4,523
Views
6
CrossRef citations to date
0
Altmetric
Review

Detection of MET exon 14 skipping mutations in non-small cell lung cancer: overview and community perspective

ORCID Icon &
Pages 877-886 | Received 23 Feb 2021, Accepted 27 Apr 2021, Published online: 24 May 2021

ABSTRACT

Introduction: Non-small cell lung cancer (NSCLC), which accounts for the majority of lung cancer diagnoses in the United States, has many known driver mutations, including MET exon 14 skipping mutation (METex14). The detection of oncogenic driver mutations in NSCLC and the development of drugs to target these alterations, including METex14, has created the need for accurate and reliable testing, of which next-generation sequencing (NGS) is the gold standard. However, detection of METex14 in patients with NSCLC can be challenging due to the complex biology of METex14 and the abilities of different NGS platforms to detect METex14.

Areas covered: This review provides an overview of METex14 biology, discusses the optimal platforms for the detection of METex14 in NSCLC, and provides an overview of the use of NGS in the community setting.

Expert opinion: Broad molecular testing is crucial for identifying actionable oncogenic drivers in NSCLC. METex14 is a complex oncogenic driver mutation requiring carefully optimized platforms for proper detection. To identify patients eligible for targeted therapies – including therapies targeting novel oncogenic drivers, such as MET inhibitors – community oncologists need to be aware of both the use of NGS platforms and the differences in their capabilities to detect certain oncogenic drivers.

1. Introduction

Non-small cell lung cancer (NSCLC) is a molecularly heterogeneous disease with many known driver mutations [Citation1,Citation2]. In recent years, many therapies have been approved for the treatment of NSCLC, which accounts for approximately 85% of lung cancer diagnoses in the United States [Citation2,Citation3]. Many of these treatments target oncogenic drivers, such as EGFR and ALK [Citation2].

MET exon 14 skipping mutation (METex14) has emerged as an oncogenic driver for NSCLC, occurring in approximately 3% to 4% of NSCLC cases [Citation1,Citation2,Citation4–9]. METex14 typically occurs in the absence of other driver mutations and is an adverse prognostic factor in patients with NSCLC [Citation1,Citation4,Citation5,Citation10]. A meta-analysis of patients with NSCLC found that among different histologies, METex14 occurred only in ~1% of squamous cell carcinoma, ~2% of adenocarcinoma, ~6% of adenosquamous cell carcinoma, and ~13% of pulmonary sarcomatoid carcinoma cases [Citation6]. In particular, the prevalence of METex14 in pulmonary sarcomatoid carcinomas can be substantial, ranging between 4.9% and 31.8% [Citation4,Citation6,Citation11,Citation12]. Patients with METex14 NSCLC are significantly older than patients with NSCLC without METex14 as well as those with EGFR, KRAS mutations, or ALK rearrangements. These patients are also more likely to be female and less likely to have a smoking history than other patients with NSCLC [Citation6].

Studies have shown that MET inhibitors prolong overall survival of patients with METex14 NSCLC; in contrast, immunotherapy agents have shown modest overall response rates in patients with METex14 NSCLC [Citation13–15]. Capmatinib, a selective MET inhibitor, was approved in 2020 for the treatment of adult patients with metastatic NSCLC whose tumors have a mutation that leads to METex14, based on results from the GEOMETRY mono-1 trial (NCT02414139) [Citation16,Citation17]. Additionally, tepotinib, another MET inhibitor, was recently approved for the treatment of adult patients with metastatic NSCLC harboring MET exon 14 skipping alterations, based on results from the VISION trial (NCT02864992) [Citation18,Citation19]. Other targeted therapies, such as crizotinib and savolitinib, are being investigated for the treatment of METex14 NSCLC in clinical trials ().

Table 1. Active studies with MET-targeted therapies in advanced or metastatic METex14 NSCLC: summary of ongoing trials [Citation17–22,Citation44,Citation77]

It is important to note that MET amplification, which is distinct from METex14, is an additional MET alteration that has been identified as an oncogenic driver associated with poor prognosis [Citation11,Citation23]. Unlike the genetic alterations leading to METex14, MET amplifications are caused by an increased copy number of the MET gene [Citation1,Citation24].

With the availability of targeted therapies, the field of oncology has evolved rapidly from the use of single-gene tests to broad molecular testing, enabling clinicians to identify actionable markers and high-risk patients in a cost-efficient manner [Citation25,Citation26]. However, studies have shown that there are still barriers to adoption of NGS testing and utilization of broad molecular testing results in guiding treatment decisions in the community setting. Accurate and reliable tests are needed for identification of NSCLC patients with METex14, who may benefit from these emerging therapies. Due to the heterogeneous nature of NSCLC, next-generation sequencing (NGS) platforms are a preferred method for screening NSCLC, as NGS enables the simultaneous detection of multiple actionable biomarkers [Citation1,Citation2,Citation27]. Here we will review the biology of METex14, discuss the abilities of available NGS platforms to detect METex14, and highlight the use of and the need for NGS testing in the community setting.

2. METex14 biology

MET is a proto-oncogene that encodes the c-Met tyrosine kinase protein [Citation28,Citation29]. The MET signaling pathway plays key roles in embryogenesis, wound healing, and organ regeneration; overactivation of this pathway has been observed in many types of cancer [Citation28,Citation29]. Several different types of alterations can occur within the MET gene that can result in exon 14 loss [Citation1,Citation30]. Exon 14 encodes a juxtamembrane domain, a key regulatory region of the MET receptor; loss of this region results in increased oncogenic signaling [Citation1,Citation28,Citation30]. Based on previous research, it is believed that without the juxtamembrane domain, MET receptors have increased stability and tyrosine kinase activity, which can lead to cell proliferation and tumor growth () [Citation31–35]. METex14 occurs most commonly in lung cancers, and the mutations are highly diverse [Citation1,Citation10,Citation34,Citation36,Citation37]. Alterations that lead to METex14 include base substitutions and insertions or deletions (indels) at the splice acceptor site, base substitutions and indels at the splice donor site, base substitutions and indels in intronic noncoding regions immediately adjacent to the splice acceptor site, and whole-exon deletion [Citation10,Citation36]. In a recent study of 1387 patients with NSCLC, more than 500 distinct genetic alterations leading to METex14 were identified [Citation10]. These alterations leading to METex14 were found at the splice donor site (42%), the splice acceptor site (~5%), the polypyrimidine tract (15%), and both the splice acceptor site and the polypyrimidine tract (13%) [Citation10]. All of these alterations result in the fusion of exon 13 and exon 15 in the messenger RNA transcript (). The loss of exon 14 due to these underlying genetic alterations may lead to increased MET stability and promote oncogenesis [Citation1,Citation6,Citation8,Citation31,Citation34].

