Abstract

Background

Impact of BRAF V600E mutations (BRAFV600E), a poor prognostic factor in metastatic colorectal cancer, is lacking in non-CRC gastrointestinal (GI) cancers including pancreatic (PDAC), gastric/gastroesophageal (GEA), hepatocellular carcinoma (HCC), and cholangiocarcinoma (CCA). Due to tumor-agnostic approvals for patients with BRAFV600E, understanding the frequency and impact of BRAF alterations across non-CRC GI cancers is essential for clinical decision-making.

Methods

Patients with PDAC, GEA, HCC, or CCA who had cell-free DNA detected on Guardant360 (Guardant Health) from 2020 to 2023 were queried. Prevalence of characterized BRAF genomic alterations (GA) was calculated; GAs were grouped by class (Class I/II/III). The Chi-squared test assessed differences between cancer types. A subset of patients had outcomes analysis using GuardantINFORM, a real-world clinicogenomic database, to derive real-world overall survival (rwOS).

Results

Of 32 480 included patients, BRAF GAs were identified in 4.4%; 19% were BRAFV600E (0.81% prevalence overall). CCA had the highest rate of BRAF GAs and BRAFV600E (P < .01); HCC and GEA had the highest rates of BRAF class II/III alterations. There were no significant differences in rwOS by alteration class or cancer type; numeric differences were observed by alteration class. Few patients were treated with BRAF inhibitors (2.2%). Prevalence of co-occurring alterations was unique by cancer type.

Conclusions

Frequency of BRAF GAs, including BRAFV600E, in non-CRC GI cancers detected by liquid biopsy is similar to tissue-based rates and can be reliably used to assess BRAF status. BRAF GAs have mixed prognostic implications on survival for patients with non-CRC GI malignancies that warrant further exploration.

Implications for practice

Novel drug therapeutics targeting BRAF alterations are under development. However, the frequency and types of BRAF alteration in non-colorectal gastrointestinal cancers are not well defined. In this study, using liquid biopsy, we found that the highest frequency of class 1 BRAF alterations is present in cholangiocarcinoma while Class II/III BRAF alterations are more common in hepatocellular cancer and gastroesophageal cancer. We did not observe the detrimental effect of BRAF mutations on overall survival in these tumor types. Despite the tumor-agnostic FDA approval of BRAF inhibitors, their utilization was low in clinical practice in non-colorectal gastrointestinal cancers.

Background

BRAF is a protein kinase that functions in cell signaling pathways impacting cellular growth and differentiation and BRAF mutations have been implicated in oncogenic pathways.1BRAF mutations have been found to have a prognostic impact and may guide therapeutic decisions in patients with multiple solid malignancies, including colorectal cancer (CRC),2-4 melanoma,5,6 and non-small cell lung cancer (NSCLC).7BRAFV600E is the most frequently observed and best-studied mutation, however, new studies have evaluated BRAFnon-V600E mutations as well, identifying distinct phenotypic and prognostic implications of these less frequently observed variants.8-11

BRAF mutations may be characterized as class I, II, or III. Class I mutations include BRAFV600E and result in constitutive activation of BRAF monomers independent of RAS, while class II mutations lead to constitutive activation of BRAF dimers independent of RAS.8 Alternatively, class III mutations lead to more avid binding of RAS and CRAF. In patients with CRC, class I mutations (BRAFV600E) are associated with proximal malignancies that are poorly differentiated, conferring poorer survival.2,12,13 More recent studies have sought to better understand BRAFnon-V600E mutations, which may be observed in approximately 2.2% of CRC cases.9 In CRC, class II mutations generally confer a poorer prognosis than BRAFWT, while class III mutations have similar survival compared to BRAFWT.8 However, BRAFV600E and other classes of BRAF alterations and their clinical implications have not been broadly assessed in other gastrointestinal (GI) malignancies, including pancreatic cancer (PDAC), gastric/gastroesophageal cancer (GEA), hepatocellular carcinoma (HCC), and cholangiocarcinoma (CCA).14

Recently, the United States Food and Drug Administration (FDA) approved the combination of dabrafenib, a BRAF inhibitor, and trametinib, a MEK inhibitor, for advanced solid malignancies harboring BRAFV600E mutation. This is based on the results of the ROAR trial, an open-label phase II basket trial that enrolled patients with rare malignancies harboring BRAFV600E mutation. Interval analysis demonstrated impressive results in multiple tumor types with overall response rates of 47% in biliary tract cancer,15 56% in anaplastic thyroid cancer,16 33% in high-grade glioma, and 69% in low-grade glioma.17 Additionally, recent studies in metastatic CRC demonstrated prolonged preservation in quality of life in those treated with BRAF inhibitors.18 As such, this further highlights the importance of better assessing BRAFV600E prevalence in expanded cancer types, including non-CRC GI malignancies.

To detect BRAF mutations, cell-free DNA (cfDNA) may be used clinically and/or in research studies. Studies have demonstrated high concordance of genomic alterations detected via cfDNA analysis with tissue biopsy in multiple GI malignancies including CRC,19 PDAC,20 and GEA.21 The advantage of cfDNA includes its minimally invasive nature, faster turnaround time, and low failure rate for next-generation sequencing testing. Assessing BRAF status via cfDNA may have utility in identifying patients who may have matched therapy or clinical trial options. As such, we sought to evaluate the prevalence of BRAFV600E and BRAFnon-V600E alterations in non-CRC GI malignancies using cfDNA and report select patient outcomes.

Methods

Prevalence assessment

Patients with advanced PDAC, GEA, HCC, or CCA who had cfDNA detected as part of routine clinical testing (Guardant360, Guardant Health) in the United States from October 1, 2020 to September 30, 2023 were retrospectively assessed to determine the prevalence of BRAF genomic alteration (GA). Detection of cfDNA may be observed in 85%–90% of patients assessed, depending on tumor shed.22,23 Only the first test for patients was included if multiple tests were performed. Guardant360 is a CLIA-certified, CAP-accredited, cfDNA next-generation sequencing (NGS) assay with prior analytical and clinical validations reported.23,24 Guardant360 can detect genomic alterations in up to 83 genes and, in select genes, identify copy number amplifications, rearrangement/fusions, and insertions/deletions.

Patients with cfDNA detected were sorted by cancer type and stratified by BRAF status: no BRAF alteration detected, BRAFV600E (class I), BRAF class II, BRAF class III, or other predicted BRAF driver alteration as previously published (listed in Supplementary Table S1).14BRAF amplifications and/or characterized alterations excluding variants of uncertain significance and synonymous alterations were removed for analysis by class. If patients had more than one BRAF alteration, then they were assigned the class with the highest alteration (eg, BRAFV600E and BRAF class II alterations detected in the same patient would assign the patient to the V600E/class I group). Clinical factors, such as diagnosis, clinical status (newly diagnosed, not responding to therapy), age, and sex were derived from clinician-completed test requisition forms; comparisons across cancer types were assessed using the chi-squared test with P-value for the significance of P < .05.

