A screen of the NIH Clinical Collection small molecule library identifies potential anti-coronavirus drugs

Jianzhong Cao, J Craig Forrest, Xuming Zhang, Jianzhong Cao, J Craig Forrest, Xuming Zhang

Abstract

With the recent emergence of Middle East Respiratory Syndrome coronavirus in humans and the outbreak of devastating porcine epidemic diarrhea coronavirus in swine, therapeutic intervention is urgently needed. However, anti-coronavirus drugs currently are not available. In an effort to assist rapid development of anti-coronavirus drugs, here we screened the NIH Clinical Collection in cell culture using a luciferase reporter-expressing recombinant murine coronavirus. Of the 727 compounds screened, 84 were found to have a significant anti-coronavirus effect. Further experiments revealed that 51 compounds blocked virus entry while 19 others inhibited viral replication. Additional validation studies with the top 3 inhibitors (hexachlorophene, nitazoxanide and homoharringtonine) demonstrated robust anti-coronavirus activities (a reduction of 6 to 8log10 in virus titer) with an IC50 ranging from 11nM to 1.2μM. Furthermore, homoharringtonine and hexachlorophene exhibited broad antiviral activity against diverse species of human and animal coronaviruses. Since the NIH Clinical Collection consists of compounds that have already been through clinical trials, these small molecule inhibitors have a great potential for rapid development as anti-coronavirus drugs.

Keywords: Coronavirus; NCC library; Screening; Small molecule.

Copyright © 2014 Elsevier B.V. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Effect of candidate drugs on MHV infection at different MOIs. DBT cells were treated with drugs at 10 μM for 1 h and then infected with MHV-2aFLS at MOI of 1 or 0.1 in the presence of the indicated drugs for 8 h. Cells were then lysed for determining luciferase activity. Cells treated with 1% DMSO were served as control. The antiviral effectiveness of the drugs is expressed as percent luciferase activity to the control, which is 100%. Data represent mean of 3 independent treatments and standard deviation of the means.
Fig. 2
Fig. 2
Correlation of inhibition on luciferase reporter expression and virus titer. DBT cells were treated with various drugs as indicated (10 μM) or DMSO (1%) as a control for 1 h and then infected with MHV-2aFLS at MOI of 1 for 8 h. The medium was harvested for determining viral titer (TCID50) (B) and cells were lysed for determining luciferase activity (A). Data indicate the mean of 3 replicates (independent treatments) and standard deviation of the mean. Statistical significance of the inhibitory effect of the drugs on luciferase activity (A) or virus titer (B) as compared to those of DMSO control is indicated by the number of asterisks (∗p < 0.05; ∗∗p < 0.01; ∗∗∗p < 0.001).
Fig. 3
Fig. 3
Inhibitory effect of hexachlorophene on MHV infection. (A) Chemical structure of hexachlorophene. (B) Determination of IC50. DBT cells were treated with hexachlorophene at various concentrations as indicated or 1% DMSO (vehicle control) for 1 h, and were infected with MHV-2aFLS at MOI of 1 in the presence of the drug for 8 h. Cells were then lysed for luciferase assay. Inhibition of MHV infection was expressed as percent reduction in luciferase activity following drug treatment compared to the control, and the IC50 was then calculated as indicated by the solid lines. (C) Inhibition of viral titer. DBT cells were treated with hexachlorophene (10 μM) or DMSO (1%) as a control for 1 h and then infected with MHV-2aFLS at MOI of 1 for 12 h. The medium was harvested for determining viral titer (TCID50). Data indicate the mean of 3 replicates and standard deviation of the mean. (D) Inhibition of viral N protein expression. The experiments were performed identically to (C), except that different concentrations of the drugs were used. Following drug treatment and viral infection, cells were lysed to evaluate viral N protein expression levels by Western blotting. Beta-actin serves as a loading control.
Fig. 4
Fig. 4
Inhibitory effect of nitazoxanide on MHV infection. (A) Chemical structure of nitazoxanide. (B) Determination of IC50. DBT cells were treated with nitazoxanide at various concentrations or 1% DMSO (vehicle control) for 1 h, and were infected with MHV-2aFLS at MOI of 1 in the presence of the drug for 8 h. Cells were then lysed for luciferase assay. Inhibition of drug on MHV infection was expressed as percent reduction on luciferase activity to the control and the IC50 was then calculated as indicated by the solid lines. (C) Inhibition of viral titer. DBT cells were treated with nitazoxanide (10 μM) or DMSO (1%) as a control for 1 h and then infected with MHV-2aFLS at MOI of 1 for 12 h. The medium was harvested for determining viral titer (TCID50). Data indicate the mean of 3 replicates and standard deviation of the mean. (D) Inhibition of viral N protein expression. The experiments were performed identically to (C), except that different concentrations of the drugs were used. Following drug treatment and viral infection, cells were lysed to evaluate viral N protein expression levels by Western blotting. Beta-actin serves as a loading control.
Fig. 5
Fig. 5
Inhibitory effect of homoharringtonine on MHV infection. (A) Chemical structure of homoharringtonine. (B) Determination of IC50. DBT cells were treated with homoharringtonine at various concentrations or 1% DMSO (vehicle control) for 1 h, and were infected with MHV-2aFLS at MOI of 1 in the presence of the drug for 8 h. Cells were then lysed for luciferase assay. Inhibition of drug on MHV infection was expressed as percent reduction on luciferase activity to the control and the IC50 was then calculated as indicated by the solid lines. (C–E) Cells were pretreated with homoharringtonine at various concentrations as indicated or 1% DMSO for 1 h. Cells were then infected with MHV-A59 for 12 h (C and D in DBT cells) or MHV-A59GFP for 16 h (E in 17Cl-1 cells) at MOI of 1 in the presence of the drug. (C) Virus titer in the medium was determined by TCID50. Data represent the mean of 3 replicates and standard deviation of the mean. (D) Viral N protein expression in cell lysates was detected by Western blotting using mAb J.3.3. Beta-actin serves as a loading control. (E) Expression of GFP was directly observed using a fluorescence microscope (Olympus IX-70), and images were captured using a digital camera (Zeiss).
Fig. 6
Fig. 6
Identification of candidate drugs that inhibit MHV infection during viral replication. (A) DBT cells were either treated with drugs (10 μm) as indicated or DMSO (1%) at 1 h before or 3 h after infection with MHV-2aFLS at MOI of 1. The cells were lysed at 8 h p.i. and luciferase activity was measured and expressed as a percentage of DMSO control. Data represent the mean of triplicate experiments and standard deviation from the mean. (B) 17Cl-1 cells were either treated with drugs (10 μm) as indicated or DMSO (1%) at 1 h before or 3 h after infection with MHV-A59GFP at MOI of 1. At 16 h p.i. EGFP expression was observed using a fluorescence microscope (Olympus IX-70), and images were captured using a digital camera (Zeiss). Representative images for 3 h post infection are shown.
Fig. 7
Fig. 7
Identification of candidate drugs that inhibit MHV infection during cell entry. (A) DBT cells were either treated with drugs (10 μM) as indicated or DMSO (1%) at 1 h before or 3 h after infection with MHV-2aFLS at MOI of 1. The cells were lysed at 8 h p.i. and luciferase activity was measured and expressed as a percentage of DMSO control. Data represent the mean of triplicate experiments and standard deviation from the mean. (B) 17Cl-1 cells were either treated with drugs (10 μM) as indicated or DMSO (1%) at 1 h before or 3 h after infection with MHV-A59GFP at MOI of 1. At 16 h p.i. EGFP expression was observed using a fluorescence microscope (Olympus IX-70), and images were captured using a digital camera (Zeiss).
Fig. 8
Fig. 8
Candidate drugs are capable of inhibiting infection with diverse coronaviruses. (A) DBT cells were infected with various MHV strains (MHV-1, MHV-2, and MHV-JHM) in the presence of the indicated drugs for 12 h, and the viral N protein was detected by Western blot using mAb J.3.3. Beta-actin serves as loading control. (B and C) HRT-18 cells were infected with either BCoV-L9 (B) or HECoV-4408 (C) in the presence of the drugs for 36 h, and the viral N protein was detected by IFA using specific mAb#46. The concentration of homoharringtonine is 1 μM and hexachlorophene is 5 μM. The control is 1% DMSO.

References

    1. Al-Tawfiq J.A., Momattin H., Dib J., Memish Z.A. Ribavirin and interferon therapy in patients infected with the Middle East respiratory syndrome coronavirus: an observational study. Int. J. Infect. Dis. 2014;20:42–46.
    1. Andruska N., Mao C., Cherian M., Zhang C., Shapiro D.J. Evaluation of a luciferase-based reporter assay as a screen for inhibitors of estrogen-ERalpha-induced proliferation of breast cancer cells. J. Biomol. Screen. 2012;17:921–932.
    1. Aulner N., Danckaert A., Rouault-Hardoin E., Desrivot J., Helynck O., Commere P.H., Munier-Lehmann H., Spath G.F., Shorte S.L., Milon G., Prina E. High content analysis of primary macrophages hosting proliferating Leishmania amastigotes: application to anti-leishmanial drug discovery. PLoS Negl. Trop. Dis. 2013;7:E2154.
    1. Barlough J.E., Shacklett B.L. Antiviral studies of feline infectious peritonitis virus in vitro. Vet. Rec. 1994;135:177–179.
    1. Boukhvalova M.S., Prince G.A., Blanco J.C. Inactivation of respiratory syncytial virus by zinc finger reactive compounds. Virol. J. 2010;7:20.
    1. Cao J., Zhang X. Comparative in vivo analysis of the nsp15 endoribonuclease of murine, porcine and severe acute respiratory syndrome coronaviruses. Virus Res. 2012;167:247–258.
    1. Caul E.O., Egglestone S.I. Further studies on human enteric coronaviruses. Arch Virol. 1977;54:107–117.
    1. CDC, 2004. SARS Basics Fact Sheet.
    1. Crance J.M., Scaramozzino N., Jouan A., Garin D. Interferon, ribavirin, 6-azauridine and glycyrrhizin: antiviral compounds active against pathogenic flaviviruses. Antiviral Res. 2003;58:73–79.
    1. Das Sarma J., Fu L., Tsai J.C., Weiss S.R., Lavi E. Demyelination determinants map to the spike glycoprotein gene of coronavirus mouse hepatitis virus. J. Virol. 2000;74:9206–9213.
    1. Das Sarma J., Scheen E., Seo S.H., Koval M., Weiss S.R. Enhanced green fluorescent protein expression may be used to monitor murine coronavirus spread in vitro and in the mouse central nervous system. J. Neurovirol. 2002;8:381–391.
    1. De Gans J., Portegies P., Tiessens G., Eeftinck Schattenkerk J.K., Van Boxtel C.J., Van Ketel R.J., Stam J. Itraconazole compared with amphotericin B plus flucytosine in AIDS patients with cryptococcal meningitis. AIDS. 1992;6:185–190.
    1. De Haan C.A., Van G.L., Stoop J.N., Volders H., Rottier P.J. Coronaviruses as vectors: position dependence of foreign gene expression. J. Virol. 2003;77:11312–11323.
    1. De La Hamaide, S., 2014. Deadly pig virus likely to ease in US by year-end. <>.
    1. Drosten C., Gunther S., Preiser W., Van Der Werf S., Brodt H.R., Becker S., Rabenau H., Panning M., Kolesnikova L., Fouchier R.A., Berger A., Burguiere A.M., Cinatl J., Eickmann M., Escriou N., Grywna K., Kramme S., Manuguerra J.C., Muller S., Rickerts V., Sturmer M., Vieth S., Klenk H.D., Osterhaus A.D., Schmitz H., Doerr H.W. Identification of a novel coronavirus in patients with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1967–1976.
    1. Efferth T., Romero M.R., Wolf D.G., Stamminger T., Marin J.J., Marschall M. The antiviral activities of artemisinin and artesunate. Clin. Infect. Dis. 2008;47:804–811.
    1. Falzarano D. Inhibition of novel beta coronavirus replication by a combination of interferon-alpha2b and ribavirin. Sci. Rep. 2013;3:1686.
