Abstract
Background
Anti-PD-1 antibodies (PD1) prolong recurrence-free survival in high-risk resected melanoma; however, approximately 25–30% of patients recur within one year. This study describes the pattern of recurrence, management and outcomes of patients who recur with adjuvant PD1 therapy.
Patients and Methods
Consecutive patients from 16 centres who recurred having received adjuvant PD1 therapy for resected stage III/IV melanoma were studied. Recurrence characteristics, management and outcomes were examined; patients with mucosal melanoma were analysed separately.
Results
Melanoma recurrence occurred in 147 (17%) of ~850 patients treated with adjuvant PD1. In those with cutaneous melanoma (n=136), median time to recurrence was 4.6 months (range 0.3–35.7); 104 (76%) recurred during (ON) adjuvant PD1 after a median 3.2 months, and 32 (24%) following (OFF) treatment cessation after a median 12.5 months, including in 21 (15%) who ceased early for toxicity. Fifty-nine (43%) recurred with locoregional disease only and 77 (57%) with distant disease. Of those who recurred locally, 22/59 (37%) subsequently recurred distantly. Eighty-nine (65%) patients received systemic therapy after recurrence. Of those who recurred ON adjuvant PD1, none (0/6) responded to PD1 alone; 8/33 evaluable patients (24%) responded to ipilimumab (alone or in combination with PD1), and 18/23 (78%) responded to BRAF/MEK inhibitors. Of those who recurred OFF adjuvant PD1, 2/5 (40%) responded to PD1 monotherapy, 2/5 (40%) responded to ipilimumab-based therapy, and 9/10 (90%) responded to BRAF/MEK inhibitors.
Conclusions
Most patients who recur early despite adjuvant PD1 develop distant metastases. In those who recur ON adjuvant PD1, there is minimal activity of further PD1 monotherapy, but ipilimumab (alone or in combination with PD1) and BRAF/MEK inhibitors have clinical utility. Retreatment with PD1 may have activity in select patients who recur OFF PD1.
Keywords: melanoma, immunotherapy, adjuvant therapy
Introduction
Outcomes for patients with advanced melanoma have been transformed in recent years with novel systemic therapies, including PD1 and anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) immunotherapy, and targeted therapy with BRAF and MAPK kinase inhibitors (BRAF/MEKi) in BRAFV600 mutant melanoma1–3. Similar improvements are observed in the adjuvant setting for resected stage III or IV melanoma4–6, with phase III trials demonstrating prolonged recurrence-free survival (RFS) for adjuvant nivolumab versus ipilimumab4 (HR 0.65) and pembrolizumab versus placebo5 (HR 0.57). Therefore, adjuvant PD1 therapy is a new standard of care for high-risk resected melanoma, however approximately 25–30% of patients recur within one year.
To date, there are no data regarding patterns of recurrence, management and outcomes of patients who recur having received adjuvant PD1 therapy. In patients who progress on PD1 in the advanced setting, ipilimumab (alone or in combination with PD1) and BRAF/MEKi have activity7–9, while those who initially respond to PD1 and then progress while off therapy may also benefit from re-treatment with PD1 therapy10,11. Whether the efficacy of systemic treatment for patients who recur following adjuvant PD1 therapy is similar to those who progress after PD1 for advanced melanoma is unknown. It is also unknown whether the success of systemic therapy at recurrence is different in patients who recur during (ON) adjuvant PD1 therapy compared to those who recur following (OFF) therapy.
The aim of this study was to describe the nature of recurrence, management and outcomes of patients who recurred ON or OFF adjuvant PD1 therapy for resected melanoma.
Methods
With local institutional board approval, data were collected retrospectively from 16 international sites. Between March 2015 and December 2018, all patients with resected stage III or IV melanoma who received at least one dose of adjuvant PD1 and had melanoma recurrence were included; new primary melanomas were not considered a recurrence. Patients who received adjuvant PD1 therapy on a clinical trial or as standard care were included, as were those who received combination adjuvant nivolumab with ipilimumab4,5 (NCT03068455). The proportion of recurrence was estimated from the total number of patients that commenced adjuvant PD1 therapy for resected melanoma at all sites by data submission at 31st December 2018. The denominator included 214 patients who remained blinded on 1:1 randomised trials, where only one arm included a PD1 agent, which was halved to estimate actual number receiving adjuvant PD1.
Clinical data regarding disease characteristics prior to adjuvant therapy, adjuvant treatments received, toxicity, timing and pattern of recurrences, method of detection, subsequent management and patient outcomes were collected. Patients had 12-weekly CT surveillance including brain imaging (CT or MRI) and medical oncology review for at least 24 months from start of adjuvant therapy or until recurrence. Regional skin, subcutaneous or nodal metastases were reported as locoregional recurrence. Any distant or visceral metastases were reported as distant recurrence, including recurrence at a previously resected distant site. Investigators determined whether recurrence was primarily detected by symptoms, clinical examination or imaging. Patients were divided into those who recurred ON adjuvant PD1 therapy (recurred while receiving adjuvant PD1 or within 1 month of last dose of therapy) and those who recurred OFF adjuvant PD1 (greater than 1 month after last dose of adjuvant PD1). Clinical outcomes after subsequent systemic therapies were assessed using investigator-determined best response according to RECIST 1.1 (unconfirmed), progression-free survival (PFS) and overall survival (OS). Efficacy data (ORR/PFS) were reported by therapy type, regardless of line of therapy, and OS data was reported from first line of systemic therapy at recurrence. ORR was calculated in evaluable patients; patients who had recently commenced therapy <12 weeks, without response assessment and without clinical signs of progression, were recorded as non-evaluable and were excluded from the denominator for ORR. Patients with cutaneous, acral or unknown primary melanoma, were analysed together and are henceforth termed cutaneous. Patients with mucosal melanoma were analysed independently given the unique biology and inferior outcomes in the metastatic setting12.
Descriptive statistics were used (stratified by initial site of recurrence or treatment at recurrence, where appropriate), except for time-to-event outcomes (OS, PFS, RFS and DMFS), which were analysed using the Kaplan-Meier method, with the log-rank test used to examine differences between subgroups. Systemic therapy responses were compared descriptively in subgroups and the exact binomial test was used to examine differences between them.
Results
Patient Characteristics and Adjuvant Therapy
From an estimated total of 850 patients treated with adjuvant PD1 therapy, 147 (17%) had melanoma recurrence during or following adjuvant PD1-based therapy (Figure S1). Those who recurred had received adjuvant nivolumab (67 patients, 46%), pembrolizumab (40 patients, 27%), nivolumab with “low dose” ipilimumab 1mg/kg every six weeks (18 patients, 12%) or nivolumab +/− “low dose” ipilimumab on a blinded trial (22 patients, 15%). Median time from starting PD1 to last follow-up was 13.3 months (range 1.4–42.3). Median time from 1st recurrence to last follow-up was 7.7 months (range 0.2–33.6).
The majority of patients had cutaneous melanoma (n=136, 93%), and eleven had mucosal melanoma (analysed separately). Most patients (119 patients, 88%) had resected stage III melanoma, 97 (71%) of whom underwent completion lymph node dissection surgery (CLND) and 17 (13%) had resected stage IV melanoma.
Timing and Nature of Initial Recurrence
Median time to first recurrence from starting adjuvant PD1 was 4.6 months (range 0.3–35.7 months). Most patients recurred ON adjuvant PD1 (104 patients, 76%), at a median 3.2 months. Of the 32 patients (24%) who recurred OFF adjuvant PD1, median time to recurrence was 12.5 months. 21/32 patients (66%) had discontinued adjuvant PD1 early for toxicity after a median 2.3 months (range 0.4–11.3), ten completed one year of adjuvant PD1, and one withdrew consent to continuing adjuvant PD1 after one month. In those who recurred OFF adjuvant PD1, median time to recurrence from ceasing adjuvant PD1 was 5.5 months (range 1.0–24.2).
