182
Views
6
CrossRef citations to date
0
Altmetric
Review

Recent advances in the development of breast cancer vaccines

, , &
Pages 159-168 | Published online: 14 Oct 2014

Abstract

The manipulation of the immune system through the administration of a vaccine to direct an effective and long-lasting immune response against breast cancer (BC) cells is an attractive strategy. Vaccines would have several theoretical advantages over standard therapies, including low toxicities, high specificity, and long-lasting efficacy due to the establishment of immunological memory. However, BC vaccines have failed to demonstrate meaningful results in clinical trials so far. This reflects the intrinsic difficulty in breaking the complex immune-escaping mechanisms developed by cancer cells. New vaccines should be able to elicit complex immunologic response involving multiple immune effectors such as cytotoxic and antibody-secreting B cells, innate immunity effectors, and memory cells. Moreover, especially in patients with large tumor burdens and metastatic disease, combining vaccines with other strategies, such as systemic BC therapies, passive immunotherapy, or immunomodulatory agents, could increase the effectiveness of each approach. Here, we review recent advances in BC vaccines, focusing on suitable targets and innovative strategies. We report results of most recent trials investigating active immunotherapy in BC and provide possible future perspectives in this field of research.

Introduction

Recently, advances in early diagnosis and more effective treatments have reduced the mortality rate due to breast cancer (BC).Citation1 However, despite this progress, BC remains a leading cause of death in the female population worldwide.Citation2 In this scenario, manipulating the immune system to direct an effective and long-term immune response against BC cells through the administration of a vaccine is an attractive strategy. Tumor vaccination would have several theoretical advantages over standard therapies. First, the ideal tumor vaccine would induce potent and durable immune reactions against a broad spectrum of tumor antigens. It could be easily administered and manufactured, with modest side effects typical of conventional chemotherapies. Moreover, it would prevent further tumor recurrences, due to the establishment of persistent immune memory. At present, however, active immunotherapeutic strategies against cancer have failed to fulfill the above expectations in clinical trials.Citation3 This reflects the intrinsic difficulty in finding optimal targets for a cancer vaccine, the most effective type of vaccination, route of administration, and the most immunologically favorable setting of disease (eg, low tumor burden, not heavily pretreated patients). Most importantly, it reflects the difficulty in breaking the complex immune-escaping mechanisms developed by cancer cells.Citation4 The aim of this review is to summarize recent advances in BC active immunotherapy, to address recent results from clinical trials, and to provide possible future perspectives in this field of research.

Targets and strategies of BC vaccines

It has been well established that the immune system plays a role in controlling tumor growth, and adaptive immunity is the main mediator of “spontaneous” regression of certain types of cancers.Citation5,Citation6 The immune system has the ability to recognize several types of antigens expressed on tumor cell surfaces, namely the tumor-associated antigens (TAAs).Citation7 TAAs are presented to immune system effectors such as T-cells by the tumor itself, through the major histocompatibility complex (MHC) or, more likely, by antigen presenting cells (APCs), in particular macrophages and dendritic cells (DCs).Citation7 These cells are essential in processing antigens into immunogenic peptides and presenting them to naive T-cells through the MHC complex. Through a complex and regulated system of co-activator and inhibitory molecules expressed on the cell surface, these cells play an essential role in priming T lymphocytes and activating an immunogenic response against specific targets.Citation8 The presence of tumor-infiltrating lymphocytes has been correlated with better prognosis in several types of cancers.Citation9,Citation10 However, tumor cells often develop the ability to circumvent the surveillance of the immune system.Citation11 In the tumor microenvironment, molecules such as vascular endothelial growth factor, transforming growth factor (TGF)-β, and interleukins are abundant and both actively downregulate the immune function and promote tumor progression, invasion, and metastasis.Citation12 In addition, tumor cells can directly downregulate T-cell function through expression of transmembrane inhibitory molecules such as FasL or B7-H1/PD-L1 or, indirectly, by promoting functionally suppressive CD4+FoxP3+ T lymphocyte (TReg) function.Citation13,Citation14 Finally, cancer cells can modulate expression or mask TAAs, reducing their availability and presentation to immune effectors.Citation15 All these mechanisms can therefore lead to altered DC and T-cell function, and, as final result, to impaired immune response against tumor cells. Immune-escaping mechanisms are particularly active in epithelial cancers such as BC.Citation16 However, some degree of immune response against TAAs can be demonstrated in BC patients.Citation17Citation19 This has prompted researchers to develop active immunotherapies to therapeutically amplify these weak responses against known immunogenic BC antigens. In fact, the aim of an effective therapeutic vaccine is to break peripheral tolerance and activate low-affinity T-cells that were not eliminated during selection in thymus.Citation20 A number of breast TAAs have been recognized and described so far (). Among them, human epithelial growth factor receptor 2 (HER2), carbohydrate antigens, telomerase reverse transcriptase (hTERT), and mucin-1 (MUC-1) have received the greatest attention for vaccine formulationsCitation7 and have been tested in clinical trials. In order to produce an effective vaccine, an antigen or a pool of antigens (as for whole-tumor-cell vaccines) should be delivered through an appropriate formulation. Activation of the immune system could be enhanced by including adjuvant compounds, and appropriate monitoring techniques should be prompted to assess the immunologic response.Citation21 Recently, new technologies such as the use of nanoparticles and liposome formulations, which may improve efficacy of vaccines have received great attention, and preclinical studies with interesting results have been published.Citation22Citation24 Indeed, various vaccine formulations have been tested so far, but none of these was shown to be superior in all situations. Distinct vaccination platforms engage different aspects of the antitumor immune response and each has its own pros and cons, as briefly summarized in .

Table 1 Main breast cancer TAAs

Table 2 Current vaccine platforms

Peptide-based vaccination

Peptide-based vaccines aim at inducing immune responses (including antibodies, cytotoxic T lymphocytes [CTLs], and helper T-cells) using antigenic epitopes derived from TAAs. Many of the first cancer vaccine strategies focused on inducing tumor-specific CD8+ cells with MHC class I restricted short peptides.Citation25Citation27 It is now clear that these CD8+ T-cell responses are typically weak and short-lived.Citation25,Citation28 Further studies have clarified that triggering the CD4+ T-cell response is critical for maximizing tumor immunity, as it both optimizes the CD8+ T-cell response and supports the humoral antitumor immune response.Citation29 Thus, researchers have focused on studying peptide-based vaccines that are able to trigger both CD4+ and CD8+ responses, using longer peptides or mixtures of epitopes.Citation30 Peptide vaccines have several potential advantages, which include easy manufacturing, easily evaluable immunological response, and low expected toxicities. These advantages have made the peptide-based vaccination widely studied and employed in clinical trials. However, this strategy presents some objective limitations. First, to be effective, peptide vaccines often require co-administration of an immunological adjuvant. Adjuvants play an important role in favoring recruitment and efficient stimulation of immune effectors. Identification of an even more efficient adjuvant for a given vaccine is crucial for the effectiveness of the formulation and has been the object of intense research. Second, most of the peptide-based vaccines tested are restricted to HLA-A2. This limits the number of potentially benefiting patients. Third, although easily monitored, immune response is directed against one or a few epitopes, possibly reducing the effectiveness of response and favoring mechanisms of immune escape. Finally, we should consider population-and patient-specific variability in antigen processing and presentation, which could affect the effectiveness of such a strategy.Citation31

DNA-based vaccination

The principle of this approach is based on the assumption that the DNA encoding for a given TAA can be taken by APCs, translated into protein, and finally processed for presentation. DNA can be delivered naked or complexed with other molecules. Frequently, the most used vectors are viruses that are able to efficiently transfect target cells.Citation32,Citation33 Recently, new technologies such as nanoparticles and liposome preparations have been successfully employed to deliver DNA vaccines.Citation34Citation36 A large body of evidence supports the idea that stimulating a coordinated immune response, involving cellular, humoral, and innate immune effectors (natural killer cells and macrophages), most effectively mediates tumor rejection.Citation37,Citation38 DNA vaccines, because of their unique mechanisms of action, could stimulate a more “physiologic” immune response against antigens and could be produced on a larger scale. However, finding an effective vector can be challenging.

Dendritic cells-based vaccination

DCs are the most important APCs. They naturally express high levels of MHC molecules, co-stimulatory proteins, and cytokines.Citation39,Citation40 Autologous DCs can be modified by fusion with cancer cells by pulsing with peptides or by transfection to express tumor antigens.Citation41 DC vaccination represents one of the most intriguing platforms in cancer vaccines. In fact, DCs are able to stimulate both class I and class II responses and can be further modified in order to co-express co-stimulatory molecules, and responses can be directed against multiple targets. This type of platform has been successfully employed and approved for clinical use in castration-resistant prostate cancer.Citation42,Citation43 However, this vaccine platform remains technically challenging due to the uncertainty related to the optimal route of administration and expansion, maturation, and/or activation of DC cells, which is not easily achievable ex vivo and, as a result, this limits larger scale manufacturing.

Whole cells-based vaccination

Another potential approach is immunizing the patient with whole tumor cells, derived from the patient herself (autologous) or from cell-line cultures (allogeneic). These vaccines have been shown to induce antigen-specific T-cell responses. However, more frequently, tumor cells act as antigenic pool for in vivo or ex vivo APCs presentation. To enhance immunological response, tumor cells can be genetically modified to express co-stimulatory molecules or cytokines.Citation44 Theoretical advantages of such approach comprise providing a pool of tumor antigens, generating immune responses to more than one antigen, and thereby possibly overcoming the tumor antigen loss.Citation45 Moreover, this could lead to a more “complex” response, involving both CD4+ and CD8+ T-cells, against different antigens. Potential drawbacks may be the triggering of autoimmunity and difficulties in monitoring the consequent immunologic response that may be directed against unknown TAAs.Citation46

Anti-HER2 vaccines

HER2 is a protein receptor with tyrosine kinase activity belonging to the human epidermal growth factor receptor family overexpressed in 25%–30% of BC.Citation47 HER2 is effectively targeted by a number of new-generation therapies, such as the monoclonal antibodies trastuzumab, pertuzumab, or T-DM1,Citation48Citation50 and small tyrosine kinase inhibitors such as Lapatinib, Neratinib, and Afatinib.Citation51Citation53 HER2 is a suitable target for BC vaccines, and several investigations have shown that some patients develop spontaneous anti-HER2-specific immunity with high levels of both cellular and humoral response.Citation17,Citation54

The largest body of data resulting from anti-HER2 vaccinations derives from clinical trials employing peptide-based vaccines in different clinical settings. The most studied HER2-derived peptide in clinical trials is E75 (HER2 amino acids [aa] 369–377) derived from the extracellular domain of HER2 and characterized by HLA-A2 restriction.Citation55 In the initial studies, E75 was demonstrated to be safe and capable of inducing peptide-specific CD8+ response. The majority of patients enrolled in these pivotal studies had breast or ovarian cancer and were vaccinated with increasing doses of vaccine administered monthly, with up to six immunizations.Citation17,Citation25,Citation26,Citation56,Citation57 In a retrospective analysis aimed at evaluating long-term outcome of BC patients enrolled in these early trials, it was shown that 21 out of 52 (40%) identified patients were alive at the time of analysis (median follow-up [FU] of 112 months).Citation58 Six out of eight (75%) evaluable patients had persistent T-cell immunity versus immunizing HER2 peptides. Moreover, seven out of eight evaluable patients (88%) developed T-cell-specific immunity against HER2 epitopes other than the one used for immunization, likely due to epitope spreading. These data showed that E75 was able to induce effective and long-lasting immunologic responses. Given the encouraging results, E75 was tested in two larger Phase II clinical trials in an adjuvant setting.Citation59 A total of 195 women with HER2 positive (with score ≥1 by immunohistochemistry) were included (100 lymph-node positive, 95 lymph-node negative) after completion of a standard course of surgery, chemotherapy, and radiotherapy.Citation60 The final results of this study have been very recently reported.Citation61 The vaccine was administered with granulocyte-macrophage colony-stimulating factor (GM-CSF) for up to six immunizations, and 49% of patients received booster inoculations. At 5 years, the disease-free survival (DFS) rate for vaccinated patients was 89.7% versus 80.2% for controls (P=0.08).Citation62 Although not statistically significant, vaccination was associated with a 48% reduction of relative risk of recurrence. Interestingly, among the node-positive population, benefits of vaccination were shown to be larger. In fact, the 24-month DFS rate was 90.2% for vaccinated patients and0 79.1% for controls (P=0.13), with a relative reduction of risk for recurrence of 53%. Interestingly, patients expressing low levels of HER2 (immunohistochemistry 1 or 2+) had more robust immunologic response,Citation63 and in this population the DFS rate was improved at 5 years of FU,Citation64 with the DFS rate of 88.1% in vaccinated patients versus 77.5% in controls (P=0.16). In contrast, in patients whose tumors overexpressed HER2, the DFS rate was 90.3% for vaccinated patients versus 83.3% for controls (P=0.44). Of 30 vaccinated patients who had HER2-overexpressing tumors, only 12 patients received trastuzumab before vaccination, and none of these patients had recurrences. Because of the trial design, not all patients received the vaccine dose that was subsequently determined to be optimal. Moreover, a booster inoculation was administered in 53 patients (49%). Intriguingly, among the patients who received booster inoculations the DFS rate at 5 years was 95.2% (P=0.11 vs controls).

AE37 is an HER2 (aa 776–779)-derived peptide modified with a “li-Key” motif in order to elicit a combined CD4+ T- and CD8+ T-cell response.Citation28 This vaccine formulation was tested at three different doses with or without GM-CSF in 15 patients with node-negative BC, in remission after standard therapy. Intriguingly, without adjuvants, AE37 was able to effectively elicit a specific combined response.Citation65Citation67

Another HER2-derived peptide tested in clinical trials is GP2. This is a nine-aa, HLA-A2-restricted peptide derived from the transmembrane domain of HER2 (aa 654–662), with a lower affinity to HLA-A2.Citation68 Preliminary results of a Phase II trial of the GP2 vaccine with GM-CSF were recently presented.Citation69 In this trial, 172 HLA-A2-positive newly diagnosed women with node-positive or high-risk node-negative disease after standard therapy were enrolled. GP2 was shown to be able to elicit strong immunologic response in the vaccinated group, and, although not statistically significant, the immune response correlated with a >50% reduction in BC recurrences (4.3% vs 11.6% in the vaccine and control group, respectively; P=0.41).

Results of the clinical trials of anti-HER2 vaccines, using different types of platforms, are summarized in .Citation70Citation73

Table 3 Clinical trials with anti-HER2 vaccinations

Anti-MUC-1 and anti-CEA vaccines

MUC-1 is a membrane glycoprotein expressed by many types of epithelial cells, including breast, lung, and the gastrointestinal tract.Citation74 In cancer cells, it is overexpressed and aberrantly glycosylated, and involved in cell-to-cell and cell-to-matrix adhesion, in signal transduction, and in modulation of the immune system. Nearly 70% of cancers overexpress MUC-1.Citation74,Citation75 It has been demonstrated that MUC-1 can be immunogenic and could be a suitable target for cancer immunotherapy.Citation76Citation78

A series of pilot studies using anti-unglycosylated MUC-1 peptide-based vaccination were undertaken, with conflicting results.Citation79Citation83 Further studies showed that directing immunologic response against glycosylated form of MUC-1 could translate into stronger humoral responses. An anti-MUC-1 vaccination conjugated with keyhole limpet hemocyanin (KLH) was shown to elicit strong humoral response and to be safe.Citation84Citation86 Based on these results, Miles et al conducted a Phase III trial in 1,028 women with metastatic BC who had previously received chemotherapy and had had either a complete or a partial response, or no disease progression.Citation86 All women received T-Reg-depleting doses of cyclophosphamide (300 mg/m2) 3 days before vaccine injection. Although the treatment was well tolerated and able to induce strong humoral response, no DFS or overall survival (OS) benefit emerged.Citation3 However, in patients treated with concomitant hormone therapy (34%), a trend toward improved time to progression and OS was observed. This observation suggests that combined therapy with anti-estrogens and anti-MUC-1 could be beneficial.

It was shown that most of the immunogenicity of MUC-1 resided in a repeated 20-aa peptide domain in the extracellular portion of the protein. This peptide, fused with oxidized mannan (M-FP) in an attempt to improve APC presentation, was able to induce both cytotoxic and humoral responses.Citation87Citation90 Hence, a pilot Phase III study in early stage BC patients using oxidized mannan–MUC-1 was undertaken.Citation91 In this small, placebo-controlled, randomized study, 31 patients with stage II and no more than four lymph nodes involved were injected with the vaccine after surgery, radiotherapy, and commencement of the adjuvant Tamoxifen. At 15 years of FU, the recurrence rate in patients receiving placebo was 60% (9 out of 15), whereas in those receiving immunotherapy it was 12.5% (2 out of 16) (P=0.002).Citation92 All patients injected with M-FP showed no evidence of toxic effects or signs of autoimmunity during the 12–15-year FU. No response to MUC-1 was seen in the patients treated with placebo, whereas 9 of 13 patients immunized with MUC-1 had developed MUC-1-specific antibodies and 4 of 10 patients MUC-1-specific T-cell responses. The clinical results obtained with this strategy are very promising, and further larger studies are warranted to confirm these results.

Carcinoembryonic antigen (CEA) is another glycoprotein aberrantly expressed in a multitude of cancer cell types, such as colorectal, lung, or breast cancer.Citation93 It is a member of adhesion molecules, and its overexpression in cancer cells promotes their adhesion and the metastatic process.Citation94 PANVAC™ is a new cancer vaccine targeting both CEA and MUC-1. The vaccine is delivered through two sequential viral vectors, recombinant vaccinia (rV−) and recombinant fowlpox (rF−) – given in sequence, containing transgenes for the targets and for three human T-cell co-stimulatory molecules (TRICOM [TRIad of COstimulatory Molecules]) to enhance immune system activation. The PANVAC vaccines are injected subcutaneously and processed by APCs.Citation95 In a pilot study, 26 heavily pretreated patients with ovarian cancer or BC were enrolled. Side effects were largely limited to mild injection-site reactions. For the 12 BC patients enrolled, the median time to progression was 2.5 months and the median OS was 13.7 months. One patient had a complete response and remained on study for 37 months and another had partial response, whereas four patients had stable disease. Patient with fewer previous lines of treatment or low tumor burden were more likely to respond to the vaccine.Citation96 In another pilot study, 25 patients with advanced metastatic carcinoma were treated with the vaccine. Two patients with BC were enrolled, one had a confirmed decrease of >20% in the size of large liver metastasis, whereas both had long-duration disease stabilization.Citation97 These preliminary results show promising activity of this vaccine in BC, and further larger studies are currently going on.

Anti-hTERT vaccines

hTERT is another potential target for cancer vaccination. Telomerase is a ribonucleoprotein complex that maintains chromosomal integrity by protecting telomeric DNA for continuous cell proliferation.Citation98 The complex contains hTERT and an RNA template. hTERT has broad expression in cancer cells, with little or no expression in normal somatic cells. A first pivotal study in 7 HLA-A2 advanced breast or prostate cancer patients showed good tolerability and promising clinical efficacy. Patients enrolled in this study were vaccinated with autologous monocyte-derived DCs pulsed ex vivo with hTERT I540 peptide and KLH as adjuvant. Among the two patients with BC, one showed a mixed response of skin lesions, previously progressed despite chemo, hormonal, and radiotherapy.Citation99 Another study was conducted on 19 HLA-A2-positive patients with metastatic BC who were refractory to conventional therapy. Following vaccination, a specific CD8+ T-cell response was seen in more than 50% of the patients. Despite this, no objective responses were shown. However, in a post hoc analysis, it was shown that the median OS was significantly greater among patients with high CD8+ T-cell response than among low or no responders (32.2 months vs 17.5 months, P=0.03).Citation100

Combining BC vaccines with other strategies

Despite the encouraging preliminary results and excellent profile of tolerability, BC vaccines still show limited clinical efficacy. Antigen variability and mechanisms of tumor immune-escaping can impair the effectiveness of active immunization. Moreover, possible difficulties of the immune effectors to reach poorly vascularized tumors and high tumor burdens may contribute to limit the efficiency of vaccines. For patients with larger burdens of tumor and disseminated disease, it is fairly clear that vaccines alone are not able to outmatch the immune tolerance mechanisms of cancer cells; moreover, these become progressively more complex with tumor progression. Thus, a possible way to overcome the known limits of active immunotherapy may be combining BC vaccines with other strategies, such as systemic BC therapies, passive immunotherapy, or immunomodulatory agents. Interaction of BC vaccines and systemic therapy could be complex and poorly predictable. Thus, any combinatorial strategy requires strong biologic rationale. For example, target-immune checkpoints and reducing activity of T-Regs could overcome immune tolerance and increase the effectiveness of vaccination. Especially for patients with advanced disease, incorporating drugs that target BC-biology-inhibiting key intracellular signaling pathway may be required to enhance the activity of vaccines. In this field, results of first pivotal studies incorporating BC vaccines with targeted therapies have already been reported. Finally, disruption of tumor cells by conventional therapies could lead to the release of tumor fragments/antigens that are otherwise not accessible for presentation and processing and proinflammatory cytokines, with the final result of an increased immune response.

The humanized anti-HER2 monoclonal antibody trastuzumab has been combined with an HER2 peptide-based vaccine in a Phase I/II study in 22 pretreated patients with HER2 overexpressing stage IV BC.Citation101 The association treatment was well tolerated and did not result in additional toxic effects. At a median FU of 36 months, the median progression free survival was 17.7 months (33% proportion of patients without events at 3 years). This finding favorably compares with the results obtained in similar settings by conventional treatments.

Patil et alCitation102 recently reported preliminary results of a Phase I study of trastuzumab in combination with GP2 vaccine plus GM-CSF in an adjuvant setting. Nineteen patients were treated. The combination was shown to be safe, with only mild reaction at the site of injection. No adverse cardiac events were reported. The vaccine was shown to be highly immunogenic, and further studies on this combination are warranted.

Trastuzumab has also been safely combined with an HER2-positive, GM-CSF-secreting, allogeneic breast tumor-cell vaccine. Twenty-two patients with HER2-positive metastatic BC were enrolled in this study. No dose-limiting toxic effects were observed. Clinical benefit rates (complete responses + partial responses + stable disease) at 6 and 12 months were 50% (95% CI, 27%–72%) and 35% (95% CI, 15%–59%), respectively.Citation103

A plasmid DNA encoding HER-2/neu together with low doses of GM-CSF and IL-2 and concurrent trastuzumab has been tested in eight patients with metastatic HER2-positive BC. The study was conducted by Norell et al.Citation104 No relevant toxic effects were observed. The treatment was shown to induce immediate and strong antibody production alongside long-lasting T-cell response. Notably, two out of six patients who completed all three vaccination cycles were long-term survivors, still alive more than 4 years after the last vaccination.

Finally, Hamilton et alCitation105 published interesting results of a small pilot trial using recombinant anti-HER2 protein vaccine with concurrent Lapatinib in 12 women with trastuzumab-refractory HER2 overexpressing metastatic BC. No unexpected adverse events were shown. Specific anti-HER2 humoral response was induced in all patients. Interestingly, OS at 300 days was 92% (95% CI, 77%–100%).

Conclusion and future perspectives

In recent years, outstanding progress has been achieved toward the cure of BC. More personalized therapies, molecularly targeted drugs, and a deeper understanding of the mechanisms of disease have allowed improving the prognosis of certain subtypes of tumor. In this rapidly changing scenario, there is a growing interest in developing an effective cancer vaccine. Unfortunately, none of the vaccine tested so far in clinical trials has turned out to be “practice changing.” Nevertheless, three important lessons can be drawn.

First, many vaccines elicit a measurable immunologic response, such as specific antibodies or specific CD8+ T-cells, but this response often has little or no impact on tumor growth. Engaging only one compartment of the immune system (eg, only cytotoxic response or humoral response) is probably not sufficient for an effective therapeutic vaccine. New vaccination strategies should therefore aim at eliciting a wide response, involving multiple immune effectors such as cytotoxic and antibody-secreting B-cells, innate immunity effectors, and memory cells. The underlying concept would be that a “complete” immunologic response may promote increased release of tumor cell fragments/antigens and proinflammatory cytokines, resulting in an immunologic virtuous cycle.

Second, the main barrier against vaccination is probably due to complex immuno-escaping mechanisms developed by cancer cells. Regulatory cells such as T-Regs and molecular immune checkpoints (eg, CTLA-4, PD1/PD1L) play crucial roles in maintaining self-tolerance, and tumors are able to exploit these elements to get protection from immune system’s attack. New strategies based on blocking antibodies, recombinant forms of ligands, or receptors should be implemented to block such modulatory checkpoints and strengthen the immune response, with promising initial translation into clinical setting. One of the most intriguing perspectives of these strategies is obviously their synergism with immunotherapy approaches such as cancer vaccines.

Third, patients with large tumor burden and very advanced disease, enrolled in most of the clinical trials on cancer vaccines, are also those who benefit less from cancer vaccines alone. In fact, as cancer progresses and spreads in spite of multiple lines of treatment, immune-tolerance mechanisms become more intricate and the immune system is less likely to counteract the tumor. Therefore, in these patients, combining vaccine with established drugs targeting cancer biology, such as endocrine therapy, tyrosine receptor inhibitors, or chemotherapy, is required to achieve satisfactory clinical results. In conclusion, in order to maximize the likelihood of success, new BC vaccines should be developed by integrating a thorough understanding of immune tolerance mechanisms and tested in well-designed clinical trials conducted in immunologically favorable settings. Moreover, an additional effort should be made to improve immunotherapy in the specific basal-like subtype, which requires novel therapeutic strategies more than luminal and HER2-positive types.

Disclosure

The authors have no conflicts of interest to declare.

Acknowledgments

This work has been funded by Grant GR-2009-1543842 and AIRC IG-2011 Grant no 11515.

References

  • WinkfieldKMHarrisJREffective local therapy and long-term survival in breast cancerOncology200923866967519711579
  • SiegelRNaishadhamDJemalACancer statistics, 2013CA Cancer J Clin2013631113023335087
  • MilesDRochéHMartinMPhase III multicenter clinical trial of the sialyl-TN (STn)-keyhole limpet hemocyanin (KLH) vaccine for metastatic breast cancerOncologist20111681092110021572124
  • EmensLABreast cancer immunobiology driving immunotherapy: vaccines and immune checkpoint blockadeExpert Rev Anticancer Ther201212121597161123253225
  • BurnetFMImmunological aspects of malignant diseaseLancet196717501117111744165129
  • ShankaranVIkedaHBruceATIFNgamma and lymphocytes prevent primary tumour development and shape tumour immunogenicityNature200141068321107111111323675
  • AndersonKSTumor vaccines for breast cancerCancer Invest200927436136819358018
  • WilcoxRATamadaKFliesDBLigation of CD137 receptor prevents and reverses established anergy of CD8+ cytolytic T lymphocytes in vivoBlood2004103117718412969968
  • HakanssonAGustafssonBKrysanderLHakanssonLTumour-infiltrating lymphocytes in metastatic malignant melanoma and response to interferon alpha treatmentBr J Cancer19967456706768845294
  • YasumotoKTakeoSYanoTRole of tumor-infiltrating lymphocytes in the host defense mechanism against lung cancerJ Surg Oncol19883842212262842545
  • RaviRFuchsEJJainAResistance of cancers to immunologic cytotoxicity and adoptive immunotherapy via X-linked inhibitor of apoptosis protein expression and coexisting defects in mitochondrial death signalingCancer Res20066631730173916452233
  • ZouWImmunosuppressive networks in the tumour environment and their therapeutic relevanceNat Rev Cancer20055426327415776005
  • MuschenMMoersCWarskulatUEvenJNiederacherDBeckmannMWCD95 ligand expression as a mechanism of immune escape in breast cancerImmunology2000991697710651943
  • WolfAMWolfDSteurerMGastlGGunsiliusEGrubeck-LoebensteinBIncrease of regulatory T-cells in the peripheral blood of cancer patientsClin Cancer Res20039260661212576425
  • RikerACormierJPanelliMImmune selection after antigen-specific immunotherapy of melanomaSurgery1999126211212010455872
  • MarchiniCKalogrisCGarulliCTailoring DNA vaccines: designing strategies against HER2-positive cancersFront Oncol2013312223675574
  • DisisMLCalenoffEMcLaughlinGExistent T-cell and antibody immunity to HER-2/neu protein in patients with breast cancerCancer Res199454116207505195
  • DisisMLPupaSMGralowJRDittadiRMenardSCheeverMAHigh-titer HER-2/neu protein-specific antibody can be detected in patients with early-stage breast cancerJ Clin Oncol19971511336333679363867
  • MilesDPapazisisKRationale for the clinical development of STn-KLH (Theratope) and anti-MUC-1 vaccines in breast cancerClin Breast Cancer20033Suppl 4S134S13812620150
  • HsiehCSLeeHMLioCWSelection of regulatory T cells in the thymusNat Rev Immunol201212315716722322317
  • CuriglianoGCriscitielloCEspositoADeveloping an effective breast cancer vaccine: challenges to achieving sterile immunity versus resetting equilibriumBreast201322Suppl 2S96S9924074802
  • PenwellASharpKMansourMSammaturLDevelopment and validation of an HPLC/UV assay for separation and quantification of peptide antigens from a liposomal vaccine delivery platformJ Pharm Biomed Anal20126617618222516680
  • MarracheSTundupSHarnDADharSEx vivo programming of dendritic cells by mitochondria-targeted nanoparticles to produce interferon-gamma for cancer immunotherapyACS Nano2013787392740223899410
  • PouyanfardSBamdadTHashemiHBandehpourMKazemiBInduction of protective anti-CTL epitope responses against HER-2-positive breast cancer based on multivalent T7 phage nanoparticlesPLoS One2012711e4953923166703
  • KnutsonKLSchiffmanKCheeverMADisisMLImmunization of cancer patients with a HER-2/neu, HLA-A2 peptide, p369-377, results in short-lived peptide-specific immunityClin Cancer Res2002851014101812006513
  • KnutsonKLSchiffmanKDisisMLImmunization with a HER-2/neu helper peptide vaccine generates HER-2/neu CD8 T-cell immunity in cancer patientsJ Clin Invest2001107447748411181647
  • ZaksTZRosenbergSAImmunization with a peptide epitope (p369-377) from HER-2/neu leads to peptide-specific cytotoxic T lymphocytes that fail to recognize HER-2/neu+ tumorsCancer Res19985821490249089809997
  • MittendorfEAHolmesJPMurrayJLvon HofeEPeoplesGECD4+ T cells in antitumor immunity: utility of an li-key HER2/neu hybrid peptide vaccine (AE37)Expert Opin Biol Ther200991717819063694
  • PardollDMTopalianSLThe role of CD4+ T cell responses in antitumor immunityCurr Opin Immunol19981055885949794842
  • ZwavelingSFerreira MotaSCNoutaJEstablished human papillomavirus type 16-expressing tumors are effectively eradicated following vaccination with long peptidesJ Immunol2002169135035812077264
  • MilaniASangioloDMontemurroFAgliettaMValabregaGActive immunotherapy in HER2 overexpressing breast cancer: current status and future perspectivesAnn Oncol20132471740174823585514
  • GalloPDharmapuriSNuzzoMXenogeneic immunization in mice using HER2 DNA delivered by an adenoviral vectorInt J Cancer20051131677715386429
  • Prud’hommeGJDNA vaccination against tumorsJ Gene Med20057131715538731
  • HuiKMAngPTHuangLTaySKPhase I study of immunotherapy of cutaneous metastases of human carcinoma using allogeneic and xenogeneic MHC DNA-liposome complexesGene Ther1997487837909338006
  • PecherGSpahnGSchirrmannTMucin gene (MUC1) transfer into human dendritic cells by cationic liposomes and recombinant adenovirusAnticancer Res2001214A2591259611724326
  • LiuZLvDLiuSAlginic acid-coated chitosan nanoparticles loaded with legumain DNA vaccine: effect against breast cancer in micePLoS One201384e6019023577091
  • CurcioCDi CarloEClynesRNonredundant roles of antibody, cytokines, and perforin in the eradication of established Her-2/neu carcinomasJ Clin Invest200311181161117012697735
  • SpadaroMAmbrosinoEIezziMCure of mammary carcinomas in Her-2 transgenic mice through sequential stimulation of innate (neoadjuvant interleukin-12) and adaptive (DNA vaccine electroporation) immunityClin Cancer Res20051151941195215756020
  • RohrbachFWethRKursarMSlootsAMittruckerHWWelsWSTargeted delivery of the ErbB2/HER2 tumor antigen to professional APCs results in effective antitumor immunityJ Immunol200517495481548915843546
  • NabekuraTNagasawaTNakauchiHOnoderaMAn immunotherapy approach with dendritic cells genetically modified to express the tumor-associated antigen, HER2Cancer Immunol Immunother200857561162217786440
  • ZhangYMaBZhouYDendritic cells fused with allogeneic breast cancer cell line induce tumor antigen-specific CTL responses against autologous breast cancer cellsBreast Cancer Res Treat2007105327728617187233
  • KantoffPWHiganoCSShoreNDSipuleucel-T immunotherapy for castration-resistant prostate cancerN Engl J Med2010363541142220818862
  • SmallEJSchellhammerPFHiganoCSPlacebo-controlled Phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancerJ Clin Oncol200624193089309416809734
  • RossiGRMautinoMRAwwadDZAllogeneic melanoma vaccine expressing alphaGal epitopes induces antitumor immunity to autologous antigens in mice without signs of toxicityJ Immunother200831654555418528300
  • KeenanBPJaffeeEMWhole cell vaccines – past progress and future strategiesSemin Oncol201239327628622595050
  • CuriglianoGSpitaleriGPietriEBreast cancer vaccines: a clinical reality or fairy tale?Ann Oncol200617575076216293674
  • SlamonDJGodolphinWJonesLAStudies of the HER-2/neu proto-oncogene in human breast and ovarian cancerScience198924449057077122470152
  • SlamonDJLeyland-JonesBShakSUse of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2N Engl J Med20013441178379211248153
  • BaselgaJCortesJKimSBPertuzumab plus trastuzumab plus docetaxel for metastatic breast cancerN Engl J Med2012366210911922149875
  • VermaSMilesDGianniLTrastuzumab emtansine for HER2-positive advanced breast cancerN Engl J Med2012367191783179123020162
  • GeyerCEForsterJLindquistDLapatinib plus capecitabine for HER2-positive advanced breast cancerN Engl J Med2006355262733274317192538
  • AwadaADirixLManso SanchezLSafety and efficacy of neratinib (HKI-272) plus vinorelbine in the treatment of patients with ErbB2-positive metastatic breast cancer pretreated with anti-HER2 therapyAnn Oncol201324110911622967996
  • GeunaEMontemurroFAgliettaMValabregaGPotential of afatinib in the treatment of patients with HER2-positive breast cancerBreast Cancer2012413113724367201
  • PeoplesGEGoedegebuurePSSmithRLinehanDCYoshinoIEberleinTJBreast and ovarian cancer-specific cytotoxic T lymphocytes recognize the same HER2/neu-derived peptideProc Natl Acad Sci USA19959224324367831305
  • FiskBBlevinsTLWhartonJTIoannidesCGIdentification of an immunodominant peptide of HER-2/neu protooncogene recognized by ovarian tumor-specific cytotoxic T lymphocyte linesJ Exp Med19951816210921177539040
  • DisisMLGooleyTARinnKGeneration of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccinesJ Clin Oncol200220112624263212039923
  • MurrayJLGilloglyMEPrzepiorkaDToxicity, immunogenicity, and induction of E75-specific tumor-lytic CTLs by HER-2 peptide E75 (369-377) combined with granulocyte macrophage colony-stimulating factor in HLA-A2+ patients with metastatic breast and ovarian cancerClin Cancer Res20028113407341812429628
  • SalazarLGGoodellVO’MearaMPersistent immunity and survival after immunization with a HER2/neu (HER2) vaccineASCO Meeting Abstracts2009Orlando, FL
  • PeoplesGEGurneyJMHuemanMTClinical trial results of a HER2/neu (E75) vaccine to prevent recurrence in high-risk breast cancer patientsJ Clin Oncol200523307536754516157940
  • PeoplesGEHolmesJPHuemanMTCombined clinical trial results of a HER2/neu (E75) vaccine for the prevention of recurrence in high-risk breast cancer patients: US Military Cancer Institute Clinical Trials Group Study I-01 and I-02Clin Cancer Res200814379780318245541
  • MittendorfEACliftonGTHolmesJPFinal report of the Phase I/II clinical trial of the E75 (nelipepimut-S) vaccine with booster inoculations to prevent disease recurrence in high-risk breast cancer patientsAnnals of Oncology20142591735174224907636
  • AminABenavidesLCHolmesJPAssessment of immunologic response and recurrence patterns among patients with clinical recurrence after vaccination with a preventive HER2/neu peptide vaccine: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02Cancer Immunol Immunother200857121817182518392824
  • BenavidesLCGatesJDCarmichaelMGThe impact of HER2/neu expression level on response to the E75 vaccine: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02Clin Cancer Res20091582895290419351776
  • MittendorfEACliftonGTHolmesJPClinical trial results of the HER-2/neu (E75) vaccine to prevent breast cancer recurrence in high-risk patients: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02Cancer2012118102594260221989902
  • HolmesJPBenavidesLCGatesJDResults of the first Phase I clinical trial of the novel II-key hybrid preventive HER-2/neu peptide (AE37) vaccineJ Clin Oncol200826203426343318612158
  • HaleDFPerezSAVreelandTJAn assessment of disease features and immune response in breast cancer patients that did not recur after receiving HER2 peptide, AE37 vaccine in a randomized Phase II trialASCO Meeting Abstracts2012Chicago, IL
  • AnastasopoulouEAPappouETzonisPBooster inoculations of the AE37 peptide vaccine enhance immunological responses in a Phase II studyASCO Meeting Abstracts201331Suppl 153095
  • CarmichaelMGBenavidesLCHolmesJPResults of the first Phase 1 clinical trial of the HER-2/neu peptide (GP2) vaccine in disease-free breast cancer patients: United States Military Cancer Institute Clinical Trials Group Study I-04Cancer2010116229230119924797
  • TrappeyFBerryJSVreelandTJRandomized Phase II clinical trial of the anti-HER2 (GP2) vaccine to prevent recurrence in high-risk breast cancer patients: a planned interim analysisASCO Meeting Abstracts201331Suppl 153005
  • DisisMLSchiffmanKGuthrieKEffect of dose on immune response in patients vaccinated with an her-2/neu intracellular domain protein – based vaccineJ Clin Oncol200422101916192515143085
  • GuardinoACassidyMPienkowskiTResults of two Phase I clinical trials of MVA-BN(R)-HER2 in HER-2 overexpressing metastatic breast cancer patientsCancer Res200969245089
  • LimentaniSDorvalTWhiteSPhase I dose-escalation trial of a recombinant HER2 vaccine in patients with stage II/III HER2+ breast cancerASCO Meeting Abstracts2005Chicago, IL
  • EmensLAAsquithJMLeathermanJMTimed sequential treatment with cyclophosphamide, doxorubicin, and an allogeneic granulocyte-macrophage colony-stimulating factor-secreting breast tumor vaccine: a chemotherapy dose-ranging factorial study of safety and immune activationJ Clin Oncol200927355911591819805669
  • YangEHuXFXingPXAdvances of MUC1 as a target for breast cancer immunotherapyHistol Histopathol200722890592217503348
  • TarpMAClausenHMucin-type O-glycosylation and its potential use in drug and vaccine developmentBiochim Biophys Acta20081780354656317988798
  • ApostolopoulosVXingPXMcKenzieIFMurine immune response to cells transfected with human MUC1: immunization with cellular and synthetic antigensCancer Res19945419518651937923138
  • ZhangSGraeberLAHellingFAugmenting the immunogenicity of synthetic MUC1 peptide vaccines in miceCancer Res19965614331533198764127
  • AcresRBHareuveniMBalloulJMKienyMPVaccinia virus MUC1 immunization of mice: immune response and protection against the growth of murine tumors bearing the MUC1 antigenJ Immunother Emphasis Tumor Immunol19931421361438280702
  • GoydosJSElderEWhitesideTLFinnOJLotzeMTA Phase I trial of a synthetic mucin peptide vaccine. Induction of specific immune reactivity in patients with adenocarcinomaJ Surg Res19966312983048667619
  • RamanathanRKLeeKMMcKolanisJPhase I study of a MUC1 vaccine composed of different doses of MUC1 peptide with SB-AS2 adjuvant in resected and locally advanced pancreatic cancerCancer Immunol Immunother200554325426415372205
  • AdluriSGilewskiTZhangSRamnathVRagupathiGLivingstonPSpecificity analysis of sera from breast cancer patients vaccinated with MUC1-KLH plus QS-21Br J Cancer19997911–121806181210206297
  • GilewskiTAdluriSRagupathiGVaccination of high-risk breast cancer patients with mucin-1 (MUC1) keyhole limpet hemocyanin conjugate plus QS-21Clin Cancer Res2000651693170110815887
  • PalmerMParkerJModiSPhase I study of the BLP25 (MUC1 peptide) liposomal vaccine for active specific immunotherapy in stage IIIB/IV non-small-cell lung cancerClin Lung Cancer2001314957 discussion 5814656392
  • MacLeanGDReddishMKogantyRRImmunization of breast cancer patients using a synthetic sialyl-Tn glycoconjugate plus Detox adjuvantCancer Immunol Immunother19933642152228439984
  • ReddishMAMacLeanGDPoppemaSBergALongeneckerBMPre-immunotherapy serum CA27.29 (MUC-1) mucin level and CD69+ lymphocytes correlate with effects of Theratope sialyl-Tn-KLH cancer vaccine in active specific immunotherapyCancer Immunol Immunother19964253033098706053
  • MilesDWTowlsonKEGrahamRA randomised Phase II study of sialyl-Tn and DETOX-B adjuvant with or without cyclophosphamide pretreatment for the active specific immunotherapy of breast cancerBr J Cancer1996748129212968883420
  • XingPMichaelMApostolopoulosVPhase-I study of synthetic muc1 peptides in breast-cancerInt J Oncol1995661283128921556671
  • KaranikasVHwangLAPearsonJAntibody and T cell responses of patients with adenocarcinoma immunized with mannan-MUC1 fusion proteinJ Clin Invest199710011278327929389743
  • KaranikasVLoddingJMainoVCMcKenzieIFFlow cytometric measurement of intracellular cytokines detects immune responses in MUC1 immunotherapyClin Cancer Res20006382983710741704
  • KaranikasVThynneGMitchellPMannan mucin-1 peptide immunization: influence of cyclophosphamide and the route of injectionJ Immunother2001242172183
  • ApostolopoulosVPieterszGATsibanisAPilot Phase III immunotherapy study in early-stage breast cancer patients using oxidized mannan-MUC1 [ISRCTN71711835]Breast Cancer Res200683R2716776849
  • VassilarosSTsibanisATsikkinisAPieterszGAMcKenzieIFApostolopoulosVUp to 15-year clinical follow-up of a pilot Phase III immunotherapy study in stage II breast cancer patients using oxidized mannan-MUC1Immunotherapy20135111177118224188672
  • TurrizianiMFantiniMBenvenutoMCarcinoembryonic antigen (CEA)-based cancer vaccines: recent patents and antitumor effects from experimental models to clinical trialsRecent Pat Anticancer Drug Discov20127326529622630596
  • CuriglianoGRescignoMGoldhirschAImmunology and breast cancer: therapeutic cancer vaccinesBreast200716Suppl 2S20S2617706425
  • MadanRAArlenPMGulleyJLPANVAC-VF: poxviral-based vaccine therapy targeting CEA and MUC1 in carcinomaExpert Opin Biol Ther20077454355417373905
  • MohebtashMTsangKYMadanRAA pilot study of MUC-1/CEA/TRICOM poxviral-based vaccine in patients with metastatic breast and ovarian cancerClin Cancer Res201117227164717322068656
  • GulleyJLArlenPMTsangKYPilot study of vaccination with recombinant CEA-MUC-1-TRICOM poxviral-based vaccines in patients with metastatic carcinomaClin Cancer Res200814103060306918483372
  • CovelerALBatesNEDisisMLProgress in the development of a therapeutic vaccine for breast cancerBreast Cancer20102253624367164
  • VonderheideRHDomchekSMSchultzeJLVaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytesClin Cancer Res200410382883914871958
  • DomchekSMRecioAMickRTelomerase-specific T-cell immunity in breast cancer: effect of vaccination on tumor immunosurveillanceCancer Res20076721105461055517974999
  • DisisMLWallaceDRGooleyTAConcurrent trastuzumab and HER2/neu-specific vaccination in patients with metastatic breast cancerJ Clin Oncol200927284685469219720923
  • PatilRCliftonGTLittonJKSafety and efficacy of the HER2-derived GP2 peptide vaccine in combination with trastuzumab for breast cancer patients in the adjuvant settingASCO Meeting Abstracts201331Suppl 153096
  • EmensLAGuptaRPetrikSA feasibility study of combination therapy with trastuzumab (T), cyclophosphamide (CY), and an allogeneic GM-CSF-secreting breast tumor vaccine for the treatment of HER2+ metastatic breast cancerASCO Meeting Abstracts2011Chicago, IL
  • NorellHPoschkeICharoJVaccination with a plasmid DNA encoding HER-2/neu together with low doses of GM-CSF and IL-2 in patients with metastatic breast carcinoma: a pilot clinical trialJ Transl Med201085320529245
  • HamiltonEBlackwellKHobeikaACPhase I clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibitionJ Transl Med2012102822325452