM. In silico molecular-docking results of compound 6f in the ATP-binding site of EGFR agreed with the in vitro results. Besides, the investigation of the physicochemical properties of compound 6f via the egg-boiled method clarified good lipophilicity, GIT absorption, and blood-brain barrier penetration properties.
Collapse
Affiliation(s)
- Hend A A Ezelarab
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519-Mini, Minia, Egypt
| | - Taha F S Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519-Mini, Minia, Egypt.
| | - Samar H Abbas
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519-Mini, Minia, Egypt.
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef, 62513, Egypt
| | - Eman A M Beshr
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519-Mini, Minia, Egypt.
| | - Heba A Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, 61519-Mini, Minia, Egypt
| |
Collapse
|
31
|
Xu Z, Zhai Y, Chang H, Yan D, Ge P, Ren G, Zhang L, Yuan Y, Wang R, Li W, Li F, Ren M, Mo H. Heterologous expression of taxane genes confers resistance to fall armyworm in Nicotiana benthamiana. PLANT CELL REPORTS 2024; 43:94. [PMID: 38472660 DOI: 10.1007/s00299-024-03169-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/05/2024] [Indexed: 03/14/2024]
Abstract
KEY MESSAGE Taxadiene synthase, taxadiene-5α-hydroxylase, and taxane 13α-hydroxylase genes were introduced into Nicotiana benthamiana, and the improved resistance to lepidoptera pest fall armyworm was reported. Fall armyworm (FAW) is a serious agricultural pest. Genetic engineering techniques have been used to create pest-resistant plant varieties for reducing pest damage. Paclitaxel is a diterpenoid natural metabolite with antineoplastic effects in medicine. However, the effects of taxanes on the growth and development of lepidoptera pests, such as the FAW, are unknown. Here, selected paclitaxel precursor biosynthesis pathway genes, taxadiene synthase, taxane 5α-hydroxylase, and taxane 13α-hydroxylase, were engineered in the heterologous host Nicotiana benthamiana plants. Bioassay experiments showed that the transgenic N. benthamiana plants displayed improved resistance to FAW infestation, with degeneration of gut tissues and induced expression of apoptosis-related genes. Cytotoxicity experiment showed that the paclitaxel precursor, 10-deacetylbaccatin III, is cytotoxic to Sf9 cells, causing cell cycle arrest at the G2/M phase and disorder of the cytoskeleton. Metabolome analysis showed that heterologous expression of taxane genes in N. benthamiana affected the digestive system, steroid hormone and purine metabolism pathways of FAW larvae. In summary, this study provides a candidate approach for FAW control.
Collapse
Affiliation(s)
- Zhenlu Xu
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yaohua Zhai
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Huimin Chang
- State Key Laboratory of Cotton Biology, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, China
| | - Da Yan
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Pengliang Ge
- College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, 430000, China
| | - Guangming Ren
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Lijun Zhang
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Ye Yuan
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Ruoyan Wang
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan Province, Zhengzhou, 450003, China
| | - Wentao Li
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Henan Province, Zhengzhou, 450003, China
| | - Fuguang Li
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- State Key Laboratory of Cotton Biology, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, China.
| | - Maozhi Ren
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- State Key Laboratory of Cotton Biology, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, China.
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, 610000, China.
| | - Huijuan Mo
- Zhengzhou Research Base, State Key Laboratory of Cotton Biology, School of Agricultural Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- State Key Laboratory of Cotton Biology, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, China.
| |
Collapse
|
32
|
Yang X. Research progress of LSD1-based dual-target agents for cancer therapy. Bioorg Med Chem 2024; 101:117651. [PMID: 38401457 DOI: 10.1016/j.bmc.2024.117651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 02/26/2024]
Abstract
Lysine-specific demethylase 1 (LSD1) is a histone lysine demethylase that is significantly overexpressed or dysregulated in different cancers and plays important roles in cell growth, invasion, migration, immune escape, angiogenesis, gene regulation, and transcription. Therefore, it is a superb target for the discovery of novel antitumor agents. However, because of their innate and acquired resistance and low selectivity, LSD1 inhibitors are associated with limited therapeutic efficacy and high toxicity. Furthermore, LSD1 inhibitors synergistically improve the efficacy of additional antitumor drugs, which encourages numerous medicinal chemists to innovate and develop new-generation LSD1-based dual-target agents. This review discusses the theoretical foundation of the design of LSD1-based dual-target agents and summarizes their possible applications in treating cancers.
Collapse
Affiliation(s)
- Xiaojuan Yang
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China.
