Cytotoxicity and pro‐apoptosis activity of synthetic 1,3-thiazole incorporated phthalimide derivatives on cancer cells

Document Type : Original Article

Authors

1 Pharmaceutical Sciences Research Center, Health Institute, School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran

2 Department of Pharmacognosy and Pharmaceutical Biotechnology, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran

3 Students Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran

4 Department of Medicinal Chemistry, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran

Abstract

Objective(s): Cancer is the second important reason for death worldwide. In spite of advances in cancer treatment, however, survival of patients stays weak. Therefore, there is a critical need for advancement of new anticancer drugs. Regarding the hopeful biological activity of phthalimide derivatives, in this study, synthesis, cytotoxicity, and pro‐apoptosis activity of eleven derivatives of thiazole bearing phthalimide structure were evaluated.
Materials and Methods: First, target derivatives were synthesized. All synthesized compounds were characterized by spectroscopic methods. Cytotoxicity and pro‐apoptosis activity of the synthesized compounds were evaluated in MDA-MB-468, PC-12, and MCF-7 cancer cell lines by MTT assay, caspase-3 activity, and TUNEL assay. Finally, expression of BAX, BCL-2, and FAS (as markers of apoptosis) was assessed by the RT-PCR procedure.
Results: Among the eleven compounds, 5b (IC50 = 0.2±0.01 µM) was found to be the most potent derivative against MCF-7 cells. Also, Compound 5k and 5g showed strong cytotoxic activity against MDA-MB-468 and PC-12 cells with IC50 value of 0.6±0.04 µM and 0.43±0.06 µM, respectively.  DNA fragmentation and activity of caspase-3 data suggest that cytotoxic activity of the compounds on cancer cells might be related to apoptosis. Also, RT-PCR of apoptosis markers indicated that these compounds induce apoptosis through the intrinsic pathway.
Conclusion: Our findings suggest that para chloro derivative (5c) may be a promising agent for treatment of cancer cells by the targeted intrinsic pathway of apoptosis and could be used as a drug candidate for in vivo assessment in the treatment of cancer.

