Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Sep 1.
Published in final edited form as: Cancer J. 2011 Sep;17(5):343–350. doi: 10.1097/PPO.0b013e318233e5b2

The Present and Future of Peptide Vaccines for Cancer: Single or Multiple, Long or Short, Alone or in Combination?

Craig L Slingluff Jr
PMCID: PMC3204371  NIHMSID: NIHMS328333  PMID: 21952285

Abstract

Peptide vaccines incorporate one or more short or long amino acid sequences as tumor antigens, combined with a vaccine adjuvant. Thus, they fall broadly into the category of defined-antigen vaccines, along with vaccines using protein, protein subunits, DNA, or RNA. They remain one of the most immunogenic approaches, based on measures of T cell response in the blood or in draining lymph nodes. However, existing peptide vaccines have had limited success at inducing clinical tumor regressions, despite reliable induction of T cell responses. Several new developments offer promise for improving peptide vaccines, including use of long peptides, optimization of adjuvants including toll-like receptor agonists, and combination with systemic therapies that may reduce tumor-associated immune dysfunction, such as blockade of PD-1/PD-L1 interactions. To apply these new approaches optimally, it will be critical to study their effects in the context of defined antigens, for which peptide vaccines are optimal.

Keywords: Peptide, Neoplasm, Melanoma, Immune therapy, TLR agonists, Vaccine, cancer vaccine


Peptide vaccines for cancer offer the promise of inducing T cells reactive to well-characterized tumor antigens and also enabling assessment of vaccination effect, by monitoring antigen-specific T cell responses. Cancer cells express peptide antigens recognized by CD8+ cytotoxic T lymphocytes (CTL)1, which are typically 8-10 amino acids long and are presented in association with Class I MHC molecules. The peptides recognized by helper (CD4+) T-cells are presented in association with Class II MHC molecules and are usually longer (13-18 amino acids in length), although peptide elution studies have indicated no apparent restriction on peptide length. For melanoma, the melanocytic differentiation proteins (MDPs) and the cancer-testis antigens (CTAs) are the most common source proteins for these defined shared peptide antigens. Now, a large number of peptide epitopes recognized by melanoma-reactive human CTL and helper T-cells are known2,3, making it possible to design vaccines using these antigens. We and others have found that selected peptides can induce circulating T cell responses in a majority of patients,4,5 and that vaccination with a mixture of peptides is immunogenic in up to 100% of patients.4 The magnitude of T cell responses sometimes is substantial, with 1-5% of circulating CD8 T cells reactive to single antigens 6-9, which rivals the magnitude of T cell responses to individual CMV antigens in humans 10; however, responses in most patients are 1-2 orders of magnitude lower, which may or may not be adequate for clinical benefit. T cell responses to vaccines may be durable for months or years, but are at least as likely to be transient, sometimes declining even while still receiving vaccines.11 However, T cells induced by vaccination can recognize and lyse melanoma cells expressing the relevant protein and MHC12,13. Thus, peptide vaccines induce promising immunogenicity. However, the transience and low magnitude of responses in many patients presents a need for improving the immunogenicity and for ensuring that memory responses are induced.

Clinically, there have been durable clinical responses in some patients receiving melanoma vaccines, suggesting the potential for clinical activity.14 However, overall clinical response rates are only about 3-5% 15. Thus, vaccines are not optimized: the antigens and the adjuvants may both be improved. Also, circulating immune responses have not consistently correlated with clinical outcome4,16. Arguably, that is not surprising, especially in the setting of advanced tumor burden. Both antigenic heterogeneity and tumor-associated immune dysfunction are characteristic of the tumor microenvironment (TME). Adoptive therapy with T cell clones specific for a single antigen has led to eradication of melanoma cells expressing that antigen, but the tumors have not regressed, because of the persistence of antigen-loss variants 17. Furthermore, T cells infiltrating tumor deposits are commonly found to be anergic or poorly responsive to antigenic stimulation, leading to the perception that the tumor microenvironment is hostile to the T cell response 18. Effective immune therapy may require induction of T cell responses to multiple numbers of antigens simultaneously, and maintenance of T cell activation in the tumor deposits. Combination with approaches to block immunoregulatory mechanisms may well also be needed for immune therapy to be most successful.

ADVANTAGES OF PEPTIDE VACCINES

There are several advantages of peptide vaccines over other cancer vaccine approaches (Table 1). Aside from the ease of synthesizing them, and their safety demonstrated in many trials, they have been effective at inducing T cell responses. Short peptides (typically nine amino acid residues) bind to Class I MHC molecules and induce CD8 T cells that can lyse melanoma cells expressing the cognate MHC and peptide 12,13. Immune response rates vary, depending on the peptides and adjuvants used, and depending on the assay method. However, immune response rates approaching 100% can be achieved 4,8,19,20, and the proportion of CD8+ cells responding to individual peptide antigens can exceed 1% 7-9,20. Though MHC-restriction of individual peptides limits their use to a subset of patients, we have found that mixtures of a dozen peptides restricted by HLA-A1, A2, A3, or A11 can be prepared as a stable mixture 21 and can induce immune responses in the 85% of patients with melanoma who express one or more of those MHC molecules4,8,20, without negative effects from competition among the peptides4. Other experience supports the ability to induce T cell responses to multiple peptides when vaccinating with peptide mixtures 22.

Table 1. Advantages and Limitations of Peptide Vaccines.

Advantages Limitations
  • Readily synthesized and purified at low cost

  • Stable in many storage conditions

  • Off-the-shelf reagent

  • Safe

  • Very effective at inducing CD8 or CD4 T cell responses in vivo in humans

  • Enables direct monitoring of T cell responses induced by the vaccine.

  • Safety in many studies

  • Using defined epitopes avoids use of uncharacterized antigens that may have nontherapeutic autoimmune activity

  • Repeated booster vaccines feasible

  • Class I MHC restriction limits relevance of individual peptides to certain HLA types.

  • Short peptides may bind directly to MHC on non-professional APC*, which may induce tolerance

  • Rapidly degraded by serum/tissue peptidases

  • Peptides with low affinity for MHC may be poorly immunogenic

  • Patients have variable repertoires for melanoma antigens: a peptide vaccine may have to include a large number of peptides to be useful across a wide range of patients.

  • Immune responses may be transient and/or of low magnitude

*

APC = antigen-presenting cell

LIMITATIONS OF PEPTIDE VACCINES

Short peptides restricted by Class I MHC molecules can bind directly into the peptide-binding groove on the exposed surface of the appropriate Class I MHC molecule. In vivo, when a peptide vaccine is administered into the subcutaneous tissue (or other sites), the peptides may be able to bind to numerous types of cells, only a few of which are professional APC. When they bind to non-professional APC (e.g. fibroblasts), they are presented without optimal co-stimulation; presentation of antigen in this way can even be toleragenic. Thus, there is concern that the effectiveness of vaccination with short peptides may be limited by this phenomenon. Also, these short peptides have little or no tertiary structure and thus are subject to rapid degradation by tissue and serum peptidases. We have estimated the half-life of a MelanA/MART-1 peptide in fresh human plasma to be approximately 22 seconds, and found that both exopeptidases and endopeptidases were involved in the degradation. It is possible that the low magnitude and transience of T cell responses observed in many patients vaccinated with short peptides may be explained in part by rapid degradation of these peptides in vivo, before they can be presented by professional APC, and also by suboptimal antigen presentation when it occurs.

