1
|
Tran C, Hamze A. Recent Advancements in the Development of HDAC/Tubulin Dual-Targeting Inhibitors. Pharmaceuticals (Basel) 2025; 18:341. [PMID: 40143119 PMCID: PMC11945613 DOI: 10.3390/ph18030341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Histone deacetylases (HDACs) have become one of the main targets in cancer therapy due to their involvement in various biological processes, including gene regulation, cell proliferation, and differentiation. Microtubules, as key elements of the cell cytoskeleton, also represent important therapeutic targets in anticancer drugs research. These proteins are involved in diverse cellular functions, especially mitosis, cell signaling, and intracellular trafficking. With the emergence of multi-target therapy during the last decades, the combination of HDAC and tubulin inhibitors has been envisioned as a practical approach for optimizing the therapeutic efficacy of antitumor molecules. HDAC/tubulin dual-targeting inhibitors offer the advantages of the synergistic action of both compounds, along with a significant decrease in their respective toxicities and drug resistance. This review will detail the major recent advancements in the development of HDAC/tubulin dual inhibitors over the last decade and their impact on anticancer drugs discovery.
Collapse
Affiliation(s)
- Christine Tran
- BioCIS, CNRS (Centre National de Recherche Scientifique), Université Paris-Saclay, 91400 Orsay, France
| | - Abdallah Hamze
- BioCIS, CNRS (Centre National de Recherche Scientifique), Université Paris-Saclay, 91400 Orsay, France
| |
Collapse
|
2
|
Lai Q, Wang Z, Wu C, Zhang R, Li L, Tao Y, Mo D, Zhang J, Gou L, Wang Y. Design, synthesis, and antitumor evaluation of quinazoline-4-tetrahydroquinoline chemotypes as novel tubulin polymerization inhibitors targeting the colchicine site. Eur J Med Chem 2025; 283:117139. [PMID: 39662284 DOI: 10.1016/j.ejmech.2024.117139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/13/2024] [Accepted: 11/24/2024] [Indexed: 12/13/2024]
Abstract
We designed, synthesized, and evaluated the antitumor activity of a series of novel quinazoline-4-(6-methoxytetrahydroquinoline) analogues. Among the tested compounds, 4a4 exhibited the most potent antiproliferative activities across four human cancer cell lines with half-maximal inhibitory concentration (IC50) values ranging from 0.4 to 2.7 nM, more potent than the lead compound. The 2.71 Å resolution co-crystal structure of 4a4 with tubulin (PDB code: 8YER) confirmed its critical binding at the colchicine site. Moreover, 4a4 inhibited the polymerization of tubulin, colony formation, and tumor cell migration, while inducing G2/M phase arrest and apoptosis. In vivo, 4a4 significantly delayed primary tumor growth in the SKOV3 xenograft model without obvious side effect. Our research enhances the structure-activity relationships (SARs) understanding of the quinazoline-4-tetrahydroquinoline scaffold and provides new insights for potential structural optimization and the development of novel colchicine binding site inhibitors (CBSIs).
Collapse
Affiliation(s)
- Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhijia Wang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruofei Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Leyan Li
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China; Course of Biological Sciences, Department of Life Science, Imperial College London, United Kingdom
| | - Yiran Tao
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Dan Mo
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jifa Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, Chinese Evidence-based Medicine Center, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, West China Hospital, Sichuan University, Chengdu, 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
3
|
Li H, Yu J, Yu G, Cheng S, Wu H, Wei J, You C, Liu K, Wang M, Meng X, Xu G, Luo H, Xu B. Design and synthesis of N-aryl-2-trifluoromethyl-quinazoline-4-amine derivatives as potential Werner-dependent antiproliferative agents. Mol Divers 2025; 29:195-214. [PMID: 38739229 DOI: 10.1007/s11030-024-10844-6] [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: 01/04/2024] [Accepted: 03/08/2024] [Indexed: 05/14/2024]
Abstract
To discover new Werner (WRN) helicase inhibitors, a series of N-aryl-2-trifluoromethyl-quinazoline-4-amine derivatives were designed and synthesized through a structural optimization strategy, and the anticancer activities of 25 new target compounds against PC3, K562, and HeLa cell lines were evaluated by the MTT assay. Some of these compounds exhibited excellent inhibitory activity against three different cancer cell lines. Compounds 6a, 8i, and 13a showed better antiproliferative activity against K562 cells, with IC50 values of 3871.5, 613.6 and 134.7 nM, respectively, than did paclitaxel (35.6 nM), doxorubicin (2689.0 nM), and NSC 617145 (20.3 nM). To further verify whether the antiproliferative activity of these compounds is dependent on WRN, PC3 cells overexpressing WRN (PC3-WRN) were constructed to further study their antiproliferative potency in vitro, and the inhibition ratio and IC20 values showed that compounds 6a, 8i, and 13a were more sensitive to PC3-WRN than were the control group cells (PC3-NC). The IC20 ratios of compounds 6a, 8i, and 13a to PC3-NC and PC3-WRN were 94.3, 153.4 and 505.5, respectively. According to the docking results, the compounds 6a, 8i, and 13a overlapped well with the binding pocket of 6YHR. Further study demonstrated that among the tested compounds, 13a was the most sensitive to PC3-WRN. In summary, our research identified a series of N-aryl-2-trifluoromethyl-quinazoline-4-amine derivatives as potential WRN-dependent anticancer agents.
Collapse
Affiliation(s)
- Huimin Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
- School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Hui Wu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Jiaomei Wei
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Chang You
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Menghan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
- College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Xueling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China
| | - Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province, Guiyang, 550014, China.
| |
Collapse
|
4
|
Kharbanda S, Alkhamayseh O, Eastham G, Weaver JD. Synthesis of Functionalized Benzocycloheptene Analogs. ESSENTIAL CHEM 2025; 2:1-8. [PMID: 40248686 PMCID: PMC12002400 DOI: 10.1080/28378083.2024.2394899] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/14/2024] [Indexed: 04/19/2025]
Abstract
The benzocycloheptene core is structurally important motif that appears within a wide array of natural products, and benzocycloheptene has recently been demonstrated to undergo [3+2] photo-sensitized cycloaddition reactions. Thus, there is a need for efficient synthetic protocols to access this motif. Herein, we provide efficient strategies to achieve highly functionalized benzocycloheptene derivatives starting from commercially available 1-benzosuberone and 1-tetralone. We anticipate that these benzocycloheptene compounds will serve as excellent precursors for a diverse array of chemical reactions.
Collapse
Affiliation(s)
- Shivangi Kharbanda
- Department of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Osaid Alkhamayseh
- Department of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Georgia Eastham
- Department of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| | - Jimmie D. Weaver
- Department of Chemistry, Oklahoma State University, Stillwater, Oklahoma 74078, United States
| |
Collapse
|
5
|
Anisimov MN, Boichenko MA, Shorokhov VV, Borzunova JN, Janibekova M, Mustyatsa VV, Lifshits IA, Plodukhin AY, Andreev IA, Ratmanova NK, Zhokhov SS, Tarasenko EA, Ipatova DA, Pisarev AR, Vorobjev IA, Trushkov IV, Ivanova OA, Gudimchuk NB. Synthesis and evaluation of tetrahydropyrrolo[1,2- a]quinolin-1(2 H)-ones as new tubulin polymerization inhibitors. RSC Med Chem 2024; 16:d4md00541d. [PMID: 39464648 PMCID: PMC11499956 DOI: 10.1039/d4md00541d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Here we explored new 1,5-disubstituted pyrrolidin-2-ones 1, 2 and 5-aryl-3,3a,4,5-tetrahydropyrrolo[1,2-a]quinoline-1(2H)-ones 3 as inhibitors of tubulin polymerization. We evaluated their effects on microtubule dynamics in vitro and on the proliferation of A549 cells, using flow cytometry-based cell cycle analysis. The results were verified with phase-contrast microscopy in three cancer cell lines: A549, HeLa and MCF-7. Guided by molecular modeling of the interactions between tubulin and the most active of the identified compounds, we designed, synthesized, and tested the 3-hydroxyphenyl-substituted compound 3c. This compound was further shown to bind to the colchicine site of tubulin and reduce microtubule growth rates in vitro. Moreover, compound 3c arrested division of the A549 cells in the low micromolar range (IC50 = 5.9 μM) and exhibited cytotoxicity against four different cell lines in the MTT assay for cell proliferation. Our findings demonstrate that 5-aryltetrahydropyrrolo[1,2-a]quinoline-1(2H)-one is a promising scaffold for the development of novel tubulin polymerization inhibitors.
Collapse
Affiliation(s)
- Mikhail N Anisimov
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
- Center for theoretical problems of physicochemical pharmacology Moscow 109029 Russia
| | - Maksim A Boichenko
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Vitaly V Shorokhov
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Julia N Borzunova
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | | | - Vadim V Mustyatsa
- Center for theoretical problems of physicochemical pharmacology Moscow 109029 Russia
- National Laboratory Astana Astana 010000 Kazakhstan
| | - Ilya A Lifshits
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Andrey Yu Plodukhin
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Ivan A Andreev
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow 117997 Russia
| | - Nina K Ratmanova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology Moscow 117997 Russia
| | - Sergey S Zhokhov
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Elena A Tarasenko
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Daria A Ipatova
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Alexander R Pisarev
- Faculty of Biology and Biotechnologies, Higher School of Economics Moscow 117418 Russia
| | - Ivan A Vorobjev
- National Laboratory Astana Astana 010000 Kazakhstan
- Department of Biology, School of Sciences and Humanities, Nazarbayev University Astana 010000 Kazakhstan
- Department of Biology, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Igor V Trushkov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences Moscow 119991 Russia
| | - Olga A Ivanova
- Department of Chemistry, M.V. Lomonosov Moscow State University Moscow 119991 Russia
| | - Nikita B Gudimchuk
- Department of Physics, M.V. Lomonosov Moscow State University Moscow 119991 Russia
- Center for theoretical problems of physicochemical pharmacology Moscow 109029 Russia
| |
Collapse
|
6
|
Wu C, Zhang L, Zhou Z, Tan L, Wang Z, Guo C, Wang Y. Discovery and mechanistic insights into thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines inhibitors targeting tubulin for cancer therapy. Eur J Med Chem 2024; 276:116649. [PMID: 38972078 DOI: 10.1016/j.ejmech.2024.116649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Guided by the X-ray cocrystal structure of the lead compound 4a, we developed a series of thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines demonstrating potent antiproliferative activity against four tumor cell lines. Two analogs, 13 and 25d, exhibited IC50 values around 1 nM and overcame P-glycoprotein (P-gp)-mediated multidrug resistance (MDR). At low concentrations, 13 and 25d inhibited both the colony formation of SKOV3 cells in vitro and tubulin polymerization. Furthermore, mechanistic studies showed that 13 and 25d induced G2/M phase arrest and apoptosis in SKOV3 cells, as well as dose-dependent inhibition of tumor cell migration and invasion at low concentrations. Most notably, the X-ray cocrystal structures of compounds 4a, 25a, and the optimal molecule 13 in complex with tubulin were elucidated. This study identifies thieno[3,2-d]pyrimidine and heterocyclic fused pyrimidines as representatives of colchicine-binding site inhibitors (CBSIs) with potent antiproliferative activity.
Collapse
Affiliation(s)
- Chengyong Wu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lele Zhang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhilan Zhou
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lun Tan
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhijia Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Cuiyu Guo
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
7
|
Cui YJ, Zhou Y, Zhang XW, Dou BK, Ma CC, Zhang J. The discovery of water-soluble indazole derivatives as potent microtubule polymerization inhibitors. Eur J Med Chem 2023; 262:115870. [PMID: 37890199 DOI: 10.1016/j.ejmech.2023.115870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/26/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
Taking a previously discovered indazole derivative 1 as a lead, systematic structural modifications were performed with an indazole core at the 1- and 6-positions to improve its aqueous solubility. Among the designed indazole derivatives, 6-methylpyridin-3-yl indazole derivative 8l and 1H-indol-4-yl indazole derivative 8m exhibited high potency in the low nanomolar range against A549, Huh-7, and T24 cancer cells, including Taxol-resistant variant cells (A549/Tax). As a hydrochloride salt, 8l exhibited much improved aqueous solubility, and its log P value fell into a favorable range. In mechanistic studies, 8l impeded tubulin polymerization through interacting with the colchicine site, resulting in cell cycle arrest and cellular apoptosis. In addition, compared to lead compound 1, 8l reduced cell migration and led to more potent inhibition of tumor growth in vivo without apparent toxicity. In summary, indazole derivative 8l could work as a potential anticancer agent and deserves further investigation for cancer therapy.
Collapse
Affiliation(s)
- Ying-Jie Cui
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Yi Zhou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Xi-Wu Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Bao-Kai Dou
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China
| | - Chen-Chen Ma
- Central Laboratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250012, China.
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250012, China.
| |
Collapse
|
8
|
Zhang Y, El Sayed S, Kang L, Sanger M, Wiegand T, Jessop PG, DeBeer S, Bordet A, Leitner W. Adaptive Catalysts for the Selective Hydrogenation of Bicyclic Heteroaromatics using Ruthenium Nanoparticles on a CO 2 -Responsive Support. Angew Chem Int Ed Engl 2023; 62:e202311427. [PMID: 37677109 DOI: 10.1002/anie.202311427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/09/2023]
Abstract
Ruthenium nanoparticles (NPs) immobilized on an amine-functionalized polymer-grafted silica support act as adaptive catalysts for the hydrogenation of bicyclic heteroaromatics. Whereas full hydrogenation of benzofuran and quinoline derivatives is achieved under pure H2 , introducing CO2 into the H2 gas phase leads to an effective shutdown of the arene hydrogenation while preserving the activity for the hydrogenation of the heteroaromatic part. The selectivity switch originates from the generation of ammonium formate species on the surface of the materials by catalytic hydrogenation of CO2 . The CO2 hydrogenation is fully reversible, resulting in a robust and rapid switch between the two states of the catalyst adapting its performance in response to the feed gas composition. A variety of benzofuran and quinoline derivatives were hydrogenated to fully or partially saturated products in high selectivity and yields simply by altering the composition of the feed gas from H2 to H2 /CO2 . The adaptive catalytic system thus provides controlled access to valuable products using a single catalyst rather than two specific and distinct catalysts with static reactivity.
Collapse
Affiliation(s)
- Yuyan Zhang
- Max Planck Institute for Chemical Energy Conversion, Department of Molecular Catalysis, Stiftstraße 34-36, 45470, Mülheim an der Ruhr, Germany
| | - Sami El Sayed
- Institut für Technische und Makromolekulare Chemie, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
| | - Liqun Kang
- Max Planck Institute for Chemical Energy Conversion, Department of Molecular Catalysis, Stiftstraße 34-36, 45470, Mülheim an der Ruhr, Germany
| | - Matthew Sanger
- Department of Chemistry, Queen's University, Kingston, Ontario, Canada
| | - Thomas Wiegand
- Max Planck Institute for Chemical Energy Conversion, Department of Molecular Catalysis, Stiftstraße 34-36, 45470, Mülheim an der Ruhr, Germany
- Institut für Technische und Makromolekulare Chemie, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
| | - Philip G Jessop
- Department of Chemistry, Queen's University, Kingston, Ontario, Canada
| | - Serena DeBeer
- Max Planck Institute for Chemical Energy Conversion, Department of Molecular Catalysis, Stiftstraße 34-36, 45470, Mülheim an der Ruhr, Germany
| | - Alexis Bordet
- Max Planck Institute for Chemical Energy Conversion, Department of Molecular Catalysis, Stiftstraße 34-36, 45470, Mülheim an der Ruhr, Germany
| | - Walter Leitner
- Max Planck Institute for Chemical Energy Conversion, Department of Molecular Catalysis, Stiftstraße 34-36, 45470, Mülheim an der Ruhr, Germany
- Institut für Technische und Makromolekulare Chemie, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
| |
Collapse
|
9
|
Sedenkova KN, Leschukov DN, Grishin YK, Zefirov NA, Gracheva YA, Skvortsov DA, Hrytseniuk YS, Vasilyeva LA, Spirkova EA, Shevtsov PN, Shevtsova EF, Lukmanova AR, Spiridonov VV, Markova AA, Nguyen MT, Shtil AA, Zefirova ON, Yaroslavov AA, Milaeva ER, Averina EB. Verubulin (Azixa) Analogues with Increased Saturation: Synthesis, SAR and Encapsulation in Biocompatible Nanocontainers Based on Ca 2+ or Mg 2+ Cross-Linked Alginate. Pharmaceuticals (Basel) 2023; 16:1499. [PMID: 37895970 PMCID: PMC10610134 DOI: 10.3390/ph16101499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Tubulin-targeting agents attract undiminished attention as promising compounds for the design of anti-cancer drugs. Verubulin is a potent tubulin polymerization inhibitor, binding to colchicine-binding sites. In the present work, a series of verubulin analogues containing a cyclohexane or cycloheptane ring 1,2-annulated with pyrimidine moiety and various substituents in positions 2 and 4 of pyrimidine were obtained and their cytotoxicity towards cancer and non-cancerous cell lines was estimated. The investigated compounds revealed activity against various cancer cell lines with IC50 down to 1-4 nM. According to fluorescent microscopy data, compounds that showed cytotoxicity in the MTT test disrupt the normal cytoskeleton of the cell in a pattern similar to that for combretastatin A-4. The hit compound (N-(4-methoxyphenyl)-N,2-dimethyl-5,6,7,8-tetrahydroquinazolin-4-amine) was encapsulated in biocompatible nanocontainers based on Ca2+ or Mg2+ cross-linked alginate and it was demonstrated that its cytotoxic activity was preserved after encapsulation.
Collapse
Affiliation(s)
- Kseniya N. Sedenkova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Denis N. Leschukov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Yuri K. Grishin
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Nikolay A. Zefirov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Yulia A. Gracheva
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Dmitry A. Skvortsov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Yanislav S. Hrytseniuk
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Lilja A. Vasilyeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia;
| | - Elena A. Spirkova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), 142432 Chernogolovka, Russia; (E.A.S.); (P.N.S.); (E.F.S.)
| | - Pavel N. Shevtsov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), 142432 Chernogolovka, Russia; (E.A.S.); (P.N.S.); (E.F.S.)
| | - Elena F. Shevtsova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences (IPAC RAS), 142432 Chernogolovka, Russia; (E.A.S.); (P.N.S.); (E.F.S.)
| | - Alina R. Lukmanova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Vasily V. Spiridonov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Alina A. Markova
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.M.); (M.T.N.)
| | - Minh T. Nguyen
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, 119334 Moscow, Russia; (A.A.M.); (M.T.N.)
| | - Alexander A. Shtil
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
- Institute of Cyber Intelligence Systems, National Research Nuclear University MEPhI, 115409 Moscow, Russia
| | - Olga N. Zefirova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Alexander A. Yaroslavov
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Elena R. Milaeva
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| | - Elena B. Averina
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (K.N.S.); (D.N.L.); (Y.K.G.); (N.A.Z.); (Y.A.G.); (D.A.S.); (Y.S.H.); (A.R.L.); (V.V.S.); (O.N.Z.); (A.A.Y.); (E.R.M.); (A.A.S.)
| |
Collapse
|
10
|
Liu K, Mo M, Yu G, Yu J, Song SM, Cheng S, Li HM, Meng XL, Zeng XP, Xu GC, Luo H, Xu BX. Discovery of novel 2-(trifluoromethyl)quinolin-4-amine derivatives as potent antitumor agents with microtubule polymerization inhibitory activity. Bioorg Chem 2023; 139:106727. [PMID: 37451147 DOI: 10.1016/j.bioorg.2023.106727] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023]
Abstract
In this work, a series of 2-(trifluoromethyl)quinolin-4-amine derivatives were designed and synthesized through structural optimization strategy as a microtubule-targeted agents (MTAs) and their cytotoxicity activity against PC3, K562 and HeLa cell lines were evaluated. The half maximal inhibitory concentration (IC50) of 5e, 5f, and 5o suggested that their potency of anti-proliferative activities against HeLa cell lines were better than the combretastatin A-4. Compound 5e showed the higher anti-proliferative activity against PC3, K562 and HeLa in vitro with IC50 values of 0.49 µM, 0.08 µM and 0.01 µM, respectively. Further mechanism study indicated that the representative compound 5e was new class of tubulin inhibitors by EBI competition assay and tubulin polymerization assays, it is similar to colchicine. Immunofluorescence staining revealed that compound 5e apparently disrupted tubulin network in HeLa cells, and compound 5e arrested HeLa cells at the G2/M phase and induced cells apoptosis in a dose-dependent manner. Molecular docking results illustrated that the hydrogen bonds of represented compounds reinforced the interactions in the pocket of colchicine binding site. Preliminary results suggested that 5e deserves further research as a promising tubulin inhibitor for the development of anticancer agents.
Collapse
Affiliation(s)
- Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Min Mo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Shan-Min Song
- Department of Food and Medicine, Guizhou Vocational College of Agriculture, Qingzhen 551400, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Hui-Min Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Xue-Ling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Xiao-Ping Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Guang-Can Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China.
| | - Bi-Xue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Academy of Sciences/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang 550014, China.
| |
Collapse
|
11
|
Wu H, Wang LS, Li P, Yu J, Cheng S, Yu G, Ahmad M, Meng XL, Luo H, Xu BX. Discovery of novel N-aryl-2-trifluoromethyl-quinazoline-4-amine derivatives as the inhibitors of tubulin polymerization in leukemia cells. Eur J Med Chem 2023; 256:115470. [PMID: 37201429 DOI: 10.1016/j.ejmech.2023.115470] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/05/2023] [Accepted: 05/05/2023] [Indexed: 05/20/2023]
Abstract
A series of new N-aryl-2-trifluoromethylquinazoline-4-amine analogs were designed and synthesized based on structure optimization of quinazoline by introducing a trifluoromethyl group into 2-position. The structures of the twenty-four newly synthesized compounds were confirmed by 1H NMR, 13C NMR and ESI-MS. The in vitro anti-cancer activity against chronic myeloid leukemia cells (K562), erythroleukemia cells (HEL), human prostate cancer cells (LNCaP), and cervical cancer cells (HeLa) of the target compounds was evaluated. Among them, compounds 15d, 15f, 15h, and 15i showed the significantly (P < 0.01) stronger growth inhibitory activity on K562 than those of the positive controls of paclitaxel and colchicine, while compounds 15a, 15d, 15e, and 15h displayed significantly stronger growth inhibitory activity on HEL than those of the positive controls. However, all the target compounds exhibited weaker growth inhibition activity against K562 and HeLa than those of the positive controls. The selectivity ratio of compounds 15h, 15d, and 15i were significantly higher than those of other active compounds, indicating that these three compounds had the lower hepatotoxicity. Several compounds displayed strong inhibition against leukemia cells. They inhibited tubulin polymerization, disrupted cellular microtubule networks by targeting the colchicine site, and promoted cell cycle arrest of leukemia cells at G2/M phase and cell apoptosis, as well as inhibiting angiogenesis. In summary, our research provided that novel synthesized N-aryl-2-trifluoromethyl-quinazoline-4-amine active derivatives as the inhibitors of tubulin polymerization in leukemia cells, which might be a valuable lead compounds for anti-leukemia agents.
Collapse
Affiliation(s)
- Hui Wu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Long-Shan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China; School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Pei Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Mashaal Ahmad
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Xue-Ling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China.
| | - Bi-Xue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China; The Key Laboratory of Chemistry for Natural Products of Guizhou Province and Chinese Academy of Sciences, Guiyang, 550014, China.
| |
Collapse
|
12
|
Pochampally S, Hartman KL, Wang R, Wang J, Yun MK, Parmar K, Park H, Meibohm B, White SW, Li W, Miller DD. Design, Synthesis, and Biological Evaluation of Pyrimidine Dihydroquinoxalinone Derivatives as Tubulin Colchicine Site-Binding Agents That Displayed Potent Anticancer Activity Both In Vitro and In Vivo. ACS Pharmacol Transl Sci 2023; 6:526-545. [PMID: 37082747 PMCID: PMC10111625 DOI: 10.1021/acsptsci.2c00108] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Indexed: 04/22/2023]
Abstract
Polymerization of tubulin dimers to form microtubules is one of the key events in cell proliferation. The inhibition of this event has long been recognized as a potential treatment option for various types of cancer. Compound 1e was previously developed by our team as a potent inhibitor of tubulin polymerization that binds to the colchicine site. To further improve the potency and therapeutic properties of compound 1e, we hypothesized based on the X-ray crystal structure that modification of the pyrimidine dihydroquinoxalinone scaffold with additional hetero-atom (N, O, and S) substituents could allow the resulting new compounds to bind more tightly to the colchicine site and display greater efficacy in cancer therapy. We therefore synthesized a series of new pyrimidine dihydroquinoxalinone derivatives, compounds 10, 12b-c, 12e, 12h, and 12j-l, and evaluated their cytotoxicity and relative ability to inhibit proliferation, resulting in the discovery of new tubulin-polymerization inhibitors. Among these, the most potent new inhibitor was compound 12k, which exhibited high cytotoxic activity in vitro, a longer half-life than the parental compound in liver microsomes (IC50 = 0.2 nM, t 1/2 = >300 min), and significant potency against a wide range of cancer cell lines including those from melanoma and breast, pancreatic, and prostate cancers. High-resolution X-ray crystal structures of the best compounds in this scaffold series, 12e, 12j, and 12k, confirmed their direct binding to the colchicine site of tubulin and revealed their detailed molecular interactions. Further evaluation of 12k in vivo using a highly taxane-resistant prostate cancer xenograft model, PC-3/TxR, demonstrated the strong tumor growth inhibition at the low dose of 2.5 mg/kg (i.v., twice per week). Collectively, these results strongly support further preclinical evaluations of 12k as a potential candidate for development.
Collapse
Affiliation(s)
- Satyanarayana Pochampally
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Kelli L. Hartman
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Rui Wang
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jiaxing Wang
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Mi-Kyung Yun
- Department
of Structural Biology, St. Jude Children’s
Research Hospital, Memphis, Tennessee 38105, United States
| | - Keyur Parmar
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Hyunseo Park
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Bernd Meibohm
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Stephen W. White
- Department
of Structural Biology, St. Jude Children’s
Research Hospital, Memphis, Tennessee 38105, United States
| | - Wei Li
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D. Miller
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
13
|
Tan L, Wu C, Zhang J, Yu Q, Wang X, Zhang L, Ge M, Wang Z, Ouyang L, Wang Y. Design, Synthesis, and Biological Evaluation of Heterocyclic-Fused Pyrimidine Chemotypes Guided by X-ray Crystal Structure with Potential Antitumor and Anti-multidrug Resistance Efficacy Targeting the Colchicine Binding Site. J Med Chem 2023; 66:3588-3620. [PMID: 36802449 DOI: 10.1021/acs.jmedchem.2c02115] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
Herein, a series of quinazoline and heterocyclic fused pyrimidine analogues were designed and synthesized based on the X-ray co-crystal structure of lead compound 3a, showing efficacious antitumor activities. Two analogues, 15 and 27a, exhibited favorable antiproliferative activities, which were more potent than lead compound 3a by 10-fold in MCF-7 cells. In addition, 15 and 27a exhibited potent antitumor efficacy and tubulin polymerization inhibition in vitro. 15 reduced the average tumor volume by 80.30% (2 mg/kg) in the MCF-7 xenograft model and 75.36% (4 mg/kg) in the A2780/T xenograft model, respectively. Most importantly, supported by structural optimization and Mulliken charge calculation, X-ray co-crystal structures of compounds 15, 27a, and 27b in complex with tubulin were resolved. In summary, our research provided the rational design strategy of colchicine binding site inhibitors (CBSIs) based on X-ray crystallography with antiproliferation, antiangiogenesis, and anti-multidrug resistance properties.
Collapse
Affiliation(s)
- Lun Tan
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengyong Wu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jifa Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Quanwei Yu
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiye Wang
- Department of Pharmacy, Western Theater Command Hospital, Chengdu 610083, Sichuan, China
| | - Lele Zhang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meiyi Ge
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Zhijia Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Liang Ouyang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuxi Wang
- Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Joint Research Institution of Altitude Health, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, Sichuan, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.,Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
14
|
Parshuram Satpute D, Shirwadkar U, Kumar Tharalla A, Dattatray Shinde S, Nikhil Vaidya G, Joshi S, Patel Vatsa P, Jain A, Singh AA, Garg R, Mandoli A, Kumar D. Discovery of fluorinated 2‑Styryl 4(3H)-quinazolinone as potential therapeutic hit for oral cancer. Bioorg Med Chem 2023; 81:117193. [PMID: 36796126 DOI: 10.1016/j.bmc.2023.117193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant epithelial neoplasm, affects the mouth and throat, and accounts for 90 % of oral cancers. Considering the associated morbidity with neck dissections and the limitation of existing therapeutic agents, the discovery and development of new anticancer drugs/drug candidates for oral cancer treatment are of the utmost need. In this context, reported here is the identification of fluorinated 2‑styryl 4(3H)-quinazolinone as a promising hit for oral cancer. Preliminary studies indicate that the compound blocks the transition of G1 to S phase, thereby leading to arrest in the G1/S phase. Subsequent RNA-seq analysis revealed that the compound induces the activation of molecular pathways involved in apoptosis (such as TNF signalling through NF-κB, p53 pathways) and cell differentiation and suppresses the pathways of cellular growth and development (such as KRAS signaling) in CAL-27 cancer cells. It is noted that identified hit complies with a favorable range of ADME properties as per the computational analysis.
Collapse
Affiliation(s)
- Dinesh Parshuram Satpute
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Urjita Shirwadkar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Anil Kumar Tharalla
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Sangita Dattatray Shinde
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Gargi Nikhil Vaidya
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Swarali Joshi
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Priyanka Patel Vatsa
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India
| | - Alok Jain
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India; Department of Bio-engineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, India
| | - Abhishek A Singh
- Department of Molecular Biology, Radboud University, Nijmegen, Netherlands
| | - Rachana Garg
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India; Division of Neurosurgery, Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA.
| | - Amit Mandoli
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India.
| | - Dinesh Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER) - Ahmadabad, Palaj, Gandhinagar-382355, Gujarat, India.
| |
Collapse
|
15
|
Goel KK, Kharb R, Rajput SK, Sharma PP, Mukherjee M. Design, Synthesis and Biological Evaluation of 2-Phenylquinazolin-4-yl 4-Methylbenzenesulfonate Derivatives as Anticancer Agents via Tubulin Inhibition. Synlett 2022. [DOI: 10.1055/s-0042-1751378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
AbstractMalignant behavior and multiple abnormal cellular functions have rendered cancer a great challenge for scientists to treat. The rising death toll presents an alarming situation, and the side effects associated with marketed drugs has further increased the quest to develop new anticancer drug molecules. We herein report a rationally designed 2,4-disubstituted quinazoline-based bioactive pharmacophore possessing different substitution patterns to obtain potent anticancer active agents targeting tubulin polymerization. In this series, two compounds showed potent cytotoxicity against all four cancer cell lines (MCF-7, MD-MBA-231, A549, and HCT-116) comparable to that of colchicine. The compounds showed cell cycle arrest in the G2/M phase and induced apoptosis, which showed these compounds might act via binding to the colchicine binding site. These results were further confirmed via tubulin polymerization inhibition, which showed a similar profile to colchicine. Compounds with a propargyl moiety showed very low cytotoxicity as compared to colchicines, even in the presence of a trimethoxy substituent at the quinazoline ring, except for compound case. Two compounds are obtained as potential lead compounds for the development of active anticancer agents, with one having a similar profile to colchicine activity on tubulin polymerization inhibition. These compounds represent promising leads that deserve further investigation and optimization.
Collapse
Affiliation(s)
- Kapil Kumar Goel
- Department of Pharmaceutical Sciences, Gurukul Kangri (Deemed to be University)
- Amity Institute of Pharmacy, Amity University
| | | | | | | | - Monalisa Mukherjee
- Amity Institute of Click Chemistry Research and Studies, Amity University
| |
Collapse
|
16
|
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]
|
17
|
Wang J, Miller DD, Li W. Molecular interactions at the colchicine binding site in tubulin: An X-ray crystallography perspective. Drug Discov Today 2022; 27:759-776. [PMID: 34890803 PMCID: PMC8901563 DOI: 10.1016/j.drudis.2021.12.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/27/2021] [Accepted: 12/02/2021] [Indexed: 01/02/2023]
Abstract
Tubulin is an important cancer drug target. Compounds that bind at the colchicine site in tubulin have attracted significant interest as they are generally less affected by multidrug resistance than other potential drugs. Modeling is useful in understanding the interactions between tubulin and colchicine binding site inhibitors (CBSIs), but because the colchicine binding site contains two flexible loops whose conformations are highly ligand-dependent, modeling has its limitations. X-ray crystallography provides experimental pictures of tubulin-ligand interactions at this challenging colchicine site. Since 2004, when the first X-ray structure of tubulin in complex with N-deacetyl-N-(2-mercaptoacetyl)-colchicine (DAMA-colchicine) was published, many X-ray crystal structures have been reported for tubulin complexes involving the colchicine binding site. In this review, we summarize the crystal structures of tubulin in complexes with various CBSIs, aiming to facilitate the discovery of new generations of tubulin inhibitors.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
18
|
Madbouly EA, Lashine ESM, Al-Karmalawy AA, Sebaiy MM, Pratsinis H, Kletsas D, Metwally K. Design and synthesis of novel quinazolinone–chalcone hybrids as potential apoptotic candidates targeting caspase-3 and PARP-1: in vitro, molecular docking, and SAR studies. NEW J CHEM 2022. [DOI: 10.1039/d2nj04053k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Novel quinazolinone–chalcone hybrids as potential apoptotic candidates targeting caspase-3 and PARP-1.
Collapse
Affiliation(s)
- Eman A. Madbouly
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - El-Sayed M. Lashine
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Ahmed A. Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt
| | - Mahmoud M. Sebaiy
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Harris Pratsinis
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre of Scientific Research “Demokritos”, Athens, Greece
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre of Scientific Research “Demokritos”, Athens, Greece
| | - Kamel Metwally
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| |
Collapse
|
19
|
Rezaei Z, Asadi M, Montazer MN, Rezaeiamiri E, Bahadorikhalili S, Amini M, Amanlou M. Synthesis, Molecular Docking, and Biological Evaluation of 2,3-Diphenylquinoxaline Derivatives as a Tubulin's Colchicine Binding Site Inhibitor Based on Primary Virtual Screening. Anticancer Agents Med Chem 2021; 22:2011-2025. [PMID: 34702157 DOI: 10.2174/1871520621666211026102307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 07/09/2021] [Accepted: 09/13/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE Tubulin inhibitors have proved to be a promising treatment against cancer. Tubulin inhibitors target different areas in microtubule structure to exert their effects. The colchicine binding site (CBS) is one of them for which there is no FDA-approved drug yet. This makes CBS a desirable target for drug design. MATERIALS AND METHODS Primary virtual screening is done by developing a possible pharmacophore model of colchicine binding site inhibitors of tubulins, and 2,3-diphenylquinoxaline is chosen as a lead compound to synthesis. In this study, 28 derivatives of 2,3-diphenylquinoxalines are synthesized, and their cytotoxicity is evaluated by the MTT assay in different human cancer cell lines, including AGS (Adenocarcinoma gastric cell line), HT-29 (Human colorectal adenocarcinoma cell line), NIH3T3 (Fibroblast cell line), and MCF-7 (Human breast cancer cell). RESULTS Furthermore, the activity of the studied compounds was investigated using computational methods involving molecular docking of the 2,3-diphenylquinoxaline derivatives to β-tubulin. The results showed that the compounds with electron donor functionalities in positions 2 and 3 and electron-withdrawing groups in position 6 are the most active tubulin inhibitors. CONCLUSION Apart from the high activity of the synthesized compounds, the advantage of this report is the ease of the synthesis, work-up, and isolation of the products in safe, effective, and high-quality isolated yields.
Collapse
Affiliation(s)
- Zahra Rezaei
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical, Sciences, Tehran. Iran
| | - Mehdi Asadi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical, Sciences, Tehran. Iran
| | - Mohammad Nazari Montazer
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical, Sciences, Tehran. Iran
| | - Elnaz Rezaeiamiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical, Sciences, Tehran. Iran
| | | | - Mohsen Amini
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical, Sciences, Tehran. Iran
| | - Massoud Amanlou
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tehran University of Medical, Sciences, Tehran. Iran
| |
Collapse
|
20
|
Banerjee S, Mahmud F, Deng S, Ma L, Yun MK, Fakayode SO, Arnst KE, Yang L, Chen H, Wu Z, Lukka PB, Parmar K, Meibohm B, White SW, Wang Y, Li W, Miller DD. X-ray Crystallography-Guided Design, Antitumor Efficacy, and QSAR Analysis of Metabolically Stable Cyclopenta-Pyrimidinyl Dihydroquinoxalinone as a Potent Tubulin Polymerization Inhibitor. J Med Chem 2021; 64:13072-13095. [PMID: 34406768 PMCID: PMC9206499 DOI: 10.1021/acs.jmedchem.1c01202] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Small molecules that interact with the colchicine binding site in tubulin have demonstrated therapeutic efficacy in treating cancers. We report the design, syntheses, and antitumor efficacies of new analogues of pyridopyrimidine and hydroquinoxalinone compounds with improved drug-like characteristics. Eight analogues, 5j, 5k, 5l, 5m, 5n, 5r, 5t, and 5u, showed significant improvement in metabolic stability and demonstrated strong antiproliferative potency in a panel of human cancer cell lines, including melanoma, lung cancer, and breast cancer. We report crystal structures of tubulin in complex with five representative compounds, 5j, 5k, 5l, 5m, and 5t, providing direct confirmation for their binding to the colchicine site in tubulin. A quantitative structure-activity relationship analysis of the synthesized analogues showed strong ability to predict potency. In vivo, 5m (4 mg/kg) and 5t (5 mg/kg) significantly inhibited tumor growth as well as melanoma spontaneous metastasis into the lung and liver against a highly paclitaxel-resistant A375/TxR xenograft model.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Physical Sciences, College of STEM, University of Arkansas Fort Smith, Fort Smith, Arkansas 72913, United States
| | - Foyez Mahmud
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Lingling Ma
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mi-Kyung Yun
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Sayo O Fakayode
- Department of Physical Sciences, College of STEM, University of Arkansas Fort Smith, Fort Smith, Arkansas 72913, United States
| | - Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Zhongzhi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Pradeep B Lukka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Keyur Parmar
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Stephen W White
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Yuxi Wang
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
21
|
Discovery of Novel Diarylamide N-Containing Heterocyclic Derivatives as New Tubulin Polymerization Inhibitors with Anti-Cancer Activity. Molecules 2021; 26:molecules26134047. [PMID: 34279387 PMCID: PMC8272053 DOI: 10.3390/molecules26134047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 01/11/2023] Open
Abstract
Tubulin has been regarded as an attractive and successful molecular target in cancer therapy and drug discovery. Vicinal diaryl is a simple scaffold found in many colchicine site tubulin inhibitors, which is also an important pharmacophoric point of tubulin binding and anti-cancer activity. As the continuation of our research work on colchicine binding site tubulin inhibitors, we designed and synthesized a series of diarylamide N-containing heterocyclic derivatives by the combination of vicinal diaryl core and N-containing heterocyclic skeletons into one hybrid though proper linkers. Among of these compounds, compound 15b containing a 5-methoxyindole group exhibited the most potent inhibitory activity against the tested three human cancer cell lines (MGC-803, PC-3 and EC-109) with IC50 values of 1.56 μM, 3.56 μM and 14.5 μM, respectively. Besides, the SARs of these compounds were preliminarily studied and summarized. The most active compound 15b produced the inhibition of tubulin polymerization in a dose-dependent manner and caused microtubule network disruption in MGC-803 cells. Therefore, compound 15b was identified as a novel tubulin polymerization inhibitor targeting the colchicine binding site. In addition, the results of molecular docking also suggested compound 15b could tightly bind into the colchicine binding site of β-tubulin.
Collapse
|
22
|
Man S, Wu Z, Sun R, Guan Q, Li Z, Zuo D, Zhang W, Wu Y. W436, a novel SMART derivative, exhibits anti-hepatocarcinoma activity by inducing apoptosis and G2/M cell cycle arrest in vitro and in vivo and induces protective autophagy. J Biochem Mol Toxicol 2021; 35:e22831. [PMID: 34155709 DOI: 10.1002/jbt.22831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/25/2021] [Accepted: 05/18/2021] [Indexed: 01/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is considered one of the most common primary liver cancers and the second leading cause of cancer-associated mortality around the world annually. Therefore, it is urgent to develop novel drugs for HCC therapy. We synthesized a novel 4-substituted-methoxybenzoyl-aryl-thiazole (SMART) analog, (5-(4-aminopiperidin-1-yl)-2-phenyl-2H-1,2,3-triazol-4-yl) (3,4,5-trimethoxyphenyl) methanone (W436), with higher solubility, stability, and antitumor activity than SMART against HCC cells in vivo. The purpose of this study was to investigate the mechanisms by which W436 inhibited cell growth in HCC cells. We observed that W436 inhibited the proliferation of HepG2 and Hep3B cells in a dose-dependent manner. Importantly, the anticancer activity of W436 against HCC cells was even higher than that of SMART in vivo. In addition, the antiproliferative effects of W436 on HCC cells were associated with G2/M cell cycle arrest and apoptosis via the activation of reactive oxygen species-mediated mitochondrial apoptotic pathway. W436 also induced protective autophagy by inhibiting the protein kinase B/mammalian target of rapamycin pathway. At the same time, W436 treatment inhibited the cell adhesion and invasion as well as the process of epithelial-to-mesenchymal transition Taken together, our results showed that W436 had the promising potential for the therapeutic treatment of HCC with improved solubility, stability, and bioavailability.
Collapse
Affiliation(s)
- Shuai Man
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zhuzhu Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Rui Sun
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Qi Guan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Zengqiang Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Daiying Zuo
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Yingliang Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
23
|
Peerzada MN, Hamel E, Bai R, Supuran CT, Azam A. Deciphering the key heterocyclic scaffolds in targeting microtubules, kinases and carbonic anhydrases for cancer drug development. Pharmacol Ther 2021; 225:107860. [PMID: 33895188 DOI: 10.1016/j.pharmthera.2021.107860] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/17/2022]
Abstract
Heterocyclic scaffolds are widely utilized for drug design by taking into account the molecular structure of therapeutic targets that are related to a broad spectrum of ailments, including tumors. Such compounds display various covalent and non-covalent interactions with the specific residues of the target proteins while causing their inhibition. There is a substantial number of heterocyclic compounds approved for cancer treatment, and these compounds function by interacting with different therapeutic targets involved in tumorogenesis. In this review, we trace and emphasize the privileged heterocyclic pharmacophores that have immense potency against several essential chemotherapeutic tumor targets: microtubules, kinases and carbonic anhydrases. Potent compounds currently undergoing pre-clinical and clinical studies have also been assessed for ascertaining the effective class of chemical scaffolds that have significant therapeutic potential against multiple malignancies. In addition, we also describe briefly the role of heterocyclic compounds in various chemotherapy regimens. The optimized molecular hybridization of delineated motifs may result in the discovery of more active anticancer therapeutics and circumvent the development of resistance by specific targets in the future.
Collapse
Affiliation(s)
- Mudasir Nabi Peerzada
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ernest Hamel
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Ruoli Bai
- Molecular Pharmacology Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Sesto Fiorentino, Florence, Italy.
| | - Amir Azam
- Medicinal Chemistry Research Laboratory, Department of Chemistry, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
24
|
Zhu H, Li W, Shuai W, Liu Y, Yang L, Tan Y, Zheng T, Yao H, Xu J, Zhu Z, Yang DH, Chen ZS, Xu S. Discovery of novel N-benzylbenzamide derivatives as tubulin polymerization inhibitors with potent antitumor activities. Eur J Med Chem 2021; 216:113316. [PMID: 33676300 DOI: 10.1016/j.ejmech.2021.113316] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 01/17/2023]
Abstract
A series of novel N-benzylbenzamide derivatives were designed and synthesized as tubulin polymerization inhibitors. Among fifty-one target compounds, compound 20b exhibited significant antiproliferative activities with IC50 values ranging from 12 to 27 nM against several cancer cell lines, and possessed good plasma stability and satisfactory physicochemical properties. Mechanism studies demonstrated that 20b bound to the colchicine binding site and displayed potent anti-vascular activity. Notably, the corresponding disodium phosphate 20b-P exhibited an excellent safety profile with the LD50 value of 599.7 mg/kg (i.v. injection), meanwhile, it significantly inhibited tumor growth and decreased microvessel density in liver cancer cell H22 allograft mouse model without obvious toxicity. Collectively, 20b and 20b-P are novel promising anti-tubulin agents with more druggable properties and deserve to be further investigated for cancer therapy.
Collapse
Affiliation(s)
- Huajian Zhu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wenlong Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Wen Shuai
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yang Liu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Limei Yang
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Yuchen Tan
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Tiandong Zheng
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, UK
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York, 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, New York, 11439, United States
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing, 210009, PR China.
| |
Collapse
|
25
|
Shaik TB, Malik MS, Routhu SR, Seddigi ZS, Althagafi II, Kamal A. Evaluation of Anticancer and Anti-Mitotic Properties of Quinazoline and Quinazolino-Benzothiadiazine Derivatives. Anticancer Agents Med Chem 2021; 20:599-611. [PMID: 31884931 DOI: 10.2174/1871520620666191224122204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cancer is one of the major health and social-economic problems despite considerable progress in its early diagnosis and treatment. Owing to the emergence and increase of multidrug resistance to various conventional drugs, and the continuing importance of health-care expenditure, many researchers have focused on developing novel and effective anticancer compounds. OBJECTIVE Chemical repositories provide a good platform to evaluate and exploit known chemical entities for the identification of other biological activities. In the present study, we have selected an in-house library of synthesized compounds based on two different pharmacophoric scaffolds to evaluate their cytotoxic potency on various cancer cell lines and mechanisms of action. METHODS A series of in-house synthesized quinazoline and quinazolino-benzothiadiazine derivatives were investigated for their anticancer efficacy against a panel of five cancer (DU145, MCF7, HepG2, SKOV3 and MDA-MB-231) and one normal (MRC5) cell lines. Furthermore, the active compound of the study was investigated to elucidate the mechanism of cytotoxicity by performing series of experiments such as cell cycle analysis, inhibition of tubulin polymerization, alteration of mitochondrial membrane potential, determination of endocytic pathway for drug uptake pathway and combination drug treatment. RESULTS Among all the tested compounds, fifteen of them exhibited promising growth-inhibitory effect (0.15- 5.0μM) and induced cell cycle arrest in the G2/M phase. In addition, the selected compounds inhibited the microtubule assembly; altered mitochondrial membrane potential and enhanced the levels of caspase-9 in MCF-7 cells. Furthermore, the active compound with a combination of drugs showed a synergistic effect at lower concentrations, and the drug uptake was mediated through clathrin-mediated endocytic pathway. CONCLUSION Our results indicated that quinazoline and quinazolino-benzothiadiazine conjugates could serve as potential leads in the development of new anticancer agents.
Collapse
Affiliation(s)
- Thoukhir B Shaik
- Department of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India.,Department of Biotechnology, Acharya Nagarjuna University, Nagarjuna Nagar, Guntur, A.P, India
| | - M Shaheer Malik
- Department of Chemistry and Central Research Laboratories, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Sunitha R Routhu
- Department of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
| | - Zaki S Seddigi
- Department of Environmental Health, College of Public Health and Health Informatics, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Ismail I Althagafi
- Department of Chemistry and Central Research Laboratories, Umm Al-Qura University, 21955 Makkah, Saudi Arabia
| | - Ahmed Kamal
- Department of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad-500007, India
| |
Collapse
|
26
|
Cui YJ, Liu C, Ma CC, Ji YT, Yao YL, Tang LQ, Zhang CM, Wu JD, Liu ZP. SAR Investigation and Discovery of Water-Soluble 1-Methyl-1,4-dihydroindeno[1,2- c]pyrazoles as Potent Tubulin Polymerization Inhibitors. J Med Chem 2020; 63:14840-14866. [PMID: 33201714 DOI: 10.1021/acs.jmedchem.0c01345] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Taking the previously discovered 1-methyl-1,4-dihydroindeno[1,2c]pyrazol derivative LL01 as a lead, systematic structural modifications were made at the phenolic 6- and 7-positions and the aniline at the 3-position of the indenopyrazole core to investigate the SARs and to improve water solubility. Among the designed indenopyrazoles ID01-ID33, a series of potent MTAs were identified. As the hydrochloride salt(s), ID09 and ID33 showed excellent aqueous solubility and favorable Log P value and displayed noteworthily low nanomolar potency against a variety of tumor cells, including those taxol-resistant ones. They inhibited tubulin polymerization, disrupted cellular microtubule networks by targeting the colchicine site, and promoted HepG2 cell cycle arrest and cell apoptosis. In the HepG2 xenograft mouse model, ID09 and ID33 effectively inhibited tumor growth at an oral dose of 25 mg/kg. At an intravenous (iv) injection dose of 10 mg/kg every other day, ID09 suppressed tumor growth by 68% without obvious toxicity.
Collapse
Affiliation(s)
- Ying-Jie Cui
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Chao Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Chen-Chen Ma
- Central Laboratory, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250012, P. R. China
| | - Ya-Ting Ji
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Yi-Li Yao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Long-Qian Tang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Cheng-Mei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Jing-De Wu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Zhao-Peng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| |
Collapse
|
27
|
Sai KKS, Prabhakaran J, Damuka N, Craft S, Rajagopal SA, Mintz A, Mann J, Kumar D. Synthesis and Initial In Vivo Evaluations of [
11
C]WX‐132‐18B, a Microtubule PET Imaging Agent. ChemistrySelect 2020. [DOI: 10.1002/slct.202001827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
| | - Jaya Prabhakaran
- Department of PsychiatryColumbia University Irving Medical Center 1051 Riverside Drive New York 10032 USA
- Area of Molecular Imaging and neuropathologyNew York State Psychiatric Institute 1051 Riverside Drive New York 10032 USA
| | - Naresh Damuka
- Department of RadiologyWake Forest University School of Medicine Winston-Salem North Carolina 27157 USA
| | - Suzanne Craft
- Department of Internal MedicineWake Forest University School of Medicine Winston Salem North Carolina 27157 USA
| | - Shamyaa A. Rajagopal
- Department of RadiologyWake Forest University School of Medicine Winston-Salem North Carolina 27157 USA
| | - Akiva Mintz
- Department of RadiologyColumbia University Irving Medical Center New York 10032 USA
| | - John Mann
- Department of PsychiatryColumbia University Irving Medical Center 1051 Riverside Drive New York 10032 USA
- Area of Molecular Imaging and neuropathologyNew York State Psychiatric Institute 1051 Riverside Drive New York 10032 USA
- Department of RadiologyColumbia University Irving Medical Center New York 10032 USA
| | - Dileep Kumar
- Area of Molecular Imaging and neuropathologyNew York State Psychiatric Institute 1051 Riverside Drive New York 10032 USA
| |
Collapse
|
28
|
Khelifi I, Pecnard S, Bernadat G, Bignon J, Levaique H, Dubois J, Provot O, Alami M. Synthesis and Anticancer Properties of Oxazepines Related to Azaisoerianin and IsoCoQuines. ChemMedChem 2020; 15:1571-1578. [PMID: 32485077 DOI: 10.1002/cmdc.202000197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Indexed: 01/07/2023]
Abstract
In this article, we report the synthesis and biological properties of a series of novel oxazepines related to isoCA-4 having significant antitumor properties. Among them, three oxazepin-9-ol derivatives display a nanomolar or a sub-nanomolar cytotoxicity level against five human cancer cell lines (HCT116, U87, A549, MCF7, and K562). It was demonstrated that the lead compound in this series inhibits tubulin assembly with an IC50 value of 1 μM and totally arrests the cellular cycle in the G2/M phase at the low concentration of 5 nM in HCT116 and K562 cells. Molecular modeling studies perfectly corroborates these promising results.
Collapse
Affiliation(s)
- Ilhem Khelifi
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Shannon Pecnard
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | | | - Jérome Bignon
- Institut de Chimie des Substances Naturelles, UPR 2301, CNRS, avenue de la terrasse, 91198, Gif sur Yvette, France
| | - Hélène Levaique
- Institut de Chimie des Substances Naturelles, UPR 2301, CNRS, avenue de la terrasse, 91198, Gif sur Yvette, France
| | - Joëlle Dubois
- Institut de Chimie des Substances Naturelles, UPR 2301, CNRS, avenue de la terrasse, 91198, Gif sur Yvette, France
| | - Olivier Provot
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Mouad Alami
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| |
Collapse
|
29
|
Suzuki Y, Otake A, Ueno S, Hayashi K, Ishii H, Miyoshi N, Kuroiwa K, Tachikawa M, Fujimaki Y, Nishiyama K, Manabe K, Yamazaki R, Asai A. Discovery of a Potent Anticancer Agent PVHD303 with in Vivo Activity. ACS Med Chem Lett 2020; 11:1287-1291. [PMID: 32551013 DOI: 10.1021/acsmedchemlett.0c00119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/08/2020] [Indexed: 01/20/2023] Open
Abstract
As a part of our continuous structure-activity relationship (SAR) studies on 1-(quinazolin-4-yl)-1-(4-methoxyphenyl)ethan-1-ols, the synthesis of derivatives and their cytotoxicity against the human lung cancer cell line A549 were explored. This led to the discovery of 1-(2-(furan-3-yl)quinazolin-4-yl)-1-(4-methoxyphenyl)ethan-1-ol (PVHD303) with potent antiproliferative activity. PVHD303 disturbed microtubule formation at the centrosomes and inhibited the growth of tumors dose-dependently in the HCT116 human colon cancer xenograft model in vivo.
Collapse
Affiliation(s)
- Yumiko Suzuki
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Ayana Otake
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Satoshi Ueno
- Pharmaceutical Research Department, Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Kensuke Hayashi
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Hirosuke Ishii
- Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Nao Miyoshi
- Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kenta Kuroiwa
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Masashi Tachikawa
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Yuki Fujimaki
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Kotaro Nishiyama
- Department of Materials and Life Sciences, Faculty of Science and Technology, Sophia University, 7-1 Kioi-cho, Chiyoda-ku, Tokyo 102-8554, Japan
| | - Kei Manabe
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ryuta Yamazaki
- Pharmaceutical Research Department, Yakult Central Institute, 5-11 Izumi, Kunitachi-shi, Tokyo 186-8650, Japan
| | - Akira Asai
- Graduate School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
30
|
Wang GK, Jin WF, Zhang N, Wang G, Cheng YY, Morris-Natschke SL, Goto M, Zhou ZY, Liu JS, Lee KH. Kalshiolin A, new lignan from Kalimeris shimadai. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2020; 22:489-495. [PMID: 31190564 PMCID: PMC6908768 DOI: 10.1080/10286020.2019.1592164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/04/2019] [Accepted: 03/04/2019] [Indexed: 06/09/2023]
Abstract
The Asian plant Kalimeris shimadai has been used as food and ethnologic medicine for over a thousand years. In this study, we isolated and identified one new lignan, kalshiolin A (1), and 12 known lignans (2-13). The structures were characterized by the comprehensive analyses of spectroscopic data (HR-ESI-MS, IR, 1D, and 2D-NMR) and the absolute configuration of 1 was determined from ECD calculations. The new compound 1 was also screened for cytotoxic activity but did not show significant potency (IC50 35.9-43.3 μM) against A549, MDA-MB-231, MCF7, KB, and KB-VIN cell lines.
Collapse
Affiliation(s)
- Guo-Kai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Anhui Key Laboratory of Modern Chinese Materia Medica, Hefei 230027, China
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
| | - Wen-Fang Jin
- School of Pharmacy, Anhui University of Chinese Medicine, Anhui Key Laboratory of Modern Chinese Materia Medica, Hefei 230027, China
| | - Nan Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Anhui Key Laboratory of Modern Chinese Materia Medica, Hefei 230027, China
| | - Gang Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Anhui Key Laboratory of Modern Chinese Materia Medica, Hefei 230027, China
| | - Yung-Yi Cheng
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
- Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung 404, Taiwan
| | - Susan L. Morris-Natschke
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
| | - Masuo Goto
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
| | - Zhong-Yu Zhou
- Key Laboratory of Plant Resources Conservation and Sustainable Utilization, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou 510070, China
| | - Jin-Song Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Anhui Key Laboratory of Modern Chinese Materia Medica, Hefei 230027, China
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599-7568, USA
- Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung 404, Taiwan
| |
Collapse
|
31
|
Hamze A, Alami M, Provot O. Developments of isoCombretastatin A-4 derivatives as highly cytotoxic agents. Eur J Med Chem 2020; 190:112110. [PMID: 32061961 DOI: 10.1016/j.ejmech.2020.112110] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 10/25/2022]
Abstract
Combretastatin A-4 (CA-4) is a natural anti-cancer agent isolated in 1989 from the African willow tree, Combretum caffrum. Due to its chemical simplicity, this (Z)-stilbene has been the subject of many structural modifications mainly to improve its chemical and metabolic stability. Beside a large number of synthetic analogues, isoCombretastatin A-4 (isoCA-4), has proved to be a solution of choice since this non-natural isomer of CA-4 is stable, easier to synthesize and has equivalent antitumor properties as CA-4. In this review, we will present the structure-activity relationships (SARs) around isoCA-4 since its discovery in 2007. In a first part, we will describe some alternatives to replace the phenol B-ring of isoCA-4, then we will focus on the variations made on the 1,1-ethylene double bond and then, we will evocate very recent exiting results concerning the possible replacements of the 3,4,5-trimethoxyphenyl A-ring of isoCA-4 by suitable heterocycles.
Collapse
Affiliation(s)
- Abdallah Hamze
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France
| | - Mouad Alami
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France.
| | - Olivier Provot
- Université Paris-Saclay, CNRS, BioCIS, 92290, Châtenay-Malabry, France.
| |
Collapse
|
32
|
Kurouchi H, Ohwada T. Synthesis of Medium-Ring-Sized Benzolactams by Using Strong Electrophiles and Quantitative Evaluation of Ring-Size Dependency of the Cyclization Reaction Rate. J Org Chem 2020; 85:876-901. [PMID: 31800245 DOI: 10.1021/acs.joc.9b02843] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Benzolactams with medium-sized rings were synthesized via the electrophilic aromatic substitution reaction of carbamoyl cations (R1R2N+═C═O) in good to high yields without dilution. These reactions were utilized to quantitatively examine the extent of retardation of medium-sized ring formation, compared to five- or six-membered ring formation. The order of reaction rates of formation of cyclic benzolactams is six- > five- > seven- > eight- > nine-membered ring at 25 °C. The present reaction provides a route to eight- and nine-membered benzolactams.
Collapse
Affiliation(s)
- Hiroaki Kurouchi
- Research Foundation Itsuu Laboratory , C1232, Kanagawa Science Park R&D Building, 3-2-1 Sakado , Takatsu-ku, Kawasaki , Kanagawa 213-0012 , Japan.,Graduate School of Pharmaceutical Sciences , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| | - Tomohiko Ohwada
- Graduate School of Pharmaceutical Sciences , The University of Tokyo , 7-3-1 Hongo , Bunkyo-ku, Tokyo 113-0033 , Japan
| |
Collapse
|
33
|
Jiang L, Goto M, Zhu DQ, Hsu PL, Li KP, Cui M, He X, Morris-Natschke SL, Lee KH, Xie L. Scaffold Hopping-Driven Optimization of 4-(Quinazolin-4-yl)-3,4-dihydroquinoxalin-2(1 H)-ones as Novel Tubulin Inhibitors. ACS Med Chem Lett 2020; 11:83-89. [PMID: 31938468 DOI: 10.1021/acsmedchemlett.9b00352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/16/2019] [Indexed: 11/28/2022] Open
Abstract
Scaffold hopping-driven lead optimizations were performed based on our prior lead 7-methoxy-4-(2-methylquinazolin-4-yl)-3,4-dihydroquinoxalin-2(1H)-one (2a) by C-ring expansion and isometric replacement of the A/B-ring, successively, aimed at finding new potential alternative drug candidates with different scaffold(s), high antitumor activity, and other improved properties to replace prior, once promising drug candidates that failed in further studies. Two series of new compounds 7 (a-d) and 13 (a-j) were synthesized and evaluated for antitumor activity, leading to the discovery of three highly potent compounds 13c, 13d, and 13e with different scaffolds. They exhibited similar high antitumor activity with single digital low nanomolar GI50 values (4.6-9.6 nM) in cellular assays, comparable to lead 2a, clinical drug candidate CA-4, and paclitaxel in the same assays. Further biological evaluations identified new active compounds as tubulin polymerization inhibitors targeting the colchicine binding site. Moreover, 13d showed better aqueous solubility than 2a and a similar log P value.
Collapse
Affiliation(s)
- Li Jiang
- Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing 100850, China
| | - Masuo Goto
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Dong-Qing Zhu
- Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing 100850, China
| | - Pei-Ling Hsu
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Kang-Po Li
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Mutian Cui
- Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing 100850, China
| | - Xiaoyang He
- Beijing Institute of Radiation Medicine, 27 Tai-Ping Road, Beijing 100850, China
| | - Susan Lynne Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung, Taiwan
| | - Lan Xie
- Beijing Institute of Pharmacology and Toxicology, 27 Tai-Ping Road, Beijing 100850, China
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
34
|
Design, synthesis and bio-evaluation of novel 2-aryl-4-(3,4,5-trimethoxy-benzoyl)-5-substituted-1,2,3-triazoles as the tubulin polymerization inhibitors. Eur J Med Chem 2020; 186:111846. [DOI: 10.1016/j.ejmech.2019.111846] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 10/31/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023]
|
35
|
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: 39] [Impact Index Per Article: 6.5] [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
|
36
|
Kasatkina S, Stepanova E, Dmitriev M, Mokrushin I, Maslivets A. Divergent synthesis of (quinoxalin-2-yl)-1,3-oxazines and pyrimido[1,6-a]quinoxalines via the cycloaddition reaction of acyl(quinoxalinyl)ketenes. Tetrahedron Lett 2019. [DOI: 10.1016/j.tetlet.2019.151088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Li W, Yin Y, Shuai W, Xu F, Yao H, Liu J, Cheng K, Xu J, Zhu Z, Xu S. Discovery of novel quinazolines as potential anti-tubulin agents occupying three zones of colchicine domain. Bioorg Chem 2019; 83:380-390. [DOI: 10.1016/j.bioorg.2018.10.027] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 10/10/2018] [Accepted: 10/15/2018] [Indexed: 11/17/2022]
|
38
|
Arnst KE, Banerjee S, Chen H, Deng S, Hwang DJ, Li W, Miller DD. Current advances of tubulin inhibitors as dual acting small molecules for cancer therapy. Med Res Rev 2019; 39:1398-1426. [PMID: 30746734 DOI: 10.1002/med.21568] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/16/2019] [Accepted: 01/19/2019] [Indexed: 12/25/2022]
Abstract
Microtubule (MT)-targeting agents are highly successful drugs as chemotherapeutic agents, and this is attributed to their ability to target MT dynamics and interfere with critical cellular functions, including, mitosis, cell signaling, intracellular trafficking, and angiogenesis. Because MT dynamics vary in the different stages of the cell cycle, these drugs tend to be the most effective against mitotic cells. While this class of drug has proven to be effective against many cancer types, significant hurdles still exist and include overcoming aspects such as dose limited toxicities and the development of resistance. Newer generations of developed drugs attack these problems and alternative approaches such as the development of dual tubulin and kinase inhibitors are being investigated. This approach offers the potential to show increased efficacy and lower toxicities. This review covers different categories of MT-targeting agents, recent advances in dual inhibitors, and current challenges for this drug target.
Collapse
Affiliation(s)
- Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hao Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Dong-Jin Hwang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
39
|
Li W, Xu F, Shuai W, Sun H, Yao H, Ma C, Xu S, Yao H, Zhu Z, Yang DH, Chen ZS, Xu J. Discovery of Novel Quinoline–Chalcone Derivatives as Potent Antitumor Agents with Microtubule Polymerization Inhibitory Activity. J Med Chem 2018; 62:993-1013. [DOI: 10.1021/acs.jmedchem.8b01755] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Wenlong Li
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Feijie Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Wen Shuai
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Honghao Sun
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Hong Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Cong Ma
- State Key Laboratory of Chemical Biology and Drug Discovery, and Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong, China
| | - Shengtao Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Hequan Yao
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| | - Zheying Zhu
- Division of Molecular Therapeutics & Formulation, School of Pharmacy, The University of Nottingham, University Park Campus, Nottingham NG7 2RD, U.K
| | - Dong-Hua Yang
- College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York 11439, United States
| | - Jinyi Xu
- State Key Laboratory of Natural Medicines and Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, P. R. China
| |
Collapse
|
40
|
Design, synthesis and biological evaluation of quinoline-indole derivatives as anti-tubulin agents targeting the colchicine binding site. Eur J Med Chem 2018; 163:428-442. [PMID: 30530194 DOI: 10.1016/j.ejmech.2018.11.070] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 11/20/2018] [Accepted: 11/28/2018] [Indexed: 01/09/2023]
Abstract
A series of novel isocombretastatin A-4 (isoCA-4) analogs were designed and synthesized by replacing 3,4,5-trimethoylphenyl and isovanillin of isoCA-4 with quinoline and indole moieties, respectively. The structure activity relationships (SARs) of these synthesized quinoline-indole derivatives have been intensively investigated. Two compounds 27c and 34b exhibited the most potent activities against five cancer cell lines with IC50 values ranging from 2 to 11 nM, which were comparable to those of Combretastatin A-4 (CA-4, 1). Further mechanism investigations revealed that 34b effectively inhibited the microtubule polymerization by binding to the colchicine site of tubulin. Further cellular mechanism studies elucidated that 34b disrupted cell microtubule networks, arrested the cell cycle at G2/M phase, induced apoptosis and depolarized mitochondria of K562 cells. Moreover, 34b displayed potent anti-vascular activity in both wound healing and tube formation assays. Importantly, 27c and 34b significantly inhibited tumor growth in H22 xenograft models without apparent toxicity, suggesting that 27c and 34b deserve further research as potent antitumor agents for cancer therapy.
Collapse
|
41
|
Wang YR, Yu Y, Li SM, Liu W, Li W, Morris-Natschke SL, Goto M, Lee KH, Huang XF. Salvisertin A, a New Hexacyclic Triterpenoid, and Other Bioactive Terpenes from Salvia deserta Root. Chem Biodivers 2018; 15:e1800019. [PMID: 29451710 DOI: 10.1002/cbdv.201800019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 02/12/2018] [Indexed: 11/07/2022]
Abstract
Using various chromatographic methods, a new hexacyclic triterpenoid, 2β,3β,24β-trihydroxy-12,13-cyclotaraxer-l4-en-28oic acid (1), together with ten known compounds, 2α,3α,23-trihydroxyurs-12,20(30)-dien-28oic acid (2), 6,7-dehydroroyleanone (3), horminone (4), 7-O-methylhorminone (5), sugiol (6), demethylcryptojaponol (7), 14-deoxycoleon U (8), 5,6-didehydro-7-hydroxy-taxodone (9), ferruginol (10), and dichroanone (11), were isolated from the roots of Salvia deserta. Their structures were identified on the basis of spectroscopic analysis and comparison with the reported data. The individual compounds (1, 3 - 8) were screened for cytotoxic activity, using the sulforhodamine B bioassay (SRB) method. As the results, Compounds 3, 5, and 8 showed cytotoxic potency against A549, MDA-MB-231, KB, KB-VIN, and MCF7 cell lines with IC50 values ranging from 6.5 to 10.2 μm.
Collapse
Affiliation(s)
- Yin-Ru Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Yun Yu
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China
| | - Shu-Ming Li
- Tasly R&D Institute, Tasly Pharmaceutical Co., Ltd., Tianjin, 300410, P. R. China
| | - Wei Liu
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA.,Department of Pharmacology, Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Wei Li
- Tasly R&D Institute, Tasly Pharmaceutical Co., Ltd., Tianjin, 300410, P. R. China
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Masuo Goto
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA.,Chinese Medicinal Research and Development Center, China Medical University and Hospital, Taiwan, 40402, Taiwan
| | - Xue-Feng Huang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, 210009, P. R. China.,Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
42
|
Banerjee S, Arnst KE, Wang Y, Kumar G, Deng S, Yang L, Li GB, Yang J, White SW, Li W, Miller DD. Heterocyclic-Fused Pyrimidines as Novel Tubulin Polymerization Inhibitors Targeting the Colchicine Binding Site: Structural Basis and Antitumor Efficacy. J Med Chem 2018; 61:1704-1718. [PMID: 29406710 DOI: 10.1021/acs.jmedchem.7b01858] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We report the design, synthesis, and biological evaluation of heterocyclic-fused pyrimidines as tubulin polymerization inhibitors targeting the colchicine binding site with significantly improved therapeutic index. Additionally, for the first time, we report high-resolution X-ray crystal structures for the best compounds in this scaffold, 4a, 4b, 6a, and 8b. These structures not only confirm their direct binding to the colchicine site in tubulin and reveal their detailed molecular interactions but also contrast the previously published proposed binding mode. Compounds 4a and 6a significantly inhibited tumor growth in an A375 melanoma xenograft model and were accompanied by elevated levels of apoptosis and disruption of tumor vasculature. Finally, we demonstrated that compound 4a significantly overcame clinically relevant multidrug resistance in a paclitaxel resistant PC-3/TxR prostate cancer xenograft model. Collectively, these studies provide preclinical and structural proof of concept to support the continued development of this scaffold as a new generation of tubulin inhibitors.
Collapse
Affiliation(s)
- Souvik Banerjee
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee 38163, United States
| | - Kinsie E Arnst
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee 38163, United States
| | - Yuxi Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041, China
| | - Gyanendra Kumar
- Structural Biology Department, St. Jude Children's Research Hospital , Memphis, Tennessee 38105, United States
| | - Shanshan Deng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee 38163, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital , Memphis, Tennessee 38105, United States
| | - Guo-Bo Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041, China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu 610041, China
| | - Stephen W White
- Structural Biology Department, St. Jude Children's Research Hospital , Memphis, Tennessee 38105, United States
| | - Wei Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee 38163, United States
| | - Duane D Miller
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center , Memphis, Tennessee 38163, United States
| |
Collapse
|
43
|
Design, synthesis, biological evaluation and cocrystal structures with tubulin of chiral β -lactam bridged combretastatin A-4 analogues as potent antitumor agents. Eur J Med Chem 2018; 144:817-842. [DOI: 10.1016/j.ejmech.2017.12.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 11/28/2017] [Accepted: 12/02/2017] [Indexed: 11/22/2022]
|
44
|
Synthesis and biological evaluation of N-substituted 3-oxo-1,2,3,4-tetrahydro-quinoxaline-6-carboxylic acid derivatives as tubulin polymerization inhibitors. Eur J Med Chem 2018; 143:8-20. [DOI: 10.1016/j.ejmech.2017.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 08/05/2017] [Accepted: 08/06/2017] [Indexed: 11/23/2022]
|
45
|
Tubulin inhibitors targeting the colchicine binding site: a perspective of privileged structures. Future Med Chem 2017; 9:1765-1794. [DOI: 10.4155/fmc-2017-0100] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The vital roles of microtubule in mitosis and cell division make it an attractive target for antitumor therapy. Colchicine binding site of tubulin is one of the most important pockets that have been focused on to design tubulin-destabilizing agents. Over the past few years, a large number of colchicine binding site inhibitors (CBSIs) have been developed inspired by natural products or synthetic origins, and many moieties frequently used in these CBSIs are structurally in common. In this review, we will classify the CBSIs into classical CBSIs and nonclassical CBSIs according to their spatial conformations and binding modes with tubulin, and highlight the privileged structures from these CBSIs in the development of tubulin inhibitors targeting the colchicine binding site.
Collapse
|
46
|
Discovery of 9 H -purins as potential tubulin polymerization inhibitors: Synthesis, biological evaluation and structure−activity relationships. Eur J Med Chem 2017; 138:1126-1134. [DOI: 10.1016/j.ejmech.2017.07.054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/19/2017] [Accepted: 07/23/2017] [Indexed: 02/08/2023]
|
47
|
Design and synthesis of bis(indolyl)ketohydrazide-hydrazones: Identification of potent and selective novel tubulin inhibitors. Eur J Med Chem 2017; 136:184-194. [DOI: 10.1016/j.ejmech.2017.04.078] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 04/25/2017] [Accepted: 04/30/2017] [Indexed: 12/26/2022]
|
48
|
Rekha T, Durgamma S, Padmaja A, Padmavathi V. Synthesis and antimicrobial activity of bis(azolyl)quinazoline-2,4-diamines. MONATSHEFTE FUR CHEMIE 2017. [DOI: 10.1007/s00706-017-1981-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
49
|
Cui MT, Jiang L, Goto M, Hsu PL, Li L, Zhang Q, Wei L, Yuan SJ, Hamel E, Morris-Natschke SL, Lee KH, Xie L. In Vivo and Mechanistic Studies on Antitumor Lead 7-Methoxy-4-(2-methylquinazolin-4-yl)-3,4-dihydroquinoxalin-2(1H)-one and Its Modification as a Novel Class of Tubulin-Binding Tumor-Vascular Disrupting Agents. J Med Chem 2017; 60:5586-5598. [PMID: 28653846 DOI: 10.1021/acs.jmedchem.7b00273] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
7-Methoxy-4-(2-methylquinazolin-4-yl)-3,4-dihydroquinoxalin-2(1H)-one (2), a promising anticancer lead previously identified by us, inhibited tumor growth by 62% in mice at 1.0 mg/kg without obvious signs of toxicity. Moreover, compound 2 exhibited extremely high antiproliferative activity in the NIH-NCI 60 human tumor cell line panel, with low to sub-nanomolar GI50 values (10-10 M level). It also showed a suitable balance between aqueous solubility and lipophilicity, as well as moderate metabolic stability in vivo. Mechanistic studies using Mayer's hematoxylin and eosin and immunohistochemistry protocols on xenograft tumor tissues showed that 2 inhibited tumor cell proliferation, induced apoptosis, and disrupted tumor vasculature. Moreover, evaluation of new synthetic analogues (6a-6t) of 2 indicated that appropriate 2-substitution on the quinazoline ring could enhance antitumor activity and improve druglike properties. Compound 2 and its analogues with a 4-(2-methylquinazolin-4-yl)-3,4-dihydroquinoxalin-2(1H)-one scaffold thus represent a novel class of tubulin-binding tumor-vascular disrupting agents (tumor-VDAs) that target established blood vessels in tumors.
Collapse
Affiliation(s)
- Mu-Tian Cui
- Beijing Institute of Pharmacology and Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Li Jiang
- Beijing Institute of Pharmacology and Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Masuo Goto
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Pei-Ling Hsu
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Linna Li
- Beijing Institute of Radiation Medicine , 27 Tai-Ping Road, Beijing 100850, China
| | - Qi Zhang
- Beijing Institute of Radiation Medicine , 27 Tai-Ping Road, Beijing 100850, China
| | - Lei Wei
- Beijing Institute of Pharmacology and Toxicology , 27 Tai-Ping Road, Beijing 100850, China
| | - Shou-Jun Yuan
- Beijing Institute of Radiation Medicine , 27 Tai-Ping Road, Beijing 100850, China
| | - Ernest Hamel
- Screening Technologies Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick National Laboratory for Cancer Research, National Institutes of Health , Frederick, Maryland 21702, United States
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States.,Chinese Medicine Research and Development Center, China Medical University and Hospital , Taichung 40402, Taiwan
| | - Lan Xie
- Beijing Institute of Pharmacology and Toxicology , 27 Tai-Ping Road, Beijing 100850, China.,Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
50
|
Guan F, Ding R, Zhang Q, Chen W, Li F, Long L, Li W, Li L, Yang D, Xie L, Yuan S, Wang L. WX-132-18B, a novel microtubule inhibitor, exhibits promising anti-tumor effects. Oncotarget 2017; 8:71782-71796. [PMID: 29069746 PMCID: PMC5641089 DOI: 10.18632/oncotarget.17710] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/24/2017] [Indexed: 11/25/2022] Open
Abstract
Cancer drug researchers have been seeking microtubule-inhibiting agents (MIAs) with higher bioactivity and lower toxicity than currently marketed drugs. WX-132-18B, a novel structural compound synthesized at our institute, specifically bound to the colchicine-binding site on tubulin rather than the vinblastine site, and concentration-dependently reduced microtubule content via depolymerization. It exhibited the same cellular phenotypic profiles as the classic MIAs (colchicine, vincristine, and taxol), including inducing cell cycle arrest at the G2/M phase, triggering tumor cell apoptosis, promoting nuclear membrane permeability, reducing mitochondrial membrane potential, and disrupting the redox system balance. Importantly, WX-132-18B displayed more potent in vitro bioactivity (IC50 0.45–0.99 nM) than did the classic MIAs; it inhibited the proliferation of human umbilical vein endothelial cells and seven types of human tumor cells, especially the taxol-resistant breast cancer cells MX-1/T. WX-132-18B also dose-dependently inhibited mice sarcoma, human lung, and gastric cancer xenograft tumors and the formation of tumor blood vessels in mice. In conclusion, WX-132-18B is a novel microtubule-depolymerizing agent that selectively acts on the colchicine-binding site of tubulin and exerts potent in vitro and in vivo anti-tumor effects. These characteristics, along with its anti-angiogenesis and anti-drug resistance properties, make WX-132-18B a promising anti-tumor drug candidate.
Collapse
Affiliation(s)
- Fang Guan
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Rui Ding
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Qi Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wei Chen
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Feifei Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Long Long
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Wei Li
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Linna Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Dexuan Yang
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lan Xie
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| | - Shoujun Yuan
- Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lili Wang
- Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, 100850, China
| |
Collapse
|