No Clinical Impact of CYP3A5 Gene Polymorphisms on the Pharmacokinetics and/or Efficacy of Maraviroc in Healthy Volunteers and HIV-1-Infected Subjects

Manoli Vourvahis, Lynn McFadyen, Sunil Nepal, Srinivas Rao Valluri, Annie Fang, Gwendolyn D Fate, Linda S Wood, Jean-Claude Marshall, Phylinda L S Chan, Angus Nedderman, Julian Haynes, Mark E Savage, Andrew Clark, Kimberly Y Smith, Jayvant Heera, Manoli Vourvahis, Lynn McFadyen, Sunil Nepal, Srinivas Rao Valluri, Annie Fang, Gwendolyn D Fate, Linda S Wood, Jean-Claude Marshall, Phylinda L S Chan, Angus Nedderman, Julian Haynes, Mark E Savage, Andrew Clark, Kimberly Y Smith, Jayvant Heera

Abstract

Maraviroc is a C-C chemokine receptor type-5 antagonist approved for the treatment of HIV-1. Previous studies show that cytochrome P450 3A5 (CYP3A5) plays a role in maraviroc metabolism. CYP3A5 is subject to a genetic polymorphism. The presence of 2 functional alleles (CYP3A5*1/*1) confers the extensive metabolism phenotype, which is rare in whites but common in blacks. The effect of CYP3A5 genotype on maraviroc and/or metabolite pharmacokinetics was evaluated in 2 clinical studies: a post hoc analysis from a phase 2b/3 study (NCT00098293) conducted in 494 HIV-1-infected subjects (study 1) in which the impact on maraviroc efficacy in 303 subjects was also assessed, and a study conducted in 47 healthy volunteers (study 2). In study 2 (NCT02625207), extensive metabolizers had 26% to 37% lower mean area under the concentration-time curve compared with poor metabolizers (no CYP3A5*1 alleles). This effect diminished to 17% in the presence of potent CYP3A inhibition. The effect of CYP3A5 genotype was greatest in the formation of the metabolite (1S,2S)-2-hydroxymaraviroc. In study 1, the CYP3A5*1/*1 genotype unexpectedly had higher maraviroc area under the curve predictions (20%) compared with those with no CYP3A5*1 alleles. The reason for this disparity remains unclear. The proportions of subjects with viral loads <50 and <400 copies/mL for maraviroc were comparable among all 3 CYP3A5 genotypes. In both studies maraviroc exposures were in the range of near-maximal viral inhibition in the majority of subjects. These results demonstrate that although CYP3A5 contributes to the metabolism of maraviroc, CYP3A5 genotype does not affect the clinical response to maraviroc in combination treatment of HIV-1 infection at approved doses.

Keywords: C-C chemokine receptor type-5 antagonist; CYP3A inhibition; CYP3A5 genotype; MERIT study; cytochrome P450; maraviroc.

© 2018, The Authors. The Journal of Clinical Pharmacology published by Wiley Periodicals, Inc. on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
Effect of CYP3A5 genotype on estimated maraviroc Cavg in HIV‐infected subjects (study 1). Upper plot, all maraviroc subjects (n = 494); middle plot, whites (n = 311); lower plot, blacks (n = 138). Dashed line represents median Cavg for distribution; solid reference line represents Cavg = 75 ng/mL, exposure associated with near‐maximal virologic efficacy.28 Cavg indicates average plasma concentration; EM, extensive metabolizer (2 CYP3A5*1 alleles); IM, intermediate metabolizer (1 CYP3A5*1 allele); PM, poor metabolizer (no CYP3A5*1 alleles).
Figure 2
Figure 2
Maraviroc plasma concentration‐time profiles by CYP3A5 genotype (study 2). A, Maraviroc plasma concentrations following maraviroc 300 mg twice‐daily dosing in part 1. B, Maraviroc plasma concentrations following maraviroc 150 mg + darunavir/cobicistat 800/150 mg once‐daily dosing in part 2. EM indicates extensive metabolizer (2 CYP3A5*1 alleles); IM, intermediate metabolizer (1 CYP3A5*1 allele); PM, poor metabolizer (no CYP3A5*1 alleles).
Figure 3
Figure 3
Maraviroc Cavg exposure by CYP3A5 genotype with and without CYP3A inhibition (study 2). Solid reference line represents Cavg = 75 ng/mL, exposure associated with near‐maximal virologic efficacy in study 1.28 Each boxplot shows the interquartile range with the median (horizontal line) and mean (×) Cavg values indicated. Whiskers show the minimum and maximum values. The circles represent individual scores. Cavg indicates average plasma concentration; EM, extensive metabolizer (2 CYP3A5*1 alleles); IM, intermediate metabolizer (1 CYP3A5*1 allele); MVC, maraviroc; PM, poor metabolizer (no CYP3A5*1 alleles).