Another factor is that when a vaccine is administered in the skin, it is generally believed that antigen presentation depends on dendritic cells bearing the antigen to migrate to the regional draining nodes and to present the antigen there to naïve circulating lymphocytes. When an antigen is presented by dendritic cells (DC) that have taken up a whole protein or DNA, the peptide of interest is going to be presented on an ongoing basis because it is generated inside the cell over time and presented on MHC. However, when a short peptide is used, and its ability to be presented to T cells in the draining node depends on its ability to remain bound to the MHC; so short peptides with low affinity for the MHC may be less immunogenic than they would be if they were more continuously being presented, for which one example may be gp100280-288 23,24.

SINGLE VS MULTIPLE PEPTIDES

Melanomas often lose expression of one or all melanocytic differentiation antigens as they progress 25, and cancer-testis antigens are expressed only in a subset of patients. Thus, no single antigen will be adequate for all melanomas. Also, antigenic heterogeneity, and antigen loss phenotypes are common; so even if a melanoma expresses a specific antigen, some of the cells in that tumor may not express it. Thus, immunologic control of melanoma likely requires a broad immune response against multiple antigens. It is hypothesized that an effective immune response against a single antigen can induce epitope-spreading – induction of immune responses against other antigens. However, it is also possible for vaccines to target multiple antigens directly.

In developing multipeptide vaccines, there has been some concern that if multiple peptides binding the same MHC molecule are co-administered, binding of lower-affinity peptides to the MHC may be competitively inhibited by higher-affinity peptides. Because of this concern, multipeptide vaccines at some centers have been administered so that each peptide is administered at a different body site 26. This approach will become increasingly unwieldy if the number of peptides exceeds three or four. In preclinical studies from our group, we have found that competition among peptides for MHC binding does not significantly inhibit T-cell induction or T-cell effector function 27,28. On the other hand, a clinical study raised concern about the effect of mixing the peptides YMDGTMSQV (tyrosinase 369-377) and IMDQVPFSV (gp100 209-2M29). That report was based on two sequential clinical trials, so that there may have been other variables at play. In a prospective randomized trial, we tested the immunogenicity of a 12-peptide vaccine compared to a vaccine containing just 4 of those 12 peptides. We found that immunogenicity of the index peptides was maintained in those patients receiving the 12-peptide vaccine, and there was also a marked and significant increase in cumulative T-cell reactivity toward the 12-peptide vaccine. Thus, addition of two to three peptides binding the same class I MHC allele does not inhibit immunogenicity of an index peptide when administered at equimolar concentrations, and use of multiple peptides in a cancer vaccine is supported.

ADDITION OF CD4 EPITOPES

The role of CD4+ helper T Lymphocytes in anti-tumor immune responses

Most peptide vaccines have been designed to activate the CD8+ cytotoxic T cell arm of the host immune system, which plays a critical role in tumor eradication 30-34. However, some recent approaches target CD4+ Th cells. This is based in part on results from earlier studies which demonstrated depletion of CD4+ T-cells abrogates all or part of protective immune response to vaccines35. Furthermore, adoptive therapy with CD4+ T-cells has been shown to induce tumor protection in some model systems36, and there has been anecdotal clinical benefit in a patient after adoptive therapy with melanoma-reactive CD4+ T-cells 37. Thus, protective immunity induced by tumor cell vaccines and by adoptive T cell therapy may be mediated both by CD4+ T-cells.

Natural immune responses to pathogens consist of an integrated response including Th responses to epitopes presented by class II MHC molecules and CTL responses to epitopes presented by class I MHC molecules 38. Th cells can activate dendritic cells (DC) for heightened antigen presentation, causing the DC to secrete IL-2 and other cytokines that may help to direct the immune response. Furthermore, strong Th1 help produces the proper cytokine milieu (IFNγ, TNFα, IL-2) which is critical to the induction of immune-mediated tumor destruction39,40. In addition, Th responses are believed to be involved in the establishment of memory responses 41. Thus, there is rationale for induction of CD4+ T cells with cancer vaccines, either on their own, or in combination with stimulation of CD8+ T cells.

Induction of non-specific T cell help

One approach to induction of CD4+ T cell help is to use molecules that stimulate CD4+ T cell responses that are not specific for cancer antigens but may stimulate recall responses or other nonspecific help. Commonly used approaches have been to add keyhole limpet hemocyanin (KLH) 42-44, PADRE peptide 45, or a tetanus toxoid helper peptide tetanus 46,47. In one study, CD8+ T cell responses to Class I MHC-associated tyrosinase peptides appeared to be increased by co-administration of KLH 44. Although, KLH may have broader adjuvant properties than simply the effects on helper T cells, these data suggest a benefit of non-specific helper peptide stimulation. We have used a peptide epitope for T-helper cells derived from tetanus toxoid. (tetanus helper peptide) modified from the reported sequence by adding an alanine residue to the N-terminus, to avoid spontaneous conversion of glutamine at the N-terminus to pyroglutamate, and to increase stability (AQYIKANSKFIGITEL). Responses to that peptide are induced in over 90% of patients when vaccinating with IFA 8,20, and these responses appear to be predominantly Th1 responses 48.

Induction of melanoma-specific T cell help

HLA-DR restricted peptides have been identified from melanoma-associated proteins,49-55 but there is limited in vivo human experience with them. Epitopes for CD4+ helper T cells are typically longer than those for CD8+ T cells, and they are promiscuous in binding to many different Class II MHC molecules. Most prior studies with these “helper” peptides used only one or two peptides, and they limited enrollment to a single MHC Class II allele 56-58. We have tested a multipeptide helper peptide vaccine to stimulate melanoma-reactive CD4+ T cells, using 6 melanoma helper peptides (6MHP) from melanocytic differentiation antigens and cancer testis antigens, restricted by HLA-DR molecules (Figure 1) 14. The findings demonstrate safety and immunogenicity. Immune responses to the 6 MHP pool were detected in over 80% of patients, across a wide range of HLA-DR molecules, suggesting these peptides may be broadly relevant to the immune response to melanoma.14 Findings also suggest promiscuity of these helper peptides across a wider range of HLA-DR molecules than originally reported. Some immune responses were detectable through week 39, more than 6 months after the last vaccine, suggesting induction of memory in those patients. However, the immune responses were transient in some other cases, suggesting immune regulatory processes that should be identified and targeted for combination immunotherapy in the future.

Figure 1.

Figure 1

Schematic of vaccine components for melanoma vaccine testing addition of 6 melanoma helper peptides (6MHP) or tetanus helper peptide in addition to 12 melanoma peptides, in the adjuvant therapy of high-risk resected melanoma.

In addition to in vitro evidence of immunogenicity of this 6-helper peptide vaccine, we also observed in vivo evidence of immune reactivity, based on DTH responses in 7 of 24 evaluable patients (29%). There were also autoimmune reactivities in 21% of patients, including vitiligo in 10%, without associated symptoms. Durable objective clinical responses were observed in 2 of 17 patients with measurable disease, and durable disease stabilization occurred in 2 additional patients.14 Immune responses were identified to 6MHP for all patients with DTH responses, all patients with autoimmune toxicities, and all four patients with partial clinical responses or stable disease. Together, these data suggest both biological activity and evidence of clinical activity. This phase I/II trial provided data supporting larger studies of this 6 helper peptide mixture with or without immunogens to stimulate CD8+ T cells.

