1,501
Views
35
CrossRef citations to date
0
Altmetric
Review

Trial Watch

DNA vaccines for cancer therapy

, , , , , , , , & show all
Article: e28185 | Received 09 Feb 2014, Accepted 10 Feb 2014, Published online: 01 Apr 2014

Abstract

During the past 2 decades, the possibility that preparations capable of eliciting tumor-specific immune responses would mediate robust therapeutic effects in cancer patients has received renovated interest. In this context, several approaches to vaccinate cancer patients against their own malignancies have been conceived, including the administration of DNA constructs coding for one or more tumor-associated antigens (TAAs). Such DNA-based vaccines conceptually differ from other types of gene therapy in that they are not devised to directly kill cancer cells or sensitize them to the cytotoxic activity of a drug, but rather to elicit a tumor-specific immune response. In spite of an intense wave of preclinical development, the introduction of this immunotherapeutic paradigm into the clinical practice is facing difficulties. Indeed, while most DNA-based anticancer vaccines are well tolerated by cancer patients, they often fail to generate therapeutically relevant clinical responses. In this Trial Watch, we discuss the latest advances on the use of DNA-based vaccines in cancer therapy, discussing the literature that has been produced around this topic during the last 13 months as well as clinical studies that have been launched in the same time frame to assess the actual therapeutic potential of this intervention.

Introduction

The possibility to employ vaccination as a therapeutic means against cancer was first proposed at the end of the 19th century by the German physician Paul Ehrlich (1854–1915) and the American surgeon William Bradley Coley (1862–1936),Citation1 but the interest in this approach quickly declined thereafter.Citation2 It’s only with the late 1990s that renovated enthusiasm has gathered around the use of vaccines in cancer therapy, at least in part owing to 2 conceptual advances: (1) Polly Matzinger’s danger theory, proposing that the immune system does not simply react against non-self constituents, but rather respond to situations of danger (irrespective of their origin),Citation3 and (2) the discovery of antigens that are expressed preferentially, when not exclusively, by malignant cells.Citation4

Thus, starting with the late 1990s, significant efforts have been dedicated at the development of preparations that would actively elicit a tumor-associated antigen (TAA)-specific immune response, including dendritic cell (DC)-based,Citation5-Citation7 purified component-based,Citation8-Citation10 and DNA-based vaccines.Citation11 The latter de facto consist in TAA-encoding circularized DNA constructs that are administered to cancer patients (most often via the intramuscular route) either in the form of naked DNA or by means of a suitable vector.Citation11-Citation14 Of note, DNA-based vaccines should be conceptually differentiated from other forms of gene therapy in that (1) they do not directly target diseased (malignant) cells, as it is the case of constructs that code for cytotoxic proteins,Citation15-Citation25 or enzymes that can transform an inactive chemical into a toxic drug;Citation26-Citation31 and (2) they do not boost the immune system in a relatively unspecific fashion, like constructs encoding immunostimulatory cytokines or ligands for co-stimulatory T-cell receptors.Citation19,Citation32-Citation55 Although their efficiency strictly relies on the achievement of high transfection rates and efficient TAA presentation in vivo (see below), DNA-based vaccines have several advantages as compared with their DC-based and purified component-based counterparts, including (but limited to) those provided by genetic engineering. We have discussed these advantages in our latest Trial Watch dealing with the use of DNA-based vaccines in cancer therapy, which has been published in the April 2013 issue of OncoImmunology.Citation56

DNA-based vaccines are expected to enter tissue-resident antigen-presenting cells and/or myocytes, resulting in (1) local TAA synthesis; (2) presentation of TAA-derived peptides to naïve T cells; and (3) the consequent activation of potentially therapeutic TAA-specific CD8+ T cell-dependent and/or humoral immune responses.Citation56,Citation57 Several vectors have been evaluated for their ability to promote efficient TAA-specific immune responses upon the administration of DNA-based vaccines. As it stands, naked constructs remain the preferred form of DNA-based vaccines for clinical applications.Citation11-Citation14 Viral vectors generally yield improved rates of transduction and stable TAA expression,Citation58-Citation61 but are associated with several disadvantages, including the facts that (1) they can elicit potent neutralizing humoral responses (driven by packaging proteins); (2) they are relatively expensive; (3) some viral genomes cannot be engineered to bear large transgenes; and (4) some vectors have been associated with a non-negligible risk of insertional mutagenesis.Citation58,Citation59,Citation62 Along similar lines, prokaryotic as well as eukaryotic vehicles are advantageous as they are compatible with oral administration, are capable of eliciting mucosal immune responses (which are generally considered superior than intramuscular ones), and endogenously express potent immunostimulatory factors (e.g., lipopolysaccharide, bacterial DNA),Citation63-Citation65 yet their large-scale application to clinical settings appears premature at this stage.Citation11-Citation14

Consistent efforts have also been dedicated at the identification of the delivery method that would allow for optimal immune responses to DNA-based vaccines.Citation11,Citation12,Citation14,Citation66,Citation67 In this context, it soon turned out that simple intramuscular injections generally result in poor TAA-specific immune responses, an effect that has been attributed to the limited hydrostatic pressure generated by standard injection volumes.Citation68 Several methods have been proposed as an alternative to intramuscular injections, including gene gun-mediated delivery,Citation69,Citation70 jet injection,Citation71,Citation72 and tattooing,Citation73 all of which involve the transdermal route, as well as oral administration,Citation74-Citation76 and electroporation.Citation69,Citation77-Citation79 Electroporation, i.e., the intramuscular delivery of naked DNA immediately followed by the application of an innocuous electrical stimulus, nowadays stands out as the delivery method of choice for DNA-based vaccines.Citation80-Citation83 This reflects the facts that electroporation (1) is associated with high transfection rates (irrespective of injection volume),Citation68,Citation79,Citation83 (2) causes some extent of local tissue injury, resulting in the release of damage-associated molecular patterns (DAMPs) that operate as endogenous immunostimulants,Citation84-Citation86 and (3) may be perceived as uncomfortable yet is associated with no significant toxicities even when employed repetitively over several vaccination sessions.Citation11,Citation14

The efficacy of all anticancer vaccines, including DNA-based preparations, obviously depends to a large extent on the TAA of choice. For illustrative purposes, TAAs can be classified into 4 mutually exclusive categories: (1) viral TAAs, which are by definition non-self but can be shared by different neoplasms caused by the same virus; (2) unique TAAs, which originate from cancer cell-specific mutations and hence are not shared by distinct tumors, not even of the same type; (3) idiotypic TAAs, another type of tumor-specific TAAs that reflect the rearrangement of transmembrane immunoglobulins expressed by clonal B-cell neoplasms; and (4) shared TAAs, which are also expressed by one or more healthy tissues (though to a limited extent). So-called cancer-testis antigens constitute bona fide shared TAAs, as they are expressed by both malignant and germline cells.Citation87,Citation88 Interestingly, efficient DNA-based vaccines have been constructed that target TAAs from each of these classes have been constructed.Citation56

In spite of a significant amount of encouraging clinical results,Citation56 no DNA-based vaccines are currently licensed by the US Food and Drug Administration (FDA) for use in cancer patients, neither as a prophylactic nor as a therapeutic intervention (source http://www.fda.gov). Conversely, 3 of such preparations have been approved for veterinary use.Citation89-Citation92 Interestingly, one of these DNA-based vaccines, which is employed for the treatment of canine melanoma, relies on a xenogenous TAA (i.e., human tyrosinase).Citation92

One year ago, in the April issue of OncoImmunology, we summarized the scientific rationale behind the use of DNA-based vaccines in cancer therapy and discussed recent clinical trials evaluating the safety and efficacy of this approach.Citation56 Here, we present the latest advances on the clinical development of tumor-targeting DNA-based vaccines.

Update on the Development of Anticancer Vaccines

Since the submission of the latest Trial Watch dealing with this topic,Citation56 i.e., February 2013, the results of only 7 clinical trials testing the safety, immunogenicity and/or therapeutic activity of DNA-based anticancer vaccines have been published in peer-reviewed scientific journals ().Citation93-Citation98 Of these studies, 3 tested naked DNA preparations,Citation93-Citation95 3 relied on viral or eukaryotic vectors,Citation96-Citation98 and 1 tested an heterologous prime-boost approach. In same period, 6 clinical trials have been launched to test the safety and therapeutic potential of DNA-based anticancer vaccines in cancer patients (source http://www.clinicaltrials.gov).

