2,976
Views
53
CrossRef citations to date
0
Altmetric
Research Papers

DNA vaccines, electroporation and their applications in cancer treatment

, &
Pages 1889-1900 | Received 01 Dec 2014, Accepted 25 Mar 2015, Published online: 11 Aug 2015

Abstract

Numerous animal studies and recent clinical studies have shown that electroporation-delivered DNA vaccines can elicit robust Ag-specific CTL responses and reduce disease severity. However, cancer antigens are generally poorly immunogenic, requiring special conditions for immune response induction. To date, many different approaches have been used to elicit Ag-specific CTL and anti-neoplastic responses to DNA vaccines against cancer. In vivo electroporation is one example, whereas others include DNA manipulation, xenogeneic antigen use, immune stimulatory molecule and immune response regulator application, DNA prime-boost immunization strategy use and different DNA delivery methods. These strategies likely increase the immunogenicity of cancer DNA vaccines, thereby contributing to cancer eradication. However, cancer cells are heterogeneous and might become CTL-resistant. Thus, understanding the CTL resistance mechanism(s) employed by cancer cells is critical to develop counter-measures for this immune escape. In this review, the use of electroporation as a DNA delivery method, the strategies used to enhance the immune responses, the cancer antigens that have been tested, and the escape mechanism(s) used by tumor cells are discussed, with a focus on the progress of clinical trials using cancer DNA vaccines.

Abbreviations

AFP=

α-fetoprotein

APCs=

antigen presenting cells

CEA=

carcinoembryonic antigen

CTLA-4=

cytotoxic T lymphocyte-associated antigen-4

DCs=

dendritic cells

EP=

electroporation

GITR=

glucocorticoid-induced tumor necrosis factor receptor family-related gene

hTERT=

human telomerase reverse transcriptase

HPV=

human papillomavirus

HSP=

heat shock protein

HSV=

herpes simplex virus

ID=

intradermal

IM=

intramuscular

MAGE=

melanoma-associated antigen

MART=

melanoma antigen recognized by T cells

PAP=

prostatic acid phosphatase

PD=

programmed death

PRAME=

preferentially expressed antigen in melanoma

PSA=

prostate-specific antigen

PSMA=

prostate-specific membrane antigen

WT1=

Wilm's tumor

Introduction

DNA vaccines are not replicating and the vaccine products are expressed within the host cells. They can be constructed to mimic the specificity and safety of subunit vaccines. Due to the production and processing of immunogenic proteins within host cells, DNA vaccines are likely to induce immune responses in a manner similar to live-attenuated vaccine types while causing no pathogenic infection in vivo. By directly administering DNA vaccines into the host, the host cells express the antigenic protein. This process has been known to induce both Ag-specific antibody and cellular responses.Citation1-3 DNA vaccines are also expected to be safe, stable and cost-effective in their applications, and are easily and rapidly engineered by recent recombinant DNA technologies.

For delivery of DNA vaccines, the majority of DNA vaccine studies have utilized muscle or skin as an immunization target. In intramuscular (IM) injection, DNA vaccines are taken up and expressed by muscle cells and local antigen presenting cells (APCs). The local APCs then migrate to the draining lymph nodes for the induction of adaptive immune responses.Citation4,5 The APCs, not muscle cells are believed to present antigen to CD4+ and CD8+ T cells by cross-presentation of secreted antigen or by direct transfection of DNA vaccines.Citation3,6,7 In this case, APCs can provide co-stimulatory signals and cytokines necessary for stimulation of naïve T cells. In addition, antigenic proteins expressed and/or secreted by APCs or non-hematogenous target tissues (e.g., muscle cells in the case of IM injection and keratinocytes in the case of intradermal injection) are presumably recognized by B cells for subsequent antibody production in association with helper functions of Ag-specific CD4+ T cells. In intradermal (ID) delivery, DNA can be taken up by Langerhans cells and/or dermal dendritic cells, which migrate to the draining lymph nodes for induction of adaptive immune responses.Citation8 In particular, DNA vaccines, which are generated from bacteria, contain un-methylated CpG motifs and stimulate the innate immune responses by interacting with Toll-like receptor 9 expressed on the surface of APCs (reviewed in Ref. Citation9). This non-specific activation of APCs likely influences Ag-specific immune responses to DNA vaccines.

For increasing the translation efficacy of DNA vaccines, the genes that code for antigens are codon-optimized and are even linked with specific DNA sequences encoding products that behave as immune-stimulatory or intracellular antigen-targeting molecules. Moreover, for the augmentation of adaptive immune responses, cancer DNA vaccines are co-delivered with cytokines and/or immune response regulators as adjuvants. The DNA prime-boost approach and xenogeneic antigen use have also been proven to be useful strategies to enhance adaptive immune responses. These molecular and biological strategies have contributed to upgrading the immunogenic quality of DNA vaccines.

Cancer antigens are often thought to be poorly immunogenic due to their self-tolerance state. Thus, overcoming immunological tolerance to cancer antigens is a pre-requisite for the induction of potent adaptive immune responses in cancer DNA vaccination. Moreover, cancer antigens are expressed in normal tissues. Thus, cancer-reactive immune cells might attack the normal tissues, possibly causing autoimmune diseases. To date, a number of cancer antigens have been reported. However, continuous work is still needed to identify more potential cancer antigens from malignancies.

In cancer immunotherapy, CTLs are thought to be a major effector cell population targeting neoplastic cells for their lysis. Cancer-reactive CTLs kill neoplastic cells through 2 major killing pathways, the perforin/granzyme B-mediated and Fas/Fas ligand-mediated pathways.Citation10,11 Despite the induction and potentiation of cancer Ag-specific CTL activity, cancer cells may become immune-resistant cells, no longer responding to the CTL recognition or attacks. This immune-resistant state of cancer cells is often observed after long-term immunotherapy or under conditions of immune selection. It has been reported that cancer cells develop many different mechanism(s) to resist host immune surveillance (reviewed in Ref. Citation12). The unresponsiveness of cancer cells to cancer-reactive CTLs undermines the ultimate usefulness of cancer DNA vaccines in treating cancer patients. Therefore, it is likely that blocking the immune escape processes of cancer cells is important for achieving the ultimate goal of curing cancer.

As the cancer-killing mechanism(s) of DNA vaccination and conventional therapy modalities such as surgery, radiation, and chemotherapy are all variable, combined therapy using these different modalities might be an option to limit the chance of cancer cells' becoming immune-resistant, thereby achieving better cancer control. In this context, DNA vaccines coding for numerous cancer-specific and cancer-associated antigens have been tested for their safety and effectiveness pre-clinically, as well as in patients. In this article, delivery methods of DNA vaccines, the manipulation of antigens and the use of xenogeneic antigens, approaches to enhance the immunogenicity of DNA vaccines, the types of cancer antigens tested, as well as the immune evasion mechanism(s) utilized by cancer cells and the possible counter-measures for this immune escape will be discussed.

Electroporation as a DNA delivery method

Because the construction and mass production of DNA vaccines are far easier than those of other conventional vaccine types (live attenuated or killed vaccines, and subunit vaccines), as well as their advantages in bio-stability and biosafety, DNA vaccines are thought of as a second-generation vaccine. Historically, striated muscles were first demonstrated to be a site of gene expression following naked DNA injection.Citation13 Subsequently, needle injection of DNAs encoding viral antigens into the muscle sites was reported to induce Ag-specific antibody and cellular responses (CTL responses, in particular) in influenza and HIV-1 animal models.Citation2,14 At the same time, ID delivery of cDNA coding for antigens was also shown to induce Ag-specific antibody and CTL responses.Citation15 These early animal studies showed that Ag-encoding DNA molecules could be taken up and expressed by striated muscle or skin cells for the induction of adaptive immune responses. Since then, human studies have been initiated to evaluate the efficacy of DNA vaccines in treating various diseases. Contrary to the data on small animal models, the immunogenicity of DNA vaccines in large animals and humans has been far below what is expected,Citation16-18 possibly due to a low level of protein expression by DNA molecules in relation to the body size and/or a lack of APCs at the DNA injection sites. To overcome this hurdle, many different approaches such as co-injection of DNA vaccines with various immune stimulatory molecules, DNA codon optimization and the targeting of antigens to different cellular compartments, ligation of immune stimulatory molecules to antigens, DNA prime-boost injection schemes and direct injection of DNA vaccines into the lymph nodes have been taken,Citation19-21 (reviewed in Ref. Citation22,23). In addition, different DNA delivery methods including gene gun delivery, jet injection, electroporation, tattooing, use of liposome, polymers and nanoparticles have also been tested,Citation24,25 (reviewed in Ref. Citation22). Among these methods, gene gun delivery and electroporation methods have been most intensively studied. For instance, a DNA/gold particle bombardment strategy using a gene gun was able to induce potent Ag-specific CTL responses even at a lower dose of DNA than is used for IM injection.Citation26 This gene gun delivery technology utilized the bombardment of DNA/gold particles on the skin surface, where the DNA molecules were taken up and expressed by local dendritic cells (DCs) for antigen processing and presentation.Citation27 Later, in vivo electroporation (EP) was employed as a DNA delivery method, resulting in a dramatic effect on adaptive immune response induction. When compared with gene gun delivery, in vivo EP of DNA plasmid vectors into the muscles was shown to be more effective for protein production and Ag-specific CTL response induction.Citation28 IM-EP is known to increase muscle cell permeability, thus permitting more DNA molecules to enter into the cells for subsequent protein expression.Citation29-31 IM-EP also increases distribution and cellular uptake of DNA molecules at the injection sites.Citation32 In addition, the injection of needle probes and the application of subsequent electric pulses act to attract immune cells, including DCs, to the DNA injection sites, thus increasing the magnitude and duration of the Ag-specific immune response.Citation33 In a phase I clinical trial, IM-EP of human papillomavirus (HPV) E6 and E7 DNA vaccines induced robust antibody and CTL responses.Citation34 In a phase II trial, the same DNA vaccine regimens displayed clinical benefits by decreasing the HPV viral load and the HPV-associated disease severity (personal communication with Bagarazzi M). Moreover, ID-EP of DNA vaccines has also been shown to induce both robust and durable immune responses in primates.Citation35 Simultaneously, intratumoral EP of either DNA coding for cytokines or chemotherapeutic drugs has also been shown to have a positive effect on treating skin tumors in animals and humans.Citation36-40 EP of cDNA expressing antibodies has also been tested as a passive immune therapy strategy. In particular, our un-reported data demonstrated that a serum anti-HER2 antibody was detectable for over 90 days after a single IM-EP of 50 μg of anti-HER2 IgG heavy chain cDNA and 50 μg of anti-HER2 IgG light chain cDNA in mice, suggesting the potential benefit of EP in the passive delivery of antibodies in the form of DNA for cancer treatment. Taken together, EP is thought to be the most potent DNA delivery method for protein expression and subsequent immune induction in small animals and humans. However, more clinical trials using EP are needed to verify the utility of EP-delivered DNA vaccines in treating cancer patients.

