Effective oral favipiravir (T-705) therapy initiated after the onset of clinical disease in a model of arenavirus hemorrhagic Fever

Michelle Mendenhall, Andrew Russell, Donald F Smee, Jeffery O Hall, Ramona Skirpstunas, Yousuke Furuta, Brian B Gowen, Michelle Mendenhall, Andrew Russell, Donald F Smee, Jeffery O Hall, Ramona Skirpstunas, Yousuke Furuta, Brian B Gowen

Abstract

Background: Lassa and Junín viruses are the most prominent members of the Arenaviridae family of viruses that cause viral hemorrhagic fever syndromes Lassa fever and Argentine hemorrhagic fever, respectively. At present, ribavirin is the only antiviral drug indicated for use in treatment of these diseases, but because of its limited efficacy in advanced cases of disease and its toxicity, safer and more effective antivirals are needed.

Methodology/principal findings: Here, we used a model of acute arenaviral infection in outbred guinea pigs based on challenge with an adapted strain of Pichindé virus (PICV) to further preclinical development of T-705 (Favipiravir), a promising broad-spectrum inhibitor of RNA virus infections. The guinea pig-adapted passage 19 PICV was uniformly lethal with an LD(50) of ∼5 plaque-forming units and disease was associated with fever, weight loss, thrombocytopenia, coagulation defects, increases in serum aspartate aminotransferase (AST) concentrations, and pantropic viral infection. Favipiravir (300 mg/kg/day, twice daily orally for 14 days) was highly effective, as all animals recovered fully from PICV-induced disease even when therapy was initiated one week after virus challenge when animals were already significantly ill with marked fevers and thrombocytopenia. Antiviral activity and reduced disease severity was evidenced by dramatic reductions in peak serum virus titers and AST concentrations in favipiravir-treated animals. Moreover, a sharp decrease in body temperature was observed shortly after the start of treatment. Oral ribavirin was also evaluated, and although effective, the slower rate of recovery may be a sign of the drug's known toxicity.

Conclusions/significance: Our findings support further development of favipiravir for the treatment of severe arenaviral infections. The optimization of the experimental favipiravir treatment regimen in the PICV guinea pig model will inform critical future studies in the same species based on challenge with highly pathogenic arenaviruses such as Lassa and Junín.

Conflict of interest statement

YF is employed by the Toyama Chemical Co., Ltd., the manufacturer of favipiravir.

Figures

Figure 1. Titration of PICV in outbred…
Figure 1. Titration of PICV in outbred guinea pigs.
Hartley guinea pigs were challenged i.p. with 50,000, 5000, 500, 50, 5, 0.5 (n = 4/group), or 0.05 PFU (n = 3) p19 PICV and survival plotted over a 28-day post-infection (p.i.) period. The data are combined from two separate experiments.
Figure 2. Natural history of disease caused…
Figure 2. Natural history of disease caused by PICV infection in guinea pigs.
Animals were challenged with 500 PFU of p19 PICV, (A) daily weights and temperatures were recorded, and groups of 3 were sacrificed on the indicated day p.i. for collection of blood and tissues for analysis of (B) PT and aPTT clot times, (C) PLT counts, (D) serum AST levels, (E) infectious serum and tissue virus titers, and (F) splenomegaly during the course of infection. Due to a technical problem, PT clot times were not detected in two of three animals on day 1. *P<0.05 compared to day 1 p.i. (day 3 p.i. for PT).
Figure 3. Treatment of lethal PICV infection…
Figure 3. Treatment of lethal PICV infection with favipiravir starting 4 days after challenge.
Guinea pigs (n = 8/group) challenged with 500 PFU of p19 PICV were treated twice daily for 14 days with favipiravir, ribavirin, or placebo beginning on day 4 of infection (capped hashed line). Animals receiving favipiravir were initially fed 100 and 30 mg/kg/d (days 4 to 7) before increasing the doses to 300 and 90 mg/kg/d, respectively, starting on day 8 (indicated by arrow). (A) Survival, (B) body weights, and (C) temperatures were monitored for 29 days. Serum was collected on day 11 for analysis of (D) AST and (E) viremia, with the exception of two moribund animals from the placebo group that had to be sacrificed on day 10. *P<0.05 compared to placebo-treated animals.
Figure 4. Favipiravir treatment of advanced PICV…
Figure 4. Favipiravir treatment of advanced PICV infection in guinea pigs.
Guinea pigs (n = 7–8/group) challenged with 500 PFU of p19 PICV were treated with the indicated dosages of favipiravir, ribavirin, or placebo beginning on day 7 of infection. The 150-mg/kg/d group received a loading dose of 300 mg/kg/d on the first day of treatment. Drugs were administered twice daily for 14 days (capped hashed line) and (A) survival, (B) body weights, and (C) temperatures were monitored for 36 days. Serum was collected on day 10 for analysis of (D) AST and (E) viremia. *P<0.05 compared to placebo-treated animals.

