ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Systemic treatment of metastatic melanoma with BRAF and other molecular alterations

Systemic treatment of metastatic melanoma with BRAF and other molecular alterations
Literature review current through: Jan 2024.
This topic last updated: Sep 13, 2023.

INTRODUCTION — In patients with metastatic melanoma, genetic sequencing has led to the identification of multiple molecular alterations, some of which are candidates for targeted drug therapy. The most common are mutations in the BRAF gene, which are identified in approximately 40 to 60 percent of patients with metastatic disease [1-4]. Additional molecular alterations include NRAS mutations and the less frequent TRK gene fusions, and KIT mutations, among others. (See "The molecular biology of melanoma".)

Systemic therapy is indicated for most patients with metastatic melanoma harboring such mutations. The choice of therapy is guided by multiple clinical factors including mutation status, patient comorbidities, performance status, and prior therapy in either the adjuvant or metastatic setting. The role of checkpoint inhibitor immunotherapy and targeted therapy in this population is evolving, and enrollment in clinical trials is encouraged, where available. (See "Overview of the management of advanced cutaneous melanoma".)

Available treatment options with clinical benefit in BRAF mutant metastatic melanoma include the following, either as single agents or in combination:

Combination BRAF (dabrafenib, encorafenib, vemurafenib) and MEK inhibitors (trametinib, binimetinib, cobimetinib)

Anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) antibodies (ipilimumab)

Programmed cell death 1 protein (PD-1) checkpoint inhibitors (nivolumab (table 1), pembrolizumab (table 2))

Programmed cell death ligand 1 (PD-L1) checkpoint inhibitors (atezolizumab) in combination with targeted therapy

Lymphocyte-activation gene 3 (LAG-3) checkpoint inhibitors in combination with PD-1 inhibitors (nivolumab-relatlimab)

The approach to systemic therapy in patients with metastatic melanoma containing a BRAF V600 mutation is reviewed here (algorithm 1).

The approach to checkpoint inhibitor immunotherapy in metastatic melanoma, adjuvant and neoadjuvant therapy for locally advanced cutaneous melanoma, and the surgical management of melanoma, is discussed separately.

(See "Systemic treatment of metastatic melanoma lacking a BRAF mutation".)

(See "Adjuvant and neoadjuvant therapy for cutaneous melanoma".)

(See "Surgical management of primary cutaneous melanoma or melanoma at other unusual sites".)

(See "Metastatic melanoma: Surgical management".)

ASSESSMENT OF ACTIONABLE MUTATIONS — An understanding of the mitogen-activated protein (MAP) kinase pathway (figure 1) has led to the development of important targeted therapeutic approaches in patients with melanoma, specifically the development of combined BRAF plus MEK inhibitor therapy. Prior to initiation of systemic therapy, patients with metastatic melanoma should have their tumors assessed for actionable mutations (ie, molecular alterations that are candidates for targeted drug therapy) as follows:

Choice of tumor sample — We typically perform genetic sequencing on tumor samples obtained from distant metastases, as these are the sites of disease that will be treated with systemic therapy. If tissue from metastatic disease is unavailable or unable to be obtained, genetic sequencing can alternatively be performed on archived, paraffin-embedded tissue from either the primary tumor or tumor-involved regional lymph nodes.

Approach to genetic assay — For patients with tissue available for genetic assays, our approach is to rapidly assess for a BRAF V600 mutation (including V600E, the most frequent mutation identified in patients with metastatic melanoma), as mutation status influences treatment decisions for systemic therapy. BRAF V600 mutations can be identified using a single gene polymerase chain reaction (PCR) assays that are available either institutionally or commercially, such as the Cobas 4800 BRAF V600 mutation test, among others [5,6].

For patients in whom a BRAF V600 mutation is not identified, we obtain next-generation DNA sequencing (NGS), which is the preferred approach to identify other potentially actionable molecular alterations. NGS platforms can be used to identify frequent driver mutations in melanoma as well as an extensive panel of other mutations that could possibly be targeted in the future [7,8]. This approach maximizes the chances of identifying actionable mutations. (See "Next-generation DNA sequencing (NGS): Principles and clinical applications".)

Mutations that should be assessed with NGS include the less common BRAF V600 mutations and non-V600 molecular variants; NRAS; KIT (frequently seen in patients with either an acral or mucosal primary site, or primary melanoma in an area of chronically sun exposed skin); and TRK gene fusions. NGS can also provide information on the level of tumor mutational burden, which influences response to checkpoint inhibitor immunotherapy but not the choice to treat with immunotherapy. (See 'BRAF V600 mutation variants' below and 'Other molecular alterations' below.)

BRAF V600 MUTANT DISEASE

BRAF V600 mutation variants — Activating mutations in BRAF are present in approximately 40 to 60 percent of metastatic melanomas, depending on the primary site on the skin [2-4]. The two most common BRAF mutations are V600E and V600K. Among all tumors harboring a BRAF mutation, the V600E mutation occurs in 80 to 90 percent of cases, and the V600K mutation occurs in approximately 15 percent of cases [9].

Less frequent BRAF mutations include V600R, V600M, V600D, and V600G, occurring in approximately 5 percent of cases. Patients with metastatic melanoma harboring different BRAF V600 mutations exhibit varying degrees of clinical response to targeted therapy with BRAF plus MEK inhibitors, with BRAF V600E tumors demonstrating higher response rates than those with other V600 mutations [10].

Previously untreated patients

Choice of initial therapy — In patients with systemic therapy-naive BRAF V600 mutant metastatic disease, we recommend combination checkpoint inhibitor immunotherapy with nivolumab plus ipilimumab (table 3) rather than combination targeted therapy with BRAF plus MEK inhibitors. In a phase III trial, initial therapy with combination immunotherapy (nivolumab plus ipilimumab) improved overall survival (OS) compared with initial targeted therapy (combination BRAF plus MEK inhibitors (algorithm 1)) [11]. (See 'Nivolumab plus ipilimumab (preferred)' below.)

Initial treatment with immunotherapy offers durable responses, long-term OS benefit, and greater treatment-free survival. By contrast, while targeted therapy may initially offer a rapid treatment response, the duration of response is more limited, and most patients with BRAF mutant disease ultimately experience disease progression after initial targeted therapy [12] and require subsequent therapy. Immunotherapy is also less effective when used after targeted therapy. However, targeted therapy is the preferred option for patients who are ineligible for or decline immunotherapy. (See 'Ineligible for immunotherapy (targeted therapy)' below.)

In a randomized phase III trial (DREAMseq; ECOG-ACRIN EA6134), 265 patients with treatment-naïve BRAF mutant melanoma were randomly assigned to receive either immunotherapy with nivolumab plus ipilimumab followed by maintenance nivolumab, or targeted therapy with dabrafenib plus trametinib [11]. Patients with disease progression were offered the alternative regimen. At median follow-up of 28 months, compared with the sequence of targeted therapy followed by immunotherapy, immunotherapy followed by targeted therapy improved OS (two-year OS 72 percent [95% CI 62-79 percent] versus 52 percent [95% CI 42-60 percent]; two-year OS benefit 20 percent [95% CI 3-38 percent]). The trial was stopped early for benefit.

This sequence also trended toward improved progression-free survival (PFS; median 11.8 months [95% CI 5.9-33.5] versus 8.5 months [95% CI 6.5-11.3]; two-year PFS 42 versus 19 percent) and median duration of response (not reached versus 13 months). Among the responders to initial immunotherapy, the proportion who remained in remission at a median follow-up of 28 months was 88 percent (37 of 42 patients); in contrast, the proportion remaining in remission among the responders to initial targeted therapy was 48 percent (18 of 37 patients). Grade ≥3 toxicity rates were similar for immunotherapy and targeted therapy (60 versus 52 percent, respectively).

Similar results were also seen in a separate randomized phase II trial (SECOMBIT) comparing varying sequencing approaches of immunotherapy (nivolumab plus ipilimumab) and targeted therapy (encorafenib plus binimetinib), although follow-up of this trial is ongoing. In this study, compared with initial targeted therapy followed by immunotherapy, there is a trend towards higher PFS and OS for those treated with initial immunotherapy followed by targeted therapy and those who received a "sandwich" approach (ie, targeted therapy for a defined period to initially reduce tumor burden, followed by immunotherapy until disease progression, followed by targeted therapy) [13].

Data also suggest that immunotherapy is less effective when used upon progression after targeted therapy, likely due to treatment resistance [11,13,14]. In the phase III trial DREAMseq, objective response rates (ORRs) were lower for patients who received immunotherapy following targeted therapy (30 percent), compared with those who received initial immunotherapy (46 percent). In contrast, ORR were similar for targeted therapy whether given as initial therapy (43 percent) or following disease progression on immunotherapy (48 percent) [11].

Defining immunotherapy eligibility — Patients eligible for checkpoint inhibitor immunotherapy typically meet the following criteria:

Eastern Cooperative Oncology Group (ECOG) performance status <2 (table 4) or Karnofsky performance status >70 (table 5).

Fitness to tolerate potential immunotherapy-related adverse events (irAEs (see "Toxicities associated with immune checkpoint inhibitors")).

No medical comorbidities that would make irAEs difficult to manage (eg, chronic obstructive pulmonary disease [COPD] with low pulmonary reserve or poorly controlled diabetes mellitus).

No active clinically significant autoimmune disease.

No immunosuppressive therapy or corticosteroids greater than the equivalent of prednisone 10 mg orally daily.

Eligible for immunotherapy

Nivolumab plus ipilimumab (preferred) — For patients with BRAF V600 mutant disease who are eligible for immunotherapy, we prefer initial treatment with the combination of nivolumab (a PD-1 inhibitor) and ipilimumab (a cytotoxic T lymphocyte-associated antigen 4 [CTLA-4] inhibitor (table 3)), over a single-agent PD-1 inhibitor, as this approach improved PFS and OS in a phase III trial (algorithm 1) [15,16]. Nivolumab plus ipilimumab is also the preferred treatment in patients with stage IV disease, regardless of BRAF V600 mutation status, who have undergone definitive treatment of their disease with either surgery or radiation therapy. These data are discussed separately. (See "Adjuvant and neoadjuvant therapy for cutaneous melanoma", section on 'Metastatic disease (stage IV)'.)

In a double-blind, placebo-controlled phase III trial (CheckMate 067) [15,16], 945 treatment-naïve patients with metastatic melanoma were randomly assigned to one of the following:

Combination nivolumab (1 mg/kg every three weeks for four doses) plus ipilimumab (3 mg/kg every three weeks for four doses), followed by nivolumab 3 mg/kg every two weeks

Nivolumab 3 mg/kg every two weeks

Ipilimumab 3 mg/kg every three weeks for four doses

In a subgroup analysis performed at follow-up of 6.5 years, the 301 patients with BRAF V600 mutant tumors appeared to derive greater benefit with the combination relative to nivolumab [16]. In these patients, compared with nivolumab alone, nivolumab plus ipilimumab improved 6.5-year PFS (37 versus 23 percent, hazard ratio [HR] 0.62, 95% CI 0.44-0.89) and 6.5-year OS (57 versus 43 percent, HR 0.68, 95% CI 0.46-1.02).