Outcomes analysis: overall survival by braf status

Outcomes of patients with BRAF alterations in the prevalence analysis were assessed using GuardantINFORM, a real-world evidence database that includes anonymized genomic data and structured payer claims from inpatient and outpatient settings across academic and community practices. GuardantINFORM does not include clinical features that are not coded as claims (eg, biomarkers assessed by other tests, clinical response to cancer treatment, etc.).

Patient treatments were recapitulated over the study period using claims data in GuardantINFORM. Patients were required to have 2 metastatic diagnosis codes for their cancer to confirm their cancer type and metastatic state as well as capture baseline characteristics. Patients were stratified by cancer type and classified BRAF alteration group as performed in the Prevalence cohort for V600E/class I, class II, and class III. Co-occurring alterations were assessed for patients with BRAF V600E alterations by cancer type regardless of availability of evaluable outcomes, where genes were altered in at least 1% of the cancer-type specific population. Then, outcomes were assessed for all BRAF classes, where patients were required to have treatment and evaluable outcomes (eg, no conflicting death dates). Real-world overall survival (rwOS) was evaluated in months and defined as the time from G360 testing report date until death from any cause; patients were censored at the last claim activity. Log-rank tests were used to analyze differences. Demographics and Elixhauser comorbidity scores were compared across BRAF groups for each cancer type using the chi-squared test. Significance was defined as P < .05 for all tests. Of note, HCC and “other putative driver BRAF alterations had a low number of outcomes available and thus were not included in outcomes analysis.

Ethics statement

De-identified research datasets generated by Guardant Health are approved by the Adverra IRB with a waiver of consent. The GuardantINFORM database is a fully deidentified database that complies with sections 164.514(a)-(n)1ii of the U.S. Health Insurance Portability and Accountability Act (HIPAA) regarding the determination and documentation of statistically deidentified data.

Results

Prevalence and features of BRAF genomic alterations

The study cohort consisted of 32 480 patients with advanced PDAC, GEA, HCC, or CCA who underwent cfDNA testing and had at least one alteration detected in circulating tumor DNA (ctDNA). Median age of the study cohort was 68 years (range: 18-102 years) with male preponderance (58.7%). Clinical status was reported for 89% (m = 29,124) with ~65% having cfDNA testing at the time of diagnosis and the remainder (~35%) performed when not responding to therapy (as reported by ordering clinicians); these rates did not differ by tumor type (Table 1).

Table 1.

Demographics and BRAF prevalence assessment by cancer type.

Total nPDACGEACCAHCC
Cohorts (n, %)32 48015 578 (48.0)8435 (26.0)6189 (19.1)2278 (7.0)
Male sex (n, %)19 084 (58.7)8251 (53.0)6080 (72.1)3055 (49.4)1697 (74.5)
Median age, years68 (18-102)69 (20-102)66 (18-100)68 (18-98)67 (16-97)
Percent newly diagnosed64.9364.1863.8666.5269.54
BRAF alterations (n,%)969 (2.98)361 (2.3)219 (2.6)328 (5.3)61 (2.7)
BRAF V600E (class I) 264 (0.81) 79 (0.51) 53 (0.63) 114 (1.8) 18 (0.79)
BRAF class II 256 (0.79) 117 (0.75) 57 (0.68) 69 (1.1) 13 (0.57)
BRAF class III 329 (1.01) 103 (0.66) 87 (1.03) 119 (1.9) 20 (0.88)
 Other predicted driver 120 (0.37) 62 (0.40) 22 (0.26) 26 (0.40) 10 (0.44)
Total nPDACGEACCAHCC
Cohorts (n, %)32 48015 578 (48.0)8435 (26.0)6189 (19.1)2278 (7.0)
Male sex (n, %)19 084 (58.7)8251 (53.0)6080 (72.1)3055 (49.4)1697 (74.5)
Median age, years68 (18-102)69 (20-102)66 (18-100)68 (18-98)67 (16-97)
Percent newly diagnosed64.9364.1863.8666.5269.54
BRAF alterations (n,%)969 (2.98)361 (2.3)219 (2.6)328 (5.3)61 (2.7)
BRAF V600E (class I) 264 (0.81) 79 (0.51) 53 (0.63) 114 (1.8) 18 (0.79)
BRAF class II 256 (0.79) 117 (0.75) 57 (0.68) 69 (1.1) 13 (0.57)
BRAF class III 329 (1.01) 103 (0.66) 87 (1.03) 119 (1.9) 20 (0.88)
 Other predicted driver 120 (0.37) 62 (0.40) 22 (0.26) 26 (0.40) 10 (0.44)
Table 1.

Demographics and BRAF prevalence assessment by cancer type.

Total nPDACGEACCAHCC
Cohorts (n, %)32 48015 578 (48.0)8435 (26.0)6189 (19.1)2278 (7.0)
Male sex (n, %)19 084 (58.7)8251 (53.0)6080 (72.1)3055 (49.4)1697 (74.5)
Median age, years68 (18-102)69 (20-102)66 (18-100)68 (18-98)67 (16-97)
Percent newly diagnosed64.9364.1863.8666.5269.54
BRAF alterations (n,%)969 (2.98)361 (2.3)219 (2.6)328 (5.3)61 (2.7)
BRAF V600E (class I) 264 (0.81) 79 (0.51) 53 (0.63) 114 (1.8) 18 (0.79)
BRAF class II 256 (0.79) 117 (0.75) 57 (0.68) 69 (1.1) 13 (0.57)
BRAF class III 329 (1.01) 103 (0.66) 87 (1.03) 119 (1.9) 20 (0.88)
 Other predicted driver 120 (0.37) 62 (0.40) 22 (0.26) 26 (0.40) 10 (0.44)
Total nPDACGEACCAHCC
Cohorts (n, %)32 48015 578 (48.0)8435 (26.0)6189 (19.1)2278 (7.0)
Male sex (n, %)19 084 (58.7)8251 (53.0)6080 (72.1)3055 (49.4)1697 (74.5)
Median age, years68 (18-102)69 (20-102)66 (18-100)68 (18-98)67 (16-97)
Percent newly diagnosed64.9364.1863.8666.5269.54
BRAF alterations (n,%)969 (2.98)361 (2.3)219 (2.6)328 (5.3)61 (2.7)
BRAF V600E (class I) 264 (0.81) 79 (0.51) 53 (0.63) 114 (1.8) 18 (0.79)
BRAF class II 256 (0.79) 117 (0.75) 57 (0.68) 69 (1.1) 13 (0.57)
BRAF class III 329 (1.01) 103 (0.66) 87 (1.03) 119 (1.9) 20 (0.88)
 Other predicted driver 120 (0.37) 62 (0.40) 22 (0.26) 26 (0.40) 10 (0.44)

BRAF alterations were identified in 1375 (4.4%) patients, of which 407 were amplifications. Of patients who had BRAF mutations (n = 969), 264 were BRAFV600E/Class I for an overall prevalence of 0.81% (Figure 1). Twenty-four patients with BRAFV600E/class I alterations also harbored BRAF amplifications (1.7%); 23 patients had MSI-H detected (1.6%). When assessing differences by tumor type, cholangiocarcinoma had the highest rate of BRAF alterations (> 5%) including the highest rate of BRAFV600E/class I alterations compared to other tumor types (Table 1, P < .01). Gastroesophageal cancers and HCC had higher rates of class II/III alterations than V600E/class I alterations.

