Effects of Strong CYP3A Inhibition and Induction on the Pharmacokinetics of Ixazomib, an Oral Proteasome Inhibitor: Results of Drug-Drug Interaction Studies in Patients With Advanced Solid Tumors or Lymphoma and a Physiologically Based Pharmacokinetic Analysis

Neeraj Gupta, Michael J Hanley, Karthik Venkatakrishnan, Alberto Bessudo, Drew W Rasco, Sunil Sharma, Bert H O'Neil, Bingxia Wang, Guohui Liu, Alice Ke, Chirag Patel, Karen Rowland Yeo, Cindy Xia, Xiaoquan Zhang, Dixie-Lee Esseltine, John Nemunaitis, Neeraj Gupta, Michael J Hanley, Karthik Venkatakrishnan, Alberto Bessudo, Drew W Rasco, Sunil Sharma, Bert H O'Neil, Bingxia Wang, Guohui Liu, Alice Ke, Chirag Patel, Karen Rowland Yeo, Cindy Xia, Xiaoquan Zhang, Dixie-Lee Esseltine, John Nemunaitis

Abstract

At clinically relevant ixazomib concentrations, in vitro studies demonstrated that no specific cytochrome P450 (CYP) enzyme predominantly contributes to ixazomib metabolism. However, at higher than clinical concentrations, ixazomib was metabolized by multiple CYP isoforms, with the estimated relative contribution being highest for CYP3A at 42%. This multiarm phase 1 study (Clinicaltrials.gov identifier: NCT01454076) investigated the effect of the strong CYP3A inhibitors ketoconazole and clarithromycin and the strong CYP3A inducer rifampin on the pharmacokinetics of ixazomib. Eighty-eight patients were enrolled across the 3 drug-drug interaction studies; the ixazomib toxicity profile was consistent with previous studies. Ketoconazole and clarithromycin had no clinically meaningful effects on the pharmacokinetics of ixazomib. The geometric least-squares mean area under the plasma concentration-time curve from 0 to 264 hours postdose ratio (90%CI) with vs without ketoconazole coadministration was 1.09 (0.91-1.31) and was 1.11 (0.86-1.43) with vs without clarithromycin coadministration. Reduced plasma exposures of ixazomib were observed following coadministration with rifampin. Ixazomib area under the plasma concentration-time curve from time 0 to the time of the last quantifiable concentration was reduced by 74% (geometric least-squares mean ratio of 0.26 [90%CI 0.18-0.37]), and maximum observed plasma concentration was reduced by 54% (geometric least-squares mean ratio of 0.46 [90%CI 0.29-0.73]) in the presence of rifampin. The clinical drug-drug interaction study results were reconciled well by a physiologically based pharmacokinetic model that incorporated a minor contribution of CYP3A to overall ixazomib clearance and quantitatively considered the strength of induction of CYP3A and intestinal P-glycoprotein by rifampin. On the basis of these study results, the ixazomib prescribing information recommends that patients should avoid concomitant administration of strong CYP3A inducers with ixazomib.

Keywords: CYP3A; PBPK modeling; drug-drug interaction; ixazomib; multiple myeloma; pharmacokinetics.

