Advancements in Drug Design Technology and Its Impact on COVID-19 Treatment

Year : 2024 | Volume :01 | Issue : 01 | Page : –
By

Satish kumar Yadav

  1. Associate professor Dept. of Pharmacy, Prasad Institute of Technology, Jaunpur Uttar Pradesh India

Abstract

The deadly coronavirus disease 19 (COVID-19) pandemic has recently spread, raising concerns about global health. The search for novel therapeutic compounds is made more necessary by the persistent problem of the absence of licensed medications or vaccinations. By saving money and time, computer-aided drug design has sped up the process of finding and developing new drugs. The structured-based and ligand-based drug discovery subcategories of computer-aided drug design (CADD) are the main topics of this review study. In ligand-based drug design, pharmacophoremodeling, quantitative structure-activity relationships (QSARs), and artificial intelligence (AI) are commonly employed molecular modeling techniques, whereas structure-based drug design often involves molecular docking and molecular dynamic simulation. We have touched on the importance of computer-aided drug design in relation to COVID-19 and how scientists are still depending on these computational methods to quickly identify molecules that show promise as potential drugs against different targets connected to the pathophysiology of SARS-CoV-2, or severe acute respiratory syndrome coronavirus. Cross-species viruses known as coronaviruses (CoVs) can quickly move from their original host species into other ones, whereupon they can cause epidemic diseases. This article provides a comprehensive examination of computational methodologies and their utilization in the realm of drug development. Chemogenomics and drug discovery (DR) are highlighted as novel and developing system-based fields that are focused on modeling protein networks versus a library of chemicals in CoV infections. Furthermore, a number of recent successes based on chemogenomics,and molecular docking are given, thoroughly examined, and interpreted to highlight the unique benefits of CADD approaches in quickly developing a treatment for this deadly virus. The review’s findings should help researchers who are creating energy-harvesting materials and systems identify new, unanticipated CoV strains or other variations in the future.

Keywords: Computer-aided drug design (CADD), COVID-19, Molecular docking, Pharmacophore modelling, Chemogenomics

[This article belongs to International Journal of Virus Studies(ijvs)]

How to cite this article: Satish kumar Yadav. Advancements in Drug Design Technology and Its Impact on COVID-19 Treatment. International Journal of Virus Studies. 2024; 01(01):-.
How to cite this URL: Satish kumar Yadav. Advancements in Drug Design Technology and Its Impact on COVID-19 Treatment. International Journal of Virus Studies. 2024; 01(01):-. Available from: https://journals.stmjournals.com/ijvs/article=2024/view=147763

References

  1. C. M. Song, S. J. Lim, and J. C. Tong, “Recent advances in computer-aided drug design,” Briefings in Bioinformatics, vol. 10, no. 5, pp. 579–591, 2009.
  2. J. A. DiMasi, H. G. Grabowski, and R. W. Hansen, “Innovation in the pharmaceutical industry: New estimates of R&D costs,” Journal of Health Economics, vol. 47, pp. 20–33, 2016.
  3. D. Vohora and G. Singh, Pharmaceutical Medicine and Translational Clinical Research, Academic Press, 2018.
  4. F. Zhong, J. Xing, X. Li et al., “Artificial intelligence in drug design,” Science China Life Sciences, vol. 61, no. 10, pp. 1191–1204, 2018.
  5. T. Hou and X. Xu, “Recent development and application of virtual screening in drug discovery: an overview,” Current Pharmaceutical Design, vol. 10, no. 9, pp. 1011–1033, 2004.
  6. W. Yu and A. D. Mac Kerell, “Computer-aided drug design methods,” in Antibiotics, Humana Press, New York, NY, 2017.
  7. S. J. Y. Macalino, V. Gosu, S. Hong, and S. Choi, “Role of computer-aided drug design in modern drug discovery,” Archives of Pharmacal Research, vol. 38, no. 9, pp. 1686–1701, 2015.
  8. W. Duch, K. Swaminathan, and J. Meller, “Artificial intelligence approaches for rational drug design and discovery,” Current Pharmaceutical Design, vol. 13, no. 14, pp. 1497–1508, 2007.
  9. H.-J. Huang, H. W. Yu, C.-Y. Chen et al., “Current developments of computer-aided drug design,” Journal of the Taiwan Institute of Chemical Engineers, vol. 41, no. 6, pp. 623–635, 2010.
  10. M. Hassan Baig, K. Ahmad, S. Roy et al., “Computer aided drug design: success and limitations,” Current Pharmaceutical Design, vol. 22, no. 5, pp. 572–581, 2016.
  11. S. Skariyachan, S. B. Challapilli, S. Packirisamy, S. T. Kumargowda, and V. S. Sridhar, “Recent aspects on the pathogenesis mechanism, animal models and novel therapeutic interventions for Middle East respiratory syndrome coronavirus infections,” Frontiers in Microbiology, vol. 10, p. 569, 2019.
  12. S. A. Amin and T. Jha, “Fight against novel coronavirus: a perspective of medicinal chemists,” European Journal of Medicinal Chemistry, vol. 201, article 112559, 2020.
  13. B. Goyal and D. Goyal, “Targeting the dimerization of the main protease of coronaviruses: A potential broad-spectrum therapeutic strategy,” ACS Combinatorial Science, vol. 22, no. 6, pp. 297–305, 2020.
  14. W. Dai, B. Zhang, X.-M. Jiang et al., “Structure-based design of antiviral drug candidates targeting the SARS-CoV-2 main protease,” Science, vol. 368, no. 6497, pp. 1331–1335, 2020.
  15. Y.-F. Tu, C.-S. Chien, A. A. Yarmishyn et al., “A review of SARS-CoV-2 and the ongoing clinical trials,” International Journal of Molecular Sciences, vol. 21, no. 7, p. 2657, 2020.
  16. D. Gopal and S. Skariyachan, Recent Perspectives on COVID-19 and Computer-Aided Virtual Screening of Natural Compounds for the Development of Therapeutic Agents Towards SARS-CoV-2, Methods in Pharmacology and Toxicology, Springer, 2020.
  17. T. Pillaiyar, S. Meenakshisundaram, and M. Manickam, “Recent discovery and development of inhibitors targeting coronaviruses,” Drug Discovery Today, vol. 25, no. 4, pp. 668–688, 2020.
  18. M. Batool, B. Ahmad, and S. Choi, “A structure-based drug discovery paradigm,” International Journal of Molecular Sciences, vol. 20, no. 11, p. 2783, 2019.
  19. E. Lionta, G. Spyrou, D. K. Vassilatis, and Z. Cournia, “Structure-based virtual screening for drug discovery: principles, applications and recent advances,” Current Topics in Medicinal Chemistry, vol. 14, no. 16, pp. 1923–1938, 2014.
  20. S. Kalyaanamoorthy and Y.-P. P. Chen, “Structure-based drug design to augment hit discovery,” Drug Discovery Today, vol. 16, no. 17–18, pp. 831–839, 2011.
  21. A. Wlodawer and J. Vondrasek, “Inhibitors of HIV-1 protease: a major success of structure-assisted drug design,” Annual Review of Biophysics and Biomolecular Structure, vol. 27, no. 1, pp. 249–284, 1998.
  22. D. E. Clark, “What has computer-aided molecular design ever done for drug discovery?” Expert Opinion on Drug Discovery, vol. 1, no. 2, pp. 103–110, 2006.
  23. A. C. Anderson, “The process of structure-based drug design,” Chemistry & Biology, vol. 10, no. 9, pp. 787–797, 2003.
  24. S. Grover, M. A. Apushkin, and G. A. Fishman, “Topical dorzolamide for the treatment of cystoid macular edema in patients with retinitis pigmentosa,” American Journal of Ophthalmology, vol. 141, no. 5, pp. 850–858, 2006.
  1. H. Marrakchi, G. Lanéelle, and A. Quémard, “InhA, a target of the antituberculous drug isoniazid, is involved in a mycobacterial fatty acid elongation system, FAS-II,” Microbiology, vol. 146, no. 2, pp. 289–296, 2000.
  2. S. Dadashpour, T. TuyluKucukkilinc, O. Unsal Tan, K. Ozadali, H. Irannejad, and S. Emami, “Design, synthesis and in vitro study of 5, 6-dDiaryl-1, 2, 4-triazine-3-ylthioacetate derivatives as COX-2 and β-amyloid aggregation inhibitors,” Archiv der Pharmazie, vol. 348, no. 3, pp. 179–187, 2015.
  3. Z. Miller, K.-S. Kim, D.-M. Lee et al., “Proteasome inhibitors with pyrazole scaffolds from structure-based virtual screening,” Journal of Medicinal Chemistry, vol. 58, no. 4, pp. 2036–2041, 2015.
  4. X. Wang, K. Song, L. Li, and L. Chen, “Structure-based drug design strategies and challenges,” Current Topics in Medicinal Chemistry, vol. 18, no. 12, pp. 998–1006, 2018.
  5. V. K. Vyas, R. D. Ukawala, C. Chintha, and M. Ghate, “Homology modeling a fast tool for drug discovery: current perspectives,” Indian Journal of Pharmaceutical Sciences, vol. 74, no. 1, pp. 1–17, 2012.
  6. C. M.-R. Lemer, M. J. Rooman, and S. J. Wodak, “Protein structure prediction by threading methods: evaluation of current techniques,” Proteins: Structure, Function, and Genetics, vol. 23, no. 3, pp. 337–355, 1995.
  7. J. Lee, P. L. Freddolino, and Y. Zhang, “Ab initio protein structure prediction,” in From protein structure to function with bioinformatics, Springer, 2017.
  8. M. T. Muhammed and E. Aki-Yalcin, “Homology modeling in drug discovery: Overview, current applications, and future perspectives,” Chemical Biology & Drug Design, vol. 93, no. 1, pp. 12–20, 2019.
  9. J. Xu, F. Jiao, and L. Yu, “Protein structure prediction using threading,” in Protein structure prediction, Springer, 2008.
  10. M. Yousef, T. Abdelkader, and K. El-Bahnasy, “Performance comparison of ab initio protein structure prediction methods,” Ain Shams Engineering Journal, vol. 10, no. 4, pp. 713–719, 2019.
  11. L. Pan, C. L. Gardner, F. A. Pagliai, C. F. Gonzalez, and G. L. Lorca, “Identification of the tolfenamic acid binding pocket in PrbP from Liberibacterasiaticus,” Frontiers in Microbiology, vol. 8, 2017.
  12. T. A. Binkowski, S. Naghibzadeh, and J. Liang, “CASTp: computed atlas of surface topography of proteins,” Nucleic Acids Research, vol. 31, no. 13, pp. 3352–3355, 2003.
  13. A. Volkamer, D. Kuhn, F. Rippmann, and M. Rarey, “DoGSiteScorer: a web server for automatic binding site prediction, analysis and druggability assessment,” Bioinformatics, vol. 28, no. 15, pp. 2074-2075, 2012.
  14. J. Sun and K. Chen, “NSiteMatch: prediction of binding sites of nucleotides by identifying the structure similarity of local surface patches,” Computational and Mathematical Methods in Medicine, vol. 2017, Article ID 5471607, 16 pages, 2017.
  15. K. P. Tan, R. Varadarajan, and M. S. Madhusudhan, “DEPTH: a web server to compute depth and predict small-molecule binding cavities in proteins,” Nucleic Acids Research, vol. 39, suppl_2, pp. W242–W248, 2011.
  1. Zhu H, Pisabarro MT. MSPocket: an orientation-independent algorithm for the detection of ligand binding pockets. Bioinformatics. 2011 Feb 1;27(3):351-8.
  2. Huang B. MetaPocket: a meta approach to improve protein ligand binding site prediction. OMICS A Journal of Integrative Biology. 2009 Aug 1;13(4):325-30.
  3. Laurie AT, Jackson RM. Q-SiteFinder: an energy-based method for the prediction of protein–ligand binding sites. Bioinformatics. 2005 Feb 8;21(9):1908-16.
  4. Hu Y, Bajorath J. Entering the ‘big data’era in medicinal chemistry: molecular promiscuity analysis revisited. Future science OA. 2017 Mar 6;3(2):FSO179.
  5. Cincilla G, Thormann M, Pons M. Structuring Chemical Space: Similarity‐Based Characterization of the PubChem Database. Molecular Informatics. 2010 Jan 12;29(1‐2):37-49.
  6. Warr W. Report on an NIH workshop on ultralarge chemistry databases.
  7. Oniani D, Hilsman J, Zang C, Wang J, Cai L, Zawala J, Wang Y. Emerging opportunities of using large language models for translation between drug molecules and indications. Scientific Reports. 2024 May 10;14(1):10738.
  8. Heinemann N, Bub S, Wolfram J, Stehle S, Petschick LL, Schulz R. A Compendium of Chemical Class and Use Type Open Access Databases. Data. 2020 Dec 4;5(4):114.
  9. Bitew M, Desalegn T, Demissie TB, Belayneh A, Endale M, Eswaramoorthy R. Pharmacokinetics and drug-likeness of antidiabetic flavonoids: Molecular docking and DFT study. Plos one. 2021 Dec 10;16(12):e0260853.
  10. Zadorozhnii PV, Kiselev VV, Kharchenko AV. In silico ADME profiling of salubrinal and its analogues. Future Pharmacology. 2022 Jun 16;2(2):160-97.
  11. Guedes IA, de Magalhães CS, Dardenne LE. Receptor–ligand molecular docking. Biophysical reviews. 2014 Mar;6:75-87.
  12. Yuriev E, Holien J, Ramsland PA. Improvements, trends, and new ideas in molecular docking: 2012–2013 in review. Journal of Molecular Recognition. 2015 Oct;28(10):581-604.
  13. Spyrakis F, BidonChanal A, Barril X, Javier Luque F. Protein flexibility and ligand recognition: challenges for molecular modeling. Current topics in medicinal chemistry. 2011 Jan 1;11(2):192-210.
  14. Morris GM, Goodsell DS, Huey R, Hart WE, Halliday S, Belew R, Olson AJ. AutoDock. Automated docking of flexible ligands to receptor-User Guide. 2001 Nov 20.
  15. Huey R, Morris GM, Forli S. Using AutoDock 4 and AutoDockvina with AutoDockTools: a tutorial. The Scripps Research Institute Molecular Graphics Laboratory. 2012 Oct 26;10550(92037):1000.
  16. Verdonk ML, Cole JC, Hartshorn MJ, Murray CW, Taylor RD. Improved protein–ligand docking using GOLD. Proteins: Structure, Function, and Bioinformatics. 2003 Sep;52(4):609-23.
  17. Wu G, Robertson DH, Brooks III CL, Vieth M. Detailed analysis of grid‐based molecular docking: A case study of CDOCKER—A CHARMm‐based MD docking algorithm. Journal of computational chemistry. 2003 Oct;24(13):1549-62.
  18. Schellhammer I, Rarey M. FlexX‐Scan: Fast, structure‐based virtual screening. PROTEINS: Structure, Function, and Bioinformatics. 2004 Nov 15;57(3):504-17.
  19. Jain AN. Surflex: fully automatic flexible molecular docking using a molecular similarity-based search engine. Journal of medicinal chemistry. 2003 Feb 13;46(4):499-511.
  20. Halgren TA, Murphy RB, Friesner RA, Beard HS, Frye LL, Pollard WT, Banks JL. Glide: a new approach for rapid, accurate docking and scoring. 2. Enrichment factors in database screening. Journal of medicinal chemistry. 2004 Mar 25;47(7):1750-9.
  21. Allen WJ, Balius TE, Mukherjee S, Brozell SR, Moustakas DT, Lang PT, Case DA, Kuntz ID, Rizzo RC. DOCK 6: Impact of new features and current docking performance. Journal of computational chemistry. 2015 Jun 5;36(15):1132-56.
  22. Bitencourt-Ferreira G, de Azevedo WF. Docking with swissdock. Docking screens for drug discovery. 2019:189-202.
  23. Adcock SA, McCammon JA. Molecular dynamics: survey of methods for simulating the activity of proteins. Chemical reviews. 2006 May 10;106(5):1589-615.
  24. Gubbins KE, Moore JD. Molecular modeling of matter: Impact and prospects in engineering. Industrial & Engineering Chemistry Research. 2010 Apr 7;49(7):3026-46.
  25. Yang Y, Zhu Z, Wang X, Zhang X, Mu K, Shi Y, Peng C, Xu Z, Zhu W. Ligand-based approach for predicting drug targets and for virtual screening against COVID-19. Briefings in Bioinformatics. 2021 Mar;22(2):1053-64.
  26. Tyagi R, Singh A, Chaudhary KK, Yadav MK. Pharmacophoremodeling and its applications. InBioinformatics 2022 Jan 1 (pp. 269-289). Academic Press.
  27. Kutlushina A, Khakimova A, Madzhidov T, Polishchuk P. Ligand-based pharmacophoremodeling using novel 3D pharmacophore signatures. Molecules. 2018 Nov 27;23(12):3094.
  28. So SS. Quantitative Structure‐Activity Relationships. Evolutionary Algorithms in Molecular Design. 2000 Jul 27:71-97.
  29. Pirhadi S, Shiri F, Ghasemi JB. Multivariate statistical analysis methods in QSAR. Rsc Advances. 2015;5(127):104635-65.
  30. Staszak M, Staszak K, Wieszczycka K, Bajek A, Roszkowski K, Tylkowski B. Machine learning in drug design: Use of artificial intelligence to explore the chemical structure–biological activity relationship. Wiley Interdisciplinary Reviews: Computational Molecular Science. 2022 Mar;12(2):e1568.
  31. Nayarisseri A, Khandelwal R, Tanwar P, Madhavi M, Sharma D, Thakur G, Speck-Planche A, Singh SK. Artificial intelligence, big data and machine learning approaches in precision medicine & drug discovery. Current drug targets. 2021 Apr 1;22(6):631-55.
  32. Shanmugam A, Muralidharan N, Velmurugan D, Gromiha MM. Therapeutic targets and computational approaches on drug development for COVID-19. Current Topics in Medicinal Chemistry. 2020 Sep 1;20(24):2210-20.
  33. Gonsalves RC, Pacharla H, Manohar S, Belliraj SK, Tripathi E, Karyala P, Pakala SB. SARS-CoV-2—host cell interactions and pathways: understanding its physiology, pathology, and targeted drug therapy. InPandemic Outbreaks in the 21st Century 2021 Jan 1 (pp. 185-210). Academic Press.
  34. Tušar L, Usenik A, Turk B, Turk D. Mechanisms applied by protein inhibitors to inhibit cysteine proteases. International Journal of Molecular Sciences. 2021 Jan 20;22(3):997.
  35. Kumar S, Paul P, Yadav P, Kaul R, Maitra SS, Jha SK, Chaari A. A multi-targeted approach to identify potential flavonoids against three targets in the SARS-CoV-2 life cycle. Computers in biology and medicine. 2022 Mar 1;142:105231.
  36. Wang Y, Anirudhan V, Du R, Cui Q, Rong L. RNA‐dependent RNA polymerase of SARS‐CoV‐2 as a therapeutic target. Journal of medical virology. 2021 Jan;93(1):300-10.

Regular Issue Subscription Review Article
Volume 01
Issue 01
Received May 13, 2024
Accepted May 21, 2024
Published May 27, 2024