1
|
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
|
2
|
Chen Z, Geng DW, Yuan TB, Yu C, Cai DW, Yin Y, Shen Q. Design and evaluation of novel N-substituent diphenylamine derivatives as tubulin colchicine binding site inhibitors. Bioorg Med Chem Lett 2025; 115:130031. [PMID: 39557311 DOI: 10.1016/j.bmcl.2024.130031] [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: 06/12/2024] [Revised: 09/30/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Novel N-substituent diphenylamine derivatives as tubulin inhibitors targeting colchicine-binding site have been designed based on structural simplification and structural fusing strategy. Most designed compounds exhibited the moderate or potent antiproliferative activities against five cancer cell lines. Among them, compound 4k displayed the significant selectivity for osteosarcoma cells MG-63 and U2OS with the IC50 value of 0.08-0.14 μM. Further investigations verified 4k could inhibit tubulin polymerization by targeting colchicine binding site. Meanwhile, compound 4k not only effectively induced tumor cell cycle arrest at the G2/M phase, but also slightly induced cell apoptosis. These results indicated that N-substituent of diphenylamine derivatives are deserved for further development as tubulin colchicine binding site inhibitors.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Orthopaedics, Shanghai General Hospital of Nanjing Medical University, Shanghai, China; Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Da-Wei Geng
- Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Tang-Bo Yuan
- Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Chen Yu
- Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Da-Wei Cai
- Department of Orthopaedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Qiang Shen
- Department of Orthopaedics, Shanghai General Hospital of Nanjing Medical University, Shanghai, China.
| |
Collapse
|
3
|
Zhu L, Zhang M, Leng J, Zhao B, Ning M, Zhang C, Kong L, Yin Y. Discovery of novel quinazoline derivatives as tubulin polymerization inhibitors targeting the colchicine binding site with potential anti-colon cancer effects. Eur J Med Chem 2024; 280:117000. [PMID: 39489984 DOI: 10.1016/j.ejmech.2024.117000] [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: 09/10/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Tubulin is a critical target for cancer therapy, with colchicine binding site inhibitors (CBSIs) being the most extensively researched. A series of quinazoline derivatives designed to target the colchicine binding site of tubulin were synthesized and evaluated for their biological activities. The antiproliferative effects of these compounds were tested against six human cancer cell lines, and compound Q19 demonstrated potent antiproliferative activity against the HT-29 cell line, with an IC50 value of 51 nM. Additionally, further investigation revealed that Q19 effectively inhibited microtubule polymerization by binding to the colchicine binding site on tubulin. Furthermore, compound Q19 arrested the HT-29 cell cycle at the G2/M phase, induced apoptosis in these cells, and disrupted angiogenesis. Finally, compound Q19 exhibited potent inhibitory effects on tumor growth in HT-29 xenografted mice while demonstrating minimal toxic side effects and acceptable pharmacokinetic properties. These findings suggested that Q19 hold promise as a potential candidate for colon cancer therapy targeting tubulin.
Collapse
Affiliation(s)
- Liqiao Zhu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Mengyu Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Jiafu Leng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Bo Zhao
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Mengdan Ning
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Chao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| | - Yong Yin
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
4
|
Omar MH, Emam SH, Mikhail DS, Elmeligie S. Combretastatin A-4 based compounds as potential anticancer agents: A review. Bioorg Chem 2024; 153:107930. [PMID: 39504638 DOI: 10.1016/j.bioorg.2024.107930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/13/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024]
Abstract
The current review discusses the importance of combretastatin A-4 (CA-4) as a lead compound of microtubule targeting agents. CA-4 holds a unique place among naturally occurring compounds having cytotoxic activity. In this review an overall picture of design strategies, structure-activity relationship, synthesis, cytotoxic activity, and binding interactions of promising CA-4 analogues, are discussed and arranged chronologically from 2016 to early 2023. Also, this review emphasizes their biological activity as anticancer agents, within an overview of clinical application limitation and suggested strategies to overcome. Dual targeting tubulin inhibitors showed highpotentialto surpass medication resistance and provide synergistic efficacy. Linking platinum (IV), amino acids, and HDAC targeting moieties to active tubulin inhibitorsproduced potent active compounds. Analogues of CA-4 bridged with azetidin-2-one, pyrazole, sulfide, or carrying selenium atom exhibited cytotoxic action against a variety of malignant cell lines through different pathways.
Collapse
Affiliation(s)
- Mai H Omar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Soha H Emam
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Demiana S Mikhail
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Salwa Elmeligie
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
5
|
Salarinejad S, Seyfi S, Hayashi S, Moghimi S, Toolabi M, Taslimi P, Firoozpour L, Usui T, Foroumadi A. Design, synthesis, and biological evaluation of new biaryl derivatives of cycloalkyl diacetamide bearing chalcone moiety as type II c-MET kinase inhibitors. Mol Divers 2024; 28:4167-4180. [PMID: 38466553 DOI: 10.1007/s11030-024-10807-x] [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: 06/27/2023] [Accepted: 01/05/2024] [Indexed: 03/13/2024]
Abstract
Many human cancers have been associated with the deregulation of the mesenchymal-epithelial transition factor tyrosine kinase (MET) receptor, a promising drug target for anticancer drug discovery. Herein, we report the discovery of a novel structure of potent chalcone-based derivatives type II c-Met inhibitors which are comparable to Foretinib (IC50 = 14 nM) as a potent reference drug. Based on our design strategy, we also expected an anti-tubulin activity for the compounds. However, the weak inhibitory effects on microtubules were confirmed by cell cycle analyses implicated that the observed cytotoxicity against HeLa cells probably was not derived from tubulin inhibition. Compounds 14q and 14k with IC50 values of 24 nM and 45 nM, respectively, demonstrated favorable inhibition of MET kinase activity, and desirable bonding interactions in the ligand-MET enzyme complex stability in molecular docking studies.
Collapse
Affiliation(s)
- Somayeh Salarinejad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Seyfi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Seiko Hayashi
- Institute of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan
| | - Setareh Moghimi
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Toolabi
- Department of Medicinal Chemistry, School of Pharmacy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74100, Bartin, Turkey
| | - Loghman Firoozpour
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran
| | - Takeo Usui
- Institute of Life and Environmental Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki, 305-8572, Japan.
| | - Alireza Foroumadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Bakchi B, Devabattula G, Maddipatla S, Singampalli A, Kumar Porna D, Nanduri S, Sharma A, Godugu C, Yaddanapudi VM. New Indole-Based Phenylthiazolyl-2,4-dihydropyrazolones as Tubulin polymerization inhibitors: Multicomponent Synthesis, Cytotoxicity Evaluation, and in silico Studies. ChemMedChem 2024:e202400817. [PMID: 39523484 DOI: 10.1002/cmdc.202400817] [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: 10/17/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
A facile multicomponent synthesis of new indole-based phenylthiazolyl-dihydropyrazolone hybrids, their structural characterization, biological evaluation, and in silico investigations as anticancer agents are reported. Lead molecule 5 i of the series showed potent activity against MCF-7 breast cancer cells with an IC50 of 3.92±0.01 μM while showing minimal toxicity to normal human lung cells (IC50=69.85±3.95 μM). Further studies show that the compound exhibits antiproliferative activity by inducing apoptosis in MCF-7 cancer cells. The wound healing assay indicated impaired cell migration under the concentration-dependent dosage. The lead molecule 5 i also successfully inhibited the tubulin polymerase enzyme with an IC50 of 4.16±0.18 μM. A flow cytometric assay indicated compound 5 i induced apoptosis through G0 phase cell cycle arrest. The binding mode and interactions of the compound with the tubulin were predicted by molecular modelling and calculating binding free energies. These findings explain the current series as a new class of microtubule polymerization inhibitors with anticancer activity suitable for developing anticancer agents targeting tubulin.
Collapse
Affiliation(s)
- Bulti Bakchi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Geetanjali Devabattula
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Sarvan Maddipatla
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Anuradha Singampalli
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Dileep Kumar Porna
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Srinivas Nanduri
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Anamika Sharma
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Chandraiah Godugu
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| | - Venkata Madhavi Yaddanapudi
- Department of Chemical Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad 500037, Telangana, India
| |
Collapse
|
7
|
Xu M, Xiao X, Chen Y, Zhou X, Parisi L, Ma R. 3D physiologically-informed deep learning for drug discovery of a novel vascular endothelial growth factor receptor-2 (VEGFR2). Heliyon 2024; 10:e35769. [PMID: 39220924 PMCID: PMC11365333 DOI: 10.1016/j.heliyon.2024.e35769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Angiogenesis is an essential process in tumorigenesis, tumor invasion, and metastasis, and is an intriguing pathway for drug discovery. Targeting vascular endothelial growth factor receptor 2 (VEGFR2) to inhibit tumor angiogenic pathways has been widely explored and adopted in clinical practice. However, most drugs, such as the Food and Drug Administration -approved drug axitinib (ATC code: L01EK01), have considerable side effects and limited tolerability. Therefore, there is an urgent need for the development of novel VEGFR2 inhibitors. In this study, we propose a novel strategy to design potential candidates targeting VEGFR2 using three-dimensional (3D) deep learning and structural modeling methods. A geometric-enhanced molecular representation learning method (GEM) model employing a graph neural network (GNN) as its underlying predictive algorithm was used to predict the activity of the candidates. In the structural modeling method, flexible docking was performed to screen data with high affinity and explore the mechanism of the inhibitors. Small -molecule compounds with consistently improved properties were identified based on the intersection of the scores obtained from both methods. Candidates identified using the GEM-GNN model were selected for in silico modeling using molecular dynamics simulations to further validate their efficacy. The GEM-GNN model enabled the identification of candidate compounds with potentially more favorable properties than the existing drug, axitinib, while achieving higher efficacy.
Collapse
Affiliation(s)
- Mengyang Xu
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, Guangdong, China
| | - Xiaoyue Xiao
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, Guangdong, China
| | - Yinglu Chen
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, Guangdong, China
| | - Xiaoyan Zhou
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, Guangdong, China
| | - Luca Parisi
- Department of Computer Science, Tutorantis, Edinburgh, EH2 4AN, Scotland, United Kingdom
| | - Renfei Ma
- Faculty of Biology, Shenzhen MSU-BIT University, Shenzhen, 518172, Guangdong, China
| |
Collapse
|
8
|
Lu L, Li K, Pu J, Wang S, Liang T, Wang J. Dual-target inhibitors of colchicine binding site for cancer treatment. Eur J Med Chem 2024; 274:116543. [PMID: 38823265 DOI: 10.1016/j.ejmech.2024.116543] [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: 04/26/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/03/2024]
Abstract
Colchicine binding site inhibitors (CBSIs) have attracted much attention due to their antitumor efficacies and the advantages of inhibiting angiogenesis and overcoming multidrug resistance. However, no CBSI has been currently approved for cancer treatment due to the insufficient efficacies, serious toxicities and poor pharmacokinetic properties. Design of dual-target inhibitors is becoming a potential strategy for cancer treatment to improve anticancer efficacy, decrease adverse events and overcome drug resistance. Therefore, we reviewed dual-target inhibitors of colchicine binding site (CBS), summarized the design strategies and the biological activities of these dual-target inhibitors, expecting to provide inspiration for developing novel dual inhibitors based on CBS.
