Skip to main content
Cancer Immunology, Immunotherapy : CII logoLink to Cancer Immunology, Immunotherapy : CII
. 2005 Oct 27;55(1):85–95. doi: 10.1007/s00262-005-0692-3

Immunogenic HER-2/neu peptides as tumor vaccines

Constantin N Baxevanis 1,, Nectaria N Sotiriadou 1, Angelos D Gritzapis 1, Panagiota A Sotiropoulou 1, Sonia A Perez 1, Nike T Cacoullos 1, Michael Papamichail 1
PMCID: PMC11030617  PMID: 15948002

Abstract

During the last decade, a large number of tumor-associated antigens (TAA) have been identified, which can be recognized by T cells. This has led to renewed interest in the use of active immunization as a modality for the treatment of cancer. HER-2/neu is a 185-KDa receptor-like glycoprotein that is overexpressed by a variety of tumors including breast, ovarian, lung, prostate and colorectal carcinomata. Several immunogenic HER-2/neu peptides recognized by cytotoxic T lymphocytes (CTL) or helper T lymphocytes (TH) have been identified thus far. Patients with HER-2/neu over-expressing cancers exhibit increased frequencies of peripheral blood T cells recognizing immunogenic HER-2/neu peptides. Various protocols for generating T cell-mediated immune responses specific for HER-2/neu peptides have been examined in pre-clinical models or in clinical trials. Vaccination studies in animals utilizing HER-2/neu peptides have been successful in eliminating tumor growth. In humans, however, although immunological responses have been detected against the peptides used for vaccination, no clinical responses have been described. Because HER-2/neu is a self-antigen, functional immune responses against it may be limited through tolerance mechanisms. Therefore, it would be interesting to determine whether abrogation of tolerance to HER-2/neu using appropriate adjuvants and/or peptide analogs may lead to the development of immune responses to HER-2/neu epitopes that can be of relevance to cancer immunotherapy. Vaccine preparations containing mixtures of HER-2/neu peptides and peptide from other tumor-related antigens might also enhance efficacy of therapeutic vaccination.

Keywords: Vaccines, CTL epitopes, TH epitopes, HER-2/neu, Tolerance

Introduction

Active immunization for cancer treatment is based on the existence of tumor-associated antigens (TAA), which are in principle both antigenic and at the same time immunogenic (i.e., they are recognized by and are capable of stimulating the immune system, respectively).

TAA have been classified into several categories including differentiation, tissue-specific, mutated and overexpressed antigens [103]. HER-2/neu, a member of the last category, is a transmembrane glycoprotein and member of the epidermal growth factor receptor family. Amplification and/or overexpression of HER-2/neu have been reported in 20–40% of primary breast cancers and also in ovarian (20–25%), colorectal and pancreatic adenocarcinomas (up to 80–85% for both). HER-2/neu has become an important target for cancer immunotherapy for several reasons. In particular, HER-2/neu is present in high proportions of tumor cells [19] and single tumor cells frequently show an intense expression of this molecule [66] suggesting that anti-HER-2/neu therapy would specifically target most tumor cells. In addition, CTL and IgG specific for HER-2/neu have been detected in 30–50% of patients with breast cancer [45] demonstrating that this molecule is capable of inducing both cellular and humoral immunologic responses in vivo. The fact that HER-2/neu-specific IgG levels are increased in patients with HER-2/neu+ breast cancer, suggests that TH cells can also be sensitized upon recognition of HER-2/neu epitopes. In this review, we shall focus on the identification and application of HER-2/neu peptides as tumor vaccines.

Identification of HLA-restricted cytotoxic T cell epitopes

The identification of HLA-restricted CTL peptides is essential for understanding the role of immunity in various circumstances, including its potential in the treatment of cancer. Specifically, it is instrumental in the development of defined vaccines that stimulate CTL responses as well as for the monitoring of immunotherapies of cancer. The peptide-binding based prediction of CTL epitopes in protein sequences has led to the identification of CTL epitopes on proteins from various sources, also including tumor antigens [44]. CTL epitopes are presented in the context of MHC class I alleles. The peptide-binding groove of HLA molecules contains highly polymorphic allele-specific pockets that accommodate side chains of the anchor residues of the bound peptide [8, 9, 88]. Peptides bound to MHC class I molecules are usually 8–10 amino acids in length, because the peptide-binding groove of the HLA class I molecules is closed at both sites [9] and thus accommodates only short peptides. Each allele has specific peptide-binding motifs, which were defined through the analysis of naturally presented peptides eluted from class I molecules [20, 22]. Each individual HLA class I molecule displays a preference for certain amino acids at the major peptide anchor positions (positions 2 and 9 for most class I alleles) that bind to the binding pockets. Amino acids at other positions in the peptide can significantly contribute to binding by their engagement in secondary pockets [50, 78, 87]. The knowledge of allele-specific peptide-binding motifs has led to the development of peptide-binding prediction algorithms by several groups [16, 67, 72]. Although peptides can bind MHC class I molecules, this does not necessarily mean that they are presented on the cell surface. Proteins are degraded intracellularly in the cytosol by the proteasome complex for presentation by MHC class I molecules. Not all of the putative MHC-binding peptides from a protein are generated in vivo and it is not an easy task to predict which peptides will be naturally processed. Various antigen-presenting cells, such as dendritic cells (DC) and tumor cells, can have different proteasome complexes that may, in turn, generate different peptides from the same protein. It has been shown that some TAA epitopes are generated by tumor cells but not by DC [57]. For these epitopes, peptide-based vaccines may be the only modality for vaccination.

Two approaches have been used to identify immunogenic CTL peptides for the HER-2/neu protein that are naturally processed and presented. First, tumor-specific CTL have been generated using tumor-infiltrating lymphocytes (TIL) and autologous tumor cells. Target cells pulsed with the peptides are then used to stimulate the same CTL and to identify peptide-specific T-cell reactivity [103]. The advantage of this method is the guarantee that peptides identified in this way are naturally processed. The disadvantage, however, is the difficulty in obtaining TIL and sufficient numbers of autologous tumor cells from many tumors.

The second approach to identifying HER-2/neu CTL peptides is “reverse immunology”. In this method, MHC class I-binding epitopes are identified using computer programs that take into account the presence of allele-specific major anchor residues, and specific secondary anchor residues [40, 98]. The corresponding synthetic peptides are then tested for their capacity to induce peptide- and tumor-specific CTL derived from healthy individuals or cancer patients [14, 40, 24, 54, 98].

Breaking tolerance to HER-2 self-protein through active immunization with HER-2/neu CTL peptides

The majority of TAA represent abnormally expressed or overexpressed normal gene products. Therefore, mechanisms responsible for self tolerance dampen immune responses against these antigens. Transgenic mouse model studies revealed that tolerance is capable of deleting reactive high-avidity T cells against the transgene, thereby leading to self-tolerance [30, 59, 60]. However, T cell elimination through tolerance is not absolute, since self-reactive T cells can be isolated from tolerant hosts [31, 39]. Moreover, it has been demonstrated that low-avidity T cells can be activated, expanded and involved in antitumor responses [57, 58]. Multiple immunizations with DC pulsed with two different HER-2/neu CTL peptides restarted tumor growth in HER-2/neu transgenic mice, despite the fact that CD8+ T cells from these mice were of low-avidity for the same peptides [56]. These results underscore the potential role of low-avidity T cells in antitumor immunity and offer an important component for vaccination immunotherapies. Similar results were also obtained in another syngeneic mouse/tumor model where injections of DC pulsed with tyrosinase- or gp100-derived CTL peptides significantly delayed melanoma outgrowth in HLA-A*0201-transgenic mice [61]. In this model, protection was enhanced by the use of peptide ligands containing conservative substitutions that were cross-reactive with the original antigens.

We have established a preclinical tumor model based on HLA-A*0201 transgenic, H-2kb knockout (HHD) mice inoculated with syngeneic ALC tumor cells transfected to co-express human HER-2/neu and HLA-A*0201 (ALC.HER.A2.1). Immunization with murine HER-2/neu peptide spanning residues 435–443 plus GM-CSF only partially protected HHD mice from the growth of ALC.HER.A2.1 tumor cells. However, when mice were repeatedly vaccinated with human HER-2 (435–443), which differs in a single amino acid at position 4 from its murine homologue, protection was dramatically enhanced (manuscript in preparation). These results demonstrate that if immune responses from tolerant hosts are properly primed and effector function is maintained over time, the immune response will contribute to the destruction of the tumor. Further evaluation of the low-avidity T cell repertoire specific for self-HER-2/neu in tolerated hosts will ultimately lead to improvements of the methods for optimization of vaccination immunotherapies against HER-2/neu.

HER-2/neu CTL peptides

Peptide HER-2 (p369-377) was originally identified by Fisk et al. [24] as an immunodominant HLA-A2-binding epitope recognized by tumor-associated lymphocytes of ovarian cancer. The characteristic feature of HER-2 (p369-377) as an immunodominant peptide recognized by effector CTL in the context of HLA-A2 molecules on the surface of HER-2/neu overexpressing tumor cells was shown in our laboratory by two different means: first, we stimulated in vitro ascitic fluid T cells from HLA-A2.1+ patients suffering from HER-2/neu overexpressing ovarian cancer with autologous DC pulsed with membrane peptides (acid cell extracts; ACE) which were extracted from their tumors [5]. After two–three rounds of restimulation in the presence of cytokine combinations, we obtained ACE-specific CTL lines and, subsequently, T-cell clones which recognized synthetic HER-2 (p369-377) expressed on T2 cells. The same HER-2 (p369-377) specific CTL lines and clones also recognized the HER-2/neu overexpressing and HLA-A2.1+ SKOV3.A2 ovarian tumor cell line, demonstrating that this CTL epitope is naturally processed and presented. In the second approach [29], we immunized HHD mice with ACE extracted from HLA-A2.1+, HER-2/neu overexpressing ovarian and breast tumors; splenocytes from vaccinated HHD mice were then restimulated in vitro with syngeneic splenocytes pulsed with ACE and tested in cytotoxicity assays. HHD splenocytes specifically recognizing ACE from five patients with cancer (three patients with breast cancer and two patients with ovarian cancer) efficiently lysed T2 cells pulsed with HER-2 (p369-377) as well as the ovarian SKOV3.A2 and breast MCF-7 (both expressing HER-2/neu and HLA-A2.1) cancer cell lines. These HER-2 (p369-377)-specific HHD CTL, upon adoptive transfer, efficiently protected SCID mice from growth of SKOV3.A2 or MCF-7 induced tumors. Later on, p369-377 was also found to be expressed by several types of HLA-A2+ tumors, including renal cell carcinoma [12], breast carcinoma [48], and melanoma cells [77].

Rongcun et al. [77] were able to generate in vitro T-cell lines and clones from ascitic fluids of HLA-A2+ patients with epithelial ovarian cancer recognizing HER-2 peptides (p435-443), (p665-673), (p689-697), and (p952-960). These peptides are also expressed on a variety of tumor cell lines including ovarian, colon and breast carcinomas, and melanomas [45]. The HER-2 (p689-697) was also found to be recognized by gastric cancer specific CTLs [51]. More recently, we have found that besides classical CTL, p369-377, p435-443, p665-673, and p689-697 can elicit NKT cells specifically recognizing autologous HLA-A2+ HER-2/neu+ ovarian tumors [5]. HER-2 (p435-443) is also an immunodominant CTL epitope recognized by ACE-specific CTL effectors either loaded onto T2 cells or naturally presented in the context of HLA-A2.1+ and HER-2/neu+ tumor cell lines [5, 29]. Peptide HER-2 (p754-762) was shown to induce CTL from healthy donor-derived PBMC that were capable of killing the colon tumor cell line SW403 expressing HLA-A3 and HER-2/neu [42]. Additional MHC-binding studies with the most common HLA molecules belonging to the HLA-A3 superfamily (HLA-A*1101, HLA-A*3101, HLA-A*3301, and HLA-A*6801) demonstrated that p754-762 was able to bind these alleles [42]. Eberlein and co-workers [69] identified HER-2 peptide (p654-662) from the transmembrane region of this protein as a common epitope presented by various HLA-A27 tumor types, including breast, ovarian, pancreatic, and non-small lung cancer. HER-2 peptides p5-13, p48-56, and p1023-1032 were demonstrated to trigger CTL responses in both HLA-A2+ humans and HLA-A2 transgenic mice. Such CTL lysed HLA-A2+ HER-2/neu+ tumor cells of different origins (breast, colon, lung, and renal cancer) irrespective of the expression levels of HER-2/neu [81]. Shiku and collaborators [87] have identified two HER-2 peptides, p63-71 and p780-788, capable of inducing HLA-A24-restricted CTL responses against various targets, including HLA-A24+ HER-2/neu+ tumor cell lines. MHC class I-presented peptides from HER-2/neu are listed in Table 1.

Table 1.

MHC class I presented CTL HER-2/neu epitopes

Peptide HLA-presenting allele Reference
HER-2 (p5-13) A2 [41]
HER-2 (p8-16) A24 [41]
HER-2 (p48-56) A2 [43]
HER-2 (p63-71) A24 [87]
HER-2 (p106-114) A2 [51]
HER-2 (p369-377) A2, A3, A26 [24, 92]
HER-2 (p435-443) A2 [77]
HER-2 (p654-662) A2 [69]
HER-2 (p665-673) A2 [77]
HER-2 (p689-697) A2 [77]
HER-2 (p754-762) A3, A11, A33 [42]
HER-2 (p773-782) A2 [55]
HER-2 (p780-788) A24 [87]
HER-2 (p785-794) A2 [77]
HER-2 (p789-797) A2 [24]
HER-2 (p799-807) A2 [24]
HER-2 (p952-961) A2 [77]
HER-2 (p1023-1032) A2 [81]

HER-2/neu peptides presented in the context of MHC class II gene products

There is now increasing evidence that optimal cancer vaccines require the collaboration between CTL and TH cells [6]. In the murine system there are several reports demonstrating that both in vitro and in vivo generation of CTL, specific for a tumor protein-epitope, requires cross-priming of tumor antigens by antigen presenting cells and is strictly TH-cell dependent, since depletion of CD4+ T cells totally abrogated the generation of peptide-specific CTL responses [15, 75, 83, 108]. In humans, we were the first to show an absolute requirement of synergistic interactions between autologous tumor-specific CD4+ and CD8+ T cells and DC for optimal killing of autologous tumor cells in patients with various types of metastatic cancers [7]. To this end, we have also shown that an initial interaction of CD4+ T cells with tumor peptide-pulsed autologous DC is required for providing all necessary conditions to the latter to induce optimal CTL killing.

There are also several studies reporting the existence of MHC class II-restricted T-cell responses to HER-2/neu. CD4+ T helper cells from HER-2/neu+ breast and ovarian cancer patients can proliferate and produce lymphokines in response to stimulation with HER-2/neu recombinant protein or synthetic peptides corresponding to immunodominant regions of HER-2/neu such as HER-2 (396-406), HER-2 (776-788), and HER-2 (884-899) [4, 18, 26, 70, 91]. Some of these patients exhibited preexistent immunity to these peptides in that they responded moderately after a short-term stimulation period [26, 48]. We have recently reported [91], on the generation and characterization of CD4+ T cell clones specifically recognizing HER-2 (p776-788). Such clones yielded specific proliferative and cytokine responses in response to DC loaded with HER-2 (p776-788). By performing blocking experiments with monoclonal antibodies and by using DC from allogeneic donors sharing certain DR-alleles, we could show that HER-2 (p776-788) is a promiscuous peptide presented by at least three different alleles (i.e., DRB5*0101, DRB1*0701 and DRB1*0405). In another series of experiments [70], we successfully generated CD4+ T-cell clones recognizing HER-2 (p884-899) in an HLA-DR4-restricted fashion. These clones also recognized human HER-2/neu+ breast, colorectal and pancreatic adenocarcinomas constitutively expressed or induced to express HLA-DR4. The data from both studies provided novel information on the role of HER-2 (p776-788) and HER-2 (p884-899) as naturally processed epitopes expressed by various types of epithelial cancers and the capacity of the HER-2/neu protein to follow the endogenous class II processing pathways. Our data also suggested that these peptides might be attractive as applicable vaccines offering a broad population coverage. MHC class II-presented epitopes from HER-2/neu are listed in Table 2.

Table 2.

MHC class II presented TH HER-2/neu epitopes

Peptide HLA-presenting allele Reference
HER-2 (p62-76) DR4/15, DR51, DR53, DQ6/7 [49]
HER-2 (p605-619) DR4/15, DR51, DR53, DQ6/7 [49]
HER-2 (p765-783) DR4/15, DR51, DR53, DQ6/7 [49]
HER-2 (p776-788) DR51, DR7, DR4 [91]
HER-2 (p777-789) DR4 [99]
HER-2 (p822-836) DR1/11, DR51, DR52, DQ5/7 [49]
HER-2 (p883-889) DR1/11, DR4, DR51, DR52, DR53, DQ6/7 [49]
HER-2 (p884-899) DR4 [70]

Vaccination of cancer patients with HER-2/neu peptides

Most published vaccination trials in humans have used HER-2 (p369-377). Knutson et al. [47] showed that immunization of six patients with breast or ovarian cancer with HER-2 (p369-377) plus GM-CSF resulted in increased peptide-specific T-cell precursor frequencies (PF) in two of them, as measured in IFN-γ ELISPOT assays. The responses were short-lived and not detectable at 5 months after vaccination. In a phase I clinical study [109], patients with metastatic breast, ovarian, or colorectal cancer were immunized with HER-2 (p369-377) in IFA. Peptide-specific CTL were obtained from patients’ peripheral blood and upon restimulation in vitro were able to recognize and lyse target cells pulsed with the peptide but not tumor cells expressing HER-2/neu. In another phase I clinical study [62], HER-2 (p369-377) was administered with GM-CSF. Thirty percent of the patients developed peptide-specific T-cell proliferative responses and the vast majority of them gave peptide-specific DTH responses. Peptide-specific CTL precursors could be detected in only one of them (eight patients were enrolled). No clinical responses were observed.

Disis group [17, 48] vaccinated patients with HER-2/neu-overexpressing breast, ovarian or non-small-cell lung cancers with peptides derived from potential TH epitopes of the HER-2/neu protein admixed with GM-CSF. One of these peptides was HER-2 (p369-384), which also contained the p369-377 CTL epitope. Patients received mixtures of peptides, which included not only HER-2 (p369-384) but also HER-2 (p688-703) and HER-2 (p971-984). Ninety-two percent of patients developed T-cell immunity to HER-2/neu peptides, which was preferentially directed against HER-2 (p369-384). At 1-year follow-up, immunity to HER-2/neu protein persisted in 38% of patients. In a similar trial [47], nineteen HLA-A2 patients with HER-2/neu overexpressing breast and ovarian cancers received a vaccine preparation consisting of putative helper HER-2/neu peptides, which contained within their sequences HLA-A2-binding CTL peptides [also including HER-2 (p369-377)]. After vaccination, the mean peptide-specific T-cell PF to HER-2 (p369-377) was increased in the majority of patients. In addition, the peptide-specific T cells were able to lyse HER-2/neu+, HLA-A2+ tumors. The responses were long-lived and detectable for more than 1 year after the final vaccination. In a similar designed study, Disis et al. [18] demonstrated that FLt3 ligand could act as a vaccine adjuvant in association with HER-2/neu peptide-based vaccines. A summary of the clinical studies based on HER-2/neu peptide vaccines is summarized in Table 3.

Table 3.

HER-2/neu peptide-based vaccinations in patients with HER-2/neu+ cancers

Peptide-vaccine Schedule Immunological effects Referencea
P369-377 in GM-CSF 500 μg/Month. Monthly×6 Increase of peptide-specific CTL precursors [46]
Mixtures of p369-384 p686-703 p971-984 in GM-CSF 500 μg of each peptide/Month. Monthly×6 Increase of peptide-specific CTL precursors. Lysis of HER-2+ tumors [17, 47]
Mixtures of p776-790 p927-941 p1166-1180 20 μg/kg FL/day × 14 days/Month. Monthly×6 500 μg of each peptide on day 7 of FL cycle alone or in GM-CSF FL was effective in boosting the GM-CSF-induced IFN-γ secreting HER-2-specific TH cells [18]
p369-377 in IFA 1 mg/3-4 Week. Weekly×4 Increased reactivity against targets pulsed with p369-377. No reactivity against HER-2+, HLA-A2+ tumors [108]
p369-377 in GM-CSF 10 μg or 500 μg or 1000 μg/Week. Weekly×4 Monthly×6 Proliferative T-cell responses specific for p369-377 DTH responses [62]

In none of these studies clinical activity was tested

a In refs. [17, 47, 108] breast, ovarian, non-small-cell lung and colorectal cancer patients were enrolled. In refs. [46, 18, 62] breast and ovarian cancer patients were enrolled

In most of the patients examined, Disis group could detect increased PF to HER-2/neu CTL epitopes only after vaccination with the relevant peptide and only rarely non-immunized patients exhibited detectable CTL PF [17, 47, 48]. We were able to demonstrate increased CTL PF specific for HER-2 (p369-377) in non-vaccinated patients with HER-2/neu overexpressing tumors [92, 93]. A reason possibly accounting for this may lie in our ability to develop a more sensitive IFN-γ ELISPOT assay favouring the detection of preexisting CTL responses, eventually also in individuals who have developed physiological mechanisms of immunologic tolerance. We used DCs pulsed with HER-2 (p369-377) to stimulate, in the presence of IL-7 and IL-12, autologous PBMCs. We could thus detect IFN-γ producing CTL in 25% of HLA-A2+, 30% of HLA-A26+ and 60% of HLA-A3+ patients with breast, lung, colorectal and prostate cancers but not in healthy individuals [92]. PBMC from patients with increased CTP PF for HER-2 (p369-377) were capable of lysing autologous HER-2/neu+, HLA-A2+ tumors. By applying the same method, we could demonstrate increased PF of CTL specific for HER-2 (p435-443), HER-2 (p665-673), HER-2 (p689-697), HER-2 (p777-785) and HER-2 (p952-960) ranging from 6% to 31% in patients suffering from breast, ovarian, lung, colorectal and prostate cancers [93]. Autologous tumor-killing by patients’ PBMC with increased CTL PF was also observed. The detection of CTL PF for certain HER-2/neu epitopes in non-vaccinated cancer patients is of major importance because it is suggestive of the peptide or mixture of peptides to be included in vaccine formulations.

HER-2/neu CTL peptide analogs with enhanced immunogenicity

The immunogenicity of synthetic peptides which correspond to CTL epitopes in patients is usually limited. Two major reasons may account for this: first, peptide-specific CTL are actively tolerized, either through deletion or anergy induction among high avidity T cells [84, 85]; second, short synthetic peptides (i.e., octamers to decamers) are susceptible to rapid degradation by serum proteases, thus requiring repeated administration of relatively high doses [100, 52, 86, 25]. For these reasons there is a clear interest in designing alternative immunization regimens aiming at a more efficient induction of CTL responses towards tumor-associated antigens. These also include CTL peptide analogs carrying single or multiple amino acid substitutions that have improved immunogenicity compared to the native CTL peptides [79]. Enhancement of immunogenicity is determined by the ability of peptide analogs (or agonists) to induce higher levels of immune responses compared to those induced by the native CTL peptides. Such responses are illustrated by higher levels of cytokine secretion and cytotoxicity upon encounter with the native CTL peptide presented by DCs or the tumor itself. Strong agonistic peptides are designed by two general approaches: (1) modification of HLA anchor residues, resulting in higher HLA binding; and (2) modification of the residues involved in the TCR contact site, resulting in increased response by T cells. The first approach has been successfully used for normally low-affinity binding HLA-A2 CTL peptides from various tumor antigens such as NY-ESO-1 [11], gp100 [65] and Melan-A [100]. With respect to HER-2/neu peptides, Fisk et al. [25] investigated whether oligopeptide analogs can induce an improved anti-CTL response. To this end, PBMC were stimulated with HER-2/neu analogs and the CTL generated recognized the peptides used as immunogen as well as HLA-A2.1+, HER-2+ tumors. Peptide GP2 [HER-2 (9654)] was found to bind poorly to HLA-A2.1 due to a lack of stabilizing contacts with the peptide-binding cleft [52]. In that case, substitutions at the peptide center had a potential impact in increasing peptide affinity. By substituting the amino acid at position 1 with a tyrosine (P1Y) Scardino et al. [81, 82] generated heteroclitic P1Y variants of CTL peptides, HER-2 (9391), HER-2 (9402), HER-2 (9466) and HER-2 (9650), all of which bound to HLA-A2.1 with much higher affinities than did the wild-type epitopes. Such heteroclitic variants were also immunogenic, generating in vivo strong antitumor responses.

However, improved immunogenicity of a given CTL peptide does not necessarily correlate with increased affinity for binding to MHC class I alleles. For instance, in a recently published report [21], HER-2 (10420) restricted by H-2Dq was shown to generate CTL lines in vitro, which upon adoptive transfer, could highly protect non-transgenic mice from spontaneous mammary carcinomas. However, when alanine was substituted for glutamate at position 2, this heteroclitic peptide demonstrated markedly improved recognition by a T cell clone in a cytotoxicity assay as the wild-type peptide. Interestingly, the heteroclitic peptide was found to have a lower binding affinity than the wild-type peptide in a T2Dq stabilization assay, suggesting that its improved stimulatory capacity was instead due to the enhanced stability of the TCR/MHC/peptide complex. In a similar manner, Vertuani et al. [101] have most recently demonstrated that a HER-2 (9369) analog generated by introduction of V at positions 2 and 9 proved to be a more potent immunogen compared to wild-type HER-2 (9369) although the analog bound to HLA-A2.1 with the same affinity as the wild-type peptide. The authors demonstrated that the high biological activity of HER-2 (9369) analog was associated with a slower dissociation kinetic profile, resulting in an epitope with greater HLA-A2.1 stability.

The second approach to designing epitopes from tumor antigens, which involves the replacement of residues responsible for TCR contact, has been mainly applied to melanoma differentiation antigens, and carcinoembryonic antigen [80, 76, 89]. Castilleja et al. [13] modeled the complex of HLA-A2.1 with HER-2 (9369) in order to identify side chain orientation of the latter. They then modified HER-2 (9369) at the central Ser (which is involved in the contact with the TCR) in different ways, producing three variants one of which was a weaker stimulator than native HER-2 (9369) for induction of lytic function, indicating that this particular variant hindered TCR activation. However, CTL raised against this variant were less apoptotic and upon restimulation with the wild-type peptide became highly active, recognizing tumors endogeneously presenting HER-2 (9369). Agonists produced by modifications at TCR contacts may circumvent apoptotic pathways and mechanisms of active tolerance and thus, may provide improved approaches to the rational design of cancer vaccines. The data from this study offer the prospect of sequential stimulation of PBMC with weak and strong gonists (produced by modifications at anchor residues 2 and 9 or at the central portion of the peptide, or a combination of both) for developing efficient and long-lasting antitumor responses.

Active immunization targeting HER-2/neu and other tumor-related antigenic molecules

Limited efficacy of tumor-specific CTL in cancer patients may be attributed to several factors including (a) active immunosuppression either via soluble factors released by the tumor cells [49] or via T regulatory cells [35], (b) down-regulation of MHC class I products and co-stimulatory molecules by the tumor cells [38, 49] and (c) inadequate TH function essential for optimal CTL activation [7, 75]. Early activation of type-1 innate effector cells such as DC, NK and NKT cells, which produce TH1-inducing cytokines, would be essential for supporting the generation and amplification of TH1-dependent tumor-specific CTL. This could be achieved by developing an efficient tumor peptide-vaccine protocol that can overcome strong immunosuppression in tumor-bearing hosts and at the same time promote the generation and activation of tumor-specific CTL and TH. Unmethylated cytosine-phosphorothioate-guanine (CpG) containing oligonucleotides are well-known to promote TH1-type immune response through binding to TLR-9 in DCs [1, 32, 36]. Internalization of CpG-ODN is a prerequisite for activation of TLR-9 and initiation of its signaling pathway [1] which suggests that encapsulation of CpG-ODN in liposomes might increase their uptake by DCs to produce higher levels of IL-12, which in turn may potentially induce higher amounts of IFN-γ producing NK and NKT cells. Such a pathway involving type-1 innate immune mediators through TLR engagement may trigger activation of bystander T cells [23, 26]. Accordingly, tumor-vaccines including CTL and TH peptides along with CpG-ODN would benefit from the generation of type-1 innate and adoptive immunity with the former exerting direct antitumor effects but also indirect via amplification of the latter.

Xenogeneic homologues of tumor-associated proteins have been shown upon immunization to induce strong in vivo antitumor responses in mice by overcoming tolerance to weakly immunogenic self-antigens [104]. DC-based xenovaccination for prostate cancer in humans has also produced strong TH1 immunity for self-prostate antigens, with no data available so far, however, on tumor response [27]. Chimeric fusion proteins resulting from translocations can be necessary for the persistence of the tumor [80, 107], thus critically marking the tumor cells as a target for immunotherapy. Peptides generated from proteolytic processing of fusion proteins would have a “non-self” sequence of amino acids, which, if displayed via MHC molecules, could result in potent T-cell-mediated immunity [106]. Cytogenetic studies have revealed a number of translocations in both breast cancer lines and primary tumors [28, 102]. There is also an association between translocations and HER-2/neu expression in tumor cells [97]. These findings suggest a potential utility of peptides from xenogeneic homologues of breast tumor antigens and for translocation-specific peptides combined with HER-2/neu peptides in novel active immunotherapies.

There is now evidence to suggest that tumor escape variants are likely to emerge after treatment with increasingly effective immunotherapies. Thus, it is important that tumor-associated antigens be part of molecules which are obligatory for the survival of tumor cells. Immunization against such tumor antigens would overcome tumor escape attributable to antigenic variation because losing the survival-related molecule may result in tumor cell death. One of the antigens that is needed by cancer cells to survive is the antiapoptotic protein, survivin, which is overexpressed in several types of cancer [105, 95] and correlates with a more aggressive disease and poor survival [90, 37]. So far, survivin CTL epitopes restricted by various HLA class I alleles have been identified and demonstrated to be naturally expressed on the surface of tumor cell lines [3, 34, 59, 74]. Telomerase is a ribonucleoprotein that mediates RNA-dependent synthesis of telomeric DNA, thus maintaining telomere length and chromosomal stability [10]. Normal tissues display little or no telomerase activity [46, 63]. Most of the human tumors express high telomerase activity [46, 63] and telomerase peptides have been recently demonstrated to induce strong CTL responses against tumor cell lines naturally expressing these epitopes [96, 68, 33, 2, 94]. Vaccines comprising of HER-2/neu CTL and TH peptides mixed with survivin and/or telomerase CTL peptides may more efficiently target tumor cells with improved clinical results.

Growth of tumor cells and metastasis are dependent on mechanisms inducing angiogenesis. Therapy against angiogenesis represents a new modality for cancer therapy. Vaccination protocols for active specific immunotherapy include endothelial cell vaccines, DNA vaccines, DC vaccines and peptide or protein vaccines [53, 64, 71, 73]. The rational for such vaccination strategies is to target antigens selectively expressed by tumor-associated endothelial cells, thus inducing tumor necrosis by blocking blood supply through angiogenesis inhibition. Combined vaccination with HER-2/neu peptides and peptides targeting angiogenesis-related molecules (e.g. vascular endothelial growth factor receptor [54]) may provide a new strategy for the rational design of cancer vaccines.

Conclusion

The HER-2/neu oncoprotein gene encompasses sequences that can activate, upon recognition in the context of certain alleles on DC, both CTL and TH cells. Such cells, with specificity for HER-2/neu peptides, have been isolated from in vitro cultures but also from patients’ peripheral blood. Moreover, HER-2/neu peptide-specific CTL have been shown to efficiently lyse only HER-2/neu+ tumor cells expressing the appropriate allele(s).

However, how such HER-2/neu specific T-cell responses can be translated into effective therapeutic responses remains so far unclear. Combination of active immunotherapy with other therapeutic modalities, such as surgery or chemotherapy, might be necessary for achieving improved clinical responses. Another topic of major importance is the definition of optimum vaccination protocols including vaccine dose, number of doses, and modus of administration. The identification and optimal use of new adjuvants which are necessary for breaking tolerance to self-proteins or peptides is also an important issue. A better understanding of host–tumor interactions will surely provide insights for the development of more effective vaccination strategies mostly based on mixtures of HER-2/neu peptides with other peptides targeting tumor cells directly or even indirectly. Moreover, in light of recent findings demonstrating the generation of negative regulatory mechanisms of anti-tumor immunity in the tumor-bearing host, approaches that overcome this immunosuppressive state should be developed. Potent adjuvants, administered along with peptide vaccines, that bridge type-1 innate and acquired immunity are essential for inducing tumor-specific CTL. Implementation of these modalities in the clinical setting will certainly help to more precisely define the role of HER-2/neu peptide vaccines as effective immunotherapy of HER-2/neu overexpressing cancers.

Acknowledgements

We wish to thank Miss Joanna Doukoumopoulou for her excellent secretarial assistance

Footnotes

This article is a symposium paper from the conference “Progress in Vaccination against Cancer 2004 (PIVAC 4)”, held in Freudenstadt-Lauterbad, Black Forest, Germany, on 22–25 September 2004

References

  • 1.Ahmad-Najad P, Hacke H, Rutz M, Bauer S, Vabulas RM, Wagner H. Bacterial DNA and lipopolysaccharides active Toll-like receptors at distinct cellular compartments. Eur J Immunol. 2002;32:1958. doi: 10.1002/1521-4141(200207)32:7<1958::AID-IMMU1958>3.0.CO;2-U. [DOI] [PubMed] [Google Scholar]
  • 2.Amarnath SM, Dyer CE, Ramesh A, Iwuagwu O, Drew PJ, Greenman J. In vitro quantification of the cytotoxic T lymphocyte response against human telomerase reverse transcriptase in breast cancer. Int J Oncol. 2004;25:211. [PubMed] [Google Scholar]
  • 3.Andersen MH, Pedersen LO, Becker JC, Straten PT. Identification of a cytotoxic T lymphocyte reponse to the apoptosis inhibitor protein survirin in cancer patients. Cancer Res. 2001;61:869. [PubMed] [Google Scholar]
  • 4.Anderson BW, Kudelka AD, Honda T, Pollack MS, Gershenson DM, Gillogly MA, Murray JI, Ioannides CG. Induction of determinant spreading and of TH1 responses by in vitro stimulation with HER-2 peptides. Cancer Immunol Immunother. 2000;49:459. doi: 10.1007/s002620000143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Baxevanis CN, Gritzapis AD, Tsitsilonis OE, Katsoulas HL, Papamichail M. HER-2/neu-derived epitopes are also recognized by cytotoxic CD3+ CD56+ (Natural killer T) lymphocytes. Int J Cancer. 2002;98:864. doi: 10.1002/ijc.10251. [DOI] [PubMed] [Google Scholar]
  • 6.Baxevanis CN, Sotiropoulou PA, Sotiriadou NN, Papamichail M. Immunobiology of HER-2/neu oncoprotein and its potential application in cancer immunotherapy. Cancer Immunol Immunother. 2004;53:166. doi: 10.1007/s00262-003-0475-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Baxevanis CN, Voutsas IF, Tsitsilonis OE, Gritzapis AD, Sotiriadou R, Papamichail M. Tumor-specific CD4+ T lymphocytes from cancer patients are required for optimal induction of cytotoxic T cells against the autologous tumor. J Immunol. 2000;164:3902. doi: 10.4049/jimmunol.164.7.3902. [DOI] [PubMed] [Google Scholar]
  • 8.Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. The foreign antigen binding site and T cell recognition regions of class I histocompatbility antigens. Nature. 1987;329:512. doi: 10.1038/329512a0. [DOI] [PubMed] [Google Scholar]
  • 9.Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. Structure of the human class I histocompatibility antigen HLA-A2. Nature. 1987;329:506. doi: 10.1038/329506a0. [DOI] [PubMed] [Google Scholar]
  • 10.Blackburn EH. Telomerases. Annu Rev Biochem. 1992;61:113. doi: 10.1146/annurev.bi.61.070192.000553. [DOI] [PubMed] [Google Scholar]
  • 11.Bownds S, Tong-On P, Rosenberg SA, Parkhurst M. Induction of tumor-reactive cytotoxic T-lymphocytes using a peptide from NY-ESO-1 modified at the carboxy-terminus to enhance HLA-A2.1 binding affinity and stability in solution. J Immunother. 2001;24:1. doi: 10.1097/00002371-200101000-00001. [DOI] [PubMed] [Google Scholar]
  • 12.Brossart P, Stuhler G, Flad T, Stevanovic S, Sammensee HG, Kanz L, Brugger W. HER-2/neu-derived peptides are tumor-associated antigens expressed by human renal cell and colon carcinoma lines and are recognized by in vitro induced specific cytotoxic T lymphocytes. Cancer Res. 1998;58:732. [PubMed] [Google Scholar]
  • 13.Castilleja A, Carter D, Efferson CL, Ward NE, Kawano K, Fisk B, Kudelka AP, Gershenson DM, Murray JI, O’Brian CA, Ioannides CG. Induciton of tumor-reactive CTL by C-side chain variants of the CTL epitope HER-2/neu proton-ogene (369-377) selected by molecular modeling of the peptide HLA-A2 complex. J Immunol. 2002;169:3545. doi: 10.4049/jimmunol.169.7.3545. [DOI] [PubMed] [Google Scholar]
  • 14.Celis E, Tsai V, Crimi C, DeMars R, Wentworth PA, Chesnut RW, Grey HM, Sette A, Serra HM. Induction of anti-tumor cytotoxic T lymphocytes in normal humans using primary cultures and synthetic peptide epitopes. Proc Natl Acad Sci USA. 1994;91:2105. doi: 10.1073/pnas.91.6.2105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Chen PW, Ananthaswamy HN. Rejection of K1735 murine melanoma in syngeneic hosts required expression of MHC class I antigens and either class II antigens of IL-2. J Immunol. 1993;151:244. [PubMed] [Google Scholar]
  • 16.D’Amaro J, Houbiers JG, Drijfhout JW, Brandt RMP, Schipper R, Bavinck JN, Melief CJM, Kast WM. A computer program for predicting possible cytotoxic T lymphocyte epitopes based on HLA class I peptide-binding motifs. Hum Immunol. 1995;43:13. doi: 10.1016/0198-8859(94)00153-H. [DOI] [PubMed] [Google Scholar]
  • 17.Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, Cheever MA, Knutson KL, Shiffmann K. Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol. 2002;20:2624. doi: 10.1200/JCO.2002.06.171. [DOI] [PubMed] [Google Scholar]
  • 18.Disis ML, Rinn K, Knutson KL, Davis D, Caron D, dela Rosa C, Schiffman K. FLt3 ligand as a vaccine adjuvant in association with HER-2/neu peptide-based vaccines in patients with HER-2/neu-overexpressing cancers. Blood. 2002;99:2845. doi: 10.1182/blood.V99.8.2845. [DOI] [PubMed] [Google Scholar]
  • 19.Eccles SA. The role c-erbB-2/HER-2/neu in breast cancer progression and metastasis. J Mammary Gland Biol Neoplasia. 2001;6:393. doi: 10.1023/A:1014730829872. [DOI] [PubMed] [Google Scholar]
  • 20.Engelhard VH. Structure of peptides associated with MHC class I molecules. Curr Opin Immunol. 1994;6:13. doi: 10.1016/0952-7915(94)90028-0. [DOI] [PubMed] [Google Scholar]
  • 21.Ercolini AM, Machiels J-PH, Chen YC, Slansky JE, Giedlen M, Reilly RT, Jaffee EM. Identification and characterization of the immunodominant rat HER-2/neu MHC class I epitope presented by spontaneous mammary tumor from HER-2/neu-transgenic mice. J Immunol. 2003;170:4273. doi: 10.4049/jimmunol.170.8.4273. [DOI] [PubMed] [Google Scholar]
  • 22.Falk K, Rotzschke O, Stevanovic S, Jung G, Rammensee HG. Allele-specific motifs revealed by sequencing of self-peptides eluted from MHC molecules. Nature. 1991;351:290. doi: 10.1038/351290a0. [DOI] [PubMed] [Google Scholar]
  • 23.Farina C, Theil D, Semlinger B, Hohlfeld R, Meinl E. Distinct responses of monocytes to Toll-like receptor ligands and imflammatory cytokines. Int Immunol. 2004;16:799. doi: 10.1093/intimm/dxh083. [DOI] [PubMed] [Google Scholar]
  • 24.Fisk B, Blevins TL, Wharton JT, Ioannides CG. Identification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor- specific cytotoxic T lymphocyte lines. J Exp Med. 1995;181:2109. doi: 10.1084/jem.181.6.2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fisk B, Chesak B, Pollack MS, Wharton JT, Ioannides CG. Oligopeptide induction of a cytotoxic T lymphocyte response to HER-2/neu proto-oncogene in vitro. Cell Immunol. 1994;157:415. doi: 10.1006/cimm.1994.1238. [DOI] [PubMed] [Google Scholar]
  • 26.Fisk B, Hudson JM, Kavanagh J, Wharton JT, Murray JL, Ioannides CG, Kudelka AP. Existent proliferative response of peripheral blood mononuclear cells from healthy donors and ovarian cancer patients to HER-2 peptides. Anticancer Res. 1997;17:45. [PubMed] [Google Scholar]
  • 27.Fong L, Brockstedt D, Benike C et al Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy from HER-2/neu-transgenic mice. J Immunol 170:4273 [DOI] [PubMed]
  • 28.Goodison S, Viars C, Urquidi V. Molecular cytogenetic analysis of a human breast metastasis model: identification of phenotype-specific chromosomal rearrangements. Cancer Genet Cytogenet. 2005;156:37. doi: 10.1016/j.cancergencyto.2004.04.005. [DOI] [PubMed] [Google Scholar]
  • 29.Gritzapis AD, Sotiriadou NN, Papamichail M, Baxevanis CN. Generation of human tumor-specific CTLs in HLA-A2.1 trangenic mice using unfractionated peptides from eluates of human primary breast and ovarian tumors. Cancer Immunol Immunother. 2004;53:1027. doi: 10.1007/s00262-004-0541-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Guy CT, Webmaster MA, Schaller M, Parsons TJ, Cardiff RD, Muller WJ. Expression of the neu protooncogene in the mammary epithelium of transgenic mice induces metastatic diseases. Proc Natl Acad Sci USA. 1992;89:10578. doi: 10.1073/pnas.89.22.10578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hausmann S, Martin M, Gauthier L, Wucherpfennig K. Structural features of autoreactive TCR that determine the degree of degenerary in peptide recognition. J Immunol. 1999;162:338. [PubMed] [Google Scholar]
  • 32.Hemmi H, Takeuchi O, Kawai T, Kaison T, Sato S, Sanjo H, Mattimoto Hoshino K, Wagner H, Takeda K, Akira SA. Toll-like receptor recognize bacterial DNA. Nature. 2000;408:740. doi: 10.1038/35047123. [DOI] [PubMed] [Google Scholar]
  • 33.Hernandez J, Schoeder P, Blondelle SE, Pons FG, Lone YC, Simora A, Langlade-Demoyen P, Wilson DB, Zanetti M. Antigenicity and immunogenicity of peptide analogues of a low affinity peptide of the human telomerase reverse transcriptase tumor antigen. Eur J Immunol. 2004;34:2331. doi: 10.1002/eji.200425134. [DOI] [PubMed] [Google Scholar]
  • 34.Hirohashi Y, Torigoe T, Maeda A, Nabeta Y, Kamiguchi K, Sato T, Yoda J, Ikeda H, Hirata K, Yamanaka N, Sato N. An HLA-A24-restricted cytotoxic T lymphocyte epitope of a tumor-associated protein, survirin. Clin Cancer Res. 2002;8:1731. [PubMed] [Google Scholar]
  • 35.Hori S, Takahashi T, Sakeguchi S. Control of autoimmunity by naturally arising regulatory CD4+ T cells. Adv Immunol. 2003;81:331. doi: 10.1016/s0065-2776(03)81008-8. [DOI] [PubMed] [Google Scholar]
  • 36.Ikeda H, Chamoto K, Tsuji T, Suzuki Y, Wakita D, Takeshima T, Takashi N. The critical role of type-1 innate and acquired immunity in tumor immunotherapy. Cancer Sci. 2004;95:697. doi: 10.1111/j.1349-7006.2004.tb03248.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ito T, Shiraki K, Sugomoto K, Yamanaka T, Fujikawa K, Ito M, Takase K, Moriyama M, Kawano H, Hayashida M, Nakano Y, Suzuki A. Survirin promotes cell proliferation in human hepatocellular carcinoma. Hepatology. 2000;31:1080. doi: 10.1053/he.2000.6496. [DOI] [PubMed] [Google Scholar]
  • 38.Kageshita T, Hirai S, Ono T, Hicklin DJ, Ferrone S. Down-regulation of HLA class I antigen-processing molecules in malignant melanoma: association with disease progression. Am J Pathol. 1999;154:745. doi: 10.1016/S0002-9440(10)65321-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kawai K, Ohashi PS. Immunological function of a defined T cell population tolerized to low-affinity self antigens. Nature. 1995;374:68. doi: 10.1038/374068a0. [DOI] [PubMed] [Google Scholar]
  • 40.Kawakami Y, Eliyadu S, Sakaguchi K, Robbins PF, Rivoltini I, Yannelli JR, Apella E, Rosenberg SA. Identification of the immunodominant peptides of the MART-1 human melanoma antigen recognized by the majority of HLA-A2-restricted tumor infiltrating lymphocytes. J Exp Med. 1994;180:347. doi: 10.1084/jem.180.1.347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kawashima I, Hudson SJ, Tsai V, Southwood S, Takesako K, Appella E, Sette A, Celis E. The multiepitope approach for immunotherapy of cancer: identification of several CTL epitopes from various tumor-associated antigens expressed on solid epithelial tumors. Hum Immunol. 1998;59:1. doi: 10.1016/S0198-8859(97)00255-3. [DOI] [PubMed] [Google Scholar]
  • 42.Kawashima I, Tsai V, Southwood S, Takesako K, Sette A, Celis E. Identification of HLA-A3-restricted cytotoxic T lymphocytes epitopes from carcinoembryonic antigen and HER-2/neu by primary in vitro immunization with peptide-pulsed dendritic cells. Cancer Res. 1999;59:431. [PubMed] [Google Scholar]
  • 43.Keogh E, Fikes J, Southwood S, Celis E, Chesnut R, Sette A. Identification of new epitopes from four different tumor-associated antigens: recognition of naturally processed epitopes correlates with HLA-A*0201-binding affinity. J Immunol. 2001;167:787. doi: 10.4049/jimmunol.167.2.787. [DOI] [PubMed] [Google Scholar]
  • 44.Kessler JH, Beekman NJ, Bres-Vloemans SA, Verdijk P, van Veelen PA, Kloosterman-Joosten AM, Vissers DC, ten Bosch GJ, Kester MG, Sijts A, Drijfhout JW, Ossendorp F, Offringa R, Melief CJM. Efficient identification of novel HLA-A*0201-presented cytotoxic T lymphocyte epitopes in the widely expressed tumor antigen PRAME by proteasome-mediated digestion analysis. J Exp Med. 2001;193:73. doi: 10.1084/jem.193.1.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kiessling R, Wel WZ, Herrmann F, Lindencrona JA, Cheudhury A, Kono K, Seliger B. Cellular immunity to the HER-2/neu protooncogene. Adv Cancer Res. 2002;85:101. doi: 10.1016/S0065-230X(02)85004-7. [DOI] [PubMed] [Google Scholar]
  • 46.Kim NW. Clinical implications of telomerase in cancer. Eur J Cancer. 1997;33:781. doi: 10.1016/S0959-8049(97)00057-9. [DOI] [PubMed] [Google Scholar]
  • 47.Knutson KL, Shiftmann K, Cheever MA, Disis ML. Immunization of cancer patients with a HER-2/neu. HLA-A2 peptide, p369-377, results in short-lived peptide-specific immunity. Clin Cancer Res. 2002;8:1014. [PubMed] [Google Scholar]
  • 48.Knutson KL, Shiftmann K, Disis ML. Immunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity to cancer patients. J Clin Invest. 2001;107:477. doi: 10.1172/JCI11752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kobayashi H, Wood M, Song Y, Appelle E, Celis E. Defining promiscuous MHC class II helper T-cell epitopes for HER-2/neu tumor antigen. Cancer Res. 2000;60:52258. [PubMed] [Google Scholar]
  • 50.Kondo A, Sidney J, Southwood S, del Guercio MF, Appella E, Sakamoto H, Celis E, Grey HM, Chesnut RW, Kubo RT. Prominent roles of secondary anchor residues in peptide binding to HLA-A24 human class I molecules. J Immunol. 1995;155:4307. [PubMed] [Google Scholar]
  • 51.Kono K, Rongcun Y, Charo J, Ichihara F, Celis E, Sette A, Appellia E, Sekikawa T, Matshumoto Y, Kiessling R. Identification of HER-2/neu-derived peptide epitopes recognized by gastric cancer-specific cytotoxic T lymphocytes. Int J Cancer. 1998;78:202. doi: 10.1002/(SICI)1097-0215(19981005)78:2<202::AID-IJC14>3.0.CO;2-C. [DOI] [PubMed] [Google Scholar]
  • 52.Kuhns JJ, Batalia MA, Shuqin Y, Collins EJ. Poor binding of a HER-2/neu epitope (GP2) to HLA-A2.1 is dues to a lack of interaction with the center of the peptide. J Biol Chem. 1999;274:26422. doi: 10.1074/jbc.274.51.36422. [DOI] [PubMed] [Google Scholar]
  • 53.Bohlen P, Hicklin DJ. against angiogenesis-associated antigens: a novel cancer immunotherapy strategy. Curr Mol Med. 2003;3:773. doi: 10.2174/1566524033479438. [DOI] [PubMed] [Google Scholar]
  • 54.Liu J-Y, Wei Y, Yang L, Zhao X, Tian L, Hou J, Niu Y, Liu F, Jiang Y, Xu B, Wy Y, Su J, Lou Y, He Q, Wen Y, Yang J, Kan B, Mao Y, Luo F, Peng F. Immunotherapy of tumors with vaccine based on quail homologous vascular endothelial growth factor receptor-2. Blood. 2003;102:1815. doi: 10.1182/blood-2002-12-3772. [DOI] [PubMed] [Google Scholar]
  • 55.Lustgarten L, Dominguez AL, Guadros C. The CD8+ T cell repertoire against HER-2/neu antigens in new transgenic mice is of low avidity with antitumor activity. Eur J Immunol. 2004;34:752. doi: 10.1002/eji.200324427. [DOI] [PubMed] [Google Scholar]
  • 56.Lustgarten J, Theobald M, Labadie C, LaFace D, Peterson P, Disis ML, Cheever MA, Sherman LA. Identification of HER-2/neu CTL epitopes using double transgenic mice expressing HLA-A2.1 and human CD8. Hum Immunol. 1997;52:109. doi: 10.1016/S0198-8859(96)00292-3. [DOI] [PubMed] [Google Scholar]
  • 57.Morel S, Levy F, Burlet-Schiltz O, Brasseur F, Probst-Kepper M, Peitrequin Al, Monsarrat B, Van Veithove R, Cerottini JC, Boon T, Gairin JE, Van den Eynde BJ. Processing of some antigens by the standard proteasome but not by the immunoproteasome results in poor presentation by dendritic cells. Immunity. 2000;12:107. doi: 10.1016/S1074-7613(00)80163-6. [DOI] [PubMed] [Google Scholar]
  • 58.Morgan DK, Kreuwell HT, Fleck S, Levitsky HI, Pardoll DM, Sherman LA. Activation of low avidity CTL specific for a self epitope results in tumor rejection but not autoimmunity. J Immunol. 1998;160:643. [PubMed] [Google Scholar]
  • 59.Moriai R, Asanuma K, Kobayashi D, Yajima T, Yagihashi A, Yamada M, Watanabe N. Quantitative analysis of this anti-apoptotic gene survivin-expression in malignant haematopoietic cells. Anticancer Res 21:595 [PubMed]
  • 60.Muller WJ, Sinn E, Pattengale PK, Wallace R, Leder P. Single-step induction of mammary adenocarcinoma in transgenic mice bearing the activated c-neu oncogene. Cell. 1988;54:104. doi: 10.1016/0092-8674(88)90184-5. [DOI] [PubMed] [Google Scholar]
  • 61.Mullins DW, Bullock TN, Colella TA, Robila VV, Engelhard VH. Immune responses to the HLA-A*0201-restricted epitopes of tyrosinase and glycoprotein 100 enable control of melanoma outgrowth in HLA-A*0201-transgenic mice. J Immunol. 2001;167:4853. doi: 10.4049/jimmunol.167.9.4853. [DOI] [PubMed] [Google Scholar]
  • 62.Murray JL, Przepiorka D, Ioannides C. Clinical trials of HER-2/neu-specific vaccines. Semin Oncol. 2000;27:71. [PubMed] [Google Scholar]
  • 63.Nakamura TM, Morin GH, Chapman KB, Weinrich SL, Andrews WH, Lingner J, Harley CB, Cech TR. Science. 1997;277:955. doi: 10.1126/science.277.5328.955. [DOI] [PubMed] [Google Scholar]
  • 64.Niethammer AG, Xiang R, Becker JC, Wodrsch H, Derte U, Karsten G, Eliceiri BP, Reisfeld RA. A DNA vaccine against VEGF receptor 2 prevents effective angiogenesis and inhibits tumor growth. Nat Med. 2002;8:1369. doi: 10.1038/nm794. [DOI] [PubMed] [Google Scholar]
  • 65.Overwijk WW, Tsung A, Irvine KR, Parkhurst MR, Goletz TJ, Tsung K, Carroll MW, Liu C, Moss B, Rosenberg SA, Restifo NP. gp100/pmel 17 is a murine tumor rejection antigen: induction of “self”-reactive, tumoricidal T cells using high-affinity, altered peptide ligand. J Exp Med. 1998;188:277. doi: 10.1084/jem.188.2.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Paik S, Hazan R, Fisher ER, Sass RE, Fisher B, Redmond C, Schlessinger J, Lippman ME, King CR. Pathologic findings from the National Surgical Adjuvant Breast and Bowell Project:prognostic significance of erbB-2 protein overexpression in primary breast cancer. J Clin Oncol. 1990;8:103. doi: 10.1200/JCO.1990.8.1.103. [DOI] [PubMed] [Google Scholar]
  • 67.Parker KC, Bednarek MA, Coligan JE. Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains. J Immunol. 1994;152:163. [PubMed] [Google Scholar]
  • 68.Parkhurst MR, Riley JP, Igarashi T, Li Y, Robbins PF, Rosenberg SA. Immunization of patients with hTERT: 540-548 peptide induces peptide-reactive T lymphocytes that do not recognize tumors endogenously expressing tolemerase. Clin Cancer Res. 2004;15:4688. doi: 10.1158/1078-0432.CCR-04-0325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Peiper M, Goedegebuure MP, Linehan DC, Ganguly E, Douville CC, Eberlein TJ. The HER-2/neu-derived peptide p654-662 is a tumor-associated antigen in human pancreatic cancer recognized by cytotoxic T lymphocytes. Eur J Immunol. 1997;27:1115. doi: 10.1002/eji.1830270511. [DOI] [PubMed] [Google Scholar]
  • 70.Perez SA, Sotiropoulou PA, Sotiriadou NM, Mamalaki A, Gritzapis AD, Echner H, Voelter W, Pawelec G, Papamichail M, Baxevanis CN. HER-2/neu-derived peptide 884-899 is expressed by human breast, colorectal and pancreatic adenocarcinomas and is recognized by in-vitro-induced specific CD4+ T cell clones. Cancer Immunol Immunother. 2002;50:615. doi: 10.1007/s002620100225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Plum SM, Fogmer WE. Anti-angiogenic vaccines as a treatment modality for cancer. Curr Opin Investig Drugs. 2004;5:1243. [PubMed] [Google Scholar]
  • 72.Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic S. SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics. 1999;50:213. doi: 10.1007/s002510050595. [DOI] [PubMed] [Google Scholar]
  • 73.Reisfeld RA, Niethammer AG, Luo Y, Xiang R. DNA vaccines suppress tumor growth and metastases by the induction of anti-angiogenesis. Immunol Rev. 2004;199:181. doi: 10.1111/j.0105-2896.2004.00137.x. [DOI] [PubMed] [Google Scholar]
  • 74.Reker S, Meier A, Holten-Andersen L, Svane IM, Becker JC, thor Straten P, Andersein MH. Identification of novel survirin-derived CTL epitopes. Cancer Biol Ther. 2004;3:173. doi: 10.4161/cbt.3.2.611. [DOI] [PubMed] [Google Scholar]
  • 75.Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic cell can be temporal bridge between a CD4+ T-helper and a T-killer cell. Nature. 1998;393:474. doi: 10.1038/30989. [DOI] [PubMed] [Google Scholar]
  • 76.Rivoltini L, Squarcina P, Loftus DJ, Castelli C, Tarsini P, Mazzocchi A, Rini F, Viggiano V, Belli F, Parmiani G. A superagonist variant of peptide MART1/Melan A27-35 elicits anti-melanoma CD8+ T cells with enhanced functional characteristics: implication for more effective immunotherapy. Cancer Res. 1999;39:301. [PubMed] [Google Scholar]
  • 77.Rongcun Y, Salazar-Onfray F, Charo J, Malmberg KJ, Evrin K, Maes H, Komo K, Hising C, Petersson M, Larsson O, Lan L, Appella E, Sette A, Celis E, Kiessling R. Identification of new HER-2/neu-derived peptide epitopes that can elicit specific CTL against autologous and allogeneic carcinomas and melanomas. J Immunol. 1999;163:1037. [PubMed] [Google Scholar]
  • 78.Ruppert J, Sidney J, Celis E, Kubo RT, Grey HM, Sette A. Prominent role of secondary anchor residues in peptide binding to HLA-A2.1 molecules. Cell. 1993;74:929. doi: 10.1016/0092-8674(93)90472-3. [DOI] [PubMed] [Google Scholar]
  • 79.Salazar E, Zaremba S, Arlen PM, Tsang KY, Schlom J. Agonist peptide from a cytotoxic T-lymphocyte epitope of human carcinoembryonic antigen stimulates production of TC1-type cytokines and increases tyrosine phosphorylation more efficiently than cognate peptide. Int J Cancer. 2000;85:829. doi: 10.1002/(SICI)1097-0215(20000315)85:6<829::AID-IJC16>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 80.Sanchez-Garcia L, Grutz G. Tumorigenic activity of the BCR-ABL oncogenes is mediated by BCL2. Proc Natl Acad Sci USA. 1995;92:5287. doi: 10.1073/pnas.92.12.5287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Scardino A, Alves P, Gross DA, Tourdot S, Graff-Dubois S, Angevin E, Firat H, Chouaib S, Lemonnier F, Nadler LM, Cardoso AA, Kosmatopoulos K. of HER-2/neu immunogenic epitopes presented by renal cell carcinoma and other human epithelial tumors. Eur J Immunol. 2001;31:3261. doi: 10.1002/1521-4141(200111)31:11<3261::AID-IMMU3261>3.0.CO;2-4. [DOI] [PubMed] [Google Scholar]
  • 82.Scardino A, Gross D-A, Alves P, Schultze JL, Graff-Dubois S, Faure O, Tourdot S, Chouaib S, Nadler LM, Lemonnier FA, Vonderheide RH, Cardoso AA, Kospatopoulos K. HER-2/neu and hTERT cryptic epitopes as novel targets for broad spectrum tumor immunotherapy. J Immunol. 2002;168:5900. doi: 10.4049/jimmunol.168.11.5900. [DOI] [PubMed] [Google Scholar]
  • 83.Schoenberger PS, Toes RE, van der Voort EI, Offringa R, Melief CJM. help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393:480. doi: 10.1038/31002. [DOI] [PubMed] [Google Scholar]
  • 84.Sercarz EE. Driver clones and determinant spreading. J Autoimmun. 2000;14:275. doi: 10.1006/jaut.2000.0380. [DOI] [PubMed] [Google Scholar]
  • 85.Sercarz EE, Lehmann PV, Ametani A, Benichou G, Miller A, Moudgil K. Dominance and crypticity of T-cell antigenic determinants. Ann Rev Immunol. 1993;11:729. doi: 10.1146/annurev.iy.11.040193.003501. [DOI] [PubMed] [Google Scholar]
  • 86.Serody JS, Collins EJ, Tisch RM, Kuhns JJ, Freilinger JA. T cell activity after dendritic cell vaccination is dependent on both the type of antigen and the mode of delivery. J Immunol. 2000;164:4961. doi: 10.4049/jimmunol.164.9.4961. [DOI] [PubMed] [Google Scholar]
  • 87.Shiku H, Wang L, Ikuta Y, Okugawa T, Schmitt M, Gu X, Akiyoshi K, Sunamoto J, Nakamura H. Development of a cancer vaccine: peptides, proteins and DNA. Cancer Chemother Pharmacol. 2000;46:77. doi: 10.1007/s002800000179. [DOI] [PubMed] [Google Scholar]
  • 88.Sidney J, Southwood S, del Guercio MF, Grey HM, Chesnut RW, Kubo RT, Sette A. Specificity and degenerary in peptide binding to HLA-B7-like class I molecules. J Immunol. 1996;157:3480. [PubMed] [Google Scholar]
  • 89.Slansky JE, Rattis FM, Boyd IF, Fahmy T, Jaffee EM, Schneck JP, Margulles DH, Pardoll DM. Enhanced antigen-specific antitumor immunity with altered peptide ligands that stabilize the MHC-peptide-TCR complex. Immunity. 2000;13:529. doi: 10.1016/S1074-7613(00)00052-2. [DOI] [PubMed] [Google Scholar]
  • 90.Smith SD, Wheeler MA, Plescia J, Colberg JW, Weiss RM, Altieri DC. Urine detection of survirin and diagnosis of bladder cancer. J Am Med Assoc. 2001;285:324. doi: 10.1001/jama.285.3.324. [DOI] [PubMed] [Google Scholar]
  • 91.Sotiriadou R, Perez SA, Gritzapis AD, Sotiropoulou PA, Echner H, Heinzel S, Mamalaki A, Pawelec G, Voelter W, Baxevanis CN, Papamichail M. Peptide HER2 (776-788) represents a naturally processed broad MHC class II-restricted T cell epitope. Br J Cancer. 2001;85:1527. doi: 10.1054/bjoc.2001.2089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Sotiropoulou PA, Perez SA, Hliopoulou EG, Missitzis I, Voelter W, Echner H, Baxevanis CN, Papamichail M. Cytotoxic T-cell precursor frequencies to HER-2 (369-377) in patients with HER-2/neu-positive epithelial tumors. Br J Cancer. 2003;80:1055. doi: 10.1038/sj.bjc.6601244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sotiropoulou PA, Perez SA, Voelter W, Echner H, Missitzis I, Tsavaris NB, Papamichail M, Baxevanis CN. Natural CD8+ T cell responses against MHC class I epitopes of the HER-2/neu oncoprotein in patients with epithelial tumors. Cancer Immunol Immunother. 2003;52:771. doi: 10.1007/s00262-003-0420-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Tajima K, Ito Y, Demachi A, Nishida K, Akatsuka Y, Tsujimura K, Hida T, Morishima Y, Kuwano H, Mitsudomi T, Takahashi T, Kuzushima K. Interferon-gamma differentially regulates susceptibility of lung cancer cells to telomerase-specific cytotoxic T lymphocytes. Int J Cancer. 2004;20:403. doi: 10.1002/ijc.20139. [DOI] [PubMed] [Google Scholar]
  • 95.Tanaka K, Iwamoto S, Gon G, Nohara T, Iwamoto M, Tanigawa N. Expression of survivin and its relationship to loss of apoptosis in breast carcinomas. Clin Cancer Res. 2000;6:127. [PubMed] [Google Scholar]
  • 96.Titu LV, Loveday RL, Maden LA, Cawkwell L, Monson JR, Greenman J. Cytotoxic T-cell immunity against telomerase reverse transcriptase in colorectal cancer patients. Oncol Rep. 2004;12:871. [PubMed] [Google Scholar]
  • 97.Thomas DG, Giordano TJ, Sanders D, Biermann S, Sondak UK, Trent JC, Pollock RE, Baker L. Expression of receptor tyrosine kinases epidermal growth factor receptor and HER-2/neu in synovial sarcoma. Cancer. 2005;103:830. doi: 10.1002/cncr.20847. [DOI] [PubMed] [Google Scholar]
  • 98.Tsai V, Southwood S, Sidney J, Sakaguchi K, Kawakami Y, Appella E, Sette A, Celis E. Identification of subdominant CTL epitopes of the gp100 melanoma-associated tumor antigen by primary in vitro immunization with peptide-pulsed dendritic cells. J Immunol. 1997;158:1796. [PubMed] [Google Scholar]
  • 99.Tuttle TM, Anderson BW, Thompson WE, Lee JE, Sahin A, Smith Tl, Grabstein KH, Wharton T, Ioannides CG, Murray JL. Proliferative and cytokine reponses to class II HER-2/neu-associated peptides in breast cancer. Clin Cancer Res. 1998;4:2015. [PubMed] [Google Scholar]
  • 100.Valmori D, Levy F, Miconnet I, Zajac P, Spagnoli GC, Rimold D, Lienard D, Cerrundolo J, Cerrotini C, Romero P. Induction of potent antitumor CTL responses by recombinant vaccinia encoding a Melan-peptide analogue. J Immunol. 2000;164:1125. doi: 10.4049/jimmunol.164.2.1125. [DOI] [PubMed] [Google Scholar]
  • 101.Vertuani S, Sette A, Sidney J, Southwood S, Fikes J, Keogh E, Lindencrona JA, Ishioka G, Levitskaya J, Kiessling R. Improved immunogenicity of an immunodominant epitope of the HER-2/neu protooncogene by alterations of MHC contact residues. J Immunol. 2004;172:3501. doi: 10.4049/jimmunol.172.6.3501. [DOI] [PubMed] [Google Scholar]
  • 102.Watson MB, Bahia H, Ashman JN, Berrieman HK, Drew P, Lind MJ, Greenman J, Cawkwell L. Chromosomal alterations in breast cancer revealed by multicolour fluorescence in situ hybridization. Int J Oncol. 2004;25:277. doi: 10.3892/ijo.25.2.277. [DOI] [PubMed] [Google Scholar]
  • 103.Wang RF. Human tumor antigens: implications for cancer vaccine development. J Mol Med. 1999;77:640. doi: 10.1007/s001099900042. [DOI] [PubMed] [Google Scholar]
  • 104.Wei YQ, Huang MJ, Yang L, Zhao X, Thian L, Lu Y, Shu JM, Lu CJ, Niu T, Kang B, Mao YQ, et al. Immunogene therapy of tumours with vaccine based on xenopus homologous vascular endothelial growth factor as a model antigen. Proc Natl Acad Sci USA. 2001;98:11545. doi: 10.1073/pnas.191112198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Williams NS, Gaynor RB, Scoggin S, Verna U, Gokaslan T, Simmang C, Flemming J, Tavana D, Frenkel E, Becerra C. Identification and validation of genes involved in the pathogenesis of colorectal cancer using cDNA microarrays and RNA interference. Clin Cancer Res. 2003;9:931. [PubMed] [Google Scholar]
  • 106.Worley BS, van den Broeke IT, Goletz TJ, Pendleton CD, Daschbach EM, Thomas EK, Marincola FM, Helman LJ, Berzolsky JA. Antigenicity of fusion proteins from sarcoma-associated chromosomal translocations. Cancer Res. 2001;61:6868. [PubMed] [Google Scholar]
  • 107.Yi H, Fujimura Y, Ouchida M, Prasad DD, Rao VN, Reddy ES. Inhibition of apoptosis by normal and aberrant Fli-1 and erg proteins involved in human solid tumors and leukemias. Oncogene. 1997;14:1259. doi: 10.1038/sj.onc.1201099. [DOI] [PubMed] [Google Scholar]
  • 108.Yoshimura A, Shiku H, Nakayama E. Rejection of an IA+ variant line of FBL-3 leukemia by cytotoxic T lymphocytes with CD4+ and CD4− CD8+ T cell receptor-αβ phenotypes generated in CD8-depleted C57BL/6 mice. J Immunol. 1993;150:4900. [PubMed] [Google Scholar]
  • 109.Zaks TZ, Rosenberg SA. Immunization with a peptide epitope (p369-377) from HER-2/neu leads to peptide-specific cytotoxic T lymphocytes that fail to recognize HER-2/neu+ tumors. Cancer Res. 1998;58:4902. [PubMed] [Google Scholar]

Articles from Cancer Immunology, Immunotherapy : CII are provided here courtesy of Springer

RESOURCES