1
|
Abdelrazek FM, Zaki ME, Al-Hussain SA, Farag B, Hebishy AM, Abdelfattah MS, Hassan SM, El-Farargy AF, Iovkova L, Mross D, Gomha SM. Facile one-pot synthesis and in silico study of new heterocyclic scaffolds with 4-pyridyl moiety: Mechanistic insights and X-ray crystallographic elucidation. Heliyon 2024; 10:e29221. [PMID: 38617929 PMCID: PMC11015136 DOI: 10.1016/j.heliyon.2024.e29221] [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: 10/19/2023] [Revised: 03/08/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024] Open
Abstract
4-Acetylpyridine 1 and malononitrile 2 were allowed to react in a 3MCRs with dimedone 3a or cyclohexa-1,3-dione 3b under reflux to afford 4-methyl-4-(pyridin-4-yl)-5,6,7,8-tetrahydro-4H-chromene derivatives 4a,b respectively. The mechanism of the reaction has been studied and the structures elucidated by analytical, spectral as well as X-ray crystallographic data. Heterocyclic compounds find widespread application in pharmaceutical and agrochemical products. Docking analyses were performed on the synthesized compounds to assess their binding modes with various amino acids of the target protein tubulin (PDB Code - 1SA0). The results indicated promising binding scores for compounds 4a and 4b, suggesting a strong affinity for the tubulin binding site. Finally, ADMET for the synthesized compounds 4a, 4b, 5, 8a and 8b were carried out. The drug likeness and pharmacokinetic properties of the prepared compounds were also evaluated. Notably, all of the novel compounds adhered to Lipinski's rule (Ro5) without any violations.
Collapse
Affiliation(s)
- Fathy M. Abdelrazek
- Chemistry Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Magdi E.A. Zaki
- Department of Chemistry, Faculty of Science, Imam Mohammed Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Sami A. Al-Hussain
- Department of Chemistry, Faculty of Science, Imam Mohammed Ibn Saud Islamic University (IMSIU), Riyadh, 11623, Saudi Arabia
| | - Basant Farag
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
| | - Ali M. Hebishy
- Chemistry Department, Faculty of Science, Helwan University, Helwan, 11795, Cairo, Egypt
| | - Mohamed S. Abdelfattah
- Chemistry Department, Faculty of Science, Helwan University, Helwan, 11795, Cairo, Egypt
| | - Safaa M. Hassan
- Chemistry Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
- Chemistry Department, Faculty of Science, Helwan University, Helwan, 11795, Cairo, Egypt
| | - Ahmed F. El-Farargy
- Department of Chemistry, Faculty of Science, Zagazig University, Zagazig, 44519, Egypt
- Fakultät für Chemie und Chemische Biologie, TU Dortmund, Dortmund, 44227, Germany
| | - Lyuba Iovkova
- Fakultät für Chemie und Chemische Biologie, TU Dortmund, Dortmund, 44227, Germany
| | - David Mross
- Fakultät für Chemie und Chemische Biologie, TU Dortmund, Dortmund, 44227, Germany
| | - Sobhi M. Gomha
- Department of Chemistry, Faculty of Science, Islamic University of Madinah, Madinah, 42351, Saudi Arabia
| |
Collapse
|
2
|
Chudasama DD, Rajput CV, Patel MS, Parekh JN, Patel HC, Chikhaliya NP, Puerta A, Padrón JM, Ram KR. Microwave-induced one-pot synthesis of 3-imidazolyl indole clubbed 1,2,3-triazole hybrids as antiproliferative agents and density functional theory study. Arch Pharm (Weinheim) 2024; 357:e2300632. [PMID: 38150663 DOI: 10.1002/ardp.202300632] [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: 10/31/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023]
Abstract
Herein, we outline a highly efficient PEG-4000-mediated one-pot three-component reaction for the synthesis of 3-imidazolyl indole clubbed 1,2,3-triazole derivatives (5a-r) at up to 96% yield as antiproliferative agents. This three-component protocol offers the advantages of an environmentally benign reaction, excellent yield, quick response time, and operational simplicity triggered by the copper catalyst under microwave irradiation. All the synthesized compounds were tested for antiproliferative activity against six human solid tumor cell lines, that is, A549 and SW1573 (nonsmall cell lung), HBL100 and T-47D (breast), HeLa (cervix), and WiDr (colon). Among them, six compounds, 5g-j, 5m, and 5p, demonstrated effective antiproliferative action with GI50 values under 10 μM. Furthermore, density functional theory (DFT) calculations were performed for all the synthesized molecules through geometry optimizations, frontier molecular orbital approach, and molecular electrostatic potential (MESP). The theoretical DFT calculation was performed using the DFT/B3LYP/6-31+G (d,p) basis set. Moreover, the biological reactivity of all the representative synthesized molecules was compared with the theoretically calculated quantum chemical descriptors and MESP 3D plots. We also investigated the drug-likeness characteristic and absorption, distribution, metabolism, excretion, and toxicity (ADMET) prediction. In general, our approach enables environmentally friendly access to 3-imidazolyl indole clubbed 1,2,3-triazole derivatives as prospective antiproliferative agents.
Collapse
Affiliation(s)
| | - Chetan V Rajput
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Manan S Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Jaydeepkumar N Parekh
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Harsh C Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Navin P Chikhaliya
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, La Laguna, Spain
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, La Laguna, Spain
| | - Kesur R Ram
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Gujarat, India
| |
Collapse
|
3
|
Abdul Hussein SA, Razzak Mahmood AA, Tahtamouni LH, Balakit AA, Yaseen YS, Al-Hasani RA. New Combretastatin Analogs as Anticancer Agents: Design, Synthesis, Microtubules Polymerization Inhibition, and Molecular Docking Studies. Chem Biodivers 2023; 20:e202201206. [PMID: 36890635 DOI: 10.1002/cbdv.202201206] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 02/16/2023] [Indexed: 03/10/2023]
Abstract
A new series of 4-(4-methoxyphenyl)-5-(3,4,5-trimethoxyphenyl)-4H-1,2,4-triazole-3-thiol derivatives were synthesized as analogs for the anticancer drug combretastatin A-4 (CA-4) and characterized using FT-IR, 1 H-NMR, 13 CNMR, and HR-MS techniques. The new CA-4 analogs were designed to meet the structural requirements of the highest expected anticancer activity of CA-4 analogs by maintaining ring A 3,4,5-trimethoxyphenyl moiety, and at the same time varying the substituents effect of the triazole moiety (ring B). In silico analysis indicated that compound 3 has higher total energy and dipole moment than colchicine and the other analogs, and it has excellent distribution of electron density and is more stable, resulting in an increased binding affinity during tubulin inhibition. Additionally, compound 3 was found to interact with three apoptotic markers, namely p53, Bcl-2, and caspase 3. Compound 3 showed strong similarity to colchicine, and it has excellent pharmacokinetics properties and a good dynamic profile. The in vitro anti-proliferation studies showed that compound 3 is the most cytotoxic CA-4 analog against cancer cells (IC50 of 6.35 μM against Hep G2 hepatocarcinoma cells), and based on its selectivity index (4.7), compound 3 is a cancer cytotoxic-selective agent. As expected and similar to colchicine, compound 3-treated Hep G2 hepatocarcinoma cells were arrested at the G2/M phase resulting in induction of apoptosis. Compound 3 tubulin polymerization IC50 (9.50 μM) and effect on Vmax of tubulin polymerization was comparable to that of colchicine (5.49 μM). Taken together, the findings of the current study suggest that compound 3, through its binding to the colchicine-binding site at β-tubulin, is a promising microtubule-disrupting agent with excellent potential to be used as cancer therapeutic agent.
Collapse
Affiliation(s)
- Shaker A Abdul Hussein
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Babylon, 51001, Babylon, Iraq
| | - Ammar A Razzak Mahmood
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, 10001, Baghdad, Iraq
| | - Lubna H Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, 13133, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, 80523 Colorado, USA
| | - Asim A Balakit
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Babylon, 51001, Babylon, Iraq
| | - Yahya S Yaseen
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Tikrit, 34001, Tikrit, Iraq
| | - Rehab A Al-Hasani
- Department of Chemistry, College of Science, Al-Mustansiriyah University, 10052, Baghdad, Iraq
| |
Collapse
|
4
|
Chalcones: Promising therapeutic agents targeting key players and signaling pathways regulating the hallmarks of cancer. Chem Biol Interact 2023; 369:110297. [PMID: 36496109 DOI: 10.1016/j.cbi.2022.110297] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The need for innovative anticancer treatments with high effectiveness and low toxicity is urgent due to the development of malignancies that are resistant to chemotherapeutic agents and the poor specificity of existing anticancer treatments. Chalcones are 1,3-diaryl-2-propen-1-ones, which are the precursors for flavonoids and isoflavonoids. Chalcones are readily available from a wide range of natural resources and consist of very basic chemical scaffolds. Because the ease with which the synthesis it allows for the production of several chalcone derivatives. Various in-vitro and in-vivo studies indicate that naturally occurring and synthetic chalcone derivatives exhibit promising biological activities against cancer hallmarks such as proliferation, angiogenesis, invasion, metastasis, inflammation, stemness, and regulation of cancer epigenetics. According to their structure and functional groups, chalcones derivatives and their hybrid compounds exert a broad range of biological activities through targeting key elements and signaling molecules relevant to cancer progression. This review will provide valuable insights into the latest updates of chalcone groups as anticancer agents and extensively discuss their underlying molecular mechanisms of action.
Collapse
|
5
|
Hawash M. Recent Advances of Tubulin Inhibitors Targeting the Colchicine Binding Site for Cancer Therapy. Biomolecules 2022; 12:biom12121843. [PMID: 36551271 PMCID: PMC9776383 DOI: 10.3390/biom12121843] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer accounts for numerous deaths each year, and it is one of the most common causes of death worldwide, despite many breakthroughs in the discovery of novel anticancer candidates. Each new year the FDA approves the use of new drugs for cancer treatments. In the last years, the biological targets of anticancer agents have started to be clearer and one of these main targets is tubulin protein; this protein plays an essential role in cell division, as well as in intracellular transportation. The inhibition of microtubule formation by targeting tubulin protein induces cell death by apoptosis. In the last years, numerous novel structures were designed and synthesized to target tubulin, and this can be achieved by inhibiting the polymerization or depolymerization of the microtubules. In this review article, recent novel compounds that have antiproliferation activities against a panel of cancer cell lines that target tubulin are explored in detail. This review article emphasizes the recent developments of tubulin inhibitors, with insights into their antiproliferative and anti-tubulin activities. A full literature review shows that tubulin inhibitors are associated with properties in the inhibition of cancer cell line viability, inducing apoptosis, and good binding interaction with the colchicine binding site of tubulin. Furthermore, some drugs, such as cabazitaxel and fosbretabulin, have been approved by FDA in the last three years as tubulin inhibitors. The design and development of efficient tubulin inhibitors is progressively becoming a credible solution in treating many species of cancers.
Collapse
Affiliation(s)
- Mohammed Hawash
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus P.O. Box 7, Palestine
| |
Collapse
|
6
|
Kuchana V, Kashetti V, Tangeda SJ, Manga V. Design, synthesis and molecular docking study of thiophenyl hydrazone derivatives as tubulin polymerization inhibitors. SYNTHETIC COMMUN 2022. [DOI: 10.1080/00397911.2022.2125324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Vinutha Kuchana
- Department of Chemistry, Sarojini Naidu Pharmacy Maha Vidyalaya, Osmania University, Hyderabad, India
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, India
| | - Vaeshnavi Kashetti
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, India
| | - Saritha Jyotsna Tangeda
- Department of Chemistry, Sarojini Naidu Pharmacy Maha Vidyalaya, Osmania University, Hyderabad, India
| | - Vijjulatha Manga
- Department of Chemistry, University College of Science, Osmania University, Hyderabad, India
| |
Collapse
|
7
|
Ranjan Dwivedi A, Singh Rawat S, Kumar V, Kumar N, Anand P, Prakash Yadav R, Barnwal S, Prasad A, Kumar V. Synthesis and Screening of Novel 4-N-Heterocyclic-2-aryl-6,7,8-trimethoxyquinazolines as Antiproliferative and Tubulin Polymerization Inhibitors. Bioorg Med Chem 2022; 72:116976. [DOI: 10.1016/j.bmc.2022.116976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/04/2022] [Accepted: 08/19/2022] [Indexed: 11/02/2022]
|
8
|
Hu L, Fan M, Shi S, Song X, Wang F, He H, Qi B. Dual target inhibitors based on EGFR: Promising anticancer agents for the treatment of cancers (2017-). Eur J Med Chem 2022; 227:113963. [PMID: 34749202 DOI: 10.1016/j.ejmech.2021.113963] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023]
Abstract
The EGFR family play a significant role in cell signal transduction and their overexpression is implicated in the pathogenesis of numerous human solid cancers. Inhibition of the EGFR-mediated signaling pathways by EGFR inhibitors is a widely used strategy for the treatment of cancers. In most cases, the EGFR inhibitors used in clinic were only effective when the cancer cells harbored specific activating EGFR mutations which appeared to preserve the ligand-dependency of receptor activation but altered the pattern of downstream signaling pathways. Moreover, cancer is a kind of multifactorial disease, and therefore manipulating a single target may result in treatment failure. Although drug combinations for the treatment of cancers proved to be successful, the use of two or more drugs concurrently still was a challenge in clinical therapy owing to various dose-limiting toxicities and drug-drug interactions caused by pharmacokinetic profiles changed. Therefore, a single drug targeting two or multiple targets could serve as an effective strategy for the treatment of cancers. In recent, drugs with diverse pharmacological effects have been shown to be more advantageous than combination therapies due to their lower incidences of side effects and more resilient therapies. Accordingly, dual target-single-agent strategy has become a popular field for cancer treatment, and researchers became more and more interest in the development of novel dual-target drugs in recent years. In this review, we briefly introduce the EGFR family proteins and synergisms between EGFR and other anticancer targets, and summarizes the development of potential dual target inhibitors based on wild-type and/or mutant EGFR for the treatment of solid cancers in the past five years. Additionally, the rational design and SARs of these dual target agents are also presented in detailed, which will lay a significant foundation for the further development of novel EGFR-based dual inhibitors with excellent druggability.
Collapse
Affiliation(s)
- Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| | - Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| |
Collapse
|
9
|
Ibrahim TS, Hawwas MM, Malebari AM, Taher ES, Omar AM, Neamatallah T, Abdel-Samii ZK, Safo MK, Elshaier YAMM. Discovery of novel quinoline-based analogues of combretastatin A-4 as tubulin polymerisation inhibitors with apoptosis inducing activity and potent anticancer effect. J Enzyme Inhib Med Chem 2021; 36:802-818. [PMID: 33730937 PMCID: PMC7993375 DOI: 10.1080/14756366.2021.1899168] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/27/2021] [Accepted: 02/27/2021] [Indexed: 02/08/2023] Open
Abstract
A new series of quinoline derivatives of combretastatin A-4 have been designed, synthesised and demonstrated as tubulin polymerisation inhibitors. These novel compounds showed significant antiproliferative activities, among them, 12c exhibited the most potent inhibitory activity against different cancer cell lines (MCF-7, HL-60, HCT-116 and HeLa) with IC50 ranging from 0.010 to 0.042 µM, and with selectivity profile against MCF-10A non-cancer cells. Further mechanistic studies suggest that 12c can inhibit tubulin polymerisation and cell migration, leading to G2/M phase arrest. Besides, 12c induces apoptosis via a mitochondrial-dependant apoptosis pathway and caused reactive oxygen stress generation in MCF-7 cells. These results provide guidance for further rational development of potent tubulin polymerisation inhibitors for the treatment of cancer.HighlightsA novel series of quinoline derivatives of combretastatin A-4 have been designed and synthesised.Compound 12c showed significant antiproliferative activities against different cancer cell lines.Compound 12c effectively inhibited tubulin polymerisation and competed with [3H] colchicine in binding to tubulin.Compound 12c arrested the cell cycle at G2/M phase, effectively inducing apoptosis and inhibition of cell migration.
