Identification of existing pharmaceuticals and herbal medicines as inhibitors of SARS-CoV-2 infection

Jia-Tsrong Jan, Ting-Jen Rachel Cheng, Yu-Pu Juang, Hsiu-Hua Ma, Ying-Ta Wu, Wen-Bin Yang, Cheng-Wei Cheng, Xiaorui Chen, Ting-Hung Chou, Jiun-Jie Shie, Wei-Chieh Cheng, Rong-Jie Chein, Shi-Shan Mao, Pi-Hui Liang, Che Ma, Shang-Cheng Hung, Chi-Huey Wong, Jia-Tsrong Jan, Ting-Jen Rachel Cheng, Yu-Pu Juang, Hsiu-Hua Ma, Ying-Ta Wu, Wen-Bin Yang, Cheng-Wei Cheng, Xiaorui Chen, Ting-Hung Chou, Jiun-Jie Shie, Wei-Chieh Cheng, Rong-Jie Chein, Shi-Shan Mao, Pi-Hui Liang, Che Ma, Shang-Cheng Hung, Chi-Huey Wong

Abstract

The outbreak of COVID-19 caused by SARS-CoV-2 has resulted in more than 50 million confirmed cases and over 1 million deaths worldwide as of November 2020. Currently, there are no effective antivirals approved by the Food and Drug Administration to contain this pandemic except the antiviral agent remdesivir. In addition, the trimeric spike protein on the viral surface is highly glycosylated and almost 200,000 variants with mutations at more than 1,000 positions in its 1,273 amino acid sequence were reported, posing a major challenge in the development of antibodies and vaccines. It is therefore urgently needed to have alternative and timely treatments for the disease. In this study, we used a cell-based infection assay to screen more than 3,000 agents used in humans and animals, including 2,855 small molecules and 190 traditional herbal medicines, and identified 15 active small molecules in concentrations ranging from 0.1 nM to 50 μM. Two enzymatic assays, along with molecular modeling, were then developed to confirm those targeting the virus 3CL protease and the RNA-dependent RNA polymerase. Several water extracts of herbal medicines were active in the cell-based assay and could be further developed as plant-derived anti-SARS-CoV-2 agents. Some of the active compounds identified in the screen were further tested in vivo, and it was found that mefloquine, nelfinavir, and extracts of Ganoderma lucidum (RF3), Perilla frutescens, and Mentha haplocalyx were effective in a challenge study using hamsters as disease model.

Keywords: SARS-CoV-2; antiviral; cell-based and animal studies; drug repurposing.

Conflict of interest statement

The authors declare no competing interest.

Copyright © 2021 the Author(s). Published by PNAS.

Figures

Fig. 1.
Fig. 1.
Representative drugs showed antiinfective effects at 10 μM. These drugs are categorized according to their potential mode of action against SARS-CoV-2.
Fig. 2.
Fig. 2.
Dose–response relationships of 15 selected antiviral compounds. Vero E6 cells were pretreated with compounds at indicated doses followed by SARS-CoV-2 infection for 48 h. The percentage of viral titer determined by antinucleocapsid antibody after drug treatment (red) and cell viability (blue) were measured and expressed as mean ± SD of at least three independent experiments.
Fig. 3.
Fig. 3.
Structures of protease inhibitors and their IC50 and Ki values for 3CL protease inhibition. The values were determined from three independent experiments using FRET-based enzymatic assays.
Fig. 4.
Fig. 4.
Computer simulation of (A) nelfinavir and (B) boceprevir binding to SARS-CoV-2 3CL protease (PDB ID code 6LU7). Pink dashed lines indicated interaction between compounds and protein.
Fig. 5.
Fig. 5.
SARS-CoV-2 spike protein sequence mutation analysis. (A) Analysis of spike protein mutation from the 196,276 sequences revealed 1,141 sites of mutation in the 1,273 amino acids of spike protein. The spike protein sequence of hCoV-19/Taiwan/4/2020 used in this study was identical to the original virus strain (UniProt Entry: P0DTC2). Single bottom line: S1 region (residues 13 to 685); double bottom line: S2 region (residues 686 to 1273); yellow bottom line: receptor binding domain (residues 319 to 541); green top line: N-glycosylation motifs; blue top line: O-glycosylation sites; pink: mutation residues; sequence representative: P0DTC2 (UniProt Entry). (B) Top and side view of the S protein indicating residue variants (from strictly conserved to highly variable: white to red), N-glycans (green stick), O-glycosylation sites (blue) in three-dimensional structure (PDB ID code 7CN9).
Fig. 6.
Fig. 6.
Evaluations of antiviral effect of Chinese herbal medicines in serial dilutions were presented as log2(dilution fold). Anti–SARS-CoV-2 infection effects of selected Chinese herbal medicines as water extracts (1.0 g/20 mL H2O) and RF3 dissolved in H2O (0.25 mg/mL) are presented. The tested results of all Chinese herbal medicines are summarized in SI Appendix, Fig. S7.
Fig. 7.
Fig. 7.
In vivo anti–SARS-CoV-2 assay conducted in female golden Syrian hamsters. (A) Virus elimination effect of drugs and extracts. Hamsters were infected with SAR-CoV-2 by intranasal instillation at day 0, and treated with drugs and extracts orally twice a day (30 mg/kg/d for drugs and 200 mg/kg/d for extracts) continuously for 3 d. After 3 d, the lungs were collected to measure the virus load (n = 5), *P < 0.05; **P < 0.005. (B) Body-weight change after 3-d treatment, n = 5 for test group and n = 6 for the control group.