Combining class I MHC restricted peptides & class II MHC restricted peptides in vaccines

Because CD4+ cells have a fundamental and comprehensive role in initiation and maintenance of cytotoxic T cell responses41, vaccination with cytotoxic T-cell epitopes may be more successful when the vaccine includes helper epitopes from the same protein(s) rather than with non-specific helper epitopes. In HIV patients, there are data suggesting that induction of CD4+ responses to HIV antigens with an HIV-specific peptide augments HIV-specific CD8+ T cell reactivity 59. Considering the strong rationale for the induction of helper T cell responses to melanoma antigens, and human data supporting the approach in the HIV setting, it is rational to test whether vaccination with melanoma-associated helper peptides that can induce antigen-reactive CD4+ T cell responses can increase CD8+ responses, compared to vaccination with a nonspecific helper peptide. Several trials have tested this concept in the setting of a single Class I restricted peptide and a single Class II restricted peptide. In a study with the gp10044-59 helper peptide, helper T cell responses were not induced, but CTL responses were paradoxically reduced compared to a prior study 60. That report raises a question of whether the addition of helper peptides may be harmful, rather than helpful, to the anti-tumor response. However, the conclusions of that report were confounded by several critical weaknesses: (1) comparison of outcomes between two non-randomized pilot studies only, (2) marked differences in the proportion of patients with prior chemotherapy between the study groups (23% vs 58%), (3) very low immunogenicity of the MART-1 peptide in both arms, raising questions about vaccine immunogenicity, (4) complete absence of immunogenicity of the DR-restricted gp100 peptide, and (5) use of high-dose IL-2 in some patients, that may alter measured responses. 11

In another study with class II peptides only, that same gp100 peptide was not immunogenic, but another helper peptide, from MART-1/MelanA, was immunogenic 61. That study did not assess the impact of the helper peptide responses on CTL responses to Class I peptides, but there were some cytotoxic CD4+ responses generated 61. In another study, in epithelial cancers, with Her-2/neu peptides comprising overlapping epitopes for CD4+ and CD8+ T cells, CD8+ T cell responses were observed, but CD4+ responses were not reported 62.

To address formally the question of whether addition of melanoma helper peptides would increase CD8 T cell responses in a multipeptide vaccine, we have performed a multicenter randomized trial, Mel44, which enrolled 167 eligible patients with resected stage IIB-IV melanoma, who were randomized to 4 vaccination groups. Patients were vaccinated with 12 MHC Class I-restricted melanoma peptides (12MP) to stimulate CD8+ T cells, and randomized to receive a tetanus helper peptide or a mixture of 6 melanoma helper peptides (6MHP) to stimulate CD4+ T cells. Prior to vaccination, patients were also randomly assigned to receive CY pretreatment or not. T cell responses were assessed by ex vivo IFN-gamma ELIspot assay. Vaccination with 12MP plus tetanus induced CD8+ T cell responses in 78% of patients and CD4+ T cell responses to tetanus peptide in 93%. Vaccination with 12MP plus 6MHP induced CD8+ responses in 19% and CD4+ responses to 6MHP in 48%. CY had no significant effect on T cell responses. Thus, in this adjuvant setting, melanoma-associated helper peptides paradoxically decreased CD8+ T cell responses to a melanoma vaccine (p < 0.001), and CY pretreatment had no immunologic or clinical effect 20. Similar negative effects of combining helper peptides with class I peptides have been observed in an Eastern Cooperative Oncology trial 1602 63. Possible explanations for negative effects on CD8 responses include modulation of homing receptor expression or induction of antigen-specific regulatory T cells, and new data also raise the possibility that these negative results may be explained in part by effects of the vaccine adjuvants, locally at the site of vaccination.

PHOSPHO-PEPTIDES

As immune therapy becomes an effective and accepted approach for durable clinical regressions of melanoma and other cancers, the other most exciting area of new drug development for cancer therapy is in targeted therapies that inhibit molecular activation that is critical to the transformed phenotype. Most of the targeted therapies, including those of the MEK/BRAF pathway, are focused on blocking the activity of bioactive phosphoproteins. As effective as these therapies can be, rapid disease progression can occur after initial responses. A promising approach would be to combine such targeted therapies with immune therapy that is active against the phosphoproteins that maintain the malignant transformed phenotype. A new class of peptides being brought to the clinic in the near future are peptides that contain phosphoserine or phosphotyrosine residues, and which thus represent biologically active phosphoproteins that may be critical to the transformed phenotype Engelhard 64-66. They offer promise for a new and important class of antigen to target in future cancer vaccines.

ADJUVANTS FOR PEPTIDE VACCINES

IFA as immunologic adjuvant

This paper is focused on peptide vaccines, but the peptides cannot be isolated from their adjuvants. Surprisingly, effects of immunologic adjuvants in vivo in humans are not well-understood. The most common adjuvant for peptide vaccines for melanoma has been an incomplete Freund’s adjuvant (IFA), commonly Montanide ISA-51. There have been questions about whether the commonly used incomplete Freund’s adjuvant (IFA) Montanide ISA-51 (Seppic, Inc.; Paris, France) is a reliable immunological adjuvant 67,68. We have found it useful even in its newer formulation.67 However, it is increasingly evident that effective immunotherapy depends on the quality of the adjuvant, whose optimization will require an understanding of its function in vivo in humans at the vaccine site microenvironment (VSME). In a study of vaccination with peptides in IFA, we found that 1 week after 1 vaccine, Th2 cells (GATA-3+), but not Th1 cells (T-bet+), were increased in number, suggesting that peptide vaccination in IFA may induce a Th2-dominant VSME; this was supported also by induction of a large number of eosinophils at the vaccine site. Only after 3 vaccines was there conversion to a more balanced Th1/Th2 microenvironment69. Also, the frequency of FoxP3+ cells (putative regulatory T cells) increased with repeated vaccination69. The impact of FoxP3+ cells here can be debated, but this adjuvant does not appear to induce an optimal immunologic milieu at the VSME. These findings may contribute to the low magnitude and transience of CD8+ T cell responses to short peptide vaccines administered in IFA. An intriguing question also is whether the chronic inflammation induced by IFA can serve to attract antigen-specific T cells back to the vaccine site, thereby depleting them from circulation and reducing the number that may traffick to sites of metastasis 70. These findings together support seeking new adjuvants.