Across the cutaneous cohorts (n=136), initial recurrences were locoregional alone in 59 (43%), distant alone in 55 (40%), and concurrent locoregional and distant recurrence in 22 (16%) (Table S1).
In the cohort with stage III melanoma at baseline (n=119), 60 (50.4%) developed distant metastases at initial recurrence, including 12 (20%) with brain metastases (Figure 1A–C). Distant metastases were more frequent in those with macroscopic versus microscopic nodal disease at baseline (p=0.04, table S1) and in older patients (median age 59 versus 54 in those with only locoregional metastases, p=0.01). Of the 52 patients with microscopic nodal disease identified on sentinel node biopsy who recurred, 39 had a prior CLND of whom 15 (38%) recurred in the nodal basin, and 13 did not have a prior CLND of whom six (46%) recurred in the nodal basin.
Figure 1.
Pattern of initial recurrence in 119 patients with resected stage III at baseline in A), including primary method of detection (n=119) in B) and organ site in those with distant metastases at initial recurrence (n=60) in C), classified by most advanced site (by AJCC M category) in those with multiple sites.
*all patients had CT staging and brain imaging at recurrence
The method of detection of first recurrence differed by the pattern and type of recurrence in patients with resected stage III at baseline (Figure 1B). Most distant recurrences were identified solely on imaging (65%; 39/60 patients), especially in those without concurrent locoregional recurrence (78%, 32/41 patients).
Of the 59 patients who recurred initially with only locoregional metastases, 22 (37%) later developed distant metastases, at a median follow-up of 8.3 months [range 1–31] from first recurrence. At the end of the study period, of the patients with stage III melanoma at baseline, 82 patients had stage IV disease (69%) and 37 still had stage III disease (31%).
Management of Resectable Locoregional Recurrence
Across the cutaneous stage III and IV cohorts (n=136), at first recurrence, 59 patients had locoregional recurrence alone, and 48 (81%) of these were resectable. All patients with resectable locoregional recurrence had surgery, either alone (29, 49%), with adjuvant PD1 (8, 14%), with adjuvant radiotherapy (12, 20%) or with adjuvant BRAF/MEKi (4, 7%) (Figure 2). Median follow-up from resectable locoregional recurrence was 8.3 months [range 1–31] and 27/48 (56%) resected patients had further recurrence, with distant metastases in 18 (38%). Of four patients treated with adjuvant BRAF/MEKi following initial recurrence, none have yet recurred at a median follow-up of 6.5 months (range 0–8).
Figure 2.
Management of resectable locoregional recurrence (n= 48), with individual patients grouped according to treatment at recurrence (Y axis), showing follow-up in months after surgery for initial recurrence (X axis), and subsequent locoregional or distant relapse or death according to key
*3 pts had adjuvant PD1 and adjuvant RT
‡2 pts had adjuvant BRAF/MEK and adjuvant RT
Management of Distant and Unresectable Locoregional Recurrence
In total, 108 (79%) patients developed unresectable locoregional or distant disease during the study. At data cut, median OS from unresectable locoregional or distant recurrence was 21.3 months (95% CI 12.3-NR) and 26 patients (24%) had died, at a median follow-up of 6.5 months (range 0–31).
Of 105 patients that received treatment, 35 (33%) had ipilimumab-based therapy first line (10 had monotherapy, 25 had combination with PD1), 32 (31%) had BRAF/MEKi, 16 (15%) had PD1 alone or in combination with an investigational agent (11 PD1 monotherapy, 5 on an PD1/L1-based combination trial), and 22 (21%) had local therapy for distant recurrence to render the patient free of measurable disease. Differences were observed between the treatment groups (Table S2; supplementary results S1).
