Exposure-response analyses for the MET inhibitor tepotinib including patients in the pivotal VISION trial: support for dosage recommendations

Wenyuan Xiong, Sofia Friberg Hietala, Joakim Nyberg, Orestis Papasouliotis, Andreas Johne, Karin Berghoff, Kosalaram Goteti, Jennifer Dong, Pascal Girard, Karthik Venkatakrishnan, Rainer Strotmann, Wenyuan Xiong, Sofia Friberg Hietala, Joakim Nyberg, Orestis Papasouliotis, Andreas Johne, Karin Berghoff, Kosalaram Goteti, Jennifer Dong, Pascal Girard, Karthik Venkatakrishnan, Rainer Strotmann

Abstract

Purpose: Tepotinib is a highly selective MET inhibitor approved for treatment of non-small cell lung cancer (NSCLC) harboring METex14 skipping alterations. Analyses presented herein evaluated the relationship between tepotinib exposure, and efficacy and safety outcomes.

Methods: Exposure-efficacy analyses included data from an ongoing phase 2 study (VISION) investigating 500 mg/day tepotinib in NSCLC harboring METex14 skipping alterations. Efficacy endpoints included objective response, duration of response, and progression-free survival. Exposure-safety analyses included data from VISION, plus four completed studies in advanced solid tumors/hepatocellular carcinoma (30-1400 mg). Safety endpoints included edema, serum albumin, creatinine, amylase, lipase, alanine aminotransferase, aspartate aminotransferase, and QT interval corrected using Fridericia's method (QTcF).

Results: Tepotinib exhibited flat exposure-efficacy relationships for all endpoints within the exposure range observed with 500 mg/day. Tepotinib also exhibited flat exposure-safety relationships for all endpoints within the exposure range observed with 30-1400 mg doses. Edema is the most frequently reported adverse event and the most frequent cause of tepotinib dose reductions and interruptions; however, the effect plateaued at low exposures. Concentration-QTc analyses using data from 30 to 1400 mg tepotinib resulted in the upper bounds of the 90% confidence interval being less than 10 ms for the mean exposures at the therapeutic (500 mg) and supratherapeutic (1000 mg) doses.

Conclusions: These analyses provide important quantitative pharmacologic support for benefit/risk assessment of the 500 mg/day dosage of tepotinib as being appropriate for the treatment of NSCLC harboring METex14 skipping alterations.

Registration numbers: NCT01014936, NCT01832506, NCT01988493, NCT02115373, NCT02864992.

Keywords: Dose selection; METex14 skipping alteration; NSCLC; Targeted therapies; Tyrosine kinase inhibitor.

