1,304
Views
74
CrossRef citations to date
0
Altmetric
Review

Trial watch

Peptide vaccines in cancer therapy

, , , , , , , , & show all
Pages 1557-1576 | Published online: 06 Dec 2012

Abstract

Prophylactic vaccination constitutes one of the most prominent medical achievements of history. This concept was first demonstrated by the pioneer work of Edward Jenner, dating back to the late 1790s, after which an array of preparations that confer life-long protective immunity against several infectious agents has been developed. The ensuing implementation of nation-wide vaccination programs has de facto abated the incidence of dreadful diseases including rabies, typhoid, cholera and many others. Among all, the most impressive result of vaccination campaigns is surely represented by the eradication of natural smallpox infection, which was definitively certified by the WHO in 1980. The idea of employing vaccines as anticancer interventions was first theorized in the 1890s by Paul Ehrlich and William Coley. However, it soon became clear that while vaccination could be efficiently employed as a preventive measure against infectious agents, anticancer vaccines would have to (1) operate as therapeutic, rather than preventive, interventions (at least in the vast majority of settings), and (2) circumvent the fact that tumor cells often fail to elicit immune responses. During the past 30 y, along with the recognition that the immune system is not irresponsive to tumors (as it was initially thought) and that malignant cells express tumor-associated antigens whereby they can be discriminated from normal cells, considerable efforts have been dedicated to the development of anticancer vaccines. Some of these approaches, encompassing cell-based, DNA-based and purified component-based preparations, have already been shown to exert conspicuous anticancer effects in cohorts of patients affected by both hematological and solid malignancies. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating purified peptides or full-length proteins as therapeutic interventions against cancer.

Introduction

Jenner’s pioneering observations

Edward Anthony Jenner (1749–1823) was an English physician nowadays considered by many as the father of modern immunology.Citation1,Citation2 In the 1790s, Jenner, who beyond medicine cultivated various interests spanning from natural history to air balloons, was practicing as a family doctor and surgeon in Berkeley (Gloucestershire), the small town he was born in some 40 y earlier. In that period, Jenner was particularly intrigued by the observation that milkmaids were generally immune to smallpox, and he postulated that such a protection would be conferred by the pus contained in blisters that milkmaids developed along with cowpox (a disease similar to, yet much less virulent than, smallpox).Citation1,Citation2 In 1796, to test his hypothesis, Jenner inoculated 8-y-old James Phipps with pus that he had scraped from the blisters of a cowpox-affected milkmaid. Sometimes later, Jenner challenged James Phipps with variolous material, i.e., material obtained from a smallpox pustule of a selected mild case (supposedly affected by the relatively less virulent Variola minor smallpox virus). The boy developed no signs of disease, nor did he after a further similar inoculation performed a few weeks later. Jenner pursued his investigations on additional 22 cases and then reported his findings to the Royal Society, which accepted to publish them only after consistent revisions.Citation1,Citation2 The term “vaccination” (from the Latin adjective “vaccinae,” which literally means “pertaining to cows, from cow”) was coined by Jenner himself for the technique he had devised to prevent smallpox, and only more than 50 y later it was attributed a more general meaning by the French microbiologist Louis Pasteur, another pioneer in the history of vaccination.Citation3,Citation4

When Jenner first inoculated James Phipps, variolation, i.e., the inoculation of variolous material into healthy subjects as a prophylactic measure against smallpox, was a well known procedure (it had been imported in 1721 from Turkey by Lady Mary Wortley Montagu), yet was associated with a very high incidence of (often lethal) smallpox cases.Citation1,Citation2 Thus, Jenner was not the first to realize that a sublethal smallpox or cowpox infection can confer protection to subsequent, potentially lethal, challenges. Similarly, he was not the first who de facto inoculated cowpox-derived material as a prophylaxis against smallpox, since at least six investigators from the UK and Germany, including the farmer Benjamin Jesty, had done so (with variable success) earlier.Citation5 Still, it is thanks to Jenner’s observations that the British government eventually banned variolation and decided to provide cowpox-based vaccination free of charge (but optional) nation-wide (Vaccination Act, 1840). This constituted the first large-scale vaccination campaign of history, paving the way to a series of similar measures taken worldwide and culminating with the eradication of natural smallpox sources, as first certified by a committee of experts in 1979 and confirmed by WHO one year later.Citation6 Since then, the development of efficient vaccines and their widespread administration has strikingly abated the incidence of life-threatening infectious diseases including (but not limited to) rabies, typhoid, cholera, measles, plague, chickenpox, mumps, poliomyelitis and hepatitis B.Citation3 Such an extraordinary medical achievement has been possible also thanks to the critical contribution of Pasteur, who in the last decades of the 19th century demonstrated for the first time that the rationale behind smallpox vaccination could be extended to several other infectious diseases.Citation3,Citation4

Ehrlich and Coley’s hypotheses

The hypothesis that—similar to infectious diseases—cancer could be treated with active immunotherapy first arose nearly one century after Jenner’s investigations, along with the work of the German physician Paul Ehrlich and the American surgeon William Bradley Coley.Citation3 On one hand, driven by the findings made a few years earlier by Pasteur, Ehrlich (who is best known for the vaccination-unrelated concept of a “magic bullet” that would specifically kill cancer cells while sparing their normal counterparts) attempted to generate immunity against cancer by injecting weakened tumor cells, with no success.Citation3 On the other hand, inspired by multiple sporadic cases of cancer patients who underwent complete (and often long-lasting) regression following acute streptococcal fevers, Coley became convinced that he could efficiently use bacteria to cure tumors. To this aim, Coley developed a mixture of heat-killed Streptococcus pyogenes and Serratia marcescens bacteria (best known as the Coley toxin), which he begun to test in cancer patients as early as in 1896.Citation7 This preparation de facto operates as an adjuvant, facilitating the maturation of dendritic cells (DCs) via Toll-like receptor (TLR)-transduced signals,Citation8 rather than as a bona fide tumor-specific vaccine. However, similar to other relatively unspecific immunotherapeutic approaches such as the administration of high-dose interleukin (IL)-2 to melanoma and renal cell carcinoma (RCC) patients,Citation9,Citation10 Coley’s toxin soon turned out to mediate potent antitumor effects.Citation11,Citation12 Of note, the use of the Coley toxin has been suspended in the early 1960s, owing to concerns following the thalidomide case (this antiemetic was withdrawn 11 y after its approval by FDA as it was found to be highly teratogenic, leading to more than 10,000 children born with deformities worldwide).Citation13 Still, both Coley and Ehrlich represent true pioneers of modern oncoimmunology, theorizing concepts that have been disregarded for nearly one century and have received renovated interest only recently.Citation14

The “self/non-self” dichotomy and the “danger theory”

One of the major impediments against the rapid development of tumor immunology as a self-standing discipline directly stemmed from one of the most central concepts in immunology: the “self/non-self” dichotomy, as first theorized by the Australian virologist Sir Frank Macfarlane Burnet in 1949.Citation15 This model has surely been instrumental for the understanding of phenomena that underpin graft rejection and several other disorders involving an immune component.Citation16 However, it has also promoted the (incorrect) view that tumors, de facto being self tissues, must be non-immunogenic and (as a corollary) insensitive to immunotherapeutic interventions. The self/non-self model was first questioned in the late 1980s, when the cellular circuitries behind the activation of T cells, and notably the requirement for antigen presentation, began to be elucidated.Citation17 A few years later, the American scientist Polly Matzinger proposed a revolutionary theory according to which the immune system would not simply react to non-self (while sparing self) constituents, but would rather respond to situations of danger, irrespective of their origin.Citation18 The first corollary of such a “danger theory” was that trauma, cancer and other conditions that had long been viewed as immunologically silent de facto are capable of activating the immune system,Citation18,Citation19 a notion that nowadays is widely accepted.Citation20,Citation21 Approximately in the same period, van der Bruggen and colleagues from the Ludwig Institute for Cancer Research (Brussels, Belgium) were the first to clone the gene coding for MZ2-E, a protein expressed by multiple distinct melanoma cell lines as well as by tumors of unrelated histological origin, but not by a panel of normal tissues.Citation22 Moreover, cytotoxic T lymphocytes (CTLs) that specifically reacted against malignant cells in vitro were being found in patients affected by a variety of hematological and solid neoplasms.Citation22,Citation23 Thus, in line with by Polly Matzinger’s model,Citation18,Citation19 it appeared that the adult T-cell repertoire preserves the ability to react against self antigens, at least in specific circumstances.

Tumor-associated antigens

Nowadays, MZ2-E, best known as melanoma-associated antigen (MAGE)-A1, is considered as the “founder” of the large family of tumor-associated antigens (TAAs), i.e., antigens that, at least in some settings, are capable of eliciting a tumor-specific immune response manifesting with the expansion of TAA-specific CTLs.Citation24-Citation27 Unfortunately, TAA-directed immune responses are most often incapable of mediating sizeable antineoplastic effects, owing to multiple reasons (see below).Citation28 Still, the findings by van der Bruggen and colleagues generated an intense wave of investigation worldwide, not only leading to the identification of dozens, if not hundreds, of additional TAAs, but also providing additional insights into the mechanisms whereby TAAs, in selected circumstances, are capable to break self-tolerance and elicit an immune response.Citation29-Citation31 So far, four distinct classes of TAAs have been described: (1) truly exogenous, non-self TAAs; (2) unique, mutated TAAs; (3) idiotypic TAAs and (4) shared TAAs.

Exogenous TAAs

Bona fide non-self TAAs are specifically expressed by neoplasms that develop as a result of (or concomitant with) viral infections. According to WHO, the viruses that are currently known to be associated with human malignancies are limited to the Epstein-Barr virus (EBV), which is linked to lymphomas and nasopharyngeal cancer, hepatitis B virus (HBV) and hepatitis C virus (HCV), both of which are associated with hepatocellular carcinoma, human papillomaviruses (HPV), in particular HPV-16 and HPV-18, which are associated with head and neck, cervical and anal carcinomas, human T lymphotropic virus type 1 (HTLV-1) and type 2 (HTLV-2), which are linked to adult T-cell leukemia and hairy-cell leukemia, respectively, and human herpesvirus 8 (HHV-8), which is associated with Kaposi’s sarcoma.Citation32-Citation34 The possibility to develop recombinant vaccines against these viruses has been extensively investigated in the last decade, and multiple clinical trials have been concluded with encouraging results.Citation35-Citation39 In this context, a special mention goes to Cervarix® and Gardasil®, two multivalent, recombinant anti-HPV vaccines that have been approved by FDA in 2009 as preventive measures against HPV infection and the consequent development of cervical carcinoma.Citation40 The success of Cervarix® and Gardasil® as compared with other vaccination strategies against viral cancers that have not yet moved from the bench to the bedside, depends—at least in part—on the fact that both these vaccines were developed as fully preventive measures, aimed at blocking de novo HPV infection, rather than as at therapeutic strategy against established cervical carcinoma. Indeed, both Cervarix® and Gardasil® induce high levels of neutralizing antibodies and result in the generation of HPV-specific long-lasting memory B cells,Citation41 which efficiently prevent infection, yet are less efficient in promoting T-cell responses that may be beneficial for cervical carcinoma patients. In line with thin notion, official documents report that Cervarix® is not efficient against histopathological endpoints in HPV-infected women (source http://www.fda.gov).

Unique TAAs

Malignant cells near-to-invariably accumulate genetic alterations, which can be as gross as chromosomal rearrangements (e.g., t(9;22)(q34;q11), resulting in the very well known Philadelphia chromosome and leading to chronic myelogenous leukemia, CML) or as specific as point mutations affecting the activity of tumor suppressor genes (e.g., ATM, TP53) or oncogenes (e.g., ALK, EGFR, KRAS).Citation42 Some of these alterations (such as the Philadelphia chromosome and the resulting fusion kinase BCR-ABL) are so prevalent among specific populations of cancer patients that their detection decisively contributes to diagnosis.Citation43,Citation44 Others (such as R175H, R248W and R273H TP53 substitutions) are highly prevalent, too, yet affect a rather heterogeneous and very large population of patients, bearing malignancies that encompass (but are not limited to) breast, lung, gastric and colorectal cancer.Citation45 Irrespective of whether these changes actually drive oncogenesis and tumor progression (driver mutations) or whether they appear alongside with carcinogenesis and are retained by tumor cells (passenger mutations),Citation46 non-synonymous mutations that affect exons are expected to generate new, tumor-specific (unique) and potentially immunogenic antigens.Citation47 In line with this notion, patients affected by neoplasms bearing one of such unique TAAs have been shown to naturally develop anti-TAA antibodies and/or TAA-specific CD8+ cells, although these responses—in the near-to-totality of cases—are unable to exert significant antitumor effects.Citation48-Citation50 As unique TAAs are only expressed by malignant cells, immune responses arising against their epitopes have a very low probability to result in autoimmune reactions. In addition, the development of efficient immunotherapies against unique TAAs that are expressed by a wide array of tumors would provide clinical benefits to a large population of cancer patients. During the last two decades, the intense wave of research stemming from these considerations has demonstrated that targeting unique TAAs constitutes a meaningful immunotherapeutic approach against cancer.Citation51-Citation55

Idiotypic TAAs

One particular class of unique TAAs is constituted by idiotypic TAAs. Hematological malignancies arising from B cells that have functionally rearranged immunoglobulin (Ig)-coding genes are characterized by the cell surface expression of a clonal B-cell receptor (BCR). Such a BCR is de facto a self protein, yet contains a unique variable region that defines its specificity (idiotype), to which the immune system has never been exposed, and hence that is potentially immunogenic.Citation56 In line with this notion, anti-idiotypic antibodies arise naturally in the course of humoral immune responses (when high levels of clonal Igs are produced by plasma cells), which they contribute to terminate.Citation57,Citation58 In 1972, Lynch et al. were the first to demonstrate that peptides corresponding to idiotypic regions of the BCR exposed by myeloma cells are capable of eliciting an efficient immune response,Citation59 de facto providing the rationale for the development of idiotypic anticancer vaccination. In practical terms, this can be achieved not only by injecting purified peptides that correspond to the idiotype expressed by malignant cells, but also by means of anti-idiotype antibodies.Citation60 The latter constitute bona fide structural mimics of TAAs (which in this specific case—but not in many other settings—are represented by the idiotype), owing to the fact that antigens and the corresponding antibodies exhibit a consistent degree of complementarity.Citation60 In general, anti-idiotype antibodies are advantageous as compared with purified peptides as they can be easily and cost-effectively produced in high amounts by immunizing laboratory animals with TAA-targeting antibodies.Citation60 Irrespective of how they are elicited, anti-idiotype immune responses are patient- and tumor-specific, implying (1) that the development of idiotypic anticancer vaccines requires the precise characterization of neoplastic cells on a per patient basis, and (2) that the efficacy of this approach can be fully compromised by the arisal of a new malignant cell clone as well as by processes of somatic (hyper)mutation, which normally affect the idiotype.Citation61 Still, following the pioneer work by Lynch and colleagues,Citation59 the fact that idiotypes constitute a meaningful target for the therapy of B-cell neoplasms has been validated in multiple preclinical and clinical settings.Citation62-Citation65

