Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Fluoxazolevir inhibits hepatitis C virus infection in humanized chimeric mice by blocking viral membrane fusion

Abstract

Fluoxazolevir is an aryloxazole-based entry inhibitor of hepatitis C virus (HCV). We show that fluoxazolevir inhibits fusion of HCV with hepatic cells by binding HCV envelope protein 1 to prevent fusion. Nine of ten fluoxazolevir resistance-associated substitutions are in envelope protein 1, and four are in a putative fusion peptide. Pharmacokinetic studies in mice, rats and dogs revealed that fluoxazolevir localizes to the liver. A 4-week intraperitoneal regimen of fluoxazolevir in humanized chimeric mice infected with HCV genotypes 1b, 2a or 3 resulted in a 2-log reduction in viraemia, without evidence of drug resistance. In comparison, daclatasvir, an approved HCV drug, suppressed more than 3 log of viraemia but is associated with the emergence of resistance-associated substitutions in mice. Combination therapy using fluoxazolevir and daclatasvir cleared HCV genotypes 1b and 3 in mice. Fluoxazolevir combined with glecaprevir and pibrentasvir was also effective in clearing multidrug-resistant HCV replication in mice. Fluoxazolevir may be promising as the next generation of combination drug cocktails for HCV treatment.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Fluoxazolevir disrupts HCV membrane fusion.
Fig. 2: Fluoxazolevir-resistant HCV substitutions generated from the in vitro resistance selection assay.
Fig. 3: Dose–response curves of fluoxazolevir against various chimeric HCV genotypes.
Fig. 4: Efficacy of fluoxazolevir in vivo against HCV genotypes 1b, 2a and 3 and multidrug-resistant HCV infection in Alb-uPA/Scid mice.

Similar content being viewed by others

Data availability

The data used to generate the HCV E1 alignment in Fig. 2b and support the findings of this study are available from the Virus Pathogen Resource database (genotypes 1–6). The two genotype 7 sequences are available in the National Center for Biotechnology Information with accession nos. YP_009272536 and ARB18146. The source data for Figs. 1b,d–f, 3 and 4, and Extended Data Figs. 1b, 2, 3b,c and 410 are included in the article. Other data supporting the findings of this study are available from the corresponding author upon request.

References

  1. Blach, S. et al. Global prevalence and genotype distribution of hepatitis C virus infection in 2015: a modelling study. Lancet Gastroenterol. Hepatol. 2, 161–176 (2017).

    Google Scholar 

  2. Jafari, S., Copes, R., Baharlou, S., Etminan, M. & Buxton, J. Tattooing and the risk of transmission of hepatitis C: a systematic review and meta-analysis. Int. J. Infect. Dis. 14, e928–e940 (2010).

    PubMed  Google Scholar 

  3. Blackard, J. T., Shata, M. T., Shire, N. J. & Sherman, K. E. Acute hepatitis C virus infection: a chronic problem. Hepatology 47, 321–331 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Liang, T. J. & Ghany, M. G. Current and future therapies for hepatitis C virus infection. N. Engl. J. Med. 368, 1907–1917 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Liang, T. J. & Ghany, M. G. Therapy of hepatitis C—back to the future. N. Engl. J. Med 370, 2043–2047 (2014).

    PubMed  PubMed Central  Google Scholar 

  6. Ward, J. W. & Hinman, A. R. What is needed to eliminate hepatitis B virus and hepatitis C virus as global health threats. Gastroenterology 156, 297–310 (2019).

    PubMed  Google Scholar 

  7. Zeuzem, S. et al. Sofosbuvir and ribavirin in HCV genotypes 2 and 3. N. Engl. J. Med 370, 1993–2001 (2014).

    PubMed  Google Scholar 

  8. Fourati, S. et al. Frequent antiviral treatment failures in patients infected with hepatitis C virus genotype 4, subtype 4r. Hepatology 69, 513–523 (2019).

