Next Article in Journal
A New Nrf2 Inhibitor Enhances Chemotherapeutic Effects in Glioblastoma Cells Carrying p53 Mutations
Next Article in Special Issue
Longitudinal Plasma Proteomics-Derived Biomarkers Predict Response to MET Inhibitors for MET-Dysregulated NSCLC
Previous Article in Journal
Genetically Modified Circulating Levels of Advanced Glycation End-Products and Their Soluble Receptor (AGEs-RAGE Axis) with Risk and Mortality of Breast Cancer
Previous Article in Special Issue
Molecular Characteristics of Radon Associated Lung Cancer Highlights MET Alterations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Landscape of Savolitinib Development for the Treatment of Non-Small Cell Lung Cancer with MET Alteration—A Narrative Review

1
Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
2
AstraZeneca China, Shanghai 201200, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2022, 14(24), 6122; https://doi.org/10.3390/cancers14246122
Submission received: 8 November 2022 / Revised: 6 December 2022 / Accepted: 7 December 2022 / Published: 12 December 2022
(This article belongs to the Special Issue Roles of MET in Cancer Development and Treatment)

Abstract

:

Simple Summary

In this article, we outline updates on the clinical development of savolitinib, a novel, reversible c-MET kinase inhibitor conditionally approved in China for treatment of advanced non-small cell lung cancer (NSCLC) patients harboring MET exon 14 skipping mutation (METex14). Savolitinib was developed as a monotherapy for NSCLC with MET alterations, and in combination with epidermal growth factor receptor (EGFR) inhibitors for patients who developed resistance to EGFR–TKIs because of MET alterations. Savolitinib showed anti-tumor activity in preclinical models. The early phase I trial established the recommended phase II dose to be 600 mg once-daily. Savolitinib plus osimertinib showed beneficial efficacy and safety in EGFR mutant patients with acquired resistance due to MET amplification and/or c-MET overexpression. Benefits were noted with savolitinib in Chinese patients with pulmonary sarcomatoid carcinoma and other NSCLC subtypes positive for METex14 mutation. Results from phase III trials are awaited to further confirm the beneficial effects from early phase trials.

Abstract

Non-small cell lung cancer (NSCLC) is increasingly being treated with targeted therapies. Savolitinib (Orpathys®) is highly selective mesenchymal epithelial transition (MET)–tyrosine kinase inhibitor (TKI), which is conditionally approved in China for advanced NSCLC with MET exon 14 skipping mutations (METex14). This article summarizes the clinical development of savolitinib, as a monotherapy in NSCLC with METex14 mutation and in combination with epidermal growth factor receptor (EGFR) inhibitor in post EGFR–TKI resistance NSCLC due to MET-based acquired resistance. Preclinical models demonstrated anti-tumor activities in MET-driven cancer cell line and xenograft tumor models. The Phase Ia/Ib study established an optimized, recommended phase II dose in Chinese NSCLC patients, while TATTON study of savolitinib plus osimertinib in patients with EGFR mutant, MET-amplified and TKI-progressed NSCLC showed beneficial efficacy with acceptable safety profile. In a pivotal phase II study, Chinese patients with pulmonary sarcomatoid carcinoma, brain metastasis and other NSCLC subtype positive for METex14 mutation showed notable responses and acceptable safety profile with savolitinib. Currently, results from ongoing clinical trials are eagerly anticipated to confirm the efficacious and safety benefits of savolitinib as monotherapy and in combination with EGFR–TKI in acquired resistance setting in advanced NSCLC and its subtypes with MET alterations.

1. Introduction

Non-small cell lung cancer (NSCLC) accounts for approximately 80% of all lung cancers with a low 5-year survival rate of about 22% [1,2]. Most NSCLC are usually diagnosed at an advanced stage with traditional chemotherapy and radiotherapy showing limited efficacy. However, recent advances in immune therapy and targeted therapy have radically improved the treatment paradigm of NSCLC over the past decade [2]. Molecular profiling of lung cancer samples for activated oncogenes, including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK) and c–ros oncogene 1 (ROS1), is considered as standard-of care to select the most appropriate up-front treatment [3]. However, the identification of new therapeutic targets remains a high priority. Recently, mesenchymal-epithelial transition (MET) exon 14 skipping mutations (METex14) and high-level MET amplification have emerged as one of the novel, actionable oncogenic alterations in NSCLC, sensitive to MET inhibitors [4,5].
MET is a receptor tyrosine kinase activated by binding ligand hepatocyte growth factor (HGF) which plays a key physiological role in the interaction between mesenchyme and epithelia during embryonic wound closure and embryogenesis [6,7,8,9]. At cellular levels, MET-TK activity transduces mitogenesis by activating Ras–Raf–MAPK signaling pathway and motogenic signals by activating phosphoinositide 3–kinase (PI–3K) pathway upon HGF binding [10]. Aberrant MET/HGF signaling promotes mitogenesis, invasion and angiogenesis, thus contributing towards tumorigenesis and progression of cancer [11]. Importantly, significant implications for tumorigenesis are observed due to crosstalk between downstream signal pathways of MET and EGFR [12]. The oncogenic role of MET was first discovered in 1984 as a part of an oncogenic fusion with the translocated promoter region gene in a mutagenized osteosarcoma cell line [13]. MET alterations, including amplification, mutations, gene fusion, MET/HGF protein over expression and the crosstalk between dysregulated MET and other signaling pathways, are associated with poor prognosis in cancers, and thus, molecularly targeted [4]. METex14 mutations are the most commonly reported oncogenic mutations. Exon 14 encodes the 47-amino acid juxtamembrane domain of the MET receptor, a key regulatory region that prevents MET over signaling. METex14 mutations include a heterogeneous group of mutations with base substitutions or indels that disrupt the branch point of intron 13, the 3′ splice site of intron 13 or the 5′ splice site of intron 14, producing a MET variant that lacks the exon 14 leading to disruption of cellular signaling [14]. The identification of MET oncogene and the journey leading to development of MET–TKIs is represented in Figure 1.
Clinical studies conducted earlier suggest that activation of MET can act as primary oncogenic driver, or secondary driver of acquired resistance to targeted therapy in subsets of lung cancer [9,10,11]. METex14 mutations occur in approximately 0.9 to 4% of NSCLC cases across all histologic subtypes [6] and are enriched in pulmonary sarcomatoid carcinoma (PSC) (20 to 31%), a rare subtype of poorly differentiated NSCLC [15,16]. Furthermore, 1 to 5% of NSCLC harbors de novo MET gene amplification, while 15% of cases in METex14-mutated NSCLC report MET amplification [17,18]. MET fusion is known to occur in 0.5% [18] and MET protein overexpression in 13.7 to 63.7% of NSCLC patients [17]. Significant cross talk between aberrant MET pathway and other signaling pathways, especially EGFR results in acquired resistance to EGFR tyrosine kinase inhibitors (TKIs) in patients with NSCLC [19]. Mechanistically, MET amplification causes EGFR–TKI resistance by activating EGFR-independent phosphorylation of ErbB3 and downstream activation of the PI3K/AKT pathway, providing a bypass pathway in the presence of an EGFR inhibitor [20]. Thus, concomitant inhibition of both EGFR and MET would be required to overcome resistance to EGFR inhibitors by MET amplification [19]. Approximately, 5–22% of NSCLC patients with first- or second-generation EGFR–TKI resistance [18,21] and 5–50% patients with third generation EGFR–TKI resistance harbor MET amplification [22], while MET amplification as a co-driver occurs in 2–11% EGFR-positive treatment-naïve NSCLC patients [23,24]. The incidence of high MET expression after EGFR–TKI resistance is as high as 30.4 to 37% [25]. The proportion of different MET alterations in NSCLC patients is summarized in Table 1.
Currently the FDA approved MET–TKIs are capmatinib and tepotinib with crizotinib granted as breakthrough therapy designation, while savolitinib is conditionally approved in China [26,27,28]. Further, for EGFR-mutated NSCLC with MET amplification treatment, efficacy of combination of MET–TKIs with EGFR–TKIs has been preliminarily approved by several clinical trials [29,30]. The clinical development strategy for savolitinib is centered both as monotherapy for advanced METex14-altered NSCLC and in combination with EGFR–TKI for correction of MET-driven acquired resistance to EGFR–TKIs [31]. In this review, we briefly describe the major milestones achieved in the clinical development of savolitinib as standard of care for NSCLC with METex14 mutation and potential treatment for NSCLC with other MET alterations.

2. Savolitinib, in Brief

Savolitinib (Orpathys®) is an orally bioavailable and highly selective small molecule MET–TKI that has demonstrated profound efficacy in preclinical and clinical studies of various cancers, including NSCLC, papillary renal cell carcinoma (PRCC) and gastric carcinoma [32,33,34]. Figure 2 demonstrates the chemical structure of savolitinib.
Early on, in vitro studies have established inhibitory effect of savolitinib on growth of gastric cells lines, while in vivo studies observed anti-tumor activity in human xenograft tumor models of MET-amplified gastric cancer and PRCC [33,34,35]. Another study by Jones and colleagues related to pharmacokinetic-pharmacodynamic (PK-PD) model observed inhibition of phosphorylated-MET by savolitinib at an effective concentration (EC)50 of 0.35 ng/mL and EC90 of 3.2 ng/mL in a cell line-derived xenograft (CDX) mice model using human lung cancer (EBC-1) and gastric cancer (MKN-45) cells [36]. Furthermore, PK studies in healthy male Chinese volunteers administered with single oral savolitinib doses of 200, 400 and 600 mg following an overnight fast or a high-fat and high-calorie breakfast prior to dosing showed no clinically relevant impact on PK and bioavailability of savolitinib [37].
In NSCLC with MET aberrations, several clinical trials have shown the potential benefit of savolitinib as a monotherapy and in combination with EGFR–TKI [30]. Savolitinib received its first approval by The National Medical Products Administration (NMPA), China for patients with METex14-altered locally advanced or metastatic NSCLC with disease-progression following systemic treatment or unable to receive chemotherapy [28]. The approval was based on a phase II trial conducted in China in patients with METex14-altered NSCLC, including patients with the more aggressive PSC subtype [38]. The key milestones in the development of savolitinib for NSCLC treatment are demonstrated in Figure 3.

