Generic placeholder image

Current Drug Discovery Technologies

Editor-in-Chief

ISSN (Print): 1570-1638
ISSN (Online): 1875-6220

General Research Article

Combined Ligand-Based and Structure-Based Virtual Screening Approach for Identification of New Dipeptidyl Peptidase 4 Inhibitors

Author(s): Jagatkumar Upadhyay*, Anuradha Gajjar and Bhanubhai N. Suhagia

Volume 16, Issue 4, 2019

Page: [426 - 436] Pages: 11

DOI: 10.2174/1570163815666180926111558

Price: $65

Abstract

Background: Dipeptidyl Peptidase 4 (DPP 4) enzyme cleaves an incretin-based glucoregulatory hormone Glucagon Like Peptide -1 from N-terminal where penultimate amino acid is either alanine or proline. Several DPP 4 inhibitors, “gliptins”, are approved for the management of Type 2 Diabetes or are under clinical trial. In the present study, combined pharmacophore and docking-based virtual screening protocol were used for the identification of new hits from the Specs Database, which would inhibit DPP 4.

Methods: The entire computational studies were performed using the Discovery Studio v. 4.1 software package, Pipeline Pilot v. 9.2 (Accelrys Inc.) and FRED v. 2.2.5 (OpenEye Scientific Software). Common feature pharmacophore model was generated from known DPP 4 inhibitors and validated by Receiver Operating curve analysis and GH-scoring method. Database search of Specs commercial database was performed using validated pharmacophore. Hits obtained from pharmacophore search were further docked into the binding site of DPP 4. Based on the analysis of docked poses of hits, 10 compounds were selected for in- vitro DPP 4 enzyme inhibition assay.

Results: Based on docking studies, virtual hits were predicted to form interaction with essential amino acid residues of DPP 4 and have an almost similar binding orientation as that of the reference molecule. Three compounds having Specs database ID- AN-465/42837213, AP-064/42049348 and AN- 465/43369427 were found to inhibit DPP 4 enzyme moderately.

Conclusion: The present study demonstrates a successful utilization of in-silico tools in the identification of new DPP 4 inhibitor, which can serve as a starting point for the development of novel DPP 4 inhibitors.

Keywords: DPP 4, Gliptins, virtual screening, specs database, pharmacophore, molecular docking.

Graphical Abstract
[1]
World Health Organization. Diabetes Factsheet. http://www.who.int/mediacentre/factsheets/fs312/en/ (Accessed March 13, 2018)
[2]
Penalver J, Timon I, Collantes C, Canizo-Gomez F. Update on the treatment of type 2 diabetes mellitus. World J Diabetes 2016; 7: 354-95.
[3]
Cahn A, Cernea S, Raz I. An update on DPP-4 inhibitors in the management of type 2 diabetes. Expert Opin Emerg Drugs 2016; 121: 1-12.
[4]
Jose T, Inzucchi S. Cardiovascular effects of the DPP-4 inhibitors. Diab Vasc Dis Res 2012; 9: 109-16.
[5]
Stonehouse A, Darsow T, Maggs D. Incretin-based therapies. J Diabetes 2012; 4: 55-67.
[6]
Daniel J, Drucker D, Nauck M. The incretin system: Glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006; 368: 1696-705.
[7]
Cox M, Chu H, Kuethe J, et al. The discovery of novel 5, 6, 5- and 5, 5, 6-tricyclic pyrrolidines as potent and selective DPP-4 inhibitors. Bioorg Med Chem Lett 2016; 26: 2622-6.
[8]
Kim D, Wang L, Beconi M, et al. (2R)-4-Oxo-4-[3-(Trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]-1-(2,4,5 trifluorophenyl)butan-2-amine: A potent, orally active dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. J Med Chem 2005; 48: 141-51.
[9]
Villhauer E, Brinkman J, Naderi G, et al. 1-[[(3-Hydroxy-1-adamantyl)amino]acetyl]-2-cyano-(S)-pyrrolidine: A potent, selective, and orally bioavailable dipeptidyl peptidase IV inhibitor with antihyperglycemic properties. J Med Chem 2003; 46: 2774-89.
[10]
Augeri D, Robl J, David A, et al. Discovery and Preclinical Profile of Saxagliptin (BMS-477118): A highly potent, long-acting, orally active dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. J Med Chem 2005; 48: 5025-37.
[11]
Feng J, Zhang Z, Wallace M, et al. Discovery of alogliptin: a potent, selective, bioavailable, and efficacious inhibitor of dipeptidyl peptidase IV. J Med Chem 2007; 50: 2297-300.
[12]
Eckhardt M, Langkopf E, Mark M, et al. 8-(3-(R)-Aminopiperidin-1-yl)-7-but-2-ynyl-3-methyl-1-(4-methyl-quinazolin-2-ylmethyl)-3,7-dihydropurine-2,6-dione (BI 1356), a highly potent, selective, long-acting, and orally bioavailable DPP-4 inhibitor for the treatment of type 2 diabetes. J Med Chem 2007; 50: 6450-3.
[13]
Yoshida T, Akahoshi F, Sakashita H, et al. Discovery and preclinical profile of teneligliptin (3-[(2s,4s)-4-[4-(3-methyl-1-phenyl-1h-pyrazol-5-yl)piperazin-1-yl]pyrrolidin-2-ylcarbonyl]thiazolidine): A highly potent, selective, long-lasting and orally active dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. Bioorg Med Chem 2012; 20: 5705-19.
[14]
Kato N, Oka M, Murase T, et al. Discovery and pharmacological characterization of N-[2-(2-[(2S)-2-cyanopyrrolidin-1-yl]-2-oxoethylamino)-2-methylpropyl]-2-methylpyrazolo[1,5-a]pyrimidine-6-carboxamide hydrochloride (anagliptin hydrochloride salt) as a potent and selective DPP-IV inhibitor. Bioorg Med Chem 2011; 19: 7221-7.
[15]
Engel M, Hoffmann T, Wagner L, et al. The crystal structure of dipeptidyl peptidase IV (CD26) reveals its functional regulation and enzymatic mechanism. Proc Natl Acad Sci 2003; 100: 5063-8.
[16]
Gupta S, Patil V. Specificity of binding with matrix metalloproteinases. In: Gupta S, Ed. Matrix metalloproteinases Inhibitors. New York: Springer International Publishing AG 2012; Vol. 103: pp. 35-56.
[17]
Nabeno M, Akahoshi F, Kishida H, et al. A comparative study of the binding modes of recently launched dipeptidyl peptidase IV inhibitors in the active site. Biochem Biophys Res Commun 2013; 434: 191-6.
[18]
Jeanneret L. Dipeptidyl peptidase IV and its inhibitors: Therapeutics for type 2 diabetes and what else? J Med Chem 2014; 57: 2197-212.
[19]
Arulmozhiraja S, Matsuo N, Ishitsubo E, Okazaki S, Shimano H, Tokiwa H. Comparative binding analysis of dipeptidyl peptidase IV (DPP-IV) with antidiabetic drugs – an ab initio fragment molecular orbital study. PLoS One 2016; 11: 1-15.
[20]
Nojima H, Kanou K, Terashi G, et al. Comprehensive analysis of the co-structures of dipeptidyl peptidase IV and its inhibitor. BMC Struct Biol 2016; 16: 11-24.
[21]
Kushwaha R, Haq W, Katti S. Sixteen-years of clinically relevant dipeptidyl peptidase-iv (DPP-IV) inhibitors for treatment of type-2 diabetes: a perspective. Curr Med Chem 2014; 21: 1-33.
[22]
Tseng C. Sitagliptin and pancreatic cancer risk in patients with type 2 diabetes. Eur J Clin Invest 2016; 46: 70-9.
[23]
Swann S, Brown P, Muchmore S, et al. A unified, probabilistic framework for structure- and ligand-based virtual screening. J Med Chem 2011; 54: 1223-32.
[24]
Lionta E, Spyrou G, Vassilatis D, Cournia Z. Structure-based virtual screening for drug discovery: principles, applications and recent advances. Curr Top Med Chem 2014; 14: 1923-38.
[25]
Noha S, Fischer K, Koeberle A, Garscha U, Werz O, Schuster D. Discovery of novel, non-acidic mPGES-1 inhibitors by virtual screening with a multistep protocol. Bioorg Med Chem 2015; 23: 4839-45.
[26]
Discovery Studio Modelling Environment, Version 4.1, Accelrys Software: San Diego, CA, 2005-2014.
[27]
PipelinePilot, Version 9.2, SciTegic, San Diego, CA, 2001-2014.
[28]
FRED, V 2.2.5, OpenEye Scientific, Santa Fe, 1997-2018.
[29]
RCSB Protein Data Bank. http://www.rcsb.org/pdb (Accessed March 1, 2018).
[30]
Edmondson S, Mastracchio A, Mathvink R, et al. (2S, 3S)-3-Amino-4-(3,3-difluoropyrrolidin-1-yl)-N,N-dimethyl-4-oxo-2-(4-[1,2,4]triazolo[1,5-a]-pyridin-6-ylphenyl)butanamide: a selective r-amino amide dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes. J Med Chem 2006; 49: 3614-27.
[31]
Biftu T, Roy R, Chen P, et al. Omarigliptin (MK-3102): A novel long-acting DPP-4 inhibitor for once-weekly treatment of type 2 diabetes. J Med Chem 2014; 57: 3205-12.
[32]
Martis E, Chandarana R, Shaikh M, et al. Quantifying ligand–receptor interactions for gorge-spanning acetylcholinesterase inhibitors for the treatment of Alzheimer’s disease. J Biomol Struct Dyn 2015; 33: 1107-25.
[33]
Guner O. History and evolution of the pharmacophore concept in computer-aided drug design. Curr Top Med Chem 2002; 2: 1321-32.
[34]
Guner O, Bowen J. Pharmacophore modeling for ADME. Curr Top Med Chem 2013; 13: 1-17.
[35]
Braga R, Andrade C. Assessing the performance of 3d pharmacophore models in virtual screening: How good are they? Curr Top Med Chem 2013; 13: 1127-38.
[36]
Yao T, Xie J, Liu X, et al. Integration of pharmacophore mapping and molecular docking in sequential virtual screening: towards the discovery of novel JAK2 inhibitors. RSC Advances 2017; 7: 10353-60.
[37]
Patel B, Ghate M. Computational studies on structurally diverse dipeptidyl peptidase IV inhibitors: An approach for new antidiabetic drug development. Med Chem Res 2013; 22: 4505-21.
[38]
The Binding Database. University of California, San Diego, US. https://www.bindingdb.org (Accessed December 10, 2017).
[39]
SPECS Database. https://www.specs.net (Accessed 13 December, 2017).
[40]
Onnis V, Kinsella G, Carta G, et al. Virtual screening for the identification of novel nonsteroidal glucocorticoid modulators. J Med Chem 2010; 53: 3065-74.
[41]
Almasri I, Taha M, Mohammad M. New leads for DPP IV inhibition: Structure-based pharmacophore mapping and virtual screening study. Arch Pharm Res 2013; 36: 1326-37.
[42]
McCann M. FRED pose prediction and virtual screening accuracy. J Chem Inf Model 2011; 51: 578-96.
[43]
Elokely K, Doerksen R. Docking challenge: Protein sampling and molecular docking performance. J Chem Inf Model 2013; 53: 1934-45.
[44]
Bickerton G, Paolini G, Besnard J, Muresan S, Andrew L, Hopkins A. Quantifying the chemical beauty of drugs. Nat Chem 2012; 4: 90-8.

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