Next Article in Journal
Proton Bragg Peak FLASH Enables Organ Sparing and Ultra-High Dose-Rate Delivery: Proof of Principle in Recurrent Head and Neck Cancer
Next Article in Special Issue
Targeting MET Amplification: Opportunities and Obstacles in Therapeutic Approaches
Previous Article in Journal
Polyphenol-Based Nanoparticles: A Promising Frontier for Enhanced Colorectal Cancer Treatment
Previous Article in Special Issue
The Development and Role of Capmatinib in the Treatment of MET-Dysregulated Non-Small Cell Lung Cancer—A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

A Systematic Review of Mesenchymal Epithelial Transition Factor (MET) and Its Impact in the Development and Treatment of Non-Small-Cell Lung Cancer

1
Department of Clinical Sciences, Lund University, 22184 Lund, Sweden
2
Wallenberg Center for Molecular Medicine, Lund University, 22184 Lund, Sweden
3
Lund Stem Cell Center, Lund University, 22184 Lund, Sweden
4
Department of Cardiothoracic Surgery and Transplantation, Skåne University Hospital, 22242 Lund, Sweden
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(15), 3827; https://doi.org/10.3390/cancers15153827
Submission received: 14 June 2023 / Revised: 20 July 2023 / Accepted: 25 July 2023 / Published: 27 July 2023
(This article belongs to the Special Issue Roles of MET in Cancer Development and Treatment)

Abstract

:

Simple Summary

Lung cancer is the type of cancer that kills the most people in the world each year. It is difficult to diagnose lung cancer in the early stages and there are only few treatment options available once the cancer has spread. The mesenchymal epithelial transition factor (MET) gene is of importance in lung cancer development, and mutations in this gene are related to poor prognosis. Consequently, it is important to develop new treatment options that specifically target the MET protein. In this systematic review, we aimed to summarize the existing knowledge on the impact of MET on lung cancer development and the effect of currently available medications. Our hope is that the findings of this systematic review will deepen the understanding of other researchers, possibly providing a guiding hand as to what may be most interesting to focus on in future research projects on this subject.

Abstract

Lung cancer represents the leading cause of annual cancer-related deaths worldwide, accounting for 12.9%. The available treatment options for patients who experience disease progression remain limited. Targeted therapeutic approaches are promising but further understanding of the role of genetic alterations in tumorigenesis is imperative. The MET gene has garnered great interest in this regard. The aim of this systematic review was to analyze the findings from multiple studies to provide a comprehensive and unbiased summary of the evidence. A systematic search was conducted in the reputable scientific databases Embase and PubMed, leading to the inclusion of twenty-two articles, following the PRISMA guidelines, elucidating the biological role of MET in lung cancer and targeted therapies. The systematic review was registered in PROSPERO with registration ID: CRD42023437714. MET mutations were detected in 7.6–11.0% of cases while MET gene amplification was observed in 3.9–22.0%. Six studies showed favorable treatment outcomes utilizing MET inhibitors compared to standard treatment or placebo, with increases in PFS and OS ranging from 0.9 to 12.4 and 7.2 to 24.2 months, respectively, and one study reporting an increase in ORR by 17.3%. Furthermore, patients with a higher mutational burden may derive greater benefit from treatment with MET tyrosine kinase inhibitors (TKIs) than those with a lower mutational burden. Conversely, two studies reported no beneficial effect from adjunctive treatment with a MET targeted therapy. Given these findings, there is an urgent need to identify effective therapeutic strategies specifically targeting the MET gene in lung cancer patients.

1. Introduction

Lung cancer is one of the most common malignancies, causing 12.9% of cancer-related deaths worldwide, resulting in 1.3 million deaths annually [1,2,3,4]. Between 80.0 and 85.0% of lung cancer cases are non-small cell lung cancer (NSCLC) [3,5,6]. Late diagnosis is a major problem, contributing to the short median survival of approximately 18 months and the overall 5-year survival rate for lung cancer of 15–21%, depending on gender [7,8]. Up to 75.0% of patients with newly diagnosed NSCLC have locally advanced or metastasized disease at diagnosis, with a 5-year survival rate below 5.0% [1]. To this day, locally advanced or metastasized NSCLC is commonly treated with platinum-based chemotherapy, offering modest efficacy, with response rates of 20.0–30.0% and a plethora of side effects, or immunotherapy [3,5,9]. Targeted therapies for several types of cancer, including NSCLC, have emerged as a beneficial option for subsets of patients. Current treatment guidelines for advanced NSCLC call for broad molecular profiling to identify and guide the choice of potential targeted therapy options [10]. The proportion of patients with NSCLC receiving next generation sequencing (NGS) is low, which is in part due to insufficient availability of tumor tissue at the time of diagnosis. Approximately 60.0–65.0% of patients undergo testing for mutations in the epidermal growth factor receptor gene (EGFR) and less than 25.0% of patients are tested for alterations in the mesenchymal epithelial transition factor gene (MET) [11].
EGFR is a transmembrane receptor that is involved in several signaling pathways; it promotes cell proliferation and is anti-apoptotic. Overexpression of the EGFR gene is a well-known pathological mechanism in NSCLC, present in 43.0–89.0% of NSCLC cases, which can lead to poorer outcomes [12]. Several EGFR TKIs exist; however acquired resistance to these therapies is common and alterations in the MET gene have been proven to be a contributing factor [13]. The protein MET is a transmembrane receptor tyrosine kinase (RTK) with a central role in cell motility, morphogenesis, proliferation, survival, and invasion (Figure 1A) [5,14,15,16]. Alterations in the MET gene, such as gene copy number (GCN) gain, mutation, or overexpression of the protein, have been reported in NSCLC [17]. The only known ligand to MET is hepatocyte growth factor (HGF) [6]. HGF is found in healthy lung tissue but is often overexpressed in NSCLC. Aberrant signaling through the HGF/MET pathway has clinically been linked to oncogenic potential and poor outcomes in NSCLC, with shortened overall survival (OS) and progression-free survival (PFS). However, studies showing the opposite also exist, introducing a contradiction in this area of research. One proven cause of unfavorable outcomes in MET altered lung cancer is acquired resistance to EGFR TKIs, underlining the need for more efficient targeted therapies [1,5,6,14,15,17,18,19,20]. The frequency of any form of dysregulation of MET in NSCLC ranges from 3.0 to 7.0%. Sporadic GCN gain of MET is detected in 1.0–4.0% of wild type EGFR NSCLC cases. MET exon 14 skipping mutations occur in approximately 3.0% of NSCLC cases [2,3,18,20]. Amplification of the MET gene is the most common type of dysregulated signaling in NSCLC with acquired resistance to EGFR TKIs, with reported frequencies between 5.0 and 26.0% [20,21]. Alterations in MET have been shown to upregulate the expression of EGFR ligands, which in turn increases EGFR signaling, promoting cell proliferation, angiogenesis, and apoptosis [15]. Increased expression of HGF can also promote resistance to EGFR TKIs by supporting clonal selection of a subpopulation of cells with MET amplification [1]. Despite the fact that acquired resistance to EGFR TKIs is very common, they remain the preferred first-line treatment for locally advanced or metastatic EGFR mutation-positive NSCLC [15,22,23]. For patients with acquired resistance to EGFR TKIs caused by upregulation or amplification of MET, it may be beneficial to treat with a combination of inhibitors of both MET and EGFR, as this has been shown to have a synergistic inhibitory effect on the proliferation of cancer cells [2,21,22]. A combination of EGFR and MET TKIs has been shown to possibly delay the occurrence of resistance to EGFR TKIs [24]. For an overview of the EGFR and MET TKIs discussed in this article, see Table 1 (Figure 1B).
An issue with these types of targeted therapies remains the inherent genetic heterogeneity of NSCLC. Several mutations may co-exist in the same patient and complicate the interpretation of specific drug effects [3,25]. An additional aspect to take into consideration is the variability in methodologies employed to quantify aberrant MET expression. Some studies use immunohistochemistry (IHC) with the definition of MET positivity ranging from 1+ to 3+ and MET negativity varying from 0 to 1+ [2,17,18,20]. Other studies apply gene copy number (GCN) ≥ 5 or MET/centromere 7 ratio ≥ 2.0 as the lower limit for defined MET positivity [20].
Genetic profiling carried out by NGS is a rapidly progressing area of research within oncology. The methods used are becoming increasingly cost and time efficient, allowing for improved individual genetic testing and tailored therapy options [26]. The aim of this systematic review was to provide a comprehensive and unbiased summary of the evidence investigating the role of MET and aberrant expression of the protein in lung cancer development and treatment thereof.

2. Methods

2.1. Search Strategy

The Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) guidelines were used in conducting this systematic review; for the full PRISMA 2020 Checklist, see Appendix B [27] (Table A1 and Table A2). A protocol was not established. The systematic review was registered in PROSPERO with registration ID CRD42023437714. Medically relevant databases containing existing literature were systematically searched by the authors for articles related to MET, lung cancer, and targeted MET therapies. The systematic search was conducted in Embase and PubMed in February of 2023 with the help of two search queries (Figure 2, Appendix A) based on the population, intervention, comparison, and outcome (PICO) model. Only clinical trials and randomized clinical trials were included.

2.2. Exclusion and Inclusion Criteria

After the initial search in Embase and PubMed, all duplicates were removed. All remaining articles were screened independently by two of the authors, E.B. and F.S., for relevance based on the title, abstract, and full article. Articles were included if one or both parties deemed it relevant to the subject. Articles that were published in 2012 or earlier were excluded as well as supplemental materials and conference abstracts. A full text screening of all eligible articles was performed independently by the two parties, excluding irrelevant and ongoing trials. A flow diagram of exclusion steps is presented in Figure 3.

2.3. Data Extraction and Processing

The included papers were grouped by investigated drug; the main characteristics are described in Table 2 and Table 3. The main outcome measures presented and analyzed in this article are PFS and OS. Data on prevalence of MET mutations and amplifications in lung cancer were also analyzed and included in this article.

3. Bias

Publication bias is a factor inevitably impacting systematic reviews due to positive selection bias in the publication of research articles. This may lead to overestimation of the therapeutic effect of the investigated drugs. In this systematic review, several articles reported no effect of the evaluated drugs on PFS and OS among patients with lung cancer. By simultaneous screening of all articles by separate parties, the risk of bias in the process of selecting articles to be included in this systematic review was minimized. Furthermore, the Cochrane Risk of Bias Tool “robvis” was adapted to create risk of bias plots for all RCT’s and clinical trials included in this systematic review, see Appendix C (Figure A1 and Figure A2) [33]. None of the authors of this systematic review declare any conflicts of interest related to the article.

4. Results

This systematic review identified a total of 786 eligible articles by the two search queries (Appendix A) employed following the PRISMA guidelines. Exclusion of 256 duplicates resulted in 530 remaining articles. After the assessment of article titles, 314 articles remained followed by further exclusion based on the abstract, resulting in 114 remaining articles. Six articles were excluded based on publication year and 71 studies were excluded due to being conference abstracts or supplements. Consequently, 37 full articles remained for the final full text screening. A total of 22 articles were judged to be relevant to this systematic review (Figure 3). Six of the included articles present data regarding the prevalence and significance of aberrant signaling through the HGF/MET pathway in lung cancer. The remaining 16 articles present data on the effect of specific targeted therapies on mortality and morbidity of lung cancer patients.