3. First Steps towards the Development of Savolitinib as Mono and Combination Therapies

The availability of substantial evidence of anti-tumor activity and acceptable safety profile led to the development of savolitinib as a treatment for advanced NSCLC with MET aberrations. The high selectivity of savolitinib for MET was confirmed using a screening platform of more than 900 cell lines of which 111 represented NSCLC [39]. In vitro study by Henry and colleagues demonstrated the ability of savolitinib as a single agent to inhibit MET activity and reduce NSCLC cell viability in a dose dependent manner [39]. Further, anti-tumor efficacy was observed with savolitinib in vivo, in lung cancer PDX model with METex14 mutation. Savolitinib showed tumor regression (tumor volume reduction: 62%) with a dose of 25 mg/kg in all mice on treatment (n = 9) as well as 98% inhibition in tumor growth (TGI) with 5 mg/kg dose in 4 out of 9 mice of PDX model (Data on file). In addition, in vivo study using H1993 and EBC-1 tumor xenografts showed considerable decrease in tumor growth, with savolitinib achieving an optimal response at doses as low as 0.3 mg/kg and 2.5 mg/kg in H1993 and EBC-1 tumors, respectively [39]. Interestingly, the same group (Henry and colleagues) concurred that savolitinib resistance in NSCLC is partially driven by MYC overexpression in H1993 cells, suggesting potential mechanism and treatment strategies for future acquired resistance to MEK–TKI.
Savolitinib, in combination with erlotinib, a first-generation EGFR–TKI inhibitor, showed substantial tumor inhibition in H441, an EGFR wild type model with MET amplification [40,41]. In addition, savolitinib treatment exhibited substantial anti-tumor activity in vivo (tumor regression: 35%) in the NSCLC cancer cell line NCI-H820 harboring an activating EGFR mutation (Ex19del), a gefitinib/erlotinib resistant mutation (T790M) as well as hyperactivated MET (data on file). Osimertinib, a third-generation, irreversible EGFR–TKI, at either 25 mg/kg daily or 12.5 mg/kg daily exhibited minimal anti-tumor activity, with TGI of 24% and 4%, respectively. However, when treated in combination with savolitinib, 25 mg/kg of savolitinib plus osimertinib at either 25 mg/kg or 12.5 mg/kg daily resulted in 94% and 90% TGI, respectively. These preclinical results highlight the beneficial anti-tumor effect of osimertinib plus savolitinib combination at optimal doses of 0.3~1.5 mg/kg savolitinib combined with 10 mg/kg osimertinib. Further, another study analyzed different doses of savolitinib, ranging from 0.02 mg/kg to 15 mg/kg (15 mg/kg equivalent to 600 mg clinical dose), in combination with a fixed dose of 10 mg/kg osimertinib (equivalent to 80 mg clinical dose). Pan–CYP inhibitor 1–aminobenzotriazole was dosed along with savolitinib and osimertinib to prolong PK half-life by reducing elimination rate so that plasma concentration time profile matches clinical exposure of the drugs (data on file). The combination of osimertinib and savolitinib demonstrated strong anti-tumor activity leading to tumor regressions. The benefit of combination treatment was observed with as low as 0.3 mg/kg dose of savolitinib. Thus, these encouraging preclinical results led to the evaluation of savolitinib’s efficacy and safety in clinical trials for NSCLC with MET aberrations.

4. Clinical Development of Savolitinib: Phase I Trials

A first in-human phase I clinical study (NCT01773018) was conducted in patients (n = 48) with locally advanced solid tumors from Australia [42]. The doses administered were 100–1000 mg once daily (OD) and 300–500 mg twice-daily (BID), and the maximum tolerated dose was 800 mg. Savolitinib showed preliminary efficacy in patients with papillary renal cell carcinoma with MET gene copy number changes. The most frequent adverse events (AE) were nausea (62.5%), vomiting (41.7%), fatigue (35.4%) and peripheral edema (27.1%). The tolerability profile of savolitinib was acceptable, and the recommended phase II dose (RP2D) was established as 600 mg OD [42]. In another open-label, multi-center, phase Ia/Ib study (NCT0198555) conducted in China in patients (n = 85) with advanced tumors bearing MET aberrations, savolitinib demonstrated a manageable safety profile and promising anti-tumor activity in NSCLC with METex14 mutation, apparent tumor shrinkage (55% and 27%) in target lesions was observed, although partial response (PR) was not achieved. The most common treatment-related AEs were nausea (29.4%), vomiting (27.1%) and peripheral edema (21.2%). The RP2D of savolitinib was established at 600 mg OD or 500 mg BID and was consistent with phase I first-in human study conducted in Australia [43]. There was certain comparability between the patients with NSCLC enrolled in the phase I study conducted in Australia and the phase I study conducted in China, and thus the results could be analyzed accordingly [42,43].
Savolitinib demonstrated the ability to overcome MET-mediated resistance in patients with EGFR-mutant, MET-amplified or c-MET overexpressed NSCLC when combined with osimertinib, and these benefits extended to those with disease that had previously progressed on a prior EGFR–TKI [29,44]. Part A of the multi-arm phase Ib TATTON study (NCT02143466) demonstrated the safety and tolerability of osimertinib plus savolitinib (n = 18) in patients with advanced NSCLC disease progression on a prior EGFR–TKI [44]. Doses of savolitinib applied were escalated from 600 to 800 mg OD with a fixed dose of osimertinib 80 mg. The most common AEs reported were nausea (67%), rash (56%) and vomiting (50%). The objective response rate (ORR) was 44% [44]. Furthermore, in the expansion cohorts of TATTON trial, investigators evaluated the safety and efficacy of osimertinib plus savolitinib in locally advanced or metastatic, MET-amplified, EGFR mutation-positive NSCLC patients who had progressed on EGFR–TKIs [29]. Part B (n = 138) was substratified into three cohorts: B1 included those who had previously received a third-generation EGFR-TKI; patients without prior third-generation EGFR–TKI treatment were separated into B2 with Thr790Met negative and B3 with Thr790Met positive, at the time of enrolment. These patients received 600 mg QD, although the protocol was later amended, causing patients who weighed lesser than 55 kg to receive a 300-mg dose of savolitinib. The Part D expansion cohort was comprised of patients (n = 42) who had not previously received a third-generation EGFR–TKI and were T790M negative, and these patients received osimertinib 80 mg plus savolitinib 300 mg OD. Objective partial responses (PR) were observed (by 4 March 2020) in 68 (49%) patients in total of part B, with 23 (33%) patients, 33 (65%) patients and 12 (67%) patients in B1, B2 and B3, respectively, while in 26 (62%) patients in part D [45]. Regarding safety, the 4 expanded cohorts had similar safety profiles with 28% in part B and 19% in part D experiencing AEs possibly related to savolitinib. Serious AEs of grade 3 or 4 were associated with 49% patients in part B and 38% patients in part D. The most common AEs of grade 1–2 in expanded cohorts included nausea (48%), peripheral edema (34%), decreased appetite (32%), vomiting (30%) and fatigue (28%). In part B cohorts, the most common grade 3 or higher AEs related to savolitinib were decreased neutrophil count (6%) and aminotransferase elevations (4%), while in part D, hypersensitivity (5%), diarrhea (5%) and myalgia (5%) are more frequent [45]. Generally, in the dose expansion cohorts of TATTON trial, savolitinib plus osimertinib showed promising anti-tumor activity in MET-amplified EGFR positive advanced NSCLC patients who received a prior third-generation EGFR–TKI. These results have now been further investigated in the phase II SAVANNAH trial.
In another phase Ib study (NCT02374645), the clinical evaluation of savolitinib plus gefitinib (a first-generation EGFR-TKI) demonstrated promising anti-tumor activity with acceptable safety profile in EGFRm, MET-amplified advanced NSCLC patients from China who had disease progression on EGFR-TKIs. Patients received savolitinib 600 or 800 mg plus gefitinib 250 mg orally OD for which no dose-limiting toxicities were reported in safety run-in. The most commonly reported AEs were vomiting (46%), nausea (40%) and increased aspartate aminotransferase (39%) [30]. ORR in EGFR T790M-negative and -positive patients were 52% and 9%, respectively, suggesting beneficial anti-tumor activity [30].

5. Clinical Development of Savolitinib: Phase II Trials

A pivotal open-label phase II clinical study (NCT02897479) conducted in China demonstrated encouraging efficacy and tolerable safety profile of savolitinib in overall and patient subsets stratified according to tumor type (PSC and other NSCLC), brain metastasis status and prior anti-tumor treatment (pretreated and treatment naïve) [38]. Unresectable or metastatic NSCLC patients (n = 70) harboring METex14 mutation were administered savolitinib monotherapy at recommended starting dose of 600 mg orally once daily (OD) for patients weighing ≥50 kg, or 400 mg OD for patients weighing <50 kg, until disease progression or unacceptable toxicity. The majority of patients were elderly with advanced NSCLC on prior systemic therapy. In both the full analysis set (FAS) and the tumor response evaluable set (TRES), independent review committee (IRC) assessments were the main analyses, while investigators’ (INV) assessments were supportive analyses. The IRC-assessed tumor response evaluable set (TRES) was comprised of 62 patients. The ECOG performance status of full analysis set (FAS) for majority of patients (81%) was 1 and in pre-specified subsets (PSC vs. other NSCLC subtypes, treatment naïve vs. previously treated), 78% to 88% patients had ECOG status of 1. Of the total PSC population (n = 25), pre-treated and treatment-naïve subsets were comprised of 29% and 46%, while brain metastasis and non-brain metastasis groups were comprised of 13% and 42% of PSC patients, respectively [38,46,47]. The primary efficacy end point was ORR (as assessed by IRC in TRES) defined as the proportion of patients with a confirmed complete response or partial response according to RECIST version 1.1. Secondary outcomes included duration of response (DoR), time to response (TTR), progression free survival (PFS), overall survival (OS) and safety. The latest results of the trial were presented at the 2022 ELCC conference and published in JTO Clinical and Research Reports [46,48]. The baseline characteristics are provided in Table 2.

5.1. Efficacy Evidence

At a median follow-up of 17.6 months, the IRC and INV assessed ORR was 49.2% and 53.2 %, respectively in TRES subset, while ORR assessed in FAS set by IRC and INV was 42.9% and 47.1%, respectively. Further, the IRC and INV assessed disease control rate (DCR) was 93.4% and 91.9%, respectively in TRES subset, while DCR assessed in FAS set by IRC and INV was 82.9% and 81.4%, respectively. The median time to response was 1.4 months across TRES and FAS sets as judged by IRC and INV. Median DoR for TRES and FAS as assessed by IRC and INV was 8.3 and 6.9 months. Savolitinib was associated with mOS of 12.5 months and a mPFS of 6.9 months in FAS at a median follow-up time of 28.4 months. The 18-month OS rate is 42.1%, dropping to 31.5% at 24 months [38,48].
In subgroup analyses (assessed in TRES set by INV, median follow-up of 28.4 months), for PSC (n = 20), 10 patients had partial response (ORR 50%) with a median duration of response of 12.4 months. In other NSCLC subtypes (n = 42), 23 patients had partial response (ORR 54.8%) with a median duration of response of 5.6 months and DCR of 92.9%. In pre-treated (n = 38) patients, partial response was observed in 20 patients (52.6%), while in treatment-naïve (n = 24) subgroup, partial response was observed in 13 patients (54.2%). Extracranial ORR for brain metastasis group was 64.3%. For survival outcomes, the PSC group showed a mPFS of 5.5 months, while with brain metastasis (n = 15), it was 7.0 months and without brain metastasis was 6.2 months. Similar values of mPFS were observed with pre-treated (6.9 months) and treatment-naïve (6.9 months) subgroups, respectively. The mOS for PSC and other NSCLC patients was 10.6 months and 17.3 months, respectively, with corresponding 24-month OS rates of 26% and 35%. Among brain metastases patients, the mOS was 17.7 months with the 24-month OS rate being 36%. The mOS for pre-treated and treatment-naïve patients was 19.4 months and 10.9 months, respectively, with corresponding 24-month OS rates of 38% and 22% [48]. However, this large difference in OS can be attributed to the higher proportion of patients with PSC in treatment-naïve population (46% vs. 29% in pre-treated patients) and a higher median age (74.5 vs. 67.7 in pre-treated patients). Patients with PSC had a short mOS vs other NSCLC patient (10.6 months vs. 17.3 months), likely due to the poor prognosis associated with PSC. These results confirmed savolitinib having beneficial efficacy towards NSCLC with METex14 mutation and its PSC subtype [32,38,48]. The PFS and OS results have been illustrated graphically in Table 3.
An earlier study reported mOS of 6.7 months in patients with METex14 mutation NSCLC on chemotherapy treatment who did not receive prior targeted therapy [49]. In addition, mOS of PSC subset in NSCLC patients treated with chemotherapy has been reported to be 4 to 8 months [32,49,50,51,52,53]. With savolitinib, mOS of NSCLC patients reaches 12.5 months with 70% maturity. In PSC subset, higher OS is seen with savolitinib treatment compared to chemotherapy, with OS reaching 10.6 months. So far, literature related to MET inhibitor treatment with PSC population is available only for savolitinib [51,52,53,54].
P–glycoprotein (gp) and breast cancer resistance protein (BRCP) are efflux proteins located in the luminal membrane of brain capillary endothelium, preventing drugs from entering the central nervous system. Most MET inhibitors, such as crizotinib and tepotinib, are known substrates of the P–gp and BRCP efflux transport system [55,56,57]. Steady concentrations of savolitnib are readily maintained in an intracerebral area which may be attributed to it not being a substrate of P–gp and BRCP efflux transport system. Promising efficacy of savolitinib was observed in brain metastasis subgroup, with ORR at 64.3%, DCR at 100% and significant survival benefit (PFS, 7.0 months; OS, 17.7 months). These encouraging results provide a treatment option for this subgroup of patients with poor prognosis and few treatment options [38,47,48].

