ABSTRACT

BRAF mutation leads to constitutive activation of the MAPK pathway and is associated with the immune-activating molecular subtype of colorectal cancer. Targeted therapy for BRAF V600E–mutant metastatic colorectal cancer (CRC) has significantly improved outcomes for these patients when combined with anti–epithelial growth factor receptor (EGFR) therapy. However, most patients ultimately develop disease progression. We report a case of a patient with metastatic-deficient mismatch repair, BRAF V600E–mutated CRC, who achieved a durable complete response to vemurafenib plus cetuximab and chemotherapy despite initial high burden of disease, including peritoneal involvement. Recent clinical trials of combined anti-EGFR/anti–BRAF V600E therapy in BRAF V600E-mutant CRCs have found a few instances of robust response. Further study of combined targeted and chemotherapeutic regimens and the immunogenic properties of BRAF mutation may yield promising results.

Mutations involving BRAF proto-oncogene are present in 3% to 4% of cancers, of which BRAF V600E is the most common variant.[1]BRAF V600E mutations have an estimated prevalence of 8% to 12% in metastatic colorectal cancer (CRC). Of note, nearly 80% of metastatic-deficient mismatch repair (MSI-H/dMMR) CRCs are BRAF mutant due to hypermethylation of the MLH1 promotor.[2,3] BRAF encodes for a serine/threonine kinase involved in the MAPK signaling pathway that, when mutated, leads to constitutive activation of the kinase, resulting in cellular proliferation. BRAF V600E mutations are generally associated with a poor prognosis, with studies demonstrating limited survival in stage II-IV CRC and limited response to conventional therapeutic agents.[3,4]

Vemurafenib, a BRAF inhibitor, was first approved in unresectable BRAF V600E-mutated melanoma after demonstrating increased overall and progression-free survival. In contrast, BRAF inhibition as a monotherapy for BRAF V600E–mutant CRC did not demonstrate clinical activity.[5] Subsequent efforts identified acquired resistance through intratumor heterogeneity of KRAS/NRAS mutations (upstream of BRAF in MAPK pathway) and rapid feedback activation of EGFR as potential mechanisms of resistance.[5,6] The BEACON trial, which assessed the combination of the BRAF inhibitor encorafenib with the anti–epithelial growth factor receptor (EGFR) antibody cetuximab demonstrated improved overall survival and progression-free survival compared with chemotherapy (8.4 vs. 5.4 median overall survival; 4.2 vs. 1.2 median progression-free survival) and lead to Food and Drug Administration approval in the second line for unresectable BRAF V600E mutant CRC.[7] Despite meaningful improvements in survival with this regimen, the vast majority of patients nevertheless develop progressive disease. Herein, we report an unusual case of a patient with metastatic BRAF V600E–mutant CRC with a durable complete response (CR) to vemurafenib plus cetuximab and chemotherapy. The patient consented to the publication of their case and photographs.

A 68-year-old man without significant medical comorbidities presented to his primary care physician with abdominal pain and anemia evaluated by colonoscopy and endoscopy, which demonstrated a circumferential mass in the ascending colon. A biopsy of the mass revealed a poorly differentiated carcinoma. Shortly thereafter, the patient was hospitalized after developing fever and abdominal tenderness. He then underwent an emergent right hemicolectomy, revealing perforation of the mass. Pathology again demonstrated a poorly differentiated carcinoma, favoring a primary colon cancer. The mass had invaded the pericolic soft tissues. Lymphovascular invasion was present. None of the 21 dissected lymph nodes were negative for carcinoma. Contemporaneous CT of the abdomen was only notable for a nonspecific liver lesion that was negative for malignancy on biopsy. The clinical stage at diagnosis was pT3N0M0.

