Effects of Maribavir on P-Glycoprotein and CYP2D6 in Healthy Volunteers

Ivy H Song, Katarina Ilic, Joseph Murphy, Kenneth Lasseter, Patrick Martin, Ivy H Song, Katarina Ilic, Joseph Murphy, Kenneth Lasseter, Patrick Martin

Abstract

Maribavir is an investigational drug being evaluated in transplant recipients with cytomegalovirus infection. To understand potential drug-drug interactions, we examined the effects of multiple doses of maribavir on cytochrome P450 (CYP) 2D6 and P-glycoprotein (P-gp) activity using probe substrates in healthy volunteers. During this phase 1 open-label study (NCT02775240), participants received the probe substrates digoxin (0.5 mg) and dextromethorphan (30 mg) before and after maribavir (400 mg twice daily for 8 days). Serial plasma samples were analyzed for digoxin, dextromethorpha, dextrorphan, and maribavir concentrations. Pharmacokinetic parameters were calculated (noncompartmental analysis) and analyzed with a linear mixed-effects model for treatment comparison to estimate geometric mean ratios (GMRs) and 90% confidence intervals (CIs). CYP2D6 polymorphisms were genotyped using polymerase chain reaction. Overall, 17 of 18 participants (94.4%) completed the study. All participants were genotyped as CYP2D6 intermediate/extensive metabolizers. GMR (90%CI) of digoxin Cmax , AUClast , and AUC0-∞ with and without maribavir was 1.257 (1.139-1.387), 1.187 (1.088-1.296), and 1.217 (1.110-1.335), respectively, outside the "no-effect" window (0.8-1.25). GMR (90%CI) of dextromethorphan AUClast and AUClast ratio of dextromethorphan/dextrorphan were 0.877 (0.692-1.112) and 0.901 (0.717-1.133), respectively, marginally outside the no-effect window, although large variability was observed in these pharmacokinetic parameters. Pharmacokinetic parameters of dextrorphan were unaffected. Maribavir inhibited P-gp activity but did not affect CYP2D6 activity. Maribavir's effect on the pharmacokinetics of P-gp substrates should be evaluated individually, and caution should be exercised with P-gp substrates with narrow therapeutic windows.

Keywords: P-glycoprotein; cytochrome P450 2D6; dextromethorphan; digoxin; maribavir; pharmacokinetics.

Conflict of interest statement

Ivy H. Song, Katarina Ilic, Joseph Murphy, and Patrick Martin are employees of Shire, a Takeda company, and Takeda stock owners. Kenneth Lasseter is an employee of Clinical Pharmacology of Miami Inc., which was funded by Shire Development LLC, Lexington, Massahusetts, a Takeda company, to perform the study. Under the direction of the authors, Amy Horne, MSc, and Yelena Lyustikman, MSc, of Caudex Health, Oxford, UK, provided writing assistance for this publication, funded by Shire International GmbH, a Takeda company. I.H.S., K.I., J.M., and P.M. have received salary and ownership of stocks from Takeda. K.L. has received study funding from Takeda. I.H.S., K.I., J.M., P.M., and K.L. have signed authorship agreements with Shire, a Takeda company. These authorship agreements are in accordance with ICMJE and GPP3 guidelines.

© 2019 Shire Plc. The Journal of Clinical Pharmacology published by Wiley Periodicals, Inc. on behalf of American College of Clinical Pharmacology.

Figures

Figure 1
Figure 1
Mean plasma concentration‐time profile for digoxin by treatment period: log scale (pharmacokinetic set, n = 18). Treatment 1: digoxin 0.5 mg and dextromethorphan 30 mg administered on day 1. Treatment 2: maribavir 400 mg (2 × 200 mg) twice daily from days 8 to 15. On day 13, digoxin 0.5 mg and dextromethorphan 30 mg were administered with the morning dose of maribavir 400 mg (2 × 200 mg). Error bars represent the standard deviation.
Figure 2
Figure 2
Mean plasma concentration‐time profile for dextromethorphan and dextrorphan by treatment, with log scale (pharmacokinetic set, n = 18).a Treatment 1: digoxin 0.5 mg and dextromethorphan 30 mg administered on day 1. Treatment 2: maribavir 400 mg (2 × 200 mg) twice daily from days 8 to 15. On day 13, digoxin 0.5 mg and dextromethorphan 30 mg was administered with the morning dose of maribavir 400 mg (2 × 200 mg). Error bars represent the standard deviation. aSamples with concentrations of less than the LLOQ (0.2 ng/mL for dextromethorphan and 2.5 ng/mL for dextrorphan) were treated as zero when calculating mean concentration.

