<<

Hematology-Oncology Board Review Manual

Targeted Therapy and Immunotherapy in the Treatment of Metastatic Cutaneous

Saro Sarkisian, MD, Corey Rearick, BSc, and Diwakar Davar, MD, MSc

Introduction 1 [PD-1]). In this review, we discuss the role of immune therapy, , and combi- The incidence of cutaneous melanoma has nations of these in the treatment of metastatic increased over the past 2 decades, with SEER cutaneous melanoma. We limit the immuno- estimates indicating that the number of new therapy discussion to approved CTLA-4/PD-1 cases of melanoma diagnosed annually rose inhibitors and the targeted therapy discussion from 38,300 in 1996 to 76,000 in 2016.1 Among to approved BRAF/NRAS/MEK inhibitors and persons younger than 50 years, the incidence is do not discuss non-checkpoint immunothera- higher in females, and younger women (aged pies including cytokines (HD IL-2), vaccines, or 15–39 years) are especially vulnerable.2 Among adoptive T-cell approaches. Interested readers persons older than 50, melanoma incidence in are directed to other excellent works covering men is nearly twice that of women, in whom these important topics.26–29 are often thicker and often associ- ated with worse outcomes.1,2 Approximately 85% of melanomas are diagnosed at early stages DEVELOPMENT OF TARGETED AND NOVEL when surgery is curative, but the lifetime prob- IMMUNE THERAPIES ability of developing invasive disease is 3% in men and 2% in women. For many years the degree of ultraviolet (UV) Prior to the advent of effective immuno- light exposure was considered the sole major therapies and targeted therapies, melanoma risk factor for melanoma oncogenesis, even was often managed with , which though its mechanism was largely unknown.3 had dismal response rates and commensurately However, clinical observations regarding the poor outcomes. Advances in the understanding occurrence of melanoma on less exposed areas of the molecular etiopathogenesis and immune (trunk and limbs) in individuals with intermit- escape responses of cutaneous metastatic mela- tent sun exposure led to the proposition that noma have transformed therapeutic approach- melanomas that arose in younger patients with es. Specifically, improved understanding of the intermittent sun exposure were distinct from genetic mutations driving melanoma tumori- melanomas that arose in older patients in asso- genesis coupled with insights into mechanisms ciation with markers of chronic sun exposure— of tumor-mediated immune evasion resulted in the “divergent pathway” hypothesis.3 Critical to development of inhibitors of mitogen-activated this understanding were whole-exome sequenc- protein (MAPK; BRAF and MEK) along ing data from multiple groups, including The with inhibitors of negative regulatory immune Cancer Genome Atlas, that identified patterns checkpoints (cytotoxic T lymphocyte–associated of mutations in oncogenic drivers that were antigen 4 [CTLA-4] and programmed cell death- distinct in patients with and without chronically

26 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

sun-damaged (CSD) skin.4–7 It is now clear that often durable, with the vast majority of patients based on its association with CSD skin, mela- progressing after 12 to 15 months of therapy. noma can be subclassified into CSD or non- In parallel, work primarily done in murine CSD melanoma. CSD and non-CSD melanoma models of chronic viral infection uncovered the have distinct clinico-pathological characteristics role played by co-inhibitory or co-excitatory im- and are associated with different driver muta- mune checkpoints in mediating T-cell immune tions. CSD melanomas typically arise in older responses. These efforts clarified that tumor- patients on sun-exposed areas (head/neck, mediated immune suppression primarily occurs dorsal surfaces of distal extremities) and are through enhancement of inhibitory signals via associated with particular driver mutations the negative T-cell immune checkpoints CTLA- (BRAF non-V600E, NRAS, NF1, or KIT) and 4 or PD-1.14,15 Blockade of negative T-cell im- genetic signatures of UV-induced DNA damage mune checkpoints resulted in activation of the (G > T [UVA] or C > T [UVB]) transitions. Con- adaptive immune system, resulting in durable versely, non-CSD melanomas typically arise in anti-tumor responses as demonstrated in studies younger (< 55 years) patients on intermittently of the CTLA-4 inhibitor (CA184- sun-exposed areas (trunk, proximal extremi- 02016 and CA184-02417) and the PD-1 inhibitors ties) and are associated with BRAF V600E/K (CA209-003,18 CheckMate 037,19 and driver mutations and often lack genetic signa- CheckMate 06620) and (KEY- tures of UV mutagenesis. NOTE-00121 and KEYNOTE-00622). Compared Identification of driver mutations in compo- to the deep but short-lived responses seen with nents of the MAPK pathway, including BRAF targeted inhibitors, patients treated with and NRAS, facilitated the development of tar- CTLA-4 or PD-1 immune checkpoint blockade geted inhibitors. The BRAF inhibitors vemu- often developed durable responses that persist- rafenib and have been shown in ed even after completion of therapy. Combined pivotal phase 3 studies to significantly improve CTLA-4 and PD-1 blockade results in greater overall and progression-free survival in patients magnitude of response with proportionately with metastatic melanoma compared with che- increased toxicity.23–25 motherapy and garnered regulatory approval (, BRIM-3;8,9 dabrafenib, BREAK- 310). Concomitant MEK and BRAF inhibition Immunotherapy extends the duration of benefit by preventing downstream kinase activation in the MAPK CTLA-4 and PD-1 Immune Checkpoint inhibitors pathway. Notably, concomitant MEK inhibition The novel success of immunotherapy in re- alters the side-effect profile of BRAF inhibi- cent decades is largely attributable to improved tors, with reduced incidence of keratoacantho- understanding of adaptive immune physiology, mas and cutaneous squamous cell carcinomas specifically T-cell activation and regulation. T-cell that are attributable to on-target, off-tumor activation requires 2 independent signaling effects of BRAF inhibitors. Combined BRAF events: it is initiated upon recognition of the and MEK inhibition (vemurafenib/ antigen-MHC class II-receptor complex on and dabrafenib/) further improved antigen-presenting cells (APC), and requires a overall and progression-free survival compared secondary co-stimulatory interaction of CD80/ to single-agent BRAF inhibition in phase 3 stud- CD86 (B7.1/B7.2) on APCs and CD28 mol- ies (COMBI-d,11 COMBI-v,12 and coBRIM13). ecule on T-cells; without this second event, Although often deep, the responses seen with T-cells enter an anergic state.30–32 Upon successful the use of targeted kinase inhibitors are not signaling and co-stimulation, newly activated

www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 27 Metastatic Cutaneous Melanoma

T-cells upregulate CTLA-4, which can bind to tion, cytolytic function, and survival. Unlike B7 molecules with a nearly 100-fold greater CTLA-4, which primarily affects the priming affinity than CD28.33,34 Unlike CD28, CTLA-4 phase of naive T-cell activation, PD-1 chiefly engagement negatively regulates T-cell activa- regulates the effector phase of T-cell function. tion. The opposing signals produced by CD28 Furthermore, because PD-L1/PD-L2 expression and CTLA-4 are integrated by the T-cell to is limited to inflammatory microenvironments, determine eventual response to activation, and the effects of PD-1 are less generalized than those provide a means by which T-cell activation is of CTLA-4. homeostatically regulated to prevent exagger- ated physiologic immune responses.35 It was hy- Single Agent Activity of CTLA-4 and PD-1 pothesized that CTLA-4 blockade would permit Inhibitors T-cell activation, which is thwarted in the tumor Ipilimumab (MDX-010) is a human IgG1 microenvironment by tumor-mediated CTLA-4 monoclonal antibody shown to inhibit CTLA- engagement, thereby unleashing an anti-tumor 4.43 Early studies tested different formulations immune response.36 (transfectoma-derived and hybridoma-derived), PD-1 is a member of the CD28 and CTLA-4 doses, and schedules of ipilimumab primar- immunoglobulin super family and, similar to ily in patients with advanced refractory mela- CTLA-4, binds activated T-cells. PD-1 has 2 li- noma.44–46 Although responses were infrequent, gands on activated T-cells: PD-L1 and PD-L2.37 responding patients experienced durable remis- PD-L1 is constitutively expressed by a variety of sions at 1- and 2-year time points. Notably, in a immune and non-immune cells, particularly in foreshadowing of changes to response criteria inflammatory environments including tumor used to evaluate these agents, several treated microenvironments, in response to the release patients who initially had radiographically sta- of inflammatory cytokines such as interferon ble disease upon completion of therapy subse- (IFN)-γ.37,38 Conversely, PD-L2 is only minimally quently experienced a gradual decline in tumor expressed constitutively, although its expression burden. on immune and non-immune cells can be in- Ipilimumab was subsequently evaluated in 2 duced by similar cues from inflammatory micro- phase 3 trials. The first study (MDX010-020/ environments. PD-L1 and PD-L2 cross-compete CA184-020), which involved 676 HLA-A*0201– for binding to PD-1, with PD-L2 exhibiting positive patients with advanced melanoma, com- 2- to 6-fold greater relative affinity than PD-L1.39 pared ipilimumab 3 mg/kg every 3 weeks for PD-L1/PD-1 binding results in phosphorylation 4 doses either singly or in combination with of 2 tyrosinases in the intracellular portion of gp100 vaccine with a gp100-only control arm.16 PD-1, which contains immunoreceptor tyrosine- Ipilimumab administration resulted in objective based inhibitory motif (ITIM) and immunore- responses in 11% of patients and improved pro- ceptor tyrosine-based switch motif (ITSM). PD-1 gression-free and overall survival compared to ITSM subsequently recruits either of 2 SH2-do- gp100 alone. Of note, ipilimumab monotherapy main–containing protein tyrosine phosphatases: was superior to ipilimumab/gp100 combination, SHP-1 and SHP-2. SHP-2 signaling suppresses possibly related to timing of vaccine in relation PI3K/Akt activation, down-regulates Bcl-xL, and to ipilimumab. A confirmatory study (CA184- suppresses expression of multiple transcrip- 024) compared a higher dose of ipilimumab tion factors that mediate T-cell effector func- (10 mg/kg) in combination with to tion including GATA-3, Eomes, and T-bet.40–42 dacarbazine monotherapy in previously untreat- The net effect of PD-L1/PD-1 engagement is to ed melanoma and was positive.17 Given the lack of suppress T-cell proliferation, cytokine produc- augmented efficacy with the higher (10 mg/kg)