    1. Fan H. The nucleocapsid protein of coronavirus infectious bronchitis virus: crystal structure of its N-terminal domain and multimerization properties. Structure. 2005;13(12):1859–1868.
    1. Flint M., Mcmullan L.K., Dodd K.A., Bird B.H., Khristova M.L., Nichol S.T., Spiropoulou C.F. Inhibitors of the tick-borne, hemorrhagic fever-associated flaviviruses. Antimicrob. Agents Chemother. 2014;58:3206–3216.
    1. Fukuoka M., Minakuchi M., Kawaguchi A., Nagata K., Kamatari Y.O., Kuwata K. Structure-based discovery of anti-influenza virus a compounds among medicines. Biochim. Biophys. Acta. 2012;1820:90–95.
    1. Gough A.H., Chen N., Shun T.Y., Lezon T.R., Boltz R.C., Reese C.E., Wagner J., Vernetti L.A., Grandis J.R., Lee A.V., Stern A.M., Schurdak M.E., Taylor D.L. Identifying and quantifying heterogeneity in high content analysis: application of heterogeneity indices to drug discovery. PLoS One. 2014;9:E102678.
    1. Hirano N., Fujiwara K., Hino S., Matumoto M. Replication and plaque formation of mouse hepatitis virus (MHV-2) in mouse cell line DBT culture. Arch. Gesamte Virusforsch. 1974;44:298–302.
    1. Hsu J.T., Kuo C.J., Hsieh H.P., Wang Y.C., Huang K.K., Lin C.P., Huang P.F., Chen X., Liang P.H. Evaluation of metal-conjugated compounds as inhibitors of 3CL protease of SARS-CoV. FEBS Lett. 2004;574:116–120.
    1. Hummel M.A., Locuson C.W., Gannett P.M., Rock D.A., Mosher C.M., Rettie A.E., Tracy T.S. CYP2C9 genotype-dependent effects on in vitro drug–drug interactions: switching of benzbromarone effect from inhibition to activation in the CYP2C9.3 variant. Mol. Pharmacol. 2005;68:644–651.
    1. Jayaram H. X-ray structures of the N- and C-terminal domains of a coronavirus nucleocapsid protein: implications for nucleocapsid formation. J. Virol. 2006;80(13):6612–6620.
    1. Keeffe E.B., Rossignol J.F. Treatment of chronic viral hepatitis with nitazoxanide and second generation thiazolides. World J. Gastroenterol. 2009;15:1805–1808.
    1. Kim S.M., Park J.H., Lee K.N., Kim S.K., Ko Y.J., Lee H.S., Cho I.S. Enhanced inhibition of foot-and-mouth disease virus by combinations of porcine interferon-alpha and antiviral agents. Antiviral Res. 2012;96:213–220.
    1. Korba B.E., Montero A.B., Farrar K., Gaye K., Mukerjee S., Ayers M.S., Rossignol J.F. Nitazoxanide, tizoxanide and other thiazolides are potent inhibitors of hepatitis B virus and hepatitis C virus replication. Antiviral Res. 2008;77:56–63.
    1. Ksiazek T.G., Erdman D., Goldsmith C.S., Zaki S.R., Peret T., Emery S., Tong S., Urbani C., Comer J.A., Lim W., Rollin P.E., Dowell S.F., Ling A.E., Humphrey C.D., Shieh W.J., Guarner J., Paddock C.D., Rota P., Fields B., Derisi J., Yang J.Y., Cox N., Hughes J.M., Leduc J.W., Bellini W.J., Anderson L.J. A novel coronavirus associated with severe acute respiratory syndrome. N. Engl. J. Med. 2003;348:1953–1966.
    1. Lim P.L., Lee T.H., Rowe E.K. Middle East respiratory syndrome coronavirus (MERS CoV): update 2013. Curr. Infect. Dis. Rep. 2013;15:295–298.
    1. Marra M.A., Jones S.J., Astell C.R., Holt R.A., Brooks-Wilson A., Butterfield Y.S., Khattra J., Asano J.K., Barber S.A., Chan S.Y., Cloutier A., Coughlin S.M., Freeman D., Girn N., Griffith O.L., Leach S.R., Mayo M., Mcdonald H., Montgomery S.B., Pandoh P.K., Petrescu A.S., Robertson A.G., Schein J.E., Siddiqui A., Smailus D.E., Stott J.M., Yang G.S., Plummer F., Andonov A., Artsob H., Bastien N., Bernard K., Booth T.F., Bowness D., Czub M., Drebot M., Fernando L., Flick R., Garbutt M., Gray M., Grolla A., Jones S., Feldmann H., Meyers A., Kabani A., Li Y., Normand S., Stroher U., Tipples G.A., Tyler S., Vogrig R., Ward D., Watson B., Brunham R.C., Krajden M., Petric M., Skowronski D.M., Upton C., Roper R.L. The genome sequence of the SARS-associated coronavirus. Science. 2003;300:1399–1404.
    1. Mitka M. Deadly MERS coronavirus not yet a global concern. JAMA. 2013;310:569.
    1. Morgenstern B., Michaelis M., Baer P.C., Doerr H.W., Cinatl J., Jr. Ribavirin and interferon-beta synergistically inhibit SARS-associated coronavirus replication in animal and human cell lines. Biochem. Biophys. Res. Commun. 2005;326:905–908.
    1. Morrey J.D., Smee D.F., Sidwell R.W., Tseng C. Identification of active antiviral compounds against a New York isolate of West Nile virus. Antiviral Res. 2002;55:107–116.
    1. Phillips J.J., Chua M.M., Lavi E., Weiss S.R. Pathogenesis of chimeric MHV4/MHV-A59 recombinant viruses: the murine coronavirus spike protein is a major determinant of neurovirulence. J. Virol. 1999;73:7752–7760.
    1. Pyrc K., Bosch B.J., Berkhout B., Jebbink M.F., Dijkman R., Rottier P., Van Der Hoek L. Inhibition of human coronavirus NL63 infection at early stages of the replication cycle. Antimicrob. Agents Chemother. 2006;50:2000–2008.
    1. Rachidi N., Taly J.F., Durieu E., Leclercq O., Aulner N., Prina E., Pescher P., Notredame C., Meijer L., Spath G.F. Pharmacological assessment defines Leishmania donovani casein kinase 1 as a drug target and reveals important functions in parasite viability and intracellular infection. Antimicrob. Agents Chemother. 2014;58:1501–1515.
    1. Reinders M.K., Van Roon E.N., Jansen T.L., Delsing J., Griep E.N., Hoekstra M., Van De Laar M.A., Brouwers J.R. Efficacy and tolerability of urate-lowering drugs in gout: a randomised controlled trial of benzbromarone versus probenecid after failure of allopurinol. Ann. Rheum. Dis. 2009;68:51–56.
    1. Resta S., Luby J.P., Rosenfeld C.R., Siegel J.D. Isolation and propagation of a human enteric coronavirus. Science. 1985;229:978–981.
    1. Romero M.R., Serrano M.A., Efferth T., Alvarez M., Marin J.J. Effect of cantharidin, cephalotaxine and homoharringtonine on “in vitro” models of hepatitis B virus (HBV) and bovine viral diarrhoea virus (BVDV) replication. Planta Med. 2007;73:552–558.
    1. Rossignol J.F., La F.S., Chiappa L., Ciucci A., Santoro M.G. Thiazolides, a new class of anti-influenza molecules targeting viral hemagglutinin at the post-translational level. J. Biol. Chem. 2009;284:29798–29808.
    1. Rota P.A., Oberste M.S., Monroe S.S., Nix W.A., Campagnoli R., Icenogle J.P., Penaranda S., Bankamp B., Maher K., Chen M.H., Tong S., Tamin A., Lowe L., Frace M., Derisi J.L., Chen Q., Wang D., Erdman D.D., Peret T.C., Burns C., Ksiazek T.G., Rollin P.E., Sanchez A., Liffick S., Holloway B., Limor J., Mccaustland K., Olsen-Rasmussen M., Fouchier R., Gunther S., Osterhaus A.D., Drosten C., Pallansch M.A., Anderson L.J., Bellini W.J. Characterization of a novel coronavirus associated with severe acute respiratory syndrome. Science. 2003;300:1394–1399.
    1. Saijo M., Morikawa S., Fukushi S., Mizutani T., Hasegawa H., Nagata N., Iwata N., Kurane I. Inhibitory effect of mizoribine and ribavirin on the replication of severe acute respiratory syndrome (SARS)-associated coronavirus. Antiviral Res. 2005;66:159–163.
    1. Sattar S.A., Raphael R.A., Lochnan H., Springthorpe V.S. Rotavirus inactivation by chemical disinfectants and antiseptics used in hospitals. Can. J. Microbiol. 1983;29:1464–1469.
    1. Seguin S.P., Ireland A.W., Gupta T., Wright C.M., Miyata Y., Wipf P., Pipas J.M., Gestwicki J.E., Brodsky J.L. A screen for modulators of large T antigen’s ATPase activity uncovers novel inhibitors of simian virus 40 and BK virus replication. Antiviral Res. 2012;96:70–81.
    1. Shi Z., Wei J., Deng X., Li S., Qiu Y., Shao D., Li B., Zhang K., Xue F., Wang X., Ma Z. Nitazoxanide inhibits the replication of Japanese encephalitis virus in cultured cells and in a mouse model. Virol. J. 2014;11:10.
    1. Siddiq D.M., Koo H.L., Adachi J.A., Viola G.M. Norovirus gastroenteritis successfully treated with nitazoxanide. J. Infect. 2011;63:394–397.
    1. Sinclair D.S., Fox I.H. The pharmacology of hypouricemic effect of benzbromarone. J. Rheumatol. 1975;2:437–445.
    1. Sorensen M.D., Sorensen B., Gonzalez-Dosal R., Melchjorsen C.J., Weibel J., Wang J., Jun C.W., Huanming Y., Kristensen P. Severe acute respiratory syndrome (SARS): development of diagnostics and antivirals. Ann. N. Y. Acad. Sci. 2006;1067:500–505.
    1. Stevenson G.W., Hoang H., Schwartz K.J., Burrough E.R., Sun D., Madson D., Cooper V.L., Pillatzki A., Gauger P., Schmitt B.J., Koster L.G., Killian M.L., Yoon K.J. Emergence of porcine epidemic diarrhea virus in the United States: clinical signs, lesions, and viral genomic sequences. J. Vet. Diagn. Invest. 2013;25:649–654.
    1. Tebas P., Powderly W.G. Nelfinavir mesylate. Expert Opin. Pharmacother. 2000;1:1429–1440.
    1. Walson J.L., Otieno P.A., Mbuchi M., Richardson B.A., Lohman-Payne B., Macharia S.W., Overbaugh J., Berkley J., Sanders E.J., Chung M.H., John-Stewart G.C. Albendazole treatment of HIV-1 and helminth co-infection: a randomized, double-blind, placebo-controlled trial. AIDS. 2008;22:1601–1609.
    1. Wang L., Byrum B., Zhang Y. New variant of porcine epidemic diarrhea virus, United States, 2014. Emerg. Infect. Dis. 2014;20:917–919.
    1. White A.C., Jr. Nitazoxanide: an important advance in anti-parasitic therapy. Am. J. Trop. Med. Hyg. 2003;68:382–383.
    1. WHO, 2014. WHO MERS-CoV. <>.
    1. Zhang X.D. A pair of new statistical parameters for quality control in RNA interference high-throughput screening assays. Genomics. 2007;89:552–561.
    1. Zhang X.M., Herbst W., Kousoulas K.G., Storz J. Biological and genetic characterization of a hemagglutinating coronavirus isolated from a diarrhoeic child. J. Med. Virol. 1994;44:152–161.
    1. Zhang X.M., Kousoulas K.G., Storz J. Comparison of the nucleotide and deduced amino acid sequences of the S genes specified by virulent and avirulent strains of bovine coronaviruses. Virology. 1991;183:397–404.
    1. Zhu H., Yu D., Zhang X. The spike protein of murine coronavirus regulates viral genome transport from the cell surface to the endoplasmic reticulum during infection. J. Virol. 2009;83:10653–10663.

Source: PubMed

3
Abonner