Skip to main content
The Oncologist logoLink to The Oncologist
. 2019 May 31;24(12):e1331–e1340. doi: 10.1634/theoncologist.2018-0914

Characteristics of BRAFV600E Mutant, Deficient Mismatch Repair/Proficient Mismatch Repair, Metastatic Colorectal Cancer: A Multicenter Series of 287 Patients

Christelle de la Fouchardière a,*, Romain Cohen b, David Malka c, Rosine Guimbaud d, Héloïse Bourien e, Astrid Lièvre f, Wulfran Cacheux g, Pascal Artru h, Eric François i, Marine Gilabert j, Emmanuelle Samalin‐Scalzi k, Aziz Zaanan l, Vincent Hautefeuille m, Benoit Rousseau n, Hélène Senellart o, Romain Coriat p, Ronan Flippot c, Françoise Desseigne a, Audrey Lardy‐Cleaud q, David Tougeron r
PMCID: PMC6975964  PMID: 31152084

This article describes the real‐life management of BRAFV600E‐mutant metastatic colorectal cancer in a large cohort of patients treated in 16 French centers from 2006 to 2017. Potential prognostic factors to steer treatment decisions are identified.

Keywords: Colorectal cancer, BRAF, Mismatch repair testing, Prognostic, Decision making

Abstract

Background.

BRAFV600E mutations occurring in about 10% of metastatic colorectal cancers (mCRCs) are usually associated with a poor outcome. However, their prognostic factors are unknown.

Materials and Methods.

We built a multicenter clinico‐biological database gathering data from patients with BRAFV600E‐mutant mCRC treated in one of the 16 French centers from 2006 to 2017. The primary endpoint was to identify prognostic factors using a Cox model.

Results.

We included 287 patients (median age, 67 years [28–95]; female, 57%). Their median overall survival was 20.8 months (95% confidence interval [CI], 17.97–27.04), and median progression‐free survival in the first‐line setting was 4.34 months (95% CI, 3.81–5.03). Chemotherapy regimen and biological agents (antiangiogenic or anti‐epidermal growth factor receptor) were not associated with overall and progression‐free survival. Stage IV disease (synchronous metastases) and absence of curative‐intent surgery were statistically associated with poor overall survival. Among the 194 patients with mismatch repair (MMR) status available, overall survival was significantly longer in patients with deficient MMR tumors compared with those with proficient MMR tumors (adjusted hazard ratio = 0.56; p = .009).

Conclusion.

Despite that BRAFV600E‐mutant mCRCs are associated with poor overall and progression‐free‐survival, patients with deficient MMR tumors and/or resectable disease experienced a longer survival. These results highlight the importance of MMR testing and resectability discussion in patients with BRAFV600E mCRC in day‐to‐day practice.

Implications for Practice.

Mismatch repair (MMR) testing and resectability discussion in patients with BRAFV600E metastatic colorectal cancer (mCRC) should be performed in day‐to‐day practice to steer treatment decision making in patients with BRAFV600E‐mutant mCRC.

Introduction

The BRAF protein is a member of the serine/threonine kinase family that transduces signals downstream of RAS via a cascade of phosphorylation from MAPK/ERK kinase 1/2 (MEK1/2) to extracellular signal‐regulated kinase 1/2 (ERK1/2). Incidence of BRAF mutations, identified in human cancers in 2002, varies according to tumor types. The highest BRAF mutation rates are observed in melanoma and papillary thyroid cancers and are generally involved the V600 codon [1]. BRAF mutations are currently classified into three categories, according to their kinase activity, RAS dependency, and dimerization status underlying the differential tumor response to epidermal growth factor receptor (EGFR), BRAF, and MEK inhibitors [2], [3]. Indeed, BRAF inhibitor monotherapies are not effective in BRAFV600E‐mutant metastatic colorectal cancer (mCRC) [4], [5], [6].

Colorectal cancer (CRC) is the third most common cancer type with more than 1,800,000 new cases and about 883,000 deaths worldwide in 2018 according to GLOBOCAN estimates [7]. RAS mutations are identified in about half of mCRC. They are used as predictive biomarker for resistance to anti‐EGFR therapies [8]. In addition, BRAFV600E mutations observed in about 10% of mCRC [9] are often associated with MLH1 promoter hypermethylation and CpG island methylator phenotype inducing a microsatellite instability (MSI) phenotype [10], [11]. A rate of mismatch repair (MMR) deficiency (dMMR) from 20% to 40% was reported in BRAFV600E‐mutant mCRC [12]. In metastatic setting, BRAFV600E mutations have a significant negative prognostic impact leading to a reduced median overall survival (OS) of about 12 months [13], [14], [15]. On the other hand, its predictive role in anti‐EGFR resistance remains controversial [16], [17], [18]. Indeed, the management of patients with BRAFV600E‐mutant mCRC is heterogeneous, based on the administration of tri‐ or bi‐chemotherapy regimen combined or not with bevacizumab, according to patient's age and performance status [19], [20]. Despite that recent studies demonstrated the benefit of immune checkpoint inhibitors in patients with dMMR tumors, no other biomarker allows to guide therapeutic choices [21], [22].