4.1. Epidemiology

The reported patient characteristics varied greatly between the included studies, see Appendix C (Table A3). Landi et al. reported the lowest mean age in all the studies (56.0 years) while Moro-Sibilot et al. reported the highest in their subgroup of patients with MET mutations (72.0 years) [18,29]. In regard to the included subjects’ gender, Matsumoto et al. included a vast overweight of women compared to men in their clinical trial (80.3% women), while Okamoto et al. reported the highest percentage of men included (77.0%) [3,19]. Similar to age and gender, smoking history also varied between the studies. The highest reported prevalence of never smokers was seen in the clinical trial conducted by Matsumoto et al., with 89.1% of the patients being never smokers in an EGFR WT population [19]. Interestingly, both of the MET-negative cohorts in a RCT by Spigel et al. held smaller fractions of patients with no smoking history (7.0% in the treated subgroup and 3.0% in the placebo subgroup) compared to the MET+ cohorts in the same study (20.0% in the treated subgroup and 19.0% in the placebo subgroup) [6]. In the study by Moro-Sibilot et al., the authors reported double the amount of patients with a no smoking history in the MET mutated cohort compared to the MET amplified cohort (MET mutated: 48.0%, MET amplified: 24.0%) (Table A3) [29].

4.2. Prevalence of Aberrant MET Expression

The reported prevalence of the different MET gene alterations varies greatly between the articles included in this systematic review. In a prospective clinical trial by Palmero et al., genetic testing of treatment naïve NSCLC patients was carried out to identify genetic alterations. Next generation sequencing (NGS) of circulating cell free tumor DNA in blood was compared to standard-of-care tissue-based biopsy testing in order to determine the patients’ mutational burden. The results showed no difference between the methods’ ability to identify mutations in NSCLC. Out of 186 tested patients, 11.0% harbored MET exon 14 skipping mutations (METex14) and 22.0% harbored amplification of the MET gene [11].
In a retrospective clinical study by Okamoto et al., 295 patients were tested for MET mutations and 229 patients were tested for MET amplification. The results showed a prevalence of 7.6% for mutations in the MET gene and 3.9% for MET amplification, with a median GCN of 8.8. The median OS was found to be non-significantly longer among patients without MET amplification [3].
Sacher et al. carried out a single arm, single center clinical trial, testing the EGFR TKI erlotinib on 22 NSCLC patients with varying EGFR mutational statuses. All included patients were naïve to any systemic anti-cancer treatment. MET amplification was determined by fluorescence in situ hybridization (FISH) and MET positivity was defined as GCN ≥ 4. Protein expression was assessed through IHC and MET positivity was defined as an H-score ≥ 100. The prevalence of any aberrant MET expression in this study was 45.0%, while MET amplification specifically was seen in 9.0% of the patients [28].
Helman et al. conducted a retrospective study that performed NGS on plasma samples from patients suffering from NSCLC harboring EGFR mutations. NGS was carried out at study enrolment before receiving treatment and after disease progression to evaluate the effect of the EGFR tyrosine kinase inhibitor (TKI) rociletinib. Prior to treatment with rociletinib, 15.0% of the NGS screened patients were positive for genetic alterations in the MET gene. After disease progression on treatment with rociletinib, another 7.6% of the patients with acquired resistance to the drug had acquired amplification of the MET gene. This amplification was found to be caused by focal amplification in 4.5% of the patients and by aneuploidy in the remaining 3.1% [25].
In a phase II study by Arrieta et al., the levels of the HGF protein were analyzed in 66 patients with either EGFR mutated or EGFR wild type lung adenocarcinoma. All patients received treatment with the EGFR TKI afatinib after disease progression on first-line platinum-based chemotherapy. Patients with reduction of HGF levels after treatment with afatinib had a significantly longer PFS, OS, and objective response rate (ORR) compared to patients with higher levels of HGF after treatment with afatinib. These differences were most prominent among patients with EGFR mutated disease. The authors of the study suggest that HGF has a direct role in acquired resistance to EGFR TKIs, making HGF an interesting target in this field of research [1].
In a different phase II study by Matsumoto et al., HGF levels were measured and MET mutational status was determined in 47 patients with EGFR wild type NSCLC treated with erlotinib. Tumors expressing HGF had a poor response to erlotinib, and the patients had a shorter median PFS compared to the HGF negative study population. MET mutational status did not impact PFS or the response to erlotinib in this clinical trial [19].

4.3. Targeted Therapies

Sixteen of the papers included in this systematic review are studies investigating targeted therapies for the MET gene. The different MET TKIs evaluated in the clinical trials are presented in Table 3. The most common MET TKIs were onartuzumab and crizotinib, occurring in five and four of the included articles each.
In a phase II study conducted by Hirsch et al., treatment with the combination of onartuzumab, paclitaxel, and carboplatin/cisplatin was compared to treatment with only paclitaxel and carboplatin/cisplatin in 106 NSCLC patients. The included patients were all EGFR wild type, with or without mutations in the MET gene. The results showed no significant benefit of added treatment with onartuzumab, regardless of MET mutational status, with similar PFS (5 months in the onartuzumab group and 5.2 in the placebo group) and OS (10.8 months vs. 7.9 months) in the two groups. In this study, MET mutational status was evaluated by immunohistochemistry, with IHC3+ and IHC2+ considered MET positive, and IHC1+ and IHC0 considered MET negative [17].
Similar results were found in a phase II study conducted by Wakelee et al., including 259 NSCLC patients in two cohorts. Cohort 1 tested the addition of onartuzumab vs. placebo to treatment with bevacizumab, carboplatin/cisplatin, and paclitaxel. In cohort 2, patients received either onartuzumab or placebo in addition to carboplatin/cisplatin and pemetrexed. The subjects had varying mutational statuses regarding both EGFR and MET. MET positivity was defined as IHC3+ or IHC2+. In cohort 1, the overall median PFS was 5.0 months in the onartuzumab group compared to 6.8 months in the placebo group. In a MET+ subgroup, median PFS was 4.8 months and median OS was 9.9 months in the onartuzumab arm compared to 6.9 months and 16.5 months in the placebo arm. Cohort 2 revealed similar results, with a median PFS of 5.1 months in the placebo group compared to 4.9 months in the onartuzumab group. Median OS was 13.7 months in the placebo treated group and 8.5 months in the onartuzumab treated group. In the MET+ subgroup of cohort 2, median PFS was 5.0 in for both onartuzumab and placebo while median OS was marginally longer in the onartuzumab arm (8.0 months) compared to the placebo arm (7.6 months) [32].
In another phase III study evaluating onartuzumab, 636 NSCLC patients with varying EGFR and MET mutational statuses were included and treated with either erlotinib and onartuzumab or erlotinib and placebo. In this study, high doses of onartuzumab were associated with a longer median PFS compared to lower doses of erlotinib or placebo (high dose PFS = 4.37; low dose PFS = 2.5 months; placebo PFS = 2.5 months). No significant differences were found in OS regardless of MET mutational status [14].
In contrast to this, a phase II study by Spigel et al. performed on NSCLC patients found that both median PFS and OS were significantly longer in a MET positive subgroup (defined as MET IHC3+ or IHC2+) treated with onartuzumab and erlotinib compared to a subgroup treated with erlotinib and placebo (dual treatment PFS = 2.9 months, OS = 12.6 months; single treatment PFS = 1.5 months, OS = 3.8 months). All 136 included participants were EGFR wild type but had varying MET mutational statuses. Interestingly, the patients without any mutations in the MET gene had earlier progression when treated with the combination of onartuzumab and erlotinib compared to erlotinib and placebo [6].
A third phase II study, titled the global METLung study (OAM4971g), presented results on treatment with onartuzumab and erlotinib without comparing the effects to a control group. This study included 61 patients with MET and EGFR mutation positive NSCLC but was terminated early due to lack of efficacy of onartuzumab. Patients with MET IHC3+ or IHC2+ as well as a total number of MET genes in 20 cancer cells ≥ 90 as determined by a gene amplification assay were considered MET positive. The preliminary results showed a median PFS of 8.5 months, a median OS of 15.6 months, and an overall response rate of 68.9% in the patients treated with onartuzumab and erlotinib [15].
In a phase II clinical trial, Landi et al. evaluated treatment with the targeted MET inhibitor crizotinib in an EGFR wild type NSCLC population of 26 patients with either MET amplification or METex14 mutation. The median PFS was 4.4 months, median OS was 5.4 months, and the ORR was 27.0%. MET amplification was defined as a MET/centromere 7 (CEP7) ratio > 2.2 [18]. Jänne et al. treated 67 NSCLC patients with mixed EGFR mutational statuses with crizotinib and dacomitinib in a phase I clinical trial. The included patients had experienced progression on first-line treatment with either chemotherapy or another targeted therapy and were included in either an escalation phase cohort or in an expansion phase cohort. The median PFS was 3.0 months in the escalation cohort, with 61.0% of these patients having stable disease during the treatment. In the expansion cohort, the median PFS was 2.1 months and 32.0% of the patients had stable disease during treatment. No association was seen between overexpression of MET and PFS. MET positivity was defined as IHC3+, IHC2+, or MET GCN ≥ 2.1 [21].
In another phase II clinical trial conducted by Moro-Sibilot et al., 53 NSCLC patients with varying EGFR mutational statuses were treated with crizotinib. The study included two cohorts with aberrant MET expression, one with MET GCN ≥ 6 (n = 25) and one with positivity for MET exon skipping mutations in exon 14 or 16–19 (n = 28). In the MET amplified cohort (GCN ≥ 6), the ORR was 16.0%, the median PFS was 3.2 months, and the median OS was 7.7 months. In the MET mutated cohort, the OS was longer while the PFS and ORR were inferior (ORR = 10.7%, median PFS = 2.4 months, median OS = 8.1 months) [29].
In a phase I study by Ou et al., crizotinib was evaluated in combination with erlotinib on 26 NSCLC patients with previous progression of disease on one or two prior treatments with chemotherapy. The patients’ EGFR and MET mutational statuses were not reported in this article. Only 20 of the 26 included patients were evaluated for response to treatment. Of these, two had partial response, eight had stable disease, and ten had progressive disease as defined by the RECIST version 1.1 guidelines [24].
One study evaluated the MET targeted drug savolitinib in combination with osimertinib in a phase Ib clinical trial. A total of 180 patients with EGFR mutation positive and MET amplified NSCLC received treatment with savolitinib and osimertinib after previous treatment with one, two, or three different EGFR TKIs. In this study, MET amplification was defined as MET GCN ≥ 5, MET/CEP7 ratio ≥ 2, MET IHC3+, or MET expression in ≥20.0% of tumor cells as determined by NGS. Patients were stratified into one of two cohorts according to type of EGFR mutation and number of prior EGFR TKI treatments. Only 161 patients were eligible for final evaluation of treatment effect. Of these, 89 patients had a partial response according to the RECIST version 1.1 guidelines. One of the two cohorts included 138 patients and had a median PFS of 7.6 months, while the other cohort, including 23 patients, had a median PFS of 9.1 months [22].
In a phase Ib/II trial by Wu et al., standard platinum doublet chemotherapy was compared to treatment with the targeted drugs tepotinib and gefitinib. In this clinical trial, 55 patients were included, all with NSCLC, positive for EGFR mutation, and MET overexpression or amplification. MET overexpression was defined as IHC2+ or IHC3+, and amplification was defined as GCN ≥ 5. All patients included in the phase II part of the trial had acquired resistance to other first or second-generation EGFR TKIs. There were no significant differences in median PFS or OS when comparing chemotherapy to targeted therapy. Subgroup analyses were carried out on groups of patients harboring either MET amplification or high MET overexpression defined as IHC3+. In the MET amplified group, both PFS and OS were significantly longer in the targeted therapy treated group compared to the chemotherapy treated group (targeted therapy median PFS = 16.6, chemotherapy median PFS = 4.2; targeted therapy OS = 37.7, chemotherapy OS = 13.1). In the MET IHC3+ group, the median PFS was 8.3 months, and the OS was 37.3 months compared to a PFS of 4.4 months and an OS of 17.9 in the chemotherapy group [23].
A phase I study evaluating capmatinib in NSCLC patients with MET amplification or MET overexpression showed that patients with a high MET GCN or METex14 mutations may benefit from treatment with MET inhibitors. Of 55 enrolled patients treated with capmatinib, 26 were included in a dose expansion group with varying EGFR mutational statuses and either MET overexpression or amplification. The remaining 29 patients were all EGFR wild type and had high overexpression of MET, defined as IHC3+. Complete response was observed in one of the 55 patients and partial response was seen in ten patients. The median PFS was 3.7 months for the entire cohort, whereas the MET IHC3+ group had a median PFS of 5.1 months. An even greater PFS was seen among patients with MET GCN ≥ 6 (median PFS of 9.3 months). Furthermore, in four patients with METex14 mutations, a reduction in the tumor burden between 14.0 and 83.0% could be seen. This trial was terminated early due to disease progression and frequent adverse events [20].
A phase II trial by Seto et al. tested the effect of capmatinib on 45 EGFR wild type NSCLC patients divided into several cohorts depending on MET mutational burden and prior systemic anti-cancer treatment. Aberrant MET expression was defined as the presence of the METex14 mutation or amplification of the MET gene. The results showed that treatment with capmatinib as a second or third-line option in METex14 positive subjects (n = 11) yielded an overall response rate of 36.4%. The overall response rate in a cohort of MET amplified patients with GCN ≥ 10 (n = 11) was found to be 45.5%. In contrast to this, in a cohort with MET GCN ≥ 4 but <6 (n = 10), the overall response rate was only 10.0%. The remainder of the patients were further subdivided into considerably smaller cohorts with inconclusive results [31].
Yoshioka et al. conducted a phase III clinical trial comparing treatment with tivantinib and erlotinib to treatment with erlotinib and placebo in 303 EGFR wild type NSCLC patients. All included patients had received one or two prior treatments, one of them being platinum-based chemotherapy. The patients had varying MET mutational statuses, including high and low expression of MET as well as elevated and normal MET GCN. The authors found no significant difference in the median OS when comparing the dual treatment group to the erlotinib plus placebo group. In the patients treated with both tivantinib and erlotinib, a significantly longer median PFS could be seen compared to the placebo group (dual treatment PFS = 2.9 months, placebo PFS = 2.0 months). Furthermore, high expression of HGF, defined as an H-score ≥ 200 as measured by IHC, was associated with a significant benefit in OS in the tivantinib plus erlotinib group compared to the placebo group. This clinical trial was terminated early due to increased incidence of interstitial lung disease, with 14 cases in the dual treatment group and 6 cases in the placebo group [5].
This is coherent with the results of a phase III study conducted by Scagliotti et al., which also compared erlotinib plus tivantinib to erlotinib plus placebo in 109 EGFR mutated NSCLC patients with varying MET mutational statuses. Here the median PFS was 13.0 months and the median OS was 25.5 months in the dual treatment group compared to a median PFS of 7.5 months and a median OS of 20.3 months in the erlotinib plus placebo group [30].
In a phase II clinical trial, Neal et al. assessed treatment with cabozantinib in EGFR wild type NSCLC patients with varying MET mutational statuses. This study comprised three treatment arms, one with both erlotinib and cabozantinib, one with only cabozantinib, and one with only erlotinib. All patients had previously been treated with one or two other therapeutic agents prior to enrolment in this trial. The median PFS was significantly longer in the erlotinib plus cabozantinib arm (median PFS = 4.7 months) and in the cabozantinib arm (median PFS = 4.3 months) compared to the erlotinib arm (median PFS = 1.8 months). The median OS was also longer in the erlotinib plus cabozantinib arm (median OS = 13.3 months) compared to the erlotinib arm (median OS = 5.1 months). The median OS in the cabozantinib arm was 9.2 months and was not significantly longer compared to the erlotinib arm. In this clinical trial, no association was found between MET IHC status and median PFS in the patient groups receiving treatment with cabozantinib or erlotinib plus cabozantinib [2].
In summary, many of the targeted drugs mentioned in this review show promising results that encourage further research. Onartuzumab, as an added treatment to erlotinib, has been shown in several trials to lead to a longer PFS and OS in NSCLC patients, compared to treatment with placebo and erlotinib. One clinical trial showed a reversed effect of onartuzumab as an added treatment to chemotherapy, as compared to placebo, with a shortened PFS and OS among the patients treated with onartuzumab. Crizotinib alone has been proven to greater increase OS among NSCLC patients harboring MET mutations compared to patients with MET amplified cancer genotypes. Crizotinib, as an addition to the EGFR TKIs dacomitinib or erlotinib, has resulted in a greater fraction of patients with stable disease. Treatment with savolitinib in combination with osimertinib has led to a fraction of patients with partial response of more than 55% in cohorts of NSCLC patients with previous progression of disease on EGFR TKIs. Tepotinib and gefitinib, as a treatment for NSCLC patients harboring high overexpression or amplification of MET, has been shown to prolong both the PFS and OS compared to treatment with chemotherapy. Similarly, in NSCLC patients with a high MET GCN or high levels of overexpression of MET, treatment with capmatinib has prolonged PFS and led to a reduced tumor burden and higher overall response rates compared to cohorts with lower grades of overexpression or amplification. The addition of tivatinib to treatment with erlotinib has been proven to be effective and has led to increases in PFS and OS compared to placebo. Lastly, use of cabozantinib alone or in combination with erlotinib has led to significantly longer PFS compared to treatment with erlotinib alone. For an overview of the results, see Figure A3 (Appendix C).