| |
Collapse
|
33
|
Marinho LL, Rached FH, Morikawa AT, Tavoni TM, Cardoso APT, Torres RVA, Assuncao AN, Serrano CV, Nomura CH, Maranhão RC. Safety and possible anti-inflammatory effect of paclitaxel associated with LDL-like nanoparticles (LDE) in patients with chronic coronary artery disease: a double-blind, placebo-controlled pilot study. Front Cardiovasc Med 2024; 11:1342832. [PMID: 38450375 PMCID: PMC10915057 DOI: 10.3389/fcvm.2024.1342832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/13/2024] [Indexed: 03/08/2024] Open
Abstract
Introduction Studies in cholesterol-fed rabbits showed that anti-proliferative chemotherapeutic agents such as paclitaxel associated with solid lipid nanoparticles (LDE) have marked anti-atherosclerotic effects. In addition, association with LDE nearly abolishes paclitaxel toxicity. We investigated whether treatment with LDE-paclitaxel changes plaque progression by coronary CT angiography and is safe in patients with chronic coronary artery disease. Methods We conducted a prospective, randomized, double-blind, placebo-controlled pilot study in patients with multi-vessel chronic coronary artery disease. Patients were randomized to receive IV infusions of LDE-paclitaxel (paclitaxel dose: 175 mg/m2 body surface) or LDE alone (placebo group), administered every 3 weeks for 18 weeks. All participants received guideline-directed medical therapy. Clinical and laboratory safety evaluations were made at baseline and every 3 weeks until the end of the study. Analysis of inflammatory biomarkers and coronary CTA was also performed at baseline and 4 weeks after treatment. Results Forty patients aged 65.6 ± 8 years, 20 in LDE-paclitaxel and 20 in placebo group were enrolled. Among those, 58% had diabetes, 50% had myocardial infarction, and 91% were in use of statin and aspirin. Baseline demographics, risk factors, and laboratory results were not different between groups. In all patients, no clinical or laboratory toxicities were observed. From the baseline to the end of follow-up, there was a non-significant trend toward a decrease in IL-6 levels and hsCRP in the LDE-paclitaxel group (-16% and -28%, respectively), not observed in placebo. Regarding plaque progression analysis, variation in plaque parameter values was wide, and no difference between groups was observed. Conclusion In patients with multivessel chronic coronary artery disease and optimized medical therapy, LDE-paclitaxel was safe and showed clues of potential benefits in reducing inflammatory biomarkers. Clinical Trial Registration https://clinicaltrials.gov/study/NCT04148833, identifier (NCT04148833).
Collapse
Affiliation(s)
- Lucas Lage Marinho
- Lipid Metabolism Laboratory, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| | - Fabiana Hanna Rached
- Department of Cardiopneumology, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| | - Aleksandra Tiemi Morikawa
- Lipid Metabolism Laboratory, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| | - Thauany Martins Tavoni
- Lipid Metabolism Laboratory, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| | | | | | | | - Carlos Vicente Serrano
- Department of Cardiopneumology, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| | - Cesar Higa Nomura
- Department of Radiology, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| | - Raul Cavalcante Maranhão
- Lipid Metabolism Laboratory, Instituto do Coracao (InCor) Universidade de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Jiang B, Gao L, Wang H, Sun Y, Zhang X, Ke H, Liu S, Ma P, Liao Q, Wang Y, Wang H, Liu Y, Du R, Rogge T, Li W, Shang Y, Houk KN, Xiong X, Xie D, Huang S, Lei X, Yan J. Characterization and heterologous reconstitution of Taxus biosynthetic enzymes leading to baccatin III. Science 2024; 383:622-629. [PMID: 38271490 DOI: 10.1126/science.adj3484] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Paclitaxel is a well known anticancer compound. Its biosynthesis involves the formation of a highly functionalized diterpenoid core skeleton (baccatin III) and the subsequent assembly of a phenylisoserinoyl side chain. Despite intensive investigation for half a century, the complete biosynthetic pathway of baccatin III remains unknown. In this work, we identified a bifunctional cytochrome P450 enzyme [taxane oxetanase 1 (TOT1)] in Taxus mairei that catalyzes an oxidative rearrangement in paclitaxel oxetane formation, which represents a previously unknown enzyme mechanism for oxetane ring formation. We created a screening strategy based on the taxusin biosynthesis pathway and uncovered the enzyme responsible for the taxane oxidation of the C9 position (T9αH1). Finally, we artificially reconstituted a biosynthetic pathway for the production of baccatin III in tobacco.
Collapse
Affiliation(s)
- Bin Jiang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Lei Gao
- Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Haijun Wang
- Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Yaping Sun
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, College of Life Sciences, South China Agricultural University, Guangzhou, China
| | - Xiaolin Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Han Ke
- Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Shengchao Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Pengchen Ma
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Qinggang Liao
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yue Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Huan Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yugeng Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Ran Du
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Torben Rogge
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wei Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yi Shang
- Yunnan Key Laboratory of Potato Biology, The CAAS-YNNU-YINMORE Joint Academy of Potato Sciences, Yunnan Normal University, Kunming, China
| | - K N Houk
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xingyao Xiong
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Daoxin Xie
- Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Sanwen Huang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Peking-Tsinghua Center for Life Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jianbin Yan
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| |
Collapse
|
35
|
Moreira J, Silva PMA, Castro E, Saraiva L, Pinto M, Bousbaa H, Cidade H. BP-M345 as a Basis for the Discovery of New Diarylpentanoids with Promising Antimitotic Activity. Int J Mol Sci 2024; 25:1691. [PMID: 38338967 PMCID: PMC10855865 DOI: 10.3390/ijms25031691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Recently, the diarylpentanoid BP-M345 (5) has been identified as a potent in vitro growth inhibitor of cancer cells, with a GI50 value between 0.17 and 0.45 µM, showing low toxicity in non-tumor cells. BP-M345 (5) promotes mitotic arrest by interfering with mitotic spindle assembly, leading to apoptotic cell death. Following on from our previous work, we designed and synthesized a library of BP-M345 (5) analogs and evaluated the cell growth inhibitory activity of three human cancer cell lines within this library in order to perform structure-activity relationship (SAR) studies and to obtain compounds with improved antimitotic effects. Four compounds (7, 9, 13, and 16) were active, and the growth inhibition effects of compounds 7, 13, and 16 were associated with a pronounced arrest in mitosis. These compounds exhibited a similar or even higher mitotic index than BP-M345 (5), with compound 13 displaying the highest antimitotic activity, associated with the interference with mitotic spindle dynamics, inducing spindle collapse and, consequently, prolonged mitotic arrest, culminating in massive cancer cell death by apoptosis.