Strategies to improve immunogenicity

As mentioned before, cancer antigens are generally poorly immunogenic and require special strategies to increase their immunogenicity. To date, various approaches such as DNA manipulation and the use of xenogeneic antigens, the use of immune response regulators, the co-injection of cytokines and other molecules, the use of DNA prime-boost immunization strategies, and different DNA delivery methods have been mainly taken to achieve this goal. summarizes the 5 major strategies that have been employed to enhance the immunogenicity of cancer DNA vaccines.

Table 1. Immune-enhancing strategies for cancer vaccines

DNA manipulation and the use of xenogeneic antigens

Advances in molecular biological technology have made DNA construction and manipulation far easier than before. Recently, most DNA vaccines are engineered to have codon-optimized antigen genes, which allow for more antigenic protein production. In addition, the strategy of linking antigen-coding genes with other DNA sequences, the products of which are associated with immune activation or the targeting of the antigens to intracellular compartments, has also been evaluated. For instance, the endolysosomal targeting of codon-optimized E7 genes resulted in more antigenic protein production, leading to enhanced Ag-specific CTL and antitumor responses to E6/E7-expressing tumor cell (TC-1) challenges.Citation41 Conjugation of heat shock protein to HPV 16 E7 antigens also improved E7 DNA vaccine potency against TC-1 cells through augmenting Ag-specific CTL responses.Citation42 Conjugation of the IgE leader sequence to codon-optimized E6 and E7 genes also resulted in the induction of much stronger CTL responses and antitumor activity against TC-1 cell challenges.Citation43 Conjugation of the CD4+ T helper cell epitope domain to CTL epitope sequences has also been tested in humans for increasing CTL responses.Citation44,45 Linking E7 DNA sequences to the genes coding for lysosome-associated membrane protein 1-targeting peptides (for endolysosomal targeting), bacterial toxin (for cellular targeting), calreticulin (for endoplasmic reticulum targeting), viral protein 22 (for intracellular antigen spreading), γ-tubulin (for centrosomal compartment targeting), and heat shock protein (HSP)70 have been demonstrated to be effective at inducing greater antitumor CTL and antitumor responses (reviewed in Ref. Citation23). These preclinical studies demonstrated that antigen codon optimization, CD4+ T helper cell epitope domain conjugation, and antigen targeting to specific intracellular compartments might be promising strategies to augment cancer Ag-specific CTL responses and antitumor activity.

In the case of non-mutated self-antigens, use of a xenogeneic antigen has been thought to be another promising approach to overcome immunological tolerance to self-antigens. Because xenogeneic antigens are an altered self-protein bearing amino acid substitutions in one or more epitopes, these antigens might overcome immune tolerance and be recognized by T cells, which can then be further activated for the induction of immune responses to both the xenogeneic antigens and the native antigens. The use of homologous proteins from a different species, called xenogeneic antigens, as an immunogen was well demonstrated with melanoma differentiation antigens, such as gp100 and tyrosinase.Citation46-48 For example, DNA vaccines expressing human gp100 but not murine gp100 induced cross-reactive and anti-melanoma protective CD8+ T cell responses in a murine model.Citation46 In a canine melanoma treatment, DNA vaccines encoding human tyrosinase showed a positive effect on the induction of cross-reactive antibodies against canine and human tyrosinases and increased the mean survival time.Citation47,48 These animal studies suggest that use of a xenogeneic antigen may be an option to overcome natural immunological tolerance to cancer-associated self-antigens.

Use of regulators of immune responses

The fate of T cells is determined by 2 types of molecules expressed on T cell surface, which act as regulators of immune responses.Citation49,50 One type comprises co-stimulatory molecules, including 4-1BB, GITR [glucocorticoid-induced tumor necrosis factor receptor family-related gene], OX40, ICOS [inducible T cell co-stimulator], CD27, and CD30, whereas the other comprises co-inhibitory molecules including CTLA-4 [cytotoxic T lymphocyte-associated antigen-4], PD [programmed death], and B and T lymphocyte attenuator. To date, numerous animal and clinical studies have been performed to test the ability of these molecules to regulate the efficacy of cancer therapy. For example, we reported using 2 separate animal tumor models that the combined delivery of tumor vaccines with agonistic anti-4-1BB Abs results in the dramatic enhancement of antitumor therapeutic activity by increasing Ag-specific CTL activity.Citation51,52 DC-based tumor vaccines, in combination with both anti-OX-40 and anti-4-1BB Abs, were also effective at enhancing antitumor activity in a HER2/neu transgenic animal model.Citation53 The combined injection of DNA vaccines coding for gp100 and Trp2 with anti-GITR Abs increased protection against a lethal challenge with B16 melanoma and induced memory CD8+ T cell responses, even in the absence of CD4+ T cells.Citation54 In contrast, blocking CTLA-4 using antagonistic anti-CTLA-4 Abs is associated with increased antitumor T cell responses in mice.Citation55,56 Trp2- and gp100-encoding DNA vaccines in combination with anti-CTLA-4 Abs also displayed enhanced Ag-specific T cell responses and anti-melanoma activity in a B16 model.Citation57 In a clinical trial, anti-CTLA-4 Abs improved overall survival in patients with previously treated metastatic melanoma.Citation58 It has recently been reported that anti-CTLA-4 Abs play a more important role in broadening tumor-reactive CD8+ T cell responses rather than increasing tumor-specific T cell reactivity,Citation59 suggesting that anti-CTLA-4 Abs are mainly associated with Ag-specific T cell activation. Taken together, these animal and clinical results strongly support the notion that the immunogenicity of cancer DNA vaccines could be modulated by combining them with the regulators of immune responses. Presently, the anti-CTLA-4 blocking Ab, Ipilimumab has been approved by the US FDA for the treatment of late stage melanoma.

Co-injection of cytokines and other molecules

The co-injection of DNA vaccines with molecular adjuvants encoding cytokines, chemokines and other immune stimulatory molecules has been known to influence the immunogenicity of DNA vaccines. Previously, our group demonstrated using a herpes simplex virus (HSV)-2 gD DNA vaccine model that the naked injection of gD DNA vaccines along with plasmid DNA encoding Th0-, Th1-, and Th2-type cytokines, chemokines and co-stimulatory molecules can modulate Ag-specific antibody and cellular responses, and enhance protective immunity against HSV-2 challenges (reviewed in Ref. Citation23). These data suggest that these immune stimulatory molecules could be useful as cancer vaccine adjuvants for enhancing Ag-specific CTL responses, which are required for cancer eradication. In this context, the co-injection of prostate-specific antigen (PSA) DNA vaccines with IL-2 and GM-CSF cDNA augments Ag-specific CTL responses and antitumor protection from a challenge with PSA-expressing tumor cells.Citation60 Our data has also shown that IM-EP of HPV E7 DNA vaccines plus IL-2 cDNA is able to enhance antitumor therapeutic activity by increasing the Ag-specific CTL activity in a TC-1 model.Citation52 IM injection of carcinoembryonic antigen (CEA) DNA vaccines plus IL-12 expression plasmids resulted in enhanced antitumor effectiveness in an animal tumor model.Citation61 Contrary to this, we observed that IM-EP of E7 DNA vaccines plus IL-12 plasmid suppressed Ag-specific CTL responses and antitumor activity against TC-1 cells, suggesting that a high level of IL-12 expressed by EP might be detrimental to immune induction.Citation62 However, direct EP of IL-12 cDNA alone into tumor sites is also capable of eradicating established B16 melanoma and MC32 colon cancer by inducing tumor Ag-specific CTL responses.Citation39,63 In a phase I clinical trial including 24 metastatic melanoma patients, intratumoral EP of IL-12 cDNA also showed minimal systemic toxicity.Citation38 In that study, 2 (10%) of the 19 patients with non-electroporated distant lesions and no other systemic therapy showed complete regression of all metastases, whereas 8 patients (42%) displayed disease stabilization or a partial response, suggesting that IL-12 alone can also be useful for achieving cancer control. In addition, the co-injection of HPV E7 DNA vaccines with plasmid DNA encoding anti-apoptotic molecules and serine protease inhibitors increased Ag-specific CTL responses and antitumor activity against TC-1 cells through increased DC functions.Citation64,65 In a phase I/II clinical study, a different dose of GM-CSF DNA was used as the adjuvant of a multipeptide vaccine (gp100 and tyrosinase) in the treatment of 19 melanoma patients (stage III/IV), and 8 patients (42%) developed T cell responses without any GM-CSF-related side effects.Citation66 In a phase I/II clinical trial of 12 patients with follicular B-cell lymphoma in remission after chemotherapy, the delivery of naked DNA vaccines expressing chimeric immunoglobulin plus GM-CSF also demonstrated both the induction of Ag-specific T cell responses and the safety of GM-CSF as an adjuvant.Citation67 Taken together, co-injection approaches using immune-stimulatory molecules and regulators of DC functions likely influence the magnitude and the duration of cancer DNA vaccines in their induction of cancer-reactive CTL responses.

