2,259
Views
92
CrossRef citations to date
0
Altmetric
Review

Trial Watch

Peptide vaccines in cancer therapy

, , , , , , , & show all
Article: e26621 | Received 25 Sep 2013, Accepted 26 Sep 2013, Published online: 04 Nov 2013

Abstract

Throughout the past 3 decades, along with the recognition that the immune system not only influences oncogenesis and tumor progression, but also determines how established neoplastic lesions respond therapy, renovated enthusiasm has gathered around the possibility of using vaccines as anticancer agents. Such an enthusiasm quickly tempered when it became clear that anticancer vaccines would have to be devised as therapeutic, rather than prophylactic, measures, and that malignant cells often fail to elicit (or actively suppress) innate and adaptive immune responses. Nonetheless, accumulating evidence indicates that a variety of anticancer vaccines, including cell-based, DNA-based, and purified component-based preparations, are capable of circumventing the poorly immunogenic and highly immunosuppressive nature of most tumors and elicit (at least under some circumstances) therapeutically relevant immune responses. Great efforts are currently being devoted to the identification of strategies that may provide anticancer vaccines with the capacity of breaking immunological tolerance and eliciting tumor-associated antigen-specific immunity in a majority of patients. In this sense, promising results have been obtained by combining anticancer vaccines with a relatively varied panels of adjuvants, including multiple immunostimulatory cytokines, Toll-like receptor agonists as well as inhibitors of immune checkpoints. One year ago, in the December issue of OncoImmunology, we discussed the biological mechanisms that underlie the antineoplastic effects of peptide-based vaccines and presented an abundant literature demonstrating the prominent clinical potential of such an approach. Here, we review the latest developments in this exciting area of research, focusing on high-profile studies that have been published during the last 13 mo and clinical trials launched in the same period to evaluate purified peptides or full-length proteins as therapeutic anticancer agents.

Introduction

Prophylactic vaccination constitutes one of the major achievements in the history of medicine, Indeed, thanks to the pioneer work of the English physician Edward Anthony Jenner (1749–1823)Citation1-Citation3 and other prominent scientists including the French microbiologist Louis Pasteur (1822–1895),Citation1,Citation4 the administration of attenuated infectious agents (or purified components thereof) in the presence of adequate immunostimulatory agents (commonly referred to as adjuvants) is nowadays employed as part of routine, nationwide programs of disease prevention.Citation1 The most significant objective attained by the implementation of these global vaccination campaigns (starting in UK in 1840, with the promulgation of the Vaccination Act) is undoubtedly constituted by the eradication of natural smallpox sources, which was first certified by a committee of experts in 1979 and confirmed by the WHO 1 y later.Citation5 In addition, nationwide vaccination programs have dramatically abated the incidence of several other life-threatening infectious diseases including rabies, typhoid, cholera, measles, plague, chickenpox, mumps, poliomyelitis, and hepatitis B.Citation1

The possibility that patients affected by neoplastic diseases might also benefit from active immunotherapy, i.e., the elicitation of an endogenous adaptive immune response, first surged at the end of the 19th century (that is, nearly 1 century after Jenner’s experiments), along with the work of Paul Ehrlich, an German physician, and William Bradley Coley, an American surgeon.Citation1,Citation6 Erlich, who is best known for the concept of a “magic bullet” that would specifically eliminate malignant cells while sparing their non-transformed counterparts, attempted intensively to generate an anticancer vaccine by using attenuated cancer cells, with no success.Citation1,Citation6 Conversely, Coley developed a preparation of heat-killed Streptococcus pyogenes and Serratia marcescens bacteria (best known as the Coley toxin) that soon turned out to mediate potent antineoplastic effects.Citation7-Citation9 Nonetheless, the Coley toxin does not constitute an anticancer vaccine sensu stricto, but rather acts as an adjuvant, hence stimulating the maturation of dendritic cells (DCs) via Toll-like receptor (TLR)-dependent signal transduction cascades.Citation10

In (large) part owing to the broad acceptance of the “self/non-self” model, as posited by Sir Frank Macfarlane Burnet (an Australian virologist) in 1949,Citation11 the possibility that attenuated neoplastic cells or components thereof might induce clinically relevant anticancer responses was disregarded until the mid-1990s, when the American scientist Polly Matzinger first proposed the so-called “danger theory.”Citation12 According to this model, the immune system does not react against non-self entities while sparing self ones, but rather responds to states of danger, irrespective of whether they are elicited by non-self or self constituents.Citation12,Citation13 Together with the identification of the first antigen specifically expressed by transformed (as opposed to normal) cellsCitation14 and the characterization of tumor-specific cytotoxic T lymphocytes (CTLs) in the circulation of cancer patients,Citation14,Citation15 the danger theory de facto laid the foundations of modern tumor immunology, alongside resurrecting the interest of researchers and clinicians in the use of vaccines as antineoplastic interventions.

Throughout the last 2 decades, a wide panel of preparations has been tested for their ability to elicit tumor-associated antigen (TAA)-specific immune responses and mediate robust antineoplastic effects in vivo, encompassing (but not limited to): (1) recombinant TAAs, in the form of either short synthetic peptides, which are expected to bind MHC molecules on the surface of antigen-presenting cells (APCs) and hence be directly presented to T cells, or full-length proteins, the presentation of which relies on the uptake and processing by APCs; (2) cancer cell lysates, containing TAAs alone or complexed with chaperones; (3) TAA-encoding vectors, in the form of naked DNA or RNA molecules as well as of viral delivery systems; (4) DC-based vaccines, including DCs loaded with TAAs ex vivo as well as fusion proteins that allow for the selective delivery of TAAs to DCs in vivo.Citation6,Citation16-Citation21 Such an intense wave of investigation has generated profound insights into the molecular and cellular mechanisms that control the elicitation of anticancer immune responses. In addition, it has prompted the development of several vaccines that are currently being investigated in the clinic, in patients affected by a large panel of neoplasms.Citation6,Citation16-Citation18 Nonetheless, with the notable exceptions of Cervarix® and Gardasil®, 2 multivalent vaccines that have been approved by the US FDA in 2009 as prophylactic measures against infection by human papillomavirus (HPV) type 16 and 18 and the consequent development of cervical carcinoma,Citation22-Citation24 no vaccines based on recombinant TAAs are currently licensed for use in subjects affected by malignant conditions. Along similar lines, no DNA-based preparation is nowadays approved by international regulatory agencies for use as prophylactic or immunotherapeutic intervention against cancer in humans, and only one cell-based product is, i.e., sipuleucel-T (also known as Provenge®), which has been licensed for the treatment of patients with asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer in 2010.Citation25,Citation26 Conversely, Oncept®, a DNA-based vaccine coding for human tyrosinase (a differentiation-associated TAA),Citation27-Citation29 is currently approved by the US Department of Agriculture for the treatment of canine melanoma.Citation30

The paucity of clinically effective anticancer vaccines of all types, in particular recombinant ones, is in stark contrast with the hundreds of preparations that have been developed and commercialized throughout the last century for the prevention of infectious diseases. In fact, the development of effective anticancer vaccines may be problematic due to (1) issues related to the intrinsically low antigenicity of transformed cells; (2) the fact that anticancer vaccines are expected to operate most often as therapeutic—as opposed to prophylactic—agents; and (3) the capacity of neoplastic cells to establish robust immunosuppressive networks, at both local and systemic levels. We dissected these points in detail one year ago, in the December issue of OncoImmunology, when we first discussed clinical efforts aimed at testing TAA-derived peptides and full-length TAAs as therapeutic anticancer agents.Citation6 Multiple factors have been shown to influence the therapeutic potential of this approach, including not only the TAA of choice, but also the vaccine administration schedule and route as well as the presence and type of adjuvants. A detailed discussion of these parameters can be found in Refs. Citation6,Citation18,Citation31-Citation33.

Here, along the lines of our monthly Trial Watch series,Citation34-Citation39 we summarize the latest advances in the development of peptide-based vaccines, focusing on preclinical studies that have been published during the last 13 mo and clinical trials initiated in the same period to evaluate this approach as a therapeutic intervention against cancer.

Literature Update

During the last 13 mo, the results of no less than 22 clinical studies investigating the therapeutic potential of full length TAAs or peptides thereof in cancer patients have been published in peer-reviewed scientific journals (source http://www.ncbi.nlm.nih.gov/pubmed). With a single exception, a clinical trial testing the safety and antineoplastic activity of autologous heat-shock protein-TAAs complexes in 12 patients affected by high-grade glioma,Citation40 all these studies involved recombinant TAA-derived peptides.Citation41-Citation63 In a majority of settings, peptides were given as standalone therapeutic interventions, most frequently in incomplete Freund’s adjuvant (IFA, a water in oil emulsion).Citation64,Citation65 Alternatively, peptide-based vaccination was combined with additional adjuvant-like interventions, including CpG oligodeoxynucleotides, which act as TLR9 agonists;Citation48,Citation66,Citation67 polyinosinic:polycytidylic acid, poly-l-lysine, and carboxymethylcellulose (polyICLC), which triggers TLR3 signaling;Citation58,Citation68,Citation69 imiquimod, a TLR7 agonist;Citation49 interferon (IFN)α, a potent immunostimulatory cytokine,Citation42,Citation47,Citation62,Citation70-Citation73 and low-dose interleukin (IL)-2 combined with metronomic cyclophosphamide, a regimen that exerts robust immunogenic effects.Citation50,Citation74-Citation77