References

    1. Del Rio C., Malani P. N., COVID-19—New insights on a rapidly changing epidemic. JAMA 323, 1339–1340 (2020).
    1. Helmy Y. A., et al. , The COVID-19 pandemic: A comprehensive review of taxonomy, genetics, epidemiology, diagnosis, treatment, and control. J. Clin. Med. 9, 1225 (2020).
    1. Hoffmann M., et al. , SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 181, 271–280.e8 (2020).
    1. Sola I., Almazán F., Zúñiga S., Enjuanes L., Continuous and discontinuous RNA synthesis in coronaviruses. Annu. Rev. Virol. 2, 265–288 (2015).
    1. Wang M., et al. , Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res. 30, 269–271 (2020).
    1. Mitjà al. .; BCN PEP-CoV-2 RESEARCH GROUP , Hydroxychloroquine for early treatment of adults with mild Covid-19: A randomized-controlled trial. Clin. Infect. Dis., ciaa1009 (2020).
    1. Gordon D. E., et al. , A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 583, 459–468 (2020).
    1. Bouhaddou M., et al. , The global phosphorylation landscape of SARS-CoV-2 infection. Cell 182, 685–712.e19 (2020).
    1. Watanabe Y., Allen J. D., Wrapp D., McLellan J. S., Crispin M., Site-specific glycan analysis of the SARS-CoV-2 spike. Science 369, 330–333 (2020).
    1. Shang J., et al. , Structural basis of receptor recognition by SARS-CoV-2. Nature 581, 221–224 (2020).
    1. Walls A. C., et al. , Structure, function, and antigenicity of the SARS-CoV-2 spike glycoprotein. Cell 181, 281–292.e6 (2020).
    1. Wu C. Y., et al. , Small molecules targeting severe acute respiratory syndrome human coronavirus. Proc. Natl. Acad. Sci. U.S.A. 101, 10012–10017 (2004).
    1. Matsuyama S., et al. , Enhanced isolation of SARS-CoV-2 by TMPRSS2-expressing cells. Proc. Natl. Acad. Sci. U.S.A. 117, 7001–7003 (2020).
    1. Yamamoto N., et al. , HIV protease inhibitor nelfinavir inhibits replication of SARS-associated coronavirus. Biochem. Biophys. Res. Commun. 318, 719–725 (2004).
    1. Cao B., et al. , A trial of lopinavir-ritonavir in adults hospitalized with severe Covid-19. N. Engl. J. Med. 382, 1787–1799 (2020).
    1. Group R. C.; RECOVERY Collaborative Group , Lopinavir-ritonavir in patients admitted to hospital with COVID-19 (RECOVERY): A randomised, controlled, open-label, platform trial. Lancet, S0140-6736(20)32013-4 (2020).
    1. Arshad U., et al. , Prioritization of anti-SARS-Cov-2 drug repurposing opportunities based on plasma and target site concentrations derived from their established human pharmacokinetics. Clin. Pharmacol. Ther. 108, 775–790 (2020).
    1. Shao Y. M., et al. , Structure-based design and synthesis of highly potent SARS-CoV 3CL protease inhibitors. ChemBioChem 8, 1654–1657 (2007).
    1. Musarrat F., et al. , The anti-HIV drug nelfinavir mesylate (Viracept) is a potent inhibitor of cell fusion caused by the SARSCoV-2 spike (S) glycoprotein warranting further evaluation as an antiviral against COVID-19 infections. J. Med. Virol. 92, 2087–2095 (2020).
    1. de Boer N. K., van Bodegraven A. A., Jharap B., de Graaf P., Mulder C. J., Drug insight: Pharmacology and toxicity of thiopurine therapy in patients with IBD. Nat. Clin. Pract. Gastroenterol. Hepatol. 4, 686–694 (2007).
    1. Chou C. Y., et al. , Thiopurine analogues inhibit papain-like protease of severe acute respiratory syndrome coronavirus. Biochem. Pharmacol. 75, 1601–1609 (2008).
    1. Swaim C. D., et al. , 6-Thioguanine blocks SARS-CoV-2 replication by inhibition of PLpro protease activities. bioRxiv [Preprint] (2020) 10.1101/2020.07.01.183020 (Accessed 1 July 2020).
    1. Shu Y., McCauley J., GISAID: Global initiative on sharing all influenza data—From vision to reality. Euro Surveill. 22, 2–4 (2017).
    1. Shajahan A., Supekar N. T., Gleinich A. S., Azadi P., Deducing the N- and O- glycosylation profile of the spike protein of novel coronavirus SARS-CoV-2. Glycobiology, cwaa042 (2020).
    1. Williams S. J., Goddard-Borger E. D., α-glucosidase inhibitors as host-directed antiviral agents with potential for the treatment of COVID-19. Biochem. Soc. Trans. 48, 1287–1295 (2020).
    1. Uhlén M., et al. , Proteomics. Tissue-based map of the human proteome. Science 347, 1260419 (2015).
    1. Starr T. N., et al. , Deep mutational scanning of SARS-CoV-2 receptor binding domain reveals constraints on folding and ACE2 binding. Cell 182, 1295–1310.e20 (2020).
    1. Casalino L., et al. , Beyond shielding: The roles of glycans in the SARS-CoV-2 spike protein. ACS Cent. Sci. 6, 1722–1734 (2020).
    1. Moore S. E., Spiro R. G., Demonstration that Golgi endo-alpha-D-mannosidase provides a glucosidase-independent pathway for the formation of complex N-linked oligosaccharides of glycoproteins. J. Biol. Chem. 265, 13104–13112 (1990).
    1. Rajsekharan S., et al. , Repurposing of miglustat to inhibit the coronavirus severe acquired respiratory syndrome SARS-CoV-2. bioRxiv [Preprint] (2020) 10.1101/2020.05.18.101691 (Accessed 20 May 2020).
    1. Bailly C., Cepharanthine: An update of its mode of action, pharmacological properties and medical applications. Phytomedicine 62, 152956 (2019).
    1. Ohashi H., et al. , Multidrug treatment with nelfinavir and cepharanthine against COVID-19. bioRxiv [Preprint] (2020) 10.1101/2020.04.14.039925 (Accessed 14 April 2020).
    1. Shen L., et al. , High-throughput screening and identification of potent broad-spectrum inhibitors of coronaviruses. J. Virol. 93, e00023-19 (2019).
    1. Choy K. T., et al. , Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro. Antiviral Res. 178, 104786 (2020).
    1. Wagstaff K. M., Sivakumaran H., Heaton S. M., Harrich D., Jans D. A., Ivermectin is a specific inhibitor of importin α/β-mediated nuclear import able to inhibit replication of HIV-1 and dengue virus. Biochem. J. 443, 851–856 (2012).
    1. Schmith V. D., Zhou J. J., Lohmer L. R. L., The approved dose of ivermectin alone is not the ideal dose for the treatment of COVID-19. Clin. Pharmacol. Ther. 108, 762–765 (2020).
    1. Liu J., et al. , Hydroxychloroquine, a less toxic derivative of chloroquine, is effective in inhibiting SARS-CoV-2 infection in vitro. Cell Discov. 6, 16 (2020).
    1. Sze C. W., Tan Y. J., Viral membrane channels: Role and function in the virus life cycle. Viruses 7, 3261–3284 (2015).
    1. Hover S., Foster B., Barr J. N., Mankouri J., Viral dependence on cellular ion channels—An emerging anti-viral target? J. Gen. Virol. 98, 345–351 (2017).
    1. Jeon S., et al. , Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs. Antimicrob. Agents Chemother. 64, e00819–e00820 (2020).
