Composite peptide‑based vaccines for cancer immunotherapy (Review)

  • Authors:
    • Jie Yang
    • Qing Zhang
    • Ke Li
    • Hong Yin
    • Jun‑Nian Zheng
  • View Affiliations

  • Published online on: November 12, 2014     https://doi.org/10.3892/ijmm.2014.2000
  • Pages: 17-23
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The use of peptide‑based vaccines as therapeutics aims to elicit immune responses through antigenic epitopes derived from tumor antigens. Peptide‑based vaccines are easily synthesized and chemically stable entities, and of note, they are absent of oncogenic potential. However, their application is more complicated as the success of an effective peptide‑based vaccine is determined by numerous parameters. The success thus far has been limited by the choice of tumor antigenic peptides, poor immunogenicity and incorporation of strategies to reverse cancer‑mediated immune suppression. In the present review, an overview of the mechanisms of peptide‑based vaccines is provided and antigenic peptides are categorized with respect to their tissue distribution in order to determine their usefulness as targets. Furthermore, certain approaches are proposed that induce and maintain T cells for immunotherapy. The recent progress indicates that peptide‑based vaccines are preferential for targeted therapy in cancer patients.

1. Introduction

Cancer, particularly malignant tumors, is the leading cause of mortality in developed countries and the second leading cause of mortality in developing countries (1). For decades, advancements have been made in traditional cancer treatment regimens, chiefly surgery, chemotherapy and radiation. Although these treatment regimens have had certain success, they are not entirely adequate or optimal for tumors that have migrated to areas that are inaccessible by surgery or chemotherapy/radiation is not permissible. Therefore, immunotherapy, which may produce fewer side-effects and prevent metastasis in comparison to traditional treatments, has become of increasing interest in the area of cancer treatment.

In the field of immunotherapy, increasing attention has been focused on the use of cancer vaccines that activate T cells to treat growing tumors (2). The development of peptide-based vaccines has taken >20 years. A vaccine specific for tumor antigens may have wide application and utility in the prevention of the recurrence in numerous different malignancies. In cancer patients, the body masks tumor antigens by the addition of carbohydrate moieties to avoid an uncontrolled autoimmune response; however, in the process, the elimination of threatening tumor cells is also impeded (3). Therefore, the development of peptide-based vaccines that directly stimulate the immune system would be highly significant.

Peptides, which are composed of several amino acids and are absent of oncogenic potential, are antigenic epitopes derived from tumor-associated antigens (TAA) or tumor-specific antigens (TSA) (3,4). Peptide-based vaccines are designed to elicit specific T cells against antigens selectively expressed by tumor cells (5). In comparison to traditional treatment, peptide-based vaccines significantly prolonged the overall survival rate and spare normal tissue due to its low toxic effect (68). Evidently, there is a large number of cancer patients requiring the development of novel approaches for immunotherapy.

2. Mechanism of antitumor immunity by peptide-based vaccines

The application of peptide-based vaccines is based on three distinct steps to create a specific antitumor immune response. To initiate immunity, dentritic cells (DCs), which are taken up exogenously as part of a therapeutic vaccine (9), differentiate into immunogenic mature DCs (10). DCs enable the presentation of peptides on major histocompatibility complex (MHC) class I and II molecules (Fig. 1). Previously, the majority of peptide-based vaccines target MHC class I peptides to stimulate cytotoxic T lymphocyte (CTL) responses. MHC class I binds with peptides that are ~8–12 amino acids in length (Fig. 1A) (11). Peptides are transported into the endoplasmic reticulum (ER) by a transporter associated with antigen presentation (12). Subsequently, peptide-MHC class I complexes go through the Golgi and are delivered to the cell surface for recognition by cluster of differentiation 8+ (CD8+) CTLs (13). Concurrently, a small proportion of MHC class II-restricted peptides stimulate CD4+ T helper cells. MHC class II-restricted peptides are generally 12–20 amino acids in length (Fig. 1B) (3). Extracellular peptides are taken up by antigen-presenting cells and placed into phagosomes, which fuse with lysosomes to form phagolysosomes (14). MHC class II assembled in the ER associates with the invariant chain (Ii), and the Ii-MHC class II complex is transported to phagolysosome and is known as the MHC class II compartment (MIIC). In the MIIC, Ii is degraded, leaving a residual class II-associated Ii peptide in the peptide-binding groove (12). MHC class II requires human leukocyte antigen (HLA)-DM (H2-DM in mice) to completely expose the peptide-binding groove, binding with a specific peptide (15). Peptide-MHC class II complexes are delivered to the cell surface for recognition by CD4+ T helper cells.

Subsequently, in lymphoid organs the peptide-loaded DCs trigger specific T-cell responses (10). These T cells, such as CD4+ T helper cells and CD8+ T cells, show potent cytotoxic effects by two signals (16). One signal is from the T cell receptor (TCR) interacting with peptide-MHC complexes on the DCs (17). The other signal is from the interaction of DC surface receptor CD80/CD86 with T-cell co-stimulatory molecule, CD28 (17,18).

