Next Article in Journal
Liposome-Based Carriers for CRISPR Genome Editing
Previous Article in Journal
Evaluating the Molecular Properties and Function of ANKHD1, and Its Role in Cancer
Previous Article in Special Issue
Therapeutical Options in ROS1—Rearranged Advanced Non Small Cell Lung Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Real-World Experience in Treatment of Patients with Non-Small-Cell Lung Cancer with BRAF or cMET Exon 14 Skipping Mutations

1
Department of Medical Oncology, University Clinic Golnik, 4204 Golnik, Slovenia
2
Medical Faculty Ljubljana, University of Ljubljana, 1000 Ljubljana, Slovenia
3
The Legacy Heritage Cancer Center & Dr. Larry Norton Institute, Soroka Medical Center, Ben Gurion University, Beer Sheva 84105, Israel
4
Department of Oncology and Radiotherapy and Early Phase Clinical Trials Centre, Medical University of Gdańsk, 80-210 Gdańsk, Poland
5
Department for Respiratory Diseases Jordanovac, University Hospital Centre Zagreb, 10 000 Zagreb, Croatia
6
Clinical Trials Unit, Fourth Oncology Department, Henry Dunant Hospital Center, 115 26 Athens, Greece
7
Department of Lung Cancer and Chest Tumours, The Maria Skłodowska-Curie National Research Institute of Oncology, 00-001 Warsaw, Poland
8
Department of Medical Oncology, CHUPorto—University Hospitalar Center of Porto, 4099-001 Porto, Portugal
9
Oncology Unit, 3rd Department of Medicine, “Sotiria” Hospital for Diseases of the Chest, National and Kapodistrian University of Athens, 106 79 Athens, Greece
10
Department of Oncology, Bnai-Zion Medical Center, 47 Golomb Avenue, Haifa 31048, Israel
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(16), 12840; https://doi.org/10.3390/ijms241612840
Submission received: 15 July 2023 / Revised: 12 August 2023 / Accepted: 15 August 2023 / Published: 16 August 2023

Abstract

:
BRAF and cMET exon 14 skipping are rare mutations of NSCLC. The treatment sequence in these cases for the first and second line is not clear. An international registry was created for patients with advanced NSCLC harboring BRAF or cMET exon 14 skipping mutations, diagnosed from January 2017 to June 2022. Clinicopathological and molecular data and treatment patterns were recorded. Data on 58 patients, from eight centers across five countries, were included in the final analysis. We found that 40 patients had the cMET exon 14 skipping mutation and 18 had the BRAF V600E mutation. In total, 53 and 28 patients received first- and second-line treatments, respectively, among which 52.8% received targeted therapy (TT) in the first line and 53.5% in the second line. The overall response rate (ORR) and disease control rate (DCR) for first-line treatment with TT vs. other treatment such as immune checkpoint inhibitors ± chemotherapy (IO ± CT) were 55.6% vs. 21.7% (p = 0.0084) and 66.7% vs. 39.1% (p = 0.04), respectively. The type of treatment in first-line TT vs. other affected time to treatment discontinuation (TTD) was 11.6 m vs. 4.6 m (p= 0.006). The overall survival for the whole group was 15.4 m and was not statistically affected by the type of treatment (19.2 m vs. 13.5 m; p = 0.83).

1. Introduction

Lung cancer (LC) is the main cause of mortality worldwide and represents approximately 18% of total deaths from cancer [1,2]. Non-small-cell lung cancer (NSCLC) accounts for about 85% of lung cancer and, despite improvements in early detection, NSCLC is often diagnosed at advanced stages, where patients have a poor prognosis [3,4].
The recent advances in cancer molecular biology have deciphered the oncogenic processes in many types of malignant diseases, including LC. The Lung Cancer Mutation Consortium has elucidated the molecular heterogeneity of NSCLC, discovering molecular alterations in key regulatory pathways that spearhead the malignant process, which, in turn, makes their effective targeting possible and the consequent impairment of cancer growth [5,6]. These molecular alterations are associated with driver mutations. In NSCLC, there are high rates of somatic mutations and genomic rearrangements. Although less frequent, rare mutations are a growing concern. The latest version (3.2022) of the National Comprehensive Cancer Network (NCCN) guidelines for NSCLC recommends that it is important to detect less frequent epidermal growth factor receptor (EGFR) mutations, such as point mutations in exon 18 (G719X), exon 19 insertions, substitution mutations in exon 20 (S768I), exon 20 insertions and substitution mutations in exon 21 (L861Q); human epidermal growth factor 2 (HER2) mutations, such as exon 20 insertions and exon 19 and exon 20 substitutions; Kirsten rat sarcoma virus (KRAS) p.G12C single-nucleotide variations; anaplastic lymphoma kinase (ALK), c-ROS proto-oncogene (ROS1) and rearranged during transfection (RET) rearrangements; B-type Raf kinase (BRAF) V600E mutations; and neurotrophic tyrosine kinase (NTRK) 1/2/3 gene fusion mutations and mesenchymal epithelial transition factor (MET) exon 14 skipping mutations [7,8].
Nowadays, it is possible to identify driver mutations through different methods of molecular analysis. Advanced assays with hybrid capture next-generation sequencing (NGS) enable us to discover a wide range of common and less common mutations, but they are not universally used. Instead, other techniques are routinely applied, such as fluorescence in situ hybridization (FISH), immunohistochemistry (IHC), Sanger sequencing, point-of-care testing or laboratory-developed tests based on PCR technology [9,10]. BRAF and cMET exon 14 skipping mutations are very rare genetic alterations in NSCLC, with incidences of 1–5% and 2–4%, respectively [10].
Genetic assessment in LC varies among different countries; therefore, it is necessary to obtain a comprehensive profiling of genetic mutations of the patients to guide diagnosis and treatment approaches for NSCLC [11]. As a result, many patients could become eligible for targeted therapies (TTs), such as specific monoclonal antibodies or tyrosine kinase inhibitors (TKIs) [12]. Using TTs in the first-line setting has been shown to bring about an improvement in progression-free survival (PFS) and overall survival (OS) in common mutations like EGFR and ALK [13,14].
The treatment paradigm in other less common NSCLC mutations, like MET and BRAF, is less clear due to their rarity and the absence of phase III clinical trials in this subset of the patient population comparing TTs to the standard of care (SOC) [15]. Both mutations affect the downstream signal pathways. MET mutation results in overexpression of its gene product—hepatocyte growth factor receptor (HGFR) [16]. Meanwhile, BRAF activates the MAPK/extracellular-signal-regulated kinase (ERK), leading to cell proliferation, migration, invasion and metastasis [17]. Treatment strategies aim to interfere with these downstream pathways [18,19].
TTs for MET mutations are divided into three types: type I and II are in clinical use, while type III inhibitors are still in preclinical experimental phase [20]. Type I inhibitors are tyrosine kinase inhibitors (TKIs), which bind competitively to the ATP pocket of the receptor, e.g., crizitonib (type Ia), capmatinib and tepotinip (type Ib). Type II inhibitors include cabozantinib, which binds to a hydrophobic pocket adjacent to the ATP binding site [20].
The phase I/II clinical trials GEOMETRY mono-1 [21] and VISION [22] have shown significant clinical activity in patients treated with capmatinib and tepotinib, respectively, including a tolerable toxicity profile. While in the GEOMETRY mono-1 trial, capmatinib was mainly active in treatment-naïve patients, tepotinib showed better antitumor activity in patients treated in advanced lines in the VISION trial [23]. Savolitinib is another MET TKI that has shown efficacy in both first- and second-line settings [24,25]. These MET TKIs have also shown some activity against brain metastasis based on their ability to cross the blood–brain barrier [26].
The use of immunotherapy (IO) as monotherapy or in combination with chemotherapy in first-line treatment in a group of NSCLC MET mutated patients is still controversial despite the high prevalence of positive PD-L1 [27]. This approach is based on small retrospective data collection, which has shown 16–36% response rates, PFS of 1.9–3.4 months and OS of 18 months [28,29,30,31].
Dabrafenib and vemurafenib are a novel generation of BRAF inhibitors [19]. Both drugs have shown antitumor activity in NSCLC patients with the BRAF V600 mutation when given as monotherapy [19].
Dabrafenib plus trametinib, a type of MEK inhibitor, when given in combination to NSCLC patients bearing the BRAF V600E mutation, has shown an ORR of 64%, PFS of 14.6 months and an OS of 24.6 months [32,33].
Regarding immunotherapy in this group of patients and based on the promising activity with IO plus BRAF targeted therapy in melanoma patients [34], the exact role of IO is still not well defined.
The place of IO treatment in the BRAF V600E-mutant NSCLC population is still unclear. Recently, only a few retrospective datasets reporting efficacy, such as longer response after treatment with atezolizumab plus chemotherapy, were published [35,36,37].
Further prospective and real-world data are needed to define the role of the best treatment modality in the first- and second-line settings in the two groups of NSCLC patients described above.
The present real-world trial compared TTs and other systemic treatments in terms of efficacy for the first-line setting in the two groups of mutations (BRAF, MET).

