1
|
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
|
2
|
Dong H, Lu L, Song X, Li Y, Zhou J, Xu Y, Zhang Y, Qi J, Liang T, Wang J. Design, synthesis and biological evaluation of tetrahydroquinoxaline sulfonamide derivatives as colchicine binding site inhibitors. RSC Adv 2023; 13:30202-30216. [PMID: 37849704 PMCID: PMC10577396 DOI: 10.1039/d3ra05720h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/09/2023] [Indexed: 10/19/2023] Open
Abstract
Colchicine binding site inhibitors (CBSIs) are potential microtubule targeting agents (MTAs), which can overcome multidrug resistance, improve aqueous solubility and reduce toxicity faced by most MTAs. Novel tetrahydroquinoxaline sulfonamide derivatives were designed, synthesized and evaluated for their antiproliferative activities. The MTT assay results demonstrated that some derivatives exhibited moderate to strong inhibitory activities against HT-29 cell line. Among them, compound I-7 was the most active compound. Moreover, I-7 inhibited tubulin polymerization, disturbed microtubule network, disrupted the formation of mitotic spindle and arrested cell cycle at G2/M phase. However, I-7 didn't induce cell apoptosis. Furthermore, the prediction of ADME demonstrated that I-7 showed favorable physiochemical and pharmacokinetic properties. And the detailed molecular docking confirmed I-7 targeted the site of colchicine through hydrogen and hydrophobic interactions.
Collapse
Affiliation(s)
- Haiyang Dong
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Lu Lu
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Xueting Song
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Youkang Li
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jinguang Zhou
- Huaihe Hospital of Henan University Kaifeng 475004 Henan China
| | - Yungen Xu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University Nanjing 211198 China
| | - Yahong Zhang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jianguo Qi
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Tingting Liang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| | - Jianhong Wang
- Key Laboratory of Natural Medicine and Immune-Engineering of Henan Province, Henan University Jinming Campus Kaifeng 475004 Henan China
| |
Collapse
|
3
|
Wang C, Shi L, Yang S, Chang J, Liu W, Zeng J, Meng J, Zhang R, Xing D. Research progress on antitumor activity of XRP44X and analogues as microtubule targeting agents. Front Chem 2023; 11:1096666. [PMID: 36936533 PMCID: PMC10014799 DOI: 10.3389/fchem.2023.1096666] [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: 11/12/2022] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Cancer threatens human health and life. Therefore, it is particularly important to develop safe and effective antitumor drugs. Microtubules, the main component of cytoskeleton, play an important role in maintaining cell morphology, mitosis, and signal transduction, which are one of important targets of antitumor drug research and development. Colchicine binding site inhibitors have dual effects of inhibiting proliferation and destroying blood vessels. In recent years, a series of inhibitors targeting this target have been studied and some progress has been made. XRP44X has a novel structure and overcomes some disadvantages of traditional inhibitors. It is also a multifunctional molecule that regulates not only the function of tubulin but also a variety of biological pathways. Therefore, the structure, synthesis, structure-activity relationship, and biological activity of XRP44X analogues reported in recent years were summarized in this paper, to provide a useful reference for the rational design of efficient colchicine binding site inhibitors.
Collapse
Affiliation(s)
- Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- *Correspondence: Chao Wang, ; Dongming Xing,
| | - Lingyu Shi
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Shanbo Yang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jing Chang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Wenjing Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jun Zeng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jingsen Meng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Renshuai Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
- *Correspondence: Chao Wang, ; Dongming Xing,
| |
Collapse
|
4
|
An update on the recent advances and discovery of novel tubulin colchicine binding inhibitors. Future Med Chem 2023; 15:73-95. [PMID: 36756851 DOI: 10.4155/fmc-2022-0212] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
Abstract
Microtubules, formed by α- and β-tubulin heterodimer, are considered as a major target to prevent the proliferation of tumor cells. Microtubule-targeted agents have become increasingly effective anticancer drugs. However, due to the relatively sophisticated chemical structure of taxane and vinblastine, their application has faced numerous obstacles. Conversely, the structure of colchicine binding site inhibitors (CBSIs) is much easier to be modified. Moreover, CBSIs have strong antiproliferative effect on multidrug-resistant tumor cells and have become the mainstream research orientation of microtubule-targeted agents. This review focuses mainly on the recent advances of CBSIs during 2017-2022, attempts to depict their biological activities to analyze the structure-activity relationships and offers new perspectives for designing next generation of novel CBSIs.