Keywords


1. Jemal A. Murray t. Ward E, Samuels A, Tiwari RC, Ghafoor A et al. Cancer Statistics 2005; Cancer J Clin 2005; 55: 10-30.
2. Srivastava V, Negi AS, Kumar JK, Gupta MM, Khanuja SP. Plant-based anticancer molecules: A chemical and biological profile of some important leads. Bioorg Med Chem 2005; 13:5892-908.
3. Leite AC, Santos LM, Moreira DR, Brondani DJ. Synthesis and characterization of new amino acyl-4-thiazolidones. Química Nova 2007; 30:284-286.
4. Ferreira PM, Lima DJ, Debiasi BW, Soares BM, da Conceição Machado K, et al. Antiproliferative activity of Rhinella marina and Rhaebo guttatus venom extracts from Southern Amazon. Toxicon 2013; 72:43-51.
5. Ferreira JR, Cavalcanti BC, da Costa PM, de Arantes FF, de Alvarenga ES, Maltha CR, de Almeida Barbosa LC, Militão GC, Pessoa C, Ferreira PM. Induction of G2/M arrest, caspase activation and apoptosis by α-santonin derivatives in HL-60 cells. Toxicol in vitro 2013; 27:1458-1466.
6. Ruchelman AL, Man HW, Zhang W, Chen R, Capone L, Kang J, Parton A, Corral L, Schafer PH, Babusis D, Moghaddam MF. Isosteric analogs of lenalidomide and pomalidomide: Synthesis and biological activity. Bioorg Med Chem Lett 2013; 23:360-365.
7. Aliabadi A, Gholamine B, Karimi T, Synthesis and antiseizure evaluation of isoindoline-1,3-dione derivatives in mice. Med Chem Res 2014; 23:2736-2743.
8. Mohammadi-Farani A, Abdi N, Moradi A, Aliabadi A, 2-(2-(4-Benzoylpiperazin-1-yl)ethyl)isoindoline-1,3-dione derivatives: Synthesis, docking and acetylcholinesterase inhibitory evaluation as anti-Alzheimer agents. Iran J Basic Med Sci 2017; 20:59-66.
9. Aliabadi A, Foroumadi A, Safavi M, Kaboudian Ardestani S, Synthesis, molecular docking and cytotoxicity evaluation of 2-(4-substituted-benzyl)isoindoline-1,3-dione derivatives as anticancer agents. J Rep Pharm Sci 2012; 1:19-22.
10. Aliabadi A, Mohammadi-Farani A, Seydi-Kangarshahi S, Ahmadi F, Discovery of 2-(1,3-Dioxoisoindolin-2-yl)-N-phenylacetamide derivatives as probable 15-lipoxygenase-1 inhibitors with potential anticancer effects. Farmacia 2017; 65: 268-274.
11. Machado AL, Lima LM, Araújo-Jr JX, Fraga CA, Koatz VL, Barreiro EJ. Design, synthesis and antiinflammatory activity of novel phthalimide derivatives, structurally related to thalidomide. Bioorg Med Chem Lett 2005; 15:1169-1172.
12. Mazzoccoli L, Cadoso SH, Amarante GW, de Souza MV, Domingues R, Machado MA, et al. Novel thalidomide analogues from diamines inhibit pro-inflammatory cytokine production and CD80 expression while enhancing IL-10. Biomed Pharmacother 2012; 66:323-329.
13. Yang YJ, Yang YN, Jiang JS, Feng ZM, Liu HY, Pan XD, et al. Synthesis and cytotoxic activity of heterocycle-substituted phthalimide derivatives. Chinese Chem Lett 2010; 21:902-904.
14. Akgün H, Karamelekoğlu İ, Berk B, Kurnaz I, Sarıbıyık G, Öktem S, Kocagöz T. Synthesis and antimycobacterial activity of some phthalimide derivatives. Bioorg Med Chem 2012; 20:4149-4154.
15. Andrade P, Visser-Vandewalle V, Del Rosario JS, Daemen MA, Buurman WA, Steinbusch HW, et al. The thalidomide analgesic effect is associated with differential TNF-α receptor expression in the dorsal horn of the spinal cord as studied in a rat model of neuropathic pain. Brain Res 2012; 1450:24-32.
16. Palencia G, Rubio C, Custodio-Ramirez V, Paz C, Sotelo J. Strong anticonvulsant effect of thalidomide on amygdaloid kindling. Epilepsy Res 2011; 95:263-269.
17. Pessoa C, Ferreira PM, Lotufo LV, de Moraes MO, Cavalcanti SM, Coêlho LC, et al. Discovery of phthalimides as immunomodulatory and antitumor drug prototypes. Chem Med Chem 2010; 5:523-8.
18. D’Amato RJ, Loughnan MS, Flynn E, Folkman J. Thalidomide is an inhibitor of angiogenesis.  Proc Natl Acad Sci 1994; 91:4082-4085.
19. Dredge K, Horsfall R, Robinson SP, Zhang LH, Lu L, Tang Y, et al. Orally administered lenalidomide (CC-5013) is anti-angiogenic in vivo and inhibits endothelial cell migration and Akt phosphorylation in vitro. Microvasc Res 2005; 69:56-63.
20. Badamtseren B, Odkhuu E, Koide N, Haque A, Naiki Y, Hashimoto S, et al. Thalidomide inhibits interferon-γ-mediated nitric oxide production in mouse vascular endothelial cells. Cell Immunol 2011; 270:19-24.
21. Kagoya Y, Nannya Y, Kurokawa M. Thalidomide maintenance therapy for patients with multiple myeloma: meta-analysis. Leuk Res 2012; 36:1016-1021.
22. Hosseinzadeh L, Aliabadi A, Kalantari M, Mostafavi A, Rahmani Khajouei M, Synthesis and cytotoxicity evaluation of some new 6-nitro derivatives of thiazole-containing 4-(3H)-quinazolinone. Res Pharm Sci 2016; 11: 210-218.
24. Hosseinzadeh L, Aliabadi A, Rahnama M, Mir Mohammad Sadeghi H, Rahmani Khajouei M, Synthesis and cytotoxic evaluation of some new 3-(2-(2-phenylthiazol-4-yl)ethyl)-quinazolin-4(3H) one derivatives with potential anticancer effects. Res Pharm Sci 2017; 12:290-298.
