Multidimensional QSAR Modeling of Amprenavir Derivatives as HIV-Protease Inhibitors
Sonal Dubey, G. Gowtham
.
DOI: 10.4236/ojmc.2011.11001   PDF    HTML     5,104 Downloads   13,687 Views   Citations

Abstract

A computational study has been performed on a series of 55 compounds having (S)-N-(3-(N-(cyclopean- tylmethyl)substituted-phenylsulfonamido)-2-hydroxypropyl)acetamide backbone as HIV-1 protease inhibitors. Various combinations of these specific inhibitors fragments were formed by breaking them at central alicyclic single bonds, while retaining the core. Standard Topomer 3D models were automatically constructed for each fragment, and a set of steric and electrostatic fields was generated for each set of topomers. The models generated showed r2 of 0.811 and crossvalidated r2 (q2) of 0.608. The other method used were Quasar and Raptor based on receptor-modelling concept (6D-QSAR) and this explicitly allows for the simulation of the induced fit, that yielded r2 of 0.574, cross-validated r2 (q2) of 0.504 and predictive r2 (q2) of 0.895 averaged over 200 models. This study has suggested the various type of substituent that can be attached to the core. The information obtained from these 3-D contour maps can be used for the design of amprenavir analogs possessing better protease inhibitory activity.

Share and Cite:

S. Dubey and G. Gowtham, "Multidimensional QSAR Modeling of Amprenavir Derivatives as HIV-Protease Inhibitors," Open Journal of Medicinal Chemistry, Vol. 1 No. 1, 2011, pp. 1-15. doi: 10.4236/ojmc.2011.11001.

Conflicts of Interest

The authors declare no conflicts of interest.

References

[1] H. Mitsuya, R. Yarchoan and S. Broder, “Molecular Targets for AIDS Therapy”, Science, Vol. 249, 1990, pp. 1533-44.
[2] R. Yarchoan, H. Mitsuya and S. Broder, “Challenges in the Therapy of HIV Infection”, Trends in Pharmacological Science, Vol. 14, 1993, pp. 196-202.
[3] C.U. Hellen, H.G. Krausslich and E. Wimmer, “Proteolytic Processing of Polyproteins in the Replication of RNA Viruses”, Biochemistry, Vol. 28, 1989, pp.. 9881-90.
[4] J. Vondrasek and A. Wlodawer, “A Database of the Structures of Human Immunodeficiency Virus Protease”, Proteins, Vol. 49, 2002, pp. 429-31.
[5] M.L.West and D.P Fairlie, “Targeting HIV-1 Protease: A Test of Drug-Design Methodologies”, Trends in Pharmacological Science , Vol. 16, 1995, pp. 67-75.
[6] T.D. Meek, “Inhibitors of HIV-1 Protease”, Enzyme Inhibition, Vol.6, 1992, pp. 65-98.
[7] C. Merry, M.G Barry, F. Mulcahy, K.L Halifax and D.J Back, “Saquinavir Pharmacokinetics Alone and In Combination With Nelfinavir in HIV-Infected Patients”, AIDS, Vol. 11, 1997, pp. F117-20.
[8] E.L. Ellsworth, J. Domagala, J.V. Prasad, S. Hagen, D. Ferguson, T. Holler, D. Hupe, N. Graham, C. Nouhan, P.J. Tummino, G. Zeikus and E.A. Lunney, “4-Hydroxy- 5,6-dihydro-2H-pyran-2-ones.3. Bicyclic and Hetero-Ar- omatic Ring Systems as 3-Position Scaffolds to Bind to S1' and S2' of the HIV-1 Protease Enzyme ”, Bioorganic Medicinal Chemistry Leters., Vol 9, 1999, pp. 2019-24.
[9] A.K. Patick, K.E. Potts, “Protease inhibitors as antiviral agents”, Clinical Microbiology Reviews., Vol. 11, 1998, pp. 614-27.
[10] J.R. Huff, “HIV protease: a novel chemotherapeutic target for AIDS”, Journal of Medicinal Chemistry, Vol.34, 1991, pp. 2305-12.
[11] N.E. Kohl, E.A. Emini, W.A. Schleif, L.J. Davis, J.C. Heimbach, R.A. Dixon, E. M. Scolnick and I. Sigal, “Active Human Immunodeficiency Virus Protease is Required for Viral”, Proceedings of the National Academy of Science USA, Vol. 85, 1988, pp. 4686-90.
[12] E.T. Brower, U.M. Bacha, Y. Kawasaki and E. Freire, “Inhibition of HIV-2 protease by HIV-1 Protease Inhibitors in Clinical Use”, Chemical Biology and Drug Design, Vol. 71, 2008, pp. 298-305.
[13] G. Bold, H.G. Capraro, R. Cozens, T. Klimkait, J. Lazdins, J. Mestan, B. Poncioni, J. Rosel, V. Stover, M. Tintelnot-Blomley, F. Acemoglu, W. Beck, B. Boss, M. Eschbachm, T. Hurlimann, E. Masso, S. Roussel, K. UcciStoll, D. Wyss and M. Lang, “New Aza-Dipeptide Analogues as Potent and Orally Absorbed HIV-1 Protease Inhibitors: Candidates for Clinical Development”, Journal of Medicinal Chemistry, Vol. 41, 1998, pp. 3387-401.
[14] D.C. Richard, “Topomer CoMFA: A Design Methodology for Rapid Lead Optimization”, Journal of Medicinal Chemistry, Vol. 46, 2003,pp. 374-388.
[15] F.G. Salituro, C.T. Baker, J.J. Court, D.D. Deininger, E.E. Kim and B. Li, “Design and Synthesis of Novel Conformationally Restricted HIV Protease Inhibitors”, Bioorganic Medicinal Chemistry Letters, Vol. 8, 1998, pp. 3637-42.
[16] C.T. Baker, F.G. Salituro, J.J. Court, D.D. Deininger, E.E. Kim, B. Li, P.M. Novak, B.G. Rao, S. Pazhanisamy, W.C. Schairer and R.D. Tung, “Design, Synthesis and Conformational Analysis of a Novel Series of HIV Protease Inhibitor” , Bioorganic Medicinal Chemistry Letters, Vol. 8, 1998, pp. 3631-36.
[17] A.K. Ghosh, J.F. Kincaid, W. Cho, D.E. Walters, K. Krishnan, K.A. Hussain, Y. Koo, H. Cho, C. Rudall, L. Holland and J. Buthod, “Potent HIV Protease Inhibitors Incorporating High-Affinity P2-Ligands and (R)-(Hyd- roxyethylamino)Sulfonamide Isoesters”, Bioorganic Medicinal Chemistry Letters, Vol. 8, 1998, pp. 687-90.
[18] A.K. Ghosh, J.F. Kincaid, W. Cho, D.E. Walters, H. Cho, Y. Koo, J. Trevino, L. Holland and J. Buthod, “Structure Based Design: Novel Spirocyclic Ethers as Nonpeptidal P2-Ligands for HIV Protease Inhibitors”, Bioorganic Medicinal Chemistry Letters, Vol. 8, 1998, pp. 979-82.
[19] A.K. Ghosh, S. Leshchenko-Yashchuk, D.D. Anderson, A. Baldridge, M. Noetzel, H.B. Miller, Y. Tie, YF. Wang, Y. Koh, I.T. Weber and H. Mitsuya, “Design of HIV-1 Protease Inhibitors with Pyrrolidinones and Oxazolidinones as Novel P1'-Ligands to Enhance Backbone- Bind- ing Interactions with Protease: Synthesis, Biological Eva- luation and Protien-Ligand X-Ray Studies”, Journal of Medicinal Chemistry, Vol. 52, 2009, pp. 3902-914.
[20] R.D. Cramer, “Topomer CoMFA: A Design Methodology for Rapid Lead Optimization”, Journal of Medicinal Chemistry, Vol. 46, 2003, PP. 374-388.
[21] R.D. Cramer, R.J. Jilek and K.M. Andrews, “Topomer Similarity Searching of Conventional Databases”, Journal of Molecular Graphics and Modeling, Vol. 20, 2002, pp. 447-462.
[22] A. Vedani and M. Dobler, “5D-QSAR: the key for simulating induced fit?”, Journal of Medicinal Chemistry, Vol. 45, 2002, pp. 2139-49.
[23] A. Vedani, M. Dobler and M.A. Lill, “Combining Protein Modeling and 6D-QSAR. Simulating the Binding of Structurally Diverse Ligands to the Estrogen Receptor”, Journal of Medicinal Chemistry, Vol. 48, 2005, pp. 3700- 3703.
[24] A. Vedani, M. Dobler, H. Dollinger, M. Hasselbach Kai, F. Birke and M.A. Lill, “Novel Ligands for the Chemokine Receptor-3 (CCR3): A Receptor-Modelling Study Based on 5D-QSAR”, Journal of Medicinal Chemistry, Vol. 48, 2005, pp. 1515-1527.
[25] A. Vedani, “QSAR:Prediction Beyond the Fourth Dimension”, Drug Discovery and Development, 2005, Available online: http://www.dddmag.com/qsar-prediction-beyond-the-fourth.aspx.
[26] Quasar.pdf- (Manual)
[27] D. Rogers and A.J. Hopfinger, “Genetic function approximation to generate a family of QSAR equations using genetic algorithms”, Journal of Chemical Information and Modeling, VOL. 34, 1994, pp. 854-66.
[28] A. Vedani, M. Dobler and M.A. Lill, “In silico prediction of harmful effects triggered by drugs and chemicals”, Toxicology and Applied Pharmacology, VOL. 207, 2005, pp. S398-S407.
[29] M.A. Lill, A. Vedani and M. Dobler, “Raptor combining dual shell representation, induced-fit simulation and hydrophobicity scoring in receptor-modeling: application towards the simulation of structutrally diverse ligand sets”, Journal of Medicinal Chemistry, Vol. 47, 2004, pp. 6174-6186.

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.