Conflict of interest statement

Wenyuan Xiong was employed by Merck Healthcare KGaA, Darmstadt, Germany for the duration of the study. Sofia Friberg Hietala and Joachim Nyberg are employees of Pharmetheus AB, Sweden. Orestis Papasouliotis, Andreas Johne, Karin Berghoff, Kosalaram Goteti, Jennifer Dong, Pascal Girard, Karthik Venkatakrishnan, and Rainer Strotmann are employees of Merck Healthcare KGaA, Darmstadt, Germany.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Association between tepotinib exposure and independently assessed (panels ac) or investigator-assessed (panel a) efficacy outcomes in patients with NSCLC and MET exon 14 skipping alterations. Objective response rate (a), duration of response (b), and progression-free survival (c) by tepotinib AUCτ,ss quartile. OR and PFS analyses include all 146 patients; duration of response is based on 66 patients who attained an objective response. The lines represent the Clopper–Pearson 95% CI and points are observed OR per AUC quartile (dark gray represents OR assessed by independent evaluation, and light gray represents OR assessed by investigator review). In panels b and c, shaded areas represent 95% confidence intervals. AUCτ,ss, area under the curve at steady state; CI, confidence interval; NSCLC, non-small cell lung cancer; OR, objective response; ORR, objective response rate; PFS, progression-free survival
Fig. 2
Fig. 2
Relationship between tepotinib AUC24h quartile and edema events and change in serum albumin levels. Panel a presents time-to-first edema event stratified by tepotinib AUC24h quartile on the day of the edema event or day of censoring. Panel b presents the distribution of mean tepotinib AUC24h during 1 week prior to an edema event according to edema severity (maximum severity per participant). Panel c presents impact of age on the predicted risk of edema based on the final TTE model with model-estimated hazard ratios for edema relative to a typical participant of median age of 66 years (the closed symbols represent the median hazard ratio for the applicable age category. The whiskers represent the 90% CI of the median values, based on 100 bootstrap datasets. The vertical black line represents the hazard ratio for a typical patient in the analysis data set, aged 66 years). Panel d presents the visual predictive check of the indirect response model of serum albumin with an inhibitory effect of tepotinib exposure on albumin formation. In panels a and d, shaded areas represent 95% CI. In panel e, solid and dashed red lines represent the observed median, 5th and 95th percentiles; the shaded red area represents the 95% CI of the model predicted median, and the shaded blue areas represent the 95% CI of the model predicted 5th and 95th percentiles. Dots are observed values. Panel e presents a Kaplan–Meier analysis of time-to-first edema event stratified by quartiles of baseline serum albumin. Panel f presents mean change from baseline serum albumin according to edema severity. AUC24h, 24-h area under the curve; CI, confidence interval; TTE, time-to-event
Fig. 2
Fig. 2
Relationship between tepotinib AUC24h quartile and edema events and change in serum albumin levels. Panel a presents time-to-first edema event stratified by tepotinib AUC24h quartile on the day of the edema event or day of censoring. Panel b presents the distribution of mean tepotinib AUC24h during 1 week prior to an edema event according to edema severity (maximum severity per participant). Panel c presents impact of age on the predicted risk of edema based on the final TTE model with model-estimated hazard ratios for edema relative to a typical participant of median age of 66 years (the closed symbols represent the median hazard ratio for the applicable age category. The whiskers represent the 90% CI of the median values, based on 100 bootstrap datasets. The vertical black line represents the hazard ratio for a typical patient in the analysis data set, aged 66 years). Panel d presents the visual predictive check of the indirect response model of serum albumin with an inhibitory effect of tepotinib exposure on albumin formation. In panels a and d, shaded areas represent 95% CI. In panel e, solid and dashed red lines represent the observed median, 5th and 95th percentiles; the shaded red area represents the 95% CI of the model predicted median, and the shaded blue areas represent the 95% CI of the model predicted 5th and 95th percentiles. Dots are observed values. Panel e presents a Kaplan–Meier analysis of time-to-first edema event stratified by quartiles of baseline serum albumin. Panel f presents mean change from baseline serum albumin according to edema severity. AUC24h, 24-h area under the curve; CI, confidence interval; TTE, time-to-event
