Next Article in Journal
The Local Microbiome in Esophageal Cancer and Treatment Response: A Review of Emerging Data and Future Directions
Next Article in Special Issue
A Systematic Review of Mesenchymal Epithelial Transition Factor (MET) and Its Impact in the Development and Treatment of Non-Small-Cell Lung Cancer
Previous Article in Journal
The Development of p53-Targeted Therapies for Human Cancers
Previous Article in Special Issue
The Role of MET in Resistance to EGFR Inhibition in NSCLC: A Review of Mechanisms and Treatment Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer—A Narrative Review

1
Division of Medical Oncology, Department of Internal Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA
2
Hoag Family Cancer Institute, Newport Beach, CA 92663, USA
3
Division of Hematology and Oncology, Department of Medicine, Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, CA 92868, USA
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(14), 3561; https://doi.org/10.3390/cancers15143561
Submission received: 24 May 2023 / Revised: 4 July 2023 / Accepted: 6 July 2023 / Published: 10 July 2023
(This article belongs to the Special Issue Roles of MET in Cancer Development and Treatment)

Abstract

:

Simple Summary

In this narrative review, we discuss the development of capmatinib, a reversible MET tyrosine kinase inhibitor that received approval for advanced non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutation. Capmatinib was first discovered in 2011 and has been shown to have promising antitumor activity. Early-phase trials identified a recommended dose of 400 mg twice daily in tablet formulation. The GEOMETRY mono-1 trial showed efficacy in MET exon 14 skipping mutation, leading to FDA approval for capmatinib. Currently, ongoing clinical trials evaluating combination therapy with capmatinib, including amivantamab, trametinib, and immunotherapy, are being conducted to improve efficacy and broaden indications of capmatinib with new drug agents such as antibody–drug conjugates being developed to treat MET dysregulated NSCLC.

Abstract

Non-small cell lung cancer (NSCLC) is a leading cause of death, but over the past decade, there has been tremendous progress in the field with new targeted therapies. The mesenchymal–epithelial transition factor (MET) proto-oncogene has been implicated in multiple solid tumors, including NSCLC, and dysregulation in NSCLC from MET can present most notably as MET exon 14 skipping mutation and amplification. From this, MET tyrosine kinase inhibitors (TKIs) have been developed to treat this dysregulation despite challenges with efficacy and reliable biomarkers. Capmatinib is a Type Ib MET TKI first discovered in 2011 and was FDA approved in August 2022 for advanced NSCLC with MET exon 14 skipping mutation. In this narrative review, we discuss preclinical and early-phase studies that led to the GEOMETRY mono-1 study, which showed beneficial efficacy in MET exon 14 skipping mutations, leading to FDA approval of capmatinib along with Foundation One CDx assay as its companion diagnostic assay. Current and future directions of capmatinib are focused on improving the efficacy, overcoming the resistance of capmatinib, and finding approaches for new indications of capmatinib such as acquired MET amplification from epidermal growth factor receptor (EGFR) TKI resistance. Clinical trials now involve combination therapy with capmatinib, including amivantamab, trametinib, and immunotherapy. Furthermore, new drug agents, particularly antibody–drug conjugates, are being developed to help treat patients with acquired resistance from capmatinib and other TKIs.

1. Introduction

Non-small cell lung cancer (NSCLC) is a leading cause of death, accounting for an estimated 1.8 million deaths according to GLOBOCAN in 2020 [1]. Over the past decade, there has been tremendous progress in the discovery and development of targeted therapies for EGFR; KRAS G12C; BRAF V600E mutations; ALK, ROS1; RET gene rearrangements; MET alterations, including MET exon 14 skipping mutations, ERBB2 (HER2) mutations, and NTRK 1/2/3 gene mutations [2,3,4,5,6,7,8,9,10]. This has led to the personalization of medicine in NSCLC.
The mesenchymal–epithelial transition factor (MET) gene is located in human chromosome 7 (7q21–q31), comprising 21 exons and 21 introns, and encodes a protein that is approximately 120 kDa in size. The ligand for MET is hepatocyte growth factor (HGF), which is a soluble cytokine and is synthesized by mesenchymal cells, fibroblasts, and smooth muscle cells [11]. HGF will bind to MET, and this will trigger the autophosphorylation of Tyr-1234 and Tyr-1235 in the intracellular tyrosine kinase domain, which then undergoes further autophosphorylation of Tyr-1340 and Tyr-1356 in the C-terminal docking site [11,12]. This then facilitates the recruitment of intracellular effector molecules such as GRB2, SRC, PIK3, and GAB1, leading to the activation of downstream pathways. Normally, MET/HGF signaling pathway mediates embryogenesis, tissue regeneration, wound healing, and the formation of nerves and muscles [11,12,13].
In cancer, the MET proto-oncogene is abnormally activated and stimulates other signaling pathways in tumor cells, notably PI3K/AKT, JAK/STAT, Ras/MAPK, SRC, and Wnt/beta-catenin [11] (Figure 1). MET overexpression can be found in inflammation and hypoxia, leading to proliferation and migration, and is seen in a large variety of cancer types, including epithelial, mesenchymal, and hematological malignancies [14]. In NSCLC, it has been shown to be overexpressed in 35–72% of cases [14]. High levels of MET expression have been found to correlate with early disease recurrence [15]. MET dysregulation in NSCLC can present in a variety of ways—gene overexpression; HGF expression that can cause ligand-induced activation, leading to sustained or altered signaling; gene amplification, which can lead to overexpression and reduce the requirement for ligand activation, leading to sustained or altered signaling of the MET receptor; gene rearrangement, which may reduce or remove the requirement for ligand activation, leading to sustained altered signaling properties of the MET receptor; and downstream MET signaling alterations [11,12,15]. Notably, cigarette smoking can upregulate c-MET and the downstream Akt pathway [16]. It also affects the sensitivity of EGFR TKIs as cigarette smoke attenuates the AMP-activated protein-kinase (AMPK)-dependent inhibition of mTOR which then decreases the sensitivity of NSCLC cells with wild-type EGFR to TKI and thereby represses the expression of liver kinase B1 (LKB1) [17]. Finally, MET dysregulation can occur via gene mutation, most notably the MET exon 14 skipping mutation seen in about 3–4% of adenocarcinoma and 2% of squamous cell carcinoma but in higher frequencies in adenosquamous carcinoma (6%) and pulmonary sarcomatoid carcinoma (9–22%) [15,18].
MET exon 14 skipping mutations are processes in which the 47-amino-acid juxtamembrane domain is deleted, altered, or disrupted by intronic regions surrounding exon 14, leading to fusion in mature mRNA between exon 13 and exon 15 [19,20]. MET exon 14 skipping mutations have been shown to be exclusive from other driver mutations but coexist with other MET amplification or copy number gains [21]. Meanwhile, the amplification of the MET gene, which is defined as a gain in copy number (GCN), has been seen both de novo and as an acquired resistance mechanism [22]. MET amplification is seen in EGFR-acquired resistance and can occur with or without the loss of T790M [23]. In the analysis of resistance mechanisms in the AURA 3 study (n = 78), MET amplification was seen in (14/78,18%) of samples, EGFR C797S (14/78,18%) of cases, and 15 patients having >1 resistance-related genomic alteration [23,24]. MET amplification is also considered an acquired resistance mechanism of ALK inhibitors, as MET amplification has been observed in about 15% of next-generation ALK inhibitor resistance [25]. Both MET exon 14 skipping mutations and MET high-level amplification have been shown to portend poor prognosis [21]. Without the use of MET inhibitors, a retrospective study by Awad et al. showed that the median OS was 8.1 months [26]. MET exon 14 skipping mutations are seen more frequently in females than in males, and the median age of MET exon 14 skip mutation patients ranged from 71.4 to 76.7 years [6,18]. Compared with other driver mutations, MET exon 14 skip mutation patients tend to be smokers, with only about 36% being never smokers in a previous retrospective analysis [27].
MET tyrosine kinase inhibitors have been developed to treat MET-dysregulated NSCLC, classified as Type I, Type II, and Type III inhibitors. Type I inhibitors compete with ATP for the binding of the ATP-binding pocket of the active conformation of MET. Specifically, Type Ia inhibitors such as crizotinib interact with the Y1230 residue in the hinge region and are dependent on binding with the G1163 residue [28,29]. Type Ib inhibitors such as capmatinib, tepotinib, and savolitinib also connect with the Y1230 residue but are not dependent on G1163 binding [28,30,31,32]. Meanwhile, Type II inhibitors, which include cabozantinib, meresitinib, and gleasatanib, bind the ATP pocket in an inactive state [32,33,34,35]. Type III inhibitors bind to allosteric sites different from the ATP site and are not competitive; tivantinib has been studied in NSCLC but was not found to show any benefit in interim analysis and therefore was discontinued [32,34,36].
This review specifically focuses on capmatinib (INC280), which received U.S. Food and Drug Administration (FDA) approval for MET exon 14 skip mutations in metastatic NSCLC on 10 August 2022 and by the European Medicines Agency (EMA) on 20 June 2022 specifically for those patients who have received immunotherapy or platinum-based chemotherapy or both [37,38]. Herein, we review clinical development trials involving capmatinib, notably the GEOMETRY mono-1 study, which led to FDA approval and the companion diagnostic assay for the detection of MET exon 14 skipping mutations.

2. Crizotinib

Prior to capmatinib, crizotinib was the first MET TKI to show efficacy in MET exon 14 skipping mutation in advanced NSCLC. The PROFILE 1001 trial showed an overall response rate (ORR) of 32% (95% CI 21–45) among 65 response-evaluable patients, with a median duration of response (DOR) of 9.1 months (95% CI 6.4–12.7) and a progression-free survival (PFS) rate of 7.3 months (95% CI 5.4–9.1), with two additional Phase II crizotinib trials showing ORR of around 30% [39,40,41]. However, crizotinib confers resistance to G1163R mutation not seen in MET Type Ib TKIs such as capmatinib, and thus treatment for MET dysregulation has shifted towards MET Type Ib TKIs [42]. Currently, crizotinib is approved for ALK- and ROS1-positive advanced NSCLC by the FDA and EMA [43,44].