28 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

Table 1. Phase 3 Studies of CTLA-4 and PD-1 Inhibitors in Advanced Melanoma

Study/Patient Population/ mPFS mOS Primary Endpoint(s) Intervention(s) N ORR (95% CI) (95% CI) Grade 3/4 AEs, %

Ipilimumab CA184-020 3:1:1 randomization to: 676 Ipilimumab + Ipilimumab + Ipilimumab + Any: 16 (MDX010-020) • Ipilimumab 3 mg/kg + gp100: 5.7% gp100: 2.86 gp100: 10.0 Ipilimumab + gp100: 45.5 HLA-A*0201-positive, gp100 (DCR 20.1%) mo (2.8–3.0) mo (8.5–11.5) Ipilimumab: 45.8 Gp100: 47.0 previously treated • Ipilimumab 3 mg/kg + Ipilimumab: Ipilimumab: Ipilimumab: advanced melanoma gp100 placebo 10.9% (DCR 2.76 mo 10.1 mo Immune-related AE: (≥ 1 prior therapy) 28.5%) (2.7–2.8) (8.0–13.8) Ipilimumab + gp100: 10.2 • Gp100 + ipilimumab Ipilimumab: 14.5 Primary endpoint: OS placebo Gp100: 1.5% Gp100: 2.76 Gp100: 6.4 (DCR 11.0%) mo (2.7–2.8) mo (5.5–8.7) Gp100: 3.0 Ipilimumab dosed 3 mg/kg Cutaneous: every 3 wk for 4 doses HR 0.81 (95% HR 0.66 (95% CI 0.66–1.00) CI 0.51–0.87) Ipilimumab + gp100: 2.4 Gp100 vaccination con- Ipilimumab: 1.5 sisted of 2 injections 1 mg Gp100: 0 of gp100:280-288(288V) GI (diarrhea/colitis): peptide and 1 mg of Ipilimumab + gp100: 5.8 emulsion comprising Ipilimumab: 7.6 incomplete Freund's Gp100: 0.8 adjuvant (Montanide ISA-51) with gp100:209- Endocrine: 217(210M) peptide Ipilimumab + gp100: 1.1 Ipilimumab: 3.8 Reinduction with assigned Gp100: 0 treatment regimen was permitted in patients whose disease progressed

CA184-024 1:1 randomization to: 502 Ipilimumab + mPFS not Ipilimumab + Any: 17 (MDX010-024) • Ipilimumab 10 mg/kg + dacarbazine: reported dacarbazine: Ipilimumab + dacarbazine: 56.7 Previously untreated dacarbazine (850 mg/m2) 15.2% HR for PFS (vs 11.2 (9.4–13.6) Dacarbazine: 27.5 advanced melanoma • Dacarbazine (850 mg/m2) Dacarbazine: dacarbazine): Dacarbazine: Immune-related AE: Primary endpoint: OS + placebo 10.3% 0.76 (P = 0.006) 9.1 (7.8–10.5) Ipilimumab + dacarbazine: 41.7 Dacarbazine: 6.0 Induction (wk 0–22): HR 0.69 (95% CI 0.57–0.84) Cutaneous: Ipilimumab dosed 10 mg/kg Ipilimumab + dacarbazine: 3.2 every 3 wk for 4 doses Dacarbazine: 0.0 2 Dacarbazine 850 mg/m GI (diarrhea/colitis): every 3 weeks for 8 doses Ipilimumab + dacarbazine: 6.0 Maintenance (starting wk 24): Dacarbazine: 0.0 Ipilimumab dosed 10 mg/kg ALT/AST increases: every 12 wk until progres- Ipilimumab + dacarbazine: 38.1 sion Dacarbazine: 1.2 Dacarbazine placebo

continued on page 30 dose, ipilimumab received regulatory approval of 22% (26% treated; 20% untreated) reached in 2011 for the treatment of melanoma at the at year 3 regardless of prior therapy or ipilim- lower dose: 3 mg/kg administered every 3 weeks umab dose, underscoring the durability of long- for 4 doses (Table 1). Survival data was strikingly term survival in ipilimumab-treated patients.47 Ipi- similar to patterns observed in prior phase 2 stud- limumab administration resulted in an unusual ies, with survival curves plateauing after 2 years spectrum of toxicities including diarrhea, rash, at 23.5% to 28.5% of treated patients. Pooled hepatitis, and hypophysitis (termed immune-re- survival data from prospective and retrospective lated adverse events, or irAEs) in up to a third of studies of ipilimumab corroborate the plateau patients. www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 29 Metastatic Cutaneous Melanoma

Table 1. (continued)

Study/Patient Population/ mPFS mOS Primary Endpoint(s) Intervention(s) N ORR (95% CI) (95% CI) Grade 3/4 AEs, %

Nivolumab CheckMate 03719 2:1 randomization to: 405 Nivolumab: Nivolumab: mOS not Any: Advanced melanoma • Nivolumab 3 mg/kg every 31.7% 4.7 mo reported Nivolumab: 9.0 following progression 2 wk ICC: 10.6% (2.3–6.5) ICC: 32.0 on ipilimumab and BRAF • ICC: ICC: 4.2 mo Cutaneous: inhibitor (if BRAF V600 Dacarbazine 1000 mg/m2 (2.1–6.3) Nivolumab: 0.0 mutated) every 3 wk or carbopla- 6-mo PFS: ICC: 0.0 Primary endpoints: ORR tin AUC 6 with paclitaxel 48% (nivolum- GI (diarrhea/colitis): and mOS 175 mg/m2 ab) vs 34% Nivolumab: < 1.0 Continued until progres- (ICC) ICC: 2.0 sion or unacceptable side HR 0.82 Anemia: effects Nivolumab: 1.0 ICC: 5.0 Neutropenia Nivolumab: 0.0 ICC: 14.0 Thrombocytopenia Nivolumab: 0.0 ICC: 6.0 Other (increased lipase) Nivolumab: 1.0 ICC: 0.0

Checkmate 06620 1:1 randomization to: 418 Nivolumab: Nivolumab: 5.1 Nivolumab: All Advanced untreated • Nivolumab 3 mg/kg every 42.9% mo (3.5–10.8) not reached Nivolumab: 11.7 melanoma, BRAF wild 2 wk + placebo every 3 wk Dacarbazine: Dacarba- Dacarbazine: Dacarbazine: 17.6 type • Dacarbazine 1000 mg 14.4% zine: 2.2 mo 11.2 mo Cutaneous: Primary endpoint: OS every 3 wk + placebo (2.1–2.4) Significant Nivolumab: 0.5 every 2 wk HR 0.43 (95% benefit in OS Dacarbazine: 0.0 CI 0.34–0.56) in nivolumab GI (diarrhea/colitis): vs dacarbazine Nivolumab: 1.0 group (HR Dacarbazine: 0.5 for death 0.42 Neutropenia [99.79% CI Nivolumab: 0.0 0.25–0.73]) Dacarbazine: 4.9 Thrombocytopenia: Nivolumab: 0.0 Dacarbazine: 4.9

continued on page 31

Pembrolizumab and nivolumab are human- IgG4 moiety neither engages Fc receptors nor ized IgG4 monoclonal antibodies that target activates complement, avoiding cytotoxic effects the PD-1 receptor found on activated T cells, of the antibody upon binding to the T cells that B cells, and myeloid cells. Both pembrolizum- it is intended to activate. Both pembrolizumab ab and nivolumab are engineered similarly: and nivolumab bind PD-1 with high affinity and by immunizing transgenic mice with recombi- specificity, effectively inhibiting the interaction nant human PD-1-Fc protein and subsequently between PD-1 and ligands PD-L1 and PD-L2. screening murine splenic cells fused with my- Nivolumab was first studied in a phase 1 study eloma cells for hybridomas producing antibod- (CA209-003) of 296 patients with advanced can- ies reactive to PD-1-Fc.48,49 Unlike IgG1, the cers who received 1, 3, or 10 mg/kg administered

30 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

Table 1. (continued)

Study/Patient Population/ mPFS mOS Primary Endpoint(s) Intervention(s) N ORR (95% CI) (95% CI) Grade 3/4 AEs, %

Pembrolizumab KEYNOTE-00621 1:1:1 randomization to: 834 Pembrolizumab Pembrolizumab Pembrolizumab Any: Advanced melanoma • Pembrolizumab 10 mg/kg 2 wk: 33.7% 2 wk: 5.5 mo (every 2 wk Pembrolizumab 2 wk: 13.3 with 0 or 1 prior sys- every 2 wk Pembrolizumab (3.4–6.9) and 3 wk): Pembrolizumab 3 wk: 10.1 not reached Ipilimumab: 19.9 temic therapy for • Pembrolizumab 10 mg/kg 3 wk: 32.9% Pembrolizumab advanced disease every 3 wk Ipilimumab: 3 wk: 4.1 mo Ipilimumab: Cutaneous (rash): Primary endpoints: PFS 11.9% (2.9–6.9) 16.0 (unknown) Pembrolizumab 2 wk: 0 • Ipilimumab 3 mg/kg every Pembrolizumab 3 wk: 0 and OS 3 wk for 4 cycles Ipilimumab: 1-yr OS: 2.8 mo (2.8–2.9) Pembrolizumab Ipilimumab: 0.4 Therapy continued until HR for disease 2 wk and GI (diarrhea): disease progression, unac- 3 wk: 74.1% Pembrolizumab 2 wk: 14.7 ceptable AEs, withdrawal of progression 0.58 for both pem- and 68.4% Pembrolizumab 3 wk: 13.4 patient consent, or investi- Ipilimumab: 14.5 gator’s decision to discon- brolizumab and Ipilimumab: tinue therapy ipilimumab (95% 58.2% Fatigue: CI 0.46–0.72 Pembrolizumab Pembrolizumab 2 wk: 20.9 and 0.47–0.72), every 2 wk Pembrolizumab 3 wk: 19.1 respectively and 3 wk HR Ipilimumab: 15.2 for death vs ipilimumab 0.69 (95% CI 0.47–0.83) and 0.69 (95% CI 0.52– 0.9), respectively

Ipilimumab + Nivolumab CheckMate 06725 1:1:1 randomization to: 945 Nivolumab+ Nivolumab + Nivolumab + Any: Advanced melanoma • Nivolumab 1 mg/kg ipilimumab: ipilimumab: ipilimumab: Nivolumab + ipilimumab: 55.0 with no prior systemic every 3 wk + ipilimum- 57.6% 11.5 mo (8.9– not reached Nivolumab: 16.3 therapy for advanced ab 3 mg/kg every 3 wk Nivolumab: 16.7) Nivolumab: Ipilimumab: 27.3 disease for 4 doses, followed 43.7% Nivolumab: 6.9 not reached Cutaneous (rash/pruritus): Primary endpoints: PFS by nivolumab 3 mg/kg Ipilimumab: mo (4.3–9.5) Ipilimumab: Nivolumab + ipilimumab: 6.7 and OS every 2 wk for cycle 5 19% Ipilimumab: 2.9 20.0 (17.1–24.6) Nivolumab: 0.6 onwards Ipilimumab: 2.2 mo (2.8–3.4) HR for death • Nivolumab 3 mg/kg or disease GI (diarrhea): every 2 wk + placebo progression Nivolumab + ipilimumab: 9.3 • Ipilimumab 3 mg/kg 0.42 (99.5% CI Nivolumab: 2.2 every 3 wk for 4 doses 0.31–0.57) Ipilimumab: 6.1 + placebo GI (colitis): Treatment continued until Nivolumab + ipilimumab: 7.7 disease progression or Nivolumab: 0.6 development of unaccept- Ipilimumab: 8.7 able AEs GI (ALT/AST abnormalities): Nivolumab + ipilimumab: 14.4 Nivolumab: 2.3 Ipilimumab: 2.2 Fatigue: Nivolumab + ipilimumab: 4.2 Nivolumab: 1.3 Ipilimumab: 1.0

AE = adverse events; CI = confidence interval; GI = gastrointestinal; HR = hazard ratio; ICC = investigator’s choice chemotherapy; mPFS = median progression-free survival; mOS = median overall survival; ORR = objective response rate.

www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 31 Metastatic Cutaneous Melanoma

every 2 weeks.18 Histologies tested included mel- treated patients. Similar to nivolumab, most re- anoma, non–small-cell lung cancer (NSCLC), sponders experienced durable remissions. renal-cell cancer (RCC), castration-resistant Pembrolizumab was subsequently compared prostate cancer (CRPC), and colorectal can- to ipilimumab in untreated patients (KEY- cer (CRC). Responses were seen in melanoma NOTE-006) in which patients were randomly and RCC and unusually in NSCLC, including assigned to receive either ipilimumab or pem- in both squamous and non-squamous tumors. brolizumab at 1 of 2 doses: 10 mg/kg every 2 Objective responses were noted in 41% of the weeks and pembrolizumab 10 mg/kg every 107 melanoma patients treated at 3 mg/kg. 3 weeks.22 Response rates were greater with Survival was improved, with 1- and 2- year sur- pembrolizumab than ipilimumab, with com- vival rates of 62% and 43% at extended follow mensurately greater 1-year survival rates. Rates up.50 of treatment-related adverse events requiring Subsequently, nivolumab was compared to discontinuation of study drug were much lower chemotherapy in a pair of phase 3 studies with pembrolizumab than ipilimumab. This involving both previously untreated (Check- trial was instrumental in proving the supe- mate 066) and ipilimumab/BRAF inhibitor– rior profile of pembrolizumab over ipilimumab. refractory (CheckMate 037) patients.19,20 In both The US Food and Drug Administration (FDA) studies, nivolumab produced durable responses granted pembrolizumab accelerated approval in 32% to 34% of patients and improved sur- for second-line treatment of melanoma in 2014, vival over chemotherapy. Compared to ipilim- and updated this to include a first-line indica- umab, the incidence of irAEs was much lower tion in 2015 (Table 1). with nivolumab. The depth and magnitude of responses observed led to regulatory approval Efficacy of Combined CTLA-4 and PD-1 Inhibition for nivolumab in both indications (untreated Preclinical studies demonstrated that PD-1 and ipilimumab/BRAF inhibitor–treated mela- blockade was more effective than CTLA-4 block- noma) in 2014. Data from both studies are sum- ade and combination PD-1/CTLA-4 blockade marized in Table 1. was synergistic, with complete rejection of tu- Pembrolizumab was first evaluated in a phase mors in approximately half of the treated ani- 1 study of 30 patients with a variety of solid mals.14 This hypothesis was evaluated in a phase 1 organ malignancies in which no dose-limiting study that explored both concurrent and se- toxicities were observed and no defined maxi- quential combinations of ipilimumab and mal tolerated dose was reached.51 Per protocol, nivolumab along with increasing doses of both maximal administered dose was 10 mg/kg every agents in PD-1/CTLA-4–naïve advanced mela- 2 weeks. Following startling responses including noma.23 Responses were greater in the concur- 2 complete responses of long duration, pembro- rent arm (40%) than in the sequential arm lizumab was evaluated in a large phase 1 study (20%) across dose-levels with a small fraction of (KEYNOTE-001) of 1260 patients that evaluated patients treated in the concurrent arm experi- 3 doses (10 mg/kg every 2 weeks, 10 mg/kg encing a profound reduction (80%) in tumor every 3 weeks, and 2 mg/kg every 3 weeks) in burden. separate melanoma and NSCLC substudies.21 The superiority of ipilimumab/nivolumab Both ipilimumab-naïve and ipilimumab-treated combination to ipilimumab monotherapy was patients were enrolled in the melanoma sub- demonstrated in a randomized blinded phase 2 study. Objective responses were seen in 38% of study (CheckMate 069).24 Of the 4 different ipili- patients across all 3 dosing schedules and were mumab/nivolumab doses explored in the phase similar in both ipilimumab-naïve and ipilimumab- 1 study (3 mg/kg and 0.3 mg/kg, 3 mg/kg

32 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma and 1 mg/kg, 1 mg/kg and 3 mg/kg, 3 mg/kg central mediators in the MAPK pathway and and 3 mg/kg), ipilimumab 3 mg/kg and exert their effect primarily through MEK phos- nivolumab 1 mg/kg (followed by nivolumab phorylation and activation following dimeriza- 3 mg/kg) was compared to ipilimumab and tion (hetero- or homo-) of RAF molecules. As nivolumab-matched placebo. Responses were sig- a result, RAF is integral to multiple cellular nificantly greater with dual PD-1/CTLA-4 block- processes, including transcriptional regulation, ade compared to CTLA-4 blockade alone (59% cellular differentiation, and cell proliferation. versus 11%). Concurrently, a 3-arm randomized MAPK pathway activation is a common event phase 3 study compared the same dose of ipilim- in many cancers, primarily due to activating umab/nivolumab to ipilimumab and nivolumab mutations in BRAF or RAS. Alternatively, MAPK in previously untreated advanced melanoma pathway activation can occur in the absence of (CheckMate 067).25 Similar to CheckMate 069, activating mutations in BRAF or NRAS through CheckMate 067 demonstrated that ipilimumab/ down-regulation of MAPK pathway inhibitory nivolumab combination resulted in more pro- proteins (RAF-1 inhibitory protein or SPRY-2), found responses (58%) than either ipilimumab C-MET overexpression, or activating mutations (19%) or nivolumab (44%) alone. Toxicity, in non-BRAF/NRAS kinases including CRAF, primarily diarrhea, fatigue, pruritus, and rash, HRAS, and NRAS.53,54 was considerable in the combination arm (55% Somatic point mutations in BRAF are fre- grade 3/4 adverse events) and resulted in treat- quently observed (37%–50%) in malignant ment discontinuation in 30% of patients. The melanomas and at lower frequency in a range profound and durable responses observed led to of human cancers including NSCLC, colorectal accelerated approval of ipilimumab/nivolumab cancer, papillary thyroid cancer, , combination in 2015 (Table 1). glioma, and gastrointestinal stromal tumor.6,55,56 Efforts to improve the toxicity/benefit ratio BRAF mutations in melanoma typically occur of ipilimumab/nivolumab combination have within the activation segment of the kinase centered around studying lower doses and/or domain (exon 15). Between 80% and 90% of extended dosing schedules of ipilimumab, includ- activating mutations result in an amino acid ing ipilimumab 1 mg/kg every 6 or 12 weeks substitution of glutamate (E) for valine (V) at with nivolumab dosed at 3 mg/kg every 2 weeks position 600: V600E.57,58 V600E mutations are or 480 mg every 4 weeks. Promising data from true oncogenic drivers, resulting in increased a first-line study in NSCLC (CheckMate 012) kinase activity with demonstrable transforma- support the evaluation of nivolumab in combi- tional capacity in vitro. BRAF mutations are nation with lower-dosed ipilimumab (1 mg/kg usually mutually exclusive, with tumors typically every 6 or 12 weeks).52 This approach is being containing no other driver mutations in NRAS, tested against platinum doublet chemotherapy KIT, NF1, or other genes. in a confirmatory phase 3 study in NSCLC NRAS mutations are less common than BRAF (CheckMate 227). mutations, having a reported frequency of 13% to 25% in melanoma.4 NRAS mutations gener- ally occur within the P-loop region of the G do- Targeted Therapy main (exon 2), or less commonly in the switch II region of the G domain (exon 3). Most NRAS MAPK Kinase Pathway in Melanoma exon 2 mutations comprise amino acid substitu- Tumorigenesis tions at position 61 from glutamine (Q) to argi- The MAPK pathway mediates cellular re- nine (R; 35%), lysine (K; 34%) and less often sponses to growth signals. RAF kinases are to glutamate (E), leucine (L), or proline (P). www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 33 Metastatic Cutaneous Melanoma

Preclinical data suggest that NRAS mutations developed, the initial evaluation of MAPK paradoxically stimulate the MAPK pathway and pathway inhibitors in advanced human can- thus enhance tumor growth in vitro.59,60 cers began with CI-1040. Preclinical data sug- Several important phenotypic differences dis- gested that CI-1040 potently and selectively tinguish NRAS- from BRAF-mutated melanoma. inhibited both MEK1 and MEK2, but phase 1 NRAS-mutated tumors are typically associated and 2 human trial results were disappointing, with increasing age and CSD skin, while BRAF- likely because these trials were not selectively mutated tumors arise in younger patients in enriched for NRAS/BRAF–mutated tumors or non-CSD skin. A large population-based study cancers in which these oncogenic mutations suggested that NRAS-mutated melanomas were were most commonly detected, such as mela- associated with mitoses and lower tumor infil- noma.67,68 The subsequent evaluation of selu- trating lymphocytes (TIL) grade, and arose in metinib (AZD6244/ARRY-142886) in a phase 2 anatomic sites other than head/neck, while study was also negative. Although investigators BRAF-mutated tumors were associated with enrolled a presumably enriched population (cu- mitoses and superficial spreading histology.61 taneous melanoma), the incidence of NRAS/ Although the lower TIL grade seen with NRAS- BRAF–mutated tumors was not ascertained to mutated melanomas suggests a more immuno- determine this, but rather assumed, which led to suppressed microenvironment and argues for a discrepancy between the assumed (prestudy) poorer responses to immune therapies, clinical and observed (on-study) proportions of BRAF/ studies comparing responses to immunothera- NRAS mutations that was not accounted for in pies in various categories of driver mutations power calculations.69,70 Lessons learned from provide conflicting results for the prognostic these earlier misadventures informed the cur- role of NRAS mutations in relation to immune rent paradigm of targeted therapy development: checkpoint blockade and other immune thera- (1) identification of a highly specific and po- pies.62–64 tent inhibitor through high-throughput screen- NF1 represents the third known driver in ing; (2) establishment of maximum tolerated cutaneous melanoma, with mutations reported dose (MTD) and recommended phase 2 dose in 12% of cases.6,7 NF1 encodes neurofibromin, (RP2D) in unselected patients; (3) confirma- which has GTPase activity and regulates RAS tion of RP2D in selected tumor types enriched proteins; NF1 loss results in increased RAS.65 for target of interest; and (4) confirmatory study Unlike BRAF or NRAS, which are usually mutu- against standard comparator to seek regulatory ally exclusive, NF1 mutations in melanoma can approval. occur singly or in combination with either BRAF Vemurafenib and dabrafenib were evaluated or NRAS mutations. In these settings, NF1 muta- in this tiered fashion in phase 1 dose-finding tions are associated with RAS activation, MEK- studies comprising unselected patients, followed dependence, and resistance to RAF inhibition.66 by phase 2 studies in advanced BRAF V600E– mutated melanoma. Both were subsequently MAPK Pathway INHIBITION Singly and In evaluated in randomized phase 3 trials (vemu- Combination rafenib, BRIM-38; dabrafenib, BREAK-310) that Although multiple MEK 1/2 inhibitors compared them with dacarbazine (1000 mg/m2 (AS703026, AZD8330/ARRY-704, AZD6244, intravenously every 3 weeks) in the treatment CH5126766, CI-1040, GSK1120212, PD0325901, of advanced BRAF V600E–mutated melanoma. RDEA119, and XL518) and RAF inhibitors Response kinetics for both agents were remark- (ARQ 680, GDC-0879, GSK2118436, PLX4032, ably similar: single-agent BRAF inhibitors re- RAF265, , XL281/BMS-908662) were sulted in rapid (time to response 2–3 months),

34 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

Table 2. Phase 3 Studies of BRAF and MEK Inhibitors in Advanced Melanoma

Study/Patient Population/ mPFS mOS Primary Endpoint(s) Intervention(s) N ORR (95% CI) (95% CI) Grade 3/4 AEs, %

Vemurafenib BRIM38,9 1:1 randomization to: 675 Vemurafenib: 5.7% HR 0.39 (0.33– HR 0.75 (0.60– Cutaneous SCC: BRAF V600E–mutated, • Vemurafenib 960 mg Dacarbazine: 1.5% 0.47, P < 0.0001) 0.93, P = 0.0085) Vemurafenib: 12 previ­ously untreated twice daily Vemurafenib: Vemurafenib: Dacarbazine: 0 2 advanced melanoma­ • Dacarbazine (1000 mg/m ) 6.9 mo (6.2–7.0) 13.3 mo (11.9–14.9) Keratoacanthoma: every 3 wk Primary endpoints: PFS Dacarbazine: Dacarbazine: Vemurafenib: 6 and OS 1.6 mo (1.6–2.1) 10.0 mo (8.0–14.0) Dacarbazine: 0 HR 0.38 (95% CI HR 0.7 (95% CI Rash: 0.32–0.46) 0.57–0.87) Vemurafenib: 8 Dacarbazine: 0 Fatigue: Vemurafenib: 2 Dacarbazine: 2 Neutropenia: Vemurafenib: 0 Dacarbazine: 8

Dabrafenib BREAK-310 3:1 randomization to: 250 Dabrafenib: 50% Dabrafenib: Dabrafenib: 18.2 All: BRAF V600E–mutated, • Dabrafenib 150 mg Chemotherapy: 7% 4.3 mo mo (16.6–not Dabrafenib: 53 previ­ously untreated twice daily Chemotherapy: reached) Chemotherapy: 44 2 advanced melanoma­ • Dacarbazine 1000 mg/m 2.7 mo Chemotherapy: Hyperkeratosis: IV every 3 wk Primary endpoint: PFS HR 0.3 (95% CI 15.6 mo (12.7–not Dabrafenib: 37 0.18–0.51) reached) Skin papillomas: HR 0.77 (95% CI Dabrafenib: 24 0.52–1.13, not Pyrexia: statistically signifi- Dabrafenib: 28 cant)

continued on page 36

profound (approximately 50% objective re- inhibitors for the treatment of BRAF V600E sponses) reductions in tumor burden that lasted (and later V600K)–mutated melanoma (vemu- 6 to 7 months. Adverse events common to both rafenib in 2011; dabrafenib in 2013). Details agents included rash, fatigue, and arthralgia, al- regarding trial populations, study interventions, though clinically significant photosensitivity was efficacy, and adverse events are summarized in more common with vemurafenib and clinically Table 2. significant pyrexia was more common with dab- Responses to BRAF inhibitors are typically rafenib. Class-specific adverse events included profound but temporary. Mechanisms of ac- the development of cutaneous squamous-cell quired resistance are diverse and include reac- carcinomas and keratoacanthomas secondary tivation of MAPK pathway–dependent signaling to paradoxical activation of MAPK pathway sig- (RAS activation or increased RAF expression), naling either through activating mutations in and development of MAPK pathway–indepen- HRAS or mutations or amplifications in receptor dent signaling (COT overexpression; increased tyrosine kinases upstream of BRAF, resulting in PI3K or AKT signaling) that permits bypass of elevated levels of RAS–guanosine triphosphate inhibited BRAF signaling within the MAPK path- complexes.71 Results of these studies resulted way.72–76 These findings suggested that upfront in regulatory approval of single-agent BRAF inhibition of both MEK and mutant BRAF may

www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 35 Metastatic Cutaneous Melanoma

Table 2. (continued)

Study/Patient Population/ mPFS mOS Primary Endpoint(s) Intervention(s) N ORR (95% CI) (95% CI) Grade 3/4 AEs, %

Trametinib METRIC77 2:1 randomization to: 322 Trametinib: 22% Trametinib: At 6 mo: All: Metastatic melanoma • Trametinib 2 mg daily Chemotherapy: 8% 4.8 mo Trametinib: 81% Trametinib: 15 with V600E or V600K • Intravenous dacarba- Chemotherapy: Chemotherapy: Chemotherapy: 15 2 BRAF mutation zine 1000 mg/m or 1.5 mo 67% Rash: 2 paclitaxel 17 5mg/m HR for death 0.54 Trametinib: 8 Primary endpoint: PFS every 3 wk HR 0.45 (95% CI 0.33–0.63) (95% CI 0.32–0.91) Chemotherapy: 0 Patients in chemotherapy Hypertension: arm were permitted to Trametinib: 12 switch to trametinib arm Chemotherapy: 3 if they had disease pro- gression Fatigue: Trametinib: 4 Chemotherapy: 3 Vomiting: Trametinib: 1 Chemotherapy: 2

Binimetinib NEMO80 2:1 randomization to: 402 Binimetinib: Binimetinib: Binimetinib: All: NRAS-mutated melanoma • Binimetinib 45 mg 15% (11%–20%) 2.8 mo (2.8–3.6) 11 mo Binimetinib: 5 previ­ously untreated or twice daily Dacarbazine: Dacarbazine: Dacarbazine: Dacarbazine: 5 2 progressed on immuno- • Dacarbazine 1000 mg/m 7% (3%–13%) 1.5 mo (1.5–1.7) 10.1 mo Hypertension: IV every 3 wk therapy HR 0.62 (95% CI HR 1 (95% CI Binimetinib: 7 Primary endpoint: PFS 0.47–0.8) 0.75–1.33) Dacarbazine: 2 Anemia: Binimetinib: 2 Dacarbazine: 5 Neutropenia: Binimetinib: 1 Dacarbazine: 9

Vemurafenib + Cobimetinib coBRIM13 1:1 randomization to: 495 Cobimetinib + Cobimetinib + Cobimetinib + All: BRAF V600 E/K-positive • Cobimetinib (60 mg vemurafenib: vemurafenib: vemurafenib: 22.3 Cob + Vem: 62 dis­ease previously once daily, 21 days 70% 12.3 mo mo Vem + placebo: 58 untreated on/7 days off) + Vemurafenib + Vemurafenib + Vemurafenib + Diarrhea: Vemurafenib (960 mg Primary endpoint: PFS placebo: 50% placebo: 7.3 mo placebo: 17.4 mo Cob + Vem: 6 twice daily (15–19.8) Vem + placebo: 0 • Vemurafenib 960 mg HR 0.58 (95% CI twice daily + Placebo 0.46–0.72) HR 0.70 (95% CI Cutaneous SCC: 0.55–0.90) Cob + Vem: 2 Vem + placebo: 11 Keratoacanthoma: Cob + Vem: 1 Vem + placebo:18

continued on page 37 produce more durable responses than BRAF versus dabrafenib/placebo), COMBI-v12 (dab- inhibition alone. Three pivotal phase 3 stud- rafenib/trametinib versus vemurafenib), and ies established the superiority of combination coBRIM13 (vemurafenib/cobimetinib versus ve- BRAF and MEK inhibition over BRAF inhibi- murafenib/placebo). As expected, compared tion alone: COMBI-d11 (dabrafenib/trametinib to BRAF inhibitor monotherapy, combination

36 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

Table 2. (continued)

Study/Patient Population/ mPFS mOS Primary Endpoint(s) Intervention(s) N ORR (95% CI) (95% CI) Grade 3/4 AEs, %

Dabrafenib + Trametinib COMBI-d11 1:1 randomization to: 423 Dabrafenib + Dabrafenib + Dabrafenib + All: BRAF V600E or • Dabrafenib 150 mg twice trametinib: trametinib: Trametinib: Dab + Tram: 48 V600K–mutated, daily + Trametinib • 2 mg 69% 11 mo (8.0–13.9) 25.1 mo Dab + placebo: 50 once daily previously untreated Dabrafenib + Dabrafenib + Dabrafenib + Pyrexia: • Dabrafenib + placebo advanced melanoma placebo: 53% placebo: 8.8 mo Placebo: 18.7 mo Dab + Tram: 59 (5.9–9.3) Dab + placebo: 25 Primary endpoint: OS HR 0.69 (95% CI HR 0.56 (95% CI 0.59–0.70) Fatigue: 0.46–0.69) Dab + Tram: 29 Dab + placebo: 37 Nausea: Dab + Tram: 36 Dab + placebo: 27 Cutaneous SCC: Dab + Tram: 4 Dab + placebo: 12 COMBI-v12 1:1 randomization to: 704 Dabrafenib + Dabrafenib + Dabrafenib + All: BRAF V600E or • Dabrafenib 150 mg twice trametinib: 64% trametinib: trametinib: Dab + Tram: 52 V600K–mutated, daily + trametinib 2 mg Vemurafenib: 51% 11.4 mo 25.6 mo Vemurafenib: 63 once daily previ­ously untreat­ed Vemurafenib: Vemurafenib: Pyrexia: • Vemurafenib 960 mg advanced melanoma 7.3 mo 18 mo Dab + Tram: 53 twice daily Vemurafenib: 21 Primary endpoint: OS HR 0.61 (95% CI HR 0.66 (95% CI 0.52–0.73) 0.53–0.81) Rash: Dab + Tram: 22 Vemurafenib: 43 Nausea: Dab + Tram: 35 Vemurafenib: 36% Arthralgia: Dab + Tram: 24 Vemurafenib: 51 + Binimetinib COLUMBUS78 1:1:1: randomization to: 921 Encorafenib + Encorafenib + Not reported All: Locally advanced or met- • Encorafenib 450 mg/day + binimetinib: 63% binimetinib: Enco + Bini: 58 astatic melanoma with binimetinib 45 mg twice Encorafenib: 51% 14.9 mo Encorafenib: 66 BRAF V600 mutation daily (11.0–18.5) Vemurafenib: 63 • Encorafenib 300 mg daily Vemurafenib: 40% Nausea: Prior therapy with Encorafenib: • Vemurafenib 960 mg 9.6 mo (7.5–14.8) Enco + Bini: 41 immuno­therapy was twice daily Encorafenib: 39 allowed Vemurafenib: Vemurafenib: 34 Primary endpoint: PFS 7.3 mo (5.6–8.2) Pyrexia: HR 0.54 (95% CI Enco + Bini:18 0.41–0.71) Encorafenib: 15 Vemurafenib: 28 Fatigue: Enco + Bini:29 Encorafenib: 25 Vemurafenib: 31 Rash: Enco + Bini: 14 Encorafenib: 21 Vemurafenib: 29

AE = adverse events; CI = confidence interval; GI = gastrointestinal; HR = hazard ratio; ICC = investigator’s choice chemotherapy; mPFS = median progression-free survival; mOS = median overall survival; ORR = objective response rate. www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 37 Metastatic Cutaneous Melanoma

BRAF and MEK inhibition produced greater Binimetinib has been compared to dacarba- responses and improved progression-free and zine in a phase 3 study (NEMO) of patients with overall survival (Table 2). Interestingly, the rate NRAS-mutant melanoma, most of whom had of cutaneous squamous-cell carcinomas was been previously treated with immunotherapy.80 much lower with combination therapy, reflect- Response rates were low in both arms, although ing the more profound degree of MAPK path- slightly greater with binimetinib than dacarba- way inhibition achieved with combination BRAF zine (15% versus 9%), commensurate with a and MEK inhibition. Based on these results, modest improvement in progression-free surviv- FDA approval was granted for both dabrafenib/ al. FDA approval has been sought and remains trametinib and vemurafenib/cobimetinib com- pending at this time. binations in 2015. Although the dabrafenib/ trametinib combination was only approved in KIT Inhibition Singly and In Combination 2015, trametinib had independently gained The KIT receptor protein tyrosine kinase is a FDA approval in 2013 for the treatment of transmembrane protein consisting of extracel- BRAF V600E/K–mutated melanoma on the lular and intracellular domains. Activating KIT basis of the phase 3 METRIC study.77 mutations occur in 2% to 8% of all melanoma Encorafenib (LGX818) and binimetinib patients and may be found in all melanoma (MEK162, ARRY-162, ARRY-438162) are new subtypes but are commonest in acral mela- BRAF and MEK inhibitors currently being evalu- nomas (10%–20%) and mucosal melanomas ated in clinical trials. Encorafenib/binimetinib (15%–20%). Activating KIT mutations primarily combination was first evaluated in a phase 3 study occur in exons 11 and 13, which code for the (COLUMBUS) that compared it with vemu- juxtamembrane and kinase domains, respec- rafenib monotherapy in BRAF-mutant melano- tively.5,81–83 ma.78 Unsurprisingly, encorafenib/binimetinib mesylate is a tyrosine kinase in- combination produced greater and more du- hibitor of the 2-phenyl amino pyrimidine class rable responses compared to vemurafenib mono- that occupies the tyrosine kinase active site therapy. The median progression-free survival with resultant blocking of tyrosine kinase activ- of the encorafenib/binimetinib combination ity. Imatinib mesylate is known to block KIT (14.9 months) was greater than vemurafenib and has been extensively studied in patients monotherapy (7.3 months) in this study, and with gastrointestinal stromal tumors (GIST), intriguingly greater than that seen with vemu- 80% of whom harbor KIT mutations, in both rafenib/cobimetinib (coBRIM 9.9 months) and the adjuvant and the metastatic settings. In dabrafenib/trametinib (COMBI-d 9.3 months; melanoma, imatinib mesylate was studied in a COMBI-v 11.4 months). Of note, although Chinese open-label, phase 2 study of imatinib encorafenib has an IC50 midway between dab- mesylate monotherapy in metastatic melano- rafenib and vemurafenib in cell-free assays ma harboring KIT mutation or amplification; (0.8 nM dabrafenib, 4 nM encorafenib, and 31 25% of the study patients had mucosal disease nM vemurafenib), it has an extremely slower and the rest had cutaneous disease, with acral off-rate from BRAF V600E, which results in sig- involvement in 50% of all patients.84 Overall nificantly greater target inhibition in cells follow- response rate was 23%, while 51% of patients ing drug wash-out.79 This may account for the remained alive at 1 year with no differences significantly greater clinical benefit seen with in response rate and/or survival being noted encorafenib/binimetinib in clinical trials. Final between patients with either KIT mutations or study data are eagerly awaited. Regulatory approv- amplifications. In a separate study of imatinib al has been sought, and is pending at this time. mesylate at 400 mg daily or 400 mg twice daily in

38 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

Caucasian patients with KIT-mutated/amplified other disease. The primary events underlying melanoma, similar response and survival rates this are advances in our understanding of the were reported, although patients with KIT mu- gene mutation landscape driving melanoma tations did nonsignificantly better than those tumorigenesis, accompanied by insights into with KIT amplifications.85 the means by which tumors circumvent the in- Other novel studies evaluating KIT inhibi- duction of effective anti-tumor T-cell responses. tors include KIT inhibition in combination These insights have resulted in the development with the VEGF inhibitor and a of inhibitors targeting MAPK pathway kinases study of selective BCR-ABL kinase inhibitor ni- BRAF, MEK, and NRAS), KIT, and regulato- lotinib in imatinib-resistant melanoma. In the ry immune checkpoints (CTLA-4 and PD-1). former phase 1/2 study, Flaherty and colleagues Although BRAF/MEK inhibition results in studied imatinib 800 mg daily and bevacizumab profound reductions and even occasional com- at 10 mg/kg every 2 weeks in 63 patients with plete responses in patients, these responses are advanced tumors, including 23 with metastatic typically short lived, rarely lasting more than 9 to melanoma. Although the combination was rela- 11 months; the encorafenib/binimetinib com- tively nontoxic, no significant efficacy signal was bination may improve that duration marginally. seen and further accrual to the phase 2 portion However, the signature therapeutic advance in was halted after the first stage was completed.86 melanoma of the past decade is immunother- is a BCR-ABL1 tyrosine kinase inhibi- apy, particularly the development of inhibitors tor intelligently designed based on the structure of CTLA-4 and PD-1 immune checkpoints. of the ABL-imatinib complex that is 10 to 30 With these agents, significant proportions of times more potent than imatinib in inhibiting treated patients remain free of progression BCR-ABL1 tyrosine kinase activity. Nilotinib is off-therapy (ipilimumab 23%; nivolumab 34%; approved for the treatment of imatinib-resistant pembrolizumab 35%; ipilimumab/nivolumab chronic myelogenous (CML), with 64%), and some patients can be successfully re- reported efficacy in patients with central ner- induced after delayed progression. Separately, vous system (CNS) involvement.87,88 Nilotinib the high response rates observed with the use has been studied in a single study of KIT-mutat- of KIT inhibitors in CML and GIST have not ed/amplified melanoma that included patients been observed in KIT mutated/amplified mela- with imatinib-resistance and those with treated noma and development of agents in this space CNS disease. Nilotinib appeared to be active in has been limited. The challenges ahead center imatinib-resistant melanoma, although no re- around identifying predictive biomarkers and sponses were seen in the CNS disease cohort.89 circumventing primary or acquired resistance, Overall, the response rates observed with KIT with the eventual goal of producing durable inhibition in melanoma are much lower than remissions in the majority of treated patients. those observed in CML and GIST. Our improved understanding of the mecha- nisms of acquired resistance to BRAF/MEK in- hibitors suggests that anti-tumor activity may be Conclusion and future directions achieved by targeting multiple pathways, possibly with combination regimens comprising other in- Prior to 2011, the only approved agents for hibitors and/or immunotherapy. Preclinical data the treatment of advanced melanoma were supports the use of combination strategies target- dacarbazine and high-dose -2. Since ing both ERK and PI3K/mTOR to circumvent 2011, drug approvals in melanoma have pro- acquired resistance.90 Ongoing studies are evalu- ceeded at a frenetic pace unmatched in any ating combinations with biguanides (metformin:

www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 39 Metastatic Cutaneous Melanoma

NCT02143050 and NCT01638676; phenformin: polymorphisms, and tumor mutation load. The NCT03026517), HSP90 inhibitors (XL888: goal is to use multiple markers to guide devel- NCT02721459; AT13387: NCT02097225), and opment of combinations and then, depend- decitabine (NCT01876641). ing on initial response, to examine tumors for One complexity affecting management of alterations to guide decisions about additional resistance in the targeted therapy landscape treatment(s) to improve responses, with the even- remains tumor heterogeneity, particularly intra- tual goal being durable clinical responses for all and intertumoral heterogeneity, which may patients. explain the apparent contradiction between continued efficacy of BRAF inhibitors in BRAF- BOARD REVIEW QUESTIONS resistant tumors and preclinical data predicting Test your knowledge of this topic. Go to slower progression of resistant tumors on cessa- www.turner-white.com/brm/bonco.htm and tion of BRAF inhibitors.91–94 These data provide click the Board Review Questions button. a rationale to investigate intermittent dosing regimens with BRAF/MEK inhibitors; several studies exploring this approach are ongoing REFERENCES (NCT01894672 and NCT02583516). Given the specificity, adaptability, and mem- 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016;66:7–30. ory response associated with immunotherapy, 2. Guy GP, Thomas CC, Thompson T, et al. Vital signs: melanoma it is likely that these agents will be used to treat incidence and mortality trends and projections - United States, the majority of patients regardless of mutational 1982-2030. MMWR Morb Mortal Wkly Rep 2015;64:591–6. 3. Anderson WF, Pfeiffer RM, Tucker MA, Rosenberg PS. Divergent status. Hence, identifying predictive biomarkers cancer pathways for early-onset and late-onset cutaneous malig- of response to immune checkpoint inhibitors nant melanoma. Cancer 2009;115:4176–85. 4. Curtin JA, Fridlyand J, Kageshita T, et al. Distinct sets of genetic is vital. The presence of CD8+ T-cell infiltrate alterations in melanoma. N Engl J Med 2005;353:2135–47. and IFN-γ gene signature, which indicate an 5. Curtin JA, Busam K, Pinkel D, Bastian BC. Somatic activation “inflamed” tumor microenvironment, are highly of KIT in distinct subtypes of melanoma. J Clin Oncol 2006;24: 4340–6. predictive of clinical benefit from PD-1 inhibi- 6. Hodis E, Watson IR, Kryukov GV, et al. A landscape of driver tors.95,96 However, not all PD-1 responders have mutations in melanoma. Cell 2012;150:251–63. 7. Cancer Genome Atlas Network. Genomic classification of cuta- “inflamed” tumor microenvironments, and not neous melanoma. Cell 2015;161:1681–96. all patients with an “inflamed” tumor micro- 8. Chapman PB, Hauschild A, Robert C, et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N environment respond to immune checkpoint Engl J Med 2011;364:2507–16. inhibitors. The complexity of the immune sys- 9. McArthur GA, Chapman PB, Robert C, et al. Safety and efficacy tem is reflected in the multiple non-redundant of vemurafenib in BRAF(V600E) and BRAF(V600K) mutation- positive melanoma (BRIM-3): extended follow-up of a phase 3, immunologic pathways, both positive and nega- randomised, open-label study. Lancet Oncol 2014;15:323–32. tive, with checkpoints and ligands that emerge 10. Hauschild A, Grob J-J, Demidov LV, et al. Dabrafenib in BRAF- dynamically in response to treatment. Given the mutated metastatic melanoma: a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012;380:358–65. dynamic nature of the immune response, it is 11. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and unlikely that any single immunologic biomarker MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med 2014;371:1877–88. identified pre-treatment will be completely pre- 12. Robert C, Karaszewska B, Schachter J, et al. Improved overall dictive. Rather, the complexity of the biomarker survival in melanoma with combined dabrafenib and trametinib. approach must match the complexity of the N Engl J Med 2015;372:30–9. 13. Larkin J, Ascierto PA, Dréno B, et al. Combined vemurafenib immune response elicited, and will likely incor- and cobimetinib in BRAF-mutated melanoma. N Engl J Med porate multifarious elements including CD8+ 2014;371:1867–76. 14. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 T-cell infiltrate, IFN-γ gene signature, and addi- combination blockade expands infiltrating T cells and reduces tional elements including microbiome, genetic regulatory T and myeloid cells within B16 melanoma tumors.

40 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

Proc Natl Acad Sci U S A 2010;107:4275–80. in tolerance and immunity. Annu Rev Immunol 2008;26:677– 15. Gubin MM, Zhang X, Schuster H, et al. Checkpoint blockade 704. targets tumour-specific mutant antigens. 38. Yamazaki T, Akiba H, Iwai H, et al. Expression of programmed Nature 2014;515:577–81. death 1 ligands by murine T cells and APC. J Immunol 16. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with 2002;169:5538–45. ipilimumab in patients with metastatic melanoma. N Engl J Med 39. Youngnak P, Kozono Y, Kozono H, et al. Differential binding 2010;363:711–23. properties of B7-H1 and B7-DC to programmed death-1. Bio- 17. Robert C, Thomas L, Bondarenko I, et al. Ipilimumab plus da- chem Biophys Res Commun 2003;307:672–7. carbazine for previously untreated metastatic melanoma. N Engl 40. Chemnitz JM, Parry RV, Nichols KE, June CH, Riley JL. SHP-1 J Med 2011;364:2517–26. and SHP-2 associate with immunoreceptor tyrosine-based switch 18. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and im- motif of programmed death 1 upon primary human T cell mune correlates of anti-PD-1 antibody in cancer. N Engl J Med stimulation, but only receptor ligation prevents T cell activation. 2012;366:2443–54. J Immunol 2004;173:945–54. 19. Weber JS, Angelo SP D’, Minor D, et al. Nivolumab versus che- 41. Parry RV, Chemnitz JM, Frauwirth KA, et al. CTLA-4 and PD-1 motherapy in patients with advanced melanoma who progressed receptors inhibit T-cell activation by distinct mechanisms. Mol after anti-CTLA-4 treatment (CheckMate 037): a randomised, Cell Biol 2005;25:9543–53. controlled, open-label, phase 3 trial. Lancet Oncol 2015;16: 42. Riley JL. PD-1 signaling in primary T cells. Immunol Rev 375–84. 2009;229:114–25. 20. Robert C, Long GV, Brady B, et al. Nivolumab in previously 43. Wolchok JD, Hodi FS, Weber JS, et al. Development of ipilim- untreated melanoma without BRAF mutation. N Engl J Med umab: a novel immunotherapeutic approach for the treatment 2015;372:320–30. of advanced melanoma. Ann N Y Acad Sci 2013;1291:1–13. 21. Hamid O, Robert C, Daud A, et al. Safety and tumor responses 44. Weber JS, O’Day S, Urba W, et al. Phase I/II study of ipi- with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med limumab for patients with metastatic melanoma. J Clin Oncol 2013;369:134–44. 2008;26:5950–6. 22. Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipi- 45. Hodi FS, Butler M, Oble DA, et al. Immunologic and clinical ef- limumab in advanced melanoma. N Engl J Med 2015;372:2521– fects of antibody blockade of cytotoxic T lymphocyte-associated 32. antigen 4 in previously vaccinated cancer patients. Proc Natl 23. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipili- Acad Sci U S A 2008;105:3005–10. mumab in advanced melanoma. N Engl J Med 2013;369:122–33. 46. Wolchok JD, Neyns B, Linette G, et al. Ipilimumab monotherapy 24. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilim- in patients with pretreated advanced melanoma: a randomised, umab versus ipilimumab in untreated melanoma. N Engl J Med double-blind, multicentre, phase 2, dose-ranging study. Lancet 2015;372:2006–17. Oncol 2010;11:155–64. 25. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolum- 47. Schadendorf D, Hodi FS, Robert C, et al. Pooled analysis of ab and ipilimumab or monotherapy in untreated melanoma. long-term survival data from phase II and phase III trials of ipi- N Engl J Med 2015;373:23–34. limumab in unresectable or metastatic melanoma. J Clin Oncol 26. Eggermont AMM. Therapeutic vaccines in solid tumours: can 2015;33:1889–94. they be harmful? Eur J Cancer 2009;45:2087–90. 48. Wang C, Thudium KB, Han M, et al. In vitro characterization 27. Rosenberg SA. Raising the bar: the curative potential of human of the anti-PD-1 antibody nivolumab, BMS-936558, and in cancer immunotherapy. Sci Transl Med 2012;4:127ps8. vivo toxicology in non-human primates. Cancer Immunol Res 28. Rosenberg SA. IL-2: the first effective immunotherapy for human 2014;2:846–56. cancer. J Immunol 2014;192:5451–8. 49. Poole RM. Pembrolizumab: first global approval. Drugs 29. Rosenberg SA, Restifo NP. Adoptive cell transfer as personalized 2014;74:1973–81. immunotherapy for human cancer. Science 2015;348:62–8. 50. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable 30. Harding FA, McArthur JG, Gross JA, Raulet DH, Allison JP. CD28- tumor remission, and long-term safety in patients with advanced mediated signalling co-stimulates murine T cells and prevents melanoma receiving nivolumab. J Clin Oncol 2014;32:1020–30. induction of anergy in T-cell clones. Nature 1992;356:607–9. 51. Patnaik A, Kang SP, Rasco D, et al. Phase I study of pembroli- 31. Greenfield EA, Nguyen KA, Kuchroo VK. CD28/B7 costimula- zumab (MK-3475; anti-PD-1 monoclonal antibody) in patients tion: a review. Crit Rev Immunol 1998;18:389–418. with advanced solid tumors. Clin Cancer Res 2015;21:4286–93. 32. Sharpe AH, Abbas AK. T-cell costimulation--biology, therapeutic 52. Hellmann MD, Rizvi NA, Goldman JW, et al. Nivolumab plus ipilim- potential, and challenges. N Engl J Med 2006;355:973–5. umab as first-line treatment for advanced non-small-cell lung cancer 33. Chambers CA, Kuhns MS, Egen JG, Allison JP. CTLA-4-mediated (CheckMate 012): results of an open-label, phase 1, multicohort inhibition in regulation of T cell responses: mechanisms and study. Lancet Oncol 2017;18:31–41. manipulation in tumor immunotherapy. Annu Rev Immunol 53. Dhomen N, Marais R. BRAF signaling and targeted therapies in 2001;19:565–94. melanoma. Hematol Oncol Clin North Am 2009;23:529–45, ix. 34. Collins AV, Brodie DW, Gilbert RJC, et al. The interaction proper- 54. Satyamoorthy K, Li G, Gerrero MR, et al. Constitutive mitogen- ties of costimulatory molecules revisited. Immunity 2002;17:201– activated protein kinase activation in melanoma is mediated by 10. both BRAF mutations and autocrine growth factor stimulation. 35. Krummel MF, Allison JP. CD28 and CTLA-4 have opposing Cancer Res 2003;63:756–9. effects on the response of T cells to stimulation. J Exp Med 55. Davies H, Bignell GR, Cox C, et al. Mutations of the BRAF gene 1995;182:459–65. in human cancer. Nature 2002;417:949–54. 36. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor 56. Krauthammer M, Kong Y, Ha BH, et al. Exome sequencing immunity by CTLA-4 blockade. Science 1996;271:1734–6. identifies recurrent somatic RAC1 mutations in melanoma. Nat 37. Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands Genet 2012;44:1006–14. www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 41 Metastatic Cutaneous Melanoma

57. Rubinstein JC, Sznol M, Pavlick AC, et al. Incidence of the V600K 77. Flaherty KT, Robert C, Hersey P, et al. Improved survival with mutation among melanoma patients with BRAF mutations, and MEK inhibition in BRAF-mutated melanoma. N Engl J Med potential therapeutic response to the specific BRAF inhibitor 2012;367:107–14. PLX4032. J Transl Med 2010;8:67. 78. Dummer R, Ascierto PA, Gogas HJ, et al. Results of COLUMBUS 58. Lovly CM, Dahlman KB, Fohn LE, et al. Routine multiplex mu- Part 1: a phase 3 trial of encorafenib (ENCO) plus binimetinib tational profiling of melanomas enables enrollment in genotype- (BINI) versus vemurafenib (VEM) or ENCO in BRAF-mutant driven therapeutic trials. PLoS ONE 2012;7:e35309. melanoma. Presented at Society for Melanoma Research 2016 59. Hatzivassiliou G, Song K, Yen I, et al. RAF inhibitors prime wild- Congress. November 6-9,­ 2016. Boston (MA). type RAF to activate the MAPK pathway and enhance growth. 79. Adelmann CH, Ching G, Du L, et al. Comparative profiles of Nature 2010;464:431–5. BRAF inhibitors: the paradox index as a predictor of clinical 60. Poulikakos PI, Zhang C, Bollag G, Shokat KM, Rosen N. RAF toxicity. Oncotarget 2016;7:30453–60. inhibitors transactivate RAF dimers and ERK signalling in cells 80. Dummer R, Schadendorf D, Ascierto PA, et al. Binimetinib ver- with wild-type BRAF. Nature 2010;464:427–30. sus dacarbazine in patients with advanced NRAS-mutant mela- 61. Thomas NE, Edmiston SN, Alexander A, et al. Association noma (NEMO): a multicentre, open-label, randomised, phase 3 between NRAS and BRAF mutational status and melanoma-spe- trial. Lancet Oncol 2017;18:435–45. cific survival among patients with higher-risk primary melanoma. 81. Willmore-Payne C, Holden JA, Tripp S, Layfield LJ. Human JAMA Oncology 2015;1:359–68. malignant melanoma: detection of BRAF- and c--activating 62. Joseph RW, Sullivan RJ, Harrell R, et al. Correlation of NRAS mutations by high-resolution amplicon melting analysis. Hum mutations with clinical response to high-dose IL-2 in patients with Pathol 2005;36:486–93. advanced melanoma. J Immunother 2012;35:66–72. 82. Beadling C, Jacobson-Dunlop E, Hodi FS, et al. KIT gene muta- 63. Johnson DB, Lovly CM, Flavin M, et al. Impact of NRAS muta- tions and copy number in melanoma subtypes. Clin Cancer Res tions for patients with advanced melanoma treated with immune 2008;14:6821–8. therapies. Cancer Immunol Res 2015;3:288–95. 83. Handolias D, Salemi R, Murray W, et al. Mutations in KIT occur 64. Johnson DB, Frampton GM, Rioth MJ, et al. Targeted next at low frequency in melanomas arising from anatomical sites generation sequencing identifies markers of response to PD-1 associated with chronic and intermittent sun exposure. Pigment blockade. Cancer Immunol Res 2016;4:959–67. Cell Melanoma Res 2010;23:210–5. 65. Kiuru M, Busam KJ. The NF1 gene in tumor syndromes and 84. Guo J, Si L, Kong Y, et al. Phase II, open-label, single-arm trial of melanoma. Lab Invest 2017;97:146–57. imatinib mesylate in patients with metastatic melanoma harbor- 66. Nissan MH, Pratilas CA, Jones AM, et al. Loss of NF1 in cutaneous ing c-Kit mutation or amplification. J Clin Oncol 2011;29:2904–9. melanoma is associated with RAS activation and MEK depen- 85. Hodi FS, Corless CL, Giobbie-Hurder A, et al. Imatinib for dence. Cancer Res 2014;74:2340–50. melanomas harboring mutationally activated or amplified KIT 67. Lorusso PM, Adjei AA, Varterasian M, et al. Phase I and pharma- arising on mucosal, acral, and chronically sun-damaged skin. J codynamic study of the oral MEK inhibitor CI-1040 in patients Clin Oncol 2013;31:3182–90. with advanced malignancies. J Clin Oncol 2005;23:5281–93. 86. Flaherty KT, Hamilton BK, Rosen MA, et al. Phase I/II trial of 68. Rinehart J, Adjei AA, Lorusso PM, et al. Multicenter phase II imatinib and bevacizumab in patients with advanced melanoma study of the oral MEK inhibitor, CI-1040, in patients with ad- and other advanced cancers. Oncologist 2015;20:952–9. vanced non-small-cell lung, breast, colon, and pancreatic cancer. 87. Giles FJ, le Coutre PD, Pinilla-Ibarz J, et al. Nilotinib in imatinib- J Clin Oncol 2004;22:4456–62. resistant or imatinib-intolerant patients with chronic myeloid 69. Kirkwood JM, Bastholt L, Robert C, et al. Phase II, open-label, leukemia in chronic phase: 48-month follow-up results of a phase randomized trial of the MEK1/2 inhibitor as mono- II study. Leukemia 2013;27:107–12. therapy versus temozolomide in patients with advanced mela- 88. Reinwald M, Schleyer E, Kiewe P, et al. Efficacy and pharmaco- noma. Clin Cancer Res 2012;18:555–67. logic data of second-generation tyrosine kinase inhibitor nilo- 70. Davar D, Kirkwood JM. CCR 20th anniversary commentary: tinib in BCR-ABL-positive leukemia patients with central nervous MAPK/ERK pathway inhibition in melanoma-kinase inhibition system relapse after allogeneic stem cell transplantation. Biomed redux. Clin Cancer Res 2015;21:5412–4. Res Int 2014;2014:637059. 71. Su F, Viros A, Milagre C, et al. RAS mutations in cutaneous 89. Carvajal RD, Lawrence DP, Weber JS, et al. Phase II study of squamous-cell carcinomas in patients treated with BRAF inhibi- nilotinib in melanoma harboring KIT alterations following tors. N Engl J Med 2012;366:207–15. progression to prior KIT inhibition. Clin Cancer Res 2015;21: 72. Johannessen CM, Boehm JS, Kim SY, et al. COT drives resistance 2289–96. to RAF inhibition through MAP kinase pathway reactivation. 90. Carlino MS, Todd JR, Gowrishankar K, et al. Differential activity Nature 2010;468:968–72. of MEK and ERK inhibitors in BRAF inhibitor resistant mela- 73. Nazarian R, Shi H, Wang Q, et al. Melanomas acquire resistance noma. Mol Oncol 2014;8:544–54. to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. 91. Carlino MS, Gowrishankar K, Saunders CAB, et al. Antiprolifera- Nature 2010;468:973–7. tive effects of continued mitogen-activated protein kinase pathway 74. Shi H, Hong A, Kong X, et al. A novel AKT1 mutant amplifies an inhibition following acquired resistance to BRAF and/or MEK adaptive melanoma response to BRAF inhibition. Cancer Discov inhibition in melanoma. Mol Cancer Ther 2013;12:1332–42. 2014;4:69–79. 92. Chan MMK, Haydu LE, Menzies AM, et al. The nature and 75. Shi H, Hugo W, Kong X, et al. Acquired resistance and clonal management of metastatic melanoma after progression on evolution in melanoma during BRAF inhibitor therapy. Cancer BRAF inhibitors: effects of extended BRAF inhibition. Cancer Discov 2014;4:80–93. 2014;120:3142–53. 76. Van Allen EM, Wagle N, Sucker A, et al. The genetic landscape 93. Thakur M Das, Salangsang F, Landman AS, et al. Modelling ve- of clinical resistance to RAF inhibition in metastatic melanoma. murafenib resistance in melanoma reveals a strategy to forestall Cancer Discov 2014;4:94–109. drug resistance. Nature 2013;494:251–5.

42 Hospital Physician Board Review Manual www.hpboardreview.com Metastatic Cutaneous Melanoma

94. Thakur M Das, Stuart DD. Molecular pathways: response and 2014;515:568–71. resistance to BRAF and MEK inhibitors in BRAF(V600E) tumors. 96. Ayers M, Lunceford J, Nebozhyn M, et al. Relationship between Clin Cancer Res 2014;20:1074–80. immune gene signatures and clinical response to PD-1 blockade 95. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces with pembrolizumab (MK-3475) in patients with advanced solid responses by inhibiting adaptive immune resistance. Nature tumors. J Immunotherapy Cancer 2015;3(Suppl 2):P80.

Access Interactive Board Review Questions and Download the Free Hematology/Oncology App at: www.turner-white.com/brm/bonco.htm

www.hpboardreview.com Hematology-Oncology Volume 12, Part 4 43