Next Article in Journal
Recent Advances in Treatment Options for Childhood Acute Lymphoblastic Leukemia
Previous Article in Journal
Feasibility of Tumor Treating Fields with Pemetrexed and Platinum-Based Chemotherapy for Unresectable Malignant Pleural Mesothelioma: Single-Center, Real-World Data
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Multicenter Observational Study on Metastatic Non-Small Cell Lung Cancer Harboring BRAF Mutations: Focus on Clinical Characteristics and Treatment Outcome of V600E and Non-V600E Subgroups

by
Fabiana Perrone
1,†,
Giulia Mazzaschi
1,2,†,
Roberta Minari
1,*,
Michela Verzè
1,
Cinzia Azzoni
3,
Lorena Bottarelli
3,
Rita Nizzoli
1,
Monica Pluchino
1,
Annalisa Altimari
4,
Elisa Gruppioni
4,
Francesca Sperandi
5,
Elisa Andrini
6,
Giorgia Guaitoli
7,8,
Federica Bertolini
7,
Fausto Barbieri
7,
Stefania Bettelli
9,
Lucia Longo
10,
Maria Pagano
11,
Candida Bonelli
11,
Elena Tagliavini
12,
Davide Nicoli
13,
Alessandro Ubiali
14,
Adriano Zangrandi
14,
Serena Trubini
14,
Manuela Proietto
15,
Letizia Gnetti
3 and
Marcello Tiseo
1,2
add Show full author list remove Hide full author list
1
Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
2
Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
3
Unit of Pathological Anatomy, University Hospital of Parma, 43126 Parma, Italy
4
Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
5
Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
6
Department of Experimental Diagnostic and Specialized Medicine (DIMES), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
7
Division of Medical Oncology, University Hospital of Modena, 41125 Modena, Italy
8
Ph.D. Program Clinical and Experimental Medicine (CEM), Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
9
Pathology Unit, University Hospital of Modena, 41125 Modena, Italy
10
Medical Oncology Unit, Sassuolo Hospital, AUSL Modena, 41121 Modena, Italy
11
Medical Oncology Unit, Clinical Cancer Centre, Azienda USL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy
12
Pathology Unit, Clinical Cancer Centre, Azienda USL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy
13
Molecular Biology, Oncology and Advanced Technology Unit, Azienda USL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy
14
Pathology Unit, AUSL Piacenza, 29121 Piacenza, Italy
15
Medical Oncology Unit, AUSL Piacenza, 29121 Piacenza, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to the work.
Submission received: 31 March 2022 / Revised: 13 April 2022 / Accepted: 14 April 2022 / Published: 16 April 2022

Abstract

:

Simple Summary

Around 2–4% of lung adenocarcinoma harbors BRAF mutations. Dabrafenib and Trametinib represent the first treatment-choice for BRAF V600Emut NSCLC, regardless of the line of therapy, while non-V600Emut receive standard immunotherapy or chemo-immunotherapy. Our real-life multicenter study on 44 BRAF mutant NSCLC responds to the urgent need to characterize this subset of patients in-depth, potentially offering new valuable biological and clinical insights. We specifically focused on similarities/discrepancies between V600E and non-V600E populations, providing consistent data about clinicopathologic characteristics, treatment response, and survival outcome.

Abstract

Introduction: BRAF mutation involved 2–4% of lung adenocarcinoma. Differences in clinicopathologic features and patient outcome exist between V600E and non-V600E BRAF mutated NSCLC. Thus, we sought to assess the frequency and clinical relevance of BRAF mutations in a real-life population of advanced-NSCLC, investigating the potential prognostic significance of distinct genetic alterations. Materials and Methods: The present multicenter Italian retrospective study involved advanced BRAF mutant NSCLC. Complete clinicopathologic data were evaluated for BRAF V600E and non-V600E patients. Results: A total of 44 BRAFmut NSCLC patients were included (V600E, n = 23; non-V600E, n = 21). No significant differences in survival outcome and treatment response were documented, according to V600E vs. non-V600E mutations, although a trend towards prolonged PFS was observed in the V600E subgroup (median PFS = 11.3 vs. 6.0 months in non-V600E). In the overall population, ECOG PS and age significantly impacted on OS, while bone lesions were associated with shorter PFS. Compared to immunotherapy, first-line chemotherapy was associated with longer OS in the overall population, and especially in the BRAF V600E subtype. Conclusions: Here, we report on real-life data from a retrospective cohort of advanced-NSCLC harboring BRAF alterations. Our study offers relevant clues on survival outcome, therapeutic response, and clinicopathologic correlations of BRAF-mutant NSCLC.