Figure 1. Biology of MET exon 14 skipping mutation (METex14). (A) Left, Exon 14 of the MET gene encodes the juxtamembrane domain of the MET receptor. Right, METex14 eliminates this key regulatory region, which could lead to increased stability and increased kinase activity of the MET receptor and, ultimately, upregulated signaling of the MET pathway [Citation34]. (B) Alterations leading to METex14 result in a messenger RNA in which exons 13 and 15 are fused [Citation10,Citation34,Citation36,Citation45]. Arrows indicate the primary regions of interest for sequencing to detect METex14. HGF, hepatocyte growth factor; MET, mesenchymal–epithelial transition; mRNA, messenger RNA; P, phosphorylation; Ub, ubiquitin. Cortot AB et al, Exon 14 Deleted MET Receptor as a New Biomarker and Target in Cancers, Journal of the National Cancer Institute, 2017, 109, 5, djw262, adapted by permission of Oxford University Press

Figure 1. Biology of MET exon 14 skipping mutation (METex14). (A) Left, Exon 14 of the MET gene encodes the juxtamembrane domain of the MET receptor. Right, METex14 eliminates this key regulatory region, which could lead to increased stability and increased kinase activity of the MET receptor and, ultimately, upregulated signaling of the MET pathway [Citation34]. (B) Alterations leading to METex14 result in a messenger RNA in which exons 13 and 15 are fused [Citation10,Citation34,Citation36,Citation45]. Arrows indicate the primary regions of interest for sequencing to detect METex14. HGF, hepatocyte growth factor; MET, mesenchymal–epithelial transition; mRNA, messenger RNA; P, phosphorylation; Ub, ubiquitin. Cortot AB et al, Exon 14 Deleted MET Receptor as a New Biomarker and Target in Cancers, Journal of the National Cancer Institute, 2017, 109, 5, djw262, adapted by permission of Oxford University Press

Some studies have also identified an association between METex14 and MET amplification, but data are inconclusive. These studies note significant associations between METex14 and MET amplification, with a co-occurrence rate between 0% and 40.5% [Citation4,Citation8,Citation10,Citation17,Citation24,Citation31,Citation37–40]. However, amplification is difficult to discern across studies because there is no standard definition; the use of a low threshold for defining MET amplification will produce overlap with a large number of known dominant oncogenes [Citation4,Citation9,Citation24,Citation38,Citation41].

In addition, METex14 is not correlated with MET overexpression, whereas mixed results surround the correlation between MET amplification and MET overexpression [Citation7,Citation11,Citation24]. Quantification of MET protein levels by immunohistochemistry has shown that increases in MET levels may be present even in the absence of METex14 or MET amplifications [Citation24,Citation31,Citation42]. Most cases of MET protein overexpression are driven by other oncogenic drivers that promote tumor growth and progression [Citation4].

3. Detecting METex14 using NGS-based approaches

METex14 has been associated with poor prognosis in patients with NSCLC () [Citation4,Citation5,Citation13,Citation14,Citation43]. To improve patient outcomes, METex14 needs to be detected accurately and reliably, given the approval of capmatinib and pending approvals of additional METex14-targeted therapies [Citation16,Citation19,Citation44]. Although multiple NGS platforms are available for screening mutations, not all are optimized for detecting METex14 [Citation27,Citation45,Citation46]. One factor to consider prior to choosing a NGS platform is whether mutations, indels, gene fusions, or gene copy number alterations will be assessed [Citation47]. For the detection of novel gene alterations, such as METex14, amplicon-based and hybrid capture–based platforms are 2 commonly used NGS approaches [Citation47], but the detection rates may vary significantly between platforms [Citation27,Citation45,Citation46].

Table 2. Association of METex14 with poor overall survival [Citation4,Citation5,Citation13,Citation14,Citation43]

Amplicon-based methods rely on the enrichment of target sequences by multiplex polymerase chain reaction (PCR) amplification () [Citation47]. In 2017, Poirot et al [Citation27] published an in silico assessment of the METex14 detection rate of commercial amplicon-based cancer panels and found that no panel detected more than 63% of METex14; only 2 of the 7 panels assessed achieved this detection rate. Of the 7 panels, 4 were predicted to have METex14 detection rates of <25%. Analysis of the commercial amplicon-based panels also revealed that different panels were predicted to detect different types of METex14 events. Only 3 of 7 panels were predicted to find splice donor-site and/or Y1003 mutations at a rate >99%, whereas 2 other panels were predicted to detect acceptor-site or branch-site mutations only at a rate as high as 65% [Citation27]. Another study found that DNA-based amplicon assays performed between 2013 and 2018 had a METex14 detection rate of 1.3%, whereas RNA-based amplicon assays performed between 2016 and 2018 had a detection rate of 4.2%. Among samples tested by both amplicon-based assays, 60% that were positive according to the RNA-based assay were negative according to the DNA-based amplicon assay [Citation45].

Amplicon-based approaches may fail to identify METex14 based on the way they are constructed. The diversity of alterations leading to METex14 can lead to allele dropout and produce false negatives if there is a single-nucleotide polymorphism or short indel in the primer region, because the primer will be mismatched and will not bind [Citation27,Citation47]. Indels that substantially affect amplicon size may also evade detection [Citation47]. Large deletions, such as those over 40 base pairs in length, can go undetected with amplicon-based methods [Citation48]. Finally, poor sequencing quality at the ends of amplicons may lead to miscalling of variants [Citation47]. However, with careful optimization, such as improved primer coverage of exon 14 and fragment-length analysis, detection rates with amplicon-based methods can be improved [Citation46,Citation48,Citation49].

