Inhibition of avian-origin influenza A(H7N9) virus by the novel cap-dependent endonuclease inhibitor baloxavir marboxil

Keiichi Taniguchi, Yoshinori Ando, Haruaki Nobori, Shinsuke Toba, Takeshi Noshi, Masanori Kobayashi, Makoto Kawai, Ryu Yoshida, Akihiko Sato, Takao Shishido, Akira Naito, Keita Matsuno, Masatoshi Okamatsu, Yoshihiro Sakoda, Hiroshi Kida, Keiichi Taniguchi, Yoshinori Ando, Haruaki Nobori, Shinsuke Toba, Takeshi Noshi, Masanori Kobayashi, Makoto Kawai, Ryu Yoshida, Akihiko Sato, Takao Shishido, Akira Naito, Keita Matsuno, Masatoshi Okamatsu, Yoshihiro Sakoda, Hiroshi Kida

Abstract

Human infections with avian-origin influenza A(H7N9) virus represent a serious threat to global health; however, treatment options are limited. Here, we show the inhibitory effects of baloxavir acid (BXA) and its prodrug baloxavir marboxil (BXM), a first-in-class cap-dependent endonuclease inhibitor, against A(H7N9), in vitro and in vivo. In cell culture, BXA at four nanomolar concentration achieved a 1.5-2.8 log reduction in virus titers of A(H7N9), including the NA-R292K mutant virus and highly pathogenic avian influenza viruses, whereas NA inhibitors or favipiravir required approximately 20-fold or higher concentrations to achieve the same levels of reduction. A(H7N9)-specific amino acid polymorphism at position 37, implicated in BXA binding to the PA endonuclease domain, did not impact on BXA susceptibility. In mice, oral administration of BXM at 5 and 50 mg/kg twice a day for 5 days completely protected from a lethal A/Anhui/1/2013 (H7N9) challenge, and reduced virus titers more than 2-3 log in the lungs. Furthermore, the potent therapeutic effects of BXM in mice were still observed when a higher virus dose was administered or treatment was delayed up to 48 hours post infection. These findings support further investigation of BXM for A(H7N9) treatment in humans.

Conflict of interest statement

The authors K.T., Y.A., H.N., S.T., T.N., M. Kawai., R.Y., A.S., T.S. and A.N. are employees of Shionogi & Co., Ltd. The authors K.M., M.O., Y.S. and H.K. were provided financial support from Shionogi & Co., Ltd. for the studies performed in the manuscript. The author M. Kobayashi. was an employee of Shionogi & Co., Ltd., and now declares no potential conflict of interest. All works reported here were financially supported by Shionogi & Co., Ltd..