3. Preclinical Studies

Capmatinib was first reported in 2011 by Liu et al., who showed that in both in vivo and in vitro mice studies using human cell lines, capmatinib had a 10,000-fold selectivity for c-met over a large panel of human kinase [45]. They showed that capmatinib can block the c-MET phosphorylation and activation of downstream targets, including HGF. They further showed that activated c-met upregulates cancer-promoting EGFR and HER-3 pathways [45]. Baltschukat et al. further investigated capmatinib in NSCLC [46]. They investigated the affinity of capmatinib in a set of 442 kinases and demonstrated a selectivity in MET of over 1000 fold [46]. Furthermore, they demonstrated that capmatinib is highly selective to Y1230 and D1228 and observed resistance when using cell lines bearing mutations to Y1230 and D1228 [46]. MET amplification and HGF expression in vitro were also associated with capmatinib sensitivity in vitro [46].

4. Pharmacodynamics/Pharmacokinetics

Capmatinib is a selective Type Ib ATP-competitive tyrosine kinase inhibitor targeting MET. Capmatinib has an average IC50 value of 0.13 nM and a cell-based IC50 of 0.3–0.7 nM in lung cancer cell lines [28,46] (Figure 2). Capmatinib has linear pharmacokinetics, with exposure increasing approximately dose-proportionally over a dose range of 200–400 mg. It is rapidly absorbed, with peak plasma concentration (Cmax) obtained about 1–2 h after a 400 mg dose is given. There is similar absorption when taken with and without food. The effective elimination half-life is 6.5 h. The plasma protein binding is 96% [38,47].
Capmatinib is metabolized by CYP3A4 and aldehyde oxidase. In a single oral dose, 78% of total radioactivity was recovered in feces with 42% as unchanged and 22% recovered in urine. There are no specific significant effects on the pharmacokinetic parameters of capmatinib identified in the following covariates assessed: age, sex, race, mild-to-moderate renal impairment, and hepatic impairment [38,47].
In drug interaction studies, coadministration with itraconazole, a strong CYP3A inhibitor, increased capmatinib’s area under the curve (AUC0-INF) by 42%, with no change in Cmax. Coadministration with rifampicin, a strong CYP3A inducer, decreased capmatinib AUC0-INF by 67% and decreased Cmax by 56%. Coadministration with protein pump inhibitors (rabeprazole) decreased capmatinib by AUC0-INF 25% and decreased Cmax by 38%. Coadministration with rosuvastatin, a BRCP substrate, increased rosuvastatin AUC0-INF by 108% and increased Cmax by 204% [38,47].

5. Phase I Clinical Trials

Multiple open-label, multicenter, Phase I studies in advanced solid tumors have evaluated capmatinib. A Phase I study comprising 44 adult Japanese patients, including 15 NSCLC patients, found that the highest studied dose determined to be safe was 400 mg administered orally (po) twice a day (b.i.d.) as a tablet. The median duration of treatment exposure was 7 weeks (range 0.4–32.3 weeks), with disease progression being the primary reason for the discontinuation occurring in 38 patients (86.4%). There were two drug-limiting toxicities (DLTs), which consisted of Grade 2 suicidal ideation in a patient taking 600 mg po b.i.d. and Grade 3 depression in a patient taking 400 mg po b.i.d. [48]. Another global Phase I study, comprising 38 patients primarily with gastrointestinal cancers, had a recommended Phase II dose (R2PD) of 600 mg po b.i.d. in a capsule formulation and 400 mg po b.i.d. in a tablet formulation. The most frequent Grade 3 or 4 adverse events were an increase in levels of blood bilirubin (11%), fatigue (8%), and AST increase (8%) [49].
Schuler et al. investigated 55 patients with advanced MET-dysregulated NSCLC, which included 40 patients with prior systemic therapies. All patients discontinued treatment, mostly due to disease progression (69.1%), with a median duration of 10.4 weeks. While the overall response rate (ORR) by RECIST for the entire cohort was 20%, MET with a gene copy number ≥6 had an ORR of 47%, with median progression-free survival (PFS) of 9.3 months, and all patients with MET exon 14 skip mutations had a response. The most common toxicities were nausea (42%), peripheral edema (33%), and vomiting (31%) [50]. Another Phase Ib/II study involving capmatinib investigated EGFR-mutated, MET-dysregulated NSCLC in combination with gefitinib, an EGFR TKI, in patients with acquired EGFR TKI resistance. The ORR across the cohort was 27%, with a 47% ORR in patients with a MET copy number ≥6. The drug was relatively well tolerated, with the most common Grade 3–4 adverse event being increased amylase and lipase levels (6% in both). The R2PD was capmatinib 400 mg po b.i.d. plus gefitinib 250 mg po daily [51] (Table 1).

6. GEOMETRY Mono-1 Trial

The GEOMETRY mono-1 trial was a multicohort Phase II study in patients with MET-dysregulated advanced NSCLC. The patients were either in Stage IIIB or IV NSCLC, had no EGFR mutation, and were negative for ALK rearrangement. All subjects took capmatinib 400 mg po b.i.d. A total of 364 patients were enrolled, with 97 having a MET exon 14 skipping mutation and 210 having MET amplification. There were seven cohorts to the study: In previously treated patients (1–2 lines of therapy), Cohort 1 consisted of MET amplification with (a) GCN ≥ 10 (n = 69) or (b) GCN 6–9 (n = 42); Cohort 2 consisted of MET amplification with GCN 4–5 (n = 54); Cohort 3 consisted of MET amplification with GCN < 4 (n = 30); Cohort 4 consisted of MET exon 14 skipping mutation with any GCN (n = 69); and Cohort 6 consisted of MET amplification with GCN > 10 (n = 3) or MET exon 14 skipping mutation with any GCN (n = 31) who had received one line of therapy (n = 34). In the untreated group, Cohort 5a consisted of MET amplification with GCN ≥ 10 (n = 15); Cohort 5b consisted of MET exon 14 skipping mutation with any GCN (n = 28); and Cohort 7 consisted of treatment-naïve MET exon 14 skipping mutation with any GCN (n = 23). MET exon 14 skipping mutation patients had a slightly higher median age (71 years) than patients with MET amplification (60–70 years) on diagnosis. Patients with MET exon 14 skipping mutation were more likely to be women and to have never smoked [6].
Among patients with MET exon 14 skip mutations, ORR was seen in 41% (95% CI 29–53) of 69 previously treated patients and 68% (95% CI 48–84) of 28 previously untreated patients. The median duration of response (DOR) was 9.7 months (95% CI 5.6–13.0) among the treated patients and 12.6 months (95% CI 5.6—not reached) in previously untreated patients. Most patients (82% in treated and 68% in untreated) had a response at the first tumor evaluation following the start of capmatinib therapy. The median PFS was 5.4 months (95% CI 4.2–7.0) in previously treated patients and 12.4 months (95% CI 8.2—not reached) in previously untreated patients. Notably, 12 of 13 patients with exon 14 skipping mutations who had brain metastasis had intracranial disease control. The primary reason for discontinuation was progressive disease (58% in previously treated patients and 46% in untreated patients) [6].
In patients with GCN < 10, the cohorts were closed due to futility, as PFS for GCN 6–9 and 4 or 5 was only 2.7 months. In GCN ≥ 10, there was activity; the ORR was 29% (95% CI 19–41) in previously treated patients and 40% (95% CI 16–68) in previously untreated patients, but this fell below the predefined clinical efficacy. The median DOR was 8.3 months (95% CI 4.2–15.4) in treated patients and 7.5 months (95% CI 2.6–14.3) in untreated patients. The median PFS was 4.1 months (95% CI 2.9–4.8) in treated patients and 4.2 months (95% CI 1.4–6.9) in untreated patients [6] (Table 2).
Across all cohorts, the most reported adverse events were peripheral edema, nausea, and vomiting. Overall, 67% of patients had adverse events of Grade 3 or 4; the most frequent of these were peripheral edema, nausea, vomiting, and increased blood creatinine level. Treatment-related adverse events led to the discontinuation of treatment in 39 patients (11%), with treatment-related peripheral edema leading to discontinuation in 6 patients (2%) [6].
The post hoc analysis involving 69 MET exon 14 skipping mutation patients that focused on 19 patients in the cohort who had previously received immunotherapy (IO) showed ORR 57.9% (n = 11/19; 95% CI 33.5–79.5%), with a median DOR of 11.2 months (95% CI 3.35—not reached). Safety findings were similar, according to which capmatinib showed efficacy irrespective of prior treatment with IO and was also well tolerated in post-IO patients [52]. Moreover, capmatinib was associated with clinically meaningful improvements in cough and preserved the quality of life in patient-reported surveys [53]. There was also a subgroup analysis on 45 Japanese patients, which showed an ORR of 36% (95% 10.9–69.2) and good tolerability [54].
A recent real-world analysis was carried out that investigated MET exon 14 skipping mutation and brain metastasis patients; of the 68 patients that fit the criteria, the real-world response rate was 90.9%, with 87.3% intracranial response along with a median PFS rate of 14.1 months [55]. Another real-world retrospective study examined 81 cases of NSCLC with advanced NSCLC and MET exon 14 skipping mutation who were treated with capmatinib from March 2019 to December 2021 [56]. The ORR to capmatinib was 58% (95% CI 47–69), including 68% (95% CI 50–82) for treatment-naïve and 50% (95% CI 35–65) for pretreated patients. The median PFS was 9.5 months (95% CI 4.7–14.3), and the median OS was 18.2 months (95% CI 13.2) for the entire cohort, including a median PFS of 10.6 months (95% CI 5.5–15.7) for untreated patients [56].
Thus, the GEOMETRY mono-1 trial evaluated MET-dysregulated, advanced NSCLC, with promising ORR and PFS seen in MET exon 14 skip mutations, though the results showed a lack of effect in MET GCN < 10, leading to FDA and EMA approval for capmatinib only in advanced NSCLC with MET exon 14 skipping mutations. Subsequent real-world data have shown response to capmatinib among patients with MET exon 14 skipping mutations, with IO exposure and brain metastasis [52,56].

