Pharmacokinetics and Safety of Amenamevir in Healthy Subjects: Analysis of Four Randomized Phase 1 Studies

Tomohiro Kusawake, James J Keirns, Donna Kowalski, Martin den Adel, Dorien Groenendaal-van de Meent, Akitsugu Takada, Yoshiaki Ohtsu, Masataka Katashima, Tomohiro Kusawake, James J Keirns, Donna Kowalski, Martin den Adel, Dorien Groenendaal-van de Meent, Akitsugu Takada, Yoshiaki Ohtsu, Masataka Katashima

Abstract

Introduction: Amenamevir (ASP2151) is a nonnucleoside antiherpesvirus compound available for the treatment of varicella-zoster virus infections. In this article we summarize the findings of four phase 1 studies in healthy participants.

Methods: Four randomized phase 1 studies investigated the safety and pharmacokinetics of single and multiple doses of amenamevir, including the assessment of age group effect (nonelderly vs elderly), food effect, and the relative bioavailability of two formulations. Amenamevir was administered orally at various doses as a single dose (5-2400 mg) or daily (300 or 600 mg/day) for 7 days.

Results: Following single and multiple oral doses, amenamevir demonstrated a less than dose proportional increase in the pharmacokinetic parameters area under the plasma drug concentration versus time curve from time zero to infinity (AUCinf) and C max. After single and multiple oral 300-mg doses of amenamevir, no apparent differences in pharmacokinetics were observed between nonelderly and elderly participants. In contrast, with the amenamevir 600-mg dose both the area under the plasma drug concentration versus time curve from time zero to 24 h and C max were slightly increased and renal clearance was decreased in elderly participants. The pharmacokinetics of amenamevir was affected by food, with AUCinf increased by about 90%. In the bioavailability study, AUCinf and C max were slightly lower following tablet versus capsule administration (decreased by 14 and 12%, respectively), with relative bioavailability of 86%. The different amenamevir doses and formulations were safe and well tolerated; no deaths or serious adverse events were reported.

Conclusion: Amenamevir had less than dose proportional pharmacokinetic characteristics. Age may have an influence on amenamevir pharmacokinetics; however, the effect was considered minimal. The pharmacokinetics of amenamevir were affected by food, with AUCinf almost doubling when amenamevir was administered with food. The concentration versus time profile of the tablet was slightly lower than that of the capsule; the relative bioavailability of the tablet versus the capsule was 86%. Amenamevir was safe and well tolerated in the dose range investigated.

Funding: Astellas Pharma.

Trial registration: ClinicalTrials.gov identifiers NCT02852876 (15L-CL-002) and NCT02796118 (15L-CL-003).

Keywords: Amenamevir; Japanese participants; Pharmacokinetics; Safety; Varicella–zoster virus.

Figures

Fig. 1
Fig. 1
Mean plasma concentration versus time profile of amenamevir in a study 15L-CL-001 and b part 1 of study 15L-CL-002 after a single capsule dose of amenamevir at 5 mg (open circles), 25 mg (closed circles), 100 mg (open triangles), 300 mg (closed triangles), 600 mg (open squares), 1200 mg (closed squares), 1800 mg (open diamonds), and 2400 mg (closed diamonds)
Fig. 2
Fig. 2
Mean plasma concentration versus time profile of amenamevir after a single capsule dose under fasting (open circles) and fed (closed circles) conditions in part 2 of study 15L-CL-002
Fig. 3
Fig. 3
Mean plasma concentration versus time profiles of amenamevir after a single capsule (open circles) or tablet (open triangles) dose under fasting conditions as well as after a single tablet dose under fed conditions (open squares) in study 15L-CL-006

References

    1. Wareham DW, Breuer J. Herpes zoster. BMJ. 2007;334(7605):1211–1215. doi: 10.1136/.
    1. Cohen JI. Herpes zoster. N Engl J Med. 2013;369(18):1766–1767.
    1. Perry CM, Faulds D. Valaciclovir. A review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy in herpesvirus infections. Drugs. 1996;52(5):754–772. doi: 10.2165/00003495-199652050-00009.
    1. Perry CM, Wagstaff AJ. Famciclovir. A review of its pharmacological properties and therapeutic efficacy in herpesvirus infections. Drugs. 1995;50(2):396–415. doi: 10.2165/00003495-199550020-00011.
    1. Wagstaff AJ, Faulds D, Goa KL. Aciclovir. A reappraisal of its antiviral activity, pharmacokinetic properties and therapeutic efficacy. Drugs. 1994;47(1):153–205. doi: 10.2165/00003495-199447010-00009.
    1. Bestman-Smith J, Schmit I, Papadopoulou B, Boivin G. Highly reliable heterologous system for evaluating resistance of clinical herpes simplex virus isolates to nucleoside analogues. J Virol. 2001;75(7):3105–3110. doi: 10.1128/JVI.75.7.3105-3110.2001.
    1. Morfin F, Thouvenot D. Herpes simplex virus resistance to antiviral drugs. J Clin Virol. 2003;26(1):29–37. doi: 10.1016/S1386-6532(02)00263-9.
    1. Larder BA, Darby G. Selection and characterisation of acyclovir-resistant herpes simplex virus type 1 mutants inducing altered DNA polymerase activities. Virology. 1985;146(2):262–271. doi: 10.1016/0042-6822(85)90009-1.
    1. Sarisky RT, Quail MR, Clark PE, et al. Characterization of herpes simplex viruses selected in culture for resistance to penciclovir or acyclovir. J Virol. 2001;75(4):1761–1769. doi: 10.1128/JVI.75.4.1761-1769.2001.
    1. Sauerbrei A, Bohn K, Heim A, et al. Novel resistance-associated mutations of thymidine kinase and DNA polymerase genes of herpes simplex virus type 1 and type 2. Antivir Ther. 2011;16(8):1297–1308. doi: 10.3851/IMP1870.
    1. Sauerbrei A, Deinhardt S, Zell R, Wutzler P. Phenotypic and genotypic characterization of acyclovir-resistant clinical isolates of herpes simplex virus. Antiviral Res. 2010;86(3):246–252. doi: 10.1016/j.antiviral.2010.03.002.
    1. Jacobson MA, Berger TG, Fikrig S, et al. Acyclovir-resistant varicella zoster virus infection after chronic oral acyclovir therapy in patients with the acquired immunodeficiency syndrome (AIDS) Ann Intern Med. 1990;112(3):187–191. doi: 10.7326/0003-4819-112-3-187.
    1. Linnemann CC, Jr, Biron KK, Hoppenjans WG, Solinger AM. Emergence of acyclovir-resistant varicella zoster virus in an AIDS patient on prolonged acyclovir therapy. AIDS. 1990;4(6):577–579. doi: 10.1097/00002030-199006000-00014.
    1. Pahwa S, Biron K, Lim W, et al. Continuous varicella-zoster infection associated with acyclovir resistance in a child with AIDS. JAMA. 1988;260(19):2879–2882. doi: 10.1001/jama.1988.03410190127035.
    1. Snoeck R, Gérard M, Sadzot-Delvaux C, et al. Meningoradiculoneuritis due to acyclovir-resistant varicella zoster virus in an acquired immune deficiency syndrome patient. J Med Virol. 1994;42(4):338–347. doi: 10.1002/jmv.1890420404.
    1. Crute JJ, Grygon CA, Hargrave KD, et al. Herpes simplex virus helicase-primase inhibitors are active in animal models of human disease. Nat Med. 2002;8(4):386–391. doi: 10.1038/nm0402-386.
    1. Kleymann G, Fischer R, Betz UA, et al. New helicase-primase inhibitors as drug candidates for the treatment of herpes simplex disease. Nat Med. 2002;8(4):392–398. doi: 10.1038/nm0402-392.
    1. Crute JJ, Mocarski ES, Lehman IR. A DNA helicase induced by herpes simplex virus type 1. Nucleic Acids Res. 1988;16(14A):6585–6596. doi: 10.1093/nar/16.14.6585.
    1. Crute JJ, Tsurumi T, Zhu LA, et al. Herpes simplex virus 1 helicase-primase: a complex of three herpes-encoded gene products. Proc Natl Acad Sci U S A. 1989;86(7):2186–2189. doi: 10.1073/pnas.86.7.2186.
    1. Dodson MS, Crute JJ, Bruckner RC, Lehman IR. Overexpression and assembly of the herpes simplex virus type 1 helicase-primase in insect cells. J Biol Chem. 1989;264(35):20835–20838.
    1. Chono K, Katsumata K, Suzuki H, Shiraki K. Synergistic activity of amenamevir (ASP2151) with nucleoside analogs against herpes simplex virus types 1 and 2 and varicella-zoster virus. Antiviral Res. 2013;97(2):154–160. doi: 10.1016/j.antiviral.2012.12.006.
    1. Ohtsu Y, van Trigt R, Takama K, et al. Quantification of ASP2151 in human plasma and urine: a pitfall associated with supersaturation of analyte in urine. Chromatographia. 2017;80(2):217–227. doi: 10.1007/s10337-016-3236-4.
    1. Katsumata K, Chono K, Kato K, et al. Pharmacokinetics and pharmacodynamics of ASP2151, a helicase-primase inhibitor, in a murine model of herpes simplex virus infection. Antimicrob Agents Chemother. 2013;57(3):1339–1346. doi: 10.1128/AAC.01803-12.
    1. US Department of Health and Human Services. Guidance for industry: food-effect bioavailability and fed bioequivalence studies. 2002. . Accessed 27 Jun 2017
    1. Tyring S, Wald A, Zadeikis N, Dhadda S, Takenouchi K, Rorig R. ASP2151 for the treatment of genital herpes: a randomized, double-blind, placebo- and valacyclovir-controlled, dose-finding study. J Infect Dis. 2012;205(7):1100–1110. doi: 10.1093/infdis/jis019.

Source: PubMed

3
구독하다