5. Discussion

Lung cancer continues to be the leading cause of global annual cancer-related mortality, and the need for more efficient therapies is evident. Targeted therapies are a promising area of research, offering new possibilities for inhibiting genetic alterations that are involved in driving tumorigenesis, but to this day, there are no known clinically useful biomarkers [34,35,36,37]. Lung cancer patients harboring alterations in the MET gene detected in circulating DNA in blood or tumor tissue have been shown to have poorer outcomes.
In a study by Andreasson et al., the authors were able to show that the MET protein could be found in exhaled breath as well as in blood plasma, and that the expression of the protein diminished after surgical removal of the lung cancer, underlining the proteins role in lung cancer tumorigenesis [34].
There is a pressing need for novel and more efficient therapies that can target the MET signaling pathway, but for this to be possible, the understanding of how abundant certain genetic alterations are, and how they affect and drive lung cancer development, needs to be further deepened. Therefore, this systematic review aimed to summarize and enlighten the current status of genetic mapping and prevalence of different alterations in the MET gene in NSCLC patients, as well as existing and pipe-line targeted therapies towards this protein.
The articles reviewed in this paper reported that MET mutations were found in 7.6–11.0% of lung cancer patients and that amplification was found in 3.9–22.0% of cases [3,11]. One study reported that genetic alterations in the MET gene were present in 15.0% of the lung cancer cases analyzed [25]. These numbers are in line with previously published data [38,39,40]. One study presented results on prevalence, which differed significantly from the rest of the included papers, with a reported frequency of aberrant MET expression of 45.0% [28]. The reported prevalence of aberrant MET expression varies in different studies. This may, in part, be due to the fact that different studies analyze samples from different genotypes of lung cancer, with differing characteristics, using different quantification methods and definitions. Some of the articles included in this systematic review analyzed the prevalence of aberrant MET signaling in EGFR mutated patients, while other studies do not specify EGFR mutational status [3,11,25]. It has previously been shown that alterations in the MET gene are a known mechanism of resistance to EGFR TKIs; therefore, considering the impact of previous treatments on genetic analyses results may offer valuable insights for included patients [22].
The varying patient characteristics in the included studies make it difficult to predict any specific groups of the population that are at increased risk of developing NSCLC with MET mutated genotypes. Some trials do not specify MET mutational status or present their data on patient characteristics in cohorts of patients with varying MET mutational statuses [6,21,24]. However, some trends can be seen among the studies presenting results on patient characteristics in cohorts with specified MET mutations [6,15,18,20,22,23,29,31]. The median age among patients harboring MET mutations ranges from 56.0 to 72.0 years. MET mutations appear equally among both genders, with the greatest difference being reported by a single study being 32% men and 68% women [29]. Cohorts with MET positive patients reported smoking history as “never smokers” ranging from 19 to 72%, compared to cohorts consisting of MET negative patients, with the lowest reported percentage of never smokers being 3.0% in the study by Spigel et al. [6]. This could be indicative of a trend with higher fractions of MET positive NSCLC patients being never smokers. However, more research that is more directly aimed at this research question needs to be conducted to verify these results.
Five of the included studies evaluated the MET TKI onartuzumab. Spigel et al. found a significantly longer OS and PFS in MET positive patients receiving treatment with both onartuzumab and erlotinib compared to patients receiving erlotinib alone. Interestingly, they also found that the MET negative patients suffered earlier progression of disease and increased mortality when treatment with onartuzumab was added compared to treatment with placebo alone [6]. This is in line with the findings of Han et al., who showed a significantly longer PFS among patients treated with high doses of onartuzumab together with erlotinib compared to single treatment with erlotinib or erlotinib plus lower doses of onartuzumab [14]. Kishi et al. presented similar results regarding OS to what Spigel et al. presented; however, there was a remarkable difference in PFS between the two studies (Kishi et al. PFS = 8.5 months, Spigel et al. PFS = 2.9 months) [6,15]. Hirsch et al. and Wakelee et al. were not able to show a longer OS or PFS with the addition of onartuzumab to standard chemotherapy in lung cancer patients with varying MET mutational statuses [17]. MET has previously been suggested to act as both a suppressor and an oncogene, which is in line with the presented results [6]. These findings underline the importance of genetic testing upon lung cancer diagnosis in order to customize targeted therapies and postpone or prevent progression caused by treatment with ill-suited therapies.
Two separate trials investigated the effect of dual treatment with tivantinib and erlotinib compared to single treatment with erlotinib. They both found a prolongation in the median PFS in the dual treatment group compared to treatment with erlotinib alone, although to very differing extents. No difference was seen in the median OS [5,30]. One study included only EGFR wild type subjects while the other had only EGFR mutated subjects. The variation in EGFR mutational status is likely a contributing factor to the varying results of the studies included in this review. Some clinical trials included only EGFR wild type lung cancers, others included only EGFR positive cases or a combination of wild type and positive cases. Two studies evaluated the effect of additional treatment with onartuzumab to erlotinib in patients with varying EGFR mutational statuses [6,14]. As erlotinib is an EGFR TKI, it seems likely that the patients with an EGFR positive cancer genotype would benefit more from treatment with erlotinib than the patients with an EGFR wild type genotype, making it difficult to evaluate the actual effect of the added onartuzumab or capmatinib treatment. Therefore, in these studies, the effect of added treatment with onartuzumab or capmatinib should be interpreted carefully and further research in larger clinical trials is needed to determine the effect.
Multiple studies performed on different MET TKIs have presented varying results on PFS, ranging from 2.1 to 9.1 months, without comparisons to control groups [18,20,21,22,29]. Interestingly, two of these articles showed that treatment with the targeted drug crizotinib had a greater effect in a ROS1 translocated NSCLC cohort compared to MET altered cohorts [18,29]. Other studies evaluating crizotinib did not report ROS1 mutational status, rendering the results more difficult to interpret [21,24]. Neal et al. reported that treatment with cabozantinib and erlotinib and single treatment with cabozantinib was superior to treatment with erlotinib alone in lung cancer patients with EGFR wild type lung cancer. These results support the statement that patients with EGFR wild type lung cancer may not respond to treatment with EGFR TKIs alone. In the clinical trial conducted by Neal et al., the authors did not find any association between MET IHC+ status and prolonged PFS in the treatment groups receiving cabozantinib. It is not possible to draw any conclusions regarding the association between prolonged time to progression and death, and inhibition of the MET receptor specifically, as cabozantinib has multiple gene targets [2,22]. Several clinical trials present similar results, indicating that patients with a high mutational burden in the MET gene (defined as high overexpression, high GCN gain, and high IHC+ status) benefit more from treatment with MET TKIs compared to patients with a lower mutational burden [5,20,23,31]. This further emphasizes the importance of thorough genetic mapping of cancer patients prior to treatment.
An important aspect in trials testing novel drugs is the fact that the included cohorts differ greatly in cancer genotype [6,14]. This puts the reliability and comparability of the results into question. Another aspect is the fact that the drugs presented in these trials typically show a gain in PFS and OS months at best [5,6,14,17]. This needs to be put in relation with the potential side effects and adverse events that the patients experience during treatment [5,20]. MET TKIs are still considered experimental, and treatment with targeted MET inhibitors is currently a second, third, or even fourth-line treatment option. It is known that cancers generally harbor fewer driver mutations in the beginning of tumorigenesis compared to the later stages of disease and so it is possible that earlier implementation of MET inhibitors could provide a greater effect on PFS and OS [41,42]. However, the timing of treatment will need to be investigated in further clinical trials.
The different cancer genotypes also contribute to potential bias in reporting more or less promising pipe-line drugs. However, there are several interesting ongoing clinical trials evaluating different pipe-line drugs on more targeted patient groups harboring different MET alterations (Table 4) [43]. Two of these ongoing trials have already presented results from their studies. The NCT02544633 trial tested the MET inhibitor MGCD265 on patients with NSCLC and MET activating mutations or MET amplifications. The patients were divided into four study arms, one with MET activating mutations in tumor tissue, one with MET activating mutations in circulating tumor DNA in the bloodstream, one with MET amplification in tumor tissue, and one with MET amplification in circulating tumor DNA. Interestingly, in the case of MET amplification, the patients with MET amplification identified in circulating tumor DNA had a poorer PFS (2.76 months) and OS (4.08 months) compared to the patients with MET amplification identified in tumor tissue (PFS 4.85 months, OS 7.04 months). The patients with MET activating mutations in tumor tissue had a longer OS of 16.32 months compared to all other study arms, but not PFS (3.95 months). The last treatment group with MET activating mutations in circulating tumor DNA had a similar PFS to the other three study arms (3.39 months). No results were available regarding OS in this fourth study arm [44]. This data is similar to results from earlier studies on MET TKIs presented in this systematic review. A possible explanation to why the patients with MET amplification detected in circulating tumor DNA had a poorer outcome is the greater tumor burden associated with circulating tumor cells compared to that of a localized tumor.
One other study had available preliminary results at the time of writing this systematic review (trial ID: NCT02648724). In this phase I/II study, the monoclonal anti MET antibody mixture entitled Sym015 was investigated. The study included 57 patients divided into three different treatment arms, one with KRAS wild type patients with MET amplifications, one with MET amplified NSCLC, and one where the patients harbored METex14 deletions. The outcome measure presented was the ORR and the results showed that the KRAS wild type cohort had 0.0% ORR while the MET amplified and the METex14 deletion cohort both had an ORR of 25.0% [45]. A vast difference between the already existing and published clinical trials and the ongoing ones is the greatly improved consistence in genotypes included in the newer trials. The majority of the current trials on pipe-line targeted therapies consist of cohorts of patients that are all genetically altered in the MET gene rather than cohorts of mixed cancer genotypes. This will make future results more easily interpreted and clinically useful.

