Evaluation of Drug-Drug Interactions of Rucaparib and CYP1A2, CYP2C9, CYP2C19, CYP3A, and P-gp Substrates in Patients With an Advanced Solid Tumor

Jim J Xiao, Dorota Nowak, Rodryg Ramlau, Monika Tomaszewska-Kiecana, Piotr J Wysocki, Jeff Isaacson, Jeri Beltman, Eileen Nash, Robert Kaczanowski, Gerhard Arold, Simon Watkins, Jim J Xiao, Dorota Nowak, Rodryg Ramlau, Monika Tomaszewska-Kiecana, Piotr J Wysocki, Jeff Isaacson, Jeri Beltman, Eileen Nash, Robert Kaczanowski, Gerhard Arold, Simon Watkins

Abstract

This phase I study (CO-338-044; NCT02740712), conducted in patients with advanced solid tumors, evaluated the effect of the poly(ADP-ribose) polymerase (PARP) inhibitor rucaparib on the pharmacokinetics (PK) of caffeine 200 mg, warfarin 10 mg, omeprazole 40 mg, and midazolam 2 mg (cytochrome P450 (CYP) 1A2, CYP2C9, CYP2C19, and CYP3A substrates; dosed as a cocktail) and digoxin 0.25 mg (P-glycoprotein (P-gp) substrate; dosed separately) without rucaparib and following oral rucaparib 600 mg b.i.d. Geometric mean (GM) ratios (90% confidence interval (CI)) of area under the concentration-time curve (AUC) from time zero to last quantifiable measurement with and without rucaparib were: caffeine, 2.26 (1.93-2.65); S-warfarin, 1.49 (1.40-1.58); omeprazole, 1.55 (1.32-1.83); midazolam, 1.39 (1.14-1.68); and digoxin, 1.20 (1.12-1.29). There was limited effect on peak concentration of the substrates (GM ratios, 0.99-1.13). At steady state, rucaparib 600 mg b.i.d. moderately inhibited CYP1A2, weakly inhibited CYP2C9, CYP2C19, and CYP3A, and marginally increased digoxin exposure.

© 2018 Clovis Oncology Inc. Clinical and Translational Science published by Wiley Periodicals, Inc. on behalf of the American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Study schema. CYP, cytochrome P450.
Figure 2
Figure 2
Arithmetic mean (SD) plasma concentration‐time profiles for (a) caffeine, (b) S‐warfarin, (c) omeprazole, (d) midazolam, and (e) digoxin administered with (blue line) and without (red line) rucaparib. *Following the caffeine dose on day 1, 9 of the 16 evaluable patients had caffeine concentrations lower than the quantification limit at 72 hours postdose. One subject had a higher than expected concentration at the same time point, presumably due to incidental caffeine intake, contributing to an apparent spike in the mean caffeine pharmacokinetic profile. The presumed incidental caffeine intake in this one patient had no impact on CYP1A2 DDI assessment based on Cmax and AUC 0‐inf. AUC 0‐inf, area under the concentration‐time curve extrapolated from time 0 to infinity; Cmax, peak plasma concentration; DDI, drug–drug interaction.
Figure 3
Figure 3
Effect of rucaparib on the pharmacokinetics of probe drugs. AUC 0‐72 h, area under the concentration‐time curve from time 0−72 hours; AUC 0‐96 h, area under the concentration‐time curve from time 0−96 hours; AUC 0‐inf, area under the concentration‐time curve extrapolated from time 0 to infinity; CI, confidence interval; CYP, cytochrome P450; Cmax, peak plasma concentration; P‐gp, P‐glycoprotein.

References

    1. Rubraca (rucaparib) tablets [prescribing information] (Clovis Oncology, Inc.: Boulder, CO, 2018).
    1. Oza, A.M. et al Antitumor activity and safety of the PARP inhibitor rucaparib in patients with high‐grade ovarian carcinoma and a germline or somatic BRCA1 or BRCA2 mutation: Integrated analysis of data from study 10 and ARIEL2. Gynecol. Oncol. 147, 267–275 (2017).
    1. Coleman, R.L. et al Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): A randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet 390, 1949–1961 (2017).
    1. Kristeleit, R. et al A phase I‐II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2‐mutated ovarian carcinoma or other solid tumors. Clin. Cancer Res. 23, 4095–4106 (2017).
    1. Shapiro, G.I. et al Pharmacokinetic study of rucaparib in patients with advanced solid tumors. Clin. Pharmacol. Drug Dev. Epub ahead of print (2018).
    1. Swisher, E.M. et al Rucaparib in relapsed, platinum‐sensitive high‐grade ovarian carcinoma (ARIEL2 part 1): An international, multicentre, open‐label, phase 2 trial. Lancet Oncol. 18, 75–87 (2017).
    1. Chainuvati, S. et al Combined phenotypic assessment of cytochrome p450 1A2, 2C9, 2C19, 2D6, and 3A, N‐acetyltransferase‐2, and xanthine oxidase activities with the “Cooperstown 5 + 1 cocktail”. Clin. Pharmacol. Ther. 74, 437–447 (2003).
    1. Dumond, J.B. et al A phenotype‐genotype approach to predicting CYP450 and P‐glycoprotein drug interactions with the mixed inhibitor/inducer tipranavir/ritonavir. Clin. Pharmacol. Ther. 87, 735–742 (2010).
    1. Tachibana, M. et al Evaluation of the pharmacokinetic drug interaction potential of tivantinib (ARQ 197) using cocktail probes in patients with advanced solid tumours. Br. J. Clin. Pharmacol. 84, 112–121 (2018).
    1. Eisenhauer, E.A. et al New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).
    1. Brown, E.G. , Wood, L. & Wood, S. The medical dictionary for regulatory activities (MedDRA). Drug Saf. 20, 109–117 (1999).
    1. European Medicines Agency. Guideline on the investigation of drug interactions. <> (2012). Accessed 30 September 2017.
    1. U.S. Food and Drug Administration. Guidance for industry: Drug interaction studies — Study design, data analysis, implications for dosing, and labeling recommendations. <> (2012). Accessed 1 August 2017.
    1. Ebner, T. , Ishiguro, N. & Taub, M.E. The use of transporter probe drug cocktails for the assessment of transporter‐based drug–drug interactions in a clinical setting—proposal of a four component transporter cocktail. J. Pharm. Sci. 104, 3220–3228 (2015).
    1. Zhang, L. & Sparreboom, A. Predicting transporter‐mediated drug interactions: Commentary on: “Pharmacokinetic evaluation of a drug transporter cocktail consisting of digoxin, furosemide, metformin and rosuvastatin” and “Validation of a microdose probe drug cocktail for clinical drug interaction assessments for drug transporters and CYP3A”. Clin. Pharmacol. Ther. 101, 447–449 (2017).
    1. Gibbons, J.A. et al Pharmacokinetic drug interaction studies with enzalutamide. Clin. Pharmacokinet. 54, 1057–1069 (2015).
    1. Goh, B.C. et al An evaluation of the drug interaction potential of pazopanib, an oral vascular endothelial growth factor receptor tyrosine kinase inhibitor, using a modified Cooperstown 5 + 1 cocktail in patients with advanced solid tumors. Clin. Pharmacol. Ther. 88, 652–659 (2010).
    1. Polyak, K. & Garber, J. Targeting the missing links for cancer therapy. Nat. Med. 17, 283–284 (2011).
    1. Wahlberg, E. et al Family‐wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nat. Biotechnol. 30, 283–288 (2012).
    1. Thomas, H.D. et al Preclinical selection of a novel poly(ADP‐ribose) polymerase inhibitor for clinical trial. Mol. Cancer Ther. 6, 945–956 (2007).
    1. Jenner, Z.B. , Sood, A.K. & Coleman, R.L. Evaluation of rucaparib and companion diagnostics in the PARP inhibitor landscape for recurrent ovarian cancer therapy. Future Oncol. 12, 1439–1456 (2016).
    1. Stockis, A. , van Lier, J.J. , Cawello, W. , Kumke, T. & Eckhardt, K. Lack of effect of lacosamide on the pharmacokinetic and pharmacodynamic profiles of warfarin. Epilepsia 54, 1161–1166 (2013).
    1. Walker, G. , Mandagere, A. , Dufton, C. & Venitz, J. The pharmacokinetics and pharmacodynamics of warfarin in combination with ambrisentan in healthy volunteers. Br. J. Clin. Pharmacol. 67, 527–534 (2009).
    1. Coumadin (warfarin sodium) tablets [prescribing information] (Bristol‐Myers Squibb Company: Princeton, NJ, 2017).
    1. Bauer, L.A . Applied Clinical Pharmacokinetics. 2nd edn. New York, NY: McGraw‐Hill Education: <> (2008). Accessed 16 May 2018.
    1. Klein, K. & Zanger, U. Pharmacogenomics of cytochrome P450 3A4: Recent progress toward the “missing heritability” problem. Front. Genet. 4, 12 (2013).
    1. de Lannoy, I.A.M. & Silverman, M. The MDR1 gene product, P‐glycoprotein, mediates the transport of the cardiac glycoside, digoxin. Biochem. Biophys. Res. Commun. 189, 551–557 (1992).
    1. Schinkel, A.H. , Wagenaar, E. , van Deemter, L. , Mol, C.A. & Borst, P. Absence of the mdr1a P‐glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J. Clin. Invest. 96, 1698–1705 (1995).
    1. Igel, S. et al Increased absorption of digoxin from the human jejunum due to inhibition of intestinal transporter‐mediated efflux. Clin. Pharmacokinet. 46, 777–785 (2007).
    1. Gorski, J.C. , Jones, D.R. , Haehner‐Daniels, B.D. , Hamman, M.A. , O'Mara, E.M. & Hall, S.D. The contribution of intestinal and hepatic CYP3A to the interaction between midazolam and clarithromycin. Clin. Pharmacol. Ther. 64, 133–143 (1998).
    1. Drew, Y. et al Phase 2 multicentre trial investigating intermittent and continuous dosing schedules of the poly(ADP‐ribose) polymerase inhibitor rucaparib in germline BRCA mutation carriers with advanced ovarian and breast cancer. Br. J. Cancer 114, 723–730 (2016).

Source: PubMed

Подписаться