1,524
Views
73
CrossRef citations to date
0
Altmetric
Review

Trial watch

DNA vaccines for cancer therapy

, , , , , , , & show all
Article: e23803 | Received 27 Jan 2013, Accepted 28 Jan 2013, Published online: 01 Apr 2013

Abstract

The foundation of modern vaccinology dates back to the 1790s, when the English physician Edward Jenner uncovered the tremendous medical potential of prophylactic vaccination. Jenner’s work ignited a wave of nationwide vaccination campaigns abating the incidence of multiple life-threatening infectious diseases and culminating with the eradication of natural smallpox virus, which was definitively certified by the WHO in 1980. The possibility of using vaccines against cancer was first proposed at the end of the 19th century by Paul Ehrlich and William Coley. However, it was not until the 1990s that such a hypothesis began to be intensively investigated, following the realization that the immune system is not completely unresponsive to tumors and that neoplastic cells express immunogenic tumor-associated antigens (TAAs). Nowadays, anticancer vaccines are rapidly moving from the bench to the bedside, and a few prophylactic and therapeutic preparations have already been approved by FDA for use in humans. In this setting, one interesting approach is constituted by DNA vaccines, i.e., TAA-encoding circularized DNA constructs, often of bacterial origin, that are delivered to patients as such or by means of specific vectors, including (but not limited to) liposomal preparations, nanoparticles, bacteria and viruses. The administration of DNA vaccines is most often performed via the intramuscular or subcutaneous route and is expected to cause (1) the endogenous synthesis of the TAA by myocytes and/or resident antigen-presenting cells; (2) the presentation of TAA-derived peptides on the cell surface, in association with MHC class I molecules; and (3) the activation of potentially therapeutic tumor-specific immune responses. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating DNA vaccines as therapeutic interventions against cancer.

Introduction

Historical perspective

In 1980, the WHO officially certified the eradication of natural smallpox infection,Citation1 representing one of the major medical triumphs of history. Such an achievement de facto originated from a series of nationwide vaccination campaigns that were launched throughout the 18th and 19th centuries following the pioneering work of the English physician Edward Anthony Jenner (1749–1823).Citation2,Citation3 In the 1790s, Jenner demonstrated indeed that a sublethal smallpox (or cowpox) infection can confer complete protection against subsequent, potentially lethal, exposures,Citation2,Citation3 establishing the foundations of modern vaccinology. In fact, the term “vaccination” (derived from the Latin adjective vaccinae, which means “pertaining to cows, from cow”) was coined by Jenner himself for the procedure he had conceived to prevent smallpox, and was given a more general meaning by the French microbiologist Louis Pasteur (1822–1895), another central figure in the history of vaccination, only 50 years later.Citation4,Citation5 The development and widespread administration of efficient prophylactic vaccines not only has resulted in the eradication of natural smallpox,Citation1 but also has strikingly abated the incidence of a large panel of life-threatening infectious diseases including (but not limited to) rabies, typhoid, cholera, measles, plague, chickenpox, mumps, poliomyelitis and hepatitis B.Citation4

One century after Jenner’s work, the German physician Paul Ehrlich (1854–1915) and the American surgeon William Bradley Coley (1862–1936) were the first to propose that vaccination might be successfully employed against cancer.Citation4 In fact, Ehrlich (who is best known for the concept of a “magic bullet” that would specifically kill malignant cells) failed in his attempts to formally demonstrate that weakened cancer cells may generate antitumor immunity.Citation4 Conversely, Coley developed a mixture of heat-killed bacteria (best known as the Coley toxin) that mediates potent antitumor effects,Citation6,Citation7 although it does so by operating as an adjuvant, hence stimulating the maturation of dendritic cells (DCs) via Toll-like receptor (TLR)-transduced signals,Citation8 rather than as a bona fide vaccine. Of note, the Coley toxin has been commercially available and administered to cancer patients until the early 1960s, when its use was discontinued following concerns raised by the thalidomide case.Citation9

Unfortunately, the hypotheses of Ehrlich and Coley have been disregarded for about one century and have generated renovated enthusiasm only recently.Citation10 One of the major theoretical hurdles against the development of anticancer vaccines (and, more in general, against the affirmation of tumor immunology as a self-standing discipline) was represented by the “self/non-self” dichotomy, as originally theorized by the Australian virologist Sir Frank Macfarlane Burnet (1899–1985) in 1949.Citation11 According to this model, tumors—as they constitute self tissues—are non-immunogenic and hence completely insensitive to immunotherapeutic interventions.Citation11 It took more than 45 years for an alternative model that globally explains the modus operandi of the immune system to be formulated. Indeed, in 1994, the American scientist Polly Matzinger proposed that the immune system would not simply recognize and react to non-self constituents but would rather be activated by situations of danger, be them of exogenous (non-self) or endogenous (self) origin.Citation12 Thus, conditions that have long been viewed as immunologically silent, including trauma and cancer, are de facto capable of activating the immune system, a concept that is nowadays widely accepted.Citation13-Citation15 Approximately in the same years, (1) the gene coding for MZ2-E, a protein expressed by malignant cells of diverse histological origin but not by a series of normal tissues, was cloned;Citation16 and (2) cytotoxic T lymphocytes (CTLs) specifically recognizing neoplastic cells in vitro were isolated from patients bearing a variety of tumors,Citation16,Citation17 lending further support to the notions that (1) malignant cells express immunogenic tumor-associated antigens (TAAs), whereby they can be discriminated from their normal counterparts, and that (2) at least under selected circumstances, the immune system indeed reacts against neoplastic cells, though in the vast majority of cases such responses are unable to control tumor growth.Citation18

Anticancer vaccines

Within the conceptual framework provided by Polly Matzinger’s danger theory,Citation12 the discovery of MZ2-E, nowadays known as melanoma-associated antigen (MAGE)-A1, ignited an intense experimental effort not only resulting in the identification and characterization of hundreds of additional TAAs, but also generating further insights into the mechanisms whereby TAAs, at least in some settings, can break tolerance and elicit an adaptive immune response.Citation19-Citation21 For didactic purposes, TAAs can be classified into four distinct classes: (1) truly exogenous, non-self TAAs (which are invariably of viral origin); (2) unique, mutated TAAs (stemming from cancer cell-specific genetic alterations); (3) idiotypic TAAs (reflecting the unique way whereby the B-cell receptor expressed by some clonal hematopoietic malignancies is rearranged); and (4) shared TAAs (which are also expressed by normal cells, though often to low levels). A detailed discussion of the properties of these four groups of TAAs largely exceeds the scope of this Trial Watch and can be found in ref. Citation22.

As soon as the first TAAs were characterized, great efforts have been dedicated to the development of anticancer vaccines, resulting in a wealth of different approaches including cell-based strategies (most often involving the loading of autologous DCs with tumor material ex vivo, followed by their re-administration to patients),Citation23 recombinant vaccines (entailing the direct administration of purified TAAs or TAA-derived peptides)Citation22 and DNA vaccines. The results of such an intense wave of research and development have been very encouraging. However, to date only three vaccines have been approved by FDA for use in humans: Cervarix® and Gardasil®, de facto constituting preventive measures against infection by human papillomavirus (HPV)-16 and HPV-18 and the consequent development of cervical carcinoma,Citation24,Citation25 and sipuleucel-T (also known as Provenge®), a cellular preparation for the therapy of asymptomatic or minimally symptomatic metastatic hormone-refractory prostate cancer.Citation26 This is in stark contrast with the huge number of vaccines that have been developed and commercialized during the last century for the prophylaxis of infectious diseases, and may stem from several reasons including (but not limited to): (1) the antigenic properties of malignant cells, (2) the fact that anticancer vaccines must operate in the vast majority of settings as therapeutic—rather than prophylactic—interventions and (3) the existence of multiple immunosuppressive mechanisms that are activated by malignant cells, both in the tumor microenvironment and systemically. A detailed discussion of these points exceeds the scope of this Trial Watch and can be found in ref. Citation22.