Median follow-up from start of systemic therapy for recurrence ranged from 5.5–8.4 months, depending on treatment group (Table 1). The overall response rate to ipilimumab-based therapy was 26%, 10/38 (95% CI 14–43%, 3 CRs), and to BRAF/MEKi was 82%, 27/33 (95% CI 64–93%, 12 CRs). The 6-months PFS was 40% and 70%, respectively (Figure 3A). Ipilimumab-based therapy had efficacy in those who recurred ON adjuvant therapy (8/33, 24% [Table S3]) and in those who recurred OFF adjuvant therapy (2/5, 40%), as did BRAF/MEKi (18/23, 78% ON therapy, 9/10, 90% OFF therapy), with similar PFS in those who recurred ON vs OFF adjuvant therapy for ipilimumab and BRAF/MEKi (Figure 3B–C). Ipilimumab activity appeared similar whether used as monotherapy or combined with PD1 (supplementary results S2).
Table 1.
Responses to first-line and subsequent systemic therapies for unresectable locoregional or distant recurrence
Ipilimumab (+/−PD1) | BRAF/MEKi | PD1 monotherapy | PD(L)1 + novel agent** | |
---|---|---|---|---|
| ||||
N | 44* | 40† | 14 | 11 |
Recurred ON PD1 | 38 | 27 | 9 | 10 |
Recurred OFF PD1 | 6 | 13 | 5 | 1 |
| ||||
Median F/U (months) | 8.4 | 5.5 | 8.4 | 6.4 |
| ||||
ORR, % (95% CI)$ | ||||
Total | 26 (14–43) | 82 (64–93) | 18 (2–52) | 11 (0–48) |
Recurred ON PD1 | 24 (12–43) | 78 (56–93) | 0 (0–46) | 13 (0–53) |
Recurred OFF PD1 | 40 (5–85) | 90 (55–100) | 40 (5–85) | 0(0–96) |
| ||||
6-month PFS, % (95% CI) | 40 (26 – 58) | 70 (52 – 93) | 26 (10 – 68) | 18 (3 – 93) |
| ||||
Median OS‡ (months, 95% CI) | 21.3 (17.6-NR) | 12.3 (8.7-NR) | NR | 5.5 (4.2 - NR) |
Abbreviations: PD(L)1, anti-programmed cell death (Ligand) 1 antibodies
includes one who was treated on a clinical trial with ipilimumab, nivolumab and IDO-inhibitor and one treated with ipilimumab + TLR9-agonist.
38/40 patients had dabrafenib and trametinib, 1 patient had vemurafenib and cobimetinib, 1 patient had encorafenib and binimetinib.
PD1 + novel agents included PD1/-LAG3, -PDL1/MEKi, PD1/TLR9-agonist and PD1/IDOi.
95% CI for ORR are based on exact binomial test.
Efficacy data (ORR/PFS) by drug in total cohort regardless of line of therapy. ORR reported for each systemic agent and in those who recurred during or following adjuvant PD1.
OS reported for 1st line therapy only
Figure 3.
Progression-free survival for standard systemic therapies at recurrence (1st line and subsequent therapies, n=98) A) and by systemic therapy, according to recurrence ON or OFF adjuvant PD1 (B-D)
Notably, there were no responses (0/6) to PD1 therapy in patients who recurred ON adjuvant PD1, while 2/5 (40%) patients who recurred OFF adjuvant PD1 responded to retreatment PD1 therapy at recurrence. Re-treatment of patients who recurred ON adjuvant PD1 with PD1 was associated with inferior PFS than those OFF adjuvant PD1 (median 2.3mo vs NR, Figure 3D). The two patients who recurred OFF treatment and responded to re-treatment with PD1 had completed the one-year standard adjuvant treatment, recurred 5.6 and 13.5 months following PD1 cessation, and remain in response on PD1 after 10.3 and 5.4 months of treatment, respectively.
In 25 patients who underwent definitive local therapy for distant recurrence without systemic therapy (as first line or subsequent treatment), median follow-up from local therapy was 16.7 months [range 0–26], and 17 (68%) have progressed.
Mucosal Melanoma Recurrence
Eleven patients with primary mucosal melanoma recurred following adjuvant PD1 therapy. Prior to adjuvant PD1, five patients had resected stage IV melanoma and six had resected stage III. One patient had a BRAFV600E mutation. Median time to recurrence from starting adjuvant PD1 therapy was 3.1 months (range 0.4–7.9), 10/11 (91%) recurred ON adjuvant PD1 and one recurred OFF adjuvant PD1, having discontinued for toxicity after 2.7 months. All eleven patients had distant metastases at recurrence, including four with concurrent locoregional metastases. At recurrence, seven patients received ipilimumab-based therapy; all with evaluable responses (5/5) had progressive disease. Of the remaining four, two had palliative radiotherapy alone, one patient had a partial response with BRAF/MEKi and one patient had progressive disease on PD1 monotherapy. Median follow-up from initial recurrence was 4.8 months (range 0.5–33.6 months); three patients have died, with estimated median OS from initial recurrence 9.2 months (95% CI 6.9-NR).
Discussion
The nature and best management of patients with melanoma who recur early, either during (ON) or following (OFF) adjuvant PD1 therapy, has not been reported to date and is challenging, as most landmark trials have been performed in systemic treatment-naïve patients. Given the extraordinary impact PD1 therapy has had across a range of advanced-stage cancers, these results in the adjuvant setting in melanoma will likely have broad implications for other solid tumours likely soon to be treated with adjuvant PD1. This multicentre, retrospective analysis of 147 patients showed that those who recur locoregionally and have local therapy often develop distant recurrence shortly thereafter. There was a lack of activity of PD1 monotherapy in those who recur ON adjuvant PD1, while further PD1 monotherapy may have activity in those who recur OFF adjuvant PD1.
In patients with melanoma recurrence despite adjuvant PD1, systemic therapy can be active, but response rates appear to vary by drug class and whether patients recur ON or OFF therapy. Ipilimumab-based immunotherapy and BRAF/MEKi are most active, with response rates of 26% and 82%, respectively. Ipilimumab activity was comparable to both that observed in advanced melanoma after PD1 progression, and that of single-agent ipilimumab in the PD1-naïve setting1,2, suggesting that PD1 resistance may not necessarily confer CTLA-4 resistance7. In our study, a limited number of patients had also received prior anti-CTLA4 (low dose and 8-weekly) in combination with PD1 as adjuvant therapy, and higher doses of ipilimumab may be important for response, especially given the dose-response relationship observed in the metastatic setting13. Response rates to ipilimumab alone or in combination with PD1 were similar in this study, whereas retrospective data in advanced melanoma suggests higher activity of combination therapy than single-agent ipilimumab following PD1 progression8. Prospective data are needed (NCT03179436, NCT03033576).
For patients with BRAFV600-mutant melanoma, BRAF/MEKi are an alternative subsequent therapy, with a high response rate (82%), however durable survival will likely be achieved only in a small subgroup of patients given data from metastatic trials14. For patients who recur OFF adjuvant PD1, re-challenge with single-agent PD1 therapy may be considered, however the small numbers of patients treated with this approach prevent the identification of a specific window period between cessation, recurrence and retreatment where this is effective. A few patients who recurred ON PD-1 continued PD-1 monotherapy at recurrence and predictably none responded; these cases had mostly recurred very early on adjuvant therapy, and PD-1 therapy was likely continued in case of an emergent delayed response. The small group of patients with mucosal melanoma included did not benefit from subsequent therapies (most often CTLA-4 blockade). This supports genomic data indicating that mucosal melanoma is a different from cutaneous melanoma, and remains an area of high unmet clinical need15,16.
In our study, 50% of the recurrences in patients with stage III melanoma were distant at first relapse, which is consistent with historical data in the era before effective adjuvant therapies17, and clinical trial data of PD1 and BRAF/MEK therapies4–6. The most common sites of initial distant recurrence were lung, liver and brain. The rate of brain metastases at initial recurrence (20%) appears higher than that reported in the era pre-effective systemic therapies17, however this is likely attributable to changes in clinical practice, whereby regular surveillance brain imaging is now routine, resulting in a higher rate of detection of asymptomatic brain metastases. Consistent with historical data17, most distant recurrences were identified on surveillance imaging, whereas most locoregional recurrences were clinically evident. Therefore, regular surveillance imaging (including brain imaging) remains worthwhile to detect distant recurrence prior to symptoms developing, particularly within 12 months, during which 88% of recurrences occurred in our study.
Most patients who recurred locoregionally underwent resection. Despite this, there was a high rate of subsequent relapse both locoregionally and distantly. These data suggest that systemic therapy is not only required after distant recurrence, but also after resected locoregional recurrence. Of note, no subsequent relapses were observed in four patients treated with BRAF/MEKi; however, follow-up is limited (median 6.5 months). In the COMBI-AD trial, recurrence was uncommon in the first 12 months while patients were on dabrafenib and trametinib, but sharply increased in the first year off therapy6.
The majority of patients with micrometastatic nodal involvement underwent CLND prior to adjuvant PD1 in our study, which is no longer standard practice following the DeCOG-SLT and MSLT-II trials18,19. This change of practice may impact contemporary recurrence patterns; locoregional recurrence in the nodal basin may become more frequent as most patients no longer undergo CLND, although we saw no difference in nodal recurrence between those who did or did not undergo CLND in our study with short follow up.
The majority of patients in this study recurred early, ON adjuvant PD1 therapy, and follow-up post-recurrence is relatively short (median 7.7 months). Similarly, some patients had short follow-up from commencement of salvage therapy such that robust response assessments could not be made. As such, this study largely represents early recurrence, which likely shares similar resistance mechanisms to those seen with primary resistance in the metastatic setting. Melanoma that recurs later, for example, many years following adjuvant therapy, may have a different biology and response to systemic treatment. Given the short follow-up and minority of recurrences OFF adjuvant PD1 (many of which were still within a few months of cessation of therapy, and due to long receptor binding time, could still be considered similar to those ON therapy), it will be particularly important to study those with late recurrence (beyond one year), especially since our data suggest that clinical activity of systemic therapies in this setting, particularly retreatment PD1, may be clinically relevant.
In conclusion, this is the first study exploring the nature and management of recurrence during or following adjuvant PD1 therapy in melanoma. With adjuvant PD1 therapy being a standard of care for high-risk resected melanoma, and now being tested in trials across a range of cancers, these data are crucial to guiding clinical management. The poor outcome of patients who recur ON adjuvant PD1 identifies a patient group in great need of new therapeutic options. Clinical trials in advanced melanoma should not exclude this growing group of patients. Furthermore, these data serve as an important framework as adjuvant PD1 therapy is introduced more broadly across oncology.
Supplementary Material
Highlights.
Melanoma recurrence occurred in 147 (17%) of 850 patients, after a median 4.6 months after starting adjuvant PD1
At initial recurrence, 57% had distant metastases, mostly detected on imaging while asymptomatic
38% of patients with initial local recurrence then recurred distantly within short follow-up (median 8.3 months)
PD1 monotherapy was not active in those who recurred ON adjuvant PD1, but had activity in those who recurred OFF adjuvant PD1
BRAF/MEK inhibitors and ipilimumab (alone or in combination with PD1) had the highest activity in this setting
Acknowledgments;
The authors wish to thank the patients and their families, the investigators, and the site personnel who participated in this study. AMM is supported by a Cancer Institute NSW Fellowship.
Declaration of interests;
Carina N. Owen reports non-financial support from Merck Sharp Dohme, outside the submitted work.
Alexander N Shoushtari reports grants and personal fees from Bristol-Myers Squibb, during the conduct of the study; grants and personal fees from Immunocore, grants from Xcovery, grants, personal fees and non-financial support from Castle Biosciences, outside the submitted work.
Dharmisha Chauhan reports personal fees from BMS, personal fees from Novartis, outside the submitted work.
David James Palmieri has nothing to disclose.
Belinda Lee has nothing to disclose.
Maartje W. Rohaan has nothing to disclose.
Joanna Mangana reports other from Merck/Pfizer, other from MSD, other from Pierre Fabre, grants and other from Ultrasun, L Oreal, Pierre Fabre, MSD, BMS, outside the submitted work.
Victoria Atkinson reports personal fees and non-financial support from BMS, personal fees from MSD, personal fees from Novartis, personal fees and non-financial support from Pierre fabre, personal fees from Merck Serono, personal fees from Roche, non-financial support from Onco-sec, outside the submitted work.
Farzana Zaman has nothing to disclose.
Arissa Young reports grants from National Heart, Lung, And Blood Institute of the National Institutes of Health under Award Number R38HL143619.
Christoph Hoeller reports personal fees from Amgen, personal fees from BMS, personal fees from Inzyte, personal fees from MSD, personal fees from Novartis, personal fees from Pierre Fabre, personal fees from Roche, outside the submitted work.
Peter Hersey has nothing to disclose.
Reinhard Dummer has intermittent, project focused consulting and/or advisory relationships with Novartis, Merck Sharp & Dhome (MSD), Bristol-Myers Squibb (BMS), Roche, Amgen, Takeda, Pierre Fabre, Sun Pharma, Sanofi, Catalym, Second Genome outside the submitted work.
Muhammad A Khattak has nothing to disclose.
Michael Millward reports personal fees and non-financial support from Merck Sharp & Dohme, personal fees and non-financial support from Bxristol Myers squibb, personal fees and non-financial support from AstraZeneca, personal fees and non-financial support from Roche, personal fees from Novartis, outside the submitted work.
Sapna P Patel reports personal fees from Merck & Co, Incyte, Castle Biosciences, and Cardinal Health, and institutional clinical trial support from Provectus, Ideaya, and Bristol Myers Squibb outside the submitted work.
Andrew Haydon reports personal fees from BMS, personal fees from Pierre Fabre, personal fees from Merck, outside the submitted work.
Douglas B. Johnson reports other from Array Biopharma, grants and other from BMS, grants and other from Incyte, other from Merck, other from Novartis, other from Genentech, outside the submitted work.
Serigne Lo has nothing to disclose.
Christian U. Blank reports personal fees from BMS, MSD, GSK, Lilly, Roche, Novartis, Pfizer, GenMab, AZ, Pierre Fabre, grants from BMS, NanoString, Novartis, outside the submitted work; and stockownership Unitit Cars, Neon Therapeutics.
Shahneen Sandhu reports grants and other from Merck Sharp and Dohme, grants and other from Bristol Myer Squibb, grants from Amgen, grants from Endocyte, grants from Astra Zeneca, other from Roche, outside the submitted work.
Matteo S. Carlino reports personal fees from BMS, personal fees from MSD, personal fees from Novartis, personal fees from Roche, personal fees from Amgen, personal fees from Pierre Fabre, personal fees from Ideaya, outside the submitted work.
James M. G. Larkin reports grants and personal fees from Achilles therapeutics, personal fees from AstraZeneca, personal fees from Boston Biomedical, grants and personal fees from BMS, personal fees from Eisai, personal fees from EUSA Pharma, personal fees from GSK, personal fees from Ipsen, personal fees from Imugene, personal fees from Incyte, personal fees from iOnctura, personal fees from Kymab, personal fees from Merck Sorono, grants and personal fees from MSD, grants and personal fees from Nektar, grants and personal fees from Novartis, personal fees from Pierre Fabre, grants and personal fees from Pfizer, grants and personal fees from Roche/Genetech, personal fees from Secarna, personal fees from Vitaccess, personal fees from Covance, grants and personal fees from Immunocore, grants from Aveo, grants from Pharmacyclics, outside the submitted work.
Alexander M. Menzies reports personal fees from BMS, personal fees from MSD, personal fees from Novartis, personal fees from Roche, personal fees from Pierre-Fabre, outside the submitted work.
Georgina V. Long is a Consultant Advisor and reports the following outside of the submitted work; personal fees from Aduro, personal fees from Amgen, personal fees from Array, personal fees from Bristol-Myers Squibb, personal fees from Merck Sharp Dohme, personal fees from Novartis, personal fees from Pierre-Fabre, personal fees from Oncosec, personal fees from Roche, personal fees from Sandoz.
Footnotes
Ethics committee approval;
All participating sites have local Human Research Ethics Committee approval.
Previous presentation;
American Society of Clinical Oncology Annual Meeting 2019. J Clin Oncol 2019 37:15_suppl, 9502–9502
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Larkin J. et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med 373, 23–34 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Robert C. et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med 372, 2521–2532 (2015). [DOI] [PubMed] [Google Scholar]
- 3.Long GV et al. Combined BRAF and MEK Inhibition versus BRAF Inhibition Alone in Melanoma. N. Engl. J. Med 371, 1877–1888 (2014). [DOI] [PubMed] [Google Scholar]
- 4.Weber J. et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N. Engl. J. Med 377, 1824–1835 (2017). [DOI] [PubMed] [Google Scholar]
- 5.Eggermont AMM et al. Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. N. Engl. J. Med 378, 1789–1801 (2018). [DOI] [PubMed] [Google Scholar]
- 6.Long GV et al. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF -Mutated Melanoma. N. Engl. J. Med 377, 1813–1823 (2017). [DOI] [PubMed] [Google Scholar]
- 7.Zimmer L. et al. Ipilimumab alone or in combination with nivolumab after progression on anti-PD-1 therapy in advanced melanoma. Eur. J. Cancer 75, 47–55 (2017). [DOI] [PubMed] [Google Scholar]
- 8.Weichenthal M. et al. Salvage therapy after failure from anti-PD-1 single agent treatment: A Study by the German ADOReg melanoma registry. J. Clin. Oncol 37, 9505–9505 (2019). [Google Scholar]
- 9.Johnson DB et al. Sequencing Treatment in BRAFV600 Mutant Melanoma: Anti-PD-1 Before and After BRAF Inhibition. J Immunother 40, 5 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jansen YJL et al. Discontinuation of anti-PD-1 antibody therapy in the absence of disease progression or treatment limiting toxicity: clinical outcomes in advanced melanoma. Ann. Oncol (2019) doi: 10.1093/annonc/mdz110. [DOI] [PubMed] [Google Scholar]
- 11.Warner AB et al. Long-Term Outcomes and Responses to Retreatment in Patients With Melanoma Treated With PD-1 Blockade. J. Clin. Oncol (2020) doi: 10.1200/JCO.19.01464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.D’Angelo SP et al. Efficacy and Safety of Nivolumab Alone or in Combination With Ipilimumab in Patients With Mucosal Melanoma: A Pooled Analysis. J. Clin. Oncol (2016) doi: 10.1200/JCO.2016.67.9258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ascierto PA et al. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 18, 611–622 (2017). [DOI] [PubMed] [Google Scholar]
- 14.Robert C. et al. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N. Engl. J. Med (2019) doi: 10.1056/NEJMoa1904059. [DOI] [PubMed] [Google Scholar]
- 15.Hayward NK et al. Whole-genome landscapes of major melanoma subtypes. Nature 545, 175–180 (2017). [DOI] [PubMed] [Google Scholar]
- 16.Newell F. et al. Whole-genome landscape of mucosal melanoma reveals diverse drivers and therapeutic targets. Nat. Commun 10, (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Romano E, Scordo M, Dusza SW, Coit DG & Chapman PB Site and Timing of First Relapse in Stage III Melanoma Patients: Implications for Follow-Up Guidelines. J. Clin.Oncol (2010) doi: 10.1200/JCO.2009.26.2063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Leiter U. et al. Complete lymph node dissection versus no dissection in patients with sentinel lymph node biopsy positive melanoma (DeCOG-SLT): a multicentre, randomised, phase 3 trial.Lancet Oncol. 17, 757–767 (2016). [DOI] [PubMed] [Google Scholar]
- 19.Faries MB et al. Completion Dissection or Observation for Sentinel-Node Metastasis in Melanoma. N. Engl. J. Med 376, 2211–2222 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.