Evaluation of Safety and Clinically Relevant Drug-Drug Interactions with Tucatinib in Healthy Volunteers

Ariel Topletz-Erickson, Anthony Lee, Evelyn L Rustia, Hao Sun, JoAl G Mayor, Layth I Abdulrasool, Luke Walker, Christopher J Endres, Ariel Topletz-Erickson, Anthony Lee, Evelyn L Rustia, Hao Sun, JoAl G Mayor, Layth I Abdulrasool, Luke Walker, Christopher J Endres

Abstract

Background and objective: Tucatinib is approved for treatment of human epidermal growth factor receptor 2-positive metastatic breast cancer. Understanding potential drug-drug interactions (DDIs) informs proper dosing when co-administering tucatinib with other therapies. The aim of this study was to evaluate DDIs between tucatinib and metabolizing enzymes and transporters in healthy volunteers.

Methods: Parts A-C assessed the impact of itraconazole (cytochrome P450 [CYP] 3A4 inhibitor), rifampin (CYP3A4/CYP2C8 inducer), or gemfibrozil (CYP2C8 inhibitor) on the pharmacokinetics of a single 300 mg dose of tucatinib administered orally and its primary metabolite, ONT-993. Parts D and E assessed the effect of steady-state tucatinib on the pharmacokinetics of repaglinide (CYP2C8 substrate), tolbutamide (CYP2C9 substrate), midazolam (CYP3A4 substrate), and digoxin (P-glycoprotein substrate).

Results: Tucatinib area under the concentration-time curve from time 0 extrapolated to infinity (AUC0-inf) increased by ~ 1.3- and 3.0-fold with itraconazole and gemfibrozil, respectively, and decreased by 48% with rifampin, indicating that tucatinib is metabolized primarily by CYP2C8, and to a lesser extent via CYP3A. Tucatinib was a strong inhibitor of CYP3A (midazolam AUC0-inf increased 5.7-fold), a weak inhibitor of CYP2C8 and P-glycoprotein, and had no impact on CYP2C9-mediated metabolism in humans. Tucatinib was well tolerated, alone and with co-administered drugs.

Conclusion: The potential DDIs identified here may be mitigated by avoiding concomitant use of tucatinib with strong CYP3A inducers, moderate CYP2C8 inducers, CYP3A substrates with a narrow therapeutic window (modifying substrate dose where concomitant use is unavoidable), and strong CYP2C8 inhibitors (decreasing tucatinib dose where concomitant use is unavoidable), or by reducing the dose of P-glycoprotein substrates with a narrow therapeutic window.

Trial registration: This trial (NCT03723395) was registered on October 29, 2018.

Conflict of interest statement

This study was funded by Seagen Inc. The authors wrote the article with the assistance of a medical writer funded by the sponsor. All the authors had full access to the relevant data, vouch for the completeness and accuracy of the data and for adherence of the trial to the protocol, assume final responsibility for the content of the article, and for the decision to submit the article for publication. A. Topletz-Erickson, A. Lee, E.L. Rustia, H. Sun, J.G. Mayor, L.I. Abdulrasool, L. Walker, and C.J. Endres are employees of Seagen and hold stocks and shares in Seagen. No authors are Fellows of the American College of Clinical Pharmacology.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Study schematics for assessing A the effect of comedications on tucatinib plasma exposure, and B the effect of steady-state tucatinib on the plasma exposure of co-administered drugs. BID twice daily, D day, ITZ itraconazole, MDZ midazolam, PK pharmacokinetics, QD once daily, RIF rifampin, TUC tucatinib
Fig. 2
Fig. 2
Tucatinib plasma concentration profiles (A, D, G), AUC0–inf values (B, E, H), and Cmax values (C, F, I) in the absence and presence of steady-state itraconazole (A–C), rifampin (D–F), or gemfibrozil (G–I). Solid lines inside the box plots represent the median. The boxes represent the 25th and 75th percentile and the whiskers represent the minimum and maximum values. AUC0–inf area under the concentration–time curve from time 0 extrapolated to infinity, Cmax maximum observed concentration, CYP cytochrome P450, GEM gemfibrozil, ITZ itraconazole, RIF rifampin
Fig. 3
Fig. 3
Plasma concentration profiles, AUC0–inf values, and Cmax values of midazolam (A–C), repaglinide (D–F), tolbutamide (G–I), or digoxin (J–L) in the absence and presence of steady-state tucatinib. Solid lines inside the box plots represent the median. The boxes represent the 25th and 75th percentile and the whiskers represent the minimum and maximum values. AUC0–inf area under the concentration–time curve from time 0 extrapolated to infinity, Cmax maximum observed concentration, CYP cytochrome P450, MDZ midazolam, P-gp P-glycoprotein, TUC tucatinib

