1
|
I A, Purawarga Matada GS, Pal R, Ghara A, Aishwarya NVSS, B K, Hosamani KR, B V M, E H. Benzothiazole a privileged scaffold for Cutting-Edges anticancer agents: Exploring drug design, structure-activity relationship, and docking studies. Eur J Med Chem 2024; 279:116831. [PMID: 39255643 DOI: 10.1016/j.ejmech.2024.116831] [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: 07/08/2024] [Revised: 08/30/2024] [Accepted: 09/01/2024] [Indexed: 09/12/2024]
Abstract
Cancer is a major societal, public health, and economic burden in the 21st century, with 9.7 million deaths in 2022 (9.96 million in 2020) and 20 million new cancer cases (19.6 million in 2020). Considering the increasing number of cancer cases and deaths, heterocyclic compounds always paved the gold mine for the development of potential anticancer drugs as these compounds have unique flexibility and dynamic cores. Benzothiazoles and their derivatives have potential anticancer properties, making them a desirable scaffold among different heterocycles. Title structures are a class of chemicals that may bind to various receptors with high affinity, particularly those engaged in oncogenic processes. The use of these compounds allows medicinal chemists to rapidly produce anticancer treatments across a large range of targets over an extended length of time. The current study presents a thorough success story of benzothiazole derivatives as anticancer agents. It discusses the current state of cancer, the profile of benzothiazole-based derivatives synthetic pathways, and its relevance as an anticancer agent on several oncogenic pathways. The structure-activity relationship was also added to offer insight into the connection of biological data with structure and the rational design of more active drugs.
Collapse
Affiliation(s)
- Aayishamma I
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Gurubasavaraja Swamy Purawarga Matada
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Rohit Pal
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India.
| | - Abhishek Ghara
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | | | - Kumaraswamy B
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Ketan R Hosamani
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Manjushree B V
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| | - Haripriya E
- Integrated Drug Discovery Centre, Department of Pharmaceutical Chemistry, Acharya & BM Reddy College of Pharmacy, Bengaluru, 560107, Karnataka, India
| |
Collapse
|
2
|
Hashem H, Hassan A, Abdelmagid WM, Habib AGK, Abdel-Aal MAA, Elshamsy AM, El Zawily A, Radwan IT, Bräse S, Abdel-Samea AS, Rabea SM. Synthesis of New Thiazole-Privileged Chalcones as Tubulin Polymerization Inhibitors with Potential Anticancer Activities. Pharmaceuticals (Basel) 2024; 17:1154. [PMID: 39338317 PMCID: PMC11435058 DOI: 10.3390/ph17091154] [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: 07/28/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
A series of novel thiazole-based chalcones were evaluated for their anticancer activity as potential tubulin polymerization inhibitors. In vitro anticancer screening for the thiazole derivatives 2a-2p exhibited broad-spectrum antitumor activity against various cancer cell lines particularly Ovar-3 and MDA-MB-468 cells with a GI50 range from 1.55 to 2.95 μΜ, respectively. Compound 2e demonstrated significant inhibition of tubulin polymerization, with an IC50 value of 7.78 μM compared to Combretastatin-A4 (CA-4), with an IC50 value of 4.93 μM. Molecular docking studies of compounds 2e, 2g, and 2h into tubulin further supported these findings, revealing that they bind effectively to the colchicine binding site, mirroring key interactions exhibited by CA-4. Computational predictions suggested favorable oral bioavailability and drug-likeness for these compounds, highlighting their potential for further development as chemotherapeutic agents.