Figures

Figure 1
Figure 1
Therapeutic effects of BXM on survival and weight loss in a lethal infection model of mice infected with a low dose of the A(H7N9) virus. Mice were intranasally inoculated with 4.0 × 105 TCID50/mouse (10.4 MLD50) of A/Anhui/1/2013 (H7N9) viruses, and treatment was started immediately after virus inoculation (n = 10/group). Survival time (a) and body weight loss (b) were monitored through a 28-day period after the infection. bid (bis in die): twice a day. The log-rank test was applied for comparison of the survival time between each group (*p < 0.05, **p < 0.001 compared to vehicle, †p < 0.01 compared to OSP at 5 mg/kg twice a day, §p < 0.05 compared to OSP at 50 mg/kg twice a day). Dunnett’s multiple-comparison method was applied for statistical analysis of body weight changes (*p < 0.01, **p < 0.001 compared to vehicle, †p < 0.001 compared to OSP at 5 mg/kg twice a day, §p < 0.001 compared to OSP at 50 mg/kg twice a day). The body weights at 5 dpi were calculated from nine mice in vehicle-treated group because one of the ten mice showed more than 30% reduction and was euthanized.
Figure 2
Figure 2
Inhibitory effect of BXM on virus titers in the lungs of mice infected with a low dose of the A(H7N9) virus. Mice were inoculated with 4.0 × 105 TCID50/mouse (10.4 MLD50) of A/Anhui/1/2013 (H7N9) virus and treatment was started immediately after virus inoculation (n = 5/group). The virus titers (TCID50) in lungs of mice at 1, 3 and 5 dpi were measured in MDCK cells. The lower limit of quantification of the virus titer is indicated by a dotted line (1.5 Log10 TCID50/mL). Dunnett’s multiple-comparison method was conducted for statistical comparison (*p < 0.05, **p < 0.001 compared to vehicle, †p < 0.05, ††p < 0.01, †††p < 0.001 compared to OSP at 5 mg/kg twice a day, §p < 0.01, §§p < 0.001 compared to OSP at 50 mg/kg twice a day).
Figure 3
Figure 3
Suppressive effects of BXM on proinflammatory cytokine and chemokine production in the lungs of mice infected with a low dose of the A(H7N9) virus. Mice were intranasally infected with 4.0 × 105 TCID50/mouse (10.4 MLD50) of A/Anhui/1/2013 (H7N9) virus, and treatment was started immediately after virus inoculation (n = 5/group). The proinflammatory cytokines and chemokines, IL-6, MCP-1, MIP-1α and IFN-γ, in the lungs at 1, 3 and 5 dpi were quantified. Dunnett’s multiple-comparison method was employed for the comparison (*p < 0.05, **p < 0.01, ***p < 0.001 compared to vehicle).
Figure 4
Figure 4
Effects of BXM on survival, weight loss and virus titers in the lungs in mice infected with a high dose of the A(H7N9) virus. Mice intranasally infected with 1.2 × 106 TCID50/mouse (31.1 MLD50) of A/Anhui/1/2013 (H7N9) virus were administrated with treatment started immediately after virus inoculation, and survival time (a) and body weight loss (b) were analyzed. The log-rank test was applied for survival time (*p < 0.001 compared to vehicle, †p < 0.001 compared to OSP at 5 mg/kg twice a day, §p < 0.001 compared to OSP at 50 mg/kg twice a day). Dunnett’s multiple-comparison method was employed for body weight changes (*p < 0.001 compared to vehicle, †p < 0.001 compared to OSP at 5 mg/kg twice a day, §p < 0.001 compared to OSP at 50 mg/kg twice a day). Body weights at 4 and 5 dpi were calculated from nine mice because one of the ten mice in vehicle-treated group showed more than 30% reduction and was euthanized. (c) The virus titers (TCID50) in the lungs of mice at 1 and 5 dpi were measured. The lower limit of quantification of the virus titer is indicated by a dotted line (1.5 Log10 TCID50/mL). Dunnett’s multiple-comparison method was applied for the comparison (*p < 0.05, **p < 0.001 compared to vehicle, †p < 0.05, ††p < 0.01, †††p < 0.001 compared to OSP at 5 mg/kg twice a day, §p < 0.01, §§p < 0.001 compared to OSP at 50 mg/kg twice a day).
Figure 5
Figure 5
Effects of delayed treatment of BXM on a low dose of A(H7N9) infection. Mice were intranasally inoculated with 4.0 × 105 TCID50/mouse (10.4 MLD50) of A/Anhui/1/2013 (H7N9) viruses, and BXM treatment was started at (a) immediately, (b) 24 or (c) 48 hours after virus inoculation (n = 5/group). Vehicle or OSP treatment was started immediately after virus inoculation (n = 5/group). Survival time and body weight loss were monitored through a 21-day period after the infection. The shaded area represents the treatment period. The log-rank test was applied for comparison of the survival time between each group (*p < 0.01 compared to vehicle, †p < 0.01 compared to OSP at 5 mg/kg twice a day).