References

    1. Teira P, Battiwalla M, Ramanathan M, et al. Early cytomegalovirus reactivation remains associated with increased transplant‐related mortality in the current era: a CIBMTR analysis. Blood. 2016;127(20):2427‐2438.
    1. Hakimi Z, Aballea S, Ferchichi S, et al. Burden of cytomegalovirus disease in solid organ transplant recipients: a national matched cohort study in an inpatient setting. Transpl Infect Dis. 2017;19(5).
    1. Azevedo LS, Pierrotti LC, Abdala E, et al. Cytomegalovirus infection in transplant recipients. Clinics (Sao Paulo). 2015;70(7):515‐523.
    1. Meesing A, Razonable RR. New developments in the management of cytomegalovirus infection after transplantation. Drugs. 2018;78(11):1085‐1103.
    1. Accessdata . Ganciclovir. Highlights of prescribing information. . Accessed April 4, 2019.
    1. Accessdata . Valcyte. Highlights of prescribing information. . Accessed March 15, 2019.
    1. Accessdata . Letermovir. . Accessed March 15, 2019.
    1. Food and Drug Administration . Cytomegalovirus in transplantation: developing drugs to treat or prevent disease guidance for industry. . Accessed April 4, 2019.
    1. Globenewswire . Shire PLC: Shire receives FDA breakthrough therapy designation for maribavir, an investigational treatment for cytomegalovirus (CMV) infection in transplant patients. . Accessed March 15, 2019.
    1. Maertens J, Cordonnier C, Jaksch P, et al. Maribavir versus valganciclovir for preemptive treatment of cytomegalovirus (CMV) viremia: a randomized, dose‐ranging, phase 2 study among hematopoietic stem cell transplant (SCT) and solid organ transplant (SOT) recipients. Open Forum Infect Dis. 2016;3(suppl 1):2287.
    1. Papanicolaou GA, Silveira FP, Langston AA, et al. Maribavir for refractory or resistant cytomegalovirus infections in hematopoietic‐cell or solid‐organ transplant recipients: a randomized, dose‐ranging, double‐blind, phase 2 study. Clin Infect Dis. 2018;68(8):1256‐1264.
    1. Biron KK, Harvey RJ, Chamberlain SC, et al. Potent and selective inhibition of human cytomegalovirus replication by 1263W94, a benzimidazole L‐riboside with a unique mode of action. Antimicrob Agents Chemother. 2002;46(8):2365‐2372.
    1. Hamirally S, Kamil JP, Ndassa‐Colday YM, et al. Viral mimicry of Cdc2/cyclin‐dependent kinase 1 mediates disruption of nuclear lamina during human cytomegalovirus nuclear egress. PLoS Pathog. 2009;5(1):e1000275.
    1. Krosky PM, Baek MC, Coen DM. The human cytomegalovirus UL97 protein kinase, an antiviral drug target, is required at the stage of nuclear egress. J Virol. 2003;77(2):905‐914.
    1. Drew WL, Miner RC, Marousek GI, Chou S. Maribavir sensitivity of cytomegalovirus isolates resistant to ganciclovir, cidofovir or foscarnet. J Clin Virol. 2006;37(2):124‐127.
    1. Duncan MD, Wilkes DS. Transplant‐related immunosuppression: a review of immunosuppression and pulmonary infections. Proc Am Thorac Soc. 2005;2(5):449‐455.
    1. Cimino FM, Snyder KA. Primary care of the solid organ transplant recipient. Am Fam Physician. 2016;93(3):203‐210.
    1. Glotzbecker B, Duncan C, Alyea E 3rd, Campbell B, Soiffer R. Important drug interactions in hematopoietic stem cell transplantation: what every physician should know. Biol Blood Marrow Transplant. 2012;18(7):989‐1006.
    1. Koszalka GW, Johnson NW, Good SS, et al. Preclinical and toxicology studies of 1263W94, a potent and selective inhibitor of human cytomegalovirus replication. Antimicrob Agents Chemother. 2002;46(8):2373‐2380.
    1. Downey R, Johnson J, Gelone S, Broom C, Villano S. Lack of a pharmacokinetic (PK) interaction between oral Maribavir, a novel anti‐cytomegalovirus agent and the CYP 2C19 substrate Voriconazole, in healthy volunteers. Clin Pharmacol Ther. 2009;85:PIII‐74.
    1. Ma JD, Nafziger AN, Villano SA, Gaedigk A, Bertino JS Jr. Maribavir pharmacokinetics and the effects of multiple‐dose maribavir on cytochrome P450 (CYP) 1A2, CYP 2C9, CYP 2C19, CYP 2D6, CYP 3A, N‐acetyltransferase‐2, and xanthine oxidase activities in healthy adults. Antimicrob Agents Chemother. 2006;50(4):1130‐1135.
    1. Wyen C, Fuhr U, Frank D, et al. Effect of an antiretroviral regimen containing ritonavir boosted lopinavir on intestinal and hepatic CYP3A, CYP2D6 and P‐glycoprotein in HIV‐infected patients. Clin Pharmacol Ther. 2008;84(1):75‐82.
    1. Frank D, Jaehde U, Fuhr U. Evaluation of probe drugs and pharmacokinetic metrics for CYP2D6 phenotyping. Eur J Clin Pharmacol. 2007;63(4):321‐333.
    1. Borges S, Li L, Hamman MA, Jones DR, Hall SD, Gorski JC. Dextromethorphan to dextrorphan urinary metabolic ratio does not reflect dextromethorphan oral clearance. Drug Metab Dispos. 2005;33(7):1052‐1055.
    1. Fuhr U, Jetter A, Kirchheiner J. Appropriate phenotyping procedures for drug metabolizing enzymes and transporters in humans and their simultaneous use in the “cocktail” approach. Clin Pharmacol Ther. 2007;81(2):270‐283.
    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. Department of Health and Human Services (DHHS) FaDA , Center for Drug Evaluation and Research (CDER) Center for Veterinary Medicine (CVM) (US). Guidance for industry: Bioanalytical method of validation. . Accessed May 23, 2018.
    1. European Medicines Agency . Guideline on bioanalytical method validation. . Accessed April 4, 2019.
    1. Kakuda TN, Van Solingen‐Ristea RM, Onkelinx J, et al. The effect of single‐ and multiple‐dose etravirine on a drug cocktail of representative cytochrome P450 probes and digoxin in healthy subjects. J Clin Pharmacol. 2014;54(4):422‐431.
    1. Sun K, Rong H, Song I, Welty D. In vitro metabolic and transporter profiling for maribavir (SHP620) [abstract]. 22nd North American ISSX Meeting. 2018.
    1. Kullak‐Ublick GA, Ismair MG, Stieger B, et al. Organic anion‐transporting polypeptide B (OATP‐B) and its functional comparison with three other OATPs of human liver. Gastroenterology. 2001;120(2):525‐533.
    1. Taub ME, Mease K, Sane RS, et al. Digoxin is not a substrate for organic anion‐transporting polypeptide transporters OATP1A2, OATP1B1, OATP1B3, and OATP2B1 but is a substrate for a sodium‐dependent transporter expressed in HEK293 cells. Drug Metab Dispos. 2011;39(11):2093‐2102.
    1. Pescovitz MD, Bloom R, Pirsch J, Johnson J, Gelone S, Villano SA. A randomized, double‐blind, pharmacokinetic study of oral maribavir with tacrolimus in stable renal transplant recipients. Am J Transplant. 2009;9(10):2324‐2330.
    1. Staatz CE, Tett SE. Clinical pharmacokinetics and pharmacodynamics of tacrolimus in solid organ transplantation. Clin Pharmacokinet. 2004;43(10):623‐653.
    1. Murakami T IM, Oda K. Modulation of P‐glycoprotein Expression and Function under Disease States in Rats and Humans. Int J Clin Pharmacol Pharmacother. 2017;2:130.
    1. Lemahieu W, Maes B, Verbeke K, Rutgeerts P, Geboes K, Vanrenterghem Y. Cytochrome P450 3A4 and P‐glycoprotein activity and assimilation of tacrolimus in transplant patients with persistent diarrhea. Am J Transplant. 2005;5(6):1383‐1391.
    1. Storset E, Hole K, Midtvedt K, Bergan S, Molden E, Asberg A. The CYP3A biomarker 4beta‐hydroxycholesterol does not improve tacrolimus dose predictions early after kidney transplantation. Br J Clin Pharmacol. 2017;83(7):1457‐1465.
    1. Winston DJ, Young JA, Pullarkat V, et al. Maribavir prophylaxis for prevention of cytomegalovirus infection in allogeneic stem cell transplant recipients: a multicenter, randomized, double‐blind, placebo‐controlled, dose‐ranging study. Blood. 2008;111(11):5403‐5410.
    1. Goldwater DR, Dougherty C, Schumacher M, Villano SA. Effect of ketoconazole on the pharmacokinetics of maribavir in healthy adults. Antimicrob Agents Chemother. 2008;52(5):1794‐1798.
    1. Winston DJ, Saliba F, Blumberg E, et al. Efficacy and safety of maribavir dosed at 100 mg orally twice daily for the prevention of cytomegalovirus disease in liver transplant recipients: a randomized, double‐blind, multicenter controlled trial. Am J Transplant. 2012;12(11):3021‐3030.
    1. Lalezari JP, Aberg JA, Wang LH, et al. Phase I dose escalation trial evaluating the pharmacokinetics, anti‐human cytomegalovirus (HCMV) activity, and safety of 1263W94 in human immunodeficiency virus‐infected men with asymptomatic HCMV shedding. Antimicrob Agents Chemother. 2002;46(9):2969‐2976.
    1. Marty FM, Ljungman P, Papanicolaou GA, et al. Maribavir prophylaxis for prevention of cytomegalovirus disease in recipients of allogeneic stem‐cell transplants: a phase 3, double‐blind, placebo‐controlled, randomised trial. Lancet Infect Dis. 2011;11(4):284‐292; Erratum: 2011;2011:2343.
    1. Wang LH, Peck RW, Yin Y, Allanson J, Wiggs R, Wire MB. Phase I safety and pharmacokinetic trials of 1263W94, a novel oral anti‐human cytomegalovirus agent, in healthy and human immunodeficiency virus‐infected subjects. Antimicrob Agents Chemother. 2003;47(4):1334‐1342.
    1. Mohammadpour N, Elyasi S, Vahdati N, Mohammadpour AH, Shamsara J. A review on therapeutic drug monitoring of immunosuppressant drugs. Iran J Basic Med Sci. 2011;14(6):485‐498.

Source: PubMed

3
Iratkozz fel