Fig. 2
Fig. 2
Relationship between tepotinib AUC24h quartile and edema events and change in serum albumin levels. Panel a presents time-to-first edema event stratified by tepotinib AUC24h quartile on the day of the edema event or day of censoring. Panel b presents the distribution of mean tepotinib AUC24h during 1 week prior to an edema event according to edema severity (maximum severity per participant). Panel c presents impact of age on the predicted risk of edema based on the final TTE model with model-estimated hazard ratios for edema relative to a typical participant of median age of 66 years (the closed symbols represent the median hazard ratio for the applicable age category. The whiskers represent the 90% CI of the median values, based on 100 bootstrap datasets. The vertical black line represents the hazard ratio for a typical patient in the analysis data set, aged 66 years). Panel d presents the visual predictive check of the indirect response model of serum albumin with an inhibitory effect of tepotinib exposure on albumin formation. In panels a and d, shaded areas represent 95% CI. In panel e, solid and dashed red lines represent the observed median, 5th and 95th percentiles; the shaded red area represents the 95% CI of the model predicted median, and the shaded blue areas represent the 95% CI of the model predicted 5th and 95th percentiles. Dots are observed values. Panel e presents a Kaplan–Meier analysis of time-to-first edema event stratified by quartiles of baseline serum albumin. Panel f presents mean change from baseline serum albumin according to edema severity. AUC24h, 24-h area under the curve; CI, confidence interval; TTE, time-to-event
Fig. 3
Fig. 3
Change in serum creatinine following tepotinib administration. Panel a presents the change from baseline in serum creatinine concentrations following first dose of study medication for all individual patients. Each line represents the data for one participant. The solid blue line is a LOESS smooth. The y-axis is truncated at -50 and 200 μmol/L and the x-axis at 365 days. Panel b presents the individual maximum change from baseline in serum creatinine concentration versus tepotinib AUC24h. Dots represent observations. The solid black line is a LOESS smooth. The vertical blue lines indicate the PK model simulated median (solid line), 5th and 95th percentiles (dashed lines) of AUCτ,ss at a dose of 500 mg. Panel c presents individual serum creatinine concentrations taken from 56 observations from 11 patients over time following a single administration of tepotinib (500 mg) in healthy volunteers in study 007. The black lines represent individual patient data and the blue line is a LOESS smooth. AUC24h, 24-h area under the curve; AUCτ,ss, area under the curve at steady state; LOESS, locally estimated scatterplot smoothing; PK, pharmacokinetics
Fig. 4
Fig. 4
Relationship of ΔQTcF interval versus tepotinib plasma concentration. The model derived predicted population ΔQTcF from baseline is shown as the continuous blue line and the two-sided 90% bootstrapped confidence limits of predicted mean ΔQTcF are shown as broken lines for pooled study patients. The vertical red lines correspond to geometric mean Cmax at steady state in the 500 mg and 1400 mg dose levels. The brown horizontal lines represent the regulatory threshold of potential concern of 10 ms, and an additional 20 ms reference line as a threshold of potential clinical relevance applicable for oncology drugs. Open symbols represent observed data. CI, confidence interval; QTcF, QT interval corrected using Fredericia’s formula
Fig. 5
Fig. 5
Relationship between tepotinib exposure and grade ≥ 3 adverse event, and dose reduction due to an adverse event. Panel a presents Kaplan–Meier analysis of time-to-first grade ≥ 3 AE stratified according to tepotinib exposure quartile. Panel b presents Kaplan–Meier analysis of time-to-first dose reduction due to an AE stratified according to tepotinib exposure quartile. Shaded areas represent 95% confidence intervals. AE, adverse event; AUCτ,ss, area under the curve at steady state