Collapse
Affiliation(s)
- Tarek S. Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed M. Hawwas
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Azizah M. Malebari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ehab S. Taher
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | - Abdelsattar M. Omar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Thikryat Neamatallah
- Department of Pharmacology and toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zakaria K. Abdel-Samii
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Martin K. Safo
- Institute for Structural Biology, Drug Discovery and Development, Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Yaseen A. M. M. Elshaier
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt
| |
Collapse
|
10
|
Ferreira LOA, Molnár S, Martins FT, Perjési P, Keng Queiroz LH, Noda-Perez C. Michael addition reaction of symmetrically halogenated bischalcones with reduced glutathione assessed by RP-HPLC and RP-HPLC-ESI-MS. Microchem J 2021. [DOI: 10.1016/j.microc.2021.106603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
11
|
Xue J, Wu G, Ejaz U, Akhtar F, Wan X, Zhu Y, Geng A, Chen Y, He S. A novel histone deacetylase inhibitor LT-548-133-1 induces apoptosis by inhibiting HDAC and interfering with microtubule assembly in MCF-7 cells. Invest New Drugs 2021; 39:1222-1231. [PMID: 33788074 DOI: 10.1007/s10637-021-01102-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/12/2021] [Indexed: 12/29/2022]
Abstract
Many studies have indicated that histone deacetylase inhibitors (HDACis) have a significant antitumor effect in cancer. Here we report a compound named LT-548-133-1 that not only acts as an HDAC inhibitor but also interferes with microtubule assembly to inhibit MCF-7 cell proliferation and induce apoptosis. Consistent with Chidamide, LT-548-133-1 inhibited HDAC activity and increased histone H3 acetylation. But the difference is that it significantly induced cell cycle G2/M arrest while Chidamide caused G0/G1 arrest in MCF-7 cells. By Western blotting, we found the accumulation of CyclinB1 and phosphorylated histone H3 in LT-548-133-1 treated cells. Immunofluorescence based microtubule-repolymerization experiments and immunofluorescence staining of cell microtubules and nuclei showed that LT-548-133-1inhibited microtubule-repolymerization and induced mitotic abnormalities. The decreased expression of Bcl-2 and the increased expression of Bax, p53, p21, and cleaved-Caspase3 indicated the occurrence of apoptosis. Flow cytometry results also showed an increase in the proportion of apoptotic cells after administration of LT-548-133-1 or Chidamide. Therefore, we demonstrated that LT-548-133-1 could act as an HDAC inhibitor while inhibiting microtubule-repolymerization, causing mitosis to be arrested in G2/M. These two effects ultimately lead to proliferation inhibition and apoptosis of MCF-7 cells.
Collapse
Affiliation(s)
- Jinbing Xue
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Gang Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Umer Ejaz
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Fahad Akhtar
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.,State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, S2-316 Building 2, West Beichan Road, Chaoyang District, Beijing, 100101, China.,School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xinyu Wan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Yong Zhu
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Aixing Geng
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Yadong Chen
- School of Science, China Pharmaceutical University, Nanjing, 210009, China.
| | - Shuying He
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
12
|
|
13
|
Ciccolini C, De Crescentini L, Mantellini F, Mari G, Santeusanio S, Favi G. Construction of Unusual Indole-Based Heterocycles from Tetrahydro-1 H-pyridazino[3,4- b]indoles. Molecules 2020; 25:molecules25184124. [PMID: 32916997 PMCID: PMC7571100 DOI: 10.3390/molecules25184124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 11/21/2022] Open
Abstract
Herein, we report the successful syntheses of scarcely represented indole-based heterocycles which have a structural connection with biologically active natural-like molecules. The selective oxidation of indoline nucleus to indole, hydrolysis of ester and carbamoyl residues followed by decarboxylation with concomitant aromatization of the pyridazine ring starting from tetrahydro-1H-pyridazino[3,4-b]indole derivatives lead to fused indole-pyridazine compounds. On the other hand, non-fused indole-pyrazol-5-one scaffolds are easily prepared by subjecting the same C2,C3-fused indoline tetrahydropyridazines to treatment with trifluoroacetic acid (TFA). These methods feature mild conditions, easy operation, high yields in most cases avoiding the chromatographic purification, and broad substrate scope. Interestingly, the formation of indole linked pyrazol-5-one system serves as a good example of the application of the umpolung strategy in the synthesis of C3-alkylated indoles.
Collapse
|
14
|
Donthiboina K, Anchi P, Gurram S, Sai Mani G, Lakshmi Uppu J, Godugu C, Shankaraiah N, Kamal A. Synthesis and biological evaluation of substituted N-(2-(1H-benzo[d]imidazol-2-yl)phenyl)cinnamides as tubulin polymerization inhibitors. Bioorg Chem 2020; 103:104191. [PMID: 32891862 DOI: 10.1016/j.bioorg.2020.104191] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 01/23/2023]
Abstract
A new series of N-(2-(1H-benzo[d]imidazol-2-yl)phenyl) cinnamides was prepared and evaluated for their in vitro cytotoxic activity using various cancer cell lines viz. A549 (human non-small cell lung cancer), MDA-MB-231 (human triple negative breast cancer), B16-F10 (mouse melanoma), BT-474 (human breast cancer), and 4 T1 (mouse triple negative breast cancer). In the series of tested compounds, 12h showed potent cytotoxic activity against non-small cell lung cancer cell line with IC50 value of 0.29 ± 0.02 µM. The cytoxicity of most potent compound 12h was also tested on NRK-52E (normal rat kidney epithelial cell line) and showed less cytotoxicity compared to cancer cells. Tubulin polymerization assay indicated that the compound 12h was able to impede the cell division by inhibiting tubulin polymerization. Moreover, molecular docking study also suggested the binding of 12h at the colchicine-binding site of the tubulin protein. Cell cycle analysis revealed that the compound 12h arrests G2/M phase. In addition, 12h induced apoptosis in A549 cell lines was evaluated by various staining studies like acridine orange, DAPI, analysis of mitochondrial membrane potential, annexin V-FITC, and DCFDA assays.