Table 1. Recent clinical trials testing the therapeutic profile of DNA-based vaccines in cancer patients*

Eriksson and colleagues reported the results of a Phase I study involving the intradermal administration of a plasmid encoding kallikrein-related peptidase 3 (KLK3, best known as prostate specific antigen, PSA)Citation99,Citation100 from Macaca mulatta (Rhesus macaque) to 15 patients with biochemical manifestations of relapsing prostate cancer who had previously been subjected to a 1 mo course of androgen deprivation therapy (ADT)Citation101-Citation103 (NCT00859729).Citation94 In this setting, the vaccine was administered by electroporation every 4 wk over 5 consecutive mo. No systemic toxicities were recorded upon vaccination, which was only associated with some discomfort (not requiring analgesia or topical anesthetics) and/or minor skin reactions at the electroporation site. T cells and antibodies specific for xenogenous PSA could be detected in some patients upon vaccination, yet were not associated with clinical efficacy, as all subjects eventually required ADT owing to raising PSA levels. Of note, all but 1 patient exhibited pre-vaccination reactivity against endogenous PSA.Citation94 This suggests that relapsing tumors had already been immunoedited by a PSA-targeting immunological pressure, possibly explaining the lack of efficacy of this approach.Citation94

Tiriveedhi and collaborators characterized the immunological effects of a plasmid coding for secretoglobin, family 2A, member 2 (SCGB2A2, best known as mammaglobin A, MAMA), which is overexpressed by a large fraction (up to 80%) of breast neoplasms,Citation104-Citation106 in 7 patients with Stage IV metastatic breast carcinoma (NCT00807781).Citation95 In this context, MAMA-coding plasmids were administered via the intramuscular route (by means of a jet injector) in 3 vaccination sessions at 4-wk intervals from each other. As this Phase I clinical trial is still ongoing, data on safety and efficacy are not available. Nonetheless, Tiriveedhi et al. reported an increase in circulating CD4+ T cells expressing high levels inducible T-cell co-stimulator (ICOS)Citation107 6 mo after vaccination, a phenomenon that was paralleled by a decline in the levels of blood-borne CD4+FOXP3+ regulatory T cells (Tregs).Citation108-Citation110 Interestingly, MAMA-specific CD4+ICOShigh T cells were found to predominantly express immunosuppressive cytokines such as interleukin-10 (IL-10) before vaccination, but immunostimulatory ones like interferon γ (IFNγ) thereafter. Such a shift in the secretory profile of CD4+ICOShigh T cells was associated with an improved ability to directly lyse MAMA-expressing malignant cells.Citation95 Thus, MAMA-targeting naked DNA-based vaccines may elicit therapeutically relevant immune responses in breast carcinoma patients. The conclusion of this clinical trial is eagerly awaited to shed light on this possibility.

Diaz and colleagues reported the results of 2 multicenter, Phase I, open-label dose-escalation trials testing a bivalent naked DNA-based vaccine targeting carcinoembryonic antigen (CEA)Citation111-Citation113 and v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 2 (ERBB2, best known as HER2),Citation114-Citation116 alone or combined with a dicistronic adenovirus type 6-based vaccine with identical specificity.Citation93 In particular, both the naked DNA-based (V930) and the adenoviral vaccine (V932) encode a truncated variant of HER2 (lacking the cytoplasmic domain) and CEA fused to the B subunit of Escherichia coli heat labile toxin (LTB), as an adjuvant.Citation117,Citation118 In the first study (NCT00250419), 28 patients bearing various HER2- and/or CEA-expressing Stage II-IV malignancies (mainly colorectal, breast, and non-small cell lung carcinomas) received 5 courses of V930 (at 2-wk intervals) by electroporation. With the exception of 2 subjects suffering from Grade 3 abdominal pain and a few individuals reporting minor adverse effects (Grade 1/2 injection site reactions, fatigue, and diarrhea), vaccination was globally well tolerated. However, 3 mo after vaccination, none of the subjects included in this study had developed detectable CEA- or HER2-specific T-cell responses.Citation93 In the second study (NCT00647114), 11 patients with HER2- and/or CEA-expressing Stage II-IV tumors were treated with V930 and V932 in a heterologous prime-boost setting. In this context, the administration of V930 (priming, performed exactly as for the first study) was followed by the intramuscular delivery of V932 in 2 distinct sessions (boosting, 4 and 6 wk after the end of the priming cycle). Similar to V930, V932 was well tolerated. However, V932 failed to improve the ability of V930 to elicit CEA-or HER2-specific adaptive immune responses.Citation93 The precise reasons for such a complete lack of efficacy have not yet been identified.

Stage I/II colorectal cancer patients are currently being recruited in the context of a Phase I clinical trial aimed at testing an adenovirus type 5-based vaccine that encodes guanylyl cyclase 2C (GUCY2C, best known as GCC), which is universally expressed by colorectal cancer cells,Citation119,Citation120 and the so-called pan HLA DR-binding epitope (PADRE), a short peptide that operates as adjuvantCitation121,Citation122 (NCT01972737). Besides evaluating the safety and immunogenicity of this approach, this study aims at investigating whether the development of anti-GCC immunity is related to (1) occult metastases in regional lymph nodes; (2) race (African American vs. Caucasian); and (3) time to recurrence and/or disease-free survival.

Hui and collaborators tested a modified vaccinia virus Ankara (MVA)-based vectorCitation123,Citation124 expressing 2 Epstein–Barr virus (EBV) antigens in patients with EBV-related nasopharyngeal carcinoma.Citation97 All the patients enrolled in this Phase I clinical trial (18 individuals who were in remission for more than 12 wk after the completion of first-line therapies) received 3 intradermal vaccinations at 3-wk intervals. No dose-limiting toxicity was observed, and T-cell responses to either or both EBV antigens, namely, Epstein–Barr nuclear antigen 1 (EBNA1) and latent membrane protein 2 (LMP2),Citation125,Citation126 could be documented in 15 out of 18 (83.3%) patients.Citation97 This vaccination strategy is currently being tested in a larger cohort of patients affected by the same malignancy (NCT01800071).

Three Phase II clinical trials testing recombinant vaccinia and fowlpox viruses as vectors for DNA-based anticancer vaccines have recently been initiated and are currently recruiting patients. Two of the studies investigate the safety and efficacy of an heterologous prime-boost vaccination combined with enzalutamide (an androgen receptor antagonist also known as MDV3100)Citation127-Citation130 in advanced prostate cancer patients (NCT01867333; NCT01875250). In both these settings, priming is achieved by the subcutaneous injection of a recombinant vaccinia virus expressing PSA plus 3 immunostimulatory molecules, namely, CD80 (also known as B7-1), CD58 (also known as LFA-3), and intercellular adhesion molecule 1 (ICAM1).Citation131-Citation135 Boosting is performed by the repeated subcutaneous delivery of a recombinant fowlpox virus encoding the same molecules. The third study aims at comparing the efficacy of a similar prime-boost approach combined with bacillus Calmette–Guérin (BCG)-based immunotherapyCitation63,Citation64 in subjects with high grade non-muscle invasive bladder carcinoma (NCT02015104). In this case, however, the viral vectors employed for vaccination do not encode PSA but CEA and mucin 1 (MUC1), a glycoprotein that is overexpressed or aberrantly glycosylated in multiple tumors.Citation136-Citation138

Slovin and colleagues investigated the safety and therapeutic potential of alphavirus-derived virus-like replicon particles (VRPs)Citation139-Citation141 expressing high levels of folate hydrolase 1 (FOLH1, best known as prostate-specific membrane antigen, PMSA)Citation142,Citation143 in subjects affected by castration-resistant metastatic prostate cancer.Citation98,Citation144 In this Phase I dose-escalation study, 12 patients received up to 4 vaccine doses (at 3-wk intervals), followed by a 5th dose 2 mo later. No systemic or local adverse effects were recorded, indicating that this regimen is well tolerated. However, although all patients were immunologically competent (as demonstrated by appropriate T-cell responses to standard mitogenic assays), none of them developed PSMA-specific cellular immunity. Accordingly, none of these individuals obtained clinical benefits from vaccination. As VRP-neutralizing antibodies were detected in some patients, dosing may have been suboptimal.Citation98 A Phase I clinical trial study has recently been launched to assess the safety and immunogenicity of AVX701, an alphavirus-derived VRP encoding CEA in Stage III colorectal cancer patients (NCT01890213).

