Skip to main content
UKPMC Funders Author Manuscripts logoLink to UKPMC Funders Author Manuscripts
. Author manuscript; available in PMC: 2026 Jan 20.
Published in final edited form as: Eur J Cancer. 2024 Dec 13;215:115160. doi: 10.1016/j.ejca.2024.115160

A comparison of real-world data on adjuvant treatment in patients with stage III BRAF V600 mutated melanoma – Results of systematic literature research

Teresa Amaral a,b,*, Lena Nanz a,#, Lina Maria Serna Higuita c,#, Paolo Ascierto d, Carola Berking e, Eva Muñoz Couselo f, Marco Donia g, Reinhard Dummer h, Ralf Gutzmer i, Axel Haushild j, Mathilde Jalving k, Rebecca Lee l,m, Paul Lorigan l,m, Ivan Marquez-Rodas n, Olivier Michelin o, Paul Nathan p, Caroline Robert q, Dirk Schadendorf r, Pawel Sobczuk s, Lukas Flatz a,b, Ulrike Leiter a, Claus Garbe a
PMCID: PMC7618644  EMSID: EMS212048  PMID: 39673834

Abstract

Background

Over the past decade, PD-1-based immune checkpoint inhibitors (ICI) and targeted therapies (TT) with BRAF and MEK inhibitors transformed melanoma treatment. Both are widely used in the adjuvant setting. However, for patients with a BRAF V600 mutation, the optimal adjuvant therapy remains unclear due to the lack of head-to-head comparison studies.

Methods

We conducted a systematic review of real-world data on adjuvant therapy in stage III melanoma to determine the best option for patients with BRAF V600 mutations. Kaplan-Meier curves were generated for TT and ICI using Digitizelt software.

Results

Nine publications with 3625 patients were included. TT showed better relapse-free survival (RFS) at 6, 12, 24, and 36 months than ICI. A similar trend was observed for distant metastasis-free survival (DMFS), with no apparent difference in overall survival.

Conclusion

Real-world data suggest that adjuvant TT may be associated with better RFS and DMFS in stage III BRAF V600-mutated melanoma compared to ICI.

Keywords: Adjuvant therapy, Targeted therapy, Checkpoint inhibitors, Stage III, Relapse-free survival, Distant metastasis-free survival, Overall survival

1. Introduction

Over the last decade, two important strategies have been developed for the systemic treatment of melanoma: PD-1 based immune checkpoint inhibition (ICI) and targeted therapy (TT) with BRAF and MEK in-hibitors. The latter is only applicable if a BRAF V600 mutation is present, with only a few exceptions. For the 40–50 % of patients with melanoma having this mutation, both ICI and TT can be offered. This raised the question of which therapy should be used in first line.

For patients with unresectable stage IV disease, clinical trials comparing ICI versus TT in first line were conducted, and the results reveal that patients receiving a combination of ICI in first line have a better outcome, with TT being given in second line. [1,2] In resected stage III melanoma, both therapeutic strategies including nivolumab or pembrolizumab (ICI) and dabrafenib plus trametinib (TT) have been approved for adjuvant treatment. [35] However, in this setting, there have been no comparative, head-to-head randomized clinical trials to date, and they are likewise not expected. On the other hand, a substantial body of real-world data on adjuvant treatment outcomes for patients with stage III melanoma is now available.

We conducted a systematic literature review of real-world data on adjuvant treatment for stage III melanoma. Our objective was to determine whether real-world data can address which adjuvant therapy offers the best survival outcomes, namely relapse-free survival (RFS), distant metastases free survival (DMFS) and overall survival (OS), for patients with a BRAF V600 mutation.

2. Methods

2.1. Search strategy and selection criteria

We searched PubMed from Jan 1, 2012, to Feb 29, 2024, with the following two algorithms: 1) “Melanoma [ti] AND adjuvant [ti] AND real world [ti]”; and 2) “Melanoma [ti] AND adjuvant [ti] AND real world”. We also searched relevant articles referenced by other publications and abstracts from clinical meetings held in this time period. All publications selected were available either as full text publications or as conference abstracts. Publications that did not include survival outcomes of TT or ICI in patients with BRAF V600 mutation were excluded. A summary of the publications included in the analysis is provided in Table 1.

Table 1. Publications (n = 9) included in the survival analysis.

Publication Analysis interval Number of patients included and treated with systemic therapy (TT; ICI) Number of patients with BRAF mut FUP RFS D+T HR RFS (95 % CI) p value RFSICI HR RFS (95 % CI) p value DMFS D+T DMFSICI Comment
van Laar, 2022[8] 01.2019 to 10.2021 122(20/102) 65 Max FUP 13 months 1y RFS 83.0 % NR 1y RFS nivo 70.3 %, 1y RFS Pembro 72.4 % NR NR NR 20 patients with Stage IV NED included. No information about subtype of V600 mutation
Bai, 2023 [16] 07.2015 to 10.2022 598 (393/205) 598 (83.8 % BRAF V600E; 9,5 % BRAF V600K; 1.3 % other; 5.3 % unknown) mFUP 33 months (IQR 21–43); TT 29 months [IQR 18–40], PD–1 38 months [IQR 29–50]) 51.0 months (95 % CI 41.0-not reached) Univariate: HR 0.66 (95 % CI 0.50–0.87); P = 0.003; multivariate: HR 0.58, (95 % CI 0.39–0.86); P = 0.007 44.8 months [95 % CI 28.5-NR] Univariate: HR 0.66 (95 % CI 0.50–0.87) P = 0.003; multivariate: HR 0.58 (95 % CI 0.39–0.86) P = 0.007 NR NR OS multivariate, HR 0.90, (95 % CI 0.48–1.70), P = 0.75. 2-yr restricted mean survival time for TT 21.8 months and 19 months for PD–1 (P < 0.001).
De Falco, 2023[9] Data cut-off for analysis was 31/5/2022 113 (58/55) 75 Total FUP 20.2 months NR 0.36 (0.16–0.77) p = 0.010 NR NR NR NR No information about subtype of V600 mutation
Haist 2023 [12] 01.2014 to 07.2022 515 (242/273) 237 (73 % -BRAF V600E; 12.2 % BRAF V600K; 3.3 % BRAF V600D/R) mFUP 27 months (22.3 to 31.7) mRFS 11 (8.7 to 13.3) Adjusted HR 0.52 (95 % CI 0.40 to 0.68) p < 0.001 mRFS 6 months (4.0 to 8.0) mDMFS 15 months (11.5 to 18.5) mDMFS 12 months (8.5 to 15.4) 3y OS rate TT 87.4 %;PD–180.5 %
Lodde 2023[13] 06.2018 to 09.2019 589 (110/479) 232 (85 % BRAF V600E; 9 % BRAF V600K; 5 % Other BRAF mutations; 1 % unknonw) mFUP 25.7 months 2y RFS 67 % NR 2y RFS 49% HR 1.99; (95 % CI 1.34–2.96) NR NR 2y MSS TT 92 %; PD–1 87 %
Placzke 2023; [11] 02.2019 to 01.2021 248 (101/147) 155 mFUP 13.9 months 2y RFS 65.9 % NR 2y RFS 56.1 % NR 2y DMFS 76.5 % 2y DMFS 64.8 % 23 patients with Stage IV NED included.OS TT 87.8 %; PD–1 85.3 %. No information about subtype of V600 mutation
Schumann 2023[14] 01.2017 to 10.2021 1198 (195/1003) 542 (78.8% BRAF V600E; 9,2% BRAF V600K; 12% Other BRAF mutations) mFUP 17 months 1y RFS 86.5 %; 2y RFS 78.1 % NR 1y RFS 78.1 %; 2y RFS 67.9 HR 1y 1.998 (95 % CI 1.335–2.991); p = 0.001 NR NR 105 patients with Stage IV NED included.2y OS TT 95.3 %; PD–1: 93.1 %
Zhong, 2023[10] 01. 2017 to 12.2021 93 (25/25) 93 (100 % BRAF V600E) mFUP 11months for TT and 22 months for PD–1 mRFS not reached1y RFS 81.7 %; 2y RFS 58.1 % NR mRFS 15 months1y RFS 59.0 %; 2y RFS 54.2 % NR Not reached Not recahed 23 patients were treated with Vemurafenib and 20 did not receive adjuvant therapy
Rigo 2024 [15] 09.2017 to 03.2021 149 (20/129) 74 (49.7 % BRAF V600 E or K) mFUP 22.4 months mRFS 38.4 %; 1y RFS 94 %; 2y RFS 63 % NR mRFS not reached.1y RFS BRAF mut patients 73 %; 2y RFS BRAF mut patients 60 % NR NR NR 7 patients with Stage IV NED included.

HR – Hazard ratio; mFUP – median follow-up; NR – not reported; RFS – relapse free survival; DMFS – distant metastases free survival; MSS – melanoma specific survival; OS – Overall survival; INF – interferon; NA – Not applicable

2.2. Statistical analysis - Kaplan Meier after digitalization

The RFS survival data were obtained by estimating individual data by Kaplan Meier curves in the selected publications. The RFS probabilities at pre-specified time intervals were extracted from the published Kaplan-Meier curves using the software Digitizelt. [6,7] Digital sampling points were created from published Kaplan-Meier survival curves, and digital curves were constructed by linear interpolation between these points. Mean survival curves were then calculated as a weighted average of the survival curve from the studies included in our analysis. The weights applied were in proportion to the number of individuals stratified by therapy (ICI vs. TT). Weighted averaging was calculated pointwise at the sampling points tk from all digitized Kaplan-Meier curves Si(t) using the following formula:

S^(tk)=niS^i(tk)ni

where: Ŝ(tk): represents the value at time point (tk) and ni represents the initial sample size estimation. [6,7] In addition, Kaplan-Meier curves stratified by individual publication were created to evaluate their consistency with the weighted Kaplan-Meier analysis.

We refrained from statistical tests for evaluating differences in survival between TT and ICI, and limited ourselves to descriptive evaluations, as the composition of the populations in the various publications was quite different.

3. Results

3.1. Population included

Data from nine publications with real-world data were included in the analysis, comprising a total of 3625 patients. Median follow up ranged between 11 and 33 months.

Fifty-seven percent of patients (n = 2071) had a BRAFV600 mutation. Fifty-six percent of all BRAF V600 mutated patients (n = 1164) were treated with TT therapy, and 44 % (n = 907) were treated with ICI. [816] Six out of nine publications reported data on the subtype of BRAF V600 mutation of the patients included, with the majority being BRAF V600E, as expected. More data on the subtype of BRAF V600 mutation is available in Table 1 and S1, for the publications that reported this information. The consort diagram can be found in Figure 1.

Fig. 1. Consort diagram.

Fig. 1

3.2. Analysis of survival outcomes

A total of 20 Kaplan Meier curves for RFS were extracted from the above-mentioned 9 publications (n = 3625 patients). Data on DMFS and OS was also extracted as follows: 5 Kaplan Meier curves for DMFS were extracted from 2 publications (n = 608 patients) and 8 Kaplan Meier curves for OS were extracted from 4 publications (n = 2559 patients).

After digitalization, the survival curves were grouped by treatment type, i.e., TT or ICI. Weighted averaging of RFS curves was performed within each therapy strategy group as described above and plotted separately for TT and ICI (Fig. 2A). Patients with BRAF V600 mutation treated with TT seem to have better RFS outcomes than patients treated with ICI. Weighted averaging of DMFS and OS curves was also performed within each therapy strategy group as described above and plotted separately for TT and ICI (Figs. 2B and 2C). We also observed a trend for better DMFS in patients with BRAF V600 mutation treated with TT, although data were reported in only two publications. In terms of OS benefit, there were no obvious differences between TT and ICI, but this conclusion is based on data from only four publications.

Fig. 2A.

Fig. 2A

RFS Kaplan Meier curve for immune checkpoint therapy and targeted therapy extracted from 9 publications identified. [816] Each publication reported data from more than 65 patients with a BRAF mutation. Total number of patients included is 3625.

Fig. 2B.

Fig. 2B

DMFS Kaplan Meier curve for immune checkpoint therapy and targeted therapy extracted from 2 publications [10,12]. Total number of patients included is 608.

Fig. 2C.

Fig. 2C

OS Kaplan Meier curve for immune checkpoint therapy and targeted therapy extracted from 4 publications [11,12,14,16]. Total number of patients included is 2559.

3.3. Survival with targeted therapy compared to immune checkpoint therapy

A key objective of this exploratory analysis was to compare the mean survival curves of patients receiving TT versus ICI.

Averaged survival proportions, specifically the RFS, DMFS and OS rates at 6, 12, 24, and 36 months for TT and ICI, were calculated and are available in Table 2. The RFS rates at the different time points indicated a higher numerical value for TT compared to ICI in patients with BRAF V600 mutation. The 6, 12, 24, and 36 months RFS rates were 95 % vs 82.3 %; 85.8 % vs 68.5 %; 67.4 % vs 54.6 % and 51.9 % vs 48.3 %, respectively for TT and ICI.

Table 2. RFS, DMFS and OS rates for 6, 12, 24 and 36 months.

6 months 12 months 24 months 36 months
%
RFS (9 publications)
ICI 82.9 69.3 55.5 48.9
TT 95.9 88.2 70.4 57.5
DMFS (2 publications)
ICI 83.4 73.9 60 53.9
TT 95.5 84.4 71.4 55.7
OS (4 publications)
ICI 99.0 96.7 89.2 85.2
TT 99.1 97.0 91.0 87.8

3.4. Survival with targeted therapy and immune checkpoint therapy

To further evaluate the concordance between the outcomes of TT and ICI among the different publications, we performed grouping of digitized Kaplan Meier RFS curves, side by side (Figures 3A and 3B). The results showed a high concordance between the different publications within each therapy.

Fig. 3A.

Fig. 3A

RFS Kaplan Meier curve for immune checkpoint therapy extracted the publications identified below, showing highly concordance among the different publications. The publication from Van Laar 2022 [8] is listed twice in the graphic as Nivolumab and Pembrolizumab were plotted separately in the original publication.

Fig. 3B.

Fig. 3B

RFS Kaplan Meier curve for targeted therapy extracted the publications identified below, showing highly concordance among the different publications. The publication from Zhong 2023 [10] is listed twice as Vemurafenib and Dabrafenib + trametinib were plotted separately.

4. Discussion

Our analysis of real-world data on the outcomes of adjuvant therapy in 3625 patients with stage III melanoma suggest that TT may provide better RFS benefit than ICI in patients with BRAF V600 mutation, and this is maintained at 1 and 2 years of follow-up (86 % vs 68 % and 67 % vs 55 %, respectively). The same trend was observed for DMFS, although the number of patients included was lower. There was no obvious difference in terms of OS between both therapies. Longer follow-up is needed to confirm these results.

Currently, based on the results from the NADINA and SWOG S1801 trials [17,18] a therapeutic option for patients with Stage III resectable macroscopic disease is 2 cycles of neoadjuvant ipilimumab plus nivolumab followed by complete surgical resection and adjuvant therapy according to pathological response and BRAF status, or 3 cycles of neoadjuvant pembrolizumab followed by complete surgical resection and 15 cycles of adjuvant pembrolizumab. Still, a significant percentage of patients present with microscopic stage III disease, only detected after complete resection (i.e., sentinel lymph node biopsy). Therefore, adjuvant TT for patients with BRAF V600 mutated melanoma and ICI for BRAF wild-type and BRAF V600 mutated melanoma can be considered. Additionally, the comparator arm in the NADINA study did not include adjuvant TT alone, therefore a direct comparison between neoadjuvant ICI and adjuvant targeted therapy cannot be made at this time point.

Recently, the first OS data in adjuvant stage III melanoma were reported in the COMBI-AD study using dabrafenib plus trametinib. It was shown that for the whole population of patients with BRAF V600-mutated melanoma there was no statistically significant OS benefit of adjuvant TT versus placebo (hazard ratio (HR) for death, 0.80; 95 % confidence interval [CI], 0.62 to 1.01; p = 0.06). [19] However, when BRAF V600 mutation types were analyzed separately, the HR for death for patients with BRAF V600E was 0.75 (95 % CI, 0.58 to 0.96) and for patients with BRAF V600K was 1.95 (95 % CI, 0.84 to 4.50). This means that patients with the more common BRAF V600E mutation experienced a significant improvement in OS with TT, whereas patients with the less frequent BRAF V600K mutation could have a survival disadvantage. However, this was a subgroup analysis, and the results should be interpreted cautiously. The primary endpoint of the COMBI-AD study was RFS which showed a clear benefit for patients receiving adjuvant TT compared to placebo; the median RFS was 93.1 months (95 % CI, 47.9 to not reached) for dabrafenib plus trametinib and 16.6 months (95 % CI, 12.7 to 22.1) for placebo (HR for relapse or death, 0.52; 95 % CI, 0.43 to 0.63). The absence of benefit in OS despite the RFS and DMFS benefit when using adjuvant therapy, also in the real-world setting, may be explained by the use of other systemic therapies, such as immune checkpoint inhibitors, at the time of relapse, which seem to be able to rescue patients in a more advanced, irresectable stage. Specifically in the COMBI-AD study, and for the safety population analysis, 97 of 213 patients receiving placebo were treated with PD-1 based therapies at any time after relapse. This relatively low number may be due to early relapses during the study when PD-1 based therapy was not available outside clinical trials. Also worth mentioning is that the COMBI-AD OS analysis was done with only 260 of the 597 events included in the initial statistical plan, which may have contributed to the absence of OS benefit seen.

The presence of BRAF V600 mutation is a prognostic factor. Indeed, patients with untreated BRAF V600 mutated melanoma have a worse prognosis than patients with BRAF wild-type tumors. [20] However, there seems to be no difference in terms of RFS or DMFS benefit in patients with BRAF V600 mutated compared to wild-type tumors treated with ICI, suggesting that patients with BRAF V600 mutated tumors may have a relative higher benefit. [21].

Currently, there are no OS data reported from the adjuvant trials investigating ICI therapy versus placebo in stage III. There is some uncertainty if the RFS/DMFS benefit in the adjuvant setting, similar to TT, will translate into OS benefit. Indeed, the first OS data from Keynote 054 initially planned to be reported in 2023 have now been postponed to Q4 of 2027. [22] Further studies on adjuvant ICI therapy in resected stage III and IV melanoma, namely Checkmate 238 and SWOG 1404 trials, showed a benefit in terms of RFS but not OS, although in both trials the comparator arms were active treatments, i.e., ipilimumab and α interferon or ipilimumab, respectively, instead of placebo. [23,24].

When discussing adjuvant therapy with patients with melanoma, the toxicity profile of the individual options must be considered. Adverse events associated with TT are generally transient and diminish or resolve with therapy pause, although they may recur when therapy is resumed. Permanent or irreversible toxicities may occur, but are rare. [25] On the other hand, toxicity associated with ICI, depending on the CTCAE grade, may require not only therapy pause but also immuno-suppression. Moreover, some toxicities associated with ICI, such as hypothyroidism, hypophysitis and diabetes mellitus, are irreversible and require lifelong hormone supplementation. [26].

Another aspect that needs to be discussed is the quality of life (QOL) reported in association with TT and ICI. In Keynote 054, a decrease in QOL was reported for patients receiving pembrolizumab compared to placebo, although this difference remained below the clinical relevance threshold. [27] Data from real-world show that adjuvant ICI seems to be associated with a significant decline of QOL at 12 months compared to baseline. [28] Rogiers et al. reported that during ICI adjuvant treatment (at 9 months), the percentage of patients with clinically significant fatigue and cognitive impairment increased compared to baseline. [29] After treatment ended (at 18 months), reports of clinically significant fatigue decreased, but there was an increase in the percentage of patients with clinically significant emotional, cognitive, and social impairment compared to during treatment. Pedersen at al. also reported that adjuvant nivolumab may affect QOL at least temporarily. [30] As for adjuvant TT, the QOL analysis of patients receiving dabrafenib and trametinib in the COMBI-AD study showed that TT did not adversely affect health-related QOL during treatment or in long-term follow-up, in the absence of disease-related symptoms. [31] Important to consider that there are no data specifically looking into the QoL of patients who relapsed on placebo therapy or comparing head-to-head QoL of adjuvant targeted and immunotherapy.

Another aspect to consider when selecting the adjuvant therapy regimen may be the potential development of resistance to systemic therapy in more advanced stages. So far, the data available is scarce. The benefit may be different depending on the type of relapse, i.e., relapse on or off adjuvant therapy. Patients who relapsed after adjuvant TT seem to benefit from subsequent anti-PD-1 based therapy with outcomes similar to first line or therapy naïve stage IV melanoma, i.e., TT did not impacted response to ICI. [32] On the other hand, patients with recurrence on PD-1 adjuvant therapy seem to have marginal benefit from PD-1 monotherapy but may benefit from ipilimumab, alone or in combination with PD-1, and TT. [33] Still, in a real-world setting comparing patients receiving and not receiving adjuvant therapy, there was no OS difference, suggesting that the use of systemic therapies may be able to rescue patients who have a recurrence. [34] More data is necessary here, before drawing definite conclusions.

Our analysis has limitations: 1) This is an indirect comparison using real-world data that is not documented as detailed as in a clinical trial. Capture of RFS events depends on the radiology evaluation schedule, which diverges among countries and centers, contrary to the standardized evaluation in clinical trials. 2) The criteria for including the publications in our analysis were similar (adjuvant therapy in stage III melanoma), but the criteria for including the patients in the different analysis differ between publications. A potential selection bias could be present in this analysis, as some patients’ basal characteristics (i.e. Breslow index, ulceration, number of involved nodes, localization of primary tumor, LDH level, age, lymphocyte counts, performance status, etc.) may be imbalanced in the treatment groups. 3) Patients treated with ICI were both BRAF wild-type and BRAF V600 mutated, as without access to the raw data from the different publications it was not possible to distinguish these populations when extracting the data from the ICI survival curves. 4) Survival data for TT in patients with BRAF V600 mutation included all patients regardless of subtype of BRAF V600 mutation. 5) Our analysis did not allow for comparison between stage III substages (AJCC v8 IIIA-D). 6) The median follow-up is almost three years, but this may still be insufficient to capture all the events in stage III melanoma.7) Four percent of patients included had melanoma stage IV NED. 8) Finally, we didn’t perform statistical tests for evaluating differences in survival between TT and ICI, but rather performed a descriptive analysis, as the composition of the collectives in the various studies was quite different.

On the other hand, our report has advantages: 1) We included data from 3625 patients receiving adjuvant therapy, 2071 of whom harbored a BRAF mutation. 2) Depending on the country of origin, the patients included in this analysis may have had access to PD-1 based therapies, namely nivolumab, pembrolizumab or the combination of ipilimumab and nivolumab, at the time of relapse, i.e., similar to what is offered nowadays. 3) Grouping of digitalized Kaplan Meier RFS curves by therapeutic strategy showed a high concordance between the survival curves within each therapy in the different publications. 4) We used weighted average calculations which improved the detailed analysis.

In the absence of a clinical trial evaluating face-to-face TT versus ICI in the adjuvant setting, an opportunity to address this question with better statistical quality could be to analyze individual patient data from all patients with BRAFV600 mutation included in the COMBI-AD and Keynote 054 studies, matched by basal staging prognostic factors. This is out of the scope of this manuscript and would require the agreement of at least three different parties (Novartis, EORTC and Merck), but it has been possible before. One recent example, where randomized trials investigating two available therapies will be challenging is that of metastatic uveal melanoma, where an individual rather than aggregated patient analysis has been conducted comparing ipilimumab and nivolumab from the Spanish Multidisciplinary Melanoma Group (GEM 1402) [35] with Tebentafusp (IMCgp100–202) [36,37], unveiling an advantage of tebentafusp over ipilimumab and nivolumab. [38].

Further data is needed, especially on 1) biomarker analysis to identify the patients who would benefit the most from adjuvant therapy, 2) on the identification of patients at risk for developing long-term, irreversible toxicities, and 3) on the potential development of resistance in the adjuvant setting that could impair rescue with systemic therapies in the more advanced, irresectable stages. Finally, longer follow-up is needed to confirm the capture of the majority of recurrences in an adjuvant setting, which depends on a non-standardized schedule of radiological evaluation.

5. Conclusion

Here we present the first indirect head-to-head comparison of survival outcomes between adjuvant targeted therapy and immune checkpoint inhibitors in stage III melanoma using real-world data. Our exploratory analysis indicates that targeted therapy may provide longer RFS and DMFS in patients with BRAF V600 mutation, confirming targeted therapy as a beneficial option in this setting.

When deciding on the choice of adjuvant therapy for patients with BRAF mutation in stage III, the following aspects should be taken into consideration when weighing up TT versus ICI: TT shows better RFS data than ICI in the real-world data analysis; TT has a lower toxicity and, in contrast to ICI, it’s not associated with lifelong loss of organ function; clinically meaningful impact in quality of life was found with ICI, but not with TT; and, last but not least, patients may prefer oral to intravenous therapy.

Supplementary Material

Appendix A. Supporting information

Supplementary data associated with this article can be found in the online version at doi:10.1016/j.ejca.2024.115160.

Supplementary Material

Acknowledgements

Not applicable.

Funding

Not applicable.

Footnotes

Ethics approval and consent to participate

Not applicable.

CRediT authorship contribution statement

Lina Serna Higuita: Writing – review & editing, Writing – original draft, Visualization, Validation, Methodology, Formal analysis, Data curation. Ivan Marquez-Rodas: Writing – review & editing, Writing – original draft, Validation. Paolo Ascierto: Writing – review & editing, Writing – original draft, Validation. Olivier Michielin: Writing – review & editing, Writing – original draft, Validation. Carola Berking: Writing – review & editing, Writing – original draft, Validation. Paul Nathan: Writing – review & editing, Writing – original draft, Validation. Eva Muñoz Couselo: Writing – review & editing, Writing – original draft, Validation. Caroline Robert: Writing – review & editing, Writing – original draft, Validation. Paul Lorigan: Writing – review & editing, Writing – original draft, Validation. Mathilde Jalving: Writing – review & editing, Writing – original draft, Validation. Claus Garbe: Writing – review & editing, Writing – original draft, Visualization, Validation, Supervision, Methodology, Investigation, Formal analysis, Data curation, Conceptualization. Teresa Amaral: Writing – review & editing, Writing – original draft, Visualization, Validation, Supervision, Project administration, Methodology, Investigation, Formal analysis, Data curation, Conceptualization. Rebecca Lee: Writing – review & editing, Writing – original draft, Validation. Lena Nanz: Writing – review & editing, Writing – original draft, Formal analysis, Data curation. Marco Donia: Writing – review & editing, Writing – original draft, Validation. Dirk Schadendorf: Writing – review & editing, Writing – original draft, Validation. Reinhard Dummer: Writing – review & editing, Writing – original draft, Validation. Pawel Sobczuk: Writing – review & editing, Writing – original draft, Validation. Ralf Gutzmer: Writing – review & editing, Writing – original draft, Validation. Lukas Flatz: Writing – review & editing, Writing – original draft, Validation. Axel Haushild: Writing – review & editing, Writing – original draft, Validation. Ulrike Leiter: Writing – review & editing, Writing – original draft, Validation.

Declaration of Competing Interest

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: TA reports personal fees for advisory board membership from Delcath and Philogen; personal fees as an invited speaker from Bristol Myers Squibb (BMS), Neracare, Novartis and Pierre Fabre; personal fees for a writing engagement from CeCaVa and Medtrix; institutional fees as local principal investigator (PI) from Agenus Inc., AstraZeneca, BioNTech, BMS, HUYA Bioscience, Immunocore, IO Biotech, MSD, Pfizer, Philogen, Regeneron, Roche and University Hospital Essen; institutional fees as coordinating PI from Unicancer; institutional research grants from iFIT and Novartis; institutional funding from MNI - Naturwissenschaftliches und Medizinisches Institut, Neracare, Novartis, Pascoe, Sanofi and Skyline-Dx; non-remunerated membership of the American Society of Clinical Oncology (ASCO) and the Portuguese Society for Medical Oncology; and a role as clinical expert in the area of medical oncology for Infarmed.LN nothing to report. LMSH nothing to report. PA reports personal fees for advisory board membership from Anaveon, Bayer and BioNTech; personal fees for a consultancy role from Bio-Al Health, BMS, Boehringer Ingelheim, Erasca, Immunocore, Italfarmaco, Lunaphore, Medicenna, Merck Serono, MSD, Nouscom, Novartis, Pfizer, Pfizer/Array, Philogen, Pierre Fabre, Regeneron, Replimmune, Roche Genentech, Sandoz, Sanofi, Sun Pharma and ValoTx; personal fees for an advisory role from BMS, Boehringer Ingelheim, Erasca, Immunocore, Merck Serono, MSD, Novartis, Pfizer/Array, Pierre Fabre, Regeneron, Replimune, Roche, Sandoz, Sanofi and Sun Pharma; institutional funding for clinical trials from BMS, Pfizer/Array, Roche, Genentech and Sanofi; travel grants from Bio-Al Health, MSD, Pfizer/Array and Replimmune; and non-remunerated roles as President of the Campania Society of ImmunoTherapy of Cancer (SCITO) Italy, President of the Fondazione Melanoma Onlus Italy, member of the steering committee for the Society for Melanoma Research (SMR), member of AIOM, ASCO, EORTC Melanoma Cooperative Group, SITC and SMR and member of the Board of Directors for the SITC (November 2017-December 2021). CB reports honoraria for consulting/advisory board and/or speakeŕs fees from Almirall Hermal, BMS, Delcath, Immunocore, Leo Pharma, MSD, Novartis, Pierre Fabre, Regeneron, and Sanofi, outside the submitted work. EMC reports consultant or Advisory Role Bristol Myers Squibb, Merck Sharp & Dohme, Novartis, Pierre Fabre, Roche, Sanofi, Regeneron; Research Funding MSD, Sanofi, BMS; Speaking Amgen, Bristol Myers Squibb, Merck Sharp & Dohme, MSD, Novartis, Pierre Fabre, Regeneron; Clinical trial participation (principal investigator) Amgen, Bristol Myers Squibb, GlaxoSmithKline, Merck Sharp & Dohme, Novartis, Pierre Fabre, Roche, Sanofi, Iovance, Regeneron. MD reports consultancy fees from Achilles Therapeutics and membership of expert network of Guidepoint LLC and Alphasights, outside of the submitted work. RD reports honoraria from Novartis, Merck Sharp & Dhome (MSD), Bristol-Myers Squibb (BMS), Roche, Amgen, Takeda, Pierre Fabre, Sun Pharma, Sanofi, Catalym, Second Genome, Regeneron, T3 Pharma, MaxiVAX SA, Pfizer and Simcere.outside the submitted work. RG Honoraria for lectures/advisory boards BristolMyers Squibb, MerckSharpDohme, Novartis, Merck-Serono, Amgen, Almirall Hermal, Pierre-Fabre, Sun Pharma, Immunocore, 4SC, Delcath, Sanofi/Regeneron; Meeting support SUN Pharma, Boehringer Ingelheim, PierreFabre; Research support (to institution) Novartis, Sanofi/Regeneron, Merck Serono, Amgen, SUN Pharma, KyowaKirin, Almirall Hermal. AH reports grants and personal fees from Amgen, grants and personal fees from BMS, grants and personal fees from MerckPfizer, grants and personal fees from MSD/Merck, grants and personal fees from Philogen, grants and personal fees from Pierre Fabre, grants and personal fees from Regeneron, personal fees from Roche, grants and personal fees from Sanofi-Genzyme, grants and personal fees from Novartis Pharma, personal fees from Eisai, personal fees from Immunocore, grants and personal fees from Replimune, personal fees from Seagen, personal fees from IO Biotech, personal fees from Dermagnostix, personal fees from Incyte, grants and personal fees from NeraCare, grants from Huya Biosciences, personal fees from Kyowa Kirin, personal fees from Highlight Therapeutics, personal fees from Iovance, personal fees from CureVac, personal fees from Xenthera, peronal fees from Agenus, personal fees from Almirall, and personal fees from Sun Pharma outside the submitted work. MJ reports fees for advisory board Pierre Fabre and AstraZeneca, paid to institution. RL has received institutional funding from Bristol Myers Squibb, Pierre Fabre, AstraZeneca, Chromition. Furthermore, speaker fees for Pierre Fabre. PL reports personal fees for advisory board membership from MLA Dx, MSD and Ultimovacs; personal fees as an invited speaker from BMS and Pierre Fabre; institutional research funding from BMS and Pierre Fabre; travel expenses from MSD; and non-remunerated roles for Cancer Research UK (clinical research committee), EORTC Melanoma Group (Chairman), Melanoma Focus charity (invited speaker) and Society for Melanoma Research (SMR) Edinburgh (invited session chair). IMR declares Advisory role: Amgen, Astra Zeneca, BiolineRx, BMS, Celegene, GSK, Highlight Therapeutics, Immunocore, Merck Serono, MSD, Novartis, Pierre Fabre, Regeneron, Roche, Sanofi, Sun Pharma and Travel accommodation and congress: Amgen, BMS, GSK, Highlight Therapeutics, MSD, Novartis, Pierre Fabre, Roche, Sun Pharma. OM reports personal fees for an advisory role from Amgen, BMS, GSL, MSD, Novartis, Pierre Fabre and Roche; personal fees for membership of the Scientific Board of Cellula Therapeutics; personal fees from stocks and shares from Cellula Therapeutics; institutional fees for advisory board membership from Amgen, BMS, MSD, Novartis, Pierre Fabre and Roche; institutional fees as an invited speaker from Amgen, BMS, Pierre Fabre and Roche, institutional funding from Amgen, BMS, MSD and Pierre Fabre, institutional research grants form Amgen, BMS, Merck, MSD and Pierre Fabre, institutional funding for a writing engagement from BMS and a non-remunerated role as PI for Amgen, BMS, MSD, Novartis, Pierre Fabre and Roche. PN reports personal fees for advisory board membership from 4SC, BMS, IDEAYA, Immunocore, Merck, Novartis and Pfizer; personal fees as an invited speaker from Immuncore and Novartis; personal fees for steering committee membership from 4SC; institutional research funding from Immunocore; and institutional funding as coordinating PI from BMS, Immunocore, Ipsen, Merck, Novartis and Pfizer. CR reports personal consultancy fees for advisory board membership from BMS, Egle Therapeutics, MaaT Pharma, Merck, MSD, Novartis, Pfizer, Philogen, Pierre Fabre, Regeneron, Roche, Sanofi, Sun Pharma and Ultimovacs; personal fees as an invited speaker from BMS, MSD, Novartis, Pierre Fabre and Sanofi; personal fees as for steering committee membership from IO Biotech, Novartis, Pfizer and Regeneron; and personal fees as member of the Independent Data Monitoring Committee for Ultimovacs. DS reports personal fees for advisory board membership from Anaveon, BioNTech, BMS, CureVac, Erasca, Immatics, Immunocore, MSD, Neracare, Novartis, NoviGenix, Pfizer, Philogen, Pierre Fabre, Replimune, Sanofi/Regeneron, Sun Pharma and Ultimovacs; personal fees for expert testimony from AstraZeneca, BioAlta, Daiichi Sankyo, Formycon, InFlarX, PamGene and Seagen; personal fees as an invited speaker from BMS, Merck Serono, MSD, Neracare, Sanofi and Sun Pharma; personal fees for steering committee membership from BMS and MSD; institutional research funding from BMS and MSD; institutional funding as coordinating PI from BMS, MSD, Novartis and Pierre Fabre; institutional funding as local PI from Philogen and Sanofi; non-remunerated steering committee membership for Novartis; non-remunerated membership of the Board of Directors for EORTC-MG; and non-renumerated leadership roles as a founding member and steering committee chair for the European Melanoma Registry (EuMelaReg). PS received honoraria for lectures and consultancies from Gilead, Bristol-Myers. Squibb and Sandoz, travel grants from Pierr Fabre, MSD and Novartis; is a stockholder of Celon Pharma. LF reports grants or contracts from Hookipa Pharma, SAKK/Immunophotonics, DFG Grant Deutsche Forschungsgemeinschaft, Philogen and Mundipharma; consulting fees from Philogen, Sanofi, Novartis, BMS; participation on Data Safety Board University of Basel and stocks or stock options from Hookipa Pharma, outside the submitted work. UL reports research support from MSD, consulting fees and honoraria from Sun Pharma, Sanofi (personal and institutional), MSD (personal and institutional), Novartis, Roche, Almirall Hermal, support for attending meeting from Sun Pharma and participation on a Data Safety Monitoring Board or Advisory Board from Sun Pharma, Sanofi, MSD, Novartis, Roche, Almirall Hermal, outside the submitted work. CG reports personal fees for advisory board membership from MSD and Philogen; personal fees for an advisory role from CeCaVa and NeraCare; and a non-remunerated role as President of the European Association of Dermato-Oncology (EADO).

Consent for publication

Not applicable.

References

  • [1].Ascierto PA, Mandalà M, Ferrucci PF, Guidoboni M, Rutkowski P, Ferraresi V, et al. Sequencing of ipilimumab plus nivolumab and encorafenib plus binimetinib for untreated braf-mutated metastatic melanoma (SECOMBIT): a randomized, three-arm, open-label phase ii trial. J Clin Oncol. 2022;41:212–21. doi: 10.1200/JCO.21.02961. [DOI] [PubMed] [Google Scholar]
  • [2].Atkins MB, Lee SJ, Chmielowski B, Tarhini AA, Cohen GI, Truong TG, et al. Combination dabrafenib and trametinib versus combination nivolumab and ipilimumab for patients with advanced braf-mutant melanoma: the DREAMseq trial-ECOG-ACRIN EA6134. J Clin Oncol: J Am Soc Clin Oncol. 2023;41:186–97. doi: 10.1200/JCO.22.01763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Eggermont Alexander MM, Blank Christian U, Mandala M, Long Georgina V, Atkinson V, Dalle S, et al. Adjuvant pembrolizumab versus placebo in resected stage iii melanoma. N Engl J Med. 2018;378:1789–801. doi: 10.1056/NEJMoa1802357. [DOI] [PubMed] [Google Scholar]
  • [4].Long Georgina V, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, et al. Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma. New England Journal of Medicine. 377:1813–1823. doi: 10.1056/NEJMoa1708539. [DOI] [PubMed] [Google Scholar]
  • [5].Weber J, Mandala M, Del Vecchio M, Gogas Helen J, Arance Ana M, Cowey CL, et al. Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. New England Journal of Medicine. 377:1824–1835. doi: 10.1056/NEJMoa1709030. [DOI] [PubMed] [Google Scholar]
  • [6].Ugurel S, Röhmel J, Ascierto PA, Flaherty KT, Grob JJ, Hauschild A, et al. Survival of patients with advanced metastatic melanoma: the impact of novel therapies–update 2017. Eur J Cancer. 2017;83:247–57. doi: 10.1016/j.ejca.2017.06.028. [DOI] [PubMed] [Google Scholar]
  • [7].Ugurel S, Röhmel J, Ascierto PA, Becker JC, Flaherty KT, Grob JJ, et al. Survival of patients with advanced metastatic melanoma: the impact of MAP kinase pathway inhibition and immune checkpoint inhibition - Update 2019. Eur J Cancer. 2020;130:126–38. doi: 10.1016/j.ejca.2020.02.021. [DOI] [PubMed] [Google Scholar]
  • [8].van Laar SA, Kapiteijn E, Gombert-Handoko KB, Guchelaar HJ, Zwaveling J. Application of electronic health record text mining: real-world tolerability, safety, and efficacy of adjuvant melanoma treatments. Cancers. 2022;14 doi: 10.3390/cancers14215426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].De Falco V, Suarato G, Napolitano R, Argenziano G, Famiglietti V, Amato A, et al. Real-world clinical outcome and safety of adjuvant therapy in stage III melanoma patients: data from two academic Italian institutions. Int J Cancer. 2023;153:133–40. doi: 10.1002/ijc.34462. [DOI] [PubMed] [Google Scholar]
  • [10].Zhong J, Sun W, Hu T, Wang C, Yan W, Luo Z, et al. Comparative analysis of adjuvant therapy for stage III BRAF-mut melanoma: a real-world retrospective study from single center in China. Cancer Med. 2023;12:11475–82. doi: 10.1002/cam4.5866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Placzke J, Rosińska M, Sobczuk P, Ziętek M, Kempa-Kamińska N, Cybulska-Stopa B, et al. Modern approach to melanoma adjuvant treatment with Anti-PD1 Immune Check Point Inhibitors or BRAF/MEK targeted therapy: multicenter real-world report. Cancers. 2023;15 doi: 10.3390/cancers15174384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Haist M, Stege H, Rogall F, Tan Y, von Wasielewski I, Klespe KC, et al. Treatment management for BRAF-mutant melanoma patients with tumor recurrence on adjuvant therapy: a multicenter study from the prospective skin cancer registry ADOREG. J Immunother Cancer. 2023;11 doi: 10.1136/jitc-2023-007630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Lodde GC, Hassel J, Wulfken LM, Meier F, Mohr P, Kähler K, et al. Adjuvant treatment and outcome of stage III melanoma patients: results of a multicenter real-world german dermatologic cooperative oncology group (DeCOG) study. Eur J Cancer. 2023;191:112957. doi: 10.1016/j.ejca.2023.112957. [DOI] [PubMed] [Google Scholar]
  • [14].Schumann K, Mauch C, Klespe KC, Loquai C, Nikfarjam U, Schlaak M, et al. Real-world outcomes using PD-1 antibodies and BRAF + MEK inhibitors for adjuvant melanoma treatment from 39 skin cancer centers in Germany, Austria and Switzerland. J Eur Acad Dermatol Venereol: JEADV. 2023;37:894–906. doi: 10.1111/jdv.18779. [DOI] [PubMed] [Google Scholar]
  • [15].Rigo R, Ding PQ, Batuyong E, Cheung WY, Walker J, Monzon JG, et al. Adjuvant systemic therapies for resected stages iii and iv melanoma: a multi-center retrospective clinical study. oncologist. 2024;29:57–66. doi: 10.1093/oncolo/oyad223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Bai X, Shaheen A, Grieco C, d’Arienzo PD, Mina F, Czapla JA, et al. Dabrafenib plus trametinib versus anti-PD-1 monotherapy as adjuvant therapy in <em>BRAF</em> V. 600-Mutant Stage III Melanoma Defin Surg: a Multicent, Retrosp cohort Study eClinicalMedicine. 2023;65 doi: 10.1016/j.eclinm.2023.102290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Blank CU, Lucas MW, Scolyer RA, Wiel BAvd, Menzies AM, Lopez-Yurda M, et al. Neoadjuvant Nivolumab and Ipilimumab in Resectable Stage III Melanoma. New England Journal of Medicine. doi: 10.1056/NEJMoa2402604. 0. [DOI] [PubMed] [Google Scholar]
  • [18].Patel SP, Othus M, Chen Y, Wright GP, Yost KJ, Hyngstrom JR, et al. Neoadjuvant–adjuvant or adjuvant-only pembrolizumab in advanced melanoma. N Engl J Med. 2023;388:813–23. doi: 10.1056/NEJMoa2211437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Long GV, Hauschild A, Santinami M, Kirkwood JM, Atkinson V, Mandala M, et al. Final Results for Adjuvant Dabrafenib plus Trametinib in Stage III Melanoma. New En9*0gland Journal of Medicine. doi: 10.1056/NEJMoa2404139. 0. [DOI] [PubMed] [Google Scholar]
  • [20].Ny L, Hernberg M, Nyakas M, Koivunen J, Oddershede L, Yoon M, et al. BRAF mutational status as a prognostic marker for survival in malignant melanoma: a systematic review and meta-analysis. Acta Oncol. 2020;59:833–44. doi: 10.1080/0284186X.2020.1747636. [DOI] [PubMed] [Google Scholar]
  • [21].Eggermont AMM, Kicinski M, Blank CU, Mandala M, Long GV, Atkinson V, et al. Five-year analysis of adjuvant pembrolizumab or placebo in stage iii melanoma. NEJM Evid. 2022;1:EVIDoa2200214. doi: 10.1056/EVIDoa2200214. [DOI] [PubMed] [Google Scholar]
  • [22].(EMA) EMA. 2024. https://www.ema.europa.eu/en/documents/product-information/keytruda-epar-product-information_en.pdf .
  • [23].Larkin J, Del Vecchio M, Mandalá M, Gogas H, Arance Fernandez AM, Dalle S, et al. Adjuvant nivolumab versus ipilimumab in resected stage III/IV melanoma: 5-year efficacy and biomarker results from checkMate 238. Clin Cancer Res: J Am Assoc Cancer Res. 2023;29:3352–61. doi: 10.1158/1078-0432.CCR-22-3145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Grossmann KF, Othus M, Patel SP, Tarhini AA, Sondak VK, Knopp MV, et al. Adjuvant pembrolizumab versus IFNα2b or ipilimumab in resected high-risk melanoma. Cancer Discov. 2022;12:644–53. doi: 10.1158/2159-8290.CD-21-1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Egeler MD, van Leeuwen M, Fraterman I, van den Heuvel NMJ, Boekhout AH, Lai-Kwon J, et al. Common toxicities associated with immune checkpoint inhibitors and targeted therapy in the treatment of melanoma: a systematic scoping review. Crit Rev Oncol/Hematol. 2023;183:103919. doi: 10.1016/j.critrevonc.2023.103919. [DOI] [PubMed] [Google Scholar]
  • [26].Haanen J, Obeid M, Spain L, Carbonnel F, Wang Y, Robert C, et al. Management of toxicities from immunotherapy: ESMO clinical practice guideline for diagnosis, treatment and follow-upx2606. Ann Oncol. 2022;33:1217–38. doi: 10.1016/j.annonc.2022.10.001. [DOI] [PubMed] [Google Scholar]
  • [27].Bottomley A, Coens C, Mierzynska J, Mandalà M, Long GV, et al. Adjuvant pembrolizumab versus placebo in resected stage III melanoma (EORTC 1325-MG/KEYNOTE-054): health-related quality-of-life results from a double-blind, randomised, controlled, phase 3 trial. Lancet Oncol. 2021;22:655–64. doi: 10.1016/S1470-2045(21)00081-4. [DOI] [PubMed] [Google Scholar]
  • [28].Egeler M, Lai-Kwon J, Tissier R, Fraterman I, Kuijpers A, Van Houdt W, et al. Real-world health-related quality of life outcomes for patients with resected stage III/IV melanoma treated with adjuvant anti-PD1 therapy. Eur J Cancer. 2024;200:113601. doi: 10.1016/j.ejca.2024.113601. [DOI] [PubMed] [Google Scholar]
  • [29].Rogiers A, Willemot L, McDonald L, Van Campenhout H, Berchem G, Jacobs C, et al. Real-world effectiveness, safety, and health-related quality of life in patients receiving adjuvant nivolumab for melanoma in belgium and luxembourg: results of PRESERV MEL. Cancers. 2023;15 doi: 10.3390/cancers15194823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Pedersen S, Holmstroem RB, von Heymann A, Tolstrup LK, Madsen K, Petersen MA, et al. Quality of life and mental health in real-world patients with resected stage III/IV melanoma receiving adjuvant immunotherapy. Acta Oncol (Stockh, Swed) 2023;62:62–9. doi: 10.1080/0284186X.2023.2165449. [DOI] [PubMed] [Google Scholar]
  • [31].Schadendorf D, Hauschild A, Santinami M, Atkinson V, Mandalà M, Chiarion-Sileni V, et al. Effect on health-related quality of life (HRQOL) of adjuvant treatment (tx) with dabrafenib plus trametinib (D + T) in patients (pts) with resected stage III BRAF-mutant melanoma. J Clin Oncol. 2018;36:9590. doi: 10.1200/JCO.18.01219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Bhave P, Pallan L, Long GV, Menzies AM, Atkinson V, Cohen JV, et al. Melanoma recurrence patterns and management after adjuvant targeted therapy: a multicentre analysis. Br J Cancer. 2021;124:574–80. doi: 10.1038/s41416-020-01121-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Owen CN, Shoushtari AN, Chauhan D, Palmieri DJ, Lee B, Rohaan MW, et al. Management of early melanoma recurrence despite adjuvant anti-PD-1 antibody therapy. Ann Oncol: J Eur Soc Med Oncol / ESMO. 2020;31:1075–82. doi: 10.1016/j.annonc.2020.04.471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Helgadottir H, Ny L, Ullenhag GJ, Falkenius J, Mikiver R, Olofsson Bagge R, et al. Survival after introduction of adjuvant treatment in stage III melanoma: a nationwide registry-based study. J Natl Cancer Inst. 2023;115:1077–84. doi: 10.1093/jnci/djad081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Piulats JM, Espinosa E, de la Cruz Merino I, Varela M, Alonso Carrión L, Martín-Algarra S, et al. Nivolumab plus ipilimumab for treatment-naïve metastatic uveal melanoma: an open-label, multicenter, phase ii trial by the Spanish multidisciplinary melanoma group (GEM-1402) J Clin Oncol. 2021;39:586–98. doi: 10.1200/JCO.20.00550. [DOI] [PubMed] [Google Scholar]
  • [36].Nathan P, Hassel Jessica C, Rutkowski P, Baurain J-F, Butler Marcus O, Schlaak M, et al. Overall survival benefit with tebentafusp in metastatic uveal melanoma. N Engl J Med. 2021;385:1196–206. doi: 10.1056/NEJMoa2103485. [DOI] [PubMed] [Google Scholar]
  • [37].Hassel Jessica C, Piperno-Neumann S, Rutkowski P, Baurain J-F, Schlaak M, Butler Marcus O, et al. Three-year overall survival with tebentafusp in metastatic uveal melanoma. N Engl J Med. 2023;389:2256–66. doi: 10.1056/NEJMoa2304753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Piulats JM, Watkins C, Costa-García M, Del Carpio L, Piperno-Neumann S, Rutkowski P, et al. Overall survival from tebentafusp versus nivolumab plus ipilimumab in first-line metastatic uveal melanoma: a propensity score-weighted analysis. Ann Oncol: J Eur Soc Med Oncol / ESMO. 2024;35:317–26. doi: 10.1016/j.annonc.2023.11.013. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplementary Material

RESOURCES