We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleFree Access

Potential drugs against COVID-19 revealed by gene expression profile, molecular docking and molecular dynamic simulation

    Claudia Cava

    *Author for correspondence:

    E-mail Address: claudia.cava@ibfm.cnr.it

    Institute of Molecular Bioimaging & Physiology, National Research Council (IBFM-CNR), Via F. Cervi 93, Segrate-Milan, Milan, 20090, Italy

    ,
    Gloria Bertoli

    Institute of Molecular Bioimaging & Physiology, National Research Council (IBFM-CNR), Via F. Cervi 93, Segrate-Milan, Milan, 20090, Italy

    &
    Isabella Castiglioni

    Department of Physics “Giuseppe Occhialini”, University of Milan-Bicocca Piazza dell'Ateneo Nuovo, Milan, 20126, Italy

    Published Online:https://doi.org/10.2217/fvl-2020-0392

    Aim: SARS-CoV-2, an emerging betacoronavirus, is the causative agent of COVID-19. Currently, there are few specific and selective antiviral drugs for the treatment and vaccines to prevent contagion. However, their long-term effects can be revealed after several years, and new drugs for COVID-19 should continue to be investigated. Materials & methods: In the first step of our study we identified, through a gene expression analysis, several drugs that could act on the biological pathways altered in COVID-19. In the second step, we performed a docking simulation to test the properties of the identified drugs to target SARS-CoV-2. Results: The drugs that showed a higher binding affinity are bardoxolone (-8.78 kcal/mol), irinotecan (-8.40 kcal/mol) and pyrotinib (-8.40 kcal/mol). Conclusion: We suggested some drugs that could be efficient in treating COVID-19.