6. Strengths and Limitations

The most apparent limitation of the articles included in this review is the lack of control groups in most of the phase I trials, leading to difficulties in interpreting the results. Furthermore, some articles have a low number of included patients and overall, the number of participants varies greatly. Since the included studies have a high grade of variation in patient characteristics, such as age, gender, and smoking history, the comparison between the studies’ results need to be interpreted with some caution. As the targeted therapies are tested on different patient categories, with varying cancer genotypes, it is difficult to compare the presented results and draw conclusions on the efficacy of the investigated drugs. On the same note, the definition of the criteria for what is considered aberrant MET expression varies between the included trials, with some research groups applying IHC, some performing NGS, and some using different methods. A strength of this review is the variation in location of the included trials, leading to more generalizable and widely applicable conclusions. The exclusion criterion of a publication year prior to 2013 might be considered a limitation; however, this also ensured that only current results were included in this systematic review.

7. Conclusions

It remains difficult to compare different targeted therapies and to draw conclusions regarding their potential place in the future treatment panorama. The inter and intra study variation in cohort composition and included cancer genotypes is large. Onartuzumab has shown prolonged PFS and OS among MET positive NSCLC patients but no convincing results in cohorts with mixed MET mutational statuses. The studies on crizotinib lacked control groups for comparison of the outcome measures. Many ongoing trials on pipe-line targeted therapies exist, which are investigating anti-MET agents on clearly defined cohorts of patients with aberrant MET expression. This systematic review summarized the current status of publications on the MET gene’s implications in lung cancer development and the status of existing and up and coming targeted therapy options. More research is needed and should be encouraged to fully understand how, when, and to whom these drugs should be recommended in order to improve patient outcomes.

Author Contributions

Conceptualization, E.B., F.S. and S.L.; methodology, E.B., F.S. and S.L.; formal analysis, E.B. and F.S.; investigation, E.B. and F.S.; resources, S.L.; data curation, E.B. and F.S.; writing—original draft preparation, E.B., F.S. and F.O.; writing—review and editing, E.B., F.S., F.O. and S.L.; visualization, E.B. and F.S.; supervision, F.O. and S.L.; project administration, S.L.; funding acquisition, S.L. All authors have read and agreed to the published version of the manuscript.

Funding

The study was funded by the Swedish Cancer Foundation and the Sjögren Foundation. The APC was funded by Lund University.

Conflicts of Interest

The authors declare no conflict of interest.

Appendix A

Search Queries

Search query 1 PubMed (n = 183):
(((lung cancer[MeSH Terms]) OR (lung cancer)) AND ((((((“met”[All Fields]) OR (“mesenchymal epithelial transition factor”[All Fields])) OR (hgfr)) OR (hepatocyte growth factor receptor)) OR (hgf receptor[MeSH Terms])) OR (hepatocyte growth factor receptor[MeSH Terms]))) AND (((((“prognosis”[All Fields]) OR (“mortality”[All Fields])) OR (“morbidity”[All Fields])) OR (“diagnosis”[All Fields])) OR (morbidity[MeSH Terms])) AND (clinicaltrial[Filter] OR randomizedcontrolledtrial[Filter])
Search query 2 PubMed (n = 157):
((((lung cancer[MeSH Terms]) OR (lung cancer)) AND ((((((“met”[All Fields]) OR (“mesenchymal epithelial transition factor”[All Fields])) OR (hgfr)) OR (hepatocyte growth factor receptor)) OR (hgf receptor[MeSH Terms])) OR (hepatocyte growth factor receptor[MeSH Terms]))) AND ((((((targeted therapy) OR (cancer therapy)) OR (targeted treatment)) OR (molecular targeted therapies[MeSH Terms])) OR (drug targeting[MeSH Terms])) OR (cancer treatment protocol[MeSH Terms]))) AND (((((“prognosis”[All Fields]) OR (“mortality”[All Fields])) OR (“morbidity”[All Fields])) OR (“diagnosis”[All Fields])) OR (morbidity[MeSH Terms])) AND (clinicaltrial[Filter] OR randomizedcontrolledtrial[Filter])
Search query 1 Embase (n = 384):
(‘lung cancer’ OR ‘lung cancer’/exp) AND (‘met’/exp OR met OR ‘mesenchymal epithelial transition factor’ OR hgfr OR ‘scatter factor receptor’) AND (‘prognosis’/exp OR prognosis OR mortality OR morbidity OR diagnosis) AND ([controlled clinical trial]/lim OR [randomized controlled trial]/lim)
Search query 2 Embase (n = 62):
(‘lung cancer’ OR ‘lung cancer’/exp) AND (‘met’/exp OR met OR ‘mesenchymal epithelial transition factor’ OR hgfr OR ‘scatter factor receptor’) AND (‘prognosis’/exp OR prognosis OR mortality OR morbidity OR diagnosis) AND (‘molecularly targeted therapy’/exp OR ‘molecularly targeted therapy’ OR ‘cancer therapy’ OR ‘antineoplastic protocol’ OR ‘drug targeting’) AND ([controlled clinical trial]/lim OR [randomized controlled trial]/lim)

