Favipiravir pharmacokinetics in Ebola-Infected patients of the JIKI trial reveals concentrations lower than targeted

Thi Huyen Tram Nguyen, Jérémie Guedj, Xavier Anglaret, Cédric Laouénan, Vincent Madelain, Anne-Marie Taburet, Sylvain Baize, Daouda Sissoko, Boris Pastorino, Anne Rodallec, Géraldine Piorkowski, Sara Carazo, Mamoudou N Conde, Jean-Luc Gala, Joseph Akoi Bore, Caroline Carbonnelle, Frédéric Jacquot, Hervé Raoul, Denis Malvy, Xavier de Lamballerie, France Mentré, JIKI study group, Thi Huyen Tram Nguyen, Jérémie Guedj, Xavier Anglaret, Cédric Laouénan, Vincent Madelain, Anne-Marie Taburet, Sylvain Baize, Daouda Sissoko, Boris Pastorino, Anne Rodallec, Géraldine Piorkowski, Sara Carazo, Mamoudou N Conde, Jean-Luc Gala, Joseph Akoi Bore, Caroline Carbonnelle, Frédéric Jacquot, Hervé Raoul, Denis Malvy, Xavier de Lamballerie, France Mentré, JIKI study group

Abstract

Background: In 2014-2015, we assessed favipiravir tolerance and efficacy in patients with Ebola virus (EBOV) disease (EVD) in Guinea (JIKI trial). Because the drug had never been used before for this indication and that high concentrations of the drugs were needed to achieve antiviral efficacy against EBOV, a pharmacokinetic model had been used to propose relevant dosing regimen. Here we report the favipiravir plasma concentrations that were achieved in participants in the JIKI trial and put them in perspective with the model-based targeted concentrations.

Methods and findings: Pre-dose drug concentrations were collected at Day-2 and Day-4 of treatment in 66 patients of the JIKI trial and compared to those predicted by the model taking into account patient's individual characteristics. At Day-2, the observed concentrations were slightly lower than the model predictions adjusted for patient's characteristics (median value of 46.1 versus 54.3 μg/mL for observed and predicted concentrations, respectively, p = 0.012). However, the concentrations dropped at Day-4, which was not anticipated by the model (median values of 25.9 and 64.4 μg/mL for observed and predicted concentrations, respectively, p<10-6). There was no significant relationship between favipiravir concentrations and EBOV viral kinetics or mortality.

Conclusions: Favipiravir plasma concentrations in the JIKI trial failed to achieve the target exposure defined before the trial. Furthermore, the drug concentration experienced an unanticipated drop between Day-2 and Day-4. The origin of this drop could be due to severe sepsis conditions and/or to intrinsic properties of favipiravir metabolism. Dose-ranging studies should be performed in healthy volunteers to assess the concentrations and the tolerance that could be achieved with high doses.

Trial registration: ClinicalTrials.gov NCT02329054.

Conflict of interest statement

I have read the journal's policy and the authors of this manuscript have the following competing interests: THTN, JG, VM, SB, HR, XdL and FM received a grant from St Luke International University (Tokyo, Japan) to perform research on favipiravir in non-human primates. All other authors declared no conflict of interest.

Figures

Fig 1. Flowchart of the patients included…
Fig 1. Flowchart of the patients included in the PK analysis of the JIKI trial
Fig 2. Observed trough concentrations of favipiravir…
Fig 2. Observed trough concentrations of favipiravir at Day-2 (n = 44) and Day-4 (n = 50) after treatment initiation.
Red points represent concentrations measured in patients who died during the trial, green points represent concentrations measured in those who survived. Concentrations obtained in patients receiving adult dose or weighted-based dose are presented in circles and stars, respectively. Lines connect data obtained in the 28 patients who had both measurements at Day-2 and Day-4. Boxplots represent the distribution of the concentrations. The lower and upper hinges correspond to the first and third quartiles. The upper whisker extends from the hinge to the highest value that is within 1.5 * IQR of the hinge, where IQR is the inter-quartile range, or distance between the first and third quartiles. The lower whisker extends from the hinge to the lowest value within 1.5 * IQR of the hinge.
Fig 3. Observed trough concentrations (y-axis) versus…
Fig 3. Observed trough concentrations (y-axis) versus predicted trough concentrations (x-axis) at Day-2 (left, n = 44 observations) and Day-4 (right, n = 50) after treatment initiation.
Red points represent concentrations measured in patients who died during the trial, green points represent concentrations measured in those who survived.
Fig 4. Difference in Ct values at…
Fig 4. Difference in Ct values at Day-2 (left) or Day-4 (right) from baseline (the larger the value the larger the viral decline) versus drug concentrations.
Top: patients with a low baseline Ct value (Ct=20) (bottom, n = 26 and 37). Red points represent concentrations measured in patients who died during the trial, green points represent concentrations measured in those who survived. Triangles indicate Ct values that were above 40 (detection limit) and were treated as equal to 40, and circles are observed values. Black lines are Loess trend lines.

References

    1. World Health Organization. WHO | Ebola situation reports [Internet]. Available:
    1. World Health Organization. Categorization and prioritization of drugs for consideration for testing or use in patients infected with Ebola. In: Ebola treatments and interventions. 3 July 2015 [Internet]. [cited 7 Sep 2015]. Available:
    1. Smither SJ, Eastaugh LS, Steward JA, Nelson M, Lenk RP, Lever MS. Post-exposure efficacy of oral T-705 (Favipiravir) against inhalational Ebola virus infection in a mouse model. Antiviral Res. 2014;104: 153–155. 10.1016/j.antiviral.2014.01.012
    1. Oestereich L, Lüdtke A, Wurr S, Rieger T, Muñoz-Fontela C, Günther S. Successful treatment of advanced Ebola virus infection with T-705 (favipiravir) in a small animal model. Antiviral Res. 2014;105: 17–21. 10.1016/j.antiviral.2014.02.014
    1. Madelain V, Nguyen THT, Olivo A, de Lamballerie X, Guedj J, Taburet A-M, et al. Ebola Virus Infection: Review of the Pharmacokinetic and Pharmacodynamic Properties of Drugs Considered for Testing in Human Efficacy Trials. Clin Pharmacokinet. 2016;55: 907–923. 10.1007/s40262-015-0364-1
    1. Japanese Pharmaceuticals and Medical Devices Agency (PMDA). Report on the Deliberation Results (English version) [Internet]. 4 Mar 2014 [cited 31 May 2016]. Available:
    1. Sissoko D, Laouenan C, Folkesson E, M’Lebing A-B, Beavogui A-H, Baize S, et al. Experimental Treatment with Favipiravir for Ebola Virus Disease (the JIKI Trial): A Historically Controlled, Single-Arm Proof-of-Concept Trial in Guinea. PLoS Med. 2016;13: e1001967 10.1371/journal.pmed.1001967
    1. Mentré F, Taburet A-M, Guedj J, Anglaret X, Keïta S, de Lamballerie X, et al. Dose regimen of favipiravir for Ebola virus disease. Lancet Infect Dis. 2015;15: 150–151. 10.1016/S1473-3099(14)71047-3
    1. Bouazza N, Treluyer J-M, Foissac F, Mentré F, Taburet A-M, Guedj J, et al. Favipiravir for children with Ebola. Lancet. 2015;385: 603–604.
    1. Bazzoli C, Jullien V, Le Tiec C, Rey E, Mentré F, Taburet A-M. Intracellular Pharmacokinetics of Antiretroviral Drugs in HIV-Infected Patients, and their Correlation with Drug Action. Clin Pharmacokinet. 2010;49: 17–45. 10.2165/11318110-000000000-00000
    1. Madelain V, Oestereich L, Graw F, Nguyen THT, de Lamballerie X, Mentré F, et al. Ebola virus dynamics in mice treated with favipiravir. Antiviral Res. 2015;123: 70–77. 10.1016/j.antiviral.2015.08.015
    1. Gowen BB, Sefing EJ, Westover JB, Smee DF, Hagloch J, Furuta Y, et al. Alterations in favipiravir (T-705) pharmacokinetics and biodistribution in a hamster model of viral hemorrhagic fever. Antiviral Res. 2015;121: 132–137. 10.1016/j.antiviral.2015.07.003
    1. Gordon AH, Green DE, Subrahmanyan V. Liver aldehyde oxidase. Biochem J. 1940;34: 764–774.
    1. Lobo DN, Stanga Z, Simpson JA, Anderson JA, Rowlands BJ, Allison SP. Dilution and redistribution effects of rapid 2-litre infusions of 0.9% (w/v) saline and 5% (w/v) dextrose on haematological parameters and serum biochemistry in normal subjects: a double-blind crossover study. Clin Sci Lond Engl 1979. 2001;101: 173–179.
    1. Marty AM, Jahrling PB, Geisbert TW. Viral hemorrhagic fevers. Clin Lab Med. 2006;26: 345–386, viii. 10.1016/j.cll.2006.05.001
    1. Roberts DJ, Hall RI. Drug absorption, distribution, metabolism and excretion considerations in critically ill adults. Expert Opin Drug Metab Toxicol. 2013;9: 1067–1084. 10.1517/17425255.2013.799137
    1. Boucher BA, Wood GC, Swanson JM. Pharmacokinetic changes in critical illness. Crit Care Clin. 2006;22: 255–271, vi. 10.1016/j.ccc.2006.02.011
    1. Hartmann T, Terao M, Garattini E, Teutloff C, Alfaro JF, Jones JP, et al. The Impact of Single Nucleotide Polymorphisms on Human Aldehyde Oxidase. Drug Metab Dispos. 2012;40: 856–864. 10.1124/dmd.111.043828
    1. Madelain V, Guedj J, Mentré F, Nguyen THT, Jacquot F, Oestereich L, et al. Favipiravir Pharmacokinetics in Nonhuman Primates and Insights for Future Efficacy Studies of Hemorrhagic Fever Viruses. Antimicrob Agents Chemother. 2017;61.

Source: PubMed

3
Subskrybuj