    References

    • 1. Hall DC Jr, Ji HF. A search for medications to treat COVID-19 via in silico molecular docking models of the SARS-CoV-2 spike glycoprotein and 3CL protease. Travel. Med. Infect. Dis. 35, 101646 (2020).
    • 2. Han DP, Penn-Nicholson A, Cho MW. Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitor. Virology 350(1), 15–25 (2006).
    • 3. Brown JD. Antihypertensive drugs and risk of COVID-19? Lancet Respir. Med. 8(5), e28 (2020).
    • 4. Lu IL, Mahindroo N, Liang PH et al. Structure-based drug design and structural biology study of novel nonpeptide inhibitors of severe acute respiratory syndrome coronavirus main protease. J. Med. Chem. 49(17), 5154–5161 (2006).
    • 5. Blanchard JE, Elowe NH, Huitema C et al. High-throughput screening identifies inhibitors of the SARS coronavirus main proteinase. Chem. Biol. 11(10), 1445–1453 (2004).
    • 6. Zhang L, Lin D, Sun X et al. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors. Science 368(6489), 409–412 (2020).
    • 7. Jin Z, Du X, Xu Y et al. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 582(7811), 289–293 (2020).
    • 8. Cava C, Bertoli G, Castiglioni I. In silico discovery of candidate drugs against COVID-19. Viruses 12(4), 404 (2020).
    • 9. Cheng F, Murray JL, Rubin DH. Drug repurposing: new treatments for Zika virus infection? Trends Mol. Med. 22, 919–921 (2016).
    • 10. Cheng F, Murray JL, Zhao J, Sheng J, Zhao Z, Rubin DH. Systems biology-based investigation of cellular antiviral drug targets identified by gene-trap insertional mutagenesis. PLoS Comput. Biol. 12, e1005074 (2016).
    • 11. Mustafa S, Balkhy H, Gabere M. Peptide–protein interaction studies of antimicrobial peptides targeting middle east respiratory syndrome coronavirus spike protein: an in silico approach. Adv. Bioinform. 2019, doi: 10.1155/2019/6815105 (2019).
    • 12. Lionta E, Spyrou G, Vassilatis DK, Cournia Z. Structure-based virtual screening for drug discovery: principles, applications and recent advances. Curr. Top. Med. Chem. 14, 1923–1938 (2014).
    • 13. Cheng LS, Amaro RE, Xu D, Li WW, Arzberger PW, McCammon JA. Ensemble-based virtual screening reveals potential novel antiviral compounds for avian influenza neuraminidase. J. Med. Chem. 51, 3878–3894 (2008).
    • 14. Zhang L, Ai HX, Li SM et al. Virtual screening approach to identifying influenza virus neuraminidase inhibitors using molecular docking combined with machine-learning-based scoring function. Oncotarget 8(47), 83142–83154 (2017).
    • 15. Shah B, Modi P, Sagar SR. In silico studies on therapeutic agents for COVID-19: drug repurposing approach. Life Sci. 252, 117652 (2020).
    • 16. Aatif M, Muteeb G, Alsultan A, Alshoaibi A, Khelif BY. Dieckol and its derivatives as potential inhibitors of SARS-CoV-2 spike protein (UK strain: VUI 202012/01): a computational study. Mar. Drugs 19(5), 242 (2021).
    • 17. Prabhu D, Rajamanikandan S, Sureshan M, Jeyakanthan J, Saraboji K. modeling studies reveal the importance of the C-terminal inter motif loop of NSP1 as a promising target site for drug discovery and screening of potential phytochemicals to combat SARS-CoV-2. J. Mol. Graph. Model. 106, 107920 (2021).
    • 18. Vivek-Ananth RP, Rana A, Rajan N, Biswal HS, Samal A. In silico identification of potential natural product inhibitors of human proteases key to SARS-CoV-2 infection. Molecules 25(17), 3822 (2020).
    • 19. Gentile F, Agrawal V, Hsing M et al. Deep docking: a deep learning platform for augmentation of structure based drug discovery. ACS Cent. Sci. 6(6), 939–949 (2020).
    • 20. Nguyen DD, Gao K, Chen J, Wang R, Wei GW. Potentially highly potent drugs for 2019-nCoV. Biorxiv doi: https://doi.org/10.1101/2020.02.05.936013 (2020).
    • 21. Salmaso V, Moro S. Bridging molecular docking to molecular dynamics in exploring ligand-protein recognition process: an overview. Front. Pharmacol. 9, 923 (2018).
    • 22. Colaprico A, Silva TC, Olsen C et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 44(8), e71 (2016).
    • 23. Günther S, Kuhn M, Dunkel M et al. SuperTarget and Matador: resources for exploring drug-target relationships. Nucleic Acids Res. 36(Database issue), D919–D922 (2008).
    • 24. Cotto KC, Wagner AH, Feng YY et al. DGIdb 3.0: a redesign and expansion of the drug-gene interaction database. Nucleic Acids Res. 46(D1), D1068–D1073 (2018).
    • 25. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B Methodol. 57, 289–300 (1995).
    • 26. Morris GM, Huey R, Lindstrom W et al. AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility. J. Comput. Chem. 30(16), 2785–2791 (2009).
    • 27. Kim S, Chen J, Cheng T et al. PubChem 2019 update: improved access to chemical data. Nucleic Acids Res. 47(D1), D1102–D1109 (2019).
    • 28. Dassault Systèmes. BIOVIA, Discovery Studio Modeling Environment, release. (2020). https://www.3ds.com/products-services/biovia/products/molecular-modeling-simulation/biovia-discovery-studio/
    • 29. Salentin S, Schreiber S, Haupt VJ, Adasme MF, Schroeder M. PLIP: fully automated protein–ligand interaction profiler. Nucleic Acids Res. 43(W1), W443–447 (2015).
    • 30. Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci. Rep. 7, 42717 (2017).
    • 31. Hastings J, Owen G, Dekker A et al. ChEBI in 2016: improved services and an expanding collection of metabolites. Nucleic Acids Res. 44(D1), D1214–D1219 (2016).
    • 32. Gentile P, Sterodimas A, Pizzicannella J, Calabrese C, Garcovich S. Research progress on mesenchymal stem cells (MSCs), adipose-derived mesenchymal stem cells (AD-MSCs), drugs, and vaccines in inhibiting COVID-19 disease. Aging Dis. 11(5), 1191–1201 (2020).
    • 33. Gentile P, Sterodimas A. Adipose stem cells (ASCs) and stromal vascular fraction (SVF) as a potential therapy in combating (COVID-19)-disease. Aging Dis. 11(3), 465–469 (2020).
    • 34. Gentile P, Sterodimas A. Adipose-derived stromal stem cells (ASCs) as a new regenerative immediate therapy combating coronavirus (COVID-19)-induced pneumonia. Expert Opin. Biol. Ther. 20(7), 711–716 (2020).
    • 35. Durand N, Mallea J, Zubair AC. Insights into the use of mesenchymal stem cells in COVID-19 mediated acute respiratory failure. NPJ Regen. Med. 5(1), 17 (2020).
    • 36. Rad F, Ghorbani M, Mohammadi Roushandeh A, Habibi Roudkenar M. Mesenchymal stem cell-based therapy for autoimmune diseases: emerging roles of extracellular vesicles. Mol. Biol. Rep. 46(1), 1533–1549 (2019).
    • 37. Food and Drug Administration. Emergency Use Authorization. https://www.fda.gov/emergency-preparedness-and-response/mcm-legal-regulatory-and-policy-framework/emergency-use-authorization#coviddrugs
    • 38. Gordon CJ, Tchesnokov EP, Feng JY, Porter DP, Götte M. The antiviral compound remdesivir potently inhibits RNA-dependent RNA polymerase from Middle East respiratory syndrome coronavirus. J. Biol. Chem. 295(15), 4773–4779 (2020).
    • 39. Ledford H. COVID antibody treatments show promise for preventing severe disease. Nature 591(7851), 513–514 (2021).
    • 40. Huang DT, McCreary EK, Bariola JR et al. The UPMC OPTIMISE-C19 (optimizing treatment and impact of monoclonal antibodies through evaluation for COVID-19) trial: a structured summary of a study protocol for an open-label, pragmatic, comparative effectiveness platform trial with response-adaptive randomization. Trials 22(1), 363 (2021).
    • 41. Hansen J, Baum A, Pascal KE et al. Studies in humanized mice and convalescent humans yield a SARS-CoV-2 antibody cocktail. Science 369(6506), 1010–1014 (2020).
    • 42. Devarasetti PK, Rajasekhar L, Baisya R, Sreejitha KS, Vardhan YK. A review of COVID-19 convalescent plasma use in COVID-19 with focus on proof of efficacy. Immunol. Res. 69(1), 18–25 (2021).
    • 43. Naqvi AAT, Fatima K, Mohammad T et al. Insights into SARS-CoV-2 genome, structure, evolution, pathogenesis and therapies: structural genomics approach. Biochim. Biophys. Acta. Mol. Basis Dis. 1866(10), 165878 (2020).
    • 44. Klemm T, Ebert G, Calleja DJ et al. Mechanism and inhibition of the papain-like protease, PLpro, of SARS-CoV-2. EMBO J. 39(18), e106275 (2020).
    • 45. Bepari AK, Reza HM. Identification of a novel inhibitor of SARS-CoV-2 3CL-PRO through virtual screening and molecular dynamics simulation. Peer J. 9, e11261 (2021).
    • 46. Nio Y, Sasai M, Akahori Y et al. Bardoxolone methyl as a novel potent antiviral agent against hepatitis B and C viruses in human hepatocyte cell culture systems. Antiviral Res. 169, 104537 (2019).
    • 47. Wyler E, Franke V, Menegatti J et al. Single-cell RNA-sequencing of herpes simplex virus 1-infected cells connects NRF2 activation to an antiviral program. Nat. Commun. 10(1), 4878 (2019).
    • 48. Patra U, Mukhopadhyay U, Sarkar R, Mukherjee A, Chawla-Sarkar M. RA-839, a selective agonist of Nrf2/ARE pathway, exerts potent anti-rotaviral efficacy in vitro. Antiviral Res. 161, 53–62 (2019).
    • 49. Kulu Y, Kawasaki H, Donahue JM et al. Concurrent chemotherapy inhibits herpes simplex virus-1 replication and oncolysis. Cancer Gene Ther. 20(2), 133–140 (2013).
    • 50. Lipid Nanocarriers for Drug Targeting Grumezescu AM (Ed.). William Andrew (2018).
    • 51. Lui LCY, Chan RTT, Kwok CCH et al. Reactivation of hepatitis B after irinotecan. J. Hong Kong College of Radiologists 7(2), 72 (2004).
    • 52. Gourd E. Pyrotinib shows activity in metastatic breast cancer. Lancet Oncol. 18(11), e643 (2017).
    • 53. Sisk JM, Frieman MB, Machamer CE. Coronavirus S protein-induced fusion is blocked prior to hemifusion by Abl kinase inhibitors. J. Gen. Virol. 99(5), 619–630 (2018).
    • 54. Curtin N, Bányai K, Thaventhiran J, Le Quesne J, Helyes Z, Bai P. Repositioning PARP inhibitors for SARS-CoV-2 infection(COVID-19); a new multi-pronged therapy for acute respiratory distress syndrome? Br. J. Pharmacol. 177(16), 3635–3645 (2020).
    • 55. Morrow DA, Brickman CM, Murphy SA et al. A randomized, placebo-controlled trial to evaluate the tolerability, safety, pharmacokinetics, and pharmacodynamics of a potent inhibitor of poly(ADP-ribose) polymerase (INO-1001) in patients with ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention: results of the TIMI 37 trial. J. Thromb. Thrombolysis. 27(4), 359–364 (2009).
    • 56. Sabetian S, Castiglioni I, Jahromi BN, Mousavi P, Cava C. In silico identification of miRNA-lncRNA interactions in male reproductive disorder associated with COVID-19 infection. Cells. 10(6), 1480 (2021).
    • 57. Cava C, Bertoli G, Castiglioni I. A protein interaction map identifies existing drugs targeting SARS-CoV-2. BMC Pharmacol Toxicol. 21(1), 65 (2020).