This figure describes our patient cohort. 32,480 patients were included in prevalence analyses and 28,895 patients were included in outcome analysis. BRAF alteration was detected in 969 (2.98%) patients. In this cohort 271 patients had evaluable survival outcomes and had treatment information.
Figure 1.

Consort diagram. aOwsley et al.14 Abbreviations: CCA, cholangiocarcinoma; GEA, gastroesophageal cancer; HCC, hepatocellular carcinoma, PDAC, pancreatic cancer.

When compared to published rates of classified BRAF alterations identified on tissue-based testing,14 rates were higher for each cancer type assessed via ctDNA when comparable: 3.8% tissue-based CCA BRAF alteration rate compared to 5.3% on ctDNA, 1.12% tissue-based rate for GEA compared to 2.6% on ctDNA, and 1.49% tissue-based rate for PDAC compared to 1.49% for ctDNA. Notably, there was not a tissue-based reference for hepatocellular carcinoma available in the literature, likely due to its diagnostic protocol that may not require tissue biopsy.

Outcomes analysis

Three-hundred fourteen patients with BRAF alterations were assessed using GuardantINFORM for co-alteration analysis (n = 155 with CCA, n = 67 with GEA, n = 92 with PDAC; Supplement Figure S1A-C).

Outcome data were available for 271 patients with class I, II, or III BRAF alterations comprising 31.9% of the prevalence cohort (n = 100 with CCA, n = 95 with PDAC, and n = 76 with GEA; Table 2). When assessing BRAF alterations by class, higher rates of class II alterations and lower rates of class III alterations were observed in the outcomes cohort compared to the overall prevalence cohort (P < .0001).

Table 2.

Real-world overall survival by cancer type and by BRAF mutational classification.

Cholangiocarcinoma
Class IClass IIClass III
Subjects 46 36 13
 Events19155
 Censored27218
Median rwOS in months 12.4711.6710.63
 95% CI10.63-24.579.57-NR9.1-NR
Log-rank P across groupsP = .45
 Pancreatic cancer
Class IClass IIClass III
Subjects 33 4720
 Events9206
 Censored242714
Median rwOS in months NR 11.30 NR
 95% CI10.17-NR7.33-NR6.10-NR
Log-rank p across groupsP = 0.36
Gastro/esophageal cancer
Class IClass IIClass III
Subjects 18 4414
 Events4144
 Censored142910
Median rwOS in months 14.70 NR 31.60
 95% CI10.90-NR8.93-NR22.67-NR
Log-rank P across groups P = .76
Cholangiocarcinoma
Class IClass IIClass III
Subjects 46 36 13
 Events19155
 Censored27218
Median rwOS in months 12.4711.6710.63
 95% CI10.63-24.579.57-NR9.1-NR
Log-rank P across groupsP = .45
 Pancreatic cancer
Class IClass IIClass III
Subjects 33 4720
 Events9206
 Censored242714
Median rwOS in months NR 11.30 NR
 95% CI10.17-NR7.33-NR6.10-NR
Log-rank p across groupsP = 0.36
Gastro/esophageal cancer
Class IClass IIClass III
Subjects 18 4414
 Events4144
 Censored142910
Median rwOS in months 14.70 NR 31.60
 95% CI10.90-NR8.93-NR22.67-NR
Log-rank P across groups P = .76

Abbreviations: CI, confidence interval; rwOS: real-world overall survival.

Table 2.

Real-world overall survival by cancer type and by BRAF mutational classification.

Cholangiocarcinoma
Class IClass IIClass III
Subjects 46 36 13
 Events19155
 Censored27218
Median rwOS in months 12.4711.6710.63
 95% CI10.63-24.579.57-NR9.1-NR
Log-rank P across groupsP = .45
 Pancreatic cancer
Class IClass IIClass III
Subjects 33 4720
 Events9206
 Censored242714
Median rwOS in months NR 11.30 NR
 95% CI10.17-NR7.33-NR6.10-NR
Log-rank p across groupsP = 0.36
Gastro/esophageal cancer
Class IClass IIClass III
Subjects 18 4414
 Events4144
 Censored142910
Median rwOS in months 14.70 NR 31.60
 95% CI10.90-NR8.93-NR22.67-NR
Log-rank P across groups P = .76
Cholangiocarcinoma
Class IClass IIClass III
Subjects 46 36 13
 Events19155
 Censored27218
Median rwOS in months 12.4711.6710.63
 95% CI10.63-24.579.57-NR9.1-NR
Log-rank P across groupsP = .45
 Pancreatic cancer
Class IClass IIClass III
Subjects 33 4720
 Events9206
 Censored242714
Median rwOS in months NR 11.30 NR
 95% CI10.17-NR7.33-NR6.10-NR
Log-rank p across groupsP = 0.36
Gastro/esophageal cancer
Class IClass IIClass III
Subjects 18 4414
 Events4144
 Censored142910
Median rwOS in months 14.70 NR 31.60
 95% CI10.90-NR8.93-NR22.67-NR
Log-rank P across groups P = .76

Abbreviations: CI, confidence interval; rwOS: real-world overall survival.

There were no statistically significant differences in real-world overall survival (rwOS) for patients with BRAFV600E compared to those without for any cancer type (P = 0.17-0.97). When assessing alterations by cancer type, patients with BRAFV600E alterations had numerically improved rwOS for CCA and PDAC; the inverse was observed for GEA [CCA BRAFV600E median survival: 34.1 months (95% CI, 22.0-46-2) vs 24.8 months (95% CI, 23.9-25.9); PDAC BRAFV600E median survival 70.6 months (95% CI, 12.9-NR) vs 20.9 (95%CI, 20.4-21.7); gastroesophageal BRAFV600E medial survival: 27.0 (95%CI, 17.8-55.9) vs 29.5 (95% CI, 28.23-31.40). While patients with CCA and BRAF Class I (V600E) alterations were significantly younger than those with Class II or III mutations (P = .0006, Table 3), there were no other demographic differences within cancer groups across age, sex, comorbidity index, or smoking status.

Table 3.

Demographics by BRAF mutational class for patients assessed for real-world overall survival

Cholangiocarcinoma
BRAF class I
(V600E, n = 46)
BRAF class II
(n = 36)
BRAF class III
(n = 13)
P-value
Age in years0.0006
 Median58.567.569
<65 (n, %)33 (71.7)14 (38.9)3 (23.1)
 65-74 (n, %)7 (17.4)11 (30.6)7 (53.9)
 75+ (n, %)5 (10.9)11 (30.6)3 (23.1)
Sex0.85
 Male, n (%)20 (43.5)17 (47.2%)5 (38.5)
Comorbidity
 ECI, mean (SD)5.3 (2.5)4.9 (3.2)5.8 (3.4)0.38
 ECI, median333
Smoking
 Ever smoker, n (%)10 (21.7)8 (22.2)4 (30.8)0.78
Pancreatic cancer
BRAF class I
(V600E, n = 33)
BRAF class II
(n = 47)
BRAF class III
(n = 20)
P-value
Age0.72
 Median706766.5
<65 (n, %)11 (33.3)18 (38.3)8 (40.0)
 65-74 (n, %)11 (33.3)13 (27.8)10 (50.0)
 75+ (n, %)11 (33.3)16 (34.0)2 (10.0)
Sex0.08
 Male, n (%)15 (45.5)32 (68.1)14 (70.0)
Comorbidity
 ECI, mean (SD)5.7 (3.0)5.0 (2.8)4.9 (1.8)0.63
 ECI, median545
Smoking
 Ever smoker, n (%)6 (18.2)13 (27.7)4 (20.0)0.057
 Gastro/esophageal cancer
BRAF class I
(V600E, n = 18)
BRAF class II (n = 44)BRAF class III
(n = 14)
P-value
Age0.9
 Median66.664.264
<658 (44.4%)18 (40.9%)8 (57.1%)
 65-743 (16.7%)17 (38.6%)2 (14.3%)
 75+7 (38.9%)9 (20.5%)4 (28.6%)
Sex0.46
 Male, n (%)11 (61.1%)30 (68.2%)7 (50.0%)
Comorbidity
 ECI, mean (SD)5.4 (2.6)4.9 (3.2)5.8 (3.4)0.38
 ECI, median546
Smoking
 Ever smoker, n (%)6 (33.3%)17 (38.6%)6 (42.9%)0.86
Cholangiocarcinoma
BRAF class I
(V600E, n = 46)
BRAF class II
(n = 36)
BRAF class III
(n = 13)
P-value
Age in years0.0006
 Median58.567.569
<65 (n, %)33 (71.7)14 (38.9)3 (23.1)
 65-74 (n, %)7 (17.4)11 (30.6)7 (53.9)
 75+ (n, %)5 (10.9)11 (30.6)3 (23.1)
Sex0.85
 Male, n (%)20 (43.5)17 (47.2%)5 (38.5)
Comorbidity
 ECI, mean (SD)5.3 (2.5)4.9 (3.2)5.8 (3.4)0.38
 ECI, median333
Smoking
 Ever smoker, n (%)10 (21.7)8 (22.2)4 (30.8)0.78
Pancreatic cancer
BRAF class I
(V600E, n = 33)
BRAF class II
(n = 47)
BRAF class III
(n = 20)
P-value
Age0.72
 Median706766.5
<65 (n, %)11 (33.3)18 (38.3)8 (40.0)
 65-74 (n, %)11 (33.3)13 (27.8)10 (50.0)
 75+ (n, %)11 (33.3)16 (34.0)2 (10.0)
Sex0.08
 Male, n (%)15 (45.5)32 (68.1)14 (70.0)
Comorbidity
 ECI, mean (SD)5.7 (3.0)5.0 (2.8)4.9 (1.8)0.63
 ECI, median545
Smoking
 Ever smoker, n (%)6 (18.2)13 (27.7)4 (20.0)0.057
 Gastro/esophageal cancer
BRAF class I
(V600E, n = 18)
BRAF class II (n = 44)BRAF class III
(n = 14)
P-value
Age0.9
 Median66.664.264
<658 (44.4%)18 (40.9%)8 (57.1%)
 65-743 (16.7%)17 (38.6%)2 (14.3%)
 75+7 (38.9%)9 (20.5%)4 (28.6%)
Sex0.46
 Male, n (%)11 (61.1%)30 (68.2%)7 (50.0%)
Comorbidity
 ECI, mean (SD)5.4 (2.6)4.9 (3.2)5.8 (3.4)0.38
 ECI, median546
Smoking
 Ever smoker, n (%)6 (33.3%)17 (38.6%)6 (42.9%)0.86

Abbreviation: ECI, Elixhauser comorbidity index.

Table 3.

Demographics by BRAF mutational class for patients assessed for real-world overall survival

Cholangiocarcinoma
BRAF class I
(V600E, n = 46)
BRAF class II
(n = 36)
BRAF class III
(n = 13)
P-value
Age in years0.0006
 Median58.567.569
<65 (n, %)33 (71.7)14 (38.9)3 (23.1)
 65-74 (n, %)7 (17.4)11 (30.6)7 (53.9)
 75+ (n, %)5 (10.9)11 (30.6)3 (23.1)
Sex0.85
 Male, n (%)20 (43.5)17 (47.2%)5 (38.5)
Comorbidity
 ECI, mean (SD)5.3 (2.5)4.9 (3.2)5.8 (3.4)0.38
 ECI, median333
Smoking
 Ever smoker, n (%)10 (21.7)8 (22.2)4 (30.8)0.78
Pancreatic cancer
BRAF class I
(V600E, n = 33)
BRAF class II
(n = 47)
BRAF class III
(n = 20)
P-value
Age0.72
 Median706766.5
<65 (n, %)11 (33.3)18 (38.3)8 (40.0)
 65-74 (n, %)11 (33.3)13 (27.8)10 (50.0)
 75+ (n, %)11 (33.3)16 (34.0)2 (10.0)
Sex0.08
 Male, n (%)15 (45.5)32 (68.1)14 (70.0)
Comorbidity
 ECI, mean (SD)5.7 (3.0)5.0 (2.8)4.9 (1.8)0.63
 ECI, median545
Smoking
 Ever smoker, n (%)6 (18.2)13 (27.7)4 (20.0)0.057
 Gastro/esophageal cancer
BRAF class I
(V600E, n = 18)
BRAF class II (n = 44)BRAF class III
(n = 14)
P-value
Age0.9
 Median66.664.264
<658 (44.4%)18 (40.9%)8 (57.1%)
 65-743 (16.7%)17 (38.6%)2 (14.3%)
 75+7 (38.9%)9 (20.5%)4 (28.6%)
Sex0.46
 Male, n (%)11 (61.1%)30 (68.2%)7 (50.0%)
Comorbidity
 ECI, mean (SD)5.4 (2.6)4.9 (3.2)5.8 (3.4)0.38
 ECI, median546
Smoking
 Ever smoker, n (%)6 (33.3%)17 (38.6%)6 (42.9%)0.86
Cholangiocarcinoma
BRAF class I
(V600E, n = 46)
BRAF class II
(n = 36)
BRAF class III
(n = 13)
P-value
Age in years0.0006
 Median58.567.569
<65 (n, %)33 (71.7)14 (38.9)3 (23.1)
 65-74 (n, %)7 (17.4)11 (30.6)7 (53.9)
 75+ (n, %)5 (10.9)11 (30.6)3 (23.1)
Sex0.85
 Male, n (%)20 (43.5)17 (47.2%)5 (38.5)
Comorbidity
 ECI, mean (SD)5.3 (2.5)4.9 (3.2)5.8 (3.4)0.38
 ECI, median333
Smoking
 Ever smoker, n (%)10 (21.7)8 (22.2)4 (30.8)0.78
Pancreatic cancer
BRAF class I
(V600E, n = 33)
BRAF class II
(n = 47)
BRAF class III
(n = 20)
P-value
Age0.72
 Median706766.5
<65 (n, %)11 (33.3)18 (38.3)8 (40.0)
 65-74 (n, %)11 (33.3)13 (27.8)10 (50.0)
 75+ (n, %)11 (33.3)16 (34.0)2 (10.0)
Sex0.08
 Male, n (%)15 (45.5)32 (68.1)14 (70.0)
Comorbidity
 ECI, mean (SD)5.7 (3.0)5.0 (2.8)4.9 (1.8)0.63
 ECI, median545
Smoking
 Ever smoker, n (%)6 (18.2)13 (27.7)4 (20.0)0.057
 Gastro/esophageal cancer
BRAF class I
(V600E, n = 18)
BRAF class II (n = 44)BRAF class III
(n = 14)
P-value
Age0.9
 Median66.664.264
<658 (44.4%)18 (40.9%)8 (57.1%)
 65-743 (16.7%)17 (38.6%)2 (14.3%)
 75+7 (38.9%)9 (20.5%)4 (28.6%)
Sex0.46
 Male, n (%)11 (61.1%)30 (68.2%)7 (50.0%)
Comorbidity
 ECI, mean (SD)5.4 (2.6)4.9 (3.2)5.8 (3.4)0.38
 ECI, median546
Smoking
 Ever smoker, n (%)6 (33.3%)17 (38.6%)6 (42.9%)0.86

Abbreviation: ECI, Elixhauser comorbidity index.

Real-world overall survival (rwOS) differed by BRAF alteration class across tumor types (Table 2), however no statistically significant differences were noted. For patients with CCA, those with class III alterations had the numerically greatest median rwOS (24.33 months, 95%CI, 9.1-NR) compared to V600E/class I and class II alterations [12.47 months (95% CI, 10.63-24.57); 11.67 months (95%CI, 9.567-NR), respectively]. Notably, only 3/46 (6.5%) patients with V600E/class I alterations were treated with BRAF inhibitors; most patients had chemotherapy and/or immunotherapy-containing regimens (Table 4) with the highest rates observed in patients with BRAF class III alterations (78.6%) compared to those with V600E/class I and class II (52.0%, 52.8%, respectively; Table 4).

Table 4.

Treatments by cancer type and BRAF mutational class in the outcomes cohort (n, %)

Cholangiocarcinoma
TherapyBRAF class 1
(n = 46)
BRAF class 2
(n = 36)
BRAF class 3
(n = 14)
Total
(n = 95)
BRAF inhibitor3 (6.5)0 (0)0 (0)3 (3.2)
Chemo only24 (52.0)19 (52.8)11 (78.6)54 (56.8)
 Chemo + Bevacizumab1 (2.1)2 (5.6)0 (0)3 (3.2)
Chemo/IO6 (13.0)11 (3.1)1 (7.7)18 (18.9)
IO only9 (19.6)2 (5.6)1 (7.7)12 (12.6)
Targeted therapy3 (6.5)2 (5.6)0 (0)5 (5.3)
Pancreatic cancer
TherapyBRAF class 1
(n = 33)
BRAF class 2
(n = 47)
BRAF class 3
(n = 20)
Total
(n = 100)
BRAF inhibitor1 (3.0)0 (0)0 (0)1 (1.0)
Chemo only22 (66.7)41 (87.2)20 (100)83 (83.0)
IO only8 (24.2)4 (8.5)0 (0)12 (12.0)
Targeted therapy2 (6.1)2 (4.2)0 (0)4 (4,0)
Gastroesophageal cancer
TherapyBRAF class 1
(n = 18)
BRAF class 2
(n = 44)
BRAF class 3
(n = 14)
Total
(n = 76)
BRAF inhibitor2 (11.1)0 (0)0 (0)2 (2.6)
Chemo only8 (44.4)27 (61.4)9 (64)44 (57.9)
 Chemo + Bevacizumab1 (5.6)1 (2.3)0 (0)2 (2.6)
 Chemo + anti-HER20 (0)1 (2.3)1 (7.1)2 (2.6)
Chemo/IO4 (22.2)7 (15.9)2 (14.2)13 (17.1)
IO only2 (11.1)4 (9.0)1 (7.1)7 (9.2)
Targeted therapy1 (5.6)4 (9.0)1 (7.1)6 (7.9)
Cholangiocarcinoma
TherapyBRAF class 1
(n = 46)
BRAF class 2
(n = 36)
BRAF class 3
(n = 14)
Total
(n = 95)
BRAF inhibitor3 (6.5)0 (0)0 (0)3 (3.2)
Chemo only24 (52.0)19 (52.8)11 (78.6)54 (56.8)
 Chemo + Bevacizumab1 (2.1)2 (5.6)0 (0)3 (3.2)
Chemo/IO6 (13.0)11 (3.1)1 (7.7)18 (18.9)
IO only9 (19.6)2 (5.6)1 (7.7)12 (12.6)
Targeted therapy3 (6.5)2 (5.6)0 (0)5 (5.3)
Pancreatic cancer
TherapyBRAF class 1
(n = 33)
BRAF class 2
(n = 47)
BRAF class 3
(n = 20)
Total
(n = 100)
BRAF inhibitor1 (3.0)0 (0)0 (0)1 (1.0)
Chemo only22 (66.7)41 (87.2)20 (100)83 (83.0)
IO only8 (24.2)4 (8.5)0 (0)12 (12.0)
Targeted therapy2 (6.1)2 (4.2)0 (0)4 (4,0)
Gastroesophageal cancer
TherapyBRAF class 1
(n = 18)
BRAF class 2
(n = 44)
BRAF class 3
(n = 14)
Total
(n = 76)
BRAF inhibitor2 (11.1)0 (0)0 (0)2 (2.6)
Chemo only8 (44.4)27 (61.4)9 (64)44 (57.9)
 Chemo + Bevacizumab1 (5.6)1 (2.3)0 (0)2 (2.6)
 Chemo + anti-HER20 (0)1 (2.3)1 (7.1)2 (2.6)
Chemo/IO4 (22.2)7 (15.9)2 (14.2)13 (17.1)
IO only2 (11.1)4 (9.0)1 (7.1)7 (9.2)
Targeted therapy1 (5.6)4 (9.0)1 (7.1)6 (7.9)
Table 4.

Treatments by cancer type and BRAF mutational class in the outcomes cohort (n, %)