7. Tepotinib and Savolitinib

Two other MET selective Type Ib inhibitors have been investigated in MET alterations, namely tepotinib and savolitinib [30,31]. Tepotinib received accelerated approval from the FDA for MET exon 14 skipping mutations in advanced NSCLC after the open-label Phase II VISION study [31]. It also received approval from the EMA for those with advanced NSCLC MET exon 14 skipping mutations who require systemic therapy following immunotherapy and/or platinum-based therapy [57]. In this study, 152 patients with MET exon 14 skipping mutations were followed, and the ORR was 46% (95% CI 36–57), including 44.2% (95% CI 29.1–60.1) in untreated patients and 48.2 (95% CI 34.7–62.0) in previously treated patients. The median DOR was 11.1 months (95% CI 7.2—not reached), and the PFS was 8.5 months (95% CI 5.1–11.0) [31]. There were 11 patients with brain metastasis in the study, with a median PFS of 10.9 months (95% CI 8.0—not reached) [31].
Meanwhile, a Phase II, single-arm, open-label study in China involved 84 patients with MET exon 14 skipping mutations who had positive pulmonary sarcomatoid carcinoma or other NSCLC subtypes and received savolitinib [30]. The ORR was 42.9% (95% CI 31.1–55.3) [20] (Table 3). Savolitinib received conditional approval in China in 2021 for the treatment of metastatic NSCLC with MET exon 14 skipping mutations in patients who have progressed after or who are unable to tolerate platinum-based chemotherapy [58].

8. Companion Diagnostic Assay

One of the challenges in the success of finding successful MET-targeted therapies has been finding a reliable biomarker. For example, in previous studies where MET GCN ≥ 6, the ORR outcomes ranged from 16% to 67%, while for immunohistochemistry (IHC) 2+ and 3+, the ORR outcomes ranged from 14% to 68% [6,39,40,41,50,51,59]. Another way to assess MET overexpression has been the MET/chromosome 7 centromere (CEP7) ratio, in which ORR outcomes range from 33% to 67% [51,59] (Table 4).
Some thoughts as to the lack of reliability in MET amplification have been that gene copy number gains can occur through both polysomy and amplification and thus the gene copy number could be a result of polysomy, not true amplification [59,60,61]. Another possible problem has been the use of NGS-based assays with a control group using CEP7 [59,61]. A previous study has shown that a MET/CEP7 ratio >5 is reliable for MET inhibitor response, but the issue is that many below this ratio have other oncogenes and may not be truly MET-addicted cases [61]. Guo et al. demonstrated that MET expression via mass spectrometry, IHC, and H-score ≥ 200 had significantly improved PFS but saw no association based on copy number [62].
Another challenging aspect of finding a reliable companion diagnostic assay has been the discrepancy between circulating tumor DNA (ctDNA) and tumor next-generation sequencing (NGS) testing. Ikeda et al. studied the ctDNA of 438 patients, and among the 31 patients with MET alterations, only 2 of the 18 patients who also received tissue testing were found to have MET alterations in the tissue [63]. Another study involving paired plasma and tissue samples in advanced NSCLC patients showed 77.6% concordance between tissue and plasma NGS; 26% of the cohort who received both ctDNA and tissue testing had MET alterations on ctDNA testing, but only 17.8% of the 26% total also had MET alterations on tissue testing [64]. Overall, when compared to tumor NGS testing, ctDNA had 67.7% sensitivity and 88.8% specificity in pretreated patients, whereas in treated patients, it revealed a sensitivity of 68.4% but only a specificity of 16.7% [64]. Yet, MET alterations have been found in both circulating-free DNA (cfDNA) and circulating tumor cells (CTCs) both at diagnosis and at resistance to EGFR TKIs [65]. Moreover, Peng et al. examined 48 paired samples and showed a 92.4% concordance between the absolute copy number variant > 6 and the NGS detection of MET amplification in tumor tissue [66]. This all has significant ramifications clinically when it comes to making sure MET dysregulation is captured on diagnosis but then also on acquired resistance because sometimes patients may not have adequate tissue for testing, which limits them only to liquid biopsy testing, or clinicians may choose to only perform liquid biopsy testing upon the progression of the disease. Thus, finding a trustable biomarker, whether it is a specific MET GCN or MET/CEP7 ratio threshold that can be used in both tissue testing and ctDNA testing, will go a long way towards determining which MET amplification patients would benefit from capmatinib and other MET-targeted agents and to ensure that as many MET exon 14 skipping mutations are detected as possible.
In MET exon 14 skipping mutations, there is also some variability in the ORR, with ranges from 32% to 64%, though these studies do originate from patients on different lines of therapy and different MET TKI inhibitors [30,31,41]. However, in the GEOMETRY mono-1 trial, a clinical bridging study was carried out to show analytical and clinical agreement between the enrollment assay and the Foundation One CDx assay [59,67]. The Foundation One CDx assay, developed by Foundation Medicine in collaboration with Novartis, is performed at Foundation Medicine Inc. using DNA isolated from fresh-frozen paraffin-embedded (FFPE) tumor tissue specimens. In previously treated patients, the positive percent agreement (PPA) was 96.8%, the negative percent agreement (NPA) was 100%, and the overall agreement (OA) was 100%. In untreated patients, the PPA, NPA, and OA were all 100%. This led to the FDA approval of the Foundation One CDx assay as the only assay associated with a MET inhibitor [59,67].

9. Toxicities

In the GEOMETRY mono-1 trial, across all cohorts, the most reported adverse events were peripheral edema, nausea, and vomiting. Notably, 67% of patients had adverse events of Grade 3 or 4; the most frequent of these were peripheral edema, nausea, vomiting, and increased blood creatinine level. Treatment-related adverse events led to the discontinuation of treatment in 39 patients (11%), with treatment-related peripheral edema leading to discontinuation in 6 patients (2%) [6].
In the VISION study, 28% of patients had Grade 3–4 adverse events, with peripheral edema (7%) being the greatest [31]. Other Grade 3–4 adverse events with greater than 1% incidence included increased amylase (3%), increased lipase (3%), pleural effusion (3%), increased ALT (3%), increased AST (2%), and general edema (3%) [31]. Meanwhile, in the study involving savolitinib, treatment-related adverse events occurred in 46% of the patients, with increased aspartate aminotransferase (n = 9), alanine aminotransferase (n = 7), and peripheral edema (n = 6) being the most common serious adverse side effect. There was one death in the study due to tumor lysis syndrome, likely treatment-related [30] (Table 5).

