Design, synthesis, and cytotoxicity evaluation of novel indole-acylhydrazone derivatives of 4-pyridinone as potential histone deacetylase-2 inhibitors

Niloofar Naghi-Ganji , Lotfollah Saghaei, Farial Tavakoli, Vajihe Azimian, Mina Mirian, Hajar Sirous, Mahboubeh Rostami

Abstract


Background and purpose: Histone deacetylation is one of the essential cellular pathways in the growth and spread of cancer, so the design of histone deacetylase (HDAC) inhibitors as anticancer agents is of great importance in pharmaceutical chemistry. Here, a series of indole acylhydrazone derivatives of 4-pyridone have been introduced as potential histone deacetylase inhibitors.

Experimental approach: Seven indole-acylhydrazone-pyridinone derivatives were synthesized via simple, straightforward chemical procedures. The molecular docking studies were accomplished on HDAC2 compared to panobinostat. The cytotoxicity of all derivatives was studied on MCF-7 and MDA-MB-231 breast cancer cell lines by MTT assay.

Findings / Results: Molecular docking studies supported excellent fitting to the HADC2 active site with binding energies in the range of -10 Kcal/mol for all derivatives. All compounds were tested for their cytotoxicity against MCF-7 and MDA-MB-231 cell lines; derivatives A, B, F, and G were the best candidates. The half-maximal inhibitory concentration (IC50) values on MCF-7 were below 25 mg/mL and much lower than those obtained on the MDA-MB-231 cell line.

Conclusion and implications: The derivatives showed selectivity toward the MCF-7 cell line, probably due to the higher HDAC expression in the MCF-7 cell line. In this regard, debenzylated derivatives F and G showed slightly better cytotoxicity, which should be more studied in the future. Derivatives A, B, F, and G were promising for future enzymatic studies.


Keywords


Acylhydrazone; Cytotoxicity; HDAC inhibitor; Indole; Molecular docking 4-Pyridone.

Full Text:

PDF

References


Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7-33. DOI: 10.3322/caac.21654.

Wan Y, Li Y, Yan C, Yan M, Tang Z. Indole: a privileged scaffold for the design of anti-cancer agents. Eur J Med Chem. 2019;183:11169. DOI: 10.1016/j.ejmech.2019.111691.

Drummond DC, Noble CO, Kirpotin DB, Guo Z, Scott GK, Benz CC. Clinical development of histone deacetylase inhibitors as anticancer agents. Annu Rev Pharmacol Toxicol. 2005;45:495-528. DOI: 10.1146/annurev.phamtox.45.120403.0955825.

Sanaei M, Kavoosi F. Histone deacetylases and histone deacetylase inhibitors: molecular mechanisms of action in various cancers. Adv Biomed Res. 2019;8:63-75. DOI: 10.4103/abr.abr_142_19.

Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, Walther TC, et al. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science. 2009;325(5942):834-840. DOI: 10.1126/science.1175371.

Zhang L, Zhang J, Jiang Q, Zhang L, Song W. Zinc binding groups for histone deacetylase inhibitors. J Enzym Inhib Med Chem. 2018;33(1):714-721. DOI: 10.1080/14756366.2017.1417274.

Harshita S, Jaya M, Anjali G. Indole derivatives as potential anticancer agents: a review. J Chil Chem Soc. 2020;65(3):4900-4907. DOI: 10.4067/s0717-97072020000204900.

Sahu R, Mishra R, Kumar R, Majee C, Salahuddin, Mazumder A, et al. Pyridine moiety: recent advances in cancer treatment. Indian J Pharm Sci. 2021;83(2):162-185. DOI: 10.36468/pharmaceutical-sciences.763.

Dai Y, Guo Y, Guo J, Pease LJ, Li J, Marcotte PA, et al. Indole amide hydroxamic acids as potent inhibitors of histone deacetylases. Bioorg Med Chem Lett. 2003;13(11):1897-1901. DOI: 10.1016/s0960-894x(03)00301-9.

Yang F, Zhao N, Hu Y, Jiang CS, Zhang H. The development process: from SAHA to hydroxamate HDAC inhibitors with branched CAP region and linear linker. Chem Biodivers. 2020;17(1):e1900427. DOI: 10.1002/cbdv.201900427.