Finally, specific T cells must migrate to the tumor microenvironment to cause the cytotoxic response. Considerable knowledge has been obtained on CD8+ CTLs that have been identified as the most powerful effector cells (19). CD4+ T helper 1 cells (Th1) secrete several cytokines, such as IFN-γ, TNF-α and interleukin-2 (IL-2) (20,21). These cytokines exert direct antitumor immunity and antiangiogenic effects (22). Notably, specific CD4+ T helper cells have been found to enhance CD8+ CTL recruitment and proliferation (21).

3. Peptide selection

During the past two decades, a majority of antigenic peptides have been discovered. In principle, there are two types of tumor antigens. One type is from TSAs, which are expressed exclusively by tumors. TSAs generally arise from viral infections or genetic mutations (23). The second more common type is derived from TAAs. TAAs are found on malignant and normal cells, but in a significantly higher number in the former.

From TSAs

Peptide-based vaccines targeting viral oncogene products are ideal candidates to elicit strong immune responses without generating autoimmunity. As these viral oncoproteins are not expressed in normal cells and their expression is required to maintain the malignant phenotype, the viral protein is considered as a potential target for cancer immunotherapy. Recently, a variety of viral oncoproteins in virus-associated cancers have been used as vaccines to induce T-cell responses. For instance, injection of human papillomavirus type 16 E5 peptide + CpG resulted in strong T-cell immunity and inhibited tumor growth, whilst prolonging the survival time in animals with cervical cancer (24). Similarly, two recombinant Epstein-Barr virus antigenic peptides, EBNA1 fused with LMP2, boosted T-cell immunity and was proven to be safe and had low immunogenicity in the phase I clinical trial for nasopharyngeal carcinoma (7).

The antigens occurring in a number of different proteins expressed by tumor cells are the result of genetic mutations. A controversy exists over the idea that a single human tumor, as in a mouse system, can express multiple mutated antigens and generate new ones during progression, thereby making their characterization even more complex. However, in the last few years, the situation has slowly changed. Several studies (6979) have described such antigens as peptide epitopes recognized by T cells in combination with MHC class I and II in human tumors, such as melanoma, non-small cell lung cancer, renal cancer and head/neck cancer (Table I). The presumed advantages of mutated antigens are based on the potential to be recognized as non-self by the immune system and their potential resistance to negative selection if the mutated protein participates in cell survival (25).

Table I

Antigens from genetic mutation recognized by HLA class I and class II restricted T cells.

Table I

Antigens from genetic mutation recognized by HLA class I and class II restricted T cells.

AntigenTumorRefs.
HLA class I-restricted
 β-cateninMelanoma(69)
 CDK-4Melanoma(70)
 MART-2Melanoma(71)
 MUM-1/2Melanoma(72)
 MUM-3Melanoma(73)
 HSP70-2Renal cancer(74)
 Caspase-8Head/neck cancer(75)
 p21/rasPancreatic, colorectal, lung cancer(76)
HLA class II-restricted
 p53Head/neck cancer(77)
 TPIMelanoma(78)
 CDC27Melanoma(79)

[i] HLA, human leukocyte antigen; CDK, cyclin-dependent kinase; MART, melanoma antigen recognized by T cells; MUM, melanoma ubiquitously mutated; HSP, heat-shock protein; TPI, triose-phosphate isomerase; CDC, cell division cycle.

From TAAs

TAAs can be divided into four major categories: i) Differentiation antigens; ii) cancer/testis antigens shared by germ and tumor cells; iii) overexpressed antigens, such as normal proteins whose expression is upregulated in tumor cells; and iv) universal tumor antigens.

Differentiation-antigens

These antigens are expressed by the normal tissue and tumor originating from these tissues. The majority of these antigens have been applied to treat melanoma, such as melanoma Ag recognized by T cells (MART-1)/Melan A, gp100, tyrosinase, tyrosinase-related protein (TRP-1) and TRP-2. Gp100, which were initially identified, were reported as non-mutated differentiation antigens expressed by a melanocytic lineage, including normal melanocytes, pigmented retinal cells and melanomas, but not in other normal tissues or non-melanoma tumors. In a current clinical trial with tumor-free lymph nodes of stage I to III melanoma patients, immunization with modified gp100209–2M peptide without co-administration of CD4+ cell-restricted antigens induced the effective expansion of tumor-reactive memory CD8+ T cells with high proliferation potential (26). In another clinical study, the response rate was higher and the progression-free survival rate was longer with gp100:209–217 (210 M) peptide vaccine plus IL-2 compared to IL-2 alone in patients with metastatic melanoma (27). These studies demonstrate that differentiation proteins may be suitable targets for immunotherapy.

