Scopus (CiteScore 2022 =3.0, Q3) , ISC

Document Type : Original Research Article

Authors

1 Doctoral Program of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya-60115, (East Java) Indonesia

2 Department of Pharmacy, Faculty of Medicine and Health Sciences, Maulana Malik Ibrahim State Islamic University, Malang-65144, (East Java) Indonesia

3 Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Surabaya-60115, (East Java) Indonesia

4 Department of Chemistry, Faculty of Science and Technology, Maulana Malik Ibrahim State Islamic University, Malang-65144, (East Java) Indonesia

Abstract

One of the critical strategies in developing new drugs is to design drugs through structural modification. The structural modification leads to changes in the structure of a compound, thereby changing the compound''s physicochemical properties, including lipophilic, electronic, and steric properties. These changes will cause changes in the biological activity of the compound. This study aimed to determine the effect of substituents on the benzoyl group of the compound N-benzoyl-N’-naphthylthiourea (BNTU) on the anti-breast cancer activity of HER-2 in silico. Molecular docking of BNTU lead compound and derivatives using Autodock tools software against HER-2 receptors (PDB ID: 3RCD). Compared to lipophilic and steric properties, electronic properties influence the anti-breast cancer activity of HER-2 on BNTU-derived compounds. The binding score (∆G) of BNTU-derived compounds with strong electronic substituents was more negative than lipophilic and steric substituents. However, there is an anomaly in BNTU derivatives with fluoro (F) substituents because they have the lowest anticancer activity compared to the other BNTU derivatives. Anti-breast cancer activity of BNTU-derived compounds is influenced by variations in substituents, especially by the electronic properties (electron withdrawing groups) of these substituents. Compounds with Br substituents have a better affinity for HER-2 receptors than the lead compound BNTU and other BNTU derivatives. 

Graphical Abstract

In Silico study of the effect of substituents on the structure of N-benzoyl-N’-naphthylthiourea as anti-breast cancer HER-2 positive candidates

Keywords

[1] W. Yang, Y. Hu, Y.S. Yang, F. Zhang, Y. Bin Zhang, X.L. Wang, J.F. Tang, W.Q. Zhong, H.L. Zhu, Design, modification and 3D QSAR studies of novel naphthalin-containing pyrazoline derivatives with/without thiourea skeleton as anticancer agents, Bioorg. Med. Chem., 2013, 21, 1050. [Crossref], [Google Scholar], [Publisher]
[2] B.E. Oyinloye, T.A. Adekiya, R.T. Aruleba, O.A. Ojo, B.O. Ajiboye, Structure-based docking studies of GLUT4 towards exploring selected phytochemicals from Solanum xanthocarpum as a therapeutic target for the treatment of cancer, Curr. Drug Discov. Technol., 2019, 16, 406. [Crossref], [Google Scholar], [Publisher]
[3] H. Sung, J. Ferlay, R. Siegel, M. Laversanne, I. Soerjomataram, A. Jemal, F. Bray, Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries, CA. Cancer J. Clin., 2021, 71, 209–249. [Crossref], [Google Scholar], [Publisher]
[4] H.-S. Seo, J.M. Ku, H.-S. Choi, Y.K. Choi, J.-K. Woo, M. Kim, I. Kim, C.H. Na, H. Hur, B.-H. Jang, Y.C. Shin, S.-G. Ko, Quercetin induces caspase-dependent extrinsic apoptosis through inhibition of signal transducer and activator of transcription 3 signaling in HER2-overexpressing BT-474 breast cancer cells, Oncol. Rep., 2016, 36, 31. [Crossref], [Google Scholar], [Publisher]
[5] T. Widiandani, Siswandono, E. Meiyanto, Anticancer evaluation of N-benzoyl-3-allylthiourea as potential antibreast cancer agent through enhances HER-2 expression, J. Adv. Pharm. Technol. Res., 2020, 11, 163. [Crossref], [Google Scholar], [Publisher]
[6] V. D’Amato, L. Raimondo, L. Formisano, M. Giuliano, S. De Placido, R. Rosa, R. Bianco, Mechanisms of lapatinib resistance in HER2-driven breast cancer, Cancer Treat. Rev., 2015, 41, 877. [Crossref], [Google Scholar], [Publisher]
[7] B. Ruprecht, E.A. Zaal, J. Zecha, W. Wu, C.R. Berkers, B. Kuster, S. Lemeer, Lapatinib resistance in breast cancer cells is accompanied by phosphorylation-mediated reprogramiming of glycolysis, Cancer Res., 2017, 77, 1842. [Crossref], [Google Scholar], [Publisher]
[8] R.L. Carpenter, H.-W. Lo, Regulation of apoptosis by HER2 in breast cancer, J. Carcinog. Mutagen., 2013, 7, 1. [Crossref], [Google Scholar], [Publisher]
[9] A. Patel, N. Unni, Y. Peng, The changing paradigm for the treatment of HER2-positive breast cancer, Cancers, 2020, 12, 1. [Crossref], [Google Scholar], [Publisher]
[10] V. Kumar, S. Krishna, M.I. Siddiqi, Virtual screening strategies: Recent advances in the identification and design of anti-cancer agents, Methods, 2015, 71, 64-70. [Crossref], [Google Scholar], [Publisher]
[11] T. Baudino, Targeted cancer therapy: the next generation of cancer treatment, Curr. Drug Discov. Technol., 2015, 12, 3–20. [Crossref], [Google Scholar], [Publisher]
[12] R. Gerl, D.L. Vaux, Apoptosis in the development and treatment of cancer, J. Carcinog., 2005, 26, 263-270. [Crossref], [Google Scholar], [Publisher]
[13] T. Widiandani, Siswandono, E. Meiyanto, M.I. Sulistyowaty, B.T. Purwanto, S. Hardjono, New N-allylthiourea derivatives: synthesis, molecular docking and in vitro cytotoxicity studies, Trop. J. Pharm. Res., 2018, 17, 1607–1613. [Crossref], [Google Scholar], [Publisher]
[14] M.M. Fallatah, S. Liu, M.B. Sevigny, H. Zou, M.C. Louie, Novel flexible heteroarotinoid, SL-1-18, promotes ERα degradation to inhibit breast cancer cell growth, Cancer Lett., 2017, 408, 82. [Crossref], [Google Scholar], [Publisher]
[15] H. Zou, M.B. Sevigny, S. Liu, D.T. Madden, M.C. Louie, Novel flexible heteroarotinoid, SL-1-39, inhibits HER2-positive breast cancer cell proliferation by promoting lysosomal degradation of HER2, Cancer Lett., 2019, 443, 157. [Crossref], [Google Scholar], [Publisher]
[16] W. Bai, J. Ji, Q. Huang, W. Wei, Synthesis and evaluation of new thiourea derivatives as antitumor and antiangiogenic agents, Tetrahedron Lett., 2020, 61, 152366. [Crossref], [Google Scholar], [Publisher]
[17] R. Pingaew, V. Prachayasittikul, N. Anuwongcharoen, S. Prachayasittikul, S. Ruchirawat, V. Prachayasittikul, Synthesis and molecular docking of N,N’-disubstituted thiourea derivatives as novel aromatase inhibitors, Bioorg. Chem., 2018, 79, 171. [Crossref], [Google Scholar], [Publisher]
[18] G. Kirishnamaline, J.D. Magdaline, T. Chithambarathanu, D. Aruldhas, A.R. Anuf, Theoretical investigation of structure, anticancer activity and molecular docking of thiourea derivatives, J. Mol. Struct., 2020, 1225, 129118. [Crossref], [Google Scholar], [Publisher]
[19] Siswandono, Pengembangan Obat Baru; Airlangga University Press: Surabaya, 2014. [Google Scholar], [Publisher]
[20] S. Hardjono, T. Widiandani, B.T. Purwanto, A.L. Nasyanka, Molecular docking of N-benzoyl-N’-(4-fluorophenyl) thiourea derivatives as anticancer drug candidate and their ADMET prediction, Res. J. Pharm. Technol., 2019, 12, 2160. [Crossref], [Google Scholar], [Publisher]
[21] A.L. Nasyanka, S. Siswodihardjo, S. Hardjono, Docking, synthesis, and cytotoxic activity of N-4-methoxybenzoyl-N’-(4-fluorophenyl)thiourea on HeLa cell line, Thai J. Pharm. Sci., 2017, 41, 99. [PDF], [Google Scholar], [Publisher]
[22] D. Kesuma, A.L. Nasyanka, M. Rudyanto, Siswandono, B.T. Purwanto, I.G.A. Sumartha, A prospective modification structure: the effect of lipophilic and electronic properties of N-(phenylcarbamothyoil)benzamide derivatives on cytotoxic activity by in silico and in vitro assay with T47D cells, Rasayan J. Chem., 2020, 13, 1914. [Crossref], [Google Scholar], [Publisher]
[23] Ruswanto, A.M. Miftah, D.H. Tjahjono, Siswandono, Synthesis and in vitro cytotoxicity of 1-benzoyl-3-methyl thiourea derivatives, Procedia Chem., 2015, 17, 157. [Crossref], [Google Scholar], [Publisher]
[24] R. Suharjo, A.M. Miftah, D.H. Tjahjono, Siswandono, Synthesis and in vitro test of 1-(4-chlorobenzoyl)-3-methyl thiourea on Hela cell, In Proceedings of The 5TH Annual Basic Science International Conference: Malang, 2015, 203. [Publisher]
[25] C. Cava, I. Castiglioni, Integration of molecular docking and in vitro studies: A powerful approach for drug discovery in breast cancer, App. Sci., 2020, 10, 698. [Crossref], [Google Scholar], [Publisher]
[26] G. Syahputra, L. Ambarsari, T. Sumaryada, Simulasi docking kurkumin enol, bismetoksikurkumin dan analognya sebagai inhibitor enzim12-Lipoksigenase, Jurnal Biofisika, 2014, 10, 55. [Google Scholar], [Publisher]
[27] M.R.F. Pratama, E.N. Praditapuspa, D. Kesuma, H. Poerwono, T. Widiandani, S. Siswodihardjo, Boesenbergia pandurata as an anti-breast cancer agent: molecular docking and ADMET study, Lett. Drug Des. Discov., 2022, 19, 606–626. [Crossref], [Google Scholar], [Publisher]
[28] M.R.F. Pratama, H. Poerwono, S. Siswodihardjo, Molecular docking of novel 5-O-benzoylpinostrobin derivatives as SARS CoV-2 main protease inhibitors, Pharm. Sci., 2020, 26, S63. [Crossref], [Google Scholar], [Publisher]
[29] M.R.F. Pratama, Siswandono, Number of runs variations on Autodock 4 do not have a significant effect on RMSD from docking results, Pharm. Pharmacol., 2020, 6, 476. [Crossref], [Google Scholar], [Publisher]
[30] M. Rudrapal, D. Chetia, Virtual screening, molecular docking and QSAR studies in drug discovery and development programme, J. Drug Deliv. Ther., 2020, 10, 225. [Crossref], [Google Scholar], [Publisher]
[31] M.R.F. Pratama, H. Poerwono, S. Siswodihardjo, Design and molecular docking of novel 5-O-Benzoylpinostrobin derivatives as anti-breast cancer, Thai J. Pharm. Sci., 2019, 43, 201. [Google Scholar], [Publisher]
[32] T. Nauli, Penentuan sisi aktif selulase Aspergillus niger dengan docking ligan, J. Kim. Terap. Indones., 2014, 16, 94. [Crossref], [Google Scholar], [Publisher]
[33] F. Azam, M.V.V. Prasad, N. Thangavel, H.I. Ali, Molecular docking studies of 1-(substituted phenyl)-3-(naphtha [1, 2-d] thiazol-2-yl) urea/thiourea derivatives with human adenosine A2A receptor, Bioinformation, 2011, 6, 330. [Crossref], [Google Scholar], [Publisher]
[34] J. G. Topliss, Utilization of operational schemes for analog synthesis in drug design, J. Med. Chem., 1972, 15, 1006–1011. [Crossref], [Google Scholar], [Publisher]