Cho M, Choi E, Yang JS, Lee C, Seo JJ, Kim BS, et al. Discovery of pyridone‐based histone deacetylase inhibitors: approaches for metabolic stability. Chem Med Chem. 2013;8(2):272-279. DOI: 10.1002/cmdc.201200529.

Frühauf A, Meyer-Almes FJ. Non-hydroxamate zinc-binding groups as warheads for histone deacetylases. Molecules. 2021;26(17):5151. DOI: 10.3390/molecules26175151.

Li X, Peterson YK, Inks ES, Himes RA, Li J, Zhang Y, et al. Class I HDAC inhibitors display different antitumor mechanism in leukemia and prostatic cancer cells depending on their p53 status. J Med Chem. 2018;61(6):2589-2603. DOI: 10.1021/acs.jmedchem.8b00136.

Ning ZQ, Li ZB, Newman MJ, Shan S, Wang XH, Pan DS, et al. Chidamide (CS055/HBI-8000): a new histone deacetylase inhibitor of the benzamide class with antitumor activity and the ability to enhance immune cell-mediated tumor cell cytotoxicity. Cancer Chemother Pharmacol. 2012;69(4):901-909. DOI: 10.1007/s00280-011-1766-x.

Katner AS. An improved synthesis of indole-3-carboxylic acids. Org Prep Proced. 1970;2(4):297-303. DOI: 10.1080/00304947009458633.

Bullock MW, Fox SW. A convenient synthesis of indole-3-acetic acids. J Am Chem Soc. 1951;73(11):5155-51557. DOI: 10.1002/chin.199437104.

Hu Y, Ruan W, Gao A, Zhou Y, Gao L, Xu M, et al. Synthesis and biological evaluation of novel 4, 5-bisindolyl-1, 2, 4-triazol-3-ones as glycogen synthase kinase-3β inhibitors and neuroprotective agents. Pharmazie. 2017;72(12):707-713. DOI: 10.1691/ph.2017.7722.

Song ZL, Zhu Y, Liu JR, Guo SK, Gu YC, Han X, et al. Diversity-oriented synthesis and antifungal activities of novel pimprinine derivative bearing a 1, 3, 4-oxadiazole-5-thioether moiety. Mol Divers. 2021;25(1):205-221. DOI: 10.1007/s11030-020-10048-8.

Jirousek M, Robert, Paal M, Ruhter G, Schotten T, Takeuchi K, Stenzel W, inventors; VONDRAN-JONES, MaCharri, Agents. Hypoglycemic imidazoline compounds 1999. Publication No. WO/1999/032482. Available from: https://patentscope.wipo.int/search/en/detail.jsf?docId=WO1999032482.

Ma Y, Luo W, Quinn PJ, Liu Z, Hider RC. Design, synthesis, physicochemical properties, and evaluation of novel iron chelators with fluorescent sensors. J Med Chem. 2004;47(25):6349-6362. DOI: 10.1021/jm049751s.

Zhao DY, Zhang MX, Dong XW, Hu YZ, Dai XY, Wei X, et al. Design and synthesis of novel hydroxypyridinone derivatives as potential tyrosinase inhibitors. Bioorg Med Chem Lett. 2016;26(13):3103-3108. DOI: 10.1016/j.bmcl.2016.05.006.

Saghaie L, Pourfarzam M, Fassihi A, Sartippour B. Synthesis and tyrosinase inhibitory properties of some novel derivatives of kojic acid. Res Pharm Sci. 2013;8(4):233-242.

PMID: 24082892.

Mohammadpour M, Sadeghi A, Fassihi A, Saghaei L, Movahedian A, Rostami M. Synthesis and antioxidant evaluation of some novel ortho-hydroxypyridine-4-one iron chelators. Res Pharm Sci. 2012;7(3):171-179.

PMID: 23181095.

Meerloo JV, Kaspers GJ, Cloos J. Cell sensitivity assays: the MTT assay. Methods Mol Biol. 2011;731:237-245. DOI: 10.1007/978-1-61779-080-5_20.

Biasini M, Bienert S, Waterhouse A, Arnold K, Studer G, Schmidt T, et al. SWISS-MODEL: modelling protein tertiary and quaternary structure using evolutionary information. Nucleic Acids Res. 2014;42(Web Server issue):W252-W258. DOI: 10.1093/nar/gku340.

Lagorce D, Bouslama L, Becot J, Miteva MA, Villoutreix BO. FAF-Drugs4: free ADME-tox filtering computations for chemical biology and early stages drug discovery. Bioinformatics. 2017;33(22):3658-3660.DOI: 10.1093/bioinformatics/btx491.

O'Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open Babel: an open chemical toolbox. J Cheminform. 2011;3(1):33,1-14. DOI: 10.1186/1758-2946-3-33.

Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 2010;31(2):455-461. DOI: 10.1002/jcc.21334.

Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods Mol Biol. 2015;1263:243-250. DOI: 10.1007/978-1-4939-2269-7_19.

Vickers CJ, Olsen CA, Leman LJ, Ghadiri MR. Discovery of HDAC inhibitors that lack an active site Zn2+-binding functional group. ACS Med Chem Let. 2012;3(6):505-508. DOI: 10.1021/ml300081u.

Irto A, Cardiano P, Cataldo S, Chand K, Maria Cigala R, Crea F, et al. Speciation studies of bifunctional 3-hydroxy-4-pyridinone ligands in the presence of Zn2+ at different ionic strengths and temperatures. Molecules. 2019;24(22):4084-4113. DOI: 10.3390/molecules24224084.

Saji S, Kawakami M, Hayashi S, Yoshida N, Hirose M, Horiguchi S, et al. Significance of HDAC6 regulation via estrogen signaling for cell motility and prognosis in estrogen receptor-positive breast cancer. Oncogene. 2005;24(28):4531-4539. DOI: 10.1038/sj.onc.1208646.

Huang M, Zhang J, Yan C, Li X, Zhang J, Ling R. Small molecule HDAC inhibitors: promising agents for breast cancer treatment. Bioorg Chem. 2019;91:103184. DOI: 10.1016/j.bioorg.2019.103184.

Ding J, Liu J, Zhang Z, Guo J, Cheng M, Wan Y, et al. Design, synthesis and biological evaluation of coumarin-based N-hydroxycinnamamide derivatives as novel histone deacetylase inhibitors with anticancer activities. Bioorg Chem. 2020;101:104023.DOI: 10.1016/j.bioorg.2020.104023.

Bingul M, Tan O, Gardner CR, Sutton SK, Arndt GM, Marshall GM, et al. Synthesis, characterization and anticancer activity of hydrazide derivatives incorporating a quinoline moiety. Molecules. 2016;21(7):916-935. DOI: 10.3390/molecules21070916.

Margueron R, Licznar A, Lazennec G, Vignon F, Cavaillès V. Oestrogen receptor alpha increases p21(WAF1/CIP1) gene expression and the antiproliferative activity of histone deacetylase inhibitors in human breast cancer cells. J Endocrinol. 2003;179(1):41-53. DOI: 10.1677/joe.0.1790041.

Im JY, Park H, Kang KW, Choi WS, Kim HS. Modulation of cell cycles and apoptosis by apicidin in estrogen receptor (ER)-positive and-negative human breast cancer cells. Chem Biol Interact. 2008;172(3):235-244. DOI: 10.1016/j.cbi.2008.01.007.

Yari H, Ganjalikhany MR, Sadegh H. In silico investigation of new binding pocket for mitogen activated kinase kinase (MEK): development of new promising inhibitors. Comput Biol Chem. 2015;59 Pt A:185-198. DOI: 10.1016/j.compbiolchem.2015.09.013.

Lauffer BEL, Mintzer R, Fong R, Mukund S, Tam C, Zilberleyb I, et al. Histone deacetylase (HDAC) inhibitor kinetic rate constants correlate with cellular histone acetylation but not transcription and cell viability. J Biol Chem. 2013;288(37):26926-26943. DOI: 10.1074/jbc.m113.490706.


Refbacks

  • There are currently no refbacks.


Creative Commons LicenseThis work is licensed under a Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International License which allows users to read, copy, distribute and make derivative works for non-commercial purposes from the material, as long as the author of the original work is cited properly.