Nivolumab plus ipilimumab was also effective in patients with more aggressive disease (eg, elevated lactate dehydrogenase [LDH], brain metastases, or symptomatic systemic metastases). Further details on the efficacy of nivolumab plus ipilimumab in the entire study population and these patient subgroups are discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Nivolumab plus ipilimumab (preferred)'.)

Nivolumab-relatlimab — For patients with BRAF V600 mutant disease who are unlikely to tolerate the potential toxicities of nivolumab plus ipilimumab, the combination of nivolumab-relatlimab is an appropriate alternative. As examples, this approach may be appropriate for older adults who are eligible for immunotherapy but seek a more tolerable regimen with lesser risk of irAEs, those with significant comorbidities or poor performance status (eg, Karnofsky scale ≤70 (table 5)), or those with a history of autoimmune disease. (See "Toxicities associated with immune checkpoint inhibitors", section on 'Patients with vulnerabilities to immunotherapy toxicities'.)

A double-blind phase III trial (RELATIVITY-047) evaluating nivolumab-relatlimab included a subset of 275 patients with BRAF V600 mutant advanced unresectable or metastatic melanoma. In the overall population, there was an improvement in PFS with the addition of relatlimab to nivolumab, an effect that was also seen in the BRAF mutant population [17,18]. Nivolumab-relatlimab also has a more favorable toxicity profile compared with that reported for nivolumab plus ipilimumab in other studies [15,16,19,20]. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Nivolumab-relatlimab'.)

Alternative immunotherapy options — For treatment-naïve patients with BRAF V600 mutant disease eligible for immunotherapy but who are unable to tolerate the potential toxicities of combination immunotherapy (ie, nivolumab plus ipilimumab or nivolumab-relatlimab) we suggest single-agent PD-1 inhibitors rather than targeted therapy (algorithm 1). Options include pembrolizumab (table 2) [21,22] or nivolumab (table 1) [15]. Data are as follows:

Pembrolizumab In a phase III trial (KEYNOTE-006), 834 patients without prior exposure to immunotherapy were randomly assigned to pembrolizumab versus ipilimumab [21,23]. Among the subset of 163 treatment-naïve patients with BRAF V600 mutant disease, pembrolizumab improved ORRs compared with ipilimumab (47 versus 18 percent) [21]. At median follow-up of 85 months, pembrolizumab also improved OS compared with ipilimumab (median 79 versus 26 months; seven-year OS 50 versus 28 percent, HR 0.58, 95% CI 0.38-0.89) [23]. Additionally, there were no differences in efficacy parameters in any other patient subsets, including those with previous exposure to BRAF or MEK inhibitors. Data for the entire study population are discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Pembrolizumab'.)

Other studies have also demonstrated similar outcomes for pembrolizumab in patients with BRAF-mutant melanoma compared with those with BRAF wild-type disease and suggested its efficacy as initial therapy. In a post-hoc analysis of three randomized trials (KEYNOTE-001, KEYNOTE-002, and KEYNOTE-006) including 1558 patients with advanced melanoma, patients with BRAF wild-type and BRAF V600E/K-mutant melanoma had similar rates of four-year PFS (23 versus 20 percent) and OS (38 versus 35 percent), although there was a small statistically significant difference in ORRs (40 versus 34 percent, respectively) [22]. Of note, patients with BRAF V600E/K-mutant melanoma previously treated with targeted therapy had worsened outcomes with pembrolizumab compared with those who had not previously received targeted therapy (ORR 28 versus 44 percent; four-year PFS 15 versus 28 percent; and OS 27 versus 49 percent).

Nivolumab – The data for nivolumab (table 1) for patients with BRAF V600 mutant disease are discussed above. (See 'Nivolumab plus ipilimumab (preferred)' above.)

The following immunotherapy options are not standardly used in treatment-naïve patients with BRAF V600 mutant disease.

Atezolizumab – The programmed cell death ligand 1 (PD-L1) inhibitor atezolizumab has been evaluated in combination with the targeted agents vemurafenib plus cobimetinib in a phase III trial and received US Food and Drug Administration (FDA) approval. However, the control arm for this trial was cobimetinib and vemurafenib rather than a single-agent PD-1 inhibitor such as pembrolizumab or nivolumab or the combination of nivolumab plus ipilimumab. (See 'Is there a role for combined immunotherapy and targeted therapy?' below.)

Pembrolizumab plus ipilimumab – We do not offer the combination of pembrolizumab and ipilimumab in these patients, as this combination does not have regulatory approval. Data for this regimen are discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Investigational agents'.)

Ineligible for immunotherapy (targeted therapy) — For previously untreated patients with BRAF V600 mutations who decline or are ineligible for immunotherapy, we recommend targeted therapy with combination BRAF plus MEK inhibitors, rather than either inhibitor as a single agent (algorithm 1). Ineligible patients include those with significant comorbidities or poor performance status (eg, Karnofsky scale ≤70 (table 5)). Such combination therapy confers a survival benefit and is less toxic compared with single-agent targeted therapy. (See 'Choice of BRAF plus MEK inhibitor therapy' below and 'Toxicities of BRAF and MEK inhibitors' below.)

Choice of BRAF plus MEK inhibitor therapy — Three different combinations of BRAF inhibitors plus MEK inhibitors are available as initial therapy:

Dabrafenib plus trametinib (see 'Dabrafenib plus trametinib' below)

Encorafenib plus binimetinib (see 'Encorafenib plus binimetinib' below)

Vemurafenib plus cobimetinib (see 'Vemurafenib plus cobimetinib' below)

All combinations are reasonable options as they have not been directly compared in randomized trials, although all appear to have similar efficacy, including ORRs of up to 70 percent. The choice between these regimens is based on multiple factors including sites of metastatic disease, patient convenience, and potential toxicities. As examples:

For patients with CNS metastases, we offer dabrafenib plus trametinib, as this combination can cross the blood-brain barrier; it has also been evaluated in clinical trials including this patient population. These data are discussed separately. (See "Management of brain metastases in melanoma", section on 'BRAF and MEK inhibitors'.)

Encorafenib plus binimetinib may be preferred for patient convenience, as this combination can be taken with or without meals and can be stored at room temperature, as opposed to the combination of dabrafenib and trametinib, which requires administration around meals and refrigeration for trametinib.

Vemurafenib plus cobimetinib is less preferred because it is the combination with the most treatment-related toxicities (eg, skin rash, photosensitivity, diarrhea, and elevations in liver enzymes). (See 'Vemurafenib plus cobimetinib' below.)

Dabrafenib plus trametinib — Dabrafenib is a specific inhibitor of BRAF kinase, and trametinib is a potent, highly specific inhibitor of MEK1/MEK2. Dabrafenib and trametinib improved survival outcomes as single agents when initially compared with chemotherapy [24-26] and delayed treatment resistance while reducing toxicity as combination therapy when initially compared with single-agent BRAF inhibition [27,28].

Based on these prior data, two subsequent phase III trials demonstrated that the combination improved PFS and OS compared with single-agent BRAF inhibition with either dabrafenib (COMBI-d) [29-31] or vemurafenib (COMBI-v) [32,33]. Extended follow-up studies combining the data from these two trials demonstrated long-term survival benefit in approximately one-third of patients treated with the combination of dabrafenib and trametinib [34-36]. Responses lasting for over a year and favorable outcomes are seen among patients with a good performance status and limited disease burden.

Data for the phase III trials (COMBI-d and COMBI-v) are as follows:

Dabrafenib plus trametinib versus dabrafenib – In a placebo-controlled phase III trial (COMBI-d), 423 treatment-naïve patients with a BRAF V600E or V600K mutation were randomly assigned to either dabrafenib (150 mg twice per day) plus trametinib (2 mg once per day) or to dabrafenib alone. At minimum three-year follow-up, compared with single-agent dabrafenib, the combination improved both three-year PFS (22 versus 12 percent, HR 0.71, 95% CI 0.57-0.88) and three-year OS (44 versus 32 percent, HR 0.75, 95% CI 0.58-0.96) [31]. ORRs were higher with the combination (68 versus 55 percent), and complete response rates were similar between the two groups (18 versus 15 percent). Patients on combination therapy were more likely to remain on treatment due to prolonged periods of disease control (19 versus 3 percent).

Cutaneous toxicities of any grade were more frequent for single-agent dabrafenib versus the combination, including dry skin (14 versus 9 percent), pruritus (11 versus 7 percent), hyperkeratosis (33 versus 6 percent), hand-foot syndrome (27 versus 6 percent), alopecia (26 versus 5 percent), skin papilloma (18 versus 1 percent), and squamous cell carcinoma (9 versus 3 percent) [30].

By contrast, other noncutaneous toxicities were more frequent for the combination versus single-agent dabrafenib, included diarrhea (18 versus 9 percent), pyrexia (52 versus 25 percent), and chills (28 versus 14 percent). Treatment discontinuation was more common with the combination (11 versus 7 percent), primarily due to pyrexia and chills. (See 'Toxicities of BRAF and MEK inhibitors' below.)

Dabrafenib plus trametinib versus vemurafenib – In an open-label phase III trial (COMBI-v) trial, 704 patients with treatment-naïve metastatic melanoma with a BRAF V600 mutation were randomly assigned to either dabrafenib plus trametinib or vemurafenib [32,33]. At median follow-up of 11 months, compared with vemurafenib, the combination improved one-year OS (72 versus 65 percent, HR 0.69, 95% CI 0.53-0.89), PFS (median 11 versus 7 months, 95% CI 0.46-0.69), and ORRs (67 versus 53 percent) [32]. In extended follow-up available in abstract form only, the combination continued to improve both PFS and OS (three-year PFS 25 versus 11 percent; three-year OS 45 versus 32 percent) [33]. Over half (58 percent) of patients on combination therapy remained on treatment.

Of note, the incidence of cutaneous squamous cell carcinoma and keratoacanthoma was lower with the combination of dabrafenib plus trametinib compared with vemurafenib alone (1 versus 18 percent) [32]. (See 'Toxicities of BRAF and MEK inhibitors' below.)

Combined analysis of COMBI-d and COMBI-v – In a combined analysis of COMBI-d and COMBI-v, the combination of dabrafenib and trametinib demonstrated a median PFS and OS of approximately 11 and 26 months, respectively [36]. Estimated PFS and OS at five years were approximately 19 and 34 percent, respectively. Among the 19 percent with a complete response, estimated five-year OS was 71 percent.

Factors associated with a favorable outcome from combination dabrafenib and trametinib include good performance status, normal LDH, and <3 organs containing metastases. In this and another study, patients with normal LDH and <3 sites of metastatic disease had estimated five-year OS rates of 51 and 55 percent [35,36].

Based on these data, dabrafenib plus trametinib was approved by the US FDA for the treatment of patients with unresectable or metastatic melanoma containing a BRAF V600E or BRAF V600K mutation [37,38].

Encorafenib plus binimetinib — Encorafenib is a specific inhibitor of BRAF kinase. Binimetinib is a specific inhibitor of MEK1 and MEK2 that has demonstrated activity in initial phase II trials of patients with advanced melanoma harboring mutations in BRAF V600 and NRAS [39,40]. In a phase III trial, the combination of encorafenib plus binimetinib improved PFS and OS over single-agent BRAF inhibition [41-44].