Collapse
Affiliation(s)
- Joana Moreira
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (J.M.); (M.P.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal
| | - Patrícia M. A. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; (P.M.A.S.); (E.C.)
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Eliseba Castro
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; (P.M.A.S.); (E.C.)
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratory of Microbiology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (J.M.); (M.P.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal
| | - Hassan Bousbaa
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; (P.M.A.S.); (E.C.)
| | - Honorina Cidade
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal; (J.M.); (M.P.)
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos S/N, 4450-208 Matosinhos, Portugal
| |
Collapse
|
36
|
Zheng L, Zou Y, Xie T, Wu X, Tan Y, Mei S, Geng Y, Chen S, Xu S, Niu MM. Discovery of a Dual Tubulin and Neuropilin-1 (NRP1) Inhibitor with Potent In Vivo Anti-Tumor Activity via Pharmacophore-based Docking Screening, Structure Optimization, and Biological Evaluation. J Med Chem 2023; 66:16187-16200. [PMID: 38093696 DOI: 10.1021/acs.jmedchem.3c01572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Dual inhibition of tubulin and neuropilin-1 (NRP1) may become an effective method for cancer treatment by simultaneously killing tumor cells and inhibiting tumor angiogenesis. Herein, we identified dual tubulin/NRP1-targeting inhibitor TN-2, which exhibited good inhibitory activity against both tubulin polymerization (IC50 = 0.71 ± 0.03 μM) and NRP1 (IC50 = 0.85 ± 0.04 μM). Importantly, it significantly inhibited the viability of several human prostate tumor cell lines. Further mechanism studies indicated that TN-2 could inhibit tubulin polymerization and cause G2/M arrest, thereby inducing cell apoptosis. It could also suppress cell tube formation, migration, and invasion. Moreover, TN-2 showed obvious antitumor effects on the PC-3 cell-derived xenograft model with negligible side effects and good pharmacokinetic profiles. These data demonstrate that TN-2 could be a promising dual-target chemotherapeutic agent for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Lufeng Zheng
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Yunting Zou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 211198, China
| | - Tianyuan Xie
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Xiuyuan Wu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yuchen Tan
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Shuang Mei
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 211198, China
| | - Yifei Geng
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 211198, China
| | - Shutong Chen
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 211198, China
| | - Shengtao Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Miao-Miao Niu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
37
|
Marunaka Y. Physiological roles of chloride ions in bodily and cellular functions. J Physiol Sci 2023; 73:31. [PMID: 37968609 PMCID: PMC10717538 DOI: 10.1186/s12576-023-00889-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/31/2023] [Indexed: 11/17/2023]
Abstract
Physiological roles of Cl-, a major anion in the body, are not well known compared with those of cations. This review article introduces: (1) roles of Cl- in bodily and cellular functions; (2) the range of cytosolic Cl- concentration ([Cl-]c); (3) whether [Cl-]c could change with cell volume change under an isosmotic condition; (4) whether [Cl-]c could change under conditions where multiple Cl- transporters and channels contribute to Cl- influx and efflux in an isosmotic state; (5) whether the change in [Cl-]c could be large enough to act as signals; (6) effects of Cl- on cytoskeletal tubulin polymerization through inhibition of GTPase activity and tubulin polymerization-dependent biological activity; (7) roles of cytosolic Cl- in cell proliferation; (8) Cl--regulatory mechanisms of ciliary motility; (9) roles of Cl- in sweet/umami taste receptors; (10) Cl--regulatory mechanisms of with-no-lysine kinase (WNK); (11) roles of Cl- in regulation of epithelial Na+ transport; (12) relationship between roles of Cl- and H+ in body functions.
Collapse
Affiliation(s)
- Yoshinori Marunaka
- Medical Research Institute, Kyoto Industrial Health Association, General Incorporated Foundation, 67 Kitatsuboi-Cho, Nishinokyo, Nakagyo-Ku, Kyoto, 604-8472, Japan.
- Research Organization of Science and Technology, Ritsumeikan University, Kusatsu, 525-8577, Japan.
- Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-Ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
38
|
Sedenkova KN, Leschukov DN, Grishin YK, Zefirov NA, Gracheva YA, Skvortsov DA, Hrytseniuk YS, Vasilyeva LA, Spirkova EA, Shevtsov PN, Shevtsova EF, Lukmanova AR, Spiridonov VV, Markova AA, Nguyen MT, Shtil AA, Zefirova ON, Yaroslavov AA, Milaeva ER, Averina EB. Verubulin (Azixa) Analogues with Increased Saturation: Synthesis, SAR and Encapsulation in Biocompatible Nanocontainers Based on Ca 2+ or Mg 2+ Cross-Linked Alginate. Pharmaceuticals (Basel) 2023; 16:1499. [PMID: 37895970 PMCID: PMC10610134 DOI: 10.3390/ph16101499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Tubulin-targeting agents attract undiminished attention as promising compounds for the design of anti-cancer drugs. Verubulin is a potent tubulin polymerization inhibitor, binding to colchicine-binding sites. In the present work, a series of verubulin analogues containing a cyclohexane or cycloheptane ring 1,2-annulated with pyrimidine moiety and various substituents in positions 2 and 4 of pyrimidine were obtained and their cytotoxicity towards cancer and non-cancerous cell lines was estimated. The investigated compounds revealed activity against various cancer cell lines with IC50 down to 1-4 nM. According to fluorescent microscopy data, compounds that showed cytotoxicity in the MTT test disrupt the normal cytoskeleton of the cell in a pattern similar to that for combretastatin A-4. The hit compound (N-(4-methoxyphenyl)-N,2-dimethyl-5,6,7,8-tetrahydroquinazolin-4-amine) was encapsulated in biocompatible nanocontainers based on Ca2+ or Mg2+ cross-linked alginate and it was demonstrated that its cytotoxic activity was preserved after encapsulation.
Collapse
Affiliation(s)
- Kseniya N. Sedenkova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Denis N. Leschukov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Yuri K. Grishin
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Nikolay A. Zefirov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Yulia A. Gracheva
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Dmitry A. Skvortsov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Yanislav S. Hrytseniuk
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Lilja A. Vasilyeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Elena A. Spirkova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), 142432 Chernogolovka, Russia; (E.A.S.); (P.N.S.); (E.F.S.)
| | - Pavel N. Shevtsov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), 142432 Chernogolovka, Russia; (E.A.S.); (P.N.S.); (E.F.S.)
| | - Elena F. Shevtsova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), 142432 Chernogolovka, Russia; (E.A.S.); (P.N.S.); (E.F.S.)
| | - Alina R. Lukmanova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Vasily V. Spiridonov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Alina A. Markova
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.M.); (M.T.N.)
| | - Minh T. Nguyen
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.M.); (M.T.N.)
| | - Alexander A. Shtil
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
- Institute of Cyber Intelligence Systems, National Research Nuclear University MEPhI, 115409 Moscow, Russia
| | - Olga N. Zefirova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Alexander A. Yaroslavov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Elena R. Milaeva
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Elena B. Averina
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| |
Collapse
|
39
|
Abstract
Targeted protein degradation (TPD) has emerged as the most promising approach for the specific knockdown of disease-associated proteins and is achieved by exploiting the cellular quality control machinery. TPD technologies are highly advantageous in overcoming drug resistance as they degrade the whole target protein. Microtubules play important roles in many cellular processes and are among the oldest and most well-established targets for tumor chemotherapy. However, the development of drug resistance, risk of hypersensitivity reactions, and intolerable toxicities severely restrict the clinical applications of microtubule-targeting agents (MTAs). Microtubule degradation agents (MDgAs) operate via completely different mechanisms compared with traditional MTAs and are capable of overcoming drug resistance. The emergence of MDgAs has expanded the scope of TPD and provided new avenues for the discovery of tubulin-targeted drugs. Herein, we summarized the development of MDgAs, and discussed their degradation mechanisms, mechanisms of action on the binding sites, potential opportunities, and challenges.
Collapse
Affiliation(s)
- Chufeng Zhang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-Induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yong Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
40
|
Bai Z, Zhou Y, Peng Y, Ye X, Ma L. Perspectives and mechanisms for targeting mitotic catastrophe in cancer treatment. Biochim Biophys Acta Rev Cancer 2023; 1878:188965. [PMID: 37625527 DOI: 10.1016/j.bbcan.2023.188965] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/14/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Mitotic catastrophe is distinct from other cell death modes due to unique nuclear alterations characterized as multi and/or micronucleation. Mitotic catastrophe is a common and virtually unavoidable consequence during cancer therapy. However, a comprehensive understanding of mitotic catastrophe remains lacking. Herein, we summarize the anticancer drugs that induce mitotic catastrophe, including microtubule-targeting agents, spindle assembly checkpoint kinase inhibitors, DNA damage agents and DNA damage response inhibitors. Based on the relationships between mitotic catastrophe and other cell death modes, we thoroughly evaluated the roles played by mitotic catastrophe in cancer treatment as well as its advantages and disadvantages. Some strategies for overcoming its shortcomings while fully utilizing its advantages are summarized and proposed in this review. We also review how mitotic catastrophe regulates cancer immunotherapy. These summarized findings suggest that the induction of mitotic catastrophe can serve as a promising new therapeutic approach for overcoming apoptosis resistance and strengthening cancer immunotherapy.