Cancer/testis (CT) antigens

Expression of these antigens is restricted to human germ cells within the testis and trophoblasts, and is also expressed on a variety of types of human cancers. The antigens in testis do not induce an immune response, as testis cells do not express MHC class I. Since CT antigens are not expressed in normal tissue, these antigens may be potentially useful targets for tumor-specific immunotherapy. More than 40 antigens have been identified as CT antigens, including melanoma antigen (MAGE), B melanoma antigen, New York oesophageal squamous cell carcinoma 1 (NY-ESO-1) and G antigen 1. The first CT antigen was discovered from a patient with melanoma who was identified as having cytotoxic T cells that recognized autologous tumor cells (28). Through DNA-cloning methodology, the gene encoding the tumor antigen MZ2-E was cloned and was termed MAGE1. Fujie et al (29) proposed that through using the MAGE-1-encoded peptide it was possible to immunize an increased number of patients by means of such peptide-based immunotherapeutic approaches to MAGE-1-positive malignant tumors. Thus far, NY-ESO-1 is the most studied due to its strong capacity to induce a tumor-specific immune response (30). Previously, a completed clinical study using co-administration of CpG 7909 and Montanide ISA-51 with peptide NY-ESO-1 p157–165 showed the vaccine to be capable of inducing CTLs, resulting in an extended survival time in the majority of vaccinated patients (31). Currently, a number of clinical trials treating various cancers are being performed using antigenic peptide NY-ESO-1 combined with differing adjuvants.

Overexpressed-antigens

In healthy tissues, these antigens are expressed at low levels on the surface of normal cells. Conversely, in the majority of cancers these antigens are overexpressed but with no preferential expression on certain tumor types, involving human epidermal growth factor receptor-2 (HER-2), human mucin 1 (MUC1) and cyclin B1. In humans, the HER-2 protein is expressed during fetal development but is weakly detectable in the epithelial cells of a number of normal tissues in adults. Overexpression of the HER-2 protein has been identified in numerous types of human cancers, such as breast, ovarian and non-small-cell lung cancer. Immunizing patients with peptides derived from HER-2/neu protein admixed with granulocyte-macrophage colony-stimulating factor (GM-CSF) have been indicated to result in the generation of T-cell immunity specific for the HER-2/neu (32). In addition, MUC1 has been studied as a target for immunotherapy following a long developmental phase. Transmembrane glycoprotein MUC1 is expressed on the apical surface of polarized epithelial cells. However, in the majority of epithelial malignancies, MUC1 is overexpressed and loses its polarity of expression (33). In pre-clinical studies using primates, MUC1 tandem repeat peptide administered with LeIF elicited T helper cells and CTL responses (34). This study showed the peptide-based vaccine to be safe and to possibly be a vaccine that induces MUC1-specific immune responses in patients with cancer.

Universal tumor antigens

Over the past decade, numerous TAAs have been reported. However, for any particular TAA, expression is restricted to several tumor types. To circumvent this, a new category of TAAs, known as ‘universal tumor antigens,’ has been described. Such universal tumor antigens are highly expressed in tumor cells of different tissue origins with minimal expression in normal counterparts. Survivin and telomerase have been reported to be suitable as target universal tumor antigens for active immunization of cancer patients. In a previous study, telomerase was expressed in 85–90% of cancer patients and was an attractive universal tumor antigen (35). Telomerase helps to mediate functional telomeres, maintaining at the end of chromosomes, and prevent cells from going into senescence, particularly in cancer cells. The majority of human cells do not express telomerase activity, but the majority of human tumors exhibit strong activity (36). In 2006, Brunsvig et al (37) conducted a phase I/II clinical study in patients with non-small cell lung cancer (NSCLC), and the results demonstrated that intradermal injections of GV1001 (hTERT: 611–626) was immunogenic, safe and induced strong specific immune responses. Based on these initial encouraging results, the study reported further clinical studies of GV1001 in NSCLC patients. Vaccination with GV1001 was indicated to exhibit low toxicity, induced considerable immune response rate and established durable T-cell memory (38).

4. Strategies to induce and maintain T cells

Peptide-based vaccines present certain objective limitations. Free peptides have poor immunogenicity or no tertiary structure, and thus are rapidly degraded by tissue and serum peptidases prior to being loaded onto DCs. Recent studies indicate that optimal strategies to induce and maintain T cells include adjuvants, cytokines, HLA class II-restricted helper epitopes, immune-modulating antibodies and low-affinity peptides combined with high-affinity peptides, which are described in the following.

Adjuvants

Peptide-based vaccines require additional adjuvant to elicit efficient immunological response. Conjugates of peptides with heat shock proteins (HSPs) (39,40) or ligands of toll-like receptors (TLRs) (4143) have been applied to a broad range of vaccines as adjuvants to enhance the immunogenicity of peptides. In 2000, the study by Cho et al (44) reported that HSP65 fusion proteins stimulated DCs to increase expression of MHC (class I and II) and co-stimulatory (B7.2) molecules. This study suggested a mechanism in which the HSP fusion proteins induced CTLs to peptides without requiring exogenous adjuvants or the participation of CD4+ T cells (44). However, TLRs may improve vaccination efficacy through activating DC maturation, thereby upregulating the expression of MHC molecules and enhancing antigen uptake. Khan et al (45) reported that TLR ligand-peptide conjugates improved intracellular trafficking and processing pathways, triggering optimal antigen presentation and T cells priming.

