touchONCOLOGY touchONCOLOGY
Dermatological Cancers
Read Time: 5 mins

Advances in Adjuvant Therapy for the Treatment of High-risk Melanoma

Copy Link
Published Online: Jan 21st 2013 Oncology & Hematology Review 2013;9(2):132–7 DOI: https://doi.org/10.17925/OHR.2013.09.2.132
Authors: Kenneth F Grossmann, Shelley Christiansen
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

The global incidence of melanoma is increasing, and the prognosis for patients remains poor. High-dose interferon-alpha (HD-IFN-a) and pegylated IFN are the only US Food and Drug Administration (FDA)-approved agents for adjuvant therapy for high-risk melanoma, and an improvement in relapse-free survival (RFS) has been observed consistently across nearly all published studies and meta-analyses. Some studies and meta-analyses have also supported an overall survival (OS) benefit. However, despite a number of adjuvant studies, controversy remains regarding the role of this treatment. As the benefits in OS are modest with IFN treatment, there is therefore a need for new therapeutic targets, new drugs, and optimum patient selection. Current research is investigating new adjuvant agents, either individually or in combination, which may advance the standard of care beyond HD-IFN. Additionally, identifying biomarkers of patients with greater likelihood of response may allow patient-specific therapeutic approaches. Following the recent FDA approval of ipilimumab, vemurafenib, dabrafenib, and trametenib for metastatic melanoma, ongoing adjuvant trials are now underway. Acknowledgements: Editorial assistance was provided by Katrina Mountfort at Touch Medical Media.

Keywords

Adjuvant therapy, interferon-alpha, ipilimumab, melanoma, vemurafenib

Article:

Melanoma represents 4% of all malignant tumors of the skin yet is responsible for 80% of deaths from skin cancer; it was estimated that 9,180 people would die of melanoma in 2012.1 The disease disproportionately targets young people: from 2005 to 2009 the age-adjusted incidence rate of melanoma in the US was 21.0 per 100,000 men and women per year, 58.5% of whom were below the age of 64.1 The number of new cases of melanoma in the US has been increasing for the last 30 years: between 1985 and 2009, the incidence of melanoma in the US has more than doubled.1 Treatment of early-stage melanoma by surgery can be curative, but patients with locally advanced disease have a high risk for recurrence and death. Localized melanoma has a 98.2% 5-year survival rate; however, if the cancer has metastasized the 5-year survival rate is 15.1%.1 The American Joint Committee on Cancer (AJCC) classification is the most widely accepted staging system for melanoma and has been recently updated.2

Despite considerable research, the treatment of advanced disease remains challenging. Historically, the alkylating agent, dacarbazine (DTIC), has been the standard therapy for patients with metastatic melanoma. High-dose interleukin-2 (HD IL-2) has been shown to achieve durable long-term complete responses in a small proportion of patients.3 In 2011, the US Food and Drug Administration (FDA) granted approval for ipilimumab (an anti-cytotoxic T-lymphocyte antigen 4 [CTLA-4] human monoclonal antibody) and vemurafenib (an inhibitor of proto-oncogene B-Raf [BRAF] kinase); in 2013, dabrafenib and trametinib were approved for the management of stage IV disease harboring BRAF mutations.

The current treatment for melanoma with lymph node involvement, but without distant metastasis, is surgical excision and lymph node dissection. However, the risk for recurrence of melanoma after surgery is reported to be approximately 60% for stage IIB patients and 75% for stage III patients.4 The probability of recurrence is defined as low, intermediate, or high risk depending on the thickness of the primary tumor, the presence of ulceration or mitoses in the primary tumor, and the presence of nodal or in-transit or satellite metastases around the primary lesion.5,6

Adjuvant therapy is offered after surgical treatment has removed all detectable disease and is given with the intent of reducing relapse risk due to occult disease. Adjuvant therapy should be considered for patients whose risk for recurrence exceeds 30%, i.e. patients with either stage IIB melanoma with a primary thickness greater than 4 mm or greater than 2 mm with ulceration or stage III melanoma.4 For patients with stage I–II disease, sentinel node mapping, a procedure that identifies micro-metastatic disease in the regional lymph nodes with greater precision than an elective lymph node dissection, may be used to select patients for adjuvant therapy.7

This article will discuss the use of IFN-a in the adjuvant setting and will outline other evolving options including vaccines, CTLA-4 blockade, chemotherapy, and radiotherapy.

Interferon Adjuvant Therapy in Melanoma HD interferon-alpha (HD-IFN-a) is the only FDA-approved agent for adjuvant therapy for high-risk melanoma and has shown clinical efficacy; however, the optimal dosing and duration of treatment are still unclear. The schedule for administration of the therapy is as follows: induction: IFN-a: 20 MU, intravenously (IV) 5 days a week for 4 weeks; followed by maintenance: IFN-a: 10 MU, subcutaneously (SC) three times a week for 48 weeks. The FDA approved the use of adjuvant HD-IFN-a in high-risk melanoma after a pivotal phase III trial, E1684, demonstrated significant reduction of recurrence and mortality in patients with high-risk (stage IIB and IIIA/B) melanoma.8 Treatment with HD-IFN-a prolonged the relapsefree survival (RFS) at 5 years by 40%, and overall survival (OS) by 28% compared with the observation group. Since the aim of the study was to administer maximally tolerated doses of IFN, toxicity was significant among those treated with HD-IFN-a. Grade III toxicity occurred in 67% of patients, grade IV toxicity in 9%, and there were two early deaths secondary to hepatotoxicity. This prompted the investigation of the efficacy of lower doses of IFN-a.

A number of alternative IFN-a regimens have been studied with varying degrees of success and are summarized in Table 1. Randomized controlled trials have utilized very low dose (LD) (1 MU every other day),9 and LD (≤3 MU/dose),10–14 but these did not demonstrate significant clinical benefit. A trial comparing LD- and HD-IFN-a showed improvements in RFS in the high-dose setting, but not with low doses.14 A French trial reported significant benefits of treatment with LD-IFN-a for 18 months in terms of OS and RFS,15 but this was in stage II patients before clinically detectable lymph node metastases had developed. Given the relatively good prognosis of these patients, the cost–benefit advantages of this approach are questionable. An intermediate dose failed to demonstrate significant benefits in terms of OS or RFS.16

The duration of therapy of HD-IFN-a has been the subject of debate. In a study to determine the potential benefit of adding a short-term highdose induction phase to conventional adjuvant LD-IFN-a treatment of patients with high-risk primary melanoma, no significant differences were observed in terms of OS, or RFS when compared with a conventionally treated group of patients.17 The Hellenic group found no differences in RFS and OS with 1 month versus 1 year of adjuvant HD-IFN-a therapy in patients with resected, high-risk (stage IIB to III) melanoma.18 However, this trial was underpowered to achieve statistical significance. Another trial compared therapy for 1 month with therapy for 1 year for patients with intermediate-risk melanoma. The trial was stopped for futility, because the 1-month HD-IFN-a was not better than observation. It was concluded that 1 month of therapy with HD-IFN-a is insufficient to improve RFS and OS.19 Further efforts to modify the IFN-a dose or schedule have not improved its efficacy.20 These data suggest that the benefit of IFN-a requires more lengthy treatment than just the induction phase of the high-dose regimen. The optimal dosing and duration of treatment with IFN-a could be improved with further study, but the promise of newer agents suggests other approaches.

In a meta-analysis, no optimal IFN-a dose and/or treatment duration or a subset of patients more responsive to adjuvant therapy was identified.21 The meta-analysis of 14 studies concluded that IFN-a treatment was associated with a statistically significant improvement in RFS in 10 of the 17 comparisons (hazard ratio [HR] for disease recurrence = 0.82, 95% confidence interval [CI] = 0.77–0.87; p<0.001) and improved OS in four of the 14 comparisons (HR for death = 0.89, 95% CI=0.83–0.96; p=0.002). These three meta-analyses and one systematic review to date have found that IFN-a treatment was associated with a statistically significant improvement RFS with a lesser impact on OS, and there is no evidence of survival gain beyond 10 years.21–24 More recently, a Cochrane review of 18 RCTs and 10,499 patients showed a RFS benefit (HR 0.83; p 0.0001) and OS benefit (HR 0.91; p=0.003).25

In 2011, the FDA-approved pegylated IFN-a-2b (PEG-IFN-a-2b) for the adjuvant treatment of patients with high-risk melanoma, following the results of a phase III trial.26 Long-term (7.6 years) follow-up data showed improved RFS but not OS in the treatment arm.27 Subgroup analysis suggests that the benefit of PEG-IFN may be limited to patients with micrometastatic disease and ulcerated primaries.27 A recent phase II trial compared PEG-IFN-a-2b with LD-IFN-a-2b in patients with resected stage IIA–IIIB melanoma. No differences were observed between the two groups in terms of RFS, OS, or distant metastasis-free survival (DMFS), although PEG-IFN-a was associated with higher rates of grade 3–4 adverse events (47.3% versus 25.2%) and discontinuations (54.3% versus 30.4%) compared with LD-IFN-a.28 PEG-IFN-a may be considered as an alternative to HD-IFN-a for patients unwilling to undergo a high-dose regimen.

In summary, in the clinical trials of IFN-a adjuvant therapy to date, only HD-IFN-a has consistently shown an improvement of RFS in all studies and has shown an increase in OS in randomized clinical trials (RCTs). However, it remains uncertain which patient population benefits most from adjuvant treatment. In addition, the use of IFN-a is associated with toxicity and requires a team with experience in its management. There is a need for other therapeutic regimes in adjuvant treatment of melanoma.

Current Research into Alternative Adjuvant Therapies
A number of alternative adjuvant therapies are being investigated. A summary is given in Table 2.

Biochemotherapy
Several trials have evaluated the use of adjuvant chemotherapy following surgical resection in high-risk patients, but none have demonstrated significant benefits in phase III clinical trials.29 Biochemotherapy (the combination of biologics and chemotherapy) has demonstrated improvements in RFS in an RCT30 and is associated with long-term survival as demonstrated by a single-institution series.31 This has led to investigation of its use in the adjuvant setting.

A phase III trial compared a combination of LD-IFN-a with DTIC and LDIFN- a alone following lymph node dissection. However, the addition of DTIC reversed the beneficial effect of adjuvant IFN-a therapy.32 A recent study aimed to determine whether a short course of biochemotherapy would be more effective than HD-IFN-a as adjuvant treatment in patients with high-risk melanoma (stage IIIB and IV patients). A study comparing HD IFN-a, LD IFN-a, and biochemotherapy (consisting of DTIC, cisplatin, vinblastine, IL-2, and IFN-a) in patients with stage III disease was unable to show a statistically significant improvement in patients receiving biochemotherapy compared with HD IFN-a or ID IFN-a.33 However, this study aimed to show a doubling of RFS and OS with biochemotherapy over IFN-a. A less-ambitious target increase in RFS may have yielded different results. Furthermore, the study stopped early due to slow accrual with only 138 of the planned 200 patients enrolled, further limiting the power of the study. A later study including a larger number of patients, also with stage III disease, revealed biochemotherapy (consisting of DTIC, cisplatin, vinblastine, IL-2, IFN-a, and granulocyte colony-stimulating factor [G-CSF]) produced a statistically significant improvement in RFS (4.31 versus 1.9 year median) compared with HD IFN-a, but no discernable difference in OS.34

These data are intriguing and support the use of biochemotherapy in the adjuvant setting for patients ineligible for clinical trials or for patients not wishing to participate in clinical studies and who have access to medical centers with expertise in the administration of this complex regimen. Unfortunately, the toxicity and complexity of the administration of this regimen is a significant barrier to making it standard practice for stage III melanoma, and make it ill-suited for a control arm in large randomized controlled trials.

Checkpoint Blockade
The use of ipilimumab as first-line treatment is well-established,35 and clinical trials are now underway to assess its utility in the adjuvant setting. Two clinical trials are currently ongoing for the adjuvant use of ipilimumab. The Eastern Cooperative Oncology Group (ECOG) 1609 clinical trial is currently underway has three treatment arms: high-dose ipilimumab, lowdose ipilimumab, and HD-IFN-a (see Figure 1).36 In Europe, the European Organisation for Research and Treatment of Cancer (EORTC) 18071 trial, comparing ipilimumab against placebo, has completed accrual and results are pending.37 Given the high cost and toxicity of ipilimumab, biomarkers of response would be useful to identify relevant patient cohorts. Elevated tumor-infiltrating lymphocyte (TIL), T reg, and indoleamine 2,3-dioxygenase (IDO) levels, have been associated with improved outcomes in patients receiving ipilimumab therapy in the first-line setting.38

Granulocyte-macrophage Colony-stimulating Factor
Granulocyte-macrophage colony-stimulating factor (GM-CSF) may be effective as an adjuvant therapy in stage III patients.39 A phase III trial in stage IIIB, IIIC, and IV disease, found that GM-CSF improved the RFS with minimal toxicity.40 A single-center study has also provided evidence for the efficacy of GM-CSF in this setting.41 There is a need for further clinical trials investigating its use and it may be that in combination with other immunotherapy we will see enhanced results.

A phase II trial found that the combination treatment regimen of GM-CSF and IL-2 in the adjuvant setting appeared to benefit high-risk melanoma patients.42 In a new approach to adjuvant therapy, a pilot study investigated the administration of a single short course of GM-CSF and IL-2 intradermally at the primary site, prior to its excision. This very small study underscores the biological activity of GM-CSF and IL-2 when given intradermally, and suggests that although newer studies are focusing on other agents, the addition of intradermal injections of IL-2 and GM-CSF to other adjuvant treatment schemes may prove beneficial.43


Melanoma vaccines have been extensively studied in the hope of obtaining durable clinical responses. Several large randomized trials of adjuvant allogeneic melanoma cell-based vaccines have been conducted, none of which have found any survival benefit.44–46 A polyvalent vaccine, Canvaxin, was evaluated as adjuvant therapy in stage III melanoma patients in a retrospective study. Median and 5-year OS were higher in vaccinated patients than in nonvaccinated patients.47 However, in a subsequent phase III trial for resected stage III/IV melanoma, Canvaxin did not increase either RFS or OS. In fact, survival was lower in the vaccine group (5% in stage IV and 9% in stage III) possibly a result of vaccine-induced immunosuppression.48 A recent phase III study, which randomized patients with stage II melanoma to adjuvant ganglioside GM2-KLH/QS-21 vaccination (n=657) or observation (n=657), found that this vaccine did not improve outcomes.49

Phase I/II trials have demonstrated an immunologic response from postoperative vaccine with melanoma -specific antigen A3 (MAGE-A3) in stage IV melanoma.50 The MAGE-A3 Antigen-Specific Cancer Immunotherapeutic (ASCI) combines a tumor-specific antigen delivered as a recombinant protein with an immunostimulant AS15. Unfortunately, a phase III trial investigating the use of MAGE-A3 ASCI as an adjuvant treatment for resected stage III MAGE-A3 positive melanoma patients has completed accrual and preliminary results suggest no benefit as reported by GlaxoSmithKline.51

Preliminary results of a trial comparing dendritic cell (DC) vaccine therapy with observation in stage III and IV melanoma patients (n=108) postsurgery presented at ASCO 2012 showed that immunotherapy with DC vaccine is safe and improves RFS compared with observation in the adjuvant treatment of stage III and IV melanoma. At a median follow up of 22 months, the HR for RFS for DC vaccine versus observation was 0.45 (95% CI 0.29–0.69; p<0.05) and for OS was 0.71 (95% CI 0.40–1.25; p=0.23).52 A peer-reviewed publication of this dataset is eagerly anticipated.

Radiotherapy
Radiotherapy may be valuable in the adjuvant setting in patients with bulky disease of involvement of nodes, particularly for those with the potential for a highly symptomatic nodal relapse.6 The use of radiotherapy as adjuvant has been investigated in a phase III trial, but although it improved regional control, it did not improve OS.53 Further work from this group presented at this last year’s ASCO confirmed the RFS advantage, but lack of OS and trend toward decreased quality of life suggest careful selection of patients where decrease in relapse outweighs the potential morbidity in terms of lymphedema risk.54

Targeted Agents
Approximately 40 to 60% of advanced melanomas have mutations in BRAF, and the BRAF inhibitor vemurafenib has demonstrated efficacy in the first-line setting,55 which has led to the suggestion that this agent may be useful as adjuvant therapy in stage IV disease. A phase III study of vemurafenib (RO5185426) adjuvant treatment of patients with surgically resected, cutaneous BRAF mutant melanoma at high risk for recurrence is currently ongoing.56 Further work has demonstrated improved response rates and PFS with combinations of b-raf and mek inhibitors.57,58 A second study comparing dabrafenib and tremetenib together versus observation is also underway.59

Prognostic Biomarkers and Predictive Factors for Response to Adjuvant Therapy in Melanoma
There is a need for prognostic biomarkers of IFN-a response, since identification of the ~15% of patients who will derive benefit from IFN-a would increase the therapeutic index of this agent. Numerous candidate molecules have been studied, including methylthioadenosine phosphorylase expression, YKL-40, S100B, melanoma-inhibiting activity, and tumor-associated antigen 90 immune complex.6 However, there is a lack of prospective data validating the use of these biomarkers. Serum protein S100B has been demonstrated to significantly correlate with mortality risk when assessed at baseline in patients with high-risk resectable melanoma.60

IFN-a-2b upregulates STAT1, a molecular molecule of progression, and downregulates STAT3 in tumor cells and host lymphocytes, The pSTAT1/ pSTAT3 ratio in tumor cells at baseline may serve as a useful predictor of clinical outcome in melanoma; the modulation of this ratio may serve as a predictor of therapeutic effect.61

Detection of circulating tumor cells (CTC) by molecular approaches may also be a promising prognostic biomarker in melanoma patients. In a recent phase III clinical trial, blood levels of three messenger RNA (mRNA) biomarkers of CTCs (MART-1, MAGE-A3, and PAX-3) status were significantly associated with RFS (HR 1.64; p=0.002) and OS (HR 1.53; p=0.028) before and during adjuvant treatment for resected stage IV melanoma patients.62

Lower tumor stage and ulceration have been noted as predictive factors for patient response to PEG-IFN-a-2b.27 Analysis of the results of the PEGIFN- a trial found that ulceration of the primary tumour was not only a strong prognostic factor, but also a significant predictive factor for patient response to adjuvant IFN-a treatment.27 There is also evidence that patients treated with IFN-a may benefit more if they have micrometastases than if they have macrometastases of the lymph nodes.63 In addition, the appearance of autoantibodies or clinical manifestations of autoimmunity during treatment with IFN-a-2b was also associated with improvements in RFS and OS in patients with melanoma.64 This observation was confirmed in a subsequent study65 and warrants further investigation. A phase III study of PEG-IFN-a-2b in high-risk, stage II patients with ulcerated melanomas (EORTC 18081) is ongoing.66

Summary and Concluding Remarks
Numerous clinical data support the efficacy of adjuvant IFN-alpha in the treatment of melanoma – although it is associated with consistent improvements in RFS, its effect on OS has been small. Future research should focus on determining the scientific basis of action of IFN-a, the optimum duration of therapy, as well as how to overcome resistance to further enhance efficacy of IFN-a. In particular, there is a need for clinical trial data in the highest risk category: resected stage IIIb-IV disease. An increased understanding of biological determinants of response, such as the induction of autoantibodies, as well as the identification of the biomarkers of subpopulations of patients who are likely to benefit the most from IFN-a therapy, will increase the effectiveness of clinical trials and provide further insights into the mechanisms of IFN antitumor activity.

The lack of treatments with proven efficacy against resected melanoma has affected the investigation of adjuvant treatment in melanoma therapy. Chemotherapy, cytokines, vaccines, and combinations of these treatments have been investigated, but with little success to date. Ongoing melanoma adjuvant trials are now testing the next generation of immunotherapy, as well as targeted agents. The complete and durable responses observed in phase III trials of ipilimumab in patients with metastatic melanoma suggest that this agent may demonstrate efficacy in the adjuvant setting. Although targeted agents, such as vemurafenib and combination treatment with dabrafenib and trametenib, may be beneficial and less toxic than HD IFN-a, their effects in stage IV disease appear to require continuous administration. In the adjuvant setting it may be that given their high response rate, micrometastatic disease will be eliminated at a similarly high rate and, if so, long-term benefits would be possible without continued treatment.

Although programmed cell death 1 (PD-1) blockade appears to be an interesting new immunotherapeutic its ability to produce treatment-free complete responses remains unknown. The favorable toxicity profile of the PD-1 inhibitors, in addition to the highest response rates yet documented for single-agent immunotherapeutics in treating stage IV patients, make them attractive agents for clinical study in the adjuvant setting. Future work in the adjuvant setting will be further informed by the efficacy of single agents, and combinations in metastatic disease.72,73

Article Information:
Disclosure

Kenneth F Grossman, MD, PhD, has received honoraria from Prometheus for travel and funding trainees to attend meetings. He has completed uncompensated advisory work for Roche/Genentech. Shelley Christiansen, MD, has no conflicts of interest to declare.

Correspondence

Kenneth F Grossmann, MD, PhD, 2000 Circle of Hope Drive, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84106, US, E: kenneth.grossmann@hci.utah.edu

Support

The publication of this article was supported by Prometheus. The views and opinions expressed are those of the authors and not necessarily those of Prometheus.

Received

2013-10-24T00:00:00

References

1. Howlader NNA, Krapcho M, Krapcho M (eds), SEER Cancer Statistics Review, 1975–2009 (Vintage 2009 Populations), National Cancer Institute. Bethesda, MD. https://seer.cancer.gov/ csr/1975_2009_pops09/ (accessed November 25, 2013).
2. Balch CM, Gershenwald JE, Soong SJ, et al., Final version of 2009 AJCC melanoma staging and classification, J Clin Oncol, 2009;27:6199–206.
3. Atkins MB, Lotze MT, Dutcher JP, et al., High-dose recombinant interleukin 2 therapy for patients with metastatic melanoma: analysis of 270 patients treated between 1985 and 1993, J Clin Oncol, 1999;17:2105–16.
4. Balch CM, Buzaid AC, Soong SJ, et al., Final version of the American Joint Committee on Cancer staging system for cutaneous melanoma, J Clin Oncol, 2001;19:3635–48.
5. Essner R, Chung MH, Bleicher R, et al., Prognostic implications of thick (>or=4-mm) melanoma in the era of intraoperative lymphatic mapping and sentinel lymphadenectomy, Ann Surg Oncol, 2002;9:754–61.
6. Davar D, Tarhini A, Kirkwood JM, Adjuvant therapy: melanoma, J Skin Cancer, 2011;2011:274382.
7. Eggermont AM, Adjuvant therapy of malignant melanoma and the role of sentinel node mapping, Recent Results Cancer Res, 2000;157:178–89.
8. Kirkwood JM, Strawderman MH, Ernstoff MS, et al., Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684, J Clin Oncol, 1996;14:7–17.
9. Kleeberg UR, Suciu S, Brocker EB, et al., Final results of the EORTC 18871/DKG 80-1 randomised phase III trial. rIFN-alpha2b versus rIFN-gamma versus ISCADOR M versus observation after surgery in melanoma patients with either high-risk primary (thickness >3 mm) or regional lymph node metastasis, Eur J Cancer, 2004;40:390–402.
10. Cascinelli N, Belli F, MacKie RM, et al., Effect of long-term adjuvant therapy with interferon alpha-2a in patients with regional node metastases from cutaneous melanoma: a randomised trial, Lancet, 2001;358:866–9.
11. Cameron DA, Cornbleet MC, Mackie RM, et al., Adjuvant interferon alpha 2b in high risk melanoma – the Scottish study, Br J Cancer, 2001;84:1146–9.
12. Hauschild A, Weichenthal M, Rass K, et al., Efficacy of low-dose interferon {alpha}2a 18 versus 60 months of treatment in patients with primary melanoma of >= 1.5 mm tumor thickness: results of a randomized phase III DeCOG trial, J Clin Oncol, 2010;28:841–6.
13. Hancock BW, Wheatley K, Harris S, et al., Adjuvant interferon in high-risk melanoma: the AIM HIGH Study—United Kingdom Coordinating Committee on Cancer Research randomized study of adjuvant low-dose extended-duration interferon Alfa-2a in highrisk resected malignant melanoma, J Clin Oncol, 2004;22:53–61.
14. Kirkwood JM, Ibrahim JG, Sondak VK, et al., High- and low-dose interferon alfa-2b in high-risk melanoma: first analysis of intergroup trial E1690/S9111/C9190, J Clin Oncol, 2000;18:2444–58.
15. Grob JJ, Dreno B, de la Salmoniere P, et al., Randomised trial of interferon alpha-2a as adjuvant therapy in resected primary melanoma thicker than 1.5 mm without clinically detectable node metastases. French Cooperative Group on Melanoma, Lancet, 1998;351:1905–10.
16. Eggermont AM, Suciu S, MacKie R, et al., Post-surgery adjuvant therapy with intermediate doses of interferon alfa 2b versus observation in patients with stage IIb/III melanoma (EORTC 18952): randomised controlled trial, Lancet, 2005;366:1189–96.
17. Hauschild A, Weichenthal M, Rass K, et al., Prospective randomized multicenter adjuvant dermatologic cooperative oncology group trial of low-dose interferon alfa-2b with or without a modified highdose interferon alfa-2b induction phase in patients with lymph node-negative melanoma, J Clin Oncol, 2009;27:3496–502.
18. Pectasides D, Dafni U, Bafaloukos D, et al., Randomized phase III study of 1 month versus 1 year of adjuvant high-dose interferon alfa-2b in patients with resected high-risk melanoma, J Clin Oncol, 2009;27:939–44.
19. Agarwala SS, Lee SJ, Flaherty LE, et al., Randomized phase III trial of high-dose interferon alfa-2b (HDI) for 4 weeks induction only in patients with intermediate- and high-risk melanoma (Intergroup trial E 1697), J Clin Oncol, 2011;(Suppl. 29):abstract 8505.
20. Mohr P, Hauschild A, Trefzer U, et al., Intermittent high-dose intravenous interferon alpha 2b (IFNa2b) for adjuvant treatment of stage III malignant melanoma: Final analysis of a randomized phase III DeCOG-trial (NCT00226408), J Clin Oncol, 2012;(Suppl. 30):abstract 8505.
21. Mocellin S, Pasquali S, Rossi CR, et al., Interferon alpha adjuvant therapy in patients with high-risk melanoma: a systematic review and meta-analysis, J Natl Cancer Inst, 2010;102:493–501.
22. Verma S, Quirt I, McCready D, et al., Systematic review of systemic adjuvant therapy for patients at high risk for recurrent melanoma, Cancer, 2006;106:1431–42.
23. Pirard D, Heenen M, Melot C, et al., Interferon alpha as adjuvant postsurgical treatment of melanoma: a meta-analysis, Dermatology, 2004;208:43–8.
24. Wheatley K, Ives N, Hancock B, et al., Does adjuvant interferonalpha for high-risk melanoma provide a worthwhile benefit? A meta-analysis of the randomised trials, Cancer Treat Rev, 2003;29:241–52.
25. Mocellin S, Lens MB, Pasquali S, et al., Interferon alpha for the adjuvant treatment of cutaneous melanoma, Cochrane Database Syst Rev, 2013;6:CD008955.
26. Eggermont AM, Suciu S, Santinami M, et al., Adjuvant therapy with pegylated interferon alfa-2b versus observation alone in resected stage III melanoma: final results of EORTC 18991, a randomised phase III trial, Lancet, 2008;372:117–26.
27. Eggermont AM, Suciu S, Testori A, et al., Ulceration and stage are predictive of interferon efficacy in melanoma: results of the phase III adjuvant trials EORTC 18952 and EORTC 18991, Eur J Cancer, 2012;48:218–25.
28. Grob JJ, Jouary T, Dreno B, et al., Adjuvant therapy with pegylated interferon alfa-2b (36 months) versus low-dose interferon alfa-2b (18 months) in melanoma patients without macrometastatic nodes: an open-label, randomised, phase 3 European Association for Dermato-Oncology (EADO) study, Eur J Cancer, 2013;49:166–74.
29. DeVita V, Lawrence TS, Rosenberg SA, DeVita, Hellman, and Rosenberg’s Cancer: Principles & Practice of Oncology, Lippincott Williams & Wilkins, 2008;1929–30.
30. Eton O, Legha SS, Bedikian AY, et al., Sequential biochemotherapy versus chemotherapy for metastatic melanoma: results from a phase III randomized trial, J Clin Oncol, 2002;20:2045–52.
31. Bedikian AY, Johnson MM, Warneke CL, et al., Systemic therapy for unresectable metastatic melanoma: impact of biochemotherapy on long-term survival, J Immunotoxicol, 2008;5:201–7.
32. Garbe C, Radny P, Linse R, et al., Adjuvant low-dose interferon {alpha}2a with or without dacarbazine compared with surgery alone: a prospective-randomized phase III DeCOG trial in melanoma patients with regional lymph node metastasis, Ann Oncol, 2008;19:1195–201.
33. Kim KB, Legha SS, Gonzalez R, et al., A randomized phase III trial of biochemotherapy versus interferon-alpha-2b for adjuvant therapy in patients at high risk for melanoma recurrence, Melanoma Res, 2009;19:42–9.
34. Flaherty L, Moon J, Atkins MB, et al., Phase III trial of high-dose interferon alpha-2b versus cisplatin, vinblastine, DTIC plus IL-2 and interferon in patients with high-risk melanoma (SWOG S0008): An intergroup study of CALGB, COG, ECOG, and SWOG, J Clin Oncol, 2012;(Suppl. 30):abstract 8504.
35. Hodi FS, O’Day SJ, McDermott DF, et al., Improved survival with ipilimumab in patients with metastatic melanoma, N Engl J Med, 2010;363:711–23.
36. Minutilli E, Feliciani C, Adjuvant therapy for resected stage III melanoma patients: high-dose interferon-alpha versus ipilimumab combined with kinases inhibitors, Tumori, 2012;98:185–90.
37. EORTC, Trial # 18071: Adjuvant immunotherapy with anti- CTLA-4 monoclonal antibody (ipilimumab) versus placebo after complete resection of high-risk Stage III melanoma: A randomized, double-blind Phase 3 trial of the EORTC Melanoma Group. Available at: https://www.melanomagroup.eu/site/ component/docman/doc_details/64-18071-open-report. html?Itemid=135 (accessed November 25, 2013).
38. Hamid O, Chasalow SD, Tsuchihashi Z, et al. , Association of baseline and on study tumor biopsy markers with clinical activity in patients (pts) with advanced melanoma treated with ipilimumab, J Clin Oncol, 2009;(Suppl. 27):abstract 9008.
39. Spitler LE, Grossbard ML, Ernstoff MS, et al., Adjuvant therapy of stage III and IV malignant melanoma using granulocytemacrophage colony-stimulating factor, J Clin Oncol, 2000;18:1614–21.
40. Lawson D, Lee SJ, Tarhini AA, et al., E4697: Phase III cooperative group study of yeast-derived granulocyte macrophage colony-stimulating factor (GM-CSF) versus placebo as adjuvant treatment of patients with completely resected stage III–IV melanoma, J Clin Oncol, 2010;(Suppl. 28):abstract 8504.
41. Markovic S, Burch, PA, LaPlant B et al, Adjuvant GM-CSF therapy for patients with resented stage III/IV melanoma: A retrospective review of a single-center experience, J Clin Oncol, 2011;(Suppl. 29):abstract 8596.
42. Elias EG, Zapas JL, Beam SL, et al., GM-CSF and IL-2 combination as adjuvant therapy in cutaneous melanoma: early results of a phase II clinical trial, Oncology (Williston Park), 2005;19:15–18.
43. Elias E, New approach to adjuvant therapy in high-risk melanoma patients, J Clin Oncol, 2012;(Suppl. 30):abstract e13129
44. Hersey P, Coates AS, McCarthy WH, et al., Adjuvant immunotherapy of patients with high-risk melanoma using vaccinia viral lysates of melanoma: results of a randomized trial, J Clin Oncol, 2002;20:4181–90.
45. Mitchell MS, Abrams J, Thompson JA, et al., Randomized trial of an allogeneic melanoma lysate vaccine with low-dose interferon Alfa-2b compared with high-dose interferon Alfa-2b for Resected stage III cutaneous melanoma, J Clin Oncol, 2007;25:2078–85.
46. Livingston PO, Wong GY, Adluri S, et al., Improved survival in stage III melanoma patients with GM2 antibodies: a randomized trial of adjuvant vaccination with GM2 ganglioside, J Clin Oncol, 1994;12:1036–44.
47. Morton DL, Hsueh EC, Essner R, et al., Prolonged survival of patients receiving active immunotherapy with Canvaxin therapeutic polyvalent vaccine after complete resection of melanoma metastatic to regional lymph nodes, Ann Surg, 2002;236:438–48; discussion 48–9.
48. Morton D, Mozzillo N, Thompson JF, et al., An international, randomized, phase III trial of bacillus Calmette-Guerin (BCG) plus allogeneic melanoma vaccine (MCV) or placebo after complete resection of melanoma metastatic to regional or distant sites, J Clin Oncol, 2007;25:8508.
49. Eggermont AM, Suciu S, Rutkowski P, et al., Adjuvant ganglioside GM2-KLH/QS-21 Vaccination versus observation after resection of primary tumor > 1.5 mm in patients with stage ii melanoma: results of the EORTC 18961 Randomized Phase III Trial, J Clin Oncol, 2013;31:3831–7.
50. Vantomme V, Dantinne C, Amrani N, et al., Immunologic analysis of a phase I/II study of vaccination with MAGE-3 protein combined with the AS02B adjuvant in patients with MAGE-3- positive tumors, J Immunother, 2004;27:124–35.
51. GlaxoSmithKline, Press release: The investigational MAGE-A3 antigen-specific cancer immunotherapeutic does not meet first co-primary endpoint in Phase III melanoma clinical trial. Available at: https://www.gsk.com/media/press-releases/2013/ the-investigational-mage-a3-antigen-specific-cancerimmunotherap. html (accessed November 6, 2013).
52. Petenko N, Mikhaylova IN, Chkadua GZ, et al., Adjuvant dendritic cell (DC)-based vaccine therapy of melanoma patients, J Clin Oncol, 2012;(Suppl. 30):abstract 2524.
53. Henderson MA, Burmeister B, Thompson, JF, et al., Adjuvant radiotherapy and regional lymph node field control in melanoma patients after lymphadenectomy: Results of an intergroup randomized trial (ANZMTG 01.02/TROG 02.01), J Clin Oncol, 2009;(Suppl. 27):abstract LBA9084.
54. Henderson MA, Adjuvant radiotherapy after lymphadenectomy in melanoma patients: Final results of an intergroup randomized trial (ANZMTG 0.1.02/TROG 02.01). Presented at the 2013 ASCO meeting, May 31–Jun 4, 2013, Chicago, IL, abstract no. 9001.
55. Chapman PB, Hauschild A, Robert C, et al., Improved survival with vemurafenib in melanoma with BRAF V600E mutation, N Engl J Med, 2011;364:2507–16.
56. A study of vemurafenib adjuvant therapy in patients with resected cutaneous BRAF mutant melanoma. Available at: https://clinicaltrials.gov/show/NCT01667419 (accessed November 6, 2013).
57. Flaherty K, Infante J, Daud A, et al., Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations, N Engl J Med, 2012;367:1694–03.
58. Gonzalez R, Ribas A, Daud A, et al., Phase 1B study of vemurafenib in combination with the MEK inhibitor, GDC-0973, in patients (pts) with unresectable or metastatic BRAFV600 mutated melanoma (BRIM7), Ann Oncol, 2012;23(Suppl. 9):ixe19(abstract LBA28_PR).
59. A study of the BRAF inhibitor dabrafenib in combination with the MEK inhibitor trametinib in the adjuvant treatment of high-risk BRAF V600 mutation-positive melanoma after surgical resection (COMBI-AD). Available at: https://clinicaltrials.gov/ show/NCT01682083 (accessed November 6, 2013).
60. Tarhini AA, Stuckert J, Lee S, et al., Prognostic significance of serum S100B protein in high-risk surgically resected melanoma patients participating in Intergroup Trial ECOG 1694, J Clin Oncol, 2009;27:38–44.
61. Wang W, Edington HD, Rao UN, et al., Modulation of signal transducers and activators of transcription 1 and 3 signaling in melanoma by high-dose IFNalpha2b, Clin Cancer Res, 2007;13:1523–31.
62. Hoshimoto S, Faries MB, Morton DL, et al., Assessment of prognostic circulating tumor cells in a phase III trial of adjuvant immunotherapy after complete resection of stage IV melanoma, Ann Surg, 2012;255:357–62.
63. Hauschild A, Adjuvant interferon alfa for melanoma: new evidence-based treatment recommendations?, Curr Oncol, 2009;16:3–6.
64. Gogas H, Ioannovich J, Dafni U, et al., Prognostic significance of autoimmunity during treatment of melanoma with interferon, N Engl J Med, 2006;354:709–18.
65. Kirkwood JM, Tarhini AA, Biomarkers of therapeutic response in melanoma and renal cell carcinoma: potential inroads to improved immunotherapy, J Clin Oncol, 2009;27:2583–5.
66. EORTC protocol 1808. Adjuvant peginterferon alpha-2b for 2 years vs Observation in patients with an ulcerated primary cutaneous melanoma with T(2-4)bN0M0: a randomized phase III trial of the EORTC Melanoma Group. Available at: https://www.eortc.be/protoc/Details.asp?Protocol=18081 (accessed November 25, 2013).
67. Creagan ET, Dalton RJ, Ahmann DL, et al., Randomized, surgical adjuvant clinical trial of recombinant interferon alfa-2a in selected patients with malignant melanoma, J Clin Oncol, 1995;13:2776–83.
68. Kirkwood JM, Ibrahim JG, Sosman JA, et al., High-dose interferon alfa-2b significantly prolongs relapse-free and overall survival compared with the GM2-KLH/QS-21 vaccine in patients with resected stage IIB-III melanoma: results of intergroup trial E1694/S9512/C509801, J Clin Oncol, 2001;19:2370–80.
69. Pehamberger H, Soyer HP, Steiner A, et al., Adjuvant interferon alfa-2a treatment in resected primary stage II cutaneous melanoma. Austrian Malignant Melanoma Cooperative Group, J Clin Oncol, 1998;16:1425–9.
70. Ipilimumab or high-dose interferon alfa-2b in treating patients with high-risk stage III or stage iv melanoma that has been removed by surgery. Available at: https://clinicaltrials.gov/show/ NCT01274338 (accessed November 6, 2013).
71. Study of a melanoma vaccine in stage IIb, IIc, and III melanoma patients (MAVIS). Available at: https://www.clinicaltrials.gov/ ct2/show/NCT01546571?term=polynoma+melanoma&rank=1; (accessed November 6, 2013).
72. Gibney GT, Weber JS, Kudchadkar RR, et al., Safety and efficacy of adjuvant anti-PD1 therapy (nivolumab) in combination with vaccine in resected high-risk metastatic melanoma, J Clin Oncol, 2013;31(Suppl.; abstr 9056).
73. Ribas A, Robert C, Daud A, Clinical efficacy and safety of lambrolizumab (MK-3475, Anti-PD-1 monoclonal antibody) in patients with advanced melanoma, J Clin Oncol, 2013;31(Suppl.; abstr 9009).

Further Resources

Share this Article
Related Content In Dermatological Cancers
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72