Purpose: Previous investigations identified transcriptional signatures associated with innate resistance to anti–programmed cell death protein 1 therapy in melanoma. This analysis aimed to increase understanding of the role of baseline genetic features in the variability of response to BRAF and MEK inhibitor therapy for BRAFV600-mutated metastatic melanoma.

Patients and Methods: This exploratory analysis compared genomic features, using whole-exome and RNA sequencing, of baseline tumors from patients who had complete response versus rapid progression (disease progression at first postbaseline assessment) on treatment with cobimetinib combined with vemurafenib or vemurafenib alone. Associations of gene expression with progression-free survival or overall survival were assessed by Cox proportional hazards modeling.

Results: Whole-exome sequencing showed that MITF and TP53 alterations were more frequent in tumors from patients with rapid progression, while NF1 alterations were more frequent in tumors from patients with complete response. However, the low frequency of alterations in any one gene precluded their characterization as drivers of response/resistance. Analysis of RNA profiles showed that expression of immune response–related genes was enriched in tumors from patients with complete response, while expression of keratinization-related genes was enriched in tumors from patients who experienced rapid progression.

Conclusions: These findings suggest that enriched immune infiltration might be a shared feature favoring response to both targeted and immune therapies, while features of innate resistance to targeted and immune therapies were distinct.

Translational Relevance

Our exploratory analysis of baseline BRAFV600-mutated melanoma samples from the BRAF inhibitor in melanoma (BRIM) studies aimed to identify genetic characteristics of patients with a complete response or no response to combined BRAF and MEK inhibition with vemurafenib and cobimetinib. No individual mutation was identified as a population-wide driver of exceptional response, but certain gene expression signatures were found to distinguish the 2 extremes of response. Melanomas from patients with complete response possessed higher preexisting tumor immunity features, while those from patients with rapid progression had a keratin signature similar to one previously associated with poor prognosis. These findings suggest that enriched immune infiltration of melanomas may be a shared feature of favorable response to both targeted and immune therapies, while features of innate resistance to immune and targeted therapies were distinct. We anticipate these findings could assist in optimization of treatment selection for patients with BRAFV600-mutated metastatic melanoma.

The introduction of small-molecule inhibitors of BRAF and MEK and mAbs targeting programmed cell death protein 1 (PD-1) and CTL–associated antigen 4 have remarkably improved treatment outcomes for patients with metastatic melanoma (1–15). With the emergence of distinct classes of therapies, there is a need to better understand baseline features associated with response to either targeted therapies or immunotherapies to allow consideration of treatment that will optimize benefits for the individual patient. An investigation of genomic features in responders compared with nonresponders identified transcriptional signatures associated with innate resistance to anti–PD-1 therapy (16, 17), although no common feature was identified to predict the response. While previous investigation of the mechanisms of acquired resistance to inhibitors of the MAPK pathway identified MAPK reactivation by various genetic and epigenetic alterations developed during treatment (18, 19), baseline features predicting response to MAPK-targeted therapies have not been examined.

Combined MEK and BRAF inhibition with cobimetinib and vemurafenib has been shown to improve response rates, progression-free survival (PFS; refs. 4, 5), and overall survival (OS; ref. 6) compared with BRAF inhibitor monotherapy. Responses to BRAF and MEK inhibitors vary between patients; while some patients achieve complete response (CR), a proportion of patients demonstrate rapid disease progression suggestive of some degree of innate resistance. A better understanding of the mechanisms underlying the variability of patient responses to these BRAF and MEK inhibitors is required to target therapy more effectively.

The objective of this retrospective, exploratory analysis was to compare genomic features of pretreatment tumors from complete responders with those from rapid progressors on treatment with cobimetinib combined with vemurafenib or vemurafenib alone in patients with BRAFV600-mutated metastatic melanoma.

Analysis population

Data from patients with BRAFV600-mutated metastatic melanoma treated with cobimetinib combined with vemurafenib or vemurafenib in the BRIM-2, BRIM-3, BRIM-7, and coBRIM studies were included in the analysis. Detailed methods have previously been reported for each study. BRIM-2 (ClinicalTrials.gov ID, NCT00949702) was a multicenter, single-arm phase II study in which patients were treated with vemurafenib 960 mg twice daily (1). BRIM-3 (ClinicalTrials.gov ID, NCT01006980) was a multicenter, randomized, open-label phase III study in which patients were randomly assigned to receive vemurafenib 960 mg twice daily or dacarbazine 1,000 mg/m2 every 3 weeks (2, 3). BRIM-7 (ClinicalTrials.gov ID, NCT01271803) was a multicenter, single-arm phase Ib dose-escalation study in which patients received cobimetinib 60, 80, or 100 mg once daily, given on a schedule of 14 days on/14 days off, 21 days on/7 days off, or continuously, in combination with vemurafenib 720 or 960 mg twice daily (4). coBRIM (ClinicalTrials.gov ID, NCT01689519) was a multicenter, randomized, double-blind phase III study in which patients were randomly assigned to receive cobimetinib 60 mg once daily for 21 days followed by 7 days off or placebo in combination with vemurafenib 960 mg twice daily (5, 6).

Key eligibility criteria for the 4 trials were similar, including age ≥18 years, unresectable stage IIIC or IV melanoma harboring a BRAFV600 mutation, an Eastern Cooperative Oncology Group performance status of 0–1, and adequate organ function. The BRIM-3 and coBRIM studies enrolled previously untreated patients only, whereas BRIM-2 enrolled patients who had received at least 1 prior systemic treatment for advanced melanoma and BRIM-7 enrolled both previously treated and untreated patients.

Baseline tumor samples were obtained from consenting patients before initiation of study treatment. Genomic features at baseline were compared between patients who had CR or rapid progression [defined as disease progression (PD) at the first tumor assessment] according to Response Evaluation Criteria in Solid Tumors, version 1.1, on treatment with cobimetinib combined with vemurafenib or vemurafenib alone.

Each of the trials was conducted in accordance with the Declaration of Helsinki and the principles of Good Clinical and Laboratory Practice and with the approval of appropriate ethics committees. All participants in each study provided written informed consent.

Whole-exome sequencing

Baseline formalin-fixed, paraffin-embedded (FFPE) tumor samples from patients with complete response and patients with rapid progression were analyzed by whole-exome sequencing (WES). Patient samples where BRAF mutations were not detected were excluded from the analysis.

DNA exomes were captured using the Agilent SureSelect 51 Mb Kit (SQ756; Agilent Technologies, Inc.) and sequenced to a target depth of 100 ×. Reads were aligned to human reference genome GRCh38 using genomic short-read nucleotide alignment program (20). Variants were called using LoFreq (21). For variant filtering, sequencing data pooled from normal tissue samples from 106 patients in the BRIM-2, BRIM-3, BRIM-7, and coBRIM studies and data from the Exome Aggregation Consortium (ExAC) database (22) were used as reference data in the variant calling algorithm. Copy-number alterations were identified using CODEX (23). Variants and copy-number alterations were annotated using the Ensembl Variant Effect Predictor tool (24).

RNA sequencing

A subset of baseline tumor samples from patients with CR and patients with PD were evaluated by RNA sequencing (RNA-Seq) to identify transcriptional signatures. mRNA libraries were prepared using TruSeq RNA Access (Illumina). Paired-end 2 × 100 base reads were generated on a HiSeq system (Illumina). Reads were aligned to human reference genome GRCh38 using a genomic short-read nucleotide alignment program, and reads that overlapped gene exonic regions were counted (20). Differentially expressed pathways were identified by gene set enrichment analysis using the Molecular Signatures Database v5.1 (25, 26). For transcriptional signature analyses, counts were normalized to library size as counts per million using the voom function of the R limma package (27, 28) and computed by taking the mean z-score of all component genes. Using RNA-Seq data, in silico cell-type enrichment analysis was performed using xCell to calculate the percentage of tumor cells, as well as stromal and infiltrating immune cells (29).

Statistical analysis

Frequencies of mutations and copy-number alterations identified by WES were compared between biopsies from patients with CR and patients with PD by Fisher exact test (2-sided). Differential expression analysis was performed using the R limma package (27). Differential gene expression was defined as a raw P value ≤ 0.05 and at least a 2-fold change (increase or decrease) in expression between biopsies from patients with CR and patients with PD. Transcriptional signatures and mutational load were compared by ANOVA (2-sided ANOVA). Associations of gene expression with PFS or OS were assessed by Cox proportional hazards modeling.

Differences in baseline genetic alterations

A total of 130 baseline tumor samples underwent WES, with a median of 46.8 million reads (interquartile range, 40.7–50.7 million). Exome sequencing data were excluded because a BRAF mutation could not be identified (n = 17) or because of a high fraction of unmapped reads (n = 4). Thus, baseline tumors from 48 patients with CR and 61 patients with PD were characterized by WES. A list of tumors, clinical features, treatment, response, and sequencing characteristics is provided in Supplementary Table S1. Raw sequencing data are provided in Supplementary Table S2.

Large heterogeneity in mutation profiles was observed across the 2 subgroups (Fig. 1). The specific genes altered in tumor biopsies from patients with a CR to therapy were different from those that were altered in tumor biopsies from patients with PD; however, the frequency of alterations in any 1 gene was too low to warrant characterization as a common driver in these responses.

Figure 1.

Alterations observed in ≥5% of tumor samples from patients with CR (n = 37) or PD (n = 70). Whole-exome sequencing results for the most frequently mutated genes are shown. CR, biopsies from patients with complete response; nsSNVs, nonsynonymous single nucleotide variants; PD, biopsies from patients with rapid progression. aUnless denoted as Del (deletion) or Amp (amplification), alterations include single nucleotide variations, insertions, and deletions.

Figure 1.

Alterations observed in ≥5% of tumor samples from patients with CR (n = 37) or PD (n = 70). Whole-exome sequencing results for the most frequently mutated genes are shown. CR, biopsies from patients with complete response; nsSNVs, nonsynonymous single nucleotide variants; PD, biopsies from patients with rapid progression. aUnless denoted as Del (deletion) or Amp (amplification), alterations include single nucleotide variations, insertions, and deletions.

Close modal

A preliminary analysis of genetic features distinguishing between biopsies from patients with CR and patients with PD showed higher rates of MITF amplification (15% vs. 4%, P = 0.11) and TP53 mutations (15% vs. 4%, P = 0.11) in biopsies from patients with PD than in those from patients with CR. However, although there was a trend of higher MITF expression (log2 fold change 0.65, P = 0.06) and lower AXL expression (log2 fold change 0.45, P = 0.16) from patients with PD compared to patients with CR, these differences were not statistically significant (Supplementary Fig. S1). Conversely, NF1 alterations were more common in biopsies from patients with CR than in those from patients with PD (13% vs. 3%, P = 0.13). No significant difference was observed between biopsies from patients with CR and patients with PD in the frequency of BRAF amplifications (10% vs. 10%, P = 1.0) or CDKN2A alterations (44% vs. 52%, P = 0.44). Tumor mutational load, as assessed by exome-wide nonsynonymous single nucleotide variants (nsSNV), was similar between patients with CR and those with rapid progression (Fig. 2).

Figure 2.

Mutational load in patients with complete response (n = 37) or rapid progression (n = 70). CR, biopsies from patients with complete response; nsSNVs, nonsynonymous single nucleotide variants; PD, biopsies from patients with rapid progression.

Figure 2.

Mutational load in patients with complete response (n = 37) or rapid progression (n = 70). CR, biopsies from patients with complete response; nsSNVs, nonsynonymous single nucleotide variants; PD, biopsies from patients with rapid progression.

Close modal

Association of response with signatures of preexisting tumor immunity

Because genomic differences could not fully account for CR versus PD, we investigated transcriptomic variation in these pretreatment tumor biopsies. Tumors from 32 patients with CR and 40 patients with PD were evaluated by RNA-Seq.

The initial analysis identified 669 genes that were differentially expressed between tumors from patients with CR and patients with PD (Fig. 3A; Supplementary Table S3). Among these differentially expressed genes, 370 were also associated with PFS (unadjusted P < 0.05; Supplementary Table S4), 59 were also associated with OS (Supplementary Table S5), and 44 were associated with both PFS and OS (Fig. 3B). Gene ontology analysis indicated that enriched expression of genes related to immune response processes was associated with CR, while enriched expression of genes related to keratinization was associated with PD (Fig. 3C; refs. 30, 31). The immune-related expression profile included gene signatures of CD8+ effector T cells, cytolytic T cells, antigen-presenting cells, and natural killer cells (Supplementary Table S6), all of which were significantly enriched in tumors from patients with CR (Fig. 3D). Differential gene expression between patients with CR and PD was similar when examined according to treatment with vemurafenib alone or cobimetinib combined with vemurafenib (Supplementary Fig. S2).

Figure 3.

Differential gene expression by RNA-Seq in patients with complete response (n = 32) or rapid progression (n = 40; A). B, Number of differentially expressed genes associated with response (CR/PD), progression-free survival (PFS), and/or overall survival (OS). Ontology analysis of differentially expressed genes (C) and immune-related gene signatures (D). Gene ontology analysis from http://geneontology.org/ (30, 31). CR, biopsies from patients with complete response; FDR, false discovery rate (Benjamini–Hochberg method); NS, not significant; PD, biopsies from patients with rapid progression.

Figure 3.

Differential gene expression by RNA-Seq in patients with complete response (n = 32) or rapid progression (n = 40; A). B, Number of differentially expressed genes associated with response (CR/PD), progression-free survival (PFS), and/or overall survival (OS). Ontology analysis of differentially expressed genes (C) and immune-related gene signatures (D). Gene ontology analysis from http://geneontology.org/ (30, 31). CR, biopsies from patients with complete response; FDR, false discovery rate (Benjamini–Hochberg method); NS, not significant; PD, biopsies from patients with rapid progression.

Close modal

Consistent with the observation that immune-related genes are associated with CR, xCell analyses revealed higher levels of immune cell types such as CD8+ T cells in tumors from patients with CR compared with those with PD (Supplementary Table S7). IHC data from a subset of patients (n = 37) also showed a trend toward higher levels of CD8+ T cells in the tumor center in patients with CR than in those with PD (P = 0.051; Supplementary Fig. S3). There was good overall concordance between CD8+ T cells in the tumor center by IHC and CD8+ T-cell levels inferred from RNA-Seq using xCell (Pearson's r = 0.88; P = 8.1 × 10−6; Supplementary Fig. S4).

Association of resistance with “keratin” subtype

There was no significant difference in innate anti–PD-1-resistant signatures (IPRES; ref. 16) between tumors from patients with CR and patients with PD (P = 0.83; Fig. 4A). However, keratin and kallikrein gene expression was higher in tumors from patients with PD than in those with CR (Fig. 4B; Supplementary Table S3). These results suggest that features for innate resistance for targeted and immune therapies were distinct.

Figure 4.

Expression of resistance gene signatures by RNA sequencing in tumors from patients with complete response (n = 32) or rapid progression (n = 40). Gene signatures constitute innate anti–PD-1–resistant signatures (A) and keratin and kallikrein genes (B). CR, biopsies from patients with complete response; PD, biopsies from patients with rapid progression.

Figure 4.

Expression of resistance gene signatures by RNA sequencing in tumors from patients with complete response (n = 32) or rapid progression (n = 40). Gene signatures constitute innate anti–PD-1–resistant signatures (A) and keratin and kallikrein genes (B). CR, biopsies from patients with complete response; PD, biopsies from patients with rapid progression.

Close modal

Previous studies of melanoma disease progression have implicated tumor genomic heterogeneity in the development of resistance, mainly involving MAPK pathway reactivation (3). Our exploratory analysis of pretreatment melanoma biopsies showed a wide heterogeneity of genomic alterations in the tumor biopsies of patients who had CR and those who progressed rapidly when treated with cobimetinib combined with vemurafenib or vemurafenib alone.

The observation of greater MITF amplification in tumors from patients who experienced rapid progression is consistent with previous evidence that high MITF expression is associated with a proliferative (rapidly progressive) phenotype in melanoma (32, 33) and can contribute to resistance to MAPK pathway inhibition (34). Supporting this hypothesis, some difference was observed in baseline expression levels of MITF between tumors from patients with CR and patients with PD. TP53 alterations were more common in tumors from patients with PD than in those with CR in the current analysis; however, previous findings suggested that TP53 mutational status has no impact on overall response rate, PFS, and OS among patients with BRAFV600-mutated metastatic melanoma treated with a first-line BRAF inhibitor (35). This difference could be owing to the overall small patient number in this study. NF1 alterations were more common in tumors from patients with CR, consistent with evidence suggesting that melanomas lacking NF1 expression are dependent on MAPK signaling and more sensitive to MAPK pathway inhibitors (17, 36, 37). Although CDKN2A mutations have been associated with worse PFS and OS outcomes in patients treated with the MEK inhibitor trametinib combined with the BRAFV600 inhibitor dabrafenib (38), we observed no significant difference in CDKN2A alterations between response groups in this analysis. However, the low frequency of occurrence of any one mutation within this study suggests that, while individual mutations may drive progression in individual patients, there are no mutations that are population-wide drivers of response. Mutational load was similar between patients with CR or rapid progression, although there was a trend toward improved survival observed in patients treated with cobimetinib combined with vemurafenib who had higher mutational load in the coBRIM trial. This is in line with previous findings that high mutational load is not associated with tumor response to treatment in melanoma, but does correlate with improved patient survival (16, 38), implying that factors beyond mutational load influence shorter-term tumor responses and longer-term patient survival.

Melanomas from patients with CR possessed higher preexisting tumor immunity features than those from patients who experienced rapid progression. Multiple lines of evidence point to the involvement of the BRAF/MEK pathway in the regulation of the host antitumor response in melanoma (39–43). Oncogenic BRAF signaling contributes to immune escape in melanoma (39, 40), while BRAF inhibition has been shown to improve the host immune response to melanoma (41–43), and this immune response appears to be downregulated prior to the emergence of resistance (43). Taken together, the evidence suggests that presence of a preexisting immune response may be an important component of the clinical activity of BRAF/MEK inhibition. Given that pretreatment melanomas from patients with CR have greater tumor immunity features, the addition of anti–PD-1 therapy to cobimetinib combined with vemurafenib is currently under investigation.

The innate PD-1 resistance (IPRES) transcriptional signature describes pretreatment genomic features associated with response to anti–PD-1 therapy in metastatic melanoma; tumors nonresponsive to anti–PD-1 therapy were enriched for upregulation of expression of genes associated with mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing (16). In the current analysis, expression of groups of genes identified as IPRES was not significantly different between tumors from patients with CR and from those with rapid progression. This suggests that there is not a complete overlap in tumor resistance to anti–PD-1 therapy and tumor response to BRAF/MEK inhibition.

Melanomas from patients with rapid progression may be overrepresented by the “keratin” molecular subtype. Melanomas expressing high levels of keratins and genes associated with epithelium have been associated with worse outcomes (17). The higher expression of keratin and kallikrein genes in tumors from patients who experienced rapid progression in this analysis is reminiscent of this “keratin” signature associated with poor prognosis.

In summary, we identified specific transcriptomic signatures distinguishing patients who have complete responses and patients who progress rapidly on treatment with the BRAF/MEK inhibitors cobimetinib and/or vemurafenib for BRAFV600-mutated metastatic melanoma. Melanomas from patients with CR possessed higher preexisting tumor immunity features, while those from patients with PD may have the “keratin” signature associated with poor prognosis. These findings suggest that enriched immune infiltration might be a shared feature favoring response to both targeted and immune therapies, while features of innate resistance for targeted and immune therapies were distinct. These results provide a rationale for the combination of BRAF and MEK inhibition with immune checkpoint inhibitors in clinical studies, and may assist in optimization of treatment selection for patients with BRAFV600-mutated metastatic melanoma.

C. Robert is a consultant/advisory board member for Roche, Bristol-Myers Squibb, Pierre Fabre, MSD, and Novartis. J. Larkin reports receiving speakers bureau honoraria from and is a consultant/advisory board member for Achilles, AstraZeneca, Boston Biomedical, Bristol-Myers Squibb, Eisai, EUSA Pharma, GlaxoSmithKline, Ipsen, Imugene, Incyte, iOnctura, Kymab, Merck Serono, MSD, Nektar, Novartis, Pierre Fabre, Pfizer, Roche, Secarna, and Vitaccess. P.A. Ascierto reports receiving commercial research grants from Bristol-Myers Squibb, Roche-Genentech, and Array, and is a consultant/advisory board member for Bristol-Myers Squibb, Roche-Genentech, MSD, Amgen, Novartis, Array, Merck Serono, Incyte, Pierre Fabre, Genmab, Newlink Genetics, Medimmune, AstraZeneca, Syndax, Sun Pharma, Sanofi, Idera, and Ultimovacs. B. Dreno reports receiving speakers bureau honoraria from and is a consultant/advisory board member for Roche, Bristol-Myers Squibb, and Fabre. M. Maio is a consultant/advisory board member for Roche and Pierre Fabre. C. Garbe reports receiving commercial research grants from Bristol-Myers Squibb, Novartis, Neracare, and Roche, and is a consultant/advisory board member for Bristol-Myers Squibb, MSD, Novartis, Neracare, Philogen, Roche, Sanofi, and Amgen. P.B. Chapman reports receiving commercial research grants from Pfizer; holds ownership interest (including patents) in Rgenix; and is a consultant/advisory board member for Merck, Immunocore, Cell Medica, and AstraZeneca. J.A. Sosman is a consultant/advisory board member for Genentech. H. Koeppen holds ownership interest (including patents) in Roche. J.J. Hsu holds ownership interest (including patents) in Roche. I. Chang holds ownership interest (including patents) in Roche. A. Ribas is a consultant/advisory board member for Genentech, Roche, and Novartis. No potential conflicts of interest were disclosed by the other authors.

Conception and design: Y. Yan, M.J. Wongchenko, C. Robert, J. Larkin, C. Garbe, I. Chang, G.A. McArthur, A. Ribas

Development of methodology: Y. Yan, M.J. Wongchenko, C. Robert, J.A. Sosman, I. Caro

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Yan, M.J. Wongchenko, C. Robert, J. Larkin, P.A. Ascierto, B. Dreno, M. Maio, C. Garbe, P.B. Chapman, I. Caro, I. Rooney, G.A. McArthur, A. Ribas

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Yan, M.J. Wongchenko, J. Larkin, P.A. Ascierto, M. Maio, C. Garbe, Z. Shi, H. Koeppen, J.J. Hsu, I. Caro, G.A. McArthur, A. Ribas

Writing, review, and/or revision of the manuscript: Y. Yan, M.J. Wongchenko, C. Robert, J. Larkin, P.A. Ascierto, B. Dreno, M. Maio, C. Garbe, P.B. Chapman, J.A. Sosman, Z. Shi, H. Koeppen, J.J. Hsu, I. Chang, I. Caro, I. Rooney, G.A. McArthur, A. Ribas

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): M.J. Wongchenko, I. Chang

Study supervision: Y. Yan, M.J. Wongchenko, B. Dreno, I. Caro, A. Ribas

Medical writing support was provided by Melanie Sweetlove, MSc (ApotheCom, San Francisco, CA) and was funded by F. Hoffmann-La Roche Ltd.

The publication costs of this article were defrayed in part by the payment of publication fees. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/).

1.
Sosman
JA
,
Kim
KB
,
Schuchter
L
,
Gonzalez
R
,
Pavlick
AC
,
Weber
JS
, et al
.
Survival in BRAFV600-mutant advanced melanoma treated with vemurafenib
.
N Engl J Med
2012
;
366
:
707
14
.
2.
Chapman
PB
,
Hauschild
A
,
Robert
C
,
Haanen
JB
,
Ascierto
P
,
Larkin
J
, et al
.
Improved survival with vemurafenib in melanoma with BRAF V600E mutation
.
N Engl J Med
2011
;
364
:
2507
16
.
3.
McArthur
GA
,
Chapman
PB
,
Robert
C
,
Larkin
J
,
Haanen
JB
,
Dummer
R
, et al
.
Safety and efficacy of vemurafenib in BRAFv600E and BRAFv600K mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study
.
Lancet Oncol
2014
;
15
:
323
32
.
4.
Ribas
A
,
Gonzalez
R
,
Pavlick
A
,
Hamid
O
,
Gajewski
TF
,
Daud
A
, et al
.
Combination of vemurafenib and cobimetinib in patients with advanced BRAF(V600)-mutated melanoma: a phase 1b study
.
Lancet Oncol
2014
;
15
:
954
65
.
5.
Long
GV
,
Stroyakovskiy
D
,
Gogas
H
,
Levchenko
E
,
de
BF
,
Larkin
J
, et al
.
Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma
.
N Engl J Med
2014
;
371
:
1877
88
.
6.
Ascierto
P
,
McArthur
GA
,
Dreno
B
,
Atkinson
V
,
Liszkay
G
,
Di Giacomo
AM
, et al
.
Cobimetinib combined with vemurafenib in advanced BRAFV600-mutant melanoma (coBRIM): updated efficacy results froma randomised, double-blind, phase 3 trial
.
Lancet
2016
;
17
:
1248
60
.
7.
Hauschild
A
,
Grob
JJ
,
Demidov
LV
,
Jouary
T
,
Gutzmer
R
,
Millward
M
, et al
.
Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial
.
Lancet
2012
;
380
:
358
65
.
8.
Masuda
S
,
Izpisua Belmonte
JC
.
Trametinib for patients with advanced melanoma
.
Lancet Oncol
2012
;
13
:
e409
10
.
9.
Robert
C
,
Karaszewska
B
,
Schachter
J
,
Rutkowski
P
,
Mackiewicz
A
,
Stroiakovski
D
, et al
.
Improved overall survival in melanoma with combined dabrafenib and trametinib
.
N Engl J Med
2015
;
372
:
30
9
.
10.
Long
GV
,
Stroyakovskiy
D
,
Gogas
H
,
Levchenko
E
,
de Braud
F
,
Larkin
J
, 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
51
.
11.
Hodi
FS
,
O'Day
SJ
,
McDermott
DF
,
Weber
RW
,
Sosman
JA
,
Haanen
JB
, et al
.
Improved survival with ipilimumab in patients with metastatic melanoma
.
N Engl J Med
2010
;
363
:
711
23
.
12.
Robert
C
,
Schachter
J
,
Long
GV
,
Arance
A
,
Grob
JJ
,
Mortier
L
, et al
.
Pembrolizumab versus ipilimumab in advanced melanoma
.
N Engl J Med
2015
;
372
:
2521
32
.
13.
Robert
C
,
Long
GV
,
Brady
B
,
Dutriaux
C
,
Maio
M
,
Mortier
L
, et al
.
Nivolumab in previously untreated melanoma without BRAF mutation
.
N Engl J Med
2015
;
372
:
320
30
.
14.
Weber
JS
,
D'Angelo
SP
,
Minor
D
,
Hodi
FS
,
Gutzmer
R
,
Neyns
B
, et al
.
Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial
.
Lancet Oncol
2015
;
16
:
375
84
.
15.
Larkin
J
,
Chiarion-Sileni
V
,
Gonzalez
R
,
Grob
JJ
,
Cowey
CL
,
Lao
CD
, et al
.
Combined nivolumab and ipilimumab or monotherapy in untreated melanoma
.
N Engl J Med
2015
;
373
:
23
34
.
16.
Hugo
W
,
Zaretsky
JM
,
Sun
L
,
Song
C
,
Moreno
BH
,
Hu-Lieskovan
S
, et al
.
Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma
.
Cell
2016
;
165
:
35
44
.
17.
Cancer Genome Atlas Network
.
Genomic classification of cutaneous melanoma
.
Cell
2015
;
161
:
1681
96
.
18.
Shi
H
,
Hugo
W
,
Kong
X
,
Hong
A
,
Koya
RC
,
Moriceau
G
, et al
.
Acquired resistance and clonal evolution in melanoma during BRAF inhibitor therapy
.
Cancer Discov
2014
;
4
:
80
93
.
19.
Trunzer
K
,
Pavlick
AC
,
Schuchter
L
,
Gonzalez
R
,
McArthur
GA
,
Hutson
TE
, et al
.
Pharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanoma
.
J Clin Oncol
2013
;
31
:
1767
74
.
20.
Wu
TD
,
Nacu
S.
.
Fast and SNP-tolerant detection of complex variants and splicing in short reads
.
Bioinformatics
2010
;
26
:
873
81
.
21.
Wilm
A
,
Aw
PP
,
Bertrand
D
,
Yeo
GH
,
Ong
SH
,
Wong
CH
, et al
.
LoFreq: a sequence-quality aware, ultra-sensitive variant caller for uncovering cell-population heterogeneity from high-throughput sequencing datasets
.
Nucl Acids Res
2012
;
40
:
11189
201
.
22.
Lek
M
,
Karczewski
KJ
,
Minikel
EV
,
Samocha
KE
,
Banks
E
,
Fennell
T
, et al
.
Analysis of protein-coding genetic variation in 60,706 humans
.
Nature
2016
;
536
:
285
.
23.
Jiang
Y
,
Oldridge
DA
,
Diskin
SJ
,
Zhang
NR
.
CODEX: a normalization and copy number variation detection method for whole exome sequencing
.
Nucleic Acids Res
2015
;
43
:
e39
.
24.
McLaren
W
,
Gil
L
,
Hunt
SE
,
Riat
HS
,
Ritchie
GR
,
Thormann
A
, et al
.
The ensembl variant effect predictor
.
Genome Biol
2016
;
17
:
122
.
25.
Subramanian
A
,
Tamayo
P
,
Mootha
VK
,
Mukherjee
S
,
Ebert
BL
,
Gillette
MA
, et al
.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles
.
Proc Natl Acad Sci U S A
2005
;
102
:
15545
50
.
26.
Mootha
VK
,
Lindgren
CM
,
Eriksson
K-F
,
Subramanian
A
,
Sihag
S
,
Lehar
J
, et al
.
PGC-1α-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes
.
Nat Genet
2003
;
34
:
267
.
27.
Ritchie
ME
,
Phipson
B
,
Wu
D
,
Hu
Y
,
Law
CW
,
Shi
W
, et al
.
limma powers differential expression analyses for RNA-sequencing and microarray studies
.
Nucleic Acids Res
2015
;
43
:
e47
.
28.
Law
CW
,
Chen
Y
,
Shi
W
,
Smyth
GK
.
voom: precision weights unlock linear model analysis tools for RNA-seq read counts
.
Genome Biol
2014
;
15
:
R29
.
29.
Aran
D
,
Hu
Z
,
Butte
AJ
.
xCell: digitally portraying the tissue cellular heterogeneity landscape
.
Genome Biol
2017
;
18
:
220
.
30.
Ashburner
M
,
Ball
CA
,
Blake
JA
,
Botstein
D
,
Butler
H
,
Cherry
JM
, et al
.
Gene ontology: tool for the unification of biology. The Gene Ontology Consortium
.
Nat Genet
2000
;
25
:
25
9
.
31.
Gene Ontology Consortium
.
Expansion of the Gene Ontology knowledgebase and resources
.
Nucleic Acids Res
2017
;
45
:
D331
8
.
32.
Konieczkowski
DJ
,
Johannessen
CM
,
Abudayyeh
O
,
Kim
JW
,
Cooper
ZA
,
Piris
A
, et al
.
A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors
.
Cancer Discov
2014
;
4
:
816
27
.
33.
Hoek
KS
,
Eichhoff
OM
,
Schlegel
NC
,
Dobbeling
U
,
Kobert
N
,
Schaerer
L
, et al
.
In vivo switching of human melanoma cells between proliferative and invasive states
.
Cancer Res
2008
;
68
:
650
6
.
34.
Muller
J
,
Krijgsman
O
,
Tsoi
J
,
Robert
L
,
Hugo
W
,
Song
C
, et al
.
Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma
.
Nat Commun
2014
;
5
:
5712
.
35.
Kim
DW
,
Haydu
LE
,
Joon
AY
,
Bassett
RL
Jr
,
Siroy
AE
,
Tetzlaff
MT
, et al
.
Clinicopathological features and clinical outcomes associated with TP53 and BRAFNon-V600 mutations in cutaneous melanoma patients
.
Cancer
2017
;
123
:
1372
81
.
36.
Maertens
O
,
Johnson
B
,
Hollstein
P
,
Frederick
DT
,
Cooper
ZA
,
Messiaen
L
, et al
.
Elucidating distinct roles for NF1 in melanomagenesis
.
Cancer Discov
2013
;
3
:
338
49
.
37.
Nissan
MH
,
Pratilas
CA
,
Jones
AM
,
Ramirez
R
,
Won
H
,
Liu
C
, et al
.
Loss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependence
.
Cancer Res
2014
;
74
:
2340
50
.
38.
Flaherty
K
,
Davies
MA
,
Grob
JJ
,
Long
GV
,
Nathan
PD
,
Ribas
A
, et al
.
Genomic analysis and 3-y efficacy and safety update of COMBI-d: a phase 3 study of dabrafenib (D) + trametinib (T) vs D monotherapy in patients (pts) with unresectable or metastatic BRAF V600E/K-mutant cutaneous melanoma
.
J Clin Oncol
2016
;
34
:
(suppl; abstr 9502)
.
39.
Sumimoto
H
,
Imabayashi
F
,
Iwata
T
,
Kawakami
Y
.
The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells
.
J Exp Med
2006
;
203
:
1651
6
.
40.
Khalili
JS
,
Liu
S
,
Rodriguez-Cruz
TG
,
Whittington
M
,
Wardell
S
,
Liu
C
, et al
.
Oncogenic BRAF(V600E) promotes stromal cell-mediated immunosuppression via induction of interleukin-1 melanoma
.
Clin Cancer Res
2012
;
18
:
5329
40
.
41.
Wilmott
JS
,
Long
GV
,
Howle
JR
,
Haydu
LE
,
Sharma
RN
,
Thompson
JF
, et al
.
Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma
.
Clin Cancer Res
2012
;
18
:
1386
94
.
42.
Schilling
B
,
Paschen
A.
.
Immunological consequences of selective BRAF inhibitors in malignant melanoma: neutralization of myeloid-derived suppressor cells
.
Oncoimmunology
2013
;
2
:
e25218
.
43.
Frederick
DT
,
Piris
A
,
Cogdill
AP
,
Cooper
ZA
,
Lezcano
C
,
Ferrone
CR
, et al
.
BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma
.
Clin Cancer Res
2013
;
19
:
1225
31
.

Supplementary data