Collapse
Affiliation(s)
- Hamada Hashem
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Sohag University, Sohag 82524, Egypt
| | - Abdelfattah Hassan
- Medicinal Chemistry Department, Faculty of Pharmacy, South Valley University, Qena 52242, Egypt
- Medicinal Chemistry Department, Clinical Pharmacy Program, South Valley National University, Qena 52242, Egypt
| | - Walid M Abdelmagid
- Medicinal Chemistry and Drug Discovery Research Centre, Swenam College, 210-6125 Sussex Avenue, Burnaby, BC V5H 4G1, Canada
| | - Ahmed G K Habib
- Department of Biotechnology and Life Sciences, Faculty of Postgraduate Studies for Advanced Sciences, Beni-Suef University, Beni-Suef 62521, Egypt
| | - Mohamed A A Abdel-Aal
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Ali M Elshamsy
- Medicinal Chemistry Department, Faculty of Pharmacy, Deraya University, New Minia 61768, Egypt
| | - Amr El Zawily
- Department of Plant and Microbiology, Faculty of Science, Damanhour University, Damanhour 22511, Egypt
- Division of Pharmaceutics and Translation Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | - Ibrahim Taha Radwan
- Supplementary General Sciences Department, Faculty of Oral and Dental Medicine, Future University in Egypt, Cairo 11835, Egypt
| | - Stefan Bräse
- Institute of Biological and Chemical Systems-Functional Molecular Systems (IBCS-FMS), Karlsruhe Institute of Technology (KIT), Kaiserstrasse 12, 76131 Karlsruhe, Germany
| | - Ahmed S Abdel-Samea
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Deraya University, New Minia 61768, Egypt
| | - Safwat M Rabea
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
- Apogee Pharmaceuticals Inc., 4475 Wayburne Dr., Suite 105, Burnaby, BC V6V2H8, Canada
| |
Collapse
|
3
|
Nagavath R, Thupurani MK, Badithapuram V, Manchal R, Vasam CS, Thirukovela NS. Organo NHC catalyzed aqueous synthesis of 4β-isoxazole-podophyllotoxins: in vitro anticancer, caspase activation, tubulin polymerization inhibition and molecular docking studies. RSC Adv 2024; 14:23574-23582. [PMID: 39070249 PMCID: PMC11276401 DOI: 10.1039/d4ra04297b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
We present, for the first time, the organo-N-heterocyclic carbene (NHC) catalyzed 1,3-dipolar cycloaddition of 4β-O-propargyl podophyllotoxin (1) with in situ aromatic nitrile oxides to afford regioselective 4β-isoxazolepodophyllotoxin hybrids (6a-n) in benign aqueous-organic media. Preliminary anticancer activity results showed that compound 6e displayed superior activity against MCF-7, HeLa and MIA PaCa2 human cell lines compared with podophyllotoxin. Compounds 6j and 6n showed greater activity against the MCF-7 cell line than the positive control. Caspase activation studies revealed that compound 6e at 20 μg ml-1 concentration had greater caspase 3/7 activation in MCF-7 and MIAPaCa2 cells than podophyllotoxin. Furthermore, in vitro tubulin polymerization inhibition studies revealed that compound 6e showed comparable activity with podophyllotoxin. Finally, in silico molecular docking studies of compounds 6e, 6j, 6n and podophyllotoxin on α,β-tubulin (pdb id 1SA0) revealed that compound 6n showed excellent binding energies and inhibition constants compared with podophyllotoxin.
Collapse
Affiliation(s)
- Rajkumar Nagavath
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Murali Krishna Thupurani
- Department of Biotechnology, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Vinitha Badithapuram
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | - Ravinder Manchal
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| | | | - Narasimha Swamy Thirukovela
- Department of Chemistry, Chaitanya (Deemed to be University) Himayathnagar (V), Moinabad (M), Ranga Reddy (D) Hyderabad India
| |
Collapse
|
4
|
Kamboj P, Mahore A, Husain A, Amir M. Benzothiazole-based apoptosis inducers: A comprehensive overview and future prospective. Arch Pharm (Weinheim) 2024; 357:e2300493. [PMID: 38212254 DOI: 10.1002/ardp.202300493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/24/2023] [Accepted: 12/06/2023] [Indexed: 01/13/2024]
Abstract
Cancer has become a major concern in healthcare globally, and over time, incidences and prevalence of cancer are increasing. To counter this, a lot of anticancer drugs are approved and are in clinical use, playing a pivotal role in its treatment. Due to drug resistance and adverse effects, a continuous demand for novel, potent, and safe candidates to treat cancer is always there. Over the last few decades, various heterocyclic ring-based derivatives have been explored and reported in the literature. In this regard, benzothiazole scaffold-based compound emerged as the versatile ring for developing novel and safe anticancer candidates. In this article, we have reported various benzothiazole heterocyclic ring-based derivatives demonstrating potent antiproliferative activity by induction of apoptosis via an intrinsic pathway in a dose-dependent manner. These compounds also displayed inhibition of different enzymes, for example, Aurora kinase, epidermal growth factor receptor, vascular endothelial growth factor receptor, phosphoinositide kinases, DNA topoisomerase, and tubulin polymerases. This study focused on a comprehensive overview of antiproliferative activity, structure-activity relationship, apoptosis induction activity, and enzyme inhibition by benzothiazole-based compounds.