Based on initial phase I and II trial data [45], a phase III trial (COLUMBUS) was performed in 577 patients with BRAF V600-mutated melanoma, comparing the combination of the BRAF inhibitor encorafenib plus the MEK inhibitor binimetinib versus either encorafenib alone or the single-agent BRAF inhibitor vemurafenib [41-44,46]. At a median follow-up of approximately 70 months, results were as follows [46]:

The combination improved both PFS and OS compared with vemurafenib (median PFS 15 versus 7 months, five-year PFS 23 versus 10 percent, HR 0.51, 95% CI 0.40-0.67; median OS 34 versus 17 months, five-year OS 35 versus 21 percent, HR 0.64, 95% CI 0.50-0.81).

The combination had a nonsignificant trend towards higher PFS and similar OS compared with encorafenib (median PFS 15 versus 10 months, five-year PFS 23 versus 19 percent, HR 0.79, 95% CI 0.61-1.02; median OS 34 versus 24 months, five-year OS 35 percent each, HR 0.93, 95% CI 0.72-1.19).

Among patients with low tumor burden (normal LDH levels and <3 organs involved), the combination demonstrated a five-year PFS and OS of 39 and 48 percent, respectively.

The ORR for the combination was 64 percent, which was higher than either encorafenib (52 percent) or vemurafenib (41 percent).

Additionally, encorafenib improved both PFS and OS compared with vemurafenib (median PFS 10 versus 7 months, HR 0.68, 95% CI 0.52-0.88; median OS 24 versus 17 months, HR 0.71, 95% CI 0.56-0.91).

Grade ≥3 toxicity rates for the combination were 70 percent, and included increased transaminases (8 percent), pyrexia (4 percent), vomiting, diarrhea, serous retinopathy (3 percent each), nausea, rash (2 percent each), and decreased ejection fraction (2 percent). In particular, any grade ocular toxicities for the combination included blurred vision (16 percent), retinal detachment, subretinal fluid, and macular edema (7 percent each). (See 'Toxicities of BRAF and MEK inhibitors' below.)

Based on these data, the combination of encorafenib plus binimetinib was approved by the US FDA for the treatment of patients with unresectable or metastatic melanoma containing a BRAF V600E or BRAF V600K mutation.

Vemurafenib plus cobimetinib — Vemurafenib is a potent inhibitor of the BRAF kinase that improved survival when initially compared with chemotherapy [12,47,48]. Cobimetinib is a potent, specific inhibitor of MEK1 and MEK2. In a phase III trial, the combination of vemurafenib plus cobimetinib improved PFS and OS over single-agent BRAF inhibition.

Based on initial phase I data from the BRIM-7 study [49,50], the combination of vemurafenib plus cobimetinib was evaluated in a placebo-controlled phase III trial (coBRIM) [51,52]. In this study, 495 patients with previously untreated advanced melanoma with a BRAF V600 mutation were randomly assigned to vemurafenib (960 mg twice per day) plus cobimetinib (60 mg once per day on days 1 to 21 of each 28-day cycle) or vemurafenib alone.

At median follow-up of 14 months, compared with vemurafenib alone, the combination improved both OS (median 22 versus 17 months, HR 0.7, 95% CI 0.55-0.9) and PFS (median 12 versus 7 months, HR 0.58, 95% CI 0.46-0.72) [52]. Objective (70 versus 50 percent) and complete response rates (16 versus 11 percent) were also higher with the combination.

Grade ≥3 toxicities occurring more frequently with the combination included diarrhea (7 versus 1 percent), photosensitivity (3 versus 0 percent), and elevations in alanine aminotransferase (11 versus 6 percent), gamma-glutamyltransferase (15 versus 10 percent), aspartate aminotransferase (9 versus 2 percent), and blood creatine phosphokinase (12 versus <1 percent) [52]. Serious adverse events reported with the combination included pyrexia and dehydration (2 percent each). Cutaneous toxicities of any grade were higher with single-agent vemurafenib compared with the combination, including cutaneous squamous cell carcinoma (13 versus 4 percent) and keratoacanthoma (9 versus 2 percent). (See 'Toxicities of BRAF and MEK inhibitors' below.)

Based on these data, cobimetinib is approved by the US FDA for use in combination with vemurafenib for patients with metastatic melanoma and a V600 mutation in BRAF.

Dosing considerations and toxicities of BRAF plus MEK inhibitors

Continuous versus intermittent therapy — For patients receiving targeted treatment with combined BRAF plus MEK inhibitors, we suggest continuous rather than intermittent therapy. Continuous therapy (ie, administration of targeted therapy without interruption until disease progression or unacceptable toxicity) improved PFS over intermittent therapy in a randomized phase II trial [53]. These data also demonstrate that intermittent therapy does not delay the development of disease resistance.

In a phase II trial (SWOG 1320), 206 patients with unresectable or metastatic BRAF V600 mutant melanoma treated with dabrafenib plus trametinib were randomly assigned to continuous or intermittent dosing (five weeks on, three weeks off) [53]. Compared with intermittent therapy, continuous therapy improved PFS (median 9 versus 5.5 months) . OS and treatment-related toxicities were similar between the two groups.

Toxicities of BRAF and MEK inhibitors — Specific treatment-related toxicities associated with BRAF and/or MEK inhibitors are as follows:

Dermatologic toxicities

Squamous cell carcinomas – Squamous cell carcinomas (SCCs), including keratoacanthomas (KAs), are common with BRAF and MEK inhibitors when administered as single agents. They occur in 19 to 26 percent of cases within weeks of initiating therapy [54]. However, the incidence of SCCs and KAs are significantly mitigated when these drugs are administered in combination.

Molecular studies indicate that the development of SCCs and KAs are due to a paradoxical activation of the mitogen-activated protein (MAP) kinase pathway that bypasses the inhibition of BRAF [55]. However, toxicity data from phase III trials evaluating combination BRAF plus MEK inhibition indicate a reduced incidence of skin toxicity (including skin cancers) compared with single-agent BRAF inhibition. These findings are presumably due to the addition of the MEK inhibitor, which blocks the paradoxical activation of the MAP kinase pathway [56]. (See 'Dabrafenib plus trametinib' above and 'Vemurafenib plus cobimetinib' above.)

Further details on SCCs and KAs in patients treated with BRAF and MEK inhibitors and their management are discussed separately. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'BRAF plus MEK inhibitors' and "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'Squamoproliferative lesions' and "Keratoacanthoma: Management and prognosis" and "Treatment and prognosis of low-risk cutaneous squamous cell carcinoma (cSCC)".)

Other dermatologic toxicities – Other nonmalignant dermatologic complications associated with single-agent BRAF inhibitors include rash, photosensitivity reactions, palmoplantar keratoderma, and delayed wound healing, among others. Such dermatologic toxicities are discussed in detail separately. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'BRAF inhibitors'.)

Acneiform rashes are associated with single-agent MEK inhibitors. Clinical manifestations and management are discussed separately. (See "Acneiform eruption secondary to epidermal growth factor receptor (EGFR) and MEK inhibitors".)

Cardiovascular toxicities

Cardiomyopathy – Cardiomyopathy with decreased cardiac ejection fraction has been identified in patients treated with MEK inhibitors as single agents (trametinib) and in combination with BRAF inhibitors (cobimetinib plus vemurafenib). Patients receiving these agents should undergo assessment of left ventricular ejection fraction (LVEF) prior to initiation of and during therapy. Further details are discussed separately. (See "Cardiotoxicity of cancer chemotherapy agents other than anthracyclines, HER2-targeted agents, and fluoropyrimidines", section on 'Cobimetinib, trametinib, and binimetinib' and "Tests to evaluate left ventricular systolic function".)

QTc prolongation – Prolongation of the QTc interval can occur with administration of the BRAF inhibitors vemurafenib and encorafenib. Vemurafenib is a cytochrome P450 3A4 (CYP3A4) substrate, and it should be used with caution in patients with congenital long QT syndrome and those who are receiving other drugs that prolong the QT interval (table 6) or inhibit CYP3A4 (table 7). Patients may be monitored with electrocardiogram (ECG) and electrolytes before treatment and after dose modification. Further details are discussed separately. (See "Cardiotoxicity of cancer chemotherapy agents other than anthracyclines, HER2-targeted agents, and fluoropyrimidines", section on 'Vemurafenib and encorafenib'.)

Constitutional toxicities – Pyrexia is relatively frequent with combination BRAF plus MEK inhibitor therapy. Other common constitutional toxicities include fatigue, weakness, and arthralgias (≥20 percent) [12,24,37,38,57].

For most patients with pyrexia, we hold the BRAF inhibitor and manage symptomatically with rest, fluids, cold compresses, acetaminophen, and occasionally low doses of corticosteroids [58,59]. Severe symptoms occur in approximately 4 percent of cases and require dose modification upon reinitiation of therapy. As pyrexia is most common with dabrafenib, some experts may switch patients on dabrafenib plus trametinib with persistent pyrexia (attributed to drug therapy) to one of the other BRAF plus MEK inhibitor combinations.

In phase III trials, the incidence of pyrexia was highest for the combination of dabrafenib plus trametinib (52 percent) [30], versus the combinations of vemurafenib plus cobimetinib (29 percent) and encorafenib plus binimetinib (18 percent) [43,52].

Gastrointestinal toxicities – Diarrhea is the most common gastrointestinal toxicity reported with the combination of BRAF and MEK inhibitors. Gastrointestinal perforation has rarely been reported (<1 percent), specifically for the combination of dabrafenib plus trametinib. Management of these toxicities are discussed separately. (See "Chemotherapy-associated diarrhea, constipation and intestinal perforation: pathogenesis, risk factors, and clinical presentation", section on 'MEK inhibitors' and "Chemotherapy-associated diarrhea, constipation and intestinal perforation: pathogenesis, risk factors, and clinical presentation", section on 'Trametinib'.)

Pulmonary toxicities – Pneumonitis, interstitial lung disease (ILD), and pulmonary embolism have been reported with both BRAF and MEK inhibitors, and these toxicities are discussed separately. (See "Pulmonary toxicity associated with antineoplastic therapy: Molecularly targeted agents", section on 'BRAF inhibitors' and "Pulmonary toxicity associated with antineoplastic therapy: Molecularly targeted agents", section on 'Trametinib' and "Treatment, prognosis, and follow-up of acute pulmonary embolism in adults".)

Hepatotoxicity – Hepatoxicity has been associated with both BRAF and MEK inhibitors, and all patients receiving these agents in combination should undergo baseline and periodic monitoring of liver function. Dose adjustment of these drugs for patients with underlying liver disease is discussed separately. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'MEK inhibitors' and "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'BRAF inhibitors'.)

Nephrotoxicity – Various renal toxicities have been reported with BRAF inhibitors as single agents and in combination with MEK inhibitors. For example, vemurafenib is associated with decreases in creatinine clearance, acute kidney injury, and Fanconi syndrome. The combination of encorafenib and binimetinib is also associated with renal impairment, hyponatremia, and rarely glomerulonephritis. Further details on these toxicities and their management in those with underlying renal impairment are discussed separately. (See "Nephrotoxicity of molecularly targeted agents and immunotherapy", section on 'BRAF inhibitors'.)

Neurologic toxicities – Neurologic toxicities reported with vemurafenib include headaches and peripheral facial palsy, which can occasionally be bilateral [57,60]. (See "Neurologic complications of cancer treatment with molecularly targeted and biologic agents", section on 'Vemurafenib'.)