References

    1. Charrel RN, de Lamballerie X. Arenaviruses other than Lassa virus. Antiviral Research. 2003;57:89–100.
    1. Delgado S, Erickson BR, Agudo R, Blair PJ, Vallejo E, et al. Chapare virus, a newly discovered arenavirus isolated from a fatal hemorrhagic fever case in Bolivia. PLoS Pathog. 2008;4:e1000047.
    1. Briese T, Paweska JT, McMullan LK, Hutchison SK, Street C, et al. Genetic detection and characterization of Lujo virus, a new hemorrhagic fever-associated arenavirus from southern Africa. PLoS Pathog. 2009;5:e1000455.
    1. Fichet-Calvet E, Rogers DJ. Risk maps of Lassa fever in West Africa. PLoS Negl Trop Dis. 2009;3:e388.
    1. Enria DA, Briggiler AM, Sanchez Z. Treatment of Argentine hemorrhagic fever. Antiviral Res. 2008;78:132–139.
    1. Maiztegui JI, Fernandez NJ, de Damilano AJ. Efficacy of immune plasma in treatment of Argentine haemorrhagic fever and association between treatment and a late neurological syndrome. Lancet. 1979;2:1216–1217.
    1. Enria DA, Briggiler AM, Levis S, Vallejos D, Maiztegui JI, et al. Tolerance and antiviral effect of ribavirin in patients with Argentine hemorrhagic fever. Antiviral Res. 1987;7:353–359.
    1. Kilgore PE, Ksiazek TG, Rollin PE, Mills JN, Villagra MR, et al. Treatment of Bolivian hemorrhagic fever with intravenous ribavirin. Clinical Infectious Diseases. 1997;24:718–722.
    1. McCormick JB, King IJ, Webb PA, Scribner CL, Craven RB, et al. Lassa fever. Effective therapy with ribavirin. New England Journal of Medicine. 1986;314:20–26.
    1. Borio L, Inglesby T, Peters CJ, Schmaljohn AL, Hughes JM, et al. Hemorrhagic fever viruses as biological weapons: medical and public health management. Jama. 2002;287:2391–2405.
    1. Bodenheimer HC, Jr, Lindsay KL, Davis GL, Lewis JH, Thung SN, et al. Tolerance and efficacy of oral ribavirin treatment of chronic hepatitis C: a multicenter trial. Hepatology. 1997;26:473–477.
    1. Chapman LE, Mertz GJ, Peters CJ, Jolson HM, Khan AS, et al. Intravenous ribavirin for hantavirus pulmonary syndrome: safety and tolerance during 1 year of open-label experience. Ribavirin Study Group. Antivir Ther. 1999;4:211–219.
    1. Fisher-Hoch SP, Gborie S, Parker L, Huggins J. Unexpected adverse reactions during a clinical trial in rural west Africa. Antiviral Res. 1992;19:139–147.
    1. Russmann S, Grattagliano I, Portincasa P, Palmieri VO, Palasciano G. Ribavirin-induced anemia: mechanisms, risk factors and related targets for future research. Curr Med Chem. 2006;13:3351–3357.
    1. Kochhar DM, Penner JD, Knudsen TB. Embryotoxic, teratogenic, and metabolic effects of ribavirin in mice. Toxicol Appl Pharmacol. 1980;52:99–112.
    1. Ferm VH, Willhite C, Kilham L. Teratogenic effects of ribavirin on hamster and rat embryos. Teratology. 1978;17:93–101.
    1. Bausch DG, Hadi CM, Khan SH, Lertora JJ. Review of the literature and proposed guidelines for the use of oral ribavirin as postexposure prophylaxis for Lassa fever. Clin Infect Dis. 2010;51:1435–1441.
    1. Furuta Y, Takahashi K, Fukuda Y, Kuno M, Kamiyama T, et al. In vitro and in vivo activities of anti-influenza virus compound T-705. Antimicrob Agents Chemother. 2002;46:977–981.
