The Effect of Hepatic Impairment on the Pharmacokinetics of Dacomitinib

Joseph Piscitelli, Joseph Chen, Robert R LaBadie, Joanne Salageanu, Chin-Hee Chung, Weiwei Tan, Joseph Piscitelli, Joseph Chen, Robert R LaBadie, Joanne Salageanu, Chin-Hee Chung, Weiwei Tan

Abstract

Background and objective: Dacomitinib is a kinase inhibitor indicated for the first-line treatment of patients with metastatic non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR)-activating mutations. To evaluate the effect of hepatic impairment on the pharmacokinetics of dacomitinib, two dedicated studies were conducted to inform optimal dosing.

Methods: Study 1 (NCT01571388) evaluated the effect of mild and moderate hepatic impairment on the plasma pharmacokinetics, safety, and tolerability after a single oral dose of dacomitinib 30 mg, and Study 2 (NCT03865446) evaluated the same endpoints in a severe hepatic impairment population. Both studies were phase I, open-label, parallel-group studies. A one-way analysis of variance (ANOVA) with unequal variance assumption and hepatic impairment group as a fixed effect was used to compare the natural log of area under the plasma concentration-time curve extrapolated to infinite time (AUCinf), AUC from time zero to the last quantifiable concentration (AUClast), and maximum plasma concentration (Cmax) for each hepatic impairment group to the respective normal hepatic function group. Since dacomitinib is a cytochrome P450 (CYP) 2D6 substrate, only participants with extensive or intermediate CYP2D6 phenotypes were included in the primary analysis.

Results: The AUCinf for participants with mild, moderate, or severe hepatic impairment decreased by 6%, decreased by 23%, and increased by 4%, respectively, compared with normal hepatic function, while the Cmax for participants with mild, moderate, or severe hepatic impairment increased by 3%, decreased by 20%, and increased by 31%, respectively, compared with normal hepatic function. A single oral dose of dacomitinib 30 mg was well tolerated in all participants.

Conclusion: Based on these pharmacokinetic results, dacomitinib pharmacokinetics of participants with mild, moderate, or severe hepatic impairment were not statistically different relative to participants with normal hepatic function based on the ANOVA analysis. No dacomitinib dose adjustments for patients with hepatic impairment are recommended.

Clinical trial registration: ClinicalTrials.gov NCT01571388, registered 5 April 2012; ClinicalTrials.gov NCT03865446, registered 6 March 2019.

Conflict of interest statement

This research was sponsored by Pfizer Inc. Joseph Chen, Robert R. LaBadie, Joanne Salageanu, Chin-Hee Chung, and Weiwei Tan are employees of Pfizer Inc. and may own Pfizer stock. Joseph Piscitelli (Postdoctoral Fellow) was an unpaid contractor to Pfizer; the fellowship program with the University of California San Diego was supported by an educational grant from Pfizer.

© 2022. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.

Figures

Fig. 1
Fig. 1
Median concentrations for dacomitinib and PF-05199265 for Study 1 and Study 2 (linear and semi-log). Concentrations that were below the LLOQ were set as LLOQ/2, 0.05 ng/mL. The vertical dashed error bars represent the 5th and 95th percentiles around the median concentrations. LLOQ lower limit of quantification
Fig. 2
Fig. 2
Boxplots for AUCinf and Cmax for Study 1 and Study 2. The red diamond represents the geometric mean value for each respective group; the black line represents the median value; black circles represent the actual parameters; and the upper and lower hinges represent the 25th and 75th percentiles. The lower whisker extends to the smallest value, no less than 1.5 times the interquartile range, and the upper whisker extends to the largest value, no greater than 1.5 times the interquartile range. Black dots outside of the whiskers represent outliers. AUCinf area under the plasma concentration–time curve from time zero extrapolated to infinite time, Cmax maximum plasma concentration