Collapse
Affiliation(s)
- Zhaoshi Bai
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, China.
| | - Yiran Zhou
- Department of General Surgery, Rui Jin Hospital, Research Institute of Pancreatic Diseases, School of Medicine, Shanghai JiaoTong University, Shanghai 200025, China
| | - Yaling Peng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Xinyue Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Lingman Ma
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| |
Collapse
|
41
|
Olivares-Ferretti P, Beltrán JF, Salazar LA, Fonseca-Salamanca F. Protein Modelling and Molecular Docking Analysis of Fasciola hepatica β-Tubulin's Interaction Sites, with Triclabendazole, Triclabendazole Sulphoxide and Triclabendazole Sulphone. Acta Parasitol 2023; 68:535-547. [PMID: 37330945 DOI: 10.1007/s11686-023-00692-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 05/29/2023] [Indexed: 06/20/2023]
Abstract
PURPOSE Fasciola hepatica is a globally distributed trematode that causes significant economic losses. Triclabendazole is the primary pharmacological treatment for this parasite. However, the increasing resistance to triclabendazole limits its efficacy. Previous pharmacodynamics studies suggested that triclabendazole acts by interacting mainly with the β monomer of tubulin. METHODS We used a high-quality method to model the six isotypes of F. hepatica β-tubulin in the absence of three-dimensional structures. Molecular dockings were conducted to evaluate the destabilization regions in the molecule against the ligands triclabendazole, triclabendazole sulphoxide and triclabendazole sulphone. RESULTS The nucleotide binding site demonstrates higher affinity than the binding sites of colchicine, albendazole, the T7 loop and pβVII (p < 0.05). We suggest that the binding of the ligands to the polymerization site of β-tubulin can lead a microtubule disruption. Furthermore, we found that triclabendazole sulphone exhibited significantly higher binding affinity than other ligands (p < 0.05) across all isotypes of β-tubulin. CONCLUSIONS Our investigation has yielded new insight on the mechanism of action of triclabendazole and its sulphometabolites on F. hepatica β-tubulin through computational tools. These findings have significant implications for ongoing scientific research ongoing towards the discovery of novel therapeutics to treat F. hepatica infections.
Collapse
Affiliation(s)
- Pamela Olivares-Ferretti
- Laboratory of Molecular Immunoparasitology, Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Temuco, Chile
| | - Jorge F Beltrán
- Chemical Engineering Department, Faculty of Engineering and Science, Universidad de La Frontera, Temuco, Chile
| | - Luis A Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Avenida Francisco Salazar 01145, 4811230, Temuco, Chile
| | - Flery Fonseca-Salamanca
- Laboratory of Molecular Immunoparasitology, Center of Excellence in Translational Medicine-Scientific and Technological Bioresource Nucleus (CEMT-BIOREN), Temuco, Chile.
- Preclinical Sciences Department, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile.
| |
Collapse
|
42
|
Zafar S, Armaghan M, Khan K, Hassan N, Sharifi-Rad J, Habtemariam S, Kieliszek M, Butnariu M, Bagiu IC, Bagiu RV, Cho WC. New insights into the anticancer therapeutic potential of maytansine and its derivatives. Biomed Pharmacother 2023; 165:115039. [PMID: 37364476 DOI: 10.1016/j.biopha.2023.115039] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/28/2023] Open
Abstract
Maytansine is a pharmacologically active 19-membered ansamacrolide derived from various medicinal plants and microorganisms. Among the most studied pharmacological activities of maytansine over the past few decades are anticancer and anti-bacterial effects. The anticancer mechanism of action is primarily mediated through interaction with the tubulin thereby inhibiting the assembly of microtubules. This ultimately leads to decreased stability of microtubule dynamics and cause cell cycle arrest, resulting in apoptosis. Despite its potent pharmacological effects, the therapeutic applications of maytansine in clinical medicine are quite limited due to its non-selective cytotoxicity. To overcome these limitations, several derivatives have been designed and developed mostly by modifying the parent structural skeleton of maytansine. These structural derivatives exhibit improved pharmacological activities as compared to maytansine. The present review provides a valuable insight into maytansine and its synthetic derivatives as anticancer agents.
Collapse
Affiliation(s)
- Sameen Zafar
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan
| | - Khushbukhat Khan
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Punjab, Pakistan.
| | - Nazia Hassan
- Department of Biochemistry, University of Agriculture Faisalabad, Pakistan
| | | | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, University of Greenwich, Central Avenue, Chatham-Maritime, Kent ME4 4TB, UK.
| | - Marek Kieliszek
- Department of Food Biotechnology and Microbiology, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159 C, 02-776 Warsaw, Poland.
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from Timisoara, 300645, Calea Aradului 119, Timis, Romania.
| | - Iulia-Cristina Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Multidisciplinary Research Center on Antimicrobial Resistance, Timisoara, Romania
| | - Radu Vasile Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania; Preventive Medicine Study Center, Timisoara, Romania
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong Special Administrative Region.
| |
Collapse
|
43
|
Gallego-Yerga L, Chiliquinga AJ, Peláez R. Novel Tetrazole Derivatives Targeting Tubulin Endowed with Antiproliferative Activity against Glioblastoma Cells. Int J Mol Sci 2023; 24:11093. [PMID: 37446273 DOI: 10.3390/ijms241311093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Increasing awareness of the structure of microtubules has made tubulin a relevant target for the research of novel chemotherapies. Furthermore, the particularly high sensitivity of glioblastoma multiforme (GBM) cells to microtubule disruption could open new doors in the search for new anti-GBM treatments. However, the difficulties in developing potent anti-tubulin drugs endowed with improved pharmacokinetic properties necessitates the expansion of medicinal chemistry campaigns. The application of an ensemble pharmacophore screening methodology helped to optimize this process, leading to the development of a new tetrazole-based tubulin inhibitor. Considering this scaffold, we have synthesized a new family of tetrazole derivatives that achieved remarkable antimitotic effects against a broad panel of cancer cells, especially against GBM cells, showing high selectivity in comparison with non-tumor cells. The compounds also exerted high aqueous solubility and were demonstrated to not be substrates of efflux pumps, thus overcoming the main limitations that are usually associated with tubulin binding agents. Tubulin polymerization assays, immunofluorescence experiments, and flow cytometry studies demonstrated that the compounds target tubulin and arrest cells at the G2/M phase followed by induction of apoptosis. The docking experiments agreed with the proposed interactions at the colchicine site and explained the structure-activity relationships.
Collapse
Affiliation(s)
- Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | | | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
44
|
Yang X, Xu L, Yang L, Xu S. Research progress of STAT3-based dual inhibitors for cancer therapy. Bioorg Med Chem 2023; 91:117382. [PMID: 37369169 DOI: 10.1016/j.bmc.2023.117382] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3), a transcription factor, regulates gene levels that are associated with cell survival, cell cycle, and immune reaction. It is correlated with the grade of malignancy and the development of various cancers and targeting STAT3 protein is a potentially promising therapeutic strategy for tumors. Over the past 20 years, various compounds have been found to directly inhibit STAT3 activity via different strategies. However, numerous difficulties exist in the development of STAT3 inhibitors, such as serious toxic effects, poor therapeutic effects, and intrinsic and acquired drug resistance. STAT3 inhibitors synergistically suppress cancer development with additional anti-tumor drugs, such as indoleamine 2,3-dioxygenase 1 inhibitors (IDO1i), histone deacetylase inhibitors (HDACi), DNA inhibitors, pro-tumorigenic cytokine inhibitors (PTCi), NF-κB inhibitors, and tubulin inhibitors. Therefore, individual molecule- based dual-target inhibitors can be the candidate alternative or complementary treatment to overcome the disadvantages of just STAT3 or other targets as a monotherapy. In this review, we discuss the theoretical basis for formulating STAT3-based dual-target inhibitors and also summarize their structure-activity relationships (SARs).
Collapse
Affiliation(s)
- Xiaojuan Yang
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China.
| | - Lu Xu
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China
| | - Li Yang
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China
| | - Shaohong Xu
- School of Pharmacy, Xinxiang University, Xinxiang 453003, China.
| |
Collapse
|
45
|
Ali YA, Soliman HA, Abdel-Gabbar M, Ahmed NA, Attia KAA, Shalaby FM, El-Nahass ES, Ahmed OM. Rutin and Hesperidin Revoke the Hepatotoxicity Induced by Paclitaxel in Male Wistar Rats via Their Antioxidant, Anti-Inflammatory, and Antiapoptotic Activities. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:2738351. [PMID: 37275575 PMCID: PMC10238143 DOI: 10.1155/2023/2738351] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/09/2022] [Accepted: 09/29/2022] [Indexed: 06/07/2023]
Abstract
Paclitaxel, one of the most effective chemotherapeutic drugs, is used to treat various cancers but it is exceedingly toxic when used long-term and can harm the liver. This study aimed to see if rutin, hesperidin, and their combination could protect male Wistar rats against paclitaxel (Taxol)-induced hepatotoxicity. Adult male Wistar rats were subdivided into 5 groups (each of six rats). The normal group was orally given the equivalent volume of vehicles for 6 weeks. The paclitaxel-administered control group received intraperitoneal injection of paclitaxel at a dose of 2 mg/Kg body weight twice a week for 6 weeks. Treated paclitaxel-administered groups were given paclitaxel similar to the paclitaxel-administered control group together with oral supplementation of rutin, hesperidin, and their combination at a dose of 10 mg/Kg body weight every other day for 6 weeks. The treatment of paclitaxel-administered rats with rutin and hesperidin significantly reduced paclitaxel-induced increases in serum alanine transaminase, aspartate transaminase, lactate dehydrogenase, alkaline phosphatase, and gamma-glutamyl transferase activities as well as total bilirubin level and liver lipid peroxidation. However, the levels of serum albumin, liver glutathione content, and the activities of liver superoxide dismutase and glutathione peroxidase increased. Furthermore, paclitaxel-induced harmful hepatic histological changes (central vein and portal area blood vessel congestion, fatty changes, and moderate necrotic changes with focal nuclear pyknosis, focal mononuclear infiltration, and Kupffer cell proliferation) were remarkably enhanced by rutin and hesperidin treatments. Moreover, the elevated hepatic proapoptotic mediator (caspase-3) and pro-inflammatory cytokine (tumor necrosis factor-α) expressions were decreased by the three treatments in paclitaxel-administered rats. The cotreatment with rutin and hesperidin was the most effective in restoring the majority of liver function and histological integrity. Therefore, rutin, hesperidin, and their combination may exert hepatic protective effects in paclitaxel-administered rats by improving antioxidant defenses and inhibiting inflammation and apoptosis.