Figure 2. Overview of the amplicon-based and hybrid capture–based NGS methods. (a) Amplicon-based NGS methods utilize PCR primers that are designed to amplify targeted regions of interest in the genome [Citation45,Citation47]. Alterations (indels or base substitutions) that affect primer binding within the target region or are located outside of the target region (such as within intronic regions flanking MET exon 14) may go undetected. (b) Hybrid capture–based NGS methods use oligonucleotide probes to isolate larger regions of interest in the tumor genome, including regions flanking the targeted area [Citation47]. Allele dropout is less likely to occur, as long probes used for hybridization can tolerate mutations within the genomic DNA. Furthermore, DNA sequences outside of the target region (such as intronic regions flanking MET exon 14) can be captured and sequenced. NGS, next-generation sequencing; PCR, polymerase chain reaction. Adapted with permission from Elsevier: Journal of Molecular Diagnostics, 19/3, Jennings LJ et al, Guidelines for Validation of Next-Generation Sequencing–Based Oncology Panels: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists, 341–365, copyright 2017

Figure 2. Overview of the amplicon-based and hybrid capture–based NGS methods. (a) Amplicon-based NGS methods utilize PCR primers that are designed to amplify targeted regions of interest in the genome [Citation45,Citation47]. Alterations (indels or base substitutions) that affect primer binding within the target region or are located outside of the target region (such as within intronic regions flanking MET exon 14) may go undetected. (b) Hybrid capture–based NGS methods use oligonucleotide probes to isolate larger regions of interest in the tumor genome, including regions flanking the targeted area [Citation47]. Allele dropout is less likely to occur, as long probes used for hybridization can tolerate mutations within the genomic DNA. Furthermore, DNA sequences outside of the target region (such as intronic regions flanking MET exon 14) can be captured and sequenced. NGS, next-generation sequencing; PCR, polymerase chain reaction. Adapted with permission from Elsevier: Journal of Molecular Diagnostics, 19/3, Jennings LJ et al, Guidelines for Validation of Next-Generation Sequencing–Based Oncology Panels: A Joint Consensus Recommendation of the Association for Molecular Pathology and College of American Pathologists, 341–365, copyright 2017

In contrast to amplicon-based platforms, hybrid capture–based platforms are constructed using long biotinylated oligonucleotides that hybridize to target regions in the genome and enable sequencing of flanking regions () [Citation47]. These probes are significantly longer than the PCR primers used in amplicon-based assays and can therefore tolerate the presence of mismatches in the binding site without interfering with target hybridization; this bypasses the issue of allele dropout that arises in amplicon-based methods [Citation47,Citation50]. The hybrid capture method generally isolates large fragments of DNA, including regions that flank the target of interest [Citation47]. Given the diversity of METex14 events and the potential for these alterations to occur in regions flanking exon 14, the ability to probe larger regions of interest makes the hybrid capture approach more amenable to detecting the alterations leading to METex14 [Citation36,Citation47]. Additionally, because of the favorable properties of hybrid capture–based assays, this methodology has been used to capture other alterations leading to oncogenic gene fusion events [Citation51,Citation52]. However, it should be noted that hybrid capture–based platforms may not be suitable for small tissue samples, because they require relatively large amounts of DNA, ranging from 50 to 250 ng, whereas amplicon-based platforms require about 10 ng of DNA input [Citation53–55].

4. RNA-based versus DNA-based approaches for testing METex14

Both RNA-based and DNA-based approaches have been used to detect METex14 in NSCLC [Citation1,Citation27,Citation39,Citation45,Citation46,Citation56]. DNA approaches detect mutations that are predicted to lead to exon 14 skipping, whereas RNA approaches detect the direct fusion of exon 13 and exon 15 transcripts [Citation45]. Studies have found that RNA-based methods detect METex14 events at a higher rate than DNA-based approaches [Citation45,Citation46,Citation56]. In a comparison of single-gene testing methods, the mRNA-based quantitative reverse transcriptase (RT)-PCR demonstrated 100% sensitivity in detecting METex14, compared with 61.5% sensitivity using conventional DNA-based Sanger sequencing [Citation56].

Two studies found that RNA-based NGS methods outperformed DNA-based NGS methods. In a study by Davies et al [Citation45], METex14 was detected at a rate of 1.3% using a DNA amplicon–based NGS assay, compared with 4.2% using an RNA-based NGS approach (P < 0.01). Similarly, Jurkiewicz et al [Citation46] reported a METex14 detection rate of 2.5% (16/644 lung cancer tumors) when a DNA amplicon–based NGS panel was used, compared with a detection rate of 3.9% (25/644 lung cancer tumors) using a NGS-based RNA fusion panel.

The higher accuracy of RNA-based assays compared with that of DNA-based assays can be attributed to the easily detectable direct fusion events between exon 13 and exon 15 found in RNA transcripts [Citation39]. In contrast, DNA-based assays need to be able to detect many potential point mutations or deletions leading to METex14 that occur not only within the MET gene but also in neighboring intronic regions [Citation39,Citation45]. Variants detected in the RNA-based panel were likely to include alterations in the intron 13 splice acceptor site, or other sites relevant to splicing but not covered by the DNA panel, whereas METex14 detected by the DNA-based NGS panel included only alterations at or around the intron 14 splice donor site, which suggests that the DNA panel had insufficient coverage [Citation46]. DNA-based testing results should be verified by RNA-based testing unless the alteration identified by DNA-based testing has been previously validated [Citation50,Citation57]. Although high levels of accuracy have been observed with RNA-based approaches, it is important to note that one limitation is the ability to obtain enough high-quality RNA for NGS assays, which can be challenging because RNA is prone to degradation.

Notably, in some cases, DNA-based NGS methods can match the sensitivity of RNA-based assays depending on the platform used. A recent retrospective analysis of NSCLC patient samples from the GEOMETRY mono-1 study found that a hybrid capture NGS approach identified 99% of METex14 events previously detected by using a single-gene quantitative RT-PCR approach [Citation39]. METex14 in 72 of 73 patients with NSCLC were successfully detected, including 41 with canonical METex14 events, 20 with indels in the intron preceding exon 14, 10 with base substitutions in the intron following the 3′ end of exon 14, 11 with indels in the intron following the 3′ end of exon 14, and 1 with a rearrangement involving MET exon 14. Thus, this study demonstrated that a wide range of METex14 events occurring within and surrounding exon 14 can be detected using a DNA-based hybrid capture NGS approach [Citation39].