References

    1. Siena S, Sartore-Bianchi A, Marsoni S, Hurwitz HI, McCall SJ, Penault-Llorca F, et al. Targeting the human epidermal growth factor receptor 2 (HER2) oncogene in colorectal cancer. Ann Oncol. 2018;29(5):1108–1119. doi: 10.1093/annonc/mdy100.
    1. Cardoso F, Paluch-Shimon S, Senkus E, Curigliano G, Aapro MS, Andre F, et al. 5th ESO-ESMO international consensus guidelines for advanced breast cancer (ABC 5) Ann Oncol. 2020;31(12):1623–1649. doi: 10.1016/j.annonc.2020.09.010.
    1. Choong GM, Cullen GD, O'Sullivan CC. Evolving standards of care and new challenges in the management of HER2-positive breast cancer. CA Cancer J Clin. 2020;70(5):355–374. doi: 10.3322/caac.21634.
    1. Murthy RK, Loi S, Okines A, Paplomata E, Hamilton E, Hurvitz SA, et al. Tucatinib, trastuzumab, and capecitabine for HER2-positive metastatic breast cancer. N Engl J Med. 2020;382(7):597–609. doi: 10.1056/NEJMoa1914609.
    1. Seagen Inc. Tukysa (tucatinib) US prescribing information. 2020. . Accessed 18 Jan 2022.
    1. Seagen Inc. Tukysa (tucatinib) European summary of product characteristics. 2021. . Accessed 18 Jan 2022.
    1. Strickler JH, Zemla T, Ou FS, Cercek A, Wu C, Sanchez FA, et al. Trastuzumab and tucatinib for the treatment of HER2 amplified metastatic colorectal cancer (mCRC): Initial results from the MOUNTAINEER trial. Ann Oncol. 2019;30:v200. doi: 10.1093/annonc/mdz246.005.
    1. Food and Drug Administration. Clinical drug interaction studies—cytochrome P450 enzyme- and transporter-mediated drug interactions: guidance for industry. 2020. . Accessed 18 Jan 2022.
    1. Food and Drug Administration. Multi-disciplinary review and evaluation-NDA 213411 (tucatinib). 2019. . Accessed 18 Jan 2022.
    1. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013;138(1):103–141. doi: 10.1016/j.pharmthera.2012.12.007.
    1. Topletz-Erickson AR, Lee AJ, Mayor JG, Rustia EL, Abdulrasool LI, Wise AL, et al. Tucatinib inhibits renal transporters OCT2 and MATE without impacting renal function in healthy subjects. J Clin Pharmacol. 2021;61(4):461–471. doi: 10.1002/jcph.1750.
    1. Metzger Filho O, Leone JP, Li T, Tan-Wasielewski Z, Trippa L, Barry WT, et al. Phase I dose-escalation trial of tucatinib in combination with trastuzumab in patients with HER2-positive breast cancer brain metastases. Ann Oncol. 2020;31(9):1231–1239. doi: 10.1016/j.annonc.2020.05.014.
    1. Murthy R, Borges VF, Conlin A, Chaves J, Chamberlain M, Gray T, et al. Tucatinib with capecitabine and trastuzumab in advanced HER2-positive metastatic breast cancer with and without brain metastases: a non-randomised, open-label, phase 1b study. Lancet Oncol. 2018;19(7):880–888. doi: 10.1016/S1470-2045(18)30256-0.
    1. Borges VF, Ferrario C, Aucoin N, Falkson C, Khan Q, Krop I, et al. Tucatinib combined with ado-trastuzumab emtansine in advanced ERBB2/HER2-positive metastatic breast cancer: a phase 1b clinical trial. JAMA Oncol. 2018;4(9):1214–1220. doi: 10.1001/jamaoncol.2018.1812.
    1. Moulder SL, Borges VF, Baetz T, McSpadden T, Fernetich G, Murthy RK, et al. Phase I study of ONT-380, a HER2 inhibitor, in patients with HER2(+)-advanced solid tumors, with an expansion cohort in HER2(+) metastatic breast cancer (MBC) Clin Cancer Res. 2017;23(14):3529–3536. doi: 10.1158/1078-0432.CCR-16-1496.
    1. Sun H, Cardinal K, Wienkers L, Chin A, Kumar V, Neace C, et al. Elimination of tucatinib, a small molecule kinase inhibitor of HER2, is primarily governed by CYP2C8 enantioselective oxidation of gem dimethyl. Cancer Chemother Pharmacol. 2022;89(6):737–50.
    1. Yu J, Wang Y, Ragueneau-Majlessi I. Pharmacokinetic drug-drug interactions with drugs approved by the U.S. Food and Drug Administration in 2020: mechanistic understanding and clinical recommendations. Drug Metab Dispos. 2021;50(1):1–7.
    1. Sanico NV. Itroconazole US prescribing information. 2010. . Accessed 5 Apr 2022.
    1. Sanofi. Rifadin (rifampin) US prescribing infomration. 2010. . Accessed 22 Mar 2022.
    1. Inc NN. Prandin (repaglinide) US prescribing information. 2008. . Accessed 22 Mar 2022.
    1. Pfizer. Lopid (gemfibrozil) US prescribing information. 2020. . Accessed 22 Mar 2022.
    1. Concordia Pharmaceuticals Inc. Lanoxin (digoxin prescribing infomration). 2016. . Accessed 22 Mar 2022.
    1. Liu L, Bello A, Dresser MJ, Heald D, Komjathy SF, O'Mara E, et al. Best practices for the use of itraconazole as a replacement for ketoconazole in drug-drug interaction studies. J Clin Pharmacol. 2016;56(2):143–151. doi: 10.1002/jcph.562.
    1. Blakey GE, Lockton JA, Perrett J, Norwood P, Russell M, Aherne Z, et al. Pharmacokinetic and pharmacodynamic assessment of a five-probe metabolic cocktail for CYPs 1A2, 3A4, 2C9, 2D6 and 2E1. Br J Clin Pharmacol. 2004;57(2):162–169. doi: 10.1046/j.1365-2125.2003.01973.x.

Source: PubMed

3
订阅