Collapse
Affiliation(s)
- Yasmine A. Ali
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Hanan A. Soliman
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Mohamed Abdel-Gabbar
- Biochemistry Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Noha A. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Kandil A. A. Attia
- Clinical Nutrition Department, College of Applied Medical Sciences, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
- Department of Evaluation of Natural Resources, Environmental Studies and Research Institute, El-Sadat City University, El-Sadat City 32897, Egypt
| | - Fatma M. Shalaby
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - El-Shaymaa El-Nahass
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| | - Osama M. Ahmed
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O. Box 62521, Beni-Suef, Egypt
| |
Collapse
|
46
|
Hurysz B, Evans BA, Laryea RN, Boyer BE, Coburn TE, Dexter MS, Edwards MA, Faulkner GV, Huss RL, Lafferty MM, Manning M, McNulty M, Melvin SJ, Mitrow CM, Patel RR, Pierce K, Russo J, Seminer AM, Sockett KA, Webster NR, Cole KE, Mowery P, Pelkey ET. Synthesis, Modeling, and Biological Evaluation of Anti-Tubulin Indole-Substituted Furanones. Bioorg Med Chem Lett 2023:129347. [PMID: 37236376 DOI: 10.1016/j.bmcl.2023.129347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/12/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023]
Abstract
Due to the central role of tubulin in various cellular functions, it is a validated target for anti-cancer therapeutics. However, many of the current tubulin inhibitors are derived from complex natural products and suffer from multidrug resistance, low solubility, toxicity issues, and/or the lack of multi-cancer efficacy. As such, there is a continued need for the discovery and development of new anti-tubulin drugs to enter the pipeline. Herein we report on a group of indole-substituted furanones that were prepared and tested for anti-cancer activity. Molecular docking studies showed positive correlations between favorable binding in the colchicine binding site (CBS) of tubulin and anti-proliferative activity, and the most potent compound was found to inhibit tubulin polymerization. These compounds represent a promising new structural motif in the search for small heterocyclic CBS cancer inhibitors.
Collapse
Affiliation(s)
- Brianna Hurysz
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Blake A Evans
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Reuben N Laryea
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, 23606
| | - Brooke E Boyer
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Taylor E Coburn
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Molly S Dexter
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456; Department of Biology, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Marissa A Edwards
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Grace V Faulkner
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Rebecca L Huss
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Megan M Lafferty
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Maegan Manning
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Matthew McNulty
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Sophia J Melvin
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Christina M Mitrow
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Roslyn R Patel
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Kelsey Pierce
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Jack Russo
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Allie M Seminer
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Kaitlynn A Sockett
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Nathan R Webster
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456
| | - Kathryn E Cole
- Department of Molecular Biology and Chemistry, Christopher Newport University, Newport News, VA, 23606.
| | - Patricia Mowery
- Department of Biology, Hobart and William Smith Colleges, Geneva, NY, 14456.
| | - Erin T Pelkey
- Department of Chemistry, Hobart and William Smith Colleges, Geneva, NY, 14456.
| |
Collapse
|
47
|
Abdul Hussein SA, Razzak Mahmood AA, Tahtamouni LH, Balakit AA, Yaseen YS, Al-Hasani RA. New Combretastatin Analogs as Anticancer Agents: Design, Synthesis, Microtubules Polymerization Inhibition, and Molecular Docking Studies. Chem Biodivers 2023; 20:e202201206. [PMID: 36890635 DOI: 10.1002/cbdv.202201206] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/16/2023] [Indexed: 03/10/2023]
Abstract
A new series of 4-(4-methoxyphenyl)-5-(3,4,5-trimethoxyphenyl)-4H-1,2,4-triazole-3-thiol derivatives were synthesized as analogs for the anticancer drug combretastatin A-4 (CA-4) and characterized using FT-IR, 1 H-NMR, 13 CNMR, and HR-MS techniques. The new CA-4 analogs were designed to meet the structural requirements of the highest expected anticancer activity of CA-4 analogs by maintaining ring A 3,4,5-trimethoxyphenyl moiety, and at the same time varying the substituents effect of the triazole moiety (ring B). In silico analysis indicated that compound 3 has higher total energy and dipole moment than colchicine and the other analogs, and it has excellent distribution of electron density and is more stable, resulting in an increased binding affinity during tubulin inhibition. Additionally, compound 3 was found to interact with three apoptotic markers, namely p53, Bcl-2, and caspase 3. Compound 3 showed strong similarity to colchicine, and it has excellent pharmacokinetics properties and a good dynamic profile. The in vitro anti-proliferation studies showed that compound 3 is the most cytotoxic CA-4 analog against cancer cells (IC50 of 6.35 μM against Hep G2 hepatocarcinoma cells), and based on its selectivity index (4.7), compound 3 is a cancer cytotoxic-selective agent. As expected and similar to colchicine, compound 3-treated Hep G2 hepatocarcinoma cells were arrested at the G2/M phase resulting in induction of apoptosis. Compound 3 tubulin polymerization IC50 (9.50 μM) and effect on Vmax of tubulin polymerization was comparable to that of colchicine (5.49 μM). Taken together, the findings of the current study suggest that compound 3, through its binding to the colchicine-binding site at β-tubulin, is a promising microtubule-disrupting agent with excellent potential to be used as cancer therapeutic agent.