5. Optimization of METex14 testing in the community setting

METex14 is estimated to occur in ~3 to 4% of patients with NSCLC [Citation1,Citation4–8]. If METex14 events are detected at a lower rate within a practice or institution, the testing methodology may need to be assessed for the ability to detect METex14 and for whether it has been optimized for detecting METex14. Due to the complexity of METex14, certain types of alterations in patients with NSCLC may elude detection on various NGS platforms, particularly amplicon-based NGS platforms, as demonstrated by multiple studies [Citation27,Citation45–47].

When using an amplicon-based NGS assay to detect METex14, optimization may be required. For example, it has been shown that panel primers can increase intronic coverage by targeting both the 3′ splice site of intron 13 and the 5′ splice site of intron 14 to ensure detection of a wide range of METex14 events [Citation46]. One group improved their METex14 detection rates from 0.3% to 2.2% after optimizing their amplicon-based NGS assays to improve coverage of METex14 in combination with fragment-length analysis [Citation49]. The fragment analysis–based approach paired with amplicon-based NGS has been further validated by Loyaux and colleagues [Citation48] as a method to detect METex14.

6. Use of NGS testing in the community setting

To assess the use of broad molecular profiling in the community setting, a 3-year study of a large community-based oncology network was conducted from 2013 to 2015 [Citation58]. Despite the fact that both the contemporary National Comprehensive Cancer Network (NCCN) guidelines and the College of American Pathologists/International Association for the Study of Lung Cancer/Association for Molecular Pathology (CAP/IASLC/AMP) guidelines recommended routine testing for EGFR and ALK alterations [Citation59,Citation60], only 59% of patients were tested for EGFR and ALK alterations in this patient population [Citation58]. Contemporary NCCN guidelines also recommended the testing of 5 additional biomarkers [Citation59]; however, only 8% of patients were tested for all 7 NCCN-recommended biomarkers in this analysis [Citation58]. It should be noted that in a recent German study, the average biomarker testing rate before start of first-line treatment was 92.2% in 2019 for patients with nonsquamous NSCLC [Citation61].

Another retrospective review assessed the use of targeted therapy among patients with NSCLC who underwent broad molecular testing between January 2011 and January 2018 [Citation62]; 85% of patients who underwent initial screening (N = 28,998) were treated in the community setting. Of the 1260 patients with an advanced NSCLC diagnosis who had an NCCN-listed driver alteration (EGFR, ALK, ROS1, MET, BRAF, RET, or ERBB2), only 48.3% (n = 609) received NCCN-recommended therapy after the advanced diagnosis. Importantly, the study showed that receiving a targeted therapy (n = 575) was significantly associated with longer overall survival compared with not receiving a targeted therapy (n = 560) for patients with a mutation in an NCCN-listed gene (median overall survival, 18.6 months [95% CI, 15.2–21.7] vs 11.4 months [95% CI, 9.7–12.5]; P < 0.001) [Citation62].

Several factors may play a significant role in limiting the use of broad molecular testing. The prioritization of tissue specimens for histopathologic diagnosis and PD-L1 expression, which may leave little tissue remaining for broad molecular testing, could be one limiting factor [Citation63]. In some cases, patients with advanced disease may have initiated nontargeted therapy prior to receiving NGS testing results due to the turnaround time needed [Citation63].

Several studies have shown that physician attitudes and knowledge may also play a role in both the use of broad molecular testing for diagnosis and utilization of testing results in guiding treatment decisions [Citation64–69]. A recent study found that community oncologists were unsure about or incorrectly matched the molecular alteration to the targeted therapy up to 69% of the time, including for some FDA-approved therapies [Citation68]. Another survey of 100 practicing oncologists showed that 36% found NGS testing extremely important, whereas 51% found NGS testing only moderately important [Citation70].

One of the largest surveys, conducted by the IASLC from May to December 2018, evaluated 2537 respondents from 102 countries on their perceptions of current molecular testing. It found that among respondents who request tests and treat patients, 33% were unaware of the most recent guidelines for molecular testing. When molecular tests were used, over 90% tested for EGFR mutation and ALK rearrangement, whereas less than 30% tested for METex14. Few providers were using multiplex assays or including all suggested biomarkers within multiplex tests [Citation69]. Oncologists may not be choosing the ideal testing platform due to the number of commercial platforms available and lack of knowledge surrounding these platforms. No studies to date have investigated oncologists’ understanding of the capabilities and limitations of different testing platforms.

Physician attitudes may be shaped by training, experience in using broad molecular testing, and patient volume. In a study using data from the 2017 National Survey of Precision Medicine in Cancer Treatment assessing confidence levels regarding use of broad molecular testing, oncologists who had received prior genomics training reported higher confidence levels than those who did not [Citation67]. Similarly, oncologists who had prior experience using genomic tests, including gene expression and NGS tests, reported the highest confidence levels, with 60.1% being very confident and 35.6% being moderately confident in using broad molecular testing to guide patient management [Citation67].

Taken together, the results from these studies underscore the need for increased awareness of broad molecular profiling use in the community setting and education on patient outcomes for those who receive the appropriate targeted therapy. Considering that the NCCN guidelines for NSCLC treatment recommendations rely on testing for METex14 and other biomarkers [Citation71], it is critical for oncologists to incorporate broad molecular testing into their clinical decision-making. To guide treatment decisions and improve patient outcomes, oncologists should have a working knowledge of the newly approved therapies and appropriate genomic testing and incorporate precision medicine into their practices.

7. Conclusions

Detection of METex14 in patients with NSCLC can be challenging due to the complex alterations leading to the loss of exon 14 and the abilities of different NGS platforms to detect these alterations. The use of broad molecular testing in patients with NSCLC is crucial for matching their tumors with the appropriate targeted therapy, especially as the list of actionable oncogenic drivers continues to grow. Although guidelines recommend routine testing of biomarkers for patients with NSCLC, not all physicians test their patients for actionable biomarkers. For patients to have the best outcome, increased awareness and education on broad molecular profiling and genetic testing is needed within the physician community.