2. Results

2.1. Clinical and Demographic Characterization of the Cohorts

This data collection identified 133 patients with rare mutations, and further analysis was conducted on a subset of these patients, namely 58 patients who were found to have BRAF and cMET mutations. Of those, 17 (29%) had the BRAF V600E mutation, 1 (2%) had a BRAF translocation and 40 (69%) had the cMET exon 14 skipping mutation (Table 1).
There was no significant difference between the genders. Regarding a history of smoking, almost two-thirds (65%) of the patients had a smoking habit.
The majority of the patients (65%) had background diseases or other chronic conditions. The initial diagnosis stage of the patients included in the study revealed a diverse distribution. Among the participants, 1.7% were diagnosed at stages IB and IIIA, while 3.4% were at stage IIA; stage IIIB accounted for 5.2% of cases and no patients were diagnosed at stage IIIC. Additionally, the majority of the patients (65%) were at stage IVA and 17.2% were diagnosed at stage IVB.
The most common histopathological diagnosis was adenocarcinoma, accounting for 86% of cases. Genetic testing was performed using next-generation sequencing (NGS) in all 58 patients.
PD-L1 levels indicated that 18% of patients had a PD-L1 score below 1%, 34% of patients had a PD-L1 score of 1–49%, and 48% of patients had PD-L1 scores over 50%.
The ECOG performance status of the majority of patients (75%) was 1.
Among the patients with advanced disease, 93% had five or fewer metastatic sites. The most common sites of metastasis included the contralateral lung (50%), bone (45%), pleura (29%), adrenal gland (24%) and brain (21%).

2.2. Treatment and Follow-Ups among the Patients

Regarding treatment and follow-up among the patients (Table 2), 9% underwent surgery as part of their treatment, while 10% received stereotactic body radiation therapy (SBRT), and 7% underwent chemoradiotherapy (CRT) as a radical treatment approach. Consolidation with IO was administered to 2% of the patients.
In terms of the first-line treatment regimen, 47% received chemotherapy, while 53% were treated with targeted therapy.
The average number of treatment cycles was 6, ranging from 2.5 to 12 cycles.
Out of 18 patients with BRAF mutation, 1 patient did not receive any systemic therapy. Out of 17 patients treated, 14 patients received targeted therapy (TT) with dabrafenib and trametinib in the first-line setting with an ORR of 78% and DCR of 86%, and only 2 patients had progressive disease as the best response to TT. Meanwhile, 2 out of 3 patients who received either chemotherapy or therapy and progressed then received TT in the second line; the best responses were partial remission and stable disease.
In total, 40 patients were identified with the cMET exon 14 skipping mutation and 36 of them received systemic therapy. First-line treatment with TT was received by 15 patients (4 capmatinib, 10 crizotinib and 1 tepotinib with the following responses: partial response, stable disease and progressive disease in 53%, 13% and 7%, respectively). Meanwhile, 21 patients received either chemotherapy, chemo-IO or IO treatment in the first-line setting and, upon progression, 12 of those patients received second-line TT (2 capmatinib, 8 crizotinib and 2 tepotinib). The best responses in the second line to TT were partial response, stable disease and progressive disease in 58%, 9% and 33%, respectively.
The treatment-related adverse events (TRAEs) following targeted therapy in NSCLC patients were reported to be mostly of grade 1 and grade 2 (n = 54, 93.1%). The skin was affected in 43.1% (n = 25) of the study participants, followed by fatigue (n = 14, 24.1%) and diarrhea (n = 11, 18.9%).
There were 42 patients who discontinued first-line treatment; the reasons for discontinuation included death (17%), end of treatment (10%), and disease progression (74%). No patients discontinued treatment due to toxicity.
Progression of the disease was noted at different sites: in the central nervous system, there was disease recurrence in 3% of patients; in only distant sites, in 3% of patients; and in local/regional plus distant sites, in 47% of patients (the same result (47%) was also noted for local/regional progression only).
Among patients progressing after first-line treatment, 27% underwent local radical treatment, and 78% received second-line treatment.

2.3. First- and Second-Line Therapy

A statistical analysis was conducted to compare various factors and their associations with different treatment aspects. The number of treatment cycles differed significantly between the groups, with a median of 5.0 cycles (range 2.0–6.5) for the other treatment (mono-chemotherapy, mono-IO, platinum-doublet, chemo-IO) group and a median of 9.5 cycles (range 4.25–14.0) for the targeted therapy group (p = 0.01). For the first-line treatment, the median duration was 133.5 days (range 62.3–178.5) for the other treatment group, whereas it was 326 days (range 122–428) for the targeted therapy group (p = 0.003) (Table 3). However, there was no significant difference in the duration of second-line treatment between the two groups, with medians of 159 days (range 99.5–285.5) and 194 days (range 106–338) for the other treatment and targeted therapy group, respectively (p = 0.51).
Only one patient with the BRAF V600E mutation had initial brain metastases and was treated with stereotactic radiosurgery (SRS) prior to systemic therapy initiation. In total, 11 out of 40 patients with the cMET exon 14 skipping mutation were diagnosed with initial brain metastases, but only 5 of them received local treatment before systemic therapy initiation.
Significant differences were observed for the drug access type of the first-line treatment. Patients receiving other treatments uniformly received first-line therapy through the national reimbursement program, whereas out of the 28 patients receiving targeted therapy, 7% accessed the treatment through extended access programs, 32% through individual reimbursement, 43% through national reimbursement, and 18% through other means like out-of-pocket payment (p < 0.001).

2.4. Treatments

In the targeted therapy group, 16 patients (57%) died, and 12 patients (43%) were alive at the time of analysis (Table 4). Meanwhile, 15 patients (60%) who received other treatments in the first-line setting experienced death during the study period and 10 patients (40%) were alive at the time of analysis.

2.5. Patients’ Survival

The median overall durations of survival for the groups of patients receiving TT (such as biological drugs) or other treatments in the first-line setting were 19.2 months (2.7–35.7 months; 95% confidence interval (CI)) and 13.6 months (8.6–18.5 months; 95% CI), respectively (Table 5).
Overall, considering both groups, the median PFS was 7.9 months (95% CI; ranged from 5.6 to 10.2 months) (Table 5).