Cytokines

Cytokines, such as GM-CSF and IL-12, are used as an adjuvant in vaccines. GM-CSF increases the number of immature DCs and migration, and it induces MHC class II expression and activation by macrophages. In melanoma patients, subcutaneous injection with GM-CSF modestly increased the immune response against peptide vaccines (46). The cytokine IL-12 augments antitumor immunity through promoting Th1 cell differentiation and stimulating the production of IFN-γ from CD4+ Th1 and CD8+ T cells (47). In preclinical and clinical studies, a significant proportion of patients with resected melanomas experienced an improved performance of the peptide vaccines when it was combined with properly dosed IL-12 therapy (48).

HLA class II-restricted helper epitopes

HLA class II-restricted helper epitopes enhance specific CTLs and generate T-cell memory (4951). CD4+ T helper cells can activate DCs to enhance antigen presentation, resulting in the secretion of IL-2 and other cytokines from DCs that may help to direct the immune response. Furthermore, cytokines secreted by Th1, such as IL-2 and IFN-γ, are required for the generation of CTLs, as well as in promoting CD8+ memory T-cell development. IL-2 induces CTL activation and proliferation (52). Simultaneously, IFN-γ controls the migration of CTLs (53). IL-2 is essential for programming the ability of CD8+ memory T cells to re-expand upon secondary infection in vivo (52,54). Knutson et al (55) evaluated whether active immunization with HER-2/neu helper peptides generated CD4+ and CD8+ T-cell responses in patients. Following vaccination, HER-2/neu-specific CD8+ T-cells increased in the majority of patients. Additionally, the specific T cells were able to lyse tumors and the immunity was long-lived. Subsequently, Gritzapis et al (56) indicated that in comparison to Her-2 (435–443) CTL peptide alone, mice vaccinated with Her-2 (435–443) plus Her-2 (776–790) exhibited longer lasting antitumor responses and induced memory immunity. Thus, there is a rationale for induction of CD4+ T cells with peptide-based vaccines, either in combination with stimulation of CD8+ T cells or on their own.

Immune-modulating antibodies

Combining peptide-based vaccines with immune-modulating antibodies may be a novel strategy to overcome immune suppression (17,57,58). Several antibody therapies that are either agonistic or inhibit receptors have shown benefits in cancer treatment (5961). Specific recognition by T cells is a two-step process. In addition to the interaction of TCRs with MHC-peptide complexes as the first signal for T cell recognition, a second signal co-stimulates the receptors that determine whether the T cell will become activated or anergic. These surface co-stimulatory receptors transmit agonistic or inhibitory signals through engagement of specific ligands. The co-stimulatory activity of these receptors can be mimicked by antibodies modulating T-cell proliferation, cytokine secretion and cytolysis (62). There are a number of known agonistic receptors, including 4–1BB (CD137), OX40, CD27, GITR and CD28 (6264). The immune response against a peptide vaccine combined with the systemic delivery of anti-4–1BB antibodies resulted in considerably improved antitumor therapeutic activity through CTLs, NK cells, neutrophils and IFN-γ (65). Receptors that serve as targets for inhibitory antibodies include CTLA-4, PD-1 and BTLA. In one study, a combination of peptide-based vaccines with blocking co-inhibitory receptor signaling, resulted in antibody blockage of the co-inhibitory receptors, CTLA-4 and PD-1, decreased T cell anergy and allowed specific T cells to carry out their effector function (66).

Low-affinity peptides combined with high-affinity peptides

Peptides that bind to MHC molecules with high affinity usually induce high-avidity T cells. Administration of high-affinity peptides of exogenous antigens (such as viruses) is necessary for their elimination. However, if the antigen is autologous, high-affinity peptides possibly lead to tolerance. Thus, for a peptide-based vaccine the most appropriate peptides may be low-to-medium-affinity peptides (67). However, low-affinity peptides are difficult to identify. In order to overcome this difficulty, attempts have been made to raise the affinity of peptides for the MHC through binding with high-affinity peptides. For example, synthetic peptides, such as TERT578–592 combined with two peptides derived from TERT, as high-affinity forms strongly stimulated antitumor immune responses (22). In a parallel study, Disis et al (68) synthesized four peptides from HER-2/neu protein and two were shown to be avid binders to HLA-A2.1, whereas the other two may be shown to elicit peptide-specific CTL in vivo.

5. Conclusions

The significant advantage of peptide-based vaccines is that they are easily synthesized, chemically stable entities and notably, are absent of oncogenic potential. Antigenic peptides offer a simple and flexible way to deal with the complexity of tumor antigens through bypassing the requirements for antigen processing.

As discussed previously, several promising preclinical and clinical studies for peptide-based vaccines are currently being carried out. Thus far however, there is no peptide-based vaccine currently available on the market. There are drawbacks that may hinder the peptide vaccine therapy. First, tumor cells can downregulate MHC molecules or disable other components of the antigen processing machinery. Second, peptides are HLA class I and II restricted and, consequently, restrict the treatment of patients in the clinical trials. The majority of previous cancer vaccines target MHC class I-restricted peptides to stimulate CTL responses. However, the clinical effect of CTL peptide-based vaccines remains modest. Third, tumor cells may upregulate surface ligands (such as PD-L1), which engage inhibitory receptors on the surfaces of activated T cells (PD-1), to mediate T-cell anergy. These drawbacks emphasize the requirement for significant changes in the applications of peptide-based vaccines. Various combinational approaches have been carried out to raise the efficacy of peptide-based therapies.

In conclusion, there is evidence that peptide-based vaccines have increased responses and prolonged survival rates in patients with cancer. Firstly, designing a peptide-based vaccine for cancer immunotherapy is challenging, involving the selection of appropriate antigenic peptides. Strategies to increase the effects to generate antitumor CD4+ cells that recognize MHC class II-restricted peptides may have impact due to the importance of CD4+ cells in enhancing activation and survival of CD8+ effector cells, as well as generating CD8+ memory T cells. Synthetic peptides that have antigenic low-affinity combined with high-affinity peptides raise the affinity of the peptides for the MHC and may significantly enhance antitumor response. Furthermore, increasing numbers of peptide-based vaccines with the co-administration of adjuvants, cytokines or immunomodulatory antibodies have been shown to induce and maintain immune responses. Finally, further studies are required to focus on the synergy of peptide vaccination with chemotherapy, involving larger studies providing evidence to evaluate the curative effects ex vivo and in vivo.

Acknowledgements

The present study was supported by grants from the Natural Science Foundation of China (no. 81301946, 81301947 and 81202015), and Nature Science Foundation of Jiangsu Province (no. BK2012146).

References

1 

Yong X, Xiao YF, Luo G, et al: Strategies for enhancing vaccine-induced CTl antitumor immune responses. J Biomed Biotechnol. 2012:6050452012. View Article : Google Scholar : PubMed/NCBI

2 

Rosenberg SA, Yang JC and Restifo NP: Cancer immunotherapy: moving beyond current vaccines. Nat Med. 10:909–915. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Lazoura E and Apostolopoulos V: Rational peptide-based vaccine design for cancer immunotherapeutic applications. Curr Med Chem. 12:629–639. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Buhrman JD, Jordan KR, Munson DJ, Moore BL, Kappler JW and Slansky JE: Improving antigenic peptide vaccines for cancer immunotherapy using a dominant tumor-specific T cell receptor. J Biol Chem. 288:33213–33225. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Milani A, Sangiolo D, Montemurro F, Aglietta M and Valabrega G: Active immunotherapy in HER2 overexpressing breast cancer: current status and future perspectives. Ann Oncol. 24:1740–1748. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Tanaka T, Kitamura H, Inoue R, et al: Potential survival benefit of anti-apoptosis protein: survivin-derived peptide vaccine with and without interferon alpha therapy for patients with advanced or recurrent urothelial cancer - results from phase I clinical trials. Clin Dev Immunol. 2013:262967l2013. View Article : Google Scholar

7 

Hui EP, Taylor GS, Jia H, et al: Phase I trial of recombinant modified vaccinia ankara encoding epstein-BARR viral tumor antigens in nasopharyngeal carcinoma patients. Cancer Res. 73:1676–1688. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Asahara S, Takeda K, Yamao K, Maguchi H and Yamaue H: Phase I/II clinical trial using HLA-A24-restricted peptide vaccine derived from KIF20A for patients with advanced pancreatic cancer. J Transl Med. 11:2912013. View Article : Google Scholar : PubMed/NCBI

9 

Basha G, Lizée G, Reinicke AT, et al: MHC class I endosomal and lysosomal trafficking coincides with exogenous antigen loading in dendritic cells. PLoS One. 3:e32472008. View Article : Google Scholar : PubMed/NCBI

10 

Mellman I, Coukos G and Dranoff G: Cancer immunotherapy comes of age. Nature. 480:480–489. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Flutter B and Gao B: MHC class I antigen presentation-recently trimmed and well presented. Cell Mol Immunol. 1:22–30. 2004.

12 

Neefjes J, Jongsma MLM, Paul P and Bakke O: Towards a systems understanding of MHC class I and MHC class II antigen presentation. Nat Rev Immunol. 11:823–836. 2011.PubMed/NCBI

13 

Van Kaer L: Major histocompatibility complex class I-restricted antigen processing and presentation. Tissue Antigens. 60:1–9. 2002. View Article : Google Scholar : PubMed/NCBI

14 

Vyas JM, Van der Veen AG and Ploegh HL: The known unknowns of antigen processing and presentation. Nat Rev Immunol. 8:607–618. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Pieters J: MHC class II-restricted antigen processing and presentation. Adv Immunol. 75:159–208. 2000. View Article : Google Scholar : PubMed/NCBI

16 

Callan MF, Fazou C, Yang H, et al: CD8(+) T-cell selection, function, and death in the primary immune response in vivo. J Clin Invest. 106:1251–1261. 2000. View Article : Google Scholar : PubMed/NCBI

17 

Vanneman M and Dranoff G: Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer. 12:237–251. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Shashidharamurthy R, Bozeman EN, Patel J, Kaur R, Meganathan J and Selvaraj P: Immunotherapeutic strategies for cancer treatment: a novel protein transfer approach for cancer vaccine development. Med Res Rev. 32:1197–1219. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Vesely MD, Kershaw MH, Schreiber RD and Smyth MJ: Natural innate and adaptive immunity to cancer. Annu Rev Immunol. 29:235–271. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Kennedy R and Celis E: Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol Rev. 222:129–144. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Bos R and Sherman LA: CD4+ T-cell help in the tumor milieu is required for recruitment and cytolytic function of CD8+ T lymphocytes. Cancer Res. 70:8368–8377. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Dosset M, Godet Y, Vauchy C, et al: Universal cancer peptide-based therapeutic vaccine breaks tolerance against telomerase and eradicates established tumor. Clin Cancer Res. 18:6284–6295. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Wei HJ, Wu AT, Hsu CH, et al: The development of a novel cancer immunotherapeutic platform using tumor-targeting mesenchymal stem cells and a protein vaccine. Mol Ther. 19:2249–2257. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Liao SJ, Deng DR, Zeng D, et al: HPV16 E5 peptide vaccine in treatment of cervical cancer in vitro and in vivo. J Huazhong Univ Sci Technolog Med Sci. 33:735–742. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Palucka K, Banchereau J and Mellman I: Designing vaccines based on biology of human dendritic cell subsets. Immunity. 33:464–478. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Walker EB, Miller W, Haley D, Floyd K, Curti B and Urba WJ: Characterization of the class I-restricted gp100 melanoma peptide-stimulated primary immune response in tumor-free vaccine-draining lymph nodes and peripheral blood. Clin Cancer Res. 15:2541–2551. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Schwartzentruber DJ, Lawson DH, Richards JM, et al: gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med. 364:2119–2127. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Knuth A, Wölfel T, Klehmann E, Boon T and Meyer zum Buschenfelde KH: Cytolytic T-cell clones against an autologous human melanoma: specificity study and definition of three antigens by immunoselection. Proc Natl Acad Sci USA. 86:2804–2808. 1989. View Article : Google Scholar : PubMed/NCBI

29 

Fujie T, Tahara K, Tanaka F, Mori M, Takesako K and Akiyoshi T: A MAGE-1-encoded HLA-A24-binding synthetic peptide induces specific anti-tumor cytotoxic T lymphocytes. Int J Cancer. 80:169–172. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Simpson AJ, Caballero OL, Jungbluth A, Chen YT and Old LJ: Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer. 5:615–625. 2005. View Article : Google Scholar : PubMed/NCBI

31 

Karbach J, Gnjatic S, Bender A, et al: Tumor-reactive CD8+ T-cell responses after vaccination with NY-ESO-1 peptide, CpG 7909 and montanide ISA-51: association with survival. Int J Cancer. 126:909–918. 2010.

32 

Disis ML, Gooley TA, Rinn K, et al: Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol. 20:2624–2632. 2002. View Article : Google Scholar : PubMed/NCBI

33 

Sangha R and Butts C: L-BLP25: a peptide vaccine strategy in non small cell lung cancer. Clin Cancer Res. 13:s4652–s4654. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Barratt-Boyes SM, Vlad A and Finn OJ: Immunization of chimpanzees with tumor antigen MUC1 mucin tandem repeat peptide elicits both helper and cytotoxic T-cell responses. Clin Cancer Res. 5:1918–1924. 1999.PubMed/NCBI

35 

Bernhardt SL, Gjertsen MK, Trachsel S, et al: Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer. 95:1474–1482. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Kim NW, Piatyszek MA, Prowse KR, et al: Specific association of human telomerase activity with immortal cells and cancer. Science. 266:2011–2015. 1994. View Article : Google Scholar : PubMed/NCBI

37 

Brunsvig PF, Aamdal S, Gjertsen MK, et al: Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer. Cancer Immunol Immunother. 55:1553–1564. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Brunsvig PF, Kyte JA, Kersten C, et al: Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial. Clin Cancer Res. 17:6847–6857. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Ding Z, Ou R, Ni B, Tang J and Xu Y: Cytolytic activity of the human papillomavirus type 16 E711-20 epitope-specific cytotoxic t lymphocyte is enhanced by heat shock protein 110 in HLA-A*0201 transgenic mice. Clin Vaccine Immunol. 20:1027–1033. 2013. View Article : Google Scholar : PubMed/NCBI

40 

Yang J, Zhang Y, Wang H, et al: Vaccination with the repeat β-hCG C-terminal peptide carried by heat shock protein-65 (HSP65) for inducing antitumor effects. Tumor Biol. 33:1777–1784. 2012. View Article : Google Scholar

41 

Koido S, Homma S, Okamoto M, et al: Combined TLR2/4-activated dendritic/tumor cell fusions induce augmented cytotoxic T lymphocytes. PLoS One. 8:e592802013. View Article : Google Scholar : PubMed/NCBI

42 

Muraoka D, Kato T, Wang L, et al: Peptide vaccine induces enhanced tumor growth associated with apoptosis induction in CD8+ T cells. J Immunol. 185:3768–3776. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Speiser DE, Liénard 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. 115:739–746. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Cho BK, Palliser D, Guillen E, et al: A proposed mechanism for the induction of cytotoxic T lymphocyte production by heat shock fusion proteins. Immunity. 12:263–272. 2000. View Article : Google Scholar

45 

Khan S, Bijker MS, Weterings JJ, et al: Distinct uptake mechanisms but similar intracellular processing of two different toll-like receptor ligand-peptide conjugates in dendritic cells. J Biol Chem. 282:21145–21159. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Weber J, Sondak VK, Scotland R, et al: Granulocyte- macrophage-colony-stimulating factor added to a multipeptide vaccine for resected stage II melanoma. Cancer. 97:186–200. 2003. View Article : Google Scholar

47 

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. 13:215–222. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Lee P, Wang F, Kuniyoshi J, et al: Effects of interleukin-12 on the immune response to a multipeptide vaccine for resected metastatic melanoma. J Clin Oncol. 19:3836–3847. 2001.PubMed/NCBI

49 

Izumoto S: Peptide vaccine. Adv Exp Med Biol. 746:166–177. 2012. View Article : Google Scholar : PubMed/NCBI

50 

May RJ, Dao T, Pinilla-Ibarz J, et al: Peptide epitopes from the wilms’ tumor 1 oncoprotein stimulate CD4+ and CD8+ T cells that recognize and kill human malignant mesothelioma tumor cells. Clin Cancer Res. 13:4547–4555. 2007. View Article : Google Scholar : PubMed/NCBI

51 

Fujiki F, Oka Y, Tsuboi A, et al: Identification and characterization of a WT1 (Wilms Tumor Gene) protein-derived HLA-DRB1*0405-restricted 16-mer helper peptide that promotes the induction and activation of WT1-specific cytotoxic T lymphocytes. J Immunother. 30:282–293. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Pipkin ME, Sacks JA, Cruz-Guilloty F, Lichtenheld MG, Bevan MJ and Rao A: Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity. 32:79–90. 2010. View Article : Google Scholar : PubMed/NCBI

53 

Nakanishi Y, Lu B, Gerard C and Iwasaki A: CD8+ T lymphocyte mobilization to virus-infected tissue requires CD4+ T-cell help. Nature. 462:510–513. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Williams MA, Tyznik AJ and Bevan MJ: Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature. 441:890–893. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Knutson KL, Schiffman K and 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. 107:477–484. 2001. View Article : Google Scholar : PubMed/NCBI

56 

Gritzapis AD, Voutsas IF, Lekka E, Papamichail M and Baxevanis CN: Peptide vaccination breaks tolerance to HER-2/neu by generating vaccine-specific FasL(+) CD4(+) T cells: first evidence for intratumor apoptotic regulatory T cells. Cancer Res. 70:2686–2696. 2010. View Article : Google Scholar : PubMed/NCBI

57 

Wang Y, Wang XY, Subjeck JR, Shrikant PA and Kim HL: Temsirolimus, an mTOR inhibitor, enhances anti-tumour effects of heat shock protein cancer vaccines. Br J Cancer. 104:643–652. 2011. View Article : Google Scholar : PubMed/NCBI

58 

Arens R, van Hall T, van der Burg SH, Ossendorp F and Melief CJM: Prospects of combinatorial synthetic peptide vaccine-based immunotherapy against cancer. Semin Immunol. 25:182–190. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Gray JC, French RR, James S, Al-Shamkhani A, Johnson PW and Glennie MJ: Optimising anti-tumour CD8 T-cell responses using combinations of immunomodulatory antibodies. Eur J Immunol. 38:2499–2511. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Fransen MF, Sluijter M, Morreau H, Arens R and Melief CJ: Local activation of CD8 T cells and systemic tumor eradication without toxicity via slow release and local delivery of agonistic CD40 antibody. Clin Cancer Res. 17:2270–2280. 2011. View Article : Google Scholar : PubMed/NCBI

61 

Ascierto PA, Simeone E, Sznol M, Fu YX and Melero I: Clinical experiences with anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol. 37:508–516. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Croft M: The role of TNF superfamily members in T-cell function and diseases. Nat Rev Immunol. 9:271–285. 2009. View Article : Google Scholar : PubMed/NCBI

63 

Croft M: Co-stimulatory members of the TNFR family: keys to effective T-cell immunity? Nat Rev Immunol. 3:609–620. 2003. View Article : Google Scholar : PubMed/NCBI

64 

Topalian SL, Weiner GJ and Pardoll DM: Cancer immunotherapy comes of age. J Clin Oncol. 29:4828–4836. 2011. View Article : Google Scholar : PubMed/NCBI