8. Expert opinion

Broad molecular testing is recommended in clinical practice guidelines for newly diagnosed NSCLC [Citation71,Citation72]. The approval of capmatinib provides a rationale to routinely include METex14 in these panels [Citation16]. METex14 is an established oncogenic driver in NSCLC, with a prevalence of about 3% to 4% in NSCLC cases [Citation1,Citation4–8].

Multiple studies have found that treatment with MET-targeted therapies is associated with better outcomes in patients with METex14 [Citation13,Citation14,Citation43]. In a recent real-world analysis, comparing clinical outcomes of treatment-naive patients with METex14 advanced NSCLC who received first-line therapy with capmatinib in the GEOMETRY mono-1 study and those of matched patients who received other standard-of-care first-line therapies, patients who received first-line capmatinib had longer median progression-free survival than patients with first-line chemotherapy and/or immunotherapy (12.0 months vs 6.2 months) [Citation43]. Another real-world analysis of patients with METex14 found that patients receiving a MET inhibitor had longer overall survival compared with patients who did not receive a MET inhibitor (25.3 months and 10.9 months, respectively) [Citation14]. Finally, in a retrospective study, patients with METex14 treated with single-agent immunotherapy had only modest overall response rates, with a median progression-free survival of 1.9 months and median overall survival of 18.2 months [Citation15].

Importantly, NGS panels used for broad molecular profiling are not equivalent in their ability to detect METex14. Choosing the correct test is critical because biopsies tend to have minimal tumor content and using a single sample for multiple tests may not be possible [Citation73–75]. Many amplicon-based NGS panels are not properly optimized to detect METex14, and the detection rate may be as low as 0.3% [Citation49]. One study found that a DNA-based assay had a METex14 detection rate of 1.3% [Citation45]. Additionally, an in silico study comparing 7 different DNA amplicon-based assays revealed that the detection of known METex14 ranged from 3% to 63% [Citation27]. The hybrid capture approach is a preferred method for detecting alterations leading to METex14 and other oncogenic gene fusions because the assay is able to isolate large fragments of DNA – including regions surrounding the target of interest – due to the probes being significantly longer than PCR primers, which avoids allele dropout [Citation45,Citation47,Citation51,Citation52]. However, this method also requires bioinformatic tools optimized to detect these events, which could be an additional obstacle for oncologists and laboratories looking to switch testing panels [Citation47]. Another obstacle oncologists face when choosing the correct testing platform is the duration between the test and when the patient can begin an appropriate targeted treatment. A real-world analysis investigating testing patterns and clinical outcomes in patients with METex14 found that NGS results had a median time from specimen collection to reported results of 27 days (interquartile range, 27–54 days) [Citation76]. The majority of patients initiated first-line therapy before receiving their NGS reports [Citation76]. Given the moderate response METex14 patients have with immunotherapy treatment, it is critical for patients to receive the correct targeted therapy as quickly as possible. Finally, clinical oncologists may also be hesitant to run certain tests due to concerns related to costs associated with molecular profiling and insurance coverage [Citation68].

Future research for patients with METex14 includes combination studies with MET inhibitors, such as capmatinib, and immunotherapy. Additional studies are currently being conducted on the use of MET inhibitors in patients with MET amplification [Citation17,Citation77,Citation78]. Further research is needed to explore the role of treatment sequencing in patients with METex14 treated with first-line MET inhibitors. Physicians may seek to retest patients for new molecular alterations upon disease progression. Preclinical studies have shown that resistance mutations to one type of MET inhibitor may remain sensitive to another class of MET inhibitor [Citation79,Citation80]. However, these findings will require clinical confirmation.

In 5 years, as targeted therapies move into earlier disease settings [Citation81–84], the practice norms should evolve to include routine broad molecular profiling of all NSCLCs, regardless of stage.

Article highlights

  • Non-small cell lung cancer (NSCLC) is a heterogeneous disease with numerous histologies and many known oncogenic drivers

  • The approval of MET tyrosine kinase inhibitors has made MET exon 14 skipping mutation (METex14) an actionable oncogenic driver in NSCLC

  • METex14 occurs across all histologies of NSCLC and can occur through a diverse set of underlying genetic alterations in and around exon 14 in the MET gene

  • Carefully optimized next-generation sequencing assays are required to properly detect the diverse alterations that lead to METex14

  • In the community setting, guideline-recommended biomarkers are not tested consistently

  • Surveys have found that community oncologists vary in confidence and understanding of different biomarker assays

  • Increased awareness and understanding of broad molecular profiling is critical in the community

Declaration of interest

Medical writing assistance was utilised in the production of this manuscript, provided by Chameleon Communications and funded by Novartis Pharmaceuticals Corporation.

J Subramanian has received personal fees for advisory boards and speakers bureaus from AstraZeneca, Boehringer Ingelheim, and Eli Lilly; personal fees for advisory boards from Novartis, Pfizer, G1 Therapeutics, Jazz Pharmaceuticals, and Cardinal Health; and grants from Paradigm and Biocept.

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgments

The authors thank Lisa Hang of Chameleon Communications International, New York, NY, for providing medical writing assistance, which was funded by Novartis Pharmaceuticals Corporation, East Hanover, NJ, in accordance with Good Publication Practice (GPP3) guidelines (http://www.ismpp.org/gpp3).

Additional information

Funding

This work was supported by Novartis Pharmaceuticals Corporation, East Hanover, NJ, in accordance with Good Publication Practice (GPP3) guidelines (http://www.ismpp.org/gpp3).