© 2017, 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
DDI study designs: study treatment and PK sampling during the PK cycle of the DDI study arms for (A) ketoconazole, (B) clarithromycin, and (C) rifampin. DDI indicates drug‐drug interaction; PK, pharmacokinetics.
Figure 2
Figure 2
Mean (± SE) plasma ixazomib concentration‐time profiles (with insets showing the first 24 hours after dosing) with and without coadministration of (A) clarithromycin or (B) rifampin.
Figure 3
Figure 3
Physiologically based pharmacokinetic model‐predicted and observed mean plasma concentration‐time profiles for (A) ixazomib after oral administration of 2.5 mg; (B) ixazomib 2.5 mg with and without clarithromycin coadministration; and (C) ixazomib 4 mg with and without rifampin coadministration. (A) The gray lines represent the outcomes of simulated individual trials (10 trials each containing 16 patients). The solid black line represents the mean concentration‐time data for the simulated population (N = 160 patients). The open circles represent the observed mean concentration‐time data after day 1 administration of ixazomib in the ketoconazole DDI study. (B) Simulated (black lines; 10 trials each containing 16 patients) and observed (circles; data from the clarithromycin DDI study) mean plasma concentration‐time profiles of ixazomib after a single oral dose of 2.5 mg in the presence (dashed black line, filled circles) and absence (solid black line, open circles) of multiple daily doses of clarithromycin (500 mg twice daily for 16 days). The solid/dashed black lines represent the mean concentration‐time data for the simulated population (N = 160 patients). The gray lines represent the outcomes of simulated individual trials. (C) Simulated (black lines; 10 trials each containing 16 patients) and observed (circles; data from the rifampin DDI study) mean plasma concentration‐time profiles of ixazomib after a single oral dose of 4 mg in the presence (dashed black line, filled circles) and absence (solid black line, open circles) of multiple daily doses of rifampin (600 mg daily for 14 days). The solid/dashed black lines represent the mean concentration‐time data for the simulated population (N = 160 patients). The gray lines represent the outcomes of simulated individual trials. DDI indicates drug‐drug interaction.
Figure 4
Figure 4
Physiologically based pharmacokinetic model‐predicted and observed geometric least‐squares mean AUC ratios for ixazomib with and without various strong CYP3A inhibitors and strong CYP3A inducers. For predicted data, error bars represent the 5th and 95th percentiles. AUC indicates area under the concentration‐time curve; CYP, cytochrome P450.

References

    1. Millennium Pharmaceuticals Inc . NINLARO® (ixazomib) capsules, for oral use. Prescribing information, November 2016. . Accessed April 13, 2017.
    1. Takeda Pharma A/S. NINLARO® European Public Assessment Report—Product Information . . Accessed December 15, 2016.
    1. Moreau P, Masszi T, Grzasko N, et al. Oral ixazomib, lenalidomide, and dexamethasone for multiple myeloma. N Engl J Med. 2016;374(17):1621–1634.
    1. Kumar SK, Bensinger WI, Zimmerman TM, et al. Phase 1 study of weekly dosing with the investigational oral proteasome inhibitor ixazomib in relapsed/refractory multiple myeloma. Blood. 2014;124(7):1047–1055.
    1. Richardson PG, Baz R, Wang M, et al. Phase 1 study of twice‐weekly ixazomib, an oral proteasome inhibitor, in relapsed/refractory multiple myeloma patients. Blood. 2014;124(7):1038–1046.
    1. Gupta N, Zhao Y, Hui AM, Esseltine DL, Venkatakrishnan K. Switching from body surface area‐based to fixed dosing for the investigational proteasome inhibitor ixazomib: a population pharmacokinetic analysis. Br J Clin Pharmacol. 2014;79(5):789–800.
    1. Gupta N, Hanley MJ, Venkatakrishnan K, et al. The effect of a high‐fat meal on the pharmacokinetics of ixazomib, an oral proteasome inhibitor, in patients with advanced solid tumors or lymphoma. J Clin Pharmacol. 2016;56(10):1288–1295.
    1. Gupta N, Hanley MJ, Harvey RD, et al. A pharmacokinetics and safety phase 1/1b study of oral ixazomib in patients with multiple myeloma and severe renal impairment or end‐stage renal disease requiring haemodialysis. Br J Haematol. 2016;174(5):748–759.
    1. Gupta N, Hanley MJ, Venkatakrishnan K, et al. Pharmacokinetics of ixazomib, an oral proteasome inhibitor, in solid tumour patients with moderate or severe hepatic impairment. Br J Clin Pharmacol. 2016;82(3):728–738.
    1. Gupta N, Diderichsen PM, Hanley MJ, et al. Population pharmacokinetic analysis of ixazomib, an oral proteasome inhibitor, including data from the phase III TOURMALINE‐MM1 study to inform labelling. Clin Pharmacokinet. e‐pub ahead of print; . 2017.
    1. Gupta N, Huh Y, Hutmacher MM, et al. Integrated nonclinical and clinical risk assessment of the investigational proteasome inhibitor ixazomib on the QTc interval in cancer patients. Cancer Chemother Pharmacol. 2015;76(3):507–516.
    1. Gupta N, Zhang S, Pusalkar S, et al. A phase 1 mass balance study of ixazomib, an oral proteasome inhibitor (PI), using accelerator mass spectrometry (AMS) in patients with advanced solid tumors [abstract]. Clin Pharmacol Ther. 2017;101:Abstract 615.
    1. Hanley MJ, Gupta N, Venkatakrishnan K, et al. Phase I study to assess the relative bioavailability of two capsule formulations of ixazomib, an oral proteasome inhibitor, in patients with advanced solid tumors or lymphoma [abstract]. Clin Pharmacol Ther. 2016;99:Abstract PII‐023.
    1. Kishimoto W, Ishiguro N, Ludwig‐Schwellinger E, Ebner T, Schaefer O. In vitro predictability of drug‐drug interaction likelihood of P‐glycoprotein‐mediated efflux of dabigatran etexilate based on [I]2/IC50 threshold. Drug Metab Dispos. 2014;42(2):257–263.
    1. Yamashita F, Sasa Y, Yoshida S, et al. Modeling of rifampicin‐induced CYP3A4 activation dynamics for the prediction of clinical drug‐drug interactions from in vitro data. PLoS One. 2013;8(9):e70330.
    1. Greiner B, Eichelbaum M, Fritz P, et al. The role of intestinal P‐glycoprotein in the interaction of digoxin and rifampin. J Clin Invest. 1999;104(2):147–153.
    1. Westphal K, Weinbrenner A, Zschiesche M, et al. Induction of P‐glycoprotein by rifampin increases intestinal secretion of talinolol in human beings: a new type of drug/drug interaction. Clin Pharmacol Ther. 2000;68(4):345–355.
    1. European Medicines Agency Committee for Human Medicinal Products . Guideline on the investigation of drug interactions. . Accessed February 15, 2017.
    1. Gorski JC, Jones DR, Haehner‐Daniels BD, et al. The contribution of intestinal and hepatic CYP3A to the interaction between midazolam and clarithromycin. Clin Pharmacol Ther. 1998;64(2):133–143.
    1. Gurley B, Hubbard MA, Williams DK, et al. Assessing the clinical significance of botanical supplementation on human cytochrome P450 3A activity: comparison of a milk thistle and black cohosh product to rifampin and clarithromycin. J Clin Pharmacol. 2006;46(2):201–213.