5.2. Safety Evidence

Savolitinib demonstrated tolerable safety profile consistent with previous trials; most AEs were grades 1–2 and resolved with dose adjustment and discontinuation. Adverse events that presented at rates of ≥30% are listed below (Table 4, median follow-up of 28.4 months). The incidence of grade3 or more AEs was 65.7%, while 50% of patients reported treatment-related serious adverse events (SAE). The top ≥ grade 3 AE was elevated AST (12.9%). The most common treatment-related AEs (TRAEs) (≥30%) are peripheral edema (55.7%), nausea (45.7%) and elevated aminotransferase (38.6% and 37.1%). The top ≥ grade 3 treatment related AE was elevated AST (12.9%) [48,58]. The common SAEs reported were abnormal liver function (4.3%, 3 patients), drug hypersensitivity reaction (2.9%, 2 patients) and fever (2.9%, 2 patients). Treatment related fatal SAE, tumor lysis syndrome was reported in one patient. Ten patients discontinued treatment due to AEs, of which drug-induced liver damage and drug hypersensitivity reactions were seen in 2.9% of patients (2 patients), respectively [38]. No occurrence of pulmonary interstitial pneumonia and interstitial lung disease (ILD) was observed with savolitinib, while ILD is seen with tepotinib (n = 2) and capmatinib (n = 1) [4,59].
The updated results further confirm that savolitinib can benefit METex14-mutated NSCLC patients and each subgroup with acceptable safety profile [38,46,47,48]. Savolitinib thus displays promising efficacy and tolerability in PSC associated with METex14 mutation and holds potential to become the first approved treatment in this setting. In addition, the study showed that savolitinib can penetrate the blood–brain barrier and is effective in patients with brain metastases.

5.3. Brief Introduction of Other Phase II Trials

Other ongoing phase II trials include SAVANNAH, SOUND and FLOWERS trials. SAVANNAH trial (NCT03778229) continues to explore the sequence of savolitinib plus osimertinib with previous osimertinib monotherapy resistance. It is a phase II, single-arm study evaluating the efficacy of osimertinib in combination with savolitinib in 259 patients with EGFRm and MET amplified and/or c-MET overexpressed locally advanced or metastatic NSCLC who have progressed on osimertinib. Patients were treated with osimertinib (80 mg OD) and savolitinib (300 mg QD, 300 mg BID or 600 mg OD) until objective disease progression. Efficacy endpoints—such as ORR (primary endpoint), PFS, OS, DoR, HRQoL, pharmacokinetics, safety points such as AEs and patient related outcomes (PROs)—were studied. This is the first phase II clinical study of the third-generation EGFR–TKI osimertinib resistance in patients with advanced NSCLC with MET amplification and/or c-MET overexpression. MET detection was performed using fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) methods. The detection criteria were set to FISH, MET GCN ≥ 5 and/or MET/CEP7 ≥ 2; IHC, ≥50% tumor cells 3+. Sixty-two percent of osimertinib resistant patients was at low threshold [IHC50+ and/or FISH5+] as well as 34%—at the high threshold [IHC90+ and/or FISH10+] subgroups. Figure S1 provides the proportion of patient population with MET amplified and/or c-MET overexpressed in this study suggesting amplification and/or overexpression is the most common osimertinib resistance mechanism. The baseline characteristics are provided in Table S1. The overall median age of patients is 63 years, 62% were female, 54% were Asian and 34% were with brain metastases at baseline. On savolitinib 300 mg OD plus osimertinib 80 mg OD treatment, advanced NSCLC patients (n = 193) with high MET amplification and/or high threshold c-MET overexpression level show a trend toward better efficacy benefit, emphasizing the necessity of patients’ selection according to appropriate MET detection criteria in this population. Among the overall population, ORR was 32%; median DoR was 8.3 months; and median PFS was 5.3 months, while among 108 patients who met the threshold for high MET amplification and/or high threshold c-MET overexpression level (IHC90+ and/or FISH10+), ORR was 49%; median DoR was 9.3 months; and median PFS was 7.1 months (Table S2). The safety results showed that the incidence of treatment-related AEs was 84%; treatment-related ≥grade 3 AEs at 20%; and treatment-related SAEs at 7% (Table S3). The incidence of hypersensitivity, ILD and pneumonia were 2% (4/196), and QT interval prolongation at 5% (10/196) [60].
In addition, the FLOWERS trial (NCT05163249) explores the efficacy and safety of osimertinib with or without savolitinib in patients with de novo MET amplified and/or c-MET overexpressed, EGFR-mutant advanced NSCLC. In SOUND trial (NCT05374603), an open-label, interventional, multi-center, exploratory trial, savolitinib combined with durvalumab will be evaluated in Chinese EGFR wild-type locally advanced or metastatic NSCLC patients with MET alterations. NSCLC patients from China with MET amplification (n = 30) and METex14 mutation (n = 30) will be treated with 1500 mg durvalumab and 300 to 600 mg savolitinib (OD) for 28-day/cycle till disease progression, death or toxicity. Efficacy endpoints will be PFS, ORR, DoR, DCR, 12 m OS rate and safety endpoints will be AEs and AEs of special interest (AESI) [61]. Further, phase III SAFFRON trial (NCT05261399) is investigating savolitinib plus osimertinib versus platinum-based doublet chemotherapy in participants with NSCLC (EGFR mutated, c-MET overexpressed and/or MET gene amplified) who have progressed on osimertinib treatment.

6. Ongoing Phase III Trials

Currently, four phase III trials evaluating savolitinib as a monotherapy and in combination with EGFR-TKIs are underway. The confirmatory phase IIIb clinical study (CTR20211151) is evaluating efficacy and safety of savolitinib in two cohorts from patients with locally advanced or metastatic NSCLC with METex14 mutation in China; patients of one cohort are with disease progression or toxicity intolerance after previous platinum-based chemotherapy regimens, and patients of another cohort are with no prior systemic antineoplastic therapy for advanced disease. The patients were treated until disease progression or intolerable toxicity. Phase III SACHI trial (CTR20211441) is a randomized, two-arm, open-label, multi-center study evaluating the efficacy and safety of savolitinib plus osimertinib versus chemotherapy in NSCLC patients from China with MET amplification who has progressed after first- to third-generation EGFR–TKI therapy and has already begun its recruitment in multiple centers. Another similar phase III trial SAFFRON is designed to evaluate the efficacy and safety of the same combined therapy as SACHI versus chemotherapy, but focus on global advanced NSCLC patients with MET amplification/c-MET overexpression that progressed after osimertinib treatment. SANOVO Phase III study is evaluating the efficacy and safety of savolitinib in combination with osimertinib in treatment-naïve patients with EGFR mutant positive and c-MET overexpression advanced NSCLC (NCT05009836).