After diagnosis, the patient was initiated on systemic chemotherapy with FOLFOX. Interval imaging after six cycles demonstrated pericolic necrotic lymphadenopathy and 5.5 × 5.2 cm local recurrence around the cecum, involving the adjacent small-bowel retroperitoneum, most consistent with carcinomatosis. Next-generation sequencing (NGS) of the tissue revealed BRAF V600E (c.1799T>A p.V600E) and PIK3CA (c.1173A>G p.I391M) mutations in addition to methylation of the MLH1 promotor consistent with an MSI-H phenotype. The patient was initiated on FOLFIRI plus cetuximab for two cycles with radiographic progression before enrolling in a Phase I clinical trial of vemurafenib in combination with cetuximab and irinotecan. Vemurafenib dosing was initially 720 mg taken orally. Approximately 2 months after the initiation of the trial, computed tomography (CT) of the abdomen revealed near-complete resolution of the recurrent disease along with significant radiologic response in lymphadenopathy. After completing five cycles, the patient developed a bowel obstruction complicated by perforation and abscess. Therapy was held. Approximately 2 months later, the patient redeveloped the bowel obstruction and underwent resection of the ileum, mesenteric mass resection, ileocolic anastomosis, and primary ileotransverse colostomy. Pathology from the surgery revealed necrosis and fibrosis of the mesenteric mass, possibly consistent with a necrotic lymph node. The resected small bowel and colon demonstrated multiple adhesions with foreign body giant cell reaction, fibrosis, and associated chronic mucosal injury, but no tumor tissue was identified. Roughly 2 months after surgery, a repeat abdomen CT demonstrated no evidence of disease, and therapy was resumed. The patient developed grade 2-3 diarrhea, requiring dose reduction of vemurafenib to 240 mg twice daily and eventual cessation of irinotecan by 18 months after the start of treatment. Cetuximab infusion was discontinued 60 months after initiation of therapy due to unclear additional benefits and patient preference.

The patient remained on the trial protocol for 89 months, with routine imaging every 3 to 6 months, before transitioning to standard-of-care use of vemurafenib at the same dosing. The patient has maintained a complete radiographic response at the last follow-up, 120 months after initiating vemurafenib (Fig. 1). Carcinoembryonic antigen levels obtained in tandem with imaging ranged between 3.3 and 11.4 ng/mL on treatment. Circulating tumor DNA was unavailable at the time of the initial treatment.

Figure 1

CT images of BRAF V600E/MSI-H case. CT images of the abdomen and pelvis illustrate the recurrent primary tumor surrounding the cecum (red arrow) and malignant pericolic lymphadenopathy (yellow arrow). Serial imaging studies at 2, 6, and 120 months show excellent, durable response to treatment with complete resolution of the primary tumor and lymphadenopathy after the resection of the mesenteric mass (possibly a necrotic lymph node) at the 6-month follow-up. CT, computed tomography.

Figure 1

CT images of BRAF V600E/MSI-H case. CT images of the abdomen and pelvis illustrate the recurrent primary tumor surrounding the cecum (red arrow) and malignant pericolic lymphadenopathy (yellow arrow). Serial imaging studies at 2, 6, and 120 months show excellent, durable response to treatment with complete resolution of the primary tumor and lymphadenopathy after the resection of the mesenteric mass (possibly a necrotic lymph node) at the 6-month follow-up. CT, computed tomography.

Close modal

We present a patient with BRAF V600E mutation and MSI-H metastatic (m)CRC, with no evidence of disease 10 years after the initiation of combined BRAF/EGFR inhibitor therapy despite the high disease burden, including peritoneal involvement. Of note, the patient had a robust and rapid response, which resulted in a small bowel obstruction, necessitating surgical intervention. While durable CRs have been observed with immune checkpoint inhibitors, such as in the case of dMMR CRC after exposure to immune checkpoint blockade, it is uncommon to see a sustained CR with targeted therapies given the heterogeneity of driver alterations and activation of feedback pathways, which lead to the eventual development of therapeutic resistance. In the BEACON trial, which assessed the BRAF inhibitor and anti-EGFR combination (without irinotecan), 4.5% (n = 10) of patients achieved a CR. However, the duration of response is unknown, with the longest follow-up at 18 months.[7] In the SWOG S1406 study, which randomized patients with BRAF V600E–mutant mCRC (n = 106) to receive either irinotecan and cetuximab with vemurafenib (VIC regimen) or irinotecan and cetuximab, no CRs were observed among the 17% of patients who had a response, indicating it is an uncommon occurrence.[8]