Appendix B

Table A1. Full PRISMA 2020 Checklist [27]. N/A: not applicable.
Table A1. Full PRISMA 2020 Checklist [27]. N/A: not applicable.
Section and Topic Item #Checklist Item Location Where Item Is Reported
TITLE
Title 1Identify the report as a systematic review.Page 1
ABSTRACT
Abstract 2See the PRISMA 2020 for Abstracts checklist.Page 1, paragraph 2
INTRODUCTION
Rationale 3Describe the rationale for the review in the context of existing knowledge.Page 2, paragraph 2
Objectives 4Provide an explicit statement of the objective(s) or question(s) the review addresses.Page 4, paragraph 2
METHODS
Eligibility criteria 5Specify the inclusion and exclusion criteria for the review and how studies were grouped for the syntheses.Page 5, paragraph 2
Information sources 6Specify all databases, registers, websites, organisations, reference lists and other sources searched or consulted to identify studies. Specify the date when each source was last searched or consulted.Page 5, paragraph 1
Search strategy7Present the full search strategies for all databases, registers and websites, including any filters and limits used.Page 22
Selection process8Specify the methods used to decide whether a study met the inclusion criteria of the review, including how many reviewers screened each record and each report retrieved, whether they worked independently, and if applicable, details of automation tools used in the process.Page 5, paragraph 2
Data collection process 9Specify the methods used to collect data from reports, including how many reviewers collected data from each report, whether they worked independently, any processes for obtaining or confirming data from study investigators, and if applicable, details of automation tools used in the process.Page 5, paragraph 2
Data items 10aList and define all outcomes for which data were sought. Specify whether all results that were compatible with each outcome domain in each study were sought (e.g., for all measures, time points, analyses), and if not, the methods used to decide which results to collect.Page 6, paragraph 1
10bList and define all other variables for which data were sought (e.g., participant and intervention characteristics, funding sources). Describe any assumptions made about any missing or unclear information.Page 6, paragraph 1
Study risk of bias assessment11Specify the methods used to assess risk of bias in the included studies, including details of the tool(s) used, how many reviewers assessed each study and whether they worked independently, and if applicable, details of automation tools used in the process.Page 11, paragraph 1
Effect measures 12Specify for each outcome the effect measure(s) (e.g., risk ratio, mean difference) used in the synthesis or presentation of results.N/A
Synthesis methods13aDescribe the processes used to decide which studies were eligible for each synthesis (e.g., tabulating the study intervention characteristics and comparing against the planned groups for each synthesis (item #5)).Page 8, Table 3
13bDescribe any methods required to prepare the data for presentation or synthesis, such as handling of missing summary statistics, or data conversions.N/A
13cDescribe any methods used to tabulate or visually display results of individual studies and syntheses.Page 8, Table 3
13dDescribe any methods used to synthesize results and provide a rationale for the choice(s). If meta-analysis was performed, describe the model(s), method(s) to identify the presence and extent of statistical heterogeneity, and software package(s) used.N/A
13eDescribe any methods used to explore possible causes of heterogeneity among study results (e.g., subgroup analysis, meta-regression).N/A
13fDescribe any sensitivity analyses conducted to assess robustness of the synthesized results.N/A
Reporting bias assessment14Describe any methods used to assess risk of bias due to missing results in a synthesis (arising from reporting biases).Page 11, paragraph 1
Certainty assessment15Describe any methods used to assess certainty (or confidence) in the body of evidence for an outcome.N/A
RESULTS
Study selection 16aDescribe the results of the search and selection process, from the number of records identified in the search to the number of studies included in the review, ideally using a flow diagram.Page 6, Figure 3
16bCite studies that might appear to meet the inclusion criteria, but which were excluded, and explain why they were excluded.N/A
Study characteristics 17Cite each included study and present its characteristics.Page 11, paragraph 4
Risk of bias in studies 18Present assessments of risk of bias for each included study.Pages 27–28
Results of individual studies 19For all outcomes, present, for each study: (a) summary statistics for each group (where appropriate) and (b) an effect estimate and its precision (e.g., confidence/credible interval), ideally using structured tables or plots.Pages 29–30
Results of syntheses20aFor each synthesis, briefly summarise the characteristics and risk of bias among contributing studies.Page 21, paragraph 2
20bPresent results of all statistical syntheses conducted. If meta-analysis was done, present for each the summary estimate and its precision (e.g., confidence/credible interval) and measures of statistical heterogeneity. If comparing groups, describe the direction of the effect.N/A
20cPresent results of all investigations of possible causes of heterogeneity among study results.Page 21, paragraph 2
20dPresent results of all sensitivity analyses conducted to assess the robustness of the synthesized results.N/A
Reporting biases21Present assessments of risk of bias due to missing results (arising from reporting biases) for each synthesis assessed.Pages 27–28
Certainty of evidence 22Present assessments of certainty (or confidence) in the body of evidence for each outcome assessed.N/A
DISCUSSION
Discussion 23aProvide a general interpretation of the results in the context of other evidence.Page 18, paragraph 1
23bDiscuss any limitations of the evidence included in the review.Page 21, paragraph 2
23cDiscuss any limitations of the review processes used.Page 21, paragraph 2
23dDiscuss implications of the results for practice, policy, and future research.Page 18, paragraph 3
OTHER INFORMATION
Registration and protocol24aProvide registration information for the review, including register name and registration number, or state that the review was not registered.Page 5, paragraph 1
24bIndicate where the review protocol can be accessed, or state that a protocol was not prepared.Page 5, paragraph 1
24cDescribe and explain any amendments to information provided at registration or in the protocol.N/A
Support25Describe sources of financial or non-financial support for the review, and the role of the funders or sponsors in the review.Page 22
Competing interests26Declare any competing interests of review authors.Page 22
Availability of data, code and other materials27Report which of the following are publicly available and where they can be found: template data collection forms; data extracted from included studies; data used for all analyses; analytic code; any other materials used in the review.Page 22
Table A2. PRISMA Abstract Checklist [27].
Table A2. PRISMA Abstract Checklist [27].
TopicNo.ItemReported?
TITLE
Title1Identify the report as a systematic review.Yes
BACKGROUND
Objectives2Provide an explicit statement of the main objective(s) or question(s) the review addresses.Yes
METHODS
Eligibility criteria3Specify the inclusion and exclusion criteria for the review.No
Information sources4Specify the information sources (e.g., databases, registers) used to identify studies and the date when each was last searched. Yes
Risk of bias5Specify the methods used to assess risk of bias in the included studies.No
Synthesis of results6Specify the methods used to present and synthesize results. No
RESULTS
Included studies7Give the total number of included studies and participants and summarise relevant characteristics of studies.Yes
Synthesis of results8Present results for main outcomes, preferably indicating the number of included studies and participants for each. If meta-analysis was done, report the summary estimate and confidence/credible interval. If comparing groups, indicate the direction of the effect (i.e., which group is favoured).Yes
DISCUSSION
Limitations of evidence9Provide a brief summary of the limitations of the evidence included in the review (e.g., study risk of bias, inconsistency and imprecision).No
Interpretation10Provide a general interpretation of the results and important implications.Yes
OTHER
Funding11Specify the primary source of funding for the review.No
Registration12Provide the register name and registration number.Yes

Appendix C

Figure A1. Risk of bias plots for all included RCTs in this systematic review, created in the Cochrane Risk of Bias Tool "robvis" [2,5,6,17,23,30,32,33]. (A) Traffic light plot, indicating the risk of bias for each included RCT in each of the five domains. (B) Summary plot, showing the summarized risk of bias in each domain, and an overall risk of bias measurement. D: domain, RCT: randomized controlled trial.
Figure A1. Risk of bias plots for all included RCTs in this systematic review, created in the Cochrane Risk of Bias Tool "robvis" [2,5,6,17,23,30,32,33]. (A) Traffic light plot, indicating the risk of bias for each included RCT in each of the five domains. (B) Summary plot, showing the summarized risk of bias in each domain, and an overall risk of bias measurement. D: domain, RCT: randomized controlled trial.
Cancers 15 03827 g0a1
Figure A2. Risk of bias plots for all included clinical trials in this systematic review, created in the Cochrane Risk of Bias Tool "robvis" [1,3,11,14,15,18,19,20,21,22,24,25,28,29,31,33]. (A): Traffic light plot, indicating the risk of bias for each included clinical trial in each of the seven domains. (B): Summary plot, showing the summarized risk of bias in each domain, and an overall risk of bias measurement. D: domain.
Figure A2. Risk of bias plots for all included clinical trials in this systematic review, created in the Cochrane Risk of Bias Tool "robvis" [1,3,11,14,15,18,19,20,21,22,24,25,28,29,31,33]. (A): Traffic light plot, indicating the risk of bias for each included clinical trial in each of the seven domains. (B): Summary plot, showing the summarized risk of bias in each domain, and an overall risk of bias measurement. D: domain.
Cancers 15 03827 g0a2
Figure A3. Forest plots presenting the median PFS and median OS with 95% confidence intervals (CI) from the included articles in this systematic review presenting results on treatment with a MET TKI. Results that are specific to a MET+ subgroup are presented separately from the overall cohort results [2,5,6,14,15,17,18,20,21,22,23,29,30,32]. (A) Median PFS with 95% CIs. (B) Median OS with 95% CIs.
Figure A3. Forest plots presenting the median PFS and median OS with 95% confidence intervals (CI) from the included articles in this systematic review presenting results on treatment with a MET TKI. Results that are specific to a MET+ subgroup are presented separately from the overall cohort results [2,5,6,14,15,17,18,20,21,22,23,29,30,32]. (A) Median PFS with 95% CIs. (B) Median OS with 95% CIs.
Cancers 15 03827 g0a3aCancers 15 03827 g0a3b
Table A3. Epidemiology reported by the included studies. Median age, percentage of participants that are male gender, and smoking history, presented as percentage of participants with a history of no smoking, of the included patients in the different studies and their subgroups. EGFR: epidermal growth factor receptor, WT: wild type, MET: mesenchymal epithelial transition factor, NS: not specified.
Table A3. Epidemiology reported by the included studies. Median age, percentage of participants that are male gender, and smoking history, presented as percentage of participants with a history of no smoking, of the included patients in the different studies and their subgroups. EGFR: epidermal growth factor receptor, WT: wild type, MET: mesenchymal epithelial transition factor, NS: not specified.
Mutational StatusMedian Age (Years)Male Gender (%)History of No Smoking (%)
Arrieta et al., 2016 [1]EGFR+/WT60.133.3%39.4%
Han et al., 2017 [14]EGFR+/WT, MET+/−Treatment: 63.0
Placebo: 63.0
Treatment: 56.0%
Placebo: 57.0%
Treatment: 18.5%
Placebo: 15.8%
Helman et al., 2018 [25]EGFR+61.028.6%NS
Hirsch et al., 2017 [17]MET+/−Treatment: 68.0
Placebo: 66.0
Treatment: 70.9%
Placebo: 74.1%
Treatment: 3.6%
Placebo: 5.6%
Jänne et al., 2016 [21]EGFR+/WT59.539.0%51.0%
Kishi et al., 2019 [15]EGFR+
MET+
67.043%NS
Landi et al., 2019 [18]EGFR WT
MET+
56.065%23%
Matsumoto et al., 2014 [19]EGFR WT64.019.7%89.1%
Moro-Sibilot et al., 2019 [29]EGFR+/WT
MET+/−
MET amplified: 59.0
MET mutated: 72.0
MET amplified: 56.0%
MET mutated: 32.0%
MET amplified: 24.0%
MET mutation: 48.0%
Neal et al., 2016 [2]EGFR WT65.345.0%15%
Okamoto et al., 2014 [3]NS64.077.0%18.0%
Ou et al., 2017 [24]NS60.030.0%52.0%
Palmero et al., 2021 [11]NS64.365.0%27.0%
Sacher et al., 2016 [28]EGFR+/WT64.045.0%18.0%
Scagliotti et al., 2018 [30]EGFR+
MET+/−
Treatment: 59.5
Placebo: 65.0
Treatment: 42.9%
Placebo: 47.2%
Treatment: 48.2%
Placebo: 60.4%
Schuler et al., 2020 [20]EGFR WT
MET+
60.060.0%NS
Sequist et al., 2020 [22]EGFR+
MET+
Cohort B: 59.0
Cohort D: 62.0
Cohort B: 59%
Cohort D: 60%
NS
Seto et al., 2021 [31]EGFR WT
MET+
68.066.7%42.2%
Spigel et al., 2013 [6]EGFR+/WT
MET+/−
Treatment MET-: 63.0
Placebo MET-: 61.0
Treatment MET+: 66.0
Placebo MET+: 64.0
Treatment MET-: 65.0%
Placebo MET-: 55.0%
Treatment MET+: 51.0%
Placebo MET+: 65.0%
Treatment MET -: 7.0%
Placebo MET -: 3.0%
Treatment MET+: 20.0%
Placebo MET+: 19.0%
Wakelee et al., 2017 [32]EGFR+/WT
MET+/−
Cohort I treatment: 60.0
Cohort I placebo: 60.5
Cohort II treatment: 66.0
Cohort II placebo: 63.0
Cohort I treatment: 68.1%
Cohort I placebo: 48.6%
Cohort II treatment: 55.9%
Cohort II placebo: 42.6%
Cohort I treatment: 21.7%
Cohort I placebo: 25.7%
Cohort II treatment: 27.1%
Cohort II placebo: 13.1%
Wu et al., 2020 [23]EGFR+
MET+
Phase Ib: 65.5
Phase II treatment: 61.0
Phase II placebo: 58.3
Phase Ib: 44.0%
Phase II treatment: 35.5%
Phase II placebo: 50.0%
Phase Ib: 72.0%
Phase II treatment: 68.0%
Phase II placebo: 67.0%
Yoshioka et al., 2015 [5]EGFR WT
MET+/−
Treatment: 63.0
Placebo: 63.0
Treatment: 66.7%
Placebo: 70.8%
Treatment: 25.5%
Placebo: 26.2%