Collapse
Affiliation(s)
- Payal Kamboj
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, India
| | - Anjali Mahore
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, India
| | - Asif Husain
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, India
| | - Mohammad Amir
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, India
| |
Collapse
|
5
|
Wu BW, Huang WJ, Liu YH, Liu QG, Song J, Hu T, Chen P, Zhang SY. Design, synthesis and biological evaluation of 1,2,3-triazole benzothiazole derivatives as tubulin polymerization inhibitors with potent anti-esophageal cancer activities. Eur J Med Chem 2024; 265:116118. [PMID: 38181651 DOI: 10.1016/j.ejmech.2023.116118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/20/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
In this work, we utilized the molecular hybridization strategy to design and synthesize novel 1,2,3-triazole benzothiazole derivatives K1-26. The antiproliferative activities against MGC-803, Kyse30 and HCT-116 cells were explored, and their structure-activity relationship were preliminarily conducted and summarized. Among them, compound K18, exhibited the strongest proliferation inhibitory activity, with esophageal cancer cells Kyse30 and EC-109 being the most sensitive to its effects (IC50 values were 0.042 and 0.038 μM, respectively). Compound K18 effectively inhibited tubulin polymerization (IC50 = 0.446 μM), thereby hindering tubulin polymerize into filamentous microtubules in Kyse30 and EC-109 cells. Additionally, compound K18 induced the degradation of oncogenic protein YAP via the UPS pathway. Based on these dual molecular-level effects, compound K18 could induce G2/M phase arrest and cell apoptosis in Kyse30 and EC-109 cells, as well as regulate the expression levels of cell cycle and apoptosis-related proteins. In summary, our findings highlight a novel 1,2,3-triazole benzothiazole derivative K18, which possesses significant potential for treating esophageal cancers.
Collapse
Affiliation(s)
- Bo-Wen Wu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wen-Jing Huang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yun-He Liu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Qiu-Ge Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Tao Hu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ping Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China; State Key Laboratory of Esophageal Cancer Prevention &Treatment, Zhengzhou 450001, China.
| |
Collapse
|
6
|
Liu W, He Y, Guo Z, Wang M, Han X, Jia H, He J, Miao S, Wang S. Discovery of potent tubulin inhibitors targeting the colchicine binding site via structure-based lead optimization and antitumor evaluation. J Enzyme Inhib Med Chem 2023; 38:2155815. [PMID: 36629423 PMCID: PMC9848350 DOI: 10.1080/14756366.2022.2155815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The colchicine binding site of tubulin is a promising target for discovering novel antitumour agents. Previously, we identified 2-aryl-4-amide-quinoline derivatives displayed moderate tubulin polymerisation inhibitory activity and broad-spectrum in vitro antitumour activity. In this study, structure based rational design and systematic structural optimisation were performed to obtain analogues C1∼J2 bearing diverse substituents and scaffolds. Among them, analogue G13 bearing a hydroxymethyl group displayed good tubulin polymerisation inhibitory activity (IC50 = 13.5 μM) and potent antiproliferative activity (IC50 values: 0.65 μM∼0.90 μM). G13 potently inhibited the migration and invasion of MDA-MB-231 cells, and displayed potent antiangiogenic activity. It efficiently increased intracellular ROS level and decreased MMP in cancer cells, and obviously induced the fragmentation and disassembly of the microtubules network. More importantly, G13 exhibited good in vivo antitumour efficacy in MDA-MB-231 xenograft model (TGI = 38.2%; i.p., 30 mg/kg).
Collapse
Affiliation(s)
- Wei Liu
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Youyou He
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China,Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Zhongjie Guo
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Miaomiao Wang
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Xiaodong Han
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Hairui Jia
- Faculty of Pharmacy, School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an, China
| | - Jin He
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Shanshan Miao
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| | - Shengzheng Wang
- Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China,CONTACT Shengzheng Wang Department of Medicinal Chemistry and Pharmaceutical Analysis, School of Pharmacy, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
7
|
Gallego-Yerga L, Ceña V, Peláez R. Potent and Selective Benzothiazole-Based Antimitotics with Improved Water Solubility: Design, Synthesis, and Evaluation as Novel Anticancer Agents. Pharmaceutics 2023; 15:1698. [PMID: 37376146 DOI: 10.3390/pharmaceutics15061698] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The design of colchicine site ligands on tubulin has proven to be a successful strategy to develop potent antiproliferative drugs against cancer cells. However, the structural requirements of the binding site endow the ligands with low aqueous solubility. In this work, the benzothiazole scaffold is used to design, synthesize, and evaluate a new family of colchicine site ligands exhibiting high water solubility. The compounds exerted antiproliferative activity against several human cancer cell lines, due to tubulin polymerization inhibition, showing high selectivity toward cancer cells in comparison with non-tumoral HEK-293 cells, as evidenced by MTT and LDH assays. The most potent derivatives, containing a pyridine moiety and ethylurea or formamide functionalities, displayed IC50 values in the nanomolar range even in the difficult-to-treat glioblastoma cells. Flow cytometry experiments on HeLa, MCF7, and U87MG cells showed that they arrest the cell cycle at the G2/M phases at an early time point (24 h), followed by apoptotic cell death 72 h after the treatment. Tubulin binding was confirmed by microtubule network disruption observed via confocal microscopy. Docking studies support favorable interaction of the synthesized ligands at the colchicine binding site. These results validate the proposed strategy to develop potent anticancer colchicine ligands with improved water solubility.