3. Discussion

The present multicenter real-world study of treatment outcomes in patients with NSCLC harboring rare genetic alterations, i.e., BRAF and cMET exon 14 skipping mutations, has clearly shown the benefit of using targeted therapies, preferably in the first-line setting. Both the response rates and survival are prolonged when targeted agents are used in comparison to other standard-of-care (SOC) therapies for non-oncogene-addicted NSCLC.
A clear advantage to the use of targeted therapy was seen with dabrafenib + trametinib therapy in the current BRAF V600E mutant NSCLC patient cohort. This TT resulted in an ORR and DCR of 78% and 86%, respectively. Most of the patients received this combination of TT in the first-line setting. About half of the patients bearing the cMET exon 14 skipping mutation received TT in first- and half in second-line therapy, and both demonstrated similar outcomes, with promising ORRs of 53% and 58%, and DCRs of 66% and 67%, respectively. Considering the similarities in response rates, the PFS and OS results were reported together and showed a substantial benefit in time to treatment discontinuation, which almost tripled from 133 to 326 days for patients treated with TT.
While the treatment of EGFR- and ALK-mutated NSCLC is well established, the treatment of other rare mutations is still under exploration. Since both types of molecular alterations studied in this retrospective analysis are rare, occurring in about 3–5% (BRAF V 6000E mutation) and 2–4% of NSCLC (MET exon 14 skipping mutation), robust and large-scale data on the characteristic and treatment of patients are missing, as are phase III randomized controlled trials [38,39].
Comparable to the pivotal phase II trial assessing both first- and further-line targeted therapy with BRAF and MEK inhibitors dabrafenib and trametinib, pretreated patients had an ORR of 68%, mPFS of 10.2 months and mOS of 18.2 months, whereas patients receiving the targeted combination in the first-line setting had an ORR of 64%, mPFS of 10.8 months and mOS of 17.3 months. It should be noted, however, that at the time this study was conducted, treatment with ICI was not yet a SOC for patients with NSCLC, which could partially alter the results [40]. As for the MET exon 14 skipping mutation, more tyrosine kinase targeted agents were studied in the phase II trials, such as crizotinib, capmatinib, tepotinib and savolitinib, and those trials displayed positive results, with ORRs spanning from 32% to 68%, mPFS from 6.8 months to 9.7 months and mOS reaching up to 24.6 months [21,41,42,43]. Further trials are also needed to assess whether patients with cMET amplifications and overexpression will derive the same benefit as patients with the exon 14 skipping mutation, since initial signals show that responses are lower and survival is shorter when using MET TKIs [44]. A recent meta-analysis showed a clear benefit of MET TKIs when treating patients diagnosed with advanced NSCLC with the MET exon 14 skipping mutation, resulting in an ORR of 39% and DCR of 78%; when taken together with other MET alterations, the ORR and DCR were 28% and 69%, respectively [18].
Retrospective data have shown a quite consistent benefit of the dabrafenib + trametinib combination treatment for BRAF V600E-positive NSCLC patients, such as our data, with an ORR and DCR of 53% and 85%, respectively, mPFS ranging from 5.0 to 17.5 months and mOS from 10.8 to 22.5 months [33,45,46].
As for patients with the cMET exon 14 skipping mutation, retrospective real-world data show promising results with either crizotinib treatment (mPFS 12.4 months, mOS 22.8 months) or capmatinib therapy (mPFS 9.5 months, mOS 18.2 months) [47,48]. Moreover, a substantial benefit is noted when using the MET inhibitor capmatinib in patients with brain metastases, demonstrating an 85% response rate both systemically and intracranially, even without the primary use of radiotherapy [49]. These data are further confirmed by the meta-analysis, which proved a high intracranial response rate of 95% [18].
New treatment options are underway, mostly phase I/II trials assessing the efficacy of dual or triple inhibition for BRAF, such as encorafenib + binimetinib or vemurafenib + cobimetinib ± atezolizumab [50]. The latter is already showing promising results, where 28 NSCLC BRAF mutant patients have shown an ORR of 18% and a median OS of 13.2 months [51]. The benefit was more prominent in the first-line treatment setting. Clinical trials assessing monoclonal antibodies are underway that target the BRAF mutation [52].
MET inhibitors are also found in further clinical development, both small-molecule TKIs such as merestinib, glesatinib and TPX-0022, as well as antibody-based therapies against MET/HGF, such as Sym-015, telisotuzumab vedotin or combining both MET TKIs with ICI therapy [52,53].
Some retrospective analyses report that patients with oncogene-addicted NSCLC with either a BRAF or MET exon 14 skipping mutation might respond to treatment with IO in monotherapy [54]. Although only about half of BRAF-mutant NSCLC patients carry the specific class I V600E mutation, treatment with IO is not detrimental in this patient population, as it is with chemotherapy [55].
The current study data showed that only 1 patient with the BRAF V600E mutation received IO monotherapy in the first line, with the best response of stable disease, and 4 patients received IO monotherapy in second line after progression on TT, with 3/4 achieving a clinical benefit. As for the MET exon 14 skipping mutation patients, 6/36 received IO monotherapy as the first-line treatment and there was a 67% DCR, whereas only 1 patient received mono-IO treatment after progression on TT, and that patient had a partial response.
Only one retrospective study showed poorer outcomes, which were noted especially in the class I cohort as opposed to the class II and III (non-V600E mutant) cohort, where they did not see a clear benefit of IO in most patients with BRAF alterations. There were ORRs of 9% and 26% for class I and II/III mutations and a higher hazard ratio (HR) for deceased patients who were ever treated with IO [56].
On the other hand, other retrospective analyses, such as the multicenter ImmunoTarget study, showed a 28% and 56% ORR and DCR, respectively, amongst patients with BRAF mutations treated with IO. Additionally, those patients achieved an mPFS of 3.1 months and mOS of 13.6 months [57]. Similarly, another analysis confirmed an ORR ranging from 25% to 33% for patients with class I and II/III BRAF-mutant tumors, and patients experienced significantly longer mOS when treated with IO (from not reported to 21.1 months). The same study also showed that there was a high prevalence of PD-L1 high expressers of 42% in this population [58]. The most recent series showed an ORR and DCR of 30% and 60%, mPFS of 5 months and mOS of 22.5 months for the BRAF V600E group of patients [59]. Another reason for higher IO efficacy in BRAF-mutant NSCLC, as opposed to EGFR or ALK, is the higher tumor mutational burden (TMB) and more frequent smoking status, as almost half of the BRAF-mutant NSCLC patients are found to be smokers [60]. Similarly, this was shown in the present analysis, with 65% of patients either current or former smokers.
In total, 50% of patients with the MET exon 14 skipping mutation had DC (with only 16% having an OR) in the ImmunoTarget study, with an mPFS and mOS of 3.4 and 18.4 months, respectively [57]. Another case series showed prolonged responses when treatment with IO was applied for patients with the MET exon 14 skipping mutation, with 6/13 prolonged responses, which were maintained for 18–49 months [60]. Furthermore, similar series have shown a moderate ORR with IO monotherapy, ranging from 12% to 36%, but with a DCR of up to 72% and mOS from 13.4 to 18.2 months [56,61]. In the post hoc analysis of the GEOMETRY trial, it was shown that patients previously receiving IO were more sensitive to capmatinib treatment versus those that received chemotherapy in prior lines (64% vs. 32%), which led to the conclusion that this is also an IO-sensitive disease [21].
This study’s limitations arose by virtue of conducting a retrospective analysis of data collected at separate medical centers in different countries, which may have varying access to therapies, clinical trials, etc., along with different health policies.
The weak points in targeted therapy for this type of NSCLC arise from the fact that these two mutations (BRAF and cMET) are very rare; therefore, experience is limited (due to the few patients) and insufficient to provide a clear-cut and unambiguous answer yet to whether first-line treatment in these cases should be targeted therapy or chemotherapy. The reports that there is a clinical response to targeted therapy, immunotherapy in patients bearing these mutations give an indication that we should continue offering this type of treatment. In addition, due to the low incidence and hence the difficulty in recruiting participants to clinical trials both for phase I and phase II, treatments of these rare mutations may eventually be recommended based on phase I/II results due to a lack of phase III clinical trials. Additional difficulty in providing a standard of care for the abovementioned patients arises from the fact that each oncologist has individual experience, though it is usually very limited due to the scarcity of these patients [62].
The recent NCCN Clinical Practice Guidelines in Oncology for Non-Small-Cell Lung Cancer (Version 3.2023, 13 April 2023) [62] detail preferred first-line and subsequent therapy following molecular testing results detecting these mutations. In case of positive BRAF V600E mutation, the suggestion is to administer dabrafenib in combination with trametinib as the first-line targeted therapy. For patients harboring the METex 14 skipping mutation, the preferred first-line therapy is capmatinib as a targeted therapy or tepotinib as a selective inhibitor (in cases where the genetic results are delayed, the recommended immediate planned therapy is systemic chemotherapy and immunotherapy). Obstacles in targeted therapy could account for the low response rate and drug-resistance mechanisms, which may lead to progression [63]. The indications for targeted therapy according to the NCCN Guidelines (Version 3.2023, Non-Small-Cell Lung Cancer) are molecular profiling in biopsies and positive detection of driver oncogenes such as the BRAF V600E mutation, METex14 skipping mutation, EGFR exon 19 deletion or exon 21 L858R, EGFR S768I L861Q and/or G719X, ALK rearrangement, ROS1 rearrangement or RET rearrangements [62].
Future prospects for the use of anticancer drugs in the treatment of NSCLC BRAF mutation or cMET exon 14 skipping mutation are under investigation and clinical trials testing the combination of targeted therapy and immunotherapy are needed. Ideally, large trials would provide better statistical interpretation of the results; however, they may take some time to achieve.
Immune checkpoint inhibitor (ICI) therapy combined with MET TKI is an avenue that is being explored, such as capmatinib and anti-PD1 drugs, e.g., nivolumab [52,64].
An ICI combined regimen with chemotherapy might be a further prospective choice for BRAF V600E-mutated NSCLC [52]. A recent review suggests that a promising regimen for NSCLC bearing the BRAF V600E mutation is ICI (e.g., atezolizumab) plus chemotherapy [19].
BRAF inhibitor targeted therapies using dabrafenib in combination are also being explored for the future.
In addition, more potent inhibitors could be developed [52]. The question remains whether the best treatment approach is sequential, or perhaps combining IO treatment with targeted therapies against BRAF or MET mutations would improve outcomes even further.