References

  1. Arrieta, O.; Cruz-Rico, G.; Soto-Perez-de-Celis, E.; Ramírez-Tirado, L.A.; Caballe-Perez, E.; Martínez-Hernández, J.N.; Martinez-Alvarez, I.; Soca-Chafre, G.; Macedo-Pérez, E.O.; Astudillo-de la Vega, H.; et al. Reduction in Hepatocyte Growth Factor Serum Levels is Associated with Improved Prognosis in Advanced Lung Adenocarcinoma Patients Treated with Afatinib: A Phase II Trial. Target Oncol. 2016, 11, 619–629. [Google Scholar] [CrossRef] [PubMed]
  2. Neal, J.W.; Dahlberg, S.E.; Wakelee, H.A.; Aisner, S.C.; Bowden, M.; Huang, Y.; Carbone, D.P.; Gerstner, G.J.; Lerner, R.E.; Rubin, J.L.; et al. Erlotinib, cabozantinib, or erlotinib plus cabozantinib as second-line or third-line treatment of patients with EGFR wild-type advanced non-small-cell lung cancer (ECOG-ACRIN 1512): A randomised, controlled, open-label, multicentre, phase 2 trial. Lancet Oncol. 2016, 17, 1661–1671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Okamoto, I.; Sakai, K.; Morita, S.; Yoshioka, H.; Kaneda, H.; Takeda, K.; Hirashima, T.; Kogure, Y.; Kimura, T.; Takahashi, T.; et al. Multiplex genomic profiling of non-small cell lung cancers from the LETS phase III trial of first-line S-1/carboplatin versus paclitaxel/carboplatin: Results of a West Japan Oncology Group study. Oncotarget 2014, 5, 2293–2304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, 359–386. [Google Scholar] [CrossRef] [PubMed]
  5. Yoshioka, H.; Azuma, K.; Yamamoto, N.; Takahashi, T.; Nishio, M.; Katakami, N.; Ahn, M.J.; Hirashima, T.; Maemondo, M.; Kim, S.W.; et al. A randomized, double-blind, placebo-controlled, phase III trial of erlotinib with or without a c-Met inhibitor tivantinib (ARQ 197) in Asian patients with previously treated stage IIIB/IV nonsquamous nonsmall-cell lung cancer harboring wild-type epidermal growth factor receptor (ATTENTION study). Ann. Oncol. 2015, 26, 2066–2072. [Google Scholar] [PubMed]
  6. Spigel, D.R.; Ervin, T.J.; Ramlau, R.A.; Daniel, D.B.; Goldschmidt, J.H., Jr.; Blumenschein, G.R., Jr.; Krzakowski, M.J.; Robinet, G.; Godbert, B.; Barlesi, F.; et al. Randomized phase II trial of Onartuzumab in combination with erlotinib in patients with advanced non-small-cell lung cancer. J. Clin. Oncol. 2013, 31, 4105–4114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Urvay, S.E.; Yucel, B.; Erdis, E.; Turan, N. Prognostic Factors in Stage III Non-Small-Cell Lung Cancer Patients. Asian. Pac. J. Cancer Prev. 2016, 17, 4693–4697. [Google Scholar]
  8. Önal, Ö.; Koçer, M.; Eroğlu, H.N.; Yilmaz, S.D.; Eroğlu, I.; Karadoğan, D. Survival analysis and factors affecting survival in patients who presented to the medical oncology unit with non-small cell lung cancer. Turk. J. Med. Sci. 2020, 50, 1838–1850. [Google Scholar] [CrossRef]
  9. Shields, M.D.; Marin-Acevedo, J.A.; Pellini, B. Immunotherapy for Advanced Non-Small Cell Lung Cancer: A Decade of Progress; American Society of Clinical Oncology Educational Book: Alexandria, VA, USA, 2021; pp. 105–127. [Google Scholar]
  10. Bodén, E.; Andreasson, J.; Hirdman, G.; Malmsjö, M.; Lindstedt, S. Quantitative Proteomics Indicate Radical Removal of Non-Small Cell Lung Cancer and Predict Outcome. Biomedicines 2022, 10, 2738. [Google Scholar] [CrossRef]
  11. Palmero, R.; Taus, A.; Viteri, S.; Majem, M.; Carcereny, E.; Garde-Noguera, J.; Felip, E.; Nadal, E.; Malfettone, A.; Sampayo, M.; et al. Biomarker Discovery and Outcomes for Comprehensive Cell-Free Circulating Tumor DNA Versus Standard-of-Care Tissue Testing in Advanced Non-Small-Cell Lung Cancer. JCO Precis. Oncol. 2021, 5, 93–102. [Google Scholar] [CrossRef]
  12. Bethune, G.; Bethune, D.; Ridgway, N.; Xu, Z. Epidermal growth factor receptor (EGFR) in lung cancer: An overview and update. J. Thorac. Dis. 2010, 2, 48–51. [Google Scholar] [PubMed]
  13. 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]
  14. Han, K.; Chanu, P.; Jonsson, F.; Winter, H.; Bruno, R.; Jin, J.; Stroh, M. Exposure-Response and Tumor Growth Inhibition Analyses of the Monovalent Anti-c-MET Antibody Onartuzumab (MetMAb) in the Second- and Third-Line Non-Small Cell Lung Cancer. AAPS J. 2017, 19, 527–533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Kishi, K.; Sakai, H.; Seto, T.; Kozuki, T.; Nishio, M.; Imamura, F.; Hiroshi, N.; Miyako, S.; Shintaro, N.; Takashi, T.; et al. First-line onartuzumab plus erlotinib treatment for patients with MET-positive and EGFR mutation-positive non-small-cell lung cancer. Cancer Treat Res. Commun. 2019, 18, 100113. [Google Scholar] [CrossRef] [PubMed]
  16. Organ, S.L.; Tsao, M. S An overview of the c-MET signaling pathway. Ther. Adv. Med. Oncol. 2011, 3, S7–S19. [Google Scholar] [CrossRef] [Green Version]
  17. Hirsch, F.R.; Govindan, R.; Zvirbule, Z.; Braiteh, F.; Rittmeyer, A.; Belda-Iniesta, C.; Isla, D.; Cosgriff, T.; Boyer, M.; Ueda, M.; et al. Efficacy and Safety Results From a Phase II, Placebo-Controlled Study of Onartuzumab Plus First-Line Platinum-Doublet Chemotherapy for Advanced Squamous Cell Non-Small-Cell Lung Cancer. Clin. Lung Cancer 2017, 18, 43–49. [Google Scholar] [CrossRef]
  18. Landi, L.; Chiari, R.; Tiseo, M.; D’Incà, F.; Dazzi, C.; Chella, A.; Delmonte, A.; Bonanno, L.; Giannarelli, 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]
  19. Matsumoto, Y.; Maemondo, M.; Ishii, Y.; Okudera, K.; Demura, Y.; Takamura, K.; Kobayashi, K.; Morikawa, N.; Gemm, A.; Ishimoto, O.; et al. A phase II study of erlotinib monotherapy in pre-treated non-small cell lung cancer without EGFR gene mutation who have never/light smoking history: Re-evaluation of EGFR gene status (NEJ006/TCOG0903). Lung Cancer 2014, 86, 195–200. [Google Scholar] [CrossRef]
  20. Schuler, M.; Berardi, R.; Lim, W.T.; de Jonge, M.; Bauer, T.M.; Azaro, A.; Gottfried, M.; Han, J.-Y.; Lee, D.; 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] [PubMed]
  21. Jänne, P.A.; Shaw, A.T.; Camidge, D.R.; Giaccone, G.; Shreeve, S.M.; Tang, Y.; Goldberg, Z.; Martini, J.-F.; Xu, H.; James, L.P.; et al. Combined Pan-HER and ALK/ROS1/MET Inhibition with Dacomitinib and Crizotinib in Advanced Non-Small Cell Lung Cancer: Results of a Phase I Study. J. Thorac. Oncol. 2016, 11, 737–747. [Google Scholar] [CrossRef]
  22. 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] [PubMed]
  23. 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 1b/2, multicentre, randomised trial. Lancet Respir. Med. 2020, 8, 1132–1143. [Google Scholar] [CrossRef]
  24. Ou, S.I.; Govindan, R.; Eaton, K.D.; Otterson, G.A.; Gutierrez, M.E.; Mita, A.C.; Argiris, A.; Brega, N.M.; Usari, T.; Tan, W.; et al. Phase I Results from a Study of Crizotinib in Combination with Erlotinib in Patients with Advanced Nonsquamous Non-Small Cell Lung Cancer. J. Thorac. Oncol. 2017, 12, 145–151. [Google Scholar] [CrossRef]
  25. Helman, E.; Nguyen, M.; Karlovich, C.A.; Despain, D.; Choquette, A.K.; Spira, A.I.; Yu, H.A.; Camidge, D.R.; Harding, T.C.; Lanman, R.B.; et al. Cell-Free DNA Next-Generation Sequencing Prediction of Response and Resistance to Third-Generation EGFR Inhibitor. Clin. Lung Cancer 2018, 19, 518–530. [Google Scholar] [CrossRef] [Green Version]
  26. Morganti, S.; Tarantino, P.; Ferraro, E.; D’Amico, P.; Duso, B.A.; Curigliano, G. Next Generation Sequencing (NGS): A Revolutionary Technology in Pharmacogenomics and Personalized Medicine in Cancer; Springer International Publishing: Berlin/Heidelberg, Germany, 2019; pp. 9–30. [Google Scholar]
  27. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Jennifer, M.; Elie, A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Syst. Rev. 2021, 88, 105906. [Google Scholar]
  28. Sacher, A.G.; Le, L.W.; Lara-Guerra, H.; Waddell, T.K.; Sakashita, S.; Chen, Z.; Kim, L.; Zhang, T.; Kamel-Reid, S.; Salvarrey, A.; et al. A window of opportunity study of potential tumor and soluble biomarkers of response to preoperative erlotinib in early stage non-small cell lung cancer. Oncotarget 2016, 7, 25632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. 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] [PubMed]
  30. Scagliotti, G.V.; Shuster, D.; Orlov, S.; von Pawel, J.; Shepherd, F.A.; Ross, J.S.; Wang, Q.; Schwartz, B.; Akerley, W. Tivantinib in Combination with Erlotinib versus Erlotinib Alone for EGFR-Mutant NSCLC: An Exploratory Analysis of the Phase 3 MARQUEE Study. J. Thorac. Oncol. 2018, 13, 849–854. [Google Scholar] [CrossRef] [Green Version]
  31. Seto, T.; Ohashi, K.; Sugawara, S.; Nishio, M.; Takeda, M.; Aoe, K.; Moizumi, S.; Nomura, S.; Tajima, T.; Hida, T.; et al. 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] [PubMed]
  32. Wakelee, H.; Zvirbule, Z.; De Braud, F.; Kingsley, C.D.; Mekhail, T.; Lowe, T.; Schütte, W.; Lena, H.; Lawler, W.; Braiteh, F.; et al. Efficacy and Safety of Onartuzumab in Combination With First-Line Bevacizumab- or Pemetrexed-Based Chemotherapy Regimens in Advanced Non-Squamous Non-Small-Cell Lung Cancer. Clin. Lung Cancer 2017, 18, 50–59. [Google Scholar] [CrossRef]
  33. McGuinness, L.A.; Higgins, J.P.T. Risk-of-bias VISualization (robvis): An R package and Shiny web app for visualizing risk-of-bias assessments. Res. Synth. Methods 2021, 12, 55–61. [Google Scholar] [CrossRef] [PubMed]
  34. Andreasson, J.; Bodén, E.; Fakhro, M.; von Wachter, C.; Olm, F.; Malmsjö, M.; Hallgren, O.; Lindstedt, S. Exhaled phospholipid transfer protein and hepatocyte growth factor receptor in lung adenocarcinoma. Respir. Res. 2022, 23, 369. [Google Scholar] [CrossRef] [PubMed]
  35. Liguori, N.R.; Lee, Y.; Borges, W.; Zhou, L.; Azzoli, C.; El-Deiry, W.S. Absence of Biomarker-Driven Treatment Options in Small Cell Lung Cancer, and Selected Preclinical Candidates for Next Generation Combination Therapies. Front. Pharmacol. 2021, 12, 747180. [Google Scholar] [CrossRef] [PubMed]
  36. Toumazis, I.; Bastani, M.; Han, S.S.; Plevritis, S.K. Risk-Based lung cancer screening: A systematic review. Lung Cancer 2020, 147, 154–186. [Google Scholar] [CrossRef] [PubMed]
  37. Scott, A.; Salgia, R. Biomarkers in lung cancer: From early detection to novel therapeutics and decision making. Biomark. Med. 2008, 2, 577–586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Smyth, E.C.; Sclafani, F.; Cunningham, D. Emerging molecular targets in oncology: Clinical potential of MET/hepatocyte growth-factor inhibitors. OncoTargets Ther. 2014, 7, 1001–1014. [Google Scholar] [CrossRef] [Green Version]
  39. Beau-Faller, M.; Ruppert, A.M.; Voegeli, A.C.; Neuville, A.; Meyer, N.; Guerin, E.; Legrain, M.; Mennecier, B.; Wihlm, J.-M.; Massard, G.; et al. MET gene copy number in non-small cell lung cancer: Molecular analysis in a targeted tyrosine kinase inhibitor naïve cohort. J. Thorac. Oncol. 2008, 3, 331–339. [Google Scholar] [CrossRef]
  40. Onozato, R.; Kosaka, T.; Kuwano, H.; Sekido, Y.; Yatabe, Y.; Mitsudomi, T. Activation of MET by gene amplification or by splice mutations deleting the juxtamembrane domain in primary resected lung cancers. J. Thorac. Oncol. 2009, 4, 5–11. [Google Scholar] [CrossRef] [Green Version]
  41. Kent, D.G.; Green, A.R. Order Matters: The Order of Somatic Mutations Influences Cancer Evolution. Cold Spring Harb. Perspect. Med. 2017, 7, a027060. [Google Scholar] [CrossRef] [Green Version]
  42. Jolly, C.; Van Loo, P. Timing somatic events in the evolution of cancer. Genome Biol. 2018, 19, 95. [Google Scholar] [CrossRef] [Green Version]
  43. Dong, Y.; Xu, J.; Sun, B.; Wang, J.; Wang, Z. MET-Targeted Therapies and Clinical Outcomes: A Systematic Literature Review. Mol. Diagn. Ther. 2022, 26, 203–227. [Google Scholar] [CrossRef] [PubMed]
  44. Kollmannsberger, C.; Hurwitz, H.; Bazhenova, L.; Cho, B.C.; Hong, D.; Park, K.; Sharma, S.; Der-Torossian, H.; Christensen, J.G.; Faltaos, D.; et al. Phase I Study Evaluating Glesatinib (MGCD265), An Inhibitor of MET and AXL, in Patients with Non-small Cell Lung Cancer and Other Advanced Solid Tumors. Target Oncol. 2023, 18, 105–118. [Google Scholar] [CrossRef] [PubMed]
  45. Camidge, D.R.; Janku, F.; Bueno, A.M.; Catenacci, D.V.; Lee, J.; Lee, S.H.; Chung, H.C.; Dowlati, A.; Rohrberg, K.S.; Font, F.E.; et al. 1490PD—A phase Ia/IIa trial of Sym015, a MET antibody mixture, in patients with advanced solid tumours. Ann. Oncol. 2019, 30, V610–V611. [Google Scholar] [CrossRef]
Figure 1. Schematic of the MET signaling pathway and the sites of action for MET targeted therapies. (A) The MET signaling pathway with downstream intracellular signaling and transcription of genes leading to enhanced morphogenesis, cell survival, motility, proliferation, and invasion. MET is activated by its ligand, HGF, and the MET receptor can interact in various ways with the EGFR receptor. (B) The MET targeted therapies presented according to specific targets, either extracellular or intracellular. The pipe-line drugs are highlighted in cursive. MET: mesenchymal epithelial transition factor, HGF: hepatocyte growth factor, EGFR: epidermal growth factor receptor, SRC: v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog, PI3K: phosphatidylinositol 3-kinase, GRB2: growth factor receptor-bound protein 2, GAB1: GRB2-associated binding protein 1, SHC: src homology 2 domain-containing, SOS: son of sevenless, RAS: rat sarcoma, RAF: rapidly accelerated fibrosarcoma, MEK: MAPK effector kinase, ERK: extracellular signal-regulated kinase, RAC1: ras-related C3 botulinum toxin substrate 1, JNK: janus kinase 1, AKT: ak strain transforming, NF-kB: nuclear factor kappa B, mTOR: mammalian target of rapamycin, FAK: focal adhesion kinase, STAT3: signal transducer and activator of transcription 3, ATP: adenosine triphosphate. Created in biorender.com.
Figure 1. Schematic of the MET signaling pathway and the sites of action for MET targeted therapies. (A) The MET signaling pathway with downstream intracellular signaling and transcription of genes leading to enhanced morphogenesis, cell survival, motility, proliferation, and invasion. MET is activated by its ligand, HGF, and the MET receptor can interact in various ways with the EGFR receptor. (B) The MET targeted therapies presented according to specific targets, either extracellular or intracellular. The pipe-line drugs are highlighted in cursive. MET: mesenchymal epithelial transition factor, HGF: hepatocyte growth factor, EGFR: epidermal growth factor receptor, SRC: v-src sarcoma (Schmidt-Ruppin A-2) viral oncogene homolog, PI3K: phosphatidylinositol 3-kinase, GRB2: growth factor receptor-bound protein 2, GAB1: GRB2-associated binding protein 1, SHC: src homology 2 domain-containing, SOS: son of sevenless, RAS: rat sarcoma, RAF: rapidly accelerated fibrosarcoma, MEK: MAPK effector kinase, ERK: extracellular signal-regulated kinase, RAC1: ras-related C3 botulinum toxin substrate 1, JNK: janus kinase 1, AKT: ak strain transforming, NF-kB: nuclear factor kappa B, mTOR: mammalian target of rapamycin, FAK: focal adhesion kinase, STAT3: signal transducer and activator of transcription 3, ATP: adenosine triphosphate. Created in biorender.com.
Cancers 15 03827 g001
Figure 2. Illustration of the PICO model used to create the search queries used in this systematic review. P: population, I: intervention, C: comparison, O: outcome, MET: mesenchymal epithelial transition factor, TKI: tyrosine kinase inhibitor.
Figure 2. Illustration of the PICO model used to create the search queries used in this systematic review. P: population, I: intervention, C: comparison, O: outcome, MET: mesenchymal epithelial transition factor, TKI: tyrosine kinase inhibitor.
Cancers 15 03827 g002
Figure 3. Flow diagram of inclusion and exclusion steps according to the PRISMA guidelines.
Figure 3. Flow diagram of inclusion and exclusion steps according to the PRISMA guidelines.
Cancers 15 03827 g003
Table 1. Overview of included EGFR and MET TKIs. EGFR and MET targeted therapies included in this systematic review. EGFR: epidermal growth factor receptor, MET: mesenchymal epithelial transition factor, TKI: tyrosine kinase receptor, VEGFR2: vascular endothelial growth factor receptor 2, RET: ret proto-oncogene, ROS1: ROS proto-oncogene 1, KIT: CD117, TIE-2: tyrosine kinase with immunoglobin and EGF homology domains 2, AXL: AXL receptor tyrosine kinase, RTK: receptor tyrosine kinase, NSCLC: non-small cell lung cancer, HGF: hepatocyte growth factor, ATP: adenosine triphosphate, CYP2C19: cytochrome P450 2C19.
Table 1. Overview of included EGFR and MET TKIs. EGFR and MET targeted therapies included in this systematic review. EGFR: epidermal growth factor receptor, MET: mesenchymal epithelial transition factor, TKI: tyrosine kinase receptor, VEGFR2: vascular endothelial growth factor receptor 2, RET: ret proto-oncogene, ROS1: ROS proto-oncogene 1, KIT: CD117, TIE-2: tyrosine kinase with immunoglobin and EGF homology domains 2, AXL: AXL receptor tyrosine kinase, RTK: receptor tyrosine kinase, NSCLC: non-small cell lung cancer, HGF: hepatocyte growth factor, ATP: adenosine triphosphate, CYP2C19: cytochrome P450 2C19.
Drug NameEffectReferences
AfatinibBinds covalently and irreversibly to the kinase domain of EGFR.Arrieta et al. [1]
CabozantinibA small molecule TKI that targets MET, VEGFR2, RET, ROS1, KIT, TIE-2, and AXL. Binds intracellularly to MET.Neal et al. [2]
Landi et al. [18]
CapmatinibA highly selective intracellular MET inhibitor.Schuler et al. [20]
Sequist et al. [22]
CrizotinibAn intracellular MET/ALK/ROS1 RTK inhibitor with high specificity for MET. Landi et al. [18]
DacomitinibA small irreversible pan-human EGFR inhibitor.Jänne et al. [21]
ErlotinibA reversible, small-molecule EGFR TKI.Spigel et al. [6]
GefitinibA reversible EGFR TKI.Arrieta et al. [1]
Wu et al. [23]
OnartuzumabA recombinant, fully humanized, one-armed anti-MET monovalent monoclonal antibody. Binds to the extracellular domain of MET without activating it and without dimerizing.Hirsch et al. [17]
Kishi et al. [15]
OsimertinibA CNS-active, irreversible EGFR TKI.Sequist et al. [22]
RociletinibAn irreversible EGFR TKI targeting mutated form of the EGFR gene.Arrieta et al. [1]
SavolitinibA small molecule, ATP competitive, selective MET TKI.Sequist et al. [22]
TepotinibA highly selective, ATP competitive MET inhibitor.Wu et al. [23]
TivantinibA selective, non-ATP-competitive MET inhibitor metabolized by CYP2C19.Yoshioka et al. [5]
Table 2. Characteristics of included articles reporting the prevalence and impact of MET and HGF expression. Summary of characteristics of the six included articles reporting numbers regarding the prevalence of aberrant MET expression and results regarding the impact of the protein HGF on outcome. NSCLC: non-small cell lung cancer, EGFR: epidermal growth factor receptor, WT: wild type, NS: mutational status not specified, TKI: tyrosine kinase inhibitor, HGF: hepatocyte growth factor, ORR: objective response rate, PFS: progression-free survival, OS: overall survival, MET: mesenchymal epithelial transition factor, GCN: gene copy number, METex14: MET exon 14 skipping mutations.
Table 2. Characteristics of included articles reporting the prevalence and impact of MET and HGF expression. Summary of characteristics of the six included articles reporting numbers regarding the prevalence of aberrant MET expression and results regarding the impact of the protein HGF on outcome. NSCLC: non-small cell lung cancer, EGFR: epidermal growth factor receptor, WT: wild type, NS: mutational status not specified, TKI: tyrosine kinase inhibitor, HGF: hepatocyte growth factor, ORR: objective response rate, PFS: progression-free survival, OS: overall survival, MET: mesenchymal epithelial transition factor, GCN: gene copy number, METex14: MET exon 14 skipping mutations.
Total
Participants
Cancer TypeMutational StatusActive DrugResults
Arrieta et al., 2016 [1] n = 66NSCLCEGFR WT
EGFR+
Afatinib (EGFR TKI)Reduced levels of HGF led to improved ORR, PFS, and OS.
Helman et al., 2018 [25]n = 77NSCLCEGFR+Rociletinib (EGFR TKI)Prevalence of MET alteration was 15.0%. MET amplification was seen in 7.6% of these, 4.5% had focal amplification, and 3.1% had aneuploidy.
Matsumoto et al., 2014 [19]n = 47NSCLCEGFR WTErlotinib (EGFR TKI)Expression of HGF resulted in poor response to erlotinib with shorter PFS. MET mutational status did not correlate to response to erlotinib or PFS.
