Design, synthesis, and biological evaluation of novel 5,6,7-trimethoxy quinolines as potential anticancer agents and tubulin polymerization inhibitors

Document Type : Original Article

Authors

1 Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran

2 Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

3 Department of Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran

Abstract

Objective(s): Microtubules have key roles in essential cellular processes such as mitosis, cell motion, and intracellular organelle transport. Increasing interest has been given to tubulin binding compounds after the introduction of taxanes into clinical oncology. The object of this study was  synthesis and biological evaluation of novel 5,6,7-trimethoxy quinolines as tubulin inhibitors.
Materials and Methods: The cytotoxicity of the newly synthesized compounds was assessed against different human cancer cell lines including MCF-7, A2780, MCF-7/MX, A2780/RCIS, and normal cells. Compounds demonstrating the most antiproliferative activity, were chosen to examine their tubulin inhibition activity and their ability to arrest the cell cycle and induce apoptosis. Molecular docking studies and molecular dynamics simulation of compound 7e in the catalytic site of tubulin were performed.
Results: Most of the synthesized quinolines showed moderate to significant cytotoxic activity against human cancer cells. Compounds 7e and 7f, possessing N-(4-benzoyl phenyl) and N-(4-phenoxy phenyl), respectively, exhibited the most antiproliferative activity more potent than the other compounds and exhibited similar antiproliferative activity on both resistant and parental cancer cells.
Conclusion: Flow cytometry analysis of A2780, A2780/RCIS, MCF-7, and MCF-7/MX cancer cells treated with 7e and 7f exhibited that these compounds arrested the cell cycle (at the G2/M phase) and induced cellular apoptosis in A2780 cancer cells. These quinolines inhibited tubulin polymerization in a way resembling that of CA-4. Molecular dynamics simulation and molecular docking studies of compound 7e into the binding site of tubulin displayed the probable interactions of 7e with the binding site of tubulin.

Keywords


1. Kirschner M, Mitchison, T. Beyond self-assembly: from microtubules to morphogenesis. Cell. 1986; 45: 329-342.
2. Mignot I, Pecqueur L, Dorléans A, Karuppasamy M, Ravelli RB, Dreier B, et al. Design and characterization of modular scaffolds for tubulin assembly. J Biol Chem 2012; 287: 31085-31094.
3. Akhmanova A, Steinmetz MO. Control of microtubule organization and dynamics: two ends in the limelight. Nat Rev Mol Cell Biol 2015; 16:711-726.
4. Kadavath H, Hofele RV, Biernat J, Kumar S, Tepper K, Urlaub H, et al. Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proc Natl Acad Sci 2015; 112: 7501-7506.
5. Chandrasekaran G, Tátrai P, Gergely F. Hitting the brakes: targeting microtubule motors in cancer. Br J Cancer 2015; 113: 693-698.
6. Negi AS, Gautam Y, Alam S, Chanda D, Luqman S, Sarkar J, et al. Natural antitubulin agents: Importance of 3, 4, 5-trimethoxyphenyl fragment. Bioorg Med Chem 2015; 23: 373-389.
7. Wang W, Kong D, Cheng H, Tan L, Zhang Z, Zhuang X, et al. New benzimidazole-2-urea derivates as tubulin inhibitors. Bioorg Med Chem lett 2014, 24: 4250-4253.
8. Gigant B, Wang C, Ravelli RB, Roussi F, Steinmetz  MO, Curmi  PA, et al. Structural basis for the regulation of tubulin by vinblastine. Nature 2005; 435: 519-522.
9. Ravelli RB, Gigant B, Curmi PA, Jourdain I, Lachkar S, Sobel A, et al. Insight into tubulin regulation from a complex with colchicine and a stathmin-like domain. Nature 2004; 428: 198-202.
10. Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer 2004; 4: 253-265.
11. Canta A, Chiorazzi A, Cavaletti G. Tubulin: a target for antineoplastic drugs into the cancer cells but also in the peripheral nervous system. Curr Med Chem 2009; 16:1315-1324.
12. Kavallaris M.  Microtubules and resistance to tubulin-binding agents. Nat Rev Cancer 2010; 10:194-204
13. Seligmann J, Twelves C. Tubulin: an example of targeted chemotherapy. Future med chem 2013; 5: 339-352.
14. Bukhari SNA. Kumar GB, Revankar HM, Qin HL. Development of combretastatins as potent tubulin polymerization inhibitors. Bioorg Chem 2017; 72: 130-147.
15. Chen P, Zhuang YX, Diao PC, Yang F, Wu SY, Lv L, et al. Synthesis, biological evaluation, and molecular docking investigation of 3-amidoindoles as potent tubulin polymerization inhibitors. Eur J Med Chem 2019; 162: 525-533.
16. Jurášek M, Černohorská M, Řehulka J, Spiwok V, Sulimenko T, Dráberová E, et al. Estradiol dimer inhibits tubulin polymerization and microtubule dynamics. J Steroid Biochem Mol Bio 2018; 183: 68-79.
17. Lai Q, Wang Y, Wang R, Lai W, Tang L, Tao Y, et al. Design, synthesis and biological evaluation of a novel tubulin inhibitor 7a3 targeting the colchicine binding site. Eur J Med Chem 2018; 156: 162-179.
18. Li W, Sun H, Xu F, Shuai W, Liu J, Xu S, et al. Synthesis, molecular properties prediction and biological evaluation of indole-vinyl sulfone derivatives as novel tubulin polymerization inhibitors targeting the colchicine binding site. Bioorg Chem 2019; 85: 49-59.
19. Wang YT, Shi TQ, Zhu HL, Liu CH. Synthesis, biological evaluation and molecular docking of benzimidazole grafted benzsulfamide-containing pyrazole ring derivatives as novel tubulin polymerization inhibitors. Bioorg Med Chem 2018; 1: 502-515.
20. Yang H, An B, Li X, Zeng W. Evaluation of 4-phenylamino-substituted naphthalene-1,2-diones as tubulin polymerization inhibitors. Bioorg Med Chem lett 2018; 28: 3057-3063.
21. Zhai M, Liu S, Gao M, Wang L, Sun J, Du J, et al. 3,5-Diaryl-1H-pyrazolo[3,4-b]pyridines as potent tubulin polymerization inhibitors: Rational design, synthesis and biological evaluation. Eur J Med Chem 2018; 168: 426-435.
22. La Regina G, Bai R, Coluccia A, Naccarato V, Famiglini V, Nalli M, et al. New 6- and 7-heterocyclyl-1H-indole derivatives as potent tubulin assembly and cancer cell growth inhibitors. Eur J Med Chem 2018; 152:283-297.
23. Li W, Yin Y, Shuai W, Xu F, Yao H, Liu J, et al. Discovery of novel quinazolines as potential anti-tubulin agents occupying three zones of colchicine domain. Bioorg Chem 2019; 83: 380-390.
24. Behbahani FS, Tabeshpour J, Mirzaei S, Golmakaniyoon S, Tayarani-Najaran Z, Ghasemi A, et al. Synthesis and biological evaluation of novel benzo[c]acridine-diones as potential anticancer agents and tubulin polymerization inhibitors. Arch Pharm 2019; 352: 1800307.
25. Mirzaei S, Hadizadeh F, Eisvand F, Mosaffa F, Ghodsi R. Synthesis, structure-activity relationship and molecular docking studies of novel quinoline-chalcone hybrids as potential anticancer agents and tubulin inhibitors. J Mol Struct 2020; 1202:127310-127322.
26. Karimikia E, Behravan J, Zarghi A, Ghandadi M, Omid Malayeri S, Ghodsi R. Colchicine-like β-acetamidoketones as inhibitors of microtubule polymerization: Design, synthesis and biological evaluation of in vitro anticancer activity. Iran J Basic Med Sci 2019; 22:1138-1146.
27. Afzal O, Kumar S, Haider MR, Ali MR, Kumar R, Jaggi M, et al: A review on anticancer potential of bioactive heterocycle quinoline. Eur J Med Chem 2015; 97: 871-910.
28. Efferth T, Fu YJ, Zu Yg, Schwarz G, Konkimalla VSB, Wink M. Molecular target-guided tumor therapy with natural products derived from traditional Chinese medicine. Curr Med Chem 2007; 14: 2024-2032.
29. Arafa RK, Hegazy GH, Piazza GA, Abadi AH. Synthesis and in vitro antiproliferative effect of novel quinoline-based potential anticancer agents. Eur J Med Chem 2013; 63: 826-832.
30. Chang FS, Chen W, Wang C, Tzeng CC, Chen YL. Synthesis and antiproliferative evaluations of certain 2-phenylvinylquinoline (2-styrylquinoline) and 2-furanylvinylquinoline derivatives. Bioorg Med Chem 2010; 18: 124-133.
31. Lockman JW, Klimova Y, Anderson MB, Willardsen JA. Synthesis of substituted quinazolines: application to the synthesis of verubulin. Synth Commun 2012; 42: 1715-1723.
32. Nagarsenkar A, Prajapti SK, Guggilapu SD, Birineni S, Kotapalli SS, Ummanni R, et al. Investigation of triazole-linked indole and oxindole glycoconjugates as potential anticancer agents: novel Akt/PKB signaling pathway inhibitors. Med ChemComm 2016; 7: 646-653.
33. Mirzaei S, Hadizadeh F, Eisvand F, Mosaffa F, Ghasemi A, Ghodsi R. Design, synthesis and biological evaluation of novel 5, 6, 7-trimethoxy-N-aryl-2-styrylquinolin-4-amines as potential anticancer agents and tubulin polymerization inhibitors. Bioorg Chem 2020; 98: 103711-103727.
34. Ghodsi R, Azizi E, Ferlin MG, Pezzi V, Zarghi, A. Design, synthesis and biological evaluation of 4-(imidazolylmethyl)-2-aryl-quinoline derivatives as aromatase inhibitors and anti-breast cancer agents. Lett Dru. De. Disc 2016; 13: 89-97.
35. Malayeri SO, Abnous K, Arab A, Akaberi M, Mehri S, Zarghi A, et al. Design, synthesis and biological evaluation of 7-(aryl)-2,3-dihydro-[1,4]dioxino[2,3-g]quinoline derivatives as potential Hsp90 inhibitors and anticancer agents. Bioorg Med Chem 2017; 25: 1294-1302.
36. Jafari F, Baghayi H, Lavaee P, Hadizadeh F, Soltani F, Moallemzadeh H, et al. Design, synthesis and biological evaluation of novel benzo- and tetrahydrobenzo-[h]quinoline derivatives as potential DNA-intercalating antitumor agents. Eur J Med Chem 2019; 164: 292-303.
37. Pang Y, Yan J, An B, Huang L, Li X. The synthesis and evaluation of new butadiene derivatives as tubulin polymerization inhibitors. Bioorg Med Chem 2017; 25: 3059-3067.
38. Malayeri SO, Tayarani-Najaran Z, Behbahani FS, Rashidi R, Delpazir S, Ghodsi R. Synthesis and biological evaluation of benzo[b]furo[3,4-e][1,4]diazepin-1-one derivatives as anti-cancer agents. Bioorg Chem 2018; 80: 631-638.
39. Wang XQ, Xia CL, Chen SB, Tan JH, Ou TM, Huang SL, et al. Design, synthesis, and biological evaluation of 2-arylethenylquinoline derivatives as multifunctional agents for the treatment of Alzheimer’s disease. Eur J Med Chem 2015; 89: 349-361.
40. Yan Y, Xu K, Fang Y, Wang Z. A catalyst-free benzylic C–H bond olefination of azaarenes for direct Mannich-like reactions. J Org Chem 2011; 76: 6849-6855.
41. Abdizadeh T, Kalani MR, Abnous K, Tayarani-Najaran Z, Khashyarmanesh BZ, Abdizadeh R, et al. Design, synthesis and biological evaluation of novel coumarin-based benzamides as potent histone deacetylase inhibitors and anticancer agents. Eur J Med Chem 2017; 132: 42-62.
42. Mirzaei S, Hadizadeh F, Eisvand F, Mosaffa F, Ghodsi R. Synthesis, structure-activity relationship and molecular docking studies of novel quinoline-chalcone hybrids as potential anticancer agents and tubulin inhibitors. J Mol Struct 2020; 1202: 127310-127322.