Population pharmacokinetic analysis of tepotinib, an oral MET kinase inhibitor, including data from the VISION study

Wenyuan Xiong, Orestis Papasouliotis, E Niclas Jonsson, Rainer Strotmann, Pascal Girard, Wenyuan Xiong, Orestis Papasouliotis, E Niclas Jonsson, Rainer Strotmann, Pascal Girard

Abstract

Purpose: Tepotinib is a highly selective, potent, mesenchymal-epithelial transition factor (MET) inhibitor, approved for the treatment of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping. Objectives of this population pharmacokinetic (PK) analysis were to evaluate the dose-exposure relationship of tepotinib and its major circulating metabolite, MSC2571109A, and to identify the intrinsic/extrinsic factors that are predictive of PK variability.

Methods: Data were included from 12 studies in patients with cancer and in healthy participants. A sequential modeling approach was used to analyze the parent and metabolite data, including covariate analyses. Potential associations between observed covariates and PK parameters were illustrated using bootstrap analysis-based forest plots.

Results: A two-compartment model with sequential zero- and first-order absorption, and a first-order elimination from the central compartment, best described the plasma PK of tepotinib in humans across the dose range of 30-1400 mg. The bioavailability of tepotinib was shown to be dose dependent, although bioavailability decreased primarily at doses above the therapeutic dose of 500 mg. The intrinsic factors of race, age, sex, body weight, mild/moderate hepatic impairment and mild/moderate renal impairment, along with the extrinsic factors of opioid analgesic and gefitinib intake, had no relevant effect on tepotinib PK. Tepotinib has a long effective half-life of ~ 32 h.

Conclusions: Tepotinib shows dose proportionality up to at least the therapeutic dose, and time-independent clearance with a profile appropriate for once-daily dosing. None of the covariates identified had a clinically meaningful effect on tepotinib exposure or required dose adjustments.

Trial registration: ClinicalTrials.gov NCT02864992.

Keywords: MET kinase inhibitor; NSCLC; Population PK; Tepotinib.

Conflict of interest statement

Wenyuan Xiong was employed by the Merck Institute of Pharmacometrics, Lausanne, Switzerland, an affiliate of Merck KGaA for the duration of the study. Orestis Papasouliotis and Pascal Girard are employees of the Merck Institute of Pharmacometrics, Lausanne, Switzerland, an affiliate of Merck KGaA. Rainer Strotmann is an employee of Merck Healthcare KGaA, Darmstadt, Germany. E. Niclas Jonsson is an employee of Pharmetheus AB, Sweden.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Illustration of the base population pharmacokinetics model for tepotinib and MSC2571109A. CL clearance, D1 zero-order absorption duration, FM fraction of tepotinib metabolized to MSC2571109A, ka first-order absorption rate constant, met metabolite, par parent, Q inter-compartmental clearance, Vc central volume of distribution, Vp peripheral volume of distribution
Fig. 2
Fig. 2
Predicted tepotinib AUCss versus dose, with and without the estimated dose effect on Fpar. The blue line represents the relationship between AUCss and dose according to the final tepotinib model, while the grey, dashed line displays the theoretical relationship between tepotinib AUCss and dose if the dose had no impact on Fpar. AUCss area under the curve at steady state, par parent, eGFR estimated glomerular filtration rate, INR international normalized ratio, NSCLC non-small cell lung cancer, QD once daily, TF tablet formulation. Note: The prediction is for a typical patient with NSCLC (59 years, 72 kg, serum albumin = 40 g/L, eGFR = 97.28 mL/min/1.73 m2, INR = 1.06) receiving 500 mg QD tepotinib TF3 with food
Fig. 3
Fig. 3
Simulation of tepotinib PK profile for a typical patient with NSCLC (59 years, 72 kg, serum albumin = 40 g/L, eGFR = 97.28 mL/min/1.73 m2, INR = 1.06) receiving 500 mg QD tepotinib TF3 with food. The solid black line represents the median prediction of the PK time profile, and the green shaded area represents a simulation-based 5–95% prediction interval for PK time profile. eGFR estimated glomerular filtration rate, INR international normalized ratio, NSCLC non-small cell lung cancer, PK pharmacokinetics, QD once daily, TF tablet formulation
Fig. 4
Fig. 4
The distribution of tepotinib AUCss stratified by selected race categories, based on the final tepotinib population PK model and using the analysis data set. The predictions of AUCτ,ss are for Caucasian, Other East Asian and Japanese participants in the tepotinib analysis data set receiving 500 mg tepotinib with the TF1 or TF2 formulation and having a standard breakfast. The horizontal line in the box indicates the median value, the box edges represent the 25th and 75th percentiles, and the whiskers extend from the box to the furthest data points still within a distance of 1.5 times the interquartile range from the box. Data points, which are jittered in the horizontal direction, show the individually predicted AUCss values. The numbers represent the number of individuals in each strata. AUCss area under the curve at steady state, PK pharmacokinetics, TF tablet formulation
Fig. 5
Fig. 5
Forest plot showing the association of the predicted tepotinib AUCss and covariates assuming a dosing regimen of 500 mg tepotinib daily, based on the final tepotinib population PK model, for cancer patients in the analysis data set. The closed symbols represent the mean ratio of individual parameter estimates for the applicable covariate category or value (5th or 95th percentile for continuous covariates) percentile relative to the mean parameter estimate (vertical solid line) for cancer patients in the analysis dataset. The whiskers represent the 90% CI of the mean values, based on 100 bootstrap samples. AUCss area under the curve at steady state, CI confidence interval, ECOG Eastern Cooperative Oncology Group, eGFR estimated glomerular filtration rate, NCI-ODG National Cancer Institute Organ Dysfunction Group, PK pharmacokinetic