Cholangiocarcinoma
TherapyBRAF class 1
(n = 46)
BRAF class 2
(n = 36)
BRAF class 3
(n = 14)
Total
(n = 95)
BRAF inhibitor3 (6.5)0 (0)0 (0)3 (3.2)
Chemo only24 (52.0)19 (52.8)11 (78.6)54 (56.8)
 Chemo + Bevacizumab1 (2.1)2 (5.6)0 (0)3 (3.2)
Chemo/IO6 (13.0)11 (3.1)1 (7.7)18 (18.9)
IO only9 (19.6)2 (5.6)1 (7.7)12 (12.6)
Targeted therapy3 (6.5)2 (5.6)0 (0)5 (5.3)
Pancreatic cancer
TherapyBRAF class 1
(n = 33)
BRAF class 2
(n = 47)
BRAF class 3
(n = 20)
Total
(n = 100)
BRAF inhibitor1 (3.0)0 (0)0 (0)1 (1.0)
Chemo only22 (66.7)41 (87.2)20 (100)83 (83.0)
IO only8 (24.2)4 (8.5)0 (0)12 (12.0)
Targeted therapy2 (6.1)2 (4.2)0 (0)4 (4,0)
Gastroesophageal cancer
TherapyBRAF class 1
(n = 18)
BRAF class 2
(n = 44)
BRAF class 3
(n = 14)
Total
(n = 76)
BRAF inhibitor2 (11.1)0 (0)0 (0)2 (2.6)
Chemo only8 (44.4)27 (61.4)9 (64)44 (57.9)
 Chemo + Bevacizumab1 (5.6)1 (2.3)0 (0)2 (2.6)
 Chemo + anti-HER20 (0)1 (2.3)1 (7.1)2 (2.6)
Chemo/IO4 (22.2)7 (15.9)2 (14.2)13 (17.1)
IO only2 (11.1)4 (9.0)1 (7.1)7 (9.2)
Targeted therapy1 (5.6)4 (9.0)1 (7.1)6 (7.9)
Cholangiocarcinoma
TherapyBRAF class 1
(n = 46)
BRAF class 2
(n = 36)
BRAF class 3
(n = 14)
Total
(n = 95)
BRAF inhibitor3 (6.5)0 (0)0 (0)3 (3.2)
Chemo only24 (52.0)19 (52.8)11 (78.6)54 (56.8)
 Chemo + Bevacizumab1 (2.1)2 (5.6)0 (0)3 (3.2)
Chemo/IO6 (13.0)11 (3.1)1 (7.7)18 (18.9)
IO only9 (19.6)2 (5.6)1 (7.7)12 (12.6)
Targeted therapy3 (6.5)2 (5.6)0 (0)5 (5.3)
Pancreatic cancer
TherapyBRAF class 1
(n = 33)
BRAF class 2
(n = 47)
BRAF class 3
(n = 20)
Total
(n = 100)
BRAF inhibitor1 (3.0)0 (0)0 (0)1 (1.0)
Chemo only22 (66.7)41 (87.2)20 (100)83 (83.0)
IO only8 (24.2)4 (8.5)0 (0)12 (12.0)
Targeted therapy2 (6.1)2 (4.2)0 (0)4 (4,0)
Gastroesophageal cancer
TherapyBRAF class 1
(n = 18)
BRAF class 2
(n = 44)
BRAF class 3
(n = 14)
Total
(n = 76)
BRAF inhibitor2 (11.1)0 (0)0 (0)2 (2.6)
Chemo only8 (44.4)27 (61.4)9 (64)44 (57.9)
 Chemo + Bevacizumab1 (5.6)1 (2.3)0 (0)2 (2.6)
 Chemo + anti-HER20 (0)1 (2.3)1 (7.1)2 (2.6)
Chemo/IO4 (22.2)7 (15.9)2 (14.2)13 (17.1)
IO only2 (11.1)4 (9.0)1 (7.1)7 (9.2)
Targeted therapy1 (5.6)4 (9.0)1 (7.1)6 (7.9)

For PDAC, median rwOS was numerically poorest for patients with class II alterations (11.30 months, 95% CI, 7.33-NR); however, many patients were censored in other groups (class I, class III). Only one patient of 33 with BRAFV600E/class I alterations received a BRAF inhibitor with >80% of the cohort receiving chemotherapy, as expected. All patients with Class III had chemo-only regimens compared to V600E/class I and class II who had lower rates of chemo-only regimens (Table 4).

Finally, patients with GEA who harbored BRAFV600E/class I alterations had the numerically poorest median rwOS (14.70 months, 95% CI, 10.90-NR), while those with class III alterations had the numerically highest rwOS (31.60 months, 95% CI, 22.67-NR). Of 18 patients with V600E/class I alterations, 2 (11%) were treated with BRAF inhibitors, the highest rate among cancer types assessed. Patients with V600E/class I alterations had lower rates of chemotherapy-only regimens (44%) and higher rates of chemo/IO (22%) regimens compared to class II and III (chemo-only: 44% vs. 61% vs. 64%, respectively; chemo/IO: 22% vs. 15% vs. 14%, respectively; Table 4).

When assessing co-occurring alterations in patients with BRAFV600E (regardless of outcomes availability), certain pathways were observed to be impacted most frequently, including the RTK pathway, p53 cell cycle pathway, and the Wnt/β-catenin pathway (Supplementary Figure S1A-C). This includes frequently harbored alterations in TP53 (41%), FGFR2 (12%), PIK3CA (12%), and KRAS (10%) in BRAFV600E CCA (Supplementary Figure S1A); TP53 (47%), KRAS (21%), APC (13%), and SMAD4 (12%) in BRAFV600E PDAC (Supplementary Figure S1B); and TP53 (61%), APC (19%), ARID1A (19%), and EGFR (15%) in BRAFV600E GEA (Supplementary Figure S1C). Alterations across these pathways can impact processes such as cell proliferation, cell survival, and differentiation, all contributing to the carcinogenic process.25,26

Discussion

In the present study, we demonstrate the feasibility of cfDNA detection to reliably identify BRAF mutations in non-CRC GI malignancies, which may have implications for clinical management. In our study, BRAF alterations were identified in approximately 4% of these non-CRC GI malignancies. The frequency of BRAF alterations in this group is lower when compared to other malignancies, including CRC (10%)3,4 and melanoma (50%).27 To our knowledge, this is one of few studies reporting prognostic implications of BRAF mutations in non-CRC GI malignancies.

Among GI malignancies evaluated, classified BRAF alterations were most frequently observed in CCA, which also had the highest rate of V600E/class I alterations. While differences in rwOS among BRAF mutation classes were not statistically significant, a numerical trend towards superior rwOS was noted among those with class III mutations, which is consistent with previously observed positive prognostic value of class III mutations in colorectal cancer; notably low numbers of patients with this type of mutation in CCA does limit analysis to some extent.28 Outcomes of those with class I and class II mutations were similar, which is not unexpected considering observations of similar behavior between these 2 mutational classes in patients with CRC.8 Other studies evaluating the prognostic implications of BRAF alterations in CCA are limited, and this requires further evaluation.

BRAF mutations were found in 2.3% of patients with PDAC, which is consistent with prior reports noting an incidence of 2-4% in those with advanced disease.29 In the present study, those with class II mutations appeared to have the shortest rwOS, although conclusions are challenging due to the high rates of censorship in this cohort. Notably, of 33 patients with a BRAFV600E mutation, only one received BRAF-targeted therapy. Considering the poor prognosis with PDAC and the lack of therapeutic options, this does demonstrate an unmet need.