10. Discussion and Future Directions

Although capmatinib has been approved by both the FDA and EMA, there has not been a Phase III trial comparing capmatinib versus chemotherapy and immunotherapy in the first-line setting for MET exon 14 skipping mutations despite the National Comprehensive Cancer Network (NCCN) recommending capmatinib as first-line therapy in advanced NSCLC with MET exon 14 skipping mutations [68]. In pretreated populations, the GEOMETRY-III (NCT04427072) trial is a study that involves approximately 90 previously treated advanced NSCLC patients harboring MET exon 14 skipping mutation and compares the efficacy of capmatinib with docetaxel [69]. Furthermore, capmatinib has been studied in 20 patients previously treated with a MET inhibitor, including 15 with MET exon 14 skipping mutation. The DCR was 80%. Notably, circulating tumor DNA analysis was carried out on these patients, and a secondary MET mutation was detected in four patients with MET D1228H and Y1230H, along with three patients having MAPK signaling alterations [70]. Furthermore, capmatinib and other Type Ib MET inhibitors have not been directly compared with Type Ia MET inhibitors.
Meanwhile, the challenge remains in finding reliable combinations to both improve the efficacy of capmatinib and broaden the indications of capmatinib use beyond MET exon 14 skipping mutations (Table 6).
Within population subgroups, there are ongoing studies on capmatinib in Asia, which may give insight into its efficacy within specific Asian subgroup populations, including one in China (GEOMETRY-C study, NCT04677595) and one in India (NCT05110196). For early-stage NSCLC, the GEOMETRY-N (NCT04926831) study is a Phase II, two-cohort, two-stage study evaluating the efficacy and safety of neoadjuvant and adjuvant capmatinib therapy in improving the major pathological rate (MPR) and outcomes in patients with MET exon 14 skipping or high-level MET amplification NSCLC [71]. As there has been success with EGFR mutations and the use of osimertinib in an adjuvant setting with the ADUARA trial, it will be interesting to note the results of the major pathological response rate in this study [72].
Currently, there is a Phase I/Ib trial underway that investigates capmatinib and trametinib, a MEK inhibitor (NCT05435846), which may be of benefit to patients with progression on crizotinib. Meanwhile, there has not been much success with capmatinib in combination with immunotherapy due to limited activity and tolerability. A retrospective study at two academic institutions showed an ORR of 17% (95% CI 6–36) in MET exon 14 skip mutations receiving PD-L1 blockade [73]. A Phase II study (NCT04323436) looking at the efficacy and safety of capmatinib plus spartalizumab, a PD-1 monoclonal antibody, did not demonstrate significant antitumor benefit, with a high dose reduction/interruption (80.6%) and discontinuation rate (35.5%) [74]. Another Phase II randomized, open-label study (NCT04139317) evaluated the efficacy and safety of combination therapy with capmatinib and pembrolizumab versus pembrolizumab alone in first-line therapy among advanced NSCLC patients with PD-L1 tumor proportion score (TPS) ≥ 50% and no EGFR mutation or ALK rearrangements. However, the trial closed due to concerns from the drug sponsor of tolerability in patients [75]. Finally, there was another study that investigated the efficacy of capmatinib plus nivolumab or nazartinib (EGF816) plus nivolumab in previously treated NSCLC patients (NCT02323126). This study was also terminated due to low accrual, but in its primary endpoint of PFS at 6 months, capmatinib plus nivolumab showed a 68.9% (95% CI 48.85–85.7) PFS at 6 months in high cMet and 50.9% (95% CI 35.6–66.4) in low cMet (NCT02323126). However, there continue to be clinical trials, particularly with cabozantinib and atezolizumab (NCT03170960 and NCT04471428) targeting the MET pathway, as MET expression has been found to be implicated through its pathway with MET/HGF and is involved in the regulation of the inflamed tumor microenvironment, leading towards the upregulation of inhibitory molecules such as PD-L1 and the downregulation of immune stimulators such as CD137, CD252, and CD70 [76].
Another important role of capmatinib in the future is in patients with acquired MET amplification, as observed in about 15% of patients who received first-line osimertinib and in 12–22% of patients receiving second-line osimertinib [60]. As mentioned earlier, Wu et al. saw efficacy using capmatinib and gefitinib, and the TATTON trial, which incorporated osmertinib and savolitinib, showed ORR of 23–66% between the two arms of treatment [51,77]. The GEOMETRY-E study (NCT04816214) was a Phase III study involving osimertinib with capmatinib but recently closed due to a business decision, but a recent Phase II LUNG-MAP trial with SWOG (NCT05642572) recently opened that investigates capmatinib with osimertinib +/− ramucirumab in EGFR mutant, MET-amplified Stage IV or recurrent NSCLC. Meanwhile, NCT03040973 is a rollover study currently accruing in patients who were part of a Novartis-sponsored clinical trial to continue receiving capmatinib as a single agent or in combination with other treatments.
As with all TKIs, it will be important to note the recurring resistance mechanisms with capmatinib to aid with future directions. Previous studies have shown that in Type I MET TKIs, secondary mutations at residue Y1230 may cause resistance, as Type I MET TKIs do interact with Y1230, specifically Y1230C [42,78,79]. However, notably, D1228 mutations have also been seen in capmatinib and other Type I TKIs [42,46,79,80].
While switching to Type II MET TKIs has been believed to help overcome resistance to capmatinib, novel drugs that can bypass the MET signaling pathway may provide the answer for treatment in the post-capmatinib treatment setting [79]. Amivantamab, a bispecific, monoclonal antibody targeting EGFR and MET is a promising combination that can be considered in conjunction with capmatinib. In the CHYRSALIS study specifically involving patients with MET exon 14 skipping mutation whose disease had progressed or had declined standard-of-care therapy, the ORR was 21% (4/19) in patients with prior MET inhibitor therapy and 46% (5/11) in patients with no prior MET inhibitor therapy. The median DOR was not reached, and 67% (8/13) had DOR ≥ 6 months [81]. Meanwhile, in another cohort of patients with EGFR exon 19 deletion or L858R NSCLC who had progressed on an EGFR TKI, ORR with amivantamab and lazertinib, an EGFR inhibitor, was 36% (95% CI 23–51), and 39% had a DOR ≥ 6 months [82]. An ongoing clinical trial (NCT05488314) is currently underway that investigates the combination of amivantamab and capmatinib in advanced NSCLC with MET exon 14 skipping mutation or MET amplification and may provide a promising new combination. Another promising class of novel drugs includes antibody–drug conjugates (ADCs) in which the monoclonal antibody binds to a specific protein and can deliver a cytotoxic drug to its intended target [83]. telisotuzumab vedotin (Teliso-V) is an antibody–drug conjugate composed of a c-Met antibody (ABT-700) and a microtubule inhibitor (monomethyl auristatin E); the ongoing Phase II M14-239 LUMINOSITY trial (NCT03539536) showed a 52% ORR in patients with previously treated c-MET overexpressors with nonsquamous pathology and EGFR wild-type [84]. ABBV-400 is another ADC, which targets c-Met and topoisomerase-1, with an ongoing Phase I study (NCT05029882) involving c-Met overexpression in advanced solid tumors. In addition, a biparatopic MET x MET ADC REGN 5093-M114 has shown promising preclinical activity in both MET-overexpressed, TKI-naïve, EGFR-mutant NSCLC cells regardless of MET gene copy number as well as cell lines of EGFR-mutant NSCLC with PTEN loss or MET Y1230C mutation after the progression of prior osimertinib and savolitinib treatment [85]. A Phase I study (NCT04982224) is ongoing that involves the study of REGN5093-M114 in MET overexpression in advanced solid tumors.
Finally, it is worth noting the tolerability of capmatinib, as 67% of patients in the GEOMETRY mono-1 trial had a Grade 3 or 4 toxicity, and 42% of patients had serious adverse events [6]. The most frequent etiologies for Grade 3–4 toxicity include peripheral edema (9%), dyspnea (7%), fatigue (4%), and asthenia (4%), which all can severely impact the quality of life in patients [6]. While some of these side effects like peripheral edema can be controlled with supportive care, the toxicity profile of capmatinib merits further comparison with other standard-of-care options in a Phase III study and real-world prospective studies that evaluate side effects of capmatinib in clinical practice [86].
Thus, future directions in capmatinib and other combinations and novel agents in MET-dysregulated NSCLC will focus on the efficacy of these drugs, tolerability, and given the multiple new drugs, the sequence of these agents.

11. Conclusions

The dysregulation of MET in NSCLC has proven challenging when it comes to finding therapeutic options given the lack of activity and reliability of biomarkers. Capmatinib, a Type Ib MET TKI that is not dependent on G1163, as crizotinib is, has proven to have efficacy, as shown in the GEOMETRY mono-1 study. Subsequent post hoc analyses have shown similar efficacy regardless of the prior treatment used and patient-reported improvement in quality of life. In addition, real-world analysis has shown similar efficacy with a promising intracranial response. The Foundation One CDx assay has been shown to be a reliable companion assay and remains the only FDA-approved assay for MET-targeted therapies. However, there have been no completed Phase III studies comparing capmatinib to first-line chemotherapy and immunotherapy or second-line chemotherapy. Furthermore, there was a notable percentage of Grade 3–4 toxicities. Future studies include investigations of capmatinib with MEK inhibition, combination therapy with amivantamab, and new classes of drugs, particularly ADCs. Capmatinib’s role in a perioperative setting in early-stage NSCLC may provide further treatment options for early stage patients with MET exon 14 skipping NSCLC, but the sequencing of these drugs and tolerability will be key factors, along with finding a more reliable biomarker.

Author Contributions

Conceptualization, M.N.; formal analysis, R.H., D.J.B. and M.N.; investigation, R.H., D.J.B. and M.N.; writing—original draft preparation, R.H., D.J.B. and M.N.; writing—review and editing, R.H., D.J.B. and M.N.; visualization, R.H. and D.J.B.; supervision, M.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

RH is a consultant for Targeted Oncology and received honoraria from DAVA Oncology and The Dedham Group. D.J.B. has received consulting fees from Seagen. MN has received consulting fees from AstraZeneca, Caris Life Sciences, Daiichi Sankyo, Novartis, EMD Serono, Pfizer, Lilly and Genentech, has received travel support from AnHeart Therapeutics and is a speaker for Takeda, Janssen, Mirati and Blueprint Medicines.

Abbreviations

NSCLCNon-small cell lung cancer
EGFREpidermal growth factor receptor
KRASKirsten rat sarcoma virus
BRAFv-raf murine sarcoma viral oncogene homolog B1
ALKAnaplastic lymphoma kinase
ROS1Proto-oncogene tyrosine–protein kinase ROS
RETRearranged during transfection proto-oncogene
METMesenchymal–epithelial transition
ERBB2erb-b2 receptor tyrosine kinase 2
NTRKNeurotrophic tyrosine receptor kinase
HGFHepatocyte growth factor
AMPKAMP-activated protein kinase
LKB1Liver kinase B1
GCNGain of copy number
FDAU.S. Food and Drug Administration
EMAEuropean Medicines Agency
poOral
DLTDrug limiting toxicity
b.i.d.Twice a day
R2PDRecommended Phase II dose
ORROverall response rate
PFSProgression-free survival
DORDuration of response
IOImmunotherapy
IHCImmunohistochemistry
CEP7Chromosome 7 centromere
ctDNACirculating tumor DNA
cfDNACirculating-free DNA
CTCsCirculating tumor cells
FFPEFresh-frozen paraffin-embedded
PPAPositive percent agreement
NPANegative percent agreement
OAOverall agreement
NCCNNational Comprehensive Cancer Network
Teliso-VTelisotuzumab vedotin

