Phase 1 study of capmatinib in MET-positive solid tumor patients: Dose escalation and expansion of selected cohorts

Yung-Jue Bang, Wu-Chou Su, Martin Schuler, Do-Hyun Nam, Wan Teck Lim, Todd M Bauer, Analia Azaro, Ronnie Tung Ping Poon, David Hong, Chia-Chi Lin, Mikhail Akimov, Samson Ghebremariam, Sylvia Zhao, Monica Giovannini, Brigette Ma, Yung-Jue Bang, Wu-Chou Su, Martin Schuler, Do-Hyun Nam, Wan Teck Lim, Todd M Bauer, Analia Azaro, Ronnie Tung Ping Poon, David Hong, Chia-Chi Lin, Mikhail Akimov, Samson Ghebremariam, Sylvia Zhao, Monica Giovannini, Brigette Ma

Abstract

Capmatinib is an oral, ATP-competitive, and highly potent, type 1b MET inhibitor. Herein, we report phase 1 dose-escalation results for capmatinib in advanced MET-positive solid tumor patients and dose expansion in advanced non-lung tumors. Capmatinib was well tolerated with a manageable safety profile across all explored doses. Dose-limiting toxicities (DLT) occurred at 200 mg twice daily (bid), 250 mg bid, and 450 mg bid capsules; however, no DLT were reported at 600 mg bid (capsules). Capmatinib tablets at 400 mg bid had comparable tolerability and exposure to that of 600 mg bid capsules. Maximum tolerated dose was not reached; recommended phase 2 dose was 400 mg bid tablets/600 mg bid capsules; at this dose, Ctrough >EC90 (90% inhibition of c-MET phosphorylation in animal models) is expected to be achieved and maintained. Among the dose-expansion patients (N = 38), best overall response across all cohorts was stable disease (gastric cancer 22%, hepatocellular carcinoma 46%, other indications 28%); two other indication patients with gene copy number (GCN) ≥6 achieved substantial tumor reduction. Near-complete immunohistochemically determined phospho-MET inhibition (H-score = 2) was shown following capmatinib 450 mg bid capsule in paired biopsies obtained from one advanced colorectal cancer patient. Incidence of high-level MET GCN (GCN ≥6) and MET-overexpressing (immunohistochemistry 3+) tumors in the expansion cohorts was 8% and 13%, respectively; no MET mutations were observed. Thus, the recommended phase 2 dose (RP2D) of capmatinib was 600 mg bid capsule/400 mg bid tablet. Capmatinib was well tolerated and showed antitumor activity and acceptable safety profile at the RP2D. (ClinicalTrials.gov Identifier: NCT01324479).

Keywords: MET amplification; MET dysregulation; capmatinib; phase 1; solid tumors.

Conflict of interest statement

Annual value of remuneration received:

  1. Mikhail Akimov from Novartis (employment).

  2. Samson Ghebremariam from Novartis (employment).

  3. Monica Giovannini (self) from Novartis (employment).

  4. Monica Giovannini (family member) from Bluebirdbio (employment).

Annual profit from shares received:

  1. Mikhail Akimov has Novartis Stock.

  2. Samson Ghebremariam has Novartis Stock.

  3. Monica Giovannini (self) has Novartis Stock.

  4. Monica Giovannini (family member) has Bluebirdbio Stock.

Total annual value of daily allowances/honoraria received:

  1. David Hong from AbbVie (research grants), Adaptimmune (research grants, and consulting or advisory role), Amgen (research grants), AstraZeneca (research grants), Bayer (research grants, and consulting or advisory role), BMS (research grants), Daiichi Sankyo (research grants), Eisai (research grants), Fate Therapeutics (research grants), Genentech (research grants, consulting or advisory role), Genmab (research grants), Ignyta (research grants), Infinity (research grants), Kite (research grants), Kyowa (research grants), Lilly (research grants), LOXO (research grants), Merck (research grants), MedImmune (research grants), Mirati (research grants), MiRNA (research grants), Molecular Templates (research grants), Mologen (research grants), NCI‐CTEP (research grants), Novartis (research grants), Pfizer (research grants, and consulting or advisory role), Seattle Genetics (research grants, and consulting or advisory role), Takeda (research grants, and consulting or advisory role), Alpha Insights (consulting or advisory role), Axiom (consulting or advisory role), Baxter (consulting or advisory role), GLG (consulting or advisory role), Group H (consulting or advisory role), Guidepoint Global (consulting or advisory role), Infinity (consulting or advisory role), Janssen (consulting or advisory role), Merrimack (consulting or advisory role), Medscape (consulting or advisory role), Numab (consulting or advisory role), and Trieza Therapeutics (consulting or advisory role).