Bilusic et al. assessed the clinical profile of a heat-inactivated Saccharomyces cerevisiae strain that had been genetically manipulated to express human CEA (GI-6207).Citation96,Citation145,Citation146 In this Phase I clinical trial, 25 subjects affected by CEA-expressing metastatic carcinomas underwent subcutaneous vaccination every 2 wk for 3 mo, and then monthly. Subcutaneous GI-6207 was well tolerated and was able to induce systemic CEA-specific T-cell responses as well as to decrease the percentage of circulating Tregs, at least in some individuals. Notably, 5 patients experienced stable disease upon vaccination, lasting for more > 3 mo (range: 3.5 to 18 mo).Citation96 A Phase II study has recently been launched to test the same experimental paradigm in patients with medullary thyroid cancer, which is often associated with increased CEA levelsCitation147,Citation148 (NCT01856920). The primary endpoint of this trial will be the effect of GI-6207 on the levels of calcitonin (a circulating marker that correlates with tumor size)Citation149-Citation151 6 mo post-vaccination.

A Phase I clinical trial has been initiated a few months ago to test the therapeutic profile of a prokaryotic vector for DNA-based anticancer vaccination (NCT02002182). This study involves the intravenous administration of a live attenuated strain of Listeria monocytogenes engineered to express the E6/E7 antigens of human papillomavirus (HPV),Citation152-Citation155 named ADX11-001, to patients with Stage II-IV HPV+ oropharyngeal cancer scheduled to undergo ablative surgery. In particular, ADX11-001 will be evaluated for its safety and ability to elicit HPV-specific circulating and intratumoral cytotoxic T-lymphocyte responses.

As for the clinical trials listed in our previous Trial Watch dealing with this topic,Citation56 the following studies have changed status: NCT00669136, now listed as “Terminated”; NCT00629057, NCT01147965, NCT01152398, and NCT01191684, now listed as “Completed”; NCT00669734 and NCT01304524, now listed as “Active, not recruiting”; and NCT01116245 and NCT01064375, whose status is “Unknown” (source http://www.clinicaltrials.gov). NCT00669136, aimed at testing an heterologous prime-boost vaccine targeting the α-fetoprotein (AFP)Citation156 in subjects with hepatocellular carcinoma, was terminated owing to poor accrual and insufficient target population for future accrual. Among “Completed” studies, preliminary results appear to be available only for NCT01147965, a Phase I/II clinical trial aimed at assessing the safety and therapeutic potential of a CEA-encoding adenoviral vector in breast, colorectal, and lung carcinoma patients. In particular, the authors of this study reported that their adenoviral vector is able to elicit TAA- (CEA-)specific cellular immune responses in colorectal cancer patients, in spite of the presence of neutralizing antibodies.Citation157 These findings indicate that some vectors for DNA-based vaccination may be less sensitive than others to natural or immunization-induced humoral responses.

Interestingly, Gavazza and colleagues have recently evaluated the therapeutic profile of a heterologous prime-boost approach in a veterinarian trial.Citation158 In this context, 42 client-owned dogs with Stage III/IV B-cell lymphosarcoma received a conventional chemotherapeutic regimen consisting of cyclophosphamide (an immunogenic alkylating agent),Citation159-Citation162 vincristine (a microtubular poison),Citation163 and prednisolone (a glucocorticoid),Citation164 either alone or combined with a prime-boost vaccine targeting endogenous telomerase reverse transcriptase (TERT), which is hyperactivated in a significant fraction of canine neoplasms.Citation165 Priming relied on 2 intramuscular injections (at a 2-wk interval) of an adenoviral vector expressing a catalytically inactive variant of canine TERT. Boosting was performed 4–6 wk later by administering a naked plasmid coding for the same protein fused to the leader sequence of the human plasminogen activator (at the N-terminus) and LTB (at the C-terminus). Boosting injections were repeated up to 3 times, at 2-wk intervals from each other. Both treatments were extremely well tolerated by the cohort (no signs of toxicities), and 19 out of 21 (90.5%) vaccinated dogs developed a robust T-cell response against canine TERT. Moreover, vaccinated dogs exhibited a significant survival advantage as compared with dogs receiving chemotherapy only (> 76.1 vs. 29.3 wk, respectively).Citation158 These results validate the efficacy of TERT-targeting vaccines in dogs bearing B-cell malignancies,Citation166,Citation167 and support the evaluation of a similar approach for other veterinary neoplasms. TERT-targeting vaccines (including DNA-based, DC-based, and purified component-based formulations) have been intensively investigated as a therapeutic measure against multiple human neoplasms. However, no TERT-targeting preparations are currently approved by the US FDA or other international regulatory agencies for use in cancer patients.Citation6,Citation9,Citation56

Concluding Remarks

As discussed above, several strategies have been devised in the past 2 decades to elicit therapeutically relevant TAA-specific immune responses in cancer patients.Citation5-Citation11 However, only one of these approaches is currently approved by the US FDA and other international regulatory agencies for use in humans. This is the DC-based preparation known as sipuleucel-T (Provenge®), which is currently licensed for the treatment of asymptomatic or minimally symptomatic metastatic castration-resistant prostate cancer.Citation168 Conversely, in spite of encouraging results, no purified component-based and DNA-based anticancer vaccine has yet entered the clinical practice.Citation9,Citation10,Citation56

During the last 13 mo, only a few clinical trials have been initiated to test the safety, immunogenicity and therapeutic potential of DNA-based anticancer vaccines, as if the initial enthusiasm about this (at least hypothetically) powerful immunotherapeutic paradigm were decreasing. Perhaps, this trend reflects a significant number of Phase I/II clinical studies in which DNA-based vaccines were shown to be well tolerated by cancer patients and exerted immunogenic effects, yet failed to elicit therapeutically relevant immune responses.Citation56 We surmise that additional insights into the molecular and systemic factors that allow for the elicitation of robust TAA-specific immunity are required to conceive not only safe, but also efficient DNA-based anticancer vaccines. As vaccines are normally administered in the presence of potent adjuvants, which generally operate via pattern recognition receptors,Citation169 we expect agents other than Toll-like receptor agonists to mediate optimal immunostimulatory effects in this context. The future will tell which, if any, of the multiple immunochemotherapeutic combinatorial regimens that are currently being developedCitation170 endows DNA-based vaccines with the ability to elicit clinically meaningful anticancer immune responses.

Abbreviations:
ADT=

androgen deprivation therapy

CEA=

carcinoembryonic antigen

DC=

dendritic cell

EBV=

Epstein-Barr virus

FDA=

Food and Drug Administration

HPV=

human papillomavirus

ICOS=

inducible T-cell co-stimulator

LTB=

E. coli heat labile toxin, B subunit

MAMA=

mammaglobin A

PSA=

prostate specific antigen

PSMA=

prostate-specific membrane antigen

TAA=

tumor-associated antigen

TERT=

telomerase reverse transcriptase

VRP=

virus-like replicon particle

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

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).

Citation: Pol J, Bloy N, Obrist F, Eggermont A, Galon J, Hervé Fridman W, Cremer I, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: DNA vaccines for cancer therapy. OncoImmunology 2014; 3:e28185; 10.4161/onci.28185