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus trametinib in patients with previously untreated BRAFV600E-mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef] [PubMed]
  3. Shaw, A.T.; Riely, G.J.; Bang, Y.-J.; Kim, D.-W.; Camidge, D.R.; Solomon, B.J.; Varella-Garcia, M.; Iafrate, A.J.; Shapiro, G.I.; Usari, T.; et al. Crizotinib in ROS1-rearranged advanced non-small-cell lung cancer (NSCLC): Updated results, including overall survival, from PROFILE 1001. Ann Oncol. 2019, 30, 1121–1126. [Google Scholar] [CrossRef] [PubMed]
  4. Drilon, A.; Siena, S.; Dziadziuszko, R.; Barlesi, F.; Krebs, M.G.; Shaw, A.T.; de Braud, F.; Rolfo, C.; Ahn, M.-J.; Wolf, J.; et al. Entrectinib in ROS1 fusion-positive non-small-cell lung cancer: Integrated analysis of three phase 1–2 trials. Lancet Oncol. 2020, 21, 261–270. [Google Scholar] [CrossRef] [PubMed]
  5. Drilon, A.; Oxnard, G.R.; Tan, D.S.W.; Loong, H.H.F.; Johnson, M.; Gainor, J.; McCoach, C.E.; Gautschi, O.; Besse, B.; Cho, B.C.; et al. Efficacy of Selpercatinib in RET Fusion-Positive Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 813–824. [Google Scholar] [CrossRef]
  6. Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14–Mutated or MET -Amplified Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
  7. Ramalingam, S.S.; Yang, J.C.-H.; Lee, C.K.; Kurata, T.; Kim, D.-W.; John, T.; Nogami, N.; Ohe, Y.; Mann, H.; Rukazenov, Y.; et al. Osimertinib As First-Line Treatment of EGFR Mutation-Positive Advanced Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 841–849. [Google Scholar] [CrossRef] [Green Version]
  8. Skoulidis, F.; Li, B.T.; Dy, G.K.; Price, T.J.; Falchook, G.S.; Wolf, J.; Italiano, A.; Schuler, M.; Borghaei, H.; Barlesi, F.; et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N. Engl. J. Med. 2021, 384, 2371–2381. [Google Scholar] [CrossRef]
  9. Doebele, R.C.; Drilon, A.; Paz-Ares, L.; Siena, S.; Shaw, A.T.; Farago, A.F.; Blakeley, C.M.; Seto, T.; Cho, B.C.; Tosi, D.; et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: Integrated analysis of three phase 1-2 trials. Lancet Oncol. 2020, 21, 271–282. [Google Scholar] [CrossRef]
  10. Li, B.T.; Smit, E.F.; Goto, Y.; Nakagawa, K.; Udagawa, H.; Mazières, J.; Nagasaka, M.; Bazhenova, L.; Saltos, A.N.; Felip, E.; et al. Trastuzumab Deruxtecan in HER2 -Mutant Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2022, 386, 241–251. [Google Scholar] [CrossRef]
  11. Zhang, Y.; Xia, M.; Jin, K.; Wang, S.; Wei, H.; Fan, C.; Wu, Y.; Li, X.; Li, X.; Li, G.; et al. Function of the c-Met receptor tyrosine kinase in carcinogenesis and associated therapeutic opportunities. Mol. Cancer 2018, 17, 45. [Google Scholar] [CrossRef] [PubMed]
  12. Cecchi, F.; Rabe, D.C.; Bottaro, D.P. Targeting the HGF/Met signalling pathway in cancer. Eur. J. Cancer 2010, 46, 1260–1270. [Google Scholar] [CrossRef] [Green Version]
  13. Pilotto, S.; Carbognin, L.; Karachaliou, N.; Ma, P.C.; Rosell, R.; Tortora, G.; Bria, E. Tracking MET de-addiction in lung cancer: A road towards the oncogenic target. Cancer Treat. Rev. 2017, 60, 1–11. [Google Scholar] [CrossRef] [PubMed]
  14. Lee, M.; Jain, P.; Wang, F.; Ma, P.C.; Borczuk, A.; Halmos, B. MET alterations and their impact on the future of non-small cell lung cancer (NSCLC) targeted therapies. Expert Opin. Ther. Targets 2021, 25, 249–268. [Google Scholar] [CrossRef] [PubMed]
  15. Salgia, R. MET in Lung Cancer: Biomarker Selection Based on Scientific Rationale. Mol. Cancer Ther. 2017, 16, 555–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Tu, C.-Y.; Cheng, F.-J.; Chen, C.-M.; Wang, S.-L.; Hsiao, Y.-C.; Chen, C.-H.; Tsia, T.-C.; He, Y.-H.; Wang, B.-W.; Hsieh, I.-S.; et al. Cigarette smoke enhances oncogene addiction to c-MET and desensitizes EGFR-expressing non-small cell lung cancer to EGFR TKIs. Mol. Oncol. 2018, 12, 705–723. [Google Scholar] [CrossRef]
  17. Cheng, F.-J.; Chen, C.-H.; Tsai, W.-C.; Wang, B.-W.; Yu, M.-C.; Hsia, T.-C.; Wei, Y.-L.; Hsiao, Y.-C.; Hu, D.-W.; Ho, C.-Y.; et al. Cigarette smoke-induced LKB1/AMPK pathway deficiency reduces EGFR TKI sensitivity in NSCLC. Oncogene 2021, 40, 1162–1175. [Google Scholar] [CrossRef]
  18. Hong, L.; Zhang, J.; Heymach, J.V.; Le, X. Current and future treatment options for MET exon 14 skipping alterations in non-small cell lung cancer. Ther. Adv. Med. Oncol. 2021, 13, 1758835921992976. [Google Scholar] [CrossRef]
  19. Cortot, A.B.; Kherrouche, Z.; Descarpentries, C.; Wislez, M.; Baldacci, S.; Furlan, A.; Tulasne, D. Exon 14 Deleted MET Receptor as a New Biomarker and Target in Cancers. JNCI J. Natl. Cancer Inst. 2017, 109, djw262. [Google Scholar] [CrossRef] [Green Version]
  20. Socinski, M.A.; Pennell, N.A.; Davies, K.D. MET Exon 14 Skipping Mutations in Non–Small-Cell Lung Cancer: An Overview of Biology, Clinical Outcomes, and Testing Considerations. JCO Precis. Oncol. 2021, 5, 653–663. [Google Scholar] [CrossRef]
  21. Tong, J.H.; Yeung, S.F.; Chan, A.W.H.; Chung, L.Y.; Chau, S.L.; Lung, R.W.M.; Tong, C.Y.; Chow, C.; Tin, E.K.Y.; Yu, Y.H.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Schubart, C.; Stöhr, R.; Tögel, L.; Fuchs, F.; Sirbu, H.; Seitz, G.; Seggewiss-Bernhardt, R.; Leistner, R.; Sterlacci, W.; Vieth, M.; et al. MET Amplification in Non-Small Cell Lung Cancer (NSCLC)—A Consecutive Evaluation Using Next-Generation Sequencing (NGS) in a Real-World Setting. Cancers 2021, 13, 5023. [Google Scholar] [CrossRef] [PubMed]
  23. Leonetti, A.; Sharma, S.; Minari, R.; Perego, P.; Giovannetti, E.; Tiseo, M. Resistance mechanisms to osimertinib in EGFR-mutated non-small cell lung cancer. Br. J. Cancer 2019, 121, 725–737. [Google Scholar] [CrossRef]
  24. Chmielecki, J.; Mok, T.; Wu, Y.-L.; Han, J.-Y.; Ahn, M.; Ramalingam, S.S.; John, T.; Okamoto, I.; Yang, J.C.-H.; Shepherd, F.A.; et al. Analysis of acquired resistance mechanisms to osimertinib in patients with EGFR-mutated advanced non-small cell lung cancer from the AURA3 trial. Nat Commun. 2023, 14, 1071. [Google Scholar] [CrossRef] [PubMed]
  25. Dagogo-Jack, I.; Yoda, S.; Lennerz, J.K.; Langenbucher, A.; Lin, J.J.; Rooney, M.M.; Prutisto-Chang, K.; Oh, A.; Adams, N.A.; Yeap, B.Y.; et al. MET Alterations Are a Recurring and Actionable Resistance Mechanism in ALK-Positive Lung Cancer. Clin. Cancer Res. 2020, 26, 2535–2545. [Google Scholar] [CrossRef]
  26. Awad, M.M.; Leonardi, G.C.; Kravets, S.; Dahlberg, S.E.; Drilon, A.; Noonan, S.A.; Camidge, D.R.; Ou, S. H-I; Costa, D.B.; Gadgeel, S.M.; et al. Impact of MET inhibitors on survival among patients with non-small cell lung cancer harboring MET exon 14 mutations: A retrospective analysis. Lung Cancer 2019, 133, 96–102. [Google Scholar] [CrossRef]
  27. Awad, M.M.; Oxnard, G.R.; Jackman, D.M.; Savukoski, D.O.; Hall, D.; Shivdasani, P.; Heng, J.C.; Dahlberg, S.E.; Janne, P.A.; Verma, S.; 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. [Google Scholar] [CrossRef] [Green Version]
  28. Bahcall, M.; Paweletz, C.P.; Kuang, Y.; Taus, L.J.; Sim, T.; Kim, N.D.; Dholakia, K.H.; Lau, C.J.; Gokhale, P.C.; Chopade, P.R.; et al. Combination of Type I and Type II MET Tyrosine Kinase Inhibitors as Therapeutic Approach to Prevent Resistance. Mol. Cancer Ther. 2022, 21, 322–335. [Google Scholar] [CrossRef]
  29. Cui, J.J. Targeting receptor tyrosine kinase MET in cancer: Small molecule inhibitors and clinical progress. J. Med. Chem. 2014, 57, 4427–4453. [Google Scholar] [CrossRef]
  30. Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Once-daily savolitinib in Chinese patients with pulmonary sarcomatoid carcinomas and other non-small-cell lung cancers harbouring MET exon 14 skipping alterations: A multicentre, single-arm, open-label, phase 2 study. Lancet Respir. Med. 2021, 9, 1154–1164. [Google Scholar] [CrossRef]
  31. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Van Meerbeeck, J.; et al. Tepotinib in Non–Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. N. Engl. J. Med. 2020, 383, 931–943. [Google Scholar] [CrossRef] [PubMed]