DNA prime-boost immunization strategies

DNA prime-boost immunization appears to be a promising strategy as a DNA vaccine regimen. This prime-boost approach has been shown to be promising in generating greater immune responses compared with DNA vaccination alone. For instance, IM-EP of CEA DNA vaccines followed by boosting with adenovirus encoding CEA induced the greatest level of Ag-specific CD4+ and CD8 + T cell responses in wild type mice, and the repeated injection of this prime-boost scheme induced Ag-specific CD8+ T cell responses in CEA transgenic mice,Citation68 suggesting that a prime-boost strategy might break through self-tolerance to engender robust immune responses. A prime-boost strategy using either a CEA DNA vaccine fused at the C-terminal end to tetanus toxin fragment C or an adenovirus expressing the fused CEA/toxin C was also more immunogenic than that using the normal CEA counterparts.Citation69 Furthermore, CEA DNA vaccine/adenovirus prime-boost schemes were safe and immunogenic in monkeys.Citation19 However, a recent clinical trial using HER2/CEA DNA vaccine/adenovirus prime-boosting strategy failed to show any measurable T cell responses to the 2 tested antigens.Citation70

Use of different DNA delivery methods

To improve the potency of cancer DNA vaccines, many different delivery methods besides aforementioned gen gun delivery and electroporation have also been tested. For instance, jet injector delivery method using HER2/neu DNA vaccines was compared with gene gun delivery, demonstrating that gene gun was superior to jet injector in eliciting Ag-specific CTL responses and antitumor protective immunity.Citation25 Similarly, tattooing delivery method using tetanus toxin fragment C-conjugated MART-1 DNA vaccines has been found to be highly immunogenic in humans.Citation24 Furthermore, direct intranodal delivery of DNA vaccines was also tested for its efficacy.Citation20,21 DNA vaccines were also formulated with chemical compounds (such as polymers and nanoparticles) and then tested for their utility. For instance, liposome-DNA vaccine complex was more effective at increasing CD8+ T cell numbers and controlling established tumors through the production of IL-12 from dendritic cells and of IFN-γ from NK cells, as compared with DNA vaccines alone.Citation71 Similarly, higher anti-melanoma immune responses were elicited by formulation of DNA vaccines with nanoparticle, as compared with DNA vaccines alone.Citation72 Thus, the use of these formulations and newer delivery strategies likely hold promise for augmenting the potency of cancer DNA vaccines.

Cancer antigens

To date, scientists have identified a number of cancer antigens. In general, cancer antigens are classified as altered self-antigens, cancer/testis antigens, cancer over-expressed antigens, cancer differentiation antigens, and cancer-specific antigens. The typical examples of these antigens are as follows: cancer overexpressed antigens: survivin, human telomerase reverse transcriptase (hTERT), CEA, HER2/neu, α-fetoprotein (AFP), Wilm's tumor (WT1) antigens, PSA, prostate-specific membrane antigen (PSMA), prostatic acid phosphatase (PAP); cancer/testis antigens: melanoma-associated antigen (MAGE), NY-ESO-1, preferentially expressed antigen in melanoma (PRAME); differentiation antigens: melanoma antigen recognized by T cells (MART)-1, gp100, tyrosinase, idiotype immunoglobulin; altered self-antigens: mutated p53; cancer-specific antigens: HPV E6 and E7. As most of these cancer antigens are derived from self-proteins, they are often weak antigens. However, these antigens are recognized by T cells, such that Ag-specific T cells are likely induced and further activated by applying various strategies. A list of cancer DNA vaccines tested in humans (based on our PubMed search) is shown in .

Table 2. DNA vaccines tested in clinical trials and the results

Cancer over-expressed antigens

Normal cellular proteins such as survivin (a member of the inhibitor of apoptosis protein family) and hTERT are overexpressed in some major types of cancer.Citation73-75 In a murine melanoma model, priming with survivin DNA vaccines plus IL-2 cDNA and boosting with rAd were effective at inducing antitumor effects.Citation76 hTERT DNA vaccines were also able to induce Ag-specific CD8+ T cell and protective antitumor responses to Ag-expressing tumor cells.Citation77,78 These animal data show that survivin and hTERT might be used to induce Ag-specific CTL responses for the eradication of malignancies expressing survivin and hTERT.

CEA is over-expressed in colorectal cancer,Citation79,80 whereas HER2/neu is highly expressed in breast cancer.Citation81 In an animal study, the co-injection of CEA DNA vaccines with IL-12 cDNA augmented antitumor resistance to CEA-expressing tumor cells in a CD8+ T cell- and perforin-dependent manner.Citation61,82 A CEA DNA priming and adenovirus boosting strategy also increased antitumor protective responses to CEA-expressing tumors.Citation68 DNA vaccines targeting 2 antigens, CEA and HER2/neu were able to induce Ag-specific immune responses by breaking immunological tolerance in CEA/HER2 double-transgenic mice and NOD/scid-DR1 mice, leading to the inhibition of CEA- and HER2/neu-expressing tumor growth.Citation83 However, prime-boost immunization strategies using HER2/CEA DNA vaccines and HER2/CEA-expressing adenovirus failed to induce measurable cellular responses to the 2 tested antigens in humans,Citation70 underscoring the importance of breaking immune tolerance prior to inducing Ag-specific immune responses.

AFP is an oncofetal protein which is overexpressed by many hepatocellular cancers.Citation84 AFP+GM-CSF DNA prime-adenovirus (expressing AFP) boost vaccines were able to induce Ag-specific Th1 type and antitumor protective responses in mice.Citation85 In a clinical trial, AFP DNA prime-adenovirus boost vaccines also induced Ag-specific T cell responses in 2 hepatocellular patients, one of which showed higher Ag-specific T cell responses and more favorable progression-free survival,Citation86 suggesting that AFP might be an antigen with potential for hepatocellular cancer immunotherapy.

The WT1 antigen is a transcription factor that is overexpressed both in leukemia cells and in some solid tumors, including lung cancer and colon cancer.Citation87–89 In humanized transgenic mice, DNA vaccines encoding WT1-derived epitopes, in particular WT137–45, were able to elicit Ag-specific CTL responses that recognized leukemia cells. The expanded human CTLs were also able to kill Ag-expressing human target cells in vitro,Citation90 suggesting a potential use for WT1 in therapeutic immunization against leukemia and other cancers expressing WT1.

PSA is a serum marker that is widely used in the detection and monitoring of prostate cancer.Citation91,92 PSMA and PAP are also known to be over-expressed in prostate cancer.Citation93,94 In a murine model, ID injection of PSA DNA vaccines resulted in the induction of potent Ag-specific antibody and cellular responses, as well as of antitumor resistance to Ag-expressing tumors.Citation95 Furthermore, IM-EP of DNA vaccines expressing PSA and PSMA as a dual antigen approach induced potent Ag-specific antibody and cellular responses to the 2 tested antigens in animals.Citation96 In patients with advanced prostate cancer, IM and ID delivery of 900 μg PSA DNA vaccines in combination with subcutaneous delivery of GM-CSF and IL-2 induced Ag-specific humoral and cellular responses.Citation97 In 41% of the treated prostate cancer patients, the ID injection of PAP DNA vaccines plus GM-CSF cDNA displayed Ag-specific CD4+/CD8+ T cell proliferative responses and the increased PSA doubling time.Citation98 Moreover, the booster ID injections of PAP DNA vaccines plus GM-CSF cDNA to the immunized patients have been shown to induce cytolytic T cell responses in 58% patients.Citation99 Furthermore, in patients with recurrent prostate cancer, IM-EP of DNA vaccines expressing the tetanus toxin DOMmain-conjugated PSMA CTL epitopes showed anti-DOM IgG production and CD4+ T cell responses, as well as PSMA-specific CD8+ T cell responses with the increased PSA doubling time.Citation44,45 In patients with Ag-positive solid tumors, PRAME+PSMA fragment DNA prime-peptide boost vaccines (by intranodal delivery) induced Ag-specific T cell responses in 63 melanoma patients and showed stable disease in 7 patients, but no partial or complete responses.Citation21 In that study, some patients showed decreased PSA levels. These promising data warrant further evaluation of IM-EP and intranodal delivery of PSA, PSMA and PAP DNA vaccines in treating patients with prostate cancer.

Cancer/testis tumor-associated antigens

Cancer/testis antigens, such as MAGE and NY-ESO-1 are expressed in many cancer cells but, among the normal tissues, are solely expressed in the testes.Citation100 In a murine model, DNA vaccines encoding HSP70/MAGE-3 fusion proteins were immunogenic and conferred potent antitumor immunity against MAGE-3-expressing tumors.Citation101 Particle-mediated ID delivery of NY-ESO-1 DNA vaccines into 16 patients with advanced cancer [diagnosed as prostate adenocarcinoma (10 patients), non-small cell lung cancer (NSCLC; 5 patients) or esophageal carcinoma (1 patient)] also led to the induction of Ag-specific CD4 and/or CD8 T cell responses without any clinical benefits in most of the tested patients.Citation102 In view of this finding, an association between NY-ESO-1-specific immune responses and clinical responses remains to be determined.

Cancer differentiation antigens

The proteins MART-1 (also called Melan-A), gp100 and Trp2 are differentiation antigens that are mainly expressed in melanoma cells and normal melanocytes.Citation103-105 DNA vaccines expressing human gp100 and Trp2 have shown antitumor protective and therapeutic activities against B16 melanoma in mice,Citation106 suggesting that a xenogeneic antigen might be effective at inducing cross-reactive antitumor responses to self-antigen-expressing tumor cells. Consistent with this finding, human Trp2-encoding DNA vaccines provided significant protection against the metastatic growth of B16 melanoma in mice through the induction of cross-reactive antibody and CD8+ T cell responses.Citation107 Intradermal and intramuscular delivery of gp100 failed to show any clinical and immune responses in melanoma patients.Citation108 Particle-mediated epidermal delivery of cDNA coding for gp100 and GM-CSG to uninvolved skin of melanoma patients resulted in modest activation of anti-melanoma responses while showing its safety and tolerability.Citation109 However, the intranodal delivery of tyrosinase epitope DNA vaccines induced Ag-specific immune responses in 42% patients without any clinical responses while overall survival was in favor of immune responders.Citation110 In patients with stage IV melanoma, moreover, the intranodal delivery of MART-1-expressing DNA vaccines resulted in induction of Ag-specific immune responses without any clinical benefits.Citation20

In the treatment of hematological malignancies, the induction of long-term memory immune responses is likely critical for preventing cancer relapse. Idiotype immunoglobulin has also been identified to have unique protein sequences in the variable regions of B cell immunoglobulin.Citation111,112 Thus, the idiotype of B-cell malignancy might serve as a tumor-specific antigen. Immunization with DNA vaccines encoding chemokine-fused short idiotype single-chain (sFv) polypeptides containing only the antigenic variable regions of light and heavy chains resulted in the induction of Ag-specific immune responses and antitumor protective effects in a T cell-dependent manner in mice.Citation113 When intramuscularly injected into patients with follicular B-cell lymphoma, DNA vaccines encoding chimeric idiotypes were also immunogenic.Citation67 In this context, differentiation antigens represent promising targets that can be employed to induce long-term tumor-specific CTL immunity for controlling the relapse of hematological malignancy.

A mutated antigen, p53

The p53 protein plays a role in cell cycle regulation, genomic stability, angiogenesis and apoptosis.Citation114,115 In cancer cells, p53 is often mutated,Citation116 and is thus thought to be a potential target for cancer immunotherapy. In an animal model, IM-EP of a DNA vaccine encoding mutated human p53 displayed mouse p53 cross-reactive CD8+ T cell responses and antitumor protective and therapeutic activities in a CD8+ T cell-dependent manner,Citation117 suggesting that mutated p53 might also represent a useful target for cancer immune therapy.

Cancer-specific antigens (HPV E6 and E7)

HPV infection is a major cause of oral cancer, as well as anogenital cancers, including cervical cancer (reviewed in Ref. Citation118). HPV E6 and E7 proteins are known to be associated with oncogenesis through the inhibition of the functions of p53 and Rb, which are cell cycle regulatory proteins.Citation119,120 In this context, HPV E6 and E7 proteins have been tested as immunization targets for HPV-associated cancers. In numerous animal studies, DNA vaccines encoding HPV E6 and E7 cDNAs have shown to induce robust antigen-specific antibody and cellular responses.Citation41-43,121,122 A clinical trial using IM-EP of HPV E6/E7 DNA vaccines showed that these vaccines were able to induce robust Ag-specific humoral and cellular responses (CTL, in particular).Citation34 A recent clinical trial further demonstrated the clinical efficacy of IM-EP of HPV E6/E7 DNA vaccines in reducing the viral load and precancerous disease severity (personal communication with Bagarazzi M). Therefore, it is likely that HPV E6 and E7 DNA vaccines might be applicable in treating HPV-associated oral and anogenital cancers, as well as in treating precancerous diseases.

Tumor immune evasion

Because cancer cells are somewhat different from their normal counterparts, these cells can possibly be recognized and eradicated by the immune system. However, cancer cells are heterogeneous and employ many different mechanisms to evade host immune surveillance (reviewed inCitation12). Clarification of the immune escape mechanism(s) utilized by cancer cells appears to be important for developing counter-measures for this immune escape. summarizes the immune evasion mechanism(s) employed by cancer cells and the cancer microenvironment.

Table 3. Immune evasion mechanisms employed by tumor cells and the tumor microenvironment

Tumor cells release TGF-β, which acts on tumor Ag-specific CTLs and inactivates the functions of perforin, granzyme B, Fas ligand, and interferon-γ, which are the effector molecules of CTLs for tumor cell killing.Citation123 Tumor cells also express indoleamine 2,3-dioxygenase, which exploits the tryptophan pool and induces tryptophan deficiency to prevent T cell proliferation.Citation124,125 In addition to these immune inhibitory molecules, tumor cells utilize immune-inhibitory ligands on the cell surface as a mechanism to resist CTL-mediated tumor cell destruction. Cancer cell-associated B7-H1 (also called PD-L1) induces the apoptosis of cancer-specific T cells in vitro and promotes the growth of tumor cells in animals.Citation126 The infusion of anti-B7-H1 Abs to block B7-H1 resulted in the increased efficacy of adoptive T cell therapy for curing mice with tumors.Citation127 Furthermore, in PD-1 deficient mice, tumor growth was suppressed in a manner similar to that of the application of anti-PD-L1 Abs,Citation128 supporting a dominant role for PD-1/PD-L1 in local immune suppression within the tumor microenvironment. These data suggest that PD-L1 expressed on tumor cells might contribute to the suppression of tumor-reactive CTLs. In patients with advanced cancers, including non-small-cell lung cancer, melanoma, and renal-cell cancer, blocking PD-L1 using anti-PD-L1 Abs induced sustained tumor regression and prolonged disease stabilization.Citation129

The mutation of p53 was suggested as a possible mechanism for the insensitivity of tumor cells to tumor-reactive CTLs.Citation130 In that study, the restoration of mutant p53 function by infecting the cells with adenovirus expressing wild type p53 resulted in increased sensitivity to CTL-mediated lysis by the Fas-mediated pathway. The serine protease inhibitor PI-9/SPI-6 (inactivator of granzyme B) is also associated with tumor cell insensitivity to Ag-specific CTL-mediated killing by blocking granzyme B proteins.Citation131 cFLIP over-expression is also responsible for tumor cell resistance to CTL-mediated lysis by blocking the death receptor-dependent tumor cell lysis pathway.Citation132 Moreover, tumor cells do not present antigens on the surface of tumor cells in the context of MHC class I antigens and thus remaining unrecognizable by tumor-reactive CTLs.Citation133 Consistent with this, we recently reported that MC32 colon cancer cells could evade CEA-specific CTLs by the loss of antigen expression on the tumor cell surface in an MC32 colon cancer model.Citation63 In addition, tumor cells lose an antigen for CTL recognition.Citation134 We also found that MC32 cells remain without antigen expression under severe immune selection conditions, thereby evading Ag-specific CTLs.Citation135 The down-regulation of MHC class I molecules is also a frequently observed mechanism for immune escape in cancer cells.Citation136 Furthermore, the overexpression of anti-apoptotic molecules causes tumor cells to become insensitive to CTL-mediated tumor cell killing.Citation137 Thus, reversing tumor cell-driven immune dysfunction is required to achieve CTL-mediated tumor destruction.

The tumor microenvironment often contains immunosuppressive cells that secrete immunosuppressive molecules and limit the functions of CTLs. For instance, tumor-associated macrophages promote tumor cell growth and metastasis and inhibit effector T cell activity, whereas removing this cell population enhances anti-cancer therapy (reviewed in Ref. Citation138). Regulatory T cells are also involved in inhibiting T cell function by many different strategies, but the suppression of these cell populations results in improved vaccine immunogenicity (reviewed in Ref. Citation139). Myeloid-derived stem cells are also known to suppress T cell responses and to induce tolerance to cancer-associated antigens (reviewed in Ref. Citation140). Blocking or removing these negative regulatory immune cells is one additional potential method to improve the immunogenicity of cancer DNA vaccines and the reactivity of Ag-specific CTLs to cancer cells. Moreover, if one antigen is lost or an antigen epitope presentation is altered during long-term immune therapy, it might be useful to use more than one cancer antigen as a pre-measure to the loss of one antigen. Combined therapy using cancer DNA vaccines and other therapy protocols with different antitumor killing mechanisms might also be required for obtaining better clinical efficacy of cancer DNA vaccines. This is based upon our previous findings that cisplatin-based chemotherapy and E7 vaccination synergized antitumor therapeutic activity by rendering tumor cells more sensitive to Ag-specific CTL-mediated killing and by eliciting long-term memory responses for preventing tumor recurrence in an animal model.Citation141 A similar finding was obtained with radiotherapy plus E7 cancer vaccination,Citation142 as well as chemotherapy plus adoptive therapy using Ag-specific CTL effector cells.Citation143 In addition, during treatment with cancer DNA vaccines, blocking or inhibiting the negative molecules such as TGF-β, indoleamine 2,3-dioxygenase, PD-L1, SPI-6, cFLIP, anti-apoptotic molecules and others involved in cancer cell immune escape likely plays an important role in augmenting cancer antigen-specific CTL and anti-neoplastic responses. Taken together, these multimodal treatment approaches are promising to limit the escape of tumor cells and simultaneously achieve cancer eradication.

Conclusion

During the last 2 decades, the possibility of using cancer DNA vaccines as a means to induce therapeutically robust cancer-specific CTL responses has been intensively evaluated. However, the clinical benefits of such vaccines in treating cancer patients have yet to be demonstrated. This lack of clinical benefits is likely attributed to a number of biological factors. For instance, cancer antigens are generally of low immunogenicity due to self-tolerance, and cancer cells employ different mechanisms to escape host immune surveillance. The applications of immune-enhancing strategies in cancer DNA vaccination might help to overcome immune tolerance and to induce robust cancer-reactive immune responses, which are required for cancer eradication. At the same time, blocking or removing negative immune cells and molecules likely leads to the development of more effective anticancer DNA vaccines for treating patients with cancers. Moreover, cancer DNA vaccines have been tested on subjects with very late stage cancer; this might be the reason why the vaccines have not exhibited any cancer-eradicating effects.

Combined therapy using cancer DNA vaccines and other strategies that limit the emergence of immune-evading cancer cells and simultaneously induce robust cancer-specific CTL responses is expected to be of benefit to cancer patients.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Funding

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF) funded by the Ministry of Education, Science and Technology (2012R1A1A2038430).