Anticancer gene therapy

Along with the recognition of the potential of recombinant DNA technologies, great efforts have been dedicated to the development of constructs that would drive the whole-body or tissue-specific expression of therapeutic genes, as well as of vectors and administration protocols that would allow for the efficient delivery of such constructs to patients.Citation27 Starting in the late 1990s, this intense wave of investigation generated a considerable number of Phase I-II clinical trials testing whether preclinical observations could be safely and efficacy translated from the bench to the bedside.Citation28-Citation30 Indeed, especially in the case of monogenic diseases affecting a relatively accessible cell compartment, such as severe immunodeficiency syndromes caused by the lack of adenosine deaminase (ADA) or the γ chain common to multiple cytokine receptors, gene therapy initially appeared to constitute a relatively safe and highly efficient therapeutic option.Citation31,Citation32 Unfortunately, a few years later the use of retroviral vectors for gene therapy was associated with an increased risk for insertional mutagenesis, de facto abating the general enthusiasm about this therapeutic approach.Citation33 In the same period, the first clinical trials investigating the possibility to employ gene therapy as an anticancer intervention were concluded.Citation34-Citation40 These studies were based on at least three distinct approaches, which continue to be actively investigated nowadays: (1) the selective delivery to malignant cells of genes coding for self-sufficient cytotoxic factors, such as the oncosuppressor protein p53,Citation35,Citation41-Citation43 a cytocydal variant of cyclin G1,Citation44 the adenovirus 5 E1A protein (which de facto functions as an oncosuppressor in breast cancer cells)Citation36,Citation40,Citation45,Citation46 and the diphtheria toxin,Citation47,Citation48 or enzymes that convert inactive drug precursors into poisonous chemicals, like the herpes simplex virus thymidine kinase (which can transform gangiclovir into a lethal triphosphate derivative)Citation49-Citation53 and cytosine deaminase (which can convert 5-fluorocytosine into 5-fluorouracil);Citation54 (2) the (most often intratumoral) administration of plasmids coding for relatively unspecific immunostimulatory factors, including, but not limited to, interleukin (IL)-2,Citation44,Citation55-Citation58 IL-12,Citation59-Citation63 interferon γ (IFNγ),Citation64-Citation66 granulocyte-macrophage colony-stimulating factor (GM-CSF),Citation67 CD40 ligand (CD40L)Citation68,Citation69 and the MHC class I molecule HLA-B7;Citation38,Citation39,Citation70-Citation76 and (3) bona fide DNA vaccines. Of note, none of these gene therapy-based approaches is currently approved by US. FDA for use in cancer patients, yet gendicine, a recombinant adenovirus engineered to express wild-type p53, has been licensed for the treatment of subjects affected by head and neck squamous cell carcinoma in China as early as in 2003.Citation77,Citation78

DNA vaccines

DNA vaccines consist in circular DNA constructs (near-to-invariably derived from bacterial plasmids) that encode one or more TAA(s),Citation79-Citation81 and their use in humans de facto represents a particular case of gene therapy. These vaccines are administered subcutaneously or intramuscularly in the form of naked DNA or within appropriate delivery vectors, resulting in their uptake by resident antigen-presenting cells (APCs), mainly DCs and/or myocytes and local TAA expression. In both scenarios, intracellular TAAs are processed and presented on MHC class I molecules to TAA-specific T cells (direct presentation). However, whereas professional APCs are very efficient at direct presentation, myocytes generally are not, as they express detectable yet rather low levels of MHC class I and co-stimulatory molecules.Citation81,Citation82 Thus, the induction of robust antitumor immunity following the expression of TAAs by myocytes must proceed via cross-presentation, the process whereby APCs take up exogenous material (most often apoptotic debris), process it and eventually present it in association with MHC class I (rather than class II) molecules, eventually resulting in the elicitation of CD8+ T-cell responses.Citation81,Citation83,Citation84 Of note, cross-presentation has been proposed to constitute a major route for the activation of immune responses by DNA vaccines even in settings in which direct presentation can occur, for instance upon the direct delivery of naked DNA to Langerhans cells by gene gun.Citation85

As compared with cell-based and recombinant preparations, DNA vaccines are advantageous in that (1) they can be generated in large amounts and with clinical grade purity in a relatively inexpensive and rapid fashion;Citation79-Citation81,Citation86 (2) they are highly stable (that is, they are relatively insensitive to temperature and have a long shelf life);Citation79-Citation81,Citation86 (3) they are safe, based on experience accumulated in more than one hundred clinical trials completed to date;Citation79-Citation81,Citation86 (4) the presence of bacterial sequences, notably unmethylated CpG islands, in the DNA backbone operates per se as an adjuvant, stimulating the activation of TLR9;Citation87 (5) they can be engineered either for the expression of TAAs fused to non-self proteins that exert adjuvant effects, such as the fragment C of the tetanus toxin,Citation88 Pseudomonas aeruginosa exotoxin,Citation89 the potato virus X coat proteinCitation90 and green fluorescent protein,Citation91 or for the co-expression of other immunostimulatory factors, such as the heat shock 70 KDa protein (HSP70)Citation92,Citation93 and various cytokines, including IL-2, IL-12 and GM-CSF;Citation93-Citation95 (6) they can be engineered so to alter the intracellular routing of TAAs, resulting in the preferential activation of humoral (when TAAs are targeted to the endoplasmic reticulum) or cellular (if TAAs are targeted to the cytosol or—even more specifically—to the proteasome) immunity;Citation96,Citation97 and (7) they can induce very robust T-cell responses (leading to the elimination of APCs at boosting) even if the amounts of TAA produced in situ is minimal.Citation79 However, the efficacy of DNA vaccines is influenced—at least in part—by the achievement of high transfection rates in vivo, raising the need of efficient vectors and administration protocols.

Vectors

Although the use of naked DNA constructs (at least in some circumstances) has been associated with acceptable transfection rates and the elicitation of TAA-specific immune responses, great efforts have recently been dedicated to the optimization of specific vectors for DNA vaccines.Citation79-Citation81,Citation86 The delivery of TAA-coding genes by lentiviral, adenoviral, retroviral and adeno-associated vectors perhaps constitutes the most investigated approach in this sense, offering high levels of transduction efficiency as well as a relatively stable and protracted TAA production.Citation98,Citation99 However, these advantages are largely overcome by the facts that (1) viral packaging proteins are immunogenic and elicit potent anti-vector immune responses, de facto precluding the possibility of efficient boosting in prime-boosting settings, and (2) viral vectors are expensive, cannot host large transgenes, have been associated with toxic side effects and are potentially at risk for insertional mutagenesis.Citation33,Citation98,Citation99 Bacterial and eukaryotic vehicles have been proposed as an alternative to viral vectors, including genetically modified, attenuated strains of Salmonella typhimurium, Pichia pastoris and Saccharomyces cerevisiae.Citation100-Citation104 In general, these systems are advantageous as they are compatible with oral administration, resulting in TAA expression by splenic APCsCitation104,Citation105 or in the induction of potent mucosal immune responses,Citation101 and as multiple bacterial products like lipopolysaccharide (LPS), diacyl lipopeptides, flagellin and bacterial DNA —at least potentially—operate as adjuvants by activating various TLRs.Citation6,Citation7,Citation87,Citation106 This said and in spite of promising preclinical results, currently available bacterial and eukaryotic vectors are generally perceived as insufficiently mature for clinical applications;Citation79-Citation81,Citation86 although a few clinical trials to test their anticancer potential have been launched (see below). Other vectors including liposomes, microparticles, nanoparticles and peculiar polymers are under investigation as a means to increase the transfection rate of DNA vaccines and their immunogenicity, with encouraging results.Citation107,Citation108 Nevertheless, the vast majority of clinical trials ever launched to date for evaluating the antineoplastic potential of DNA vaccines has been based on naked DNA.Citation79-Citation81,Citation86

Delivery methods

Preclinical and clinical data collected during the last two decades demonstrate that the administration route constitutes a critical determinant for the efficacy of DNA vaccines.Citation79,Citation81,Citation86,Citation109,Citation110 Intramuscular injections were commonly employed during early tests with large animals and humans, resulting in relatively poor efficacy. In retrospective, this could have been predicted, as the efficacy of DNA vaccines administered i.m. strictly depends on the injected volume.Citation79,Citation81 Thus, while the intramuscular administration of a DNA vaccine in 50 μL vehicle results in the elicitation of robust immune responses in mice, efficacy is gradually lost along with the decrease in injection volumes.Citation111 Presumably, this stems from the fact that a high hydrostatic pressure not only augments the uptake of the DNA vaccine by myocytes and resident APCs (de facto increasing transfection efficacy) but also promotes (a limited degree of) tissue damage, resulting in the release of danger signals that (1) attract additional APCs and other immune cells to the injection sites and (2) provide immunostimulatory signals via TLRs and other pattern recognition receptors.Citation6,Citation7,Citation15,Citation112-Citation115 Unfortunately, scaling this volume up for the intramuscular administration of DNA vaccines to humans is unfeasible, raising the need for alternative delivery routes. In this sense, several options have been investigated during the last two decades, including (but not limited to) gene gun-mediated delivery,Citation108,Citation116 jet injectionCitation117,Citation118 and tattooing,Citation119 all of which involve the skin route, oral deliveryCitation120-Citation122 and electroporation.Citation116,Citation123-Citation125 Of note, although most (if not all) of these strategies have already entered the clinical phase of development, nowadays electroporation has emerged as a preferred and efficient delivery method.Citation126