References

  • Waldmann TA. Immunotherapy: past, present and future. Nat Med 2003; 9:269 - 77; http://dx.doi.org/10.1038/nm0303-269; PMID: 12612576
  • Finn OJ. Tumor immunology at the service of cancer immunotherapy. Curr Opin Immunol 2004; 16:127 - 9; http://dx.doi.org/10.1016/j.coi.2004.02.006; PMID: 15023402
  • 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
  • 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
  • Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, Zitvogel L, Kroemer G, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2013; 2:e25771; http://dx.doi.org/10.4161/onci.25771; PMID: 24286020
  • 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
  • Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12:265 - 77; http://dx.doi.org/10.1038/nrc3258; PMID: 22437871
  • 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
  • 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; http://dx.doi.org/10.4161/onci.26621; PMID: 24498550
  • 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
  • Rice J, Ottensmeier CH, Stevenson FK. DNA vaccines: precision tools for activating effective immunity against cancer. Nat Rev Cancer 2008; 8:108 - 20; http://dx.doi.org/10.1038/nrc2326; PMID: 18219306
  • Fioretti D, Iurescia S, Fazio VM, Rinaldi M. DNA vaccines: developing new strategies against cancer. J Biomed Biotechnol 2010; 2010:174378; http://dx.doi.org/10.1155/2010/174378; PMID: 20368780
  • Liu MA. DNA vaccines: an historical perspective and view to the future. Immunol Rev 2011; 239:62 - 84; http://dx.doi.org/10.1111/j.1600-065X.2010.00980.x; PMID: 21198665
  • Stevenson FK, Ottensmeier CH, Rice J. DNA vaccines against cancer come of age. Curr Opin Immunol 2010; 22:264 - 70; http://dx.doi.org/10.1016/j.coi.2010.01.019; PMID: 20172703
  • Roth JA, Nguyen D, Lawrence DD, Kemp BL, Carrasco CH, Ferson DZ, Hong WK, Komaki R, Lee JJ, Nesbitt JC, et al. Retrovirus-mediated wild-type p53 gene transfer to tumors of patients with lung cancer. Nat Med 1996; 2:985 - 91; http://dx.doi.org/10.1038/nm0996-985; PMID: 8782455
  • Maiuri MC, Galluzzi L, Morselli E, Kepp O, Malik SA, Kroemer G. Autophagy regulation by p53. Curr Opin Cell Biol 2010; 22:181 - 5; http://dx.doi.org/10.1016/j.ceb.2009.12.001; PMID: 20044243
  • 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
  • INGN. INGN 201: Ad-p53, Ad5CMV-p53, adenoviral p53, p53 gene therapy--introgen, RPR/INGN 201. Drugs R D 2007; 8:176 - 87; http://dx.doi.org/10.2165/00126839-200708030-00005; PMID: 17472413
  • Chawla SP, Chua VS, Fernandez L, Quon D, Saralou A, Blackwelder WC, Hall FL, Gordon EM. Phase I/II and phase II studies of targeted gene delivery in vivo: intravenous Rexin-G for chemotherapy-resistant sarcoma and osteosarcoma. Mol Ther 2009; 17:1651 - 7; http://dx.doi.org/10.1038/mt.2009.126; PMID: 19532136
  • Madhusudan S, Tamir A, Bates N, Flanagan E, Gore ME, Barton DP, Harper P, Seckl M, Thomas H, Lemoine NR, et al. A multicenter Phase I gene therapy clinical trial involving intraperitoneal administration of E1A-lipid complex in patients with recurrent epithelial ovarian cancer overexpressing HER-2/neu oncogene. Clin Cancer Res 2004; 10:2986 - 96; http://dx.doi.org/10.1158/1078-0432.CCR-03-0291; PMID: 15131034
  • Yoo GH, Hung MC, Lopez-Berestein G, LaFollette S, Ensley JF, Carey M, Batson E, Reynolds TC, Murray JL. Phase I trial of intratumoral liposome E1A gene therapy in patients with recurrent breast and head and neck cancer. Clin Cancer Res 2001; 7:1237 - 45; PMID: 11350889
  • Xing X, Liu V, Xia W, Stephens LC, Huang L, Lopez-Berestein G, Hung MC. Safety studies of the intraperitoneal injection of E1A--liposome complex in mice. Gene Ther 1997; 4:238 - 43; http://dx.doi.org/10.1038/sj.gt.3300376; PMID: 9135737
  • Xing X, Zhang S, Chang JY, Tucker SD, Chen H, Huang L, Hung MC. Safety study and characterization of E1A-liposome complex gene-delivery protocol in an ovarian cancer model. Gene Ther 1998; 5:1538 - 44; http://dx.doi.org/10.1038/sj.gt.3300771; PMID: 9930307
  • Smaldone MC, Davies BJ. BC-819, a plasmid comprising the H19 gene regulatory sequences and diphtheria toxin A, for the potential targeted therapy of cancers. Curr Opin Mol Ther 2010; 12:607 - 16; PMID: 20886393
  • Hanna N, Ohana P, Konikoff FM, Leichtmann G, Hubert A, Appelbaum L, Kopelman Y, Czerniak A, Hochberg A. Phase 1/2a, dose-escalation, safety, pharmacokinetic and preliminary efficacy study of intratumoral administration of BC-819 in patients with unresectable pancreatic cancer. Cancer Gene Ther 2012; 19:374 - 81; http://dx.doi.org/10.1038/cgt.2012.10; PMID: 22498722
  • Trask TW, Trask RP, Aguilar-Cordova E, Shine HD, Wyde PR, Goodman JC, Hamilton WJ, Rojas-Martinez A, Chen SH, Woo SL, et al. Phase I study of adenoviral delivery of the HSV-tk gene and ganciclovir administration in patients with current malignant brain tumors. Mol Ther 2000; 1:195 - 203; http://dx.doi.org/10.1006/mthe.2000.0030; PMID: 10933931
  • Singh S, Cunningham C, Buchanan A, Jolly DJ, Nemunaitis J. Toxicity assessment of intratumoral injection of the herpes simplex type I thymidine kinase gene delivered by retrovirus in patients with refractory cancer. Mol Ther 2001; 4:157 - 60; http://dx.doi.org/10.1006/mthe.2001.0430; PMID: 11482988
  • Germano IM, Fable J, Gultekin SH, Silvers A. Adenovirus/herpes simplex-thymidine kinase/ganciclovir complex: preliminary results of a phase I trial in patients with recurrent malignant gliomas. J Neurooncol 2003; 65:279 - 89; http://dx.doi.org/10.1023/B:NEON.0000003657.95085.56; PMID: 14682378
  • Voges J, Reszka R, Gossmann A, Dittmar C, Richter R, Garlip G, Kracht L, Coenen HH, Sturm V, Wienhard K, et al. Imaging-guided convection-enhanced delivery and gene therapy of glioblastoma. Ann Neurol 2003; 54:479 - 87; http://dx.doi.org/10.1002/ana.10688; PMID: 14520660
  • Kubo H, Gardner TA, Wada Y, Koeneman KS, Gotoh A, Yang L, Kao C, Lim SD, Amin MB, Yang H, et al. Phase I dose escalation clinical trial of adenovirus vector carrying osteocalcin promoter-driven herpes simplex virus thymidine kinase in localized and metastatic hormone-refractory prostate cancer. Hum Gene Ther 2003; 14:227 - 41; http://dx.doi.org/10.1089/10430340360535788; PMID: 12639303
  • Nemunaitis J, Cunningham C, Senzer N, Kuhn J, Cramm J, Litz C, Cavagnolo R, Cahill A, Clairmont C, Sznol M. Pilot trial of genetically modified, attenuated Salmonella expressing the E. coli cytosine deaminase gene in refractory cancer patients. Cancer Gene Ther 2003; 10:737 - 44; http://dx.doi.org/10.1038/sj.cgt.7700634; PMID: 14502226
  • Trudel S, Trachtenberg J, Toi A, Sweet J, Li ZH, Jewett M, Tshilias J, Zhuang LH, Hitt M, Wan Y, et al. A phase I trial of adenovector-mediated delivery of interleukin-2 (AdIL-2) in high-risk localized prostate cancer. Cancer Gene Ther 2003; 10:755 - 63; http://dx.doi.org/10.1038/sj.cgt.7700626; PMID: 14502228
  • Galanis E, Hersh EM, Stopeck AT, Gonzalez R, Burch P, Spier C, Akporiaye ET, Rinehart JJ, Edmonson J, Sobol RE, et al. Immunotherapy of advanced malignancy by direct gene transfer of an interleukin-2 DNA/DMRIE/DOPE lipid complex: phase I/II experience. J Clin Oncol 1999; 17:3313 - 23; PMID: 10506635
  • Belldegrun A, Tso CL, Zisman A, Naitoh J, Said J, Pantuck AJ, Hinkel A, deKernion J, Figlin R. Interleukin 2 gene therapy for prostate cancer: phase I clinical trial and basic biology. Hum Gene Ther 2001; 12:883 - 92; http://dx.doi.org/10.1089/104303401750195854; PMID: 11387054
  • Horton HM, Lalor PA, Rolland AP. IL-2 plasmid electroporation: from preclinical studies to phase I clinical trial. Methods Mol Biol 2008; 423:361 - 72; http://dx.doi.org/10.1007/978-1-59745-194-9_28; PMID: 18370214
  • Heinzerling L, Burg G, Dummer R, Maier T, Oberholzer PA, Schultz J, Elzaouk L, Pavlovic J, Moelling K. Intratumoral injection of DNA encoding human interleukin 12 into patients with metastatic melanoma: clinical efficacy. Hum Gene Ther 2005; 16:35 - 48; http://dx.doi.org/10.1089/hum.2005.16.35; PMID: 15703487
  • Mahvi DM, Henry MB, Albertini MR, Weber S, Meredith K, Schalch H, Rakhmilevich A, Hank J, Sondel P. Intratumoral injection of IL-12 plasmid DNA--results of a phase I/IB clinical trial. Cancer Gene Ther 2007; 14:717 - 23; http://dx.doi.org/10.1038/sj.cgt.7701064; PMID: 17557109
  • Daud AI, DeConti RC, Andrews S, Urbas P, Riker AI, Sondak VK, Munster PN, Sullivan DM, Ugen KE, Messina JL, et al. Phase I trial of interleukin-12 plasmid electroporation in patients with metastatic melanoma. J Clin Oncol 2008; 26:5896 - 903; PMID: 19029422
  • Anwer K, Barnes MN, Fewell J, Lewis DH, Alvarez RD. Phase-I clinical trial of IL-12 plasmid/lipopolymer complexes for the treatment of recurrent ovarian cancer. Gene Ther 2010; 17:360 - 9; http://dx.doi.org/10.1038/gt.2009.159; PMID: 20033066
  • Hernandez-Alcoceba R, Berraondo P. Immunochemotherapy against colon cancer by gene transfer of interleukin-12 in combination with oxaliplatin. Oncoimmunology 2012; 1:97 - 9; http://dx.doi.org/10.4161/onci.1.1.17930; PMID: 22720223
  • Khorana AA, Rosenblatt JD, Sahasrabudhe DM, Evans T, Ladrigan M, Marquis D, Rosell K, Whiteside T, Phillippe S, Acres B, et al. A phase I trial of immunotherapy with intratumoral adenovirus-interferon-gamma (TG1041) in patients with malignant melanoma. Cancer Gene Ther 2003; 10:251 - 9; http://dx.doi.org/10.1038/sj.cgt.7700568; PMID: 12679797
  • Dummer R, Hassel JC, Fellenberg F, Eichmüller S, Maier T, Slos P, Acres B, Bleuzen P, Bataille V, Squiban P, et al. Adenovirus-mediated intralesional interferon-gamma gene transfer induces tumor regressions in cutaneous lymphomas. Blood 2004; 104:1631 - 8; http://dx.doi.org/10.1182/blood-2004-01-0360; PMID: 15161670
  • Nemunaitis J, Fong T, Robbins JM, Edelman G, Edwards W, Paulson RS, Bruce J, Ognoskie N, Wynne D, Pike M, et al. Phase I trial of interferon-gamma (IFN-gamma) retroviral vector administered intratumorally to patients with metastatic melanoma. Cancer Gene Ther 1999; 6:322 - 30; http://dx.doi.org/10.1038/sj.cgt.7700019; PMID: 10419050
  • Merrick AE, Ilett EJ, Melcher AA. JX-594, a targeted oncolytic poxvirus for the treatment of cancer. Curr Opin Investig Drugs 2009; 10:1372 - 82; PMID: 19943208
  • Malmström PU, Loskog AS, Lindqvist CA, Mangsbo SM, Fransson M, Wanders A, Gårdmark T, Tötterman TH. AdCD40L immunogene therapy for bladder carcinoma--the first phase I/IIa trial. Clin Cancer Res 2010; 16:3279 - 87; http://dx.doi.org/10.1158/1078-0432.CCR-10-0385; PMID: 20448220
  • Castro JE, Melo-Cardenas J, Urquiza M, Barajas-Gamboa JS, Pakbaz RS, Kipps TJ. Gene immunotherapy of chronic lymphocytic leukemia: a phase I study of intranodally injected adenovirus expressing a chimeric CD154 molecule. Cancer Res 2012; 72:2937 - 48; http://dx.doi.org/10.1158/0008-5472.CAN-11-3368; PMID: 22505652
  • Nabel GJ, Nabel EG, Yang ZY, Fox BA, Plautz GE, Gao X, Huang L, Shu S, Gordon D, Chang AE. Direct gene transfer with DNA-liposome complexes in melanoma: expression, biologic activity, and lack of toxicity in humans. Proc Natl Acad Sci U S A 1993; 90:11307 - 11; http://dx.doi.org/10.1073/pnas.90.23.11307; PMID: 8248244
  • Nabel GJ, Gordon D, Bishop DK, Nickoloff BJ, Yang ZY, Aruga A, Cameron MJ, Nabel EG, Chang AE. Immune response in human melanoma after transfer of an allogeneic class I major histocompatibility complex gene with DNA-liposome complexes. Proc Natl Acad Sci U S A 1996; 93:15388 - 93; http://dx.doi.org/10.1073/pnas.93.26.15388; PMID: 8986821
  • Rubin J, Galanis E, Pitot HC, Richardson RL, Burch PA, Charboneau JW, Reading CC, Lewis BD, Stahl S, Akporiaye ET, et al. Phase I study of immunotherapy of hepatic metastases of colorectal carcinoma by direct gene transfer of an allogeneic histocompatibility antigen, HLA-B7. Gene Ther 1997; 4:419 - 25; http://dx.doi.org/10.1038/sj.gt.3300396; PMID: 9274718
  • Stopeck AT, Hersh EM, Akporiaye ET, Harris DT, Grogan T, Unger E, Warneke J, Schluter SF, Stahl S. Phase I study of direct gene transfer of an allogeneic histocompatibility antigen, HLA-B7, in patients with metastatic melanoma. J Clin Oncol 1997; 15:341 - 9; PMID: 8996161
  • Gleich LL, Gluckman JL, Armstrong S, Biddinger PW, Miller MA, Balakrishnan K, Wilson KM, Saavedra HI, Stambrook PJ. Alloantigen gene therapy for squamous cell carcinoma of the head and neck: results of a phase-1 trial. Arch Otolaryngol Head Neck Surg 1998; 124:1097 - 104; http://dx.doi.org/10.1001/archotol.124.10.1097; PMID: 9776187
  • Rini BI, Selk LM, Vogelzang NJ. Phase I study of direct intralesional gene transfer of HLA-B7 into metastatic renal carcinoma lesions. Clin Cancer Res 1999; 5:2766 - 72; PMID: 10537340
  • Stopeck AT, Jones A, Hersh EM, Thompson JA, Finucane DM, Gutheil JC, Gonzalez R. Phase II study of direct intralesional gene transfer of allovectin-7, an HLA-B7/beta2-microglobulin DNA-liposome complex, in patients with metastatic melanoma. Clin Cancer Res 2001; 7:2285 - 91; PMID: 11489803
  • Gonzalez R, Hutchins L, Nemunaitis J, Atkins M, Schwarzenberger PO. Phase 2 trial of Allovectin-7 in advanced metastatic melanoma. Melanoma Res 2006; 16:521 - 6; http://dx.doi.org/10.1097/01.cmr.0000232299.44902.41; PMID: 17119453
  • Bedikian AY, Richards J, Kharkevitch D, Atkins MB, Whitman E, Gonzalez R. A phase 2 study of high-dose Allovectin-7 in patients with advanced metastatic melanoma. Melanoma Res 2010; 20:218 - 26; PMID: 20354459
  • 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
  • Haniffa M, Collin M, Ginhoux F. Identification of human tissue cross-presenting dendritic cells: A new target for cancer vaccines. Oncoimmunology 2013; 2:e23140; http://dx.doi.org/10.4161/onci.23140; PMID: 23802067
  • Larocca C, Schlom J. Viral vector-based therapeutic cancer vaccines. Cancer J 2011; 17:359 - 71; http://dx.doi.org/10.1097/PPO.0b013e3182325e63; PMID: 21952287
  • 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; http://dx.doi.org/10.1016/j.molmed.2012.07.007; PMID: 22917663
  • Bridle BW, Clouthier D, Zhang L, Pol J, Chen L, Lichty BD, Bramson JL, Wan Y. Oncolytic vesicular stomatitis virus quantitatively and qualitatively improves primary CD8(+) T-cell responses to anticancer vaccines. Oncoimmunology 2013; 2:e26013; http://dx.doi.org/10.4161/onci.26013; PMID: 24083086
  • Zhou Q, Buchholz CJ. Cell type specific gene delivery by lentiviral vectors: New options in immunotherapy. Oncoimmunology 2013; 2:e22566; http://dx.doi.org/10.4161/onci.22566; PMID: 23483777
  • 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; http://dx.doi.org/10.1126/science.1088547; PMID: 14564000
  • 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
  • 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
  • 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
  • Lu S, Wang S, Grimes-Serrano JM. Current progress of DNA vaccine studies in humans. Expert Rev Vaccines 2008; 7:175 - 91; http://dx.doi.org/10.1586/14760584.7.2.175; PMID: 18324888
  • Nardelli-Haefliger D, Romero P, Jichlinski P. What is the influence of vaccination’s routes on the regression of tumors located at mucosal sites?. Oncoimmunology 2012; 1:242 - 3; http://dx.doi.org/10.4161/onci.1.2.18204; PMID: 22720257
  • 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
  • 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; http://dx.doi.org/10.1016/j.vaccine.2009.07.005; PMID: 19622402
  • Fuller DH, Loudon P, Schmaljohn C. Preclinical and clinical progress of particle-mediated DNA vaccines for infectious diseases. Methods 2006; 40:86 - 97; http://dx.doi.org/10.1016/j.ymeth.2006.05.022; PMID: 16997717
  • Hallermalm K, Johansson S, Bråve A, Ek M, Engström 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; http://dx.doi.org/10.1111/j.1365-3083.2007.01945.x; PMID: 17587345
  • Nguyen-Hoai T, Kobelt D, Hohn O, Vu MD, Schlag PM, Dörken 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; http://dx.doi.org/10.4161/onci.22563; PMID: 23264900
  • 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; http://dx.doi.org/10.1089/hum.2008.073; PMID: 19301471
  • Fest S, Huebener N, Bleeke M, Durmus T, Stermann A, Woehler A, Baykan B, Zenclussen AC, Michalsky E, Jaeger IS, et al. Survivin minigene DNA vaccination is effective against neuroblastoma. Int J Cancer 2009; 125:104 - 14; http://dx.doi.org/10.1002/ijc.24291; PMID: 19291796
  • 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; http://dx.doi.org/10.1186/1471-2407-12-361; PMID: 22906006
  • Meng JZ, Dong YJ, Huang H, Li S, Zhong Y, Liu SL, Wang YD. Oral vaccination with attenuated Salmonella enterica strains encoding T-cell epitopes from tumor antigen NY-ESO-1 induces specific cytotoxic T-lymphocyte responses. Clin Vaccine Immunol 2010; 17:889 - 94; http://dx.doi.org/10.1128/CVI.00044-10; PMID: 20375244
  • Liu J, Kjeken R, Mathiesen I, Barouch DH. Recruitment of antigen-presenting cells to the site of inoculation and augmentation of human immunodeficiency virus type 1 DNA vaccine immunogenicity by in vivo electroporation. J Virol 2008; 82:5643 - 9; http://dx.doi.org/10.1128/JVI.02564-07; PMID: 18353952
  • Ahlén G, Söderholm J, Tjelle T, Kjeken R, Frelin L, Höglund U, Blomberg P, Fons M, Mathiesen I, Sällberg M. In vivo electroporation enhances the immunogenicity of hepatitis C virus nonstructural 3/4A DNA by increased local DNA uptake, protein expression, inflammation, and infiltration of CD3+ T cells. J Immunol 2007; 179:4741 - 53; PMID: 17878373
  • Buchan S, Grønevik 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
  • Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol 1998; 16:867 - 70; http://dx.doi.org/10.1038/nbt0998-867; PMID: 9743122
  • Mathiesen I. Electropermeabilization of skeletal muscle enhances gene transfer in vivo. Gene Ther 1999; 6:508 - 14; http://dx.doi.org/10.1038/sj.gt.3300847; PMID: 10476210
  • Mir LM, Bureau MF, Gehl J, Rangara R, Rouy D, Caillaud JM, Delaere P, Branellec D, Schwartz B, Scherman D. High-efficiency gene transfer into skeletal muscle mediated by electric pulses. Proc Natl Acad Sci U S A 1999; 96:4262 - 7; http://dx.doi.org/10.1073/pnas.96.8.4262; PMID: 10200250
  • Murakami T, Sunada Y. Plasmid DNA gene therapy by electroporation: principles and recent advances. Curr Gene Ther 2011; 11:447 - 56; http://dx.doi.org/10.2174/156652311798192860; PMID: 22023474
  • Galluzzi L, Kepp O, Kroemer G. Immunogenic cell death in radiation therapy. Oncoimmunology 2013; 2:e26536; http://dx.doi.org/10.4161/onci.26536; PMID: 24404424
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2013; 31:51 - 72; http://dx.doi.org/10.1146/annurev-immunol-032712-100008; PMID: 23157435
  • Galluzzi L, Kepp O, Kroemer G. Mitochondria: master regulators of danger signalling. Nat Rev Mol Cell Biol 2012; 13:780 - 8; http://dx.doi.org/10.1038/nrm3479; PMID: 23175281
  • Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/testis antigens, gametogenesis and cancer. Nat Rev Cancer 2005; 5:615 - 25; http://dx.doi.org/10.1038/nrc1669; PMID: 16034368
  • Suri A, Saini S, Sinha A, Agarwal S, Verma A, Parashar D, Singh S, Gupta N, Jagadish N. Cancer testis antigens: A new paradigm for cancer therapy. Oncoimmunology 2012; 1:1194 - 6; http://dx.doi.org/10.4161/onci.20686; PMID: 23170277
  • 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; http://dx.doi.org/10.1128/JVI.75.9.4040-4047.2001; PMID: 11287553
  • 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
  • Diaz CM, Chiappori A, Aurisicchio L, Bagchi A, Clark J, Dubey S, Fridman A, Fabregas JC, Marshall J, Scarselli E, et al. Phase 1 studies of the safety and immunogenicity of electroporated HER2/CEA DNA vaccine followed by adenoviral boost immunization in patients with solid tumors. J Transl Med 2013; 11:62; http://dx.doi.org/10.1186/1479-5876-11-62; PMID: 23497415
  • Eriksson F, Tötterman 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; http://dx.doi.org/10.1016/j.vaccine.2013.06.063; PMID: 23831327
  • 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; http://dx.doi.org/10.1007/s10549-012-2110-9; PMID: 22678162
  • 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; http://dx.doi.org/10.1007/s00262-013-1505-8; PMID: 24327292
  • Hui EP, Taylor GS, Jia H, Ma BB, Chan SL, Ho R, Wong WL, Wilson S, Johnson BF, Edwards C, et al. Phase I trial of recombinant modified vaccinia ankara encoding Epstein-Barr viral tumor antigens in nasopharyngeal carcinoma patients. Cancer Res 2013; 73:1676 - 88; http://dx.doi.org/10.1158/0008-5472.CAN-12-2448; PMID: 23348421
  • Slovin SF, Kehoe M, Durso R, Fernandez C, Olson W, Gao JP, Israel R, Scher HI, Morris S. A phase I dose escalation trial of vaccine replicon particles (VRP) expressing prostate-specific membrane antigen (PSMA) in subjects with prostate cancer. Vaccine 2013; 31:943 - 9; http://dx.doi.org/10.1016/j.vaccine.2012.11.096; PMID: 23246260
  • Catalona WJ, Smith DS, Ratliff TL, Dodds KM, Coplen DE, Yuan JJ, Petros JA, Andriole GL. Measurement of prostate-specific antigen in serum as a screening test for prostate cancer. N Engl J Med 1991; 324:1156 - 61; http://dx.doi.org/10.1056/NEJM199104253241702; PMID: 1707140
  • Stamey TA, Yang N, Hay AR, McNeal JE, Freiha FS, Redwine E. Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate. N Engl J Med 1987; 317:909 - 16; http://dx.doi.org/10.1056/NEJM198710083171501; PMID: 2442609
  • Hussain M, Tangen CM, Berry DL, Higano CS, Crawford ED, Liu G, Wilding G, Prescott S, Kanaga Sundaram S, Small EJ, et al. Intermittent versus continuous androgen deprivation in prostate cancer. N Engl J Med 2013; 368:1314 - 25; http://dx.doi.org/10.1056/NEJMoa1212299; PMID: 23550669
  • Sartor O. Androgen deprivation--continuous, intermittent, or none at all?. N Engl J Med 2012; 367:945 - 6; http://dx.doi.org/10.1056/NEJMe1206814; PMID: 22931264