References

    1. Taubenberger JK, Morens DM. Influenza: the once and future pandemic. Public Health Rep. 2010;125:16–26. doi: 10.1177/00333549101250S305.
    1. Su S, et al. Epidemiology, Evolution, and Pathogenesis of H7N9 Influenza Viruses in Five Epidemic Waves since 2013 in China. Trends Microbiol. 2017;25:713–728. doi: 10.1016/j.tim.2017.06.008.
    1. Gao R, et al. Human infection with a novel avian-origin influenza A (H7N9) virus. N. Engl. J. Med. 2013;368:1888–97. doi: 10.1056/NEJMoa1304459.
    1. Zaraket H, et al. Mammalian adaptation of influenza A(H7N9) virus is limited by a narrow genetic bottleneck. Nat. Commun. 2015;6:1–10. doi: 10.1038/ncomms7553.
    1. Watanabe T, et al. Characterization of H7N9 influenza A viruses isolated from humans. Nature. 2013;501:551–555. doi: 10.1038/nature12392.
    1. Chan LLY, et al. Evaluation of the human adaptation of influenza A/H7N9 virus in PB2 protein using human and swine respiratory tract explant cultures. Sci. Rep. 2016;6:1–11. doi: 10.1038/s41598-016-0001-8.
    1. de Vries RP, et al. Three mutations switch H7N9 influenza to human-type receptor specificity. PLoS Pathog. 2017;13:1–16.
    1. Iuliano AD, et al. Increase in Human Infections with Avian Influenza A(H7N9) Virus During the Fifth Epidemic - China, October 2016-February 2017. MMWR. Morb. Mortal. Wkly. Rep. 2017;66:254–255. doi: 10.15585/mmwr.mm6609e2.
    1. Liu J, et al. H7N9: A low pathogenic avian influenza A virus infecting humans. Curr. Opin. Virol. 2014;5:91–97. doi: 10.1016/j.coviro.2014.03.001.
    1. Zhou, L. et al. Preliminary Epidemiology of Human Infections with Highly Pathogenic Avian Influenza A(H7N9) Virus, China, 2017. Emerg. Infect. Dis. J. 23 (2017).
    1. Qi, W. et al. Emergence and adaptation of a novel highly pathogenic H7N9 influenza virus in birds and humans from a 2013-human-infecting low pathogenic ancestor. J. Virol. JVI.00921-17, 10.1128/JVI.00921-17 (2017).
    1. Food and Agriculture Organization of the United Nations. H7N9 situation update. Available at: . (Accessed: 3rd October 2018) (2018).
    1. Centers for Disease Control and Prevention. Interim Guidance on the Use of Antiviral Medications for Treatment of Human Infections with Novel Influenza A Viruses Associated with Severe Human Disease. Available at, . (Accessed: 27th July 2018) (2016).
    1. Farooqui A, et al. Assessment of antiviral properties of peramivir against H7N9 avian influenza virus in an experimental mouse model. Antimicrob. Agents Chemother. 2015;59:7255–7264. doi: 10.1128/AAC.01885-15.
    1. Hu Y, et al. Association between adverse clinical outcome in human disease caused by novel influenza A H7N9 virus and sustained viral shedding and emergence of antiviral resistance. Lancet. 2013;381:2273–2279. doi: 10.1016/S0140-6736(13)61125-3.
    1. Treanor JJ, et al. Efficacy and safety of the oral neuraminidase inhibitor oseltamivir in treating acute influenza: a randomized controlled trial. US Oral Neuraminidase Study Group. JAMA. 2000;283:1016–24. doi: 10.1001/jama.283.8.1016.
    1. Lee N, et al. A prospective intervention study on higherdose oseltamivir treatment in adults hospitalized with influenza A and B infections. Clin. Infect. Dis. 2013;57:1511–1519. doi: 10.1093/cid/cit597.
    1. Kondo H, et al. Influenza virus shedding in laninamivir-treated children upon returning to school. Tohoku J. Exp. Med. 2016;238:113–121. doi: 10.1620/tjem.238.113.
    1. Zhang F, et al. Human infections with recently-emerging highly pathogenic H7N9 avian influenza virus in China. J. Infect. 2017;75:71–75. doi: 10.1016/j.jinf.2017.04.001.
    1. Takashita E, et al. Antiviral susceptibility of influenza viruses isolated from patients pre- and post-administration of favipiravir. Antiviral Res. 2016;132:170–177. doi: 10.1016/j.antiviral.2016.06.007.
    1. de Jong MD, et al. Fatal outcome of human influenza A (H5N1) is associated with high viral load and hypercytokinemia. Nat. Med. 2006;12:1203–1207. doi: 10.1038/nm1477.
    1. McKimm-Breschkin JL, et al. Mutations in a conserved residue in the influenza virus neuraminidase active site decreases sensitivity to Neu5Ac2en-derived inhibitors. J. Virol. 1998;72:2456–62.