Collapse
|
5
|
Dehbid M, Tahmasvand R, Tasharofi M, Shojaie F, Aghamaali M, Almasirad A, Salimi M. Synthesis of oxamide-hydrazone hybrid derivatives as potential anticancer agents. Res Pharm Sci 2022; 18:24-38. [PMID: 36846733 PMCID: PMC9951783 DOI: 10.4103/1735-5362.363593] [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: 08/13/2022] [Revised: 09/04/2022] [Accepted: 09/11/2022] [Indexed: 12/25/2022] Open
Abstract
Background and purpose Considering various studies implying anticancer activity of the hydrazone and oxamide derivatives through different mechanisms such as kinases and calpain inhibition, herein, we report the synthesis, characterization, and evaluation of the antiproliferative effect of a series of hydrazones bearing oxamide moiety compounds (7a-7n) against a panel of cancer cell lines to explore a novel and promising anticancer agent (7k). Experimental approach Chemical structures of the synthesized compounds were confirmed by FTIR, 1H-NMR, 13C-NMR, and mass spectra. The antiproliferative activity and cell cycle progression of the target compound were investigated using the MTT assay and flow cytometry. Findings/Results Compound 7k with 2-hydroxybenzylidene structure was found to have a significant in vitro anti-proliferative influence on MDA-MB-231 (human adenocarcinoma breast cancer) and 4T1 (mouse mammary tumor) cells as the model of triple-negative breast cancer, with the IC50-72h values of 7.73 ± 1.05 and 1.82 ± 1.14 μM, respectively. Following 72-h incubation with compound 7k, it caused MDA-MB-231 cell death through G1/S cell cycle arrest at high concentrations (12 and 16 μM). Conclusion and implications Conclusively, this study for the first time reports the anti-proliferative efficacy of compound 7k possessing 2-hydroxyphenyl moiety, which may serve as a potent candidate in triple-negative breast cancer treatment.
Collapse
Affiliation(s)
- Mina Dehbid
- Department of Biology, Faculty of Science, University of Guilan, Rasht, I.R. Iran
| | - Raheleh Tahmasvand
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, I.R. Iran
| | - Marzieh Tasharofi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, I.R. Iran
| | - Fatemeh Shojaie
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, I.R. Iran
| | | | - Ali Almasirad
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, I.R. Iran,Corresponding authors: A. Almasirad, Tel: +98-2122640051, Fax: +98-22602059
M. Salimi, Tel: +98-2164112264, Fax: +98-2164112834
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, I.R. Iran,Corresponding authors: A. Almasirad, Tel: +98-2122640051, Fax: +98-22602059
M. Salimi, Tel: +98-2164112264, Fax: +98-2164112834
| |
Collapse
|
6
|
Yang S, Wang C, Shi L, Chang J, Zhang Y, Meng J, Liu W, Zeng J, Zhang R, Shao Y, Xing D. Design, synthesis and biological evaluation of novel diarylpyridine derivatives as tubulin polymerisation inhibitors. J Enzyme Inhib Med Chem 2022; 37:2755-2764. [PMID: 36196773 PMCID: PMC9553186 DOI: 10.1080/14756366.2022.2130284] [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] [Indexed: 10/25/2022] Open
Abstract
A set of novel diarylpyridines as anti-tubulin agents were designed, synthesised using a rigid pyridine as a linker to fix the cis-orientation of ring-A and ring-B. All of the target compounds were evaluated for their in vitro antiproliferative activities. Among them, 10t showed remarkable antiproliferative activities against three cancer cell lines (HeLa, MCF-7 and SGC-7901) in sub-micromolar concentrations. Consistent with its potent antiproliferative activity, 10t also displayed potent anti-tubulin activity. Cellular mechanism investigation elucidated 10t disrupted the cellular microtubule structure, arrested cell cycle at G2/M phase and induces apoptosis. Molecular modelling studies showed that 10t could bind to the colchicine binding site on microtubules. These results provide motivation and further guidance for the development of new CA-4 analogues.