Radiation sensitization and recall – Radiation sensitization and recall, in some cases severe, involving cutaneous and visceral organs have been reported in patients treated with radiation prior to, during, or subsequent to treatment with vemurafenib and dabrafenib [61-64]. Holding treatment with a BRAF inhibitor (with or without an MEK inhibitor) for one to three days before and one day after stereotactic radiosurgery appears to minimize the risk of significant toxicity. The same approach is used for both intracranial and extracranial systemic metastases. (See "Management of brain metastases in melanoma", section on 'Radiation sensitization with BRAF inhibitors'.)

Ocular toxicities – Ocular toxicity (including uveitis, conjunctivitis, dry eyes) has been reported with both vemurafenib and dabrafenib. (See "Ocular side effects of systemically administered chemotherapy", section on 'BRAF inhibitors'.)

Visual problems associated with MEK inhibitors are known as MEK inhibitor-associated retinopathy. Retinal vein occlusion is an uncommon but potentially severe side effect that occurs in less than 1 percent of cases. Patients receiving MEK inhibitors should undergo ophthalmologic exams regularly during treatment and in the event of visual disturbances. (See "Ocular side effects of systemically administered chemotherapy", section on 'Mitogen-activated protein kinase inhibitors'.)

Hematologic toxicity Dabrafenib is not recommended for patients with known glucose-6-phosphate dehydrogenase (G6PD) deficiency, as it contains a sulfonamide moiety and may result in hemolytic anemia in these patients [58].

RAS-mutant chronic myelomonocytic leukemia occurring with the initiation of single-agent vemurafenib therapy has been reported [65]. (See "Chronic myelomonocytic leukemia: Clinical features, evaluation, and diagnosis".)

Hemophagocytic lymphohistiocytosis (HLH) has been observed in patients treated with dabrafenib plus trametinib [66,67]. Dabrafenib plus trametinib should be held if the diagnosis of HLH is suspected and discontinued if HLH is confirmed [37,38]. The diagnosis and management of HLH are discussed separately. (See "Clinical features and diagnosis of hemophagocytic lymphohistiocytosis" and "Treatment and prognosis of hemophagocytic lymphohistiocytosis".)

Endocrinologic toxicity – The incidence of grade 3 hyperglycemia in those receiving BRAF and MEK inhibitors is approximately 2 to 6 percent, but this toxicity mainly impacts patients with diabetes mellitus or hyperglycemia [26,58]. Such patients should have regular monitoring of glucose levels while on therapy.

Is there a role for combined immunotherapy and targeted therapy? — In patients with previously untreated advanced BRAF V600 mutant melanoma, we do not combine checkpoint inhibitor immunotherapy with molecularly targeted therapy (using BRAF and MEK inhibitors). For most patients with BRAF mutation-positive metastatic melanoma, our approach is to use combination immunotherapy as initial therapy, rather than molecularly targeted therapies in combination with immunotherapy, because of the ability of immunotherapy to provide long-term treatment-free survival. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Nivolumab plus ipilimumab (preferred)'.)

The rationale for combining these treatments in patients with BRAF V600 mutant melanoma was to achieve both the high response rates of targeted therapy and the durable responses of immunotherapy [68]. Data from randomized phase II and phase III trials suggest that the addition of immunotherapy to targeted therapy improves PFS and duration of response [69-72], which led the US FDA to approve the triplet combination of atezolizumab and vemurafenib plus cobimetinib. However, the triplet combination spartalizumab and dabrafenib plus trametinib showed a similar but nonstatistically significant improvement in PFS [73]. Furthermore, OS and ORRs are not significantly improved, and toxicity is greater with this approach. These triplet combinations have also not been directly compared with the sequential use of initial immunotherapy followed by targeted therapy upon disease progression, which is the preferred treatment strategy for many patients. Therefore, despite regulatory approval of the atezolizumab and vemurafenib plus cobimetinib triplet regimen, we do not routinely use combined immunotherapy and targeted therapy in clinical practice.

Atezolizumab and vemurafenib plus cobimetinib — In a randomized, placebo-controlled phase III trial (IMspire150), 514 treatment-naïve patients with advanced, unresectable stage IIIC or stage IV BRAF V600 mutant melanoma were randomly assigned to the combination of the PD-L1 inhibitor atezolizumab and vemurafenib plus cobimetinib (immunotherapy plus targeted therapy) versus placebo and vemurafenib plus cobimetinib (targeted therapy alone) [69,72,74]. During the first cycle of therapy, all patients received vemurafenib and cobimetinib only, with either atezolizumab or placebo added during subsequent cycles.

At median follow-up of 29 months, the addition of atezolizumab to targeted therapy improved PFS based on investigator assessment (median PFS 15 versus 11 months, HR 0.79, 95% CI 0.64-0.97), although in the initial publication the statistical significance of these data was not confirmed on independent review [69,72]. OS was similar between the treatment arms (median OS 39 versus 26 months; two-year OS 62 versus 53 percent, HR 0.84, 95% CI 0.66-1.06). Median duration of response was longer for those receiving both immunotherapy and targeted therapy (21 versus 13 months). Objective (67 versus 65 percent) and complete (18 versus 19 percent) response rates were similar between the two groups.

The use of combination versus sequential immunotherapy and targeted therapy has not been directly compared in a randomized trial. However, based on indirect comparison of data from a separate randomized trial (DREAMseq) which compared initial therapy of metastatic melanoma with nivolumab plus ipilimumab followed by targeted therapy upon disease progression versus the opposite treatment sequence [11], two-year OS (72 percent) and median duration of response (not reached) were more favorable for the sequential use of nivolumab plus ipilimumab and targeted therapy compared with that of combined atezolizumab plus targeted therapy in IMspire150. Further details of the DREAMseq trial are discussed separately. (See 'Choice of initial therapy' above.)

Serious treatment-related adverse events were similar between the groups (48 versus 42 percent); however, this data may be confounded, as the toxicities seen in patients receiving combination immunotherapy and targeted therapy but treated with vemurafenib plus cobimetinib during the initial run-in period were combined with the toxicities seen in those receiving placebo with vemurafenib plus cobimetinib, leading to a potentially lower toxicity than would be expected for the combination therapy regimen. No new toxicity signals for the individual therapies were noted.

Based on these data, atezolizumab, in combination with cobimetinib and vemurafenib, is approved by the US FDA for patients with BRAF V600 mutation-positive melanoma [75].

The efficacy of atezolizumab plus cobimetinib and vemurafenib in patients with BRAF mutation-positive melanoma and brain metastases is discussed separately. (See "Management of brain metastases in melanoma", section on 'Is there a role for combined immunotherapy and targeted therapy for melanoma brain metastases?'.)

Other combinations

Spartalizumab and dabrafenib plus trametinib – The combination of the PD-1 inhibitor spartalizumab with dabrafenib and trametinib (targeted therapy) did not significantly improve PFS and increased toxicity in a placebo-controlled phase III trial (COMBI-i) [73,76]. Among 532 patients with advanced, unresectable BRAF V600 mutant melanoma, at median follow-up of 27 months, the addition of spartalizumab to targeted therapy numerically increased PFS, but these results did not achieve statistical significance (16 versus 12 months; HR 0.82, 95% CI 0.66-1.03) [73]. Although OS was not formally tested because the primary endpoint for the trial (PFS) was not significant, median OS was not reached (NR) for either treatment arm. ORRs were 69 and 64 percent, respectively. Median duration of response was NR and 21 months, respectively. The addition of spartalizumab to targeted therapy also increased grade ≥3 toxicity rates (55 versus 33 percent), which frequently led to dose adjustments and treatment discontinuation (12 versus 8 percent).

Pembrolizumab and dabrafenib plus trametinib – In a post-hoc analysis of a randomized placebo-controlled phase II trial (KEYNOTE-022) of 120 treatment-naïve patients with advanced BRAF V600 mutant melanoma, at a median follow-up of 37 months, the addition of the PD-1 inhibitor pembrolizumab to dabrafenib plus trametinib improved PFS (median 17 versus 11 months, HR 0.53, 0.34-0.83) and duration of response (median 25 versus 12 months) [70,71]. Two-year OS rates were higher for immunotherapy plus targeted therapy compared with targeted therapy alone, although the results were not statistically significant (63 versus 52 percent, HR 0.64, 95% CI 0.38-1.06), and ORRs were lower (63 versus 72 percent, respectively). Grade ≥3 treatment-related toxicity rates were higher among those receiving combined immunotherapy plus targeted therapy (58 versus 25 percent).

Prior adjuvant systemic therapy — Adjuvant systemic therapy is administered to patients at high risk for recurrence after initial definitive surgical resection. For such patients with BRAF V600 mutant disease, options for adjuvant therapy include checkpoint inhibitor immunotherapy or targeted therapy with combination BRAF plus MEK inhibition. (See "Adjuvant and neoadjuvant therapy for cutaneous melanoma".)

Although adjuvant therapy reduces the risk of recurrence, some patients may still relapse with metastatic disease. There are limited prospective data evaluating the optimal treatment in these patients, and enrollment in clinical trials is encouraged, where available.

The approach to systemic therapy in these patients is based on multiple clinical factors including:

Type of adjuvant therapy originally received

Tolerance of prior adjuvant therapy

Interval between completion of adjuvant therapy and disease progression

Patient performance status and comorbidities

Prior adjuvant immunotherapy

Eligible for immunotherapy — For patients with BRAF V600 mutant metastatic melanoma who previously received adjuvant immunotherapy with a single-agent PD-1 inhibitor (eg, nivolumab or pembrolizumab), relapse with metastatic disease, and are eligible for further immunotherapy, we suggest initial treatment with nivolumab plus ipilimumab rather than other systemic therapies, similar to those with BRAF wildtype disease. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Prior treatment with single-agent PD-1 inhibitors (including adjuvant therapy)'.)

However, in the absence of comparative data, initial targeted therapy with combination BRAF plus MEK inhibitors is a reasonable alternative. We do not retreat these patients with single-agent PD-1 inhibitors because such treatment is unlikely to be curative, given the demonstrated disease resistance. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Other approaches'.)

Combination immunotherapy with CTLA-4 and PD-1 inhibitors results in objective responses in approximately 30 percent of patients with PD-1 inhibitor refractory disease and may offer the opportunity for long-term treatment-free survival. Patients offered this approach must have tolerated prior adjuvant immunotherapy without significant irAEs. (See "Toxicities associated with immune checkpoint inhibitors".)

As an example, a phase II trial evaluated the efficacy of pembrolizumab plus ipilimumab in patients with metastatic disease resistant to PD-1 inhibitors [77]. Among the subset of patients with BRAF V600 mutant tumors, the ORR was 25 percent (5 of 20 evaluable patients). Further details of this trial in the entire study population and other supporting data are discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Other approaches'.)

For those receiving prior adjuvant therapy with a single-agent PD-1 inhibitor (eg, pembrolizumab (table 2) or nivolumab (table 1)), we do not retreat with a PD-1 inhibitor. Observational retrospective data, which included patients with BRAF mutant advanced or metastatic disease, demonstrated limited activity with this retreatment approach and suggest some form of treatment resistance [78]. While single-agent ipilimumab is also an option in these patients, it is less preferred due to toxicity profile and the availability of other effective treatments. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Ipilimumab'.)

Ineligible for immunotherapy — For those who decline or are ineligible for further immunotherapy, we offer targeted therapy with combination BRAF plus MEK inhibitors. Ineligible patients include those with significant comorbidities or poor performance status (eg, Karnofsky scale ≤70 (table 5)). (See 'Ineligible for immunotherapy (targeted therapy)' above.)

Prior adjuvant BRAF plus MEK inhibitors — For patients with BRAF V600 mutant disease previously treated with adjuvant targeted therapy (ie, combination BRAF plus MEK inhibitors), we typically administer checkpoint inhibitor immunotherapy. However, specific treatment options depend upon previous tolerance of targeted therapy and the interval between completion of adjuvant therapy and disease progression. For example, patients with longer intervals between completion of adjuvant targeted therapy and disease progression may be rechallenged with targeted therapy.

Relapsed disease ≤6 months – For patients who relapse within six months of completing adjuvant targeted therapy, we offer immunotherapy rather than retreatment with targeted therapy, given patients likely have disease resistance to the previously received targeted therapy [79,80]. While we prefer combination immunotherapy with nivolumab plus ipilimumab, single-agent PD-1 inhibitors (eg, pembrolizumab (table 2), nivolumab (table 1)) are also an option in these patients, who are treatment-naïve to immunotherapy. Patients ineligible for immunotherapy should be offered enrollment in clinical trials, were available. (See 'Investigational options' below.)

Relapsed disease >6 months – We offer immunotherapy to patients who relapse more than six months after completing adjuvant therapy and/or were intolerant of targeted therapy. Options include combination immunotherapy (eg, nivolumab plus ipilimumab) or single-agent immunotherapy with PD-1 inhibitors (eg, pembrolizumab (table 2), nivolumab (table 1)) or the CTLA-4 inhibitor ipilimumab.

Rechallenging with targeted therapy is an acceptable alternative to immunotherapy for select patients who relapse more than six months after completing adjuvant targeted therapy, if they tolerated it previously without significant toxicity. (See 'Choice of BRAF plus MEK inhibitor therapy' above.)

Subsequent therapy — Although the survival of patients with BRAF V600 mutant metastatic melanoma has dramatically improved with the use of targeted therapy and immunotherapy, some patients may eventually relapse. The choice of treatment regimen is influenced by prior therapy and associated toxicities, patient performance status and comorbidities, and tolerance of the proposed regimen. Patients with symptoms related to rapid disease progression may also require concurrent palliative, symptom-directed care.

Our general approach is to offer an alternative treatment strategy, depending upon the prior therapy (eg, targeted therapy for those previously treated with immunotherapy; immunotherapy for those previously treated with targeted therapy). However, the choice of subsequent therapy is evolving, and clinical trials are encouraged, where available. (See 'Investigational options' below.)

Prior immunotherapy — For patients with BRAF V600 mutant metastatic disease previously treated with immunotherapy who relapse, we propose the following approach:

Prior nivolumab plus ipilimumab – For patients previously treated with nivolumab plus ipilimumab, we offer targeted therapy with combination BRAF plus MEK inhibitors. (See 'Choice of BRAF plus MEK inhibitor therapy' above.)

For those who decline or are ineligible for targeted therapy, options include enrollment in clinical trials or subsequent therapy with nivolumab-relatlimab [81]. (See 'Investigational options' below and "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Prior treatment with nivolumab plus ipilimumab'.)

Prior nivolumab-relatlimab – For patients who previously received nivolumab-relatlimab, options include targeted therapy with combination BRAF plus MEK inhibitors, or nivolumab plus ipilimumab, although data are limited for the optimal approach. For most patients, we prefer targeted therapy, especially for those who relapse shortly (eg, within six months) after initiating nivolumab-relatlimab; this may indicate some underlying disease resistance to immunotherapy. Nivolumab plus ipilimumab is also a reasonable alternative, particularly for patients who relapse more than six months after discontinuing nivolumab-relatlimab. In patients with disease refractory to nivolumab-relatlimab, observational studies suggest limited efficacy for subsequent therapy with regimens involving ipilimumab [82], but further prospective studies are needed. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Prior treatment with nivolumab-relatlimab'.)

Prior single-agent immunotherapy – For patients who previously received single-agent immunotherapy with a PD-1 inhibitor, have no prior exposure to ipilimumab, and remain eligible for immunotherapy, we offer the combination of nivolumab plus ipilimumab, as this approach offers patients the potential for long-term treatment-free survival. In addition, in a randomized phase II trial (SWOG 1616), nivolumab plus ipilimumab improved PFS in patients who progressed on single-agent immunotherapy. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Treatment-free survival' and "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Prior treatment with single-agent PD-1 inhibitors (including adjuvant therapy)'.)

For patients who progress on single-agent ipilimumab, we offer single-agent PD-1 inhibitors such as nivolumab (table 1) or pembrolizumab (table 2); however, such patients are rare since initial therapy with ipilimumab is less preferred due to toxicity profile and the availability of other effective treatments. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Prior treatment with single-agent ipilimumab'.)

For all of these patients, targeted therapy with BRAF plus MEK inhibitors is also a reasonable alternative. As an example, in a post-hoc analysis of a phase III trial (KEYNOTE-006), among a subset of patients initially treated with single-agent pembrolizumab who received subsequent therapy with BRAF plus MEK inhibitors, ORRs were approximately 30 percent [83]. (See 'Choice of BRAF plus MEK inhibitor therapy' above.)

Ineligible for immunotherapy – For patients who decline or are ineligible for further immunotherapy, we offer combination targeted therapy with BRAF plus MEK inhibitors. Ineligible patients include those with significant comorbidities or poor performance status (eg, Karnofsky scale ≤70 (table 5)). (See 'Ineligible for immunotherapy (targeted therapy)' above.)

Prior BRAF plus MEK inhibitors — For patients with BRAF V600 mutant disease previously treated with combination BRAF plus MEK inhibitor who relapse, we prefer combination immunotherapy with nivolumab plus ipilimumab. Single-agent PD-1 inhibitors are an alternative option for those who decline or are anticipated to not tolerate the potential toxicities of nivolumab plus ipilimumab.

Patients ineligible for immunotherapy should be evaluated for clinical trials. (See 'Investigational options' below.)

Prior immunotherapy and BRAF plus MEK inhibitors — For patients with BRAF V600 mutant disease who progress on both immunotherapy and targeted therapies, we refer for clinical trials. (See 'Investigational options' below.)

Retreatment with targeted therapy using combination BRAF plus MEK inhibitors is also an option in these patients [84,85]. We reserve this approach for patients who received their last dose of targeted therapy at least three months prior to initiating retreatment and did not experience significant treatment-related toxicities.

In a phase II trial, 25 patients with a driver mutation in BRAF were treated with the combination of dabrafenib and trametinib [84]. All patients had progressed on both checkpoint inhibitor immunotherapy and BRAF inhibitor (with or without trametinib), and a minimum of 12 weeks had elapsed since receiving their last dose of targeted therapy. In this study, partial responses were seen in eight patients (32 percent), with a majority of patients previously treated with a combination of dabrafenib plus trametinib. Stable disease was seen in 10 patients (40 percent). The median PFS during retreatment was 4.9 months.

Prognosis of BRAF V600 mutant disease — The prognosis of patients with BRAF V600 mutant disease has dramatically improved, with up to two-thirds of patients experiencing long-term survival. Among patients treated with immunotherapy or targeted therapy with BRAF plus MEK inhibitors, five-year OS rates are between 34 and 60 percent [15,36,50]. By contrast, initial studies conducted prior to the availability of these treatments reported five-year OS rates of 10 percent or less [4,50].

As examples, in a combined analysis of two phase III trials in patients with BRAF V600 mutant disease treated with dabrafenib plus trametinib, five-year OS in the entire study population and those with a complete response to therapy was 34 and 71 percent, respectively [36]. Similar results were seen in long-term follow-up of a phase Ib trial of vemurafenib plus cobimetinib, with a five-year OS of approximately 40 percent [50].

Immunotherapy also improves survival in patients with BRAF V600 mutant disease. In a phase III trial of patients treated with combination immunotherapy (nivolumab plus ipilimumab), five-year OS was 60 percent in this patient population [15].

OTHER MOLECULAR ALTERATIONS — Molecular alterations other than BRAF V600 may be present in metastatic melanoma. Since these mutations are rare, enrollment in clinical trials is encouraged, where available. The approach to systemic therapy in these patients is discussed below.

BRAF non-V600 mutations — For patients with BRAF non-V600 mutations (L597, K601, or BRAF fusions), we offer initial therapy with checkpoint inhibitor immunotherapy, using a similar treatment approach to those with BRAF V600 mutations. For patients who are ineligible for immunotherapy, there are no established treatments, and patients should be enrolled on clinical trials, where available. Targeted therapy with BRAF plus MEK inhibitors have minimal activity in such patients. (See 'BRAF V600 mutant disease' above.)

Rare BRAF mutations other than those in the 600th codon (V600) are found in approximately 4 to 14 percent of patients with patients with primary cutaneous melanoma [10]. Examples of such mutations include BRAF L597, K601, and BRAF fusion genes [86]. (See 'Assessment of actionable mutations' above.)

Limited data in patients with these rare mutations suggest minimal activity for MEK inhibitors as single agents and in combination with BRAF inhibitors [10,87]. As an example, in an observational study of 103 patients with non-BRAF V600 mutations treated with either BRAF and/or MEK inhibitors, the incidence of BRAF L597 and K601 mutations was 15 and 11 percent, respectively [10]. Among the 38 patients with these two mutations, overall response rates to BRAF inhibition alone, MEK inhibition alone, and combination therapy was 0, 40, and 28 percent respective; median PFS was only two, four, and three months, respectively.

NRAS mutation — Patients with BRAF wild-type disease may harbor mutations in NRAS, a driver mutation found in the mitogen-activated protein (MAP) kinase pathway [88]. NRAS mutations are found in approximately 15 to 20 percent of patients with primary cutaneous melanoma [39]. (See 'Assessment of actionable mutations' above.)

Patients with NRAS mutant metastatic melanoma are responsive to initial treatment with immunotherapy-based regimens [88]. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Treatment-naive patients'.)

For those who progress on immunotherapy, we suggest enrollment in clinical trials, where available. Patients who are ineligible for or decline clinical trials may be offered subsequent therapy using a similar approach to those with BRAF wild-type metastatic melanoma [88], which is discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Subsequent therapy'.)

The off-label use of binimetinib, a MEK inhibitor, is also an alternative option which modestly improved progression-free survival (PFS) but not overall survival (OS) in a phase III trial (NEMO). This study was conducted in 402 patients with advanced NRAS mutation-positive melanoma, including a subset (21 percent) previously treated with immunotherapy [89]. Patients were randomly assigned to either binimetinib or dacarbazine. Compared with dacarbazine, binimetinib improved PFS (median 2.8 versus 1.5 months, hazard ratio [HR] 0.62, 95% CI 0.47-0.8) and objective response rates (ORRs; 15 versus 7 percent). However, OS was similar between the two treatment arms (median 11 versus 10 months). In the subset of 57 patients previously receiving immunotherapy, binimetinib improved PFS more impressively (median 5.5 versus 1.6 months). Objective responses were seen in 9 of 57 patients (16 percent).