References

  • Kuhober A, Pudollek HP, Reifenberg K, Chisari FV, Schlicht HJ, Reimann J, Schirmbeck R. DNA immunization induces antibody and cytotoxic T cell responses to hepatitis B core antigen in H-2b mice. J Immunol 1996; 156:3687-95; PMID:8621903
  • Wang B, Ugen KE, Srikantan V, Agadjanyan MG, Dang K, Refaeli Y, Sato A, Boyer JD, Williams WV, Weiner DB. Gene inoculation generates immune responses against human immunodeficiency virus type 1. Proc Natl Acad Sci USA 1993; 90:4156-60; PMID:8483929; http://dx.doi.org/10.1073/pnas.90.9.4156
  • Corr M, Lee DJ, Carson DA, Tighe H. Gene vaccination with naked plasmid DNA: mechanism of CTL priming. J Exp Med 1996; 184:1555-60; PMID:8879229; http://dx.doi.org/10.1084/jem.184.4.1555
  • Casares S, Inaba K, Brumeanu TD, Steinman RM, Bona CA. Anitigen presentation by dendritic cells after immunization with DNA encoding a major histocompatibility complex class II-restricted viral epitope. J Exp Med 1997; 186:1481-6; PMID:9348305; http://dx.doi.org/10.1084/jem.186.9.1481
  • Chattergoon MA, Robinson TM, Boyer JD, Weiner DB. Specific immune induction following DNA-based immunization through in vivo transfection and activation of macrophages. J Immunol 1998; 160:5707-18; PMID:9637479
  • Fu T, Ulmer JB, Caulfield MJ, Deck RR, Friedman A, Wang S, Liu X, Donnelly JJ, Liu MA. Priming of cytotoxic T lymphocytes by DNA vaccines: requirement for professional antigen presenting cells and evidence for antigen transfer from myocytes. Mol Med 1997; 3:362-71; PMID:9234241
  • Iwasaki A, Torres CA, Ohashi PS, Robinson HL, Barber BH. The dominant role of bone marrow-derived cells in CTL induction following plasmid DNA immunization at different sites. J Immunol 1997; 159:11-4; PMID:9200432
  • Condon C, Watkins S, Celluzzi CM, Thompson K, Faldo L Jr. DNA-based immunization by in vivo transfection of dendritic cells. Nature Med 1996; 2:1122-8; PMID:8837611; http://dx.doi.org/10.1038/nm1096-1122
  • Liu M, Ulmer JB. Human clinical trials of plasmid DNA vaccines. Adv Genet 2005; 55:25-40; PMID:16291211; http://dx.doi.org/10.1016/S0065-2660(05)55002-8
  • Atkinson EA, Bleackley RC. Mechanisms of lysis by cytotoxic T cells. Crit Rev Immunol 1995; 15:359-84; PMID:8834456; http://dx.doi.org/10.1615/CritRevImmunol.v15.i3-4.90
  • Groscurth P, Filgueira L. Killing mechanisms of cytotoxic T lymphocytes. News Physiol Sci 1998; 13:17-21; PMID:11390753
  • Igney FH, Krammer PH. Immune escape of tumors: apoptosis resistance and tumor counterattack. J Leukoc Biol 2002; 71:907-20; PMID:12050175
  • Wolff JA, Malone RW, Williams P, Chong W, Acsadi G, Jani A, Felgner PL. Direct gene transfer into mouse muscle in vivo. Science 1990; 247:1465-8; PMID:1690918; http://dx.doi.org/10.1126/science.1690918
  • Ulmer JB, Donnelly J, Parker SE, Rhodes GH, Felgner PL, Dwarki VL, Gromkowski SH, Deck R, DeVitt CM, Friedman A, et al. Heterologous protection against influenza by injection of DNA encoding a viral protein. Science 1993; 259:1745-9; PMID:8456302; http://dx.doi.org/10.1126/science.8456302
  • Raz E, Carson DA, Parker SE, Parr TB, Abai AM, Aichinger G, Gromkowski SH, Singh M, Lew D, Yankauckas MA, et al. Intradermal gene immunization: the possible role of DNA uptake in the induction of cellular immunity to viruses. Proc Natl Acad Sci U S A 1994; 91:9519-23; PMID:7937799; http://dx.doi.org/10.1073/pnas.91.20.9519
  • MacGregor RR, Boyer JD, Ugen KE, Lacy KE, Gluckman SJ, Bagarazzi ML, Chattergoon MA, Baine Y, Higgins TJ, Ciccarelli RB, et al. First human trial of a DNA-based vaccine for treatment of human immunodeficiency virus-1 infection: safety and host response. J Infect Dis 1998; 178:92-100; PMID:9652427; http://dx.doi.org/10.1086/515613
  • Wang R, Doolan DL, Le TP, Hedstrom RC, Coonan KM, Charoenvit Y, Jones TR, Hobart P, Margalith M, Ng J, et al. Induction of antigen-specific cytotoxic T lymphocytes in humans by a malaria DNA vaccine. Science 1998; 282:476-80; PMID:9774275; http://dx.doi.org/10.1126/science.282.5388.476
  • Trimble CL, Peng S, Kos F, Gravitt P, Viscidi R, Sugar E, Pardoll D, Wu TC. A phase I trial of a human papillomavirus DNA vaccine for HPV16+ cervical intraepithelial neoplasia 2/3. Clin Cancer Res 2009; 15:361-7; PMID:19118066; http://dx.doi.org/10.1158/1078-0432.CCR-08-1725
  • Aurisicchio L, Mennuni C, Giannetti P, Calvaruso F, Nuzzo M, Cipriani B, Palombo F, Monaci P, Ciliberto G, La Monica N. Immunogenicity and safety of a DNA prime/adenovirus boost vaccine against rhesus CEA in nonhuman primates. Int J Cancer 2007; 120:2290-300; PMID:17304509; http://dx.doi.org/10.1002/ijc.22555
  • Weber J, Boswell W, Smith J, Hersh E, Snively J, Diaz M, Miles S, Liu X, Obrocea M, Qiu Z, 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; PMID:18481391; http://dx.doi.org/10.1097/CJI.0b013e3181611420
  • Weber J, Vogelzang NJ, Ernstoff MS, Goodman OB, Cranmer LD, Marshall JL, Miles S, Rosario D, Diamond DC, Qiu Z, et al. A phase 1 study of a vaccine targeting preferentially expressed antigen in melanoma and prostate-specific membrane antigen in patients with advanced solid tumors. J Immunother 2011; 34:556-67; PMID:21760528; http://dx.doi.org/10.1097/CJI.0b013e3182280db1
  • Greenland J, Letvin NL. Chemical adjuvants for plasmid DNA vaccines. Vaccine 2007; 25:3731-41; PMID:17350735; http://dx.doi.org/10.1016/j.vaccine.2007.01.120
  • Han DJ, Weiner DB, Sin JI. DNA vaccines against infectious diseases and cancer. Biomol Ther 2010; 18:1-15; http://dx.doi.org/10.4062/biomolther.2010.18.1.001
  • Quaak S, van den Berg JH, Toebes M, Schumacher TN, Haanen JB, Beijnen JH, Nuijen B. GMP production of pDERMATT for vaccination against melanoma in a phase I clinical trial. Eur J Pharm Biopharm 2008; 70:429-38; PMID:18606527; http://dx.doi.org/10.1016/j.ejpb.2008.05.002
  • 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; PMID:23264900; http://dx.doi.org/10.4161/onci.22563
  • Trimble C, Lin CT, Hung CF, Pai S, Juang J, He L, Gillison M, Pardoll D, Wu L, Wu TC. Comparison of the CD8+ T cell responses and antitumor effects generated by DNA vaccine administered through gene gun, biojector, and syringe. Vaccine 2003; 21:4036-42; PMID:12922140; http://dx.doi.org/10.1016/S0264-410X(03)00275-5
  • Porgador A, Irvine KR, Iwasaki A, Barber BH, Restifo NP, Germain RN. Predominant role for directly transfected dendritic cells in antigen presentation to CD8+ T cells after gene gun immunization. J Exp Med 1998; 188:1075-82; PMID:9743526; http://dx.doi.org/10.1084/jem.188.6.1075
  • Best S, Peng S, Juang CM, Hung CF, Hannaman D, Saunders JR, Wu TC, Pai SI. Administration of HPV DNA vaccine via electroporation elicits the strongest CD8+ T cell immune responses compared to intramuscular injection and intradermal gene gun delivery. Vaccine 2009; 27:5450-9; PMID:19622402; http://dx.doi.org/10.1016/j.vaccine.2009.07.005
  • Aihara H, Miyazaki J. Gene transfer into muscle by electroporation in vivo. Nat Biotechnol 1998; 16:867-70; PMID:9743122; http://dx.doi.org/10.1038/nbt0998-867
  • Rizzuto G, Cappelletti M, Maione D, Savino R, Lazzaro D, Costa P, Mathiesen I, Cortese R, Ciliberto G, Laufer R, et al. Efficient and regulated erythropoietin production by naked DNA injection and muscle electroporation. Proc Natl Acad Sci USA 1999; 96:6417-22; PMID:10339602; http://dx.doi.org/10.1073/pnas.96.11.6417
  • Gronevik E, von Steyern FV, Kalhovde JM, Tjelle TE, Mathiesen I. Gene expression and immune response kinetics using electroporation-mediated DNA delivery to muscle. J Gene Med 2005; 7:218-27; PMID:15515140; http://dx.doi.org/10.1002/jgm.650
  • Dupuis M, Denis-Mize K, Woo C, Goldbeck C, Selby MJ, Chen M, Otten GR, Ulmer JB, Donnelly JJ, Ott G, et al. Distribution of DNA vaccines determines their immunogenicity after intramuscular injection in mice. J Immunol 2000; 165:2850-8; PMID:10946318; http://dx.doi.org/10.4049/jimmunol.165.5.2850
  • 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; http://dx.doi.org/10.4049/jimmunol.179.7.4741
  • Bagarazzi ML, Yan J, Morrow MP, Shen X, Parker RL, Lee JC, Giffear M, Pankhong P, Khan AS, Broderick KE, et al. Immunotherapy against HPV16/18 generates potent TH1 and cytotoxic cellular immune responses. Science Transl Med 2012; 4:155ra38; http://dx.doi.org/10.1126/scitranslmed.3004414
  • Kulkarni V, Rosati M, Bear J, Pilkington GR, Jalah R, Bergamaschi C, Singh AK, Alicea C, Chowdhury B, Zhang GM, et al. Comparison of intradermal and intramuscular delivery followed by in vivo electroporation of SIV Env DNA in macaques. Hum Vaccin Immunother 2013; 9:2081-94; PMID:23811579; http://dx.doi.org/10.4161/hv.25473
  • Lucas ML, Heller L, Coppola D, Heller R. IL-12 plasmid delivery by in vivo electroporation for the successful treatment of established subcutaneous B16.F10 melanoma. Mol Ther 2002; 5:668-75
  • Ugen KE, Kutzler MA, Marrero B, Westover J, Coppola D, Weiner DB, Heller R. Regression of subcutaneous B16 melanoma tumors after intratumoral delivery of an IL-15-expressing plasmid followed by in vivo electroporation. Cancer Gene Ther 2006; 13:969-74; PMID:16763607; http://dx.doi.org/10.1038/sj.cgt.7700973