References

    1. Fukuoka M, Wu YL, Thongprasert S, Sunpaweravong P, Leong SS, Sriuranpong V, et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS) J Clin Oncol. 2011;29:2866–2874. doi: 10.1200/JCO.2010.33.4235.
    1. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380–2388. doi: 10.1056/NEJMoa0909530.
    1. Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–128. doi: 10.1016/S1470-2045(09)70364-X.
    1. Sequist LV, Yang JC, Yamamoto N, O'Byrne K, Hirsh V, Mok T, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31:3327–3334. doi: 10.1200/JCO.2012.44.2806.
    1. Wu YL, Zhou C, Hu CP, Feng J, Lu S, Huang Y, et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): an open-label, randomised phase 3 trial. Lancet Oncol. 2014;15:213–222. doi: 10.1016/S1470-2045(13)70604-1.
    1. Zhou C, Wu YL, Chen G, Feng J, Liu XQ, Wang C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12:735–742. doi: 10.1016/s1470-2045(11)70184-x.
    1. Mok TS, Wu YL, Thongprasert S, Yang CH, Chu DT, Saijo N, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947–957. doi: 10.1056/NEJMoa0810699.
    1. Engelman JA, Zejnullahu K, Gale CM, Lifshits E, Gonzales AJ, Shimamura T, et al. PF00299804, an irreversible pan-ERBB inhibitor, is effective in lung cancer models with EGFR and ERBB2 mutations that are resistant to gefitinib. Cancer Res. 2007;67:11924–11932. doi: 10.1158/0008-5472.CAN-07-1885.
    1. Wu Y-L, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:1454–1466. doi: 10.1016/s1470-2045(17)30608-3.
    1. Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Improvement in overall survival in a randomized study that compared dacomitinib with gefitinib in patients with advanced non-small-cell lung cancer and EGFR-activating mutations. J Clin Oncol. 2018;36:2244–2250. doi: 10.1200/JCO.2018.78.7994.
    1. Mok TS, Cheng Y, Zhou X, Lee KH, Nakagawa K, Niho S, et al. Updated overall survival in a randomized study comparing dacomitinib with gefitinib as first-Line treatment in patients with advanced non-small-cell lung cancer and EGFR-activating mutations. Drugs. 2021;81:257–266. doi: 10.1007/s40265-020-01441-6.
    1. Food and Drug Administration (FDA). VIZIMPRO® (dacomitinib) prescribing information. 2018. . Accessed 10 Dec 2021.
    1. Bello CL, Smith E, Ruiz-Garcia A, Ni G, Alvey C, Loi CM. A phase I, open-label, mass balance study of [(14)C] dacomitinib (PF-00299804) in healthy male volunteers. Cancer Chemother Pharmacol. 2013;72:379–385. doi: 10.1007/s00280-013-2207-9.
    1. Rodighiero V. Effects of liver disease on pharmacokinetics. An update. Clin Pharmacokinet. 1999;37:399–431. doi: 10.2165/00003088-199937050-00004.
    1. Giri N, Masters JC, Plotka A, Liang Y, Boutros T, Pardo P, et al. Investigation of the impact of hepatic impairment on the pharmacokinetics of dacomitinib. Invest New Drugs. 2015;33:931–941. doi: 10.1007/s10637-015-0256-0.
    1. Gaedigk A, Ingelman-Sundberg M, Miller NA, Leeder JS, Whirl-Carrillo M, Klein TE, et al. The Pharmacogene Variation (PharmVar) Consortium: incorporation of the Human Cytochrome P450 (CYP) Allele Nomenclature Database. Clin Pharmacol Ther. 2018;103:399–401. doi: 10.1002/cpt.910.
    1. Whirl-Carrillo M, McDonagh EM, Hebert JM, Gong L, Sangkuhl K, Thorn CF, et al. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2012;92:414–417. doi: 10.1038/clpt.2012.96.
    1. US Food and Drug Administration (FDA). Pharmacokinetics in Patients with Impaired Hepatic Function: Study Design, Data Analysis, and Impact on Dosing and Labeling. 2003. . Accessed 10 Dec 2021.
    1. Fostvedt L, Nickens D, Tan W. Abstract 2103: Application of longitudinal exposure-response modelling to support dacomitinib starting dose in patients with EGFR mutation-positive NSCLC. Worldwide Virtual Event. World Conference on Lung Cancer; 28–31 January 2021.
    1. Tan W, Giri N, Quinn S, Wilner K, Parivar K. Evaluation of the potential effect of dacomitinib, an EGFR tyrosine kinase inhibitor, on ECG parameters in patients with advanced non-small cell lung cancer. Invest New Drugs. 2020;38:874–884. doi: 10.1007/s10637-019-00887-0.
    1. Blaschke TF. Protein binding and kinetics of drugs in liver diseases. Clin Pharmacokinet. 1977;2:32–44. doi: 10.2165/00003088-197702010-00003.
    1. Shah R, Lester JF. Tyrosine kinase inhibitors for the treatment of EGFR mutation-positive non-small-cell lung cancer: a clash of the generations. Clin Lung Cancer. 2020;21:e216–e228. doi: 10.1016/j.cllc.2019.12.003.
    1. US Food and Drug Administration (FDA). IRESSA® (gefitinib) prescribing information. 2018. . Accessed 10 Dec 2021.
    1. Li J, Zhao M, He P, Hidalgo M, Baker SD. Differential metabolism of gefitinib and erlotinib by human cytochrome P450 enzymes. Clin Cancer Res. 2007;13:3731–3737. doi: 10.1158/1078-0432.CCR-07-0088.
    1. Horak J, White J, Harris AL, Verrill M, Carmichael J, Holt A, et al. The effect of different etiologies of hepatic impairment on the pharmacokinetics of gefitinib. Cancer Chemother Pharmacol. 2011;68:1485–1495. doi: 10.1007/s00280-011-1611-2.
    1. US Food and Drug Administration (FDA). TARCEVA® (erlotinib) prescribing information. 2016. . Accessed 10 Dec 2021.
    1. O'Bryant CL, Haluska P, Rosen L, Ramanathan RK, Venugopal B, Leong S, et al. An open-label study to describe pharmacokinetic parameters of erlotinib in patients with advanced solid tumors with adequate and moderately impaired hepatic function. Cancer Chemother Pharmacol. 2012;69:605–612. doi: 10.1007/s00280-011-1733-6.
    1. US Food and Drug Administration (FDA). TAGRISSO™ (osimertinib) prescribing information. . Accessed 10 Dec 2021.
    1. Grande E, Harvey RD, You B, Batlle JF, Galbraith H, Sarantopoulos J, et al. Pharmacokinetic study of osimertinib in cancer patients with mild or moderate hepatic impairment. J Pharmacol Exp Ther. 2019;369:291–299. doi: 10.1124/jpet.118.255919.
    1. Peters S, Zimmermann S, Adjei AA. Oral epidermal growth factor receptor tyrosine kinase inhibitors for the treatment of non-small cell lung cancer: comparative pharmacokinetics and drug-drug interactions. Cancer Treat Rev. 2014;40:917–926. doi: 10.1016/j.ctrv.2014.06.010.
    1. Alfieri RR, Galetti M, Tramonti S, Andreoli R, Mozzoni P, Cavazzoni A, et al. Metabolism of the EGFR tyrosin kinase inhibitor gefitinib by cytochrome P450 1A1 enzyme in EGFR-wild type non small cell lung cancer cell lines. Mol Cancer. 2011;10:143. doi: 10.1186/1476-4598-10-143.
    1. Dickinson PA, Cantarini MV, Collier J, Frewer P, Martin S, Pickup K, et al. Metabolic disposition of osimertinib in rats, dogs, and humans: insights into a drug designed to bind covalently to a cysteine residue of epidermal growth factor receptor. Drug Metab Dispos. 2016;44:1201–1212. doi: 10.1124/dmd.115.069203.

Source: PubMed

3
Předplatit