Okamoto et al., 2014 [3]n = 295NSCLCNSChemotherapyTotally, 21 patients (7.6%) had MET mutations. MET amplifications were present in 9 (3.9%) cases. Median GCN was 8.8 among MET amplified patients.
Palmero et al., 2021 [11]n = 186NSCLCNSNoneTotally, 22.0% of patients had MET amplifications and 11.0% had METex14 mutations.
Sacher et al., 2016 [28]n = 22NSCLCEGFR WT
EGFR+
ErlotinibTotally, 45.0% of subjects harbored a MET alteration. MET amplification was present in 9.0% of the patients.
Table 3. Characteristics of included articles reporting effect of targeted therapies. Summary of characteristics and results of the 16 included articles that reported the effect of specific targeted therapies. NSCLC: non-small cell lung cancer, EGFR: epidermal growth factor receptor, WT: wild type, MET: mesenchymal epithelial transition factor, NS: mutational status not specified, IHC: immunohistochemistry, GCN: gene copy number, CEP7: centromere 7, METex14: MET exon 14 skipping mutations, NGS: next generation sequencing, TKI: tyrosine kinase inhibitor, VEGF-A: vascular endothelial growth factor A, PFS: progression-free survival, OS: overall survival, ns: not significant, ORR: objective response rate, 2/3L: second- or third-line.
Table 3. Characteristics of included articles reporting effect of targeted therapies. Summary of characteristics and results of the 16 included articles that reported the effect of specific targeted therapies. NSCLC: non-small cell lung cancer, EGFR: epidermal growth factor receptor, WT: wild type, MET: mesenchymal epithelial transition factor, NS: mutational status not specified, IHC: immunohistochemistry, GCN: gene copy number, CEP7: centromere 7, METex14: MET exon 14 skipping mutations, NGS: next generation sequencing, TKI: tyrosine kinase inhibitor, VEGF-A: vascular endothelial growth factor A, PFS: progression-free survival, OS: overall survival, ns: not significant, ORR: objective response rate, 2/3L: second- or third-line.
Study
Design
Total
Participants
Cancer TypeMutational StatusDefinition of MET+Active DrugResults
Han et al., 2017 [14]Phase IIIn = 636NSCLCEGFR+/WT, MET+/−Not defined Onartuzumab (MET TKI) + erlotinib (EGFR TKI) vs. erlotinibHigh dose onartuzumab resulted in longer PFS (4.4 months) compared to low dose (2.5 months) and erlotinib (2.5 months). No significant difference in OS.
Hirsch et al., 2017 [17]Phase IIn = 106NSCLCMET+/−MET IHC2+ or IHC3+Onartuzumab (MET TKI) + chemotherapy vs. chemotherapy + placeboMedian PFS was 5 months for onartuzumab and 5.2 months for placebo (ns). Median OS was 10.8 months for onartuzumab and 7.9 months for placebo (ns).
Jänne et al., 2016 [21]Phase In = 67NSCLCEGFR+/WTMET IHC2+ or IHC3+, MET GCN ≥ 2.1Crizotinib (MET TKI) + dacomitinib (EGFR TKI)Median PFS was 3 months with 61.0% stable disease in the escalation phase. Median PFS was 2.1 months with 32.0% stable disease in the expansion phase.
Kishi et al., 2019 [15]Phase IIn = 61NSCLCEGFR+
MET+
MET IHC2+ or IHC3+, the total number of MET genes in 20 cancer cells ≥ 90Onartuzumab (MET TKI) + erlotinib (EGFR TKI)Median PFS was 8.5 months, median OS 15.6 months, and ORR 68.9%.
Landi et al., 2019 [18]Phase IIn = 26NSCLCEGFR WT
MET+
MET-CEP7/ratio ≥ 2.2, METex14 mutationCrizotinib (MET TKI)ORR of 27.0%, median PFS 4.4 months, and median OS 5.4 months.
Moro-Sibilot et al., 2019 [29]Phase IIn = 53NSCLCEGFR WT
EGFR+
MET+/−
MET IHC2+ or IHC3+, MET GCN ≥ 6, MET exon skipping mutations in exon 14, 16–19 determined by NGSCrizotinib (MET TKI)ORR of 16.0%, median PFS of 3.2 months, and median OS of 7.7 months in the MET GCN ≥ 6 cohort. ORR of 10.7%, median PFS of 2.4 months, and median OS of 8.1 months in the MET mutated cohort.
Neal et al., 2016 [2]Phase IIn = 111NSCLCEGFR WTTested through IHC, positive if MET was expressed in either membrane or cytoplasmCabozantinib (MET TKI) + erlotinib (EGFR TKI) vs. cabozantinib vs. erlotinibMedian PFS of 1.8 months and OS of 5.1 months for erlotinib. PFS was 4.7 months and OS was 12.3 months for erlotinib + cabozantinib. PFS was 4.3 months and OS was 9.2 months (ns) for cabozantinib. No association between MET IHC+ and PFS in the cabozantinib group.
Ou et al., 2017 [24]Phase In = 26NSCLCNSMutational status not mentionedCrizotinib (MET TKI) + erlotinib (EGFR TKI)Two patients had partial response, 8 had stable disease, and 10 had progressive disease.
Scagliotti et al., 2018 [30] Phase IIIn = 109NSCLCEGFR+
MET+/−
MET IHC2+ or IHC3+, MET GCN ≥ 4Tivantinib (MET TKI) + erlotinib (EGFR TKI) vs. erlotinib + placeboGreater overall response rate (60.7%) and median PFS (13.0 months) for tivantinib + erlotinib compared to erlotinib + placebo (43.4%, 7.5 months). Similar median OS between groups (25.5 months for tivantinib and 20.3 months for placebo).
Schuler et al., 2020 [20]Phase In = 55NSCLCEGFR WT
MET+
MET H-score ≥ 150, MET/CEP7 ≥ 2.0, MET GCN ≥ 5, MET IHC 2+ or IHC3+Capmatinib (MET TKI)Median PFS was 3.7 months. In patients with MET GCN ≥ 6 median PFS was 9.3 months.
Sequist et al., 2020 [22]Phase Ib n = 180NSCLCEGFR+
MET+
MET GCN ≥ 5, MET/CEP7 ratio ≥ 2, MET IHC3+ or ≥20.0% tumor cells in NGSSavolitinib (MET TKI) + osimertinib (EGFR TKI)Partial response in 89 patients treated with savolitinib and osimertinib. PFS was 7.6 months and 9.1 months in two different subgroups.
Seto et al., 2021 [31]Phase IIn = 45NSCLCEGFR WT
MET+
METex14 mutation, MET amplification Capmatinib (MET TKI)In a subcohort with METex14 mutations receiving second or third-line (2/3L) treatment with capmatinib, overall response rate was 36.4%. In a second cohort with MET GCN ≥ 10, overall response rate to 2/3L capmatinib was 45.5%.
Spigel et al., 2013 [6]Phase IIn = 136NSCLCEGFR+/WT
MET+/−
MET IHC2+ or IHC3+Onartuzumab (MET TKI) + erlotinib (EGFR TKI) vs. erlotinibNo difference in PFS and OS between groups. In a MET+ subgroup, PFS was significantly longer for onartuzumab compared to erlotinib + placebo (2.9 vs. 1.5 months). Longer OS in the MET+ subgroup receiving onartuzumab (12.6 vs. 3.8 months).
Wakelee et al., 2017 [32]Phase IIn = 259NSCLCEGFR+/WT
MET+/−
MET IHC2+ or ICH3+Cohort 1: onartuzumab (MET TKI) + bevacizumab (VEGF-A TKI) + chemotherapy vs. placebo + bevacizumab + chemotherapy.
Cohort 2: onartuzumab + chemotherapy vs. placebo + chemotherapy
Cohort 1: longer median PFS on placebo (6.8 months) compared to onartuzumab (5.0 months). A MET+ subgroup had a median PFS of 4.8 months and OS of 9.9 months on onartuzumab vs. 6.9 months and 16.5 months on placebo.
Cohort 2: median PFS of 4.9 months and median OS of 8.5 months on onartuzumab vs. 5.1 months and 13.7 months on placebo. In a MET+ subgroup, median PFS was 5.0 months and OS was 8.0 months on onartuzumab vs. 5.0 months and 7.6 months on placebo.
Wu et al., 2020 [23]Phase Ib/IIn = 55NSCLCEGFR+
MET+
MET IHC2+ or IHC3+, MET GCN ≥ 5Tepotinib (MET TKI) + gefitinib (EGFR TKI) vs. chemotherapySignificantly longer OS (37.3 months vs. 13.1 months) and PFS (16.6 months vs. 4.2 months) for tepotinib + gefitinib in patients with MET IHC3+ or MET GCN ≥ 5.
Yoshioka et al., 2015 [5]Phase IIIn = 303NSCLCEGFR WT
MET+/−
IHC with moderate/strong intensity ≥ 50.0% of tumor cells, MET GCN ≥ 4Tivantinib (MET TKI) + erlotinib (EGFR TKI) vs. erlotinib + placeboSignificantly longer PFS for tivantinib + erlotinib (2.9 months) compared to erlotinib + placebo (2 months). No effect on OS.
Table 4. Ongoing clinical trials. Overview of currently ongoing clinical trials investigating potential pipe-line targeted therapies for the treatment of lung cancer with aberrant MET expression. NSCLC: non-small cell lung cancer, MET: mesenchymal epithelial transition factor, METex14: MET exon 14 skipping mutations, RTK: receptor tyrosine kinase, ATP: adenosine triphosphate, EGFR: epidermal growth factor receptor.
Table 4. Ongoing clinical trials. Overview of currently ongoing clinical trials investigating potential pipe-line targeted therapies for the treatment of lung cancer with aberrant MET expression. NSCLC: non-small cell lung cancer, MET: mesenchymal epithelial transition factor, METex14: MET exon 14 skipping mutations, RTK: receptor tyrosine kinase, ATP: adenosine triphosphate, EGFR: epidermal growth factor receptor.
Clinical
Trial ID
Study
Design
Study TypeTotal
Participants
Cancer TypeMutational StatusActive Drug and Effect
NCT02544633Phase IINon-randomized n = 68NSCLCMET activating mutation
MET amplification
MGCD265: oral RTK inhibitor targeting MET
NCT02920996Phase IISingle armn = 12NSCLCMETex14 mutationMerestinib: reversible type II ATP-competitive MET inhibitor
NCT02896231Phase IDose escalationn = 37NSCLCMET+PLB1001: selective MET inhibitor
NCT04270591Phase Ib/IISingle armn = 183NSCLCMETex14 mutation
MET amplification
MET overexpression
Glumetinib: selective MET inhibitor
NCT02648724Phase I/IINon-randomizedn = 57NSCLCMET amplification
METex14 deletion
Sym015: monoclonal antibody mixture targeting MET
NCT03539536Phase IISingle armn = 275NSCLCMET+Telisotuzumab vedotin: antibody-drug conjugate targeting MET
NCT02609776Phase INon-randomizedn = 780Advanced NSCLCVaryingAmivantamab: human bispecific antibody targeting EGFR and MET
NCT04077099Phase I/IISingle armn = 82NSCLCAny MET alterationREGN5093: human bispecific antibody targeting MET, inducing internalization and degradation
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

Bodén, E.; Sveréus, F.; Olm, F.; Lindstedt, S. A Systematic Review of Mesenchymal Epithelial Transition Factor (MET) and Its Impact in the Development and Treatment of Non-Small-Cell Lung Cancer. Cancers 2023, 15, 3827. https://doi.org/10.3390/cancers15153827

AMA Style

Bodén E, Sveréus F, Olm F, Lindstedt S. A Systematic Review of Mesenchymal Epithelial Transition Factor (MET) and Its Impact in the Development and Treatment of Non-Small-Cell Lung Cancer. Cancers. 2023; 15(15):3827. https://doi.org/10.3390/cancers15153827

Chicago/Turabian Style

Bodén, Embla, Fanny Sveréus, Franziska Olm, and Sandra Lindstedt. 2023. "A Systematic Review of Mesenchymal Epithelial Transition Factor (MET) and Its Impact in the Development and Treatment of Non-Small-Cell Lung Cancer" Cancers 15, no. 15: 3827. https://doi.org/10.3390/cancers15153827

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