References

  • Frampton GM, Ali SM, Rosenzweig M, et al., Activation of MET via diverse exon 14 splicing alterations occurs in multiple tumor types and confers clinical sensitivity to MET inhibitors. Cancer Discov. 2015;5(8):850–859.
  • Hirsch FR, Scagliotti GV, Mulshine JL, et al. Lung cancer: current therapies and new targeted treatments. Lancet. 2017;389(10066):299–311.
  • American Cancer Society. About lung cancer [ cited 2020 23 July]. Available from: https://www.cancer.org/cancer/lung-cancer/about.html
  • Tong JH, Yeung SF, Chan AW, et al. MET amplification and exon 14 splice site mutation define unique molecular subgroups of non-small cell lung carcinoma with poor prognosis. Clin Cancer Res. 2016;22(12):3048–3056.
  • Yeung SF, Tong JHM, Law PPW, et al. Profiling of oncogenic driver events in lung adenocarcinoma revealed MET mutation as independent prognostic factor. J Thorac Oncol. 2015;10(9):1292–1300.
  • Vuong HG, Ho ATN, Altibi AMA, et al. Clinicopathological implications of MET exon 14 mutations in non-small cell lung cancer – a systematic review and meta-analysis. Lung Cancer. 2018;123:76–82.
  • Awad MM, Oxnard GR, Jackman DM, et al. MET exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression. J Clin Oncol. 2016;34(7):721–730.
  • Schrock AB, Frampton GM, Suh J, et al. Characterization of 298 patients with lung cancer harboring MET exon 14 skipping alterations. J Thorac Oncol. 2016;11(9):1493–1502.
  • Drilon A, Cappuzzo F, Ou SH, et al. Targeting MET in lung cancer: will expectations finally be MET? J Thorac Oncol. 2016;12(1):15–26.
  • Awad M, Lee J, Madison R, et al. Characterization of 1,387 NSCLCs with MET exon 14 (METex14) skipping alterations (SA) and potential acquired resistance (AR) mechanisms. Paper presented at: Annual Meeting of the American Society of Clinical Oncology; 2020 May 29–Jun 29–Jun 2; Virtual.
  • Liu X, Chen X, Rong Y, et al. MET exon 14 skipping mutation, amplification and overexpression in pulmonary sarcomatoid carcinoma: a multi-center study. Transl Oncol. 2020;13(12):100868.
  • Saffroy R, Fallet V, Girard N, et al. MET exon 14 mutations as targets in routine molecular analysis of primary sarcomatoid carcinoma of the lung. Oncotarget. 2017;8(26):42428–42437.
  • Awad MM, Leonardi GC, Kravets S, et al. Impact of MET inhibitors on survival among patients with non-small cell lung cancer harboring MET exon 14 mutations: a retrospective analysis. Lung Cancer. 2019;133:96–102.
  • Wolf J, Balk C, Heist RS, et al. Natural history, treatment (Tx) patterns, and outcomes in MET dysregulated non-small cell lung cancer (NSCLC) patients (Pts). Poster presented at: 30th EORTC-NCI-AACR Symposium; 2018 November 13–16; Dublin, Ireland.
  • Sabari JK, Leonardi GC, Shu CA, et al. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers. Ann Oncol. 2018;29(10):2085–2091.
  • Tabrecta [prescribing information]. East Hanover, NJ: Novartis Pharmaceuticals Corp; 2020.
  • Wolf J, Seto T, Han JY, et al. Capmatinib in MET exon 14-mutated or MET-amplified non-small-cell lung cancer. N Engl J Med. 2020;383(10):944–957.
  • Tepmetko [prescribing information]. EMD Serono, Inc.; 2021.
  • Paik PK, Felip E, Veillon R, et al. Tepotinib in non–small-cell lung cancer with MET exon 14 skipping mutations. N Engl J Med. 2020;383(10):931–943.
  • Drilon A, Clark JW, Weiss J, et al. Antitumor activity of crizotinib in lung cancers harboring a MET exon 14 alteration. Nat Med. 2020;26(1):47–51.
  • Wolf J, Seto T, Han J-Y, et al. Capmatinib in METΔex14-mutated advanced non-small cell lung cancer (NSCLC): efficacy data from the phase II GEOMETRY mono-1 study. Paper presented at: Annual Meeting of the American Society of Clinical Oncology; 2019 May 31–June 4; Chicago, IL.
  • Paik PK, Sakai H, Felip E, et al. Tepotinib in patients with MET exon 14 (METex14) skipping advanced NSCLC: updated efficacy results from VISION Cohort A. Oral presented at: 2020 World Conference on Lung Cancer Singapore; 2021 January 28–31. p. MA11.05.
  • Overbeck TR, Cron DA, Schmitz K, et al. Top-level MET gene copy number gain defines a subtype of poorly differentiated pulmonary adenocarcinomas with poor prognosis. Transl Lung Cancer Res. 2020;9(3):603–616.
  • Mignard X, Ruppert A, Antoine M, et al. c-MET overexpression as a poor predictor of MET amplifications or exon 14 mutations in lung sarcomatoid carcinomas. J Thorac Oncol. 2020;13(12):1962–1967.
  • El-Deiry WS, Goldberg RM, Lenz H-J, et al. The current state of molecular testing in the treatment of patients with solid tumors, 2019. CA Cancer J Clin. 2019;69(4):305–343.
  • Freedman AN, Klabunde CN, Wiant K, et al. Use of next-generation sequencing tests to guide cancer treatment: results from a nationally representative survey of oncologists in the United States. JCO Precis Oncol. 2018;2:1–13.
  • Poirot B, Doucet L, Benhenda S, et al., MET exon 14 alterations and new resistance mutations to tyrosine kinase inhibitors: risk of inadequate detection with current amplicon-based NGS panels. J Thorac Oncol. 2017;12(10):1582–1587.
  • Smyth EC, Sclafani F, Cunningham D. Emerging molecular targets in oncology: clinical potential of MET/hepatocyte growth-factor inhibitors. Onco Targets Ther. 2014;7:1001–1014.
  • Van Der Steen N, Giovannetti E, Pauwels P, et al. cMET exon 14 skipping: from the structure to the clinic. J Thorac Oncol. 2016;11(9):1423–1432.
  • Sadiq AA, Salgia R. MET as a possible target for non-small-cell lung cancer. J Clin Oncol. 2013;31(8):1089–1096.