    1. Gowen BB, Wong MH, Jung KH, Sanders AB, Mendenhall M, et al. In vitro and in vivo activities of T-705 against arenavirus and bunyavirus infections. Antimicrob Agents Chemother. 2007;51:3168–3176.
    1. Morrey JD, Taro BS, Siddharthan V, Wang H, Smee DF, et al. Efficacy of orally administered T-705 pyrazine analog on lethal West Nile virus infection in rodents. Antiviral research. 2008;80:377–379.
    1. Julander JG, Shafer K, Smee DF, Morrey JD, Furuta Y. Activity of T-705 in a hamster model of yellow fever virus infection in comparison with that of a chemically related compound, T-1106. Antimicrob Agents Chemother. 2009;53:202–209.
    1. Kiso M, Takahashi K, Sakai-Tagawa Y, Shinya K, Sakabe S, et al. T-705 (favipiravir) activity against lethal H5N1 influenza A viruses. Proc Natl Acad Sci U S A. 2010;107:882–887.
    1. Sleeman K, Mishin VP, Deyde VM, Furuta Y, Klimov AI, et al. In Vitro antiviral activity of favipiravir (T-705) against drug-resistant influenza and 2009 A(H1N1) viruses. Antimicrob Agents Chemother. 2010;54:2517–2524.
    1. Gowen BB, Smee DF, Wong MH, Hall JO, Jung KH, et al. Treatment of late stage disease in a model of arenaviral hemorrhagic fever: T-705 efficacy and reduced toxicity suggests an alternative to ribavirin. PLoS One. 2008;3:e3725.
    1. Mendenhall M, Russell A, Juelich T, Messina EL, Smee DF, et al. T-705 (Favipiravir) Inhibition of Arenavirus Replication in Cell Culture. 2010. Antimicrob Agents Chemother: In press.
    1. Flatz L, Rieger T, Merkler D, Bergthaler A, Regen T, et al. T cell-dependence of Lassa fever pathogenesis. PLoS Pathog. 2010;6:e1000836.
    1. Kolokoltsova OA, Yun NE, Poussard AL, Smith JK, Smith JN, et al. Mice lacking interferon {alpha}/{beta} and {gamma} receptors are susceptible to Junin virus infection. J Virol 2010
    1. Jahrling PB, Smith S, Hesse RA, Rhoderick JB. Pathogenesis of Lassa virus infection in guinea pigs. Infect Immun. 1982;37:771–778.
    1. Oubina JR, Carballal G, Videla CM, Cossio PM. The guinea pig model for Argentine hemorrhagic fever. Am J Trop Med Hyg. 1984;33:1251–1257.
    1. Yun NE, Linde NS, Dziuba N, Zacks MA, Smith JN, et al. Pathogenesis of XJ and Romero strains of Junin virus in two strains of guinea pigs. Am J Trop Med Hyg. 2008;79:275–282.
    1. Hall WC, Geisbert TW, Huggins JW, Jahrling PB. Experimental infection of guinea pigs with Venezuelan hemorrhagic fever virus (Guanarito): a model of human disease. Am J Trop Med Hyg. 1996;55:81–88.
    1. Jahrling PB, Hesse RA, Rhoderick JB, Elwell MA, Moe JB. Pathogenesis of a pichinde virus strain adapted to produce lethal infections in guinea pigs. Infection and Immunity. 1981;32:872–880.
    1. Aronson JF, Herzog NK, Jerrells TR. Pathological and virological features of arenavirus disease in guinea pigs. Comparison of two Pichinde virus strains. American Journal of Pathology. 1994;145:228–235.
    1. Enria DA, Maiztegui JI. Antiviral treatment of Argentine hemorrhagic fever. Antiviral Research. 1994;23:23–31.
    1. Reed LJ, Muench H. A simple method of estimating fifty percent endpoints. American Journal of Hygiene. 1938;27:493–497.
