Development of CKI and TT have dramatically changed the treatment options for patients with metastatic melanoma. Regardless of treatment choice, eventual disease progression in the majority of patients remains a clinical challenge. For example, patients whose best response to ipilimumab and nivolumab was progressive or stable disease had a median survival of only 3 and 6 months respectively [Long et al., Characteristics of Long-Term Survivors and Subgroup Analyses with Combination Nivolumab Plus Ipilimumab for Advanced Melanoma (CheckMate 067), presented at Society for Melanoma Research 2019]. In a retrospective reanalysis of 3 clinical trials, patients treated second line TT after initial CKI failure had a PFS of only 18% when treated with 2nd line BRAF ± MEK inhibitors [32]. Thus, better treatment options for patients with progression of melanoma after initial CKI therapy are still badly needed.
Since targeted agents induce a rapid and deep response with eventual development of adaptive resistance and checkpoint inhibitors produce more gradual onset of activity with a significant fraction of complete and durable responses, combining CKI and TT is highly attractive in BRAF mutant patients. Unfortunately, early attempts at combination therapy led to unacceptably high levels of toxicity [33, 23, 34]
This toxicity appeared, in part, due to the unanticipated immunologic activity of BRAF and MEK inhibitors. BRAF ± MEK inhibitor therapy appears to result in significant immunologic potentiation, by increasing immune infiltration into tumors and enhancing T cell cytotoxicity [20, 21], in part via increased PD-1 ligand expression on melanoma cells [22]. In addition TT may increase melanoma specific antigen expression, enhance dendritic cell function, increase NK and T cell function, and overcome inhibitor influences in the tumor microenvironment [19]. More recently, a number of front-line combination trials of BRAF + MEK inhibitors have reported results, most of which have not met their primary endpoint goals. However, these reports have provided tantalizing hints of potential effectiveness of these regimens [35, 34, 36].
In the Keynote 022 trial, patients were randomly assigned 1:1 to receive dabrafenib (150 mg orally two times per day) and trametinib (2 mg orally one time a day) with intravenous pembrolizumab (200 mg every 3 weeks) or placebo. The primary endpoint was PFS. With 36.6 months of follow-up, median PFS was 16.9 months (95% CI 11.3 to 27.9) with triplet and 10.7 months (95% CI 7.2 to 16.8) with doublet (HR 0.53; 95% CI 0.34 to 0.83)(p = 0.53). With triplet and doublet, respectively, PFS at 24 months was 41.0% and 16.3%; median response duration were 25.1 months and 12.1 months, respectively. Median OS was not reached with triplet and was 26.3 months with doublet (HR 0.64; 95% CI 0.38 to 1.06). Grade 3–5 treatment-related adverse events (TRAEs) occurred in 35 patients (58%, including one death) receiving triplet and 15 patients (25%) receiving doublet [36].
The IMspire150 trial was a randomized, double-blind, placebo-controlled phase 3 study that enrolled 514 patients. Patients with unresectable stage IIIc–IV, BRAF V600E mutant melanoma were randomly assigned 1:1 to 28-day cycles of vemurafenib (960 mg b.i.d.), and cobimetinib (60 mg/d for 21 days) with either atezolizumab (840 mg) or placebo. At a median follow-up of 18.9 months, progression-free survival as assessed by the study investigator was significantly prolonged with atezolizumab versus control (15.1 vs 10.6 months; hazard ratio [HR] 0.78; 95% CI 0.63–0.97; p = 0·025). Treatment-related adverse events occurred in 99% of both the atezolizumab and control groups, with 13% of patients in the atezolizumab group and 16% in the control group stopping all treatment because of adverse events [35].
In the COMBI-I trial, 532 pts were randomized to receive spartalizumab 400 mg or placebo IV Q4W with dabrafenib 150 mg orally BID and trametinib 2 mg orally QD. At median follow-up 27.2 mo., the spartalizumab-based regimen did not significantly improve PFS (median PFS, 16.2 mo. vs 12.0 mo.; HR, 0.82 [95% CI, 0.655–1.027]; P = .042). Estimated 12- and 24-mo PFS rates with the spartalizumab regimen versus placebo were 58% vs 50% and 44% vs 36%, respectively. The objective response rate was 69% in the spartalizumab arm (CR rate 20%) vs 64% in the placebo arm (CR rate, 18%); median duration of response was NR vs 20.7 mo., respectively. Tx-related adverse events (TRAEs) grade ≥ 3 occurred in 55% vs 33% of pts treated with spartalizumb arm versus placebo. TRAEs leading to discontinuation of all 3 study drugs occurred in 12% vs 8% of pts in the two arms, respectively [37].
We approached the problem of immunotherapy resistant patients somewhat differently than most current trials. In a community setting, there are frequently significant delays in identifying BRAF mutant patients, as reflex BRAF mutation testing is not routinely being performed by community pathologists. Also, V600E-specific mutation testing by monoclonal antibody staining is known to miss a percentage of patients with other BRAF gene mutations (including V600K, V600R and internal gene rearrangements or gene fusions) as well as other potentially targetable mutations, which are more accurately pinpointed via next-gen sequencing panels [38]. Thus, initiating treatment with CKI avoids lengthy treatment delays while awaiting molecular test results. In addition, there is developing data in BRAF mutant patients that initial CKI therapy followed by BRAF inhibition at relapse may have a higher progression free survival than the reverse sequence [39–41, 32].
We also recognized that a substantial percentage of patients (30–40%) treated with initial immunotherapy would achieve complete and durable remissions without any additional therapy. Inclusion of patients with potential CKI response may have confounded analysis of the randomized trials described above and exposed these patients to unnecessary risks for toxicity. BRAF mutant patients may have a modestly increased response potential to CKI treatment compared to non-BRAF mutated patients [6]. It is also known that the majority of patients with best response of progressive or stable disease progression after initial CKI treatment will rapidly die of their disease, with a median survival of 3–6 months [Long et al, Characteristics of Long-Term Survivors and Subgroup Analyses with Combination Nivolumab Plus Ipilimumab for Advanced Melanoma (CheckMate 067), Society for Melanoma Research 2019]. Thus, we chose to treat only patients who had clearly progressed on immunotherapy with cautious addition of TT to ongoing PD-1 directed treatment. This avoided overtreatment of patients who would achieve complete response without addition of TT, avoiding unnecessary additive toxicity of combining CKI and TT. We also discarded the chemotherapy concept of “maximum tolerated dose” unlike the trials cited above, as it is not clear that this applies to either CKI or TT therapy [42–44], instead seeking a minimum effective dose of TT with CKI. Our belief was that this might arrest tumor progression while enhancing potentially synergistic immune activation and decrease the risk of additive toxicity.
Our results support the clinical activity of a sequential treatment approach. We were able to successfully combine CKI with cautious escalation of TT. Our approach was able to produce a significant frequency of complete responses in 55% of patients including a significant number of patients with “hyperprogression”. This included patients with either a BRAF V600E and V600K mutation. Progression free survival at a median follow-up at 33 months was 34.1 months. Median overall survival was appeared to show a median survival of 34.1 months with a plateau of 48% after 3 years. Of patients achieving a complete response, 10/13 remain in long term remission. Many of these patients have been able to successfully discontinue all treatment based on criteria published by Robert et al [31], and remain in ongoing remission. One patient died of a late CNS relapse; two others remain in complete remission on ongoing TT. In progressing patients, brain metastases proved to be a significant component of progression in 70%. This is a common challenge in BRAF mutant melanomas [45].
The toxicity of our approach seemed modest compared to data from the trials described above. In our series toxicity mostly reached Grade 1–2 intensity. Most toxicity was managed with temporary treatment interruption and steroid administration, if toxicity was typical for CKI immunologic toxicity, or withholding TT and subsequent dose reduction if toxicity was believed to be TT related (e.g., a typical non-pruritic MEK inhibitor rash). In these patients, treatment was resumed once toxicity reached grade 1 or less. In patients with recurrence of TT toxicity after re-challenge, patients were converted to alternate BRAF or MEK inhibitors (6 patients, one required a trial of 3 separate combinations to achieve acceptable levels of toxicity). A total of 21/23 patients was able to remain on therapy after toxicity resolved. Only 2/23 patients had to discontinue treatment due to toxicity. These two patients required hospitalization for grade 4 biopsy-proven interstitial nephritis. In the first patient, renal impairment responded completely to treatment cessation and high dose steroids. This patient did not receive further combined therapy and continues in long-term complete response. The second patient had both interstitial nephritis, ataxia, and concomitant acute sensory-motor neuropathy. He responded dramatically to high dose steroids with the addition of IV immunoglobulin. Treatment with TT was resumed after recovery, however this patient (with a BRAF insertion) eventually died due to melanoma progression. One patient died suddenly of a sudden cardiopulmonary event.
Unfortunately, a subset (44%) of patients failed to respond to the addition of TT to PD-1 treatment. Further work will be needed to understand the mechanism(s) of resistance and to identify more active treatment options in this subset of patients. A number of these progressing patients were characterized by the presence of elevated LDH or brain metastases at progression (known adverse prognostic markers). Patients with internal BRAF rearrangements or fusions also appeared to have an extremely poor prognosis after treatment with both CKI and CKI + TT. The dichotomous response pattern in our patients is curious: Most responding patients eventually reached a complete response and virtually all initially progressing patients have died. The basis for this observation remains to be elucidated. Our clinical trial suggests that it may be possible to identify the eventual response pattern at early times (e.g., within 2–3 months) after adding TT to CKI. If verified, this potentially would allow non-responding patients to transition to clinical trials of new agents, before disease progression results in deterioration of performance status.
Our current study is intended to be hypothesis generating, with a goal of providing preliminary data to supporting development of confirmatory clinical trials. Potential limitations of this study include that it is a retrospective review of patient outcomes over a number of years involving a relatively small number of patients. Patients were treated with a number of different CKI regimens as well as TT agents based on availability over this time. It is possible that an attempt to further escalate TT agent doses might further increase responses, as may adding additional agents to overcome CKI or TT resistance mechanisms. We are hopeful that our promising results will stimulate further trials of concurrent versus sequential CKI and TT.