References

    1. FDA (2021) TEPMETKO (tepotinib) US Prescribing Information. . Accessed Nov 2021
    1. Bladt F, Faden B, Friese-Hamim M, et al. EMD 1214063 and EMD 1204831 constitute a new class of potent and highly selective c-Met inhibitors. Clin Cancer Res. 2013;19(11):2941–2951. doi: 10.1158/1078-0432.ccr-12-3247.
    1. Anvisa (2021) Tepotinib approval (Brazil). . Accessed Jul 2021
    1. Government of Canada (2021) Qualifying Notice for Tepmetko. . Accessed Jul 2021
    1. PMDA (2020) TEPMETKO approval in Japan. . Accessed Jul 2021
    1. SwissMedic (2021) TEPMETKO approval (Switzerland). . Accessed Jul 2021
    1. Paik P, Felip E, Veillon R, et al. Tepotinib in non-small-cell lung cancer with MET exon 14 skipping mutations. N Engl J Med. 2020;383(10):931–943. doi: 10.1056/NEJMoa2004407.
    1. Falchook GS, Kurzrock R, Amin HM, et al. First-in-man phase I trial of the selective MET inhibitor tepotinib in patients with advanced solid tumors. Clin Cancer Res. 2020;26(6):1237–1246. doi: 10.1158/1078-0432.CCR-19-2860.
    1. Wu YL, Cheng Y, Zhou J, et al. Tepotinib plus gefitinib in patients with EGFR-mutant non-small-cell lung cancer with MET overexpression or MET amplification and acquired resistance to previous EGFR inhibitor (INSIGHT study): an open-label, phase 1b/2, multicentre, randomised trial. Lancet Respir Med. 2020;8(11):1132–1143. doi: 10.1016/S2213-2600(20)30154-5.
    1. Shitara K, Yamazaki K, Tsushima T, et al. Phase I trial of the MET inhibitor tepotinib in Japanese patients with solid tumors. Jpn J Clin Oncol. 2020;50(8):859–866. doi: 10.1093/jjco/hyaa042.
    1. Strotmann R, Heuer J, Becker A, Krebs-Brown A, Mammasse N, Yalkinoglu Ö. Bioavailability of tepotinib: impact of omeprazole and food. Ann Oncol. 2019;30(Suppl. 5):v182. doi: 10.1093/annonc/mdz244.043.
    1. Yalkinoglu Ö, Heuer J, Becker A, Krebs-Brown A, Strotmann R. Drug-drug interaction profile of tepotinib with CYP3A and P-gp substrates. Ann Oncol. 2019;30(Suppl. 5):v182. doi: 10.1093/annonc/mdz244.042.
    1. Fink C, Lecomte M, Badolo L, Wagner K, Mäder K, Peters SA. Identification of solubility-limited absorption of oral anticancer drugs using PBPK modeling based on rat PK and its relevance to human. Eur J Pharm Sci. 2020 doi: 10.1016/j.ejps.2020.105431.
    1. Johne A, Scheible H, Becker A, van Lier JJ, Wolna P, Meyring M. Open-label, single-center, phase I trial to investigate the mass balance and absolute bioavailability of the highly selective oral MET inhibitor tepotinib in healthy volunteers. Invest New Drugs. 2020;38(5):1507–1519. doi: 10.1007/s10637-020-00926-1.