  • Baldacci S, Figeac M, Antoine M, et al. High MET overexpression does not predict the presence of MET exon 14 splice mutations in NSCLC: results from the IFCT PREDICT.amm study. J Thorac Oncol. 2020;15(1):120–124.
  • Ma PC, Jagadeeswaran R, Jagadeesh S, et al. Functional expression and mutations of c-Met and its therapeutic inhibition with SU11274 and small interfering RNA in non-small cell lung cancer. Cancer Res. 2005 Feb 15;65(4):1479–1488.
  • Kong-Beltran M, Seshagiri S, Zha J, et al. Somatic mutations lead to an oncogenic deletion of met in lung cancer. Cancer Res. 2006 Jan 1;66(1):283–289.
  • Cortot AB, Kherrouche Z, Descarpentries C, et al. Exon 14 deleted MET receptor as a new biomarker and target in cancers. J Natl Cancer Inst. 2017;109(5):djw262.
  • Recondo G, Che J, Jänne PA, et al. Targeting MET dysregulation in cancer. Cancer Discov. 2020;10(7):922–934.
  • Drilon A. MET exon 14 alterations in lung cancer: exon skipping extends half-life. Clin Cancer Res. 2016;22(12):2832–2834.
  • Reungwetwattana T, Liang Y, Zhu V, et al. The race to target MET exon 14 skipping alterations in non-small cell lung cancer: the why, the how, the who, the unknown, and the inevitable. Lung Cancer. 2017;103:27–37.
  • Noonan SA, Berry L, Lu X, et al. Identifying the appropriate FISH criteria for defining MET copy number-driven lung adenocarcinoma through oncogene overlap analysis. J Thorac Oncol. 2016;11(8):1293–1304.
  • Heist RS, Garon EB, Tan DSW, et al. Accurate detection of METex14 mutations in non-small-cell lung cancer (NSCLC) with comprehensive genomic sequencing: results from the GEOMETRY mono-1 study. Poster presented at: Sixth AACR-IASLC International Joint Conference: Lung Cancer Translational Science from the Bench to the Clinic; 2020 January 11–14; San Diego, CA.
  • Le X, Kowalski D, Cho BC, et al. Liquid biopsy to detect MET exon 14 skipping (METex14) and MET amplification in patients with advanced NSCLC: biomarker analysis from VISION study. Cancer Res. 2020;80(16 suppl):Abstract 3385.
  • Schildhaus H-U, Schultheis AM, Ruschoff J, et al. MET amplification status in therapy-naive adeno- and squamous cell carcinomas of the lung. Clin Cancer Res. 2015;21(4):907–915.
  • Guo R, Berry LD, Aisner DL, et al. MET IHC is a poor screen for MET amplification or MET exon 14 mutations in lung adenocarcinomas: data from a tri-institutional cohort of the Lung Cancer Mutation Consortium. J Thorac Oncol. 2019;14(9):1666–1671.
  • Wolf J, Neal J, Mansfield A, et al. Comparison of clinical outcomes of patients with METΔex14 NSCLC treated with first-line capmatinib in the GEOMETRY mono-1 study with those of a cohort of real-world patients. Poster presented at: European Society for Medical Oncology Virtual Meeting; 2020 September 19–21.
  • Lu S, Fang J, Li X, et al. Phase II study of savolitinib in patients (pts) with pulmonary sarcomatoid carcinoma (PSC) and other types of non-small cell lung cancer (NSCLC) harboring MET exon 14 skipping mutations (METex14+). J Clin Oncol. 2020;38(suppl):Abstract 9519.
  • Davies KD, Lomboy A, Lawrence CA, et al., DNA-based versus RNA-based detection of MET exon 14 skipping events in lung cancer. J Thorac Oncol. 2019;14(4):737–741.
  • Jurkiewicz M, Saqi A, Mansukhani MM, et al. Efficacy of DNA versus RNA NGS-based methods in MET exon 14 skipping mutation detection. J Clin Oncol. 2020;38(15 suppl):9036.
  • Jennings LJ, Arcila ME, Corless C, et al. Guidelines for validation of next-generation sequencing-based oncology panels: a joint consensus recommendation of the Association for Molecular Pathology and College of American Pathologists. J Mol Diagn. 2017;19(3):341–365.
  • Loyaux R, Blons H, Garinet P, et al. MET exon 14 screening strategy: how not to miss large deletions. Ann Oncol. 2020;31(suppl 5):S1218.
  • Descarpentries C, Leprêtre F, Escande F, et al. Optimization of routine testing for MET exon 14 splice site mutations in NSCLC patients. J Thorac Oncol. 2018;13(12):1873–1883.
  • Solomon JP, Benayed R, Hechtman JF, et al. Identifying patients with NTRK fusion cancer. Ann Oncol. 2019;30(suppl 8):viii16–viii22.
  • Ruan X, Sun Y, Wang W, et al. Multiplexed molecular profiling of lung cancer with malignant pleural effusion using next generation sequencing in Chinese patients. Oncol Lett. 2020;19(5):3495–3505.
  • Reeser J, Martin D, Miya J, et al. Validation of a targeted RNA sequencing assay for kinase fusion detection in solid tumors. J Mol Diagn. 2017;19(5):682–696.
  • Williams HL, Walsh K, Diamond A, et al. Validation of the Oncomine™ focus panel for next-generation sequencing of clinical tumour samples. Virchows Arch. 2018;473(4):489–503.
  • Wen S, Dai L, Wang L, et al. Genomic signature of driver genes identified by target next-generation sequencing in Chinese non-small cell lung cancer. Oncologist. 2019;24(11):e1070–e1081.
  • Cao J, Chen L, Li H, et al. An accurate and comprehensive clinical sequencing assay for cancer targeted and immunotherapies. Oncologist. 2019;24(12):e1294–e1302.
  • Kim EK, Kim KA, Lee CY, et al. Molecular diagnostic assays and clinicopathologic implications of MET exon 14 skipping mutation in non-small-cell lung cancer. Clin Lung Cancer. 2019;20(1):e123–e132.
  • Benayed R, Offin M, Mullaney K, et al. High yield of RNA sequencing for targetable kinase fusions in lung adenocarcinomas with no mitogenic driver alteration detected by DNA sequencing and low tumor mutation burden. Clin Cancer Res. 2019;25(15):4712–4722.
  • Gutierrez ME, Choi K, Lanman RB, et al. Genomic profiling of advanced non-small cell lung cancer in community settings: gaps and opportunities. Clin Lung Cancer. 2017;18(6):651–659.