TRK fusions — In the rare patients with metastatic melanoma whose tumors test positive for a tropomyosin receptor kinase (TRK) gene fusion and have no other actionable driver mutations, we offer subsequent-line therapy with TRK-gene fusion inhibitors (larotrectinib and entrectinib) after prior treatment with checkpoint inhibitor immunotherapy.

Neurotrophic tyrosine receptor kinase (NTRK) genes encode for TRK proteins. When an NTRK gene fuses with a separate, unrelated gene, it produces an altered TRK fusion protein. Such NTRK gene fusions occur rarely (<1 percent) in patients with primary cutaneous melanoma, whose tumors express TRK oncogenic fusion proteins.

TRK inhibitors such as larotrectinib [90] and entrectinib have clinical activity and manageable toxicity profiles in patients with metastatic melanoma, including those with central nervous system (CNS) metastases [91,92]. As an example, in a combined analysis of three phase I to II studies, 159 patients with various malignancies receiving previous systemic therapy were treated with larotrectinib [90]. Among the seven patients with metastatic melanoma, objective responses were seen in three patients (43 percent).

Further details on the diagnosis and management of patients with TRK gene fusions are discussed separately. (See "TRK fusion-positive cancers and TRK inhibitor therapy".)

KIT mutations (acral and mucosal melanoma) — For patients with metastatic acral or mucosal melanoma with an activating c-kit mutation who progress on or are ineligible for immunotherapy, we offer treatment with imatinib.

Mutations in c-kit are seen most commonly among patients with acral or mucosal melanomas (15 to 20 percent) and in a smaller percentage of melanomas arising in areas of chronic skin damage (2 to 3 percent) [93]. Patients with BRAF wild-type disease may harbor mutations in c-kit. (See 'Assessment of actionable mutations' above.)

KIT inhibitors have clinical activity specifically in patients with melanoma harboring activating mutations of the c-kit gene (eg, exon 11 [codon 596] or exon 13 [codon 642]). Tumors with these mutations are more sensitive to KIT inhibitors, and treatment responses are more durable [94,95]. Otherwise, KIT inhibitors have limited efficacy in unselected groups [96-98]. Further details on these studies in patients with mucosal melanoma are discussed separately. (See "Treatment of metastatic mucosal melanoma", section on 'KIT mutation'.)

Tumor mutational burden — In patients with metastatic melanoma, testing for tumor mutational burden (TMB) is not required to guide treatment approach, as most melanomas demonstrate clinical response to checkpoint inhibitor immunotherapy regardless of TMB status. Patients with cutaneous melanoma typically exhibit high levels of TMB due to ultraviolet (UV) radiation damage from sun exposure, which is associated with improved response to checkpoint inhibitor immunotherapy. However, responses to immunotherapy have also been demonstrated frequently in patients with melanoma expressing lower TMB and/or negative programmed cell death ligand 1 (PD-L1) expression.

The diagnosis and treatment of other cancers expressing high levels of TMB are discussed separately. (See "Tissue-agnostic cancer therapy: DNA mismatch repair deficiency, tumor mutational burden, and response to immune checkpoint blockade in solid tumors".)

INVESTIGATIONAL OPTIONS — Other systemic therapies have been evaluated in patients with metastatic melanoma, and their use remains investigational. Further prospective studies are necessary.

Pembrolizumab plus lenvatinib — The combination of pembrolizumab and lenvatinib is effective in patients who have progressed on immunotherapy with a programmed cell death 1 protein (PD-1)/programmed cell death ligand 1 (PD-L1) inhibitor alone or in combination with a cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) inhibitor. This is based on an open-label, single arm trial (LEAP-004) of 103 patients with immunotherapy-refractory, unresectable or metastatic melanoma, including approximately one-third with BRAF V600 mutant disease (37 percent) [99]. The results of this study are discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'Investigational agents'.)

Antiangiogenic therapy — The use of VEGFR inhibitors as single agents also remains an investigational treatment approach, as tumors that are resistant to immunotherapy may respond to these agents. VEGF inhibitors such as axitinib, bevacizumab, and other tyrosine kinase inhibitors (TKIs) of this pathway have demonstrated activity in clinical trials [100-109]. As an example, in a phase II trial of patients with metastatic melanoma (which included a subset of patients refractory to immunotherapy), axitinib demonstrated an ORR of 19 percent, median PFS of seven months, and one-year OS of 28 percent [105].

Adoptive cell therapy — Adoptive cell therapy (ACT) is a promising investigational therapy in patients with metastatic melanoma who have progressed on immunotherapy and/or targeted therapies. Various ACT approaches include chimeric antigen receptor (CAR) T-cell therapy, T-cell receptor (TCR)-transduced T-cells, and tumor infiltrating lymphocytes (TILs). Further details on the mechanisms of action for ACT are discussed separately. (See "Principles of cancer immunotherapy", section on 'Manipulating T cells'.)

Lifileucel (LN-144) is a form of TIL therapy, a T-cell product produced ex vivo from tumor specimens. A phase II trial (C-144-01) demonstrated the efficacy of this agent in a subset of 66 patients with immunotherapy-refractory melanoma (including 15 patients with BRAF V600 mutant disease with prior exposure to BRAF plus MEK inhibitor therapy) [110]. Further details on TIL therapy are discussed separately. (See "Systemic treatment of metastatic melanoma lacking a BRAF mutation", section on 'What is the role of tumor-infiltrating lymphocytes?'.)

SPECIAL POPULATIONS

Brain metastases — The efficacy of combination targeted therapy with BRAF plus MEK inhibitors and the role of these agents in the management of brain metastases in patients with melanoma are discussed separately. (See "Management of brain metastases in melanoma".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Melanoma screening, prevention, diagnosis, and management".)

SUMMARY AND RECOMMENDATIONS

Molecular alterations in metastatic melanoma – Genetic sequencing has led to the identification of multiple molecular alterations, some of which are candidates for targeted drug therapy in patients with metastatic melanoma. While the most common are mutations in the BRAF gene, additional molecular alterations include NRAS mutations, TRK gene fusions, and KIT mutations, among others. (See 'Introduction' above.)

Assessment of actionable mutations – Prior to initiation of systemic therapy, patients with metastatic melanoma should have their tumors assessed for actionable mutations. Our approach is to rapidly assess for a BRAF V600 mutation, as mutation status influences treatment decisions for systemic therapy. For patients in whom a BRAF V600 mutation is not identified, we obtain next-generation DNA sequencing (NGS) to identify other potentially actionable molecular alterations. (See 'Assessment of actionable mutations' above.)

Initial systemic therapy for previously untreated BRAF V600 mutant disease – For previously untreated patients with BRAF V600 mutant metastatic melanoma, our approach is as follows (algorithm 1) (see 'BRAF V600 mutant disease' above):

Eligible for immunotherapy For treatment-naïve patients, we recommend combination immunotherapy with nivolumab and ipilimumab (table 3) rather than targeted therapy with combination BRAF plus MEK inhibitors (Grade 1B), as this approach improves overall survival and offers a greater opportunity for long-term treatment-free survival. For those unlikely to tolerate nivolumab and ipilimumab (eg, older adults who are eligible for immunotherapy but seek a more tolerable regimen; patients with a history of autoimmune disease), the combination of nivolumab-relatlimab is an appropriate alternative. (See 'Previously untreated patients' above and 'Eligible for immunotherapy' above.)

For those who are anticipated to not tolerate the potential toxicities of combination immunotherapy (ie, nivolumab plus ipilimumab or nivolumab-relatlimab) but are candidates for immunotherapy, we suggest single agent PD-1 inhibitors such as nivolumab (table 1) or pembrolizumab (table 2) rather than targeted agents (Grade 2C). (See 'Alternative immunotherapy options' above.)

Ineligible for immunotherapy – For patients who decline or are ineligible for immunotherapy, we recommend targeted therapy with combination BRAF plus MEK inhibitors, rather than either inhibitor as a single agent (Grade 1B) (algorithm 1). Ineligible patients include those with multiple comorbidities or poor performance status (eg, Karnofsky scale ≤70 (table 5)). (See 'Ineligible for immunotherapy (targeted therapy)' above.)

Available options include dabrafenib plus trametinib, encorafenib plus binimetinib, and vemurafenib plus cobimetinib. All combinations are reasonable options as they have similar efficacy and have not been directly compared in randomized trials. The choice between these regimens is based on multiple factors including sites of metastatic disease, patient convenience, and potential toxicities. (See 'Choice of BRAF plus MEK inhibitor therapy' above and 'Toxicities of BRAF and MEK inhibitors' above.)

For patients receiving targeted therapy with combined BRAF plus MEK inhibitors, we suggest continuous rather than intermittent therapy (Grade 2C). (See 'Continuous versus intermittent therapy' above.)

Prior adjuvant systemic therapy – The approach to systemic therapy in patients with BRAF V600 mutant disease who were previously treated with adjuvant systemic therapy and relapse with metastatic disease is based on the type and tolerance of the adjuvant therapy originally received, interval between completion of adjuvant therapy and disease progression, and patient performance status and comorbidities. (See 'Prior adjuvant systemic therapy' above.)

For patients who previously received adjuvant systemic therapy (eg, either with a single-agent PD-1 inhibitor or combination BRAF plus MEK inhibitor therapy) and are eligible for further immunotherapy, we suggest initial treatment with nivolumab and ipilimumab rather than other systemic therapies (Grade 2C). (See 'Eligible for immunotherapy' above.)

For those who are ineligible for further immunotherapy, we offer targeted therapy with combination BRAF plus MEK inhibitors. (See 'Ineligible for immunotherapy' above.)

For those who received adjuvant targeted therapy with BRAF plus MEK inhibitors, while we prefer combination immunotherapy with nivolumab plus ipilimumab, single-agent PD-1 inhibitors (eg, pembrolizumab (table 2), nivolumab (table 1)) are also an option for these patients who did not receive immunotherapy in the adjuvant setting. (See 'Prior adjuvant BRAF plus MEK inhibitors' above.)

For select patients who relapse more than six months after completing adjuvant targeted therapy, rechallenge with targeted therapy is an acceptable alternative to immunotherapy.

Subsequent therapy – For patients with BRAF V600 mutant metastatic melanoma who progress on initial therapy, we typically offer an alternative treatment approach, depending upon prior therapy. Patients are encouraged to enroll in clinical trials, where available. (See 'Subsequent therapy' above and 'Investigational options' above.)

Prior nivolumab plus ipilimumab – For patients previously treated with nivolumab plus ipilimumab, we offer targeted therapy with combination BRAF plus MEK inhibitors. For those who decline or are ineligible for targeted therapy, options include enrollment in clinical trials or subsequent therapy with nivolumab-relatlimab. (See 'Prior immunotherapy' above.)

Prior nivolumab-relatlimab – For those who previously received nivolumab-relatlimab, options include targeted therapy with combination BRAF plus MEK inhibitors or nivolumab plus ipilimumab. For most patients, we prefer targeted therapy, especially for those who relapse shortly (eg, within six months) after initiating nivolumab-relatlimab. Nivolumab plus ipilimumab is also a reasonable alternative, particularly for patients who relapse more than six months after discontinuing nivolumab-relatlimab. (See 'Prior immunotherapy' above.)

