Immunogenicity and Antitumor Effects of Vaccination with Peptide Vaccine +/− Granulocyte-Monocyte Colony-Stimulating Factor and/or IFN-α2b in Advanced Metastatic Melanoma: Eastern Cooperative Oncology Group Phase II Trial E1696 (original) (raw)

Skip Nav Destination

Cancer Therapy: Clinical| February 19 2009

John M. Kirkwood;

1University of Pittsburgh, Pittsburgh, Pennsylvania;

Search for other works by this author on:

Sandra Lee;

2Dana-Farber Cancer Institute, Boston, Massachusetts;

Search for other works by this author on:

Stergios J. Moschos;

1University of Pittsburgh, Pittsburgh, Pennsylvania;

Search for other works by this author on:

Mark R. Albertini;

3University of Wisconsin, Madison, Wisconsin;

Search for other works by this author on:

John C. Michalak;

4Siouxland Hematology Oncology Associates, Sioux City, Idaho; and

Search for other works by this author on:

Cindy Sander;

1University of Pittsburgh, Pittsburgh, Pennsylvania;

Search for other works by this author on:

Theresa Whiteside;

1University of Pittsburgh, Pittsburgh, Pennsylvania;

Search for other works by this author on:

Lisa H. Butterfield;

1University of Pittsburgh, Pittsburgh, Pennsylvania;

Search for other works by this author on:

Louis Weiner

5Fox Chase Cancer Center, Philadelphia, Pennsylvania

Search for other works by this author on:

Crossmark: Check for Updates

Requests for reprints: John M. Kirkwood, Department of Medicine, Melanoma and Skin Cancer Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Research Pavilion, Suite 132, 5117 Centre Avenue, Pittsburgh, PA 15213-2584. Phone: 412-623-7707; Fax: 412-623-7704; E-mail: KirkwoodJM@upmc.edu.

Received: June 25 2008

Revision Received: November 03 2008

Accepted: November 06 2008

Online ISSN: 1557-3265

Print ISSN: 1078-0432

American Association for Cancer Research

2009

Clin Cancer Res (2009) 15 (4): 1443–1451.

Article history

Revision Received:

November 03 2008

Accepted:

November 06 2008

Abstract

Purpose: No therapy has ever shown prolongation of survival in stage IV metastatic melanoma. The association of cytokine-induced autoimmunity with improved prognosis led us to investigate the effect of multi-epitope melanoma vaccines alone and in combination with cytokines in this Eastern Cooperative Oncology Group multicenter phase II trial.

Experimental Design: Eligible patients were required to have failed prior therapies and to be HLA-A2 positive. Three HLA class I-restricted lineage antigen epitopes were administered in a factorial 2 × 2 design. Peptide vaccine alone (arm A), or combined with granulocyte-monocyte colony-stimulating factor (GM-CSF; Immunex) 250 μg/d subcutaneously for 14 of 28 days each month (arm B), or combined with IFN-α2b (Intron A; Schering-Plough) 10 million units/m2 three times a week (arm C), or combined with both IFN-α2b and GM-CSF (arm D). The primary endpoint was immune response measured by enzyme-linked immunospot assay; secondary endpoints were clinical antitumor response, disease-free survival, and overall survival.

Results: One hundred twenty patients enrolled and 115 patients were analyzed. Immune responses to at least one melanoma antigen were observed in 26 of 75 (35%) patients with serial samples. Neither IFN-α2b nor GM-CSF significantly improved immune responses. Six objective clinical responses were documented. At a median follow-up of 25.4 months, the median overall survival of patients with vaccine immune response was significantly longer than that of patients with no immune response (21.3 versus 13.4 months; P = 0.046).

Conclusion: Immune response to vaccination correlates with prolonged survival in patients with metastatic melanoma and is not enhanced by immunomodulatory cytokines as tested in this trial.

Translational Relevance

The results of the Eastern Cooperative Oncology Group multi-epitope peptide vaccine trial E1696 trial reported in this article have translational implications for the future therapy of melanoma in several respects: the results show the feasibility and immunologic efficacy of multi-epitope peptide vaccination, providing the foundation for adjuvant studies of this multi-epitope peptide vaccine that are ongoing in the intergroup. In summary, the results (a) show the feasibility of vaccination with rigorous immunologic assessments in the national cooperative groups, coupled with either granulocyte-monocyte colony-stimulating factor or IFN-α2b systemic therapy tested in a factorial 2 × 2 design. This study of 120 subjects who had failed prior therapy shows (b) that multi-epitope peptide vaccination induces immune response in a significant fraction of patients determined using complex enzyme-linked immunospot immunologic assays done in a central reference laboratory. The modulation of immune responses assessed by enzyme-linked immunospot assays of T-cell IFN-γ production is correlated with improved overall survival. Finally, this study (c) provides the foundation in advanced metastatic melanoma for multi-epitope peptide vaccination that has now been evaluated using a placebo-controlled design testing granulocyte-monocyte colony-stimulating factor and/or peptide vaccination in 815 patients with operable stage III to IV melanoma (Intergroup E4697).

There is no therapy to date that has shown a significant effect on overall survival (OS) in patients with metastatic melanoma. Only high-dose bolus interleukin-2 has been approved by the U.S. Food and Drug Administration for therapy of advanced melanoma based on durable responses induced in 4.7% to 6% of patients (1). Furthermore, in the adjuvant setting of operable high-risk melanoma, rigorous randomized phase III trials of high-dose IFN-α2b have shown both consistent relapse-free survival (RFS) and OS benefit in two of these trials (2). Granulocyte-monocyte colony-stimulating factor (GM-CSF) is another cytokine that has suggested RFS and OS benefit in a single phase II study of patients with operable resected stages III and IV melanoma compared with historic controls (3). These observations led us to design an intergroup trial of GM-CSF given in monthly cycles for treatment of patients with resectable advanced stage III and limited stage IV melanoma (4).

The common denominator of therapies of benefit in melanoma has been the augmentation of host immunity. High-dose IFN given in the neoadjuvant setting induces higher increases in the number of tumor-infiltrating CD3+ and CD11c+ immune cells in clinical responders compared with nonresponders (5). Similarly, high-dose bolus interleukin-2 induces inflammation at the tumor site (6), whereas GM-CSF has potent effects on myeloid dendritic cells (7, 8). These cytokines have been shown to induce autoimmunity that is associated with clinical benefit (911). Concurrent or sequential administration of cytokines with melanoma vaccines in small studies has suggested augmented immunologic response in association with clinically significant antitumor responses (12, 13).

Vaccines have become the focus of major investigative efforts across the world, driven by the expanding understanding of the immunogenicity of melanoma antigens (14). Most previously reported vaccine studies have been conducted at single institutions, employing one or a limited panel of peptide antigens, with a variety of immunologic adjuvants, resulting in occasional clinical benefit (1519). These limited benefits have been attributed to regulatory T cells, the low frequencies of antigen-specific T cells, and to defects in immune cell trafficking and function (20). A critical need has therefore developed for well-powered randomized studies testing the immunodominant melanoma peptide epitopes along with novel vaccination strategies to produce higher frequencies and more durable immune responses. No cooperative group trial has yet evaluated the immunogenicity of vaccines composed of multiple lineage antigens, or compared vaccination alone, versus in combination with immunomodulatory cytokines.

E1696 was designed to test immune responses of peptide-specific CD8+ T-cell precursors by enzyme-linked immunospot (ELISPOT) assay against epitopes derived from three lineage-restricted antigens MART-1, gp100, and tyrosinase in patients with metastatic unresectable melanoma. The peptide epitopes were selected based on the high frequency of the anti-melanoma CTL clones derived from HLA-A2-positive tumor-infiltrating lymphocytes against antigenic sequences identified by both cDNA expression cloning and protein biochemical approaches (18, 2128). Given the promise of two immunomodulatory biological agents available in the outpatient setting, this study was also designed to evaluate the immunomodulatory effects of GM-CSF, IFNα, or the combination of these two agents on vaccine immune response and toxicity. Finally, analyses of antitumor responses and correlations of immune response with survival outcome were explored in this multicenter randomized trial.

Patients and Methods

Patients. Eligible patients had histologically confirmed stage IV melanoma [American Joint Committee on Cancer (AJCC) 5th edition], absent brain metastases by magnetic resonance imaging or computed tomography scans within 4 weeks before randomization, and measurable disease. Prior treatments were allowed if administered >4 weeks before randomization, including IFN or GM-CSF. Patients needed to have an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, to have adequate hematologic (WBC >4,000/mm3, platelet count >100,000/mm3, and lymphocytes >700/mm3), hepatic (aspartate aminotransferase, serum bilirubin, alkaline phosphatase, and lactate dehydrogenase ≤2 times institutional upper limit), and renal (serum creatinine <1.8 mg/dL) functions within 4 weeks before randomization, and to be HLA-A2 positive by serologic or genotypic analysis. Patients with HIV, other current malignancies active autoimmune disorders, or other conditions requiring chronic corticosteroid use were excluded. All patients provided written informed consent, and the study was approved by the institutional review board of each participating ECOG-affiliated institution.

Peptides. HLA-A2-restricted peptides used in this study included AAGIGILTV (MART-127-35; ref. 27), the enhanced gp100 peptide analogue (gp100 209, T210M) IMDQVPFSV (gp100209-217; ref. 18), and the tyrosinase peptide analogue tyrosinase 368-376 (370D) [NSC 699048] YMDGTMSQV (tyrosinase368-376; ref. 22). The peptides were synthesized and placed in vials containing 1 mL of a sterile 1 mg/mL solution for injections. All vaccine peptides and those used for ELISPOT assays were provided by the National Cancer Institute/Cancer Therapy Evaluation Program as Investigational New Drug 6123.

Immunization protocol. The schema of this study is illustrated in Fig. 1. After patients provided written informed consent, immunophenotypic or genotypic analysis for HLA-A2 status and radiographic studies were done. If HLA-A2 positive, additional peripheral blood was obtained for baseline immunologic assays, and patients were randomized at the ECOG operations office by telephone or through ECOG Web registration with equal weighting into one of the four treatment arms:

Fig. 1.

Fig. 1. Schema for ECOG phase II trial E1696.

Schema for ECOG phase II trial E1696.

Fig. 1.

Fig. 1. Schema for ECOG phase II trial E1696.

Schema for ECOG phase II trial E1696.

Close modal

The duration of therapy for all arms was 13 cycles (52 weeks). Patients who did not progress during treatment were followed every 3 to 24 months and every 6 to 60 months following study enrollment with physical exam and whole-body computed tomography scans.

HLA typing. HLA typing was done either genotypically using PCR (HLA-0201) or serologically using multiple antibodies (MA2.1, W6.32, BB7.2, and L243) for HLA-A2 with the respective isotype controls in the Immunological Monitoring Laboratory of ECOG at the University of Pittsburgh Cancer Institute. For genotypic analysis, we used PCR methods (HLA-A2 0201).

ELISPOT assays. Peripheral blood from patients was sent by overnight courier to the ECOG Core Immunological Reference Laboratory at the University of Pittsburgh Cancer Institute. Peripheral blood mononuclear cells were isolated by a Ficoll gradient centrifugation and cryopreserved. After completion of the vaccine regimen, samples from baseline and the days 43 and 85 time points were evaluated simultaneously by an IFN-γ (ELISPOT) assay (30). No in vitro sensitization of lymphocytes to enhance detection of low-frequency precursor T cells directed against melanoma lineage and other antigens was done, as this has been used for various intervals in multiple prior studies. We chose instead to avoid the artifacts associated with in vitro priming in this study where evaluation of T-cell responses was based on previously published definitions (16). The IFN-γ ELISPOT was done by carefully trained technologists, using standard operating procedures and validated reagents. Performance of different technologists involved in assay conduct was cross-compared. All plates were read by automated ELISPOT plate readers. Each assay contained wells in which normal healthy donor cells (made in large, multi-use batches) were tested for spontaneous and phorbol 12-myristate 13-acetate/ionomycin-stimulated cytokine release, serving as an assay control. Medium-only wells served as controls for nonspecific spots from other reagents.

Patient cells stimulated with OKT3 served as patient cell function controls over and above percent recovery and percent viability measures. Recall responses to flu peptide from CD8+ cells and nonspecific responses to Ova were also included. Wells were plated in triplicate and SDs were measured. All samples were assayed on the same plate on the same day to avoid interassay variation. Several internal controls were used to test for spontaneous release of IFN-γ as well as stimulation to test for the ability to mount an immune response. For example, test wells were plated with the responders alone, responders + antigen-presenting cells (no peptide), and responders + Ova protein to test for spontaneous IFN-γ production in the absence of a stimulus. In addition, test wells were plated with responders + OKT3, and a set of wells with flu peptide to insure an immune response could be elicited. All reagent lot numbers were tested before patient assays for quality assurance.

Evaluation of clinical response and statistical considerations. Objective antitumor response was evaluated in measurable disease by computed tomography or magnetic resonance imaging scans and by clinical measurement of cutaneous lesions using the standard ECOG tumor response criteria (ECOG Policies and Procedures Manual).

The primary endpoint of the study was peptide-specific immune response by treatment (IFN versus no IFN or GM-CSF versus no GM-CSF) in a 2 × 2 factorial design with IFN on one axis and GM-CSF on the other axis. ELISPOT assays were the sole measure of treatment-induced immune modulation in this study. For an increase of immunologic response from 5% in non-IFN or non-GM-CSF treatment arms to 30% in IFN or GM-CSF treatment arms, 18 fully evaluable patients per treatment arm, and a two-sided significance level of 10%, the power to detect such a difference using Fisher's exact test was 81%. The secondary endpoints of the study were assessment of progression-free survival (PFS) and OS for patients in all four treatment arms.

In analyzing the time-to-event data, the method of Kaplan-Meier was used and significant differences observed were tested by the log-rank test. The Cox proportional hazards model was used for a multivariate time-to-event analysis. Fisher's exact test was used to compare proportions. All P values were reported for two-sided tests.

Results

Accrual. This study accrued 120 patients in seven major institutions of ECOG between September 22, 2000 and May 14, 2003. One patient randomized to arm B never received any therapy due to insurance issues. Four patients were ineligible. For E1696, 115 cases were included in the analysis of clinical outcomes, whereas toxicity data were summarized for all 117 treated cases. Seventy-five patients had ELISPOT data taken at baseline and at least one later time point (day 43 or 85). The stage breakdown for 72 of 75 patients with full staging data was M1a [15 (20.8%)], M1b [30 (41.7%)], and M1c [27 (37.5%)].

Patient characteristics and immunologic response.Table 1 displays the on-study characteristics of the 115 treated patients who were analyzable for immune response. Seventy-five patients had ELISPOT data at baseline and for at least one later time point. Of these 75 patients, 66 patients had ELISPOT data at day 43 and 54 patients at day 85. Table 2 summarizes immune responses by ELISPOT data for each lineage-restricted melanoma antigen. The immune response rate to at least one of the peptides [overall immune response rate (OIRR)] was 21.2% at day 43 and 37% at day 85. The OIRR at day 43 or 85 was 34.7% (95 confidence interval, 24-46.6%). The immune response rate to gp100 was the highest compared with MART-1 and tyrosinase. There was a marginally significant association between the OIRR and the disease stage (AJCC 6th edition) on study enrollment (46.7% and 43.3% in patients with M1a and M1b disease, respectively, versus 20.7% in patients with M1c disease; P = 0.112). Neither IFN nor GM-CSF significantly increased the immune response rate. OIRR was 16 of 37 (43.2%) with IFN treatment in comparison with 9 of 36 (25%) without IFN treatment. By GM-CSF treatment, OIRR was 13 of 34 (38.2%) with GM-CSF and 12 of 39 (30.8%) without GM-CSF.

Table 1.

On-study characteristics (N = 115)

n (%)
Gender
Male 69 (60)
Female 46 (40)
Race (n = 110)
White 110 (100)
Performance status (n = 113)
0 74 (65.5)
1 39 (34.5)
Depth of invasion (n = 80)
I 1 (1.3)
II 8 (10.0)
III 22 (27.5)
IV 32 (40.0)
V 17 (21.3)
Primary tumor site (n = 106)
Head and neck 19 (17.9)
Upper limb 5 (4.7)
Lower limb 17 (16)
Trunk 46 (43.4)
Anagenital 2 (1.9)
Ocular 8 (7.6)
Mucosal 4 (3.8)
Other 5 (4.7)
Histologic type (n = 79)
Amelanotic 6 (7.6)
Melanotic 73 (92.4)
Sites of distant metastasis
Visceral involvement Measurable (n) Nonmeasurable (n)
Lung 70 11
Pleura 1 1
Liver 24 5
Brain/central nervous system 0 1
Bone 0 6
Other visceral 18 6
Nonvisceral involvement Measurable (n) Nonmeasurable (n)
Distant nodes 29 5
Soft tissue 23 6
Skin 19 2
Other nonvisceral 12 4
AJCC M stage (n = 111)
M1a 20 (18)
M1b 47 (42.4)
M1c 44 (39.6)
n (%)
Age, y
Median (range) 62 (23-83)
Breslow thickness, mm (n = 81)
Median (range) 2.5 (28-24)
Prior treatment
Surgery 109
Chemotherapy 24
Radiotherapy 22
Immunotherapy 44
Hormonal therapy 1
Gene therapy 1
Other 9
n (%)
Gender
Male 69 (60)
Female 46 (40)
Race (n = 110)
White 110 (100)
Performance status (n = 113)
0 74 (65.5)
1 39 (34.5)
Depth of invasion (n = 80)
I 1 (1.3)
II 8 (10.0)
III 22 (27.5)
IV 32 (40.0)
V 17 (21.3)
Primary tumor site (n = 106)
Head and neck 19 (17.9)
Upper limb 5 (4.7)
Lower limb 17 (16)
Trunk 46 (43.4)
Anagenital 2 (1.9)
Ocular 8 (7.6)
Mucosal 4 (3.8)
Other 5 (4.7)
Histologic type (n = 79)
Amelanotic 6 (7.6)
Melanotic 73 (92.4)
Sites of distant metastasis
Visceral involvement Measurable (n) Nonmeasurable (n)
Lung 70 11
Pleura 1 1
Liver 24 5
Brain/central nervous system 0 1
Bone 0 6
Other visceral 18 6
Nonvisceral involvement Measurable (n) Nonmeasurable (n)
Distant nodes 29 5
Soft tissue 23 6
Skin 19 2
Other nonvisceral 12 4
AJCC M stage (n = 111)
M1a 20 (18)
M1b 47 (42.4)
M1c 44 (39.6)
n (%)
Age, y
Median (range) 62 (23-83)
Breslow thickness, mm (n = 81)
Median (range) 2.5 (28-24)
Prior treatment
Surgery 109
Chemotherapy 24
Radiotherapy 22
Immunotherapy 44
Hormonal therapy 1
Gene therapy 1
Other 9

NOTE: Accrual by institution >5%. (a) University of Pittsburgh, 23.3%; (b) University of Wisconsin, 10.00%; (c) Mayo Clinic Rochester, 10.0%; (d) University of Pennsylvania, 8.3%; (e) Johns Hopkins University, 7.5%; (f) Beth Israel Deaconess Medical Center, 6.7%; (g) Emory University, 6.7%.

Table 2.

Immune response by peptide (n = 75)

Day gp100, % MART-1, % Tyrosine, %
Day 43 12.3 (8/65) 9.1 (6/66) 5.1 (3/59)
Day 85 28 (14/50) 5.6 (3/54) 10.6 (5/47)
Overall response to any of three peptides
Day 43 21.2 (14/66)
Day 85 37 (20/54)
Day 43 or 85 34.7 (26/75)
Day gp100, % MART-1, % Tyrosine, %
Day 43 12.3 (8/65) 9.1 (6/66) 5.1 (3/59)
Day 85 28 (14/50) 5.6 (3/54) 10.6 (5/47)
Overall response to any of three peptides
Day 43 21.2 (14/66)
Day 85 37 (20/54)
Day 43 or 85 34.7 (26/75)

Clinical antitumor responses. Of the 115 analyzable patients, 2 patients had a complete response (CR), 4 had a partial response (PR), 30 had stable disease, and 73 had progressive disease, whereas 6 patients were nonevaluable. Table 3A and B summarizes the baseline characteristics of the 6 patients with CR or PR and the correlation of clinical and immunologic response. There was no significant difference across treatment arms in terms of best overall response, defined as nonprogression (CR + PR + stable disease). As expected, only AJCC staging of disease on study enrollment was significantly correlated with best overall response [13 of 20 (65%) for M1a patients versus 12 of 47 (25.5%) for M1b patients versus 11 of 44 (25%) for M1c patients; P = 0.04].

(A) Baseline characteristics of six clinical responders (CR and PR)
Clinical response Immune response Diagnosis Stage Age Gender Metastatic Sites
1 CR Yes A M1a 47 M Soft tissue
2 CR No C M1a 50 F Distant node
3 PR Yes D M1c 68 F Lung, soft tissue, other
4 PR Yes A M1b 51 F Lung
5 PR Missing C M1c 74 M Distant node, other
6 PR Yes D M1b 65 F Lung
(B) Correlation of clinical response and immunologic response to peptide vaccination
Immune response CR PR Stable disease Progressive disease Unevaluable
No 1 1 15 29 2
Yes 1 2 9 13 0
(A) Baseline characteristics of six clinical responders (CR and PR)
Clinical response Immune response Diagnosis Stage Age Gender Metastatic Sites
1 CR Yes A M1a 47 M Soft tissue
2 CR No C M1a 50 F Distant node
3 PR Yes D M1c 68 F Lung, soft tissue, other
4 PR Yes A M1b 51 F Lung
5 PR Missing C M1c 74 M Distant node, other
6 PR Yes D M1b 65 F Lung
(B) Correlation of clinical response and immunologic response to peptide vaccination
Immune response CR PR Stable disease Progressive disease Unevaluable
No 1 1 15 29 2
Yes 1 2 9 13 0

Survival. The median follow-up for the 115 analyzable cases was 25.4 months (range, 19.7-45.3) to final analysis. For the entire study cohort, the median PFS was 2.8 months (95% confidence interval, 2.7-3.0) and the OS was 11.9 months (95% confidence interval, 9.5-14.6). Figure 2 (1 and 2) show the Kaplan-Meier plots for PFS and OS by treatment status, respectively. There was no significant difference in OS by IFN and/or GM-CSF treatment status (data not shown).

Fig. 2.

Fig. 2. PFS and OS by treatment (n = 115) and PFS by treatment (n = 115). 1, A, progression of survival of patients according to treatment with IFN-α2b; B, progression of survival of patients according to treatment with GM-CSF. OS by treatment (n = 115). 2, A, OS by treatment with IFN-α2b; B, OS by treatment with GM-CSF.

PFS and OS by treatment (n = 115) and PFS by treatment (n = 115). 1, A, progression of survival of patients according to treatment with IFN-α2b; B, progression of survival of patients according to treatment with GM-CSF. OS by treatment (n = 115). 2, A, OS by treatment with IFN-α2b; B, OS by treatment with GM-CSF.

Fig. 2.

Fig. 2. PFS and OS by treatment (n = 115) and PFS by treatment (n = 115). 1, A, progression of survival of patients according to treatment with IFN-α2b; B, progression of survival of patients according to treatment with GM-CSF. OS by treatment (n = 115). 2, A, OS by treatment with IFN-α2b; B, OS by treatment with GM-CSF.

PFS and OS by treatment (n = 115) and PFS by treatment (n = 115). 1, A, progression of survival of patients according to treatment with IFN-α2b; B, progression of survival of patients according to treatment with GM-CSF. OS by treatment (n = 115). 2, A, OS by treatment with IFN-α2b; B, OS by treatment with GM-CSF.

Close modal

Associations between immune and clinical data. The immune response data were associated with clinical data. Of the 75 patients with available ELISPOT data, 73 patients were clinically analyzable. Of these 73 patients, 25 patients had an immune response at any of the two time points studied (day 43 or 85) and to at least one of the three peptides administered.

There was a significant difference in OS by immune response status. Immune responders lived longer than the nonimmune responders (median OS, 21.3 versus 10.8 months; P = 0.033). The Cox model was used to evaluate the significance of immune response status for OS while adjusting for age, sex, performance status, AJCC stage at diagnosis, primary disease information (including Breslow depth, nodal involvement, and metastatic involvement), and ulceration status. Due to missing data on primary disease information, the total sample size for this analysis was smaller (n = 44). As expected, AJCC stage at diagnosis was the most significant predictor of OS (P = 0.002). Immune response status was marginally significant (P = 0.073). No other covariates were significant. The Kaplan-Meier plot for OS by immune response status is displayed in Fig. 3. No significant difference in PFS by immune response status was noted (data not shown). The data regarding immune response in relation to clinical response have been summarized in Table 3B.

Fig. 3.

Fig. 3. OS by ELISPOT response (n = 73). OS according to immune response to one or more of the three peptide vaccine antigens measured by ELISPOT.

OS by ELISPOT response (n = 73). OS according to immune response to one or more of the three peptide vaccine antigens measured by ELISPOT.

Fig. 3.

Fig. 3. OS by ELISPOT response (n = 73). OS according to immune response to one or more of the three peptide vaccine antigens measured by ELISPOT.

OS by ELISPOT response (n = 73). OS according to immune response to one or more of the three peptide vaccine antigens measured by ELISPOT.

Close modal

Toxicity.Table 4 summarizes treatment-related toxicities based on 117 evaluable patients. No treatment-related deaths occurred. Grade ≥3 treatment-related toxicity rates were 41.9%, 48.3%, 83.3%, and 48.7% for arms A to D, respectively. As expected, arm C had a significantly higher rate of toxicity with lymphopenia (9 of 30), neutropenia (7 of 30), and fatigue (11 of 30) being the most frequent.

Table 4.

Treatment-related toxicity (n = 117)

Toxicity type Arm A (n = 31) Arm B (n = 29) Arm C (n = 30) Arm D (n = 27)
Grade Grade Grade Grade
3 4 5 3 4 5 3 4 5 3 4 5
Hemoglobin 2
Leukocytes 1 3
Lymphopenia 2 5 9 7
Neutrophils 1 1 7 1
Transfusion: pRBCs 1 1
Supraventricular arrhythmias 1 1 1
Vasovagal episode 1
Cardiac-ischemia 1
Edema 1 1 1
Hypertension 1
Hypotension 2
Pericardial effusion/pericarditis 1
Thrombosis/embolism 1 1
Fatigue 2 10 1 4
Rigors/chills 1
Weight loss 1
DIC 1
PTT 1
PT 1
Injection site reaction 1 2
Rash/desquamation 1
Wound-infectious 1
Anorexia 2
Colitis 1
Constipation 1
Dehydration 1 1
Ileus 1 1
Nausea 1 1
Stomatitis 1
Vomiting 1 1 2
Diarrhea without prior colostomy 2 1
Central nervous system hemorrhage 1
SGPT 1
Infection with unknown ANC 1
Infection without neutropenia 1
Infection-other 1
Lymphatics 2
Hyperglycemia 1
Hypokalemia 1
Hyponatremia 1
Hypophosphatemia 1
Joint, muscle, bone-other 1
Confusion 1 1
Dizziness/lightheadedness 1 3
Hallucinations 1 1
Depression 1 2
Neuropathy-motor 1 1 1
Neuropathy-sensory 1
Syncope 3 1
Vertigo 1
Ocular-other 1
Abdominal pain 1
Arthralgia 1 1
Bone pain 1 1 3
Headache 2
Myalgia 1 1
Neuropathic pain 1 1
Pleuritic pain 1
Pain-other 2 1
Dyspnea 1 1 2 2 1
Hypoxia 1
Pneumonitis/pulmonary infiltrates 1
Pulmonary-other 1
Worst degree 8 5 11 3 22 3 13 3
Toxicity type Arm A (n = 31) Arm B (n = 29) Arm C (n = 30) Arm D (n = 27)
Grade Grade Grade Grade
3 4 5 3 4 5 3 4 5 3 4 5
Hemoglobin 2
Leukocytes 1 3
Lymphopenia 2 5 9 7
Neutrophils 1 1 7 1
Transfusion: pRBCs 1 1
Supraventricular arrhythmias 1 1 1
Vasovagal episode 1
Cardiac-ischemia 1
Edema 1 1 1
Hypertension 1
Hypotension 2
Pericardial effusion/pericarditis 1
Thrombosis/embolism 1 1
Fatigue 2 10 1 4
Rigors/chills 1
Weight loss 1
DIC 1
PTT 1
PT 1
Injection site reaction 1 2
Rash/desquamation 1
Wound-infectious 1
Anorexia 2
Colitis 1
Constipation 1
Dehydration 1 1
Ileus 1 1
Nausea 1 1
Stomatitis 1
Vomiting 1 1 2
Diarrhea without prior colostomy 2 1
Central nervous system hemorrhage 1
SGPT 1
Infection with unknown ANC 1
Infection without neutropenia 1
Infection-other 1
Lymphatics 2
Hyperglycemia 1
Hypokalemia 1
Hyponatremia 1
Hypophosphatemia 1
Joint, muscle, bone-other 1
Confusion 1 1
Dizziness/lightheadedness 1 3
Hallucinations 1 1
Depression 1 2
Neuropathy-motor 1 1 1
Neuropathy-sensory 1
Syncope 3 1
Vertigo 1
Ocular-other 1
Abdominal pain 1
Arthralgia 1 1
Bone pain 1 1 3
Headache 2
Myalgia 1 1
Neuropathic pain 1 1
Pleuritic pain 1
Pain-other 2 1
Dyspnea 1 1 2 2 1
Hypoxia 1
Pneumonitis/pulmonary infiltrates 1
Pulmonary-other 1
Worst degree 8 5 11 3 22 3 13 3

Discussion

This study is the first large, randomized, multi-institutional trial of multi-epitope peptide vaccination for patients who have failed prior therapy for metastatic unresectable melanoma. The study addresses laboratory as well as traditional clinical endpoints that were considered critical to future vaccine development. Previous large randomized multicenter melanoma vaccine trials either could not evaluate therapeutic effect because they were conducted in the adjuvant setting (3133) or they have not generally employed current immunologic monitoring against defined immunogenic peptides (34). We tested the immune response to lineage antigen-derived peptides administered either alone or in combination with two immunomodulatory agents whose efficacy has been or is being tested in the adjuvant clinical arena. We have shown the feasibility and safety of multicenter vaccine studies in conjunction with laboratory immunologic analyses for patients with metastatic melanoma.

The primary endpoints of this study were to define the immune response rate to the multi-epitope peptide vaccination and to determine the influence of immunomodulatory agents on peptide-specific immune response. The OIRR was lower compared with prior multi-epitope peptide vaccination studies in patients with unresectable metastatic melanoma (15, 16). This inferior immune response rate may be a laboratory artifact and could be attributable to the in vitro sensitization of lymphocytes to enhance detection of low-frequency melanoma antigen-specific precursor T cells used in other studies.

This study was large enough to have adequate statistical power to confirm observations from smaller vaccine studies previously reported in metastatic melanoma. First, the objective response rate (CR + PR) observed in this study was similar to reported results (20) suggesting limited epitope spreading, with insufficient duration and/or magnitude of immune responses induced to achieve tumor regression. Second, gp100209-217 induced higher rates of vaccination response than the other two lineage-specific peptides, and this high immunogenicity has been reported previously in other studies (35, 36). There are two possible explanations for this observation: a higher immunogenicity of the gp100209-217 peptide analogue has been noted and an increased abundance of gp100-positive melanoma cell lines has been described (37). However, no enhancement of immunogenicity was observed with the modified tyrosinase peptide analogue used in this study. Third, the survival of HLA-A2-positive patients enrolled into this trial was longer than historically anticipated (38, 39). HLA-A2-positive patients have been noted to have improved outcome in the setting of operable high-risk disease (40). The prolonged OS observed in this series of patients, all of whom were HLA-A2 positive, will be of interest to compare with patients other series of patients with metastatic melanoma according to MHC type (41).

This is the first study large enough to have adequate power to assess the immunomodulatory effects of cytokines on antigen-specific immune responses and antitumor responses of patients with refractory advanced metastatic melanoma. In this study, neither IFN-α nor GM-CSF significantly augmented the immunologic response to peptide vaccination of patients with active measurable metastatic melanoma. This may be explained by counter-regulatory mechanisms that may be triggered by the systemic administration of immunomodulatory cytokines. Alternatively, and given the effect of IFN-α and GM-CSF on tumor-infiltrating immune cells (5, 8), it may be that the most relevant assessment of tumor immunity and vaccination would have focused on tumor-infiltrating rather than peripheral blood antigen-specific lymphocyte responses.

The most promising finding that emerges from this study is that patients with immune response to at least one of the vaccine peptides lived longer than patients without detected immune responses, as has been noted previously in smaller vaccine studies (38), and is remarkable for several reasons. First, it suggests that therapies that augment host immune response may improve the outcome of metastatic melanoma. Second, this finding suggests that immune response to vaccination may be used as a marker of improved prognosis in patients with metastatic melanoma. Our results concord well with the recent results of a separate adjuvant study on the prognostic significance of autoimmunity in patients with resected high-risk melanoma who receive high-dose IFN and suggest that autoimmunity is an intermediate marker of benefit from high-dose IFN-α2b through the induction of a more specific immunity to lineage and other antigens of melanoma (9). Third, understanding the molecular mechanisms regulating the development of antigen-specific immune response to vaccination may lead to further improvement of treatment for metastatic melanoma.

In summary, this large multicenter randomized phase II study of vaccination with lineage peptide antigens of melanoma administered in conjunction with two different immunomodulatory cytokines has shown that large studies with complex intermediate immunologic endpoints are feasible in the cooperative groups using a central reference immunologic monitoring laboratory. Neither of the cytokines tested in this study was capable of significantly augmenting the immunologic or therapeutic response to vaccination. However, immune response to vaccination was associated with improved OS. This provides support for the pursuit of these same peptide vaccines in the adjuvant setting, which has been accomplished in the E4697 intergroup trial that enrolled 815 patients to rigorously test the benefit of vaccination versus placebo using the exact same peptides and schedule tested in E1696. A better understanding of the mechanisms that regulate immune responses to melanoma vaccines is necessary before larger phase III randomized vaccine trials are conducted.

Disclosure of Potential Conflicts of Interest

J.M. Kirkwood: commercial research grant, Schering Plough, Berlex; speaker's bureau, Schering Plough.

Grant support: NIH grants CA23318, CA66636, CA21115, and CA39229 and research awards to Eastern Cooperative Oncology Group from Schering Plough and Immunex.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

References

1

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.

2

Kirkwood JM, Manola J, Ibrahim J, et al. A pooled analysis of Eastern Cooperative Oncology Group and intergroup trials of adjuvant high-dose interferon for melanoma.

Clin Cancer Res

2004

;

10

:

1670

–7.

3

Spitler LE, Grossbard ML, Ernstoff MS, et al. Adjuvant therapy of stage III and IV malignant melanoma using granulocyte-macrophage colony-stimulating factor (GM-CSF).

J Clin Oncol

2000

;

18

:

1614

–21.

4

Lawson D, Kirkwood JM. Granulocyte-macrophage colony-stimulating factor: another cytokine with adjuvant therapeutic benefit in melanoma?

J Clin Oncol

2000

;

18

:

1603

–5.

5

Moschos SJ, Edington HD, Land SR, et al. Neoadjuvant treatment of regional stage IIIB melanoma with high-dose interferon α-2b induces tumor regression via an immunomodulatory mechanism.

J Clin Oncol

2006

;

24

:

3164

–71.

6

Panelli MC, Wang E, Phan G, et al. Gene-expression profiling of the response of peripheral blood mononuclear cells and melanoma metastases to systemic IL-2 administration.

Genome Biol

2002

;

3

:

0035.1

–0035.17.

7

Vuylsteke RJCLM, Molenkamp BG, Gietema HA, et al. Local administration of granulocyte/macrophage colony-stimulating factor increases the number and activation state of dendritic cells in the sentinel lymph node of early-stage melanoma.

Cancer Res

2004

;

64

:

8456

–60.

8

Soiffer R, Hodi FS, Haluska F, et al. Vaccination with irradiated, autologous melanoma cells engineered to secrete granulocyte-macrophage colony-stimulating factor by adenoviral-mediated gene transfer augments antitumor immunity in patients with metastatic melanoma.

J Clin Oncol

2003

;

21

:

3343

–50.

9

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.

10

Atkins M, Mier JW, Parkinson DR, et al. Hypothyroidism after treatment with interleukin-2 and lymphokine-activated killer cells.

N Engl J Med

1988

;

318

:

1557

–63.

11

Luiten RM, Kueter EWM, Mooi W, et al. Immunogenicity, including vitiligo, and feasibility of vaccination with autologous GM-CSF-transduced tumor cells in metastatic melanoma patients.

J Clin Oncol

2005

;

23

:

8978

–91.

12

Astsaturov I, Petrella T, Bagriacik EU, et al. Amplication of virus-induced antimelanoma T-cell reactivity by high-dose interferon-α2b: implications for cancer vaccines.

Clin Cancer Res

2003

;

9

:

4347

–55.

13

Chianese-Bullock KA, Pressley J, Garbee C, et al. MAGE-A1-, MAGE-A10-, and gp100-derived peptides are immunogenic when combined with granulocyte-macrophage colony stimulating factor and Montanide ISA-51 adjuvant and administered as part of a multipeptide vaccine for melanoma.

J Immunol

2005

;

174

:

3080

–6.

14

Zarour H, Kirkwood JM. Melanoma vaccines: early progress and future promises.

Semin Cutan Med Surg

2003

;

22

:

68

–75.

15

Slingluff CL, Petroni GR, Yamshchikov GV, et al. Immunologic and clinical outcomes of vaccination with a multiepitope melanoma peptide vaccine plus low-dose interleukin-2 administered either concurrently or on a delayed schedule.

J Clin Oncol

2004

;

22

:

4474

–85.

16

Slingluff CL, Jr., Petroni GR, Yamshchikov GV, et al. Clinical and immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocyte-macrophage colony-stimulating factor in adjuvant or pulsed on dendritic cells.

J Clin Oncol

2003

;

21

:

4016

–26.

17

Wang F, Bade E, Kuniyoski C, et al. Phase I trial of a MART-1 peptide vaccine with incomplete Freund's adjuvant for resected high-risk melanoma.

Clin Cancer Res

1999

;

5

:

2756

–65.

18

Rosenberg SA, Yang JC, Schwartzentruber DJ, et al. Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma.

Nat Med

1998

;

4

:

321

–7.

19

Davis ID, Chen W, Jackson H, et al. Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4+ and CD8+ T cell responses in humans.

Proc Nat Acad Sci U S A

2004

;

101

:

10697

–702.

20

Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines.

Nat Med

2004

;

10

:

909

–15.

21

Traversari C, van der Bruggen P, Leuscher IF, et al. A nonapeptide encoded by human gene MAGE-1 is recognized on HLA-A1 by cytolytic T lymphocytes directed against tumor antigen MZ2-E.

J Exp Med

1992

;

176

:

1453

–7.

22

Wolfel T, van Pel A, Brichard V, et al. Two tyrosinase nonapeptides recognized on HLA-A2 melanomas by autologous cytolytic T lymphocytes.

Eur J Immunol

1994

;

24

:

759

–64.

23

Cox AL, Skipper J, Chen Y, et al. Identification of a peptide recognized by five melanoma-specific human cytotoxic T cell lines.

Science

1994

;

264

:

716

–9.

24

van der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma.

Science

1991

;

254

:

1643

–7.

25

Kawakami Y, Eliyahu S, Delgado CH, et al. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection.

Proc Natl Acad Sci U S A

1994

;

91

:

6458

–62.

26

Coulie PG, Brichard V, van Pel A, et al. A new gene coding for a differentiation antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas.

J Exp Med

1994

;

180

:

35

–42.

27

Kawakami Y, Eliyahu S, Sakaguchi K, et al. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes.

J Exp Med

1994

;

180

:

347

–52.

28

Castelli C, Storkus WJ, Maeurer MJ, et al. Mass spectrometric identification of a naturally processed melanoma peptide recognized by CD8+ cytotoxic T lymphocytes.

J Exp Med

1995

;

181

:

363

–8.

29

Kirkwood JM, Bender C, Agarwala SS, et al. Mechanisms and management of toxicities associated with high-dose interferon α-2b therapy.

J Clin Oncol

2002

;

20

:

3703

–18.

30

Yamshchikov G, Barnd DL, Eastham S, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and peripheral blood of melanoma patients.

Int J Cancer

2001

;

92

:

703

–11.

31

Sondak VK, Liu P-Y, Tuthill RJ, et al. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: overall results of a randomized trial of the Southwest Oncology Group.

J Clin Oncol

2002

;

20

:

2058

–66.

32

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.

33

Hsueh EC, Morton DL. Antigen-based immunotherapy of melanoma: canvaxin therapeutic polyvalent cancer vaccine.

Semin Cancer Biol

2003

;

13

:

401

–7.

34

Kirkwood JM, Ibrahim J, Sosman JA, et al. High-dose interferon α-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.

35

Pass HA, Schwarz SL, Wunderlich JR, et al. Immunization of patients with melanoma peptide vaccines: immunologic assessment using the ELISPOT assay.

Cancer J Sci Am

1998

;

4

:

316

–23.

36

Markovic SN, Suman VJ, Ingle JN, et al. Peptide vaccination of patients with metastatic melanoma: improved clinical outcome in patients demonstrating effective immunization.

Am J Clin Oncol

2006

;

29

:

352

–60.

37

Lotem M, Zhao Y, Riley J, et al. Presentation of tumor antigens by dendritic cells genetically modified with viral and nonviral vectors.

J Immunother

2006

;

29

:

616

–27.

38

Kaufmann R, Spieth K, Leiter U, et al. Temozolomide in combination with interferon-α versus temozolomide alone in patients with advanced metastatic melanoma: a randomized, phase III, multicenter study from the Dermatologic Cooperative Oncology Group.

J Clin Oncol

2005

;

23

:

9001

–7.

39

Keiholz U, Punt CJA, Gore M, et al. Dacarbazine, cisplatin, and interferon-α-2b with or without interleukin-2 in metastatic melanoma: a randomized phase III trial (18951) of the European Organization for Research and Treatment of Cancer Melanoma Group.

J Clin Oncol

2005

;

23

:

6747

–55.

40

Kirkwood JM, Richards T, Zarour HM, et al. Immunomodulatory effects of high-dose and low-dose interferon α2b in patients with high-risk resected melanoma: the E2690 laboratory corollary of intergroup adjuvant trial E1690.

Cancer

2002

;

95

:

1101

–12.

41

Peterson AC, Gajewski TF. Immunization with Melan-A peptide-pulsed peripheral blood mononuclear cells plus recombinant human interleukin-12 induced clinical activity and T-cell responses in advanced melanoma.

J Clin Oncol

2003

;

21

:

2342

–8.

American Association for Cancer Research

2009