Electroporation consists in the electrical stimulation of a skeletal muscle immediately after the intramuscular delivery of naked DNA.Citation127-Citation129 De facto, electroporation is associated with (1) a consistent increase in transfection efficiency and (2) local tissue injury, resulting in the release of danger signals by dying myocytes, the recruitment of immune cells and the establishment of a pro-inflammatory milieu that stimulates robust humoral and cellular immune responses.Citation124,Citation130-Citation132 Of note, the efficacy of DNA vaccines administered via electroporation is not compromised by the use of low injection volumes.Citation111,Citation125 Moreover, although generally perceived as uncomfortable, repeated electroporation appears to cause no major side effects and is accepted by patients with no need for anesthetic procedures.Citation79,Citation81 Finally, although increased transfection efficiencies as achieved with electroporation elevate the risk of (potentially oncogenic) integration, this appears to remain within acceptable levels.Citation133 As it stands, electroporation constitutes the delivery method for DNA vaccines best suited for clinical applications; though ever more encouraging results are being obtained with preparation that exploit the oral route, including bacterial and eukaryotic vectors.Citation121,Citation134

Along the lines of our monthly Trial Watch series,Citation6,Citation7,Citation22,Citation23,Citation135-Citation142 here we will briefly discuss the results of recent clinical trials that have investigated/are investigating the antineoplastic potential of DNA vaccines. As mentioned above, no DNA-based preparation is approved by FDA for use in cancer patients as a prophylactic or immunotherapeutic intervention to date (source www.fda.gov). Conversely, three distinct DNA vaccines have been licensed for veterinary use, including one for the prophylaxis of West Nile virus in horses,Citation143 one for the prophylaxis of infectious hematopoietic necrosis virus (IHNV) in salmonid fishCitation144,Citation145 and one for the therapy of malignant melanoma in dogs.Citation146 Intriguingly, the latter relies on the expression of a xenogenous TAA (i.e., human tyrosinase), resulting in the breakdown of tolerance against the endogenous protein and hence in the development of an efficient humoral response that significantly prolongs the overall survival of melanoma-bearing dogs.Citation147

Naked DNA-Based Anticancer Vaccines

So far, the safety and efficacy of naked DNA vaccines have been evaluated in a relatively restricted number of clinical settings. In particular, constructs coding for autogenic TAAs or allogeneic factors that would exert cross-immunizing functions have been tested in cohorts of B-cell lymphoma patients (TAA: idiotypic B-cell receptor regions),Citation148 head and neck cancer (HNC) patients (immunogen: Mycobacterium leprae HSP65),Citation149 melanoma patients (TAAs: gp100, MART-1-derived peptides, tyrosinase or tyrosinase-derived peptides),Citation150-Citation156 colorectal carcinoma patients (TAA: carcinoembryonic antigen, CEA),Citation157 HPV-16+ cervical intraepithelial neoplasia (CIN) patients (TAA: HPV-16 E6)Citation92 and individuals affected by prostate carcinoma (TAA: prostate-specific antigen, PSA).Citation158,Citation159 The results of these studies (all of which were conducted in a Phase I clinical setting) suggest that the intramuscular, intratumoral and intranodal administration of naked DNA vaccines to cancer patients is safe and can elicit TAA-specific immune responses that—at in least in a fraction of patients—exert bona fide therapeutic effects.

Nowadays (January 2013), official sources list 15 recent (started after January 1, 2008), ongoing (not withdrawn, terminated or completed at the day of submission) clinical trials assessing the safety and efficacy of naked DNA-based vaccines as therapeutic interventions against cancer (). Five of these studies are investigating the therapeutic potential of constructs encoding the E6 and/or E7 proteins of HPV variants that are associated with an increased risk for HNC, cervical cancer and anal carcinoma (i.e., HPV-16 and HPV-18)Citation24,Citation160 either (1) as a plasmid co-encoding the immunostimulatory protein FLT3 ligand, administered i.m. via electroporation, in patients affected by grade 3 CIN (NCT01634503); (2) as a construct co-encoding the immunostimulatory protein calreticulin (CRT),Citation112,Citation161,Citation162 administered as a standalone agent i.m., s.c. or i.t., in subjects affected by grade 2/3 CIN (NCT00988559), or delivered i.m. via electroporation in combination with the immunostimulatory drug cyclophosphamide i.v.Citation13,Citation112,Citation136,Citation142 to HNC patients (NCT01493154); (3) as a plasmid co-encoding the immunostimulatory factor HSP70,Citation112,Citation163 administered i.m. together with a viral vector coding for the same TAAs and topical imiquimodCitation6,Citation7,Citation164 to women bearing grade 3 CIN (NCT00788164); or (4) delivered i.m. via electroporation to patients affected by grade 2/3 CIN (NCT01304524). Of the remaining 10 studies, (1) three are evaluating the safety and efficacy of a construct coding for prostate acid phosphatase (PAP),Citation165,Citation166 administered s.c. in combination with sipuleucel-T and/or GM-CSF to prostate cancer patients (NCT00849121; NCT01341652; NCT01706458); (2) one is investigating the clinical profile of a plasmid coding for mammaglobin A (a secretoglobin that is often overexpressed by breast carcinoma cells),Citation167 administered i.m. as a standalone intervention, in women affected by metastatic breast carcinoma (NCT00807781); (3) one is testing a CEA-coding plasmid,Citation168,Citation169 delivered s.c. via electroporation as a standalone agent or combined with GM-CSF s.c. and cyclophosphamide i.v., in colorectal carcinoma patients (NCT01064375); (4) one is evaluating the therapeutic profile of a prime-boost strategy based on a construct encoding the common TAA α fetoprotein (AFP),Citation170 administered i.m. together with a GM-CSF-coding plasmid (prime) and an AFP-expressing adenoviral vector given i.m. (boost), in hepatocellular carcinoma patients (NCT00669136); (5) one is assessing the safety and efficacy of a plasmid coding for patient-specific, lymphoma-derived single-chain variable fragments (idiotypic vaccination)Citation22,Citation171 fused to a not-better specified chemokine, administered i.m. as a standalone intervention, in subjects affected by lymphoplasmacytic lymphoma (NCT01209871); (6) one is investigating the therapeutic potential of a construct that encodes a tyrosinase-related protein 2 (TRP2) epitope fused to a modified monoclonal antibody targeting the chimera to DCs,Citation172,Citation173 delivered i.m. via electroporation as a standalone intervention to melanoma patients (NCT01138410); (7) one is testing a plasmid coding for residues 1–163 of insulin-like growth factor binding protein 2 (IGFBP-2),Citation174,Citation175 administered s.c. as a single agent, in patients affected by Stage III-IV ovarian cancer (NCT01322802); and (8) one is assessing the safety and efficacy of a construct coding for Macaca mulatta PSA, which is highly homologous to its human counterpart,Citation176-Citation178 delivered s.c. via electroporation to patients bearing relapsed prostate cancer (NCT00859729). Of note, all these naked DNA-based vaccination strategies are currently being tested in Phase I-II clinical settings ().

Table 1. Clinical trials testing naked DNA-based vaccines as therapeutic interventions against cancer*

Vector-Based Anticancer Vaccines

Similar to the case of naked DNA vaccines, the safety and therapeutic potential of vector-based anticancer vaccines have been investigated in a relatively low number of clinical scenarios. In particular, the oral administration of bacterial vectors has only been tested in a cohort of pancreatic cancer patients (TAA: vascular endothelial growth factor receptor 2, VEGFR2);Citation121,Citation179 adenoviral or poxviral vectors (given i.m. or s.c.) have been evaluated in cohorts of non-small cell lung carcinoma (NSCLC) patients (TAA: L523S),Citation180 melanoma patients (TAA: multiple epitopes from distinct melanoma antigens)Citation150,Citation181 and prostate carcinoma patients (TAAs: prostate-specific membrane antigen, PSMA);Citation182,Citation183 and biodegradable polymeric materials have been tested in cohorts of anal dysplasia patients (TAA: HPV-16 E7),Citation184 CIN patients (TAAs: HPV-16 E6/E7)Citation185 and individuals bearing advanced solid tumors (TAA: cytochrome P450 1B1).Citation186 Cumulatively, these clinical trials reported a very low incidence of (near-to-invariably) mild side effects, as well as the development of TAA-specific immune responses that, at least in a subset of patients, translated into a clinical benefit.