Collapse
Affiliation(s)
- Shanbo Yang
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Chao Wang
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Lingyu Shi
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Jing Chang
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jingsen Meng
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Wenjing Liu
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Jun Zeng
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Yingchun Shao
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao, China.,Qingdao Cancer Institute, Qingdao, China.,School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Chang J, Wang C, Yang S, Shi L, Zhang Y, Liu W, Meng J, Zeng J, Zhang R, Liu N, Xing D. Design, synthesis, and biological evaluation of diarylpyrazole derivatives as antitumor agents targeting microtubules. ARAB J CHEM 2022. [DOI: 10.1016/j.arabjc.2022.104253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
8
|
Ebenezer O, Shapi M, Tuszynski JA. A Review of the Recent Developments of Molecular Hybrids Targeting Tubulin Polymerization. Int J Mol Sci 2022; 23:4001. [PMID: 35409361 PMCID: PMC8999808 DOI: 10.3390/ijms23074001] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/17/2022] [Accepted: 03/29/2022] [Indexed: 12/17/2022] Open
Abstract
Microtubules are cylindrical protein polymers formed from αβ-tubulin heterodimers in the cytoplasm of eukaryotic cells. Microtubule disturbance may cause cell cycle arrest in the G2/M phase, and anomalous mitotic spindles will form. Microtubules are an important target for cancer drug action because of their critical role in mitosis. Several microtubule-targeting agents with vast therapeutic advantages have been developed, but they often lead to multidrug resistance and adverse side effects. Thus, single-target therapy has drawbacks in the effective control of tubulin polymerization. Molecular hybridization, based on the amalgamation of two or more pharmacophores of bioactive conjugates to engender a single molecular structure with enhanced pharmacokinetics and biological activity, compared to their parent molecules, has recently become a promising approach in drug development. The practical application of combined active scaffolds targeting tubulin polymerization inhibitors has been corroborated in the past few years. Meanwhile, different designs and syntheses of novel anti-tubulin hybrids have been broadly studied, illustrated, and detailed in the literature. This review describes various molecular hybrids with their reported structural-activity relationships (SARs) where it is possible in an effort to generate efficacious tubulin polymerization inhibitors. The aim is to create a platform on which new active scaffolds can be modeled for improved tubulin polymerization inhibitory potency and hence, the development of new therapeutic agents against cancer.
Collapse
Affiliation(s)
- Oluwakemi Ebenezer
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Michael Shapi
- Department of Chemistry, Faculty of Natural Science, Mangosuthu University of Technology, Umlazi 4031, South Africa; (O.E.); (M.S.)
| | - Jack A. Tuszynski
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB T6G 1Z2, Canada
- Department of Physics, University of Alberta, Edmonton, AB T6G 2E1, Canada
- DIMEAS, Politecnico di Torino, 10129 Turin, Italy
| |
Collapse
|
9
|
López-López E, Cerda-García-Rojas CM, Medina-Franco JL. Tubulin Inhibitors: A Chemoinformatic Analysis Using Cell-Based Data. Molecules 2021; 26:2483. [PMID: 33923169 PMCID: PMC8123128 DOI: 10.3390/molecules26092483] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
Inhibiting the tubulin-microtubules (Tub-Mts) system is a classic and rational approach for treating different types of cancers. A large amount of data on inhibitors in the clinic supports Tub-Mts as a validated target. However, most of the inhibitors reported thus far have been developed around common chemical scaffolds covering a narrow region of the chemical space with limited innovation. This manuscript aims to discuss the first activity landscape and scaffold content analysis of an assembled and curated cell-based database of 851 Tub-Mts inhibitors with reported activity against five cancer cell lines and the Tub-Mts system. The structure-bioactivity relationships of the Tub-Mts system inhibitors were further explored using constellations plots. This recently developed methodology enables the rapid but quantitative assessment of analog series enriched with active compounds. The constellations plots identified promising analog series with high average biological activity that could be the starting points of new and more potent Tub-Mts inhibitors.
Collapse
Affiliation(s)
- Edgar López-López
- Departamento de Química y Programa de Posgrado en Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Apartado 14-740, Mexico City 07000, Mexico;
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de México, Mexico City 04510, 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, Apartado 14-740, 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
|