References

    1. Referenced with permission from the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Non-Small Cell Lung Cancer V.3.2022. © National Comprehensive Cancer Network, Inc. 2022. All rights reserved. Accessed May 6, 2022. To view the most recent and complete version of the guideline, go online to . NCCN makes no warranties of any kind whatsoever regarding their content, use or application and disclaims any responsibility for their application or use in any way.
    1. Hanna NH, Robinson AG, Temin S, et al. Therapy for stage IV non–small-cell lung cancer with driver alterations: ASCO and OH (CCO) joint guideline update. J Clin Oncol. 2021;39:1040–1091. doi: 10.1200/JCO.20.03570.
    1. Salgia R, Sattler M, Scheele J, et al. The promise of selective MET inhibitors in non-small cell lung cancer with MET exon 14 skipping. Cancer Treat Rev. 2020;87:102022. doi: 10.1016/j.ctrv.2020.102022.
    1. Frampton GM, Ali SM, Rosenzweig M, et al. Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015;5:850–859. doi: 10.1158/-15-0285.
    1. Cortot AB, Kherrouche Z, Descarpentries C, et al. Exon 14 deleted MET receptor as a new biomarker and target in cancers. J Natl Cancer Inst. 2017;109:djq262. doi: 10.1093/jnci/djw262.
    1. Paik PK, Felip E, Veillon R, et al. Tepotinib in non-small-cell lung cancer with MET exon 14 skipping mutations. N Engl J Med. 2020;383:931–943. doi: 10.1056/NEJMoa2004407.
    1. Bladt F, Friese-Hamim M, Ihling C, et al. The c-Met inhibitor MSC2156119J effectively inhibits tumor growth in liver cancer models. Cancers (Basel) 2014;6:1736–1752. doi: 10.3390/cancers6031736.
    1. Friese-Hamim M, Bladt F, Locatelli G, et al. The selective c-Met inhibitor tepotinib can overcome epidermal growth factor receptor inhibitor resistance mediated by aberrant c-Met activation in NSCLC models. Am J Cancer Res. 2017;7:962–972.
    1. Wu ZX, Teng QX, Cai CY, et al. Tepotinib reverses ABCB1-mediated multidrug resistance in cancer cells. Biochem Pharmacol. 2019;166:120–127. doi: 10.1016/J.BCP.2019.05.015.
    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;19:2941–2951. doi: 10.1158/1078-0432.ccr-12-3247.
    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;26:1237–1246. doi: 10.1158/1078-0432.CCR-19-2860.
    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;50:859–866. doi: 10.1093/jjco/hyaa042.
    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;8:1132–1143. doi: 10.1016/S2213-2600(20)30154-5.
    1. Xiong W, Papasouliotis O, Jonsson EN. Population pharmacokinetic analysis of tepotinib, an oral MET kinase inhibitor, including data from the VISION study. Cancer Chemother Pharmacol. 2022;89:655–669. doi: 10.1007/s00280-022-04423-5.
    1. Mazieres J, Paik PK, Felip E, et al (2020) 1283P—Tepotinib in patients (pts) with advanced NSCLC with MET exon 14 (METex14) skipping: overall efficacy results from VISION cohort A. Ann Oncol 31: Poster 1283P. 10.1016/j.annonc.2020.08.1597
    1. Gastonguay MR (2011) Full covariate models as an alternative to methods relying on statistical significance for inferences about covariate effects: a review of methodology and 42 case studies. Presented at: PAGE, Athens, Greece
    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;125:190–199. doi: 10.1038/s41416-021-01334-9.
    1. Ryoo B-Y, Cheng A-L, Ren Z, et al. Randomised Phase 1b/2 trial of tepotinib vs sorafenib in Asian patients with advanced hepatocellular carcinoma with MET overexpression. Br J Cancer. 2021;125:200–208. doi: 10.1038/S41416-021-01380-3.
    1. Johne A, Scheible H, Becker A, et al. 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;38:1507–1519. doi: 10.1007/s10637-020-00926-1.
    1. National Cancer Institute (2009) Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0. . Accessed 21 Oct 2021
    1. Jonsson EN, Karlsson MO. Automated covariate model building within NONMEM. Pharm Res. 1998;15:1463–1468. doi: 10.1023/A:1011970125687.
    1. Jonsson EN, Harling K (2018) Increasing the efficiency of the covariate search algorithm in the SCM. Presented at: PAGE, Athens, Greece
    1. Garnett C, Bonate PL, Dang Q, et al. Scientific white paper on concentration-QTc modeling. J Pharmacokinet Pharmacodyn. 2017;45:383–397. doi: 10.1007/S10928-017-9558-5.
    1. Veillon R, Sakai H, Le X, et al (2020) FP14.09 – Tepotinib safety in MET exon 14 (METex14) skipping NSCLC: updated results from the VISION trial. J Thorac Oncol 16: FP14.09. 10.1016/j.jtho.2021.01.152