Today (January 2013), official sources list 17 recent, ongoing clinical trials investigating the therapeutic potential of vector-based DNA vaccines in cancer patients (). Five of these studies are based on bacterial vectors, as (1) four are testing a live attenuated variant of Listeria monocytogenes engineered to express E7 from HPV-16 (ADXS11-001),Citation187 delivered i.v. either as a standalone intervention to individuals affected by grade 2/3 CIN (NCT01116245), persistent/recurrent cervical carcinoma (NCT01266460) and oropharyngeal cancer (NCT01598792), or in combination with 5-fluorouracil, mitomycin and intensity-modulated radiation therapy to anal carcinoma patients (NCT01671488); and (2) one is assessing the safety and efficacy of an attenuated strain of Salmonella typhimurium encoding VEGFR2 (VXM01),Citation121,Citation179 administered p.o. to patients affected by locally advanced, inoperable Stage IV pancreatic cancer (NCT01486329). Of the remaining 12 studies, all involving (at least in part) viral delivery systems, six are testing Vaccinia virus- or Modified Vaccinia Ankara (MVA) virus-derived vectors (1) either expressing HPV-16 E6 and E7 and co-administered i.m. with a naked plasmid coding for the same TAAs plus HSP70Citation112,Citation163 and topical imiquimodCitation6,Citation7,Citation164 to women affected by grade 3 CIN (NCT00788164);Citation92 (2) either coding for the breast cancer-associated TAA v-erb-b2 erythroblastic leukemia viral oncogene homolog 2 (ERBB2, best known as HER2)Citation188-Citation191 and delivered s.c. as a standalone intervention following adjuvant chemotherapy to individuals affected by ERBB2+ breast cancer (NCT01152398); (3) either encoding both PAPCitation165,Citation166 and PSACitation178 and administered s.c. to androgen-insensitive prostate cancer patients (NCT00629057); (4) either coding for two antigens of the Epstein-Barr virus (i.e., EBNA1, LMP2), which is associated with a fraction of HNC cases,Citation192,Citation193 and delivered s.c. to nasopharyngeal cancer patients with residual viral load after conventional therapy (NCT01094405); (5) either encoding p53,Citation41,Citation42,Citation194 which is frequently overexpressed by a wide variety of neoplasms as a result of inactivating TP53 mutations,Citation195-Citation199 and administered s.c. to subjects affected by gastric, pancreatic or colorectal carcinoma (NCT01191684); (6) or coding for mucin 1 (MUC1)Citation200 plus IL-2 (TG4010)Citation201 and delivered s.c. in combination with conventional chemotherapeutic regimens to Stage IV NSCLC patients (NCT01383148). In addition, (1) three studies are assessing the therapeutic profile of the co-administration of fowlpox virus- and vaccinia virus-derived vectors, either coding for PSACitation178 plus three T-cell co-stimulatory molecules (TRICOM)Citation202,Citation203 and delivered s.c. in association with the microtubular poison docetaxel plus prednisone to metastatic, hormone-resistant prostate cancer patients (NCT01145508), either coding for PSACitation178 plus TRICOMCitation202,Citation203 and delivered together with GM-CSF to subjects affected by metastatic, castration-resistant prostate cancer (NCT01322490), or coding for CEACitation168,Citation169 plus MUC1Citation200 and delivered i.t. and s.c. in combination with GM-CSF to individuals bearing unresectable pancreatic carcinoma (NCT00669734); (2) two trials are testing adenoviral vectors, either encoding AFPCitation170 and delivered i.m. as a boosting strategy following the intramuscular co-administration of AFP- and GM-CSF-coding plasmids (prime) to hepatocellular carcinoma patients (NCT00669136), or coding for CEACitation168,Citation169 (ETBX-011) and administered s.c. as a standalone intervention to patients affected by advanced CEA-expressing breast, lung and colorectal carcinoma (NCT01147965); and (3) one study is investigating the therapeutic potential of a live attenuated strain of the Measles virus (Attenuvax®) delivered s.c. as a single agent to Stage IIIB/IV, Measles virus-positive NSCLC patientsCitation204,Citation205 (NCT00828022). Of note, only two of these approaches are in a relatively advanced stage of clinical development and are tested in Phase III settings (NCT01322490; NCT01383148), i.e., (1) the subcutaneous co-administration of fowlpox virus- and vaccinia virus-derived vectors coding for PSA plus TRICOM in combination with recombinant GM-CSF (for the treatment of prostate cancer); and (2) the subcutaneous delivery of an MVA-derived vector encoding MUC1 plus IL-2 in combination with conventional chemotherapy (for the treatment of NSCLC) (). Future will tell whether either of these strategies will become the first therapeutic DNA vaccine to be approved by FDA for use in cancer patients.

Table 2. Clinical trials testing vector-based DNA vaccines as therapeutic interventions against cancer*

Concluding Remarks

Preclinical and clinical evidence accumulated during the last two decades indicates that DNA vaccines have the potential to induce tumor-specific immune responses that—at least in a fraction of patients—may translate into a therapeutic benefit.Citation79-Citation81,Citation86 Thus, although no DNA vaccines are currently approved by FDA for use in cancer patients, great expectations are reposited on this technology, also linked to the fact that three distinct DNA-based preparations (of which one is employed in a therapeutic—as opposed to prophylactic—setting) have already been licensed for veterinary use.Citation80

DNA vaccines offer great possibilities in that they can be engineered (1) so to express not only the TAA(s) of choice but also immunostimulatory molecules, including cytokines and xenogenous proteins that operate as adjuvants;Citation88-Citation95 and (2) so that the intracellular routing of the TAA(s) of choice is pre-determined, resulting in the preferential elicitation of humoral or cellular immune responses.Citation96,Citation97

The progress of anticancer DNA vaccines toward clinical applications is confronted with the very same issues that complicate the development of other vaccination strategies.Citation22,Citation23 These include the limited availability of clinical grade TLR agonists for use adjuvantsCitation6,Citation7 as well as the problems posed by the immunosuppressive tumor microenvironment, raising the need for the delivery of co-stimulatory signals, such as those elicited by CD40 agonists,Citation206,Citation207 or immune checkpoint inhibitors, such as anti-CTLA4 or anti-PD1 antibodies.Citation135,Citation141 In addition, the ability of DNA-based preparations to elicit TAA-specific immunity is dramatically influenced by transfection efficacy and delivery route, as these two factors dictate not only the amount of TAA that is available for (direct of cross-) presentation, but also the type and intensity of immunostimulatory signals that are released in situ to promote immune responses.Citation79,Citation81 Nowadays, the electroporation of naked DNA is perceived as the approach with a more straightforward path to clinical applications,Citation79,Citation81 whereas the efficacy of viral vectors is limited by the development of anti-vector immune responses that de facto preclude boosting.Citation98,Citation99 Promising results have been also obtained with bacterial and eukaryotic vectors,Citation100-Citation104 yet these tools appear to require a consistent degree of refinement before entering the clinical routine.

Only future will tell whether DNA vaccines will ever make their way from the bench to the bedside and transform from a promising investigational approach into a brilliant clinical reality.

Abbreviations:
ADA=

adenosine deaminase

AFP=

α fetoprotein

APC=

antigen-presenting cell

CD40L=

CD40 ligand

CEA=

carcinoembryonic antigen

CIN=

cervical intraepithelial neoplasia

CRT=

calreticulin

CTL=

cytotoxic T lymphocyte

DC=

dendritic cell

ERBB2=

v-erb-b2 erythroblastic leukemia viral oncogene homolog 2

GM-CSF=

granulocyte-macrophage colony-stimulating factor

HNC=

head and neck cancer

HPV=

human papillomavirus

HSP70=

heat shock 70 KDa protein

IFNγ=

interferon γ

IGFBP-2=

insulin-like growth factor binding protein 2

IHNV, infectious hematopoietic necrosis virus=

IL, interleukin

i.m.=

intra musculum

i.t.=

intra tumorem

i.v.=

intra venam

LPS=

lipopolysaccharide

MAGE=

melanoma-associated antigen

MUC1=

mucin 1

MVA=

Modified Vaccinia Ankara

NSCLC=

non-small cell lung carcinoma

PAP=

prostate acid phosphatase

PSA=

prostate-specific antigen

PSMA=

prostate-specific membrane antigen

p.o.=

per os

s.c.=

sub cutem

TAA=

tumor-associated antigen

TLR=

Toll-like receptor

TRP2=

tyrosinase-related protein 2

VEGFR2=

vascular endothelial growth factor receptor 2

Acknowledgments

The authors are supported by the European Commission (ArtForce); Agence National de la Recherche (ANR); Ligue contre le Cancer (Equipe labelisée); Fondation pour la Recherche Médicale (FRM); Institut National du Cancer (INCa, label SIRIC to SOCRATE project); LabEx Immuno-Oncologie; Fondation de France; Fondation Bettencourt-Schueller; AXA Chair for Longevity Research; Cancéropôle Ile-de-France; and Paris Alliance of Cancer Research Institutes (PACRI).

Disclosure of Potential Conflicts of Interest

No conflicts of interest were disclosed.

Notes

These authors contributed equally to this article.

These authors share senior co-authorship.

References

  • Breman JG, Arita I. The confirmation and maintenance of smallpox eradication. N Engl J Med 1980; 303:1263 - 73; http://dx.doi.org/10.1056/NEJM198011273032204; PMID: 6252467
  • Riedel S. Edward Jenner and the history of smallpox and vaccination. Proc (Bayl Univ Med Cent) 2005; 18:21 - 5; PMID: 16200144
  • Smith KA. Edward jenner and the small pox vaccine. Front Immunol 2011; 2:21; http://dx.doi.org/10.3389/fimmu.2011.00021; PMID: 22566811
  • Waldmann TA. Immunotherapy: past, present and future. Nat Med 2003; 9:269 - 77; http://dx.doi.org/10.1038/nm0303-269; PMID: 12612576
  • Smith KA. Louis pasteur, the father of immunology?. Front Immunol 2012; 3:68; http://dx.doi.org/10.3389/fimmu.2012.00068; PMID: 22566949
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Experimental Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:699 - 716; http://dx.doi.org/10.4161/onci.20696; PMID: 22934262
  • Vacchelli E, Galluzzi L, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 2012; 1:894 - 907; http://dx.doi.org/10.4161/onci.20931; PMID: 23162757
  • Oblak A, Jerala R. Toll-like receptor 4 activation in cancer progression and therapy. Clin Dev Immunol 2011; 2011:609579; http://dx.doi.org/10.1155/2011/609579; PMID: 22110526
  • Ito T, Ando H, Suzuki T, Ogura T, Hotta K, Imamura Y, et al. Identification of a primary target of thalidomide teratogenicity. Science 2010; 327:1345 - 50; http://dx.doi.org/10.1126/science.1177319; PMID: 20223979
  • Finn OJ. Tumor immunology at the service of cancer immunotherapy. Curr Opin Immunol 2004; 16:127 - 9; http://dx.doi.org/10.1016/j.coi.2004.02.006; PMID: 15023402
  • Burgio GR. Commentary on the biological self: Toward a “Biological Ego”. From Garrod’s “chemical individuality” to Burnet’s “self”. Thymus 1990; 16:99 - 117; PMID: 2256127
  • 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
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798
  • Fridman WH, Pagès F, Sautès-Fridman C, Galon J. The immune contexture in human tumours: impact on clinical outcome. Nat Rev Cancer 2012; 12:298 - 306; http://dx.doi.org/10.1038/nrc3245; PMID: 22419253
  • 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
  • van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991; 254:1643 - 7; http://dx.doi.org/10.1126/science.1840703; PMID: 1840703
  • Parmiani G. Tumor immunity as autoimmunity: tumor antigens include normal self proteins which stimulate anergic peripheral T cells. Immunol Today 1993; 14:536 - 8; http://dx.doi.org/10.1016/0167-5699(93)90183-L; PMID: 8274196
  • Rabinovich GA, Gabrilovich D, Sotomayor EM. Immunosuppressive strategies that are mediated by tumor cells. Annu Rev Immunol 2007; 25:267 - 96; http://dx.doi.org/10.1146/annurev.immunol.25.022106.141609; PMID: 17134371
  • Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol 2009; 27:83 - 117; http://dx.doi.org/10.1146/annurev.immunol.021908.132544; PMID: 19007331
  • Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer 2012; 12:265 - 77; http://dx.doi.org/10.1038/nrc3258; PMID: 22437871
  • Vanneman M, Dranoff G. Combining immunotherapy and targeted therapies in cancer treatment. Nat Rev Cancer 2012; 12:237 - 51; http://dx.doi.org/10.1038/nrc3237; PMID: 22437869
  • Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2012; 1:1557 - 76; http://dx.doi.org/10.4161/onci.22428; PMID: 23264902
  • Galluzzi L, Senovilla L, Vacchelli E, Eggermont A, Fridman WH, Galon J, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology 2012; 1:1111 - 34; http://dx.doi.org/10.4161/onci.21494; PMID: 23170259
  • Agosti JM, Goldie SJ. Introducing HPV vaccine in developing countries--key challenges and issues. N Engl J Med 2007; 356:1908 - 10; http://dx.doi.org/10.1056/NEJMp078053; PMID: 17494923
  • Lehtinen M, Paavonen J. Sound efficacy of prophylactic HPV vaccination: Basics and implications. Oncoimmunology 2012; 1:995 - 6; http://dx.doi.org/10.4161/onci.20011; PMID: 23162784
  • Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al, IMPACT Study Investigators. Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med 2010; 363:411 - 22; http://dx.doi.org/10.1056/NEJMoa1001294; PMID: 20818862
  • Yi Y, Noh MJ, Lee KH. Current advances in retroviral gene therapy. Curr Gene Ther 2011; 11:218 - 28; http://dx.doi.org/10.2174/156652311795684740; PMID: 21453283
  • Wasil T, Buchbinder A. Gene therapy in human cancer: report of human clinical trials. Cancer Invest 2000; 18:740 - 6; http://dx.doi.org/10.3109/07357900009012206; PMID: 11107444
  • Sobol RE, Scanlon KJ. Cancer gene therapy clinical trials. Cancer Gene Ther 1995; 2:5 - 6; PMID: 7621255
  • Human gene therapy clinical trials in Europe. Hum Gene Ther 1996; 7:1258 - 9; http://dx.doi.org/10.1089/hum.1996.7.10-1258; PMID: 8793550
  • Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, Gross F, Yvon E, Nusbaum P, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science 2000; 288:669 - 72; http://dx.doi.org/10.1126/science.288.5466.669; PMID: 10784449
  • Bordignon C, Mavilio F, Ferrari G, Servida P, Ugazio AG, Notarangelo LD, et al. Transfer of the ADA gene into bone marrow cells and peripheral blood lymphocytes for the treatment of patients affected by ADA-deficient SCID. Hum Gene Ther 1993; 4:513 - 20; http://dx.doi.org/10.1089/hum.1993.4.4-513; PMID: 8399494
  • Hacein-Bey-Abina S, Von Kalle C, Schmidt M, McCormack MP, Wulffraat N, Leboulch P, 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
  • Quetglas JI, John LB, Kershaw MH, Alvarez-Vallina L, Melero I, Darcy PK, et al. Virotherapy, gene transfer and immunostimulatory monoclonal antibodies. Oncoimmunology 2012; 1:1344 - 54; http://dx.doi.org/10.4161/onci.21679; PMID: 23243597
  • Roth JA, Nguyen D, Lawrence DD, Kemp BL, Carrasco CH, Ferson DZ, 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
  • Yoo GH, Hung MC, Lopez-Berestein G, LaFollette S, Ensley JF, Carey M, et al. 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
  • Hui KM, Ang PT, Huang L, Tay SK. Phase I study of immunotherapy of cutaneous metastases of human carcinoma using allogeneic and xenogeneic MHC DNA-liposome complexes. Gene Ther 1997; 4:783 - 90; http://dx.doi.org/10.1038/sj.gt.3300455; PMID: 9338006
  • Rubin J, Galanis E, Pitot HC, Richardson RL, Burch PA, Charboneau JW, 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, et al. 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
  • Xing X, Zhang S, Chang JY, Tucker SD, Chen H, Huang L, et al. 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
  • 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, et al. 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, 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
  • Xing X, Liu V, Xia W, Stephens LC, Huang L, Lopez-Berestein G, et al. 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
  • 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, et al. 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, 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, 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, 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, et al. 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, 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, 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, et al. 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, et al. 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, et al. 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, 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, 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, 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, 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, et al. 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, et al. 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, et al. 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
  • Gleich LL, Gluckman JL, Armstrong S, Biddinger PW, Miller MA, Balakrishnan K, et al. 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; 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, et al. 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