One possible explanation for the durable CR is that inhibiting BRAF heralded a robust anti-tumor immune response. BRAF mutation correlates with increased PD-L1 expression and with the immune-activating molecular subtype of CRC, consensus molecular subtype 1.[9,10] Moreover, there appear to be higher levels of immune-cell infiltration on BRAF-mutated tumors compared with their wild-type counterparts.[11] The patient in the case also had an MSI-H tumor, which is a phenotype associated with increased immunogenicity and immune infiltrate, possibly explaining the improved overall survival with BRAF V600E-mutant/MSI-H CRC compared with BRAF V600E–mutant/microsatellite stable (MSS) phenotype.[12] A single-arm, phase I/II trial studying the combination of encorafenib (BRAF inhibitor), cetuximab, and nivolumab (PD-L1 inhibitor) produced encouraging results, with overall response rate (ORR) of 45% and disease control rate (DCR) of 95% in MSS, BRAF V600E–mutated mCRC suggesting a possible benefit with the addition of immunotherapy in this context.[13] Furthermore, a Phase II study of combined PD-L1, BRAF, and MEK inhibition in BRAF V600E–mutant mCRCs revealed greater upregulation of T-cell signatures in treated tumor cells of patients with better clinical outcomes, with an ORR of 24.3% and DCR of 70.3%.[14,15] Results from the ongoing randomized phase III SEAMARK trial (NCT05217446), looking at the combination of encorafenib (BRAF inhibitor), cetuximab, and pembrolizumab (PD-L1 inhibitor) in MSI-H mCRC, are awaited.[16] An alternative hypothesis is that the combination of irinotecan with EGFR/BRAF-combined may have sufficiently eradicated the tumor cells before developing resistance. A subset of CRCs may rely extensively on a singular pathway, in which case inhibition produces a robust response. For many targeted therapies, it may be a matter of sufficient blockade of the signaling pathway to prevent resistance. Therefore, more potent and selective inhibitors and drug combinations would be of interest.

There are few biomarkers that have been shown to identify patients with tumors responsive to BRAF-targeted therapy. In the SWOG S1406 trial, a decline in circulating tumor DNA BRAF V600E–mutant variant allele frequency corresponded with improved progression-free survival.[8,17] Other biomarkers that may be of significance include RNF43 and transcriptomic signatures.[18,19]

Given the patient’s complex clinical course, the contribution of various therapeutics toward his outcome is unclear. For example, it is difficult to exclude the possibility that the patient would have achieved a similar response to irinotecan, vemurafenib, or cetuximab alone or as a part of a doublet regimen. However, a study of BRAF V600E–mutant mCRC tumor xenografts found the most tumor growth inhibition in specimens receiving a combination of FOLFIRI with encorafenib plus cetuximab, compared with FOLFIRI or encorafenib plus cetuximab alone, suggesting an additive benefit.[20] Additionally, molecular features associated with response beyond NGS were not examined in this case. In summary, this case demonstrates that therapies targeting genomic alterations present in CRC, such as BRAF V600E, can confer sustained CR in the right clinical context. Further work is needed to harness the potential of targeted therapy in more patients.