Our study describes the real‐life management of patients with BRAFV600E‐mutant mCRC in a large retrospective multicenter French cohort. We aimed to identify potential prognostic factors to steer treatment decision making in patients with BRAFV600E‐mutant mCRC.

Materials and Methods

Study Design

We retrospectively enrolled data from consecutive patients with BRAFV600E‐mutant mCRC identified from January 2006 to September 2017 in 16 centers. Eligible patients were older than 18 years with BRAFV600E‐mutation identified by polymerase chain reaction (PCR) or next‐generation sequencing analysis on tumor sample. Patients with mCRC who had experienced a curative resection of their metastases were also included. Noninclusion criteria were patients without metastases or for whom treatment and follow‐up information were not available.

Patient and Tumor Characteristics

Initial CRC stage was categorized according to the 7th Tumor Node Metastasis (TNM) classification system [23]. Somatic mutations in RAS genes and MMR status were collected, when available. MMR status was determined by MSI testing (pentaplex PCR) and/or analysis of immunohistochemistry [24]. dMMR status was defined as the presence of an instability for more than 20% of the microsatellites or a loss of MLH1, MSH2, PMS2, and/or MSH6 expression. Routine follow‐up consisted of physical examination, biological tests, and computed tomography scan every 2–3 months to evaluate treatment response and toxicity.

Statistical Analysis

Descriptive statistics were used to summarize patients’ characteristics. Median OS was defined as the time between the date of metastase(s) diagnosis and date of death (from any cause) or censored at the date of last follow‐up (September 1, 2017). Progression‐free survival (PFS) of the first‐line treatment (PFS1) was defined as the time between the initiation date of first‐line treatment and date of first disease progression, or the initiation date of second‐line treatment, or death, or censored at the date of last follow‐up. Patients with curative surgery for metastase(s) and primary tumor were also censored at the surgery date. Patients with curative surgery first and then “adjuvant” chemotherapy were excluded for PFS1 analysis. PFS of chemotherapy lines 2, 3, and 4 were calculated with the same definition. Survival curves for OS and PFS with associated log‐rank tests were generated using the Kaplan‐Meier method. Median follow‐up was calculated using reverse Kaplan‐Meier estimation.

A Cox proportional hazards model was used to investigate prespecified factors for OS and PFS. The variables considered were age at metastases diagnosis, gender, stage IV disease (synchronous or metachronous metastase(s)), primary tumor site, metastatic site (liver, lung, bone, and brain), number of metastatic sites, surgery of primary tumor and/or of metastase(s), and type of palliative treatment. For OS and PFS1, the sufficiently informed variables (less than 10% of missing data) and significant at a 0.20 level were included in a backward selection procedure to keep factors significant at 5% level in the final multivariate Cox model. SAS version 9.4 was used for all statistical analyses (SAS Institute Inc., Cary, NC).

Results

Clinical Characteristics

A total of 287 patients from 16 French centers (8 comprehensive cancer care centers, 7 university hospitals, and 1 private hospital) were included (Table 1). The median age was 67 years, and 57.1% of patients were women. Two (0.69%) patients had a known germinal mutation in one MMR gene. Nearly two thirds of patients (65.9 %) had synchronous metastases, and the primary tumor localization was mainly ascendant colon (65.4%). The most frequent metastatic sites were liver (51.9%), followed by peritoneum (37.3%), lymph nodes (31.0%), and lung (25.8%). More than half of patients (55.4%) had one metastatic site, including 82 patients (28.6%) with liver‐only metastases.

Table 1. Demographic and clinical characteristics.

image

Global population of BRAFV600E‐mutant patients. Data are median (minimum–maximum) or n (%).

a

According to International Union Against Cancer.

Abbreviations: dMMR, deficient mismatch repair; M, metastasis; MMR, mismatch repair; N, node; pMMR, proficient mismatch repair; T, tumor.

Pathological and Molecular Characteristics

Most patients had advanced T and N stage at diagnosis (T3–4: 74.6%; N1–2: 57.9%). A total of 98 (34.1%) patients had a specific pathological component, including 79 (27.5%) with mucinous component in >50% of the tumor. Of note, other rare histological CRC subtypes such as signet ring cells carcinoma (n = 7) or sarcomatoid carcinoma (n = 2) were identified. The MMR status was determined for 194 patients and identified 85 (43.8%) patients with dMMR and 109 patients with proficient MMR (pMMR), among whom were 42 with MLH1 promoter methylation. Among the patients with KRAS status determined (n = 279), only two tumors harbored a RAS mutation (n = 2).