  32. Santarpia, M.; Massafra, M.; Gebbia, V.; D’Aquino, A.; Garipoli, C.; Altavilla, G.; Rosell, R. A narrative review of MET inhibitors in non-small cell lung cancer with MET exon 14 skipping mutations. Transl. Lung Cancer Res. 2021, 10, 1536–1556. [Google Scholar] [CrossRef] [PubMed]
  33. Yan, S.B.; Um, S.L.; Peek, V.L.; Stephens, J.R.; Zeng, W.; Konicek, B.W.; Ling, L.; Manro, J.R.; Wacheck, W.; Walgren, R.A. MET-targeting antibody (emibetuzumab) and kinase inhibitor (merestinib) as single agent or in combination in a cancer model bearing MET exon 14 skipping. Investig. New Drugs 2018, 36, 536–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Calles, A.; Kwiatkowski, N.; Cammarata, B.K.; Ercan, D.; Gray, N.S.; Jänne, P.A. Tivantinib (ARQ 197) efficacy is independent of MET inhibition in non-small-cell lung cancer cell lines. Mol. Oncol. 2015, 9, 260–269. [Google Scholar] [CrossRef]
  35. Klempner, S.J.; Borghei, A.; Hakimian, B.; Ali, S.M.; Ou, S.-H.I. Intracranial Activity of Cabozantinib in MET Exon 14-Positive NSCLC with Brain Metastases. J. Thorac. Oncol. 2017, 12, 152–156. [Google Scholar] [CrossRef] [Green Version]
  36. Scagliotti, G.; von Pawel, J.; Novello, S.; Ramlau, R.; Favaretto, A.; Barlesi, F.; Akerley, W.; Orlov, S.; Santoro, A.; Spigel, D.; et al. Phase III Multinational, Randomized, Double-Blind, Placebo-Controlled Study of Tivantinib (ARQ 197) Plus Erlotinib Versus Erlotinib Alone in Previously Treated Patients With Locally Advanced or Metastatic Nonsquamous Non–Small-Cell Lung Cancer. J. Clin. Oncol. 2015, 33, 2667–2674. [Google Scholar] [CrossRef]
  37. Tabrecta. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/tabrecta (accessed on 1 July 2023).
  38. Novartis. TABRECTA (Capmatinib): US Prescribing Information. 2020. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/213591s000lbl.pdf (accessed on 8 March 2023).
  39. Moro-Sibilot, D.; Cozic, N.; Pérol, M.; Mazières, J.; Otto, J.; Souquet, P.J.; Bahleda, R.; Wislez, M.; Zalcman, G.; Guibert, S.D. Crizotinib in c-MET- or ROS1-positive NSCLC: Results of the AcSé phase II trial. Ann. Oncol. 2019, 30, 1985–1991. [Google Scholar] [CrossRef]
  40. Landi, L.; Chiari, R.; Tiseo, M.; D’Incà, F.; Dazzi, C.; Chella, A.; Delmonte, A.; Bonanno, L.; Giannarell, D.; Cortinovis, D.L.; et al. Crizotinib in MET -Deregulated or ROS1 -Rearranged Pretreated Non–Small Cell Lung Cancer (METROS): A Phase II, Prospective, Multicenter, Two-Arms Trial. Clin. Cancer Res. 2019, 25, 7312–7319. [Google Scholar] [CrossRef] [Green Version]
  41. Drilon, A.; Clark, J.W.; Weiss, J.; Ou, S.-H.I.; Camidge, D.R.; Solomon, B.J.; Otterson, G.A.; Villaruz, L.C.; Riely, G.J.; Heist, R.S.; et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat. Med. 2020, 26, 47–51. [Google Scholar] [CrossRef]
  42. Recondo, G.; Bahcall, M.; Spurr, L.F.; Che, J.; Ricciuti, B.; Leonardi, G.C.; Lo, Y.-C.; Li, Y.Y.; Lamberti, G.; Nguyen, T.; et al. Molecular Mechanisms of Acquired Resistance to MET Tyrosine Kinase Inhibitors in Patients with MET Exon 14-Mutant NSCLC. Clin. Cancer Res. 2020, 26, 2615–2625. [Google Scholar] [CrossRef]
  43. Xalkori (Crizotinib). Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/202570s021lbl.pdf (accessed on 1 July 2023).
  44. Xalkori, INN-Crizotinib. Available online: https://www.ema.europa.eu/en/documents/product-information/xalkori-epar-product-information_en.pdf (accessed on 1 July 2023).
  45. Liu, X.; Wang, Q.; Yang, G.; Marando, C.; Koblish, H.K.; Hall, L.M.; Fridman, J.S.; Behshad, E.; Wynn, R.; Li, Y.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Baltschukat, S.; Engstler, B.S.; Huang, A.; Hao, H.-X.; Tam, A.; Wang, H.Q.; Liang, J.; DiMare, M.T.; Bhang, H.-E.C.; Wang, Y.; 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Dhillon, S. Capmatinib: First Approval. Drugs 2020, 80, 1125–1131. [Google Scholar] [CrossRef] [PubMed]
  48. Esaki, T.; Hirai, F.; Makiyama, A.; Seto, T.; Bando, H.; Naito, Y.; Yoh, K.; Ishihara, K.; Kakizume, T.; Natsume, K.; et al. Phase I dose-escalation study of capmatinib (INC280) in Japanese patients with advanced solid tumors. Cancer Sci. 2019, 110, 1340–1351. [Google Scholar] [CrossRef]
  49. Bang, Y.-J.; Su, W.-C.; Schuler, M.; Nam, D.-H.; Lim, W.T.; Bauer, T.M.; Azaro, A.; Poon, R.T.P.; Hong, D.; Lin, C.-C.; et al. Phase 1 study of capmatinib in MET-positive solid tumor patients: Dose escalation and expansion of selected cohorts. Cancer Sci. 2020, 111, 536–547. [Google Scholar] [CrossRef] [Green Version]
  50. Schuler, M.; Berardi, R.; Lim, W.-T.; de Jonge, M.; Bauer, T.M.; Azaro, A.; Gottfried, M.; Han, J.-Y.; Lee, D.-H.; Wollner, M.; et al. Molecular correlates of response to capmatinib in advanced non-small-cell lung cancer: Clinical and biomarker results from a phase I trial. Ann. Oncol. 2020, 31, 789–797. [Google Scholar] [CrossRef]
  51. Wu, Y.-L.; Zhang, L.; Kim, D.-W.; Liu, X.; Lee, D.H.; Yang, J.C.-H.; Ahn, M.-J.; Vansteenkiste, J.F.; Su, W.-C.; Felip, E.; et al. Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients With EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2018, 36, 3101–3109. [Google Scholar] [CrossRef]
  52. Vansteenkiste, J.F.; Smit, E.F.; Groen, H.J.M.; Garon, E.B.; Heist, R.S.; Hida, T.; Nishio, M.; Kokowski, K.; Grohe, C.; Reguart, N.; et al. 1285P Capmatinib in patients with METex14-mutated advanced non-small cell lung cancer who received prior immunotherapy: The phase II GEOMETRY mono-1 study. Ann. Oncol. 2020, 31, S830. [Google Scholar] [CrossRef]
  53. Wolf, J.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Gilloteau, I.; Le Mouhaer, S.; Hampe, M.; Cai, C.; Chassot-Agostinho, A.; Reynolds, M.; et al. Patient-reported outcomes in capmatinib-treated patients with METex14-mutated advanced NSCLC: Results from the GEOMETRY mono-1 study. Eur. J. Cancer 2023, 183, 98–108. [Google Scholar] [CrossRef]
  54. Seto, T.; Ohashi, K.; Sugawara, S.; Nishio, M.; Takeda, M.; Aoe, K.; Moizumi, S.; Nomura, S.; Tajima, T.; Hida, T. Capmatinib in Japanese patients with MET exon 14 skipping-mutated or MET-amplified advanced NSCLC: GEOMETRY mono-1 study. Cancer Sci. 2021, 112, 1556–1566. [Google Scholar] [CrossRef]
  55. Paik, P.K.; Goyal, R.K.; Cai, B.; Price, M.A.; Davis, K.L.; Ansquer, V.D.; Caro, N.; Saliba, T.R. Real-world outcomes in non-small-cell lung cancer patients with MET Exon 14 skipping mutation and brain metastases treated with capmatinib. Future Oncol. 2023, 19, 217–228. [Google Scholar] [CrossRef]
  56. Illini, O.; Fabikan, H.; Swalduz, A.; Vikström, A.; Krenbek, D.; Schumacher, M.; Dudnik, E.; Studnicka, M.; Ohman, R.; Wurm, R.; et al. Real-world experience with capmatinib in MET exon 14-mutated non-small cell lung cancer (RECAP): A retrospective analysis from an early access program. Ther. Adv. Med. Oncol. 2022, 14, 17588359221103206. [Google Scholar] [CrossRef]
  57. TEPMETKO, INN. Available online: https://www.ema.europa.eu/en/documents/product-information/tepmetko-epar-product-information_en.pdf (accessed on 1 July 2023).
  58. Markham, A. Savolitinib: First Approval. Drugs 2021, 81, 1665–1670. [Google Scholar] [CrossRef] [PubMed]
  59. Jørgensen, J.T.; Mollerup, J. Companion Diagnostics and Predictive Biomarkers for MET-Targeted Therapy in NSCLC. Cancers 2022, 14, 2150. [Google Scholar] [CrossRef] [PubMed]
  60. Coleman, N.; Hong, L.; Zhang, J.; Heymach, J.; Hong, D.; Le, X. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open 2021, 6, 100319. [Google Scholar] [CrossRef] [PubMed]
  61. Noonan, S.A.; Berry, L.; Lu, X.; Gao, D.; Barón, A.E.; Chesnut, P.; Sheren, J.; Aisner, D.L.; Merrick, D.; Doebele, R.C.; et al. Identifying the Appropriate FISH Criteria for Defining MET Copy Number-Driven Lung Adenocarcinoma through Oncogene Overlap Analysis. J. Thorac. Oncol. 2016, 11, 1293–1304. [Google Scholar] [CrossRef] [Green Version]
  62. Guo, R.; Offin, M.; Brannon, A.R.; Chang, J.; Chow, A.; Delasos, L.; Girshman, J.; Wilkins, O.; McCarthy, C.G.; Makhnin, A. MET Exon 14-altered Lung Cancers and MET Inhibitor Resistance. Clin. Cancer Res. 2021, 27, 799–806. [Google Scholar] [CrossRef]