4. Materials and Methods

4.1. Study Design

We conducted a real-world retrospective, multicenter, non-interventional observational study, utilizing secondary data collection from medical records/applicable registries of patients with NSCLC harboring a rare targetable genetic alteration, diagnosed and having started treatment up to June 2022. Next-generation sequencing was employed in detecting the BRAF V600E mutation and MET exon 14 skipping mutation. Eight medical centers from five countries (Croatia, Greece, Israel, Slovenia and Poland) took part in this study. Data were reported on patients with the BRAF V600E mutation and MET exon 14 skipping mutation in this analysis. The characterization of patients comprised of gender, age, smoking habits, background disease, stage at diagnosis, ECOG performance status, genetic testing methodology, PD-L1 score and number of metastatic sites. Patient cohorts were devised according to the specific targeted therapy: first-line treatment of BRAF mutation, first-line treatment of cMET exon 14 skipping mutation and second-line treatment of cMET exon 14 skipping mutation.

4.2. Study Population

We sampled adults over the age of 18, who were diagnosed with NSCLC harboring a rare targetable genetic alteration (BRAF V600E mutation or MET exon 14 skipping mutation), diagnosed and having started treatment up to June 2022. Participants provided written informed consent for us to collect anonymized data for the study or the ethics committee provided a waiver for the informed consent form. Exclusion criteria were mixed SCLC or neuroendocrine histology. Given the retrospective design of this study, the necessary approvals were secured from the respective Helsinki committees. In Israel, approvals were obtained with IRB code 0023-22-BNZ for the Bnai-Zion Medical Center and approval number 0316 for the Soroka Medical Center. In Greece, permission was granted under reference number 14042/8-6-18. In Poland, the Independent Ethical Committee of the Medical University of Gdansk granted approval with reference number NKEBN/376/2014 for molecular and clinical research involving patients with lung cancer. In Croatia and Slovenia, ethical approval was not required due to the retrospective nature of the study, which did not involve any interventions.

4.3. Statistical Analysis

Descriptive statistics in terms of means, medians, percentages, ranges and interquartile ranges (IQRs) were calculated for all the parameters in the study. The normal distribution of the continuous parameters was determined using the Kolmogorov–Smirnov test. As a result of this test, the Mann–Whitney U test was employed to compare groups. For categorical parameters, the Pearson chi-squared and Fisher exact tests were used. Overall survival and progression-free survival were computed using the log rank test. p < 0.05 was considered significant. SPSS version 28 was used for all statistical analysis. Data were analyzed as frequencies (n) and percentages (%) or medians and ranges. A confidence interval (CI) of 95% and p-values were calculated.

5. Conclusions

Targeted therapy is the best choice for NSCLC patients bearing rare mutations. TT was shown to prolong both the duration of treatment and overall survival.

Author Contributions

Conceptualization, A.A. (Abed Agbarya), U.J., W.S., R.D. and A.P.; methodology, A.A. (Abed Agbarya), W.S. and U.J.; formal analysis A.A. (Abed Agbarya), W.S. and U.J.; investigation, U.J., W.S., K.S., R.D., M.J., G.M., A.P., A.A. (Antonio Araujo), A.C. and A.A. (Abed Agbarya); resources, A.A. (Abed Agbarya) and W.S.; data curation, W.S.; writing—original draft preparation, A.A. (Abed Agbarya); W.S. and U.J.; writing—review and editing, U.J., W.S., K.S., R.D., M.J., G.M., A.P., A.A. (Antonio Araujo), A.C. and A.A. (Abed Agbarya); supervision, A.A. (Abed Agbarya) and W.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by the local Institutional Review Boards. All data gathered were anonymized and cannot be linked back to individual patients. In Israel, approvals were obtained with IRB code 0023-22-BNZ for the Bnai-Zion Medical Center and approval number 0316 for the Soroka Medical Center. In Greece, permission was granted under reference number 14042/8-6-18. In Poland, the Independent Ethical Committee of the Medical University of Gdansk granted approval with reference number NKEBN/376/2014 for molecular and clinical research involving patients with lung cancer. In Croatia and Slovenia, ethical approval was not required due to the retrospective nature of the study, which did not involve any interventions.

Informed Consent Statement

The study was approved by the local IRB with a waiver of signed consent forms due to the retrospective nature.

Data Availability Statement

The data presented in this study are available upon reasonable request from the corresponding author.

Acknowledgments

The authors would like to acknowledge Judith Elbaz and Sara Figueriedo from AstraZeneca for the logistic support provided.

Conflicts of Interest

U.J. reports honoraria for consultancy or lectures from MSD, AstraZeneca, Roche, Takeda, Pfizer, Novartis, BMS, Amgen and Boehringer Ingelheim. W.S. reports honoraria for consultancy or lectures from AstraZeneca, Roche, Novartis, MSD, Pfizer, Merck and Bristol Myers Squibb. K.S. reports honoraria for consultancy or lectures from Roche, AstraZeneca, MSD, Novartis, BMS, Amgen and Vipharm. R.D. reports honoraria for consultancy or lectures from AstraZeneca, Roche, Novartis, MSD, Takeda, Pfizer, Amgen and Bristol Myers Squibb. M.J. reports honoraria for consultancy or lectures from AstraZeneca, Roche, Novartis, MSD, Takeda, Pfizer, Amgen, Boehringer Ingelheim and Bristol Myers Squibb. G.M. reports honoraria for consultancy or lectures from Roche, MSD, BMS, Amgen, AstraZeneca, Sanofi, Novartis, Takeda, Pfizer, Amgen and Janssen. A.P. reports honoraria for consultancy or lectures from BMS, MSD, AstraZeneca and Roche. A.A. (Antonio Araujo) reports honoraria for consultancy or lectures from Merck, IPSEN, Roche, Eli Lily, Pfizer, Janssen and Servier. A.A. (Abed Agbarya) reports honoraria for consultancy or lectures from AstraZeneca, Roche, Novartis, MSD, Takeda, Pfizer, Amgen and Bristol Myers Squibb.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Szero, C.H.; Shalata, W.; Yakobson, A.; Agbarya, A. Neoadjuvant and Adjuvant Immunotherapy in Early-Stage Non-Small-Cell Lung Cancer, Past, Present and Future. J. Clin. Med. 2021, 10, 5614. [Google Scholar] [CrossRef]
  3. Cunha Santos, G.; Shepherd, F.A.; Tsao, M.S. EGFR mutations and lung cancer. Annu. Rev. Pathol. Mech. Dis. 2011, 6, 49–69. [Google Scholar] [CrossRef] [PubMed]
  4. Shalata, W.; Zolnoorian, J.; Migliozzi, G.; Jama, A.A.; Dudnik, Y.; Cohen, A.Y.; Meirovitz, A.; Yakobson, A. Long-Lasting Therapeutic Response following Treatment with Pembrolizumab in Patients with Non-Small Cell Lung Cancer: A Real-World Experience. Int. J. Mol. Sci. 2023, 24, 5938. [Google Scholar] [CrossRef]
  5. D’Arcangelo, M.; D’Incecco, A.; Cappuzzo, F. Rare mutations in non-small-cell lung cancer. Future Oncol. 2013, 9, 699–711. [Google Scholar] [CrossRef]
  6. Shalata, W.; Jacob, B.M.; Agbarya, A. Adjuvant Treatment with Tyrosine Kinase Inhibitors in Epidermal Growth Factor Receptor Mutated Non-Small-Cell Lung Carcinoma Patients, Past, Present and Future. Cancers 2021, 13, 4119. [Google Scholar] [CrossRef]
  7. Ettinger, D.S.; Wood, D.E.; Aggarwal, C.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; et al. NCCN Guidelines Insights: Non-Small Cell Lung Cancer, Version 1.2020. J. Natl. Compr. Cancer Netw. 2019, 17, 1464–1472. [Google Scholar] [CrossRef] [PubMed]
  8. Gkolfinopoulos, S.; Mountzios, G. Beyond EGFR and ALK: Targeting rare mutations in advanced non-small cell lung cancer. Ann. Transl. Med. 2018, 6, 142. [Google Scholar] [CrossRef]
  9. Sholl, L. Molecular diagnostics of lung cancer in the clinic. Transl. Lung Cancer Res. 2017, 6, 560–569. [Google Scholar] [CrossRef]
  10. Taha, T.; Khoury, R.; Brenner, R.; Nasrallah, H.; Shofaniyeh, I.; Yousef, S.; Agbarya, A. Treatment of rare mutations in patients with lung cancer. Biomedicines 2021, 9, 534. [Google Scholar] [CrossRef]
  11. Chen, X.; Xu, B.; Li, Q.; Xu, X.; Li, X.; You, X.; Yu, Z. Genetic profile of non-small cell lung cancer (NSCLC): A hospital-based survey in Jinhua. Mol. Genet. Genom. Med. 2020, 8, e1398. [Google Scholar] [CrossRef] [PubMed]
  12. Chevallier, M.; Borgeaud, M.; Addeo, A.; Friedlaender, A. Oncogenic driver mutations in non-small cell lung cancer: Past, present and future. World J. Clin. Oncol. 2021, 12, 217–237. [Google Scholar] [CrossRef] [PubMed]
  13. Mielgo-Rubio, X.; Azkona Uribelarrea, E.; Quintanta Cortés, L.; Sereno Moyano, M. Immunotherapy in non-small cell lung cancer: Update and new insights. J. Clin. Transl. Res. 2021, 7, 1–21. [Google Scholar]
  14. Memmott, M.R.; Wolfe, A.R.; Carbone, D.P.; Williams, T.M. Predictors of Response, Progression-Free Survival, and Overall Survival in Patients with Lung Cancer Treated with Immune Checkpoint Inhibitors. J. Thorac. Oncol. 2021, 16, 1086–1098. [Google Scholar] [CrossRef] [PubMed]
  15. Tsamis, I.; Gomatou, G.; Chachali, S.P.; Trontzas, I.P.; Patriarcheas, V.; Panagiotou, E.; Kotteas, E. BRAF/MEK inhibition in NSCLC: Mechanisms of resistance and how to overcome it. Clin. Transl. Oncol. 2023, 25, 10–20. [Google Scholar] [CrossRef]
  16. Navab, R.; Liu, J.; Seiden-Long, I.; Shih, W.; Li, M.; Bandarchi, B.; Chen, Y.; Lau, D.; Zu, Y.F.; Cescon, D.; et al. Co-overexpression of Met and hepatocyte growth factor promotes systemic metastasis in NCI-H460 non-small cell lung carcinoma cells. Neoplasia 2009, 11, 1292–1300. [Google Scholar] [CrossRef]
  17. Zhang, L.; Zheng, L.; Yang, Q.; Sun, J. The Evolution of BRAF Activation in Non-Small-Cell Lung Cancer. Front. Oncol. 2022, 12, 882940. [Google Scholar] [CrossRef]
  18. Xu, L.; Wang, F.; Luo, F. MET-Targeted Therapies for the Treatment of Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. Front. Oncol. 2022, 12, 1013299. [Google Scholar] [CrossRef]
  19. Yan, M.; Guo, S.; Zhang, H.; Zhang, Z.; Shen, S.; Li, X. BRAF-Mutated Non-Small Cell Lung Cancer: Current Treatment Status and Future Perspective. Front. Oncol. 2022, 12, 863043. [Google Scholar] [CrossRef]
  20. Santarpia, M.; Massafra, M.; Gebbia, V.; D’Aquino, A.; Garipoli, C.; Altavilla, G.; Rosell, R. A narrative review of MET inhibitors in non-small cell lung cancer with MET exon skipping mutations. Transl. Lung Cancer Res. 2021, 10, 1536–1556. [Google Scholar] [CrossRef]
  21. Wolf, J.; Seto, T.; Han, J.-Y.; Reguart, N.; Garon, E.B.; Groen, H.J.M.; Tan, D.S.W.; Hida, T.; de Jonge, M.; Orlov, S.V.; et al. Capmatinib in MET Exon 14–Mutated or MET -Amplified Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2020, 383, 944–957. [Google Scholar] [CrossRef]
  22. Le, X.; Sakai, H.; Felip, E.; Veillon, R.; Garassino, M.C.; Raskin, J.; Cortot, A.B.; Viteri, S.; Mazieres, J.; Smit, E.F.; et al. Tepotinib Efficacy and Safety in Patients with MET Exon 14 Skipping NSCLC: Outcomes in Patient Subgroups from the VISION Study with Relevance for Clinical Practice. Clin. Cancer Res. 2022, 28, 1117–1126. [Google Scholar] [CrossRef] [PubMed]
  23. Drusbosky, L.M.; Dawar, R.; Rodriguez, E.; Ikpeazu, C.V. Therapeutic strategies in METex14 skipping mutated non-small cell lung cancer. J. Hematol. Oncol. 2021, 14, 129. [Google Scholar] [CrossRef] [PubMed]
  24. Lu, S.; Fang, J.; Li, X.; Cao, L.; Zhou, J.; Guo, Q.; Liang, Z.; Cheng, Y.; Jiang, L.; Yang, N.; et al. Once-daily savolitinib in Chinese patients with pulmonary sarcomatoid carcinomas and other non-small cell lung cancers harbouring MET exon 14 skipping alterations: A multicentre, single-arm, open-label, phase 2 study. Lancet Respir. Med. 2021, 9, 1154–1164. [Google Scholar] [CrossRef] [PubMed]
  25. Markham, A. Savolitinib: First Approval. Drugs 2021, 81, 1665–1670. [Google Scholar] [CrossRef]
  26. Dodson, C.; Richards, T.J.; Smith, D.A.; Ramaiya, N.H. Tyrosine Kinase Inhibitor Therapy for Brain Metastases in Non-Small-Cell Lung Cancer: A Primer for Radiologists. AJNR Am. J. Neuroradiol. 2020, 41, 738–750. [Google Scholar] [CrossRef]
  27. Saxena, P.; Singh, P.K.; Malik, P.S.; Singh, N. Immunotherapy Alone or in Combination with Chemotherapy as First-Line Treatment of Non-Small Cell Lung Cancer. Curr. Treat. Options Oncol. 2020, 21, 69. [Google Scholar] [CrossRef]
  28. Shafique, M.; Tanvetyanon, T. Immunotherapy alone or chemo-immunotherapy as front-line treatment for advanced non-small cell lung cancer. Expert Opin. Biol. Ther. 2019, 19, 225–232. [Google Scholar] [CrossRef]
  29. Proto, C.; Ferrara, R.; Signorelli, D.; Lo Russo, G.; Galli, G.; Imbimbo, M.; Prelaj, A.; Zilembo, N.; Ganzinelli, M.; Pallavicini, L.M.; et al. Choosing wisely first line immunotherapy in non-small cell lung cancer (NSCLC): What to add and what to leave out. Cancer Treat. Rev. 2019, 75, 39–51. [Google Scholar] [CrossRef]
  30. Shields, M.D.; Marin-Acevedo, J.A.; Pellini, B. Immunotherapy for Advanced Non-Small Cell Lung Cancer: A decade of Progress. Am. Soc. Clin. Oncol. Educ. Book 2021, 41, e105–e127. [Google Scholar] [CrossRef]
  31. Fucà, G.; de Braud, F.; Di Nicola, M. Immunotherapy-based combinations: An update. Curr. Opin. Oncol. 2018, 30, 345–351. [Google Scholar] [CrossRef] [PubMed]
  32. Kelly, R.J. Dabrafenib and trametinib for the treatment of non-small cell lung cancer. Expert Rev. Anticancer Ther. 2018, 18, 1063–1068. [Google Scholar] [CrossRef] [PubMed]
  33. Auliac, J.B.; Bayle, S.; Vergnenegre, A.; Le Caer, H.; Falchero, L.; Gervais, R.; Doubre, H.; Vinas, F.; Marin, B.; Chouaid, C. Patients with Non-Small-Cell Lung Cancer Harbouring a Braf Mutation: A Multicentre Study Exploring Clinical Characteristics, Management, and Outcomes in a Real-Life Setting: Explore Gfpc 02-14. Curr. Oncol. 2018, 25, e398–e402. [Google Scholar] [CrossRef]
  34. Ferrucci, P.F.; Lens, M.; Cocorocchio, E. Combined BRAF-Targeted Therapy with Immunotherapy in BRAF-Mutated Advanced Melanoma Patients. Curr. Oncol. Rep. 2021, 23, 138. [Google Scholar] [CrossRef]
  35. Lu, S.; Fang, J.; Wang, Z.; Fan, Y.; Liu, Y.; He, J.; Zhou, J.; Hu, J.; Xia, J.; Liu, W.; et al. Results from Impower132 China cohort: Atezolizumab plus platinum-based chemotherapy in advanced non-small cell lung cancer. Cancer Med. 2023, 12, 2666–2676. [Google Scholar] [CrossRef] [PubMed]
  36. Nishio, M.; Saito, H.; Goto, K.; Watanabe, S.; Sueoka-Aragane, N.; Okuma, Y.; Kasahara, K.; Chikamori, K.; Nakagawa, Y.; Kawakami, T. IMpower132: Atezolizumab plus platinum-based chemotherapy vs chemotherapy for advanced NSCLC in Japanese patients. Cancer Sci. 2021, 112, 1534–1544. [Google Scholar] [CrossRef]
  37. Niu, X.; Sun, Y.; Planchard, D.; Chiu, L.; Bai, J.; Ai, X.; Lu, S. Durable Response to the Combination of Atezolizumab with Platinum-Based Chemotherapy in an Untreated Non-Smoking Lung Adenocarcinoma Patient with BRAF V600E Mutation: A Case Report. Front. Oncol. 2021, 11, 634920. [Google Scholar] [CrossRef]
  38. Liang, H.; Wang, M. MET Oncogene in Non-Small Cell Lung Cancer: Mechanism of MET Dysregulation and Agents Targeting the HGF/c-Met Axis. OncoTargets Ther. 2020, 13, 2491–2510. [Google Scholar] [CrossRef]
  39. Frisone, D.; Friedlaender, A.; Malapelle, U.; Banna, G.; Addeo, A. Critical Reviews in Oncology/Hematology A BRAF New World. Crit. Rev. Oncol./Hematol. 2023, 152, 103008. [Google Scholar] [CrossRef]
  40. Planchard, D.; Besse, B.; Groen, H.J.M.; Hashemi, S.M.S.; Mazieres, J.; Kim, T.M.; Quoix, E.; Souquet, P.J.; Barlesi, F.; Baik, C.; et al. Phase 2 Study of Dabrafenib Plus Trametinib in Patients with BRAF V600E-Mutant Metastatic NSCLC: Updated 5-Year Survival Rates and Genomic Analysis. J. Thorac. Oncol. 2022, 17, 103–115. [Google Scholar] [CrossRef]
  41. Drilon, A.; Clark, J.W.; Weiss, J.; Ou, S.H.I.; Camidge, D.R.; Solomon, B.J.; Otterson, G.A.; Villaruz, L.C.; Riely, G.J.; Heist, R.S.; et al. Antitumor Activity of Crizotinib in Lung Cancers Harboring a MET Exon 14 Alteration. Nat. Med. 2020, 26, 47–51. [Google Scholar] [CrossRef] [PubMed]
  42. Błaszkowska, M.; Specht-Szwoch, Z.; Dziadziuszko, R. Tepotinib for Advanced Non-Small-Cell Lung Cancer with MET Exon 14 Skipping Mutations. ESMO Open 2022, 7, 2021–2022. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, Y.; Liu, T.; Chen, G.; Gong, J.; Bai, Y.; Zhang, T.; Xu, N.; Liu, L.; Xu, J.; He, J.; et al. Phase Ia/Ib Study of the Selective MET Inhibitor, Savolitinib, in Patients with Advanced Solid Tumors: Safety, Efficacy, and Biomarkers. Oncologist 2022, 27, 342–383. [Google Scholar] [CrossRef] [PubMed]
  44. Brazel, D.; Zhang, S.; Nagasaka, M. Spotlight on Tepotinib and Capmatinib for Non-Small Cell Lung Cancer with MET Exon 14 Skipping Mutation. Lung Cancer Targets Ther. 2022, 13, 33–45. [Google Scholar] [CrossRef]
  45. Wiesweg, M.; Preuß, C.; Roeper, J.; Metzenmacher, M.; Eberhardt, W.; Stropiep, U.; Wedeken, K.; Reis, H.; Herold, T.; Darwiche, K.; et al. BRAF Mutations and BRAF Mutation Functional Class Have No Negative Impact on the Clinical Outcome of Advanced NSCLC and Associate with Susceptibility to Immunotherapy. Eur. J. Cancer 2021, 149, 211–221. [Google Scholar] [CrossRef]
  46. Gautschi, O.; Milia, J.; Cabarrou, B.; Bluthgen, M.V.; Besse, B.; Smit, E.F.; Wolf, J.; Peters, S.; Früh, M.; Koeberle, D.; et al. Targeted Therapy for Patients with BRAF-Mutant Lung Cancer Results from the European EURAF Cohort. J. Thorac. Oncol. 2015, 10, 1451–1457. [Google Scholar] [CrossRef]
  47. Shalata, W.; Yakobson, A.; Weissmann, S.; Oscar, E.; Iraqi, M.; Kian, W.; Peled, N.; Agbarya, A. Crizotinib in MET Exon 14-Mutated or MET-Amplified in Advanced Disease Non-Small Cell Lung Cancer: A Retrospective, Single Institution Experience. Oncology 2022, 100, 467–474. [Google Scholar] [CrossRef]
  48. Illini, O.; Fabikan, H.; Swalduz, A.; Vikström, A.; Krenbek, D.; Schumacher, M.; Dudnik, E.; Studnicka, M.; Öhman, R.; Wurm, R.; et al. Real-World Experience with Capmatinib in MET Exon 14-Mutated Non-Small Cell Lung Cancer (RECAP): A Retrospective Analysis from an Early Access Program. Ther. Adv. Med. Oncol. 2022, 14, 259–261. [Google Scholar] [CrossRef]
  49. Paik, P.K.; Goyal, R.K.; Cai, B.; Price, M.A.; Davis, K.L.; Ansquer, V.D.; Caro, N.; Saliba, T.R. Real-World Outcomes in Non-Small-Cell Lung Cancer Patients with MET Exon 14 Skipping Mutation and Brain Metastases Treated with Capmatinib. Future Oncol. 2023, 19, 217–228. [Google Scholar] [CrossRef]
  50. Sforza, V.; Palumbo, G.; Cascetta, P.; Carillio, G.; Manzo, A.; Montanino, A.; Sandomenico, C.; Costanzo, R.; Esposito, G.; Laudato, F.; et al. BRAF Inhibitors in Non-Small Cell Lung Cancer. Cancers 2022, 14, 4863. [Google Scholar] [CrossRef]
  51. Hellmann, M.D.; Kim, T.W.; Lee, C.B.; Goh, B.C.; Miller, W.H.; Oh, D.Y.; Jamal, R.; Chee, C.E.; Chow, L.Q.M.; Gainor, J.F.; et al. Phase Ib Study of Atezolizumab Combined with Cobimetinib in Patients with Solid Tumors. Ann. Oncol. 2019, 30, 1134–1142. [Google Scholar] [CrossRef] [PubMed]
  52. Hong, L.; Zhang, J.; Heymach, J.V.; Le, X. Current and Future Treatment Options for MET Exon 14 Skipping Alterations in Non-Small Cell Lung Cancer. Ther. Adv. Med. Oncol. 2021, 13, 1758835921992976. [Google Scholar] [CrossRef] [PubMed]
  53. Desai, A.; Cuellar, S. The Current Landscape for MET ex14 Skipping Mutations in Non–Small Cell Lung Cancer. J. Adv. Pract. Oncol. 2022, 13, 539–544. [Google Scholar] [CrossRef] [PubMed]
  54. Guaitoli, G.; Tiseo, M.; Di Maio, M.; Friboulet, L.; Facchinetti, F. Immune Checkpoint Inhibitors in Oncogene-Addicted Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Transl. Lung Cancer Res. 2021, 10, 2890–2916. [Google Scholar] [CrossRef]
  55. Cardarella, S.; Ogino, A.; Nishino, M.; Butaney, M.; Shen, J.; Lydon, C.; Yeap, B.Y.; Sholl, L.M.; Johnson, B.E.; Jänne, P.A. Clinical, Pathological and Biological Features Associated with BRAF Mutations in Non-Small Cell Lung Cancer Stephanie. Clin. Cancer Res. 2013, 19, 4532–4540. [Google Scholar] [CrossRef]
  56. Murciano-Goroff, Y.R.; Pak, T.; Mondaca, S.; Flynn, J.R.; Montecalvo, J.; Rekhtman, N.; Halpenny, D.; Plodkowski, A.J.; Wu, S.L.; Kris, M.G.; et al. Immune Biomarkers and Response to Checkpoint Inhibition of BRAF V600 and BRAF Non-V600 Altered Lung Cancers. Br. J. Cancer 2022, 126, 889–898. [Google Scholar] [CrossRef]
  57. Mazieres, J.; Drilon, A.; Lusque, A.; Mhanna, L.; Cortot, A.B.; Mezquita, L.; Thai, A.A.; Mascaux, C.; Couraud, S.; Veillon, R.; 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. [Google Scholar] [CrossRef]
  58. Dudnik, E.; Peled, N.; Nechushtan, H.; Wollner, M.; Onn, A.; Agbarya, A.; Moskovitz, M.; Keren, S.; Popovits-Hadari, N.; Urban, D.; et al. BRAF Mutant Lung Cancer: Programmed Death Ligand 1 Expression, Tumor Mutational Burden, Microsatellite Instability Status, and Response to Immune Check-Point Inhibitors. J. Thorac. Oncol. 2018, 13, 1128–1137. [Google Scholar] [CrossRef]
  59. Guisier, F.; Dubos-Arvis, C.; Viñas, F.; Doubre, H.; Ricordel, C.; Ropert, S.; Janicot, H.; Bernardi, M.; Fournel, P.; Lamy, R.; 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. [Google Scholar] [CrossRef]
  60. Rihawi, K.; Giannarelli, D.; Galetta, D.; Delmonte, A.; Giavarra, M.; Turci, D.; Garassino, M.; Tiseo, M.; Barbieri, F.; Panni, S.; et al. BRAF Mutant NSCLC and Immune Checkpoint Inhibitors: Results from a Real-World Experience. J. Thorac. Oncol. 2019, 14, e57–e59. [Google Scholar] [CrossRef]
  61. Mayenga, M.; Assié, J.B.; Monnet, I.; Massiani, M.A.; Tabeze, L.; Friard, S.; Fraboulet, S.; Métivier, A.C.; Chouaïd, C.; Zemoura, L.; et al. Durable Responses to Immunotherapy of Non-Small Cell Lung Cancers Harboring MET Exon-14–Skipping Mutation: A Series of 6 Cases. Lung Cancer 2020, 150, 21–25. [Google Scholar] [CrossRef] [PubMed]
  62. Ettinger, D.S.; Wood, D.E.; Aisner, D.L.; Akerley, W.; Bauman, J.R.; Bharat, A.; Bruno, D.S.; Chang, J.Y.; Chirieac, L.R.; D’Amico, T.A.; et al. Non–Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2022, 20, 497–530. [Google Scholar] [CrossRef] [PubMed]
  63. Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T.; et al. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021, 6, 201. [Google Scholar] [CrossRef] [PubMed]