1.
Owsley
J,
Stein
MK,
Porter
J,
et al
Prevalence of class I-III BRAF mutations among 114,662 cancer patients in a large genomic database
.
Exp Biol Med (Maywood)
.
2021
;
246
:
31
39
.
2.
Tabernero
J,
Ros
J,
Élez
E.
The evolving treatment landscape in BRAF-V600E-mutated metastatic colorectal cancer
.
Am Soc Clin Oncol Educ Book
.
2022
;
42
:
1
10
.
3.
Clarke
CN,
Kopetz
ES.
BRAF mutant colorectal cancer as a distinct subset of colorectal cancer: clinical characteristics, clinical behavior, and response to targeted therapies
.
J Gastrointest Oncol
.
2015
;
6
:
660
667
.
4.
De Roock
W,
Claes
B,
Bernasconi
D,
et al
Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal cancer: a retrospective consortium analysis
.
Lancet Oncol
.
2010
;
11
:
753
762
.
5.
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
.
6.
Prahallad
A,
Sun
C,
Huang
S,
et al
Unresponsiveness of colon cancer to BRAF(V600E) inhibition through feedback activation of EGFR
.
Nature
.
2012
;
483
:
100
103
.
7.
Kopetz
S,
Grothey
A,
Yaeger
R,
et al
Encorafenib, binimetinib, and cetuximab in BRAF V600E-mutated colorectal cancer
.
N Engl J Med
.
2019
;
381
:
1632
1643
.
8.
Kopetz
S,
Guthrie
KA,
Morris
VK,
et al
Randomized trial of irinotecan and cetuximab with or without vemurafenib in BRAF-mutant metastatic colorectal cancer (SWOG S1406)
.
J Clin Oncol
.
2021
;
39
:
285
294
.
9.
Guinney
J,
Dienstmann
R,
Wang
X,
et al
The consensus molecular subtypes of colorectal cancer
.
Nat Med
.
2015
;
21
:
1350
1356
.
10.
Rosenbaum
MW,
Bledsoe
JR,
Morales-Oyarvide
V,
et al
PD-L1 expression in colorectal cancer is associated with microsatellite instability, BRAF mutation, medullary morphology and cytotoxic tumor-infiltrating lymphocytes
.
Mod Pathol
.
2016
;
29
:
1104
1112
.
11.
Edin
S,
Gylling
B,
Li
X,
et al
Opposing roles by KRAS and BRAF mutation on immune cell infiltration in colorectal cancer - possible implications for immunotherapy
.
Br J Cancer
.
2024
;
130
:
143
150
.
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
.
13.
Morris
VK,
Parseghian
CM,
Escano
M,
et al
Phase I/II trial of encorafenib, cetuximab, and nivolumab in patients with microsatellite stable, BRAFV600E metastatic colorectal cancer
. Presented at Proceedings from the American Society of Clinical Oncology Gastrointestinal Cancers Symposium,
San Francisco, CA
; January 20–22,
2022
.
14.
Tian
J,
Chen
JH,
Chao
SX,
et al
Combined PD-1, BRAF and MEK inhibition in BRAFV600E colorectal cancer: a phase 2 trial
.
Nat Med
.
2023
;
29
:
458
466
.
15.
Ros
J,
Rodríguez-Castells
M,
Saoudi
N,
et al
Treatment of BRAF-V600E mutant metastatic colorectal cancer: new insights and biomarkers
.
Expert Rev Anticancer Ther
.
2023
;
23
:
797
806
.
16.
Elez
E,
Kopetz
S,
Tabernero
J,
et al
SEAMARK: phase II study of first-line encorafenib and cetuximab plus pembrolizumab for MSI-H/dMMR BRAFV600E-mutant mCRC
.
Future Oncol
.
2024
;
20
:
653
663
.
17.
Ros
J,
Matito
J,
Villacampa
G,
et al
Plasmatic BRAF-V600E allele fraction as a prognostic factor in metastatic colorectal cancer treated with BRAF combinatorial treatments [published correction appears in Ann Oncol. 2024 Aug 12:S0923-7534(23)00796-2
. ].
Ann Oncol
.
2023
;
34
:
543
552
.
18.
Kopetz
S,
et al
Molecular correlates of clinical benefit in previously treated patients (pts) with BRAF V600E-mutant metastatic colorectal cancer (mCRC) from the BEACON study
.
J Clin Oncol
.
2021
;
39
(
suppl; abstr 3513
):
15s
.
19.
Elez
E,
et al
RNF43 mutations predict response to anti-BRAF/EGFR combinatory therapies in BRAFV600E metastatic colorectal cancer
.
Nat Med
.
2022
;
28
:
2162
2170
.
20.
Napolitano
S,
Woods
M,
Lee
HM,
et al
Antitumor efficacy of dual blockade with encorafenib + cetuximab in combination with chemotherapy in human BRAFV600E-mutant colorectal cancer
.
Clin Cancer Res
.
2023
;
29
:
2299
2309
.

Author notes

Sources of Support: None. Conflicts of Interest: Scott Kopetz reports consultant for Genentech, EMD Serono, Merck, Holy Stone Healthcare, Novartis, Lilly, Boehringer Ingelheim, AstraZeneca/MedImmune, Bayer Health, Redx Pharma, Ipsen, HalioDx, Lutris, Jacobio, Pfizer, Repare Therapeutics, Inivata, GlaxoSmithKline, Jazz Pharmaceuticals, Iylon, Xilis, Abbvie, Amal Therapeutics, Gilead Sciences, Mirati Therapeutics, Flame Biosciences, Servier, Carina Biotech, Bicara Therapeutics, Endeavor BioMedicines, Numab, Johnson and Johnson/Janssen, Genomic Health, Frontier Medicines, Replimune, Taiho Pharmaceutical, Cardiff Oncology, Ono Pharmaceutical, Bristol-Myers Squibb,-Medarex, Amgen, Tempus, Foundation Medicine, Harbinger Oncology, Takeda, CureTeq, Zentalis, Black Stone Therapeutics, NeoGenomics Laboratories, Accademia Nazionale Di Medicina, and Tachyon Therapeutics; research support from Sanofi, Biocartis, Guardant Health, Array BioPharma, Genentech/Roche, EMD Serono, MedImmune, Novartis, Amgen, Lilly, and Daiichi Sankyo; and ownership in Lutris, Iylon, Frontier Medicines, Xilis, and Navire outside the submitted work. David S. Hong reports research supportfrom Bayer, Lilly, Genentech, LOXO, Pfizer, Amgen, Mirati, Ignyta, Merck, Daichii-Sanko, Eisai, Adaptimmune, Abbvie, Astra-Zeneca, BMS, Genmab, Infinity, Kite, Kyowa, Medimmune, Molecular Template, Novartis, and Takeda; personal fees from Mirna and LOXO; and other support from Bayer, Baxter, Guidepoint Global, Oncoresponse, Janssen, and Molecular Match outside the submitted work. The remaining authors have no disclosures.

This work is published under a CC-BY-NC-ND 4.0 International License.