  • Wilson JM. Gendicine: the first commercial gene therapy product. Hum Gene Ther 2005; 16:1014 - 5; http://dx.doi.org/10.1089/hum.2005.16.1014; PMID: 16149899
  • Peng Z. Current status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers. Hum Gene Ther 2005; 16:1016 - 27; http://dx.doi.org/10.1089/hum.2005.16.1016; PMID: 16149900
  • 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
  • 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
  • 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
  • Shirota H, Petrenko L, Hong C, Klinman DM. Potential of transfected muscle cells to contribute to DNA vaccine immunogenicity. J Immunol 2007; 179:329 - 36; PMID: 17579053
  • Heath WR, Belz GT, Behrens GM, Smith CM, Forehan SP, Parish IA, et al. Cross-presentation, dendritic cell subsets, and the generation of immunity to cellular antigens. Immunol Rev 2004; 199:9 - 26; http://dx.doi.org/10.1111/j.0105-2896.2004.00142.x; PMID: 15233723
  • Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature 1998; 392:86 - 9; http://dx.doi.org/10.1038/32183; PMID: 9510252
  • Stoitzner P, Tripp CH, Eberhart A, Price KM, Jung JY, Bursch L, et al. Langerhans cells cross-present antigen derived from skin. Proc Natl Acad Sci U S A 2006; 103:7783 - 8; http://dx.doi.org/10.1073/pnas.0509307103; PMID: 16672373
  • 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
  • Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, Sanjo H, et al. A Toll-like receptor recognizes bacterial DNA. Nature 2000; 408:740 - 5; http://dx.doi.org/10.1038/35047123; PMID: 11130078
  • King CA, Spellerberg MB, Zhu D, Rice J, Sahota SS, Thompsett AR, et al. DNA vaccines with single-chain Fv fused to fragment C of tetanus toxin induce protective immunity against lymphoma and myeloma. Nat Med 1998; 4:1281 - 6; http://dx.doi.org/10.1038/3266; PMID: 9809552
  • Hung CF, Cheng WF, Hsu KF, Chai CY, He L, Ling M, et al. Cancer immunotherapy using a DNA vaccine encoding the translocation domain of a bacterial toxin linked to a tumor antigen. Cancer Res 2001; 61:3698 - 703; PMID: 11325841
  • Savelyeva N, Munday R, Spellerberg MB, Lomonossoff GP, Stevenson FK. Plant viral genes in DNA idiotypic vaccines activate linked CD4+ T-cell mediated immunity against B-cell malignancies. Nat Biotechnol 2001; 19:760 - 4; http://dx.doi.org/10.1038/90816; PMID: 11479570
  • Wolkers MC, Toebes M, Okabe M, Haanen JB, Schumacher TN. Optimizing the efficacy of epitope-directed DNA vaccination. J Immunol 2002; 168:4998 - 5004; PMID: 11994451
  • Trimble CL, Peng S, Kos F, Gravitt P, Viscidi R, Sugar E, et al. A phase I trial of a human papillomavirus DNA vaccine for HPV16+ cervical intraepithelial neoplasia 2/3. Clin Cancer Res 2009; 15:361 - 7; http://dx.doi.org/10.1158/1078-0432.CCR-08-1725; PMID: 19118066
  • Chen CH, Wang TL, Hung CF, Yang Y, Young RA, Pardoll DM, et al. Enhancement of DNA vaccine potency by linkage of antigen gene to an HSP70 gene. Cancer Res 2000; 60:1035 - 42; PMID: 10706121
  • Perales MA, Yuan J, Powel S, Gallardo HF, Rasalan TS, Gonzalez C, et al. Phase I/II study of GM-CSF DNA as an adjuvant for a multipeptide cancer vaccine in patients with advanced melanoma. Mol Ther 2008; 16:2022 - 9; http://dx.doi.org/10.1038/mt.2008.196; PMID: 18797450
  • Biragyn A, Tani K, Grimm MC, Weeks S, Kwak LW. Genetic fusion of chemokines to a self tumor antigen induces protective, T-cell dependent antitumor immunity. Nat Biotechnol 1999; 17:253 - 8; http://dx.doi.org/10.1038/6995; PMID: 10096292
  • Boyle JS, Koniaras C, Lew AM. Influence of cellular location of expressed antigen on the efficacy of DNA vaccination: cytotoxic T lymphocyte and antibody responses are suboptimal when antigen is cytoplasmic after intramuscular DNA immunization. Int Immunol 1997; 9:1897 - 906; http://dx.doi.org/10.1093/intimm/9.12.1897; PMID: 9466317
  • Rice J, King CA, Spellerberg MB, Fairweather N, Stevenson FK. Manipulation of pathogen-derived genes to influence antigen presentation via DNA vaccines. Vaccine 1999; 17:3030 - 8; http://dx.doi.org/10.1016/S0264-410X(99)00171-1; PMID: 10462238
  • 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, et al. 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
  • Zhang T, Sun L, Xin Y, Ma L, Zhang Y, Wang X, et al. A vaccine grade of yeast Saccharomyces cerevisiae expressing mammalian myostatin. BMC Biotechnol 2012; 12:97; http://dx.doi.org/10.1186/1472-6750-12-97; PMID: 23253888
  • Shin SJ, Bae JL, Cho YW, Lee DY, Kim DH, Yang MS, et al. Induction of antigen-specific immune responses by oral vaccination with Saccharomyces cerevisiae expressing Actinobacillus pleuropneumoniae ApxIIA. FEMS Immunol Med Microbiol 2005; 43:155 - 64; http://dx.doi.org/10.1016/j.femsim.2004.07.004; PMID: 15681145
  • Ruitenberg KM, Gilkerson JR, Wellington JE, Love DN, Whalley JM. Equine herpesvirus 1 glycoprotein D expressed in Pichia pastoris is hyperglycosylated and elicits a protective immune response in the mouse model of EHV-1 disease. Virus Res 2001; 79:125 - 35; http://dx.doi.org/10.1016/S0168-1702(01)00337-9; PMID: 11551653
  • Xiang R, Silletti S, Lode HN, Dolman CS, Ruehlmann JM, Niethammer AG, et al. Protective immunity against human carcinoembryonic antigen (CEA) induced by an oral DNA vaccine in CEA-transgenic mice. Clin Cancer Res 2001; 7:Suppl 856s - 64s; PMID: 11300483
  • Paglia P, Medina E, Arioli I, Guzman CA, Colombo MP. Gene transfer in dendritic cells, induced by oral DNA vaccination with Salmonella typhimurium, results in protective immunity against a murine fibrosarcoma. Blood 1998; 92:3172 - 6; PMID: 9787153
  • Kiflmariam MG, Yang H, Zhang Z. Gene delivery to dendritic cells by orally administered recombinant Saccharomyces cerevisiae in mice. Vaccine 2012; In press http://dx.doi.org/10.1016/j.vaccine.2012.11.048; PMID: 23200937
  • Napolitani G, Rinaldi A, Bertoni F, Sallusto F, Lanzavecchia A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nat Immunol 2005; 6:769 - 76; http://dx.doi.org/10.1038/ni1223; PMID: 15995707
  • 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
  • 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
  • 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, et al. Distribution of DNA vaccines determines their immunogenicity after intramuscular injection in mice. J Immunol 2000; 165:2850 - 8; PMID: 10946318
  • Kroemer G, Galluzzi L, Kepp O, Zitvogel L. Immunogenic cell death in cancer therapy. Annu Rev Immunol 2012; In press PMID: 23157435
  • Vacchelli E, Galluzzi L, Rousseau V, Rigoni A, Tesniere A, Delahaye N, et al. Loss-of-function alleles of P2RX7 and TLR4 fail to affect the response to chemotherapy in non-small cell lung cancer. Oncoimmunology 2012; 1:271 - 8; http://dx.doi.org/10.4161/onci.18684; PMID: 22737602
  • Cirone M, Di Renzo L, Lotti LV, Conte V, Trivedi P, Santarelli R, et al. Activation of dendritic cells by tumor cell death. Oncoimmunology 2012; 1:1218 - 9; http://dx.doi.org/10.4161/onci.20428; PMID: 23170286
  • Pinto A, Rega A, Crother TR, Sorrentino R. Plasmacytoid dendritic cells and their therapeutic activity in cancer. Oncoimmunology 2012; 1:726 - 34; http://dx.doi.org/10.4161/onci.20171; PMID: 22934264
  • Best SR, Peng S, Juang CM, Hung CF, Hannaman D, Saunders JR, et al. 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
  • Hallermalm K, Johansson S, Bråve A, Ek M, Engström G, Boberg A, 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, 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, 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, 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, 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, et al. 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, et al. 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
  • 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
  • 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, et al. 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
  • Babiuk S, Baca-Estrada ME, Foldvari M, Storms M, Rabussay D, Widera G, et al. Electroporation improves the efficacy of DNA vaccines in large animals. Vaccine 2002; 20:3399 - 408; http://dx.doi.org/10.1016/S0264-410X(02)00269-4; PMID: 12213410
  • Tollefsen S, Tjelle T, Schneider J, Harboe M, Wiker H, Hewinson G, et al. Improved cellular and humoral immune responses against Mycobacterium tuberculosis antigens after intramuscular DNA immunisation combined with muscle electroporation. Vaccine 2002; 20:3370 - 8; http://dx.doi.org/10.1016/S0264-410X(02)00289-X; PMID: 12213407
  • Widera G, Austin M, Rabussay D, Goldbeck C, Barnett SW, Chen M, et al. Increased DNA vaccine delivery and immunogenicity by electroporation in vivo. J Immunol 2000; 164:4635 - 40; PMID: 10779767
  • Wang Z, Troilo PJ, Wang X, Griffiths TG, Pacchione SJ, Barnum AB, et al. Detection of integration of plasmid DNA into host genomic DNA following intramuscular injection and electroporation. Gene Ther 2004; 11:711 - 21; http://dx.doi.org/10.1038/sj.gt.3302213; PMID: 14724672
  • Qian BJ, Yan F, Li N, Liu QL, Lin YH, Liu CM, et al. MTDH/AEG-1-based DNA vaccine suppresses lung metastasis and enhances chemosensitivity to doxorubicin in breast cancer. Cancer Immunol Immunother 2011; 60:883 - 93; http://dx.doi.org/10.1007/s00262-011-0997-3; PMID: 21400023
  • Galluzzi L, Vacchelli E, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial Watch: Monoclonal antibodies in cancer therapy. Oncoimmunology 2012; 1:28 - 37; http://dx.doi.org/10.4161/onci.1.1.17938; PMID: 22720209
  • Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, et al. Trial watch: Chemotherapy with immunogenic cell death inducers. Oncoimmunology 2012; 1:179 - 88; http://dx.doi.org/10.4161/onci.1.2.19026; PMID: 22720239
  • Galluzzi L, Vacchelli E, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, et al. Trial Watch: Adoptive cell transfer immunotherapy. Oncoimmunology 2012; 1:306 - 15; http://dx.doi.org/10.4161/onci.19549; PMID: 22737606
  • Vacchelli E, Galluzzi L, Eggermont A, Galon J, Tartour E, Zitvogel L, et al. Trial Watch: Immunostimulatory cytokines. Oncoimmunology 2012; 1:493 - 506; http://dx.doi.org/10.4161/onci.20459; PMID: 22754768
  • Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, et al. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2012; 1:1323 - 43; http://dx.doi.org/10.4161/onci.22009; PMID: 23243596
  • Menger L, Vacchelli E, Kepp O, Eggermont A, Tartour E, Zitvogel L, et al. Trial watch: cardiac glycosides and cancer therapy. Oncoimmunology 2013; 2 In press
  • Vacchelli E, Galluzzi L, Fridman WH, Galon J, Sautes-Fridman C, Tartour E, et al. Trial watch: monoclonal antibodies in cancer therapy. Oncoimmunology 2013; 2; In press http://dx.doi.org/10.4161/onci.22789
  • Vacchelli E, Senovilla L, Eggermont A, Fridman WH, Galon J, Zitvogel L, et al. Trial watch: chemotherapy with immunogenic cell death inducers. Oncoimmunology 2012; 1:179 - 88; PMID: 22720239
  • Davis BS, Chang GJ, Cropp B, Roehrig JT, Martin DA, Mitchell CJ, et al. 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, et al. Long-term survival of dogs with advanced malignant melanoma after DNA vaccination with xenogeneic human tyrosinase: a phase I trial. Clin Cancer Res 2003; 9:1284 - 90; PMID: 12684396
  • Liao JC, Gregor P, Wolchok JD, Orlandi F, Craft D, Leung C, et al. Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma. Cancer Immun 2006; 6:8; PMID: 16626110
  • Timmerman JM, Singh G, Hermanson G, Hobart P, Czerwinski DK, Taidi B, et al. Immunogenicity of a plasmid DNA vaccine encoding chimeric idiotype in patients with B-cell lymphoma. Cancer Res 2002; 62:5845 - 52; PMID: 12384547
  • Victora GD, Socorro-Silva A, Volsi EC, Abdallah K, Lima FD, Smith RB, et al. Immune response to vaccination with DNA-Hsp65 in a phase I clinical trial with head and neck cancer patients. Cancer Gene Ther 2009; 16:598 - 608; http://dx.doi.org/10.1038/cgt.2009.9; PMID: 19197326
  • Dangoor A, Lorigan P, Keilholz U, Schadendorf D, Harris A, Ottensmeier C, et al. Clinical and immunological responses in metastatic melanoma patients vaccinated with a high-dose poly-epitope vaccine. Cancer Immunol Immunother 2010; 59:863 - 73; http://dx.doi.org/10.1007/s00262-009-0811-7; PMID: 20043222
  • Rosenberg SA, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, et al. Inability to immunize patients with metastatic melanoma using plasmid DNA encoding the gp100 melanoma-melanocyte antigen. Hum Gene Ther 2003; 14:709 - 14; http://dx.doi.org/10.1089/104303403765255110; PMID: 12804135
  • Tagawa ST, Lee P, Snively J, Boswell W, Ounpraseuth S, Lee S, et al. Phase I study of intranodal delivery of a plasmid DNA vaccine for patients with Stage IV melanoma. Cancer 2003; 98:144 - 54; http://dx.doi.org/10.1002/cncr.11462; PMID: 12833467
  • Triozzi PL, Aldrich W, Allen KO, Carlisle RR, LoBuglio AF, Conry RM. Phase I study of a plasmid DNA vaccine encoding MART-1 in patients with resected melanoma at risk for relapse. J Immunother 2005; 28:382 - 8; http://dx.doi.org/10.1097/01.cji.0000162779.88687.4c; PMID: 16000957
  • Weber J, Boswell W, Smith J, Hersh E, Snively J, Diaz M, et al. Phase 1 trial of intranodal injection of a Melan-A/MART-1 DNA plasmid vaccine in patients with stage IV melanoma. J Immunother 2008; 31:215 - 23; http://dx.doi.org/10.1097/CJI.0b013e3181611420; PMID: 18481391
  • Wolchok JD, Yuan J, Houghton AN, Gallardo HF, Rasalan TS, Wang J, et al. Safety and immunogenicity of tyrosinase DNA vaccines in patients with melanoma. Mol Ther 2007; 15:2044 - 50; http://dx.doi.org/10.1038/sj.mt.6300290; PMID: 17726460
  • Yuan J, Ku GY, Gallardo HF, Orlandi F, Manukian G, Rasalan TS, et al. Safety and immunogenicity of a human and mouse gp100 DNA vaccine in a phase I trial of patients with melanoma. Cancer Immun 2009; 9:5; PMID: 19496531
  • Conry RM, Curiel DT, Strong TV, Moore SE, Allen KO, Barlow DL, et al. Safety and immunogenicity of a DNA vaccine encoding carcinoembryonic antigen and hepatitis B surface antigen in colorectal carcinoma patients. Clin Cancer Res 2002; 8:2782 - 7; PMID: 12231517
  • Pavlenko M, Roos AK, Lundqvist A, Palmborg A, Miller AM, Ozenci V, et al. A phase I trial of DNA vaccination with a plasmid expressing prostate-specific antigen in patients with hormone-refractory prostate cancer. Br J Cancer 2004; 91:688 - 94; PMID: 15280930
  • Miller AM, Ozenci V, Kiessling R, Pisa P. Immune monitoring in a phase 1 trial of a PSA DNA vaccine in patients with hormone-refractory prostate cancer. J Immunother 2005; 28:389 - 95; http://dx.doi.org/10.1097/01.cji.0000165353.19171.41; PMID: 16000958
  • Marur S, D’Souza G, Westra WH, Forastiere AA. HPV-associated head and neck cancer: a virus-related cancer epidemic. Lancet Oncol 2010; 11:781 - 9; http://dx.doi.org/10.1016/S1470-2045(10)70017-6; PMID: 20451455
  • Senovilla L, Vitale I, Martins I, Tailler M, Pailleret C, Michaud M, et al. An immunosurveillance mechanism controls cancer cell ploidy. Science 2012; 337:1678 - 84; http://dx.doi.org/10.1126/science.1224922; PMID: 23019653
  • Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, et al. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 2011; 334:1573 - 7; http://dx.doi.org/10.1126/science.1208347; PMID: 22174255
  • Garg AD, Krysko DV, Vandenabeele P, Agostinis P. Hypericin-based photodynamic therapy induces surface exposure of damage-associated molecular patterns like HSP70 and calreticulin. Cancer Immunol Immunother 2012; 61:215 - 21; http://dx.doi.org/10.1007/s00262-011-1184-2; PMID: 22193987
  • Holcmann M, Drobits B, Sibilia M. How imiquimod licenses plasmacytoid dendritic cells to kill tumors. Oncoimmunology 2012; 1:1661 - 3; http://dx.doi.org/10.4161/onci.22033; PMID: 23264929
  • 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
  • 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; http://dx.doi.org/10.1016/j.canlet.2005.05.021; PMID: 16046056
  • 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
  • Hammarström S. The carcinoembryonic antigen (CEA) family: structures, suggested functions and expression in normal and malignant tissues. Semin Cancer Biol 1999; 9:67 - 81; http://dx.doi.org/10.1006/scbi.1998.0119; PMID: 10202129
  • Tiernan JP, Perry SL, Verghese ET, West NP, Yeluri S, Jayne DG, et al. Carcinoembryonic antigen is the preferred biomarker for in vivo colorectal cancer targeting. Br J Cancer 2013; In press http://dx.doi.org/10.1038/bjc.2012.605; PMID: 23322207
  • Zhao L, Mou DC, Leng XS, Peng JR, Wang WX, Huang L, et al. Expression of cancer-testis antigens in hepatocellular carcinoma. World J Gastroenterol 2004; 10:2034 - 8; PMID: 15237429
  • Bendandi M. Idiotype vaccines for lymphoma: proof-of-principles and clinical trial failures. Nat Rev Cancer 2009; 9:675 - 81; http://dx.doi.org/10.1038/nrc2717; PMID: 19701243
  • Prins RM, Odesa SK, Liau LM. Immunotherapeutic targeting of shared melanoma-associated antigens in a murine glioma model. Cancer Res 2003; 63:8487 - 91; PMID: 14679014
  • Sarantou T, Chi DD, Garrison DA, Conrad AJ, Schmid P, Morton DL, et al. Melanoma-associated antigens as messenger RNA detection markers for melanoma. Cancer Res 1997; 57:1371 - 6; PMID: 9102226
  • Visintin I, Feng Z, Longton G, Ward DC, Alvero AB, Lai Y, et al. Diagnostic markers for early detection of ovarian cancer. Clin Cancer Res 2008; 14:1065 - 72; http://dx.doi.org/10.1158/1078-0432.CCR-07-1569; PMID: 18258665
  • Kanety H, Kattan M, Goldberg I, Kopolovic J, Ravia J, Menczer J, et al. Increased insulin-like growth factor binding protein-2 (IGFBP-2) gene expression and protein production lead to high IGFBP-2 content in malignant ovarian cyst fluid. Br J Cancer 1996; 73:1069 - 73; http://dx.doi.org/10.1038/bjc.1996.206; PMID: 8624265
  • Lundberg K, Roos AK, Pavlenko M, Leder C, Wehrum D, Guevara-Patiño J, et al. A modified epitope identified for generation and monitoring of PSA-specific T cells in patients on early phases of PSA-based immunotherapeutic protocols. Vaccine 2009; 27:1557 - 65; http://dx.doi.org/10.1016/j.vaccine.2009.01.011; PMID: 19171173
  • Mubiru JN, Hubbard GB, Dick EJ Jr., Furman J, Troyer DA, Rogers J. Nonhuman primates as models for studies of prostate specific antigen and prostatic diseases. Prostate 2008; 68:1546 - 54; http://dx.doi.org/10.1002/pros.20814; PMID: 18668524
  • Dale W. Prostate cancer: PSA testing in older men--are we following the guidelines?. Nat Rev Urol 2012; 9:357 - 8; http://dx.doi.org/10.1038/nrurol.2012.115; PMID: 22641163
  • Smith NR, Baker D, James NH, Ratcliffe K, Jenkins M, Ashton SE, et al. Vascular endothelial growth factor receptors VEGFR-2 and VEGFR-3 are localized primarily to the vasculature in human primary solid cancers. Clin Cancer Res 2010; 16:3548 - 61; http://dx.doi.org/10.1158/1078-0432.CCR-09-2797; PMID: 20606037
  • Nemunaitis J, Meyers T, Senzer N, Cunningham C, West H, Vallieres E, et al. Phase I Trial of sequential administration of recombinant DNA and adenovirus expressing L523S protein in early stage non-small-cell lung cancer. Mol Ther 2006; 13:1185 - 91; http://dx.doi.org/10.1016/j.ymthe.2006.01.013; PMID: 16581300
  • Smith CL, Dunbar PR, Mirza F, Palmowski MJ, Shepherd D, Gilbert SC, et al. Recombinant modified vaccinia Ankara primes functionally activated CTL specific for a melanoma tumor antigen epitope in melanoma patients with a high risk of disease recurrence. Int J Cancer 2005; 113:259 - 66; http://dx.doi.org/10.1002/ijc.20569; PMID: 15386406
  • Todorova K, Ignatova I, Tchakarov S, Altankova I, Zoubak S, Kyurkchiev S, et al. Humoral immune response in prostate cancer patients after immunization with gene-based vaccines that encode for a protein that is proteasomally degraded. Cancer Immun 2005; 5:1; PMID: 15641767
  • Mincheff M, Tchakarov S, Zoubak S, Loukinov D, Botev C, Altankova I, et al. Naked DNA and adenoviral immunizations for immunotherapy of prostate cancer: a phase I/II clinical trial. Eur Urol 2000; 38:208 - 17; http://dx.doi.org/10.1159/000020281; PMID: 10895014
  • Klencke B, Matijevic M, Urban RG, Lathey JL, Hedley ML, Berry M, et al. Encapsulated plasmid DNA treatment for human papillomavirus 16-associated anal dysplasia: a Phase I study of ZYC101. Clin Cancer Res 2002; 8:1028 - 37; PMID: 12006515
  • Garcia F, Petry KU, Muderspach L, Gold MA, Braly P, Crum CP, et al. ZYC101a for treatment of high-grade cervical intraepithelial neoplasia: a randomized controlled trial. Obstet Gynecol 2004; 103:317 - 26; http://dx.doi.org/10.1097/01.AOG.0000110246.93627.17; PMID: 14754702
  • Gribben JG, Ryan DP, Boyajian R, Urban RG, Hedley ML, Beach K, et al. Unexpected association between induction of immunity to the universal tumor antigen CYP1B1 and response to next therapy. Clin Cancer Res 2005; 11:4430 - 6; http://dx.doi.org/10.1158/1078-0432.CCR-04-2111; PMID: 15958627
  • 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
  • Du JW, Xu KY, Fang LY, Qi XL. Clinical significance of Mena and Her-2 expression in breast cancer. Eur J Gynaecol Oncol 2012; 33:455 - 8; PMID: 23185786
  • Dawood S, Broglio K, Gong Y, Yang WT, Cristofanilli M, Kau SW, et al, Inflammatory Breast Cancer Research Group. Prognostic significance of HER-2 status in women with inflammatory breast cancer. Cancer 2008; 112:1905 - 11; http://dx.doi.org/10.1002/cncr.23350; PMID: 18300243
  • Foy KC, Miller MJ, Moldovan N, Bozanovic T, Carson Iii WE, Kaumaya PT. Immunotherapy with HER-2 and VEGF peptide mimics plus metronomic paclitaxel causes superior antineoplastic effects in transplantable and transgenic mouse models of human breast cancer. Oncoimmunology 2012; 1:1004 - 16; http://dx.doi.org/10.4161/onci.21057; PMID: 23170249
  • Foy KC, Miller MJ, Moldovan N, Carson Iii WE, Kaumaya PT. Combined vaccination with HER-2 peptide followed by therapy with VEGF peptide mimics exerts effective anti-tumor and anti-angiogenic effects in vitro and in vivo. Oncoimmunology 2012; 1:1048 - 60; http://dx.doi.org/10.4161/onci.20708; PMID: 23170253
  • Chu EA, Wu JM, Tunkel DE, Ishman SL. Nasopharyngeal carcinoma: the role of the Epstein-Barr virus. Medscape J Med 2008; 10:165; PMID: 18769688
  • 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
  • Li H, Lakshmikanth T, Carbone E, Selivanova G. A novel facet of tumor suppression by p53: Induction of tumor immunogenicity. Oncoimmunology 2012; 1:541 - 3; http://dx.doi.org/10.4161/onci.19409; PMID: 22754780
  • Vojtĕsek B, Lane DP. Regulation of p53 protein expression in human breast cancer cell lines. J Cell Sci 1993; 105:607 - 12; PMID: 8408290
  • Yaginuma Y, Westphal H. Abnormal structure and expression of the p53 gene in human ovarian carcinoma cell lines. Cancer Res 1992; 52:4196 - 9; PMID: 1638534
  • Bodner SM, Minna JD, Jensen SM, D’Amico D, Carbone D, Mitsudomi T, et al. Expression of mutant p53 proteins in lung cancer correlates with the class of p53 gene mutation. Oncogene 1992; 7:743 - 9; PMID: 1565469
  • Soussi T. p53 alterations in human cancer: more questions than answers. Oncogene 2007; 26:2145 - 56; http://dx.doi.org/10.1038/sj.onc.1210280; PMID: 17401423
  • Soussi T, Lozano G. p53 mutation heterogeneity in cancer. Biochem Biophys Res Commun 2005; 331:834 - 42; http://dx.doi.org/10.1016/j.bbrc.2005.03.190; PMID: 15865939
  • Kufe DW. Mucins in cancer: function, prognosis and therapy. Nat Rev Cancer 2009; 9:874 - 85; http://dx.doi.org/10.1038/nrc2761; PMID: 19935676
  • Limacher JM, Quoix E. TG4010: A therapeutic vaccine against MUC1 expressing tumors. Oncoimmunology 2012; 1:791 - 2; http://dx.doi.org/10.4161/onci.19863; PMID: 22934285
  • 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
  • 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
  • Zhao D, Chen P, Yang H, Wu Y, Zeng X, Zhao Y, et al. Live attenuated measles virus vaccine induces apoptosis and promotes tumor regression in lung cancer. Oncol Rep 2013; 29:199 - 204; PMID: 23129111
  • Sion-Vardy N, Lasarov I, Delgado B, Gopas J, Benharroch D, Ariad S. Measles virus: evidence for association with lung cancer. Exp Lung Res 2009; 35:701 - 12; http://dx.doi.org/10.3109/01902140902853176; PMID: 19895323
  • Diehl L, den Boer AT, Schoenberger SP, van der Voort EI, Schumacher TN, Melief CJ, et al. CD40 activation in vivo overcomes peptide-induced peripheral cytotoxic T-lymphocyte tolerance and augments anti-tumor vaccine efficacy. Nat Med 1999; 5:774 - 9; http://dx.doi.org/10.1038/10495; PMID: 10395322
  • Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF, Heath WR. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature 1998; 393:478 - 80; http://dx.doi.org/10.1038/30996; PMID: 9624004