Collapse
Affiliation(s)
- Lu Lu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan Province, 475004, China
| | - Keke Li
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan Province, 475004, China
| | - Jiaxin Pu
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan Province, 475004, China
| | - Shaochi Wang
- Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Tingting Liang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan Province, 475004, China; The Zhongzhou Laboratory for Integrative Biology, Henan University, Zhengzhou, Henan Province, 450000, China.
| | - Jianhong Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng, Henan Province, 475004, China.
| |
Collapse
|
9
|
Liu Y, Zhang J, Tian J, Wang C, Wang T, Gong J, Hu L. Structure-activity relationship study of new carbazole sulfonamide derivatives as anticancer agents with dual-target mechanism. Eur J Med Chem 2024; 273:116509. [PMID: 38781920 DOI: 10.1016/j.ejmech.2024.116509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/04/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024]
Abstract
A series of novel carbazole sulfonamide derivatives were synthesized and evaluated for antiproliferative activity. Among them, compounds 7 and 15 showed strong potency (IC50 values of 0.81-31.19 nM) against five different cancer cells including multidrug-resistant MCF7/ADR cells. Compound 15 displayed a high cancer cell selectivity (IC50(L02)/average IC50: SI = 7.7). The l-valine prodrug 7a and the phosphate prodrug 15a exerted rohust in vivo antitumor efficacies and accepted safety prolifes. Further mechanism studies revealed that 7 and 15 directly bind to the colchicine site in tubulin to block tubulin polymerization, promote microtubule fragmentation at the cellular level, and induce apoptosis with G2/M cell cycle arrest. These compounds also inhibit HEMC-1 cells migration and vascular tube formation. Additionally, compound 7 displayed a selective inhibition of Topo I. Collectively, these studies suggest that 7 and 15 represents a promising new generation of tubulin inhibitors for cancer treatment.
Collapse
Affiliation(s)
- Yonghua Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Junyi Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaqi Tian
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengxi Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tianqi Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianhua Gong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Laixing Hu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Ma Y, Wang T, Cheng L, Ma X, Li R, Zhang M, Chen J, Zhao P. Design, concise synthesis and evaluation of novel amide-based combretastatin A-4 analogues as potent tubulin inhibitors. Bioorg Med Chem Lett 2024; 108:129816. [PMID: 38806101 DOI: 10.1016/j.bmcl.2024.129816] [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: 04/08/2024] [Revised: 05/23/2024] [Accepted: 05/25/2024] [Indexed: 05/30/2024]
Abstract
As our ongoing work, a novel series of the amide-based CA-4 analogues were successfully designed, synthesized, and explored for their biological evaluation. Among these compounds, 7d and 8a illustrated most potent antiproliferative activity toward A549, HeLa, HCT116, and HT-29 cell lines. Most importantly, these two compounds didn't display noticeable cytotoxic activity on the non-tumoural cell line HEK-293. Further mechanism studies revealed that analogue 8a was identified as a novel tubulin polymerization inhibitor with an IC50 value of 6.90 μM, which is comparable with CA-4. The subsequent investigations unveiled that analogue 8a not only effectively caused cell cycle arrest at the G2/M phase but also induced apoptosis in A549 cells via a concentration-dependent manner. The molecular docking revealed that 8a could occupy well the colchicine-binding site of tubulin. Collectively, these findings indicate that amide-based CA-4 scaffold could be worthy of further evaluation for development of novel tubulin inhibitors with improved safety profile.
Collapse
Affiliation(s)
- Yufeng Ma
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China
| | - Ting Wang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China
| | - Li Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China
| | - Xuanxuan Ma
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China
| | - Rou Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China
| | - Mengting Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China
| | - Jingkao Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China.
| | - Peiliang Zhao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
11
|
O'Brien NS, Gilbert J, McCluskey A, Sakoff JA. 2,3-Dihydroquinazolin-4(1 H)-ones and quinazolin-4(3 H)-ones as broad-spectrum cytotoxic agents and their impact on tubulin polymerisation. RSC Med Chem 2024; 15:1686-1708. [PMID: 38784470 PMCID: PMC11110758 DOI: 10.1039/d3md00600j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/27/2024] [Indexed: 05/25/2024] Open
Abstract
Tubulin plays a central role in mitosis and has been the target of multiple anticancer drugs, including paclitaxel. Herein two separate families of 2,3-dihydroquinazoline-4(1H)-ones and quinazoline-4(3H) ones, comprising 57 compounds in total, were synthesised. Screening against a broad panel of human cancer cell lines (HT29 colon, U87 and SJ-G2 glioblastoma, MCF-7 breast, A2780 ovarian, H460 lung, A431 skin, Du145 prostate, BE2-C neuroblastoma, and MIA pancreas) reveals these analogues to be broad spectrum cytotoxic compounds. Of particular note, 2-styrylquinazolin-4(3H)-one 51, 2-(4-hydroxystyryl)quinazolin-4(3H)-one 63, 2-(2-methoxystyryl)quinazolin-4(3H)-one 64 and 2-(3-methoxystyryl)quinazolin-4(3H)-one 65 and 2-(naphthalen-1-yl)-2,3-dihydroquinazolin-4(1H)-one 39 exhibited sub-μM potency growth inhibition values. Of these 1-naphthyl 39 has activity <50 nM against the HT29, U87, A2780, H460 and BE2-C cell lines. Molecular modelling of these compounds, e.g. 2-(naphthalen-1-yl)-2,3-dihydroquinazolin-4(1H)-one 39, 2-(2-methoxystyryl)quinazolin-4(3H)-one 64, 2-(3-methoxystyryl)quinazolin-4(3H)-one 65, and 2-(4-methoxystyryl)quinazolin-4(3H)-one 50 docked to the known tubulin polymerisation inhibitor sites highlighted well conserved interactions within the colchicine binding pocket. These compounds were examined in a tubulin polymerisation assay alongside the known tubulin polymerisation promotor, paclitaxel (69), and tubulin inhibitor, nocodazole (68). Of the analogues examined, indoles 43 and 47 were modest promotors of tubulin polymerisation, but less effective than paclitaxel. Analogues 39, 64, and 65 showed reduced microtubule formation consistent with tubulin inhibition. The variation in ring methoxy substituent with 50, 64 and 65, from o- to m- to p-, results in a concomitant reduction in cytotoxicity and a reduction in tubulin polymerisation, with p-OCH350 being the least active in this series of analogues. This presents 64 as a tubulin polymerisation inhibitor possessing novel chemotype and sub micromolar cytotoxicity. Naphthyl 39, with complete inhibition of tubulin polymerisation, gave rise to a sub 0.2 μM cell line cytotoxicity. Compounds 39 and 64 induced G2 + M cell cycle arrest indicative of inhibition of tubulin polymerisation, with 39 inducing an equivalent effect on cell cycle arrest as nocodazole (68).
Collapse
Affiliation(s)
- Nicholas S O'Brien
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia +61(0)249215472 +61(0)249216486
| | - Jayne Gilbert
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah 2298 NSW Australia
| | - Adam McCluskey
- Chemistry, School of Environmental & Life Sciences, The University of Newcastle University Drive Callaghan NSW 2308 Australia +61(0)249215472 +61(0)249216486
| | - Jennette A Sakoff
- Experimental Therapeutics Group, Department of Medical Oncology, Calvary Mater Newcastle Hospital Edith Street Waratah 2298 NSW Australia
| |
Collapse
|
12
|
Shan P, Ye T, Tang YD, Song H, Wang C, Zhu K, Yang F, Zhang SL, Su PW, Gao S, Zhang H. First total synthesis, antitumor evaluation and target identification of mornaphthoate E: A new tubulin inhibitor template acting on PI3K/Akt signaling pathway. Acta Pharm Sin B 2024; 14:2177-2193. [PMID: 38799630 PMCID: PMC11120283 DOI: 10.1016/j.apsb.2024.02.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 05/29/2024] Open
Abstract
Mornaphthoate E (MPE) is a prenylated naphthoic acid methyl ester isolated from the roots of a famous Chinese medicinal plant Morinda officinalis and shows remarkable cytotoxicity against several human tumor cell lines. In the current project, the first total synthesis of (±)-MPE was achieved in seven steps and 5.6% overall yield. Then the in vitro anti-tumor activity of MPE was first assessed for both enantiomers in two breast cancer cells, with the levoisomer exerting slightly better potency. The in vivo anti-tumor effect was further verified by applying the racemate in an orthotopic autograft mouse model. Notably, MPE exerted promising anti-metastasis activity both in vitro and in vivo and showed no obvious toxicity on mice at the therapeutic dosage. Mechanistic investigations demonstrated that MPE acted as a tubulin polymerization stabilizer and disturbed the dynamic equilibrium of microtubules via regulating PI3K/Akt signaling. In conclusion, our work has provided a new chemical template for the future design and development of next-generation tubulin-targeting chemotherapies.
Collapse
Affiliation(s)
- Peipei Shan
- Institute of Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Tao Ye
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Ying-De Tang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Hui Song
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Chao Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kongkai Zhu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Feifei Yang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Shi-Lei Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215127, China
| | - Pei-Wen Su
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Shuanhu Gao
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| |
Collapse
|
13
|
Yakkala PA, Rahaman S, Soukya PSL, Begum SA, Kamal A. An update on the development on tubulin inhibitors for the treatment of solid tumors. Expert Opin Ther Targets 2024; 28:193-220. [PMID: 38618889 DOI: 10.1080/14728222.2024.2341630] [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: 12/29/2023] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Abstract
INTRODUCTION Microtubules play a vital role in cancer therapeutics. They are implicated in tumorigenesis, thus inhibiting tubulin polymerization in cancer cells, and have now become a significant target for anticancer drug development. A plethora of drug molecules has been crafted to influence microtubule dynamics and presently, numerous tubulin inhibitors are being investigated. This review discusses the recently developed inhibitors including natural products, and also examines the preclinical and clinical data of some potential molecules. AREA COVERED The current review article summarizes the development of tubulin inhibitors while detailing their specific binding sites. It also discusses the newly designed inhibitors that may be useful in the treatment of solid tumors. EXPERT OPINION Microtubules play a crucial role in cellular processes, especially in cancer therapy where inhibiting tubulin polymerization holds promise. Ongoing trials signify a commitment to revolutionizing cancer treatment and exploring targeted therapies. Challenges in microtubule modulation, like resistance and off-target effects, demand focused efforts, emphasizing combination therapies and personalized treatments. Beyond microtubules, promising avenues in cancer research include immunotherapy, genomic medicine, CRISPR gene editing, liquid biopsies, AI diagnostics, and stem cell therapy, showcasing a holistic approach for future advancements.