    1. Venkatakrishnan K, Friberg L, Ouellet D, et al. Optimizing oncology therapeutics through quantitative translational and clinical pharmacology: challenges and opportunities. Clin Pharmacol Ther. 2015;97:37–54. doi: 10.1002/CPT.7.
    1. Bullock JM, Lin T, Bilic S. Clinical pharmacology tools and evaluations to facilitate comprehensive dose finding in oncology: a continuous risk-benefit approach. J Clin Pharmacol. 2017;57:S105–S115. doi: 10.1002/JCPH.908.
    1. Bullock JM, Rahman A, Liu Q. Lessons learned: dose selection of small molecule-targeted oncology drugs. Clin Cancer Res. 2016;22:2630–2638. doi: 10.1158/1078-0432.CCR-15-2646.
    1. Faucette S, Wagh S, Trivedi A, et al. Reverse translation of US Food and Drug Administration reviews of oncology new molecular entities approved in 2011–2017: lessons learned for anticancer drug development. Clin Transl Sci. 2018;11:123–146. doi: 10.1111/CTS.12527.
    1. Parchment RE, Doroshow JH. Pharmacodynamic endpoints as clinical trial objectives to answer important questions in oncology drug development. Semin Oncol. 2016;43:514–525. doi: 10.1053/J.SEMINONCOL.2016.07.002.
    1. Venkatakrishnan K, Ecsedy J. Enhancing value of clinical pharmacodynamics in oncology drug development: an alliance between quantitative pharmacology and translational science. Clin Pharmacol Ther. 2017;101:99–113. doi: 10.1002/CPT.544.
    1. Blanc-Durand F, Alameddine R, Iafrate AJ, et al. Tepotinib efficacy in a patient with non-small cell lung cancer with brain metastasis harboring an HLA-DRB1-MET gene fusion. Oncologist. 2020;25:916–920. doi: 10.1634/theoncologist.2020-0502.
    1. Paik P, Sakai H, Felip E, et al. MA11.05 – Tepotinib in patients with MET exon 14 (METex14) skipping advanced NSCLC: updated efficacy results from VISION cohort A. J Thorac Oncol. 2021;16:174. doi: 10.1016/j.jtho.2021.01.250.
    1. Tong JH, Yeung SF, Chan AWH, et al. MET amplification and exon 14 splice site mutation define unique molecular subgroups of non-small cell lung carcinoma with poor prognosis. Clin Cancer Res. 2016;22:3048–3056. doi: 10.1158/1078-0432.CCR-15-2061.
    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;383:944–957. doi: 10.1056/nejmoa2002787.
    1. Choueiri TK, Heng DYC, Lee JL, et al. Efficacy of savolitinib vs sunitinib in patients with MET-driven papillary renal cell carcinoma: the SAVOIR phase 3 randomized clinical trial. JAMA Oncol. 2020;6:1247–1255. doi: 10.1001/JAMAONCOL.2020.2218.
    1. Heigener DF, Reck M. Crizotinib. Recent Results Cancer Res. 2018;211:57–65. doi: 10.1007/978-3-319-91442-8_4.
    1. Usatyuk PV, Fu P, Mohan V, et al. Role of c-Met/Phosphatidylinositol 3-Kinase (PI3k)/Akt signaling in hepatocyte growth factor (HGF)-mediated lamellipodia formation, reactive oxygen species (ROS) generation, and motility of lung endothelial cells *. J Biol Chem. 2014;289:13476–13491. doi: 10.1074/JBC.M113.527556.
    1. Yamada N, Nakagawa S, Horai S, et al. Hepatocyte growth factor enhances the barrier function in primary cultures of rat brain microvascular endothelial cells. Microvasc Res. 2014;92:41–49. doi: 10.1016/J.MVR.2013.12.004.
    1. Morley R, Cardenas A, Hawkins P, et al. Safety of onartuzumab in patients with solid tumors: experience to date from the onartuzumab clinical trial program. PLoS One. 2015;10:e0139679. doi: 10.1371/JOURNAL.PONE.0139679.
    1. Tabernero J, Elez ME, Herranz M, et al. A pharmacodynamic/pharmacokinetic study of ficlatuzumab in patients with advanced solid tumors and liver metastases. Clin Cancer Res. 2014;20:2793–2804. doi: 10.1158/1078-0432.CCR-13-1837.
    1. Mathialagan S, Rodrigues AD, Feng B. Evaluation of renal transporter inhibition using creatinine as a substrate in vitro to assess the clinical risk of elevated serum creatinine. J Pharm Sci. 2017;106:2535–2541. doi: 10.1016/J.XPHS.2017.04.009.
    1. Mohan A, Herrmann S. Capmatinib-induced pseudo-acute kidney injury: a case report. Am J Kidney Dis. 2021 doi: 10.1053/J.AJKD.2021.04.009.
    1. Topletz-Erickson AR, Lee AJ, Mayor JG, et al. Tucatinib inhibits renal transporters OCT2 and MATE without impacting renal function in healthy subjects. J Clin Pharmacol. 2021;61:461–471. doi: 10.1002/JCPH.1750.
    1. Scotcher D, Arya V, Yang X, et al. A novel physiologically based model of creatinine renal disposition to integrate current knowledge of systems parameters and clinical observations. CPT Pharmacomet Syst Pharmacol. 2020;9:310–321. doi: 10.1002/PSP4.12509.

Source: PubMed

3
구독하다