Shared TAAs

Obviously, cancer cells express (and sometimes overexpress) a majority of self antigens, which they share with the normal tissue they originated from.Citation66 According to the “self/non-self” theory, these antigens should not elicit an immune response, due to central and/or peripheral tolerance mechanisms that are in place to prevent autoimmune reactions.Citation17 This prediction is actually inaccurate, as (1) both antibodies and CD8+ T cells recognizing shared TAAs (e.g., wild type epidermal growth factor receptor, EGFR and p53) appear to be enriched in the circulation of cancer patients as compared with healthy subjects;Citation67,Citation68 and (2) a consistent fraction of paraneoplastic syndromes derives from tumor-elicited autoimmune reactions targeting normal tissues.Citation69 Thus, as postulated by the “danger theory,” self-shared TAAs are capable of eliciting an immune response, most likely because they are presented to the immune system in the context of appropriate activation signals.Citation18,Citation19 Such an immune response is frequently held in check by local immunosuppressive mechanisms (see below),Citation70,Citation71 and hence does not exert antitumor effects, yet it may be functional at distant sites, thus underlying life-threatening paraneoplastic syndromes.Citation69 During the last two decades, great efforts have been dedicated at understanding whether and based on which strategies shared TAAs would constitute meaningful targets for the elicitation of antitumor immune responses. Promising results have been obtained in both preclinical and clinical models.Citation52,Citation72,Citation73 Of note, although so-called “cancer-testis” antigens (CTAs) are expressed not only by a variety of malignant cells but also by germline cells,Citation74 they are most often considered as unique, rather than shared, TAAs, mostly due to the fact that testes represent an immune privileged site and are de facto spared by most, if not all, autoimmune reactions.Citation75

Considerations on the development of anticancer vaccines

Along with the recognition that the immune system is not completely irresponsive to tumors (as it was initially thought to be) and that malignant cells express antigens that are capable of eliciting a tumor-specific immune response, great efforts have been dedicated to the development of anticancer vaccines.Citation29 Thus, several approaches have been evaluated for their potential to elicit efficient, tumor-specific immune responses, including (but not limited to): recombinant TAAs, in the form of short synthetic epitopes (expected to directly bind, and hence be presented to T cells on, MHC molecules); recombinant full-length proteins (whose presentation requires the uptake and processing by antigen-presenting cells, APCs) or tumor cell-purified preparations (containing TAAs alone or in complex with chaperon proteins), administered as such or via multiple delivery systems (e.g., nanoparticles, DC-derived exosomes, DC-targeting vectors); TAA-encoding vectors; and DC preparations. The results of such an intense wave of investigation/vaccine development have been encouraging. Still, exception made for Cervarix® and Gardasil® (which are approved for prophylactic use, see above), only one product is currently commercialized as a therapeutic anticancer vaccine, namely, sipuleucel-T (also known as Provenge®), a cellular preparation for the treatment of asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer.Citation76 This is in strike contrast with the large array of vaccines that have been developed against infectious agents during the last century. Indeed, there are at least three major obstacles that complicate the development of anticancer vaccines as compared with prophylactic vaccines against infectious diseases. First: the antigenic properties of cancer cells. Although a number of specific and potentially immunogenic TAAs have been identified (see above), only a few of them operate as bona fide tumor rejection antigens (TRAs) as they elicit an immune response that leads to tumor eradication.Citation26,Citation77 Of note, it has recently been shown that TRAs not necessarily correspond to TAAs that arise as a result of driver mutations, indicating (1) that there is no direct correlation between the oncogenic potential of mutations and their immunogenicity, and (2) that passenger mutations might generate therapeutically useful targets for immunotherapy.Citation78 Second: the fact that anticancer vaccines must operate, in the vast majority of cases, as therapeutic interventions. Conventional prophylactic vaccines against infectious agents elicit strong humoral responses and promote the establishment of long-term B-cell memory.Citation79 While this results in an efficient protection against invading pathogens (including HPV strains associated with cervical carcinoma, see above), it has limited (if any) efficacy against established tumors. Indeed, the rejection of established neoplastic lesions requires the activation of robust cell-mediated immune responses, which can be achieved only by specific vaccination strategies.Citation3,Citation80 In particular, the elicitation of cell-mediated immunity requires TAAs to be conveniently processed by APCs, mainly DCs, and presented to T cells in vivo in the context of appropriate stimulatory signals.Citation30 This is a critical point and explains why vaccines are invariably administered in the presence of adjuvants (encompassing classical agents such as alum, montanide and incomplete Freund’s adjuvant as well as recently developed TLR agonists like monophosphoryl lipid A, MPLA and imiquimod).Citation11,Citation12 Indeed, in the absence of activation signals, immature DCs present TAAs to T cells in the context of inhibitory interactions, hence promoting the establishment of tolerance via multiple mechanisms.Citation81-Citation84 Third, the existence of distinct immunosuppressive pathways that are elicited by tumor cells, both locally and systemically. Cancer cells not only co-opt the stromal components of the neoplastic lesion to serve their metabolic and structural needs,Citation85,Citation86 but also secrete a wide array of mediators that (1) stimulate the bone marrow to release specific subsets of (relatively immature) myeloid cells into the bloodstream; (2) attract such cells and others to the tumor microenvironment and promote their expansion; (3) condition the differentiation program and/or functional behavior of tumor-infiltrating leukocytes.Citation87-Citation91 Overall, this results not only in the establishment of a potently immunosuppressive tumor microenvironment but also in some extent of systemic immunosuppression, and explains, at least in part, why natural TAA-directed immune responses are near-to-always unable to exert antitumor effects.

Along the lines of our Trial Watch series,Citation11,Citation12,Citation92-Citation97 here we will discuss recently published and ongoing clinical trials that have investigated/are investigating the safety and efficacy of purified peptides or full-length proteins as therapeutic interventions against cancer.

Hematological Malignancies

During the past 15 y, the safety and efficacy of recombinant peptides/proteins employed as therapeutic vaccines against hematological neoplasms have been evaluated in a few clinical trials. Peptides derived from Wilms’ tumor 1 (WT1), a transcription factor that is overexpressed by several neoplasms,Citation98 have been tested (most often combined with the carrier keyhole limpet hemocyanin, KLH) in CML patients (n = 1)Citation99 acute myeloid leukemia (AML) patients (n = 10 and n = 10),Citation100,Citation101 as well as in a mixed cohort of AML and myelodysplastic syndrome (MDS) patients (n = 19).Citation102 A peptide derived from receptor for hyaluronic acid-mediated motility (RHAMM, a hyaluronate-binding protein that influences cell motility) has been evaluated in AML, MDS and multiple myeloma (MM) patients (n = 10 and n = 9).Citation103,Citation104 Idiotype vaccines have been investigated in cohorts of myeloma (n = 5 and n = 6)Citation105,Citation106 and lymphoma (n = 20, n = 16 and n = 177) patients.Citation63,Citation107,Citation108 Finally, two clinical trials have investigated the therapeutic potential of autologous, tumor-derived heat-shock protein (HSP)-complexed antigens in CML (n = 20) and non-Hodgkin’s lymphoma (n = 20) patients.Citation109,Citation110 Altogether, these studies demonstrated that recombinant TAA-derived peptides are well tolerated by patients bearing hematological malignancies. These vaccines elicited TAA-specific immune responses in a variable fraction of patients, some of whom also exhibited partial or complete clinical responses.

Nowadays (September 2012), official sources list 11 recent (started after January, 1st 2008), ongoing (not withdrawn, terminated or completed at the day of submission) Phase I-II clinical studies assessing the safety and efficacy of recombinant peptides as therapeutic interventions against hematological neoplasms (). Six of these studies are investigating WT1-derived peptides, either as a standalone intervention or combined with granulocyte macrophage colony-stimulating factor (GM-CSF) or regimens for the depletion of immunosuppressive FOXP3+ regulatory T cells (Tregs), in cohorts of AML and MDS patients. The remaining 5 studies involve MM patients or subjects affected by various hematological malignancies, who are receiving, either as single agents or in combination with various immunostimulatory strategies, peptides derived from the MAGE-A1-related protein MAGE-A3,Citation111 from mucin 1 (MUC1, an extensively glycosylated transmembrane protein that is overexpressed by a wide variety of cancers),Citation112 from the catalytic subunit of human telomerase reverse transcriptase (hTERT)Citation113 or from the anti-apoptotic protein survivinCitation114 (source www.clinicaltrials.gov).

Table 1. Clinical trials testing TAA-derived peptides as therapeutic interventions in patients affected by hematological neoplasms.*

Neurological and Pulmonary Cancers

To the best of our knowledge, the first clinical trials investigating the safety and therapeutic potential of TAA-derived peptides in brain and lung cancer patients have been completed in the mid 2000s,Citation115-Citation118 followed by a few additional studies addressing the same question.Citation119-Citation123 In particular, a personalized multi-peptide preparation combined with a mineral oil-based adjuvant (Montanide ISA51) has been tested in glioma patients (n = 25),Citation118 tumor-derived peptides complexed with HSPs have been evaluated in astroglyoma, oligodendrocytoma and meningioma patients (n = 5),Citation120 and a WT1-derived 9mer has been tested in individuals affected by glioblastoma multiforme (GBM) (n = 21).Citation122 In addition, cohorts of non-small cell lung carcinoma (NSCLC) patients have been treated with peptides derived from ERBB2/HER2 (a member of the epidermal growth factor receptor family frequently overexpressed in lung and breast cancer patients),Citation124 in combination with GM-CSF (n = 2 and n = 1),Citation115,Citation117 with hTERT-derived peptides, combined with either GM-CSF or radiotherapy (n = 26 and n = 23),Citation119,Citation123 and with peptides corresponding to mutated regions of RAS (n = 18).Citation121 Taken together, these studies demonstrated that the administration of TAA-derived peptides to patients affected by neurological or pulmonary malignancies is safe and has the potential of inducing — in a fraction of cases — immunological and clinical responses.

Today (September 2012), official sources list 13 recent, ongoing, Phase I-III clinical trials investigating the safety profile and efficacy of TAA-derived vaccines as therapeutic interventions against neurological neoplasms (). Six of these studies involve GBM patients, 4 glioma patients, 1 astrocytoma patients, 1 neuroblastoma patients and 1 individuals bearing not-better specified brain tumors. In four trials, a peptide corresponding to the EGFR in-frame deletion mutant EGFRvIII (rindopepimut, also known as CDX-110)Citation125,Citation126 is employed, either as a single agent or in combination with GM-CSF, temozolomide or radiotherapy. Alternatively, patients are administered with glioma-associated antigens (GAAs), frequently associated to the TLR3 activator polyriboinosinic-polyribocytidylic acid stabilized with poly-L-lysine in carboxymethylcellulose (polyICLC), with survivin-derived peptides, with HSP-TAA complexes or with a multi-peptide vaccine containing 11 distinct TAAs (IMA950)Citation127 (source www.clinicaltrials.gov). In addition, official sources list 17 recent, ongoing, Phase I-III clinical trials investigating the potential of TAA-derived peptides for the treatment of lung cancer, mainly NSCLC, patients (). These studies involve a variety of recombinant vaccines, including (but not limited to) peptides derived from MUC1, MAGE-A3, hTERT, kinesin family member 20A (KIF20A), cell division cycle-associated 1 (CDCA1), vascular endothelial growth factor receptor 1 and 2 (VEGFR1 and VEGFR2) and CTAs (such as NY-ESO-1 and upregulated in lung cancer 10, URLC10).Citation74 In the majority of cases, peptides or full-length proteins are administered as standalone adjuvanted agents, with the exceptions of trial NCT01579188, in which hTERT-derived peptides are combined with GM-CSF, trials NCT00409188 and NCT01015443, in which MUC1-derived peptides are administered after a single dose of cyclophosphamide, and trial NCT00455572, in which recombinant full-length MAGE-A3 is combined with radiotherapy, cisplatin (a DNA-damaging agent) or vinorelbine (a semi-synthetic vinca alkaloid). Importantly, trial NCT00480025, in which advanced NSCLC patients are treated with adjuvanted full-length MAGE-A3 upon tumor resection, constitutes the (or at least one of the) largest clinical study(ies) ever commenced to evaluate the efficacy of an immunotherapeutic intervention against lung cancer.Citation128 Another particularly intriguing approach in this context is represented by trial NCT00655161, in which NSCLC patients receive an inactivated strain of Saccharomyces cerevisiae that has been engineered for the expression of mutant RAS (GI-4000) (source www.clinicaltrials.gov).

Table 2. Clinical trials testing TAA-derived peptides and/or full length proteins as therapeutic interventions in patients affected by neurological and pulmonary malignancies

Breast, Ovarian and Prostate Carcinoma

During the last two decades, the potential of recombinant vaccines employed as therapeutic interventions against breast, ovarian and prostate carcinoma patients has been extensively investigated. Thus, cohorts of breast carcinoma patients have been administered with HER2-derived peptides in combination with GM-CSF (n = 31, n = 9, n = 9 and n = 195),Citation115-Citation117,Citation129 with peptides derived from a specific splicing variant of survivin (n = 14),Citation130 with a broad panel of peptides naturally presented by ovarian cancer cells in combination with GM-CSF (n = 7),Citation131 with full-length CA15–3, CA125 and carcinoembryonic antigen (CEA), three circulating markers of breast cancer recurrence,Citation132 combined with autologous breast cancer cells, allogeneic breast cancer MCF-7 cells, GM-CSF and recombinant IL-2 (n = 42),Citation133 and with Sialyl-Tn (a MUC1-associated carbohydrate) chemically coupled to KLH (n = 33).Citation134 Some of these approaches have alongside been tested in ovarian cancer patients,Citation115,Citation116,Citation131,Citation134 owing to the fact that breast and ovarian carcinomas share a relatively consistent number of TAAs.Citation135 Moreover, ovarian carcinoma patients have been treated with a synthetic form of an immunodominant disaccharide of the Thomsen-Friedenreich antigen conjugated to KLH (n = 10),Citation136 with not better specified pre-designated or evidence-based peptides (n = 5),Citation137 with a p53-derived synthetic long peptide (SLP) coupled to immunostimulatory doses of cyclophosphamide (n = 10),Citation138 and with multiple courses of recombinant poxviruses encoding full-length NY-ESO-1 (n = 22).Citation139 Finally, prostate carcinoma patients have received HER2-derived peptides, as such or in the form of hybrids with a moiety of the MHC class II-associated invariant chain, plus GM-CSF (n = 40 and n = 32),Citation140,Citation141 prostate-specific antigen (PSA)-derived peptides, as a single adjuvanted agent (n = 5) or combined with GM-CSF (n = 28),Citation142,Citation143 full-length NY-ESO-1 complexed with cholesterol-bearing hydrophobized pullulan (CHP) (n = 4, n = 4 and n = 2),Citation144-Citation146 an adjuvanted globo H hexasaccharide-KLH fusion (n = 20),Citation147 and a number of multi-peptide preparations often, but not always, including PSA- and squamous cell carcinoma antigen recognized by T cells (SART)-derived peptides and combined with GM-CSF or estramustine phosphate, an alkylating estradiol derivative (n = 13, n = 10, n = 16, n = 19 and n = 23).Citation148-Citation153 Altogether, these studies demonstrated that the administration of recombinant peptides or full length proteins to breast, ovarian and prostate carcinoma patients is generally safe and can induce, in a fraction of cases, immunological and clinical responses.