References

    1. Abel S, Back DJ, Vourvahis M. Maraviroc: pharmacokinetics and drug interactions. Antivir Ther. 2009;14(5):607–618.
    1. Abel S, Russell D, Whitlock LA, Ridgway CE, Nedderman AN, Walker DK. Assessment of the absorption, metabolism and absolute bioavailability of maraviroc in healthy male subjects. Br J Clin Pharmacol. 2008;65(suppl 1):60–67.
    1. Hyland R, Dickins M, Collins C, Jones H, Jones B. Maraviroc: in vitro assessment of drug‐drug interaction potential. Br J Clin Pharmacol. 2008;66(4):498–507.
    1. Walker DK, Abel S, Comby P, Muirhead GJ, Nedderman AN, Smith DA. Species differences in the disposition of the CCR5 antagonist, UK‐427,857, a new potential treatment for HIV. Drug Metab Dispos. 2005;33(4):587–595.
    1. Tseng E, Fate GD, Walker GS, Goosen TC, Obach RS. Biosynthesis and identification of metabolites of maraviroc and their use in experiments to delineate the relative contributions of cytochrome P4503A4 versus 3A5. Drug Metab Dispos. 2018;46(5):493–502.
    1. Abel S, van der Ryst E, Rosario MC, et al. Assessment of the pharmacokinetics, safety and tolerability of maraviroc, a novel CCR5 antagonist, in healthy volunteers. Br J Clin Pharmacol. 2008;65(suppl 1):5–18.
    1. Siccardi M, D'Avolio A, Nozza S, et al. Maraviroc is a substrate for OATP1B1 in vitro and maraviroc plasma concentrations are influenced by SLCO1B1 521 T>C polymorphism. Pharmacogenet Genomics. 2010;20(12):759–765.
    1. Lu Y, Hendrix CW, Bumpus NN. Cytochrome P450 3A5 plays a prominent role in the oxidative metabolism of the anti‐human immunodeficiency virus drug maraviroc. Drug Metab Dispos. 2012;40(12):2221–2230.
    1. Tseng E, Walsky RL, Luzietti RA Jr, et al. Relative contributions of cytochrome CYP3A4 versus CYP3A5 for CYP3A‐cleared drugs assessed in vitro using a CYP3A4‐selective inactivator (CYP3cide). Drug Metab Dispos. 2014;42(7):1163–1173.
    1. Lu Y, Fuchs EJ, Hendrix CW, Bumpus NN. CYP3A5 genotype impacts maraviroc concentrations in healthy volunteers. Drug Metab Dispos. 2014;42(11):1796–1802.
    1. The International HapMap Consortium . dbSNP short genetic variations. National Center for Biotechnology Information. . Accessed April 25, 2018.
    1. Centers for Disease Control and Prevention. HIV Surveillance Report, 2015; volume 27. . Published November 2016. Accessed April 25, 2018.
    1. UNAIDS. Global AIDS Update 2016. . Published July 2017. Accessed April 25, 2018.
    1. Fatkenheuer G, Nelson M, Lazzarin A, et al, the MOTIVATE 1 and MOTIVATE 2 Study Teams . Subgroup analyses of maraviroc in previously treated R5 HIV‐1 infection. N Engl J Med. 2008;359(14):1442–1455.
    1. Jacqmin P, Wade JR, Weatherley B, Snoeck E, Marshall S, McFadyen L. Assessment of maraviroc exposure‐response relationship at 48 weeks in treatment‐experienced HIV‐1‐infected patients in the MOTIVATE studies. CPT Pharmacometrics Syst Pharmacol. 2013;2:e64.
    1. Rosario MC, Poland B, Sullivan J, Westby M, van der Ryst E. A pharmacokinetic‐pharmacodynamic model to optimize the phase IIa development program of maraviroc. J Acquir Immune Defic Syndr. 2006;42(2):183–191.
    1. Mathias AA, German P, Murray BP, et al. Pharmacokinetics and pharmacodynamics of GS‐9350: a novel pharmacokinetic enhancer without anti‐HIV activity. Clin Pharmacol Ther. 2010;87(3):322–329.
    1. Cooper DA, Heera J, Goodrich J, et al. Maraviroc versus efavirenz, both in combination with zidovudine‐lamivudine, for the treatment of antiretroviral‐naive subjects with CCR5‐tropic HIV‐1 infection. J Infect Dis. 2010;201(6):803–813.
    1. Kakuda TN, Van De Casteele T, Petrovic R, et al. Bioequivalence of a darunavir/cobicistat fixed‐dose combination tablet versus single agents and food effect in healthy volunteers. Antivir Ther. 2014;19(6):597–606.
