Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 1.
Published in final edited form as: Semin Immunopathol. 2011 Jan 31;33(4):385–391. doi: 10.1007/s00281-011-0247-y

Prognostic significance of autoimmunity during treatment of melanoma with interferon

Michal T Krauze 1,2, Ahmad Tarhini 3,4, Helen Gogas 5, John M Kirkwood 6,7,8
PMCID: PMC8635120  NIHMSID: NIHMS1753207  PMID: 21279809

Abstract

Since the pivotal cooperative group trials in the 1980’s-90’s, high-dose interferon (HDI) has been the standard of adjuvant therapy. Despite multiple other trials evaluating potential new therapies in melanoma, HDI remains the only FDA-approved therapy for stage IIB and III melanoma. Initial reports from the more recent phase III international trials of modifications of the original HDI regimen linked the appearance of autoimmunity with improved outcomes of disease. Trials of high-dose interleukin-2, many years earlier, reported anecdotal observations that were consistent with the hypothesis that autoimmunity and clinical benefit of immunotherapies of melanoma are linked with one another. The only prospectively conducted study examining the appearance of clinical and laboratory evidence of autoimmunity during HDI therapy was published by Gogas and colleagues, demonstrating statistically significant impact on relapse-free survival and overall survival. Retrospectively conducted studies of different intermediate dosage regimens of interferon (IFN) have not fully confirmed the linkage of serological evidence of autoimmunity and improved survival outcomes. With the emergence of new immunotherapies in treatment of melanoma, this review highlights the importance of autoimmunity for future applications in melanoma and reviews significant differences of past studies evaluating the appearance of autoimmunity during IFN therapy in high-risk melanoma.

Keywords: Melanoma, Autoimmunity, Interferon, High-dose interferon, Cancer

Introduction

Despite increasing efforts in prevention and therapy, metastatic melanoma at the time of diagnosis remains a very aggressive and deadly disease. Soaring 50% in young women since 1980, the incidence among people under age 30 is increasing faster than any other demographic group. Melanoma is the most common form of cancer for young adults between the ages of 25 and 29 and the second most common cancer in adolescents and young adults between ages 15 and 29 [1].

Melanoma is most common in Caucasians, striking men and women of all ages, races, and skin types. The biology of melanoma differs for intermittent, sun-damaged skin and for acral/mucosal sites where solar radiation appears to be a less important etiologic factor, and the mutation of genes such as c-KIT and BRAF distinguish these forms of melanoma [2, 3]. Malignant transformation of melanocytes in the basal layer of the epidermis gives rise to cutaneous melanoma. Although clearly linked to ultraviolet radiation, other risk factors such as family history and the presence of clinically atypical and histopathologically dysplastic nevi, skin complexion also plays an important role.

Additional risk for development of melanoma is associated with the melanocortin-1 receptor gene variant (MC1R) that results in red hair, fair skin and freckles. In affected individuals, [lighter] pheomelanin pigment is produced instead of the [darker] eumelanin pigment [4]. While eumelanin has the ability to absorb UV radiation, pheomelanin gives rise to free radicals upon contact with UV radiation contributing to further skin damage. The development of more potent genetic analysis tools allows new insights into gene mutations that occur in melanoma. This recently discovered genetic information has given rise to many new drugs that have already made substantial progress in clinical trials [5].

The use of high-dose interferon (HDI) in high-risk melanoma has been controversial as adjuvant treatment for patients with high-risk melanoma. High-risk disease is defined as all melanomas with thick primary lesions (T4 N0 M0, greater than 4 mm depth) or any invasion depth with positive lymph nodes (T1–4 N1 M0) with 5-year recurrence risks of 60% and 75%, respectively (see Table 1). Adjuvant HDI therapy is associated with significant toxicity in relation to the constitutional, hematologic, hepatic, neurologic, cutaneous, musculoskeletal, gastrointestinal, cardiovascular, and renal systems, potentially affecting the ability to deliver the full course of therapy [6]. Despite these toxicities of the HDI regimen, several studies from 1999 to 2001 have documented the feasibility of this treatment for patients treated in cooperative group settings across the US, where 90% of patients were able to complete therapy in the largest and most recently completed E1694 HDI trial [7]. HDI toxicity is now largely manageable, using the published guidelines that have emerged from the USA, Canada, and Europe [6, 8, 9].

Table 1.

Melanoma-risk groups based on 5-year relapse risk

Melanoma risk group Stage

Low IA–B
Intermediate IIA–IIIA
High IIIB–IV

The immune system plays a crucial role in the progression and pathogenesis of melanoma, but the exact mechanisms necessary to determine the outcome of the early disease are not fully understood. Rare cases of spontaneous regression of metastatic melanoma have been reported. Recent literature reviews in 2009 analyzed 76 published cases but did not find a particular mechanism for this phenomenon [10]. Between 10% and 35% of all melanomas show regression upon pathology review; however, such regression is generally considered to be associated with adverse outcomes [11]. Although other published studies do not uniformly support this belief [12, 13], the most recent 2009 American Joint Committee on Cancer compilation reported that no adverse impact of regression is apparent [14]. This suggests the direction of the tumor evasion from appropriate host immune response.

During the first trials with high-dose interleukin-2 (IL-2) and lymphokine-activated killer cells in patients with metastatic melanoma and renal cell cancer, a percentage of patients developed autoimmune thyroiditis as a result of the treatment [15]. This prompted a clearer focus on the phenomenon of autoimmunity than previous findings associated with favorable outcomes in the context of IL-2 therapy [16]. Autoimmunity can be seen as an example of overreaction of the immune system. A fine balance between regulatory (CD 25+, CD4+) and self-reactive/effector T cells prevents immune response to the normal host tissue in the human body [17].

Evidence implicates that regulatory T cells fulfill a unique role, controlling immune responses that are, in general, either physiologic or pathologic [18]. Depletion of regulatory T cells in otherwise normal mice causes autoimmune diseases [19, 20]. Regulatory mechanisms involving T regulatory cells that are marked by FOXP3—a transcription factor that is essential for development and function of regulatory T cells—are one component that has only recently been evaluated [21, 22]. Mutation of the FOXP3 gene causes a clinical syndrome called IPEX (immune dysregulation, polyendocrinopathy, enteropathy, and X-linked), that provides further evidence of its importance in the regulation of the immune system [23].

Another important player in immune regulation is the regulatory T cell receptor CTLA-4, already prevalent as a target in ongoing malignant melanoma trials [5]. FOXP3 and CTLA-4 expression on T cells appear to be corollaries. For example, terminally differentiated CD4+ T cells express FOXP3 highly and CD25+ constitutively expresses CTLA-4. Blockage of CTLA-4 with monoclonal antibodies causes organ-specific autoimmune disease and inflammatory bowel disease in otherwise healthy mice. Furthermore, mice lacking CTLA-4 on their regulatory T cells develop similar autoimmune disease as their FOXP3-deficient counterparts. T cell inhibitory receptor programmed death-1 (PD-1) is a more recent target for antibody blockage (MDX-1106, Bristol–Myers Squibb) currently undergoing a phase-I trial in solid tumors [24]. Recent laboratory evidence suggests that blocking the T cell immunoglobulin mucin (TIM) 3 receptor together with PD-1 receptor is capable of reversing T cell dysfunction in melanoma patients [25]. The importance of immunoregulation and the emergence of pharmacological means to modulate these, in context with current melanoma therapies, make it critical to evaluate the process of autoimmunity.

Interferon

Interferon (IFN) was discovered more than 50 years ago as a substance that showed inhibition of viral replication [26]. Two subtypes have been developed for therapeutic purposes so far, interferon-α−2-a (Roferon-A®, Roche Pharmaceuticals, Basel, Switzerland) and interferon-α−2-b (Intron-A) Schering Plough, Kenilworth, NJ, USA). These two IFNs differ from each other in only 2 of the 166 amino acids, and are nearly identical in therapeutic effects and toxicity profiles. All type I IFNs bind to a specific cell surface receptor complex known as the IFN-α receptor (IFNAR) [27]. The IFN-α receptor is widely expressed on both tumor cells and immune effector cells. It mediates most of its effects via activation of the Janus kinase (JAK) that further recruits transcription of the STAT pathway [2830]. The STAT factors form dimers, which are transported to the nucleus where they modulate the interferon response elements (ISREs) in combination with stabilizing molecules. These IFN-stimulated response elements act as transcription factors to activate a number of genes that ultimately lead to interferon-α production. Effects include the inhibition of viral growth, growth inhibition of tumor cells, and immunostimulatory effects by activation of different cytokines [28].