    1. Gaymard A, et al. Impact on antiviral resistance of E119V, I222L and R292K substitutions in influenza A viruses bearing a group 2 neuraminidase (N2, N3, N6, N7 and N9) J. Antimicrob. Chemother. 2016;71:3036–3045. doi: 10.1093/jac/dkw275.
    1. Kageyama T, et al. Genetic analysis of novel avian A(H7N9) influenza viruses isolated from patients in China, February to April 2013. Euro Surveill. Bull. Eur. sur les Mal. Transm.=Eur. Commun. Dis. Bull. 2013;18:20453.
    1. Marjuki H, et al. Characterization of drug-resistant influenza a(H7N9) variants isolated from an oseltamivir-treated patient in Taiwan. J. Infect. Dis. 2015;211:249–257. doi: 10.1093/infdis/jiu447.
    1. Dias A, et al. The cap-snatching endonuclease of influenza virus polymerase resides in the PA subunit. Nature. 2009;458:914–918. doi: 10.1038/nature07745.
    1. Stevaert A, Naesens L. The Influenza Virus Polymerase Complex: An Update on Its Structure, Functions, and Significance for Antiviral Drug Design. Med. Res. Rev. 2016;36:1127–1173. doi: 10.1002/med.21401.
    1. Stevaert A, et al. Mutational Analysis of the Binding Pockets of the Diketo Acid Inhibitor L-742,001 in the Influenza Virus PA Endonuclease. J. Virol. 2013;87:10524–10538. doi: 10.1128/JVI.00832-13.
    1. Tomassini J, et al. Inhibition of Cap (m7GpppXm) -Dependent Endonuclease of Influenza Virus by 4-Substituted 2, 4-Dioxobutanoic Acid Compounds. Antimicrob. Agents Chemother. 1994;38:2827–2837. doi: 10.1128/AAC.38.12.2827.
    1. Jones JC, et al. A novel endonuclease inhibitor exhibits broad-spectrum anti-influenza virus activity In Vitro. Antimicrob. Agents Chemother. 2016;60:5504–5514. doi: 10.1128/AAC.00888-16.
    1. McKimm-Breschkin JL, et al. Prevention and treatment of respiratory viral infections: Presentations on antivirals, traditional therapies and host-directed interventions at the 5th ISIRV Antiviral Group conference. Antiviral Res. 2018;149:118–142. doi: 10.1016/j.antiviral.2017.11.013.
    1. Omoto, S. et al. Characterization of influenza virus variants induced by treatment with the endonuclease inhibitor baloxavir marboxil. Sci. Rep. 1–15, 10.1038/s41598-018-27890-4 (2018).
    1. Hayden FG, et al. Baloxavir Marboxil for Uncomplicated Influenza in Adults and Adolescents. N. Engl. J. Med. 2018;379:913–923. doi: 10.1056/NEJMoa1716197.
    1. Uehara, T. et al. S-033188, a Small Molecule Inhibitor of Cap-dependent Endonuclease of Influenza A and B Virus, Leads to Rapid and Profound Viral Load Reduction. In OPTIONS IX. (2016).
    1. Noshi, T. et al. In vitro characterization of baloxavir acid, a first-in-class cap-dependent endonuclease inhibitor of the influenza virus polymerase PA subunit. Antiviral Res., 10.1016/j.antiviral.2018.10.008 (2018).
    1. Yamayoshi, S. et al. Virulence-Affecting Amino Acid Changes in the PA Protein of H7N9 Influenza A Viruses. J. Virol., 10.1128/JVI.03155-13 (2014).
    1. Hu, M. et al. Amino acid substitutions V63I or A37S/I61T/V63I/V100A in the PA N-terminal domain increase the virulence of H7N7 influenza Avirus. Sci. Rep., 10.1038/srep37800 (2016).
    1. Jones, J. C. et al. Identification of the I38T PA substitution as a resistance marker for next-generation influenza virus endonuclease inhibitors. Mbio, 10.1128/mBio.00430-18 (2018).
    1. Ward, P., Small, I., Smith, J., Suter, P. & Dutkowski, R. Oseltamivir (Tamiflu®) and its potential for use in the event of an influenza pandemic. J. Antimicrob. Chemother. 55 (2005).
    1. Chi Y, et al. Cytokine and chemokine levels in patients infected with the novel avian influenza a (H7N9) virus in China. J. Infect. Dis. 2013;208:1962–1967. doi: 10.1093/infdis/jit440.
    1. Zhou J, et al. Biological features of novel avian influenza A (H7N9) virus. Nature. 2013;499:500–3. doi: 10.1038/nature12379.
    1. Lee ACY, et al. Avian influenza virus a H7N9 infects multiple mononuclear cell types in peripheral blood and induces dysregulated cytokine responses and apoptosis in infected monocytes. J. Gen. Virol. 2017;98:922–934. doi: 10.1099/jgv.0.000691.
    1. Wang XF, et al. Clinical features of three avian influenza H7N9 virus-infected patients in Shanghai. Clin. Respir. J. 2014;8:410–416. doi: 10.1111/crj.12087.