  • Ettinger DS, Wood DE, Akerley W, et al. Non-small cell lung cancer, version 6.2015. J Natl Compr Canc Netw. 2015;13(5):515–524.
  • Lindeman NI, Cagle PT, Beasley MB, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol. 2013;8(7):823–859.
  • Griesinger F, Eberhardt WE, Nusch A, et al. Biomarker testing in non-small cell lung cancer in routine care: analysis of the first 3,717 patients in the German prospective, observational, nation-wide CRISP registry (AIO-TRK-0315). Lung Cancer. 2021;152:174–184.
  • Singal G, Miller PG, Agarwala V, et al. Association of patient characteristics and tumor genomics with clinical outcomes among patients with non-small cell lung cancer using a clinicogenomic database. JAMA. 2019;321(14):1391–1399.
  • Pennell NA, Arcila ME, Gandara DR, et al. Biomarker testing for patients with advanced non-small cell lung cancer: real-world issues and tough choices. Am Soc Clin Oncol Educ Book. 2019;39(39):531–542.
  • Miller FA, Krueger P, Christensen RJ, et al. Postal survey of physicians and laboratories: practices and perceptions of molecular oncology testing. BMC Health Serv Res. 2009;9(1):131.
  • Blazer KR, Nehoray B, Solomon I, et al. Next-generation testing for cancer risk: perceptions, experiences, and needs among early adopters in community healthcare settings. Genet Test Mol Biomarkers. 2015;19(12):657–665.
  • Gray SW, Hicks-Courant K, Cronin A, et al. Physicians’ attitudes about multiplex tumor genomic testing. J Clin Oncol. 2014;32(13):1317–1323.
  • De Moor JS, Gray SW, Mitchell SA, et al. Oncologist confidence in genomic testing and implications for using multimarker tumor panel tests in practice. JCO Precis Oncol. 2020;4:620–631.
  • Singh BP, Britton SL, Prins P, et al. Molecular profiling for malignancies: knowledge gaps and variable practice patterns among United States oncologists. Annual Meeting of the American Society of Clinical Oncology; 2019 May 31–June 4; Chicago, IL.
  • Smeltzer MP, Wynes MW, Lantuejoul S, et al., The International Association for the Study of Lung Cancer global survey on molecular testing in lung cancer. J Thorac Oncol. 2020;15(9):1434–1448.
  • Menezes J, Joy V, Vora A What can we learn from oncologists? A survey of molecular testing patterns. Poster presented at: Association for Molecular Pathology Annual Meeting; 2018 November 1–3; San Antonio, TX.
  • National Comprehensive Cancer Network. Non-small cell lung cancer (version 2.2021) [ cited 2021 January 20]. Available from: https://www.nccn.org/professionals/physician_gls/default.aspx#nscl
  • Lindeman NI, Cagle PT, Aisner DL, et al. Updated molecular testing guideline for the selection of lung cancer patients for treatment with targeted tyrosine kinase inhibitors: guideline from the College of American Pathologists, the International Association for the Study of Lung Cancer, and the Association for Molecular Pathology. J Mol Diagn. 2018;20(2):129–159.
  • Yu TM, Morrison C, Gold EJ, et al. Multiple biomarker testing tissue consumption and completion rates with single-gene tests and investigational use of Oncomine Dx target test for advanced non-small-cell lung cancer: a single-center analysis. Clin Lung Cancer. 2019;20(1):20–29.e8.
  • Pennell NA, Mutebi A, Zhou Z-Y, et al. Economic impact of next-generation sequencing versus single-gene testing to detect genomic alterations in metastatic non–small-cell lung cancer using a decision analytic model. JCO Precis Oncol. 2019;3:1–9.
  • Drilon A, Wang L, Arcila ME, et al. Broad, hybrid capture-based next-generation sequencing identifies actionable genomic alterations in lung adenocarcinomas otherwise negative for such alterations by other genomic testing approaches. Clin Cancer Res. 2015;21(16):3631–3639.
  • Asad Zadeh Vosta Kolaei F, Cai B, Kanakamedala H, et al. Biomarker testing patterns and treatment outcomes in patients with advanced non-small cell lung cancer and MET exon 14 skipping mutations. Poster presented at: IASLC North America Conference on Lung Cancer; 2020 October 16–17; Virtual.
  • Paik PK, Veillon R, Cortot AB, et al. Phase II study of tepotinib in NSCLC patients with METex14 mutations. J Clin Oncol. 2019;37(15 suppl):9005.
  • Tan D, Lee D, Soo R, et al. Phase Ib results from a study of capmatinib (INC280) + EGF816 in patients with EGFR-mutant non-small cell lung cancer (NSCLC). IASLC World Conference on Lung Cancer; 2016 December 4–7; Vienna, Austria.
  • Fujino T, Kobayashi Y, Suda K, et al. Sensitivity and resistance of MET exon 14 mutations in lung cancer to eight MET tyrosine kinase inhibitors in vitro. J Thorac Oncol. 2019;14(10):1753–1765.
  • Jamme P, Fernandes M, Copin MC, et al. Alterations in the PI3K pathway drive resistance to MET inhibitors in NSCLC harboring MET exon 14 skipping mutations. J Thorac Oncol. 2020;15(5):741–751.
  • Wu YL, Tsuboi M, John T, et al. Osimertinib in resected EGFR-mutated non-small cell lung cancer. N Engl J Med. 2020;383(18):1711–1723.
  • Recondo G, Guo R, Cravero P, et al. Clinical characteristics, genomic features, and recurrence risk of early-stage MET exon 14 mutant non-small cell lung cancer (NSCLC). Poster presented at: American Society of Clinical Oncology; 2020 May 29–Jun 2; Virtual.
  • Chen AP, Eljanne M, Harris L, et al. National Cancer Institute basket/umbrella clinical trials: MATCH, LungMAP, and beyond. Cancer J. 2019;25(4):272–281.
  • Gerber DE, Oxnard GR, Mandrekar SJ, et al. ALCHEMIST: a clinical trial platform to bring genomic discovery and molecularly targeted therapies to early-stage lung cancer. J Clin Oncol. 2015;33(suppl): abstr TPS7583. 2015.