Nowadays (September 2012), official sources list 16 recent, ongoing Phase I-III clinical trials assessing the safety and efficacy of recombinant peptides in breast carcinoma patients (). A majority of these studies involve the administration of HER2-derived peptides, either as adjuvanted standalone interventions or combined with additional immunostimulatory agents, including low doses of cyclophosphamide, GM-CSF and polyICLC. Alternatively, vaccination regimens based on CDCA1-, CEA-, hTERT-, KIF20A-, MUC1-, survivin-, URLC10- and WT1-derived peptides are being evaluated (source www.clinicaltrials.gov). In addition, official sources list 8 recent, ongoing, Phase I-II clinical trials investigating TAA-derived peptides for the therapeutic vaccination of ovarian (3 studies) and prostate (5 studies) carcinoma patients (). The trials enrolling ovarian carcinoma patients involve the administration a p53-derived SLP combined with pegylated interferon (IFN), full-length NY-ESO-1 adjuvanted with MPLA or a peptide derived from folate-binding protein (FBP, which is often overexpressed by ovarian neoplasms)Citation154 in association with GM-CSF. The studies recruiting prostate carcinoma patients are based on peptides derived from T-cell receptor gamma chain alternate reading frame protein (TARP, a nuclear protein overexpressed in a large proportion of prostate carcinomas),Citation155,Citation156 administered either as a single agent or combined with ex vivo TARP peptide-pulsed DCs, peptides derived from prostate membrane-specific antigen (PMSA, a glycoprotein specifically expressed by normal and malignant prostate cells), CDCA1-derived epitopes, a synthetic peptide derived corresponding to amino acids 22–31 of mouse gonadotropin releasing hormone (GnRH), or full-length NY-ESO-1, all given as standalone adjuvanted interventions (source www.clinicaltrials.gov).

Table 3. Clinical trials testing TAA-derived peptides and/or full length proteins as therapeutic interventions in patients affected by breast, ovarian and prostate carcinoma

Melanoma

Together with RCC, melanoma constitutes by far the clinical setting in which immunotherapeutic interventions have been most extensively investigated, at least in part due to the fact that both these neoplasms naturally generate immune responses and appear to be very sensitive to immunostimulatory interventions, even as unspecific as the systemic administration of high-dose IL-2.Citation9,Citation10 This intense research effort has lead not only to an improved understanding of the biology of melanoma cells, but also to the detailed characterization of a wide panel of melanocyte differentiation antigens (MDAs), underpinning the development of potential anticancer vaccines.Citation157 The safety and therapeutic profiles of many of such vaccination strategies have been tested in clinical trials starting from the late 1990s. These studies involved peptides derived from MDAs including, but not limited to: the type I transmembrane glycoprotein gp100 (n = 22, n = 15, n = 26, n = 12, n = 60, n = 25, n = 24, n = 8, n = 11, n = 51, n = 12, n = 121, n = 197 and n = 185),Citation158-Citation171 the 18 KDa transmembrane protein melan A (also known as melanoma antigen recognized by T cells 1, MART-1) (n = 1, n = 3, n = 15, n = 28, n = 12, n = 60, n = 25, n = 6, n = 24, n = 8, n = 11, n = 12, n = 17, n = 18 and n = 15),Citation159,Citation161,Citation163-Citation166,Citation168,Citation172-Citation178 several members of the MAGE-A protein family such as MAGE-A1, MAGE-A3 and MAGE-A10 (n = 24, n = 51, n = 121 and n = 197),Citation164,Citation167,Citation169,Citation170 and tyrosinase, an enzyme required for melanin synthesis (n = 18, n = 43, n = 15, n = 26, n = 60, n = 25, n = 24, n = 11, n = 51, n = 121, n = 197 and n = 18).Citation159,Citation160,Citation162-Citation164,Citation166,Citation167,Citation169,Citation170,Citation177,Citation179,Citation180 In addition, clinical trials enrolling melanoma patients have been performed to assess the safety profile and therapeutic potential of NY-ESO-1-derived peptides (n = 37, n = 8, n = 13 and n = 121),Citation169,Citation181-Citation183 hTERT-derived peptides (n = 25),Citation184 full-length recombinant NY-ESO-1 (n = not available, n = 51, n = 1, n = 1 and n = 18),Citation144,Citation145,Citation185-Citation187 HSP-complexed antigens (n = not available),Citation188 and subsequent courses of recombinant poxviruses encoding full-length NY-ESO-1 (n = 25).Citation139 Most often, MDA- and/or TAA-derived peptides were administered as part of multi-peptide preparations and combined with immunostimulatory interventions including conventional adjuvants, GM-CSF, IL-2 and cyclophosphamide. In line with the high sensitivity of melanoma cells to immunostimulatory approaches, the vast majority of these clinical trials reported no significant side effects and satisfactory rates of durable clinical responses.

Today (September 2012), official sources list 25 recent, ongoing Phase I-III clinical trials assessing the safety and efficacy of recombinant peptides/proteins in melanoma patients (). Most of these studies are based on various MDA- or TAA-derived peptides, given either as single adjuvanted agents or combined with additional immunostimulatory interventions including, but not limited to, IL-2, IL-12, pegylated IFNα, IFNγ, GM-CSF, TLR agonists (e.g., polyICLC, imiquimod, resiquimod, lipopolysaccharide) and monoclonal antibodies targeting CD40 or PD1. In this setting, particularly interesting strategies are being undertaken by trial NCT01331915, investigating the safety and anticancer profile of a recombinant, detoxified toxin from Bordetella pertussis coupled to a tyrosinase epitope,Citation189 and by trial NCT00706992, testing the clinical potential of a replication-defective recombinant canarypox virus encoding a melan A-derived epitope coupled to T cells genetically engineered to express a melan A-targeting T-cell receptor (TCR)Citation190 (source www.clinicaltrials.gov).

Table 4. Clinical trials testing TAA-derived peptides and/or full-length proteins as therapeutic interventions in melanoma patients

Gastrointestinal, Pancreatic and Colorectal Tumors

The results of the first clinical trials investigating the safety and efficacy of TAA-derived peptides or proteins as therapeutic interventions in cohort of patients affected by gastrointestinal, pancreatic and colorectal neoplasms have been published no earlier than in 2004.Citation191,Citation192 Since then, the following therapeutic and clinical settings have been investigated: survivin-derived peptides, given to colorectal carcinoma (CRC) (n = 15) or pancreatic cancer (n = 1) patients as a single adjuvanted agent,Citation192,Citation193 a multi-peptide vaccine including epitopes from distinct SART proteins administered to CRC patients as a standalone adjuvanted intervention (n = 10),Citation191 a personalized, peptide-based vaccine, given to CRC patients in combination with uracil, tegafur and calcium folinate (n = 8),Citation194 a personalized combination of maximum 4 peptides derived from 16 distinct TAAs including (but not limited to) HER2, CEA, PAP, PSA, SART2 and SART3, given to advanced gastric carcinoma or CRC patients in combination with a 5-fluorouracil derivative (n = 11),Citation195 full-length NY-ESO-1, administered as a CHP complex to esophageal cancer patients (n = 4, n = 8, n = 4 and n = 8),Citation144-Citation146,Citation196 an artificially synthesized helper/killer-hybrid epitope long peptide derived from MAGE-A4, given as a dually adjuvanted standalone intervention to a patient with CRC pulmonary metastasis,Citation197 and three peptides derived from the protein kinase TTK, lymphocyte antigen 6 complex locus K (LY6K), and insulin-like growth factor II mRNA-binding protein 3 (IMP3), administered in incomplete Freund’s adjuvant to esophageal cancer patients (n = 10 and n = 60).Citation198,Citation199 In all these settings, vaccination with TAA-peptides was well tolerated and, in multiple instances, it also elicited immunological and clinical responses.

Nowadays (September 2012), official sources list 9 recent, ongoing Phase I-II clinical trials investigating the safety and efficacy of recombinant peptides/proteins in esophageal cancer (5 trials), gastric cancer (1 trial), pancreatic carcinoma (5 trials) and CRC (4 trials) patients (). CHP-complexed full-length NY-ESO-1 as a single agent as well as peptides derived from common TAAs such as CDCA1, TTK, URLC10, VEGFR1 and VEGFR2, either as standalone interventions or combined with TLR9 agonists, are being tested in esophageal cancer patients. The safety and therapeutic profile of VEGFR1-derived peptides, as single agents, is being investigated in gastric carcinoma patients. CRC patients are being enrolled in trials involving MUC1-derived peptides combined with either chemoradiation therapy plus cyclophosphamide or polyICLC, peptides derived from the CTA RNF43, given as standalone agents, as well as GI-4000 (an inactivated strain of S. cerevisiae engineered for the expression of mutant RAS, see above), in combination with conventional chemotherapy or bevacizumab (a VEGF-targeting monoclonal antibody). Finally, peptides derived from hTERT and VEGFR1/2 are being tested in pancreatic carcinoma patients, in combination with GM-CSF plus tadalafil (a phosphodiesterase type 5 inhibitor currently approved for the therapy of erectile dysfunction and commercialized under the label of Cialis®) and/or gemcitabine (a nucleoside analog) (source www.clinicaltrials.gov).

Table 5. Clinical trials testing TAA-derived peptides and/or full-length proteins as therapeutic interventions in patients affected by esophageal, gastric, pancreatic and colorectal carcinoma

Renal, Bladder and Reproductive Tract Tumors

So far, a few clinical studies have investigated the profile of TAA-derived peptides or proteins employed as therapeutic interventions in cohort of patients affected by RCC and distinct malignancies of the reproductive tract, including cervical carcinoma, endometrial cancer, uterine sarcoma and vulvar intraepithelial neoplasia.Citation137,Citation200-Citation204 In particular, multi-peptide vaccination strategies involving up to six peptides derived from a broad panel of RCC-associated antigens have been tested, invariably in combination with immunostimulatory interventions (including IL-2, IFNα, GM-CSF and low-dose cyclophosphamide), in RCC patients (n = 10 and n = 96).Citation203,Citation204 In addition, the efficacy of peptides corresponding to distinct regions of the HPV-16 protein E7 has been evaluated in patients affected by cervical carcinoma or vulvar intraepithelial neoplasia, most often as standalone adjuvanted agents or combined with pan-HLA-DR-binding T helper epitopes (n = 19, n = 18 and n = 15).Citation200-Citation202 Finally, not better specified pre-designated or evidence-based peptides have been tested in a cohort of patients affected by cervical carcinoma or various other neoplasms of the reproductive tract (n = 9).Citation137 The administration of recombinant peptides combined to immunostimulatory interventions was well tolerated by RCC patients and yielded immunological responses that, at least in some cases, were associated with improved patient survival.Citation203,Citation204 Conversely, E7-derived peptides induced potent immune responses that, in one trial, led to viral clearance from cervical scrapings by the fourth vaccine course,Citation200 yet were unable to promote efficient antitumor immunity.Citation137,Citation200-Citation202 These results are in line with the fact that—according to official sources—preventive anti-HPV vaccines (i.e., Cervarix® and Gardasil®) are not efficient against histopathological endpoints when used as therapeutic agents in HPV-infected women (source http://www.fda.gov).

Today (September 2012), official sources list 10 recent, ongoing Phase I-II clinical trials investigating the safety and efficacy of recombinant peptides/proteins in bladder carcinoma (3 trials) and reproductive tract cancer (7 trials) patients (). In the former clinical setting, MAGE-A3-derived peptides, recombinant full-length MAGE-A3 or epitopes derived from DEP domain containing 1 (DEPDC1) and M phase phosphoprotein 1 (MPHOSPH1) are being tested, either as standalone adjuvanted agents or in combination with the bacillus Calmette-Guérin (BCG), an attenuated strain of Mycobacterium bovis that is currently employed against superficial bladder carcinoma.Citation205 In the latter clinical setting, 2 studies involve full-length NY-ESO-1 combined with GM-CSF, the demethylating agents decitabine and doxorubicine (an anthracycline that has recently been shown to promote the immunogenic death of tumor cells),Citation20,Citation206,Citation207 2 studies involve a lyophilized liposomal preparation containing either 7 different TAA-derived peptides (DPX-0907, given as a standalone adjuvanted agent) or survivin-derived epitopes (administered in combination with cyclophosphamide), 1 study involves the administration of folate receptor α-derived peptides plus cyclophosphamide, 1 study involves FBP-derived epitopes given together with GM-CSF and 1 study is based on a replication-defective NY-ESO-1-coding canarypox virus combined with GM-CSF and the mammalian target or rapamycin (mTOR) inhibitor sirolimus (source www.clinicaltrials.gov).

Table 6. Clinical trials testing TAA-derived peptides and/or full-length proteins as therapeutic interventions in patients affected by bladder carcinoma and tumors of the reproductive tract

Additional Neoplasms and Mixed Clinical Cohorts

Recombinant TAA-derived peptides and full-length proteins have been tested in a few additional clinical settings, encompassing oral and urothelial cancer patientsCitation208,Citation209 as well as rather heterogeneous cohorts including subjects affected by wide arrays of solid neoplasms.Citation101,Citation210-Citation219 Thus, oral and urothelial cancer patients (n = 11 and n = 9, respectively) have been treated with a survivin-derived 9-mer, either as a subcutaneous or as a intratumoral adjuvanted injection.Citation208,Citation209 In addition, WT1-derived 9-mers, HER2-derived short epitopes or long peptides complexed with CHP, and not better indicated peptides recognized by circulating T cells in the periphery have been tested, as adjuvanted standalone interventions, in cohort of patients affected by not better specified solid tumors (n = 5, n = 10, n = 9, n = 24 and n = 14),Citation101,Citation210-Citation212,Citation219 NY-ESO-1-derived peptides have been evaluated in patients bearing metastatic NY-ESO-1-expressing cancers (n = 12),Citation213 and epitopes corresponding to mutated regions of RAS, CEA-derived peptides, complex multi-peptide preparations as well as HSP-complexed antigens have been used to vaccinate patients affected by distinct types of carcinoma or advanced neoplasms (n = 8, n = 10, n = not available, n = 113 and n = 16).Citation214-Citation218 In general, the administration of purified peptides/proteins to these patients was well tolerated and promoted—in a few cases—immunological and clinical responses.

Today (September 2012), official sources list 12 recent, ongoing Phase I-II clinical trials investigating the safety and efficacy of recombinant peptides/proteins in patients affected by various tumor types encompassing head and neck carcinoma (1 trial), hepatocellular carcinoma (1 trial), mesothelioma (2 trials), bile duct cancer (1 trial), as well as in relatively heterogeneous patient cohorts (7 trials) (). The vast majority of these studies involves the administration of TAA-derived peptides, either as standalone adjuvanted agents or combined with immunostimulatory compounds such as GM-CSF, TLR agonists or low doses of cyclophosphamide. Two notable exceptions are constituted by NCT01569919, testing a recombinant modified vaccinia Ankara viral vector encoding the 5T4 fetal oncoprotein in mesothelioma patients and NCT01526473, evaluating a non-infective variant of the Venezuelan equine encephalitis virus encoding the extracellular domain and transmembrane region of HER2 in patients affected by not better specific HER2+ neoplasms (www.clinicaltrials.gov).

Table 7. Clinical trials testing TAA-derived peptides and/or full length proteins as therapeutic interventions in patients affected by additional tumor type and in mixed patient cohorts

Concluding Remarks

During the last two decades, the molecular and cellular circuitries whereby malignant cells and the immune system mutually interact have been the subject of in-depth investigation. Such a renovated interest, stemming within the conceptual framework provided by Polly Matzinger’s danger theory, has been paralleled by the development of multiple strategies for anticancer vaccination. These approaches, involving the use of recombinant proteins, TAA-encoding vectors or DC preparations, have generated encouraging results in both preclinical and clinical settings. However, only a few trials assessing the efficacy of TAA-derived peptides and/or full length proteins have reported consistent rates of objective, long-term clinical responses.Citation108,Citation129,Citation171,Citation204,Citation220 In line with this notion, no more than three anticancer vaccines are currently approved by FDA for use in humans: Provenge®, employed as a therapeutic intervention in a limited subset of prostate carcinoma patients; Cervarix® and Gardasil®, both given as prophylactic agents against HPV infection (and hence against HPV-associated cervical carcinoma). At least in part, this is due to the fact that the eradication of established malignant lesions requires a robust tumor-specific, cell-mediated immune response that is relatively difficult to obtain, owing to multiple reasons (see above). Moreover, it appears that several TAA-derived peptides and/or full-length protein exhibit (at least some degree of) clinical activity when administered as adjuvant therapy or to patients with minimal residual disease, yet fail to provide any clinical benefit to individuals bearing advanced and/or metastatic lesions.Citation80,Citation108,Citation220-Citation222 We believe that (1) the discovery of novel bona fide TRAs, (2) the optimization of adjuvant strategies that potently activate DCs in vivo, (3) the rational combination of anticancer vaccines with immunomodulatory agents (such as anti-CTLA4 and anti-PD1 antibodies), (4) the precise identification of the subsets of patients that are most likely to respond to vaccination with robust immune responses and (5) the establishment of standardized protocols to evaluate the nature, breadth and quality of antigen-specific T-cell responses, an objective recently proposed by the MIATA (Minimal Information About T Cell Assays) project,Citation223-Citation225 are the keys toward the development of new, efficient and (perhaps) clinically useful anticancer vaccines.

Abbreviations:
AML=

acute myeloid leukemia

APC=

antigen-presenting cell

BCG=

bacillus Calmette-Guérin

BCR=

B-cell receptor

CDCA1=

cell division cycle-associated 1

CEA=

carcinoembryonic antigen

CHP=

cholesterol-bearing hydrophobized pullulan

CML=

chronic myelogenous leukemia

CRC=

colorectal carcinoma

CTA=

cancer-testis antigen

CTL=

cytotoxic T lymphocyte

DC=

dendritic cell

DEPDC1=

DEP domain containing 1

EBV=

Epstein-Barr virus

EGFR=

epidermal growth factor receptor

FBP=

folate-binding protein

GAA=

glioma-associated antigen

GBM=

glioblastoma multiforme

GM-CSF=

granulocyte macrophage colony-stimulating factor

GnRH=

gonadotropin releasing hormone

HBV=

hepatitis B virus

HCV=

hepatitis C virus

HHV-8=

human herpesvirus 8

HPV=

human papillomavirus

HSP=

heat-shock protein

hTERT=

human telomerase reverse transcriptase

HTLV=

human T lymphotropic virus

IFN=

interferon

Ig=

immunoglobulin

IL=

interleukin

IMP3=

insulin-like growth factor II mRNA-binding protein 3

KIF20A=

kinesin family member 20A

KLH=

keyhole limpet hemocyanin

LY6K=

lymphocyte antigen 6 complex locus K

MAGE=

melanoma-associated antigen

MART-1=

melanoma antigen recognized by T cells 1

MDA=

melanoma differentiation antigen

MDS=

myelodysplastic syndrome

MIATA=

Minimal Information About T Cell Assays

MM=

multiple myeloma

MPHOSPH1=

M phase phosphoprotein 1

MPLA=

monophosphoryl lipid A

mTOR=

mammalian target of rapamycin

MUC1=

mucin 1

NSCLC=

non-small cell lung carcinoma patients

PMSA=

prostate membrane-specific antigen

polyICLC=

polyriboinosinic-polyribocytidylic acid stabilized with poly-L-lysine in carboxymethylcellulose

PSA=

prostate-specific antigen

RCC=

renal cell carcinoma

RHAMM=

receptor for hyaluronic acid-mediated motility

SART=

squamous cell carcinoma antigen recognized by T cells

SLP=

synthetic long peptide

TAA=

tumor-associated antigen

TARP=

T-cell receptor gamma chain alternate reading frame protein

TCR=

T-cell receptor

TLR=

Toll-like receptor

TRA=

tumor rejection antigen

Treg=

FOXP3+ regulatory T cells

URLC10=

upregulated in lung cancer 10

VEGF=

vascular endothelial growth factor

VEGFR=

VEGF receptor

WT1=

Wilms’ tumor 1

Acknowledgments

Authors are supported by the Ligue contre le Cancer (équipes labelisées), AXA Chair for Longevity Research, Cancéropôle Ile-de-France, Institut National du Cancer (INCa), Fondation Bettencourt-Schueller, Fondation de France, Fondation pour la Recherche Médicale, Agence National de la Recherche, the European Commission (Apo-Sys, ArtForce, ChemoRes. Death-Train) and the LabEx Immuno-Oncology.

References

  • Riedel S. Edward Jenner and the history of smallpox and vaccination. Proc (Bayl Univ Med Cent) 2005; 18:21 - 5; PMID: 16200144
  • Smith KA. Edward jenner and the small pox vaccine. Front Immunol 2011; 2:21; http://dx.doi.org/10.3389/fimmu.2011.00021; PMID: 22566811
  • Waldmann TA. Immunotherapy: past, present and future. Nat Med 2003; 9:269 - 77; http://dx.doi.org/10.1038/nm0303-269; PMID: 12612576
  • Smith KA. Louis pasteur, the father of immunology?. Front Immunol 2012; 3:68; http://dx.doi.org/10.3389/fimmu.2012.00068; PMID: 22566949
  • Plett PC. [Peter Plett and other discoverers of cowpox vaccination before Edward Jenner]. Sudhoffs Arch 2006; 90:219 - 32; PMID: 17338405
  • Breman JG, Arita I. The confirmation and maintenance of smallpox eradication. N Engl J Med 1980; 303:1263 - 73; http://dx.doi.org/10.1056/NEJM198011273032204; PMID: 6252467
  • Hoption Cann SA, van Netten JP, van Netten C. Dr William Coley and tumour regression: a place in history or in the future. Postgrad Med J 2003; 79:672 - 80; PMID: 14707241
  • Oblak A, Jerala R. Toll-like receptor 4 activation in cancer progression and therapy. Clin Dev Immunol 2011; 2011:609579; http://dx.doi.org/10.1155/2011/609579; PMID: 22110526
  • Lotze MT, Chang AE, Seipp CA, Simpson C, Vetto JT, Rosenberg SA. High-dose recombinant interleukin 2 in the treatment of patients with disseminated cancer. Responses, treatment-related morbidity, and histologic findings. JAMA 1986; 256:3117 - 24; http://dx.doi.org/10.1001/jama.1986.03380220083027; PMID: 3491225
  • Rosenberg SA, Yang JC, Topalian SL, Schwartzentruber DJ, Weber JS, Parkinson DR, et al. Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. JAMA 1994; 271:907 - 13; http://dx.doi.org/10.1001/jama.1994.03510360033032; PMID: 8120958
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:699 - 716; http://dx.doi.org/10.4161/onci.20696; PMID: 22934262
  • Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: FDA-approved Toll-like receptor agonists for cancer therapy. OncoImmunology 2012; 1; In press http://dx.doi.org/10.4161/onci.20931
  • Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, et al. Identification of a primary target of thalidomide teratogenicity. Science 2010; 327:1345 - 50; http://dx.doi.org/10.1126/science.1177319; PMID: 20223979
  • Finn OJ. Tumor immunology at the service of cancer immunotherapy. Curr Opin Immunol 2004; 16:127 - 9; http://dx.doi.org/10.1016/j.coi.2004.02.006; PMID: 15023402
  • Burgio GR. Commentary on the biological self: Toward a “Biological Ego”. From Garrod’s “chemical individuality” to Burnet’s “self”. Thymus 1990; 16:99 - 117; PMID: 2256127
  • Mason DW, Morris PJ. Effector mechanisms in allograft rejection. Annu Rev Immunol 1986; 4:119 - 45; http://dx.doi.org/10.1146/annurev.iy.04.040186.001003; PMID: 3518743
  • Janeway C. Immunogenicity signals 1,2,3 ... and 0. Immunol Today 1989; 10:283 - 6; http://dx.doi.org/10.1016/0167-5699(89)90081-9; PMID: 2590379
  • Matzinger P. Tolerance, danger, and the extended family. Annu Rev Immunol 1994; 12:991 - 1045; http://dx.doi.org/10.1146/annurev.iy.12.040194.005015; PMID: 8011301
  • Matzinger P. The danger model: a renewed sense of self. Science 2002; 296:301 - 5; http://dx.doi.org/10.1126/science.1071059; PMID: 11951032
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798
  • Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12:298 - 306; http://dx.doi.org/10.1038/nrc3245; PMID: 22419253
  • van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991; 254:1643 - 7; http://dx.doi.org/10.1126/science.1840703; PMID: 1840703
  • Parmiani G. Tumor immunity as autoimmunity: tumor antigens include normal self proteins which stimulate anergic peripheral T cells. Immunol Today 1993; 14:536 - 8; http://dx.doi.org/10.1016/0167-5699(93)90183-L; PMID: 8274196
  • Berzofsky JA, Ahlers JD, Belyakov IM. Strategies for designing and optimizing new generation vaccines. Nat Rev Immunol 2001; 1:209 - 19; http://dx.doi.org/10.1038/35105075; PMID: 11905830
  • Boon T, Cerottini JC, Van den Eynde B, van der Bruggen P, Van Pel A. Tumor antigens recognized by T lymphocytes. Annu Rev Immunol 1994; 12:337 - 65; http://dx.doi.org/10.1146/annurev.iy.12.040194.002005; PMID: 8011285
  • Gilboa E. The makings of a tumor rejection antigen. Immunity 1999; 11:263 - 70; http://dx.doi.org/10.1016/S1074-7613(00)80101-6; PMID: 10514004
  • Rosenberg SA. A new era for cancer immunotherapy based on the genes that encode cancer antigens. Immunity 1999; 10:281 - 7; http://dx.doi.org/10.1016/S1074-7613(00)80028-X; PMID: 10204484
  • Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 2007; 25:267 - 96; http://dx.doi.org/10.1146/annurev.immunol.25.022106.141609; PMID: 17134371
  • Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol 2009; 27:83 - 117; http://dx.doi.org/10.1146/annurev.immunol.021908.132544; PMID: 19007331
  • Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12:265 - 77; http://dx.doi.org/10.1038/nrc3258; PMID: 22437871
  • Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer 2012; 12:237 - 51; http://dx.doi.org/10.1038/nrc3237; PMID: 22437869
  • Mesri EA, Cesarman E, Boshoff C. Kaposi’s sarcoma and its associated herpesvirus. Nat Rev Cancer 2010; 10:707 - 19; http://dx.doi.org/10.1038/nrc2888; PMID: 20865011
  • Moody CA, Laimins LA. Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer 2010; 10:550 - 60; http://dx.doi.org/10.1038/nrc2886; PMID: 20592731
  • Moore PS, Chang Y. Why do viruses cause cancer? Highlights of the first century of human tumour virology. Nat Rev Cancer 2010; 10:878 - 89; http://dx.doi.org/10.1038/nrc2961; PMID: 21102637
  • Rees L, Tizard EJ, Morgan AJ, Cubitt WD, Finerty S, Oyewole-Eletu TA, et al. A phase I trial of epstein-barr virus gp350 vaccine for children with chronic kidney disease awaiting transplantation. Transplantation 2009; 88:1025 - 9; http://dx.doi.org/10.1097/TP.0b013e3181b9d918; PMID: 19855249
  • Klade CS, Wedemeyer H, Berg T, Hinrichsen H, Cholewinska G, Zeuzem S, et al. Therapeutic vaccination of chronic hepatitis C nonresponder patients with the peptide vaccine IC41. Gastroenterology 2008; 134:1385 - 95; http://dx.doi.org/10.1053/j.gastro.2008.02.058; PMID: 18471515
  • Yutani S, Komatsu N, Shichijo S, Yoshida K, Takedatsu H, Itou M, et al. Phase I clinical study of a peptide vaccination for hepatitis C virus-infected patients with different human leukocyte antigen-class I-A alleles. Cancer Sci 2009; 100:1935 - 42; http://dx.doi.org/10.1111/j.1349-7006.2009.01256.x; PMID: 19604246
  • Paavonen J, Naud P, Salmerón J, Wheeler CM, Chow SN, Apter D, et al, HPV PATRICIA Study Group. Efficacy of human papillomavirus (HPV)-16/18 AS04-adjuvanted vaccine against cervical infection and precancer caused by oncogenic HPV types (PATRICIA): final analysis of a double-blind, randomised study in young women. Lancet 2009; 374:301 - 14; http://dx.doi.org/10.1016/S0140-6736(09)61248-4; PMID: 19586656
  • FUTURE II Study Group. Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med 2007; 356:1915 - 27; http://dx.doi.org/10.1056/NEJMoa061741; PMID: 17494925
  • Agosti JM, Goldie SJ. Introducing HPV vaccine in developing countries--key challenges and issues. N Engl J Med 2007; 356:1908 - 10; http://dx.doi.org/10.1056/NEJMp078053; PMID: 17494923
  • Einstein MH, Baron M, Levin MJ, Chatterjee A, Edwards RP, Zepp F, et al, HPV-010 Study Group. Comparison of the immunogenicity and safety of Cervarix and Gardasil human papillomavirus (HPV) cervical cancer vaccines in healthy women aged 18-45 years. Hum Vaccin 2009; 5:705 - 19; http://dx.doi.org/10.4161/hv.5.10.9518; PMID: 19684472
  • Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011; 144:646 - 74; http://dx.doi.org/10.1016/j.cell.2011.02.013; PMID: 21376230
  • Nowell PC, Hungerford D. A minute chromosome in chronic granulocytic leukemia. Science 1960; 132:1497
  • Rowley JD. Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature 1973; 243:290 - 3; http://dx.doi.org/10.1038/243290a0; PMID: 4126434
  • Soussi T. TP53 mutations in human cancer: database reassessment and prospects for the next decade. Adv Cancer Res 2011; 110:107 - 39; http://dx.doi.org/10.1016/B978-0-12-386469-7.00005-0; PMID: 21704230
  • Liu ET. Functional genomics of cancer. Curr Opin Genet Dev 2008; 18:251 - 6; http://dx.doi.org/10.1016/j.gde.2008.07.014; PMID: 18691651
  • Mumberg D, Wick M, Schreiber H. Unique tumor antigens redefined as mutant tumor-specific antigens. Semin Immunol 1996; 8:289 - 93; http://dx.doi.org/10.1006/smim.1996.0037; PMID: 8956457
  • Passoni L, Gallo B, Biganzoli E, Stefanoni R, Massimino M, Di Nicola M, et al. In vivo T-cell immune response against anaplastic lymphoma kinase in patients with anaplastic large cell lymphomas. Haematologica 2006; 91:48 - 55; PMID: 16434370
  • Fossum B, Gedde-Dahl T 3rd, Breivik J, Eriksen JA, Spurkland A, Thorsby E, et al. p21-ras-peptide-specific T-cell responses in a patient with colorectal cancer. CD4+ and CD8+ T cells recognize a peptide corresponding to a common mutation (13Gly-->Asp). Int J Cancer 1994; 56:40 - 5; http://dx.doi.org/10.1002/ijc.2910560108; PMID: 7903287
  • Rusakiewicz S, Madrigal A, Travers P, Dodi AI. BCR/ABL-specific CD8+ T cells can be detected from CML patients, but are only expanded from healthy donors. Cancer Immunol Immunother 2009; 58:1449 - 57; http://dx.doi.org/10.1007/s00262-009-0703-x; PMID: 19360407
  • Yanuck M, Carbone DP, Pendleton CD, Tsukui T, Winter SF, Minna JD, et al. A mutant p53 tumor suppressor protein is a target for peptide-induced CD8+ cytotoxic T-cells. Cancer Res 1993; 53:3257 - 61; PMID: 7686815
  • Mayordomo JI, Loftus DJ, Sakamoto H, De Cesare CM, Appasamy PM, Lotze MT, et al. Therapy of murine tumors with p53 wild-type and mutant sequence peptide-based vaccines. J Exp Med 1996; 183:1357 - 65; http://dx.doi.org/10.1084/jem.183.4.1357; PMID: 8666894
  • Heimberger AB, Crotty LE, Archer GE, Hess KR, Wikstrand CJ, Friedman AH, et al. Epidermal growth factor receptor VIII peptide vaccination is efficacious against established intracerebral tumors. Clin Cancer Res 2003; 9:4247 - 54; PMID: 14519652
  • Carbone DP, Ciernik IF, Kelley MJ, Smith MC, Nadaf S, Kavanaugh D, et al. Immunization with mutant p53- and K-ras-derived peptides in cancer patients: immune response and clinical outcome. J Clin Oncol 2005; 23:5099 - 107; http://dx.doi.org/10.1200/JCO.2005.03.158; PMID: 15983396
  • Del Vecchio CA, Li G, Wong AJ. Targeting EGF receptor variant III: tumor-specific peptide vaccination for malignant gliomas. Expert Rev Vaccines 2012; 11:133 - 44; http://dx.doi.org/10.1586/erv.11.177; PMID: 22309662
  • Bendandi M. Idiotype vaccines for lymphoma: proof-of-principles and clinical trial failures. Nat Rev Cancer 2009; 9:675 - 81; http://dx.doi.org/10.1038/nrc2717; PMID: 19701243
  • Jerne NK. Towards a network theory of the immune system. Ann Immunol (Paris) 1974; 125C:373 - 89; PMID: 4142565
  • Siskind GW, Arreaza E. Network regulation of the immune response. Allergy Proc 1989; 10:387 - 91; http://dx.doi.org/10.2500/108854189778935872; PMID: 2697632
  • Lynch RG, Graff RJ, Sirisinha S, Simms ES, Eisen HN. Myeloma proteins as tumor-specific transplantation antigens. Proc Natl Acad Sci U S A 1972; 69:1540 - 4; http://dx.doi.org/10.1073/pnas.69.6.1540; PMID: 4113870
  • de Cerio AL, Zabalegui N, Rodríguez-Calvillo M, Inogés S, Bendandi M. Anti-idiotype antibodies in cancer treatment. Oncogene 2007; 26:3594 - 602; http://dx.doi.org/10.1038/sj.onc.1210371; PMID: 17530013
  • Peled JU, Kuang FL, Iglesias-Ussel MD, Roa S, Kalis SL, Goodman MF, et al. The biochemistry of somatic hypermutation. Annu Rev Immunol 2008; 26:481 - 511; http://dx.doi.org/10.1146/annurev.immunol.26.021607.090236; PMID: 18304001
  • Stevenson GT, Elliott EV, Stevenson FK. Idiotypic determinants on the surface immunoglobulin of neoplastic lymphocytes: a therapeutic target. Fed Proc 1977; 36:2268 - 71; PMID: 69552
  • Bendandi M, Gocke CD, Kobrin CB, Benko FA, Sternas LA, Pennington R, et al. Complete molecular remissions induced by patient-specific vaccination plus granulocyte-monocyte colony-stimulating factor against lymphoma. Nat Med 1999; 5:1171 - 7; http://dx.doi.org/10.1038/13928; PMID: 10502821
  • Miller RA, Maloney DG, Warnke R, Levy R. Treatment of B-cell lymphoma with monoclonal anti-idiotype antibody. N Engl J Med 1982; 306:517 - 22; http://dx.doi.org/10.1056/NEJM198203043060906; PMID: 6173751
  • Syrengelas AD, Chen TT, Levy R. DNA immunization induces protective immunity against B-cell lymphoma. Nat Med 1996; 2:1038 - 41; http://dx.doi.org/10.1038/nm0996-1038; PMID: 8782465
  • Mufson RA. Tumor antigen targets and tumor immunotherapy. Front Biosci 2006; 11:337 - 43; http://dx.doi.org/10.2741/1801; PMID: 16146735
  • Hoffmann TK, Donnenberg AD, Finkelstein SD, Donnenberg VS, Friebe-Hoffmann U, Myers EN, et al. Frequencies of tetramer+ T cells specific for the wild-type sequence p53(264-272) peptide in the circulation of patients with head and neck cancer. Cancer Res 2002; 62:3521 - 9; PMID: 12067999
  • Schuler PJ, Boeckers P, Engers R, Boelke E, Bas M, Greve J, et al. EGFR-specific T cell frequencies correlate with EGFR expression in head and neck squamous cell carcinoma. J Transl Med 2011; 9:168; http://dx.doi.org/10.1186/1479-5876-9-168; PMID: 21970318
  • Honnorat J, Viaccoz A. New concepts in paraneoplastic neurological syndromes. Rev Neurol (Paris) 2011; 167:729 - 36; http://dx.doi.org/10.1016/j.neurol.2011.08.001; PMID: 21890156
  • Pere H, Montier Y, Bayry J, Quintin-Colonna F, Merillon N, Dransart E, et al. A CCR4 antagonist combined with vaccines induces antigen-specific CD8+ T cells and tumor immunity against self antigens. Blood 2011; 118:4853 - 62; http://dx.doi.org/10.1182/blood-2011-01-329656; PMID: 21908423
  • Pere H, Tanchot C, Bayry J, Terme M, Taieb J, Badoual C, et al. Comprehensive analysis of current approaches to inhibit regulatory T cells in cancer. Oncoimmunology 2012; 1:326 - 33; http://dx.doi.org/10.4161/onci.18852; PMID: 22737608
  • Eura M, Chikamatsu K, Katsura F, Obata A, Sobao Y, Takiguchi M, et al. A wild-type sequence p53 peptide presented by HLA-A24 induces cytotoxic T lymphocytes that recognize squamous cell carcinomas of the head and neck. Clin Cancer Res 2000; 6:979 - 86; PMID: 10741724
  • Andrade Filho PA, López-Albaitero A, Gooding W, Ferris RL. Novel immunogenic HLA-A*0201-restricted epidermal growth factor receptor-specific T-cell epitope in head and neck cancer patients. J Immunother 2010; 33:83 - 91; http://dx.doi.org/10.1097/CJI.0b013e3181b8f421; PMID: 19952953
  • Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 2005; 5:615 - 25; http://dx.doi.org/10.1038/nrc1669; PMID: 16034368
  • Fijak M, Meinhardt A. The testis in immune privilege. Immunol Rev 2006; 213:66 - 81; http://dx.doi.org/10.1111/j.1600-065X.2006.00438.x; PMID: 16972897
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al, IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010; 363:411 - 22; http://dx.doi.org/10.1056/NEJMoa1001294; PMID: 20818862
  • Kroemer G, Zitvogel L. Can the exome and the immunome converge on the design of efficient cancer vaccines?. Oncoimmunology 2012; 1:579 - 80; http://dx.doi.org/10.4161/onci.20730; PMID: 22934249
  • Castle JC, Kreiter S, Diekmann J, Löwer M, van de Roemer N, de Graaf J, et al. Exploiting the mutanome for tumor vaccination. Cancer Res 2012; 72:1081 - 91; http://dx.doi.org/10.1158/0008-5472.CAN-11-3722; PMID: 22237626
  • Defrance T, Taillardet M, Genestier L. T cell-independent B cell memory. Curr Opin Immunol 2011; 23:330 - 6; http://dx.doi.org/10.1016/j.coi.2011.03.004; PMID: 21482090
  • Melief CJ, van der Burg SH. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat Rev Cancer 2008; 8:351 - 60; http://dx.doi.org/10.1038/nrc2373; PMID: 18418403
  • Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol 2003; 21:685 - 711; http://dx.doi.org/10.1146/annurev.immunol.21.120601.141040; PMID: 12615891
  • Probst HC, McCoy K, Okazaki T, Honjo T, van den Broek M. Resting dendritic cells induce peripheral CD8+ T cell tolerance through PD-1 and CTLA-4. Nat Immunol 2005; 6:280 - 6; http://dx.doi.org/10.1038/ni1165; PMID: 15685176
  • Stranges PB, Watson J, Cooper CJ, Choisy-Rossi CM, Stonebraker AC, Beighton RA, et al. Elimination of antigen-presenting cells and autoreactive T cells by Fas contributes to prevention of autoimmunity. Immunity 2007; 26:629 - 41; http://dx.doi.org/10.1016/j.immuni.2007.03.016; PMID: 17509906
  • Zou T, Caton AJ, Koretzky GA, Kambayashi T. Dendritic cells induce regulatory T cell proliferation through antigen-dependent and -independent interactions. J Immunol 2010; 185:2790 - 9; http://dx.doi.org/10.4049/jimmunol.0903740; PMID: 20686126
  • Galluzzi L, Kepp O, Kroemer G. Reverse Warburg: straight to cancer. Cell Cycle 2012; 11:1059; http://dx.doi.org/10.4161/cc.11.6.19746; PMID: 22343921
  • Galluzzi L, Vitale I, Kroemer G. Past, present, and future of molecular and cellular oncology. Front Oncol 2011; 1:1; http://dx.doi.org/10.3389/fonc.2011.00001; PMID: 22655224
  • Sakaguchi S, Miyara M, Costantino CM, Hafler DA. FOXP3+ regulatory T cells in the human immune system. Nat Rev Immunol 2010; 10:490 - 500; http://dx.doi.org/10.1038/nri2785; PMID: 20559327
  • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9:162 - 74; http://dx.doi.org/10.1038/nri2506; PMID: 19197294
  • Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12:253 - 68; http://dx.doi.org/10.1038/nri3175; PMID: 22437938
  • Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 2009; 27:451 - 83; http://dx.doi.org/10.1146/annurev.immunol.021908.132532; PMID: 19105661
  • Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008; 8:958 - 69; http://dx.doi.org/10.1038/nri2448; PMID: 19029990
  • Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial Watch: Dendritic cell-based interventions for cancer therapy. OncoImmunology 2012; 1 In press
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012; 1:306 - 15; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2012; 1:28 - 37; http://dx.doi.org/10.4161/onci.1.1.17938; PMID: 22720209
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, et al. Trial Watch: Prognostic and predictive value of the immune infiltrate in cancer. OncoImmunology 2012; 1 In press
  • Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2012; 1:493 - 506; http://dx.doi.org/10.4161/onci.20459; PMID: 22754768
  • Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2012; 1:179 - 88; http://dx.doi.org/10.4161/onci.1.2.19026; PMID: 22720239
  • Huff V. Wilms’ tumours: about tumour suppressor genes, an oncogene and a chameleon gene. Nat Rev Cancer 2011; 11:111 - 21; http://dx.doi.org/10.1038/nrc3002; PMID: 21248786
  • Kawakami M, Oka Y, Tsuboi A, Harada Y, Elisseeva OA, Furukawa Y, et al. Clinical and immunologic responses to very low-dose vaccination with WT1 peptide (5 microg/body) in a patient with chronic myelomonocytic leukemia. Int J Hematol 2007; 85:426 - 9; http://dx.doi.org/10.1532/IJH97.06194; PMID: 17562620
  • Busse A, Letsch A, Scheibenbogen C, Nonnenmacher A, Ochsenreither S, Thiel E, et al. Mutation or loss of Wilms’ tumor gene 1 (WT1) are not major reasons for immune escape in patients with AML receiving WT1 peptide vaccination. J Transl Med 2010; 8:5; http://dx.doi.org/10.1186/1479-5876-8-5; PMID: 20092642
  • Hashii Y, Sato E, Ohta H, Oka Y, Sugiyama H, Ozono K. WT1 peptide immunotherapy for cancer in children and young adults. Pediatr Blood Cancer 2010; 55:352 - 5; http://dx.doi.org/10.1002/pbc.22522; PMID: 20582983
  • Keilholz U, Letsch A, Busse A, Asemissen AM, Bauer S, Blau IW, et al. A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood 2009; 113:6541 - 8; http://dx.doi.org/10.1182/blood-2009-02-202598; PMID: 19389880
  • Greiner J, Schmitt A, Giannopoulos K, Rojewski MT, Götz M, Funk I, et al. High-dose RHAMM-R3 peptide vaccination for patients with acute myeloid leukemia, myelodysplastic syndrome and multiple myeloma. Haematologica 2010; 95:1191 - 7; http://dx.doi.org/10.3324/haematol.2009.014704; PMID: 20081055
  • Schmitt M, Schmitt A, Rojewski MT, Chen J, Giannopoulos K, Fei F, et al. RHAMM-R3 peptide vaccination in patients with acute myeloid leukemia, myelodysplastic syndrome, and multiple myeloma elicits immunologic and clinical responses. Blood 2008; 111:1357 - 65; http://dx.doi.org/10.1182/blood-2007-07-099366; PMID: 17978170
  • Osterborg A, Yi Q, Henriksson L, Fagerberg J, Bergenbrant S, Jeddi-Tehrani M, et al. Idiotype immunization combined with granulocyte-macrophage colony-stimulating factor in myeloma patients induced type I, major histocompatibility complex-restricted, CD8- and CD4-specific T-cell responses. Blood 1998; 91:2459 - 66; PMID: 9516146
  • Rasmussen T, Hansson L, Osterborg A, Johnsen HE, Mellstedt H. Idiotype vaccination in multiple myeloma induced a reduction of circulating clonal tumor B cells. Blood 2003; 101:4607 - 10; http://dx.doi.org/10.1182/blood-2002-06-1925; PMID: 12576327
  • McCormick AA, Reddy S, Reinl SJ, Cameron TI, Czerwinkski DK, Vojdani F, et al. Plant-produced idiotype vaccines for the treatment of non-Hodgkin’s lymphoma: safety and immunogenicity in a phase I clinical study. Proc Natl Acad Sci U S A 2008; 105:10131 - 6; http://dx.doi.org/10.1073/pnas.0803636105; PMID: 18645180
  • Schuster SJ, Neelapu SS, Gause BL, Janik JE, Muggia FM, Gockerman JP, et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. J Clin Oncol 2011; 29:2787 - 94; http://dx.doi.org/10.1200/JCO.2010.33.3005; PMID: 21632504
  • Li Z, Qiao Y, Liu B, Laska EJ, Chakravarthi P, Kulko JM, et al. Combination of imatinib mesylate with autologous leukocyte-derived heat shock protein and chronic myelogenous leukemia. Clin Cancer Res 2005; 11:4460 - 8; http://dx.doi.org/10.1158/1078-0432.CCR-05-0250; PMID: 15958631
  • Oki Y, McLaughlin P, Fayad LE, Pro B, Mansfield PF, Clayman GL, et al. Experience with heat shock protein-peptide complex 96 vaccine therapy in patients with indolent non-Hodgkin lymphoma. Cancer 2007; 109:77 - 83; http://dx.doi.org/10.1002/cncr.22389; PMID: 17133412
  • Meek DW, Marcar L. MAGE-A antigens as targets in tumour therapy. Cancer Lett 2012; 324:126 - 32; http://dx.doi.org/10.1016/j.canlet.2012.05.011; PMID: 22634429
  • Kufe DW. Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer 2009; 9:874 - 85; http://dx.doi.org/10.1038/nrc2761; PMID: 19935676
  • Vonderheide RH, Hahn WC, Schultze JL, Nadler LM. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity 1999; 10:673 - 9; http://dx.doi.org/10.1016/S1074-7613(00)80066-7; PMID: 10403642
  • Schmidt SM, Schag K, Müller MR, Weck MM, Appel S, Kanz L, et al. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood 2003; 102:571 - 6; http://dx.doi.org/10.1182/blood-2002-08-2554; PMID: 12576330
  • Disis ML, Gooley TA, Rinn K, Davis D, Piepkorn M, Cheever MA, et al. Generation of T-cell immunity to the HER-2/neu protein after active immunization with HER-2/neu peptide-based vaccines. J Clin Oncol 2002; 20:2624 - 32; http://dx.doi.org/10.1200/JCO.2002.06.171; PMID: 12039923
  • Disis ML, Rinn K, Knutson KL, Davis D, Caron D, dela Rosa C, et al. 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 - 50; http://dx.doi.org/10.1182/blood.V99.8.2845; PMID: 11929774
  • Salazar LG, Fikes J, Southwood S, Ishioka G, Knutson KL, Gooley TA, et al. Immunization of cancer patients with HER-2/neu-derived peptides demonstrating high-affinity binding to multiple class II alleles. Clin Cancer Res 2003; 9:5559 - 65; PMID: 14654536
  • Yajima N, Yamanaka R, Mine T, Tsuchiya N, Homma J, Sano M, et al. Immunologic evaluation of personalized peptide vaccination for patients with advanced malignant glioma. Clin Cancer Res 2005; 11:5900 - 11; http://dx.doi.org/10.1158/1078-0432.CCR-05-0559; PMID: 16115932
  • Brunsvig PF, Aamdal S, Gjertsen MK, Kvalheim G, Markowski-Grimsrud CJ, Sve I, et al. Telomerase peptide vaccination: a phase I/II study in patients with non-small cell lung cancer. Cancer Immunol Immunother 2006; 55:1553 - 64; http://dx.doi.org/10.1007/s00262-006-0145-7; PMID: 16491401
  • Ciocca DR, Frayssinet P, Cuello-Carrión FD. A pilot study with a therapeutic vaccine based on hydroxyapatite ceramic particles and self-antigens in cancer patients. Cell Stress Chaperones 2007; 12:33 - 43; http://dx.doi.org/10.1379/CSC-218R.1; PMID: 17441505
  • Meyer RG, Korn S, Micke P, Becker K, Huber C, Wölfel T, et al. An open-label, prospective phase I/II study evaluating the immunogenicity and safety of a ras peptide vaccine plus GM-CSF in patients with non-small cell lung cancer. Lung Cancer 2007; 58:88 - 94; http://dx.doi.org/10.1016/j.lungcan.2007.05.003; PMID: 17599645
  • Izumoto S, Tsuboi A, Oka Y, Suzuki T, Hashiba T, Kagawa N, et al. Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme. J Neurosurg 2008; 108:963 - 71; http://dx.doi.org/10.3171/JNS/2008/108/5/0963; PMID: 18447714
  • Brunsvig PF, Kyte JA, Kersten C, Sundstrøm S, Møller M, Nyakas M, et al. Telomerase peptide vaccination in NSCLC: a phase II trial in stage III patients vaccinated after chemoradiotherapy and an 8-year update on a phase I/II trial. Clin Cancer Res 2011; 17:6847 - 57; http://dx.doi.org/10.1158/1078-0432.CCR-11-1385; PMID: 21918169
  • Baselga J, Swain SM. Novel anticancer targets: revisiting ERBB2 and discovering ERBB3. Nat Rev Cancer 2009; 9:463 - 75; http://dx.doi.org/10.1038/nrc2656; PMID: 19536107
  • Pao W, Chmielecki J. Rational, biologically based treatment of EGFR-mutant non-small-cell lung cancer. Nat Rev Cancer 2010; 10:760 - 74; http://dx.doi.org/10.1038/nrc2947; PMID: 20966921
  • Choi BD, Archer GE, Mitchell DA, Heimberger AB, McLendon RE, Bigner DD, et al. EGFRvIII-targeted vaccination therapy of malignant glioma. Brain Pathol 2009; 19:713 - 23; http://dx.doi.org/10.1111/j.1750-3639.2009.00318.x; PMID: 19744042
  • Dutoit V, Herold-Mende C, Hilf N, Schoor O, Beckhove P, Bucher J, et al. Exploiting the glioblastoma peptidome to discover novel tumour-associated antigens for immunotherapy. Brain 2012; 135:1042 - 54; http://dx.doi.org/10.1093/brain/aws042; PMID: 22418738
  • Tyagi P, Mirakhur B. MAGRIT: the largest-ever phase III lung cancer trial aims to establish a novel tumor-specific approach to therapy. Clin Lung Cancer 2009; 10:371 - 4; http://dx.doi.org/10.3816/CLC.2009.n.052; PMID: 19808198
  • Mittendorf EA, Clifton GT, Holmes JP, Clive KS, Patil R, Benavides LC, et al. Clinical trial results of the HER-2/neu (E75) vaccine to prevent breast cancer recurrence in high-risk patients: from US Military Cancer Institute Clinical Trials Group Study I-01 and I-02. Cancer 2012; 118:2594 - 602; http://dx.doi.org/10.1002/cncr.26574; PMID: 21989902
  • Tsuruma T, Iwayama Y, Ohmura T, Katsuramaki T, Hata F, Furuhata T, et al. Clinical and immunological evaluation of anti-apoptosis protein, survivin-derived peptide vaccine in phase I clinical study for patients with advanced or recurrent breast cancer. J Transl Med 2008; 6:24; http://dx.doi.org/10.1186/1479-5876-6-24; PMID: 18471305
  • Morse MA, Secord AA, Blackwell K, Hobeika AC, Sinnathamby G, Osada T, et al. MHC class I-presented tumor antigens identified in ovarian cancer by immunoproteomic analysis are targets for T-cell responses against breast and ovarian cancer. Clin Cancer Res 2011; 17:3408 - 19; http://dx.doi.org/10.1158/1078-0432.CCR-10-2614; PMID: 21300761
  • Yerushalmi R, Tyldesley S, Kennecke H, Speers C, Woods R, Knight B, et al. Tumor markers in metastatic breast cancer subtypes: frequency of elevation and correlation with outcome. Ann Oncol 2012; 23:338 - 45; http://dx.doi.org/10.1093/annonc/mdr154; PMID: 21543625
  • Jiang XP, Yang DC, Elliott RL, Head JF. Vaccination with a mixed vaccine of autogenous and allogeneic breast cancer cells and tumor associated antigens CA15-3, CEA and CA125--results in immune and clinical responses in breast cancer patients. Cancer Biother Radiopharm 2000; 15:495 - 505; http://dx.doi.org/10.1089/cbr.2000.15.495; PMID: 11155821
  • Sandmaier BM, Oparin DV, Holmberg LA, Reddish MA, MacLean GD, Longenecker BM. Evidence of a cellular immune response against sialyl-Tn in breast and ovarian cancer patients after high-dose chemotherapy, stem cell rescue, and immunization with Theratope STn-KLH cancer vaccine. J Immunother 1999; 22:54 - 66; http://dx.doi.org/10.1097/00002371-199901000-00008; PMID: 9924700
  • Ghadersohi A, Chitta K, Greco WR, Harvey S, Winston J, Slocum H, et al. Tumor antigens and markers for breast and ovarian cancers. Front Biosci 2002; 7:e48 - 57; http://dx.doi.org/10.2741/ghader; PMID: 11815284
  • MacLean GD, Bowen-Yacyshyn MB, Samuel J, Meikle A, Stuart G, Nation J, et al. Active immunization of human ovarian cancer patients against a common carcinoma (Thomsen-Friedenreich) determinant using a synthetic carbohydrate antigen. J Immunother (1991) 1992; 11:292 - 305; http://dx.doi.org/10.1097/00002371-199205000-00008; PMID: 1599915
  • Tsuda N, Mochizuki K, Harada M, Sukehiro A, Kawano K, Yamada A, et al. Vaccination with predesignated or evidence-based peptides for patients with recurrent gynecologic cancers. J Immunother 2004; 27:60 - 72; http://dx.doi.org/10.1097/00002371-200401000-00006; PMID: 14676634
  • Vermeij R, Leffers N, Hoogeboom BN, Hamming IL, Wolf R, Reyners AK, et al. Potentiation of a p53-SLP vaccine by cyclophosphamide in ovarian cancer: a single-arm phase II study. Int J Cancer 2012; 131:E670 - 80; http://dx.doi.org/10.1002/ijc.27388; PMID: 22139992
  • Odunsi K, Matsuzaki J, Karbach J, Neumann A, Mhawech-Fauceglia P, Miller A, et al. Efficacy of vaccination with recombinant vaccinia and fowlpox vectors expressing NY-ESO-1 antigen in ovarian cancer and melanoma patients. Proc Natl Acad Sci U S A 2012; 109:5797 - 802; http://dx.doi.org/10.1073/pnas.1117208109; PMID: 22454499
  • Gates JD, Carmichael MG, Benavides LC, Holmes JP, Hueman MT, Woll MM, et al. Longterm followup assessment of a HER2/neu peptide (E75) vaccine for prevention of recurrence in high-risk prostate cancer patients. J Am Coll Surg 2009; 208:193 - 201; http://dx.doi.org/10.1016/j.jamcollsurg.2008.10.018; PMID: 19228530
  • Perez SA, Kallinteris NL, Bisias S, Tzonis PK, Georgakopoulou K, Varla-Leftherioti M, et al. Results from a phase I clinical study of the novel Ii-Key/HER-2/neu(776-790) hybrid peptide vaccine in patients with prostate cancer. Clin Cancer Res 2010; 16:3495 - 506; http://dx.doi.org/10.1158/1078-0432.CCR-10-0085; PMID: 20466887
  • Kouiavskaia DV, Berard CA, Datena E, Hussain A, Dawson N, Klyushnenkova EN, et al. Vaccination with agonist peptide PSA: 154-163 (155L) derived from prostate specific antigen induced CD8 T-cell response to the native peptide PSA: 154-163 but failed to induce the reactivity against tumor targets expressing PSA: a phase 2 study in patients with recurrent prostate cancer. J Immunother 2009; 32:655 - 66; http://dx.doi.org/10.1097/CJI.0b013e3181a80e0d; PMID: 19483644
  • Perambakam S, Hallmeyer S, Reddy S, Mahmud N, Bressler L, DeChristopher P, et al. Induction of specific T cell immunity in patients with prostate cancer by vaccination with PSA146-154 peptide. Cancer Immunol Immunother 2006; 55:1033 - 42; http://dx.doi.org/10.1007/s00262-005-0090-x; PMID: 16283303
  • Uenaka A, Wada H, Isobe M, Saika T, Tsuji K, Sato E, et al. T cell immunomonitoring and tumor responses in patients immunized with a complex of cholesterol-bearing hydrophobized pullulan (CHP) and NY-ESO-1 protein. Cancer Immun 2007; 7:9; PMID: 17441676
  • Kawabata R, Wada H, Isobe M, Saika T, Sato S, Uenaka A, et al. Antibody response against NY-ESO-1 in CHP-NY-ESO-1 vaccinated patients. Int J Cancer 2007; 120:2178 - 84; http://dx.doi.org/10.1002/ijc.22583; PMID: 17278093
  • Kawada J, Wada H, Isobe M, Gnjatic S, Nishikawa H, Jungbluth AA, et al. Heteroclitic serological response in esophageal and prostate cancer patients after NY-ESO-1 protein vaccination. Int J Cancer 2012; 130:584 - 92; http://dx.doi.org/10.1002/ijc.26074; PMID: 21413013
  • Slovin SF, Ragupathi G, Adluri S, Ungers G, Terry K, Kim S, et al. Carbohydrate vaccines in cancer: immunogenicity of a fully synthetic globo H hexasaccharide conjugate in man. Proc Natl Acad Sci U S A 1999; 96:5710 - 5; http://dx.doi.org/10.1073/pnas.96.10.5710; PMID: 10318949
  • Noguchi M, Itoh K, Suekane S, Morinaga A, Sukehiro A, Suetsugu N, et al. Immunological monitoring during combination of patient-oriented peptide vaccination and estramustine phosphate in patients with metastatic hormone refractory prostate cancer. Prostate 2004; 60:32 - 45; http://dx.doi.org/10.1002/pros.20011; PMID: 15129427
  • Noguchi M, Itoh K, Suekane S, Yao A, Suetsugu N, Katagiri K, et al. Phase I trial of patient-oriented vaccination in HLA-A2-positive patients with metastatic hormone-refractory prostate cancer. Cancer Sci 2004; 95:77 - 84; http://dx.doi.org/10.1111/j.1349-7006.2004.tb03174.x; PMID: 14720331
  • Noguchi M, Itoh K, Yao A, Mine T, Yamada A, Obata Y, et al. Immunological evaluation of individualized peptide vaccination with a low dose of estramustine for HLA-A24+ HRPC patients. Prostate 2005; 63:1 - 12; http://dx.doi.org/10.1002/pros.20157; PMID: 15378520
  • Noguchi M, Kobayashi K, Suetsugu N, Tomiyasu K, Suekane S, Yamada A, et al. Induction of cellular and humoral immune responses to tumor cells and peptides in HLA-A24 positive hormone-refractory prostate cancer patients by peptide vaccination. Prostate 2003; 57:80 - 92; http://dx.doi.org/10.1002/pros.10276; PMID: 12886526
  • Feyerabend S, Stevanovic S, Gouttefangeas C, Wernet D, Hennenlotter J, Bedke J, et al. Novel multi-peptide vaccination in Hla-A2+ hormone sensitive patients with biochemical relapse of prostate cancer. Prostate 2009; 69:917 - 27; http://dx.doi.org/10.1002/pros.20941; PMID: 19267352
  • Uemura H, Fujimoto K, Mine T, Uejima S, de Velasco MA, Hirao Y, et al. Immunological evaluation of personalized peptide vaccination monotherapy in patients with castration-resistant prostate cancer. Cancer Sci 2010; 101:601 - 8; http://dx.doi.org/10.1111/j.1349-7006.2009.01459.x; PMID: 20128819
  • Toffoli G, Cernigoi C, Russo A, Gallo A, Bagnoli M, Boiocchi M. Overexpression of folate binding protein in ovarian cancers. Int J Cancer 1997; 74:193 - 8; http://dx.doi.org/10.1002/(SICI)1097-0215(19970422)74:2<193::AID-IJC10>3.0.CO;2-F; PMID: 9133455
  • Wolfgang CD, Essand M, Lee B, Pastan I. T-cell receptor gamma chain alternate reading frame protein (TARP) expression in prostate cancer cells leads to an increased growth rate and induction of caveolins and amphiregulin. Cancer Res 2001; 61:8122 - 6; PMID: 11719440
  • Wolfgang CD, Essand M, Vincent JJ, Lee B, Pastan I. TARP: a nuclear protein expressed in prostate and breast cancer cells derived from an alternate reading frame of the T cell receptor gamma chain locus. Proc Natl Acad Sci U S A 2000; 97:9437 - 42; http://dx.doi.org/10.1073/pnas.160270597; PMID: 10931945
  • Slingluff CL Jr., Chianese-Bullock KA, Bullock TN, Grosh WW, Mullins DW, Nichols L, et al. Immunity to melanoma antigens: from self-tolerance to immunotherapy. Adv Immunol 2006; 90:243 - 95; http://dx.doi.org/10.1016/S0065-2776(06)90007-8; PMID: 16730266
  • Slingluff CL Jr., Yamshchikov G, Neese P, Galavotti H, Eastham S, Engelhard VH, et al. Phase I trial of a melanoma vaccine with gp100(280-288) peptide and tetanus helper peptide in adjuvant: immunologic and clinical outcomes. Clin Cancer Res 2001; 7:3012 - 24; PMID: 11595689
  • Pullarkat V, Lee PP, Scotland R, Rubio V, Groshen S, Gee C, et al. A phase I trial of SD-9427 (progenipoietin) with a multipeptide vaccine for resected metastatic melanoma. Clin Cancer Res 2003; 9:1301 - 12; PMID: 12684398
  • Slingluff CL Jr., Petroni GR, Yamshchikov GV, Barnd DL, Eastham S, Galavotti H, et al. Clinical and immunologic results of a randomized phase II trial of vaccination using four melanoma peptides either administered in granulocyte-macrophage colony-stimulating factor in adjuvant or pulsed on dendritic cells. J Clin Oncol 2003; 21:4016 - 26; http://dx.doi.org/10.1200/JCO.2003.10.005; PMID: 14581425
  • Wong R, Lau R, Chang J, Kuus-Reichel T, Brichard V, Bruck C, et al. Immune responses to a class II helper peptide epitope in patients with stage III/IV resected melanoma. Clin Cancer Res 2004; 10:5004 - 13; http://dx.doi.org/10.1158/1078-0432.CCR-04-0241; PMID: 15297401
  • Chen DS, Soen Y, Stuge TB, Lee PP, Weber JS, Brown PO, et al. Marked differences in human melanoma antigen-specific T cell responsiveness after vaccination using a functional microarray. PLoS Med 2005; 2:e265; http://dx.doi.org/10.1371/journal.pmed.0020265; PMID: 16162034
  • Markovic SN, Suman VJ, Ingle JN, Kaur JS, Pitot HC, Loprinzi CL, et al. Peptide vaccination of patients with metastatic melanoma: improved clinical outcome in patients demonstrating effective immunization. Am J Clin Oncol 2006; 29:352 - 60; http://dx.doi.org/10.1097/01.coc.0000217877.78473.a4; PMID: 16891861
  • Atzpodien J, Reitz M. GM-CSF plus antigenic peptide vaccination in locally advanced melanoma patients. Cancer Biother Radiopharm 2007; 22:551 - 5; http://dx.doi.org/10.1089/cbr.2007.376; PMID: 17803450
  • Powell DJ Jr., Felipe-Silva A, Merino MJ, Ahmadzadeh M, Allen T, Levy C, et al. Administration of a CD25-directed immunotoxin, LMB-2, to patients with metastatic melanoma induces a selective partial reduction in regulatory T cells in vivo. J Immunol 2007; 179:4919 - 28; PMID: 17878392
  • Bins A, Mallo H, Sein J, van den Bogaard C, Nooijen W, Vyth-Dreese F, et al. Phase I clinical study with multiple peptide vaccines in combination with tetanus toxoid and GM-CSF in advanced-stage HLA-A*0201-positive melanoma patients. J Immunother 2007; 30:234 - 9; http://dx.doi.org/10.1097/01.cji.0000211333.06762.47; PMID: 17471170
  • Slingluff CL Jr., Petroni GR, Chianese-Bullock KA, Smolkin ME, Hibbitts S, Murphy C, et al. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res 2007; 13:6386 - 95; http://dx.doi.org/10.1158/1078-0432.CCR-07-0486; PMID: 17975151
  • Nisticò P, Capone I, Palermo B, Del Bello D, Ferraresi V, Moschella F, et al. Chemotherapy enhances vaccine-induced antitumor immunity in melanoma patients. Int J Cancer 2009; 124:130 - 9; http://dx.doi.org/10.1002/ijc.23886; PMID: 18839429
  • Slingluff CL Jr., Petroni GR, Olson WC, Smolkin ME, Ross MI, Haas NB, et al. Effect of granulocyte/macrophage colony-stimulating factor on circulating CD8+ and CD4+ T-cell responses to a multipeptide melanoma vaccine: outcome of a multicenter randomized trial. Clin Cancer Res 2009; 15:7036 - 44; http://dx.doi.org/10.1158/1078-0432.CCR-09-1544; PMID: 19903780
  • Slingluff CL, Petroni GR, Smolkin ME, Chianese-Bullock KA, Smith K, Murphy C, et al. Immunogenicity for CD8+ and CD4+ T cells of 2 formulations of an incomplete freund’s adjuvant for multipeptide melanoma vaccines. J Immunother 2010; 33:630 - 8; http://dx.doi.org/10.1097/CJI.0b013e3181e311ac; PMID: 20551833
  • Schwartzentruber DJ, Lawson DH, Richards JM, Conry RM, Miller DM, Treisman J, et al. gp100 peptide vaccine and interleukin-2 in patients with advanced melanoma. N Engl J Med 2011; 364:2119 - 27; http://dx.doi.org/10.1056/NEJMoa1012863; PMID: 21631324
  • Pittet MJ, Speiser DE, Liénard D, Valmori D, Guillaume P, Dutoit V, et al. Expansion and functional maturation of human tumor antigen-specific CD8+ T cells after vaccination with antigenic peptide. Clin Cancer Res 2001; 7:Suppl 796s - 803s; PMID: 11300475
  • Ayyoub M, Zippelius A, Pittet MJ, Rimoldi D, Valmori D, Cerottini JC, et al. Activation of human melanoma reactive CD8+ T cells by vaccination with an immunogenic peptide analog derived from Melan-A/melanoma antigen recognized by T cells-1. Clin Cancer Res 2003; 9:669 - 77; PMID: 12576434
  • Cebon J, Jäger E, Shackleton MJ, Gibbs P, Davis ID, Hopkins W, et al. Two phase I studies of low dose recombinant human IL-12 with Melan-A and influenza peptides in subjects with advanced malignant melanoma. Cancer Immun 2003; 3:7; PMID: 12862418
  • Appay V, Voelter V, Rufer N, Reynard S, Jandus C, Gasparini D, et al. Combination of transient lymphodepletion with busulfan and fludarabine and peptide vaccination in a phase I clinical trial for patients with advanced melanoma. J Immunother 2007; 30:240 - 50; http://dx.doi.org/10.1097/01.cji.0000211332.68643.98; PMID: 17471171
  • Lienard D, Avril MF, Le Gal FA, Baumgaertner P, Vermeulen W, Blom A, et al. Vaccination of melanoma patients with Melan-A/Mart-1 peptide and Klebsiella outer membrane protein p40 as an adjuvant. J Immunother 2009; 32:875 - 83; http://dx.doi.org/10.1097/CJI.0b013e3181b56ad9; PMID: 19752746
  • Ribas A, Weber JS, Chmielowski B, Comin-Anduix B, Lu D, Douek M, et al. Intra-lymph node prime-boost vaccination against Melan A and tyrosinase for the treatment of metastatic melanoma: results of a phase 1 clinical trial. Clin Cancer Res 2011; 17:2987 - 96; http://dx.doi.org/10.1158/1078-0432.CCR-10-3272; PMID: 21385924
  • Speiser DE, Wieckowski S, Gupta B, Iancu EM, Baumgaertner P, Baitsch L, et al. Single cell analysis reveals similar functional competence of dominant and nondominant CD8 T-cell clonotypes. Proc Natl Acad Sci U S A 2011; 108:15318 - 23; http://dx.doi.org/10.1073/pnas.1105419108; PMID: 21876175