  63. Ikeda, S.; Schwaederle, M.; Mohindra, M.; Fontes Jardim, D.L.; Kurzrock, R. MET alterations detected in blood-derived circulating tumor DNA correlate with bone metastases and poor prognosis. J. Hematol. Oncol. 2018, 11, 76. [Google Scholar] [CrossRef] [Green Version]
  64. Park, S.; Olsen, S.; Ku, B.M.; Lee, M.-S.; Jung, H.-A.; Sun, J.-M.; Lee, S.-H.; Ahn, J.S.; Park, K.; Choi, Y.-L.; et al. High concordance of actionable genomic alterations identified between circulating tumor DNA–based and tissue-based next-generation sequencing testing in advanced non–small cell lung cancer: The Korean Lung Liquid Versus Invasive Biopsy Program. Cancer 2021, 127, 3019–3028. [Google Scholar] [CrossRef]
  65. Mondelo-Macía, P.; Rodríguez-López, C.; Valiña, L.; Aguín, S.; León-Mateos, L.; García-González, J.; Abalo, A.; Rapado-Gonzalez, O.; Suarez-Cunqueiro, M.; Díaz-Lagares, A.; et al. Detection of MET Alterations Using Cell Free DNA and Circulating Tumor Cells from Cancer Patients. Cells 2020, 9, 522. [Google Scholar] [CrossRef] [Green Version]
  66. Peng, H.; Lu, L.; Zhou, Z.; Liu, J.; Zhang, D.; Nan, K.; Zhao, X.; Li, F.; Tian, L.; Dong, H.; et al. CNV Detection from Circulating Tumor DNA in Late Stage Non-Small Cell Lung Cancer Patients. Genes 2019, 10, 926. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. FDA Summary of Safety and Effectiveness Data. FoundationOne CDx. 2020. Available online: https://www.accessdata.fda.gov/cdrh_docs/pdf17/P170019S011B.pdf (accessed on 8 March 2023).
  68. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®) for Non-Small Cell Lung Cancer V.3.2023. Available online: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf (accessed on 15 June 2023).
  69. Souquet, P.; Kim, S.; Solomon, B.; Vansteenkiste, J.; Carbini, M.; Kenny, S.; Glaser, S.; Chassot Agostinho, A.; Wolf, J. P47.17 Capmatinib vs Docetaxel in Pretreated Patients With MET Exon 14 Skipping–mutated Stage IIIB/IIIC or IV NSCLC (GeoMETry-III). J. Thorac. Oncol. 2021, 16, S1104. [Google Scholar] [CrossRef]
  70. Dagogo-Jack, I.; Moonsamy, P.; Gainor, J.F.; Lennerz, J.K.; Piotrowska, Z.; Lin, J.J.; Lenne, I.T.; Sequist, L.V.; Shaw, A.T.; Goodwin, K.; et al. A Phase 2 Study of Capmatinib in Patients With MET-Altered Lung Cancer Previously Treated With a MET Inhibitor. J. Thorac. Oncol. 2021, 16, 850–859. [Google Scholar] [CrossRef]
  71. Lee, J.M.; Awad, M.M.; Saliba, T.R.; Caro, N.; Banerjee, H.; Kelly, K. Neoadjuvant and adjuvant capmatinib in resectable non–small cell lung cancer with MET exon 14 skipping mutation or high MET amplification: GEOMETRY-N trial. J. Clin. Oncol. 2022, 40, TPS8590. [Google Scholar] [CrossRef]
  72. Wu, Y.-L.; Tsuboi, M.; He, J.; John, T.; Grohe, C.; Majem, M.; Goldman, J.W.; Laktionov, K.; Kim, S.-W.; Kato, T.; et al. Osimertinib in Resected EGFR -Mutated Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 1711–1723. [Google Scholar] [CrossRef]
  73. Sabari, J.K.; Leonardi, G.C.; Shu, C.A.; Umeton, R.; Montecalvo, J.; Ni, A.; Chen, R.; Dienstag, J.; Mrad, C.; Bergagnini, I.; et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann. Oncol. 2018, 29, 2085–2091. [Google Scholar] [CrossRef]
  74. Wolf, J.; Heist, R.; Kim, T.M.; Nishio, M.; Dooms, C.; Kanthala, R.R.; Leo, E.; Giorgetti, E.; Mardjuadi, F.I.; Corto, A. 994P Efficacy and safety of capmatinib plus spartalizumab in treatment-naïve patients with advanced NSCLC harboring MET exon 14 skipping mutation. Ann. Oncol. 2022, 33, S1007–S1008. [Google Scholar] [CrossRef]
  75. Mok, T.S.K.; Cortinovis, D.L.; Majem, M.; Johnson, M.L.; Mardjuadi, F.I.; Zhao, X.; Siripurapu, S.V.; Jiang, Z.; Wolf, J. Efficacy and safety of capmatinib plus pembrolizumab in treatment (tx)-naïve patients with advanced non–small cell lung cancer (NSCLC) with high tumor PD-L1 expression: Results of a randomized, open-label, multicenter, phase 2 study. J. Clin. Oncol. 2022, 40, 9118. [Google Scholar] [CrossRef]
  76. Dempke, W.C.M.; Fenchel, K. Has programmed cell death ligand-1 MET an accomplice in non-small cell lung cancer?—A narrative review. Transl. Lung Cancer Res. 2021, 10, 2667–2682. [Google Scholar] [CrossRef]
  77. Sequist, L.V.; Han, J.-Y.; Ahn, M.-J.; Cho, B.C.; Yu, H.; Kim, S.-W.; Yang, J.C.-H.; Lee, J.S.; Su, W.-C.; Kowalski, D.; et al. Osimertinib plus savolitinib in patients with EGFR mutation-positive, MET-amplified, non-small-cell lung cancer after progression on EGFR tyrosine kinase inhibitors: Interim results from a multicentre, open-label, phase 1b study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef]
  78. Ou, S.-H.I.; Young, L.; Schrock, A.B.; Johnson, A.; Klempner, S.J.; Zhu, V.W.; Miller, V.A.; Ali, S.M. Emergence of Preexisting MET Y1230C Mutation as a Resistance Mechanism to Crizotinib in NSCLC with MET Exon 14 Skipping. J. Thorac. Oncol. 2017, 12, 137–140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Fujino, T.; Suda, K.; Koga, T.; Hamada, A.; Ohara, S.; Chiba, M.; Shimoji, M.; Takemoto, T.; Soh, J.; Mitsudomi, T. Foretinib can overcome common on-target resistance mutations after capmatinib/tepotinib treatment in NSCLCs with MET exon 14 skipping mutation. J. Hematol. Oncol. 2022, 15, 79. [Google Scholar] [CrossRef]
  80. Bahcall, M.; Sim, T.; Paweletz, C.P.; Patel, J.D.; Alden, R.S.; Kuang, Y.; Sacher, A.G.; Kim, N.D.; Lydon, C.A.; Awad, M.M.; et al. Acquired METD1228V Mutation and Resistance to MET Inhibition in Lung Cancer. Cancer Discov. 2016, 6, 1334–1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Krebs, M.; Spira, A.I.; Cho, B.C.; Besse, B.; Goldman, J.W.; Janne, P.A.; Ma, Z.; Mansfield, A.S.; Minchom, A.R.; Ou, S.-H.I.; et al. Amivantamab in patients with NSCLC with MET exon 14 skipping mutation: Updated results from the CHRYSALIS study. J. Clin. Oncol. 2022, 40, 9008. [Google Scholar] [CrossRef]
  82. Shu, C.A.; Goto, K.; Ohe, Y.; Besse, B.; Lee, S.-H.; Wang, Y.; Griesinger, F.; Yang, J.C.-H.; Felip, E.; Sanborn, R.E.; et al. Amivantamab and lazertinib in patients with EGFR-mutant non–small cell lung (NSCLC) after progression on osimertinib and platinum-based chemotherapy: Updated results from CHRYSALIS-2. J. Clin. Oncol. 2022, 40, 9006. [Google Scholar] [CrossRef]
  83. Fu, Z.; Li, S.; Han, S.; Shi, C.; Zhang, Y. Antibody drug conjugate: The “biological missile” for targeted cancer therapy. Signal Transduct. Target. Ther. 2022, 7, 93. [Google Scholar] [CrossRef]
  84. Camidge, D.R.; Bar, J.; Horinouchi, H.; Goldman, J.W.; Moiseenko, F.V.; Filippova, E.; Cicin, I.; Bradbury, P.A.; Daaboul, N.; Tomasini, P.; et al. Telisotuzumab vedotin (Teliso-V) monotherapy in patients (pts) with previously treated c-Met–overexpressing (OE) advanced non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2022, 40, 9016. [Google Scholar] [CrossRef]
  85. Oh, S.Y.; Lee, Y.W.; Lee, E.J.; Kim, J.H.; Park, Y.; Heo, S.G.; Yu, M.R.; Hong, M.H.; DaSilva, J.; Daly, C.; et al. Preclinical Study of a Biparatopic METxMET Antibody–Drug Conjugate, REGN5093-M114, Overcomes MET-driven Acquired Resistance to EGFR TKIs in EGFR-mutant NSCLC. Clin. Cancer Res. 2023, 29, 221–232. [Google Scholar] [CrossRef]
  86. Goodwin, K.; Ledezma, B.; Heist, R.; Garon, E. MO01.04 Management of Selected Adverse Events With Capmatinib: Institutional Experiences From the GEOMETRY Mono-1 Trial. J. Thorac. Oncol. 2021, 16, S16–S17. [Google Scholar] [CrossRef]
Figure 1. MET signaling pathway and blockade by MET inhibitors. In cancer, the MET proto-oncogene is abnormally activated and stimulates other signaling pathways in tumor cells, notably PI3K/AKT, JAK/STAT, Ras/MAPK, SRC, and Wnt/beta-catenin [11]. Type 1a inhibitor crizotinib blocks ATP binding to prevent the phosphorylation of the receptor, whereas type 1b inhibitors such as capmatinib are more specific and bind to a pocket adjacent to the ATP binding site. This figure was generated by BioRender.