    1. Quinney SK, Haehner BD, Rhoades MB, et al. Interaction between midazolam and clarithromycin in the elderly. Br J Clin Pharmacol. 2008;65(1):98–109.
    1. US Food and Drug Administration Center for Drug Evaluation and Research (CDER) . Guidance for Industry—Drug Interaction Studies—Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations. . Accessed February 15, 2017.
    1. Yeates RA, Laufen H, Zimmermann T. Interaction between midazolam and clarithromycin: comparison with azithromycin. Int J Clin Pharmacol Ther. 1996;34(9):400–405.
    1. Gay F, Rajkumar SV, Coleman M, et al. Clarithromycin (Biaxin)‐lenalidomide‐low‐dose dexamethasone (BiRd) versus lenalidomide‐low‐dose dexamethasone (Rd) for newly diagnosed myeloma. Am J Hematol. 2010;85(9):664–669.
    1. Niesvizky R, Jayabalan DS, Christos PJ, et al. BiRD (Biaxin [clarithromycin]/Revlimid [lenalidomide]/dexamethasone) combination therapy results in high complete‐ and overall‐response rates in treatment‐naive symptomatic multiple myeloma. Blood. 2008;111(3):1101–1109.
    1. Backman JT, Olkkola KT, Neuvonen PJ. Rifampin drastically reduces plasma concentrations and effects of oral midazolam. Clin Pharmacol Ther. 1996;59(1):7–13.
    1. Gorski JC, Vannaprasaht S, Hamman MA, et al. The effect of age, sex, and rifampin administration on intestinal and hepatic cytochrome P450 3A activity. Clin Pharmacol Ther. 2003;74(3):275–287.
    1. Blassmann U, Roehr AC, Frey OR, et al. Decreased linezolid serum concentrations in three critically ill patients: clinical case studies of a potential drug interaction between linezolid and rifampicin. Pharmacology. 2016;98(1‐2):51–55.
    1. Hoyo I, Martinez‐Pastor J, Garcia‐Ramiro S, et al. Decreased serum linezolid concentrations in two patients receiving linezolid and rifampicin due to bone infections. Scand J Infect Dis. 2012;44(7):548–550.
    1. Self T, Corley CR, Nabhan S, Abell T. Case report: interaction of rifampin and nortriptyline. Am J Med Sci. 1996;311(2):80–81.
    1. von Bahr C, Steiner E, Koike Y, Gabrielsson J. Time course of enzyme induction in humans: effect of pentobarbital on nortriptyline metabolism. Clin Pharmacol Ther. 1998;64(1):18–26.
    1. Venkatakrishnan K, von Moltke LL, Greenblatt DJ. Nortriptyline E‐10‐hydroxylation in vitro is mediated by human CYP2D6 (high affinity) and CYP3A4 (low affinity): implications for interactions with enzyme‐inducing drugs. J Clin Pharmacol. 1999;39(6):567–577.
    1. Gandelman K, Zhu T, Fahmi OA, et al. Unexpected effect of rifampin on the pharmacokinetics of linezolid: in silico and in vitro approaches to explain its mechanism. J Clin Pharmacol. 2011;51(2):229–236.
    1. Cotreau MM, Siebers NM, Miller J, Strahs AL, Slichenmyer W. Effects of ketoconazole or rifampin on the pharmacokinetics of tivozanib hydrochloride, a vascular endothelial growth factor receptor tyrosine kinase inhibitor. Clin Pharmacol Drug Dev. 2015;4(2):137–142.
    1. Venkatakrishnan K. DDI risk assessment and evaluation in clinical development: interfacing drug metabolism and clinical pharmacology In: Lyubimov A, ed. Encyclopedia of Drug Metabolism and Interactions. Hoboken, NJ: John Wiley & Sons; 2012. . Accessed February 15, 2017.
    1. Venkatakrishnan K, Pickard MD, von Moltke LL. A quantitative framework and strategies for management and evaluation of metabolic drug‐drug interactions in oncology drug development: new molecular entities as object drugs. Clin Pharmacokinet. 2010;49(11):703–727.
    1. Gupta N, Goh YT, Min CK, et al. Pharmacokinetics and safety of ixazomib plus lenalidomide‐dexamethasone in Asian patients with relapsed/refractory myeloma: a phase 1 study. J Hematol Oncol. 2015;8:103.
    1. Kumar SK, Berdeja JG, Niesvizky R, et al. Safety and tolerability of ixazomib, an oral proteasome inhibitor, in combination with lenalidomide and dexamethasone in patients with previously untreated multiple myeloma: an open‐label phase 1/2 study. Lancet Oncol. 2014;15(13):1503–1512.
    1. Merlini G, Sanchorawala V, Zonder JA, et al. Long‐term outcome of a phase 1 study of the investigational oral proteasome inhibitor (PI) ixazomib at the recommended phase 3 dose (RP3D) in patients (pts) with relapsed or refractory systemic light‐chain (AL) amyloidosis (RRAL) [abstract]. Blood. 2014;124:3450.
    1. Battisti WP, Wager E, Baltzeret L, et al. Good publication practice for communicating company‐sponsored medical research: GPP3. Ann Intern Med. 2015;163:461–464.

Source: PubMed

3
Se inscrever