332
Views
114
CrossRef citations to date
0
Altmetric
Review

BRAF-mutant melanoma: treatment approaches, resistance mechanisms, and diagnostic strategies

, , , , , , & show all
Pages 157-168 | Published online: 16 Jan 2015

Abstract

BRAF inhibitors vemurafenib and dabrafenib achieved improved overall survival over chemotherapy and have been approved for the treatment of BRAF-mutated metastatic melanoma. More recently, the combination of BRAF inhibitor dabrafenib with MEK inhibitor trametinib has shown improved progression-free survival, compared to dabrafenib monotherapy, in a Phase II study and has received approval by the US Food and Drug Administration. However, even when treated with the combination, most patients develop mechanisms of acquired resistance, and some of them do not achieve tumor regression at all, because of intrinsic resistance to therapy. Along with the development of BRAF inhibitors, immunotherapy made an important step forward: ipilimumab, an anti-CTLA-4 monoclonal antibody, was approved for the treatment of metastatic melanoma; anti-PD-1 agents achieved promising results in Phase I/II trials, and data from Phase III studies will be ready soon. The availability of such drugs, which are effective regardless of BRAF status, has made the therapeutic approach more complex, as first-line treatment with BRAF inhibitors may not be the best choice for all BRAF-mutated patients. The aim of this paper is to review the systemic therapeutic options available today for patients affected by BRAF V600-mutated metastatic melanoma, as well as to summarize the mechanisms of resistance to BRAF inhibitors and discuss the possible strategies to overcome them. Moreover, since the molecular analysis of tumor specimens is now a pivotal and decisional factor in the treatment strategy of metastatic melanoma patients, the advances in the molecular detection techniques for the BRAF V600 mutation will be reported.

Background

Cutaneous melanoma is the most aggressive form of skin cancer, with a global incidence of about 200,000 new cases per year, likely to increase over the next years. Although melanoma represents only 4% of all types of skin cancers, it is correlated with about 80% of skin cancer-related deaths (about 65,000 per year). Survival rates depend on the clinical stage at the diagnosis, with 5-year survival ranging from 15% to 60% in patients with distant and local metastases, respectively.Citation1

The prognosis of metastatic melanoma has recently changed substantially thanks to the approval of kinase inhibitors vemurafenib,Citation2 dabrafenib,Citation3 and trametinib,Citation4 and the immune checkpoint inhibitor ipilimumab.Citation5,Citation6 More recently, immune checkpoint inhibitors nivolumabCitation7 and pembrolizumabCitation8 have achieved promising results in clinical trials, which will probably lead to the approval of these drugs by the regulatory agencies.

Vemurafenib and dabrafenib are selective inhibitors of BRAF V600, a mutation carried by almost half of melanomas,Citation9 and are approved by the US Food and Drug Administration (FDA) and European Medicines Agency for the treatment of unresectable or metastatic melanoma with mutant BRAF V600. In the Phase III trial of vemurafenib,Citation2 median overall survival (OS) was 13.6 months in the vemurafenib group, compared with 9.7 months in the dacarbazine group; median progression-free survival (PFS) was 6.9 months in the vemurafenib group and only 1.6 months in the dacarbazine group; response rate for vemurafenib was 57%, compared with 9% for dacarbazine. Dabrafenib achieved similar results, with a PFS of 5.1 months in the Phase III study.Citation3

Trametinib is an MEK inhibitor that achieved improved PFS (4.8 versus 1.5 months) compared to chemotherapy in a randomized Phase III studyCitation4 and, more importantly, was investigated as combination therapy with dabrafenib. In the Phase II trial comparing trametinib plus dabrafenib with dabrafenib alone,Citation10 the median PFS were 9.4 months and 5.8 months for patients treated with dabrafenib 150 mg twice daily plus trametinib 2 mg daily and dabrafenib monotherapy, respectively. On the basis of these results, at the beginning of 2014 the FDA approved the combination of dabrafenib plus trametinib for the treatment of unresectable or metastatic melanoma with a BRAF V600E or V600K mutation.

Other kinase inhibitors are in late stages of clinical development. LGX818 is a potent and selective BRAF inhibitor with a dissociation half-time about 10 times longer than other BRAF inhibitors; in the Phase I trial a response rate as high as 67% was achieved among BRAF inhibitors-naïve patients.Citation11 A three-arm Phase III trial is currently recruiting participants in order to compare the efficacy and safety of LGX818 monotherapy and LGX818 in combination with MEK inhibitor MEK162 as compared to vemurafenib in patients with locally advanced unresectable or metastatic melanoma with BRAF V600 mutation (NCT01909453). Moreover, a Phase III clinical trial comparing vemurafenib in combination with MEK inhibitor cobimetinib versus vemurafenib monotherapy (NCT01689519) met its primary endpoint (PFS), and these data are planned to be submitted to health authorities.Citation12

Along with the development of BRAF and MEK inhibitors, immunotherapy made some steps forward as well: ipilimumab, a fully human IgG1 monoclonal antibody that blocks cytotoxic T-lymphocyte antigen (CTLA)-4 to elicit antitumor T-cell-mediated responses, was approved for the treatment of metastatic melanoma as it achieved a statistically significant improvement in OS in two different randomized Phase III trials in pretreatedCitation5 and in treatment-naïveCitation6 patients with metastatic melanoma; nivolumab and pembrolizumab, monoclonal antibodies targeting the programmed cell death-1 (PD-1) receptor on infiltrating T-cells, which otherwise produces an inhibition of T-cells directed against melanoma antigens, showed promising clinical activity and efficacy, and pembrolizumab was recently approved by the FDA for the treatment of patients with unresectable or metastatic melanoma and disease progression following ipilimumab and, if BRAF V600 mutation positive, a BRAF inhibitor. A compassionate use of both drugs is available in several countries all over the world.