Total annual value of manuscript fees received:

  1. Todd M. Bauer from Pfizer (paid to third part vendor for medical writing/editorial support).

Total annual value of research funds, endowments, endowed chairs, and researcher‐employment costs received:

  1. Yung‐Jue Bang (for clinical trials to the institution) from AstraZeneca, Novartis, Genentech/Roche, MSD, Merck Serano, Bayer, BMS, GSK, Pfizer, Eli Lilly, Boehringer‐Ingelheim, MacroGenics, Boston Biomedical, FivePrime, Curis, Taiho, Takeda, Ono, Daiichi Sankyo, Astellas, BeiGene, Green Cross, CKD Pharma, Genexine. Martin Schuler (research grants provided to academic institution from AstraZeneca, Bristol‐Myers Squibb, Novartis. Wan Teck Lim from Novartis (personal fees). Todd M. Bauer (to the institution) from Daiichi Sankyo, Medpacto (grants), Incyte (grants), Mirati Therapeutics (grants), MedImmune (grants), Abbvie (grants), AstraZeneca (grants), MabVax (grants), Stemline Therapeutics (grants), Merck (grants), Lilly (grants), GlaxoSmithKline (grants), Novartis (grants), Genentech (grants), Deciphera (grants), Merrimack (grants), Immunogen (grants), Millennium (grants), Phosplatin Therapeutics (grants), Calithera Biosciences (grants), Kolltan Pharmaceuticals (grants), Principia Biopharma (grants), Peloton (grants), Immunocore (grants), Roche (grants), Aileron Therapeutics (grants), Bristol‐Myers Squibb (grants), Amgen (grants), Onyx (grants), Sanofi (grants), Boehringer‐Ingelheim (grants), Astellas Pharma (grants), Five Prime Therapeutics (grants), Jacobio (grants), Top Alliance BioScience (grants), Janssen (grants), Clovis Oncology (grants), Takeda (grants), Karyopharm Therapeutics (grants), Foundation Medicine (grants), ARMO Biosciences (grants), Leap Therapeutics (grants and other), Ignyta (grants, non‐financial support and other), Moderna Therapeutics (grants, non‐financial support and other), Pfizer (grants, personal fees and other), Loxo (grants, personal fees and non‐financial support), Bayer (grants, personal fees and non‐financial support), Guardant Health (personal fees and non‐financial support from) outside the submitted work. David Hong from Molecular Match (Advisor), OncoResponse (founder), and Presagia Inc. (Advisor). Brigette Ma from Novartis (personal fees [advisory role] and research grant), BI (personal fees [advisory role]), BMS (personal fees [advisory role]), and MSD (personal fees [advisory role]), and Roche (personal fees [speaker]).

Other annual remuneration received:

  1. David Hong from LOXO, MiRNA, ASCO, AACR, SITC, and Genmab.

  2. Mikhail Akimov from Novartis.

  3. Samson Ghebremariam from Novartis.

  4. Monica Giovannini (self) from Novartis (employment).

  5. Monica Giovannini (family member) from Bluebirdbio (employment).

© 2019 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

Figures

Figure 1
Figure 1
Best percentage change from baseline in sum of tumor diameters according to investigator assessment in dose‐expansion cohorts (N = 18)*. *Patients with measurable baseline disease and at least one valid postbaseline (BIRC) assessment (best percentage change from baseline 0 [n = 11]). BIRC, blinded independent review committee; GCN, gene copy number; HCC, hepatocellular carcinoma; IHC, immunohistochemistry; unk, unknown.

References

    1. Sierra JR, Tsao MS. c‐MET as a potential therapeutic target and biomarker in cancer. Ther Adv Med Oncol. 2011;3(1 Suppl):S21‐35.
    1. Smolen GA, Sordella R, Muir B, et al. Amplification of MET may identify a subset of cancers with extreme sensitivity to the selective tyrosine kinase inhibitor PHA‐665752. Proc Natl Acad Sci USA. 2006;103:2316‐2321.
    1. Bradley CA, Salto‐Tellez M, Laurent‐Puig P, et al. Targeting c‐MET in gastrointestinal tumours: rationale, opportunities and challenges. Nat Rev Clin Oncol. 2017;14:562‐576.
    1. Sadiq AA, Salgia R. MET as a possible target for non‐small‐cell lung cancer. J Clin Oncol. 2013;31:1089‐1096.
    1. Cappuzzo F, Marchetti A, Skokan M, et al. Increased MET gene copy number negatively affects survival of surgically resected non‐small‐cell lung cancer patients. J Clin Oncol. 2009;27:1667‐1674.
    1. Kawakami H, Okamoto I, Okamoto W, Tanizaki J, Nakagawa K, Nishio K. Targeting MET amplification as a new oncogenic driver. Cancers (Basel). 2014;6:1540‐1552.
    1. Schildhaus HU, Schultheis AM, Ruschoff J, et al. MET amplification status in therapy‐naive adeno‐ and squamous cell carcinomas of the lung. Clin Cancer Res. 2015;21:907‐915.
    1. Bean J, Brennan C, Shih JY, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proc Natl Acad Sci USA. 2007;104:20932‐20937.
    1. Chen HJ, Mok TS, Chen ZH, et al. Clinicopathologic and molecular features of epidermal growth factor receptor T790M mutation and c‐MET amplification in tyrosine kinase inhibitor‐resistant Chinese non‐small cell lung cancer. Pathol Oncol Res. 2009;15:651‐658.
    1. Engelman JA, Zejnullahu K, Mitsudomi T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science. 2007;316:1039‐1043.
    1. Sequist LV, Waltman BA, Dias‐Santagata D, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3:75ra26.
    1. Yu HA, Arcila ME, Rekhtman N, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR‐TKI therapy in 155 patients with EGFR‐mutant lung cancers. Clin Cancer Res. 2013;19:2240‐2247.
    1. Minari R, Bordi P, Tiseo M. Third‐generation epidermal growth factor receptor‐tyrosine kinase inhibitors in T790M‐positive non‐small cell lung cancer: review on emerged mechanisms of resistance. Transl Lung Cancer Res. 2016;5:695‐708.
    1. Lorenzato A, Olivero M, Patane S, et al. Novel somatic mutations of the MET oncogene in human carcinoma metastases activating cell motility and invasion. Cancer Res. 2002;62:7025‐7030.
    1. Lee JH, Han SU, Cho H, et al. A novel germ line juxtamembrane Met mutation in human gastric cancer. Oncogene. 2000;19:4947‐4953.
    1. Drilon AE, Camidge DR, Ou S‐HI, et al. Efficacy and safety of crizotinib in patients (pts) with advanced MET exon 14‐altered non‐small cell lung cancer (NSCLC). J Clin Oncol. 2016;34(15_suppl): 108.
    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.
    1. Schrock AB, Frampton GM, Suh J, et al. Characterization of 298 patients with lung cancer harboring MET exon 14 skipping alterations. J Thorac Oncol. 2016;11:1493‐1502.
    1. Awad MM, Oxnard GR, Jackman DM, et al. MET exon 14 mutations in non‐small‐cell lung cancer are associated with advanced age and stage‐dependent MET genomic amplification and c‐Met overexpression. J Clin Oncol. 2016;34:721‐730.
    1. Liu X, Jia Y, Stoopler MB, et al. Next‐generation sequencing of pulmonary sarcomatoid carcinoma reveals high frequency of actionable MET gene mutations. J Clin Oncol. 2016;34:794‐802.