Lastly, BRAF mutations were seen in 2.6% of patients with GEA. Previous studies of gastric or esophageal cancers report a low frequency of 0-1%.30,31 A higher frequency of BRAF alterations may be observed here due to differences in clinical status compared to other studies (newly diagnosed vs. not responding to therapy) and/or heterogeneity in GEA which has been well documented. While not significant, there was a numerical trend toward longer rwOS in those with class II/III alterations, reflecting the prognostic pattern of CRC, although low patient count does limit this analysis. Baseline characteristics among those with various BRAF statuses were not explanatory of the observed differences. One prior study reported BRAF overexpression to be correlated with poorer survival in esophageal cancer, however, it is difficult to draw comparisons between this study and the present one.32

In addition to defining the prevalence and prognostic implications of BRAF mutations in non-CRC GI malignancies, this study demonstrates the ability of ctDNA to reliably characterize somatic BRAF mutations. Among patients with cfDNA detected, rates of BRAF mutation detection were comparable to those observed via tissue-based assays, suggesting the validity of this liquid biopsy assay.33,34 Rates in this study may have been slightly elevated given tissue-based assays are often deployed at the time of diagnosis; this cohort overall was assessed using liquid biopsy after diagnosis in 35% of cases, suggesting perhaps higher rates of BRAF due to heterogeneity and/or treatment pressure not captured at tissue biopsy at the time of diagnosis. However, the validity of using liquid biopsy for BRAF detection has been supported by additional studies.35

Notably, ~1% of the cohort had V600E/class I alterations yet only 6 patients were treated with BRAF inhibitors While it is unclear as to why BRAF inhibitors were infrequently used in this population, one explanation may be the underwhelming efficacy of BRAF inhibition in management of GI cancers when compared to melanoma. In trials assessing the efficacy of BRAF inhibition in combination with MEK inhibition in the treatment of melanoma, response rates of 63-69% were seen.36-39 However, in the case of CRC, the BEACON trial found that those who received a combination of encorafenib plus cetuximab (EGFR inhibitor) exhibited an objective response rate of 20%.3 In the case of biliary tract cancers, dabrafenib plus trametinib has resulted in an ORR of 51%, however, these were all partial responses in the ROAR trial.15 Prospective data for the use of BRAF inhibitors in other GI malignancies is limited. Additionally, BRAF inhibitors have only been approved in patients with GI malignancies at the time of disease progression, which may also reduce the number of patients receiving therapy. An additional consideration is the relatively recent approval of BRAF inhibitors in non-CRC GI malignancies which may impact rates of utilization.

Limitations of this study include its retrospective nature and lack of clinical data for all patients, particularly those with HCC. Additionally, the relative infrequency of various BRAF mutations in these malignancies does make it more challenging to draw definitive conclusions comparing rwOS, particularly given the high censorship rate in the cohorts presented here. In this study, we were not able to elucidate the utility of BRAF inhibitors in this patient population as too few in our study cohort were treated with BRAF inhibitors. Further, while we report prognostic implications of various BRAF mutations among non-CRC GI malignancies, implications of these mutations may change with increased utilization of BRAF inhibitors among patients with BRAFV600E mutations. Additionally, missingness of data may occur in real-world datasets due to lack of continuous enrollment in health plans and/or loss-to-follow-up; rwOS should be interpreted accordingly.

Conclusions

In this study, we demonstrate the feasibility of the detection of classified BRAF alterations among patients with non-CRC GI malignancies via liquid biopsy. To the best of our knowledge, this is the largest study to assess the prevalence and potential impact of BRAF alterations in patients with non-CRC GI cancers, and one of the only studies to publish prevalence for hepatocellular carcinoma. While these alterations are relatively rare, they may have prognostic implications, as well as implications in treatment considering recent FDA tumor-agnostic approval of dabrafenib and trametinib. This study further demonstrates the importance of biomarker testing and the utility of liquid biopsy in patients with advanced GI cancers and the ongoing need to develop and assess the impact of BRAF-targeted therapies on patient outcomes in these subpopulations.

Prior Presentation: Presented in part at the 2023 ASCO Gastrointestinal Cancers Symposium.

Author contributions

Amit Mahipal, Leslie Bucheit, Sakti Chakrabarti (Conceptualization). Leslie Bucheit, Nicole Zhang, Reagan M. Barnett (Data curation). Leslie Bucheit, Nicole Zhang (Formal analysis). None (Funding acquisition). Amit Mahipal, Leslie Bucheit, Nicole Zhang (Investigation Methodology). Amit Mahipal, Leslie Bucheit (Project administration). Leslie Bucheit (Resources). Nicole Zhang (Software). Amit Mahipal, Leslie Bucheit (Supervision). Leslie Bucheit, Nicole Zhang (Validation). None (Visualization). Amit Mahipal, Leslie Bucheit, Michael H. Storandt (Writing—original draft). All authors (Writing—review & editing)

Funding

None.

Conflicts of interest

A.M. is on advisory board on Taiho oncology, Merck and Exelixis. A.M. is on speakers bureau with Astrazeneca, Exelixis, Guardant Health, Astellas and Taiho Oncology. L.B., N.Z., and R.B. are employees and shareholders of Guardant Health Inc. MHS and SC reports no financial disclosures.

Data availability

The data underlying this article will be shared on reasonable request to the corresponding author.

References

1.

Degirmenci
U
,
Wang
M
,
Hu
J.
Targeting aberrant RAS/RAF/MEK/ERK signaling for cancer therapy
.
Cells
.
2020
;
9
:
198
. https://doi.org/

2.

Sinicrope
FA
,
Mahoney
MR
,
Yoon
HH
, et al. ;
Alliance for Clinical Trials in Oncology
.
Analysis of molecular markers by anatomic tumor site in stage iii colon carcinomas from adjuvant chemotherapy trial NCCTG N0147 (Alliance)
.
Clin Cancer Res
.
2015
;
21
:
5294
-
5304
. https://doi.org/

3.

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
. https://doi.org/

4.

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
. https://doi.org/

5.

Robert
C
,
Grob
JJ
,
Stroyakovskiy
D
, et al.
Five-year outcomes with Dabrafenib plus Trametinib in metastatic melanoma
.
N Engl J Med
.
2019
;
381
:
626
-
636
. https://doi.org/

6.

Atkins
MB
,
Lee
SJ
,
Chmielowski
B
, et al.
Combination dabrafenib and trametinib versus combination Nivolumab and Ipilimumab for patients with advanced BRAF-mutant melanoma: the DREAMseq trial-ECOG-ACRIN EA6134
.
J Clin Oncol
.
2023
;
41
:
186
-
197
. https://doi.org/

7.

Yan
N
,
Guo
S
,
Zhang
H
, et al.
BRAF-mutated non-small cell lung cancer: current treatment status and future perspective
.
Front Oncol
.
2022
;
12
:
863043
. https://doi.org/

8.

Schirripa
M
,
Biason
P
,
Lonardi
S
, et al.
Class 1, 2, and 3 BRAF-mutated metastatic colorectal cancer: a detailed clinical, pathologic, and molecular characterization
.
Clin Cancer Res
.
2019
;
25
:
3954
-
3961
. https://doi.org/

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
. https://doi.org/

10.

Cremolini
C
,
Di Bartolomeo
M
,
Amatu
A
, et al.
BRAF codons 594 and 596 mutations identify a new molecular subtype of metastatic colorectal cancer at favorable prognosis
.
Ann Oncol
.
2015
;
26
:
2092
-
2097
. https://doi.org/

11.

Dagogo-Jack
I
,
Martinez
P
,
Yeap
BY
, et al.
Impact of BRAF mutation class on disease characteristics and clinical outcomes in BRAF-mutant lung cancer
.
Clin Cancer Res
.
2019
;
25
:
158
-
165
. https://doi.org/

12.

Safaee Ardekani
G
,
Jafarnejad
SM
,
Tan
L
,
Saeedi
A
,
Li
G.
The prognostic value of BRAF mutation in colorectal cancer and melanoma: a systematic review and meta-analysis
.
PLoS One
.
2012
;
7
:
e47054
. https://doi.org/

13.

Clancy
C
,
Burke
JP
,
Kalady
MF
,
Coffey
JC.
BRAF mutation is associated with distinct clinicopathological characteristics in colorectal cancer: a systematic review and meta-analysis
.
Colorectal Dis
.
2013
;
15
:
e711
-
e718
. https://doi.org/

14.

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
. https://doi.org/

15.

Subbiah
V
,
Lassen
U
,
Élez
E
, et al.
Dabrafenib plus trametinib in patients with BRAFV600E-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial
.
Lancet Oncol
.
2020
;
21
:
1234
-
1243
. https://doi.org/

16.

Subbiah
V
,
Kreitman
RJ
,
Wainberg
ZA
, et al.
Dabrafenib plus trametinib in patients with BRAF V600E-mutant anaplastic thyroid cancer: updated analysis from the phase II ROAR basket study
.
Ann Oncol.
2022
;
33
:
406
-
415
. https://doi.org/

17.

Wen
PY
,
Stein
A
,
van den Bent
M
, et al.
Dabrafenib plus trametinib in patients with BRAFV600E-mutant low-grade and high-grade glioma (ROAR): a multicentre, open-label, single-arm, phase 2, basket trial
.
Lancet Oncol
.
2022
;
23
:
53
-
64
. https://doi.org/

18.

Kopetz
S
,
Grothey
A
,
Van Cutsem
E
, et al.
Quality of life with encorafenib plus cetuximab with or without binimetinib treatment in patients with BRAF V600E-mutant metastatic colorectal cancer: patient-reported outcomes from BEACON CRC
.
ESMO Open.
2022
;
7
:
100477
. https://doi.org/

19.

Strickler
JH
,
Loree
JM
,
Ahronian
LG
, et al.
Genomic landscape of cell-free DNA in patients with colorectal cancer
.
Cancer Discov.
2018
;
8
:
164
-
173
. https://doi.org/

20.

Chung
C
,
Galvin
R
,
Achenbach
E
,
Dziadkowiec
O
,
Sen
S.
Characterization of blood-based molecular profiling in pancreatic adenocarcinoma
.
Oncology (Williston Park).
2021
;
35
:
794
-
803
. https://doi.org/

21.

Kato
S
,
Okamura
R
,
Baumgartner
JM
, et al.
Analysis of circulating tumor DNA and clinical correlates in patients with esophageal, gastroesophageal junction, and gastric adenocarcinoma
.
Clin Cancer Res
.
2018
;
24
:
6248
-
6256
. https://doi.org/

22.

Botrus
G
,
Uson Junior
PLS
,
Raman
P
, et al.
Circulating cell-free tumor DNA in advanced pancreatic adenocarcinoma identifies patients with worse overall survival
.
Front Oncol
.
2022
;
11
:
794009
. https://doi.org/

23.

Odegaard
JI
,
Vincent
JJ
,
Mortimer
S
, et al.
Validation of a plasma-based comprehensive cancer genotyping assay utilizing orthogonal tissue- and plasma-based methodologies
.
Clin Cancer Res
.
2018
;
24
:
3539
-
3549
. https://doi.org/

24.

Lanman
RB
,
Mortimer
SA
,
Zill
OA
, et al.
Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA
.
PLoS One
.
2015
;
10
:
e0140712
. https://doi.org/

25.

Zhang
N
,
Li
Y.
Receptor tyrosine kinases: biological functions and anticancer targeted therapy
.
MedComm (Beijing.
2023
;
4
:
446
. https://doi.org/

26.

Sasaki
K
,
Takahashi
S
,
Ouchi
K
, et al.
Different impacts of TP53 mutations on cell cycle-related gene expression among cancer types
.
Sci Rep
.
2023
;
13
:
4868
. https://doi.org/

27.

Davies
H
,
Bignell
GR
,
Cox
C
, et al.
Mutations of the BRAF gene in human cancer
.
Nature
.
2002
;
417
:
949
-
954
. https://doi.org/

28.

Chen
Y
,
Sun
H
,
Deng
Y
, et al.
The clinical and genomic distinctions of Class1/2/3 BRAF-mutant colorectal cancer and differential prognoses
.
Biomark Res
2023
;
11
:
11
. https://doi.org/

29.

Ciner
AT
,
Jiang
Y
,
Hausner
P.
BRAF-driven pancreatic cancer: prevalence, molecular features, and therapeutic opportunities
.
Mol Cancer Res
.
2023
;
21
:
293
-
300
. https://doi.org/

30.

Takahashi
N
,
Yamada
Y
,
Taniguchi
H
, et al.
Clinicopathological features and prognostic roles of KRAS, BRAF, PIK3CA and NRAS mutations in advanced gastric cancer
.
BMC Res Notes.
2014
;
7
:
271
. https://doi.org/

31.

Maeng
CH
,
Lee
J
,
van Hummelen
P
, et al.
High-throughput genotyping in metastatic esophageal squamous cell carcinoma identifies phosphoinositide-3-kinase and BRAF mutations
.
PLoS One
.
2012
;
7
:
e41655
. https://doi.org/

32.

Kuerbanjiang
A
,
Maimaituerxun
M
,
Zhang
Y
, et al.
V-Raf murine sarcoma viral oncogene homolog B1 (BRAF) as a prognostic biomarker of poor outcomes in esophageal cancer patients
.
BMC Gastroenterol
.
2021
;
21
:
86
. https://doi.org/

33.

Campbell
PJ
,
Getz
G
,
Korbel
JO
, et al.
Pan-cancer analysis of whole genomes
.
Nature
.
2020
;
578
:
82
-
93
.

34.

Zehir
A
,
Benayed
R
,
Shah
RH
, et al.
Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients
.
Nat Med
.
2017
;
23
:
703
-
713
. https://doi.org/

35.

Sobczuk
P
,
Kozak
K
,
Kopeć
S
, et al.
The use of ctDNA for BRAF mutation testing in routine clinical practice in patients with advanced melanoma
.
Cancers (Basel).
2022
;
14
:
777
. https://doi.org/

36.

Long
GV
,
Stroyakovskiy
D
,
Gogas
H
, et al.
Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial
.
Lancet
.
2015
;
386
:
444
-
451
. https://doi.org/

37.

Robert
C
,
Karaszewska
B
,
Schachter
J
, et al.
Improved overall survival in melanoma with combined dabrafenib and trametinib
.
N Engl J Med
.
2015
;
372
:
30
-
39
. https://doi.org/

38.

Larkin
J
,
Ascierto
PA
,
Dréno
B
, et al.
Combined vemurafenib and cobimetinib in BRAF-mutated melanoma
.
N Engl J Med
.
2014
;
371
:
1867
-
1876
. https://doi.org/

39.

Dummer
R
,
Ascierto
PA
,
Gogas
HJ
, et al.
Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): a multicentre, open-label, randomised phase 3 trial
.
Lancet Oncol
.
2018
;
19
:
603
-
615
. https://doi.org/

This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any medium, provided the original work is properly cited.