Signaling pathways in BRAF-mutated melanoma

The mitogen-activated protein kinase (MAPK) pathway plays an important role in the pathogenesis of melanoma. This pathway is physiologically activated when extracellular signals bind to their cognate membrane receptor, typically a receptor tyrosine kinase (RTK). RAF kinases are components of the pathway: their activity requires the formation of dimers, which is promoted by RAS activation. Activated RAF kinases phosphorylate and activate MEK1/2, which in turn phosphorylate and activate ERK1/2, leading to cellular proliferation, survival, and differentiation, and to an inhibitory feedback toward upstream components of the pathway.Citation13

About 50% of melanomas harbor an activating mutation in BRAF, the most common being BRAFV600E,Citation9 which renders the kinase constitutively active. In BRAF-mutated melanomas, RAS is negatively regulated by ERK-dependent feedback and BRAFV600E exists mainly as a monomer.Citation13

BRAF inhibitors inhibit ERK signaling only in BRAF-mutated tumors. In wild-type (WT) cells, BRAF and CRAF form homo- and heterodimers on RAS activation; BRAF inhibitors binding to one member of the dimer causes an allosteric transactivation of the drug-free protomer, leading to ERK activation. This is enhanced when RAS, which promotes RAF dimerization, is overexpressed:Citation14 in fact, most cutaneous tumors developing in patients treated with BRAF inhibitors harbor RAS mutations.Citation15

The PI3K/AKT/mTOR pathway is a key regulator of cellular growth and protein synthesis.Citation16,Citation17 The MAPK pathway interacts with the PI3K/AKT/mTOR pathway at multiple points: for example, RAS directly activates PI3K, and the inhibition of a pathway may upregulate the other one.Citation16,Citation18

Diagnostic strategies

Detection of BRAF V600 mutations has recently become mandatory to treat patients with advanced or metastatic melanoma. Several methods have been used to detect BRAF mutations, including Sanger sequencing, mismatch ligation assay, ligase detection reaction, denaturating high-performance liquid chromatography, SNAPshot®, high-resolution melting, mutation-specific polymerase chain reaction (PCR) and mutation-specific real-time (RT) PCR, including EntroGen molecular probes (CE-IVD), pyrosequencing, and mass spectrometry.Citation19Citation24 Each method has its own sensitivity, specificity, cost, and response delay. Several studies have been performed to compare the methods for detection of these mutations.Citation24Citation29

Recently, detection of BRAF p.V600E mutation was also performed by immunohistochemistry (IHC) with VE1 antibody, and next-generation sequencing (NGS) technology is currently being used to analyze melanoma specimens in many research institutions. Below, we compare the most commonly used methods for the detection of BRAF mutations in melanomas to determine the method or combination of methods that should be used in diagnostic daily practice (). Our suggestion is that sequential analysis, with initial detection of p.V600E-positive cases by IHC and/or Sanger sequencing, followed by pyrosequencing or RT-PCR-based tests in negative or uninterpretable cases, is the most efficient method to use in daily practice, in certified laboratories, with validated techniques.

Table 1 Sensitivity and specificity of the described methods

Immunohistochemistry

IHC with VE1 monoclonal antibody was found to be efficient to detect p.V600E mutations.Citation30 The advantages of this technique are that only two slides are needed, no specialized equipment is needed, it is cheaper than other techniques, and results are easily obtained within 48 hours. Interestingly, IHC with VE1 monoclonal antibody was reported to have a 100% sensitivity and specificity for detection of p.V600E, even when compared with methods more sensitive than Sanger sequencing.Citation31 However, important disadvantages are that staining interpretation is easy in most, but not all, cases, and that the VE1 antibody is highly specific for the V600E mutation, but other clinically relevant BRAF mutations are missed. For these reasons, it could be used as a cost-effective first-line method for BRAF V600E detection in a daily practice sequential combination of methods.Citation32

Sanger sequencing

For many years, Sanger sequencing has been considered the reference method for identification of acquired mutations in tumors. However, because of its low sensitivity (direct sequencing cannot detect the presence of mutant alleles when the mutant/WT ratio is less than 1:5),Citation28 detection of mutations from tumor DNA requires a high percentage of tumor cells within the samples, a requirement that cannot always be met in routine diagnostic testing of human samples. A recent study compared four methods for the detection of BRAF mutations in metastatic melanomas.Citation31 Microdissection of metastatic melanomas was performed to increase the number of tumor cells to at least 60%. However, 3 of the 40 melanomas with p.V600E mutation detected by other methods were negative with Sanger sequencing. The sensitivity of Sanger sequencing was 92.5% (95% confidence interval [CI], 78.5%–98.0%) for p.V600E detection. Thus, 7.5% of patients eligible for treatment with BRAF inhibitors would have been excluded. In line with this, another recently published series found a false-negative rate of Sanger detection of BRAF mutations of 9.2%.Citation24 Interestingly, false-negative cases with Sanger sequencing corresponded to tumors with a high proportion of tumor cells within the samples. This finding may be related to tumor heterogeneity, with only some subclones containing p.V600E mutation and others being WT. These data indicate that Sanger sequencing failed to detect BRAF mutations in melanomas and therefore should no longer be considered as the reference test, but as a first screening or a confirmation test. Alternatively, in samples negative with Sanger sequencing, one of the tests described below should be performed.

Pyrosequencing

Pyrosequencing is an RT sequencing by synthesis approach which allows the quantification of mutated alleles. It is a rapid and more sensitive method compared with direct sequencing for quantifying the BRAF V600 mutation. In a study comparing different techniques, pyrosequencing has been shown to have 100% sensitivity (95% CI, 92.4%–100%) and specificity (95% CI, 91.6%–100%) for p.V600 detection.Citation31 Another recent study comparing different detection methods found that there was no difference in sensitivity between the high-resolution melting analysis and Sanger sequencing (98%). All mutations down to 6.6% allele frequency could be detected with 100% specificity. In contrast, pyrosequencing detected 100% of the mutations down to 5% allele frequency but exhibited only 90% specificity, being prone to errors without using a customer-designed setup to identify all BRAF mutations at codon V600.Citation29 Overall, pyrosequencing could be performed only on cases not interpretable or negative, corresponding to approximately 50%–60% of samples analyzed by IHC and Sanger sequencing.

RT-PCR-based tests

Several RT-PCR-based tests, certified or not, have been developed, with an overall sensitivity of 97.5% (higher than that of Sanger sequencing) and variable specificity (depending on their design specific for V600E mutation or not, and thus ranging from 87.8% to 100%). The two FDA/CE-IVD-approved tests for BRAF mutations (cobas® 4800 BRAF V600 mutation test and THxID®-BRAF) are both RT-PCR-based assays. Major disadvantages are that they are optimized for the most common BRAF mutation, and less common BRAF mutations that may still be responsive to BRAF inhibitor therapy may be missed. Further, although these assays require only a small amount of DNA, if the specimen contains <10% of tumor cells, these assays may fail to detect the mutation (the cobas® test guidelines recommend a tumor content of 50% in samples).