Toll-like receptor (TLR) agonists as vaccine adjuvants

The critical functions of vaccine adjuvants are not known, but may include activation of innate immunity, optimization of antigen presentation, recruitment of dendritic cells, and creating a cytokine environment that supports the desired immunologic outcome. Toll-like receptors are early mediators of innate immune responses to pathogens. They may improve vaccination efficacy, through activation of innate immune mechanisms, mediated in part by IFN-alpha signaling and by activating dendritic cells. Toll-like receptor (TLR) agonists offer the potential to improve the magnitude and persistence of antitumor T cell responses9; however, most trials of TLR agonists have been limited to use of one TLR agonist, and have not defined molecular and cellular effects at the vaccine site microenvironment (VSME). Several TLR agonists may be effective vaccine adjuvants, including agonists for TLR 3, 4, 7, 8, and 9. In murine models, the combination of TLR agonists and IFA has strong immunological adjuvant properties with peptide vaccines. Murine and human studies have also suggested value of combining agonists for 2-3 TLRs.71,72 Future human studies need to evaluate whether individual agonists for TLRs 3, 4, 7, 8, or 9 are better adjuvants than IFA alone, and also whether these TLR agonists alone or together may be more immunogenic with or without IFA. TLR agonists are likely to support a strong Th1 environment73-75. Some data suggest TLR8 agonists may decrease regulatory T cells76, though these data have not been replicated. Vaccination of humans with the oligonucleotide CpG7909 (TLR9 agonist) increases CD8 T cell responses to a short peptide when combined with IFA9, and vaccination with peptides and the TLR3 agonist polyICLC also shows promise in murine and human studies 77. Current trials with a MAGE-A3 protein use a complex adjuvant system that includes the TLR4 agonist Monophosphoryl Lipid A and the TLR9 agonist CpG oligonucleotide, in addition to the saponin QS-21 78. It is not yet known if this approach is optimal, or if it will be feasible or effective also for peptide vaccines. However, it does seem likely that some use of TLR agonists will be helpful to increase the immunogenicity of peptide vaccines.

Ligation of CD40

TLR activation synergizes with ligation of CD40 on DC; combining TLR agonists and CD40 ligation may augment immune responses to vaccines 79. In humans, antibody to CD40 may induce CD40 ligation, but many cells express CD40 and thus serve as sinks for systemic CD40 antibody. Another possible approach to ligate CD40 specifically on DC in the vaccine-site microenvironment (VSME) and in vaccine-draining lymph nodes (VDLN) is to take a lesson from physiologic immune responses to pathogens. When pathogens are encountered naturally in the skin, CD4 T cells are activated and upregulate CD40L, which then binds and activates CD40 to license DC. Thus, activation of CD4+ cells in the VSME and VDLN will upregulate CD40L on those cells. CD40L+ CD4 cells in turn license professional APC (DC) in tissues where antigen is presented. Progress in cancer vaccine development thus likely requires optimizing approaches to induce CD4+ T cells in the VSME and VDLN. The encouraging results of vaccinating with melanoma helper peptides alone are in contrast to the disappointing findings after vaccinating with melanoma helper peptides mixed with peptides to stimulate CD8+ T cells. Alteration in adjuvants may be critical to improving the ability to combine approaches to stimulate CD4+ T cells and CD8+ T cells.

LONG VS SHORT PEPTIDES

Short peptides may bind directly to MHC molecules on cells that are not professional antigen-presenting cells (APC), thereby potentially inducing tolerance or anergy80,81. In contrast, recent work with long (30-mer) peptides that encompass short minimal epitopes suggests that these longer peptides may be more effective immunogens than the minimal peptides. The extra length contributes to a tertiary structure that may protect from exopeptidase-mediated degradation, and they are too long to be presented directly on MHC; so they must be internalized by professional APC and processed for presentation (eg CD11c+ DC).82-85 Unlike short peptides, long peptides induce memory CD8+ T cell responses that are boosted dramatically on repeat vaccination in mice, and induce substantially improved tumor control compared to vaccination with short peptides.83,86 Induction of helper T cells reactive to epitopes within the long peptide have been implicated as necessary for longterm T cell memory86. This is supported by the finding that the improved immunologic responses and tumor control are blocked in mice knocked out for CD4 or for CD40. A vaccine using long (30-mer) peptides from HPV-16 for squamous vulvar neoplasia has induced clinical regressions in most patients, supporting clinical activity of long peptide vaccines.87 Using these long peptides promises to induce a broad and more durable adaptive immune responses against multiple antigens.

COMBINATION IMMUNE THERAPY

Even from the most optimistic perspective, it is unlikely that vaccines alone will provide durable clinical benefit for the majority of melanoma patients; however, it is likely that combination with other effective agents may lead to cumulative or synergistic benefit in large proportions of patients. New immunomodulatory agents with clinical activity are now available for use in melanoma. Among these, perhaps the most exciting are antibodies that block PD-1/PD-L1 interactions. The NCI’s Immunotherapy Agent Workshop prioritized therapeutic agents for use in cancer immunotherapy. Anti-PD-1 agents were ranked #2 88. There are at least 3 antibodies to PD-1 in clinical trials (MDX-1106, CT-011, and MK-3475). Clinical experience with one of these is that it induces objective clinical responses in 30% of patients with advanced melanoma, with high durability, and a safety profile that may be better than that of CTLA-4 antibody, and with MTD not reached in initial studies 89,90. PD-1 is expressed by activated T cells, B cells, and some myeloid cells. Its ligand, PD-L1 is expressed on many peripheral tissues and cell types, including melanoma cells. Its unique effects (as compared to CTLA-4, for example) may be mediated in part by its expression in peripheral tissues, including the tumor.91 PD-L1’s expression in peripheral tissues appears critical to maintaining peripheral tolerance91. PD-1/PD-L1 interaction can limit T cell reactivity even long after initial activation, and its blockade can restore immune function91,92. PD-1 ligation inhibits signaling through TCR activation; it depends on stimulation of the TCR at the time of PD-1/PD-L1/L2 ligation93. Thus, inhibition of PD-1/PD-L1 can be expected to improve dysfunction of tumor-reactive T cells in situ in the presence of tumor antigen, may improve reactivity to vaccines given simultaneously, or to chronic viral antigen (eg CMV) but may not affect memory responses to viral antigens in the absence of viral antigen (eg influenza). PD-L1 expression also is induced by antigen-mediated immune reactivity. PD-1 also can alter T cell movement 94; and blockade of PD-1 can increase T cell infiltration of peripheral tissues (eg in autoimmunity models)95,96 and thus may be expected to affect T cell homing receptor (HR) expression or expression of homing receptor ligands (HRL), and may be expected to augment T cell infiltration of melanoma metastases. Thus, PD-1/PD-L1 blockade has promise to increase T cell activation in the VSME and in the tumor microenvironment, while also enabling activated T cells to infiltrate metastases and to remain activated. Thus, there is strong rationale to combine an optimized peptide vaccine with an inhibitor of PD-1/PD-L1 interactions.

SUMMARY

Prospects for improving peptide vaccines include use of long peptides, modification of adjuvants, inclusion of new antigens, and combination therapy with other immunologically active agents. Evidence suggests that long peptides may be more immunogenic than short peptides, and studies evaluating them are underway. Current data suggest that long peptides may overcome many of the challenges with short peptide vaccines, by inducing both CD4+ and CD8+ responses with more optimal antigen presentation, and by enabling continued presentation of immunogenic but low affinity peptides as antigen-loaded DC migrate to regional nodes. TLR agonists may be more effective adjuvants than IFA, local or systemic GM-CSF, or systemic interferon-gamma. Preclinical studies support the value of TLR agonists as vaccine adjuvants, and some clinical studies support the value of TLR3, TLR4, and TLR9 agonists, in particular. However, the optimal TLR agonist(s), dose, timing, and mode of delivery remain to be determined. Very little is known about the molecular and cellular effects of various adjuvants, at the vaccine site and in the vaccine-draining node; an important area of future studies is to understand the local effects of TLR agonists and how they mediate adjuvanticity. A large number of antigens are known, but few have been studied in vaccines. A new class of peptides being brought to the clinic in the near future are peptides that contain phosphoserine or phosphotyrosine residues, which represent biologically active phosphoproteins that may be critical to the transformed phenotype.