  • 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; http://dx.doi.org/10.1200/JCO.2007.13.9048
  • Sin JI, Park JB, Lee IH, Park D, Choi YS, Choe J, Celis E. Intratumoral electroporation of IL-12 cDNA eradicates established melanomas by Trp2180-188-specific CD8+ CTLs in a perforin/granzyme-mediated and IFN-g dependent manner: application of Trp2180-188 peptides. Cancer Immunol Immunother 2012; 61:1671-82; PMID:22382361; http://dx.doi.org/10.1007/s00262-012-1214-8
  • Kim H, Sin JI. Electroporation driven delivery of both an IL-12 expressing plasmid and cisplatin synergizes to inhibit B16 melanoma tumor growth through an NK cell mediated tumor killing mechanism. Hum Vaccin Immunother 2012; 8:1714-21; PMID:23151450; http://dx.doi.org/10.4161/hv.22346
  • Kim MS, Sin JI. Both antigen optimization and lysosomal targeting are required for enhanced anti-tumour protective immunity in a human papillomavirus E7-expressing animal tumour model. Immunol 2005; 116:255-66; http://dx.doi.org/10.1111/j.1365-2567.2005.02219.x
  • Hsu KF, Hung CF, Cheng WF, He L, Slater LA, Ling M, Wu TC. Enhancement of suicidal DNA vaccine potency by linking Mycobacterium tuberculosis heat shock protein 70 to an antigen. Gene Ther 2001; 8:376-83; PMID:11313814; http://dx.doi.org/10.1038/sj.gt.3301408
  • Yan J, Reichenbach DK, Corbitt N, Hokey DA, Ramanathan MP, McKinney KA, Weiner DB, Sewell D. Induction of antitumor immunity in vivo following delivery of a novel HPV-16 DNA vaccine encoding an E6/E7 fusion antigen. Vaccine 2009; 27:431-40; PMID:19022315; http://dx.doi.org/10.1016/j.vaccine.2008.10.078
  • Low L, Mander A, McCann K, Dearnaley D, Tjelle T, Mathiesen I, Stevenson F, Ottensmeier CH. DNA vaccination with electroporation induces increased antibody responses in patients with prostate cancer. Hum Gene Ther 2009; 20:1269-78; PMID:19619001; http://dx.doi.org/10.1089/hum.2009.067
  • Chudley L, McCann K, Mander A, Tjelle T, Campos-Perez J, Godeseth R, Creak A, Dobbyn J, Johnson B, Bass P, et al. DNA fusion-gene vaccination in patients with prostate cancer induces high-frequency CD8(+) T-cell responses and increases PSA doubling time. Cancer Immunol Immunother 2012; 61:2161-70; PMID:22729556; http://dx.doi.org/10.1007/s00262-012-1270-0
  • Gold JS, Ferrone CR, Guevara-Patiño JA, Hawkins WG, Dyall R, Engelhorn ME, Wolchok JD, Lewis JJ, Houghton AN. A single heteroclitic epitope determines cancer immunity after xenogeneic DNA immunization against a tumor differentiation antigen. J Immunol 2003; 170:5188-94; PMID:12734366; http://dx.doi.org/10.4049/jimmunol.170.10.5188
  • Bergman PJ, Camps-Palau MA, McKnight JA, Leibman NF, Craft DM, Leung C, Liao J, Riviere I, Sadelain M, Hohenhaus AE, et al. Development of a xenogeneic DNA vaccine program for canine malignant melanoma at the Animal Medical Center. Vaccine 2006; 24:4582-5; PMID:16188351; http://dx.doi.org/10.1016/j.vaccine.2005.08.027
  • Liao J, Gregor P, Wolchok JD, Orlandi F, Craft D, Leung C, Houghton AN, Bergman PJ. Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma. Cancer Immun 2006; 6:8; PMID:16626110
  • Peggs KS, Quezada SA, Allison JP. Cancer immunotherapy: co-stimulatory agonists and co-inhibitory antagonists. Clin Exp Immunol 2009; 157:9-19; PMID:19659765; http://dx.doi.org/10.1111/j.1365-2249.2009.03912.x
  • Pentcheva-Hoang T, Corse E, Allison JP. Negative regulators of T-cell activation: potential targets for therapeutic intervention in cancer, autoimmune disease, and persistent infections. Immunol Rev 2009; 229:67-87; PMID:19426215; http://dx.doi.org/10.1111/j.1600-065X.2009.00763.x
  • Sin JI, Kim H, Ahn E, Jeon YH, Park WS, Lee SY, Kwon B. Combined stimulation of TLR9 and 4.1BB augments Trp2 peptide vaccine-mediated melanoma rejection by increasing Ag-specific CTLs and their infiltration into tumor sites. Cancer Lett 2013; 330:190-9; PMID:23219755; http://dx.doi.org/10.1016/j.canlet.2012.11.045
  • Kim H, Kwon B, Sin JI. Combined stimulation of IL-2 and 4-1BB receptors augments the antitumor activity of E7 DNA vaccines by increasing Ag-specific CTL responses. PLoS One 2013; 8:e83765; PMID:24391824; http://dx.doi.org/10.1371/journal.pone.0083765
  • Cuadros C, Dominguez AL, Lollini PL, Croft M, Mittler RS, Borgström P, Lustgarten J. Vaccination with dendritic cells pulsed with apoptotic tumors in combination with anti-OX40 and anti-4-1BB monoclonal antibodies induces T cell-mediated protective immunity in Her-2/neu transgenic mice. Int J Cancer 2005; 116:934-43; PMID:15856473; http://dx.doi.org/10.1002/ijc.21098
  • Cohen AD, Diab A, Perales MA, Wolchok JD, Rizzuto G, Merghoub T, Huggins D, Liu C, Turk MJ, Restifo NP, et al. Agonist anti-GITR antibody enhances vaccine-induced CD8(+) T-cell responses and tumor immunity. Cancer Res 2006; 66:4904-12; PMID:16651447; http://dx.doi.org/10.1158/0008-5472.CAN-05-2813
  • van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied by autoimmune depigmentation. J Exp Med 1999; 190:355-66; PMID:10430624; http://dx.doi.org/10.1084/jem.190.3.355
  • Quezada SA, Peggs KS, Curran MA, Allison JP. CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. J Clin Invest 2006; 116:1935-45; PMID:16778987; http://dx.doi.org/10.1172/JCI27745
  • Gregor PD, Wolchok JD, Ferrone CR, Buchinshky H, Guevara-Patiño JA, Perales MA, Mortazavi F, Bacich D, Heston W, Latouche JB, et al. CTLA-4 blockade in combination with xenogeneic DNA vaccines enhances T-cell responses, tumor immunity and autoimmunity to self antigens in animal and cellular model systems. Vaccine 2004; 22:1700-8; PMID:15068853; http://dx.doi.org/10.1016/j.vaccine.2003.10.048
  • Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010; 363:711-23; PMID:20525992; http://dx.doi.org/10.1056/NEJMoa1003466
  • Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, Welters MJ, van der Burg S, Kapiteijn E, Michielin O, et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 2014; 6:254ra128; http://dx.doi.org/10.1126/scitranslmed.3008918
  • Roos AK, Pavlenko M, Charo J, Egevad L, Pisa P. Induction of PSA-specific CTLs and anti-tumor immunity by a genetic prostate cancer vaccine. Prostate 2005; 62:217-23; PMID:15389792; http://dx.doi.org/10.1002/pros.20135
  • Song K, Chang Y, Prud'homme GJ. IL-12 plasmid-enhanced DNA vaccination against carcinoembryonic antigen (CEA) studied in immune-gene knockout mice. Gene Ther 2000; 7:1527-35; PMID:11021590; http://dx.doi.org/10.1038/sj.gt.3301274
  • Sin JI. Suppression of antitumor protective cytotoxic T lymphocyte responses to a human papillomavirus 16 E7 DNA vaccine by coinjection of interleukin-12 cDNA: involvement of nitric oxide in immune suppression. Immunology 2009; 128:e707-e17; PMID:19740332; http://dx.doi.org/10.1111/j.1365-2567.2009.03068.x
  • Ahn E, Kim H, Han KT, Sin JI. A loss of antitumor therapeutic activity of CEA DNA vaccines is associated with the lack of tumor cells' antigen presentation to Ag-specific CTLs in a colon cancer model. Cancer Lett 2015; 356:676-85; PMID:25449428; http://dx.doi.org/10.1016/j.canlet.2014.10.019
  • Kim TW, Hung CF, Ling M, Juang J, He L, Hardwick JM, Kumar S, Wu TC. Enhancing DNA vaccine potency by coadministration of DNA encoding antiapoptotic proteins. J Clin Invest 2003; 112:109-17; PMID:12840065; http://dx.doi.org/10.1172/JCI200317293
  • Kim TW, Hung CF, Boyd DA, He L, Lin CT, Kaiserman D, Bird PI, Wu TC. Enhancement of DNA vaccine potency by coadministration of a tumor antigen gene and DNA encoding serine protease inhibitor-6. Cancer Res 2004; 64:400-5; PMID:14729651; http://dx.doi.org/10.1158/0008-5472.CAN-03-1475
  • Perales MA, Yuan J, Powel S, Gallardo HF, Rasalan TS, Gonzalez C, Manukian G, Wang J, Zhang Y, Chapman PB, 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; PMID:18797450; http://dx.doi.org/10.1038/mt.2008.196
  • Timmerman JM, Singh G, Hermanson G, Hobart P, Czerwinski DK, Taidi B, Rajapaksa R, Caspar CB, Van Beckhoven A, Levy R. Immunogenicity of a plasmid DNA vaccine encoding chimeric idiotype in patients with B-cell lymphoma. Cancer Res 2002; 62:5845-52; PMID:12384547
  • Mennuni C, Calvaruso F, Facciabene A, Aurisicchio L, Storto M, Scarselli E, Ciliberto G, La Monica N. Efficient induction of T-cell responses to carcinoembryonic antigen by a heterologous prime-boost regimen using DNA and adenovirus vectors carrying a codon usage optimized cDNA. Int J Cancer 2005; 117:444-55; PMID:15906358; http://dx.doi.org/10.1002/ijc.21188
  • Facciabene A, Aurisicchio L, Elia L, Palombo F, Mennuni C, Ciliberto G, La Monica N. DNA and adenoviral vectors encoding carcinoembryonic antigen fused to immunoenhancing sequences augment antigen-specific immune response and confer tumor protection. Hum Gene Ther 2006; 17:81-92; PMID:16409127; http://dx.doi.org/10.1089/hum.2006.17.81
  • Diaz-Montero 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; PMID:23497415; http://dx.doi.org/10.1186/1479-5876-11-62
  • U'Ren L, Kedl R, Dow S. Vaccination with liposome-DNA complexes elicits enhanced antitumor immunity. Cancer Gene Ther 2006; 13:1033-44; PMID:16841080; http://dx.doi.org/10.1038/sj.cgt.7700982
  • Li A, Qin L, Wang W, Zhu R, Yu Y, Liu H, Wang S. The use of layered double hydroxides as DNA vaccine delivery vector for enhancement of anti-melanoma immune response. Biomaterials 2011; 32:469-77; PMID:20934217; http://dx.doi.org/10.1016/j.biomaterials.2010.08.107
  • Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer 2008; 8:61-70; PMID:18075512; http://dx.doi.org/10.1038/nrc2293
  • Kim NW, Piatyszek MA, Prowse KR, Harley CB, West MD, Ho PL, Coviello GM, Wright WE, Weinrich SL, Shay JW. Specific association of human telomerase activity with immortal cells and cancer. Science 1994; 266:2011-5; PMID:7605428; http://dx.doi.org/10.1126/science.7605428
  • Hiyama E, Hiyama K. Telomerase as tumor marker. Cancer Lett 2003; 194:221-33; PMID:12757980; http://dx.doi.org/10.1016/S0304-3835(02)00709-7
  • Zhang H, Wang Y, Liu C, Zhang L, Xia Q, Zhang Y, Wu J, Jiang C, Chen Y, Wu Y, et al. DNA and adenovirus tumor vaccine expressing truncated survivin generates specific immune responses and anti-tumor effects in a murine melanoma model. Cancer Immunol Immunother 2012; 61:1857-67; PMID:22706381; http://dx.doi.org/10.1007/s00262-012-1296-3
  • Yan J, Pankhong P, Shin TH, Obeng-Adjei N, Morrow MP, Walters JN, Khan AS, Sardesai NY, Weiner DB. Highly optimized DNA vaccine targeting human telomerase reverse transcriptase stimulates potent antitumor immunity. Cancer Immunol Res 2013; 1:179-89; PMID:24777680; http://dx.doi.org/10.1158/2326-6066.CIR-13-0001
  • Tsuboi A, Oka Y, Ogawa H, Elisseeva OA, Li H, Kawasaki K, Aozasa K, Kishimoto T, Udaka K, Sugiyama H. Cytotoxic T-lymphocyte responses elicited to Wilms' tumor gene WT1 product by DNA vaccination. J Clin Immunol 2000; 20:195-202; PMID:10941827; http://dx.doi.org/10.1023/A:1006637529995
  • Thompson J, Zimmermann W. The carcinoembryonic antigen gene family: structure, expression and evolution. Tumor Biol 1988; 9:63-83; http://dx.doi.org/10.1159/000217547
  • Thompson JA, Grunert F, Zimmermann W. Carcinoembryonic antigen gene family: molecular biology and clinical perspectives. J Clin Lab Anal 1991; 5:344-66; PMID:1941355; http://dx.doi.org/10.1002/jcla.1860050510
  • Slamon DJ, Godolphin W, Jones LA, Holt JA, Wong SG, Keith DE, Levin WJ, Stuart SG, Udove J, Ullrich A, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science 1989; 244:707-12; PMID:2470152; http://dx.doi.org/10.1126/science.2470152
  • Song K, Chang Y, Prud'homme GJ. Regulation of T-helper-1 versus T-helper-2 activity and enhancement of tumor immunity by combined DNA-based vaccination and nonviral cytokine gene transfer. Gene Ther 2000; 7:481-92; PMID:10757021; http://dx.doi.org/10.1038/sj.gt.3301123
  • Aurisicchio L, Peruzzi D, Koo G, Wei WZ, La Monica N, Ciliberto G. Immunogenicity and therapeutic efficacy of a dual-component genetic cancer vaccine cotargeting carcinoembryonic antigen and HER2/neu in preclinical models. Hum Gene Ther 2014; 25:121-31; PMID:24195644; http://dx.doi.org/10.1089/hum.2013.103
  • Ruoslahti E, Seppälä M. α-Fetoprotein in cancer and fetal development. Adv Cancer Res 1979; 29:275-346; PMID:89800; http://dx.doi.org/10.1016/S0065-230X(08)60849-0
  • Meng W, Butterfield LH, Ribas A, Dissette VB, Heller JB, Miranda GA, Glaspy JA, McBride WH, Economou JS. alpha-fetoprotein-specific tumor immunity induced by plasmid prime-adenovirus boost genetic vaccination. Cancer Res 2001; 61:8782-6; PMID:11751399
  • Butterfield L, Economou JS, Gamblin TC, Geller DA. Alpha fetoprotein DNA prime and adenovirus boost immunization of two hepatocellular cancer patients. J Transl Med 2014; 12:86; PMID:24708667; http://dx.doi.org/10.1186/1479-5876-12-86
  • Inoue K, Ogawa H, Sonoda Y, Kimura T, Sakabe H, Oka Y, Miyake S, Tamaki H, Oji Y, Yamagami T, et al. Aberrant overexpression of the Wilms tumor gene (WT1) in human leukemia. Blood 1997; 89:1405-12; PMID:9028964
  • Oji Y, Miyoshi S, Maeda H, Hayashi S, Tamaki H, Nakatsuka S, Yao M, Takahashi E, Nakano Y, Hirabayashi H, et al. Overexpression of the Wilms' tumor gene WT1 in de novo lung cancers. Int J Cancer 2002; 100:297-303; PMID:12115544; http://dx.doi.org/10.1002/ijc.10476
  • Koesters R, Linnebacher M, Coy JF, Germann A, Schwitalle Y, Findeisen P, von Knebel Doeberitz M. WT1 is a tumor-associated antigen in colon cancer that can be recognized by in vitro stimulated cytotoxic T cells. Int J Cancer 2004; 109:385-92; PMID:14961577; http://dx.doi.org/10.1002/ijc.11721
  • Chaise C, Buchan SL, Rice J, Marquet J, Rouard H, Kuentz M, Vittes GE, Molinier-Frenkel V, Farcet JP, Stauss HJ, et al. DNA vaccination induces WT1-specific T-cell responses with potential clinical relevance. Blood 2008; 112:2956-64; PMID:18502835; http://dx.doi.org/10.1182/blood-2008-02-137695
  • Wang M, Kuriyama M, Papsidero L, Loor R, Valenzyela L, Murphy G, Chu T. Prostate antigen of human cancer patients. Methods Cancer Res 1982; 19:179-97
  • Moul JW. Prostate specific antigen only progression of prostate cancer. J Urol 2000; 163:1632-42; PMID:10799151; http://dx.doi.org/10.1016/S0022-5347(05)67511-8
  • Schmittgen TD, Teske S, Vessella RL, True LD, Zakrajsek BA. Expression of prostate specific membrane antigen and three alternatively spliced variants of PSMA in prostate cancer patients. Int J Cancer 2003; 107:323-9; PMID:12949815; http://dx.doi.org/10.1002/ijc.11402
  • Gutman AB, Gutman EB. An acid phosphatase in the serum of patients with metastasizing carcinoma of the prostate gland. J Clin Invest 1938; 17; PMID:16694594; http://dx.doi.org/10.1172/JCI100974
  • Marshall D, San Mateo LR, Rudnick KA, McCarthy SG, Harris MC, McCauley C, Schantz A, Geng D, Cawood P, Snyder LA. Induction of Th1-type immunity and tumor protection with a prostate-specific antigen DNA vaccine. Cancer Immunol Immunother 2005; 54:1082-94; PMID:16047142; http://dx.doi.org/10.1007/s00262-005-0687-0
  • Ferraro B, Cisper NJ, Talbott KT, Philipson-Weiner L, Lucke CE, Khan AS, Sardesai NY, Weiner DB. Co-delivery of PSA and PSMA DNA vaccines with electroporation induces potent immune responses. Human Vaccin 2011; 7:(Suppl):120-7; http://dx.doi.org/10.4161/hv.7.0.14574
  • Pavlenko M, Roos AK, Lundqvist A, Palmborg A, Miller AM, Ozenci V, Bergman B, Egevad L, Hellström M, Kiessling R, 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
  • McNeel DG, Dunphy EJ, Davies JG, Frye TP, Johnson LE, Staab MJ, Horvath DL, Straus J, Alberti D, Marnocha R, et al. Safety and immunological efficacy of a DNA vaccine encoding prostatic acid phosphatase in patients with stage D0 prostate cancer. J Clin Oncol 2009; 27:4047-54; PMID:19636017; http://dx.doi.org/10.1200/JCO.2008.19.9968
  • Becker JT, Olson BM, Johnson LE, Davies JG, Dunphy EJ, McNeel DG. DNA vaccine encoding prostatic acid phosphatase (PAP) elicits long-term T-cell responses in patients with recurrent prostate cancer. J Immunother 2010; 33:639-47; PMID:20551832; http://dx.doi.org/10.1097/CJI.0b013e3181dda23e
  • Yoshida N, Abe H, Ohkuri T, Wakita D, Sato M, Noguchi D, Miyamoto M, Morikawa T, Kondo S, Ikeda H, et al. Expression of the MAGE-A4 and NY-ESO-1 cancer-testis antigens and T cell infiltration in non-small cell lung carcinoma and their prognostic significance. Int J Oncol 2006; 28:1089-98; PMID:16596224
  • Ma J, Sui YF, Ye J, Huang YY, Li ZS, Chen GS, Qu P, Song HP, Zhang XM. Heat shock protein 70/MAGE-3 fusion protein vaccine can enhance cellular and humoral immune responses to MAGE-3 in vivo. Cancer Immunol Immunother 2005; 54:907-14; PMID:15756604; http://dx.doi.org/10.1007/s00262-004-0660-3
  • Gnjatic S, Altorki NK, Tang DN, Tu SM, Kundra V, Ritter G, Old LJ, Logothetis CJ, Sharma P. NY-ESO-1 DNA vaccine induces T-cell responses that are suppressed by regulatory T cells. Clin Cancer Res 2009; 15:2130-9; PMID:19276258; http://dx.doi.org/10.1158/1078-0432.CCR-08-2632
  • Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Rivoltini L, Topalian SL, Miki T, Rosenberg SA. Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci U S A 1994; 91:3515-9; PMID:8170938; http://dx.doi.org/10.1073/pnas.91.9.3515
  • Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Sakaguchi K, Appella E, Yannelli JR, Adema GJ, Miki T, Rosenberg SA. Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc Natl Acad Sci U S A 1994; 91:6458-62; PMID:8022805; http://dx.doi.org/10.1073/pnas.91.14.6458
  • Brichard V, Van Pel A, Wölfel T, Wölfel C, De Plaen E, Lethé B, Coulie P, Boon T. The tyrosinase gene codes for an antigen recognized by autologous cytolytic T lymphocytes on HLA-A2 melanomas. J Exp Med 1993; 178:489-95; PMID:8340755; http://dx.doi.org/10.1084/jem.178.2.489
  • Tanaka M, Kaneda Y, Fujii S, Yamano T, Hashimoto K, Huang SK, Hoon DS. Induction of a systemic immune response by a polyvalent melanoma-associated antigen DNA vaccine for prevention and treatment of malignant melanoma. Mol Ther 2002; 5:291-9; PMID:11863419; http://dx.doi.org/10.1006/mthe.2002.0537
  • Steitz J, Brück J, Steinbrink K, Enk A, Knop J, Tüting T. Genetic immunization of mice with human tyrosinase-related protein 2: implications for the immunotherapy of melanoma. Int J Cancer 2000; 86:89-94; PMID:10728600; http://dx.doi.org/10.1002/(SICI)1097-0215(20000401)86:1%3c89::AID-IJC14%3e3.0.CO;2-I
  • Rosenberg S, Yang JC, Sherry RM, Hwu P, Topalian SL, Schwartzentruber DJ, Restifo NP, Haworth LR, Seipp CA, Freezer LJ, 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; PMID:12804135; http://dx.doi.org/10.1089/104303403765255110
  • Cassaday R, Sondel PM, King DM, Macklin MD, Gan J, Warner TF, Zuleger CL, Bridges AJ, Schalch HG, Kim KM, et al. A phase I study of immunization using particle-mediated epidermal delivery of genes for gp100 and GM-CSF into uninvolved skin of melanoma patients. Clin Cancer Res 2007; 13:540-9; PMID:17255276; http://dx.doi.org/10.1158/1078-0432.CCR-06-2039
  • Tagawa S, Lee P, Snively J, Boswell W, Ounpraseuth S, Lee S, Hickingbottom B, Smith J, Johnson D, Weber JS. Phase I study of intranodal delivery of a plasmid DNA vaccine for patients with Stage IV melanoma. Cancer 2003; 98:144-54; PMID:12833467; http://dx.doi.org/10.1002/cncr.11462
  • Sirisinha S, Eisen HN. Autoimmune-like antibodies to the ligand-binding sites of myeloma proteins. Proc Natl Acad Sci U S A 1971; 68:3130-5; PMID:4108872; http://dx.doi.org/10.1073/pnas.68.12.3130
  • Lynch RG, Graff RJ, Sirisinha S, Simms ES, Eisen HN. Myeloma proteins as tumor-specific transplantation antigens. Proc Natl Acad Sci U S A 1972; 69:1540-4; PMID:4113870; http://dx.doi.org/10.1073/pnas.69.6.1540
  • Qin H, Cha SC, Neelapu SS, Lou Y, Wei J, Liu YJ, Kwak LW. Vaccine site inflammation potentiates idiotype DNA vaccine-induced therapeutic T cell-, and not B cell-, dependent antilymphoma immunity. Blood 2009; 114:4142-9; PMID:19749091; http://dx.doi.org/10.1182/blood-2009-05-219683
  • Levine AJ. p53, the cellular gatekeeper for growth and division. Cell 1997; 88:323-31; PMID:9039259; http://dx.doi.org/10.1016/S0092-8674(00)81871-1
  • Oren M. Regulation of the p53 tumor suppressor protein. J Biol Chem 1999; 274:36031-4; PMID:10593882; http://dx.doi.org/10.1074/jbc.274.51.36031
  • Hollstein M, Shomer B, Greenblatt M, Soussi T, Hovig E, Montesano R, Harris CC. Somatic point mutations in the p53 gene of human tumors and cell lines: updated compilation. Nucleic Acids Res 1996; 24:141-6; PMID:8594564; http://dx.doi.org/10.1093/nar/24.1.141
  • Soong RS, Trieu J, Lee SY, He L, Tsai YC, Wu TC, Hung CF. Xenogeneic human p53 DNA vaccination by electroporation breaks immune tolerance to control murine tumors expressing mouse p53. PLoS One 2013; 8:e56912; PMID:23457640; http://dx.doi.org/10.1371/journal.pone.0056912
  • Han KT, Sin JI. DNA vaccines targeting human papillomavirus-associated diseases: progresses in animal and clinical studies. Clin Exp Vaccine Res 2013; 2:106-14; PMID:23858401; http://dx.doi.org/10.7774/cevr.2013.2.2.106
  • Dyson N, Howley PM, Munger K, Harlow E. The human papillomavirus-16 E7 oncoprotein is able to bind the retinoblastoma gene product. Science 1989; 243:934-7; PMID:2537532; http://dx.doi.org/10.1126/science.2537532
  • Werness BA, Levine AJ, Howley PM. Association of human papillomavirus type 16 and 18 E6 protein with p53. Science 1990; 248:76-9; PMID:2157286; http://dx.doi.org/10.1126/science.2157286
  • Cheng WF, Hung CF, Chai CY, Hsu KF, He L, Ling M, Wu TC. Tumor-specific immunity and antiangiogenesis generated by a DNA vaccine encoding calreticulin linked to a tumor antigen. J Clin Invest 2001; 108:669-78; PMID:11544272; http://dx.doi.org/10.1172/JCI200112346
  • Lee IH, Park JB, Cheong M, Choi YS, Park D, Sin JI. Antitumor therapeutic and antimetastatic activity of electroporation-delivered human papillomavirus 16 E7 DNA vaccines: a possible mechanism for enhanced tumor control. DNA Cell Biol 2011; 30:975-85; http://dx.doi.org/10.1089/dna.2011.1266
  • Thomas DA, Massagué J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005; 8:369-80; PMID:16286245; http://dx.doi.org/10.1016/j.ccr.2005.10.012
  • Mansfield AS, Heikkila PS, Vaara AT, von Smitten KA, Vakkila JM, Leidenius MH. Simultaneous Foxp3 and IDO expression is associated with sentinel lymph node metastases in breast cancer. BMC Cancer 2009; 9:231; PMID:19604349; http://dx.doi.org/10.1186/1471-2407-9-231
  • Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2, 3-dioxygenase. J Exp Med 2002; 196:459-68; PMID:12186838; http://dx.doi.org/10.1084/jem.20020121
  • Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 2002; 8:793-800; PMID:12091876; http://dx.doi.org/10.1038/nm0902-1039c
  • Strome SE, Dong HD, Tamura HH, Voss SG, Flies DB, Tamada K, Salomao D, Cheville J, Hirano F, Lin W, et al. B7-H1 blockade augments adoptive T-cell immunotherapy for squamous cell carcinoma. Cancer Res 2003; 63:6501-5; PMID:14559843
  • Iwai Y, Ishida M, Tanaka Y, Okazaki T, Honjo T, Minato N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci U S A 2002; 99:12293-7; PMID:12218188; http://dx.doi.org/10.1073/pnas.192461099
  • Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366:2455-65; PMID:22658128; http://dx.doi.org/10.1056/NEJMoa1200694
  • Thiery J, Dorothée G, Haddada H, Echchakir H, Richon C, Stancou R, Vergnon I, Benard J, Mami-Chouaib F, Chouaib S. Potentiation of a tumor cell susceptibility to autologous CTL killing by restoration of wild-type p53 function. J Immunol 2003; 170:5919-26; PMID:12794118; http://dx.doi.org/10.4049/jimmunol.170.12.5919
  • Medema JP, de Jong J, Peltenburg LT, Verdegaal EM, Gorter A, Bres SA, Franken KL, Hahne M, Albar JP, Melief CJ, et al. Blockade of the granzyme B/perforin pathway through overexpression of the serine protease inhibitor PI-9/SPI-6 constitutes a mechanism for immune escape by tumors. Proc Natl Acad Sci USA 2001; 98:11515-20; PMID:11562487; http://dx.doi.org/10.1073/pnas.201398198
  • Medema JP, de Jong J, van Hall T, Melief CJ, Offringa R. Immune escape of tumors in vivo by expression of cellular FLICE-inhibitory protein. J Exp Med 1999; 190:1033-8; PMID:10510093; http://dx.doi.org/10.1084/jem.190.7.1033
  • Bai XF, Liu J, Li O, Zheng P, Liu Y. Antigenic drift as a mechanism for tumor evasion of destruction by cytolytic T lymphocytes. J Clin Invest 2003; 111:1487-96; PMID:12750398; http://dx.doi.org/10.1172/JCI17656
  • Douawho CK, Pride MW, Kripke ML. Persistence of immunogenic pulmonary metastases in the presence of protective anti-melanoma immunity. Cancer Res 2001; 61:215-21; PMID:11196164
  • Lee SY, Sin JI. MC32 tumor cells acquire Ag-specific CTL resistance through the loss of CEA in a colon cancer model. Hum Vaccin Immunother 2015; In press
  • Angell TE, Lechner MG, Jang JK, LoPresti JS, Epstein AL. MHC class I loss is a frequent mechanism of immune escape in papillary thyroid cancer that is reversed by interferon and selumetinib treatment in vitro. Clin Cancer Res 2014; 20(23):6034-44; pii: clincanres.0879.2014; PMID:25294906; http://dx.doi.org/10.1158/1078-0432.CCR-14-0879
  • Jazirehi AR, Baritaki S, Koya RC, Bonavida B, Economou JS. Molecular mechanism of MART-1+/A*0201+ human melanoma resistance to specific CTL-killing despite functional tumor-CTL interaction. Cancer Res 2011; 71:1406-17; PMID:21159666; http://dx.doi.org/10.1158/0008-5472.CAN-10-1296
  • Ruffell B, Affara NI, Coussens LM. Differential macrophage programming in the tumor microenvironment. Trends Immunol 2012; 33:119-26; PMID:22277903; http://dx.doi.org/10.1016/j.it.2011.12.001
  • Ndure J, Flanagan KL. Targeting regulatory T cells to improve vaccine immunogenecity in early life. Front Microbiol 2014; 5:477; PMID:25309517; http://dx.doi.org/10.3389/fmicb.2014.00477
  • Solito S, Bronte V, Mandruzzato S. Antigen specificity of immune suppression by myeloid-derived suppressor cells. J Leukoc Biol 2011; 90:31-6; PMID:21486906; http://dx.doi.org/10.1189/jlb.0111021
  • Bae SH, Park YJ, Park JB, Choi YS, Kim MS, Sin JI. Therapeutic synergy of human papillomavirus E7 subunit vaccines plus cisplatin in an animal tumor model: causal involvement of increased sensitivity of cisplatin-treated tumors to CTL-mediated killing in therapeutic synergy. Clin Cancer Res 2007; 13:341-9; PMID:17200373; http://dx.doi.org/10.1158/1078-0432.CCR-06-1838
  • Ye GW, Park JB, Park YJ, Choi YS, Sin JI. Increased sensitivity of radiated murine cervical cancer tumors to E7 subunit vaccine-driven CTL-mediated killing induces synergistic antitumor activity. Mol Ther 2007; 15:1564-70; PMID:17505485; http://dx.doi.org/10.1038/sj.mt.6300149
  • Sin JI, Kim JM, Bae SH, Lee IH, Park JS, Ryoo HM. Adoptive transfer of human papillomavirus E7-specific CTL enhances tumor chemoresponse through the perforin/granzyme-mediated pathway. Mol Ther 2009; 17:906-13; PMID:19277009; http://dx.doi.org/10.1038/mt.2009.32
  • Norell H, Poschke I, Charo J, Wei WZ, Erskine C, Piechocki MP, Knutson KL, Bergh J, Lidbrink E, Kiessling R. Vaccination with a plasmid DNA encoding HER-2/neu together with low doses of GM-CSF and IL-2 in patients with metastatic breast carcinoma: a pilot clinical trial. J Transl Med 2010; 8:53; PMID:20529245; http://dx.doi.org/10.1186/1479-5876-8-53

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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