Recently, the cobas® test was found to detect only 70% of p.V600K, whereas 100% of p.V600E was detected.Citation24 Interestingly, in a study comparing different techniques, RT-PCR detected 39 of the 40 p.V600E mutations and all WT cases. The sensitivity of RT PCR for p.V600E detection was 97.5% (95% CI, 87.1% to 99.6%), and specificity was 87.8% (95% CI, 75.8%–94.3%). Surprisingly, the six cases with p.V600K mutation were also positive, although with a lower signal. The four other BRAF mutations, including p.V600R, were not detected by RT-PCR. The RT-PCR detection of p.V600K was possible only after inhibition of WT allele amplification. This may be the cause of a lower sensitivity.Citation31 Inhibition of the WT allele amplification, as performed with peptide nucleic acids (PNAs) to analyze the mutated allele, is in fact an added value to these techniques.

Recently, CE-IVD RT-PCR tests based on PNA inhibition have been developed to detect all the mutations at codon V600, with a sensitivity comparable to that of pyrosequencing.Citation33 PNA-mediated clamping PCR (PNA-clamping PCR) is based on the principle that PNA inhibits WT by hybridizing normal sequences, and therefore mutant DNA is preferentially amplified.Citation34 Indeed, PNA clamp RT-PCR detected a 0.5% BRAF V600E mutant in the background of the WT with high sensitivity. PNA-clamping PCR may offer a sensitive and reliable alternative method to pyrosequencing, particularly for the detection of a small amount of mutant.

Overall, RT-PCR-based assays, however, provide qualitative information only on BRAF at codon 600; no other genes are characterized, and the results are not quantitative (how much mutated BRAF is present). Further, when they are optimized for the most common BRAF mutation (V600E), less common BRAF mutations that may still be responsive to inhibitor therapy may be missed with the exception of PNA-clamp PCR, which detects all mutations at p.V600 with a sensitivity comparable to that of pyrosequencing.

A recent study comparing different methods for detecting BRAF mutations concluded that in their present setup, the cobas® 4800 BRAF V600 test as well as the therascreen® BRAF Pyro Kit (Qiagen NV, Venlo, the Netherlands) are not sufficient for the European approval of vemurafenib because there is a therapeutic option for melanoma patients with any mutation in codon 600 of the BRAF gene.Citation35 The authors suggest a combination of VE1 antibody staining and high-resolution melting or sequencing for p.V600E mutation analysis, combining the lowest detection limit with a fast, reliable method with 100% sensitivity for routine diagnostics at the moment.

Next-generation sequencing

NGS is currently being used to analyze melanoma specimens in many research institutions. Many platforms are available, but what they have in common is that they are massively parallel sequencing techniques in which relatively small stretches (which may cover an exon, a gene, or the whole genome) of DNA are sequenced many times (typically 20 to several hundred times). NGS has several advantages over RT-PCR techniques, the most important being that it provides far more genetic information: besides mutations, NGS can detect rearrangements, amplifıcations, and deletions, and can analyze many genes. This will become increasingly important as we discover other actionable mutations/mechanisms of resistance to BRAF inhibition. NGS is also more sensitive than many RT-PCR assays, and can detect mutations even when tumor DNA represents less than 10% of the total DNA. NGS could analyze 100% of the cases with 100% specificity and exhibited 98.6% sensitivity in a recent study comparing different methods.Citation29 Among disadvantages, NGS generally requires more tumor material, has a longer turn-around time, and requires a higher expertise in computational biology than any other established methods. Most of the information obtained is not yet clinically relevant, and the assays are not FDA approved. International validation is ongoing. NGS requires specialized equipment, computers, and bioinformatics, making it more expensive. This makes NGS largely a research tool at this time. In the near future and with growing experiences, it is an inevitable fact that NGS will replace all established methods for molecular diagnostics, in view of the high sensitivity and multiplexing options of this method allowing generation of a molecular profile of each tumor sample analyzed.Citation29 A recent study performing whole cancer genome sequencing by NGS methods states that almost 75% of cancer gene variations may be missed by an approach analyzing only hotspot mutations.Citation36

Treatment approaches

The presence of a BRAF V600 mutation is an important factor to decide the treatment approach that is the best for each patient, but it is not the only one. In fact, first-line treatment with BRAF inhibitors or BRAF inhibitors in combination with MEK inhibitors may not be the best therapeutic strategy for all patients, and the possibility to start with immunotherapy must be considered. FDA approval of ipilimumab includes the first-line treatment of metastatic melanoma, and even if in Europe, initially, the indication was in pretreated patients only, since October 2013 it has been broadened to first-line as well. Moreover, anti-PD-1 agents achieved promising results in clinical trialsCitation7,Citation8,Citation37 and may be recommended soon as a first-line treatment.

The role of chemotherapy as a frontline approach for BRAF-mutated metastatic melanoma has been limited by the introduction of targeted therapies. Currently, dacarbazine is the only FDA-approved chemotherapeutic drug for the treatment of metastatic melanoma, and it has not been shown to improve PFS or OS. BRAF inhibitors and BRAF inhibitors in combination with MEK inhibitors achieve tumor regression in a high rate of patients (50%–76%),Citation2,Citation3,Citation10 PFSCitation2,Citation3 and OSCitation2 are improved compared to those seen with chemotherapy, and the onset of tumor regression is early,Citation41 allowing to treat successfully even symptomatic patients with low performance status and a rapidly evolving disease. Even if mechanisms of resistance, which will be discussed in the “Resistance mechanisms” section, arise in most patients, leading to tumor regrowth, there is the chance for long-term survival at least for a subset of patients, with 26% of patients from the Phase I study being alive at 3 years.Citation38 Emerging clinical evidence suggests that extended BRAF inhibition after progression on BRAF inhibitors may prolong survival.Citation42,Citation43 In a series of 114 patients treated with vemurafenib and dabrafenib within clinical trials, continued therapy with BRAF inhibitors after progressive disease was associated with prolonged survival;Citation42 similar results were observed in the Phase I study of vemurafenib.Citation38 However, these data may be biased by patient selection, and prospective randomized trials are needed to investigate the role that prolonged BRAF inhibition may have in the treatment strategies for BRAF-mutated patients. A prospective, single-arm Phase II study was designed to evaluate the activity of treatment after progression during the therapy with vemurafenib with the combination of vemurafenib and fotemustine (NCT01983124).

Preliminary results of the Phase III trial of the combination of dabrafenib and trametinib versus dabrafenib monotherapy were presented at ASCO 2014 annual meeting:Citation39 even if PFS was only slightly better in the combination arm (9.3 versus 8.8 months), dabrafenib and trametinib achieved 29% improvement in response rate and 37% reduction in risk of death over the monotherapy. In addition, the analysis ad interim of the other Phase III trial comparing the combination of dabrafenib and trametinib with vemurafenib monotherapy showed an OS benefit in the combination arm, allowing the crossover to the combination arm for the patients in treatment with vemurafenib.Citation40

In contrast, the response rate and PFS with ipilimumab are lower than those with BRAF/MEK inhibitors and the onset of tumor regression is slow, as it may take time to build an immune antitumor response. However, even if the number of objective responses is relatively low, ipilimumab can induce long-lasting disease control and long-term survival: 18.2% of patients treated within Phase II studies with the approved dose of ipilimumab were alive after 4 years.Citation44

Retrospective clinical data seem to indicate that the activity of ipilimumab is not influenced by BRAF mutational status,Citation45 but the administration of ipilimumab after the failure of BRAF inhibitors may have suboptimal results;Citation46Citation48 however, no prospective data are available to date. In the ECOG E1612 trial, patients with BRAF-mutated metastatic melanoma were randomized to receive either ipilimumab followed by vemurafenib at progression or vice versa; patients were stratified based on ECOG performance status (0 or 1), stage (III and M1a/b or M1c), and prior treatment (yes or no). ECOG trial E4613 will similarly investigate sequential treatment with the combinations ipilimumab and nivolumab versus dabrafenib and trametinib. Until the availability of prospective data from these studies, the choice of first-line treatment for BRAF-mutated metastatic melanoma patients relies on retrospective data and expert opinion. Patients with poor performance status and rapidly evolving disease, whose estimated life expectancy is less than 3 months, may not benefit from front-line treatment with ipilimumab, as may patients with high LDH levels.Citation49

The presence of brain metastasis is historically associated with lack of efficacy of systemic therapies and poor prognosis.Citation50 Even if ipilimumab showed clinical activity in patients with pretreated, asymptomatic, and not steroid-dependent brain metastasis,Citation51,Citation52 only BRAF inhibitors have evidence of activity in case of active brain metastasis.Citation53Citation55

The identification of new biomarkers may also help selecting patients who are likely to respond to ipilimumab: for example, CTLA-4 gene polymorphisms seem to influence the response to anti-CTLA-4 antibodies,Citation56 and some immunological signature may predict response to immunotherapy in general.Citation57

Anti-PD-1 agents nivolumab and pembrolizumab achieved higher response rates in Phase I–II studies than did ipilimumab (25%–38%),Citation7,Citation8 and the onset of response was shorter. The duration of response is also impressive, with most responses lasting more than 12 months. Their availability in daily clinical practice may influence the therapeutic strategies, as also patients unfit to be treated with ipilimumab may benefit from treatment with anti-PD-1 drugs. Until then, patients with short life expectancy or who are unlikely to respond to ipilimumab should be treated with BRAF inhibitors or BRAF and MEK inhibitors in combination as a first-line treatment. Two BRAF inhibitors are currently available: vemurafenib and dabrafenib. Data from clinical trials showed that they have substantially the same clinical activity in patients with the BRAF V600E mutation, but they slightly differ in toxicity. Both treatments are well-tolerated, and dose reductions were needed in the range of 28% for dabrafenibCitation3 and 38% for vemurafenibCitation2 in the Phase III studies. Cutaneous side effects, fatigue, arthralgia, and nausea are the most common adverse events shared by the two drugs;Citation2,Citation3 however, vemurafenib causes a higher rate of hepatic transaminitis, photosensitivity, and cutaneous hyperproliferative lesions (including squamous cell carcinomas and keratoacanthomas), whereas pyrexia is more commonly seen with dabrafenib.Citation2,Citation3 As for the BRAF V600K mutation, which is the second most common BRAF V600 mutation in melanoma (19% as compared with 73% for BRAF V600E),Citation58 in the Phase III study, vemurafenib achieved similar PFS and OS irrespective of the mutation;Citation2 in the Phase III trial of dabrafenib,Citation3 patients whose melanoma harbored a V600K mutation were excluded from the study, but some data are available from the Phase II trial:Citation59 only 2 patients with the V600K mutation had a response (13%), compared with 45 patients (59%) harboring V600E. However, median PFS (4.5 versus 6.3 months) and OS (12.9 versus 13.1 months) were similar in the two groups. About 8% of melanomas harbor other genotypes than V600E and V600K,Citation58 such as V600R, and some clinical evidence suggest that vemurafenib and dabrafenib may have clinical activity in this setting.Citation60 The clinical activity of BRAF inhibitors in patients whose melanomas harbor a non-V600E mutation underlines the importance of using a diagnostic tool that detects all BRAF V600 mutations.

The role of trametinib monotherapy may be limited as a first-line treatment of BRAF-mutated melanoma, as well as after the failure of therapy with BRAF inhibitors.Citation61 In the Phase II study of MEK inhibitor trametinib in BRAF-mutated patients,Citation61 there were no confirmed objective responses in the cohort of patients previously treated with a BRAF inhibitor. On the contrary, combination therapy with trametinib and dabrafenib achieved improved clinical activity over dabrafenib monotherapy, but adverse events leading to treatment discontinuation (9% versus 5%), dose reduction (24% versus 13%), and dose interruption (45% versus 30%) were more frequent in patients treated with the combination than in those receiving only dabrafenib.Citation43 Pyrexia was the most notable risk for the combination compared with dabrafenib, while fewer cutaneous hyperproliferative events were observed in the combination arm, consistent with the identification of activating RAS mutations in most skin tumors developing during therapy with BRAF inhibitors.Citation13

Resistance mechanisms

About 15% of patients treated with BRAF inhibitors do not achieve tumor regression, because of intrinsic/primary mechanisms of resistance, and most patients who respond to therapy ultimately develop a mechanism of acquired/secondary resistance, leading to progressive disease.

Mechanisms of primary resistance include RAC1P29S mutations,Citation62 COT overexpression,Citation63 alterations in RTK signaling,Citation64,Citation65 loss of function of NF1,Citation66Citation69 alterations in the RB1 pathway,Citation70,Citation71 and alterations in the PI3K-AKT-mTOR pathway (loss of function of PTEN) ().Citation71Citation74

Table 2 Mechanisms of intrinsic/primary resistance

The reactivation of the MAPK pathway is the most frequent cause of acquired/secondary resistance; it may be driven by events that occur upstream (upregulation and activation of the RTKs,Citation75Citation79 NRAS activating mutationsCitation75), downstream (activating MEK1/2 mutationsCitation62,Citation74,Citation80Citation83), or at the level of BRAF (alternative splicing of V600E BRAF,Citation84 BRAFV600E copy number amplification,Citation85 elevated CRAF levelsCitation86) (). The PI3K-PTEN-AKT pathway is a second core resistance pathway: AKT1/3 mutations and mutations in PI3K-AKT positive-regulatory and negative-regulatory genes may upregulate this pathway,Citation62,Citation80,Citation87 driving resistance to BRAF inhibitor.

Table 3 Mechanisms of acquired/secondary resistance

No association was observed between clinical outcome (best response and PFS) and specific mechanisms of resistance.Citation80 Some tumors develop multiple mechanisms of resistance simultaneously in the same patient (intrapatient heterogeneity) or even in the same lesion (intratumor heterogeneity).Citation60,Citation87

In addition to intrinsic and acquired resistance, mechanisms of adaptive response to BRAF inhibition limit the efficacy of treatment with BRAF inhibitors, leading mostly to partial responses, with complete response rate being in the range of only 3%–6% in the Phase III studies of vemurafenib and dabrafenib.Citation2,Citation3 In BRAF-mutated cells, ERK transcriptional products are upregulated, including negative-feedback components, which suppress RAS activation. As a result, RAS does not promote RAF dimerization and BRAF exists predominantly as an active monomer. Treatment with BRAF inhibitors, in addition to arresting tumor growth, relieves ERK negative feedback, partially restoring the sensitivity to extracellular signaling and the activity of RAS, promoting the formation of RAF dimers. BRAF inhibitors bind to one component of the dimer and cause an allosteric activation of the other one.Citation14 ERK is reactivated and negative-feedback pathways are partially restored over time, leading to the formation of a new steady state of reactivated ERK signaling, which is different among different cell lines.Citation13 The PI3K-AKT-mTOR pathway is also involved in the mechanisms of adaptive resistance: in fact, the inhibition of the MAPK pathway leads to early, adaptive AKT signaling, unleashing a rebound activation of PI3K-AKT pathway.Citation88

Discussion and conclusion

Intrapatient and intratumor heterogeneity of resistance,Citation62,Citation87,Citation89,Citation90 cross-resistance,Citation17,Citation91 and alternative pathways activationCitation88 are a challenge for personalized targeted therapies. Preclinical and clinical evidence suggest that one strategy to overcome resistance to BRAF inhibition may be the combination of multiple inhibitors. The combination of BRAF and MEK inhibitors dabrafenib and trametinib achieved a slightly better PFS over dabrafenib monotherapy in the Phase III study. Nevertheless, most mechanisms of resistance may confer cross-resistance to MEK inhibition and may lead to the activation of additional pathways, such as PI3K-AKT-mTOR, suggesting that multiple pathways may be needed to be targeted to achieve durable responses.Citation17,Citation91 Phase I/II trials are evaluating the safety and clinical activity of such combination regimens in patients ().

Table 4 Ongoing Phase I/II studies investigating multitargeted combinations

An alternative to combination strategies may be an adaptive sequential approach based on the biopsy of progressing tumors during therapy with BRAF inhibitors. A Phase II study (NCT01820364) is currently recruiting metastatic melanoma patients who progress on treatment with BRAF inhibitor LGX818: resistant tumors will be biopsied and compared with a pretreatment biopsy to identify the mechanism of resistance. On the basis of the alterations identified in the tumor samples, a second agent from a list of MEK, CDK4/6, FGFR, PI3K, and c-MET inhibitors will be added to the regimen. A limitation of this study is that single biopsy may underestimate the tumor genomics landscape due to tumor heterogeneity. In the future, the analysis of circulating tumor cells or circulating tumor-derived DNA may provide a complete genetic profile compared to single tumor biopsies,Citation92 but no standard method for their detection and molecular analysis is currently available.Citation93,Citation94

In addition to inhibiting multiple molecular targets, either in combination or in sequence, the combination of BRAF inhibitors and immunotherapy may be a strategy to provide durable responses in a high rate of metastatic melanoma patients. BRAF inhibitors do not seem to impair the immune systemCitation95,Citation96 and, on the contrary, may enhance immune activation.Citation97Citation102 Combined BRAF inhibitor vemurafenib and anti-CTLA-4 antibody ipilimumab are not tolerated.Citation103 Nevertheless, anti-PD-1 antibodies pembrolizumab and nivolumab seem to be more tolerated than ipilimumab: their safety in combination with BRAFi and BRAFi + MEKi is under investigation in Phase I–II clinical studies. In addition, clinical studies are underway to determine the safety and clinical activity of the combination of BRAFi with other immunotherapeutic agents such as anti-PDL-1 antibodies, interleukin, adoptive cell therapy, and interferon ().

Table 5 Ongoing Phase I/II studies investigating the combination of MAPK inhibitors with immunotherapy

As mentioned in the “Treatment approaches” section, clinical evidence suggests that prolonged BRAF inhibition after progression on BRAF inhibitors may prolong survival in a subset of patients.Citation42,Citation43 In contrast with these clinical observations, Das Thakur et al demonstrated that cessation of BRAF inhibition may lead to regression of tumors expressing BRAF alternative splicing variants or amplified BRAF and that, in vivo, a discontinuous dosing strategy delayed the onset of resistance over continuous administration.Citation104Citation106 The safety and efficacy of an intermittent regimen with BRAF inhibitors will be prospectively evaluated in a Phase II trial (NCT01894672): LGX818 will be administered on a daily schedule dosing for the first 6 weeks; this will be followed by a 2-week break and, thereafter, patients will resume LGX818 on a 2-weeks-on/2-weeks-off schedule.

Until further investigations and availability of new drugs, in clinical practice patients with rapid and extensive progressive disease during treatment with BRAF inhibitors, alone or in combination with MEK inhibitors, are unlikely to benefit from extended MAPK inhibition and should switch to another treatment; patients with isolated progression, on the other hand, could continue treatment with BRAF inhibitors after local treatment of resistant lesions, as resistance mechanisms are not always shared by all metastases.Citation89

Disclosure

The authors have no financial interests in any of the products, devices, or drugs mentioned in this paper. Paola Queirolo has received lecture fees from Bristol-Myers Squibb and Roche and served on advisory boards for Bristol-Myers Squibb, Roche, and GSK; Francesco Spagnolo has received lecture fees from Bristol-Myers Squibb, GSK, and Roche. The authors declare no other conflicts of interest.

References

  • SiegelRMaJZouZJemalACancer statistics, 2014CA Cancer J Clin201464192924399786
  • McArthurGAChapmanPBRobertCSafety and efficacy of vemurafenib in BRAFV600E and BRAFV600K mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label studyLancet Oncol201415332333224508103
  • HauschildAGrobJ-JDemidovLVDabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trialLancet2012380983935836522735384
  • FlahertyKTRobertCHerseyPImproved survival with MEK inhibition in BRAF-mutated melanomaN Engl J Med2012367210711422663011
  • HodiFSO’DaySJMcDermottDFImproved survival with Ipilimumab in patients with metastatic melanomaN Engl J Med2010363871172320525992
  • RobertCThomasLBondarenkoIIpilimumab plus Dacarbazine for previously untreated metastatic melanomaN Engl J Med2011364262517252621639810
  • TopalianSLSznolMMcDermottDFSurvival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving NivolumabJ Clin Oncol201432101020103024590637
  • HamidORobertCDaudASafety and tumor responses with Lambrolizumab (Anti–PD-1) in melanomaN Engl J Med2013369213414423724846
  • DaviesHBignellGRCoxCMutations of the BRAF gene in human cancerNature2002417689294995412068308
  • FlahertyKTInfanteJRDaudACombined BRAF and MEK inhibition in melanoma with BRAF V600 mutationsN Engl J Med2012367181694170323020132
  • DummerRRobertCNyakasMInitial results from a phase I, open-label, dose escalation study of the oral BRAF inhibitor LGX818 in patients with BRAF V600 mutant advanced or metastatic melanomaJ Clin Oncol201331suppl abstr 9028
  • LarkinJAsciertoPADrénoBCombined vemurafenib and cobimetinib in BRAF-mutated melanomaN Engl J Med20141113371201867187625265494
  • LitoPRosenNSolitDBTumor adaptation and resistance to RAF inhibitorsNat Med201319111401140924202393
  • PoulikakosPIZhangCBollagGShokatKMRosenNRAF inhibitors transactivate RAF dimers and ERK signalling in cells with wild-type BRAFNature2010464728742743020179705
  • SuFVirosAMilagreCRAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitorsN Engl J Med2012366320721522256804
  • MendozaMCErEEBlenisJThe Ras-ERK and PI3K-mTOR pathways: cross-talk and compensationTrends Biochem Sci201136632032821531565
  • BrittenCDPI3K and MEK inhibitor combinations: examining the evidence in selected tumor typesCancer Chemother Pharmacol20137161395140923443307
  • Sánchez-HernándezIBaqueroPCallerosLChiloechesADual inhibition of V600EBRAF and the PI3K/AKT/mTOR pathway cooperates to induce apoptosis in melanoma cells through a MEK-independent mechanismCancer Lett2012314224425522056813
  • XingMTufanoRPTufaroAPDetection of BRAF mutation on fine needle aspiration biopsy specimens: a new diagnostic tool for papillary thyroid cancerJ Clin Endocrinol Metab20048962867287215181070
  • SpittleCWardMRNathansonKLApplication of a BRAF pyrosequencing assay for mutation detection and copy number analysis in malignant melanomaJ Mol Diagn20079446447117690212
  • ThomasRKBakerACDeBiasiRMHigh-throughput oncogene mutation profiling in human cancerNat Genet200739334735117293865
  • PichlerMBalicMStadelmeyerEEvaluation of high-resolution melting analysis as a diagnostic tool to detect the BRAF V600E mutation in colorectal tumorsJ Mol Diagn200911214014719213871
  • JarryAMassonDCassagnauEParoisSLaboisseCDenisMGReal-time allele-specific amplification for sensitive detection of the BRAF mutation V600EMol Cell Probes200418534935215294323
  • AndersonSBloomKJValleraDUMultisite analytic performance studies of a real-time polymerase chain reaction assay for the detection of BRAF V600E mutations in formalin-fixed, paraffin-embedded tissue specimens of malignant melanomaArch Pathol Lab Med20121361113851391Accessed September 1, 201422332713
  • IbrahemSSethRO’SullivanBFadhilWTanierePIlyasMComparative analysis of pyrosequencing and QMC-PCR in conjunction with high resolution melting for KRAS/BRAF mutation detectionInt J Exp Pathol201091650050521199003
  • HalaitHDeMartinKShahSAnalytical performance of a real-time PCR-based assay for V600 mutations in the BRAF gene, used as the companion diagnostic test for the novel BRAF Inhibitor Vemurafenib in metastatic melanomaDiagn Mol Pathol201221118 Available at: http://journals.lww.com/molecularpathology/Full-text/2012/03000/Analytical_Performance_of_a_Real_time_PCR_based.1.aspxAccessed September 1, 201422306669
  • HeidemanDAMLurkinIDoelemanMKRAS and BRAF mutation analysis in routine molecular diagnosticsJ Mol Diagn14324725522425762
  • CarbonellPTurpinMCTorres-MorenoDComparison of allelic discrimination by dHPLC, HRM, and TaqMan in the detection of BRAF mutation V600EJ Mol Diagn2011135467473Accessed September 1, 201421708284
  • IhleMFassunkeJKonigKComparison of high resolution melting analysis, pyrosequencing, next generation sequencing and immunohistochemistry to conventional Sanger sequencing for the detection of p.V600E and non-p.V600E BRAF mutationsBMC Cancer20141411324410877
  • LongGVWilmottJSCapperDImmunohistochemistry is highly sensitive and specific for the detection of V600E BRAF mutation in melanomaAm J Surg Pathol20133716165 Available at: http://journals.lww.com/ajsp/Fulltext/2013/01000/Immunohistochemis-try_Is_Highly_Sensitive_and.7.aspxAccessed September 1, 201423026937
  • ColombaEHélias-RodzewiczZVon DeimlingADetection of BRAF p.V600E mutations in melanomas: comparison of four methods argues for sequential use of immunohistochemistry and pyrosequencingJ Mol Diagn20131519410023159108
  • BoursaultLHaddadVVergierBTumor homogeneity between primary and metastatic sites for BRAF status in metastatic melanoma determined by immunohistochemical and molecular testingPLoS ONE201388e7082623976959
  • JeongDJeongYParkJBRAF V600E mutation analysis in papillary thyroid carcinomas by peptide nucleic acid clamp real-time PCRAnn Surg Oncol201320375976623179992
  • KangS-HPyoJYYangS-WHongSWDetection of BRAF V600E mutation with thyroid tissue using pyrosequencing: comparison with PNA-clamping and real-time PCRAm J Clin Pathol2013139675976423690118
  • Da Rocha DiasSSalmonsonTvan Zwieten-BootBThe European Medicines Agency review of vemurafenib (Zelboraf®) for the treatment of adult patients with BRAF V600 mutation-positive unresectable or metastatic melanoma: summary of the scientific assessment of the Committee for Medicinal Products for Human UseEur J Cancer20134971654166123481513
  • RossJSCroninMWhole cancer genome sequencing by next-generation methodsAm J Clin Pathol2011136452753921917674
  • RobertCLongGVBradyBNivolumab in Previously Untreated Melanoma without BRAF MutationN Engl J Med11162014 [Epub ahead of print]
  • KimKAmaravadiRKFlahertyKSignificant long-term survival benefit demonstrated with vemurafenib in ongoing Phase I studyPigment Cell Melanoma Res201225866 [abstract]
  • LongGVStroyakovskyDLGogasHCOMBI-d: A randomized, double-blinded, Phase III study comparing the combination of dabrafenib and trametinib to dabrafenib and trametinib placebo as first-line therapy in patients (pts) with unresectable or metastatic BRAFV600E/K mutation-positive cutaneous melanomaJ Clin Oncol325s2014suppl abstr 9011
  • RobertCKaraszewskaBSchachterJImproved Overall Survival in Melanoma with Combined Dabrafenib and TrametinibN Engl J Med20141116 [Epub ahead of print]
  • McArthurGAPuzanovIAmaravadiRMarked, homogeneous, and early [18F]fluorodeoxyglucose–positron emission tomography responses to vemurafenib in BRAF-mutant advanced melanomaJ Clin Oncol201230141628163422454415
  • ChanMMKHayduLEMenziesAMThe nature and management of metastatic melanoma after progression on BRAF inhibitors: effects of extended BRAF inhibition: BRAF inhibitor progression in melanomaCancer2014120203142315324985732
  • CarlinoMSGowrishankarKSaundersCABAntiproliferative effects of continued mitogen-activated protein kinase pathway inhibition following acquired resistance to BRAF and/or MEK inhibition in melanomaMol Cancer Ther20131271332134223645591
  • WolchokJDWeberJSMaioMFour-year survival rates for patients with metastatic melanoma who received ipilimumab in phase II clinical trialsAnn Oncol201324821742180Accessed September 1, 201423666915
  • AsciertoPSimeoneESileniVClinical experience with ipilimumab 3mg/kg: real-world efficacy and safety data from an expanded access programme cohortJ Transl Med201412111624885479
  • AckermanAKleinOMcDermottDFOutcomes of patients with metastatic melanoma treated with immunotherapy prior to or after BRAF inhibitorsCancer2014120111695170124577748
  • AsciertoPAMargolinKIpilimumab before BRAF inhibitor treatment may be more beneficial than vice versa for the majority of patients with advanced melanomaCancer2014120111617161924577788
  • AsciertoPASimeoneESileniVCSequential treatment with ipilimumab and BRAF inhibitors in patients with metastatic melanoma: data from the Italian cohort of the ipilimumab expanded access programCancer Invest201432414414924484235
  • KeldermanSHeemskerkBvan TinterenHLactate dehydrogenase as a selection criterion for ipilimumab treatment in metastatic melanomaCancer Immunol Immunother201463544945824609989
  • VecchioSSpagnoloFMerloDFThe treatment of melanoma brain metastases before the advent of targeted therapies: associations between therapeutic choice, clinical symptoms and outcome with survivalMelanoma Res20142416167 Available at: http://journals.lww.com/melanomaresearch/Fulltext/2014/02000/The_treatment_of_melanoma_brain_metastases_before.8.aspxAccessed September 1, 201424121190
  • QueiroloPSpagnoloFAsciertoPEfficacy and safety of ipilimumab in patients with advanced melanoma and brain metastasesJ Neurooncol2014118110911624532241
  • MargolinKErnstoffMSHamidOIpilimumab in patients with melanoma and brain metastases: an open-label, phase 2 trialLancet Oncol201213545946522456429
  • LongGVTrefzerUDaviesMADabrafenib in patients with Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the brain (BREAK-MB): a multicentre, open-label, phase 2 trialLancet Oncol201213111087109523051966
  • DummerRGoldingerSMTurtschiCPVemurafenib in patients with BRAFV600 mutation-positive melanoma with symptomatic brain metastases: final results of an open-label pilot studyEur J Cancer50361162124295639
  • RochetNMKottschadeLAMarkovicSNVemurafenib for melanoma metastases to the brainN Engl J Med2011365252439244122188003
  • QueiroloPMorabitoALaurentSAssociation of CTLA-4 polymorphisms with improved overall survival in melanoma patients treated with CTLA-4 Blockade: a pilot studyCancer Invest201331533634523641913
  • GajewskiTFMolecular profiling of melanoma and the evolution of patient-specific therapySemin Oncol201138223624221421113
  • MenziesAMHayduLEVisintinLDistinguishing clinicopathologic features of patients with V600E and V600K BRAF-mutant metastatic melanomaClin Cancer Res201218123242324922535154
  • AsciertoPAMinorDRibasAPhase II trial (BREAK-2) of the BRAF inhibitor dabrafenib (GSK2118436) in patients with metastatic melanomaJ Clin Oncol201331263205321123918947
  • KleinOClementsAMenziesAMO’TooleSKeffordRFLongGVBRAF inhibitor activity in V600R metastatic melanomaEur J Cancer20134951073107923237741
  • KimKBKeffordRPavlickACPhase II study of the MEK1/MEK2 inhibitor trametinib in patients with metastatic BRAF-mutant cutaneous melanoma previously treated with or without a BRAF inhibitorJ Clin Oncol201331448248923248257
  • Van AllenEMWagleNSuckerAThe genetic landscape of clinical resistance to RAF inhibition in metastatic melanomaCancer Discov2014419410924265153
  • JohannessenCMBoehmJSKimSYCOT drives resistance to RAF inhibition through MAP kinase pathway reactivationNature2010468732696897221107320
  • WilsonTRFridlyandJYanYWidespread potential for growth-factor-driven resistance to anticancer kinase inhibitorsNature2012487740850550922763448
  • StraussmanRMorikawaTSheeKTumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretionNature2012487740850050422763439
  • GibneyGTSmalleyKSMAn unholy alliance: cooperation between BRAF and NF1 in melanoma development and BRAF inhibitor resistanceCancer Discov20133326026323475878