Collapse
Affiliation(s)
- Kavitha Donthiboina
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Pratibha Anchi
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Sowmyasree Gurram
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Geeta Sai Mani
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Jaya Lakshmi Uppu
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India
| | - Chandraiah Godugu
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India.
| | - Ahmed Kamal
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, India; School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
15
|
Discovery of isopenicin A, a meroterpenoid as a novel inhibitor of tubulin polymerization. Biochem Biophys Res Commun 2020; 525:303-307. [DOI: 10.1016/j.bbrc.2020.02.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 02/04/2020] [Indexed: 01/09/2023]
|
16
|
Zhan S, Yan C. Efficient Synthesis of Functionalized 6‐(2‐Oxoindolin‐3‐yl)‐5‐azaspiro[2.4]heptanes. ChemistrySelect 2019. [DOI: 10.1002/slct.201902785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Shao‐Cong Zhan
- School of Chemistry & Chemical EngineeringYangzhou University Yangzhou 225002 China
| | - Chao‐Guo Yan
- School of Chemistry & Chemical EngineeringYangzhou University Yangzhou 225002 China
| |
Collapse
|
17
|
Duan Y, Liu W, Tian L, Mao Y, Song C. Targeting Tubulin-colchicine Site for Cancer Therapy: Inhibitors, Antibody- Drug Conjugates and Degradation Agents. Curr Top Med Chem 2019; 19:1289-1304. [PMID: 31210108 DOI: 10.2174/1568026619666190618130008] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/22/2019] [Accepted: 05/01/2019] [Indexed: 12/14/2022]
Abstract
Microtubules are essential for the mitotic division of cells and have been an attractive target
for antitumour drugs due to the increased incidence of cancer and significant mitosis rate of tumour cells.
In the past few years, tubulin-colchicine binding site, as one of the three binding pockets including taxol-,
vinblastine- and colchicine-binding sites, has been focused on to design tubulin-destabilizing agents including
inhibitors, antibody-drug conjugates and degradation agents. The present review is the first to
cover a systemic and recent synopsis of tubulin-colchicine binding site agents. We believe that it would
provide an increase in our understanding of receptor-ligand interaction pattern and consciousness of a
series of challenges about tubulin target druggability.
Collapse
Affiliation(s)
- Yongtao Duan
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Wei Liu
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Liang Tian
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yanna Mao
- Henan Provincial Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Chuanjun Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Zhengzhou, 450001, China
| |
Collapse
|
18
|
Schmid M, Grossmann AS, Mayer P, Müller T, Magauer T. Ring-Expansion Approaches for the Total Synthesis of Salimabromide. Tetrahedron 2019; 75:3195-3215. [PMID: 31160829 PMCID: PMC6544523 DOI: 10.1016/j.tet.2019.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
We describe the evolution of a synthetic strategy for the construction of the marine polyketide salimabromide. Combining a bicyclo[3.1.0]hexan-2-one ring-expansion to build up a functionalized naphthalene and an unprecedented rearrangement/cyclization cascade, enabled synthesis of a dearomatized tricyclic subunit of the target compound. Alternatively, an intramolecular ketiminium [2+2]-cycloaddition and subsequent Baeyer-Villiger ring-expansion gave access to the sterically encumbered architecture of salimabromide. Sequential oxidation of the carbon framework finally enabled the total synthesis of this unusual natural product.
Collapse
Affiliation(s)
- Matthias Schmid
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstraße 5-13, Munich 81377, Germany
- Institute of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Adriana S Grossmann
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstraße 5-13, Munich 81377, Germany
| | - Peter Mayer
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University Munich, Butenandtstraße 5-13, Munich 81377, Germany
| | - Thomas Müller
- Institute of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| | - Thomas Magauer
- Institute of Organic Chemistry and Center for Molecular Biosciences, University of Innsbruck, Innrain 80-82, 6020 Innsbruck, Austria
| |
Collapse
|
19
|
Anti-tubulin agents of natural origin: Targeting taxol, vinca, and colchicine binding domains. Eur J Med Chem 2019; 171:310-331. [PMID: 30953881 DOI: 10.1016/j.ejmech.2019.03.025] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/09/2019] [Accepted: 03/10/2019] [Indexed: 12/24/2022]
Abstract
Microtubules are a protein which is made of α- and β-heterodimer. It is one of the main components of the cell which play a vital role in cell division especially in G2/M-phase. It exists in equilibrium dynamic of polymerization and depolymerization of α- and β-heterodimer. It is one of the best targets for developing anti-cancer drugs. Various natural occurring molecules are well known for their anti-tubulin effect such as vinca, paclitaxel, combretastatin, colchicine etc. These microtubule-targeted drugs are acted through two processes (i) inhibiting depolymerization of tubulin (tubulin stabilizing agents) and (ii) inhibiting polymerization of tubulin (tubulin destabilizing agents). Now days, various binding domains have been explore through which these molecules are binding to tubulin but the three major binding domain of tubulin are taxol, vinca and colchicine binding domain. The present article mainly focus on the classification of various naturally occurring compounds on the basis of their inhibition processes (depolymerization and polymerization) and the site of interaction (targets taxol, vinca and colchicine binding domain) which has been hitherto reported. By placing all the naturally occurring taxol, vinca and colchicine binding site analogues at one place makes a better understanding of the tubulin interactions with known natural tubulin binders that would helps in the discovery of new and potent natural, semi-synthetic and synthetic analogues for treating cancer.