65 

Sin JI, Kim H, Ahn E, et al: Combined stimulation of TLR9 and 4.1BB augments Trp2 peptide vaccine-mediated melanoma rejection by increasing Ag-specific CTl activity and infiltration into tumor sites. Cancer Lett. 330:190–199. 2013. View Article : Google Scholar

66 

Curran MA, Montalvo W, Yagita H and Allison JP: PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci USA. 107:4275–4280. 2010. View Article : Google Scholar : PubMed/NCBI

67 

Apostolopoulos V: Peptide-based vaccines for cancer: are we choosing the right peptides? Expert Rev Vaccines. 8:259–260. 2009. View Article : Google Scholar : PubMed/NCBI

68 

Disis ML, Smith JW, Murphy AE, Chen W and Cheever MA: In vitro generation of human cytolytic T-cells specific for peptides derived from the HER-2/neu protooncogene protein. Cancer Res. 54:1071–1076. 1994.PubMed/NCBI

69 

Robbins PF, El-Gamil M, Li YF, et al: A mutated beta-catenin gene encodes a melanoma-specific antigen recognized by tumor infiltrating lymphocytes. J Exp Med. 183:1185–1192. 1996. View Article : Google Scholar : PubMed/NCBI

70 

Wölfel T, Hauer M, Schneider J, et al: A p16INK4a-insensitive CDK4 mutant targeted by cytolytic T lymphocytes in a human melanoma. Science. 269:1281–1284. 1995. View Article : Google Scholar : PubMed/NCBI

71 

Kawakami Y, Wang X, Shofuda T, et al: Isolation of a new melanoma antigen, MART-2, containing a mutated epitope recognized by autologous tumor-infiltrating T lymphocytes. J Immunol. 166:2871–2877. 2001. View Article : Google Scholar : PubMed/NCBI

72 

Coulie PG, Lehmann F, Lethé B, et al: A mutated intron sequence codes for an antigenic peptide recognized by cytolytic T lymphocytes on a human melanoma. Proc Natl Acad Sci USA. 92:7976–7980. 1995. View Article : Google Scholar : PubMed/NCBI

73 

Baurain JF, Colau D, van Baren N, et al: High frequency of autologous anti-melanoma CTL directed against an antigen generated by a point mutation in a new helicase gene. J Immunol. 164:6057–6066. 2000. View Article : Google Scholar : PubMed/NCBI

74 

Gaudin C, Kremer F, Angevin E, Scott V and Triebel F: A hsp70-2 mutation recognized by CTl on a human renal cell carcinoma. J Immunol. 162:1730–1738. 1999.PubMed/NCBI

75 

Mandruzzato S, Brasseur F, Andry G, Boon T and van der Bruggen P: A CASP-8 mutation recognized by cytolytic T lymphocytes on a human head and neck carcinoma. J Exp Med. 186:785–793. 1997. View Article : Google Scholar : PubMed/NCBI

76 

Bristol JA, Schlom J and Abrams SI: Development of a murine mutant ras CD8+ CTL peptide epitope variant that possesses enhanced MHC class I binding and immunogenic properties. J Immunol. 160:2433–2441. 1998.PubMed/NCBI

77 

Couch ME, Ferris RL, Brennan JA, et al: Alteration of cellular and humoral immunity by mutant p53 protein and processed mutant peptide in head and neck cancer. Clin Cancer Res. 13:7199–7206. 2007. View Article : Google Scholar : PubMed/NCBI

78 

Pieper R, Christian RE, Gonzales MI, et al: Biochemical identification of a mutated human melanoma antigen recognized by CD4(+) T cells. J Exp Med. 189:757–766. 1999. View Article : Google Scholar : PubMed/NCBI

79 

Wang RF, Wang X, Atwood AC, Topalian SL and Rosenberg SA: Cloning genes encoding MHC class II-restricted antigens: mutated CDC27 as a tumor antigen. Science. 284:1351–1354. 1999. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2015
Volume 35 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang J, Zhang Q, Li K, Yin H and Zheng JN: Composite peptide‑based vaccines for cancer immunotherapy (Review). Int J Mol Med 35: 17-23, 2015
APA
Yang, J., Zhang, Q., Li, K., Yin, H., & Zheng, J. (2015). Composite peptide‑based vaccines for cancer immunotherapy (Review). International Journal of Molecular Medicine, 35, 17-23. https://doi.org/10.3892/ijmm.2014.2000
MLA
Yang, J., Zhang, Q., Li, K., Yin, H., Zheng, J."Composite peptide‑based vaccines for cancer immunotherapy (Review)". International Journal of Molecular Medicine 35.1 (2015): 17-23.
Chicago
Yang, J., Zhang, Q., Li, K., Yin, H., Zheng, J."Composite peptide‑based vaccines for cancer immunotherapy (Review)". International Journal of Molecular Medicine 35, no. 1 (2015): 17-23. https://doi.org/10.3892/ijmm.2014.2000