1. Introduction

V-Raf murine sarcoma viral oncogene homolog (BRAF) mutations have been described in 8% of cancer, with higher incidence reported in melanoma (40–60%), papillary thyroid carcinomas (30–70%), and colorectal cancers (5–20%) [1,2]. The BRAF gene was localized on chromosome 7 and encodes for the BRAF protein, a RAF kinase which plays a key role in the MAPK cascade, a signaling pathway involved in cell growth, differentiation, proliferation, senescence, and apoptosis [3,4,5,6]. The most common activating BRAF mutation found in cancer is a point mutation in exon 15 (c.1799T > A) that corresponds to a valine to glutamate substitution at codon 600 (V600E) [7]. The mutation leads to a 500-fold increase in the kinase activity of BRAF and allows BRAF to be active, regardless of RAS-mediated activation [8,9]. In lung cancer, BRAF is mutated in approximately 2–4% of cases [10]. Different from melanoma, in which the vast majority of BRAF mutations occur at the V600 site, only approximately 50% of BRAF mutant lung adenocarcinomas harbor V600 mutations, with the rest of the cases carrying non-V600 mutations in exons 11 and 15 [1,10]. Compared to other oncogenic drivers, BRAF mutations are more frequent in current/former smokers, less frequently mutually exclusive, and associated with higher PD-L1 expression and better response to immunotherapy [11,12,13,14]. The prognostic role of BRAF V600E mutation in lung cancer is still unclear. However, multiple evidence reported poorer outcomes and lower response rate to platinum-based chemotherapy in BRAF-mutated NSCLC patients compared to BRAF-wild type [15]. The therapeutic landscape of BRAF V600 mutant NSCLC has been dramatically revolutionized in the last few years. Indeed, based on the encouraging results achieved in melanoma, several studies have investigated the association between BRAF and MEK inhibitors in BRAF mutant NSCLC. Additionally, as a consequence of the favorable results of a phase II, single-arm study in both pre-treated and treatment-naïve NSCLC patients harboring BRAF V600E mutations, the combination of dabrafenib plus trametinib, has become the standard of care for these patients [16,17,18]. On the other hand, chemotherapy and immunotherapy represent the treatment of choice in non-V600E mutant NSCLC, given the undefined efficacy of BRAF and MEK inhibitor against this molecular alteration [19]. However, in light of the association of some non-V600E mutations, such as G469A and L597R, with high BRAF kinase activity, dual BRAF ± MEK blockage could be conceivably employed, also in this setting [10,20]. Several previous studies have investigated the incidence, distribution, and prognosis of BRAF mutant lung adenocarcinoma, but, partly due to relatively small sample size and the retrospective nature, the results have been demonstrated as inconclusive and/or contradictory [11,12,21,22].
Here, we present real-world data on a large cohort of advanced NSCLC patients harboring BRAF mutations from five Italian Institutions in the attempt to describe clinical features and survival outcomes, with particular attention given to potential differences between BRAF V600E mutated and non-V600E mutated subgroups.

2. Materials and Methods

2.1. Patients and Study Objectives

The present multicenter retrospective observational study (DETECTION_KB study) was carried out in five Italian Centers (Piacenza, Parma, Reggio Emilia, Modena, and Bologna), involving Oncology and Pathology Units, and included consecutive patients with advanced NSCLC with a BRAF mutation diagnosed between 1 January 2018 and 28 February 2020. Patients had to be >18 years old and have data available for clinical follow-up.
The primary aim of the study was to assess the incidence of BRAF mutation in a real-life cohort of advanced-NSCLC patients and to illustrate the different type of detected mutations. As exploratory aims, we explored the impact of distinct BRAF mutations (V600E vs. non-V600E) on survival outcome and treatment response and the potential association with specific clinicopathological characteristics.

2.2. Study Design and Data Collection

Participating centers entered data into a database and completed a survey on the molecular procedures for the diagnostic approach in testing BRAF mutation (see Appendix A). The BRAF molecular testing used to analyze the tissue biopsy or cytological specimens had to have similar sensitivity in all participating centers. The following data were extracted from medical records: patient characteristics (date of diagnosis, age at diagnosis, sex, Eastern Cooperative Oncology Group performance status, smoking status); disease history (disease stage at diagnosis, tumor histology, presence of BRAF mutations, site of diagnostic biopsy). Outcome data for analysis were also collected from medical records, including the start and end dates of systemic therapy, number of lines of treatment, type of response (complete response, partial response, stable disease, and progression of disease (CR, PR, SD and PD)), date of radiologic disease progression (according to the RECIST criteria version 1.1), date of death, and survival outcomes (PFS, progression-free survival and OS, overall survival). The study was conducted in accordance with the current revisions of the Declaration of Helsinki (Fortaleza, Brazil, 2013) and Italian laws on observational studies; ethical approval was obtained for all five Centers. Written informed consent for trial participation was obtained from all patients (expect for deceased patients).

2.3. Statistical Analysis

Anonymous data were collected by each participating Center and treated according to Italian national laws and General Data Protection Regulations. Data were subsequently analyzed by the Medical Oncology Unit of Parma University Hospital.
Comparative analyses were performed for patients with BRAF V600E versus non-V600E mutations, and to assess differences between groups, Fisher’s exact test and the Mann-Whitney test were employed for categorical variables and continuous variables, respectively.
OS and PFS were estimated according to the Kaplan Meier method. OS was defined as the interval from the date of diagnosis of metastatic disease to the date of death or last follow-up for alive patients. PFS was defined as the interval from the date of diagnosis of metastatic disease to the date of radiological/clinical progression or death due to any cause, whichever occurred first, or last follow-up visit for patients alive without disease progression. Cut-off for survival analysis was set at 30 March 2021. Median follow-up was calculated according to the so-termed ‘reverse Kaplan Meier’ (Kaplan Meier estimate of potential follow-up) technique [23].
Log-rank test (Mantel Cox) was applied to evaluate statistical differences in PFS and OS between subgroups. Univariate and multivariate Cox proportional hazards regression models were used to test potential prognostic factors both in terms of PFS and OS, and results were presented with hazard ratios (HRs), 95% CI, and p values.
Complete and partial responses (CR and PR, respectively) were gathered, with stable diseases (SD) lasting at least 6 months in the disease control group (DC = CR + PR + SD ≥ 6 months), set against the progressive disease (PD) group, which comprises PD and SD lasting less than 6 months.
For all the analyses described above, a p value of 0.05 was set as a threshold of statistical significance. To minimize the risk of multiplicity, Bonferroni correction test was applied to all our multiple comparisons. All statistical analyses were carried out using the Statistical Package for the Social Sciences program (SPSS), version 25.0, (IBM, Armonk, NY, USA).

3. Results

3.1. Baseline Patient Characteristics

From 1 January 2016 to 28 February 2020, an overall number of 2342 patients with advanced NSCLC were tested for BRAF mutation, as per clinical practice, by the five Pathology Units involved in the DETECTION_KB study. Of all patients, 127 (5.4%) were BRAF mutated: 68 (2.9%) carried a V600E mutation and 59 (2.5%) another (non-V600E) mutation in the BRAF gene (Consort Diagram—Figure 1).
At data cut-off, 44 NSCLC cases with available clinical data and adequate follow-up were included, of which 23 presented BRAF V600E mutation (V600E group, 52.2%) and 21 (47.8%) displayed BRAF mutations other than V600E (non-V600E). Out of 21 non-V600E NSCLC, a considerable proportion (38.1%, n = 8) harbored G469A/V/S mutations and an equal fraction carried G466A (n = 5, 23.8%) and D594G/N (n = 5, 23.8%) alterations. Two cases (9.5%) displayed BRAF G464G mutation, while only one had Q257K mutation (Consort Diagram—Figure 1).
Baseline clinicopathologic characteristics of our patient population are reported in Table 1. Median age was 67 years, ranging from 47 to 84, and male patients accounted for 64% of the overall population. The vast majority of patients had 0 or 1 performance status (89%), and 27 (61%) cases presented with more than three metastatic sites at the time of diagnosis. Concerning the histological subtypes, 36 patients had adenocarcinoma (ADC) histology (82%), while 16% and 2% presented not otherwise specified (NOS) NSCLC and squamous cell carcinoma (SCC), respectively. Whitin tested cases (71%), PD-L1 Tumor Proportional Score (TPS) resulted in ≥50% in 16 patients (37%), while the proportion of PD-L1 negative (TPS < 1%) and intermediate (TPS 1–49%) samples was essentially comparable.
Chemotherapy-based regimens represented the most employed first-line treatment (68%), while 21% and 11% patients were administered with single-agent immunotherapy and BRAF/MEK inhibitors, respectively (four cases underwent Dabrafenib plus Trametinib and one patient Encorafenib plus Binimetinib).
Concerning the therapeutic course, among 22 patients who started a second-line therapy, 9 (41%) received single-agent immunotherapy, 6 (27.3%) chemotherapy, and only 1 (4.5%) chemo-immunotherapy combination. Six BRAF V600E mutant NSCLCs were treated with anti-BRAF/MEK combination of Dabrafenib and Trametinib. The proportion of cases undergoing third- and fourth-line was 18.2% (n = 8) and 2.3% (n = 1), with an equal distribution among chemotherapy- and immunotherapy-treated groups.
With a median follow-up of 36.1 months (95% CI 17.9–54.3), median OS and PFS were 13.9 (95% CI 8.4–19.6) and 6.2 (95% CI 5.6–6.9), respectively (Figure 2Ai,Aii). In terms of response to first-line therapy, 24 patients (64.9% of evaluable patients) experienced DC, while 13 (35.1%) experienced PD; 7 were not evaluable.

3.2. BRAF Mutations and Baseline Clinicopathologic Characteristics

We subsequently evaluated whether the distribution of clinicopathologic features differed according to specific BRAF mutations (V600E vs. non-V600E). As reported in Table 1, no significant variations in terms of number or location of metastatic sites, gender, performance status, age, and first-line treatment were detected between V600E and non-V600E subgroups. Intriguingly, a strong predominance of non-smokers was present in V600E subset (27% vs. 0% in non-V600E, p = 0.032), and NSCLC NOS appeared to be less represented in the non-V600E subgroup (22% vs. 9%, p = 0.024). Moreover, when evaluating PD-L1 status, BRAF V600E samples displayed a trend towards lower levels of PD-L1 expression, with a relatively higher proportion of TPS < 1% (39% vs. 29% in non-V600E, not significant (NS)). Nonetheless, considering the relatively limited sample size and the high number of comparisons, when the obtained statistically significant p values were adjusted following Bonferroni’s correction test, no meaningful differences were observed (Supplementary Table S1).

3.3. BRAF Mutational Status and Impact on Clinical Outcome

Partly as a consequence of the relatively limited sample size, BRAF mutational status did not display a striking impact on OS, with a median OS of 13.3 months (95% CI 10.3–16.3) for BRAF V600E and 15.1 months (95% CI 4.4–29.7) for BRAF non-V600E (Figure 2Bi). Nevertheless, a tendency towards prolonged PFS was apparent in patients carrying BRAF V600E mutation, with a median PFS of 11.3 months (95% CI 3.3–19.2) compared to 6.0 months (95% CI 5.4–6.6) in BRAF non-V600E (Figure 2Bii). When focusing on disease response to first-line treatment, the proportion of patients experiencing DC and PD was largely comparable between the two BRAF subgroups (Figure 2C).

3.4. Impact of Clinicopathologic Characteristics on Clinical Outcome

In order to provide useful information about the prognostic and predictive value of widely recognized clinicopathologic characteristics, we assessed the impact of baseline clinical and histopathologic features on patient outcome in the overall population and in BRAF V600E vs. non-V600E subgroups. In terms of OS, only ECOG PS and age appeared to significantly condition patient outcome (Table 2). Specifically, patients with ECOG 0–1 and age below the median (<67 years) displayed prolonged OS compared to their counterpart (Cox regression univariate analysis, p < 0.05). Although without reaching statistical significance, a trend towards favorable prognosis was documented in female patients (p = 0.110, HR = 0.51) and without liver metastatic involvement (p = 0.095, HR = 2.26).
With regards to PFS, while most of clinicopathologic parameters did not significantly affect the outcome, the evidence of bone lesions was meaningfully associated with a shorter PFS (p = 0.020) (Table 2).
When challenged on multivariate analysis, only ECOG PS maintained their statistically significant impact on OS (data not shown).
We next explored the prognostic value of the abovementioned clinicopathologic characteristics within the two BRAF mutational subgroups, thus revealing slight differences. In terms of OS, ECOG PS and age confirmed their prognostic value only in the BRAF non-V600E subgroup, while histotype (NSCLC NOS vs. ADC/SCC, p = 0.04) and presence of brain metastases (p = 0.032) negatively conditioned OS of BRAF V600E cases. When evaluating PFS, male sex and lymph nodal involvement resulted significant only in BRAF non-V600E, while in BRAF V600E subgroup, we did not identify any prognostically relevant factor (Table 2).
Even following Bonferroni’s correction test, the statistical significance of our results was maintained (Supplementary Table S2).

3.5. Impact of First-Line Therapeutic Strategy on Clinical Outcome

In the perspective of patient management optimization, we finally sought to investigate the putative prognostic role of chemotherapy- vs. immunotherapy-based regimens in our cohort of BRAF mutant NSCLC, excluding those patients treated with first-line targeted agents. As shown in Figure 3Ai, in the overall population, the positive impact of CT-based treatment was remarkable in terms of OS (Median OS: 17.6 vs. 7.1 mos. in IT-treated patients, p = 0.074), although not translated in an analogous PFS advantage (Figure 3Aii). Conversely, the survival outcome of IT-treated patients was disappointing, registering median OS and PFS of 7.1 and 6.2 months, respectively.
When stratifying patients in BRAF V600E and non-V600E subgroups, we demonstrated that BRAF V600E mutant NSCLC treated with first-line chemotherapy displayed a significantly prolonged OS compared to those receiving single-agent immunotherapy (Median OS = 20.4 vs. 7.0 mos. in IT-treated patients, p = 0.019; Figure 3Bi), while any difference was observed in BRAF non-V600E subset (Figure 3Ci). In both BRAF V600E and non-V600E patients, PFS was not meaningfully affected (Figure 3Bii,Cii).
Finally, we focused on BRAF V600E mutant NSCLC patients receiving first-line target therapies (TT). Nevertheless, mainly due to the limited sample size and the unbalanced distribution of first-line treatments (CT-treated group, n = 14; IT-treated group, n = 4; TT-treated group, n = 5), we did not document any statistically significant difference.

4. Discussion

The present retrospective multicenter Italian study reports clinical outcomes of metastatic NSCLC patients harboring BRAF mutations. To date, despite available evidence in this setting of patients, data demonstrated is largely inconclusive, likely due to the relatively limited sample size and the retrospective nature of the studies. Thus, we sought to delve into this subset of lung cancer patients in an attempt to provide a real-life portrayal of the BRAF mutant population, particularly highlighting similarities and discrepancies between V600E and non-V600E groups.
The overall frequency of BRAF mutations was 5.4%, slightly higher than expected [10], but in line with a recent Italian survey [24]. At data cut-off, 44 consecutive BRAF mutant advanced NSCLC patients were included and the proportion of cases harboring BRAF V600E alteration resulted in 52.2%, consistent with other reported observations [10,11,12,21].
In addition to V600E, we explored the frequencies of rarer BRAF mutations, such as G469A/V/S, G466A, D594G/N, G464G, and Q257K.
When we analyzed the distribution of clinicopathologic features according to BRAF mutational subtypes (V600E vs. non-V600E), we observed that V600E mutations preferentially occurred in never-/light-smokers. This finding is in line with a previous meta-analysis, including 10 studies, in which no significant differences in BRAF mutation frequency were found in former/current smokers versus never smokers (OR = 0.95, 95% CI: 0.45–2.02), whereas the incidence of BRAF V600E significantly differed in former or current smokers versus never smokers (OR = 0.14, 95% CI: 0.05–042) [25]. No other relevant clinical profiles emerged to be associated with BRAF mutation subtype in our study. Specifically, we did not document any associations between gender, performance status, age, number, and location of metastatic sites, or BRAF mutational status. This evidence are similar to previously published works, confirming the smoking status as unique distinctive clinical characteristics of V600E mutant NSCLC [11,12,21].
The clinical outcome of patients with BRAF V600E mutation compared to those carrying non-V600E alterations was essentially comparable, with no significant differences in terms of survival and response to therapy. A favorable trend in PFS was observed in the V600E population, although without reaching statistical significance. This finding is partly in contrast with previous reports, demonstrating shorter PFS in V600E mutant patients, likely attributable to a more aggressive histotype, characterized by micropapillary features, typical of the V600E subgroup [13,21,26]. Considering the lack of such detailed histological information in our study, a conclusive judgment is not conceivable and future research involving a larger cohort of cases are warranted.
When focusing on the clinical outcome according to type of BRAF mutation and type of therapy, we found that median OS in the overall population treated with first-line platinum-based chemotherapy was 17.6 months, in line with that reported by previous works [21,27].
A worse performance of first-line immunotherapy versus chemotherapy was observed in terms of OS, either in the overall population or in the BRAF V600E subset. This latter finding partly mimics the preliminary results of Tan et al., although acknowledging their limited sample size [27]. Conversely, it has been demonstrated that, compared to other NSCLC harboring oncogene driver alterations, BRAF mutant patients could benefit from immunotherapy. Dudnik et al. conducted a retrospective study on a cohort of 39 patients positive for BRAF alterations and treated with ICIs. The study revealed a benefit from immunotherapy administered in first or subsequent lines, with an objective response rate (ORR) of 25–30% and a median PFS of 3.7–4.1, which are comparable to the results observed in the second-line setting in the unselected population of NSCLC [14]. No difference was observed in ORR or PFS according to the type of BRAF mutation. Similarly, a better outcome compared to other oncogene alteration was reported in terms of RR and PFS in the Immunotarget trial [13]. Finally, Rihawi et al. carried out a retrospective study on an Italian expanded access program comprising patients with advanced non-squamous NSCLC treated with second-line nivolumab [28]. The median OS were 10.3 and 11.2 months in BRAF mutated and wild type patients, respectively. The above-mentioned data demonstrates that immunotherapy could represent a reasonable treatment option after target therapy and chemotherapy in BRAF mutated NSCLC. At present, there are no data from randomized controlled trials comparing different first-line therapeutic strategies in BRAF mutated NSCLC and updated ESMO guidelines, although, when designating dabrafenib plus trametinib as the preferred regimen in a BRAF setting, it did not mention chemo-immunotherapy combinations, thus leaving this critical issue partly unanswered [29]. A recent retrospective study presented at ESMO 2021 Annual Congress using a propensity score weighting method showed that the first-line target therapy with dabrafenib plus trametinib reported a numerically longer OS and PFS over single-agent immunotherapy and chemo-immunotherapy [30]. In this regard, we did not detect any significant difference according to first-line regimens, likely depending on the relatively small sample size and the unbalanced distribution of first-line treatments among NSCLC patients.
Finally, we explored the association between BRAF variants and expression of PD-L1. A trend towards lower levels of PD-L1 was observed in the V600E population compared to the non-V600E subgroup. This result is inconsistent with previous evidence reporting high level of PD-L1 expression (TPS ≥ 50%) in the BRAF mutant population, with a similar proportion between V600E and the non-V600E subgroup and could partly explain the poor outcome obtained with immunotherapy [14].
Although the present multicenter Italian study addressed the clinically relevant goal to provide a real-life portrait of a still partially explored subset of advanced NSCLC carrying BRAF mutation, some limitations have to be acknowledged. First, the retrospective and non-interventional nature of our research, as well as the relatively limited sample size, prevents any conclusive judgement on different BRAF-driven subgroups. Second, the absence of a control group includes BRAF wild-type patients. Nonetheless, consistent with the literature, these fundamental unsolved issues require large prospective studies, involving BRAF mutant and wild-type cases, to be fully addressed.

5. Conclusions

This is one of the largest Italian series of advanced NSCLC patients with BRAF mutation, highlighting distinctive features of BRAF V600E and non-V600E mutant NSCLC in terms of survival outcome, therapeutic response, and clinicopathologic correlations. In particular, our study suggests that immunotherapy could not be the best option in BRAF mutant NSCLC patients, especially in V600E subtype, underlining the opportunity to consider a targeted therapy approach. To completely dissect the impact of target therapies, first-line chemotherapy- or immunotherapy-based treatment strategies, larger prospective studies are warranted.

Supplementary Materials

The following supporting information can be downloaded at: https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/cancers14082019/s1, Supplementary Table S1: Patient Population Characteristics of the overall population and according to BRAF mutational status, Table S2: Explanatory prognostic factors in Cox proportional hazard models: uni- and multivariate analysis illustrating the impact on OS and PFS of clinico-pathological characteristics.

Author Contributions

Conceptualization, R.M. and M.T.; methodology, F.P., G.M., R.M. and M.T.; formal analysis, F.P. and G.M.; data curation, M.V., M.P. (Monica Pluchino), F.S., E.A., G.G., F.B. (Federica Bertolini), F.B. (Fausto Barbieri), L.L., M.P. (Maria Pagano), C.B., M.P. (Manuela Proietto), and L.G.; resources, C.A., L.B., R.N., A.A., E.G., S.B., E.T., D.N., A.U., A.Z. and S.T.; writing—original draft preparation, F.P. and G.M.; writing—review and editing, F.P., G.M., R.M. and M.T.; supervision, R.M. and M.T. 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 according to the guidelines of the Declaration of Helsinki and approved by the Ethics Committee of the University Hospital of Parma (protocol code: 17028, and date of approval: 19 May 2020).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available in order to protect patients privacy.

Conflicts of Interest

Marcello Tiseo: Advisory boards and/or speakers’ fee for Astra-Zeneca, Pfizer, Eli-Lilly, BMS, Novartis, Roche, MSD, Boehringer Ingelheim, Otsuka, Takeda, Pierre Fabre, Amgen, Merck, Sanofi. Research Grants from Astra-Zeneca and Boehringer Ingelheim. For the other authors, no potential conflicts of interest were disclosed.

Appendix A

BRAF testing was performed during the molecular characterization of advanced NSCLC as per clinical practice with the diagnostic test currently in use at each Pathological Unit of participating centers.
The methods used, divided by technology, were:
Next Generation Sequencing: Oncomine Dx Target Test (cod. A32452, Thermo Fisher Scientific, Waltham, MA, USA); Myriapod NGS Cancer Panel DNA (cod. NG033, Diatech Pharmacogenetics, Jesi, Italy) and Myriapod® NGS 56G onco panel (cod. NG052, Diatech Pharmacogenetics); real-time PCR: EasyPGX ready BRAF (cod. RT022, Diatech Pharmacogenetics); Sequenom: Myriapod Lung status (cod. SQ011, Diatech Pharmacogenetics) and Myriapod Colon status (cod. SQ010, Diatech Pharmacogenetics); Pyrosequencing: Anti-EGFR MoAb response (BRAF status) (cod. UP033, Diatech Pharmacogenetics) and therascreen BRAF Pyro kit (cod. 971470, Qiagen, Germantown, MD, USA).

References

  1. Davies, H.; Bignell, G.R.; Cox, C.; Stephens, P.; Edkins, S.; Clegg, S.; Teague, J.; Woffendin, H.; Garnett, M.J.; Bottomley, W.; et al. Mutations of the BRAF gene in human cancer. Nature 2002, 417, 949–954. [Google Scholar] [CrossRef] [PubMed]
  2. Dankner, M.; Rose, A.A.N.; Rajkumar, S.; Siegel, P.M.; Watson, I.R. Classifying BRAF alterations in cancer: New rational therapeutic strategies for actionable mutations. Oncogene 2018, 37, 3183–3199. [Google Scholar] [CrossRef] [PubMed]
  3. Peyssonnaux, C.; Eychène, A. The Raf/MEK/ERK pathway: New concepts of activation. Biol. Cell 2001, 93, 53–62. [Google Scholar] [CrossRef]
  4. Wasylyk, C.; Wasylyk, B.; Heidecker, G.; Huleihel, M.; Rapp, U.R. Expression of raf oncogenes activates the PEA1 transcription factor motif. Mol. Cell. Biol. 1989, 9, 2247. [Google Scholar] [PubMed] [Green Version]
  5. Jamal, S.; Ziff, E. Transactivation of c-fos and β-actin genes by raf as a step in early response to transmembrane signals. Nature 1990, 344, 463–466. [Google Scholar] [CrossRef] [PubMed]
  6. Kolch, W.; Heidecker, G.; Lloyd, P.; Rapp, U.R. Raf-1 protein kinase is required for growth of induced NIH/3T3 cells. Nature 1991, 349, 426–428. [Google Scholar] [CrossRef]
  7. Tan, Y.H.; Liu, Y.; Eu, K.W.; Ang, P.W.; Li, W.Q.; Salto-Tellez, M.; Iacopetta, B.; Soong, R. Detection of BRAF V600E mutation by pyrosequencing. Pathology 2008, 40, 295–298. [Google Scholar] [CrossRef]
  8. Wan, P.T.C.; Garnett, M.J.; Roe, S.M.; Lee, S.; Niculescu-Duvaz, D.; Good, V.M.; Project, C.G.; Jones, C.M.; Marshall, C.J.; Springer, C.J.; et al. Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF. Cell 2004, 116, 855–867. [Google Scholar] [CrossRef] [Green Version]
  9. Yao, Z.; Torres, N.M.; Tao, A.; Gao, Y.; Luo, L.; Li, Q.; de Stanchina, E.; Abdel-Wahab, O.; Solit, D.B.; Poulikakos, P.I.; et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer Cell 2015, 28, 370–383. [Google Scholar] [CrossRef] [Green Version]
  10. Russo, A.; Lopes, A.R.; McCusker, M.G.; Garrigues, S.G.; Ricciardi, G.R.; Arensmeyer, K.E.; Scilla, K.A.; Mehra, R.; Rolfo, C. New Targets in Lung Cancer (Excluding EGFR, ALK, ROS1). Curr. Oncol. Rep. 2020, 22, 48. [Google Scholar] [CrossRef]
  11. Litvak, A.M.; Paik, P.K.; Woo, K.M.; Sima, C.S.; Hellmann, M.D.; Arcila, M.E.; Ladanyi, M.; Rudin, C.M.; Kris, M.G.; Riely, G.J. Clinical characteristics and course of 63 patients with BRAF mutant lung cancers. J. Thorac. Oncol. 2014, 9, 1669–1674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Marchetti, A.; Felicioni, L.; Malatesta, S.; Sciarrotta, M.G.; Guetti, L.; Chella, A.; Viola, P.; Pullara, C.; Mucilli, F.; Buttitta, F. Clinical features and outcome of patients with non-small-cell lung cancer harboring BRAF mutations. J. Clin. Oncol. 2011, 29, 3574–3579. [Google Scholar] [CrossRef] [PubMed]
  13. 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] [PubMed]
  14. 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] [Green Version]
  15. Ferrara, M.G.; Di Noia, V.; D’argento, E.; Vita, E.; Damiano, P.; Cannella, A.; Ribelli, M.; Pilotto, S.; Milella, M.; Tortora, G.; et al. Oncogene-Addicted Non-Small-Cell Lung Cancer: Treatment Opportunities and Future Perspectives. Cancers 2020, 12, 1196. [Google Scholar] [CrossRef]
  16. Planchard, D.; Besse, B.; Groen, H.J.M.; Souquet, P.J.; Quoix, E.; Baik, C.S.; Barlesi, F.; Kim, T.M.; Mazieres, J.; Novello, S.; et al. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: An open-label, multicentre phase 2 trial. Lancet Oncol. 2016, 17, 984–993. [Google Scholar] [CrossRef] [Green Version]
  17. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus trametinib in patients with previously untreated BRAF V600E-mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef]
  18. 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]
  19. Leonetti, A.; Facchinetti, F.; Rossi, G.; Minari, R.; Conti, A.; Friboulet, L.; Tiseo, M.; Planchard, D. BRAF in non-small cell lung cancer (NSCLC): Pickaxing another brick in the wall. Cancer Treat. Rev. 2018, 66, 82–94. [Google Scholar] [CrossRef]
  20. Tabbò, F.; Pisano, C.; Mazieres, J.; Mezquita, L.; Nadal, E.; Planchard, D.; Pradines, A.; Santamaria, D.; Swalduz, A.; Ambrogio, C.; et al. How far we have come targeting BRAF-mutant non-small cell lung cancer (NSCLC). Cancer Treat. Rev. 2022, 103, 102335. [Google Scholar] [CrossRef]
  21. 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, pathologic, and biologic features associated with BRAF mutations in non-small cell lung cancer. Clin. Cancer Res. 2013, 19, 4532–4540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Paik, P.K.; Arcila, M.E.; Fara, M.; Sima, C.S.; Miller, V.A.; Kris, M.G.; Ladanyi, M.; Riely, G.J. Clinical characteristics of patients with lung adenocarcinomas harboring BRAF mutations. J. Clin. Oncol. 2011, 29, 2046–2051. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Schemper, M.; Smith, T.L. A note on quantifying follow-up in studies of failure time. Control. Clin. Trials 1996, 17, 343–346. [Google Scholar] [CrossRef]
  24. Malapelle, U.; Rossi, G.; Pisapia, P.; Barberis, M.; Buttitta, F.; Castiglione, F.; Cecere, F.L.; Grimaldi, A.M.; Iaccarino, A.; Marchetti, A.; et al. BRAF as a positive predictive biomarker: Focus on lung cancer and melanoma patients. Crit. Rev. Oncol. Hematol. 2020, 156, 103118. [Google Scholar] [CrossRef]
  25. Chen, D.; Zhang, L.Q.; Huang, J.F.; Liu, K.; Chuai, Z.R.; Yang, Z.; Wang, Y.X.; Shi, D.C.; Liu, Q.; Huang, Q.; et al. BRAF mutations in patients with non-small cell lung cancer: A systematic review and meta-analysis. PLoS ONE 2014, 9, e101354. [Google Scholar] [CrossRef] [Green Version]
  26. Ding, X.; Zhang, Z.; Jiang, T.; Li, X.; Zhao, C.; Su, B.; Zhou, C. Clinicopathologic characteristics and outcomes of Chinese patients with non-small-cell lung cancer and BRAF mutation. Cancer Med. 2017, 6, 555–562. [Google Scholar] [CrossRef]
  27. Tan, I.; Stinchcombe, T.E.; Ready, N.E.; Crawford, J.; Datto, M.B.; Nagy, R.J.; Lanman, R.B.; Gu, L.; Clarke, J.M. Therapeutic outcomes in non-small cell lung cancer with BRAF mutations: A single institution, retrospective cohort study. Transl. Lung Cancer Res. 2019, 8, 258–267. [Google Scholar] [CrossRef]
  28. 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] [Green Version]
  29. Planchard, D.; Popat, S.; Kerr, K.; Novello, S.; Smit, E.F.; Faivre-Finn, C.; Mok, T.S.; Reck, M.; Van Schil, P.E.; Hellmann, M.D. Updated version published 15 September 2020 by the ESMO Guidelines Committee, Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29, 192–237. [Google Scholar] [CrossRef]
  30. Melosky, B.; Knoll, S.; Souef, I.S.; Wu, W.H.; Rhodes, W.; Martinalbo, J.; Ye, F. Clinical outcomes of patients with BRAFv600-mutated metastatic NSCLC (mNSCLC) receiving first-line (1L) dabrafenib-trametinib vs other standard of care in real-world practice. Ann. Oncol. 2021, 32, S949–S1039. [Google Scholar] [CrossRef]
Figure 1. Consort diagram of DETECTION_KB study population: Advanced NSCLC patients included in this analysis, from 1 January 2016 to 28 February 2020.
Figure 1. Consort diagram of DETECTION_KB study population: Advanced NSCLC patients included in this analysis, from 1 January 2016 to 28 February 2020.
Cancers 14 02019 g001
Figure 2. (Ai,Aii) Median overall survival (OS) and progression-free survival (PFS) in the overall population. (Bi,Bii) Kaplan Meier survival curves illustrating the survival outcome according to BRAF mutational status (V600E vs. non-V600E). (C) Stacked charts showing the treatment response (DC vs. PD) in BRAF V600E vs. non-V600E subsets. DC = CR + PR + SD ≥ 6 months; PD = PD + SD < 6 months.
Figure 2. (Ai,Aii) Median overall survival (OS) and progression-free survival (PFS) in the overall population. (Bi,Bii) Kaplan Meier survival curves illustrating the survival outcome according to BRAF mutational status (V600E vs. non-V600E). (C) Stacked charts showing the treatment response (DC vs. PD) in BRAF V600E vs. non-V600E subsets. DC = CR + PR + SD ≥ 6 months; PD = PD + SD < 6 months.
Cancers 14 02019 g002
Figure 3. (Ai,Aii): Kaplan Meier survival curves reporting the impact of first-line treatment (CT vs. IT) on OS and PFS in the overall population. (Bi,Bii): Kaplan Meier survival curves reporting the impact of first-line treatment (CT vs. IT) on OS and PFS in the BRAF V600E group. (Ci,Cii): Kaplan Meier survival curves reporting the impact of first-line treatment (CT vs. IT) on OS and PFS in BRAF non-V600E group. Abbreviations: CT, chemotherapy; IT, immunotherapy.
Figure 3. (Ai,Aii): Kaplan Meier survival curves reporting the impact of first-line treatment (CT vs. IT) on OS and PFS in the overall population. (Bi,Bii): Kaplan Meier survival curves reporting the impact of first-line treatment (CT vs. IT) on OS and PFS in the BRAF V600E group. (Ci,Cii): Kaplan Meier survival curves reporting the impact of first-line treatment (CT vs. IT) on OS and PFS in BRAF non-V600E group. Abbreviations: CT, chemotherapy; IT, immunotherapy.
Cancers 14 02019 g003
Table 1. Patient Population Characteristics. SCC: Squamous Cell Carcinoma; ADC: Adenocarcinoma; NSCLC: Non-Small Cell Lung Cancer; NOS: Not Otherwise Specified, ECOG: Eastern Cooperative Group; TPS: Tumor Proportional Score.
Table 1. Patient Population Characteristics. SCC: Squamous Cell Carcinoma; ADC: Adenocarcinoma; NSCLC: Non-Small Cell Lung Cancer; NOS: Not Otherwise Specified, ECOG: Eastern Cooperative Group; TPS: Tumor Proportional Score.
Patient CharacteristicsOverall
(n = 44)
BRAF V600E
(n = 23)
BRAF Non-V600E
(n = 21)
p-Value
Age, years (Median, range) 67 (47–84)79 (47–81)65 (49–84)0.459
n (%)
HistotypeSCC1 (2)1 (4)0 (0)0.024
ADC36 (82)17 (74)19 (91)
NSCLC NOS7 (16)5 (22)2 (9)
SexMale28 (64)16 (70)12 (57)0.533
Female16 (36)7 (30)9 (43)
Smoking status *Smokers9 (24)3 (14)6 (37)0.032
Ex-Smokers23 (60)13 (59)10 (63)
Non Smokers6 (16)6 (27)0 (0)
ECOG PS0–139 (89)21 (91)18 (86)0.658
25 (11)2 (9)3 (14)
Stage of the diseaseIIIB4 (9)2 (9)2 (9)0.946
IV40 (91)21 (91)19 (91)
Number of metastatic sites<317 (39)10 (43)7 (33)0.548
≥327 (61)13 (57)14 (67)
Metastatic InvolvementBone14 (32)7 (30)7 (33)1.000
Brain8 (18)3 (13)5 (24)0.448
Liver7 (16)5 (22)2 (9)0.416
Lung20 (45)11 (48)9 (43)0.771
Lymph nodes32 (73)15 (65)17 (81)0.318
Adrenal gland5 (11)4 (17)1 (5)0.348
Pleura14 (32)9 (39)5 (24)0.342
First-line TreatmentChemotherapy30 (68)14 (61)16 (76)0.081
Immunotherapy9 (21)4 (17)5 (24)
Targeted therapy §5 (11)5 (22)0 (0)
PD-L1 statusTPS ≥ 50%16 (37)30 (23)47 (28)0.513