  64. Remon, J.; Hendricks, L.E.L.; Mountzios, G.; García-Campelo, R.; Saw, S.P.L.; Uprety, D.; Recondo, G.; Villacampa, G.; Reck, M. MET alterations in NSCLC—Current Perspectives and Future Challenges. J. Thorac. Oncol. 2023, 18, 419–435. [Google Scholar] [CrossRef] [PubMed]
Table 1. Patients’ demographics and clinical characteristics (n = 58).
Table 1. Patients’ demographics and clinical characteristics (n = 58).
CharacteristicFrequency (%)
Gender 
  Female28 (48)
  Male30 (52)
Age at diagnosis (years [range])66.7 [36–91]
Family history of cancer (n = 54)12 (22)
Smoking habit 
  Current17 (29)
  Former21 (36)
  Never20 (35)
Background diseases38 (65)
  Hypertension 26 (45)
  Diabetes mellitus7 (12)
  Peripheral vascular disease3 (5)
  Other chronic conditions17 (29)
  COPD 15 (9)
  Hepatic disease1 (2)
Stage at initial diagnosis 
  IB1 (2)
  IIA2 (3)
  IIB3 (5)
  IIIA1 (2)
  IIIB3 (5)
  IVA38 (66)
  IVB10 (17)
Histological diagnosis: adenocarcinoma50 (86)
Mutation type  
  BRAF V600E point mutation17 (29)
  BRAF translocation1 (2)
  MET exon 14 skipping mutation 40 (69)
Genetic testing method, NGS 258 (100)
PD-L1 score (n = 44) 
  <1%8 (18)
  1–49%15 (34)
  ≥50%21 (48)
ECOG performance status (n = 57) 
  05 (9)
  143 (75)
  27 (12)
  32 (4)
Number of metastatic sites at time of advanced disease (n = 57) 
  ≤553 (93)
  >54 (7)
  Brain12 (21)
  Contralateral lung29 (50)
  Lymph nodes, extra-thoracic13 (22)
  Pleural17 (29)
  Pericardial2 (4)
  Bone26 (45)
  Adrenal14 (24)
  Liver12 (21)
  Spleen1 (1)
  Peritoneal1 (1)
Abbreviation: 1 COPD, chronic obstructive pulmonary disease; 2 NGS, next-generation sequencing.
Table 2. Patients’ treatment and follow-up (n = 58).
Table 2. Patients’ treatment and follow-up (n = 58).
TreatmentFrequency (%)
Surgery 5 (8.6)
SRBT 16 (10.3)
CRT 2 (radical treatment)4 (6.9)
Consolidation with immunotherapy1 (1.7)
First-line treatment (n = 53) 
  Chemotherapy16 (30)
  Chemo-IO 31 (2)
  IO monotherapy7 (13)
  Targeted therapy (TT)29 (55)
First-line treatment BRAF mutation (n = 17) 
  Targeted therapy (TT)14 (82)
  Chemotherapy doublet2 (12)
  IO monotherapy1 (6)
Best response to first-line TT BRAF mutation (n = 14) 
  Complete response1 (7)
  Partial response10 (71)
  Stable disease1 (7)
  Progressive disease2 (15)
First-line treatment cMET exon 14 skipping mutation (n = 36) 
  Targeted therapy15 (42)
  Chemotherapy14 (39)
  Chemo-IO1 (3)
  IO monotherapy6 (16)
Best response to first-line TT cMET exon 14 skipping mutation (n = 15) 
  Partial response8 (53)
  Stable disease2 (13)
  Progressive disease4 (27)
  NA1 (7)
Best response to second-line TT cMET exon 14 skipping mutation (n = 12) 
  Partial response7 (58)
  Stable disease1 (9)
  Progressive disease4 (33)
No. of cycles (n [range])6 [2.5–12]
Reason for discontinuation (n = 42) 
  Death7 (17)
  End of treatment4 (9)
  Progression31 (74)
  Toxicity0
Type of progression (n = 36) 
CNS disease recurrence1 (3)
Distant only1 (3)
Local/regional only17 (47)
Local/regional and distant17 (47)
Local radical treatment (n = 45)12 (27)
Second-line treatment28 (48)
Abbreviation: 1 SRBT, stereotactic body radiation therapy; 2 CRT, chemoradiotherapy; 3 Chemo-IO, combined chemotherapy and immunotherapy.
Table 3. Patients’ therapy at first and second line.
Table 3. Patients’ therapy at first and second line.
 Targeted TherapyOther Therapiesp
Median no. of cycles [range]9.5 [4.25–14.0]5 [2–6.5]p = 0.01
First-line median duration of treatment (days) [range]326 [122–428]133.5 [62.3–178.5]p = 0.003
Second-line median duration of treatment [range] (days)194 [106–338]159 [99.5–285.5]p = 0.51
Drug access type at first line, n (%)n = 28n = 25p < 0.001
Extended access program2 (7)0 
Individual reimbursement9 (32)0 
National reimbursement12 (43)25 (100) 
Other5 (18)0 
Drug access type at second linen = 15n = 13NS 1
Extended access program4 (26.7)0 
National reimbursement9 (60)12 (92) 
Other2 (13)0 
Private insurance01 (8) 
Abbreviation: 1 NS, not significant; n, number.
Table 4. Patients’ treatment regimens.
Table 4. Patients’ treatment regimens.
 Frequency (n)Death Events (n)Censored (n)Percent (%)
Targeted therapy2816 (57)1243
Other treatment2515 (60)1040
Overall53312242
Table 5. Patients’ survival for the treatment regimens (n = 53) (months).
Table 5. Patients’ survival for the treatment regimens (n = 53) (months).
TreatmentMedian ± SD 195% Confidence Interval
Survival  
  Biological drugs19.2 ± 8.42–35.7
  All other13.6 ± 2.58.6–18.5
  Overall15.5 ± 3.58.6–22.3
Progression-free survival  
  Biological drugs11.6 ± 3.25.4–17.8
  All other4.6 ± 1.31.9–7.2
  Overall7.9 ± 1.25.6–10.2
Abbreviation: 1 SD, standard deviation.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Janzic, U.; Shalata, W.; Szymczak, K.; Dziadziuszko, R.; Jakopovic, M.; Mountzios, G.; Płużański, A.; Araujo, A.; Charpidou, A.; Agbarya, A. Real-World Experience in Treatment of Patients with Non-Small-Cell Lung Cancer with BRAF or cMET Exon 14 Skipping Mutations. Int. J. Mol. Sci. 2023, 24, 12840. https://doi.org/10.3390/ijms241612840

AMA Style

Janzic U, Shalata W, Szymczak K, Dziadziuszko R, Jakopovic M, Mountzios G, Płużański A, Araujo A, Charpidou A, Agbarya A. Real-World Experience in Treatment of Patients with Non-Small-Cell Lung Cancer with BRAF or cMET Exon 14 Skipping Mutations. International Journal of Molecular Sciences. 2023; 24(16):12840. https://doi.org/10.3390/ijms241612840

Chicago/Turabian Style

Janzic, Urska, Walid Shalata, Katarzyna Szymczak, Rafał Dziadziuszko, Marko Jakopovic, Giannis Mountzios, Adam Płużański, Antonio Araujo, Andriani Charpidou, and Abed Agbarya. 2023. "Real-World Experience in Treatment of Patients with Non-Small-Cell Lung Cancer with BRAF or cMET Exon 14 Skipping Mutations" International Journal of Molecular Sciences 24, no. 16: 12840. https://doi.org/10.3390/ijms241612840

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

Article Metrics

Back to TopTop