In the early 1980s [31], Bart et al. demonstrated promising effects of IFN-α upon melanoma cells in vitro in the murine B16 melanoma cell line. The observation of clinical anti-tumor effects of IFN in the first phase I and phase II trials ultimately led to the pursuit of this agent in earlier stages of melanoma in the cooperative groups [32, 33]. Three US cooperative group studies have evaluated high-dose IFN-α−2b (HDI) for the treatment of high-risk melanoma in the past [7, 34, 35]. The pivotal ECOG trial E1684 reported in 1996 was the first prospective trial (n=280) that investigated the HDI regimen currently approved in the USA this regimen [of IFN-α−2b at 20 million units (MU)/m2/d intravenously (IV) 5 days/week for 4 weeks]) followed by 10 MU/m2/d subcutaneously (SC) three times weekly for 48 weeks was compared versus observation [34].

The median relapse-free survival (RFS) time in patients receiving IFN was 1.72 years, while median for those in the observation arm was 0.98 years, respectively. The overall median survival time was 3.82 years in the IFN group as opposed to 2.78 years in the observation group. The following ECOG trial E1690 was a prospective, randomized and three-arm study that evaluated HDI, low-dose IFN, and observation in high-risk melanoma (clinical stage IIIB/IV) [35] (see Table 2). Adjuvant IFN-α therapy has been hypothesized to operate through multiple mechanisms including antiviral, antivascular, pro-apoptotic, and immunomodulatory effects. To date, immunomodulatory effects appear to be most closely linked to its clinical anti-tumor effects in node-metastatic melanoma [36, 37].

Table 2.

Comparison of study characteristics analyzing statistical significance of autoimmunity in melanoma in interferon therapy

Study characteristics Gogas et al. 2006 Bouwhuis et al. 2009 Bouwhuis et al. 2010

Study size/patient samples analyzed 364/200a 1388 +855/187 +356b 1256/296
Pathology review Centralized Multiple centers Multiple centers
Autoantibody testing Accredited clinical laboratory Research laboratory Research laboratory
Interferon dosing Modified high-dose regimen (IV, SC) Intermediate-dose (SC only) Intermediate-dose (SC only)
a

Substudy of a larger clinical trial that was conducted prospectively at a single institution

b

In this study, patient samples were obtained from two separate clinical trials (EORTC 18952 and Nordic IFN trial)

Therapy in the adjuvant setting of potentially curative operable disease aims at inducing host response that may eradicate micrometastatic disease, i.e., the presumed source of future disease recurrence [7]. Results showed a dose-dependent and significant benefit on RFS in the HDI group. The ECOG trial E1694 high-dose IFN showed significant benefit in terms of RFS and overall survival when compared to GMK ganglioside vaccine [7]. Less-intensive regimens tested in the melanoma adjuvant setting have not demonstrated durable effects upon relapse or death as shown with HDI.

These include intermediate-dose IFN (given subcutaneously) regimens tested in the European Organization for Research and Treatment of Cancer (EORTC) 18952 (T4, N1–2) [38]. The EORTC 18952 trial randomized patients to intermediate-dose IFN (10 MU/d for 5 days per week for total of 4 weeks followed by 10 MU three times a week for 1 year or 5 MU for three times a week for 2 years) versus observation only. Results of this trial, using lower IFN dosing when compared to the previously mentioned ECOG studies, did not show statistically significant distant metastasis-free interval and overall survival. Despite positive data from recent clinical trials of other immunotherapies in advanced inoperable melanoma, Interferon-α−2b (IFN-α−2b) remains the only agent approved by regulatory authorities worldwide (United States Food and Drug Administration [FDA]) for adjuvant therapy of high-risk melanoma to date.

The first large study that looked at autoimmunity associated with HDI therapy in patients with high-risk melanoma was published by Gogas et al. in 2006 [39]. The parent trial enrolled 364 patients in 13 institutions, and the autoimmunity sub-study analyzed 200 patients enrolled at 1 institution. Results showed significant survival benefit in patients that developed serum auto-antibodies on HDI therapy and will be discussed in more details in a later section.

Pegylated interferon

Pegylated interferon (PEG-IFN), widely used for the treatment of hepatitis, has not been studied as extensively as recombinant HDI in the treatment of high-risk melanoma [40, 41]. Available evidence suggests that the schedule of administration for this agent is more convenient than HDI, but the acute and chronic toxicity do not appear to differ qualitatively. The favorable pharmacokinetic properties of PEG-IFN allow its administration on a weekly basis with sustained exposure to IFN during that entire period at modest ambient levels [42].

The large size of the PEG-IFN molecule results in a 100-fold reduction in renal clearance when compared with conventional IFN [43]. Peak plasma levels of PEG-IFN are achieved approximately 80 h post-administration, whereas conventional intravenous IFN reaches its peak serum levels within minutes; subcutaneous administration does so within 7–12 h after administration, due to absorption and elimination kinetics given by the IV and SC routes. The role of the high concentrations of IFN achieved by the IV route of administration in the induction phase of the HDI therapy has no counterpart in therapy given with lower doses administered by the SC route alone or the kinetics of PEG-IFN, and the induction phase has been a component of both trials that have shown independent positive results in terms of overall survival [34, 35].

The largest clinical trial to date using PEG-IFN reported by Eggermont and colleagues showed a significant improvement in recurrence-free survival, but did not show a significant impact on overall survival. Subset analyses suggest that the impact of this regimen is observed almost entirely among patients with microscopic lymph node disease [40].

Autoimmunity induced during interferon-α therapy

The evasion of host immunity appears to be crucial in the development and progression of melanoma. As reported nearly three decades ago [4446], clinical observations have suggested that the appearance of autoimmune vitiligo is a favorable prognostic factor in patients with melanoma. Results of later trials using high-dose IL-2 and HDI have further linked the appearance of autoimmunity with improved outcome—although none of these trials were specifically designed to analyze the appearance of autoimmunity [15, 16, 4751].

Until this time, the only large trial that prospectively investigated the appearance of clinical and laboratory features of autoimmunity during HDI treatment in patients with high-risk melanoma was published by Gogas and colleagues in 2006 [39]. This substudy was partially conducted by the Hellenic Cooperative Group (HECOG) trial at 13 institutions that enrolled 364 patients with high-risk melanoma. Patients received either a month-long or year-long course of a modified HDI regimen. Blood samples from the substudy population of 200 patients enrolled at the First University of Athens, were collected for autoantibody testing against six autoantigens prior to, during, and up to 12 months after initiation of HDI therapy. Patients with preexisting autoantibodies were excluded. Regardless of the IFN arm, autoantibodies (or clinical manifestations of autoimmunity) were detected in 52 patients. At 45.6 months of median follow-up, only 7 of 52 patients in the autoimmunity group developed relapse, whereas 108 of 148 patients without autoimmunity relapsed.

Similarly, outcomes in overall survival were 2 of 52 patients dying in the autoimmunity group, whereas 80 of 148 patients without autoimmunity died. In multivariate analyses, autoimmunity was found to be an independent prognostic marker for improved RFS and overall survival (P<0.001). A retrospective analysis exploring evidence autoimmunity (testing for three autoantibodies) in patients from the EORTC 18952 and the Nordic IFN trial did not demonstrate a statistically significant correlation of autoimmunity and RFS [52].

A more detailed evaluation of the study characteristics reveals some important differences from the Gogas study. Although enrollment criteria for high-risk melanoma patients was very similar, the EORTC study tested only a small subset of patients enrolled in the two clinical trials (see Table 2). Patient samples were obtained from 27 different collaborating centers for central testing of autoantibodies, but each enrolled patient had local pathology assessment of their melanoma at the collaborating center. Patient sera were analyzed by the same enzyme-linked immunoabsorbent assay (Quanta Lite; Inova Diagnostics, San Diego, CA). Tests by the Gogas study were performed in an accredited hospital laboratory, whereas those in the Bouwhuis study were completed in a non-accredited/non-standardized research laboratory.

Unlike the Gogas et al. study, Bouwhuis and colleagues did not report data on clinical manifestations of autoimmunity, only three autoantibodies were screened (see Table 3) and seroconversion occurred in 34% and 32% of the EORTC 18952 and Nordic IFN trials, respectively. This is a significantly higher number than the 26% rate of overall seroconversion and clinical autoimmune disorders reported by Gogas et al. Bouwhuis’ study further analyzed results using three different statistical models. Model 1, analyzed for “any positive autoantibody test” (time-independent Cox model), showed statistical significance for recurrence-free survival. To address guarantee-time bias phenomenon, they used time-dependent Cox models that tested in model 2 for “latest positive autoantibody status” and in model 3 for “latest autoantibody status.” Analysis using models 2 and 3 did not enable this group to confirm the previously reported statistical significance for correlations between autoimmunity and improved recurrence-free survival.

Table 3.

Laboratory and clinical parameters assessed in each study to detect autoimmunity

Gogas et al. 2006 Bouwhuis et al. 2009

Antithyroglobulin antibodies Antityroglobulin antibodies
Antinuclear antibodies Antinuclear antibodies
Anticardiolipin antibodies (IgM and IgG) Anticardiolipin (IgA, IgM, and IgG)
Anti-DNA antibodies
TSH
Thyroxine
Triiodothyronine
Clinical evaluation for manifestations of autoimmunity done prospectively No prospective clinical evaluation

Further differences between these studies include the doses of IFN tested: Gogas used high dosages of interferon in the induction phase (IV) and maintenance dosages (SC), whereas the Bouwhuis group studied therapy employing intermediate doses of interferon for induction and maintenance phases. A more recent study by Bouwhuis examined the prognostic value of autoantibody responses in patients with high-risk melanoma receiving PEG-IFN, which also did not detect improved outcomes associated with autoimmunity [53]. As with the prior study of this group, only a small subset of patients from the original study cohort was analyzed using research laboratory tests for autoantibodies, rather than GLP clinical laboratory assays. Direct comparison between the two studies is difficult, as PEG-IFN has different pharmacokinetics compared to non-pegylated IFN.

Discussion

Despite ongoing clinical trials and reports of new promising therapeutics, HDI remains the only regimen approved for adjuvant treatment of high-risk melanoma (Table 4). Based on the results of three ECOG and intergroup studies with HDI, the therapeutic efficacy of interferon appears to exceed other regimens, and is associated with treatment of 1 year’s duration. Rate-limiting features of this regimen include the side effects for patients, although more than 90% of patients now complete the planned regimen.

Table 4.

Interferon-α-2b-dosing regimens used in treatment of high-risk melanoma

Interferon regimen Duration/dosing schedule

High-dose IFN
 Induction IFN, 20 MU/m2 iv, 5 times per week for 4 weeks
 Maintenance IFN, 10 MU/m2 sc, 3 times per week for 48 weeks
Intermediate-dose IFN
 Induction IFN, 10 MU/m2 iv, 5 times per week for 4 weeks
 Maintenance IFN, 5–10 MU/m2 sc, 3 times per week for 48 weeks
 Low-dose IFN IFN 3 MU/m2 sc, 3 times per week (varying duration)
PEG-IFN
 Induction PEG-IFN 6 μgram/kg sc, weekly for 8 weeks
 Maintenance PEG-IFN 3 μgram/kg sc, weekly for up to 5 years

Overall, clinical benefits from HDI therapy amount to a reduction in the vulnerability for relapse by one-third (E1684, E1690, E1694) and reduction of mortality by one- quarter to one-third (E1684, E1694). Attempts to select a population of melanoma patients that might respond more favorably to HDI treatment has led to the exploration of autoimmunity, which has anecdotally been associated with improved outcome of autoimmunity as reported by multiple investigators [15]. Gogas and colleagues conducted a well-designed prospective study that analyzed 200 patients and showed a significant benefit of HDI therapy in patients who developed autoimmunity during therapy.

Subsequently, Bouwhuis and colleagues reported that a subset of patients from larger EORTC studies did not show the same statistical correlation of favorable outcomes associated with autoimmunity. Stuckert and colleagues presented further evidence during the 2007 ASCO annual meeting. Their findings showed autoimmunity as a predictive biomarker in recurrent-free survival in HDI of the E2696 trial [54]. Final analyses of this trial are pending. Staging of malignant melanoma specimens in the trials of HECOG, ECOG, and EORTC include centralized staging, whereas Bouwhuis et al. conducted staging in various institutions.

Differences are also seen in laboratory tests of specimens from the previously discussed clinical studies. While Gogas and colleagues were using an accredited clinical laboratory for all their specimen testing, Bouwhuis and colleagues used non-accredited experimental laboratories. Thus, it is difficult to ascertain if the analyses limited to only three autoantibodies were able to generate enough statistical data for comparison with more rigorous tests from the Gogas experiment.

Interestingly, the rates of seroconversion are much higher in the Bouwhuis study. Bouwhuis acknowledges these differences between the two studies, and while the same assays were reportedly used, there is no clear explanation for this discrepancy. Finally, the trials have employed different dosage regimens. The Gogas study used a higher-dose IFN regimen, while E18952 used intermediate-IFN dosages and E18991 used PEG-IFN at intermediate dosages. Several points require exploration in future studies. Larger trials using prospective enrollment with centralized pathology review and homogenous evaluation of specimens from all patients in clinically approved laboratories would avoid several pitfalls of the study design. In the absence of an overall survival impact of PEG-IFN, EORTC 18991 may not be the most promising regimen in which to validate a role of predictive biomarkers of therapeutic benefit.

Acknowledgments

This work was supported by Award Number P50CA121973 from the National Cancer Institute. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Cancer Institute or the National Institutes of Health.

Contributor Information

Michal T. Krauze, Melanoma Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.

Ahmad Tarhini, Melanoma Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.

Helen Gogas, Medical Oncology Division, 2nd Department of Medicine, University of Athens, Athens, Greece.

John M. Kirkwood, Melanoma Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA Hillman Cancer Center, University of Pittsburgh Cancer Institute, Research Suite L1.32c, 5117 Centre Avenue, Pittsburgh, PA 15213-2584, USA; Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.

References

  • 1.Weinstock MA (2006) Cutaneous melanoma: public health approach to early detection. Dermatol Ther 19:26–31 [DOI] [PubMed] [Google Scholar]
  • 2.Curtin JA, Busam K, Pinkel D, Bastian BC (2006) Somatic activation of KIT in distinct subtypes of melanoma. J Clin Oncol 24:4340–4346 [DOI] [PubMed] [Google Scholar]
  • 3.Davies H, Bignell GR, Cox C et al. (2002) Mutations of the BRAF gene in human cancer. Nature 417:949–954 [DOI] [PubMed] [Google Scholar]
  • 4.Valverde P, Healy E, Sikkink S et al. (1996) The Asp84Glu variant of the melanocortin 1 receptor (MC1R) is associated with melanoma. Hum Mol Genet 5:1663–1666 [DOI] [PubMed] [Google Scholar]
  • 5.Hodi FS, O’Day SJ, McDermott DF et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hauschild A, Gogas H, Tarhini A et al. (2008) Practical guidelines for the management of interferon-alpha-2b side effects in patients receiving adjuvant treatment for melanoma: expert opinion. Cancer 112:982–994 [DOI] [PubMed] [Google Scholar]
  • 7.Kirkwood JM, Ibrahim JG, Sosman JA et al. (2001) 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 19:2370–2380 [DOI] [PubMed] [Google Scholar]
  • 8.Caraceni A, Gangeri L, Martini C et al. (1998) Neurotoxicity of interferon-alpha in melanoma therapy: results from a randomized controlled trial. Cancer 83:482–489 [DOI] [PubMed] [Google Scholar]
  • 9.Kirkwood JM, Bender C, Agarwala S et al. (2002) Mechanisms and management of toxicities associated with high-dose interferon alfa-2b therapy. J Clin Oncol 20:3703–3718 [DOI] [PubMed] [Google Scholar]
  • 10.Kalialis LV, Drzewiecki KT, Klyver H (2009) Spontaneous regression of metastases from melanoma: review of the literature. Melanoma Res 19:275–282 [DOI] [PubMed] [Google Scholar]
  • 11.Requena C, Botella-Estrada R, Traves V, Nagore E, Almenar S, Guillen C (2009) Problems in defining melanoma regression and prognostic implication. Actas Dermosifiliogr 100:759–766 [PubMed] [Google Scholar]
  • 12.Morris KT, Busam KJ, Bero S, Patel A, Brady MS (2008) Primary cutaneous melanoma with regression does not require a lower threshold for sentinel lymph node biopsy. Ann Surg Oncol 15:316–322 [DOI] [PubMed] [Google Scholar]
  • 13.Fontaine D, Parkhill W, Greer W, Walsh N (2003) Partial regression of primary cutaneous melanoma: is there an association with sub-clinical sentinel lymph node metastasis? Am J Dermatopathol 25:371–376 [DOI] [PubMed] [Google Scholar]
  • 14.Balch CM, Gershenwald JE, Soong SJ et al. (2009) Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol 27:6199–6206 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Atkins MB, Mier JW, Parkinson DR, Gould JA, Berkman EM, Kaplan MM (1988) Hypothyroidism after treatment with interleukin-2 and lymphokine-activated killer cells. N Engl J Med 318:1557–1563 [DOI] [PubMed] [Google Scholar]
  • 16.Weijl NI, Van der Harst D, Brand A et al. (1993) Hypothyroidism during immunotherapy with interleukin-2 is associated with antithyroid antibodies and response to treatment. J Clin Oncol 11:1376–1383 [DOI] [PubMed] [Google Scholar]
  • 17.Wing K, Sakaguchi S (2010) Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol 11:7–13 [DOI] [PubMed] [Google Scholar]
  • 18.Sakaguchi S, Sakaguchi N (2005) Regulatory T cells in immunologic self-tolerance and autoimmune disease. Int Rev Immunol 24:211–226 [DOI] [PubMed] [Google Scholar]
  • 19.Singh B, Read S, Asseman C et al. (2001) Control of intestinal inflammation by regulatory T cells. Immunol Rev 182:190–200 [DOI] [PubMed] [Google Scholar]
  • 20.Sakaguchi S (2005) Naturally arising Foxp3-expressing CD25+ CD4+ regulatory T cells in immunological tolerance to self and non-self. Nat Immunol 6:345–352 [DOI] [PubMed] [Google Scholar]
  • 21.Ascierto PA, Streicher HZ, Sznol M (2010) Melanoma: a model for testing new agents in combination therapies. J Transl Med 8:38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hori S, Takahashi T, Sakaguchi S (2003) Control of autoimmunity by naturally arising regulatory CD4+ T cells. Adv Immunol 81:331–371 [DOI] [PubMed] [Google Scholar]
  • 23.Gambineri E, Torgerson TR, Ochs HD (2003) Immune dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX), a syndrome of systemic autoimmunity caused by mutations of FOXP3, a critical regulator of T-cell homeostasis. Curr Opin Rheumatol 15:430–435 [DOI] [PubMed] [Google Scholar]
  • 24.Sznol M, Powderly JP, Smith DC et al. (2010) Safety and antitumor activity of biweekly MDX-1106 (Anti-PD-1, BMS-936558/ONO-4538) in patients with advanced refractory malignancies (ASCO 2010). J Clin Oncol 28:15s, suppl; abstr2506 [Google Scholar]
  • 25.Fourcade J, Sun Z, Benallaoua M et al. (2010) Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med 207:2175–2186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Isaacs A, Lindenmann J (1957) Virus interference. I. The interferon. Proc R Soc Lond B Biol Sci 147:258–267 [PubMed] [Google Scholar]
  • 27.Uze G, Lutfalla G, Mogensen KE (1995) Alpha and beta interferons and their receptor and their friends and relations. J Interferon Cytokine Res 15:3–26 [DOI] [PubMed] [Google Scholar]
  • 28.Darnell JE Jr, Kerr IM, Stark GR (1994) Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264:1415–1421 [DOI] [PubMed] [Google Scholar]
  • 29.Haque SJ, Williams BR (1998) Signal transduction in the interferon system. Semin Oncol 25:14–22 [PubMed] [Google Scholar]
  • 30.Kaehler KC, Sondak VK, Schadendorf D, Hauschild A (2010) Pegylated interferons: prospects for the use in the adjuvant and palliative therapy of metastatic melanoma. Eur J Cancer 46:41–46 [DOI] [PubMed] [Google Scholar]
  • 31.Bart RS, Porzio NR, Kopf AW, Vilcek JT, Cheng EH, Farcet Y (1980) Inhibition of growth of B16 murine malignant melanoma by exogenous interferon. Cancer Res 40:614–619 [PubMed] [Google Scholar]
  • 32.Creagan ET, Ahmann DL, Green SJ et al. (1984) Phase II study of recombinant leukocyte A interferon (rIFN-alpha A) in disseminated malignant melanoma. Cancer 54:2844–2849 [DOI] [PubMed] [Google Scholar]
  • 33.Ernstoff MS, Trautman T, Davis CA et al. (1987) A randomized phase I/II study of continuous versus intermittent intravenous interferon gamma in patients with metastatic melanoma. J Clin Oncol 5:1804–1810 [DOI] [PubMed] [Google Scholar]
  • 34.Kirkwood JM, Strawderman MH, Ernstoff MS, Smith TJ, Borden EC, Blum RH (1996) Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol 14:7–17 [DOI] [PubMed] [Google Scholar]
  • 35.Kirkwood JM, Ibrahim JG, Sondak VK et al. (2000) High- and low-dose interferon alfa-2b in high-risk melanoma: first analysis of intergroup trial E1690/S9111/C9190. J Clin Oncol 18:2444–2458 [DOI] [PubMed] [Google Scholar]
  • 36.Moschos SJ, Edington HD, Land SR et al. (2006) Neoadjuvant treatment of regional stage IIIB melanoma with high-dose interferon alfa-2b induces objective tumor regression in association with modulation of tumor infiltrating host cellular immune responses. J Clin Oncol 24:3164–3171 [DOI] [PubMed] [Google Scholar]
  • 37.Wang W, Edington HD, Rao UN et al. (2007) Modulation of signal transducers and activators of transcription 1 and 3 signaling in melanoma by high-dose IFNalpha2b. Clin Cancer Res 13:1523–1531 [DOI] [PubMed] [Google Scholar]
  • 38.Eggermont AM, Suciu S, MacKie R et al. (2005) 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 366:1189–1196 [DOI] [PubMed] [Google Scholar]
  • 39.Gogas H, Ioannovich J, Dafni U et al. (2006) Prognostic significance of autoimmunity during treatment of melanoma with interferon. N Engl J Med 354:709–718 [DOI] [PubMed] [Google Scholar]
  • 40.Eggermont AM, Suciu S, Santinami M et al. (2008) 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 372:117–126 [DOI] [PubMed] [Google Scholar]
  • 41.Kirkwood JM, Tawbi HA, Tarhini AA, Moschos SJ (2009) Does pegylated interferon alpha-2b confer additional benefit in the adjuvant treatment of high-risk melanoma? Nat Clin Pract Oncol 6:70–71 [DOI] [PubMed] [Google Scholar]
  • 42.Daud AI, Xu C, Hwu WJ, et al. (2010) Pharmacokinetic/pharmacodynamic analysis of adjuvant pegylated interferon alpha-2b in patients with resected high-risk melanoma. Cancer Chemother Pharmacol (in press) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zeuzem S, Welsch C, Herrmann E (2003) Pharmacokinetics of peginterferons. Semin Liver Dis 23(Suppl 1):23–28 [DOI] [PubMed] [Google Scholar]
  • 44.Nordlund JJ, Kirkwood JM, Forget BM, Milton G, Albert DM, Lerner AB (1983) Vitiligo in patients with metastatic melanoma: a good prognostic sign. J Am Acad Dermatol 9:689–696 [DOI] [PubMed] [Google Scholar]
  • 45.Bystryn JC, Rigel D, Friedman RJ, Kopf A (1987) Prognostic significance of hypopigmentation in malignant melanoma. Arch Dermatol 123:1053–1055 [PubMed] [Google Scholar]
  • 46.Schallreuter KU, Levenig C, Berger J (1991) Vitiligo and cutaneous melanoma. A case study. Dermatologica 183:239–245 [DOI] [PubMed] [Google Scholar]
  • 47.Becker JC, Winkler B, Klingert S, Brocker EB (1994) Anti-phospholipid syndrome associated with immunotherapy for patients with melanoma. Cancer 73:1621–1624 [DOI] [PubMed] [Google Scholar]
  • 48.Krouse RS, Royal RE, Heywood G et al. (1995) Thyroid dysfunction in 281 patients with metastatic melanoma or renal carcinoma treated with interleukin-2 alone. J Immunother Emphasis Tumor Immunol 18:272–278 [DOI] [PubMed] [Google Scholar]
  • 49.Phan GQ, Attia P, Steinberg SM, White DE, Rosenberg SA (2001) Factors associated with response to high-dose interleukin-2 in patients with metastatic melanoma. J Clin Oncol 19:3477–3482 [DOI] [PubMed] [Google Scholar]
  • 50.Scalzo S, Gengaro A, Boccoli G et al. (1990) Primary hypothyroidism associated with interleukin-2 and interferon alpha-2 therapy of melanoma and renal carcinoma. Eur J Cancer 26:1152–1156 [DOI] [PubMed] [Google Scholar]
  • 51.Vallisa D, Cavanna L, Berte R, Merli F, Ghisoni F, Buscarini L (1995) Autoimmune thyroid dysfunctions in hematologic malignancies treated with alpha-interferon. Acta Haematol 93:31–35 [DOI] [PubMed] [Google Scholar]
  • 52.Bouwhuis MG, Suciu S, Collette S et al. (2009) Autoimmune antibodies and recurrence-free interval in melanoma patients treated with adjuvant interferon. J Natl Cancer Inst 101:869–877 [DOI] [PubMed] [Google Scholar]
  • 53.Bouwhuis MG, Suciu S, Testori A et al. (2010) Phase III trial comparing adjuvant treatment with pegylated interferon Alfa-2b versus observation: prognostic significance of autoantibodies–EORTC 18991. J Clin Oncol 28:2460–2466 [DOI] [PubMed] [Google Scholar]
  • 54.Stuckert JJ, Tarhini AA, Kirkwood JM (2007) Interferon alfa-induced autoimmunity and serum S100 levels as predictive and prognostic biomarkers in high-risk melanoma in the ECOG-intergroup phase II trial E2696 (ASCO 2007). J Clin Oncol 25:473s [Google Scholar]

RESOURCES