These studies were performed on patients affected by a large panel of solid (but not hematological) neoplasms, including glioma,Citation40 lung carcinoma,Citation48,Citation52 sarcoma,Citation59,Citation62 melanoma,Citation41,Citation50,Citation61 esophageal squamous cell carcinoma,Citation51 gastric cancer,Citation55 hepatocellular carcinoma,Citation43,Citation56,Citation60 pancreatic cancer,Citation47 colorectal carcinoma,Citation42,Citation45,Citation53 metastatic renal cell carcinoma,Citation49,Citation54 castration-resistant prostate cancer,Citation26,Citation46,Citation49 ovarian carcinoma,Citation58 gynecologic malignancies,Citation63 and various other tumors.Citation44,Citation57 The TAAs specifically targeted in these clinical trials (most of which were Phase I studies) encompassed cancer-testis antigens such as NY-ESO-1,Citation46,Citation48,Citation50,Citation58,Citation78 TTK protein kinase (also known as MOS),Citation51,Citation52,Citation79 lymphocyte antigen 6 complex, locus K (LY6K, best known as URLC10),Citation51,Citation52,Citation55,Citation80,Citation81 insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3, best known as IMP3),Citation51,Citation82,Citation83 ring finger protein 43 (RNF43),Citation45,Citation53,Citation84-Citation86 and translocase of outer mitochondrial membrane 34 (TOMM34);Citation45,Citation53,Citation87,Citation88 carcinoembryonic antigens like glypican-3;Citation43,Citation56,Citation60,Citation89,Citation90 differentiation antigens such as melan-A (MLANA) and premelanosome protein (PMEL, best known as gp100);Citation50,Citation91-Citation93 tumor-restricted TAAs, such as the SYT-SSX fusion (which is selectively expressed by synovial sarcomas as a result of a t(X;18)(p11;q11) chromosomal translocation);Citation62,Citation94,Citation95 as well as so-called “shared TAAs” (antigens that are overexpressed by malignant cells but also produced in normal amounts by one or several healthy tissues), including vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2,Citation44,Citation52-Citation54,Citation57,Citation96,Citation97 survivin,Citation47,Citation50,Citation61,Citation98,Citation99 Wilms tumor 1 (WT1),Citation63,Citation100,Citation101 telomerase reverse transcriptase (TERT),Citation49,Citation102 and p53.Citation42,Citation103,Citation104 Thus, a relatively heterogeneous panel of approaches and indications has been explored during the last 13 mo to get further insights into the safety and clinical potential of peptide-based anticancer vaccines.

Taken together, the results of these clinical trials demonstrate that the local administration of recombinant TAAs or peptides thereof is generally well tolerated by cancer patients, the most common adverse effects being grade 1–3 erythema or induration at the injection site,Citation44,Citation51,Citation53,Citation54,Citation61,Citation63 grade 1–2 proteinuria,Citation44,Citation57 and hypertension.Citation44,Citation57 Moreover, the authors of most of these studies could document the ability of TAA-derived peptides to elicit an immune response, most often as (1) an increase in circulating TAA-specific CTLs,Citation42,Citation43,Citation45-Citation54,Citation56-Citation58,Citation61,Citation94 (2) a surge in serum TAA-targeting antibodies,Citation42,Citation58 (3) a delayed type hypersensitivity (DTH) reaction developing at the injection site;Citation55,Citation62,Citation63,Citation94 (4) the infiltration of neoplastic lesions by CD4+ or CD8+ T lymphocytes,Citation40,Citation60 or (5) the activation of transcriptional programs associated with CTL effector functions in peripheral blood mononuclear cells.Citation44 Often, but not always,Citation50 such an immune response to vaccination was associated with (at least some degree of) clinical benefit.Citation40,Citation47,Citation51-Citation54,Citation56,Citation61 This correlation was particularly robust in the Phase II study published by Becker and collaborators, pointing to survivin-specific T-cell reactivity as an independent predictor of survival in a cohort of metastatic melanoma patients receiving survivin-derived peptides.Citation61 Along similar lines, Miyatake and colleagues reported a statistically significant correlation between the development of a WT1-specific DTH reaction upon the administration of WT1-derived peptides and the overall survival of patients with gynecologic malignancies.Citation63 These findings add to the large amount of clinical data suggesting that the implementation of appropriate immunomonitoring procedures is of the utmost importance not only to correctly interpret the results of clinical trials testing immunotherapeutic anticancer regimens, but also to prospectively identify the subsets of patients who are likely to obtain (at least some degree of) clinical benefit from treatment.Citation105,Citation106 This said, the possibility that immunological responders might exhibit a survival advantage over non-responders irrespective of vaccination has not yet been ruled out.

We have also found of particular interest a few recent studies that investigated fundamental aspects of the molecular and cellular circuitries underlying the therapeutic efficacy of TAA-derived anticancer vaccines. Salerno et al. revealed that activated (CD69+) effector memory CD8+ T cells expressing chemokine (C-X-C motif) receptor 3 (CXCR3) as well as retention molecules such as integrins αEβ7 and α1β1 tend to accumulate at recall vaccination sites irrespective of the presence of TAA-derived peptides, presumably owing to the immunostimulatory effect of IFA.Citation41 As these cells appear to be strikingly dysfunctional, producing minimal amounts of IFNγ in response to stimulation, the transient immune reactions and relatively low rates of clinical responses associated with (at least some) IFA-adjuvanted peptide vaccines may reflect the capacity of IFA to recruit and retain effector T cells, eventually resulting in their functional impairment.Citation41 In line with this notion, Hailemichael and collaborators demonstrated that TAA-specific CD8+ T lymphocytes accumulate at vaccination sites to become functionally impaired and undergo antigen-driven, IFNγ- and FAS ligand (FASL)-dependent apoptosis.Citation107 In this setting, the provision of CD40 agonist antibodies, a TLR7 agonist or IL-2 limited the demise of T cells but not their accumulation at vaccination sites. Conversely, the capacity of TAA-derived peptides to elicit robust anticancer immune responses was significantly ameliorated when short-lived vaccines were employed, a notion that may have important therapeutic implications.Citation107 Ma and colleagues developed a molecular platform for the selective delivery of anticancer peptide vaccines to B lymphocytes.Citation108 To this aim, they fused the extracellular domain of v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2, also known as HER2/neu) to a single chain variable fragment specific for CD19. Such a chimeric peptide activated B cells to produce high titers of ERBB2-specific antibodies and elicit robust CD4+ helper T-cell responses, hence supporting the activation and differentiation of CD8+ T cells and mediating antineoplastic effects in vivo.Citation108 These findings suggest that the therapeutic efficacy of anticancer peptide vaccines may be enhanced by strategies that allow for the simultaneous activation of B and T lymphocytes. The importance of humoral immunity in cancer immunosurveillance is further underlined by recent results from Zhang and collaborators, demonstrating that a multipeptide vaccine based on B-cell epitopes from heparanase (a metastasis-associated antigen)Citation109 exerts prominent antimetastatic effects in vivo.Citation110 Finally, Dosset et al. obtained further data in support of the notion that TERT elicits CTL responses in patients affected by a wide panel of solid and malignancies.Citation111,Citation112 In particular, these authors demonstrated that a mixture of TERT-derived peptides can induce high-avidity CD4+ T-cell responses in vivo, resulting in the breakdown of tolerance and the activation of therapeutic CTL antitumor reactivity.Citation111,Citation112

TERT-targeting anticancer vaccines have spurred great interest among clinicians throughout the past decade, as demonstrated by the consistent number of Phase I-II clinical trials completed in this period.Citation49,Citation102,Citation112-Citation134 In spite of encouraging safety data and clinical findings, however, only a few Phase III trials have been launched to test the antineoplastic potential of TAA-derived peptides in cancer patients, including TELOVAC (NCT00425360), a randomized study testing a promiscuous class II epitope of TERT (TERT611–626, best known as GV1001) in patients with locally advanced or metastatic pancreatic cancer previously treated with gemcitabine and capecitabine,Citation135,Citation136 and DERMA (NCT00796445), a randomized, double-blind, placebo-controlled study investigating the efficacy of recombinant melanoma antigen, family A, 3 (MAGEA3) administered in AS15 (a liposomal formulation of QS-21 Stimulon®, monophosphoryl lipid A and CpG-7909, a TLR9 agonist) as adjuvant therapy to patients with Stage IIIB/C resected MAGEA3+ melanoma.Citation137,Citation138 As recently disclosed at the Annual Meeting of the American Society of Clinical Oncology (ASCO) held in Chicago last June, the administration of GV1001 fails to ameliorate the overall survival of pancreatic cancer patients previously treated with conventional chemotherapy.Citation135 However, the CEO of GemVax and KAEL Co., the company that is currently developing GV1001, declared that 2 candidate biomarkers were identified that may predict immunological responses to the vaccine as well as increased survival in a subset of patients (source http://online.wsj.com/article/PR-CO-20130603-906288.html). On 2013, September 5th, GlaxoSmithKline plc announced that the DERMA study also failed to meet its first co-primary endpoint as recombinant MAGEA3 administered in AS15 did not extend the disease-free survival of patients with Stage IIIB/C resected MAGEA3+ melanoma as compared with placebo (source http://www.gsk.com/media/press-releases/2013/the-investigational-mage-a3-antigen-specific-cancer-immunotherap.html). Nonetheless, following the Independent Data Monitoring Committee’s (IDMC) unanimous recommendation, the DERMA study will be protracted until the assessment of the second co-primary endpoint, i.e., disease-free survival in a patient subset characterized by a 84-component gene signature that may predict the clinical efficacy of recombinant MAGEA3.Citation138 The preliminary results of these randomized studies lend further support to the notion that clinical immunomonitoring may constitute a critical step for the development of novel anticancer immunotherapeutics (see above).

Update on Clinical Trials

When our latest Trial Watch dealing with the use of recombinant TAAs or peptides thereof as anticancer vaccines was submitted for publication to OncoImmunology (September, 2012), official sources listed 118 recent (started after 2008, January 1st) clinical trials (not withdrawn, terminated or completed at the day of submission) that would assess the safety and efficacy of this immunotherapeutic strategy in cancer patients.Citation6 Of these studies, 11 involved patients affected by hematological neoplasms, 30 subjects bearing neurological or pulmonary malignancies, 24 individuals with breast, ovarian or prostate carcinomas, 25 melanoma patients, 15 subjects affected by esophageal, gastric, pancreatic or colorectal tumors, 10 patients bearing bladder carcinoma or neoplasms of the reproductive tract, and 12 individuals with other tumors or mixed patients cohorts. The status of most of these trials has remained unchanged since, exception made for NCT01232712, NCT00633724, NCT00655785, NCT00711334, NCT00773097, NCT00874588, NCT00887796, NCT00892567, NCT00952692, NCT01003808, NCT01219348, and NCT01673217 (all of which have been completed), as well as of NCT00706992 (which has been terminated) (source http://www.clinicaltrials.gov). Of note, the preliminary results of this latter Phase II study, demonstrating the ability of T cells genetically engineered to express a MLANA-targeting T-cell receptor (TCR) coupled to a MLANA-derived peptide to induce objective clinical responses in 4 out of 31 high-risk melanoma patients, had first been reported as early as in 2006.Citation139,Citation140 Along similar lines, the findings of NCT00633724 and NCT00874588 (testing 4 HLA-A*2402-restricted TAA-derived peptides in non-small cell lung carcinoma, NSCLC, patients), NCT00952692 (investigating the clinical profile of a truncated variant of ERBB2 in women with metastatic breast carcinoma) as well as NCT01219348 (assessing the safety and efficacy of an indoleamine 2,3-dioxygenase-derived peptide in NSCLC patients) have already been published, either in the form of a scientific report,Citation52,Citation141 or within a review article.Citation142 Conversely, the results of the other (recently completed) clinical trials mentioned above have not yet been disseminated.

When this Trial Watch was being redacted (September 2013), official sources listed 20 clinical trials launched after 2012, September 1st to investigate the safety and therapeutic potential of purified/recombinant TAAs or peptide thereof in cancer patients () (source http://www.clinicaltrials.gov). The vast majority of these studies (18 trials) involves recombinant preparations, be them TAA-derived peptides (14 trials), full-length TAAs (1 trial) or TAA-containing fusion proteins (3 trials), most often administered as standalone adjuvanted interventions. In addition, 2 trials intend to investigate the therapeutic profile of recombinant TAAs or tumor cell lysates in complex with chaperones of the heat-shock protein family. Thus, (1) ERBB2-derived peptides are being evaluated, as single immunotherapeutic agents or combined with the ERBB2-targeting antibody trastuzumab,Citation143,Citation144 in subjects with breast carcinoma (NCT01729884; NCT01922921); (2) the safety of and antineoplastic profile of WT1-derived peptides delivered in combination with granulocyte macrophage colony-stimulating factor (GM-CSF) are being assessed in cohorts of multiple myeloma and malignant pleural mesothelioma patients (NCT01827137; NCT01890980); (3) peptides derived from mucin 1 (MUC1), which is overexpressed as an aberrantly glycosylated glycoprotein by multiple tumors of epithelial derivation,Citation145,Citation146 are being tested as standalone adjuvanted interventions or as a liposomal formulation combined with cyclophosphamideCitation147 in NSCLC patients (NCT01720836; NCT01731587); (4) the safety and efficacy of TERT-derived peptides combined with local GM-CSF are being tested in cohorts of NSCLC and prostate carcinoma patients (NCT01789099; NCT01784913); (5) the clinical profile or recombinant NY-ESO-1 or overlapping peptides thereof in combination with cytotoxic T lymphocyte-associated protein 4 (CTLA4)-targeting antibodies is being evaluated in individuals with unresectable or metastatic melanoma (NCT01810016); (6) peptides derived from MAGEA3Citation148 and NA17-A2Citation149 are being tested, alone or combined with each other and/or with a galectin-3 inhibitor, in metastatic melanoma patients (NCT01723813); (7) MLANA epitopes are being tested in combination with a peptide corresponding to residues 267–274 of HIV-1 GagCitation150 and the TLR7 agonist resiquimodCitation151,Citation152 in subjects with stage II-IV melanoma who underwent surgical tumor resection (NCT01748747); and (8) the safety and immunogenicity of phosphorylated peptides derived from breast cancer anti-estrogen resistance 3 (BCAR3) or insulin receptor substrate 2 (IRS2), 2 proteins that are frequently upregulated by malignant cells,Citation153,Citation154 are being assessed in a cohort of melanoma patients (NCT01846143).

Table 1. Recent clinical trials testing purified TAAs or peptides thereof as therapeutic interventions in cancer patients.*

In addition, (1) multipeptide preparations encompassing a panel of recombinant TAA-derived or cytomegalovirus-derived epitopes are being tested in glioblastoma and multiple myeloma patients, either in combination with the DNA-damaging agent temozolomide or as a standalone therapeutic intervention, respectively (NCT01854099; NCT01920191; NCT01718899); (2) the safety and therapeutic profile of a gp100-derived peptide in complex with heat shock 105kDa/110kDa protein 1 (HSPH1), administered as a standalone adjuvanted intervention, are being assessed in patients with advanced Stage III-IV melanoma (NCT01744171); and (3) TAA-derived peptides complexed with HSP96 are being tested, in combination with the vascular endothelial growth factor (VEGF)-targeting antibody bevacizumab,Citation155,Citation156 in individuals bearing resectable recurrent glioblastoma (NCT01814813). Finally, (1) the safety, tolerability, and immunogenicity of a fusion protein comprising HPV E7, administered as a standalone adjuvanted intervention, are being investigated in patients with cervical intraepithelial neoplasia (NCT01880411); (2) the clinical profile of a fusion protein comprising WT1 and MAGE-A10, a nuclear protein frequently expressed by lung, skin and urothelial neoplasms,Citation157-Citation159 is being assessed in patients with acute myeloid leukemia (AML), chronic myeloid leukemia (CML) and myelodysplastic syndrome (MDS) subjected to hematopoietic stem cell transplantation (NCT01819558); and (3) full-length NY-ESO-1 fused to a monoclonal antibody specific for lymphocyte antigen 75 (LY75, a DC receptor best known as DEC-205),Citation160,Citation161given in combination with the demethylating agent decitabine,Citation162,Citation163 is being tested in AML and MDS patients (NCT01834248).

Taken together, these observations suggest that the interest of clinicians in the possibility of employing purified/recombinant TAAs or peptides thereof as therapeutic anticancer interventions remains high.

Concluding Remarks

During the past decade, the possibility of employing purified/recombinant TAAs or peptides thereof as a means to elicit therapeutically relevant tumor-specific immune responses has been intensively investigated, in both preclinical and clinical settings.Citation6 Nonetheless, no peptide-based preparation is currently approved by the US FDA or other international regulatory agencies for use as a therapeutic intervention in cancer patients. Indeed, Cervarix® and Gardasil®, both of which have been licensed by the US FDA as soon as in 2009, are to be employed as preventive, as opposed to therapeutic, measures, de facto constituting conventional antiviral vaccines.Citation22-Citation24

As mentioned above, multiple biological factors contribute to the limited efficacy of anticancer, as opposed to antiviral, vaccines, including the generally low antigenicity of malignant cells and the robust immunosuppressive mechanisms that are established in the course of oncogenesis and tumor progression. In addition, the scarce success of the clinical trials that so far have tested the therapeutic profile of recombinant TAAs or TAA-derived peptides at least in part reflects: (1) the limited availability of novel, clinical grade adjuvants that operate via specific TLRs;Citation7,Citation8,Citation164 (2) the lack of immunological biomarkers that would reliably predict the propensity of specific patients to mount a robust immune response upon vaccination;Citation105,Citation106 and (3) the fact that often, if not always, the clinical evaluation of innovative (immunotherapeutic) regimens is initially performed on subjects bearing very advanced malignancies. As a matter of fact, several TAA-derived peptides and/or full-length TAAs have been shown to exhibit (at least some degree of) clinical activity when administered to patients with minimal residual disease, yet fail to provide any benefit to individuals affected by advanced and/or metastatic tumors.Citation31,Citation165-Citation168

Peptide-based anticancer vaccines hold great promise, but their potential has not been fully developed yet. The characterization of novel tumor-rejection antigens, i.e., TAAs that can elicit an immune response leading to tumor eradication,Citation169,Citation170 an improved understanding of the molecular and cellular circuitries that regulate the mutual interaction between neoplastic cells and the immune system, the development of potent clinical grade adjuvants, the design of combinatorial immunotherapeutic regimens involving peptide-based anticancer vaccines and strategies that limit tumor-associated immunosuppressionCitation171,Citation172 or stimulate the immunogenic demise of cancer cells,Citation76,Citation77 as well as the initiation of clinical trials that comprise meticulous immunomonitoring programs, stand out as the main avenues through which a promising therapeutic paradigm may soon be converted into a robust clinical reality.

Abbreviations:
AML=

acute myeloid leukemia

APC=

antigen-presenting cell

CTL=

cytotoxic T lymphocyte

DC=

dendritic cell

DTH=

delayed type hypersensitivity

ERBB2=

v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2

GM-CSF=

granulocyte macrophage colony-stimulating factor

HPV=

human papillomavirus

IFA=

incomplete Freund’s adjuvant

IFN=

interferon

MAGEA3=

melanoma antigen, family A, 3

MDS=

myelodysplastic syndrome

MLANA=

melan-A

NSCLC=

non-small cell lung carcinoma

TAA=

tumor-associated antigen

TERT=

telomerase reverse transcriptase

TLR=

Toll-like receptor

VEGFR=

vascular endothelial growth factor receptor

WT1=

Wilms tumor 1

Acknowledgments

Authors are supported by the European Commission (ArtForce); European Research Council (ERC); Agence National de la Recherche (ANR); Ligue Nationale contre le Cancer; Fondation pour la Recherche Médicale (FRM); Institut National du Cancer (INCa); Association pour la Recherche sur le Cancer (ARC), LabEx Immuno-Oncologie; Fondation de France; Fondation Bettencourt-Schueller; AXA Chair for Longevity Research; Cancéropôle Ile-de-France, Paris Alliance of Cancer Research Institutes (PACRI) and Cancer Research for Personalized Medicine (CARPEM).

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Citation: Aranda F, Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Peptide vaccines in cancer therapy. OncoImmunology 2013; 2:e26621; 10.4161/onci.26621

References

  • Waldmann TA. Immunotherapy: past, present and future. Nat Med 2003; 9:269 - 77; http://dx.doi.org/10.1038/nm0303-269; PMID: 12612576
  • Riedel S. Edward Jenner and the history of smallpox and vaccination. [Bayl Univ Med Cent] 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
  • Smith KA. Louis pasteur, the father of immunology?. Front Immunol 2012; 3:68; http://dx.doi.org/10.3389/fimmu.2012.00068; PMID: 22566949
  • 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
  • Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2012; 1:1557 - 76; http://dx.doi.org/10.4161/onci.22428; PMID: 23264902
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. 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, Tartour E, Zitvogel L, Kroemer G. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:894 - 907; http://dx.doi.org/10.4161/onci.20931; PMID: 23162757
  • Vacchelli E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists for cancer therapy. Oncoimmunology 2013; 2:e25238; http://dx.doi.org/10.4161/onci.25238; PMID: 24083080
  • 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
  • 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
  • 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
  • van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, Knuth A, Boon T. 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
  • Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2012; 1:1111 - 34; http://dx.doi.org/10.4161/onci.21494; PMID: 23170259
  • Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, et al. Trial Watch: Dendritic cell-based interventions for cancer therapy. OncoImmunology 2013; 2:e25771;; http://dx.doi.org/10.4161/onci.25771; PMID: 23170259
  • Senovilla L, Vacchelli E, Garcia P, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: DNA vaccines for cancer therapy. Oncoimmunology 2013; 2:e23803; http://dx.doi.org/10.4161/onci.23803; PMID: 23734328
  • Bonifaz LC, Bonnyay DP, Charalambous A, Darguste DI, Fujii S, Soares H, Brimnes MK, Moltedo B, Moran TM, Steinman RM. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J Exp Med 2004; 199:815 - 24; http://dx.doi.org/10.1084/jem.20032220; PMID: 15024047
  • Kreutz M, Tacken PJ, Figdor CG. Targeting dendritic cells--why bother?. Blood 2013; 121:2836 - 44; http://dx.doi.org/10.1182/blood-2012-09-452078; PMID: 23390195
  • Tacken PJ, de Vries IJ, Torensma R, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol 2007; 7:790 - 802; http://dx.doi.org/10.1038/nri2173; PMID: 17853902
  • 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
  • Paavonen J, Naud P, Salmerón J, Wheeler CM, Chow SN, Apter D, Kitchener H, Castellsague X, Teixeira JC, Skinner SR, 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
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, 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
  • Galluzzi L. New immunotherapeutic paradigms for castration-resistant prostate cancer. OncoImmunology 2013; 2:e26084; http://dx.doi.org/10.4161/onci.26084
  • Bergman PJ, Camps-Palau MA, McKnight JA, Leibman NF, Craft DM, Leung C, Liao J, Riviere I, Sadelain M, Hohenhaus AE, et al. Development of a xenogeneic DNA vaccine program for canine malignant melanoma at the Animal Medical Center. Vaccine 2006; 24:4582 - 5; http://dx.doi.org/10.1016/j.vaccine.2005.08.027; PMID: 16188351
  • Bergman PJ, McKnight J, Novosad A, Charney S, Farrelly J, Craft D, Wulderk M, Jeffers Y, Sadelain M, Hohenhaus AE, et al. Long-term survival of dogs with advanced malignant melanoma after DNA vaccination with xenogeneic human tyrosinase: a phase I trial. Clin Cancer Res 2003; 9:1284 - 90; PMID: 12684396
  • Liao JC, Gregor P, Wolchok JD, Orlandi F, Craft D, Leung C, Houghton AN, Bergman PJ. Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma. Cancer Immun 2006; 6:8; PMID: 16626110
  • USDA licenses DNA vaccine for treatment of melanoma in dogs. J Am Vet Med Assoc 2010; 236:495; PMID: 20187810
  • 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
  • Nizard M, Sandoval F, Badoual C, Pere H, Terme M, Hans S, Benhamouda N, Granier C, Brasnu D, Tartour E. Immunotherapy of HPV-associated head and neck cancer: Critical parameters. Oncoimmunology 2013; 2:e24534; http://dx.doi.org/10.4161/onci.24534; PMID: 23894716
  • Sandoval F, Terme M, Nizard M, Badoual C, Bureau MF, Freyburger L, Clement O, Marcheteau E, Gey A, Fraisse G, et al. Mucosal imprinting of vaccine-induced CD8⁺ T cells is crucial to inhibit the growth of mucosal tumors. Sci Transl Med 2013; 5:172ra20; http://dx.doi.org/10.1126/scitranslmed.3004888; PMID: 23408053
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Adoptive cell transfer for anticancer immunotherapy. Oncoimmunology 2013; 2:e24238; http://dx.doi.org/10.4161/onci.24238; PMID: 23762803
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012; 1:306 - 15; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautes-Fridman C, Galon J, et al. Trial Watch: Anticancer radioimmunotherapy. OncoImmunology 2013; 2:e24238; http://dx.doi.org/10.4161/onci.24238
  • Menger L, Vacchelli E, Kepp O, Eggermont A, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Cardiac glycosides and cancer therapy. Oncoimmunology 2013; 2:e23082; http://dx.doi.org/10.4161/onci.23082; PMID: 23525565
  • Vacchelli E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Oncolytic viruses for cancer therapy. Oncoimmunology 2013; 2:e24612; http://dx.doi.org/10.4161/onci.24612; PMID: 23894720
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2012; 1:1323 - 43; http://dx.doi.org/10.4161/onci.22009; PMID: 23243596
  • Crane CA, Han SJ, Ahn B, Oehlke J, Kivett V, Fedoroff A, Butowski N, Chang SM, Clarke J, Berger MS, et al. Individual patient-specific immunity against high-grade glioma after vaccination with autologous tumor derived peptides bound to the 96 KD chaperone protein. Clin Cancer Res 2013; 19:205 - 14; http://dx.doi.org/10.1158/1078-0432.CCR-11-3358; PMID: 22872572
  • Salerno EP, Shea SM, Olson WC, Petroni GR, Smolkin ME, McSkimming C, Chianese-Bullock KA, Slingluff CL Jr.. Activation, dysfunction and retention of T cells in vaccine sites after injection of incomplete Freund’s adjuvant, with or without peptide. Cancer Immunol Immunother 2013; 62:1149 - 59; http://dx.doi.org/10.1007/s00262-013-1435-5; PMID: 23657629
  • Zeestraten EC, Speetjens FM, Welters MJ, Saadatmand S, Stynenbosch LF, Jongen R, Kapiteijn E, Gelderblom H, Nijman HW, Valentijn AR, et al. Addition of interferon-α to the p53-SLP® vaccine results in increased production of interferon-γ in vaccinated colorectal cancer patients: a phase I/II clinical trial. Int J Cancer 2013; 132:1581 - 91; http://dx.doi.org/10.1002/ijc.27819; PMID: 22948952
  • Tada Y, Yoshikawa T, Shimomura M, Sawada Y, Sakai M, Shirakawa H, Nobuoka D, Nakatsura T. Analysis of cytotoxic T lymphocytes from a patient with hepatocellular carcinoma who showed a clinical response to vaccination with a glypican‑3‑derived peptide. Int J Oncol 2013; 43:1019 - 26; PMID: 23903757
  • Okamoto I, Arao T, Miyazaki M, Satoh T, Okamoto K, Tsunoda T, Nishio K, Nakagawa K. Clinical phase I study of elpamotide, a peptide vaccine for vascular endothelial growth factor receptor 2, in patients with advanced solid tumors. Cancer Sci 2012; 103:2135 - 8; http://dx.doi.org/10.1111/cas.12014; PMID: 22957712
  • Sugiura F, Inoue K, Okuno K, Sukegawa Y. [Clinical study of Peptide-cocktail vaccination with tegafur-uracil/leucovorin for advanced colorectal cancer]. Gan To Kagaku Ryoho 2012; 39:1760 - 2; PMID: 23267878
  • Sonpavde G, Wang M, Peterson LE, Wang HY, Joe T, Mims MP, Kadmon D, Ittmann MM, Wheeler TM, Gee AP, et al. HLA-restricted NY-ESO-1 peptide immunotherapy for metastatic castration resistant prostate cancer. Invest New Drugs 2013; Forthcoming http://dx.doi.org/10.1007/s10637-013-9960-9; PMID: 23609828
  • Kameshima H, Tsuruma T, Kutomi G, Shima H, Iwayama Y, Kimura Y, Imamura M, Torigoe T, Takahashi A, Hirohashi Y, et al. Immunotherapeutic benefit of α-interferon (IFNα) in survivin2B-derived peptide vaccination for advanced pancreatic cancer patients. Cancer Sci 2013; 104:124 - 9; http://dx.doi.org/10.1111/cas.12046; PMID: 23078230
  • Eikawa S, Kakimi K, Isobe M, Kuzushima K, Luescher I, Ohue Y, Ikeuchi K, Uenaka A, Nishikawa H, Udono H, et al. Induction of CD8 T-cell responses restricted to multiple HLA class I alleles in a cancer patient by immunization with a 20-mer NY-ESO-1f (NY-ESO-1 91-110) peptide. Int J Cancer 2013; 132:345 - 54; http://dx.doi.org/10.1002/ijc.27682; PMID: 22729530
  • Fenoglio D, Traverso P, Parodi A, Tomasello L, Negrini S, Kalli F, Battaglia F, Ferrera F, Sciallero S, Murdaca G, et al. A multi-peptide, dual-adjuvant telomerase vaccine (GX301) is highly immunogenic in patients with prostate and renal cancer. Cancer Immunol Immunother 2013; 62:1041 - 52; http://dx.doi.org/10.1007/s00262-013-1415-9; PMID: 23591981
  • Filipazzi P, Pilla L, Mariani L, Patuzzo R, Castelli C, Camisaschi C, Maurichi A, Cova A, Rigamonti G, Giardino F, et al. Limited induction of tumor cross-reactive T cells without a measurable clinical benefit in early melanoma patients vaccinated with human leukocyte antigen class I-modified peptides. Clin Cancer Res 2012; 18:6485 - 96; http://dx.doi.org/10.1158/1078-0432.CCR-12-1516; PMID: 23032742
  • Kono K, Iinuma H, Akutsu Y, Tanaka H, Hayashi N, Uchikado Y, Noguchi T, Fujii H, Okinaka K, Fukushima R, 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
  • Suzuki H, Fukuhara M, Yamaura T, Mutoh S, Okabe N, Yaginuma H, Hasegawa T, Yonechi A, Osugi J, Hoshino M, et al. Multiple therapeutic peptide vaccines consisting of combined novel cancer testis antigens and anti-angiogenic peptides for patients with non-small cell lung cancer. J Transl Med 2013; 11:97; http://dx.doi.org/10.1186/1479-5876-11-97; PMID: 23578144
  • Matsushita N, Aruga A, Inoue Y, Kotera Y, Takeda K, Yamamoto M. Phase I clinical trial of a peptide vaccine combined with tegafur-uracil plus leucovorin for treatment of advanced or recurrent colorectal cancer. Oncol Rep 2013; 29:951 - 9; PMID: 23314271
  • Yoshimura K, Minami T, Nozawa M, Uemura H. Phase I clinical trial of human vascular endothelial growth factor receptor 1 peptide vaccines for patients with metastatic renal cell carcinoma. Br J Cancer 2013; 108:1260 - 6; http://dx.doi.org/10.1038/bjc.2013.90; PMID: 23470466
  • Ishikawa H, Imano M, Shiraishi O, Yasuda A, Peng YF, Shinkai M, Yasuda T, Imamoto H, Shiozaki H. Phase I clinical trial of vaccination with LY6K-derived peptide in patients with advanced gastric cancer. Gastric Cancer 2013; Forthcoming;; http://dx.doi.org/10.1007/s10120-013-0258-6; PMID: 23613128
  • Sawada Y, Yoshikawa T, Nobuoka D, Shirakawa H, Kuronuma T, Motomura Y, Mizuno S, Ishii H, Nakachi K, Konishi M, et al. Phase I trial of a glypican-3-derived peptide vaccine for advanced hepatocellular carcinoma: immunologic evidence and potential for improving overall survival. Clin Cancer Res 2012; 18:3686 - 96; http://dx.doi.org/10.1158/1078-0432.CCR-11-3044; PMID: 22577059
  • Hayashi H, Kurata T, Fujisaka Y, Kawakami H, Tanaka K, Okabe T, Takeda M, Satoh T, Yoshida K, Tsunoda T, et al. Phase I trial of OTS11101, an anti-angiogenic vaccine targeting vascular endothelial growth factor receptor 1 in solid tumor. Cancer Sci 2013; 104:98 - 104; http://dx.doi.org/10.1111/cas.12034; PMID: 23020774
  • Sabbatini P, Tsuji T, Ferran L, Ritter E, Sedrak C, Tuballes K, Jungbluth AA, Ritter G, Aghajanian C, Bell-McGuinn K, et al. Phase I trial of overlapping long peptides from a tumor self-antigen and poly-ICLC shows rapid induction of integrated immune response in ovarian cancer patients. Clin Cancer Res 2012; 18:6497 - 508; http://dx.doi.org/10.1158/1078-0432.CCR-12-2189; PMID: 23032745
  • Takahashi R, Ishibashi Y, Hiraoka K, Matsueda S, Kawano K, Kawahara A, Kage M, Ohshima K, Yamanaka R, Shichijo S, et al. Phase II study of personalized peptide vaccination for refractory bone and soft tissue sarcoma patients. Cancer Sci 2013; Forthcoming; http://dx.doi.org/10.1111/cas.12226; PMID: 23829867
  • Sawada Y, Yoshikawa T, Fujii S, Mitsunaga S, Nobuoka D, Mizuno S, et al. Remarkable tumor lysis in a hepatocellular carcinoma patient immediately following glypican-3-derived peptide vaccination: An autopsy case. Hum Vaccin Immunother 2013; Forthcoming http://dx.doi.org/10.4161/hv.24179; PMID: 23570049
  • Becker JC, Andersen MH, Hofmeister-Müller V, Wobser M, Frey L, Sandig C, Walter S, Singh-Jasuja H, Kämpgen E, Opitz A, et al. Survivin-specific T-cell reactivity correlates with tumor response and patient survival: a phase-II peptide vaccination trial in metastatic melanoma. Cancer Immunol Immunother 2012; 61:2091 - 103; http://dx.doi.org/10.1007/s00262-012-1266-9; PMID: 22565484
  • Bloom JE, McNeel DG, Olson BM. Vaccination using peptides spanning the SYT-SSX tumor-specific translocation. Expert Rev Vaccines 2012; 11:1401 - 4; http://dx.doi.org/10.1586/erv.12.122; PMID: 23252384
  • Miyatake T, Ueda Y, Morimoto A, Enomoto T, Nishida S, Shirakata T, Oka Y, Tsuboi A, Oji Y, Hosen N, et al. WT1 peptide immunotherapy for gynecologic malignancies resistant to conventional therapies: a phase II trial. J Cancer Res Clin Oncol 2013; 139:457 - 63; http://dx.doi.org/10.1007/s00432-012-1348-2; PMID: 23160854
  • Miller LH, Saul A, Mahanty S. Revisiting Freund’s incomplete adjuvant for vaccines in the developing world. Trends Parasitol 2005; 21:412 - 4; http://dx.doi.org/10.1016/j.pt.2005.07.005; PMID: 16043410
  • Jensen FC, Savary JR, Diveley JP, Chang JC. Adjuvant activity of incomplete Freund’s adjuvant. Adv Drug Deliv Rev 1998; 32:173 - 86; http://dx.doi.org/10.1016/S0169-409X(98)00009-X; PMID: 10837643
  • Murad YM, Clay TM, Lyerly HK, Morse MA. CPG-7909 (PF-3512676, ProMune): toll-like receptor-9 agonist in cancer therapy. Expert Opin Biol Ther 2007; 7:1257 - 66; http://dx.doi.org/10.1517/14712598.7.8.1257; PMID: 17696823
  • Ashkar AA, Rosenthal KL. Toll-like receptor 9, CpG DNA and innate immunity. Curr Mol Med 2002; 2:545 - 56; http://dx.doi.org/10.2174/1566524023362159; PMID: 12243247
  • Ming Lim C, Stephenson R, Salazar AM, Ferris RL. TLR3 agonists improve the immunostimulatory potential of cetuximab against EGFR(+) head and neck cancer cells. Oncoimmunology 2013; 2:e24677; http://dx.doi.org/10.4161/onci.24677; PMID: 23894722
  • Chew V, Abastado JP. Immunomodulation of the tumor microenvironment by Toll-like receptor-3 (TLR3) ligands. Oncoimmunology 2013; 2:e23493; http://dx.doi.org/10.4161/onci.23493; PMID: 23734310
  • Smits EL, Anguille S, Berneman ZN. Interferon α may be back on track to treat acute myeloid leukemia. Oncoimmunology 2013; 2:e23619; http://dx.doi.org/10.4161/onci.23619; PMID: 23734314
  • Chen LL, Gouw L, Sabripour M, Hwu WJ, Benjamin RS. Combining targeted therapy with immunotherapy (interferon-α): Rational, efficacy in gastrointestinal stromal tumor model and implications in other malignancies. Oncoimmunology 2012; 1:773 - 6; http://dx.doi.org/10.4161/onci.19729; PMID: 22934279
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2013; 2:e24850; http://dx.doi.org/10.4161/onci.24850; PMID: 24073369
  • Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2012; 1:493 - 506; http://dx.doi.org/10.4161/onci.20459; PMID: 22754768
  • 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
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74 - 88; http://dx.doi.org/10.1016/j.immuni.2013.06.014; PMID: 23890065
  • Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Kroemer G. 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
  • Vacchelli E, Senovilla L, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2013; 2:e23510; http://dx.doi.org/10.4161/onci.23510; PMID: 23687621
  • Lai JP, Rosenberg AZ, Miettinen MM, Lee CC. NY-ESO-1 expression in sarcomas: A diagnostic marker and immunotherapy target. Oncoimmunology 2012; 1:1409 - 10; http://dx.doi.org/10.4161/onci.21059; PMID: 23243610
  • Vitale I, Senovilla L, Jemaà M, Michaud M, Galluzzi L, Kepp O, Nanty L, Criollo A, Rello-Varona S, Manic G, et al. Multipolar mitosis of tetraploid cells: inhibition by p53 and dependency on Mos. EMBO J 2010; 29:1272 - 84; http://dx.doi.org/10.1038/emboj.2010.11; PMID: 20186124
  • Matsuda R, Enokida H, Chiyomaru T, Kikkawa N, Sugimoto T, Kawakami K, Tatarano S, Yoshino H, Toki K, Uchida Y, et al. LY6K is a novel molecular target in bladder cancer on basis of integrate genome-wide profiling. Br J Cancer 2011; 104:376 - 86; http://dx.doi.org/10.1038/sj.bjc.6605990; PMID: 21063397
  • Ishikawa N, Takano A, Yasui W, Inai K, Nishimura H, Ito H, Miyagi Y, Nakayama H, Fujita M, Hosokawa M, et al. Cancer-testis antigen lymphocyte antigen 6 complex locus K is a serologic biomarker and a therapeutic target for lung and esophageal carcinomas. Cancer Res 2007; 67:11601 - 11; http://dx.doi.org/10.1158/0008-5472.CAN-07-3243; PMID: 18089789
  • Sitnikova L, Mendese G, Liu Q, Woda BA, Lu D, Dresser K, Mohanty S, Rock KL, Jiang Z. IMP3 predicts aggressive superficial urothelial carcinoma of the bladder. Clin Cancer Res 2008; 14:1701 - 6; http://dx.doi.org/10.1158/1078-0432.CCR-07-2039; PMID: 18347170
  • Jiang Z, Chu PG, Woda BA, Rock KL, Liu Q, Hsieh CC, Li C, Chen W, Duan HO, McDougal S, et al. Analysis of RNA-binding protein IMP3 to predict metastasis and prognosis of renal-cell carcinoma: a retrospective study. Lancet Oncol 2006; 7:556 - 64; http://dx.doi.org/10.1016/S1470-2045(06)70732-X; PMID: 16814207
  • Koo BK, Spit M, Jordens I, Low TY, Stange DE, van de Wetering M, van Es JH, Mohammed S, Heck AJ, Maurice MM, et al. Tumour suppressor RNF43 is a stem-cell E3 ligase that induces endocytosis of Wnt receptors. Nature 2012; 488:665 - 9; http://dx.doi.org/10.1038/nature11308; PMID: 22895187
  • Hao HX, Xie Y, Zhang Y, Charlat O, Oster E, Avello M, Lei H, Mickanin C, Liu D, Ruffner H, et al. ZNRF3 promotes Wnt receptor turnover in an R-spondin-sensitive manner. Nature 2012; 485:195 - 200; http://dx.doi.org/10.1038/nature11019; PMID: 22575959
  • Uchida N, Tsunoda T, Wada S, Furukawa Y, Nakamura Y, Tahara H. Ring finger protein 43 as a new target for cancer immunotherapy. Clin Cancer Res 2004; 10:8577 - 86; http://dx.doi.org/10.1158/1078-0432.CCR-04-0104; PMID: 15623641
  • Aleskandarany MA, Negm OH, Rakha EA, Ahmed MA, Nolan CC, Ball GR, Caldas C, Green AR, Tighe PJ, Ellis IO. TOMM34 expression in early invasive breast cancer: a biomarker associated with poor outcome. Breast Cancer Res Treat 2012; 136:419 - 27; http://dx.doi.org/10.1007/s10549-012-2249-4; PMID: 23053644
  • Shimokawa T, Matsushima S, Tsunoda T, Tahara H, Nakamura Y, Furukawa Y. Identification of TOMM34, which shows elevated expression in the majority of human colon cancers, as a novel drug target. Int J Oncol 2006; 29:381 - 6; PMID: 16820880
  • Komori H, Nakatsura T, Senju S, Yoshitake Y, Motomura Y, Ikuta Y, Fukuma D, Yokomine K, Harao M, Beppu T, et al. Identification of HLA-A2- or HLA-A24-restricted CTL epitopes possibly useful for glypican-3-specific immunotherapy of hepatocellular carcinoma. Clin Cancer Res 2006; 12:2689 - 97; http://dx.doi.org/10.1158/1078-0432.CCR-05-2267; PMID: 16675560
  • Nakatsura T, Komori H, Kubo T, Yoshitake Y, Senju S, Katagiri T, Furukawa Y, Ogawa M, Nakamura Y, Nishimura Y. Mouse homologue of a novel human oncofetal antigen, glypican-3, evokes T-cell-mediated tumor rejection without autoimmune reactions in mice. Clin Cancer Res 2004; 10:8630 - 40; http://dx.doi.org/10.1158/1078-0432.CCR-04-1177; PMID: 15623647
  • Rosenberg SA, Zhai Y, Yang JC, Schwartzentruber DJ, Hwu P, Marincola FM, Topalian SL, Restifo NP, Seipp CA, Einhorn JH, et al. Immunizing patients with metastatic melanoma using recombinant adenoviruses encoding MART-1 or gp100 melanoma antigens. J Natl Cancer Inst 1998; 90:1894 - 900; PMID: 9862627
  • Overwijk WW, Tsung A, Irvine KR, Parkhurst MR, Goletz TJ, Tsung K, Carroll MW, Liu C, Moss B, Rosenberg SA, et al. gp100/pmel 17 is a murine tumor rejection antigen: induction of “self”-reactive, tumoricidal T cells using high-affinity, altered peptide ligand. J Exp Med 1998; 188:277 - 86; http://dx.doi.org/10.1084/jem.188.2.277; PMID: 9670040
  • Zhai Y, Yang JC, Spiess P, Nishimura MI, Overwijk WW, Roberts B, Restifo NP, Rosenberg SA. Cloning and characterization of the genes encoding the murine homologues of the human melanoma antigens MART1 and gp100. J Immunother 1997; 20:15 - 25; http://dx.doi.org/10.1097/00002371-199701000-00002; PMID: 9101410
  • Kawaguchi S, Tsukahara T, Ida K, Kimura S, Murase M, Kano M, Emori M, Nagoya S, Kaya M, Torigoe T, et al. SYT-SSX breakpoint peptide vaccines in patients with synovial sarcoma: a study from the Japanese Musculoskeletal Oncology Group. Cancer Sci 2012; 103:1625 - 30; http://dx.doi.org/10.1111/j.1349-7006.2012.02370.x; PMID: 22726592
  • Kawai A, Woodruff J, Healey JH, Brennan MF, Antonescu CR, Ladanyi M. SYT-SSX gene fusion as a determinant of morphology and prognosis in synovial sarcoma. N Engl J Med 1998; 338:153 - 60; http://dx.doi.org/10.1056/NEJM199801153380303; PMID: 9428816
  • Masuzawa T, Fujiwara Y, Okada K, Nakamura A, Takiguchi S, Nakajima K, Miyata H, Yamasaki M, Kurokawa Y, Osawa R, et al. Phase I/II study of S-1 plus cisplatin combined with peptide vaccines for human vascular endothelial growth factor receptor 1 and 2 in patients with advanced gastric cancer. Int J Oncol 2012; 41:1297 - 304; PMID: 22842485
  • Ishizaki H, Tsunoda T, Wada S, Yamauchi M, Shibuya M, Tahara H. Inhibition of tumor growth with antiangiogenic cancer vaccine using epitope peptides derived from human vascular endothelial growth factor receptor 1. Clin Cancer Res 2006; 12:5841 - 9; http://dx.doi.org/10.1158/1078-0432.CCR-06-0750; PMID: 17020992
  • Andersen MH, Svane IM, Becker JC, Straten PT. The universal character of the tumor-associated antigen survivin. Clin Cancer Res 2007; 13:5991 - 4; http://dx.doi.org/10.1158/1078-0432.CCR-07-0686; PMID: 17947459
  • Schmidt SM, Schag K, Müller MR, Weck MM, Appel S, Kanz L, Grünebach F, Brossart P. 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
  • Pospori C, Xue SA, Holler A, Voisine C, Perro M, King J, Fallah-Arani F, Flutter B, Chakraverty R, Stauss HJ, et al. Specificity for the tumor-associated self-antigen WT1 drives the development of fully functional memory T cells in the absence of vaccination. Blood 2011; 117:6813 - 24; http://dx.doi.org/10.1182/blood-2010-08-304568; PMID: 21447831
  • Koesters R, Linnebacher M, Coy JF, Germann A, Schwitalle Y, Findeisen P, von Knebel Doeberitz M. WT1 is a tumor-associated antigen in colon cancer that can be recognized by in vitro stimulated cytotoxic T cells. Int J Cancer 2004; 109:385 - 92; http://dx.doi.org/10.1002/ijc.11721; PMID: 14961577
  • Inderberg-Suso EM, Trachsel S, Lislerud K, Rasmussen AM, Gaudernack G. Widespread CD4+ T-cell reactivity to novel hTERT epitopes following vaccination of cancer patients with a single hTERT peptide GV1001. Oncoimmunology 2012; 1:670 - 86; http://dx.doi.org/10.4161/onci.20426; PMID: 22934259
  • Vermeij R, Leffers N, van der Burg SH, Melief CJ, Daemen T, Nijman HW. Immunological and clinical effects of vaccines targeting p53-overexpressing malignancies. J Biomed Biotechnol 2011; 2011:702146; http://dx.doi.org/10.1155/2011/702146; PMID: 21541192
  • Galluzzi L, Morselli E, Kepp O, Tajeddine N, Kroemer G. Targeting p53 to mitochondria for cancer therapy. Cell Cycle 2008; 7:1949 - 55; http://dx.doi.org/10.4161/cc.7.13.6222; PMID: 18642442
  • Umansky V, Malyguine A, Shurin M. New perspectives in cancer immunotherapy and immunomonitoring. Future Oncol 2009; 5:941 - 4; http://dx.doi.org/10.2217/fon.09.62; PMID: 19792962
  • van der Burg SH. Therapeutic vaccines in cancer: moving from immunomonitoring to immunoguiding. Expert Rev Vaccines 2008; 7:1 - 5; http://dx.doi.org/10.1586/14760584.7.1.1; PMID: 18251686
  • Hailemichael Y, Dai Z, Jaffarzad N, Ye Y, Medina MA, Huang XF, Dorta-Estremera SM, Greeley NR, Nitti G, Peng W, et al. Persistent antigen at vaccination sites induces tumor-specific CD8⁺ T cell sequestration, dysfunction and deletion. Nat Med 2013; 19:465 - 72; http://dx.doi.org/10.1038/nm.3105; PMID: 23455713
  • Ma Y, Xiang D, Sun J, Ding C, Liu M, Hu X, Li G, Kloecker G, Zhang HG, Yan J. Targeting of antigens to B lymphocytes via CD19 as a means for tumor vaccine development. J Immunol 2013; 190:5588 - 99; http://dx.doi.org/10.4049/jimmunol.1203216; PMID: 23630363
  • Sommerfeldt N, Beckhove P, Ge Y, Schütz F, Choi C, Bucur M, Domschke C, Sohn C, Schneeweis A, Rom J, et al. Heparanase: a new metastasis-associated antigen recognized in breast cancer patients by spontaneously induced memory T lymphocytes. Cancer Res 2006; 66:7716 - 23; http://dx.doi.org/10.1158/0008-5472.CAN-05-2363; PMID: 16885374
  • Zhang J, Yang JM, Wang HJ, Ru GQ, Fan DM. Synthesized multiple antigenic polypeptide vaccine based on B-cell epitopes of human heparanase could elicit a potent antimetastatic effect on human hepatocellular carcinoma in vivo. PLoS One 2013; 8:e52940; http://dx.doi.org/10.1371/journal.pone.0052940; PMID: 23308126
  • Dosset M, Vauchy C, Beziaud L, Adotevi O, Godet Y. Universal tumor-reactive helper peptides from telomerase as new tools for anticancer vaccination. Oncoimmunology 2013; 2:e23430; http://dx.doi.org/10.4161/onci.23430; PMID: 23802085
  • Dosset M, Godet Y, Vauchy C, Beziaud L, Lone YC, Sedlik C, Liard C, Levionnois E, Clerc B, Sandoval F, et al. Universal cancer peptide-based therapeutic vaccine breaks tolerance against telomerase and eradicates established tumor. Clin Cancer Res 2012; 18:6284 - 95; http://dx.doi.org/10.1158/1078-0432.CCR-12-0896; PMID: 23032748
  • Brunsvig PF, Kyte JA, Kersten C, Sundstrøm S, Møller M, Nyakas M, Hansen GL, Gaudernack G, Aamdal S. 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
  • Kotsakis A, Vetsika EK, Christou S, Hatzidaki D, Vardakis N, Aggouraki D, Konsolakis G, Georgoulias V, Christophyllakis Ch, Cordopatis P, et al. Clinical outcome of patients with various advanced cancer types vaccinated with an optimized cryptic human telomerase reverse transcriptase (TERT) peptide: results of an expanded phase II study. Ann Oncol 2012; 23:442 - 9; http://dx.doi.org/10.1093/annonc/mdr396; PMID: 21873272
  • Vetsika EK, Konsolakis G, Aggouraki D, Kotsakis A, Papadimitraki E, Christou S, Menez-Jamet J, Kosmatopoulos K, Georgoulias V, Mavroudis D. Immunological responses in cancer patients after vaccination with the therapeutic telomerase-specific vaccine Vx-001. Cancer Immunol Immunother 2012; 61:157 - 68; http://dx.doi.org/10.1007/s00262-011-1093-4; PMID: 21858533
  • Hunger RE, Kernland Lang K, Markowski CJ, Trachsel S, Møller M, Eriksen JA, Rasmussen AM, Braathen LR, Gaudernack G. Vaccination of patients with cutaneous melanoma with telomerase-specific peptides. Cancer Immunol Immunother 2011; 60:1553 - 64; http://dx.doi.org/10.1007/s00262-011-1061-z; PMID: 21681371
  • 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
  • Schlapbach C, Yerly D, Daubner B, Yawalkar N, Hunger RE. Telomerase-specific GV1001 peptide vaccination fails to induce objective tumor response in patients with cutaneous T cell lymphoma. J Dermatol Sci 2011; 62:75 - 83; http://dx.doi.org/10.1016/j.jdermsci.2011.02.001; PMID: 21377838
  • Suso EM, Dueland S, Rasmussen AM, Vetrhus T, Aamdal S, Kvalheim G, Gaudernack G. hTERT mRNA dendritic cell vaccination: complete response in a pancreatic cancer patient associated with response against several hTERT epitopes. Cancer Immunol Immunother 2011; 60:809 - 18; http://dx.doi.org/10.1007/s00262-011-0991-9; PMID: 21365467
  • Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, Horger MS, Maksimovic O, Stenzl A, Hoerr I, et al. Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients. Mol Ther 2011; 19:990 - 9; http://dx.doi.org/10.1038/mt.2010.289; PMID: 21189474
  • Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, Janofsky S, Chew A, Storek J, Akpek G, et al. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood 2011; 117:788 - 97; http://dx.doi.org/10.1182/blood-2010-08-299396; PMID: 21030558
  • Greten TF, Forner A, Korangy F, N’Kontchou G, Barget N, Ayuso C, Ormandy LA, Manns MP, Beaugrand M, Bruix J. A phase II open label trial evaluating safety and efficacy of a telomerase peptide vaccination in patients with advanced hepatocellular carcinoma. BMC Cancer 2010; 10:209; http://dx.doi.org/10.1186/1471-2407-10-209; PMID: 20478057
  • Trepiakas R, Berntsen A, Hadrup SR, Bjørn J, Geertsen PF, Straten PT, Andersen MH, Pedersen AE, Soleimani A, Lorentzen T, et al. Vaccination with autologous dendritic cells pulsed with multiple tumor antigens for treatment of patients with malignant melanoma: results from a phase I/II trial. Cytotherapy 2010; 12:721 - 34; http://dx.doi.org/10.3109/14653241003774045; PMID: 20429791
  • Soleimani A, Berntsen A, Svane IM, Pedersen AE. Immune responses in patients with metastatic renal cell carcinoma treated with dendritic cells pulsed with tumor lysate. Scand J Immunol 2009; 70:481 - 9; http://dx.doi.org/10.1111/j.1365-3083.2009.02322.x; PMID: 19874553
  • Aloysius MM, Mc Kechnie AJ, Robins RA, Verma C, Eremin JM, Farzaneh F, Habib NA, Bhalla J, Hardwick NR, Satthaporn S, et al. Generation in vivo of peptide-specific cytotoxic T cells and presence of regulatory T cells during vaccination with hTERT (class I and II) peptide-pulsed DCs. J Transl Med 2009; 7:18; http://dx.doi.org/10.1186/1479-5876-7-18; PMID: 19298672
  • Berntsen A, Trepiakas R, Wenandy L, Geertsen PF, thor Straten P, Andersen MH, Pedersen AE, Claesson MH, Lorentzen T, Johansen JS, et al. Therapeutic dendritic cell vaccination of patients with metastatic renal cell carcinoma: a clinical phase 1/2 trial. J Immunother 2008; 31:771 - 80; http://dx.doi.org/10.1097/CJI.0b013e3181833818; PMID: 18779742
  • Bolonaki I, Kotsakis A, Papadimitraki E, Aggouraki D, Konsolakis G, Vagia A, Christophylakis C, Nikoloudi I, Magganas E, Galanis A, et al. Vaccination of patients with advanced non-small-cell lung cancer with an optimized cryptic human telomerase reverse transcriptase peptide. J Clin Oncol 2007; 25:2727 - 34; http://dx.doi.org/10.1200/JCO.2006.10.3465; PMID: 17602077
  • Thorn M, Wang M, Kløverpris H, Schmidt EG, Fomsgaard A, Wenandy L, Berntsen A, Brunak S, Buus S, Claesson MH. Identification of a new hTERT-derived HLA-A*0201 restricted, naturally processed CTL epitope. Cancer Immunol Immunother 2007; 56:1755 - 63; http://dx.doi.org/10.1007/s00262-007-0319-y; PMID: 17464507
  • Bernhardt SL, Gjertsen MK, Trachsel S, Møller M, Eriksen JA, Meo M, Buanes T, Gaudernack G. Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: A dose escalating phase I/II study. Br J Cancer 2006; 95:1474 - 82; http://dx.doi.org/10.1038/sj.bjc.6603437; PMID: 17060934
  • Mavroudis D, Bolonakis I, Cornet S, Myllaki G, Kanellou P, Kotsakis A, Galanis A, Nikoloudi I, Spyropoulou M, Menez J, et al. A phase I study of the optimized cryptic peptide TERT(572y) in patients with advanced malignancies. Oncology 2006; 70:306 - 14; http://dx.doi.org/10.1159/000096252; PMID: 17047402
  • Wierecky J, Müller MR, Wirths S, Halder-Oehler E, Dörfel D, Schmidt SM, Häntschel M, Brugger W, Schröder S, Horger MS, et al. Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Res 2006; 66:5910 - 8; http://dx.doi.org/10.1158/0008-5472.CAN-05-3905; PMID: 16740731
  • Brunsvig PF, Aamdal S, Gjertsen MK, Kvalheim G, Markowski-Grimsrud CJ, Sve I, Dyrhaug M, Trachsel S, Møller M, Eriksen JA, 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
  • Märten A, Sievers E, Albers P, Müller S, Franchy C, von Ruecker A, Strunk H, Schild HH, Schmiedel A, Sommer T, et al. Telomerase-pulsed dendritic cells: preclinical results and outcome of a clinical phase I/II trial in patients with metastatic renal cell carcinoma. Ger Med Sci 2006; 4:Doc02; PMID: 19675693
  • Vonderheide RH, Domchek SM, Schultze JL, George DJ, Hoar KM, Chen DY, Stephans KF, Masutomi K, Loda M, Xia Z, et al. Vaccination of cancer patients against telomerase induces functional antitumor CD8+ T lymphocytes. Clin Cancer Res 2004; 10:828 - 39; http://dx.doi.org/10.1158/1078-0432.CCR-0620-3; PMID: 14871958
  • Middleton GW, Walle JW, Wadsley J, Propper D, Coxon FY, Ross PJ, et al. A phase III randomized trial of chemoimmunotherapy comprising gemcitabine and capecitabine with or without telomerase vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer. J Clin Oncol 2013; 31:abstr LBA4004
  • Kyte JA. Cancer vaccination with telomerase peptide GV1001. Expert Opin Investig Drugs 2009; 18:687 - 94; http://dx.doi.org/10.1517/13543780902897631; PMID: 19388882
  • Kruit WH, Suciu S, Dreno B, Mortier L, Robert C, Chiarion-Sileni V, Maio M, Testori A, Dorval T, Grob JJ, et al. Selection of immunostimulant AS15 for active immunization with MAGE-A3 protein: results of a randomized phase II study of the European Organisation for Research and Treatment of Cancer Melanoma Group in Metastatic Melanoma. J Clin Oncol 2013; 31:2413 - 20; http://dx.doi.org/10.1200/JCO.2012.43.7111; PMID: 23715572
  • Ulloa-Montoya F, Louahed J, Dizier B, Gruselle O, Spiessens B, Lehmann FF, Suciu S, Kruit WH, Eggermont AM, Vansteenkiste J, et al. Predictive gene signature in MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol 2013; 31:2388 - 95; http://dx.doi.org/10.1200/JCO.2012.44.3762; PMID: 23715562
  • Burns WR, Zheng Z, Rosenberg SA, Morgan RA. Lack of specific gamma-retroviral vector long terminal repeat promoter silencing in patients receiving genetically engineered lymphocytes and activation upon lymphocyte restimulation. Blood 2009; 114:2888 - 99; http://dx.doi.org/10.1182/blood-2009-01-199216; PMID: 19589923
  • Morgan RA, Dudley ME, Wunderlich JR, Hughes MS, Yang JC, Sherry RM, Royal RE, Topalian SL, Kammula US, Restifo NP, et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006; 314:126 - 9; http://dx.doi.org/10.1126/science.1129003; PMID: 16946036
  • Hamilton E, Blackwell K, Hobeika AC, Clay TM, Broadwater G, Ren XR, Chen W, Castro H, Lehmann F, Spector N, et al. Phase 1 clinical trial of HER2-specific immunotherapy with concomitant HER2 kinase inhibition [corrected]. [corrected] J Transl Med 2012; 10:28; http://dx.doi.org/10.1186/1479-5876-10-28; PMID: 22325452
  • Andersen MH. The specific targeting of immune regulation: T-cell responses against Indoleamine 2,3-dioxygenase. Cancer Immunol Immunother 2012; 61:1289 - 97; http://dx.doi.org/10.1007/s00262-012-1234-4; PMID: 22388712
  • Mavilio D, Galluzzi L, Lugli E. Novel multifunctional antibody approved for the treatment of breast cancer. Oncoimmunology 2013; 2:e24567; http://dx.doi.org/10.4161/onci.24567; PMID: 23802090
  • Kute T, Stehle JR Jr., Ornelles D, Walker N, Delbono O, Vaughn JP. Understanding key assay parameters that affect measurements of trastuzumab-mediated ADCC against Her2 positive breast cancer cells. Oncoimmunology 2012; 1:810 - 21; http://dx.doi.org/10.4161/onci.20447; PMID: 23162748
  • Van Elssen CH, Frings PW, Bot FJ, Van de Vijver KK, Huls MB, Meek B, Hupperets P, Germeraad WT, Bos GM. Expression of aberrantly glycosylated Mucin-1 in ovarian cancer. Histopathology 2010; 57:597 - 606; http://dx.doi.org/10.1111/j.1365-2559.2010.03667.x; PMID: 20955385
  • Lavrsen K, Madsen CB, Rasch MG, Woetmann A, Ødum N, Mandel U, Clausen H, Pedersen AE, Wandall HH. Aberrantly glycosylated MUC1 is expressed on the surface of breast cancer cells and a target for antibody-dependent cell-mediated cytotoxicity. Glycoconj J 2013; 30:227 - 36; http://dx.doi.org/10.1007/s10719-012-9437-7; PMID: 22878593
  • Kroemer G, Zitvogel L, Galluzzi L. Victories and deceptions in tumor immunology: Stimuvax(®). Oncoimmunology 2013; 2:e23687; http://dx.doi.org/10.4161/onci.23687; PMID: 23483762
  • Schultz ES, Lethé B, Cambiaso CL, Van Snick J, Chaux P, Corthals J, Heirman C, Thielemans K, Boon T, van der Bruggen P. A MAGE-A3 peptide presented by HLA-DP4 is recognized on tumor cells by CD4+ cytolytic T lymphocytes. Cancer Res 2000; 60:6272 - 5; PMID: 11103782
  • Trakatelli M, Toungouz M, Blocklet D, Dodoo Y, Gordower L, Laporte M, Vereecken P, Sales F, Mortier L, Mazouz N, et al. A new dendritic cell vaccine generated with interleukin-3 and interferon-beta induces CD8+ T cell responses against NA17-A2 tumor peptide in melanoma patients. Cancer Immunol Immunother 2006; 55:469 - 74; http://dx.doi.org/10.1007/s00262-005-0056-z; PMID: 16133111
  • Gahéry-Ségard H, Pialoux G, Figueiredo S, Igéa C, Surenaud M, Gaston J, Gras-Masse H, Lévy JP, Guillet JG. Long-term specific immune responses induced in humans by a human immunodeficiency virus type 1 lipopeptide vaccine: characterization of CD8+-T-cell epitopes recognized. J Virol 2003; 77:11220 - 31; http://dx.doi.org/10.1128/JVI.77.20.11220-11231.2003; PMID: 14512570
  • Hackstein H, Knoche A, Nockher A, Poeling J, Kubin T, Jurk M, Vollmer J, Bein G. The TLR7/8 ligand resiquimod targets monocyte-derived dendritic cell differentiation via TLR8 and augments functional dendritic cell generation. Cell Immunol 2011; 271:401 - 12; http://dx.doi.org/10.1016/j.cellimm.2011.08.008; PMID: 21889130
  • Igartua M, Pedraz JL. Topical resiquimod: a promising adjuvant for vaccine development?. Expert Rev Vaccines 2010; 9:23 - 7; http://dx.doi.org/10.1586/erv.09.135; PMID: 20021302
  • Reuveni H, Flashner-Abramson E, Steiner L, Makedonski K, Song R, Shir A, Herlyn M, Bar-Eli M, Levitzki A. Therapeutic destruction of insulin receptor substrates for cancer treatment. Cancer Res 2013; 73:4383 - 94; http://dx.doi.org/10.1158/0008-5472.CAN-12-3385; PMID: 23651636
  • Wilson AL, Schrecengost RS, Guerrero MS, Thomas KS, Bouton AH. Breast cancer antiestrogen resistance 3 (BCAR3) promotes cell motility by regulating actin cytoskeletal and adhesion remodeling in invasive breast cancer cells. PLoS One 2013; 8:e65678; http://dx.doi.org/10.1371/journal.pone.0065678; PMID: 23762409
  • Mansfield AS, Nevala WK, Lieser EA, Leontovich AA, Markovic SN. The immunomodulatory effects of bevacizumab on systemic immunity in patients with metastatic melanoma. Oncoimmunology 2013; 2:e24436; http://dx.doi.org/10.4161/onci.24436; PMID: 23762809
  • Michielsen AJ, Ryan EJ, O’Sullivan JN. Dendritic cell inhibition correlates with survival of colorectal cancer patients on bevacizumab treatment. Oncoimmunology 2012; 1:1445 - 7; http://dx.doi.org/10.4161/onci.21318; PMID: 23243624
  • Gure AO, Chua R, Williamson B, Gonen M, Ferrera CA, Gnjatic S, Ritter G, Simpson AJ, Chen YT, Old LJ, et al. Cancer-testis genes are coordinately expressed and are markers of poor outcome in non-small cell lung cancer. Clin Cancer Res 2005; 11:8055 - 62; http://dx.doi.org/10.1158/1078-0432.CCR-05-1203; PMID: 16299236
  • Lin J, Lin L, Thomas DG, Greenson JK, Giordano TJ, Robinson GS, Barve RA, Weishaar FA, Taylor JM, Orringer MB, et al. Melanoma-associated antigens in esophageal adenocarcinoma: identification of novel MAGE-A10 splice variants. Clin Cancer Res 2004; 10:5708 - 16; http://dx.doi.org/10.1158/1078-0432.CCR-04-0468; PMID: 15355897
  • Schultz-Thater E, Piscuoglio S, Iezzi G, Le Magnen C, Zajac P, Carafa V, Terracciano L, Tornillo L, Spagnoli GC. MAGE-A10 is a nuclear protein frequently expressed in high percentages of tumor cells in lung, skin and urothelial malignancies. Int J Cancer 2011; 129:1137 - 48; http://dx.doi.org/10.1002/ijc.25777; PMID: 21710496
  • Tsuji T, Matsuzaki J, Kelly MP, Ramakrishna V, Vitale L, He LZ, Keler T, Odunsi K, Old LJ, Ritter G, et al. Antibody-targeted NY-ESO-1 to mannose receptor or DEC-205 in vitro elicits dual human CD8+ and CD4+ T cell responses with broad antigen specificity. J Immunol 2011; 186:1218 - 27; http://dx.doi.org/10.4049/jimmunol.1000808; PMID: 21149605
  • Birkholz K, Schwenkert M, Kellner C, Gross S, Fey G, Schuler-Thurner B, Schuler G, Schaft N, Dörrie J. Targeting of DEC-205 on human dendritic cells results in efficient MHC class II-restricted antigen presentation. Blood 2010; 116:2277 - 85; http://dx.doi.org/10.1182/blood-2010-02-268425; PMID: 20566893
  • Lainey E, Wolfromm A, Marie N, Enot D, Scoazec M, Bouteloup C, Leroy C, Micol JB, De Botton S, Galluzzi L, et al. Azacytidine and erlotinib exert synergistic effects against acute myeloid leukemia. Oncogene 2013; 32:4331 - 42; http://dx.doi.org/10.1038/onc.2012.469; PMID: 23085751
  • Thépot S, Lainey E, Cluzeau T, Sébert M, Leroy C, Adès L, Tailler M, Galluzzi L, Baran-Marszak F, Roudot H, et al. Hypomethylating agents reactivate FOXO3A in acute myeloid leukemia. Cell Cycle 2011; 10:2323 - 30; http://dx.doi.org/10.4161/cc.10.14.16399; PMID: 21654193
  • Weeratna RD, Makinen SR, McCluskie MJ, Davis HL. TLR agonists as vaccine adjuvants: comparison of CpG ODN and Resiquimod (R-848). Vaccine 2005; 23:5263 - 70; http://dx.doi.org/10.1016/j.vaccine.2005.06.024; PMID: 16081189
  • Hale DF, Clifton GT, Sears AK, Vreeland TJ, Shumway N, Peoples GE, Mittendorf EA. 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
  • Kenter GG, Welters MJ, Valentijn AR, Lowik MJ, Berends-van der Meer DM, Vloon AP, Drijfhout JW, Wafelman AR, Oostendorp J, Fleuren GJ, 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
  • 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
  • Schuster SJ, Neelapu SS, Gause BL, Janik JE, Muggia FM, Gockerman JP, Winter JN, Flowers CR, Nikcevich DA, Sotomayor EM, 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
  • 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
  • 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
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zucman-Rossi J, Zitvogel L, Kroemer G. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2012; 1:28 - 37; http://dx.doi.org/10.4161/onci.1.1.17938; PMID: 22720209
  • Vacchelli E, Eggermont A, Galon J, Sautès-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2013; 2:e22789; http://dx.doi.org/10.4161/onci.22789; PMID: 23482847