    1. Baranovich T, et al. The neuraminidase inhibitor oseltamivir is effective against A/Anhui/1/2013 (H7N9) influenza virus in a mouse model of acute respiratory distress syndrome. J. Infect. Dis. 2014;209:1343–1353. doi: 10.1093/infdis/jit554.
    1. Shibata A, et al. Repeated detection of H7N9 avian influenza viruses in raw poultry meat illegally brought to Japan by international flight passengers. Virology. 2018;524:10–17. doi: 10.1016/j.virol.2018.08.001.
    1. Zhang, X. et al. Drug susceptibility profile and pathogenicity of H7N9 influenza virus (Anhui1 lineage) with R292K substitution. Emerg. Microbes Infect., 10.1038/emi.2014.80 (2014).
    1. Hay, A. J. & Hayden, F. G. Oseltamivir resistance during treatment of H7N9 infection. The Lancet, 10.1016/S0140-6736(13)61209-X (2013).
    1. Hai, R. et al. Influenza A(H7N9) virus gains neuraminidase inhibitor resistance without loss of in vivo virulence or transmissibility. Nat. Commun., 10.1038/ncomms3854 (2013).
    1. Noshi, T. et al. Pharmacokinetic and Pharmacodynamic Analysis of S-033188/S-033447, a Novel Inhibitor of Influenza Virus Cap-dependent Endonuclease, in Mice Infected with Influenza A Virus. In ECCMID 2017. (2017).
    1. Fukao, K. et al. Combination treatment with the cap-dependent endonuclease inhibitor baloxavir marboxil and a neuraminidase inhibitor in a mouse model of influenza A virus infection. J. Antimicrob. Chemother. 1–9, 10.1093/jac/dky462 (2018).
    1. Koshimichi, H. et al. Safety, Tolerability, and Pharmacokinetics of the Novel Anti-influenza Agent Baloxavir Marboxil in Healthy Adults: Phase I Study Findings. Clinical Drug Investigation, 10.1007/s40261-018-0710-9 (2018).
    1. Tsang, T. K. et al. Individual correlates of infectivity of influenza A virus infections in households. PLoS One11 (2016).
    1. Byrn RA, et al. Preclinical activity of VX-787, a first-in-class, orally bioavailable inhibitor of the influenza virus polymerase PB2 subunit. Antimicrob. Agents Chemother. 2015;59:1569–1582. doi: 10.1128/AAC.04623-14.
    1. Govorkova EA, et al. Efficacy of oseltamivir therapy in ferrets inoculated with different clades of H5N1 influenza virus. Antimicrob. Agents Chemother. 2007;51:1414–1424. doi: 10.1128/AAC.01312-06.
    1. Bi Y, et al. A new reassortment of influenza A (H7N9) virus causing human infection in Beijing, 2014. Sci. Rep. 2016;6:26624. doi: 10.1038/srep26624.
    1. Wang Z, et al. Early hypercytokinemia is associated with interferon-induced transmembrane protein-3 dysfunction and predictive of fatal H7N9 infection. Proc. Natl. Acad. Sci. USA. 2014;111:769–774. doi: 10.1073/pnas.1321748111.
    1. Ka C, et al. Pathogenicity of the Novel A/H7N9 Influenza Virus in Mice. MBio. 2013;4:1–10. doi: 10.3391/mbi.2013.4.1.01.
    1. Kobayashi M, et al. Therapeutic efficacy of peramivir against H5N1 highly pathogenic avian influenza viruses harboring the neuraminidase H275Y mutation. Antiviral Res. 2017;139:41–48. doi: 10.1016/j.antiviral.2016.12.011.
    1. Guo J, et al. The Serum Profile of Hypercytokinemia Factors Identified in H7N9-Infected Patients can Predict Fatal Outcomes. Sci. Rep. 2015;5:10942. doi: 10.1038/srep10942.
    1. Woo, P. C. Y. et al. Cytokine Profiles Induced by the Novel Swine-Origin Influenza A/H1N1 Virus: Implications for Treatment Strategies. Cytokine201 (2010).
    1. Baumgarth N, Kelso A. In vivo blockade of gamma interferon affects the influenza virus-induced humoral and the local cellular immune response in lung tissue. J. Virol. 1996;70:4411–4418.
    1. Hoffmann E, Neumann G, Kawaoka Y, Hobom G, Webster RGA. DNA transfection system for generation of influenza A virus from eight plasmids.pdf. 2000;97:6108–6113.
    1. Kumar, S., Stecher, G. & Tamura, K. MEGA7: Molecular Evolutionary Genetics Analysis Version 7.0 for Bigger Datasets. Mol. Biol. Evol., 10.1093/molbev/msw054 (2016).
    1. Chu, D. H. et al. Potency of an inactivated influenza vaccine prepared from A/duck/Mongolia/119/2008 (H7N9) against the challenge with A/Anhui/1/2013 (H7N9). Vaccine, 10.1016/j.vaccine.2014.04.060 (2014).

Source: PubMed

3
Tilaa