Figure 1. MET signaling pathway and blockade by MET inhibitors. In cancer, the MET proto-oncogene is abnormally activated and stimulates other signaling pathways in tumor cells, notably PI3K/AKT, JAK/STAT, Ras/MAPK, SRC, and Wnt/beta-catenin [11]. Type 1a inhibitor crizotinib blocks ATP binding to prevent the phosphorylation of the receptor, whereas type 1b inhibitors such as capmatinib are more specific and bind to a pocket adjacent to the ATP binding site. This figure was generated by BioRender.
Cancers 15 03561 g001
Figure 2. Chemical structure of capmatinib; the asterisk (*) represents the chiral carbons that are part of the chemical structure.. The chemical name for capmatinib is 2-Fluoro-N-methyl-4-[7-(quinolin-6-ylmethyl)imidazo[1,2 b][1,2,4]triazin-2-yl]benzamide—hydrogen chloride—water (1/2/1). The molecular formula for capmatinib hydrochloride is C23H21Cl2FN6O2 [38].
Figure 2. Chemical structure of capmatinib; the asterisk (*) represents the chiral carbons that are part of the chemical structure.. The chemical name for capmatinib is 2-Fluoro-N-methyl-4-[7-(quinolin-6-ylmethyl)imidazo[1,2 b][1,2,4]triazin-2-yl]benzamide—hydrogen chloride—water (1/2/1). The molecular formula for capmatinib hydrochloride is C23H21Cl2FN6O2 [38].
Cancers 15 03561 g002
Table 1. Early-stage studies on capmatinib.
Table 1. Early-stage studies on capmatinib.
PublicationnIndicationR2PDORR
Esaki et al. [48]44 (15 NSCLC)Advanced solid tumors400 mg po bid
Bang et al. [49]38 (1 NSCLC)Advanced solid tumors600 mg po bid (capsule)/400 mg po bid (tablet)
Schuler et al. [50]55Advanced NSCLC600 mg po bid (capsule)/400 mg po bid (tablet)47%
Wu et al. [51]61 Phase Ib/100 Phase IIAdvanced NSCLC in patients with acquired EGFR TKI resistance400 mg po b.i.d. plus gefitinib 250 mg po daily27% (47% in patients with MET GCN ≥ 6)
Table 2. Responses to capmatinib treatment relative to the cohort in GEOMETRY mono-1 trial (6).
Table 2. Responses to capmatinib treatment relative to the cohort in GEOMETRY mono-1 trial (6).
ResponseNSCLC with MET Exon 14 Skipping Mutation NSCLC with MET Amplification
Best Response—No (%)Cohort 4 n = 69,
any GCN with 1–2 Lines of Therapy
Cohort 5b n = 28, any GCN with No Previous TherapyCohort 1a n = 69, GCN ≥ 10 with 1–2 Lines of Therapy Cohort 5a n = 15, GCN ≥ 10 with No Previous Therapy Cohort 1b n = 42, GCN 6–9 with 1–2 Lines of TherapyCohort 2 n = 54, GCN 4 or 5 with 1–2 Lines of TherapyCohort 3 n = 30, GCN < 4 with 1–2 Lines of Therapy
Complete response01 (4)1 (1)0000
Partial Response28 (41)18 (64)19 (28)6 (40)5 (12)5 (9)2 (7)
Stable disease 25 (36)7 (25)28 (41)4 (27)17 (40)20 (37)14 (47)
Incomplete response or nonprogressive disease1 (1)1 (4)1 (1)01 (2)00
Unknown or could not be evaluated9 (13)08 (12)1 (7)4 (10)8 (15)8 (27)
Overall response
No. of patients with overall response 2819206552
Percent of patients (95% CI)41 (29–53)68 (48–84)29 (19–41)40 (16–68)12 (4–26)9 (3–20)7 (1–22)
Disease control
No. of patients with disease control54274910232516
Percent of patients (95% CI)78 (67–87)96 (82–100)71 (59–81)67 (38–88)55 (39–70)46 (33–60)53 (34–72)
Duration of Response
No. of events/No. of patients with response23/2811/1915/206/63/54/52/2
Median duration of response (95% CI)—mo9.7 (5.6–13.0)12.6 (5.6–NE)8.3 (4.2–15.4)7.5 (2.6–14.3)24.9 (2.7–24.9)9.7 (4.2–NE)4.2 (4.2–4.2)
Progression-free survival
Progression or death—No. of patients60175815345022
Median progression-free survival (95% CI)—mo5.4 (4.2–7.0)12.4 (8.2–NE)4.1 (2.9–4.8)4.2 (1.4–6.9)2.7 (1.4–3.1)2.7 (1.4–4.1)3.6 (2.2–4.2)
Table 3. Key trials involving MET selective Type 1b inhibitors.
Table 3. Key trials involving MET selective Type 1b inhibitors.
Capmatinib [6]Tepotinib [31]Savolitinib [30]
N (with MET exon 14 skipping mutation)97152 (99 evaluable)84 (70 evaluable)
Overall response rate (%) (95% CI)68% (48–84) in untreated patients (n = 28) and 41 (29–53) in previously treated patients (n = 69) 46 (36–57); 44.2% (29.1–60.1) in untreated patients (n = 43) and 48.2 (34.7–62.0) in previously treated patients (n = 56)42.9 (31.1–53.3); 46.4 (27.5–66.1) in untreated patients (n = 28) and 40.5 (25.6–56.7) in previously treated patients (n = 42)
Duration of response mo (95% CI)12.6 (5.6—NE) in untreated patients and 9.7 (5.6–13.0) in previously treated patients 11.1 (7.2—NE)8.3 (5.3–16.6); 5.6 (4.1–9.6) in untreated patients and 9.7 (4.9—NE) In previously treated patients
Progression-free survival mo (95% CI)12.4 (8.2—NE) in untreated patients and 5.4 (4.2–7.0) in previously treated patients 8.5 (5.1–11.0)6.8 (4.2–9.6); 5.6 (4.1–9.6) in untreated patients and 6.9 (4.1–9.3) in previously treated patients
Table 4. Predictive biomarkers and methods for FDA-approved, MET-targeted drugs in NSCLC [59].
Table 4. Predictive biomarkers and methods for FDA-approved, MET-targeted drugs in NSCLC [59].
PublicationDrugMethodBiomarkerNORR%
Moro-Sibilot et al. [39]CrizotinibFISHMET GCN ≥ 62516
NGSMET exon 14 skip2512
Landi et al. [40]CrizotinibFISHMET/CEP7 > 2.21631
NGSMET exon 14 skip1020
Drilon et al. [41]CrizotinibNGSMET exon 14 skip6532
Schuler et al. [50]CapmatinibFISHMET GCN < 4176
MET GCN 4–61225
MET GCN ≥ 61547
MET/CEP7 > 2.0944
MET/CEP7 < 2.03222
IHCMET IHC 2+1414
MET IHC 3_3727
Wu et al. [51]Capmatinib with gefitinibFISHMET GCN < 44112
MET GCN 4–61822
MET GCN ≥ 63647
IHCMET IHC 2+1619
MET IHC 3+_3727
Wolf et al. [6]CapmatinibNGSMET exon 14 skip (Previously treated)6941
MET exon 14 skip (Untreated)2864
NGSMET GCN < 4 (Previously treated)307
MET GCN 4–5 (Previously treated)549
MET GCN > 6–9 (Previously treated)4212
MET GCN ≥ 10 (Previously treated)6928
MET GCN ≥ 10 (Untreated)1540
Paik et al. [31]TepotinibNGSMET exon 14 skip9946
Table 5. Adverse events in all cohorts (n = 364) in the GEOMETRY mono-1 trial [6].
Table 5. Adverse events in all cohorts (n = 364) in the GEOMETRY mono-1 trial [6].
Adverse EventTotalGrade 3 or 4
Any event—No. (%)355 (98)244 (67)
Most common events—No. (%)
Peripheral edema186 (51)33 (9)
Nausea163 (45)9 (2)
Vomiting 102 (28)9 (2)
Blood creatinine increased89 (24)0
Dyspnea 84 (23)24 (7)
Fatigue80 (22)16 (4)
Decreased appetite76 (21)3 (1)
Constipation66 (18)3 (1)
Diarrhea64 (18)2 (1)
Cough58 (16)2 (1)
Back Pain54 (15)3 (1)
Pyrexia50 (14)3 (1)
ALT increased48 (13)23 (6)
Asthenia42 (12)13 (4)
Pneumonia39 (11)17 (5)
Weight loss36 (10)2 (1)
Noncardiac chest pain35 (10)4 (1)
Serious adverse event—No. (%)184 (51)152 (42)
Event leading to discontinuation—No. (%)56 (15)35 (10)
Table 6. Current key ongoing studies involving capmatinib.
Table 6. Current key ongoing studies involving capmatinib.
Clinical Trial Number PhasePurpose
NCT04427072Phase IIIPreviously treated advanced NSCLC patients with MET exon 14 skipping mutation treated with capmatinib versus docetaxel
NCT04926831Phase IIEfficacy and safety of neoadjuvant and adjuvant capmatinib
NCT05435846Phase I/IbCapmatinib plus trametinib in patients with MET exon 14 skipping mutation
NCT04677595Phase IIChinese patients who are EGFR wt and ALK rearrangement negative with MET exon 14 skipping mutation
NCT05110196Phase IVIndian patients with MET exon 14 skipping mutation
NCT05488314Phase I/IICombination therapy of capmatinib and amivantamab in unresectable Stage IV NSCLC in patients with MET exon 14 skipping mutations or MET amplification
NCT05642572Phase IICombination therapy of capmatinib with osimertinib +/− ramucirumab in EGFR mutant, MET-amplified, Stage IV or recurrent NSCLC
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hsu, R.; Benjamin, D.J.; Nagasaka, M. The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer—A Narrative Review. Cancers 2023, 15, 3561. https://doi.org/10.3390/cancers15143561

AMA Style

Hsu R, Benjamin DJ, Nagasaka M. The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer—A Narrative Review. Cancers. 2023; 15(14):3561. https://doi.org/10.3390/cancers15143561

Chicago/Turabian Style

Hsu, Robert, David J. Benjamin, and Misako Nagasaka. 2023. "The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer—A Narrative Review" Cancers 15, no. 14: 3561. https://doi.org/10.3390/cancers15143561

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop