Purpose: This multicenter randomized trial was designed to evaluate whether melanoma helper peptides augment cytotoxic T lymphocyte (CTL) responses to a melanoma vaccine and improve clinical outcome in patients with advanced melanoma.

Experimental Design: One hundred seventy-five patients with measurable stage IV melanoma were enrolled into 4 treatment groups, vaccinated with 12 MHC class I-restricted melanoma peptides to stimulate CTL (12MP, group A), plus a tetanus peptide (group B), or a mixture of 6 melanoma helper peptides (6MHP, group C) to stimulate helper T lymphocytes (HTL), or with 6 melanoma helper peptide (6MHP) alone (group D), in incomplete Freund's adjuvant plus granulocyte macrophage colony-stimulating factor. CTL responses were assessed using an in vitro-stimulated IFN-γ ELIspot assay, and HTL responses were assessed using a proliferation assay.

Results: In groups A to D, respectively, CTL response rates to 12 melanoma peptides were 43%, 47%, 28%, and 5%, and HTL response rates to 6MHP were in 3%, 0%, 40%, and 41%. Best clinical response was partial response in 7 of 148 evaluable patients (4.7%) without significant difference among study arms. Median overall survival (OS) was 11.8 months. Immune response to 6 MHP was significantly associated with both clinical response (P = 0.036) and OS (P = 0.004).

Conclusion: Each vaccine regimen was immunogenic, but MHPs did not augment CTL responses to 12 melanoma peptides. The association of survival and immune response to 6MHP supports further investigation of helper peptide vaccines. For patients with advanced melanoma, multipeptide vaccines should be studied in combination with other potentially synergistic active therapies. Clin Cancer Res; 19(15); 4228–38. ©2013 AACR.

See related commentary by Woods and Cebon, p. 4021

Translational Relevance

This article reports results of a 4-arm randomized multicenter phase II clinical trial testing whether multipeptide melanoma vaccines that stimulate CD4+ helper T lymphocytes (HTL) would augment CD8+ cytotoxic T lymphocyte (CTL) responses for patients with advanced melanoma. Each vaccine regimen was immunogenic, but melanoma helper peptides (MHP) did not augment CTL responses to 12 melanoma peptides. However, there was a strong and significant association between survival and HTL responses to vaccines containing a mixture of 6MHP, whereas a similar association was not observed with HTL responses to a tetanus helper peptide. These data suggest that HTL responses to melanoma antigens have clinical relevance and deserve further investigation, but require consideration of different vaccine preparations or adjuvants. The low rates of objective clinical responses may be enhanced by combination with immune checkpoint blockade or other combination immune therapies.

Therapeutic cancer vaccines using defined tumor antigens show promise for melanoma and other cancers (1–5). Melanoma cells express peptides presented by MHC class I molecules, recognized by CD8+ cytotoxic T lymphocytes (CTL), or by MHC class II molecules, recognized by CD4+ helper T lymphocytes (HTL). Common melanoma peptides are from melanocytic differentiation proteins (MDP) and cancer-testis antigens (CTA; refs. 6–13). MDPs are expressed in 70% to 90% of metastatic melanomas and may be downregulated with progression (14–17); several CTAs are expressed in 30% to 60% of melanomas, and their expression may increase in more advanced melanomas (18, 19). Thus, there is rationale for vaccines targeting both MDPs and CTAs (20).

Most peptide vaccines have targeted only CTL; however, natural immune responses to pathogens induce both HTLs and CTLs. The present trial tests vaccines targeting both. A mixture of 12 melanoma peptides (12MP) restricted by HLA-A1, A2, or A3 has induced CTLs in up to 70% to 100% of patients (20–22). A mixture of 6 melanoma helper peptides (6MHP) has been immunogenic in 80% of patients and associated with clinical activity (22, 23), and a tetanus helper peptide has been immunogenic and safe. We proposed to test, in advanced melanoma, whether the immunogenicity and immune protection of a 12MP melanoma vaccine would be augmented by addition of melanoma-specific helper epitopes (6MHP).

Patients

Eligible patients had histologically proven American Joint Committee on Cancer Stage IV melanoma with measurable disease, HLA-A1, A2, or A3 expression, age 18 years or more, Eastern Cooperative Oncology Group (ECOG) performance status 0 and 1, adequate organ function, and lactate dehydrogenase (LDH) ≤2 times the upper limits of normal (ULN). Exclusions included: pregnancy; other therapy within 4 weeks; allergy to vaccine components; other cancers within 5 years, untreated or progressive brain metastases; steroids; immunosuppression; or severe autoimmune disease. Patients provided informed consent, and the study was approved by the institutional review boards and the US Food and Drug Administration and registered at ClinicalTrials.gov (NCT00071981).

Vaccines

Patients received vaccines composed of multiple peptides from MDP and CTA, including 12MP (20; Arms A–C) restricted by HLA-A1, A2, or A3, to stimulate CTLs, 6MHP (23; Arms C and D), or a tetanus helper peptide (tet; 24, Arm B) to stimulate HTLs. Peptide sequences and details of Good Manufacturing Practice preparation and compliance (25) are provided in Supplementary Text.

Each vaccine was administered as a 2 mL stable water-in-oil emulsion, prepared by the two-syringe method, containing 100 mcg (12MP) and/or 200 mcg (6MHP, tet) of each peptide, plus 110 mcg granulocyte macrophage colony-stimulating factor (GM-CSF; Berlex/Genzyme) plus 1 mL Montanide ISA-51 or ISA-51 VG (Seppic, Inc.). On days 1, 8, and 15, vaccines were administered (half-intradermally and half-subcutaneously) at 2 injection sites (primary and replicate sites), on extremities uninvolved with tumor. On day 22, an optional biopsy was conducted of a lymph node draining the replicate vaccine site [sentinel immunized node, SIN (26)]. On days 29, 36, and 43, one vaccine was administered at the primary vaccine site only. Patients were reassessed at weeks 8 and 12 for evidence and confirmation of clinical response via Response Evaluation Criteria in Solid Tumors (RECIST) v1.0. Patients without progression were eligible for 6 cycles of 3 weekly booster vaccines, until progression or for 2 years.

Trial design

This was an open-label, multicenter phase II study, with randomization equal to vaccination with 12MP, 12MP/Tet, 12MP/6MHP, or 6MHP (Supplementary Fig. S1), through the ECOG web-registration program, using permuted blocks within strata with dynamic balancing within main institutions and affiliate networks. Patients were stratified by HLA status (HLA-A1+ only, HLA-A2+ only, HLA-A1+ and HLA-A2+, other) and sentinel immunized node (SIN) biopsy to be conducted (yes, no).

The primary objective was to determine the CTL response to 12MP. Secondary objectives were: (i) to determine the HTL response to 6 MHP; (ii) to estimate whether addition of 6MHP to 12MP augments CTL responses to 12MP; (iii) to obtain preliminary data on whether booster vaccination may maintain immune responses, (iv) to estimate rates of clinical response and survival, and (v) to obtain preliminary data on whether cellular immune responses may correlate with clinical outcome.

The primary endpoint was the CTL response, with the main comparisons of CTL response rates in arms A, B, and C versus D. We expected CTL response rates less than 10% in arm D and at least 40% in arms A to C. The sample size of 40 evaluable patients in each arm provided at least 85% power to detect a difference of at least 30% in CTL response rate, based on a 2-sided type I error of 0.05 using Fisher exact test. Secondary endpoints included HTL responses in peripheral blood mononuclear cells (PBMC), clinical response, and outcome. We expected HTL responses to be higher in arms B, C, and D (at least 30%) than A (5%), with the main comparisons between HTL immune response rates in arms B, C, and D versus A. There would be at least 82% power to detect a difference of at least 25% using Fisher exact test with a 2-sided type I error of 0.05.

Clinical endpoints

Tumor response was evaluated per RECIST v1.0 at weeks 8 and 12, then every 12 weeks, with survival follow-up thereafter. Objective response rate (ORR) was defined as complete response (CR) + partial response (PR) rates. Disease control rate (DCR) was defined as CR+PR+SD (stable disease at 8 weeks). Overall survival (OS) was defined as the time from randomization to death from any cause. Patients still alive were censored at the date last known alive. Progression-free survival (PFS) was defined as time from randomization to disease progression or death, censoring cases without progression at the date of last disease assessment.

Correlative studies

Peripheral blood (100 mL) was collected pretreatment, at weeks 1, 3, 5, 7, and 11, and at the first and third of each set of booster vaccines. PBMCs were isolated from 90 mL of blood and viably cryopreserved, and serum was collected from 10 mL blood. Lymphocytes were isolated from the SIN (26). CTL response was determined by IFN-γ ELIspot assay conducted 14 days after 1 in vitro sensitization, as described (in vitro-stimulated ELIspot assay; ref. 20). CTL response required at least a 2-fold increase over background and 30 responding cells per 100,000 (details in Supplementary Text) HTL response was evaluated in a 5-day tritiated thymidine proliferation assay, as described in ref. (23) and in Supplementary Text, and required at least a 4-fold increase in proliferation. Cytokine [interleukin (IL)-12 p70, IL-1b, IL-2, IL-5, IL-6, IL-10, IL-17, TNFα, IFN-γ, GM-CSF], and chemokine (CCL3, CCL4, CCL5, CXCL9, CXCL10) production was measured by Luminex in supernatants collected from PBMC after 2-day or 5-day culture with helper peptides or mitogens [phytohaemagglutinin (PHA) and phorbol myristate acetate (PMA)]. Treatment samples (week 3–7) were compared with pretreatment samples with paired Student t tests.

Toxicity assessment and stopping rules

The trial was monitored continuously by the ECOG Data Monitoring Committee for treatment-related adverse events, using NCI Common Terminology Criteria for Adverse Events version 3.0. Protocol treatment was to be discontinued for unexpected treatment-related grade 4 hematologic, grade 3 nonhematologic, grade 2 allergic, or grade 1 ocular events, or for disease progression requiring other therapy. If 3 of the first 9 patients in any arm experienced dose-limiting toxicities (DLT), we would consider closing that arm. If the true DLT rate was 5%, the probability of observing 3 patients or more experiencing DLT events of 9 patients was 0.008. Interim analyses were conducted for immune response and clinical outcome, as detailed in Supplementary Text.

Statistical analysis

Immune response rate, objective clinical response rate, DCR, and toxicity incidence were compared between treatment arms using the Fisher exact test (27). Distributions of OS and PFS were estimated using the Kaplan–Meier method (28), with 95% confidence intervals (CI) calculated using Greenwood formula. Treatment effect on survival was tested using log-rank tests. Associations between immune response and clinical response were explored using Fisher exact test and logistic regression. Logistic regression models for rare events were also fit for testing association between immune response and clinical response as sensitivity analysis (29). Multivariable Cox proportional-hazards models (30) were built to explore associations between immune response and OS using the landmark method (31), with 2 months from randomization as a landmark time point. In the landmark analysis, OS was defined from landmark to the event (death); patients with OS events before the landmark were excluded from that analysis. All reported P values were for 2-sided tests. Significance level was set at 0.05. Given the exploratory nature of this study, no adjustments were made for multiple comparisons.

Patients

One hundred seventy-five participants were enrolled from March 2005 to January 2009 (Table 2, Consort Diagram Fig. 1). At interim analysis, Arm C met protocol criteria for stopping based on clinical outcome, after accruing 37 patients. On eligibility review, 21 patients were ineligible (19 treated). Reasons for ineligibility are provided in Supplementary Text. Among the 154 eligible participants, 6 never started assigned therapy; thus, 148 eligible and treated patients were used for clinical outcome analysis. Toxicity analysis was based on the 167 participants who received protocol therapy and had toxicity reported, regardless of eligibility. Immune response analyses were based on 140 eligible participants with CTL data, and 128 with HTL data. Forty-five patients (30%) received booster vaccines (16, 8, 7, and 14, on arms A–D, respectively); 7 completed all 6 cycles. Study arms were well matched for known prognostic factors (Supplementary Table S1), but prior immunotherapy was less frequent for arm B than others (P = 0.01). Overall, serum LDH was elevated in 28%; 80% had 2 or more metastatic sites; M-stage was 7% M1a, 22% M1b, and 72% M1c.

Figure 1.

CONSORT diagram for E1602 trial.

Figure 1.

CONSORT diagram for E1602 trial.

Close modal

Immune response

CTL response rate.

CTL response rates to 12 melanoma peptides for Arms A to D were 43%, 47%, 28%, and 5%, respectively (Fig. 2A, Supplementary Table S2), and differed among treatment arms (P < 0.001) and for arms A to C versus D (P < 0.05), but not for A versus C or B versus C. In multivariable logistic analysis, the odds of CTL response was lower for patients on arm D, patients with more metastases, and increased age (Table 1). Responses to each peptide and responses to booster vaccines are summarized in Supplementary Text and Supplementary Table S3.

Figure 2.

Immune responses to 12MP and 6MHP. A, the proportions of analyzable patients with CTL response to 12MP (blue bars), HTL response to tetanus peptide (yellow bars), and HTL response to 6MHP (maroon bars) are shown for each study arm. These are based on 140 eligible participants with CTL data (40, 30, 29, and 41 on arms A–D, respectively), and 128 eligible participants had HTL response data (40, 30, 29, and 41 on arms A–D, respectively). B, the objective clinical response rate is shown for patients based on the presence or absence of CTL or HTL response to peptides in the vaccines (n = 140 for response to 12MP, and n = 128 for response to 6 MHP or tetanus peptides; patients in each category are listed under the x-axis); C, median survival and 1-year survival based on 2-month Landmark analyses, by CTL response to 12MP, HTL response to 6MHP or tetanus, and considering just patients vaccinated with the respective peptides.

Figure 2.

Immune responses to 12MP and 6MHP. A, the proportions of analyzable patients with CTL response to 12MP (blue bars), HTL response to tetanus peptide (yellow bars), and HTL response to 6MHP (maroon bars) are shown for each study arm. These are based on 140 eligible participants with CTL data (40, 30, 29, and 41 on arms A–D, respectively), and 128 eligible participants had HTL response data (40, 30, 29, and 41 on arms A–D, respectively). B, the objective clinical response rate is shown for patients based on the presence or absence of CTL or HTL response to peptides in the vaccines (n = 140 for response to 12MP, and n = 128 for response to 6 MHP or tetanus peptides; patients in each category are listed under the x-axis); C, median survival and 1-year survival based on 2-month Landmark analyses, by CTL response to 12MP, HTL response to 6MHP or tetanus, and considering just patients vaccinated with the respective peptides.

Close modal
Table 1.

Multivariable logistic regression for CTL response to 12MP (N = 140)

Baseline CharacteristicsLevelsORP-value
Treatment Arm A vs. Arm D 11.9 0.009 
 Arm B vs. Arm D 33.1 <0.001 
 Arm C vs. Arm D 30.2 <0.001 
Age >65 vs. < = 65 0.2 0.015 
Sex Female vs. Male 1.3 0.657 
ECOG performance status 1 vs. 0 0.6 0.289 
Number of sites 2–3 vs. 1 0.9 0.856 
 > = 4 vs. 1 0.1 0.005 
Ulceration Yes vs. No 0.8 0.782 
 Unknown vs. No 0.6 0.469 
Clark level IV vs. V 0.6 0.486 
 Other vs. V 0.4 0.432 
 Unknown vs. V 1.3 0.736 
Serum LDH Elevated vs. Normal 2.4 0.160 
Prior chemotherapy Yes vs. No 0.4 0.115 
Prior radiotherapy Yes vs. No 0.3 0.063 
Prior surgery Yes vs. No 0.6 0.288 
Chronic disease pre-registration Yes vs. No 2.9 0.086 
Baseline CharacteristicsLevelsORP-value
Treatment Arm A vs. Arm D 11.9 0.009 
 Arm B vs. Arm D 33.1 <0.001 
 Arm C vs. Arm D 30.2 <0.001 
Age >65 vs. < = 65 0.2 0.015 
Sex Female vs. Male 1.3 0.657 
ECOG performance status 1 vs. 0 0.6 0.289 
Number of sites 2–3 vs. 1 0.9 0.856 
 > = 4 vs. 1 0.1 0.005 
Ulceration Yes vs. No 0.8 0.782 
 Unknown vs. No 0.6 0.469 
Clark level IV vs. V 0.6 0.486 
 Other vs. V 0.4 0.432 
 Unknown vs. V 1.3 0.736 
Serum LDH Elevated vs. Normal 2.4 0.160 
Prior chemotherapy Yes vs. No 0.4 0.115 
Prior radiotherapy Yes vs. No 0.3 0.063 
Prior surgery Yes vs. No 0.6 0.288 
Chronic disease pre-registration Yes vs. No 2.9 0.086 

NOTE: Bold highlights P < 0.05.

Abbreviations: ECOG, Eastern Cooperative Oncology Group; LDH, serum lactate dehydrogenase.

HTL responses.

HTL response rates to 6 MHP for Arms A to D, respectively, were 3%, 0%, 40%, and 41% (P < 0.001); for tetanus peptide, they were 11%, 59%, 4%, and 0%, respectively (P < 0.001, Fig. 2A, Supplementary Table S2).

Cytokine and chemokine production.

Antigen-specific cytokine and chemokine production by PBMC was evaluated in the 47 analyzable patients with HTL responses to 6 MHP (26), tet (20), or both (1). Among arm B patients, there were treatment-related increases in tetanus-specific production of IL-2 (d2, P = 0.0383), IL-5 (d2, P = 0.0149; d5, P = 0.0088), IFN-γ (d2, P = 0.006; d5, P = 0.0118), GM-CSF (d5, P = 0.0377), CXCL9 (d2, P = 0.0144; d5, P = 0.0280), and CXCL10 (d2, P = 0.0064; d5, P = 0.0055). Among 29 arm C+D patients, there were significant treatment-related increases in 6MHP–specific production of IL-2 (d2, P = 0.021), IL-5 (d2, P = 0.0212; d5, P = 0.0072), IL-12p70 (d5, P = 0.0359), CXCL9 (d2, P = 0.0176; d5, P = 0.0434), and CXCL10 (d5, P = 0.0076). These data for IL-2, IL-5, CXCL9, and CXCL10 are shown in Fig. 3. Within groups C+D, for patients with partial response or SD (n = 9), after stimulation with 6MHP, there were treatment-related increases in CCL3 (d2, P = 0.0212) and CCL4 (d2, 0.0125); for patients with PD (n = 19), there were increases in IL-2 (d2, P = 0.0413), IL-5 (d2, P = 0.0292; d5, P = 0.0096), GM-CSF (d5, P = 0.0238), and CXCL10 (d5, P = 0.0151, data not shown).

Figure 3.

Cytokine/chemokine produced by HTLs. From 47 patients with proliferative responses to tetanus peptide or 6MHP, 2-day (A, B) and 5-day (C, D) supernatants of PBMC from patients on arm B (A, C) or arms C+D (B, D) stimulated in vitro with 6MHP (6 MHP-0, 6MHP +) or tetanus peptide (Tet-0 or Tet +) were assayed by Luminex for 10 cytokines and 5 chemokines, with increases in IL-2, IL-5, CXCL9, and CXCL10, shown here. Each line represents data from PBMC taken pretreatment (6 MHP-0 or Tet-0) or after 3 to 6 vaccines (6MHP + or Tet +). The median concentrations induced by PHA for IL-2, IL-5, CXCL9, and CXCL10 were 4,600, 198, 2,248, and 760 pg/mL, respectively.

Figure 3.

Cytokine/chemokine produced by HTLs. From 47 patients with proliferative responses to tetanus peptide or 6MHP, 2-day (A, B) and 5-day (C, D) supernatants of PBMC from patients on arm B (A, C) or arms C+D (B, D) stimulated in vitro with 6MHP (6 MHP-0, 6MHP +) or tetanus peptide (Tet-0 or Tet +) were assayed by Luminex for 10 cytokines and 5 chemokines, with increases in IL-2, IL-5, CXCL9, and CXCL10, shown here. Each line represents data from PBMC taken pretreatment (6 MHP-0 or Tet-0) or after 3 to 6 vaccines (6MHP + or Tet +). The median concentrations induced by PHA for IL-2, IL-5, CXCL9, and CXCL10 were 4,600, 198, 2,248, and 760 pg/mL, respectively.

Close modal

Clinical outcome

Seven patients had PR (4.7%, 95% CI: 1.9–9.5), and 27% had SD at 8 weeks (Table 2). There were no CRs. ORRs (CR+PR, ORR) did not differ significantly across study arms (P = 0.74). The DCR (CR+PR+SD, DCR) was 32% (47/148; 95% CI: 24–40) overall. For arms A to D, PR rates were 2, 3, 6, and 7%, respectively. DCRs were 46%, 15%, 25%, and 36%, respectively, differing among arms (P = 0.026), with A > B (P = 0.006). SD persisted to 6 months in 5, 2, 3, and 2 patients on arms A to D, respectively; thus, PR or durable SD were observed in 15%, 9%, 16%, and 12%, respectively.

Table 2.

Clinical response by treatment arm

Best overall responseArm A n (%)Arm B n (%)Arm C n (%)Arm D n (%)Total n (%)P-value
Analyzable (eligible and treated) patients n = 41 n = 33 n = 32 n = 42 n = 148  
CR  
PR 1 (2.4) 1 (3.0) 2 (6.3) 3 (7.1) 7 (4.7)  
SD (at 8 weeks) 18 (43.9) 4 (12.1) 6 (18.8) 12 (28.6) 40 (27.0)  
PD 22 (53.7) 27 (81.8) 23 (71.9) 27 (64.3) 99 (66.9)  
Un-evaluable 1 (3.0) 1 (3.1) 2 (1.4)  
ORR (CR+PR) 1 (2.4) 1 (3.0) 2 (6.3) 3 (7.1) 7 (4.7) 0.741 
DCR (CR+PR+SD) 19 (46.3) 5 (15.1) 8 (25.1) 15 (35.7) 47 (31.7) 0.026 
All patients n = 47 n = 43 n = 37 n = 48 n = 175  
CR  
PR 1 (2.1) 1 (2.3) 2 (5.4) 3 (6.2) 7 (4.0)  
SD (at 8 weeks) 19 (40.4) 4 (9.3) 7 (18.9) 13 (27.1) 43 (24.6)  
PD 24 (51.1) 31 (72.1) 26 (70.3) 29 (60.4) 110 (62.9)  
Un-evaluable 3 (6.4) 7 (16.3) 2 (5.4) 3 (6.2) 15 (8.6)  
ORR (CR+PR) 1 (2.1) 1 (2.3) 2 (5.4) 3 (6.2) 7 (4.0) 0.662 
DCR (CR+PR+SD) 20 (42.5) 5 (11.6) 9 (24.3) 16 (33.3) 50 (28.6) 0.008 
Best overall responseArm A n (%)Arm B n (%)Arm C n (%)Arm D n (%)Total n (%)P-value
Analyzable (eligible and treated) patients n = 41 n = 33 n = 32 n = 42 n = 148  
CR  
PR 1 (2.4) 1 (3.0) 2 (6.3) 3 (7.1) 7 (4.7)  
SD (at 8 weeks) 18 (43.9) 4 (12.1) 6 (18.8) 12 (28.6) 40 (27.0)  
PD 22 (53.7) 27 (81.8) 23 (71.9) 27 (64.3) 99 (66.9)  
Un-evaluable 1 (3.0) 1 (3.1) 2 (1.4)  
ORR (CR+PR) 1 (2.4) 1 (3.0) 2 (6.3) 3 (7.1) 7 (4.7) 0.741 
DCR (CR+PR+SD) 19 (46.3) 5 (15.1) 8 (25.1) 15 (35.7) 47 (31.7) 0.026 
All patients n = 47 n = 43 n = 37 n = 48 n = 175  
CR  
PR 1 (2.1) 1 (2.3) 2 (5.4) 3 (6.2) 7 (4.0)  
SD (at 8 weeks) 19 (40.4) 4 (9.3) 7 (18.9) 13 (27.1) 43 (24.6)  
PD 24 (51.1) 31 (72.1) 26 (70.3) 29 (60.4) 110 (62.9)  
Un-evaluable 3 (6.4) 7 (16.3) 2 (5.4) 3 (6.2) 15 (8.6)  
ORR (CR+PR) 1 (2.1) 1 (2.3) 2 (5.4) 3 (6.2) 7 (4.0) 0.662 
DCR (CR+PR+SD) 20 (42.5) 5 (11.6) 9 (24.3) 16 (33.3) 50 (28.6) 0.008 

NOTE: P-values were from Fisher's exact test. Among analyzable patients, SD at 3 months was observed in 10, 4, 5, and 5 patients on arms A to D, respectively; SD at 6 months was observed in 5, 2, 3, and 2 patients on arms A to D, respectively.

Abbreviations: PD, progressive disease; ORR, objective response rate; DCR, disease control rate.

Of the 148 analyzable patients, median OS was 11.8 (95% CI: 10.2–14.0) months. For arms A to D, median OS and 95% CI were 14.9 (10.1–18.6), 10.2 (6.7–12.2), 12.4 (4.8–16.8), and 11.1 (8.8–14.2) months, respectively. The respective 1-year OS probabilities were 59% (42–72), 38% (21–54), 53% (35–69), and 49% (33–63) [50% (42–58) overall], and were assessed in the context of prior outcomes in multicenter cooperative group trials (32): correcting for cited variables (gender, visceral disease, performance status), the observed 1-year OS rate exceeds prediction by at least 2-fold, for each study arm (Supplementary Tables S4 and S5) and the observed values fall above the 95th percentile curves for arms A, C, D, and overall (Fig. 4D). PFS was not a designated endpoint of the study and did not differ significantly among study arms (Supplementary Text).

Figure 4.

Survival outcomes. Kaplan–Meier estimates of OS, for analyzable patients (n = 148) by study arm treatment (A, n = 148), by CTL immune response (B, n = 139, landmark method), and by HTL immune response to 6 MHP in arms C and D (C, n = 64, landmark method). D, one-year survival for each treatment arm and for the full study population is shown on the plot of 1-year OS for all melanoma trials.

Figure 4.

Survival outcomes. Kaplan–Meier estimates of OS, for analyzable patients (n = 148) by study arm treatment (A, n = 148), by CTL immune response (B, n = 139, landmark method), and by HTL immune response to 6 MHP in arms C and D (C, n = 64, landmark method). D, one-year survival for each treatment arm and for the full study population is shown on the plot of 1-year OS for all melanoma trials.

Close modal

Association between immune response and clinical outcomes

For eligible and analyzable patients with and without CTL responses to 12 MP, ORRs were 9.8% and 3.0% (P = 0.194), and DCRs were 37% and 32% (P = 0.695), respectively (Fig. 2B). Among patients vaccinated with 12MP (groups A–C), ORRs were 10.3% and 0% (P = 0.022) and DCRs 36% and 30% (P = 0.661), respectively. Landmark analysis showed no significant association between OS and CTL response (P = 0.253, Fig. 4B). In multivariable regression analyses for groups A to D, CTL response was not associated with clinical response (OR = 5.9; 95% CI: 0.7–48.6; P = 0.102) or OS (HR 0.87; P = 0.557; Table 3, Model 1). The landmark analysis data are comparable if eligible and ineligible patients were assessed (P = 0.312 for 12 MP; data not shown)

Table 3.

Multivariable Cox landmark regression analysis for OS by immune response and clinical variables

Model 1: OS by CTL response to 12MP, n = 139, Arms A–DModel 2: OS by helper T-cell response to 6MHP, n = 64, Arms C, D
VariablesLevelHRP valueHRP-value
CTL response Yes vs. No 0.87 0.557 — — 
Helper T-cell response to 6 MHP Yes vs. No — — 0.50 0.038 
Treatment Arm B vs. Arm A 1.57 0.124 — — 
 Arm C vs. Arm A 1.05 0.863 — — 
 Arm D vs. Arm A 1.42 0.198 — — 
 Arm D vs. Arm C — — 1.36 0.305 
Age Years (continuous) 1.02 0.041 1.00 0.830 
Sex Female vs. Male 0.92 0.684 0.62 0.164 
ECOG performance status 1 vs. 0 0.95 0.810 0.63 0.174 
Number of sites 2–3 vs. 1 1.74 0.033 2.32 0.037 
 ≥4 vs. 1 2.66 0.003 2.55 0.046 
Ulceration Yes vs. No 0.81 0.390 0.75 0.525 
 Unknown vs. No 0.75 0.310 0.58 0.209 
Clark level IV vs. V 0.75 0.335 1.07 0.877 
 Other vs. V 0.43 0.027 0.28 0.036 
 Unknown vs. V 1.06 0.859 1.04 0.932 
Serum LDH Elevated vs. normal 1.45 0.107 1.45 0.334 
Model 1: OS by CTL response to 12MP, n = 139, Arms A–DModel 2: OS by helper T-cell response to 6MHP, n = 64, Arms C, D
VariablesLevelHRP valueHRP-value
CTL response Yes vs. No 0.87 0.557 — — 
Helper T-cell response to 6 MHP Yes vs. No — — 0.50 0.038 
Treatment Arm B vs. Arm A 1.57 0.124 — — 
 Arm C vs. Arm A 1.05 0.863 — — 
 Arm D vs. Arm A 1.42 0.198 — — 
 Arm D vs. Arm C — — 1.36 0.305 
Age Years (continuous) 1.02 0.041 1.00 0.830 
Sex Female vs. Male 0.92 0.684 0.62 0.164 
ECOG performance status 1 vs. 0 0.95 0.810 0.63 0.174 
Number of sites 2–3 vs. 1 1.74 0.033 2.32 0.037 
 ≥4 vs. 1 2.66 0.003 2.55 0.046 
Ulceration Yes vs. No 0.81 0.390 0.75 0.525 
 Unknown vs. No 0.75 0.310 0.58 0.209 
Clark level IV vs. V 0.75 0.335 1.07 0.877 
 Other vs. V 0.43 0.027 0.28 0.036 
 Unknown vs. V 1.06 0.859 1.04 0.932 
Serum LDH Elevated vs. normal 1.45 0.107 1.45 0.334 

NOTE: Two models for OS are summarized in the table: Model 1 includes all patients and includes the measures of CTL response; Model 2 includes patients vaccinated with 6MHP (arms C and D) and includes measures of the immune response to 6MHP.

One patient died within 2 months and was not included in the landmark analysis for OS. Bold highlights P < 0.05.

Among 128 analyzable patients with or without HTL response to 6MHP, ORRs were 15% (4/27) and 3% (3/101; P = 0.036), respectively (Fig. 2B). Evaluating only the 64 patients on arms C and D, ORRs were similar: 15.4% and 2.6%, respectively (P = 0.149 due to the smaller sample size). In contrast, response to tetanus peptide was not associated with clinical response (P = 1.00). Landmark analyses showed that OS, for 64 patients on arms C and D, was strongly and significantly associated with HTL response to 6MHP (P = 0.0045), with 1-year survival rate of 65% and 24%, and median OS of 14.7 and 7.9 months, respectively, for patients with or without HTL response (Figs. 2C and 4C). This association remained significant in multivariable Cox landmark analysis for arms C+D (HR 0.50; 95% CI: 0.26–0.96; P = 0.038; Table 3, Model 2). In contrast, of 27 analyzable patients on arm B, landmark analysis showed no significant association of survival with response to tetanus peptide (P = 0.153, Fig. 2C). The landmark analysis data are comparable if eligible and ineligible patients were assessed (P = 0.0183 for 6MHP; data not shown); and the effect is in the same direction when groups C and D are assessed individually, though not significant for arm D given the smaller sample size (Arm C: P = 0.009, Arm D: P = 0.127, Supplementary Fig. S2).

Of the 128 analyzable patients with data on both CTL response and HTL response, patients without response to 12MP or 6MHP had an ORR of 1.5%; corresponding rates were 6.9% with response to either, and 50% for response to both (P = 0.005, Fig. 2B).

Toxicity

Treatment-related toxicities are summarized in Supplementary Table S6. Rates of grade 3 or higher toxicity were 13%, 24%, 19%, and 8% on arms A to D, respectively. There was no significant difference in toxicity rates among treatment arms (P > 0.05). There were 6 grade 4 toxicities in 4 patients (neutrophils, troponin elevation, lung infection, lymphopenia, CNS ischemia, and AST elevation) and no treatment-related mortality. The most common toxicities of grade 3 or higher were fatigue and injection site reaction.

The cytotoxic T-lymphocyte response rates in arms A and B met primary study objectives by exceeding 40%; however, the study failed to show increased immunogenicity of a 12 MP melanoma vaccine by addition of melanoma-specific helper epitopes (6MHP), as only 28% had cytotoxic T-lymphocyte responses in arm C. This negative result is consistent with a parallel study conducted in the adjuvant setting (22). Possible explanations may include expansion of antigen-specific regulatory T cells, suboptimal cytokine induction, homing of T cells to tumor deposits, or sequestration of T cells at vaccine sites (33). Prior experience with 6 MHP vaccines argues against a TH2-dominant cytokine response (Dillon and colleagues, manuscript in preparation) and against expansion of total regulatory T cells (23). However, effects on melanoma-specific regulatory T cells and T-cell trafficking warrant further study.

Data from the HIV literature show that induction of HTLs can restore function in CTLs (34). However, combining epitopes for HTLs and CTLs into one colinear peptide may be preferable to using them separately in vaccines (35–37). Short peptides (9–11 residues) can bind directly to class I MHC molecules of cells in the vaccine-site microenvironment (VSME; ref. 38). On the other hand, longer peptides require uptake and intracellular processing before presentation on nascent MHC molecules in professional APC, which migrate from the VSME to draining lymph nodes after vaccination (38). It is possible that the intermediate length helper peptides in 6MHP (14–23 amino acids) may be presented primarily by professional APC in the vaccine-draining lymph node (VDLN). If peptides for CTLs and HTLs are presented in different compartments (VSME and VDLN), intended synergy may not be achieved.

Despite a body of literature that supports the use of GM-CSF as a vaccine adjuvant (4, 39, 40), 2 prospective randomized phase II trials have shown negative effects of GM-CSF in combination with other vaccine adjuvants (21, 41). The overall CTL response rates (43 and 47% in arms A and B) are based on ELIspot assays conducted after in vitro stimulation; which has been required for other studies when GM-CSF was used in the adjuvant (4, 20, 21); we expect that the magnitude of these responses would be greater using incomplete Freund's adjuvant without GM-CSF. It is also notable that the CTL response rates of 43% to 47% fall short of the circulating CTL response rate of 83% to the same 12 class I MHC-restricted peptides in a prior study (20); this may be due to effects of shipping blood from many centers or to cryopreservation methods (manuscript in preparation), or to the more advanced stage of the patients in the present study. However, this CTL response rate is at least as high as observed with a prior ECOG multipeptide vaccine trial (42).

Although the MHPs did not increase circulating CTL responses in this study, the HTL response rates to 6MHP in arm C (40%) and D (41%) were similar and were comparable with prior studies (22, 23). Thus, HTL responses to 6 MHP are not impacted negatively by addition of 12 melanoma peptides. The response to tetanus peptide and to 6MHP includes specific induction of low levels of TH1-associated cytokine IL-2, but also significant production of the TH2 cytokine IL-5. In prior studies, HTL responses to tetanus peptide were primarily TH1 dominant (24), as were responses to 6MHP (Dillon P and colleagues, manuscript in preparation). Regardless, it is anticipated that improved vaccine adjuvants are needed to induce T cells with stronger TH1 bias. Also, TH1-associated chemokines, CXCL9–10, implicated in T-cell homing to tumor (43, 44), were produced (Fig. 3); so if these vaccine-induced CD4+ T cells infiltrate melanoma metastases, they may be helpful in recruiting melanoma-reactive CD8+ T cells to the tumor. Notably, there was a strong association between immune response to the 6MHP and clinical response and patient survival, unlike tetanus peptide. These findings suggest a more important clinical role of HTL responses to melanoma antigens than previously appreciated.

The overall OR rate of 4.7% is consistent with prior experience with peptide vaccines (45); however, 1-year survival for all study groups exceeded prior experience for advanced melanoma in cooperative group trials (32), even after correcting for multiple risk factors. The encouraging 1-year survival data may be impacted by patient selection, by the fact that 6 untreated patients were not included, and by capping LDH in the eligibility; however, 72% of analyzable patients had M1c disease, 28% had elevated LDH, 80% had 2 or more metastatic sites, 43% had PS 1, and prior systemic therapy included immunotherapy in 44% and chemotherapy in 49%; these features may be compared with patients treated with carboplatin and taxol on the recent E2603 trial, for which 57% had M1c disease, 39% had elevated LDH, 39% had PS1, and 58% were treatment-naïve; overall median survival with carboplatin and taxol was 11.1 months (46); overall median survival for the present trial E1602 was 11.8 months. Despite modestly encouraging early survival data with these multipeptide vaccines, most patients still died of their melanoma within 2 years. Thus, continued investigation of multipeptide vaccines in patients with advanced melanoma should be in combination with other active agents. In particular, BRAF inhibitors may transiently improve antigen expression by melanoma cells and T-cell infiltration into melanoma metastases (47–49). So, combination with vaccines may be considered as an approach to improve clinical activity of both agents.

C.L. Slingluff has a commercial research grant from Glaxo Smith Kline, has ownership interest (including patents) in UVA Licensing and Ventures Group (patents on peptides, through UVA), and is a consultant/advisory board member of Immatics and Polynoma. No potential conflicts of interest were disclosed by the other authors.

Conception and design: C.L. Slingluff, S. Lee, K.A. Chianese-Bullock, J.M. Kirkwood

Development of methodology: C.L. Slingluff, T.L. Whiteside

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): C.L. Slingluff, L.H Butterfield, T.L. Whiteside, P.D. Leming, J.M. Kirkwood

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): C.L. Slingluff, S. Lee, F. Zhao, L.H Butterfield

Writing, review, and/or revision of the manuscript: C.L. Slingluff, S. Lee, F. Zhao, K.A. Chianese-Bullock, W.C. Olson, L.H Butterfield, P.D. Leming, J.M. Kirkwood

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): C.L. Slingluff, F. Zhao, W.C. Olson, J.M. Kirkwood

Study supervision: C.L. Slingluff, J.M. Kirkwood

Providing peptides for use in the trial, from our research program at UVA: C.L. Slingluff

Development of the clinical trial protocol: K.A. Chianese-Bullock

This study was supported by the Eastern Cooperative Oncology Group (ECOG; Principal investigator and Director of ECOG: Robert L. Comis) and supported in part by Public Health Service Grants CA23318, CA66636, CA21115, CA49957, CA39229, and CA104362 from the National Cancer Institute, NIH, and the Department of Health and Human Services. This study was also supported in part by NIH/NCI grant NIH R01 CA118386 (to C.L. Slingluff), by the University of Virginia Cancer Center Support Grant (NIH/NCI P30 CA44579: Clinical Trials Office, Biorepository and Tissue Research Facility, Flow Cytometry Core, and Biomolecular Core Facility); and the UVA General Clinical Research Center (NIH M01 RR00847). The ECOG Central Immunology Laboratory is supported by Frontier Science and Technology Research Foundation. GM-CSF used in the vaccine adjuvant was provided by Berlex/Genzyme. Peptides used in this vaccine were prepared with philanthropic support from the Commonwealth Foundation for Cancer Research and Alice and Bill Goodwin. Additional philanthropic support was provided by Frank and Jane Batten, the James and Rebecca Craig Foundation, George S. Suddock, Richard and Sherry Sharp, and the Patients and Friends Research Fund of the University of Virginia Cancer Center. The Cancer Therapy Evaluation Program (CTEP) of the National Cancer Institute managed distribution of the vaccine preparations and adjuvant to participating centers.

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.

1.
Higano
CS
,
Schellhammmer
PF
,
Small
EJ
,
Burch
PA
,
Nemunaitis
J
,
Yuh
L
, et al
Integrated data from 2 randomized, double-blind, placebo-controlled, phase 3 trials of active cellular immunotherapy with sipuleucel-T in advanced prostate cancer
.
Cancer
2009
;
115
:
3670
9
.
2.
Sipuleucel-T: APC 8015, APC-8015, prostate cancer vaccine–Dendreon
.
Drugs R D
2006
;
7
:
197
201
.
3.
Small
EJ
,
Schellhammmer
PF
,
Higano
CS
,
Redfern
CH
,
Nemunaitis
JJ
,
Valone
FH
, et al
Placebo-controlled phase 3 trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asympotomiatic hormone refractory prostate cancer
.
J Clin Oncol
2006
;
24
:
3089
94
.
4.
Walter
S
,
Weinschenk
T
,
Stenzl
A
,
Zdrojowy
R
,
Pluzanska
A
,
Szczylik
C
, et al
Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival
.
Nat Med
2012
;
18
:
1254
61
.
5.
Schwartzentruber
DJ
,
Lawson
DH
,
Richards
JM
,
Conry
RM
,
Miller
DM
,
Treisman
J
, et al
gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma
.
N Engl J Med
2011
;
364
:
2119
27
.
6.
Slingluff
CL
 Jr
,
Darrow
T
,
Vervaert
C
,
Quinn-Allen
MA
,
Seigler
HF
. 
Human cytotoxic T cells specific for autologous melanoma cells: successful generation from lymph node cells in seven consecutive cases
.
J Natl Cancer Inst
1988
;
80
:
1016
26
.
7.
Slingluff
CL
 Jr
,
Darrow
TL
,
Seigler
HF
. 
Melanoma-specific cytotoxic T cells generated from peripheral blood lymphocytes. Implications of a renewable source of precursors for adoptive cellular immunotherapy [see comments]
.
Ann Surg
1989
;
210
:
194
202
.
8.
Darrow
TL
,
Slingluff
CL
 Jr
,
Seigler
HF
. 
The role of HLA class I antigens in recognition of melanoma cells by tumor-specific cytotoxic T lymphocytes. Evidence for shared tumor antigens
.
J Immunol
1989
;
142
:
3329
35
.
9.
Topalian
SL
,
Hom
SS
,
Kawakami
Y
,
Mancini
M
,
Schwartzentruber
DJ
,
Zakut
R
, et al
Recognition of shared melanoma antigens by human tumor-infiltrating lymphocytes. [Review] [11 refs]
.
J Immunother
1992
;
12
:
203
6
.
10.
van der Bruggen
P
,
Traversari
C
,
Chomez
P
,
Lurquin
C
,
De Plaen
E
,
Van den Eynde
B
, et al
A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma
.
Science
1991
;
254
:
1643
7
.
11.
Cox
AL
,
Skipper
J
,
Chen
Y
,
Henderson
RA
,
Darrow
TL
,
Shabanowitz
J
, et al
Identification of a peptide recognized by five melanoma-specific human cytotoxic T cell lines
.
Science
1994
;
264
:
716
9
.
12.
Renkvist
N
,
Castelli
C
,
Robbins
PF
,
Parmiani
G
. 
A listing of human tumor antigens recognized by T cells
.
Cancer Immunol Immunother
2001
;
50
:
3
15
.
13.
Brinckerhoff
LH
,
Thompson
LW
,
Slingluff
CL
 Jr
. 
Melanoma vaccines. [Review] [142 refs]
.
Curr Opin Oncol
2000
;
12
:
163
73
.
14.
de Vries
TJ
,
Smeets
M
,
de Graaf
R
,
Hou-Jensen
K
,
Brocker
EB
,
Renard
N
, et al
Expression of gp100, MART-1, tyrosinase, and S100 in paraffin-embedded primary melanomas and locoregional, lymph node, and visceral metastases: implications for diagnosis and immunotherapy. A study conducted by the EORTC Melanoma Cooperative Group. [see comments]
.
J Pathol
2001
;
193
:
13
20
.
15.
Hofbauer
GF
,
Kamarashev
J
,
Geertsen
R
,
Boni
R
,
Dummer
R
. 
Tyrosinase immunoreactivity in formalin-fixed, paraffin-embedded primary and metastatic melanoma: frequency and distribution
.
J Cutan Pathol
1998
;
25
:
204
9
.
16.
Cormier
JN
,
Abati
A
,
Fetsch
P
,
Hijazi
YM
,
Rosenberg
SA
,
Marincola
FM
, et al
Comparative analysis of the in vivo expression of tyrosinase, MART-1/Melan-A, and gp100 in metastatic melanoma lesions: implications for immunotherapy
.
J Immunother
1998
;
21
:
27
31
.
17.
Slingluff
CL
 Jr
,
Colella
TA
,
Thompson
L
,
Graham
DD
,
Skipper
JC
,
Caldwell
J
, et al
Melanomas with concordant loss of multiple melanocytic differentiation proteins: immune escape that may be overcome by targeting unique or undefined antigens
.
Cancer Immunol Immunother
2000
;
48
:
661
72
.
18.
Hofbauer
GF
,
Schaefer
C
,
Noppen
C
,
Boni
R
,
Kamarashev
J
,
Nestle
FO
, et al
MAGE-3 immunoreactivity in formalin-fixed, paraffin-embedded primary and metastatic melanoma: frequency and distribution
.
Am J Pathol
1997
;
151
:
1549
53
.
19.
Brasseur
F
,
Rimoldi
D
,
Lienard
D
,
Lethe
B
,
Carrel
S
,
Arienti
F
, et al
Expression of MAGE genes in primary and metastatic cutaneous melanoma
.
Int J Cancer
1995
;
63
:
375
80
.
20.
Slingluff
CL
 Jr
,
Petroni
GR
,
Chianese-Bullock
KA
,
Smolkin
ME
,
Hibbitts
S
,
Murphy
C
, et al
Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting
.
Clin Cancer Res
2007
;
13
:
6386
95
.
21.
Slingluff
CL
 Jr
,
Petroni
GR
,
Olson
WC
,
Smolkin
ME
,
Ross
MI
,
Haas
NB
, et al
Effect of GM-CSF on circulating CD8+ and CD4+ T cell responses to a multipeptide melanoma vaccine: Outcome of a multicenter randomized trial
.
Clin Cancer Res
2009
;
15
:
7036
44
.
22.
Slingluff
CL
 Jr
,
Petroni
GR
,
Chianese-Bullock
KA
,
Smolkin
ME
,
Ross
MI
,
Haas
NB
, et al
A randomized multicenter trial of the effects of melanoma-associated helper peptides and cyclophosphamide on the immunogenicity of a multipeptide melanoma vaccine
.
J Clin Oncol
2011
;
29
:
2924
32
.
23.
Slingluff
CL
 Jr
,
Petroni
GR
,
Olson
W
,
Czarkowski
AR
,
Grosh
WW
,
Smolkin
M
, et al
Helper T cell responses and clinical activity of a melanoma vaccine with multiple peptides from MAGE and melanocytic differentiation antigens
.
J Clin Oncol
2008
;
26
:
4973
80
.
24.
Slingluff
CL
 Jr
,
Yamshchikov
G
,
Neese
P
,
Galavotti
H
,
Eastham
S
,
Engelhard
VH
, et al
Phase I trial of a melanoma vaccine with gp100(280–288) peptide and tetanus helper peptide in adjuvant: immunologic and clinical outcomes
.
Clin Cancer Res
2001
;
7
:
3012
24
.
25.
Chianese-Bullock
KA
,
Lewis
ST
,
Sherman
NE
,
Shannon
JD
,
Slingluff
CL
 Jr
. 
Multi-peptide vaccines vialed as peptide mixtures can be stable reagents for use in peptide-based immune therapies
.
Vaccine
2009
;
27
:
1764
70
.
26.
Slingluff
CL
 Jr
,
Yamshchikov
GV
,
Hogan
KT
,
Hibbitts
SC
,
Petroni
GR
,
Bissonette
EA
, et al
Evaluation of the sentinel immunized node for immune monitoring of cancer vaccines
.
Ann Surg Oncol
2008
;
15
:
3538
49
.
27.
Agresti
A
. 
Analysis of ordinal categorical data
. Hoboken (NJ):
Wiley and Sons
; 
1984
.
28.
Kaplan
EL
,
Meier
P
. 
Nonparametric estimation from incomplete observations
.
J Am Stat Assoc
1958
;
53
:
457
81
.
29.
King
G
,
Zeng
L
. 
Logistic regression in rare events data
.
Pol Anal
2001
;
9
:
137
63
.
30.
Cox
DR
. 
Regression models and life tables (with discussion)
.
J R Stat Soc B
1972
;
34
:
187
220
.
31.
Anderson
JR
,
Cain
KC
,
Gelber
RD
. 
Analysis of survival by tumor response
.
J Clin Oncol
1983
;
1
:
710
9
.
32.
Korn
EL
,
Liu
PY
,
Lee
SJ
,
Chapman
JA
,
Niedzwiecki
D
,
Suman
VJ
, et al
Meta-analysis of phase II cooperative group trials in metastatic stage IV melanoma to determine progression-free and overall survival benchmarks for future phase II trials
.
J Clin Oncol
2009
;
26
:
527
34
.
33.
Hailemichael
Y
,
Dail
Z
,
Jaffarzad
N
,
Ye
Y
,
Medina
MA
,
Huang
X-F
, et al
Persistent antigen at vaccination sites induces tumor-specific CD8+ T cell sequestration, dysfunction and deletion
.
J Exp Med
2013
;
19
:
465
72
.
34.
Lichterfeld
M
,
Kaufmann
DE
,
Yu
XG
,
Mui
SK
,
Addo
MM
,
Johnston
MN
, et al
Loss of HIV-1-specific CD8+ T cell proliferation after acute HIV-1 infection and restoration by vaccine-induced HIV-1-specific CD4+ T cells
.
J Exp Med
2004
;
200
:
701
12
.
35.
Shirai
M
,
Chen
M
,
Arichi
T
,
Masaki
T
,
Nishioka
M
,
Newman
M
, et al
Use of intrinsic and extrinsic helper epitopes for in vivo induction of anti-hepatitis C virus cytotoxic T lymphocytes (CTL) with CTL epitope peptide vaccines
.
J Infect Dis
1996
;
173
:
24
31
.
36.
Shirai
M
,
Pendleton
CD
,
Ahlers
J
,
Takeshita
T
,
Newman
M
,
Berzofsky
JA
. 
Helper-cytotoxic T lymphocyte (CTL) determinant linkage required for priming of anti-HIV CD8 +CTL in vivo with peptide vaccine constructs
.
J Immunol
1994
;
152
:
549
56
.
37.
Ahlers
JD
,
Dunlop
N
,
Pendleton
CD
,
Newman
M
,
Nara
PL
,
Berzofsky
JA
. 
Candidate HIV type 1 multideterminant cluster peptide-P18MN vaccine constructs elicit type 1 helper T cells, cytotoxic T cells, and neutralizing antibody, all using the same adjuvant immunization
.
AIDS Res Hum Retroviruses
1996
;
12
:
259
72
.
38.
Bijker
MS
,
van den Eeden
SJ
,
Franken
KL
,
Melief
CJ
,
Offringa
R
,
van der Burg
SH
. 
CD8+ CTL priming by exact peptide epitopes in incomplete Freund's adjuvant induces a vanishing CTL response, whereas long peptides induce sustained CTL reactivity
.
J Immunol
2007
;
179
:
5033
40
.
39.
Zarei
S
,
Schwenter
F
,
Luy
P
,
Aurrand-Lions
M
,
Morel
P
,
Kopf
M
, et al
Role of GM-CSF signaling in cell-based tumor immunization
.
Blood
2009
;
113
:
6658
68
.
40.
Mittendorf
EA
,
Clifton
GT
,
Holmes
JP
,
Clive
KS
,
Patil
R
,
Benavides
LC
, et al
Clinical trial results of the HER-2/neu (E75) vaccine to prevent breast cancer recurrence in high-risk patients: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02
.
Cancer
2012
;
118
:
2594
602
.
41.
Faries
MB
,
Hsueh
EC
,
Ye
X
,
Hoban
M
,
Morton
DL
. 
Effect of granulocyte/macrophage colony-stimulating factor on vaccination with an allogeneic whole-cell melanoma vaccine
.
Clin Cancer Res
2009
;
15
:
7029
35
.
42.
Kirkwood
JM
,
Lee
S
,
Moschos
SJ
,
Albertini
MR
,
Michalak
JC
,
Sander
C
, et al
Immunogenicity and antitumor effects of vaccination with peptide vaccine+/-granulocyte-monocyte colony-stimulating factor and/or IFN-alpha2b in advanced metastatic melanoma: Eastern Cooperative Oncology Group Phase II Trial E1696
.
Clin Cancer Res
2009
;
15
:
1443
51
.
43.
Harlin
H
,
Meng
Y
,
Peterson
AC
,
Zha
Y
,
Tretiakova
M
,
Slingluff
C
, et al
Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment
.
Cancer Res
2009
;
69
:
3077
85
.
44.
Dengel
LT
,
Norrod
AG
,
Gregory
BL
,
Burdick
MD
,
Strieter
RM
,
Slingluff
CL
 Jr
, et al
Interferons induce CXCR3-cognate chemokine production by human metastatic melanoma
.
J Immunother
2010
;
33
:
965
74
.
45.
Rosenberg
SA
,
Yang
JC
,
Restifo
NP
. 
Cancer immunotherapy: moving beyond current vaccines
.
Nat Med
2004
;
10
:
909
15
.
46.
Flaherty
KT
,
Lee
SJ
,
Zhao
F
,
Schuchter
LM
,
Flaherty
L
,
Kefford
R
, et al
Phase III trial of carboplatin and paclitaxel with or without sorafenib in metastatic melanoma
.
J Clin Oncol
2013
;
20
;
31
:
373
9
.
47.
Frederick
DT
,
Piris
A
,
Cogdill
AP
,
Cooper
ZA
,
Lezcano
C
,
Ferrone
CR
, et al
BRAF inhibition is associated with enhanced melanoma antigen expression and a more favorable tumor microenvironment in patients with metastatic melanoma
.
Clin Cancer Res
2013
;
19
:
1225
31
.
48.
Boni
A
,
Cogdill
AP
,
Dang
P
,
Udayakumar
D
,
Njauw
CN
,
Sloss
CM
, et al
Selective BRAFV600E inhibition enhances T-cell recognition of melanoma without affecting lymphocyte function
.
Cancer Res
2010
;
70
:
5213
9
.
49.
Wilmott
JS
,
Long
GV
,
Howle
JR
,
Haydu
LE
,
Sharma
RN
,
Thompson
JF
, et al
Selective BRAF inhibitors induce marked T-cell infiltration into human metastatic melanoma
.
Clin Cancer Res
2012
;
18
:
1386
94
.