Generic placeholder image

Current Enzyme Inhibition

Editor-in-Chief

ISSN (Print): 1573-4080
ISSN (Online): 1875-6662

Research Article

Rational Drug Design and In vitro Cell Line Studies of Some N-(4-(1Hbenzo[ d]imidazol-2-yl)phenyl)arylamine Derivatives as Aromatase Inhibitors for the Treatment of Cancer

Author(s): Nusrat B. Sayyad and Prafulla M. Sabale*

Volume 19, Issue 1, 2023

Published on: 23 December, 2022

Page: [38 - 48] Pages: 11

DOI: 10.2174/1573408019666221028142316

Price: $65

Open Access Journals Promotions 2
Abstract

Background: Aromatase is a catalytic enzyme involved in the biosynthesis of estrogen from androgen. It catalyzes the last rate-limiting/crucial critical step in estrogen biosynthesis. Following the success of the aromatase inhibitor, researchers are working on developing a small physiologically active molecule with fewer side effects and improved tolerance.

Objectives: Inhibition of the aromatase enzyme, which plays a major role in the rate-limiting phase, is one strategy to prevent estrogen synthesis. After knowing the importance of nitrogen atom containing moieties in the treatment of breast cancer, we have designed some N-(4-(1H-benzo[d]imidazol-2- yl)phenyl)arylamine derivatives through in silico screening such as ADMET analysis and molecular docking studies. From the present investigation, we aimed for the synthesis and biological evaluation of the most potent derivatives obtained in this study.

Methods: The selected derivatives were synthesized and confirmed by spectral analysis (FTIR, 1H NMR, and Mass). Cytotoxic activity of the compounds was evaluated by colorimetric MTT assay on MDA-MB-231 (breast adenocarcinoma), MCF-7(breast adenocarcinoma), A549 (lung adenocarcinoma) NCI-H23 (Lung carcinoma) and A-498 (Renal carcinoma) cell line using Doxorubicin hydrochloride as a positive control.

Results: From the present investigation, we have concluded that compound 10 [N-(4-(1Hbenzo[ d]imidazol-2-yl)phenyl)-1H-benzo[d]imidazol-5-amine) is most potent and exhibited -9.5 kcal/mol binding affinity. It has formed conventional hydrogen bonds with ALA306 and THR310. It displayed most promising activity with GI50 values 0.796 ± 0.06 μM, 0.695 ± 0.05 μM, 1.14 ± 0.06 μM, 2.15 ± 0.04 μM, and 0.987 ± 0.07 μM against MDAMB-231, MCF-7, A-549, NCI-H23, and A- 498, respectively when compared with Doxorubicin (0.306 ± 0.04 μM, 0.270 ± 0.02 μM, 0.297 ± 0.04 μM, 0.305 ± 0.04 μM, and 0.345 ± 0.09 μM).

Conclusion: From the present investigation, it is concluded that the designed molecules had the potential to be developed as broad-spectrum anticancer agents.

Keywords: Aromatase, inhibitors, breast cancer, ADMET, in silico, imidazole.

Graphical Abstract
[1]
Yadav MR, Barmade MA, Tamboli RS, Murumkar PR. Developing steroidal aromatase inhibitors-an effective armament to win the battle against breast cancer. Eur J Med Chem 2015; 105: 1-38.
[http://dx.doi.org/10.1016/j.ejmech.2015.09.038] [PMID: 26469743]
[2]
Bulard J, Mowszowicz I, Schaison G. Increased aromatase activity in pubic skin fibroblasts from patients with isolated gynecomastia. J Clin Endocrinol Metab 1987; 64(3): 618-23.
[http://dx.doi.org/10.1210/jcem-64-3-618] [PMID: 3818893]
[3]
Berkovitz GD, Guerami A, Brown TR, MacDonald PC, Migeon CJ. Familial gynecomastia with increased extraglandular aromatization of plasma carbon19-steroids. J Clin Invest 1985; 75(6): 1763-9.
[http://dx.doi.org/10.1172/JCI111888] [PMID: 3924954]
[4]
Ji J, Lao K, Hu J, et al. Discovery of novel aromatase inhibitors using a homogeneous time-resolved fluorescence assay. Acta Pharmacol Sin 2014; 35(8): 1082-92.
[http://dx.doi.org/10.1038/aps.2014.53] [PMID: 25047514]
[5]
Zubeldia-Brenner L, Roselli CE, Recabarren SE, Gonzalez Deniselle MC, Lara HE. Developmental and functional effects of steroid hormones on the neuroendocrine axis and spinal cord. J Neuroendocrinol 2016; 28(7): jne.12401.
[http://dx.doi.org/10.1111/jne.12401] [PMID: 27262161]
[6]
Ratre P, Mishra K, Dubey A, Vyas A, Jain A, Thareja S. Aromatase inhibitors for the treatment of breast cancer: A journey from the scratch. Anticancer Agents Med Chem 2020; 20(17): 1994-2004.
[http://dx.doi.org/10.2174/1871520620666200627204105] [PMID: 32593281]
[7]
Bradley R, Burrett J, Clarke M, et al. Aromatase inhibitors versus tamoxifen in early breast cancer: Patient-level meta-analysis of the randomised trials. Lancet 2015; 386(10001): 1341-52.
[http://dx.doi.org/10.1016/S0140-6736(15)61074-1] [PMID: 26211827]
[8]
Chumsri S, Howes T, Bao T, Sabnis G, Brodie A. Aromatase, aromatase inhibitors, and breast cancer. J Steroid Biochem Mol Biol 2011; 125(1-2): 13-22.
[http://dx.doi.org/10.1016/j.jsbmb.2011.02.001] [PMID: 21335088]
[9]
Ghodsi R, Hemmateenejad B. QSAR study of diarylalkylimidazole and diarylalkyltriazole aromatase inhibitors. Med Chem Res 2016; 25(5): 834-42.
[http://dx.doi.org/10.1007/s00044-016-1530-1]
[10]
Bae SH, Park JH, Choi HG, Kim H, Kim SH. Imidazole antifungal drugs inhibit the cell proliferation and invasion of human breast cancer cells. Biomol Ther 2018; 26(5): 494-502.
[http://dx.doi.org/10.4062/biomolther.2018.042] [PMID: 30092625]
[11]
Ballard SA, Lodola A, Tarbit MH. A comparative study of 1-substituted imidazole and 1,2,4-triazole antifungal compounds as inhibitors of testosterone hydroxylations catalysed by mouse hepatic musomal cytochromes P-450. Biochem Pharmacol 1988; 37(24): 4643-51.
[http://dx.doi.org/10.1016/0006-2952(88)90333-4] [PMID: 3202901]
[12]
Trösken ER, Fischer K, Völkel W, Lutz WK. Inhibition of human CYP19 by azoles used as antifungal agents and aromatase inhibitors, using a new LC–MS/MS method for the analysis of estradiol product formation. Toxicology 2006; 219(1-3): 33-40.
[http://dx.doi.org/10.1016/j.tox.2005.10.020] [PMID: 16330141]
[13]
Avendaño C, Menéndez JC. Anticancer drugs that modulate hormone action.In: Medicinal Chemistry of Anticancer Drugs. Amsterdam: Elsevier 2015; pp. 81-131.
[http://dx.doi.org/10.1016/B978-0-444-62649-3.00003-X]
[14]
Kim S, Chen J, Cheng T, et al. PubChem in 2021: New data content and improved web interfaces. Nucleic Acids Res 2021; 49(D1): D1388-95.
[http://dx.doi.org/10.1093/nar/gkaa971] [PMID: 33151290]
[15]
Daina A, Michielin O, Zoete V. SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep 2017; 7(1): 42717.
[http://dx.doi.org/10.1038/srep42717] [PMID: 28256516]
[16]
Banerjee P, Eckert AO, Schrey AK, Preissner R. ProTox-II: A webserver for the prediction of toxicity of chemicals. Nucleic Acids Res 2018; 46(W1)W257-63
[http://dx.doi.org/10.1093/nar/gky318] [PMID: 29718510]
[17]
Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods Mol Biol 2015; 1263(1263): 243-50.
[http://dx.doi.org/10.1007/978-1-4939-2269-7_19] [PMID: 25618350]
[18]
Rappé AK, Casewit CJ, Colwell KS, Goddard WA III, Skiff WM. UFF, a full periodic table force field for molecular mechanics and molecular dynamics simulations. J Am Chem Soc 1992; 114(25): 10024-35.