    1. Brewer E, Felix T, Clarke P, Edgington A, Muirhead D. An LC‐MS‐MS method for quantitative determination of maraviroc (UK‐427,857) in human plasma, urine and cerebrospinal fluid. Biomed Chromatogr. 2010;24(12):1316–1323.
    1. Chan PL, Weatherley B, McFadyen L. A population pharmacokinetic meta‐analysis of maraviroc in healthy volunteers and asymptomatic HIV‐infected subjects. Br J Clin Pharmacol. 2008;65(suppl 1):76–85.
    1. Weatherley B, McFadyen L, Chan PL, Marsahall S. Population pharmacokinetic covariate analysis of maraviroc in the MERIT study in treatment naïve subjects. Poster presented at: 9th International Workshop on Clinical Pharmacology of HIV Therapy; April 7–9, 2008; New Orleans, LA.
    1. HapMap. The International HapMap Consortium . dbSNP Short Genetic Variations. . 2016. Accessed September 3, 2017.
    1. Jetter A, Fatkenheuer G, Frank D, et al. Do activities of cytochrome P450 (CYP)3A, CYP2D6 and P‐glycoprotein differ between healthy volunteers and HIV‐infected patients? Antivir Ther. 2010;15(7):975–983.
    1. Jones AE, Brown KC, Werner RE, et al. Variability in drug metabolizing enzyme activity in HIV‐infected patients. Eur J Clin Pharmacol. 2010;66(5):475–485.
    1. Kis O, Sankaran‐Walters S, Hoque MT, Walmsley SL, Dandekar S, Bendayan R. HIV‐1 alters intestinal expression of drug transporters and metabolic enzymes: implications for antiretroviral drug disposition. Antimicrob Agents Chemother. 2016;60(5):2771–2781.
    1. Gulick RM, Lalezari J, Goodrich J, et al, the MOTIVATE Study Teams . Maraviroc for previously treated patients with R5 HIV‐1 infection. N Engl J Med. 2008;359(14):1429–1441.
    1. McFadyen L, Jacqmin P, Weatherley B, Chan PL, Craig C, Westby M. Lack of adherence to treatment drives maraviroc exposure‐response in the merit study in treatment naïve HIV‐1 infected subjects. Abstract presented at: 11th International Workshop on Clinical Pharmacology of HIV Therapy; April 7‐9, 2010; Sorrento, Italy.
    1. Bernal E, Verdu JM, Vera F, et al. Improvement of endothelial function after switching previously treated HIV‐infected patients to an NRTI‐sparing bitherapy with maraviroc. J Int AIDS Soc. 2014;17(4 Suppl 3):19726.
    1. Macias J, Recio E, Marquez M, et al, on behalf of VEMAN Study Group . Efficacy and safety of once‐daily maraviroc plus ritonavir‐boosted darunavir in pretreated HIV‐infected patients in a real‐life setting. HIV Med. 2014;15(7):417–424.
    1. Nozza S, Galli L, Antinori A, et al, on behalf of VEMAN Study Group . Maraviroc 150 mg daily plus lopinavir/ritonavir, a nucleoside/nucleotide reverse transcriptase inhibitor‐sparing regimen for HIV‐infected naive patients: 48‐week final results of VEMAN study. Clin Microbiol Infect. 2015;21(5):510;e1–e9.
    1. Saumoy M, Llibre JM, Terron A, et al. Short communication: maraviroc once‐daily: experience in routine clinical practice. AIDS Res Hum Retroviruses. 2017;33(1):29–32.
    1. Stellbrink HJ, Le Fevre E, Carr A, et al. Once‐daily maraviroc versus tenofovir/emtricitabine each combined with darunavir/ritonavir for initial HIV‐1 treatment. AIDS. 2016;30(8):1229–1238.
    1. Taiwo B, Acosta EP, Ryscavage P, et al. Virologic response, early HIV‐1 decay, and maraviroc pharmacokinetics with the nucleos(t)ide‐free regimen of maraviroc plus darunavir/ritonavir in a pilot study. J Acquir Immune Defic Syndr. 2013;64(2):167–173.
    1. Gulick RM, Wilkin TJ, Chen YQ, et al. Safety and tolerability of maraviroc‐containing regimens to prevent HIV infection in women: a phase 2 randomized trial. Ann Intern Med. 2017;167(6):384–393.
    1. Gulick RM, Wilkin TJ, Chen YQ, et al. Phase 2 study of the safety and tolerability of maraviroc‐containing regimens to prevent HIV infection in men who have sex with men (HPTN 069/ACTG A5305). J Infect Dis. 2017;215(2):238–246.

Source: PubMed

3
Iratkozz fel