A major limitation of all immune therapy is the fact that the tumor microenvironment is hostile to T cell infiltration, function and survival. Thus, in addition to optimizing the immune response induced by peptide vaccines, there is a need to understand critical molecular mediators of T cell trafficking to the tumor microenvironment and mediators of immune dysfunction in the tumor microenvironment. As evidence begins to accumulate for the therapeutic value of defined-antigen vaccines 87,97-99, it seems likely that improvements in vaccine immunogenicity, in T cell persistence, and reversal of tumor-associated immune dysfunction, will lead to improved therapeutic value of peptide vaccines in combination with optimal adjuvants and systemic immune modulation. Combinations of vaccines with IFN-alpha, GM-CSF, and CTLA-4 antibodies have not improved immune responses or clinical outcome compared to vaccine alone 8,16,100,101. On the other hand, combination of a peptide vaccine with high-dose IL-2 improved clinical response rate and progression-free survival, and induced a strong trend to improved survival (p=0.06). 99 Other clinically active agents offer promise to improve T cell responses and clinical outcome. Candidates include antibodies to PD-1 or PD-L1, agonistic antibody to CD137, and cytokines that may support T cell expansion and persistence, such as IL-7 and IL-15. As all of these new immune modulators are explored to augment cancer immunotherapy, studies will peptide vaccines will be particularly helpful because they are ideal for correlative studies of immune response to enable an understanding of the effects of each intervention.

Acknowledgments

Funding support and other disclosures. This work is funded by NIH/NCI grant NIH R01 CA118386 and R01 CA57653. Support for work presented here includes philanthropic support from the Commonwealth Foundation for Cancer Research and Alice and Bill Goodwin, 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. Dr. Slingluff has ongoing collaborations with Glaxo-Smith Kline Biologicals and Bristol-Myers Squibb, and is on scientific advisory boards for Roche, Curetech, and Immatics. Dr. Slingluff is an inventor on licensed patents for several peptides used in melanoma vaccine trials.

Footnotes

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.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. [PubMed] [Google Scholar]
  • 2.Brinckerhoff LH, Thompson LW, Slingluff CL., Jr. Melanoma vaccines. Curr Opin Oncol. 2000;12:163–173. doi: 10.1097/00001622-200003000-00012. [DOI] [PubMed] [Google Scholar]
  • 3.Novellino L, Castelli C, Parmiani G. A listing of human tumor antigens recognized by T cells: March 2004 update. Cancer Immunol Immunother. 2004 doi: 10.1007/s00262-004-0560-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Slingluff CL, Jr., Petroni GR, Chianese-Bullock KA, et al. Immunologic and clinical utcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res. 2007;13:6386–6395. doi: 10.1158/1078-0432.CCR-07-0486. [DOI] [PubMed] [Google Scholar]
  • 5.Slingluff CL, Jr., Chianese-Bullock KA, Bullock TN, et al. Immunity to melanoma antigens: from self-tolerance to immunotherapy. Adv Immunol. 2006;90:243–95. 243–295. doi: 10.1016/S0065-2776(06)90007-8. [DOI] [PubMed] [Google Scholar]
  • 6.Speiser DE, Pittet MJ, Rimoldi D, et al. Evaluation of melanoma vaccines with molecularly defined antigens by ex vivo monitoring of tumor-specific T cells. Semin Cancer Biol. 2003;13:461–472. doi: 10.1016/j.semcancer.2003.09.010. [DOI] [PubMed] [Google Scholar]
  • 7.Rosenberg SA, Sherry RM, Morton KE, et al. Tumor progression can occur despite the induction of very high levels of self/tumor antigen-specific CD8+ T cells in patients with melanoma. JI. 2005;175:6169–6176. doi: 10.4049/jimmunol.175.9.6169. [DOI] [PubMed] [Google Scholar]
  • 8.Slingluff CL, Jr., Petroni GR, Olson WC, 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;15:7036–7044. doi: 10.1158/1078-0432.CCR-09-1544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Speiser DE, Lienard D, Rufer N, et al. Rapid and strong human CD8+ T cell responses to vaccination with peptide, IFA, and CpG oligodeoxynucleotide 7909. J Clin Invest. 2005;115:739–746. doi: 10.1172/JCI23373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Slezak SL, Bettinotti M, Selleri S, Adams S, Marincola FM, Stroncek DF. CMV pp65 and IE-1 T cell epitopes recognized by healthy subjects. J Transl Med. 2007;5:17. doi: 10.1186/1479-5876-5-17. 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Slingluff CL, Jr., 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;22:4474–4485. doi: 10.1200/JCO.2004.10.212. [DOI] [PubMed] [Google Scholar]
  • 12.Yamshchikov GV, Barnd DL, Eastham S, et al. Evaluation of peptide vaccine immunogenicity in draining lymph nodes and blood of melanoma patients. Int J Cancer. 2001;92:703–711. doi: 10.1002/1097-0215(20010601)92:5<703::aid-ijc1250>3.0.co;2-5. [DOI] [PubMed] [Google Scholar]
  • 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. JI. 2005;174:3080–3086. doi: 10.4049/jimmunol.174.5.3080. [DOI] [PubMed] [Google Scholar]
  • 14.Slingluff CL, Jr., Petroni GR, Olson W, 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;26:4973–4980. doi: 10.1200/JCO.2008.17.3161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving beyond current vaccines. Nat Med. 2004;10:909–915. doi: 10.1038/nm1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Kirkwood JM, Lee S, Land S, et al. E1696: Final analysis of the clinical and immunological results of a multicenter ECOG phase II trial of multi-epitope peptide vaccination for stage IV melanoma with MART-1 (27-35), gp100 (209-217, 210M), and tyrosinase (368-376, 370D) (MGT) +/− IFNa2b and GM-CSF. J Clin Oncol. 2004;22:7502. Meeting Abstracts. [Google Scholar]
  • 17.Yee C, Thompson JA, Byrd D, et al. Adoptive T cell therapy using antigen-specific CD8+ T cell clones for the treatment of patients with metastatic melanoma: in vivo persistence, migration, and antitumor effect of transferred T cells. Proc Natl Acad Sci U S A. 2002;99:16168–16173. doi: 10.1073/pnas.242600099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zippelius A, Batard P, Rubio-Godoy V, et al. Effector function of human tumor-specific CD8 T cells in melanoma lesions: a state of local functional tolerance. Cancer Res. 2004;64:2865–2873. doi: 10.1158/0008-5472.can-03-3066. [DOI] [PubMed] [Google Scholar]
  • 19.Hamid O, Solomon JC, Scotland R, et al. Alum with interleukin-12 augments immunity to a melanoma peptide vaccine: correlation with time to relapse in patients with resected high-risk disease. Clin Cancer Res. 2007;13:215–222. doi: 10.1158/1078-0432.CCR-06-1450. [DOI] [PubMed] [Google Scholar]
  • 20.Slingluff CL, Jr., Petroni GR, Chianese-Bullock KA, 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 doi: 10.1200/JCO.2010.33.8053. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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;27:1764–1770. doi: 10.1016/j.vaccine.2009.01.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Baumgaertner P, Rufer N, Devevre E, et al. Ex vivo detectable human CD8 T-cell responses to cancer-testis antigens. Cancer Res. 2006;66:1912–1916. doi: 10.1158/0008-5472.CAN-05-3793. [DOI] [PubMed] [Google Scholar]
  • 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;264:716–719. doi: 10.1126/science.7513441. [DOI] [PubMed] [Google Scholar]
  • 24.Slingluff CL, Jr., Yamshchikov G, Neese P, 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;7:3012–3024. [PubMed] [Google Scholar]
  • 25.Slingluff CL, Jr., Colella TA, Thompson L, 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;48:661–672. doi: 10.1007/s002620050015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Powell DJ, Jr., Rosenberg SA. Phenotypic and functional maturation of tumor antigen-reactive CD8+ T lymphocytes in patients undergoing multiple course peptide vaccination. Journal of Immunotherapy. 2004;27:36–47. doi: 10.1097/00002371-200401000-00004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Thompson LW, Garbee CF, Hibbitts S, et al. Competition among peptides in melanoma vaccines for binding to MHC molecules. Journal of Immunotherapy. 2004;27:425–431. doi: 10.1097/00002371-200411000-00002. [DOI] [PubMed] [Google Scholar]
  • 28.Mullins DW, Engelhard VH. Limited infiltration of exogenous dendritic cells and naive T cells restricts immune responses in peripheral lymph nodes. JI. 2006;176:4535–4542. doi: 10.4049/jimmunol.176.8.4535. [DOI] [PubMed] [Google Scholar]
  • 29.Rosenberg SA, Sherry RM, Morton KE, et al. Altered CD8(+) T-cell responses when immunizing with multiepitope peptide vaccines. Journal of Immunotherapy. 2006;29:224–231. doi: 10.1097/01.cji.0000190399.98802.10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Fearon ER, Pardoll DM, Itaya T, et al. Interleukin-2 production by tumor cells bypasses T helper function in the generation of an antitumor response. Cell. 1990;60:397–403. doi: 10.1016/0092-8674(90)90591-2. [DOI] [PubMed] [Google Scholar]
  • 31.Golumbek PT, Lazenby AJ, Levitsky HI, et al. Treatment of established renal cancer by tumor cells engineered to secrete interleukin-4. Science. 1991;254:713–716. doi: 10.1126/science.1948050. [DOI] [PubMed] [Google Scholar]
  • 32.Dranoff G, Jaffee E, Lazenby A, et al. Vaccination with irradiated tumor cells engineered to secrete murine granulocyte-macrophage colony-stimulating factor stimulates potent, specific, and long-lasting anti-tumor immunity. Proc Natl Acad Sci USA. 1993;90:3539–3543. doi: 10.1073/pnas.90.8.3539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Kast WM, Offringa R, Peters PJ, et al. Eradication of adenovirus E1-induced tumors by E1A-specific cytotoxic T lymphocytes. Cell. 1989;59:603–614. doi: 10.1016/0092-8674(89)90006-8. [DOI] [PubMed] [Google Scholar]
  • 34.Greenberg PD, Riddell SR. Principles for adoptive T cell therapy of human viral diseases. Annual Review in Immunology. 1995;13:545–586. doi: 10.1146/annurev.iy.13.040195.002553. [DOI] [PubMed] [Google Scholar]
  • 35.Kayaga J, Souberbielle BE, Sheikh N, et al. Anti-tumour activity against B16-F10 melanoma with a GM-CSF secreting allogeneic tumour cell vaccine. Gene Therapy. 1999;6:1475–1481. doi: 10.1038/sj.gt.3300961. erratum appears in Gene Ther 1999 Nov;6(11):1905. [DOI] [PubMed] [Google Scholar]
  • 36.Kahn M, Sugawara H, McGowan P, et al. CD4+ T cell clones specific for the human p97 melanoma-associated antigen can eradicate pulmonary metastases from a murine tumor expressing the p97 antigen. J Immunol. 1991;146:3235–3241. [PubMed] [Google Scholar]
  • 37.Hunder NN, Wallen H, Cao J, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008;19(358):2698–2703. doi: 10.1056/NEJMoa0800251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Weiss WR, Sedegah M, Berzofsky JA, Hoffman SL. The role of CD4+ T cells in immunity to malaria sporozoites. J Immunol. 1993;151:2690–2698. [PubMed] [Google Scholar]
  • 39.Hung K, Hayashi R, Lafond-Walker A, Lowenstein C, Pardoll D, Levitsky H. The central role of CD4+ T cells in the antitumor immune response. Journal of Experimental Medicine. 1998;188:2357–2368. doi: 10.1084/jem.188.12.2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Matsui S, Ahlers JD, Vortmeyer AO, et al. A model for CD8+ CTL tumor immunosurveillance and regulation of tumor escape by CD4 T cells through an effect on quality of CTL. J Immunol. 1999;163:184–193. [PubMed] [Google Scholar]
  • 41.Pardoll DM, Topalian Sl. The role of CD4+ T cell responses in antitumor immunity. Current Opinion in Immunology. 1998;10:588–594. doi: 10.1016/s0952-7915(98)80228-8. [DOI] [PubMed] [Google Scholar]
  • 42.Na IK, Keilholz U, Letsch A, et al. Addition of GM-CSF to a peptide/KLH vaccine results in increased frequencies of CXCR3-expressing KLH-specific T cells. Cancer Immunol Immunother. 2007;56:391–396. doi: 10.1007/s00262-006-0198-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Helling F, Zhang S, Shang A, et al. GM2-KLH conjugate vaccine: increased immunogenicity in melanoma patients after administration with immunological adjuvant QS-21. Cancer Res. 1995;55:2783–2788. [PubMed] [Google Scholar]
  • 44.Scheibenbogen C, Schadendorf D, Bechrakis NE, et al. Effects of granulocyte-macrophage colony-stimulating factor and foreign helper protein as immunologic adjuvants on the T-cell response to vaccination with tyrosinase peptides. Int J Cancer. 2003;104:188–194. doi: 10.1002/ijc.10961. [DOI] [PubMed] [Google Scholar]
  • 45.Del Guercio MF, Alexander J, Kubo RT, et al. Potent immunogenic short linear peptide constructs composed of B cell epitopes and Pan DR T helper epitopes (PADRE) for antibody responses in vivo. Vaccine. 1997;15:441–448. doi: 10.1016/s0264-410x(97)00186-2. [DOI] [PubMed] [Google Scholar]
  • 46.Valmori D, Pessi A, Bianchi E, Corradin G. Use of human universally antigenic tetanus toxin T cell epitopes as carriers for human vaccination. JI. 1992;149:717–721. [PubMed] [Google Scholar]
  • 47.Demotz S. Delineation of several DR-restricted tetanus toxin T cell epitopes. JI. 1989;142:394–402. [PubMed] [Google Scholar]
  • 48.Slingluff CL, Jr, Yamshchikov G, Neese P, 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;7:3012–3024. [PubMed] [Google Scholar]
  • 49.Topalian SL, Gonzales MI, Parkhurst M, et al. Melanoma-specific CD4+ T cells recognize nonmutated HLA-DR-restricted tyrosinase epitopes. J Exp Med. 1996;183:1965–1971. doi: 10.1084/jem.183.5.1965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Kobayashi H, Kokubo T, Sato K, et al. CD4+ T cells from peripheral blood of a melanoma patient recognize peptides derived from nonmutated tyrosinase. Cancer Res. 1998;58:296–301. [PubMed] [Google Scholar]
  • 51.Zarour HM, Kirkwood JM, Kierstead LS, et al. Melan-A/MART-1(51-73) represents an immunogenic HLA-DR4-restricted epitope recognized by melanoma-reactive CD4(+) T cells. Proc Natl Acad Sci USA. 2000;97:400–405. doi: 10.1073/pnas.97.1.400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Manici S, Sturniolo T, Imro MA, et al. Melanoma cells present a MAGE-3 epitope to CD4(+) cytotoxic T cells in association with histocompatibility leukocyte antigen DR11. J Exp Med. 1999;189:871–876. doi: 10.1084/jem.189.5.871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Chaux P, Vantomme V, Stroobant V, et al. Identification of MAGE-3 epitopes presented by HLA-DR molecules to CD4(+) T lymphocytes. J Exp Med. 1999;189:767–778. doi: 10.1084/jem.189.5.767. see comments. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Halder T, Pawelec G, Kirkin AF, et al. Isolation of novel HLA-DR restricted potential tumor-associated antigens from the melanoma cell line FM3. Cancer Res. 1997;57:3238–3244. [PubMed] [Google Scholar]
  • 55.Li K, Adibzadeh M, Halder T, et al. Tumour-specific MHC-class-II-restricted responses after in vitro sensitization to synthetic peptides corresponding to gp100 and Annexin II eluted from melanoma cells. Cancer Immunol Immunother. 1998;47:32–38. doi: 10.1007/s002620050501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Khong HT, Yang JC, Topalian SL, et al. Immunization of HLA-A*0201 and/or HLA-DPbeta1*04 patients with metastatic melanoma using epitopes from the NY-ESO-1 antigen. Journal of Immunotherapy. 2004;27:472–477. doi: 10.1097/00002371-200411000-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Phan GQ, Touloukian CE, Yang JC, et al. Immunization of patients with metastatic melanoma using both class I- and class II-restricted peptides from melanoma-associated antigens. Journal of Immunotherapy. 2003;26:349–356. doi: 10.1097/00002371-200307000-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wong R, Lau R, Chang J, et al. Immune responses to a class II helper peptide epitope in patients with stage III/IV resected melanoma. Clin Cancer Res. 2004;10:5004–5013. doi: 10.1158/1078-0432.CCR-04-0241. [DOI] [PubMed] [Google Scholar]
  • 59.Lichterfeld M, Kaufmann DE, Yu XG, 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;200:701–712. doi: 10.1084/jem.20041270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Phan GQ, Touloukian CE, Yang JC, et al. Immunization of patients with metastatic melanoma using both class I- and class II-restricted peptides from melanoma-associated antigens. J Immunother. 2003;26:349–356. doi: 10.1097/00002371-200307000-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wong R, Lau R, Chang J, et al. Immune responses to a class II helper peptide epitope in patients with stage III/IV resected melanoma. Clin Cancer Res. 2004;10:5004–5013. doi: 10.1158/1078-0432.CCR-04-0241. [DOI] [PubMed] [Google Scholar]
  • 62.Knutson KL, Schiffman K, Disis Ml. Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patients. J Clin Invest. 2001;107:477–484. doi: 10.1172/JCI11752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Slingluff CL, Jr, Lee SJ, Chianese-Bullock KA, et al. First report of a randomized phase II trial of multi-epitope vaccination with melanoma peptides for cytotoxic T cells and helper T cells in patients with metastatic melanoma: An Eastern Cooperative Oncology Group Study (E1602) J Clin Oncol. 2010;28 abstract. Abstract 8508. [Google Scholar]
  • 64.Mohammed F, Cobbold M, Zarling AL, et al. Phosphorylation-dependent interaction between antigenic peptides and MHC class I: a molecular basis for the presentation of transformed self. Nat Immunol. 2008;9:1236–1243. doi: 10.1038/ni.1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Depontieu FR, Qian J, Zarling AL, et al. Identification of tumor-associated, MHC class II-restricted phosphopeptides as targets for immunotherapy. Proc Natl Acad Sci U S A. 2009;106:12073–12078. doi: 10.1073/pnas.0903852106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Li Y, Depontieu FR, Sidney J, et al. Structural basis for the presentation of tumor-associated MHC class II-restricted phosphopeptides to CD4+ T cells. J Mol Biol. 2010;399:596–603. doi: 10.1016/j.jmb.2010.04.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Slingluff CL, Jr., Petroni GR, Smolkin ME, et al. Immunogenicity for CD8+ and CD4+ T cells of two formulations of an incomplete Freund’s adjuvant for multipeptide melanoma vaccines. Journal of Immunotherapy. 2010 doi: 10.1097/CJI.0b013e3181e311ac. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Rosenberg SA, Yang JC, Kammula US, et al. Different adjuvanticity of incomplete freund’s adjuvant derived from beef or vegetable components in melanoma patients immunized with a peptide vaccine. J Immunother. 2010;33:626–629. doi: 10.1097/CJI.0b013e3181dac9de. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Schaefer JT, Patterson JW, Deacon DH, et al. Dynamic changes in cellular infiltrates with repeated cutaneous vaccination: A histologic and immunophenotypic analysis. Journal of Translational Medicine. 2010;8:79. doi: 10.1186/1479-5876-8-79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Overwijk W, Hailemichael Y, Dai Z, Jaffarzad N, Hwu P. Peptide/incomplete Freund adjuvant emulsion depots are a graveyard for tumor antigen-specific CD8+ T cells. Journal of Immunotherapy. 2009;32:971. abstract. [Google Scholar]
  • 71.Zhu Q, Egelston C, Gagnon S, et al. Using 3 TLR ligands as a combination adjuvant induces qualitative changes in T cell responses needed for antiviral protection in mice. J Clin Invest. 2010;120:607–616. doi: 10.1172/JCI39293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Makela SM, Strengell M, Pietila TE, Osterlund P, Julkunen I. Multiple signaling pathways contribute to synergistic TLR ligand-dependent cytokine gene expression in human monocyte-derived macrophages and dendritic cells. J Leukoc Biol. 2009;85:664–672. doi: 10.1189/jlb.0808503. [DOI] [PubMed] [Google Scholar]
  • 73.Lore K, Betts MR, Brenchley JM, et al. Toll-like receptor ligands modulate dendritic cells to augment cytomegalovirus- and HIV-1-specific T cell responses. J Immunol. 2003;171:4320–4328. doi: 10.4049/jimmunol.171.8.4320. [DOI] [PubMed] [Google Scholar]
  • 74.Puggioni F, Durham SR, Francis JN. Monophosphoryl lipid A (MPL) promotes allergen-induced immune deviation in favour of Th1 responses. Allergy. 2005;60:678–684. doi: 10.1111/j.1398-9995.2005.00762.x. [DOI] [PubMed] [Google Scholar]
  • 75.Mata-Haro V, Cekic C, Martin M, Chilton PM, Casella CR, Mitchell TC. The vaccine adjuvant monophosphoryl lipid A as a TRIF-biased agonist of TLR4. Science. 2007;316:1628–1632. doi: 10.1126/science.1138963. [DOI] [PubMed] [Google Scholar]
  • 76.Peng G, Guo Z, Kiniwa Y, et al. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science. 2005;309:1380–1384. doi: 10.1126/science.1113401. [DOI] [PubMed] [Google Scholar]
  • 77.Zhu X, Fallert-Junecko BA, Fujita M, et al. Poly-ICLC promotes the infiltration of effector T cells into intracranial gliomas via induction of CXCL10 in IFN-alpha and IFN-gamma dependent manners. Cancer Immunol Immunother. 2010;59:1401–1409. doi: 10.1007/s00262-010-0876-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Cluff CW. Monophosphoryl Lipid A (MPL) as an Adjuvant for Anti-Cancer Vaccines: Clinical Results. Adv Exp Med Biol. 2010;667:111–23. doi: 10.1007/978-1-4419-1603-7_10. 111-123. [DOI] [PubMed] [Google Scholar]
  • 79.Ahonen CL, Wasiuk A, Fuse S, et al. Enhanced efficacy and reduced toxicity of multifactorial adjuvants compared with unitary adjuvants as cancer vaccines. Blood. 2008;111:3116–3125. doi: 10.1182/blood-2007-09-114371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Toes RE, Blom RJ, Offringa R, Kast WM, Melief CJ. Enhanced tumor outgrowth after peptide vaccination. Functional deletion of tumor-specific CTL induced by peptide vaccination can lead to the inability to reject tumors. J Immunol. 1996;156:3911–3918. [PubMed] [Google Scholar]
  • 81.Toes RE, Offringa R, Blom RJ, Melief CJ, Kast WM. Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc Natl Acad Sci U S A. 1996;93:7855–7860. doi: 10.1073/pnas.93.15.7855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Srinivasan M, Domanico SZ, Kaumaya PT, Pierce SK. Peptides of 23 residues or greater are required to stimulate a high affinity class II-restricted T cell response. Eur J Immunol. 1993;23:1011–1016. doi: 10.1002/eji.1830230504. [DOI] [PubMed] [Google Scholar]
  • 83.Zwaveling S, Ferreira Mota SC, Nouta J, et al. Established human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptides. J Immunol. 2002;169:350–358. doi: 10.4049/jimmunol.169.1.350. [DOI] [PubMed] [Google Scholar]
  • 84.Sercarz EE, Maverakis E. Mhc-guided processing: binding of large antigen fragments. Nat Rev Immunol. 2003;3:621–629. doi: 10.1038/nri1149. [DOI] [PubMed] [Google Scholar]
  • 85.Bijker MS, van den Eeden SJ, Franken KL, Melief CJ, van der Burg SH, Offringa R. Superior induction of anti-tumor CTL immunity by extended peptide vaccines involves prolonged, DC-focused antigen presentation. Eur J Immunol. 2008;38:1033–1042. doi: 10.1002/eji.200737995. [DOI] [PubMed] [Google Scholar]
  • 86.Janssen EM, Droin NM, Lemmens EE, et al. CD4+ T cell help helps control CD8+ T-cell memory via TRAIL-mediated activation-induced cell death. Nature. 2010;434:88–93. doi: 10.1038/nature03337. [DOI] [PubMed] [Google Scholar]
  • 87.Kenter GG, Welters MJ, Valentijn AR, et al. Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia. N Engl J Med. 2009;361:1838–1847. doi: 10.1056/NEJMoa0810097. [DOI] [PubMed] [Google Scholar]
  • 88.National Cancer Institute Immunotherapy Agent Workshop: 2011
  • 89.Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–3175. doi: 10.1200/JCO.2009.26.7609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sznol M, Powderly JD, Smith DC, et al. Safety and antitumor activity of biweekly MDX-1106 (Anti-PD-1, BMS-936558/ONO-4538) in patients with advanced refractory malignancies. J Clin Oncol. 2010;28:2506. abstract # 2506. [Google Scholar]
  • 91.Fife BT, Pauken KE. The role of the PD-1 pathway in autoimmunity and peripheral tolerance. Annals of the New York Academy of Science. 2011;1217:45–59. doi: 10.1111/j.1749-6632.2010.05919.x. [DOI] [PubMed] [Google Scholar]
  • 92.Barber DL, Wherry EJ, Masopust D, et al. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature. 2006;439:682–687. doi: 10.1038/nature04444. [DOI] [PubMed] [Google Scholar]
  • 93.Bennett F, Luxenberg D, Ling V, et al. Program death-1 engagement upon TCR activation has distinct effects on costimulation and cytokine-driven proliferation: attenuation of ICOS, IL-4, and IL-21, but not CD28, IL-7, and IL-15 responses. J Immunol. 2003;170:711–718. doi: 10.4049/jimmunol.170.2.711. [DOI] [PubMed] [Google Scholar]
  • 94.Fife BT, Pauken KE, Eagar TN, et al. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol. 2009;10:1185–1192. doi: 10.1038/ni.1790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hirata S, Senju S, Matsuyoshi H, Fukuma D, Uemura Y, Nishimura Y. Prevention of experimental autoimmune encephalomyelitis by transfer of embryonic stem cell-derived dendritic cells expressing myelin oligodendrocyte glycoprotein peptide along with TRAIL or programmed death-1 ligand. J Immunol. 2005;174:1888–1897. doi: 10.4049/jimmunol.174.4.1888. [DOI] [PubMed] [Google Scholar]
  • 96.Fife BT, Guleria I, Gubbels BM, et al. Insulin-induced remission in new-onset NOD mice is maintained by the PD-1-PD-L1 pathway. J Exp Med. 2006;203:2737–2747. doi: 10.1084/jem.20061577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Higano CS, Schellhammmer PF, Small EJ, 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;115:3670–3679. doi: 10.1002/cncr.24429. [DOI] [PubMed] [Google Scholar]
  • 98.Schwartzentruber DJ, Lawson D, Richards J, et al. A phase III multi-institutional randomized study of immunization with the gp100:209-217(210M) peptide followed by high-dose IL-2 compared with high-dose IL-2 alone in patients with metastatic melanoma. J Clin Oncol. 2009;27 Meeting Abstracts. abstract CRA9011. [Google Scholar]
  • 99.Schwartzentruber DJ, Lawson DH, Richards JM, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 2011;364:2119–2127. doi: 10.1056/NEJMoa1012863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.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;15:7029–7035. doi: 10.1158/1078-0432.CCR-09-1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lawson DH, Lee SJ, Tarhini AA, Margolin KA, Ernstoff MS, Kirkwood JM. E4697: Phase III cooperative group study of yeast-derived granulocyte macrophage colony-stimulating factor (GM-CSF) versus placebo as adjuvant treatment of patients with completely resected stage III-IV melanoma. J Clin Oncol. 2010;28 doi: 10.1200/JCO.2015.62.0500. abstract. abstract 8504. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES