2,862
Views
24
CrossRef citations to date
0
Altmetric
Review

Trial watch: Naked and vectored DNA-based anticancer vaccines

, , , , , , , , , , , , & show all
Article: e1026531 | Received 27 Feb 2015, Accepted 27 Feb 2015, Published online: 21 May 2015

Abstract

One type of anticancer vaccine relies on the administration of DNA constructs encoding one or multiple tumor-associated antigens (TAAs). The ultimate objective of these preparations, which can be naked or vectored by non-pathogenic viruses, bacteria or yeast cells, is to drive the synthesis of TAAs in the context of an immunostimulatory milieu, resulting in the (re-)elicitation of a tumor-targeting immune response. In spite of encouraging preclinical results, the clinical efficacy of DNA-based vaccines employed as standalone immunotherapeutic interventions in cancer patients appears to be limited. Thus, efforts are currently being devoted to the development of combinatorial regimens that allow DNA-based anticancer vaccines to elicit clinically relevant immune responses. Here, we discuss recent advances in the preclinical and clinical development of this therapeutic paradigm.

Introduction

During the last 2 decades, several approaches have been conceived to (re-)activate a therapeutically relevant immune response against malignant cells, including dendritic cell (DC)-, peptide- and DNA-based vaccines.Citation1-8 The goal of all such strategies is to endow host antigen-presenting cells (APCs) with the capacity to prime a robust and specific, cellular immune response against one or several tumor-associated antigens (TAAs).Citation9-12 In the context of DC-based vaccination, this is achieved as circulating monocytes are collected from cancer patients, expanded ex vivo in the presence of a source of TAAs and appropriate maturation stimuli, and evenutally re-infused (to the same individual).Citation13-15 Peptide-based vaccination involves the direct administration to cancer patients of recombinant full-length TAAs or peptide thereof, near to invariably in conjunction with potent immunostimulatory agents commonly referred to as adjuvants.Citation16-21 Finally, DNA-based vaccines consist of circularized DNA constructs encoding one or several TAAs, which are delivered to cancer patients as naked plasmids or within appropriate vectors.Citation7,22-25 Vectored DNA-based vaccines should be conceptually differentiated from oncolytic viruses, be they natural or genetically manipulated, as well as from other forms of viral-based anticancer gene therapy, for at least 2 reasons. First, oncolytic viruses, as well as other vectors for gene therapy, target cancer cells, whereas TAA-coding constructs are taken up and expressed by non-malignant cells including APCs, myocytes and epithelial cells (depending on the specific vaccine and administration route).Citation26-28 Second, while oncolytic virotherapy and the delivery of specific gene products to cancer cells aim at provoking their demise (ideally, but not invariably, accompanied by the elicitation of an immune response), DNA-based vaccines mediate antineoplastic effects solely through the immune system.Citation5,9,29

The delivery of naked constructs via the intramuscular route (along with an innocuous electric impulse that provokes the electroporation of myocytes and tissue-resident APCs) is the form of DNA-based anticancer vaccination currently preferred in clinical settings.Citation30 Indeed, although promising results have been obtained with viral, bacterial and eukaryotic vectors, each of these approaches is associated with obstacles that have not yet been completely overcome.Citation31-35 Viral vectors generally ensure increased transduction rates, yet are susceptible to neutralization by natural antibodies (elicited by packaging proteins), are relatively expensive, are not always compatible with the insertion of a large transgene, and are not completely devoid of risks of insertional mutagenesis.Citation36-39 Along similar lines, the development of prokaryotic and eukaryotic (yeast) vectors is not sufficiently advanced for clinical applications.Citation7,22-24 Nonetheless, both of these vectors stand out as promising alternatives to their viral counterparts for at least 2 reasons. First, they are compatible with oral administration.Citation40-42 Second, they both have been shown to elicit potent mucosal immune responses,Citation43-45 which are superior to intramuscular ones, possibly owing to endogenous immunostimulatory factors that trigger Toll-like receptor (TLR) signaling (such as bacterial lipopolysaccharide).Citation46-50 As an alternative to electroporation, DNA-based vaccines can be delivered via the transdermal route, by gene gun,Citation51,52 jet injection,Citation53,54 and tattooing.Citation55,56 None of these delivery methods, however, appears to be superior to electroporation which has been associated with elevated transfection rates,Citation57-59 a minimal extent of tissue injury that exerts immunostimulatory effects (upon the release of damage-associated molecular patterns, DAMPs),Citation60-62 and no significant toxicities.Citation22,24

The target of DNA-based vaccines obviously determines their efficacy as well as their toxicity, a notion that we discussed in previous Trial Watches dealing with this and other TAA-specific active immunotherapies against cancer.Citation9,63,64 One important obstacle against clinical efficiency is indeed represented by the emergence of so-called antigen-loss tumor variants, i.e., malignant cells that do not express the TAA targeted by the vaccine, and–under the selective pressure imposed by vaccine-elicited immunity–emerge and progressively substitute their TAA-expressing counterparts.Citation65-68 Ultimately, this process is responsible for the formation of neoplastic lesions that are completely insensitive to vaccination. Vaccines simultaneously targeting 2 or more TAAs, or targeting so-called tumor rejection antigens (TRAs), i.e., antigens that are critically required for the survival of malignant cells, may partially circumvent this issue.Citation69-74 However, the number of TRAs that are selectively expressed by neoplastic cells, (i.e., not by their non-transformed counterparts or by other healthy tissues) is limited.Citation69,75-77 In addition, progressive tumors often establish potent immunosuppressive networks that limit the efficacy of DNA-based vaccines and several other forms of immunotherapy.Citation78-84 These immunosuppressive circuitries operate both locally and systemically and generally develop along with natural tumor progression, although the presence of an accrued immune reaction, such as that elicited by TAA-targeting vaccines, is expected to accelerate this process.Citation85,86 The emergence of antigen-loss tumor variants and/or the establishment of local and systemic immunosuppression explains why – in spite of promising preclinical results – most DNA-based anticancer vaccines are not efficient in patients when employed as standalone immunotherapeutic interventions.Citation5,9,25,87

As it stands, no DNA-based vaccine is currently approved for use in cancer patients by the US Food and Drug Administration (FDA) or equivalent agencies worldwide. On the contrary, at least 3 preparations of this type are licensed for use in dogs.Citation88-92 Interestingly, one of the DNA-based vaccines approved for veterinary use relies on a xenogenous TAA, i.e., human tyrosinase.Citation91 This said, it seems unlikely that xenogenous TAAs may always be superior to their endogenous counterparts at eliciting therapeutically relevant immune responses.

Along the lines of our Trial Watch series,Citation93,94 here we discuss recent preclinical, translational and clinical advances in the development of DNA-based vaccines for oncological indications.

Update on the development of DNA-based anticancer vaccines

Completed clinical studies

Since the submission of our latest Trial Watch dealing with this topic (March 2014),Citation5 the results of 4 clinical studies assessing the clinical profile of DNA-based anticancer vaccines have been published in international, peer-reviewed scientific journals (source http://www.ncbi.nlm.nih.gov/pubmed), and preliminary data from 3 additional studies have been presented at the American Society of Clinical Oncology (ASCO) annual meeting (source http://meetinglibrary.asco.org/).

Bilusic and colleagues (National Cancer Institute, National Institutes of Health, Bethesda, MD, USA) ran a Phase I clinical trial to test the safety and efficacy of a heat-killed Saccharomyces cerevisiae strain genetically modified to express carcinoembryonic antigen (CEA), a TAA that is overexpressed by several epithelial cancers,Citation95-97 in adult individuals with metastatic, CEA-expressing carcinomas (NCT00924092).Citation33 Twenty-five patients were enrolled in the study and allocated to receive the vaccine (named GI-6207) s.c. every 2 weeks (we) for 3 months (mo) and then monthly thereafter. Vaccination was well tolerated, the most common toxicities being Grade 1/2 injection-site reactions. Five patients experienced disease stabilization for more than 3 mo (range 3.5-18 mo). Moreover, some subjects exhibited increased amounts of CEA-specific CD8+ T lymphocytes coupled to decreased levels of CD4+CD25+FOXP3+ regulatory T cells (Tregs) after vaccination.Citation33

Butterfield and collaborators (University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA) investigated the clinical profile of a prime-boost vaccination strategy relying on a pVAX1-based plasmid encoding full-length human α-fetoprotein (AFP), an oncofetal antigen frequently re-expressed by hepatocellular carcinoma (HCC),Citation98-101 co-injected i.m. with a granulocyte macrophage colony-stimulating factor (GM-CSF)-coding plasmid (prime), followed by a single intramuscular administration of an AFP-coding adenovirus (boost) (NCT00093548).Citation102 Two patients with AFP+ HCC were vaccinated, and neither of them experienced adverse side effects. Signs of immunization could be documented in both individuals, in spite of pre-existing adenovirus-specific antibodies, including an increased amount of AFP-targeting CD8+ T lymphocytes. Of note, tumors in both patients eventually recurred, one of which was after 18 mo and with an AFP lesion.Citation102

Tiriveedhi and co-workers (Washington University School of Medicine, St. Louis, MO, USA) investigated the safety and efficacy of a pING-based plasmid coding for the breast cancer-associated antigen secretoglobin, family 2A, member 2 (SCGB2A2, best known as mammaglobin A)Citation103-106 administered i.m. (3 times, with at least 21-d interval between vaccinations) to subjects with SCGB2A2+ breast carcinoma (NCT00807781).Citation107 Fourteen individuals out of 52 originally enrolled in the study could be vaccinated, none of whom experienced severe (Grade 3-4) toxicities. Common Grade 1-2 side effects included vaccine site tenderness (in 1 out of 14 vaccinated patients), rash (in 1 out of 14 vaccinated patients) and the precipitation of shingle episodes (in 2 out of 14 vaccinated patients). Eight of these patients expressed HLA-A2, allowing for precise immunological monitoring. In these individuals, vaccination efficiently elicited SCGB2A2-specific CD8+ T lymphocytes that were cytotoxic, in vitro, against SCGB2A2+HLA-A2+ breast carcinoma cells. Moreover, vaccinated subjects experienced a statistically significant improvement in progression-free survival as compared to enrolled patients who could not be vaccinated.Citation107