Collapse
|
20
|
Lu S, Obianom ON, Ai Y. Novel hybrids derived from aspirin and chalcones potently suppress colorectal cancer in vitro and in vivo. MEDCHEMCOMM 2018; 9:1722-1732. [PMID: 30429977 DOI: 10.1039/c8md00284c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/23/2018] [Indexed: 01/06/2023]
Abstract
Colorectal cancer (CRC) remains the fourth leading cause of cancer deaths around the world despite the availability of many approved small molecules for treatment. The issues lie in the potency, selectivity and targeting of these compounds. Therefore, new strategies and targets are needed to optimize and develop novel treatments for CRC. Here, a group of novel hybrids derived from aspirin and chalcones were designed and synthesized based on recent reports of their individual benefits to CRC targeting and selectivity. The most active compound 7h inhibited proliferation of CRC cell lines with better potency compared to 5-fluorouracil, a currently used therapeutic agent for CRC. Importantly, 7h had 8-fold less inhibitory activity against non-cancer CCD841 cells. In addition, 7h inhibited CRC growth via the inhibition of the cell cycle in the G1 phase. Furthermore, 7h induced apoptosis by activating caspase 3 and PARP cleavage, as well as increasing ROS in CRC cells. Finally, 7h significantly retarded the CRC cell growth in a mouse xenograft model. These findings suggest that 7h may have potential to treat CRC.
Collapse
Affiliation(s)
- Shan Lu
- College of Pharmacy , Hubei University of Chinese Medicine , Hubei 430065 , PR China .
| | - Obinna N Obianom
- Department of Pharmaceutical Sciences , University of Maryland School of Pharmacy , Baltimore , MD 21201 , USA .
| | - Yong Ai
- Department of Pharmaceutical Sciences , University of Maryland School of Pharmacy , Baltimore , MD 21201 , USA .
| |
Collapse
|
21
|
Li L, Jiang S, Li X, Liu Y, Su J, Chen J. Recent advances in trimethoxyphenyl (TMP) based tubulin inhibitors targeting the colchicine binding site. Eur J Med Chem 2018; 151:482-494. [PMID: 29649743 DOI: 10.1016/j.ejmech.2018.04.011] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 12/16/2022]
Abstract
Microtubules (composed of α- and β-tubulin heterodimers) play a pivotal role in mitosis and cell division, and are regarded as an excellent target for chemotherapeutic agents to treat cancer. There are four unique binding sites in tubulin to which taxanes, vinca alkaloids, laulimalide and colchicine bind respectively. While several tubulin inhibitors that bind to the taxane or vinca alkaloid binding sites have been approved by FDA, currently there are no FDA approved tubulin inhibitors targeting the colchicine binding site. Tubulin inhibitors that bind to the colchicine binding site have therapeutic advantages over taxanes and vinca alkaloids, for example, they can be administered orally, have less drug-drug interaction potential, and are less prone to develop multi-drug resistance. Typically, tubulin inhibitors that bind to the colchicine binding site bear the trimethoxyphenyl (TMP) moiety which is essential for interaction with tubulin. Over the last decade, a variety of molecules bearing the TMP moiety have been designed and synthesized as tubulin inhibitors for cancer treatment. In this review, we focus on the TMP analogs that are designed based on CA-4, indole, chalcone, colchicine and natural product scaffolds which are known to interact with the colchicine binding site in tubulin. The challenges and future direction of the TMP based tubulin inhibitors are also discussed in detail.
Collapse
Affiliation(s)
- Ling Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Sibo Jiang
- College of Pharmacy, University of Florida, Orlando, FL 32827, USA
| | - Xiaoxun Li
- Chengdu Easton Biopharmaceuticals Co., Ltd., Chengdu 611731, China
| | - Yao Liu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
22
|
Design, Synthesis and Cytotoxic Evaluation of Novel Chalcone Derivatives Bearing Triazolo[4,3-a]-quinoxaline Moieties as Potent Anticancer Agents with Dual EGFR Kinase and Tubulin Polymerization Inhibitory Effects. Molecules 2017; 23:molecules23010048. [PMID: 29280968 PMCID: PMC5943945 DOI: 10.3390/molecules23010048] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/20/2017] [Accepted: 12/22/2017] [Indexed: 12/03/2022] Open
Abstract
A series of hybrid of triazoloquinoxaline-chalcone derivatives 7a–k were designed, synthesized, fully characterized, and evaluated for their cytotoxic activity against three target cell lines: human breast adenocarcinoma (MCF-7), human colon carcinoma (HCT-116), and human hepatocellular carcinoma (HEPG-2). The preliminary results showed that some of these chalcones like 7b–c, and 7e–g exhibited significant antiproliferative effects against most of the cell lines, with selective or non-selective behavior, indicated by IC50 values in the 1.65 to 34.28 µM range. In order to investigate the mechanistic aspects of these active compounds, EGFR TK and tubulin inhibitory activities were measured as further biological assays. The EGFR TK assay results revealed that the derivatives 7a–c, 7e, and 7g could inhibit the EGFR TK in the submicromolar range (0.093 to 0.661 µM). Moreover, an antitubulin polymerization effect was noted for the active derivatives compared to the reference drug colchicine, with compounds 7e and 7g displaying 14.7 and 8.4 micromolar activity, respectively. Furthermore, a molecular docking study was carried out to explain the observed effects and the binding modes of these chalcones with the EGFR TK and tubulin targets.
Collapse
|
23
|
Zhu C, Wang R, Zheng W, Chen D, Yue X, Cao Y, Qin W, Sun H, Wang Y, Liu Z, Li B, Du J, Bu X, Zhou B. Synthesis and evaluation of anticancer activity of BOC26P, an ortho-aryl chalcone sodium phosphate as water-soluble prodrugs in vitro and in vivo. Biomed Pharmacother 2017; 96:551-562. [PMID: 29032339 DOI: 10.1016/j.biopha.2017.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 09/14/2017] [Accepted: 10/02/2017] [Indexed: 12/14/2022] Open
Abstract
Major limitations of chalcones as clinical anticancer agents are water insolubility and poor bioavailability, which may be improved by a classic phosphate prodrug strategy that targets non-specific alkaline phosphatase (ALP) for releasing the parent drug in vivo. In this study, we found that BOC26P, a phosphate prodrug of chalcone OC26, exhibits excellent water solubility and improved plasma concentration in vivo by either i.v. or p.o. compared with the parent drug. In pace with decreased inhibitory activity of BOC26P against microtubule polymerization in vitro and in cells, the antiproliferative activity of BOC26P is attenuated in A549 and HLF cells. However, the antitumor effect of BOC26P increases in an A549 xenograft model as compared to the equimolar concentration of OC26, suggesting that complex tumor microenvironment would be another important influence factor to regulate the antitumor activity of BOC26Pin vivo. In conclusion, these observations showed that the traditional phosphate prodrug strategy would be a promising and easy method to increase water solubility and anticancer activity of chalcones for the clinical developments of anticancer agents.
Collapse
Affiliation(s)
- Cuige Zhu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Ruimin Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China; Department of Nuclear Medicine, Guangzhou General Hospital of Guangzhou Military Command Guangzhou, Guangdong, 510010, PR China
| | - Weichao Zheng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Daoyuan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xin Yue
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yingnan Cao
- School of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, PR China
| | - Wenjing Qin
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Haixia Sun
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Youqiao Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Ziyi Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Baojian Li
- Shenzhen Davoos tech. Ltd.Co., Room A611, Silver star tech. building, 1301 Guanguang Road, Guanlan, Longhua District, Shenzhen, PR China
| | - Jun Du
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xianzhang Bu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Binhua Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
24
|
Zhou Y, Yan W, Cao D, Shao M, Li D, Wang F, Yang Z, Chen Y, He L, Wang T, Shen M, Chen L. Design, synthesis and biological evaluation of 4-anilinoquinoline derivatives as novel potent tubulin depolymerization agents. Eur J Med Chem 2017; 138:1114-1125. [DOI: 10.1016/j.ejmech.2017.07.040] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/19/2017] [Accepted: 07/20/2017] [Indexed: 12/24/2022]
|
25
|
Singh H, Singh JV, Gupta MK, Saxena AK, Sharma S, Nepali K, Bedi PMS. Triazole tethered isatin-coumarin based molecular hybrids as novel antitubulin agents: Design, synthesis, biological investigation and docking studies. Bioorg Med Chem Lett 2017; 27:3974-3979. [DOI: 10.1016/j.bmcl.2017.07.069] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 07/15/2017] [Accepted: 07/27/2017] [Indexed: 12/17/2022]
|
26
|
Hu F, Chen H, Zhang M, Yu S, Xu X, Yuan W, Zhang X. Catalyst-free Synthesis of Spiro[indoline-3,1′-pyrazolo[5,1-a]isoquinolines]viaDiastereoselective 1,3-dipolar Cycloaddition under Mild Conditions. J Heterocycl Chem 2017. [DOI: 10.1002/jhet.2903] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Fangzhi Hu
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Hui Chen
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Minmin Zhang
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Shuowen Yu
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Xiaoying Xu
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
| | - Weicheng Yuan
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
| | - Xiaomei Zhang
- Key Laboratory for Asymmetric Synthesis & Chirotechnology of Sichuan Province, Chengdu Institute of Organic Chemistry; Chinese Academy of Sciences; Chengdu 610041 China
| |
Collapse
|
27
|
Guggilapu SD, Lalita G, Reddy TS, Prajapti SK, Nagarsenkar A, Ramu S, Brahma UR, Lakshmi UJ, Vegi GMN, Bhargava SK, Babu BN. Synthesis of C 5 -tethered indolyl-3-glyoxylamide derivatives as tubulin polymerization inhibitors. Eur J Med Chem 2017; 128:1-12. [DOI: 10.1016/j.ejmech.2017.01.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 01/19/2017] [Accepted: 01/19/2017] [Indexed: 01/19/2023]
|
28
|
Wang Y, Hedblom A, Koerner SK, Li M, Jernigan FE, Wegiel B, Sun L. Novel synthetic chalcones induce apoptosis in the A549 non-small cell lung cancer cells harboring a KRAS mutation. Bioorg Med Chem Lett 2016; 26:5703-5706. [PMID: 27810244 DOI: 10.1016/j.bmcl.2016.10.063] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 10/19/2016] [Accepted: 10/21/2016] [Indexed: 12/12/2022]
Abstract
A series of novel chalcones were synthesized by the Claisen-Schmidt condensation reaction of tetralones and 5-/6-indolecarboxaldehydes. Treatment of human lung cancer cell line harboring KRAS mutation (A549) with the chalcones induced dose-dependent apoptosis. Cell cycle analyses and Western blotting suggested the critical role of the chalcones in interrupting G2/M transition of cell cycle. SAR study demonstrated that substituent on the indole N atom significantly affects the anticancer activity of the chalcones, with methyl and ethyl providing the more active compounds (EC50: 110-200nM), Compound 1g was found to be >4-fold more active in the A549 cells (EC50: 110nM) than in prostate (PC3) or pancreatic cancer (CLR2119, PAN02) cells. Furthermore, compound 1l selectively induced apoptosis of lung cancer cells A549 (EC50: 0.55μM) but did not show measurable toxicity in the normal lung bronchial epithelial cells (hBEC) at doses as high as 10μM, indicating specificity towards cancer cells.