  • WhittakerSRTheurillatJ-PVan AllenEA genome-scale RNA interference screen implicates NF1 loss in resistance to RAF inhibitionCancer Discov20133335036223288408
  • MaertensOJohnsonBHollsteinPElucidating distinct roles for NF1 in melanomagenesisCancer Discov20133333834923171796
  • NissanMHPratilasCAJonesAMLoss of NF1 in cutaneous melanoma is associated with RAS activation and MEK dependenceCancer Res20147482340235024576830
  • SmalleyKSLioniMDalla PalmaMIncreased cyclin D1 expression can mediate BRAF inhibitor resistance in BRAF V600E–mutated melanomasMol Cancer Ther2008792876288318790768
  • NathansonKLMartinA-MWubbenhorstBTumor genetic analyses of patients with metastatic melanoma treated with the BRAF inhibitor dabrafenib (GSK2118436)Clin Cancer Res201319174868487823833299
  • ParaisoKHTXiangYRebeccaVWPTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expressionCancer Res20117172750276021317224
  • XingFPersaudYPratilasCAConcurrent loss of the PTEN and RB1 tumor suppressors attenuates RAF dependence in melanomas harboring V600EBRAFOncogene201231444645721725359
  • TrunzerKPavlickACSchuchterLPharmacodynamic effects and mechanisms of resistance to vemurafenib in patients with metastatic melanomaJ Clin Oncol201331141767177423569304
  • NazarianRShiHWangQMelanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulationNature2010468732697397721107323
  • PoulikakosPIRosenNMutant BRAF melanomas – dependence and resistanceCancer Cell2011191111521251612
  • ShiHKongXRibasALoRSCombinatorial treatments that overcome PDGFR -driven resistance of melanoma cells to V600EB-RAF inhibitionCancer Res201171155067507421803746
  • SunCWangLHuangSReversible and adaptive resistance to BRAF(V600E) inhibition in melanomaNature2014508749411812224670642
  • GirottiMRPedersenMSanchez-LaordenBInhibiting EGF receptor or SRC family kinase signaling overcomes BRAF inhibitor resistance in melanomaCancer Discov20133215816723242808
  • RizosHMenziesAMPupoGMBRAF inhibitor resistance mechanisms in metastatic melanoma: spectrum and clinical impactClin Cancer Res20142071965197724463458
  • WagleNEmeryCBergerMFDissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profilingJ Clin Oncol201129223085309621383288
  • ShiHMoriceauGKongXPreexisting MEK1 exon 3 mutations in V600E/KBRAF melanomas do not confer resistance to BRAF inhibitorsCancer Discov20122541442422588879
  • EmeryCMVijayendranKGZipserMCMEK1 mutations confer resistance to MEK and B-RAF inhibitionProc Natl Acad Sci200910648204112041619915144
  • PoulikakosPIPersaudYJanakiramanMRAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E)Nature2011480737738739022113612
  • ShiHMoriceauGKongXMelanoma whole-exome sequencing identifies V600EB-RAF amplification-mediated acquired B-RAF inhibitor resistanceNat Commun2012372422395615
  • MontagutCSharmaSVShiodaTElevated CRAF as a potential mechanism of acquired resistance to BRAF inhibition in melanomaCancer Res200868124853486118559533
  • ShiHHugoWKongXAcquired resistance and clonal evolution in melanoma during BRAF inhibitor therapyCancer Discov201441809324265155
  • ShiHHongAKongXA novel AKT1 mutant amplifies an adaptive melanoma response to BRAF inhibitionCancer Discov201441697924265152
  • RomanoEPradervandSPaillussonAIdentification of multiple mechanisms of resistance to vemurafenib in a patient with BRAFV600E-mutated cutaneous melanoma successfully rechallenged after progressionClin Cancer Res201319205749575723948972
  • WilmottJSMenziesAMHayduLEBRAFV600E protein expression and outcome from BRAF inhibitor treatment in BRAFV600E metastatic melanomaBr J Cancer2013108492493123403819
  • GregerJGEastmanSDZhangVCombinations of BRAF, MEK, and PI3K/mTOR inhibitors overcome acquired resistance to the BRAF inhibitor GSK2118436 dabrafenib, mediated by NRAS or MEK mutationsMol Cancer Ther201211490992022389471
  • SmalleyKSMWeberJSUp close and personal: the challenges of precision medicine in melanomaJNCI J Natl Cancer Inst20141062djt443
  • RodicSMihalcioiuCSalehRRDetection methods of circulating tumor cells in cutaneous melanoma: a systematic reviewCrit Rev Oncol Hematol2014911749224530125
  • LuoXMitraDSullivanRJIsolation and molecular characterization of circulating melanoma cellsCell Rep20147364565324746818
  • HongDSVenceLFalchookGBRAF(V600) inhibitor GSK2118436 targeted inhibition of mutant BRAF in cancer patients does not impair overall immune competencyClin Cancer Res20121882326233522355009
  • Comin-AnduixBChodonTSazegarHThe oncogenic BRAF kinase inhibitor PLX4032/RG7204 does not affect the viability or function of human lymphocytes across a wide range of concentrationsClin Cancer Res201016246040604821169256
  • FrederickDTPirisACogdillAPBRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanomaClin Cancer Res20131951225123123307859
  • BoniACogdillAPDangPSelective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte functionCancer Res201070135213521920551059
  • LiuCPengWXuCBRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in miceClin Cancer Res201319239340323204132
  • WilmottJSLongGVHowleJRSelective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanomaClin Cancer Res20121851386139422156613
  • CooperZAJunejaVRSagePTResponse to BRAF inhibition in melanoma is enhanced when combined with immune checkpoint blockadeCancer Immunol Res20142764365424903021
  • CallahanMKMastersGPratilasCAParadoxical activation of T cells via augmented ERK signaling mediated by a RAF inhibitorCancer Immunol Res201421707924416731
  • RibasAHodiFSCallahanMKontoCWolchokJHepatotoxicity with combination of vemurafenib and ipilimumabN Engl J Med2013368141365136623550685
  • Das ThakurMStuartDDThe evolution of melanoma resistance reveals therapeutic opportunitiesCancer Res201373206106611024097822
  • Das ThakurMSalangsangFLandmanASModelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistanceNature2013494743625125523302800
  • Das ThakurMStuartDDMolecular pathways: response and resistance to BRAF and MEK inhibitors in BRAFV600E tumorsClin Cancer Res20142051074108024352648