1% ≤ TPS < 50%7 (16)42 (32)59 (35)
TPS < 1%8 (18)51 (39)48 (29)
Unknown13 (29)7 (6)13 (8)
* In six patients, the data was not available. § Four patients were treated with Dabrafenib + Trametinib, one patient with Encorafenib + Binimetinib. p values are referred to the comparison between BRAF V600E and non-V600E groups (Fisher’s exact test for categorical variables; Mann Whitney test for continuous variables). Significant p values were reported in bold.
Table 2. Explanatory prognostic factors in Cox proportional hazard models: uni- and multivariate analysis illustrating the impact on OS and PFS of clinico-pathological characteristics.
Table 2. Explanatory prognostic factors in Cox proportional hazard models: uni- and multivariate analysis illustrating the impact on OS and PFS of clinico-pathological characteristics.
OS,
Univariate Analysis a
OverallBRAF V600EBRAF Non-V600E
HRCI (95%)χ2p ValueHRCI (95%)χ2p ValueHRCI (95%)χ2p Value
Age1.051.01–1.105.730.0231.020.96–1.080.540.4771.071.01–1.154.750.030
Histotype1.180.78–1.790.580.4202.161.04–4.493.970.0400.880.45–1.750.140.725
Sex0.510.22–1.212.400.1100.481.14–1.701.490.2550.611.18–2.010.690.414
Smoking status1.060.53–2.100.030.8691.320.54–3.230.380.5391.240.29–5.290.080.772
ECOG PS4.171.38–12.634.770.0111.390.18–11.040.090.75418.63.0–75.688.890.002
N of metastatic
sites
0.980.46–2.080.030.9561.180.43–3.300.741.1850.880.27–2.940.040.841
Bone metastasis1.480.64–3.390.820.3561.910.55–6.630.970.3071.180.37–3.780.080.775
Brain metastasis0.940.35–2.490.020.9014.671.14–19.073.710.0320.520.11–2.430.790.406
Liver metastasis 2.260.87–5.912.450.0952.180.62–7.601.370.2222.680.54–13.11.210.229
Lymphnodes
metastasis
1.190.53–2.670.180.6682.090.70–6.251.880.1840.581.15–2.140.620.410
Adrenal
metastasis
1.480.43–5.080.360.5312.890.71–11.791.910.1390.040.03–9.111.640.537
I line treatment1.130.82–1.550.490.4581.140.80–1.610.480.4701.380.37–1.120.220.631
PD-L1 status, TPS1.270.72–2.230.710.4081.160.48–2.790.110.7421.290.57–2.960.380.541
PFS,
Univariate Analysis a
OverallBRAF V600EBRAF Non-V600E
HRCI (95%)χ2p ValueHRCI (95%)χ2p ValueHRCI (95%)χ2p Value
Age0.980.94–1.021.250.2570.980.93–1.030.610.4300.980.91–1.060.180.673
Histotype0.830.52–1.340.650.4481.110.52–2.380.080.7800.760.38–1.510.750.440
Sex0.910.42–1.970.060.8101.470.48–4.540.430.5040.190.05–0.795.970.022
Smoking status1.440.77–2.681.320.2540.890.37–2.170.060.8051.610.54–4.820.710.393
ECOG PS3.560.42–29.841.020.2428.640.78–15.82.210.0790.040.01–7.890.350.776
N of metastatic
sites
1.390.62–3.110.660.4221.530.49–4.720.570.4550.870.27–2.810.050.813
Bone metastasis2.601.16–5.815.090.0202.220.64–7.691.450.2092.600.89–7.562.990.079
Brain metastasis2.310.84–6.372.280.1032.280.47–11.10.890.3063.630.79–16.52.480.096
Liver metastasis 1.190.47–2.990.130.7141.750.51–6.030.730.3780.990.21–4.540.010.986
Lymphnodes
metastasis
0.960.43–2.150.010.9250.990.33–2.990.010.9932.170.04–2.784.350.023
Adrenal metastasis1.600.54–4.750.650.3950.480.61–9.011.430.2041.370.17–11.090.080.766
I line treatment0.790.51–1.211.480.2780.870.56–1.330.480.5120.630.14–2.820.410.545
PD-L1 status, TPS0.720.40–1.281.210.2670.690.26–1.830.530.4550.960.44–2.090.010.910
PFS: progression free survival; OS: overall survival; Age (continue variable), Histotype (SCC = 0, ADC = 1, NOS = 3), Smoking status (negative smoking history = 0, positive smoking history = 1), Sex (Male = 0, Female = 1), Eastern Cooperative Oncology Group performance status (ECOG PS, 0–1 vs. 2), number of metastatic sites (<3 = 0, ≥3 = 1), bone/brain/liver/lymphnode/adrenal gland metastasis (absence = 0, presence = 1), first-line treatment (chemotherapy = 0, immunotherapy = 1, chemo-immunotherapy = 2), PD-L1 status (TPS < 1% = 0, 1–49% = 1, ≥50% = 2); Statistical results with p < 0.05 are bolded. a Univariate analysis is carried out without any adjustment.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Perrone, F.; Mazzaschi, G.; Minari, R.; Verzè, M.; Azzoni, C.; Bottarelli, L.; Nizzoli, R.; Pluchino, M.; Altimari, A.; Gruppioni, E.; et al. Multicenter Observational Study on Metastatic Non-Small Cell Lung Cancer Harboring BRAF Mutations: Focus on Clinical Characteristics and Treatment Outcome of V600E and Non-V600E Subgroups. Cancers 2022, 14, 2019. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14082019

AMA Style

Perrone F, Mazzaschi G, Minari R, Verzè M, Azzoni C, Bottarelli L, Nizzoli R, Pluchino M, Altimari A, Gruppioni E, et al. Multicenter Observational Study on Metastatic Non-Small Cell Lung Cancer Harboring BRAF Mutations: Focus on Clinical Characteristics and Treatment Outcome of V600E and Non-V600E Subgroups. Cancers. 2022; 14(8):2019. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14082019

Chicago/Turabian Style

Perrone, Fabiana, Giulia Mazzaschi, Roberta Minari, Michela Verzè, Cinzia Azzoni, Lorena Bottarelli, Rita Nizzoli, Monica Pluchino, Annalisa Altimari, Elisa Gruppioni, and et al. 2022. "Multicenter Observational Study on Metastatic Non-Small Cell Lung Cancer Harboring BRAF Mutations: Focus on Clinical Characteristics and Treatment Outcome of V600E and Non-V600E Subgroups" Cancers 14, no. 8: 2019. https://0-doi-org.brum.beds.ac.uk/10.3390/cancers14082019

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