  • Scheibenbogen C, Schmittel A, Keilholz U, Allgäuer T, Hofmann U, Max R, et al. Phase 2 trial of vaccination with tyrosinase peptides and granulocyte-macrophage colony-stimulating factor in patients with metastatic melanoma. J Immunother 2000; 23:275 - 81; http://dx.doi.org/10.1097/00002371-200003000-00012; PMID: 10746554
  • Scheibenbogen C, Schadendorf D, Bechrakis NE, Nagorsen D, Hofmann U, Servetopoulou F, et al. Effects of granulocyte-macrophage colony-stimulating factor and foreign helper protein as immunologic adjuvants on the T-cell response to vaccination with tyrosinase peptides. Int J Cancer 2003; 104:188 - 94; http://dx.doi.org/10.1002/ijc.10961; PMID: 12569574
  • Khong HT, Yang JC, Topalian SL, Sherry RM, Mavroukakis SA, White DE, et al. Immunization of HLA-A*0201 and/or HLA-DPbeta1*04 patients with metastatic melanoma using epitopes from the NY-ESO-1 antigen. J Immunother 2004; 27:472 - 7; http://dx.doi.org/10.1097/00002371-200411000-00007; PMID: 15534491
  • Fourcade J, Kudela P, Andrade Filho PA, Janjic B, Land SR, Sander C, et al. Immunization with analog peptide in combination with CpG and montanide expands tumor antigen-specific CD8+ T cells in melanoma patients. J Immunother 2008; 31:781 - 91; http://dx.doi.org/10.1097/CJI.0b013e318183af0b; PMID: 18779741
  • Ebert LM, Liu YC, Clements CS, Robson NC, Jackson HM, Markby JL, et al. A long, naturally presented immunodominant epitope from NY-ESO-1 tumor antigen: implications for cancer vaccine design. Cancer Res 2009; 69:1046 - 54; http://dx.doi.org/10.1158/0008-5472.CAN-08-2926; PMID: 19176376
  • Kyte JA, Gaudernack G, Dueland S, Trachsel S, Julsrud L, Aamdal S. Telomerase peptide vaccination combined with temozolomide: a clinical trial in stage IV melanoma patients. Clin Cancer Res 2011; 17:4568 - 80; http://dx.doi.org/10.1158/1078-0432.CCR-11-0184; PMID: 21586625
  • Chen Q, Jackson H, Parente P, Luke T, Rizkalla M, Tai TY, et al. Immunodominant CD4+ responses identified in a patient vaccinated with full-length NY-ESO-1 formulated with ISCOMATRIX adjuvant. Proc Natl Acad Sci U S A 2004; 101:9363 - 8; http://dx.doi.org/10.1073/pnas.0403271101; PMID: 15197261
  • Davis ID, Chen W, Jackson H, Parente P, Shackleton M, Hopkins W, et al. Recombinant NY-ESO-1 protein with ISCOMATRIX adjuvant induces broad integrated antibody and CD4(+) and CD8(+) T cell responses in humans. Proc Natl Acad Sci U S A 2004; 101:10697 - 702; http://dx.doi.org/10.1073/pnas.0403572101; PMID: 15252201
  • Valmori D, Souleimanian NE, Tosello V, Bhardwaj N, Adams S, O’Neill D, et al. Vaccination with NY-ESO-1 protein and CpG in Montanide induces integrated antibody/Th1 responses and CD8 T cells through cross-priming. Proc Natl Acad Sci U S A 2007; 104:8947 - 52; http://dx.doi.org/10.1073/pnas.0703395104; PMID: 17517626
  • Cohen L, Parker PA, Sterner J, De Moor C. Quality of life in patients with malignant melanoma participating in a phase I trial of an autologous tumour-derived vaccine. Melanoma Res 2002; 12:505 - 11; http://dx.doi.org/10.1097/00008390-200209000-00013; PMID: 12394193
  • Dadaglio G, Morel S, Bauche C, Moukrim Z, Lemonnier FA, Van Den Eynde BJ, et al. Recombinant adenylate cyclase toxin of Bordetella pertussis induces cytotoxic T lymphocyte responses against HLA*0201-restricted melanoma epitopes. Int Immunol 2003; 15:1423 - 30; http://dx.doi.org/10.1093/intimm/dxg144; PMID: 14645151
  • Motta I, André F, Lim A, Tartaglia J, Cox WI, Zitvogel L, et al. Cross-presentation by dendritic cells of tumor antigen expressed in apoptotic recombinant canarypox virus-infected dendritic cells. J Immunol 2001; 167:1795 - 802; PMID: 11466405
  • Sato Y, Maeda Y, Shomura H, Sasatomi T, Takahashi M, Une Y, et al. A phase I trial of cytotoxic T-lymphocyte precursor-oriented peptide vaccines for colorectal carcinoma patients. Br J Cancer 2004; 90:1334 - 42; http://dx.doi.org/10.1038/sj.bjc.6601711; PMID: 15054451
  • Tsuruma T, Torigoe T, Hata F, Furuhata T, Sato N, Hirata K. [Anti-apoptosis protein, survivin-2B-derived peptide vaccine therapy]. Gan To Kagaku Ryoho 2004; 31:1634 - 6; PMID: 15553667
  • Wobser M, Keikavoussi P, Kunzmann V, Weininger M, Andersen MH, Becker JC. Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunol Immunother 2006; 55:1294 - 8; http://dx.doi.org/10.1007/s00262-005-0102-x; PMID: 16315030
  • Hattori T, Okuno K, Yoshida K, Kokubu T, Mine T, Yamada R, et al. [A phase I study of combination-therapy using personalized peptide vaccine and UFT/UZEL for advanced or recurrent colorectal cancer]. Gan To Kagaku Ryoho 2006; 33:1745 - 7; PMID: 17212094
  • Sato Y, Fujiwara T, Mine T, Shomura H, Homma S, Maeda Y, et al. Immunological evaluation of personalized peptide vaccination in combination with a 5-fluorouracil derivative (TS-1) for advanced gastric or colorectal carcinoma patients. Cancer Sci 2007; 98:1113 - 9; http://dx.doi.org/10.1111/j.1349-7006.2007.00498.x; PMID: 17459063
  • Wada H, Sato E, Uenaka A, Isobe M, Kawabata R, Nakamura Y, et al. Analysis of peripheral and local anti-tumor immune response in esophageal cancer patients after NY-ESO-1 protein vaccination. Int J Cancer 2008; 123:2362 - 9; http://dx.doi.org/10.1002/ijc.23810; PMID: 18729190
  • Takahashi N, Ohkuri T, Homma S, Ohtake J, Wakita D, Togashi Y, et al. First clinical trial of cancer vaccine therapy with artificially synthesized helper/ killer-hybrid epitope long peptide of MAGE-A4 cancer antigen. Cancer Sci 2012; 103:150 - 3; http://dx.doi.org/10.1111/j.1349-7006.2011.02106.x; PMID: 22221328
  • Kono K, Iinuma H, Akutsu Y, Tanaka H, Hayashi N, Uchikado Y, et al. Multicenter, phase II clinical trial of cancer vaccination for advanced esophageal cancer with three peptides derived from novel cancer-testis antigens. J Transl Med 2012; 10:141; http://dx.doi.org/10.1186/1479-5876-10-141; PMID: 22776426
  • Kono K, Mizukami Y, Daigo Y, Takano A, Masuda K, Yoshida K, et al. Vaccination with multiple peptides derived from novel cancer-testis antigens can induce specific T-cell responses and clinical responses in advanced esophageal cancer. Cancer Sci 2009; 100:1502 - 9; http://dx.doi.org/10.1111/j.1349-7006.2009.01200.x; PMID: 19459850
  • Muderspach L, Wilczynski S, Roman L, Bade L, Felix J, Small LA, et al. A phase I trial of a human papillomavirus (HPV) peptide vaccine for women with high-grade cervical and vulvar intraepithelial neoplasia who are HPV 16 positive. Clin Cancer Res 2000; 6:3406 - 16; PMID: 10999722
  • Ressing ME, van Driel WJ, Brandt RM, Kenter GG, de Jong JH, Bauknecht T, et al. Detection of T helper responses, but not of human papillomavirus-specific cytotoxic T lymphocyte responses, after peptide vaccination of patients with cervical carcinoma. J Immunother 2000; 23:255 - 66; http://dx.doi.org/10.1097/00002371-200003000-00010; PMID: 10746552
  • van Driel WJ, Ressing ME, Kenter GG, Brandt RM, Krul EJ, van Rossum AB, et al. Vaccination with HPV16 peptides of patients with advanced cervical carcinoma: clinical evaluation of a phase I-II trial. Eur J Cancer 1999; 35:946 - 52; http://dx.doi.org/10.1016/S0959-8049(99)00048-9; PMID: 10533477
  • Suekane S, Nishitani M, Noguchi M, Komohara Y, Kokubu T, Naitoh M, et al. Phase I trial of personalized peptide vaccination for cytokine-refractory metastatic renal cell carcinoma patients. Cancer Sci 2007; 98:1965 - 8; http://dx.doi.org/10.1111/j.1349-7006.2007.00631.x; PMID: 17919310
  • Walter S, Weinschenk T, Stenzl A, Zdrojowy R, Pluzanska A, Szczylik C, et al. Multipeptide immune response to cancer vaccine IMA901 after single-dose cyclophosphamide associates with longer patient survival. Nat Med 2012; In press http://dx.doi.org/10.1038/nm.2883; PMID: 22842478
  • Hoffman ES, Smith RE, Renaud RC Jr.. From the analyst’s couch: TLR-targeted therapeutics. Nat Rev Drug Discov 2005; 4:879 - 80; http://dx.doi.org/10.1038/nrd1880; PMID: 16299917
  • Kepp O, Galluzzi L, Martins I, Schlemmer F, Adjemian S, Michaud M, et al. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev 2011; 30:61 - 9; http://dx.doi.org/10.1007/s10555-011-9273-4; PMID: 21249425
  • Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011; 334:1573 - 7; http://dx.doi.org/10.1126/science.1208347; PMID: 22174255
  • Honma I, Kitamura H, Torigoe T, Takahashi A, Tanaka T, Sato E, et al. Phase I clinical study of anti-apoptosis protein survivin-derived peptide vaccination for patients with advanced or recurrent urothelial cancer. Cancer Immunol Immunother 2009; 58:1801 - 7; http://dx.doi.org/10.1007/s00262-009-0691-x; PMID: 19294381
  • Miyazaki A, Kobayashi J, Torigoe T, Hirohashi Y, Yamamoto T, Yamaguchi A, et al. Phase I clinical trial of survivin-derived peptide vaccine therapy for patients with advanced or recurrent oral cancer. Cancer Sci 2011; 102:324 - 9; http://dx.doi.org/10.1111/j.1349-7006.2010.01789.x; PMID: 21143701
  • Morita S, Oka Y, Tsuboi A, Kawakami M, Maruno M, Izumoto S, et al. A phase I/II trial of a WT1 (Wilms’ tumor gene) peptide vaccine in patients with solid malignancy: safety assessment based on the phase I data. Jpn J Clin Oncol 2006; 36:231 - 6; http://dx.doi.org/10.1093/jjco/hyl005; PMID: 16611662
  • Kitano S, Kageyama S, Nagata Y, Miyahara Y, Hiasa A, Naota H, et al. HER2-specific T-cell immune responses in patients vaccinated with truncated HER2 protein complexed with nanogels of cholesteryl pullulan. Clin Cancer Res 2006; 12:7397 - 405; http://dx.doi.org/10.1158/1078-0432.CCR-06-1546; PMID: 17189412
  • Kaumaya PT, Foy KC, Garrett J, Rawale SV, Vicari D, Thurmond JM, et al. Phase I active immunotherapy with combination of two chimeric, human epidermal growth factor receptor 2, B-cell epitopes fused to a promiscuous T-cell epitope in patients with metastatic and/or recurrent solid tumors. J Clin Oncol 2009; 27:5270 - 7; http://dx.doi.org/10.1200/JCO.2009.22.3883; PMID: 19752336
  • Jäger E, Gnjatic S, Nagata Y, Stockert E, Jäger D, Karbach J, et al. Induction of primary NY-ESO-1 immunity: CD8+ T lymphocyte and antibody responses in peptide-vaccinated patients with NY-ESO-1+ cancers. Proc Natl Acad Sci U S A 2000; 97:12198 - 203; http://dx.doi.org/10.1073/pnas.220413497; PMID: 11027314
  • Arlen P, Tsang KY, Marshall JL, Chen A, Steinberg SM, Poole D, et al. The use of a rapid ELISPOT assay to analyze peptide-specific immune responses in carcinoma patients to peptide vs. recombinant poxvirus vaccines. Cancer Immunol Immunother 2000; 49:517 - 29; http://dx.doi.org/10.1007/s002620000145; PMID: 11129322
  • Janetzki S, Palla D, Rosenhauer V, Lochs H, Lewis JJ, Srivastava PK. Immunization of cancer patients with autologous cancer-derived heat shock protein gp96 preparations: a pilot study. Int J Cancer 2000; 88:232 - 8; http://dx.doi.org/10.1002/1097-0215(20001015)88:2<232::AID-IJC14>3.0.CO;2-8; PMID: 11004674
  • Abrams SI, Khleif SN, Bergmann-Leitner ES, Kantor JA, Chung Y, Hamilton JM, et al. Generation of stable CD4+ and CD8+ T cell lines from patients immunized with ras oncogene-derived peptides reflecting codon 12 mutations. Cell Immunol 1997; 182:137 - 51; http://dx.doi.org/10.1006/cimm.1997.1224; PMID: 9514698
  • Mine T, Sato Y, Noguchi M, Sasatomi T, Gouhara R, Tsuda N, et al. Humoral responses to peptides correlate with overall survival in advanced cancer patients vaccinated with peptides based on pre-existing, peptide-specific cellular responses. Clin Cancer Res 2004; 10:929 - 37; http://dx.doi.org/10.1158/1078-0432.CCR-1117-3; PMID: 14871969
  • Khleif SN, Abrams SI, Hamilton JM, Bergmann-Leitner E, Chen A, Bastian A, et al. A phase I vaccine trial with peptides reflecting ras oncogene mutations of solid tumors. J Immunother 1999; 22:155 - 65; http://dx.doi.org/10.1097/00002371-199903000-00007; PMID: 10093040
  • Tanaka S, Harada M, Mine T, Noguchi M, Gohara R, Azuma K, et al. Peptide vaccination for patients with melanoma and other types of cancer based on pre-existing peptide-specific ctotoxic T-lymphocyte precursors in the periphery. J Immunother 2003; 26:357 - 66; http://dx.doi.org/10.1097/00002371-200307000-00008; PMID: 12843798
  • Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, et al. Phase I immunotherapeutic trial with long peptides spanning the E6 and E7 sequences of high-risk human papillomavirus 16 in end-stage cervical cancer patients shows low toxicity and robust immunogenicity. Clin Cancer Res 2008; 14:169 - 77; http://dx.doi.org/10.1158/1078-0432.CCR-07-1881; PMID: 18172268
  • Hale DF, Clifton GT, Sears AK, Vreeland TJ, Shumway N, Peoples GE, et al. Cancer vaccines: should we be targeting patients with less aggressive disease?. Expert Rev Vaccines 2012; 11:721 - 31; http://dx.doi.org/10.1586/erv.12.39; PMID: 22873128
  • Levy A, Massard C, Gross-Goupil M, Fizazi K. Carcinomas of an unknown primary site: a curable disease?. Ann Oncol 2008; 19:1657 - 8; http://dx.doi.org/10.1093/annonc/mdn430; PMID: 18647966
  • Britten CM, Janetzki S, Butterfield LH, Ferrari G, Gouttefangeas C, Huber C, et al. T cell assays and MIATA: the essential minimum for maximum impact. Immunity 2012; 37:1 - 2; http://dx.doi.org/10.1016/j.immuni.2012.07.010; PMID: 22840835
  • Britten CM, Janetzki S, van der Burg SH, Huber C, Kalos M, Levitsky HI, et al. Minimal information about T cell assays: the process of reaching the community of T cell immunologists in cancer and beyond. Cancer Immunol Immunother 2011; 60:15 - 22; http://dx.doi.org/10.1007/s00262-010-0940-z; PMID: 21080166
  • Janetzki S, Britten CM, Kalos M, Levitsky HI, Maecker HT, Melief CJ, et al. “MIATA”-minimal information about T cell assays. Immunity 2009; 31:527 - 8; http://dx.doi.org/10.1016/j.immuni.2009.09.007; PMID: 19833080