Generic placeholder image

Current Drug Research Reviews

Editor-in-Chief

ISSN (Print): 2589-9775
ISSN (Online): 2589-9783

Research Article

In-silico Design, ADMET Screening, MM-GBSA Binding Free Energy of Some Novel Isoxazole Substituted 9-Anilinoacridines as HER2 Inhibitors Targeting Breast Cancer

Author(s): Rajagopal Kalirajan*, Arumugasamy Pandiselvi, Byran Gowramma and Pandiyan Balachandran

Volume 11, Issue 2, 2019

Page: [118 - 128] Pages: 11

DOI: 10.2174/2589977511666190912154817

Abstract

Background: Human Epidermal development factor Receptor-2 (HER2) is a membrane tyrosine kinase which is overexpressed and gene amplified in human breast cancers. HER2 amplification and overexpression have been linked to important tumor cell proliferation and survival pathways for 20% of instances of breast cancer. 9-aminoacridines are significant DNA-intercalating agents because of their antiproliferative properties.

Objective: Some novel isoxazole substituted 9-anilinoacridines(1a-z) were designed by in-silico technique for their HER2 inhibitory activity. Docking investigations of compounds 1a-z are performed against HER2 (PDB id-3PP0) by using Schrodinger suit 2016-2.

Methods: Molecular docking study for the designed molecules 1a-z are performed by Glide module, in-silico ADMET screening by QikProp module and binding free energy by Prime-MMGBSA module of Schrodinger suit. The binding affinity of designed molecules 1a-z towards HER2 was chosen based on GLIDE score.

Results: Many compounds showed good hydrophobic communications and hydrogen bonding associations to hinder HER2. The compounds 1a-z, aside from 1z have significant Glide scores in the scope of - 4.91 to - 10.59 when compared with the standard Ethacridine (- 4.23) and Tamoxifen (- 3.78). The in-silico ADMET properties are inside the suggested about drug likeness. MM-GBSA binding of the most intense inhibitor is positive.

Conclusion: The outcomes reveal that this study provides evidence for the consideration of isoxazole substituted 9-aminoacridine derivatives as potential HER2 inhibitors. The compounds, 1s,x,v,a,j,r with significant Glide scores may produce significant anti breast cancer activity and further in vitro and in vivo investigations may prove their therapeutic potential.

Keywords: Acridine, isoxazole, docking studies, in-silico ADMET screening, MM-GBSA, HER2.

Graphical Abstract
[1]
Baselga J, Tripathy D, Mendelsohn J, et al. Phase II study of weekly intravenous recombinant humanized anti-p185her2 monoclonal antibody in patients with HER2/neu-overexpressing metastatic breast cancer. J Clin Oncol 1996; 14(3): 737-44.
[http://dx.doi.org/10.1200/JCO.1996.14.3.737] [PMID: 8622019]
[2]
Baselga J, Swain SM. CLEOPATRA, a phase III evaluation of pertuzumab and trastuzumab for HER2-positive metastatic breast cancer. Clin Breast Cancer 2010; 10(6): 489-91.
[http://dx.doi.org/10.3816/CBC.2010.n.065] [PMID: 21147694]
[3]
Burris HA, Rugo HS, Vukelja SJ, et al. Phase II study of the antibody drug conjugate trastuzumab-DM1 for the treatment of H Human epidermal growth factor R Receptor 2 (HER2)-positive breast cancer after prior HER2-directed therapy. J Clin Oncol 2011; 29(4): 398-405.
[http://dx.doi.org/10.1200/JCO.2010.29.5865] [PMID: 21172893]
[4]
Gajria D, Chandarlapaty S. HER2-amplified breast cancer: Mechanisms of trastuzumab resistance and novel targeted therapies. Expert Rev Anticancer Ther 2011; 11(2): 263-75.
[http://dx.doi.org/10.1586/era.10.226] [PMID: 21342044]
[5]
Eccles SA. The role of c-erbb-2/HER2/neu in breast cancer progression and metastasis. J Mammary Gland Biol Neoplasia 2001; 6(4): 393-406.
[http://dx.doi.org/10.1023/A:1014730829872] [PMID: 12013529]
[6]
Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol 2010; 28(7): 1138-44.
[http://dx.doi.org/10.1200/JCO.2009.24.2024] [PMID: 20124182]
[7]
Cardoso F, Durbecq V, Laes JF, et al. Bortezomib (PS-341, Velcade) increases the efficacy of trastuzumab (Herceptin) in HER-2-positive breast cancer cells in a synergistic manner. Mol Cancer Ther 2006; 5(12): 3042-51.
[http://dx.doi.org/10.1158/1535-7163.MCT-06-0104] [PMID: 17148762]
[8]
Nielsen DL, Andersson M, Kamby C. HER2-targeted therapy in breast cancer. Monoclonal antibodies and tyrosine kinase inhibitors. Cancer Treat Rev 2009; 35(2): 121-36.
[http://dx.doi.org/10.1016/j.ctrv.2008.09.003] [PMID: 19008049]
[9]
Hoppea S, Sparidans RW, Wagenaar E, Beijnenb JH, Schinkel AH. Breast cancer resistance protein (BCRP/ABCG2) and P-glycoprotein (P-gp/ABCB1) transport afatinib and restrict its oral availability andbrain accumulation. Pharmacol Res 2017; 120: 43-50.
[http://dx.doi.org/10.1016/j.phrs.2017.01.035] [PMID: 28288939]
[10]
Cheng Q, Chang JT, Geradts J, et al. Amplification and high-level expression of heat shock protein 90 marks aggressive phenotypes of human epidermal growth factor receptor 2 negative breast cancer. Breast Cancer Res 2012; 14(2): R62.
[http://dx.doi.org/10.1186/bcr3168] [PMID: 22510516]
[11]
Chang BY, Kim SA, Malla B, Kim SY. The effect of Selective Estrogen Receptor Modulators (SERMS) on the tamoxifen resistant breast cancer cells. Toxicol Res 2011; 27(2): 85-93.
[http://dx.doi.org/10.5487/TR.2011.27.2.085] [PMID: 24278556]
[12]
Musiliyu A, Musaa M, Omar F, Khanb JSC. Synthesis and antiproliferative activity of coumarin-estrogen conjugates against breast cancer cell lines. Lett Drug Des Discov 2012; 76: 211-20.
[13]
Luo G, Chen M, Lyu W, et al. Design, synthesis, biological evaluation and molecular docking studies of novel 3-aryl-4-anilino-2H-chromen-2-one derivatives targeting ERα as anti-breast cancer agents. Bioorg Med Chem Lett 2017; 27(12): 2668-73.
[http://dx.doi.org/10.1016/j.bmcl.2017.04.029] [PMID: 28460819]
[14]
Berger C, Qian Y, Chen X. The p53-estrogen receptor loop in cancer. Curr Mol Med 2013; 13(8): 1229-40.
[http://dx.doi.org/10.2174/15665240113139990065] [PMID: 23865427]
[15]
Menendez JA, Lupu R. Fatty acid synthase regulates estrogen receptor-α signaling in breast cancer cells. Oncogenesis 2017; 6(2)e299
[http://dx.doi.org/10.1038/oncsis.2017.4] [PMID: 28240737]
[16]
Harmey JH, Dimitriadis E, Kay E, Redmond HP, Bouchier-Hayes D. Regulation of macrophage production of Vascular Endothelial Growth Factor (VEGF) by hypoxia and transforming growth factor beta-1. Ann Surg Oncol 1998; 5(3): 271-8.
[http://dx.doi.org/10.1007/BF02303785] [PMID: 9607631]
[17]
Lai L, Liu J, Zhai D, et al. Plumbagin inhibits tumour angiogenesis and tumour growth through the Ras signalling pathway following activation of the VEGF receptor-2. Br J Pharmacol 2012; 165(4b): 1084-96.
[http://dx.doi.org/10.1111/j.1476-5381.2011.01532.x] [PMID: 21658027]
[18]
Miller KD, Trigo JM, Wheeler C, et al. A multicenter phase II trial of ZD6474, a vascular endothelial growth factor receptor-2 and epidermal growth factor receptor tyrosine kinase inhibitor, in patients with previously treated metastatic breast cancer. Clin Cancer Res 2005; 11(9): 3369-76.
[http://dx.doi.org/10.1158/1078-0432.CCR-04-1923] [PMID: 15867237]
[19]
Gianni L, Lladó A, Bianchi G, et al. Open-label, phase II, multicenter, randomized study of the efficacy and safety of two dose levels of Pertuzumab, a human epidermal growth factor receptor 2 dimerization inhibitor, in patients with human epidermal growth factor receptor 2-negative metastatic breast cancer. J Clin Oncol 2010; 28(7): 1131-7.
[http://dx.doi.org/10.1200/JCO.2009.24.1661] [PMID: 20124183]
[20]
Perez EA, Romond EH, Suman VJ, et al. Four-year follow-up of trastuzumab plus adjuvant chemotherapy for operable human epidermal growth factor receptor 2-positive breast cancer: Joint analysis of data from NCCTG N9831 and NSABP B-31. J Clin Oncol 2011; 29(25): 3366-73.
[http://dx.doi.org/10.1200/JCO.2011.35.0868] [PMID: 21768458]
[21]
Bhargava R, Gerald WL, Li AR, et al. EGFR gene amplification in breast cancer: Correlation with epidermal growth factor receptor mrna and protein expression and HER-2 status and absence of EGFR-activating mutations. Mod Pathol 2005; 18(8): 1027-33.
[http://dx.doi.org/10.1038/modpathol.3800438] [PMID: 15920544]
[22]
Sasaki T, Hiroki K, Yamashita Y. The role of epidermal growth factor receptor in cancer metastasis and microenvironment. BioMed Res Int 2013; 2013546318
[http://dx.doi.org/10.1155/2013/546318] [PMID: 23986907]
[23]
Buzdar AU, Ibrahim NK, Francis D, et al. Significantly higher pathologic complete remission rate after neoadjuvant therapy with trastuzumab, paclitaxel, and epirubicin chemotherapy: Results of a randomized trial in human epidermal growth factor receptor 2-positive operable breast cancer. J Clin Oncol 2005; 23(16): 3676-85.
[http://dx.doi.org/10.1200/JCO.2005.07.032] [PMID: 15738535]
[24]
Real PJ, Benito A, Cuevas J, et al. Blockade of epidermal growth factor receptors chemosensitizes breast cancer cells through up-regulation of BNIP3L. Cancer Res 2005; 65(18): 8151-7.
[http://dx.doi.org/10.1158/0008-5472.CAN-05-1134] [PMID: 16166289]
[25]
Langelier MF, Ruhl DD, Planck JL, Kraus WL, Pascal JM. The Zn3 domain of human poly (ADP-ribose) polymerase-1 (PARP-1) functions in both DNA-dependent poly (ADP-ribose) synthesis activity and chromatin compaction. J Biol Chem 2010; 285(24): 18877-87.
[http://dx.doi.org/10.1074/jbc.M110.105668] [PMID: 20388712]
[26]
Lord CJ, Ashworth A. Targeted therapy for cancer using PARP inhibitors. Curr Opin Pharmacol 2008; 8(4): 363-9.
[http://dx.doi.org/10.1016/j.coph.2008.06.016] [PMID: 18644251]
[27]
Yuan Z, Chen S, Chen C, et al. Design, synthesis and biological evaluation of 4-amidobenzimidazoleacridine derivatives as dual PARP and Topoinhibitors for cancer therapy. Eur J Med Chem 2017; 138: 1135-46.
[http://dx.doi.org/10.1016/j.ejmech.2017.07.050] [PMID: 28763648]
[28]
Murai J, Zhang Y, Morris J, et al. Rationale for Poly (ADP-Ribose) Polymerase (PARP) inhibitors in combination therapy with camptothecins or temozolomide based on PARP trapping versus catalytic inhibition. J Pharmacol Exp Ther 2014; 349(3): 408-16.
[http://dx.doi.org/10.1124/jpet.113.210146] [PMID: 24650937]
[29]
Bai P, Cantó C. The role of PARP-1 and PARP-2 enzymes in metabolic regulation and disease. Cell Metab 2012; 16(3): 290-5.
[http://dx.doi.org/10.1016/j.cmet.2012.06.016] [PMID: 22921416]
[30]
Lee JM, Hays JL, Annunziata CM, et al. Phase I/Ib study of olaparib and carboplatin in BRCA1 or BRCA2 mutation-associated breast or ovarian cancer with biomarker analyses. J Natl Cancer Inst 2014; 106(6)dju089
[http://dx.doi.org/10.1093/jnci/dju089] [PMID: 24842883]
[31]
Qin T, Huang G, Chi L, et al. Exceptionally high UBE2C expression is a unique phenomenon in basal-like type breast cancer and is regulated by BRCA1. Biomed Pharmacother 2017; 95: 649-55.
[http://dx.doi.org/10.1016/j.biopha.2017.08.095] [PMID: 28881292]
[32]
Yun MH, Hiom K. Ctip-BRCA1 modulates the choice of DNA double-strand-break repair pathway throughout the cell cycle. Nature 2009; 459(7245): 460-3.
[http://dx.doi.org/10.1038/nature07955] [PMID: 19357644]
[33]
Zarrizi R, Menard J, Belting M, Massoumi R. Deubiquitination of γ -tubulin by BAP1 prevents chromosome instability in breast cancer cells. Cancer Res 2014; 0221-27.
[34]
Papoutsis AJ, Lamore SD, Wondrak GT, Selmin OI, Romagnolo DF. Resveratrol prevents epigenetic silencing of BRCA-1 by the aromatic hydrocarbon receptor in human breast cancer cells. J Nutr 2010; 140(9): 1607-14.
[http://dx.doi.org/10.3945/jn.110.123422] [PMID: 20631324]
[35]
Ocaña A, Amir E. Irreversible pan-erbb tyrosine kinase inhibitors and breast cancer: Current status and future directions. Cancer Treat Rev 2009; 35(8): 685-91.
[http://dx.doi.org/10.1016/j.ctrv.2009.08.001] [PMID: 19733440]
[36]
Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther 2005; 315(3): 971-9.
[http://dx.doi.org/10.1124/jpet.105.084145] [PMID: 16002463]
[37]
Pentassuglia L, Graf M, Lane H, et al. Inhibition of erbb2 by receptor tyrosine kinase inhibitors causes myofibrillar structural damage without cell death in adult rat cardiomyocytes. Exp Cell Res 2009; 315(7): 1302-12.
[http://dx.doi.org/10.1016/j.yexcr.2009.02.001] [PMID: 19331811]
[38]
Samir HR, Ratn DS, Dilip VJ, Hitesh BP, Shailesh KM. Recent developments in receptor tyrosine kinases targeted anticancer therapy. Vet World 2016; 9: 80-90.
[http://dx.doi.org/10.14202/vetworld.2016.80-90]
[39]
O’Sullivan CC. CDK4/6 inhibitors for the treatment of advanced hormone receptor positive breast cancer and beyond: 2016 update. Expert Opin Pharmacother 2016; 17(12): 1657-67.
[http://dx.doi.org/10.1080/14656566.2016.1201072] [PMID: 27322766]
[40]
Prall OW, Sarcevic B, Musgrove EA, Watts CK, Sutherland RL. Estrogen-induced activation of CDK4 and CDK2 during G1-S phase progression is accompanied by increased cyclin D1 expression and decreased cyclin-dependent kinase inhibitor association with cyclin E-Cdk2. J Biol Chem 1997; 272(16): 10882-94.
[http://dx.doi.org/10.1074/jbc.272.16.10882] [PMID: 9099745]
[41]
Tagliabue E, Balsari A, Campiglio M, Pupa SM. HER2 as a target for breast cancer therapy. Expert Opin Biol Ther 2010; 10(5): 711-24.
[http://dx.doi.org/10.1517/14712591003689972] [PMID: 20214497]
[42]
Gutierrez C, Schiff R. HER2 biology, detection, and clinical implications. Arch Pathol Lab Med 2011; 135(1): 55-62.
[PMID: 21204711]
[43]
Kallioniemi OP, Kallioniemi A, Kurisu W, et al. ERBB2 amplification in breast cancer analyzed by fluorescence in situ hybridization. Proc Natl Acad Sci USA 1992; 89(12): 5321-5.
[http://dx.doi.org/10.1073/pnas.89.12.5321] [PMID: 1351679]
[44]
Kapuriya N, Kapuriya K, Zhang X, et al. Synthesis and biological activity of stable and potent antitumor agents, aniline nitrogen mustards linked to 9-anilinoacridines via a urea linkage. Bioorg Med Chem 2008; 16(10): 5413-23.
[http://dx.doi.org/10.1016/j.bmc.2008.04.024] [PMID: 18450456]
[45]
Sun YW, Chen KY, Kwon CH, Chen KM. CK0403, a 9-aminoacridine, is a potent anti-cancer agent in human breast cancer cells. Mol Med Rep 2016; 13(1): 933-8.
[http://dx.doi.org/10.3892/mmr.2015.4604] [PMID: 26648164]
[46]
Wakelin LPG, Bu X, Eleftheriou A, Parmar A, Hayek C, Stewart BW. Bisintercalating threading diacridines: Relationships between DNA binding, cytotoxicity, and cell cycle arrest. J Med Chem 2003; 46(26): 5790-802.
[http://dx.doi.org/10.1021/jm030253d] [PMID: 14667232]
[47]
Atwell GJ, Cain BF, Baguley BC, Finlay GJ, Denny WA. Potential antitumor agents. Part 43. Synthesis and biological activity of dibasic 9-aminoacridine-4-carboxamides, a new class of antitumor agent. J Med Chem 1984; 27(11): 1481-5.
[http://dx.doi.org/10.1021/jm00377a017] [PMID: 6492078]
[48]
Bacherikov VA, Chang JY, Lin YW, et al. Synthesis and antitumor activity of 5-(9-acridinylamino)anisidine derivatives. Bioorg Med Chem 2005; 13(23): 6513-20.
[http://dx.doi.org/10.1016/j.bmc.2005.07.018] [PMID: 16140018]
[49]
Yang P, Yang Q, Qian X. Novel DNA bis-inteclators of isoquinolino [4, 5-bc] acridines: Design, synthesis and evaluation of cytotoxic activities. Tetrahedron 2005; 61: 11895-901.
[http://dx.doi.org/10.1016/j.tet.2005.09.065]
[50]
Rewcastle GW, Atwell GJ, Chambers D, Baguley BC, Denny WA. Potential antitumor agents. 46. Structure-activity relationships for acridine monosubstituted derivatives of the antitumor agent N-[2-(dimethylamino)ethyl]-9-aminoacridine-4-carboxamide. J Med Chem 1986; 29(4): 472-7.
[http://dx.doi.org/10.1021/jm00154a008] [PMID: 3959026]
[51]
Plouvier B, Houssin R, Hecquet B, et al. Antitumor combilexin. A thiazole-containing analogue of netropsin linked to an acridine chromophore. Bioconjug Chem 1994; 5(5): 475-81.
[http://dx.doi.org/10.1021/bc00029a016] [PMID: 7849080]
[52]
Kalirajan R, Gaurav K, Pandiselvi A, Gowramma B, Sankar S. Novel Thiazine substituted 9-Anilinoacridines: Synthesis, antitumour activity and structure-activity relationships. Anticancer Agents Med Chem 2019; 11: 1350-8.
[http://dx.doi.org/10.2174/1871520619666190408134224]
[53]
Cholody WM, Hernandez L, Hassner L, Scudiero DA, Djurickovic DB, Michejda CJ. Bisimidazoacridones and related compounds: New antineoplastic agents with high selectivity against colon tumors. J Med Chem 1995; 38(16): 3043-52.
[http://dx.doi.org/10.1021/jm00016a007] [PMID: 7636867]
[54]
Tabarrini O, Cecchetti V, Fravolini A, et al. Design and synthesis of modified quinolones as antitumoral acridones. J Med Chem 1999; 42(12): 2136-44.
[http://dx.doi.org/10.1021/jm980324m] [PMID: 10377219]
[55]
Antonini I, Polucci P, Jenkins TC, et al. 1-[(ω-aminoalkyl)amino]-4-[N-(ω-aminoalkyl)carbamoyl]-9-oxo-9, 10-dihydroacridines as intercalating cytotoxic agents, synthesis, DNA binding and biological evaluation. J Med Chem 1997; 40(23): 3749-55.
[http://dx.doi.org/10.1021/jm970114u] [PMID: 9371240]
[56]
Gary DJ, Gordon WG, Miles PH. Potential DNA bis-intercalating agents: Synthesis and antitumor activity of novel, conformationally restricted bis(9-Aminoacridine). J Het Chem 1987; 24: 1405-8.
[57]
Nadaraj V, Selvi ST, Mohan S. Microwave-induced synthesis and anti-microbial activities of 7, 10, 11, 12-tetrahydrobenzo[c]acridin-8(9H)-one derivatives. Eur J Med Chem 2009; 44(3): 976-80.
[http://dx.doi.org/10.1016/j.ejmech.2008.07.004] [PMID: 18718695]
[58]
Kalirajan R. Mohammed Rafick MH, Sankar S, Gowramma B. Green synthesis of some novel chalcone and isoxazole substituted 9-anilinoacridine derivatives and evaluation of their antimicrobial and larvicidal activities. Indian J Chem 2018; 57B: 583-90.
[59]
Kalirajan R, Muralidharan V, Selvaraj J, Sankar S. Microwave assisted synthesis, characterization and evaluation for their antimicrobial activities of some novel pyrazole substituted 9-anilino acridine derivatives. Int J Health Allied Sci 2013; 2(2): 81-7.
[http://dx.doi.org/10.4103/2278-344X.115682]
[60]
Dickens BF, Weglicki WB, Boehme PA, Mak TI. Antioxidant and lysosomotropic properties of acridine-propranolol: Protection against oxidative endothelial cell injury. J Mol Cell Cardiol 2002; 34(2): 129-37.
[http://dx.doi.org/10.1006/jmcc.2001.1495] [PMID: 11851353]
[61]
Kalirajan R, Muralidharan V, Jubie S, et al. Synthesis of some novel pyrazole substituted 9-anilinoacridine derivatives and evaluation for their antioxidant and cytotoxic activities. J Heterocycl Chem 2012; 49: 748-54.
[http://dx.doi.org/10.1002/jhet.848]
[62]
Kalirajan R, Rafick MH, Sankar S, Jubie S. Docking studies, synthesis, characterization and evaluation of their antioxidant and cytotoxic activities of some novel isoxazole-substituted 9-anilinoacridine derivatives. ScientificWorldJournal 2012; 2012165258
[http://dx.doi.org/10.1100/2012/165258] [PMID: 22593663]
[63]
Gamage SA, Tepsiri N, Wilairat P, et al. Synthesis and in vitro evaluation of 9-anilino-3, 6-diaminoacridines active against a multidrug-resistant strain of the malaria parasite Plasmodium falciparum. J Med Chem 1994; 37(10): 1486-94.
[http://dx.doi.org/10.1021/jm00036a014] [PMID: 8182707]
[64]
Anderson MO, Sherrill J, Madrid PB, et al. Parallel synthesis of 9-aminoacridines and their evaluation against chloroquine-resistant Plasmodium falciparum. Bioorg Med Chem 2006; 14(2): 334-43.
[http://dx.doi.org/10.1016/j.bmc.2005.08.017] [PMID: 16216519]
[65]
Sondhi SM, Johar M, Nirupama S, Sukla R, Raghubir R, Dastidar SG. Synthesis of sulpha drug acridine derivatives and their evaluation for anti-anflammatory, analgesic and anticancer activity. Indian J Chem 2002; 41B: 2659-66.
[66]
Gamage SA, Figgitt DP, Wojcik SJ, et al. Structure-activity relationships for the antileishmanial and antitrypanosomal activities of 1′-substituted 9-anilinoacridines. J Med Chem 1997; 40(16): 2634-42.
[http://dx.doi.org/10.1021/jm970232h] [PMID: 9258370]
[67]
Di Giorgio C, Shimi K, Boyer G, Delmas F, Galy JP. Synthesis and antileishmanial activity of 6-mono-substituted and 3, 6-di-substituted acridines obtained by acylation of proflavine. Eur J Med Chem 2007; 42(10): 1277-84.
[http://dx.doi.org/10.1016/j.ejmech.2007.02.010] [PMID: 17418916]
[68]
Llama EF, Campo CD, Capo M, Anadon M. Synthesis and antinociceptive activity of 9-phenyl-oxy or 9-acyl-oxy derivatives of xanthene, thioxanthene and acridine. Eur J Med Chem 1989; 24: 391-6.
[http://dx.doi.org/10.1016/0223-5234(89)90083-4]
[69]
Recanatini M, Cavalli A, Belluti F, et al. SAR of 9-amino-1,2,3,4-tetrahydroacridine-based acetylcholinesterase inhibitors: Synthesis, enzyme inhibitory activity, QSAR, and structure-based comfa of tacrine analogues. J Med Chem 2000; 43(10): 2007-18.
[http://dx.doi.org/10.1021/jm990971t] [PMID: 10821713]
[70]
Goodell JR, Madhok AA, Hiasa H, Ferguson DM. Synthesis and evaluation of acridine- and acridone-based anti-herpes agents with topoisomerase activity. Bioorg Med Chem 2006; 14(16): 5467-80.
[http://dx.doi.org/10.1016/j.bmc.2006.04.044] [PMID: 16713270]
[71]
Hemalatha V, Sakila L, Balaji M. Molecular modelling and insilico drug docking studies on breast cancer target protein (TNRC9) using cheminformatics software and tools. Int J Novel Trends Pharm Sci 2015; 5(3): 55-63.
[72]
Rastogi K, Chang JY, Pan WY, et al. Antitumor AHMA linked to DNA minor groove binding agents: Synthesis and biological evaluation. J Med Chem 2002; 45(20): 4485-93.
[http://dx.doi.org/10.1021/jm0200714] [PMID: 12238927]
[73]
Baruah H, Wright MW, Bierbach U. Solution structural study of a DNA duplex containing the guanine-N7 adduct formed by a cytotoxic platinum-acridine hybrid agent. Biochemistry 2005; 44(16): 6059-70.
[http://dx.doi.org/10.1021/bi050021b] [PMID: 15835895]
[74]
Harrison RJ, Cuesta J, Chessari G, et al. Trisubstituted acridine derivatives as potent and selective telomerase inhibitors. J Med Chem 2003; 46(21): 4463-76.
[http://dx.doi.org/10.1021/jm0308693] [PMID: 14521409]
[75]
Kalirajan R, Sivakumar SU, Jubie S, Gowramma B, Suresh B. Synthesis and biological evaluation of some heterocyclic derivatives of chalcones. Int J Chem Sci 2009; 1(1): 27-34.
[76]
Padmaja A, Payani T, Reddy GD, Padmavathi V. Synthesis, antimicrobial and antioxidant activities of substituted pyrazoles, isoxazoles, pyrimidine and thioxopyrimidine derivatives. Eur J Med Chem 2009; 44(11): 4557-66.
[http://dx.doi.org/10.1016/j.ejmech.2009.06.024] [PMID: 19631423]
[77]
Rajanarendar E, Mohan G, Kalyan E, Siva A. The synthesis, antimicrobial and mosquito larvicidal activity of N- protected amino acid/peptide isoxazoles. Indian J Chem 2008; 47B: 781-6.
[http://dx.doi.org/10.1002/chin.200836193]
[78]
Edwin W. Kamal, Michal Z, Wanda S. Synthesis and pharmacological screening of derivatives of isoxazole [4,5-d] pyrimidine. Eur J Med Chem 2008; 43: 2498-504.
[http://dx.doi.org/10.1016/j.ejmech.2008.01.035] [PMID: 18358570]
[79]
Kalirajan R. Microwave irradiated synthesis, characterization and evaluation for their antibacterial and larvicidal activities of some novel chalcone and isoxazole substituted 9-anilino Acridines. Open J Chem 2015; 1(1): 1-7.
[80]
Sahu S, Banerjee M, Sahu D, Behera C. Synthesis, analgesic and antimicrobial activities of some novel isoxazole derivatives. Dhaka Univ J Pharm Sci 2008; 7(1): 113-8.
[81]
Osama S, Moustafa S, Raga A. Synthesis and antimicrobial activity of some new cyanopyrimidines, isoxazoles, pyrazoles and pyrimidines bearing sulfonamide moiety Phosphorus Sulfur 2003 178: 475-84.
[82]
Panda S. chowdary P, jayashree B. Synthesis, anti-inflammatory and antibacterial activity of novel indolyl-isoxazoles. Indian J Pharm Sci 2009; 71(6): 684-7.
[http://dx.doi.org/10.4103/0250-474X.59554] [PMID: 20376225]
[83]
Kalirajan R. Docking studies, Synthesis, Characterization and Evaluation of their Antioxidant and cytotoxic activities of some Novel Isoxazole substituted 9-anilinoacridine derivatives. ScientificWorldJournal 2012; 165258: 6.
[http://dx.doi.org/10.1100/2012/165258]
[84]
Kalirajan R, Vivek K, Sankar S, Jubie S. Docking studies, synthesis, characterization of some novel oxazine substituted 9-anilinoacridine derivatives and evaluation for their anti oxidant and anticancer activities as topo isomerase II inhibitors. Eur J Med Chem 2012; 56: 217-24.
[http://dx.doi.org/10.1016/j.ejmech.2012.08.025] [PMID: 22982526]
[85]
Kalirajan R, Leela R, Jubie S, Gowramma B, Gomathy S, Sankar S. Microwave assisted synthesis of some novel pyrazole substituted benzimidazoles and evaluation of their biological activities. Indian J Chem 2011; 50B: 1794-801.
[86]
Kalirajan R, Sankar S, Jubie S, Gowramma B. Molecular Docking studies and in-silico ADMET Screening of Some novel Oxazine substituted 9-Anilinoacridines as Topoisomerase II Inhibitors. Indian J Pharm Educ Res 2017; 51(1): 110-5.
[http://dx.doi.org/10.5530/ijper.51.1.15]
[87]
Kalirajan R, Gowramma B, Jubie S, Sankar S. Molecular docking Studies and in silico ADMET Screening of some novel heterocyclic substituted 9-anilinoacridines as topoisomerase ii inhibitors. JSM Chem 2017; 5(1): 1039-44.
[88]
Kalirajan R, Gaurav K, Pandiselvi A, Gowramma B, Sankar S. Novel thiazine substituted 9-anilinoacridines: Synthesis, antitumour activity and structure-activity relationships. Anticancer Agents Med Chem 2019; 19(11): 1350-8.
[http://dx.doi.org/10.2174/1871520619666190408134224] [PMID: 30961512]
[89]
Gowramma B, Praveen TK, Gomathy S, Kalirajan R, Babu B, Nagappan K. Synthesis of 2-(Bis (2-Chloroethyl) Amino)-N- (5-substituedphenyl)-1,3,4- Thiadiazol- 2-Yl) Aceto hydrazide and Evaluation of Anticancer activity. Curr Bioact Compd 2018; 14(3): 309-16.
[90]
Kalirajan R, Vivek K, Sankar S. Synthesis, characterization and evaluation for antitumour activity of some novel oxazine substituted 9-anilinoacridines and their 3D-QSAR studies. Indian J Pharm Sci 2018; 80(5): 921-9.
[http://dx.doi.org/10.4172/pharmaceutical-sciences.1000439]
[91]
kalirajan R, Rathire L, Jubie S, et al. Microwave assisted synthesis and biological evaluation of pyrazole derivatives of benzimidazoles. Indian J Pharm Educ Res 2010; 44(4): 358-62.
[92]
Kalirajan R. Chitra, Jubie S, Gowramma B. Synthesis and biological evaluation of Mannich bases of 2-substituted Benzimidazoles. Asian J Chem 2009; 21(7): 5207-11.
[93]
Jubie S, Gayathri R, Srividya AR, et al. Synthesis and characterization of some novel quinoxaline-2, 3-dione derivatives: A preliminary investigation on their activity against a human epithelial carcinoma cell line. Lett Drug Des Discov 2011; 8: 317-20.
[http://dx.doi.org/10.2174/157018011794839385]
[94]
Jubie S, Nilesh RP, Dhanabal P, Kalirajan R, Muruganantham N, Antony SA. Synthesis, antidepressant and antimicrobial activities of some novel stearicacid analogues. Eur J Med Chem 2012; 54: 931-5.
[http://dx.doi.org/10.1016/j.ejmech.2012.06.025] [PMID: 22770606]
[95]
Jubie S, Dhanabal P, Afzal AM, Muruganantham N, Kalirajan R, Elango K. Synthesis and characterization of some novel fatty acid analogues: A preliminary investigation on their activity against human lung carcinoma cell line. Lipids Health Dis 2013; 12: 45-52.
[http://dx.doi.org/10.1186/1476-511X-12-45] [PMID: 23537396]
[96]
Kalirajan R, Pandiselvi A, Gowramma B. In-silico drug design by docking studies, admet screening, mm-gbsa binding free energy of some chalcone substituted 9-anilinoacridines as her2 inhibitors targeting breast cancer. Int J Comp Theo Chem 2019; 7(1): 6-13.
[97]
Halperin I, Ma B, Wolfson H, Nussinov R. Principles of docking: An overview of search algorithms and a guide to scoring functions. Proteins 2002; 47(4): 409-43.
[http://dx.doi.org/10.1002/prot.10115] [PMID: 12001221]
[98]
Naga ST, Mohammed AA. Pharmacophore modelling, 3D-QSAR and docking study of 2-phenylpyrimidine analogues as selective PDE4B inhibitors. J Theor Biol 2016; 394: 117-26.
[http://dx.doi.org/10.1016/j.jtbi.2016.01.007] [PMID: 26804643]
[99]
Lengaur T, Rarey M. Computational method for bio molecular docking. Curr Opin Struct Biol 1996; 6(3): 402-6.
[http://dx.doi.org/10.1016/S0959-440X(96)80061-3]
[100]
Kalirajan R, Pandiselvi A, Sankar S, Gowramma B. Molecular docking studies and in silico ADMET screening of some novel chalcone substituted 9-anilinoacridines as topoisomerase ii inhibitors. SF J Pharm Anal Chem 2018; 1(1): 1004-9.
[101]
Reetu VK. Computer aided design of selective calcium channel blockers: Using pharmacophore - Based and docking simulations. Indian J Pharm Sci Res 2012; 3(3): 805-10.
[102]
Furrer D. Claudie Paquet, Simon Jacob, Caroline Diorio. Cancer prognosis 2018.
[103]
Pal S. An in silico drug designing approach to target the BRCA1 protein involved in breast cancer. Helix 2016; 16(1): 761-5.
[104]
Mehta S, Seema R. Pathak. In silico drug design and molecular docking studies of novel coumarin derivatives as anticancer agents. Asian J Pharm Clin Res 2017; 10(4): 335-40.
[105]
Sahu M, Amit GN. In silico design, synthesis and pharmacological screening of some Quinazolinone metal complexes as dihydrofolate reductase inhibitors for anticancer activity: Part-II. Int J Pharm Pharm Sci 2014; 6(5): 509-14.
[106]
Alejandro SP, Natália MDSC. Fragment-based in silico modeling of multi-target inhibitors against breast cancer-related proteins. Mol Divers 2017; 21(3): 511-23.
[http://dx.doi.org/10.1007/s11030-017-9731-1]

© 2024 Bentham Science Publishers | Privacy Policy