Treatments

Most patients (75.6%) had experienced a resection of primary tumor. Among the 96 patients with a stage II–III tumor at diagnosis, 57.3% received adjuvant chemotherapy (CT), mainly oxaliplatin‐based regimen (65.5%). Forty‐four patients (15.3%) had a curative metastasectomy, including liver metastases (n = 41) and complete cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (n = 5). Of the 44 patients, 19 experienced upfront metastasectomy without neoadjuvant or perioperative chemotherapy. Most patients (n = 256, 89.2%) had a first‐line CT (supplemental online Table 1). The 31 patients (10.8%) who did not begin CT were either too old (80.5 [49–95]) or had poor performance status. The median duration of the first‐line CT was 3.2 months (0.1–38.9). Only 25 patients (9.7%) received tri‐CT (FOLFOXIRI or FOLFIRINOX), in combination with either antiangiogenic or anti‐EGFR therapy for 16 of them. Doublet with oxaliplatin‐based CT was the most used protocol (49.2%), followed by irinotecan‐based CT (28.9%). Cytotoxic drugs were combined with bevacizumab in 42.6% or anti‐EGFR therapies in 17.6% of the patients. The main reasons for first‐line CT discontinuation were disease progression (51.5%), treatment holiday (24.3%), and toxicity (11.1%).

Of the 256 patients who received a first‐line CT, 158 (61.7%) initiated a second‐line CT for a median duration of 2.3 (0.03–47.0) months. In the second‐line setting, oxaliplatin‐based bi‐CT and irinotecan‐based CT were used in 18.6% and 44.2% of the patients, respectively. Of note, in the 109 patients treated with an antiangiogenic drug in the first‐line setting, 29 (26.6%) continued the antiangiogenic drug in the second‐line setting while switching chemotherapy regimen. Tumor progression was the main reason for second‐line CT discontinuation (70.8%). A total of 81 (28.2%) patients subsequently initiated a third line and 34 (11.8%) a fourth line of chemotherapy. Moreover, 7 patients received regorafenib, 8 patients received an anti‐BRAF, and 10 patients have been treated with an immune checkpoint inhibitors in clinical trials.

OS and PFS

With a median follow‐up of 47.5 months (3.3–210.7), the median OS was 20.8 months (95% CI, 18.0–27.0; Fig. 1). Overall, 68.9% of the patients were alive at 12 months (95% CI, 63.1–74) and 47.7% at 24 months (95% CI, 41.5–53.6). In the multivariate analysis, metachronous metastases (hazard ratio [HR] = 0.697; 95% CI, 0.499–0.973; p = .0341), metastasectomy (HR = 0.469; 95% CI, 0.288–0.765; p = .0024), and primary tumor surgery (HR = 0.306; 95% CI, 0.214–0.438; p < .0001) were statistically associated with better OS (Table 2). Regimen of first‐line treatment had no impact on OS (oxaliplatin‐ vs. +irinotecan‐based CT, antiangiogenic vs. anti‐EGFR therapies). The median OS for BRAF‐mutant patients who had a surgery for their metastases was 47.4 months (28.5–nonevaluable) versus 19.5 months (15.9–22.0) for those who had no metastasectomy (p < .001; Fig. 2). The median OS for patients with no surgery (neither primary tumor nor metastases) and at least one first‐line chemotherapy was 9.3 (95% CI, 6.9–11.1) months.

Figure 1.

image

OS.

Abbreviations: CI, confidence interval; OS, overall survival.

Table 2. Cox model for overall survival.

image

a

Variable eliminated during the backward selection procedure.

b

Order of elimination during the backward selection procedure.

Abbreviations: CI, confidence interval; CT, chemotherapy; EGFR, epidermal growth factor receptor; FP, fluoropyrimidine; HR, hazard ratio; L1, first chemotherapy line; M, metastasis; NS, not significant.

Figure 2.

image

OS and metastasectomy.

Abbreviations: IQR, interquartile range; NE, nonevaluable; OS, overall survival

The median PFS1 was 4.3 months (95% CI, 3.8–5.0; Fig. 3). The PFS1 rates were 22.8% (17.8–28.2) and 12.6% (8.7–17.2) at 12 and 24 months, respectively. The chemotherapy regimen and antiangiogenic/anti‐EGFR therapy did not influence the PFS1 (supplemental online Table 2). In multivariate analysis, the number of metastatic sites (HR = 1.328; 95% CI, 1.012–1.742; p = .04), primary tumor surgery (HR = 0.627; 95% CI, 0.460–0.855; p = .0031), and the metastasectomy (HR = 0.586; 95% CI, 0.395–0.868; p = .0077) were significantly associated with PFS1. In the second‐line setting, the median progression‐free survival was 3.0 months (95% CI, 2.6–3.9). The PFS rates were 12.4% (95% CI, 7.5–18.5) and 2.3% (95% CI, 0.6–6.1) at 12 and 24 months.

Figure 3.

image

PFS1.

Abbreviations: CI, confidence interval; PFS, progression‐free survival; PFS1, progression‐free survival of the first‐line treatment.

Focus on dMMR Tumors

The Cox model did not include the MMR status because too many statuses were not specified (n = 93, 32%). However, we performed an exploratory analysis in the population of 194 patients with MMR status available. Of note, this subgroup was slightly different from the entire population (Table 1). Indeed, patients were younger (65.5 vs. 67.0 years), and fewer had an advanced disease at diagnostic (62.9% vs. 65.9%). Mucinous carcinoma tended to be more frequent (82.2% vs. 80.6%), and the ratio liver/peritoneum metastasis was inferior (1.4 vs. 1.0). The OS of the BRAFV600E‐mutant dMMR population was longer than that of patients with pMMR tumors (30.5 [95% CI, 24.6–42.2] vs. 16.5 [95% CI, 12.5–22.5] months; p = .0087; Fig. 4). The impact of MMR status on OS was still significant even after adjustment on primitive tumor surgery, use of antiangiogenic drug in first‐line metastatic setting, or use of immune checkpoint inhibitors (HR = 0.557; 95% CI, 0.358–0.865; p = .0091). Of note, metastasectomy was the last variable to be eliminated in the multivariate model. The PFS of the first‐line chemotherapy was 4.0 months (95% CI, 3.1–6.4) in dMMR vs. 4.2 months (95% CI, 3.3–5.1) in pMMR tumors.

Figure 4.

image

OS according to MMR status in BRAFV600E‐mutant patients.

Abbreviations: CI, confidence interval; dMMR, deficient mismatch repair; MMR, mismatch repair; pMMR, proficient mismatch repair; OS, overall survival.

Discussion

To our knowledge, the present study is the largest real‐life cohort of BRAFV600‐mutant mCRC. Most analyses published so far are retrospective studies using uni‐ or pauci‐center databases or subgroups analyses from clinical trials. Only few prospective clinical trials dedicated to patients with BRAF mutant mCRC with limited sample size (n = 19–43) have been reported [25], [26], [27]. Our study confirmed the hallmarks of BRAFV600 mutant mCRC tumors and their association with poor prognosis and chemoresistance. The median OS of 20.8 months and the median PFS in the first‐line setting of 4.3 months are consistent with results from clinical trials, even though no selection was prospectively performed. Stage IV disease (metachronous metastases), primary tumor surgery, and metastasectomy were factors significantly associated with OS, confirming prior results observed in mCRC [28]. Furthermore, we pointed out that dMMR and tumor resection are associated with a better prognosis. In this series, the median OS (20.8 months) was longer than the 15 months’ survival duration previously published [12], [29], [30], [31]. One possible explanation is the high rate of patients treated with metastasectomy in our cohort (15.3%). Indeed, these patients are usually excluded from therapeutic clinical trials administrating new drugs and chemotherapy regimens. Excluding patients who have had complete resection of their metastases and median OS of 8.9 (95% CI, 6.57–10.77) months is consistent with published results. When we also left out patients without any chemotherapy (treated with best supportive care only), the median OS was 9.3 months (95% CI, 6.93–11.10). The median PFS in the first‐line setting of 4.3 months was in accordance with literature results [32]. No obvious advantage was observed for irinotecan‐ versus oxaliplatin‐based CT in the first‐line setting, even focusing on patients who received bi‐chemotherapy (data not shown). Moreover, no clinical benefit of antiangiogenic versus anti‐EGFR therapy was observed. It must nevertheless be interpreted with caution because less than 20% of the patients had received anti‐EGFR therapies in the first‐line setting. Contrary to what was previously published, patients with dMMR tumors did not derive any statistically significant survival benefit from the addition of bevacizumab [33], [34]. Furthermore, we included patients in the present cohort before the publication of the TRIBE study, which explains the low percentage of patients treated with tri‐CT (<10%) [20]. However, these 25 patients treated with tri‐CT did not appear to profit from intensification in terms of neither PFS nor OS, whereas 28 patients in the TRIBE study experienced a better overall survival with FOLFOXIRI + bevacizumab (19.0 months) versus FOLFIRI + bevacizumab (10.7 months) [35]. The continuous administration of bevacizumab beyond progression after first‐line chemotherapy did not benefit the few patients treated (n = 29). Although more than half of the patients received a second line of chemotherapy (55.1%), only few of them received a third line (28.2%) in our series, as previously reported in the literature. Indeed, the pooled analysis of the population of 2,530 patients issued from three randomized clinical trials—FOCUS, COIN, and PICCOLO—showed that BRAF‐mutant patients tend to progress rapidly during or following the first‐line chemotherapy, with only 33% of the patients with BRAF‐mutant mCRC having received second‐line treatment versus 51% of the patients with BRAF wild‐type tumors (p < .001) [32]. This point may be relevant and useful for the management and used to design future clinical trials to help in the prioritization of investigational options for early treatment phases in these patients with poor prognosis. We identified two subsets of patients with BRAF‐mutant mCRC with a better outcome, the first being patients with dMMR tumors who experienced a longer OS than those with pMMR tumors. In mCRC, several studies highlighted the poor prognosis of patients with dMMR CRC versus those with pMMR CRC in the metastatic setting, but the prognostic impact of the MMR status in the subgroup of BRAFV600E mutant mCRC has been poorly studied [36], [37]. Indeed, dMMR tumors are rare (≈5%), and dMMR‐BRAF‐mutant tumors consequently constitute a very small subgroup accounting for about 1%–2% of mCRCs [38]. To date, Venderbosch et al. published the largest study evaluating the effect of BRAFV600E mutation and MMR status in patients with mCRC as a pooled analysis of four clinical trials including 3,063 tumors [12]: 153 (5%) were dMMR, 250 (8.2%) harbored a BRAFV600E mutation, and only 53 (1.7%) were dMMR CRC with BRAFV600E‐mutation. Median PFS and OS significantly decreased in patients with dMMR tumors compared with those with pMMR tumors, and in patients with BRAFV600E‐mutant tumors compared with patients harboring BRAF‐wild type tumors. However, the poor prognosis of BRAFV600E‐mutation was exclusively observed in patients with pMMR tumors, in contrast to previous publications that did not show any significant survival difference between patients with dMMR and those with pMMR mCRC. Similarly, Tougeron et al. reported no prognostic impact of BRAF mutation, as well as Lynch syndrome status, in a multivariate analysis of prognostic factors from a large cohort of 284 patients with dMMR mCRC [34]. Our exploratory analysis in the population of patients with a specified MMR status reported that dMMR was significantly associated with better OS, independently of immune checkpoint inhibitor treatment. Median overall survival of patients with mCRC with BRAFV600E and dMMR tumors was 30.5 months compared with 16.5 months in those with pMMR BRAF‐mutant mCRC. This benefit persisted after adjusting on metastasectomy and antiangiogenic treatment. Of note, MMR status was not statistically significantly associated with PFS1. Finally, our results showed that patients for whom resection of liver or peritoneal metastases could have been performed had a better OS. Recent results showed worse prognosis and increased risk of recurrence in patients with BRAF‐mutated tumors who had experienced hepatic resection compared with patients with BRAF‐wild‐type tumors [39]. Gagnière et al. recently identified node‐negative primary tumors, carcinoembryonic antigen (CEA) ≤200 μg/L, and the Clinical Risk Score < 4 as favorable prognostic factors following hepatectomy in BRAF‐mutant patients, allowing them to be proposed aggressive strategies in the metastatic setting [40]. According to the few results reported in patients with peritoneal carcinomatosis, peritoneal resection does not seem to be contraindicated for BRAF‐mutant patients [41]. The surgical resection of liver metastasis contributes to improve survival in patients with BRAF‐mutant mCRC, and surgical resection of peritoneal carcinomatosis for these patients should also be considered as an option and discussed within a multidisciplinary team, especially for young patients with disease control with chemotherapy and low peritoneal carcinomatosis index [42]. Only few patients received either immune checkpoint inhibitors or anti‐BRAF therapies in this cohort. Pembrolizumab, as well as nivolumab with or without ipilimumab, has been approved by the U.S. Food and Drug Administration for the treatment of metastatic refractory dMMR tumors, whatever their BRAF mutational status, and therefore is an option for dMMR BRAF‐mutant metastatic CRC [43], [44]. For BRAF‐mutant tumors, the efficacy of the combination of the anti‐BRAF monoclonal antibody (like vemurafenib) with irinotecan and anti‐EGFR monoclonal antibody (like cetuximab) has been demonstrated in a randomized phase II study, leading to the implementation in the National Comprehensive Cancer Network guidelines of this regimen in BRAFV600E‐mutant mCRC [45]. Furthermore, the results of the BEACON trial, evaluating the combination of a MEK inhibitor, a BRAF inhibitor, and an anti‐EGFR, are greatly awaited because of the magnitude of the effect observed in the first 30 patients [46]. The best therapeutic option for dMMR BRAF‐mutant tumors is unknown at present, but may include immune checkpoint inhibitors or MEK inhibitor plus BRAF inhibitor and an anti‐EGFR. The present cohort is the largest series of BRAF‐mutant mCRC, with very few missing data and patients unselected in contrast to clinical trials proceedings. Therefore, our study contributes to a better knowledge of the population of patients with BRAF‐mutant mCRC known to be associated with poor prognosis [29], [47], [48], [49]. We hypothesize that the poor prognosis in these patients with BRAF‐mutant mCRC may result from the development of chemoresistance mechanisms.

Conclusion

We identify stage IV disease (metachronous metastases), metastasectomy, and primary tumor surgery as prognostic factors that could help treatment decision making. Indeed, when metastases can be resected, metastasectomy should be performed to prolong survival. Chemotherapy regimen and biological agent (antiangiogenic or anti‐EGFR) were not associated with OS or PFS, suggesting that a cytotoxic doublet plus an antiangiogenic or an anti‐EGFR should be a therapeutic option for frail patients not eligible for tri‐CT. Finally, patients with dMMR BRAF‐mutant tumors seem to have a better prognosis and are good candidates for immunotherapy.

See http://www.TheOncologist.com for supplemental material available online.

Acknowledgments

We thank all the clinical research technicians who participated in the data collection, as well as Sophie Darnis, Ph.D., who provided medical writing assistance. This research did not receive any specific grant from research agencies in the public, commercial, or nonprofit sectors.

Author Contributions

Conception/design: Christelle de la Fouchardière

Provision of study material or patients: Christelle de la Fouchardière, Romain Cohen, David Malka, Rosine Guimbaud, Héloïse Bourien, Astrid Lièvre, Wulfran Cacheux, Pascal Artru, Eric François, Marine Gilabert, Emmanuelle Samalin‐Scalzi, Aziz Zaanan, Vincent Hautefeuille, Benoit Rousseau, Hélène Senellart, Romain Coriat, Ronan Flippot, Françoise Desseigne, David Tougeron

Collection and/or assembly of data: Christelle de la Fouchardière, Romain Cohen, David Malka, Rosine Guimbaud, Héloïse Bourien, Astrid Lièvre, Wulfran Cacheux, Pascal Artru, Eric François, Marine Gilabert, Emmanuelle Samalin‐Scalzi, Aziz Zaanan, Vincent Hautefeuille, Benoit Rousseau, Hélène Senellart, Romain Coriat, Ronan Flippot, Françoise Desseigne, David Tougeron

Data analysis and interpretation: Christelle de la Fouchardière, Audrey Lardy‐Cleaud, David Tougeron

Manuscript writing: Christelle de la Fouchardière, Romain Cohen, David Malka, Astrid Lièvre, David Tougeron

Final approval of manuscript: Christelle de la Fouchardière, Romain Cohen, David Malka, Rosine Guimbaud, Héloïse Bourien, Astrid Lièvre, Wulfran Cacheux, Pascal Artru, Eric François, Marine Gilabert, Emmanuelle Samalin‐Scalzi, Aziz Zaanan, Vincent Hautefeuille, Benoit Rousseau, Hélène Senellart, Romain Coriat, Ronan Flippot, Françoise Desseigne, Audrey Lardy‐Cleaud, David Tougeron

Disclosures

Pascal Artru: Roche, Servier, Amgen, Merck, Pierre Fabre, Bayer (C/A); Eric François: Roche, Sanofi, Servier (C/A), Novartis, Amgen, Servier (H); Aziz Zaanan: Baxter, Roche, Merck Serono, Merck Sharp & Dohme, Amgen, Servier, Sanofi, Eli Lilly and Company (C/A); Benoit Rousseau: Bayer, Servier, Roche (C/A). The other authors indicated no financial relationships.

(C/A) Consulting/advisory relationship; (RF) Research funding; (E) Employment; (ET) Expert testimony; (H) Honoraria received; (OI) Ownership interests; (IP) Intellectual property rights/inventor/patent holder; (SAB) Scientific advisory board

References

  • 1.Davies H, Bignell GR, Cox C et al. Mutations of the BRAF gene in human cancer. Nature 2002;417:949–954. [DOI] [PubMed] [Google Scholar]
  • 2.Yao Z, Yaeger R, Rodrik‐Outmezguine VS et al. Tumours with class 3 BRAF mutants are sensitive to the inhibition of activated RAS. Nature 2017;548:234–238. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dankner M, Rose AAN, Rajkumar S et al. Classifying BRAF alterations in cancer: New rational therapeutic strategies for actionable mutations. Oncogene 2018;37:3183–3199. [DOI] [PubMed] [Google Scholar]
  • 4.Gomez‐Roca CA, Delord J, Robert C et al. 535‐Pencorafenib (LGX818), an oral BRAF inhibitor, in patients (patients) with BRAF V600E metastatic colorectal cancer (MCRC): Results of dose expansion in an open‐label phase 1 study. Ann Oncol 2014;25:iv182–iv183. [Google Scholar]
  • 5.Hyman DM, Puzanov I, Subbiah V et al. Vemurafenib in multiple nonmelanoma cancers with BRAF V600 mutations. N Engl J Med 2015;373:726–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Kopetz S, Desai J, Chan E et al. Phase II pilot study of vemurafenib in patients with metastatic BRAF‐mutated colorectal cancer. J Clin Oncol 2015;33:4032–4038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bray F, Ferlay J, Soerjomataram I et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394–424. [DOI] [PubMed] [Google Scholar]
  • 8.Van CE, Lenz HJ, Kohne CH et al. Fluorouracil, leucovorin, and irinotecan plus cetuximab treatment and RAS mutations in colorectal cancer. J Clin Oncol 2015;33:692–700. [DOI] [PubMed] [Google Scholar]
  • 9.Jones JC, Renfro LA, Al‐Shamsi HO et al. (Non‐V600) BRAF mutations define a clinically distinct molecular subtype of metastatic colorectal cancer. J Clin Oncol 2017;35:2624–2630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Rajagopalan H, Bardelli A, Lengauer C et al. Tumorigenesis: RAF/RAS oncogenes and mismatch‐repair status. Nature 2002;418:934. [DOI] [PubMed] [Google Scholar]
  • 11.Ogino S, Nosho K, Kirkner GJ et al. CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut 2009;58:90–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Venderbosch S, Nagtegaal ID, Maughan TS et al. Mismatch repair status and BRAF mutation status in metastatic colorectal cancer patients: A pooled analysis of the CAIRO, CAIRO2, COIN, and FOCUS studies. Clin Cancer Res 2014;20:5322–5330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Samowitz WS, Sweeney C, Herrick J et al. Poor survival associated with the BRAF V600E mutation in microsatellite‐stable colon cancers. Cancer Res 2005;65:6063–6069. [DOI] [PubMed] [Google Scholar]
  • 14.Di NF, Martini M, Molinari F et al. Wild‐type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. J Clin Oncol 2008;26:5705–5712. [DOI] [PubMed] [Google Scholar]
  • 15.Mao C, Liao RY, Qiu LX et al. BRAF V600E mutation and resistance to anti‐EGFR monoclonal antibodies in patients with metastatic colorectal cancer: A meta‐analysis. Mol Biol Rep 2011;38:2219–2223. [DOI] [PubMed] [Google Scholar]
  • 16.Wang Q, Hu WG, Song QB et al. BRAF V600E mutation as a predictive factor of anti‐EGFR monoclonal antibodies therapeutic effects in metastatic colorectal cancer: A meta‐analysis. Chin Med Sci J 2014;29:197–203. [DOI] [PubMed] [Google Scholar]
  • 17.Pietrantonio F, Petrelli F, Coinu A et al. Predictive role of BRAF mutations in patients with advanced colorectal cancer receiving cetuximab and panitumumab: A meta‐analysis. Eur J Cancer 2015;51:587–594. [DOI] [PubMed] [Google Scholar]
  • 18.Rowland A, Dias MM, Wiese MD et al. Meta‐analysis of BRAF mutation as a predictive biomarker of benefit from anti‐EGFR monoclonal antibody therapy for RAS wild‐type metastatic colorectal cancer. Br J Cancer 2015;112:1888–1894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Geissler M, Martens UM, Knorrenschield R et al. 475OmFOLFOXIRI + panitumumab versus FOLFOXIRI as first‐line treatment in patients with RAS wild‐type metastatic colorectal cancer m(CRC): A randomized phase II VOLFI trial of the AIO (AIO‐KRK0109). Ann Oncol 2017;28:mdx393.002. [Google Scholar]
  • 20.Loupakis F, Cremolini C, Lonardi S et al. Subgroup analyses in RAS mutant, BRAF mutant and all‐wt mCRC patients treated with FOLFOXIRI plus bevacizumab (bev) or FOLFIRI plus bev in the TRIBE study. J Clin Oncol 2014;32(suppl 15):3519a. [Google Scholar]
  • 21.Overman MJ, Lonardi S, Wong KYM et al. Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair‐deficient/microsatellite instability‐high metastatic colorectal cancer. J Clin Oncol 2018;36:773–779. [DOI] [PubMed] [Google Scholar]
  • 22.Overman MJ, McDermott R, Leach JL et al. Nivolumab in patients with metastatic DNA mismatch repair‐deficient or microsatellite instability‐high colorectal cancer (CheckMate 142): An open‐label, multicentre, phase 2 study. Lancet Oncol 2017;18:1182–1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Edge SB, Compton CC. The American Joint Committee on Cancer: The 7th edition of the AJCC cancer staging manual and the future of TNM. Ann Surg Oncol 2010;17:1471–1474. [DOI] [PubMed] [Google Scholar]
  • 24.Suraweera N, Duval A, Reperant M et al. Evaluation of tumor microsatellite instability using five quasimonomorphic mononucleotide repeats and pentaplex PCR. Gastroenterology 2002;123:1804–1811. [DOI] [PubMed] [Google Scholar]
  • 25.Bendell JC, Zakari A, Peyton JD et al. A phase II study of FOLFOXIRI plus panitumumab followed by evaluation for resection in patients with metastatic KRAS wild‐type colorectal cancer with liver metastases only. The Oncologist 2016;21:279–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Corcoran RB, Atreya CE, Falchook GS et al. Combined BRAF and MEK inhibition with dabrafenib and trametinib in BRAF V600‐mutant colorectal cancer. J Clin Oncol 2015;33:4023–4031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Cremolini C, Pietrantonio F, Tomasello G et al. Vinorelbine in BRAF V600E mutated metastatic colorectal cancer: A prospective multicentre phase II clinical study. ESMO Open 2017;2:e000241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Faron M, Pignon JP, Malka D et al. Is primary tumour resection associated with survival improvement in patients with colorectal cancer and unresectable synchronous metastases? A pooled analysis of individual data from four randomised trials. Eur J Cancer 2015;51:166–176. [DOI] [PubMed] [Google Scholar]
  • 29.Tol J, Nagtegaal ID, Punt CJ. BRAF mutation in metastatic colorectal cancer. N Engl J Med 2009;361:98–99. [DOI] [PubMed] [Google Scholar]
  • 30.Sanz‐Garcia E, Argiles G, Elez E et al. BRAF mutant colorectal cancer: Prognosis, treatment, and new perspectives. Ann Oncol 2017;28:2648–2657. [DOI] [PubMed] [Google Scholar]
  • 31.Stintzing S, Miller‐Phillips L, Modest DP et al. Impact of BRAF and RAS mutations on first‐line efficacy of FOLFIRI plus cetuximab versus FOLFIRI plus bevacizumab: Analysis of the FIRE‐3 (AIO KRK‐0306) study. Eur J Cancer 2017;79:50–60. [DOI] [PubMed] [Google Scholar]
  • 32.Seligmann JF, Fisher D, Smith CG et al. Investigating the poor outcomes of BRAF‐mutant advanced colorectal cancer: Analysis from 2530 patients in randomised clinical trials. Ann Oncol 2017;28:562–568. [DOI] [PubMed] [Google Scholar]
  • 33.Lenz HJ, Ou FS, Venook AP et al. Impact of consensus molecular subtyping (CMS) on overall survival (OS) and progression free survival (PFS) in patients (pts) with metastatic colorectal cancer (mCRC): Analysis of CALGB/SWOG 80405 (Alliance). J Clin Oncol 2017;35(suppl 15):3511a. [Google Scholar]
  • 34.Tougeron D, Cohen R, Sueur B et al. 533PA large retrospective multicenter study evaluating prognosis and chemosensitivity of metastatic colorectal cancer with microsatellite instability. Ann Oncol 2017;28:mdx393.059. [Google Scholar]
  • 35.Cremolini C, Loupakis F, Antoniotti C et al. FOLFOXIRI plus bevacizumab versus FOLFIRI plus bevacizumab as first‐line treatment of patients with metastatic colorectal cancer: Updated overall survival and molecular subgroup analyses of the open‐label, phase 3 TRIBE study. Lancet Oncol 2015;16:1306–1315. [DOI] [PubMed] [Google Scholar]
  • 36.Tran B, Kopetz S, Tie J et al. Impact of BRAF mutation and microsatellite instability on the pattern of metastatic spread and prognosis in metastatic colorectal cancer. Cancer 2011;117:4623–4632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Goldstein J, Tran B, Ensor J et al. Multicentre retrospective analysis of metastatic colorectal cancer (CRC) with high‐level microsatellite instability (MSI‐H). Ann Oncol 2014;25:1032–1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Blaker H, Alwers E, Arnold A et al. The association between mutations in BRAF and colorectal cancer‐specific survival depends on microsatellite status and tumor stage. Clin Gastroenterol Hepatol 2019;17:455–462. [DOI] [PubMed] [Google Scholar]
  • 39.Margonis GA, Buettner S, Andreatos N et al. Association of BRAF mutations with survival and recurrence in surgically treated patients with metastatic colorectal liver cancer. JAMA Surg 2018;153:e180996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gagniere J, Dupre A, Gholami SS et al. Is hepatectomy justified for BRAF mutant colorectal liver metastases? A multi‐institutional analysis of 1497 patients. Ann Surg 2018. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
  • 41.Franko J, Shi Q, Meyers JP et al. Prognosis of patients with peritoneal metastatic colorectal cancer given systemic therapy: An analysis of individual patient data from prospective randomised trials from the Analysis and Research in Cancers of the Digestive System (ARCAD) database. Lancet Oncol 2016;17:1709–1719. [DOI] [PubMed] [Google Scholar]
  • 42.Péron J, Mercier F, Tuech JJ et al. The location of the primary colon cancer has no impact on outcomes in patients undergoing cytoreductive surgery for peritoneal metastasis. Surgery 2019;165:476–484. [DOI] [PubMed] [Google Scholar]
  • 43.Le DT, Durham JN, Smith KN et al. Mismatch repair deficiency predicts response of solid tumors to PD‐1 blockade. Science 2017;357:409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Le DT, Uram JN, Wang H et al. PD‐1 blockade in tumors with mismatch‐repair deficiency. N Engl J Med 2015;372:2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kopetz S, McDonough SL, Lenz H‐J et al. Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF‐mutant metastatic colorectal cancer (SWOG S1406). J Clin Oncol 2017;35(suppl 15):3505a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kopetz S, Grothey A, Yaeger R et al. Updated results of the BEACON CRC safety lead‐in: Encorafenib (ENCO)+binimetinib (BINI)+cetuximab (CETUX) for BRAFV600E‐mutant metastatic colorectal cancer (mCRC). J Clin Oncol 2019;37(suppl 4):688a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Richman SD, Seymour MT, Chambers P et al. KRAS and BRAF mutations in advanced colorectal cancer are associated with poor prognosis but do not preclude benefit from oxaliplatin or irinotecan: Results from the MRC FOCUS trial. J Clin Oncol 2009;27:5931–5937. [DOI] [PubMed] [Google Scholar]
  • 48.Van Cutsem E, Köhne CH, Láng I et al. Cetuximab plus irinotecan, fluorouracil, and leucovorin as first‐line treatment for metastatic colorectal cancer: updated analysis of overall survival according to tumor KRAS and BRAF mutation status. J Clin Oncol 2011;29:2011–2019. [DOI] [PubMed] [Google Scholar]
  • 49.Bokemeyer C, Van Cutsem E, Rougier P et al. Addition of cetuximab to chemotherapy as first‐line treatment for KRAS wild‐type metastatic colorectal cancer: Pooled analysis of the CRYSTAL and OPUS randomised clinical trials. Eur J Cancer 2012;48:1466–1475. [DOI] [PubMed] [Google Scholar]

Articles from The Oncologist are provided here courtesy of Oxford University Press

RESOURCES