[http://dx.doi.org/10.1021/ja00051a040]
[19]
Discovery Studio Modeling Environment. San Diego: Accelrys Software Inc. 2012.
[20]
Khan SL, Siddiqui FA, Jain SP, Sonwane GM. Discovery of potential inhibitors of SARS-CoV-2 (COVID-19) Main Protease (Mpro) from Nigella sativa (Black Seed) by molecular docking study. Coronaviruses 2021; 2(3): 384-402.
[http://dx.doi.org/10.2174/2666796701999200921094103]
[21]
Chaudhari RN, Khan SL, Chaudhary RS, Jain SP, Siddiqui FA. B-sitosterol: Isolation from Muntingia calabura linn bark extract, structural elucidation and molecular docking studies as potential inhibitor of sars-cov-2 Mpro (COVID-19). Asian J Pharm Clin Res 2020; 13(5): 204-9.
[http://dx.doi.org/10.22159/ajpcr.2020.v13i5.37909]
[22]
Khan SL, Siddiqui FA, Shaikh MS, Nema NV, Shaikh AA. Discovery of potential inhibitors of the Receptor-Binding Domain (RBD) of pandemic disease-causing sars-cov-2 spike glycoprotein from triphala through molecular docking. Curr Chinese Chem 2021; 01.
[http://dx.doi.org/10.2174/2666001601666210322121802]
[23]
Khan SL, Sonwane GM, Siddiqui FA, Jain SP, Kale MA, Borkar VS. Discovery of naturally occurring flavonoids as human cytochrome P450 (CYP3A4) inhibitors with the aid of computational chemistry. Indo Global J Pharm Sci 2020; 10(4): 58-69.
[http://dx.doi.org/10.35652/IGJPS.2020.10409]
[24]
Siddiqui FA, Khan SL, Marathe RP, Nema NV. Design, synthesis, and in silico studies of novel N-(2-aminophenyl)-2,3- diphenylquinoxaline-6-sulfonamide derivatives targeting Receptor- Binding Domain (RBD) of sars-cov-2 spike glycoprotein and their evaluation as antimicrobial and antimalarial agents. Lett Drug Des Discov 2021; 18(9): 915-31.
[http://dx.doi.org/10.2174/1570180818666210427095203]
[25]
Baviskar B, Baviskar B, Chuadhary S, et al. Synthesis of novel benzimidazole derivatives as potent antimicrobial agent. Rasayan J Chem 2009; 2(1): 186-90.
[26]
Khattab M, Ragab F, Galal S, El Diwani H. Synthesis of 4-(1H-Benzo[d]Imidazol-2-Yl) aniline derivatives of expected anti-HCV activity. Int J Res Pharm Chem 2012; 2(4): 937-46.
[27]
Shenvi S, Kumar K, Hatti KS, Rijesh K, Diwakar L, Reddy GC. Synthesis, anticancer and antioxidant activities of 2,4,5-trimethoxy chalcones and analogues from asaronaldehyde: Structure–activity relationship. Eur J Med Chem 2013; 62: 435-42.
[http://dx.doi.org/10.1016/j.ejmech.2013.01.018] [PMID: 23395966]
[28]
Basu P, Maier C. Phytoestrogens and breast cancer: In vitro anticancer activities of isoflavones, lignans, coumestans, stilbenes and their analogs and derivatives. Biomed Pharmacother 2018; 107: 1648-66.
[http://dx.doi.org/10.1016/j.biopha.2018.08.100] [PMID: 30257383]
[29]
Ameta KL, Rathore N, Kumar B. Synthesis and in vitro anti breast cancer activity of some novel 1,5-benzothiazepine derivatives. J Serb Chem Soc 2012; 77(6): 725-31.
[http://dx.doi.org/10.2298/JSC110715219A]
[30]
Khan A, Unnisa A, Sohel M, et al. Investigation of phytoconstituents of Enicostemma littorale as potential glucokinase activators through molecular docking for the treatment of type 2 diabetes mellitus. In Silico Pharmacol 2022; 10(1): 1.
[http://dx.doi.org/10.1007/s40203-021-00116-8] [PMID: 34926125]
[31]
Khan S, Kale M, Siddiqui F, Nema N. Novel pyrimidine-benzimidazole hybrids with antibacterial and antifungal properties and potential inhibition of SARS-CoV-2 main protease and spike glycoprotein. Digital Chinese Med 2021; 4(2): 102-19.
[http://dx.doi.org/10.1016/j.dcmed.2021.06.004]
[32]
Krzywinski M, Altman N. Significance, P values and t-tests. Nat Methods 2013; 10(11): 1041-2.
[http://dx.doi.org/10.1038/nmeth.2698] [PMID: 24344377]
[33]
Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2012; 64: 4-17.
[http://dx.doi.org/10.1016/j.addr.2012.09.019] [PMID: 11259830]
[34]
Shntaif AH, Khan S, Tapadiya G, et al. Rational drug design, synthesis, and biological evaluation of novel N-(2-arylaminophenyl)-2,3-diphenylquinoxaline-6-sulfonamides as potential antimalarial, antifungal, and antibacterial agents. Digital Chinese Med 2021; 4(4): 290-304.
[http://dx.doi.org/10.1016/j.dcmed.2021.12.004]

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