26. Aliabadi A, Foroumadi A, Safavi M, K. Ardestani S, Synthesis, cytotoxicity assessment and molecular docking of 4-Substituted-2-p-tolylthiazole derivatives as probable c-Src and erb tyrosine kinase inhibitors. Croat Chem Acta 2013; 86:245-251.
27. Mohammadi-Farani A, Foroumadi A, Rezvani Kashani M, Aliabadi A, N-Phenyl-2-p-tolylthiazole-4-carboxamide derivatives: Synthesis and cytotoxicity evaluation as anticancer agents. Iran J Basic Med Sci 2014; 17:502-508.
28. Yang Y, Zhu YQ, Jiang L, Li LF, Ge JP. Thalidomide induces apoptosis in human oral squamous cell carcinoma cell line with altered expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Oral Oncol 2011; 47:927-928.
29. Nabhan C, Petrylak DP. The role of IMiDs alone or in combination in prostate cancer. Clin Genitourin cancer 2012; 10:141-146.
30. Macpherson GR, Franks M, Tomoaia-Cotisel A, Ando Y, Price DK, Figg WD. Current status of thalidomide and its role in the treatment of metastatic prostate cancer. Crit Rev Oncol Hematol 2003; 46:49-57.
31.  Silva Júnior JG, Holanda VN, Rodrigues Gamb DS, Siqueira do Monte TV, Andrade de Araújo HD, Alves do Nascimento Júnior JA, Felipe da Silva Araújo V, et al. Therapeutic potential of phthalimide derivatives: A Review. Am J Biomed Sci Res 2019; 3:378-384.
32. Huang P, Robertson LE, Wright S, Plunkett W. High molecular weight DNA fragmentation: A critical event in nucleoside analogue-induced apoptosis in leukemia cells. Clin Cancer Res. 1995; 1:1005-1013.
33. Safavi M, Esmati N, Ardestani SK, Emami S, Ajdari S, Davoodi J, Shafiee A, Foroumadi A. Halogenated flavanones as potential apoptosis-inducing agents: synthesis and biological activity evaluation. Eur J Med Chem 2012; 58:573-580.
34. Lu Y, Mahato RI, editors. Pharmaceutical perspectives of cancer therapeutics. Springer; 2009. p. 623-694.
35. Xie K, Huang S. Contribution of nitric oxide-mediated apoptosis to cancer metastasis inefficiency. Free Rad Biol Med 2003; 34:969-986.
36. Mahdavi M, Davoodi J, Zali MR, Foroumadi A. Concomitant activation of caspase-9 and down-regulation of IAP proteins as a mechanism of apoptotic death in HepG2, T47D and HCT-116 cells upon exposure to a derivative from 4-aryl-4H-chromenes family. Biomed Pharmacother 2011; 65:175-182.
37. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol 2007; 35:495-516.
38. Cáceres-Castillo D, M. Carballo R, A. Tzec-Interián J, J. Mena-Rejón G, Solvent-free synthesis of 2-amino-4-arylthiazoles under microwave irradiation. Tetrahedron Lett 2012; 53:3934-3936.
39. Mohammadi G, Shakeri A, Fattahi A, Mohammadi P, Mikaeili A, Aliabadi A,  et al. Preparation, physicochemical characterization and anti-fungal    evaluation of nystatin-loaded PLGA-glucosamine nanoparticles. Pharm Res 2017; 34:301-309.
41. Rahmani-Khajouei M, Mohammadi-Farani A, Moradi A, Aliabadi A, Synthesis and evaluation of anticonvulsant activity of (Z)-4-(2-oxoindolin-3-ylideneamino)-N-phenylbenzamide derivatives in mice. Res Pharm Sci 2018; 13:262-272.
42. Kang MH, Reynolds CP. Bcl-2 inhibitors: targeting mitochondrial apoptotic pathways in cancer therapy. Clin Cancer Res 2009; 15:1126-1132.
43. Nakamura S, Kobayashi M, Shibata K, Sahara N, Shigeno K, Shinjo K, et al. Leuk Res 2006; 30:123-135.
44. Brady HJ, Gil-Gomez G. The pro-apoptotic Bcl-2 family member, Bax. Int J Biochem Cell Biol 1998; 30:647-650.
45. Nagata S, Golstein P. The Fas death factor. Science 1995; 267:1449-1456.
46. Nagata S. Apoptosis by death factor. Cell 1997; 88:355-365.
47. Tanaka M, Suda T, Takahashi T, Nagata S. Expression of the functional soluble form of human fas ligand in activated lymphocytes. EMBO J. 1995; 14:1129-1135.
48. Suda T, Takahashi T, Golstein P, Nagata S. Molecular cloning and expression of the Fas ligand, a novel member of the tumor necrosis factor family. Cell 1993; 75:1169-1178.
49. Xerri L, Devilard E, Hassoun J, Mawas C, Birg F. Fas ligand is not only expressed in immuneprivileged human organs but is also coexpressed with Fas in various epithelial tissues. Mol Pathol 1997; 50:87-91.
50. Poppema S, Potters M, Visser L, Van den Berg AM. Immune escape mechanisms in Hodgkin’s disease. Ann Oncol 1998; 9 Suppl 5: S21-24.
51. Bennett MW, O’Connell J, O’Sullivan GC, Roche D, Brady C, Kelly J, et al. Expression of Fas ligand by human gastric adenocarcinomas: a potential mechanism of immune escape in stomach cancer. Gut 1999; 44:156-162.
52. Lee SH, Shin MS, Park WS, Kim SY, Dong SM Lee HK, et al. Immunohistochemical analysis of Fas ligand expression in normal human tissues. APMIS 1999; 107:1013-1009.
53. Loreto C, La Rocca G, Anzalone R, Caltabiano R, Vespasiani G, Castorina S, et al. The role of intrinsic pathway in apoptosis activation and progression in Peyronie’s disease. Bio Med Res Int 2014:1-10.