Collapse
Affiliation(s)
- Prasanna Anjaneyulu Yakkala
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shaik Rahaman
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - P S Lakshmi Soukya
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad, India
| | - Sajeli Ahil Begum
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad, India
| | - Ahmed Kamal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Hyderabad, India
- Department of Environment, Forests, Science & Technology, Telangana State Council of Science & Technology, Hyderabad, India
| |
Collapse
|
14
|
Al Sbihi A, Alasfour M, Pongas G. Innovations in Antibody-Drug Conjugate (ADC) in the Treatment of Lymphoma. Cancers (Basel) 2024; 16:827. [PMID: 38398219 PMCID: PMC10887180 DOI: 10.3390/cancers16040827] [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: 01/21/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Chemoimmunotherapy and cellular therapy are the mainstay of the treatment of relapsed/refractory (R/R) lymphomas. Development of resistance and commonly encountered toxicities of these treatments limit their role in achieving desired response rates and durable remissions. The Antibody-Drug Conjugate (ADC) is a novel class of targeted therapy that has demonstrated significant efficacy in treating various cancers, including lymphomas. To date, three ADC agents have been approved for different lymphomas, marking a significant advancement in the field. In this article, we aim to review the concept of ADCs and their application in lymphoma treatment, provide an analysis of currently approved agents, and discuss the ongoing advancements of ADC development.
Collapse
Affiliation(s)
| | | | - Georgios Pongas
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
15
|
Podolak M, Holota S, Deyak Y, Dziduch K, Dudchak R, Wujec M, Bielawski K, Lesyk R, Bielawska A. Tubulin inhibitors. Selected scaffolds and main trends in the design of novel anticancer and antiparasitic agents. Bioorg Chem 2024; 143:107076. [PMID: 38163424 DOI: 10.1016/j.bioorg.2023.107076] [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: 10/04/2023] [Revised: 12/02/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Design of tubulin inhibitors as anticancer drugs dynamically developed over the past 20 years. The modern arsenal of potential tubulin-targeting anticancer agents is represented by small molecules, monoclonal antibodies, and antibody-drug conjugates. Moreover, targeting tubulin has been a successful strategy in the development of antiparasitic drugs. In the present review, an overall picture of the research and development of potential tubulin-targeting agents using small molecules between 2018 and 2023 is provided. The data about some most often used and prospective chemotypes of small molecules (privileged heterocycles, moieties of natural molecules) and synthetic methodologies (analogue-based, fragment-based drug design, molecular hybridization) applied for the design of novel agents with an impact on the tubulin system are summarized. The design and prospects of multi-target agents with an impact on the tubulin system were also highlighted. Reported in the review data contribute to the "structure-activity" profile of tubulin-targeting small molecules as anticancer and antiparasitic agents and will be useful for the application by medicinal chemists in further exploration, design, improvement, and optimization of this class of molecules.
Collapse
Affiliation(s)
- Magdalena Podolak
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine
| | - Yaroslava Deyak
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; Department of Pharmaceutical Disciplines, Uzhhorod National University, Narodna Square 3, 88000 Uzhhorod, Ukraine
| | - Katarzyna Dziduch
- Doctoral School, Medical University of Lublin, Chodzki 7, 20-093 Lublin, Poland
| | - Rostyslav Dudchak
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Monika Wujec
- Department of Organic Chemistry, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine.
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| |
Collapse
|
16
|
Ren W, Deng Y, Ward JD, Vairin R, Bai R, Wanniarachchi HI, Hamal KB, Tankoano PE, Tamminga CS, Bueno LMA, Hamel E, Mason RP, Trawick ML, Pinney KG. Synthesis and biological evaluation of structurally diverse 6-aryl-3-aroyl-indole analogues as inhibitors of tubulin polymerization. Eur J Med Chem 2024; 263:115794. [PMID: 37984295 PMCID: PMC11019941 DOI: 10.1016/j.ejmech.2023.115794] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 11/22/2023]
Abstract
The synthesis and evaluation of small-molecule inhibitors of tubulin polymerization remains a promising approach for the development of new therapeutic agents for cancer treatment. The natural products colchicine and combretastatin A-4 (CA4) inspired significant drug discovery campaigns targeting the colchicine site located on the beta-subunit of the tubulin heterodimer, but so far these efforts have not yielded an approved drug for cancer treatment in human patients. Interest in the colchicine site was enhanced by the discovery that a subset of colchicine site agents demonstrated dual functionality as both potent antiproliferative agents and effective vascular disrupting agents (VDAs). Our previous studies led to the discovery and development of a 2-aryl-3-aroyl-indole analogue (OXi8006) that inhibited tubulin polymerization and demonstrated low nM IC50 values against a variety of human cancer cell lines. A water-soluble phosphate prodrug salt (OXi8007), synthesized from OXi8006, displayed promising vascular disrupting activity in mouse models of cancer. To further extend structure-activity relationship correlations, a series of 6-aryl-3-aroyl-indole analogues was synthesized and evaluated for their inhibition of tubulin polymerization and cytotoxicity against human cancer cell lines. Several structurally diverse molecules in this small library were strong inhibitors of tubulin polymerization and of MCF-7 and MDA-MB-231 human breast cancer cells. One of the most promising analogues (KGP591) caused significant G2/M arrest of MDA-MB-231 cells, disrupted microtubule structure and cell morphology in MDA-MB-231 cells, and demonstrated significant inhibition of MDA-MB-231 cell migration in a wound healing (scratch) assay. A phosphate prodrug salt, KGP618, synthesized from its parent phenolic precursor, KGP591, demonstrated significant reduction in bioluminescence signal when evaluated in vivo against an orthotopic model of kidney cancer (RENCA-luc) in BALB/c mice, indicative of VDA efficacy. The most active compounds from this series offer promise as anticancer therapeutic agents.
Collapse
Affiliation(s)
- Wen Ren
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Yuling Deng
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Jacob D Ward
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Rebecca Vairin
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Hashini I Wanniarachchi
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Khagendra B Hamal
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Pouguiniseli E Tankoano
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Caleb S Tamminga
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Lorena M A Bueno
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, MD, 21702, United States.
| | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-9058, United States.
| | - Mary Lynn Trawick
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| | - Kevin G Pinney
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place, No. 97348, Waco, TX, 76798-7348, United States.
| |
Collapse
|
17
|
Trivedi A, Hasan A, Ahmad R, Siddiqui S, Srivastava A, Misra A, Mir SS. Flavonoid Myricetin as Potent Anticancer Agent: A Possibility towards Development of Potential Anticancer Nutraceuticals. Chin J Integr Med 2024; 30:75-84. [PMID: 37340205 DOI: 10.1007/s11655-023-3701-5] [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] [Accepted: 03/01/2023] [Indexed: 06/22/2023]
Abstract
Good nutrition plays a crucial role in maintaining a balanced lifestyle. The beneficial effects of nutrition have been found to counteract nutritional disturbances with the expanded use of nutraceuticals to treat and manage cardiovascular diseases, cancer, and other developmental defects over the last decade. Flavonoids are found abundantly in plant-derived foods such as fruits, vegetables, tea, cocoa, and wine. Fruits and vegetables contain phytochemicals like flavonoids, phenolics, alkaloids, saponins, and terpenoids. Flavonoids can act as anti-inflammatory, anti-allergic, anti-microbial (antibacterial, antifungal, and antiviral) antioxidant, anti-cancer, and anti-diarrheal agents. Flavonoids are also reported to upregulate apoptotic activity in several cancers such as hepatic, pancreatic, breast, esophageal, and colon. Myricetin is a flavonol which is naturally present in fruits and vegetables and has shown possible nutraceutical value. Myricetin has been portrayed as a potent nutraceutical that may protect against cancer. The focus of the present review is to present an updated account of studies demonstrating the anticancer potential of myricetin and the molecular mechanisms involved therein. A better understanding of the molecular mechanism(s) underlying its anticancer activity would eventually help in its development as a novel anticancer nutraceutical having minimal side effects.
Collapse
Affiliation(s)
- Anchal Trivedi
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Adria Hasan
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India
| | - Rumana Ahmad
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Sahabjada Siddiqui
- Department of Biotechnology, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Aditi Srivastava
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Aparna Misra
- Department of Biochemistry, Era's Lucknow Medical College & Hospital, Era University, Lucknow, 226003, India
| | - Snober S Mir
- Molecular Cell Biology Laboratory, Integral Information and Research Centre-4 (IIRC-4), Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, 226026, India.
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 226026, India.
| |
Collapse
|
18
|
Peerzada MN, Dar MS, Verma S. Development of tubulin polymerization inhibitors as anticancer agents. Expert Opin Ther Pat 2023; 33:797-820. [PMID: 38054831 DOI: 10.1080/13543776.2023.2291390] [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: 05/17/2023] [Accepted: 12/01/2023] [Indexed: 12/07/2023]
Abstract
INTRODUCTION Microtubules are intracellular, filamentous, polymeric structures that extend throughout the cytoplasm, composed of α-tubulin and β-tubulin subunits. They regulate many cellular functions including cell polarity, cell shape, mitosis, intracellular transport, cell signaling, gene expression, cell integrity, and are associated with tumorigenesis. Inhibition of tubulin polymerization within tumor cells represents a crucial focus in the pursuit of developing anticancer treatments. AREAS COVERED This review focuses on the natural product and their synthetic congeners as tubulin inhibitors along with their site of interaction on tubulin. This review also covers the developed novel tubulin inhibitors and important patents focusing on the development of tubulin inhibition for cancer treatment reported from 2018 to 2023. The scientific and patent literature has been searched on PubMed, Espacenet, ScienceDirect, and Patent Guru from 2018-2023. EXPERT OPINION Tubulin is one of the promising targets explored extensively for drug discovery. Compounds binding in the colchicine site could be given importance because they can elude resistance mediated by the P-glycoprotein efflux pump and no colchicine site binding inhibitor is approved by FDA so far. The research on the development of antibody drug conjugates (ADCs) for tubluin polymerization inhibition could be significant strategy for cancer treatment.
Collapse
Affiliation(s)
- Mudasir Nabi Peerzada
- Tumor Biology Department, Drug Discovery Laboratory, National Institute of Pathology, Indian Council of Medical Research, Safdarjung Hospital Campus, New Delhi, India
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Mohammad Sultan Dar
- Department of Neurosurgery, Sub-District Hospital Sopore, Jammu and Kashmir, India
| | - Saurabh Verma
- Tumor Biology Department, Drug Discovery Laboratory, National Institute of Pathology, Indian Council of Medical Research, Safdarjung Hospital Campus, New Delhi, India
| |
Collapse
|
19
|
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
|
20
|
Ahsan MJ, Gautam K, Ali A, Ali A, Altamimi ASA, Salahuddin, Alossaimi MA, Lakshmi SVVNSM, Ahsan MF. Synthesis, Anticancer Activity, and In Silico Studies of 5-(3-Bromophenyl)- N-aryl-4 H-1,2,4-triazol-3-amine Analogs. Molecules 2023; 28:6936. [PMID: 37836779 PMCID: PMC10574406 DOI: 10.3390/molecules28196936] [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: 08/25/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023] Open
Abstract
In the current study, we described the synthesis of ten new 5-(3-Bromophenyl)-N-aryl-4H-1,2,4-triazol-3-amine analogs (4a-j), as well as their characterization, anticancer activity, molecular docking studies, ADME, and toxicity prediction. The title compounds (4a-j) were prepared in three steps, starting from substituted anilines in a satisfactory yield, followed by their characterization via spectroscopic techniques. The National Cancer Institute (NCI US) protocol was followed to test the compounds' (4a-j) anticancer activity against nine panels of 58 cancer cell lines at a concentration of 10-5 M, and growth percent (GP) as well as percent growth inhibition (PGI) were calculated. Some of the compounds demonstrated significant anticancer activity against a few cancer cell lines. The CNS cancer cell line SNB-75, which showed a PGI of 41.25 percent, was discovered to be the most sensitive cancer cell line to the tested compound 4e. The mean GP of compound 4i was found to be the most promising among the series of compounds. The five cancer cell lines that were found to be the most susceptible to compound 4i were SNB-75, UO-31, CCRF-CEM, EKVX, and OVCAR-5; these five cell lines showed PGIs of 38.94, 30.14, 26.92, 26.61, and 23.12 percent, respectively, at 10-5 M. The inhibition of tubulin is one of the primary molecular targets of many anticancer agents; hence, the compounds (4a-j) were further subjected to molecular docking studies looking at the tubulin-combretastatin A-4 binding site (PDB ID: 5LYJ) of tubulin. The binding affinities were found to be efficient, ranging from -6.502 to -8.341 kcal/mol, with two major electrostatic interactions observed: H-bond and halogen bond. Ligand 4i had a binding affinity of -8.149 kcal/mol with the tubulin-combretastatin A-4 binding site and displayed a H-bond interaction with the residue Asn258. The ADME and toxicity prediction studies for each compound were carried out using SwissADME and ProTox-II software. None of the compounds' ADME predictions showed that they violated Lipinski's rule of five. All of the compounds were also predicted to have LD50 values between 440 and 500 mg/kg, putting them all in class IV toxicity, according to the toxicity prediction. The current discovery could potentially open up the opportunity for further developments in cancer.
Collapse
Affiliation(s)
- Mohamed Jawed Ahsan
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Ambabari Circle, Jaipur 302039, Rajasthan, India; (M.J.A.); (K.G.)
| | - Krishna Gautam
- Department of Pharmaceutical Chemistry, Maharishi Arvind College of Pharmacy, Ambabari Circle, Jaipur 302039, Rajasthan, India; (M.J.A.); (K.G.)
| | - Amena Ali
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia;
| | | | - Salahuddin
- Department of Pharmaceutical Chemistry, Noida Institute of Engineering and Technology (Pharmacy Institute), Knowledge Park-2, Greater Noida 201306, Uttar Pradesh, India;
| | - Manal A. Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - S. V. V. N. S. M. Lakshmi
- Department of Pharmacognosy, Vishnu Institute of Pharmaceutical Education & Research, Narsapur 502313, Medak Dist., Telangana, India;
| | - Md. Faiyaz Ahsan
- Department of Chemistry, Bihar National College, Patna 800004, Bihar, India;
| |
Collapse
|
21
|
Thammathong J, Chisam KB, Tessmer GE, Womack CB, Sidrak MM, Weissmiller AM, Banerjee S. Fused Imidazopyrazine-Based Tubulin Polymerization Inhibitors Inhibit Neuroblastoma Cell Function. ACS Med Chem Lett 2023; 14:1284-1294. [PMID: 37736192 PMCID: PMC10510670 DOI: 10.1021/acsmedchemlett.3c00298] [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: 07/07/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
Targeting the colchicine binding site on tubulin is a promising approach for cancer treatment to overcome the limitations of current tubulin polymerization inhibitors. New classes of colchicine binding site inhibitors (CBSIs) are continually being uncovered; however, balancing metabolic stability and cellular potency remains an issue that needs to be resolved. Therefore, we designed and synthesized a series of novel fused imidazopyridine and -pyrazine CBSIs and evaluated their cellular activity, metabolic stability, and tubulin-binding properties. Evidence shows that the imidazo[1,2-a]pyrazine series are effective against neuroblastoma cell lines marked by MYCN amplification. Further assessment shows that a combination of an imidazo[1,2-a]pyrazine core with a trimethoxyphenyl ring D results in the highest cellular activity and binding characteristics compared with a dichloromethoxyphenyl or difluoromethoxyphenyl ring D. However, the metabolic stability of compounds with a dichloromethoxyphenyl or difluoromethoxyphenyl ring D is significantly higher than that of those containing a trimethoxyphenyl ring D, suggesting that improved metabolic stability is achieved with a moderate impact on potency.
Collapse
Affiliation(s)
- Joshua Thammathong
- Department
of Chemistry, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | - Kaylee B. Chisam
- Department
of Biology, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | - Garrett E. Tessmer
- Department
of Biology, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | - Carl B. Womack
- Department
of Biology, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | - Mario M. Sidrak
- Department
of Biology, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | - April M. Weissmiller
- Department
of Biology, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| | - Souvik Banerjee
- Department
of Chemistry, Middle Tennessee State University, Murfreesboro, Tennessee 37132, United States
| |
Collapse
|
22
|
Fu DJ, Wang T. Targeting NEDD8-activating enzyme for cancer therapy: developments, clinical trials, challenges and future research directions. J Hematol Oncol 2023; 16:87. [PMID: 37525282 PMCID: PMC10388525 DOI: 10.1186/s13045-023-01485-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/20/2023] [Indexed: 08/02/2023] Open
Abstract
NEDDylation, a post-translational modification through three-step enzymatic cascades, plays crucial roles in the regulation of diverse biological processes. NEDD8-activating enzyme (NAE) as the only activation enzyme in the NEDDylation modification has become an attractive target to develop anticancer drugs. To date, numerous inhibitors or agonists targeting NAE have been developed. Among them, covalent NAE inhibitors such as MLN4924 and TAS4464 currently entered into clinical trials for cancer therapy, particularly for hematological tumors. This review explains the relationships between NEDDylation and cancers, structural characteristics of NAE and multistep mechanisms of NEDD8 activation by NAE. In addition, the potential approaches to discover NAE inhibitors and detailed pharmacological mechanisms of NAE inhibitors in the clinical stage are explored in depth. Importantly, we reasonably investigate the challenges of NAE inhibitors for cancer therapy and possible development directions of NAE-targeting drugs in the future.
Collapse
Affiliation(s)
- Dong-Jun Fu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
23
|
Wang S, Malebari AM, Greene TF, Kandwal S, Fayne D, Nathwani SM, Zisterer DM, Twamley B, O'Boyle NM, Meegan MJ. Antiproliferative and Tubulin-Destabilising Effects of 3-(Prop-1-en-2-yl)azetidin-2-Ones and Related Compounds in MCF-7 and MDA-MB-231 Breast Cancer Cells. Pharmaceuticals (Basel) 2023; 16:1000. [PMID: 37513912 PMCID: PMC10385824 DOI: 10.3390/ph16071000] [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: 05/23/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
A series of novel 3-(prop-1-en-2-yl)azetidin-2-one, 3-allylazetidin-2-one and 3-(buta-1,3-dien-1-yl)azetidin-2-one analogues of combretastatin A-4 (CA-4) were designed and synthesised as colchicine-binding site inhibitors (CBSI) in which the ethylene bridge of CA-4 was replaced with a β-lactam (2-azetidinone) scaffold. These compounds, together with related prodrugs, were evaluated for their antiproliferative activity, cell cycle effects and ability to inhibit tubulin assembly. The compounds demonstrated significant in vitro antiproliferative activities in MCF-7 breast cancer cells, particularly for compounds 9h, 9q, 9r, 10p, 10r and 11h, with IC50 values in the range 10-33 nM. These compounds were also potent in the triple-negative breast cancer (TBNC) cell line MDA-MB-231, with IC50 values in the range 23-33 nM, and were comparable with the activity of CA-4. The compounds inhibited the polymerisation of tubulin in vitro, with significant reduction in tubulin polymerization, and were shown to interact at the colchicine-binding site on tubulin. Flow cytometry demonstrated that compound 9q arrested MCF-7 cells in the G2/M phase and resulted in cellular apoptosis. The antimitotic properties of 9q in MCF-7 human breast cancer cells were also evaluated, and the effect on the organization of microtubules in the cells after treatment with compound 9q was observed using confocal microscopy. The immunofluorescence results confirm that β-lactam 9q is targeting tubulin and resulted in mitotic catastrophe in MCF-7 cells. In silico molecular docking supports the hypothesis that the compounds interact with the colchicine-binding domain of tubulin. Compound 9q is a novel potent microtubule-destabilising agent with potential as a promising lead compound for the development of new antitumour agents.
Collapse
Affiliation(s)
- Shu Wang
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Azizah M Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Thomas F Greene
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Shubhangi Kandwal
- Molecular Design Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Darren Fayne
- Molecular Design Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Seema M Nathwani
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, D02 PN40 Dublin, Ireland
| | - Niamh M O'Boyle
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Mary J Meegan
- School of Pharmacy and Pharmaceutical Sciences, Trinity Biomedical Sciences Institute, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| |
Collapse
|
24
|
Mohamed SM, Abou-Ghadir OMF, El-Mokhtar MA, Aboraia AS, Abdel Aal ABM. Fatty Acid Conjugated Chalcones as Tubulin Polymerization Inhibitors: Design, Synthesis, QSAR, and Apoptotic and Antiproliferative Activity. JOURNAL OF NATURAL PRODUCTS 2023; 86:1150-1158. [PMID: 37098901 DOI: 10.1021/acs.jnatprod.2c00793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Cancer is often associated with an aberrant increase in tubulin and microtubule activity required for cell migration, invasion, and metastasis. A new series of fatty acid conjugated chalcones have been designed as tubulin polymerization inhibitors and anticancer candidates. These conjugates were designed to harness the beneficial physicochemical properties, ease of synthesis, and tubulin inhibitory activity of two classes of natural components. New lipidated chalcones were synthesized from 4-aminoacetophenone via N-acylation followed by condensation with different aromatic aldehydes. All new compounds showed strong inhibition of tubulin polymerization and antiproliferative activity against breast and lung cancer cell lines (MCF-7 and A549) at low or sub-micromolar concentrations. A significant apoptotic effect was shown using a flow cytometry assay that corresponded to cytotoxicity against cancer cell lines, as indicated by a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide assay. Decanoic acid conjugates were more potent than longer lipid analogues, with the most active being more potent than the reference tubulin inhibitor, combretastatin-A4 and the anticancer drug, doxorubicin. None of the newly synthesized compounds caused any detectable cytotoxicity against the normal cell line (Wi-38) or hemolysis of red blood cells below 100 μM. It is unlikely that the new conjugates described would affect normal cells or interrupt with cell membranes due to their lipidic nature. A quantitative structure-activity relationship analysis was performed to determine the influence of 315 descriptors of the physicochemical properties of the new conjugates on their tubulin inhibitory activity. The obtained model revealed a strong correlation between the tubulin inhibitory activity of the investigated compounds and their dipole moment and degree of reactivity.
Collapse
Affiliation(s)
- Samia M Mohamed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Ola M Fahmy Abou-Ghadir
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Mohamed A El-Mokhtar
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Ahmed S Aboraia
- Department of Therapeutic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Abu-Baker M Abdel Aal
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Department of Chemistry, Faculty of Science and Mathematics, Universiti Pendidikan Sultan Idris, Tanjong Malim 35900, Perak, Malaysia
| |
Collapse
|
25
|
Borys F, Tobiasz P, Poterała M, Fabczak H, Krawczyk H, Joachimiak E. Systematic Studies on Anti-Cancer Evaluation of Stilbene and Dibenzo[ b,f]oxepine Derivatives. Molecules 2023; 28:molecules28083558. [PMID: 37110792 PMCID: PMC10146957 DOI: 10.3390/molecules28083558] [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: 03/06/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Cancer is one of the most common causes of human death worldwide; thus, numerous therapies, including chemotherapy, have been and are being continuously developed. In cancer cells, an aberrant mitotic spindle-a microtubule-based structure necessary for the equal splitting of genetic material between daughter cells-leads to genetic instability, one of the hallmarks of cancer. Thus, the building block of microtubules, tubulin, which is a heterodimer formed from α- and β-tubulin proteins, is a useful target in anti-cancer research. The surface of tubulin forms several pockets, i.e., sites that can bind factors that affect microtubules' stability. Colchicine pockets accommodate agents that induce microtubule depolymerization and, in contrast to factors that bind to other tubulin pockets, overcome multi-drug resistance. Therefore, colchicine-pocket-binding agents are of interest as anti-cancer drugs. Among the various colchicine-site-binding compounds, stilbenoids and their derivatives have been extensively studied. Herein, we report systematic studies on the antiproliferative activity of selected stilbenes and oxepine derivatives against two cancer cell lines-HCT116 and MCF-7-and two normal cell lines-HEK293 and HDF-A. The results of molecular modeling, antiproliferative activity, and immunofluorescence analyses revealed that compounds 1a, 1c, 1d, 1i, 2i, 2j, and 3h were the most cytotoxic and acted by interacting with tubulin heterodimers, leading to the disruption of the microtubular cytoskeleton.
Collapse
Affiliation(s)
- Filip Borys
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Piotr Tobiasz
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland
| | - Marcin Poterała
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland
| | - Hanna Fabczak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Hanna Krawczyk
- Department of Organic Chemistry, Faculty of Chemistry, Warsaw University of Technology, 00-664 Warsaw, Poland
| | - Ewa Joachimiak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| |
Collapse
|
26
|
Deng S, Banerjee S, Chen H, Pochampally S, Wang Y, Yun MK, White SW, Parmar K, Meibohm B, Hartman KL, Wu Z, Miller DD, Li W. SB226, an inhibitor of tubulin polymerization, inhibits paclitaxel-resistant melanoma growth and spontaneous metastasis. Cancer Lett 2023; 555:216046. [PMID: 36596380 PMCID: PMC10321023 DOI: 10.1016/j.canlet.2022.216046] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 12/15/2022] [Accepted: 12/24/2022] [Indexed: 01/02/2023]
Abstract
Extensive preclinical studies have shown that colchicine-binding site inhibitors (CBSIs) are promising drug candidates for cancer therapy. Although numerous CBSIs were generated and evaluated, but so far the FDA has not approved any of them due to undesired adverse events or insufficient efficacies. We previously reported two very potent CBSIs, the dihydroquinoxalinone compounds 5 m and 5t. In this study, we further optimized the structures of compounds 5 m and 5t and integrated them to generate a new analog, SB226. X-ray crystal structure studies and a tubulin polymerization assay confirmed that SB226 is a CBSI that could disrupt the microtubule dynamics and interfere with microtubule assembly. Biophysical measurements using surface plasmon resonance (SPR) spectroscopy verified the high binding affinity of SB226 to tubulin dimers. The in vitro studies showed that SB226 possessed sub-nanomolar anti-proliferative activities with an average IC50 of 0.76 nM against a panel of cancer cell lines, some of which are paclitaxel-resistant, including melanoma, breast cancer and prostate cancer cells. SB226 inhibited the colony formation and migration of Taxol-resistant A375/TxR cells, and induced their G2/M phase arrest and apoptosis. Our subsequent in vivo studies confirmed that 4 mg/kg SB226 strongly inhibited the tumor growth of A375/TxR melanoma xenografts in mice and induced necrosis, anti-angiogenesis, and apoptosis in tumors. Moreover, SB226 treatment significantly inhibited spontaneous axillary lymph node, lung, and liver metastases originating from subcutaneous tumors in mice without any obvious toxicity to the animals' major organs, demonstrating the therapeutic potential of SB226 as a novel anticancer agent for cancer therapy.
Collapse
Affiliation(s)
- Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Souvik Banerjee
- Department of Chemistry, Middle Tennessee State University, 1301 E. Main Street, Murfreesboro, TN, 37132, United States; Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main Street, Murfreesboro, TN, 37132, United States
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Satyanarayana Pochampally
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mi-Kyung Yun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, United States
| | - Stephen W White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, United States
| | - Keyur Parmar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Kelli L Hartman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States.
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, 38163, United States.
| |
Collapse
|
27
|
Han HH, Wang HM, Jangili P, Li M, Wu L, Zang Y, Sedgwick AC, Li J, He XP, James TD, Kim JS. The design of small-molecule prodrugs and activatable phototherapeutics for cancer therapy. Chem Soc Rev 2023; 52:879-920. [PMID: 36637396 DOI: 10.1039/d2cs00673a] [Citation(s) in RCA: 77] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Cancer remains as one of the most significant health problems, with approximately 19 million people diagnosed worldwide each year. Chemotherapy is a routinely used method to treat cancer patients. However, current treatment options lack the appropriate selectivity for cancer cells, are prone to resistance mechanisms, and are plagued with dose-limiting toxicities. As such, researchers have devoted their attention to developing prodrug-based strategies that have the potential to overcome these limitations. This tutorial review highlights recently developed prodrug strategies for cancer therapy. Prodrug examples that provide an integrated diagnostic (fluorescent, photoacoustic, and magnetic resonance imaging) response, which are referred to as theranostics, are also discussed. Owing to the non-invasive nature of light (and X-rays), we have discussed external excitation prodrug strategies as well as examples of activatable photosensitizers that enhance the precision of photodynamic therapy/photothermal therapy. Activatable photosensitizers/photothermal agents can be seen as analogous to prodrugs, with their phototherapeutic properties at a specific wavelength activated in the presence of disease-related biomarkers. We discuss each design strategy and illustrate the importance of targeting biomarkers specific to the tumour microenvironment and biomarkers that are known to be overexpressed within cancer cells. Moreover, we discuss the advantages of each approach and highlight their inherent limitations. We hope in doing so, the reader will appreciate the current challenges and available opportunities in the field and inspire subsequent generations to pursue this crucial area of cancer research.
Collapse
Affiliation(s)
- Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Han-Min Wang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China
| | - Paramesh Jangili
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Mingle Li
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| | - Luling Wu
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Yi Zang
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,Lingang laboratory, Shanghai 201203, China
| | - Adam C Sedgwick
- Chemistry Research Laboratory, University of Oxford, Mansfield Road, OX1 3TA, UK.
| | - Jia Li
- State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China. .,University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, P. R. China.,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, P. R. China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, P. R. China. .,The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China.,National Center for Liver Cancer, Shanghai 200438, China
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, BA2 7AY, UK. .,School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
28
|
Zhang M, Zhao M, Wang Y, Chen L, Li G, Liu B, You X, Sun W, Hong L. Synthesis and Biological Evaluation of Phthalideisoquinoline Derivatives. J Org Chem 2023; 88:1720-1729. [PMID: 36651751 DOI: 10.1021/acs.joc.2c02702] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A photo and Cu-mediated radical-radical approach enabling the one-step synthesis of the phthalideisoquinoline skeleton has been reported. Under mild reaction conditions, a series of N-aryl phthalideisoquinolines containing various substituents were synthesized in moderate to good yields. Bioactivity data demonstrated that a new compound 4x can efficiently inhibit the growth of multiple tumor cell lines with enhancements of more than 10-fold by significantly increasing G2/M arrest compared with noscapine.
Collapse
Affiliation(s)
- Ming Zhang
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou510006, China
| | - Man Zhao
- School of Pharmaceutical Sciences, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen518060, China
| | - Ying Wang
- School of Pharmaceutical Sciences, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen518060, China
| | - Lu Chen
- School of Pharmaceutical Sciences, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen518060, China
| | - Guofeng Li
- School of Pharmaceutical Sciences, Shenzhen University Health Science Centre, Shenzhen University, Shenzhen518060, China
| | - Bohan Liu
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou510006, China
| | - Xiaobin You
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou510006, China
| | - Wangsheng Sun
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou730000, China
| | - Liang Hong
- Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou510006, China
| |
Collapse
|
29
|
Man RJ, Lu T, Zheng CC, Li T, Wu MK, Li DD, He XM. Discovery of pyrazole-carbohydrazide with indole moiety as tubulin polymerization inhibitors and anti-tumor candidates. Drug Dev Res 2023; 84:110-120. [PMID: 36433708 DOI: 10.1002/ddr.22016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022]
Abstract
In this work, a series of indole-containing pyrazole-carbohydrazide derivatives A1-A25 were synthesized, and their biological activity on tubulin polymerization inhibition and mitotic catastrophe was evaluated. For introducing indole group to CA-4 pattern, the carbohydrazide linker was used for the first time. As the top hit, A18 suggested notable antiproliferation efficacy and tubulin polymerization inhibitory activity. Inferring comparable antitubulin effect with the positive control Colchicine, A18 indicated obviously lower cyto-toxicity. The cell scratch test showed that A18 could block the cell migration, while the confocal imaging depicted that A18 could induce the mitotic catastrophe via a Colchicine-like approach. The docking simulation visualized the probable binding pattern of A18. With the information in this work, some new hints on modification might be involved in further tubulin-related investigations.
Collapse
Affiliation(s)
- Ruo-Jun Man
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning, China
| | - Tian Lu
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning, China.,Agro-Food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China.,College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Chi-Chong Zheng
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning, China.,Agro-Food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China.,College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Tong Li
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning, China.,Agro-Food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China.,College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Meng-Ke Wu
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi Minzu University, Nanning, China.,Agro-Food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China.,College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Dong-Dong Li
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Xue-Mei He
- Agro-Food Science and Technology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning, China.,Guangxi Key Laboratory of Fruits and Vegetables Storage-processing Technology, Nanning, China
| |
Collapse
|
30
|
Khachatryan H, Olszowy B, Barrero CA, Gordon J, Perez-Leal O. Identification of Inhibitors of Tubulin Polymerization Using a CRISPR-Edited Cell Line with Endogenous Fluorescent Tagging of β-Tubulin and Histone H1. Biomolecules 2023; 13:249. [PMID: 36830618 PMCID: PMC9953358 DOI: 10.3390/biom13020249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/25/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Tubulin is a protein that plays a critical role in maintaining cellular structure and facilitating cell division. Inhibiting tubulin polymerization has been shown to be an effective strategy for inhibiting the proliferation of cancer cells. In the past, identifying compounds that could inhibit tubulin polymerization has required the use of in vitro assays utilizing purified tubulin or immunofluorescence of fixed cells. This study presents a novel approach for identifying tubulin polymerization inhibitors using a CRISPR-edited cell line that expresses fluorescently tagged β-tubulin and a nuclear protein, enabling the visualization of tubulin polymerization dynamics via high-content imaging analysis (HCI). The cells were treated with known tubulin polymerization inhibitors, colchicine, and vincristine, and the resulting phenotypic changes indicative of tubulin polymerization inhibition were confirmed using HCI. Furthermore, a library of 429 kinase inhibitors was screened, resulting in the identification of three compounds (ON-01910, HMN-214, and KX2-391) that inhibit tubulin polymerization. Live cell tracking analysis confirmed that compound treatment leads to rapid tubulin depolymerization. These findings suggest that CRISPR-edited cells with fluorescently tagged endogenous β-tubulin can be utilized to screen large compound libraries containing diverse chemical families for the identification of novel tubulin polymerization inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Oscar Perez-Leal
- Department of Pharmaceutical Sciences, Moulder Center for Drug Discovery, School of Pharmacy, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
31
|
López-López E, Cerda-García-Rojas CM, Medina-Franco JL. Consensus Virtual Screening Protocol Towards the Identification of Small Molecules Interacting with the Colchicine Binding Site of the Tubulin-microtubule System. Mol Inform 2023; 42:e2200166. [PMID: 36175374 PMCID: PMC10078098 DOI: 10.1002/minf.202200166] [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: 07/18/2022] [Accepted: 09/29/2022] [Indexed: 01/12/2023]
Abstract
Modification of the tubulin-microtubule (Tub-Mts) system has generated effective strategies for developing different treatments for cancer. A huge amount of clinical data about inhibitors of the tubulin-microtubule system have supported and validated the studies on this pharmacological target. However, many tubulin-microtubule inhibitors have been developed from representative and common scaffolds that cover a small region of the chemical space with limited structural innovation. The main goal of this study is to develop the first consensus virtual screening protocol for natural products (ligand- and structure-based drug design methods) tuned for the identification of new potential inhibitors of the Tub-Mts system. A combined strategy that involves molecular similarity, molecular docking, pharmacophore modeling, and in silico ADMET prediction has been employed to prioritize the selections of potential inhibitors of the Tub-Mts system. Five compounds were selected and further studied using molecular dynamics and binding energy predictions to characterize their possible binding mechanisms. Their structures correspond to 5-[2-(4-hydroxy-3-methoxyphenyl) ethyl]-2,3-dimethoxyphenol (1), 9,10-dihydro-3,4-dimethoxy-2,7-phenanthrenediol (2), 2-(3,4-dimethoxyphenyl)-5,7-dihydroxy-6-methoxy-4H-1-benzopyran-4-one (3), 13,14-epoxyparvifoline-4',5',6'-trimethoxybenzoate (4), and phenylmethyl 6-hydroxy-2,3-dimethoxybenzoate (5). Compounds 1-3 have been associated with literature reports that confirm their activity against several cancer cell lines, thus supporting the utility of this protocol.
Collapse
Affiliation(s)
- Edgar López-López
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico.,Departamento de Química y Programa de Posgrado en Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, 07000, Mexico
| | - Carlos M Cerda-García-Rojas
- Departamento de Química y Programa de Posgrado en Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, 07000, Mexico
| | - José L Medina-Franco
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Mexico City, 04510, Mexico
| |
Collapse
|
32
|
Guerraoui A, Goudjil M, Direm A, Guerraoui A, Şengün İY, Parlak C, Djedouani A, Chelazzi L, Monti F, Lunedei E, Boumaza A. A rhodanine derivative as a potential antibacterial and anticancer agent: crystal structure, spectral characterization, DFT calculations, Hirshfeld surface analysis, in silico molecular docking and ADMET studies. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
33
|
Cao X, Li R, Wang H, Guo C, Wang S, Chen X, Zhao R. Novel indole–chalcone platinum(IV) complexes as tubulin polymerization inhibitors to overcome oxaliplatin resistance in colorectal cancer. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
34
|
Schlesinger T, Stockfleth E, Grada A, Berman B. Tirbanibulin for Actinic Keratosis: Insights into the Mechanism of Action. Clin Cosmet Investig Dermatol 2022; 15:2495-2506. [PMID: 36415541 PMCID: PMC9675993 DOI: 10.2147/ccid.s374122] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/19/2022] [Indexed: 08/13/2023]
Abstract
Actinic keratosis (AK) is a common pre-neoplastic skin lesion constituted by uncontrolled proliferation of atypical keratinocytes that may evolve to squamous cell carcinoma. With global prevalence increasing, AK is expected to be the most common carcinoma of the skin. Tirbanibulin is a reversible tubulin polymerization inhibitor with potent anti-proliferative and anti-tumoral effects. In-vivo and in-vitro studies have shown that tirbanibulin significantly inhibits cell proliferation, tumor growth and downregulates Src signaling with no overt toxicity. Early phase and Phase III trials have shown high lesion clearance, compliance, and few side effects of once daily tirbanibulin treatment. This review discusses tirbanibulin anti-cancer activity, focusing on tubulin polymerization and Src signaling inhibitory effects, highlighting relevant literature and novel preclinical results from the ATNXUS-KX01-001 study. Furthermore, we address the relevant findings obtained in recent clinical trials to evaluate the safety, efficacy, pharmacokinetics, clearance efficacy, and side effects of the 1% tirbanibulin ointment applied once daily. In summary, we highlight preclinical and clinical evidence on the use of tirbanibulin as an effective and safe treatment option for AK.
Collapse
Affiliation(s)
| | - Eggert Stockfleth
- Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum, Bochum, Germany
| | - Ayman Grada
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Brian Berman
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
35
|
Constantinescu T, Mihis AG. Two Important Anticancer Mechanisms of Natural and Synthetic Chalcones. Int J Mol Sci 2022; 23:11595. [PMID: 36232899 PMCID: PMC9570335 DOI: 10.3390/ijms231911595] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
ATP-binding cassette subfamily G and tubulin pharmacological mechanisms decrease the effectiveness of anticancer drugs by modulating drug absorption and by creating tubulin assembly through polymerization. A series of natural and synthetic chalcones have been reported to have very good anticancer activity, with a half-maximal inhibitory concentration lower than 1 µM. By modulation, it is observed in case of the first mechanism that methoxy substituents on the aromatic cycle of acetophenone residue and substitution of phenyl nucleus by a heterocycle and by methoxy or hydroxyl groups have a positive impact. To inhibit tubulin, compounds bind to colchicine binding site. Presence of methoxy groups, amino groups or heterocyclic substituents increase activity.
Collapse
Affiliation(s)
- Teodora Constantinescu
- Department of Chemistry, Faculty of Pharmacy, Iuliu Hatieganu University, 400012 Cluj-Napoca, Romania
| | - Alin Grig Mihis
- Advanced Materials and Applied Technologies Laboratory, Institute of Research-Development-Innovation in Applied Natural Sciences, “Babes-Bolyai” University, Fantanele Str. 30, 400294 Cluj-Napoca, Romania
| |
Collapse
|
36
|
Peng J, Zeng Y, Hu X, Huang S, Gao X, Tian D, Tian S, Qiu L, Liu J, Dong R, Zhan W, Qin C, Guang B, Yang T. KC-180-2 Exerts Anti-SCLC Effects via Dual Inhibition of Tubulin Polymerization and Src Signaling. ACS OMEGA 2022; 7:32164-32175. [PMID: 36120000 PMCID: PMC9476193 DOI: 10.1021/acsomega.2c03408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
In this study, a series of N-benzyl-2-(5-phenylpyridin-2-yl) acetamide-based derivatives were successfully designed and synthesized as anti-cancer agents. KC-180-2 was screened as a potentially leading compound with dual mechanisms of action: Src signaling and tubulin polymerization inhibition. It efficiently suppressed the proliferation of five cancer cell lines (MDA-MB-231, H446, SKOV-3, HepG2, and HT29), with IC50 values ranging from 5 to 188 nM, especially small-cell lung cancer (SCLC) cells (IC50, 5 nM). Correspondingly, it exerted a significant therapeutic effect on the H446 small-cell lung cancer xenograft model, significantly reducing the volume of tumors without obvious toxicity. Mechanistically, this compound significantly inhibited the polymerization of purified tubulin in vitro, inducing G2/M cell cycle arrest and binding to the kinase catalytic domain of the Src protein, which reduced the phosphorylation of Src. Thus, KC-180-2 is a potential lead compound for the further development of a new anti-tumor drug against SCLC.
Collapse
Affiliation(s)
- Jian Peng
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Yisheng Zeng
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Xiaojun Hu
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Sheng Huang
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Xiaofang Gao
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Dong Tian
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Shuting Tian
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Lan Qiu
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Jin Liu
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| | - Renhan Dong
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Wei Zhan
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Chuanjun Qin
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Bing Guang
- Chengdu
Biobel Biotechnology Co., Ltd., No. 88, Keyuan South Road, New and High-Tech Zone, Chengdu, Sichuan Province 610094, China
| | - Tai Yang
- School
of Pharmacy, Chengdu Medical College, No. 783, Xindu Avenue, Xindu District, Chengdu, Sichuan Province 610500, China
| |
Collapse
|
37
|
Banerjee S, Lee S, Norman DD, Tigyi GJ. Designing Dual Inhibitors of Autotaxin-LPAR GPCR Axis. Molecules 2022; 27:5487. [PMID: 36080255 PMCID: PMC9458164 DOI: 10.3390/molecules27175487] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
The ATX-LPA-LPAR1 signaling pathway plays a universal role in stimulating diverse cellular responses, including cell proliferation, migration, survival, and invasion in almost every cell type. The ATX-LPAR1 axis is linked to several metabolic and inflammatory diseases including cancer, fibrosis, and rheumatoid arthritis. Numerous selective ATX or LPAR1 inhibitors have been developed and so far, their clinical efficacy has only been evaluated in idiopathic pulmonary fibrosis. None of the ATX and LPAR1 inhibitors have advanced to clinical trials for cancer and rheumatoid arthritis. Nonetheless, several research groups, including ours, have shown considerable benefit of simultaneous ATX and LPAR1 inhibition through combination therapy. Recent research suggests that dual-targeting therapies are superior to combination therapies that use two selective inhibitors. However, limited reports are available on ATX-LPAR1 dual inhibitors, potentially due to co-expression of multiple different LPARs with close structural similarities at the same target. In this review, we discuss rational design and future directions of dual ATX-LPAR1 inhibitors.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Chemistry, Middle Tennessee State University, 1301 E. Main Street, Murfreesboro, TN 37132, USA
- Molecular Biosciences Program, Middle Tennessee State University, 1301 E. Main Street, Murfreesboro, TN 37132, USA
| | - Suechin Lee
- Department of Physiology, University of Tennessee Health Science Center Memphis, 3 N. Dunlap Street, Memphis, TN 38163, USA
| | - Derek D. Norman
- Department of Physiology, University of Tennessee Health Science Center Memphis, 3 N. Dunlap Street, Memphis, TN 38163, USA
| | - Gabor J. Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, 3 N. Dunlap Street, Memphis, TN 38163, USA
| |
Collapse
|
38
|
Malebari AM, Duffy Morales G, Twamley B, Fayne D, Khan MF, McLoughlin EC, O’Boyle NM, Zisterer DM, Meegan MJ. Synthesis, Characterisation and Mechanism of Action of Anticancer 3-Fluoroazetidin-2-ones. Pharmaceuticals (Basel) 2022; 15:1044. [PMID: 36145265 PMCID: PMC9501633 DOI: 10.3390/ph15091044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
The stilbene combretastatin A-4 (CA-4) is a potent microtubule-disrupting agent interacting at the colchicine-binding site of tubulin. In the present work, the synthesis, characterisation and mechanism of action of a series of 3-fluoro and 3,3-difluoro substituted β-lactams as analogues of the tubulin-targeting agent CA-4 are described. The synthesis was achieved by a convenient microwave-assisted Reformatsky reaction and is the first report of 3-fluoro and 3,3-difluoro β-lactams as CA-4 analogues. The β-lactam compounds 3-fluoro-4-(3-hydroxy-4-methoxyphenyl)-1-(3,4,5-trimethoxy phenyl)azetidin-2-one 32 and 3-fluoro-4-(3-fluoro-4-methoxyphenyl)-1-(3,4,5-trimethoxyphenyl)azetidin-2-one) 33 exhibited potent activity in MCF-7 human breast cancer cells with IC50 values of 0.075 µM and 0.095 µM, respectively, and demonstrated low toxicity in non-cancerous cells. Compound 32 also demonstrated significant antiproliferative activity at nanomolar concentrations in the triple-negative breast cancer cell line Hs578T (IC50 0.033 μM), together with potency in the invasive isogenic subclone Hs578Ts(i)8 (IC50 = 0.065 μM), while 33 was also effective in MDA-MB-231 cells (IC50 0.620 μM). Mechanistic studies demonstrated that 33 inhibited tubulin polymerisation, induced apoptosis in MCF-7 cells, and induced a downregulation in the expression of anti-apoptotic Bcl2 and survivin with corresponding upregulation in the expression of pro-apoptotic Bax. In silico studies indicated the interaction of the compounds with the colchicine-binding site, demonstrating the potential for further developing novel cancer therapeutics as microtubule-targeting agents.
Collapse
Affiliation(s)
- Azizah M. Malebari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Trinity Biomedical Sciences Institute, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Gabriela Duffy Morales
- Trinity Biomedical Sciences Institute, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Brendan Twamley
- School of Chemistry, Trinity College Dublin, Dublin 2, D02 PN40 Dublin, Ireland
| | - Darren Fayne
- Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Mohemmed Faraz Khan
- Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Eavan C. McLoughlin
- Trinity Biomedical Sciences Institute, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Niamh M. O’Boyle
- Trinity Biomedical Sciences Institute, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Daniela M. Zisterer
- Trinity Biomedical Sciences Institute, School of Biochemistry and Immunology, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| | - Mary J. Meegan
- Trinity Biomedical Sciences Institute, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, 152-160 Pearse Street, Dublin 2, D02 R590 Dublin, Ireland
| |
Collapse
|
39
|
Romagnoli R, Oliva P, Prencipe F, Manfredini S, Budassi F, Brancale A, Ferla S, Hamel E, Corallo D, Aveic S, Manfreda L, Mariotto E, Bortolozzi R, Viola G. Design, Synthesis and Biological Investigation of 2-Anilino Triazolopyrimidines as Tubulin Polymerization Inhibitors with Anticancer Activities. Pharmaceuticals (Basel) 2022; 15:1031. [PMID: 36015179 PMCID: PMC9415608 DOI: 10.3390/ph15081031] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 11/16/2022] Open
Abstract
A further investigation aiming to generate new potential antitumor agents led us to synthesize a new series of twenty-two compounds characterized by the presence of the 7-(3',4',5'-trimethoxyphenyl)-[1,2,4]triazolo[1,5-a]pyrimidine pharmacophore modified at its 2-position. Among the synthesized compounds, three were significantly more active than the others. These bore the substituents p-toluidino (3d), p-ethylanilino (3h) and 3',4'-dimethylanilino (3f), and these compounds had IC50 values of 30-43, 160-240 and 67-160 nM, respectively, on HeLa, A549 and HT-29 cancer cells. The p-toluidino derivative 3d was the most potent inhibitor of tubulin polymerization (IC50: 0.45 µM) and strongly inhibited the binding of colchicine to tubulin (72% inhibition), with antiproliferative activity superior to CA-4 against A549 and HeLa cancer cell lines. In vitro investigation showed that compound 3d was able to block treated cells in the G2/M phase of the cell cycle and to induce apoptosis following the intrinsic pathway, further confirmed by mitochondrial depolarization and caspase-9 activation. In vivo experiments conducted on the zebrafish model showed good activity of 3d in reducing the mass of a HeLa cell xenograft. These effects occurred at nontoxic concentrations to the animal, indicating that 3d merits further developmental studies.
Collapse
Affiliation(s)
- Romeo Romagnoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paola Oliva
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Filippo Prencipe
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127 Trieste, Italy
| | - Stefano Manfredini
- Department of Life Sciences and Biotechnology, University of Ferrara, 44121 Ferrara, Italy
| | - Federica Budassi
- Medicinal Chemistry Department, Integrated Drug Discovery, Aptuit-An Evotec Company, 37135 Verona, Italy
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff CF10 3NB, UK
| | - Salvatore Ferla
- Faculty of Medicine, Health and Life Science, Swansea University Medical School, Grove Building, Swansea University, Swansea SA2 8PP, UK
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Lorenzo Manfreda
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Elena Mariotto
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Roberta Bortolozzi
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| | - Giampietro Viola
- Department of Woman’s and Child’s Health, Hemato-Oncology Lab, University of Padova, 35131 Padova, Italy
- Laboratory of Experimental Pharmacology, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35128 Padova, Italy
| |
Collapse
|
40
|
Jackson TR, Vuorinen A, Josa-Culleré L, Madden KS, Conole D, Cogswell TJ, Wilkinson IV, Kettyle LM, Zhang D, O’Mahony A, Gracias D, McCall L, Westwood R, Terstappen GC, Davies SG, Tate EW, Wynne GM, Vyas P, Russell AJ, Milne TA. A tubulin binding molecule drives differentiation of acute myeloid leukemia cells. iScience 2022; 25:104787. [PMID: 35992086 PMCID: PMC9385704 DOI: 10.1016/j.isci.2022.104787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/31/2022] [Accepted: 07/13/2022] [Indexed: 12/01/2022] Open
Abstract
Despite much progress in developing better drugs, many patients with acute myeloid leukemia (AML) still die within a year of diagnosis. This is partly because it is difficult to identify therapeutic targets that are effective across multiple AML subtypes. One common factor across AML subtypes is the presence of a block in differentiation. Overcoming this block should allow for the identification of therapies that are not dependent on a specific mutation for their efficacy. Here, we used a phenotypic screen to identify compounds that stimulate differentiation in genetically diverse AML cell lines. Lead compounds were shown to decrease tumor burden and to increase survival in vivo. Using multiple complementary target deconvolution approaches, these compounds were revealed to be anti-mitotic tubulin disruptors that cause differentiation by inducing a G2-M mitotic arrest. Together, these results reveal a function for tubulin disruptors in causing differentiation of AML cells.
Collapse
Affiliation(s)
- Thomas R. Jackson
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Aini Vuorinen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Laia Josa-Culleré
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Katrina S. Madden
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Daniel Conole
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Thomas J. Cogswell
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Isabel V.L. Wilkinson
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Laura M. Kettyle
- Axis Bioservices, 189 Castleroe Rd, Coleraine, Co. Londonderry BT51 3RP, Northern Ireland
| | - Douzi Zhang
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Alison O’Mahony
- Eurofins Discovery Phenotypic Services, St. Charles, MO 63304 and Burlingame, CA 94010, USA
- Discovery Platform at Recursion, 41 S Rio Grande Street, Salt Lake City, UT 84101, USA
| | - Deanne Gracias
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Lorna McCall
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Robert Westwood
- Oxstem Ltd, Midland House West Way, Botley, Oxford OX2 0PH, UK
| | | | - Stephen G. Davies
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
| | - Edward W. Tate
- Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, 82 Wood Lane, London W12 0BZ, UK
| | - Graham M. Wynne
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Oxstem Ltd, Midland House West Way, Botley, Oxford OX2 0PH, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Angela J. Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, UK
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Thomas A. Milne
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
41
|
Combination of microtubule targeting agents with other antineoplastics for cancer treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188777. [PMID: 35963551 DOI: 10.1016/j.bbcan.2022.188777] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/04/2022] [Accepted: 08/07/2022] [Indexed: 11/22/2022]
Abstract
Microtubule targeting agents (MTAs) have attracted extensive attention for cancer treatment. However, their clinical efficacies are limited by intolerable toxicities, inadequate efficacy and acquired multidrug resistance. The combination of MTAs with other antineoplastics has become an efficient strategy to lower the toxicities, overcome resistance and improve the efficacies for cancer treatment. In this article, we review the combinations of MTAs with some other anticancer drugs, such as cytotoxic agents, kinases inhibitors, histone deacetylase inhibitors, immune checkpoints inhibitors, to overcome these obstacles. We strongly believe that this review will provide helpful information for combination therapy based on MTAs.
Collapse
|
42
|
A novel aromatic amide derivative SY-65 co-targeted tubulin and histone deacetylase 1 with potent anticancer activity in vitro and in vivo. Biochem Pharmacol 2022; 201:115070. [DOI: 10.1016/j.bcp.2022.115070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/27/2022]
|
43
|
de la Roche NM, Mühlethaler T, Di Martino RMC, Ortega JA, Gioia D, Roy B, Prota AE, Steinmetz MO, Cavalli A. Novel fragment-derived colchicine-site binders as microtubule-destabilizing agents. Eur J Med Chem 2022; 241:114614. [DOI: 10.1016/j.ejmech.2022.114614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/16/2022]
|
44
|
Karthika C, Sureshkumar R, Zehravi M, Akter R, Ali F, Ramproshad S, Mondal B, Tagde P, Ahmed Z, Khan FS, Rahman MH, Cavalu S. Multidrug Resistance of Cancer Cells and the Vital Role of P-Glycoprotein. Life (Basel) 2022; 12:897. [PMID: 35743927 PMCID: PMC9227591 DOI: 10.3390/life12060897] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
P-glycoprotein (P-gp) is a major factor in the multidrug resistance phenotype in cancer cells. P-gp is a protein that regulates the ATP-dependent efflux of a wide range of anticancer medicines and confers resistance. Due to its wide specificity, several attempts have been made to block the action of P-gp to restore the efficacy of anticancer drugs. The major goal has been to create molecules that either compete with anticancer medicines for transport or function as a direct P-gp inhibitor. Despite significant in vitro success, there are presently no drugs available in the clinic that can "block" P-gp-mediated resistance. Toxicity, unfavourable pharmacological interactions, and a variety of pharmacokinetic difficulties might all be the reason for the failure. On the other hand, P-gp has a significant effect in the body. It protects the vital organs from the entry of foreign bodies and other toxic chemicals. Hence, the inhibitors of P-gp should not hinder its action in the normal cells. To develop an effective inhibitor of P-gp, thorough background knowledge is needed in this field. The main aim of this review article was to set forth the merits and demerits of the action of P-gp on cancer cells as well as on normal cells. The influence of P-gp on cancer drug delivery and the contribution of P-gp to activating drug resistance were also mentioned.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Raman Sureshkumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University Alkharj, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Faraat Ali
- Department of Licensing and Enforcement, Laboratory Services, Botswana Medicines Regulatory Authority (BoMRA), Gaborone 999106, Botswana;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Priti Tagde
- Amity Institute of Pharmacy, Amity University, Noida 201303, Uttar Pradesh, India;
| | - Zubair Ahmed
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia;
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Mahala Campus, Community College, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Abha 61413, Saudi Arabia;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
45
|
Design, Synthesis, In Vitro Biological Activity Evaluation and Stabilized Nanostructured Lipid Carrier Formulation of Newly Synthesized Schiff Bases-Based TMP Moieties. Pharmaceuticals (Basel) 2022; 15:ph15060679. [PMID: 35745599 PMCID: PMC9230623 DOI: 10.3390/ph15060679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/03/2022] Open
Abstract
A series of novel Schiff bases-based TMP moieties have been designed and synthesized as potential anticancer agents. The target Schiff bases were screened for their cytotoxic activity against the MDA-MB-231 breast cancer cell line. Most of the tested molecules revealed good cytotoxic activity, especially compounds 4h, 4j and 5d. Being the most potent, compound 4h showed good tubulin polymerization inhibition activity as revealed by immunofluorescence analysis and ELISA assay. Additionally, compound 4h was screened for cell cycle disturbance and apoptosis induction. Pre-G1 apoptosis and cell growth halt at the G2/M phase were discovered to be caused by it. Moreover, compound 4h induced apoptosis via p53 and Bax activation, as well as reduced the level of Bcl-2. Additionally, the most potent compound 4h was lodged on nanostructured lipid carriers (NLCs). 23 full factorial design was involved to govern the influence of the fabrication variables on the in vitro characters of the casted NLCs. F3 was picked as the optimum formula exhibiting dominant desirability value 0.805, EE% 95.6 ± 2.4, PS 222.4 ±18.7, PDI 0.23 ± 0.05 and ZP −39.2 ± 3.9 Mv. Furthermore, F3 affirmed improved solubility and release over the drug suspension. In the comparative cytotoxic activity, F3 was capable of diminishing the IC50 by around 2.15 times for pure 4h, while nearly close to the IC50 of the reference drug. Thus, NLCs could be a potential platform for boosted antitumor activity.
Collapse
|
46
|
Swaminathan S, Haribabu J, Balakrishnan N, Vasanthakumar P, Karvembu R. Piano stool Ru(II)-arene complexes having three monodentate legs: A comprehensive review on their development as anticancer therapeutics over the past decade. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
47
|
Boichuk S, Syuzov K, Bikinieva F, Galembikova A, Zykova S, Gankova K, Igidov S, Igidov N. Computational-Based Discovery of the Anti-Cancer Activities of Pyrrole-Based Compounds Targeting the Colchicine-Binding Site of Tubulin. Molecules 2022; 27:2873. [PMID: 35566235 PMCID: PMC9101527 DOI: 10.3390/molecules27092873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 12/10/2022] Open
Abstract
Despite the tubulin-binding agents (TBAs) that are widely used in the clinic for cancer therapy, tumor resistance to TBAs (both inherited and acquired) significantly impairs their effectiveness, thereby decreasing overall survival (OS) and progression-free survival (PFS) rates, especially for the patients with metastatic, recurrent, and unresectable forms of the disease. Therefore, the development of novel effective drugs interfering with the microtubules' dynamic state remains a big challenge in current oncology. We report here about the novel ethyl 2-amino-1-(furan-2-carboxamido)-5-(2-aryl/tert-butyl-2-oxoethylidene)-4-oxo-4,5-dihydro-1H-pyrrole-3-carboxylates (EAPCs) exhibiting potent anti-cancer activities against the breast and lung cancer cell lines in vitro. This was due to their ability to inhibit tubulin polymerization and induce cell cycle arrest in M-phase. As an outcome, the EAPC-treated cancer cells exhibited a significant increase in apoptosis, which was evidenced by the expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin-V-positive cells. By using the in silico molecular modeling methods (e.g., induced-fit docking, binding metadynamics, and unbiased molecular dynamics), we found that EAPC-67 and -70 preferentially bind to the colchicine-binding site of tubulin. Lastly, we have shown that the EAPCs indicated above and colchicine utilizes a similar molecular mechanism to inhibit tubulin polymerization via targeting the T7 loop in the β-chain of tubulin, thereby preventing the conformational changes in the tubulin dimers required for their polymerization. Collectively, we identified the novel and potent TBAs that bind to the colchicine-binding site and disrupt the microtubule network. As a result of these events, the compounds induced a robust cell cycle arrest in M-phase and exhibited potent pro-apoptotic activities against the epithelial cancer cell lines in vitro.
Collapse
Affiliation(s)
- Sergei Boichuk
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
- Department of Radiotherapy and Radiology, Russian Medical Academy of Continuous Professional Education, 125993 Moscow, Russia
- Biologically Active Terpenoids Laboratory, Kazan Federal University, 18 Kremlyovskaya St., 420008 Kazan, Russia
| | - Kirill Syuzov
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Firuza Bikinieva
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Aigul Galembikova
- Department of Pathology, Kazan State Medical University, 420012 Kazan, Russia; (K.S.); (F.B.); (A.G.)
| | - Svetlana Zykova
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Ksenia Gankova
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Sergei Igidov
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| | - Nazim Igidov
- Department of Pharmacology, Perm State Academy of Pharmacy, 614990 Perm, Russia; (S.Z.); (K.G.); (S.I.); (N.I.)
| |
Collapse
|
48
|
Computational-Based Discovery of the Anti-Cancer Activities of Pyrrole-Based Compounds Targeting the Colchicine-Binding Site of Tubulin. Molecules 2022. [PMID: 35566235 DOI: 10.3390/molecules27092873.(] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Despite the tubulin-binding agents (TBAs) that are widely used in the clinic for cancer therapy, tumor resistance to TBAs (both inherited and acquired) significantly impairs their effectiveness, thereby decreasing overall survival (OS) and progression-free survival (PFS) rates, especially for the patients with metastatic, recurrent, and unresectable forms of the disease. Therefore, the development of novel effective drugs interfering with the microtubules' dynamic state remains a big challenge in current oncology. We report here about the novel ethyl 2-amino-1-(furan-2-carboxamido)-5-(2-aryl/tert-butyl-2-oxoethylidene)-4-oxo-4,5-dihydro-1H-pyrrole-3-carboxylates (EAPCs) exhibiting potent anti-cancer activities against the breast and lung cancer cell lines in vitro. This was due to their ability to inhibit tubulin polymerization and induce cell cycle arrest in M-phase. As an outcome, the EAPC-treated cancer cells exhibited a significant increase in apoptosis, which was evidenced by the expression of cleaved forms of PARP, caspase-3, and increased numbers of Annexin-V-positive cells. By using the in silico molecular modeling methods (e.g., induced-fit docking, binding metadynamics, and unbiased molecular dynamics), we found that EAPC-67 and -70 preferentially bind to the colchicine-binding site of tubulin. Lastly, we have shown that the EAPCs indicated above and colchicine utilizes a similar molecular mechanism to inhibit tubulin polymerization via targeting the T7 loop in the β-chain of tubulin, thereby preventing the conformational changes in the tubulin dimers required for their polymerization. Collectively, we identified the novel and potent TBAs that bind to the colchicine-binding site and disrupt the microtubule network. As a result of these events, the compounds induced a robust cell cycle arrest in M-phase and exhibited potent pro-apoptotic activities against the epithelial cancer cell lines in vitro.
Collapse
|
49
|
Gouw AM, Kumar V, Resendez A, Alvina FB, Liu NS, Margulis K, Tong L, Zare RN, Malhotra SV, Felsher DW. Azapodophyllotoxin Causes Lymphoma and Kidney Cancer Regression by Disrupting Tubulin and Monoglycerols. ACS Med Chem Lett 2022; 13:615-622. [PMID: 35450373 PMCID: PMC9014495 DOI: 10.1021/acsmedchemlett.1c00673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/18/2022] [Indexed: 11/28/2022] Open
Abstract
A natural compound screen identified several anticancer compounds, among which azapodophyllotoxin (AZP) was found to be the most potent. AZP caused decreased viability of both mouse and human lymphoma and renal cell cancer (RCC) tumor-derived cell lines. Novel AZP derivatives were synthesized and screened identifying compound NSC750212 to inhibit the growth of both lymphoma and RCC both in vitro and in vivo. A nanoimmunoassay was used to assess the NSC750212 mode of action in vivo. On the basis of the structure of AZP and its mode of action, AZP disrupts tubulin polymerization. Through desorption electrospray ionization mass spectrometry imaging, NSC750212 was found to inhibit lipid metabolism. NSC750212 suppresses monoglycerol metabolism depleting lipids and thereby inhibits tumor growth. The dual mode of tubulin polymerization disruption and monoglycerol metabolism inhibition makes NSC750212 a potent small molecule against lymphoma and RCC.
Collapse
Affiliation(s)
- Arvin M. Gouw
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Vineet Kumar
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Angel Resendez
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, United States
| | - Fidelia B. Alvina
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Natalie S. Liu
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Katherine Margulis
- Department of Chemistry, School of Humanities and Sciences, Stanford University, Stanford, California 94305, United States
| | - Ling Tong
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Richard N. Zare
- Department of Chemistry, School of Humanities and Sciences, Stanford University, Stanford, California 94305, United States
| | - Sanjay V. Malhotra
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Cell, Developmental and Cancer Biology, Oregon health and Science University, Portland, Oregon 97201, United States
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon health and Science University, Portland, Oregon 97201, United States
| | - Dean W. Felsher
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
50
|
Peng Y, Shi Z, Liang Y, Ding K, Wang Y. Targeting the tumor microenvironment by an enzyme-responsive prodrug of tubulin destabilizer for triple-negative breast cancer therapy with high safety. Eur J Med Chem 2022; 236:114344. [PMID: 35405397 DOI: 10.1016/j.ejmech.2022.114344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 11/04/2022]
Abstract
In response to the long-term potential toxicity concerns of tubulin destabilizer, an enzyme-responsive prodrug therapy for triple-negative breast cancer was developed based on the different β-glucuronidase levels between tumor and normal tissues in this study. All the prodrugs synthesized herein showed remarkable stability in phosphate buffer and bovine serum solution, among which 17a was found to be more susceptible to enzymatic cleavage. 17a exhibited excellent selectivity between the in vitro antiproliferative activities against β-glucuronidase-pretreated and -untreated cancer cells (IC50 (+Enz) = 8.9-15.7 nM, IC50 (-Enz) > 50 μM), along with favorable liver microsomal metabolic stability and improved aqueous solubility. Furthermore, as a candidate prodrug 17a showed potent antitumor efficacy in MDA-MB-231 xenograft mouse model without causing perceptible injury to organs. Importantly, 17a exhibited superior safety profiles with higher LD50 value and no perceivable cardiotoxicity, which was a major dose-limiting adverse effect for the parent compound 1. These salient toxicity-reduced effects of 17a would merit further in-depth assessment of this compound for preclinical therapeutic usages.
Collapse
Affiliation(s)
- Yingyuan Peng
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhixian Shi
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuru Liang
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Kuiling Ding
- State Key Laboratory of Organometallic Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Yang Wang
- School of Pharmacy, Fudan University, Shanghai, 201203, China; School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, 453007, China; Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|