Abstract
Non-small cell lung cancer (NSCLC) represents the most prevalent histological subtype of lung cancer. Within this disease, the MET gene emerges as a critical therapeutic target, exhibiting various forms of dysregulation. Although MET tyrosine kinase inhibitors, HGF/c-MET targeting antibodies, and antibody-drug conjugates constitute the primary treatment modalities for patients with MET-altered NSCLC, numerous questions remain regarding their optimal application. The advent of immunotherapy holds promise for enhancing therapeutic outcomes in patients with MET-altered NSCLC. MET mutations can reshape the tumor immune microenvironment of NSCLC by reducing tumor immunogenicity, inducing exhaustion in immune-activated cells, and promoting immune evasion, which are crucial for modulating treatment responses. Furthermore, we emphasize the promising synergy of immunotherapy with emerging treatments and the challenges and opportunities in refining these approaches to improve patient outcomes.
Keywords: antibody-drug conjugate, c-MET, immunotherapy, non-small cell lung cancer (NSCLC), tumor immune microenvironment (TIME)
Backgrounds
In non-small cell lung cancer (NSCLC), MET gene alterations, such as amplification, exon 14 skipping, fusions, and overexpression, are observed. MET overexpression, the most common alteration at 35%–72% prevalence, 1 can be identified by selecting a suitable cutoff, potentially benefiting treatment and clinical trials. MET amplification occurs in 2%–4% of cases, and exon 14 skipping mutations in 3%–4%, 2 both being actionable alterations. MET fusions are less common, found in only 0.2%–0.3% of lung cancer patients. 3 At present, the primary therapeutic approach for NSCLC patients exhibiting MET alterations involves the use of MET tyrosine kinase inhibitors (MET-TKIs). However, the clinical outcomes achieved with this method have not met expectations. Moreover, challenges persist in the detection of MET alterations, particularly regarding the determination of cut-off values, which complicates the interpretation of test results. Consequently, there is a pressing need to investigate alternative therapeutic strategies for NSCLC cases characterized by MET mutations. One promising avenue for future exploration is the integration of immunotherapy with existing treatment modalities to enhance therapeutic efficacy.
The MET and MET pathways
One of the leading causes of global cancer fatalities is lung cancer. Approximately 85% of all patients have a histological type of NSCLC. 4 The MET gene, recognized as an oncogenic driver in this disease, becomes activated by its natural ligand HGF, thereby triggering other signaling pathways like RAS, NF-κB, PI3K/AKT, and JAK/STAT. 5 These pathways regulate complex physiological processes, including angiogenesis, embryonic development, epithelial-mesenchymal transition, and antiapoptotic signaling. 6
Variants and detection methods of MET in NSCLC
MET amplification, resulting from polysomy and local gene amplification, 7 can be identified using next-generation sequencing (NGS), fluorescence in situ hybridization (FISH), and quantitative reverse transcription polymerase chain reaction (qRT-PCR; Table 1), although establishing definitive amplification thresholds remains challenging. 8 METex14 skipping, induced by mutations that disrupt the regulatory juxtamembrane domain, 9 leads to hyperactive MET signaling, promoting uncontrolled cell proliferation and potentially oncogenic growth. 10 Kurtis et al. 11 reported that RNA-based methods have a significant advantage in sensitivity compared to DNA-based methods. This advantage is primarily due to the ability of RNA sequencing to bypass the inherent limitations of DNA sequencing technology, thereby more effectively detecting mutations and variations associated with MET. However, the high sensitivity of RNA sequencing also means that it has stricter requirements for the quality of RNA in the samples. The stability of RNA molecules is relatively low and can be easily affected by various conditions, including the methods of sample preservation and processing, all of which can significantly impact the accuracy of sequencing results (Table 1). MET fusions arise from chromosomal rearrangements, predominantly involving exon 15 and downstream kinase domains, 12 and are identified using hybridization-based technologies or anchored multiplex PCR (AMP) in RNA-NGS, with FISH and qRT-PCR as alternative detection methods (Table 1) 13 ; DNA-NGS, however, is less sensitive for this purpose. MET overexpression, associated with ligand-independent phosphorylation and aberrant activation of signaling pathways, correlates with tumor metastasis, increased invasiveness, and reduced patient survival. 14 It is assessed using various immunohistochemical antibodies and scoring systems, with a common grading scale of 0–3+ for staining intensity, and the H-score (ranging from 0 to 300) as criteria for identifying overexpression, where a score above 200 typically indicates increased MET expression levels (Table 1). 15 Different detection methods come with their own set of advantages and constraints. In clinical applications, clinicians need to choose the appropriate test for the situation and understand the limitations of the testing methods for accurate interpretation of results. When feasible, integrating a variety of detection methods can augment the sensitivity of the diagnostics.
Table 1.
MET mutation types and detection methods.
| MET alteration type | Description | Detection methods | Challenges |
|---|---|---|---|
| MET Amplification | Results from polysomy and local gene amplification | NGS, FISH, qRT-PCR | Definitive amplification thresholds challenging to establish |
| METex14 Skipping | Induced by mutations disrupting the regulatory juxtamembrane domain | Sanger Sequencing, RNA-based NGS, RT-PCR | / |
| MET Fusions | Arise from chromosomal rearrangements, often involving exon 15 and kinase domains | Hybridization-based technologies, AMP in RNA-NGS, FISH, qRT-PCR | DNA-NGS is less sensitive for detecting fusions |
| MET Overexpression | Associated with ligand-independent phosphorylation and aberrant signaling activation | IHC | Inconsistent truncation point settings for MET overexpression |
AMP, Anchored Multiplex PCR; FISH, Fluorescence in Situ Hybridization; IHC, immunohistochemical; NGS, Next-Generation Sequencing; qRT-PCR, Quantitative Reverse Transcription Polymerase Chain Reaction.
Current treatment for patients with MET-altered NSCLC
MET tyrosine kinase inhibitors
MET-TKIs can be classified as type Ia, type Ib, or type II. Type Ia relies on binding to a G1163 residue and is susceptible to mutations in this residue, while type Ib has the opposite effect. 16 Both type I and II inhibitors are ATP-competitive TKI inhibitors, but type II inhibitors have greater potential for slowing down rates and are stronger in kinase specificity than type I inhibitors; this is because type II inhibitors can expose an additional hydrophobic binding site adjacent to the MET ATP binding site by recognizing the inactive conformation of the kinase. 17 MET-TKI are currently widely used in the treatment of NSCLC with MET alterations (Figure 1), including crizotinib, capmatinib, tepotinib, savolitinib, and cabozantinib, among others. A multitude of studies on these drugs are actively underway, with the potential for some to significantly inform clinical guidance on medication use. For instance, studies such as INSIGHT 2 (NCT03940703), SAVANNAH (NCT03778229), and SACHI (NCT05015608) are currently underway to assess the efficacy of tepotinib or savolitinib in combination with osimertinib in patients with epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) resistance and MET positivity. These studies are expected to provide effective resistance overcoming strategies for MET-driven EGFR-TKI resistant patients. In addition, studies have shown that cabozantinib can compensate for crizotinib’s lack of ability to infiltrate the central nervous system, offering greater benefits to patients with brain metastases. 18 Presently, the Phase II clinical study CABinMET (NCT03911193) on cabozantinib for the patients with primary or treated MET-altered (METex14, METamp) NSCLC is underway, anticipated to yield hopeful therapeutic options for this patient group.
Figure 1.
Therapeutic approaches for diverse MET alterations in NSCLC patients. The standard pharmacological interventions for conditions characterized by MET amplification, METex14 skipping, MET fusion, and MET overexpression encompass MET-TKIs (e.g., crizotinib, capmatinib, tepotinib, savolitinib), bispecific antibodies (e.g., amivantamab), ADC drugs (e.g., Teliso-V), and monoclonal antibodies (e.g., emibetuzumab, ficlatuzumab). The concurrent administration of MET-TKIs in conjunction with immunotherapy has been utilized in the treatment of NSCLC presenting with METex14 mutations, while its efficacy in other mutational contexts remains to be elucidated.
NSCLC, non-small cell lung cancer.
Antibody therapy targeting MET
With the increasing understanding of the structure–function relationships of ligands, receptors, and activators, the development of HGF/SF-MET inhibitors for cancer therapy continues to advance. In a phase II randomized controlled clinical study, the combination of the MET monoclonal antibody emibetuzumab with erlotinib significantly prolonged overall survival (OS) (34.3 vs 25.4 months). In a subgroup analysis, the combination therapy was found to have a significant survival benefit in patients with high levels of MET protein overexpression (mPFS: 15 vs 5.4 months; Table 2). 19 Onartuzumab (MetMab), a humanized, monovalent monoclonal antibody targeting MET receptors, primarily inhibits HGF/MET binding without inducing excitatory activity or MET dimerization. 20 In a phase II randomized trial, the combination of onartuzumab and erlotinib was used to treat advanced NSCLC patients. Compared to erlotinib monotherapy alone, the combination therapy significantly improved the median progression-free survival (mPFS) (2.9 vs 1.5 months; Table 2). 21 In addition, therapies targeting the extracellular domain of MET are actively being developed; among them, REGN5093 is an METxMET bispecific antibody (Table 3), 22 and Amivantamab (JNJ-61186372) is also a potential therapeutic drug for MET. Amivantamab is an EGFR-MET bispecific antibody with immune cell targeting activity. In a phase I clinical trial CHRYSALIS (NCT02609776), NSCLC patients carrying METex14 mutations showed a good response to amivantamab treatment. The objective response rate (ORR) reached 33%, with a median PFS of 6.7 months (Table 2). 23 These results provide strong evidence for the potential of amivantamab in treating patients with METex14 mutations.
Table 2.
Advancements in main drug therapies for NSCLC with MET alterations.
| Category | Drugs | Classify | Trial number | Phase | Types of variation | Subgroup | ORR | mPFS |
|---|---|---|---|---|---|---|---|---|
| MET-TKIs | Crizotinib | Type Ia | NCT0058519 | Phase II | METex14 | / | 32% (21/65) | 7.3 months 46 |
| Capmatinib | Type Ib | NCT02414139 | Phase II | METex14 | Treatment-naive patients | 68% (19/28) | 12.4 months 41 | |
| Previously-treated patients | 41% (28/69) | 5.4 months 41 | ||||||
| Savolitinib | Type Ib | NCT02897479 | Phase II | METex14 | / | 49.2% (30/61) | 6.8 months 116 | |
| Tepotinib | Type Ib | NCT02864992 | Phase II | METex14 | Liquid-biopsy group | 48.5% (32/66) | 8.5 months 117 | |
| Tissue-biopsy group | 50% (30/60) | 11 months 117 | ||||||
| Gumarontinib | Type Ib | NCT04270591 | Phase II | METex14 | Treatment-naive patients | 71% (31/44) | 11.7 months 118 | |
| Previously-treated patients | 60% (21/35) | 7.6 months 118 | ||||||
| Antibody therapy targeting MET | Emibetuzumab | Monoclonal antibodies | / | Phase II | MET overexpression | Combination of emibetuzumab and erlotinib | / | 20.7 months 19 |
| Erlotinib alone | / | 5.4 months 19 | ||||||
| Onartuzumab | Monoclonal antibodies | / | Phase II | MET positive | Combination of onartuzumab and erlotinib | / | 2.9 months 21 | |
| Erlotinib alone | / | 1.5 months 21 | ||||||
| Amivantamab | EGFR-MET bispecific antibody | NCT02609776 | Phase I | METex14 | / | 33% (15/46) | 6.7 months 23 | |
| Antibody-drug Conjugates Targeting MET | Teliso-V | ADC | NCT02099058 | Phase Ib | MET positive | Combination of Teliso-V and erlotinib | 30.6% (11/36) | 5.9 months 27 |
| NCT02099058 | Phase II | MET positive | / | 23% (9/40) | 5.2 months 26 | |||
| NCT03539536 | Phase II | MET overexpression | high MET expression | 52.2% (12/23) | / | |||
| medium MET expression | 24.1% (7/29) 28 | / |
NSCLC, non-small cell lung cancer.
Table 3.
Ongoing studies on drug therapies for NSCLC with MET alterations.
| Category | Classify | Trial number | Phase | Types of variation | Primary outcome measures |
|---|---|---|---|---|---|
| MET-TKIs | |||||
| Crizotinib | Type Ia | NCT04084717 | Phase II | METex14, MET amplification | RR, PFS, OS |
| Capmatinib | Type Ib | NCT02750215 | Phase II | METex14 | ORR |
| NCT05110196 | Phase IV | METex14 | AEs, Percentage of participants with dose modifications, Dose intensity | ||
| NCT04677595 | Phase II | METex14 | ORR | ||
| NCT05567055 | Phase II | METex14, MET amplification | CORR | ||
| NCT05435846 | Phase I/Ib | METex14 | DLTs | ||
| Savolitinib | Type Ib | NCT04606771 | Phase II | MET amplification | ORR |
| NCT05777278 | Phase I/II | METex14 | ORR | ||
| NCT05009836 | Phase III | MET positive | PFS | ||
| NCT05015608 | Phase III | MET amplification | PFS | ||
| NCT03778229 | Phase II | MET amplification, MET overexpression | ORR | ||
| Tepotinib | Type Ib | NCT06083857 | Phase I/II | METex14 | IAEs |
| NCT03940703 | Phase II | MET amplification | DLTs, ORR | ||
| NCT04647838 | Phase II | METex14, MET amplification | ORR | ||
| NCT02864992 | Phase II | METex14, MET amplification | ORR | ||
| Cabozantinib | Type II | NCT03911193 | Phase II | METex14, MET amplification | RR |
| NCT01639508 | Phase II | Increased MET activity | ORR | ||
| Merestinib | Type II | NCT02920996 | Phase II | METex14 | ORR |
| Antibody therapy targeting MET | |||||
| Amivantamab | EGFR-MET bispecific antibody | NCT06083857 | Phase I/II | METex14 | IAEs |
| REGN5093 | MET-MET bispecific antibody | NCT04077099 | Phase I/II | METex14, MET amplification, MET overexpression | AESIs, SAEs, ORR |
| Antibody-drug Conjugates Targeting MET | |||||
| REGN5093-M114 | METxMET ADC | NCT04982224 | Phase II | MET overexpression | DLTs, TEAEs, SAEs, ORR |
| Antibody Therapy Targeting the Ligand HGF | |||||
| Ficlatuzumab | Monoclonal antibodies | NCT01039948 | Phase II | EGFR mutations combined with high MET expression | ORR |
AEs, Adverse Events; AESIs, adverse events of special interest; CORR, CNS Overall response rate; DLTs, Dose limiting toxicities; IAEs, Incidence of adverse events; NSCLC, non-small cell lung cancer; ORR, Objective response rate; OS, Overall survival; PFS, Progression-free survival; RR, Response rate; SAEs, Serious adverse events; TEAEs, Treatment-emergent adverse events.
Antibody–drug conjugates targeting MET
Antibody-drug conjugate (ADC) drugs have some advantages over traditional treatments. They combine the tumor cell-specific targeting ability of monoclonal antibodies with the potent killing activity of the drug through their unique structure and can therefore be tailored to different tumor types and antigen expression levels to enable personalized treatment. 24 Its payload released within the tumor cell not only kills cells expressing the target antigen, but may also affect surrounding tumor cells that do not express the target antigen through a bystander effect. The targeted nature of ADCs helps to reduce the risk of tumor resistance and allows for therapeutic efficacy to be achieved at lower doses, with reduced systemic toxicity and an improved therapeutic window. 25 Particularly importantly, ADC drugs offer new treatment options for patients with tumors that have failed to respond to, or have developed resistance to, conventional or targeted therapies. Teliso-V (ABBV-399) is an ADC composed of the anti-MET monoclonal antibody ABT-700 connected to the microtubule inhibitor Monomethyl auristatin E (MMAE) via a valine–citrulline linker. This drug has been granted Breakthrough Therapy Designation by the US Food and Drug Administration (FDA) for the treatment of EGFR wild-type, nonsquamous NSCLC patients with advanced/metastatic c-MET overexpression and previously treated. Progress has been made in several clinical trials. In a phase I study of 52 patients treated with ABBV-399 ⩾ 1.6 mg/kg every 2 weeks (n = 28) or ⩾ 2.4 mg/kg every 3 weeks (n = 24), among the 40 evaluable c-MET-positive patients, the ORR was 23%, and the mPFS was 5.2 months (Table 2). 26 A phase Ib study evaluated ABBV-399 in combination with erlotinib in patients with advanced NSCLC who were c-MET-positive. Among 36 evaluable patients, the overall ORR reached 23%, and the median PFS was 5.9 months (Table 2). 27 This study revealed that in EGFR-TKI-treated patients with positive c-MET and EGFR mutations, combination therapy comprising ABBV-399 and erlotinib had good antitumor activity and acceptable toxicity. The current phase II trial aimed to identify the optimal population of NSCLC patients with c-MET overexpression treated with ABBV-399 (phase I) and expand the selected population for further evaluation of efficacy (phase II). The latest data were reported at the 2022 American Society of Clinical Oncology (ASCO) Congress: the ORR was 36.5% in the EGFR wild-type nonsquamous NSCLC group, 52.2% in the high c-MET expression group and 24.1% in the medium c-M expression group (Table 2). 28 REGN5093-M114 is an ADC based on REGN5093 that is conjugated with the toxin M24 (a derivative of metformin) through the M114 linker on the surface of the antibody. In vitro experiments, REGN5093-M114 significantly reduced tumor cells in MET-driven EGFR-TKI-resistant patients. 22 A phase II study (NCT04982224) is currently being conducted on patients with MET overexpression to evaluate its clinical efficacy (Table 3), and the data will contribute to the clinical application of the novel therapy.
Antibody therapy targeting the ligand HGF
New drugs targeting HGF (HGF antagonists) or hepatocyte growth factor receptor (HGFR) can block MET signaling at the ligand–receptor level and inhibit overactivation of the MET pathway (Figure 2). 20 Ficlazumab (AV-299) is an anti-HGF monoclonal antibody. In a phase I study, good treatment tolerability was demonstrated when this drug was in combination of an anti-EGFR drug. 29 In another phase II clinical trial involving an Asian population, the evaluation of ficlatuzumab combined with gefitinib versus gefitinib monotherapy in the treatment of advanced NSCLC was revealed. By analyzing specific subgroups, it was found that the ORR in people with EGFR mutations and high MET protein expression was approximately 20% higher than monotherapy, and the mPFS was 5.5 months longer, with a significant benefit from the combination therapy. 30
Figure 2.
Possible treatment options in combination with immunotherapy. Treatment methods that have the potential to be combined with immunotherapy such as targeted MET/HGF antibodies, and MET- TKIs are designed to intercept different domains of the c-MET receptor. TPD-PROTACs are molecules engineered to induce MET degradation by forming a ternary complex with the POI and an E3 ubiquitin ligase, which triggers ubiquitination and subsequent proteasomal degradation. ADCs deliver cytotoxic payloads to tumor cells through receptor-mediated endocytosis, followed by lysosomal release, leading to cell apoptosis. TIL and CAR-T therapy involve extracting, modifying, and reinfusing immune cells to enhance antitumor activity. In targeted TAM therapy, RP-182 activates signaling pathways that promote phagocytosis and autophagy, as well as providing costimulatory signals for NF-κB activation, thereby aiding in the immune response against tumors.
POI, protein of interest; TAM, tumor-associated macrophages; TILs, tumor-infiltrating lymphocytes; TPD, targeted protein degradation.
Current problems
Inconsistency between MET mutations and MET overexpression
The overexpression of MET often occurs alongside various cancer-causing mutations, being detected in 35%–72% of NSCLC cases through immunohistochemistry (IHC). However, its significance as a separate prognostic element for this disease remains a topic of debate. 14 Thus far, the use of MET protein expression as a biomarker in targeted treatment for MET via monoclonal antibodies and MET-TKIs has not been successful 12 ; moreover, a phase III clinical study (NCT01456325) 31 comparing the efficacy of an MET monoclonal antibody combined with erlotinib versus placebo combined with erlotinib in advanced MET-positive (⩾50% of tumor cells with MET IHC scores of 2+ or 3+) NSCLC patients did not show significant improvement in clinical outcomes. This result may indeed suggest that the impact of MET as the primary biomarker for NSCLC is still unclear.
MET overexpression was correlated with METex14 skipping and MET amplification but was not strongly correlated. The three-center cohort study data of the Lung Cancer Mutation Alliance (LCMC) 32 demonstrated that out of the 71 (39%) MET IHC-positive (H-score⩾200) patients screened, only 1 (2%) was detected among the 3 MET-amplified (MET/CEP7>2.2) patients. In addition, although both (1%) patients with METex14-related alterations were detected, considering the high frequency of MET IHC positivity in lung cancer and because METex14-mutated lung cancer is usually IHC positive, MET IHC is a poor screening strategy for detecting METex14 mutations. 33 Because of the low number of patients, exhibiting MET amplification and METex14 mutation, coupled with the absence of MET fusion data, necessitates the need for additional assay data to the significance of MET overexpression as a screening tool for identifying MET alterations.
MET amplification and cut-off points
There are currently various methods available for detecting changes in MET copy number and MET expression, but unfortunately, the cut-off points for defining MET amplification and MET overexpression vary according to each detection method. MET amplification is mainly defined through two methods of FISH, which is the gold standard for MET amplification detection. The first method relies on the gene copy number (GCN) and defines MET amplification as the presence of five or more MET copies per cell (METGCN ⩾ 5) 34 ; other studies have defined this as METGCN⩾6 35 or METGCN⩾15. 36 However, MET amplification includes both true amplification and polyploid amplification and cannot be distinguished by GCN-dependent methods. Polyploid amplification of MET occurs when MET occurs with a regional or local increase in copy number on a specific arm of chromosome 7 (located at 7q31). Therefore, taking the ratio of MET to CEP7 (centromere seven counting probe) is another more accurate method in the absence of chromosome duplication. 37 Generally, MET/CEP7 ⩾ 2.0 is used to define focal amplification, while METGCN⩾5 with MET/CEP7 <2 is considered polyploid amplification. 38 Numerous studies are delving into the identification of cutoff points that would benefit patients, thereby refining detection methodologies. In one such study, the efficacy of crizotinib was assessed across varying degrees of MET gene amplification 39 : low-level amplification (MET/CEP7 between 1.8 and 2.2), moderate-level amplification (ratio between 2.2 and 5), and high-level amplification (ratio above 5). The experimental data 40 revealed that patients with high-level amplification had the highest ORR, reaching 50%, whereas the ORR for patients with low and moderate amplification were 33% and 20%, respectively. In subsequent updates to this study, the threshold between moderate and high amplification was adjusted to an MET/CEP 7 ratio of 4, and it was found that patients with high-level amplification consistently achieved the best therapeutic outcomes (ORR of 40%, with an mPFS of 6.7 months). Similar conclusions have been drawn from other studies.41,42 This suggests that, in general, patients with higher levels of MET amplification respond better to MET inhibitors. Establishing a relatively high cutoff value helps to identify a patient population that is more sensitive to the medication and tends to have better therapeutic outcomes, although this may exclude some patients who could potentially benefit. The “NCCN Clinical Practice Guidelines in Oncology – Non-Small Cell Lung Cancer (Version 4.2024)” 43 recommend defining high-level MET amplification as GCN⩾10 detected by NGS. Conversely, setting a lower cutoff value to include more patients might encompass many who do not respond to the treatment. For instance, in the onartuzumab study, half of the patients selected based on MET expression levels participated in the research, a proportion that significantly exceeds the likely number of patients who are genuinely reliant on the MET pathway. 44
It has to be said that the level of MET expression, as a continuous variable, has a certain degree of arbitrariness in the setting of any cutoff point, rather than being entirely based on biological mechanisms. 2 In clinical practice, it may be necessary to select cutoff points in a more personalized manner, establishing inclusive thresholds. Reducing interobserver variability in FISH interpretation through digital pathology or confirming MET amplification through orthogonal experiments may be effective solutions. 8
MET-targeted therapy exhibits limited PFS and susceptibility to drug resistance in NSCLC
Since PAIK et al. 45 first reported crizotinib and cabozantinib as third-line treatment options for patients with stage IV lung adenocarcinoma carrying METex14 mutations in a small retrospective series of studies in 2015, many studies targeting MET-TKIs have been initiated in the field since, also mostly focusing on METex14 mutations. However, many clinical studies related to MET-TKIs have shown that although the ORR is high, PFS is not ideal. For example, a subsequent additionally reported phase I clinical trial by PAIK et al. (Profile 1001) 46 showed an ORR of 32.3%, a duration of response (DOR) of 9.1 months, and a PFS of only 7.3 months. MET amplification has been shown to exhibit sensitivity to MET-TKI, but there is a lack of studies based on large samples to further clarify this. 2 In a clinical study utilizing capmatinib for the treatment of NSCLC patients with METex14 mutations or MET amplifications, the ORR was 68% and the PFS was 12.4 months for untreated patients with METex14 mutations. For patients with MET amplification, those with a GCN of 10 or higher had an ORR of 40% and a PFS of 4.2 months; however, patients with a GCN below 10 exhibited a limited therapeutic response. 41 This study not only pointed out that the level of MET amplification is an important factor influencing the response to treatment, but also revealed a limited PFS. In patients with lung cancer, MET fusion is a rare event. A study found that patients with MET fusions responded to crizotinib treatment, with PFS ranging from 4 to 14 months. 3
Drug resistance significantly impedes the efficacy of MET inhibitors, with research primarily centered on MET-TKIs. This resistance is categorized into primary and secondary forms, yet the underlying mechanisms remain not fully understood. Studies have shown that the presence of MET gene alterations accompanied by other co-mutated genes may be one of the primary resistance mechanisms, such as PIK3CA mutations. 47 The occurrence of secondary drug resistance may be due to changes in the MET domain after initial treatment with MET inhibitors. Type I and type II TKIs act at different sites on the MET molecule, and their resistance mutations occur at different residues. For example, a study reported that a patient with METex14 had an acquired mutation in the MET kinase domain D1228N during the progression of crizotinib treatment. 48 Combining MET-TKIs with immunotherapy to improve PFS and overcome drug resistance is a promising therapeutic direction. In recent years, research on organoids has developed rapidly, and using organoids to screen sensitive drugs may be an effective tool for further studying MET-TKIs. 49
Features of the TIME in patients with MET-altered NSCLC
Antigen presentation and immunogenicity characteristics of TIME
The tumor mutational burden (TMB) is an important indicator of the number of mutations in cancer. The presence of high TMB suggests that tumor cells have undergone numerous mutations, leading to the creation of additional new antigens that are immunogenic and more likely to activate T-cell responses. After analyzing The Cancer Genome Atlas (TCGA online dataset, a study revealed that patients with MET amplification had a significantly greater TMB than did patients without MET alterations. These findings could lead to the use of MET amplification as a representative predictive biomarker for immunotherapy, providing new perspectives and evaluation tools for clinical treatment. 50 A comparative analysis was carried out for 138 patients with METex14-mutant and 5162 patients with MET-WT NSCLC at the 2022 World Conference on Lung Cancer. Among METex14 lung cancer patients, the proportion of patients with high TMB (TMB-H, defined as TMB ⩾ 10 mt/Mb) was significantly lower, at 16.3%, than that of patients with wild-type MET. 51 Another study targeting three groups of NSCLC patients with oncogene alterations (a total of 4189 patients) revealed similar characteristics of TMB in the METex14 cohort. 52 Thus, it is evident that even within the MET mutation spectrum, the immunological features vary among different types due to their distinct underlying characteristics. This diversity has led to a lack of consensus on the efficacy of immunotherapy in studies focusing on NSCLC patients with MET alterations. This difference may be due to the expansion of myeloid-derived suppressor cells (MDSCs) by mesenchymal stem cells (mediated by downstream STAT3 phosphorylation of the HGF/c-MET signaling pathway). MDSCs can inhibit cytokine production, inhibit T-cell proliferation, and significantly inhibit immune cell responses, NK cell cytotoxicity, and dendritic cell (DC) antigen presentation. 53 Another possible potential reason is the correlation of high MET copy number with lower stimulation of interferon gene (STING) signaling. The MET gene weakens the immune system’s ability to recognize and attack tumors by inhibiting the STING signals, reducing the immunogenicity of tumors and thus affecting the effectiveness of immunotherapy. 54 Overall, although MET mutations are linked to a higher TMB in NSCLC patients, these mutations reduce the immunogenicity of tumors by altering the immune microenvironment, which may be the reason for the poor efficacy of immunotherapy. This discovery suggests that for NSCLC patients with MET alterations, a deep understanding of the characteristics of their immune microenvironment is crucial for optimizing immunotherapy regimens.
Infiltration of immune cells
CD8+ T cells are the most essential component of tumor-infiltrating lymphocytes (TILs) when killing tumor cells in the body, 55 and a low infiltration level of CD8+ T cells in lesions is closely related to poor prognosis. 56 A study showed a significant increase in the number of TILs in patients with MET-altered tumors, and MET amplification was independently correlated with the infiltration levels of CD8+ T cells and TILs in tumors. 57
Among immune cells, tumor-associated macrophages (TAMs) have the highest infiltration rate in the tumor microenvironment (TME). TAMs can stimulate invasion, proliferation, and metastasis, as well as tumor angiogenesis and TME immunosuppression, in NSCLC cells to promote tumor occurrence and development. 58 Therefore, NSCLC patients with higher levels of TAM infiltration often have poorer prognoses, 59 indicating that TAMs play an important role in the occurrence and development of NSCLC. Macrophages are highly malleable and can differentiate specifically into M1 and M2 phenotypes in different tissue environments and produce different effects. M1-type macrophages have antitumor activity and help activate adaptive immune and inflammatory responses, while M2-type macrophages promote tumor occurrence and development by inhibiting the immune function of the TME and promoting angiogenesis, tissue reconstruction, and damage repair. 60 Research has shown that activation of the downstream PI3K pathway by the HGF/c-MET signaling pathway can lead to the dependent induction of Arg-1 expression, which transforms tumor-inhibiting M1-type macrophages into tumor-promoting M2-type macrophages. 61 CSF-1R is a protein that mediates the survival of macrophages in tumor tissue, and its inhibition is considered a therapeutic strategy for eliminating TAMs. However, studies have shown that although CSF-1R inhibitors can effectively eliminate TAMs, they can cause tumor-associated fibroblasts to release more of the chemokine CXCL1, thereby recruiting more immunosuppressive polymorphonuclear MDSCs and promoting tumor growth. 62
Tregs are a subgroup of T cells that can downregulate or inhibit the induction and proliferation of effector T cells and participate in regulatory immunity. A study revealed that under HGF induction, monocytes can differentiate into tolerant DCs. In addition, HGF also enhances the ability of DCs to produce the immunosuppressive cytokine IL-10. These findings further confirm that the HGF/c-MET pathway is involved in Treg accumulation. 63 Tregs can upregulate immunosuppressive signals, such as T-cell immunoglobulin and mucin domain-3 (TIM-3) and lymphocyte activation gene 3 (LAG-3), 64 to produce the immunosuppressive cytokines IL-10 and TGF-β, which can inhibit the activity of CD8+ T cells. In addition, Treg cells can also contact and interact with DCs, causing DC depletion, further leading to reduced activation of CD8+ T cells and inhibiting CD8+ T-cell-mediated cytotoxicity. 65 Moreover, as inhibitory cytokines of NK cells, TGF-β and IL-10 can inhibit the migration of NK cells to the TME and its antitumor effects, leading to NK cell dysfunction. 66
Therefore, MET mutations cause extensive infiltration of immunosuppressive cells, including Tregs, TAMs, MDSCs, etc., as well as depletion of immune-activated cells, inhibition of the antitumor immune response, and therapeutic methods targeting the HGF/c-MET pathway are effective strategies.
Tumor cells evade immune killing
PD-1 and PD-L1 play important negative regulatory roles in the immune response to tumors, enabling tumor cells to successfully evade immune system attacks. Research has shown that in lung adenocarcinoma patients, MET mutations are significantly positively correlated (p < 0.001) with high PD-L1 expression. 67 According to previous studies, there is a positive correlation between PD-L1 expression and MET amplification in NSCLC patients, 68 and patients with METex14 skipping have a high positivity rate. 69 These studies have some guiding significance for the application of immune checkpoint inhibitors (ICIs) in patients with MET-altered NSCLC. However, as previously mentioned, different types of MET mutations exhibit similar yet distinct immunological characteristics, which suggests that for patients with genomic alterations in the MET gene, selecting individuals based on biomarkers associated with immunotherapy prognosis can more precisely identify those who are likely to benefit.
TAMs are also interrelated with the expression of PD-L1; almost all PD-1+ TAMs express the M2 type, while PD-1- TAMs mainly express the M1 type, and high expression of PD-L1 can induce M2 polarization of TAMs. 70 M2-type TAMs can inhibit NK cell activity and promote the expression of PD-L1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), hindering effector T-cell infiltration by blocking immune checkpoints 71 ; however, the secretion of TGF-β upregulates Tregs, the Foxp3 gene induces CTLA-4 expression on the surface of Tregs, and CTLA-4 binds to CD80 and CD86 on antigen-presenting cells, resulting in their downregulation and inhibition of T-cell activation. 72 In addition, the surface-characteristic marker molecule CD25 has a high affinity for IL-2 and limits the activation and proliferation of effector T cells by binding to IL-2. 73 Most tumor-infiltrating Tregs exhibit increased PD-1 expression, thereby inducing immunosuppression through the interaction of PD-1 and PD-L. 74 Furthermore, MDSCs can also induce T-cell tolerance by expressing inhibitory receptors such as PD-L1 and CTLA-4. 75
In summary, MET mutations promote immune escape mechanisms by inducing and recruiting immunosuppressive cells and increasing the expression of various immunosuppressive molecules. By blocking these immunosuppressive mechanisms, the underlying antitumor immune response can be restored. Immunotherapy with ICIs, such as CTLA-4 monoclonal antibodies and PD-1 or PD-L1 monoclonal antibodies, can rescue cytotoxic effector CD8+ T-cell dysfunction and kill tumor cells.
Opportunities for immunotherapy via the MET pathway
Immunotherapy
ICIs have become one of the first-line treatment options for NSCLC patients who lack driver mutations. 76 Retrospective studies indicate that patients with NSCLC harboring KRAS, BRAF mutations, or co-occurring TP53 mutations derive more favorable outcomes from ICIs, whereas those with EGFR, ALK, or MET mutations exhibit suboptimal benefits from immunotherapy. 77 However, notably, certain studies have demonstrated a positive correlation between PD-L1 expression and MET amplification in NSCLC patients, suggesting that ICIs may be effective in treating MET-amplified tumors. 68 In patients with NSCLC harboring MET exon 14 skipping, the efficacy of immunotherapy remains a contentious issue. The IMMUNOTARGET study 78 indicated that patients with MET-mutated subgroups, including MET amplification and MET exon 14 skipping mutations, derive relatively limited clinical benefits from immunotherapy, with an ORR of 16% and an mPFS of 3.4 months. A study specifically targeting MET exon 14 skipping also reported a similar ORR of 17% and an mPFS of 1.9 months, 69 consistent with another study’s finding that NSCLC patients with MET mutations may have a lower response to immunotherapy, although the sample size of that study was small. 79 Despite the aforementioned studies showing that patients with MET genomic alterations treated with ICIs have an ORR of less than 20%, suggesting that ICI therapy has some efficacy but is not highly desirable, a retrospective study (GFPC 01-2018) 80 reported a higher ORR. In this study, 30 patients with MET exon 14 skipping treated with ICIs achieved an ORR of 35.7% and an mPFS of 4.9 months. Researchers believe that this may be related to the relatively low number of lines of ICI treatment (63% were first- or second-line treatments) and the higher proportion of patients with high PD-L1 expression (⩾50%) included in the study (37%). Further analysis of the three GFPCs also revealed a higher ORR, which was 43%. 81 This suggests that additional biomarker data are needed in order to determine which patients are most likely to benefit from ICIs.
In recent years, a study 82 has reported a case of an NSCLC patient with an MET exon 14 skipping mutation who experienced a brief period of PFS and rapidly developed resistance following treatment with crizotinib. Upon switching to a regimen combining immunotherapy with chemotherapy, the patient’s PFS was extended to 15.0 months. Another study also found that chemoimmunotherapy prolonged PFS compared to chemotherapy. 83 This outcome suggests that the chemoimmunotherapy approach may enhance patient benefit. A recent retrospective study 84 has further corroborated this conclusion. The study found that in the subgroup of patients with MET mutations, including MET amplification and MET exon 14 skipping, the ORR to first-line chemoimmunotherapy was 60%, with an mPFS of 6.2 months. In the second-line treatment, the ORR was lower, at 30.8%, with a median PFS of 5.7 months, and both first- and second-line treatments demonstrated manageable safety profiles.
Immunotherapy combined with targeted therapy
Given the suboptimal outcomes of monotherapy with ICIs in patients with MET-mutated NSCLC, a combination or sequential approach with targeted therapies may represent a promising strategy. Preclinical research utilizing lung cancer cell lines and models from other cancer types has demonstrated that MET inhibitors can reduce the number of neutrophils in tumors and draining lymph nodes that increase reactively due to immunotherapy. These neutrophils rapidly acquire immunosuppressive characteristics within the T-cell inflammatory microenvironment and limit the expansion and effector functions of T cells. By modulating the behavior of immune cells in the tumor microenvironment, particularly neutrophils, MET inhibitors enhance the antitumor immune response mediated by T cells, thereby improving the efficacy of cancer immunotherapy. 85 However, combination therapy may be associated with more serious adverse effects. Several clinical trials have attempted to combine EGFR-TKIs or ALK-TKIs with PD-1/PD-L1 monoclonal antibodies; however, these endeavors have been unsuccessful due to a higher incidence of adverse events, such as interstitial pneumonia and hepatic injury.86,87 Furthermore, two clinical trials exploring the combination of ICIs with MET-TKIs (NCT04323436, NCT04139317) were prematurely terminated due to toxicity associated with the combined therapy. In one of these trials, out of 51 patients who received the combined treatment, 19 (37.3%) had to discontinue therapy, and there were four suspected treatment-related fatalities. 88 Future research will continue to explore the potential of the combination of ICIs and MET-TKIs, aiming to provide new therapeutic strategies for patients with lung cancer harboring MET alterations. Concurrently, it is imperative to closely monitor the safety profiles of these medications.
Immunotherapy combined with ADC drugs
Previously reported ADC drugs are often associated with dose-limiting toxicity, but the emergence of trastuzumab deruxtecan (DS-8201), a new ADC drug designed using a novel linker peptide-loading technology, has overcome the limitations of the previous generation of compounds and has a synergistic attenuating effect, heralding the arrival of the next generation of ADCs and showing potential for combination with PD-1/L1. Datopotamab deruxtecan (Dato-DXd) is an innovative ADC designed to target human trophoblast surface antigen 2 (TROP2). This drug consists of multiple components, including humanized anti-TROP2 IgG1 monoclonal antibodies, potent topoisomerase I inhibitors that can be effectively loaded, and a stable tetrapeptide-cleavable linker. 89 At the 2023 ASCO Congress, the midterm analysis results of the clinical study TROPION-Lung02 (NCT04526691) were announced, which evaluated the efficacy of Dato-DXd + pembrolizumab dual and Dato-DXd + pembrolizumab + platinum-based chemotherapy triple in the treatment of newly diagnosed advanced/metastatic NSCLC patients without driver gene mutations. The results showed that the ORRs were 50% and 57%, respectively. 90 Research on TROPION-Lung04 (NCT04612751), TROPION-Lung07, and TROPION-Lung08 (NCT05215340) is currently underway to further evaluate the potential of Dato-DXd in combination with immunotherapy.91,92 As a “highly effective, targeted chemotherapy” drug, ADC drugs hold promise in exerting synergistic effects when combined with immunotherapy and could become a new direction for next-generation tumor treatment.
Immunotherapy combined with targeted protein degradation
Targeted protein degradation (TPD) is an emerging therapeutic approach with enormous therapeutic potential. For certain proteins that are difficult to target with conventional small molecules, TPD can alter cellular protein homeostasis mechanisms. 93 PROTAC is a bifunctional small molecule composed of two ligands covalently linked by linkers of 5~15 carbon atoms or other atoms; one ligand recruits and binds to the cancer cell-dependent protein of interest (POI), while the other recruits and binds to the E3 ubiquitin ligase. 93 PROTAC proteins simultaneously bind to POI and ligases, leading to ubiquitination of the POI and subsequent degradation by the ubiquitin–proteasome system (UPS). Following degradation, the PROTAC is recycled to target another copy of the POI (Figure 2). 94 PROTAC technology has several advantages over traditional small molecule inhibitors due to its unique mechanism of action. Unlike traditional inhibitors that rely on the active site of target proteins, PROTACs are able to degrade proteins that lack active sites or are difficult to target by traditional means, such as transcription factors and other non-enzymatic proteins, effectively solving the problem of “undruggable.” In addition, the catalytic properties of PROTACs allow a single molecule to degrade multiple target proteins in a cyclic manner, reducing the dependence on prolonged drug exposure and sustained high drug concentrations. Compared to non-catalytic, occupation-driven mechanisms of drug action, PROTAC molecules can act at lower doses, reducing the risk of side effects. They also have the advantage of a lower probability of resistance development and a more rapid efficacy response. 95 Two studies have evaluated c-MET PROTACs via the multitargeted kinase inhibitor foretinib, which includes two different E3 ubiquitin ligases, von Hippel–Lindau (VHL) recruitment and cereblon (CRBN) recruitment; both of these studies revealed that foretinib-based c-MET PROTACs effectively degrade c-MET, inhibit tumor cell proliferation, and counter the increased c-met stability of exon 14 deletion and resistance to HGF-mediated degradation. 96 These findings suggest that the application of PROTACs in patients with MET-altered NSCLC may have considerable potential. Subsequently, other studies have designed, synthesized, and evaluated highly potent orally active c-MET PROTACs using tepotinib and thalidomide, which have higher pharmacological activity, broader selectivity, and lower cytotoxicity than previously reported. 97 Targeting immunosuppressive molecules on TAMs is also a promising therapeutic approach that can effectively inhibit endocytosis by interfering with the phagocytic receptor MerTK. This process can lead to the accumulation of apoptotic cells in tumor cells, triggering a type I interferon response and ultimately enhancing antitumor immunity. In addition, other emerging TPD technologies, including lysosome-targeting chimaera, antibody-based PROTAC (AbTAC), autophagy-targeting chimaera (AUTAC), autophagosome-tethering compound (ATTEC), molecular glue, photocontrolled PROTACs, and hydrophobic labeled small molecules, are also important additions to the chemical regulation of intracellular protein homeostasis. 98
Targeted treatment of TAMs
Given that MET mutations are significantly associated with high PD-L1 expression and that high PD-L1 expression can induce M2 polarization of TAMs,67,70 MET mutations can promote the polarization of TAMs from the M1 to M2 phenotype through high PD-L1 expression. One of the key characteristics of macrophages is their plasticity, which allows them to alter their phenotype in response to the tumor microenvironment; therefore, the repolarization strategy of reprogramming TAMs to an antitumor phenotype is very promising in tumor therapy. RP-182 is a synthetic 10-mer amphiphilic analog of a host defense peptide that selectively induces a conformational switch to the mannose receptor CD206 expressed on M2-type TAMs, reprogramming M2-type TAMs to the antitumor M1 type (Figure 2). 99 Combination therapy comprising RP-182 expression and chemotherapy or ICIs may be an effective strategy for inhibiting tumor growth and prolonging survival. In addition, targeting immunosuppressive molecules on TAMs is an effective method that can selectively inhibit endocytosis by blocking the phagocytic receptor MerTK, leading to the accumulation of apoptotic cells in tumor cells and triggering a type I interferon response, enhancing antitumor immunity. 58 PD-1-PD-L1 therapy can act directly on macrophages or synergistically with anti-MerTK antibody therapy to treat tumor-bearing mice and stimulate T-cell activation. 100
Adoptive cell immunotherapy
TIL therapy refers to the isolation of infiltrating lymphocytes (TILs) from tumor tissue; these cells are cultured and amplified in vitro before being reintroduced into the body (Figure 2). TIL therapy has the advantages of multiple targets, tumor targeting, and minimal side effects and has unique advantages in the treatment of solid tumors. 101 In a phase I clinical trial, 102 20 patients with advanced NSCLC were administered TIL or IL-2 along with nivolumab after lymphodepletion, 11 of the 13 evaluable efficacy patients experienced a decrease in tumor burden, 3 responded, and 2 achieved a complete response after 1.5 years. Studies have shown that the immune system’s response to neoantigens formed by mutations and the subsequent development of tumor-specific lymphocytes increase the likelihood of benefiting from TIL therapy in patients with NSCLC with a high mutational burden, such as those with MET amplification. 103 TIL therapy for solid tumors has unique advantages, but its promotion and popularization face some challenges, mainly including expensive and time-consuming tissue collection and production processes, difficulties in achieving TIL standardization for different patients, and difficulties in selecting prognostic markers. There is still room for further improvement and application of TIL therapy. 101
CAR-T-cell therapy is a novel therapy that involves the genetic modification of specific tumor antigens and precisely kills tumor cells. CAR-T-cell-targeting c-MET has been developed and verified for its specific killing activity against tumor cells. 104 However, CAR-T-cell therapy for lung cancer, while potentially effective, faces numerous challenges in its widespread use. These include severe targeted toxicity, antigen evasion, and CAR-T-cell persistence. 105 Innovative CAR designs are urgently needed to be developed. CAR-NK therapy appears to have a more favorable safety profile compared to CAR-T-cell therapy, as demonstrated in a clinical trial where no adverse immune-related events were associated with CAR-NK-cell therapy, 106 suggesting that it has certain therapeutic potential. However, the inherent characteristics of NK cells, such as increased difficulty in amplification, isolation, and genetic engineering compared to those of T cells, as well as limited persistence in vivo after infusion, remain unresolved. 107
Biomarker discovery and patient selection
TMB, microsatellite instability (MSI), and PD-L1 expression are commonly used biomarkers in the field of immunotherapy. Studies have confirmed that MSI can serve as a predictive biomarker for the efficacy of immunotherapy.108,109 However, a retrospective study has indicated that in NSCLC patients with rare driver genes such as BRAF, MET, RET, HER2, etc., the MSI status is mostly microsatellite stable, similar to the overall situation in lung adenocarcinoma. 110 This suggests that in the population of NSCLC patients with MET mutations, the MSI status may not be as significantly predictive of the efficacy of ICIs as it is in MSI-High tumors.
Regarding PD-L1 expression, there is disagreement among multiple studies. In the GFPC study, the higher ORR may be related to the higher proportion of patients with PD-L1 expression⩾50% (37%). 80 However, another study has suggested that the efficacy of immunotherapy is not directly related to PD-L1 expression but is associated with TP53 mutations. 83 In addition, a meta-analysis did not observe significant differences in PD-L1 expression among patients with different levels of MET expression, 111 but this finding should be interpreted with caution due to the small number of studies included. It is worth noting that PD-L1 expression is correlated with the specific gene expression characteristics of CD8+ T cells, 112 and combining PD-L1 expression with the infiltration of CD8+ T cells in tumor tissue may improve the accuracy of predicting the efficacy of ICIs. 113
As for the research results on TMB, they also show certain differences. A studies show no difference in TMB between mutated and wild types, 114 while another study found that TMB in mutated types is lower than in wild type. 69 In addition, the expression level of MET itself is also related to the efficacy of immunotherapy. Studies have found that patients with a high number of MET copies respond poorly to ICIs.54,115 These findings suggest that for patients with MET genomic alterations, screening based on prognostic biomarkers related to immunotherapy may help to more accurately identify the population that can benefit from the treatment.
Conclusion
MET mutations reshape the immune microenvironment of NSCLC through intricate mechanisms, including the immunogenicity of the TIME, depletion of immune-activated cells, infiltration of immunosuppressive cells, and evasion of immune killing, affecting the response of patients to immunotherapy. The application of immunotherapy in patients with MET-altered NSCLC holds promise, yet monotherapy presents numerous challenges. Combination therapy offers the potential to overcome these limitations and unlock the full potential of immunotherapy in this setting.
Acknowledgments
None.
Footnotes
ORCID iD: Xiaoling Xu
https://orcid.org/0000-0003-0618-494X
Contributor Information
Lisha Ye, Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
Wenjing Wang, Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
Huihui Li, Department of Medical Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China.
Yongling Ji, Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 Banshan East Road, Hangzhou, Zhejiang 310022, China.
Xiuning Le, Division of Internal Medicine, Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4000, USA.
Xiaoling Xu, Postgraduate Training Base Alliance, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Zhenmin Road, Shanghai 200433, China.
Declarations
Ethics approval and consent to participate: Not applicable.
Consent for publication: Not applicable.
Author contributions: Lisha Ye: Conceptualization; Writing – original draft; Writing – review & editing.
Wenjing Wang: Writing – review & editing.
Huihui Li: Writing - review & editing.
Yongling Ji: Supervision; Writing – review & editing.
Xiuning Le: Writing – review & editing.
Xiaoling Xu: Conceptualization; Funding acquisition; Supervision; Writing – review & editing.
Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: This research was supported by Zhejiang Provincial Natural Science Foundation of China (Grant No. LTGY23H160011), Program of Province TCM Sci-Tech Plan (Grant No. 2024ZL298), Shanghai Municipal Science and Technology Innovation Action Plan (23Y11908077), Shanghai Pulmonary Hospital Institutional Project (FKLY20006), Establishment and Application of Precision Diagnosis and Treatment System for Thoracic Tumors, and Beijing Xisike Clinical Oncology Research Foundation (Grant No. Y-Q201802-074).
The authors declare that there is no conflict of interest.
Availability of data and materials: Not applicable.
References
- 1. Salgia R. MET in lung cancer: biomarker selection based on scientific rationale. Mol Cancer Ther 2017; 16: 555–565. [DOI] [PubMed] [Google Scholar]
- 2. Sacco JJ, Clague MJ. Dysregulation of the Met pathway in non-small cell lung cancer: implications for drug targeting and resistance. Transl Lung Cancer Res 2015; 4: 242–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Sun D, Wu W, Wang L, et al. Identification of MET fusions as novel therapeutic targets sensitive to MET inhibitors in lung cancer. J Transl Med 2023; 21: 150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Herbst RS, Morgensztern D, Boshoff C. The biology and management of non-small cell lung cancer. Nature 2018; 553: 446–454. [DOI] [PubMed] [Google Scholar]
- 5. Owusu BY, Galemmo R, Janetka J, et al. Hepatocyte growth factor, a key tumor-promoting factor in the tumor microenvironment. Cancers (Basel) 2017; 9: 35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Ma PC, Maulik G, Christensen J, Salgia R. c-Met: structure, functions and potential for therapeutic inhibition. Cancer Metastasis Rev 2003; 22: 309–325. [DOI] [PubMed] [Google Scholar]
- 7. Drilon A, Cappuzzo F, Ou SHI, et al. Targeting MET in lung cancer: will expectations finally be MET? J Thorac Oncol 2017; 12(1): 15–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Remon J, Hendriks LEL, Mountzios G, et al. MET alterations in NSCLC-current perspectives and future challenges. J Thorac Oncol 2023; 18: 419–435. [DOI] [PubMed] [Google Scholar]
- 9. Lu X, Peled N, Greer J, et al. MET Exon 14 mutation encodes an actionable therapeutic target in lung adenocarcinoma. Cancer Res 2017; 77: 4498–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Socinski MA, Pennell NA, Davies KD. MET exon 14 skipping mutations in non–small-cell lung cancer: an overview of biology, clinical outcomes, and testing considerations. JCO Precis Oncol 2021; 5: PO.20.00516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. 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: 737–741. [DOI] [PubMed] [Google Scholar]
- 12. Heydt C, Ihle MA, Merkelbach-Bruse S. Overview of molecular detection technologies for MET in lung cancer. Cancers (Basel) 2023; 15: 2932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Zheng Z, Liebers M, Zhelyazkova B, et al. Anchored multiplex PCR for targeted next-generation sequencing. Nat Med 2014; 20: 1479–1484. [DOI] [PubMed] [Google Scholar]
- 14. Recondo G, Che J, Jänne PA, et al. Targeting MET dysregulation in cancer. Cancer Discov 2020; 10: 922–934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Mignard X, Ruppert A-M, 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 2018; 13: 1962–1967. [DOI] [PubMed] [Google Scholar]
- 16. Bahcall M, Paweletz CP, Kuang Y, et al. Combination of type I and type II MET tyrosine kinase inhibitors as therapeutic approach to prevent resistance. Mol Cancer Ther 2022; 21: 322–335. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Zhang J, Yang PL, Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer 2009; 9: 28–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Klempner SJ, Borghei A, Hakimian B, et al. Intracranial Activity of Cabozantinib in MET Exon 14-Positive NSCLC with Brain Metastases. J Thorac Oncol 2017; 12: 152–156. [DOI] [PubMed] [Google Scholar]
- 19. Scagliotti G, Moro-Sibilot D, Kollmeier J, et al. A randomized-controlled phase 2 study of the MET antibody emibetuzumab in combination with erlotinib as first-line treatment for EGFR Mutation-Positive NSCLC Patients. J Thorac Oncol 2020; 15: 80–90. [DOI] [PubMed] [Google Scholar]
- 20. Gelsomino F, Facchinetti F, Haspinger ER, et al. Targeting the MET gene for the treatment of non-small-cell lung cancer. Crit Rev Oncol Hematol 2014; 89: 284–299. [DOI] [PubMed] [Google Scholar]
- 21. Spigel DR, Ervin TJ, Ramlau RA, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Oh SY, Lee YW, Lee EJ, et al. Preclinical study of a biparatopic METxMET antibody-drug conjugate, REGN5093-M114, overcomes MET-driven acquired resistance to EGFR TKIs in EGFR-mutant NSCLC. Clin Cancer Res 2023; 29: 221–232. [DOI] [PubMed] [Google Scholar]
- 23. Leslie M. Amivantamab emerges positively from Its “Chrysalis.” Cancer Discov 2022; 12: OF2. [DOI] [PubMed] [Google Scholar]
- 24. Passaro A, Jänne PA, Peters S. Antibody-drug conjugates in lung cancer: recent advances and implementing strategies. J Clin Oncol 2023; 41: 3747–3761. 10.1200/JCO.23.00013. [DOI] [PubMed] [Google Scholar]
- 25. Fu Z, Li S, Han S, et al. Antibody drug conjugate: the “biological missile” for targeted cancer therapy. Signal Transduct Target Ther 2022; 7: 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Camidge DR, Morgensztern D, Heist RS, et al. Phase I Study of 2- or 3-week dosing of telisotuzumab vedotin, an antibody-drug conjugate targeting c-Met, monotherapy in patients with advanced non-small cell lung carcinoma. Clin Cancer Res 2021; 27: 5781–5792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Camidge DR, Barlesi F, Goldman JW, et al. Phase Ib study of telisotuzumab vedotin in combination with erlotinib in patients with c-Met protein-expressing non-small-cell lung cancer. J Clin Oncol 2023; 41: 1105–1115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Camidge DR, Barlesi F, Goldman JW, et al. Telisotuzumab vedotin (Teliso-V) monotherapy in patients (pts) with previously treated c-Met–overexpressing (OE) advanced non-small cell lung cancer (NSCLC). American Society of Clinical Oncology (ASCO) Abstract 9016 2022.
- 29. Tan E-H, Lim W-T, Ahn M-J, et al. Phase 1b trial of ficlatuzumab, a humanized hepatocyte growth factor inhibitory monoclonal antibody, in combination with gefitinib in asian patients with NSCLC. Clin Pharmacol Drug Dev 2018; 7: 532–542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Mok TSK, Geater SL, Su W-C, et al. A randomized phase 2 study comparing the combination of ficlatuzumab and gefitinib with gefitinib alone in Asian patients with advanced stage pulmonary adenocarcinoma. J Thorac Oncol 2016; 11: 1736–1744. [DOI] [PubMed] [Google Scholar]
- 31. Spigel DR, Edelman MJ, O’Byrne K, et al. Results from the phase III randomized trial of onartuzumab plus erlotinib versus erlotinib in previously treated stage IIIB or IV non-small-cell lung cancer: METLung. J Clin Oncol 2017; 35: 412–420. [DOI] [PubMed] [Google Scholar]
- 32. 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: 1666–16671. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Awad MM, Oxnard GR, Jackman DM, et al. MET Exon 14 mutations in non-small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression. J Clin Oncol 2016; 34: 721–730. [DOI] [PubMed] [Google Scholar]
- 34. Cappuzzo F, Marchetti A, Skokan M, et al. Increased MET gene copy number negatively affects survival of surgically resected non-small-cell lung cancer patients. J Clin Oncol 2009; 27: 1667–1674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Yin X, Zhang T, Su X, et al. Relationships between Chromosome 7 Gain, MET Gene Copy Number Increase and MET Protein Overexpression in Chinese Papillary Renal Cell Carcinoma Patients. PLoS One 2015; 10: e0143468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Lee HE, Kim MA, Lee HS, et al. MET in gastric carcinomas: comparison between protein expression and gene copy number and impact on clinical outcome. Br J Cancer 2012; 107: 325–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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: 1293–1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Chinese Society of Pathology, Pathology Quality Control Center, Lung Cancer Group of Chinese Medical Association Chinese Society of Oncology, China Anti-cancer Association Chinese Society of Lung Cancer, Chinese Thoracic Oncology Group. [Chinese expert consensus on clinical practice of MET detection in non-small cell lung cancer]. Zhonghua Bing Li Xue Za Zhi 2022; 51: 1094–103. [DOI] [PubMed] [Google Scholar]
- 39. Camidge DR, Ou S-HI, Shapiro G, et al. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). JCO 2014; 32: 8001–8001. [Google Scholar]
- 40. Camidge DR, Otterson GA, Clark JW, et al. Crizotinib in patients (pts) with MET-amplified non-small cell lung cancer (NSCLC): updated safety and efficacy findings from a phase 1 trial. JCO 2018; 36: 9062–9062. [Google Scholar]
- 41. Wolf J, Seto T, Han J-Y, et al. Capmatinib in MET exon 14-mutated or MET-amplified non-small-cell lung cancer. N Engl J Med 2020; 383: 944–957. [DOI] [PubMed] [Google Scholar]
- 42. Schuler MH, Berardi R, Lim W-T, et al. Phase (Ph) I study of the safety and efficacy of the cMET inhibitor capmatinib (INC280) in patients (pts) with advanced cMET+ non-small cell lung cancer (NSCLC). JCO 2016; 34: 9067–9067. [Google Scholar]
- 43. Network NCC. NCCN Clinical Practice Guidelines in Oncology – Non-Small Cell Lung Cancer (Version 4.2024) 2024.
- 44. Garber K. MET inhibitors start on road to recovery. Nat Rev Drug Discov 2014; 13: 563–565. [DOI] [PubMed] [Google Scholar]
- 45. Paik PK, Drilon A, Fan P-D, et al. Response to MET inhibitors in patients with stage IV lung adenocarcinomas harboring MET mutations causing exon 14 skipping. Cancer Discov 2015; 5: 842–849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. 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: 47–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Jamme P, Fernandes M, Copin M-C, 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: 741–751. [DOI] [PubMed] [Google Scholar]
- 48. Heist RS, Sequist LV, Borger D, et al. Acquired resistance to crizotinib in NSCLC with MET exon 14 skipping. J Thorac Oncol 2016; 11: 1242–1245. [DOI] [PubMed] [Google Scholar]
- 49. Li H, Chen Z, Chen N, et al. Applications of lung cancer organoids in precision medicine: from bench to bedside. Cell Commun Signal 2023; 21: 350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015; 348: 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Wolf J, Souquet P-J, Goto K, et al. Improved survival outcomes in patients with MET-dysregulated advanced NSCLC treated with MET inhibitors: results of a multinational retrospective chart review. Clin Lung Cancer 2023: S1525-7304(23)00172-9. [DOI] [PMC free article] [PubMed]
- 52. Negrao MV, Skoulidis F, Montesion M, et al. Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer. J Immunother Cancer 2021; 9: e002891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Veglia F, Perego M, Gabrilovich D. Myeloid-derived suppressor cells coming of age. Nat Immunol 2018; 19: 108–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Zhang Y, Yang Q, Zeng X, et al. MET Amplification attenuates lung tumor response to immunotherapy by inhibiting STING. Cancer Discov 2021; 11: 2726–2737. [DOI] [PubMed] [Google Scholar]
- 55. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of immune monitoring for cancer immunotherapies in the era of checkpoint inhibition. Clin Cancer Res 2016; 22: 1865–1874. [DOI] [PubMed] [Google Scholar]
- 56. Lo Presti E, Dieli F, Meraviglia S. Tumor-infiltrating γδ T lymphocytes: pathogenic role, clinical significance, and differential programing in the tumor microenvironment. Front Immunol 2014; 5: 607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Yoshimura K, Inoue Y, Tsuchiya K, et al. Elucidation of the relationships of MET protein expression and gene copy number status with PD-L1 expression and the immune microenvironment in non-small cell lung cancer. Lung Cancer 2020; 141: 21–31. [DOI] [PubMed] [Google Scholar]
- 58. Pan Y, Yu Y, Wang X, et al. Tumor-associated macrophages in tumor immunity. Front Immunol 2020; 11: 583084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Jackute J, Zemaitis M, Pranys D, et al. Distribution of M1 and M2 macrophages in tumor islets and stroma in relation to prognosis of non-small cell lung cancer. BMC Immunol 2018; 19: 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Sumitomo R, Hirai T, Fujita M, et al. M2 tumor-associated macrophages promote tumor progression in non-small-cell lung cancer. Exp Ther Med 2019; 18: 4490–4498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Nishikoba N, Kumagai K, Kanmura S, et al. HGF-MET Signaling shifts M1 macrophages toward an M2-like phenotype through PI3K-mediated induction of arginase-1 expression. Front Immunol 2020; 11: 2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Kumar V, Donthireddy L, Marvel D, et al. Cancer-associated fibroblasts neutralize the anti-tumor effect of CSF1 receptor blockade by inducing PMN-MDSC infiltration of tumors. Cancer Cell 2017; 32: 654–668.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Palle J, Hirsch L, Lapeyre-Prost A, et al. Targeting HGF/c-Met axis decreases circulating regulatory T cells accumulation in gastric cancer patients. Cancers (Basel) 2021; 13: 5562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Sakaguchi S, Wing K, Onishi Y, et al. Regulatory T cells: how do they suppress immune responses? Int Immunol 2009; 21: 1105–1111. [DOI] [PubMed] [Google Scholar]
- 65. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity 2010; 32: 593–604. [DOI] [PubMed] [Google Scholar]
- 66. Gaggero S, Witt K, Carlsten M, et al. Cytokines orchestrating the natural killer-myeloid cell crosstalk in the tumor microenvironment: implications for natural killer cell-based cancer immunotherapy. Front Immunol 2020; 11: 621225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Schoenfeld AJ, Rizvi H, Bandlamudi C, et al. Clinical and molecular correlates of PD-L1 expression in patients with lung adenocarcinomas. Ann Oncol 2020; 31: 599–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Peng S, Wang R, Zhang X, et al. EGFR-TKI resistance promotes immune escape in lung cancer via increased PD-L1 expression. Mol Cancer 2019; 18: 165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. 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: 2085–2091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Gordon SR, Maute RL, Dulken BW, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017; 545: 495–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Mondal P, Kaur B, Natesh J, et al. The emerging role of miRNA in the perturbation of tumor immune microenvironment in chemoresistance: therapeutic implications. Semin Cell Dev Biol 2022; 124: 99–113. [DOI] [PubMed] [Google Scholar]
- 72. Qureshi OS, Zheng Y, Nakamura K, et al. Trans-endocytosis of CD80 and CD86: a molecular basis for the cell-extrinsic function of CTLA-4. Science 2011; 332: 600–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. J K, J H, J L, S FL, Yh K. Recent advances in tumor microenvironment-targeted nanomedicine delivery approaches to overcome limitations of immune checkpoint blockade-based immunotherapy. J Control Release 2021; 332: 109–126. [DOI] [PubMed] [Google Scholar]
- 74. Sugiyama D, Hinohara K, Nishikawa H. Significance of regulatory T cells in cancer immunology and immunotherapy. Exp Dermatol 2023; 32: 256–263. [DOI] [PubMed] [Google Scholar]
- 75. Bronte V, Brandau S, Chen S-H, et al. Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 2016; 7: 12150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Gandhi L, Rodríguez-Abreu D, Gadgeel S, et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N Engl J Med 2018; 378: 2078–2092. [DOI] [PubMed] [Google Scholar]
- 77. Otano I, Ucero AC, Zugazagoitia J, et al. At the crossroads of immunotherapy for oncogene-addicted subsets of NSCLC. Nat Rev Clin Oncol 2023; 20: 143–159. [DOI] [PubMed] [Google Scholar]
- 78. Mazieres J, Drilon A, Lusque A, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann Oncol 2019; 30: 1321–1328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Reis H, Metzenmacher M, Goetz M, et al. MET expression in advanced non-small-cell lung cancer: effect on clinical outcomes of chemotherapy, targeted therapy, and immunotherapy. Clin Lung Cancer 2018; 19: e441–e463. [DOI] [PubMed] [Google Scholar]
- 80. Guisier F, Dubos-Arvis C, Viñas F, et al. Efficacy and safety of anti-PD-1 immunotherapy in patients with advanced NSCLC With BRAF, HER2, or MET mutations or RET translocation: GFPC 01-2018. J Thorac Oncol 2020; 15: 628–636. [DOI] [PubMed] [Google Scholar]
- 81. Guisier F, Descourt R, Babey H, et al. Brief report: first-line pembrolizumab in metastatic non-small cell lung cancer habouring MET Exon 14 skipping mutation and PD-L1 ⩾50% (GFPC 01-20 Study). Clin Lung Cancer 2022; 23: e545–e549. [DOI] [PubMed] [Google Scholar]
- 82. Gu L, Wei X, Zhang Z, et al. Treatment response to immunotherapy after crizotinib resistance in a patient with pulmonary sarcomatoid carcinoma harboring MET Exon 14 skipping mutation: a case report. Clin Med Insights Oncol 2022; 16: 11795549211067185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Blasi M, Jonas K, Lüders H, et al. First-line immunotherapy for lung cancer with MET exon 14 skipping and the relevance of TP53 mutations. Eur J Cancer (Oxford, England : 1990) 2024; 199: 113556. [DOI] [PubMed] [Google Scholar]
- 84. Qin H, Yan H, Chen Y, et al. Clinical outcomes for immune checkpoint inhibitors plus chemotherapy in non-small-cell lung cancer patients with uncommon driver gene alterations. BMC Cancer 2024; 24: 952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Glodde N, Bald T, van den Boorn-Konijnenberg D, et al. Reactive neutrophil responses dependent on the receptor tyrosine kinase c-MET limit cancer immunotherapy. Immunity 2017; 47: 789–802.e9. [DOI] [PubMed] [Google Scholar]
- 86. Spigel DR, Reynolds C, Waterhouse D, et al. Phase 1/2 study of the safety and tolerability of nivolumab plus crizotinib for the first-line treatment of anaplastic lymphoma kinase translocation – positive advanced non-small cell lung cancer (CheckMate 370). J Thorac Oncol 2018; 13: 682–688. [DOI] [PubMed] [Google Scholar]
- 87. Yang JC-H, Shepherd FA, Kim D-W, et al. Osimertinib plus durvalumab versus osimertinib monotherapy in EGFR T790M-positive NSCLC following previous EGFR TKI therapy: CAURAL brief report. J Thorac Oncol 2019; 14: 933–939. [DOI] [PubMed] [Google Scholar]
- 88. Mok TSK, Cortinovis DL, Majem M, et al. Efficacy and safety of capmatinib plus pembrolizumab in treatment (tx)-naïve patients with advanced non–small cell lung cancer (NSCLC) with high tumor PD-L1 expression: results of a randomized, open-label, multicenter, phase 2 study. JCO 2022; 40: 9118–9118. [Google Scholar]
- 89. Okajima D, Yasuda S, Maejima T, et al. Datopotamab deruxtecan, a Novel TROP2-directed antibody-drug conjugate, demonstrates potent antitumor activity by efficient drug delivery to tumor cells. Mol Cancer Ther 2021; 20: 2329–2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Goto Y, Su W-C, Levy BP, et al. TROPION-Lung02: Datopotamab deruxtecan (Dato-DXd) plus pembrolizumab (pembro) with or without platinum chemotherapy (Pt-CT) in advanced non-small cell lung cancer (aNSCLC). JCO 2023; 41: 9004–9004. [Google Scholar]
- 91. Borghaei H, Waqar SN, Bruno DS, et al. TROPION-Lung04: Phase 1b, multicenter study of datopotamab deruxtecan (Dato-DXd) in combination with immunotherapy ± carboplatin in advanced/metastatic non-small cell lung cancer (mNSCLC). JCO 2023; 41: TPS3158–TPS3158. [Google Scholar]
- 92. Levy BP, Reck M, Yang JC-H, et al. Datopotamab deruxtecan (Dato-DXd) plus pembrolizumab in treatment-naive advanced/metastatic (adv/met) non–small cell lung cancer (NSCLC) with PD-L1 ⩾ 50% and without actionable genomic alterations. JCO 2022; 40: TPS3162–TPS3162. [Google Scholar]
- 93. Békés M, Langley DR, Crews CM. PROTAC targeted protein degraders: the past is prologue. Nat Rev Drug Discov 2022; 21: 181–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Li X, Song Y. Proteolysis-targeting chimera (PROTAC) for targeted protein degradation and cancer therapy. J Hematol Oncol 2020; 13: 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Dale B, Cheng M, Park K-S, et al. Advancing targeted protein degradation for cancer therapy. Nat Rev Cancer 2021; 21: 638–654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Bondeson DP, Smith BE, Burslem GM, et al. Lessons in PROTAC design from selective degradation with a promiscuous warhead. Cell Chem Biol 2018; 25: 78–87.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Li P, Jia C, Fan Z, et al. Discovery of novel exceptionally potent and orally active c-MET PROTACs for the treatment of tumors with MET alterations. Acta Pharm Sin B 2023; 13: 2715–2735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Zhao H-Y, Xin M, Zhang S-Q. Progress of small molecules for targeted protein degradation: PROTACs and other technologies. Drug Dev Res 2023; 84: 337–394. [DOI] [PubMed] [Google Scholar]
- 99. Jaynes JM, Sable R, Ronzetti M, et al. Mannose receptor (CD206) activation in tumor-associated macrophages enhances adaptive and innate antitumor immune responses. Sci Transl Med 2020; 12: eaax6337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Zhou Y, Fei M, Zhang G, et al. Blockade of the phagocytic receptor MerTK on tumor-associated macrophages enhances P2X7R-dependent sting activation by tumor-derived cGAMP. Immunity 2020; 52: 357–373.e9. [DOI] [PubMed] [Google Scholar]
- 101. Granhøj JS, Witness Præst Jensen A, Presti M, et al. Tumor-infiltrating lymphocytes for adoptive cell therapy: recent advances, challenges, and future directions. Expert Opin Biol Ther 2022; 22: 627–641. [DOI] [PubMed] [Google Scholar]
- 102. Caushi JX, Zhang J, Ji Z, et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 2021; 596: 126–132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Galvano A, Gristina V, Malapelle U, et al. The prognostic impact of tumor mutational burden (TMB) in the first-line management of advanced non-oncogene addicted non-small-cell lung cancer (NSCLC): a systematic review and meta-analysis of randomized controlled trials. ESMO Open 2021; 6: 100124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Min J, Long C, Zhang L, et al. c-Met specific CAR-T cells as a targeted therapy for non-small cell lung cancer cell A549. Bioengineered 2022; 13: 9216–9232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Houot R, Schultz LM, Marabelle A, et al. T-cell-based Immunotherapy: adoptive cell transfer and checkpoint inhibition. Cancer Immunol Res 2015; 3: 1115–1122. [DOI] [PubMed] [Google Scholar]
- 106. Liu E, Marin D, Banerjee P, et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N Engl J Med 2020; 382: 545–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Bald T, Krummel MF, Smyth MJ, et al. The NK cell-cancer cycle – advances and new challenges in NK cell-based immunotherapies. Nat Immunol 2020; 21: 835–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Dudley JC, Lin M-T, Le DT, et al. Microsatellite instability as a biomarker for PD-1 blockade. Clin Cancer Res 2016; 22: 813–820. [DOI] [PubMed] [Google Scholar]
- 109. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017; 357: 409–413. 10.1126/science.aan6733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Luchini C, Bibeau F, Ligtenberg MJL, et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol 2019; 30: 1232–1243. [DOI] [PubMed] [Google Scholar]
- 111. Lan B, Ma C, Zhang C, et al. Association between PD-L1 expression and driver gene status in non-small-cell lung cancer: a meta-analysis. Oncotarget 2018; 9: 7684–7699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Palmer C, Diehn M, Alizadeh AA, et al. Cell-type specific gene expression profiles of leukocytes in human peripheral blood. BMC Genom 2006; 7: 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Greenplate AR, Johnson DB, Ferrell PB, et al. Systems immune monitoring in cancer therapy. Eur J Cancer 2016; 61: 77–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. 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: 1391–1399. 10.1001/jama.2019.3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Bar J, Cai MH, Choi YC, et al. Prevalence, molecular characterization, and prognosis of MET–overexpressing non-small cell lung cancer (NSCLC) in a real-world patient cohort. ESMO Congress 2023: 1397P. [Google Scholar]
- 116. Lu S, Fang J, Li X, et al. Once-daily savolitinib in Chinese patients with pulmonary sarcomatoid carcinomas and other non-small-cell lung cancers harbouring MET exon 14 skipping alterations: a multicentre, single-arm, open-label, phase 2 study. Lancet Respir Med 2021; 9: 1154–1164. [DOI] [PubMed] [Google Scholar]
- 117. 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: 931–943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Yu Y, Zhou J, Li X, et al. Gumarontinib in patients with non-small-cell lung cancer harbouring MET exon 14 skipping mutations: a multicentre, single-arm, open-label, phase 1b/2 trial. eClinicalMedicine 2023; 59: 101952. [DOI] [PMC free article] [PubMed] [Google Scholar]


