SYNTHESIS OF NOVEL CYCLOHEXANONE DERIVATIVES AS BCR-ABL T1351 INHIBITORS

Authors

  • Sulaiman Ali Muhammad Department of Chemistry, Karpagam Academy of Higher Education, Coimbatore 641021 Tamilnadu, India
  • Subash Suganya Department of Chemistry, Karpagam Academy of Higher Education, Coimbatore 641021 Tamilnadu, India
  • Subban Ravi Department of Chemistry, Karpagam Academy of Higher Education, Coimbatore 641021 Tamilnadu, India
  • Senniappan Venkatachalapathi Department of Chemistry, Karpagam Academy of Higher Education, Coimbatore 641021 Tamilnadu, India

Keywords:

Synthesis, Phenothiazine, Cyclohexanone derivatives, Chronic myelogenous leukemia, Molecular docking, Anti-proliferative activity

Abstract

Objective: Several 3(rd) generation inhibitors are being developed for the treatment of patients with Chronic myelogenous leukemia (CML). The present work mainly aims to discover novel small molecular inhibitors against important molecular target T3151 ABL mutant involved in leukemia.

Methods: Docking study was carried out and the binding affinity of the proteins with the phenothiazine compounds 3a-h and 7a-c was measured. The docking scores of the N-acylated compounds 7a-c are higher than 3a-h. The drug likeliness of these compounds was tested by the Lipinski's rule of five. The phenothiazine compounds with good docking scores and 7a-c were synthesized and screened by in-vitro methods for inducing antiproliferative effect by trypan blue and MTT assay and induction of apoptosis in K562 cells.

Results: All the N-acylated compounds and, in particular, 7c with a chloro substituent in the para position of the phenyl ring appeared to be most potent molecule with an IC50 value of 32.44 and 24.01(µg/ml) by trypan blue and MTT assay respectively. Further, a dose-dependent increase in LDH release was observed, confirming the antiproliferative potential of the compounds.

Conclusion: The compounds 7a-c was tested for antiproliferative effect against K562 cell lines by MTT assay LDH assay and Trypan blue assay. All the compounds 7a-h behaves as 3(rd) generation inhibitors for the treatment of patients with Chronic myelogenous leukemia (CML). These can act as a template for the further development and optimization studies.

 

Downloads

Download data is not yet available.

Author Biography

Sulaiman Ali Muhammad, Department of Chemistry, Karpagam Academy of Higher Education, Coimbatore 641021 Tamilnadu, India

Research Scholar, Department of Chemistry

Karpagam Academy of Higher education, Coimbatore-21

References

Feig M, Onufriev A, Lee MS, Im W, Case DA, Brooks CL. Performance comparison of generalized born and Poisson methods in the calculation of electrostatic solvation energies for protein structures. J Comb Chem 2004;25:265–84.

Friesner RA, Banks JL, Murphy RB, Halgren TA, Klicic JJ, Mainz DT, et al. Glide: a new approach for rapid, accurate docking and scoring. Method and assessment of docking accuracy. J Med Chem 2004;47:1739–49.

Goldman BB, Wipke WT. QSD quadratic shape descriptors. 2. Molecular docking using quadratic shape descriptors (QSDock). Proteins 2000;38:79–94.

Jorgensen WL. Rusting of the lock and key model for protein-ligand binding. Science 1991;254:954.

Kahraman A, Morris RJ, Laskowski RA, Thornton JM. Shape variation in protein binding pockets and their ligands. J Mol Biol 2007;368:283–301.

Kitchen DB, Decornez H, Furr JR, Bajorath J. Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discovery 2004;3:935–49.

Klebe G, Mietzner T. A fast and efficient method to generate biologically relevant conformations". J Comput Aided Mol Des 1994;8:583–606.

Lengauer T, Rarey M. Computational methods for biomolecular docking. Curr Opin Struct Biol 1996;6:402–6.

Longenecker KL, Stamper GF, Hajduk PJ, Fry EH, Jakob CG, Harlan JE, et al. Structure of MurF from streptococcus pneumoniae co-crystallized with a small molecule inhibitor exhibits interdomain closure. Protein Sci 2005;14:3039–47.

Meng EC, Shoichet BK, Kuntz ID. Automated docking with grid-based energy evaluation. J Comp Chem 2004;13:505–24.

Modungo M, Casale E, Soncini C, Rosettani P, Colombo R, Lupi R, et al. Crystal Structure of the T315I Abl Mutant in Complex with the Aurora Kinases Inhibitor PHA-739358. Cancer Res 2007;67:7987.

Morris GM, Goodsell DS, Halliday RS, Huey R, Hart WE, Belew RK, et al. Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J Comp Chem 1998;19:1639–62.

Shoichet BK, Kuntz ID, Bodian DL. Molecular docking using shape descriptors. J Comp Chem 2004;13:380–97.

Sousa SF, Ribeiro AJ, Coimbra JTS, Neves RPP, Martins SA, Moorthy HNS, et al. Protein-ligand docking in the new millennium-A retrospective of 10 y in the field. Curr Med Chem 2013;20:2296–14.

Wang Q, Pang YP. Romesberg, Floyd. ed. Preference of small molecules for local minimum conformations when binding to proteins. PLoS One 2007;2:e820.

Wei BQ, Weaver LH, Ferrari AM, Matthews BW, Shoichet BK. Testing a flexible-receptor docking algorithm in a model binding site. J Mol Biol 2004;337:1161–82.

Zengming Z, Yu Li, Biaoyang Lin. Michael Schroeder and Bingding Huang, Identification of cavities on protein surface using multiple computational approaches for drug binding site prediction. Bioinform 2011;27:2083-8.

An X, Tiwari A, Sun Y, Ding P, Ashby Jr C, Chen Z. BCR-ABL tyrosine kinase inhibitors in the treatment of Philadelphia chromosome-positive chronic myeloid leukemia: a review. Leuk Res 2010;34:1255–68.

Bixby D, Talpaz M. Mechanisms of resistance to tyrosine kinase inhibitors in chronic myeloid leukemia and recent therapeutic strategies to overcome resistance. Hematology 2009;1:461-76.

Manley PW, Cowan-Jacob SW, Buchdunger E, Fabbro D, Fendrich G, Furet P, et al. Imatinib: a selective tyrosine kinase inhibitor. Eur J Cancer 2002:S19-27.

Shawver LK, Slamon D, Ullrich A. Smart drugs: tyrosine kinase inhibitors in cancer therapy. Cancer Cell 2002;1:117-23.

Druker BJ, Lydon NB. Lessons learned from the development of an ABL tyrosine kinase inhibitor for chronic myelogenous leukemia. J Clin Invest 2000;105:3–7.

Buchanan SG. Protein structure: discovering selective protein kinase inhibitors. Targets 2003;2:101-8.

Eck M, Manley P. The interplay of structural information and functional studies in kinase drug design: insights from BCR-ABL. Curr Opin Cell Biol 2009;21:288–95.

Mandal S, Moudgil M, Mandal S. Rational drug design. Eur J Pharm 2009;625:90–100.

Asaki T, Sugiyama Y, Hamamoto T, Higashioka M, Umehara M, Naito H, et al. Design and synthesis of 3-substituted benzamide derivatives as BCR-ABL kinase inhibitors. Bioorgn Med Chem Lett 2006;16:1421–5.

Manley P, Cowan-Jacob S, Mestan J. Advances in the structural biology, design and clinical development of BCR-ABL kinase inhibitors for the treatment of chronic myeloid leukaemia. Biochim Biophys Acta 1754;1–2:3–13.

Manley P, Stiefl N, Cowan-Jacob S, Kaufman S, Mestan J, Wartmann M, et al. "Structural resemblances and comparisons of the relative pharmacological properties of imatinib and nilotinib". Bioorg Med Chem 2010;18:6977–86.

Stein B, Smith BD. Treatment options for patients with chronic myeloid leukemia who are resistant to or unable to tolerate imatinib. Clin Ther 2010;32:804-20.

Gorre M, Mohammed M, Ellwood K, Hsu N, Paquette RP, Rao PN, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science 2001;293:876-80.

Thomas J, Wang L, Clark R, Pirmohamed M. Active transport of imatinib into and out of cells: implications for drug resistance. Blood 2004;104:3739–45.

Jabbour E, Cortes J, Kantarjian H. Nilotinib for the treatment of chronic myeloid leukemia: an evidence-based review. Core Evidence 2009;4:207-13.

Olivieri A, Manzione L. "Dasatinib: a new step in molecular target therapy". Annal oncol 2007;18 Suppl 6:42–6.

Breccia M, Alimena G. "Nilotinib: a second-generation tyrosine kinase inhibitor for chronic myeloid leukemia". Leuk Res 2010;34:129–34.

http://www.rscb.org/pdb for protein structure data base. [Last accessed on 11 oct 2015].

Meta Pocket 2.0 Finder program. Available from: http://metpocket.eml.org. [Last accessed on 11 oct 2015].

Saranya AV, Ravi S. Synthesis of 5-phenyl-3-(10H-phenothiazinyl)-Δ2-cyclohexen-1-ones by conventional and microwave-assisted methods and their antifungal activity. Res Chem Int 2014;40:3085-93.

Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 1983;65:55-63.

Shahabuddin MS, Nambiar M, Choudhary B, Advirao GM, Raghavan SC. A novel DNA intercalator, butylamino-pyrimido[4',5':4,5]selenolo(2,3-b)quinoline, induces cell cycle arrest and apoptosis in leukemic cells. Invest New Drugs 2009;28:35-48.

Korzeniewski C, Callewaert DM. An enzyme-release assay for natural cytotoxicity. J Immunol Methods 1983;64:313–20.

Published

01-12-2015

How to Cite

Muhammad, S. A., S. Suganya, S. Ravi, and S. Venkatachalapathi. “SYNTHESIS OF NOVEL CYCLOHEXANONE DERIVATIVES AS BCR-ABL T1351 INHIBITORS”. International Journal of Pharmacy and Pharmaceutical Sciences, vol. 7, no. 12, Dec. 2015, pp. 195-9, https://journals.innovareacademics.in/index.php/ijpps/article/view/8995.

Issue

Section

Original Article(s)