    1. Xiong W, Friese-Hamim M, Johne A, et al. Translational pharmacokinetic-pharmacodynamic modeling of preclinical and clinical data of the oral MET inhibitor tepotinib to determine the recommended phase II dose. CPT Pharmacometrics Syst Pharmacol. 2021;10(5):428–440. doi: 10.1002/psp4.12602.
    1. Patel H, Egorin MJ, Remick SC, et al. Comparison of Child-Pugh (CP) criteria and NCI organ dysfunction working group (NCI-ODWG) criteria for hepatic dysfunction (HD): implications for chemotherapy dosing. J Clin Oncol. 2004;22(14_suppl):6051–6051. doi: 10.1200/jco.2004.22.90140.6051.
    1. Hunsicker L, Adler S, Caggiula A, et al. Predictors of the progression of renal disease in the modification of diet in renal disease study. Kidney Int. 1997;51(6):1908–1919. doi: 10.1038/KI.1997.260.
    1. Jonsson EN, Harling K (2018) Increasing the efficiency of the covariate search algorithm in the SCM. Presented at PAGE, Athens, Greece, Accessed Jul 2021
    1. Mould D, Upton R. Basic concepts in population modeling, simulation, and model-based drug development-part 2: introduction to pharmacokinetic modeling methods. CPT Pharmacometrics Syst Pharmacol. 2013;2(4):38. doi: 10.1038/psp.2013.14.
    1. Wolf J, Seto T, Han J-YY, et al. Capmatinib in MET exon 14–mutated or MET–amplified non-small-cell lung cancer. N Engl J Med. 2020;383(10):944–957. doi: 10.1056/nejmoa2002787.
    1. Paik P, Xiong W, Hietala SF, et al. 584P Tepotinib exposure–response analyses of safety and efficacy in patients with solid tumours. Ann Oncol. 2020;31:S494–S495. doi: 10.1016/j.annonc.2020.08.698.
    1. EMA (2017) ICH guideline E17 on general principles for planning and design of multi-regional clinical trials. . Accessed Aug 2021
    1. Wesolowski R, Zhao M, Geyer SM, et al. Phase I trial of the PARP inhibitor veliparib (V) in combination with carboplatin (C) in metastatic breast cancer (MBC) J Clin Oncol. 2014;32(15_suppl):1074. doi: 10.1200/JCO.2014.32.15_SUPPL.1074.
    1. Decaens T, Barone C, Assenat E, et al. Phase 1b/2 trial of tepotinib in sorafenib pretreated advanced hepatocellular carcinoma with MET overexpression. Br J Cancer. 2021;125(2):190–199. doi: 10.1038/s41416-021-01334-9.
    1. (2018) c-Met Second-Line Hepatocellular Carcinoma. . Accessed Aug 2021
    1. (2019) Tepotinib Hepatic Impairment Trial. . Accessed Jul 2021
    1. (2018) Effect of a Proton Pump Inhibitor on the Pharmacokinetics (PK) of Tepotinib. . Accessed Aug 2021
    1. (2019) Bioequivalence of Tablet Formulation 3 (TF3) and Tablet Formulation 2 (TF2) and Effect of Food on the Pharmacokinetics (PK) of Tepotinib. . Accessed Aug 2021

Source: PubMed

3
Abonnieren