Collapse
Affiliation(s)
- Yiqiang Wang
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Andreas Hedblom
- Transplant Institute & Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Steffi K Koerner
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mailin Li
- Transplant Institute & Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Finith E Jernigan
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Barbara Wegiel
- Transplant Institute & Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - Lijun Sun
- Center for Drug Discovery and Translational Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
29
|
Indole molecules as inhibitors of tubulin polymerization: potential new anticancer agents, an update (2013–2015). Future Med Chem 2016; 8:1291-316. [DOI: 10.4155/fmc-2016-0047] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Discovery of new indole-based tubulin polymerization inhibitors will continue to dominate the synthetic efforts of many medicinal chemists working in the field. The indole ring system is an essential part of several tubulin inhibitors identified in the recent years. The present review article will update the synthesis, anticancer and tubulin inhibition activities of several important new indole classes such as 2-phenylindoles (28, 29 & 30), oxindoles (35 & 38), indole-3-acrylamides (44), indolines (46), aroylindoles (49), carbozoles (75, 76 & 82), azacarbolines (87) and annulated indoles (100–105).
Collapse
|
30
|
Mirzaei H, Emami S. Recent advances of cytotoxic chalconoids targeting tubulin polymerization: Synthesis and biological activity. Eur J Med Chem 2016; 121:610-639. [PMID: 27318983 DOI: 10.1016/j.ejmech.2016.05.067] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 05/28/2016] [Accepted: 05/30/2016] [Indexed: 02/07/2023]
Abstract
Since microtubules have an important role in mitosis and other vital cellular functions, tubulin-targeting chemotherapy has been received growing attention in anticancer drug design and development. It was found that a number of naturally occurring compounds including distinct chalcones exert their effect by inhibition of tubulin polymerization. After the identification of tubulin polymerization as potential target for chalcone-type compounds, extensive researches have been made to design and synthesis of new anti-tubulin chalconoids. Although diverse chalcones have found to be potent anticancer agents but in the present review, we focused on the recently reported tubulin polymerization inhibitors from chalcone origin and related synthetic compounds, and their detailed synthetic methods and biological activities.
Collapse
Affiliation(s)
- Hassan Mirzaei
- Student Research Committee, Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
31
|
Singh H, Kumar M, Nepali K, Gupta MK, Saxena AK, Sharma S, Bedi PMS. Triazole tethered C5-curcuminoid-coumarin based molecular hybrids as novel antitubulin agents: Design, synthesis, biological investigation and docking studies. Eur J Med Chem 2016; 116:102-115. [PMID: 27060762 DOI: 10.1016/j.ejmech.2016.03.050] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 11/15/2022]
Abstract
Keeping in view the confines allied with presently accessible antitumor agents and success of C5-curcuminoid based bifunctional hybrids as novel antitubulin agnets, molecular hybrids of C5-curcuminoid and coumarin tethered by triazole ring have been synthesized and investigated for in-vitro cytotoxicity against THP-1, COLO-205, HCT-116 and PC-3 human tumor cell lines. The results revealed that the compounds A-2 to A-9, B-2, B-3, B-7 showed significant cytotoxic potential against THP-1, COLO-205 and HCT-116 cell lines, while the PC-3 cell line among these was found to be almost resistant. Structure activity relationship revealed that the nature of Ring X and the length of carbon-bridge (n) connecting triazole ring with coumarin moiety considerably influence the activity. Methoxy substituted phenyl ring as Ring X and two carbon-bridges were found to be the ideal structural features. The most potent compounds (A-2, A-3 and A-7) were further tested for tubulin polymerization inhibition. Compound A-2 was found to significantly inhibit the tubulin polymerization (IC50 = 0.82 μM in THP-1 tumor cells). The significant cytotoxicity and tubulin polymerization inhibition by A-2 was further rationalized by docking studies where it was docked at the curcumin binding site of tubulin.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Mandeep Kumar
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Kunal Nepali
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Manish K Gupta
- Lloyd Institute of Management and Technology, Greater Noida, UP, India
| | - Ajit K Saxena
- Indian Institute of Integrative Medicine, Jammu, India
| | - Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India.
| | - Preet Mohinder S Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| |
Collapse
|
32
|
Sharma S, Gupta MK, Saxena AK, Bedi PMS. Triazole linked mono carbonyl curcumin-isatin bifunctional hybrids as novel anti tubulin agents: Design, synthesis, biological evaluation and molecular modeling studies. Bioorg Med Chem 2015; 23:7165-80. [PMID: 26515041 DOI: 10.1016/j.bmc.2015.10.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/06/2015] [Accepted: 10/11/2015] [Indexed: 12/13/2022]
Abstract
Keeping in view the limitations associated with currently available anticancer drugs, molecular hybrids of mono carbonyl curcumin and isatin tethered by triazole ring have been synthesized and evaluated for in vitro cytotoxicity against THP-1, COLO-205, HCT-116, A549, HeLa, CAKI-I, PC-3, MiaPaca-2 human cancer cell lines. The results revealed that the compounds SA-1 to SA-9, SB-2, SB-3, SB-4, SB-7 and SC-2 showed a good range of IC50 values against THP-1, COLO-205, HCT-116 and PC-3 cell lines, while the other four cell lines among these were found to be almost resistant. Structure activity relationship revealed that the nature of Ring X and substitution at position R influences the activity. Methoxy substituted phenyl ring as Ring X and H as R were found to be the ideal structural features. The most potent compounds (SA-2, SA-3, SA-4, SA-7) were further tested for tubulin inhibition. Compound SA-2 was found to significantly inhibit the tubulin polymerization (IC50=1.2 μM against HCT-116). Compound SA-2, moreover, lead to the disruption of microtubules as confirmed by immunofluorescence technique. The significant cytotoxicity and tubulin inhibition by SA-2 was streamlined by molecular modeling studies where it was docked at the curcumin binding site of tubulin.
Collapse
Affiliation(s)
- Sahil Sharma
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| | - Manish K Gupta
- Lloyd Institute of Management and Technology, Greater Noida, UP, India
| | - Ajit K Saxena
- Indian Institute of Integrative Medicine, Jammu, India
| | - Preet Mohinder S Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, Punjab 143005, India
| |
Collapse
|