The impact of co-administration of ketoconazole and rifampicin on the pharmacokinetics of apremilast in healthy volunteers

Yong Liu, Simon Zhou, Yuntao Wan, Anfan Wu, Maria Palmisano, Yong Liu, Simon Zhou, Yuntao Wan, Anfan Wu, Maria Palmisano

Abstract

Aims: Two clinical studies were conducted to determine possible drug-drug interactions between apremilast and a strong CYP3A4 inhibitor, ketoconazole, or a potent CYP3A4 inducer, rifampicin. The main objectives of these two studies were to evaluate the impact of multiple doses of ketoconazole on the pharmacokinetics of apremilast and its metabolites, and the effect of multiple oral doses of rifampicin on the pharmacokinetics of apremilast.

Methods: These single centre, open label, sequential treatment studies in healthy subjects included two treatment periods for ketoconazole and three treatment periods for rifampicin. Apremilast was administered as a 20 mg (ketoconazole study) or 30 mg (rifampicin study) single oral dose.

Results: Ketoconazole increases overall exposure (AUC(0,∞)) of apremilast by ≈36% (2827 vs. 2072 ng ml(-1) h, 90% CI = 126.2, 147.5) and peak exposure (Cmax ) by 5% (247 vs. 236 ng ml(-1) ). Multiple doses of rifampicin increase apremilast clearance ≈3.6-fold and decrease apremilast mean AUC(0,∞) by ≈72% (3120 vs. 869 ng ml(-1) h, 90% CI = 25.7, 30.4) and Cmax (from 290 vs. 166 ng ml(-1) ) relative to that of apremilast given alone. A 30 min intravenous infusion of rifampicin 600 mg had negligible effects on the overall exposure (AUC(0,∞)) of apremilast (2980 vs. 3120 ng ml(-1) h, 90% CI = 88.0, 104.1).

Conclusion: Ketoconazole slightly decreased apremilast clearance, resulting in a small increase in AUC which is probably not meaningful clinically. However, the effect of CYP3A4 induction by rifampicin on apremilast clearance is much more pronounced than that of CYP3A4 inhibition by ketoconazole. Strong CYP3A4 inducers may result in a loss of efficacy of apremilast because of decreased drug exposure.

Keywords: drug interaction; inflammatory autoimmune disorders; phosphodiesterase 4 inhibitor.

© 2014 Celgene. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of The British Pharmacological Society.

Figures

Figure 1
Figure 1
Mean (± SD) plasma concentration (ng ml−1) of apremilast (APR) vs. time by treatment with and without ketoconazole (KETO) on a semi-logarithmic scale. , APR; , KETO + APR
Figure 2
Figure 2
Mean (± SD) plasma concentration (ng ml−1) of apremilast (APR) vs. time by treatment with and without multiple oral dose and single intravenous (i.v.) rifampicin (RIF) on a semi-logarithmic scale. , APR; , APR + i.v. RIF; , APR + oral RIF

References

    1. Schafer P. Apremilast mechanism of action and application to psoriasis and psoriatic arthritis. Biochem Pharmacol. 2012;83:1583–1590.
    1. Kavanaugh A, Mease PJ, Gomez-Reino JJ, Adebajo AO, Wollenhaupt J, Gladman DD, Lespessailles E, Hall S, Hochfeld M, Hu C, Hough D, Stevens RM, Schett G. Treatment of psoriatic arthritis in a phase 3 randomised, placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. 2014;73:1020–1026.
    1. Papp K, Cather J, Rosoph L, Sofen H, Langley RG, Matheson RT, Hu A, Day RM. The efficacy of apremilast, a phosphodiesterase-4 inhibitor, in the treatment of moderate to severe psoriasis: results of a phase 2 randomised study. Lancet. 2012;380:738–746.
    1. Reich K, Papp K, Leonardi C, Kircik L, Chimenti S, Hu CC, Stevens RM, Day RM, Griffiths CEM. Apremilast, an oral phosphodiesterase 4 inhibitor, in patients with moderate to severe psoriasis: 16-week results of a phase 3, randomized, controlled trial (ESTEEM 1) [oral presentation] Annual Meeting of the American Academy of Dermatology; March 1–5, 2013; Miami, FL.
    1. Schett G, Wollenhaupt J, Papp K, Joos R, DeVlam KL, Rodrigues JF, Vessey A, Hu CC, Stevens R. Oral apremilast in the treatment of active psoriatic arthritis: results of a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum. 2012;64:3156–3167.
    1. Hoffmann M, Kumar G, Schafer P, Cedzik D, Capone L, Fong KL, Gu Z, Heller D, Feng H, Surapaneni S, Laskin O, Wu A. Disposition, metabolism and mass balance of [(14)C]apremilast following oral administration. Xenobiotica. 2011;41:1063–1075.
    1. Wu A, Wan Y, Laille E, Liangang L, Wang X, Stevens LA, Sidhu S, Palmisano M. Absolute bioavailability of apremilast using a [14C] labeled microtracer IV solution concomitantly with an oral dose [abstract] 2011 American Association of Pharmaceutical Sciences Annual Meeting and Exposition; October 23–27, 2011; Washington, DC.
    1. U.S. Food and Drug Administration, Center for Drugs Evaluation Research (CDER) 2012. Guidance for industry: drug interaction studies - study design, data analysis, implications for dosing, and labeling recommendations. Available at (last accessed 13 December 2013)
    1. European Medicines Agency Committee for Human Medicinal Products. 2012. Guideline on the investigation of drug interactions. Available at (last accessed 13 December 2013)
    1. Reitman ML, Chu X, Cai X, Yabut J, Venkatasubramanian R, Zajic S, Stone JA, Ding Y, Witter R, Gibson C, Roupe K, Evers R, Wagner JA, Stoch A. Rifampin's acute inhibitory and chronic inductive drug interactions: experimental and model-based approaches to drug-drug interaction trial design. Clin Pharmacol Ther. 2011;89:234–242.
    1. Vavricka SR, Van Montfoort J, Ha HR, Meier PJ, Fattinger K. Interactions of rifamycin SV and rifampicin with organic anion uptake systems of human liver. Hepatology. 2002;36:164–172.
    1. Smith SJ, Brookes-Fazakerley S, Donnelly LE, Barnes PJ, Barnette MS, Giembycz MA. Ubiquitous expression of phosphodiesterase 7A in human proinflammatory and immune cells. Am J Physiol Lung Cell Mol Physiol. 2003;284:L279–L289.
    1. Xiong H, Carr RA, Locke CS, Katz DA, Achari R, Doan TT, Wang P, Jankowski JR, Sleep DJ. Dual effects of rifampin on the pharmacokinetics of atrasentan. J Clin Pharmacol. 2007;47:423–429.
    1. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, McGrath JC, Catterall WA, Spedding M, Peters JA Harmar AJ and CGTP Collaborators. The Concise Guide to PHARMACOLOGY 2013/14. Br J Pharmacol. 2013a;170:1449–1867.
    1. Wu A, Scheffler M. First-time-in-man, safety/tolerability and pharmacokinetics of ascending oral doses of apremilast (APR) in healthy subjects (HS) [abstract 515] J Invest Dermatol. 2011;131:S86.
    1. Gottlieb AB, Strober B, Krueger JG, Rohane P, Zeldis JB, Hu CC, Kipnis C. An open-label, single-arm pilot study in patients with severe plaque-type psoriasis treated with an oral anti-inflammatory agent, apremilast. Curr Med Res Opin. 2008;24:1529–1538.
    1. Liu Y, Zhou S, Nissel J, Wu A, Lau H, Palmisano M. The pharmacokinetic effect of coadministration of apremilast and methotrexate in individuals with rheumatoid arthritis and psoriatic arthritis. Clin Pharmacol Drug Dev. 2014 doi: .
    1. Olkkola KT, Backman JT, Neuvonen PJ. Midazolam should be avoided in patients receiving the systemic antimycotics ketoconazole or itraconazole. Clin Pharmacol Ther. 1994;55:481–485.
    1. Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivisto KT. Pharmacokinetic interactions with rifampicin : clinical relevance. Clin Pharmacokinet. 2003;42:819–850.
    1. Rae JM, Johnson MD, Lippman ME, Flockhart DA. Rifampin is a selective, leiotropic inducer of drug metabolism genes in human hepatocytes: studies with cDNA and oligonucleotide expression arrays. JPET. 2001;299:849–857.
    1. Backman JT, Olkkola KT, Neuvonen PJ. Rifampin drastically reduces plasma concentrations and effects of oral midazolam. Clin Pharmacol Ther. 1996;59:7–13.

Source: PubMed

3
Předplatit