  • Tang S, Dubey P. Opposing effects of androgen ablation on immune function in prostate cancer. Oncoimmunology 2012; 1:1220 - 1; http://dx.doi.org/10.4161/onci.20448; PMID: 23170287
  • Schmidt HH, Ge Y, Hartmann FJ, Conrad H, Klug F, Nittel S, Bernhard H, Domschke C, Schuetz F, Sohn C, et al. HLA Class II tetramers reveal tissue-specific regulatory T cells that suppress T-cell responses in breast carcinoma patients. Oncoimmunology 2013; 2:e24962; http://dx.doi.org/10.4161/onci.24962; PMID: 23894725
  • 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
  • Zehentner BK, Carter D. Mammaglobin: a candidate diagnostic marker for breast cancer. Clin Biochem 2004; 37:249 - 57; http://dx.doi.org/10.1016/j.clinbiochem.2003.11.005; PMID: 15003725
  • Faget J, Sisirak V, Blay JY, Caux C, Bendriss-Vermare N, Ménétrier-Caux C. ICOS is associated with poor prognosis in breast cancer as it promotes the amplification of immunosuppressive CD4(+) T cells by plasmacytoid dendritic cells. Oncoimmunology 2013; 2:e23185; http://dx.doi.org/10.4161/onci.23185; PMID: 23802069
  • Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T cells: mechanisms of differentiation and function. Annu Rev Immunol 2012; 30:531 - 64; http://dx.doi.org/10.1146/annurev.immunol.25.022106.141623; PMID: 22224781
  • von Boehmer H, Daniel C. Therapeutic opportunities for manipulating T(Reg) cells in autoimmunity and cancer. Nat Rev Drug Discov 2013; 12:51 - 63; http://dx.doi.org/10.1038/nrd3683; PMID: 23274471
  • Blatner NR, Gounari F, Khazaie K. The two faces of regulatory T cells in cancer. Oncoimmunology 2013; 2:e23852; http://dx.doi.org/10.4161/onci.23852; PMID: 23762787
  • Wang D, Rayani S, Marshall JL. Carcinoembryonic antigen as a vaccine target. Expert Rev Vaccines 2008; 7:987 - 93; http://dx.doi.org/10.1586/14760584.7.7.987; PMID: 18767948
  • Sarobe P, Huarte E, Lasarte JJ, Borrás-Cuesta F. Carcinoembryonic antigen as a target to induce anti-tumor immune responses. Curr Cancer Drug Targets 2004; 4:443 - 54; http://dx.doi.org/10.2174/1568009043332916; PMID: 15320719
  • 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
  • Stern HM. Improving treatment of HER2-positive cancers: opportunities and challenges. Sci Transl Med 2012; 4:127rv2; http://dx.doi.org/10.1126/scitranslmed.3001539; PMID: 22461643
  • Hurvitz SA, Hu Y, O’Brien N, Finn RS. Current approaches and future directions in the treatment of HER2-positive breast cancer. Cancer Treat Rev 2013; 39:219 - 29; http://dx.doi.org/10.1016/j.ctrv.2012.04.008; PMID: 22658319
  • Arteaga CL, Sliwkowski MX, Osborne CK, Perez EA, Puglisi F, Gianni L. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol 2011; 9:16 - 32; http://dx.doi.org/10.1038/nrclinonc.2011.177; PMID: 22124364
  • Facciabene A, Aurisicchio L, Elia L, Palombo F, Mennuni C, Ciliberto G, La Monica N. Vectors encoding carcinoembryonic antigen fused to the B subunit of heat-labile enterotoxin elicit antigen-specific immune responses and antitumor effects. Vaccine 2007; 26:47 - 58; http://dx.doi.org/10.1016/j.vaccine.2007.10.060; PMID: 18055074
  • Weltzin R, Guy B, Thomas WD Jr., Giannasca PJ, Monath TP. Parenteral adjuvant activities of Escherichia coli heat-labile toxin and its B subunit for immunization of mice against gastric Helicobacter pylori infection. Infect Immun 2000; 68:2775 - 82; http://dx.doi.org/10.1128/IAI.68.5.2775-2782.2000; PMID: 10768972
  • Hyslop T, Waldman SA. Guanylyl cyclase C as a biomarker in colorectal cancer. Biomark Med 2013; 7:159 - 67; http://dx.doi.org/10.2217/bmm.12.90; PMID: 23387497
  • Carrithers SL, Barber MT, Biswas S, Parkinson SJ, Park PK, Goldstein SD, Waldman SA. Guanylyl cyclase C is a selective marker for metastatic colorectal tumors in human extraintestinal tissues. Proc Natl Acad Sci U S A 1996; 93:14827 - 32; http://dx.doi.org/10.1073/pnas.93.25.14827; PMID: 8962140
  • Alexander J, del Guercio MF, Maewal A, Qiao L, Fikes J, Chesnut RW, Paulson J, Bundle DR, DeFrees S, Sette A. Linear PADRE T helper epitope and carbohydrate B cell epitope conjugates induce specific high titer IgG antibody responses. J Immunol 2000; 164:1625 - 33; PMID: 10640784
  • Rosa DS, Tzelepis F, Cunha MG, Soares IS, Rodrigues MM. The pan HLA DR-binding epitope improves adjuvant-assisted immunization with a recombinant protein containing a malaria vaccine candidate. Immunol Lett 2004; 92:259 - 68; http://dx.doi.org/10.1016/j.imlet.2004.01.006; PMID: 15081621
  • Gilbert SC. Clinical development of Modified Vaccinia virus Ankara vaccines. Vaccine 2013; 31:4241 - 6; http://dx.doi.org/10.1016/j.vaccine.2013.03.020; PMID: 23523410
  • Acres B, Bonnefoy JY. Clinical development of MVA-based therapeutic cancer vaccines. Expert Rev Vaccines 2008; 7:889 - 93; http://dx.doi.org/10.1586/14760584.7.7.889; PMID: 18767940
  • Klein E, Nagy N, Rasul AE. EBV genome carrying B lymphocytes that express the nuclear protein EBNA-2 but not LMP-1: Type IIb latency. Oncoimmunology 2013; 2:e23035; http://dx.doi.org/10.4161/onci.23035; PMID: 23526738
  • Smith C, Khanna R. A new approach for cellular immunotherapy of nasopharyngeal carcinoma. Oncoimmunology 2012; 1:1440 - 2; http://dx.doi.org/10.4161/onci.21286; PMID: 23243622
  • Payton S. Prostate cancer: MDV3100 has antitumor activity in castration-resistant disease. Nat Rev Urol 2010; 7:300; http://dx.doi.org/10.1038/nrurol.2010.69; PMID: 20545033
  • Scher HI, Beer TM, Higano CS, Anand A, Taplin ME, Efstathiou E, Rathkopf D, Shelkey J, Yu EY, Alumkal J, et al, Prostate Cancer Foundation/Department of Defense Prostate Cancer Clinical Trials Consortium. Antitumour activity of MDV3100 in castration-resistant prostate cancer: a phase 1-2 study. Lancet 2010; 375:1437 - 46; http://dx.doi.org/10.1016/S0140-6736(10)60172-9; PMID: 20398925
  • Tsao CK, Oh WK. Urological cancer: Enzalutamide in metastatic CRPC-old dog, new tricks. Nat Rev Clin Oncol 2012; 9:613 - 4; http://dx.doi.org/10.1038/nrclinonc.2012.181; PMID: 23044775
  • Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, de Wit R, Mulders P, Chi KN, Shore ND, et al, AFFIRM Investigators. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med 2012; 367:1187 - 97; http://dx.doi.org/10.1056/NEJMoa1207506; PMID: 22894553
  • Doehn C, Kausch I, Böhmer T, Sommerauer M, Jocham D. Drug evaluation: Therion’s rV-PSA-TRICOM + rF-PSA-TRICOM prime-boost prostate cancer vaccine. Curr Opin Mol Ther 2007; 9:183 - 9; PMID: 17458173
  • 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; http://dx.doi.org/10.1186/1479-5876-4-1; PMID: 16390546
  • Geary SM, Salem AK. Prostate cancer vaccines: Update on clinical development. Oncoimmunology 2013; 2:e24523; http://dx.doi.org/10.4161/onci.24523; PMID: 23762812
  • Gerritsen WR. The evolving role of immunotherapy in prostate cancer. Ann Oncol 2012; 23:Suppl 8 viii22 - 7; http://dx.doi.org/10.1093/annonc/mds259; PMID: 22918924
  • Madan RA, Bilusic M, Heery C, Schlom J, Gulley JL. Clinical evaluation of TRICOM vector therapeutic cancer vaccines. Semin Oncol 2012; 39:296 - 304; http://dx.doi.org/10.1053/j.seminoncol.2012.02.010; PMID: 22595052
  • Brockhausen I. Mucin-type O-glycans in human colon and breast cancer: glycodynamics and functions. EMBO Rep 2006; 7:599 - 604; http://dx.doi.org/10.1038/sj.embor.7400705; PMID: 16741504
  • Devine PL, McKenzie IF. Mucins: structure, function, and associations with malignancy. Bioessays 1992; 14:619 - 25; http://dx.doi.org/10.1002/bies.950140909; PMID: 1365918
  • Reichenbach DK, Finn OJ. Early in vivo signaling profiles in MUC1-specific CD4(+) T cells responding to two different MUC1-targeting vaccines in two different microenvironments. Oncoimmunology 2013; 2:e23429; http://dx.doi.org/10.4161/onci.23429; PMID: 23802084
  • Avogadri F, Merghoub T, Maughan MF, Hirschhorn-Cymerman D, Morris J, Ritter E, Olmsted R, Houghton AN, Wolchok JD. Alphavirus replicon particles expressing TRP-2 provide potent therapeutic effect on melanoma through activation of humoral and cellular immunity. PLoS One 2010; 5:e12670; http://dx.doi.org/10.1371/journal.pone.0012670; PMID: 20844763
  • Morse MA, Hobeika AC, Osada T, Berglund P, Hubby B, Negri S, Niedzwiecki D, Devi GR, Burnett BK, Clay TM, et al. An alphavirus vector overcomes the presence of neutralizing antibodies and elevated numbers of Tregs to induce immune responses in humans with advanced cancer. J Clin Invest 2010; 120:3234 - 41; http://dx.doi.org/10.1172/JCI42672; PMID: 20679728
  • Atkins GJ, Fleeton MN, Sheahan BJ. Therapeutic and prophylactic applications of alphavirus vectors. Expert Rev Mol Med 2008; 10:e33; http://dx.doi.org/10.1017/S1462399408000859; PMID: 19000329
  • Elsässer-Beile U, Bühler P, Wolf P. Targeted therapies for prostate cancer against the prostate specific membrane antigen. Curr Drug Targets 2009; 10:118 - 25; http://dx.doi.org/10.2174/138945009787354601; PMID: 19199907
  • Chang SS. Overview of prostate-specific membrane antigen. Rev Urol 2004; 6:Suppl 10 S13 - 8; PMID: 16985927
  • Galluzzi L. New immunotherapeutic paradigms for castration-resistant prostate cancer. OncoImmunology 2013; 2:e26084; http://dx.doi.org/10.4161/onci.26084
  • Remondo C, Cereda V, Mostböck S, Sabzevari H, Franzusoff A, Schlom J, Tsang KY. Human dendritic cell maturation and activation by a heat-killed recombinant yeast (Saccharomyces cerevisiae) vector encoding carcinoembryonic antigen. Vaccine 2009; 27:987 - 94; http://dx.doi.org/10.1016/j.vaccine.2008.12.002; PMID: 19110021
  • Wansley EK, Chakraborty M, Hance KW, Bernstein MB, Boehm AL, Guo Z, Quick D, Franzusoff A, Greiner JW, Schlom J, et al. Vaccination with a recombinant Saccharomyces cerevisiae expressing a tumor antigen breaks immune tolerance and elicits therapeutic antitumor responses. Clin Cancer Res 2008; 14:4316 - 25; http://dx.doi.org/10.1158/1078-0432.CCR-08-0393; PMID: 18594015
  • Isaacson P, Judd MA. Carcinoembryonic antigen in medullary carcinoma of thyroid. Lancet 1976; 2:1016 - 7; http://dx.doi.org/10.1016/S0140-6736(76)90847-3; PMID: 62227
  • Ishikawa N, Hamada S. Association of medullary carcinoma of the thyroid with carcinoembryonic antigen. Br J Cancer 1976; 34:111 - 5; http://dx.doi.org/10.1038/bjc.1976.133; PMID: 962990
  • Meyer JS, Abdel-Bari W. Granules and thyrocalcitonin-like activity in medullary carcinoma of the thyroid gland. N Engl J Med 1968; 278:523 - 9; http://dx.doi.org/10.1056/NEJM196803072781002; PMID: 5637238
  • Melvin KE, Tashjian AH Jr.. The syndrome of excessive thyrocalcitonin produced by medullary carcinoma of the thyroid. Proc Natl Acad Sci U S A 1968; 59:1216 - 22; http://dx.doi.org/10.1073/pnas.59.4.1216; PMID: 5240025
  • Tashjian AH Jr., Melvin EW. Medullary carcinoma of the thyroid gland. Studies of thyrocalcitonin in plasma and tumor extracts. N Engl J Med 1968; 279:279 - 83; http://dx.doi.org/10.1056/NEJM196808082790602; PMID: 5660301
  • Guirnalda P, Wood L, Goenka R, Crespo J, Paterson Y. Interferon γ-induced intratumoral expression of CXCL9 alters the local distribution of T cells following immunotherapy with Listeria monocytogenes. Oncoimmunology 2013; 2:e25752; http://dx.doi.org/10.4161/onci.25752; PMID: 24083082
  • Rothman J, Paterson Y. Live-attenuated Listeria-based immunotherapy. Expert Rev Vaccines 2013; 12:493 - 504; http://dx.doi.org/10.1586/erv.13.34; PMID: 23659298
  • Guirnalda PD, Paterson Y. Vaccination with immunotherapeutic Listeria monocytogenes induces IL-17(+) γδ T cells in a murine model for HPV associated cancer. Oncoimmunology 2012; 1:822 - 8; http://dx.doi.org/10.4161/onci.20491; PMID: 23162749
  • Mustafa W, Maciag PC, Pan ZK, Weaver JR, Xiao Y, Isaacs SN, Paterson Y. Listeria monocytogenes delivery of HPV-16 major capsid protein L1 induces systemic and mucosal cell-mediated CD4+ and CD8+ T-cell responses after oral immunization. Viral Immunol 2009; 22:195 - 204; http://dx.doi.org/10.1089/vim.2008.0071; PMID: 19435416
  • 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; http://dx.doi.org/10.1111/j.1572-0241.2006.00443.x; PMID: 16542289
  • Morse MA, Chaudhry A, Gabitzsch ES, Hobeika AC, Osada T, Clay TM, Amalfitano A, Burnett BK, Devi GR, Hsu DS, et al. Novel adenoviral vector induces T-cell responses despite anti-adenoviral neutralizing antibodies in colorectal cancer patients. Cancer Immunol Immunother 2013; 62:1293 - 301; http://dx.doi.org/10.1007/s00262-013-1400-3; PMID: 23624851
  • Gavazza A, Lubas G, Fridman A, Peruzzi D, Impellizeri JA, Luberto L, Marra E, Roscilli G, Ciliberto G, Aurisicchio L. Safety and efficacy of a genetic vaccine targeting telomerase plus chemotherapy for the therapy of canine B-cell lymphoma. Hum Gene Ther 2013; 24:728 - 38; http://dx.doi.org/10.1089/hum.2013.112; PMID: 23902422
  • 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
  • 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
  • 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
  • Vitale I, Galluzzi L, Castedo M, Kroemer G. Mitotic catastrophe: a mechanism for avoiding genomic instability. Nat Rev Mol Cell Biol 2011; 12:385 - 92; http://dx.doi.org/10.1038/nrm3115; PMID: 21527953
  • Hsu A, Ritchie DS, Neeson P. Are the immuno-stimulatory properties of Lenalidomide extinguished by co-administration of Dexamethasone?. Oncoimmunology 2012; 1:372 - 4; http://dx.doi.org/10.4161/onci.18963; PMID: 22737619
  • Nasir L. Telomeres and telomerase: Biological and clinical importance in dogs. Vet J 2008; 175:155 - 63; http://dx.doi.org/10.1016/j.tvjl.2007.01.024; PMID: 17398127
  • Peruzzi D, Gavazza A, Mesiti G, Lubas G, Scarselli E, Conforti A, Bendtsen C, Ciliberto G, La Monica N, Aurisicchio L. A vaccine targeting telomerase enhances survival of dogs affected by B-cell lymphoma. Mol Ther 2010; 18:1559 - 67; http://dx.doi.org/10.1038/mt.2010.104; PMID: 20531395
  • Peruzzi D, Mesiti G, Ciliberto G, La Monica N, Aurisicchio L. Telomerase and HER-2/neu as targets of genetic cancer vaccines in dogs. Vaccine 2010; 28:1201 - 8; http://dx.doi.org/10.1016/j.vaccine.2009.11.031; PMID: 19944791
  • 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
  • Greenland JR, Letvin NL. Chemical adjuvants for plasmid DNA vaccines. Vaccine 2007; 25:3731 - 41; http://dx.doi.org/10.1016/j.vaccine.2007.01.120; PMID: 17350735
  • Vacchelli E, Prada N, Kepp O, Galluzzi L. Current trends of anticancer immunochemotherapy. Oncoimmunology 2013; 2:e25396; http://dx.doi.org/10.4161/onci.25396; PMID: 23894726