    1. Gassen N. C. P., et al. , Analysis of SARS-CoV-2-controlled autophagy reveals spermidine, MK-2206, and niclosamide as putative antiviral therapeutics. bioRxiv[Preprint] (2020) 10.1101/2020.04.15.997254 (Accessed 16 April 2020).
    1. Kondratskyi A., Kondratska K., Skryma R., Klionsky D. J., Prevarskaya N., Ion channels in the regulation of autophagy. Autophagy 14, 3–21 (2018).
    1. Lippi G., South A. M., Henry B. M., Electrolyte imbalances in patients with severe coronavirus disease 2019 (COVID-19). Ann. Clin. Biochem. 57, 262–265 (2020).
    1. Yu M.-S., et al. , Identification of myricetin and scutellarein as novel chemical inhibitors of the SARS coronavirus helicase, nsP13. Bioorg. Med. Chem. Lett. 22, 4049–4054 (2012).
    1. Joseph J., et al. , Green tea and Spirulina extracts inhibit SARS, MERS, and SARS-2 spike pseudotyped virus entry in vitro. bioRxiv [Preprint] (2020) 10.1101/2020.06.20.162701 (Accessed 20 June 2020).
    1. Menegazzi M., et al. , Protective effect of epigallocatechin-3-gallate (EGCG) in diseases with uncontrolled immune activation: Could such a scenario Be helpful to counteract COVID-19? Int. J. Mol. Sci. 21, E5171 (2020).
    1. Chen F., et al. , In vitro susceptibility of 10 clinical isolates of SARS coronavirus to selected antiviral compounds. J. Clin. Virol. 31, 69–75 (2004).
    1. Kulczyński B., Kobus-Cisowska J., Taczanowski M., Kmiecik D., Gramza-Michałowska A., The chemical composition and nutritional value of chia seeds-current state of knowledge. Nutrients 11, 1242 (2019).
    1. Kubiça T. F., Alves S. H., Weiblen R., Lovato L. T., In vitro inhibition of the bovine viral diarrhoea virus by the essential oil of Ocimum basilicum (basil) and monoterpenes. Braz. J. Microbiol. 45, 209–214 (2014).
    1. Birtić S., Dussort P., Pierre F. X., Bily A. C., Roller M., Carnosic acid. Phytochemistry 115, 9–19 (2015).
    1. Kiyohara H., et al. , Patchouli alcohol: In vitro direct anti-influenza virus sesquiterpene in Pogostemon cablin Benth. J. Nat. Med. 66, 55–61 (2012).
    1. Mani J. S., et al. , Natural product-derived phytochemicals as potential agents against coronaviruses: A review. Virus Res. 284, 197989 (2020).
    1. Liao S. F., et al. , Immunization of fucose-containing polysaccharides from Reishi mushroom induces antibodies to tumor-associated Globo H-series epitopes. Proc. Natl. Acad. Sci. U.S.A. 110, 13809–13814 (2013).
    1. V’Kovski P., Kratzel A., Steiner S., Stalder H., Thiel V., Coronavirus biology and replication: Implications for SARS-CoV-2. Nat. Rev. Microbiol., 10.1038/s41579-020-00468-6 (2020).
    1. Sia S. F., et al. , Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature 583, 834–838 (2020).
    1. Imai M., et al. , Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development. Proc. Natl. Acad. Sci. U.S.A. 117, 16587–16595 (2020).
    1. K. H. Dinnon, 3rd, et al. , A mouse-adapted model of SARS-CoV-2 to test COVID-19 countermeasures. Nature 586, 560–566 (2020).
    1. Munster V. J., et al. , Respiratory disease in rhesus macaques inoculated with SARS-CoV-2. Nature 585, 268–272 (2020).
    1. Clausen T. M., et al. , SARS-CoV-2 infection depends on cellular heparan sulfate and ACE2. Cell 183, 1043–1057.e15 (2020).

Source: PubMed

3
購読する