DiPaola et al. (Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA) tested a multimodal immunotherapeutic strategy relying on the sequential administration of a vaccinia virus co-encoding kallikrein-related peptidase 3 (KLK3, best known as prostate-specific antigen, PSA)Citation108,109 and 3 immunostimulatory molecules (i.e., CD58, CD80, and intercellular adhesion molecule 1, ICAM1),Citation110-112 a fowlpox virus modified with the same transgenesCitation113 (both in combination with recombinant GM-CSF),Citation114-116 and androgen ablation therapy, in prostate carcinoma patients with PSA progression but no visible metastasis (NCT00108732).Citation117 These viral vectors were developed by Bavarian Nordic (Washington, DC, USA) under the names of PROSTVAC®-V/TRICOM and PROSTVAC®-F/TRICOM, respectively.Citation118-122 Forty patients were vaccinated, 25 of which did not experience disease progression prior to androgen ablation therapy. Of 27 patients eligible for this therapeutic option, 20 achieved a complete response at 7-mo follow-up. Of note, no Grade 4 toxicities related to treatment were documented.Citation117

Disis and colleagues (University of Washington, Seattle, WA, USA) tested the safety and immunogenicity of a DNA-based vaccine encoding the intracellular domain of erb-b2 receptor tyrosine kinase 2 (ERBB2, best known as HER2), which is overexpressed by a sizeable fraction of breast carcinomas,Citation123-126 in individuals with HER2+ breast carcinoma (NCT00436254).Citation127 Sixty-six patients with Stage III-IV breast carcinoma in remission were enrolled, 64 of which received 3 intradermal vaccine injections administered in combination with recombinant GM-CSF. Minimal toxicities were documented, and the patients who received an intermediate vaccine dose (100 μg/injection versus 10 and 500 μg/injection) were those exhibiting the highest incidence of immunological responders, the immune responses of longest duration and superior overall survival (OS). Interestingly, high doses were associated with increased DNA persistence at injection sites, but this corresponded to comparatively less robust and shorter immune responses.Citation127 Possibly, this relates to the sequestration of antigen-specific CD8+ T lymphocytes at the vaccination site, which has previously been shown to prevent effective antitumor immune responses in preclinical models.Citation128

Patel and collaborators (University of Nottingham, Nottingham, UK) investigated the clinical profile of a peculiar DNA-based anticancer vaccine encoding cytotoxic and helper T-cell epitopes from 2 melanoma-associated antigens (an immunotherapeutic paradigm developed by Scancell, Notthingam, UK, under the name of SCIB1), administered i.m. by electroporation to melanoma patients (NCT01138410).Citation129 SCIB1 is a so-called “ImmunoBody®,” i.e., a DNA-based vaccine in which TAA-derived epitopes are cloned within the complementarity-determining regions (CDRs) of a human antibody.Citation130 In particular, SCIB1 codes from epitopes derived from premelanosome protein (PMEL, best known as gp100)Citation131-133 and tyrosinase-related protein 2 (TRP2)Citation134,135 within the CDRs of an IgG1 molecule. A total of 28 individuals with Stage III-IV melanoma were vaccinated (3 sequential vaccinations at 3 we intervals, then 2 additional vaccination at 3 and 6 mo), none of whom experienced dose-limiting toxicities. The most common side effect was pain at the injection site. Of 25 evaluable patients, 23 exhibited immunological responses to vaccination, and 1 achieved an objective clinical response. The study has not yet concluded although the OS times so far are encouraging.Citation129

Singh and coworkers (National Cancer Institute, National Institutes of Health, Bethesda, MD, USA) assessed the safety and efficacy of a heat-killed S. cerevisiae strain genetically modified to express brachyury homolog (T) in subjects with advanced neoplasms (NCT01519817).Citation136 T is a TAA implicated in both tumor progression and resistance to chemotherapy.Citation137-141 Twenty-seven patients were enrolled and received escalating doses of the vaccine (named GI-6301) every 2 we for 7 injections and then bimonthly. No Grade 3-4 side effects related to vaccination were observed. Of 21 patients evaluable for immune responses, 7 exhibited T-specific CD8+ or CD4+ T lymphocytes. Moreover, treated patients with metastatic colon carcinoma experienced disease stabilization for 15 mo.

Preclinical and translational advances

Consistent efforts are being devoted to identify approaches that may endow TAA-targeting DNA-based vaccines with the ability to elicit robust and specific (and hence therapeutically relevant) immune responses in patients. This can be achieved by: (1) developing DNA constructs and vectors with improved specificity, transfection efficiency and/or immunostimulatory potential; (2) identifying novel TAAs/TRAs as vaccine targets; and (3) optimizing combinatorial immunochemotherapeutic regimens that significantly boost vaccine-elicited immune responses. All of these approaches have been investigated during the last 13 months with promising results.

The immunogenicity of DNA-based anticancer vaccines has been successfully ameliorated by altering the sequence of TAAs targeted by vaccination in several ways, including random shuffling (which allowed for the generation of one construct with superior efficacy as compared to several other containing the same sequences arranged in a different manner),Citation142 the removal of epitopes that elicit IL-10-producing, immunosuppressive TH2 cells (as demonstrated in a vaccine targeting insulin-like growth factor binding protein 2, IGFBP2),Citation143 and the fusion of the TAA-coding sequence with sequences encoding portions of cytotoxic T lymphocyte protein 4 (CTLA4),Citation144 possibly because of the development of endogenous CTLA4-targeting antibodies (which may boost immune responses similar to their recombinant, FDA-approved counterpart ipilimumab).Citation145-148 In addition, encouraging results have recently been obtained by delivering DNA-based vaccines within novel vectors including (but not limited to): a Semliki Forest virus-derived vector co-encoding GM-CSF, as demonstrated by targeting survivin and chorionic gonadotropin, β polypeptide (CGB) in a murine model of melanoma;Citation149 an integrating plasmid based on the PiggyBac system, as shown by targeting transgenic enhanced green fluorescent protein (eGFP) in mice;Citation150 heat-killed Pichia pastoris, as proved by targeting an human papillomavirus (HPV)-16 antigen in a mouse model of cervical carcinoma prevention;Citation35 attenuated Listeria monocytogenes, as demonstrated by targeting CD24 in a murine model of transplantable HCC;Citation151 living Lactococcus lactis, as shown by targeting HPV-16 E7 in a mouse model of cervical cancer;Citation152 a chitosan-based nanodelivery system, as proved by targeting E7 in mice receiving E7-expressing TC-1 cancer cells;Citation153 and perhaps synthetic “pathogen-like” nanoparticles that preferentially target Langerhans cells, although immunological responses were not assessed in this study.Citation154

In an alternative approach, various molecules that (at least hypothetically) may serve as adjuvants to boost the ability of DNA-based anticancer vaccines to elicit robust immune responses have been identified. These molecules include (but are not limited to): IL-15, as demonstrated by targeting a viral protein in Il15−/- vs. wild-type mice;Citation155 the DAMPsCitation61,156,157 IL-33 and high mobility group nucleosomal binding domain 1 (HMGN1), as shown by targeting the exogenous TAA ovalbumin (OVA) in mice receiving OVA-expressing EG7 cells or HPV-16 E7 in mice administered with TC-1 cells, respectively;Citation158,159 the co-stimulatory receptor CD40, as proved in murine models of anticancer vaccination directed against tumor protein 53 (TP53, best known as p53) and gp100;Citation160 the FDA-approved TLR7/TLR8 mixed agonist imiquimod, as demonstrated by targeting HPV-16 E7 in a murine model of cervical carcinoma;Citation161 and the FDA-approved immunomodulatory drug lenalidomide,Citation162,163 as shown in a model of DNA-based vaccination against murine lymphoma.Citation164

During the last 13 months, several laboratories demonstrated the ability of novel DNA-based anticancer vaccines to elicit therapeutically relevant immune responses in mice. These vaccines target well known TAAs, such as HER2, glypican 3 (GPC3), which is generally overexpressed by HCC cells,Citation165 and tyrosinase (TYR), which is selectively expressed by melanocytes,Citation166 as well as proteins that were not considered as therapeutically targetable TAAs, including sequestosome 1 (SQSTM1), an autophagic adaptor with oncogenic functions,Citation167 SRY (sex determining region Y)-box 2 (SOX2), a transcription factor important for the maintenance of cancer stem cells,Citation168 and CD248, a protein that is not expressed by malignant cells but by the tumor endothelium.Citation169 Finally, recent efforts have been devoted to the characterization of the cellular and immunological mechanisms that may underlie the therapeutic efficacy of DNA-based anticancer vaccination. In this context, it has been shown that a DNA vaccine targeting Wilms tumor 1 (WT1) affords mice with complete protection against an otherwise lethal challenge with WT1-expressing mesothelioma cells as it elicits WT1-specific CD8+ T cells while reducing the amounts of immunosuppressive Tregs and myeloid-derived suppressor cells (MDSCs).Citation170 Moreover, the failure of a CEA-targeting DNA vaccine to exert therapeutic activity against colorectal carcinoma cells has been associated with the ability of the latter to avoid the presentation of CEA to CEA-specific CD8+ T lymphocytes.

Recently initiated clinical trials

Since the submission of our latest Trial Watch dealing with this topic (March 2014),Citation5 only 8 clinical trials have been initiated to test the safety and efficacy of DNA-based anticancer vaccines (source http://clinicaltrials.gov/). Seven of these studies are based on naked DNA constructs near to invariably administered i.m. by electroporation (NCT02139267; NCT02157051; NCT02163057; NCT02172911; NCT02204098; NCT02241369; NCT02348320). In addition, one trial relies on semi-allogenic human fibroblasts (MRC-5 cells) as vectors (NCT02211027). All these studies are Phase I or Phase I/II trials ().

Table 1 Clinical trials recently started to evaluate the therapeutic profile of DNA-based anticancer vaccines*

Naked constructs encoding proteins from HPV strains that are causally associated with the development of head and neck cancer and cervical neoplasmsCitation171-173 are being tested in patients affected by these conditions. In particular, the safety and efficacy of a plasmid encoding the E6-E7 fusion protein from HPV-16 and -18 (developed by Inovio, Plymouth Meeting, PA, US, under the name of VGX-3100 SynCon®) administered i.m. by electroporation together with an interleukin (IL)-12-coding construct (called INO-9012) and optionally combined with external beam radiation therapy, are being assessed in cohorts of head and neck squamous cell carcinoma (HNSCC) or cervical carcinoma patients (NCT02163057; NCT02172911). Another naked plasmid coding for HPV-16/18 E6-E7 (developed by Genexine, Gyeonggi-do, Republic of Korea, under the name of GX-188E) as well as for the immunostimulatory molecule fms-like tyrosine kinase-3 ligand (FLT3LG),Citation174-176 administered i.m. by electroporation, is being tested as a standalone immunotherapeutic intervention in subjects with Grade III cervical intraepithelial neoplasia (CIN) (NCT02139267). Finally, the clinical profile of a naked DNA-based vaccine targeting non-disclosed antigens from HPV-6 (developed by Inovio under the name of INO-3106), administered i.m. by electroporation together with INO-9012, is being assessed in patients with HPV-6-associated aerodigestive malignancies, including HNSCCCitation177,178 (NCT02241369).

The safety and efficacy of a naked DNA construct encoding mammaglobin-A administered i.m. by electroporation, are being investigated in breast carcinoma patients undergoing neoadjuvant endocrine therapy (NCT02204098). A plasmid coding for various patient-specific tumor-derived epitopes is being tested as a single immunotherapeutic regimens in subjects with triple-negative (i.e., estrogen receptor-, progesterone receptor- and HER2-negative) breast carcinoma (NCT02348320). Along similar lines, the clinical profile of a naked DNA plasmid encoding various TAAs, including MDM2 proto-oncogene, E3 ubiquitin protein ligase (MDM2), SOX2, and Y box-binding protein 1 (YBX1),Citation179-182 administered intradermally in combination with recombinant human GM-CSF,Citation183,184 is being assessed in women with HER2-negative Stage III-IV breast carcinoma (NCT02157051). Finally, the safety and therapeutic properties of irradiated MRC-5 cells transfected with genomic DNA isolated from autologous tumors, administered intradermally at 4 distinct anatomical locations (i.e., both arms and thighs), are being investigated in subjects with HNSCC (NCT02211027). Of note, to the best of our knowledge, this is the first attempt to employ human cells as vectors for a DNA-based anticancer vaccine.

As for the clinical trials discussed in our previous Trial Watches dealing with this topic,Citation5,9 the following studies have changed status: NCT01493154, which has been “Terminated”; NCT01598792, whose status is “Unknown;" NCT01671488, now listed as “Recruiting”; NCT00849121, NCT01145508, NCT01266460, NCT01304524, NCT01322490, NCT01322802, NCT01486329 and NCT01972737, which are now “Active, not recruiting”; as well as NCT00859729 and NCT01634503, which have been “Completed” (source http://clinicaltrials.gov/). NCT01493154, a Phase I clinical trials testing a naked DNA construct encoding HPV-16/18 E7 in combination with cyclophosphamide (an immunogenic alkylating agent)Citation185-189 in subjects with head and neck carcinoma, has been terminated owing to financial issues. To the best of our knowledge, the results of NCT00859729, a Phase I/II study testing the clinical profile of a naked DNA construct encoding PSA from Rhesus macaque as a standalone immunotherapeutic intervention against prostate carcinoma,Citation190 and NCT01634503, a Phase I trial investigating the safety profile of GX-188E administrered i.m. by electroporation to women with Grade III CIN, have not been disclosed yet (source http://clinicaltrials.gov/).

Although not yet completed, both NCT00849121 and NCT01145508 have preliminary results. NCT00849121 (a Phase II trial) is assessing the safety and efficacy of a naked DNA construct encoding for acid phosphatase, prostate (ACPP, best known as prostatic acid phosphatase, PAP)Citation191,192 administered i.d. to subjects with prostate carcinoma in combination with recombinant human GM-CSF. So far, 17 subjects have been enrolled in this study, of which only 1 developed Grade 2-4 autoimmune reactions or toxicities related to vaccination. In addition, 9 out 17 individuals exhibited an increase in PAP-specific T-cell frequency of at least 3-fold one year after vaccination, and 7 individuals manifested an increase of PSA-doubling time of at least 2-fold. Finally, 12 out of 17 subjects involved in this study were metastasis-free one year after vaccination (source http://clinicaltrials.gov/). NCT01145508 (a Phase II study) is investigating the therapeutic profile of a vaccination strategy based on the sequential, subcutaneous administration of PROSTVAC®-V/TRICOM and PROSTVAC®-F/TRICOM, alone of combined with conventional chemotherapy, to subjects with metastatic, castration-resistant prostate carcinoma.Citation193-195 So far, 10 individuals have been enrolled in the study, 8 of which effectively started treatment with either chemotherapy only (2 patients) or chemotherapy plus vaccination (6 patients). Only 3 patients completed the entire course of therapy (1 from the chemotherapy arm, 2 from the chemotherapy plus vaccination arm), owing to adverse effects, death or physician's decision. The median OS of the chemotherapy arm could not be calculated because of the insufficient number of patients, while the median OS in the chemotherapy plus vaccination arm was 20.8 mo. Of note, virtually all participants in the study experienced serious adverse effects, including febrile neutropenia, fatigue, fatigue, syncope and thrombolytic events. The limited number of patients enrolled in the study, however, prevents concluding to which extent these toxicities truly relate to vaccination (source http://clinicaltrials.gov/).

Concluding Remarks

Accumulating preclinical and clinical evidence indicates that DNA-based anticancer vaccines are relatively ineffective when employed as standalone immunotherapeutic interventions.Citation5,9 Along with the unprecedented clinical success recently obtained by checkpoint-blocking antibodiesCitation196 and with the development of several other immunostimulatory agents,Citation183,197 this deficit has shifted the attention of both researchers and clinicians toward the development of combinatorial immuno(chemo)therapeutic regimens.Citation8,198,199 As it stands, the clinical interest in DNA-based anticancer vaccines remains relatively low, as witnessed by the reduced number of trials initiated during the last 13 months to test this immunotherapeutic paradigm. Nonetheless, we are positive that combining DNA-based anticancer vaccines with appropriate immunostimulants, be them checkpoint blockers, other immunomodulatory monoclonal antibodies, TLR agonists or cytokines, may pave the way to the development of next-generation combinatorial regimens with improved clinical efficacy.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Funding

Authors are supported by the Ligue contre le Cancer (équipe labelisée); Agence National de la Recherche (ANR); Association pour la recherche sur le cancer (ARC); Cancéropôle Ile-de-France; AXA Chair for Longevity Research; Institut National du Cancer (INCa); Fondation Bettencourt-Schueller; Fondation de France; Fondation pour la Recherche Médicale (FRM); the European Commission (ArtForce); the European Research Council (ERC); the LabEx Immuno-Oncology; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); the SIRIC Cancer Research and Personalized Medicine (CARPEM); and the Paris Alliance of Cancer Research Institutes (PACRI).

References

  • Aranda F, Vacchelli E, Eggermont A, Galon J, Sautes-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: peptide vaccines in cancer therapy. Oncoimmunology 2013; 2:e26621; PMID:24498550; http://dx.doi.org/10.4161/onci.26621
  • Ueno H, Schmitt N, Klechevsky E, Pedroza-Gonzalez A, Matsui T, Zurawski G, Oh S, Fay J, Pascual V, Banchereau J, Palucka K. Harnessing human dendritic cell subsets for medicine. Immunol Rev 2010; 234:199-212; PMID:20193020; http://dx.doi.org/10.1111/j.0105-2896.2009.00884.x
  • Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013; 39:38-48; PMID:23890062; http://dx.doi.org/10.1016/j.immuni.2013.07.004
  • Palucka K, Banchereau J. SnapShot: cancer vaccines. Cell 2014; 157:516-e1; PMID:24725415; http://dx.doi.org/10.1016/j.cell.2014.03.044
  • Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Herve Fridman W, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: DNA vaccines for cancer therapy. Oncoimmunology 2014; 3:e28185; PMID:24800178; http://dx.doi.org/10.4161/onci.28185
  • Bloy N, Pol J, Aranda F, Eggermont A, Cremer I, Fridman WH, et al. Trial watch: dendritic cell-based anticancer therapy. Oncoimmunology 2014; 3: e963424; http://dx.doi.org/10.4161/21624011.2014.963424
  • Liu MA. DNA vaccines: an historical perspective and view to the future. Immunol Rev 2011; 239:62-84; PMID:21198665; http://dx.doi.org/10.1111/j.1600-065X.2010.00980.x
  • Galluzzi L, Vacchelli E, Bravo-San Pedro JM, Buque A, Senovilla L, Baracco EE, Bloy N, Castoldi F, Abastado JP, Agostinis P, et al. Classification of current anticancer immunotherapies. Oncotarget 2014; 5:12472-508; PMID:25537519
  • 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; PMID:23734328; http://dx.doi.org/10.4161/onci.23803
  • Haniffa M, Collin M, Ginhoux F. Identification of human tissue cross-presenting dendritic cells: A new target for cancer vaccines. Oncoimmunology 2013; 2:e23140; PMID:23802067; http://dx.doi.org/10.4161/onci.23140
  • Couzin-Frankel J. Breakthrough of the year 2013. Cancer Immunother Sci 2013; 342:1432-3
  • Pardoll D. Immunotherapy: it takes a village. Science 2014; 344:149; PMID:24723594; http://dx.doi.org/10.1126/science.344.6180.149-a
  • Banchereau J, Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol 2005; 5:296-306; PMID:15803149; http://dx.doi.org/10.1038/nri1592
  • Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12:265-77; PMID:22437871; http://dx.doi.org/10.1038/nrc3258
  • Coosemans A, Vergote I, Van Gool SW. Dendritic cell-based immunotherapy in ovarian cancer. Oncoimmunology 2013; 2:e27059; PMID:24501688; http://dx.doi.org/10.4161/onci.27059
  • Aruga A. Vaccination of biliary tract cancer patients with four peptides derived from cancer-testis antigens. Oncoimmunology 2013; 2:e24882; PMID:24073370; http://dx.doi.org/10.4161/onci.24882
  • Ricupito A, Grioni M, Calcinotto A, Bellone M. Boosting anticancer vaccines: too much of a good thing? Oncoimmunology 2013; 2:e25032; PMID:24073378; http://dx.doi.org/10.4161/onci.25032
  • Hailemichael Y, Overwijk WW. Peptide-based anticancer vaccines: the making and unmaking of a T-cell graveyard. Oncoimmunology 2013; 2:e24743; PMID:24073366; http://dx.doi.org/10.4161/onci.24743
  • Bijker MS, Melief CJ, Offringa R, van der Burg SH. Design and development of synthetic peptide vaccines: past, present and future. Expert Rev Vaccines 2007; 6:591-603; PMID:17669012; http://dx.doi.org/10.1586/14760584.6.4.591
  • Yaddanapudi K, Mitchell RA, Eaton JW. Cancer vaccines: looking to the future. Oncoimmunology 2013; 2:e23403; PMID:23802081; http://dx.doi.org/10.4161/onci.23403
  • Valmori D, Souleimanian NE, Tosello V, Bhardwaj N, Adams S, O'Neill D, Pavlick A, Escalon JB, Cruz CM, Angiulli A, 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; PMID:17517626; http://dx.doi.org/10.1073/pnas.0703395104
  • Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev Cancer 2008; 8:108-20; PMID:18219306; http://dx.doi.org/10.1038/nrc2326
  • Fioretti D, Iurescia S, Fazio VM, Rinaldi M. DNA vaccines: developing new strategies against cancer. J Biomed Biotechnol 2010; 2010:174378; PMID:20368780; http://dx.doi.org/10.1155/2010/174378
  • Stevenson FK, Ottensmeier CH, Rice J. DNA vaccines against cancer come of age. Curr Opin Immunol 2010; 22:264-70; PMID:20172703; http://dx.doi.org/10.1016/j.coi.2010.01.019
  • Teja Colluru V, Johnson LE, Olson BM, McNeel DG. Preclinical and clinical development of DNA vaccines for prostate cancer. Urol Oncol 2013
  • Russell SJ, Peng KW, Bell JC. Oncolytic virotherapy. Nat Biotechnol 2012; 30:658-70; PMID:22781695; http://dx.doi.org/10.1038/nbt.2287
  • Vaha-Koskela MJ, Heikkila JE, Hinkkanen AE. Oncolytic viruses in cancer therapy. Cancer Lett 2007; 254:178-216; PMID:17383089; http://dx.doi.org/10.1016/j.canlet.2007.02.002
  • Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Cremer I, Erbs P, Limacher JM, Preville X, Zitvogel L, et al. Trial watch: oncolytic viruses for cancer therapy. Oncoimmunology 2014; 3:e28694; PMID:25097804; http://dx.doi.org/10.4161/onci.28694
  • Collet G, Grillon C, Nadim M, Kieda C. Trojan horse at cellular level for tumor gene therapies. Gene 2013; 525:208-16; PMID:23542073; http://dx.doi.org/10.1016/j.gene.2013.03.057
  • Khan AS, Broderick KE, Sardesai NY. Clinical development of intramuscular electroporation: providing a "boost" for DNA vaccines. Methods Mol Biol 2014; 1121:279-89; PMID:24510832; http://dx.doi.org/10.1007/978-1-4614-9632-8_25
  • Mohler VL, Heithoff DM, Mahan MJ, Hornitzky MA, Thomson PC, House JK. Development of a novel in-water vaccination protocol for DNA adenine methylase deficient Salmonella enterica serovar Typhimurium vaccine in adult sheep. Vaccine 2012; 30:1481-91; PMID:22214887; http://dx.doi.org/10.1016/j.vaccine.2011.12.079
  • Lewis GK. Live-attenuated Salmonella as a prototype vaccine vector for passenger immunogens in humans: are we there yet? Expert Rev Vaccines 2007; 6:431-40; PMID:17542757; http://dx.doi.org/10.1586/14760584.6.3.431
  • Bilusic M, Heery CR, Arlen PM, Rauckhorst M, Apelian D, Tsang KY, Tucker JA, Jochems C, Schlom J, Gulley JL, et al. Phase I trial of a recombinant yeast-CEA vaccine (GI-6207) in adults with metastatic CEA-expressing carcinoma. Cancer Immunol Immunother 2014; 63:225-34; PMID:24327292; http://dx.doi.org/10.1007/s00262-013-1505-8
  • Bernstein MB, Chakraborty M, Wansley EK, Guo Z, Franzusoff A, Mostbock S, Tucker JA, Jochems C, Schlom J, Gulley JL, et al. Recombinant Saccharomyces cerevisiae (yeast-CEA) as a potent activator of murine dendritic cells. Vaccine 2008; 26:509-21; PMID:18155327; http://dx.doi.org/10.1016/j.vaccine.2007.11.033
  • Bolhassani A, Muller M, Roohvand F, Motevalli F, Agi E, Shokri M, Rad MM, Hosseinzadeh S. Whole recombinant Pichia pastoris expressing HPV16 L1 antigen is superior in inducing protection against tumor growth as compared to killed transgenic Leishmania. Hum Vaccin Immunother 2014; 10:3499-508; PMID:25668661; http://dx.doi.org/10.4161/21645515.2014.979606
  • Larocca C, Schlom J. Viral vector-based therapeutic cancer vaccines. Cancer J 2011; 17:359-71; PMID:21952287; http://dx.doi.org/10.1097/PPO.0b013e3182325e63
  • Cawood R, Hills T, Wong SL, Alamoudi AA, Beadle S, Fisher KD, Seymour LW. Recombinant viral vaccines for cancer. Trends Mol Med 2012; 18:564-74; PMID:22917663; http://dx.doi.org/10.1016/j.molmed.2012.07.007
  • Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, Lim A, Osborne CS, Pawliuk R, Morillon E, et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 2003; 302:415-9; PMID:14564000; http://dx.doi.org/10.1126/science.1088547
  • Majhen D, Calderon H, Chandra N, Fajardo CA, Rajan A, Alemany R, Custers J. Adenovirus-based vaccines for fighting infectious diseases and cancer: progress in the field. Hum Gene Ther 2014; 25(4):301-17; PMID:24580050
  • Pasetti MF, Levine MM, Sztein MB. Animal models paving the way for clinical trials of attenuated Salmonella enterica serovar Typhi live oral vaccines and live vectors. Vaccine 2003; 21:401-18; PMID:12531639; http://dx.doi.org/10.1016/S0264-410X(02)00472-3
  • Niethammer AG, Lubenau H, Mikus G, Knebel P, Hohmann N, Leowardi C, Beckhove P, Akhisaroglu M, Ge Y, Springer M, et al. Double-blind, placebo-controlled first in human study to investigate an oral vaccine aimed to elicit an immune reaction against the VEGF-Receptor 2 in patients with stage IV and locally advanced pancreatic cancer. BMC Cancer 2012; 12:361; PMID:22906006; http://dx.doi.org/10.1186/1471-2407-12-361
  • Lin CW, Lee JY, Tsao YP, Shen CP, Lai HC, Chen SL. Oral vaccination with recombinant Listeria monocytogenes expressing human papillomavirus type 16 E7 can cause tumor growth in mice to regress. Int J Cancer 2002; 102:629-37; PMID:12448006; http://dx.doi.org/10.1002/ijc.10759
  • Hamilton DH, Litzinger MT, Jales A, Huang B, Fernando RI, Hodge JW, Ardiani A, Apelian D, Schlom J, Palena C. Immunological targeting of tumor cells undergoing an epithelial-mesenchymal transition via a recombinant brachyury-yeast vaccine. Oncotarget 2013; 4:1777-90; PMID:24125763
  • Tanaka A, Jensen JD, Prado R, Riemann H, Shellman YG, Norris DA, Chin L, Yee C, Fujita M. Whole recombinant yeast vaccine induces antitumor immunity and improves survival in a genetically engineered mouse model of melanoma. Gene Ther 2011; 18:827-34; PMID:21390072; http://dx.doi.org/10.1038/gt.2011.28
  • Zhang T, Sun L, Xin Y, Ma L, Zhang Y, Wang X, Xu K, Ren C, Zhang C, Chen Z, et al. A vaccine grade of yeast Saccharomyces cerevisiae expressing mammalian myostatin. BMC Biotechnol 2012; 12:97; PMID:23253888; http://dx.doi.org/10.1186/1472-6750-12-97
  • Kugelberg E. Dendritic cells: TLR agonists trigger rapid metabolic changes. Nat Rev Immunol 2014; 14:209; PMID:24662378; http://dx.doi.org/10.1038/nri3652
  • Orr MT, Beebe EA, Hudson TE, Moon JJ, Fox CB, Reed SG, Coler RN. A dual TLR agonist adjuvant enhances the immunogenicity and protective efficacy of the tuberculosis vaccine antigen ID93. PLoS One 2014; 9:e83884; PMID:24404140; http://dx.doi.org/10.1371/journal.pone.0083884
  • Vacchelli E, Eggermont A, Sautes-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists for cancer therapy. Oncoimmunology 2013; 2:e25238; PMID:24083080; http://dx.doi.org/10.4161/onci.25238
  • Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Sautes-Fridman C, Cremer I, Henrik Ter Meulen J, Zitvogel L, Kroemer G, et al. Trial Watch: Toll-like receptor agonists in oncological indications. Oncoimmunology 2014; 3:e29179; PMID:25083332; http://dx.doi.org/10.4161/onci.29179
  • 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; PMID:16081189; http://dx.doi.org/10.1016/j.vaccine.2005.06.024
  • Best SR, Peng S, Juang CM, Hung CF, Hannaman D, Saunders JR, Wu TC, Pai SI. Administration of HPV DNA vaccine via electroporation elicits the strongest CD8+ T cell immune responses compared to intramuscular injection and intradermal gene gun delivery. Vaccine 2009; 27:5450-9; PMID:19622402; http://dx.doi.org/10.1016/j.vaccine.2009.07.005
  • Fuller DH, Loudon P, Schmaljohn C. Preclinical and clinical progress of particle-mediated DNA vaccines for infectious diseases. Methods 2006; 40:86-97; PMID:16997717; http://dx.doi.org/10.1016/j.ymeth.2006.05.022
  • Hallermalm K, Johansson S, Brave A, Ek M, Engstrom G, Boberg A, Gudmundsdotter L, Blomberg P, Mellstedt H, Stout R, et al. Pre-clinical evaluation of a CEA DNA prime/protein boost vaccination strategy against colorectal cancer. Scand J Immunol 2007; 66:43-51; PMID:17587345; http://dx.doi.org/10.1111/j.1365-3083.2007.01945.x
  • Nguyen-Hoai T, Kobelt D, Hohn O, Vu MD, Schlag PM, Dorken B, Norley S, Lipp M, Walther W, Pezzutto A, et al. HER2/neu DNA vaccination by intradermal gene delivery in a mouse tumor model: Gene gun is superior to jet injector in inducing CTL responses and protective immunity. Oncoimmunology 2012; 1:1537-45; PMID:23264900; http://dx.doi.org/10.4161/onci.22563
  • van den Berg JH, Nujien B, Beijnen JH, Vincent A, van Tinteren H, Kluge J, Woerdeman LA, Hennink WE, Storm G, Schumacher TN, et al. Optimization of intradermal vaccination by DNA tattooing in human skin. Hum Gene Ther 2009; 20:181-9; PMID:19301471; http://dx.doi.org/10.1089/hum.2008.073
  • van den Berg JH, Oosterhuis K, Schumacher TN, Haanen JB, Bins AD. Intradermal vaccination by DNA tattooing. Methods Mol Biol 2014; 1143:131-40; PMID:24715286; http://dx.doi.org/10.1007/978-1-4939-0410-5_9
  • Murakami T, Sunada Y. Plasmid DNA gene therapy by electroporation: principles and recent advances. Curr Gene Ther 2011; 11:447-56; PMID:22023474; http://dx.doi.org/10.2174/156652311798192860
  • Buchan S, Gronevik E, Mathiesen I, King CA, Stevenson FK, Rice J. Electroporation as a "prime/boost" strategy for naked DNA vaccination against a tumor antigen. J Immunol 2005; 174:6292-8; PMID:15879128; http://dx.doi.org/10.4049/jimmunol.174.10.6292
  • Dupuis M, Denis-Mize K, Woo C, Goldbeck C, Selby MJ, Chen M, Otten GR, Ulmer JB, Donnelly JJ, Ott G, et al. Distribution of DNA vaccines determines their immunogenicity after intramuscular injection in mice. J Immunol 2000; 165:2850-8; PMID:10946318; http://dx.doi.org/10.4049/jimmunol.165.5.2850
  • Galluzzi L, Kepp O, Kroemer G. Immunogenic cell death in radiation therapy. Oncoimmunology 2013; 2:e26536; PMID:24404424; http://dx.doi.org/10.4161/onci.26536
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013; 31:51-72; PMID:23157435; http://dx.doi.org/10.1146/annurev-immunol-032712-100008
  • Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol 2012; 13:780-8; PMID:23175281; http://dx.doi.org/10.1038/nrm3479
  • Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2012; 1:1111-34; PMID:23170259; http://dx.doi.org/10.4161/onci.21494
  • Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2012; 1:1557-76; PMID:23264902; http://dx.doi.org/10.4161/onci.22428
  • Olson BM, McNeel DG. Antigen loss and tumor-mediated immunosuppression facilitate tumor recurrence. Expert Rev Vaccines 2012; 11:1315-7; PMID:23249231; http://dx.doi.org/10.1586/erv.12.107
  • Monjazeb AM, Zamora AE, Grossenbacher SK, Mirsoian A, Sckisel GD, Murphy WJ. Immunoediting and antigen loss: overcoming the achilles heel of immunotherapy with antigen non-specific therapies. Front Oncol 2013; 3:197; PMID:23898464; http://dx.doi.org/10.3389/fonc.2013.00197
  • Maletzki C, Stier S, Linnebacher M. Microsatellite instability in hematological malignancies: Hypermutation vs. immune control-who is challenging who? Oncoimmunology 2013; 2:e25419; PMID:24167765; http://dx.doi.org/10.4161/onci.25419
  • Spiotto MT, Rowley DA, Schreiber H. Bystander elimination of antigen loss variants in established tumors. Nat Med 2004; 10:294-8; PMID:14981514; http://dx.doi.org/10.1038/nm999
  • Wang Y, Yang J, Li Z, Yang S. Evolution of the strategies for screening and identifying human tumor antigens. Curr Protein Pept Sci 2014; 15:819-27; PMID:25345657; http://dx.doi.org/10.2174/1389203715666141027100331
  • Gilboa E. The makings of a tumor rejection antigen. Immunity 1999; 11:263-70; PMID:10514004; http://dx.doi.org/10.1016/S1074-7613(00)80101-6
  • Wolfers J, Lozier A, Raposo G, Regnault A, Thery C, Masurier C, Flament C, Pouzieux S, Faure F, Tursz T, et al. Tumor-derived exosomes are a source of shared tumor rejection antigens for CTL cross-priming. Nat Med 2001; 7:297-303; PMID:11231627; http://dx.doi.org/10.1038/85438
  • Trajanoski Z, Maccalli C, Mennonna D, Casorati G, Parmiani G, Dellabona P. Somatically mutated tumor antigens in the quest for a more efficacious patient-oriented immunotherapy of cancer. Cancer Immunol Immunother 2014; PMID:25164877
  • Buonaguro L, Petrizzo A, Tornesello ML, Buonaguro FM. Translating tumor antigens into cancer vaccines. Clin Vaccine Immunol 2011; 18:23-34; PMID:21048000; http://dx.doi.org/10.1128/CVI.00286-10
  • Kaluza KM, Kottke T, Diaz RM, Rommelfanger D, Thompson J, Vile R. Adoptive transfer of cytotoxic T lymphocytes targeting two different antigens limits antigen loss and tumor escape. Hum Gene Ther 2012; 23:1054-64; PMID:22734672; http://dx.doi.org/10.1089/hum.2012.030
  • Okuyama R, Aruga A, Hatori T, Takeda K, Yamamoto M. Immunological responses to a multi-peptide vaccine targeting cancer-testis antigens and VEGFRs in advanced pancreatic cancer patients. Oncoimmunology 2013; 2:e27010; PMID:24498547; http://dx.doi.org/10.4161/onci.27010
  • Dao T, Liu C, Scheinberg DA. Approaching untargetable tumor-associated antigens with antibodies. Oncoimmunology 2013; 2:e24678; PMID:24073363; http://dx.doi.org/10.4161/onci.24678
  • Scanlon CS, D'Silva NJ. Personalized medicine for cancer therapy: lessons learned from tumor-associated antigens. Oncoimmunology 2013; 2:e23433; PMID:23734304; http://dx.doi.org/10.4161/onci.23433
  • Senovilla L, Aranda F, Galluzzi L, Kroemer G. Impact of myeloid cells on the efficacy of anticancer chemotherapy. Curr Opin Immunol 2014; 30C:24-31; http://dx.doi.org/10.1016/j.coi.2014.05.009
  • 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; PMID:23243596; http://dx.doi.org/10.4161/onci.22009
  • Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 2009; 9:162-74; PMID:19197294; http://dx.doi.org/10.1038/nri2506
  • Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol 2012; 12:253-68; PMID:22437938; http://dx.doi.org/10.1038/nri3175
  • Gajewski TF. Failure at the effector phase: immune barriers at the level of the melanoma tumor microenvironment. Clin Cancer Res 2007; 13:5256-61; PMID:17875753; http://dx.doi.org/10.1158/1078-0432.CCR-07-0892
  • Fallarino F, Grohmann U, Hwang KW, Orabona C, Vacca C, Bianchi R, Belladonna ML, Fioretti MC, Alegre ML, Puccetti P. Modulation of tryptophan catabolism by regulatory T cells. Nat Immunol 2003; 4:1206-12; PMID:14578884; http://dx.doi.org/10.1038/ni1003
  • Wang RF. Regulatory T cells and innate immune regulation in tumor immunity. Springer Semin Immunopathol 2006; 28:17-23; PMID:16838179; http://dx.doi.org/10.1007/s00281-006-0022-7
  • Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion. Science 2011; 331:1565-70; PMID:21436444; http://dx.doi.org/10.1126/science.1203486
  • Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002; 3:991-8; PMID:12407406; http://dx.doi.org/10.1038/ni1102-991
  • Fioretti D, Iurescia S, Rinaldi M. Recent advances in design of immunogenic and effective naked DNA vaccines against cancer. Recent Pat Anticancer Drug Discov 2014; 9:66-82; PMID:23444943
  • Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, Bowen R, Bunning ML. West Nile virus recombinant DNA vaccine protects mouse and horse from virus challenge and expresses in vitro a noninfectious recombinant antigen that can be used in enzyme-linked immunosorbent assays. J Virol 2001; 75:4040-7; PMID:11287553; http://dx.doi.org/10.1128/JVI.75.9.4040-4047.2001
  • Anderson ED, Mourich DV, Leong JA. Gene expression in rainbow trout (Oncorhynchus mykiss) following intramuscular injection of DNA. Mol Mar Biol Biotechnol 1996; 5:105-13; PMID:8680523
  • Anderson ED, Mourich DV, Fahrenkrug SC, LaPatra S, Shepherd J, Leong JA. Genetic immunization of rainbow trout (Oncorhynchus mykiss) against infectious hematopoietic necrosis virus. Mol Mar Biol Biotechnol 1996; 5:114-22; PMID:8680524
  • 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
  • USDA licenses DNA vaccine for treatment of melanoma in dogs. J Am Vet Med Assoc 2010; 236:495; PMID:20187810; http://dx.doi.org/10.2460/javma.236.5.488
  • Bloy N, Pol J, Manic G, Vitale I, Eggermont A, Galon J, Cremer I, Erbs P, Limacher JM, Preville X, et al. Trial watch: padioimmunotherapy for oncological indications. Oncoimmunology 2014; 3:e28694. in press; PMID:25097804
  • Vacchelli E, Aranda F, Eggermont A, Galon J, Sautes-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: tumor-targeting monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27048; PMID:24605265; http://dx.doi.org/10.4161/onci.27048
  • Compte M, Alvarez-Cienfuegos A, Nunez-Prado N, Sainz-Pastor N, Blanco-Toribio A, Pescador N, Sanz L, Alvarez-Vallina L. Functional comparison of single-chain and two-chain anti-CD3-based bispecific antibodies in gene immunotherapy applications. Oncoimmunology 2014; 3:e28810; PMID:25057445; http://dx.doi.org/10.4161/onci.28810
  • Kass ES, Greiner JW, Kantor JA, Tsang KY, Guadagni F, Chen Z, Clark B, De Pascalis R, Schlom J, Van Waes C. Carcinoembryonic antigen as a target for specific antitumor immunotherapy of head and neck cancer. Cancer Res 2002; 62:5049-57; PMID:12208760
  • Wang D, Rayani S, Marshall JL. Carcinoembryonic antigen as a vaccine target. Expert Rev Vaccines 2008; 7:987-93; PMID:18767948; http://dx.doi.org/10.1586/14760584.7.7.987
  • Mizejewski GJ. Biological role of alpha-fetoprotein in cancer: prospects for anticancer therapy. Expert Rev Anticancer Ther 2002; 2:709-35; PMID:12503217; http://dx.doi.org/10.1586/14737140.2.6.709
  • Zhao L, Mou DC, Leng XS, Peng JR, Wang WX, Huang L, Li S, Zhu JY. Expression of cancer-testis antigens in hepatocellular carcinoma. World J Gastroenterol 2004; 10:2034-8; PMID:15237429
  • Llovet JM, Pena CE, Lathia CD, Shan M, Meinhardt G, Bruix J; SHARP Investigators Study Group. Plasma biomarkers as predictors of outcome in patients with advanced hepatocellular carcinoma. Clin Cancer Res 2012; 18:2290-300; PMID:22374331; http://dx.doi.org/10.1158/1078-0432.CCR-11-2175
  • Farinati F, Marino D, De Giorgio M, Baldan A, Cantarini M, Cursaro C, Rapaccini G, Del Poggio P, Di Nolfo MA, Benvegnù L, et al. Diagnostic and prognostic role of alpha-fetoprotein in hepatocellular carcinoma: both or neither? Am J Gastroenterol 2006; 101:524-32; PMID:16542289; http://dx.doi.org/10.1111/j.1572-0241.2006.00443.x
  • Butterfield LH, Economou JS, Gamblin TC, Geller DA. Alpha fetoprotein DNA prime and adenovirus boost immunization of two hepatocellular cancer patients. J Transl Med 2014; 12:86; PMID:24708667; http://dx.doi.org/10.1186/1479-5876-12-86
  • Tiriveedhi V, Fleming TP, Goedegebuure PS, Naughton M, Ma C, Lockhart C, Gao F, Gillanders WE, Mohanakumar T. Mammaglobin-A cDNA vaccination of breast cancer patients induces antigen-specific cytotoxic CD4+ICOShi T cells. Breast Cancer Res Treat 2013; 138:109-18; PMID:22678162; http://dx.doi.org/10.1007/s10549-012-2110-9
  • Watson MA, Dintzis S, Darrow CM, Voss LE, DiPersio J, Jensen R, Fleming TP. Mammaglobin expression in primary, metastatic, and occult breast cancer. Cancer Res 1999; 59:3028-31; PMID:10397237
  • Watson MA, Fleming TP. Mammaglobin, a mammary-specific member of the uteroglobin gene family, is overexpressed in human breast cancer. Cancer Res 1996; 56:860-5; PMID:8631025
  • Zehentner BK, Carter D. Mammaglobin: a candidate diagnostic marker for breast cancer. Clin Biochem 2004; 37:249-57; PMID:15003725; http://dx.doi.org/10.1016/j.clinbiochem.2003.11.005
  • Tiriveedhi V, Tucker N, Herndon J, Li L, Sturmoski M, Ellis M, MaC, Naughton M, Lockhart AC, Gao F, et al. Safety and preliminary evidence of biologic efficacy of a mammaglobin-a DNA vaccine in patients with stable metastatic breast cancer. Clin Cancer Res 2014; 20:5964-75; PMID:25451106; http://dx.doi.org/10.1158/1078-0432.CCR-14-0059
  • Cunha AC, Weigle B, Kiessling A, Bachmann M, Rieber EP. Tissue-specificity of prostate specific antigens: comparative analysis of transcript levels in prostate and non-prostatic tissues. Cancer Lett 2006; 236:229-38; PMID:16046056; http://dx.doi.org/10.1016/j.canlet.2005.05.021
  • Dale W. Prostate cancer: PSA testing in older men–are we following the guidelines? Nat Rev Urol 2012; 9:357-8; PMID:22641163; http://dx.doi.org/10.1038/nrurol.2012.115
  • Hodge JW, Sabzevari H, Yafal AG, Gritz L, Lorenz MG, Schlom J. A triad of costimulatory molecules synergize to amplify T-cell activation. Cancer Res 1999; 59:5800-7; PMID:10582702
  • Hodge JW, Rad AN, Grosenbach DW, Sabzevari H, Yafal AG, Gritz L, Schlom J. Enhanced activation of T cells by dendritic cells engineered to hyperexpress a triad of costimulatory molecules. J Natl Cancer Inst 2000; 92:1228-39; PMID:10922408; http://dx.doi.org/10.1093/jnci/92.15.1228
  • Hodge JW, Poole DJ, Aarts WM, Gomez Yafal A, Gritz L, Schlom J. Modified vaccinia virus ankara recombinants are as potent as vaccinia recombinants in diversified prime and boost vaccine regimens to elicit therapeutic antitumor responses. Cancer Res 2003; 63:7942-9; PMID:14633725
  • Kim JW, Gulley JL. Poxviral vectors for cancer immunotherapy. Expert Opin Biol Ther 2012; 12:463-78; PMID:22413824; http://dx.doi.org/10.1517/14712598.2012.668516
  • Simmons SJ, Tjoa BA, Rogers M, Elgamal A, Kenny GM, Ragde H, Troychak MJ, Boynton AL, Murphy GP. GM-CSF as a systemic adjuvant in a phase II prostate cancer vaccine trial. Prostate 1999; 39:291-7; PMID:10344219; http://dx.doi.org/10.1002/(SICI)1097-0045(19990601)39:4%3c291::AID-PROS10%3e3.0.CO;2-9
  • Arellano M, Lonial S. Clinical uses of GM-CSF, a critical appraisal and update. Biologics 2008; 2:13-27; PMID:19707424
  • Thorne SH. The role of GM-CSF in enhancing immunotherapy of cancer. Immunotherapy 2013; 5:817-9; PMID:23902549; http://dx.doi.org/10.2217/imt.13.65
  • DiPaola RS, Chen YH, Bubley GJ, Stein MN, Hahn NM, Carducci MA, Lattime EC, Gulley JL, Arlen PM, Butterfield LH, et al. A National Multicenter Phase 2 Study of Prostate-specific Antigen (PSA) Pox Virus Vaccine with Sequential Androgen ablation therapy in patients with PSA progression: ECOG 9802. Eur Urol 2014; pii: S0302-2838(14):01265-2; PMID:25533418
  • Madan RA, Heery CR, Gulley JL. Poxviral-based vaccine elicits immunologic responses in prostate cancer patients. Oncoimmunology 2014; 3:e28611; PMID:25097802; http://dx.doi.org/10.4161/onci.28611
  • DiPaola RS, Plante M, Kaufman H, Petrylak DP, Israeli R, Lattime E, Manson K, Schuetz T. A phase I trial of pox PSA vaccines (PROSTVAC-VF) with B7-1, ICAM-1, and LFA-3 co-stimulatory molecules (TRICOM) in patients with prostate cancer. J Transl Med 2006; 4:1; PMID:16390546; http://dx.doi.org/10.1186/1479-5876-4-1
  • Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand M, Manson K, Panicali DL, Laus R, Schlom J, et al. Overall survival analysis of a phase II randomized controlled trial of a Poxviral-based PSA-targeted immunotherapy in metastatic castration-resistant prostate cancer. J Clin Oncol 2010; 28:1099-105; PMID:20100959; http://dx.doi.org/10.1200/JCO.2009.25.0597
  • May KF, Jr., Gulley JL, Drake CG, Dranoff G, Kantoff PW. Prostate cancer immunotherapy. Clin Cancer Res 2011; 17:5233-8; PMID:21700764; http://dx.doi.org/10.1158/1078-0432.CCR-10-3402
  • Schweizer MT, Drake CG. Immunotherapy for prostate cancer: recent developments and future challenges. Cancer Metastasis Rev 2014; 33(2-3):641-55; PMID:24477411
  • De Giovanni C, Nicoletti G, Quaglino E, Landuzzi L, Palladini A, Ianzano ML, Dall'Ora M, Grosso V, Ranieri D, Laranga R, et al. Vaccines against human HER2 prevent mammary carcinoma in mice transgenic for human HER2. Breast Cancer Res 2014; 16:R10; PMID:24451168; http://dx.doi.org/10.1186/bcr3602
  • Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005; 353:1659-72; PMID:16236737; http://dx.doi.org/10.1056/NEJMoa052306
  • Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Jr., Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005; 353:1673-84; PMID:16236738; http://dx.doi.org/10.1056/NEJMoa052122
  • Spicer J, Harries M, Ellis P. Adjuvant trastuzumab for HER2-positive breast cancer. Lancet 2005; 366:634; PMID:16112298; http://dx.doi.org/10.1016/S0140-6736(05)67134-6
  • Disis ML, Coveler AL, Higgins D, Fintak P, Waisman JR, Reichow J, Slota M, Childs J, Dang Y, Salazar LG. A phase I trial of the safety and immunogenicity of a DNA-based vaccine encoding the HER2/neu (HER2) intracellular domain in subjects with HER2+ breast cancer. ASCO Meeting Abstracts 2014; 32:616
  • 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; PMID:23455713; http://dx.doi.org/10.1038/nm.3105
  • Patel PM, Durrant LG, Ottensmeier C, Mulatero C, Lorigan P, Plummer R, Cunnell M, Metheringham R, Brentville V, Machado L, et al. Phase I trial of ImmunoBody in melanoma patients. ASCO Meeting Abstracts 2014; 32:3061
  • Durrant LG, Pudney VA, Spendlove I. Using monoclonal antibodies to stimulate antitumor cellular immunity. Expert Rev Vaccines 2011; 10:1093-106; PMID:21806402; http://dx.doi.org/10.1586/erv.11.33
  • Abdel-Wahab Z, DeMatos P, Hester D, Dong XD, Seigler HF. Human dendritic cells, pulsed with either melanoma tumor cell lysates or the gp100 peptide(280-288), induce pairs of T-cell cultures with similar phenotype and lytic activity. Cell Immunol 1998; 186:63-74; PMID:9637766; http://dx.doi.org/10.1006/cimm.1998.1298
  • Overwijk WW, Tsung A, Irvine KR, Parkhurst MR, Goletz TJ, Tsung K, Carroll MW, Liu C, Moss B, Rosenberg SA, Restifo NP, 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; PMID:9670040; http://dx.doi.org/10.1084/jem.188.2.277
  • Di Pucchio T, Pilla L, Capone I, Ferrantini M, Montefiore E, Urbani F, Patuzzo R, Pennacchioli E, Santinami M, Cova A, et al. Immunization of stage IV melanoma patients with Melan-A/MART-1 and gp100 peptides plus IFN-alpha results in the activation of specific CD8(+) T cells and monocyte/dendritic cell precursors. Cancer Res 2006; 66:4943-51; PMID:16651452; http://dx.doi.org/10.1158/0008-5472.CAN-05-3396
  • Tuting T, Steitz J, Bruck J, Gambotto A, Steinbrink K, DeLeo AB, Robbins P, Knop J, Enk AH. Dendritic cell-based genetic immunization in mice with a recombinant adenovirus encoding murine TRP2 induces effective anti-melanoma immunity. J Gene Med 1999; 1:400-6; PMID:10753065; http://dx.doi.org/10.1002/(SICI)1521-2254(199911/12)1:6%3c400::AID-JGM68%3e3.0.CO;2-D
  • Mahnke K, Qian Y, Fondel S, Brueck J, Becker C, Enk AH. Targeting of antigens to activated dendritic cells in vivo cures metastatic melanoma in mice. Cancer Res 2005; 65:7007-12; PMID:16061687; http://dx.doi.org/10.1158/0008-5472.CAN-05-0938
  • Singh H, Heery CR, Marte JL, Farsaci B, Madan RA, O'Sullivan Coyne GH, Palena C, Rodell TC, Schlom J, Gulley JL. A phase I study of a yeast-based therapeutic cancer vaccine, GI-6301, targeting brachyury in patients with metastatic carcinoma. ASCO Meeting Abstracts 2014; 32:e14026
  • Sarkar D, Shields B, Davies ML, Muller J, Wakeman JA. Brachyury confers cancer stem cell characteristics on colorectal cancer cells. Int J Cancer 2012; 130:328-37; PMID:21365650; http://dx.doi.org/10.1002/ijc.26029
  • Palena C, Polev DE, Tsang KY, Fernando RI, Litzinger M, Krukovskaya LL, Baranova AV, Kozlov AP, Schlom J. The human T-box mesodermal transcription factor Brachyury is a candidate target for T-cell-mediated cancer immunotherapy. Clin Cancer Res 2007; 13:2471-8; PMID:17438107; http://dx.doi.org/10.1158/1078-0432.CCR-06-2353
  • Pires MM, Aaronson SA. Brachyury: a new player in promoting breast cancer aggressiveness. J Natl Cancer Inst 2014; 106:pii: dju094; PMID:24815862; http://dx.doi.org/10.1093/jnci/dju094
  • Thoma C. Prostate cancer: Brachyury–a biomarker for progression and prognosis? Nat Rev Urol 2014; 11:485; PMID:25069730; http://dx.doi.org/10.1038/nrurol.2014.184
  • Tucker JA, Jochems C, Boyerinas B, Fallon J, Greiner JW, Palena C, Rodell TC, Schlom J, Tsang KY. Identification and characterization of a cytotoxic T-lymphocyte agonist epitope of brachyury, a transcription factor involved in epithelial to mesenchymal transition and metastasis. Cancer Immunol Immunother 2014; 63:1307-17; PMID:25186612; http://dx.doi.org/10.1007/s00262-014-1603-2
  • Almajhdi FN, Senger T, Amer HM, Gissmann L, Ohlschlager P. Design of a highly effective therapeutic HPV16 E6/E7-specific DNA vaccine: optimization by different ways of sequence rearrangements (shuffling). PLoS One 2014; 9:e113461; PMID:25422946; http://dx.doi.org/10.1371/journal.pone.0113461
  • Cecil DL, Holt GE, Park KH, Gad E, Rastetter L, Childs J, Higgins D, DisisML. Elimination of IL-10-inducing T-helper epitopes from an IGFBP-2 vaccine ensures potent antitumor activity. Cancer Res 2014; 74:2710-8; PMID:24778415; http://dx.doi.org/10.1158/0008-5472.CAN-13-3286
  • Gan L, Jia R, Zhou L, Guo J, Fan M. Fusion of CTLA-4 with HPV16 E7 and E6 enhanced the potency of therapeutic HPV DNA vaccine. PLoS One 2014; 9:e108892; PMID:25265018; http://dx.doi.org/10.1371/journal.pone.0108892
  • Aranda F, Vacchelli E, Eggermont A, Galon J, Fridman WH, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies in cancer therapy. Oncoimmunology 2014; 3:e27297; PMID:24701370; http://dx.doi.org/10.4161/onci.27297
  • Vacchelli E, Eggermont A, Galon J, Sautes-Fridman C, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: monoclonal antibodies in cancer therapy. Oncoimmunology 2013; 2:e22789; PMID:23482847; http://dx.doi.org/10.4161/onci.22789
  • Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711-23; PMID:20525992; http://dx.doi.org/10.1056/NEJMoa1003466
  • Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, Garbe C, Lebbe C, Baurain JF, Testori A, Grob JJ, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 2011; 364:2517-26; PMID:21639810; http://dx.doi.org/10.1056/NEJMoa1104621
  • Zhang L, Wang Y, Xiao Y, Wang Y, Dong J, Gao K, Gao Y, Wang X, Zhang W, Xu Y, et al. Enhancement of antitumor immunity using a DNA-based replicon vaccine derived from Semliki Forest virus. PLoS One 2014; 9:e90551; PMID:24608380; http://dx.doi.org/10.1371/journal.pone.0090551
  • Bertino P, Urschitz J, Hoffmann FW, You BR, Rose AH, Park WH, Moisyadi S, Hoffmann PR. Vaccination with a piggyBac plasmid with transgene integration potential leads to sustained antigen expression and CD8(+) T cell responses. Vaccine 2014; 32:1670-7; PMID:24513010; http://dx.doi.org/10.1016/j.vaccine.2014.01.063
  • Yang Y, Hou J, Lin Z, Zhuo H, Chen D, Zhang X, Chen Y, Sun B. Attenuated Listeria monocytogenes as a cancer vaccine vector for the delivery of CD24, a biomarker for hepatic cancer stem cells. Cell Mol Immunol 2014; 11:184-96; PMID:24488178; http://dx.doi.org/10.1038/cmi.2013.64
  • Rangel-Colmenero BR, Gomez-Gutierrez JG, Villatoro-Hernandez J, Zavala-Flores LM, Quistian-Martinez D, Rojas-Martinez A, Arce-Mendoza AY, Guzmán-López S, Montes-de-Oca-Luna R, Saucedo-Cárdenas O. Enhancement of Ad-CRT/E7-mediated antitumor effect by preimmunization with L. lactis expressing HPV-16 E7. Viral Immunol 2014; 27:463-7; PMID:25216057; http://dx.doi.org/10.1089/vim.2014.0055
  • Tahamtan A, Ghaemi A, Gorji A, Kalhor HR, Sajadian A, Tabarraei A, Moradi A, Atyabi F, Kelishadi M. Antitumor effect of therapeutic HPV DNA vaccines with chitosan-based nanodelivery systems. J Biomed Sci 2014; 21:69; PMID:25077570; http://dx.doi.org/10.1186/s12929-014-0069-z
  • Toke ER, Lorincz O, Csiszovszki Z, Somogyi E, Felfoldi G, Molnar L, Szipőcs R, Kolonics A, Malissen B, Lori F, et al. Exploitation of Langerhans cells for in vivo DNA vaccine delivery into the lymph nodes. Gene Ther 2014; 21:566-74; PMID:24694539; http://dx.doi.org/10.1038/gt.2014.29
  • Li J, Valentin A, Ng S, Beach RK, Alicea C, Bergamaschi C, Felber BK, Pavlakis GN. Differential effects of IL-15 on the generation, maintenance and cytotoxic potential of adaptive cellular responses induced by DNA vaccination. Vaccine 2015; 33:1188-96; PMID:25559187; http://dx.doi.org/10.1016/j.vaccine.2014.12.046
  • Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol 2013; 59:583-94; PMID:23567086; http://dx.doi.org/10.1016/j.jhep.2013.03.033
  • Krysko DV, Garg AD, Kaczmarek A, Krysko O, Agostinis P, Vandenabeele P. Immunogenic cell death and DAMPs in cancer therapy. Nat Rev Cancer 2012; 12:860-75; PMID:23151605; http://dx.doi.org/10.1038/nrc3380
  • Wei F, Yang D, Tewary P, Li Y, Li S, Chen X, Howard OM, Bustin M, Oppenheim JJ. The Alarmin HMGN1 contributes to antitumor immunity and is a potent immunoadjuvant. Cancer Res 2014; 74:5989-98; PMID:25205103; http://dx.doi.org/10.1158/0008-5472.CAN-13-2042
  • Villarreal DO, Wise MC, Walters JN, Reuschel EL, Choi MJ, Obeng-Adjei N, Yan J, Morrow MP, Weiner DB. Alarmin IL-33 acts as an immunoadjuvant to enhance antigen-specific tumor immunity. Cancer Res 2014; 74:1789-800; PMID:24448242; http://dx.doi.org/10.1158/0008-5472.CAN-13-2729
  • Soong RS, Song L, Trieu J, Lee SY, He L, Tsai YC, Wu TC, Hung CF. Direct T cell activation via CD40 ligand generates high avidity CD8+ T cells capable of breaking immunological tolerance for the control of tumors. PLoS One 2014; 9:e93162; PMID:24664420; http://dx.doi.org/10.1371/journal.pone.0093162
  • Soong RS, Song L, Trieu J, Knoff J, He L, Tsai YC, Huh W, Chang YN, Cheng WF, Roden RB, et al. Toll-like receptor agonist imiquimod facilitates antigen-specific CD8+ T-cell accumulation in the genital tract leading to tumor control through IFNgamma. Clin Cancer Res 2014; 20:5456-67; PMID:24893628; http://dx.doi.org/10.1158/1078-0432.CCR-14-0344
  • Semeraro M, Galluzzi L. Novel insights into the mechanism of action of lenalidomide. Oncoimmunology 2014; 3:e28386; PMID:25340011; http://dx.doi.org/10.4161/onci.28386
  • Semeraro M, Vacchelli E, Eggermont A, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: lenalidomide-based immunochemotherapy. Oncoimmunology 2013; 2:e26494; PMID:24482747; http://dx.doi.org/10.4161/onci.26494
  • Sakamaki I, Kwak LW, Cha SC, Yi Q, Lerman B, Chen J, Surapaneni S, Bateman S, Qin H. Lenalidomide enhances the protective effect of a therapeutic vaccine and reverses immune suppression in mice bearing established lymphomas. Leukemia 2014; 28:329-37; PMID:23765229; http://dx.doi.org/10.1038/leu.2013.177
  • Li SQ, Lin J, Qi CY, Fu SJ, Xiao WK, Peng BG, Liang LJ. GPC3 DNA vaccine elicits potent cellular antitumor immunity against HCC in mice. Hepatogastroenterology 2014; 61:278-84; PMID:24901124
  • Yan J, Tingey C, Lyde R, Gorham TC, Choo DK, Muthumani A, Myles D, Weiner LP, Kraynyak KA, Reuschel EL, et al. Novel and enhanced anti-melanoma DNA vaccine targeting the tyrosinase protein inhibits myeloid-derived suppressor cells and tumor growth in a syngeneic prophylactic and therapeutic murine model. Cancer Gene Ther 2014; 21:507-17; PMID:25394503; http://dx.doi.org/10.1038/cgt.2014.56
  • Gabai V, Venanzi FM, Bagashova E, Rud O, Mariotti F, Vullo C, Catone G, Sherman MY, Concetti A, Chursov A, et al. Pilot study of p62 DNA vaccine in dogs with mammary tumors. Oncotarget 2014; 5:12803-10; PMID:25296974
  • Polakova I, Duskova M, Smahel M. Antitumor DNA vaccination against the Sox2 transcription factor. Int J Oncol 2014; 45:139-46; PMID:24789529
  • Facciponte JG, Ugel S, De Sanctis F, Li C, Wang L, Nair G, Sehgal S, Raj A, Matthaiou E, Coukos G, et al. Tumor endothelial marker 1-specific DNA vaccination targets tumor vasculature. J Clin Invest 2014; 124:1497-511; PMID:24642465; http://dx.doi.org/10.1172/JCI67382
  • Tan Z, Zhou J, Cheung AK, Yu Z, Cheung KW, Liang J, Wang H, Lee BK, Man K, Liu L, et al. Vaccine-elicited CD8+ T cells cure mesothelioma by overcoming tumor-induced immunosuppressive environment. Cancer Res 2014; 74:6010-21; PMID:25125656; http://dx.doi.org/10.1158/0008-5472.CAN-14-0473
  • Agosti JM, Goldie SJ. Introducing HPV vaccine in developing countries–key challenges and issues. N Engl J Med 2007; 356:1908-10; PMID:17494923; http://dx.doi.org/10.1056/NEJMp078053
  • Paavonen J, Naud P, Salmeron J, Wheeler CM, Chow SN, Apter D, Kitchener H, Castellsague X, Teixeira JC, Skinner SR, et al. 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; PMID:19586656; http://dx.doi.org/10.1016/S0140-6736(09)61248-4
  • FUTURE II Study Group. Quadrivalent vaccine against human papillomavirus to prevent high-grade cervical lesions. N Engl J Med 2007; 356:1915-27; PMID:17494925; http://dx.doi.org/10.1056/NEJMoa061741
  • King GD, Muhammad AK, Larocque D, Kelson KR, Xiong W, Liu C, Sanderson NS, Kroeger KM, Castro MG, Lowenstein PR. Combined Flt3L/TK gene therapy induces immunological surveillance which mediates an immune response against a surrogate brain tumor neoantigen. Mol Ther 2011; 19:1793-801; PMID:21505426; http://dx.doi.org/10.1038/mt.2011.77
  • Guermonprez P, Helft J, Claser C, Deroubaix S, Karanje H, Gazumyan A, Darasse-Jèze G, Telerman SB, Breton G, Schreiber HA, et al. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection. Nat Med 2013; 19:730-8; PMID:23685841; http://dx.doi.org/10.1038/nm.3197
  • Kutzler MA, Weiner DB. Developing DNA vaccines that call to dendritic cells. J Clin Invest 2004; 114:1241-4; PMID:15520855; http://dx.doi.org/10.1172/JCI23467
  • Michaud DS, Langevin SM, Eliot M, Nelson HH, Pawlita M, McClean MD, Kelsey KT. High-risk HPV types and head and neck cancer. Int J Cancer 2014; 135:1653-61; PMID:24615247; http://dx.doi.org/10.1002/ijc.28811
  • Syrjanen S. The role of human papillomavirus infection in head and neck cancers. Ann Oncol 2010; 21 Suppl 7:vii243-5; PMID:20943622
  • Maciejczyk A, Szelachowska J, Ekiert M, Matkowski R, Halon A, Lage H, Surowiak P. Elevated nuclear YB1 expression is associated with poor survival of patients with early breast cancer. Anticancer Res 2012; 32:3177-84; PMID:22843890
  • Woolley AG, Algie M, Samuel W, Harfoot R, Wiles A, Hung NA, Tan PH, Hains P, Valova VA, Huschtscha L, et al. Prognostic association of YB-1 expression in breast cancers: a matter of antibody. PLoS One 2011; 6:e20603; PMID:21695211; http://dx.doi.org/10.1371/journal.pone.0020603
  • Dhodapkar KM, Gettinger SN, Das R, Zebroski H, Dhodapkar MV. SOX2-specific adaptive immunity and response to immunotherapy in non-small cell lung cancer. Oncoimmunology 2013; 2:e25205; PMID:24073380; http://dx.doi.org/10.4161/onci.25205
  • Leis O, Eguiara A, Lopez-Arribillaga E, Alberdi MJ, Hernandez-Garcia S, Elorriaga K, Pandiella A, Rezola R, Martin AG. Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene 2012; 31:1354-65; PMID:21822303; http://dx.doi.org/10.1038/onc.2011.338
  • Vacchelli E, Aranda F, Obrist F, Eggermont A, Galon J, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory cytokines in cancer therapy. Oncoimmunology 2014; 3:e29030; PMID:25083328; http://dx.doi.org/10.4161/onci.29030
  • Vacchelli E, Eggermont A, Fridman WH, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: immunostimulatory cytokines. Oncoimmunology 2013; 2:e24850; PMID:24073369; http://dx.doi.org/10.4161/onci.24850
  • Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, Bracci L, et al. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3: e955691; http://dx.doi.org/10.4161/21624011.2014.955691
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215-33; PMID:22301798; http://dx.doi.org/10.1038/nrd3626
  • Zitvogel L, Galluzzi L, Smyth MJ, Kroemer G. Mechanism of action of conventional and targeted anticancer therapies: reinstating immunosurveillance. Immunity 2013; 39:74-88; PMID:23890065; http://dx.doi.org/10.1016/j.immuni.2013.06.014
  • Schiavoni G, Sistigu A, Valentini M, Mattei F, Sestili P, Spadaro F, Sanchez M, Lorenzi S, D'Urso MT, Belardelli F, et al. Cyclophosphamide synergizes with type I interferons through systemic dendritic cell reactivation and induction of immunogenic tumor apoptosis. Cancer Res 2011; 71:768-78; PMID:21156650; http://dx.doi.org/10.1158/0008-5472.CAN-10-2788
  • Young BA, Spencer JF, Ying B, Tollefson AE, Toth K, Wold WS. The role of cyclophosphamide in enhancing antitumor efficacy of an adenovirus oncolytic vector in subcutaneous Syrian hamster tumors. Cancer Gene Ther 2013; 20:521-30; PMID:23928731; http://dx.doi.org/10.1038/cgt.2013.49
  • Eriksson F, Totterman T, Maltais AK, Pisa P, Yachnin J. DNA vaccine coding for the rhesus prostate specific antigen delivered by intradermal electroporation in patients with relapsed prostate cancer. Vaccine 2013; 31:3843-8; PMID:23831327; http://dx.doi.org/10.1016/j.vaccine.2013.06.063
  • Becker JT, Olson BM, Johnson LE, Davies JG, Dunphy EJ, McNeel DG. DNA vaccine encoding prostatic acid phosphatase (PAP) elicits long-term T-cell responses in patients with recurrent prostate cancer. J Immunother 2010; 33:639-47; PMID:20551832; http://dx.doi.org/10.1097/CJI.0b013e3181dda23e
  • Lin AM, Hershberg RM, Small EJ. Immunotherapy for prostate cancer using prostatic acid phosphatase loaded antigen presenting cells. Urol Oncol 2006; 24:434-41; PMID:16962496; http://dx.doi.org/10.1016/j.urolonc.2005.08.010
  • Massari F, Maines F, Modena A, Brunelli M, Bria E, Artibani W, Martignoni G, Tortora G. Castration resistant prostate cancer (CRPC): state of the art, perspectives and new challenges. Anticancer Agents Med Chem 2013; 13:872-86; PMID:23272970; http://dx.doi.org/10.2174/18715206113139990077
  • Galluzzi L. New immunotherapeutic paradigms for castration-resistant prostate cancer. Oncoimmunology 2013; 2:e26084; http://dx.doi.org/10.4161/onci.26084
  • Lubaroff DM. Prostate cancer vaccines in clinical trials. Expert Rev Vaccines 2012; 11:857-68; PMID:22913261; http://dx.doi.org/10.1586/erv.12.54
  • Galluzzi L, Kroemer G, Eggermont A. Novel immune checkpoint blocker approved for the treatment of advanced melanoma. Oncoimmunology 2014; 3:e967147; http://dx.doi.org/10.4161/21624011.2014.967147
  • Vacchelli E, Aranda F, Eggermont A, Sautès-Fridman C, Tartour E, Kennedy EP, Platten M, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2014; 3: e957994; http://dx.doi.org/10.4161/21624011.2014.957994
  • Vacchelli E, Prada N, Kepp O, Galluzzi L. Current trends of anticancer immunochemotherapy. Oncoimmunology 2013; 2:e25396; PMID:23894726; http://dx.doi.org/10.4161/onci.25396
  • Riedmann EM. News studies: combining PROSTVAC with conventional cancer therapy. Hum Vaccin Immunother 2012; 8:848-9; http://dx.doi.org/10.4161/hv.22583

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.