7. Discussion

Savolitinib, an investigational MET highly selective agent, has shown pronounced efficacy in preclinical and clinical studies. Savolitinib demonstrated preclinical anti-tumor activity against MET-dependent cancer cell line growth and MET-driven tumor growth in xenograft models. Following which, data from a phase I clinical trial established recommended phase II dose in patients with METex14-mutated NSCLC. Further, the TATTON study established utility of savolitinib with osimertinib in advanced NSCLC with MET-mediated acquired resistance to EGFR-TKIs. Final results of the phase II study (NCT02897479) further confirmed the benefit of savolitinib in patients with METex14-mutated NSCLC across all predefined subgroups. In addition, phase IIIb clinical study CTR20211151 is confirming the result of phase II study on METex14-mutated NSCLC, while three ongoing phase II trials, SAVANNAH, SOUND and FLOWERS, as well as three phase IIIB trials, SAFFRON, SACHI and SANOVO, are actively exploring solutions for different types of savolitinib combination regimens against EGFR resistance mechanisms. Preliminary results of the SAVANNAH trial have demonstrated the beneficial efficacy of osimertinib plus savolitinib in EGFRm NSCLC patients with MET amplified and/or c-MET overexpressed, supporting the results of TATTON study and paving the way for phase III SACHI and SAFFRON study.
In the hallmark phase II registry trial, savolitinib displayed promising efficacy and tolerability in patients with METex14-altered advanced NSCLC, with mOS reaching 12.5 months. The effect of savolitinib was rapid, substantiated by time to response (TTR) of 1.4 months. Promising results with PFS of 5.5 months and OS of 10.6 months were also seen in the PSC subtype, which does not respond well to chemotherapy and has limited effective treatments. By now, savolitinib is the only MET inhibitor with data related to PSC associated with METex14 mutation and is becoming the first approved agent in this setting. For patients of treatment naïve population, the PFS and OS of savolitinib were 6.9 months and 10.9 months, respectively, while PFS and OS of prior treatment patients reached 6.9 months and 19.4 months, respectively. In the current scenario, the reported ORR of savolitinib is the highest in the prior treatment population compared to other treatments (52.6% vs. 44.0% of capmatinib, 49.5% of tepotinib and 21% of amivantamab) [48,62,63,64]. Savolitinib is also currently the only MET inhibitor that has recorded beneficial OS data in brain metastases, with PFS of 7.0 months and OS of 17.7 months. In addition, savolitinib has the best tumor response in brain metastasis population with ORR at 64.3% and DCR at 100% [38,48]. Based on these promising results, savolitinib received its first conditional approval by NMPA, China in June 2021, for patients with METex14-altered NSCLC after systemic treatment resistance or unable to receive chemotherapy. Post-marketing phase IIIb trial is now undergoing (HutchMed) in larger population of NSCLC patients and is expected to provide more clinical evidences for savolitinib in first-line therapy. Furthermore, latest post hoc analysis based on ctDNA detection suggests undetectable baseline METex14 or post-treatment clearance in ctDNA being relevant to favourable clinical outcomes, including better PFS and OS results, while secondary MET mutations and other acquired gene alterations after treatment (e.g., RTK–RASP–I3K pathway) may explain resistance mechanism to savolitinib [65].
Table 5 summarizes the data for MET-TKIs developed for METex14-altered advanced NSCLC population as well as subtypes [4,38,47,48,58,59,62,63,64,66,67,68,69,70,71,72,73]. Patient population of Chinese Phase II registry trial were from China. In other global trials, east Asian population varied from 15.9 to 50.9%. Proportion of NSCLC patients with brain metastases was higher (28.9%) in Chinese Phase II registry trial compared to other trials [48]. Tumor response of different types of MET–TKIs shows ORR (54.8%) and DCR (92.9%) to be highest with savolitinib. Among AEs, most commonly, elevated transaminases were seen with savolitinib, tepotinib and crizotinib; peripheral edema with savolitnib, capmatinib and tepotinib; ILD with capmitnib, tepotinib and crizotinib; difficulty in breathing in tepotinib, crizotinib and amivantamab [38,48,58,59,62,63,64,67,68,69,70,71,72,73].
Bypass activation mediated by the MET signaling pathway is one of the important mechanisms leading to EGFR–TKI resistance. MET-driven resistance can be manifested as gene-level amplification or protein-level overexpression with previous treatment regimens such as chemotherapy, immunotherapy and targeted therapies including EGFR, BRAF and MEK [74,75]. The efficacy of tepotinib on NSCLC with T790M-negative MET amplification and/or c-MET overexpression after first/second-generation EGFR–TKI resistance is limited, with a mPFS of only 4.9 months [76,77]. The current immunotherapy efficacy for advanced NSCLC after EGFR–TKI resistance needs further improvement, and there is a lack of MET amplification and/or c-MET overexpression subgroup data. Nivolumab monotherapy has limited efficacy after EGFR–TKI resistance, with a mPFS of only 1.5–1.7 months [78,79]. In IMpower 150 and ORIENT-31 studies, EGFR–TKI resistance, followed by immunotherapy combined with bevacizumab and chemotherapy, showed a mPFS of 6.9–9.7 months but no subgroup data on MET amplification and/or c-MET overexpression was reported; meanwhile, safety of the combination therapy regimen needs attention [80,81]. MET-amplified and/or c-MET overexpressed advanced NSCLC patients with EGFR–TKI resistance have limited therapeutic effect with MET inhibitor monotherapy. Only 1 of 12 evaluable patients on inhibitor monotherapy reported an objective response [82]. Dual-target inhibition of EGFR and MET pathways may bring synergistic therapeutic benefit in MET-driven EGFR–TKI-resistant advanced NSCLC patients [83]. Meanwhile, efficacy of savolitinib combined with durvalumab in EGFR wild-type NSCLC with MET alterations is also under exploration in SOUND trial, as previously described.
The combination of EGFR inhibitor and MET-highly selective TKI possesses the potential to prevent or overcome MET-driven resistance to EGFR–TKIs. Acquired resistance to first- and second-generation EGFR–TKIs is often caused by the acquisition of the T790M mutation, which accounts for approximately 60% of resistant cases and has been overcome by third-generation EGFR–TKIs such as osimertinib. For first- and second-generation EGFR–TKIs, acquired resistance for MET-amplification is at least 5% (for example, gefitinib), while up to 25% of acquired resistance is observed with third generation EGFR–TKI (for example, osimertinib) [84]. TATTON study, set up in the back drop of acquired MET amplification associated with EGFR–TKI resistance offered explicit benefit with savolitinib in NSCLC patients without prior third-generation EGFR–TKI, while those who were administered with a prior third-generation EGFR–TKI had a relatively lower rate of response regardless of T790 status, possibly related to larger proportion of patients with ≥3 lines of treatment comprising the prior third-generation EGFR–TKI group (56.5% vs. 22.6% in partB2 + partD). Nonetheless, TATTON program demonstrated beneficial efficacy of savolitinib plus osimertinib combination in the MET-amplified, EGFR mutation–positive setting with acceptable safety profile which is a first in this setting [44]. Further, SAVANNAH phase II trial validates TATTON results with advanced NSCLC patients with MET amplification or c-MET overexpression due to osimertinib-acquired resistance. Initial results from the SAVANNAH trial show a trend toward improved response rates, with increasing level of MET amplified and/or c-MET overexpressed. Across all patients in this analysis, ORR was 32%; mDoR was 8.3 months; and mPFS was 5.3 months, while in high level MET amplification and/or c-MET overexpression subgroup, ORR was 49%; mDoR was 9.3 months; and mPFS was 7.1 months [60]. A summary of key data after EGFR–TKI resistance with secondary MET alterations treated with combination therapies available so far is provided in Table 6.
MET amplification can be detected by using fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC). With MET amplification, MET/CEP7 ratio is as follows: low: ≥1.8 to ≤2.2; intermediate: >2.2 to <5; or high: ≥5 will be applied in clinical settings when treating patients with MET inhibitors [90]. The frequency of MET amplification in NSCLC ranges from 3% to 10% depending on the cut-off of MET copies per cell [91]. c-MET overexpression score of 2+ or 3+ as determined by IHC is considered as MET positive [60]. The TATTON study conducted an exploratory analysis of the relationship between the MET detection method and the dual-target efficacy after third-generation EGFR–TKI resistance: Based on FISH detection, the ORR value of MET local amplification was higher than that of MET polysomy patients although polysomy patients benefited from the treatment. In the MET-amplified population, patients with higher gene copy numbers detected by FISH had a better treatment benefit [44]. Further in SAVANNAH trial, promising clinical efficacy in a population with high MET amplification and/or high threshold c-MET overexpression level (IHC 90+ and/or FISH 10+) with an ORR 49%, mDoR of 9.3 months, and mPFS of 7.1 months was observed. The safety profile was acceptable, similar to that of TATTON study [60]. Further results of the SAVANNAH trial are awaited. However, the sample sizes of these studies are limited. Hence the need to interpret the results with caution is warranted, and further verification is required with larger clinical trials. Further phase III confirmatory trials, SAFFRON and SACHI have been initiated in patients whose disease progressed following treatment with any EGFR-TKI.

8. Conclusions

The conditional approval of savolitinib for the treatment of metastatic METex14-mutated NSCLC is based on encouraging results from phase 2 trial conducted in China including patients with the more aggressive PSC subtype and brain metastasis. Savolitinib is a potent, highly selective MET inhibitor with robust response in advanced NSCLC. Preclinical and clinical data have shown savolitinib as effective and tolerable treatment in advanced NSCLC patients with METex14 skipping mutations. When used in combination with EGFR-TKIs, savolitinib has the potential to overcome resistance to these treatments driven through MET amplifications and/or c-MET overexpression, with future clinical trials verification needed. In conclusion, savolitinib offer another promising targeted treatment in the paradigm of metastatic NSCLC.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/cancers14246122/s1, Figure S1: Proportion of MET-altered NSCLC patients using different detection methods in SAVANNAH trail; Table S1: Baseline characteristics and clinical demographics of SAVANNAH study; Table S2: Efficacy parameters of SAVANNAH study; Table S3: Safety results of SAVANNAH trial.

Author Contributions

Conceptualization, X.Z., Y.L. and S.L.; methodology, X.Z. and Y.L.; investigation, X.Z. and Y.L.; data curation, X.Z. and Y.L.; writing—original draft preparation, X.Z. and Y.L.; writing—review and editing, X.Z., Y.L. and S.L.; supervision, S.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Yao Lu is an employee of AstraZeneca and works as a medical advisor in AstraZeneca, China.

References

  1. Cancer.Net. Lung Cancer—Non-Small Cell: Statistics. Available online: https://www.cancer.net/cancer-types/lung-cancer-non-small-cell/statistics (accessed on 12 June 2022).
  2. Yuan, M.; Huang, L.L.; Chen, J.H.; Wu, J.; Xu, Q. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal. Transduct. Target Ther. 2019, 4, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Liam, C.K.; Stone, E.; Andarini, S.; Liam, Y.S.; Lam, D.C.L.; Lee, P. Molecular testing of metastatic non-small cell lung cancer in the Asia-Pacific region. Respirology 2020, 25, 685–687. [Google Scholar] [CrossRef] [PubMed]
  4. Drilon, A.; Cappuzzo, F.; Ou, S.H.I.; Camidge, D.R. Targeting MET in Lung Cancer: Will Expectations Finally Be MET? J. Thorac. Oncol. 2017, 12, 15–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. 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]
  6. Davies, K.D.; Ritterhouse, L.L.; Snow, A.N.; Sidiropoulos, N. MET Exon 14 Skipping Mutations: Essential Considerations for Current Management of Non-Small Cell Lung Cancer. J. Mol. Diagn. 2022, 24, 841–843. [Google Scholar] [CrossRef]
  7. Stoker, M.; Gherardi, E.; Perryman, M.; Gray, J. Scatter factor is a fibroblast-derived modulator of epithelial cell mobility. Nature 1987, 327, 239–242. [Google Scholar] [CrossRef] [PubMed]
  8. Weidner, K.M.; Behrens, J.; Vandekerckhove, J.; Birchmeier, W. Scatter factor: Molecular characteristics and effect on the invasiveness of epithelial cells. J. Cell Biol. 1990, 111, 2097–2108. [Google Scholar] [CrossRef]
  9. Kaposi-Novak, P.; Lee, J.S.; Gòmez-Quiroz, L.; Coulouarn, C.; Factor, V.M.; Thorgeirsson, S.S. Met-regulated expression signature defines a subset of human hepatocellular carcinomas with poor prognosis and aggressive phenotype. J. Clin. Investig. 2006, 116, 1582–1595. [Google Scholar] [CrossRef]
  10. Baldanzi, G.; Graziani, A. Physiological Signaling and Structure of the HGF Receptor MET. Biomedicines 2014, 3, 1–31. [Google Scholar] [CrossRef] [Green Version]
  11. Raghav, K.; Bailey, A.M.; Loree, J.M.; Kopetz, S.; Holla, V.; Yap, T.A.; Wang, F.; Chen, K.; Salgia, R.; Hong, D. Untying the gordion knot of targeting MET in cancer. Cancer Treat. Rev. 2018, 66, 95–103. [Google Scholar] [CrossRef]
  12. Jo, M.; Stolz, D.B.; Esplen, J.E.; Dorko, K.; Michalopoulos, G.K.; Strom, S.C. Cross-talk between epidermal growth factor receptor and c-Met signal pathways in transformed cells. J. Biol. Chem. 2000, 275, 8806–8811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Soman, N.R.; Correa, P.; Ruiz, B.A.; Wogan, G.N. The TPR-MET oncogenic rearrangement is present and expressed in human gastric carcinoma and precursor lesions. Proc. Natl. Acad. Sci. USA 1991, 88, 4892–4896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. 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] [PubMed]
  15. Mo, H.N.; Liu, P. Targeting MET in cancer therapy. Chronic Dis. Transl. Med. 2017, 3, 148–153. [Google Scholar] [CrossRef]
  16. 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] [Green Version]
  17. Guo, R.; Luo, J.; Chang, J.; Rekhtman, N.; Arcila, M.; Drilon, A. MET-dependent solid tumours—Molecular diagnosis and targeted therapy. Nat. Rev. Clin. Oncol. 2020, 17, 569–587. [Google Scholar] [CrossRef]
  18. Recondo, G.; Che, J.; Jänne, P.A.; Awad, M.M. Targeting MET Dysregulation in Cancer. Cancer Discov. 2020, 10, 922–934. [Google Scholar] [CrossRef]
  19. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef]
  20. Zhang, Z.; Yang, S.; Wang, Q. Impact of MET alterations on targeted therapy with EGFR-tyrosine kinase inhibitors for EGFR-mutant lung cancer. Biomark. Res. 2019, 7, 27. [Google Scholar] [CrossRef]
  21. Bean, J.; Brennan, C.; Shih, J.Y.; Riely, G.; Viale, A.; Wang, L.; Chitale, D.; Motoi, N.; Szoke, J.; Broderick, S.; 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. [Google Scholar] [CrossRef]
  22. Wang, Y.; Li, L.; Han, R.; Jiao, L.; Zheng, J.; He, Y. Clinical analysis by next-generation sequencing for NSCLC patients with MET amplification resistant to osimertinib. Lung Cancer 2018, 118, 105–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Li, X.M.; Li, W.F.; Lin, J.T.; Yan, H.H.; Tu, H.Y.; Chen, H.J.; Wang, B.C.; Wang, Z.; Zhou, Q.; Zhang, X.C.; et al. Predictive and Prognostic Potential of TP53 in Patients with Advanced Non-Small-Cell Lung Cancer Treated With EGFR-TKI: Analysis of a Phase III Randomized Clinical Trial (CTONG 0901. Clin. Lung Cancer 2021, 22, 100–109. [Google Scholar] [CrossRef] [PubMed]
  24. Lai, G.G.Y.; Lim, T.H.; Lim, J.; Liew, P.J.R.; Kwang, X.L.; Nahar, R.; Aung, Z.W.; Takano, A.; Lee, Y.Y.; Lau, D.P.X.; et al. Clonal MET Amplification as a Determinant of Tyrosine Kinase Inhibitor Resistance in Epidermal Growth Factor Receptor-Mutant Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2019, 37, 876–884. [Google Scholar] [CrossRef] [PubMed]
  25. Camidge, D.R.; Barlesi, F.; Goldman, J.W.; Morgensztern, D.; Heist, R.; Vokes, E.; Angevin, E.; Hong, D.S.; Rybkin, I.I.; Barve, M.; et al. A Phase Ib Study of 25 in Combination with Nivolumab in Patients With NSCLC. JTO Clin. Res. Rep. 2022, 3, 100262. [Google Scholar] [CrossRef] [PubMed]
  26. Mathieu, L.N.; Larkins, E.; Akinboro, O.; Roy, P.; Amatya, A.K.; Fiero, M.H.; Mishra-Kalyani, P.S.; Helms, W.S.; Myers, C.E.; Skinner, A.M.; et al. FDA Approval Summary: Capmatinib and Tepotinib for the Treatment of Metastatic NSCLC Harboring MET Exon 14 Skipping Mutations or Alterations. Clin. Cancer Res. 2022, 28, 249–254. [Google Scholar] [CrossRef]
  27. Pfizer. Pfizer’s XALKORI® (Crizotinib) Receives FDA Breakthrough Therapy Designation in Two New Indications. Available online: https://www.pfizer.com/news/press-release/press-release-detail/pfizer_s_xalkori_crizotinib_receives_fda_breakthrough_therapy_designation_in_two_new_indications-0 (accessed on 12 June 2022).
  28. AstraZeneca. Orpathys Approved in China for Patients with Lung Cancer and MET Gene Alterations. Available online: https://www.astrazeneca.com/media-centre/press-releases/2021/orpathys-approved-in-china-for-patients-with-lung-cancer-and-met-gene-alterations.html (accessed on 12 June 2022).
  29. Sequist, L.V.; Han, J.Y.; Ahn, M.J.; Cho, B.C.; Yu, H.; Kim, S.W.; Yang, J.C.; 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 Ib study. Lancet Oncol. 2020, 21, 373–386. [Google Scholar] [CrossRef]
  30. Yang, J.J.; Fang, J.; Shu, Y.Q.; Chang, J.H.; Chen, G.Y.; He, J.X.; Li, W.; Liu, X.Q.; Yang, N.; Zhou, C.; et al. A phase Ib study of the highly selective MET-TKI savolitinib plus gefitinib in patients with EGFR-mutated, MET-amplified advanced non-small-cell lung cancer. Investig. New Drugs. 2021, 39, 477–487. [Google Scholar] [CrossRef]
  31. Markham, A. Savolitinib: First Approval. Drugs 2021, 81, 1665–1670. [Google Scholar] [CrossRef]
  32. 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]
  33. Schuller, A.G.; Barry, E.R.; Jones, R.D.O.; Henry, R.E.; Frigault, M.M.; Beran, G.; Linsenmayer, D.; Hattersley, M.; Smith, A.; Wilson, J.; et al. The MET Inhibitor AZD6094 (Savolitinib, HMPL-504) Induces Regression in Papillary Renal Cell Carcinoma Patient-Derived Xenograft Models. Clin. Cancer Res. 2015, 21, 2811–2819. [Google Scholar] [CrossRef]
  34. Gavine, P.R.; Ren, Y.; Han, L.; Lv, J.; Fan, S.; Zhang, W.; Xu, W.; Liu, Y.J.; Zhang, T.; Fu, H.; et al. Volitinib, a potent and highly selective c-Met inhibitor, effectively blocks c-Met signaling and growth in c-MET amplified gastric cancer patient-derived tumor xenograft models. Mol. Oncol. 2015, 9, 323–333. [Google Scholar] [CrossRef] [PubMed]
  35. Gu, Y.; Sai, Y.; Wang, J.; Yu, M.; Wang, G.; Zhang, L.; Ren, H.; Fan, S.; Ren, Y.; Qing, W.; et al. Preclinical pharmacokinetics, disposition, and translational pharmacokinetic/pharmacodynamic modeling of savolitinib, a novel selective cMet inhibitor. Eur. J. Pharm. Sci. 2019, 136, 104938. [Google Scholar] [CrossRef] [PubMed]
  36. Jones, R.D.O.; Grondine, M.; Borodovsky, A.; San Martin, M.; DuPont, M.; D’Cruz, C.; Schuller, A.; Henry, R.; Barry, E.; Castriotta, L.; et al. A pharmacokinetic-pharmacodynamic model for the MET tyrosine kinase inhibitor, savolitinib, to explore target inhibition requirements for anti-tumour activity. Br. J. Pharmacol. 2021, 178, 600–613. [Google Scholar] [CrossRef] [PubMed]
  37. Ding, Q.; Ou, M.; Zhu, H.; Wang, Y.; Jia, J.; Sai, Y.; Chen, Q.; Wang, J. Effect of food on the single-dose pharmacokinetics and tolerability of savolitinib in Chinese healthy volunteers. Fundam. Clin. Pharmacol. 2022, 36, 210–217. [Google Scholar] [CrossRef] [PubMed]
  38. 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]
  39. Henry, R.E.; Barry, E.R.; Castriotta, L.; Ladd, B.; Markovets, A.; Beran, G.; Ren, Y.; Zhou, F.; Adam, A.; Zinda, M.; et al. Acquired savolitinib resistance in non-small cell lung cancer arises via multiple mechanisms that converge on MET-independent mTOR and MYC activation. Oncotarget 2016, 7, 57651–57670. [Google Scholar] [CrossRef] [Green Version]
  40. Dua, R.; Zhang, J.; Parry, G.; Penuel, E. Detection of hepatocyte growth factor (HGF) ligand-c-MET receptor activation in formalin-fixed paraffin embedded specimens by a novel proximity assay. PLoS ONE 2011, 6, e15932. [Google Scholar] [CrossRef] [Green Version]
  41. Li, Y.; Dong, S.; Tamaskar, A.; Wang, H.; Zhao, J.; Ma, H.; Zhao, Y. Proteasome Inhibitors Diminish c-Met Expression and Induce Cell Death in Non-Small Cell Lung Cancer Cells. Oncol. Res. 2020, 28, 497–507. [Google Scholar] [CrossRef]
  42. Gan, H.K.; Millward, M.; Hua, Y.; Qi, C.; Sai, Y.; Su, W.; Wang, J.; Zhang, L.; Frigault, M.M.; Morgan, S.; et al. First-in-Human Phase I Study of the Selective MET Inhibitor, Savolitinib, in Patients with Advanced Solid Tumors: Safety, Pharmacokinetics, and Antitumor Activity. Clin. Cancer Res. 2019, 25, 4924–4932. [Google Scholar] [CrossRef] [Green Version]
  43. Wang, Y.; Liu, T.; Chen, G.; Gong, J.; Bai, Y.; Zhang, T.; Xu, N.; Liu, L.; Xu, J.; He, J.; et al. Phase Ia/Ib Study of the Selective MET Inhibitor, Savolitinib, in Patients with Advanced Solid Tumors: Safety, Efficacy, and Biomarkers. Oncologist 2022, 27, e342–e383. [Google Scholar] [CrossRef]
  44. Oxnard, G.R.; Yang, J.C.H.; Yu, H.; Kim, S.W.; Saka, H.; Horn, L.; Goto, K.; Ohe, Y.; Mann, H.; Thress, K.S.; et al. TATTON: A multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 2020, 31, 507–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Hartmaier, R.J.; Markovets, A.A.; Ahn, M.J.; Sequist, L.V.; Han, J.Y.; Cho, B.C.; Yu, H.A.; Kim, S.W.; Yang, J.C.; Lee, J.S.; et al. Osimertinib+Savolitinib to Overcome Acquired MET-Mediated Resistance in Epidermal Growth Factor Receptor Mutated MET-Amplified Non-Small Cell Lung Cancer: TATTON. Cancer Discov. 2022, 20, CD-22-0586. [Google Scholar] [CrossRef] [PubMed]
  46. Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Final OS results and subgroup analysis of savolitinib in patients with MET exon 14 skipping mutations (METex14+) NSCLC. In Proceedings of the ELCC 2022 Virtual Meeting, Virtual, 30 March–2 April 2022. [Google Scholar]
  47. Lu, S.; Fang, J.; Cao, L.; Li, X.; Guo, Q.; Zhou, J.; Cheng, Y.; Jiang, L.; Chen, Y.; Zhang, H.; et al. Abstract CT031: Preliminary efficacy and safety results of savolitinib treating patients with pulmonary sarcomatoid carcinoma (PSC) and other types of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutations. In Proceedings of the AACR Annual Meeting 2019, Atlanta, GA, USA, 29 March–3 April 2019. [Google Scholar]
  48. Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Long-Term Efficacy, Safety and Subgroup Analysis of Savolitinib in Chinese Patients with Non-Small Cell Lung Cancers Harboring MET Exon 14 Skipping Alterations. JTO Clin. Res. Rep. 2022, 3, 100407. [Google Scholar] [CrossRef] [PubMed]
  49. Gow, C.H.; Hsieh, M.S.; Wu, S.G.; Shih, J.Y. A comprehensive analysis of clinical outcomes in lung cancer patients harboring a MET exon 14 skipping mutation compared to other driver mutations in an East Asian population. Lung Cancer 2017, 103, 82–89. [Google Scholar] [CrossRef] [PubMed]
  50. Chen, R.; Manochakian, R.; James, L.; Azzouqa, A.G.; Shi, H.; Zhang, Y.; Zhao, Y.; Zhou, K.; Lou, Y. Emerging therapeutic agents for advanced non-small cell lung cancer. J. Hematol. Oncol. 2020, 13, 58. [Google Scholar] [CrossRef]
  51. Bae, H.M.; Min, H.S.; Lee, S.H.; Kim, D.W.; Chung, D.H.; Lee, J.S.; Kim, Y.W.; Heo, D.S. Palliative chemotherapy for pulmonary pleomorphic carcinoma. Lung Cancer 2007, 58, 112–115. [Google Scholar] [CrossRef]
  52. Ung, M.; Rouquette, I.; Filleron, T.; Taillandy, K.; Brouchet, L.; Bennouna, J.; Delord, J.P.; Milia, J.; Mazières, J. Characteristics and Clinical Outcomes of Sarcomatoid Carcinoma of the Lung. Clin. Lung Cancer. 2016, 17, 391–397. [Google Scholar] [CrossRef]
  53. Vieira, T.; Girard, N.; Ung, M.; Monnet, I.; Cazes, A.; Bonnette, P.; Duruisseaux, M.; Mazieres, J.; Antoine, M.; Cadranel, J.; et al. Efficacy of first-line chemotherapy in patients with advanced lung sarcomatoid carcinoma. J. Thorac. Oncol. 2013, 8, 1574–1577. [Google Scholar] [CrossRef] [Green Version]
  54. Hong, J.Y.; Choi, M.K.; Uhm, J.E.; Park, M.J.; Lee, J.; Park, Y.H.; Ahn, J.S.; Park, K.; Han, J.H.; Ahn, M.J. The role of palliative chemotherapy for advanced pulmonary pleomorphic carcinoma. Med. Oncol. 2009, 26, 287–291. [Google Scholar] [CrossRef]
  55. Cheng, H.; Perez-Soler, R. Leptomeningeal metastases in non-small-cell lung cancer. Lancet Oncol. 2018, 19, e43–e55. [Google Scholar] [CrossRef]
  56. Deeken, J.F.; Löscher, W. The blood-brain barrier and cancer: Transporters, treatment, and Trojan horses. Clin. Cancer Res. 2007, 13, 1663–1674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Choueiri, T.K.; Heng, D.Y.C.; Lee, J.L.; Cancel, M.; Verheijen, R.B.; Mellemgaard, A.; Ottesen, L.H.; Frigault, M.M.; L’Hernault, A.; Szijgyarto, Z.; 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. [Google Scholar] [CrossRef] [PubMed]
  58. Cortot, A.; Le, X.; Smit, E.; Viteri, S.; Kato, T.; Sakai, H.; Park, K.; Camidge, D.R.; Berghoff, K.; Vlassak, S.; et al. Safety of MET Tyrosine Kinase Inhibitors in Patients with MET Exon 14 Skipping Non-small Cell Lung Cancer: A Clinical Review. Clin. Lung Cancer 2022, 3, 195–207. [Google Scholar] [CrossRef] [PubMed]
  59. Paik, P.K.; Felip, E.; Veillon, R.; Sakai, H.; Cortot, A.B.; Garassino, M.C.; Mazieres, J.; Viteri, S.; Senellart, H.; Meerbeeck, J.V.; 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]
  60. Ahn, M.J.; De Marinis, F.; Bonanno, L.; Cho, B.C.; Kim, T.M.; Cheng, S.; Novello, S.; Proto, C.; Kim, S.W.; Lee, J.S.; et al. MET Biomarker-based Preliminary Efficacy Analysis in SAVANNAH: Savolitinib+osimertinib in EGFRm NSCLC Post-Osimertinib. In Proceedings of the WCLC 2022, Vienna, Austria, 6–9 August 2022. [Google Scholar]
  61. Li, A.; Chen, H.J.; Yang, J.J. Design and Rationale for a Phase II, Randomized, Open-Label, Two-Cohort Multicenter Interventional Study of Osimertinib with or Without Savolitinib in De Novo MET Aberrant, EGFR-Mutant Patients with Advanced Non-Small-Cell Lung Cancer: The FLOWERS Trial. Clin. Lung Cancer 2022, 22, 00205-4. [Google Scholar] [CrossRef]
  62. Wolf, J.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Robeva, A.; Le Mouhaer, S.; Carbini, M.; Chassot-Agostinho, A.; Heist, R.S. Capmatinib in MET exon 14-mutated, advanced NSCLC: Updated results from the GEOMETRY mono-1 study. In Proceedings of the 2021 ASCO Annual Meeting I, Virtual, 4–8 June 2021. [Google Scholar]
  63. Thomas, M.; Garassino, M.; Felip, E.; Sakai, H.; Le, X.; Veillon, R.; Smit, E.; Mazieres, J.; Cortot, A.; Raskin, J.; et al. Tepotinib in Patients with MET Exon 14 (METex14) Skipping NSCLC: Primary Analysis of the Confirmatory VISION Cohort C. In Proceedings of the WCLC 2022, Vienna, Austria, 6–9 August 2022. [Google Scholar]
  64. Krebs, M.; Spira, A.I.; Cho, B.C.; Besee, 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. In Proceedings of the 2022 ASCO Annual Meeting, Chicago, IL, USA, 3–7 June 2022. [Google Scholar]
  65. Yu, Y.; Ren, Y.; Fang, J.; Cao, L.; Liang, Z.; Guo, Q.; Han, S.; Ji, Z.; Wang, Y.; Sun, Y.; et al. Circulating tumour DNA biomarkers in savolitinib-treated patients with non-small cell lung cancer harbouring MET exon 14 skipping alterations: A post hoc analysis of a pivotal phase 2 study. Ther. Adv. Med. Oncol. 2022, 14, 17588359221133546. [Google Scholar] [CrossRef]
  66. Le, X.; Sakai, H.; Felip, E.; Veillon, R.; Garassino, M.C.; Raskin, J.; Cortot, A.B.; Viteri, S.; Mazieres, J.; Smit, E.F.; et al. Tepotinib Efficacy and Safety in Patients with MET Exon 14 Skipping NSCLC: Outcomes in Patient Subgroups from the VISION Study with Relevance for Clinical Practice. Clin. Cancer Res. 2022, 28, 1117–1126. [Google Scholar] [CrossRef]
  67. 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]
  68. Paik, P.K.; Sakai, H.; Felip, E.; Veillon, R.; Garassino, M.C.; Raskin, J.; Viteri, S.; Mazieres, J.; Cortot, A.; Smit, E.; et al. Tepotinib in patients with MET exon 14 (METex14) skipping advanced NSCLC: Updated efficacy from VISION Cohort A. In Proceedings of the WCLC 2020, Virtual, 28–31 January 2021. [Google Scholar]
  69. Spira, A.; Krebs, M.; Cho, B.C.; Besse, B.; Goldman, J.; Janne, P.; Lee, C.K.; Ma, Z.; Mansfield, A.; Minchom, A.; et al. Amivantamab in non-small cell lung cancer with MET exon 14 skipping muta- tion: Initial results from CHRYSALIS. In Proceedings of the WCLC 2021, Virtual, 8–14 September 2021. [Google Scholar]
  70. Fujino, T.; Kobayashi, Y.; Suda, K.; Koga, T.; Nishino, M.; Ohara, S.; Chiba, M.; Shimoji, M.; Tomizawa, K.; Takemoto, T.; et al. Sensitivity and Resistance of MET Exon 14 Mutations in Lung Cancer to Eight MET Tyrosine Kinase Inhibitors In Vitro. J. Thorac. Oncol. 2019, 14, 1753–1765. [Google Scholar] [CrossRef]
  71. Wolf, J.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Robeva, A.; Le Mouhaer, S.; Carbini, M.; Yovine, A.; Heist, R. Capmatinib in treatment (Tx)-naive MET exon 14-mutated (METex14) advanced non-small cell lung cancer (aNSCLC): Updated results from GEOMETRY mono-1. In Proceedings of the ELCC 2022 Virtual Meeting, Virtual, 30 March–2 April 2022. [Google Scholar]
  72. Moro-Sibilot, D.; Cozic, N.; Pérol, M.; Mazières, J.; Otto, J.; Souquet, P.J.; Bahleda, R.; Wislez, M.; Zalcman, G.; Guibert, S.D.; et al. Crizotinib in c-MET- or ROS1-positive NSCLC: Results of the AcSé phase II trial. Ann. Oncol. 2019, 30, 1985–1991. [Google Scholar] [CrossRef]
  73. Drilon, A.; Clark, J.W.; Weiss, J.; Ou, S.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] [PubMed]
  74. Reis, H.; Metzenmacher, M.; Goetz, M.; Savvidou, N.; Darwiche, K.; Aigner, C.; Herold, T.; Eberhardt, W.E.; Skiba, C.; Hense, J.; et al. MET Expression in Advanced Non-Small-Cell Lung Cancer: Effect on Clinical Outcomes of Chemotherapy, Targeted Therapy, and Immunotherapy. Clin. Lung Cancer 2018, 19, e441–e463. [Google Scholar] [CrossRef] [PubMed]
  75. Wood, G.E.; Hockings, H.; Hilton, D.M.; Kermorgant, S. The role of MET in chemotherapy resistance. Oncogene 2021, 40, 1927–1941. [Google Scholar] [CrossRef] [PubMed]
  76. Bauml, J.; Cho, B.C.; Park, K.; Lee, K.H.; Cho, E.K.; Kim, D.W.; Kim, S.W.; Haura, E.B.; Sabari, J.K.; Sanborn, R.E.; et al. Amivantamab in combination with lazertinib for the treatment of osimertinib-relapsed, chemotherapy-naïve EGFR mutant (EGFRm) non-small cell lung cancer (NSCLC) and potential biomarkers for response. In Proceedings of the 2021 ASCO Annual Meeting I, Virtual, 4–8 June 2021. [Google Scholar]
  77. Park, K.; Zhou, J.; Kim, D.; Ahmad, A.R.; Soo, R.A.; Bruns, R.; Straub, J.; Johne, A.; Scheele, J.; Yang, J.C.; et al. Tepotinib plus gefitinib in patients with MET-amplified EGFR-mutant NSCLC: Long-term outcomes of the INSIGHT study. In Proceedings of the ESMO Asia Congress 2019, Singapore, 22–24 November 2019. [Google Scholar]
  78. Haratani, K.; Hayashi, H.; Tanaka, T.; Kaneda, H.; Togashi, Y.; Sakai, K.; Hayashi, K.; Tomida, S.; Yonesaka, K.; Nonagase, Y.; et al. Tumor immune microenvironment and nivolumab efficacy in EGFR mutation-positive non-small-cell lung cancer based on T790M status after disease progression during EGFR-TKI treatment. Ann. Oncol. 2017, 28, 1532–1539. [Google Scholar] [CrossRef] [PubMed]
  79. Hayashi, H.; Sugawara, S.; Fukuda, Y.; Fujimoto, D.; Miura, S.; Ota, K.; Ozawa, Y.; Hara, S.; Tanizaki, J.; Azuma, K.; et al. A Randomized Phase II Study Comparing Nivolumab with Carboplatin-Pemetrexed for EGFR-Mutated NSCLC with Resistance to EGFR Tyrosine Kinase Inhibitors (WJOG8515L. Clin. Cancer Res. 2022, 28, 893–902. [Google Scholar] [CrossRef] [PubMed]
  80. Reck, M.; Mok, T.; Socinski, M.A.; Jotte, R.M.; Lim, D.W.; Cappuzzo, F.; Orlandi, F.J.; Stroyakovskiy, D.; Nogami, N.; Rodriguez-Abreu, D.; et al. IMpower150: Updated efficacy analysis in patients with EGFR mutations. In Proceedings of the ESMO Virtual Congress 2020, Virtual, 19–21 September 2020. [Google Scholar]
  81. Lu, S.; Wu, L.; Jian, H.; Chen, Y.; Wang, Q.; Fang, J.; Wang, Z.; Hu, Y.; Sun, M.; Han, L.; et al. Sintilimab plus bevacizumab biosimilar IBI305 and chemotherapy for patients with EGFR-mutated non-squamous non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy (ORIENT-31): First interim results from a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2022, 23, 1167–1179. [Google Scholar] [CrossRef] [PubMed]
  82. Baldacci, S.; Mazieres, J.; Tomasini, P.; Girard, N.; Guisier, F.; Audigier-Valette, C.; Monnet, I.; Wislez, M.; Pérol, M.; Dô, P.; et al. Outcome of EGFR-mutated NSCLC patients with MET-driven resistance to EGFR tyrosine kinase inhibitors. Oncotarget 2017, 8, 105103–105114. [Google Scholar] [CrossRef]
  83. Baldacci, S.; Kherrouche, Z.; Cockenpot, V.; Stoven, L.; Copin, M.C.; Werkmeister, E.; Marchand, N.; Kyheng, M.; Tulasne, D.; Cortot, A.B. MET amplification increases the metastatic spread of EGFR-mutated NSCLC. Lung Cancer 2018, 125, 57–67. [Google Scholar] [CrossRef]
  84. Wang, Q.; Yang, S.; Wang, K.; Sun, S.Y. MET inhibitors for targeted therapy of EGFR TKI-resistant lung cancer. J. Hematol. Oncol. 2019, 12, 63. [Google Scholar] [CrossRef] [Green Version]
  85. Yu, H.A.; Ambrose, H.; Baik, C.; Cho, B.C.; Cocco, E.; Goldberg, S.B.; Goldman, J.W.; Kraljevic, S.; de Langen, A.J.; Okamoto, I.; et al. ORCHARD osimertinib + savolitinib interim analysis: A biomarker-directed phase II platform study in patients (pts) with advanced non-small cell lung cancer (NSCLC) whose disease has progressed on first-line (1L) osimertinib. In Proceedings of the ESMO Congress 2021, Paris, France, 16–21 September 2021. [Google Scholar]
  86. 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]
  87. Wu, Y.L.; Cheng, Y.; Zhou, J.; Lu, S.; Zhang, Y.; Zhao, J.; Kim, D.W.; Soo, R.A.; Kim, S.W.; Pan, H.; 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 Ib/2, multicentre, randomised trial. Lancet Respir Med. 2020, 8, 1132–1143. [Google Scholar] [CrossRef] [PubMed]
  88. Liam, C.K.; Ahmad, A.R.; Hsia, T.C.; Zhou, J.; Kim, D.W.; Soo, R.A.; Cheng, Y.; Lu, S.; Shin, S.W.; Yang, J.C.H.; et al. Tepotinib + gefitinib in patients with EGFR-mutant NSCLC with MET amplification: Final analysis of INSIGHT. In Proceedings of the 2022 AACR Annual Meeting, New Orleans, LA, USA, 8–13 April 2022. [Google Scholar]
  89. Goldman, J.W.; Horinouchi, H.; Cho, B.C.; Tomasini, P.; Dunbar, M.; Hoffman, D.; Parikh, A.; Blot, V.; Camidge, D.R. Phase 1/1b study of telisotuzumab vedotin (Teliso-V) + osimertinib (Osi), after failure on prior Osi, in patients with advanced, c-Met overexpressing, EGFR-mutated non-small cell lung cancer (NSCLC. In Proceedings of the 2022 ASCO Annual Meeting, Chicago, IL, USA, 3–7 June 2022. [Google Scholar]
  90. Yin, W.; Cheng, J.; Tang, Z.; Toruner, G.; Hu, S.; Guo, M.; Robinson, M.; Medeiros, L.J.; Tang, G. MET Amplification (MET/CEP7 Ratio ≥ 1.8) Is an Independent Poor Prognostic Marker in Patients with Treatment-naive Non-Small-cell Lung Cancer. Clin. Lung Cancer 2021, 22, e512–e518. [Google Scholar] [CrossRef] [PubMed]
  91. Fang, L.; Chen, H.; Tang, Z.; Kalhor, N.; Liu, C.H.; Yao, H.; Hu, S.; Lin, P.; Zhao, J.; Luthra, R.; et al. MET amplification assessed using optimized FISH reporting criteria predicts early distant metastasis in patients with non-small cell lung cancer. Oncotarget 2018, 9, 12959–12970. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Exploration of MET as oncogene and the journey leading to the development of MET–TKI. 1 Based on overall NSCLC population. 2 Based on treatment-naïve NSCLC population. 3 Based on EGFR-positive treatment-naïve NSCLC population.
Figure 1. Exploration of MET as oncogene and the journey leading to the development of MET–TKI. 1 Based on overall NSCLC population. 2 Based on treatment-naïve NSCLC population. 3 Based on EGFR-positive treatment-naïve NSCLC population.
Cancers 14 06122 g001
Figure 2. Chemical structure of Savolitinib.
Figure 2. Chemical structure of Savolitinib.
Cancers 14 06122 g002
Figure 3. Key milestones and clinical trials in the development of savolitinib for non-small cell lung cancer.
Figure 3. Key milestones and clinical trials in the development of savolitinib for non-small cell lung cancer.
Cancers 14 06122 g003
Table 1. Proportion of different MET alterations in NSCLC patients.
Table 1. Proportion of different MET alterations in NSCLC patients.
MET AlterationsProportion, %Publication [Reference]
METex14NSCLC 10.9–4Davies KD et al. [6]
PSC subtype20–31.8Mo HN et al. [15]
Tong JH et al. [16]
MET Fusion 1 0.5Recondo G et al. [18]
MET Overexpression 1 13.7–63.7Guo R et al. [17]
MET Amplification 2 1–5Guo R et al. [17]
Secondary MET Amplification1/2G EGFR–TKI resistance5–22Recondo G et al. [18]
Bean J et al. [21]
3G EGFR–TKI resistance5–50Wang Y et al. [22]
MET Amplification Co-occurrence with EGFR Mutation 3 2–11Li XM et al. [23]
Lai GGY et al. [24]
1 Based on overall NSCLC population. 2 Based on treatment-naïve NSCLC population. 3 Based on EGFR-positive treatment-naïve NSCLC population. EGFR, Epidermal Growth Factor Receptor; TKI, Tyrosine Kinase Inhibitor; MET, Mesenchymal Epithelial Transition; NSCLC, Non-Small Cell Lung Cancer; PSC, Pulmonary Sarcomatoid Carcinoma.
Table 2. Baseline characteristics of phase II trial conducted in China [48].
Table 2. Baseline characteristics of phase II trial conducted in China [48].
AgeSexSmoking HistoryECOG Performance Status
Median Age, Years<75 Years≥75 YearsFemaleMaleNon-SmokersSmokers013
Full Analysis Set
(n = 70)
68.7 (51.7–85.0)54 (77%)16 (23%)29 (41%)41 (59%)42 (60%)28 (40%)12 (17%)57 (81%)1 (1%)
Type of Primary TumorPSC
(n = 25)
69.3 (54.1–84.8)19 (76%)6 (24%)8 (32%)17 (68%)13 (52%)12 (48%)3 (12%)22 (88%)0
Other NSCLC
(n = 45)
68.1 (51.7–85.0)35 (78%)10 (22%)21 (47%)24 (53%)29 (64%)16 (36%)9 (20%)35 (78%)1 (2%)
Prior Anti-tumor TreatmentPre-treated
(n = 42)
67.7 (51.7–84.8)38 (90%)4 (10%)17 (40%)25 (60%)28 (67%)14 (33%)8 (19%)34 (81%)0
Treatment-naïve
(n = 28)
74.5 (56.0–85.0)16 (57%)12 (43%)17 (40%)16 (57%)14 (50%)14 (50%)4 (14%)23 (82%)1 (4%)
Brain Metastases StatusBrain metastases
(n = 15)
68.6 (51.7–84.8)11 (73%)4 (27%)7 (47%)8 (53%)11 (73%)4 (27%)3 (20%)12 (80%)0
Non-brain metastases
(n = 55)
68.7 (51.9–85.0)43 (78%)12 (22%)22 (40%)33 (60%)31 (56%)24 (44%)9 (16%)45 (82%)1 (2%)
HistologyPrior Anti-tumor TreatmentBrain Involvement at Baseline
Pulmonary sarcomatoid carcinomaOther NSCLC subtypesYesNo
AdenocarcinomaSquamous cell carcinomaAdenosquamous carcinomaNSCLC, not otherwise specified
Full Analysis Set
(n = 70)
25 (36%)40 (57%)3 (3%)1 (1%)1 (1%)42 (60%)28 (40%)15 (21%)
Type of Primary TumorPSC
(n = 25)
25 (100%)-12 (48%)13 (52%)2 (8%)
Other NSCLC
(n = 45)
-40 (89%)3 (7%)1 (2%)1 (2%)30 (67%)15 (33%)13 (29%)
Prior Anti-tumor TreatmentPre-treated
(n = 42)
12 (29%)27 (64%)2 (5%)1 (2%)042 (100%)-11 (26%)
Treatment-naïve
(n = 28)
13 (46%)13 (46%)1 (4%)01 (4%)-28 (100%)4 (14%)
Brain Metastases StatusBrain metastases
(n = 15)
2 (13%)13 (87%)00011 (73%)4 (27%)15 (100%)
Non-brain metastases
(n = 55)
23 (42%)27 (49%)3 (5%)1 (2%)1 (2%)33 (60%)22 (40%)-
Data in median (IQR) or n (%). NSCLC, Non-Small Cell Lung Cancer; PSC, Pulmonary Sarcomatoid Carcinoma; ECOG, Eastern Cooperative Oncology Group.
Table 3. Investigator-Assessed Responses in the Tumor-Response-Evaluable Set and the Full Analysis Set of Phase II Trial Conducted in China [48].
Table 3. Investigator-Assessed Responses in the Tumor-Response-Evaluable Set and the Full Analysis Set of Phase II Trial Conducted in China [48].
ORR, n (%)DCR, n (%)Median DOR, Months 1Median TTR, Months 1
Tumor-Response-Evaluable Set (n = 62)Total (n = 62)33 (53.2%)57 (91.9%)6.91.4
PSC (n = 20)10 (50.0%)18 (90.0%)12.41.4
Other NSCLC subtypes (n = 42)23 (54.8%)39 (92.9%)5.61.4
Pretreated (n = 38)20 (52.6%)34 (89.5%)10.91.4
Treatment-naive (n = 24)13 (54.2%)23 (95.8%)5.61.4
Brain metastases (n = 14)9 (64.3%)14 (100.0%)4.91.5
Non-brain metastases (n = 48)24 (50.0%)43 (89.6%)7.01.4
Full Analysis Set (n = 70)Total (n = 70)33 (47.1%)57 (81.4%)n/An/A
PSC (n = 25)10 (40.0%)18 (72.0%)n/An/A
Other NSCLC subtypes (n = 45)23 (51.1%)39 (86.7%)n/An/A
Pretreated (n = 42)20 (47.6%)34 (81.0%)n/An/A
Treatment-naive (n = 28)13 (46.4%)23 (82.1%)n/An/A
Brain metastases (n = 15)9 (60.0%)14 (93.3%)n/An/A
Non-brain metastases (n = 55)24 (43.6%)43 (78.2%)n/An/A
1 DOR and TTR were analyzed in the tumor-response-evaluable set. DCR, disease control rate; DOR, duration of response; n/A, not applicable; NSCLC, non-small cell lung cancer; ORR, objective response rate; PSC, pulmonary sarcomatoid carcinoma; TTR, time to response.
Table 4. Adverse events (>30%) in the full analysis set of phase II trials conducted in China (n = 70) [48].
Table 4. Adverse events (>30%) in the full analysis set of phase II trials conducted in China (n = 70) [48].
Any Grade≥Grade 3
All-cause adverse eventsAny event70 (100.0%)46 (65.7%)
Peripheral edema40 (57.1%)6 (8.6%)
Nausea37 (52.9%)0
Hypoalbuminemia29 (41.4%)1 (1.4%)
Elevated alanine aminotransferase27 (38.6%)7 (10.0%)
Elevated aspartate aminotransferase27 (38.6%)9 (12.9%)
Decreased appetite24 (34.3%)0
Vomiting23 (32.9%)0
Pyrexia21 (30.0%)1 (1.4%)
Treatment-related adverse eventsAny event70 (100.0%)32 (45.7%)
Peripheral edema39 (55.7)6 (8.6)
Nausea32 (45.7)0
Hypoalbuminemia16 (22.9)0
Elevated alanine aminotransferase27 (38.6)7 (10.0%)
Elevated aspartate aminotransferase26 (37.1)9 (12.9%)
Decreased appetite14 (20.0%)0
Vomiting18 (25.7%)0
Pyrexia11 (15.7%)1 (1.4%)
Data in n (%). Derived from latest safety analysis of phase II trial (NCT02897479) [48].
Table 5. Data summary of MET inhibitors in METex14 mutation.
Table 5. Data summary of MET inhibitors in METex14 mutation.
Savolitinib 1Capmatinib 2Tepotinib 3Crizotinib 4Amivantamab 5
ApprovalChina approved in June 2021Approved in the US in 2020Approved in Japan in 2020FDA breakthrough therapy designationApproved in the US in May 2021
MechanismMETi IbMETi IbMETi IbALK/ROS1/METi IaAnti-MET and EGFR antibody
n4516031325/6946
Population100% Chinese patients20.2% Asian patients33.9% Asian patientsUnknown/15.9% Asian patients50.9% Asian patients
Proportion of brain metastases28.9%16.9%18.2%Unknown18.2%
Dose600 mg (BW ≥ 50 kg), or
400 mg (BW < 50 kg) OD
400 mg BID500 mg OD250 mg BID1050 mg (<80 kg), or
1400 mg (≥80 kg)
ORR54.8%52.5%50.8%12.0%/32.3%32.6%
DCR92.9%88.1%75.4%44.0%/unknown76.1%
Median PFS, Months6.912.4/12.5/5.4/6.911.23.6/7.36.7
Common Grade 3/4 AEsElevated AST
Elevated ALT
Peripheral edema
(No interstitial lung disease occurred in registry studies)
Peripheral edema
Difficulty breathing
Fatigue
Elevated ALT
Weak
Pneumonia
Peripheral edema
Generalized edema
Vomit
Nausea
Interstitial lung disease
Elevated transaminases
Difficulty breathing
Hypophosphatemia
Lymphopenia
Pulmonary embolism
Interstitial lung disease
Rash
Hypoalbuminemia
Difficulty breathing
1 The number of patients and the proportion of patients with brain metastases are based on other types of NSCLC in general, and the ORR, DCR and median PFS data are derived from data from other types of NSCLC in the efficacy-evaluable set [48]; safety data is analyzed based on the overall patient (n = 70) [38,48]. 2 Data derived from the latest analysis of four different cohorts from GEOMETRY mono-1 study: cohort 4, expansion cohort 6, cohort 5b and expansion cohort 7. Number of patients, proportion of brain metastases, ORR and DCR represent four cohorts in total; proportion of population based on cohort 4, 5b and 7; mPFS reflect results of four cohorts, respectively [62,71]. 3 Data based on VISION study cohort A + cohort C latest overall analysis [63]. 4 Patient population, number of patients, ORR, DCR and median PFS data are derived from two different studies of AcSé [72] and PROFILE-1001 [73]; safety data is based on combination of these two trials. 5 Data from latest analysis of CHRYSALIS study [64]. EGFR, Epidermal Growth Factor Receptor; TKI, Tyrosine Kinase Inhibitor; MET, Mesenchymal Epithelial Transition; ALK, Anaplastic Lymphoma Kinase; ROS1, ROS proto-oncogene 1; OD, Once Daily; BID, twice daily; ORR, Objective Response Rate; DCR, Disease Control Rate; PFS, Progression Free Survival; AE, Adverse Event; AST, Aspartate aminotransferase; ALT, Alanine aminotransferase.
Table 6. Summary of key data after EGFR–TKI resistance with secondary MET alterations treated with combination therapies.
Table 6. Summary of key data after EGFR–TKI resistance with secondary MET alterations treated with combination therapies.
CombinationPublication [Reference]nPatient PopulationMET StatusORRMedian PFS, Months
-Sequist LV et al. [29]
Hartmaier RJ et al. [45] 1
931/2G EGFR–TKI resistance, T790M-FISH: MET GCN ≥ 5 or MET/CEP7 ≥ 2;
IHC: 3+ in ≥50% tumor cell;
NGS: ≥ 20% tumor cell, ≥200X seq, GCN ≥ 5
Part B2: 64.7%Part B2: 9.1
Part D: 61.9%Part D: 9.0
Hartmaier RJ et al. [45] 2693G EGFR–TKI (osimertinib) resistanceFISH: MET GCN ≥ 5 or MET/CEP7 ≥ 2;
IHC: 3+ in ≥50% tumor cell;
NGS: ≥ 20% tumor cell, ≥200X seq, GCN ≥ 5
33.3%5.5
Yu HA et al. [85]173G EGFR–TKI (osimertinib) resistanceNGS: GCN range from 7 to 6841.2%Unknown
Ahn MJ et al. [60]1933G EGFR–TKI (osimertinib) resistanceFISH: MET GCN ≥ 5 or MET/CEP7 ≥ 2;
IHC: 3+ in ≥50% tumor cell
Overall: 32%Overall: 5.3
FISH10+ or IHC90+: 49.1% 3FISH10+ or IHC90+: 7.1
Capmatinib + GefitinibWu YL et al. [86]1001/2G EGFR–TKI resistance, T790M-FISH: GCN ≥ 44 ≤ GCN < 6: 22.2%4 ≤ GCN < 6: 5.4
GCN ≥ 6: 47.2%GCN ≥ 6: 5.5
IHC: 3+ in ≥50% tumor cellIHC3+: 32.1%IHC3+: 5.5
Tepotinib + GefitinibWu YL et al. [87]
Liam CK et al. [88]
311/2G EGFR–TKI resistance, T790M-FISH: GCN ≥ 5 or MET/CEP7 ≥ 2Overall: 45.2%Overall: 4.9
MET amp: 66.7%MET amp: 16.6
IHC: 2+ or 3+IHC3+: 68.4%IHC3+: 8.3
Amivantamab + LazertinibBauml J et al. [76]453G EGFR–TKI (osimertinib) resistance, without previous chemotherapyNo MET selectionOverall: 35.6%Overall: 4.9
EGFR/MET dependent: 47.1% 4EGFR/MET dependent: 6.7
Unknown/non-EGFR/MET: 28.6%Unknown/non-EGFR/MET: 4.1
EGFR/MET IHC+: 90.0%EGFR/MET IHC+: 12.5
Telisotuzumab vedotin + OsimertinibGoldman JW et al. [89]193G EGFR–TKI (osimertinib) resistanceIHC: 3+ in ≥25% tumor cell57.9%Unknown
1 Data based on part B2 (n = 51) and part D (n = 42) of TATTON study. 2 Data based on part B1 of TATTON study. 3 Represents high MET amplification and/or high c-MET overexpression subgroup (n = 108); FISH10+: MET GCN ≥ 10; IHC90+: 3+ in ≥90% tumor cell. 4 EGFR/MET dependent, EGFR/MET dependent mechanism of resistance (n = 17); unknown/non-EGFR/MET, unknown mechanism or non-EGFR/MET mechanism of resistance to osimertinib (n = 28); EGFR/MET IHC+, high IHC results (combined EGFR + MET H score > 400) (n = 10). EGFR, Epidermal Growth Factor Receptor; TKI, Tyrosine Kinase Inhibitor; MET, Mesenchymal Epithelial Transition; FISH, Fluorescence In Situ Hybridization; IHC, Immunohistochemistry; GCN, Gene Copy Number; CEP7, Centromere 7; ORR, Objective Response Rate; PFS, Progression Free Survival.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zhu, X.; Lu, Y.; Lu, S. Landscape of Savolitinib Development for the Treatment of Non-Small Cell Lung Cancer with MET Alteration—A Narrative Review. Cancers 2022, 14, 6122. https://doi.org/10.3390/cancers14246122

AMA Style

Zhu X, Lu Y, Lu S. Landscape of Savolitinib Development for the Treatment of Non-Small Cell Lung Cancer with MET Alteration—A Narrative Review. Cancers. 2022; 14(24):6122. https://doi.org/10.3390/cancers14246122

Chicago/Turabian Style

Zhu, Xiaokuan, Yao Lu, and Shun Lu. 2022. "Landscape of Savolitinib Development for the Treatment of Non-Small Cell Lung Cancer with MET Alteration—A Narrative Review" Cancers 14, no. 24: 6122. https://doi.org/10.3390/cancers14246122

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