Generic placeholder image

Anti-Cancer Agents in Medicinal Chemistry

Editor-in-Chief

ISSN (Print): 1871-5206
ISSN (Online): 1875-5992

General Research Article

Synthesis and Investigation of the Role of Benzopyran Dihydropyrimidinone Hybrids in Cell Proliferation, Migration and Tumor Growth

Author(s): Ashutosh K. Dash, Debasis Nayak, Nazar Hussain, Mubashir J. Mintoo, Sumera Bano, Archana Katoch, Dilip M. Mondhe, Anindya Goswami* and Debaraj Mukherjee*

Volume 19, Issue 2, 2019

Page: [276 - 288] Pages: 13

DOI: 10.2174/1871520618666180903101422

Price: $65

Abstract

Background: Cancer is the second leading cause of mortality worldwide after heart diseases, and lung cancer is the topmost cause of all cancer-related deaths in both sexes. Dihydropyrimidinones (DHPMs) are medicinally important class of molecules with diverse pharmacological activities including anticancer activity. The present study focuses on the molecular hybridization of novel Benzopyran with Dihydropyrimidinone and evaluation of the resulting hybrids for cancer cell proliferation, migration and tumor growth.

Methods: We have synthesized a focused library of dihydropyrimidinone benzopyran hybrids (compounds 1-11) by joining the aromatic as well as pyran portions of the benzopyran core with dihydropyrimidinone. All the synthesized hybrid molecules were evaluated for their cytotoxic activities against a panel of four human cancer cell lines of diverse tissue origin, viz: A549 (lung carcinoma), MCF7 (mammary gland adenocarcinoma), HCT-116 (colorectal carcinoma), and PANC-1 (pancreatic duct carcinoma) with the help of MTT cell viability assay. A structure-activity relationship was made on the basis of IC50 values of different hybrids. Effect on cell proliferation was examined through colony formation assay, reactive oxygen species generation and mitochondrial membrane potential studies. Wound healing assays and cell scattering assays were employed to check the effect on cell migration. Western blotting experiments were performed to find out the molecular mechanism of action and anti-tumor studies were carried out to evaluate the in vivo efficacy of the selected lead molecule.

Results: Two types of novel hybrids were synthesized efficiently from benzopyran aldehydes, ethylacetoacetate and urea under heteropolyacid catalysis. Compound 3 was found to be the most potent hybrid among the synthesized compounds with consistent cytotoxic activities against four human cancer cell lines (IC50 values: 0.139 - 2.32 μM). Compound 3 strongly inhibited proliferation abilities of A549 cells in colony formation assay. Compound 3 exerted oxidative stress-mediated mitochondrial dysfunction, in which mitochondrial reactive oxygen species (ROS) generation as a mechanism of its anti-proliferative effects was analysed. Further, the molecule abrogated migration and cell scattering properties of aggressive PANC-1 cells. Mechanistic studies revealed that compound 3 modulated NF-kB expression and its downstream oncogenic proteins involved in cancer cell proliferation and invasion. Finally, compound 3 confirmed its in vivo anti-tumor efficacy; there observed 41.87% tumor growth inhibition at a dose of 30 mg/kg/body weight against a mouse model of Ehrlich solid tumor.

Conclusion: Our study unravels a potential anticancer lead (compound 3) from DHPMs that have opened up new research avenues for the development of promising anticancer therapeutic agents.

Keywords: Dihydropyrimidinones, benzopyran, molecular hybridization, cell proliferation, migration, tumor growth.

« Previous
Graphical Abstract
[1]
Prokopiou, E.M.; Ryder, S.A.; Walsh, J.J. Tumour vasculature targeting agents in hybrid/conjugate drugs. Angiogenesis, 2013, 16(3), 503-524.
[2]
Singh, R.K.; Prasad, D.N.; Bhardwaj, T.R. Hybrid pharmacophore-based drug design, synthesis, and antiproliferative activity of 1, 4-dihydropyridines-linked alkylating anticancer agents. Med. Chem. Res., 2014, 24(4), 1534-1545.
[3]
Gourdeau, H.; Leblond, L.; Hamelin, B.; Desputeau, C.; Dong, K.; Kianicka, I.; Custeau, D.; Boudreau, C.; Geerts, L.; Cai, S-X. Antivascular and antitumor evaluation of 2-amino-4-(3-bromo-4, 5-dimethoxy-phenyl)-3-cyano-4H-chromenes, a novel series of anticancer agents. Mol. Cancer Ther., 2004, 3(11), 1375-1384.
[4]
Mladenovi, Ä. M.; MihailoviÄ, M.; BogojeviÄ, D.; MatiÄ, S.; NićiforoviÄ, N.; MihailoviÄ, V.; VukoviÄ, N.; Sukdolak, S.; SolujiÄ, S. In vitro antioxidant activity of selected 4-hydroxy-chromene-2-one derivatives-SAR, QSAR and DFT studies. Int. J. Mol. Sci., 2011, 12(5), 2822-2841.
[5]
Nicolaou, K.C.; Pfefferkorn, J.A.; Roecker, A.J.; Cao, G.Q.; Barluenga, S.; Mitchell, H.J. Natural product-like combinatorial libraries based on privileged structures. 1. General principles and solid-phase synthesis of benzopyrans. J. Am. Chem. Soc., 2000, 122(41), 9939-9953.
[6]
Doshi, J.M.; Tian, D.; Xing, C. Structure-activity relationship studies of ethyl 2-amino-6-bromo-4-(1-cyano-2-ethoxy-2-oxoethyl)-4 H-chromene-3-carboxylate (HA 14-1), an antagonist for antiapoptotic Bcl-2 proteins to overcome drug resistance in cancer. J. Med. Chem., 2006, 49(26), 7731-7739.
[7]
Ghorbani-Vaghei, R.; Toghraei-Semiromi, Z.; Karimi-Nami, R. One-pot synthesis of 4H-chromene and dihydropyrano [3, 2-c] chromene derivatives in hydroalcoholic media. J. Braz. Chem. Soc., 2011, 22(5), 905-909.
[8]
Wilson, L.; Jordan, M.A. Microtubule dynamics: Taking aim at a moving target. Chem. Biol., 1995, 2(9), 569-573.
[9]
Madari, H.; Panda, D.; Wilson, L.; Jacobs, R.S. Dicoumaro. Cancer Res., 2003, 63(6), 1214-1220.
[10]
Kim, S-N.; Kim, N.H.; Park, Y.S.; Kim, H.; Lee, S.; Wang, Q.; Kim, Y.K. 7-Diethylamino-3 (2′-benzoxazolyl)-coumarin is a novel microtubule inhibitor with antimitotic activity in multidrug resistant cancer cells. Biochem. Pharmacol., 2009, 77(12), 1773-1779.
[11]
Atwal, K.S.; Rovnyak, G.C.; Kimball, S.D.; Floyd, D.M.
Moreland, S.; Swanson, B.N.; Gougoutas, J.Z.; Schwartz, J.; Smillie, K.M.; Malley, M.F. Dihydropyrimidine calcium channel blockers. II. 3-Substituted-4-aryl-1, 4-dihydro-6-methyl-5-pyrimidinecarboxylic acid esters as potent mimics of dihydropyridines. J. Med. Chem., 1990, 33(9), 2629-2635.
[12]
Hulubei, V.; Meikrantz, S.B.; Quincy, D.A.; Houle, T.; McKenna, J.I.; Rogers, M.E.; Steiger, S.; Natale, N.R. 4-Isoxazolyl-1, 4-dihydropyridines exhibit binding at the multidrug-resistance transporter. Bioorg. Med. Chem., 2012, 20(22), 6613-6620.
[13]
Matsuno, K.; Sawada, J.I.; Sugimoto, M.; Ogo, N.; Asai, A. Bis (hetero) aryl derivatives as unique kinesin spindle protein inhibitors. Bioorg. Med. Chem. Lett., 2009, 19(4), 1058-1061.
[14]
Myers, S.M.; Collins, I. Recent findings and future directions for interpolar mitotic kinesin inhibitors in cancer therapy. Future Med. Chem., 2016, 8(4), 463-489.
[15]
Tcherniuk, S.; Van Lis, R.; Kozielski, F.; Skoufias, D.A. Mutations in the human kinesin Eg5 that confer resistance to monastrol and S-trityl-L-cysteine in tumor derived cell lines. Biochem. Pharmacol., 2010, 79(6), 864-872.
[16]
Kozhevnikova, E.F.; Rafiee, E.; Kozhevnikov, I.V. Fries rearrangement of aryl esters catalysed by heteropoly acid: catalyst regeneration and reuse. Appl. Catal. A Gen., 2004, 260(1), 25-34.
[17]
Zilla, M.K.; Nayak, D.; Vishwakarma, R.A.; Sharma, P.R.; Goswami, A.; Ali, A. A convergent synthesis of alkyne-azide cycloaddition derivatives of 4-α, β-2-propyne podophyllotoxin depicting potent cytotoxic activity. Eur. J. Med. Chem., 2014, 77, 47-55.
[18]
Rah, B.; Lone, S.H.; Rasool, R.U.; Farooq, S.; Nayak, D.; Chikan, N.A.; Chakraborty, S.; Behl, A.; Mondhe, D.M.; Goswami, A. Design and synthesis of antitumor heck-coupled Sclareol analogues: Modulation of BH3 family members by SS-12 in autophagy and apoptotic cell death. J. Med. Chem., 2015, 58(8), 3432-3444.
[19]
Sinha, S.; Mishra, P.; Amin, H.; Rah, B.; Nayak, D.; Goswami, A.; Kumar, N.; Vishwakarma, R.; Ghosal, S. A new cytotoxic quinolone alkaloid and a pentacyclic steroidal glycoside from the stem bark of Crataeva nurvala: Study of anti-proliferative and apoptosis inducing property. Eur. J. Med. Chem., 2013, 60, 490-496.
[20]
Zilla, M.K.; Nayak, D.; Amin, H.; Nalli, Y.; Rah, B.; Chakraborty, S.; Kitchlu, S.; Goswami, A.; Ali, A. 4′-Demethyl-deoxypodophyllotoxin glucoside isolated from Podophyllum hexandrum exhibits potential anticancer activities by altering Chk-2 signaling pathway in MCF-7 breast cancer cells. Chem. Biol. Interact., 2014, 224, 100-107.
[21]
Sinha, S.; Amin, H.; Nayak, D.; Bhatnagar, M.; Kacker, P.; Chakraborty, S.; Kitchlu, S.; Vishwakarma, R.; Goswami, A.; Ghosal, S. Assessment of microtubule depolymerization property of flavonoids isolated from tanacetum gracile in breast cancer cells by biochemical and molecular docking approach. Chem. Biol. Interact., 2015, 239, 1-11.
[22]
Amin, H.; Nayak, D. ur Rasool, R.; Chakraborty, S.; Kumar, A.; Yousuf, K.; Sharma, P.R.; Ahmed, Z.; Sharma, N.; Magotra, A. Par-4 dependent modulation of cellular β-catenin by medicinal plant natural product derivative 3-azido Withaferin A. Mol. Carcinog., 2015, 55(5), 864-881.
[23]
Dash, A.K.; Jaladanki, C.K.; Maiti, D.K.; Singh, D.; Tripathi, A.K.; Gupta, V.K.; Bharatam, P.V.; Mukherjee, D. Tandem gem-dichlorination and nitrile oxide generation from chlorochromene aldoximes: synthesis of a new class of room temperature fluxional 4-chromanone derivatives. ChemistrySelect, 2016, 1(3), 567-571.
[24]
Kappe, C.O. Recent advances in the biginelli dihydropyrimidine synthesis. New tricks from an old dog. Acc. Chem. Res., 2000, 33(12), 879-888.
[25]
Rafiee, E.; Shahbazi, F. One-pot synthesis of dihydropyrimidones using silica-supported heteropoly acid as an efficient and reusable catalyst: Improved protocol conditions for the Biginelli reaction. J. Mol. Catal. Chem., 2006, 250(1), 57-61.
[26]
Fouda, A.M. Synthesis of several 4H-chromene derivatives of expected antitumor activity. Med. Chem. Res., 2016, 25(6), 1229-1238.
[27]
Focher, F.; Ubiali, D.; Pregnolato, M.; Zhi, C.; Gambino, J.; Wright, G.E.; Spadari, S. Novel nonsubstrate inhibitors of human thymidine phosphorylase, a potential target for tumor-dependent angiogenesis. J. Med. Chem., 2000, 43(13), 2601-2607.
[28]
Giovannetti, E.; Mey, V.; Nannizzi, S.; Pasqualetti, G.; Marini, L.; Del Tacca, M.; Danesi, R. Cellular and pharmacogenetics foundation of synergistic interaction of pemetrexed and gemcitabine in human non-small-cell lung cancer cells. Mol. Pharmacol., 2005, 68(1), 110-118.
[29]
Huang, C-l.; Yokomise, H.; Fukushima, M.; Kinoshita, M. Tailor-made chemotherapy for non-small cell lung cancer patients; Future Med, 2006, pp. 289-299.
[30]
Canto, R.M.F.S.; Bernardi, A.; Battastini, A.M.O.; Russowsky, D.; Eifler-Lima, V.L. Synthesis of dihydropyrimidin-2-one/thione library and cytotoxic activity against the human U138-MG and Rat C6 glioma cell lines. J. Braz. Chem. Soc., 2011, 22(7), 1379-1388.
[31]
Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell, 2011, 144(5), 646-674.
[32]
Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J., 2009, 417(1), 1-13.
[33]
Chen, Q.; Chai, Y.C.; Mazumder, S.; Jiang, C.; Macklis, R.M.; Chisolm, G.M.; Almasan, A. The late increase in intracellular free radical oxygen species during apoptosis is associated with cytochrome c release, caspase activation, and mitochondrial dysfunction. Cell Death Differ., 2003, 10(3), 323-334.
[34]
Gupta, G.P.; Massagu, Ã. ©, J. Cancer metastasis: Building a framework. Cell, 2006, 127(4), 679-695.
[35]
Valastyan, S.; Weinberg, R.A. Tumor metastasis: molecular insights and evolving paradigms. Cell, 2011, 147(2), 275-292.
[36]
Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol., 2014, 15(3), 178.
[37]
Beg, A.A.; Baltimore, D. An essential role for NF-kB in preventing TNF-alpha-induced cell death. Science, 1996, 274(5288), 782.
[38]
Catz, S.D.; Johnson, J.L. Transcriptional regulation of bcl-2 by nuclear factor [kappa] B and its significance in prostate cancer. Oncogene, 2001, 20(50), 7342.
[39]
Rosa, J.; Canovas, P.; Islam, A.; Altieri, D.C.; Doxsey, S.J. Survivin modulates microtubule dynamics and nucleation throughout the cell cycle. Mol. Biol. Cell, 2006, 17(3), 1483-1493.
[40]
Suzuki, Y.; Nakabayashi, Y.; Nakata, K.; Reed, J.C.; Takahashi, R. X-linked Inhibitor of Apoptosis Protein (XIAP) inhibits caspase-3 and-7 in distinct modes. J. Biol. Chem., 2001, 276(29), 27058-27063.
[41]
Kawakami, H.; Tomita, M.; Matsuda, T.; Ohta, T.; Tanaka, Y.; Fujii, M.; Hatano, M.; Tokuhisa, T.; Mori, N. Transcriptional activation of survivin through the NF-kB pathway by human T-cell leukemia virus type I tax. Int. J. Cancer, 2005, 115(6), 967-974.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy