Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Research Article

Repositioning Therapeutics for SARS-CoV-2: Virtual Screening of Plant-based Anti-HIV Compounds as Possible Inhibitors against COVID-19 Viral RdRp

Author(s): Mahadevamurthy Murali, Hittanahallikoppal Gajendramurthy Gowtham, Mohammad Azam Ansari*, Mohammad N. Alomary, Saad Alghamdi, Mazen Almehmadi, Sudarshana Brijesh Singh, Natarajamurthy Shilpa, Mohammed Aiyaz, Nataraj Kalegowda, Ana E. Ledesma* and Kestur Nagaraj Amruthesh*

Volume 28, Issue 12, 2022

Published on: 02 June, 2022

Page: [969 - 980] Pages: 12

DOI: 10.2174/1381612828666220428120939

Price: $65

Abstract

Background: Coronavirus disease 2019 (COVID-19) has caused a global pandemic with a high mortality and morbidity rate worldwide. The COVID-19 vaccines that are currently in development or already approved are expected to provide at least some protection against the emerging variants of the virus, but the mutations may reduce the efficacy of the existing vaccines. Purified phytochemicals from medicinal plants provide a helpful framework for discovering new therapeutic leads as they have long been employed in traditional medicine to treat many disorders.

Objective: The objectives of the study are to exploit the anti-HIV bioactive compounds against SARS-CoV-2 RNA-dependent RNA polymerase (RdRp) through molecular docking studies and to evaluate the Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) properties of potential compounds.

Methods: Molecular docking was performed to study the interaction of ligands with the target sites of RdRp protein (PDB: 6M71) using AutoDock Vina. The ADMET properties of potential compounds were predicted using the pkCSM platform.

Results: A total of 151 phytochemicals derived from the medicinal plants with recognized antiviral activity and 18 anti-HIV drugs were virtually screened against COVID-19 viral RdRp to identify putative inhibitors that facilitate the development of potential anti-COVID-19 drug candidates. The computational studies identified 34 compounds and three drugs inhibiting viral RdRp with binding energies ranging from -10.2 to -8.5 kcal/mol. Among them, five compounds, namely Michellamine B, Quercetin 3-O-(2'',6''-digalloyl)-beta-Dgalactopyranoside, Corilagin, Hypericin, and 1,2,3,4,6-Penta-O-galloyl-beta-D-glucose residues, bound efficiently with the binding site of RdRp. Besides, Lopinavir, Maraviroc, and Remdesivir drugs also inhibited SARS-CoV-2 polymerase. In addition, the ADMET properties of top potential compounds were also predicted in comparison to the drugs.

Conclusion: The present study suggested that these potential drug candidates can be further subjected to in vitro and in vivo studies that may help develop effective anti-COVID-19 drugs.

Keywords: Anti-COVID-19 drugs, anti-HIV compounds, coronaviruses, molecular docking, RNA-dependent RNA polymerase, hypericin.

[1]
Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: A descriptive study. Lancet 2020; 395(10223): 507-13.
[http://dx.doi.org/10.1016/S0140-6736(20)30211-7] [PMID: 32007143]
[2]
Pal M, Berhanu G, Desalegn C, Kandi V. Severe acute respiratory syndrome Coronavirus-2 (SARS-CoV-2): An update. Cureus 2020; 12(3): e7423.
[http://dx.doi.org/10.7759/cureus.7423] [PMID: 32337143]
[3]
Kumar R, Mishra S. Shreya, Maurya SK. Recent advances in the discovery of potent RNA-dependent RNA-polymerase (RdRp) inhibitors targeting viruses. RSC Med Chem 2020; 12(3): 306-20.
[http://dx.doi.org/10.1039/D0MD00318B] [PMID: 34046618]
[4]
Gao Y, Yan L, Huang Y, et al. Structure of the RNA-dependent RNA polymerase from COVID-19 virus. Science 2020; 368(6492): 779-82.
[http://dx.doi.org/10.1126/science.abb7498] [PMID: 32277040]
[5]
Wang Y, Anirudhan V, Du R, Cui Q, Rong L. RNA-dependent RNA polymerase of SARS-CoV-2 as a therapeutic target. J Med Virol 2021; 93(1): 300-10.
[http://dx.doi.org/10.1002/jmv.26264] [PMID: 32633831]
[6]
Venkataraman S, Prasad BVLS, Selvarajan R. RNA dependent RNA polymerases: Insights from structure, function and evolution. Viruses 2018; 10(2): 76.
[http://dx.doi.org/10.3390/v10020076] [PMID: 29439438]
[7]
Zhou Z, Liu T, Zhang J, Zhan P, Liu X. Influenza A virus polymerase: An attractive target for next-generation anti-influenza therapeutics. Drug Discov Today 2018; 23(3): 503-18.
[http://dx.doi.org/10.1016/j.drudis.2018.01.028] [PMID: 29339107]
[8]
Tian L, Qiang T, Liang C, et al. RNA-dependent RNA polymerase (RdRp) inhibitors: The current landscape and repurposing for the COVID-19 pandemic. Eur J Med Chem 2021; 213: 113201.
[http://dx.doi.org/10.1016/j.ejmech.2021.113201] [PMID: 33524687]
[9]
Chien M, Anderson TK, Jockusch S, et al. Nucleotide analogues as inhibitors of SARS-CoV-2 polymerase, a key drug target for COVID-19. J Proteome Res 2020; 19(11): 4690-7.
[http://dx.doi.org/10.1021/acs.jproteome.0c00392] [PMID: 32692185]
[10]
Elfiky AA. Ribavirin, Remdesivir, Sofosbuvir, Galidesivir, and Tenofovir against SARS-CoV-2 RNA dependent RNA polymerase (RdRp): A molecular docking study. Life Sci 2020; 253: 117592.
[http://dx.doi.org/10.1016/j.lfs.2020.117592] [PMID: 32222463]
[11]
Bhuiyan FR, Howlader S, Raihan T, Hasan M. Plants metabolites: Possibility of natural therapeutics against the COVID-19 pandemic. Front Med (Lausanne) 2020; 7: 444.
[http://dx.doi.org/10.3389/fmed.2020.00444] [PMID: 32850918]
[12]
Puttaswamy H, Gowtham HG, Ojha MD, et al. In silico studies evidenced the role of structurally diverse plant secondary metabolites in reducing SARS-CoV-2 pathogenesis. Sci Rep 2020; 10(1): 20584.
[http://dx.doi.org/10.1038/s41598-020-77602-0] [PMID: 33239694]
[13]
Forni G, Mantovani A. COVID-19 Commission of Accademia Nazionale dei Lincei, Rome. COVID-19 vaccines: Where we stand and challenges ahead. Cell Death Differ 2021; 28(2): 626-39.
[http://dx.doi.org/10.1038/s41418-020-00720-9] [PMID: 33479399]
[14]
Kurapati KR, Atluri VS, Samikkannu T, Garcia G, Nair MPN. Natural products as anti-HIV agents and role in HIV-Associated Neurocognitive Disorders (HAND): A brief overview. Front Microbiol 2016; 6: 1444.
[http://dx.doi.org/10.3389/fmicb.2015.01444] [PMID: 26793166]
[15]
Laila U, Akram M, Shariati MA, et al. Role of medicinal plants in HIV/AIDS therapy. Clin Exp Pharmacol Physiol 2019; 46(12): 1063-73.
[http://dx.doi.org/10.1111/1440-1681.13151] [PMID: 31365763]
[16]
Pettersen EF, Goddard TD, Huang CC, et al. UCSF Chimera--a visualization system for exploratory research and analysis. J Comput Chem 2004; 25(13): 1605-12.
[http://dx.doi.org/10.1002/jcc.20084] [PMID: 15264254]
[17]
Trott O, Olson AJ. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31(2): 455-61.
[http://dx.doi.org/10.1002/jcc.21334] [PMID: 19499576]
[18]
Dallakyan S, Olson AJ. Small-molecule library screening by docking with PyRx. Methods Mol Biol 2015; 1263: 243-50.
[http://dx.doi.org/10.1007/978-1-4939-2269-7_19] [PMID: 25618350]
[19]
Pires DEV, Blundell TL, Ascher DB. pkCSM: Predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J Med Chem 2015; 58(9): 4066-72.
[http://dx.doi.org/10.1021/acs.jmedchem.5b00104] [PMID: 25860834]
[20]
Appleby TC, Perry JK, Murakami E, et al. Viral replication. Structural basis for RNA replication by the hepatitis C virus polymerase. Science 2015; 347(6223): 771-5.
[http://dx.doi.org/10.1126/science.1259210] [PMID: 25678663]
[21]
Gong P, Peersen OB. Structural basis for active site closure by the poliovirus RNA-dependent RNA polymerase. Proc Natl Acad Sci USA 2010; 107(52): 22505-10.
[http://dx.doi.org/10.1073/pnas.1007626107] [PMID: 21148772]
[22]
Khan A, Khan M, Saleem S, et al. Phylogenetic analysis and structural perspectives of RNA-dependent RNA-polymerase inhibition from SARS-CoV-2 with natural products. Interdiscip Sci 2020; 12(3): 335-48.
[http://dx.doi.org/10.1007/s12539-020-00381-9] [PMID: 32617855]
[23]
Shaikh VS, Shaikh Y, Ahmed K. Lopinavir as a potential inhibitor for SARS-CoV-2 target protein: A molecular docking study. SSRN 2020.
[http://dx.doi.org/10.2139/ssrn.3596820]
[24]
Mothay D, Ramesh KV. Binding site analysis of potential protease inhibitors of COVID-19 using AutoDock. Virusdisease 2020; 31(2): 194-9.
[http://dx.doi.org/10.1007/s13337-020-00585-z] [PMID: 32363219]
[25]
Mamidala E, Davella R, Gurrapu S, Shivakrishna P. In silico identification of clinically approved medicines against the main protease of SARS-CoV-2, causative agent of COVID-19. arXiv preprint 2020.
[26]
Bansal V, Mahapure KS, Bhurwal A, et al. Mortality benefit of remdesivir in COVID-19: A systematic review and meta-analysis. Front Med (Lausanne) 2021; 7: 606429.
[http://dx.doi.org/10.3389/fmed.2020.606429] [PMID: 33585508]
[27]
Khan FI, Kang T, Ali H, Lai D. Remdesivir strongly binds to RNA-dependent RNA polymerase, membrane protein, and main protease of SARS-CoV-2: Indication from molecular modeling and simulations. Front Pharmacol 2021; 12: 710778.
[http://dx.doi.org/10.3389/fphar.2021.710778] [PMID: 34305617]
[28]
Schaftenaar G, de Vlieg J. Quantum mechanical polar surface area. J Comput Aided Mol Des 2012; 26(3): 311-8.
[http://dx.doi.org/10.1007/s10822-012-9557-y] [PMID: 22391921]
[29]
Arnott JA, Planey SL. The influence of lipophilicity in drug discovery and design. Expert Opin Drug Discov 2012; 7(10): 863-75.
[http://dx.doi.org/10.1517/17460441.2012.714363] [PMID: 22992175]
[30]
Amin ML. P-glycoprotein inhibition for optimal drug delivery. Drug Target Insights 2013; 7: 27-34.
[http://dx.doi.org/10.4137/DTI.S12519] [PMID: 24023511]
[31]
Watanabe R, Esaki T, Kawashima H, et al. Predicting fraction unbound in human plasma from chemical structure: Improved accuracy in the low value ranges. Mol Pharm 2018; 15(11): 5302-11.
[http://dx.doi.org/10.1021/acs.molpharmaceut.8b00785] [PMID: 30259749]
[32]
Pardridge WM. Drug transport across the blood-brain barrier. J Cereb Blood Flow Metab 2012; 32(11): 1959-72.
[http://dx.doi.org/10.1038/jcbfm.2012.126] [PMID: 22929442]
[33]
McDonnell AM, Dang CH. Basic review of the cytochrome p450 system. J Adv Pract Oncol 2013; 4(4): 263-8.
[http://dx.doi.org/10.6004/jadpro.2013.4.4.7] [PMID: 25032007]
[34]
Baell JB, Holloway GA. New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. J Med Chem 2010; 53(7): 2719-40.
[http://dx.doi.org/10.1021/jm901137j] [PMID: 20131845]

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