Collapse
Affiliation(s)
- Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | - Valentín Ceña
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Unidad Asociada Neurodeath, Facultad de Medicina, Universidad de Castilla-La Mancha, 02006 Albacete, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
- Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| |
Collapse
|
8
|
Sahil, Kaur K, Jaitak V. Thiazole and Related Heterocyclic Systems as Anticancer Agents: A Review on Synthetic Strategies, Mechanisms of Action and SAR Studies. Curr Med Chem 2022; 29:4958-5009. [DOI: 10.2174/0929867329666220318100019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
Background:
Cancer is the second leading cause of death throughout the world. Many anticancer drugs are commercially available, but lack of selectivity, target specificity, cytotoxicity and development of resistance lead to serious side effects. There have been several experiments going on to develop compounds with minor or no side effects.
Objective:
This review mainly emphasizes synthetic strategies, SAR studies, and mechanism of action for thiazole, benzothiazole, and imidazothiazole containing compounds as anticancer agents.
Methods:
Recent literature related to thiazole and thiazole-related derivatives endowed with encouraging anticancer potential is reviewed. This review emphasizes contemporary strategies used for the synthesis of thiazole and related derivatives, mechanistic targets, and comprehensive structural activity relationship studies to provide perspective into the rational design of high-efficiency thiazole-based anticancer drug candidates.
Results:
Exhaustive literature survey indicated that thiazole derivatives are associated with properties of inducing
apoptosis and disturbing tubulin assembly. Thiazoles are also associated with the inhibition of NFkB/mTOR/PI3K/AkT and regulation of estrogen-mediated activity. Furthermore, thiazole derivatives have been found to modulate critical targets such as topoisomerase and HDAC.
Conclusion:
Thiazole derivatives seem to be quite competent and act through various mechanisms. Some of the thiazole derivatives, such as compounds 29, 40, 62, and 74a with IC50 values of 0.05 μM, 0.00042 μM, 0.18 μM, and 0.67 μM, respectively not only have anticancer activity but they also have lower toxicity and better absorption. Therefore, some other similar compounds could be investigated to aid in the development of anticancer pharmacophores.
Collapse
Affiliation(s)
- Sahil
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), India
| | - Kamalpreet Kaur
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), India
| | - Vikas Jaitak
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghudda, Bathinda (Pb.), India
| |
Collapse
|
9
|
BP-M345, a New Diarylpentanoid with Promising Antimitotic Activity. Molecules 2021; 26:molecules26237139. [PMID: 34885726 PMCID: PMC8659070 DOI: 10.3390/molecules26237139] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/20/2021] [Accepted: 11/23/2021] [Indexed: 01/09/2023] Open
Abstract
Previously, we reported the in vitro growth inhibitory effect of diarylpentanoid BP-M345 on human cancer cells. Nevertheless, at that time, the cellular mechanism through which BP-M345 exerts its growth inhibitory effect remained to be explored. In the present work, we report its mechanism of action on cancer cells. The compound exhibits a potent tumor growth inhibitory activity with high selectivity index. Mechanistically, it induces perturbation of the spindles through microtubule instability. As a consequence, treated cells exhibit irreversible defects in chromosome congression during mitosis, which induce a prolonged spindle assembly checkpoint-dependent mitotic arrest, followed by massive apoptosis, as revealed by live cell imaging. Collectively, the results indicate that the diarylpentanoid BP-M345 exerts its antiproliferative activity by inhibiting mitosis through microtubule perturbation and causing cancer cell death, thereby highlighting its potential as antitumor agent.
Collapse
|
10
|
Discovery of novel tubulin inhibitors targeting the colchicine binding site via virtual screening, structural optimization and antitumor evaluation. Bioorg Chem 2021; 118:105486. [PMID: 34801948 DOI: 10.1016/j.bioorg.2021.105486] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/31/2021] [Accepted: 11/09/2021] [Indexed: 11/21/2022]
Abstract
The colchicine binding site of tubulin is a promising target for discovering novel antitumor agents which exert the antiangiogenic effect and are not susceptible to multidrug resistance. For identifying novel tubulin inhibitors, structure-based virtual screening was applied to identify hit 9 which displayed moderate tubulin polymerization inhibition and broad-spectrum in vitro antitumor activity. Structural optimization was performed, and biological assay revealed analog E27 displayed the best antitumor activity with IC50 values ranging from 7.81 μM to 10.36 μM, and improved tubulin polymerization inhibitory activity (IC50 = 16.1 μM). It significantly inhibited cancer cell migration and invasion, induced cell apoptosis and arrested the cell cycle at G2/M phase. Moreover, the apoptotic effect of E27 is related to the increased ROS level, the decrease of MMP, and the abnormal expression of apoptosis-related proteins. Taken together, these results suggested E27 was a promising lead compound for discovering novel tubulin-targeted antitumor agents.
Collapse
|
11
|
Beč A, Hok L, Persoons L, Vanstreels E, Daelemans D, Vianello R, Hranjec M. Synthesis, Computational Analysis, and Antiproliferative Activity of Novel Benzimidazole Acrylonitriles as Tubulin Polymerization Inhibitors: Part 2. Pharmaceuticals (Basel) 2021; 14:1052. [PMID: 34681276 PMCID: PMC8540608 DOI: 10.3390/ph14101052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/09/2021] [Accepted: 10/13/2021] [Indexed: 11/23/2022] Open
Abstract
We used classical linear and microwave-assisted synthesis methods to prepare novel N-substituted, benzimidazole-derived acrylonitriles with antiproliferative activity against several cancer cells in vitro. The most potent systems showed pronounced activity against all tested hematological cancer cell lines, with favorable selectivity towards normal cells. The selection of lead compounds was also tested in vitro for tubulin polymerization inhibition as a possible mechanism of biological action. A combination of docking and molecular dynamics simulations confirmed the suitability of the employed organic skeleton for the design of antitumor drugs and demonstrated that their biological activity relies on binding to the colchicine binding site in tubulin. In addition, it also underlined that higher tubulin affinities are linked with (i) bulkier alkyl and aryl moieties on the benzimidazole nitrogen and (ii) electron-donating substituents on the phenyl group that allow deeper entrance into the hydrophobic pocket within the tubulin's β-subunit, consisting of Leu255, Leu248, Met259, Ala354, and Ile378 residues.
Collapse
Affiliation(s)
- Anja Beč
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia;
| | - Lucija Hok
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia;
| | - Leentje Persoons
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, 3000 Leuven, Belgium; (L.P.); (E.V.); (D.D.)
| | - Els Vanstreels
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, 3000 Leuven, Belgium; (L.P.); (E.V.); (D.D.)
| | - Dirk Daelemans
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, 3000 Leuven, Belgium; (L.P.); (E.V.); (D.D.)
| | - Robert Vianello
- Laboratory for the Computational Design and Synthesis of Functional Materials, Division of Organic Chemistry and Biochemistry, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia;
| | - Marijana Hranjec
- Department of Organic Chemistry, Faculty of Chemical Engineering and Technology, University of Zagreb, Marulićev trg 19, HR-10000 Zagreb, Croatia;
| |
Collapse
|
12
|
Arya GC, Kaur K, Jaitak V. Isoxazole derivatives as anticancer agent: A review on synthetic strategies, mechanism of action and SAR studies. Eur J Med Chem 2021; 221:113511. [PMID: 34000484 DOI: 10.1016/j.ejmech.2021.113511] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/16/2021] [Accepted: 04/29/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is the second most leading cause of death among women. Multiple drugs have been approved by FDA for the treatment of BC. The major drawbacks of existing drugs are the development of resistance, toxicity, selectivity problem. The other therapies like hormonal therapy, surgery, radiotherapy, and immune therapy are in use but showed many side effects like bioavailability issues, non-selectivity, pharmacokinetic-pharmacodynamic problems. Therefore, there is an urgent need to develop new moieties that are nonviolent and more effective in the treatment of cancer. Isoxazole derivatives have gain popularity in recent years due to anticancer potential with the least side effects. These derivatives act as an anticancer agent with different mechanisms like inducing apoptosis, aromatase inhibition, disturbing tubulin congregation, topoisomerase inhibition, HDAC inhibition, and ERα inhibition. In this article, we have explored the synthetic strategies, anticancer mechanism of action along with SAR studies of isoxazole derivatives.
Collapse
Affiliation(s)
- Girish Chandra Arya
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghhudha, Bathinda, Pb, 151401, India
| | - Kamalpreet Kaur
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghhudha, Bathinda, Pb, 151401, India
| | - Vikas Jaitak
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, Ghhudha, Bathinda, Pb, 151401, India.
| |
Collapse
|
13
|
Komuraiah B, Ren Y, Xue M, Cheng B, Liu J, Liu Y, Chen J. Design, synthesis and biological evaluation of benz-fused five-membered heterocyclic compounds as tubulin polymerization inhibitors with anticancer activities. Chem Biol Drug Des 2021; 97:1109-1116. [PMID: 33638903 DOI: 10.1111/cbdd.13832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 01/31/2021] [Indexed: 01/03/2023]
Abstract
A series of benz-fused five-membered heterocyclic compounds were designed and synthesized as novel tubulin inhibitors targeting the colchicine binding site. Among them, compound 4d displayed the highest antiproliferative activity against four cancer cell lines with an IC50 value of 4.9 μM in B16-F10 cells. Compound 4d effectively inhibited tubulin polymerization in vitro (IC50 of 13.1 μM). Further, 4d induced cell cycle arrest in G2/M phase. Finally, 4d inhibited the migration of cancer cells in a dose-dependent manner. In summary, these results suggest that compound 4d represents a new class of tubulin inhibitors deserving further investigation.
Collapse
Affiliation(s)
- Buduma Komuraiah
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| | - Yichang Ren
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| | - Mingming Xue
- Tianjin Tiancheng Chemical Co., Ltd, Tianjin, China
| | - Binbin Cheng
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| | - Jin Liu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| | - Yao Liu
- Instrumental Analysis Center, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Sardaru MC, Craciun AM, Al Matarneh CM, Sandu IA, Amarandi RM, Popovici L, Ciobanu CI, Peptanariu D, Pinteala M, Mangalagiu II, Danac R. Cytotoxic substituted indolizines as new colchicine site tubulin polymerisation inhibitors. J Enzyme Inhib Med Chem 2020; 35:1581-1595. [PMID: 32752898 PMCID: PMC7470029 DOI: 10.1080/14756366.2020.1801671] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/22/2020] [Accepted: 07/19/2020] [Indexed: 12/20/2022] Open
Abstract
A potential microtubule destabilising series of new indolizine derivatives was synthesised and tested for their anticancer activity against a panel of 60 human cancer cell lines. Compounds 11a, 11b, 15a, and 15j showed a broad spectrum of growth inhibitory activity against cancer cell lines representing leukaemia, melanoma and cancer of lung, colon, central nervous system, ovary, kidney, breast, and prostate. Among them, compound 11a was distinguishable by its excellent cytostatic activity, showing GI50 values in the range of 10-100 nM on 43 cell lines. The less potent compounds 15a and 15j in terms of GI50 values showed a high cytotoxic effect against tested colon cancer, CNS cancer, renal cancer and melanoma cell lines and only on few cell lines from other types of cancer. In vitro assaying revealed tubulin polymerisation inhibition by all active compounds. Molecular docking showed good complementarity of active compounds with the colchicine binding site of tubulin.
Collapse
Affiliation(s)
- Monica-Cornelia Sardaru
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
- “Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy, Iași, Romania
| | - Anda Mihaela Craciun
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
- “Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy, Iași, Romania
| | - Cristina-Maria Al Matarneh
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
- “Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy, Iași, Romania
| | - Isabela Andreea Sandu
- “Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy, Iași, Romania
| | - Roxana Maria Amarandi
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
- TRANSCEND Research Center, Regional Institute of Oncology, Iași, Romania
| | - Lacramioara Popovici
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
| | | | - Dragos Peptanariu
- “Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy, Iași, Romania
| | - Mariana Pinteala
- “Petru Poni” Institute of Macromolecular Chemistry of Romanian Academy, Iași, Romania
| | - Ionel I. Mangalagiu
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
| | - Ramona Danac
- Department of Chemistry, Faculty of Chemistry, “Al. I. Cuza” University of Iasi, Iași, Romania
| |
Collapse
|