    CAS  PubMed  Google Scholar 

  9. Childs, K. et al. Suboptimal SVR rates in African patients with atypical genotype 1 subtypes: implications for global elimination of hepatitis C. J. Hepatol. 71, 1099–1105 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Wei, L. et al. Sofosbuvir–velpatasvir for treatment of chronic hepatitis C virus infection in Asia: a single-arm, open-label, phase 3 trial. Lancet Gastroenterol. Hepatol. 4, 127–134 (2019).

    PubMed  Google Scholar 

  11. Garrison, K. L., German, P., Mogalian, E. & Mathias, A. The drug–drug interaction potential of antiviral agents for the treatment of chronic hepatitis C infection. Drug Metab. Dispos. 46, 1212–1225 (2018).

    CAS  PubMed  Google Scholar 

  12. Voelker, R. The 8-week cure for hepatitis C. JAMA 318, 996 (2017).

    PubMed  Google Scholar 

  13. Pawlotsky, J.-M. Hepatitis C virus resistance to direct-acting antiviral drugs in interferon-free regimens. Gastroenterology 151, 70–86 (2016).

    CAS  PubMed  Google Scholar 

  14. Kurosaki, M. et al. Nation-wide real world study of NS5A resistance‐associated substitutions in patients who failed prior daclatasvir plus asunaprevir: identification of specific pattern that impacts re‐treatment outcome by ledipasvir/sofosbuvir. Hepatology 68, abstr. 200 (2018).

  15. Di Maio, V. C. et al. Frequent NS5A and multiclass resistance in almost all HCV genotypes at DAA failures: what are the chances for second-line regimens? J. Hepatol. 68, 597–600 (2018).

    PubMed  Google Scholar 

  16. Howe, A. et al. A real world resistance profile of virologic failures collected from an international collaboration (SHARED). Hepatology 68, abstr. 204 (2018).

  17. Teegen, E. M., Maurer, M. M., Globke, B., Pratschke, J. & Eurich, D. Liver transplantation for hepatitis-B-associated liver disease—three decades of experience. Transpl. Infect. Dis. 21, e12997 (2019).

    PubMed  Google Scholar 

  18. Hu, Z. et al. Novel cell-based hepatitis C virus infection assay for quantitative high-throughput screening of anti-hepatitis C virus compounds. Antimicrob. Agents Chemother. 58, 995–1004 (2014).

    PubMed  PubMed Central  Google Scholar 

  19. He, S. et al. Development of an aryloxazole class of hepatitis C virus inhibitors targeting the entry stage of the viral replication cycle. J. Med. Chem. 60, 6364–6383 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. He, S. et al. Repurposing of the antihistamine chlorcyclizine and related compounds for treatment of hepatitis C virus infection. Sci. Transl. Med. 7, 282ra249 (2015).

    Google Scholar 

  21. Tscherne, D. M. et al. Time- and temperature-dependent activation of hepatitis C virus for low-pH-triggered entry. J. Virol. 80, 1734–1741 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ashfaq, U. A., Javed, T., Rehman, S., Nawaz, Z. & Riazuddin, S. Lysosomotropic agents as HCV entry inhibitors. Virol. J. 8, 163 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sharma, N. R. et al. Hepatitis C virus is primed by CD81 protein for low pH-dependent fusion. J. Biol. Chem. 286, 30361–30376 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Bush, C. O. et al. A small-molecule inhibitor of hepatitis C virus infectivity. Antimicrob. Agents Chemother. 58, 386–396 (2014).

    PubMed  PubMed Central  Google Scholar 

  25. Zhou, N. et al. Characterization of NS5A polymorphisms and their impact on response rates in patients with HCV genotype 2 treated with daclatasvir-based regimens. J. Antimicrob. Chemother. 71, 3495–3505 (2016).

    CAS  PubMed  Google Scholar 

  26. Gottwein, J. M. et al. Development and characterization of hepatitis C virus genotype 1–7 cell culture systems: role of CD81 and scavenger receptor class B type I and effect of antiviral drugs. Hepatology 49, 364–377 (2009).

    CAS  PubMed  Google Scholar 

  27. Lin, B., He, S., Yim, H. J., Liang, T. J. & Hu, Z. Evaluation of antiviral drug synergy in an infectious HCV system. Antivir. Ther. 21, 595–603 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Bijnsdorp, I. V., Giovannetti, E. & Peters, G. J. in Cancer Cell Culture: Methods and Protocols (ed. Cree, I. A.) 421–434 (Springer Science+Business Media, 2011).

  29. Prichard, M. N. & Shipman, C.Jr. A three-dimensional model to analyze drug-drug interactions. Antivir. Res. 14, 181–205 (1990).

    CAS  PubMed  Google Scholar 

  30. Puoti, M. et al. High SVR12 with 8-week and 12-week glecaprevir/pibrentasvir therapy: an integrated analysis of HCV genotype 1–6 patients without cirrhosis. J. Hepatol. 69, 293–300 (2018).

    CAS  PubMed  Google Scholar 

  31. Ng, T. I. et al. In vitro antiviral activity and resistance profile of the next-generation hepatitis C virus NS3/4A protease inhibitor glecaprevir. Antimicrob. Agents Chemother. 62, e01620-17 (2017).

    PubMed  PubMed Central  Google Scholar 

  32. Ng, T. I. et al. In vitro antiviral activity and resistance profile of the next-generation hepatitis C virus NS5A inhibitor pibrentasvir. Antimicrob. Agents Chemother. 61, e02558-16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Osawa, M. et al. Real-world efficacy of glecaprevir plus pibrentasvir for chronic hepatitis C patient with previous direct-acting antiviral therapy failures. J. Gastroenterol. 54, 291–296 (2019).

    CAS  PubMed  Google Scholar 

  34. Osawa, M. et al. Efficacy of glecaprevir and pibrentasvir treatment for genotype 1b hepatitis C virus drug resistance-associated variants in humanized mice. J. Gen. Virol. 100, 1123–1131 (2019).

    CAS  PubMed  Google Scholar 

  35. Krishnan, P. et al. Pooled resistance analysis in patients with hepatitis C virus genotype 1 to 6 infection treated with glecaprevir-pibrentasvir in phase 2 and 3 clinical trials. Antimicrob. Agents Chemother. 62, e01249-18 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, H. F., Huang, C.-H., Ai, L.-S., Chuang, C.-K. & Chen, S. S. L. Mutagenesis of the fusion peptide-like domain of hepatitis C virus E1 glycoprotein: involvement in cell fusion and virus entry. J. Biomed. Sci. 16, 89 (2009).

    PubMed  PubMed Central  Google Scholar 

  37. Tong, Y., Lavillette, D., Li, Q. & Zhong, J. Role of hepatitis C virus envelope glycoprotein E1 in virus entry and assembly. Front. Immunol. 9, 1411 (2018).

    PubMed  PubMed Central  Google Scholar 

  38. Hu, Z. et al. Chlorcyclizine inhibits viral fusion of hepatitis C virus entry by directly targeting HCV envelope glycoprotein 1. Cell Chem. Biol. 27, 780–792 (2020).

    CAS  PubMed  Google Scholar 

  39. Cocquerel, L., Wychowski, C., Minner, F., Penin, F. & Dubuisson, J. Charged residues in the transmembrane domains of hepatitis C virus glycoproteins play a major role in the processing, subcellular localization, and assembly of these envelope proteins. J. Virol. 74, 3623–3633 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Freedman, H. et al. Computational prediction of the heterodimeric and higher-order structure of gpE1/gpE2 envelope glycoproteins encoded by hepatitis C virus. J. Virol. 91, e02309-16 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Partridge, A. W., Therien, A. G. & Deber, C. M. Missense mutations in transmembrane domains of proteins: phenotypic propensity of polar residues for human disease. Proteins 54, 648–656 (2004).

    CAS  PubMed  Google Scholar 

  42. Kong, L. et al. Hepatitis C virus E2 envelope glycoprotein core structure. Science 342, 1090–1094 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Serre, S. B. N., Krarup, H. B., Bukh, J. & Gottwein, J. M. Identification of alpha interferon-induced envelope mutations of hepatitis C virus in vitro associated with increased viral fitness and interferon resistance. J. Virol. 87, 12776–12793 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Bogomolov, P. et al. Treatment of chronic hepatitis D with the entry inhibitor myrcludex B: first results of a phase Ib/IIa study. J. Hepatol. 65, 490–498 (2016).

    CAS  PubMed  Google Scholar 

  45. Bethea, E. D. et al. Pre-emptive pangenotypic direct acting antiviral therapy in donor HCV-positive to recipient HCV-negative heart transplantation: an open-label study. Lancet Gastroenterol. Hepatol. 4, 771–780 (2019).

    PubMed  Google Scholar 

  46. Woolley, A. E. et al. Heart and lung transplants from HCV-infected donors to uninfected recipients. N. Engl. J. Med. 380, 1606–1617 (2019).

    PubMed  PubMed Central  Google Scholar 

  47. Rolt, A. et al. Preclinical pharmacological development of chlorcyclizine derivatives for the treatment of hepatitis C virus infection. J. Infect. Dis. 217, 1761–1769 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Perin, P. M. et al. Flunarizine prevents hepatitis C virus membrane fusion in a genotype-dependent manner by targeting the potential fusion peptide within E1. Hepatology 63, 49–62 (2016).

    CAS  PubMed  Google Scholar 

  49. Vausselin, T. et al. Identification of a new benzimidazole derivative as an antiviral against hepatitis C virus. J. Virol. 90, 8422–8434 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Harrison, S. C. Viral membrane fusion. Virology 479–480, 498–507 (2015).

    PubMed  Google Scholar 

  51. Rybak, J.-N., Scheurer, S. B., Neri, D. & Elia, G. Purification of biotinylated proteins on streptavidin resin: a protocol for quantitative elution. Proteomics 4, 2296–2299 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Intramural Research Program of the National Institute of Diabetes and Digestive and Kidney Diseases and National Center for Advancing Translational Sciences, NIH, the Molecular Libraries Initiative funding to the University of Kansas Specialized Chemistry Center (grant no. U54HG005031) and the Japan Agency for Medical Research and Development (grant no. JP18fk0210020h0002).

Author information

Authors and Affiliations

Authors

Contributions

T.J.L., C.D.M., Z.H. and K.J.F. conceptualized and designed the study. C.D.M., M.I., D.C.T., A.R., X.X., A.Q.W., D.L., T.U., M.O., Y.T., K.L., X.H., S.B.P., N.C., P.H.I., A.E.D., N.S., J.J.M., Z.H., K.C. and K.J.F. performed, analysed and contributed to all the experiments. C.D.M., Z.H. and T.J.L. wrote the manuscript. All other authors reviewed and contributed to the manuscript.

Corresponding author

Correspondence to Tsanyang Jake Liang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Synthesis, efficacy, and photolysis of the fluoxazolevir-diazirine-biotin probe.

a, The general synthetic scheme of the fluoxazolevir-diazirine-biotin (fluoxazolevir-DB) probe is shown. Each intermediate was confirmed with 1H NMR and LCMS. See supplemental document for more information on each synthetic step. b, Fluoxazolevir-DB probe retains anti-HCV activity in vitro and shows inhibition against HCV infection in a dose-dependent manner. Data are presented as mean values ± SEM of 6 biologically independent replicates. c, The degradation of fluoxazolevir-DB via UV irradiation is shown. d, The fluoxazolevir-DB was exposed to UV irradiation with a 100 W mercury lamp with a 365 nm bypass filter. Disappearance of fluoxazolevir-DB was measured over time via LCMS and underwent a complete conversion to the carbene insertion product within 10 min. All results are representative of three independent experiments.

Source data

Extended Data Fig. 2 Dose-response curves of fluoxazolevir against amplified HCV from the in vitro drug resistance selection assay.

Among the 8 serial passages with potential RAS-containing HCV generated from the drug resistance selection assay (Fig. 2a), the viruses in the following wells (and their identified mutations) showed moderate resistance with EC50 values increasing by at least two-fold comparing to the wild-type control: A1 (F291L, V414A), B1 (I374T), C1 (D382E, T395A, M405V, P616A), E1 (F291V), G1 (A274S) and H1 (M267V, V284A). The same viruses were tested against sofosbuvir as a control and were equally sensitive to sofosbuvir as the wild type virus. Data are presented as mean values ± SEM of 3 biologically independent replicates. All results are representative of three independent experiments.

Source data

Extended Data Fig. 3 Viral fitness of the generated RAS-containing HCV.

a, The viral fitness assay scheme is shown here. Huh7.5.1 cells were electroporated with the RNA of each HCV RAS-RLuc construct. b, The first part of the assay assesses the replication capacity for each RAS-containing HCV. Luminescence was measured 4 h and 3 days after electroporation and the readings obtained 4 h after electroporation was used as background. c, The second part of the assay assesses infectivity of each RAS. Viral medium harvested 3 days after electroporation from part b was used to reinfect 104 naïve Huh7.5.1 cells in a 96-well plate. Luminescence was measured 48 h after reinfection and all measurements were normalized to HCV-WT. Data are presented as mean values ± SEM of 3 biologically independent replicates. All results are representative of three independent experiments.

Extended Data Fig. 4 Dose-response curves of fluoxazolevir against HCV mutants with putative RASs in core, E1 and E2 regions.

Huh7.5.1 cells in 96-well plates were infected with wild-type HCV-RLuc (GT 2a) and HCV-RLuc mutants with various putative RASs (R9G, V140L, M267V, A274S, V284A, F291L, F291V, I374T, D382E, T395A, M405V, V414A and P616A) in the presence of various fluoxazolevir concentrations as indicated. Cells were harvested 48 h after infection and luminescence assessed via the luciferase assay. The EC50 values for wild-type HCV-RLuc (black circles) and the HCV mutants (red squares) were calculated with Prism 7. Data are presented as mean values ± SEM of 8 biologically independent replicates. All results are representative of three independent experiments.

Source data

Extended Data Fig. 5 Cytotoxicity of fluoxazolevir against primary human hepatocytes, HepG2 cells, MT-4 cells and peripheral blood mononuclear cells.

Cells were treated with fluoxazolevir for 3 days and processed for the ATPlite cytotoxicity assay. CC50 values were calculated with the software, Prism 7. Data are presented as mean values ± SEM of 3 biologically independent replicates. All results are representative of three independent experiments.

Source data

Extended Data Fig. 6 Pharmacokinetics of fluoxazolevir.

Pharmacokinetic studies of fluoxazolevir were performed in (a) male CD-1 mouse, (b) male SD rat and (c) male beagle dog models (n = 3 animals). The concentration profiles of fluoxazolevir were measured after either a single PO dose of 10 mg/kg or a single IV dose of 3 mg/kg. Compound concentrations were measured by UPLC-MS/MS. d, Serum alanine aminotransferase (ALT) levels were measured in each animal model to assess the potential toxicity of fluoxazolevir in vivo. For CD-1 mice and SD rats, ALTs from the 10 mg/kg PO groups were shown, and for beagle dogs, the 3 mg/kg IV group was shown. Data are presented as mean values ± standard deviations.

Source data

Extended Data Fig. 7 Tissue distribution of fluoxazolevir after PO administration in rodents.

1The plasma and tissue concentrations of fluoxazolevir were measured after a single PO dose of fluoxazolevir. Thirty-nine mice and fifteen rats (n = 3/time point) for tissue collection. 2 AUC0-∞: area under the curve from zero to infinity; t1/2: half-life; Tmax: time to reach the maximal concentration; Cmax: maximal concentration after PO administration.

Source data

Extended Data Fig. 8 Maximal tolerable dose of fluoxazolevir in mice.

The study was performed by Pharmaron Inc. (Beijing, PR China). Single doses of fluoxazolevir (50 mg/kg, 100 mg/kg, 500 mg/kg and 1000 mg/kg) were administered via oral gavage to CD-1 mice (n = 3 mice per group) and observed for 3 days. Body weights of all animals were recorded daily. All study animals were monitored behavior such as respite, food and water consumption (by cage side checking), circling, eye/hair matting and any other abnormal effect. Any mortality and/or abnormal clinical signs were recorded. All animals were sacrificed for necropsy on day 3. Data are presented as mean values ± SEM.

Source data

Extended Data Fig. 9 Lack of toxicity of fluoxazolevir monotherapy in genotypes 1b, 2a and 3-infected Alb-uPA/Scid mice.

The body weights of the humanized Alb-uPA/Scid mice infected with HCV genotypes (a) 1b (n = 2-4 mice), (b) 2a (n = 3-4 mice) and (c) 3 (n = 3 mice) were monitored during and after fluoxazolevir treatment as described in Fig. 4a, b, Supplementary Figure 35. All mice in each group were weighed regularly for evidence of toxicity.

Source data

Extended Data Fig. 10 HCV RNA and serum human albumin levels of mice infected with multidrug-resistant HCV.

Humanized Alb-uPA/Scid mice were infected with the multidrug-resistant HCV strain and were either untreated (n = 4 mice) or treated with fluoxazolevir (n = 5 mice), GLE/PIB (n = 4 mice) or combination (n = 5 mice). Serum HCV RNA and human serum albumin levels were monitored weekly. a, Serum HCV RNA levels of untreated humanized Alb-uPA/Scid mice showed steady levels during follow-up. Time 0 is comparable to the time of initiation of treatment in (b). Mouse serum samples at the end of the 20 weeks were sequenced and the same NS3 and NS5a mutations as the inoculum virus were identified. b, Human serum albumin levels of untreated mice and mice treated with fluoxazolevir (5 mg/kg), glecaprevir (60 mg/kg) and pibrentasvir (24 mg/kg). Weekly serum levels of human albumin of individual mice were plotted. Weekly HCV RNA measurements of individual mice for each time point are shown in Fig. 4c. Serum human albumin graphs that end before the 10 weeks are due to death of the mice.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–6, Table 1 and Materials and Methods.

Reporting Summary

Peer Review Information

Source data

Source Data Fig. 1b,d

Statistical source data for Fig. 1b,d.

Source Data Fig. 1e,f

Full-length, unprocessed western blot for Fig. 1e,f.

Source Data Fig. 3

Statistical source data for Fig. 3.

Source Data Fig. 4a

Statistical source data for Fig. 4a.

Source Data Fig. 4b

Statistical source data for Fig. 4b.

Source Data Fig. 4c

Statistical source data for Fig. 4c.

Source Data Fig. 4d

Statistical source data for Fig. 4d.

Source Data Extended Data Fig. 1b

Statistical source data for Extended Data Fig. 1b.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3b,c

Statistical source data for Extended Data Fig. 3b,c.

Source Data Extended Data Fig. 4

Statistical source data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical source data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical source data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical source data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical source data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical source data for Extended Data Fig. 9.

Source Data Extended Data Fig. 10

Statistical source data for Extended Data Fig. 10.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ma, C.D., Imamura, M., Talley, D.C. et al. Fluoxazolevir inhibits hepatitis C virus infection in humanized chimeric mice by blocking viral membrane fusion. Nat Microbiol 5, 1532–1541 (2020). https://doi.org/10.1038/s41564-020-0781-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-0781-2

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research