Prior single-agent immunotherapy – For those who previously received single-agent immunotherapy with a PD-1 inhibitor, have no prior exposure to ipilimumab, and remain eligible for immunotherapy, we offer the combination of nivolumab plus ipilimumab. Targeted therapy with BRAF plus MEK inhibitors is also a reasonable alternative. (See 'Prior immunotherapy' above.)

Prior BRAF plus MEK inhibitors – For those previously treated with combination BRAF plus MEK inhibitor, we prefer combination immunotherapy with nivolumab plus ipilimumab. Single-agent PD-1 inhibitors are an alternative option for those who decline or are anticipated to not tolerate the potential toxicities of nivolumab plus ipilimumab. (See 'Prior BRAF plus MEK inhibitors' above.)

Other molecular alterations – Molecular alterations other than BRAF V600 may be present in metastatic melanoma, including BRAF non-V600 variants, NRAS, tropomyosin receptor kinase (TRK) fusions, KIT, and tumor mutational burden (TMB). (See 'Other molecular alterations' above.)

  1. Flaherty KT, Puzanov I, Kim KB, et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N Engl J Med 2010; 363:809.
  2. Wellbrock C, Hurlstone A. BRAF as therapeutic target in melanoma. Biochem Pharmacol 2010; 80:561.
  3. Smalley KS, Sondak VK. Melanoma--an unlikely poster child for personalized cancer therapy. N Engl J Med 2010; 363:876.
  4. Long GV, Menzies AM, Nagrial AM, et al. Prognostic and clinicopathologic associations of oncogenic BRAF in metastatic melanoma. J Clin Oncol 2011; 29:1239.
  5. KNOW NOW: BRAF status matters. https://www.knownowbraf.com/patient/ (Accessed on February 01, 2021).
  6. Cobas 4800 BRAF V600 Mutation Test. US Food and Drug Administration. https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?start_search=1&PMANumber=P110020&SupplementType=NONE (Accessed on February 01, 2021).
  7. Reiman A, Kikuchi H, Scocchia D, et al. Validation of an NGS mutation detection panel for melanoma. BMC Cancer 2017; 17:150.
  8. Chakravarty D, Johnson A, Sklar J, et al. Somatic Genomic Testing in Patients With Metastatic or Advanced Cancer: ASCO Provisional Clinical Opinion. J Clin Oncol 2022; 40:1231.
  9. Rubinstein JC, Sznol M, Pavlick AC, et al. Incidence of the V600K mutation among melanoma patients with BRAF mutations, and potential therapeutic response to the specific BRAF inhibitor PLX4032. J Transl Med 2010; 8:67.
  10. Menzer C, Menzies AM, Carlino MS, et al. Targeted Therapy in Advanced Melanoma With Rare BRAF Mutations. J Clin Oncol 2019; 37:3142.
  11. 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.
  12. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med 2011; 364:2507.
  13. Ascierto PA, Mandalà M, Ferrucci PF, et al. Sequencing of Ipilimumab Plus Nivolumab and Encorafenib Plus Binimetinib for Untreated BRAF-Mutated Metastatic Melanoma (SECOMBIT): A Randomized, Three-Arm, Open-Label Phase II Trial. J Clin Oncol 2023; 41:212.
  14. Lau PKH, Feran B, Smith L, et al. Melanoma brain metastases that progress on BRAF-MEK inhibitors demonstrate resistance to ipilimumab-nivolumab that is associated with the Innate PD-1 Resistance Signature (IPRES). J Immunother Cancer 2021; 9.
  15. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 2019; 381:1535.
  16. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Long-Term Outcomes With Nivolumab Plus Ipilimumab or Nivolumab Alone Versus Ipilimumab in Patients With Advanced Melanoma. J Clin Oncol 2022; 40:127.
  17. Tawbi HA, Schadendorf D, Lipson EJ, et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N Engl J Med 2022; 386:24.
  18. Long GV, Hodi FS, Lipson EJ, et al. Overall Survival and Response with Nivolumab and Relatlimab in Advanced Melanoma. NEJM Evid 2023; 2.
  19. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015; 373:23.
  20. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 2017; 377:1345.
  21. Robert C, Ribas A, Schachter J, et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol 2019; 20:1239.
  22. Puzanov I, Ribas A, Robert C, et al. Association of BRAF V600E/K Mutation Status and Prior BRAF/MEK Inhibition With Pembrolizumab Outcomes in Advanced Melanoma: Pooled Analysis of 3 Clinical Trials. JAMA Oncol 2020; 6:1256.
  23. Robert C, Carlino MS, McNeil C, et al. Seven-Year Follow-Up of the Phase III KEYNOTE-006 Study: Pembrolizumab Versus Ipilimumab in Advanced Melanoma. J Clin Oncol 2023; 41:3998.
  24. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012; 380:358.
  25. Hauschild A, Ascierto PA, Schadendorf D, et al. Long-term outcomes in patients with BRAF V600-mutant metastatic melanoma receiving dabrafenib monotherapy: Analysis from phase 2 and 3 clinical trials. Eur J Cancer 2020; 125:114.
  26. Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N Engl J Med 2012; 367:107.
  27. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N Engl J Med 2012; 367:1694.
  28. Flaherty K, Daud A, Weber JS, et al. Updated overall survival (OS) for BRF113220, a phase 1-2 study of dabrafenib (D) alone versus combined dabrafenib and trametinib (D+T) in pts with BRAF V600 mutation-positive (+) metastatic melanoma (MM). J Clin Oncol 2014; 32S: ASCO #9010.
  29. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med 2014; 371:1877.
  30. 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.
  31. Long GV, Flaherty KT, Stroyakovskiy D, et al. Dabrafenib plus trametinib versus dabrafenib monotherapy in patients with metastatic BRAF V600E/K-mutant melanoma: long-term survival and safety analysis of a phase 3 study. Ann Oncol 2017; 28:1631.
  32. 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.
  33. Robert C, Karaszewska B, Schacter J, et al. Three-year estimate of overall survival in COMBI-v, a randomized phase 3 study evaluating first-line dabrafenib (D) + trametinib (T) in patients (pts) with unresectable or metastatic BRAF V600E/K–mutant cutaneous melanoma. Ann Oncol 2016; 27S: ESMO #LBA40.
  34. Long GV, Grob JJ, Nathan P, et al. Factors predictive of response, disease progression, and overall survival after dabrafenib and trametinib combination treatment: a pooled analysis of individual patient data from randomised trials. Lancet Oncol 2016; 17:1743.
  35. Long GV, Eroglu Z, Infante J, et al. Long-Term Outcomes in Patients With BRAF V600-Mutant Metastatic Melanoma Who Received Dabrafenib Combined With Trametinib. J Clin Oncol 2018; 36:667.
  36. 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.
  37. United States (US) Food and Drug Administration (FDA) Prescribing Label for Dabrafenib https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/202806s027lbl.pdf (Accessed on September 06, 2023).
  38. Trametinib: United States Food and Drug Administration Prescribing Label https://www.accessdata.fda.gov/drugsatfda_docs/label/2023/204114s029lbl.pdf (Accessed on September 06, 2023).
  39. Ascierto PA, Schadendorf D, Berking C, et al. MEK162 for patients with advanced melanoma harbouring NRAS or Val600 BRAF mutations: a non-randomised, open-label phase 2 study. Lancet Oncol 2013; 14:249.
  40. Ascierto PA, Berking C, Agarwala SS, et al. Efficacy and safety of oral MEK162 in patients with locally advanced and unresectable or metastatic cutaneous melanoma harboring BRAFV600 or NRAS mutations. J Clin Oncol 2012; 30S: ASCO #8511.
  41. 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.
  42. Dummer R, Ascierto PA, Gogas HJ, et al. Overall survival in patients with BRAF-mutant melanoma receiving encorafenib plus binimetinib versus vemurafenib or encorafenib (COLUMBUS): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2018; 19:1315.
  43. Gogas HJ, Flaherty KT, Dummer R, et al. Adverse events associated with encorafenib plus binimetinib in the COLUMBUS study: incidence, course and management. Eur J Cancer 2019; 119:97.
  44. Ascierto PA, Dummer R, Gogas HJ, et al. Update on tolerability and overall survival in COLUMBUS: landmark analysis of a randomised phase 3 trial of encorafenib plus binimetinib vs vemurafenib or encorafenib in patients with BRAF V600-mutant melanoma. Eur J Cancer 2020; 126:33.
  45. Sullivan RJ, Weber J, Patel S, et al. A Phase Ib/II Study of the BRAF Inhibitor Encorafenib Plus the MEK Inhibitor Binimetinib in Patients with BRAFV600E/K -mutant Solid Tumors. Clin Cancer Res 2020; 26:5102.
  46. Dummer R, Flaherty KT, Robert C, et al. COLUMBUS 5-Year Update: A Randomized, Open-Label, Phase III Trial of Encorafenib Plus Binimetinib Versus Vemurafenib or Encorafenib in Patients With BRAF V600-Mutant Melanoma. J Clin Oncol 2022; 40:4178.
  47. McArthur GA, Chapman PB, Robert C, et al. Safety and efficacy of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation-positive melanoma (BRIM-3): extended follow-up of a phase 3, randomised, open-label study. Lancet Oncol 2014; 15:323.
  48. Chapman PB, Robert C, Larkin J, et al. Vemurafenib in patients with BRAFV600 mutation-positive metastatic melanoma: final overall survival results of the randomized BRIM-3 study. Ann Oncol 2017; 28:2581.
  49. Ribas A, Gonzalez R, Pavlick 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.
  50. Ribas A, Daud A, Pavlick AC, et al. Extended 5-Year Follow-up Results of a Phase Ib Study (BRIM7) of Vemurafenib and Cobimetinib in BRAF-Mutant Melanoma. Clin Cancer Res 2020; 26:46.
  51. Larkin J, Ascierto PA, Dréno B, et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N Engl J Med 2014; 371:1867.
  52. Ascierto PA, McArthur GA, Dréno B, et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol 2016; 17:1248.
  53. Algazi AP, Othus M, Daud AI, et al. Continuous versus intermittent BRAF and MEK inhibition in patients with BRAF-mutated melanoma: a randomized phase 2 trial. Nat Med 2020; 26:1564.
  54. Lacouture ME, Duvic M, Hauschild A, et al. Analysis of dermatologic events in vemurafenib-treated patients with melanoma. Oncologist 2013; 18:314.
  55. Su F, Viros A, Milagre C, et al. RAS mutations in cutaneous squamous-cell carcinomas in patients treated with BRAF inhibitors. N Engl J Med 2012; 366:207.
  56. Carlos G, Anforth R, Clements A, et al. Cutaneous Toxic Effects of BRAF Inhibitors Alone and in Combination With MEK Inhibitors for Metastatic Melanoma. JAMA Dermatol 2015; 151:1103.
  57. Larkin J, Del Vecchio M, Ascierto PA, et al. Vemurafenib in patients with BRAF(V600) mutated metastatic melanoma: an open-label, multicentre, safety study. Lancet Oncol 2014; 15:436.
  58. Welsh SJ, Corrie PG. Management of BRAF and MEK inhibitor toxicities in patients with metastatic melanoma. Ther Adv Med Oncol 2015; 7:122.
  59. Atkinson V, Robert C, Grob JJ, et al. Improved pyrexia-related outcomes associated with an adapted pyrexia adverse event management algorithm in patients treated with adjuvant dabrafenib plus trametinib: Primary results of COMBI-APlus. Eur J Cancer 2022; 163:79.
  60. Klein O, Ribas A, Chmielowski B, et al. Facial palsy as a side effect of vemurafenib treatment in patients with metastatic melanoma. J Clin Oncol 2013; 31:e215.
  61. Anker CJ, Grossmann KF, Atkins MB, et al. Avoiding Severe Toxicity From Combined BRAF Inhibitor and Radiation Treatment: Consensus Guidelines from the Eastern Cooperative Oncology Group (ECOG). Int J Radiat Oncol Biol Phys 2016; 95:632.
  62. Merten R, Hecht M, Haderlein M, et al. Increased skin and mucosal toxicity in the combination of vemurafenib with radiation therapy. Strahlenther Onkol 2014; 190:1169.
  63. Boussemart L, Boivin C, Claveau J, et al. Vemurafenib and radiosensitization. JAMA Dermatol 2013; 149:855.
  64. Satzger I, Degen A, Asper H, et al. Serious skin toxicity with the combination of BRAF inhibitors and radiotherapy. J Clin Oncol 2013; 31:e220.
  65. Callahan MK, Rampal R, Harding JJ, et al. Progression of RAS-mutant leukemia during RAF inhibitor treatment. N Engl J Med 2012; 367:2316.
  66. Dudda M, Mann C, Heinz J, et al. Hemophagocytic lymphohistiocytosis of a melanoma patient under BRAF/MEK-inhibitor therapy following anti-PD1 inhibitor treatment: a case report and review to the literature. Melanoma Res 2021; 31:81.
  67. Samaran Q, Belakebi D, Theret S, et al. Hemophagocytic lymphohistiocytosis in advanced melanoma treated with dabrafenib and trametinib combination: two cases. Melanoma Res 2020; 30:519.
  68. Yu C, Liu X, Yang J, et al. Combination of Immunotherapy With Targeted Therapy: Theory and Practice in Metastatic Melanoma. Front Immunol 2019; 10:990.
  69. Gutzmer R, Stroyakovskiy D, Gogas H, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAFV600 mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020; 395:1835.
  70. Ascierto PA, Ferrucci PF, Fisher R, et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat Med 2019; 25:941.
  71. Ferrucci PF, Di Giacomo AM, Del Vecchio M, et al. KEYNOTE-022 part 3: a randomized, double-blind, phase 2 study of pembrolizumab, dabrafenib, and trametinib in BRAF-mutant melanoma. J Immunother Cancer 2020; 8.
  72. Ascierto PA, Stroyakovskiy D, Gogas H, et al. Overall survival with first-line atezolizumab in combination with vemurafenib and cobimetinib in BRAFV600 mutation-positive advanced melanoma (IMspire150): second interim analysis of a multicentre, randomised, phase 3 study. Lancet Oncol 2023; 24:33.
  73. Dummer R, Long GV, Robert C, et al. Randomized Phase III Trial Evaluating Spartalizumab Plus Dabrafenib and Trametinib for BRAF V600-Mutant Unresectable or Metastatic Melanoma. J Clin Oncol 2022; 40:1428.
  74. Robert C, Lewis KD, Gutzmer R, et al. Biomarkers of treatment benefit with atezolizumab plus vemurafenib plus cobimetinib in BRAFV600 mutation-positive melanoma. Ann Oncol 2022; 33:544.
  75. Atezolizumab injection. United States Prescribing Information. US National Library of Medicine. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761034s028lbl.pdf (Accessed on August 07, 2020).
  76. Dummer R, Lebbé C, Atkinson V, et al. Combined PD-1, BRAF and MEK inhibition in advanced BRAF-mutant melanoma: safety run-in and biomarker cohorts of COMBI-i. Nat Med 2020; 26:1557.
  77. Olson DJ, Eroglu Z, Brockstein B, et al. Pembrolizumab Plus Ipilimumab Following Anti-PD-1/L1 Failure in Melanoma. J Clin Oncol 2021; 39:2647.
  78. Betof Warner A, Palmer JS, Shoushtari AN, et al. Long-Term Outcomes and Responses to Retreatment in Patients With Melanoma Treated With PD-1 Blockade. J Clin Oncol 2020; 38:1655.
  79. Sosman JA, Pavlick AC, Schuchter LM, et al. Analysis of molecular mechanisms of response and resistance to vemurafenib (vem) in BRAFV600E melanoma. J Clin Oncol 2012; 30S: ASCO #8503.
  80. Griffin M, Scotto D, Josephs DH, et al. BRAF inhibitors: resistance and the promise of combination treatments for melanoma. Oncotarget 2017; 8:78174.
  81. Ascierto PA, Lipson EJ, Dummer R, et al. Nivolumab and Relatlimab in Patients With Advanced Melanoma That Had Progressed on Anti-Programmed Death-1/Programmed Death Ligand 1 Therapy: Results From the Phase I/IIa RELATIVITY-020 Trial. J Clin Oncol 2023; 41:2724.
  82. Menzies AM, Pires da Silva I, Trojaniello C, et al. CTLA-4 Blockade Resistance after Relatlimab and Nivolumab. N Engl J Med 2022; 386:1668.
  83. Long GV, Arance A, Mortier L, et al. Antitumor activity of ipilimumab or BRAF ± MEK inhibition after pembrolizumab treatment in patients with advanced melanoma: analysis from KEYNOTE-006. Ann Oncol 2022; 33:204.
  84. Schreuer M, Jansen Y, Planken S, et al. Combination of dabrafenib plus trametinib for BRAF and MEK inhibitor pretreated patients with advanced BRAF(V600)-mutant melanoma: an open-label, single arm, dual-centre, phase 2 clinical trial. Lancet Oncol 2017; 18:464.
  85. Cybulska-Stopa B, Rogala P, Czarnecka AM, et al. BRAF and MEK inhibitors rechallenge as effective treatment for patients with metastatic melanoma. Melanoma Res 2020; 30:465.
  86. Hutchinson KE, Lipson D, Stephens PJ, et al. BRAF fusions define a distinct molecular subset of melanomas with potential sensitivity to MEK inhibition. Clin Cancer Res 2013; 19:6696.
  87. Yao Z, Torres NM, Tao A, et al. BRAF Mutants Evade ERK-Dependent Feedback by Different Mechanisms that Determine Their Sensitivity to Pharmacologic Inhibition. Cancer Cell 2015; 28:370.
  88. Zaremba A, Mohr P, Gutzmer R, et al. Immune checkpoint inhibition in patients with NRAS mutated and NRAS wild type melanoma: a multicenter Dermatologic Cooperative Oncology Group study on 637 patients from the prospective skin cancer registry ADOREG. Eur J Cancer 2023; 188:140.
  89. Dummer R, Schadendorf D, Ascierto PA, et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2017; 18:435.
  90. Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020; 21:531.
  91. Drilon A, Siena S, Ou SI, et al. Safety and Antitumor Activity of the Multitargeted Pan-TRK, ROS1, and ALK Inhibitor Entrectinib: Combined Results from Two Phase I Trials (ALKA-372-001 and STARTRK-1). Cancer Discov 2017; 7:400.
  92. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020; 21:271.
  93. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol 2006; 24:4340.
  94. Hodi FS, Friedlander P, Corless CL, et al. Major response to imatinib mesylate in KIT-mutated melanoma. J Clin Oncol 2008; 26:2046.
  95. Lutzky J, Bauer J, Bastian BC. Dose-dependent, complete response to imatinib of a metastatic mucosal melanoma with a K642E KIT mutation. Pigment Cell Melanoma Res 2008; 21:492.
  96. Penel N, Delcambre C, Durando X, et al. O-Mel-Inib: a Cancéro-pôle Nord-Ouest multicenter phase II trial of high-dose imatinib mesylate in metastatic uveal melanoma. Invest New Drugs 2008; 26:561.
  97. Wyman K, Atkins MB, Prieto V, et al. Multicenter Phase II trial of high-dose imatinib mesylate in metastatic melanoma: significant toxicity with no clinical efficacy. Cancer 2006; 106:2005.
  98. Ugurel S, Hildenbrand R, Zimpfer A, et al. Lack of clinical efficacy of imatinib in metastatic melanoma. Br J Cancer 2005; 92:1398.
  99. Arance A, de la Cruz-Merino L, Petrella TM, et al. Phase II LEAP-004 Study of Lenvatinib Plus Pembrolizumab for Melanoma With Confirmed Progression on a Programmed Cell Death Protein-1 or Programmed Death Ligand 1 Inhibitor Given as Monotherapy or in Combination. J Clin Oncol 2023; 41:75.
  100. Flaherty KT, Brose M, Schuchter L. Phase I/II trial of BAY 43-9006, carboplatin and paclitaxel demonstrates preliminary amtotumor activity in the expansion cohort of patients with metastatic melanoma. Proc Am Soc Clin Oncol 2004; 23:708a.
  101. Amaravadi RK, Schuchter LM, McDermott DF, et al. Phase II Trial of Temozolomide and Sorafenib in Advanced Melanoma Patients with or without Brain Metastases. Clin Cancer Res 2009; 15:7711.
  102. Eisen T, Marais R, Affolter A, et al. Sorafenib and dacarbazine as first-line therapy for advanced melanoma: phase I and open-label phase II studies. Br J Cancer 2011; 105:353.
  103. Hauschild A, Agarwala SS, Trefzer U, et al. Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma. J Clin Oncol 2009; 27:2823.
  104. Flaherty KT, Lee SJ, Zhao F, et al. Phase III trial of carboplatin and paclitaxel with or without sorafenib in metastatic melanoma. J Clin Oncol 2013; 31:373.
  105. Fruehauf J, Lutzky J, McDermott D, et al. Multicenter, phase II study of axitinib, a selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, and 3, in patients with metastatic melanoma. Clin Cancer Res 2011; 17:7462.
  106. Varker KA, Biber JE, Kefauver C, et al. A randomized phase 2 trial of bevacizumab with or without daily low-dose interferon alfa-2b in metastatic malignant melanoma. Ann Surg Oncol 2007; 14:2367.
  107. González-Cao M, Viteri S, Díaz-Lagares A, et al. Preliminary results of the combination of bevacizumab and weekly Paclitaxel in advanced melanoma. Oncology 2008; 74:12.
  108. Perez DG, Suman VJ, Fitch TR, et al. Phase 2 trial of carboplatin, weekly paclitaxel, and biweekly bevacizumab in patients with unresectable stage IV melanoma: a North Central Cancer Treatment Group study, N047A. Cancer 2009; 115:119.
  109. Kim KB, Sosman JA, Fruehauf JP, et al. BEAM: a randomized phase II study evaluating the activity of bevacizumab in combination with carboplatin plus paclitaxel in patients with previously untreated advanced melanoma. J Clin Oncol 2012; 30:34.
  110. Sarnaik AA, Hamid O, Khushalani NI, et al. Lifileucel, a Tumor-Infiltrating Lymphocyte Therapy, in Metastatic Melanoma. J Clin Oncol 2021; 39:2656.
Topic 15408 Version 141.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