    1. Zhang L, Marriott K, Aronson JF. Sequence analysis of the small RNA segment of guinea pig-passaged Pichinde virus variants. Am J Trop Med Hyg. 1999;61:220–225.
    1. Zhang L, Marriott KA, Harnish DG, Aronson JF. Reassortant analysis of guinea pig virulence of pichinde virus variants. Virology. 2001;290:30–38.
    1. Scott EP, Aronson JF. Cytokine patterns in a comparative model of arenavirus haemorrhagic fever in guinea pigs. J Gen Virol. 2008;89:2569–2579.
    1. Lan S, McLay Schelde L, Wang J, Kumar N, Ly H, et al. Development of infectious clones for virulent and avirulent pichinde viruses: a model virus to study arenavirus-induced hemorrhagic fevers. J Virol. 2009;83:6357–6362.
    1. Vagdatli E, Gounari E, Lazaridou E, Katsibourlia E, Tsikopoulou F, et al. Platelet distribution width: a simple, practical and specific marker of activation of coagulation. Hippokratia. 2010;14:28–32.
    1. Choudhury A, Chung I, Blann AD, Lip GY. Platelet surface CD62P and CD63, mean platelet volume, and soluble/platelet P-selectin as indexes of platelet function in atrial fibrillation: a comparison of “healthy control subjects” and “disease control subjects” in sinus rhythm. J Am Coll Cardiol. 2007;49:1957–1964.
    1. Geisbert TW, Jahrling PB. Exotic emerging viral diseases: progress and challenges. Nat Med. 2004;10:S110–121.
    1. Kunz S. The role of the vascular endothelium in arenavirus haemorrhagic fevers. Thromb Haemost. 2009;102:1024–1029.
    1. Connolly BM, Jenson AB, Peters CJ, Geyer SJ, Barth JF, et al. Pathogenesis of Pichinde virus infection in strain 13 guinea pigs: an immunocytochemical, virologic, and clinical chemistry study. American Journal of Tropical Medicine and Hygiene. 1993;49:10–24.
    1. Walker DH, McCormick JB, Johnson KM, Webb PA, Komba-Kono G, et al. Pathologic and virologic study of fatal Lassa fever in man. American Journal of Pathology. 1982;107:349–356.
    1. Gowen BB, Wong MH, Jung KH, Smee DF, Morrey JD, et al. Efficacy of favipiravir (T-705) and T-1106 pyrazine derivatives in phlebovirus disease models. Antiviral Res. 2010;86:121–127.
    1. Gowen BB, Holbrook MR. Animal models of highly pathogenic RNA viral infections: hemorrhagic fever viruses. Antiviral Res. 2008;78:79–90.
    1. Lucia HL, Coppenhaver DH, Baron S. Arenavirus infection in the guinea pig model: antiviral therapy with recombinant interferon-alpha, the immunomodulator CL246,738 and ribavirin. Antiviral Research. 1989;12:279–292.
    1. Soares MM, King SW, Thorpe PE. Targeting inside-out phosphatidylserine as a therapeutic strategy for viral diseases. Nat Med. 2008;14:1357–1362.
    1. Gowen BB, Julander JG, London NR, Wong MH, Larson D, et al. Assessing changes in vascular permeability in a hamster model of viral hemorrhagic fever. Virol J. 2010;7:240.
    1. Johnson KM, McCormick JB, Webb PA, Smith ES, Elliott LH, et al. Clinical virology of Lassa fever in hospitalized patients. J Infect Dis. 1987;155:456–464.
    1. Reagan-Shaw S, Nihal M, Ahmad N. Dose translation from animal to human studies revisited. FASEB J. 2008;22:659–661.
    1. Furuta Y, Takahashi K, Kuno-Maekawa M, Sangawa H, Uehara S, et al. Mechanism of action of T-705 against influenza virus. Antimicrob Agents Chemother. 2005;49:981–986.

Source: PubMed

3
Předplatit