Collapse
Affiliation(s)
- Shaker A Abdul Hussein
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Babylon, 51001, Babylon, Iraq
| | - Ammar A Razzak Mahmood
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, 10001, Baghdad, Iraq
| | - Lubna H Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, 13133, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, 80523 Colorado, USA
| | - Asim A Balakit
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Babylon, 51001, Babylon, Iraq
| | - Yahya S Yaseen
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Tikrit, 34001, Tikrit, Iraq
| | - Rehab A Al-Hasani
- Department of Chemistry, College of Science, Al-Mustansiriyah University, 10052, Baghdad, Iraq
| |
Collapse
|
48
|
Dogra A, Kumar J. Biosynthesis of anticancer phytochemical compounds and their chemistry. Front Pharmacol 2023; 14:1136779. [PMID: 36969868 PMCID: PMC10034375 DOI: 10.3389/fphar.2023.1136779] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
Cancer is a severe health issue, and cancer cases are rising yearly. New anticancer drugs have been developed as our understanding of the molecular mechanisms behind diverse solid tumors, and metastatic malignancies have increased. Plant-derived phytochemical compounds target different oncogenes, tumor suppressor genes, protein channels, immune cells, protein channels, and pumps, which have attracted much attention for treating cancer in preclinical studies. Despite the anticancer capabilities of these phytochemical compounds, systemic toxicity, medication resistance, and limited absorption remain more significant obstacles in clinical trials. Therefore, drug combinations of new phytochemical compounds, phytonanomedicine, semi-synthetic, and synthetic analogs should be considered to supplement the existing cancer therapies. It is also crucial to consider different strategies for increased production of phytochemical bioactive substances. The primary goal of this review is to highlight several bioactive anticancer phytochemical compounds found in plants, preclinical research, their synthetic and semi-synthetic analogs, and clinical trials. Additionally, biotechnological and metabolic engineering strategies are explored to enhance the production of bioactive phytochemical compounds. Ligands and their interactions with their putative targets are also explored through molecular docking studies. Therefore, emphasis is given to gathering comprehensive data regarding modern biotechnology, metabolic engineering, molecular biology, and in silico tools.
Collapse
|
49
|
Yuan M, Su J, Zhang Y, Qin J, Yang H, Duan Y, Yao Y, Sun M. Design, synthesis and biological evaluation of novel tubulin inhibitors targeting colchicine sites. Bioorg Med Chem Lett 2023; 83:129166. [PMID: 36736495 DOI: 10.1016/j.bmcl.2023.129166] [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: 09/19/2022] [Revised: 01/26/2023] [Accepted: 01/28/2023] [Indexed: 02/01/2023]
Abstract
Tubulin, a potential target for antitumor drug discovery, contains three main binding sites for clinical inhibitors: colchicine, vinblastine, and paclitaxel. CA-4 has been reported to be a classic tubulin inhibitor targeting the colchicine site. Herein, based on the structural modification of CA-4, 48 novel compounds were designed and synthesized by selecting structural fragments with various biological activities to replace the cis double bond of CA-4. Among these compounds, compound 8p was the most effective tubulin inhibitor (IC50 = 65 nM aganist HepG2 cells). Immunofluorescence experiment confirmed the anti-tumor effect of 8p by destroying the network structure of microtubules. Further studies showed that 8p induced tumor cell apoptosis, arrested cell cycle, inhibited tumor cell migration and invasion.
Collapse
Affiliation(s)
- Minghua Yuan
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Jingtian Su
- 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
| | - Hua Yang
- 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
| | - Yongfang Yao
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Moran Sun
- School of Pharmaceutical Sciences, and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
50
|
Nicolella HD, Ribeiro AB, Munari CC, Melo MR, Ozelin SD, da Silva LHD, Marquele-Oliveira F, Orenha RP, Veneziani RCS, Parreira RLT, Tavares DC. Antimelanoma effect of manool in 2D cell cultures and reconstructed human skin models. J Biochem Mol Toxicol 2023; 37:e23282. [PMID: 36541366 DOI: 10.1002/jbt.23282] [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: 03/28/2022] [Revised: 11/03/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Melanoma is the most aggressive and lethal type of skin cancer, characterized by therapeutic resistance. In this context, the present study aimed to investigate the cytotoxic potential of manool, a diterpene from Salvia officinalis L., in human (A375) and murine (B16F10) melanoma cell lines. The analysis of cytotoxicity using the XTT assay showed the lowest IC50 after 48 h of treatment with the manool, being 17.6 and 18.2 µg/ml for A375 and B16F10, respectively. A selective antiproliferative effect of manool was observed on the A375 cells based on the colony formation assay, showing an IC50 equivalent to 5.6 µg/ml. The manool treatments led to 43.5% inhibition of the A375 cell migration at a concentration of 5.0 µg/ml. However, it did not affect cell migration in the B16F10 cells. Cell cycle analysis revealed that the manool interfered in the cell cycle of the A375 cells, blocking the G2/M phase. No changes in the cell cycle were observed in the B16F10 cells. Interestingly, manool did not induce apoptosis in the A375 cells, but apoptosis was observed after treatment of the B16F10 cells. Additionally, manool showed an antimelanoma effect in a reconstructed human skin model. Furthermore, in silico studies, showed that manool is stabilized in the active sites of the tubulin dimer with comparable energy concerning taxol, indicating that both structures can inhibit the proliferation of cancer cells. Altogether, it is concluded that manool, through the modulation of the cell cycle, presents a selective antiproliferative activity and a potential antimelanoma effect.
Collapse
|