    1. Raghav KP, Gonzalez‐Angulo AM, Blumenschein GR Jr. Role of HGF/MET axis in resistance of lung cancer to contemporary management. Transl Lung Cancer Res. 2012;1:179‐193.
    1. Dimou A, Non L, Chae YK, Tester WJ, Syrigos KN. MET gene copy number predicts worse overall survival in patients with non‐small cell lung cancer (NSCLC); a systematic review and meta‐analysis. PLoS ONE. 2014;9:e107677.
    1. Guo B, Cen H, Tan X, Liu W, Ke Q. Prognostic value of MET gene copy number and protein expression in patients with surgically resected non‐small cell lung cancer: a meta‐analysis of published literatures. PLoS ONE. 2014;9:e99399.
    1. Awad MM, Leonardi GC, Kravets S, et al. Impact of MET inhibitors on survival among patients (pts) with MET exon 14 mutant (METdel14) non‐small cell lung cancer (NSCLC). J Clin Oncol. 2017;35(15_suppl): 8511.
    1. Lal B, Xia S, Abounader R, Laterra J. Targeting the c‐Met pathway potentiates glioblastoma responses to gamma‐radiation. Clin Cancer Res. 2005;11:4479‐4486.
    1. Akervall J, Guo X, Qian CN, et al. Genetic and expression profiles of squamous cell carcinoma of the head and neck correlate with cisplatin sensitivity and resistance in cell lines and patients. Clin Cancer Res. 2004;10:8204‐8213.
    1. Backes A, Zech B, Felber B, et al. Small‐molecule. Part I: exceptions from the traditional pharmacophore approach of type I inhibition inhibitors binding to protein kinases. Expert Opin Drug Discov. 2008;3:1409‐1425.
    1. Gherardi E, Birchmeier W, Birchmeier C, Woude GV. Targeting MET in cancer: rationale and progress. Nat Rev Cancer. 2012;12:89‐103.
    1. Backes A, Zech B, Felber B, Klebl B, Müller G. Small‐molecule inhibitors binding to protein kinase. Part II: the novel pharmacophore approach of type II and type III inhibition. Expert Opin Drug Discov. 2008;3(12):1427‐1449.
    1. Liu X, Wang Q, Yang G, et al. A novel kinase inhibitor, INCB28060, blocks c‐MET‐dependent signaling, neoplastic activities, and cross‐talk with EGFR and HER‐3. Clin Cancer Res. 2011;17:7127‐7138.
    1. Baltschukat S, Engstler BS, Huang A, et al. Capmatinib (INC280) is active against models of non‐small cell lung cancer and other cancer types with defined mechanisms of MET activation. Clin Cancer Res. 2019;25:3164‐3175.
    1. Babb J, Rogatko A, Zacks S. Cancer phase I clinical trials: efficient dose escalation with overdose control. Stat Med. 1998;17:1103‐1120.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med. 2008;27:2420‐2439.
    1. Esaki T, Hirai F, Makiyama A, et al. Phase I dose‐escalation study of capmatinib (INC280) in Japanese patients with advanced solid tumors. Cancer Sci. 2019;110:1340‐1351.
    1. Hu X, Zheng X, Mo H, et al. P1.01-37 BPI-9016M, a novel c-Met inhibitor, in pretreated advanced solid tumor: results from a first-in-human, phase 1, dose-escalation study. J Thorac Oncol. 2018; 13:S474.
    1. Drilon A, Clark J, Weiss J, et al. OA12.02 Updated antitumor activity of crizotinib in patients with MET exon 14-altered advanced non-small cell lung cancer. J Thorac Oncol. 2018;13:S348.
    1. Camidge DR, Otterson GA, Clark JW, et al. Crizotinib in patients (pts) with MET‐amplified non‐small cell lung cancer (NSCLC): updated safety and efficacy findings from a phase 1 trial. J Clin Oncol. 2018;36(15_suppl):9062‐9062.
    1. Felip E, Sakai H, Patel J, et al. OA12.01 Phase II data for the MET inhibitor tepotinib in patients with advanced NSCLC and MET exon 14-skipping mutations. J Thorac Oncol. 2018;13:S347.

Source: PubMed

3
Sottoscrivi